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.

  • Letter
  • Published:

Pneumolysin binds to the mannose receptor C type 1 (MRC-1) leading to anti-inflammatory responses and enhanced pneumococcal survival

Abstract

Streptococcus pneumoniae (the pneumococcus) is a major cause of mortality and morbidity globally, and the leading cause of death in children under 5 years old. The pneumococcal cytolysin pneumolysin (PLY) is a major virulence determinant known to induce pore-dependent pro-inflammatory responses. These inflammatory responses are driven by PLY–host cell membrane cholesterol interactions, but binding to a host cell receptor has not been previously demonstrated. Here, we discovered a receptor for PLY, whereby pro-inflammatory cytokine responses and Toll-like receptor signalling are inhibited following PLY binding to the mannose receptor C type 1 (MRC-1) in human dendritic cells and mouse alveolar macrophages. The cytokine suppressor SOCS1 is also upregulated. Moreover, PLY–MRC-1 interactions mediate pneumococcal internalization into non-lysosomal compartments and polarize naive T cells into an interferon-γlow, interleukin-4high and FoxP3+ immunoregulatory phenotype. In mice, PLY-expressing pneumococci colocalize with MRC-1 in alveolar macrophages, induce lower pro-inflammatory cytokine responses and reduce neutrophil infiltration compared with a PLY mutant. In vivo, reduced bacterial loads occur in the airways of MRC-1-deficient mice and in mice in which MRC-1 is inhibited using blocking antibodies. In conclusion, we show that pneumococci use PLY–MRC-1 interactions to downregulate inflammation and enhance bacterial survival in the airways. These findings have important implications for future vaccine design.

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

Fig. 1: PLY inhibits cytokine responses and inflammatory signalling in DCs by upregulating SOCS1.
Fig. 2: MRC-1 colocalizes with PLY and intracellular pneumococci in DCs.
Fig. 3: Depletion of MRC-1 abolishes PLY-induced cytokine inhibition and enhances T cell activation.
Fig. 4: MRC-1 mediates PLY-induced suppression of early inflammatory responses in vivo.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  1. Black, R. E. et al. Global, regional, and national causes of child mortality in 2008: a systematic analysis. Lancet 375, 1969–1987 (2010).

    Article  PubMed  Google Scholar 

  2. Berry, A. M., Yother, J., Briles, D. E., Hansman, D. & Paton, J. C. Reduced virulence of a defined pneumolysin-negative mutant of Streptococcus pneumoniae. Infect. Immun. 57, 2037–2042 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Hirst, R. A. et al. Streptococcus pneumoniae deficient in pneumolysin or autolysin has reduced virulence in meningitis. J. Infect. Dis. 197, 744–751 (2008).

    Article  PubMed  Google Scholar 

  4. Zafar, M. A., Wang, Y., Hamaguchi, S. & Weiser, J. N. Host-to-host transmission of Streptococcus pneumoniae is driven by its inflammatory toxin, pneumolysin. Cell Host Microbe 21, 73–83 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Paton, J. C., Rowan-Kelly, B. & Ferrante, A. Activation of human complement by the pneumococcal toxin pneumolysin. Infect. Immun. 43, 1085–1087 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Iliev, A. I., Djannatian, J. R., Nau, R., Mitchell, T. J. & Wouters, F. S. Cholesterol-dependent actin remodeling via RhoA and Rac1 activation by the Streptococcus pneumoniae toxin pneumolysin. Proc. Natl Acad. Sci. USA 104, 2897–2902 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. McNeela, E. A. et al. Pneumolysin activates the NLRP3 inflammasome and promotes proinflammatory cytokines independently of TLR4. PLoS Pathog. 6, e1001191 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Price, K. E. & Camilli, A. Pneumolysin localizes to the cell wall of Streptococcus pneumoniae. J. Bacteriol. 191, 2163–2168 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 3, 23–35 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Kang, P. B. et al. The human macrophage mannose receptor directs Mycobacterium tuberculosis lipoarabinomannan-mediated phagosome biogenesis. J. Exp. Med. 202, 987–999 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Taylor, P. R., Gordon, S. & Martinez-Pomares, L. The mannose receptor: linking homeostasis and immunity through sugar recognition. Trends Immunol. 26, 104–110 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. Zamze, S. et al. Recognition of bacterial capsular polysaccharides and lipopolysaccharides by the macrophage mannose receptor. J. Biol. Chem. 277, 41613–41623 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. McGreal, E. P. et al. The carbohydrate-recognition domain of Dectin-2 is a C-type lectin with specificity for high mannose. Glycobiology 16, 422–430 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Macedo-Ramos, H. et al. Evidence of involvement of the mannose receptor in the internalization of Streptococcus pneumoniae by Schwann cells. BMC Microbiol. 14, 211 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Macedo-Ramos, H. et al. Olfactory ensheathing cells as putative host cells for Streptococcus pneumoniae: evidence of bacterial invasion via mannose receptor-mediated endocytosis. Neurosci. Res. 69, 308–313 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Kadioglu, A., Weiser, J. N., Paton, J. C. & Andrew, P. W. The role of Streptococcus pneumoniae virulence factors in host respiratory colonization and disease. Nat. Rev. Microbiol. 6, 288–301 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Yoshimura, A., Naka, T. & Kubo, M. SOCS proteins, cytokine signalling and immune regulation. Nat. Rev. Immunol. 7, 454–465 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Yasukawa, H., Sasaki, A. & Yoshimura, A. Negative regulation of cytokine signaling pathways. Annu. Rev. Immunol. 18, 143–164 (2000).

    Article  CAS  PubMed  Google Scholar 

  19. Chieppa, M. et al. Cross-linking of the mannose receptor on monocyte-derived dendritic cells activates an anti-inflammatory immunosuppressive program. J. Immunol. 171, 4552–4560 (2003).

    Article  CAS  Google Scholar 

  20. Paton, J. C. et al. Purification and immunogenicity of genetically obtained pneumolysin toxoids and their conjugation to Streptococcus pneumoniae type 19F polysaccharide. Infect. Immun. 59, 2297–2304 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Guilliams, M., Bruhns, P., Saeys, Y., Hammad, H. & Lambrecht, B. N. The function of Fcγ receptors in dendritic cells and macrophages. Nat. Rev. Immunol. 14, 94–108 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Littmann, M. et al. Streptococcus pneumoniae evades human dendritic cell surveillance by pneumolysin expression. EMBO Mol. Med. 1, 211–222 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rajaram, M. V. S. et al. M. tuberculosis-initiated human mannose receptor signaling regulates macrophage recognition and vesicle trafficking by FcRγ-chain, Grb2, and SHP-1.Cell Rep. 21, 126–140 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sakaguchi, S., Miyara, M., Costantino, C. M. & Hafler, D. A. FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 10, 490–500 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Schuette, V. et al. Mannose receptor induces T-cell tolerance via inhibition of CD45 and up-regulation of CTLA-4. Proc. Natl Acad. Sci. USA 113, 10649–10654 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Neill, D. R. et al. Density and duration of pneumococcal carriage is maintained by transforming growth factor β1 and T regulatory cells. Am. J. Respir. Crit. Care Med. 189, 1250–1259 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Dorrington, M. G. et al. MARCO is required for TLR2- and Nod2-mediated responses to Streptococcus pneumoniae and clearance of pneumococcal colonization in the murine nasopharynx. J. Immunol. 190, 250–258 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Shak, J. R. et al. Novel role for the Streptococcus pneumoniae toxin pneumolysin in the assembly of biofilms. mBio 4, e00655-13 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  29. McKenzie, E. J. et al. Mannose receptor expression and function define a new population of murine dendritic cells. J. Immunol. 178, 4975–4983 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Martinez-Pomares, L. et al. Carbohydrate-independent recognition of collagens by the macrophage mannose receptor. Eur. J. Immunol. 36, 1074–1082 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The work in Sweden was supported by grants from the Swedish Research Council, Stockholm County Council, the Swedish Foundation for Strategic Research (SSF), and the Knut and Alice Wallenberg Foundation. The work in Liverpool was supported by funding from the UK Medical Research Council (programme grant number MR/P011284/1), a Sir Henry Dale Fellowship (awarded to D.R.N.) and jointly funded by the Wellcome Trust and the Royal Society (grant number 204457/Z/16/Z), a British Commonwealth Scholarship (awarded to S.K.), a Embassy of the Kingdom of Saudi Arabia Scholarship (awarded to H.M.), and the Institute of Infection & Global Health, University of Liverpool. The authors thank the Science for Life Laboratory Mass Spectrometry Based Proteomics Facility in Uppsala for the liquid chromatography–mass spectrometry analysis.

Author information

Authors and Affiliations

Authors

Contributions

K.S., D.R.N., L.S., A. Kadioglu and B.H.-N. designed the study. K.S., L.S., G.D.L.M., H.M., S.K., E.D., M.Y., D.R.N., A.A., J.N., P.-Å.N., A. Kirby and L.P. performed the experiments. K.S., L.S., D.R.N., A. Kadioglu and B.H.-N. wrote the manuscript, and the other authors contributed to the writing. All authors read and approved the final version of the manuscript.

Corresponding authors

Correspondence to Aras Kadioglu or Birgitta Henriques-Normark.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Figures 1–12, Supplementary Table 1.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Subramanian, K., Neill, D.R., Malak, H.A. et al. Pneumolysin binds to the mannose receptor C type 1 (MRC-1) leading to anti-inflammatory responses and enhanced pneumococcal survival. Nat Microbiol 4, 62–70 (2019). https://doi.org/10.1038/s41564-018-0280-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-018-0280-x

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