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:

Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation

A Corrigendum to this article was published on 01 November 2017

This article has been updated

Abstract

Respiratory viral infections represent the most common cause of allergic asthma exacerbations. Amplification of the type-2 immune response is strongly implicated in asthma exacerbation, but how virus infection boosts type-2 responses is poorly understood. We report a significant correlation between the release of host double-stranded DNA (dsDNA) following rhinovirus infection and the exacerbation of type-2 allergic inflammation in humans. In a mouse model of allergic airway hypersensitivity, we show that rhinovirus infection triggers dsDNA release associated with the formation of neutrophil extracellular traps (NETs), known as NETosis. We further demonstrate that inhibiting NETosis by blocking neutrophil elastase or by degrading NETs with DNase protects mice from type-2 immunopathology. Furthermore, the injection of mouse genomic DNA alone is sufficient to recapitulate many features of rhinovirus-induced type-2 immune responses and asthma pathology. Thus, NETosis and its associated extracellular dsDNA contribute to the pathogenesis and may represent potential therapeutic targets of rhinovirus-induced asthma exacerbations.

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: Host dsDNA is released during human RV infection, is increased in RV-induced asthma exacerbation and is correlated with type-2 cytokine production and exacerbation severity.
Figure 2: RV-induced exacerbation of allergic airway inflammation and type-2 immune responses are associated with host dsDNA release in mice.
Figure 3: DNase treatment prevents RV-induced type-2-mediated exacerbation of allergic airway inflammation.
Figure 4: Host dsDNA is sufficient to exacerbate type-2 immune responses in allergic mice.
Figure 5: DNase treatment inhibits monocyte-derived DC recruitment during RV-induced exacerbation of allergic airway inflammation.
Figure 6: NETs released during RV infection promote type-2-immune-mediated exacerbation of allergic airway inflammation.

Similar content being viewed by others

Change history

  • 12 July 2017

    In the version of this article initially published, Dr. Nathan W Bartlett was inadvertently omitted from the author list and the Contributions section. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Johnston, S.L. The role of viral and atypical bacterial pathogens in asthma pathogenesis. Pediatr. Pulmonol. Suppl. 18, 141–143 (1999).

    Article  CAS  PubMed  Google Scholar 

  2. Johnston, S.L. et al. Community study of role of viral infections in exacerbations of asthma in 9-11 year old children. Br. Med. J. 310, 1225–1229 (1995).

    Article  CAS  Google Scholar 

  3. Busse, W.W., Lemanske, R.F. Jr. & Gern, J.E. Role of viral respiratory infections in asthma and asthma exacerbations. Lancet 376, 826–834 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Green, R.M. et al. Synergism between allergens and viruses and risk of hospital admission with asthma: case-control study. Br. Med. J. 324, 763 (2002).

    Article  Google Scholar 

  5. Murray, C.S. et al. Study of modifiable risk factors for asthma exacerbations: virus infection and allergen exposure increase the risk of asthma hospital admissions in children. Thorax 61, 376–382 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Bartlett, N.W. et al. Mouse models of rhinovirus-induced disease and exacerbation of allergic airway inflammation. Nat. Med. 14, 199–204 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Beale, J. et al. Rhinovirus-induced IL-25 in asthma exacerbation drives type 2 immunity and allergic pulmonary inflammation. Sci. Transl. Med. 6, 256ra134 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Collison, A. et al. The E3 ubiquitin ligase midline 1 promotes allergen and rhinovirus-induced asthma by inhibiting protein phosphatase 2A activity. Nat. Med. 19, 232–237 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Jackson, D.J. et al. IL-33-dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo. Am. J. Respir. Crit. Care Med. 190, 1373–1382 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Gern, J.E., Vrtis, R., Grindle, K.A., Swenson, C. & Busse, W.W. Relationship of upper and lower airway cytokines to outcome of experimental rhinovirus infection. Am. J. Respir. Crit. Care Med. 162, 2226–2231 (2000).

    Article  CAS  PubMed  Google Scholar 

  11. Braciale, T.J., Sun, J. & Kim, T.S. Regulating the adaptive immune response to respiratory virus infection. Nat. Rev. Immunol. 12, 295–305 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Message, S.D. et al. Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production. Proc. Natl. Acad. Sci. USA 105, 13562–13567 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wark, P.A. et al. Neutrophil degranulation and cell lysis is associated with clinical severity in virus-induced asthma. Eur. Respir. J. 19, 68–75 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Zhu, J. et al. Airway inflammation and illness severity in response to experimental rhinovirus infection in asthma. Chest 145, 1219–1229 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Brinkmann, V. et al. Neutrophil extracellular traps kill bacteria. Science 303, 1532–1535 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Papayannopoulos, V., Metzler, K.D., Hakkim, A. & Zychlinsky, A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J. Cell Biol. 191, 677–691 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Schönrich, G. & Raftery, M.J. Neutrophil extracellular traps go viral. Front. Immunol. 7, 366 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Imanishi, T. et al. Nucleic acid sensing by T cells initiates Th2 cell differentiation. Nat. Commun. 5, 3566 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Marichal, T. et al. DNA released from dying host cells mediates aluminum adjuvant activity. Nat. Med. 17, 996–1002 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. McKee, A.S. et al. Host DNA released in response to aluminum adjuvant enhances MHC class II-mediated antigen presentation and prolongs CD4 T-cell interactions with dendritic cells. Proc. Natl. Acad. Sci. USA 110, E1122–E1131 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bonhagen, K. et al. ICOS+ Th cells produce distinct cytokines in different mucosal immune responses. Eur. J. Immunol. 33, 392–401 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Plantinga, M. et al. Conventional and monocyte-derived CD11b+ dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity 38, 322–335 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Janss, T. et al. Interferon response factor-3 promotes the pro-Th2 activity of mouse lung CD11b+ conventional dendritic cells in response to house dust mite allergens. Eur. J. Immunol. 46, 2614–2628 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Mesnil, C. et al. Resident CD11b+Ly6C lung dendritic cells are responsible for allergic airway sensitization to house dust mite in mice. PLoS One 7, e53242 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Goritzka, M. et al. Alveolar macrophage-derived type I interferons orchestrate innate immunity to RSV through recruitment of antiviral monocytes. J. Exp. Med. 212, 699–714 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jenne, C.N. et al. Neutrophils recruited to sites of infection protect from virus challenge by releasing neutrophil extracellular traps. Cell Host Microbe 13, 169–180 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Branzk, N. et al. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Nat. Immunol. 15, 1017–1025 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wong, S.L. et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med. 21, 815–819 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yu, C. et al. Targeted deletion of a high-affinity GATA-binding site in the GATA-1 promoter leads to selective loss of the eosinophil lineage in vivo. J. Exp. Med. 195, 1387–1395 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Caudrillier, A. et al. Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J. Clin. Invest. 122, 2661–2671 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fuchs, T.A. et al. Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 107, 15880–15885 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hakkim, A. et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA 107, 9813–9818 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Noges, L.E., White, J., Cambier, J.C., Kappler, J.W. & Marrack, P. Contamination of DNase preparations confounds analysis of the role of DNA in alum-adjuvanted vaccines. J. Immunol. 197, 1221–1230 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chan, T.K. et al. House dust mite-induced asthma causes oxidative damage and DNA double-strand breaks in the lungs. J. Allergy Clin. Immunol. 138, 84–96 e81 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Chamilos, G. et al. Cytosolic sensing of extracellular self-DNA transported into monocytes by the antimicrobial peptide LL37. Blood 120, 3699–3707 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lande, R. et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra19 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Lande, R. et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 449, 564–569 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Ishii, K.J. & Akira, S. Innate immune recognition of, and regulation by, DNA. Trends Immunol. 27, 525–532 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Funchal, G.A. et al. Respiratory syncytial virus fusion protein promotes TLR-4-dependent neutrophil extracellular trap formation by human neutrophils. PLoS One 10, e0124082 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Cheng, O.Z. & Palaniyar, N. NET balancing: a problem in inflammatory lung diseases. Front. Immunol. 4, 1 (2013).

    PubMed  PubMed Central  Google Scholar 

  41. Garcia-Romo, G.S. et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra20 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Koga, H. et al. Inhibition of neutrophil elastase attenuates airway hyperresponsiveness and inflammation in a mouse model of secondary allergen challenge: neutrophil elastase inhibition attenuates allergic airway responses. Respir. Res. 14, 8 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Suzuki, T. et al. Aerosolized human neutrophil elastase induces airway constriction and hyperresponsiveness with protection by intravenous pretreatment with half-length secretory leukoprotease inhibitor. Am. J. Respir. Crit. Care Med. 153, 1405–1411 (1996).

    Article  CAS  PubMed  Google Scholar 

  44. de Heer, H.J. et al. Essential role of lung plasmacytoid dendritic cells in preventing asthmatic reactions to harmless inhaled antigen. J. Exp. Med. 200, 89–98 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Skrzeczynska-Moncznik, J. et al. DNA structures decorated with cathepsin G/secretory leukocyte proteinase inhibitor stimulate IFNI production by plasmacytoid dendritic cells. Am. J. Clin. Exp. Immunol. 2, 186–194 (2013).

    PubMed  PubMed Central  Google Scholar 

  46. Maffia, P.C. et al. Neutrophil elastase converts human immature dendritic cells into transforming growth factor-beta1-secreting cells and reduces allostimulatory ability. Am. J. Pathol. 171, 928–937 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Roghanian, A., Drost, E.M., MacNee, W., Howie, S.E. & Sallenave, J.M. Inflammatory lung secretions inhibit dendritic cell maturation and function via neutrophil elastase. Am. J. Respir. Crit. Care Med. 174, 1189–1198 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Tateosian, N.L. et al. Neutrophil elastase treated dendritic cells promote the generation of CD4+FOXP3+ regulatory T cells in vitro. Cell. Immunol. 269, 128–134 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Jenne, C.N. & Kubes, P. Virus-induced NETs--critical component of host defense or pathogenic mediator? PLoS Pathog. 11, e1004546 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Yipp, B.G. & Kubes, P. NETosis: how vital is it? Blood 122, 2784–2794 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Hahn, S., Giaglis, S., Chowdhury, C.S., Hösli, I. & Hasler, P. Modulation of neutrophil NETosis: interplay between infectious agents and underlying host physiology. Semin. Immunopathol. 35, 439–453 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chrysanthopoulou, A. et al. Neutrophil extracellular traps promote differentiation and function of fibroblasts. J. Pathol. 233, 294–307 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Grabcanovic-Musija, F. et al. Neutrophil extracellular trap (NET) formation characterises stable and exacerbated COPD and correlates with airflow limitation. Respir. Res. 16, 59 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Jackson, D.J. & Johnston, S.L. The role of viruses in acute exacerbations of asthma. J. Allergy Clin. Immunol. 125, 1178–1187; quiz 1188–1179 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Toussaint, M. et al. Myeloid hypoxia-inducible factor 1α prevents airway allergy in mice through macrophage-mediated immunoregulation. Mucosal Immunol. 6, 485–497 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Marichal, T. et al. Interferon response factor 3 is essential for house dust mite-induced airway allergy. J. Allergy Clin. Immunol. 126, 836–844 e813 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Singanayagam, A. et al. A short-term mouse model that reproduces the immunopathological features of rhinovirus-induced exacerbation of COPD. Clin. Sci. (Lond.) 129, 245–258 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank all the CBS team from Imperial College London for animal management, M. Paulsen of the Flow Cytometry Core Facility from St Mary's Campus (Imperial College London) for giving advice, and C. Tytgat for secretarial assistance. The authors also thank S. Ormenese and J.J. Goval of the Cell Imaging Platform of the Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA, Liège, Belgium) for help with confocal microscopy. M.T. is a postdoctoral fellow who has been supported by the Wallonie-Bruxelles International organization and by the European Academy of Allergy and Clinical Immunology. T.M. is a Research Associate of the F.R.S-FNRS and is supported by the Acteria Foundation. This work was supported by the European Research Council (ERC FP7 grant number 233015) to S.L.J. a Chair from Asthma UK (number CH11SJ) to S.L.J. the Medical Research Council Centre (grant number G1000758); National Institute of Health Research (NIHR) Biomedical Research Centre (grant number P26095); Predicta FP7 Collaborative Project (grant number 260895); and the NIHR Biomedical Research Centre at Imperial College London. S.L.J. is a NIHR Senior Investigator.

Author information

Authors and Affiliations

Authors

Contributions

M.T. made the initial observation by analyzing human samples, performed most of the experiments and analyzed the data. M.T. designed the experiments with the help of M.R.E., R.S., T.M., F.B., N.W.B. and S.L.J. D.J.J. conducted the human experimental infection study, provided associated data and designed Supplementary Table 1. S.L.J. was the principal investigator for the human experimental-infection study. M.T., D.S. and A.G. performed the western blot experiments. D.S., Y.M.C., N.W.B. and H.M. helped M.T. to process mouse samples in the laboratory. C.R. participated in confocal studies and experiments involving ΔdblGATA BALB/c mice. J.A. grew and purified virus with assistance from N.W.B. F.F. performed human and part of mouse statistical analyses. T.-D.T. and V.P. contributed to experiments and discussion related to NET detection and inhibition. M.T. and T.M. designed the figures. All authors provided feedback on the manuscript; M.T., T.M., F.B. and S.L.J. wrote the manuscript. T.M., F.B. and S.L.J. supervised the project; F.B. and S.L.J. secured funding.

Corresponding authors

Correspondence to Fabrice Bureau, Thomas Marichal or Sebastian L Johnston.

Ethics declarations

Competing interests

D.J.J. has received support for travel expenses to attend Respiratory Conferences from AstraZeneca, Boehringer Ingelheim (UK) and GSK. S.L.J. reports grants and/or personal fees from Centocor; Sanofi Pasteur; GSK; Chiesi; Boehringer Ingelheim; Novartis; grants, personal fees and shareholding from Synairgen; personal fees from Bioforce outside the submitted work; In addition, S.L.J. is involved in patents relating to use of interferon-β and interferon-λ for the treatment and prevention of virally induced exacerbation in asthma and chronic pulmonary obstructive disease, and for the induction of cross-reactive cellular responses against rhinovirus antigens.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 and Supplementary Table 1 (PDF 16399 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Toussaint, M., Jackson, D., Swieboda, D. et al. Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation. Nat Med 23, 681–691 (2017). https://doi.org/10.1038/nm.4332

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4332

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