Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Med Invest ; 71(1.2): 102-112, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38735705

RESUMO

Vibrio vulnificus (V. vulnificus) is a halophilic gram-negative bacterium that inhabits coastal warm water and induce severe diseases such as primary septicemia. To investigate the mechanisms of rapid bacterial translocation on intestinal infection, we focused on outer membrane vesicles (OMVs), which are extracellular vesicles produced by Gram-negative bacteria and deliver virulence factors. However, there are very few studies on the pathogenicity or contents of V. vulnificus OMVs (Vv-OMVs). In this study, we investigated the effects of Vv-OMVs on host cells. Epithelial cells INT407 were stimulated with purified OMVs and morphological alterations and levels of lactate dehydrogenase (LDH) release were observed. In cells treated with OMVs, cell detachment without LDH release was observed, which exhibited different characteristics from cytotoxic cell detachment observed in V. vulnificus infection. Interestingly, OMVs from a Vibrio Vulnificus Hemolysin (VVH) and Multifunctional-autoprocessing repeats-in -toxin (MARTX) double-deletion mutant strain also caused cell detachment without LDH release. Our results suggested that the proteolytic function of a serine protease contained in Vv-OMVs may contribute to pathogenicity of V. vulnificus by assisting bacterial translocation. This study reveals a new pathogenic mechanism during V. vulnificus infections. J. Med. Invest. 71 : 102-112, February, 2024.


Assuntos
Vesículas Extracelulares , Vibrio vulnificus , Vibrio vulnificus/patogenicidade , Vibrio vulnificus/metabolismo , Humanos , Vesículas Extracelulares/metabolismo , Proteínas Hemolisinas/metabolismo , L-Lactato Desidrogenase/metabolismo , Membrana Externa Bacteriana/metabolismo , Células Epiteliais/microbiologia
2.
J Biol Chem ; 296: 100777, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33992647

RESUMO

Opportunistic bacteria strategically dampen their virulence to allow them to survive and propagate in hosts. However, the molecular mechanisms underlying virulence control are not clearly understood. Here, we found that the opportunistic pathogen Vibrio vulnificus biotype 3, which caused an outbreak of severe wound and intestinal infections associated with farmed tilapia, secretes significantly less virulent multifunctional autoprocessing repeats-in-toxin (MARTX) toxin, which is the most critical virulence factor in other clinical Vibrio strains. The biotype 3 MARTX toxin contains a cysteine protease domain (CPD) evolutionarily retaining a unique autocleavage site and a distinct ß-flap region. CPD autoproteolytic activity is attenuated following its autocleavage because of the ß-flap region. This ß-flap blocks the active site, disabling further autoproteolytic processing and release of the modularly structured effector domains within the toxin. Expression of this altered CPD consequently results in attenuated release of effectors by the toxin and significantly reduces the virulence of V. vulnificus biotype 3 in cells and in mice. Bioinformatic analysis revealed that this virulence mechanism is shared in all biotype 3 strains. Thus, these data provide new insights into the mechanisms by which opportunistic bacteria persist in an environmental reservoir, prolonging the potential to cause outbreaks.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Vibrioses/metabolismo , Vibrio vulnificus/patogenicidade , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Modelos Moleculares , Vibrio vulnificus/fisiologia , Fatores de Virulência/química
3.
mSphere ; 5(4)2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32817457

RESUMO

To understand toxin-stimulated host-pathogen interactions, we performed dual-transcriptome sequencing experiments using human epithelial (HT-29) and differentiated THP-1 (dTHP-1) immune cells infected with the sepsis-causing pathogen Vibrio vulnificus (either the wild-type [WT] pathogen or a multifunctional-autoprocessing repeats-in-toxin [MARTX] toxin-deficient strain). Gene set enrichment analyses revealed MARTX toxin-dependent responses, including negative regulation of extracellular related kinase 1 (ERK1) and ERK2 (ERK1/2) signaling and cell cycle regulation in HT-29 and dTHP-1 cells, respectively. Further analysis of the expression of immune-related genes suggested that the MARTX toxin dampens immune responses in gut epithelial cells but accelerates inflammation and nuclear factor κB (NF-κB) signaling in immune cells. With respect to the pathogen, siderophore biosynthesis genes were significantly more highly expressed in WT V. vulnificus than in the MARTX toxin-deficient mutant upon infection of dTHP-1 cells. Consistent with these results, iron homeostasis genes that limit iron levels for invading pathogens were overexpressed in WT V. vulnificus-infected dTHP-1 cells. Taken together, these results suggest that MARTX toxin regulates host inflammatory responses during V. vulnificus infection while also countering host defense mechanisms such as iron limitation.IMPORTANCEV. vulnificus is an opportunistic human pathogen that can cause life-threatening sepsis in immunocompromised patients via seafood poisoning or wound infection. Among the toxic substances produced by this pathogen, the MARTX toxin greatly contributes to disease progression by promoting the dysfunction and death of host cells, which allows the bacteria to disseminate and colonize the host. In response to this, host cells mount a counterattack against the invaders by upregulating various defense genes. In this study, the gene expression profiles of both host cells and V. vulnificus were analyzed by RNA sequencing to gain a comprehensive understanding of host-pathogen interactions. Our results suggest that V. vulnificus uses the MARTX toxin to subvert host cell immune responses as well as to oppose host counterattacks such as iron limitation.


Assuntos
Toxinas Bacterianas/metabolismo , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno/genética , Vibrio vulnificus/genética , Toxinas Bacterianas/genética , Perfilação da Expressão Gênica , Células HT29 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Ferro/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Sideróforos/genética , Células THP-1 , Vibrio vulnificus/patogenicidade , Fatores de Virulência/genética
4.
Proc Natl Acad Sci U S A ; 116(36): 18031-18040, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31427506

RESUMO

Upon invading target cells, multifunctional autoprocessing repeats-in-toxin (MARTX) toxins secreted by bacterial pathogens release their disease-related modularly structured effector domains. However, it is unclear how a diverse repertoire of effector domains within these toxins are processed and activated. Here, we report that Makes caterpillars floppy-like effector (MCF)-containing MARTX toxins require ubiquitous ADP-ribosylation factor (ARF) proteins for processing and activation of intermediate effector modules, which localize in different subcellular compartments following limited processing of holo effector modules by the internal cysteine protease. Effector domains structured tandemly with MCF in intermediate modules become disengaged and fully activated by MCF, which aggressively interacts with ARF proteins present at the same location as intermediate modules and is converted allosterically into a catalytically competent protease. MCF-mediated effector processing leads ultimately to severe virulence in mice via an MCF-mediated ARF switching mechanism across subcellular compartments. This work provides insight into how bacteria take advantage of host systems to induce systemic pathogenicity.


Assuntos
Fatores de Ribosilação do ADP , ADP-Ribosilação , Toxinas Bacterianas , Vibrio vulnificus , Fatores de Ribosilação do ADP/química , Fatores de Ribosilação do ADP/metabolismo , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Domínios Proteicos , Vibrio vulnificus/genética , Vibrio vulnificus/metabolismo , Vibrio vulnificus/patogenicidade
5.
Int J Mol Med ; 41(1): 531-540, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29115398

RESUMO

Vibrio vulnificus (V. vulnificus) is a gram-negative bacterium, which causes life-threatening septicemia and gastroenteritis through the consumption of contaminated seafood or wound infection. In addition, V. vulnificus infection is known to stimulate the production of several pro-inflammatory cytokines, which are associated with inflammatory responses mediated predominantly by dendritic cells (DCs), functioning as antigen-presenting cells. The present study aimed to investigate whether V. vulnificus infection induced the maturation and activation of murine DCs, which have the ability to polarize T helper (Th) cells into Th17 cells. Dysregulated Th17 cell responses are known to cause tissue damage, promoting the penetration of pathogens; however, Th17 cells are also involved in host defense against infection. Infection with V. vulnificus significantly increased the expression of cell surface molecules, including CD40, CD80 and major histocompatibility complex class II, leading to the maturation and activation of DCs. In the present study, the analysis of the cytokine profiles of DCs upon infection with V. vulnificus revealed the preferential production of interleukin-1ß (IL-1ß) and IL-6, through which V. vulnificus-infected DCs induced the polarization of Th17 cells when naïve CD4+ T cells were co-incubated. The reduction of Th17 cell generation through the use of anti-IL-6 neutralizing antibodies indicated that the Th17-polarizing capacity of V. vulnificus was predominantly dependent on DC-derived IL-6. The in vivo administration of V. vulnificus-infected DCs consistently increased the Th17 cell population in the lymph nodes of mice. Finally, the oral administration of V. vulnificus in mice also increased Th17 cell responses in the lamina propria of the small intestine. These results collectively demonstrated that V. vulnificus induced inflammatory Th17 cell responses via DCs, which may be associated with the immunopathological effects caused by V. vulnificus infection.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/imunologia , Inflamação/imunologia , Células Th17/imunologia , Vibrioses/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Antígeno B7-1/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Polaridade Celular/imunologia , Humanos , Inflamação/genética , Inflamação/microbiologia , Inflamação/patologia , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Linfonodos/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Camundongos , Vibrioses/genética , Vibrioses/microbiologia , Vibrioses/patologia , Vibrio vulnificus/imunologia , Vibrio vulnificus/patogenicidade
6.
Artigo em Inglês | MEDLINE | ID: mdl-28848713

RESUMO

An inflammatory form of phagocyte death evoked by the Gram-negative bacterium Vibrio (V.) vulnificus (WT) is one of hallmarks to promote their colonization, but the virulence factor and infectious mechanism involved in this process remain largely unknown. Here, we identified extracellular metalloprotease VvpM as a new virulence factor and investigated the molecular mechanism of VvpM which acts during the regulation of the inflammatory form of macrophage death and bacterial colonization. Mutation of the vvpM gene appeared to play major role in the prevention of IL-1ß production due to V. vulnificus infection in macrophage. However, the recombinant protein (r) VvpM caused IL-1ß production coupled with necrotic cell death, which is highly susceptible to the knockdown of annexin A2 (ANXA2) located in both membrane lipid and non-lipid rafts. In lipid rafts, rVvpM recruited NOX enzymes coupled with ANXA2 to facilitate the production of ROS responsible for the epigenetic and transcriptional regulation of NF-κB in the IL-1ß promoter. rVvpM acting on non-lipid rafts increased LC3 puncta formation and autophagic flux, which are required for the mRNA expression of Atg5 involved in the autophagosome formation process. The autophagy activation caused by rVvpM induced NLRP3 inflammasome-dependent caspase-1 activation in the promoting of IL-1ß production. In mouse models of V. vulnificus infection, the VvpM mutant failed to elevate the level of pro-inflammatory responses closely related to IL-1ß production and prevented bacterial colonization. These findings delineate VvpM efficiently regulates two pathogenic pathways that stimulate NF-κB-dependent IL-1ß production and autophagy-mediated NLRP3 inflammasome via distinct spatial targeting by ANXA2.


Assuntos
Anexina A2/metabolismo , Apoptose , Interleucina-1beta/metabolismo , Macrófagos/fisiologia , Vibrioses/microbiologia , Vibrio vulnificus/patogenicidade , Fatores de Virulência/metabolismo , Animais , Anexina A2/genética , Células CACO-2 , Modelos Animais de Doenças , Humanos , Interleucina-1beta/genética , Microdomínios da Membrana/metabolismo , Metaloproteases/genética , Metaloproteases/metabolismo , Camundongos , NADPH Oxidases/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vibrioses/patologia , Fatores de Virulência/genética
7.
J Biomed Sci ; 24(1): 58, 2017 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-28822352

RESUMO

BACKGROUND: Vibrio vulnificus is a marine bacterial species that causes opportunistic infections manifested by serious skin lesions and fulminant septicemia in humans. We have previously shown that the multifunctional autoprocessing repeats in toxin (MARTXVv1) of a biotype 1 V. vulnificus strain promotes survival of this organism in the host by preventing it from engulfment by the phagocytes. The purpose of this study was to further explore how MARTXVv1 inhibits phagocytosis of this microorganism by the macrophage. METHODS: We compared between a wild-type V. vulnificus strain and its MARTXVv1-deficient mutant for a variety of phagocytosis-related responses, including morphological change and activation of signaling molecules, they induced in the macrophage. We also characterized a set of MARTXVv1 domain-deletion mutants to define the regions associated with antiphagocytosis activity. RESULTS: The RAW 264.7 cells and mouse peritoneal exudate macrophages underwent cell rounding accompanied by F-actin disorganization in the presence of MARTXVv1. In addition, phosphorylation of some F-actin rearrangement-associated signaling molecules, including Lyn, Fgr and Hck of the Src family kinases (SFKs), focal adhesion kinase (FAK), proline-rich tyrosine kinase 2 (Pyk2), phosphoinositide 3-kinase (PI3K) and Akt, but not p38, was decreased. By using specific inhibitors, we found that these kinases were all involved in the phagocytosis of MARTXVv1-deficient mutant in an order of SFKs-FAK/Pyk2-PI3K-Akt. Deletion of the effector domains in the central region of MARTXVv1 could lead to reduced cytotoxicity, depending on the region and size of deletion, but did not affect the antiphagocytosis activity and ability to cause rounding of macrophage. Reduced phosphorylation of Akt was closely associated with inhibition of phagocytosis by the wild-type strain and MARTXVv1 domain-deletion mutants, and expression of the constitutively active Akt, myr-Akt, enhanced the engulfment of these strains by macrophage. CONCLUSIONS: MARTXVv1 could inactivate the SFKs-FAK/Pyk2-PI3K-Akt signaling pathway in the macrophages. This might lead to impaired phagocytosis of the V. vulnificus-infected macrophage. The majority of the central region of MARTXVv1 is not associated with the antiphagocytosis activity.


Assuntos
Toxinas Bacterianas/imunologia , Fagocitose/imunologia , Vibrioses/microbiologia , Vibrio vulnificus/imunologia , Vibrio vulnificus/patogenicidade , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Citotoxinas/imunologia , Citotoxinas/metabolismo , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos BALB C , Vibrioses/patologia , Vibrio vulnificus/genética
8.
Mol Cells ; 40(4): 299-306, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28427249

RESUMO

The transcriptional activator AphB has been implicated in acid resistance and pathogenesis in the food borne pathogens Vibrio vulnificus and Vibrio cholerae. To date, the full-length AphB crystal structure of V. cholerae has been determined and characterized by a tetrameric assembly of AphB consisting of a DNA binding domain and a regulatory domain (RD). Although acidic pH and low oxygen tension might be involved in the activation of AphB, it remains unknown which ligand or stimulus activates AphB at the molecular level. In this study, we determine the crystal structure of the AphB RD from V. vulnificus under aerobic conditions without modification at the conserved cysteine residue of the RD, even in the presence of the oxidizing agent cumene hydroperoxide. A cysteine to serine amino acid residue mutant RD protein further confirmed that the cysteine residue is not involved in sensing oxidative stress in vitro. Interestingly, an unidentified small molecule was observed in the inter-subdomain cavity in the RD when the crystal was incubated with cumene hydroperoxide molecules, suggesting a new ligand-binding site. In addition, we confirmed the role of AphB in acid tolerance by observing an aphB-dependent increase in cadC transcript level when V. vulnificus was exposed to acidic pH. Our study contributes to the understanding of the AphB molecular mechanism in the process of recognizing the host environment.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Genes Reguladores , Transativadores/química , Transativadores/fisiologia , Vibrioses/microbiologia , Vibrio vulnificus/patogenicidade , Derivados de Benzeno/química , Sítios de Ligação , Cristalografia por Raios X , Cisteína/química , Cisteína/genética , Regulação Bacteriana da Expressão Gênica , Concentração de Íons de Hidrogênio , Ligantes , Modelos Moleculares , Estresse Oxidativo/fisiologia , Domínios e Motivos de Interação entre Proteínas , Análise de Sequência de Proteína , Serina/química , Serina/genética , Vibrio vulnificus/genética , Virulência/genética
9.
Int Wound J ; 14(5): 818-822, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28052529

RESUMO

Vibrio vulnificus can cause severe skin and soft tissue infection (SSTI). The pathogen is an opportunistic marine bacterium that is likely to infect patients with chronic liver disease, patients in an immunocompromised state, and those in end-stage renal disease. V. vulnificus gains entry through soft tissues by direct penetration of a wound by infected marine organisms, such as raw oysters, shellfish and other seafood, or by exposing a wound to contaminated water. Despite its ease of entry, V. vulnificus necrotising fasciitis with compartment syndrome has rarely been described. We report a case of an elderly patient with end-stage renal disease undergoing haemodialysis, who developed necrotising fasciitis following infection by V. vulnificus through a puncture injury while cleaning fish. A successful salvage and reconstruction surgery was performed using fenestrated-type artificial dermis followed by negative pressure wound therapy. This case presents a reasonable treatment option for threatening V. vulnificus necrotising fasciitis with compartment syndrome.


Assuntos
Fasciite Necrosante/etiologia , Fasciite Necrosante/cirurgia , Dedos/cirurgia , Tratamento de Ferimentos com Pressão Negativa , Pele Artificial , Vibrio vulnificus/patogenicidade , Infecção dos Ferimentos/cirurgia , Idoso de 80 Anos ou mais , Feminino , Dedos/microbiologia , Humanos , Falência Renal Crônica , Resultado do Tratamento
10.
PLoS Pathog ; 13(1): e1006119, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28060924

RESUMO

Vibrio vulnificus causes highly lethal bacterial infections in which the Multifunctional Autoprocessing Repeats-in-Toxins (MARTX) toxin product of the rtxA1 gene is a key virulence factor. MARTX toxins are secreted proteins up to 5208 amino acids in size. Conserved MARTX N- and C-terminal repeat regions work in concert to form pores in eukaryotic cell membranes, through which the toxin's central region of modular effector domains is translocated. Upon inositol hexakisphosphate-induced activation of the of the MARTX cysteine protease domain (CPD) in the eukaryotic cytosol, effector domains are released from the holotoxin by autoproteolytic activity. We previously reported that the native MARTX toxin effector domain repertoire is dispensable for epithelial cellular necrosis in vitro, but essential for cell rounding and apoptosis prior to necrotic cell death. Here we use an intragastric mouse model to demonstrate that the effector domain region is required for bacterial virulence during intragastric infection. The MARTX effector domain region is essential for bacterial dissemination from the intestine, but dissemination occurs in the absence of overt intestinal tissue pathology. We employ an in vitro model of V. vulnificus interaction with polarized colonic epithelial cells to show that the MARTX effector domain region induces rapid intestinal barrier dysfunction and increased paracellular permeability prior to onset of cell lysis. Together, these results negate the inherent assumption that observations of necrosis in vitro directly predict bacterial virulence, and indicate a paradigm shift in our conceptual understanding of MARTX toxin function during intestinal infection. Results implicate the MARTX effector domain region in mediating early bacterial dissemination from the intestine to distal organs-a key step in V. vulnificus foodborne pathogenesis-even before onset of overt intestinal pathology.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Junções Íntimas/patologia , Vibrioses/transmissão , Vibrio vulnificus/patogenicidade , Animais , Apoptose/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/ultraestrutura , Toxinas Bacterianas/genética , Membrana Celular/patologia , Modelos Animais de Doenças , Epitélio/microbiologia , Epitélio/patologia , Feminino , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/ultraestrutura , Camundongos , Camundongos Endogâmicos ICR , Ácido Fítico/farmacologia , Estrutura Terciária de Proteína , Vibrioses/microbiologia , Vibrio vulnificus/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
11.
J Biol Chem ; 291(27): 14213-14230, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27151217

RESUMO

Vibrio vulnificus is a marine bacterium that causes human infections resulting in high mortality. This pathogen harbors five quorum-regulatory RNAs (Qrr1-5) that affect the expression of pathogenicity genes by modulating the expression of the master regulator SmcR. The qrr genes are activated by phosphorylated LuxO to different degrees; qrr2 is strongly activated; qrr3 and qrr5 are moderately activated, and qrr1 and qrr4 are marginally activated and are the only two that do not respond to cell density-dependent regulation. Qrrs function redundantly to inhibit SmcR at low cell density and fully repress when all five are activated. In this study, we found that iron inhibits qrr expression in three distinct ways. First, the iron-ferric uptake regulator (Fur) complex directly binds to qrr promoter regions, inhibiting LuxO activation by competing with LuxO for cis-acting DNA elements. Second, qrr transcription is repressed by iron independently of Fur. Third, LuxO expression is repressed by iron independently of Fur. We also found that, under iron-limiting conditions, the five Qrrs functioned additively, not redundantly, to repress SmcR, suggesting that cells lacking iron enter a high cell density mode earlier and could thereby modulate expression of virulence factors sooner. This study suggests that iron and quorum sensing, along with their cognate regulatory circuits, are linked together in the coordinated expression of virulence factors.


Assuntos
Regulação Bacteriana da Expressão Gênica , Ferro/metabolismo , Percepção de Quorum , Vibrio vulnificus/patogenicidade , Sequência de Bases , Genes Bacterianos , Homologia de Sequência do Ácido Nucleico , Vibrio vulnificus/genética , Virulência
12.
Infect Immun ; 84(1): 254-65, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26527216

RESUMO

TonB systems actively transport iron-bound substrates across the outer membranes of Gram-negative bacteria. Vibrio vulnificus CMCP6, which causes fatal septicemia and necrotizing wound infections, possesses three active TonB systems. It is not known why V. vulnificus CMCP6 has maintained three TonB systems throughout its evolution. The TonB1 and TonB2 systems are relatively well characterized, while the pathophysiological function of the TonB3 system is still elusive. A reverse transcription-PCR (RT-PCR) study showed that the tonB1 and tonB2 genes are preferentially induced in vivo, whereas tonB3 is persistently transcribed, albeit at low expression levels, under both in vitro and in vivo conditions. The goal of the present study was to elucidate the raison d'être of these three TonB systems. In contrast to previous studies, we constructed in-frame single-, double-, and triple-deletion mutants of the entire structural genes in TonB loci, and the changes in various virulence-related phenotypes were evaluated. Surprisingly, only the tonB123 mutant exhibited a significant delay in killing eukaryotic cells, which was complemented in trans with any TonB operon. Very interestingly, we discovered that flagellum biogenesis was defective in the tonB123 mutant. The loss of flagellation contributed to severe defects in motility and adhesion of the mutant. Because of the difficulty of making contact with host cells, the mutant manifested defective RtxA1 toxin production, which resulted in impaired invasiveness, delayed cytotoxicity, and decreased lethality for mice. Taken together, these results indicate that a series of virulence defects in all three TonB systems of V. vulnificus CMCP6 coordinately complement each other for iron assimilation and full virulence expression by ensuring flagellar biogenesis.


Assuntos
Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Flagelos/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Proteínas de Membrana/genética , Vibrio vulnificus/patogenicidade , Animais , Proteínas de Bactérias/biossíntese , Toxinas Bacterianas/biossíntese , Transporte Biológico/genética , Linhagem Celular Tumoral , Feminino , Flagelos/genética , Células HeLa , Humanos , Ferro/metabolismo , Proteínas de Membrana/biossíntese , Camundongos , Cavidade Peritoneal/microbiologia , Ratos , Ratos Sprague-Dawley , Vibrioses/microbiologia , Vibrio vulnificus/genética , Vibrio vulnificus/crescimento & desenvolvimento
13.
Arq. Inst. Biol ; 83: e1252013, 2016.
Artigo em Português | LILACS, VETINDEX | ID: biblio-1006986

RESUMO

As bactérias do gênero Vibrio habitam ambiente tipicamente marinho e estuarino, sendo comumente isoladas de pescados. As principais espécies de Vibrio reportadas como agentes de infecções em humanos são V. vulnificus , V. parahaemolyticus , V. cholerae e V. mimicus . V. vulnificus é considerado o mais perigoso, podendo causar septicemia e levar à morte. V. parahaemolyticus é um patógeno importante nas regiões costeiras de clima temperado e tropical em todo o mundo e tem sido responsável por casos de gastroenterites associadas ao consumo de peixes, moluscos e crustáceos marinhos. V. cholerae causa surtos, epidemias e pandemias relacionados com ambientes estuarinos. V. mimicus pode causar episódios esporádicos de gastroenterite aguda e infecções de ouvido. A patogenicidade das bactérias está ligada à habilidade do micro-organismo em iniciar uma doença (incluindo entrada, colonização e multiplicação no corpo humano). Para que isso ocorra, os micro-organismos fazem uso de diversos fatores. O objetivo desta revisão foi sintetizar o conhecimento disponível na literatura sobre os fatores de patogenicidade de V. vulnificus , V. parahaemolyticus , V. cholerae e V. mimicus.(AU)


Bacteria of the genus Vibrio typically habitat marine and estuarine environment and are commonly isolated from fish. The main Vibrio species reported as agents of infections in humans are V. vulnificus , V. parahaemolyticus , V. cholerae and V. mimicus . V. vulnificus is considered the most dangerous, may cause sepsis and lead to death. V. parahaemolyticus is an important pathogen in coastal regions of temperate and tropical climates around the world and has been responsible for cases of gastroenteritis associated with consumption of fish, shellfish and marine crustaceans. V. cholerae causes outbreaks, epidemics and pandemics related to estuarine environments. V. mimicus can cause sporadic episodes of acute gastroenteritis and infections. The pathogenicity of the bacteria is linked to the ability of the micro-organism to initiate a disease (including entry, colonization and multiplication in the human body). For this to occur, the micro-organisms make use of several factors. The objective of this review is to summarize the knowledge available in the literature on the factors of pathogenicity of V. vulnificus , V. parahaemolyticus , V. cholerae and V. mimicus.(AU)


Assuntos
Bactérias , Vibrio parahaemolyticus/patogenicidade , Virulência , Vibrio vulnificus/patogenicidade , Vibrio mimicus/patogenicidade , Peixes
14.
Microbiol Spectr ; 3(3)2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26185080

RESUMO

Vibrio vulnificus biotype 2 is the etiological agent of warm-water vibriosis, a disease that affects eels and other teleosts, especially in fish farms. Biotype 2 is polyphyletic and probably emerged from aquatic bacteria by acquisition of a transferable virulence plasmid that encodes resistance to innate immunity of eels and other teleosts. Interestingly, biotype 2 comprises a zoonotic clonal complex designated as serovar E that has extended worldwide. One of the most interesting virulence factors produced by serovar E is RtxA13, a multifunctional protein that acts as a lethal factor for fish, an invasion factor for mice, and a survival factor outside the host. Two practically identical copies of rtxA13 are present in all biotype 2 strains regardless of the serovar, one in the virulence plasmid and the other in chromosome II. The plasmid also contains other genes involved in survival and growth in eel blood: vep07, a gene for an outer membrane (OM) lipoprotein involved in resistance to eel serum and vep20, a gene for an OM receptor specific for eel-transferrin and, probably, other related fish transferrins. All the three genes are highly conserved within biotype 2, which suggests that they are under a strong selective pressure. Interestingly, the three genes are related with transferable plasmids, which emphasizes the role of horizontal gene transfer in the evolution of V. vulnificus in nutrient-enriched aquatic environments, such as fish farms.


Assuntos
Toxinas Bacterianas/metabolismo , Enguias/microbiologia , Doenças dos Peixes/microbiologia , Vibrioses/microbiologia , Vibrio vulnificus/patogenicidade , Fatores de Virulência/metabolismo , Microbiologia da Água , Animais , Toxinas Bacterianas/genética , Doenças dos Peixes/epidemiologia , Doenças dos Peixes/transmissão , Transferência Genética Horizontal , Humanos , Imunidade Inata , Camundongos , Filogenia , Plasmídeos/genética , Receptores da Transferrina/metabolismo , Vibrioses/epidemiologia , Vibrioses/transmissão , Vibrio vulnificus/classificação , Vibrio vulnificus/genética , Fatores de Virulência/genética
15.
J Microbiol Biotechnol ; 25(2): 302-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25649984

RESUMO

VvpM, one of the extracellular metalloproteases produced by Vibrio vulnificus, induces apoptotic cell death via a pathway consisting of ERK activation, cytochrome c release, and activation of caspases-9 and -3. VvpM-treated cells also showed necrotic cell death as stained by propidium iodide (PI). The percentage of PI-stained cells was decreased by pretreatment with Necrostatin-1, indicating that VvpM-mediated cell death occurs through necroptosis. The appearance of autophagic vesicles and lipidated form of light-chain-3B in rVvpM-treated cells suggests an involvement of autophagy in this process. Therefore, the multifarious action of VvpM might be one of the factors responsible for V. vulnificus pathogenesis.


Assuntos
Apoptose , Autofagia , Morte Celular , Metaloproteases/metabolismo , Vibrio vulnificus/enzimologia , Vibrio vulnificus/patogenicidade , Caspase 3/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Citocromos c/metabolismo , Humanos , Imidazóis/farmacologia , Indóis/farmacologia , Necrose , Vibrio vulnificus/metabolismo
16.
Infect Immun ; 83(3): 1150-61, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25561711

RESUMO

Cyclo(Phe-Pro) (cFP) is a secondary metabolite produced by certain bacteria and fungi. Although recent studies highlight the role of cFP in cell-to-cell communication by bacteria, its role in the context of the host immune response is poorly understood. In this study, we investigated the role of cFP produced by the human pathogen Vibrio vulnificus in the modulation of innate immune responses toward the pathogen. cFP suppressed the production of proinflammatory cytokines, nitric oxide, and reactive oxygen species in a lipopolysaccharide (LPS)-stimulated monocyte/macrophage cell line and in bone marrow-derived macrophages. Specifically, cFP inhibited inhibitory κB (IκB) kinase (IKK) phosphorylation, IκBα degradation, and nuclear factor κB (NF-κB) translocation to the cell nucleus, indicating that cFP affects the NF-κB pathway. We searched for genes that are responsible for cFP production in V. vulnificus and identified VVMO6_03017 as a causative gene. A deletion of VVMO6_03017 diminished cFP production and decreased virulence in subcutaneously inoculated mice. In summary, cFP produced by V. vulnificus actively suppresses the innate immune responses of the host, thereby facilitating its survival and propagation in the host environment.


Assuntos
Dipeptídeos/farmacologia , Genes Bacterianos , Peptídeos Cíclicos/farmacologia , Pele/imunologia , Vibrioses/imunologia , Vibrio vulnificus/imunologia , Animais , Linhagem Celular , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Dipeptídeos/biossíntese , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Imunidade Inata , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos ICR , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/imunologia , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Peptídeos Cíclicos/biossíntese , Fosforilação , Transporte Proteico/efeitos dos fármacos , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Pele/microbiologia , Pele/patologia , Vibrioses/microbiologia , Vibrioses/patologia , Vibrio vulnificus/genética , Vibrio vulnificus/patogenicidade
17.
J Biol Chem ; 289(52): 36263-74, 2014 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-25398878

RESUMO

Peroxiredoxins (Prxs) are ubiquitous antioxidant enzymes that reduce toxic peroxides. A new Vibrio vulnificus Prx, named Prx3, was identified and characterized in this study. Biochemical and mutational analyses revealed that Prx3 reduces H2O2, utilizing glutaredoxin 3 (Grx3) and glutathione (GSH) as reductants, and requires only N-terminal peroxidatic cysteine for its catalysis. These results, combined with the monomeric size of Prx3 observed under non-reducing conditions, suggested that Prx3 is a Grx3/GSH-dependent 1-Cys Prx and oxidized without forming intermolecular disulfide bonds. The prx3 mutation impaired growth in the medium containing peroxides and reduced virulence in mice, indicating that Prx3 is essential for survival under oxidative stress and pathogenesis of V. vulnificus. The Fe-S cluster regulator IscR activates prx3 by direct binding to a specific binding sequence centered at -44 from the transcription start site. The binding sequence was homologous to the Type 2 IscR-binding sequence, most likely recognized by the Fe-S clusterless apo-IscR in Escherichia coli. The iscR3CA mutant, chromosomally encoding the apo-locked IscR, exhibited 3-fold higher levels of activation of prx3 than the wild type and accumulated more IscR3CA protein in cells. The IscR-dependent activation of prx3 by aerobic growth and iron starvation was also associated with the increase in cellular levels of IscR protein. Taken together, the results suggested that IscR senses iron starvation as well as reactive oxygen species and shifts to the apo-form, which leads to the increase of cellular IscR and in turn prx3 expression, contributing to the survival and virulence of V. vulnificus during pathogenesis.


Assuntos
Proteínas de Bactérias/fisiologia , Peroxirredoxina III/fisiologia , Vibrio vulnificus/enzimologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Sequência de Bases , Sítios de Ligação , Feminino , Regulação Bacteriana da Expressão Gênica , Ferro/metabolismo , Camundongos Endogâmicos ICR , Viabilidade Microbiana , Dados de Sequência Molecular , Estresse Oxidativo , Peroxirredoxina III/química , Regiões Promotoras Genéticas , Transcrição Gênica , Vibrioses/microbiologia , Vibrio vulnificus/patogenicidade , Virulência
18.
Mar Drugs ; 12(4): 1732-56, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24675728

RESUMO

Although human exposure to Gram-negative Vibrio vulnificus (V. vulnificus) lipopolysaccharide (LPS) has been reported to result in septic shock, its impact on the central nervous system's innate immunity remains undetermined. The purpose of this study was to determine whether V. vulnificus MO6-24/O LPS might activate rat microglia in vitro and stimulate the release of superoxide anion (O2⁻), a reactive oxygen species known to cause oxidative stress and neuronal injury in vivo. Brain microglia were isolated from neonatal rats, and then treated with either V. vulnificus MO6-24/O LPS or Escherichia coli O26:B6 LPS for 17 hours in vitro. O2⁻ was determined by cytochrome C reduction, and matrix metalloproteinase-2 (MMP-2) and MMP-9 by gelatinase zymography. Generation of cytokines tumor necrosis factor alpha (TNF-α), interleukin-1 alpha (IL-1α), IL-6, and transforming growth factor-beta 1 (TGF-ß1), chemokines macrophage inflammatory protein (MIP-1α)/chemokine (C-C motif) ligand 3 (CCL3), MIP-2/chemokine (C-X-C motif) ligand 2 (CXCL2), monocyte chemotactic protein-1 (MCP-1)/CCL2, and cytokine-induced neutrophil chemoattractant-2alpha/beta (CINC-2α/ß)/CXCL3, and brain-derived neurotrophic factor (BDNF), were determined by specific immunoassays. Priming of rat microglia by V. vulnificus MO6-24/O LPS in vitro yielded a bell-shaped dose-response curve for PMA (phorbol 12-myristate 13-acetate)-stimulated O2⁻ generation: (1) 0.1-1 ng/mL V. vulnificus LPS enhanced O2⁻ generation significantly but with limited inflammatory mediator generation; (2) 10-100 ng/mL V. vulnificus LPS maximized O2⁻ generation with concomitant release of thromboxane B2 (TXB2), matrix metalloproteinase-9 (MMP-9), and several cytokines and chemokines; (3) 1000-100,000 ng/mL V. vulnificus LPS, with the exception of TXB2, yielded both attenuated O2⁻ production, and a progressive decrease in MMP-9, cytokines and chemokines investigated. Thus concentration-dependent treatment of neonatal brain microglia with V. vulnificus MO6-24/O LPS resulted in a significant rise in O2⁻ production, followed by a progressive decrease in O2⁻ release, with concomitant release of lactic dehydrogenase (LDH), and generation of TXB2, MMP-9, cytokines and chemokines. We hypothesize that the inflammatory mediators investigated may be cytotoxic to microglia in vitro, by an as yet undetermined autocrine mechanism. Although V. vulnificus LPS was less potent than E. coli LPS in vitro, inflammatory mediator release by the former was clearly more efficacious. Finally, we hypothesize that should V. vulnificus LPS gain entry into the CNS, it would be possible that microglia might become activated, resulting in high levels of O2⁻ as well as neuroinflammatory TXB2, MMP-9, cytokines and chemokines.


Assuntos
Escherichia coli/patogenicidade , Lipopolissacarídeos/administração & dosagem , Microglia/metabolismo , Vibrio vulnificus/patogenicidade , Animais , Animais Recém-Nascidos , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/microbiologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Imunidade Inata , Lipopolissacarídeos/isolamento & purificação , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Microglia/imunologia , Microglia/microbiologia , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo , Tromboxano B2/metabolismo
19.
Infect Immun ; 82(2): 569-78, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478072

RESUMO

A mutant that exhibited less cytotoxic activity toward INT-407 human intestinal epithelial cells than the wild type was screened from a random transposon mutant library of Vibrio vulnificus, and an open reading frame encoding an Fe-S cluster regulator, IscR, was identified using a transposon-tagging method. A mutational analysis demonstrated that IscR contributes to mouse mortality as well as cytotoxicity toward the INT-407 cells, indicating that IscR is essential for the pathogenesis of V. vulnificus. A whole-genome microarray analysis revealed that IscR influenced the expression of 67 genes, of which 52 were upregulated and 15 were downregulated. Among these, 12 genes most likely involved in motility and adhesion to host cells, hemolytic activity, and survival under oxidative stress of the pathogen during infection were selected and experimentally verified to be upregulated by IscR. Accordingly, the disruption of iscR resulted in a significant reduction in motility and adhesion to INT-407 cells, in hemolytic activity, and in resistance to reactive oxygen species (ROS) such as H2O2 and tert-butyl hydroperoxide (t-BOOH). Furthermore, the present study demonstrated that iscR expression was induced by exposure of V. vulnificus to the INT-407 cells, and the induction appeared to be mediated by ROS generated by the host cells during infection. Consequently, the combined results indicated that IscR is a global regulator that contributes to the overall success in the pathogenesis of V. vulnificus by regulating the expression of various virulence and survival genes in addition to Fe-S cluster genes.


Assuntos
Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Fatores de Transcrição/metabolismo , Vibrio vulnificus/genética , Animais , Linhagem Celular , Análise Mutacional de DNA , Elementos de DNA Transponíveis , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Feminino , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos ICR , Análise em Microsséries , Mutagênese Insercional , Análise de Sobrevida , Fatores de Transcrição/genética , Vibrioses/microbiologia , Vibrioses/patologia , Vibrio vulnificus/patogenicidade , Fatores de Virulência/biossíntese
20.
Infect Immun ; 82(2): 731-44, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478087

RESUMO

The zoonotic serovar of Vibrio vulnificus (known as biotype 2 serovar E) is the etiological agent of human and fish vibriosis. The aim of the present work was to discover the role of the vulnibactin- and hemin-dependent iron acquisition systems in the pathogenicity of this zoonotic serovar under the hypothesis that both are host-nonspecific virulence factors. To this end, we selected three genes for three outer membrane receptors (vuuA, a receptor for ferric vulnibactin, and hupA and hutR, two hemin receptors), obtained single and multiple mutants as well as complemented strains, and tested them in a series of in vitro and in vivo assays, using eels and mice as animal models. The overall results confirm that hupA and vuuA, but not hutR, are host-nonspecific virulence genes and suggest that a third undescribed host-specific plasmid-encoded system could also be used by the zoonotic serovar in fish. hupA and vuuA were expressed in the internal organs of the animals in the first 24 h of infection, suggesting that they may be needed to achieve the population size required to trigger fatal septicemia. vuuA and hupA were sequenced in strains representative of the genetic diversity of this species, and their phylogenies were reconstructed by multilocus sequence analysis of selected housekeeping and virulence genes as a reference. Given the overall results, we suggest that both genes might form part of the core genes essential not only for disease development but also for the survival of this species in its natural reservoir, the aquatic environment.


Assuntos
Amidas/metabolismo , Hemina/metabolismo , Ferro/metabolismo , Oxazóis/metabolismo , Vibrio vulnificus/metabolismo , Vibrio vulnificus/patogenicidade , Fatores de Virulência/metabolismo , Animais , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Teste de Complementação Genética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Vibrioses/microbiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA