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

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Pathol ; 184(5): 1489-502, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24650563

RESUMEN

Fibrotic lung diseases represent a diverse group of progressive and often fatal disorders with limited treatment options. Although the pathogenesis of these conditions remains incompletely understood, receptor type protein tyrosine phosphatase α (PTP-α encoded by PTPRA) has emerged as a key regulator of fibroblast signaling. We previously reported that PTP-α regulates cellular responses to cytokines and growth factors through integrin-mediated signaling and that PTP-α promotes fibroblast expression of matrix metalloproteinase 3, a matrix-degrading proteinase linked to pulmonary fibrosis. Here, we sought to determine more directly the role of PTP-α in pulmonary fibrosis. Mice genetically deficient in PTP-α (Ptpra(-/-)) were protected from pulmonary fibrosis induced by intratracheal bleomycin, with minimal alterations in the early inflammatory response or production of TGF-ß. Ptpra(-/-) mice were also protected from pulmonary fibrosis induced by adenoviral-mediated expression of active TGF-ß1. In reciprocal bone marrow chimera experiments, the protective phenotype tracked with lung parenchymal cells but not bone marrow-derived cells. Because fibroblasts are key contributors to tissue fibrosis, we compared profibrotic responses in wild-type and Ptpra(-/-) mouse embryonic and lung fibroblasts. Ptpra(-/-) fibroblasts exhibited hyporesponsiveness to TGF-ß, manifested by diminished expression of αSMA, EDA-fibronectin, collagen 1A, and CTGF. Ptpra(-/-) fibroblasts exhibited markedly attenuated TGF-ß-induced Smad2/3 transcriptional activity. We conclude that PTP-α promotes profibrotic signaling pathways in fibroblasts through control of cellular responsiveness to TGF-ß.


Asunto(s)
Fibroblastos/patología , Pulmón/patología , Fibrosis Pulmonar/patología , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adenoviridae , Animales , Bleomicina , Citocinas/biosíntesis , Eliminación de Gen , Genes Reporteros , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Neumonía/complicaciones , Neumonía/patología , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/prevención & control , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Transcripción Genética
2.
Am J Pathol ; 179(4): 1733-45, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21871427

RESUMEN

Idiopathic pulmonary fibrosis (IPF) may be triggered by epithelial injury that results in aberrant production of growth factors, cytokines, and proteinases, leading to proliferation of myofibroblasts, excess deposition of collagen, and destruction of the lung architecture. The precise mechanisms and key signaling mediators responsible for this aberrant repair process remain unclear. We assessed the importance of matrix metalloproteinase-3 (MMP-3) in the pathogenesis of IPF through i) determination of MMP-3 expression in patients with IPF, ii) in vivo experiments examining the relevance of MMP-3 in experimental models of fibrosis, and iii) in vitro experiments to elucidate possible mechanisms of action. Gene expression analysis, quantitative RT-PCR, and Western blot analysis of explanted human lungs revealed enhanced expression of MMP-3 in IPF, compared with control. Transient adenoviral vector-mediated expression of recombinant MMP-3 in rat lung resulted in accumulation of myofibroblasts and pulmonary fibrosis. Conversely, MMP-3-null mice were protected against bleomycin-induced pulmonary fibrosis. In vitro treatment of cultured lung epithelial cells with purified MMP-3 resulted in activation of the ß-catenin signaling pathway, via cleavage of E-cadherin, and induction of epithelial-mesenchymal transition. These processes were inhibited in bleomycin-treated MMP-3-null mice, as assessed by cytosolic translocation of ß-catenin and cyclin D1 expression. These observations support a novel role for MMP-3 in the pathogenesis of IPF, through activation of ß-catenin signaling and induction of epithelial-mesenchymal transition.


Asunto(s)
Metaloproteinasa 3 de la Matriz/metabolismo , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología , Adenoviridae/genética , Animales , Bleomicina , Cadherinas/metabolismo , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/enzimología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Femenino , Regulación Enzimológica de la Expresión Génica , Vectores Genéticos/administración & dosificación , Humanos , Pulmón/enzimología , Pulmón/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/deficiencia , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas , Fibrosis Pulmonar/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA