Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Infect Immun ; 82(12): 5293-307, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25287924

RESUMO

The important human pathogen Streptococcus pyogenes (group A Streptococcus [GAS]) produces a hyaluronic acid (HA) capsule that plays critical roles in immune evasion. Previous studies showed that the hasABC operon encoding the capsule biosynthesis enzymes is under the control of a single promoter, P1, which is negatively regulated by the two-component regulatory system CovR/S. In this work, we characterize the sequence upstream of P1 and identify a novel regulatory region controlling transcription of the capsule biosynthesis operon in the M1 serotype strain MGAS2221. This region consists of a promoter, P2, which initiates transcription of a novel small RNA, HasS, an intrinsic transcriptional terminator that inefficiently terminates HasS, permitting read-through transcription of hasABC, and a putative promoter which lies upstream of P2. Electrophoretic mobility shift assays, quantitative reverse transcription-PCR, and transcriptional reporter data identified CovR as a negative regulator of P2. We found that the P1 and P2 promoters are completely repressed by CovR, and capsule expression is regulated by the putative promoter upstream of P2. Deletion of hasS or of the terminator eliminates CovR-binding sequences, relieving repression and increasing read-through, hasA transcription, and capsule production. Sequence analysis of 44 GAS genomes revealed a high level of polymorphism in the HasS sequence region. Most of the HasS variations were located in the terminator sequences, suggesting that this region is under strong selective pressure. We discovered that the terminator deletion mutant is highly resistant to neutrophil-mediated killing and is significantly more virulent in a mouse model of GAS invasive disease than the wild-type strain. Together, these results are consistent with the naturally occurring mutations in this region modulating GAS virulence.


Assuntos
Cápsulas Bacterianas/metabolismo , Ácido Hialurônico/biossíntese , Óperon , Elementos Reguladores de Transcrição , Streptococcus pyogenes/genética , Transcrição Gênica , Animais , Atividade Bactericida do Sangue , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Perfilação da Expressão Gênica , Genes Reporter , Variação Genética , Humanos , Evasão da Resposta Imune , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Deleção de Sequência , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Virulência
2.
Infect Immun ; 81(4): 1316-24, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23381999

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Nuclease do Micrococo/biossíntese , Proteínas Quinases/metabolismo , Staphylococcus aureus/patogenicidade , Fatores de Virulência/biossíntese , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Peritonite/microbiologia , Peritonite/patologia , Regulon , Staphylococcus aureus/genética , Fatores de Transcrição
3.
Mol Microbiol ; 79(3): 814-25, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21255120

RESUMO

Staphylococcus aureus is an important pathogen that continues to be a significant global health threat because of the prevalence of methicillin-resistant S. aureus strains (MRSA). The pathogenesis of this organism is partly attributed to the production of a large repertoire of cytotoxins that target and kill innate immune cells, which provide the first line of defence against S. aureus infection. Here we demonstrate that leukocidin A/B (LukAB) is required and sufficient for the ability of S. aureus, including MRSA, to kill human neutrophils, macrophages and dendritic cells. LukAB targets the plasma membrane of host cells resulting in cellular swelling and subsequent cell death. We found that S. aureus lacking lukAB are severely impaired in their ability to kill phagocytes during bacteria-phagocyte interaction, which in turn renders the lukAB-negative staphylococci more susceptible to killing by neutrophils. Notably, we show that lukAB is expressed in vivo within abscesses in a murine infection model and that it contributes significantly to pathogenesis of MRSA in an animal host. Collectively, these results extend our understanding of how S. aureus avoids phagocyte-mediated clearance, and underscore LukAB as an important factor that contributes to staphylococcal pathogenesis.


Assuntos
Citotoxinas/metabolismo , Staphylococcus aureus/patogenicidade , Morte Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Citoproteção , Células HL-60 , Humanos , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Fagócitos/citologia , Fagócitos/metabolismo , Fagócitos/ultraestrutura
4.
J Innate Immun ; 6(1): 21-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23816635

RESUMO

The ability of Staphylococcus aureus to infect tissues is dependent on precise control of virulence through gene-regulatory systems. While the SaeR/S two-component system has been shown to be a major regulator of S. aureus virulence, the influence of the host environment on SaeR/S-regulated genes (saeR/S targets) remains incompletely defined. Using QuantiGene 2.0 transcriptional assays, we examined expression of genes with the SaeR binding site in USA300 exposed to human and mouse neutrophils and host-derived peptides and during subcutaneous skin infection. We found that only some of the saeR/S targets, as opposed to the entire SaeR/S virulon, were activated within 5 and 10 min of interacting with human neutrophils as well as α-defensin. Furthermore, mouse neutrophils promoted transcription of saeR/S targets despite lacking α-defensin, and the murine skin environment elicited a distinctive expression profile of saeR/S targets. These findings indicate that saeR/S-mediated transcription is unique to and dependent on specific host stimuli. By using isogenic USA300ΔsaeR/S and USA300Δagr knockout strains, we also determined that SaeR/S is the major regulator of virulence factors, while Agr, a quorum-sensing two-component system, has moderate influence on transcription of the saeR/S targets under the tested physiological conditions.


Assuntos
Proteínas de Bactérias/metabolismo , Neutrófilos/imunologia , Pele/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Regulação Bacteriana da Expressão Gênica , Técnicas de Inativação de Genes , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Camundongos , Análise em Microsséries , Neutrófilos/microbiologia , Pele/microbiologia , Staphylococcus aureus/patogenicidade , Transativadores , Fatores de Transcrição , Transcriptoma , Virulência/genética , alfa-Defensinas/metabolismo
5.
PLoS One ; 9(5): e97742, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24840307

RESUMO

This study investigated the potential antibacterial activity of three series of compounds synthesized from 12 linear and branched polyamines with 2-8 amino groups, which were substituted to produce the corresponding guanides, biguanides, or phenylguanides, against Acinetobacter baumannii, Enterococcus faecalis, Escherichia coli, Pseudomonas aeruginosa and Staphylococcus aureus. Antibacterial activity was measured for each compound by determining the minimum inhibitory concentration against the bacteria, and the toxicity towards mammalian cells was determined. The most effective compound, THAM trisphenylguanide, was studied in time-to-kill and cytoplasmic leakage assays against methicillin-resistant Staphylococcus aureus (MRSA, USA300) in comparison to chlorhexidine. Preliminary toxicity and MRSA challenge studies in mice were also conducted on this compound. THAM trisphenylguanide showed significant antibacterial activity (MIC ∼1 mg/L) and selectivity against MRSA relative to all the other bacteria examined. In time-to-kill assays it showed increased antimicrobial activity against MRSA versus chlorhexidine. It induced leakage of cytoplasmic content at concentrations that did not reduce cell viability, suggesting the mechanism of action may involve membrane disruption. Using an intraperitoneal mouse model of invasive MRSA disease, THAM trisphenylguanide reduced bacterial burden locally and in deeper tissues. This study has identified a novel guanide compound with selective microbicidal activity against Staphylococcus aureus, including a methicillin-resistant (MRSA) strain.


Assuntos
Antibacterianos/farmacologia , Biguanidas/química , Guanidinas/química , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Acinetobacter baumannii/efeitos dos fármacos , Animais , Biguanidas/farmacologia , Enterococcus faecalis/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Guanidinas/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Pseudomonas aeruginosa
6.
Microbes Infect ; 15(10-11): 749-54, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23792139

RESUMO

Invasive Staphylococcus aureus (S. aureus) disease is associated with neutrophil activity and pro-inflammatory cytokine expression, including interferon-gamma (IFNγ). Using a mouse model of S. aureus peritonitis, we identify neutrophils as the predominant source of IFNγ and link this induction with the SaeR/S two-component gene regulatory system. Relative to wild-type (BALB/c) mice, IFNγ-deficient mice demonstrated increased bacterial clearance and reduced cellular cytotoxicity following intraperitoneal challenge with S. aureus. Interestingly, bacterial burden and cytotoxicity were similar in BALB/c and IFNγ-deficient mice when infected with an isogenic saeR/S mutant strain. These findings suggest saeR/S-mediated neutrophil-derived IFNγ diminishes innate antibacterial mechanisms against S. aureus.


Assuntos
Proteínas de Bactérias/imunologia , Interferon gama/metabolismo , Neutrófilos/imunologia , Neutrófilos/microbiologia , Peritonite/imunologia , Proteínas Quinases/imunologia , Infecções Estafilocócicas/imunologia , Animais , Carga Bacteriana , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Peritonite/microbiologia , Peritonite/patologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Fatores de Transcrição
7.
PLoS One ; 7(5): e36532, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22574180

RESUMO

This investigation examines the influence of alpha-toxin (Hla) during USA300 infection of human leukocytes. Survival of an USA300 isogenic deletion mutant of hla (USA300Δhla) in human blood was comparable to the parental wild-type strain and polymorphonuclear leukocyte (PMN) plasma membrane permeability caused by USA300 did not require Hla. Flow cytometry analysis of peripheral blood mononuclear cells (PBMCs) following infection by USA300, USA300Δhla, and USA300Δhla transformed with a plasmid over-expressing Hla (USA300Δhla Comp) demonstrated this toxin plays a significant role inducing plasma membrane permeability of CD14(+), CD3(+), and CD19(+) PBMCs. Rapid plasma membrane permeability independent of Hla was observed for PMNs, CD14(+) and CD19(+) PBMCs following intoxication with USA300 supernatant while the majority of CD3(+) PBMC plasma membrane permeability induced by USA300 required Hla. Addition of recombinant Hla to USA300Δhla supernatant rescued CD3(+) and CD19(+) PBMC plasma membrane permeability generated by USA300 supernatant. An observed delay in plasma membrane permeability caused by Hla in conjunction with Annexin V binding and ApoBrdU Tunel assays examining PBMCs intoxicated with recombinant Hla or infected with USA300, USA300Δhla, USA300Δhla Comp, and USA300ΔsaeR/S suggest Hla induces programmed cell death of monocytes, B cells, and T cells that results in plasma membrane permeability. Together these findings underscore the importance of Hla during S. aureus infection of human tissue and specifically demonstrate Hla activity during USA300 infection triggers programmed cell death of human monocytes, T cells and B cells that leads to plasma membrane permeability.


Assuntos
Apoptose/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/fisiologia , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/microbiologia , Toxinas Bacterianas/genética , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/metabolismo , Humanos , Leucócitos Mononucleares/microbiologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/microbiologia , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidade , Deleção de Sequência , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/microbiologia
8.
PLoS One ; 6(5): e19939, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21603642

RESUMO

Community-associated methicillin-resistant Staphylococcus aureus accounts for a large portion of the increased staphylococcal disease incidence and can cause illness ranging from mild skin infections to rapidly fatal sepsis syndromes. Currently, we have limited understanding of S. aureus-derived mechanisms contributing to bacterial pathogenesis and host inflammation during staphylococcal disease. Herein, we characterize an influential role for the saeR/S two-component gene regulatory system in mediating cytokine induction using mouse models of S. aureus pathogenesis. Invasive S. aureus infection induced the production of localized and systemic pro-inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interferon gamma (IFN-γ), interleukin (IL)-6 and IL-2. In contrast, mice infected with an isogenic saeR/S deletion mutant demonstrated significantly reduced pro-inflammatory cytokine levels. Additionally, secreted factors influenced by saeR/S elicited pro-inflammatory cytokines in human blood ex vivo. Our study further demonstrated robust saeR/S-mediated IFN-γ production during both invasive and subcutaneous skin infections. Results also indicated a critical role for saeR/S in promoting bacterial survival and enhancing host mortality during S. aureus peritonitis. Taken together, this study provides insight into specific mechanisms used by S. aureus during staphylococcal disease and characterizes a relationship between a bacterial global regulator of virulence and the production of pro-inflammatory mediators.


Assuntos
Proteínas de Bactérias/fisiologia , Citocinas/biossíntese , Infecções Estafilocócicas/patologia , Staphylococcus aureus/patogenicidade , Animais , Humanos , Mediadores da Inflamação , Camundongos , Peritonite/microbiologia , Peritonite/mortalidade , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA