Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Infect Dis ; 217(6): 943-952, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29272502

RESUMO

Background: The ability of Staphylococcus aureus to evade killing by human neutrophils significantly contributes to disease progression. In this study, we characterize an influential role for the S. aureus SaeR/S 2-component gene regulatory system in suppressing monocyte production of tumor necrosis factor alpha (TNF-α) to subsequently influence human neutrophil priming. Methods: Using flow cytometry and TNF-α specific enzyme-linked immunosorbent assays we identify the primary cellular source of TNF-α in human blood and in purified peripheral blood mononuclear cells (PBMCs) during interaction with USA300 and an isogenic saeR/S deletion mutant (USA300∆saeR/S). Assays with conditioned media from USA300 and USA300∆saeR/S exposed PBMCs were used to investigate priming on neutrophil bactericidal activity. Results: TNF-α production from monocytes was significantly reduced following challenge with USA300 compared to USA300∆saeR/S. We observed that priming of neutrophils using conditioned medium from peripheral blood mononuclear cells stimulated with USA300∆saeR/S significantly increased neutrophil bactericidal activity against USA300 relative to unprimed neutrophils and neutrophils primed with USA300 conditioned medium. The increased neutrophil bactericidal activity was associated with enhanced reactive oxygen species production that was significantly influenced by elevated TNF-α concentrations. Conclusions: Our findings identify an immune evasion strategy used by S. aureus to impede neutrophil priming and subsequent bactericidal activity.


Assuntos
Proteínas de Bactérias/farmacologia , Staphylococcus aureus Resistente à Meticilina , Monócitos/metabolismo , Neutrófilos/imunologia , Proteínas Quinases/farmacologia , Fatores de Transcrição/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas de Bactérias/metabolismo , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Staphylococcus aureus Resistente à Meticilina/imunologia , Monócitos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Proteínas Quinases/metabolismo , Fatores de Transcrição/metabolismo
2.
Nat Commun ; 7: 13696, 2016 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-27982078

RESUMO

Asthma has multiple features, including airway hyperreactivity, inflammation and remodelling. The TNF superfamily member TNFSF14 (LIGHT), via interactions with the receptor TNFRSF14 (HVEM), can support TH2 cell generation and longevity and promote airway remodelling in mouse models of asthma, but the mechanisms by which TNFSF14 functions in this setting are incompletely understood. Here we find that mouse and human mast cells (MCs) express TNFRSF14 and that TNFSF14:TNFRSF14 interactions can enhance IgE-mediated MC signalling and mediator production. In mouse models of asthma, TNFRSF14 blockade with a neutralizing antibody administered after antigen sensitization, or genetic deletion of Tnfrsf14, diminishes plasma levels of antigen-specific IgG1 and IgE antibodies, airway hyperreactivity, airway inflammation and airway remodelling. Finally, by analysing two types of genetically MC-deficient mice after engrafting MCs that either do or do not express TNFRSF14, we show that TNFRSF14 expression on MCs significantly contributes to the development of multiple features of asthma pathology.


Assuntos
Asma/induzido quimicamente , Asma/metabolismo , Mastócitos/fisiologia , Receptores de IgE/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Remodelação das Vias Aéreas , Animais , Anticorpos , Antígenos de Dermatophagoides/imunologia , Antígenos de Dermatophagoides/toxicidade , Asma/patologia , Líquido da Lavagem Broncoalveolar/citologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genótipo , Imunoglobulina E , Imunoglobulina G , Camundongos , Camundongos Knockout , Ovalbumina/imunologia , Ovalbumina/toxicidade , Receptores de IgE/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/genética
3.
J Infect Dis ; 212(6): 934-8, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25722299

RESUMO

While Staphylococcus aureus accelerates human neutrophil cell death, the underlying host- and pathogen-derived mechanisms remain incompletely defined. Previous studies demonstrated that the S. aureus SaeR/S sensory system is essential for pathogen survival following neutrophil phagocytosis. Herein, we demonstrate that the SaeR/S system promoted accelerated cell death, suppressed phosphorylation of nuclear factor-κB, and reduced interleukin-8 (IL-8) production in human neutrophils. Treatment of neutrophils with recombinant IL-8 significantly reduced bacterial burden and apoptosis. Our findings demonstrate a mechanism by which S. aureus suppresses the early neutrophil-derived IL-8 response to disrupt cell fate and promote disease.


Assuntos
Morte Celular/fisiologia , Interleucina-8/metabolismo , Neutrófilos/fisiologia , Staphylococcus aureus/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Humanos , Interleucina-8/genética , NF-kappa B/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Fatores de Transcrição
4.
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
5.
Proc Natl Acad Sci U S A ; 111(19): E2037-45, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24782537

RESUMO

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.


Assuntos
Neutrófilos/microbiologia , Proteínas Quinases/genética , Proteínas Quinases/imunologia , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Sequência de Aminoácidos , Proteínas de Bactérias , Membrana Celular/metabolismo , Ativação Enzimática , Imunidade Inata/imunologia , Dados de Sequência Molecular , Neutrófilos/imunologia , Proteínas Quinases/química , Estrutura Terciária de Proteína , Transdução de Sinais/imunologia , Staphylococcus aureus/patogenicidade , Virulência
6.
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
7.
J Leukoc Biol ; 94(5): 971-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24026286

RESUMO

This investigation examines the influence of α-toxin (Hla) expression by CA-MRSA on host immune cell integrity and cytokine expression during infection of human blood. Flow cytometry analysis of human blood infected by Staphylococcus aureus PFGE type USA300 or a USA300Δhla demonstrated that Hla expression significantly increased plasma membrane permeability of human CD14(+) monocytes. The increased susceptibility of human CD14(+) monocytes to Hla toxicity paralleled the high cell-surface expression on these cell types of ADAM10. USA300 rapidly associated with PMNs and monocytes but not T cells following inoculation of human blood. Transcription analysis indicated a strong up-regulation of proinflammatory cytokine transcription following infection of human blood by USA300 and USA300Δhla. CBAs and ELISAs determined that IL-6, IL-10, TNF-α, IFN-γ, IL-1ß, IL-8, and IL-4 are significantly up-regulated during the initial phases of human blood infection by USA300 relative to mock-infected blood but failed to distinguish any significant differences in secreted cytokine protein concentrations during infection by USA300Δhla relative to USA300. Collectively, these findings demonstrate that expression of Hla by USA300 has a significant impact on human CD14(+) monocyte plasma membrane integrity but is not exclusively responsible for the proinflammatory cytokine profile induced by USA300 during the initial stages of human blood infection.


Assuntos
Bacteriemia/imunologia , Toxinas Bacterianas/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Citocinas/genética , Proteínas Hemolisinas/farmacologia , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas/imunologia , Proteínas ADAM/sangue , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/sangue , Humanos , Receptores de Lipopolissacarídeos/sangue , Proteínas de Membrana/sangue
8.
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
9.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA