Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 13(6): e0273322, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36286551

RESUMO

Capsular polysaccharides are common virulence factors of extracellular, but not intracellular bacterial pathogens, due to the antiphagocytic properties of these surface structures. It is therefore paradoxical that Salmonella enterica subspecies enterica serovar Typhi, an intracellular pathogen, synthesizes a virulence-associated (Vi) capsule, which exhibits antiphagocytic properties. Here, we show that the Vi capsular polysaccharide has different functions when S. Typhi interacts with distinct subsets of host phagocytes. The Vi capsular polysaccharide allowed S. Typhi to selectively evade phagocytosis by human neutrophils while promoting human macrophage phagocytosis. A screen of C-type lectin receptors identified human DC-SIGN as the receptor involved in macrophage binding and phagocytosis of capsulated S. Typhi. Consistent with the anti-inflammatory activity of DC-SIGN, purified Vi capsular polysaccharide reduced inflammatory responses in macrophages. These data suggest that binding of the human C-type lectin receptor DC-SIGN by the Vi capsular polysaccharide contributes to the pathogenesis of typhoid fever. IMPORTANCE Salmonella enterica subspecies enterica serovar Typhi is the causative agent of typhoid fever. The recent emergence of S. Typhi strains which are resistant to antibiotic therapy highlights the importance of vaccination in managing typhoid fever. The virulence-associated (Vi) capsular polysaccharide is an effective vaccine against typhoid fever, but the role the capsule plays during pathogenesis remains incompletely understood. Here, we identify the human C-type lectin receptor DC-SIGN as the receptor for the Vi capsular polysaccharide. Binding of capsulated S. Typhi to DC-SIGN resulted in phagocytosis of the pathogen by macrophages and induction of an anti-inflammatory cytokine response. Thus, the interaction of the Vi capsular polysaccharide with human DC-SIGN contributes to the pathogenesis of typhoid fever and should be further investigated in the context of vaccine development.


Assuntos
Salmonella typhi , Febre Tifoide , Humanos , Febre Tifoide/microbiologia , Polissacarídeos Bacterianos/metabolismo , Lectinas Tipo C/metabolismo , Fagocitose , Macrófagos/metabolismo
2.
Cell Host Microbe ; 30(6): 836-847.e6, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35568027

RESUMO

Changes in the microbiota composition are associated with many human diseases, but factors that govern strain abundance remain poorly defined. We show that a commensal Escherichia coli strain and a pathogenic Salmonella enterica serovar Typhimurium isolate both utilize nitrate for intestinal growth, but each accesses this resource in a distinct biogeographical niche. Commensal E. coli utilizes epithelial-derived nitrate, whereas nitrate in the niche occupied by S. Typhimurium is derived from phagocytic infiltrates. Surprisingly, avirulent S. Typhimurium was shown to be unable to utilize epithelial-derived nitrate because its chemotaxis receptors McpB and McpC exclude the pathogen from the niche occupied by E. coli. In contrast, E. coli invades the niche constructed by S. Typhimurium virulence factors and confers colonization resistance by competing for nitrate. Thus, nutrient niches are not defined solely by critical resources, but they can be further subdivided biogeographically within the host into distinct microhabitats, thereby generating new niche opportunities for distinct bacterial species.


Assuntos
Microbioma Gastrointestinal , Salmonella typhimurium , Escherichia coli , Humanos , Nitratos , Nutrientes
3.
Cell Host Microbe ; 30(2): 163-170.e6, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34951948

RESUMO

Intracellular pathogens commonly reside within macrophages to find shelter from humoral defenses, but host cell death can expose them to the extracellular milieu. We find intracellular pathogens solve this dilemma by using virulence factors to generate a complement-dependent find-me signal that initiates uptake by a new phagocyte through efferocytosis. During macrophage death, Salmonella uses a type III secretion system to perforate the membrane of the pathogen-containing vacuole (PCV), thereby triggering complement deposition on bacteria entrapped in pore-induced intracellular traps (PITs). In turn, complement activation signals neutrophil efferocytosis, a process that shelters intracellular bacteria from the respiratory burst. Similarly, Brucella employs its type IV secretion system to perforate the PCV membrane, which induces complement deposition on bacteria entrapped in PITs. Collectively, this work identifies virulence factor-induced perforation of the PCV as a strategy of intracellular pathogens to generate a find-me signal for efferocytosis.


Assuntos
Vacúolos , Fatores de Virulência , Fagocitose , Sistemas de Secreção Tipo III , Sistemas de Secreção Tipo IV/metabolismo , Vacúolos/metabolismo
4.
Microbiol Immunol ; 64(3): 167-181, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31850542

RESUMO

Vibrio parahaemolyticus is a leading cause of seafood-borne bacterial gastroenteritis in humans. Since its discovery in 1950, this bacterium has been isolated in widespread outbreaks and in sporadic cases of gastroenteritis worldwide. Although the exotoxin, thermostable direct hemolysin, had been the focus of extensive research on the pathogenicity of V. parahaemolyticus, the whole-genome sequencing of a clinical isolate, RIMD2210633 strain, was a breakthrough in this field. The possession of two sets of gene clusters for type III secretion systems (T3SS1 and T3SS2) was unveiled by that genome project. T3SS is a protein export apparatus that delivers bacterial proteins, called effectors, directly into the host's cytosol, to disrupt host cell function. The subsequent studies have established that T3SS2, which is encoded in an 80 kb pathogenicity island called V. parahaemolyticus pathogenicity island (Vp-PAI), is closely related to enteropathogenicity. Recent functional analyses of Vp-PAI-encoded genes revealed the sophisticated mechanisms in V. parahaemolyticus for sensing the intestinal environment and host cell contact, and a dozen T3SS2-exported proteins encoded in Vp-PAI. In this review, we summarize recent advances in V. parahaemolyticus research regarding the control of the expression of Vp-PAI-encoded genes, structural components and the secretory regulation of T3SS2, and the biological activities of T3SS2-exported effectors. Thus, Vp-PAI-encoded T3SS2 becomes an important key in the postgenomic era to shed light on the enteropathogenic mechanism of V. parahaemolyticus.


Assuntos
Ilhas Genômicas/genética , Sistemas de Secreção Tipo III , Vibrioses/microbiologia , Vibrio parahaemolyticus , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Interações entre Hospedeiro e Microrganismos , Humanos , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/metabolismo , Vibrio parahaemolyticus/patogenicidade
5.
Nat Microbiol ; 4(5): 781-788, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30778145

RESUMO

Many Gram-negative pathogens utilize dedicated secretion systems to export virulence factors such as exotoxins and effectors1-4. Several exotoxins are synthesized as precursors containing amino-terminal Sec signal peptides and are exported through the inner-membrane-bound Sec machinery to the periplasm, followed by secretion across the outer membrane to the exterior using a type II secretion system (T2SS)3,5. Here, we report that thermostable direct haemolysin (TDH), an exotoxin of the food-borne pathogen Vibrio parahaemolyticus, can be exported through the type III secretion system (T3SS), which engages in one-step secretion of effectors4, despite possessing a Sec signal peptide and being mainly secreted via the T2SS. Although the precursor of TDH is targeted to the Sec pathway, a fraction of mature TDH was observed to re-enter the bacterial cytoplasm. The N terminus of mature TDH comprises a T3SS signal sequence, allowing it to be loaded into the T3SS. We also show that T3SS-delivered TDH as an effector contributes to intestinal fluid accumulation in a rabbit diarrhoeal model of V. parahaemolyticus infection. Thus, our results show that an unconventional export mechanism for a bacterial toxin via the T3SS in tandem with the Sec machinery facilitates the virulence trait of V. parahaemolyticus.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Hemolisinas/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Vibrioses/microbiologia , Vibrio parahaemolyticus/metabolismo , Animais , Proteínas de Bactérias/genética , Toxinas Bacterianas/metabolismo , Transporte Biológico , Feminino , Humanos , Camundongos Endogâmicos C3H , Coelhos , Sistemas de Secreção Tipo III/genética , Vibrio parahaemolyticus/genética
6.
mBio ; 9(4)2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-30042203

RESUMO

Many Gram-negative bacterial symbionts and pathogens employ a type III secretion system (T3SS) to live in contact with eukaryotic cells. Because T3SSs inject bacterial proteins (effectors) directly into host cells, the switching of secretory substrates between translocators and effectors in response to host cell attachment is a crucial step for the effective delivery of effectors. Here, we show that the protein secretion switch of Vibrio parahaemolyticus T3SS2, which is a main contributor to the enteropathogenicity of a food poisoning bacterium, is regulated by two gatekeeper proteins, VgpA and VgpB. In the absence of these gatekeepers, effector secretion was activated, but translocator secretion was abolished, causing the loss of virulence. We found that the K+ concentration, which is high inside the host cell but low outside, is a key factor for VgpA- and VgpB-mediated secretion switching. Exposure of wild-type bacteria to K+ ions provoked both gatekeeper and effector secretions but reduced the level of secretion of translocators. The secretion protein profile of wild-type bacteria cultured with 0.1 M KCl was similar to that of gatekeeper mutants. Furthermore, depletion of K+ ions in host cells diminished the efficiency of T3SS2 effector translocation. Thus, T3SS2 senses the high intracellular concentration of K+ of the host cell so that T3SS2 effectors can be effectively injected.IMPORTANCE The pathogenesis of many Gram-negative bacterial pathogens arises from a type III secretion system (T3SS), whereby bacterial proteins (effectors) are directly injected into host cells. The injected effectors then modify host cell functions. For effective delivery of effector proteins, bacteria need to both recognize host cell attachment and switch the type of secreted proteins. Here, we identified gatekeeper proteins that play important roles in a T3SS2 secretion switch of Vibrio parahaemolyticus, a causative agent of food-borne gastroenteritis. We also found that K+, which is present in high concentrations inside the host cell but in low concentrations outside, is a key factor for the secretion switch. Thus, V. parahaemolyticus senses the high intracellular K+ concentration, triggering the effective injection of effectors.


Assuntos
Proteínas de Bactérias/genética , Potássio/metabolismo , Sistemas de Secreção Tipo III/genética , Vibrio parahaemolyticus/genética , Proteínas de Bactérias/metabolismo , Citoplasma/química , Regulação Bacteriana da Expressão Gênica , Potássio/farmacologia , Cloreto de Potássio/metabolismo , Cloreto de Potássio/farmacologia , Transporte Proteico , Sistemas de Secreção Tipo III/metabolismo , Vibrio parahaemolyticus/efeitos dos fármacos , Vibrio parahaemolyticus/metabolismo
7.
FEMS Microbiol Rev ; 42(4): 527-541, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29790924

RESUMO

Typhoid and paratyphoid fever are severe systemic infections caused by human-adapted typhoidal Salmonella serovars that are indistinguishable in their clinical presentation, but differ from human gastroenteritis caused by zoonotic non-typhoidal Salmonella serovars. Typhoidal Salmonella serovars evolved from ancestral gastrointestinal pathogens through genetic changes that supported a change in pathogen ecology. Typhoidal Salmonella serovars share virulence properties that were acquired through convergent evolution and therefore this group is not defined by the presence of shared virulence genes that are absent from non-typhoidal Salmonella serovars. One feature distinguishing typhoidal Salmonella serovars from gastrointestinal pathogens is their ability to avert the respiratory burst of neutrophils. Furthermore, typhoidal Salmonella serovars possess several mechanisms to moderate intestinal inflammation, which are absent from non-typhoidal Salmonella serovars. Collectively, these shared virulence mechanisms enable typhoidal Salmonella serovars to breach an intact mucosal barrier and reach the gall bladder, a new ecological niche that is important because chronic gall bladder carriage promotes disease transmission. Thus, the morbidity and mortality resulting from the severe systemic infection that enables typhoidal Salmonella serovars to reach the gall bladder is coupled to their capacity for infectious transmission, which is the principal driving force of natural selection directing the emergence of this pathovar.


Assuntos
Evolução Biológica , Interações Hospedeiro-Patógeno/fisiologia , Salmonella/fisiologia , Animais , Humanos , Salmonella/patogenicidade , Sorogrupo , Febre Tifoide/microbiologia
8.
Cell Rep ; 22(7): 1787-1797, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29444431

RESUMO

Typhoid fever caused by Salmonella enterica serovar (S.) Typhi differs in its clinical presentation from gastroenteritis caused by S. Typhimurium and other non-typhoidal Salmonella serovars. The different clinical presentations are attributed in part to the virulence-associated capsular polysaccharide (Vi antigen) of S. Typhi, which prevents phagocytes from triggering a respiratory burst by preventing antibody-mediated complement activation. Paradoxically, the Vi antigen is absent from S. Paratyphi A, which causes a disease that is indistinguishable from typhoid fever. Here, we show that evasion of the phagocyte respiratory burst by S. Paratyphi A required very long O antigen chains containing the O2 antigen to inhibit antibody binding. We conclude that the ability to avoid the phagocyte respiratory burst is a property distinguishing typhoidal from non-typhoidal Salmonella serovars that was acquired by S. Typhi and S. Paratyphi A independently through convergent evolution.


Assuntos
Evolução Biológica , Fagócitos/microbiologia , Explosão Respiratória , Salmonella typhi/fisiologia , Sorogrupo , Febre Tifoide/microbiologia , Febre Tifoide/patologia , Adulto , Animais , Anticorpos/metabolismo , Antígenos de Bactérias/metabolismo , Ativação do Complemento , Células HL-60 , Humanos , Camundongos , Modelos Biológicos , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
9.
Nihon Saikingaku Zasshi ; 71(4): 199-208, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-27980291

RESUMO

To understand how bacterial pathogens cause diseases is the most important step in order to prevent the infection and develop an effective treatment. However, the past proceeding studies make us aware of quite-complicated interactions between the host and pathogenic bacteria. Vibrio parahaemolyticus, a food-born pathogen that is a subject of our study, causes inflammatory diarrhea in human upon ingestion of contaminated raw or undercooked seafood. Many virulence factors has been proposed since its discovery in Osaka around 70 years ago, while our research group has revealed that one of these virulence factors, type 3 secretion system 2 (T3SS2), is necessary for diarrhea induced by this bacterium. In addition, we recently found two novel T3SS2 effectors (VopO and VopV) that manipulate the actin cytoskeleton in infected host cells. In this article, I would like to show our findings with regard to biological activities of the effectors and their contributions to the T3SS2-induced enterotoxicity.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Diarreia/microbiologia , Sistemas de Secreção Tipo II/fisiologia , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/patogenicidade , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/fisiologia , Virulência/genética
10.
Sci Rep ; 5: 10870, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26039684

RESUMO

A novel bacterial type III secretion effector, VopV, from the enteric pathogen Vibrio parahaemolyticus has been identified as a key factor in pathogenicity due to its interaction with cytoskeletal actin. One of the repeat units in the long repetitive region of VopV, named VopV(rep1), functions as an actin-binding module. Despite its importance in pathogenesis, the manner in which the effector binds to actin and the subsequent effects on actin dynamics remain unclear. Here, we report the molecular basis of the VopV(rep1)/actin interaction. VopV(rep1) exists as an unstructured protein in solution but potently and specifically binds filamentous actin (F-actin) and not globular actin (G-actin). The F-actin/VopV(rep1) complex was directly visualized at 9.6-Å resolution using electron cryomicroscopy (cryoEM) and helical image reconstitution. The density map revealed the binding site of VopV(rep1) at the interface between two actin strands, which is close to the binding site of the bicyclic heptapeptide toxin phalloidin. Consistent with this observation, VopV(rep1) alone prevented the depolymerization of F-actin. Overall, VopVr(ep1) demonstrated unique characteristics in comparison to known actin-binding proteins, but was relatively similar to phalloidin. The phalloidin-like behavior, targeting the interstrand region of actin filaments to stabilize the filament structure, likely contributes to the pathogenicity of V. parahaemolyticus.


Assuntos
Citoesqueleto de Actina/metabolismo , Cólera/microbiologia , Citoesqueleto/metabolismo , Homeostase , Sistemas de Secreção Tipo III/metabolismo , Vibrio/fisiologia , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Proteínas de Bactérias/metabolismo , Citoesqueleto/química , Citoesqueleto/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Estabilidade Proteica
11.
PLoS Pathog ; 11(3): e1004694, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25738744

RESUMO

Vibrio parahaemolyticus is an important pathogen that causes food-borne gastroenteritis in humans. The type III secretion system encoded on chromosome 2 (T3SS2) plays a critical role in the enterotoxic activity of V. parahaemolyticus. Previous studies have demonstrated that T3SS2 induces actin stress fibers in various epithelial cell lines during infection. This stress fiber formation is strongly related to pathogenicity, but the mechanisms that underlie T3SS2-dependent actin stress fiber formation and the main effector have not been elucidated. In this study, we identified VopO as a critical T3SS2 effector protein that activates the RhoA-ROCK pathway, which is an essential pathway for the induction of the T3SS2-dependent stress fiber formation. We also determined that GEF-H1, a RhoA guanine nucleotide exchange factor (GEF), directly binds VopO and is necessary for T3SS2-dependent stress fiber formation. The GEF-H1-binding activity of VopO via an alpha helix region correlated well with its stress fiber-inducing capacity. Furthermore, we showed that VopO is involved in the T3SS2-dependent disruption of the epithelial barrier. Thus, VopO hijacks the RhoA-ROCK pathway in a different manner compared with previously reported bacterial toxins and effectors that modulate the Rho GTPase signaling pathway.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais/fisiologia , Vibrio parahaemolyticus/metabolismo , Actinas/metabolismo , Humanos , Microtúbulos/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
12.
Cell Microbiol ; 17(2): 183-90, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25495647

RESUMO

Vibrio parahaemolyticus is a leading causative agent of seafood-borne gastroenteritis worldwide. Most clinical isolates from patients with diarrhoea possess two sets of genes for the type III secretion system (T3SS) on each chromosome (T3SS1 and T3SS2). T3SS is a protein secretion system that delivers effector proteins directly into eukaryotic cells. The injected effectors modify the normal cell functions by altering or disrupting the normal cell signalling pathways. Of the two sets of T3SS genes present in V. parahaemolyticus, T3SS2 is essential for enterotoxicity in several animal models. Recent studies have elucidated the biological activities of several T3SS2 effectors and their roles in virulence. This review focuses on the regulation of T3SS2 gene expression and T3SS2 effectors that specifically target the actin cytoskeleton.


Assuntos
Citoesqueleto de Actina/metabolismo , Sistemas de Secreção Bacterianos , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Vibrio parahaemolyticus/fisiologia , Animais , Diarreia/microbiologia , Humanos , Vibrioses/microbiologia , Vibrio parahaemolyticus/metabolismo
13.
Infect Immun ; 82(6): 2390-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24664508

RESUMO

Clostridium perfringens is a causative agent of food-borne gastroenteritis for which C. perfringens enterotoxin (CPE) has been considered an essential factor. Recently, we experienced two outbreaks of food-borne gastroenteritis in which non-CPE producers of C. perfringens were strongly suspected to be the cause. Here, we report a novel enterotoxin produced by C. perfringens isolates, BEC (binary enterotoxin of C. perfringens). Culture supernatants of the C. perfringens strains showed fluid-accumulating activity in rabbit ileal loop and suckling mouse assays. Purification of the enterotoxic substance in the supernatants and high-throughput sequencing of genomic DNA of the strains revealed BEC, composed of BECa and BECb. BECa and BECb displayed limited amino acid sequence similarity to other binary toxin family members, such as the C. perfringens iota toxin. The becAB genes were located on 54.5-kb pCP13-like plasmids. Recombinant BECb (rBECb) alone had fluid-accumulating activity in the suckling mouse assay. Although rBECa alone did not show enterotoxic activity, rBECa enhanced the enterotoxicity of rBECb when simultaneously administered in suckling mice. The entertoxicity of the mutant in which the becB gene was disrupted was dramatically decreased compared to that of the parental strain. rBECa showed an ADP-ribosylating activity on purified actin. Although we have not directly evaluated whether BECb delivers BECa into cells, rounding of Vero cells occurred only when cells were treated with both rBECa and rBECb. These results suggest that BEC is a novel enterotoxin of C. perfringens distinct from CPE, and that BEC-producing C. perfringens strains can be causative agents of acute gastroenteritis in humans. Additionally, the presence of becAB on nearly identical plasmids in distinct lineages of C. perfringens isolates suggests the involvement of horizontal gene transfer in the acquisition of the toxin genes.


Assuntos
Clostridium perfringens/metabolismo , Enterotoxinas/metabolismo , Gastroenterite/microbiologia , ADP Ribose Transferases/genética , Doença Aguda , Análise de Variância , Animais , Modelos Animais de Doenças , Surtos de Doenças , Enterotoxinas/genética , Humanos , Camundongos , Peso Molecular , Coelhos , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA
14.
Cell Microbiol ; 16(6): 938-47, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24345190

RESUMO

Vibrio parahaemolyticus is a Gram-negative marine bacterium that causes acute gastroenteritis in humans. The virulence of V. parahaemolyticus is dependent upon a type III secretion system (T3SS2). One effector for T3SS2, VopC, is a homologue of the catalytic domain of cytotoxic necrotizing factor (CNF), and was recently reported to be a Rho family GTPase activator and to be linked to internalization of V. parahaemolyticus by non-phagocytic cultured cells. Here, we provide direct evidence that VopC deamidates Rac1 and CDC42, but not RhoA, in vivo. Our results alsosuggest that VopC, through its activation of Rac1, contributes to formation of actin stress fibres in infected cells. Invasion of host cells, which occurs at a low frequency, does not seem linked to Rac1 activation, but instead appears to require CDC42. Finally, using an infant rabbit model of V. parahaemolyticus infection, we show that the virulence of V. parahaemolyticus is not dependent upon VopC-mediated invasion. Genetic inactivation of VopC did not impair intestinal colonization nor reduce signs of disease, including fluid accumulation, diarrhoea and tissue destruction. Thus, although VopC can promote host cell invasion, such internalization is not a critical step of the disease process, consistent with the traditional view of V. parahaemolyticus as an extracellular pathogen.


Assuntos
Proteínas de Bactérias/metabolismo , Endocitose , Interações Hospedeiro-Patógeno , Vibrioses/microbiologia , Vibrio parahaemolyticus/fisiologia , Fatores de Virulência/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Células CACO-2 , Modelos Animais de Doenças , Humanos , Coelhos , Vibrioses/patologia , Vibrio parahaemolyticus/patogenicidade , Virulência , Proteínas rac1 de Ligação ao GTP/metabolismo
15.
Microbiologyopen ; 2(5): 725-42, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23894076

RESUMO

Resistance nodulation cell division (RND)-type efflux transporters play the main role in intrinsic resistance to various antimicrobial agents in many gram-negative bacteria. Here, we estimated 12 RND-type efflux transporter genes in Vibrio parahaemolyticus. Because VmeAB has already been characterized, we cloned the other 11 RND-type efflux transporter genes and characterized them in Escherichia coli KAM33 cells, a drug hypersusceptible strain. KAM33 expressing either VmeCD, VmeEF, or VmeYZ showed increased minimum inhibitory concentrations (MICs) for several antimicrobial agents. Additional four RND-type transporters were functional as efflux pumps only when co-expressed with VpoC, an outer membrane component in V. parahaemolyticus. Furthermore, VmeCD, VmeEF, and VmeYZ co-expressed with VpoC exhibited a broader substrate specificity and conferred higher resistance than that with TolC of E. coli. Deletion mutants of these transporter genes were constructed in V. parahaemolyticus. TM32 (ΔvmeAB and ΔvmeCD) had significantly decreased MICs for many antimicrobial agents and the number of viable cells after exposure to deoxycholate were markedly reduced. Strains in which 12 operons were all disrupted had very low MICs and much lower fluid accumulation in rabbit ileal loops. These results indicate that resistance nodulation cell division-type efflux transporters contribute not only to intrinsic resistance but also to exerting the virulence of V. parahaemolyticus.


Assuntos
Proteínas de Bactérias/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Filogenia , Transgenes , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/patogenicidade , Animais , Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/classificação , Proteínas de Bactérias/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Deleção de Genes , Expressão Gênica , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/classificação , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Óperon , Coelhos , Vibrioses/tratamento farmacológico , Vibrioses/microbiologia , Vibrio parahaemolyticus/efeitos dos fármacos , Vibrio parahaemolyticus/metabolismo , Virulência
16.
Microbiol Immunol ; 57(5): 334-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23668604

RESUMO

Twelve Vibrio cholerae isolates with genes for a type III secretion system (T3SS) were detected among 110 environmental and 14 clinical isolates. T3SS-related genes were distributed among the various serogroups and pulsed-field gel electrophoresis of NotI-digested genomes showed genetic diversity in these strains. However, the restriction fragment length polymorphism profiles of the T3SS-related genes had similar patterns. Additionally, naturally competent T3SS-negative V. cholerae incorporated the ca. 47 kb gene cluster of T3SS, which had been integrated into a site on the chromosome by recombination. Therefore, it is suggested that horizontal gene transfer of T3SS-related genes occurs among V. cholerae in natural ecosystems.


Assuntos
Transferência Genética Horizontal , Ilhas Genômicas , Proteínas de Membrana Transportadoras/genética , Vibrio cholerae/genética , Fatores de Virulência/genética , Cólera/microbiologia , Cromossomos Bacterianos , Eletroforese em Gel de Campo Pulsado , Microbiologia Ambiental , Variação Genética , Genótipo , Humanos , Tipagem Molecular , Família Multigênica , Polimorfismo de Fragmento de Restrição , Vibrio cholerae/classificação , Vibrio cholerae/isolamento & purificação , Vibrio cholerae/patogenicidade
17.
Infect Immun ; 80(8): 2940-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22585964

RESUMO

The pathogenesis of the diarrheal disease caused by Vibrio parahaemolyticus, a leading cause of seafood-associated enteritis worldwide, is dependent upon a type III secretion system, T3SS2. This apparatus enables the pathogen to inject bacterial proteins (effectors) into the cytosol of host cells and thereby modulate host processes. T3SS effector proteins transit into the host cell via a membrane pore (translocon) typically formed by 3 bacterial proteins. We have identified the third translocon protein for T3SS2: VopW, which was previously classified as an effector protein for a homologous T3SS in V. cholerae. VopW is a hydrophilic translocon protein; like other such proteins, it is not inserted into the host cell membrane but is required for insertion of the two hydrophobic translocators, VopB2 and VopD2, that constitute the membrane channel. VopW is not required for secretion of T3SS2 effectors into the bacterial culture medium; however, it is essential for transfer of these proteins into the host cell cytoplasm. Consequently, deletion of vopW abrogates the virulence of V. parahaemolyticus in several animal models of diarrheal disease. Unlike previously described hydrophilic translocators, VopW is itself translocated into the host cell cytoplasm, raising the possibility that it functions as both a translocator and an effector.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Vibrio parahaemolyticus/metabolismo , Animais , Proteínas de Bactérias/genética , Células CACO-2 , Proteínas de Transporte/genética , Deleção de Genes , Regulação Bacteriana da Expressão Gênica/fisiologia , Humanos , Íleo/microbiologia , Íleo/patologia , Família Multigênica , Transporte Proteico , Coelhos , Vibrioses/imunologia , Vibrioses/microbiologia , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/patogenicidade , Virulência
18.
Microbiology (Reading) ; 158(Pt 3): 746-758, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22194351

RESUMO

Among three haemolysins identified thus far in Escherichia coli, alpha-haemolysin (HlyA) is encoded on the pathogenicity islands of extraintestinal pathogenic strains, while enterohaemolysin (EhxA) is encoded on the virulence plasmids of enterohaemorrhagic E. coli (EHEC) strains. In contrast, the gene for haemolysin E (HlyE) is located on the E. coli chromosome backbone and is therefore widely distributed among E. coli strains. However, because hlyE gene expression is repressed by the H-NS protein and because the gene has been disrupted in many strains, its haemolytic activity cannot be detected in wild-type strains by routine screening on blood agar plates. In this study, we found that the HlyE-derived haemolytic activity of enteropathogenic E. coli (EPEC) O55 : H7 can be detected after anaerobic cultivation on a washed blood agar plate (EHX plate) that is used to detect the production of EhxA. We also found that the haemolytic activity of EHEC O157 : H7 observed on EHX plates under aerobic and anaerobic growth conditions is derived from EhxA and HlyE, respectively; this differential expression of the two haemolysins occurs at the transcriptional level. Our analysis of 60 E. coli strains of various pathotypes and phylogenies for their repertoires of haemolysin genes, haemolytic phenotypes and hlyE gene sequences revealed that HlyE activity can generally be detected on EHX plates under anaerobic growth conditions if the gene is intact. Furthermore, our results indicate that hlyE gene inactivation occurred in three of the five E. coli lineages (phylogroups A, B1 and B2), which demonstrates phylogroup-specific gene disruption patterns.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Proteínas Hemolisinas/metabolismo , Hemólise , Aerobiose , Anaerobiose , Meios de Cultura/química , DNA Bacteriano/química , DNA Bacteriano/genética , Proteínas de Escherichia coli/genética , Proteínas Hemolisinas/genética , Dados de Sequência Molecular , Análise de Sequência de DNA
19.
Cell Host Microbe ; 10(4): 401-9, 2011 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-22018240

RESUMO

Vibrio parahaemolyticus, a Gram-negative halophilic bacterium that causes acute gastroenteritis in humans, is characterized by two type III secretion systems (T3SS), namely T3SS1 and T3SS2. T3SS2 is indispensable for enterotoxicity but the effector(s) involved are unknown. Here, we identify VopV as a critical effector that is required to mediate V. parahaemolyticus T3SS2-dependent enterotoxicity. VopV was found to possess multiple F-actin-binding domains and the enterotoxicity caused by VopV correlated with its F-actin-binding activity. Furthermore, a T3SS2-related secretion system and a vopV homologous gene were also involved in the enterotoxicity of a non-O1/non-O139 V. cholerae strain. These results indicate that the F-actin-targeting effector VopV is involved in enterotoxic activity of T3SS2-possessing bacterial pathogens.


Assuntos
Actinas/metabolismo , Proteínas de Bactérias/metabolismo , Vibrio parahaemolyticus/patogenicidade , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Sítios de Ligação , Histocitoquímica , Intestinos/microbiologia , Intestinos/patologia , Microscopia , Estrutura Terciária de Proteína , Transporte Proteico , Coelhos , Vibrio cholerae/patogenicidade , Vibrio parahaemolyticus/genética , Fatores de Virulência/genética
20.
PLoS One ; 5(10): e13365, 2010 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-20967223

RESUMO

Vibrio parahaemolyticus, a bacterial pathogen, causes human gastroenteritis. A type III secretion system (T3SS2) encoded in pathogenicity island (Vp-PAI) is the main contributor to enterotoxicity and expression of Vp-PAI encoded genes is regulated by two transcriptional regulators, VtrA and VtrB. However, a host-derived inducer for the Vp-PAI genes has not been identified. Here, we demonstrate that bile induces production of T3SS2-related proteins under osmotic conditions equivalent to those in the intestinal lumen. We also show that bile induces vtrA-mediated vtrB transcription. Transcriptome analysis of bile-responsive genes revealed that bile strongly induces expression of Vp-PAI genes in a vtrA-dependent manner. The inducing activity of bile was diminished by treatment with bile acid sequestrant cholestyramine. Finally, we demonstrate an in vivo protective effect of cholestyramine on enterotoxicity and show that similar protection is observed in infection with a different type of V. parahaemolyticus or with non-O1/non-O139 V. cholerae strains of vibrios carrying the same kind of T3SS. In summary, these results provide an insight into how bacteria, through the ingenious action of Vp-PAI genes, can take advantage of an otherwise hostile host environment. The results also reveal a new therapeutic potential for widely used bile acid sequestrants in enteric bacterial infections.


Assuntos
Ácidos e Sais Biliares/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Vibrio parahaemolyticus/genética , Virulência/genética , Western Blotting , Eletroforese em Gel de Poliacrilamida , Genes Bacterianos , Transcrição Gênica , Vibrio parahaemolyticus/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...