Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Infect Dis ; 212(1): 95-105, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25525050

RESUMO

BACKGROUND: Streptococcus suis has emerged as an important cause of bacterial meningitis in adults. The ingestion of undercooked pork is a risk factor for human S. suis serotype 2 (SS2) infection. Here we provide experimental evidence indicating that the gastrointestinal tract is an entry site of SS2 infection. METHODS: We developed a noninvasive in vivo model to study oral SS2 infection in piglets. We compared in vitro interaction of S. suis with human and porcine intestinal epithelial cells (IEC). RESULTS: Two out of 15 piglets showed clinical symptoms compatible with S. suis infection 24-48 hours after ingestion of SS2. SS2 was detected in mesenteric lymph nodes of 40% of challenged piglets. SS2 strains isolated from patients showed significantly higher adhesion to human IEC compared to invasive strains isolated from pigs. In contrast, invasive SS9 strains showed significantly higher adhesion to porcine IEC. Translocation across human IEC, which occurred predominately via a paracellular route, was significantly associated with clonal complex 1, the predominant zoonotic genotype. Adhesion and translocation were dependent on capsular polysaccharide production. CONCLUSIONS: SS2 should be considered a food-borne pathogen. S. suis interaction with human and pig IEC correlates with S. suis serotype and genotype, which can explain the zoonotic potential of SS2.


Assuntos
Interações Hospedeiro-Patógeno , Mucosa Intestinal/microbiologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Zoonoses/microbiologia , Adulto , Animais , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Humanos , Masculino , Meningites Bacterianas/microbiologia , Meningites Bacterianas/veterinária , Suínos
3.
PLoS Pathog ; 9(12): e1003816, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24348255

RESUMO

Upon contact with human plasma, bacteria are rapidly recognized by the complement system that labels their surface for uptake and clearance by phagocytic cells. Staphylococcus aureus secretes the 16 kD Extracellular fibrinogen binding protein (Efb) that binds two different plasma proteins using separate domains: the Efb N-terminus binds to fibrinogen, while the C-terminus binds complement C3. In this study, we show that Efb blocks phagocytosis of S. aureus by human neutrophils. In vitro, we demonstrate that Efb blocks phagocytosis in plasma and in human whole blood. Using a mouse peritonitis model we show that Efb effectively blocks phagocytosis in vivo, either as a purified protein or when produced endogenously by S. aureus. Mutational analysis revealed that Efb requires both its fibrinogen and complement binding residues for phagocytic escape. Using confocal and transmission electron microscopy we show that Efb attracts fibrinogen to the surface of complement-labeled S. aureus generating a 'capsule'-like shield. This thick layer of fibrinogen shields both surface-bound C3b and antibodies from recognition by phagocytic receptors. This information is critical for future vaccination attempts, since opsonizing antibodies may not function in the presence of Efb. Altogether we discover that Efb from S. aureus uniquely escapes phagocytosis by forming a bridge between a complement and coagulation protein.


Assuntos
Proteínas de Bactérias/metabolismo , Complemento C3b/metabolismo , Fibrinogênio/metabolismo , Evasão da Resposta Imune , Fagocitose/imunologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Animais , Fatores de Coagulação Sanguínea/metabolismo , Células Cultivadas , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo
4.
Cell Host Microbe ; 13(5): 584-594, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23684309

RESUMO

Panton-Valentine Leukocidin (PVL) is a staphylococcal bicomponent pore-forming toxin linked to severe invasive infections. Target-cell and species specificity of PVL are poorly understood, and the mechanism of action of this toxin in Staphylococcus aureus virulence is controversial. Here, we identify the human complement receptors C5aR and C5L2 as host targets of PVL, mediating both toxin binding and cytotoxicity. Expression and interspecies variations of the C5aR determine cell and species specificity of PVL. The C5aR binding PVL component, LukS-PV, is a potent inhibitor of C5a-induced immune cell activation. These findings provide insight into leukocidin function and staphylococcal virulence and offer directions for future investigations into individual susceptibility to severe staphylococcal disease.


Assuntos
Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Interações Hospedeiro-Patógeno , Leucocidinas/metabolismo , Receptor da Anafilatoxina C5a/metabolismo , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/toxicidade , Células Cultivadas , Exotoxinas/toxicidade , Humanos , Leucocidinas/toxicidade , Receptores de Quimiocinas/metabolismo
5.
EMBO J ; 31(17): 3607-19, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22850671

RESUMO

The CXC chemokine receptor 2 (CXCR2) on neutrophils, which recognizes chemokines produced at the site of infection, plays an important role in antimicrobial host defenses such as neutrophil activation and chemotaxis. Staphylococcus aureus is a successful human pathogen secreting a number of proteolytic enzymes, but their influence on the host immune system is not well understood. Here, we identify the cysteine protease Staphopain A as a chemokine receptor blocker. Neutrophils treated with Staphopain A are unresponsive to activation by all unique CXCR2 chemokines due to cleavage of the N-terminal domain, which can be neutralized by specific protease inhibitors. Moreover, Staphopain A inhibits neutrophil migration towards CXCR2 chemokines. By comparing a methicillin-resistant S. aureus (MRSA) strain with an isogenic Staphopain A mutant, we demonstrate that Staphopain A is the only secreted protease with activity towards CXCR2. Although the inability to cleave murine CXCR2 limits in-vivo studies, our data indicate that Staphopain A is an important immunomodulatory protein that blocks neutrophil recruitment by specific cleavage of the N-terminal domain of human CXCR2.


Assuntos
Proteínas de Bactérias/imunologia , Cisteína Endopeptidases/imunologia , Neutrófilos/imunologia , Receptores de Interleucina-8B/imunologia , Animais , Células Cultivadas , Quimiotaxia de Leucócito/imunologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ativação de Neutrófilo/imunologia , Infiltração de Neutrófilos/imunologia , Receptores de Interleucina-8B/antagonistas & inibidores , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA