Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
Intervalo de ano de publicação
1.
Cell Microbiol ; 16(9): 1339-53, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24617613

RESUMO

Neutrophil (polymorphonuclear leucocytes; PMN) transmigration across mucosal surfaces contributes to dysfunction of epithelial barrier properties, a characteristic underlying many mucosal inflammatory diseases. Using Salmonella enterica serovar Typhimurium (S. Typhimurium) as a prototypic proinflammatory insult, we have previously reported that the eicosanoid hepoxilin A3 (HXA3 ), an endogenous product of 12-lipoxygenase (12-LOX) activity, is secreted from the apical surface of the intestinal epithelium to establish a chemotactic gradient that guides PMN across the epithelial surface. Since little is known regarding the molecular mechanisms that regulate 12-LOX during S. Typhimurium infection, we investigated this pathway. We found that expression of phospholipid glutathione peroxidase (GPX4), which is known to have an inhibitory effect on 12-LOX activity, is significantly decreased at both the mRNA and protein level during infection with S. Typhimurium. Moreover, employing intestinal epithelial cell monolayers expressing siRNA against GPX4 mRNA, S. Typhimurium-induced PMN migration was significantly increased compared with the non-specific siRNA control cells. Conversely, in cells engineered to overexpress GPX4, S. Typhimurium-induced PMN migration was significantly decreased, which is consistent with the finding that partial depletion of GPX4 by RNAi resulted in a significant increase in HXA3 secretion during S. Typhimurium infection. Mechanistically, although we found Salmonella entry not to be required for the induced decrease in GPX4, the secreted effector, SipA, which is known to induce epithelial responses leading to stimulation of HXA3 , governed the decrease in GPX4 in a process that does not lead to an overall increase in the levels of ROS. Taken together, these results suggest that S. Typhimurium induces apical secretion of HXA3 by decreasing the expression of phospholipid GPX, which in turn leads to an increase in 12-LOX activity, and hence HXA3 synthesis.


Assuntos
Glutationa Peroxidase/metabolismo , Mucosa Intestinal/enzimologia , Neutrófilos/citologia , Neutrófilos/metabolismo , Salmonella typhimurium/fisiologia , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Mucosa Intestinal/citologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Espécies Reativas de Oxigênio/metabolismo , Migração Transendotelial e Transepitelial/genética , Migração Transendotelial e Transepitelial/fisiologia
2.
Cell Microbiol ; 14(1): 120-32, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21951973

RESUMO

Enteroaggregative Escherichia coli (EAEC) induces release of pro-inflammatory markers and disruption of intestinal epithelial barriers in vitro, suggesting an inflammatory aspect to EAEC infection. However, the mechanisms underlying EAEC-induced mucosal inflammatory responses and the extent to which these events contribute to pathogenesis is not well characterized. Employing an established in vitro model we demonstrated that EAEC prototype strain 042 induces migration of polymorphonuclear neutrophils (PMNs) across polarized T84 cell monolayers. This event was mediated through a conserved host cell signalling cascade involving the 12/15-LOX pathway and led to apical secretion of an arachidonic acid-derived lipid PMN chemoattractant, guiding PMNs across the epithelia to the site of infection. Moreover, supporting the hypothesis that inflammatory responses may contribute to EAEC pathogenesis, we found that PMN transepithelial migration promoted enhanced attachment of EAEC 042 to T84 cells. These findings suggest that EAEC-induced PMN infiltration may favour colonization and thus pathogenesis of EAEC.


Assuntos
Araquidonato 12-Lipoxigenase/metabolismo , Quimiotaxia de Leucócito , Escherichia coli/imunologia , Neutrófilos/metabolismo , Migração Transendotelial e Transepitelial/imunologia , Ácidos Araquidônicos , Aderência Bacteriana , Linhagem Celular Tumoral , Escherichia coli/patogenicidade , Humanos , Inflamação , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Proteína Quinase C/metabolismo , Transdução de Sinais
3.
J Infect Dis ; 206(5): 714-22, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22723643

RESUMO

BACKGROUND: Enteroaggregative Escherichia coli (EAEC) are increasingly recognized as an important agent of inflammatory and often persistent diarrhea. Although previous studies report on the inflammatory aspects of EAEC pathogenesis, the mechanisms by which EAEC trigger these events are not well understood. METHODS: EAEC strains harboring mutations in known EAEC virulence determinants were tested in an in vitro model of transepithelial migration of polymorphonuclear neutrophils (PMNs) and in human intestinal xenografts in severe-combined immunodeficient (SCID-HU-INT) mice, a novel model for studying EAEC disease in vivo. RESULTS: Expression of aggregative adherence fimbriae (AAFs), the principal adhesins of EAEC, was required for EAEC-induced PMN transepithelial migration in vitro. Moreover, constructed plasmids encoding AAF gene clusters demonstrated that the AAF adhesins are sufficient for triggering this event in a nonpathogenic E. coli background. Furthermore, with use of the SCID-HU-INT mouse model, severe tissue damage and infiltration of inflammatory cells was observed in the human tissue after EAEC infection. These pathological marks were strongly related to AAF expression, thus clearly confirming our in vitro findings. CONCLUSIONS: The present work establishes EAEC as an important inflammatory pathogen and the AAF adhesins as inducers of potentially detrimental immune responses.


Assuntos
Adesinas de Escherichia coli/imunologia , Diarreia/microbiologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Fímbrias Bacterianas/imunologia , Adesinas de Escherichia coli/genética , Animais , Aderência Bacteriana/imunologia , Movimento Celular/imunologia , Clonagem Molecular , Diarreia/imunologia , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/microbiologia , Histocitoquímica , Humanos , Imunidade Inata/imunologia , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos SCID , Neutrófilos/imunologia , Transplante Heterólogo
4.
Cell Microbiol ; 13(12): 2007-21, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21899702

RESUMO

In human disease induced by Salmonella enterica serovar Typhimurium (S. Typhimurium), transepithelial migration of neutrophils rapidly follows attachment of the bacteria to the epithelial apical membrane. We have previously shown that during S. Typhimurium infection the multidrug resistance-associated protein 2 (MRP2) is highly expressed at the apical surface of the intestinal epithelia, and that it functions as an efflux pump for the potent neutrophil chemoattractant hepoxilin A(3) . However, the molecular mechanisms regulating its apical localization during active states of inflammation remain unknown. Thus, our objective was to determine the mechanistic basis for the translocation of MRP2 to the apical surface of intestinal epithelial cells during S. Typhimurium infection. We show that suppression of ezrin, through either RNAi or truncation of the C-terminus, results not only in a decrease in S. Typhimurium-induced neutrophil transmigration but also significantly attenuates the apical membrane expression of MRP2 during Salmonella infection. In addition, we determined that S. Typhimurium induces the activation of ezrin via a PKC-α-dependent pathway and that ezrin activation is coupled to apical localization of MRP2. Based on these results we propose that activation of ezrin is required for the apical localization of MRP2 during S. Typhimurium infection.


Assuntos
Proteínas de Bactérias/metabolismo , Movimento Celular , Proteínas do Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Neutrófilos/metabolismo , Infecções por Salmonella/metabolismo , Salmonella typhimurium/patogenicidade , Proteínas de Bactérias/genética , Benzofenantridinas/farmacologia , Western Blotting , Carbazóis/farmacologia , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/genética , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Proteínas dos Microfilamentos/genética , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Fosforilação , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Transfecção
5.
J Clin Invest ; 128(9): 4044-4056, 2018 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-30102254

RESUMO

Neutrophil influx into the intestinal lumen is a critical response to infectious agents, but is also associated with severe intestinal damage observed in idiopathic inflammatory bowel disease. The chemoattractant hepoxilin A3, an eicosanoid secreted from intestinal epithelial cells by the apically restricted efflux pump multidrug resistance protein 2 (MRP2), mediates this neutrophil influx. Information about a possible counterbalance pathway that could signal the lack of or resolution of an apical inflammatory signal, however, has yet to be described. We now report a system with such hallmarks. Specifically, we identify endocannabinoids as the first known endogenous substrates of the apically restricted multidrug resistance transporter P-glycoprotein (P-gp) and reveal a mechanism, which we believe is novel, for endocannabinoid secretion into the intestinal lumen. Knockdown or inhibition of P-gp reduced luminal secretion levels of N-acyl ethanolamine-type endocannabinoids, which correlated with increased neutrophil transmigration in vitro and in vivo. Additionally, loss of CB2, the peripheral cannabinoid receptor, led to increased pathology and neutrophil influx in models of acute intestinal inflammation. These results define a key role for epithelial cells in balancing the constitutive secretion of antiinflammatory lipids with the stimulated secretion of proinflammatory lipids via surface efflux pumps in order to control neutrophil infiltration into the intestinal lumen and maintain homeostasis in the healthy intestine.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Endocanabinoides/metabolismo , Mucosa Intestinal/metabolismo , Infiltração de Neutrófilos/fisiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/deficiência , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Transporte Biológico Ativo , Linhagem Celular , Modelos Animais de Doenças , Feminino , Homeostase , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/prevenção & controle , Mucosa Intestinal/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Receptor CB2 de Canabinoide/deficiência , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/metabolismo , Transdução de Sinais
6.
Science ; 330(6002): 390-393, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20947770

RESUMO

The enteric pathogen Salmonella enterica serovar Typhimurium causes food poisoning resulting in gastroenteritis. The S. Typhimurium effector Salmonella invasion protein A (SipA) promotes gastroenteritis by functional motifs that trigger either mechanisms of inflammation or bacterial entry. During infection of intestinal epithelial cells, SipA was found to be responsible for the early activation of caspase-3, an enzyme that is required for SipA cleavage at a specific recognition motif that divided the protein into its two functional domains and activated SipA in a manner necessary for pathogenicity. Other caspase-3 cleavage sites identified in S. Typhimurium appeared to be restricted to secreted effector proteins, which indicates that this may be a general strategy used by this pathogen for processing of its secreted effectors.


Assuntos
Proteínas de Bactérias/metabolismo , Caspase 3/metabolismo , Mucosa Intestinal/microbiologia , Proteínas dos Microfilamentos/metabolismo , Salmonelose Animal/microbiologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Ativação Enzimática , Gastroenterite/metabolismo , Gastroenterite/microbiologia , Gastroenterite/patologia , Humanos , Mucosa Intestinal/enzimologia , Intestinos/enzimologia , Intestinos/microbiologia , Intestinos/patologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Infiltração de Neutrófilos , Salmonelose Animal/patologia , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA