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:

CtIP tetramer assembly is required for DNA-end resection and repair

Abstract

Mammalian CtIP protein has major roles in DNA double-strand break (DSB) repair. Although it is well established that CtIP promotes DNA-end resection in preparation for homology-dependent DSB repair, the molecular basis for this function has remained unknown. Here we show by biophysical and X-ray crystallographic analyses that the N-terminal domain of human CtIP exists as a stable homotetramer. Tetramerization results from interlocking interactions between the N-terminal extensions of CtIP's coiled-coil region, which lead to a 'dimer-of-dimers' architecture. Through interrogation of the CtIP structure, we identify a point mutation that abolishes tetramerization of the N-terminal domain while preserving dimerization in vitro. Notably, we establish that this mutation abrogates CtIP oligomer assembly in cells, thus leading to strong defects in DNA-end resection and gene conversion. These findings indicate that the CtIP tetramer architecture described here is essential for effective DSB repair by homologous recombination.

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: CtIP-NTD (amino acids 18–145) exists as a tetramer in solution.
Figure 2: Crystal structure of the tetramerization domain of human CtIP (CtIP-nNTD; amino acids 18–52).
Figure 3: Details of the homotypic interactions responsible for tetrameric assembly and dimerization of CtIP.
Figure 4: CtIP mutation L27E abrogates tetramer assembly while preserving dimerization.
Figure 5: CtIP mutation L27E impairs HR and, to a lesser extent, MMEJ.
Figure 6: CtIP mutation L27E impairs CtIP accumulation at DNA-damage sites and DNA-end resection.
Figure 7: Model for the functional architecture of human CtIP.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

References

  1. Jackson, S.P. & Bartek, J. The DNA-damage response in human biology and disease. Nature 461, 1071–1078 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Krejci, L., Altmannova, V., Spirek, M. & Zhao, X. Homologous recombination and its regulation. Nucleic Acids Res. 40, 5795–5818 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 

  4. Fusco, C., Reymond, A. & Zervos, A.S. Molecular cloning and characterization of a novel retinoblastoma-binding protein. Genomics 51, 351–358 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Schaeper, U., Subramanian, T., Lim, L., Boyd, J.M. & Chinnadurai, G. Interaction between a cellular protein that binds to the C-terminal region of adenovirus E1A (CtBP) and a novel cellular protein is disrupted by E1A through a conserved PLDLS motif. J. Biol. Chem. 273, 8549–8552 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Wong, A.K. et al. Characterization of a carboxy-terminal BRCA1 interacting protein. Oncogene 17, 2279–2285 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Li, S. et al. Functional link of BRCA1 and ataxia telangiectasia gene product in DNA damage response. Nature 406, 210–215 (2000).

    Article  CAS  PubMed  Google Scholar 

  8. Chen, P.L. et al. Inactivation of CtIP leads to early embryonic lethality mediated by G1 restraint and to tumorigenesis by haploid insufficiency. Mol. Cell. Biol. 25, 3535–3542 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu, F. & Lee, W.-H. CtIP activates its own and cyclin D1 promoters via the E2F/RB pathway during G1/S progression. Mol. Cell. Biol. 26, 3124–3134 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yu, X. & Chen, J. DNA damage-induced cell cycle checkpoint control requires CtIP, a phosphorylation-dependent binding partner of BRCA1 C-terminal domains. Mol. Cell. Biol. 24, 9478–9486 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sartori, A.A. et al. Human CtIP promotes DNA end resection. Nature 450, 509–514 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yun, M.H. & Hiom, K. CtIP-BRCA1 modulates the choice of DNA double-strand-break repair pathway throughout the cell cycle. Nature 459, 460–463 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Huertas, P. et al. CDK targets Sae2 to control DNA-end resection and homologous recombination. Nature 455, 689–692 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhang, Y. & Jasin, M. An essential role for CtIP in chromosomal translocation formation through an alternative end-joining pathway. Nat. Struct. Mol. Biol. 18, 80–84 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Lee-Theilen, M., Matthews, A.J., Kelly, D., Zheng, S. & Chaudhuri, J. CtIP promotes microhomology-mediated alternative end joining during class-switch recombination. Nat. Struct. Mol. Biol. 18, 75–79 (2011).

    CAS  PubMed  Google Scholar 

  16. You, Z. et al. CtIP links DNA double-strand break sensing to resection. Mol. Cell 36, 954–969 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Huertas, P. & Jackson, S.P. Human CtIP mediates cell cycle control of DNA end resection and double strand break repair. J. Biol. Chem. 284, 9558–9565 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yuan, J. & Chen, J. N terminus of CtIP is critical for homologous recombination-mediated double-strand break repair. J. Biol. Chem. 284, 31746–31752 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Dubin, M.J. et al. Dimerization of CtIP, a BRCA1- and CtBP-interacting protein, is mediated by an N-terminal coiled-coil motif. J. Biol. Chem. 279, 26932–26938 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Stokes, P.H., Thompson, L.S., Marianayagam, N.J. & Matthews, J.M. Dimerization of CtIP may stabilize in vivo interactions with the Retinoblastoma-pocket domain. Biochem. Biophys. Res. Commun. 354, 197–202 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Wang, H. et al. CtIP protein dimerization is critical for its recruitment to chromosomal DNA double-stranded breaks. J. Biol. Chem. 287, 21471–21480 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Säbel, C.E., Shepherd, J.L. & Siemann, S. A direct spectrophotometric method for the simultaneous determination of zinc and cobalt in metalloproteins using 4-(2-pyridylazo)resorcinol. Anal. Biochem. 391, 74–76 (2009).

    Article  PubMed  Google Scholar 

  23. Certo, M.T. et al. Tracking genome engineering outcome at individual DNA breakpoints. Nat. Methods 8, 671–676 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gomez-Cabello, D., Jimeno, S., Fernández-Ávila, M.J. & Huertas, P. New tools to study DNA double-strand break repair pathway choice. PLoS ONE 8, e77206 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. McVey, M. & Lee, S.E. MMEJ repair of double-strand breaks (director's cut): deleted sequences and alternative endings. Trends Genet. 24, 529–538 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sharma, A., Singh, K. & Almasan, A. Histone H2AX phosphorylation: a marker for DNA damage. Methods Mol. Biol. 920, 613–626 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Huertas, P. DNA resection in eukaryotes: deciding how to fix the break. Nat. Struct. Mol. Biol. 17, 11–16 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Forment, J.V., Walker, R.V. & Jackson, S.P. A high-throughput, flow cytometry-based method to quantify DNA-end resection in mammalian cells. Cytometry A 81, 922–928 (2012).

    Article  PubMed  Google Scholar 

  29. Pommier, Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat. Rev. Cancer 6, 789–802 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Saleh-Gohari, N. et al. Spontaneous homologous recombination is induced by collapsed replication forks that are caused by endogenous DNA single-strand breaks. Mol. Cell. Biol. 25, 7158–7169 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Makharashvili, N. et al. Catalytic and noncatalytic roles of the CtIP endonuclease in double-strand break end resection. Mol. Cell 54, 1022–1033 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang, H. et al. CtIP maintains stability at common fragile sites and inverted repeats by end resection-independent endonuclease activity. Mol. Cell 54, 1012–1021 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hopfner, K.-P. et al. The Rad50 zinc-hook is a structure joining Mre11 complexes in DNA recombination and repair. Nature 418, 562–566 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Lobachev, K.S., Gordenin, D.A. & Resnick, M.A. The Mre11 complex is required for repair of hairpin-capped double-strand breaks and prevention of chromosome rearrangements. Cell 108, 183–193 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Clerici, M., Mantiero, D., Lucchini, G. & Longhese, M.P. The Saccharomyces cerevisiae Sae2 protein promotes resection and bridging of double strand break ends. J. Biol. Chem. 280, 38631–38638 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Lengsfeld, B.M., Rattray, A.J., Bhaskara, V., Ghirlando, R. & Paull, T.T. Sae2 Is an endonuclease that processes hairpin DNA cooperatively with the Mre11/Rad50/Xrs2 complex. Mol. Cell 28, 638–651 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fu, Q. et al. Phosphorylation-regulated transitions in an oligomeric state control the activity of the Sae2 DNA repair enzyme. Mol. Cell. Biol. 34, 778–793 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Blanc, E. et al. Refinement of severely incomplete structures with maximum likelihood in BUSTER-TNT. Acta Crystallogr. D Biol. Crystallogr. 60, 2210–2221 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Peränen, J., Rikkonen, M., Hyvonen, M. & Kaariainen, L. T7 vectors with modified T7lac promoter for expression of proteins in Escherichia coli. Anal. Biochem. 236, 371–373 (1996).

    Article  PubMed  Google Scholar 

  40. Sreerama, N. & Woody, R.W. Estimation of protein secondary structure from circular dichroism spectra: comparison of CONTIN, SELCON, and CDSSTR methods with an expanded reference set. Anal. Biochem. 287, 252–260 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Whitmore, L. & Wallace, B.A. Protein secondary structure analyses from circular dichroism spectroscopy: methods and reference databases. Biopolymers 89, 392–400 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Kabsch, W. Xds. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Collaborative Computational Project, N. 4. The CCP4 suite: programs for protein crystallography. Acta Crystallogr. D Biol. Crystallogr. 50, 760–763 (1994).

  44. McCoy, A.J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kelley, L.A. & Sternberg, M.J.E. Protein structure prediction on the Web: a case study using the Phyre server. Nat. Protoc. 4, 363–371 (2009).

    CAS  PubMed  Google Scholar 

  46. Adams, P.D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Emsley, P. & Cowtan, K. Coot: Model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    PubMed  Google Scholar 

  48. Dignam, J.D., Lebovitz, R.M. & Roeder, R.G. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 11, 1475–1489 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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 

  50. Limoli, C.L. & Ward, J.F. A new method for introducing double-strand breaks into cellular DNA. Radiat. Res. 134, 160–169 (1993).

    Article  CAS  PubMed  Google Scholar 

  51. Bekker-Jensen, S. et al. Spatial organization of the mammalian genome surveillance machinery in response to DNA strand breaks. J. Cell Biol. 173, 195–206 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Kilkenny for help with the collection of X-ray diffraction data, A. Sharff and P. Keller for help with X-ray data processing and J.D. Maman for assistance with SEC-MALS. This work was supported by a Wellcome Trust Senior Research Fellowship award in basic biomedical sciences (L.P.), an Isaac Newton Trust research grant (L.P. and O.R.D.) and a Cambridge Overseas Trust PhD studentship (M.D.S.). Research in the laboratory of S.P.J. is funded by Cancer Research UK (CRUK; programme grant C6/A11224), the European Research Council and the European Community Seventh Framework Programme (grant agreement no. HEALTH-F2-2010-259893 (DDResponse)). Core funding is provided by Cancer Research UK (C6946/A14492) and the Wellcome Trust (WT092096). S.P.J. receives his salary from the University of Cambridge, supplemented by CRUK. J.V.F. is funded by Cancer Research UK programme grant C6/A11224 and the Ataxia Telangiectasia Society. R.B. and J.C. are funded by Cancer Research UK programme grant C6/A11224. Y.G. and M.D. are funded by the European Research Council grant DDREAM.

Author information

Authors and Affiliations

Authors

Contributions

O.R.D. and M.S. expressed and purified the CtIP proteins and performed the biochemical and biophysical experiments; C.R.M. and N.J.R. performed the electrophoretic mobility shift assays; O.R.D. determined the X-ray crystal structure of CtIP-nNTD; J.V.F. performed the TLR and MMEJ assays, the DNA-end resection assays, the immunofluorescence assays (with help from Y.G.), the GFP-CtIP quantification on fluorescence-activated cell sorting and the coimmunoprecipitation experiments; R.B. performed the gel-filtration analysis of Flag-CtIP from HEK-293T cell extracts and created the Flag-tagged CtIP constructs; J.C. established the U2OS-TLR and the U2OS-MMEJ stable cell lines; M.D. established the U2OS GFP-CtIP cell lines; J.V.F. and Y.G. established the TLR system in U2OS cells; O.R.D., J.V.F., S.P.J. and L.P. designed experiments and wrote the paper.

Corresponding authors

Correspondence to Stephen P Jackson or Luca Pellegrini.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Multiple sequence alignment of the CtIP N-terminal domain, purified CtIP protein constructs and details of the 2FoFc electron density map of CtIP-NTD.

(a) Multiple sequence alignment, including the predicted secondary structure (red rods indicate regions of predicted α-helix) and coiled-coil regions; the probability of coiled-coil formation is indicated by ‘-’ (<50%), ‘c’ (50-90%) or ‘C’ (>90%). Protein sequence analysis was performed with the Jalview suite of bioinformatics tools (www.jalview.org). Hs: Homo sapiens, Mm: Mus musculus, Ec: Equus caballus, Oc: Oryctolagus cuniculus, Gg: Gallus gallus, Xl: Xenopus laevis, Dr: Danio rerio. (b) SDS-PAGE analysis of purified recombinant CtIP protein samples: NTD, NTD-DM (C89A, C92A), nNTD, cNTD, cNTD-DM (C89A, C92A) and CTD visualised by Coomassie staining. (c) Details of the 2Fo - Fc electron density map, contoured at 1.2 rmsd, superimposed on the refined crystallographic model of CtIP-NTD. The figure shows the map in the vicinity of amino acid W24. The map is shown as thin mesh in light purple, the protein atoms are shown in ball-and-stick representation and coloured according to chemical identity. The figure was prepared in Coot

Supplementary Figure 2 CD analysis of CtIP N-terminal-domain constructs and microPIXE analysis of the CtIP N-terminal domain.

Circular dichroism (CD) analysis of CtIP N-terminal domain constructs. (a, c, e) Far-UV CD spectra recorded between 260 and 185 nm, in mean residue ellipticity, MRE ([θ]) (1000x deg.cm2.dmol-1.residue-1), with α-helical content calculated through deconvolution using the CDSSTR algorithm with normalised root-mean-square deviation values (nrmsd) as shown. (b, d, f) CD thermal denaturation data recorded between 5 and 95°C, in mean residue ellipticity at 222 nm ([θ]222). (a) CD spectra and (b) thermal denaturation of CtIP-NTD (solid line) and CtIP-NTD-DM (dashed line). (c) CD spectra and (d) thermal denaturation of CtIP-cNTD (solid line) and CtIP-cNTD-DM (dashed line). (e) CD spectra and (f) thermal denaturation of CtIP-nNTD. (g) CtIP-NTD was analysed in buffer containing KBr. Rutherford backscattering (RBS) spectrum and (h) PIXE spectrum with raw data shown as black dots and the modelling fitting as red lines. (i) Zinc was detected at a mean atomic ratio to sulphur of 0.095, which given the presence of three methionine and three cysteine residues per chain, corresponds to a CtIP-NTD:Zn2+ ratio of 1.8:1.

Supplementary Figure 3 SEC-MALS analysis of MBP-CtIP-nNTD and F20E CtIP-NTD.

(a) MBP-CtIP-nNTD eluted in a single peak corresponding to a molecular weight of 187 kDa; its theoretical tetramer size is 197 kDa. For comparison, free MBP eluted in a single peak of 44.5 kDa, corresponding to its theoretical monomer size of 44.7 kDa. The light scattering (LS) as relative Raleigh ratio and the differential Reflective Index (dRI) are drawn as solid and dashed lines, respectively. The value for the fitted molecular mass (Mr) of MBP-CtIP-nNTD is shown as diamond shapes across its elution peak. The predicted molecular masses of monomeric MBP-CtIP-nNTD and free MBP are shown in brackets above the respective elution peak. (b) F20E CtIP-NTD forms two distinct species of 62.3 kDa and 33.2 kDa, corresponding to its theoretical tetramer and dimer sizes of 63.2 kDa and 31.6 kDa, respectively. The light scattering (LS) as relative Raleigh ratio and the differential Reflective Index (dRI) are drawn as solid and dashed lines, respectively. The values for the fitted molecular masses (Mr) of dimeric and tetrameric F20E CtIP-NTD are shown as diamond shapes across the respective elution peaks. The predicted molecular mass of monomeric F20E CtIP-nNTD is shown in brackets above the elution peaks.

Supplementary Figure 4 Experimental procedure and gating scheme for the TLR system.

(a) TLR cells are not fluorescent due to the fact that the TLR cassette contains an in-frame truncated version of the eGFP gene and full-length mCherry is out of frame. When TLR cells are transfected with an I-SceI nuclease encoded in a plasmid also expressing an infrared fluorescent protein (IFP) and an exogenous donor template containing the missing part of eGFP that also expresses a blue fluorescent protein (BFP), the different repair outcomes will give different fluorescent cells. If the double-strand break (DSB) is repaired by gene conversion (homologous recombination), eGFP is restored and the cells fluoresce in green. If the DSB is repaired by mutagenic end joining causing a +2 frameshift, mCherry gets in frame and its expression yields red fluorescence. The T2A “dis-linker” between eGFP and mCherry allows the downstream-encoded mCherry to escape degradation of the misfolded protein encoded in the +3 reading frame of eGFP. (b) Experimental outline of the TLR assay. (c-h) Gating scheme for a TLR assay. (c) Discrimination of cells over debris using a forward scatter vs. side scatter plot (areas). (d) Discrimination of cell singlets over doublets using a forward scatter (area) vs. side scatter (width) plot. (e) Selection of events for quantification. Dot plot showing the intensity of BFP (donor; x axis) vs. intensity of IFP (nuclease; y axis). Cells with negative staining for both BFP and IFP are depicted in (f) to show the absence of eGFP or mCherry fluorescence in non-transfected cells, and to establish quantification gates. Cells counted positive for both BFP and IFP (that is, transfected with both the donor and the nuclease) are represented in (g), where quantification gates are the same as the ones established in (f). At least 10,000 events were analysed in the double-positive population. (h) Due to the fact that the amount of donor transfected in the cells affects the outcome of the assay, the intensity of the BFP signal in the double-positive cells (geometric mean of BFP signal in ‘BFP+ IFP+’ cells) was calculated for normalisation purposes (see Methods). (i) Mutagenic end-joining (mutEJ) rates in the TLR-CtIP complementation system. Only expression of the wild-type version of FLAG-CtIP reduced the rate of mutEJ to values similar to the control (cells transfected with control siRNA). EV: empty vector. All quantifications are shown as the average of three independent experiments. Error bars are ± SEM.

Supplementary Figure 5 CtIP expression levels in the different stable cell lines generated and fluorescence-activated cell sorting quantification of GFP-CtIP accumulation on damaged cells.

(a) CtIP expression levels in the different U2OS-MMEJ FLAG-CtIP clones. Samples were collected 48 h after siRNA transfection. CtIP antibody was a gift from R. Baer. PARP-1 antibody (Cell Signalling) was used as loading control. (b) GFP-CtIP expression levels in the U2OS stable cell lines. RPA34-20 (Merck) antibody was used as loading control. (c) A representative complete dataset for quantification of GFP-CtIP accumulation on damaged cells. Dot plots representing, for each dataset, gating and quantification (in the y axis) of the GFP-positive cells (top panels) and the γH2AX-positive cells (bottom panels) taking into account their DNA content (x axis). Quantification gates were always established using the untreated samples of each dataset. At least 10,000 events were analysed per sample.

Supplementary Figure 6 A representative complete dataset for the DNA-end resection assay.

Dot plots representing, for each dataset, gating and quantification (in the y axis) of the RPA-positive cells (top panels) and the γH2AX-positive cells (bottom panels) taking into account their DNA content (x axis). Quantification gates were always established using the untreated samples of each dataset and were defined as to avoid quantification of RPA chromatinization due to DNA replication. Note that as camptothecin (CPT) only causes DNA double-strand breaks in S-phase cells, both RPA- and γH2AX-positive cells only appear in that cell cycle stage. At least 10,000 events were analyzed per sample.

Supplementary Figure 7 CtIP-CTD, but not CtIP-NTD, interacts with double-stranded DNA.

Electrophoretic mobility shift assay measuring the ability of CtIP NTD and CTD regions to interact with linear, double-stranded DNA. In the experiment, increasing amounts (0, 0.5, 1.0, 2.0, 5.0 and 10μM) of the CtIP protein were incubated with a 1μM sample of 5’ fluorescein-labeled 200bp double-stranded DNA. After incubation, each sample was resolved by electrophoresis on agarose gel and visualised under UV light.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 5362 kb)

Supplementary Data Set 1

Uncropped versions of western blots presented in Figures 4 and 6. (PDF 7301 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Davies, O., Forment, J., Sun, M. et al. CtIP tetramer assembly is required for DNA-end resection and repair. Nat Struct Mol Biol 22, 150–157 (2015). https://doi.org/10.1038/nsmb.2937

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2937

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