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:

Activation of Bacteroides fragilis toxin by a novel bacterial protease contributes to anaerobic sepsis in mice

Abstract

Bacteroides fragilis is the leading cause of anaerobic bacteremia and sepsis1. Enterotoxigenic strains that produce B. fragilis toxin (BFT, fragilysin) contribute to colitis2 and intestinal malignancy3, yet are also isolated in bloodstream infection4,5. It is not known whether these strains harbor unique genetic determinants that confer virulence in extra-intestinal disease. We demonstrate that BFT contributes to sepsis in mice, and we identify a B. fragilis protease called fragipain (Fpn) that is required for the endogenous activation of BFT through the removal of its auto-inhibitory prodomain. Structural analysis of Fpn reveals a His–Cys catalytic dyad that is characteristic of C11-family cysteine proteases that are conserved in multiple pathogenic Bacteroides spp. and Clostridium spp. Fpn-deficient, enterotoxigenic B. fragilis has an attenuated ability to induce sepsis in mice; however, Fpn is dispensable in B. fragilis colitis, wherein host proteases mediate BFT activation. Our findings define a role for B. fragilis enterotoxin and its activating protease in the pathogenesis of bloodstream infection, which indicates a greater complexity of cellular targeting and activity of BFT than previously recognized. The expression of fpn by both toxigenic and nontoxigenic strains suggests that this protease may contribute to anaerobic sepsis in ways that extend beyond its role in toxin activation. It could thus potentially serve as a target for disease 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: BFT contributes to lethal sepsis.
Figure 2: BFT is activated by a novel B. fragilis cysteine protease.
Figure 3: Sequence and molecular structure of B. fragilis fragipain (Fpn).
Figure 4: Selective requirement for Fpn in lethal sepsis.

Similar content being viewed by others

Accession codes

Primary accessions

NCBI Reference Sequence

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Robert, R., Deraignac, A., Le Moal, G., Ragot, S. & Grollier, G. Prognostic factors and impact of antibiotherapy in 117 cases of anaerobic bacteraemia. Eur. J. Clin. Microbiol. Infect. Dis. 27, 671–678 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Rhee, K.J. et al. Induction of persistent colitis by a human commensal, enterotoxigenic Bacteroides fragilis, in wild-type C57BL/6 mice. Infect. Immun. 77, 1708–1718 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Wu, S. et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat. Med. 15, 1016–1022 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kato, N., Kato, H., Watanabe, K. & Ueno, K. Association of enterotoxigenic Bacteroides fragilis with bacteremia. Clin. Infect. Dis. 23 (suppl. 1), S83–S86 (1996).

    Article  PubMed  Google Scholar 

  5. Claros, M.C. et al. Characterization of the Bacteroides fragilis pathogenicity island in human blood culture isolates. Anaerobe 12, 17–22 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Redondo, M.C., Arbo, M.D., Grindlinger, J. & Snydman, D.R. Attributable mortality of bacteremia associated with the Bacteroides fragilis group. Clin. Infect. Dis. 20, 1492–1496 (1995).

    Article  CAS  PubMed  Google Scholar 

  7. Ngo, J.T. et al. Population-based assessment of the incidence, risk factors, and outcomes of anaerobic bloodstream infections. Infection 41, 41–48 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Rocha, E.R. & Smith, C.J. Ferritin-like family proteins in the anaerobe Bacteroides fragilis: when an oxygen storm is coming, take your iron to the shelter. Biometals 26, 577–591 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Brook, I. & Frazier, E.H. Aerobic and anaerobic microbiology in intra-abdominal infections associated with diverticulitis. J. Med. Microbiol. 49, 827–830 (2000).

    Article  CAS  PubMed  Google Scholar 

  10. Vena, A. et al. Are incidence and epidemiology of anaerobic bacteremia really changing? Eur. J. Clin. Microbiol. Infect. Dis. 34, 1621–1629 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Ani, C., Farshidpanah, S., Bellinghausen Stewart, A. & Nguyen, H.B. Variations in organism-specific severe sepsis mortality in the United States: 1999–2008. Crit. Care Med. 43, 65–77 (2015).

    Article  PubMed  Google Scholar 

  12. Moghadamyeghaneh, Z. et al. A comparison of outcomes of emergent, urgent, and elective surgical treatment of diverticulitis. Am. J. Surg. 210, 838–845 (2015).

    Article  PubMed  Google Scholar 

  13. Snydman, D.R. et al. Lessons learned from the anaerobe survey: historical perspective and review of the most recent data (2005–2007). Clin. Infect. Dis. 50 (suppl. 1), S26–S33 (2010).

    Article  PubMed  Google Scholar 

  14. Kasper, D.L., Onderdonk, A.B., Crabb, J. & Bartlett, J.G. Protective efficacy of immunization with capsular antigen against experimental infection with Bacteroides fragilis. J. Infect. Dis. 140, 724–731 (1979).

    Article  CAS  PubMed  Google Scholar 

  15. Hartmeyer, G.N., Sóki, J., Nagy, E. & Justesen, U.S. Multidrug-resistant Bacteroides fragilis group on the rise in Europe? J. Med. Microbiol. 61, 1784–1788 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Wang, F.D., Liao, C.H., Lin, Y.T., Sheng, W.H. & Hsueh, P.R. Trends in the susceptibility of commonly encountered clinically significant anaerobes and susceptibilities of blood isolates of anaerobes to 16 antimicrobial agents, including fidaxomicin and rifaximin, 2008–2012, northern Taiwan. Eur. J. Clin. Microbiol. Infect. Dis. 33, 2041–2052 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Franco, A.A. et al. Molecular evolution of the pathogenicity island of enterotoxigenic Bacteroides fragilis strains. J. Bacteriol. 181, 6623–6633 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Moncrief, J.S. et al. The enterotoxin of Bacteroides fragilis is a metalloprotease. Infect. Immun. 63, 175–181 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Franco, A.A. et al. Cloning and characterization of the Bacteroides fragilis metalloprotease toxin gene. Infect. Immun. 65, 1007–1013 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wu, S., Lim, K.C., Huang, J., Saidi, R.F. & Sears, C.L. Bacteroides fragilis enterotoxin cleaves the zonula adherens protein, E-cadherin. Proc. Natl. Acad. Sci. USA 95, 14979–14984 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Goulas, T., Arolas, J.L. & Gomis-Rüth, F.X. Structure, function and latency regulation of a bacterial enterotoxin potentially derived from a mammalian adamalysin/ADAM xenolog. Proc. Natl. Acad. Sci. USA 108, 1856–1861 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kim, J.M. et al. Nuclear factor-κB activation pathway in intestinal epithelial cells is a major regulator of chemokine gene expression and neutrophil migration induced by Bacteroides fragilis enterotoxin. Clin. Exp. Immunol. 130, 59–66 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Powers, M.E., Kim, H.K., Wang, Y. & Bubeck Wardenburg, J. ADAM10 mediates vascular injury induced by Staphylococcus aureus α-hemolysin. J. Infect. Dis. 206, 352–356 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Inoshima, I. et al. A Staphylococcus aureus pore-forming toxin subverts the activity of ADAM10 to cause lethal infection in mice. Nat. Med. 17, 1310–1314 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Powers, M.E., Becker, R.E., Sailer, A., Turner, J.R. & Bubeck Wardenburg, J. Synergistic action of Staphylococcus aureus α-toxin on platelets and myeloid lineage cells contributes to lethal sepsis. Cell Host Microbe 17, 775–787 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Franco, A.A., Buckwold, S.L., Shin, J.W., Ascon, M. & Sears, C.L. Mutation of the zinc-binding metalloprotease motif affects Bacteroides fragilis toxin activity but does not affect propeptide processing. Infect. Immun. 73, 5273–5277 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Van Tassell, R.L., Lyerly, D.M. & Wilkins, T.D. Purification and characterization of an enterotoxin from Bacteroides fragilis. Infect. Immun. 60, 1343–1350 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Labrou, N.E. & Rigden, D.J. The structure-function relationship in the clostripain family of peptidases. Eur. J. Biochem. 271, 983–992 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. McLuskey, K. & Mottram, J.C. Comparative structural analysis of the caspase family with other clan CD cysteine peptidases. Biochem. J. 466, 219–232 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Dargatz, H., Diefenthal, T., Witte, V., Reipen, G. & von Wettstein, D. The heterodimeric protease clostripain from Clostridium histolyticum is encoded by a single gene. Mol. Gen. Genet. 240, 140–145 (1993).

    Article  CAS  PubMed  Google Scholar 

  31. Shokhen, M., Khazanov, N. & Albeck, A. Challenging a paradigm: theoretical calculations of the protonation state of the Cys25-His159 catalytic diad in free papain. Proteins 77, 916–926 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Vivares, D., Arnoux, P. & Pignol, D. A papain-like enzyme at work: native and acyl-enzyme intermediate structures in phytochelatin synthesis. Proc. Natl. Acad. Sci. USA 102, 18848–18853 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wei, D., Huang, X., Liu, J., Tang, M. & Zhan, C.G. Reaction pathway and free energy profile for papain-catalyzed hydrolysis of N-acetyl-Phe-Gly 4-nitroanilide. Biochemistry 52, 5145–5154 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Angus, D.C. & van der Poll, T. Severe sepsis and septic shock. N. Engl. J. Med. 369, 840–851 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Ortega, E., Abriouel, H., Lucas, R. & Gálvez, A. Multiple roles of Staphylococcus aureus enterotoxins: pathogenicity, superantigenic activity, and correlation to antibiotic resistance. Toxins (Basel) 2, 2117–2131 (2010).

    Article  CAS  Google Scholar 

  36. Mayer, C.L., Leibowitz, C.S., Kurosawa, S. & Stearns-Kurosawa, D.J. Shiga toxins and the pathophysiology of hemolytic uremic syndrome in humans and animals. Toxins (Basel) 4, 1261–1287 (2012).

    Article  CAS  Google Scholar 

  37. Inoshima, N., Wang, Y. & Bubeck Wardenburg, J. Genetic requirement for ADAM10 in severe Staphylococcus aureus skin infection. J. Invest. Dermatol. 132, 1513–1516 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Cui, L. et al. An ADAM10 promoter polymorphism is a functional variant in severe sepsis patients and confers susceptibility to the development of sepsis. Crit. Care 19, 73 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Rawlings, N.D., Barrett, A.J. & Bateman, A. MEROPS: the database of proteolytic enzymes, their substrates and inhibitors. Nucleic Acids Res. 40, D343–D350 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Comstock, L.E. et al. Analysis of a capsular polysaccharide biosynthesis locus of Bacteroides fragilis. Infect. Immun. 67, 3525–3532 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Melton-Celsa, A.R., Darnell, S.C. & O'Brien, A.D. Activation of Shiga-like toxins by mouse and human intestinal mucus correlates with virulence of enterohemorrhagic Escherichia coli O91:H21 isolates in orally infected, streptomycin-treated mice. Infect. Immun. 64, 1569–1576 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Goodman, A.L. et al. Identifying genetic determinants needed to establish a human gut symbiont in its habitat. Cell Host Microbe 6, 279–289 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Koropatkin, N.M., Martens, E.C., Gordon, J.I. & Smith, T.J. Starch catabolism by a prominent human gut symbiont is directed by the recognition of amylose helices. Structure 16, 1105–1115 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Winter, G. xia2: an expert system for macromolecular crystallography data reduction. J. Appl. Cryst. 43, 186–190 (2010).

    Article  CAS  Google Scholar 

  45. Kabsch, W. Xds. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Evans, P.R. & Murshudov, G.N. How good are my data and what is the resolution? Acta Crystallogr. D Biol. Crystallogr. 69, 1204–1214 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Morin, A. et al. Collaboration gets the most out of software. eLife 2, e01456 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Adams, P.D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Arnold, K., Bordoli, L., Kopp, J. & Schwede, T. The SWISS-MODEL workspace: a web-based environment for protein structure homology modelling. Bioinformatics 22, 195–201 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Emsley, P., Lohkamp, B., Scott, W.G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the Department of Pediatrics at the University of Chicago and with a Pilot and Feasibility Award from the Digestive Diseases Research Core Center at the University of Chicago to J.B.W. (NIDDK P30DK42086); by a Burroughs Wellcome Foundation Investigators in the Pathogenesis of Infectious Disease Fellowship (J.B.W.); and by the US National Institutes of Health Medical Scientist Training Program at the University of Chicago (GM007281 to V.M.C. and A.L.H.). We thank D. Hecht (Loyola University) for his generous guidance on the establishment of a B. fragilis program and for B. fragilis strain 638R/TM4000; D. Missiakas (University of Chicago) for α-RpoA antisera; J. Gordon (Washington University) for the pSAM-Bt and pKNOCK vectors and Escherichia coli S17 strain; and Y. Fu (University of Texas Southwestern Medical Center) for germ-free mice.

Author information

Authors and Affiliations

Authors

Contributions

V.M.C. developed the experimental concepts and approach, generated all bacterial strains, performed transposon-mutagenesis screening and studies on colonic epithelial injury and sepsis in vivo and in vitro, and contributed to writing the manuscript. J.H. generated and analyzed the Fpn crystal structure and contributed to writing the manuscript. A.L.H. contributed essential bacterial genetic reagents and expertise. W.P.T. performed studies on the endothelium. J.R.T. performed pathology analysis of colonic tissues. S.C. analyzed the Fpn crystal structure and contributed to writing the manuscript. J.B.W. developed the experimental concepts and approach and contributed to writing the manuscript.

Corresponding author

Correspondence to Juliane Bubeck Wardenburg.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Tables 1–2 (PDF 1595 kb)

Source data

Source data for Supplementary Figures 1, 3, 4 (XLS 35 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Choi, V., Herrou, J., Hecht, A. et al. Activation of Bacteroides fragilis toxin by a novel bacterial protease contributes to anaerobic sepsis in mice. Nat Med 22, 563–567 (2016). https://doi.org/10.1038/nm.4077

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4077

This article is cited by

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