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:

UvrD facilitates DNA repair by pulling RNA polymerase backwards

Abstract

UvrD helicase is required for nucleotide excision repair, although its role in this process is not well defined. Here we show that Escherichia coli UvrD binds RNA polymerase during transcription elongation and, using its helicase/translocase activity, forces RNA polymerase to slide backward along DNA. By inducing backtracking, UvrD exposes DNA lesions shielded by blocked RNA polymerase, allowing nucleotide excision repair enzymes to gain access to sites of damage. Our results establish UvrD as a bona fide transcription elongation factor that contributes to genomic integrity by resolving conflicts between transcription and DNA repair complexes. Furthermore, we show that the elongation factor NusA cooperates with UvrD in coupling transcription to DNA repair by promoting backtracking and recruiting nucleotide excision repair enzymes to exposed lesions. Because backtracking is a shared feature of all cellular RNA polymerases, we propose that this mechanism enables RNA polymerases to function as global DNA damage scanners in bacteria and eukaryotes.

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: UvrD promotes RNAP backtracking.
Figure 2: RNAP backtracking facilitates NER.
Figure 3: Anti-backtracking factors obstruct UvrD activity in NER.
Figure 4: Mapping UvrD interactions with the elongation complex.
Figure 5: UvrD and NusA cooperate in backtracking-mediated NER.

Similar content being viewed by others

References

  1. Reardon, J. T. & Sancar, A. Nucleotide excision repair. Prog. Nucleic Acid Res. Mol. Biol. 79, 183–235 (2005)

    CAS  PubMed  Google Scholar 

  2. Van Houten, B. & McCullough, A. Nucleotide excision repair in E. coli. Ann. NY Acad. Sci. 726, 236–251 (1994)

    ADS  CAS  PubMed  Google Scholar 

  3. Ganesan, A., Spivak, G. & Hanawalt, P. C. Transcription-coupled DNA repair in prokaryotes. Prog. Mol. Biol. Transl. Sci. 110, 25–40 (2012)

    CAS  PubMed  Google Scholar 

  4. Truglio, J. J., Croteau, D. L., Van Houten, B. & Kisker, C. Prokaryotic nucleotide excision repair: the UvrABC system. Chem. Rev. 106, 233–252 (2006)

    CAS  PubMed  Google Scholar 

  5. Mellon, I. & Hanawalt, P. C. Induction of the Escherichia coli lactose operon selectively increases repair of its transcribed DNA strand. Nature 342, 95–98 (1989)

    ADS  CAS  PubMed  Google Scholar 

  6. Gaillard, H. & Aguilera, A. Transcription coupled repair at the interface between transcription elongation and mRNP biogenesis. Biochim. Biophys. Acta 1829, 141–150 (2013)

    CAS  PubMed  Google Scholar 

  7. Deaconescu, A. M. et al. Structural basis for bacterial transcription-coupled DNA repair. Cell 124, 507–520 (2006)

    CAS  PubMed  Google Scholar 

  8. Park, J. S., Marr, M. T. & Roberts, J. W. E. coli transcription repair coupling factor (Mfd protein) rescues arrested complexes by promoting forward translocation. Cell 109, 757–767 (2002)

    CAS  PubMed  Google Scholar 

  9. Savery, N. Prioritizing the repair of DNA damage that is encountered by RNA polymerase. Transcription 2, 168–172 (2011)

    PubMed  PubMed Central  Google Scholar 

  10. Selby, C. P. & Sancar, A. Molecular mechanism of transcription–repair coupling. Science 260, 53–58 (1993)

    ADS  CAS  PubMed  Google Scholar 

  11. Kumura, K. & Sekiguchi, M. Identification of the uvrD gene product of Escherichia coli as DNA helicase II and its induction by DNA-damaging agents. J. Biol. Chem. 259, 1560–1565 (1984)

    CAS  PubMed  Google Scholar 

  12. Lee, J. Y. & Yang, W. UvrD helicase unwinds DNA one base pair at a time by a two-part power stroke. Cell 127, 1349–1360 (2006)

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Matson, S. W. & George, J. W. DNA helicase II of Escherichia coli. Characterization of the single-stranded DNA-dependent NTPase and helicase activities. J. Biol. Chem. 262, 2066–2076 (1987)

    CAS  PubMed  Google Scholar 

  14. Cohen, S. E. et al. Roles for the transcription elongation factor NusA in both DNA repair and damage tolerance pathways in Escherichia coli. Proc. Natl Acad. Sci. USA 107, 15517–15522 (2010)

    ADS  CAS  PubMed  Google Scholar 

  15. Ishihama, Y. et al. Exponentially modified protein abundance index (emPAI) for estimation of absolute protein amount in proteomics by the number of sequenced peptides per protein. Mol. Cell. Proteomics 4, 1265–1272 (2005)

    CAS  PubMed  Google Scholar 

  16. Nudler, E., Gusarov, I. & Bar-Nahum, G. Methods of walking with the RNA polymerase. Methods Enzymol. 371, 160–169 (2003)

    CAS  PubMed  Google Scholar 

  17. Nudler, E. RNA polymerase backtracking in gene regulation and genome instability. Cell 149, 1438–1445 (2012)

    CAS  PubMed  Google Scholar 

  18. Borukhov, S., Sagitov, V. & Goldfarb, A. Transcript cleavage factors from E. coli. Cell 72, 459–466 (1993)

    CAS  PubMed  Google Scholar 

  19. Nudler, E., Mustaev, A., Lukhtanov, E. & Goldfarb, A. The RNA–DNA hybrid maintains the register of transcription by preventing backtracking of RNA polymerase. Cell 89, 33–41 (1997)

    CAS  PubMed  Google Scholar 

  20. Brosh, R. M., Jr & Matson, S. W. Mutations in motif II of Escherichia coli DNA helicase II render the enzyme nonfunctional in both mismatch repair and excision repair with differential effects on the unwinding reaction. J. Bacteriol. 177, 5612–5621 (1995)

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Proshkin, S., Rahmouni, A. R., Mironov, A. & Nudler, E. Cooperation between translating ribosomes and RNA polymerase in transcription elongation. Science 328, 504–508 (2010)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Donahue, B. A., Yin, S., Taylor, J. S., Reines, D. & Hanawalt, P. C. Transcript cleavage by RNA polymerase II arrested by a cyclobutane pyrimidine dimer in the DNA template. Proc. Natl Acad. Sci. USA 91, 8502–8506 (1994)

    ADS  CAS  PubMed  Google Scholar 

  23. Selby, C. P., Drapkin, R., Reinberg, D. & Sancar, A. RNA polymerase II stalled at a thymine dimer: footprint and effect on excision repair. Nucleic Acids Res. 25, 787–793 (1997)

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Selby, C. P. & Sancar, A. Transcription preferentially inhibits nucleotide excision repair of the template DNA strand in vitro. J. Biol. Chem. 265, 21330–21336 (1990)

    CAS  PubMed  Google Scholar 

  25. Manelyte, L., Kim, Y. I., Smith, A. J., Smith, R. M. & Savery, N. J. Regulation and rate enhancement during transcription-coupled DNA repair. Mol. Cell 40, 714–724 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Batty, D. P. & Wood, R. D. Damage recognition in nucleotide excision repair of DNA. Gene 241, 193–204 (2000)

    CAS  PubMed  Google Scholar 

  27. Doudney, C. O. & Rinaldi, C. N. Chloramphenicol-promoted increase in resistance to UV damage in Escherichia coli B/r WP2 trpE65: development of the capacity for successful repair of otherwise mutagenic or lethal lesions in DNA. Mutat. Res. 143, 29–34 (1985)

    CAS  PubMed  Google Scholar 

  28. Hanawalt, P. C. The U.V. sensitivity of bacteria: its relation to the DNA replication cycle. Photochem. Photobiol. 5, 1–12 (1966)

    CAS  PubMed  Google Scholar 

  29. Tomko, E. J. et al. 5′-Single-stranded/duplex DNA junctions are loading sites for E. coli UvrD translocase. EMBO J. 29, 3826–3839 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Korzheva, N. et al. A structural model of transcription elongation. Science 289, 619–625 (2000)

    ADS  CAS  PubMed  Google Scholar 

  31. Toulokhonov, I., Artsimovitch, I. & Landick, R. Allosteric control of RNA polymerase by a site that contacts nascent RNA hairpins. Science 292, 730–733 (2001)

    ADS  CAS  PubMed  Google Scholar 

  32. Bar-Nahum, G. et al. A ratchet mechanism of transcription elongation and its control. Cell 120, 183–193 (2005)

    CAS  PubMed  Google Scholar 

  33. Ahn, B. A physical interaction of UvrD with nucleotide excision repair protein UvrB. Mol. Cells 10, 592–597 (2000)

    CAS  PubMed  Google Scholar 

  34. Manelyte, L. et al. The unstructured C-terminal extension of UvrD interacts with UvrB, but is dispensable for nucleotide excision repair. DNA Repair 8, 1300–1310 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Tornaletti, S. Transcription arrest at DNA damage sites. Mutat. Res. 577, 131–145 (2005)

    CAS  PubMed  Google Scholar 

  36. Trautinger, B. W., Jaktaji, R. P., Rusakova, E. & Lloyd, R. G. RNA polymerase modulators and DNA repair activities resolve conflicts between DNA replication and transcription. Mol. Cell 19, 247–258 (2005)

    CAS  PubMed  Google Scholar 

  37. Arthur, H. M. & Eastlake, P. B. Transcriptional control of the uvrD gene of Escherichia coli. Gene 25, 309–316 (1983)

    CAS  PubMed  Google Scholar 

  38. Maluf, N. K., Fischer, C. J. & Lohman, T. M. A dimer of Escherichia coli UvrD is the active form of the helicase in vitro. J. Mol. Biol. 325, 913–935 (2003)

    CAS  PubMed  Google Scholar 

  39. Dutta, D., Shatalin, K., Epshtein, V., Gottesman, M. E. & Nudler, E. Linking RNA polymerase backtracking to genome instability in E. coli. Cell 146, 533–543 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Witkin, E. M. Mutation and the repair of radiation damage in bacteria. Radiat. Res. 6, (suppl.). 30–53 (1966)

    Google Scholar 

  41. Schalow, B. J., Courcelle, C. T. & Courcelle, J. Mfd is required for rapid recovery of transcription following UV-induced DNA damage but not oxidative DNA damage in Escherichia coli. J. Bacteriol. 194, 2637–2645 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Brueckner, F. & Cramer, P. DNA photodamage recognition by RNA polymerase II. FEBS Lett. 581, 2757–2760 (2007)

    CAS  PubMed  Google Scholar 

  43. Churchman, L. S. & Weissman, J. S. Nascent transcript sequencing visualizes transcription at nucleotide resolution. Nature 469, 368–373 (2011)

    ADS  CAS  PubMed  Google Scholar 

  44. Compe, E. & Egly, J. M. TFIIH: when transcription met DNA repair. Nature Rev. Mol. Cell Biol. 13, 343–354 (2012)

    CAS  Google Scholar 

  45. Drapkin, R. et al. Dual role of TFIIH in DNA excision repair and in transcription by RNA polymerase II. Nature 368, 769–772 (1994)

    ADS  CAS  PubMed  Google Scholar 

  46. Yang, B. et al. Identification of cross-linked peptides from complex samples. Nature Methods 9, 904–906 (2012)

    CAS  PubMed  Google Scholar 

  47. Murakami, K. S. X-ray crystal structure of Escherichia coli RNA polymerase σ70 holoenzyme. J. Biol. Chem. 288, 9126–9134 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Runyon, G. T., Wong, I. & Lohman, T. M. Overexpression, purification, DNA binding, and dimerization of the Escherichia coli uvrD gene product (helicase II). Biochemistry 32, 602–612 (1993)

    CAS  PubMed  Google Scholar 

  49. Pakotiprapha, D., Samuels, M., Shen, K., Hu, J. H. & Jeruzalmi, D. Structure and mechanism of the UvrA-UvrB DNA damage sensor. Nature Struct. Mol. Biol. 19, 291–298 (2012)

    CAS  Google Scholar 

  50. Li, D. et al. pFind: a novel database-searching software system for automated peptide and protein identification via tandem mass spectrometry. Bioinformatics 21, 3049–3050 (2005)

    CAS  PubMed  Google Scholar 

  51. Lutz, R. & Bujard, H. Independent and tight regulation of transcriptional units in Escherichia coli via the LacR/O, the TetR/O and AraC/I1-I2 regulatory elements. Nucleic Acids Res. 25, 1203–1210 (1997)

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Epshtein, V., Toulme, F., Rahmouni, A. R., Borukhov, S. & Nudler, E. Transcription through the roadblocks: the role of RNA polymerase cooperation. EMBO J. 22, 4719–4727 (2003)

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Pósfai, G. et al. Emergent properties of reduced-genome Escherichia coli. Science 312, 1044–1046 (2006)

    ADS  PubMed  Google Scholar 

  54. Cardinale, C. J. et al. Termination factor Rho and its cofactors NusA and NusG silence foreign DNA in E. coli. Science 320, 935–938 (2008)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Jeruzalmi for materials. This work was supported by the Russian Foundation for Basic Research (A.M.) and the NIH, BGRF, Dynasty foundation and by the Howard Hughes Medical Institute (E.N.).

Author information

Authors and Affiliations

Authors

Contributions

V.E., V.K., K.M., V.S., B.U., S.P. and A.M. conducted the experimental work, discussed the results and commented on the manuscript. E.N. designed the study and wrote the paper.

Corresponding author

Correspondence to Evgeny Nudler.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 UvrD binds RNAP.

a, A sample list of RNAP-bound proteins from exponentially grown E. coli. Abundance is based on emPAI score15. b, UvrD + 6His–RNAP, UvrD, or 6His–RNAP were affinity-purified on nickel beads and electrophoresed alongside pure UvrD, for reference. The asterisk indicates UvrD. c, RNAP–UvrD complex isolated by size-exclusion chromatography. UvrD–RNAP were mixed 3:1, and run over a Superdex 200 10/300GL column. RNAP core (red) and UvrD (green) chromatograms are overlaid for comparison. The inlaid polyacrylamide gel displays fractions taken from each chromatographic peak.

Extended Data Figure 2 UvrD promotes long-range backtracking.

Biotinylated RNAP was used to prepare the startup EC11 immobilizing on beads. It was walked to position 39, followed by incubation with UvrD, washed (to remove UvrD and NTPs) and then treated with GreB for the indicated times. Numbers on the right indicate the size of 5′-labelled RNAs.

Extended Data Figure 3 Thymine dimer in the template strand blocks the elongation complex.

Schematic diagram of the T7A1 promoter template shows the position of CPD (red). Both control (no TT) and CPD-bearing template strands were radiolabelled at their 5′ ends (top band). The sequencing gel also shows the radiolabelled RNA products before and after chase of EC11. The EC was completely halted by CPD (indicated by red TT). On the control template it formed the runoff products.

Extended Data Figure 4 Effect of greAB, mfd and ribosome inactivation on uvrD sensitivity to DNA damaging agents and ultraviolet.

a, Representative efficiencies of colony formation of wild-type (MG1655) and mutant E. coli cells in the presence of the indicated amounts of mitomycin C, 4NQO and cisplatin. Cells were spotted on LB agar plates in serial tenfold dilutions and incubated at 30 °C for 24 h. b, Representative efficiencies of colony formation of wild-type and ΔuvrD cells in the presence of the indicated amounts of mitomycin, 4NQO and chloramphenicol. Cells were spotted on LB agar plates in serial tenfold dilutions and incubated at 30 °C for 24 h. c, Representative efficiencies of colony formation of wild-type (MG1655) and mutant E. coli cells after ultraviolet irradiation. Cells were spotted on LB agar plates in serial tenfold dilutions and incubated at 30 °C for 24 h. d, Representative efficiencies of colony formation of wild-type and mutant E. coli cells after ultraviolet irradiation in the presence of a sublethal dose of chloramphenicol. Cells were spotted on LB agar plates in serial tenfold dilutions and incubated at 30 °C for 24 h.

Extended Data Figure 5 UvrD inactivation suppresses temperature sensitivity of greAB cells.

E. coli strains were streaked on LB agar plates and incubated at the indicated temperatures for 24 h.

Extended Data Figure 6 UvrD–RNAP crosslinks.

a, Three inter-protein crosslinks indicate that UvrD binds near the β flap on RNAP. UvrD (grey, PDB accession code 2IS4) crosslinks to RNAP (PDB accession code 4IGC) at three distinct positions that span the β (pale yellow) and β′ (light blue) subunits of RNAP. The non-template strand (blue, PDB accession code 4G7O) is indicated for reference. Crosslinked lysines are colored magenta and pairs are connected with a black line. b, MS2 spectrum of a representative crosslinked pair (β′ K40)–(UvrD K448). The peptide sequences with crosslinked lysine residues are shown (top right). Observed peptide backbone cleavage is indicated and b- and y-type fragment ions are labelled in the spectrum. The m/z tolerances of fragment ions are presented in the inset below the spectra. Spectra were annotated using pLabel50.

Extended Data Figure 7 GreB inactivation suppresses nusA sensitivity to genotoxic chemicals and ultraviolet.

a, Representative efficiencies of colony formation of wild-type (MG1655) and mutant E. coli cells in the presence of indicated amounts of NFZ and 4NQO. Cells were spotted on LB agar plates in serial tenfold dilutions and incubated at 30 °C for 24 h. b, Data from three independent experiments are presented as the mean ± s.e.m.; **P < 0.01. c, Representative efficiencies of colony formation of MDS42 and mutant E. coli cells in the presence of indicated amounts of mitomycin, 4NQO, NFZ and after ultraviolet irradiation. Cells were spotted on LB agar plates in serial tenfold dilutions and incubated at 30 °C for 24 h.

Extended Data Figure 8 Deletion of mfd partially suppresses uvrD sensitivity to mitomycin C.

a, Representative efficiencies of colony formation of wild-type (MG1655) and mutant E. coli cells in the presence of indicated amounts of mitomycin C. Cells were spotted on LB agar plates in serial tenfold dilutions and incubated at 30 °C for 24 h. b, Data from three independent experiments are presented as the mean ±s.e.m.; **P < 0.01.

Extended Data Figure 9 Transcriptional arrest as a function of UvrD concentration.

a, A representative chase experiment demonstrating multiple transcriptional arrests as a function of UvrD concentration. b, Data from three independent experiments are plotted as the mean ± s.e.m. Arrest efficiency (%) was calculated as a fraction of all arrested complexes in relation to the full-length runoff.

Extended Data Table 1 Escherichia coli strains used in this study

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Epshtein, V., Kamarthapu, V., McGary, K. et al. UvrD facilitates DNA repair by pulling RNA polymerase backwards. Nature 505, 372–377 (2014). https://doi.org/10.1038/nature12928

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12928

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