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:

DNA mismatch repair is required for the host innate response and controls cellular fate after influenza virus infection

Abstract

Despite the cytopathic nature of influenza A virus (IAV) replication, we recently reported that a subset of lung epithelial club cells is able to intrinsically clear the virus and survive infection. However, the mechanisms that drive cell survival during a normally lytic infection remained unclear. Using a loss-of-function screening approach, we discovered that the DNA mismatch repair (MMR) pathway is essential for club cell survival of IAV infection. Repair of virally induced oxidative damage by the DNA MMR pathway not only allowed cell survival of infection, but also facilitated host gene transcription, including the expression of antiviral and stress response genes. Enhanced viral suppression of the DNA MMR pathway prevented club cell survival and increased the severity of viral disease in vivo. Altogether, these results identify previously unappreciated roles for DNA MMR as a central modulator of cellular fate and a contributor to the innate antiviral response, which together control influenza viral disease severity.

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: NCI-H441 cells survive direct infection with influenza A virus.
Fig. 2: An siRNA screen of the druggable genome reveals host factors that control H441 cell survival during IAV infection.
Fig. 3: DNA MMR genes are important for survival and their expression is maintained in H441 cells during infection.
Fig. 4: DNA MMR activity remains high in H441 cells allowing repair of virally induced ROS-mediated DNA damage.
Fig. 5: Loss of DNA MMR activity reduces the innate antiviral transcriptional response against influenza A virus.
Fig. 6: DNA MMR is required for cellular survival and protection from virulence in mice.

Similar content being viewed by others

Data availability

The raw RNA-seq data files from Fig. 5i–k are available at NCBI GEO (series GSE130189). The raw data for Figs. 2b–d, 3d and 5i–k are available in Supplementary Tables 15. Raw data from all other figures and unique materials, including viruses and plasmids, are available from the corresponding authors upon request.

References

  1. Downey, J., Pernet, E., Coulombe, F. & Divangahi, M. Dissecting host cell death programs in the pathogenesis of influenza. Microbes Infect. 20, 560–569 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Turpin, E. et al. Influenza virus infection increases p53 activity: role of p53 in cell death and viral replication. J. Virol. 79, 8802–8811 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Orzalli, M. H. & Kagan, J. C. Apoptosis and necroptosis as host defense strategies to prevent viral infection. Trends Cell Biol. 27, 800–809 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ehrhardt, C. et al. Influenza A virus NS1 protein activates the PI3K/Akt pathway to mediate antiapoptotic signaling responses. J. Virol. 81, 3058–3067 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zhirnov, O. P. & Klenk, H. D. Control of apoptosis in influenza virus-infected cells by up-regulation of Akt and p53 signaling. Apoptosis 12, 1419–1432 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. van de Sandt, C. E., Kreijtz, J. H. & Rimmelzwaan, G. F. Evasion of influenza A viruses from innate and adaptive immune responses. Viruses 4, 1438–1476 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Heaton, N. S. et al. Long-term survival of influenza virus infected club cells drives immunopathology. J. Exp. Med. 211, 1707–1714 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Reuther, P. et al. Generation of a variety of stable influenza A reporter viruses by genetic engineering of the NS gene segment. Sci. Rep. 5, 11346 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Burdeinick-Kerr, R. & Griffin, D. E. Gamma interferon-dependent, noncytolytic clearance of sindbis virus infection from neurons in vitro. J. Virol. 79, 5374–5385 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Guidotti, L. G. et al. Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity 4, 25–36 (1996).

    Article  CAS  PubMed  Google Scholar 

  11. Guidotti, L. G. et al. Noncytopathic Clearance of Lymphocytic Choriomeningitis Virus from the Hepatocyte. J. Exp. Med. 189, 1555–1564 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chisari, F. V. Viruses, immunity, and cancer: lessons from hepatitis B. Am. J. Pathol. 156, 1117–1132 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Griffin, D. E. Recovery from viral encephalomyelitis: immune-mediated noncytolytic virus clearance from neurons. Immunol. Res. 47, 123–133 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kudchodkar, S. B. & Levine, B. Viruses and autophagy. Rev. Med Virol. 19, 359–378 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hamilton, J. R. et al. Club cells surviving influenza A virus infection induce temporary nonspecific antiviral immunity. Proc. Natl Acad. Sci. USA 113, 3861–3866 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Bridge, G., Rashid, S. & Martin, S. A. DNA mismatch repair and oxidative DNA damage: implications for cancer biology and treatment. Cancers (Basel) 6, 1597–1614 (2014).

    Article  CAS  Google Scholar 

  17. Lei, X., Zhu, Y., Tomkinson, A. & Sun, L. Measurement of DNA mismatch repair activity in live cells. Nucleic Acids Res. 32, e100 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Macpherson, P. et al. 8-Oxoguanine incorporation into DNA repeats in vitro and mismatch recognition by MutSalpha. Nucleic Acids Res. 33, 5094–5105 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nencioni, L. et al. Influenza A virus replication is dependent on an antioxidant pathway that involves GSH and Bcl-2. FASEB J. 17, 758–760 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Sgarbanti, R. et al. Redox regulation of the influenza hemagglutinin maturation process: a new cell-mediated strategy for anti-influenza therapy. Antioxid. Redox Signal. 15, 593–606 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Amatore, D. et al. Influenza virus replication in lung epithelial cells depends on redox-sensitive pathways activated by NOX4-derived ROS. Cell Microbiol. 17, 131–145 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Li, N. et al. Influenza infection induces host DNA damage and dynamic DNA damage responses during tissue regeneration. Cell Mol. Life Sci. 72, 2973–2988 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lin, X. et al. The influenza virus H5N1 infection can induce ROS production for viral replication and host cell death in A549 cells modulated by human Cu/Zn superoxide dismutase (sod1) overexpression. Viruses 8, E13 (2016).

    Article  PubMed  CAS  Google Scholar 

  24. Russell, A. B., Trapnell, C. & Bloom, J. D. Extreme heterogeneity of influenza virus infection in single cells. eLife 7, e32303 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Shin, N., Pyo, C. W., Jung, K. I. & Choi, S. Y. Influenza A virus PB1-F2 is involved in regulation of cellular redox state in alveolar epithelial cells. Biochem. Biophys. Res. Commun. 459, 699–705 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Wang, Q. W. et al. Anti-influenza A virus activity of rhein through regulating oxidative stress, TLR4, Akt, MAPK, and NF-kappaB signal pathways. PLoS ONE 13, e0191793 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Valavanidis, A., Vlachogianni, T. & Fiotakis, C. 8-Hydroxy-2’-deoxyguanosine (8-OHdG): a critical biomarker of oxidative stress and carcinogenesis. J. Environ. Sci. Health C 27, 120–139 (2009).

    Article  CAS  Google Scholar 

  28. Singh, N. P., McCoy, M. T., Tice, R. R. & Schneider, E. L. A simple technique for quantitation of low levels of DNA damage in individual cells. Exp. Cell Res. 175, 184–191 (1988).

    Article  CAS  PubMed  Google Scholar 

  29. Collins, A. R., Dusinska, M. & Horska, A. Detection of alkylation damage in human lymphocyte DNA with the comet assay. Acta Biochim. Pol. 48, 611–614 (2001).

    CAS  PubMed  Google Scholar 

  30. Hamad, I., Arda, N., Pekmez, M., Karaer, S. & Temizkan, G. Intracellular scavenging activity of Trolox (6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid) in the fission yeast, Schizosaccharomyces pombe. J. Nat. Sci. Biol. Med. 1, 16–21 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Kuraoka, I. et al. Effects of endogenous DNA base lesions on transcription elongation by mammalian RNA polymerase II. Implications for transcription-coupled DNA repair and transcriptional mutagenesis. J. Biol. Chem. 278, 7294–7299 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Charlet-Berguerand, N. et al. RNA polymerase II bypass of oxidative DNA damage is regulated by transcription elongation factors. EMBO J. 25, 5481–5491 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mellon, I. & Champe, G. N. Products of DNA mismatch repair genes mutS and mutL are required for transcription-coupled nucleotide-excision repair of the lactose operon in Escherichia coli. Proc. Natl Acad. Sci. USA 93, 1292–1297 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Mellon, I., Rajpal, D. K., Koi, M., Boland, C. R. & Champe, G. N. Transcription-coupled repair deficiency and mutations in human mismatch repair genes. Science 272, 557–560 (1996).

    Article  CAS  PubMed  Google Scholar 

  35. Ni, T. T., Marsischky, G. T. & Kolodner, R. D. MSH2 and MSH6 are required for removal of adenine misincorporated opposite 8-oxo-guanine in S. cerevisiae. Mol. Cell 4, 439–444 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Bercovich-Kinori, A. et al. A systematic view on influenza induced host shutoff. eLife 5, e18311 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Domingues, P. et al. Global reprogramming of host SUMOylation during influenza virus Infection. Cell Rep. 13, 1467–1480 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Noh, H., Shoemaker, J. E. & Gunawan, R. Network perturbation analysis of gene transcriptional profiles reveals protein targets and mechanism of action of drugs and influenza A viral infection. Nucleic Acids Res. 46, e34 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Varble, A. et al. An in vivo RNAi screening approach to identify host determinants of virus replication. Cell Host Microbe 14, 346–356 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Khanna, M. et al. Detection of influenza virus induced ultrastructural changes and DNA damage. Indian J. Virol. 21, 50–55 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Vijaya Lakshmi, A. N., Ramana, M. V., Vijayashree, B., Ahuja, Y. R. & Sharma, G. Detection of influenza virus induced DNA damage by Comet assay. Mutat. Res. 442, 53–58 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Ryan, E. L., Hollingworth, R. & Grand, R. J. Activation of the DNA damage response by RNA viruses. Biomolecules 6, 2 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Machida, K. et al. Hepatitis C virus infection activates the immunologic (type II) isoform of nitric oxide synthase and thereby enhances DNA damage and mutations of cellular genes. J. Virol. 78, 8835–8843 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Machida, K. et al. Hepatitis C virus inhibits DNA damage repair through reactive oxygen and nitrogen species and by interfering with the ATM-NBS1/Mre11/Rad50 DNA repair pathway in monocytes and hepatocytes. J. Immunol. 185, 6985–6998 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Clavarino, G. et al. Induction of GADD34 is necessary for dsRNA-dependent interferon-beta production and participates in the control of Chikungunya virus infection. PLoS Pathog. 8, e1002708 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Nargi-Aizenman, J. L., Simbulan-Rosenthal, C. M., Kelly, T. A., Smulson, M. E. & Griffin, D. E. Rapid activation of poly(ADP-ribose) polymerase contributes to Sindbis virus and staurosporine-induced apoptotic cell death. Virology 293, 164–171 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Datta, A. & Jinks-Robertson, S. Association of increased spontaneous mutation rates with high levels of transcription in yeast. Science 268, 1616–1619 (1995).

    Article  CAS  PubMed  Google Scholar 

  48. Morey, N. J., Greene, C. N. & Jinks-Robertson, S. Genetic analysis of transcription-associated mutation in Saccharomyces cerevisiae. Genetics 154, 109–120 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Naganuma, A., Dansako, H., Nakamura, T., Nozaki, A. & Kato, N. Promotion of microsatellite instability by hepatitis C virus core protein in human non-neoplastic hepatocyte cells. Cancer Res. 64, 1307–1314 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Olejnik, J. et al. Ebolaviruses associated with differential pathogenicity induce distinct host responses in human macrophages. J. Virol. 91, e00179–17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Xue, J. et al. Dynamic interactions between Bombyx mori nucleopolyhedrovirus and its host cells revealed by transcriptome analysis. J. Virol. 86, 7345–7359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Beard, P. M. et al. A loss of function analysis of host factors influencing Vaccinia virus replication by RNA interference. PLoS ONE 9, e98431 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Maddocks, O. D., Scanlon, K. M. & Donnenberg, M. S. An Escherichia coli effector protein promotes host mutation via depletion of DNA mismatch repair proteins. mBio 4, e00152–00113 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sauvonnet, N., Pradet-Balade, B., Garcia-Sanz, J. A. & Cornelis, G. R. Regulation of mRNA expression in macrophages after Yersinia enterocolitica infection. Role of different Yop effectors. J. Biol. Chem. 277, 25133–25142 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Kim, J. J. et al. Helicobacter pylori impairs DNA mismatch repair in gastric epithelial cells. Gastroenterology 123, 542–553 (2002).

    Article  CAS  PubMed  Google Scholar 

  56. Iyer, R. R., Pluciennik, A., Burdett, V. & Modrich, P. L. DNA mismatch repair: functions and mechanisms. Chem. Rev. 106, 302–323 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Marteijn, J. A., Lans, H., Vermeulen, W. & Hoeijmakers, J. H. Understanding nucleotide excision repair and its roles in cancer and ageing. Nat. Rev. Mol. Cell Biol. 15, 465–481 (2014).

    Article  CAS  Google Scholar 

  58. Robertson, A. B., Klungland, A., Rognes, T. & Leiros, I. DNA repair in mammalian cells: base excision repair: the long and short of it. Cell Mol. Life Sci. 66, 981–993 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Jagger, B. W. et al. An overlapping protein-coding region in influenza A virus segment 3 modulates the host response. Science 337, 199–204 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Nemeroff, M. E., Barabino, S. M., Li, Y., Keller, W. & Krug, R. M. Influenza virus NS1 protein interacts with the cellular 30 kDa subunit of CPSF and inhibits 3’ end formation of cellular pre-mRNAs. Mol. Cell 1, 991–1000 (1998).

    Article  CAS  PubMed  Google Scholar 

  61. Vreede, F. T., Chan, A. Y., Sharps, J. & Fodor, E. Mechanisms and functional implications of the degradation of host RNA polymerase II in influenza virus infected cells. Virology 396, 125–134 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Quinlivan, M. et al. Attenuation of equine influenza viruses through truncations of the NS1 protein. J. Virol. 79, 8431–8439 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Heaton, N. S. et al. In vivo bioluminescent imaging of influenza A virus infection and characterization of novel cross-protective monoclonal antibodies. J. Virol. 87, 8272–8281 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Heaton, B. E. et al. A CRISPR activation screen identifies a pan-avian influenza virus inhibitory host factor. Cell Rep. 20, 1503–1512 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank H. Bogerd and B. Cullen (Duke University) for their help with the amiRNA northern blots. We would like to thank P. Palese (Mt. Sinai) for support and reagents during preliminary optimization experiments. We would also like to thank B. tenOever (Mt. Sinai) for his help in designing the amiRNA-expressing viruses. We are also grateful for contributions made by H. Froggatt (Duke University) in researching the literature on other pathogens that downregulate DNA MMR. The RNA-seq mapping pipeline was developed by David Sachs. N.S.H. is partially supported by NIH K22-AI116509-01, R21-AI133444-01, R01-HL142985, R01-AI137031 and the Duke School of Medicine Whitehead Scholarship. B.S.C. is supported by NIH training grant T32-CA009111. R.E.D. is supported by NIH training grant T32-GM007184-41. S.C. is supported by NIH grants R01AI074951, R01AI140539 and R01AI122749, and is a recipient of the Burroughs Wellcome Investigators in the Pathogenesis of Infectious Disease Award.

Author information

Authors and Affiliations

Authors

Contributions

B.S.C., S.C. and N.S.H. designed the study and experiments. B.S.C. generated many of the reagents and performed and analysed the majority of the biochemical and mouse experiments. B.E.H. performed some of the interferon gene expression experiments. K.R. and S.C. performed and analysed the siRNA screen data. R.E.D. performed and analysed the phospho-H2AX and ALI culture experiments. J.R.H. and N.S.H. generated the Cre-reporter assays and optimized screening conditions. N.S.H. performed and analysed the 8-OHdG experiment, most of the screen validation experiments and most of the experiments characterizing H441 cells as models for cell survival. B.S.C., S.C. and N.S.H. wrote the manuscript.

Corresponding authors

Correspondence to Sara Cherry or Nicholas S. Heaton.

Ethics declarations

Competing interests

Duke University has filed a provisional patent for targeting DNA MMR as a method to enhance the growth of influenza vaccine strains.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–7.

Reporting Summary

Supplementary Table 1

Average Z-scores of all genes tested in both replicates of the primary siRNA screen, related to Fig. 2b,c.

Supplementary Table 2

Validation siRNA sequence information and results of statistical analysis of secondary siRNA screen, related to Figs. 2d and 2g.

Supplementary Table 3

Relative mRNA levels of DNA MMR genes at 9 h postinfection with WT PR8 in A549 and H441 cells compared to mock controls, related to Fig. 3d.

Supplementary Table 4

Raw read counts for all genes detected in RNA-seq of WT PR8-infected H441 cells with control or DNA MMR knockdown, related to Fig. 5i–k and Supplementary Fig. 4.

Supplementary Table 5

RNA-seq data and analysis for all genes induced >5-fold in WT PR8-infected H441 cells, related to Fig. 5i–k.

Supplementary Table 6

List of primers used for RT–qPCR analyses, related to Figs. 3d, 5c and 5l–o.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chambers, B.S., Heaton, B.E., Rausch, K. et al. DNA mismatch repair is required for the host innate response and controls cellular fate after influenza virus infection. Nat Microbiol 4, 1964–1977 (2019). https://doi.org/10.1038/s41564-019-0509-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0509-3

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