Your browser doesn't support javascript.
loading
Nedd4L suppression in lung fibroblasts facilitates pathogenesis of lung fibrosis.
Li, Shuang; Ye, Qinmao; Wei, Jianxin; Taleb, Sarah J; Wang, Heather; Zhang, Yingze; Kass, Daniel J; Horowitz, Jeffrey C; Zhao, Jing; Zhao, Yutong.
Afiliação
  • Li S; Department of Medicine, The University of Pittsburgh, Pittsburgh, PA.
  • Ye Q; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH.
  • Wei J; Department of Medicine, The University of Pittsburgh, Pittsburgh, PA.
  • Taleb SJ; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH.
  • Wang H; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH.
  • Zhang Y; Department of Medicine, The University of Pittsburgh, Pittsburgh, PA.
  • Kass DJ; Department of Medicine, The University of Pittsburgh, Pittsburgh, PA.
  • Horowitz JC; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH; The Department of Internal Medicine, The Ohio State University, Columbus, OH.
  • Zhao J; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH; The Department of Internal Medicine, The Ohio State University, Columbus, OH.
  • Zhao Y; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH; The Department of Internal Medicine, The Ohio State University, Columbus, OH. Electronic address: yutong.zhao@osumc.edu.
Transl Res ; 253: 1-7, 2023 03.
Article em En | MEDLINE | ID: mdl-36257596
ABSTRACT
Ubiquitination-mediated protein degradation is associated with the development of pulmonary fibrosis. We and others have shown that Nedd4L plays anti-inflammatory and anti-fibrotic roles by targeting lysophosphatidic acid receptor 1 (LPAR1), p-Smad2/3, and ß-catenin, and other molecules for their degradation in lung epithelial cells and fibroblasts. However, the molecular regulation of Nedd4L expression in lung fibroblasts has not been studied. In this study, we find that Nedd4L levels are significantly suppressed in lung myofibroblasts in IPF patients and in experimental pulmonary fibrosis, and in TGF-ß1-treated lung fibroblasts. Nedd4L knockdown promotes TGF-ß1-mediated phosphorylation of Smad2/3 and lung myofibroblast differentiation. Mechanistically, Nedd4L targets TGF-ß receptor II (TßRII), the first key enzyme of TGF-ß1-mediated signaling, for its ubiquitination and degradation. Further, we show that inhibition of transcriptional factor E2F rescues Nedd4L levels and mitigates experimental pulmonary fibrosis. Together, our data reveal insight into mechanisms by which E2F-mediated Nedd4L suppression contributes to the pathogenesis of lung fibrosis. This study provides evidence showing that upregulation of Nedd4L is a potential therapeutic strategy to treat fibrotic disorders including lung fibrosis.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Fibrose Pulmonar Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Fibrose Pulmonar Idioma: En Ano de publicação: 2023 Tipo de documento: Article