Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 308(3): L259-69, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480336

RESUMO

Endothelial dysfunction underlies the pathophysiology of vascular disorders such as acute lung injury (ALI) syndromes. Recent work has identified the Abl family kinases (c-Abl and Arg) as important regulators of endothelial cell (EC) barrier function and suggests that their inhibition by currently available pharmaceutical agents such as imatinib may be EC protective. Here we describe novel and differential effects of imatinib in regulating lung pathophysiology in two clinically relevant experimental models of ALI. Imatinib attenuates endotoxin (LPS)-induced vascular leak and lung inflammation in mice but exacerbates these features in a mouse model of ventilator-induced lung injury (VILI). We next explored these discrepant observations in vitro through investigation of the roles for Abl kinases in cultured lung EC. Imatinib attenuates LPS-induced lung EC permeability, restores VE-cadherin junctions, and reduces inflammation by suppressing VCAM-1 expression and inflammatory cytokine (IL-8 and IL-6) secretion. Conversely, in EC exposed to pathological 18% cyclic stretch (CS) (in vitro model of VILI), imatinib decreases VE-cadherin expression, disrupts cell-cell junctions, and increases IL-8 levels. Downregulation of c-Abl expression with siRNA attenuates LPS-induced VCAM-1 expression, whereas specific reduction of Arg reduces VE-cadherin expression in 18% CS-challenged ECs to mimic the imatinib effects. In summary, imatinib exhibits pulmonary barrier-protective and anti-inflammatory effects in LPS-injured mice and lung EC; however, imatinib exacerbates VILI as well as dysfunction in 18% CS-EC. These findings identify the Abl family kinases as important modulators of EC function and potential therapeutic targets in lung injury syndromes.


Assuntos
Anti-Inflamatórios/farmacologia , Benzamidas/farmacologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Animais , Anti-Inflamatórios/uso terapêutico , Benzamidas/uso terapêutico , Fenômenos Biomecânicos , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Citocinas/biossíntese , Avaliação Pré-Clínica de Medicamentos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Humanos , Mesilato de Imatinib , Lipopolissacarídeos/farmacologia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Camundongos Endogâmicos C57BL , Piperazinas/uso terapêutico , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/uso terapêutico , Estresse Fisiológico , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia , alfa-Fetoproteínas/metabolismo
2.
Sci Rep ; 11(1): 9529, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33953279

RESUMO

Microvesicles (MVs) are cell-derived extracellular vesicles that have emerged as markers and mediators of acute lung injury (ALI). One of the most common pathogens in pneumonia-induced ALI is Streptococcus pneumoniae (Spn), but the role of MVs during Spn lung infection is largely unknown. In the first line of defense against Spn and its major virulence factor, pneumolysin (PLY), are the alveolar epithelial cells (AEC). In this study, we aim to characterize MVs shed from PLY-stimulated AEC and explore their contribution in mediating crosstalk with neutrophils. Using in vitro cell and ex vivo (human lung tissue) models, we demonstrated that Spn in a PLY-dependent manner stimulates AEC to release increased numbers of MVs. Spn infected mice also had higher levels of epithelial-derived MVs in their alveolar compartment compared to control. Furthermore, MVs released from PLY-stimulated AEC contain mitochondrial content and can be taken up by neutrophils. These MVs then suppress the ability of neutrophils to produce reactive oxygen species, a critical host-defense mechanism. Taken together, our results demonstrate that AEC in response to pneumococcal PLY release MVs that carry mitochondrial cargo and suggest that these MVs regulate innate immune responses during lung injury.


Assuntos
Células Epiteliais Alveolares/imunologia , Micropartículas Derivadas de Células/imunologia , Neutrófilos/imunologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Estreptolisinas/imunologia , Células A549 , Adulto , Proteínas de Bactérias/imunologia , Células Cultivadas , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Pulmão/citologia , Pulmão/imunologia , Mitocôndrias/imunologia , Pneumonia Pneumocócica/imunologia , Explosão Respiratória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA