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:

Replication fork uncoupling causes nascent strand degradation and fork reversal

Abstract

Genotoxins cause nascent strand degradation (NSD) and fork reversal during DNA replication. NSD and fork reversal are crucial for genome stability and are exploited by chemotherapeutic approaches. However, it is unclear how NSD and fork reversal are triggered. Additionally, the fate of the replicative helicase during these processes is unknown. We developed a biochemical approach to study synchronous, localized NSD and fork reversal using Xenopus egg extracts and validated this approach with experiments in human cells. We show that replication fork uncoupling stimulates NSD of both nascent strands and progressive conversion of uncoupled forks to reversed forks. Notably, the replicative helicase remains bound during NSD and fork reversal. Unexpectedly, NSD occurs before and after fork reversal, indicating that multiple degradation steps take place. Overall, our data show that uncoupling causes NSD and fork reversal and elucidate key events that precede fork reversal.

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: Induction of synchronous and localized NSD in vitro.
Fig. 2: NSD involves DNA2 but not MRE11.
Fig. 3: NSD involves fork reversal.
Fig. 4: Uncoupling promotes NSD and fork reversal.
Fig. 5: The replisome remains associated with DNA during NSD.
Fig. 6: DNA2 degrades replication forks and reversed forks.
Fig. 7: Model for NSD in Xenopus egg extracts.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are available within the article and its Supplementary Information files. Source data are provided with this paper.

References

  1. Berti, M., Cortez, D. & Lopes, M. The plasticity of DNA replication forks in response to clinically relevant genotoxic stress. Nat. Rev. Mol. Cell Biol. 21, 633–651 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Quinet, A., Lemaçon, D. & Vindigni, A. Replication Fork Reversal: Players and Guardians. Mol. Cell 68, 830–833 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Rickman, K. & Smogorzewska, A. Advances in understanding DNA processing and protection at stalled replication forks. J. Cell Biol. 218, 1096–1107 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ait Saada, A., Lambert, S. A. E. & Carr, A. M. Preserving replication fork integrity and competence via the homologous recombination pathway. DNA Repair (Amst.) 71, 135–147 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Mutreja, K. et al. ATR-Mediated Global Fork Slowing and Reversal Assist Fork Traverse and Prevent Chromosomal Breakage at DNA Interstrand Cross-Links. Cell Rep. 24, 2629–2642.e2625 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Manosas, M., Perumal, S. K., Croquette, V. & Benkovic, S. J. Direct observation of stalled fork restart via fork regression in the T4 replication system. Science 338, 1217–1220 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Thangavel, S. et al. DNA2 drives processing and restart of reversed replication forks in human cells. J. Cell Biol. 208, 545–562 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Schlacher, K. et al. Double-strand break repair-independent role for BRCA2 in blocking stalled replication fork degradation by MRE11. Cell 145, 529–542 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hashimoto, Y., Ray Chaudhuri, A., Lopes, M. & Costanzo, V. Rad51 protects nascent DNA from Mre11-dependent degradation and promotes continuous DNA synthesis. Nat. Struct. Mol. Biol. 17, 1305–1311 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dias, M. P., Moser, S. C., Ganesan, S. & Jonkers, J. Understanding and overcoming resistance to PARP inhibitors in cancer therapy. Nat Rev Clin Oncol, https://doi.org/10.1038/s41571-021-00532-x (2021).

  11. Taglialatela, A. et al. Restoration of Replication Fork Stability in BRCA1- and BRCA2-Deficient Cells by Inactivation of SNF2-Family Fork Remodelers. Mol. Cell 68, 414–430.e418 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ray Chaudhuri, A. et al. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature 535, 382–387 (2016).

    Article  PubMed  Google Scholar 

  13. Mijic, S. et al. Replication fork reversal triggers fork degradation in BRCA2-defective cells. Nat. Commun. 8, 859 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Zellweger, R. et al. Rad51-mediated replication fork reversal is a global response to genotoxic treatments in human cells. J. Cell Biol. 208, 563–579 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fugger, K. et al. FBH1 Catalyzes Regression of Stalled Replication Forks. Cell Rep. 10, 1749–1757 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Kolinjivadi, A. M. et al. Smarcal1-Mediated Fork Reversal Triggers Mre11-Dependent Degradation of Nascent DNA in the Absence of Brca2 and Stable Rad51 Nucleofilaments. Mol. Cell 67, 867–881.e867 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Vujanovic, M. et al. Replication Fork Slowing and Reversal upon DNA Damage Require PCNA Polyubiquitination and ZRANB3 DNA Translocase Activity. Mol. Cell 67, 882–890.e885 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lemaçon, D. et al. MRE11 and EXO1 nucleases degrade reversed forks and elicit MUS81-dependent fork rescue in BRCA2-deficient cells. Nat. Commun. 8, 860 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Hu, J. et al. The intra-S phase checkpoint targets Dna2 to prevent stalled replication forks from reversing. Cell 149, 1221–1232 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Mason, J. M., Chan, Y. L., Weichselbaum, R. W. & Bishop, D. K. Non-enzymatic roles of human RAD51 at stalled replication forks. Nat. Commun. 10, 4410 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Higgs, M. R. et al. BOD1L Is Required to Suppress Deleterious Resection of Stressed Replication Forks. Mol. Cell 59, 462–477 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Ray Chaudhuri, A. et al. Topoisomerase I poisoning results in PARP-mediated replication fork reversal. Nat. Struct. Mol. Biol. 19, 417–423 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Berti, M. et al. Human RECQ1 promotes restart of replication forks reversed by DNA topoisomerase I inhibition. Nat. Struct. Mol. Biol. 20, 347–354 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Amunugama, R. et al. Replication Fork Reversal during DNA Interstrand Crosslink Repair Requires CMG Unloading. Cell Rep. 23, 3419–3428 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Iannascoli, C., Palermo, V., Murfuni, I., Franchitto, A. & Pichierri, P. The WRN exonuclease domain protects nascent strands from pathological MRE11/EXO1-dependent degradation. Nucleic Acids Res. 43, 9788–9803 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Vallerga, M. B., Mansilla, S. F., Federico, M. B., Bertolin, A. P. & Gottifredi, V. Rad51 recombinase prevents Mre11 nuclease-dependent degradation and excessive PrimPol-mediated elongation of nascent DNA after UV irradiation. Proc. Natl Acad. Sci. USA 112, E6624–E6633 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Timson, J. Hydroxyurea. Mutat. Res 32, 115–132 (1975).

    Article  CAS  PubMed  Google Scholar 

  28. Couch, F. B. et al. ATR phosphorylates SMARCAL1 to prevent replication fork collapse. Genes Dev. 27, 1610–1623 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Toledo, L. I. et al. ATR prohibits replication catastrophe by preventing global exhaustion of RPA. Cell 155, 1088–1103 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Sparks, J. L. et al. The CMG Helicase Bypasses DNA-Protein Cross-Links to Facilitate Their Repair. Cell 176, 167–181.e121 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Graham, J. E., Marians, K. J. & Kowalczykowski, S. C. Independent and Stochastic Action of DNA Polymerases in the Replisome. Cell 169, 1201–1213.e1217 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rickman, K. A. et al. Distinct roles of BRCA2 in replication fork protection in response to hydroxyurea and DNA interstrand cross-links. Genes Dev. 34, 832–846 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Somyajit, K. et al. Homology-directed repair protects the replicating genome from metabolic assaults. Dev. Cell 56, 461–477.e467 (2021).

    Article  CAS  PubMed  Google Scholar 

  34. Wasserman, M. R., Schauer, G. D., O’Donnell, M. E. & Liu, S. Replication Fork Activation Is Enabled by a Single-Stranded DNA Gate in CMG Helicase. Cell 178, 600–611.e616 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Vrtis, K. B. et al. Single-strand DNA breaks cause replisome disassembly. Mol. Cell 81, 1309–1318.e1306 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Dungrawala, H. et al. RADX Promotes Genome Stability and Modulates Chemosensitivity by Regulating RAD51 at Replication Forks. Mol. Cell 67, 374–386.e375 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, W., Krishnamoorthy, A., Zhao, R. & Cortez, D. Two replication fork remodeling pathways generate nuclease substrates for distinct fork protection factors. Sci Adv 6, https://doi.org/10.1126/sciadv.abc3598 (2020).

  38. Byun, T. S., Pacek, M., Yee, M. C., Walter, J. C. & Cimprich, K. A. Functional uncoupling of MCM helicase and DNA polymerase activities activates the ATR-dependent checkpoint. Genes Dev. 19, 1040–1052 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Walter, J., Sun, L. & Newport, J. Regulated chromosomal DNA replication in the absence of a nucleus. Mol. Cell 1, 519–529 (1998).

