Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-28607020

RESUMEN

There has been major interest by the scientific community in antivirulence approaches against bacterial infections. However, partly due to a lack of viable lead compounds, antivirulence therapeutics have yet to reach the clinic. Here we investigate the development of an antivirulence lead targeting quorum sensing signal biosynthesis, a process that is conserved in Gram-positive bacterial pathogens. Some preliminary studies suggest that the small molecule ambuic acid is a signal biosynthesis inhibitor. To confirm this, we constructed a methicillin-resistant Staphylococcus aureus (MRSA) strain that decouples autoinducing peptide (AIP) production from regulation and demonstrate that AIP production is inhibited in this mutant. Quantitative mass spectrometric measurements show that ambuic acid inhibits signal biosynthesis (50% inhibitory concentration [IC50] of 2.5 ± 0.1 µM) against a clinically relevant USA300 MRSA strain. Quantitative real-time PCR confirms that this compound selectively targets the quorum sensing regulon. We show that a 5-µg dose of ambuic acid reduces MRSA-induced abscess formation in a mouse model and verify its quorum sensing inhibitory activity in vivo Finally, we employed mass spectrometry to identify or confirm the structure of quorum sensing signaling peptides in three strains each of S. aureus and Staphylococcus epidermidis and single strains of Enterococcus faecalis, Listeria monocytogenes, Staphylococcus saprophyticus, and Staphylococcus lugdunensis By measuring AIP production by these strains, we show that ambuic acid possesses broad-spectrum efficacy against multiple Gram-positive bacterial pathogens but does not inhibit quorum sensing in some commensal bacteria. Collectively, these findings demonstrate the promise of ambuic acid as a lead for the development of antivirulence therapeutics.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/biosíntesis , Ciclohexanonas/farmacología , Bacterias Grampositivas/efectos de los fármacos , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Péptidos Cíclicos/biosíntesis , Animales , Antibacterianos/química , Ciclohexanonas/química , Modelos Animales de Enfermedad , Bacterias Grampositivas/genética , Bacterias Grampositivas/patogenicidad , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Ratones Endogámicos BALB C , Percepción de Quorum/efectos de los fármacos , Transducción de Señal , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Factores de Virulencia
2.
Proc Natl Acad Sci U S A ; 111(19): E2037-45, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24782537

RESUMEN

Two-component systems (TCSs) are highly conserved across bacteria and are used to rapidly sense and respond to changing environmental conditions. The human pathogen Staphylococcus aureus uses the S. aureus exoprotein expression (sae) TCS to sense host signals and activate transcription of virulence factors essential to pathogenesis. Despite its importance, the mechanism by which the histidine kinase SaeS recognizes specific host stimuli is unknown. After mutagenizing the predicted extracellular loop of SaeS, we discovered one methionine residue (M31) was essential for the ability of S. aureus to transcribe sae target genes, including hla, lukAB/lukGH, and hlgA. This single M31A mutation also significantly reduced cytotoxicity in human neutrophils to levels observed in cells following interaction with ΔsaeS. Another important discovery was that mutation of two aromatic anchor residues (W32A and F33A) disrupted the normal basal signaling of SaeS in the absence of inducing signals, yet both mutant kinases had appropriate activation of effector genes following exposure to neutrophils. Although the transcriptional profile of aromatic mutation W32A was consistent with that of WT in response to human α-defensin 1, mutant kinase F33A did not properly transcribe the γ-toxin genes in response to this stimulus. Taken together, our results provide molecular evidence for how SaeS recognizes host signals and triggers activation of select virulence factors to facilitate evasion of innate immunity. These findings have important implications for signal transduction in prokaryotes and eukaryotes due to conservation of aromatic anchor residues across both of these domains and the important role they play in sensor protein structure and function.


Asunto(s)
Neutrófilos/microbiología , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Staphylococcus aureus/enzimología , Staphylococcus aureus/genética , Secuencia de Aminoácidos , Proteínas Bacterianas , Membrana Celular/metabolismo , Activación Enzimática , Inmunidad Innata/inmunología , Datos de Secuencia Molecular , Neutrófilos/inmunología , Proteínas Quinasas/química , Estructura Terciaria de Proteína , Transducción de Señal/inmunología , Staphylococcus aureus/patogenicidad , Virulencia
3.
EMBO J ; 31(17): 3607-19, 2012 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-22850671

RESUMEN

The CXC chemokine receptor 2 (CXCR2) on neutrophils, which recognizes chemokines produced at the site of infection, plays an important role in antimicrobial host defenses such as neutrophil activation and chemotaxis. Staphylococcus aureus is a successful human pathogen secreting a number of proteolytic enzymes, but their influence on the host immune system is not well understood. Here, we identify the cysteine protease Staphopain A as a chemokine receptor blocker. Neutrophils treated with Staphopain A are unresponsive to activation by all unique CXCR2 chemokines due to cleavage of the N-terminal domain, which can be neutralized by specific protease inhibitors. Moreover, Staphopain A inhibits neutrophil migration towards CXCR2 chemokines. By comparing a methicillin-resistant S. aureus (MRSA) strain with an isogenic Staphopain A mutant, we demonstrate that Staphopain A is the only secreted protease with activity towards CXCR2. Although the inability to cleave murine CXCR2 limits in-vivo studies, our data indicate that Staphopain A is an important immunomodulatory protein that blocks neutrophil recruitment by specific cleavage of the N-terminal domain of human CXCR2.


Asunto(s)
Proteínas Bacterianas/inmunología , Cisteína Endopeptidasas/inmunología , Neutrófilos/inmunología , Receptores de Interleucina-8B/inmunología , Animales , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Activación Neutrófila/inmunología , Infiltración Neutrófila/inmunología , Receptores de Interleucina-8B/antagonistas & inhibidores , Células U937
4.
PLoS Pathog ; 9(12): e1003819, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367264

RESUMEN

Staphylococcus aureus is a prominent bacterial pathogen that is known to agglutinate in the presence of human plasma to form stable clumps. There is increasing evidence that agglutination aids S. aureus pathogenesis, but the mechanisms of this process remain to be fully elucidated. To better define this process, we developed both tube based and flow cytometry methods to monitor clumping in the presence of extracellular matrix proteins. We discovered that the ArlRS two-component system regulates the agglutination mechanism during exposure to human plasma or fibrinogen. Using divergent S. aureus strains, we demonstrated that arlRS mutants are unable to agglutinate, and this phenotype can be complemented. We found that the ebh gene, encoding the Giant Staphylococcal Surface Protein (GSSP), was up-regulated in an arlRS mutant. By introducing an ebh complete deletion into an arlRS mutant, agglutination was restored. To assess whether GSSP is the primary effector, a constitutive promoter was inserted upstream of the ebh gene on the chromosome in a wildtype strain, which prevented clump formation and demonstrated that GSSP has a negative impact on the agglutination mechanism. Due to the parallels of agglutination with infective endocarditis development, we assessed the phenotype of an arlRS mutant in a rabbit combined model of sepsis and endocarditis. In this model the arlRS mutant displayed a large defect in vegetation formation and pathogenesis, and this phenotype was partially restored by removing GSSP. Altogether, we have discovered that the ArlRS system controls a novel mechanism through which S. aureus regulates agglutination and pathogenesis.


Asunto(s)
Proteínas Bacterianas/fisiología , Staphylococcus aureus/fisiología , Staphylococcus aureus/patogenicidad , Aglutinación/genética , Animales , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Endocarditis Bacteriana/genética , Endocarditis Bacteriana/microbiología , Femenino , Fibrinógeno/fisiología , Regulación Bacteriana de la Expresión Génica , Humanos , Masculino , Organismos Modificados Genéticamente , Conejos , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética
5.
J Biol Chem ; 287(17): 13889-98, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22371493

RESUMEN

Staphylococcus aureus causes a wide range of human disease ranging from localized skin and soft tissue infections to potentially lethal systemic infections. S. aureus has the biosynthetic ability to generate numerous virulence factors that assist in circumventing the innate immune system during disease pathogenesis. Recent studies have uncovered a set of extracellular peptides produced by community-associated methicillin-resistant S. aureus (CA-MRSA) with homology to the phenol-soluble modulins (PSMs) from Staphylococcus epidermidis. CA-MRSA PSMs contribute to skin infection and recruit and lyse neutrophils, and truncated versions of these peptides possess antimicrobial activity. In this study, novel CA-MRSA PSM derivatives were discovered by the use of microbial imaging mass spectrometry. The novel PSM derivatives are compared with their parent full-length peptides for changes in hemolytic, cytolytic, and neutrophil-stimulating activity. A potential contribution of the major S. aureus secreted protease aureolysin in processing PSMs is demonstrated. Finally, we show that PSM processing occurs in multiple CA-MRSA strains by structural confirmation of additional novel derivatives. This work demonstrates that IMS can serve as a useful tool to go beyond genome predictions and expand our understanding of the important family of small peptide virulence factors.


Asunto(s)
Espectrometría de Masas/métodos , Staphylococcus aureus Resistente a Meticilina/metabolismo , Fenol/química , Secuencia de Aminoácidos , Animales , Antiinfecciosos/farmacología , Infecciones Bacterianas/metabolismo , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Eritrocitos/citología , Hemólisis , Humanos , Inmunosupresores/farmacología , Metaloendopeptidasas/química , Ratones , Datos de Secuencia Molecular , Neutrófilos/citología , Neutrófilos/metabolismo , Homología de Secuencia de Aminoácido , Ovinos , Piel/metabolismo , Piel/microbiología , Infecciones Cutáneas Estafilocócicas/microbiología , Factores de Virulencia/química
6.
Infect Immun ; 81(9): 3227-38, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23798534

RESUMEN

Staphylococcus aureus is a known cause of chronic biofilm infections that can reside on medical implants or host tissue. Recent studies have demonstrated an important role for proteinaceous material in the biofilm structure. The S. aureus genome encodes many secreted proteases, and there is growing evidence that these enzymes have self-cleavage properties that alter biofilm integrity. However, the specific contribution of each protease and mechanism of biofilm modulation is not clear. To address this issue, we utilized a sigma factor B (ΔsigB) mutant where protease activity results in a biofilm-negative phenotype, thereby creating a condition where the protease(s) responsible for the phenotype could be identified. Using a plasma-coated microtiter assay, biofilm formation was restored to the ΔsigB mutant through the addition of the cysteine protease inhibitor E-64 or by using Staphostatin inhibitors that specifically target the extracellular cysteine proteases SspB and ScpA (called Staphopains). Through construction of gene deletion mutants, we determined that an sspB scpA double mutant restored ΔsigB biofilm formation, and this recovery could be replicated in plasma-coated flow cell biofilms. Staphopain levels were also found to be decreased under biofilm-forming conditions, possibly allowing biofilm establishment. The treatment of S. aureus biofilms with purified SspB or ScpA enzyme inhibited their formation, and ScpA was also able to disperse an established biofilm. The antibiofilm properties of ScpA were conserved across S. aureus strain lineages. These findings suggest an underappreciated role of the SspB and ScpA cysteine proteases in modulating S. aureus biofilm architecture.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteasas de Cisteína/genética , Proteasas de Cisteína/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Mutación/genética , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
7.
J Immunol ; 186(11): 6445-53, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21502375

RESUMEN

Complement is one of the first host defense barriers against bacteria. Activated complement attracts neutrophils to the site of infection and opsonizes bacteria to facilitate phagocytosis. The human pathogen Staphylococcus aureus has successfully developed ways to evade the complement system, for example by secretion of specific complement inhibitors. However, the influence of S. aureus proteases on the host complement system is still poorly understood. In this study, we identify the metalloprotease aureolysin as a potent complement inhibitor. Aureolysin effectively inhibits phagocytosis and killing of bacteria by neutrophils. Furthermore, we show that aureolysin inhibits the deposition of C3b on bacterial surfaces and the release of the chemoattractant C5a. Cleavage analyses show that aureolysin cleaves the central complement protein C3. Strikingly, there was a clear difference between the cleavages of C3 in serum versus purified conditions. Aureolysin cleaves purified C3 specifically in the α-chain, close to the C3 convertase cleavage site, yielding active C3a and C3b. However, in serum we observe that the aureolysin-generated C3b is further degraded by host factors. We pinpointed these factors to be factor H and factor I. Using an aureolysin mutant in S. aureus USA300, we show that aureolysin is essential and sufficient for C3 cleavage by bacterial supernatant. In short, aureolysin acts in synergy with host regulators to inactivate C3 thereby effectively dampening the host immune response.


Asunto(s)
Proteínas Bacterianas/inmunología , Complemento C3/inmunología , Evasión Inmune/inmunología , Metaloendopeptidasas/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Western Blotting , Activación de Complemento/inmunología , Complemento C3/metabolismo , Convertasas de Complemento C3-C5/inmunología , Convertasas de Complemento C3-C5/metabolismo , Complemento C3a/inmunología , Complemento C3a/metabolismo , Complemento C3b/inmunología , Complemento C3b/metabolismo , Citotoxicidad Inmunológica/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Mutación , Neutrófilos/inmunología , Fagocitosis/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/enzimología , Staphylococcus aureus/fisiología , Células U937
8.
J Bacteriol ; 193(19): 5279-91, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21784926

RESUMEN

Lipoteichoic acid (LTA) is a crucial cell envelope component in Gram-positive bacteria. In Staphylococcus aureus, the polyglycerolphosphate LTA molecule is synthesized by LtaS, a membrane-embedded enzyme with five N-terminal transmembrane helices (5TM domain) that are connected via a linker region to the C-terminal extracellular enzymatic domain (eLtaS). The LtaS enzyme is processed during bacterial growth, and the eLtaS domain is released from the bacterial membrane. Here we provide experimental evidence that the proteolytic cleavage following residues 215Ala-Leu-Ala217 is performed by the essential S. aureus signal peptidase SpsB, as depletion of spsB results in reduced LtaS processing. In addition, the introduction of a proline residue at the +1 position with respect to the cleavage site, a substitution known to inhibit signal peptidase-dependent cleavage, abolished LtaS processing at this site. It was further shown that the 5TM domain is crucial for enzyme function. The observation that the construction of hybrid proteins between two functional LtaS-type enzymes resulted in the production of proteins unable to synthesize LTA suggests that specific interactions between the 5TM and eLtaS domains are required for function. No enzyme activity was detected upon expression of the 5TM and eLtaS domains as separate fragments, indicating that the two domains cannot assemble postsynthesis to form a functional enzyme. Taken together, our data suggest that only the full-length LtaS enzyme is active in the LTA synthesis pathway and that the proteolytic cleavage step is used as a mechanism to irreversibly inactivate the enzyme.


Asunto(s)
Proteínas Bacterianas/metabolismo , Lipopolisacáridos/biosíntesis , Staphylococcus aureus/metabolismo , Ácidos Teicoicos/biosíntesis , Proteínas Bacterianas/genética , Western Blotting , Cromatografía en Capa Delgada , Glicerofosfatos/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Lipopolisacáridos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Modelos Biológicos , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Staphylococcus aureus/genética , Ácidos Teicoicos/metabolismo
9.
PLoS One ; 9(4): e95574, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24752186

RESUMEN

Staphylococcus aureus is a prominent bacterial pathogen that causes a diverse range of acute and chronic infections. Recently, it has been demonstrated that the secreted nuclease (Nuc) enzyme is a virulence factor in multiple models of infection, and in vivo expression of nuc has facilitated the development of an infection imaging approach based on Nuc-activatable probes. Interestingly, S. aureus strains encode a second nuclease (Nuc2) that has received limited attention. With the growing interest in bacterial nucleases, we sought to characterize Nuc2 in more detail through localization, expression, and biochemical studies. Fluorescence microscopy and alkaline phosphatase localization approaches using Nuc2-GFP and Nuc2-PhoA fusions, respectively, demonstrated that Nuc2 is membrane bound with the C-terminus facing the extracellular environment, indicating it is a signal-anchored Type II membrane protein. Nuc2 enzyme activity was detectable on the S. aureus cell surface using a fluorescence resonance energy transfer (FRET) assay, and in time courses, both nuc2 transcription and enzyme activity peaked in early logarithmic growth and declined in stationary phase. Using a mouse model of S. aureus pyomyositis, Nuc2 activity was detected with activatable probes in vivo in nuc mutant strains, demonstrating that Nuc2 is produced during infections. To assess Nuc2 biochemical properties, the protein was purified and found to cleave both single- and double-stranded DNA, and it exhibited thermostability and calcium dependence, paralleling the properties of Nuc. Purified Nuc2 prevented biofilm formation in vitro and modestly decreased biomass in dispersal experiments. Altogether, our findings confirm that S. aureus encodes a second, surface-attached and functional DNase that is expressed during infections and displays similar biochemical properties to the secreted Nuc enzyme.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Endonucleasas/metabolismo , Espacio Extracelular/enzimología , Staphylococcus aureus/enzimología , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Biopelículas , Endonucleasas/química , Endonucleasas/aislamiento & purificación , Ratones , Transporte de Proteínas , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/fisiología , Homología Estructural de Proteína
10.
PLoS One ; 6(11): e26714, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22096493

RESUMEN

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is an emerging contributor to biofilm-related infections. We recently reported that strains lacking sigma factor B (sigB) in the USA300 lineage of CA-MRSA are unable to develop a biofilm. Interestingly, when spent media from a USA300 sigB mutant was incubated with other S. aureus strains, biofilm formation was inhibited. Following fractionation and mass spectrometry analysis, the major anti-biofilm factor identified in the spent media was secreted thermonuclease (Nuc). Considering reports that extracellular DNA (eDNA) is an important component of the biofilm matrix, we investigated the regulation and role of Nuc in USA300. The expression of the nuc gene was increased in a sigB mutant, repressed by glucose supplementation, and was unaffected by the agr quorum-sensing system. A FRET assay for Nuc activity was developed and confirmed the regulatory results. A USA300 nuc mutant was constructed and displayed an enhanced biofilm-forming capacity, and the nuc mutant also accumulated more high molecular weight eDNA than the WT and regulatory mutant strains. Inactivation of nuc in the USA300 sigB mutant background partially repaired the sigB biofilm-negative phenotype, suggesting that nuc expression contributes to the inability of the mutant to form biofilm. To test the generality of the nuc mutant biofilm phenotypes, the mutation was introduced into other S. aureus genetic backgrounds and similar increases in biofilm formation were observed. Finally, using multiple S. aureus strains and regulatory mutants, an inverse correlation between Nuc activity and biofilm formation was demonstrated. Altogether, our findings confirm the important role for eDNA in the S. aureus biofilm matrix and indicates Nuc is a regulator of biofilm formation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Staphylococcus aureus Resistente a Meticilina/enzimología , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Nucleasa Microcócica/metabolismo , Proteínas Bacterianas/genética , Staphylococcus aureus Resistente a Meticilina/genética , Nucleasa Microcócica/genética , Mutación , Factor sigma/genética , Factor sigma/metabolismo
11.
J Orthop Res ; 28(1): 55-61, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19610092

RESUMEN

Orthopedic implant-related bacterial infections are associated with high morbidity that may lead to limb amputation and exert significant financial burden on the healthcare system. Staphylococcus aureus is a dominant cause of these infections, and increased incidence of community-associated methicillin-resistant S. aureus (CA-MRSA) is being reported. The ability of S. aureus to attach to the foreign body surface and develop a biofilm is an important determinant of resistance to antibiotic prophylaxis. To gain insight on CA-MRSA biofilm properties, USA300 biofilm maturation and dispersal was examined, and these biofilms were found to exhibit pronounced, quorum-sensing mediated dispersal from a glass surface. For comparison of biofilm maturation on different surface chemistries, USA300 biofilm growth was examined on glass, polycarbonate, and titanium, and minimal differences were apparent in thickness, total biomass, and substratum coverage. Importantly, USA300 biofilms grown on titanium possessed a functional dispersal mechanism, and the dispersed cells regained susceptibility to rifampicin and levofloxacin treatment. The titanium biofilms were also sensitive to proteinase K and DNaseI, suggesting the matrix is composed of proteinaceous material and extracellular DNA. These studies provide new insights on the properties of CA-MRSA biofilms on implant materials, and indicate that quorum-sensing dispersion could be an effective therapeutic strategy.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Infecciones Comunitarias Adquiridas/microbiología , Fijadores Internos/microbiología , Staphylococcus aureus Resistente a Meticilina/fisiología , Percepción de Quorum , Infecciones Estafilocócicas/microbiología , Artroplastia de Reemplazo/instrumentación , Materiales Biocompatibles , Infecciones Comunitarias Adquiridas/prevención & control , Humanos , Modelos Biológicos , Diseño de Prótesis , Infecciones Relacionadas con Prótesis/microbiología , Infecciones Relacionadas con Prótesis/prevención & control , Infecciones Estafilocócicas/prevención & control , Propiedades de Superficie , Titanio
12.
J Microbiol Methods ; 77(3): 251-60, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19264102

RESUMEN

With the emergence of Staphylococcus aureus as a prominent pathogen in community and healthcare settings, there is a growing need for effective reporter tools to facilitate physiology and pathogenesis studies. Fluorescent proteins are ideal as reporters for their convenience in monitoring gene expression, performing host interaction studies, and monitoring biofilm growth. We have developed a suite of fluorescent reporter plasmids for labeling S. aureus cells. These plasmids encode either green fluorescent protein (GFP) or higher wavelength reporter variants for yellow (YFP) and red (mCherry) labeling. The reporters were placed under control of characterized promoters to enable constitutive or inducible expression. Additionally, plasmids were assembled with fluorescent reporters under control of the agr quorum-sensing and sigma factor B promoters, and the fluorescent response with wildtype and relevant mutant strains was characterized. Interestingly, reporter expression displayed a strong dependence on ribosome binding site (RBS) sequence, with the superoxide dismutase RBS displaying the strongest expression kinetics of the sequences examined. To test the robustness of the reporter plasmids, cell imaging was performed with fluorescence microscopy and cell populations were separated using florescence-activated cell sorting (FACS), demonstrating the possibilities of simultaneous monitoring of multiple S. aureus properties. Finally, a constitutive YFP reporter displayed stable, robust labeling of biofilm growth in a flow-cell apparatus. This toolbox of fluorescent reporter plasmids will facilitate cell labeling for a variety of different experimental applications.


Asunto(s)
Genes Reporteros , Proteínas Luminiscentes/metabolismo , Staphylococcus aureus/citología , Biopelículas , Citometría de Flujo , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Plásmidos/genética , Staphylococcus aureus/genética , Staphylococcus aureus/fisiología
13.
J Biol Chem ; 283(4): 1962-73, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-18048366

RESUMEN

The yeast Sln1p sensor kinase is best known as an osmosensor involved in the regulation of the hyperosmolarity glycerol mitogen-activated protein kinase cascade. Down-regulation of Sln1 kinase activity occurs under hypertonic conditions and leads to phosphorylation of the Hog1p mitogen-activated protein kinase and increased osmotic stress-response gene expression. Conditions leading to kinase up-regulation include osmotic imbalance caused by glycerol retention in the glycerol channel mutant, fps1 (Tao, W., Deschenes, R. J., and Fassler, J. S. (1999) J. Biol. Chem. 274, 360-367). The hypothesis that Sln1p kinase activity is responsive to turgor was first suggested by the increased Sln1p kinase activity in mutants lacking Fps1p in which glycerol accumulation leads to water uptake. Also consistent with the turgor hypothesis is the observation that reduced turgor caused by treatment of cells with nystatin, a drug that increases membrane permeability and causes cell shrinkage, reduced Sln1p kinase activity (Tao, W., Deschenes, R. J., and Fassler, J. S. (1999) J. Biol. Chem. 274, 360-367; Reiser, V., Raitt, D. C., and Saito, H. (2003) J. Cell Biol. 161, 1035-1040). The turgor hypothesis is revisited here in the context of the identification and characterization of the cell wall gene, CCW12, as a determinant of Sln1p activity. Results of this analysis suggest that the activity of the plasma membrane localized Sln1p is affected by the presence or absence of specific outer cell wall proteins and that this effect is independent of turgor.


Asunto(s)
Pared Celular/metabolismo , Glicerol/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Antifúngicos/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/fisiología , Pared Celular/genética , Glicoproteínas , Péptidos y Proteínas de Señalización Intracelular , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nistatina/farmacología , Ósmosis/fisiología , Proteínas Quinasas/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Equilibrio Hidroelectrolítico/fisiología
14.
Appl Environ Microbiol ; 73(19): 6036-44, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17693565

RESUMEN

The Agr quorum-sensing system of Staphylococcus aureus modulates the expression of virulence factors in response to autoinducing peptides (AIPs). The peptides are seven to nine residues in length and have the C-terminal five residues constrained in a thiolactone ring. We have developed a new method to generate AIP structures using an engineered DnaB mini-intein from Synechocystis sp. strain PCC6803. In the method, an oligonucleotide encoding the AIP is ligated to the intein and the fusion protein is expressed and purified by affinity chromatography. To produce the correct AIP structure, intein splicing is interrupted, allowing the cysteine side chain to catalyze thiolactone ring formation and release AIP from the resin. The technique is simple and robust, and we have successfully produced the three main classes of AIPs using the intein system. The intein-generated AIPs possessed the correct thiolactone ring modification based on biochemical analysis, and, importantly, all the samples were bioactive against S. aureus. The AIP activity was confirmed through Agr interference and activation profiling with developed S. aureus reporter strains. The simplicity of the method, benefits of DNA encoding, and scalable nature enable the production of S. aureus AIPs for many biological applications.


Asunto(s)
Proteínas Bacterianas/biosíntesis , AdnB Helicasas/metabolismo , Inteínas/fisiología , Péptidos/metabolismo , Staphylococcus aureus/metabolismo , Synechocystis/química , Regulación Bacteriana de la Expresión Génica , Inteínas/genética , Péptidos Cíclicos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/genética
15.
Mol Microbiol ; 43(2): 459-73, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11985722

RESUMEN

The yeast histidine kinase, Sln1p, is a plasma membrane-associated osmosensor that regulates the activity of the osmotic stress MAP kinase pathway. Changes in the osmotic environment of the cell influence the autokinase activity of the cytoplasmic kinase domain of Sln1p. Neither the nature of the stimulus, the mechanism by which the osmotic signal is transduced nor the manner in which the kinase is regulated is currently clear. We have identified several mutations located in the linker region of the Sln1 kinase (just upstream of the kinase domain) that cause hyperactivity of the Sln1 kinase. This region of histidine kinases is largely uncharacterized, but its location between the transmembrane domains and the cytoplasmic kinase domain suggests that it may have a potential role in signal transduction. In this study, we have investigated the Sln1 linker region in order to understand its function in signal transduction and regulation of Sln1 kinase activity. Our results indicate that the linker region forms a coiled-coil structure and suggest a mechanism by which alterations induced by osmotic stress influence kinase activity by altering the alignment of the phospho-accepting histidine with respect to the catalytic domain of the kinase.


Asunto(s)
Proteínas Fúngicas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/enzimología , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Citoplasma , Proteínas Fúngicas/genética , Péptidos y Proteínas de Señalización Intracelular , Leucina Zippers , Datos de Secuencia Molecular , Mutagénesis , Fenotipo , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA