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:

Pathogen blocks host death receptor signalling by arginine GlcNAcylation of death domains

Abstract

The tumour necrosis factor (TNF) family is crucial for immune homeostasis, cell death and inflammation. These cytokines are recognized by members of the TNF receptor (TNFR) family of death receptors, including TNFR1 and TNFR2, and FAS and TNF-related apoptosis-inducing ligand (TRAIL) receptors1. Death receptor signalling requires death-domain-mediated homotypic/heterotypic interactions between the receptor and its downstream adaptors, including TNFR1-associated death domain protein (TRADD) and FAS-associated death domain protein (FADD)2. Here we discover that death domains in several proteins, including TRADD, FADD, RIPK1 and TNFR1, were directly inactivated by NleB, an enteropathogenic Escherichia coli (EPEC) type III secretion system effector known to inhibit host nuclear factor-κB (NF-κB) signalling3,4. NleB contained an unprecedented N-acetylglucosamine (GlcNAc) transferase activity that specifically modified a conserved arginine in these death domains (Arg 235 in the TRADD death domain). NleB GlcNAcylation (the addition of GlcNAc onto a protein side chain) of death domains blocked homotypic/heterotypic death domain interactions and assembly of the oligomeric TNFR1 complex, thereby disrupting TNF signalling in EPEC-infected cells, including NF-κB signalling, apoptosis and necroptosis. Type-III-delivered NleB also blocked FAS ligand and TRAIL-induced cell death by preventing formation of a FADD-mediated death-inducing signalling complex (DISC). The arginine GlcNAc transferase activity of NleB was required for bacterial colonization in the mouse model of EPEC infection. The mechanism of action of NleB represents a new model by which bacteria counteract host defences, and also a previously unappreciated post-translational modification.

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: NleB blocks TNF-α signalling by directly targeting the TRADD death domain.
Figure 2: NleB GlcNAcylates the TRADD death domain and disrupts its oligomerization.
Figure 3: NleB GlcNAcylates Arg 235 that is required for TRADD function.
Figure 4: NleB GlcNAcylates FADD and other death domain proteins, and blocks FAS- and TRAIL-induced apoptosis.
Figure 5: Disrupting several death receptor pathways by NleB GlcNAc transferase activity that is required for bacterial colonization in vivo.

Similar content being viewed by others

References

  1. Wilson, N. S., Dixit, V. & Ashkenazi, A. Death receptor signal transducers: nodes of coordination in immune signaling networks. Nature Immunol. 10, 348–355 (2009)

    Article  CAS  Google Scholar 

  2. Kersse, K., Verspurten, J., Vanden Berghe, T. & Vandenabeele, P. The death-fold superfamily of homotypic interaction motifs. Trends Biochem. Sci. 36, 541–552 (2011)

    Article  CAS  Google Scholar 

  3. Nadler, C. et al. The type III secretion effector NleE inhibits NF-κB activation. PLoS Pathog. 6, e1000743 (2010)

    Article  Google Scholar 

  4. Newton, H. J. et al. The type III effectors NleE and NleB from enteropathogenic E. coli and OspZ from Shigella block nuclear translocation of NF-kappaB p65. PLoS Pathog. 6, e1000898 (2010)

    Article  Google Scholar 

  5. Yen, H. et al. NleC, a type III secretion protease, compromises NF-κB activation by targeting p65/RelA. PLoS Pathog. 6, e1001231 (2010)

    Article  CAS  Google Scholar 

  6. Baruch, K. et al. Metalloprotease type III effectors that specifically cleave JNK and NF-κB. EMBO J. 30, 221–231 (2011)

    Article  CAS  Google Scholar 

  7. Mühlen, S., Ruchaud-Sparagano, M. H. & Kenny, B. Proteasome-independent degradation of canonical NFκB complex components by the NleC protein of pathogenic Escherichia coli. J. Biol. Chem. 286, 5100–5107 (2011)

    Article  Google Scholar 

  8. Pearson, J. S., Riedmaier, P., Marches, O., Frankel, G. & Hartland, E. L. A type III effector protease NleC from enteropathogenic Escherichia coli targets NF-κB for degradation. Mol. Microbiol. 80, 219–230 (2011)

    Article  CAS  Google Scholar 

  9. Ruchaud-Sparagano, M. H., Muhlen, S., Dean, P. & Kenny, B. The enteropathogenic E. coli (EPEC) Tir effector inhibits NF-κB activity by targeting TNFα receptor-associated factors. PLoS Pathog. 7, e1002414 (2011)

    Article  CAS  Google Scholar 

  10. Yan, D., Wang, X., Luo, L., Cao, X. & Ge, B. Inhibition of TLR signaling by a bacterial protein containing immunoreceptor tyrosine-based inhibitory motifs. Nature Immunol. 13, 1063–1071 (2012)

    Article  CAS  Google Scholar 

  11. Vossenkämper, A. et al. Inhibition of NF-κB signaling in human dendritic cells by the enteropathogenic Escherichia coli effector protein NleE. J. Immunol. 185, 4118–4127 (2010)

    Article  Google Scholar 

  12. Zhang, L. et al. Cysteine methylation disrupts ubiquitin-chain sensing in NF-κB activation. Nature 481, 204–208 (2012)

    Article  ADS  CAS  Google Scholar 

  13. Kelly, M. et al. Essential role of the type III secretion system effector NleB in colonization of mice by Citrobacter rodentium. Infect. Immun. 74, 2328–2337 (2006)

    Article  CAS  Google Scholar 

  14. Misyurina, O. et al. The role of Tir, EspA, and NleB in the colonization of cattle by Shiga toxin producing Escherichia coli O26:H11. Can. J. Microbiol. 56, 739–747 (2010)

    Article  CAS  Google Scholar 

  15. Wickham, M. E. et al. Bacterial genetic determinants of non-O157 STEC outbreaks and hemolytic-uremic syndrome after infection. J. Infect. Dis. 194, 819–827 (2006)

    Article  CAS  Google Scholar 

  16. Kujat Choy, S. L. et al. SseK1 and SseK2 are novel translocated proteins of Salmonella enterica serovar Typhimurium. Infect. Immun. 72, 5115–5125 (2004)

    Article  Google Scholar 

  17. Lamkanfi, M. & Dixit, V. M. Manipulation of host cell death pathways during microbial infections. Cell Host Microbe 8, 44–54 (2010)

    Article  CAS  Google Scholar 

  18. Aggarwal, B. B. Signalling pathways of the TNF superfamily: a double-edged sword. Nature Rev. Immunol. 3, 745–756 (2003)

    Article  CAS  Google Scholar 

  19. Chen, N. J. et al. Beyond tumor necrosis factor receptor: TRADD signaling in toll-like receptors. Proc. Natl Acad. Sci. USA 105, 12429–12434 (2008)

    Article  ADS  CAS  Google Scholar 

  20. Ermolaeva, M. A. et al. Function of TRADD in tumor necrosis factor receptor 1 signaling and in TRIF-dependent inflammatory responses. Nature Immunol. 9, 1037–1046 (2008)

    Article  CAS  Google Scholar 

  21. Pobezinskaya, Y. L. et al. The function of TRADD in signaling through tumor necrosis factor receptor 1 and TRIF-dependent Toll-like receptors. Nature Immunol. 9, 1047–1054 (2008)

    Article  CAS  Google Scholar 

  22. Park, H. H. et al. The death domain superfamily in intracellular signaling of apoptosis and inflammation. Annu. Rev. Immunol. 25, 561–586 (2007)

    Article  CAS  Google Scholar 

  23. Hsu, H., Shu, H. B., Pan, M. G. & Goeddel, D. V. TRAdeath domain-TRAF2 and TRAdeath domain-FADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell 84, 299–308 (1996)

    Article  CAS  Google Scholar 

  24. Belyi, Y. et al. Legionella pneumophila glucosyltransferase inhibits host elongation factor 1A. Proc. Natl Acad. Sci. USA 103, 16953–16958 (2006)

    Article  ADS  CAS  Google Scholar 

  25. Lairson, L. L., Henrissat, B., Davies, G. J. & Withers, S. G. Glycosyltransferases: structures, functions, and mechanisms. Annu. Rev. Biochem. 77, 521–555 (2008)

    Article  CAS  Google Scholar 

  26. Belyi, Y. & Aktories, K. Bacterial toxin and effector glycosyltransferases. Biochim. Biophys. Acta 1800, 134–143 (2010)

    Article  CAS  Google Scholar 

  27. Gao, X. et al. NleB, a bacterial effector with glycosyltransferase activity, targets GAPDH function to inhibit NF-κB activation. Cell Host Microbe 13, 87–99 (2013)

    Article  CAS  Google Scholar 

  28. Love, D. C. & Hanover, J. A. The hexosamine signaling pathway: deciphering the “O-GlcNAc code”. Sci. STKE 2005, re13 (2005)

    PubMed  Google Scholar 

  29. Hart, G. W., Slawson, C., Ramirez-Correa, G. & Lagerlof, O. Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease. Annu. Rev. Biochem. 80, 825–858 (2011)

    Article  CAS  Google Scholar 

  30. Singh, D. G. et al. β-Glucosylarginine: a new glucose-protein bond in a self-glucosylating protein from sweet corn. FEBS Lett. 376, 61–64 (1995)

    Article  ADS  CAS  Google Scholar 

  31. Ge, J. et al. A Legionella type IV effector activates the NF-κB pathway by phosphorylating the IκB family of inhibitors. Proc. Natl Acad. Sci. USA 106, 13725–13730 (2009)

    Article  ADS  CAS  Google Scholar 

  32. Li, H. et al. The phosphothreonine lyase activity of a bacterial type III effector family. Science 315, 1000–1003 (2007)

    Article  ADS  CAS  Google Scholar 

  33. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012)

    Article  CAS  Google Scholar 

  34. Wang, Z., Jiang, H., Chen, S., Du, F. & Wang, X. The mitochondrial phosphatase PGAM5 functions at the convergence point of multiple necrotic death pathways. Cell 148, 228–243 (2012)

    Article  CAS  Google Scholar 

  35. Wang, G. et al. Small-molecule activation of the TRAIL receptor DR5 in human cancer cells. Nature Chem. Biol. 9, 84–89 (2013)

    Article  ADS  Google Scholar 

  36. Wang, L., Du, F. & Wang, X. TNF-α induces two distinct caspase-8 activation pathways. Cell 133, 693–703 (2008)

    Article  CAS  Google Scholar 

  37. Cui, J. et al. Glutamine deamidation and dysfunction of ubiquitin/NEDD8 induced by a bacterial effector family. Science 329, 1215–1218 (2010)

    Article  ADS  CAS  Google Scholar 

  38. Horton, D., Hughes, J. B., Jewell, J. S., Philips, K. D. & Turner, W. N. Anomeric equilibria in derivatives of amino sugars. Nuclear magnetic resonance studies on acetylated amino sugars and specifically deuterated analogs. J. Org. Chem. 32, 1073–1080 (1967)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank I. Rosenshine for providing nleBE deletion EPEC strains, H. Wu for the TNFR1/2 plasmid, K. Iwai for LUBAC antibodies, H. Sakurai for the p-TAK1 antibody, Z. Yang for yeast two-hybrid screen, L. Feng for synthesizing Ac4GlcNAc-d3, M. Dong for accessing the ETD instrument and X. Liu for assistance with mass spectrometry data analyses. We also thank members of the Shao laboratory for discussions and technical assistance. The research was supported in part by an International Early Career Scientist grant from the Howard Hughes Medical Institute to F.S. This work was also supported by the National Basic Research Program of China 973 Programs 2010CB835400 and 2012CB518700 to F.S. and 2010CB835204 to S.C.

Author information

Authors and Affiliations

Authors

Contributions

S.L. and L.Z. performed most experiments, assisted by Q.Y., N.D. and W.G. L.L., X.D. and S.C. performed mass spectrometry analyses and S.C. analysed the data. L.S., J.R. and X.C. contributed reagents and analytic tools. S.L., L.Z. and F.S. analysed the data and wrote the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to She Chen or Feng Shao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-26. (PDF 5078 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Li, S., Zhang, L., Yao, Q. et al. Pathogen blocks host death receptor signalling by arginine GlcNAcylation of death domains. Nature 501, 242–246 (2013). https://doi.org/10.1038/nature12436

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12436

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 Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology