Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Bacteriol ; 192(19): 4894-903, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20675476

RESUMO

Salmonella enterica species are exposed to envelope stresses due to their environmental and infectious lifestyles. Such stresses include amphipathic cationic antimicrobial peptides (CAMPs), and resistance to these peptides is an important property for microbial virulence for animals. Bacterial mechanisms used to sense and respond to CAMP-induced envelope stress include the RcsFCDB phosphorelay, which contributes to survival from polymyxin B exposure. The Rcs phosphorelay includes two inner membrane (IM) proteins, RcsC and RcsD; the response regulator RcsB; the accessory coregulator RcsA; and an outer membrane bound lipoprotein, RcsF. Transcriptional activation of the Rcs regulon occurred within minutes of exposure to CAMP and during the first detectable signs of CAMP-induced membrane disorder. Rcs transcriptional activation by CAMPs required RcsF and preservation of its two internal disulfide linkages. The rerouting of RcsF to the inner membrane or its synthesis as an unanchored periplasmic protein resulted in constitutive activation of the Rcs regulon and RcsCD-dependent phosphorylation. These findings suggest that RcsFCDB activation in response to CAMP-induced membrane disorder is a result of a change in structure or availability of RcsF to the IM signaling constituents of the Rcs phosphorelay.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Lipoproteínas/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Western Blotting , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica/genética , Lipoproteínas/genética , Reação em Cadeia da Polimerase , Ligação Proteica , Regulon/genética , Regulon/fisiologia , Salmonella enterica/efeitos dos fármacos , Salmonella enterica/genética , Salmonella enterica/metabolismo
2.
Mol Microbiol ; 69(2): 503-19, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18532985

RESUMO

The PhoQ sensor kinase is essential for Salmonella typhimurium virulence for animals, and a homologue exists in the environmental organism and opportunistic pathogen Pseudomonas aeruginosa. S. typhimurium PhoQ (ST-PhoQ) is repressed by millimolar concentrations of divalent cations and activated by antimicrobial peptides and at acidic pH. ST-PhoQ has a periplasmic Per-ARNT-Sim domain, a fold commonly employed for ligand binding. However, substrate binding is instead accomplished by an acidic patch in the periplasmic domain that interacts with the inner membrane through divalent cation bridges. The DNA sequence encoding this acidic patch is absent from Pseudomonas phoQ (PA-PhoQ). Here, we demonstrate that PA-PhoQ binds and is repressed by divalent cations, and can functionally complement a S. typhimurium phoQ-null mutant. Mutational analysis and NMR spectroscopy of the periplasmic domains of ST-PhoQ and PA-PhoQ indicate distinct mechanisms of binding divalent cation. The data are consistent with PA-PhoQ binding metal in a specific ligand-binding pocket. PA-PhoQ was partially activated by acidic pH but not by antimicrobial peptides. S. typhimurium expressing PA-PhoQ protein were attenuated for virulence in a mouse model, suggesting that the ability of Salmonella to sense host environments via antimicrobial peptides and acidic pH is an important contribution to pathogenesis.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/enzimologia , Salmonella typhimurium/química , Salmonella typhimurium/enzimologia , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Sítios de Ligação , Cátions Bivalentes/metabolismo , Análise Mutacional de DNA , Feminino , Deleção de Genes , Teste de Complementação Genética , Histidina Quinase , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Salmonelose Animal/microbiologia , Salmonella typhimurium/patogenicidade , Alinhamento de Sequência , Virulência
3.
Nat Rev Microbiol ; 3(1): 36-46, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15608698

RESUMO

Innate immune receptors recognize microorganism-specific motifs. One such receptor-ligand complex is formed between the mammalian Toll-like receptor 4 (TLR4)-MD2-CD14 complex and bacterial lipopolysaccharide (LPS). Recent research indicates that there is significant phylogenetic and individual diversity in TLR4-mediated responses. In addition, the diversity of LPS structures and the differential recognition of these structures by TLR4 have been associated with several bacterial diseases. This review will examine the hypothesis that the variability of bacterial ligands such as LPS and their innate immune receptors is an important factor in determining the outcome of infectious disease.


Assuntos
Infecções Bacterianas/metabolismo , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Antígenos de Superfície/metabolismo , Infecções Bacterianas/imunologia , Infecções Bacterianas/patologia , Proteínas de Transporte/metabolismo , Progressão da Doença , Suscetibilidade a Doenças , Humanos , Imunidade Inata , Lipídeo A/química , Lipídeo A/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/química , Antígeno 96 de Linfócito , Glicoproteínas de Membrana/química , Estrutura Molecular , Receptores de Superfície Celular/química , Transdução de Sinais , Especificidade da Espécie , Receptor 4 Toll-Like , Receptores Toll-Like
4.
J Mol Biol ; 356(5): 1193-206, 2006 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-16406409

RESUMO

Bacterial histidine kinases respond to environmental stimuli by transducing a signal from an extracytosolic sensor domain to a cytosolic catalytic domain. Among them, PhoQ promotes bacterial virulence and is tightly repressed by the divalent cations such as calcium and magnesium. We have determined the crystal structure of the PhoQ sensor domain from Salmonella typhimurium in the Ca2+-bound state, which reveals a highly negatively charged surface that is in close proximity to the inner membrane. This acidic surface binds at least three Ca2+, which mediate the PhoQ-membrane interaction. Mutagenesis analysis indicates that structural integrity at the membrane proximal region of the PhoQ sensor domain promotes metal-mediated repression. We propose that depletion or displacement of divalent cations leads to charge repulsion between PhoQ and the membrane, which initiates transmembrane signaling through a change in orientation between the PhoQ sensor domain and membrane. Therefore, both PhoQ and the membrane are required for extracytosolic sensing and transmembrane signaling.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cálcio/química , Membrana Celular/metabolismo , Magnésio/química , Transdução de Sinais/fisiologia , Ácidos/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Cristalografia por Raios X , Dimerização , Regulação Bacteriana da Expressão Gênica , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Salmonella typhimurium , Alinhamento de Sequência
5.
Mol Cell ; 26(2): 165-74, 2007 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-17466620

RESUMO

The Salmonellae PhoQ sensor kinase senses the mammalian phagosome environment to activate a transcriptional program essential for virulence. The PhoQ periplasmic domain binds divalent cations, forming bridges with inner membrane phospholipids to maintain PhoQ repression. PhoQ also binds and is activated by cationic antimicrobial peptides. In this work, PhoQ is directly activated by exposure of the sensor domain to pH 5.5. NMR spectroscopy indicates that at acidic pH, the PhoQ periplasmic domain adopts a conformation different from that in the presence of divalent cations or antimicrobial peptides. The conformation is partially simulated by mutation of histidine 157, which is part of an interaction network that distinguishes the repressed conformation. The effects of antimicrobial peptides and pH on PhoQ activity are additive. We propose a model of activation by antimicrobial peptides via disruption of the cation bridges and/or by acidification of the periplasm through destabilization of the interaction network.


Assuntos
Proteínas de Bactérias/metabolismo , Salmonella/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Cátions Bivalentes/metabolismo , Cátions Bivalentes/farmacologia , Ativação Enzimática/efeitos dos fármacos , Histidina/química , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Macrófagos/enzimologia , Macrófagos/microbiologia , Magnésio/metabolismo , Camundongos , Modelos Biológicos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Salmonella/genética , Salmonella/patogenicidade , Virulência
6.
Nat Immunol ; 7(6): 569-75, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16648853

RESUMO

Macrophages respond to Salmonella typhimurium infection via Ipaf, a NACHT-leucine-rich repeat family member that activates caspase-1 and secretion of interleukin 1beta. However, the specific microbial salmonella-derived agonist responsible for activating Ipaf is unknown. We show here that cytosolic bacterial flagellin activated caspase-1 through Ipaf but was independent of Toll-like receptor 5, a known flagellin sensor. Stimulation of the Ipaf pathway in macrophages after infection required a functional salmonella pathogenicity island 1 type III secretion system but not the flagellar type III secretion system; furthermore, Ipaf activation could be recapitulated by the introduction of purified flagellin directly into the cytoplasm. These observations raise the possibility that the salmonella pathogenicity island 1 type III secretion system cannot completely exclude 'promiscuous' secretion of flagellin and that the host capitalizes on this 'error' by activating a potent host-defense pathway.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Caspase 1/metabolismo , Flagelina/imunologia , Interleucina-1/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/patogenicidade , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Ligação ao Cálcio/genética , Citoplasma/metabolismo , Citoplasma/microbiologia , Ativação Enzimática , Flagelina/genética , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Mutantes , Mutação , Transporte Proteico , Infecções por Salmonella/enzimologia , Infecções por Salmonella/genética , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/fisiologia
7.
J Mol Evol ; 60(4): 462-74, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15883881

RESUMO

The PhoPQ two-component system acts as a transcriptional regulator that responds to Mg(2+) starvation both in Escherichia coli and Salmonella typhimurium (Garcia et al. 1996; Kato et al. 1999). By monitoring the availability of extracellular Mg(2+), this two-component system allows S. typhimurium to sense the transition from an extracellular environment to a subcellular location. Concomitantly with this transition, a set of virulence factors essential for survival in the intracellular environment is activated by the PhoPQ system (Groisman et al. 1989; Miller et al. 1989). Compared to nonpathogenic strains, such as E. coli K12, the PhoPQ regulon in pathogens must contain target genes specifically contributing to the virulence phenotype. To verify this hypothesis, we compared the composition of the PhoPQ regulon between E. coli and S. typhimurium using a combination of expression experiments and motif data. PhoPQ-dependent genes in both organisms were identified from PhoPQ-related microarray experiments. To distinguish between direct and indirect targets, we searched for the presence of the regulatory motif in the promoter region of the identified PhoPQ-dependent genes. This allowed us to reconstruct the direct PhoPQ-dependent regulons in E. coli K12 and S. typhimurium LT2. Comparison of both regulons revealed a very limited overlap of PhoPQ-dependent genes between both organisms. These results suggest that the PhoPQ system has acquired a specialized function during evolution in each of these closely related species that allows adaptation to the specificities of their lifestyles (e.g., pathogenesis in S. typhimurium).


Assuntos
Proteínas de Bactérias/genética , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Regulon , Salmonella typhimurium/genética , Sequência de Bases , Análise de Sequência com Séries de Oligonucleotídeos , Homologia de Sequência do Ácido Nucleico
8.
Cell ; 122(3): 461-72, 2005 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-16096064

RESUMO

PhoQ is a membrane bound sensor kinase important for the pathogenesis of a number of Gram-negative bacterial species. PhoQ and its cognate response regulator PhoP constitute a signal-transduction cascade that controls inducible resistance to host antimicrobial peptides. We show that enzymatic activity of Salmonella typhimurium PhoQ is directly activated by antimicrobial peptides. A highly acidic surface of the PhoQ sensor domain participates in both divalent-cation and antimicrobial-peptide binding as a first step in signal transduction across the bacterial membrane. Identification of PhoQ signaling mutants, binding studies with the PhoQ sensor domain, and structural analysis of this domain can be incorporated into a model in which antimicrobial peptides displace divalent cations from PhoQ metal binding sites to initiate signal transduction. Our findings reveal a molecular mechanism by which bacteria sense small innate immune molecules to initiate a transcriptional program that promotes bacterial virulence.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Bactérias/imunologia , Proteínas Quinases/imunologia , Salmonella typhimurium/imunologia , Adaptação Fisiológica/imunologia , Peptídeos Catiônicos Antimicrobianos/imunologia , Proteínas de Bactérias/efeitos dos fármacos , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Parasita/imunologia , Humanos , Magnésio/imunologia , Modelos Biológicos , Ligação Proteica , Conformação Proteica , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/genética , Salmonella typhimurium/enzimologia , Salmonella typhimurium/genética , Transdução de Sinais/imunologia , Fatores de Tempo , Transcrição Gênica/genética
9.
Mol Microbiol ; 51(3): 849-59, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14731284

RESUMO

Escherichia coli Hsp31 is a homodimeric protein that exhibits chaperone activity in vitro and is a representative member of a recently recognized family of heat shock proteins (Hsps). To gain insights on Hsp31 cellular function, we deleted the hchA gene from the MC4100 chromosome and combined the resulting null allele with lesions in other cytoplasmic chaperones. Although the hchA mutant only exhibited growth defects when cultivated at 48 degrees C, loss of Hsp31 had a strong deleterious effect on the ability of cells to survive and recover from transient exposure to 50 degrees C, and led to the enhanced aggregation of a subset of host proteins at this temperature. The absence of Hsp31 did not significantly affect the ability of the ClpB-DnaK-DnaJ-GrpE system to clear thermally aggregated proteins at 30 degrees C suggesting that Hsp31 does not possess disaggregase activity. Although it had no effect on the growth of groES30, Delta clpB or Delta ibpAB cells at high temperatures, the hchA deletion aggravated the temperature sensitive phenotype of dnaK756 and grpE280 mutants and led to increased aggregation in stressed dnaK756 cells. On the basis of biochemical, structural and genetic data, we propose that Hsp31 acts as a modified holding chaperone that captures early unfolding intermediates under prolonged conditions of severe stress and releases them when cells return to physiological conditions. This additional line of defence would complement the roles of DnaK-DnaJ-GrpE, ClpB and IbpB in the management of thermally induced cellular protein misfolding.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Divisão Celular , Sobrevivência Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Choque Térmico HSP40 , Temperatura Alta , Modelos Moleculares , Chaperonas Moleculares/genética
10.
J Biol Chem ; 277(30): 26886-92, 2002 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-12004064

RESUMO

Isomerization of disulfide bonds is vital for the proper folding of proteins that possess multiple disulfides. In prokaryotes, the catalytic pathway responsible for disulfide isomerization involves thioredoxin, thioredoxin reductase, and the DsbC, DsbG, and DsbD proteins. To be active as isomerases, DsbC and DsbG must be kept reduced. This task is performed by the cytoplasmic membrane protein DsbD. DsbD in turn is reduced by the cytoplasmic thioredoxin and is composed of three domains. The beta domain is membrane-embedded, whereas the alpha and gamma domains are localized to the periplasm. It had been proposed that electrons are transferred within DsbD by a succession of disulfide exchange reactions between the three domains. To test this model using biochemical methods, we purified to homogeneity different polypeptides corresponding to the alpha, beta, gamma, and betagamma domains. Using these domains, we could reconstitute a DsbD activity and, for the first time, reconstitute in vitro the electron transport pathway from NADPH and thioredoxin to DsbC and DsbG. We showed that electrons are transferred from thioredoxin to the beta domain then successively to the gamma domain, the alpha domain, and finally on to DsbC or DsbG. We also determined the redox potential of the gamma domain to be -241 mV, and that of the alpha domain was found to be -229 mV. This shows that the direction of electron flow within DsbD is thermodynamically driven.


Assuntos
Dissulfetos , Proteínas de Escherichia coli , Proteínas Periplásmicas , Catálise , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Citoplasma/metabolismo , Relação Dose-Resposta a Droga , Elétrons , Eletroforese em Gel de Poliacrilamida , Cinética , Modelos Biológicos , Oxirredução , Oxirredutases/metabolismo , Plasmídeos/metabolismo , Ligação Proteica , Isomerases de Dissulfetos de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Termodinâmica , Tiorredoxinas/metabolismo , Fatores de Tempo
11.
J Biol Chem ; 277(3): 1649-52, 2002 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-11698406

RESUMO

Disulfide bond (Dsb) formation is catalyzed in the periplasm of prokaryotes by the Dsb proteins. DsbB, a key enzyme in this process, generates disulfides de novo by using the oxidizing power of quinones. To explore the mechanism of this newly described enzymatic activity, we decided to study the ubiquinone-protein interaction and identify the ubiquinone-binding domain in DsbB by cross-linking to photoactivatable quinone analogues. When purified Escherichia coli DsbB was incubated with an azidoubiquinone derivative, 3-azido-2-methyl-5-[(3)H]methoxy-6-decyl-1,4-benzoquinone ([(3)H]azido-Q), and illuminated with long wavelength UV light, the decrease in enzymatic activity correlated with the amount of 3-azido-2-methyl-5-methoxy-6-decyl-1,4-benzoquinone (azido-Q) incorporated into the protein. One azido-Q-linked peptide with a retention time of 33.5 min was obtained by high performance liquid chromatography of the V8 digest of [(3)H]azido-Q-labeled DsbB. This peptide has a partial NH(2)-terminal amino acid sequence of NH(2)-HTMLQLY corresponding to residues 91-97. This sequence occurs in the second periplasmic domain of the inner membrane protein DsbB in a loop connecting transmembrane helices 3 and 4. We propose that the quinone-binding site is within or very near to this sequence.


Assuntos
Proteínas de Bactérias/metabolismo , Dissulfetos/metabolismo , Proteínas de Membrana/metabolismo , Ubiquinona/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Cromatografia Líquida de Alta Pressão , Escherichia coli/metabolismo , Proteínas de Membrana/química , Dados de Sequência Molecular , Marcadores de Fotoafinidade , Ligação Proteica
12.
Mol Microbiol ; 50(1): 219-30, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14507376

RESUMO

Cationic antimicrobial peptides (CAMP) represent a conserved and highly effective component of innate immunity. During infection, the Gram-negative pathogen Salmonella typhimurium induces different mechanisms of CAMP resistance that promote pathogenesis in animals. This study shows that exposure of S. typhimurium to sublethal concentrations of CAMP activates the PhoP/PhoQ and RpoS virulence regulons, while repressing the transcription of genes required for flagella synthesis and the invasion-associated type III secretion system. We further demonstrate that growth of S. typhimurium in low doses of the alpha-helical peptide C18G induces resistance to CAMP of different structural classes. Inducible resistance depends on the presence of PhoP, indicating that the PhoP/PhoQ system can sense sublethal concentrations of cationic antimicrobial peptides. Growth of S. typhimurium in the presence of CAMP also leads to RpoS-dependent protection against hydrogen peroxide. Because bacterial resistance to oxidative stress and CAMP are induced during infection of animals, CAMP may be an important signal recognized by bacteria on colonization of animal tissues.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Farmacorresistência Bacteriana , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Virulência/genética , Fosfatase Alcalina/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Eletroforese em Gel Bidimensional , Perfilação da Expressão Gênica , Genes Bacterianos , Genes Reporter/genética , Estresse Oxidativo/genética , Mapeamento de Peptídeos , Polimixinas/metabolismo , Polimixinas/farmacologia , Proteoma/análise , Regulon/fisiologia , Salmonella typhimurium/efeitos dos fármacos , Fator sigma/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA