Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Bacteriol ; 198(21): 2914-2924, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27501984

RESUMEN

Regulation of icaADBC-encoded polysaccharide intercellular adhesin (PIA)/poly-N-acetylglucosasmine (PNAG) production in staphylococci plays an important role in biofilm-associated medical-device-related infections. Here, we report that the AraC-type transcriptional regulator Rbf activates icaADBC operon transcription and PIA production in Staphylococcus epidermidis Purified recombinant Rbf did not bind to the ica operon promoter region in electrophoretic mobility shift assays (EMSAs), indicating that Rbf regulates ica transcription indirectly. To identify the putative transcription factor(s) involved in Rbf-mediated icaADBC regulation, the ability of recombinant Rbf to interact with the promoter sequences of known icaADBC regulators was investigated. Recombinant Rbf bound to the sarR promoter and not the sarX, sarA, sarZ, spx, and srrA promoters. Reverse transcription (RT)-PCR demonstrated that Rbf acts as a repressor of sarR transcription. PIA expression and biofilm production were restored to wild-type levels in an rbf sarR double mutant grown in brain heart infusion (BHI) medium supplemented with NaCl, which is known to activate the ica locus, but not in BHI medium alone. RT-PCR further demonstrated that although Rbf does not bind the sarX promoter, it nevertheless exerted a negative effect on sarX expression. Apparently, direct downregulation of the SarR repressor by Rbf has a dominant effect over indirect repression of the SarX activator by Rbf in the control of S. epidermidis PIA production and biofilm formation. IMPORTANCE: The importance of Staphylococcus epidermidis as an opportunistic pathogen in hospital patients with implanted medical devices derives largely from its capacity to form biofilm. Expression of the icaADBC-encoded extracellular polysaccharide is the predominant biofilm mechanism in S. epidermidis clinical isolates and is tightly regulated. Here, we report that the transcriptional regulator Rbf promotes icaADBC expression by negatively regulating expression of sarR, which encodes an ica operon repressor. Furthermore, Rbf indirectly represses the ica operon activator, SarX. The data reveal complicated interplay between Rbf and two Sar family proteins in fine-tuning regulation of the biofilm phenotype and indicate that in the hierarchy of biofilm regulators, IcaR is dominant over the Rbf-SarR-SarX axis.


Asunto(s)
Amidohidrolasas/metabolismo , Biopelículas , Regulación Bacteriana de la Expresión Génica , Operón , Polisacáridos Bacterianos/metabolismo , Proteínas Represoras/genética , Staphylococcus epidermidis/fisiología , Factores de Transcripción/metabolismo , Amidohidrolasas/genética , Regulación hacia Abajo , Fenotipo , Polisacáridos Bacterianos/genética , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Staphylococcus epidermidis/genética , Factores de Transcripción/genética
2.
J Bacteriol ; 196(19): 3482-93, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25070736

RESUMEN

Staphylococcus epidermidis is an opportunistic pathogen that is one of the leading causes of medical device infections. Global regulators like the agr quorum-sensing system in this pathogen have received a limited amount of attention, leaving important questions unanswered. There are three agr types in S. epidermidis strains, but only one of the autoinducing peptide (AIP) signals has been identified (AIP-I), and cross talk between agr systems has not been tested. We structurally characterized all three AIP types using mass spectrometry and discovered that the AIP-II and AIP-III signals are 12 residues in length, making them the largest staphylococcal AIPs identified to date. S. epidermidis agr reporter strains were developed for each system, and we determined that cross-inhibitory interactions occur between the agr type I and II systems and between the agr type I and III systems. In contrast, no cross talk was observed between the type II and III systems. To further understand the outputs of the S. epidermidis agr system, an RNAIII mutant was constructed, and microarray studies revealed that exoenzymes (Ecp protease and Geh lipase) and low-molecular-weight toxins were downregulated in the mutant. Follow-up analysis of Ecp confirmed the RNAIII is required to induce protease activity and that agr cross talk modulates Ecp activity in a manner that mirrors the agr reporter results. Finally, we demonstrated that the agr system enhances skin colonization by S. epidermidis using a porcine model. This work expands our knowledge of S. epidermidis agr system function and will aid future studies on cell-cell communication in this important opportunistic pathogen.


Asunto(s)
Proteínas Bacterianas/metabolismo , Péptidos Cíclicos/metabolismo , Percepción de Quorum , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/fisiología , Animales , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Péptidos Cíclicos/genética , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/crecimiento & desarrollo , Porcinos
3.
Infect Genet Evol ; 117: 105545, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38160879

RESUMEN

Staphylococcus aureus are gram-positive bacteria responsible for a wide array of diseases, ranging from skin and soft tissue infections to more chronic illnesses such as toxic shock syndrome, osteomyelitis, and endocarditis. Vancomycin is currently one of the most effective antibiotics available in treating patients infected with methicillin-resistant S. aureus (MRSA), however the emergence of vancomycin-resistant S. aureus (VRSA), and more commonly vancomycin-intermediate S. aureus (VISA), threaten the future efficacy of vancomycin. Intermediate resistance to vancomycin occurs due to mutations within the loci of Staphylococcal genes involved in cell wall formation such as rpoB, graS, and yycG. We hypothesized the VISA phenotype may also arise as a result of the natural stress occurring within S. aureus biofilms, and that this phenomenon is mediated by the RecA/SOS response. Wildtype and recA null mutant/lexAG94E strains of S. aureus biofilms were established in biofilm microtiter assays or planktonic cultures with or without the addition of sub-inhibitory concentrations of vancomycin (0.063 mg/l - 0.25 mg/L ciprofloxacin, 0.5 mg/l vancomycin). Efficiency of plating techniques were used to quantify the subpopulation of biofilm-derived S. aureus cells that developed vancomycin-intermediate resistance. The results indicated that a greater subpopulation of cells from wildtype biofilms (4.16 × 102 CFUs) emerged from intermediate-resistant concentrations of vancomycin (4 µg/ml) compared with the planktonic counterpart (1.53 × 101 CFUs). Wildtype biofilms (4.16 × 102 CFUs) also exhibited greater resistance to intermediate-resistant concentrations of vancomycin compared with strains deficient in the recA null mutant (8.15 × 101 CFUs) and lexA genes (8.00 × 101 CFUs). While the VISA phenotype would be an unintended consequence of genetic diversity and potentially gene transfer in the biofilm setting, it demonstrates that mutations occurring within biofilms allow for S. aureus to adapt to new environments, including the presence of widely used antibiotics.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Humanos , Vancomicina/farmacología , Staphylococcus aureus/genética , Staphylococcus aureus Resistente a Vancomicina , Staphylococcus aureus Resistente a Meticilina/genética , Antibacterianos/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Biopelículas , Pruebas de Sensibilidad Microbiana
4.
J Bacteriol ; 195(8): 1779-88, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23396916

RESUMEN

Staphylococcus aureus is a ubiquitous bacterial pathogen that is the causative agent of numerous acute and chronic infections. S. aureus colonizes the anterior nares of a significant portion of the healthy adult population, but the mechanisms of colonization remain incompletely defined. Sialic acid (N-acetylneuraminic acid [Neu5Ac]) is a bioavailable carbon and nitrogen source that is abundant on mucosal surfaces and in secretions in the commensal environment. Our findings demonstrate that Neu5Ac can serve as an S. aureus carbon source, and we have identified a previously uncharacterized chromosomal locus (nan) that is required for Neu5Ac utilization. Molecular characterization of the nan locus indicates that it contains five genes, organized into four transcripts, and the genes were renamed nanE, nanR, nanK, nanA, and nanT. Initial studies with gene deletions indicate that nanT, predicted to encode the Neu5Ac transporter, and nanA and nanE, predicted to encode catabolic enzymes, are essential for growth on Neu5Ac. Furthermore, a nanE deletion mutant exhibits a growth inhibition phenotype in the presence of Neu5Ac. Transcriptional fusions and Northern blot analyses indicate that NanR represses the expression of both the nanAT and nanE transcripts, which can be relieved with Neu5Ac. Electrophoretic mobility studies demonstrate that NanR binds to the nanAT and nanE promoter regions, and the Neu5Ac catabolic intermediate N-acetylmannosamine-6-phosphate (ManNAc-6P) relieves NanR promoter binding. Taken together, these data indicate that the nan gene cluster is essential for Neu5Ac utilization and may perform an important function for S. aureus survival in the host.


Asunto(s)
Ácido N-Acetilneuramínico/metabolismo , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Northern Blotting , Carbono/metabolismo , ADN Bacteriano/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/fisiología , Familia de Multigenes , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Staphylococcus aureus/genética , Sitio de Iniciación de la Transcripción
5.
Infect Immun ; 81(4): 1316-24, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23381999

RESUMEN

Several prominent bacterial pathogens secrete nuclease (Nuc) enzymes that have an important role in combating the host immune response. Early studies of Staphylococcus aureus Nuc attributed its regulation to the agr quorum-sensing system. However, recent microarray data have indicated that nuc is under the control of the SaeRS two-component system, which is a major regulator of S. aureus virulence determinants. Here we report that the nuc gene is directly controlled by the SaeRS two-component system through reporter fusion, immunoblotting, Nuc activity measurements, promoter mapping, and binding studies, and additionally, we were unable identify a notable regulatory link to the agr system. The observed SaeRS-dependent regulation was conserved across a wide spectrum of representative S. aureus isolates. Moreover, with community-associated methicillin-resistant S. aureus (CA MRSA) in a mouse model of peritonitis, we observed in vivo expression of Nuc activity in an SaeRS-dependent manner and determined that Nuc is a virulence factor that is important for in vivo survival, confirming the enzyme's role as a contributor to invasive disease. Finally, natural polymorphisms were identified in the SaeRS proteins, one of which was linked to Nuc regulation in a CA MRSA USA300 endocarditis isolate. Altogether, our findings demonstrate that Nuc is an important S. aureus virulence factor and part of the SaeRS regulon.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Nucleasa Microcócica/biosíntesis , Proteínas Quinasas/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/biosíntesis , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Viabilidad Microbiana , Peritonitis/microbiología , Peritonitis/patología , Regulón , Staphylococcus aureus/genética , Factores de Transcripción
6.
Biochemistry ; 50(33): 7157-67, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21749039

RESUMEN

Pulsed-field gel electrophoresis (PFGE) clonal type USA200 is the most widely disseminated Staphylococcus aureus colonizer of the nose and is a major cause of toxic shock syndrome (TSS). Exoproteins derived from these organisms have been suggested to contribute to their colonization and causation of human diseases but have not been well-characterized. Two representative S. aureus USA200 isolates, MNPE (α-toxin positive) and CDC587 (α-toxin mutant), isolated from pulmonary post-influenza TSS and menstrual vaginal TSS, respectively, were evaluated. Biochemical, immunobiological, and cell-based assays, including mass spectrometry, were used to identify key exoproteins derived from the strains that are responsible for proinflammatory and cytotoxic activity on human vaginal epithelial cells. Exoproteins associated with virulence were produced by both strains, and cytolysins (α-toxin and γ-toxin), superantigens, and proteases were identified as the major exoproteins, which caused epithelial cell inflammation and cytotoxicity. Exoprotein fractions from MNPE were more proinflammatory and cytotoxic than those from CDC587 due to high concentrations of α-toxin. CDC587 produced a small amount of α-toxin, despite the presence of a stop codon (TAG) at codon 113. Additional exotoxin identification studies of USA200 strain [S. aureus MN8 (α-toxin mutant)] confirmed that MN8 also produced low levels of α-toxin despite the same stop codon. The differences observed in virulence factor profiles of two USA200 strains provide insight into environmental factors that select for specific virulence factors. Cytolysins, superantigens, and proteases were identified as potential targets, where toxin neutralization may prevent or diminish epithelial damage associated with S. aureus.


Asunto(s)
Citotoxinas/inmunología , Enterotoxinas/inmunología , Exotoxinas/inmunología , Choque Séptico/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Vagina/inmunología , Animales , Cromatografía Líquida de Alta Presión , Citotoxinas/metabolismo , Electroforesis en Gel de Campo Pulsado , Enterotoxinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Exotoxinas/metabolismo , Femenino , Humanos , Immunoblotting , Inmunoglobulina G/inmunología , Interleucina-8/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Conejos , Choque Séptico/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/aislamiento & purificación , Superantígenos/metabolismo , Porcinos , Vagina/metabolismo , Vagina/microbiología , Factores de Virulencia
7.
Methods Mol Biol ; 2341: 153-159, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34264471

RESUMEN

Biofilms provide an environment in which bacteria can survive adverse conditions such as nutrient or oxygen deficiencies, and antibiotic treatments. Bacterial survival of antibiotic treatments can often result in antimicrobial resistance, which can make treating infections substantially more difficult, increase the burden of healthcare costs, and hinder the healing of infected wounds. As Staphylococcus aureus is a bacterium that commonly causes skin infections, can be found in infected skin wounds, and is prone to developing antimicrobial resistance-especially within a biofilm microenvironment, the study and development of methodologies to treat infected wounds have become an important topic of research. To study the development of bacterial biofilm in a skin wound, this chapter discusses an in vitro model to access biofilm growth in an environment that mimics a human skin wound. This model serves as a tool to study the biofilm growth and efficacy of antibiotic use in an in vitro system that more closely resembles human skin tissue, rather than a polystyrene plate.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Staphylococcus aureus/patogenicidad , Infección de Heridas/microbiología , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Farmacorresistencia Bacteriana , Humanos , Viabilidad Microbiana , Modelos Biológicos , Piel/microbiología , Staphylococcus aureus/efectos de los fármacos
8.
J Antimicrob Chemother ; 65(10): 2164-71, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20719763

RESUMEN

OBJECTIVES AND METHODS: This study addressed the efficacy of daptomycin, vancomycin, rifampicin, daptomycin/rifampicin and vancomycin/rifampicin against a polysaccharide intercellular adhesin (PIA)-dependent and -independent Staphylococcus epidermidis biofilm using flow cell and guinea pig tissue cage models. RESULTS: The flow cell model of both PIA-dependent and -independent biofilms demonstrated that the viable cell count after treatment with daptomycin/rifampicin was significantly lower (P<0.05) than after treatment with vancomycin, vancomycin/rifampicin, daptomycin or rifampicin alone. To validate these observations, a guinea pig tissue cage model was used. The results demonstrated that the addition of rifampicin to daptomycin or vancomycin sterilized 5/6 tissues cages colonized with S. epidermidis 1457 (PIA producing). Similar results were noted with S. epidermidis 1457 icaADBC::dhfr (non-PIA producing), where daptomycin/rifampicin and vancomycin/rifampicin sterilized 5/6 and 6/6 tissue cages, respectively. There was no statistical difference in comparison with the no-treatment control when both 1457 and 1457 icaADBC::dhfr were treated with vancomycin and daptomycin alone. Furthermore, treatment with rifampicin alone sterilized 5/6 and 3/6 1457 and 1457 icaADBC::dhfr tissue cages, respectively. CONCLUSIONS: Interpretation of these data suggests that rifampicin is highly active against S. epidermidis biofilms and both vancomycin and daptomycin are effective at reducing the subpopulation of bacteria that develop rifampicin resistance.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Daptomicina/farmacología , Rifampin/farmacología , Staphylococcus epidermidis/efectos de los fármacos , Vancomicina/farmacología , Animales , Biopelículas/crecimiento & desarrollo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Cobayas , Pruebas de Sensibilidad Microbiana , Polisacáridos Bacterianos/metabolismo , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Infecciones Cutáneas Estafilocócicas/microbiología , Staphylococcus epidermidis/crecimiento & desarrollo , Factores de Virulencia/metabolismo
9.
J Bacteriol ; 190(23): 7621-32, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18820013

RESUMEN

Staphylococcus epidermidis is a major nosocomial pathogen primarily infecting immunocompromised individuals or those with implanted biomaterials (e.g., catheters). Biomaterial-associated infections often involve the formation of a biofilm on the surface of the medical device. In S. epidermidis, polysaccharide intercellular adhesin (PIA) is an important mediator of biofilm formation and pathogenesis. Synthesis of PIA is regulated by at least three DNA binding proteins (IcaR, SarA, and sigma(B)) and several environmental and nutritional conditions. Previously, we observed the environmental conditions that increased PIA synthesis decreased tricarboxylic acid (TCA) cycle activity. In this study, S. epidermidis TCA cycle mutants were constructed, and the function of central metabolism in PIA biosynthesis was examined. TCA cycle inactivation altered the metabolic status of S. epidermidis, resulting in a massive derepression of PIA biosynthetic genes and a redirection of carbon from growth into PIA biosynthesis. These data demonstrate that the bacterial metabolic status is a critical regulatory determinant of PIA synthesis. In addition, these data lead us to propose that the TCA cycle acts as a signal transduction pathway to translate external environmental cues into intracellular metabolic signals that modulate the activity of transcriptional regulators.


Asunto(s)
Ciclo del Ácido Cítrico/fisiología , Polisacáridos Bacterianos/biosíntesis , Staphylococcus epidermidis/metabolismo , Aconitato Hidratasa/genética , Aconitato Hidratasa/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citratos , Medios de Cultivo , Regulación Bacteriana de la Expresión Génica/fisiología , Silenciador del Gen , Mutación , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/crecimiento & desarrollo , Tiempo
10.
J Med Microbiol ; 57(Pt 8): 1036-1038, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18628509

RESUMEN

Daptomycin is a novel bactericidal agent active against Gram-positive pathogens including meticillin-resistant Staphylococcus aureus (MRSA). Our case is unique in the description of an MRSA USA 300 isolate that developed decreased susceptibility to daptomycin during daptomycin and vancomycin therapy. Directed sequencing detected a previously reported mutation in mprF, resulting in a T345A substitution, associated with non-susceptibility to daptomycin.


Asunto(s)
Daptomicina/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Adulto , Antibacterianos , Hepatitis C/complicaciones , Humanos , Masculino , Resistencia a la Meticilina , Staphylococcus aureus/aislamiento & purificación , Abuso de Sustancias por Vía Intravenosa/complicaciones
11.
Methods Mol Biol ; 1373: 69-74, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25646608

RESUMEN

The genetic manipulation of Staphylococcus aureus for molecular experimentation is a valuable tool for assessing gene function and virulence. Genetic variability between strains coupled with difficult laboratory techniques for strain construction is a frequent roadblock in S. aureus research. Bacteriophage transduction greatly increases the speed and ease of S. aureus studies by allowing movement of chromosomal markers and plasmids between strains. This technique enables the S. aureus research community to focus investigations on clinically relevant isolates.


Asunto(s)
Bacteriófagos/genética , Infecciones Estafilocócicas/genética , Staphylococcus aureus/genética , Transducción Genética/métodos , Humanos , Lisogenia/genética , Plásmidos/genética , Infecciones Estafilocócicas/microbiología
12.
Methods Mol Biol ; 1106: 167-72, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24222465

RESUMEN

The genetic manipulation of Staphylococcus epidermidis for molecular experimentation has long been an area of difficulty. Many of the traditional laboratory techniques for strain construction are laborious and hampered by poor efficiency. The ability to move chromosomal genetic markers and plasmids using bacteriophage transduction has greatly increased the speed and ease of S. epidermidis studies. These molecular genetic advances have advanced the S. epidermidis research field beyond a select few genetically tractable strains and facilitated investigations of clinically relevant isolates.


Asunto(s)
Bacteriófagos/genética , Staphylococcus epidermidis/virología , Transducción Genética , Genes Bacterianos , Staphylococcus epidermidis/genética , Cultivo de Virus
13.
Nat Med ; 20(3): 301-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24487433

RESUMEN

Technologies that enable the rapid detection and localization of bacterial infections in living animals could address an unmet need for infectious disease diagnostics. We describe a molecular imaging approach for the specific, noninvasive detection of S. aureus based on the activity of the S. aureus secreted nuclease, micrococcal nuclease (MN). Several short synthetic oligonucleotides, rendered resistant to mammalian serum nucleases by various chemical modifications and flanked with a fluorophore and quencher, were activated upon degradation by purified MN and in S. aureus culture supernatants. A probe consisting of a pair of deoxythymidines flanked by several 2'-O-methyl-modified nucleotides was activated in culture supernatants of S. aureus but not in culture supernatants of several other pathogenic bacteria. Systemic administration of this probe to mice bearing S. aureus muscle infections resulted in probe activation at the infection sites in an MN-dependent manner. This new bacterial imaging approach has potential clinical applicability for infections with S. aureus and several other medically important pathogens.


Asunto(s)
Nucleasa Microcócica/química , Infecciones Estafilocócicas/diagnóstico , Infecciones Estafilocócicas/patología , Animales , Bacteriófagos/metabolismo , Femenino , Fluorescencia , Colorantes Fluorescentes/química , Cinética , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Sondas de Oligonucleótidos/química , Oligonucleótidos/química , Piomiositis/diagnóstico , Piomiositis/microbiología
14.
Cell Host Microbe ; 13(6): 629-31, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23768487

RESUMEN

Osteomyelitis is a debilitating bone infection often caused by the bacterial pathogen Staphylococcus aureus. In this issue, Cassat et al. (2013) develop a high-resolution microcomputed tomography (microCT) method to visualize bone remodeling during S. aureus infection and discover that the metalloprotease aureolysin plays a critical role in modulating osteomyelitis pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Remodelación Ósea , Metaloendopeptidasas/metabolismo , Osteomielitis/patología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo , Animales
15.
Future Microbiol ; 5(6): 917-33, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20521936

RESUMEN

Staphylococcus epidermidis is a highly significant nosocomial pathogen mediating infections primarily associated with indwelling biomaterials (e.g., catheters and prostheses). In contrast to Staphylococcus aureus, virulence properties associated with S. epidermidis are few and biofilm formation is the defining virulence factor associated with disease, as demonstrated by animal models of biomaterial-related infections. However, other virulence factors, such as phenol-soluble modulins and poly-gamma-DL-glutamic acid, have been recently recognized that thwart innate immune system mechanisms. Formation of S. epidermidis biofilm is typically considered a four-step process consisting of adherence, accumulation, maturation and dispersal. This article will discuss recent advances in the study of these four steps, including accumulation, which can be either polysaccharide or protein mediated. It is hypothesized that studies focused on understanding the biological function of each step in staphylococcal biofilm formation will yield new treatment modalities to treat these recalcitrant infections.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Staphylococcus epidermidis/fisiología , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Infección Hospitalaria/microbiología , Humanos , Polisacáridos Bacterianos/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/crecimiento & desarrollo , Staphylococcus epidermidis/metabolismo , Virulencia
16.
J Clin Microbiol ; 43(9): 4751-7, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16145137

RESUMEN

Staphylococcus epidermidis is part of the normal microflora of the human skin but is also a leading cause of device-associated infections in critically ill patients. Commensal and clinical S. epidermidis isolates differ in their ability to form biofilms on medical devices; the synthesis of biofilms is mediated by the icaADBC operon. Currently, the epidemiological relatedness between ica-positive and -negative isolates is not known; neither is it known whether the ica genes can spread to biofilm-negative strains through horizontal gene transfer. In this study, multilocus sequence typing (MLST) was employed for the clonal analysis of 118 S. epidermidis ica-positive and -negative strains. MLST revealed that the majority of ica-positive and -negative strains were closely related and formed a single clonal complex. Within this complex one sequence type (ST27) was identified which contained exclusively ica-positive isolates and represented the majority of clinical strains tested. ST27 and related ica-positive clones carried different SCCmec cassettes (conferring methicillin resistance) and the insertion sequence IS256. The findings suggest that the S. epidermidis infections analyzed in this report are mainly caused by a single clone (ST27) which occurs preferentially in hospitals and differs from clones in the community. It is hypothesized that the successful establishment of ST27 within nosocomial environments has been facilitated by the presence of genes encoding biofilm and resistance traits.


Asunto(s)
Proteínas Bacterianas/genética , Técnicas de Tipificación Bacteriana , Biopelículas/crecimiento & desarrollo , Análisis de Secuencia de ADN , Staphylococcus epidermidis/clasificación , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Elementos Transponibles de ADN , Humanos , Datos de Secuencia Molecular , Operón , Recombinación Genética , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/aislamiento & purificación , Staphylococcus epidermidis/metabolismo
17.
Appl Environ Microbiol ; 70(11): 6846-54, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15528552

RESUMEN

Non-sorbitol-fermenting, beta-glucuronidase-negative Escherichia coli O157:H7 strains are regarded as a clone complex, and populations from different geographical locations are believed to share a recent common ancestor. Despite their relatedness, high-resolution genotyping methods can detect significant genome variation among different populations. Phylogenetic analysis of high-resolution genotyping data from these strains has shown that subpopulations from geographically unlinked continents can be divided into two primary phylogenetic lineages, termed lineage I and lineage II, and limited studies of the distribution of these lineages suggest there could be differences in their propensity to cause disease in humans or to be transmitted to humans. Because the genotyping methods necessary to discriminate the two lineages are tedious and subjective, these methods are not particularly suited for studying the large sets of strains that are required to systematically evaluate the ecology and transmission characteristics of these lineages. To overcome this limitation, we have developed a lineage-specific polymorphism assay (LSPA) that can readily distinguish between the lineage I and lineage II subpopulations. In the studies reported here, we describe the development of a six-marker test (LSPA-6) and its validation in a side-by-side comparison with octamer-based genome scanning. Analysis of over 1,400 O157:H7 strains with the LSPA-6 demonstrated that five genotypes comprise over 91% of the strains, suggesting that these subpopulations may be widespread.


Asunto(s)
Técnicas de Tipificación Bacteriana , Enfermedades de los Bovinos/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/clasificación , Glucuronidasa/metabolismo , Polimorfismo Genético , Sorbitol/metabolismo , Animales , Bovinos , Infecciones por Escherichia coli/veterinaria , Escherichia coli O157/enzimología , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Genotipo , Humanos , Filogenia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA