Your browser doesn't support javascript.
loading
Conditional overexpression of TGFß1 promotes pulmonary inflammation, apoptosis and mortality via TGFßR2 in the developing mouse lung.
Sureshbabu, Angara; Syed, Mansoor A; Boddupalli, Chandra Sekhar; Dhodapkar, Madhav V; Homer, Robert J; Minoo, Parviz; Bhandari, Vineet.
Afiliação
  • Sureshbabu A; Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA. sua2011@med.cornell.edu.
  • Syed MA; Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA. Mansoor.syed@yale.edu.
  • Boddupalli CS; Department of Medicine and Yale Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA. sekhar.boddupalli@yale.edu.
  • Dhodapkar MV; Department of Medicine and Yale Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA. madhav.dhodapkar@yale.edu.
  • Homer RJ; Department of Pathology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA. Robert.homer@yale.edu.
  • Minoo P; Department of Pediatrics, University of Southern California, 1200 North State Street, Los Angeles, CA, 90033, USA. minoo@usc.edu.
  • Bhandari V; Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA. vineet.bhandari@yale.edu.
Respir Res ; 16: 4, 2015 Jan 16.
Article em En | MEDLINE | ID: mdl-25591994
BACKGROUND: Earlier studies have reported that transforming growth factor beta 1(TGFß1) is a critical mediator of hyperoxia-induced acute lung injury (HALI) in developing lungs, leading to impaired alveolarization and a pulmonary phenotype of bronchopulmonary dysplasia (BPD). However, the mechanisms responsible for the TGFß1-induced inflammatory signals that lead to cell death and abnormal alveolarization are poorly understood. We hypothesized that TGFß1 signaling via TGFßR2 is necessary for the pathogenesis of the BPD pulmonary phenotype resulting from HALI. METHODS: We utilized lung epithelial cell-specific TGFß1 overexpressing transgenic and TGFßR2 null mutant mice to evaluate the effects on neonatal mortality as well as pulmonary inflammation and apoptosis in developing lungs. Lung morphometry was performed to determine the impaired alveolarization and multicolor flow cytometry studies were performed to detect inflammatory macrophages and monocytes in lungs. Apoptotic cell death was measured with TUNEL assay, immunohistochemistry and western blotting and protein expression of angiogenic mediators were also analyzed. RESULTS: Our data reveals that increased TGFß1 expression in newborn mice lungs leads to increased mortality, macrophage and immature monocyte infiltration, apoptotic cell death specifically in Type II alveolar epithelial cells (AECs), impaired alveolarization, and dysregulated angiogenic molecular markers. CONCLUSIONS: Our study has demonstrated the potential role of inhibition of TGFß1 signaling via TGFßR2 for improved survival, reduced inflammation and apoptosis that may provide insights for the development of potential therapeutic strategies targeted against HALI and BPD.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Pneumonia / Proteínas Serina-Treonina Quinases / Apoptose / Receptores de Fatores de Crescimento Transformadores beta / Fator de Crescimento Transformador beta1 / Lesão Pulmonar Aguda / Pulmão Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2015 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Pneumonia / Proteínas Serina-Treonina Quinases / Apoptose / Receptores de Fatores de Crescimento Transformadores beta / Fator de Crescimento Transformador beta1 / Lesão Pulmonar Aguda / Pulmão Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2015 Tipo de documento: Article