Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Curr Biol ; 34(1): 106-116.e6, 2024 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-38141614

RESUMO

Cellulose is the world's most abundant biopolymer, and similar to its role as a cell wall component in plants, it is a prevalent constituent of the extracellular matrix in bacterial biofilms. Although bacterial cellulose (BC) was first described in the 19th century, it was only recently revealed that it is produced by several distinct types of Bcs secretion systems that feature multiple accessory subunits in addition to a catalytic BcsAB synthase tandem. We recently showed that crystalline cellulose secretion in the Gluconacetobacter genus (α-Proteobacteria) is driven by a supramolecular BcsH-BcsD scaffold-the "cortical belt"-which stabilizes the synthase nanoarrays through an unexpected inside-out mechanism for secretion system assembly. Interestingly, while bcsH is specific for Gluconacetobacter, bcsD homologs are widespread in Proteobacteria. Here, we examine BcsD homologs and their gene neighborhoods from several plant-colonizing ß- and γ-Proteobacteria proposed to secrete a variety of non-crystalline and/or chemically modified cellulosic polymers. We provide structural and mechanistic evidence that through different quaternary structure assemblies BcsD acts with proline-rich BcsH, BcsP, or BcsO partners across the proteobacterial clade to form synthase-interacting intracellular scaffolds that, in turn, determine the biofilm strength and architecture in species with strikingly different physiology and secreted biopolymers.


Assuntos
Celulose , Gluconacetobacter , Proteobactérias/metabolismo , Gluconacetobacter/química , Gluconacetobacter/genética , Gluconacetobacter/metabolismo , Bactérias/metabolismo , Biofilmes
2.
Nat Commun ; 12(1): 5751, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599171

RESUMO

While the major virulence factors for Vibrio cholerae, the cause of the devastating diarrheal disease cholera, have been extensively studied, the initial intestinal colonization of the bacterium is not well understood because non-human adult animals are refractory to its colonization. Recent studies suggest the involvement of an interbacterial killing device known as the type VI secretion system (T6SS). Here, we tested the T6SS-dependent interaction of V. cholerae with a selection of human gut commensal isolates. We show that the pathogen efficiently depleted representative genera of the Proteobacteria in vitro, while members of the Enterobacter cloacae complex and several Klebsiella species remained unaffected. We demonstrate that this resistance against T6SS assaults was mediated by the production of superior T6SS machinery or a barrier exerted by group I capsules. Collectively, our data provide new insights into immunity protein-independent T6SS resistance employed by the human microbiota and colonization resistance in general.


Assuntos
Cólera/microbiologia , Enterobacter cloacae/imunologia , Microbioma Gastrointestinal/imunologia , Klebsiella/imunologia , Sistemas de Secreção Tipo VI/metabolismo , Cápsulas Bacterianas/imunologia , Cápsulas Bacterianas/metabolismo , Cólera/imunologia , Resistência à Doença/imunologia , Enterobacter cloacae/metabolismo , Humanos , Klebsiella/metabolismo , Vibrio cholerae/imunologia , Vibrio cholerae/patogenicidade , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(34): E5044-51, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27503894

RESUMO

The mammalian gastrointestinal tract is colonized by a high-density polymicrobial community where bacteria compete for niches and resources. One key competition strategy includes cell contact-dependent mechanisms of interbacterial antagonism, such as the type VI secretion system (T6SS), a multiprotein needle-like apparatus that injects effector proteins into prokaryotic and/or eukaryotic target cells. However, the contribution of T6SS antibacterial activity during pathogen invasion of the gut has not been demonstrated. We report that successful establishment in the gut by the enteropathogenic bacterium Salmonella enterica serovar Typhimurium requires a T6SS encoded within Salmonella pathogenicity island-6 (SPI-6). In an in vitro setting, we demonstrate that bile salts increase SPI-6 antibacterial activity and that S Typhimurium kills commensal bacteria in a T6SS-dependent manner. Furthermore, we provide evidence that one of the two T6SS nanotube subunits, Hcp1, is required for killing Klebsiella oxytoca in vitro and that this activity is mediated by the specific interaction of Hcp1 with the antibacterial amidase Tae4. Finally, we show that K. oxytoca is killed in the host gut in an Hcp1-dependent manner and that the T6SS antibacterial activity is essential for Salmonella to establish infection within the host gut. Our findings provide an example of pathogen T6SS-dependent killing of commensal bacteria as a mechanism to successfully colonize the host gut.


Assuntos
Antibiose , Proteínas de Bactérias/toxicidade , Salmonelose Animal/microbiologia , Salmonella typhimurium/patogenicidade , Sistemas de Secreção Tipo VI/genética , Fatores de Virulência/toxicidade , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Ácidos e Sais Biliares/farmacologia , Meios de Cultura/química , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Ilhas Genômicas , Klebsiella oxytoca/efeitos dos fármacos , Klebsiella oxytoca/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Salmonelose Animal/patologia , Salmonella typhimurium/genética , Salmonella typhimurium/crescimento & desenvolvimento , Sistemas de Secreção Tipo VI/metabolismo , Fatores de Virulência/biossíntese , Fatores de Virulência/genética
6.
Artigo em Inglês | MEDLINE | ID: mdl-27376031

RESUMO

Pseudomonas aeruginosa is an opportunistic pathogen responsible for many diseases such as chronic lung colonization in cystic fibrosis patients and acute infections in hospitals. The capacity of P. aeruginosa to be pathogenic toward several hosts is notably due to different secretion systems. Amongst them, P. aeruginosa encodes three Type Six Secretion Systems (T6SS), named H1- to H3-T6SS, that act against either prokaryotes and/or eukaryotic cells. They are independent from each other and inject diverse toxins that interact with different components in the host cell. Here we summarize the roles of these T6SSs in the PAO1 strain, as well as the toxins injected and their targets. While H1-T6SS is only involved in antiprokaryotic activity through at least seven different toxins, H2-T6SS and H3-T6SS are also able to target prokaryotic as well as eukaryotic cells. Moreover, recent studies proposed that H2- and H3-T6SS have a role in epithelial cells invasion by injecting at least three different toxins. The diversity of T6SS effectors is astounding and other effectors still remain to be discovered. In this review, we present a table with other putative P. aeruginosa strain PAO1 T6SS-dependent effectors. Altogether, the T6SSs of P. aeruginosa are important systems that help fight other bacteria for their ecological niche, and are important in the pathogenicity process.


Assuntos
Toxinas Bacterianas/metabolismo , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Sistemas de Secreção Tipo VI/metabolismo , Fatores de Virulência/metabolismo , Antibiose , Transporte Proteico , Virulência
8.
Artigo em Inglês | MEDLINE | ID: mdl-26636043

RESUMO

Pseudomonas aeruginosa is a human opportunistic pathogen that causes mortality in cystic fibrosis and immunocompromised patients. While many virulence factors of this pathogen have already been identified, several remain to be discovered. In this respect we set an unprecedented genome-wide screen of a P. aeruginosa expression library based on a yeast growth phenotype. Fifty-one candidates were selected in athree-round screening process. The robustness of the screen was validated by the selection of three well known secreted proteins including one demonstrated virulence factor, the protease LepA. Further in silico sorting of the 51 candidates highlighted three potential new Pseudomonas effector candidates (Pec). By testing the cytotoxicity of wild type P. aeruginosa vs. pec mutants toward macrophages and the virulence in the Caenorhabditis elegans model, we demonstrated that the three selected Pecs are novel virulence factors of P. aeruginosa. Additional cellular localization experiments in the host revealed specific localization for Pec1 and Pec2 that could inform about their respective functions.


Assuntos
Testes Genéticos/métodos , Interações Hospedeiro-Patógeno , Pseudomonas aeruginosa/patogenicidade , Saccharomyces cerevisiae/crescimento & desenvolvimento , Fatores de Virulência/genética , Animais , Caenorhabditis elegans/microbiologia , Linhagem Celular , Humanos , Macrófagos/microbiologia , Camundongos , Pseudomonas aeruginosa/genética , Saccharomyces cerevisiae/genética
9.
mBio ; 6(3): e00712, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26037124

RESUMO

UNLABELLED: Invasion of nonphagocytic cells through rearrangement of the actin cytoskeleton is a common immune evasion mechanism used by most intracellular bacteria. However, some pathogens modulate host microtubules as well by a still poorly understood mechanism. In this study, we aim at deciphering the mechanisms by which the opportunistic bacterial pathogen Pseudomonas aeruginosa invades nonphagocytic cells, although it is considered mainly an extracellular bacterium. Using confocal microscopy and immunofluorescence, we show that the evolved VgrG2b effector of P. aeruginosa strain PAO1 is delivered into epithelial cells by a type VI secretion system, called H2-T6SS, involving the VgrG2a component. An in vivo interactome of VgrG2b in host cells allows the identification of microtubule components, including the γ-tubulin ring complex (γTuRC), a multiprotein complex catalyzing microtubule nucleation, as the major host target of VgrG2b. This interaction promotes a microtubule-dependent internalization of the bacterium since colchicine and nocodazole, two microtubule-destabilizing drugs, prevent VgrG2b-mediated P. aeruginosa entry even if the invasion still requires actin. We further validate our findings by demonstrating that the type VI injection step can be bypassed by ectopic production of VgrG2b inside target cells prior to infection. Moreover, such uncoupling between VgrG2b injection and bacterial internalization also reveals that they constitute two independent steps. With VgrG2b, we provide the first example of a bacterial protein interacting with the γTuRC. Our study offers key insight into the mechanism of self-promoting invasion of P. aeruginosa into human cells via a directed and specific effector-host protein interaction. IMPORTANCE: Innate immunity and specifically professional phagocytic cells are key determinants in the ability of the host to control P. aeruginosa infection. However, among various virulence strategies, including attack, this opportunistic bacterial pathogen is able to avoid host clearance by triggering its own internalization in nonphagocytic cells. We previously showed that a protein secretion/injection machinery, called the H2 type VI secretion system (H2-T6SS), promotes P. aeruginosa uptake by epithelial cells. Here we investigate which H2-T6SS effector enables P. aeruginosa to enter nonphagocytic cells. We show that VgrG2b is delivered by the H2-T6SS machinery into epithelial cells, where it interacts with microtubules and, more particularly, with the γ-tubulin ring complex (γTuRC) known as the microtubule-nucleating center. This interaction precedes a microtubule- and actin-dependent internalization of P. aeruginosa. We thus discovered an unprecedented target for a bacterial virulence factor since VgrG2b constitutes, to our knowledge, the first example of a bacterial protein interacting with the γTuRC.


Assuntos
Proteínas de Bactérias/metabolismo , Endocitose , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Interações Hospedeiro-Patógeno , Microtúbulos/metabolismo , Pseudomonas aeruginosa/fisiologia , Células HeLa , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Ligação Proteica , Mapeamento de Interação de Proteínas , Transporte Proteico , Sistemas de Secreção Tipo VI , Fatores de Virulência/metabolismo
10.
Methods Mol Biol ; 1149: 17-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24818893

RESUMO

Conjugation is a gene transfer process in which a recipient bacterium receives DNA from a donor bacterium by cell-to-cell contact through conjugative pili. Conjugation is mediated by certain plasmids or transposons. Here, we describe plasmid conjugation.


Assuntos
Conjugação Genética , Técnicas de Transferência de Genes , Antibacterianos/farmacologia , Conjugação Genética/efeitos dos fármacos , Plasmídeos/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos
11.
Trends Microbiol ; 22(6): 304-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24836109

RESUMO

Two type VI secreted phospholipases D of Pseudomonas aeruginosa were identified as trans-kingdom virulence effectors, targeting both prokaryotic and eukaryotic host cells. Each of them triggers killing bacterial competitors and internalization into non-phagocytic cells. These type VI lipolytic enzymes are widely distributed among pathogens and may constitute a conserved strategy.


Assuntos
Células Eucarióticas/microbiologia , Fosfolipase D/metabolismo , Pseudomonas aeruginosa/patogenicidade , Fenômenos Fisiológicos Bacterianos , Interações Hospedeiro-Patógeno , Interações Microbianas , Transdução de Sinais
12.
PLoS One ; 8(10): e76030, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24204589

RESUMO

Three Type VI Secretion System (T6SS) loci called H1- to H3-T6SS coexist in Pseudomonas aeruginosa. H1-T6SS targets prokaryotic cells whereas H2-T6SS mediates interactions with both eukaryotic and prokaryotic host cells. Little is known about the third system, except that it may be connected to H2-T6SS during the host infection. Here we show that H3-T6SS is required for P. aeruginosa PAO1 virulence in the worm model. We demonstrate that the two putative H3-T6SS operons, called "left" and "right", are coregulated with H2-T6SS by the Las and Rhl Quorum Sensing systems. Interestingly, the RpoN σ54 factor has divergent effects on the three operons. As for many T6SSs, RpoN activates the expression of H3-T6SS left. However, RpoN unexpectedly represses the expression of H3-T6SS right and also H2-T6SS. Sfa2 and Sfa3 are putative enhancer binding proteins encoded on H2-T6SS and H3-T6SS left. In other T6SSs EBPs can act as σ54 activators to promote T6SS transcription. Strikingly, we found that the RpoN effects of H3-T6SS are Sfa-independent while the RpoN mediated repression of H2-T6SS is Sfa2-dependent. This is the first example of RpoN repression of a T6SS being mediated by a T6SS-encoded EBP.


Assuntos
Sistemas de Secreção Bacterianos/genética , Pseudomonas aeruginosa/fisiologia , Fator sigma/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sequência de Bases , Caenorhabditis elegans/microbiologia , Regulação Bacteriana da Expressão Gênica , Ordem dos Genes , Dados de Sequência Molecular , Família Multigênica , Mutação , Pseudomonas aeruginosa/patogenicidade , Percepção de Quorum/genética , Virulência/genética
13.
J Biol Chem ; 287(32): 27095-105, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22665491

RESUMO

The genome of Pseudomonas aeruginosa PAO1 contains three type VI secretion systems (T6SSs) called H1-, H2-, and H3-T6SS. The H1-T6SS secretes three identified toxins that target other bacteria, providing a fitness advantage for P. aeruginosa, and likely contributes to bacterial pathogenesis in chronic infections. However, no specific substrates or defined roles have been described for the two other systems. Here, we demonstrate that the expression of H2-T6SS genes of strain PAO1 is up-regulated during the transition from exponential to stationary phase growth and regulated by the Las and Rhl quorum sensing systems. In addition, we identify two putative Fur boxes in the promoter region and find that H2-T6SS transcription is negatively regulated by iron. We also show that the H2-T6SS system enhances bacterial uptake into HeLa cells (75% decrease in internalization with a H2-T6SS mutant) and into lung epithelial cells through a phosphatidylinositol 3-kinase-dependent pathway that induces Akt activation in the host cell (50% decrease in Akt phosphorylation). Finally, we show that H2-T6SS plays a role in P. aeruginosa virulence in the worm model. Thus, in contrast to H1-T6SS, H2-T6SS modulates interaction with eukaryotic host cells. Together, T6SS can carry out different functions that may be important in establishing chronic P. aeruginosa infections in the human host.


Assuntos
Endocitose , Células Epiteliais/metabolismo , Pseudomonas aeruginosa/metabolismo , Percepção de Quorum , Sequência de Bases , DNA Bacteriano , Genes Bacterianos , Células HeLa , Humanos , Pseudomonas aeruginosa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA