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:

The selection process of licensing a DNA mismatch for repair

Abstract

DNA mismatch repair detects and removes mismatches from DNA by a conserved mechanism, reducing the error rate of DNA replication by 100- to 1,000-fold. In this process, MutS homologs scan DNA, recognize mismatches and initiate repair. How the MutS homologs selectively license repair of a mismatch among millions of matched base pairs is not understood. Here we present four cryo-EM structures of Escherichia coli MutS that provide snapshots, from scanning homoduplex DNA to mismatch binding and MutL activation via an intermediate state. During scanning, the homoduplex DNA forms a steric block that prevents MutS from transitioning into the MutL-bound clamp state, which can only be overcome through kinking of the DNA at a mismatch. Structural asymmetry in all four structures indicates a division of labor between the two MutS monomers. Together, these structures reveal how a small conformational change from the homoduplex- to heteroduplex-bound MutS acts as a licensing step that triggers a dramatic conformational change that enables MutL binding and initiation of the repair cascade.

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: Structures of MutS at consecutive steps of the repair cascade.
Fig. 2: Licensing of DNA mismatch repair.
Fig. 3: An intermediate state between mismatch and MutL binding.
Fig. 4: MutS−DNA−MutLLN40 complex.
Fig. 5: Kinking of the lever domain promotes clamp formation.
Fig. 6: MutS heterodimers are mismatch-repair competent.

Similar content being viewed by others

Data availability

Cryo-EM maps and atomic models have been deposited in the Electron Microscopy Database and Protein Data Bank, respectively, under accession codes EMD-11791, PDB 7AI5, EMD-11792, PDB 7AI6, EMD-11793, PDB 7AI7, EMD-11794, PDB 7AIB, EMD-11795 and PDB 7AIC. Other requests should be addressed to Meindert Lamers (m.h.lamers@lumc.nl), Titia K. Sixma (t.sixma@nki.nl) or Rafael Fernández-Leiro (rfleiro@cnio.es). Source data are provided with this paper.

References

  1. Li, Z., Pearlman, A. H. & Hsieh, P. DNA mismatch repair and the DNA damage response. DNA Repair (Amst.) 38, 94–101 (2016).

    Article  CAS  Google Scholar 

  2. Jiricny, J. Postreplicative mismatch repair. Cold Spring Harb. Perspect. Biol. 5, a012633 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Lamers, M. H. et al. The crystal structure of DNA mismatch repair protein MutS binding to a G·T mismatch. Nature 407, 711–717 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Natrajan, G. et al. Structures of Escherichia coli DNA mismatch repair enzyme MutS in complex with different mismatches: a common recognition mode for diverse substrates. Nucleic Acids Res. 31, 4814–4821 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Obmolova, G., Ban, C., Hsieh, P. & Yang, W. Crystal structures of mismatch repair protein MutS and its complex with a substrate DNA. Nature 407, 703–710 (2000).

    Article  CAS  PubMed  Google Scholar 

  6. Gupta, S., Gellert, M. & Yang, W. Mechanism of mismatch recognition revealed by human MutSβ bound to unpaired DNA loops. Nat. Struct. Mol. Biol. 19, 72–78 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Warren, J. J. et al. Structure of the human MutSα DNA lesion recognition complex. Mol. Cell 26, 579–592 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Gradia, S. et al. hMSH2–hMSH6 forms a hydrolysis-independent sliding clamp on mismatched DNA. Mol. Cell 3, 255–261 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Blackwell, L. J., Bjornson, K. P., Allen, D. J. & Modrich, P. Distinct MutS DNA-binding modes that are differentially modulated by ATP binding and hydrolysis. J. Biol. Chem. 276, 34339–34347 (2001).

  10. Heo, S.-D., Cho, M., Ku, J. K. & Ban, C. Steady-state ATPase activity of E. coli MutS modulated by its dissociation from heteroduplex DNA. Biochem. Biophys. Res. Commun. 364, 264–269 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Jeong, C. et al. MutS switches between two fundamentally distinct clamps during mismatch repair. Nat. Struct. Mol. Biol. 18, 379–385 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Schofield, M. J., Nayak, S., Scott, T. H., Du, C. & Hsieh, P. Interaction of Escherichia coli MutS and MutL at a DNA mismatch. J. Biol. Chem. 276, 28291–28299 (2001).

  13. Acharya, S., Foster, P. L., Brooks, P. & Fishel, R. The coordinated functions of the E. coli MutS and MutL proteins in mismatch repair. Mol. Cell 12, 233–246 (2003).

  14. Junop, M. S., Yang, W., Funchain, P., Clendenin, W. & Miller, J. H. In vitro and in vivo studies of MutS, MutL and MutH mutants: correlation of mismatch repair and DNA recombination. DNA Repair (Amst.) 2, 387–405 (2003).

    Article  CAS  Google Scholar 

  15. Hall, M. C. & Matson, S. W. The Escherichia coli MutL protein physically interacts with MutH and stimulates the MutH-associated endonuclease activity. J. Biol. Chem. 274, 1306–1312 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Kadyrov, F. A., Dzantiev, L., Constantin, N. & Modrich, P. Endonucleolytic function of MutLα in human mismatch repair. Cell 126, 297–308 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Fukui, K., Nishida, M., Nakagawa, N., Masui, R. & Kuramitsu, S. Bound nucleotide controls the endonuclease activity of mismatch repair enzyme MutL. J. Biol. Chem. 283, 12136–12145 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Pluciennik, A. et al. PCNA function in the activation and strand direction of MutLα endonuclease in mismatch repair. Proc. Natl Acad. Sci. USA 107, 16066–16071 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pillon, M. C., Miller, J. H. & Guarné, A. The endonuclease domain of MutL interacts with the β sliding clamp. DNA Repair (Amst.) 10, 87–93 (2011).

    Article  CAS  Google Scholar 

  20. Ban, C. & Yang, W. Crystal structure and ATPase activity of MutL: implications for DNA repair and mutagenesis. Cell 95, 541–552 (1998).

    Article  CAS  PubMed  Google Scholar 

  21. Groothuizen, F. S. et al. MutS/MutL crystal structure reveals that the MutS sliding clamp loads MutL onto DNA. Elife 4, e06744 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Gorman, J. et al. Single-molecule imaging reveals target-search mechanisms during DNA mismatch repair. Proc. Natl Acad. Sci. USA 109, E3074–E3083 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cho, W.-K. et al. ATP alters the diffusion mechanics of MutS on mismatched DNA. Structure 20, 1264–1274 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nakane, T., Kimanius, D., Lindahl, E. & Scheres, S. H. Characterisation of molecular motions in cryo-EM single-particle data by multi-body refinement in RELION. Elife 7, e36861 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Yang, Y., Sass, L. E., Du, C., Hsieh, P. & Erie, D. A. Determination of protein–DNA binding constants and specificities from statistical analyses of single molecules: MutS–DNA interactions. Nucleic Acids Res. 33, 4322–4334 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tessmer, I. et al. Mechanism of MutS searching for DNA mismatches and signaling repair. J. Biol. Chem. 283, 36646–36654 (2008).

  27. Qiu, R. et al. MutL traps MutS at a DNA mismatch. Proc. Natl Acad. Sci. USA 112, 10914–10919 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. LeBlanc, S. J. et al. Coordinated protein and DNA conformational changes govern mismatch repair initiation by MutS. Nucleic Acids Res. 46, 10782–10795 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Winkler, I. et al. Chemical trapping of the dynamic MutS-MutL complex formed in DNA mismatch repair in Escherichia coli. J. Biol. Chem. 286, 17326–17337 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mendillo, M. L. et al. A conserved MutS homolog connector domain interface interacts with MutL homologs. Proc. Natl Acad. Sci. USA 106, 22223–22228 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Seifert, F. U., Lammens, K., Stoehr, G., Kessler, B. & Hopfner, K.-P. Structural mechanism of ATP-dependent DNA binding and DNA end bridging by eukaryotic Rad50. EMBO J. 35, 759–772 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Liu, Y. et al. ATP-dependent DNA binding, unwinding, and resection by the Mre11/Rad50 complex. EMBO J. 35, 743–758 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Käshammer, L. et al. Mechanism of DNA end sensing and processing by the Mre11-Rad50 complex. Mol. Cell 76, 382–394.e6 (2019).

    Article  PubMed  CAS  Google Scholar 

  34. Robertson, A., Pattishall, S. R. & Matson, S. W. The DNA binding activity of MutL is required for methyl-directed mismatch repair in Escherichia coli. J. Biol. Chem. 281, 8399–8408 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Bende, S. M. & Grafström, R. H. The DNA binding properties of the MutL protein isolated from Escherichia coli. Nucleic Acids Res. 19, 1549–1555 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ban, C., Junop, M. & Yang, W. Transformation of MutL by ATP binding and hydrolysis: a switch in DNA mismatch repair. Cell 97, 85–97 (1999).

    Article  CAS  PubMed  Google Scholar 

  37. Hermans, N. et al. Dual daughter strand incision is processive and increases the efficiency of DNA mismatch repair. Nucleic Acids Res. 44, 6770–6786 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bhairosing-Kok, D. et al. Sharp kinking of a coiled-coil in MutS allows DNA binding and release. Nucleic Acids Res. 74, 681 (2019).

    Google Scholar 

  39. Yamamoto, A., Schofield, M. J., Biswas, I. & Hsieh, P. Requirement for Phe36 for DNA binding and mismatch repair by Escherichia coli MutS protein. Nucleic Acids Res. 28, 3564–3569 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Groothuizen, F. S. et al. Using stable MutS dimers and tetramers to quantitatively analyze DNA mismatch recognition and sliding clamp formation. Nucleic Acids Res. 41, 8166–8181 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Howarth, M. et al. A monovalent streptavidin with a single femtomolar biotin binding site. Nat. Methods 3, 267–273 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Feng, G. & Winkler, M. E. Single-step purifications of His6-MutH, His6-MutL and His6-MutS repair proteins of Escherichia coli K-12. Biotechniques 19, 956–965 (1995).

    CAS  PubMed  Google Scholar 

  43. Monakhova, M. et al. Chromatographic isolation of the functionally active MutS protein covalently linked to deoxyribonucleic acid. J. Chromatogr. A 1389, 19–27 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  45. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. Elife 7, e42166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Zhang, K. Gctf: Real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tegunov, D. & Cramer, P. Real-time cryo-electron microscopy data preprocessing with Warp. Nat. Methods 16, 1146–1152 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Murshudov, G. N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D Biol. Crystallogr. 67, 355–367 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Nicholls, R. A., Tykac, M., Kovalevskiy, O. & Murshudov, G. N. Current approaches for the fitting and refinement of atomic models into cryo-EM maps using CCP-EM. Acta Crystallogr D Struct. Biol. 74, 492–505 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Liebschner, D. et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr D Struct. Biol. 75, 861–877 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Jakobi, A. J., Wilmanns, M. & Sachse, C. Model-based local density sharpening of cryo-EM maps. Elife 6, e27131 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

All cryo-EM data were collected at the LMB cryo-EM facility. We thank C. Savva for cryo-EM support; and J. Grimmett, T. Darling, J. L. Fernández and P. Reviriego for IT support. This work has been supported by a UK Medical Research Council grant U105197143 and LUMC Research Fellowship to M.H.L.; the Oncode Institute, NWO-TOP 714.016.002 and NWO-Gravity CGC.nl to T.K.S.; a Spanish Ministry of Economy and Competitiveness Grant BFU2017-87316 to R.F.L.; NWO-Gravity CGC.nl and the Oncode Institute to J.H.G.L.; and European Community’s Horizon2020 Marie Skłodowska Curie grant [722433] to P.F.

Author information

Authors and Affiliations

Authors

Contributions

R.F.-L., T.K.S. and M.H.L. conceived the overall experimental design; R.F.L. prepared samples and collected and processed cryo-EM data; D.B.-K. purified proteins and performed SPR experiments. A.F. helped with SPR data analysis. V.K. performed crosslinking experiments and MutH nicking assays on crosslinked MutS; P.F. designed crosslinking experiments; J.H.G.L and C.L. performed MutH nicking assays on MutS heterodimers; F.G. and H.H.W. prepared the MutS−MutLLN40 crosslinked complex; R.F.-L., T.K.S and M.H.L. wrote the manuscript, with contributions from all authors.

Corresponding authors

Correspondence to Rafael Fernandez-Leiro, Titia K. Sixma or Meindert H. Lamers.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Structural & Molecular Biology thanks Jean-Baptiste Charbonnier, Elizabeth Kellogg and Cynthia McMurray for their contribution to the peer review of this work. Peer reviewer reports are available. Beth Moorefield was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 CryoEM data analysis of scanning state MutS.

a, Representative micrograph (0° tilt). Plasmid DNA can be observed on the micrograph with MutS dimers bound like beads-on-string. b, 2D class averages from full dataset. c, Schematic representation of main data processing procedures. See methods section for more details. d, Principle Component Analysis of multibody refinement data. e, Detail of model fit to map. f, Final refinement map colored by local resolution. g, Fourier Shell Correlation between half-maps from final refinement. Green line: unmasked. Blue line: masked. Red line: phase randomised. Grey line: corrected h, Model-vs-map Fourier Shell Correlation. Grey line: model vs full-map. Blue line: model refined against half-map 1, FSC against half-map 1. Red-line: shaked model refined against half-map 1, FSC against half-map 2.

Extended Data Fig. 2 CryoEM data analysis of mismatch-bound MutS.

a, Representative micrograph. b, 2D class averages from full dataset. c, Schematic representation of main data processing procedures. See methods section for more details. d, Detail of model fit to map. e, Final refinement map colored by local resolution. f, Fourier Shell Correlation between half-maps from final refinement. Green line: unmasked. Blue line: masked. Red line: phase randomized. Grey line: corrected g, Model-vs-map Fourier Shell Correlation. Grey line: model vs full-map. Blue line: model refined against half-map 1, FSC against half-map 1. Red line: shaken model refined against half-map 1, FSC against half-map 2.

Extended Data Fig. 3 CryoEM data analysis of intermediate state MutS.

a, Representative micrograph. b, 2D class averages from full dataset. c, Schematic representation of main data processing procedures. See methods section for more details. d, Detail of model fit to map. e, Final refinement map colored by local resolution. f, Fourier Shell Correlation between half-maps from final refinement. Green line: unmasked. Blue line: masked. Red line: phase randomized. Grey line: corrected g, Model-vs-map Fourier Shell Correlation. Grey line: model vs full-map. Blue line: model refined against half-map 1, FSC against half-map 1. Red line: shaken model refined against half-map 1, FSC against half-map 2.

Extended Data Fig. 4 CryoEM data analysis of MutLLN40-bound MutS.

a, Representative micrograph. b, 2D class averages from full dataset. c, Schematic representation of main data processing procedures. See methods section for more details. d, Detail of model fit to map. e, Final refinement map colored by local resolution. f, Fourier Shell Correlation between half-maps from final refinement. Green line: unmasked. Blue line: masked. Red line: phase randomized. Grey line: corrected g, Model-vs-map Fourier Shell Correlation. Grey line: model vs full-map. Blue line: model refined against half-map 1, FSC against half-map 1. Red-line: shaken model refined against half-map 1, FSC against half-map 2.

Extended Data Fig. 5 Comparison of crystal and cryo-EM structure of mismatch-bound MutS.

a, Front view of the crystal structure of mismatch-bound MutS (1E3M) fitted into in the cryo-EM map of mismatch-bound MutS. b, Side view, with additional stretches of DNA shown in red. c, Close up of the mismatch binding domain of monomer B with former position of the mismatch binding domain shown in yellow and the ~22o domain rotation indicated by a black arrow. Additional visible loops, modeled after the mismatch binding of monomer A, are highlighted in red. d, Same view as in panel c, showing the cryo-EM map for the DNA, mismatch binding domain and connector domain.

Extended Data Fig. 6 Mismtach and MutL binding surfaces in MutS.

a, Mismatch-bound MutS showing the mismatch and MutL binding surfaces. MutS monomer A in green, monomer B in light-blue, mismatch binding in dark blue, MutL Interface 1 in dark red, and MutL Interface 2 in light red. b, Binding of ATP transforms MutS into a sliding clamp that brings MutL-Interface 1 and 2 together, creating the binding site for MutL c, MutS bound to MutLLN40.

Extended Data Fig. 7 Engineered MutS homo- and heterodimers.

Front view (top row) and reverse view (bottom row) of the MutS dimer a, WT MutS showing mismatch and MutL binding surface on both sides of the dimer b, Interface 1 mutant lacks one half of MutL-binding site on either side of the MutS dimer. c, Similarly, Interface 2 mutant also lacks one half of the MutL-binding site on either side of the MutS dimer. d, Mismatch mutant lacks the mismatch binding site in both monomers. e, Mixing of Interface 1 and Interface 2 mutant creates a heterodimer with an intact MutL binding site on one side, but lacks the MutL-binding interface on the reverse side. f, A double Mismatch-Interface 1 mutant mixed with an Interface 2 mutant generates a MutS dimer with an intact MutL binding site on one side and the mismatch binding site on the reverse side. This configuration is similar to the eukaryotic MSH2-MSH6 and MSH2-MSH3 hetero dimers. g, A double Mismatch-Interface 2 mutant mixed with an Interface 1 mutant generates a MutS dimer with both MutL and mismatch binding sites on the same face of the MutS dimer.

Supplementary information

Supplementary Information

Supplementary Table 1.

Reporting Summary

Peer Review Information

Supplementary Video 1

Molecular mechanism of DNA mismatch repair initiation, related to Fig. 1. Front and side views of MutS passing through the first four stages of the repair cascade: DNA scanning, mismatch recognition, intermediate state, and MutL recruitment. The movements show a computational morphing between the four cryo-EM structures described in this work and are labeled in the movie. MutS monomer A is shown in a pale-green color, monomer B in pale blue, DNA in dark gray and MutLLN40 in yellow.

Supplementary Video 2

Mismatch repair licensing at a mismatch, related to Fig. 2. Top and side views of MutS as it transforms from the DNA-scanning state to the mismatch-bound state. The initial part of the movie represents the movement of monomer B relative to monomer A during the scanning state, as derived from the principal component analysis of the multibody refinement. Note that the MutS dimer explores multiple conformations, attempting to distort the DNA, without crossing over the opposite monomer. When a mismatch is present on the DNA, it allows MutS to deform and kink the DNA and the two MutS monomers to cross over in a clockwise manner. Movements show a computational morphing between the different states. MutS monomer A is shown in a pale-green color, monomer B in pale blue, and DNA in gray. The DNA mismatch is highlighted in pink.

Supplementary Video 3

Multiple conformational changes of mismatch and connector domains tracking DNA, related to Fig. 3. Front and side views of MutS as it goes from the mismatch-bound state to the MutLLN40-bound clamp state via the intermediate state. MutS monomer A is shown in a pale green color, monomer B in pale blue, and DNA in dark gray. DNA mismatch is highlighted in pink. The mismatch domain is shown in dark green and the connector domain in light green. The ends of a central helix in the connector domain are colored in red and blue for clarity. Movements show a computational morphing between the different states.

Source data

Source Data Fig. 3

Uncropped gels.

Source Data Fig. 4

Uncropped gels.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data for SPR.

Source Data Fig. 6

Uncropped gels for nicking assays.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fernandez-Leiro, R., Bhairosing-Kok, D., Kunetsky, V. et al. The selection process of licensing a DNA mismatch for repair. Nat Struct Mol Biol 28, 373–381 (2021). https://doi.org/10.1038/s41594-021-00577-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-021-00577-7

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