Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 1399, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658521

RESUMO

Staphylococcus aureus is a prominent human pathogen that readily adapts to host immune defenses. Here, we show that, in contrast to Gram-negative pathogens, S. aureus induces a distinct airway immunometabolic response dominated by the release of the electrophilic metabolite, itaconate. The itaconate synthetic enzyme, IRG1, is activated by host mitochondrial stress, which is induced by staphylococcal glycolysis. Itaconate inhibits S. aureus glycolysis and selects for strains that re-direct carbon flux to fuel extracellular polysaccharide (EPS) synthesis and biofilm formation. Itaconate-adapted strains, as illustrated by S. aureus isolates from chronic airway infection, exhibit decreased glycolytic activity, high EPS production, and proficient biofilm formation even before itaconate stimulation. S. aureus thus adapts to the itaconate-dominated immunometabolic response by producing biofilms, which are associated with chronic infection of the human airway.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/fisiologia , Staphylococcus aureus/patogenicidade , Succinatos/metabolismo , Adulto , Animais , Biofilmes/crescimento & desenvolvimento , Líquido da Lavagem Broncoalveolar , Metabolismo dos Carboidratos , Fibrose Cística/microbiologia , Regulação Bacteriana da Expressão Gênica , Glicólise/efeitos dos fármacos , Glicólise/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Hidroliases/metabolismo , Camundongos Endogâmicos C57BL , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Escarro/microbiologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/isolamento & purificação , Estresse Fisiológico , Succinatos/farmacologia , Ácido Succínico/metabolismo , Adulto Jovem
2.
mBio ; 12(1)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33563839

RESUMO

Respiratory distress in severe malaria is associated with high mortality, but its pathogenesis remains unclear. The malaria pigment hemozoin (HZ) is abundant in target organs of severe malaria, including the lungs, and is known to be a potent innate immune activator of phagocytes. We hypothesized that HZ might also stimulate lung epithelial activation and thereby potentiate lung inflammation. We show here that airway epithelium stimulated with HZ undergoes global transcriptional reprogramming and changes in cell surface protein expression that comprise an epithelial activation phenotype. Proinflammatory signaling is induced, and key cytoadherence molecules are upregulated, including several associated with severe malaria, such as CD36 and ICAM1. Epithelial and extracellular matrix remodeling pathways are transformed, including induction of key metalloproteases and modulation of epithelial junctions. The overall program induced by HZ serves to promote inflammation and neutrophil transmigration, and is recapitulated in a murine model of HZ-induced acute pneumonitis. Together, our data demonstrate a direct role for hemozoin in stimulating epithelial activation that could potentiate lung inflammation in malaria.IMPORTANCE Respiratory distress (RD) is a complication of severe malaria associated with a particularly high risk for death in African children infected with the parasite Plasmodium falciparum The pathophysiology underlying RD remains poorly understood, and the condition is managed supportively. The parasite-derived factor HZ accumulates in target organs of severe malaria, including the lungs, and is a potent stimulator of immune cells. Our findings demonstrate that HZ causes global activation of lung epithelial cells, a response that directly promotes lung inflammation. HZ stimulates expression of key proinflammatory and cell surface molecules, alters signaling pathways involved in epithelial-matrix remodeling, and promotes neutrophil transmigration and airway inflammation. The lung epithelial activation induced by HZ mimics patterns seen in malarial lung injury and provides new insights into the molecular pathogenesis of RD.


Assuntos
Células Epiteliais/efeitos dos fármacos , Hemeproteínas/administração & dosagem , Hemeproteínas/farmacologia , Interações Hospedeiro-Parasita , Inflamação/etiologia , Pulmão/efeitos dos fármacos , Animais , Brônquios/citologia , Antígenos CD36/genética , Linhagem Celular , Movimento Celular , Feminino , Perfilação da Expressão Gênica , Inflamação/genética , Molécula 1 de Adesão Intercelular/genética , Pulmão/imunologia , Pulmão/patologia , Malária Falciparum/complicações , Malária Falciparum/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Plasmodium falciparum/metabolismo , Regulação para Cima
3.
J Clin Invest ; 128(3): 1074-1086, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29431734

RESUMO

Infection by Staphylococcus aureus strain USA300 causes tissue injury, multiorgan failure, and high mortality. However, the mechanisms by which the bacteria adhere to, then stabilize on, mucosal surfaces before causing injury remain unclear. We addressed these issues through the first real-time determinations of USA300-alveolar interactions in live lungs. We found that within minutes, inhaled USA300 established stable, self-associated microaggregates in niches at curved, but not at flat, regions of the alveolar wall. The microaggregates released α-hemolysin toxin, causing localized alveolar injury, as indicated by epithelial dye loss, mitochondrial depolarization, and cytosolic Ca2+ increase. Spread of cytosolic Ca2+ through intercellular gap junctions to adjoining, uninfected alveoli caused pulmonary edema. Systemic pretreatment with vancomycin, a USA300-cidal antibiotic, failed to protect mice infected with inhaled WT USA300. However, vancomycin pretreatment markedly abrogated mortality in mice infected with mutant USA300 that lacked the aggregation-promoting factor PhnD. We interpret USA300-induced mortality as having resulted from rapid bacterial aggregation in alveolar niches. These findings indicate, for the first time to our knowledge, that alveolar microanatomy is critical in promoting the aggregation and, hence, in causing USA300-induced alveolar injury. We propose that in addition to antibiotics, strategies for bacterial disaggregation may constitute novel therapy against USA300-induced lung injury.


Assuntos
Lesão Pulmonar/microbiologia , Lesão Pulmonar/prevenção & controle , Pulmão/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia , Animais , Antibacterianos/farmacologia , Cálcio/metabolismo , Citosol/metabolismo , Junções Comunicantes/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Perfusão , Alvéolos Pulmonares/microbiologia
4.
PLoS One ; 10(3): e0119823, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25798590

RESUMO

The contribution of specific factors to bacterial virulence is generally investigated through creation of genetic "knockouts" that are then compared to wild-type strains or complemented mutants. This paradigm is useful to understand the effect of presence vs. absence of a specific gene product but cannot account for concentration-dependent effects, such as may occur with some bacterial toxins. In order to assess threshold and dose-response effects of virulence factors, robust systems for tunable expression are required. Recent evidence suggests that the folding free energy (ΔG) of the 5' end of mRNA transcripts can have a significant effect on translation efficiency and overall protein abundance. Here we demonstrate that rational alteration of 5' mRNA folding free energy by introduction of synonymous mutations allows for predictable changes in pneumolysin (PLY) expression by Streptococcus pneumoniae without the need for chemical inducers or heterologous promoters. We created a panel of isogenic S. pneumoniae strains, differing only in synonymous (silent) mutations at the 5' end of the PLY mRNA that are predicted to alter ΔG. Such manipulation allows rheostat-like control of PLY production and alters the cytotoxicity of whole S. pneumoniae on primary and immortalized human cells. These studies provide proof-of-principle for further investigation of mRNA ΔG manipulation as a tool in studies of bacterial pathogenesis.


Assuntos
Eritrócitos/metabolismo , Hemólise , Infecções Pneumocócicas/metabolismo , Dobramento de RNA , RNA Mensageiro/genética , Streptococcus pneumoniae/genética , Estreptolisinas/metabolismo , Apoptose , Proteínas de Bactérias/metabolismo , Sequência de Bases , Western Blotting , Proliferação de Células , Células Cultivadas , Eritrócitos/citologia , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Dados de Sequência Molecular , Infecções Pneumocócicas/genética , Infecções Pneumocócicas/microbiologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Streptococcus pneumoniae/isolamento & purificação , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
5.
J Infect Dis ; 211(5): 835-45, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25240171

RESUMO

We postulated that the activation of proinflammatory signaling by methicillin-resistant Staphylococcus aureus (MRSA) strain USA300 is a major factor in the pathogenesis of severe pneumonia and a target for immunomodulation. Local activation of T cells in the lung was a conserved feature of multiple strains of S. aureus, in addition to USA300. The pattern of Vß chain activation was consistent with known superantigens, but deletion of SelX or SEK and SEQ was not sufficient to prevent T-cell activation, indicating the participation of multiple genes. Using Rag2(-/-), Cd4(-/-), and Cd28(-/-) mice, we observed significantly improved clearance of MRSA from the airways and decreased lung pathology, compared with findings for wild-type controls. The improved outcome correlated with decreased production of proinflammatory cytokines (tumor necrosis factor, KC, interleukin 6, and interleukin 1ß). Our data suggest that T-cell-mediated hypercytokinemia induced by infection with MRSA strain USA300 contributes to pathogenesis and may be a therapeutic target for improving outcomes of this common infection in a clinical setting.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Citocinas/metabolismo , Staphylococcus aureus Resistente à Meticilina/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/patologia , Animais , Antígenos CD28/deficiência , Antígenos CD4/genética , Citocinas/sangue , Proteínas de Ligação a DNA/deficiência , Modelos Animais de Doenças , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Superantígenos/genética , Superantígenos/imunologia
6.
PLoS Pathog ; 9(10): e1003682, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098127

RESUMO

The type III interferon (IFNλ) receptor IL-28R is abundantly expressed in the respiratory tract and has been shown essential for host defense against some viral pathogens, however no data are available concerning its role in the innate immune response to bacterial pathogens. Staphylococcus aureus and Pseudomonas aeruginosa induced significant production of IFNλ in the lung, and clearance of these bacteria from the lung was significantly increased in IL-28R null mice compared to controls. Improved bacterial clearance correlated with reduced lung pathology and a reduced ratio of pro- vs anti-inflammatory cytokines in the airway. In human epithelial cells IFNλ inhibited miR-21 via STAT3 resulting in upregulation of PDCD4, a protein known to promote inflammatory signaling. In vivo 18 hours following infection with either pathogen, miR-21 was significantly reduced and PDCD4 increased in the lungs of wild type compared to IL-28R null mice. Infection of PDCD4 null mice with USA300 resulted in improved clearance, reduced pathology, and reduced inflammatory cytokine production. These data suggest that during bacterial pneumonia IFNλ promotes inflammation by inhibiting miR-21 regulation of PDCD4.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Citocinas/metabolismo , Pneumonia Estafilocócica/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas de Ligação a RNA/biossíntese , Mucosa Respiratória/metabolismo , Staphylococcus aureus/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular , Citocinas/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Pneumonia Estafilocócica/genética , Pneumonia Estafilocócica/patologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/genética , Proteínas de Ligação a RNA/genética , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia , Staphylococcus aureus/genética
7.
J Clin Invest ; 123(4): 1630-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23478406

RESUMO

The respiratory tract is exceptionally well defended against infection from inhaled bacteria, with multiple proinflammatory signaling cascades recruiting phagocytes to clear airway pathogens. However, organisms that efficiently activate damaging innate immune responses, such as those mediated by the inflammasome and caspase-1, may cause pulmonary damage and interfere with bacterial clearance. The extracellular, opportunistic pathogen Pseudomonas aeruginosa expresses not only pathogen-associated molecular patterns that activate NF-κB signaling in epithelial and immune cells, but also flagella that activate the NLRC4 inflammasome. We demonstrate that induction of inflammasome signaling, ascribed primarily to the alveolar macrophage, impaired P. aeruginosa clearance and was associated with increased apoptosis/pyroptosis and mortality in a murine model of acute pneumonia. Strategies that limited inflammasome activation, including infection by fliC mutants, depletion of macrophages, deletion of NLRC4, reduction of IL-1ß and IL-18 production, inhibition of caspase-1, and inhibition of downstream signaling in IL-1R- or IL-18R-null mice, all resulted in enhanced bacterial clearance and diminished pathology. These results demonstrate that the inflammasome provides a potential target to limit the pathological consequences of acute P. aeruginosa pulmonary infection.


Assuntos
Inflamassomos/imunologia , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/fisiologia , Doença Aguda , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Ácido Clodrônico/farmacologia , Flagelina/imunologia , Técnicas de Inativação de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Interações Hospedeiro-Patógeno , Inflamassomos/metabolismo , Interferon beta/fisiologia , Interleucina-17/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Depuração Mucociliar , Neutrófilos/imunologia , Neutrófilos/microbiologia , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Transdução de Sinais
8.
J Infect Dis ; 206(1): 81-90, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22535996

RESUMO

Staphylococcus aureus protein A (SpA) plays a critical role in the induction of inflammation. This study was aimed to determine whether the number of short sequence repeats (SSRs) present in the polymorphic region modulates the inflammatory response induced by SpA. We demonstrated that there is a dose-response effect in the activation of interferon (IFN)-ß signaling in airway epithelial and immune cells, depending on the number of SSRs, which leads to differences in neutrophil recruitment. We also determined that a significant proportion of isolates from patients with chronic infections such as osteomyelitis and cystic fibrosis carry fewer SSRs than do isolates from patients with acute infections or healthy carriers and that there was an inverse correlation between the number of SSRs and the length of disease course. Given the importance of IFN signaling in eradication of S. aureus, loss of SSRs may represent an advantageous mechanism to adapt to and persist in the host.


Assuntos
Inflamação/genética , Infecções Estafilocócicas/microbiologia , Proteína Estafilocócica A/metabolismo , Adolescente , Adulto , Animais , Linhagem Celular , Criança , Pré-Escolar , Doença Crônica , Fibrose Cística/genética , Fibrose Cística/imunologia , Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Relação Dose-Resposta Imunológica , Humanos , Lactente , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Interferon beta/imunologia , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Repetições de Microssatélites , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Osteomielite/genética , Osteomielite/imunologia , Osteomielite/metabolismo , Osteomielite/microbiologia , Polimorfismo Genético , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Adulto Jovem
9.
J Bacteriol ; 190(8): 2814-21, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18165298

RESUMO

The type III secreted toxins of Pseudomonas aeruginosa are important virulence factors associated with clinically important infection. However, their effects on bacterial invasion across mucosal surfaces have not been well characterized. One of the most commonly expressed toxins, ExoS, has two domains that are predicted to affect cytoskeletal integrity, including a GTPase-activating protein (GAP) domain, which targets Rho, a major regulator of actin polymerization; and an ADP-ribosylating domain that affects the ERM proteins, which link the plasma membrane to the actin cytoskeleton. The activities of these toxins, and ExoS specifically, on the permeability properties of polarized airway epithelial cells with intact tight junctions were examined. Strains expressing type III toxins altered the distribution of the tight junction proteins ZO-1 and occludin and were able to transmigrate across polarized airway epithelial monolayers, in contrast to DeltaSTY mutants. These effects on epithelial permeability were associated with the ADP-ribosylating domain of ExoS, as bacteria expressing plasmids lacking expression of the ExoS GAP activity nonetheless increased the permeation of fluorescent dextrans, as well as bacteria, across polarized airway epithelial cells. Treatment of epithelial cells with cytochalasin D depolymerized actin filaments and increased permeation across the monolayers but did not eliminate the differential effects of wild-type and toxin-negative mutants on the epithelial cells, suggesting that additional epithelial targets are involved. Confocal imaging studies demonstrated that ZO-1, occludin, and ezrin undergo substantial redistribution in human airway cells intoxicated by ExoS, -T, and -Y. These studies support the hypothesis that type III toxins enhance P. aeruginosa's invasive capabilities by interacting with multiple eukaryotic cytoskeletal components.


Assuntos
ADP Ribose Transferases/toxicidade , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Células Epiteliais/microbiologia , Proteínas Ativadoras de GTPase/toxicidade , Glucosiltransferases/toxicidade , Pseudomonas aeruginosa/patogenicidade , Junções Íntimas/fisiologia , ADP Ribose Transferases/química , ADP Ribose Transferases/genética , Proteínas de Bactérias/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Linhagem Celular , Proteínas do Citoesqueleto/análise , Células Epiteliais/química , Células Epiteliais/efeitos dos fármacos , Deleção de Genes , Humanos , Proteínas de Membrana/análise , Ocludina , Permeabilidade , Fosfoproteínas/análise , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/crescimento & desenvolvimento , Proteína da Zônula de Oclusão-1
10.
EMBO J ; 26(3): 701-9, 2007 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-17255933

RESUMO

Among the many adhesins and toxins expressed by Staphylococcus aureus, protein A is an exceptionally complex virulence factor, known to interact with multiple eukaryotic targets, particularly those with immunological functions. Protein A acts as a ligand that can mimic TNF-alpha to activate TNFR1 and subsequent proinflammatory signaling. It also stimulates the cleavage of TNFR1 from the surface of epithelial cells and macrophages, which serves to limit TNF-alpha signaling. We characterized the signaling pathway responsible for TNFR1 shedding and identified protein A mutants which could activate TNFR1-dependent signaling, but were unable to activate TACE, the TNFR1 sheddase. Activation of TACE was dependent upon a discrete interaction between the previously defined IgG-binding domain of protein A and the epidermal growth factor receptor (EGFR), which in turn induced TACE phosphorylation through a c-Src-erk1/2-mediated cascade. This novel interaction was independent of the autocrine activation of EGFR and protein A-induced TGF-alpha was neither required nor sufficient to activate TNFR1 shedding. Thus, staphylococci exploit the ubiquitous and multifunctional EGFR to regulate the availability of TNFR1 on mucosal and immune cells.


Assuntos
Proteínas ADAM/metabolismo , Receptores ErbB/metabolismo , Transdução de Sinais/fisiologia , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/genética , Fatores de Virulência/metabolismo , Proteína ADAM17 , Western Blotting , Primers do DNA , Ativação Enzimática/fisiologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunoprecipitação , Interleucina-8/metabolismo , Fosforilação , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Proteína Estafilocócica A/genética , Staphylococcus aureus/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Fatores de Virulência/genética
11.
J Biol Chem ; 281(29): 20190-6, 2006 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16709567

RESUMO

Staphylococcus aureus continues to be a major cause of infection in normal as well as immunocompromised hosts, and the increasing prevalence of highly virulent community-acquired methicillin-resistant strains is a public health concern. A highly expressed surface component of S. aureus, protein A (SpA), contributes to its success as a pathogen by both activating inflammation and by interfering with immune clearance. SpA is known to bind to IgG Fc, which impedes phagocytosis. SpA is also a potent activator of tumor necrosis factor alpha (TNF-alpha) receptor 1 (TNFR1) signaling, inducing both chemokine expression and TNF-converting enzyme-dependent soluble TNFR1 (sTNFR1) shedding, which has anti-inflammatory consequences, particularly in the lung. Using a collection of glutathione S-transferase fusions to the intact IgG binding region of SpA and to each of the individual binding domains, we found that the SpA IgG binding domains also mediate binding to human airway cells. TNFR1-dependent CXCL8 production could be elicited by any one of the individual SpA IgG binding domains as efficiently as by either the entire SpA or the intact IgG binding region. SpA induction of sTNFR1 shedding required the entire IgG binding region and tolerated fewer substitutions in residues known to interact with IgG. Each of the repeated domains of the IgG binding domain can affect multiple immune responses independently, activating inflammation through TNFR1 and thwarting opsonization by trapping IgG Fc domains, while the intact IgG binding region can limit further signaling through sTNFR1 shedding.


Assuntos
Imunoglobulina G/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Proteína Estafilocócica A/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Clonagem Molecular , Escherichia coli/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Estrutura Secundária de Proteína , Receptores Tipo I de Fatores de Necrose Tumoral/efeitos dos fármacos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes/farmacologia , Mucosa Respiratória/fisiologia , Proteína Estafilocócica A/química , Proteína Estafilocócica A/genética
12.
Am J Physiol Lung Cell Mol Physiol ; 291(3): L297-300, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16648241

RESUMO

The symposium addressed the burgeoning interest in fundamental mechanisms underlying the onset of pneumonia. Bacteria exploit the lung's innate immune mechanism, resulting in pathophysiological cell signaling. As a consequence inflammation develops, leading to pneumonia. New mechanisms have been identified by which bacteria or bacterial products in the airway induce cross-compartmental signaling that leads to inflammatory consequences. The speakers addressed activation of the transcription factor, NF-kappaB occurring as a consequence of bacterial interactions with specific receptors, such as the Toll-like receptors and the TNF receptor 1 (Prince), or as a consequence of cytokine induction (Mizgerd). Also considered were mechanisms of bacterial virulence in the clinical setting (Wiener-Kronish) and the role of alveolar-capillary signaling mechanisms in the initiation of lung inflammation.


Assuntos
Citocinas/metabolismo , Pneumonia/etiologia , Alvéolos Pulmonares/microbiologia , Transdução de Sinais , Comunicação Celular , Epitélio/metabolismo , Interações Hospedeiro-Parasita , Humanos , Modelos Biológicos , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/microbiologia , Infecções por Pseudomonas/genética , Pseudomonas aeruginosa/genética , Alvéolos Pulmonares/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
13.
J Immunol ; 175(3): 1930-6, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16034137

RESUMO

Airway epithelial cells have a major role in initiating inflammation in response to bacterial pathogens. Through the immediate induction of CXCL8 and cytokine expression, polymorphonuclear cells are mobilized and activated to eradicate the infecting organisms. However, the influx of polymorphonuclear cells and the effects of their toxic exoproducts impede respiratory function. We postulated that respiratory epithelial cells must also participate in the regulation of their own proinflammatory signaling. Both Staphylococcus aureus and Pseudomonas aeruginosa were found to potently activate IL-6 expression immediately upon contact with epithelial cells, and by 1 h induced TNF-alpha converting enzyme (TACE) transcription. By 4 h of bacterial exposure, TACE colocalized with IL-6Ralpha on the apical surface of airway cells, and by 24 h, soluble IL-6Ralpha accumulated in the cell culture supernatant. Epithelial IL-6 and soluble IL-6Ralpha were shown to participate in trans-signaling, interacting with membrane-associated gp130 to activate CCL-2 expression and inhibit additional CXCL8 production. Thus, bacteria are physiological activators of TACE expression, which provides a mechanism to regulate inflammatory signaling that is initiated by airway epithelial cells.


Assuntos
Mediadores da Inflamação/fisiologia , Interleucina-6 , Metaloendopeptidases/biossíntese , Pseudomonas aeruginosa/imunologia , Receptores de Interleucina-6/metabolismo , Mucosa Respiratória/imunologia , Transdução de Sinais/imunologia , Staphylococcus aureus/imunologia , Proteínas ADAM , Proteína ADAM17 , Brônquios/imunologia , Brônquios/microbiologia , Brônquios/patologia , Linhagem Celular , Quimiocina CCL2/biossíntese , Ativação Enzimática/imunologia , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/biossíntese , Interleucina-6/fisiologia , Interleucina-8/biossíntese , Metaloendopeptidases/metabolismo , Pólipos Nasais/enzimologia , Pólipos Nasais/imunologia , Pólipos Nasais/microbiologia , Pólipos Nasais/patologia , Receptores de Interleucina-6/biossíntese , Receptores de Interleucina-6/fisiologia , Mucosa Respiratória/enzimologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia , Solubilidade , Especificidade por Substrato/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/fisiologia
14.
Am J Respir Crit Care Med ; 171(11): 1209-23, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15695491

RESUMO

Pseudomonas aeruginosa is an important pathogen causing a wide range of acute and chronic infections. P. aeruginosa rarely causes infection in the normal host, but is an efficient opportunistic pathogen causing serious infections in patients who are mechanically ventilated, individuals who are immunocompromised, and patients with malignancies or HIV infection. Among these risk groups, the most vulnerable hosts are neutropenic and patients who are mechanically ventilated. In addition, P. aeruginosa is the most prevalent chronic infection contributing to the pathogenesis of cystic fibrosis. Because of the ubiquitous nature of P. aeruginosa and its ability to develop resistance to antibiotics, it continues to be problematic from a treatment perspective. The pathogenicity of P. aeruginosa is largely caused by multiple bacterial virulence factors and genetic flexibility enabling it to survive in varied environments. Lung injury associated with P. aeruginosa infection results from both the direct destructive effects of the organism on the lung parenchyma and exuberant host immune responses. This article focuses on the major bacterial virulence factors and important aspects of the host immunity that are involved in the pathogenesis of serious P. aeruginosa infection. In addition to antibiotic therapy, strategies directed toward enhancing host defense and/or limiting excessive inflammation could be important to improve outcome in P. aeruginosa lung infections.


Assuntos
Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Doença Aguda , Doença Crônica , Infecção Hospitalar/microbiologia , Fibrose Cística/complicações , Interações Hospedeiro-Parasita/imunologia , Humanos , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia
15.
J Clin Invest ; 112(5): 656-8, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12952914

RESUMO

Bacillus anthracis, the causative agent of anthrax, is believed to induce disease and death in humans in an endotoxic shock-like manner. A comprehensive study of the effects of anthrax toxin in mice demonstrates that toxin-induced death is mediated not by cytokine release, as previously thought, but by hypoxia-induced liver failure. The study strongly suggests that the therapies developed for treatment of cytokine-mediated septic shock will not be appropriate for the treatment of anthrax.


Assuntos
Antígenos de Bactérias , Toxinas Bacterianas/toxicidade , Fator de Necrose Tumoral alfa/fisiologia , Animais , Antraz/complicações , Antraz/etiologia , Camundongos , Plasmídeos , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA