1
|
Finn LM, Cummer R, Castagner B, Keller BG. Allosterically switchable network orients β-flap in Clostridioides difficile toxins. Proc Natl Acad Sci U S A 2025; 122:e2419263122. [PMID: 40172960 PMCID: PMC12002228 DOI: 10.1073/pnas.2419263122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025] Open
Abstract
Allosteric proteins exhibit a functional response upon ligand binding far from the active site. Clostridioides difficile toxins use allosteric binding by the endogenous cofactor myo-inositol hexakisphosphate to orchestrate self-cleavage from within the target cell. This binding event induces a conformational shift, primarily effecting a lever-like β-flap region, with two known orientations. We uncovered a mechanism for this allosteric transition using extensive atomistic molecular dynamics simulations and computational and experimental mutagenesis. The mechanism relies on a switchable interaction network. The most prominent interaction pair is K600-E743, with K600 interactions explaining ∼70% of the allosteric effect. Rather than gradually morphing between two end states, the interaction network adopts two mutually exclusive configurations in the active and inactive state. Similar switchable networks may explain allostery more broadly. This mechanism in particular could aid in drug development targeting the C. difficile toxins autoproteolysis.
Collapse
Affiliation(s)
- Lauren M. Finn
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin14195, Germany
| | - Rebecca Cummer
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QCH3G 1Y6, Canada
| | - Bastien Castagner
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QCH3G 1Y6, Canada
| | - Bettina G. Keller
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin14195, Germany
| |
Collapse
|
2
|
Lee J, Goker S, Lim S, Hong CI. Development of circadian rhythms in mammalian systems. Biochem J 2024; 481:1967-1976. [PMID: 39714414 DOI: 10.1042/bcj20210060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
In mammals, molecular mechanisms of circadian rhythms involve a time-delayed negative feedback loop generating autonomous oscillations of ∼24 h. Most cell types in mammals possess circadian rhythms regulating temporal organization of cellular and physiological processes. Intriguingly, pluripotent stem cells do not possess circadian rhythms and oscillations arise after a defined period of differentiation. Previous studies demonstrated that post-transcriptional regulations of core clock components, CLOCK and PER2, play critical roles in inducing circadian rhythms. In this article, we review the development of circadian rhythms in mammalian systems and provide a theoretical understanding of potential mechanisms regulating the birth of circadian rhythms using mathematical modeling.
Collapse
Affiliation(s)
- Junghyun Lee
- Department of Mathematical Sciences, University of Cincinnati, Cincinnati, OH, U.S.A
| | - Sevde Goker
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, U.S.A
| | - Sookkyung Lim
- Department of Mathematical Sciences, University of Cincinnati, Cincinnati, OH, U.S.A
| | - Christian I Hong
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, U.S.A
| |
Collapse
|
3
|
Matylitsky J, Krieg A, Schumacher J, Borho J, Barth H, Papatheodorou P. Inhibition of Clostridioides difficile toxins TcdA and TcdB by the amiodarone derivative dronedarone. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9877-9885. [PMID: 38935126 PMCID: PMC11582217 DOI: 10.1007/s00210-024-03248-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
The dreaded nosocomial pathogen Clostridioides difficile causes diarrhea and severe inflammation of the colon, especially after the use of certain antibiotics. The bacterium releases two deleterious toxins, TcdA and TcdB, into the gut, which are mainly responsible for the symptoms of C. difficile-associated diseases (CDADs). Both toxins are capable of entering independently into various host cells, e.g., intestinal epithelial cells, where they mono-O-glucosylate and inactivate Rho and/or Ras GTPases, important molecular switches for various cellular functions. We have shown recently that the cellular uptake of the Clostridioides difficile toxins TcdA and TcdB (TcdA/B) is inhibited by the licensed class III antiarrhythmic drug amiodarone (Schumacher et al. in Gut Microbes 15(2):2256695, 2023). Mechanistically, amiodarone delays the cellular uptake of both toxins into target cells most likely by lowering membrane cholesterol levels and by interfering with membrane insertion and/or pore formation of TcdA/B. However, serious side effects, such as thyroid dysfunction and severe pulmonary fibrosis, limit the clinical use of amiodarone in patients with C. difficile infection (CDI). For that reason, we aimed to test whether dronedarone, an amiodarone derivative with a more favorable side effect profile, is also capable of inhibiting TcdA/B. To this end, we tested in vitro with various methods the impact of dronedarone on the intoxication of Vero and CaCo-2 cells with TcdA/B. Importantly, preincubation of both cell lines with dronedarone for 1 h at concentrations in the low micromolar range rendered the cells less sensitive toward TcdA/B-induced Rac1 glucosylation, collapse of the actin cytoskeleton, cell rounding, and cytopathic effects, respectively. Our study points toward the possibility of repurposing the already approved drug dronedarone as the preferable safer-to-use alternative to amiodarone for inhibiting TcdA/B in the (supportive) therapy of CDADs.
Collapse
Affiliation(s)
- Jauheni Matylitsky
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Anica Krieg
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Judith Schumacher
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Joscha Borho
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Panagiotis Papatheodorou
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
4
|
Lietz S, Sokolowski LM, Barth H, Ernst K. Alpha-1 antitrypsin inhibits Clostridium botulinum C2 toxin, Corynebacterium diphtheriae diphtheria toxin and B. anthracis fusion toxin. Sci Rep 2024; 14:21257. [PMID: 39261531 PMCID: PMC11390955 DOI: 10.1038/s41598-024-71706-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
The bacterium Clostridium botulinum, well-known for producing botulinum neurotoxins, which cause the severe paralytic illness known as botulism, produces C2 toxin, a binary AB-toxin with ADP-ribosyltranferase activity. C2 toxin possesses two separate protein components, an enzymatically active A-component C2I and the binding and translocation B-component C2II. After proteolytic activation of C2II to C2IIa, the heptameric structure binds C2I and is taken up via receptor-mediated endocytosis into the target cells. Due to acidification of endosomes, the C2IIa/C2I complex undergoes conformational changes and consequently C2IIa forms a pore into the endosomal membrane and C2I can translocate into the cytoplasm, where it ADP-ribosylates G-actin, a key component of the cytoskeleton. This modification disrupts the actin cytoskeleton, resulting in the collapse of cytoskeleton and ultimately cell death. Here, we show that the serine-protease inhibitor α1-antitrypsin (α1AT) which we identified previously from a hemofiltrate library screen for PT from Bordetella pertussis is a multitoxin inhibitor. α1AT inhibits intoxication of cells with C2 toxin via inhibition of binding to cells and inhibition of enzyme activity of C2I. Moreover, diphtheria toxin and an anthrax fusion toxin are inhibited by α1AT. Since α1AT is commercially available as a drug for treatment of the α1AT deficiency, it could be repurposed for treatment of toxin-mediated diseases.
Collapse
Affiliation(s)
- Stefanie Lietz
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081, Ulm, Germany
| | - Lena-Marie Sokolowski
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081, Ulm, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081, Ulm, Germany.
| | - Katharina Ernst
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081, Ulm, Germany.
| |
Collapse
|
5
|
Kempher ML, Shadid TM, Larabee JL, Ballard JD. A sequence invariable region in TcdB2 is required for toxin escape from Clostridioides difficile. J Bacteriol 2024; 206:e0009624. [PMID: 38888328 PMCID: PMC11323933 DOI: 10.1128/jb.00096-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Sequence differences among the subtypes of Clostridioides difficile toxin TcdB (2,366 amino acids) are broadly distributed across the entire protein, with the notable exception of 76 residues at the protein's carboxy terminus. This sequence invariable region (SIR) is identical at the DNA and protein level among the TcdB variants, suggesting this string of amino acids has undergone selective pressure to prevent alterations. The functional role of the SIR domain in TcdB has not been determined. Analysis of a recombinantly constructed TcdB mutant lacking the SIR domain did not identify changes in TcdB's enzymatic or cytopathic activities. To further assess the SIR region, we constructed a C. difficile strain with the final 228 bp deleted from the tcdB gene, resulting in the production of a truncated form of TcdB lacking the SIR (TcdB2∆2291-2366). Using a combination of approaches, we found in the absence of the SIR sequence TcdB2∆2291-2366 retained cytotoxic activity but was not secreted from C. difficile. TcdB2∆2291-2366 was not released from the cell under autolytic conditions, indicating the SIR is involved in a more discrete step in toxin escape from the bacterium. Fractionation experiments combined with antibody detection found that TcdB2∆2291-2366 accumulates at the cell membrane but is unable to complete steps in secretion beyond this point. These data suggest conservation of the SIR domain across variants of TcdB could be influenced by the sequence's role in efficient escape of the toxin from C. difficile. IMPORTANCE Clostridioides difficile is a leading cause of antibiotic associated disease in the United States. The primary virulence factors produced by C. difficile are two large glucosylating toxins TcdA and TcdB. To date, several sequence variants of TcdB have been identified that differ in various functional properties. Here, we identified a highly conserved region among TcdB subtypes that is required for release of the toxin from C. difficile. This study reveals a putative role for the longest stretch of invariable sequence among TcdB subtypes and provides new details regarding toxin release into the extracellular environment. Improving our understanding of the functional roles of the conserved regions of TcdB variants aids in the development of new, broadly applicable strategies to treat CDI.
Collapse
Affiliation(s)
- Megan L. Kempher
- Department of
Microbiology and Immunology, University of Oklahoma Health Sciences
Center, Oklahoma City,
Oklahoma, USA
- Department of
Chemistry and Biochemistry, University of
Oklahoma, Norman,
Oklahoma, USA
| | - Tyler M. Shadid
- Department of
Microbiology and Immunology, University of Oklahoma Health Sciences
Center, Oklahoma City,
Oklahoma, USA
| | - Jason L. Larabee
- Department of
Microbiology and Immunology, University of Oklahoma Health Sciences
Center, Oklahoma City,
Oklahoma, USA
| | - Jimmy D. Ballard
- Department of
Microbiology and Immunology, University of Oklahoma Health Sciences
Center, Oklahoma City,
Oklahoma, USA
| |
Collapse
|
6
|
Pourliotopoulou E, Karampatakis T, Kachrimanidou M. Exploring the Toxin-Mediated Mechanisms in Clostridioides difficile Infection. Microorganisms 2024; 12:1004. [PMID: 38792835 PMCID: PMC11124097 DOI: 10.3390/microorganisms12051004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Clostridioides difficile infection (CDI) is the leading cause of nosocomial antibiotic-associated diarrhea, and colitis, with increasing incidence and healthcare costs. Its pathogenesis is primarily driven by toxins produced by the bacterium C. difficile, Toxin A (TcdA) and Toxin B (TcdB). Certain strains produce an additional toxin, the C. difficile transferase (CDT), which further enhances the virulence and pathogenicity of C. difficile. These toxins disrupt colonic epithelial barrier integrity, and induce inflammation and cellular damage, leading to CDI symptoms. Significant progress has been made in the past decade in elucidating the molecular mechanisms of TcdA, TcdB, and CDT, which provide insights into the management of CDI and the future development of novel treatment strategies based on anti-toxin therapies. While antibiotics are common treatments, high recurrence rates necessitate alternative therapies. Bezlotoxumab, targeting TcdB, is the only available anti-toxin, yet limitations persist, prompting ongoing research. This review highlights the current knowledge of the structure and mechanism of action of C. difficile toxins and their role in disease. By comprehensively describing the toxin-mediated mechanisms, this review provides insights for the future development of novel treatment strategies and the management of CDI.
Collapse
Affiliation(s)
- Evdokia Pourliotopoulou
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | | | - Melania Kachrimanidou
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| |
Collapse
|
7
|
Papatheodorou P, Minton NP, Aktories K, Barth H. An Updated View on the Cellular Uptake and Mode-of-Action of Clostridioides difficile Toxins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1435:219-247. [PMID: 38175478 DOI: 10.1007/978-3-031-42108-2_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Research on the human gut pathogen Clostridioides (C.) difficile and its toxins continues to attract much attention as a consequence of the threat to human health posed by hypervirulent strains. Toxin A (TcdA) and Toxin B (TcdB) are the two major virulence determinants of C. difficile. Both are single-chain proteins with a similar multidomain architecture. Certain hypervirulent C. difficile strains also produce a third toxin, namely binary toxin CDT (C. difficile transferase). C. difficile toxins are the causative agents of C. difficile-associated diseases (CDADs), such as antibiotics-associated diarrhea and pseudomembranous colitis. For that reason, considerable efforts have been expended to unravel their molecular mode-of-action and the cellular mechanisms responsible for their uptake. Many of these studies have been conducted in European laboratories. Here, we provide an update on our previous review (Papatheodorou et al. Adv Exp Med Biol, 2018) on important advances in C. difficile toxins research.
Collapse
Affiliation(s)
- Panagiotis Papatheodorou
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany.
| | - Nigel P Minton
- BBSRC/EPSRC Synthetic Biology Research Centre, University of Nottingham, Nottingham, UK
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
8
|
Tannoo RM, Richert L, Koschut D, Tomishige N, Treffert SM, Kobayashi T, Mély Y, Orian-Rousseau V. Quantitative live imaging reveals a direct interaction between CD44v6 and MET in membrane domains upon activation with both MET ligands, HGF and internalin B. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184236. [PMID: 37793560 DOI: 10.1016/j.bbamem.2023.184236] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
Deregulation of the receptor tyrosine kinase MET/hepatocyte growth factor (HGF) pathway results in several pathological processes involved in tumor progression and metastasis. In a different context, MET can serve as an entry point for the bacterium Listeria monocytogenes, when activated by the internalin B (InlB) protein during infection of non-phagocytic cells. We have previously demonstrated that MET requires CD44v6 for its ligand-induced activation. However, the stoichiometry and the steps required for the formation of this complex, are still unknown. In this work, we studied the dynamics of the ligand-induced interaction of CD44v6 with MET at the plasma membrane. Using Förster resonance energy transfer-based fluorescence lifetime imaging microscopy in T-47D cells, we evidenced a direct interaction between MET and CD44v6 promoted by HGF and InlB in live cells. In the absence of MET, fluorescence correlation spectroscopy experiments further showed the dimerization of CD44v6 and the increase of its diffusion induced by HGF and InlB. In the presence of MET, stimulation of the cells by HGF or InlB significantly decreased the diffusion of CD44v6, in line with the formation of a ternary complex of MET with CD44v6 and HGF/InlB. Finally, similarly to HGF/InlB, disruption of liquid-ordered domains (Lo) by methyl-β-cyclodextrin increased CD44v6 mobility suggesting that these factors induce the exit of CD44v6 from the Lo domains. Our data led us to propose a model for MET activation, where CD44v6 dimerizes and diffuses rapidly out of Lo domains to form an oligomeric MET/ligand/CD44v6 complex that is instrumental for MET activation.
Collapse
Affiliation(s)
- Ryshtee Mary Tannoo
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France; Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany
| | - Ludovic Richert
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France.
| | - David Koschut
- Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany; Disease Intervention Technology Lab (DITL), Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Nario Tomishige
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France
| | - Sven Máté Treffert
- Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany
| | - Toshihide Kobayashi
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France
| | - Yves Mély
- Laboratory of Bioimaging and Pathologies (LBP), University of Strasbourg (UNISTRA), France.
| | - Véronique Orian-Rousseau
- Institute of Biological and Chemical systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Germany.
| |
Collapse
|
9
|
Buddle JE, Fagan RP. Pathogenicity and virulence of Clostridioides difficile. Virulence 2023; 14:2150452. [PMID: 36419222 DOI: 10.1080/21505594.2022.2150452] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Clostridioides difficile is the most common cause of nosocomial antibiotic-associated diarrhea, and is responsible for a spectrum of diseases characterized by high levels of recurrence, morbidity, and mortality. Treatment is complex, since antibiotics constitute both the main treatment and the major risk factor for infection. Worryingly, resistance to multiple antibiotics is becoming increasingly widespread, leading to the classification of this pathogen as an urgent threat to global health. As a consummate opportunist, C. difficile is well equipped for promoting disease, owing to its arsenal of virulence factors: transmission of this anaerobe is highly efficient due to the formation of robust endospores, and an array of adhesins promote gut colonization. C. difficile produces multiple toxins acting upon gut epithelia, resulting in manifestations typical of diarrheal disease, and severe inflammation in a subset of patients. This review focuses on such virulence factors, as well as the importance of antimicrobial resistance and genome plasticity in enabling pathogenesis and persistence of this important pathogen.
Collapse
Affiliation(s)
- Jessica E Buddle
- Molecular Microbiology, School of Biosciences, University of Sheffield, Sheffield, UK
| | - Robert P Fagan
- Molecular Microbiology, School of Biosciences, University of Sheffield, Sheffield, UK
| |
Collapse
|
10
|
Schumacher J, Nienhaus A, Heber S, Matylitsky J, Chaves-Olarte E, Rodríguez C, Barth H, Papatheodorou P. Exploring the inhibitory potential of the antiarrhythmic drug amiodarone against Clostridioides difficile toxins TcdA and TcdB. Gut Microbes 2023; 15:2256695. [PMID: 37749884 PMCID: PMC10524773 DOI: 10.1080/19490976.2023.2256695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/05/2023] [Indexed: 09/27/2023] Open
Abstract
The intestinal pathogen Clostridioides difficile is the leading cause of antibiotic-associated diarrhea and pseudomembranous colitis in humans. The symptoms of C. difficile-associated diseases (CDADs) are directly associated with the pathogen's toxins TcdA and TcdB, which enter host cells and inactivate Rho and/or Ras GTPases by glucosylation. Membrane cholesterol is crucial during the intoxication process of TcdA and TcdB, and likely involved during pore formation of both toxins in endosomal membranes, a key step after cellular uptake for the translocation of the glucosyltransferase domain of both toxins from endosomes into the host cell cytosol. The licensed drug amiodarone, a multichannel blocker commonly used in the treatment of cardiac dysrhythmias, is also capable of inhibiting endosomal acidification and, as shown recently, cholesterol biosynthesis. Thus, we were keen to investigate in vitro with cultured cells and human intestinal organoids, whether amiodarone preincubation protects from TcdA and/or TcdB intoxication. Amiodarone conferred protection against both toxins independently and in combination as well as against toxin variants from the clinically relevant, epidemic C. difficile strain NAP1/027. Further mechanistic studies suggested that amiodarone's mode-of-inhibition involves also interference with the translocation pore of both toxins. Our study opens the possibility of repurposing the licensed drug amiodarone as a novel pan-variant antitoxin therapeutic in the context of CDADs.
Collapse
Affiliation(s)
- Judith Schumacher
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Astrid Nienhaus
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Sebastian Heber
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Jauheni Matylitsky
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - César Rodríguez
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Panagiotis Papatheodorou
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
11
|
Childress KO, Cencer CS, Tyska MJ, Lacy DB. Nectin-3 and shed forms of CSPG4 can serve as epithelial cell receptors for Clostridioides difficile TcdB. mBio 2023; 14:e0185723. [PMID: 37747247 PMCID: PMC10653914 DOI: 10.1128/mbio.01857-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 09/26/2023] Open
Abstract
IMPORTANCE Toxin B (TcdB) is a major virulence factor of Clostridioides difficile, a Gram-positive pathogen that is a leading cause of hospital-acquired diarrhea. While previous studies have established that TcdB can engage multiple cell surface receptors in vitro, little is known about how these interactions promote disease and where these receptors localize on colonic tissue. Here, we used immunofluorescence microscopy to visualize Nectin-3 and CSPG4 on tissue, revealing unexpected localization of both receptors on colonic epithelial cells. We show that Nectin-3, which was previously characterized as an adherens junction protein, is also localized to the brush border of colonocytes. Staining for CSPG4 revealed that it is present along epithelial cell junctions, suggesting that it is shed by fibroblasts along the crypt-surface axis. Collectively, our study provides new insights into how TcdB can gain access to the receptors Nectin-3 and CSPG4 to intoxicate colonic epithelial cells.
Collapse
Affiliation(s)
- Kevin O. Childress
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Caroline S. Cencer
- Department of Cell and Development Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew J. Tyska
- Department of Cell and Development Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - D. Borden Lacy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
12
|
Prischich D, Camarero N, Encinar del Dedo J, Cambra-Pellejà M, Prat J, Nevola L, Martín-Quirós A, Rebollo E, Pastor L, Giralt E, Geli MI, Gorostiza P. Light-dependent inhibition of clathrin-mediated endocytosis in yeast unveils conserved functions of the AP2 complex. iScience 2023; 26:107899. [PMID: 37766990 PMCID: PMC10520943 DOI: 10.1016/j.isci.2023.107899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Clathrin-mediated endocytosis (CME) is an essential cellular process, conserved among eukaryotes. Yeast constitutes a powerful genetic model to dissect the complex endocytic machinery, yet there is a lack of specific pharmacological agents to interfere with CME in these organisms. TL2 is a light-regulated peptide inhibitor targeting the AP2-β-adaptin/β-arrestin interaction and that can photocontrol CME with high spatiotemporal precision in mammalian cells. Here, we study endocytic protein dynamics by live-cell imaging of the fluorescently tagged coat-associated protein Sla1-GFP, demonstrating that TL2 retains its inhibitory activity in S. cerevisiae spheroplasts. This is despite the β-adaptin/β-arrestin interaction not being conserved in yeast. Our data indicate that the AP2 α-adaptin is the functional target of activated TL2. We identified as interacting partners for the α-appendage, the Eps15 and epsin homologues Ede1 and Ent1. This demonstrates that endocytic cargo loading and sensing can be executed by conserved molecular interfaces, regardless of the proteins involved.
Collapse
Affiliation(s)
- Davia Prischich
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red – Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Núria Camarero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red – Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Javier Encinar del Dedo
- Department of Cell Biology, Institute for Molecular Biology of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Maria Cambra-Pellejà
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Judit Prat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Laura Nevola
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Andrés Martín-Quirós
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Institute for Molecular Biology of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Laura Pastor
- Department of Cell Biology, Institute for Molecular Biology of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Inorganic and Organic Chemistry, University of Barcelona (UB), Barcelona, Spain
| | - María Isabel Geli
- Department of Cell Biology, Institute for Molecular Biology of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red – Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
13
|
Braune-Yan M, Jia J, Wahba M, Schmid J, Papatheodorou P, Barth H, Ernst K. Domperidone Protects Cells from Intoxication with Clostridioides difficile Toxins by Inhibiting Hsp70-Assisted Membrane Translocation. Toxins (Basel) 2023; 15:384. [PMID: 37368685 DOI: 10.3390/toxins15060384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Clostridioides difficile infections cause severe symptoms ranging from diarrhea to pseudomembranous colitis due to the secretion of AB-toxins, TcdA and TcdB. Both toxins are taken up into cells through receptor-mediated endocytosis, autoproteolytic processing and translocation of their enzyme domains from acidified endosomes into the cytosol. The enzyme domains glucosylate small GTPases such as Rac1, thereby inhibiting processes such as actin cytoskeleton regulation. Here, we demonstrate that specific pharmacological inhibition of Hsp70 activity protected cells from TcdB intoxication. In particular, the established inhibitor VER-155008 and the antiemetic drug domperidone, which was found to be an Hsp70 inhibitor, reduced the number of cells with TcdB-induced intoxication morphology in HeLa, Vero and intestinal CaCo-2 cells. These drugs also decreased the intracellular glucosylation of Rac1 by TcdB. Domperidone did not inhibit TcdB binding to cells or enzymatic activity but did prevent membrane translocation of TcdB's glucosyltransferase domain into the cytosol. Domperidone also protected cells from intoxication with TcdA as well as CDT toxin produced by hypervirulent strains of Clostridioides difficile. Our results reveal Hsp70 requirement as a new aspect of the cellular uptake mechanism of TcdB and identified Hsp70 as a novel drug target for potential therapeutic strategies required to combat severe Clostridioides difficile infections.
Collapse
Affiliation(s)
- Maria Braune-Yan
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jinfang Jia
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081 Ulm, Germany
| | - Mary Wahba
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081 Ulm, Germany
| | - Johannes Schmid
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081 Ulm, Germany
| | - Panagiotis Papatheodorou
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081 Ulm, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081 Ulm, Germany
| | - Katharina Ernst
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
14
|
Godbold GD, Hewitt FC, Kappell AD, Scholz MB, Agar SL, Treangen TJ, Ternus KL, Sandbrink JB, Koblentz GD. Improved understanding of biorisk for research involving microbial modification using annotated sequences of concern. Front Bioeng Biotechnol 2023; 11:1124100. [PMID: 37180048 PMCID: PMC10167326 DOI: 10.3389/fbioe.2023.1124100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
Regulation of research on microbes that cause disease in humans has historically been focused on taxonomic lists of 'bad bugs'. However, given our increased knowledge of these pathogens through inexpensive genome sequencing, 5 decades of research in microbial pathogenesis, and the burgeoning capacity of synthetic biologists, the limitations of this approach are apparent. With heightened scientific and public attention focused on biosafety and biosecurity, and an ongoing review by US authorities of dual-use research oversight, this article proposes the incorporation of sequences of concern (SoCs) into the biorisk management regime governing genetic engineering of pathogens. SoCs enable pathogenesis in all microbes infecting hosts that are 'of concern' to human civilization. Here we review the functions of SoCs (FunSoCs) and discuss how they might bring clarity to potentially problematic research outcomes involving infectious agents. We believe that annotation of SoCs with FunSoCs has the potential to improve the likelihood that dual use research of concern is recognized by both scientists and regulators before it occurs.
Collapse
Affiliation(s)
| | | | | | | | - Stacy L. Agar
- Signature Science, LLC, Charlottesville, VA, United States
| | - Todd J. Treangen
- Department of Computer Science, Rice University, Houston, TX, United States
| | | | - Jonas B. Sandbrink
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Gregory D. Koblentz
- Schar School of Policy and Government, George Mason University, Arlington, VA, United States
| |
Collapse
|
15
|
Rosselot AE, Park M, Kim M, Matsu‐Ura T, Wu G, Flores DE, Subramanian KR, Lee S, Sundaram N, Broda TR, McCauley HA, Hawkins JA, Chetal K, Salomonis N, Shroyer NF, Helmrath MA, Wells JM, Hogenesch JB, Moore SR, Hong CI. Ontogeny and function of the circadian clock in intestinal organoids. EMBO J 2022; 41:e106973. [PMID: 34704277 PMCID: PMC8762567 DOI: 10.15252/embj.2020106973] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Circadian rhythms regulate diverse aspects of gastrointestinal physiology ranging from the composition of microbiota to motility. However, development of the intestinal circadian clock and detailed mechanisms regulating circadian physiology of the intestine remain largely unknown. In this report, we show that both pluripotent stem cell-derived human intestinal organoids engrafted into mice and patient-derived human intestinal enteroids possess circadian rhythms and demonstrate circadian phase-dependent necrotic cell death responses to Clostridium difficile toxin B (TcdB). Intriguingly, mouse and human enteroids demonstrate anti-phasic necrotic cell death responses to TcdB. RNA-Seq analysis shows that ~3-10% of the detectable transcripts are rhythmically expressed in mouse and human enteroids. Remarkably, we observe anti-phasic gene expression of Rac1, a small GTPase directly inactivated by TcdB, between mouse and human enteroids, and disruption of Rac1 abolishes clock-dependent necrotic cell death responses. Our findings uncover robust functions of circadian rhythms regulating clock-controlled genes in both mouse and human enteroids governing organism-specific, circadian phase-dependent necrotic cell death responses, and lay a foundation for human organ- and disease-specific investigation of clock functions using human organoids for translational applications.
Collapse
Affiliation(s)
- Andrew E Rosselot
- Department of Pharmacology & Systems PhysiologyUniversity of CincinnatiCincinnatiOHUSA
| | - Miri Park
- Department of Pharmacology & Systems PhysiologyUniversity of CincinnatiCincinnatiOHUSA
| | - Mari Kim
- Department of Pharmacology & Systems PhysiologyUniversity of CincinnatiCincinnatiOHUSA
| | - Toru Matsu‐Ura
- Department of Pharmacology & Systems PhysiologyUniversity of CincinnatiCincinnatiOHUSA
| | - Gang Wu
- Division of Human Genetics and ImmunobiologyCenter for ChronobiologyDepartment of PediatricsCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Danilo E Flores
- Division of Human Genetics and ImmunobiologyCenter for ChronobiologyDepartment of PediatricsCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | | | - Suengwon Lee
- Department of Pharmacology & Systems PhysiologyUniversity of CincinnatiCincinnatiOHUSA
| | - Nambirajan Sundaram
- Department of Pediatric SurgeryCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Taylor R Broda
- Center for Stem Cell and Organoid MedicineDivision of Developmental BiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Heather A McCauley
- Center for Stem Cell and Organoid MedicineDivision of Developmental BiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Jennifer A Hawkins
- Department of Pediatric SurgeryCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Kashish Chetal
- Division of Biomedical InformaticsCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Nathan Salomonis
- Division of Biomedical InformaticsCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Noah F Shroyer
- Gastroenterology and HepatologyBaylor College of MedicineHoustonTXUSA
| | - Michael A Helmrath
- Department of Pediatric SurgeryCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
- Center for Stem Cell and Organoid MedicineDivision of Developmental BiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - James M Wells
- Center for Stem Cell and Organoid MedicineDivision of Developmental BiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
- Division of EndocrinologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - John B Hogenesch
- Division of Human Genetics and ImmunobiologyCenter for ChronobiologyDepartment of PediatricsCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
- Center for ChronobiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| | - Sean R Moore
- Division of Pediatric Gastroenterology, Hepatology, and NutritionDepartment of PediatricsUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Christian I Hong
- Department of Pharmacology & Systems PhysiologyUniversity of CincinnatiCincinnatiOHUSA
- Center for Stem Cell and Organoid MedicineDivision of Developmental BiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
- Center for ChronobiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
- Division of Developmental BiologyCincinnati Children’s Hospital Medical CenterCincinnatiOHUSA
| |
Collapse
|
16
|
Chen B, Perry K, Jin R. Neutralizing epitopes on Clostridioides difficile toxin A revealed by the structures of two camelid VHH antibodies. Front Immunol 2022; 13:978858. [PMID: 36466927 PMCID: PMC9709291 DOI: 10.3389/fimmu.2022.978858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022] Open
Abstract
Toxin A (TcdA) and toxin B (TcdB) are two key virulence factors secreted by Clostridioides difficile, which is listed as an urgent threat by the CDC. These two large homologous exotoxins are mainly responsible for diseases associated with C. difficile infection (CDI) with symptoms ranging from diarrhea to life threatening pseudomembranous colitis. Single-domain camelid antibodies (VHHs) AH3 and AA6 are two potent antitoxins against TcdA, which when combined with two TcdB-targeting VHHs showed effective protection against both primary and recurrent CDI in animal models. Here, we report the co-crystal structures of AH3 and AA6 when they form complexes with the glucosyltransferase domain (GTD) and a fragment of the delivery and receptor-binding domain (DRBD) of TcdA, respectively. Based on these structures, we find that AH3 binding enhances the overall stability of the GTD and interferes with its unfolding at acidic pH, and AA6 may inhibit the pH-dependent conformational changes in the DRBD that is necessary for pore formation of TcdA. These studies reveal two functionally critical epitopes on TcdA and shed new insights into neutralizing mechanisms and potential development of epitope-focused vaccines against TcdA.
Collapse
Affiliation(s)
- Baohua Chen
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Kay Perry
- NE-CAT, Advanced Photon Source, Argonne National Laboratory, Argonne, IL, United States,Department of Chemistry and Chemical Biology, Cornell University, Argonne, IL, United States
| | - Rongsheng Jin
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States,*Correspondence: Rongsheng Jin,
| |
Collapse
|
17
|
Plakoglobin and High-Mobility Group Box 1 Mediate Intestinal Epithelial Cell Apoptosis Induced by Clostridioides difficile TcdB. mBio 2022; 13:e0184922. [PMID: 36043787 DOI: 10.1128/mbio.01849-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clostridioides difficile infection (CDI) is the leading cause of antibiotic-associated intestinal disease, resulting in severe diarrhea and fatal pseudomembranous colitis. TcdB, one of the essential virulence factors secreted by this bacterium, induces host cell apoptosis through a poorly understood mechanism. Here, we performed an RNA interference (RNAi) screen customized to Caco-2 cells, a cell line model of the intestinal epithelium, to discover host factors involved in TcdB-induced apoptosis. We identified plakoglobin, also known as junction plakoglobin (JUP) or γ-catenin, a member of the catenin family, as a novel host factor and a previously known cell death-related chromatin factor, high-mobility group box 1 (HMGB1). Disruption of those host factors by RNAi and CRISPR resulted in resistance of cells to TcdB-mediated and mitochondrion-dependent apoptosis. JUP was redistributed from adherens junctions to the mitochondria and colocalized with the antiapoptotic factor Bcl-XL. JUP proteins could permeabilize the mitochondrial membrane, resulting in the release of cytochrome c. Our results reveal a novel role of JUP in targeting the mitochondria to promote the mitochondrial apoptotic pathway. Treatment with glycyrrhizin, an HMGB1 inhibitor, resulted in significantly increased resistance to TcdB-induced epithelial damage in cultured cells and a mouse ligated colon loop model. These findings demonstrate the critical roles of JUP and HMGB1 in TcdB-induced epithelial cell apoptosis. IMPORTANCE Clostridioides difficile infection (CDI) is the leading cause of hospital-acquired diarrhea. Toxins, especially TcdB, cause epithelial cell apoptosis, but the underlying cell death mechanism is less clear. Through an apoptosis-focused RNAi screen using a bacterium-made small interfering (siRNA) library customized to a human colonic epithelial cell model, we found a novel host factor, plakoglobin (γ-catenin), as a key factor required for cell apoptosis induced by TcdB. Plakoglobin targets and permeabilizes mitochondria after stimulation by TcdB, demonstrating a hitherto underappreciated role of this catenin family member in the apoptosis of intestinal epithelial cells. We also found a previously known cell death-related chromatin factor, HMGB1, and explored the inhibition of HMGB1 for CDI therapy in vivo.
Collapse
|
18
|
Chen B, Liu Z, Perry K, Jin R. Structure of the glucosyltransferase domain of TcdA in complex with RhoA provides insights into substrate recognition. Sci Rep 2022; 12:9028. [PMID: 35637242 PMCID: PMC9151644 DOI: 10.1038/s41598-022-12909-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/17/2022] [Indexed: 01/05/2023] Open
Abstract
Clostridioides difficile is one of the most common causes of antibiotic-associated diarrhea in developed countries. As key virulence factors of C. difficile, toxin A (TcdA) and toxin B (TcdB) act by glucosylating and inactivating Rho and Ras family small GTPases in host cells, which leads to actin cytoskeleton disruption, cell rounding, and ultimately cell death. Here we present the co-crystal structure of the glucosyltransferase domain (GTD) of TcdA in complex with its substrate human RhoA at 2.60-angstrom resolution. This structure reveals that TcdA GTD grips RhoA mainly through its switch I and switch II regions, which is complemented by interactions involving RhoA's pre-switch I region. Comprehensive structural comparisons between the TcdA GTD-RhoA complex and the structures of TcdB GTD in complex with Cdc42 and R-Ras reveal both the conserved and divergent features of these two toxins in terms of substrate recognition. Taken together, these findings establish the structural basis for TcdA recognition of small GTPases and advance our understanding of the substrates selectivity of large clostridial toxins.
Collapse
Affiliation(s)
- Baohua Chen
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Zheng Liu
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Argonne National Laboratory, Cornell University, Argonne, IL, 60439, USA
| | - Rongsheng Jin
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
19
|
Kordus SL, Thomas AK, Lacy DB. Clostridioides difficile toxins: mechanisms of action and antitoxin therapeutics. Nat Rev Microbiol 2022; 20:285-298. [PMID: 34837014 PMCID: PMC9018519 DOI: 10.1038/s41579-021-00660-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 01/03/2023]
Abstract
Clostridioides difficile is a Gram-positive anaerobe that can cause a spectrum of disorders that range in severity from mild diarrhoea to fulminant colitis and/or death. The bacterium produces up to three toxins, which are considered the major virulence factors in C. difficile infection. These toxins promote inflammation, tissue damage and diarrhoea. In this Review, we highlight recent biochemical and structural advances in our understanding of the mechanisms that govern host-toxin interactions. Understanding how C. difficile toxins affect the host forms a foundation for developing novel strategies for treatment and prevention of C. difficile infection.
Collapse
Affiliation(s)
- Shannon L. Kordus
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Structural Biology, Vanderbilt University, Nashville, TN, USA,These authors contributed equally: Shannon L. Kordus, Audrey K. Thomas
| | - Audrey K. Thomas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Structural Biology, Vanderbilt University, Nashville, TN, USA,These authors contributed equally: Shannon L. Kordus, Audrey K. Thomas
| | - D. Borden Lacy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA,Center for Structural Biology, Vanderbilt University, Nashville, TN, USA,The Veterans Affairs, Tennessee Valley Healthcare, System, Nashville, TN, USA,
| |
Collapse
|
20
|
Chen B, Basak S, Chen P, Zhang C, Perry K, Tian S, Yu C, Dong M, Huang L, Bowen ME, Jin R. Structure and conformational dynamics of Clostridioides difficile toxin A. Life Sci Alliance 2022; 5:5/6/e202201383. [PMID: 35292538 PMCID: PMC8924006 DOI: 10.26508/lsa.202201383] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 01/05/2023] Open
Abstract
This study presents a complete structural model of TcdA holotoxin and sheds new lights into the conformational dynamics of TcdA and its roles in TcdA intoxication. Clostridioides difficile toxin A and B (TcdA and TcdB) are two major virulence factors responsible for diseases associated with C. difficile infection (CDI). Here, we report the 3.18-Å resolution crystal structure of a TcdA fragment (residues L843–T2481), which advances our understanding of the complete structure of TcdA holotoxin. Our structural analysis, together with complementary single molecule FRET and limited proteolysis studies, reveal that TcdA adopts a dynamic structure and its CROPs domain can sample a spectrum of open and closed conformations in a pH-dependent manner. Furthermore, a small globular subdomain (SGS) and the CROPs protect the pore-forming region of TcdA in the closed state at neutral pH, which could contribute to modulating the pH-dependent pore formation of TcdA. A rationally designed TcdA mutation that trapped the CROPs in the closed conformation showed drastically reduced cytotoxicity. Taken together, these studies shed new lights into the conformational dynamics of TcdA and its roles in TcdA intoxication.
Collapse
Affiliation(s)
- Baohua Chen
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Sujit Basak
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Peng Chen
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Changcheng Zhang
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, IL, USA
| | - Songhai Tian
- Department of Urology, Boston Children's Hospital, Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Clinton Yu
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Lan Huang
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Mark E Bowen
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Rongsheng Jin
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
21
|
Papatheodorou P, Kindig S, Badilla-Lobo A, Fischer S, Durgun E, Thuraisingam T, Witte A, Song S, Aktories K, Chaves-Olarte E, Rodríguez C, Barth H. The Compound U18666A Inhibits the Intoxication of Cells by Clostridioides difficile Toxins TcdA and TcdB. Front Microbiol 2021; 12:784856. [PMID: 34912322 PMCID: PMC8667575 DOI: 10.3389/fmicb.2021.784856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
The intestinal pathogen Clostridioides (C.) difficile is a major cause of diarrhea both in hospitals and outpatient in industrialized countries. This bacterium produces two large exotoxins, toxin A (TcdA) and toxin B (TcdB), which are directly responsible for the onset of clinical symptoms of C. difficile-associated diseases (CDADs), such as antibiotics-associated diarrhea and the severe, life-threatening pseudomembranous colitis. Both toxins are multidomain proteins and taken up into host eukaryotic cells via receptor-mediated endocytosis. Within the cell, TcdA and TcdB inactivate Rho and/or Ras protein family members by glucosylation, which eventually results in cell death. The cytotoxic mode of action of the toxins is the main reason for the disease. Thus, compounds capable of inhibiting the cellular uptake and/or mode-of-action of both toxins are of high therapeutic interest. Recently, we found that the sterol regulatory element-binding protein 2 (SREBP-2) pathway, which regulates cholesterol content in membranes, is crucial for the intoxication of cells by TcdA and TcdB. Furthermore, it has been shown that membrane cholesterol is required for TcdA- as well as TcdB-mediated pore formation in endosomal membranes, which is a key step during the translocation of the glucosyltransferase domain of both toxins from endocytic vesicles into the cytosol of host cells. In the current study, we demonstrate that intoxication by TcdA and TcdB is diminished in cultured cells preincubated with the compound U18666A, an established inhibitor of cholesterol biosynthesis and/or intracellular transport. U18666A-pretreated cells were also less sensitive against TcdA and TcdB variants from the epidemic NAP1/027 C. difficile strain. Our study corroborates the crucial role of membrane cholesterol for cell entry of TcdA and TcdB, thus providing a valuable basis for the development of novel antitoxin strategies in the context of CDADs.
Collapse
Affiliation(s)
| | - Selina Kindig
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Adriana Badilla-Lobo
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Ebru Durgun
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Tharani Thuraisingam
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Alexander Witte
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Shuo Song
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - César Rodríguez
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Holger Barth
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
22
|
Simpson HL, Roberts CL, Thompson LM, Leiper CR, Gittens N, Trotter E, Duckworth CA, Papoutsopoulou S, Miyajima F, Roberts P, O’Kennedy N, Rhodes JM, Campbell BJ. Soluble Non-Starch Polysaccharides From Plantain ( Musa x paradisiaca L.) Diminish Epithelial Impact of Clostridioides difficile. Front Pharmacol 2021; 12:766293. [PMID: 34955836 PMCID: PMC8707065 DOI: 10.3389/fphar.2021.766293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
Clostridioides difficile infection (CDI) is a leading cause of antibiotic-associated diarrhoea. Adhesion of this Gram-positive pathogen to the intestinal epithelium is a crucial step in CDI, with recurrence and relapse of disease dependent on epithelial interaction of its endospores. Close proximity, or adhesion of, hypervirulent strains to the intestinal mucosa are also likely to be necessary for the release of C. difficile toxins, which when internalized, result in intestinal epithelial cell rounding, damage, inflammation, loss of barrier function and diarrhoea. Interrupting these C. difficile-epithelium interactions could therefore represent a promising therapeutic strategy to prevent and treat CDI. Intake of dietary fibre is widely recognised as being beneficial for intestinal health, and we have previously shown that soluble non-starch polysaccharides (NSP) from plantain banana (Musa spp.), can block epithelial adhesion and invasion of a number of gut pathogens, such as E. coli and Salmonellae. Here, we assessed the action of plantain NSP, and a range of alternative soluble plant fibres, for inhibitory action on epithelial interactions of C. difficile clinical isolates, purified endospore preparations and toxins. We found that plantain NSP possessed ability to disrupt epithelial adhesion of C. difficile vegetative cells and spores, with inhibitory activity against C. difficile found within the acidic (pectin-rich) polysaccharide component, through interaction with the intestinal epithelium. Similar activity was found with NSP purified from broccoli and leek, although seen to be less potent than NSP from plantain. Whilst plantain NSP could not block the interaction and intracellular action of purified C. difficile toxins, it significantly diminished the epithelial impact of C. difficile, reducing both bacteria and toxin induced inflammation, activation of caspase 3/7 and cytotoxicity in human intestinal cell-line and murine intestinal organoid cultures. Dietary supplementation with soluble NSP from plantain may therefore confer a protective effect in CDI patients by preventing adhesion of C. difficile to the mucosa, i.e. a "contrabiotic" effect, and diminishing its epithelial impact. This suggests that plantain soluble dietary fibre may be a therapeutically effective nutritional product for use in the prevention or treatment of CDI and antibiotic-associated diarrhoea.
Collapse
Affiliation(s)
- Hannah L. Simpson
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carol L. Roberts
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Louise M. Thompson
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Cameron R. Leiper
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Nehana Gittens
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Ellie Trotter
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carrie A. Duckworth
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Stamatia Papoutsopoulou
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Fabio Miyajima
- Wolfson Centre for Personalised Medicine, Department of Molecular & Clinical Pharmacology, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Oswaldo Cruz Foundation (Fiocruz), Eusébio, Brazil
| | - Paul Roberts
- Department of Microbiology, Liverpool Clinical Laboratories, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
- School for Medicine and Clinical Practice, University of Wolverhampton, Wolverhampton, United Kingdom
| | - Niamh O’Kennedy
- Provexis PLC, c/o The University of Aberdeen, Aberdeen, United Kingdom
| | - Jonathan M. Rhodes
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Barry J. Campbell
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
23
|
Orrell KE, Melnyk RA. Translocation expands the scope of the large clostridial toxin family. Trends Biochem Sci 2021; 46:953-959. [PMID: 34429235 DOI: 10.1016/j.tibs.2021.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/20/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022]
Abstract
Large clostridial toxins (LCTs) are a family of six homologous disease-causing proteins characterised by their large size (>200 kDa) and conserved multidomain architectures. Using their central translocation and receptor-binding domain (T domain), LCTs bind host cell receptors and translocate their upstream glycosyltransferase and cysteine protease domain across the endosomal membrane and into the cytosol. The recent discovery of hundreds of LCT-like T domains in diverse genomic contexts and domain architectures from bacteria other than clostridia has provided significant new insights into the enigmatic process of LCT translocation, but also has put the definition of what constitutes an LCT into question. In this opinion article, we discuss how these findings have expanded our understanding of LCT translocation and reshaped the scope of the LCT family.
Collapse
Affiliation(s)
- Kathleen E Orrell
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, Toronto M5G 0A4, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto M5S 1A8, Ontario, Canada
| | - Roman A Melnyk
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, Toronto M5G 0A4, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto M5S 1A8, Ontario, Canada.
| |
Collapse
|
24
|
Opportunities for Nanomedicine in Clostridioides difficile Infection. Antibiotics (Basel) 2021; 10:antibiotics10080948. [PMID: 34438998 PMCID: PMC8388953 DOI: 10.3390/antibiotics10080948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022] Open
Abstract
Clostridioides difficile, a spore-forming bacterium, is a nosocomial infectious pathogen which can be found in animals as well. Although various antibiotics and disinfectants were developed, C. difficile infection (CDI) remains a serious health problem. C. difficile spores have complex structures and dormant characteristics that contribute to their resistance to harsh environments, successful transmission and recurrence. C. difficile spores can germinate quickly after being exposed to bile acid and co-germinant in a suitable environment. The vegetative cells produce endospores, and the mature spores are released from the hosts for dissemination of the pathogen. Therefore, concurrent elimination of C. difficile vegetative cells and inhibition of spore germination is essential for effective control of CDI. This review focused on the molecular pathogenesis of CDI and new trends in targeting both spores and vegetative cells of this pathogen, as well as the potential contribution of nanotechnologies for the effective management of CDI.
Collapse
|
25
|
Guo S, Chen Y, Liu J, Zhang X, Liu Z, Zhou Z, Wei W. Low-density lipoprotein receptor-related protein 1 is a CROPs-associated receptor for Clostridioides difficile toxin B. SCIENCE CHINA-LIFE SCIENCES 2021; 65:107-118. [PMID: 34279819 DOI: 10.1007/s11427-021-1943-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/28/2021] [Indexed: 12/23/2022]
Abstract
As the leading cause of worldwide hospital-acquired infection, Clostridioides difficile (C. difficile) infection has caused heavy economic and hospitalized burden, while its pathogenesis is not fully understood. Toxin B (TcdB) is one of the major virulent factors of C. difficile. Recently, CSPG4 and FZD2 were reported to be the receptors that mediate TcdB cellular entry. However, genetic ablation of genes encoding these receptors failed to completely block TcdB entry, implicating the existence of alternative receptor(s) for this toxin. Here, by employing the CRISPR-Cas9 screen in CSPG4-deficient HeLa cells, we identified LDL receptor-related protein-1 (LRP1) as a novel receptor for TcdB. Knockout of LRP1 in both CSPG4-deficient HeLa cells and colonic epithelium Caco2 cells conferred cells with increased TcdB resistance, while LRP1 overexpression sensitized cells to TcdB at a low concentration. Co-immunoprecipitation assay showed that LRP1 interacts with full-length TcdB. Moreover, CROPs domain, which is dispensable for TcdB's interaction with CSPG4 and FZD2, is sufficient for binding to LRP1. As such, our study provided evidence for a novel mechanism of TcdB entry and suggested potential therapeutic targets for treating C. diff.
Collapse
Affiliation(s)
- Shengjie Guo
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yiou Chen
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Jingze Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xinyi Zhang
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zhiheng Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zhuo Zhou
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China.
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
26
|
Abstract
Large clostridial toxins (LCTs) are a family of bacterial exotoxins that infiltrate and destroy target cells. Members of the LCT family include Clostridioides difficile toxins TcdA and TcdB, Paeniclostridium sordellii toxins TcsL and TcsH, Clostridium novyi toxin TcnA, and Clostridium perfringens toxin TpeL. Since the 19th century, LCT-secreting bacteria have been isolated from the blood, organs, and wounds of diseased individuals, and LCTs have been implicated as the primary virulence factors in a variety of infections, including C. difficile infection and some cases of wound-associated gas gangrene. Clostridia express and secrete LCTs in response to various physiological signals. LCTs invade host cells by binding specific cell surface receptors, ultimately leading to internalization into acidified vesicles. Acidic pH promotes conformational changes within LCTs, which culminates in translocation of the N-terminal glycosyltransferase and cysteine protease domain across the endosomal membrane and into the cytosol, leading first to cytopathic effects and later to cytotoxic effects. The focus of this review is on the role of LCTs in infection and disease, the mechanism of LCT intoxication, with emphasis on recent structural work and toxin subtyping analysis, and the genomic discovery and characterization of LCT homologues. We provide a comprehensive review of these topics and offer our perspective on emerging questions and future research directions for this enigmatic family of toxins.
Collapse
|
27
|
Ernst K, Landenberger M, Nieland J, Nørgaard K, Frick M, Fois G, Benz R, Barth H. Characterization and Pharmacological Inhibition of the Pore-Forming Clostridioides difficile CDTb Toxin. Toxins (Basel) 2021; 13:toxins13060390. [PMID: 34071730 PMCID: PMC8226936 DOI: 10.3390/toxins13060390] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022] Open
Abstract
The clinically highly relevant Clostridioides (C.) difficile releases several AB-type toxins that cause diseases such as diarrhea and pseudomembranous colitis. In addition to the main virulence factors Rho/Ras-glycosylating toxins TcdA and TcdB, hypervirulent strains produce the binary AB-type toxin CDT. CDT consists of two separate proteins. The binding/translocation B-component CDTb facilitates uptake and translocation of the enzyme A-component CDTa to the cytosol of cells. Here, CDTa ADP-ribosylates G-actin, resulting in depolymerization of the actin cytoskeleton. We previously showed that CDTb exhibits cytotoxicity in the absence of CDTa, which is most likely due to pore formation in the cytoplasmic membrane. Here, we further investigated this cytotoxic effect and showed that CDTb impairs CaCo-2 cell viability and leads to redistribution of F-actin without affecting tubulin structures. CDTb was detected at the cytoplasmic membrane in addition to its endosomal localization if CDTb was applied alone. Chloroquine and several of its derivatives, which were previously identified as toxin pore blockers, inhibited intoxication of Vero, HCT116, and CaCo-2 cells by CDTb and CDTb pores in vitro. These results further strengthen pore formation by CDTb in the cytoplasmic membrane as the underlying cytotoxic mechanism and identify pharmacological pore blockers as potent inhibitors of cytotoxicity induced by CDTb and CDTa plus CDTb.
Collapse
Affiliation(s)
- Katharina Ernst
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany; (M.L.); (J.N.); (K.N.)
- Correspondence: (K.E.); (H.B.)
| | - Marc Landenberger
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany; (M.L.); (J.N.); (K.N.)
| | - Julian Nieland
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany; (M.L.); (J.N.); (K.N.)
| | - Katharina Nørgaard
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany; (M.L.); (J.N.); (K.N.)
| | - Manfred Frick
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (M.F.); (G.F.)
| | - Giorgio Fois
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (M.F.); (G.F.)
| | - Roland Benz
- Department of Life Sciences and Chemistry, Jacobs-University Bremen, 28759 Bremen, Germany;
| | - Holger Barth
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081 Ulm, Germany; (M.L.); (J.N.); (K.N.)
- Correspondence: (K.E.); (H.B.)
| |
Collapse
|
28
|
Ranftler C, Nagl D, Sparer A, Röhrich A, Freissmuth M, El-Kasaby A, Nasrollahi Shirazi S, Koban F, Tschegg C, Nizet S. Binding and neutralization of C. difficile toxins A and B by purified clinoptilolite-tuff. PLoS One 2021; 16:e0252211. [PMID: 34043688 PMCID: PMC8158989 DOI: 10.1371/journal.pone.0252211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/11/2021] [Indexed: 12/31/2022] Open
Abstract
Clostridioides difficile (C. difficile) infection is a major public health problem worldwide. The current treatment of C. difficile-associated diarrhea relies on the use of antibacterial agents. However, recurrences are frequent. The main virulence factors of C. difficile are two secreted cytotoxic proteins toxin A and toxin B. Alternative research exploring toxin binding by resins found a reduced rate of recurrence by administration of tolevamer. Hence, binding of exotoxins may be useful in preventing a relapse provided that the adsorbent is innocuous. Here, we examined the toxin binding capacity of G-PUR®, a purified version of natural clinoptilolite-tuff. Our observations showed that the purified clinoptilolite-tuff adsorbed clinically relevant amounts of C. difficile toxins A and B in vitro and neutralized their action in a Caco-2 intestinal model. This conclusion is based on four independent sets of findings: G-PUR® abrogated toxin-induced (i) RAC1 glucosylation, (ii) redistribution of occludin, (iii) rarefaction of the brush border as visualized by scanning electron microscopy and (iv) breakdown of the epithelial barrier recorded by transepithelial electrical resistance monitoring. Finally, we confirmed that the epithelial monolayer tolerated G-PUR® over a wide range of particle densities. Our findings justify the further exploration of purified clinoptilolite-tuff as a safe agent in the treatment and/or prevention of C. difficile-associated diarrhea.
Collapse
Affiliation(s)
- Carmen Ranftler
- GLOCK Health, Science and Research G.m.b.H., Deutsch-Wagram, Austria
| | - Dietmar Nagl
- GLOCK Health, Science and Research G.m.b.H., Deutsch-Wagram, Austria
| | - Andreas Sparer
- GLOCK Health, Science and Research G.m.b.H., Deutsch-Wagram, Austria
| | - Andreas Röhrich
- GLOCK Health, Science and Research G.m.b.H., Deutsch-Wagram, Austria
| | - Michael Freissmuth
- Institute of Pharmacology & Gaston H. Glock Research Laboratories for Explorative Drug Development, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology & Gaston H. Glock Research Laboratories for Explorative Drug Development, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Shahrooz Nasrollahi Shirazi
- Institute of Pharmacology & Gaston H. Glock Research Laboratories for Explorative Drug Development, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Florian Koban
- Institute of Pharmacology & Gaston H. Glock Research Laboratories for Explorative Drug Development, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Cornelius Tschegg
- GLOCK Health, Science and Research G.m.b.H., Deutsch-Wagram, Austria
| | - Stephane Nizet
- GLOCK Health, Science and Research G.m.b.H., Deutsch-Wagram, Austria
- * E-mail:
| |
Collapse
|
29
|
Landenberger M, Nieland J, Roeder M, Nørgaard K, Papatheodorou P, Ernst K, Barth H. The cytotoxic effect of Clostridioides difficile pore-forming toxin CDTb. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183603. [PMID: 33689753 DOI: 10.1016/j.bbamem.2021.183603] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 01/05/2023]
Abstract
Clostridioides (C.) difficile is clinically highly relevant and produces several AB-type protein toxins, which are the causative agents for C. difficile-associated diarrhea and pseudomembranous colitis. Treatment with antibiotics can lead to C. difficile overgrowth in the gut of patients due to the disturbed microbiota. C. difficile releases large Rho/Ras-GTPase glucosylating toxins TcdA and TcdB, which are considered as the major virulence factors for C. difficile-associated diseases. In addition to TcdA and TcdB, C. difficile strains isolated from severe cases of colitis produce a third toxin called CDT. CDT is a member of the family of clostridial binary actin ADP-ribosylating toxins and consists of two separate protein components. The B-component, CDTb, binds to the receptor and forms a complex with and facilitates transport and translocation of the enzymatically active A-component, CDTa, into the cytosol of target cells by forming trans-membrane pores through which CDTa translocates. In the cytosol, CDTa ADP-ribosylates G-actin causing depolymerization of the actin cytoskeleton and, eventually, cell death. In the present study, we report that CDTb exhibits a cytotoxic effect in the absence of CDTa. We show that CDTb causes cell rounding and impairs cell viability and the epithelial integrity of CaCo-2 monolayers in the absence of CDTa. CDTb-induced cell rounding depended on the presence of LSR, the specific cellular receptor of CDT. The isolated receptor-binding domain of CDTb was not sufficient to cause cell rounding. CDTb-induced cell rounding was inhibited by enzymatically inactive CDTa or a pore-blocker, implying that CDTb pores in cytoplasmic membranes contribute to cytotoxicity.
Collapse
Affiliation(s)
- Marc Landenberger
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Julian Nieland
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Maurice Roeder
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | - Katharina Nørgaard
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| | | | - Katharina Ernst
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany.
| | - Holger Barth
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
30
|
Asadpoor M, Ithakisiou GN, Henricks PAJ, Pieters R, Folkerts G, Braber S. Non-Digestible Oligosaccharides and Short Chain Fatty Acids as Therapeutic Targets against Enterotoxin-Producing Bacteria and Their Toxins. Toxins (Basel) 2021; 13:175. [PMID: 33668708 PMCID: PMC7996226 DOI: 10.3390/toxins13030175] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Enterotoxin-producing bacteria (EPB) have developed multiple mechanisms to disrupt gut homeostasis, and provoke various pathologies. A major part of bacterial cytotoxicity is attributed to the secretion of virulence factors, including enterotoxins. Depending on their structure and mode of action, enterotoxins intrude the intestinal epithelium causing long-term consequences such as hemorrhagic colitis. Multiple non-digestible oligosaccharides (NDOs), and short chain fatty acids (SCFA), as their metabolites produced by the gut microbiota, interact with enteropathogens and their toxins, which may result in the inhibition of the bacterial pathogenicity. NDOs characterized by diverse structural characteristics, block the pathogenicity of EPB either directly, by inhibiting bacterial adherence and growth, or biofilm formation or indirectly, by promoting gut microbiota. Apart from these abilities, NDOs and SCFA can interact with enterotoxins and reduce their cytotoxicity. These anti-virulent effects mostly rely on their ability to mimic the structure of toxin receptors and thus inhibiting toxin adherence to host cells. This review focuses on the strategies of EPB and related enterotoxins to impair host cell immunity, discusses the anti-pathogenic properties of NDOs and SCFA on EPB functions and provides insight into the potential use of NDOs and SCFA as effective agents to fight against enterotoxins.
Collapse
Affiliation(s)
- Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Georgia-Nefeli Ithakisiou
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Paul A. J. Henricks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Roland Pieters
- Division of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands;
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| |
Collapse
|
31
|
Ernst K, Sailer J, Braune M, Barth H. Intoxication of mammalian cells with binary clostridial enterotoxins is inhibited by the combination of pharmacological chaperone inhibitors. Naunyn Schmiedebergs Arch Pharmacol 2020; 394:941-954. [PMID: 33284399 PMCID: PMC8102464 DOI: 10.1007/s00210-020-02029-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/18/2020] [Indexed: 01/05/2023]
Abstract
Binary enterotoxins Clostridioides difficile CDT toxin, Clostridium botulinum C2 toxin, and Clostridium perfringens iota toxin consist of two separate protein components. The B-components facilitate receptor-mediated uptake into mammalian cells and form pores into endosomal membranes through which the enzymatic active A-components translocate into the cytosol. Here, the A-components ADP-ribosylate G-actin which leads to F-actin depolymerization followed by rounding of cells which causes clinical symptoms. The protein folding helper enzymes Hsp90, Hsp70, and peptidyl-prolyl cis/trans isomerases of the cyclophilin (Cyp) and FK506 binding protein (FKBP) families are required for translocation of A-components of CDT, C2, and iota toxins from endosomes to the cytosol. Here, we demonstrated that simultaneous inhibition of these folding helpers by specific pharmacological inhibitors protects mammalian, including human, cells from intoxication with CDT, C2, and iota toxins, and that the inhibitor combination displayed an enhanced effect compared to application of the individual inhibitors. Moreover, combination of inhibitors allowed a concentration reduction of the individual compounds as well as decreasing of the incubation time with inhibitors to achieve a protective effect. These results potentially have implications for possible future therapeutic applications to relieve clinical symptoms caused by bacterial toxins that depend on Hsp90, Hsp70, Cyps, and FKBPs for their membrane translocation into the cytosol of target cells.
Collapse
Affiliation(s)
- Katharina Ernst
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081, Ulm, Germany.
| | - Judith Sailer
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Maria Braune
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Holger Barth
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89081, Ulm, Germany.
| |
Collapse
|
32
|
Schöttelndreier D, Langejürgen A, Lindner R, Genth H. Low Density Lipoprotein Receptor-Related Protein-1 (LRP1) Is Involved in the Uptake of Clostridioides difficile Toxin A and Serves as an Internalizing Receptor. Front Cell Infect Microbiol 2020; 10:565465. [PMID: 33194803 PMCID: PMC7604483 DOI: 10.3389/fcimb.2020.565465] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/28/2020] [Indexed: 01/24/2023] Open
Abstract
Toxin producing Clostridioides difficile strains cause gastrointestinal infections with the large glucosylating protein toxins A (TcdA) and B (TcdB) being major virulence factors responsible for the onset of symptoms. TcdA and TcdB enter their target cells via receptor-mediated endocytosis. Inside the cell, the toxins glucosylate and thereby inactivate small GTPases of the Rho-/Ras subfamilies resulting in actin reorganization and cell death. The receptors of TcdA are still elusive, glycoprotein 96 (gp96), the low density lipoprotein receptor family (LDLR) and sulfated glycosaminoglycans (sGAGs) have most recently been suggested as receptors for TcdA. In this study, we provide evidence on rapid endocytosis of Low density lipoprotein Receptor-related Protein-1 (LRP1) into fibroblasts and Caco-2 cells by exploiting biotinylation of cell surface proteins. In contrast, gp96 was not endocytosed either in the presence or absence of TcdA. The kinetics of internalization of TfR and LRP1 were comparable in the presence and the absence of TcdA, excluding that TcdA facilitates its internalization by triggering internalization of its receptors. Exploiting fibroblasts with a genetic deletion of LRP1, TcdA was about one order of magnitude less potent in LRP1-deficient cells as compared to the corresponding control cells. In contrast, TcdB exhibited a comparable potency in LRP1-proficient and -deficient fibroblasts. These findings suggested a role of LRP1 in the cellular uptake of TcdA but not of TcdB. Correspondingly, binding of TcdA to the cell surface of LRP1-deficient fibroblasts was reduced as compared with LRP1-proficient fibroblasts. Finally, TcdA bound to LRP1 ligand binding type repeat cluster II (amino acid 786–1,165) and cluster IV (amino acid 3332-3779). In conclusion, LRP1 appears to serve as an endocytic receptor and gp96 as a non-endocytic receptor for TcdA.
Collapse
Affiliation(s)
| | - Anna Langejürgen
- Institutes for Toxicology, Hannover Medical School, Hannover, Germany
| | - Robert Lindner
- Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Harald Genth
- Institutes for Toxicology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
33
|
Korbmacher M, Fischer S, Landenberger M, Papatheodorou P, Aktories K, Barth H. Human α-Defensin-5 Efficiently Neutralizes Clostridioides difficile Toxins TcdA, TcdB, and CDT. Front Pharmacol 2020; 11:1204. [PMID: 32903430 PMCID: PMC7435013 DOI: 10.3389/fphar.2020.01204] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
Infections with the pathogenic bacterium Clostridioides (C.) difficile are coming more into focus, in particular in hospitalized patients after antibiotic treatment. C. difficile produces the exotoxins TcdA and TcdB. Since some years, hypervirulent strains are described, which produce in addition the binary actin ADP-ribosylating toxin CDT. These strains are associated with more severe clinical presentations and increased morbidity and frequency. Once in the cytosol of their target cells, the catalytic domains of TcdA and TcdB glucosylate and thereby inactivate small Rho-GTPases whereas the enzyme subunit of CDT ADP-ribosylates G-actin. Thus, enzymatic activity of the toxins leads to destruction of the cytoskeleton and breakdown of the epidermal gut barrier integrity. This causes clinical symptoms ranging from mild diarrhea to life-threatening pseudomembranous colitis. Therefore, pharmacological inhibition of the secreted toxins is of peculiar medical interest. Here, we investigated the neutralizing effect of the human antimicrobial peptide α-defensin-5 toward TcdA, TcdB, and CDT in human cells. The toxin-neutralizing effects of α-defensin-5 toward TcdA, TcdB, and CDT as well as their medically relevant combination were demonstrated by analyzing toxins-induced changes in cell morphology, intracellular substrate modification, and decrease of trans-epithelial electrical resistance. For TcdA, the underlying mode of inhibition is most likely based on the formation of inactive toxin-defensin-aggregates whereas for CDT, the binding- and transport-component might be influenced. The application of α-defensin-5 delayed intoxication of cells in a time- and concentration-dependent manner. Due to its effect on the toxins, α-defensin-5 should be considered as a candidate to treat severe C. difficile-associated diseases.
Collapse
Affiliation(s)
- Michael Korbmacher
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Marc Landenberger
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | | | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| |
Collapse
|
34
|
Lee H, Beilhartz GL, Kucharska I, Raman S, Cui H, Lam MHY, Liang H, Rubinstein JL, Schramek D, Julien JP, Melnyk RA, Taipale M. Recognition of Semaphorin Proteins by P. sordellii Lethal Toxin Reveals Principles of Receptor Specificity in Clostridial Toxins. Cell 2020; 182:345-356.e16. [PMID: 32589945 PMCID: PMC7316060 DOI: 10.1016/j.cell.2020.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/06/2020] [Accepted: 06/01/2020] [Indexed: 01/03/2023]
Abstract
Pathogenic clostridial species secrete potent toxins that induce severe host tissue damage. Paeniclostridium sordellii lethal toxin (TcsL) causes an almost invariably lethal toxic shock syndrome associated with gynecological infections. TcsL is 87% similar to C. difficile TcdB, which enters host cells via Frizzled receptors in colon epithelium. However, P. sordellii infections target vascular endothelium, suggesting that TcsL exploits another receptor. Here, using CRISPR/Cas9 screening, we establish semaphorins SEMA6A and SEMA6B as TcsL receptors. We demonstrate that recombinant SEMA6A can protect mice from TcsL-induced edema. A 3.3 Å cryo-EM structure shows that TcsL binds SEMA6A with the same region that in TcdB binds structurally unrelated Frizzled. Remarkably, 15 mutations in this evolutionarily divergent surface are sufficient to switch binding specificity of TcsL to that of TcdB. Our findings establish semaphorins as physiologically relevant receptors for TcsL and reveal the molecular basis for the difference in tissue targeting and disease pathogenesis between highly related toxins.
Collapse
Affiliation(s)
- Hunsang Lee
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Greg L Beilhartz
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Swetha Raman
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Hong Cui
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Mandy Hiu Yi Lam
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Huazhu Liang
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - John L Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada; Molecular Architecture of Life Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada.
| | - Roman A Melnyk
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Mikko Taipale
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Molecular Architecture of Life Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
35
|
Varela-Chavez C, Blondel A, Popoff MR. Bacterial intracellularly active toxins: Membrane localisation of the active domain. Cell Microbiol 2020; 22:e13213. [PMID: 32353188 DOI: 10.1111/cmi.13213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/06/2020] [Accepted: 04/17/2020] [Indexed: 12/31/2022]
Abstract
Numerous bacterial toxins exert their activity by inactivating or modulating a specific intracellular host target. For this purpose, these toxins have developed efficient strategies to overcome the different host cell defences including specific binding to cell surface, internalisation, passage through the endosome or plasma membrane, exploiting intracellular trafficking and addressing to intracellular targets. Several intracellularly active toxins deliver an active domain into the cytosol that interacts with a target localised to the inner face of the plasma membrane. Thus, the large clostridial glucosylating toxins (LCGTs) target Rho/Ras-GTPases, certain virulence factors of Gram negative bacteria, Rho-GTPases, while Pasteurella multocida toxin (PMT) targets trimeric G-proteins. Others such as botulinum neurotoxins and tetanus neurotoxin have their substrate on synaptic vesicle membrane. LCGTs, PMT, and certain virulence factors from Vibrio sp. show a particular structure constituted of a four-helix bundle membrane (4HBM) protruding from the catalytic site that specifically binds to the membrane phospholipids and then trap the catalytic domain at the proximity of the membrane anchored substrate. Structural and functional analysis indicate that the 4HBM tip of the Clostridium sordellii lethal toxin (TcsL) from the LCGT family contain two loops forming a cavity that mediates the binding to phospholipids and more specifically to phosphatidylserine.
Collapse
Affiliation(s)
| | - Arnaud Blondel
- Unité de Bio-Informatique Structurale, Institut Pasteur, Paris, France
| | | |
Collapse
|
36
|
Identification of the role of toxin B in the virulence of Clostridioides difficile based on integrated bioinformatics analyses. Int Microbiol 2020; 23:575-587. [PMID: 32388701 DOI: 10.1007/s10123-020-00128-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/03/2020] [Accepted: 04/13/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Clostridioides difficile toxin B (TcdB) plays a critical role in C. difficile infection (CDI), a common and costly healthcare-associated disease. The aim of the current study was to explore the intracellular and potent systemic effects of TcdB on human colon epithelial cells utilizing Gene Expression Omnibus and bioinformatic methods. METHODS Two datasets (GSE63880 and GSE29008) were collected to extract data components of mRNA of TcdB-treated human colon epithelial cells; "limma" package of "R" software was used to screen the differential genes, and "pheatmap" package was applied to construct heat maps for the differential genes; Metascape website was utilized for protein-protein interaction network and Molecular Complex Detection analysis, and Genome Ontology (GO) was used to analyze the selected differential genes. Quantitative real-time PCR (qRT-PCR) and Western blot were performed to validate the expression of hub genes. RESULTS GO terms involved in DNA replication and cell cycle were identified significantly enriched in TcdB-treated human colon epithelial cells. Moreover, the decreased expression of DNA replication-related genes, MCM complex, and CDC45 in C. difficile (TcdA-/TcdB+)-infected Caco-2 cells were validated via qRT-PCR and Western blot assays. CONCLUSIONS In conclusion, the integrated analysis of different gene expression datasets allowed us to identify a set of genes and GO terms underlying the mechanisms of CDI induced by TcdB. It would aid in understanding of the molecular mechanisms underlying TcdB-exposed colon epithelial cells and provide the basis for developing diagnosis biomarkers, treatment, and prevention strategies.
Collapse
|
37
|
Shi J, Peng D, Zhang F, Ruan L, Sun M. The Caenorhabditis elegans CUB-like-domain containing protein RBT-1 functions as a receptor for Bacillus thuringiensis Cry6Aa toxin. PLoS Pathog 2020; 16:e1008501. [PMID: 32369532 PMCID: PMC7228132 DOI: 10.1371/journal.ppat.1008501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/15/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Plant-parasitic nematodes cause huge agricultural economic losses. Two major families of Bacillus thuringiensis crystal proteins, Cry5 and Cry6, show nematicidal activity. Previous work showed that binding to midgut receptors is a limiting step in Cry toxin mode of action. In the case of Cry5Ba, certain Caenorhabditis elegans glycolipids were identified as receptors of this toxin. However, the receptors for Cry6 toxin remain unknown. In this study, the C. elegans CUB-like-domain containing protein RBT-1, released by phosphatidylinositol-specific phospholipase C (PI-PLC), was identified as a Cry6Aa binding protein by affinity chromatography. RBT-1 contained a predicted glycosylphosphatidylinositol (GPI) anchor site and was shown to locate in lipid rafts in the surface of the midgut cells. Western ligand blot assays and ELISA binding analysis confirmed the binding interaction between Cry6Aa and RBT-1 showing high affinity and specificity. In addition, the mutation of rbt-1 gene decreased the susceptibility of C. elegans to Cry6Aa but not that of Cry5Ba. Furthermore, RBT-1 mediated the uptake of Cry6Aa into C. elegans gut cells, and was shown to be involved in triggering pore-formation activity, indicating that RBT-1 is required for the interaction of Cry6Aa with the nematode midgut cells. These results support that RBT-1 is a functional receptor for Cry6Aa. Bacillus thuringiensis (Bt) crystal proteins belong to pore-forming toxins (PFTs), which display virulence against target hosts by forming holes in the cell membrane. Cry6A is a nematicidal PFT, which exhibits unique protein structure and different mode of action than Cry5B, another nematicidal PFT. However, little is known about the mode of action of Cry6A. Although an intracellular nematicidal necrosis pathway of Cry6A was reported, its extracellular mode of action remains unknown. We here demonstrate that the CUB-like-domain containing protein RBT-1 acts as a functional receptor of Cry6A, which mediates the intestinal cell interaction and nematicidal activity of this toxin. RBT-1 represents a new class of crystal protein receptors. RBT-1 is dispensable for Cry5B toxicity against nematodes, consistent with that Cry6A and Cry5B have different nematicidal mechanisms. We also find that Cry6A kills nematodes by complex mechanism since rbt-1 mutation did not affect Cry6A-mediated necrosis signaling pathway. This work not only enhances the understanding of Bt crystal protein-nematode mechanism, but is also in favor for the application of Cry6A in nematode control.
Collapse
Affiliation(s)
- Jianwei Shi
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Donghai Peng
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- * E-mail: (DP); (MS)
| | - Fengjuan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Lifang Ruan
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ming Sun
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- * E-mail: (DP); (MS)
| |
Collapse
|
38
|
Lin B, Qing X, Liao J, Zhuo K. Role of Protein Glycosylation in Host-Pathogen Interaction. Cells 2020; 9:E1022. [PMID: 32326128 PMCID: PMC7226260 DOI: 10.3390/cells9041022] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/11/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
Host-pathogen interactions are fundamental to our understanding of infectious diseases. Protein glycosylation is one kind of common post-translational modification, forming glycoproteins and modulating numerous important biological processes. It also occurs in host-pathogen interaction, affecting host resistance or pathogen virulence often because glycans regulate protein conformation, activity, and stability, etc. This review summarizes various roles of different glycoproteins during the interaction, which include: host glycoproteins prevent pathogens as barriers; pathogen glycoproteins promote pathogens to attack host proteins as weapons; pathogens glycosylate proteins of the host to enhance virulence; and hosts sense pathogen glycoproteins to induce resistance. In addition, this review also intends to summarize the roles of lectin (a class of protein entangled with glycoprotein) in host-pathogen interactions, including bacterial adhesins, viral lectins or host lectins. Although these studies show the importance of protein glycosylation in host-pathogen interaction, much remains to be discovered about the interaction mechanism.
Collapse
Affiliation(s)
- Borong Lin
- Laboratory of Plant Nematology, South China Agricultural University, Guangzhou 510642, China; (B.L.); (J.L.)
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou 510642, China
| | - Xue Qing
- College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China;
| | - Jinling Liao
- Laboratory of Plant Nematology, South China Agricultural University, Guangzhou 510642, China; (B.L.); (J.L.)
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou 510642, China
- Guangdong Eco-Engineering Polytechnic, Guangzhou 510520, China
| | - Kan Zhuo
- Laboratory of Plant Nematology, South China Agricultural University, Guangzhou 510642, China; (B.L.); (J.L.)
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
39
|
Kester JC, Brubaker DK, Velazquez J, Wright C, Lauffenburger DA, Griffith LG. Clostridioides difficile-Associated Antibiotics Alter Human Mucosal Barrier Functions by Microbiome-Independent Mechanisms. Antimicrob Agents Chemother 2020; 64:e01404-19. [PMID: 31988098 PMCID: PMC7179307 DOI: 10.1128/aac.01404-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/04/2020] [Indexed: 12/16/2022] Open
Abstract
A clinically relevant risk factor for Clostridioides difficile-associated disease (CDAD) is recent antibiotic treatment. Although broad-spectrum antibiotics have been shown to disrupt the structure of the gut microbiota, some antibiotics appear to increase CDAD risk without being highly active against intestinal anaerobes, suggesting direct nonantimicrobial effects. We examined cell biological effects of antibiotic exposure that may be involved in bacterial pathogenesis using an in vitro germfree human colon epithelial culture model. We found a marked loss of mucosal barrier and immune function with exposure to the CDAD-associated antibiotics clindamycin and ciprofloxacin, distinct from the results of pretreatment with an antibiotic unassociated with CDAD, tigecycline, which did not reduce innate immune or mucosal barrier functions. Importantly, pretreatment with CDAD-associated antibiotics sensitized mucosal barriers to C. difficile toxin activity in primary cell-derived enteroid monolayers. These data implicate commensal-independent gut mucosal barrier changes in the increased risk of CDAD with specific antibiotics and warrant further studies in in vivo systems. We anticipate this work to suggest potential avenues of research for host-directed treatment and preventive therapies for CDAD.
Collapse
Affiliation(s)
- Jemila C Kester
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Douglas K Brubaker
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jason Velazquez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Charles Wright
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
40
|
Fischer S, Ückert AK, Landenberger M, Papatheodorou P, Hoffmann-Richter C, Mittler AK, Ziener U, Hägele M, Schwan C, Müller M, Kleger A, Benz R, Popoff MR, Aktories K, Barth H. Human peptide α-defensin-1 interferes with Clostridioides difficile toxins TcdA, TcdB, and CDT. FASEB J 2020; 34:6244-6261. [PMID: 32190927 DOI: 10.1096/fj.201902816r] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/23/2019] [Accepted: 02/27/2020] [Indexed: 12/15/2022]
Abstract
The human pathogenic bacterium Clostridioides difficile produces two exotoxins TcdA and TcdB, which inactivate Rho GTPases thereby causing C. difficile-associated diseases (CDAD) including life-threatening pseudomembranous colitis. Hypervirulent strains produce additionally the binary actin ADP-ribosylating toxin CDT. These strains are hallmarked by more severe forms of CDAD and increased frequency and severity. Once in the cytosol, the toxins act as enzymes resulting in the typical clinical symptoms. Therefore, targeting and inactivation of the released toxins are of peculiar interest. Prompted by earlier findings that human α-defensin-1 neutralizes TcdB, we investigated the effects of the defensin on all three C. difficile toxins. Inhibition of TcdA, TcdB, and CDT was demonstrated by analyzing toxin-induced changes in cell morphology, substrate modification, and decrease in transepithelial electrical resistance. Application of α-defensin-1 protected cells and human intestinal organoids from the cytotoxic effects of TcdA, TcdB, CDT, and their combination which is attributed to a direct interaction between the toxins and α-defensin-1. In mice, the application of α-defensin-1 reduced the TcdA-induced damage of intestinal loops in vivo. In conclusion, human α-defensin-1 is a specific and potent inhibitor of the C. difficile toxins and a promising agent to develop novel therapeutic options against C. difficile infections.
Collapse
Affiliation(s)
- Stephan Fischer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Anna-Katharina Ückert
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Marc Landenberger
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | | | | | - Ann-Katrin Mittler
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Ulrich Ziener
- Institute of Organic Chemistry III, Ulm University, Ulm, Germany
| | - Marlen Hägele
- Department of Internal Medicine I, University of Ulm Medical Center, Ulm, Germany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Martin Müller
- Department of Internal Medicine I, University of Ulm Medical Center, Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, University of Ulm Medical Center, Ulm, Germany
| | - Roland Benz
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Michel R Popoff
- Department of Anaerobic Bacteria, Pasteur Institute, Paris, France
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| |
Collapse
|
41
|
The Antibiotic Bacitracin Protects Human Intestinal Epithelial Cells and Stem Cell-Derived Intestinal Organoids from Clostridium difficile Toxin TcdB. Stem Cells Int 2019; 2019:4149762. [PMID: 31467562 PMCID: PMC6701344 DOI: 10.1155/2019/4149762] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/11/2019] [Indexed: 01/05/2023] Open
Abstract
Bacitracin is an established antibiotic for local application and inhibits the cell wall synthesis of Gram-positive bacteria. Recently, we discovered a completely different mode of action of bacitracin and reported that this drug protects human cells from intoxication by a variety of medically relevant bacterial protein toxins including CDT, the binary actin ADP-ribosylating toxin of Clostridium (C.) difficile. Bacitracin prevents the transport of CDT into the cytosol of target cells, most likely by inhibiting the transport function of the binding subunit of this toxin. Here, we tested the effect of bacitracin towards TcdB, a major virulence factor of C. difficile contributing to severe C. difficile-associated diseases (CDAD) including pseudomembranous colitis. Bacitracin protected stem cell-derived human intestinal organoids as well as human gut epithelial cells from intoxication with TcdB. Moreover, it prevented the TcdB-induced disruption of epithelia formed by gut epithelium cells in vitro and maintained the barrier function as detected by measuring transepithelial electrical resistance (TEER). In the presence of bacitracin, TcdB was not able reach its substrate Rac1 in the cytosol of human epithelial cells, most likely because its pH-dependent transport across cell membranes into the cytosol is decreased by bacitracin. In conclusion, in addition to its direct antibiotic activity against C. difficile and its inhibitory effect towards the toxin CDT, bacitracin neutralizes the exotoxin TcdB of this important pathogenic bacterium.
Collapse
|
42
|
López-Ureña D, Orozco-Aguilar J, Chaves-Madrigal Y, Ramírez-Mata A, Villalobos-Jimenez A, Ost S, Quesada-Gómez C, Rodríguez C, Papatheodorou P, Chaves-Olarte E. Toxin B Variants from Clostridium difficile Strains VPI 10463 and NAP1/027 Share Similar Substrate Profile and Cellular Intoxication Kinetics but Use Different Host Cell Entry Factors. Toxins (Basel) 2019; 11:toxins11060348. [PMID: 31212980 PMCID: PMC6628394 DOI: 10.3390/toxins11060348] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
Clostridium difficile induces antibiotic-associated diarrhea due to the release of toxin A (TcdA) and toxin B (TcdB), the latter being its main virulence factor. The epidemic strain NAP1/027 has an increased virulence attributed to different factors. We compared cellular intoxication by TcdBNAP1 with that by the reference strain VPI 10463 (TcdBVPI). In a mouse ligated intestinal loop model, TcdBNAP1 induced higher neutrophil recruitment, cytokine release, and epithelial damage than TcdBVPI. Both toxins modified the same panel of small GTPases and exhibited similar in vitro autoprocessing kinetics. On the basis of sequence variations in the frizzled-binding domain (FBD), we reasoned that TcdBVPI and TcdBNAP1 might have different receptor specificities. To test this possibility, we used a TcdB from a NAP1 variant strain (TcdBNAP1v) unable to glucosylate RhoA but with the same receptor-binding domains as TcdBNAP1. Cells were preincubated with TcdBNAP1v to block cellular receptors, prior to intoxication with either TcdBVPI or TcdBNAP1. Preincubation with TcdBNAP1v blocked RhoA glucosylation by TcdBNAP1 but not by TcdBVPI, indicating that the toxins use different host factors for cell entry. This crucial difference might explain the increased biological activity of TcdBNAP1 in the intestine, representing a contributing factor for the increased virulence of the NAP1/027 strain.
Collapse
Affiliation(s)
- Diana López-Ureña
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, 10101 San José, Costa Rica.
| | - Josué Orozco-Aguilar
- Facultad de Farmacia and Laboratorio de Ensayos Biológicos, Escuela de Medicina, Universidad de Costa Rica, 10101 San José, Costa Rica.
| | - Yendry Chaves-Madrigal
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, 10101 San José, Costa Rica.
| | - Andrea Ramírez-Mata
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, 10101 San José, Costa Rica.
| | - Amanda Villalobos-Jimenez
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, 10101 San José, Costa Rica.
| | - Stefan Ost
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, D-79104 Freiburg, Germany.
| | - Carlos Quesada-Gómez
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, 10101 San José, Costa Rica.
| | - César Rodríguez
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, 10101 San José, Costa Rica.
| | | | - Esteban Chaves-Olarte
- Facultad de Microbiología and Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, 10101 San José, Costa Rica.
| |
Collapse
|
43
|
Mileto S, Das A, Lyras D. Enterotoxic Clostridia: Clostridioides difficile Infections. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0015-2018. [PMID: 31124432 PMCID: PMC11026080 DOI: 10.1128/microbiolspec.gpp3-0015-2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Indexed: 12/17/2022] Open
Abstract
Clostridioides difficile is a Gram-positive, anaerobic, spore forming pathogen of both humans and animals and is the most common identifiable infectious agent of nosocomial antibiotic-associated diarrhea. Infection can occur following the ingestion and germination of spores, often concurrently with a disruption to the gastrointestinal microbiota, with the resulting disease presenting as a spectrum, ranging from mild and self-limiting diarrhea to severe diarrhea that may progress to life-threating syndromes that include toxic megacolon and pseudomembranous colitis. Disease is induced through the activity of the C. difficile toxins TcdA and TcdB, both of which disrupt the Rho family of GTPases in host cells, causing cell rounding and death and leading to fluid loss and diarrhea. These toxins, despite their functional and structural similarity, do not contribute to disease equally. C. difficile infection (CDI) is made more complex by a high level of strain diversity and the emergence of epidemic strains, including ribotype 027-strains which induce more severe disease in patients. With the changing epidemiology of CDI, our understanding of C. difficile disease, diagnosis, and pathogenesis continues to evolve. This article provides an overview of the current diagnostic tests available for CDI, strain typing, the major toxins C. difficile produces and their mode of action, the host immune response to each toxin and during infection, animal models of disease, and the current treatment and prevention strategies for CDI.
Collapse
Affiliation(s)
- S Mileto
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| | - A Das
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| | - D Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| |
Collapse
|
44
|
Schnell L, Felix I, Müller B, Sadi M, Bank F, Papatheodorou P, Popoff MR, Aktories K, Waltenberger E, Benz R, Weichbrodt C, Fauler M, Frick M, Barth H. Revisiting an old antibiotic: bacitracin neutralizes binary bacterial toxins and protects cells from intoxication. FASEB J 2019; 33:5755-5771. [DOI: 10.1096/fj.201802453r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Leonie Schnell
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical Center Ulm Germany
| | - Ina Felix
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical Center Ulm Germany
| | - Bastian Müller
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical Center Ulm Germany
| | - Mirko Sadi
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical Center Ulm Germany
| | - Franziska Bank
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical Center Ulm Germany
| | | | | | - Klaus Aktories
- Institute of ExperimentalClinical Pharmacology and ToxicologyUniversity of Freiburg Freiburg Germany
| | - Eva Waltenberger
- Department of Life Sciences and ChemistryJacobs University Bremen Bremen Germany
| | - Roland Benz
- Department of Life Sciences and ChemistryJacobs University Bremen Bremen Germany
| | | | - Michael Fauler
- Institute of General PhysiologyUniversity of Ulm Ulm Germany
| | - Manfred Frick
- Institute of General PhysiologyUniversity of Ulm Ulm Germany
| | - Holger Barth
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical Center Ulm Germany
| |
Collapse
|
45
|
Papatheodorou P, Song S, López-Ureña D, Witte A, Marques F, Ost GS, Schorch B, Chaves-Olarte E, Aktories K. Cytotoxicity of Clostridium difficile toxins A and B requires an active and functional SREBP-2 pathway. FASEB J 2018; 33:4883-4892. [PMID: 30592645 DOI: 10.1096/fj.201801440r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Clostridium difficile is associated with antibiotic-associated diarrhea and pseudomembranous colitis in humans. Its 2 major toxins, toxins A and B, enter host cells and inactivate GTPases of the Ras homologue/rat sarcoma family by glucosylation. Pore formation of the toxins in the endosomal membrane enables the translocation of their glucosyltransferase domain into the cytosol, and membrane cholesterol is crucial for this process. Here, we asked whether the activity of the sterol regulatory element-binding protein 2 (SREBP-2) pathway, which regulates the cholesterol content in membranes, affects the susceptibility of target cells toward toxins A and B. We show that the SREBP-2 pathway is crucial for the intoxication process of toxins A and B by using pharmacological inhibitors (PF-429242, 25-hydroxycholesterol) and cells that are specifically deficient in SREBP-2 pathway signaling. SREBP-2 pathway inhibition disturbed the cholesterol-dependent pore formation of toxin B in cellular membranes. Preincubation with the cholesterol-lowering drug simvastatin protected cells from toxin B intoxication. Inhibition of the SREBP-2 pathway was without effect when the enzyme portion of toxin B was introduced into target cells via the cell delivery property of anthrax protective antigen. Taken together, these findings allowed us to identify the SREBP-2 pathway as a suitable target for the development of antitoxin therapeutics against C. difficile toxins A and B.-Papatheodorou, P., Song, S., López-Ureña, D., Witte, A., Marques, F., Ost, G. S., Schorch, B., Chaves-Olarte, E., Aktories, K. Cytotoxicity of Clostridium difficile toxins A and B requires an active and functional SREBP-2 pathway.
Collapse
Affiliation(s)
- Panagiotis Papatheodorou
- Institut für Experimentelle and Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany; and
| | - Shuo Song
- Institut für Experimentelle and Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany; and
| | - Diana López-Ureña
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Alexander Witte
- Institut für Experimentelle and Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany; and
| | - Felícia Marques
- Institut für Experimentelle and Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany; and
| | - Gerhard Stefan Ost
- Institut für Experimentelle and Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany; and
| | - Björn Schorch
- Institut für Experimentelle and Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany; and
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Klaus Aktories
- Institut für Experimentelle and Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany; and
| |
Collapse
|
46
|
Genth H, Junemann J, Lämmerhirt CM, Lücke AC, Schelle I, Just I, Gerhard R, Pich A. Difference in Mono-O-Glucosylation of Ras Subtype GTPases Between Toxin A and Toxin B From Clostridioides difficile Strain 10463 and Lethal Toxin From Clostridium sordellii Strain 6018. Front Microbiol 2018; 9:3078. [PMID: 30622517 PMCID: PMC6308379 DOI: 10.3389/fmicb.2018.03078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/29/2018] [Indexed: 12/18/2022] Open
Abstract
Clostridioides difficile toxin A (TcdA) and Toxin B (TcdB) trigger inflammasome activation with caspase-1 activation in cultured cells, which in turn induce the release of IL-6, IFN-γ, and IL-8. Release of these proinflammatory responses is positively regulated by Ras-GTPases, which leads to the hypothesis that Ras glucosylation by glucosylating toxins results in (at least) reduced proinflammatory responses. Against this background, data on toxin-catalyzed Ras glucosylation are required to estimate of pro-inflammatory effect of the glucosylating toxins. In this study, a quantitative evaluation of the GTPase substrate profiles glucosylated in human colonic (Caco-2) cells treated with either TcdA, TcdB, or the related Clostridium sordellii lethal toxin (TcsL) was performed using multiple reaction monitoring (MRM) mass spectrometry. (H/K/N)Ras are presented to be glucosylated by TcsL and TcdA but by neither TcdB isoform tested. Furthermore, the glucosylation of (H/K/N)Ras was detected in TcdA-(not TcdB)-treated cells, as analyzed exploiting immunoblot analysis using the Ras glucosylation-sensitive 27H5 antibody. Furthermore, [14C]glucosylation of substrate GTPase was found to be increased in a cell-free system complemented with Caco-2 lysates. Under these conditions, (H/K/N)Ras glucosylation by TcdA was detected. In contrast, TcdB-catalyzed (H/K/N)Ras glucosylation was detected by neither MRM analysis, immunoblot analysis nor [14C]glucosylation in a cell-free system. The observation that TcdA (not TcdB) glucosylates Ras subtype GTPases correlates with the fact that TcdB (not TcdA) is primarily responsible for inflammatory responses in CDI. Finally, TcsL more efficaciously glucosylated Ras subtype GTPase as compared with TcdA, reinforcing the paradigm that TcsL is the prototype of a Ras glucosylating toxin.
Collapse
Affiliation(s)
- Harald Genth
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | | | | | | | - Ilona Schelle
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | - Ingo Just
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
47
|
Schöttelndreier D, Seeger K, Grassl GA, Winny MR, Lindner R, Genth H. Expression and (Lacking) Internalization of the Cell Surface Receptors of Clostridioides difficile Toxin B. Front Microbiol 2018; 9:1483. [PMID: 30022975 PMCID: PMC6039548 DOI: 10.3389/fmicb.2018.01483] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/14/2018] [Indexed: 12/30/2022] Open
Abstract
Toxin-producing strains of Clostridioides difficile and Clostridium perfringens cause infections of the gastrointestinal tract in humans and ruminants, with the toxins being major virulence factors, essential for the infection, and responsible for the onset of severe symptoms. C. difficile toxin A (TcdA) and toxin B (TcdB), and the large cytotoxin (TpeL) from C. perfringens are single chain bacterial protein toxins with an AB-like toxin structure. The C-terminal delivery domain mediates cell entry of the N-terminal glycosyltransferase domain by receptor-mediated endocytosis. Several cell surface proteins have been proposed to serve as toxin receptors, including chondroitin-sulfate proteoglycan 4 (CSPG4), poliovirus receptor-like 3 (PVRL3), and frizzled-1/2/7 (FZD1/2/7) for TcdB and LDL-receptor-related protein-1 (LRP1) for TpeL. The expression of the TcdB receptors was investigated in human intestinal organoids (HIOs) and in cultured cell lines. HIOs from four human donors exhibited a comparable profile of receptor expression, with PVRL3, LRP1, and FZD7 being expressed and CSPG4 and FZD2 not being expressed. In human epithelial Caco-2 cells and HT29 cells as well as in immortalized murine fibroblasts, either receptor FZD2/7, CSPG4, PVRL3, and LRP1 was expressed. The question whether the toxins take advantage of the normal turnover of their receptors (i.e., constitutive endocytosis and recycling) from the cell surface or whether the toxins activity induce the internalization of their receptors has not yet been addressed. For the analysis of receptor internalization, temperature-induced uptake of biotinylated toxin receptors into immortalized mouse embryonic fibroblasts (MEFs) and Caco-2 cells was exploited. Solely LRP1 exhibited constitutive endocytosis from the plasma membrane to the endosome, which might be abused by TpeL (and possibly TcdB as well) for cell entry. Furthermore, internalization of CSPG4, PVRL3, FZD2, and FZD7 was observed neither in MEFs nor in Caco-2 cells. FZD2/7, CSPG4, and PVRL3 did thus exhibit no constitutive recycling. The presence of TcdB and the p38 activation induced by anisomycin were not able to induce or enhance CSPG4 or PVRL3 uptake in MEFs. In conclusion, FZD2/7, CSPG4, and PVRL3 seem to serve as cell surface binding receptors rather than internalizing receptors of TcdB.
Collapse
Affiliation(s)
| | - Katrin Seeger
- Institute of Medical Microbiology and Hospital Epidemiology and DZIF Partner Site Hannover-Braunschweig, Hannover Medical School, Hanover, Germany
| | - Guntram A Grassl
- Institute of Medical Microbiology and Hospital Epidemiology and DZIF Partner Site Hannover-Braunschweig, Hannover Medical School, Hanover, Germany
| | - Markus R Winny
- Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hanover, Germany
| | - Robert Lindner
- Neuroanatomy and Cell Biology, Hannover Medical School, Hanover, Germany
| | - Harald Genth
- Institute for Toxicology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
48
|
Beer LA, Tatge H, Reich N, Tenspolde M, Olling A, Goy S, Rottner K, Alekov AK, Gerhard R. Early cell death induced by Clostridium difficile TcdB: Uptake and Rac1-glucosylation kinetics are decisive for cell fate. Cell Microbiol 2018; 20:e12865. [PMID: 29904993 DOI: 10.1111/cmi.12865] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/23/2018] [Accepted: 05/26/2018] [Indexed: 12/29/2022]
Abstract
Toxin A and Toxin B (TcdA/TcdB) are large glucosyltransferases produced by Clostridium difficile. TcdB but not TcdA induces reactive oxygen species-mediated early cell death (ECD) when applied at high concentrations. We found that nonglucosylated Rac1 is essential for induction of ECD since inhibition of Rac1 impedes this effect. ECD only occurs when TcdB is rapidly endocytosed. This was shown by generation of chimeras using the trunk of TcdB from a hypervirulent strain. TcdB from hypervirulent strain has been described to translocate from endosomes at higher pH values and thus, meaning faster than reference type TcdB. Accordingly, intracellular delivery of the glucosyltransferase domain of reference TcdB by the trunk of TcdB from hypervirulent strain increased ECD. Furthermore, proton transporters such as sodium/proton exchanger (NHE) or the ClC-5 anion/proton exchanger, both of which contribute to endosomal acidification, also affected cytotoxic potency of TcdB: Specific inhibition of NHE reduced cytotoxicity, whereas transfection of cells with the endosomal anion/proton exchanger ClC-5 increased cytotoxicity of TcdB. Our data suggest that both the uptake rate of TcdB into the cytosol and the status of nonglucosylated Rac1 are key determinants that are decisive for whether ECD or delayed apoptosis is triggered.
Collapse
Affiliation(s)
| | - Helma Tatge
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Nicole Reich
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Michel Tenspolde
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Alexandra Olling
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Sebastian Goy
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
49
|
Ziesemer S, Eiffler I, Schönberg A, Müller C, Hochgräfe F, Beule AG, Hildebrandt JP. Staphylococcus aureusα-Toxin Induces Actin Filament Remodeling in Human Airway Epithelial Model Cells. Am J Respir Cell Mol Biol 2018; 58:482-491. [DOI: 10.1165/rcmb.2016-0207oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Sabine Ziesemer
- Animal Physiology and Biochemistry, Zoological Institute, and
| | - Ina Eiffler
- Animal Physiology and Biochemistry, Zoological Institute, and
| | | | | | - Falko Hochgräfe
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Achim G. Beule
- Department of Otorhinolaryngology, Head and Neck Surgery, Greifswald University Hospital, Greifswald, Germany; and
- Department of Otorhinolaryngology, University Hospital, Münster, Germany
| | | |
Collapse
|
50
|
Rees WD, Steiner TS. Adaptive immune response toClostridium difficileinfection: A perspective for prevention and therapy. Eur J Immunol 2018; 48:398-406. [DOI: 10.1002/eji.201747295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/20/2017] [Accepted: 12/14/2017] [Indexed: 11/06/2022]
Affiliation(s)
- William D. Rees
- Department of Medicine; University of British Columbia; Vancouver BC Canada
| | | |
Collapse
|