Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Chin Med J (Engl) ; 127(11): 2129-37, 2014.
Article in English | MEDLINE | ID: mdl-24890166

ABSTRACT

BACKGROUND: Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are the first steps in the development of multiple organ failure induced by sepsis. A systemic excessive inflammatory reaction is currently the accepted mechanism of the pathogenesis of sepsis. Several studies have suggested a protective role of the peroxisome proliferator activated receptor-ß/δ (PPAR-ß/δ) in related inflammatory diseases. But the role of PPARß/δ in ALI remains uncertain. The aim of this study was to investigate the role and possible mechanism of PPARß/δ in ALI induced by sepsis. METHODS: Cecal ligation and puncture (CLP) was used as a sepsis model. Rats were randomly divided into four groups, the control group (CON, n = 6), sham-operation group (SHAM, n = 12), cecal ligation and puncture group (CLP, n = 30), GW501516 group (CLP+GW, n = 25), which underwent CLP and were subcutaneously injected with the PPAR-ß/δ agonist GW501516 (0.05 mg/100 g body weight). Survival was monitored to 24 hours after operation. Blood pressure, serum creatinine, blood urea nitrogen, aspartate aminotrasferase and alanine aminotrasferase were measured after CLP. Concentrations of tumor necrosis factor α (TNF-α) and interleukin (IL)-1ß in serum were detected by enzyme linked immunosorbent assay (ELISA) kits. Lung tissue samples were stained with H&E and scored according to the degree of inflammation. Bacterial colonies were counted in the peritoneal fluid. Alveolar macrophages were cultured and incubated with GW501516 (0.15 µmol/L) and PPARß/δ adenovirus and then treated with Lipopolysaccharide (2 µg/ml) for 2 hours. The TNF-α, IL-1ß and IL-6 RNA in lung and alveolar macrophages were determined by real-time PCR. Phosphorylation of signal transducer and activator of transcription 3 (STAT3) in lung and alveolar macrophages was detected by Western blotting. RESULTS: GW501516 significantly increased the survival of septic rats, decreased histological damage of the lungs, reduced inflammatory cytokines in serum and lung tissues of septic rats and did not increase counts of peritoneal bacteria. In vitro, GW501516 and over-expression of PPARß/δ attenuated gene expression of TNF-α, IL-1ß and IL-6 in alveolar macrophages. Both in vivo and in vitro, PPARß/δ inhibited the phosphorylation of STAT3. CONCLUSION: PPARß/δ plays a protective role in sepsis induced ALI via suppressing excessive inflammation.


Subject(s)
Acute Lung Injury/drug therapy , PPAR delta/metabolism , PPAR-beta/metabolism , Sepsis/drug therapy , Acute Lung Injury/etiology , Animals , Cells, Cultured , Male , PPAR delta/agonists , PPAR-beta/agonists , Rats , Rats, Sprague-Dawley , Sepsis/complications , Thiazoles/therapeutic use
2.
Molecules ; 19(4): 4967-85, 2014 Apr 21.
Article in English | MEDLINE | ID: mdl-24756129

ABSTRACT

Microsomal prostaglandin E2 synthase-1 (mPGES-1), an inducible enzyme that converts prostaglandin H2 (PGH2) to prostaglandin E2 (PGE2), plays an important role in a variety of diseases. So far, the role of mPGES-1 in idiopathic pulmonary fibrosis (IPF) remained unknown. The current study aimed to investigate the role of mPGES-1 in pulmonary fibrosis induced by bleomycin in mice. We found that mPGES-1 deficient (mPGES-1-/-) mice exhibited more severe fibrotic lesions with a decrease in PGE2 content in lungs after bleomycin treatment when compared with wild type (mPGES-1+/+) mice. The mPGES-1 expression levels and PGE2 content were also decreased in bleomycin-treated mPGES-1+/+ mice compared to saline-treated mPGES-1+/+ mice. Moreover, in both mPGES-1-/- and mPGES-1+/+ mice, bleomycin treatment reduced the expression levels of E prostanoid receptor 2 (EP2) and EP4 receptor in lungs, whereas had little effect on EP1 and EP3. In cultured human lung fibroblast cells (MRC-5), siRNA-mediated knockdown of mPGES-1 augmented transforming growth factor-ß1 (TGF-ß1)-induced α-smooth muscle actin (α-SMA) protein expression, and the increase was reversed by treatment of PGE2, selective EP2 agonist and focal adhesion kinase (FAK) inhibitor. In conclusion, these findings revealed mPGES-1 exerts an essential effect against pulmonary fibrogenesis via EP2-mediated signaling transduction, and activation of mPGES-1-PGE2-EP2-FAK signaling pathway may represent a new therapeutic strategy for treatment of IPF patients.


Subject(s)
Intramolecular Oxidoreductases/genetics , Lung/metabolism , Pulmonary Fibrosis/metabolism , Actins/genetics , Actins/metabolism , Animals , Bleomycin , Cell Line , Dinoprostone/metabolism , Dinoprostone/pharmacology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/deficiency , Lung/pathology , Mice , Mice, Knockout , Microsomes/enzymology , Prostaglandin-E Synthases , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...