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.

Regulation of DNA repair pathway choice in S and G2 phases by the NHEJ inhibitor CYREN

Abstract

Classical non-homologous end joining1 (cNHEJ) and homologous recombination2 compete for the repair of double-stranded DNA breaks during the cell cycle. Homologous recombination is inhibited during the G1 phase of the cell cycle, but both pathways are active in the S and G2 phases. However, it is unclear why cNHEJ does not always outcompete homologous recombination during the S and G2 phases. Here we show that CYREN (cell cycle regulator of NHEJ) is a cell-cycle-specific inhibitor of cNHEJ. Suppression of CYREN allows cNHEJ to occur at telomeres and intrachromosomal breaks during the S and G2 phases, and cells lacking CYREN accumulate chromosomal aberrations upon damage induction, specifically outside the G1 phase. CYREN acts by binding to the Ku70/80 heterodimer and preferentially inhibits cNHEJ at breaks with overhangs by protecting them. We therefore propose that CYREN is a direct cell-cycle-dependent inhibitor of cNHEJ that promotes error-free repair by homologous recombination during cell cycle phases when sister chromatids are present.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: CYREN depletion reactivates cNHEJ in phases S and G2 at deprotected telomeres.
Figure 2: CYREN inhibits NHEJ at intrachromosomal breaks.
Figure 3: CYREN interaction with Ku in S and G2 inhibits cNHEJ.
Figure 4: CYREN prevents overhang processing.

Similar content being viewed by others

References

  1. Lieber, M. R. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu. Rev. Biochem. 79, 181–211 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Heyer, W. D., Ehmsen, K. T. & Liu, J. Regulation of homologous recombination in eukaryotes. Annu. Rev. Genet. 44, 113–139 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Panier, S. & Boulton, S. J. Double-strand break repair: 53BP1 comes into focus. Nat. Rev. Mol. Cell Biol. 15, 7–18 (2014)

    Article  CAS  PubMed  Google Scholar 

  4. Celli, G. B. & de Lange, T. DNA processing is not required for ATM-mediated telomere damage response after TRF2 deletion. Nat. Cell Biol. 7, 712–718 (2005)

    Article  CAS  PubMed  Google Scholar 

  5. Konishi, A. & de Lange, T. Cell cycle control of telomere protection and NHEJ revealed by a ts mutation in the DNA-binding domain of TRF2. Genes Dev. 22, 1221–1230 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Escribano-Díaz, C. et al. A cell cycle-dependent regulatory circuit composed of 53BP1-RIF1 and BRCA1-CtIP controls DNA repair pathway choice. Mol. Cell 49, 872–883 (2013)

    Article  PubMed  Google Scholar 

  7. Zimmermann, M., Lottersberger, F., Buonomo, S. B., Sfeir, A. & de Lange, T. 53BP1 regulates DSB repair using Rif1 to control 5′ end resection. Science 339, 700–704 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chapman, J. R., Taylor, M. R. & Boulton, S. J. Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 47, 497–510 (2012)

    Article  CAS  PubMed  Google Scholar 

  9. Beucher, A. et al. ATM and Artemis promote homologous recombination of radiation-induced DNA double-strand breaks in G2. EMBO J. 28, 3413–3427 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Symington, L. S. & Gautier, J. Double-strand break end resection and repair pathway choice. Annu. Rev. Genet. 45, 247–271 (2011)

    Article  CAS  PubMed  Google Scholar 

  11. Agarwal, S. et al. Isolation, characterization, and genetic complementation of a cellular mutant resistant to retroviral infection. Proc. Natl Acad. Sci. USA 103, 15933–15938 (2006)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Slavoff, S. A., Heo, J., Budnik, B. A., Hanakahi, L. A. & Saghatelian, A. A human short open reading frame (sORF)-encoded polypeptide that stimulates DNA end joining. J. Biol. Chem. 289, 10950–10957 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. van Steensel, B., Smogorzewska, A. & de Lange, T. TRF2 protects human telomeres from end-to-end fusions. Cell 92, 401–413 (1998)

    Article  CAS  PubMed  Google Scholar 

  14. Doksani, Y., Wu, J. Y., de Lange, T. & Zhuang, X. Super-resolution fluorescence imaging of telomeres reveals TRF2-dependent T-loop formation. Cell 155, 345–356 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Okamoto, K. et al. A two-step mechanism for TRF2-mediated chromosome-end protection. Nature 494, 502–505 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Karlseder, J., Broccoli, D., Dai, Y., Hardy, S. & de Lange, T. p53- and ATM-dependent apoptosis induced by telomeres lacking TRF2. Science 283, 1321–1325 (1999)

    Article  CAS  PubMed  Google Scholar 

  17. Smogorzewska, A., Karlseder, J., Holtgreve-Grez, H., Jauch, A. & de Lange, T. DNA ligase IV-dependent NHEJ of deprotected mammalian telomeres in G1 and G2. Curr. Biol. 12, 1635–1644 (2002)

    Article  CAS  PubMed  Google Scholar 

  18. Celli, G. B., Denchi, E. L. & de Lange, T. Ku70 stimulates fusion of dysfunctional telomeres yet protects chromosome ends from homologous recombination. Nat. Cell Biol. 8, 885–890 (2006)

    Article  PubMed  Google Scholar 

  19. Sfeir, A., Kabir, S., van Overbeek, M., Celli, G. B. & de Lange, T. Loss of Rap1 induces telomere recombination in the absence of NHEJ or a DNA damage signal. Science 327, 1657–1661 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sfeir, A. & de Lange, T. Removal of shelterin reveals the telomere end-protection problem. Science 336, 593–597 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lazzerini Denchi, E., Celli, G. & de Lange, T. Hepatocytes with extensive telomere deprotection and fusion remain viable and regenerate liver mass through endoreduplication. Genes Dev. 20, 2648–2653 (2006)

    Article  PubMed  PubMed Central  Google Scholar 

  22. Denchi, E. L. & de Lange, T. Protection of telomeres through independent control of ATM and ATR by TRF2 and POT1. Nature 448, 1068–1071 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  23. Wu, P., van Overbeek, M., Rooney, S. & de Lange, T. Apollo contributes to G overhang maintenance and protects leading-end telomeres. Mol. Cell 39, 606–617 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lam, Y. C. et al. SNMIB/Apollo protects leading-strand telomeres against NHEJ-mediated repair. EMBO J. 29, 2230–2241 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hickson, I. et al. Identification and characterization of a novel and specific inhibitor of the ataxia-telangiectasia mutated kinase ATM. Cancer Res. 64, 9152–9159 (2004)

    Article  CAS  PubMed  Google Scholar 

  26. Leahy, J. J. et al. Identification of a highly potent and selective DNA-dependent protein kinase (DNA-PK) inhibitor (NU7441) by screening of chromenone libraries. Bioorg. Med. Chem. Lett. 14, 6083–6087 (2004)

    Article  CAS  PubMed  Google Scholar 

  27. Menear, K. A. et al. 4-[3-(4-cyclopropanecarbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin-1-one: a novel bioavailable inhibitor of poly(ADP-ribose) polymerase-1. J. Med. Chem. 51, 6581–6591 (2008)

    Article  CAS  PubMed  Google Scholar 

  28. Budke, B. et al. RI-1: a chemical inhibitor of RAD51 that disrupts homologous recombination in human cells. Nucleic Acids Res. 40, 7347–7357 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Grundy, G. J. et al. The Ku-binding motif is a conserved module for recruitment and stimulation of non-homologous end-joining proteins. Nat. Commun. 7, 11242 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Shibata, A. et al. Factors determining DNA double-strand break repair pathway choice in G2 phase. EMBO J. 30, 1079–1092 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Le Poole, I. C. et al. Generation of a human melanocyte cell line by introduction of HPV16 E6 and E7 genes. In Vitro Cell. Dev. Biol. Anim. 33, 42–49 (1997)

    Article  CAS  PubMed  Google Scholar 

  32. Sarbassov, D. D., Guertin, D. A., Ali, S. M. & Sabatini, D. M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307, 1098–1101 (2005)

    Article  ADS  CAS  PubMed  Google Scholar 

  33. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Richardson, C., Moynahan, M. E. & Jasin, M. Double-strand break repair by interchromosomal recombination: suppression of chromosomal translocations. Genes Dev. 12, 3831–3842 (1998)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Campeau, E. et al. A versatile viral system for expression and depletion of proteins in mammalian cells. PLoS One 4, e6529 (2009)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  36. O’Sullivan, R. J. et al. Rapid induction of alternative lengthening of telomeres by depletion of the histone chaperone ASF1. Nat. Struct. Mol. Biol. 21, 167–174 (2014)

    Article  PubMed  PubMed Central  Google Scholar 

  37. O’Sullivan, R. J., Kubicek, S., Schreiber, S. L. & Karlseder, J. Reduced histone biosynthesis and chromatin changes arising from a damage signal at telomeres. Nat. Struct. Mol. Biol. 17, 1218–1225 (2010)

    Article  PubMed  PubMed Central  Google Scholar 

  38. Vannier, J.-B., Pavicic-Kaltenbrunner, V., Petalcorin, M. I. R., Ding, H. & Boulton, S. J. RTEL1 dismantles T loops and counteracts telomeric G4-DNA to maintain telomere integrity. Cell 149, 795–806 (2012)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

All data are archived at the Salk Institute. We thank E. Hendrickson, J. Stark and D. A. Ramsden for support and N. O’Reilly for peptide arrays. N.A. was supported by the Human Frontiers Science Program (LT000284/2013) and N.A. and A.M. by the Paul F. Glenn Center for Biology of Aging Research. J.M. is supported by a Larry Hillblom Foundation Fellowship Grant. S.J.B. is supported by a Wellcome Trust Senior Investigator Award and the Francis Crick Institute (Cancer Research UK), the UK Medical Research Council (FC0010048), and the Wellcome Trust (FC0010048). A.S. is supported by NIH (R01 GM102491), the NCI Cancer Center Support Grant P30 (CA014195), The Leona M. and Harry B. Helmsley Charitable Trust (#2012-PG-MED002), and Dr. Frederick Paulsen Chair/Ferring Pharmaceuticals. The Salk Institute Cancer Center Core Grant (P30CA014195), the NIH (R01GM087476, R01CA174942), the Donald and Darlene Shiley Chair, the Highland Street Foundation, the Fritz B. Burns Foundation and the Emerald Foundation support J.K.

Author information

Authors and Affiliations

Authors

Contributions

Experiments were designed and performed by N.A. (all except Fig. 3c, Extended Data Figs 7b–d, 9), J.M. (Fig. 3c, Extended Data Figs 7b–d, 9), S.J.B. (Extended Data Figs 7a, 9), A.S. (Fig. 3c, Extended Data Figs 7b–d, 9), A.M. (Fig. 2a, b) and J.K. Experiments were performed by A.C. (Figs 2c, d, 3d, f–h, 4, Extended Data Figs 3c, 4, 6, 7f, 8), M.T. (Fig. 1b, c, f, Fig. 3b, Extended Data Figs 1, 3a, b, f, 5), S.G.G. (Extended Data Fig. 9) and M.M. (Extended Data Fig. 9), C.W.B. analysed data (Fig. 4b, c, Extended Data Fig. 8) and N.A. and J.K. wrote the manuscript.

Corresponding author

Correspondence to Jan Karlseder.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

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

Extended data figures and tables

Extended Data Figure 1 CYREN depletion leads to chromatid-type fusions at deprotected telomeres.

a, qRT–PCR measurement of CYREN isoforms expression for Fig. 1b–d. Normalized to ACTB qRT–PCR. CYREN-1, PCR primers target mRNA transcript variant 1. CYREN-2, PCR primers target mRNA transcript variants 2, 3, 4 and 5. CYREN-3, PCR primers target mRNA transcript variant 7. b, Western blot showing TRF2 depletion. For gel source data, see Supplementary Fig. 1. c, Experimental outline of Fig. 1c. HT1080 6TG cells stably transduced with an inducible control shLuci or one of three shCYREN RNAs were infected with shControl or shTRF2 on day 0. shTRF2-transduced cells were selected with puromycin and shCYREN expression was induced with doxycycline on day 2. Cells were collected for fusion analysis on day 5. d, Partial metaphase spreads of functional (shControl) and deprotected (shTRF2) telomeres after CYREN depletion. Green arrows, chromosome-type fusions. Blue arrows, chromatid-type fusions. e, Percentage of cells with fusions ± upper and lower value of 95% confidence intervals, Wilson–Brown test. ****P < 0.0001, ***P < 0.001. Fisher’s exact test, two-sided. n, number of metaphases analysed. f, Mean percentage of chromosome ends fused by sister telomere associations. Error bars, s.e.m. One-way ANOVA, Sidak’s multiple comparison test. n, number of metaphases analysed. g, Number of metaphases analysed, total telomere and fusions counted.

Extended Data Figure 2 Chromatid-type fusions induced by CYREN depletion in IMR90 fibroblasts.

a, Western blot showing TRF2 depletion. For gel source data, see Supplementary Fig. 1. b, qRT–PCR measurement of CYREN isoforms expression after siRNA knockdown. Normalized to ACTB qRT–PCR. CYREN-1, PCR primers target mRNA transcript variant 1. CYREN-2, PCR primers target mRNA transcript variants 2, 3, 4 and 5. CYREN-3, PCR primers target mRNA transcript variant 7. c, Representative images of partial metaphase spreads of functional (shControl) and deprotected (shTRF2) telomeres after CYREN depletion. d, Mean percentage of fused chromosome ends per metaphase. Error bars, s.e.m. **P < 0.01. One-way ANOVA, Sidak’s multiple comparison test. n, number of metaphases analysed.

Extended Data Figure 3 CYREN prevents cNHEJ in S and G2 at deprotected telomeres.

a, Schematic of CO-FISH. Chromatid-type fusions involving leading and lagging strands. b, Percentage of fusions ± upper and lower value of 95% confidence intervals, Wilson–Brown test. 126 fusions counted. c, Western blot showing knockdown of ATM, ligase 4, DNA-PKcs and ligase 3 in Fig. 2c. For gel source data, see Supplementary Fig. 1. d, Experimental timeline for Fig. 2c. CYRENWT and CYRENKO HT1080 cells were infected with shTRF2 on day 0, followed by transfection with siRNAs on day 2. On day 3, shTRF2-infected cells were selected with puromycin and cells were collected for fusion analysis on day 5. e, Experimental timeline for panel f. HT1080 6TG cells were stably transduced with shTRF2 on day 0, followed by transfection with non-targeting (NT) or CYREN siRNAs on day 2. On day 3, shTRF2-infected cells were selected with puromycin and inhibitors were added. Cells were collected for fusion analysis on day 5. f, Percentage of cells with fusions ± upper and lower value of 95% confidence intervals, Wilson–Brown test. Cells were treated for 48 h with DMSO or the following inhibitors: ATMi (KU-55933) 10 μM, DNA-PKcsi (NU-7441) 1 μM, PARPi (olaparib) 10 μM, RAD51i (RI-1) 20 μM. ****P < 0.0001, NS, not significant. Fisher’s exact test, two-sided. Experiment shown is representative of two biological replicates.

Extended Data Figure 4 CYREN regulates the DSB pathway choice at intrachromosomal breaks.

a, Experimental timeline for Fig. 2a, b. CYRENWT and CYRENKO clonal HT1080 were synchronized by double thymdine block, and irradiated at 2 Gy at 2 h, 6 h or 10 h after thymidine release, corresponding to the S, G2 or G1 phases of the cell cycle, respectively. Cells were arrested for immunofluorescence or chromosome spreads 26 h after thymidine release. b, Cell cycle profiles of cells used in Fig. 2a, b, 2 h, 6 h and 10 h after thymidine release. 20,000 cells were analysed. c, Representative flow cytometry controls for the DSB repair reporter. One million cells per sample were analysed. d, Experimental outline of Fig. 2d. A single clone of HT1080 cells transduced with the DSB repair reporter was isolated and transfected with Cas9 and sgCYREN. Single clones were isolated and genotyped. Selected CYRENWT and CYRENKO clones were then transfected with ISce1 and the HR donor, followed by flow cytometry analysis 48 h later. e, Cell cycle distribution of the CYRENWT and CYRENKO clones obtained by flow cytometry of propidium iodide and BrdU-labelled cells. 80,000 cells were analysed.

Extended Data Figure 5 CYREN does not regulate repair of replication-induced DSBs.

a, Representative images of chromosomes from b. b, Percentage of metaphases with radial chromosomes ± upper and lower value of 95% confidence intervals, Wilson–Brown test. n, number of metaphases analysed. Experiment shown is representative of two biological replicates. c, Per cent survival with increasing concentrations of PARP inhibitor olaparib.

Extended Data Figure 6 CYREN isoforms 1 and 2 inhibit cNHEJ.

a, Anti-Flag western blot on whole-cell extracts of HT1080 6TG cells expressing Flag–CYREN isoforms used in Fig. 4b. Asterisk indicates two nonspecific bands. For gel source data, see Supplementary Fig. 1. b, Experimental outline of Fig. 4b. HT1080 6TG cells were stably transfected with pcDNA3 empty Flag vector or pcDNA3 expressing CYREN-1–Flag, CYREN-2–Flag or CYREN-3–Flag. Cells were selected and infected with shTRF2 on day 0, followed by transfection with a control pool of non-targeting (NT) siRNAs or a pool of siRNAs targeting the 3′ UTR of CYREN. shTRF2-infected cells were selected on day 3 and cells were collected for fusion analysis on day 5. c, Schematic representation of N-terminal 3×Flag endogenous tagging of CYREN-1 and CYREN-2. d, Sequencing of the N-terminal 3×Flag–CYREN tagged allele. e, Anti-Flag western blot of whole-cell extracts from HT1080 cells and HT1080 cells with endogenous N-terminally 3×Flag-tagged exon 1 of C7orf49. Upper band, CYREN-1. Lower band, CYREN-2. Increasing amounts of protein extract were loaded (5, 10, 15, 20 μl). For gel source data, see Supplementary Fig. 1. f, Schematic representation of C-terminal 3×Flag endogenous tagging of CYREN-1 and CYREN-3. g, Sequencing of the C-terminal CYREN–3×Flag tagged allele. h, Flag western blot of HT1080 6TG cells without and with endogenous tagged C-terminal CYREN–3×Flag. Increasing amounts of protein extracts were loaded (5, 10, 15, 20 μl). For gel source data, see Supplementary Fig. 1. i, Flag western blot of 3×Flag–CYREN tagged HT1080 cells following CYREN knockdown by siRNA. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 7 CYREN interaction with Ku in S and G2 inhibits cNHEJ.

a, Immunoblotting of a peptide binding array of full-length CYREN-1. Each dot represents 20 amino acids of CYREN-1 with a 19-amino acid overlap with the previous and following peptides. Upper panel, Ponceau stain. Middle panels, duplicate incubation with Ku70/80 recombinant proteins and immunoblotting with anti-Ku70 antibody. Lower panel, control immunoblotting with anti-Ku70 antibody without incubation with recombinant Ku70/80. For gel source data, see Supplementary Fig. 1. b, Alanine scan of CYREN-1 on residues 9–46. Flag immunoprecipitation of protein extracts from HEK293T cells transfected with pCDNA3.1 plasmids expressing wild-type CYREN-1–Flag or each of the single residues mutated to alanine. Total lysate and Flag-immunoprecipitate were then immunoblotted with anti-Flag or Ku70 antibodies. For gel source data, see Supplementary Fig. 1. c, Protein alignment of CYREN-1 KBM among vertebrates. d, Photo-crosslinked pulldown of a BPA–BIO–CYREN(2–24) peptide in HEK293T cell protein extract, followed by immunoblotting with anti-Ku70 and Ku80 antibodies. Single and double plus symbols, 25 μM and 50 μM of BPA–BIO–CYREN, respectively. 100 μM of CYREN(2–24) free peptide was used as a competitor. For gel source data, see Supplementary Fig. 1. e, Experimental outline of Fig. 3d. HT1080 6TG cells stably expressing an inducible control GFP or wild-type or mutant CYREN-1–3×Flag were transduced with shTRF2 on day 0 and transfected with a control (NT) pool of siRNAs or a pool of siRNAs targeting the 3′ UTR of CYREN on day 2. Expression of wild-type or mutant CYREN-1 was induced on day 3 and cells were collected on day 5. f, Flag western blot of endogenous N-terminal 3×Flag-tagged CYREN-1 and CYREN-2 cells following double thymidine synchronization. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 8 CYREN inhibits cNHEJ preferentially at breaks with overhangs by preventing processing.

a, Percentage of cells using HR to repair Cas9-induced breaks. Details of four CYRENWT and four CYRENKO clones used in Fig. 4b. b, Deletion profiles of repair of Cas9-induced breaks. Detail profiles of four CYRENWT and four CYRENKO clones used in Fig. 4c. Blue line in blunt ends: break site. Blue area: overhang region created by the pair of sgRNAs. c, Average percentage of mClover+ cells in four CYRENWT clones, four CYRENKO clones, four CYRENKO clones complemented with wild-type CYREN and four CYRENKO clones complemented with mutant CYREN (RPW-AAA), in three independent experiments, normalized to wild-type. Tukey box and whiskers; box represents 25th to 75th percentiles, upper whiskers represent 75th percentile plus 1.5 interquartile distance, lower whiskers represent 25th percentile minus 1.5 interquartile distance. *P < 0.05. Unpaired t-test. In each experiment, 100,000 cells per sample were analysed. d, Deletion profiles of repair of Cas9 breaks. Average percentage of deletion in CYRENWT clones, CYRENKO clones, CYRENKO clones complemented with wild-type CYREN and CYRENKO clones complemented with mutant CYREN (RPW-AAA). Error bars, s.e.m.

Extended Data Figure 9 CYREN does not promote cNHEJ in vitro.

a, In vitro cNHEJ assay using CYRENWT and CYRENKO cells. Left, immunoblots of extracts used in the assay. Middle, in vitro ligation assay. Right, quantification. Error bars, s.d., three independent experiments. For gel source data, see Supplementary Fig. 1. b, In vitro cNHEJ assay using CYRENWT cells and increasing amounts of recombinant wild-type CYREN and CYRENΔKu mutant. Right, quantification. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 10 Maps of plasmids used in Figs 1, 2, 3, 4.

Supplementary information

Supplementary Figure

This file contains the uncropped blots. (PDF 3394 kb)

Reporting Summary (PDF 94 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Arnoult, N., Correia, A., Ma, J. et al. Regulation of DNA repair pathway choice in S and G2 phases by the NHEJ inhibitor CYREN. Nature 549, 548–552 (2017). https://doi.org/10.1038/nature24023

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature24023

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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