Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
PLoS Pathog ; 19(10): e1011496, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37871122

ABSTRACT

Clostridioides difficile is a leading cause of antibiotic-associated diarrhea and nosocomial infection in the United States. The symptoms of C. difficile infection (CDI) are associated with the production of two homologous protein toxins, TcdA and TcdB. The toxins are considered bona fide targets for clinical diagnosis as well as the development of novel prevention and therapeutic strategies. While there are extensive studies that document these efforts, there are several gaps in knowledge that could benefit from the creation of new research tools. First, we now appreciate that while TcdA sequences are conserved, TcdB sequences can vary across the span of circulating clinical isolates. An understanding of the TcdA and TcdB epitopes that drive broadly neutralizing antibody responses could advance the effort to identify safe and effective toxin-protein chimeras and fragments for vaccine development. Further, an understanding of TcdA and TcdB concentration changes in vivo can guide research into how host and microbiome-focused interventions affect the virulence potential of C. difficile. We have developed a panel of alpaca-derived nanobodies that bind specific structural and functional domains of TcdA and TcdB. We note that many of the potent neutralizers of TcdA bind epitopes within the delivery domain, a finding that could reflect roles of the delivery domain in receptor binding and/or the conserved role of pore-formation in the delivery of the toxin enzyme domains to the cytosol. In contrast, neutralizing epitopes for TcdB were found in multiple domains. The nanobodies were also used for the creation of sandwich ELISA assays that allow for quantitation of TcdA and/or TcdB in vitro and in the cecal and fecal contents of infected mice. We anticipate these reagents and assays will allow researchers to monitor the dynamics of TcdA and TcdB production over time, and the impact of various experimental interventions on toxin production in vivo.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Single-Domain Antibodies , Animals , Mice , Bacterial Toxins/genetics , Bacterial Toxins/chemistry , Enterotoxins/genetics , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Epitopes/metabolism , Bacterial Proteins/metabolism
2.
PLoS Pathog ; 18(11): e1010997, 2022 11.
Article in English | MEDLINE | ID: mdl-36409774

ABSTRACT

Human infections caused by the toxin-producing, anaerobic and spore-forming bacterium Paeniclostridium sordellii are associated with a treatment-refractory toxic shock syndrome (TSS). Reproductive-age women are at increased risk for P. sordellii infection (PSI) because this organism can cause intrauterine infection following childbirth, stillbirth, or abortion. PSI-induced TSS in this setting is nearly 100% fatal, and there are no effective treatments. TcsL, or lethal toxin, is the primary virulence factor in PSI and shares 70% sequence identity with Clostridioides difficile toxin B (TcdB). We therefore reasoned that a neutralizing monoclonal antibody (mAB) against TcdB might also provide protection against TcsL and PSI. We characterized two anti-TcdB mABs: PA41, which binds and prevents translocation of the TcdB glucosyltransferase domain into the cell, and CDB1, a biosimilar of bezlotoxumab, which prevents TcdB binding to a cell surface receptor. Both mABs could neutralize the cytotoxic activity of recombinant TcsL on Vero cells. To determine the efficacy of PA41 and CDB1 in vivo, we developed a transcervical inoculation method for modeling uterine PSI in mice. In the process, we discovered that the stage of the mouse reproductive cycle was a key variable in establishing symptoms of disease. By synchronizing the mice in diestrus with progesterone prior to transcervical inoculation with TcsL or vegetative P. sordellii, we observed highly reproducible intoxication and infection dynamics. PA41 showed efficacy in protecting against toxin in our transcervical in vivo model, but CDB1 did not. Furthermore, PA41 could provide protection following P. sordellii bacterial and spore infections, suggesting a path for further optimization and clinical translation in the effort to advance treatment options for PSI infection.


Subject(s)
Clostridium sordellii , Chlorocebus aethiops , Female , Humans , Mice , Animals , Vero Cells , Virulence Factors/metabolism , Bacterial Proteins/metabolism , Glucosyltransferases/metabolism , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/metabolism , Estrous Cycle
3.
PLoS Pathog ; 18(8): e1010720, 2022 08.
Article in English | MEDLINE | ID: mdl-35951533

ABSTRACT

Bacterial type IV secretion systems (T4SSs) are a versatile group of nanomachines that can horizontally transfer DNA through conjugation and deliver effector proteins into a wide range of target cells. The components of T4SSs in gram-negative bacteria are organized into several large subassemblies: an inner membrane complex, an outer membrane core complex, and, in some species, an extracellular pilus. Cryo-electron tomography has been used to define the structures of T4SSs in intact bacteria, and high-resolution structural models are now available for isolated core complexes from conjugation systems, the Xanthomonas citri T4SS, the Helicobacter pylori Cag T4SS, and the Legionella pneumophila Dot/Icm T4SS. In this review, we compare the molecular architectures of these T4SSs, focusing especially on the structures of core complexes. We discuss structural features that are shared by multiple T4SSs as well as evolutionary strategies used for T4SS diversification. Finally, we discuss how structural variations among T4SSs may confer specialized functional properties.


Subject(s)
Helicobacter pylori , Legionella pneumophila , Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Electron Microscope Tomography , Helicobacter pylori/metabolism , Legionella pneumophila/metabolism , Type IV Secretion Systems/genetics
4.
PLoS Pathog ; 18(2): e1010323, 2022 02.
Article in English | MEDLINE | ID: mdl-35176123

ABSTRACT

Clostridioides difficile infection (CDI) is the leading cause of nosocomial diarrhea and pseudomembranous colitis in the USA. In addition to these symptoms, patients with CDI can develop severe inflammation and tissue damage, resulting in life-threatening toxic megacolon. CDI is mediated by two large homologous protein toxins, TcdA and TcdB, that bind and hijack receptors to enter host cells where they use glucosyltransferase (GT) enzymes to inactivate Rho family GTPases. GT-dependent intoxication elicits cytopathic changes, cytokine production, and apoptosis. At higher concentrations TcdB induces GT-independent necrosis in cells and tissue by stimulating production of reactive oxygen species via recruitment of the NADPH oxidase complex. Although GT-independent necrosis has been observed in vitro, the relevance of this mechanism during CDI has remained an outstanding question in the field. In this study we generated novel C. difficile toxin mutants in the hypervirulent BI/NAP1/PCR-ribotype 027 R20291 strain to test the hypothesis that GT-independent epithelial damage occurs during CDI. Using the mouse model of CDI, we observed that epithelial damage occurs through a GT-independent process that does not involve immune cell influx. The GT-activity of either toxin was sufficient to cause severe edema and inflammation, yet GT activity of both toxins was necessary to produce severe watery diarrhea. These results demonstrate that both TcdA and TcdB contribute to disease pathogenesis when present. Further, while inactivating GT activity of C. difficile toxins may suppress diarrhea and deleterious GT-dependent immune responses, the potential of severe GT-independent epithelial damage merits consideration when developing toxin-based therapeutics against CDI.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Animals , Antibodies, Bacterial , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridium Infections/pathology , Diarrhea , Enterotoxins/metabolism , Enterotoxins/toxicity , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Humans , Inflammation , Mice , Necrosis
5.
Infect Immun ; 91(1): e0047622, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36448839

ABSTRACT

Clostridioides difficile causes antibiotic-associated diseases in humans, ranging from mild diarrhea to severe pseudomembranous colitis and death. A major clinical challenge is the prevention of disease recurrence, which affects nearly ~20 to 30% of the patients with a primary C. difficile infection (CDI). During CDI, C. difficile forms metabolically dormant spores that are essential for recurrence of CDI (R-CDI). In prior studies, we have shown that C. difficile spores interact with intestinal epithelial cells (IECs), which contribute to R-CDI. However, this interaction remains poorly understood. Here, we provide evidence that C. difficile spores interact with E-cadherin, contributing to spore adherence and internalization into IECs. C. difficile toxins TcdA and TcdB lead to adherens junctions opening and increase spore adherence to IECs. Confocal micrographs demonstrate that C. difficile spores associate with accessible E-cadherin; spore-E-cadherin association increases upon TcdA and TcdB intoxication. The presence of anti-E-cadherin antibodies decreased spore adherence and entry into IECs. By enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and immunogold labeling, we observed that E-cadherin binds to C. difficile spores, specifically to the hairlike projections of the spore, reducing spore adherence to IECs. Overall, these results expand our knowledge of how C. difficile spores bind to IECs by providing evidence that E-cadherin acts as a spore adherence receptor to IECs and by revealing how toxin-mediated damage affects spore interactions with IECs.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Humans , Adherens Junctions , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides , Spores, Bacterial , Cadherins/metabolism
6.
Proc Natl Acad Sci U S A ; 117(11): 6139-6144, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123082

ABSTRACT

Clostridioides difficile is a Gram-positive, pathogenic bacterium and a prominent cause of hospital-acquired diarrhea in the United States. The symptoms of C. difficile infection are caused by the activity of three large toxins known as toxin A (TcdA), toxin B (TcdB), and the C. difficile transferase toxin (CDT). Reported here is a 3.8-Å cryo-electron microscopy (cryo-EM) structure of CDT, a bipartite toxin comprised of the proteins CDTa and CDTb. We observe a single molecule of CDTa bound to a CDTb heptamer. The formation of the CDT complex relies on the interaction of an N-terminal adaptor and pseudoenzyme domain of CDTa with six subunits of the CDTb heptamer. CDTb is observed in a preinsertion state, a conformation observed in the transition of prepore to ß-barrel pore, although we also observe a single bound CDTa in the prepore and ß-barrel conformations of CDTb. The binding interaction appears to prime CDTa for translocation as the adaptor subdomain enters the lumen of the preinsertion state channel. These structural observations advance the understanding of how a single protein, CDTb, can mediate the delivery of a large enzyme, CDTa, into the cytosol of mammalian cells.


Subject(s)
Bacterial Toxins/metabolism , Clostridioides difficile/metabolism , Enterotoxins/metabolism , Transferases/ultrastructure , Cryoelectron Microscopy , Protein Conformation, beta-Strand , Protein Multimerization , Transferases/metabolism
7.
Proc Natl Acad Sci U S A ; 117(12): 6792-6800, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32152097

ABSTRACT

Intestinal bile acids are known to modulate the germination and growth of Clostridioides difficile Here we describe a role for intestinal bile acids in directly binding and neutralizing TcdB toxin, the primary determinant of C. difficile disease. We show that individual primary and secondary bile acids reversibly bind and inhibit TcdB to varying degrees through a mechanism that requires the combined oligopeptide repeats region to which no function has previously been ascribed. We find that bile acids induce TcdB into a compact "balled up" conformation that is no longer able to bind cell surface receptors. Lastly, through a high-throughput screen designed to identify bile acid mimetics we uncovered nonsteroidal small molecule scaffolds that bind and inhibit TcdB through a bile acid-like mechanism. In addition to suggesting a role for bile acids in C. difficile pathogenesis, these findings provide a framework for development of a mechanistic class of C. difficile antitoxins.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Bile Acids and Salts/metabolism , Clostridioides difficile/metabolism , Intestines/physiology , Receptors, Cell Surface/metabolism , Caco-2 Cells , Clostridioides difficile/growth & development , Clostridium Infections/microbiology , HCT116 Cells , Humans
8.
Proc Natl Acad Sci U S A ; 117(14): 8064-8073, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32198200

ABSTRACT

Gastrointestinal infections often induce epithelial damage that must be repaired for optimal gut function. While intestinal stem cells are critical for this regeneration process [R. C. van der Wath, B. S. Gardiner, A. W. Burgess, D. W. Smith, PLoS One 8, e73204 (2013); S. Kozar et al., Cell Stem Cell 13, 626-633 (2013)], how they are impacted by enteric infections remains poorly defined. Here, we investigate infection-mediated damage to the colonic stem cell compartment and how this affects epithelial repair and recovery from infection. Using the pathogen Clostridioides difficile, we show that infection disrupts murine intestinal cellular organization and integrity deep into the epithelium, to expose the otherwise protected stem cell compartment, in a TcdB-mediated process. Exposure and susceptibility of colonic stem cells to intoxication compromises their function during infection, which diminishes their ability to repair the injured epithelium, shown by altered stem cell signaling and a reduction in the growth of colonic organoids from stem cells isolated from infected mice. We also show, using both mouse and human colonic organoids, that TcdB from epidemic ribotype 027 strains does not require Frizzled 1/2/7 binding to elicit this dysfunctional stem cell state. This stem cell dysfunction induces a significant delay in recovery and repair of the intestinal epithelium of up to 2 wk post the infection peak. Our results uncover a mechanism by which an enteric pathogen subverts repair processes by targeting stem cells during infection and preventing epithelial regeneration, which prolongs epithelial barrier impairment and creates an environment in which disease recurrence is likely.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/pathogenicity , Clostridium Infections/pathology , Colon/pathology , Intestinal Mucosa/pathology , Stem Cells/pathology , Animals , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Cells, Cultured , Clostridioides difficile/metabolism , Clostridium Infections/microbiology , Colon/cytology , Colon/microbiology , Disease Models, Animal , Female , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Mice , Organoids , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stem Cells/microbiology
9.
Gastroenterology ; 161(1): 255-270.e4, 2021 07.
Article in English | MEDLINE | ID: mdl-33844988

ABSTRACT

BACKGROUND AND AIMS: The molecular mechanisms underlying successful fecal microbiota transplantation (FMT) for recurrent Clostridioides difficile infection (rCDI) remain poorly understood. The primary objective of this study was to characterize alterations in microRNAs (miRs) following FMT for rCDI. METHODS: Sera from 2 prospective multicenter randomized controlled trials were analyzed for miRNA levels with the use of the Nanostring nCounter platform and quantitative reverse-transcription (RT) polymerase chain reaction (PCR). In addition, rCDI-FMT and toxin-treated animals and ex vivo human colonoids were used to compare intestinal tissue and circulating miRs. miR inflammatory gene targets in colonic epithelial and peripheral blood mononuclear cells were evaluated by quantitative PCR (qPCR) and 3'UTR reporter assays. Colonic epithelial cells were used for mechanistic, cytoskeleton, cell growth, and apoptosis studies. RESULTS: miRNA profiling revealed up-regulation of 64 circulating miRs 4 and 12 weeks after FMT compared with screening, of which the top 6 were validated in the discovery cohort by means of RT-qPCR. In a murine model of relapsing-CDI, RT-qPCR analyses of sera and cecal RNA extracts demonstrated suppression of these miRs, an effect reversed by FMT. In mouse colon and human colonoids, C difficile toxin B (TcdB) mediated the suppressive effects of CDI on miRs. CDI dysregulated DROSHA, an effect reversed by FMT. Correlation analyses, qPCR ,and 3'UTR reporter assays revealed that miR-23a, miR-150, miR-26b, and miR-28 target directly the 3'UTRs of IL12B, IL18, FGF21, and TNFRSF9, respectively. miR-23a and miR-150 demonstrated cytoprotective effects against TcdB. CONCLUSIONS: These results provide novel and provocative evidence that modulation of the gut microbiome via FMT induces alterations in circulating and intestinal tissue miRs. These findings contribute to a greater understanding of the molecular mechanisms underlying FMT and identify new potential targets for therapeutic intervention in rCDI.


Subject(s)
Circulating MicroRNA/blood , Clostridium Infections/therapy , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Intestines/microbiology , Reinfection , Adult , Aged , Aged, 80 and over , Animals , Circulating MicroRNA/genetics , Clostridium Infections/blood , Clostridium Infections/genetics , Clostridium Infections/microbiology , Disease Models, Animal , Female , Humans , Male , Middle Aged , Randomized Controlled Trials as Topic , Tissue Culture Techniques , Transcriptome , Treatment Outcome
10.
Anaerobe ; 75: 102520, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35085782

ABSTRACT

We present a case of persistent bacteremia and psoas abscess from Paeniclostridium sordellii without severe symptoms or the classically associated toxic shock syndrome. Further laboratory evaluation demonstrated that the Paeniclostridium sordellii isolate lacked the lethal toxin gene and there was no cytotoxicity to exposed Vero cells.


Subject(s)
Bacteremia , Clostridium sordellii , Psoas Abscess , Shock, Septic , Animals , Bacteremia/diagnosis , Bacteremia/drug therapy , Chlorocebus aethiops , Psoas Abscess/diagnosis , Psoas Abscess/drug therapy , Shock, Septic/diagnosis , Vero Cells
11.
Infect Immun ; 89(12): e0034821, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34543122

ABSTRACT

Helicobacter pylori VacA is a secreted toxin that assembles into water-soluble oligomeric structures and forms anion-selective membrane channels. Acidification of purified VacA enhances its activity in cell culture assays. Sites of protomer-protomer contact within VacA oligomers have been identified by cryoelectron microscopy, and in the current study, we validated several of these interactions by chemical cross-linking and mass spectrometry. We then mutated amino acids at these contact sites and analyzed the effects of the alterations on VacA oligomerization and activity. VacA proteins with amino acid charge reversals at interprotomer contact sites retained the capacity to assemble into water-soluble oligomers and retained cell-vacuolating activity. Introduction of paired cysteine substitutions at these sites resulted in formation of disulfide bonds between adjacent protomers. Negative-stain electron microscopy and single-particle two-dimensional class analysis revealed that wild-type VacA oligomers disassemble when exposed to acidic pH, whereas the mutant proteins with paired cysteine substitutions retain an oligomeric state at acidic pH. Acid-activated wild-type VacA caused vacuolation of cultured cells, whereas acid-activated mutant proteins with paired cysteine substitutions lacked cell-vacuolating activity. Treatment of these mutant proteins with both low pH and a reducing agent resulted in VacA binding to cells, VacA internalization, and cell vacuolation. Internalization of a nonoligomerizing mutant form of VacA by host cells was detected without a requirement for acid activation. Collectively, these results enhance our understanding of the molecular interactions required for VacA oligomerization and support a model in which toxin activity depends on interactions of monomeric VacA with host cells.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Protein Conformation , Protein Multimerization , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship
12.
Infect Immun ; 89(4)2021 03 17.
Article in English | MEDLINE | ID: mdl-33468584

ABSTRACT

Clostridioides difficile is linked to nearly 225,000 antibiotic-associated diarrheal infections and almost 13,000 deaths per year in the United States. Pathogenic strains of C. difficile produce toxin A (TcdA) and toxin B (TcdB), which can directly kill cells and induce an inflammatory response in the colonic mucosa. Hirota et al. (S. A. Hirota et al., Infect Immun 80:4474-4484, 2012) first introduced the intrarectal instillation model of intoxication using TcdA and TcdB purified from VPI 10463 (VPI 10463 reference strain [ATCC 43255]) and 630 C. difficile strains. Here, we expand this technique by instilling purified, recombinant TcdA and TcdB, which allows for the interrogation of how specifically mutated toxins affect tissue. Mouse colons were processed and stained with hematoxylin and eosin for blinded evaluation and scoring by a board-certified gastrointestinal pathologist. The amount of TcdA or TcdB needed to produce damage was lower than previously reported in vivo and ex vivo Furthermore, TcdB mutants lacking either endosomal pore formation or glucosyltransferase activity resemble sham negative controls. Immunofluorescent staining revealed how TcdB initially damages colonic tissue by altering the epithelial architecture closest to the lumen. Tissue sections were also immunostained for markers of acute inflammatory infiltration. These staining patterns were compared to slides from a human C. difficile infection (CDI). The intrarectal instillation mouse model with purified recombinant TcdA and/or TcdB provides the flexibility needed to better understand structure/function relationships across different stages of CDI pathogenesis.


Subject(s)
Clostridioides difficile/pathogenicity , Disease Susceptibility , Enterocolitis, Pseudomembranous/microbiology , Enterotoxins/administration & dosage , Recombinant Proteins/administration & dosage , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Colon , Disease Models, Animal , Enterotoxins/genetics , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Mice , Mutant Proteins
13.
J Pediatr Gastroenterol Nutr ; 72(3): 378-383, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32925555

ABSTRACT

OBJECTIVES: Recent Infectious Disease Society of America guidelines recommend multistep testing algorithms to diagnose Clostridioides difficile infection (CDI), including a combination of nucleic acid amplification-based testing (NAAT) and toxin enzyme immunoassay (EIA). The use of these algorithms in children, including the ability to differentiate between C. difficile colonization and CDI, however, has not been evaluated. METHODS: We prospectively enrolled asymptomatic pediatric patients with cancer, cystic fibrosis (CF), or inflammatory bowel disease (IBD) and obtained a stool sample for NAAT testing. If positive by NAAT (colonized), EIA was performed. In addition, children with symptomatic CDI who tested positive by NAAT via the clinical laboratory were enrolled, and EIA was performed on residual stool. A functional cell cytotoxicity neutralization assay (CCNA) was also applied to stool samples from both the colonized and symptomatic cohorts. RESULTS: Of the 225 asymptomatic children enrolled in the study, 47 (21%) were colonized with C. difficile including 9/59 (15.5%) with cancer, 30/92 (32.6%) with CF, and 8/74 (10.8%) with IBD. An additional 41 children with symptomatic CDI were enrolled. When symptomatic and colonized children were compared, neither EIA positivity (44% vs 26%, P = 0.07) nor CCNA positivity (49% vs 45%, P = 0.70) differed significantly or were able to predict disease severity in the symptomatic cohort. CONCLUSIONS: Use of a multistep testing algorithm with NAAT followed by EIA failed to differentiate symptomatic CDI from asymptomatic colonization in our pediatric cohort. As multistep algorithms are moved into clinical care, the pediatric provider will need to be aware of their limitations.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Child , Clostridioides , Clostridium Infections/diagnosis , Feces , Humans , Immunoenzyme Techniques
14.
Nature ; 522(7555): 173-8, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26040716

ABSTRACT

Stem cells of the gastrointestinal tract, pancreas, liver and other columnar epithelia collectively resist cloning in their elemental states. Here we demonstrate the cloning and propagation of highly clonogenic, 'ground state' stem cells of the human intestine and colon. We show that derived stem-cell pedigrees sustain limited copy number and sequence variation despite extensive serial passaging and display exquisitely precise, cell-autonomous commitment to epithelial differentiation consistent with their origins along the intestinal tract. This developmentally patterned and epigenetically maintained commitment of stem cells is likely to enforce the functional specificity of the adult intestinal tract. Using clonally derived colonic epithelia, we show that toxins A or B of the enteric pathogen Clostridium difficile recapitulate the salient features of pseudomembranous colitis. The stability of the epigenetic commitment programs of these stem cells, coupled with their unlimited replicative expansion and maintained clonogenicity, suggests certain advantages for their use in disease modelling and regenerative medicine.


Subject(s)
Intestines/cytology , Stem Cells/cytology , Stem Cells/metabolism , Bacterial Toxins/pharmacology , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Clone Cells/cytology , Clone Cells/metabolism , Clostridioides difficile/physiology , Colon/cytology , Colon/drug effects , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Epigenesis, Genetic/genetics , Epithelium/drug effects , Epithelium/metabolism , Fetus/cytology , Genomic Instability/genetics , Humans , Intestine, Small/cytology , Intestines/drug effects , Organoids/cytology , Organoids/growth & development
15.
Proc Natl Acad Sci U S A ; 114(32): E6652-E6659, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739897

ABSTRACT

Gram-positive bacteria cause the majority of skin and soft tissue infections (SSTIs), resulting in the most common reason for clinic visits in the United States. Recently, it was discovered that Gram-positive pathogens use a unique heme biosynthesis pathway, which implicates this pathway as a target for development of antibacterial therapies. We report here the identification of a small-molecule activator of coproporphyrinogen oxidase (CgoX) from Gram-positive bacteria, an enzyme essential for heme biosynthesis. Activation of CgoX induces accumulation of coproporphyrin III and leads to photosensitization of Gram-positive pathogens. In combination with light, CgoX activation reduces bacterial burden in murine models of SSTI. Thus, small-molecule activation of CgoX represents an effective strategy for the development of light-based antimicrobial therapies.


Subject(s)
Bacterial Proteins/metabolism , Coproporphyrinogen Oxidase/metabolism , Coproporphyrins/biosynthesis , Photosensitizing Agents/metabolism , Phototherapy , Staphylococcal Skin Infections/enzymology , Staphylococcal Skin Infections/therapy , Staphylococcus aureus/metabolism , Animals , Bacterial Proteins/genetics , Coproporphyrinogen Oxidase/genetics , Coproporphyrins/genetics , Disease Models, Animal , Mice , Staphylococcus aureus/genetics
16.
J Biol Chem ; 293(3): 941-952, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29180448

ABSTRACT

Clostridium difficile infection is the leading cause of hospital-acquired diarrhea and is mediated by the actions of two toxins, TcdA and TcdB. The toxins perturb host cell function through a multistep process of receptor binding, endocytosis, low pH-induced pore formation, and the translocation and delivery of an N-terminal glucosyltransferase domain that inactivates host GTPases. Infection studies with isogenic strains having defined toxin deletions have established TcdB as an important target for therapeutic development. Monoclonal antibodies that neutralize TcdB function have been shown to protect against C. difficile infection in animal models and reduce recurrence in humans. Here, we report the mechanism of TcdB neutralization by PA41, a humanized monoclonal antibody capable of neutralizing TcdB from a diverse array of C. difficile strains. Through a combination of structural, biochemical, and cell functional studies, involving X-ray crystallography and EM, we show that PA41 recognizes a single, highly conserved epitope on the TcdB glucosyltransferase domain and blocks productive translocation and delivery of the enzymatic cargo into the host cell. Our study reveals a unique mechanism of C. difficile toxin neutralization by a monoclonal antibody, which involves targeting a process that is conserved across the large clostridial glucosylating toxins. The PA41 antibody described here provides a valuable tool for dissecting the mechanism of toxin pore formation and translocation across the endosomal membrane.


Subject(s)
Antibodies, Neutralizing/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/metabolism , Enterotoxins/metabolism , Antibodies, Monoclonal/metabolism , Bacterial Toxins/chemistry , Caco-2 Cells , Clostridioides difficile/enzymology , Crystallography, X-Ray , Cytosol/metabolism , Enterotoxins/chemistry , Humans , Hydrogen-Ion Concentration , Microscopy, Electron , Rubidium/chemistry , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/metabolism
17.
J Biol Chem ; 292(35): 14401-14412, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28705932

ABSTRACT

Clostridium difficile is a clinically significant pathogen that causes mild-to-severe (and often recurrent) colon infections. Disease symptoms stem from the activities of two large, multidomain toxins known as TcdA and TcdB. The toxins can bind, enter, and perturb host cell function through a multistep mechanism of receptor binding, endocytosis, pore formation, autoproteolysis, and glucosyltransferase-mediated modification of host substrates. Monoclonal antibodies that neutralize toxin activity provide a survival benefit in preclinical animal models and prevent recurrent infections in human clinical trials. However, the molecular mechanisms involved in these neutralizing activities are unclear. To this end, we performed structural studies on a neutralizing monoclonal antibody, PA50, a humanized mAb with both potent and broad-spectrum neutralizing activity, in complex with TcdA. Electron microscopy imaging and multiangle light-scattering analysis revealed that PA50 binds multiple sites on the TcdA C-terminal combined repetitive oligopeptides (CROPs) domain. A crystal structure of two PA50 Fabs bound to a segment of the TcdA CROPs helped define a conserved epitope that is distinct from previously identified carbohydrate-binding sites. Binding of TcdA to the host cell surface was directly blocked by either PA50 mAb or Fab and suggested that receptor blockade is the mechanism by which PA50 neutralizes TcdA. These findings highlight the importance of the CROPs C terminus in cell-surface binding and a role for neutralizing antibodies in defining structural features critical to a pathogen's mechanism of action. We conclude that PA50 protects host cells by blocking the binding of TcdA to cell surfaces.


Subject(s)
Anti-Bacterial Agents/metabolism , Antibodies, Neutralizing/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/enzymology , Enterocytes/metabolism , Enterotoxins/metabolism , Glucosyltransferases/metabolism , Models, Molecular , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Neutralizing/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Binding Sites, Antibody , Caco-2 Cells , Conserved Sequence , Crystallography, X-Ray , Enterocytes/drug effects , Enterotoxins/chemistry , Enterotoxins/genetics , Enterotoxins/toxicity , Epitope Mapping , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Glucosyltransferases/toxicity , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Repetitive Sequences, Amino Acid
18.
J Biol Chem ; 292(42): 17290-17301, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28842504

ABSTRACT

Clostridium difficile is a major nosocomial pathogen that produces two exotoxins, TcdA and TcdB, with TcdB thought to be the primary determinant in human disease. TcdA and TcdB are large, multidomain proteins, each harboring a cytotoxic glucosyltransferase domain that is delivered into the cytosol from endosomes via a translocation domain after receptor-mediated endocytosis of toxins from the cell surface. Although there are currently no known host cell receptors for TcdA, three cell-surface receptors for TcdB have been identified: CSPG4, NECTIN3, and FZD1/2/7. The sites on TcdB that mediate binding to each receptor are not defined. Furthermore, it is not known whether the combined repetitive oligopeptide (CROP) domain is involved in or required for receptor binding. Here, in a screen designed to identify sites in TcdB that are essential for target cell intoxication, we identified a region at the junction of the translocation and the CROP domains that is implicated in CSPG4 binding. Using a series of C-terminal truncations, we show that the CSPG4-binding site on TcdB extends into the CROP domain, requiring three short repeats for binding and for full toxicity on CSPG4-expressing cells. Consistent with the location of the CSPG4-binding site on TcdB, we show that the anti-TcdB antibody bezlotoxumab, which binds partially within the first three short repeats, prevents CSPG4 binding to TcdB. In addition to establishing the binding region for CSPG4, this work ascribes for the first time a role in TcdB CROPs in receptor binding and further clarifies the relative roles of host receptors in TcdB pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Clostridioides difficile/enzymology , Glucosyltransferases/metabolism , Membrane Proteins/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/genetics , Broadly Neutralizing Antibodies , CHO Cells , Caco-2 Cells , Chlorocebus aethiops , Chondroitin Sulfate Proteoglycans/genetics , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Cricetinae , Cricetulus , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Protein Binding , Protein Domains
19.
PLoS Pathog ; 12(12): e1006070, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27942025

ABSTRACT

Clostridium difficile infection affects a significant number of hospitalized patients in the United States. Two homologous exotoxins, TcdA and TcdB, are the major virulence factors in C. difficile pathogenesis. The toxins are glucosyltransferases that inactivate Rho family-GTPases to disrupt host cellular function and cause fluid secretion, inflammation, and cell death. Toxicity depends on receptor binding and subsequent endocytosis. TcdB has been shown to enter cells by clathrin-dependent endocytosis, but the mechanism of TcdA uptake is still unclear. Here, we utilize a combination of RNAi-based knockdown, pharmacological inhibition, and cell imaging approaches to investigate the endocytic mechanism(s) that contribute to TcdA uptake and subsequent cytopathic and cytotoxic effects. We show that TcdA uptake and cellular intoxication is dynamin-dependent but does not involve clathrin- or caveolae-mediated endocytosis. Confocal microscopy using fluorescently labeled TcdA shows significant colocalization of the toxin with PACSIN2-positive structures in cells during entry. Disruption of PACSIN2 function by RNAi-based knockdown approaches inhibits TcdA uptake and toxin-induced downstream effects in cells indicating that TcdA entry is PACSIN2-dependent. We conclude that TcdA and TcdB utilize distinct endocytic mechanisms to intoxicate host cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bacterial Toxins/metabolism , Clostridium Infections/metabolism , Endocytosis/physiology , Enterotoxins/metabolism , Animals , Blotting, Western , Caco-2 Cells , Clathrin , Clostridioides difficile , Fluorescent Antibody Technique , Gene Knockdown Techniques , HEK293 Cells , Humans , Image Processing, Computer-Assisted , Mice , Microscopy, Confocal , Protein Transport/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Virulence Factors/metabolism
20.
EMBO Rep ; 17(3): 428-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26882549

ABSTRACT

Staphylococcus aureus (S. aureus) is a human pathogen that relies on the subversion of host phagocytes to support its pathogenic lifestyle. S. aureus strains can produce up to five beta-barrel, bi-component, pore-forming leukocidins that target and kill host phagocytes. Thus, preventing immune cell killing by these toxins is likely to boost host immunity. Here, we describe the identification of glycine-rich motifs within the membrane-penetrating stem domains of the leukocidin subunits that are critical for killing primary human neutrophils. Remarkably, leukocidins lacking these glycine-rich motifs exhibit dominant-negative inhibitory effects toward their wild-type toxin counterparts as well as other leukocidins. Biochemical and cellular assays revealed that these dominant-negative toxins work by forming mixed complexes that are impaired in pore formation. The dominant-negative leukocidins inhibited S. aureus cytotoxicity toward primary human neutrophils, protected mice from lethal challenge by wild-type leukocidin, and reduced bacterial burden in a murine model of bloodstream infection. Thus, we describe the first example of staphylococcal bi-component dominant-negative toxins and their potential as novel therapeutics to combat S. aureus infection.


Subject(s)
Cytotoxins/genetics , Leukocidins/genetics , Mutation , Staphylococcal Infections/prevention & control , Staphylococcus aureus/pathogenicity , Animals , Cytotoxins/chemistry , Cytotoxins/metabolism , Cytotoxins/therapeutic use , Female , Glycine/chemistry , Glycine/genetics , Humans , Leukocidins/chemistry , Leukocidins/metabolism , Leukocidins/therapeutic use , Mice , Neutrophils/microbiology , Phagocytes/microbiology , Protein Domains , Protein Multimerization , Staphylococcus aureus/genetics , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL