Your browser doesn't support javascript.
loading
Inflammatory levels of nitric oxide inhibit airway epithelial cell migration by inhibition of the kinase ERK1/2 and activation of hypoxia-inducible factor-1 alpha.
Bove, Peter F; Hristova, Milena; Wesley, Umadevi V; Olson, Nels; Lounsbury, Karen M; van der Vliet, Albert.
Affiliation
  • Bove PF; Department of Pathology, College of Medicine, University of Vermont, Burlington, VT 05405, USA.
J Biol Chem ; 283(26): 17919-28, 2008 Jun 27.
Article de En | MEDLINE | ID: mdl-18424783
ABSTRACT
Increased synthesis of NO during airway inflammation, caused by induction of nitric-oxide synthase 2 in several lung cell types, may contribute to epithelial injury and permeability. To investigate the consequence of elevated NO production on epithelial function, we exposed cultured monolayers of human bronchial epithelial cells to the NO donor diethylenetriaamine NONOate. At concentrations generating high nanomolar levels of NO, representative of inflammatory conditions, diethylenetriaamine NONOate markedly reduced wound closure in an in vitro scratch injury model, primarily by inhibiting epithelial cell migration. Analysis of signaling pathways and gene expression profiles indicated a rapid induction of the mitogen-activated protein kinase phosphatase (MPK)-1 and decrease in extracellular signal-regulated kinase (ERK)1/2 activation, as well as marked stabilization of hypoxia-inducible factor (HIF)-1alpha and activation of hypoxia-responsive genes, under these conditions. Inhibition of ERK1/2 signaling using U0126 enhanced HIF-1alpha stabilization, implicating ERK1/2 dephosphorylation as a contributing mechanism in NO-mediated HIF-1alpha activation. Activation of HIF-1alpha by the hypoxia mimic cobalt chloride, or cell transfection with a degradation-resistant HIF-1alpha mutant construct inhibited epithelial wound repair, implicating HIF-1alpha in NO-mediated inhibition of cell migration. Conversely, NO-mediated inhibition of epithelial wound closure was largely prevented after small interfering RNA suppression of HIF-1alpha. Finally, NO-mediated inhibition of cell migration was associated with HIF-1alpha-dependent induction of PAI-1 and activation of p53, both negative regulators of epithelial cell migration. Collectively, our results demonstrate that inflammatory levels of NO inhibit epithelial cell migration, because of suppression of ERK1/2 signaling, and activation of HIF-1alpha and p53, with potential consequences for epithelial repair and remodeling during airway inflammation.
Sujet(s)

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Régulation de l'expression des gènes codant pour des enzymes / Protéine p53 suppresseur de tumeur / Mitogen-Activated Protein Kinase 1 / Mitogen-Activated Protein Kinase 3 / Cellules épithéliales / Sous-unité alpha du facteur-1 induit par l'hypoxie / Inflammation / Monoxyde d'azote Type d'étude: Prognostic_studies Limites: Humans Langue: En Journal: J Biol Chem Année: 2008 Type de document: Article Pays d'affiliation: États-Unis d'Amérique

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Régulation de l'expression des gènes codant pour des enzymes / Protéine p53 suppresseur de tumeur / Mitogen-Activated Protein Kinase 1 / Mitogen-Activated Protein Kinase 3 / Cellules épithéliales / Sous-unité alpha du facteur-1 induit par l'hypoxie / Inflammation / Monoxyde d'azote Type d'étude: Prognostic_studies Limites: Humans Langue: En Journal: J Biol Chem Année: 2008 Type de document: Article Pays d'affiliation: États-Unis d'Amérique