Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 10(6): e1004174, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24945495

RESUMEN

Bacterial signaling systems are prime drug targets for combating the global health threat of antibiotic resistant bacterial infections including those caused by Staphylococcus aureus. S. aureus is the primary cause of acute bacterial skin and soft tissue infections (SSTIs) and the quorum sensing operon agr is causally associated with these. Whether efficacious chemical inhibitors of agr signaling can be developed that promote host defense against SSTIs while sparing the normal microbiota of the skin is unknown. In a high throughput screen, we identified a small molecule inhibitor (SMI), savirin (S. aureus virulence inhibitor) that disrupted agr-mediated quorum sensing in this pathogen but not in the important skin commensal Staphylococcus epidermidis. Mechanistic studies employing electrophoretic mobility shift assays and a novel AgrA activation reporter strain revealed the transcriptional regulator AgrA as the target of inhibition within the pathogen, preventing virulence gene upregulation. Consistent with its minimal impact on exponential phase growth, including skin microbiota members, savirin did not provoke stress responses or membrane dysfunction induced by conventional antibiotics as determined by transcriptional profiling and membrane potential and integrity studies. Importantly, savirin was efficacious in two murine skin infection models, abating tissue injury and selectively promoting clearance of agr+ but not Δagr bacteria when administered at the time of infection or delayed until maximal abscess development. The mechanism of enhanced host defense involved in part enhanced intracellular killing of agr+ but not Δagr in macrophages and by low pH. Notably, resistance or tolerance to savirin inhibition of agr was not observed after multiple passages either in vivo or in vitro where under the same conditions resistance to growth inhibition was induced after passage with conventional antibiotics. Therefore, chemical inhibitors can selectively target AgrA in S. aureus to promote host defense while sparing agr signaling in S. epidermidis and limiting resistance development.


Asunto(s)
Antibacterianos/uso terapéutico , Proteínas Bacterianas/antagonistas & inhibidores , Inmunidad Innata/efectos de los fármacos , Quinazolinonas/uso terapéutico , Percepción de Quorum/efectos de los fármacos , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Transactivadores/antagonistas & inhibidores , Triazoles/uso terapéutico , Animales , Antibacterianos/efectos adversos , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular Transformada , Descubrimiento de Drogas , Genes Reporteros/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Ratones Pelados , Ratones Noqueados , Conformación Molecular , Simulación del Acoplamiento Molecular , Terapia Molecular Dirigida/efectos adversos , Mutación , Fagocitosis/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Quinazolinonas/efectos adversos , Quinazolinonas/química , Quinazolinonas/farmacología , Piel/efectos de los fármacos , Piel/microbiología , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/microbiología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/inmunología , Staphylococcus aureus/fisiología , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/crecimiento & desarrollo , Staphylococcus epidermidis/inmunología , Staphylococcus epidermidis/fisiología , Transactivadores/química , Transactivadores/genética , Transactivadores/metabolismo , Triazoles/efectos adversos , Triazoles/química , Triazoles/farmacología
2.
PLoS Pathog ; 9(2): e1003166, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23459693

RESUMEN

Staphylococcus aureus contains an autoinducing quorum-sensing system encoded within the agr operon that coordinates expression of virulence genes required for invasive infection. Allelic variation within agr has generated four agr specific groups, agr I-IV, each of which secretes a distinct autoinducing peptide pheromone (AIP1-4) that drives agr signaling. Because agr signaling mediates a phenotypic change in this pathogen from an adherent colonizing phenotype to one associated with considerable tissue injury and invasiveness, we postulated that a significant contribution to host defense against tissue damaging and invasive infections could be provided by innate immune mechanisms that antagonize agr signaling. We determined whether two host defense factors that inhibit AIP1-induced agrI signaling, Nox2 and apolipoprotein B (apoB), also contribute to innate control of AIP3-induced agrIII signaling. We hypothesized that apoB and Nox2 would function differently against AIP3, which differs from AIP1 in amino acid sequence and length. Here we show that unlike AIP1, AIP3 is resistant to direct oxidant inactivation by Nox2 characteristic ROS. Rather, the contribution of Nox2 to defense against agrIII signaling is through oxidation of LDL. ApoB in the context of oxLDL, and not LDL, provides optimal host defense against S. aureus agrIII infection by binding the secreted signaling peptide, AIP3, and preventing expression of the agr-driven virulence factors which mediate invasive infection. ApoB within the context of oxLDL also binds AIP 1-4 and oxLDL antagonizes agr signaling by all four agr alleles. Our results suggest that Nox2-mediated oxidation of LDL facilitates a conformational change in apoB to one sufficient for binding and sequestration of all four AIPs, demonstrating the interdependence of apoB and Nox2 in host defense against agr signaling. These data reveal a novel role for oxLDL in host defense against S. aureus quorum-sensing signaling.


Asunto(s)
Apolipoproteínas B/metabolismo , Proteínas Bacterianas/metabolismo , Glicoproteínas de Membrana/fisiología , NADPH Oxidasas/fisiología , Percepción de Quorum/fisiología , Receptores de LDL/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Transactivadores/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Regulación Bacteriana de la Expresión Génica , Inmunidad Innata , Inmunoensayo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2 , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/patología , Resonancia por Plasmón de Superficie
3.
Cell Host Microbe ; 4(6): 555-66, 2008 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19064256

RESUMEN

Staphylococcus aureus is both a colonizer of humans and a cause of severe invasive infections. Although the genetic basis for phenotype switching from colonizing to invasive has received significant study, knowledge of host factors that antagonize the switch is limited. We show that VLDL and LDL lipoproteins interfere with this switch by antagonizing the S. aureus agr quorum-sensing system that upregulates genes required for invasive infection. The mechanism of antagonism entails binding of the major structural protein of these lipoproteins, apolipoprotein B, to an S. aureus autoinducing pheromone, preventing attachment of this pheromone to the bacteria and subsequent signaling through its receptor, AgrC. Mice deficient in plasma apolipoprotein B, either genetically or pharmacologically, are more susceptible to invasive agr+ bacterial infection, but not to infection with an agr deletion mutant. Therefore, apolipoprotein B at homeostatic levels in blood is an essential innate defense effector against invasive S. aureus infection.


Asunto(s)
Apolipoproteínas B/inmunología , Apolipoproteínas B/metabolismo , Staphylococcus aureus/inmunología , Animales , Apolipoproteínas B/deficiencia , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Peso Corporal , Portador Sano/microbiología , Eliminación de Gen , Humanos , Pulmón/microbiología , Ratones , Ratones Noqueados , Feromonas/metabolismo , Unión Proteica , Proteínas Quinasas/metabolismo , Bazo/microbiología , Infecciones Estafilocócicas/microbiología , Análisis de Supervivencia , Transactivadores/genética , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...