Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 62
1.
Sci Rep ; 10(1): 11657, 2020 07 15.
Article En | MEDLINE | ID: mdl-32669620

Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by interstitial remodeling and pulmonary dysfunction. The etiology of IPF is not completely understood but involves pathologic inflammation and subsequent failure to resolve fibrosis in response to epithelial injury. Treatments for IPF are limited to anti-inflammatory and immunomodulatory agents, which are only partially effective. Prostaglandin E2 (PGE2) disrupts TGFß signaling and suppresses myofibroblast differentiation, however practical strategies to raise tissue PGE2 during IPF have been limited. We previously described the discovery of a small molecule, (+)SW033291, that binds with high affinity to the PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) and increases PGE2 levels. Here we evaluated pulmonary 15-PGDH expression and activity and tested whether pharmacologic 15-PGDH inhibition (PGDHi) is protective in a mouse model of bleomycin-induced pulmonary fibrosis (PF). Long-term PGDHi was well-tolerated, reduced the severity of pulmonary fibrotic lesions and extracellular matrix remodeling, and improved pulmonary function in bleomycin-treated mice. Moreover, PGDHi attenuated both acute inflammation and weight loss, and decreased mortality. Endothelial cells and macrophages are likely targets as these cell types highly expressed 15-PGDH. In conclusion, PGDHi ameliorates inflammatory pathology and fibrosis in murine PF, and may have clinical utility to treat human disease.


Anti-Inflammatory Agents/pharmacology , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Idiopathic Pulmonary Fibrosis/drug therapy , Pyridines/pharmacology , Thiophenes/pharmacology , Animals , Bleomycin/administration & dosage , Body Weight/drug effects , Dinoprostone/agonists , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Extracellular Matrix/drug effects , Extracellular Matrix/enzymology , Female , Gene Expression , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/mortality , Inflammation , Lung/drug effects , Lung/enzymology , Lung/pathology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Respiratory Function Tests , Survival Analysis
2.
Parasite Immunol ; 42(6): e12713, 2020 06.
Article En | MEDLINE | ID: mdl-32173875

Canine leishmaniasis (CanL) is caused by the intracellular parasite Leishmania infantum. Prostaglandin E2 (PGE2 ) exerts potent regulatory effects on the immune system in experimental model Leishmania infection, but this influence has not yet been studied in CanL. In this study, PGE2 and PGE2 receptor levels and the regulatory effect of PGE2 on arginase activity, NO2 , IL-10, IL-17, IFN-γ, TNF-α and parasite load were evaluated in cultures of splenic leucocytes obtained from dogs with CanL in the presence of agonists and inhibitors. Our results showed that splenic leucocytes from dogs with CanL had lower EP2 receptor levels than those of splenic leucocytes from healthy animals. We observed that NO2 levels decreased when the cells were treated with a PGE2 receptor agonist (EP1/EP2/EP3) or COX-2 inhibitor (NS-398) and that TNF-α, IL-17 and IFN-γ cytokine levels decreased when the cells were treated with a PGE2 receptor agonist (EP2) or PGE2 itself. The parasite load in splenic leucocyte cell cultures from dogs with CanL decreased after stimulation of the cells with PGE2 . We conclude that Leishmania infection of dogs modulates PGE2 receptors and speculate that the binding of PGE2 to its receptors may activate the microbicidal capacity of cells.


Cytokines/immunology , Dinoprostone/metabolism , Dog Diseases/drug therapy , Leishmania infantum/immunology , Leishmaniasis/veterinary , Receptors, Prostaglandin E/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Dog Diseases/immunology , Dog Diseases/parasitology , Dogs , Leishmaniasis/drug therapy , Leishmaniasis/immunology , Nitric Oxide/analysis , Nitrobenzenes/pharmacology , Parasite Load , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/physiology , Sulfonamides/pharmacology , Tumor Necrosis Factor-alpha/immunology
3.
Prostaglandins Other Lipid Mediat ; 144: 106349, 2019 10.
Article En | MEDLINE | ID: mdl-31229524

BACKGROUND: Prostaglandin E2 (PGE2) signals through 4 separate G-protein coupled receptor sub-types to elicit a variety of physiologic and pathophysiological effects. We have previously reported that mice lacking the EP4 receptor in the cardiomyocytes develop heart failure with a phenotype of dilated cardiomyopathy. Also, these mice have increased levels of chemokines, like MCP-5, in their left ventricles. We have recently reported that overexpression of the EP4 receptor could improve cardiac function in the myocardial infarction model. Furthermore, we showed that overexpression of EP4 had an anti-inflammatory effect in the whole left ventricle. It has also been shown that PGE2 can antagonize lipopolysaccharide-induced secretion of chemokines/cytokines in various cell types. We therefore hypothesized that PGE2 inhibits lipopolysaccharide (LPS)-induced MCP-5 secretion in adult mouse cardiac fibroblasts via its EP4 receptor. METHODS AND RESULTS: Our hypothesis was tested using isolated mouse adult ventricular fibroblasts (AVF) treated with LPS. Pre-treatment of the cells with PGE2 and the EP4 agonist CAY10598 resulted in reductions of the pro-inflammatory response induced by LPS. Specifically, we observed reductions in MCP-5 secretion. Western blot analysis showed reductions in phosphorylated Akt and IκBα indicating reduced NF-κB activation. The anti-inflammatory effects of PGE2 and EP4 agonist signaling appeared to be independent of cAMP, p-44/42, or p38 pathways. CONCLUSION: Exogenous treatment of PGE2 and the EP4 receptor agonist blocked the pro-inflammatory actions of LPS. Mechanistically, this was mediated via reduced Akt phosphorylation and inhibition of NF-κB.


Dinoprostone/agonists , Fibroblasts/drug effects , Lipopolysaccharides/pharmacology , Monocyte Chemoattractant Proteins/biosynthesis , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Prostaglandin E, EP4 Subtype/agonists , Animals , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Monocyte Chemoattractant Proteins/genetics , Myocardium/cytology , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , Signal Transduction/drug effects
4.
J Cell Mol Med ; 21(12): 3540-3551, 2017 Dec.
Article En | MEDLINE | ID: mdl-28699682

Endothelial injury or dysfunction is an early event in the pathogenesis of atherosclerosis. Epidemiological and animal studies have shown that 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) exposure increases morbidity and mortality from chronic cardiovascular diseases, including atherosclerosis. However, whether or how TCDD exposure causes endothelial injury or dysfunction remains largely unknown. Cultured human umbilical vein endothelial cells (HUVECs) were exposed to different doses of TCDD, and cell apoptosis was examined. We found that TCDD treatment increased caspase 3 activity and apoptosis in HUVECs in a dose-dependent manner,at doses from 10 to 40 nM. TCDD increased cyclooxygenase enzymes (COX)-2 expression and its downstream prostaglandin (PG) production (mainly PGE2 and 6-keto-PGF1α ) in HUVECs. Interestingly, inhibition of COX-2, but not COX-1, markedly attenuated TCDD-triggered apoptosis in HUVECs. Pharmacological inhibition or gene silencing of the PGE2 receptor subtype 3 (EP3) suppressed the augmented apoptosis in TCDD-treated HUVECs. Activation of the EP3 receptor enhanced p38 MAPK phosphorylation and decreased Bcl-2 expression following TCDD treatment. Both p38 MAPK suppression and Bcl-2 overexpression attenuated the apoptosis in TCDD-treated HUVECs. TCDD increased EP3-dependent Rho activity and subsequently promoted p38MAPK/Bcl-2 pathway-mediated apoptosis in HUVECs. In addition, TCDD promoted apoptosis in vascular endothelium and delayed re-endothelialization after femoral artery injury in wild-type (WT) mice, but not in EP3-/- mice. In summary, TCDD promotes endothelial apoptosis through the COX-2/PGE2 /EP3/p38MAPK/Bcl-2 pathway. Given the cardiovascular hazard of a COX-2 inhibitor, our findings indicate that the EP3 receptor and its downstream pathways may be potential targets for prevention of TCDD-associated cardiovascular diseases.


Endothelium, Vascular/drug effects , Environmental Pollutants/toxicity , Polychlorinated Dibenzodioxins/toxicity , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Prostaglandin E, EP3 Subtype/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/agonists , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Femoral Artery/injuries , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Re-Epithelialization/physiology , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Article En | MEDLINE | ID: mdl-28163255

This study investigated the effects of caffeine in the behavioral and inflammatory alterations caused by copper in zebrafish larvae, attempting to correlate these changes with the modulation of adenosine receptors. To perform a survival curve, 7dpf larvae were exposed to 10µM CuSO4, combined to different concentrations of caffeine (100µM, 500µM and 1mM) for up to 24h. The treatment with copper showed lower survival rates only when combined with 500µM and 1mM of caffeine. We selected 4 and 24h as treatment time-points. The behavior evaluation was done by analyzing the traveled distance, the number of entries in the center, and the length of permanence in the center and the periphery of the well. The exposure to 10µM CuSO4 plus 500µM caffeine at 4 and 24h changed the behavioral parameters. To study the inflammatory effects of caffeine, we assessed the PGE2 levels by using UHPLC-MS/MS, and TNF, COX-2, IL-6 and IL-10 gene expression by RT-qPCR. The expression of adenosine receptors was also evaluated with RT-qPCR. When combined to copper, caffeine altered inflammatory markers depending on the time of exposure. Adenosine receptors expression was significantly increased, especially after 4h exposure to copper and caffeine together or separately. Our results demonstrated that caffeine enhances the inflammation induced by copper by decreasing animal survival, altering inflammatory markers and promoting behavioral changes in zebrafish larvae. We also conclude that alterations in adenosine receptors are related to those effects.


Caffeine/adverse effects , Copper/toxicity , Larva/drug effects , Purinergic P1 Receptor Antagonists/adverse effects , Receptors, Purinergic P1/metabolism , Water Pollutants, Chemical/toxicity , Zebrafish/physiology , Animals , Behavior, Animal/drug effects , Biomarkers/metabolism , Caffeine/agonists , Caffeine/antagonists & inhibitors , Copper/agonists , Copper/chemistry , Copper Sulfate/administration & dosage , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Gene Expression Regulation, Developmental/drug effects , Inflammation Mediators/agonists , Inflammation Mediators/metabolism , Larva/growth & development , Larva/immunology , Larva/metabolism , Osmolar Concentration , Purinergic P1 Receptor Agonists/chemistry , Purinergic P1 Receptor Agonists/toxicity , Purinergic P1 Receptor Antagonists/chemistry , Receptors, Purinergic P1/chemistry , Receptors, Purinergic P1/genetics , Survival Analysis , Water Pollutants, Chemical/agonists , Water Pollutants, Chemical/antagonists & inhibitors , Zebrafish/growth & development , Zebrafish/immunology , Zebrafish Proteins/agonists , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
J Nutr Biochem ; 38: 93-101, 2016 12.
Article En | MEDLINE | ID: mdl-27732914

Exposure to the halogenated hydrocarbon carbon tetrachloride (CCl4) leads to hepatic lipid peroxidation, inflammation and fibrosis. Dietary supplementation of ω-3 fatty acids has been increasingly advocated as being generally anti-inflammatory, though its effect in models of liver fibrosis is mixed. This raises the question of whether diets high in ω-3 fatty acids will result in a greater sensitivity or resistance to liver fibrosis as a result of environmental toxicants like CCl4. In this study, we fed CCl4-treated mice a high ω-3 diet (using a mix of docosahexaenoic acid and eicosapentaenoic acid ethyl esters). We also co-administered an inhibitor of soluble epoxide hydrolase, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), which has been shown to boost anti-inflammatory epoxy fatty acids that are produced from both ω-3 and ω-6 dietary lipids. We showed that soluble epoxide inhibitors reduced CCl4-induced liver fibrosis. Three major results were obtained. First, the ω-3-enriched diet did not attenuate CCl4-induced liver fibrosis as judged by collagen deposition and collagen mRNA expression. Second, the ω-3-enriched diet raised hepatic tissue levels of several inflammatory lipoxygenase metabolites and prostaglandins, including PGE2. Third, treatment with TPPU in drinking water in conjunction with the ω-3-enriched diet resulted in a reduction in liver fibrosis compared to all other groups. Taken together, these results indicate that dietary ω-3 supplementation alone did not attenuate CCl4-induced liver fibrosis. Additionally, oxylipin signaling molecules may play role in the CCl4-induced liver fibrosis in the high ω-3 diet groups.


Carbon Tetrachloride Poisoning/diet therapy , Dietary Supplements , Enzyme Inhibitors/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Fatty Acids, Omega-3/therapeutic use , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/metabolism , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Combined Modality Therapy , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Down-Regulation/drug effects , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Epoxide Hydrolases/metabolism , Female , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/immunology , Male , Mice, Inbred C57BL , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/therapeutic use , Piperidines/blood , Piperidines/pharmacokinetics , Piperidines/therapeutic use , RNA, Messenger/metabolism , Reproducibility of Results
7.
Prostaglandins Other Lipid Mediat ; 121(Pt B): 218-26, 2015 Sep.
Article En | MEDLINE | ID: mdl-26482178

We examined whether lysophosphatidic acid affects prostaglandin biosynthesis, transport, and signalling in bovine steroidogenic luteal cells. The aim of the present study was to determine the influence of LPA on PGE2 and PGF2α synthesis and on the expression of enzymes involved in PG biosynthesis (PTGS2, mPGES-1, cPGES, mPGES-2, PGFS and 9-KPR), prostaglandin transporter (PGT), and prostaglandin receptors (EP1, EP2, EP3, EP4 and FP) in bovine steroidogenic luteal cells. We found that LPA inhibited PGF2α synthesis in steroidogenic luteal cells. Moreover, LPA increased mPGES1 and cPGES and decreased PGFS expression in cultured bovine steroidogenic luteal cells. Additionally, LPA stimulated EP2 and EP4 receptor and PGT expression. This study suggests that LPA activity in the bovine CL directs the physiological intraluteal balance between the two main prostanoids towards luteotropic PGE2.


Intramolecular Oxidoreductases/metabolism , Luteal Cells/metabolism , Lysophospholipids/metabolism , Organic Anion Transporters/agonists , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/agonists , Abattoirs , Animals , Biological Transport , Cattle , Cells, Cultured , Dairying , Dinoprost/antagonists & inhibitors , Dinoprost/metabolism , Dinoprostone/agonists , Dinoprostone/metabolism , Estrous Cycle/metabolism , Female , Gene Expression Regulation , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Luteal Cells/cytology , Luteal Cells/enzymology , Luteinizing Hormone/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Prostaglandin-E Synthases , 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
8.
Prostaglandins Other Lipid Mediat ; 121(Pt B): 163-9, 2015 Sep.
Article En | MEDLINE | ID: mdl-26209241

BACKGROUND: Isoprostanes are bioactive compounds formed by non-enzymatic oxidation of polyunsaturated fatty acids, mostly arachidonic, and markers of free radical generation during inflammation. In aspirin exacerbated respiratory disease (AERD), asthmatic symptoms are precipitated by ingestion of non-steroid anti-inflammatory drugs capable for pharmacologic inhibition of cyclooxygenase-1 isoenzyme. We investigated whether aspirin-provoked bronchoconstriction is accompanied by changes of isoprostanes in exhaled breath condensate (EBC). METHODS: EBC was collected from 28 AERD subjects and 25 aspirin-tolerant asthmatics before and after inhalatory aspirin challenge. Concentrations of 8-iso-PGF2α, 8-iso-PGE2, and prostaglandin E2 were measured using gas chromatography/mass spectrometry. Leukotriene E4 was measured by immunoassay in urine samples collected before and after the challenge. RESULTS: Before the challenge, exhaled 8-iso-PGF2α, 8-iso-PGE2, and PGE2 levels did not differ between the study groups. 8-iso-PGE2 level increased in AERD group only (p=0.014) as a result of the aspirin challenge. Urinary LTE4 was elevated in AERD, both in baseline and post-challenge samples. Post-challenge airways 8-iso-PGE2 correlated positively with urinary LTE4 level (p=0.046), whereas it correlated negatively with the provocative dose of aspirin (p=0.027). CONCLUSION: A significant increase of exhaled 8-iso-PGE2 after inhalatory challenge with aspirin was selective and not present for the other isoprostane measured. This is a novel finding in AERD, suggesting that inhibition of cyclooxygenase may elicit 8-iso-PGE2 production in a specific mechanism, contributing to bronchoconstriction and systemic overproduction of cysteinyl leukotrienes.


Anti-Inflammatory Agents, Non-Steroidal/toxicity , Aspirin/analogs & derivatives , Asthma, Aspirin-Induced/metabolism , Cyclooxygenase Inhibitors/toxicity , Dinoprostone/analogs & derivatives , Isoprostanes/agonists , Lung/drug effects , Lysine/analogs & derivatives , Respiratory Mucosa/drug effects , Adult , Aspirin/toxicity , Asthma/metabolism , Asthma/physiopathology , Asthma, Aspirin-Induced/physiopathology , Asthma, Aspirin-Induced/urine , Biomarkers/analysis , Biomarkers/metabolism , Biomarkers/urine , Breath Tests , Bronchial Provocation Tests , Bronchoconstriction/drug effects , Dinoprostone/agonists , Dinoprostone/analysis , Dinoprostone/metabolism , Female , Forced Expiratory Volume/drug effects , Humans , Isoprostanes/analysis , Isoprostanes/metabolism , Leukotriene E4/antagonists & inhibitors , Leukotriene E4/urine , Lung/metabolism , Lung/physiopathology , Lysine/toxicity , Male , Middle Aged , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiopathology , Severity of Illness Index , Single-Blind Method
9.
Cell Signal ; 27(3): 568-77, 2015 Mar.
Article En | MEDLINE | ID: mdl-25562426

Claudin 1 (CLDN1) is a critical component of tight junction adhesion complexes that maintains the structural integrity of epithelial cell layers. Dysregulation of CLDN1 is associated with the growth and metastasis of human lung adenocarcinoma. TNF-α treatment was previously shown to increase expression of CLDN1 that mediated lung cancer cell morphology changes and migration. This study aimed to elucidate the molecular mechanisms involved in TNF-α induced CLDN1 expression in human lung carcinoma A549 cells. Chemical inhibition or siRNA downregulation of Src, PI3K, Akt, MAPKs, NFκB, caspase and PKC demonstrated that PKC, specifically PKCδ, is required for TNF-α induced CLDN1 expression. Further investigation of the PKC pathway revealed that CLDN1 expression is enhanced by the downstream molecules iPLA2, PGE2, 15-keto PGE2 and PPARγ. Conversely, inhibition of these molecules decreased CLDN1 expression. Additionally, a wound-healing assay demonstrated that TNF-α stimulation, PKC activation, prostaglandin treatment or PPARγ activation enhanced cell migration. In conclusion, TNF-α induced CLDN1 expression is regulated by the PKCδ-iPLA2-PGE2-PPARγ signaling cascade in human lung carcinoma A549 cells.


Claudin-1/genetics , Claudin-1/metabolism , Gene Expression Regulation, Neoplastic/physiology , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Caspase 3/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dinoprostone/agonists , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Dinoprostone/pharmacology , Group VI Phospholipases A2/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , PPAR gamma/agonists , PPAR gamma/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/metabolism
10.
PLoS One ; 8(12): e82942, 2013.
Article En | MEDLINE | ID: mdl-24376609

BACKGROUND: Bisphenol A-glycidyl-methacrylate (BisGMA) employs as a monomer in dental resins. The leakage of BisGMA from composite resins into the peripheral environment can result in inflammation via macrophage activation. Prostaglandin E2 (PGE2) is a key regulator of immunopathology in inflammatory reactions. Little is known about the mechanisms of BisGMA-induced PGE2 expression in macrophage. The aim of this study was to evaluate the signal transduction pathways of BisGMA-induced PGE2 production in murine RAW264.7 macrophages. METHODOLOGY/PRINCIPAL FINDINGS: Herein, we demonstrate that BisGMA can exhibit cytotoxicity to RAW264.7 macrophages in a dose- and time-dependent manner (p<0.05). In addition, PGE2 production, COX-2 expression, and cPLA2 phosphorylation were induced by BisGMA on RAW264.7 macrophages in a dose- and time-dependent manner (p<0.05). Moreover, BisGMA could induce the phosphorylation of ERK1/2 pathway (MEK1/2, ERK1/2, and Elk), p38 pathway (MEK3/6, p38, and MAPKAPK2), and JNK pathway (MEK4, JNK, and c-Jun) in a dose- and time-dependent manner (p<0.05). Pretreatment with AACOCF3, U0126, SB203580, and SP600125 significantly diminished the phosphorylation of cPLA2, ERK1/2, p38, and JNK stimulated by BisGMA, respectively (p<0.05). BisGMA-induced cytotoxicity, cPLA2 phosphorylation, PGE2 generation, and caspases activation were reduced by AACOCF3, U0126, SB203580, and SP600125, respectively (p<0.05). CONCLUSIONS: These results suggest that BisGMA induced-PGE2 production may be via COX-2 expression, cPLA2 phosphorylation, and the phosphorylation of MAPK family. Cytotoxicity mediated by BisGMA may be due to caspases activation through the phosphorylation of cPLA2 and MAPKs family.


Acrylic Resins/pharmacology , Bisphenol A-Glycidyl Methacrylate/pharmacology , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Macrophages/drug effects , Phospholipases A2, Cytosolic/genetics , Animals , Caspases/genetics , Caspases/metabolism , Cell Line , Cyclooxygenase 2/metabolism , Dinoprostone/agonists , Dose-Response Relationship, Drug , Gene Expression Regulation , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phospholipases A2, Cytosolic/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction
11.
PLoS One ; 8(10): e76735, 2013.
Article En | MEDLINE | ID: mdl-24098559

Several studies have addressed the antiepileptic mechanisms of levetiracetam (LEV); however, its effect on catecholamines and the inflammatory mediators that play a role in epilepsy remain elusive. In the current work, lithium (Li) pretreated animals were administered LEV (500 mg/kg i.p) 30 min before the induction of convulsions by pilocarpine (PIL). Li-PIL-induced seizures were accompanied by increased levels of hippocampal prostaglandin (PG) E2, myeloperoxidase (MPO), tumor necrosis factor-α, and interleukin-10. Moreover, it markedly elevated hippocampal lipid peroxides and nitric oxide levels, while it inhibited the glutathione content. Li-PIL also reduced hippocampal noradrenaline, as well as dopamine contents. Pretreatment with LEV protected against Li-PIL-induced seizures, where it suppressed the severity and delayed the onset of seizures in Li-PIL treated rats. Moreover, LEV reduced PGE2 and MPO, yet it did not affect the level of both cytokines in the hippocampus. LEV also normalized hippocampal noradrenaline, dopamine, glutathione, lipid peroxides, and nitric oxide contents. In conclusion, alongside its antioxidant property, LEV anticonvulsive effect involves catecholamines restoration, as well as inhibition of PGE2, MPO, and nitric oxide.


Anticonvulsants/pharmacology , Hippocampus/drug effects , Piracetam/analogs & derivatives , Seizures/drug therapy , Animals , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Dopamine/metabolism , Glutathione/agonists , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Interleukin-10/agonists , Interleukin-10/antagonists & inhibitors , Interleukin-10/metabolism , Levetiracetam , Lipid Peroxidation/drug effects , Lithium Chloride , Male , Nitric Oxide/agonists , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Norepinephrine/agonists , Norepinephrine/antagonists & inhibitors , Norepinephrine/metabolism , Peroxidase/antagonists & inhibitors , Peroxidase/metabolism , Pilocarpine , Piracetam/pharmacology , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/metabolism , Seizures/physiopathology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
12.
Endocrinology ; 154(10): 3864-76, 2013 Oct.
Article En | MEDLINE | ID: mdl-23861370

The conceptus and endometrium secrete large amounts of prostaglandin E2 (PGE2) into the porcine uterine lumen during the periimplantation period. We hypothesized that PGE2 acts on conceptus/trophoblast cells through auto- and paracrine mechanisms. Real-time RT-PCR analysis revealed that PGE2 receptor (PTGER)2 mRNA was 14-fold greater in conceptuses/trophoblasts on days 14-25 (implantation and early placentation period) vs preimplantation day 10-13 conceptuses (P < .05). Similarly, expression of PTGER2 protein increased during implantation. Conceptus expression of PTGER4 mRNA and protein did not differ on days 10-19. PGE2 stimulated PTGER2 mRNA expression in day 15 trophoblast cells through PTGER2 receptor signaling. PGE2 elevated aromatase expression and estradiol-17ß secretion by trophoblast cells. Moreover, PGE2 and the PTGER2 agonist, butaprost, increased the adhesive capacity of both human HTR-8/SVneo trophoblast and primary porcine trophoblast cells to extracellular matrix. This PGE2-induced alteration in trophoblast cell adhesion to extracellular matrix was abolished by incubation of these cells with AH6809 (PTGER2 antagonist), ITGAVB3-directed tetrapeptide arg-gly-asp-ser or integrin ITGAVB3 antibody. PGE2 stimulated adhesion of porcine trophoblast cells via the estrogen receptor and MEK/MAPK signaling pathway. PGE2 induced phosphorylation of MAPK1/MAPK3 through PTGER2 and up-regulated expression of cell adhesion proteins such as focal adhesion kinase and intercellular adhesion molecule-1. Our study indicates that elevated PGE2 in the periimplantation uterine lumen stimulates conceptus PTGER2 expression, which in turn promotes trophoblast adhesion via integrins, and synthesis and secretion of the porcine embryonic signal estradiol-17ß. Moreover, the mechanism through which PGE2 increases trophoblast adhesion is not species specific because it is PTGER2- and integrin-dependent in both porcine and human trophoblast cells.


Autocrine Communication , Dinoprostone/metabolism , Embryonic Development , Paracrine Communication , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Trophoblasts/metabolism , Animals , Autocrine Communication/drug effects , Cell Adhesion/drug effects , Cell Line , Cells, Cultured , Crosses, Genetic , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Estradiol/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Humans , Integrins/antagonists & inhibitors , Integrins/metabolism , MAP Kinase Signaling System/drug effects , Paracrine Communication/drug effects , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/genetics , Sus scrofa , Trophoblasts/cytology , Trophoblasts/drug effects
13.
Food Chem Toxicol ; 59: 731-8, 2013 Sep.
Article En | MEDLINE | ID: mdl-23871789

Bronchial hyperresponsiveness is a hallmark of asthma and many factors modulate bronchoconstriction episodes. A potential correlation of formaldehyde (FA) inhalation and asthma has been observed; however, the exact role of FA remains controversial. We investigated the effects of FA inhalation on Ovalbumin (OVA) sensitisation using a parameter of respiratory mechanics. The involvement of nitric oxide (NO) and cyclooxygenase-derived products were also evaluated. The rats were submitted, or not, to FA inhalation (1%, 90 min/day, 3 days) and were OVA-sensitised and challenged 14 days later. Our data showed that previous FA exposure in allergic rats reduced bronchial responsiveness, respiratory resistance (Rrs) and elastance (Ers) to methacholine. FA exposure in allergic rats also increased the iNOS gene expression and reduced COX-1. L-NAME treatment exacerbated the bronchial hyporesponsiveness and did not modify the Ers and Rrs, while Indomethacin partially reversed all of the parameters studied. The L-NAME and Indomethacin treatments reduced leukotriene B4 levels while they increased thromboxane B2 and prostaglandin E2. In conclusion, FA exposure prior to OVA sensitisation reduces the respiratory mechanics and the interaction of NO and PGE2 may be representing a compensatory mechanism in order to protect the lung from bronchoconstriction effects.


Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bronchial Hyperreactivity/drug therapy , Disease Models, Animal , Eicosanoids/metabolism , Nitric Oxide/metabolism , Respiratory Insufficiency/prevention & control , Respiratory Mucosa/drug effects , Administration, Inhalation , Airway Resistance/drug effects , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/physiopathology , Bronchoconstrictor Agents/pharmacology , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Dinoprostone/agonists , Dinoprostone/metabolism , Formaldehyde/administration & dosage , Formaldehyde/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Leukotriene B4/antagonists & inhibitors , Leukotriene B4/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Wistar , Respiratory Insufficiency/etiology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiopathology , Thromboxane B2/agonists , Thromboxane B2/metabolism
14.
Pharmacol Rev ; 65(4): 1135-47, 2013.
Article En | MEDLINE | ID: mdl-23893067

Prostamide (prostaglandin ethanolamide) research emerged from two distinct lines of research: 1) the unique pharmacology of the antiglaucoma drug bimatoprost and 2) the discovery that endocannabinoid anandamide was converted by COX-2 to a series of electrochemically neutral prostaglandin (PG) ethanolamides. Bimatoprost pharmacology was found to be virtually identical to that of prostamide F2α. The earliest studies relied on comparison of agonist potencies compared with PGF2α and synthetic prostaglandin F2α (FP) receptor agonists. The subsequent discovery of selective and potent prostamide receptor antagonists (AGN 211334-6, as shown in Fig. 3) was critical for distinguishing between prostamide and FP receptor-mediated effects. The prostamide F2α receptor was then modeled by cotransfecting the wild-type FP receptor with an mRNA splicing variant (altFP4).Bimatoprost is now used therapeutically for treating both glaucoma and eyelash hypotrichosis. Bimatoprost also stimulates hair growth in isolated human scalp hair follicles. A strong effect is also seen in mouse pelage hair, where bimatoprost essentially halves the onset of hair regrowth and the time to achieve full hair regrowth in shaved mice. Beyond glaucoma and hair growth, bimatoprost has potential for reducing fat deposition. Studies to date suggest that preadipocytes are the cellular target for bimatoprost. The discovery of the enzyme prostamide/PGF synthase was invaluable in elucidating the anatomic distribution of prostamide F2α. High expression in the central nervous system provided the impetus for later studies that described prostamide F2α as a nociceptive mediator in the spinal cord. At the translational level, bimatoprost has already provided therapeutics in two distinct areas and the use of both prostamide agonists and antagonists may provide other useful medicaments.


Dinoprostone/analogs & derivatives , Adipose Tissue/metabolism , Amides/pharmacology , Amides/therapeutic use , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Bimatoprost , Cloprostenol/analogs & derivatives , Cloprostenol/pharmacology , Cloprostenol/therapeutic use , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Glaucoma/drug therapy , Glaucoma/metabolism , Hair/growth & development , Humans
15.
Clin Exp Allergy ; 43(7): 741-51, 2013 Jul.
Article En | MEDLINE | ID: mdl-23786281

BACKGROUND: Prostaglandin E2 (PGE2 ) has been shown to inhibit IgE-dependent histamine release from human lung mast cells. This effect of PGE2 is believed to be mediated by EP2 receptors. However, definitive evidence that this is the case has been lacking in the absence of EP2 -selective antagonists. Moreover, recent evidence has suggested that PGE2 activates EP4 receptors to inhibit respiratory cell function. OBJECTIVE: The aim of this study was to determine the receptor by which PGE2 inhibits human lung mast cell responses by using recently developed potent and selective EP2 and EP4 receptor antagonists alongside other established EP receptor ligands. METHODS: The effects of non-selective (PGE2 , misoprostol), EP2 -selective (ONO-AE1-259, AH13205, butaprost-free acid) and EP4 -selective (L-902,688, TCS251) agonists on IgE-dependent histamine release and cyclic-AMP generation in mast cells were determined. The effects of EP2 -selective (PF-04418948, PF-04852946) and EP4 -selective (CJ-042794, L-161,982) antagonists on PGE2 responses of mast cells were studied. The expression of EP receptor subtypes was determined by RT-PCR. RESULTS: Prostaglandin E2 , EP2 agonists and EP4 agonists inhibited IgE-dependent histamine release from mast cells. PGE2 and EP2 agonists, but not EP4 agonists, increased cyclic-AMP levels in mast cells. EP4 -selective antagonists did not affect the PGE2 inhibition of histamine release, whereas EP2 -selective antagonists caused rightward shifts in the PGE2 concentration-response curves. RT-PCR studies indicated that mast cells expressed EP2 and EP4 receptors. CONCLUSIONS AND CLINICAL RELEVANCE: Although human lung mast cells may express both EP2 and EP4 receptors, the principal mechanism by which PGE2 inhibits mediator release in mast cells is by activating EP2 receptors.


Dinoprostone/metabolism , Histamine Release/physiology , Histamine/metabolism , Immunoglobulin E/metabolism , Mast Cells/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Histamine Release/drug effects , Humans , Lung , Mast Cells/cytology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
16.
J Physiol Pharmacol ; 64(1): 47-54, 2013 Feb.
Article En | MEDLINE | ID: mdl-23568971

Peroxisome proliferator activated receptors (PPARs) are ligand-dependent transcriptional factors which are expressed in distinct tissues of the female reproductive system, including the ovary, uterus and placenta. An important role of PPARs in the regulation of reproductive processes has been previously highlighted in rodents. In the present study we investigated the in vitro effect of PPAR ligands on prostaglandin E2 (PGE2) release and prostaglandin E synthase (PGES) gene expression in the endometrial explants collected from cyclic (days 10-12 and 14-16 of the estrous cycle) or pregnant (days 10-12 and 14-16) pigs. A stimulatory (p<0.05) effect of rosiglitazone (PPARγ agonist) on PGE2 accumulation was noted during both stages of the estrous cycle and both stages of pregnancy, whereas a higher (p<0.05) PGES mRNA level was observed on days 10-12 of the estrous cycle and on days 14-16 of gestation when compared to the controls. The activation of PPARß by L-165,041 augmented (p<0.05) PGE2 release by the endometrium on days 14-16 of the estrous cycle and on days 14-16 of pregnancy, but the increase (p<0.05) in PGES mRNA abundance was noted on days 10-12 of the estrous cycle and during both stages of pregnancy. A stimulatory (p<0.05) effect of WY-14643 (agonist) and MK 886 (antagonist) on PGE2 release was noted on days 10-12 of the estrous cycle, and days 14-16 of pregnancy, respectively. There was a lack of change in PGES mRNA abundance in the endometrium exposed to PPARα ligands. We conclude that PPARs are mediators of prostaglandin E2 synthesis/accumulation in porcine endometrium during the luteal phase of the estrous cycle and the time of periimplantation.


Dinoprostone/biosynthesis , Endometrium/drug effects , Endometrium/metabolism , Estrous Cycle/drug effects , Estrous Cycle/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Estrous Cycle/genetics , Female , Indoles/pharmacology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Ligands , Peroxisome Proliferator-Activated Receptors/agonists , Phenoxyacetates/pharmacology , Pregnancy , Prostaglandin-E Synthases , Pyrimidines/pharmacology , RNA, Messenger/genetics , Rosiglitazone , Swine , Thiazolidinediones/pharmacology
17.
Int J Oncol ; 42(5): 1833-41, 2013 May.
Article En | MEDLINE | ID: mdl-23525457

The prostaglandin E2 (PGE2) EP1 receptor has been implicated in hepatocellular carcinoma (HCC) cell invasion. However, little is known about the mechanisms of EP1 receptor-mediated cell adhesion and migration. We previously showed that PGE2 promotes cell adhesion and migration by activating focal adhesion kinase (FAK). The present study was designed to elucidate the association between the EP1 receptor and FAK activation in HCC cells and to investigate the related signaling pathways. The effects of PGE2, EP1 agonist 17-phenyl trinor-PGE2 (17-PT-PGE2), PKC and EGFR inhibitors on FAK activation were investigated by treatment of Huh-7 cells. Phosphorylation of FAK Y397 and c-Src Y416 was investigated by western blotting. Cell adhesion and migration were analyzed by WST and transwell assays, respectively. Protein kinase C (PKC) activity was measured with a PKC assay kit. The results showed that 17-PT-PGE2 (3 µM) increased FAK Y397 phosphorylation by more than 2-fold and promoted cell adhesion and migration in Huh-7 cells. In transfected 293 cells, expression of the EP1 receptor was confirmed to upregulate FAK phosphorylation, while the EP1 receptor antagonist sc-19220 decreased PGE2-mediated FAK activation. PKC activity and c-Src Y416 phosphorylation were enhanced after 17-PT-PGE2 treatment. Both PKC and c-Src inhibitor suppressed the 17-PT-PGE2-upregulated FAK phosphorylation, as well as 17-PT-PGE2-induced cell adhesion and migration. In addition, exogenous epidermal growth factor (EGF) treatment increased FAK phosphorylation. The EGF receptor (EGFR) inhibitor also suppressed 17-PT-PGE2-upregulated FAK phosphorylation. Our study suggests that the PGE2 EP1 receptor regulates FAK phosphorylation by activating the PKC/c-Src and EGFR signal pathways, which may coordinately regulate adhesion and migration in HCC.


Carcinoma, Hepatocellular/metabolism , Dinoprostone/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Liver Neoplasms/metabolism , Protein Kinase C/metabolism , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Carcinoma, Hepatocellular/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Dinoprostone/agonists , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/genetics , Phosphorylation/drug effects , Protein Kinase C/genetics , Receptors, Prostaglandin E, EP1 Subtype/genetics , Signal Transduction
18.
J Biol Chem ; 288(3): 1953-66, 2013 Jan 18.
Article En | MEDLINE | ID: mdl-23184961

Fetal fibronectin (fFN) in cervical and vaginal secretions has been used as a predictor of preterm delivery. Here, we clarified the pathological function of fFN on cell type-specific matrix metalloproteinases (MMPs) and prostaglandin synthesis in fetal membranes. Treatment of amnion mesenchymal cells with fFN resulted in dramatic increases in MMP-1 and MMP-9 mRNA and enzymatic activity as well as COX-2 mRNA and prostaglandin E(2) synthesis, activating both NFκB and ERK1/2 signaling. Fetal FN-induced increases in MMPs and COX-2 were mediated through its extra domain A and Toll-like receptor 4 expressed in mesenchymal cells. Lipopolysaccharide and TNF-α increased the release of free FN in medium of amnion epithelial cells in culture. Finally, injection of fFN in pregnant mice resulted in preterm birth. Collectively, these results indicate that fFN is not only a marker of preterm delivery but also plays a significant role in the pathogenesis of preterm labor and premature rupture of fetal membranes.


Cyclooxygenase 2/metabolism , Fetal Proteins/metabolism , Fibronectins/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 9/metabolism , Obstetric Labor, Premature/enzymology , Premature Birth/enzymology , Amnion/cytology , Amnion/drug effects , Amnion/metabolism , Animals , Cyclooxygenase 2/genetics , Dinoprostone/agonists , Dinoprostone/biosynthesis , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extraembryonic Membranes/drug effects , Extraembryonic Membranes/metabolism , Female , Fetal Proteins/genetics , Fetal Proteins/pharmacology , Fetus , Fibronectins/genetics , Fibronectins/pharmacology , Humans , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 9/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Obstetric Labor, Premature/chemically induced , Pregnancy , Premature Birth/chemically induced , RNA, Messenger/biosynthesis , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
19.
Am J Physiol Endocrinol Metab ; 303(1): E111-21, 2012 Jul 01.
Article En | MEDLINE | ID: mdl-22535744

The cause of lower prevalence of duodenal ulcer in young women compared with men is largely unknown. We recently found that sex difference in duodenal mucosal HCO3⁻ secretion existed in humans and mice, but the mechanisms are not clear. Prostaglandin E2 (PGE2) is an important endogenous mediator that plays an important role in the regulation of duodenal HCO3⁻ secretion. Therefore, in the present study, we investigated the effect of estrogen on PGE2-stimulated duodenal HCO3⁻ secretion and the underlying mechanisms. The results showed that 17ß-estradiol at the physiological concentration (1 nM) had no significant effects on duodenal mucosal HCO3⁻ secretion or short-circuit current (I(sc)) in mice. However, the pretreatment of 17ß-estradiol (1 nM) markedly potentiated PGE2-stimulated duodenal HCO3⁻ secretion and I(sc) (P < 0.01 and P < 0.05). Global estrogen receptor (ER) antagonist ICI-182,780 and ERα-specific antagonist MPP, but not the ERß-specific antagonist PHTPP, abolished estrogen-potentiated PGE2-stimulated duodenal HCO3⁻ secretion and I(sc). 17ß-Estradiol and PGE2 additively increased phosphatidylinositol 3-kinase (PI3K) activity and Akt phosphorylation. Wortmannin, a specific PI3K inhibitor, inhibited estrogen-potentiated PGE2-stimulated duodenal HCO3⁻ secretion and I(sc). In conclusion, estrogen at the physiological concentration potentiates PGE2-stimulated duodenal mucosal HCO3⁻ secretion through the activation of ERα and the PI3K-dependent mechanism, which may contribute to the sex difference in duodenal mucosal HCO3⁻ secretion and the lower prevalence of duodenal ulcer in young women.


Bicarbonates/metabolism , Dinoprostone/metabolism , Duodenum/metabolism , Estradiol/metabolism , Intestinal Mucosa/metabolism , Secretory Pathway , Animals , Dinoprostone/agonists , Duodenum/drug effects , Enzyme Inhibitors/pharmacology , Estradiol/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrous Cycle , Female , In Vitro Techniques , Intestinal Mucosa/drug effects , Male , Mice , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Secretory Pathway/drug effects , Sex Characteristics , Signal Transduction/drug effects
20.
PLoS One ; 6(3): e17772, 2011 Mar 16.
Article En | MEDLINE | ID: mdl-21436889

The Wnt signaling pathway is a robust regulator of skeletal homeostasis. Gain-of-function mutations promote high bone mass, whereas loss of Lrp5 or Lrp6 co-receptors decrease bone mass. Similarly, mutations in antagonists of Wnt signaling influence skeletal integrity, in an inverse relation to Lrp receptor mutations. Loss of the Wnt antagonist Sclerostin (Sost) produces the generalized skeletal hyperostotic condition of sclerosteosis, which is characterized by increased bone mass and density due to hyperactive osteoblast function. Here we demonstrate that prostaglandin E(2) (PGE(2)), a paracrine factor with pleiotropic effects on osteoblasts and osteoclasts, decreases Sclerostin expression in osteoblastic UMR106.01 cells. Decreased Sost expression correlates with increased expression of Wnt/TCF target genes Axin2 and Tcf3. We also show that the suppressive effect of PGE(2) is mediated through a cyclic AMP/PKA pathway. Furthermore, selective agonists for the PGE(2) receptor EP2 mimic the effect of PGE(2) upon Sost, and siRNA reduction in Ptger2 prevents PGE(2)-induced Sost repression. These results indicate a functional relationship between prostaglandins and the Wnt/ß-catenin signaling pathway in bone.


Dinoprostone/pharmacology , Gene Expression Regulation/drug effects , Glycoproteins/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Signal Transduction/drug effects , Animals , Bone Morphogenetic Proteins/metabolism , Cyclic AMP/pharmacology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dinoprostone/agonists , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Myogenic Regulatory Factors/metabolism , Parathyroid Hormone/pharmacology , Transcription, Genetic/drug effects , Wnt Proteins/metabolism
...