Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Eur J Immunol ; 45(2): 383-95, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25378230

RESUMO

Peptides presented by MHC class I molecules are mostly derived from proteins synthesized by the antigen-presenting cell itself, while peptides presented by MHC class II molecules are predominantly from materials acquired by endocytosis. External antigens can also be presented by MHC class I molecules in a process referred to as cross-presentation. Here, we report that mouse dendritic cell (DC) engagement to a phagocytic target alters endocytic processing and inhibits the proteolytic activities. During phagocytosis, endosome maturation is delayed, shows less progression toward the lysosome, and the endocytosed soluble antigen is targeted for MHC class I cross-presentation. The antigen processing in these arrested endosomes is under the control of NAPDH oxidase associated ROS. We also show that cathepsin S is responsible for the generation of the MHC class I epitope. Taken together, our results suggest that in addition to solid structure uptake, DC phagocytosis simultaneously modifies the kinetics of endosomal trafficking and maturation. As a consequence, external soluble antigens are targeted into the MHC class I cross-presentation pathway.


Assuntos
Apresentação de Antígeno , Apresentação Cruzada , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Fagocitose , Animais , Catepsinas/imunologia , Catepsinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Endocitose , Endossomos/imunologia , Endossomos/metabolismo , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Lisossomos/imunologia , Lisossomos/metabolismo , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/imunologia , Complexos Multienzimáticos/metabolismo , NADH NADPH Oxirredutases/imunologia , NADH NADPH Oxirredutases/metabolismo , Ovalbumina/imunologia , Ovalbumina/farmacologia , Cultura Primária de Células , Espécies Reativas de Oxigênio/metabolismo
2.
Cell Microbiol ; 17(12): 1883-99, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26119044

RESUMO

Plasmodium falciparum-infected erythrocytes (IRBC) expressing the domain cassettes (DC) 8 and 13 of the cytoadherent ligand P. falciparum erythrocyte membrane protein 1 adhere to the endothelial protein C receptor (EPCR). By interfering with EPCR anti-coagulant and pro-endothelial barrier functions, IRBC adhesion could promote coagulation and vascular permeability that contribute to the pathogenesis of cerebral malaria. In this study, we examined the adhesion of DC8- and DC13-expressing parasite lines to endothelial cells from different microvasculature, and the consequences of EPCR engagement on endothelial cell function. We found that IRBC from IT4var19 (DC8) and IT4var07 (DC13) parasite lines adhered to human brain, lung and dermal endothelial cells under shear stress. However, the relative contribution of EPCR to parasite cytoadherence on different types of endothelial cell varied. We also observed divergent functional outcomes for DC8 cysteine-rich interdomain region (CIDR)α1.1 and DC13 CIDRα1.4 domains. IT4var07 CIDRα1.4 inhibited generation of activated protein C (APC) on lung and dermal endothelial cells and blocked the APC-EPCR binding interaction on brain endothelial cells. IT4var19 CIDRα1.1 inhibited thrombin-induced endothelial barrier dysfunction in lung endothelial cells, whereas IT4var07 CIDRα1.4 inhibited the protective effect of APC on thrombin-induced permeability. Overall, these findings reveal a much greater complexity of how CIDRα1-expressing parasites may modulate malaria pathogenesis through EPCR adhesion.


Assuntos
Antígenos CD/metabolismo , Adesão Celular , Células Endoteliais/fisiologia , Eritrócitos/parasitologia , Interações Hospedeiro-Patógeno , Plasmodium falciparum/fisiologia , Receptores de Superfície Celular/metabolismo , Células Cultivadas , Receptor de Proteína C Endotelial , Humanos , Ligadura , Resultado do Tratamento
3.
Infect Immun ; 83(12): 4571-81, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26371123

RESUMO

Campylobacter jejuni is the most common cause of bacterium-induced gastroenteritis, and while typically self-limiting, C. jejuni infections are associated with postinfectious intestinal disorders, including flares in patients with inflammatory bowel disease and postinfectious irritable bowel syndrome (PI-IBS), via mechanisms that remain obscure. Based on the hypothesis that acute campylobacteriosis may cause pathogenic microbiota dysbiosis, we investigated whether C. jejuni may activate dormant virulence genes in noninvasive Escherichia coli and examined the epithelial pathophysiological consequences of these alterations. Microarray and quantitative real-time PCR analyses revealed that E. coli adhesin, flagellum, and hemolysin gene expression were increased when E. coli was exposed to C. jejuni-conditioned medium. Increased development of bacterial flagella upon exposure to live C. jejuni or C. jejuni-conditioned medium was observed under transmission electron microscopy. Atomic force microscopy demonstrated that the forces of bacterial adhesion to colonic T84 enterocytes, and the work required to rupture this adhesion, were significantly increased in E. coli exposed to C. jejuni-conditioned media. Finally, C. jejuni-modified E. coli disrupted TLR4 gene expression and induced proinflammatory CXCL-8 gene expression in colonic enterocytes. Together, these data suggest that exposure to live C. jejuni, and/or to its secretory-excretory products, may activate latent virulence genes in noninvasive E. coli and that these alterations may directly trigger proinflammatory signaling in intestinal epithelia. These observations shed new light on mechanisms that may contribute, at least in part, to postcampylobacteriosis inflammatory disorders.


Assuntos
Campylobacter jejuni/metabolismo , Meios de Cultivo Condicionados/farmacologia , Enterócitos/efeitos dos fármacos , Interleucina-8/imunologia , Receptor 4 Toll-Like/imunologia , Campylobacter jejuni/patogenicidade , Linhagem Celular , Enterócitos/imunologia , Enterócitos/microbiologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Flagelos/efeitos dos fármacos , Flagelos/genética , Flagelos/metabolismo , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Interleucina-8/agonistas , Interleucina-8/genética , Transdução de Sinais , Simbiose , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética , Virulência
4.
PLoS Pathog ; 9(8): e1003590, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24009511

RESUMO

The adhesion of Plasmodium falciparum-infected erythrocytes (IRBC) to receptors on different host cells plays a divergent yet critical role in determining the progression and outcome of the infection. Based on our ex vivo studies with clinical parasite isolates from adult Thai patients, we have previously proposed a paradigm for IRBC cytoadherence under physiological shear stress that consists of a recruitment cascade mediated largely by P-selectin, ICAM-1 and CD36 on primary human dermal microvascular endothelium (HDMEC). In addition, we detected post-adhesion signaling events involving Src family kinases and the adaptor protein p130CAS in endothelial cells that lead to CD36 clustering and cytoskeletal rearrangement which enhance the magnitude of the adhesive strength, allowing adherent IRBC to withstand shear stress of up to 20 dynes/cm². In this study, we addressed whether CD36 supports IRBC adhesion as part of an assembly of membrane receptors. Using a combination of flow chamber assay, atomic force and confocal microscopy, we showed for the first time by loss- and gain-of function assays that in the resting state, the integrin α5ß1 does not support adhesive interactions between IRBC and HDMEC. Upon IRBC adhesion to CD36, the integrin is recruited either passively as part of a molecular complex with CD36, or actively to the site of IRBC attachment through phosphorylation of Src family kinases, a process that is Ca²âº-dependent. Clustering of ß1 integrin is associated with an increase in IRBC recruitment as well as in adhesive strength after attachment (∼40% in both cases). The adhesion of IRBC to a multimolecular complex on the surface of endothelial cells could be of critical importance in enabling adherent IRBC to withstand the high shear stress in the microcirculations. Targeting integrins may provide a novel approach to decrease IRBC cytoadherence to microvascular endothelium.


Assuntos
Antígenos CD36/metabolismo , Endotélio Vascular/metabolismo , Eritrócitos/metabolismo , Integrina alfa5beta1/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Antígenos CD36/genética , Cálcio/metabolismo , Adesão Celular/genética , Células Cultivadas , Endotélio Vascular/patologia , Eritrócitos/parasitologia , Eritrócitos/patologia , Feminino , Humanos , Integrina alfa5beta1/genética , Malária Falciparum/genética , Malária Falciparum/patologia , Masculino , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fosforilação/genética , Plasmodium falciparum/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
5.
Am J Pathol ; 180(3): 1028-1039, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22260922

RESUMO

Plasmodium falciparum is a protozoan parasite of human erythrocytes that causes the most severe form of malaria. Severe P. falciparum infection is associated with endothelial activation and permeability, which are important determinants of the outcome of the infection. How endothelial cells become activated is not fully understood, particularly with regard to the effects of parasite subcomponents. We demonstrated that P. falciparum histones extracted from merozoites (HeH) directly stimulated the production of IL-8 and other inflammatory mediators by primary human dermal microvascular endothelial cells through a signaling pathway that involves Src family kinases and p38 MAPK. The stimulatory effect of HeH and recombinant P. falciparum H3 (PfH3) was abrogated by histone-specific antibodies. The release of nuclear contents on rupture of infected erythrocytes was captured by live cell imaging and confirmed by detecting nucleosomes in the supernatants of parasite cultures. HeH and recombinant parasite histones also induced endothelial permeability through a charge-dependent mechanism that resulted in disruption of junctional protein expression and cell death. Recombinant human activated protein C cleaved HeH and PfH3 and abrogated their proinflammatory effects. Circulating nucleosomes of both human and parasite origin were detected in the plasma of patients with falciparum malaria and correlated positively with disease severity. These results support a pathogenic role for both host- and pathogen-derived histones in P. falciparum-caused malaria.


Assuntos
Endotélio Vascular/metabolismo , Histonas/farmacologia , Interleucina-8/biossíntese , Merozoítos , Plasmodium falciparum , Animais , Permeabilidade Capilar/fisiologia , Células Cultivadas , Endotélio Vascular/parasitologia , Humanos , Estágios do Ciclo de Vida , Pulmão/irrigação sanguínea , Pulmão/parasitologia , Sistema de Sinalização das MAP Quinases/fisiologia , Malária Falciparum/parasitologia , Microvasos , Proteína C/farmacologia , Proteínas Recombinantes , Pele/irrigação sanguínea , Pele/parasitologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Quinases da Família src/fisiologia
6.
Am J Pathol ; 180(3): 1308-1323, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22203054

RESUMO

Increased permeability of the microvascular endothelium to fluids and proteins is the hallmark of inflammatory conditions such as sepsis. Leakage can occur between (paracellular) or through (transcytosis) endothelial cells, yet little is known about whether these pathways are linked. Understanding the regulation of microvascular permeability is essential for the identification of novel therapies to combat inflammation. We investigated whether transcytosis and paracellular leakage are co-regulated. Using molecular and pharmacologic approaches, we inhibited transcytosis of albumin in primary human microvascular endothelium and measured paracellular permeability. Blockade of transcytosis induced a rapid increase in paracellular leakage that was not explained by decreases in caveolin-1 or increases in activity of nitric oxide synthase. The effect required caveolin-1 but was observed in cells depleted of clathrin, indicating that it was not due to the general inhibition of endocytosis. Inhibiting transcytosis by dynamin blockade increased paracellular leakage concomitantly with the loss of cortical actin from the plasma membrane and the displacement of active Rac from the plasmalemma. Importantly, inhibition of paracellular leakage by sphingosine-1-phosphate, which activates Rac and induces cortical actin, caused a significant increase in transcytosis of albumin in vitro and in an ex vivo whole-lung model. In addition, dominant-negative Rac significantly diminished albumin uptake by endothelia. Our findings indicate that transcytosis and paracellular permeability are co-regulated through a signaling pathway linking dynamin, Rac, and actin.


Assuntos
Albuminas/farmacocinética , Permeabilidade Capilar/fisiologia , Dinaminas/antagonistas & inibidores , Endotélio Vascular/metabolismo , Transcitose/fisiologia , Proteínas rac de Ligação ao GTP/antagonistas & inibidores , Citoesqueleto de Actina/fisiologia , Animais , Caveolina 1/metabolismo , Conexinas/metabolismo , Células Endoteliais/metabolismo , Glicocálix/metabolismo , Humanos , Hidrazonas/farmacologia , Lisofosfolipídeos/farmacologia , Camundongos , Microvasos , Proteínas SNARE/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Transcitose/efeitos dos fármacos , Proteínas rac de Ligação ao GTP/metabolismo
7.
FASEB J ; 26(3): 1119-30, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22106368

RESUMO

The adhesion of infected red blood cells (IRBCs) to microvascular endothelium is critical in the pathogenesis of severe malaria. Here we used atomic force and confocal microscopy to examine the adhesive forces between IRBCs and human dermal microvascular endothelial cells. Initial contact of the cells generated a mean ± sd adhesion force of 167 ± 208 pN from the formation of single or multiple bonds with CD36. The strength of adhesion increased by 5- to 6-fold within minutes of contact through a signaling pathway initiated by CD36 ligation by live IRBCs, or polystyrene beads coated with anti-CD36 or PpMC-179, a recombinant peptide representing the minimal binding domain of the parasite ligand PfEMP1 to CD36. Engagement of CD36 led to localized phosphorylation of Src family kinases and the adaptor protein p130CAS, resulting in actin recruitment and CD36 clustering by 50-60% of adherent beads. Uninfected red blood cells or IgG-coated beads had no effect. Inhibition of the increase in adhesive strength by the Src family kinase inhibitor PP1 or gene silencing of p130CAS decreased adhesion by 39 ± 12 and 48 ± 20%, respectively, at 10 dyn/cm(2) in a flow chamber assay. Modulation of adhesive strength at PfEMP1-CD36-actin cytoskeleton synapses could be a novel target for antiadhesive therapy.


Assuntos
Antígenos CD36/metabolismo , Proteína Substrato Associada a Crk/metabolismo , Citoesqueleto/metabolismo , Eritrócitos/metabolismo , Plasmodium falciparum/metabolismo , Actinas/metabolismo , Antígenos CD36/genética , Adesão Celular/efeitos dos fármacos , Comunicação Celular , Células Cultivadas , Proteína Substrato Associada a Crk/genética , Células Endoteliais/metabolismo , Eritrócitos/parasitologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interações Hospedeiro-Parasita , Humanos , Recém-Nascido , Masculino , Microscopia de Força Atômica , Microscopia Confocal , Fosforilação , Plasmodium falciparum/fisiologia , Ligação Proteica , Proteínas de Protozoários/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , Análise de Célula Única/métodos , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
8.
Eur J Immunol ; 40(6): 1639-50, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20306471

RESUMO

The response of leukocytes to lipoteichoic acid (LTA), a TLR2-dependent major cell wall component of Staphylococcus aureus, is linked to the outcome of an infection. In this study we investigated the role of nonhematopoietic TLR2 in response to LTA and S. aureus by creating bone marrow chimeras. Significant leukocyte recruitment in response to LTA required IFN-gamma priming in WT C57BL/6 and TLR2(-/-)-->WT mice, but was not observed in TLR2(-/-) or WT-->TLR2(-/-) animals. LTA also induced a proinflammatory response in IFN-gamma primed primary human microvascular endothelial cells leading to leukocyte recruitment in vitro. When mice were infected with S. aureus, the most profound elevation of TNF-alpha and IL-6 was seen in TLR2(-/-) and TLR2(-/-)-->WT mice. TLR2(-/-), but not chimeric mice, demonstrated increased IL-17, blood leukocytosis and pulmonary neutrophilia compared to WT mice. Collectively, the results suggest an essential role for IFN-gamma and nonhematopoietic TLR2 for leukocyte recruitment in response to LTA. In contrast, TLR2 on both hematopoietic and nonhematopoietic cells appears to orchestrate an inhibitory response to S. aureus such that in complete TLR2 deficiency, there is an exaggerated proinflammatory response and/or skewing of the immune response towards a Th17 phenotype that may contribute to the decreased survival of TLR2(-/-) mice.


Assuntos
Quimiotaxia de Leucócito/imunologia , Lipopolissacarídeos/imunologia , Infecções Estafilocócicas/imunologia , Ácidos Teicoicos/imunologia , Receptor 2 Toll-Like/imunologia , Animais , Células Endoteliais/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/deficiência , Quimeras de Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA