101
|
Translocation of Clostridium difficile toxin B across polarized Caco-2 cell monolayers is enhanced by toxin A. Can J Infect Dis 2011; 15:83-8. [PMID: 18159481 DOI: 10.1155/2004/292580] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2003] [Accepted: 10/10/2003] [Indexed: 01/25/2023] Open
Abstract
Clostridium difficile is the etiological agent of antibiotic-associated diarrhea; the most common form of nosocomial infectious diarrhea. The basis for the shock-like systemic symptoms observed in severe cases of this infection are not known. It is hypothesized that the invasion of C difficile toxins A and/or B from the gut mucosa may contribute to these symptoms.A polarized tissue culture model employing Caco-2 cells grown on transwell inserts was established to study the translocation of purified C difficile toxins A and B. C difficile toxins were (125)I labelled and inoculated onto confluent polarized Caco-2 cell monolayers to study translocation dynamics. Electrical resistance measurements were used to monitor monolayer confluence and tight junction integrity. Samples were taken from the apical and basal sides of the insert, as well as the insert itself, and tested using the human foreskin fibroblasts cell cytotoxicity assay to monitor partitioning of the radiolabelled toxins. Toxin A produced a 50% reduction in electrical resistance in 3 h whereas the same concentration of toxin B required at least 7 h to achieve the same effect. Both toxins A and B were able to translocate across confluent monolayers of Caco-2 cells. The combination of toxin A and B together was synergistic with respect to promoting the translocation of toxin B. Although the addition of toxin A resulted in a 100% increase in the amount of toxin B able to translocate, no increases in toxin A translocation were observed. These findings suggest a model of pathogenesis in which C difficile toxin A facilitates the translocation of toxin B from the gut into submucosal areas where it may play a role in inflammatory damage.
Collapse
|
102
|
Molecular Diagnosis of Gastrointestinal Infections. Mol Microbiol 2011. [DOI: 10.1128/9781555816834.ch48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
103
|
Super toxins from a super bug: structure and function of Clostridium difficile toxins. Biochem J 2011; 436:517-26. [DOI: 10.1042/bj20110106] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Clostridium difficile, a highly infectious bacterium, is the leading cause of antibiotic-associated pseudomembranous colitis. In 2009, the number of death certificates mentioning C. difficile infection in the U.K. was estimated at 3933 with 44% of certificates recording infection as the underlying cause of death. A number of virulence factors facilitate its pathogenicity, among which are two potent exotoxins; Toxins A and B. Both are large monoglucosyltransferases that catalyse the glucosylation, and hence inactivation, of Rho-GTPases (small regulatory proteins of the eukaryote actin cell cytoskeleton), leading to disorganization of the cytoskeleton and cell death. The roles of Toxins A and B in the context of C. difficile infection is unknown. In addition to these exotoxins, some strains of C. difficile produce an unrelated ADP-ribosylating binary toxin. This toxin consists of two independently produced components: an enzymatic component (CDTa) and the other, the transport component (CDTb) which facilitates translocation of CDTa into target cells. CDTa irreversibly ADP-ribosylates G-actin in target cells, which disrupts the F-actin:G-actin equilibrium leading to cell rounding and cell death. In the present review we provide a summary of the current structural understanding of these toxins and discuss how it may be used to identify potential targets for specific drug design.
Collapse
|
104
|
Immunization with Bacillus spores expressing toxin A peptide repeats protects against infection with Clostridium difficile strains producing toxins A and B. Infect Immun 2011; 79:2295-302. [PMID: 21482682 DOI: 10.1128/iai.00130-11] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Clostridium difficile is a leading cause of nosocomial infection in the developed world. Two toxins, A and B, produced by most strains of C. difficile are implicated as virulence factors, yet only recently has the requirement of these for infection been investigated by genetic manipulation. Current vaccine strategies are focused mostly on parenteral delivery of toxoids. In this work, we have used bacterial spores (Bacillus subtilis) as a delivery vehicle to evaluate the carboxy-terminal repeat domains of toxins A and B as protective antigens. Our findings are important and show that oral immunization of the repeat domain of toxin A is sufficient to confer protection in a hamster model of infection designed to closely mimic the human course of infection. Importantly, neutralizing antibodies to the toxin A repeat domain were shown to be cross-reactive with the analogous domain of toxin B and, being of high avidity, provided protection against challenge with a C. difficile strain producing toxins A and B (A(+)B(+)). Thus, although many strains produce both toxins, antibodies to only toxin A can mediate protection. Animals vaccinated with recombinant spores were fully able to survive reinfection, a property that is particularly important for a disease with which patients are prone to relapse. We show that mucosal immunization, not parenteral delivery, is required to generate secretory IgA and that production of these neutralizing polymeric antibodies correlates with protection. This work demonstrates that an effective vaccine against C. difficile can be designed around two attributes, mucosal delivery and the repeat domain of toxin A.
Collapse
|
105
|
Vohra P, Poxton IR. Efficacy of decontaminants and disinfectants against Clostridium difficile. J Med Microbiol 2011; 60:1218-1224. [PMID: 21474613 DOI: 10.1099/jmm.0.030288-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Clostridium difficile is a common nosocomial pathogen transmitted mainly via its spores. These spores can remain viable on contaminated surfaces for several months and are resistant to most commonly used cleaning agents. Thus, effective decontamination of the environment is essential in preventing the transmission of C. difficile in health-care establishments. However, this emphasis on decontamination must also be extended to laboratories due to risk of exposure of staff to potentially virulent strains. Though few cases of laboratory-acquired infection have been reported, the threat of infection by C. difficile in the laboratory is real. Our aim was to test the efficacy of four disinfectants, Actichlor, MicroSol 3+, TriGene Advance and Virkon, and one laboratory decontaminant, Decon 90, against vegetative cells and spores of C. difficile. Five strains were selected for the study: the three most commonly encountered epidemic strains in Scotland, PCR ribotypes 106, 001 and 027, and control strains 630 and VPI 10463. MICs were determined by agar dilution and broth microdilution. All the agents tested inhibited the growth of vegetative cells of the selected strains at concentrations below the recommended working concentrations. Additionally, their effect on spores was determined by exposing the spores of these strains to different concentrations of the agents for different periods of time. For some of the agents, an exposure of 10 min was required for sporicidal activity. Further, only Actichlor was able to bring about a 3 log(10) reduction in spore numbers under clean and dirty conditions. It was also the only agent that decontaminated different hard, non-porous surfaces artificially contaminated with C. difficile spores. However, this too required an exposure time of more than 2 min and up to 10 min. In conclusion, only the chlorine-releasing agent Actichlor was found to be suitable for the elimination of C. difficile spores from the environment, making it the agent of choice for the decontamination of laboratory surfaces.
Collapse
Affiliation(s)
- Prerna Vohra
- Centre for Infectious Diseases, University of Edinburgh College of Medicine and Veterinary Medicine, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Ian R Poxton
- Centre for Infectious Diseases, University of Edinburgh College of Medicine and Veterinary Medicine, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
106
|
Ballard TE, Wang X, Olekhnovich I, Koerner T, Seymour C, Salamoun J, Warthan M, Hoffman PS, Macdonald TL. Synthesis and antimicrobial evaluation of nitazoxanide-based analogues: identification of selective and broad spectrum activity. ChemMedChem 2011; 6:362-77. [PMID: 21275058 PMCID: PMC3089805 DOI: 10.1002/cmdc.201000475] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Indexed: 01/20/2023]
Abstract
A library composed of nitazoxanide-based analogues was synthesized and assayed for increased antibacterial efficacy against the pyruvate-ferredoxin oxidoreductase (PFOR) using microorganisms Helicobacter pylori, Campylobacter jejuni and Clostridium difficile. Derivatives were found to recapitulate and improve activity against these organisms and select analogues were tested for their ability to disrupt the PFOR enzyme directly. The library was also screened for activity against staphylococci and resulted in the identification of analogues capable of inhibiting both staphylococci and all PFOR organisms at low micromolar minimum inhibitory concentrations with low toxicity to human foreskin cells.
Collapse
Affiliation(s)
- T Eric Ballard
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904-4319, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Adenosine deaminase inhibition prevents Clostridium difficile toxin A-induced enteritis in mice. Infect Immun 2010; 79:653-62. [PMID: 21115723 DOI: 10.1128/iai.01159-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Toxin A (TxA) is able to induce most of the classical features of Clostridium difficile-associated disease in animal models. The objective of this study was to determine the effect of an inhibitor of adenosine deaminase, EHNA [erythro-9-(2-hydroxy-3-nonyl)-adenine], on TxA-induced enteritis in C57BL6 mice and on the gene expression of adenosine receptors. EHNA (90 μmol/kg) or phosphate-buffered saline (PBS) was injected intraperitoneally (i.p.) 30 min prior to TxA (50 μg) or PBS injection into the ileal loop. A(2A) adenosine receptor agonist (ATL313; 5 nM) was injected in the ileal loop immediately before TxA (50 μg) in mice pretreated with EHNA. The animals were euthanized 3 h later. The changes in the tissue were assessed by the evaluation of ileal loop weight/length and secretion volume/length ratios, histological analysis, myeloperoxidase assay (MPO), the local expression of inducible nitric oxide synthase (NOS2), pentraxin 3 (PTX3), NF-κB, tumor necrosis factor alpha (TNF-α), and interleukin-1β (IL-1β) by immunohistochemistry and/or quantitative reverse transcription-PCR (qRT-PCR). The gene expression profiles of A₁, A(2A), A(2B), and A₃ adenosine receptors also were evaluated by qRT-PCR. Adenosine deaminase inhibition, by EHNA, reduced tissue injury, neutrophil infiltration, and the levels of proinflammatory cytokines (TNF-α and IL-1β) as well as the expression of NOS2, NF-κB, and PTX3 in the ileum of mice injected with TxA. ATL313 had no additional effect on EHNA action. TxA increased the gene expression of A₁ and A(2A) adenosine receptors. Our findings show that the inhibition of adenosine deaminase by EHNA can prevent Clostridium difficile TxA-induced damage and inflammation possibly through the A(2A) adenosine receptor, suggesting that the modulation of adenosine/adenosine deaminase represents an important tool in the management of C. difficile-induced disease.
Collapse
|
108
|
Corthésy B. Role of secretory immunoglobulin A and secretory component in the protection of mucosal surfaces. Future Microbiol 2010; 5:817-29. [PMID: 20441552 DOI: 10.2217/fmb.10.39] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The contribution of secretory immunoglobulin A (SIgA) antibodies in the defense of mucosal epithelia plays an important role in preventing pathogen adhesion to host cells, therefore blocking dissemination and further infection. This mechanism, referred to as immune exclusion, represents the dominant mode of action of the antibody. However, SIgA antibodies combine multiple facets, which together confer properties extending from intracellular and serosal neutralization of antigens, activation of non-inflammatory pathways and homeostatic control of the endogenous microbiota. The sum of these features suggests that future opportunities for translational application from research-based knowledge to clinics include the mucosal delivery of bioactive antibodies capable of preserving immunoreactivity in the lung, gastrointestinal tract, the genito-urinary tract for the treatment of infections. This article covers topics dealing with the structure of SIgA, the dissection of its mode of action in epithelia lining different mucosal surfaces and its potential in immunotherapy against infectious pathogens.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory of the Department of Immunology & Allergy, University State Hospital (CHUV), Rue du Bugnon 46, 1011 Lausanne, Switzerland.
| |
Collapse
|
109
|
Sun X, Savidge T, Feng H. The enterotoxicity of Clostridium difficile toxins. Toxins (Basel) 2010; 2:1848-80. [PMID: 22069662 PMCID: PMC3153265 DOI: 10.3390/toxins2071848] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 06/23/2010] [Accepted: 07/09/2010] [Indexed: 02/06/2023] Open
Abstract
The major virulence factors of Clostridium difficile infection (CDI) are two large exotoxins A (TcdA) and B (TcdB). However, our understanding of the specific roles of these toxins in CDI is still evolving. It is now accepted that both toxins are enterotoxic and proinflammatory in the human intestine. Both purified TcdA and TcdB are capable of inducing the pathophysiology of CDI, although most studies have focused on TcdA. C. difficile toxins exert a wide array of biological activities by acting directly on intestinal epithelial cells. Alternatively, the toxins may target immune cells and neurons once the intestinal epithelial barrier is disrupted. The toxins may also act indirectly by stimulating cells to produce chemokines, proinflammatory cytokines, neuropeptides and other neuroimmune signals. This review considers the mechanisms of TcdA- and TcdB-induced enterotoxicity, and recent developments in this field.
Collapse
Affiliation(s)
- Xingmin Sun
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA;
| | - Tor Savidge
- The University of Texas Medical Branch, Galveston, TX, 77555, USA;
| | - Hanping Feng
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA;
- Author to whom correspondence should be addressed; ; Tel.: +1-508-887-4252; Fax: +1-508-839-7911
| |
Collapse
|
110
|
Structural organization of the functional domains of Clostridium difficile toxins A and B. Proc Natl Acad Sci U S A 2010; 107:13467-72. [PMID: 20624955 DOI: 10.1073/pnas.1002199107] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Clostridium difficile toxins A and B are members of an important class of virulence factors known as large clostridial toxins (LCTs). Toxin action involves four major steps: receptor-mediated endocytosis, translocation of a catalytic glucosyltransferase domain across the membrane, release of the enzymatic moiety by autoproteolytic processing, and a glucosyltransferase-dependent inactivation of Rho family proteins. We have imaged toxin A (TcdA) and toxin B (TcdB) holotoxins by negative stain electron microscopy to show that these molecules are similar in structure. We then determined a 3D structure for TcdA and mapped the organization of its functional domains. The molecule has a "pincher-like" head corresponding to the delivery domain and two tails, long and short, corresponding to the receptor-binding and glucosyltransferase domains, respectively. A second structure, obtained at the acidic pH of an endosome, reveals a significant structural change in the delivery and glucosyltransferase domains, and thus provides a framework for understanding the molecular mechanism of LCT cellular intoxication.
Collapse
|
111
|
Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea. J Clin Gastroenterol 2010; 44:354-60. [PMID: 20048681 DOI: 10.1097/mcg.0b013e3181c87e02] [Citation(s) in RCA: 481] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Clostridium difficile-associated disease (CDAD) is the major known cause of antibiotic-induced diarrhea and colitis, and the disease is thought to result from persistent disruption of commensal gut microbiota. Bacteriotherapy by way of fecal transplantation can be used to treat recurrent CDAD, which is thought to reestablish the normal colonic microflora. However, limitations of conventional microbiologic techniques have, until recently, precluded testing of this idea. In this study, we used terminal-restriction fragment length polymorphism and 16S rRNA gene sequencing approaches to characterize the bacterial composition of the colonic microflora in a patient suffering from recurrent CDAD before and after treatment by fecal transplantation from a healthy donor. Although the patient's residual colonic microbiota, prior to therapy was deficient in members of the bacterial divisions-Firmicutes and Bacteriodetes, transplantation had a dramatic impact on the composition of the patient's gut microbiota. By 14 days posttransplantation, the fecal bacterial composition of the recipient was highly similar to that of the donor and was dominated by Bacteroides spp. strains and an uncharacterized butyrate producing bacterium. The change in bacterial composition was accompanied by resolution of the patient's symptoms. The striking similarity of the recipient's and donor's intestinal microbiota following after bacteriotherapy suggests that the donor's bacteria quickly occupied their requisite niches resulting in restoration of both the structure and function of the microbial communities present.
Collapse
|
112
|
Co-culture with potentially probiotic microorganisms antagonises virulence factors of Clostridium difficile in vitro. Antonie van Leeuwenhoek 2010; 98:19-29. [DOI: 10.1007/s10482-010-9424-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 03/03/2010] [Indexed: 10/19/2022]
|
113
|
Sundriyal A, Roberts AK, Shone CC, Acharya KR. Structural basis for substrate recognition in the enzymatic component of ADP-ribosyltransferase toxin CDTa from Clostridium difficile. J Biol Chem 2009; 284:28713-9. [PMID: 19692332 DOI: 10.1074/jbc.m109.043018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
ADP-ribosylation is one of the favored modes of cell intoxication employed by several bacteria. Clostridium difficile is recognized to be an important nosocomial pathogen associated with considerable morbidity and attributable mortality. Along with its two well known toxins, Toxin A and Toxin B, it produces an ADP-ribosylating toxin that targets monomeric actin of the target cell. Like other Clostridial actin ADP-ribosylating toxins, this binary toxin, known as C. difficile toxin (CDT), is composed of two subunits, CDTa and CDTb. In this study, we present high resolution crystal structures of CDTa in its native form (at pH 4.0, 8.5, and 9.0) and in complex with ADP-ribose donors, NAD and NADPH (at pH 9.0). The crystal structures of the native protein show "pronounced conformational flexibility" confined to the active site region of the protein and "enhanced" disorder at low pH, whereas the complex structures highlight significant differences in "ligand specificity" compared with the enzymatic subunit of a close homologue, Clostridium perfringens iota toxin. Specifically in CDTa, two of the suggested catalytically important residues (Glu-385 and Glu-387) seem to play no role or a less important role in ligand binding. These structural data provide the first detailed information on protein-donor substrate complex stabilization in CDTa, which may have implications in understanding CDT recognition.
Collapse
Affiliation(s)
- Amit Sundriyal
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | | | | | | |
Collapse
|
114
|
Seddon SV, Krishna M, Davies HA, Borriello SP. Effect of Nutrition on the Expression of Known and Putative Virulence Factors ofClostridium difficile. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609109140280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | - H. A. Davies
- Electron Microscopy Research Group, Clinical Research Centre, Watford Road, Harrow, Middlesex, HA1 3 UJ, UK
| | | |
Collapse
|
115
|
Gomez-Trevino M, Boureau H, Karjalainen T, Bourlioux P. Clostridium difficileAdherence to Mucus: Results of anin vivoandex vivoAssay. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609609166474] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- M. Gomez-Trevino
- Laboratoire de Microbiologie, Centre d'Etudes Pharmaceutiques, 5 rue Jean-Baptiste-Clément, 92296, Chaˇtenay-Malabry Cedex, France
| | - H. Boureau
- Laboratoire de Microbiologie, Centre d'Etudes Pharmaceutiques, 5 rue Jean-Baptiste-Clément, 92296, Chaˇtenay-Malabry Cedex, France
| | - T. Karjalainen
- Laboratoire de Microbiologie, Centre d'Etudes Pharmaceutiques, 5 rue Jean-Baptiste-Clément, 92296, Chaˇtenay-Malabry Cedex, France
| | - P. Bourlioux
- Laboratoire de Microbiologie, Centre d'Etudes Pharmaceutiques, 5 rue Jean-Baptiste-Clément, 92296, Chaˇtenay-Malabry Cedex, France
| |
Collapse
|
116
|
Barc MC, Depitre C, Corthier G, Karjalainen T, Bourlioux P. Barrier Effect of Normal Microbiota AgainstClostridium difficilemay be Influenced by Drugs Devoid of Antibiotic Activity. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609409141370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- M.-C. Barc
- Laboratoire de Microbiologie, Faculté de Pharmacie, Centre d'Etudes Pharmaceutiques, 92296, Chaˇtenay-Malabry Cedex, France
| | - C. Depitre
- Laboratoire de Microbiologie, Faculté de Pharmacie, Centre d'Etudes Pharmaceutiques, 92296, Chaˇtenay-Malabry Cedex, France
- Laboratoire d'Ecologie et de Physiologie du Système Digestif, LEPSD-INRA, 78352, Jouy-en-Josas, France
| | - G. Corthier
- Laboratoire d'Ecologie et de Physiologie du Système Digestif, LEPSD-INRA, 78352, Jouy-en-Josas, France
| | - T. Karjalainen
- Laboratoire de Microbiologie, Faculté de Pharmacie, Centre d'Etudes Pharmaceutiques, 92296, Chaˇtenay-Malabry Cedex, France
| | - P. Bourlioux
- Laboratoire de Microbiologie, Faculté de Pharmacie, Centre d'Etudes Pharmaceutiques, 92296, Chaˇtenay-Malabry Cedex, France
| |
Collapse
|
117
|
Pruitt RN, Chagot B, Cover M, Chazin WJ, Spiller B, Lacy DB. Structure-function analysis of inositol hexakisphosphate-induced autoprocessing in Clostridium difficile toxin A. J Biol Chem 2009; 284:21934-21940. [PMID: 19553670 DOI: 10.1074/jbc.m109.018929] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The action of Clostridium difficile toxins A and B depends on inactivation of host small G-proteins by glucosylation. Cellular inositol hexakisphosphate (InsP6) induces an autocatalytic cleavage of the toxins, releasing an N-terminal glucosyltransferase domain into the host cell cytosol. We have defined the cysteine protease domain (CPD) responsible for autoprocessing within toxin A (TcdA) and report the 1.6 A x-ray crystal structure of the domain bound to InsP6. InsP6 is bound in a highly basic pocket that is separated from an unusual active site by a beta-flap structure. Functional studies confirm an intramolecular mechanism of cleavage and highlight specific residues required for InsP6-induced TcdA processing. Analysis of the structural and functional data in the context of sequences from similar and diverse origins highlights a C-terminal extension and a pi-cation interaction within the beta-flap that appear to be unique among the large clostridial cytotoxins.
Collapse
Affiliation(s)
- Rory N Pruitt
- Departments of Microbiology and Immunology, Nashville, Tennessee 37232
| | | | - Michael Cover
- Departments of Microbiology and Immunology, Nashville, Tennessee 37232
| | | | - Ben Spiller
- Departments of Microbiology and Immunology, Nashville, Tennessee 37232; Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - D Borden Lacy
- Departments of Microbiology and Immunology, Nashville, Tennessee 37232; Biochemistry, Nashville, Tennessee 37232
| |
Collapse
|
118
|
Gardiner DF, Rosenberg T, Zaharatos J, Franco D, Ho DD. A DNA vaccine targeting the receptor-binding domain of Clostridium difficile toxin A. Vaccine 2009; 27:3598-604. [PMID: 19464540 PMCID: PMC2709243 DOI: 10.1016/j.vaccine.2009.03.058] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 03/07/2009] [Accepted: 03/17/2009] [Indexed: 12/18/2022]
Abstract
Clostridium difficile is a pathogen with increasing severity for which host antibody responses provide protection from disease. DNA vaccination has several advantages compared to traditional vaccine methods, however no study has examined this platform against C. difficile toxins. A synthetic gene was created encoding the receptor-binding domain (RBD) of C. difficile toxin A, optimized for expression in human cells. Gene expression was examined in vitro. Mice were inoculated and then challenged with parenteral toxin A. Vaccination provided high titer antibodies and protected mice from death. This represents the first report of DNA vaccine inducing neutralizing antibodies to C. difficile toxin A.
Collapse
Affiliation(s)
- David F Gardiner
- Division of International Medicine and Infectious Diseases, Weill Cornell Medical College, New York, NY, United States.
| | | | | | | | | |
Collapse
|
119
|
Wolff D, Brüning T, Gerritzen A. Rapid detection of the Clostridium difficile ribotype 027 tcdC gene frame shift mutation at position 117 by real-time PCR and melt curve analysis. Eur J Clin Microbiol Infect Dis 2009; 28:959-62. [PMID: 19333630 DOI: 10.1007/s10096-009-0731-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 03/17/2009] [Indexed: 10/21/2022]
Abstract
The emergence of the hypervirulent strain Clostridium difficile PCR ribotype 027 has increased the necessity for rapid C. difficile typing tests for clinical and epidemiological purposes. We developed a rapid real-time polymerase chain reaction (PCR) test for the detection of C. difficile. As the target, we chose the tcdC gene, which encodes for a negative regulator in toxin production. A deletion at position 117 of the tcdC gene, which is associated with severe tcdC truncation, is well conserved in all PCR ribotype 027 isolates. Probe sequences of the real-time PCR test were designed to result in distinct melt profiles for sequence variations at positions 117 to 120 of the tcdC gene. The tcdC gene deletion at position 117 was easily detected with real-time PCR and melt curve analysis in all C. difficile ribotype 027 isolates. In five non-027 strains and 46 hospitalised patient samples, melt curve analysis detected no deletion. PCR results were confirmed by DNA sequencing. The combination of real-time PCR and melt curve analysis is a rapid and accurate method for the detection of C. difficile DNA and simultaneous screening for the tcdC gene deletion at position 117, which is closely related to the C. difficile PCR ribotype 027 strain.
Collapse
Affiliation(s)
- D Wolff
- Medical Laboratory Bremen, Haferwende 12, 28357 Bremen, Germany.
| | | | | |
Collapse
|
120
|
Ochsner UA, Bell SJ, O'Leary AL, Hoang T, Stone KC, Young CL, Critchley IA, Janjic N. Inhibitory effect of REP3123 on toxin and spore formation in Clostridium difficile, and in vivo efficacy in a hamster gastrointestinal infection model. J Antimicrob Chemother 2009; 63:964-71. [PMID: 19251726 DOI: 10.1093/jac/dkp042] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES REP3123 is a fully synthetic methionyl-tRNA synthetase inhibitor in pre-clinical development as a novel agent to treat Clostridium difficile infection (CDI). This novel agent was investigated for its ability to block the production of toxins and spores, and was tested for efficacy in vivo in a hamster model. METHODS Clostridial toxin levels were determined qualitatively using monoclonal antibodies and by cytotoxicity assays. Spores were detected by staining and by quantitative dilution plating after ethanol treatment. Efficacy of REP3123 was tested in a clindamycin-induced C. difficile hamster gastrointestinal (GI) infection model. RESULTS REP3123 at concentrations as low as 1 mg/L inhibited de novo toxin production in high cell density, stationary phase cultures of C. difficile. Among comparator agents currently used for CDI therapy, vancomycin required much higher levels of 20 mg/L, and metronidazole had no effect on toxin levels. REP3123 caused a >10-fold reduction of the sporulation rate in vitro. Vancomycin and, in particular, metronidazole appeared to promote the formation of spores. REP3123, at concentrations as low as 0.5 mg/kg, demonstrated efficacy in the hamster model of CDI and was superior to vancomycin in the overall survival of the animals at the end of the study (33 days). CONCLUSIONS REP3123 inhibited growth of C. difficile, affected the production of toxins and spores and demonstrated superior efficacy compared with vancomycin in the hamster GI infection model. This agent may be a promising candidate for CDI treatment; in particular, the inhibition of toxin production and spore formation may reduce the severity and spread of the disease, respectively.
Collapse
|
121
|
Jaber MR, Olafsson S, Fung WL, Reeves ME. Clinical review of the management of fulminant clostridium difficile infection. Am J Gastroenterol 2008; 103:3195-203; quiz 3204. [PMID: 18853982 DOI: 10.1111/j.1572-0241.2008.02198.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Clostridium difficile infection (CDI) is a frequent cause of morbidity and mortality among elderly hospitalized patients. A small but increasing number of patients have developed fulminant CDI, and a significant number of these patients require emergency colectomy. In this review, we discuss the risk factors, pathophysiology, diagnosis, and management of fulminant CDI. DATA SOURCES A literature search (Medline, Embase, Cochrane Library, Biosis, Science Citation Index, Ovid Journals) was performed from the period between January 1980 and June 2008 using the key words "Clostridium difficile,""pseudomembranous enterocolitis,""colectomy,""acute abdomen,""antibiotic-associated diarrhea," or "fulminant Clostridium difficile colitis." Articles not in English or not related to human subjects were excluded. For this review, we analyzed the articles identified in our original search and those articles cited in the original review articles. No randomized trials were found on the surgical management of fulminant CDI and only retrospective studies with a minimum of five patients were used in the review. With respect to medical treatment, we based our review on guideline articles, systematic reviews, and available randomized trials. CONCLUSION Both the incidence and severity of CDI are increasing. Fulminant CDI is underappreciated as a life-threatening disease because of a lack of awareness of its severity and its nonspecific clinical syndrome. Early diagnosis and treatment are essential for a good outcome, and early surgical intervention should be used in patients who are unresponsive to medical therapy. The surgical procedure of choice is a total abdominal colectomy with end ileostomy, although the mortality rate remains high.
Collapse
Affiliation(s)
- M Raffat Jaber
- Department of Surgery, Loma Linda University Medical Center, Loma, Linda, California 92354, USA
| | | | | | | |
Collapse
|
122
|
Persson S, Torpdahl and M, Olsen K. New multiplex PCR method for the detection of Clostridium difficile toxin A (tcdA) and toxin B (tcdB) and the binary toxin (cdtA/cdtB) genes applied to a Danish strain collection. Clin Microbiol Infect 2008; 14:1057-64. [DOI: 10.1111/j.1469-0691.2008.02092.x] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
123
|
Genetic relatedness of Clostridium difficile isolates from various origins determined by triple-locus sequence analysis based on toxin regulatory genes tcdC, tcdR, and cdtR. J Clin Microbiol 2008; 46:3703-13. [PMID: 18832125 DOI: 10.1128/jcm.00866-08] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A triple-locus nucleotide sequence analysis based on toxin regulatory genes tcdC, tcdR and cdtR was initiated to assess the sequence variability of these genes among Clostridium difficile isolates and to study the genetic relatedness between isolates. A preliminary investigation of the variability of the tcdC gene was done with 57 clinical and veterinary isolates. Twenty-three isolates representing nine main clusters were selected for tcdC, tcdR, and cdtR analysis. The numbers of alleles found for tcdC, tcdR and cdtR were nine, six, and five, respectively. All strains possessed the cdtR gene except toxin A-negative toxin B-positive variants. All but one binary toxin CDT-positive isolate harbored a deletion (>1 bp) in the tcdC gene. The combined analyses of the three genes allowed us to distinguish five lineages correlated with the different types of deletion in tcdC, i.e., 18 bp (associated or not with a deletion at position 117), 36 bp, 39 bp, and 54 bp, and with the wild-type tcdC (no deletion). The tcdR and tcdC genes, though located within the same pathogenicity locus, were found to have evolved separately. Coevolution of the three genes was noted only with strains harboring a 39-bp or a 54-bp deletion in tcdC that formed two homogeneous, separate divergent clusters. Our study supported the existence of the known clones (PCR ribotype 027 isolates and toxin A-negative toxin B-positive C. difficile variants) and evidence for clonality of isolates with a 39-bp deletion (toxinotype V, PCR ribotype 078) that are frequently isolated worldwide from human infections and from food animals.
Collapse
|
124
|
Abstract
Clostridium difficile is the leading cause of antibiotic-associated diarrhea and pseudomembranous colitis, which have significant morbidity and mortality. Accurate and timely diagnosis is critical. Repeat enzyme immunoassay testing for C. difficile toxin has been recommended because of <100% sensitivity. All C. difficile tests between 1 January 2006 and 31 December 2006 were retrospectively analyzed for results and testing patterns. The Wampole C. difficile Tox A/B II enzyme immunoassay kit was used. There were a total of 8,256 tests from 3,112 patients; 49% of tests were repeated. Of the 3,749 initially negative patient tests, 96 were positive upon repeat testing within 10 days of the first test. Of repeat tests, 0.9% repeated on day 0 (same day as the first test), 1.8% on day 1, 3.8% on day 2, 2.6% on day 3, 5.4% on days 4 to 6, and 10.6% on days 7 to 10 were positive. Thirty-eight patients had a positive test within 48 h of an initial negative test, and based on chart review, 18 patients were treated empirically while 16 were treated following the new result. None had evidence of medical complications. Of initially positive patients, 91% were positive upon repeat testing on day 0, 75% on day 1, and 58% on day 2, to a low of 14% on days 7 to 10. Depending on the clinical setting, these data support not repeating C. difficile tests within 2 days of a negative result and limiting repeat testing to >/=1 week of a positive result.
Collapse
|
125
|
Chow WHA, McCloskey C, Tong Y, Hu L, You Q, Kelly CP, Kong H, Tang YW, Tang W. Application of isothermal helicase-dependent amplification with a disposable detection device in a simple sensitive stool test for toxigenic Clostridium difficile. J Mol Diagn 2008; 10:452-8. [PMID: 18669881 DOI: 10.2353/jmoldx.2008.080008] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enzyme immunoassays (EIAs) are commonly used for the diagnosis of cases of Clostridium difficile-associated diarrhea (CDAD). However, these EIAs have high false-negative rates, even in patients with severe clinical disease. We have developed an IsoAmp CDAD test using a simple and user-friendly procedure to identify toxigenic C. difficile in feces. After DNA extraction from fecal samples, both the conserved sequence of the 5'-end fragment of the C. difficile tcdA toxin gene and competitive amplification internal control sequence were amplified using helicase-dependent amplification. Amplification products were detected using a novel amplicon-containment detection device. The analytical sensitivity of the assay was 20 copies of C. difficile genomic DNA per reaction. Evaluation of the clinical sensitivity and specificity of the IsoAmp CDAD test versus an EIA method using a PCR method as the reference standard revealed 100% sensitivity and 100% specificity for the IsoAmp CDAD test compared with 90.9% sensitivity and 100% specificity for the EIA method. Because the IsoAmp CDAD test requires no expensive equipments for nucleic acid amplification or detection and can be performed on a random access basis, the test provides a practical alternative to immunoassays for the diagnosis of CDAD with improved sensitivity.
Collapse
|
126
|
Dupuy B, Govind R, Antunes A, Matamouros S. Clostridium difficile toxin synthesis is negatively regulated by TcdC. J Med Microbiol 2008; 57:685-689. [DOI: 10.1099/jmm.0.47775-0] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Clostridium difficile toxin synthesis is growth phase-dependent and is regulated by various environmental signals. The toxin genes tcdA and tcdB are located in a pathogenicity locus, which also includes three accessory genes, tcdR, tcdC and tcdE. TcdR has been shown to act as an alternative σ factor that mediates positive regulation of both the toxin genes and its own gene. The tcdA, tcdB and tcdR genes are transcribed during the stationary growth phase. The tcdC gene, however, is expressed during exponential phase. This expression pattern suggested that TcdC may act as a negative regulator of toxin gene expression. TcdC is a small acidic protein without any conserved DNA-binding motif. It is able to form dimers and its N-terminal region includes a putative transmembrane domain. Genetic and biochemical evidence showed that TcdC negatively regulates C. difficile toxin synthesis by interfering with the ability of TcdR-containing RNA polymerase to recognize the tcdA and tcdB promoters. In addition, the C. difficile NAP1/027 epidemic strains that produce higher levels of toxins have mutations in tcdC. Interestingly, a frameshift mutation at position 117 of the tcdC coding sequence seems to be, at least in part, responsible for the hypertoxigenicity phenotype of these epidemic strains.
Collapse
Affiliation(s)
- B. Dupuy
- Unité des Toxines et Pathogénie Bactérienne, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - R. Govind
- Unité des Toxines et Pathogénie Bactérienne, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - A. Antunes
- Unité des Toxines et Pathogénie Bactérienne, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - S. Matamouros
- Unité des Toxines et Pathogénie Bactérienne, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
127
|
Barreto ARF, Cavalcante IC, Castro MV, Junqueira AFTA, Vale MR, Ribeiro RA, Souza MHLP, Brito GAC. Fucoidin prevents Clostridium difficile toxin-A-induced ileal enteritis in mice. Dig Dis Sci 2008; 53:990-6. [PMID: 17805968 DOI: 10.1007/s10620-007-9957-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Accepted: 08/01/2007] [Indexed: 12/18/2022]
Abstract
Recent reports suggest increased incidence and severity of Clostridium difficile-associated diseases. These facts have raised the need for additional clarification of pathogenesis and for a search for new therapeutic strategies. This study evaluated the effects of the polysaccharide fucoidin, an L-selectin blocker, on toxin-A-induced mouse enteritis. Fucoidin (25 mg/kg) or saline (0.1 ml) were injected systemically (ocular plexus) 5 min prior to local challenge with toxin A (5 microg/ileal loop) or phosphate-buffered saline (PBS). Intestinal fluid volume/length and ileal loop weight/length ratios were calculated 3 h later. Ileal tissues were collected for histopathology and measurement of myeloperoxidase and adenosine deaminase activity. Fucoidin significantly (P < 0.05) prevented the toxin-A-induced increase in weight/length and volume/length ratios and reduced mucosal disruption, as shown in histopathology. Fucoidin also significantly (P < 0.05) reduced toxin-A-induced myeloperoxidase and adenosine deaminase activities. In conclusion, fucoidin reduces tissue injury and inflammation in toxin-A-induced mouse enteritis.
Collapse
Affiliation(s)
- A R F Barreto
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, Rua Delmiro de Farias, sn, Fortaleza, CE CEP 60.416-030, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Song HJ, Shim KN, Jung SA, Choi HJ, Lee MA, Ryu KH, Kim SE, Yoo K. Antibiotic-associated diarrhea: candidate organisms other than Clostridium difficile. Korean J Intern Med 2008; 23:9-15. [PMID: 18363274 PMCID: PMC2686956 DOI: 10.3904/kjim.2008.23.1.9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND/AIMS The direct toxic effects of antibiotics on the intestine can alter digestive functions and cause pathogenic bacterial overgrowth leading to antibiotic-associated diarrhea (AAD). Clostridium difficile (C. difficile) is widely known to be responsible for 10 approximately 20% of AAD cases. However, Klebsiella oxytoca, Clostridium perfringens, Staphylococcus aureus, and Candida species might also contribute to AAD. METHODS We prospectively analyzed the organisms in stool and colon tissue cultures with a C. difficile toxin A assay in patients with AAD between May and December 2005. In addition, we performed the C. difficile toxin A assays using an enzyme-linked fluorescent assay technique. Patients were enrolled who had diarrhea with more than three stools per day for at least 2 days after the initiation of antibiotic treatment for up to 6 approximately 8 weeks after antibiotic discontinuation. RESULTS Among 38 patients (mean age 59 +/- 18 years, M:F =18:20), the organism isolation rates were 28.9% (11/38) for stool culture, 18.4% (7/38) for colon tissue cultures and 13.2% (5/38) for the C. difficile toxin A assay. The overall rate of identification of organisms was 50.0% (19/38). Of the five patients that had a positive result by the C. difficile toxin A assay, two had no organism isolated by the stool or colon tissue culture. The organisms isolated from the stool cultures were C difficile (4), Klebsiella pneumoniae (K. pneumoniae) (3), Candida species (3), and Staphylococcus aureus (1). C. difficile (4) and K. pneumoniae (3) were isolated from the colon tissue culture. CONCLUSIONS For C. difficile negative AAD patients, K. pneumoniae, Candida species and Staphylococcus aureus were found to be potential causative organisms.
Collapse
Affiliation(s)
- Hyun Joo Song
- Department of Internal Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Ki-Nam Shim
- Department of Internal Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Sung-Ae Jung
- Department of Internal Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hee Jung Choi
- Department of Internal Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Mi Ae Lee
- Department of Laboratory Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Kum Hei Ryu
- Department of Internal Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Seong-Eun Kim
- Department of Internal Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Kwon Yoo
- Department of Internal Medicine, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Seoul, Korea
| |
Collapse
|
129
|
Barbut F. [Clostridium difficile infections: an unexpected re-emergence]. PATHOLOGIE-BIOLOGIE 2008; 56:6-9. [PMID: 18178330 DOI: 10.1016/j.patbio.2007.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Accepted: 09/19/2007] [Indexed: 05/25/2023]
|
130
|
C-terminal repeats of Clostridium difficile toxin A induce production of chemokine and adhesion molecules in endothelial cells and promote migration of leukocytes. Infect Immun 2007; 76:1170-8. [PMID: 18160482 DOI: 10.1128/iai.01340-07] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The C-terminal repeating sequences of Clostridium difficile toxin A (designated ARU) are homologous to the carbohydrate-binding domain of streptococcal glucosyltransferases (GTFs) that were recently identified as potent modulins. To test the hypothesis that ARU might exert a similar biological activity on endothelial cells, recombinant ARU (rARU), which was noncytotoxic to cell cultures, was analyzed using human umbilical vein endothelial cells. The rARU could bind directly to endothelial cells in a serum- and calcium-dependent manner and induce the production of interleukin-6 (IL-6), IL-8, and monocyte chemoattractant protein 1 in a dose-dependent manner. An oligosaccharide binding assay indicated that rARU, but not GTFC, binds preferentially to Lewis antigens and 3'HSO3-containing oligosaccharides. Binding of rARU to human endothelial or intestinal cells correlated directly with the expression of Lewis Y antigen. Bound rARU directly activated mitogen-activated protein kinases and the NF-kappaB signaling pathway in endothelial cells to release biologically active chemokines and adhesion molecules that promoted migration in a transwell assay and the adherence of polymorphonuclear and mononuclear cells to the endothelial cells. These results suggest that ARU may bind to multiple carbohydrate motifs to exert its biological activity on human endothelial cells.
Collapse
|
131
|
Gougoulias C, Tuohy KM, Gibson GR. Dietary-based gut flora modulation againstClostridium difficileonset. ACTA ACUST UNITED AC 2007. [DOI: 10.1616/1476-2137.14986] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
132
|
Taha S, Johansson O, Rivera Jonsson S, Heimer D, Krovacek K. Toxin production by and adhesive properties of Clostridium difficile isolated from humans and horses with antibiotic-associated diarrhea. Comp Immunol Microbiol Infect Dis 2007; 30:163-74. [PMID: 17239950 DOI: 10.1016/j.cimid.2006.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2006] [Indexed: 11/25/2022]
Abstract
Clostridium difficile is a common nosocomial pathogen in humans and animals that causes diarrhea and colitis following antibiotic therapy. Isolates of C. difficile obtained from faecal material from 20 human patients and 6 equine subjects with antibiotic-associated diarrhea were investigated regarding production of toxins A and B, their capacity to adhere to the human intestinal Caco-2 cell line and equine intestinal cells, and for the presence of fimbriae. The results showed that most (17/20) of the human clinical isolates produced both toxins A and B. One of the human isolates proved toxin A-negative/toxin B-positive. All (6/6) horse isolates were positive for both toxins A and B. Both the human and horse isolates possessed the capacity to adhere, to varying degree, to human and equine intestinal cells. It appeared that human isolates produced greater amounts of toxin B, and that there was a host-species dependency on ability to attach to intestinal epithelial cells. No fimbriae were found in any of the investigated isolates.
Collapse
Affiliation(s)
- Sawsan Taha
- Department of Biomedical Sciences and Veterinary Public Health, Faculty of Veterinary Medicine and Animal Science, SLU, Box 7036, 750 07 Uppsala, Sweden
| | | | | | | | | |
Collapse
|
133
|
Ghose C, Kalsy A, Sheikh A, Rollenhagen J, John M, Young J, Rollins SM, Qadri F, Calderwood SB, Kelly CP, Ryan ET. Transcutaneous immunization with Clostridium difficile toxoid A induces systemic and mucosal immune responses and toxin A-neutralizing antibodies in mice. Infect Immun 2007; 75:2826-32. [PMID: 17371854 PMCID: PMC1932889 DOI: 10.1128/iai.00127-07] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clostridium difficile is the leading cause of nosocomial infectious diarrhea. C. difficile produces two toxins (A and B), and systemic and mucosal anti-toxin A antibodies prevent or limit C. difficile-associated diarrhea. To evaluate whether transcutaneous immunization with formalin-treated C. difficile toxin A (CDA) induces systemic and mucosal anti-CDA immune responses, we transcutaneously immunized three cohorts of mice with CDA with or without immunoadjuvantative cholera toxin (CT) on days 0, 14, 28, and 42. Mice transcutaneously immunized with CDA and CT developed prominent anti-CDA and anti-CT immunoglobulin G (IgG) and IgA responses in serum and anti-CDA and anti-CT IgA responses in stool. Sera from immunized mice were able to neutralize C. difficile toxin A activity in an in vitro cell culture assay. CDA itself demonstrated adjuvant activity and enhanced both serum and stool anti-CT IgA responses. Our results suggest that transcutaneous immunization with CDA toxoid may be a feasible immunization strategy against C. difficile, an important cause of morbidity and mortality against which current preventative strategies are failing.
Collapse
Affiliation(s)
- Chandrabali Ghose
- Division of Infectious Diseases, Massachusetts General Hospital, Jackson 504, 55 Fruit Street, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Reineke J, Tenzer S, Rupnik M, Koschinski A, Hasselmayer O, Schrattenholz A, Schild H, von Eichel-Streiber C. Autocatalytic cleavage of Clostridium difficile toxin B. Nature 2007; 446:415-9. [PMID: 17334356 DOI: 10.1038/nature05622] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Accepted: 01/25/2007] [Indexed: 02/04/2023]
Abstract
Clostridium difficile, the causative agent of nosocomial antibiotic-associated diarrhoea and pseudomembranous colitis, possesses two main virulence factors: the large clostridial cytotoxins A and B. It has been proposed that toxin B is cleaved by a cytosolic factor of the eukaryotic target cell during its cellular uptake. Here we report that cleavage of not only toxin B, but also all other large clostridial cytotoxins, is an autocatalytic process dependent on host cytosolic inositolphosphate cofactors. A covalent inhibitor of aspartate proteases, 1,2-epoxy-3-(p-nitrophenoxy)propane, completely blocked toxin B function on cultured cells and was used to identify its catalytically active protease site. To our knowledge this is the first report on a bacterial toxin that uses eukaryotic signals for induced autoproteolysis to deliver its toxic domain into the cytosol of target cells. On the basis of our data, we present an integrated model for the uptake and inositolphosphate-induced activation of toxin B.
Collapse
Affiliation(s)
- Jessica Reineke
- Johannes-Gutenberg Universität Mainz, Institut für medizinische Mikrobiologie and Hygiene, Hochhaus am Augustusplatz, 55131 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Curry SR, Marsh JW, Muto CA, O'Leary MM, Pasculle AW, Harrison LH. tcdC genotypes associated with severe TcdC truncation in an epidemic clone and other strains of Clostridium difficile. J Clin Microbiol 2006; 45:215-21. [PMID: 17035492 PMCID: PMC1828959 DOI: 10.1128/jcm.01599-06] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Severe Clostridium difficile associated disease is associated with outbreaks of the recently described BI/NAP1 epidemic clone. This clone is characterized by an 18-bp deletion in the tcdC gene and increased production of toxins A and B in vitro. TcdC is a putative negative regulator of toxin A&B production. We characterized tcdC genotypes from a collection of C. difficile isolates from a hospital that experienced an outbreak caused by the BI/NAP1 epidemic clone. Sequence analysis of tcdC was performed on DNA samples isolated from 199 toxigenic C. difficile isolates (31% BI/NAP1) from 2001 and 2005. Sequences obtained from 36 (18.6%) isolates predicted wild-type TcdC (232 amino acid residues), whereas 12 (6.1%) isolates had tcdC genotypes with previously described 18- or 39-bp deletions. The remaining isolates comprised 15 unique genotypes. Of these, 5 genotypes contain 18- or 36-bp deletions. Of these five genotypes, one is characterized by a single nucleotide deletion at position 117 resulting in a frameshift that introduces a stop codon at position 196, truncating the predicted TcdC to 65 amino acid residues. All 62 of the isolates in this collection comprising the epidemic clone are characterized by this genotype. This result suggests that severe truncation of TcdC is responsible for the increased toxin production observed in strains belonging to the BI/NAP1 clone and that the 18-bp deletion is probably irrelevant to TcdC function. Further investigations are required to determine the effect of this and other tcdC genotypes on toxin production and clinical disease.
Collapse
Affiliation(s)
- Scott R Curry
- Department of Medicine, University of Pittsburgh, School of Medicine, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
136
|
Chae S, Eckmann L, Miyamoto Y, Pothoulakis C, Karin M, Kagnoff MF. Epithelial cell I kappa B-kinase beta has an important protective role in Clostridium difficile toxin A-induced mucosal injury. THE JOURNAL OF IMMUNOLOGY 2006; 177:1214-20. [PMID: 16818780 DOI: 10.4049/jimmunol.177.2.1214] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Toxin A released by Clostridium difficile interacts with the single layer of intestinal epithelial cells that lines the host's intestinal tract and leads to mucosal damage and inflammation that manifests clinically as antibiotic-associated diarrhea and pseudomembranous colitis. Activation of the transcription factor NF-kappaB in intestinal epithelial cells is important for regulating the expression of epithelial cell proinflammatory genes and cell survival. However, the role of NF-kappaB activation in the pathogenesis of C. difficile toxin A-induced colitis is unknown. To determine the functional importance in vivo of NF-kappaB activation in intestinal epithelium in the pathogenesis of C. difficile-induced colitis, we used mutant mice that do not activate the classical NF-kappaB signaling pathway in intestinal epithelial cells due to a conditional deficiency in those cells of the IkappaB-kinase beta (IKKbeta) subunit of IKK. C. difficile toxin A challenge of intestinal loops in intestinal epithelial cell IKKbeta-deficient mice induced a rapid and significant increase in intestinal epithelial apoptosis compared with littermate controls. This was accompanied by a significant increase in acute mucosal inflammation, mucosal injury, luminal fluid secretion, and bacterial translocation. We conclude that activation of intestinal epithelial cell NF-kappaB by toxin A plays an important host mucosal protective role after C. difficile toxin A exposure that is mediated, at least in part, through promoting epithelial cell survival by abrogating epithelial cell apoptosis.
Collapse
Affiliation(s)
- Sungwon Chae
- Department of Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
137
|
Cavalcante IC, Castro MV, Barreto ARF, Sullivan GW, Vale M, Almeida PRC, Linden J, Rieger JM, Cunha FQ, Guerrant RL, Ribeiro RA, Brito GAC. Effect of novel A2A adenosine receptor agonist ATL 313 on Clostridium difficile toxin A-induced murine ileal enteritis. Infect Immun 2006; 74:2606-12. [PMID: 16622196 PMCID: PMC1459724 DOI: 10.1128/iai.74.5.2606-2612.2006] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Clostridium difficile is a spore-forming, anaerobic, gram-positive bacillus that releases two main virulence factors: toxins A and B. Toxin A plays an important pathogenic role in antibiotic-induced diarrhea and pseudomembranous colitis, a condition characterized by intense mucosal inflammation and secretion. Agonist activity at A2A adenosine receptors attenuates inflammation and damage in many tissues. This study evaluated the effects of a new selective A2A adenosine receptor agonist (ATL 313) on toxin A-induced injury in murine ileal loops. ATL 313 (0.5 to 5 nM) and/or the A2A adenosine receptor antagonist (ZM241385; 5 nM) or phosphate-buffered saline (PBS) were injected into ileal loops immediately prior to challenge with toxin A (1 to 10 microg/loop) or PBS. Intestinal fluid volume/length and weight/length ratios were calculated 3 h later. Ileal tissues were collected for the measurement of myeloperoxidase, adenosine deaminase activity, tumor necrosis factor alpha (TNF-alpha) production, histopathology, and detection of cell death by the TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling) method. Toxin A significantly increased volume/length and weight/length ratios in a dose-dependent fashion. ATL 313 treatment significantly (P < 0.05) reduced toxin A-induced secretion and edema, prevented mucosal disruption, and neutrophil infiltration as measured by myeloperoxidase activity. ATL 313 also reduced the toxin A-induced TNF-alpha production and adenosine deaminase activity and prevented toxin A-induced cell death. These protective effects of ATL 313 were reversed by ZM241385. In conclusion, the A2A adenosine receptor agonist, ATL 313, reduces tissue injury and inflammation in mice with toxin A-induced enteritis. The finding of increased ileal adenosine deaminase activity following the administration of toxin A is new and might contribute to the pathogenesis of the toxin A-induced enteritis by deaminating endogenous adenosine.
Collapse
Affiliation(s)
- I C Cavalcante
- Faculdade de Medicina, Universidade Federal do Ceará, Rua Delmiro de Farias, sn CEP 60.416-030, Fortaleza, CE, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Rexach CE, Tang-Feldman YJ, Cantrell MC, Cohen SH. Epidemiologic surveillance of Clostridium difficile diarrhea in a freestanding pediatric hospital and a pediatric hospital at a university medical center. Diagn Microbiol Infect Dis 2006; 56:109-14. [PMID: 16678379 DOI: 10.1016/j.diagmicrobio.2006.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2005] [Revised: 03/04/2006] [Accepted: 03/08/2006] [Indexed: 01/09/2023]
Abstract
To describe the epidemiology of Clostridium difficile in children, we cultured stool specimens from patients at the Children's Hospital Central California, Madera, CA (CHCC, n = 676) and at the University of California Davis Medical Center Pediatric Hospital, Sacramento, CA (UCDMC-PH, n = 301) for C. difficile, and toxins A and B genes and strain identity of the isolates were determined by polymerase chain reaction assays. A higher percentage of patients from UCDMC-PH were culture positive (148/301, 49%) and colonized with toxigenic strains (45/301, 15%) compared with CHCC (colonized = 178/676, 26%; toxigenic = 96/676, 14%, P < or = .001). Multiple logistic regression analysis showed decreased colonization with inpatient status (odds ratio [OR] = 0.64; 95% confidence interval [CI] = 0.46, 0.89; P = .007) and use of H-2 antagonists (OR = 0.55; 95% CI = 0.36, 0.84; P = .006), whereas underlying conditions (colonization: OR = 1.42; 95% CI = 1.02, 1.96; P = .04; toxin positive: OR = 1.60; 95% CI = 1.04, 2.44; P = .03) and exposure to > or =2 antiinfectives (colonization: OR = 1.56; 95% CI = 1.10, 2.20; P = .01; toxin positive: OR = 1.71; 95% CI = 1.10, 2.66; P = .02) increased colonization. Most isolates appear to be community acquired, although molecular analysis suggests some nosocomial transmission at UCDMC-PH. These data suggest that the epidemiology of colonization with C. difficile in children is different than previously reported.
Collapse
Affiliation(s)
- Carmen E Rexach
- Division of Natural Sciences, University of California, Merced, CA 95344, USA.
| | | | | | | |
Collapse
|
139
|
Akerlund T, Svenungsson B, Lagergren A, Burman LG. Correlation of disease severity with fecal toxin levels in patients with Clostridium difficile-associated diarrhea and distribution of PCR ribotypes and toxin yields in vitro of corresponding isolates. J Clin Microbiol 2006; 44:353-8. [PMID: 16455883 PMCID: PMC1392646 DOI: 10.1128/jcm.44.2.353-358.2006] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated in vivo and in vitro yields of toxins A and B from and PCR ribotypes of Clostridium difficile isolates from 164 patients with differing severities of C. difficile-associated diarrhea (CDAD) (patients were grouped as follows: <3 loose stools per day, n = 45; 3 to 10 per day, n = 97; >10 per day, n = 22). The median fecal toxin levels in each group were 0.5, 6.8, and 149 U/g feces (P < 0.001), respectively. Patients with severe diarrhea also had more-frequent occurrence of blood in stool and vomiting, but there was no association with fecal toxin levels per se. There was no correlation between fecal toxin level and toxin yield in vitro for the corresponding C. difficile isolate or between its PCR ribotype and disease severity. A broad range of toxin yields among isolates belonging to major PCR ribotypes indicated a presence of many subtypes. We hypothesize that bacterial and host factors that affect C. difficile toxin levels in feces are important determinants of symptoms in CDAD patients. An inverse correlation between toxin yield and spore count (r = 0.66) in stationary-phase cultures supported the notion that toxin production and sporulation represent opposite alternative survival strategies for C. difficile cells facing nutrient shortage.
Collapse
Affiliation(s)
- Thomas Akerlund
- Department of Bacteriology, Swedish Institute for Infectious Disease Control, S-17182, Solna, Sweden.
| | | | | | | |
Collapse
|
140
|
Nascimento SB, Sousa RB, Martins MJB, Souza Gomes A, Souza MHLP, Guerrant RL, Cunha FQ, Ribeiro RA, Brito GAC. Glutamine depletion potentiates leucocyte-dependent inflammatory events induced by carrageenan or Clostridium difficile toxin A in rats. Immunology 2005; 116:328-36. [PMID: 16236122 PMCID: PMC1802418 DOI: 10.1111/j.1365-2567.2005.02232.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
This research investigated the effect of glutamine (Gln) depletion on leucocyte-dependent inflammatory events. Rats were treated intraperitoneally, 16 hr prior to the peak of every parameter evaluated, with either 0.9% NaCl, methionine-sulphoximine (MSO, an inhibitor of endogenous Gln synthesis, 25 mg/kg) or with MSO + Gln (MSO as above plus Gln 3 g/kg in three doses). MSO-induced Gln depletion increased paw oedema induced both by carrageenan (Cg) and by Clostridium difficile toxin A (TxA) (66.2% and 45.5%, respectively; P < 0.05). In dextran-injected animals, oedema and myeloperoxidase (MPO) activity were not modified by Gln depletion. In Cg-treated paws, Gln depletion increased MPO activity by 44% (P < 0.05), interleukin-1beta (IL-1beta) and tumour necrosis factor-alpha (TNF-alpha) concentrations by 47% and 52%, respectively (P < 0.05), and immunostaining for TNF-alpha in paw tissue. In TxA-injected paws, Gln depletion increased MPO activity (46%; P < 0.05). Gln depletion increased Cg- and TxA-induced neutrophil migration to subcutaneous air pouches by 56% and 77% (P < 0.05), respectively, but did not affect migration induced by N-formyl-methionyl-leucyl-phenylalanine (fMLP). Gln infusions reversed all the effects of MSO. Leucocyte counts did not differ between groups. Gln depletion potentiates acute inflammation, possibly by increasing neutrophil migration through resident cell activation and production of IL-1beta and TNF-alpha. Gln supplementation reverses these effects and may be useful during inflammatory catabolic stress.
Collapse
Affiliation(s)
- Silvia B Nascimento
- Department of Physiology and Pharmacology, The Federral University of Ceará, Fortaleza, CE, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Abstract
Rho proteins are master regulators of a large array of cellular functions, including control of cell morphology, cell migration and polarity, transcriptional activation, and cell cycle progression. They are the eukaryotic targets of various bacterial protein toxins and effectors, which activate or inactivate the GTPases. Here Rho-inactivating toxins and effectors are reviewed, including the families of large clostridial cytotoxins and C3-like transferases, which inactivate Rho GTPases by glucosylation and ADP-ribosylation, respectively.
Collapse
Affiliation(s)
- K Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albertstrasse 25, 79104 Freiburg, Germany.
| | | |
Collapse
|
142
|
Dhalluin A, Bourgeois I, Pestel-Caron M, Camiade E, Raux G, Courtin P, Chapot-Chartier MP, Pons JL. Acd, a peptidoglycan hydrolase of Clostridium difficile with N-acetylglucosaminidase activity. Microbiology (Reading) 2005; 151:2343-2351. [PMID: 16000724 DOI: 10.1099/mic.0.27878-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A gene encoding a putative peptidoglycan hydrolase was identified by sequence similarity searching in the Clostridium difficile 630 genome sequence, and the corresponding protein, named Acd (autolysin of C. difficile) was expressed in Escherichia coli. The deduced amino acid sequence of Acd shows a modular structure with two main domains: an N-terminal domain exhibiting repeated sequences and a C-terminal catalytic domain. The C-terminal domain exhibits sequence similarity with the glucosaminidase domains of Staphylococcus aureus Atl and Bacillus subtilis LytD autolysins. Purified recombinant Acd produced in E. coli was confirmed to be a cell-wall hydrolase with lytic activity on the peptidoglycan of several Gram-positive bacteria, including C. difficile. The hydrolytic specificity of Acd was studied by RP-HPLC analysis and MALDI-TOF MS using B. subtilis cell-wall extracts. Muropeptides generated by Acd hydrolysis demonstrated that Acd hydrolyses peptidoglycan bonds between N-acetylglucosamine and N-acetylmuramic acid, confirming that Acd is an N-acetylglucosaminidase. The transcription of the acd gene increased during vegetative cellular growth of C. difficile 630. The sequence of the acd gene appears highly conserved in C. difficile strains. Regarding deduced amino acid sequences, the C-terminal domain with enzymic function appears to be the most conserved of the two main domains. Acd is the first known autolysin involved in peptidoglycan hydrolysis of C. difficile.
Collapse
Affiliation(s)
- Anne Dhalluin
- Groupe de Recherche sur les Antimicrobiens et les Micro-organismes (UPRES EA 2656, IFR 23), Université de Rouen, UFR Médecine-Pharmacie, 22 Boulevard Gambetta, F-76183 Rouen Cedex, France
| | - Ingrid Bourgeois
- Groupe de Recherche sur les Antimicrobiens et les Micro-organismes (UPRES EA 2656, IFR 23), Université de Rouen, UFR Médecine-Pharmacie, 22 Boulevard Gambetta, F-76183 Rouen Cedex, France
| | - Martine Pestel-Caron
- Groupe de Recherche sur les Antimicrobiens et les Micro-organismes (UPRES EA 2656, IFR 23), Université de Rouen, UFR Médecine-Pharmacie, 22 Boulevard Gambetta, F-76183 Rouen Cedex, France
| | - Emilie Camiade
- Groupe de Recherche sur les Antimicrobiens et les Micro-organismes (UPRES EA 2656, IFR 23), Université de Rouen, UFR Médecine-Pharmacie, 22 Boulevard Gambetta, F-76183 Rouen Cedex, France
| | - Gregory Raux
- INSERM U 614 (IFR 23), Université de Rouen, UFR Médecine-Pharmacie, 76183 Rouen Cedex, France
| | - Pascal Courtin
- Unité de Biochimie et Structure des Protéines, INRA, 78352 Jouy-en-Josas Cedex, France
| | | | - Jean-Louis Pons
- Groupe de Recherche sur les Antimicrobiens et les Micro-organismes (UPRES EA 2656, IFR 23), Université de Rouen, UFR Médecine-Pharmacie, 22 Boulevard Gambetta, F-76183 Rouen Cedex, France
| |
Collapse
|
143
|
Abstract
As the leading cause of hospital-acquired diarrhea, Clostridium difficile colonizes the large bowel of patients undergoing antibiotic therapy and produces two toxins, which cause notable disease pathologies. These two toxins, TcdA and TcdB, are encoded on a pathogenicity locus along with negative and positive regulators of their expression. Following expression and release from the bacterium, TcdA and TcdB translocate to the cytosol of target cells and inactivate small GTP-binding proteins, which include Rho, Rac, and Cdc42. Inactivation of these substrates occurs through monoglucosylation of a single reactive threonine, which lies within the effector-binding loop and coordinates a divalent cation critical to binding GTP. By glucosylating small GTPases, TcdA and TcdB cause actin condensation and cell rounding, which is followed by death of the cell. TcdA elicits effects primarily within the intestinal epithelium, while TcdB has a broader cell tropism. Important advances in the study of these toxins have been made in the past 15 years, and these are detailed in this review. The domains, subdomains, and residues of these toxins important for receptor binding and enzymatic activity have been elegantly studied and are highlighted herein. Furthermore, there have been major advances in defining the role of these toxins in modulating the inflammatory events involving the disruption of cell junctions, neuronal activation, cytokine production, and infiltration by polymorphonuclear cells. Collectively, the present review provides a comprehensive update on TcdA and TcdB's mechanism of action as well as the role of these toxins in disease.
Collapse
Affiliation(s)
- Daniel E Voth
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
144
|
Gonenne J, Pardi DS. Clostridium difficile: an update. ACTA ACUST UNITED AC 2005; 30:134-40. [PMID: 15793312 DOI: 10.1007/s12019-004-0009-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Accepted: 02/16/2004] [Indexed: 01/11/2023]
Abstract
Clostridium difficile causes a spectrum of diarrheal illness with the potential for major medical consequences. Although most cases respond quickly to treatment, C. difficile colitis may be severe and life threatening. Recurrent disease represents a particularly challenging problem.
Collapse
Affiliation(s)
- Jonathan Gonenne
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | |
Collapse
|
145
|
Alonso R, Martín A, Peláez T, Marín M, Rodríguez-Creixéms M, Bouza E. Toxigenic status of Clostridium difficile in a large Spanish teaching hospital. J Med Microbiol 2005; 54:159-162. [PMID: 15673510 DOI: 10.1099/jmm.0.45809-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The aim of this study was to evaluate the toxigenic status of circulating strains of Clostridium difficile in a large teaching hospital. Overall 220 isolates were studied of which 199 (90.5 %) produced both large clostridial toxins detected by conventional methods. Ten more strains (4.5 %) had toxin A and B genes detectable by PCR. Eleven (5.0 %) variant strains (A- B+) were detected among the isolates studied and 10 strains (4.5 %) had the binary toxin genes (cdtA and cdtB).
Collapse
Affiliation(s)
- R Alonso
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario ''Gregorio Marañón'', C/Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - A Martín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario ''Gregorio Marañón'', C/Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - T Peláez
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario ''Gregorio Marañón'', C/Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - M Marín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario ''Gregorio Marañón'', C/Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - M Rodríguez-Creixéms
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario ''Gregorio Marañón'', C/Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - E Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario ''Gregorio Marañón'', C/Doctor Esquerdo, 46, 28007 Madrid, Spain
| |
Collapse
|
146
|
Demarest SJ, Salbato J, Elia M, Zhong J, Morrow T, Holland T, Kline K, Woodnutt G, Kimmel BE, Hansen G. Structural characterization of the cell wall binding domains of Clostridium difficile toxins A and B; evidence that Ca2+ plays a role in toxin A cell surface association. J Mol Biol 2005; 346:1197-206. [PMID: 15713474 DOI: 10.1016/j.jmb.2004.12.059] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2004] [Revised: 12/21/2004] [Accepted: 12/22/2004] [Indexed: 11/15/2022]
Abstract
Clostridium difficile (C.difficile) is a nosocomially acquired intestinal bacillus which can cause chronic diarrhea and life-threatening colitis. The pathogenic effects of the bacillus are mediated by the release of two toxins, A and B. The C-terminal portions of both toxins are composed of 20 and 30 residue repeats known as cell wall binding (CWB) domains. We have cloned and expressed the CWB-domains of toxins A and B and several truncated CWB-domain constructs to investigate their structure and function. The smallest CWB-domain that folded in a cooperative manner was an 11 repeat construct of toxin A. This differentiates the C-terminal domains of toxins A and B from the CWB-domain of Streptococcus pneumoniae LytA, which only requires six repeats to fold. The 11 repeat toxin A construct bound Ca2+ directly with millimolar affinity and interacted with mammalian cell surfaces in a concentration and Ca2+-dependent fashion. Millimolar Ca2+ levels also accelerated toxin mediated CHO cell killing in an in vitro cell assay. Together, the data suggest a role for extracellular Ca2+ in the sensitization of toxin A/cell-surface interactions.
Collapse
Affiliation(s)
- Stephen J Demarest
- Department of Protein Therapeutics, Diversa Corp., 4955 Directors Place, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Lemee L, Dhalluin A, Testelin S, Mattrat MA, Maillard K, Lemeland JF, Pons JL. Multiplex PCR targeting tpi (triose phosphate isomerase), tcdA (Toxin A), and tcdB (Toxin B) genes for toxigenic culture of Clostridium difficile. J Clin Microbiol 2005; 42:5710-4. [PMID: 15583303 PMCID: PMC535266 DOI: 10.1128/jcm.42.12.5710-5714.2004] [Citation(s) in RCA: 264] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A multiplex PCR toxigenic culture approach was designed for simultaneous identification and toxigenic type characterization of Clostridium difficile isolates. Three pairs of primers were designed for the amplification of (i) a species-specific internal fragment of the tpi (triose phosphate isomerase) gene, (ii) an internal fragment of the tcdB (toxin B) gene, and (iii) an internal fragment of the tcdA (toxin A) gene allowing distinction between toxin A-positive, toxin B-positive (A+B+) strains and toxin A-negative, toxin B-positive (A-B+) variant strains. The reliability of the multiplex PCR was established by using a panel of 72 C. difficile strains including A+B+, A-B-, and A-B+ toxigenic types and 11 other Clostridium species type strains. The multiplex PCR assay was then included in a toxigenic culture approach for the detection, identification, and toxigenic type characterization of C. difficile in 1,343 consecutive human and animal stool samples. Overall, 111 (15.4%) of 721 human samples were positive for C. difficile; 67 (60.4%) of these samples contained A+B+ toxigenic isolates, and none of them contained A-B+ variant strains. Fifty (8%) of 622 animal samples contained C. difficile strains, which were toxigenic in 27 (54%) cases, including 1 A-B+ variant isolate. Eighty of the 721 human stool samples (37 positive and 43 negative for C. difficile culture) were comparatively tested by Premier Toxins A&B (Meridian Bioscience) and Triage C. difficile Panel (Biosite) immunoassays, the results of which were found concordant with toxigenic culture for 82.5 and 92.5% of the samples, respectively. The multiplex PCR toxigenic culture scheme described here allows combined diagnosis and toxigenic type characterization for human and animal C. difficile intestinal infections.
Collapse
Affiliation(s)
- Ludovic Lemee
- Groupe de Recherche sur les Antimicrobiens et les Microorganismes, Université de Rouen, UFR Médecine-Pharmacie, and Service de Bactériologie, Centre Hospitalier Universitaire, France
| | | | | | | | | | | | | |
Collapse
|
148
|
Péchiné S, Gleizes A, Janoir C, Gorges-Kergot R, Barc MC, Delmée M, Collignon A. Immunological properties of surface proteins of Clostridium difficile. J Med Microbiol 2005; 54:193-196. [PMID: 15673516 DOI: 10.1099/jmm.0.45800-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sera from patients with Clostridium difficile-associated disease (CDAD) and sera from a control group were analysed by an ELISA to detect antibodies directed against four surface proteins and toxins A and B of C. difficile. The surface proteins were the flagellar cap protein FliD, the flagellin FliC, the adhesin Cwp66 divided into two domains, Cwp66-Nterminal and Cwp66-Cterminal, and the fibronectin-binding protein Fbp68. For each antigen, antibody levels in the CDAD patient group and in the control group were compared. In the CDAD patient group, the mean of the antibody levels decreased from Cwp66-Cterminal to Fbp68, FliD, toxins B and A, Cwp66-Nterminal and finally FliC, suggesting different immunogenic properties among these adhesins. For Cwp66-Nterminal, FliC, FliD and Fbp68, the antibody level observed in the control group was higher than in the CDAD group with a statistically significant difference whereas the antibody level for toxins A and B was not statistically different. In conclusion, this study suggests that during the clinical course of disease, C. difficile adhesins are able to induce an immune response which could play a role in the defence mechanism of the host.
Collapse
Affiliation(s)
- Séverine Péchiné
- Université de Paris-Sud, Faculté de Pharmacie, Département de Microbiologie, 5 rue JB Clement, F-92296 Châtenay-Malabry cedex, France 2Université Catholique de Louvain, Faculté de Médecine, Unité de Microbiologie, Avenue Hippocrate 54.90, Bruxelles 1200, Belgium
| | - Aude Gleizes
- Université de Paris-Sud, Faculté de Pharmacie, Département de Microbiologie, 5 rue JB Clement, F-92296 Châtenay-Malabry cedex, France 2Université Catholique de Louvain, Faculté de Médecine, Unité de Microbiologie, Avenue Hippocrate 54.90, Bruxelles 1200, Belgium
| | - Claire Janoir
- Université de Paris-Sud, Faculté de Pharmacie, Département de Microbiologie, 5 rue JB Clement, F-92296 Châtenay-Malabry cedex, France 2Université Catholique de Louvain, Faculté de Médecine, Unité de Microbiologie, Avenue Hippocrate 54.90, Bruxelles 1200, Belgium
| | - Roseline Gorges-Kergot
- Université de Paris-Sud, Faculté de Pharmacie, Département de Microbiologie, 5 rue JB Clement, F-92296 Châtenay-Malabry cedex, France 2Université Catholique de Louvain, Faculté de Médecine, Unité de Microbiologie, Avenue Hippocrate 54.90, Bruxelles 1200, Belgium
| | - Marie-Claude Barc
- Université de Paris-Sud, Faculté de Pharmacie, Département de Microbiologie, 5 rue JB Clement, F-92296 Châtenay-Malabry cedex, France 2Université Catholique de Louvain, Faculté de Médecine, Unité de Microbiologie, Avenue Hippocrate 54.90, Bruxelles 1200, Belgium
| | - Michel Delmée
- Université de Paris-Sud, Faculté de Pharmacie, Département de Microbiologie, 5 rue JB Clement, F-92296 Châtenay-Malabry cedex, France 2Université Catholique de Louvain, Faculté de Médecine, Unité de Microbiologie, Avenue Hippocrate 54.90, Bruxelles 1200, Belgium
| | - Anne Collignon
- Université de Paris-Sud, Faculté de Pharmacie, Département de Microbiologie, 5 rue JB Clement, F-92296 Châtenay-Malabry cedex, France 2Université Catholique de Louvain, Faculté de Médecine, Unité de Microbiologie, Avenue Hippocrate 54.90, Bruxelles 1200, Belgium
| |
Collapse
|
149
|
Braunlin W, Xu Q, Hook P, Fitzpatrick R, Klinger JD, Burrier R, Kurtz CB. Toxin binding of tolevamer, a polyanionic drug that protects against antibiotic-associated diarrhea. Biophys J 2005; 87:534-9. [PMID: 15240486 PMCID: PMC1304374 DOI: 10.1529/biophysj.104.041277] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tolevamer, (GT160-246), is a sodium salt of styrene sulfonate polymer that is under development for the treatment of diarrhea caused by infection with Clostridium difficile. Pulsed ultrafiltration binding experiments in phosphate buffer containing 0.15 M Na(+) provide per polymer chain dissociation constants of 133 nM and 8.7 microM for the binding of tolevamer to C. difficile toxins A and B, respectively. At 0.05 M Na(+), the binding of toxin A to tolevamer is irreversible, whereas the dissociation constant to toxin B under these conditions is 120 nM. Binding constants obtained from fluorescence polarization data for toxin A binding to tolevamer at 0.15 M Na(+) agree substantially with those obtained by pulsed ultrafiltration. The binding activity of tolevamer reported here correlates well with previously reported results for the inhibition of the biological activity of C. difficile toxins A and B. From the fluorescence polarization data, it is estimated that one toxin A molecule interacts with between 600 to 1000 monomer units on tolevamer at 0.15 M Na(+). Thus, the data suggest a very large interaction surface between polymer and toxin A.
Collapse
|
150
|
Ozaki E, Kato H, Kita H, Karasawa T, Maegawa T, Koino Y, Matsumoto K, Takada T, Nomoto K, Tanaka R, Nakamura S. Clostridium difficile colonization in healthy adults: transient colonization and correlation with enterococcal colonization. J Med Microbiol 2004; 53:167-172. [PMID: 14729940 DOI: 10.1099/jmm.0.05376-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The aim of the present study was to investigate the colonization status of Clostridium difficile in healthy individuals. In total, 139 healthy adults from two study groups were examined at intervals of 3 months. Among the 18 positive subjects, the number of subjects from whom C. difficile was isolated once, twice, three times or four times was 10 (55.6%), three (16.7%), two (11.1%) and three (16.7%), respectively. In the student group, different subjects were colonized by different PCR ribotype/PFGE types. However, the same PCR ribotype/PFGE types of C. difficile were isolated from different subjects in the employee group, indicating that cross-transmission may have occurred in this group. Continuous colonization by the same PCR ribotype/PFGE type was only observed in three subjects. C. difficile-positive subjects were significantly more densely colonized by enterococci (P<0.05) than C. difficile-negative subjects: subjects that were found to be C. difficile-positive three or four times appeared to have higher concentrations of enterococci. The present results demonstrate that, although colonization by a C. difficile strain is transient in many cases, there are healthy individuals that are colonized persistently by C. difficile. They also suggest that dense colonization of the intestine by enterococci may be associated with C. difficile colonization.
Collapse
Affiliation(s)
- Eijiro Ozaki
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Haru Kato
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Hiroyuki Kita
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Tadahiro Karasawa
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Tsuneo Maegawa
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Youko Koino
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Kazumasa Matsumoto
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Toshihiko Takada
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Koji Nomoto
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Ryuichiro Tanaka
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Shinichi Nakamura
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan 2Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| |
Collapse
|