Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity (original) (raw)

Nature volume 477, pages 592–595 (2011)Cite this article

Subjects

Abstract

Inflammasomes are a family of cytosolic multiprotein complexes that initiate innate immune responses to pathogenic microbes by activating the caspase 1 protease1,2. Although genetic data support a critical role for inflammasomes in immune defence and inflammatory diseases3, the molecular basis by which individual inflammasomes respond to specific stimuli remains poorly understood. The inflammasome that contains the NLRC4 (NLR family, CARD domain containing 4) protein was previously shown to be activated in response to two distinct bacterial proteins, flagellin4,5 and PrgJ6, a conserved component of pathogen-associated type III secretion systems. However, direct binding between NLRC4 and flagellin or PrgJ has never been demonstrated. A homologue of NLRC4, NAIP5 (NLR family, apoptosis inhibitory protein 5), has been implicated in activation of NLRC4 (refs 7–11), but is widely assumed to have only an auxiliary role1,2, as NAIP5 is often dispensable for NLRC4 activation7,8. However, Naip5 is a member of a small multigene family12, raising the possibility of redundancy and functional specialization among Naip genes. Here we show in mice that different NAIP paralogues determine the specificity of the NLRC4 inflammasome for distinct bacterial ligands. In particular, we found that activation of endogenous NLRC4 by bacterial PrgJ requires NAIP2, a previously uncharacterized member of the NAIP gene family, whereas NAIP5 and NAIP6 activate NLRC4 specifically in response to bacterial flagellin. We dissected the biochemical mechanism underlying the requirement for NAIP proteins by use of a reconstituted NLRC4 inflammasome system. We found that NAIP proteins control ligand-dependent oligomerization of NLRC4 and that the NAIP2–NLRC4 complex physically associates with PrgJ but not flagellin, whereas NAIP5–NLRC4 associates with flagellin but not PrgJ. Our results identify NAIPs as immune sensor proteins and provide biochemical evidence for a simple receptor–ligand model for activation of the NAIP–NLRC4 inflammasomes.

This is a preview of subscription content, access via your institution

Access options

Subscribe to this journal

Receive 51 print issues and online access

$199.00 per year

only $3.90 per issue

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Additional access options:

Similar content being viewed by others

References

  1. Latz, E. The inflammasomes: mechanisms of activation and function. Curr. Opin. Immunol. 22, 28–33 (2010)
    Article CAS Google Scholar
  2. Schroder, K. & Tschopp, J. The inflammasomes. Cell 140, 821–832 (2010)
    Article CAS Google Scholar
  3. Ting, J. P., Kastner, D. L. & Hoffman, H. M. CATERPILLERs, pyrin and hereditary immunological disorders. Nature Rev. Immunol. 6, 183–195 (2006)
    Article CAS Google Scholar
  4. Franchi, L. et al. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1β in salmonella-infected macrophages. Nature Immunol. 7, 576–582 (2006)
    Article CAS Google Scholar
  5. Miao, E. A. et al. Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1β via Ipaf. Nature Immunol. 7, 569–575 (2006)
    Article CAS Google Scholar
  6. Miao, E. A. et al. Innate immune detection of the type III secretion apparatus through the NLRC4 inflammasome. Proc. Natl Acad. Sci. USA 107, 3076–3080 (2010)
    Article ADS CAS Google Scholar
  7. Lightfield, K. L. et al. Critical function for Naip5 in inflammasome activation by a conserved carboxy-terminal domain of flagellin. Nature Immunol. 9, 1171–1178 (2008)
    Article CAS Google Scholar
  8. Lightfield, K. L. et al. Differential requirements for NAIP5 in activation of the NLRC4 (IPAF) inflammasome. Infect. Immun. 79, 1606–1614 (2011)
    Article CAS Google Scholar
  9. Molofsky, A. B. et al. Cytosolic recognition of flagellin by mouse macrophages restricts Legionella pneumophila infection. J. Exp. Med. 203, 1093–1104 (2006)
    Article CAS Google Scholar
  10. Ren, T., Zamboni, D. S., Roy, C. R., Dietrich, W. F. & Vance, R. E. Flagellin-deficient Legionella mutants evade caspase-1- and Naip5-mediated macrophage immunity. PLoS Pathog. 2, e18 (2006)
    Article Google Scholar
  11. Zamboni, D. S. et al. The Birc1e cytosolic pattern-recognition receptor contributes to the detection and control of Legionella pneumophila infection. Nature Immunol. 7, 318–325 (2006)
    Article CAS Google Scholar
  12. Growney, J. D. & Dietrich, W. F. High-resolution genetic and physical map of the Lgn1 interval in C57BL/6J implicates Naip2 or Naip5 in Legionella pneumophila pathogenesis. Genome Res. 10, 1158–1171 (2000)
    Article CAS Google Scholar
  13. Bergsbaken, T., Fink, S. L. & Cookson, B. T. Pyroptosis: host cell death and inflammation. Nature Rev. Microbiol. 7, 99–109 (2009)
    Article CAS Google Scholar
  14. Wright, E. K. et al. Naip5 affects host susceptibility to the intracellular pathogen Legionella pneumophila . Curr. Biol. 13, 27–36 (2003)
    Article CAS Google Scholar
  15. Broz, P., von Moltke, J., Jones, J. W., Vance, R. E. & Monack, D. M. Differential requirement for Caspase-1 autoproteolysis in pathogen-induced cell death and cytokine processing. Cell Host Microbe 8, 471–483 (2010)
    Article CAS Google Scholar
  16. Martinon, F., Burns, K. & Tschopp, J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell 10, 417–426 (2002)
    Article CAS Google Scholar
  17. Schagger, H., Cramer, W. A. & von Jagow, G. Analysis of molecular masses and oligomeric states of protein complexes by blue native electrophoresis and isolation of membrane protein complexes by two-dimensional native electrophoresis. Anal. Biochem. 217, 220–230 (1994)
    Article CAS Google Scholar
  18. Damiano, J. S., Oliveira, V., Welsh, K. & Reed, J. C. Heterotypic interactions among NACHT domains: implications for regulation of innate immune responses. Biochem. J. 381, 213–219 (2004)
    Article CAS Google Scholar
  19. Hornung, V. et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nature Immunol. 9, 847–856 (2008)
    Article CAS Google Scholar
  20. Zhou, R., Tardivel, A., Thorens, B., Choi, I. & Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nature Immunol. 11, 136–140 (2010)
    Article CAS Google Scholar
  21. Zhou, R., Yazdi, A. S., Menu, P. & Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221–225 (2011)
    Article ADS CAS Google Scholar
  22. Chisholm, S. T., Coaker, G., Day, B. & Staskawicz, B. J. Host-microbe interactions: shaping the evolution of the plant immune response. Cell 124, 803–814 (2006)
    Article CAS Google Scholar
  23. Lu, C. et al. Nucleotide binding to CARD12 and its role in CARD12-mediated caspase-1 activation. Biochem. Biophys. Res. Commun. 331, 1114–1119 (2005)
    Article CAS Google Scholar
  24. Romanish, M. T., Lock, W. M., de Lagemaat, L. N., Dunn, C. A. & Mager, D. L. Repeated recruitment of LTR retrotransposons as promoters by the anti-apoptotic locus NAIP during mammalian evolution. PLoS Genet. 3, e10 (2007)
    Article Google Scholar
  25. Diez, E. et al. Birc1e is the gene within the Lgn1 locus associated with resistance to Legionella pneumophila . Nature Genet. 33, 55–60 (2003)
    Article CAS Google Scholar
  26. Krantz, B. A. et al. A phenylalanine clamp catalyzes protein translocation through the anthrax toxin pore. Science 309, 777–781 (2005)
    Article ADS CAS Google Scholar

Download references

Acknowledgements

Work in R.E.V.’s laboratory is supported by Investigator Awards from the Burroughs Wellcome Fund and the Cancer Research Institute and by NIH grants AI075039, AI080749 and AI063302. We thank J. von Moltke, A. Kintzer and B. Krantz for provision of reagents; S. Mariathasan and V. Dixit for the gift of anti-NLRC4 antibodies and _Nlrc4_−/− mice; J. D. Sauer and D. Portnoy for development of Listeria strains to deliver PrgJ and FlaA; and E. Michelle Long and W. Dietrich for pCDNA3-NAIP constructs and anti-NAIP antibodies. We thank A. Roberts for her initial efforts to knock down NAIP2, M. Fontana for validating NAIP knockdowns, and J. von Moltke and members of the Barton and Vance laboratories for discussions.

Author information

Authors and Affiliations

  1. Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, 94720, California, USA
    Eric M. Kofoed & Russell E. Vance

Authors

  1. Eric M. Kofoed
    You can also search for this author inPubMed Google Scholar
  2. Russell E. Vance
    You can also search for this author inPubMed Google Scholar

Contributions

E.M.K. and R.E.V. conceived the experiments and wrote the paper. E.M.K. performed the experiments.

Corresponding author

Correspondence toRussell E. Vance.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

PowerPoint slides

Rights and permissions

About this article

Cite this article

Kofoed, E., Vance, R. Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity.Nature 477, 592–595 (2011). https://doi.org/10.1038/nature10394

Download citation

This article is cited by