    Article  CAS  PubMed  Google Scholar 

  40. Hanada, K. et al. The structure-specific endonuclease Mus81 contributes to replication restart by generating double-strand DNA breaks. Nat. Struct. Mol. Biol. 14, 1096–1104 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dewar, J. M., Budzowska, M. & Walter, J. C. The mechanism of DNA replication termination in vertebrates. Nature 525, 345–350 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, W. et al. A Selective Small Molecule DNA2 Inhibitor for Sensitization of Human Cancer Cells to Chemotherapy. EBioMedicine 6, 73–86 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Dupré, A. et al. A forward chemical genetic screen reveals an inhibitor of the Mre11-Rad50-Nbs1 complex. Nat. Chem. Biol. 4, 119–125 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Zhu, Z., Chung, W. H., Shim, E. Y., Lee, S. E. & Ira, G. Sgs1 helicase and two nucleases Dna2 and Exo1 resect DNA double-strand break ends. Cell 134, 981–994 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chaudhury, I., Stroik, D. R. & Sobeck, A. FANCD2-controlled chromatin access of the Fanconi-associated nuclease FAN1 is crucial for the recovery of stalled replication forks. Mol. Cell. Biol. 34, 3939–3954 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Coquel, F. et al. SAMHD1 acts at stalled replication forks to prevent interferon induction. Nature 557, 57–61 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Long, D. T. & Kreuzer, K. N. Regression supports two mechanisms of fork processing in phage T4. Proc. Natl Acad. Sci. USA 105, 6852–6857 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hunter, N. & Kleckner, N. The single-end invasion: an asymmetric intermediate at the double-strand break to double-holliday junction transition of meiotic recombination. Cell 106, 59–70 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Tian, T. et al. The ZATT-TOP2A-PICH Axis Drives Extensive Replication Fork Reversal to Promote Genome Stability. Mol. Cell 81, 198–211.e196 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Walter, J. & Newport, J. Initiation of eukaryotic DNA replication: origin unwinding and sequential chromatin association of Cdc45, RPA, and DNA polymerase alpha. Mol. Cell 5, 617–627 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Deng, L. et al. Mitotic CDK Promotes Replisome Disassembly, Fork Breakage, and Complex DNA Rearrangements. Mol. Cell 73, 915–929.e916 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dewar, J. M., Low, E., Mann, M., Räschle, M. & Walter, J. C. CRL2(Lrr1) promotes unloading of the vertebrate replisome from chromatin during replication termination. Genes Dev. 31, 275–290 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wong, R. P., García-Rodríguez, N., Zilio, N., Hanulová, M. & Ulrich, H. D. Processing of DNA Polymerase-Blocking Lesions during Genome Replication Is Spatially and Temporally Segregated from Replication Forks. Mol. Cell 77, 3–16.e14 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Quinet, A. et al. PRIMPOL-Mediated Adaptive Response Suppresses Replication Fork Reversal in BRCA-Deficient Cells. Mol. Cell 77, 461–474.e469 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Cong, K. et al. Replication gaps are a key determinant of PARP inhibitor synthetic lethality with BRCA deficiency. Mol. Cell 81, 3128–3144.e3127 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Tirman, S. et al. Temporally distinct post-replicative repair mechanisms fill PRIMPOL-dependent ssDNA gaps in human cells. Mol. Cell 81, 4026–4040.e4028 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Low, E., Chistol, G., Zaher, M. S., Kochenova, O. V. & Walter, J. C. The DNA replication fork suppresses CMG unloading from chromatin before termination. Genes Dev. 34, 1534–1545 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Deegan, T. D., Mukherjee, P. P., Fujisawa, R., Polo Rivera, C. & Labib, K. CMG helicase disassembly is controlled by replication fork DNA, replisome components and a ubiquitin threshold. Elife 9, https://doi.org/10.7554/eLife.60371 (2020).

  60. Kose, H. B., Xie, S., Cameron, G., Strycharska, M. S. & Yardimci, H. Duplex DNA engagement and RPA oppositely regulate the DNA-unwinding rate of CMG helicase. Nat. Commun. 11, 3713 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Masuda-Ozawa, T., Hoang, T., Seo, Y. S., Chen, L. F. & Spies, M. Single-molecule sorting reveals how ubiquitylation affects substrate recognition and activities of FBH1 helicase. Nucleic Acids Res. 41, 3576–3587 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bétous, R. et al. SMARCAL1 catalyzes fork regression and Holliday junction migration to maintain genome stability during DNA replication. Genes Dev. 26, 151–162 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Biebricher, A. et al. PICH: a DNA translocase specially adapted for processing anaphase bridge DNA. Mol. Cell 51, 691–701 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Paudyal, S. C., Li, S., Yan, H., Hunter, T. & You, Z. Dna2 initiates resection at clean DNA double-strand breaks. Nucleic Acids Res. 45, 11766–11781 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Nieminuszczy, J. et al. EXD2 Protects Stressed Replication Forks and Is Required for Cell Viability in the Absence of BRCA1/2. Mol. Cell 75, 605–619.e606 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhang, J. et al. DNA interstrand cross-link repair requires replication-fork convergence. Nat. Struct. Mol. Biol. 22, 242–247 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Huang, J. et al. The DNA translocase FANCM/MHF promotes replication traverse of DNA interstrand crosslinks. Mol. Cell 52, 434–446 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Bai, G. et al. HLTF Promotes Fork Reversal, Limiting Replication Stress Resistance and Preventing Multiple Mechanisms of Unrestrained DNA Synthesis. Mol. Cell 78, 1237–1251.e1237 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lebofsky, R., Takahashi, T. & Walter, J. C. DNA replication in nucleus-free Xenopus egg extracts. Methods Mol. Biol. 521, 229–252 (2009).

    Article  CAS  PubMed  Google Scholar 

  70. Klein Douwel, D. et al. XPF-ERCC1 acts in Unhooking DNA interstrand crosslinks in cooperation with FANCD2 and FANCP/SLX4. Mol. Cell 54, 460–471 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Heintzman, D. R., Campos, L. V., Byl, J. A. W., Osheroff, N. & Dewar, J. M. Topoisomerase II Is Crucial for Fork Convergence during Vertebrate Replication Termination. Cell Rep. 29, 422–436.e425 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grant R35GM128696 to J.M.D., ACS grant IRG-15-169-56 to J.M.D., funding provided by the Breast Cancer Research Foundation to D.C. and Vanderbilt-Ingram Cancer Center Support Grant P30CA068485. We thank V. Costanzo (IFOM, FIRC Institute for Molecular Oncology, Italy and University of Milan, Italy) for antibodies to Xenopus SAMHD1 and SMARCAL1 as well as for helpful discussion about the work. We thank J. Walter (Harvard Medical School, USA and Howard Hughes Medical Institute, USA) for antibody to Xenopus FAN1.

Author information

Authors and Affiliations

Authors

Contributions

W.L. performed the experiments in Figs. 2d and 6h and Extended Data Figs. 1b, 3j,k, 4f and 8f. T.M.M. performed the experiments in Fig. 3h and Extended Data Fig. 1c. T.K. performed all other experiments. T.K. and J.M.D. designed the experiments, analyzed the data and wrote the paper with input from D.C., T.M.M. and W.L.

Corresponding author

Correspondence to James M. Dewar.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Stephan Hamperl and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Beth Moorefield, in collaboration with the Nature Structural & Molecular Biology team.

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 Nascent strand degradation is an initial response to fork stalling.

(A) A current model for fork reversal and nascent strand degradation. Note that fork reversal enzymes are thought to directly convert reversed forks back to replication forks1,2,3. (B) U2OS cells were pulse-labeled with CldU then IdU followed by treatment with the indicated concentrations of aphidicolin so that loss of the IdU labeled tracks could be monitored as a measurement of NSD. DNA fiber analysis was performed to determine the lengths of CldU and IdU labeled DNA tracks. n indicates the number of DNA fibers measured for each condition. P-values from a one-way ANOVA performed using Dunnett’s multiple comparisons test are reported for each condition. Non-significant comparisons are indicated as ‘ns’. Although lower concentrations of aphidicolin (0.2–1 µM) did not impact the IdU/CldU ratio higher concentrations (4–10 µM) significantly reduced the IdU/CldU ratio indicating that NSD took place. Thus, we observed a dose-dependent induction of NSD by aphidicolin. (C) U2OS cells were pulse labeled with CldU and IdU, then treated with the indicated concentrations of aphidicolin and hydroxyurea (HU). DNA fiber analysis was performed to determine the lengths of CldU and IdU labeled DNA tracks. n indicates the number of DNA fibers measured for each condition. P-values from a one-way ANOVA performed using Dunnett’s multiple comparisons test are reported for each condition. Non-significant comparisons are indicated as ‘ns’. Co-addition of the RAD51 inhibitor B02 (RAD51-i)4 was also performed as a positive control for degradation in the presence of HU. RAD51-i addition inactivated fork protection and led to extensive degradation in the presence of HU, as previously described5. In contrast, 1, 4, and 8 mM HU did not result in detectable NSD, consistent with limited degradation in the presence of HU alone6. Notably, the extent of NSD following 10 µM aphidicolin treatment was comparable to the level observed following HU treatment and RAD51-i addition. (D) To stall replication forks in vitro, plasmid DNA was replicated using Xenopus egg extracts and newly-synthesized nascent strands were radiolabeled by inclusion of [α-32P]dATP. In this system, plasmid templates replicate semi-synchronously from a single origin per plasmid7. 6 minutes after initiation, reactions were treated with aphidicolin, to stall DNA synthesis, or vehicle control. As a loading control (Ctrl) the reactions include a smaller plasmid that was radiolabeled prior to the experiment and did not undergo replication. (E) DNA structures from (d) were purified and digested with XmnI, which cuts the plasmid once. (F) DNA taken from (e) at the moment of aphidicolin addition (6 minutes after initiation) was separated by 2-D gel electrophoresis and visualized by autoradiography. The entire Double-Y and Bubble arcs (i) were visible (ii), indicating that DNA molecules of all sizes, and thus at all stages of replication, were present. Note that origin firing generates bubble structures, which are converted to double-Ys once one of the two forks moves beyond the restriction site (iii). (G) Samples from (e) were separated on an agarose gel and visualized by autoradiography. Time after aphidicolin addition is indicated. (H) Quantification of DNA synthesis from (g) normalized to the maximum signal across all time points and conditions. Mean ± S.D., n = 3 independent experiments. (I) Quantification of DNA synthesis in (g) normalized to the maximum signal for each condition across all time points. Mean ± S.D., n = 3 independent experiments. Following vehicle treatment in (h)-(i), radioactive signal increased and then plateaued, indicating that a single round of replication was completed. In contrast, following aphidicolin treatment in (h)-(i) signal declined, indicating that degradation took place. Importantly, signal was appreciably reduced by 60 minutes, indicating that degradation was an initial response and did not require prolonged fork stalling. (J) To test whether degradation in (h)-(i) corresponded to both strands or nascent strands only, pre-radiolabeled plasmid DNA was replicated using Xenopus egg extracts. 6 minutes after initiation, DNA synthesis was inhibited by the addition of Aphidicolin, as in (e). (K) XmnI digested molecules were separated on an agarose gel and visualized by autoradiography. As a loading control (Ctrl) the reactions include a smaller radiolabeled plasmid that did not undergo replication. (L) Quantification of (k) as in (h). Mean ± S.D., n = 3 independent experiments. (M) Quantification of (k) as in (i). Mean ± S.D., n = 3 independent experiments. In (l)-(m) parental strand signal was relatively stable and there was little difference between aphidicolin and vehicle treatment. Moreover, there was no appreciable loss of parental strand signal for the first 120 minutes, during which time most of the nascent strands were degraded in (h)-(i). Thus, most signal loss was due to degradation of nascent DNA strands. Overall, (d)-(l) show that degradation of nascent strands is an initial response to fork stalling.

Source data

Extended Data Fig. 2 Characterization of localized, synchronous nascent strand degradation.

(A) Cartoon depicting the source of θ* structures in Fig. 1B. The XmnI site is located 1013 base pairs away from the closest edge of the lacO array. (B) DNA structures from Fig. 1A were treated with human topoisomerase II (hTop2) or buffer control, then separated on an agarose gel and visualized by autoradiography. hTop2 treatment converted θ* signal back to θs, demonstrating that θ* structures are topoisomers of θs. Note that in lane 4 an additional band appears below the θ, suggesting that replication forks undergo remodeling (see Fig. 3 and related discussion in the main text). (C) Quantification of Replication Intermediates (RIs) as a % of total lane signal from Fig. 1E. Mean ± S.D., n = 5 independent experiments. (D) Quantification of total lane signal from Fig. 1E. Mean ± S.D., n = 5 independent experiments. (E) Plasmid DNA harboring a 32xlacO array (p[lacO]) or a 50xlacO array (p[lacOx50]) was incubated with LacR then replicated in Xenopus egg extracts. Once forks were localized to the LacR barrier, NSD was induced by addition of IPTG and aphidicolin. Purified DNA was subjected to restriction digest so that replication fork structures (RIs) could be visualized. (F) Samples from (e) were separated on an agarose gel and visualized by autoradiography. Note that a mobility shift can readily be detected for p[lacOx50], suggesting that fork remodeling occurs (see Fig. 4 and related main text). (G) Quantification of the amount of DNA degraded in (f). Normalized degradation accounts for the different backbone sizes of the 50x and 32x lacO plasmids. Mean ± S.D., n = 3 independent experiments. The 50xlacO array is ~60% larger than the 32xlacO array so if NSD was influenced by the proximity of the replication forks on either side of the lacO array there would be a substantial difference in NSD between p[lacO] and p[lacOx50]. However, the difference in normalized degradation between the two plasmids is negligible. Thus, NSD does not appear to be influenced by the proximity of the two converging forks.

Source data

Extended Data Fig. 3 Characterization of MRE11 and DNA2 activity during nascent strand degradation.

(A) Cartoon depicting the roles of MRE11 and DNA2 in resection at double-strand breaks. (B) Cartoon depicting the roles of MRE11 and DNA2 in resection at reversed forks. (C) Linear radiolabeled DNA was incubated in Xenopus egg extracts in the presence of MRE11 inhibitor (MRE11-i) or DNA2 inhibitor (DNA2-i) or both. Double-Strand Break (DSB) resection involves both enzymes in Xenopus egg extracts (b) and is blocked by inactivation of both DNA2 and MRE118. Thus, if both MRE11-i and DNA2-i effectively inhibit their targets then combination of both drugs should block resection. (D) Samples from (c) were separated on an agarose gel and visualized by autoradiography. In the vehicle control the linear substrate was rapidly degraded (lanes 1-4). We also observed a small number of additional products that arose from end-joining (EJ(L) and EJ(C)). As expected9, DNA2-i and MRE11-i each inhibited resection (lanes 5-8, 9-12), while a combination of both inhibitors almost completely blocked resection (lanes 13-16). (E) Quantification of total signal from (d). Mean ± S.D., n = 3 independent experiments. (F) Depiction of the DNA structures generated by Fig. 2A, prior to restriction digest. (G) DNA intermediates from (f) were separated on an agarose gel and visualized by autoradiography. Uncoupled species (θ*) are indicated. (H) Quantification of total lane signal from Fig. 2B. Mean ± S.D., n = 3 independent experiments. (I) Quantification of RI signal in Fig. 2B as a percentage of total signal for each lane. Mean ± S.D., n = 3 independent experiments. (J) U2OS cells were pulse labeled with CldU and IdU, then treated with either aphidicolin, or hydroxyurea combined with RAD51-i. MRE11-i, DNA2-i, or vehicle control were also added, as indicated. DNA fiber analysis was performed to determine the lengths of IdU and CldU labeled DNA tracks. n indicates the number of DNA fibers measured for each condition. P-values from a one-way ANOVA performed using Dunnett’s multiple comparisons test are reported for each condition. Non-significant comparisons are indicated as ‘ns’. Treatment with Rad51-i compromised fork protection and led to NSD in the presence of HU, as previously reported5. Additionally, NSD was rescued by either DNA2-i or MRE11-i, as expected10, confirming that both drugs were able to effectively inhibit their targets. Aphidicolin treatment resulted in NSD (as in Extended Data Fig. 1B) and this was rescued by DNA2-i but not MRE11-i. Thus, NSD caused by aphidicolin treatment results in DNA2-dependent but MRE11-independent degradation. (K) HCT116 cells were transfected with the indicated siRNA. 72 hours later cells were pulse labeled with CldU and IdU, then treated with aphidicolin. DNA fiber analysis was performed to determine the lengths of IdU and CldU labeled DNA tracks. n indicates the number of DNA fibers measured for each condition. P-values from a one-way ANOVA performed using Dunnett’s multiple comparisons test are reported for each condition. Non-significant comparisons are indicated as ‘ns’.

Source data

Extended Data Fig. 4 SAMHD1 and FAN1 are not involved in nascent strand degradation.

(A) Forks were localized to a LacR barrier and NSD was induced by addition of IPTG and aphidicolin in SAMHD1- and FAN1-immunodepleted Xenopus egg extracts. Purified DNA was subjected to restriction digest so that replication fork structures could be visualized (RIs). (B) Immunodepleted extracts and the corresponding immunoprecipitates (beads) from (a) were analyzed by Western blotting to determine the extent of SAMHD1 and FAN1 immunodepletion. In both cases the immunodepletion went to completion because no detectable protein was recovered by round 3 (compare lanes 7-8 to lane 9). Additionally, at least 95% of protein was removed in each case (compare lanes 1 and 5). (C) Samples from (a) were separated on an agarose gel and visualized by autoradiography. (D) Quantification of RI signal from (c). (E) Quantification of RI signal from an experimental replicate of (c). In both (d) and (e) neither SAMHD1 nor FAN1 depletion altered the rate of degradation, indicating that they do not participate in NSD. (F) U2OS cells were transfected with the indicated siRNA, pulse labeled with CldU and IdU, then treated with aphidicolin. DNA fiber analysis was performed to determine the lengths of CldU and IdU labeled DNA tracks. n indicates the number of DNA fibers measured for each condition. P-values from a one-way ANOVA performed using Dunnett’s multiple comparisons test are reported for each condition. Non-significant comparisons are indicated as ‘ns’. Neither SAMHD1 nor FAN1 affected degradation, consistent with (d)-(e).

Source data

Extended Data Fig. 5 Characterization of DNA structures detected by 2-D gel during nascent strand degradation.

(A) Quantification of Double-Ys from Fig. 3C expressed as a percentage of total signal at T = 0. Mean ± S.D., n = 3 independent experiments. (B) Quantification of remodeled forks from Fig. 3C expressed as a percentage of total signal at T = 0. Mean ± S.D., n = 3 independent experiments. (C) Cartoon indicating the different DNA structures that the remodeled forks in Fig. 3C could correspond to. (D) Expected 2-D gel migration pattern of the structures depicted in (c). The region highlighted in red indicates where each structure is expected to migrate. ‘?’ indicates structures whose expected migration on a 2-D gel is unclear. The remodeled forks (Fig. 3C) did not correspond to canonical Holliday junctions (i) or hemicatenanes (ii). However, these species could have reflected reversed forks (iii), hemicatenanes (iv), or Holliday junctions (v) in the wake of one or both forks. (E) Samples from Fig. 3C,iii were treated with buffer or RuvC, which cleaves reversed forks and Holliday junctions, but not hemicatenanes or other replication fork structures11. RuvC-treated samples were then separated by 2-D gel electrophoresis and visualized by autoradiography. (F) Quantification of DYs and RFs following RuvC treatment in (e) expressed relative to the abundance of each structure in the buffer treated condition. RuvC treatment had essentially no effect on Double-Ys, as expected, but reduced remodeled forks by ~5-fold. Thus, the remodeled forks contain four-way junctions that arise from reversed forks or Holliday junctions that is D-loops. (G) Quantification of RFs and DYs from (c) expressed relative to total signal on each gel. (H) Cartoon indicating the structure of a D-loop and a reversed fork. (I) Expected 2-D gel migration pattern of the structures depicted in (h). The region highlighted in red indicates where each structure is expected to migrate. (J) Cartoon indicating the restriction digest strategy used to excise individual replication forks for 2-D gel analysis. The DraIII site is 450 bp away from the nearest edge of the lacO array and 1250 bp away from the XhoI site.(K) Forks were localized to a LacR barrier and NSD was induced by addition of IPTG and aphidicolin in SMARCAL1-immunodepleted Xenopus egg extracts. Purified DNA was subjected to restriction digest so that replication fork structures could be visualized (RIs). (L) Immunodepleted extracts and the corresponding immunoprecipitates (beads) from (k) were analyzed by Western blotting to determine the extent of SMARCAL1 immunodepletion. Immunoblotting was performed using an antibody generated by this study (i) and the previously published antibody (ii)12. No detectable protein was recovered by the final round of depletion (compare lanes 7-8 to lane 9), indicating that the depletion was successful. Additionally, at least 99% of protein was removed (compare lanes 1 and 6 for (ii).(M) Samples from (k) were separated on an agarose gel and visualized by autoradiography. (N) Quantification of RI signal from (m). (O) Quantification of RI signal from an experimental replicate of (m). In both (n) and (o) SMARCAL1 immunodepletion had no impact on degradation.

Source data

Extended Data Fig. 6 Characterization of stalled and uncoupled replication forks.

(A) Cartoon depicting the strategy to test the role of helicase stalling during NSD. If helicase stalling triggers NSD then NSD should be increased in the absence of IPTG. If uncoupling triggers NSD then addition of IPTG should increase NSD. (B) Depiction of the DNA structures generated by Fig. 4A, prior to restriction digest. (C) Samples from (e) were separated on an agarose gel and visualized by autoradiography. θ* species arise from uncoupling. θ- species arise from the low level of uncoupling that occurs even in the presence of LacR, which is not a complete block to helicase progression13. (D) Depiction of different models for degradation of leading and lagging strands during NSD observed in Fig. 4 and the expected effect on degradation of LWS and RWS depicted in Fig. 4A. In (i) both leading and lagging strand are degraded simultaneously and synchronously, which should result in disappearance of RWS before LWS. In (ii) lagging strands only are degraded synchronously, which should result in persistence of ~50% of RWS and LWS until the leading strand are degraded by nuclease activity that initiated at lagging strands of the opposite fork. This would result in biphasic kinetics of degradation. In (iii) lagging strands only are degraded asynchronously, which should result in loss of RWS and LWS at the same rate. (E) Uncropped gel of Fig. 4B lanes 1-4 shown after a longer exposure so that the weaker rightward strands (RWS) could be visualized. (F) Quantification of LWS and RWS from (b). RWS are degraded before LWS, with no evidence of biphasic kinetics, indicating that both strands are degraded simultaneously, as in (a,i). Mean ± S.D., n = 3 independent experiments. (G) NSD was induced as in Fig. 4A in the presence or absence of DNA2-i. (H) Samples from (g) were separated on a native agarose gel and visualized by autoradiography. (I) Quantification of RI signal from (h). Mean ± S.D., n = 3 independent experiments. (J) Quantification of reversed forks from Fig. 4E expressed as a percentage of total signal at T = 0. Mean ± S.D., n = 3 independent experiments.

Source data

Extended Data Fig. 7 Binding of replication fork proteins during NSD.

(A) Cartoon depicting two different models of CMG helicase behavior during fork reversal and NSD. In (i) the CMG helicase translocates onto double-stranded DNA, as suggested1,14, which would result in its removal from DNA15,16,17,18. In (ii) the replisome remains on DNA, suggesting that it resides in a ssDNA bubble ahead of the reversed fork. (B) In parallel to Fig. 5A chromatin bound proteins were analyzed in the presence of DNA2-i. RPA was detected by Western Blotting. (C) Quantification of RPA signal from Fig. 5B. and (b). Mean ± S.D., n = 3 independent experiments. Between 0 and 60 minutes, when most fork reversal took place (Fig. 3D), most RPA signal was due to DNA2 activity. Thus, most RPA signal during this time was due to DNA2-dependent NSD, which may mask any dissociation of RPA that occurred due to reannealing of parental DNA strands.

Source data

Extended Data Fig. 8 Additional degradation steps during NSD.

(A) Cartoon of different models to explain a role for DNA2 in degrading Y-shaped forks during NSD and the expected impact of loss of DNA2 activity on Y-shaped forks in each case. (i) depicts a model where DNA2 degrades reversed forks to generate Y-shaped forks. In this model the Y-shaped forks are degraded as consequence of prior DNA2 activity at reversed forks. (ii) depicts the impact of impaired DNA2 activity on (i). Y-shaped forks should decrease in abundance as less degradation of reversed forks should reduce the formation of Y-shaped forks. (iii) depicts a model where DNA2 degrades forks prior to fork reversal. (iv) depicts the impact of impaired DNA2 activity on (iii). Y-shaped forks should increase in abundance due to less degradation of nascent strands, which is the source of the radioactive signal that is measured. (B) Quantification of Double-Ys from Fig. 6B expressed as a percentage of total replication intermediates (RIs). Mean ± S.D., n = 3 independent experiments. (C) Quantification of reversed forks from Fig. 6B expressed as a percentage of total replication intermediates (RIs). Mean ± S.D., n = 3 independent experiments. (D) U2OS cells treated with control or RAD51 siRNA were lysed and analyzed by western blotting for RAD51. Total protein levels were also determined by stain-free imaging (Bio-Rad). (E) HCT116 cells were transfected with RAD51 siRNA, pulse labeled with CldU and IdU, and then treated with aphidicolin. DNA fiber analysis was performed to determine the lengths of CldU and IdU labeled DNA tracks in each condition. (F) HCT116 cells were transfected with control or RAD51 siRNA, pulse labeled with CldU and IdU, then treated with aphidicolin. DNA fiber analysis was performed to determine the lengths of the CldU and IdU labeled DNA tracks. n indicates the number of DNA fibers measured for each condition. P-values from a one-way ANOVA performed using Dunnett’s multiple comparisons test are reported for each condition. Non-significant comparisons are indicated as ‘ns’. Degradation still occurred even following knockdown of RAD51, consistent with results from U2OS cells in Fig. 6H. (G) Potential models for template switching, based on our new model for NSD (Fig. 7). (i) After encountering a polymerase-blocking lesion (yellow) replication forks uncouple, resulting in a stalled replisome (red). (ii) The replisome is retained on DNA and parental DNA strands anneal to create a second replication fork that is the substrate for fork reversal. This places the replisome within a single-stranded DNA bubble. (iii) The nascent lagging strands serve as template for leading strand synthesis (green). (iv) Exonuclease activity at the extruded DNA end generates a 3′ overhang. (v) restoration of the reversed fork results in a lagging strand gap that allows loading of a 5′-3′ single-stranded DNA helicase that can unwind the re-annealed parental strands. (vi) nascent strands are extended to the replisome, which resumes unwinding (green). (vii) Without exonuclease activity at the extruded DNA end (in (iv)) restoration of the reversed fork would result in a 3-way fully duplex fork structure that could not be readily unwound. (viii) An alternative model of the response replication fork uncoupling by a polymerase-blocking lesion. (ix) Nascent DNA strands are unwound by single-stranded DNA helicases, allowing them to anneal together. (x) The annealed duplex generated in (ix) is the substrate for fork reversal enzymes, which reanneal parental duplex as the nascent DNA strands are extruded. (xi) Exonuclease activity at the extruded end generates a 3′ overhang, as in (iv), but the bubble structure is more extensive and the parental duplex is more limited. (xii) Restoration of the reversed fork removes all parental duplex, provided that only one of the two strands remains intact. (xiii) nascent strands are extended to the replisome, which resumes unwinding.

Source data

Supplementary information

Reporting Summary

Supplementary Note

References for Extended Data figure legends.

Source data

Source Data Fig. 1

Unprocessed gels.

Source Data Fig. 1

Numerical data.

Source Data Fig. 2

Unprocessed gels.

Source Data Fig. 2

Numerical data.

Source Data Fig. 3

Unprocessed gels.

Source Data Fig. 3

Numerical data.

Source Data Fig. 4

Unprocessed gels.

Source Data Fig. 4

Numerical data.

Source Data Fig. 5

Unprocessed gels/western blots.

Source Data Fig. 5

Numerical data.

Source Data Fig. 6

Unprocessed gels.

Source Data Fig. 6

Numerical data.

Source Data Extended Data Fig. 1

Unprocessed gels.

Source Data Extended Data Fig. 1

Numerical data.

Source Data Extended Data Fig. 2

Unprocessed gels.

Source Data Extended Data Fig. 2

Numerical data.

Source Data Extended Data Fig. 3

Unprocessed gels.

Source Data Extended Data Fig. 3

Numerical data.

Source Data Extended Data Fig. 4

Unprocessed gels/western blots.

Source Data Extended Data Fig. 4

Numerical data.

Source Data Extended Data Fig. 5

Unprocessed gels/western blots.

Source Data Extended Data Fig. 5

Numerical data.

Source Data Extended Data Fig. 6

Unprocessed gels.

Source Data Extended Data Fig. 6

Numerical data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 7

Numerical data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 8

Numerical 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

Kavlashvili, T., Liu, W., Mohamed, T.M. et al. Replication fork uncoupling causes nascent strand degradation and fork reversal. Nat Struct Mol Biol 30, 115–124 (2023). https://doi.org/10.1038/s41594-022-00871-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00871-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