Selection and characterization of ultrahigh potency designed ankyrin repeat protein inhibitors of C. difficile toxin B

PLoS Biol. 2019 Jun 24;17(6):e3000311. doi: 10.1371/journal.pbio.3000311. eCollection 2019 Jun.

Abstract

Clostridium difficile infection (CDI) is a major nosocomial disease associated with significant morbidity and mortality. The pathology of CDI stems primarily from the 2 C. difficile-secreted exotoxins-toxin A (TcdA) and toxin B (TcdB)-that disrupt the tight junctions between epithelial cells leading to the loss of colonic epithelial barrier function. Here, we report the engineering of a series of monomeric and dimeric designed ankyrin repeat proteins (DARPins) for the neutralization of TcdB. The best dimeric DARPin, DLD-4, inhibited TcdB with a half maximal effective concentration (EC50) of 4 pM in vitro, representing an approximately 330-fold higher potency than the Food and Drug Administration (FDA)-approved anti-TcdB monoclonal antibody bezlotoxumab in the same assay. DLD-4 also protected mice from a toxin challenge in vivo. Cryo-electron microscopy (cryo-EM) studies revealed that the 2 constituent DARPins of DLD-4-1.4E and U3-bind the central and C-terminal regions of the delivery domain of TcdB. Competitive enzyme-linked immunosorbent assay (ELISA) studies showed that the DARPins 1.4E and U3 interfere with the interaction between TcdB and its receptors chondroitin sulfate proteoglycan 4 (CSPG4) and frizzled class receptor 2 (FZD2), respectively. Our cryo-EM studies revealed a new conformation of TcdB (both apo- and DARPin-bound at pH 7.4) in which the combined repetitive oligopeptides (CROPS) domain points away from the delivery domain. This conformation of the CROPS domain is in stark contrast to that seen in the negative-stain electron microscopy (EM) structure of TcdA and TcdB at the same pH, in which the CROPS domain bends toward and "kisses" the delivery domain. The ultrapotent anti-TcdB molecules from this study serve as candidate starting points for CDI drug development and provide new biological tools for studying the pathogenicity of C. difficile. The structural insights regarding both the "native" conformation of TcdB and the putative sites of TcdB interaction with the FZD2 receptor, in particular, should help accelerate the development of next-generation anti-C. difficile toxin therapeutics.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Ankyrin Repeat / genetics
  • Antibodies, Monoclonal
  • Bacterial Proteins / antagonists & inhibitors*
  • Bacterial Proteins / genetics
  • Bacterial Proteins / metabolism
  • Bacterial Proteins / physiology*
  • Bacterial Toxins / antagonists & inhibitors*
  • Bacterial Toxins / genetics
  • Bacterial Toxins / metabolism
  • Broadly Neutralizing Antibodies
  • Caco-2 Cells
  • Clostridioides difficile / metabolism
  • Clostridioides difficile / pathogenicity
  • Clostridium Infections / metabolism*
  • Cryoelectron Microscopy
  • Enterotoxins / metabolism
  • Humans
  • Mice
  • Protein Engineering / methods

Substances

  • Antibodies, Monoclonal
  • Bacterial Proteins
  • Bacterial Toxins
  • Broadly Neutralizing Antibodies
  • Enterotoxins
  • toxB protein, Clostridium difficile
  • bezlotoxumab