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:

Manipulation of small Rho GTPases is a pathogen-induced process detected by NOD1

Abstract

Our innate immune system distinguishes microbes from self by detecting conserved pathogen-associated molecular patterns1. However, these are produced by all microbes, regardless of their pathogenic potential. To distinguish virulent microbes from those with lower disease-causing potential the innate immune system detects conserved pathogen-induced processes2, such as the presence of microbial products in the host cytosol, by mechanisms that are not fully resolved. Here we show that NOD1 senses cytosolic microbial products by monitoring the activation state of small Rho GTPases. Activation of RAC1 and CDC42 by bacterial delivery or ectopic expression of SopE, a virulence factor of the enteric pathogen Salmonella, triggered the NOD1 signalling pathway, with consequent RIP2 (also known as RIPK2)-mediated induction of NF-κB-dependent inflammatory responses. Similarly, activation of the NOD1 signalling pathway by peptidoglycan required RAC1 activity. Furthermore, constitutively active forms of RAC1, CDC42 and RHOA activated the NOD1 signalling pathway. Our data identify the activation of small Rho GTPases as a pathogen-induced process sensed through the NOD1 signalling pathway.

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

Figure 1: SopE-induced NF-κB activation requires RIP2 and NOD1.
Figure 2: NOD1 senses the activation state of small Rho GTPases.
Figure 3: SopE forms a multiprotein complex with RAC1, CDC42, NOD1 and HSP90.

Similar content being viewed by others

References

  1. Janeway, C. A., Jr Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb. Symp. Quant. Biol. 54, 1–13 (1989)

    Article  CAS  Google Scholar 

  2. Vance, R. E., Isberg, R. R. & Portnoy, D. A. Patterns of pathogenesis: discrimination of pathogenic and nonpathogenic microbes by the innate immune system. Cell Host Microbe 6, 10–21 (2009)

    Article  CAS  Google Scholar 

  3. Fu, Y. & Galan, J. E. The Salmonella typhimurium tyrosine phosphatase SptP is translocated into host cells and disrupts the actin cytoskeleton. Mol. Microbiol. 27, 359–368 (1998)

    Article  CAS  Google Scholar 

  4. Zhou, D., Mooseker, M. S. & Galan, J. E. An invasion-associated Salmonella protein modulates the actin-bundling activity of plastin. Proc. Natl Acad. Sci. USA 96, 10176–10181 (1999)

    Article  ADS  CAS  Google Scholar 

  5. Hardt, W. D., Chen, L. M., Schuebel, K. E., Bustelo, X. R. & Galan, J. E. S. typhimurium encodes an activator of Rho GTPases that induces membrane ruffling and nuclear responses in host cells. Cell 93, 815–826 (1998)

    Article  CAS  Google Scholar 

  6. Hapfelmeier, S. et al. Role of the Salmonella pathogenicity island 1 effector proteins SipA, SopB, SopE, and SopE2 in Salmonella enterica subspecies 1 serovar Typhimurium colitis in streptomycin-pretreated mice. Infect. Immun. 72, 795–809 (2004)

    Article  CAS  Google Scholar 

  7. Bruno, V. M. et al. Salmonella Typhimurium type III secretion effectors stimulate innate immune responses in cultured epithelial cells. PLoS Pathog. 5, e1000538 (2009)

    Article  Google Scholar 

  8. Figueiredo, J. F. et al. Salmonella enterica Typhimurium SipA induces CXC-chemokine expression through p38MAPK and JUN pathways. Microbes Infect. 11, 302–310 (2009)

    Article  CAS  Google Scholar 

  9. Hobbie, S., Chen, L. M., Davis, R. J. & Galan, J. E. Involvement of mitogen-activated protein kinase pathways in the nuclear responses and cytokine production induced by Salmonella typhimurium in cultured intestinal epithelial cells. J. Immunol. 159, 5550–5559 (1997)

    CAS  PubMed  Google Scholar 

  10. Smith, M. F., Jr et al. Toll-like receptor (TLR) 2 and TLR5, but not TLR4, are required for Helicobacter pylori-induced NF-κB activation and chemokine expression by epithelial cells. J. Biol. Chem. 278, 32552–32560 (2003)

    Article  CAS  Google Scholar 

  11. Lu, W. et al. Cutting edge: enhanced pulmonary clearance of Pseudomonas aeruginosa by Muc1 knockout mice. J. Immunol. 176, 3890–3894 (2006)

    Article  CAS  Google Scholar 

  12. Keestra, A. M. et al. A Salmonella virulence factor activates the NOD1/NOD2 signaling pathway. mBio 2, e00266–00211 (2011)

    CAS  PubMed  Google Scholar 

  13. Coso, O. A. et al. The small GTP-binding proteins Rac1 and Cdc42 regulate the activity of the JNK/SAPK signaling pathway. Cell 81, 1137–1146 (1995)

    Article  CAS  Google Scholar 

  14. Schlumberger, M. C. et al. Amino acids of the bacterial toxin SopE involved in G nucleotide exchange on Cdc42. J. Biol. Chem. 278, 27149–27159 (2003)

    Article  CAS  Google Scholar 

  15. Müller, A. J., Hoffmann, C. & Hardt, W. D. Caspase-1 activation via Rho GTPases: a common theme in mucosal infections? PLoS Pathog. 6, e1000795 (2010)

    Article  Google Scholar 

  16. Hapfelmeier, S. et al. The Salmonella pathogenicity island (SPI)-2 and SPI-1 type III secretion systems allow Salmonella serovar typhimurium to trigger colitis via MyD88-dependent and MyD88-independent mechanisms. J. Immunol. 174, 1675–1685 (2005)

    Article  CAS  Google Scholar 

  17. Le Bourhis, L. et al. Role of Nod1 in mucosal dendritic cells during Salmonella pathogenicity island 1-independent Salmonella enterica serovar Typhimurium infection. Infect. Immun. 77, 4480–4486 (2009)

    Article  CAS  Google Scholar 

  18. Geddes, K. et al. Nod1 and Nod2 regulation of inflammation in the Salmonella colitis model. Infect. Immun. 78, 5107–5115 (2010)

    Article  CAS  Google Scholar 

  19. Inohara, N. et al. An induced proximity model for NF-κB activation in the Nod1/RICK and RIP signaling pathways. J. Biol. Chem. 275, 27823–27831 (2000)

    CAS  PubMed  Google Scholar 

  20. Girardin, S. E. et al. CARD4/Nod1 mediates NF-κB and JNK activation by invasive Shigella flexneri. EMBO Rep. 2, 736–742 (2001)

    Article  CAS  Google Scholar 

  21. Argast, G. M., Fausto, N. & Campbell, J. S. Inhibition of RIP2/RIck/CARDIAK activity by pyridinyl imidazole inhibitors of p38 MAPK. Mol. Cell. Biochem. 268, 129–140 (2005)

    Article  CAS  Google Scholar 

  22. Barthel, M. et al. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar Typhimurium colitis model that allows analysis of both pathogen and host. Infect. Immun. 71, 2839–2858 (2003)

    Article  CAS  Google Scholar 

  23. Wong, K. W., Mohammadi, S. & Isberg, R. R. The polybasic region of Rac1 modulates bacterial uptake independently of self-association and membrane targeting. J. Biol. Chem. 283, 35954–35965 (2008)

    Article  CAS  Google Scholar 

  24. Aktories, K., Schmidt, G. & Just, I. Rho GTPases as targets of bacterial protein toxins. Biol. Chem. 381, 421–426 (2000)

    Article  CAS  Google Scholar 

  25. Lopez, C. A. et al. Phage-mediated acquisition of a type III secreted effector protein boosts growth of Salmonella by nitrate respiration. mBio 3, e00143–12 (2012)

    Article  CAS  Google Scholar 

  26. Boyer, L. et al. Pathogen-derived effectors trigger protective immunity via activation of the Rac2 enzyme and the IMD or Rip kinase signaling pathway. Immunity 35, 536–549 (2011)

    Article  CAS  Google Scholar 

  27. Meylan, E. & Tschopp, J. The RIP kinases: crucial integrators of cellular stress. Trends Biochem. Sci. 30, 151–159 (2005)

    Article  CAS  Google Scholar 

  28. Tattoli, I., Travassos, L. H., Carneiro, L. A., Magalhaes, J. G. & Girardin, S. E. The Nodosome: Nod1 and Nod2 control bacterial infections and inflammation. Semin. Immunopathol. 29, 289–301 (2007)

    Article  CAS  Google Scholar 

  29. Fukazawa, A. et al. GEF-H1 mediated control of NOD1 dependent NF-κB activation by Shigella effectors. PLoS Pathogens 4, e1000228 (2008)

    Article  Google Scholar 

  30. Sambrook, J., Fritsch, E. F. & Maniatis, T. Molecular Cloning: a Laboratory Manual (Cold Spring Harb. Lab. Press, 1989)

    Google Scholar 

  31. Schmieger, H. Phage P22-mutants with increased or decreased transduction abilities. Mol. Gen. Genet. 119, 75–88 (1972)

    Article  CAS  Google Scholar 

  32. Lawes, M. & Maloy, S. MudSacI, a transposon with strong selectable and counterselectable markers: use for rapid mapping of chromosomal mutations in Salmonella typhimurium. J. Bacteriol. 177, 1383–1387 (1995)

    Article  CAS  Google Scholar 

  33. Keller, A., Nesvizhskii, A. I., Kolker, E. & Aebersold, R. Empirical statistical model to estimate the accuracy of peptide identifications made by MS/MS and database search. Anal. Chem. 74, 5383–5392 (2002)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank S.-P. Nuccio for providing PCR primers for the construction of the bacterial strains. This work was supported by Public Health Service Grants AI044170 and AI076246. A.M.K. is supported by the American Heart Association Grant 12SDG12220022.

Author information

Authors and Affiliations

Authors

Contributions

A.M.K. contributed to the experimental design, performed experiments and contributed to Figs 1b–f, 2a–e, g, 3a–c, f, g and Supplementary Figs 1, 2d, 3a, b, d, 4a–c, 7 and 8. M.G.W. performed experiments, constructed bacterial strains and contributed to Figs 1a, e, f, 3d, g and Supplementary Figs 2a, b, e, h, g, e, 4c, 5b and 8. J.J.A. constructed expression plasmids and contributed to Figs 2f and 3c and Supplementary Fig. 7. S.P.F. constructed expression plasmids and contributed to Fig. 2g and Supplementary Figs 2f, 3c and 6a, b. M.N.X. contributed to Fig. 1e, f and Supplementary Fig. 5a. S.E.W. constructed bacterial strains, contributed to Supplementary Fig. 5b and critically read the manuscript. A.K. constructed expression plasmids and contributed to Supplementary Figs 2c and 7. V.P. constructed bacterial strains. M.M.R. contributed to Supplementary Fig. 2d. J.F.T.W. constructed expression plasmids. R.A.E. performed mass spectrometry. A.M.K., A.J.B. and R.M.T. provided financial support for the study and contributed to the experimental design. A.M.K. and A.J.B. were responsible for the overall study design and for writing the manuscript.

Corresponding author

Correspondence to Andreas J. Bäumler.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-3, Supplementary References, and Supplementary Figures 1-8. (PDF 1254 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Keestra, A., Winter, M., Auburger, J. et al. Manipulation of small Rho GTPases is a pathogen-induced process detected by NOD1. Nature 496, 233–237 (2013). https://doi.org/10.1038/nature12025

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12025

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