Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Infect Immun ; 91(12): e0030323, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37982617

RESUMO

Klebsiella pneumoniae is a Gram-negative bacterium that causes a variety of human diseases, ranging from pneumonia to urinary tract infections and invasive diseases. The emergence of K. pneumoniae strains that are resistant to multiple antibiotics has made treatment more complex and led to K. pneumoniae becoming a global health threat. Addressing this threat necessitates the development of new therapeutic strategies to combat this pathogen, including strategies to overcome antimicrobial resistance and therapeutics for novel targets such as antivirulence. Here, we investigated the function of TolC, an outer membrane protein essential for the function of tripartite transporters, in K. pneumoniae. Mutation of tolC rendered K. pneumoniae hypersensitive to multiple antibiotics. Moreover, the tolC mutation impaired capsule production and affected the expression of key capsule biosynthetic genes, indicating a regulatory role for TolC in capsule biosynthesis. Additionally, TolC was essential for growth under iron-limiting conditions, suggesting its involvement in iron acquisition. The tolC mutant exhibited increased adherence to human enterocytes and enhanced serum sensitivity. In the Galleria mellonella infection model, the tolC mutant displayed reduced virulence compared to the wild type. Our findings highlight the pleiotropic role of TolC in K. pneumoniae pathobiology, influencing antimicrobial resistance, capsule production, iron homeostasis, adherence to host cells, and virulence. Understanding the multifaceted role of TolC in K. pneumoniae may guide the development of new therapeutic strategies against this pathogen. .


Assuntos
Infecções por Klebsiella , Klebsiella pneumoniae , Humanos , Klebsiella pneumoniae/genética , Virulência , Antibacterianos , Farmacorresistência Bacteriana , Infecções por Klebsiella/tratamento farmacológico , Infecções por Klebsiella/microbiologia , Ferro
2.
Infect Immun ; 89(7): e0014721, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33941578

RESUMO

Multidrug efflux systems belonging to the resistance-nodulation-cell division (RND) family are ubiquitous in Gram-negative bacteria and are critical for antimicrobial resistance. This realization has led to efforts to develop efflux pump inhibitors (EPI) for use as adjuvants for antibiotic treatment of resistant organisms. However, the functions of RND transporters extend beyond antimicrobial resistance to include physiological functions that are critical for pathogenesis, suggesting that EPIs could also be used as antivirulence therapeutics. This was documented in the enteric pathogen Vibrio cholerae, in which EPIs were shown to attenuate the production of the critical virulence factors cholera toxin (CT) and the toxin-coregulated pilus (TCP). In this study, we investigated the antivirulence mechanism of action of the EPI phenyl-arginine-ß-naphthylamide (PAßN) on V. cholerae. Using bioassays, we documented that PAßN inhibited virulence factor production in three epidemic V. cholerae isolates. Transcriptional reporter studies and mutant analysis indicated that PAßN initiated a ToxR-dependent regulatory circuit to activate leuO expression and that LeuO repressed the expression of the critical virulence activator aphA to attenuate CT and TCP production. The antivirulence activity of PAßN was found to be dependent on the ToxR periplasmic sensing domain (PPD), suggesting that a feedback mechanism was involved in its activity. Collectively, the data indicated that PAßN inhibited V. cholerae virulence factor production by activating a ToxR-dependent metabolic feedback mechanism to repress the expression of the ToxR virulence regulon. This suggests that efflux pump inhibitors could be used as antivirulence therapeutics for the treatment of cholera and perhaps that of other Gram-negative pathogens.


Assuntos
Proteínas de Bactérias/genética , Cólera/microbiologia , Proteínas de Ligação a DNA/genética , Dipeptídeos/metabolismo , Fatores de Transcrição/genética , Vibrio cholerae/fisiologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Dipeptídeos/farmacologia , Suscetibilidade a Doenças , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Vibrio cholerae/efeitos dos fármacos , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
3.
Infect Immun ; 89(10): e0024221, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34310890

RESUMO

Vibrio cholerae is a Gram-negative bacterium that causes the enteric disease cholera. V. cholerae colonization of the human intestine is dependent on the expression of both virulence genes and environmental adaptation genes involved in antimicrobial resistance. The expression of virulence genes, including the genes encoding the main virulence factors cholera toxin (CT) and the toxin-coregulated pilus (TCP), are coordinately regulated by the ToxR regulon. Tripartite transport systems belonging to the ATP binding cassette, major facilitator, and resistance-nodulation-division families are critical for V. cholerae pathogenesis. Transport systems belonging to these families contribute to myriad phenotypes, including protein secretion, antimicrobial resistance, and virulence. TolC plays a central role in bacterial physiology by functioning as the outer membrane pore protein for tripartite transport systems. Consistent with this, V. cholerae tolC was previously found to be required for MARTX toxin secretion and antimicrobial resistance. Here, we investigated the contribution of TolC to V. cholerae virulence. We documented that tolC was required for CT and TCP production in O1 El Tor V. cholerae. This phenotype was linked to repression of the critical ToxR regulon transcription factor aphA. Decreased aphA transcription correlated with increased expression of the LysR-family transcription factor leuO. Deletion of leuO restored aphA expression, and CT and TCP production, in a tolC mutant. The collective results document that tolC is required for ToxR regulon expression and further suggest that tolC participates in an efflux-dependent feedback circuit to regulate virulence gene expression.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Regulon/genética , Fatores de Transcrição/genética , Vibrio cholerae/genética , Animais , Cólera/microbiologia , Toxina da Cólera/genética , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica/genética , Fenótipo , Suínos , Virulência/genética , Fatores de Virulência/genética
4.
Infect Immun ; 89(12): e0044121, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34543121

RESUMO

Cholera is an epidemic disease caused by the Gram-negative bacterium Vibrio cholerae. V. cholerae is found in aquatic ecosystems and infects people through the consumption of V. cholerae-contaminated food or water. Following ingestion, V. cholerae responds to host cues to activate the expression of critical virulence genes that are under the control of a hierarchical regulatory system called the ToxR regulon. The ToxR regulon is tightly regulated and is expressed in vitro only under special growth conditions referred to as AKI conditions. AKI conditions have been instrumental in elucidating V. cholerae virulence regulation, but the chemical cues within AKI medium that activate virulence gene expression are unknown. In this study, we fractionated AKI medium on a reverse-phase chromatography column (RPCC) and showed that the virulence-activating molecules were retained on the RPCC column and recovered in the eluate. Liquid chromatography-high-resolution mass spectrometry (LC-HRMS) analysis of the eluate revealed the presence of a known ToxR regulon activator, taurocholate, and other bile salts. The RPCC eluate activated the ToxR regulon when added to noninducing medium and promoted TcpP dimerization in a two-hybrid system, consistent with taurocholate being responsible for the virulence-inducing activity of AKI medium. Additional experiments using purified bile salts showed that the ToxR regulon was preferentially activated in response to primary bile acids. The results of this study shed light on the chemical cues involved in V. cholerae virulence activation and suggested that V. cholerae virulence genes are modulated in response to regionally specific bile acid species in the intestine.


Assuntos
Proteínas de Bactérias/genética , Ácidos e Sais Biliares/metabolismo , Cólera/metabolismo , Cólera/microbiologia , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Regulon , Fatores de Transcrição/genética , Vibrio cholerae/fisiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/isolamento & purificação , Cromatografia Líquida , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/isolamento & purificação , Humanos , Espectrometria de Massas , Fatores de Transcrição/química , Fatores de Transcrição/isolamento & purificação , Vibrio cholerae/patogenicidade , Virulência/genética , Fatores de Virulência/genética
5.
PLoS Pathog ; 14(1): e1006804, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29304169

RESUMO

Resistance-nodulation-division (RND) efflux systems are ubiquitous transporters in Gram-negative bacteria that are essential for antibiotic resistance. The RND efflux systems also contribute to diverse phenotypes independent of antimicrobial resistance, but the mechanism by which they affect most of these phenotypes is unclear. This is the case in Vibrio cholerae where the RND systems function in antimicrobial resistance and virulence factor production. Herein, we investigated the linkage between RND efflux and V. cholerae virulence. RNA sequencing revealed that the loss of RND efflux affected the activation state of periplasmic sensing systems including the virulence regulator ToxR. Activation of ToxR in an RND null mutant resulted in ToxR-dependent transcription of the LysR-family regulator leuO. Increased leuO transcription resulted in the repression of the ToxR virulence regulon and attenuated virulence factor production. Consistent with this, leuO deletion restored virulence factor production in an RND-null mutant, but not its ability to colonize infant mice; suggesting that RND efflux was epistatic to virulence factor production for colonization. The periplasmic sensing domain of ToxR was required for the induction of leuO transcription in the RND null mutant, suggesting that ToxR responded to metabolites that accumulated in the periplasm. Our results suggest that ToxR represses virulence factor production in response to metabolites that are normally effluxed from the cell by the RND transporters. We propose that impaired RND efflux results in periplasmic metabolite accumulation, which then activates periplasmic sensors including ToxR and two-component regulatory systems to initiate the expression of adaptive responses.


Assuntos
Adaptação Fisiológica/fisiologia , Proteínas de Bactérias/fisiologia , Farmacorresistência Bacteriana , Proteínas de Membrana Transportadoras/fisiologia , Proteínas Periplásmicas/fisiologia , Vibrio cholerae , Fatores de Virulência/metabolismo , Adaptação Fisiológica/genética , Animais , Animais Recém-Nascidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica , Proteínas de Membrana Transportadoras/genética , Camundongos , Organismos Geneticamente Modificados , Proteínas Periplásmicas/genética , Proteínas Periplásmicas/metabolismo , Vibrio cholerae/genética , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidade , Fatores de Virulência/genética
6.
Infect Immun ; 87(3)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30617203

RESUMO

Indole is a degradation product of tryptophan that functions as a signaling molecule in many bacteria. This includes Vibrio cholerae, where indole was shown to regulate biofilm and type VI secretion in nontoxigenic environmental isolates. Indole is also produced by toxigenic V. cholerae strains in the human intestine, but its significance in the host is unknown. We investigated the effects of indole on toxigenic V. cholerae O1 El Tor during growth under virulence inducing conditions. The indole transcriptome was defined by RNA sequencing and showed widespread changes in the expression of genes involved in metabolism, biofilm production, and virulence factor production. In contrast, genes involved in type VI secretion were not affected by indole. We subsequently found that indole repressed genes involved in V. cholerae pathogenesis, including the ToxR virulence regulon. Consistent with this, indole inhibited cholera toxin and toxin-coregulated pilus production in a dose-dependent manner. The effects of indole on virulence factor production and biofilm were linked to ToxR and the ToxR-dependent regulator LeuO. The expression of leuO was increased by exogenous indole and linked to repression of the ToxR virulence regulon. This process was dependent on the ToxR periplasmic domain, suggesting that indole was a ToxR agonist. This conclusion was further supported by results showing that the ToxR periplasmic domain contributed to indole-mediated increased biofilm production. Collectively, our results suggest that indole may be a niche-specific cue that can function as a ToxR agonist to modulate virulence gene expression and biofilm production in V. cholerae.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Indóis/farmacologia , Fatores de Transcrição/metabolismo , Vibrio cholerae/efeitos dos fármacos , Vibrio cholerae/metabolismo , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Proteínas de Bactérias/genética , Toxina da Cólera/genética , Toxina da Cólera/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Mutação , Regulon , Fatores de Transcrição/genética , Vibrio cholerae/genética
7.
Infect Immun ; 84(11): 3161-3171, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27550934

RESUMO

Vibrio cholerae is an intestinal pathogen that causes the diarrheal disease cholera. Colonization of the intestine depends upon the expression of genes that allow V. cholerae to overcome host barriers, including low pH, bile acids, and the innate immune system. ToxR is a major contributor to this process. ToxR is a membrane-spanning transcription factor that coordinates gene expression in response to environmental cues. In previous work we showed that ToxR upregulated leuO expression in response to bile salts. LeuO is a LysR family transcription factor that contributes to acid tolerance, bile resistance, and biofilm formation in V. cholerae Here, we investigated the function of ToxR and LeuO in cationic antimicrobial peptide (CAMP) resistance. We report that ToxR and LeuO contribute to CAMP resistance by regulating carRS transcription. CarRS is a two-component regulatory system that positively regulates almEFG expression. AlmEFG confers CAMP resistance by glycinylation of lipid A. We found that the expression of carRS and almEFG and the polymyxin B MIC increased in mutants lacking toxRS or leuO Conversely, leuO overexpression decreased the polymyxin B MIC. Furthermore, we found that LeuO directly bound to the carRS promoter and that ToxR-dependent activation of leuO transcription regulated carRS transcription in response to bile salts. Our results suggest that LeuO functions downstream of ToxR to modulate carRS expression in response to environmental cues. This study extends the functional role of ToxR and LeuO in environmental adaptation to include cell surface remodeling and CAMP resistance.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Lipídeo A/metabolismo , Regulon/fisiologia , Fatores de Transcrição/genética , Vibrio cholerae/genética , Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Lipídeo A/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Vibrio cholerae/metabolismo
8.
J Bacteriol ; 197(22): 3499-510, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26303831

RESUMO

UNLABELLED: Vibrio cholerae is an aquatic organism and facultative human pathogen that colonizes the small intestine. In the small intestine, V. cholerae is exposed to a variety of antimicrobial compounds, including bile. V. cholerae resistance to bile is multifactorial and includes alterations in the membrane permeability barrier that are mediated by ToxR, a membrane-associated transcription factor. ToxR has also been shown to be required for activation of the LysR family transcription factor leuO in response to cyclic dipeptides. LeuO has been implicated in the regulation of multiple V. cholerae phenotypes, including biofilm production and virulence. In this study, we investigated the effects of bile on leuO expression. We show that leuO transcription increased in response to bile and bile salts but not in response to other detergents. The bile-dependent increase in leuO expression was dependent on ToxR, which was found to bind directly to the leuO promoter. The periplasmic domain of ToxR was required for basal leuO expression and for the bile-dependent induction of both leuO and ompU transcription. V. cholerae mutants that did not express leuO exhibited increased bile susceptibility, suggesting that LeuO contributes to bile resistance. Our collective results demonstrate that ToxR activates leuO expression in response to bile and that LeuO is a component of the ToxR-dependent responses that contribute to bile resistance. IMPORTANCE: The success of Vibrio cholerae as a human pathogen is dependent upon its ability to rapidly adapt to changes in its growth environment. Growth in the human gastrointestinal tract requires the expression of genes that provide resistance to host antimicrobial compounds, including bile. In this work, we show for the first time that the LysR family regulator LeuO mediates responses in V. cholerae that contribute to bile resistance.


Assuntos
Proteínas de Bactérias/metabolismo , Ácidos e Sais Biliares/farmacologia , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Fatores de Transcrição/metabolismo , Vibrio cholerae/efeitos dos fármacos , Vibrio cholerae/metabolismo , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Fatores de Transcrição/genética , Vibrio cholerae/genética
9.
Microbiology (Reading) ; 161(12): 2434-43, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26424466

RESUMO

Vibrio cholerae is a neutrophilic enteric pathogen that is extremely sensitive to acid. As V. cholerae passages through the host gastrointestinal tract it is exposed to a variety of environmental stresses including low pH and volatile fatty acids. Exposure to acidic environments induces expression of the V. cholerae acid tolerance response. A key component of the acid tolerance response is the cad system, which is encoded by cadC and the cadBA operon. CadB is a lysine/cadaverine antiporter and CadA is a lysine decarboxylase and these function together to counter low intracellular and extracellular pH. CadC is a membrane-associated transcription factor that activates cadBA expression in response to acidic conditions. Herein we investigated the role of the LysR-type transcriptional regulator LeuO in the V. cholerae acid tolerance response. Transcriptional reporter assays revealed that leuO expression repressed cadC transcription, indicating that LeuO was a cadC repressor. Consistent with this, leuO expression was inversely linked to lysine decarboxylase production and leuO overexpression resulted in increased sensitivity to organic acids. Overexpression of leuO in a cadA mutant potentiated killing by organic acids, suggesting that the function of leuO in the acid tolerance response extended beyond its regulation of the cad system. Collectively, these studies have identified a new physiological role for LeuO in V. cholerae acid tolerance.


Assuntos
Ácidos/metabolismo , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Óperon , Transativadores/metabolismo , Vibrio cholerae/metabolismo , Proteínas de Bactérias/genética , Transativadores/genética , Vibrio cholerae/genética
10.
Proc Natl Acad Sci U S A ; 109(44): 18084-9, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23071317

RESUMO

Cytosolic bacterial pathogens require extensive metabolic adaptations within the host to replicate intracellularly and cause disease. In phagocytic cells such as macrophages, these pathogens must respond rapidly to nutrient limitation within the harsh environment of the phagosome. Many cytosolic pathogens escape the phagosome quickly (15-60 min) and thereby subvert this host defense, reaching the cytosol where they can replicate. Although a great deal of research has focused on strategies used by bacteria to resist antimicrobial phagosomal defenses and transiently pass through this compartment, the metabolic requirements of bacteria in the phagosome are largely uncharacterized. We previously identified a Francisella protein, FTN_0818, as being essential for intracellular replication and involved in virulence in vivo. We now show that FTN_0818 is involved in biotin biosynthesis and required for rapid escape from the Francisella-containing phagosome (FCP). Addition of biotin complemented the phagosomal escape defect of the FTN_0818 mutant, demonstrating that biotin is critical for promoting rapid escape during the short time that the bacteria are in the phagosome. Biotin also rescued the attenuation of the FTN_0818 mutant during infection in vitro and in vivo, highlighting the importance of this process. The key role of biotin in phagosomal escape implies biotin may be a limiting factor during infection. We demonstrate that a bacterial metabolite is required for phagosomal escape of an intracellular pathogen, providing insight into the link between bacterial metabolism and virulence, likely serving as a paradigm for other cytosolic pathogens.


Assuntos
Biotina/metabolismo , Francisella/metabolismo , Fagossomos/metabolismo , Animais , Macrófagos/microbiologia , Camundongos
11.
Infect Immun ; 82(7): 2980-91, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24799626

RESUMO

The Cpx two-component regulatory system has been shown in Escherichia coli to alleviate stress caused by misfolded cell envelope proteins. The Vibrio cholerae Cpx system was previously found to respond to cues distinct from those in the E. coli system, suggesting that this system fulfills a different physiological role in the cholera pathogen. Here, we used microarrays to identify genes that were regulated by the V. cholerae Cpx system. Our observations suggest that the activation of the V. cholerae Cpx system does not induce expression of genes involved in the mitigation of stress generated by misfolded cell envelope proteins but promotes expression of genes involved in antimicrobial resistance. In particular, activation of the Cpx system induced expression of the genes encoding the VexAB and VexGH resistance-nodulation-division (RND) efflux systems and their cognate outer membrane pore protein TolC. The promoters for these loci contained putative CpxR consensus binding sites, and ectopic cpxR expression activated transcription from the promoters for the RND efflux systems. CpxR was not required for intrinsic antimicrobial resistance, but CpxR activation enhanced resistance to antimicrobial substrates of VexAB and VexGH. Mutations that inactivated VexAB or VexGH efflux activity resulted in the activation of the Cpx response, suggesting that vexAB and vexGH and the cpxP-cpxRA system are reciprocally regulated. We speculate that the reciprocal regulation of the V. cholerae RND efflux systems and the Cpx two-component system is mediated by the intracellular accumulation of an endogenously produced metabolic by-product that is normally extruded from the cell by the RND efflux systems.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Vibrio cholerae/metabolismo , Proteínas de Bactérias/genética , Sequência de Bases , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ligação Genética , Proteínas de Membrana Transportadoras , Dados de Sequência Molecular , Mutação , Vibrio cholerae/genética
12.
Microbiology (Reading) ; 160(Pt 6): 1054-1062, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24644247

RESUMO

Vibrio cholerae has been shown to produce a cyclic dipeptide, cyclo(phenylalanine-proline) (cFP), that functions to repress virulence factor production. The objective of this study was to determine if heterologous cyclic dipeptides could repress V. cholerae virulence factor production. To that end, three synthetic cyclic dipeptides that differed in their side chains from cFP were assayed for virulence inhibitory activity in V. cholerae. The results revealed that cyclo(valine-valine) (cVV) inhibited virulence factor production by a ToxR-dependent process that resulted in the repression of the virulence regulator aphA. cVV-dependent repression of aphA was found to be independent of known aphA regulatory genes. The results demonstrated that V. cholerae was able to respond to exogenous cyclic dipeptides and implicated the hydrophobic amino acid side chains on both arms of the cyclo dipeptide scaffold as structural requirements for inhibitory activity. The results further suggest that cyclic dipeptides have potential as therapeutics for cholera treatment.


Assuntos
Antibacterianos/farmacologia , Expressão Gênica/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Valina/farmacologia , Vibrio cholerae/efeitos dos fármacos , Fatores de Virulência/biossíntese
13.
Plasmid ; 76: 87-94, 2014 11.
Artigo em Inglês | MEDLINE | ID: mdl-25451701

RESUMO

We report the construction of a tetracycline inducible expression vector that allows regulated gene expression in the enteric pathogen Vibrio cholerae. The expression vector, named pXB300, contains the tetracycline regulatory elements from Tn10, a multiple cloning site downstream of the tetA promoter and operator sequences, a ColE1 origin of replication, a ß-lactamase resistance gene for positive selection, and the hok/sok addiction system for selection in the absence of antibiotic. The function of the tetracycline expression system was demonstrated by cloning lacZ under control of the tetA promoter and quantifying ß-galactosidase expression in Escherichia coli and V. cholerae. The utility for pXB300 was documented by complementation of V. cholerae virulence mutants during growth under virulence inducing conditions. The results showed that pXB300 allowed high-level expression of recombinant genes with linear induction in response to the exogenous concentration of the inducer anhydrotetracycline. We further show that pXB300 was reliably maintained in V. cholerae during growth in the absence of antibiotic selection.


Assuntos
Regulação Bacteriana da Expressão Gênica , Vetores Genéticos/efeitos dos fármacos , Tetraciclina/farmacologia , Vibrio cholerae/genética , Antiporters/genética , Proteínas de Bactérias/genética , Toxina da Cólera/genética , Toxina da Cólera/metabolismo , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Teste de Complementação Genética , Vetores Genéticos/genética , Mutação , Plasmídeos/efeitos dos fármacos , Plasmídeos/genética , Regiões Promotoras Genéticas , Tetraciclinas/farmacologia , Fatores de Transcrição/genética , Vibrio cholerae/efeitos dos fármacos , Vibrio cholerae/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
14.
Mol Microbiol ; 86(3): 611-27, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22966934

RESUMO

Modification of specific Gram-negative bacterial cell envelope components, such as capsule, O-antigen and lipid A, are often essential for the successful establishment of infection. Francisella species express lipid A molecules with unique characteristics involved in circumventing host defences, which significantly contribute to their virulence. In this study, we show that NaxD, a member of the highly conserved YdjC superfamily, is a deacetylase required for an important modification of the outer membrane component lipid A in Francisella. Mass spectrometry analysis revealed that NaxD is essential for the modification of a lipid A phosphate with galactosamine in Francisella novicida, a model organism for the study of highly virulent Francisella tularensis. Significantly, enzymatic assays confirmed that this protein is necessary for deacetylation of its substrate. In addition, NaxD was involved in resistance to the antimicrobial peptide polymyxin B and critical for replication in macrophages and in vivo virulence. Importantly, this protein is also required for lipid A modification in F. tularensis as well as Bordetella bronchiseptica. Since NaxD homologues are conserved among many Gram-negative pathogens, this work has broad implications for our understanding of host subversion mechanisms of other virulent bacteria.


Assuntos
Amidoidrolases/metabolismo , Proteínas de Bactérias/metabolismo , Francisella/enzimologia , Francisella/patogenicidade , Infecções por Bactérias Gram-Negativas/microbiologia , Lipídeo A/metabolismo , Amidoidrolases/química , Amidoidrolases/genética , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular , Feminino , Francisella/genética , Francisella/metabolismo , Francisella tularensis/enzimologia , Francisella tularensis/genética , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Alinhamento de Sequência , Virulência
15.
Front Cell Infect Microbiol ; 13: 1203487, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37256112

RESUMO

Resistance Nodulation Division (RND) efflux systems are ubiquitous transporters in gram-negative bacteria that provide protection against antimicrobial agents and thereby enhance survival in virtually all environments these prokaryotes inhabit. Vibrio cholerae is a dual lifestyle enteric pathogen that spends much of its existence in aquatic environments. An unwitting encounter with a human host can lead to V. cholerae intestinal colonization by strains that encode cholera toxin and toxin co-regulated pilus virulence factors leading to potentially fatal cholera diarrhea and dissemination in the environment. Adaptive response mechanisms to host factors encountered by these pathogens are therefore critical both to engage survival mechanisms such as RND-mediated transporters and to induce timely expression of virulence factors. Sensing of cues encountered in the host may therefore activate more than protective responses such as efflux systems, but also be coordinated to initiate expression of virulence factors. This review summarizes recent advances that contribute towards the understanding of RND efflux physiological functions and how the transport systems interface with the regulation of virulence factor production in V. cholerae.


Assuntos
Cólera , Vibrio cholerae , Humanos , Vibrio cholerae/metabolismo , Proteínas de Bactérias/genética , Toxina da Cólera/metabolismo , Fatores de Virulência/metabolismo , Transporte Biológico , Cólera/microbiologia , Proteínas de Membrana Transportadoras/metabolismo , Regulação Bacteriana da Expressão Gênica
16.
BMC Microbiol ; 11: 179, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21819572

RESUMO

BACKGROUND: A number of studies have revealed that Francisella tularensis (FT) suppresses innate immune responses such as chemokine/cytokine production and neutrophil recruitment in the lungs following pulmonary infection via an unidentified mechanism. The ability of FT to evade early innate immune responses could be a very important virulence mechanism for this highly infectious bacterial pathogen. RESULTS: Here we describe the characterization of a galU mutant strain of FT live vaccine strain (LVS). We show that the galU mutant was highly attenuated in a murine model of tularemia and elicited more robust innate immune responses than the wild-type (WT) strain. These studies document that the kinetics of chemokine expression and neutrophil recruitment into the lungs of mice challenged with the galU mutant strain are significantly more rapid than observed with WT FT, despite the fact that there were no observed differences in TLR2 or TLR4 signaling or replication/dissemination kinetics during the early stages of infection. We also show that the galU mutant had a hypercytotoxic phenotype and more rapidly induced the production of IL-1ß following infection either in vitro or in vivo, indicating that attenuation of the galU mutant strain may be due (in part) to more rapid activation of the inflammasome and/or earlier death of FT infected cells. Furthermore, we show that infection of mice with the galU mutant strain elicits protective immunity to subsequent challenge with WT FT. CONCLUSIONS: Disruption of the galU gene of FTLVS has little (if any) effect on in vivo infectivity, replication, or dissemination characteristics, but is highly attenuating for virulence. The attenuated phenotype of this mutant strain of FT appears to be related to its increased ability to induce innate inflammatory responsiveness, resulting in more rapid recruitment of neutrophils to the lungs following pneumonic infection, and/or to its ability to kill infected cells in an accelerated fashion. These results have identified two potentially important virulence mechanisms used by FT. These findings could also have implications for design of a live attenuated vaccine strain of FT because sublethal infection of mice with the galU mutant strain of FTLVS promoted development of protective immunity to WT FTLVS.


Assuntos
Francisella tularensis/genética , Francisella tularensis/patogenicidade , Tularemia/microbiologia , Tularemia/patologia , UTP-Glucose-1-Fosfato Uridililtransferase/deficiência , Fatores de Virulência/deficiência , Animais , Quimiocinas/metabolismo , Modelos Animais de Doenças , Francisella tularensis/imunologia , Humanos , Interleucina-1beta/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Doenças dos Roedores/microbiologia , Doenças dos Roedores/patologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Virulência
17.
Microbiol Resour Announc ; 10(3)2021 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-33478998

RESUMO

Vibrio cholerae is a global health threat and a model enteric pathogen that causes the human disease cholera. Here, we report the complete genome sequence of the seventh-pandemic V. cholerae O1 El Tor strain C6706.

18.
Microbiol Resour Announc ; 10(5)2021 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-33541885

RESUMO

Klebsiella pneumoniae is a member of Enterobacteriaceae that causes a multitude of infections in compromised and healthy individuals. The rise of hypervirulent and multiple-drug-resistant K. pneumoniae strains has made this organism a global health threat. Here, we report the complete genome sequence of K. pneumoniae strain ATCC 43816.

19.
J Bacteriol ; 192(14): 3829-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20453095

RESUMO

Cyclo(Phe-Pro) is a cyclic dipeptide produced by multiple Vibrio species. In this work, we present evidence that cyclo(Phe-Pro) inhibits the production of the virulence factors cholera toxin (CT) and toxin-coregulated pilus (TCP) in O1 El Tor Vibrio cholerae strain N16961 during growth under virulence gene-inducing conditions. The cyclo(Phe-Pro) inhibition of CT and TCP production correlated with reduced transcription of the virulence regulator tcpPH and was alleviated by overexpression of tcpPH.


Assuntos
Proteínas de Bactérias/metabolismo , Toxina da Cólera/biossíntese , Dipeptídeos/farmacologia , Fímbrias Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Vibrio cholerae/metabolismo , Proteínas de Bactérias/genética , Fímbrias Bacterianas/genética , Vibrio cholerae/genética
20.
BMC Microbiol ; 10: 76, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20226053

RESUMO

BACKGROUND: Francisella tularensis (FT) is a gram-negative facultative intracellular coccobacillus and is the causal agent of a life-threatening zoonotic disease known as tularemia. Although FT preferentially infects phagocytic cells of the host, recent evidence suggests that a significant number of bacteria can be found extracellularly in the plasma fraction of the blood during active infection. This observation suggests that the interaction between FT and host plasma components may play an important role in survival and dissemination of the bacterium during the course of infection. Plasminogen (PLG) is a protein zymogen that is found in abundance in the blood of mammalian hosts. A number of both gram-positive and gram-negative bacterial pathogens have the ability to bind to PLG, giving them a survival advantage by increasing their ability to penetrate extracellular matrices and cross tissue barriers. RESULTS: We show that PLG binds to the surface of FT and that surface-bound PLG can be activated to plasmin in the presence of tissue PLG activator in vitro. In addition, using Far-Western blotting assays coupled with proteomic analyses of FT outer membrane preparations, we have identified several putative PLG-binding proteins of FT. CONCLUSIONS: The ability of FT to acquire surface bound PLG that can be activated on its surface may be an important virulence mechanism that results in an increase in initial infectivity, survival, and/or dissemination of this bacterium in vivo.


Assuntos
Proteínas de Bactérias/metabolismo , Francisella tularensis/metabolismo , Proteínas de Membrana/metabolismo , Plasminogênio/metabolismo , Aminocaproatos/farmacologia , Far-Western Blotting , Ensaio de Imunoadsorção Enzimática , Fibrinolisina/metabolismo , Fibronectinas/metabolismo , Francisella tularensis/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Microscopia Confocal , Ligação Proteica , Tularemia/sangue , Tularemia/microbiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa