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.

  • Article
  • Published:

Structure of the full-length Clostridium difficile toxin B

Abstract

Clostridium difficile is an opportunistic pathogen that establishes in the colon when the gut microbiota are disrupted by antibiotics or disease. C. difficile infection (CDI) is largely caused by two virulence factors, TcdA and TcdB. Here, we report a 3.87-Å-resolution crystal structure of TcdB holotoxin that captures a unique conformation of TcdB at endosomal pH. Complementary biophysical studies suggest that the C-terminal combined repetitive oligopeptides (CROPs) domain of TcdB is dynamic and can sample open and closed conformations that may facilitate modulation of TcdB activity in response to environmental and cellular cues during intoxication. Furthermore, we report three crystal structures of TcdB–antibody complexes that reveal how antibodies could specifically inhibit the activities of individual TcdB domains. Our studies provide novel insight into the structure and function of TcdB holotoxin and identify intrinsic vulnerabilities that could be exploited to develop new therapeutics and vaccines for the treatment of CDI.

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

Fig. 1: Overall structure of the full-length TcdB holotoxin.
Fig. 2: The unique structure of the CROPs of TcdB.
Fig. 3: The CROPs undergoes pH-dependent conformational changes.
Fig. 4: TcdB shows a novel conformation in the pore-forming region at endosomal pH.
Fig. 5: Three modulatory components are clustered at the ‘heart’ of TcdB holotoxin.

Similar content being viewed by others

Data availability

Atomic coordinates and structure factors of the TcdB–5D–E3–7F, TcdB1072–1433–5D, GTD–E3, and GTDVPI10463–7F complexes have been deposited in the Protein Data Bank under accession codes PDB 6OQ5, PDB 6OQ6, PDB 6OQ7, and PDB 6OQ8, respectively. Source data for Fig. 4e,f are available with the paper online. Other data are available upon request.

References

  1. Rupnik, M., Wilcox, M. H. & Gerding, D. N. Š infection: new developments in epidemiology and pathogenesis. Nat. Rev. Microbiol. 7, 526–536 (2009).

    Article  CAS  Google Scholar 

  2. Lessa, F. C. et al. Burden of Clostridium difficile infection in the United States. N. Engl. J. Med. 372, 825–834 (2015).

    Article  CAS  Google Scholar 

  3. Jank, T. & Aktories, K. Structure and mode of action of clostridial glucosylating toxins: the ABCD model. Trends Microbiol. 16, 222–229 (2008).

    Article  CAS  Google Scholar 

  4. Seal, D. et al. Treatment of relapsing Clostridium difficile diarrhoea by administration of a non-toxigenic strain. Eur. J. Clin. Microbiol. 6, 51–53 (1987).

    Article  CAS  Google Scholar 

  5. Kuehne, S. A. et al. The role of toxin A and toxin B in Clostridium difficile infection. Nature 467, 711–713 (2010).

    Article  CAS  Google Scholar 

  6. Lyras, D. et al. Toxin B is essential for virulence of Clostridium difficile. Nature 458, 1176–1179 (2009).

    Article  CAS  Google Scholar 

  7. Carter, G. P. et al. Defining the roles of TcdA and TcdB in localized gastrointestinal disease, systemic organ damage, and the host response during Clostridium difficile infections. MBio 6, e00551 (2015).

    Article  CAS  Google Scholar 

  8. Carter, G. P., Rood, J. I. & Lyras, D. The role of toxin A and toxin B in the virulence of Clostridium difficile. Trends Microbiol. 20, 21–29 (2012).

    Article  CAS  Google Scholar 

  9. Wilcox, M. H. et al. Bezlotoxumab for prevention of recurrent Clostridium difficile infection. N. Engl. J. Med. 376, 305–317 (2017).

    Article  CAS  Google Scholar 

  10. Ho, J. G., Greco, A., Rupnik, M. & Ng, K. K. Crystal structure of receptor-binding C-terminal repeats from Clostridium difficile toxin A. Proc. Natl Acad. Sci. USA 102, 18373–18378 (2005).

    Article  CAS  Google Scholar 

  11. Aktories, K., Schwan, C. & Jank, T. Clostridium difficile toxin biology. Annu Rev. Microbiol. 71, 281–307 (2017).

    Article  CAS  Google Scholar 

  12. Greco, A. et al. Carbohydrate recognition by Clostridium difficile toxin A. Nat. Struct. Mol. Biol. 13, 460–461 (2006).

    Article  CAS  Google Scholar 

  13. Chen, P. et al. Structural basis for recognition of frizzled proteins by Clostridium difficile toxin B. Science 360, 664–669 (2018).

    Article  CAS  Google Scholar 

  14. Yuan, P. et al. Chondroitin sulfate proteoglycan 4 functions as the cellular receptor for Clostridium difficile toxin B. Cell Res. 25, 157–168 (2015).

    Article  CAS  Google Scholar 

  15. Genisyuerek, S. et al. Structural determinants for membrane insertion, pore formation and translocation of Clostridium difficile toxin B. Mol. Microbiol. 79, 1643–1654 (2011).

    Article  CAS  Google Scholar 

  16. Zhang, Z. et al. Translocation domain mutations affecting cellular toxicity identify the Clostridium difficile toxin B pore. Proc. Natl Acad. Sci. USA 111, 3721–3726 (2014).

    Article  CAS  Google Scholar 

  17. Qa’Dan, M., Spyres, L. M. & Ballard, J. D. pH-induced conformational changes in Clostridium difficile toxin B. Infect. Immun. 68, 2470–2474 (2000).

    Article  Google Scholar 

  18. Li, S. et al. Critical roles of Clostridium difficile toxin B enzymatic activities in pathogenesis. Infect. Immun. 83, 502–513 (2015).

    Article  Google Scholar 

  19. Sehr, P. et al. Glucosylation and ADP ribosylation of rho proteins: effects on nucleotide binding, GTPase activity, and effector coupling. Biochemistry 37, 5296–5304 (1998).

    Article  CAS  Google Scholar 

  20. Egerer, M., Giesemann, T., Jank, T., Satchell, K. J. & Aktories, K. Auto-catalytic cleavage of Clostridium difficile toxins A and B depends on cysteine protease activity. J. Biol. Chem. 282, 25314–25321 (2007).

    Article  CAS  Google Scholar 

  21. Reineke, J. et al. Autocatalytic cleavage of Clostridium difficile toxin B. Nature 446, 415–419 (2007).

    Article  CAS  Google Scholar 

  22. Just, I. et al. Glucosylation of Rho proteins by Clostridium difficile toxin B. Nature 375, 500–503 (1995).

    Article  CAS  Google Scholar 

  23. Hofmann, F., Busch, C., Prepens, U., Just, I. & Aktories, K. Localization of the glucosyltransferase activity of Clostridium difficile toxin B to the N-terminal part of the holotoxin. J. Biol. Chem. 272, 11074–11078 (1997).

    Article  CAS  Google Scholar 

  24. Chumbler, N. M. et al. Crystal structure of Clostridium difficile toxin A. Nat. Microbiol. 1, 15002 (2016).

    Article  CAS  Google Scholar 

  25. Murase, T. et al. Structural basis for antibody recognition in the receptor-binding domains of toxins A and B from Clostridium difficile. J. Biol. Chem. 289, 2331–2343 (2014).

    Article  CAS  Google Scholar 

  26. Reinert, D. J., Jank, T., Aktories, K. & Schulz, G. E. Structural basis for the function of Clostridium difficile toxin B. J. Mol. Biol. 351, 973–981 (2005).

    Article  CAS  Google Scholar 

  27. Shen, A. et al. Defining an allosteric circuit in the cysteine protease domain of Clostridium difficile toxins. Nat. Struct. Mol. Biol. 18, 364–371 (2011).

    Article  CAS  Google Scholar 

  28. Orth, P. et al. Mechanism of action and epitopes of Clostridium difficile toxin B-neutralizing antibody bezlotoxumab revealed by X-ray crystallography. J. Biol. Chem. 289, 18008–18021 (2014).

    Article  CAS  Google Scholar 

  29. Pruitt, R. N., Chambers, M. G., Ng, K. K., Ohi, M. D. & Lacy, D. B. Structural organization of the functional domains of Clostridium difficile toxins A and B. Proc. Natl Acad. Sci. USA 107, 13467–13472 (2010).

    Article  CAS  Google Scholar 

  30. Yang, G. L. et al. Expression of recombinant Clostridium difficile toxin A and B in Bacillus megaterium. BMC Microbiol. 8, 192 (2008).

    Article  Google Scholar 

  31. Yang, Z. et al. A novel multivalent, single-domain antibody targeting TcdA and TcdB prevents fulminant Clostridium difficile infection in mice. J. Infect. Dis. 210, 964–972 (2014).

    Article  CAS  Google Scholar 

  32. Kobe, B. & Kajava, A. V. When protein folding is simplified to protein coiling: the continuum of solenoid protein structures. Trends Biochem. Sci. 25, 509–515 (2000).

    Article  CAS  Google Scholar 

  33. Fernandez-Tornero, C., Lopez, R., Garcia, E., Gimenez-Gallego, G. & Romero, A. A novel solenoid fold in the cell wall anchoring domain of the pneumococcal virulence factor LytA. Nat. Struct. Biol. 8, 1020–1024 (2001).

    Article  CAS  Google Scholar 

  34. Zhang, Y. et al. A segment of 97 amino acids within the translocation domain of Clostridium difficile toxin B is essential for toxicity. PLoS One 8, e58634 (2013).

    Article  CAS  Google Scholar 

  35. Lanis, J. M., Barua, S. & Ballard, J. D. Variations in TcdB activity and the hypervirulence of emerging strains of Clostridium difficile. PLoS Pathog. 6, e1001061 (2010).

    Article  Google Scholar 

  36. Lanis, J. M., Hightower, L. D., Shen, A. & Ballard, J. D. TcdB from hypervirulent Clostridium difficile exhibits increased efficiency of autoprocessing. Mol. Microbiol. 84, 66–76 (2012).

    Article  CAS  Google Scholar 

  37. Larabee, J. L., Krumholz, A., Hunt, J. J., Lanis, J. M. & Ballard, J. D. Exposure of neutralizing epitopes in the carboxyl-terminal domain of TcdB is altered by a proximal hypervariable region. J. Biol. Chem. 290, 6975–6985 (2015).

    Article  CAS  Google Scholar 

  38. Putnam, C. D., Hammel, M., Hura, G. L. & Tainer, J. A. X-ray solution scattering (SAXS) combined with crystallography and computation: defining accurate macromolecular structures, conformations and assemblies in solution. Q Rev. Biophys. 40, 191–285 (2007).

    Article  CAS  Google Scholar 

  39. Kao, A. H. et al. Development of a novel cross-linking strategy for fast and accurate identification of cross-linked peptides of protein complexes. Mol. Cell. Proteomics 10, M110.002212 (2011).

    Article  Google Scholar 

  40. Yu, C. & Huang, L. Cross-linking mass spectrometry: an emerging technology for interactomics and structural biology. Anal. Chem. 90, 144–165 (2018).

    Article  CAS  Google Scholar 

  41. Hellenkamp, B. et al. Precision and accuracy of single-molecule FRET measurements-a multi-laboratory benchmark study. Nat. Methods 15, 669–676 (2018).

    Article  CAS  Google Scholar 

  42. McCann, J. J., Choi, U. B., Zheng, L., Weninger, K. & Bowen, M. E. Optimizing methods to recover absolute FRET efficiency from immobilized single molecules. Biophys. J. 99, 961–970 (2010).

    Article  CAS  Google Scholar 

  43. Gopich, I. V. & Szabo, A. FRET efficiency distributions of multistate single molecules. J. Phys. Chem. B 114, 15221–15226 (2010).

    Article  CAS  Google Scholar 

  44. Chumbler, N. M. et al. Clostridium difficile Toxin B causes epithelial cell necrosis through an autoprocessing-independent mechanism. PLoS Pathog. 8, e1003072 (2012).

    Article  CAS  Google Scholar 

  45. Lam, K. H. et al. A viral-fusion-peptide-like molecular switch drives membrane insertion of botulinum neurotoxin A1. Nat. Commun. 9, 5367 (2018).

    Article  Google Scholar 

  46. Schmidt, D. J. et al. A tetraspecific VHH-based neutralizing antibody modifies disease outcome in three animal models of Clostridium difficile infection. Clin. Vaccin. Immunol. 23, 774–784 (2016).

    Article  CAS  Google Scholar 

  47. Just, I. & Gerhard, R. Large clostridial cytotoxins. Rev. Physiol. Biochem. Pharm. 152, 23–47 (2004).

    Article  CAS  Google Scholar 

  48. Egerer, M. & Satchell, K. J. Inositol hexakisphosphate-induced autoprocessing of large bacterial protein toxins. PLoS Pathog. 6, e1000942 (2010).

    Article  Google Scholar 

  49. Lupardus, P. J., Shen, A., Bogyo, M. & Garcia, K. C. Small molecule-induced allosteric activation of the Vibrio cholerae RTX cysteine protease domain. Science 322, 265–268 (2008).

    Article  CAS  Google Scholar 

  50. Puri, A. W. et al. Rational design of inhibitors and activity-based probes targeting Clostridium difficile virulence factor TcdB. Chem. Biol. 17, 1201–1211 (2010).

    Article  CAS  Google Scholar 

  51. Olling, A. et al. The combined repetitive oligopeptides of Clostridium difficile toxin A counteract premature cleavage of the glucosyl-transferase domain by stabilizing protein conformation. Toxins (Basel) 6, 2162–2176 (2014).

    Article  CAS  Google Scholar 

  52. Zhang, Y., Hamza, T., Gao, S. & Feng, H. Masking autoprocessing of Clostridium difficile toxin A by the C-terminus combined repetitive oligo peptides. Biochem. Biophys. Res Commun. 459, 259–263 (2015).

    Article  CAS  Google Scholar 

  53. Geissler, B., Tungekar, R. & Satchell, K. J. Identification of a conserved membrane localization domain within numerous large bacterial protein toxins. Proc. Natl Acad. Sci. USA 107, 5581–5586 (2010).

    Article  CAS  Google Scholar 

  54. Varela Chavez, C. et al. The tip of the four N-terminal alpha-helices of Clostridium sordellii lethal toxin contains the interaction site with membrane phosphatidylserine facilitating small GTPases glucosylation. Toxins (Basel) 8, 90 (2016).

    Article  Google Scholar 

  55. Mesmin, B. et al. A phosphatidylserine-binding site in the cytosolic fragment of Clostridium sordellii lethal toxin facilitates glucosylation of membrane-bound Rac and is required for cytotoxicity. J. Biol. Chem. 279, 49876–49882 (2004).

    Article  CAS  Google Scholar 

  56. Schorch, B. et al. LRP1 is a receptor for Clostridium perfringens TpeL toxin indicating a two-receptor model of clostridial glycosylating toxins. Proc. Natl Acad. Sci. USA 111, 6431–6436 (2014).

    Article  CAS  Google Scholar 

  57. Kreimeyer, I. et al. Autoproteolytic cleavage mediates cytotoxicity of Clostridium difficile toxin A. Naunyn Schmiede. Arch. Pharm. 383, 253–262 (2011).

    Article  CAS  Google Scholar 

  58. Gupta, P. et al. Functional defects in Clostridium difficile TcdB toxin uptake identify CSPG4 receptor-binding determinants. J. Biol. Chem. 292, 17290–17301 (2017).

    Article  CAS  Google Scholar 

  59. Tao, L. et al. Frizzled proteins are colonic epithelial receptors for C. difficile toxin B. Nature 538, 350–355 (2016).

    Article  CAS  Google Scholar 

  60. Laursen, N. S. et al. Universal protection against influenza infection by a multidomain antibody to influenza hemagglutinin. Science 362, 598–602 (2018).

    Article  CAS  Google Scholar 

  61. Otwinowski, Z. & Minor, W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  Google Scholar 

  62. Kabsch, W. XDS. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  Google Scholar 

  63. 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  Google Scholar 

  64. Gu, S. et al. Botulinum neurotoxin is shielded by NTNHA in an interlocked complex. Science 335, 977–981 (2012).

    Article  CAS  Google Scholar 

  65. 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  Google Scholar 

  66. Murshudov, G. N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D Biol. Crystallogr. 67, 355–367 (2011).

    Article  CAS  Google Scholar 

  67. DiMaio, F. et al. Improved low-resolution crystallographic refinement with Phenix and Rosetta. Nat. Methods 10, 1102–1104 (2013).

    Article  CAS  Google Scholar 

  68. Strong, M. et al. Toward the structural genomics of complexes: crystal structure of a PE/PPE protein complex from Mycobacterium tuberculosis. Proc. Natl Acad. Sci. USA 103, 8060–8065 (2006).

    Article  CAS  Google Scholar 

  69. Brunger, A. T. Free R value: a novel statistical quantity for assessing the accuracy of crystal structures. Nature 355, 472–475 (1992).

    Article  CAS  Google Scholar 

  70. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 66, 12–21 (2010).

    Article  CAS  Google Scholar 

  71. Petoukhov, M. V., Konarev, P. V., Kikhney, A. G. & Svergun, D. I. ATSAS 2.1 - towards automated and web-supported small-angle scattering data analysis. J. Appl. Crystallogr. 40, S223–S228 (2007).

    Article  CAS  Google Scholar 

  72. Svergun, D. I. Determination of the regularization parameter in indirect-transform methods using perceptual criteria. J. Appl. Crystallogr. 25, 495–503 (1992).

    Article  CAS  Google Scholar 

  73. Svergun, D., Barberato, C. & Koch, M. H. J. CRYSOL - A program to evaluate x-ray solution scattering of biological macromolecules from atomic coordinates. J. Appl. Crystallogr. 28, 768–773 (1995).

    Article  CAS  Google Scholar 

  74. Leitner, A., Walzthoeni, T. & Aebersold, R. Lysine-specific chemical cross-linking of protein complexes and identification of cross-linking sites using LC-MS/MS and the xQuest/xProphet software pipeline. Nat. Protoc. 9, 120–137 (2014).

    Article  CAS  Google Scholar 

  75. Yu, C., Kandur, W., Kao, A., Rychnovsky, S. & Huang, L. Developing new isotope-coded mass spectrometry-cleavable cross-linkers for elucidating protein structures. Anal. Chem. 86, 2099–2106 (2014).

    Article  CAS  Google Scholar 

  76. Yu, C. et al. Developing a multiplexed quantitative cross-linking mass spectrometry platform for comparative structural analysis of protein complexes. Anal. Chem. 88, 10301–10308 (2016).

    Article  CAS  Google Scholar 

  77. Swoboda, M. et al. Enzymatic oxygen scavenging for photostability without pH drop in single-molecule experiments. ACS Nano. 6, 6364–6369 (2012).

    Article  CAS  Google Scholar 

  78. Guttenberg, G. et al. Inositol hexakisphosphate-dependent processing of Clostridium sordellii lethal toxin and Clostridium novyi alpha-toxin. J. Biol. Chem. 286, 14779–14786 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was partly supported by National Institute of Health grants R01AI139087, R01AI125704, R21AI123920, R21AI139690, and R21CA235533 to R.J., R01GM074830 and R01GM130144 to L.H., R01DK084509, R01AI088748, R01AI132207, and U19AI109776 to H.F, and R01 MH081923 to M.E.B. NE-CAT at the Advanced Photon Source (APS) is supported by a grant from the National Institute of General Medical Sciences (P30GM124165). The Pilatus 6 M detector on 24-ID-C beam line is funded by an NIH-ORIP HEI grant (S10 RR029205). Use of the APS, an Office of Science User Facility operated for the U.S. Department of Energy (DOE) Office of Science by Argonne National Laboratory, was supported by the U.S. DOE under Contract No. DE-AC02-06CH11357. Use of the Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, is supported by the U.S. DOE, Office of Science, Office of Basic Energy Sciences under Contract No. DE-AC02-76SF00515. The SSRL Structural Molecular Biology Program is supported by the DOE Office of Biological and Environmental Research, and by the National Institutes of Health, National Institute of General Medical Sciences (including P41GM103393). The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of NIH.

Author information

Authors and Affiliations

Authors

Contributions

P.C. and R.J. conceived the project. P.C., K.L., Z.L., B.C., and R.J. carried out the protein expression, purification, characterization, crystallization, structure determination and analysis, and all related biochemical studies. K.P. collected the X-ray diffraction data. T.M. carried out the SAXS studies. P.C., C.B.G., and L.H. performed the XL-MS. F.A.M. and M.E.B. carried out the smFRET studies. Y.Z., T.H., and H.F. provided the plasmids of TcdB and VHHs (5D, E3, 7F) and TcdB-expressing Bacillus megaterium. P.C. and R.J. wrote the manuscript with input from other authors.

Corresponding author

Correspondence to Rongsheng Jin.

Ethics declarations

Competing interests

A provisional patent application has been filed by The Regents of the University of California on the use of the structural information described in this manuscript to prevent and/or treat CDI. R.J. is a cofounder of DesignerT Inc., and H.F. is a co-founder of FZata Inc., which had no role in this study.

Additional information

Peer review information: Beth Moorefield was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Crystal structure of the TcdB–5D–E3–7F complex and the representative electron density maps.

(a) A 2Fo-Fc electron density map for the CROPs (residues G1835–E2367) contoured at 1.0 σ. (b–d) Representative 2Fo-Fc electron density maps for the newly identified SR (residues G1815–S1834) and two other regions in the CROPs (residues G1898–T1927 and G2345–E2367) contoured at 1.0 σ. The bulky residue Tyr was used as one of the markers to confirm the sequence register of the structural model. (e) An anomalous-difference electron density map generated from a crystal of the TcdB–5D–E3–7F complex soaked in tantalum bromide. The map was calculated using a 5.44 Å resolution data collected at λ = 1.2524 Å and phases derived from the final structure model. We found 10 tantalum bromide molecules in one asymmetric unit, which all bind to negatively charged glutamate or aspartate residues and thus confirmed the amino acid register of the complex. One representative tantalum bromide peak was shown in the inset. (f) Overall structure of the TcdB–5D–E3–7F complex. The color scheme for the TcdB domains are the same as that shown in Fig. 1. The three VHHs are colored purple (5D), brown (E3), and green (7F). (g) The structure of TcdB holotoxin is fitted into the negative stain EM map of TcdA that is displayed as a grey surface model (map was generously provided by Dr. Lacy D.B.) (Chumbler, N.M. et al., Nat Microbiol. 1, 15002, 2016). The CROPs of TcdA interacts with the DRBD and adopts a closed conformation.

Supplementary Figure 2 Sequence alignment in the CROPs and the hinge region.

(a) Sequence alignment of all the SRs and LRs in the CROPs of TcdB* (M68 strain), with the sequence conservation displayed on the top of the alignment. (b) Sequence alignment of the hinge region among TcdB*, TcdB (VPI 10463 strain), and TcdB2 (BI/NAP1/027 strain). (c)The hinge region (olive) directly interacts with the 3-HB (blue) and the β-flap (pink). Residues involved in the interactions are colored cyan for the hinge and green for the 3-HB and the β-flap.

Supplementary Figure 3 SEC-SAXS studies on TcdB holotoxin at pH 7.4 and 5.0.

(a, b) Rg (blue) and I(0) (orange) plots for SEC-SAXS at pH 7.4 and 5.0. (c) SAXS profile at pH 7.4 (blue) and pH 5.0 (orange) were superimposed with a curve fitting using the crystal structure of TcdB holotoxin (black). (d) Pairwise distribution function, P(r), of TcdB holotoxin at pH 7.4 (blue) and 5.0 (orange). (e) The maximum dimension of TcdB holotoxin at acidic pH and the core of TcdB composed of GTD, CPD, and DRBD.

Supplementary Figure 4 XL-MS and FRET studies of TcdB holotoxin.

(a) Crosslinking workflow used to unambiguously identify DSSO crosslinks from 3 replicates (Rep1–3) of cross-linked TcdB holotoxin. (b) MSn identification of a DSSO inter-linked peptide from TcdB: K1117–K2249. The cross-linked peptide α–β4+ (m/z 510.9914+) was detected during MS1. Next, it was selected for MS2 where it was fragmented into two characteristic fragment ion pairs, that is αAT (m/z 308.672+/704.312+) and αTA (m/z 324.652+/688.322+). Finally, these peaks were selected for MS3 analysis. αA (m/z 308.672+) identified the sequence DKAATK, in which the lysine residue at position 2 was modified with an alkene moiety. βT (m/z 704.312+) identified the sequence as YYFDEKTGIMR, in which the lysine residue at position 6 was modified with unsaturated thiol moiety. (c) Dynamic light scattering showed a monodispersed peak for TcdB holotoxin at neutral pH. (d) The distribution plot of identified linkages versus their spatial distances measured in the structure of TcdB holotoxin. (e) 5D has no effect on the fluorescence emission of the dye-labeled VHHs and the TcdB–B39–7F complex based on an ensemble FRET study. TcdB holotoxin was incubated with equimolar ratio of the Alexa-555-labeled B39, the Alexa-647-labeled 7F, or both VHHs, with or without 5D at pH 7 or pH 5. Fluorescence emissions upon excitation by 540 nm were measured. Data are the average of duplicated experiments. (f) Fluorescence intensity over time for single molecules of TcdB in complex with dye-labeled VHHs. Alexa 555, donor-labeled B39 (magenta). Alexa-647, acceptor-labeled F7 (cyan). Data was recorded at a frame rate of 10 Hz. Only molecules showing single step photobleaching to baseline for both dyes were included in the analysis. From the magnitude of the anticorrelated bleaching event, we obtained the γ-correction factor for each molecule that allows us to report the absolute FRET efficiencies. FRET was calculated for each frame until acceptor photobleaching. (g) Histogram of average FRET efficiency for each molecule at either pH 5.0 (filled circles, n = 498) or pH 7.0 (open circles, n = 594). This reports a single value for each molecule, which averages out any conformational dynamics. The mean FRET efficiency was unchanged by changing the time binning of the data. (h) Mean smFRET efficiency for TcdB in complex with dye-labeled VHHs at intermediate pH. FRET was calculated from each 100 ms frame of the movie until acceptor photobleaching, compiled into histograms and fit to a Gaussian function to obtain the FRET efficiencies. Error values represent the standard deviation from replicate measurements.

Supplementary Figure 5 VHHs 5D, 7F, and E3 recognize distinct epitopes and exploit different mechanisms to neutralize TcdB.

(a, b) pH-dependent conformational change of TcdB1072–1433 probed by ANS. TcdB1072–1433, the TcdB1072–1433–5D complex, or 5D alone was incubated with ANS in buffers with different pH, and the peak fluorescence intensities of ANS at 474 nm are shown. Error bar represents SD of three replicate experiments. (c) The structure of the TcdB1072–1433–5D complex (light blue, residues 1090–1431 of TcdB were fully resolved in this crystal structure) was superimposed to the full length TcdB–5D complex (TcdB and 5D were colored yellow and pink, respectively). The overall Cα atom r.m.s.d. is ~0.608 Å. (d) Structural superposition to compare the conformations of the pore-forming region among the TcdB1072–1433–5D complex that was crystallized at pH 8.5 (light blue), the TcdB–5D complex that was crystallized at acidic pH (yellow), and TcdA at neutral pH (orange). (e, f) A close-up view into the interface between TcdB and 5D in the structure of the TcdB1072–1433–5D complex, where the CDR2 of 5D (e), as well as its CDR1 and CDR3 (f) bind TcdB with extensive interactions. (g) 7F fixes the conformation of the Helix 1 (residues 525–539) and the Helix 2 (residues 137–158) in the GTD, which are in close proximity to the active site of the CPD and the scissile bond (L544–G545). GTD, CPD, and 7F are colored red, light blue, and green, respectively. The peptide linker between the GTD and the CPD is colored blue. A close-up view of the boxed area is shown in (h). (i) Detailed interactions between GTDVPI10463 and 7F. The interacting residues are colored green and pink for 7F and GTD, respectively. The 7F-binding interfaces are largely identical between GTDVPI10463 and GTD. (j) Interactions between GTD and E3. The residues involved in interactions are colored wheat and pink for E3 and GTD, respectively.

Supplementary Figure 6 Autoprocessing is self-inhibited in TcdB holotoxin.

(a) A close-up view of a zinc atom bound in the active site of the CPD in TcdB holotoxin. (b, c) A simplified cartoon model showing the co-localization of the β-flap, the 3-HB, and the hinge. In TcdB holotoxin, the β-flap is kept in an inactive conformation (pink). Cytosolic InsP6 triggers a ~90° rotation of the β-flap (green) to activate CPD. (d) TcdB1–1805 showed a higher autoprocessing efficiency than the full length TcdB. The autoprocessing of TcdB was examined at 37°C for 1 hour incubation in the presence of various concentrations of InsP6. The results were assessed by SDS-PAGE. 7F potently inhibited GTD cleavage. The molecular weight of TcdB holotoxin, TcdB1–1805, GTD, and 7F is 270 kDa, 205 kDa, 64 kDa, and 15.2 kDa, respectively. The gel is representative of three independent experiments.

Supplementary Figure 7 Mapping the binding sites for CSPG4, FZDs, and bezlotoxumab in TcdB holotoxin.

(a) The reported CSPG4-binding site in TcdB, involving residues 1500–1900 (Gupta, P. et al., J Biol Chem. 292, 17290-17301, 2017 and Yuan, P. et al., Cell Res. 25, 157-68, 2015), was colored blue. (b) Binding of TcdB to the immobilized biotin-labeled CRD2 was examined using a pull-down assay at neutral and acidic pH. Samples were analyzed by SDS-PAGE and Coomassie Blue staining. (c) The structure of the cysteine-rich domain of FZD2 (CRD2, green cartoon model) in complex with TcdB1285–1804 (PDB code: 6C0B) (Chen, P. et al., Science, 360, 664-669, 2018) was superimposed to TcdB holotoxin. The pore-forming region of TcdB is shown in a purple ribbon model while the rest of the toxin is shown in a surface model. (d) The structure of the CROPs I–II of TcdB in complex with bezlotoxumab Fabs (PDB code: 4NP4) (Orth, P. et al., J Biol Chem, 289, 18008-21, 2014) was superimposed to TcdB holotoxin. Bezlotoxumab Fabs were shown as cartoon models, which bind to two sites on the CROPs (blue and pink). Bezlotoxumab clashes with the GTD and the DRBD when TcdB adopts the acidic conformation. A close-up view of the boxed area is shown in (e).

Supplementary information

Supplementary Information

Supplementary Figs. 1–7, Supplementary Tables 1–3, and Supplementary Note 1

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, P., Lam, Kh., Liu, Z. et al. Structure of the full-length Clostridium difficile toxin B. Nat Struct Mol Biol 26, 712–719 (2019). https://doi.org/10.1038/s41594-019-0268-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0268-0

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