Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Bacteriol ; 192(19): 4894-903, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20675476

ABSTRACT

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.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bacterial Outer Membrane Proteins/metabolism , Lipoproteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blotting, Western , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/genetics , Lipoproteins/genetics , Polymerase Chain Reaction , Protein Binding , Regulon/genetics , Regulon/physiology , Salmonella enterica/drug effects , Salmonella enterica/genetics , Salmonella enterica/metabolism
2.
Mol Microbiol ; 69(2): 503-19, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18532985

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/enzymology , Salmonella typhimurium/chemistry , Salmonella typhimurium/enzymology , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Binding Sites , Cations, Divalent/metabolism , DNA Mutational Analysis , Female , Gene Deletion , Genetic Complementation Test , Histidine Kinase , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Kinases/chemistry , Protein Kinases/metabolism , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/pathogenicity , Sequence Alignment , Virulence
3.
Mol Cell ; 26(2): 165-74, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17466620

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Salmonella/metabolism , Animals , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cations, Divalent/metabolism , Cations, Divalent/pharmacology , Enzyme Activation/drug effects , Histidine/chemistry , Hydrogen-Ion Concentration , In Vitro Techniques , Macrophages/enzymology , Macrophages/microbiology , Magnesium/metabolism , Mice , Models, Biological , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Salmonella/genetics , Salmonella/pathogenicity , Virulence
4.
Nat Immunol ; 7(6): 569-75, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16648853

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Calcium-Binding Proteins/physiology , Caspase 1/metabolism , Flagellin/immunology , Interleukin-1/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/pathogenicity , Animals , Apoptosis Regulatory Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Calcium-Binding Proteins/genetics , Cytoplasm/metabolism , Cytoplasm/microbiology , Enzyme Activation , Flagellin/genetics , Macrophages/enzymology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Mutant Strains , Mutation , Protein Transport , Salmonella Infections/enzymology , Salmonella Infections/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/physiology
5.
J Mol Biol ; 356(5): 1193-206, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16406409

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Calcium/chemistry , Cell Membrane/metabolism , Magnesium/chemistry , Signal Transduction/physiology , Acids/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Crystallography, X-Ray , Dimerization , Gene Expression Regulation, Bacterial , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Salmonella typhimurium , Sequence Alignment
6.
Cell ; 122(3): 461-72, 2005 Aug 12.
Article in English | MEDLINE | ID: mdl-16096064

ABSTRACT

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.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/immunology , Protein Kinases/immunology , Salmonella typhimurium/immunology , Adaptation, Physiological/immunology , Antimicrobial Cationic Peptides/immunology , Bacterial Proteins/drug effects , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Host-Parasite Interactions/immunology , Humans , Magnesium/immunology , Models, Biological , Protein Binding , Protein Conformation , Protein Kinases/drug effects , Protein Kinases/genetics , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Signal Transduction/immunology , Time Factors , Transcription, Genetic/genetics
7.
J Mol Evol ; 60(4): 462-74, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15883881

ABSTRACT

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).


Subject(s)
Bacterial Proteins/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Regulon , Salmonella typhimurium/genetics , Base Sequence , Oligonucleotide Array Sequence Analysis , Sequence Homology, Nucleic Acid
8.
Nat Rev Microbiol ; 3(1): 36-46, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15608698

ABSTRACT

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.


Subject(s)
Bacterial Infections/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, Surface/metabolism , Bacterial Infections/immunology , Bacterial Infections/pathology , Carrier Proteins/metabolism , Disease Progression , Disease Susceptibility , Humans , Immunity, Innate , Lipid A/chemistry , Lipid A/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/chemistry , Lymphocyte Antigen 96 , Membrane Glycoproteins/chemistry , Molecular Structure , Receptors, Cell Surface/chemistry , Signal Transduction , Species Specificity , Toll-Like Receptor 4 , Toll-Like Receptors
9.
Mol Microbiol ; 51(3): 849-59, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14731284

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Folding , Cell Division , Cell Survival , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , HSP40 Heat-Shock Proteins , Hot Temperature , Models, Molecular , Molecular Chaperones/genetics
10.
Mol Microbiol ; 50(1): 219-30, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14507376

ABSTRACT

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.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Virulence/genetics , Alkaline Phosphatase/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Genes, Bacterial , Genes, Reporter/genetics , Oxidative Stress/genetics , Peptide Mapping , Polymyxins/metabolism , Polymyxins/pharmacology , Proteome/analysis , Regulon/physiology , Salmonella typhimurium/drug effects , Sigma Factor/genetics , Signal Transduction/genetics , Signal Transduction/physiology , beta-Galactosidase/metabolism
11.
J Biol Chem ; 277(30): 26886-92, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12004064

ABSTRACT

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.


Subject(s)
Disulfides , Escherichia coli Proteins , Periplasmic Proteins , Catalysis , Chromatography, Gel , Chromatography, High Pressure Liquid , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Electrons , Electrophoresis, Polyacrylamide Gel , Kinetics , Models, Biological , Oxidation-Reduction , Oxidoreductases/metabolism , Plasmids/metabolism , Protein Binding , Protein Disulfide-Isomerases/metabolism , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Thermodynamics , Thioredoxins/metabolism , Time Factors
12.
J Biol Chem ; 277(3): 1649-52, 2002 Jan 18.
Article in English | MEDLINE | ID: mdl-11698406

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Disulfides/metabolism , Membrane Proteins/metabolism , Ubiquinone/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Chromatography, High Pressure Liquid , Escherichia coli/metabolism , Membrane Proteins/chemistry , Molecular Sequence Data , Photoaffinity Labels , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...