Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Lett Appl Microbiol ; 61(2): 158-64, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25963798

RESUMEN

UNLABELLED: α-Defensins produced by neutrophils are important effector molecules of the innate immune system. In addition to their microbicidal effects, α-defensins have the ability to neutralize bacterial toxins. Panton-Valentine leukocidin (PVL) is the hallmark of community-acquired methicillin-resistant Staphylococcus aureus. Staphylococcus aureus that produce PVL are responsible for severe diseases, including necrotizing pneumonia. Polymorphonuclear neutrophils (PMNs) are the target cells of PVL action. The goal of this study was to elucidate the effect of a group of α-defensins known as the human neutrophil peptides (HNPs) on the interactions between LukS-PV and LukF-PV, which compose PVL, and human PMNs. We observed that HNPs bound to both subunits of PVL and significantly decreased PVL pore formation in PMNs, with a maximum inhibition of 27%. When various HNP molecules were tested individually under the same conditions, we observed that HNP3, but not HNP1 or 2, decreased pore formation. Similarly, HNP3 significantly decreased PVL-induced PMN lysis, with a maximum inhibition of 31%. Interestingly, HNPs did not affect LukS-PV LukF-PV oligomerization, LukS-PV LukF-PV binding to PMNs or calcium influx induced by PVL in PMNs. Our results suggest that HNP3 partially protects neutrophils against PVL by interfering with the conformational changes of PVL required to form a functional pore. SIGNIFICANCE AND IMPACT OF THE STUDY: Panton-Valentine leukocidin (PVL) is a pore-forming toxin produced by Staphylococcus aureus, responsible for neutrophil damage and key player of severe staphylococcal diseases. Antimicrobial peptides produced by neutrophils (HNP1-3) neutralize several other bacterial cytotoxins. We examined the impact of human neutrophil peptides (HNPs) on PVL cytotoxicity against human neutrophils and we found that HNPs bind to both LukS and LukF components of PVL, thereby inhibiting pore formation and neutrophil lysis. Our results suggest that HNP3 may impair PVL conformational changes required to form a functional pore and provide insight into the pathogenesis of PVL-related staphylococcal infection, with potential impact on the disease outcome.


Asunto(s)
Toxinas Bacterianas/toxicidad , Exotoxinas/toxicidad , Leucocidinas/toxicidad , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neutrófilos/enzimología , Infecciones Estafilocócicas/inmunología , alfa-Defensinas/metabolismo , Proteínas Bacterianas/metabolismo , Humanos , Leucocidinas/metabolismo , Neutrófilos/inmunología
2.
Eur J Clin Microbiol Infect Dis ; 34(7): 1443-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25864191

RESUMEN

Staphylococcus aureus is both a component of the normal skin flora and an important pathogen. It expresses a range of recognized and putative virulence factors, such as enterotoxins with superantigenic properties. Several superantigen genes, i.e., seg, sei, selm, seln, and selo, are encoded by the enterotoxin gene cluster (egc), which is found in the majority of S. aureus isolates. Carriage of egc is associated with fitness of S. aureus in the gut microbiota, but it is not known if it contributes to pathogenicity. We constructed egc+ (functional for the seg, selm, and selo genes) and isogenic egc- S. aureus mutants, and investigated their virulence profiles in murine infection models. No effect of egc was seen in a local skin and soft tissue infection model, but in an invasive infection model, increased weight loss was observed after infection with the egc+ as compared to the egc- mutant. Mortality and arthritis were not affected by egc status. Our data suggest that egc has limited effects on the virulence of S. aureus. It may primarily function as a colonization factor increasing commensal fitness, although it might have some aggravating effects on the infection when the bacteria reach the blood.


Asunto(s)
Enterotoxinas/genética , Familia de Multigenes , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Superantígenos/genética , Animales , Modelos Animales de Enfermedad , Femenino , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Sitios Genéticos , Humanos , Ratones , Mutación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Infecciones Estafilocócicas/mortalidad , Infecciones Estafilocócicas/patología , Staphylococcus aureus/patogenicidad
3.
Eur J Clin Microbiol Infect Dis ; 34(5): 927-33, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25575949

RESUMEN

Recent research on Staphylococcus aureus vaccine development has focused on active immunization against Panton-Valentine leukocidin (PVL), a potent leukotoxin associated with both superficial and severe deep-seated infections. PVL prevalence is highly variable worldwide, but it is unknown to what extent immunity to PVL varies between patients from geographic areas with different PVL-positive S. aureus prevalences. We conducted a retrospective multicentric study of anti-PVL and anti-alpha-toxin (Hla) antibody levels in uninfected adult patients from France (low PVL prevalence; n = 200), Algeria (moderate prevalence; n = 143), and Senegal (high prevalence; n = 228). The antibody levels were quantified by an enzyme-linked immunosorbent assay (ELISA) procedure. Because Hla is present in virtually all S. aureus strains, its corresponding antibody levels were considered to reflect population exposure to S. aureus. Compared with French participants, the average anti-PVL antibody levels were 2.5-fold and 8.2-fold higher in Algerian and Senegalese participants, respectively (p < 0.001). Conversely, anti-Hla antibody levels did not differ between participants from the three countries, suggesting that the observed differences in anti-PVL antibody levels were not biased by variations in population exposure to S. aureus. Hence, anti-PVL antibody levels in the general populations of France, Algeria, and Senegal vary widely and match variations in PVL-positive S. aureus strain prevalence, with an increasing north-to-south gradient. To conclude, immunity to PVL in a given population correlates with local PVL prevalence. This finding can help to inform PVL vaccine strategies.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Toxinas Bacterianas/inmunología , Exotoxinas/inmunología , Leucocidinas/inmunología , Infecciones Estafilocócicas/epidemiología , Infecciones Estafilocócicas/inmunología , Adulto , Anciano , Argelia/epidemiología , Estudios Transversales , Ensayo de Inmunoadsorción Enzimática , Femenino , Francia/epidemiología , Geografía , Proteínas Hemolisinas/inmunología , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Senegal/epidemiología , Estudios Seroepidemiológicos
4.
Int J Antimicrob Agents ; 45(1): 19-24, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25450803

RESUMEN

Community-acquired meticillin-resistant Staphylococcus aureus (CA-MRSA) causes severe diseases through virulence factors such as staphylococcal protein A (SpA), which favours immune evasion. We have previously shown that antimicrobial peptides (AMPs) and antibiotics decrease SpA expression in CA-MRSA strains. Here we examined the effects of antibiotics and AMPs, alone and in combination, on SpA expression in various CA-MRSA strains. Six S. aureus isolates corresponding to the major worldwide CA-MRSA clones (ST8-USA300, ST80 and ST30) were selected. Strains were cultured to exponential growth phase and were subsequently incubated with antibiotics (tigecycline, linezolid, clindamycin and vancomycin) at 0.25× MIC or with AMPs [human neutrophil peptide (HNP)-1-3] at the LD50, alone and in combination. After 6h, cultures were assessed for spa mRNA by RT-PCR, whilst SpA protein was measured by specific ELISA after 18h. When used alone, antibiotics (clindamycin, linezolid and tigecycline) or HNPs significantly reduced both SpA production and mRNA levels in ST30 and ST80 strains. When used in combination, HNPs and clindamycin, linezolid or tigecycline synergistically reduced SpA production (6-100-fold) and spa mRNA levels (4-20-fold) in ST80 and ST30 strains. In contrast, for USA300 strains, among all antibiotics, clindamycin alone reduced SpA production (3.5-fold), whereas with combined treatments including HNPs, only a slight reduction in SpA production (1.7-2.2-fold) was observed. In conclusion, antibiotics and AMPs do not modulate SpA expression in USA300, unlike in other CA-MRSA clones. This observation suggests that the virulence and successful spread of USA300 strains is associated with a specific regulatory network.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Proteína Estafilocócica A/biosíntesis , Factores de Virulencia/biosíntesis , Infecciones Comunitarias Adquiridas/microbiología , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Humanos , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones Estafilocócicas/microbiología , Proteína Estafilocócica A/genética , Factores de Virulencia/genética
5.
Artículo en Inglés | MEDLINE | ID: mdl-23964349

RESUMEN

The egcSEs comprise five genetically linked staphylococcal enterotoxins, SEG, SEI, SElM, SElN, and SElO and two pseudotoxins which constitute an operon present in up to 80% of Staphylococcus aureus isolates. A preparation containing these proteins was recently used to treat advanced lung cancer with pleural effusion. We investigated the hypothesis that egcSEs induce nitrous oxide (NO) and associated cytokine production and that these agents may be involved in tumoricidal effects against a broad panel of clinically relevant human tumor cells. Preliminary studies showed that egcSEs and SEA activated T cells (range: 11-25%) in a concentration dependent manner. Peripheral blood mononuclear cells (PBMCs) stimulated with equimolar quantities of egcSEs expressed NO synthase and generated robust levels of nitrite (range: 200-250 µM), a breakdown product of NO; this reaction was inhibited by NG-monomethyl-L-arginine (L-NMMA) (0.3 mM), an NO synthase antagonist. Cell free supernatants (CSFs) of all egcSE-stimulated PBMCs were also equally effective in inducing concentration dependent tumor cell apoptosis in a broad panel of human tumor cells. The latter effect was due in part to the generation of NO and TNF-α since it was significantly abolished by L-NMMA, anti-TNF-α antibodies, respectively, and a combination thereof. A hierarchy of tumor cell sensitivity to these CFSs was as follows: lung carcinoma > osteogenic sarcoma > melanoma > breast carcinoma >neuroblastoma. Notably, SEG induced robust activation of NO/TNFα-dependent tumor cell apoptosis comparable to the other egcSEs and SEA despite TNF-α and IFN-γ levels that were 2 and 8 fold lower, respectively, than the other egcSEs and SEA. Thus, egcSEs produced by S. aureus induce NO synthase and the increased NO formation together with TNF-α appear to contribute to egcSE-mediated apoptosis against a broad panel of human tumor cells.


Asunto(s)
Apoptosis , Toxinas Bacterianas/toxicidad , Citocinas/metabolismo , Enterotoxinas/toxicidad , Óxido Nitroso/metabolismo , Staphylococcus aureus/metabolismo , Toxinas Bacterianas/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Enterotoxinas/inmunología , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Familia de Multigenes
6.
Clin Microbiol Infect ; 15(2): 156-64, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19154186

RESUMEN

Methicillin-resistant Staphylococcus aureus is increasingly responsible for staphylococcal infections in the community. A large percentage of the community-acquired methicillin-resistant (CA-MRSA) strains in the USA produce Panton-Valentine leukocidin (PVL), which is associated with severe infections. The virulence of the clinical CA-MRSA strain USA300 was compared to that of its isogenic pvl-deleted mutant, and it was shown that PVL contributes to lung and muscle tissue destruction, respectively, in murine necrotizing pneumonia and skin infection models. Mice infected with the USA300 strain developed a dominant anti-PVL response. The PVL subunits were therefore tested as vaccinogens against this isolate, and their vaccine efficacy correlated with both the route of vaccination and infection. These data suggest that PVL is a virulence factor in murine CA-MRSA infections.


Asunto(s)
Toxinas Bacterianas/inmunología , Exotoxinas/inmunología , Leucocidinas/inmunología , Neumonía Bacteriana/prevención & control , Infecciones Estafilocócicas/prevención & control , Infecciones Cutáneas Estafilocócicas/prevención & control , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Factores de Virulencia/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Peso Corporal , Exotoxinas/fisiología , Femenino , Leucocidinas/fisiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Piel/patología , Staphylococcus aureus/patogenicidad , Análisis de Supervivencia , Virulencia , Factores de Virulencia/fisiología
7.
Clin Microbiol Infect ; 15(2): 144-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19154491

RESUMEN

To determine whether Staphylococcus aureus Panton-Valentine leukocidin (PVL) is expressed during human infection, anti-PVL antibody titres were compared in patients with PVL-positive and PVL-negative staphylococcal infections, and in patients with no evidence of S. aureus infection. Patients with PVL-positive strains had higher levels of anti-PVL antibodies than individuals of both control groups. The median anti-PVL titre increased 8.6-fold during the course of PVL-positive infection and 1.4-fold during PVL-negative infection. These results indicate that only PVL-positive S. aureus strains elicit significant anti-PVL antibody production in humans, and demonstrate the production of PVL during PVL-positive S. aureus infection. The protective role of this immune response remains to be established.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Toxinas Bacterianas/inmunología , Exotoxinas/inmunología , Leucocidinas/inmunología , Suero/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , Adulto Joven
8.
Clin Microbiol Infect ; 14(12): 1180-3, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19046173

RESUMEN

Pus samples were prospectively collected from patients with Staphylococcus aureus skin infections and tested for Panton-Valentine leukocidin (PVL). PVL was detected at concentrations that were toxic for rabbit skin in all specimens from patients infected with strains harbouring PVL genes.


Asunto(s)
Absceso/microbiología , Absceso/patología , Toxinas Bacterianas/análisis , Exotoxinas/análisis , Leucocidinas/análisis , Infecciones Cutáneas Estafilocócicas/patología , Staphylococcus aureus/aislamiento & purificación , Humanos , Supuración/microbiología
9.
Clin Microbiol Infect ; 14(4): 384-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18261123

RESUMEN

The capacity of Staphylococcus aureus strain LUG855 to release Panton-Valentine leukocidin (PVL) in the presence of sub-inhibitory concentrations of anti-staphylococcal drugs was examined. Oxacillin enhanced PVL release 2.5-fold, while clindamycin, linezolid, fusidic acid and rifampicin were inhibitory, and vancomycin, pristinamycin, tetracycline, ofloxacin and co-trimoxazole had no effect. In combination with oxacillin, sub-inhibitory concentrations of clindamycin or rifampicin inhibited PVL induction significantly, linezolid was less inhibitory, and fusidic acid did not inhibit PVL induction by oxacillin. These data support the use of oxacillin in combination with clindamycin, rifampicin or linezolid for the treatment of PVL-positive S. aureus infections.


Asunto(s)
Antibacterianos/farmacología , Toxinas Bacterianas/biosíntesis , Exotoxinas/biosíntesis , Regulación Bacteriana de la Expresión Génica , Leucocidinas/biosíntesis , Staphylococcus aureus/efectos de los fármacos , Acetamidas/farmacología , Toxinas Bacterianas/genética , Clindamicina/farmacología , Sinergismo Farmacológico , Quimioterapia Combinada , Exotoxinas/genética , Ácido Fusídico/farmacología , Humanos , Leucocidinas/genética , Linezolid , Pruebas de Sensibilidad Microbiana/métodos , Oxacilina/farmacología , Oxazolidinonas/farmacología , Estándares de Referencia , Rifampin/farmacología , Staphylococcus aureus/metabolismo
10.
Leuk Lymphoma ; 48(3): 605-15, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17454606

RESUMEN

Indirubin-3'-monoxime (IO) is a derivative of Indirubin, compound of a Chinese medicinal recipe used to treat various diseases including leukemia. In this study, we investigated to what extent IO inhibits the growth of normal human lymphocytes. We defined various experimental conditions of peripheral blood lymphocyte treatment: IO introduced (i) on unstimulated lymphocytes, (ii) or on stimulated lymphocytes at the time of phytohemagglutinin stimulation (L1 protocol), (iii) 48 h after the beginning of stimulation (L2 protocol), and (iv) after nocodazole synchronization of stimulated lymphocytes. IO induces a concentration dependent cytotoxic effect yielding a characteristic sub-G1 peak in normal stimulated lymphocytes. Cell death was partly due to necrosis and apoptosis. Normal unstimulated lymphocytes remained insensitive to the cytotoxic effect of 10 microM IO treatment. A cell cycle inhibition was observed after IO treatment, stronger for the L1 than for the L2 protocol, without induction of polyploidy after Nocodazole synchronization. These cellular consequences were associated with a decrease in CDK activity, and with CDK and cyclin gene expression modifications. The inhibition of lymphocyte proliferation by IO indicates that indirubin derivatives may be potent immunosuppressive agents.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Indoles/farmacología , Linfocitos/efectos de los fármacos , Oximas/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas/efectos de los fármacos , Humanos , Immunoblotting , Activación de Linfocitos/efectos de los fármacos , Linfocitos/citología , Linfocitos/metabolismo , Mitógenos/farmacología , Necrosis , Nocodazol/farmacología , Fitohemaglutininas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...