1
|
Mondal AK, Sengupta N, Singh M, Biswas R, Lata K, Lahiri I, Dutta S, Chattopadhyay K. Glu289 residue in the pore-forming motif of Vibrio cholerae cytolysin is important for efficient β-barrel pore formation. J Biol Chem 2022; 298:102441. [PMID: 36055404 PMCID: PMC9520032 DOI: 10.1016/j.jbc.2022.102441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 11/19/2022] Open
Abstract
Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging β-barrel pore-forming toxin (β-PFT). Upon binding to the target membranes, VCC monomers first assemble into oligomeric pre-pore intermediates, and subsequently transform into transmembrane β-barrel pores. VCC harbors a designated pore-forming motif, which, during oligomeric pore formation, inserts into the membrane and generates a transmembrane β-barrel scaffold. It remains an enigma how the molecular architecture of the pore-forming motif regulates the VCC pore-formation mechanism. Here, we show that a specific pore-forming motif residue, E289, plays crucial regulatory roles in the pore-formation mechanism of VCC. We find that the mutation of E289A drastically compromises pore-forming activity, without affecting the structural integrity and membrane-binding potential of the toxin monomers. Although our single-particle cryo-EM analysis reveals wild type-like oligomeric β-barrel pore formation by E289A-VCC in the membrane, we demonstrate that the mutant shows severely delayed kinetics in terms of pore-forming ability that can be rescued with elevated temperature conditions. We find that the pore-formation efficacy of E289A-VCC appears to be more profoundly dependent on temperature as compared to that of the wild type toxin. Our results suggest that the E289A mutation traps membrane-bound toxin molecules in the pre-pore-like intermediate state that is hindered from converting into the functional β-barrel pores by a large energy barrier, thus highlighting the importance of this residue for the pore-formation mechanism of VCC.
Collapse
Affiliation(s)
- Anish Kumar Mondal
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Manauli, Punjab, India
| | - Nayanika Sengupta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Mahendra Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Manauli, Punjab, India
| | - Rupam Biswas
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Kusum Lata
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Manauli, Punjab, India
| | - Indrajit Lahiri
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Manauli, Punjab, India
| | - Somnath Dutta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Kausik Chattopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Manauli, Punjab, India.
| |
Collapse
|
2
|
Yip CH, Mahalingam S, Wan KL, Nathan S. Prodigiosin inhibits bacterial growth and virulence factors as a potential physiological response to interspecies competition. PLoS One 2021; 16:e0253445. [PMID: 34161391 PMCID: PMC8221495 DOI: 10.1371/journal.pone.0253445] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 06/06/2021] [Indexed: 11/19/2022] Open
Abstract
Prodigiosin, a red linear tripyrrole pigment, has long been recognised for its antimicrobial property. However, the physiological contribution of prodigiosin to the survival of its producing hosts still remains undefined. Hence, the aim of this study was to investigate the biological role of prodigiosin from Serratia marcescens, particularly in microbial competition through its antimicrobial activity, towards the growth and secreted virulence factors of four clinical pathogenic bacteria (methicillin-resistant Staphylococcus aureus (MRSA), Enterococcus faecalis, Salmonella enterica serovar Typhimurium and Pseudomonas aeruginosa) as well as Staphylococcus aureus and Escherichia coli. Prodigiosin was first extracted from S. marcescens and its purity confirmed by absorption spectrum, high performance liquid chromatography (HPLC) and liquid chromatography-tandem mass spectrophotometry (LC-MS/MS). The extracted prodigiosin was antagonistic towards all the tested bacteria. A disc-diffusion assay showed that prodigiosin is more selective towards Gram-positive bacteria and inhibited the growth of MRSA, S. aureus and E. faecalis and Gram-negative E. coli. A minimum inhibitory concentration of 10 μg/μL of prodigiosin was required to inhibit the growth of S. aureus, E. coli and E. faecalis whereas > 10 μg/μL was required to inhibit MRSA growth. We further assessed the effect of prodigiosin towards bacterial virulence factors such as haemolysin and production of protease as well as on biofilm formation. Prodigiosin did not inhibit haemolysis activity of clinically associated bacteria but was able to reduce protease activity for MRSA, E. coli and E. faecalis as well as decrease E. faecalis, Salmonella Typhimurium and E. coli biofilm formation. Results of this study show that in addition to its role in inhibiting bacterial growth, prodigiosin also inhibits the bacterial virulence factor protease production and biofilm formation, two strategies employed by bacteria in response to microbial competition. As clinical pathogens were more resistant to prodigiosin, we propose that prodigiosin is physiologically important for S. marcescens to compete against other bacteria in its natural soil and surface water environments.
Collapse
Affiliation(s)
- Chee-Hoo Yip
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Sobina Mahalingam
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Kiew-Lian Wan
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Sheila Nathan
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
- * E-mail:
| |
Collapse
|
3
|
Mondal AK, Verma P, Sengupta N, Dutta S, Bhushan Pandit S, Chattopadhyay K. Tyrosine in the hinge region of the pore-forming motif regulates oligomeric β-barrel pore formation by Vibrio cholerae cytolysin. Mol Microbiol 2020; 115:508-525. [PMID: 33089544 DOI: 10.1111/mmi.14631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/03/2020] [Indexed: 01/27/2023]
Abstract
β-barrel pore-forming toxins perforate cell membranes by forming oligomeric β-barrel pores. The most crucial step is the membrane-insertion of the pore-forming motifs that create the transmembrane β-barrel scaffold. Molecular mechanism that regulates structural reorganization of these pore-forming motifs during β-barrel pore-formation still remains elusive. Using Vibrio cholerae cytolysin as an archetypical example of the β-barrel pore-forming toxin, we show that a key tyrosine residue (Y321) in the hinge region of the pore-forming motif plays crucial role in this process. Mutation of Y321 abrogates oligomerization of the membrane-bound toxin protomers, and blocks subsequent steps of pore-formation. Our study suggests that the presence of Y321 in the hinge region of the pore-forming motif is crucial for the toxin molecule to sense membrane-binding, and to trigger essential structural rearrangements required for the subsequent oligomerization and pore-formation process. Such a regulatory mechanism of pore-formation by V. cholerae cytolysin has not been documented earlier in the structurally related β-barrel pore-forming toxins.
Collapse
Affiliation(s)
- Anish Kumar Mondal
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Paras Verma
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Nayanika Sengupta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Somnath Dutta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Shashi Bhushan Pandit
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Kausik Chattopadhyay
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
4
|
Mukherjee A, Ganguly S, Chatterjee NS, Banerjee KK. Vibrio cholerae hemolysin: The β-trefoil domain is required for folding to the native conformation. Biochem Biophys Rep 2016; 8:242-248. [PMID: 28955962 PMCID: PMC5614477 DOI: 10.1016/j.bbrep.2016.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/04/2016] [Accepted: 09/17/2016] [Indexed: 11/16/2022] Open
Abstract
Vibrio cholerae cytolysin/hemolysin (VCC) is a 65 kDa β-pore-forming toxin causing lysis and death of eukaryotic cells. Apart from the core cytolysin domain, VCC has two lectin domains with β-trefoil and β-prism folds. The β-prism domain binds to cell surface carbohydrate receptors; the role of the β-trefoil domain is unknown. Here, we show that the pro-VCC mutant without the β-trefoil domain formed aggregates highly susceptible to proteolysis, suggesting lack of a properly folded compact structure. The VCC variants with Trp532Ala or Trp534Ala mutation in the β-trefoil domain formed hemolytically inactive, protease-resistant, ring-shaped SDS-labile oligomers with diameters of ~19 nm. The Trp mutation induced a dramatic change in the global conformation of VCC, as indicated by: (a) the change in surface polarity from hydrophobic to hydrophilic; (b) movement of core Trp residues to the protein-water interface; and (c) decrease in reactivity to the anti-VCC antibody by >100-fold. In fact, the mutant VCC had little similarity to the wild toxin. However, the association constant for the carbohydrate-dependent interaction mediated by the β-prism domain decreased marginally from ~3×108 to ~5×107 M-1. We interpret the observations by proposing: (a) the β-trefoil domain is critical to the folding of the cytolysin domain to its active conformation; (b) the β-prism domain is an autonomous folding unit.
Collapse
Affiliation(s)
| | | | | | - Kalyan K. Banerjee
- Division of Biochemistry, National Institute of Cholera and Enteric Diseases, Kolkata 700 010, India
| |
Collapse
|
5
|
Proteomics Analysis Reveals Previously Uncharacterized Virulence Factors in Vibrio proteolyticus. mBio 2016; 7:mBio.01077-16. [PMID: 27460800 PMCID: PMC4981721 DOI: 10.1128/mbio.01077-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Members of the genus Vibrio include many pathogens of humans and marine animals that share genetic information via horizontal gene transfer. Hence, the Vibrio pan-genome carries the potential to establish new pathogenic strains by sharing virulence determinants, many of which have yet to be characterized. Here, we investigated the virulence properties of Vibrio proteolyticus, a Gram-negative marine bacterium previously identified as part of the Vibrio consortium isolated from diseased corals. We found that V. proteolyticus causes actin cytoskeleton rearrangements followed by cell lysis in HeLa cells in a contact-independent manner. In search of the responsible virulence factor involved, we determined the V. proteolyticus secretome. This proteomics approach revealed various putative virulence factors, including active type VI secretion systems and effectors with virulence toxin domains; however, these type VI secretion systems were not responsible for the observed cytotoxic effects. Further examination of the V. proteolyticus secretome led us to hypothesize and subsequently demonstrate that a secreted hemolysin, belonging to a previously uncharacterized clan of the leukocidin superfamily, was the toxin responsible for the V. proteolyticus-mediated cytotoxicity in both HeLa cells and macrophages. Clearly, there remains an armory of yet-to-be-discovered virulence factors in the Vibrio pan-genome that will undoubtedly provide a wealth of knowledge on how a pathogen can manipulate host cells. The pan-genome of the genus Vibrio is a potential reservoir of unidentified toxins that can provide insight into how members of this genus have successfully risen as emerging pathogens worldwide. We focused on Vibrio proteolyticus, a marine bacterium that was previously implicated in virulence toward marine animals, and characterized its interaction with eukaryotic cells. We found that this bacterium causes actin cytoskeleton rearrangements and leads to cell death. Using a proteomics approach, we identified a previously unstudied member of the leukocidin family of pore-forming toxins as the virulence factor responsible for the observed cytotoxicity in eukaryotic cells, as well as a plethora of additional putative virulence factors secreted by this bacterium. Our findings reveal a functional new clan of the leukocidin toxin superfamily and establish this pathogen as a reservoir of potential toxins that can be used for biomedical applications.
Collapse
|
6
|
De S, Bubnys A, Alonzo F, Hyun J, Lary JW, Cole JL, Torres VJ, Olson R. The Relationship between Glycan Binding and Direct Membrane Interactions in Vibrio cholerae Cytolysin, a Channel-forming Toxin. J Biol Chem 2015; 290:28402-28415. [PMID: 26416894 DOI: 10.1074/jbc.m115.675967] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Indexed: 12/19/2022] Open
Abstract
Bacterial pore-forming toxins (PFTs) are structurally diverse pathogen-secreted proteins that form cell-damaging channels in the membranes of host cells. Most PFTs are released as water-soluble monomers that first oligomerize on the membrane before inserting a transmembrane channel. To modulate specificity and increase potency, many PFTs recognize specific cell surface receptors that increase the local toxin concentration on cell membranes, thereby facilitating channel formation. Vibrio cholerae cytolysin (VCC) is a toxin secreted by the human pathogen responsible for pandemic cholera disease and acts as a defensive agent against the host immune system. Although it has been shown that VCC utilizes specific glycan receptors on the cell surface, additional direct contacts with the membrane must also play a role in toxin binding. To better understand the nature of these interactions, we conducted a systematic investigation of the membrane-binding surface of VCC to identify additional membrane interactions important in cell targeting. Through cell-based assays on several human-derived cell lines, we show that VCC is unlikely to utilize high affinity protein receptors as do structurally similar toxins from Staphylococcus aureus. Next, we identified a number of specific amino acid residues that greatly diminish the VCC potency against cells and investigated the interplay between glycan binding and these direct lipid contacts. Finally, we used model membranes to parse the importance of these key residues in lipid and cholesterol binding. Our study provides a complete functional map of the VCC membrane-binding surface and insights into the integration of sugar, lipid, and cholesterol binding interactions.
Collapse
Affiliation(s)
- Swastik De
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, Connecticut 06459
| | - Adele Bubnys
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, Connecticut 06459
| | - Francis Alonzo
- Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Jinsol Hyun
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, Connecticut 06459
| | - Jeffrey W Lary
- Biotechnology-Bioservices Center, University of Connecticut, Storrs, Connecticut 06269
| | - James L Cole
- Biotechnology-Bioservices Center, University of Connecticut, Storrs, Connecticut 06269; Department of Molecular and Cell Biology and Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Rich Olson
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, Connecticut 06459.
| |
Collapse
|
7
|
Khilwani B, Chattopadhyay K. Signaling beyond Punching Holes: Modulation of Cellular Responses by Vibrio cholerae Cytolysin. Toxins (Basel) 2015; 7:3344-58. [PMID: 26308054 PMCID: PMC4549754 DOI: 10.3390/toxins7083344] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/12/2015] [Accepted: 08/14/2015] [Indexed: 01/20/2023] Open
Abstract
Pore-forming toxins (PFTs) are a distinct class of membrane-damaging cytolytic proteins that contribute significantly towards the virulence processes employed by various pathogenic bacteria. Vibrio cholerae cytolysin (VCC) is a prominent member of the beta-barrel PFT (beta-PFT) family. It is secreted by most of the pathogenic strains of the intestinal pathogen V. cholerae. Owing to its potent membrane-damaging cell-killing activity, VCC is believed to play critical roles in V. cholerae pathogenesis, particularly in those strains that lack the cholera toxin. Large numbers of studies have explored the mechanistic basis of the cell-killing activity of VCC. Consistent with the beta-PFT mode of action, VCC has been shown to act on the target cells by forming transmembrane oligomeric beta-barrel pores, thereby leading to permeabilization of the target cell membranes. Apart from the pore-formation-induced direct cell-killing action, VCC exhibits the potential to initiate a plethora of signal transduction pathways that may lead to apoptosis, or may act to enhance the cell survival/activation responses, depending on the type of target cells. In this review, we will present a concise view of our current understanding regarding the multiple aspects of these cellular responses, and their underlying signaling mechanisms, evoked by VCC.
Collapse
Affiliation(s)
- Barkha Khilwani
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences Indian Institute of Science Education and Research Mohali Sector 81, S. A. S. Nagar, Manauli PO 140306, Punjab, India.
| | - Kausik Chattopadhyay
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences Indian Institute of Science Education and Research Mohali Sector 81, S. A. S. Nagar, Manauli PO 140306, Punjab, India.
| |
Collapse
|
8
|
Physicochemical constraints of elevated pH affect efficient membrane interaction and arrest an abortive membrane-bound oligomeric intermediate of the beta-barrel pore-forming toxin Vibrio cholerae cytolysin. Arch Biochem Biophys 2015; 583:9-17. [PMID: 26235489 DOI: 10.1016/j.abb.2015.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 07/24/2015] [Accepted: 07/24/2015] [Indexed: 11/21/2022]
Abstract
Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging cytotoxic protein. VCC causes permeabilization of the target cell membranes by forming transmembrane oligomeric beta-barrel pores. Membrane pore formation by VCC involves following key steps: (i) membrane binding, (ii) formation of a pre-pore oligomeric intermediate, (iii) membrane insertion of the pore-forming motifs, and (iv) formation of the functional transmembrane pore. Membrane binding, oligomerization, and subsequent pore-formation process of VCC appear to be facilitated by multiple regulatory mechanisms that are only partly understood. Here, we have explored the role(s) of the physicochemical constraints, specifically imposed by the elevated pH conditions, on the membrane pore-formation mechanism of VCC. Elevated pH abrogates efficient interaction of VCC with the target membranes, and blocks its pore-forming activity. Under the elevated pH conditions, membrane-bound fractions of VCC remain trapped in the form of abortive oligomeric species that fail to generate the functional transmembrane pores. Such an abortive oligomeric assembly appears to represent a distinct, more advanced intermediate state than the pre-pore state. The present study offers critical insights regarding the implications of the physicochemical constraints for regulating the efficient membrane interaction and pore formation by VCC.
Collapse
|
9
|
Transmembrane oligomeric form of Vibrio cholerae cytolysin triggers TLR2/TLR6–dependent proinflammatory responses in monocytes and macrophages. Biochem J 2015; 466:147-61. [DOI: 10.1042/bj20140718] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We show that the transmembrane oligomeric form of VCC evokes potent proinflammatory responses in the monocytes and macrophages of the innate immune system. VCC oligomer-induced proinflammatory responses depend critically on the TLR2/TLR6-dependent signalling pathways.
Collapse
|
10
|
Vibrio cholerae Cytolysin: Structure–Function Mechanism of an Atypical β-Barrel Pore-Forming Toxin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 842:109-25. [DOI: 10.1007/978-3-319-11280-0_7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
11
|
Rai AK, Chattopadhyay K. Trapping of Vibrio cholerae cytolysin in the membrane-bound monomeric state blocks membrane insertion and functional pore formation by the toxin. J Biol Chem 2014; 289:16978-87. [PMID: 24794872 DOI: 10.1074/jbc.m114.567099] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging cytolytic toxin that belongs to the family of β barrel pore-forming protein toxins. VCC induces lysis of its target eukaryotic cells by forming transmembrane oligomeric β barrel pores. The mechanism of membrane pore formation by VCC follows the overall scheme of the archetypical β barrel pore-forming protein toxin mode of action, in which the water-soluble monomeric form of the toxin first binds to the target cell membrane, then assembles into a prepore oligomeric intermediate, and finally converts into the functional transmembrane oligomeric β barrel pore. However, there exists a vast knowledge gap in our understanding regarding the intricate details of the membrane pore formation process employed by VCC. In particular, the membrane oligomerization and membrane insertion steps of the process have only been described to a limited extent. In this study, we determined the key residues in VCC that are critical to trigger membrane oligomerization of the toxin. Alteration of such key residues traps the toxin in its membrane-bound monomeric state and abrogates subsequent oligomerization, membrane insertion, and functional transmembrane pore-formation events. The results obtained from our study also suggest that the membrane insertion of VCC depends critically on the oligomerization process and that it cannot be initiated in the membrane-bound monomeric form of the toxin. In sum, our study, for the first time, dissects membrane binding from the subsequent oligomerization and membrane insertion steps and, thus, defines the exact sequence of events in the membrane pore formation process by VCC.
Collapse
Affiliation(s)
- Anand Kumar Rai
- From the Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Sector 81, SAS Nagar, Manauli, Mohali, 140306 Punjab, India
| | - Kausik Chattopadhyay
- From the Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Sector 81, SAS Nagar, Manauli, Mohali, 140306 Punjab, India
| |
Collapse
|
12
|
Zhou H, Zhao X, Wu R, Cui Z, Diao B, Li J, Wang D, Kan B, Liang W. Population structural analysis of O1 El Tor Vibrio cholerae isolated in China among the seventh cholera pandemic on the basis of multilocus sequence typing and virulence gene profiles. INFECTION GENETICS AND EVOLUTION 2014; 22:72-80. [PMID: 24448269 DOI: 10.1016/j.meegid.2013.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 12/12/2013] [Accepted: 12/16/2013] [Indexed: 11/25/2022]
Abstract
Serogroup O1 Vibrio cholerae is the most common agents to cause epidemic and pandemic cholera disease. In this study, multilocus sequence typing (MLST) was performed on 160 serogroup O1 strains (including 42 toxigenic and 118 non-toxigenic), and the virulence/fitness gene profiles of 16 loci were further analysed for 60 strains of these. Eighty-four sequence types (STs) with 14 clonal complexes were distinguished, and 29 STs were unique. Except SD19771005, all toxigenic strains were well-separated from the non-toxigenic strains. While a group of non-toxigenic strains clustered closer to the toxigenic strains compared to the other strains. Overall the examined gene loci showed higher presence rates in the toxigenic strains compared to the non-toxigenic strains. It is worth noting that the presence rates of VPI, TLC, VSP-I and VSP-II in the non-toxigenic strains that were clustered closer to the toxigenic strains were much higher compared to the other non-toxigenic strains. Our study indicated the complex population structure of O1 strains, and parts of non-toxigenic strains are genetically more closely related to toxigenic strains than other non-toxigenic strains, suggesting that these strains may have a higher potential for infection with CTXФ in the environment or host intestine and is more efficient to become new pathogenic or epidemic clones.
Collapse
Affiliation(s)
- Haijian Zhou
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China; Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 866 Yuhangtang Road, Hangzhou 310003, People's Republic of China
| | - Xuan Zhao
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China
| | - Rui Wu
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China
| | - Zhigang Cui
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China; Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 866 Yuhangtang Road, Hangzhou 310003, People's Republic of China
| | - Baowei Diao
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China
| | - Jie Li
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China
| | - Duochun Wang
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China; Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 866 Yuhangtang Road, Hangzhou 310003, People's Republic of China
| | - Biao Kan
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China; Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 866 Yuhangtang Road, Hangzhou 310003, People's Republic of China.
| | - Weili Liang
- National Institute for Communicable Disease Control and Prevention, and State Key Laboratory for Infectious Disease Prevention and Control, Chinese Centre for Disease Control and Prevention, 155, Changbai Road, Changping, Beijing 102206, People's Republic of China; Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, 866 Yuhangtang Road, Hangzhou 310003, People's Republic of China.
| |
Collapse
|
13
|
Vaidyanathan MS, Sathyanarayana P, Maiti PK, Visweswariah SS, Ayappa KG. Lysis dynamics and membrane oligomerization pathways for Cytolysin A (ClyA) pore-forming toxin. RSC Adv 2014. [DOI: 10.1039/c3ra45159c] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
14
|
Paul K, Chattopadhyay K. Pre-pore oligomer formation by Vibrio cholerae cytolysin: Insights from a truncated variant lacking the pore-forming pre-stem loop. Biochem Biophys Res Commun 2014; 443:189-93. [DOI: 10.1016/j.bbrc.2013.11.078] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 11/19/2013] [Indexed: 12/26/2022]
|
15
|
Ganguly S, Mukherjee A, Mazumdar B, Ghosh AN, Banerjee KK. The β-prism lectin domain of Vibrio cholerae hemolysin promotes self-assembly of the β-pore-forming toxin by a carbohydrate-independent mechanism. J Biol Chem 2013; 289:4001-8. [PMID: 24356964 DOI: 10.1074/jbc.m113.522284] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vibrio cholerae cytolysin/hemolysin (VCC) is an amphipathic 65-kDa β-pore-forming toxin with a C-terminal β-prism lectin domain. Because deletion or point mutation of the lectin domain seriously compromises hemolytic activity, it is thought that carbohydrate-dependent interactions play a critical role in membrane targeting of VCC. To delineate the contributions of the cytolysin and lectin domains in pore formation, we used wild-type VCC, 50-kDa VCC (VCC(50)) without the lectin domain, and mutant VCC(D617A) with no carbohydrate-binding activity. VCC and its two variants with no carbohydrate-binding activity moved to the erythrocyte stroma with apparent association constants on the order of 10(7) M(-1). However, loss of the lectin domain severely reduced the efficiency of self-association of the VCC monomer with the β-barrel heptamer in the synthetic lipid bilayer from ∼83 to 27%. Notably, inactivation of the carbohydrate-binding activity by the D617A mutation marginally reduced oligomerization to ∼77%. Oligomerization of VCC(50) was temperature-insensitive; by contrast, VCC self-assembly increased with increasing temperature, suggesting that the process is driven by entropy and opposed by enthalpy. Asialofetuin, the β1-galactosyl-terminated glycoprotein inhibitor of VCC-induced hemolysis, promoted oligomerization of 65-kDa VCC to a species that resembled the membrane-inserted heptamer in stoichiometry and morphology but had reduced global amphipathicity. In conclusion, we propose (i) that the β-prism lectin domain facilitated toxin assembly by producing entropy during relocation in the heptamer and (ii) that glycoconjugates inhibited VCC by promoting its assembly to a water-soluble, less amphipathic oligomer variant with reduced ability to penetrate the bilayer.
Collapse
|
16
|
Ikigai H, Otsuru H, Yamamoto K, Shimamura T. Structural Requirements of Cholesterol for Binding toVibrio choleraeHemolysin. Microbiol Immunol 2013; 50:751-7. [PMID: 17053310 DOI: 10.1111/j.1348-0421.2006.tb03848.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cholesterol is necessary for the conversion of Vibrio cholerae hemolysin (VCH) monomers into oligomers in liposome membranes. Using different sterols, we determined the stereochemical structures of the VCH-binding active groups present in cholesterol. The VCH monomers are bound to cholesterol, diosgenin, campesterol, and ergosterol, which have a hydroxyl group at position C-3 (3betaOH) in the A ring and a C-C double bond between positions C-5 and C-6 (C-C Delta(5)) in the B ring. They are not bound to epicholesterol and dihydrocholesterol, which form a covalent link with a 3alphaOH group and a C-C single bond between positions C-5 and C-6, respectively. This result suggests that the 3betaOH group and the C-CDelta(5) bond in cholesterol are required for VCH monomer binding. We further examined VCH oligomer binding to cholesterol. However, this oligomer did not bind to cholesterol, suggesting that the disappearance of the cholesterol-binding potential of the VCH oligomer might be a result of the conformational change caused by the conversion of the monomer into the oligomer. VCH oligomer formation was observed in liposomes containing sterols with the 3betaOH group and the C-C Delta(5) bond, and it correlated with the binding affinity of the monomer to each sterol. Therefore, it seems likely that monomer binding to membrane sterol leads to the assembly of the monomer. However, since oligomer formation was induced by liposomes containing either epicholesterol or dihydrocholesterol, the 3betaOH group and the C-C Delta(5) bond were not essential for conversion into the oligomer.
Collapse
Affiliation(s)
- Hajime Ikigai
- Department of Chemistry and Biochemistry, Suzuka National College of Technology, Suzuka, Mie 510-0294, Japan.
| | | | | | | |
Collapse
|
17
|
Dutta S, Banerjee KK, Ghosh AN. Cryo-electron microscopy reveals the membrane insertion mechanism of V. cholerae hemolysin. J Biomol Struct Dyn 2013; 32:1434-42. [PMID: 24102290 DOI: 10.1080/07391102.2013.823564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Vibrio cholerae hemolysin (HlyA) is a 65 kDa pore-forming toxin which causes lysis of target eukaryotic cells by forming heptameric channels in the plasma membrane. Deletion of the 15 kDa C-terminus β-prism carbohydrate-binding domain generates a 50 kDa truncated variant (HlyA50) with 1000-fold-reduced pore-forming activity. Previously, we showed by cryo-electron microscopy that the two toxin oligomers have central channels, but the 65 kDa toxin oligomer is a seven-fold symmetric structure with bowl-, ring-, and arm-like domains, whereas the 50 kDa oligomer is an asymmetric jar-like heptamer. In the present study, we determined three-dimensional(3D) structures of HlyA and HlyA50 in presence of erythrocyte stroma and observed that interaction of the 65 kDa toxin with the stroma induced a significant decrease in the height of the β-barrel oligomer with a change in conformation of the ring- and arm-like domains of HlyA. These features were absent in interaction of HlyA50 with stroma. We propose that this conformational transition is critical for membrane-insertion of the toxin.
Collapse
Affiliation(s)
- Somnath Dutta
- a Division of Electron Microscopy , National Institute of Cholera and Enteric Diseases , P-33, C.I.T. Road, Scheme-XM, Beleghata, Kolkata , 700010 , India
| | | | | |
Collapse
|
18
|
Los FCO, Randis TM, Aroian RV, Ratner AJ. Role of pore-forming toxins in bacterial infectious diseases. Microbiol Mol Biol Rev 2013; 77:173-207. [PMID: 23699254 PMCID: PMC3668673 DOI: 10.1128/mmbr.00052-12] [Citation(s) in RCA: 308] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pore-forming toxins (PFTs) are the most common bacterial cytotoxic proteins and are required for virulence in a large number of important pathogens, including Streptococcus pneumoniae, group A and B streptococci, Staphylococcus aureus, Escherichia coli, and Mycobacterium tuberculosis. PFTs generally disrupt host cell membranes, but they can have additional effects independent of pore formation. Substantial effort has been devoted to understanding the molecular mechanisms underlying the functions of certain model PFTs. Likewise, specific host pathways mediating survival and immune responses in the face of toxin-mediated cellular damage have been delineated. However, less is known about the overall functions of PFTs during infection in vivo. This review focuses on common themes in the area of PFT biology, with an emphasis on studies addressing the roles of PFTs in in vivo and ex vivo models of colonization or infection. Common functions of PFTs include disruption of epithelial barrier function and evasion of host immune responses, which contribute to bacterial growth and spreading. The widespread nature of PFTs make this group of toxins an attractive target for the development of new virulence-targeted therapies that may have broad activity against human pathogens.
Collapse
Affiliation(s)
| | - Tara M. Randis
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Raffi V. Aroian
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego, La Jolla, California, USA
| | - Adam J. Ratner
- Department of Pediatrics, Columbia University, New York, New York, USA
| |
Collapse
|
19
|
Levan S, De S, Olson R. Vibrio cholerae cytolysin recognizes the heptasaccharide core of complex N-glycans with nanomolar affinity. J Mol Biol 2013; 425:944-57. [PMID: 23274141 PMCID: PMC3578121 DOI: 10.1016/j.jmb.2012.12.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/16/2012] [Accepted: 12/20/2012] [Indexed: 01/30/2023]
Abstract
Pathogens selectively target host cells using adhesion molecules and secreted virulence factors that may utilize protein, lipid, or carbohydrate ligands on the cell surface. The human intestinal pathogen Vibrio cholerae secretes a pore-forming toxin, V.cholerae cytolysin (VCC), which contains two domains that are structurally similar to known carbohydrate-binding proteins. These tandem domains are attached to the carboxy-terminus of the cytolytic domain and contain a β-trefoil fold and a β-prism fold. VCC has been shown to bind glycosylated proteins, and removal of the β-prism domain leads to a large decrease in lytic activity against rabbit erythrocytes. Despite these clues, the identity of the glycan receptors of VCC and the role of glycan binding in toxin activity remain unknown. To better understand this specificity, we used a combination of structural and functional approaches to characterize the carbohydrate-binding activity of the VCC toxin. We first probed the monosaccharide-binding activity of VCC and demonstrated that the toxin exhibits millimolar affinity for aldohexoses. To understand this specificity, we solved the crystal structure of the VCC β-prism domain bound to methyl-α-mannose. Next, we utilized a mammalian glycan screen to determine that the β-prism domain preferentially binds complex N-glycans with a heptasaccharide GlcNAc(4)Man(3) core (NGA2). Fluorescence anisotropy and surface plasmon resonance indicated an approximately 100-nM affinity of the β-prism domain for the heptasaccharide core. Our results suggest that carbohydrate-binding domains on the VCC toxin facilitate high-affinity targeting of mammalian cell membranes, which may contribute to the ability of VCC to lyse cells at picomolar concentrations.
Collapse
Affiliation(s)
- Sophia Levan
- Department of Molecular Biology and Biochemistry, Wesleyan University, 52 Lawn Avenue, Middletown, Connecticut, USA
| | - Swastik De
- Department of Molecular Biology and Biochemistry, Wesleyan University, 52 Lawn Avenue, Middletown, Connecticut, USA
| | - Rich Olson
- Department of Molecular Biology and Biochemistry, Wesleyan University, 52 Lawn Avenue, Middletown, Connecticut, USA
| |
Collapse
|
20
|
Rai AK, Paul K, Chattopadhyay K. Functional mapping of the lectin activity site on the β-prism domain of vibrio cholerae cytolysin: implications for the membrane pore-formation mechanism of the toxin. J Biol Chem 2012; 288:1665-73. [PMID: 23209283 DOI: 10.1074/jbc.m112.430181] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vibrio cholerae cytolysin (VCC) is a prominent member in the family of β-barrel pore-forming toxins. It induces lysis of target eukaryotic cells by forming transmembrane oligomeric β-barrel channels. VCC also exhibits prominent lectin-like activity in interacting with β1-galactosyl-terminated glycoconjugates. Apart from the cytolysin domain, VCC harbors two lectin-like domains: the β-Trefoil and the β-Prism domains; however, precise contribution of these domains in the lectin property of VCC is not known. Also, role(s) of these lectin-like domains in the mode of action of VCC remain obscure. In the present study, we show that the β-Prism domain of VCC acts as the structural scaffold to determine the lectin activity of the protein toward β1-galactosyl-terminated glycoconjugates. Toward exploring the physiological implication of the β-Prism domain, we demonstrate that the presence of the β-Prism domain-mediated lectin activity is crucial for an efficient interaction of the toxin toward the target cells. Our results also suggest that such lectin activity may act to regulate the oligomerization ability of the membrane-bound VCC toxin. Based on the data presented here, and also consistent with the existing structural information, we propose a novel mechanism of regulation imposed by the β-Prism domain's lectin activity, implicated in the process of membrane pore formation by VCC.
Collapse
Affiliation(s)
- Anand Kumar Rai
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Sector 81, SAS Nagar, Manauli, 140306, Punjab, India
| | | | | |
Collapse
|
21
|
Paul K, Chattopadhyay K. Single point mutation inVibrio choleraecytolysin compromises the membrane pore-formation mechanism of the toxin. FEBS J 2012; 279:4039-51. [DOI: 10.1111/j.1742-4658.2012.08809.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 08/08/2012] [Accepted: 08/24/2012] [Indexed: 11/29/2022]
Affiliation(s)
- Karan Paul
- Department of Biological Sciences; Indian Institute of Science Education and Research (IISER) Mohali; SAS Nagar; Manauli; 140306; Punjab; India
| | - Kausik Chattopadhyay
- Department of Biological Sciences; Indian Institute of Science Education and Research (IISER) Mohali; SAS Nagar; Manauli; 140306; Punjab; India
| |
Collapse
|
22
|
Paul K, Chattopadhyay K. Unfolding Distinguishes the Vibrio cholerae Cytolysin Precursor from the Mature Form of the Toxin. Biochemistry 2011; 50:3936-45. [DOI: 10.1021/bi200332g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Karan Paul
- Indian Institute of Science Education and Research (IISER) Mohali, Transit Campus: MGSIPAP Complex, Sector 26, Chandigarh 160019, India
| | - Kausik Chattopadhyay
- Indian Institute of Science Education and Research (IISER) Mohali, Transit Campus: MGSIPAP Complex, Sector 26, Chandigarh 160019, India
| |
Collapse
|
23
|
Igbinosa EO, Okoh AI. Vibrio fluvialis: an unusual enteric pathogen of increasing public health concern. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2010; 7:3628-43. [PMID: 21139853 PMCID: PMC2996184 DOI: 10.3390/ijerph7103628] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 10/08/2010] [Indexed: 01/22/2023]
Abstract
In developing countries, the fraction of treated wastewater effluents being discharged into watersheds have increased over the period of time, which have led to the deteriorations of the qualities of major rivers in developing nations. Consequently, high densities of disease causing bacteria in the watersheds are regularly reported including incidences of emerging Vibrio fluvialis. Vibrio fluvialis infection remains among those infectious diseases posing a potentially serious threat to public health. This paper addresses the epidemiology of this pathogen; pathogenesis of its disease; and its clinical manifestations in humans.
Collapse
Affiliation(s)
- Etinosa O. Igbinosa
- Applied and Environmental Microbiology Research Group (AEMREG), Department of Biochemistry and Microbiology, University of Fort Hare, P/Bag X1314, Alice 5700, South Africa; E-Mail:
| | - Anthony I. Okoh
- Applied and Environmental Microbiology Research Group (AEMREG), Department of Biochemistry and Microbiology, University of Fort Hare, P/Bag X1314, Alice 5700, South Africa; E-Mail:
| |
Collapse
|
24
|
Cho KH, Wang HS, Kim YK. Temperature-dependent hemolytic activity of membrane pore-forming peptide toxin, tolaasin. J Pept Sci 2010; 16:85-90. [PMID: 19960443 DOI: 10.1002/psc.1199] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tolaasin, a pore-forming peptide toxin produced by Pseudomonas tolaasii, causes brown blotch disease on cultivated mushrooms. Hemolysis using red blood cells was measured to evaluate the cytotoxicity of tolaasin. To investigate the mechanism of tolaasin-induced cell disruption, we studied the effect of temperature on the hemolytic process. At 4 degrees Celsius, poor binding of the tolaasin molecules to the erythrocyte membrane was observed and most of the tolaasin molecules stayed in the solution. However, once tolaasin bound to erythrocytes at 37 degrees Celsius and the temperature was decreased, complete hemolysis was observed even at 4 degrees Celsius. These results indicate that tolaasin binding to cell membrane is temperature-sensitive while tolaasin-induced membrane disruption is less sensitive to temperature change. The effect of erythrocyte concentration was measured to understand the membrane binding and pore-forming properties of tolaasin. The percentage of hemolysis measured by both hemoglobin release and cell lysis decreased as erythrocyte concentration increased in the presence of a fixed amount of tolaasin. The result shows that hemolysis is dependent on the amount of tolaasin and multiple binding of tolaasin is required for the hemolysis of a single cell. In analysis of dose-dependence, the hemolysis was proportional to the tenth power of the amount of tolaasin, implying that tolaasin-induced hemolysis can be explained by a multi-hit model.
Collapse
Affiliation(s)
- Kwang-Hyun Cho
- Department of Agricultural Chemistry, Chungbuk National University, Cheongju, Chungbuk, 361-763, Korea
| | | | | |
Collapse
|
25
|
Toma C, Higa N, Koizumi Y, Nakasone N, Ogura Y, McCoy AJ, Franchi L, Uematsu S, Sagara J, Taniguchi S, Tsutsui H, Akira S, Tschopp J, Núñez G, Suzuki T. Pathogenic Vibrio activate NLRP3 inflammasome via cytotoxins and TLR/nucleotide-binding oligomerization domain-mediated NF-kappa B signaling. THE JOURNAL OF IMMUNOLOGY 2010; 184:5287-97. [PMID: 20348425 DOI: 10.4049/jimmunol.0903536] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vibrio vulnificus and Vibrio cholerae are Gram-negative pathogens that cause serious infectious disease in humans. The beta form of pro-IL-1 is thought to be involved in inflammatory responses and disease development during infection with these pathogens, but the mechanism of beta form of pro-IL-1 production remains poorly defined. In this study, we demonstrate that infection of mouse macrophages with two pathogenic Vibrio triggers the activation of caspase-1 via the NLRP3 inflammasome. Activation of the NLRP3 inflammasome was mediated by hemolysins and multifunctional repeat-in-toxins produced by the pathogenic bacteria. NLRP3 activation in response to V. vulnificus infection required NF-kappaB activation, which was mediated via TLR signaling. V. cholerae-induced NLRP3 activation also required NF-kappaB activation but was independent of TLR stimulation. Studies with purified V. cholerae hemolysin revealed that toxin-stimulated NLRP3 activation was induced by TLR and nucleotide-binding oligomerization domain 1/2 ligand-mediated NF-kappaB activation. Our results identify the NLRP3 inflammasome as a sensor of Vibrio infections through the action of bacterial cytotoxins and differential activation of innate signaling pathways acting upstream of NF-kappaB.
Collapse
Affiliation(s)
- Claudia Toma
- Division of Bacterial Pathogenesis, Graduate School of Medicine, University of the Ryukyus, Okinawa
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Harris JR, Palmer M. Cholesterol specificity of some heptameric beta-barrel pore-forming bacterial toxins: structural and functional aspects. Subcell Biochem 2010; 51:579-596. [PMID: 20213559 DOI: 10.1007/978-90-481-8622-8_21] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Apart from the thiol-specific/cholesterol-dependent cytolysin family of toxins (see Chapter 20) there are a number of other unrelated bacterial toxins that also have an affinity for plasma membrane cholesterol. Emphasis is given here on the Vibrio cholerae cytolysin (VCC) and the cytolysins from related Vibrio species. The inhibition of the cytolytic activity of these toxins by prior incubation with extracellular cholesterol or low density lipoprotein emerges as a unifying feature, as does plasma membrane cholesterol depletion. Incubation of VCC with cholesterol produces a heptameric oligomer, which is not equivalent to the pre-pore since it is unable to penetrate the plasma membrane. In structural terms, the precise sequence of VCC monomer binding to membrane, oligomer formation and pore insertion through the bilayer has yet to be fully defined. Several other bacterial toxins have a dependency for cholesterol, although the available data is limited in most cases.
Collapse
Affiliation(s)
- J Robin Harris
- Institute of Zoology, University of Mainz, Mainz, D-55099, Germany.
| | | |
Collapse
|
27
|
Dutta S, Mazumdar B, Banerjee KK, Ghosh AN. Three-dimensional structure of different functional forms of the Vibrio cholerae hemolysin oligomer: a cryo-electron microscopic study. J Bacteriol 2010; 192:169-78. [PMID: 19854900 PMCID: PMC2798276 DOI: 10.1128/jb.00930-09] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Accepted: 10/12/2009] [Indexed: 11/20/2022] Open
Abstract
Vibrio cholerae hemolysin (HlyA) is a 65-kDa water-soluble pore-forming toxin that causes lysis of eukaryotic cells by destroying selective permeability of the plasma membrane bilayer. The HlyA monomer self-assembles on the target cell surface to the more stable beta-barrel amphipathic heptamer, which inserts into the membrane bilayer to form a diffusion channel. Deletion of the 15-kDa beta-prism lectin domain at the C terminus generates a 50-kDa hemolysin variant (HlyA50) with an approximately 1,000-fold decrease in hemolytic activity. Because functional differences are eventually dictated by structural differences, we determined three-dimensional structures of 65- and 50-kDa HlyA oligomers, using cryo-electron microscopy and single-particle methods. Our study clearly shows that the HlyA oligomer has sevenfold symmetry but that the HlyA50 oligomer is an asymmetric molecule. The HlyA oligomer has bowl-like, arm-like, and ring-like domains. The bowl-like domain is coupled with the ring-like domain, and seven side openings are present just beneath the ring-like domain. Although a central channel is present in both HlyA and HlyA50 oligomers, they differ in pore size as well as in shape of the molecules and channel. These structural differences may be relevant to the striking difference in efficiencies of functional channel formation by the two toxin forms.
Collapse
Affiliation(s)
- Somnath Dutta
- Division of Electron Microscopy, Division of Biochemistry, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beleghata, Kolkata 700010, India
| | - Budhaditya Mazumdar
- Division of Electron Microscopy, Division of Biochemistry, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beleghata, Kolkata 700010, India
| | - Kalyan K. Banerjee
- Division of Electron Microscopy, Division of Biochemistry, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beleghata, Kolkata 700010, India
| | - Amar N. Ghosh
- Division of Electron Microscopy, Division of Biochemistry, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beleghata, Kolkata 700010, India
| |
Collapse
|
28
|
Tagami Y, Narita T, Ikigai H, Oishi Y. Penetration behavior of Vibrio cholerae hemolysin into (DMPC/cholesterol) mixed monolayer. Colloids Surf A Physicochem Eng Asp 2009. [DOI: 10.1016/j.colsurfa.2009.04.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Löhner S, Walev I, Boukhallouk F, Palmer M, Bhakdi S, Angela V. Pore formation by
Vibrio cholerae
cytolysin follows the same archetypical mode as β‐barrel toxins from gram‐positive organisms. FASEB J 2009; 23:2521-8. [DOI: 10.1096/fj.08-127688] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Sabine Löhner
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Iwan Walev
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Michael Palmer
- Department of ChemistryUniversity of WaterlooWaterlooOntarioCanada
| | - Sucharit Bhakdi
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Valeva Angela
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| |
Collapse
|
30
|
Vibrio cholerae cytolysin is essential for high enterotoxicity and apoptosis induction produced by a cholera toxin gene-negative V. cholerae non-O1, non-O139 strain. Microb Pathog 2008; 44:118-28. [DOI: 10.1016/j.micpath.2007.08.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Accepted: 08/10/2007] [Indexed: 12/21/2022]
|
31
|
Valeva A, Walev I, Weis S, Boukhallouk F, Wassenaar TM, Bhakdi S. Pro-inflammatory feedback activation cycle evoked by attack of Vibrio cholerae cytolysin on human neutrophil granulocytes. Med Microbiol Immunol 2007; 197:285-93. [PMID: 17882454 DOI: 10.1007/s00430-007-0061-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Indexed: 11/28/2022]
Abstract
Vibrio cholerae cytolysin (VCC) is a pore-forming toxin that is secreted in precursor form (pro-VCC) and requires proteolytic cleavage in order to attain membrane-permeabilizing properties. Pro-VCC can be activated both in solution and membrane-bound state. Processing of membrane-bound pro-VCC can in turn be achieved through the action of both cell-associated and soluble proteases. The current investigation describes the interaction of VCC with human neutrophil granulocytes. It is shown that pro-VCC binds to these cells and is cleaved by cell-bound serine proteases. Membrane permeabilization leads to granulocyte activation, as witnessed by the generation of reactive oxygen metabolites and liberation of granule constituents. A mutant toxin with unaltered binding properties but devoid of pore-forming activity did not elicit these effects. The secreted proteases cleave and activate further bound- and non-bound pro-VCC. A positive feedback loop is thus created that results in enhanced cytotoxicity towards both the targeted granulocytes and towards bystander cells that are not primarily killed by the protoxin. Thus, activation of neutrophil granulocytes by VCC fuels a positive feedback cycle that will cripple immune defence, augment inflammation, and enhance the cytotoxic action of the toxin on neighbouring tissue cells.
Collapse
Affiliation(s)
- Angela Valeva
- Institute of Medical Microbiology and Hygiene, University of Mainz, Augustusplatz, Mainz, Germany.
| | | | | | | | | | | |
Collapse
|
32
|
From C, Hormazabal V, Hardy SP, Granum PE. Cytotoxicity in Bacillus mojavensis is abolished following loss of surfactin synthesis: implications for assessment of toxicity and food poisoning potential. Int J Food Microbiol 2007; 117:43-9. [PMID: 17467096 DOI: 10.1016/j.ijfoodmicro.2007.01.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 01/15/2007] [Accepted: 01/22/2007] [Indexed: 11/17/2022]
Abstract
Bacillus subtilis and the closely related species Bacillus pumilus and Bacillus licheniformis have periodically been suggested to play a role in the aetiology of food poisoning despite the fact that the organisms do not possess the genes associated with enteropathogenicity in Bacillus cereus. We show here that Bacillus mojavensis, an organism closely related to B. subtilis, is able to produce toxic components which identify as a complex of three different surfactin analogues. These cyclic lipopeptides were soluble in methanol, heat stable after treatment in a boiling water bath for 10 min, resistant to enzymatic degradation by pepsin, trypsin, endoprotease V8 and proteinase K and formed pores in planar lipid bilayers. They were cytotoxic when tested in a series of commonly used laboratory cytotoxicity assays, namely, lactate dehydrogenase release, haemolysis, inhibition of both protein synthesis in Vero cells and motility in boar sperm. We show that such in vitro markers of enterotoxicity are due entirely to production of cyclic lipopeptides since deletion of sfp, a gene essential for surfactin synthesis which abolished the cytotoxicity to Vero cells, boar sperm motility and haemolytic activity. Thus, the relevance of cyclic lipopeptides as food poisoning toxins needs to be evaluated in assays other than the cell cytotoxicity assays in common use.
Collapse
Affiliation(s)
- C From
- Department of Food Safety and Infection Biology, Norwegian School of Veterinary Science, NO-0033 Oslo, Norway
| | | | | | | |
Collapse
|
33
|
Krasilnikov OV, Merzlyak PG, Lima VLM, Zitzer AO, Valeva A, Yuldasheva LN. Pore formation by Vibrio cholerae cytolysin requires cholesterol in both monolayers of the target membrane. Biochimie 2007; 89:271-7. [PMID: 17303303 DOI: 10.1016/j.biochi.2006.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Accepted: 12/18/2006] [Indexed: 11/19/2022]
Abstract
Vibrio cholerae cytolysin (VCC) forms oligomeric transmembrane pores in cholesterol-rich membranes. To better understand this process, we used planar bilayer membranes. In symmetric membranes, the rate of the channel formation by VCC has a superlinear dependency on the cholesterol membrane fraction. Thus, more than one cholesterol molecule can facilitate VCC-pore formation. In asymmetric membranes, the rate of pore formation is limited by the leaflet with the lower cholesterol content. Methyl-beta-cyclodextrin, which removes cholesterol from membranes, rapidly inhibits VCC pore formation, even when it is added to the side opposite that of VCC addition. The results suggest that cholesterol in both membrane leaflets aid VCC-pore formation and that either leaflet can function as a kinetic bottleneck with respect to the rate of pore-formation.
Collapse
Affiliation(s)
- Oleg V Krasilnikov
- Laboratory of Membrane Biophysics, Department of Biophysics and Radiobiology, Federal University of Pernambuco, Av. prof. Moraes Rego, S/N, 50670-901 Recife, PE, Brazil.
| | | | | | | | | | | |
Collapse
|
34
|
Pantano S, Montecucco C. A molecular model of the Vibrio cholerae cytolysin transmembrane pore. Toxicon 2005; 47:35-40. [PMID: 16330061 DOI: 10.1016/j.toxicon.2005.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2005] [Revised: 08/31/2005] [Accepted: 09/22/2005] [Indexed: 01/05/2023]
Abstract
The cytotoxic activity of some of the most pathogenic strains of Vibrio cholerae is associated with a cytolysin protein (VCC), which forms oligomeric transmembrane pores and changes the permeability of intestinal cells. We present here a model structure of the transmembrane pore of VCC based on sequence comparison with other pore-forming toxins. VCC is suggested to form a transmembrane beta-barrel pore with a relatively large trans vestibule region. Calculations of the electrostatic profile within the pore lumen provide a rationale for the low conductance and selectivity of the VCC ion channel.
Collapse
Affiliation(s)
- Sergio Pantano
- Venetian Institute of Molecular Medicine (VIMM),Via Orus 2, 35129, Padova, Italy.
| | | |
Collapse
|
35
|
Olson R, Gouaux E. Crystal Structure of the Vibrio cholerae Cytolysin (VCC) Pro-toxin and its Assembly into a Heptameric Transmembrane Pore. J Mol Biol 2005; 350:997-1016. [PMID: 15978620 DOI: 10.1016/j.jmb.2005.05.045] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Revised: 05/10/2005] [Accepted: 05/12/2005] [Indexed: 11/25/2022]
Abstract
Pathogenic Vibrio cholerae secrete V. cholerae cytolysin (VCC), an 80 kDa pro-toxin that assembles into an oligomeric pore on target cell membranes following proteolytic cleavage and interaction with cell surface receptors. To gain insight into the activation and targeting activities of VCC, we solved the crystal structure of the pro-toxin at 2.3A by X-ray diffraction. The core cytolytic domain of VCC shares a fold similar to the staphylococcal pore-forming toxins, but in VCC an amino-terminal pro-domain and two carboxy-terminal lectin domains decorate the cytolytic domain. The pro-domain masks a protomer surface that likely participates in inter-protomer interactions in the cytolytic oligomer, thereby explaining why proteolytic cleavage and movement of the pro-domain is necessary for toxin activation. A single beta-octyl glucoside molecule outlines a possible receptor binding site on one lectin domain, and removal of this domain leads to a tenfold decrease in lytic activity toward rabbit erythrocytes. VCC activated by proteolytic cleavage assembles into an oligomeric species upon addition of soybean asolectin/cholesterol liposomes and this oligomer was purified in detergent micelles. Analytical ultracentrifugation and crystallographic analysis indicate that the resulting VCC oligomer is a heptamer. Taken together, these studies define the architecture of a pore forming toxin and associated lectin domains, confirm the stoichiometry of the assembled oligomer as heptameric, and suggest a common mechanism of assembly for staphylococcal and Vibrio cytolytic toxins.
Collapse
Affiliation(s)
- Rich Olson
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 W. 168th Street, New York, NY 10032, USA
| | | |
Collapse
|
36
|
Valeva A, Walev I, Boukhallouk F, Wassenaar TM, Heinz N, Hedderich J, Lautwein S, Möcking M, Weis S, Zitzer A, Bhakdi S. Identification of the membrane penetrating domain of Vibrio cholerae cytolysin as a β-barrel structure. Mol Microbiol 2005; 57:124-31. [PMID: 15948954 DOI: 10.1111/j.1365-2958.2005.04684.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vibrio cholerae cytolysin (VCC) is an oligomerizing pore-forming toxin that is related to cytolysins of many other Gram-negative organisms. VCC contains six cysteine residues, of which two were found to be present in free sulphydryl form. The positions of two intramolecular disulphide bonds were mapped, and one was shown to be essential for correct folding of protoxin. Mutations were created in which the two free cysteines were deleted, so that single cysteine substitution mutants could be generated for site-specific labelling. Employment of polarity-sensitive fluorophores identified amino acid side-chains that formed part of the pore-forming domain of VCC. The sequence commenced at residue 311, and was deduced to form a beta-barrel in the assembled oligomer with the subsequent odd-numbered residues facing the lipid bilayer and even-numbered residues facing the lumen. Pro328/Lys329 were tentatively identified as the position at which the sequence turns back into the membrane and where the antiparallel beta-strand commences. This was deduced from fluorimetric analyses combined with experiments in which the pore was reversibly occluded by derivatization of sulphydryl groups with a bulky moiety. Our data support computer-based predictions that the membrane-permeabilizing amino acid sequence of VCC is homologous to the beta-barrel-forming sequence of staphylococcal cytolysins and identify the beta-barrel as a membrane-perforating structure that is highly conserved in evolution.
Collapse
Affiliation(s)
- Angela Valeva
- Institute of Medical Microbiology and Hygiene, University of Mainz, Augustusplatz, D55101 Mainz, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Galdiero S, Gouaux E. High resolution crystallographic studies of alpha-hemolysin-phospholipid complexes define heptamer-lipid head group interactions: implication for understanding protein-lipid interactions. Protein Sci 2005; 13:1503-11. [PMID: 15152085 PMCID: PMC2279993 DOI: 10.1110/ps.03561104] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The alpha-hemolysin is an archetypal pore-forming protein that is secreted from Staphylococcus aureus as a water-soluble monomer. When the monomer binds to the membrane of a susceptible cell, the membrane-bound molecules assemble into the lytic heptamer. Although a bilayer or a bilayer-like environment are essential to toxin assembly, there is no high resolution information on toxin-phospholipid complexes. We have determined the structures of detergent-solubilized alpha-hemolysin heptamer bound to glycerophosphocholine or dipropanoyl glycerophosphocholine at 1.75-1.80 A resolution and 110 K. The phosphocholine head group binds to each subunit in a crevice between the rim and the stem domains. The quaternary ammonium group interacts primarily with aromatic residues, whereas the phosphodiester moiety interacts with a conserved arginine residue. These structures provide a molecular basis for understanding why alpha-hemolysin preferentially assembles on membranes comprised of phosphocholine lipids.
Collapse
Affiliation(s)
- Stefania Galdiero
- Howard Hughes Medical Institute and Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA.
| | | |
Collapse
|
38
|
Valeva A, Walev I, Weis S, Boukhallouk F, Wassenaar TM, Endres K, Fahrenholz F, Bhakdi S, Zitzer A. A cellular metalloproteinase activates Vibrio cholerae pro-cytolysin. J Biol Chem 2004; 279:25143-8. [PMID: 15066987 DOI: 10.1074/jbc.m313913200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many strains of Vibrio cholerae produce a cytolysin (VCC) that forms oligomeric transmembrane pores in animal cells. The molecule is secreted as a procytolysin (pro-VCC) of 79 kDa that must be cleaved at the N terminus to generate the active 65-kDa toxin. Processing can occur in solution, and previous studies have described the action of mature VCC thus generated. However, little is known about the properties of pro-VCC itself. In this study, it is shown that pro-VCC exist as a monomer in solution and binds as a monomer to eukaryotic cells. Bound pro-VCC can then be activated either by exogenous, extracellular, or by endogenous, cell-bound proteases. In both cases, cleavage generates the 65-kDa VCC that oligomerizes to form transmembrane pores. A wide variety of exogenous proteinases can mediate activation. In contrast, the activating cellular protease is selectively inhibited by the hydroxamate inhibitor TAPI, and thus probable candidates are members of the ADAM-metalloproteinase family. Furin, MMP-2, MMP-9, and serine proteinases were excluded. Cells over-expressing ADAM-17, also known as tumor necrosis factor alpha converting enzyme, displayed increased activation of VCC, and knockout cells lacking ADAM-17 had a markedly decreased capacity to cleave the protoxin. The possibility is raised that pro-VCC is targeted to membrane sites that selectively contain or are accessible to cellular ADAM-metalloproteinases. Although many microbial toxins are activated by furin, this is the first evidence for processing by a cellular metalloproteinase. We identified ADAM-17 as a potent activator of pro-VCC.
Collapse
Affiliation(s)
- Angela Valeva
- Institute of Medical Microbiology and Hygiene, University of Mainz, D55101 Mainz, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Laohachai KN, Bahadi R, Hardo MB, Hardo PG, Kourie JI. The role of bacterial and non-bacterial toxins in the induction of changes in membrane transport: implications for diarrhea. Toxicon 2003; 42:687-707. [PMID: 14757199 DOI: 10.1016/j.toxicon.2003.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bacterial toxins induce changes in membrane transport which underlie the loss of electrolyte homeostasis associated with diarrhea. Bacterial- and their secreted toxin-types which have been linked with diarrhea include: (a) Vibrio cholerae (cholera toxin, E1 Tor hemolysin and accessory cholera enterotoxin); (b) Escherichia coli (heat stable enterotoxin, heat-labile enterotoxin and colicins); (c) Shigella dysenteriae (shiga-toxin); (d) Clostridium perfringens (C. perfringens enterotoxin, alpha-toxin, beta-toxin and theta-toxin); (e) Clostridium difficile (toxins A and B); (f) Staphylococcus aureus (alpha-haemolysin); (g) Bacillus cereus (cytotoxin K and haemolysin BL); and (h) Aeromonas hydrophila (aerolysin, heat labile cytotoxins and heat stable cytotoxins). The mechanisms of toxin-induced diarrhea include: (a) direct effects on ion transport in intestinal epithelial cells, i.e. direct toxin interaction with intrinsic ion channels in the membrane and (b) indirect interaction with ion transport in intestinal epithelial cells mediated by toxin binding to a membrane receptor. These effects consequently cause the release of second messengers, e.g. the release of adenosine 3',5'-cyclic monophosphate/guanosine 3',5'-monophosphate, IP(3), Ca2+ and/or changes in second messengers that are the result of toxin-formed Ca2+ and K+ permeable channels, which increase Ca2+ flux and augment changes in Ca2+ homeostasis and cause depolarisation of the membrane potential. Consequently, many voltage-dependent ion transport systems, e.g. voltage-dependent Ca2+ influx, are affected. The toxin-formed ion channels may act as a pathway for loss of fluid and electrolytes. Although most of the diarrhea-causing toxins have been reported to act via cation and anion channel formation, the properties of these channels have not been well studied, and the available biophysical properties that are needed for the characterization of these channels are inadequate.
Collapse
Affiliation(s)
- Karina N Laohachai
- Membrane Transport Group, Department of Chemistry, Building 33, The Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia
| | | | | | | | | |
Collapse
|
40
|
Montoya M, Gouaux E. Beta-barrel membrane protein folding and structure viewed through the lens of alpha-hemolysin. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1609:19-27. [PMID: 12507754 DOI: 10.1016/s0005-2736(02)00663-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The beta-barrel is a transmembrane structural motif commonly encountered in bacterial outer membrane proteins and pore-forming toxins (PFTs). Alpha-hemolysin (alphaHL) is a cytotoxin secreted by Staphylococcus aureus that assembles from a water-soluble monomer to form a membrane-bound heptameric beta-barrel on the surface of susceptible cells, perforating the cell membranes, leading to cell death and lysis. The mechanism of heptamer assembly, which has been studied extensively, occurs in a stepwise manner, and the structures of the initial, monomeric form and final, membrane-embedded pore are known. The toxin's ability to assemble from an aqueous, hydrophilic species to a membrane-inserted oligomer is of interest in understanding the assembly of PFTs in particular and the folding and structure of beta-barrel membrane proteins in general. Here we review the structures of the monomeric and heptamer states of LukF and alphaHL, respectively, the mechanism of toxin assembly, and the relationships between alphaHL and nontoxin beta-barrel membrane proteins.
Collapse
Affiliation(s)
- Michelle Montoya
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA.
| | | |
Collapse
|
41
|
Pichel M, Rivas M, Chinen I, Martín F, Ibarra C, Binsztein N. Genetic diversity of Vibrio cholerae O1 in Argentina and emergence of a new variant. J Clin Microbiol 2003; 41:124-34. [PMID: 12517837 PMCID: PMC149600 DOI: 10.1128/jcm.41.1.124-134.2003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The genetic diversity of Vibrio cholerae O1 strains from Argentina was estimated by random amplified polymorphic DNA (RAPD) analysis and pulsed-field gel electrophoresis (PFGE). Twenty-nine isolates carrying the virulence genes ctxA, zot, ace, and tcpA appeared to represent a single clone by both typing methods; while 11 strains lacking these virulence genes exhibited several heterogeneous RAPD and PFGE patterns. Among the last group, a set of isolates from the province Tucumán showed a single RAPD pattern and four closely related PFGE profiles. These strains, isolated from patients with diarrhea, did not produce the major V. cholerae O1 virulence determinants, yet cell supernatants of these isolates caused a heat-labile cytotoxic effect on Vero and Y-1 cells and elicited significant variations on the water flux and short-circuit current in human small intestine mounted in an Ussing chamber. All these effects were completely abolished by incubation with a specific antiserum against El Tor hemolysin, suggesting that this virulence factor was responsible for the toxic activity on both the epithelial cells and the small intestine specimens and may hence be involved in the development of diarrhea. We propose "Tucumán variant" as the designation for this new cluster of cholera toxin-negative V. cholerae O1 strains.
Collapse
Affiliation(s)
- Mariana Pichel
- Departamento de Bacteriología, Instituto Nacional de Enfermedades Infecciosas, ANLIS Dr. Carlos G. Malbrán, Ministerio de Salud, 1281 Capital Federal, Argentina
| | | | | | | | | | | |
Collapse
|
42
|
Han JH, Lee JH, Choi YH, Park JH, Choi TJ, Kong IS. Purification, characterization and molecular cloning of Vibrio fluvialis hemolysin. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1599:106-14. [PMID: 12479411 DOI: 10.1016/s1570-9639(02)00407-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hemolysin of Vibrio fluvialis (VFH) was purified from culture supernatants by ammonium sulfate precipitation and successive column chromatographies on DEAE-cellulose and Mono-Q. N-terminal amino acid sequences of the purified VFH were determined. The purified protein exhibited hemolytic activity on many mammalian erythrocytes with rabbit erythrocytes being the most sensitive to VFH. Activity of the native VFH was inhibited by the addition of Zn2+, Ni2+, Cd2+ and Cu2+ ions at low concentrations. Pores formed on rabbit erythrocytes were approximately 2.8-3.7 nm in diameter, as demonstrated by osmotic protection assay. Nucleotide sequence analysis of the vfh gene revealed an open reading frame (ORF) consisting of 2200 bp which encodes a protein of 740 amino acids with a molecular weight of 82 kDa. Molecular weight of the purified VFH was estimated to be 79 kDa by SDS-PAGE and N-terminal amino acid sequence revealed that the 82 kDa prehemolysin is synthesized in the cytoplasm and is then secreted into the extracellular environment as the 79 kDa mature hemolysin after cleavage of 25 N-terminal amino acids. Deletion of 70 amino acids from the C-terminus exhibited a smaller hemolytic activity, while deletion of 148 C-terminal amino acids prevented hemolytic activity.
Collapse
Affiliation(s)
- Jeong-Hyun Han
- Department of Biotechnology and Bioengineering, Pukyong National University, Pusan, 608-737, South Korea
| | | | | | | | | | | |
Collapse
|
43
|
Harris JR, Bhakdi S, Meissner U, Scheffler D, Bittman R, Li G, Zitzer A, Palmer M. Interaction of the Vibrio cholerae cytolysin (VCC) with cholesterol, some cholesterol esters, and cholesterol derivatives: a TEM study. J Struct Biol 2002; 139:122-35. [PMID: 12406694 DOI: 10.1016/s1047-8477(02)00563-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Vibrio cholerae cytolysin (VCC) 63-kDa monomer has been shown to interact in aqueous suspension with cholesterol microcystals to produce a ring/pore-like heptameric oligomer approximately 8 nm in outer diameter. Transmission electron microscopy data were produced from cholesterol samples adsorbed to carbon support films, spread across the holes of holey carbon films, and negatively stained with ammonium molybdate. The VCC oligomers initially attach to the edge of the stacked cholesterol bilayers and with increasing time cover the two planar surfaces. VCC oligomers are also released into solution, with some tendency to cluster, possibly via the hydrophobic membrane-spanning domain. At the air/water interface, the VCC oligomers are likely to be selectively oriented with the hydrophobic domain facing the air. Despite some molecular disorder/plasticity within the oligomers, multivariate statistical analysis and rotational self-correlation using IMAGIC-5 strongly suggest the presence of sevenfold rotational symmetry. To correlate the electron microscopy data with on-going biochemical and permeability studies using liposomes of varying lipid composition, the direct interaction of VCC with several cholesterol derivatives and other steroids has been examined. 19-Hydroxycholesterol and 7 beta-hydroxycholesterol both induce VCC oligomerization. beta-Estradiol, which does not possess an aliphatic side chain, also efficiently induces VCC oligomer formation, as does cholesteryl acetate. Cholesteryl stearate and oleate and the C22 (2-trifluoroacetyl)naphthyloxy analogue of cholesterol fail to induce VCC oligomerization, but binding of the monomer to the surface of these steroids does occur. Stigmasterol has little tendency to induce oligomer formation, and oligomers are largely confined to the edge of the bilayers; ergosterol has even less oligomerization ability. Attempts to solubilize and stabilize the VCC oligomers from cholesterol suspensions have been pursued using the neutral surfactant octylglucoside. Although individual solubilized oligomers have been defined which exhibit a characteristic cytolysin channel conformation in the side-on orientation, a tendency remains for the oligomers to cluster via their hydrophobic domains.
Collapse
Affiliation(s)
- J Robin Harris
- Institute of Zoology, University of Mainz, 55099 Mainz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Moschioni M, Tombola F, de Bernard M, Coelho A, Zitzer A, Zoratti M, Montecucco C. The Vibrio cholerae haemolysin anion channel is required for cell vacuolation and death. Cell Microbiol 2002; 4:397-409. [PMID: 12102686 DOI: 10.1046/j.1462-5822.2002.00199.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Several strains of Vibrio cholerae secrete a haemolytic toxin of 63 kDa, termed V. cholerae cytolysin (VCC). This toxin causes extensive vacuolation and death of cells in culture and forms an anion-selective channel in planar lipid bilayers and in cells. Here, we identify inhibitors of the VCC anion channel and show that the formation of the anion channel is necessary for the development of the vacuoles and for the cell death induced by this toxin. Using markers of cell organelles, we show that vacuoles derive from different intracellular compartments and we identify the contribution of late endosomes and of the trans-Golgi network in vacuole biogenesis.
Collapse
Affiliation(s)
- Monica Moschioni
- Centro CNR Biomembrane and Dipartimento di Scienze Biomediche Sperimentali, Università di Padova, Via G. Colombo 3, 35121, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
45
|
Kwon KB, Yang JY, Ryu DG, Rho HW, Kim JS, Park JW, Kim HR, Park BH. Vibrio vulnificus cytolysin induces superoxide anion-initiated apoptotic signaling pathway in human ECV304 cells. J Biol Chem 2001; 276:47518-23. [PMID: 11591724 DOI: 10.1074/jbc.m108645200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previous studies showed that exposure to Vibrio vulnificus cytolysin (VVC) caused characteristic morphologic changes and dysfunction of vascular structures in lung. VVC showed cytotoxicity for mammalian cells in culture and acted as a vascular permeability factor. In this study, the underlying mechanisms of VVC-induced cytotoxicity was investigated on ECV304 cell, a human vascular endothelial cell line. When cells were exposed to 0.4 hemolytic units (HU) of VVC, consecutive apoptotic events were observed; the elevation of superoxide anion (O (-.)(2)), the release of cytochrome c, the activation of caspase-3, the cleavage of poly(ADP-ribose) polymerase, and the DNA fragmentation. The pretreatment with 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (TEMPO), O(-.) 2) scavenger, completely abolished O(-.)(2) levels and downstream apoptotic events. Moreover, pretreatment with cyclosporin A (CsA), a mitochondrial permeability transition inhibitor, was capable of attenuating O(-.)(2)-mediated cytochrome c release and caspase-3 activation, and consequent apoptosis. Apoptosis, as demonstrated by oligonucleosomal DNA fragmentation and fluorescence microscopy, was induced 24 h after VVC treatment, which was also prevented by caspase-3 inhibitor, Ac-DEVD-CHO. Caspase-1 inhibitor, Ac-YVAD-CHO, did not protect ECV 304 cells from apoptosis. These results suggest a scenario where VVC-induced apoptosis is triggered by the generation of O(-.)(2), release of cytochrome c from mitochondria, activation of caspase-3, degradation of poly(ADP-ribose) polymerase, and DNA fragmentation. The induction of apoptosis in endothelial cells by VVC may provide a pivotal mechanism for understanding the pathophysiology of septicemia.
Collapse
Affiliation(s)
- K B Kwon
- Department of Physiology, School of Oriental Medicine, Won-Kwang University, Iksan 570-749, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Yuldasheva LN, Merzlyak PG, Zitzer AO, Rodrigues CG, Bhakdi S, Krasilnikov OV. Lumen geometry of ion channels formed by Vibrio cholerae EL Tor cytolysin elucidated by nonelectrolyte exclusion. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1512:53-63. [PMID: 11334624 DOI: 10.1016/s0005-2736(01)00302-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vibrio cholerae EL Tor cytolysin, a water-soluble protein with a molecular mass of 63 kDa, forms small pores in target cell membranes. In this communication, planar lipid bilayers under voltage clamp conditions were used to investigate the geometric properties of the pores. It was established that all cytolysin channels were inserted into membranes with the same orientation. Sharp asymmetry in the I-V curve of fully open cytolysin channels persisting at high electrolyte concentrations indicated asymmetry in the geometry of the channel lumen. Using the nonelectrolyte exclusion method, evidence was obtained that the cis opening of the channel had a larger diameter (< or = 1.9 nm) than the trans opening (< or = 1.6 nm). The channel lumen appeared constricted, with a diameter of < or = 1.2 nm. Cup-shaped lumen geometry was deduced for both channel openings, which appeared to be connected to each other via a central narrow part. The latter contributed significantly to the total electrical resistance and determined the discontinuous character of channel filling with nonelectrolytes. Comparisons of the properties of pores formed by cytolysins of two V. cholerae biotypes (EL Tor and non-O1) indicated that the two ion channels possessed a similar geometry.
Collapse
Affiliation(s)
- L N Yuldasheva
- Laboratory of Membrane Biophysics, Center of Biological Sciences, Department of Biophysics and Radiobiology, Federal University of Pernambuco, Recife, PE, Brazil
| | | | | | | | | | | |
Collapse
|
47
|
Zitzer A, Harris JR, Kemminer SE, Zitzer O, Bhakdi S, Muething J, Palmer M. Vibrio cholerae cytolysin: assembly and membrane insertion of the oligomeric pore are tightly linked and are not detectably restricted by membrane fluidity. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1509:264-74. [PMID: 11118538 DOI: 10.1016/s0005-2736(00)00303-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemolytic strains of Vibrio cholerae secrete a cytolysin that, upon binding as a monomer, forms pentameric pores in animal cell membranes. Pore formation is inhibited at low temperature and in the absence of cholesterol. We here posed the following questions: firstly, can oligomerization be observed in the absence of pore formation? Secondly, is membrane fluidity responsible for the effect of temperature or of cholesterol upon pore formation? The first issue was approached by chemical cross-linking, by electrophoretic heteromer analysis, and by electron microscopy. None of these methods yielded any evidence of a non-lytic pre-pore oligomer. The second question was addressed by the use of two susceptible liposome models, consisting of cholesterol admixed to bovine brain lipids and to asolectin, respectively. The two liposome species clearly differed in membrane fluidity as judged by diphenylhexatriene fluorescence polarization. Nevertheless, their permeabilization by the cytolysin decreased with temperature in a closely parallel fashion, virtually vanishing at 5 degrees C. Omission of cholesterol from the liposomes uniformly led to an increase in membrane fluidity but prevented permeabilization by the cytolysin. The effects of temperature and of cholesterol upon cytolysin activity are thus not mediated by fluidization of the target membrane. The findings of our study distinguish V. cholerae cytolysin from several previously characterized pore-forming toxins.
Collapse
Affiliation(s)
- A Zitzer
- Institute of Medical Microbiology, University of Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Lesieur C, Frutiger S, Hughes G, Kellner R, Pattus F, van der Goot FG. Increased stability upon heptamerization of the pore-forming toxin aerolysin. J Biol Chem 1999; 274:36722-8. [PMID: 10593978 DOI: 10.1074/jbc.274.51.36722] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aerolysin is a bacterial pore-forming toxin that is secreted as an inactive precursor, which is then processed at its COOH terminus and finally forms a circular heptameric ring which inserts into membranes to form a pore. We have analyzed the stability of the precursor proaerolysin and the heptameric complex. Equilibrium unfolding induced by urea and guanidinium hydrochloride was monitored by measuring the intrinsic tryptophan fluorescence of the protein. Proaerolysin was found to unfold in two steps corresponding to the unfolding of the large COOH-terminal lobe followed by the unfolding of the small NH(2)-terminal domain. We show that proaerolysin contains two disulfide bridges which strongly contribute to the stability of the toxin and protect it from proteolytic attack. The stability of aerolysin was greatly enhanced by polymerization into a heptamer. Two regions of the protein, corresponding to amino acids 180-307 and 401-427, were identified, by limited proteolysis, NH(2)-terminal sequencing and matrix-assisted laser desorption ionization-time of flight, as being responsible for stability and maintenance of the heptamer. These regions are presumably involved in monomer/monomer interactions in the heptameric protein and are exclusively composed of beta structure. The stability of the aerolysin heptamer is reminiscent of that of pathogenic, fimbrial protein aggregates found in a variety of neurodegenerative diseases.
Collapse
Affiliation(s)
- C Lesieur
- Department of Biochemistry, University of Geneva, 30 quai E. Ansermet, 1211 Genève 4, Switzerland
| | | | | | | | | | | |
Collapse
|
49
|
Zhang D, Honda T. Disappearance of glyceraldehyde 3-phosphate dehydrogenase from erythrocyte membrane by hemolysis with thermostable direct hemolysin of Vibrio parahaemolyticus or Vibrio cholerae El Tor hemolysin. Microbiol Immunol 1999; 43:303-5. [PMID: 10338202 DOI: 10.1111/j.1348-0421.1999.tb02408.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
It is believed that the thermostable direct hemolysin (TDH) of Vibrio parahaemolyticus and El Tor hemolysin (ETH) of Vibrio cholerae damage erythrocytes and other cells by acting as pore-forming toxins. In this study, we found that a protein band with a molecular weight of 37,000 daltons specifically disappeared from erythrocyte membrane after hemolysis by TDH and ETH, but not after hypotonic hemolysis. The 37 kDa band was identified as glyceraldehyde 3-phosphate dehydrogenase (G3PD), a glycolytic enzyme, based on N-terminal 14 amino acid sequencing. The role of G3PD in hemolysis is discussed.
Collapse
Affiliation(s)
- D Zhang
- Department of Bacterial Infections, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.
| | | |
Collapse
|
50
|
Kim JS, Chae MR, Chang K, Park KH, Rho HW, Park BH, Park JW, Kim HR. Cytotoxicity of Vibrio vulnificus cytolysin on rat peritoneal mast cells. Microbiol Immunol 1999; 42:837-43. [PMID: 10037218 DOI: 10.1111/j.1348-0421.1998.tb02359.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Histamine has been thought to be a permeability enhancing factor in Vibrio vulnificus infection. The injection of living bacteria or purified V. vulnificus cytolysin (VVC) can cause lethality in mice by inducing hemoconcentration and increased vascular permeability. In the present study, we tried to identify whether histamine release causes the increased vascular permeability that is responsible for the lethal effect of VVC. Treatment of rat peritoneal mast cells with high concentrations of VVC caused the release of whole cellular histamine and lactate dehydrogenase (LDH). At concentrations less than 10 HU/ml, histamine and LDH were not released whereas preloaded 2-deoxy-D-glucose was rapidly effluxed with the concomitant decrease in cellular ATP. VVC-treated mast cells were refractory to the stimulation of histamine secretion by Compound 48/80 but remained fully responsive to Ca2+ plus GTP-gamma-S. These results indicate that histamine can be released from mast cells only when the concentration of VVC is high enough to cause the lysis of cells. At low concentrations, VVC does not induce the release of stored histamine from damaged cells. The intravenous injection of 80 HU purified VVC to rats, which can produce the calculated blood concentration of about 3 HU/ml, caused a marked increase in pulmonary vascular permeability, hemoconcentration and death. However, no increase in blood histamine level was detected. This level of VVC in rat blood was enough to cause severe hemoconcentration and lethality but might not be enough to cause cytolysis of the mast cells and resulting histamine release.
Collapse
Affiliation(s)
- J S Kim
- Department of Biochemistry, Chonbuk National University Medical School and Institutes for Medical Sciences, Chonju, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|