Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(7): e0133453, 2015.
Article in English | MEDLINE | ID: mdl-26192616

ABSTRACT

Fibrotic remodeling is a process common to chronic lung diseases such as chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, acute respiratory distress syndrome and asthma. Based on preclinical studies phosphodiesterase 4 (PDE4) inhibitors may exhibit beneficial anti-inflammatory and anti-remodeling properties for the treatment of these respiratory disorders. Effects of PDE4 inhibitors on changes in the lung metabolome in models of pulmonary fibrotic remodeling have not yet been explored. This work studies the effects of the PDE4 inhibitor roflumilast on changes in the lung metabolome in the common murine model of bleomycin-induced lung fibrosis by nuclear magnetic resonance (NMR) metabolic profiling of intact lung tissue. Metabolic profiling reveals strong differences between fibrotic and non-fibrotic tissue. These differences include increases in proline, glycine, lactate, taurine, phosphocholine and total glutathione and decreases in global fatty acids. In parallel, there was a loss in plasma BH4. This profile suggests that bleomycin produces alterations in the oxidative equilibrium, a strong inflammatory response and activation of the collagen synthesis among others. Roflumilast prevented most of these metabolic effects associated to pulmonary fibrosis suggesting a favorable anti-fibrotic profile.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Lung/drug effects , Metabolome , Pulmonary Fibrosis/metabolism , Animals , Bleomycin , Cyclopropanes/pharmacology , Disease Models, Animal , Lung/metabolism , Lung/pathology , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology
2.
COPD ; 12(3): 320-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25207459

ABSTRACT

BACKGROUND: Cigarette smoking contributes to epithelial-mesenchymal transition (EMT) in COPD small bronchi as part of the lung remodeling process. We recently observed that roflumilast N-oxide (RNO), the active metabolite of the PDE4 inhibitor roflumilast, prevents cigarette smoke-induced EMT in differentiated human bronchial epithelial cells. Further, statins were shown to protect renal and alveolar epithelial cells from EMT. OBJECTIVES: To analyze how RNO and simvastatin (SIM) interact on CSE-induced EMT in well-differentiated human bronchial epithelial cells (WD-HBEC) from small bronchi in vitro. METHODS: WD-HBEC were stimulated with CSE (2.5%). The mesenchymal markers vimentin, collagen type I and α-SMA, the epithelial markers E-cadherin and ZO-1, as well as ß-catenin were quantified by real time quantitative PCR or Western blotting. Intracellular reactive oxygen species (ROS) were measured using the H2DCF-DA probe. GTP-Rac1 and pAkt were evaluated by Western blotting. RESULTS: The combination of RNO at 2 nM and SIM at 100 nM was (over) additive to reverse CSE-induced EMT. CSE-induced EMT was partially mediated by the generation of ROS and the activation of the PI3K/Akt/ß-catenin pathway. Both RNO at 2 nM and SIM at 100 nM partially abrogated this pathway, and its combination almost abolished ROS/ PI3K/Akt/ß-catenin signaling and therefore EMT. CONCLUSIONS: The PDE4 inhibitor roflumilast N-oxide acts (over)additively with simvastatin to prevent CSE-induced EMT in WD-HBEC in vitro.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Bronchi/cytology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Simvastatin/pharmacology , Cells, Cultured , Cyclopropanes/pharmacology , Humans , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Smoking/adverse effects , beta Catenin/metabolism
3.
Pulm Pharmacol Ther ; 28(2): 138-48, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24525294

ABSTRACT

BACKGROUND: Epithelial to mesenchymal transition (EMT) is under discussion as a potential mechanism of small airway remodelling in COPD. In bronchial epithelium of COPD and smokers markers of EMT were described. In vitro, EMT may be reproduced by exposing well-differentiated human bronchial epithelial cells (WD-HBEC) to cigarette smoke extract (CSE). EMT may be mitigated by an increase in cellular cAMP. OBJECTIVE: This study explored the effects of roflumilast N-oxide, a PDE4 inhibitor on CSE-induced EMT in WD-HBEC and in primary bronchial epithelial cells from smokers and COPD in vitro. METHODS: WD-HBEC from normal donors were stimulated with CSE (2.5%) for 72 h in presence of roflumilast N-oxide (2 nM or 1 µM) or vehicle. mRNA and protein of EMT markers αSMA, vimentin, collagen-1, E-cadherin, ZO-1, KRT5 as well as NOX4 were quantified by real-time quantitative PCR or protein array, respectively. Phosphorylated and total ERK1/2 and Smad3 were assessed by protein array. cAMP and TGFß1 were measured by ELISA. Reactive oxygen species (ROS) were determined by DCF fluorescence, after 30 min CSE (2.5%). Apoptosis was measured with Annexin V/PI labelling. In some experiments, EMT markers were determined in monolayers of bronchial epithelial cells from smokers, COPD versus controls. RESULTS: Roflumilast N-oxide protected from CSE-induced EMT in WD-HBEC. The PDE4 inhibitor reversed both the increase in mesenchymal and the loss in epithelial EMT markers. Roflumilast N-oxide restored the loss in cellular cAMP following CSE, reduced ROS, NOX4 expression, the increase in TGFß1 release, phospho ERK1/2 and Smad3. The PDE4 inhibitor partly protected from the increment in apoptosis with CSE. Finally the PDE4 inhibitor decreased mesenchymal yet increased epithelial phenotype markers in HBEC of COPD and smokers. CONCLUSIONS: Roflumilast N-oxide may mitigate epithelial-mesenchymal transition in bronchial epithelial cells in vitro.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , Aged , Apoptosis/drug effects , Bronchi/cytology , Bronchi/drug effects , Cyclic AMP/metabolism , Cyclopropanes/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , In Vitro Techniques , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/metabolism , Reactive Oxygen Species/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Smoke/adverse effects , Smoking/adverse effects , Transforming Growth Factor beta1/metabolism
4.
PLoS One ; 8(7): e69670, 2013.
Article in English | MEDLINE | ID: mdl-23936072

ABSTRACT

Respiratory syncytial virus (RSV) causes acute exacerbations in COPD and asthma. RSV infects bronchial epithelial cells (HBE) that trigger RSV associated lung pathology. This study explores whether the phosphodiesterase 4 (PDE4) inhibitor Roflumilast N-oxide (RNO), alters RSV infection of well-differentiated HBE (WD-HBE) in vitro. WD-HBE were RSV infected in the presence or absence of RNO (0.1-100 nM). Viral infection (staining of F and G proteins, nucleoprotein RNA level), mRNA of ICAM-1, ciliated cell markers (digital high speed videomicroscopy, ß-tubulin immunofluorescence, Foxj1 and Dnai2 mRNA), Goblet cells (PAS), mRNA of MUC5AC and CLCA1, mRNA and protein level of IL-13, IL-6, IL-8, TNFα, formation of H2O2 and the anti-oxidative armamentarium (mRNA of Nrf2, HO-1, GPx; total antioxidant capacity (TAC) were measured at day 10 or 15 post infection. RNO inhibited RSV infection of WD-HBE, prevented the loss of ciliated cells and markers, reduced the increase of MUC5AC and CLCA1 and inhibited the increase of IL-13, IL-6, IL-8, TNFα and ICAM-1. Additionally RNO reversed the reduction of Nrf2, HO-1 and GPx mRNA levels and consequently restored the TAC and reduced the H2O2 formation. RNO inhibits RSV infection of WD-HBE cultures and mitigates the cytopathological changes associated to this virus.


Subject(s)
Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Bronchi/pathology , Cell Differentiation/drug effects , Epithelial Cells/pathology , Epithelial Cells/virology , Respiratory Syncytial Virus Infections/drug therapy , Antioxidants/metabolism , Axonemal Dyneins/metabolism , Biomarkers/metabolism , Cell Count , Chloride Channels/genetics , Chloride Channels/metabolism , Cilia/drug effects , Cilia/metabolism , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Cytokines/metabolism , Epithelial Cells/drug effects , Forkhead Transcription Factors/metabolism , Goblet Cells/drug effects , Goblet Cells/pathology , Humans , Metaplasia , Mucin 5AC/genetics , Mucin 5AC/metabolism , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/physiology , Tubulin/metabolism , Viral Load/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...