Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 367
Filter
1.
J Med Chem ; 66(14): 9313-9324, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37458373

ABSTRACT

Cyclooxygenase-1 and -2 (COX1 and COX2) derived endogenous ligand prostaglandin-E2 (PGE2) triggers several physiological and pathological conditions. It mediates signaling through four G-protein coupled receptors, EP1, EP2, EP3, and EP4. Among these, EP2 is expressed throughout the body including the brain and uterus. The functional role of EP2 has been extensively studied using EP2 gene knockout mice, cellular models, and selective small molecule agonists and antagonists for this receptor. The efficacy data from in vitro and in vivo animal models indicate that EP2 receptor is a major proinflammatory mediator with deleterious functions in a variety of diseases suggesting a path forward for EP2 inhibitors as the next generation of selective anti-inflammatory and antiproliferative agents. Interestingly in certain diseases, EP2 action is beneficial; therefore, EP2 agonists seem to be clinically useful. Here, we highlight the strengths, weaknesses, opportunities, and potential threats (SWOT analysis) for targeting EP2 receptor for therapeutic development for a variety of unmet clinical needs.


Subject(s)
Dinoprostone , Receptors, Prostaglandin E , Animals , Mice , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/genetics , Dinoprostone/pharmacology , Dinoprostone/physiology , Cyclooxygenase 2 , Drug Discovery , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype
2.
Cell Rep ; 40(11): 111323, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103815

ABSTRACT

Prostaglandin receptors have been implicated in a wide range of functions, including inflammation, immune response, reproduction, and cancer. Our group has previously determined the crystal structure of the active-like EP3 bound to its endogenous agonist, prostaglandin E2. Here, we present the single-particle cryoelectron microscopy (cryo-EM) structure of the human EP3-Gi signaling complex at a resolution of 3.4 Å. The structure reveals the binding mode of Gi to EP3 and the structural changes induced in EP3 by Gi binding. In addition, we compare the structure of the EP3-Gi complex with other subtypes of prostaglandin receptors (EP2 and EP4) bound to Gs that have been previously reported and examine the differences in amino acid composition at the receptor-G protein interface. Mutational analysis reveals that the selectivity of the G protein depends on specific amino acid residues in the second intracellular loop and TM5.


Subject(s)
Dinoprostone , Receptors, Prostaglandin E , Amino Acids , Cryoelectron Microscopy , Dinoprostone/pharmacology , Humans , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism
3.
Neuroscience ; 494: 25-37, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35550162

ABSTRACT

This study aimed to re-examine the receptor subtype that mediates the fever-producing effects of prostaglandin E2 (PGE2) in the rostral ventromedial preoptic area (rvmPOA) of the hypothalamus. Among the four subtypes of PGE2 receptors (EP1, EP2, EP3, and EP4), EP3 receptor is crucially involved in the febrile effects of PGE2. However, it is possible for other subtypes of PGE2 receptor to contribute in the central mechanism of fever generation. Accordingly, effects of microinjection of PGE2 receptor subtype-specific agonists or antagonists were examined at the locus where a microinjection of a small amount (420 fmol) of PGE2 elicited prompt increases in the O2 consumption rate (VO2), heart rate, and colonic temperature (Tc) in the rvmPOA of urethane-chloralose-anesthetized rats. The EP3 agonist sulprostone mimicked, whereas its antagonist L-798,106 reduced, the febrile effects of PGE2 microinjected into the same site. Similarly, the EP4 agonist rivenprost mimicked, whereas its antagonist ONO-AE3-208 reduced, the effects of PGE2 microinjected into the same site. In contrast, microinjection of the EP1 agonist iloprost induced a very small increase in VO2 but did not have significant influences on the heart rate and Tc, whereas its antagonist, AH6809, did not affect the PGE2-induced responses. Microinjection of the EP2 agonist butaprost had no effects on the VO2, heart rate, and Tc. The results suggest that the EP3 and EP4 receptor subtypes are both involved in the fever generated by PGE2 in the rvmPOA.


Subject(s)
Preoptic Area , Receptors, Prostaglandin E , Animals , Hypothalamus/metabolism , Preoptic Area/metabolism , Rats , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype
4.
Parasite Immunol ; 42(6): e12713, 2020 06.
Article in English | MEDLINE | ID: mdl-32173875

ABSTRACT

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.


Subject(s)
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
5.
Endocrinology ; 159(10): 3482-3491, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30085093

ABSTRACT

During the periovulatory period, the profile of fibroblast growth factor 2 (FGF2) coincides with elevated prostaglandin E2 (PGE2) levels. We investigated whether PGE2 can directly stimulate FGF2 production in bovine granulosa cells and, if so, which prostaglandin E2 receptor (PTGER) type and signaling cascades are involved. PGE2 temporally stimulated FGF2. Accordingly, endoperoxide-synthase2-silenced cells, exhibiting low endogenous PGE2 levels, had reduced FGF2. Furthermore, elevation of viable granulosa cell numbers by PGE2 was abolished with FGF2 receptor 1 inhibitor, suggesting that FGF2 mediates this action of PGE2. Epiregulin (EREG), a known PGE2-inducible gene, was studied alongside FGF2. PTGER2 agonist elevated cAMP as well as FGF2 and EREG levels. However, a marked difference between cAMP-induced downstream signaling was observed for FGF2 and EREG. Whereas FGF2 upregulated by PGE2, PTGER2 agonist, or forskolin was unaffected by the protein kinase A (PKA) inhibitor H89, EREG was significantly inhibited. FGF2 was dose-dependently stimulated by the exchange protein directly activated by cAMP (EPAC) activator; a similar induction was observed for EREG. However, forskolin-stimulated FGF2, but not EREG, was inhibited in EPAC1-silenced cells. These findings ascribe a novel autocrine role for PGE2, namely, elevating FGF2 production in granulosa cells. This study also reveals that cAMP-activated EPAC1, rather than PKA, mediates the effect of PGE2/PTGER2 on the expression of FGF2. Stimulation of EREG by PGE2 is also mediated by PTGER2 but, in contrast to FGF2, EREG was found to be PKA sensitive. PGE2-stimulated FGF2 can act to maintain granulosa cell survival; it can also act on ovarian endothelial cells to promote angiogenesis.


Subject(s)
Cyclic AMP/metabolism , Dinoprostone/pharmacology , Fibroblast Growth Factor 2/metabolism , Granulosa Cells/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Animals , Cattle , Cells, Cultured , Epiregulin/genetics , Epiregulin/metabolism , Female , Fibroblast Growth Factor 2/genetics , Gene Expression/drug effects , Granulosa Cells/cytology , Granulosa Cells/metabolism , Guanine Nucleotide Exchange Factors/genetics , Models, Biological , RNA Interference , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/metabolism , Signal Transduction
6.
Pulm Pharmacol Ther ; 49: 75-87, 2018 04.
Article in English | MEDLINE | ID: mdl-29408043

ABSTRACT

Arachidonic acid is metabolized by cyclooxygenases (COX-1 and COX-2) into various prostanoids which exert different functions in mammalian physiology. One of these prostanoids, prostaglandin E2 (PGE2), interacts with four different G protein-coupled receptors, named EP1, EP2, EP3 and EP4, to initiate different downstream signalling pathways. Prostanoid receptors are diversely expressed throughout different tissues all over the body and PGE2 is responsible for a large variety of beneficial and disadvantageous effects. We have recently achieved a greater understanding of the biology of prostanoid E receptors and the potential for specific drug targeting with the advent of potent and selective EP receptor agonists and antagonists. This has important implications for lung health and disease as PGE2-mediated EP receptor activation impacts upon migration of airway smooth muscle cells, airway microvascular leak, tone regulation of pulmonary blood vessels, mast cell degranulation, bronchodilatation, cough, angiogenesis and airway inflammation, to name a few. In this review, we overview the EP receptor family and the related signalling pathways, summarize a variety of EP1-4 receptor agonists and antagonists, provide an overview of pharmacological tools used to implicate EP receptor function in the context of respiratory health and disease and finally highlight some of the more selective pharmacological reagents that have recently been developed. The availability of selective pharmacological agonists and antagonists for the distinct EP receptors, as well as the development of specific prostanoid receptor knock-out mice, offer hitherto unattainable opportunities for achieving an in depth understanding of the role and function of PGE2 in respiratory disease and the exciting potential of targeting EP receptors more broadly.


Subject(s)
Drug Development/methods , Lung Diseases/drug therapy , Receptors, Prostaglandin E/metabolism , Animals , Arachidonic Acid/metabolism , Humans , Lung/drug effects , Lung/metabolism , Lung Diseases/physiopathology , Mice , Mice, Knockout , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Signal Transduction/drug effects
7.
Theriogenology ; 95: 8-17, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28460684

ABSTRACT

In previous work, an EP2 prostanoid receptor (EP2R) agonist in vivo increased mRNA expression of luteal LH receptors (LHR), unoccupied and occupied luteal; LHR, and circulating progesterone, while an EP3R or FPR agonist decreased; mRNA expression of luteal LHR, unoccupied and occupied luteal LHR, and; circulating progesterone. An EP4R and lysophosphatidic acid (LPA) LPA2R and LPA3R agonists were reported to inhibit luteal function and sirtuins have been proposed to increase prostaglandin synthesis. The objectives were to determine; whether an EP4R, LPA2R, or LPA3R agonist affect ovine luteal function in vivo or; in vitro. In addition, whether sirtuin (SIRT)-1, 2, or 3; LPA2R or LPA3R; or EP1R, EP2R, EP3R, or EP4R agonists affect caruncular endometrial PGF2α or PGE (PGE1+PGE2) secretion in vitro. Day-10 nonpregnant ewes received a single injection of Vehicle (N = 5); an LPA2R (N = 5); LPA3R (N = 6); or EP4R (N = 5) agonist given into the interstitial tissue of the ovarian vascular pedicle adjacent to the luteal-containing ovary to determine effects on circulating progesterone, mRNA expression of luteal LHR, and luteal unoccupied and occupied LHR. In addition, agonists for LPA2R, LPA3R, EP1R, EP2R, EP3R, or EP4R or SIRT-1, SIRT-2, or SIRT-3 activators were incubated with caruncular endometrial slices in vitro to determine their effect on caruncular endometrial PGF2α, or PGE secretion. LPA2R, LPA3R, or an EP4R agonist in vivo did not affect (P ≥ 0.05) luteal weight, circulating progesterone, or occupied luteal LHR. However, an LPA2R or EP4R agonist, but; not LPA3R agonist, in vivo increased (P ≤ 0.05) mRNA expression of luteal LHR. An; LPA2R, LPA3R, or EP4R agonist increased (P ≤ 0.05) luteal unoccupied LHR, but; not occupied LHR. An LPA2R, LPA3R, or an EP4R agonist did not affect (P ≥ 0.05); luteal progesterone secretion in vitro. An LPA2R or LPA3R agonist did not affect (P ≥ 0.05) luteal PGF2α, or PGE secretion in vitro. However, an EP4R agonist tended to decrease (P < 0.066) luteal PGF2α secretion and increased (P ≤ 0.05) luteal PGE; secretion in vitro. EP1R, EP2R, EP3R, or an EP4R agonist did not affect (P ≥ 0.05); caruncular endometrial PGF2α secretion in vitro. However, EP1R, EP3R, or an EP4R agonist increased caruncular endometrial PGE secretion in vitro, while two different EP2R agonists did not affect (P ≥ 0.05) caruncular endometrial PGE; secretion. A SIRT-1 activator, but not SIRT-2 or SIRT-3 activators, increased (P ≤ 0.05) caruncular endometrial PGE secretion, while sirtuin 1, 2, or 3 activators did not affect (P ≥ 0.05) caruncular endometrial PGF2α secretion. In conclusion, receptors for EP4, LPA2, and LPA3 do not appear to be involved; in luteolysis, but EP4R and LPA2R might participate in preventing luteolysis by maintaining luteal mRNA expression for LHR and preventing loss of unoccupied luteal LHR. In addition, SIRT-1, EP1R, EP3R, and EP4R might be involved in; regulating caruncular endometrial PGE secretion, but not PGF2α secretion.


Subject(s)
Corpus Luteum/drug effects , Endometrium/drug effects , Prostaglandins E/metabolism , Receptors, Lysophosphatidic Acid/agonists , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E/agonists , Sirtuins/pharmacology , Alprostadil/metabolism , Animals , Corpus Luteum/physiology , Dinoprost/metabolism , Endometrium/physiology , Female , Lysophospholipids/agonists , Lysophospholipids/antagonists & inhibitors , Progesterone/blood , Progesterone/metabolism , RNA, Messenger/metabolism , Sheep
8.
Neurobiol Aging ; 36(3): 1439-50, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25623334

ABSTRACT

Intracerebral hemorrhage (ICH) is a devastating form of stroke. Misoprostol, a synthetic prostaglandin E1 (PGE1) analog and PGE2 receptor agonist, has shown protection against cerebral ischemia. In this study, we tested the efficacy of misoprostol in the 12-month-old mice subjected to 1 of 2 complementary ICH models, the collagenase model (primary study) and blood model (secondary study, performed in an independent laboratory). We also investigated its potential mechanism of action. Misoprostol posttreatment decreased brain lesion volume, edema, and brain atrophy and improved long-term functional outcomes. In the collagenase-induced ICH model, misoprostol decreased cellular inflammatory response; attenuated oxidative brain damage and gelatinolytic activity; and decreased high-mobility group box 1 (HMGB1) expression, Src kinase activity, and interleukin-1ß expression without affecting cyclooxygenase-2 expression. Furthermore, HMGB1 inhibition with glycyrrhizin decreased Src kinase activity, gelatinolytic activity, neuronal death, and brain lesion volume. Src kinase inhibition with 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2) decreased gelatinolytic activity and brain edema and improved neurologic function but did not decrease HMGB1 protein level. These results indicate that misoprostol protects brain against ICH injury through mechanisms that may involve the HMGB1, Src kinase, and matrix metalloproteinase-2/9 pathways.


Subject(s)
Cerebral Hemorrhage/drug therapy , Misoprostol/pharmacology , Misoprostol/therapeutic use , Neuroprotective Agents , Receptors, Prostaglandin E/agonists , Animals , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/pathology , Disease Models, Animal , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Gene Expression/drug effects , Glycyrrhizic Acid/pharmacology , Inflammation , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , src-Family Kinases/genetics , src-Family Kinases/metabolism
9.
Mol Med Rep ; 11(6): 4454-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25625500

ABSTRACT

Inflammation is a potent promoter of tumor metastasis. The aim of the present study was to explore the function of systemic inflammation in the formation of lung metastasis of breast cancer cells in a mouse model. BALB/c mice were injected intraperitoneally with lipopolysaccharide (LPS) in order to establish an inflammatory animal model and 4T1 murine breast cancer cells were injected through the tail vein to induce lung metastasis. The levels of proinflammatory cytokines were evaluated by ELISA. Metastases on the surface of the lungs were counted and histologically analyzed by hematoxylin and eosin staining. Angiogenesis in the lungs was examined by CD31 immunofluorescence. Mouse pulmonary endothelial cells (MPVECs) were isolated and used to assay endothelial tube formation and determine the protein expression levels of vascular endothelial growth factor (VEGF) in vitro. Serum levels of VEGF and prostaglandin E2 (PGE2), the number and size of metastatic lesions, and the expression levels of cyclooxygenase­2 were significantly greater in the lungs of LPS­treated mice, as compared with those in control mice threated with phosphate­buffered saline. Blood vessel density was also markedly increased in the LPS­treated mice. These increases were reversed by treatment with celecoxib. In vitro, the protein expression levels of VEGF produced by the PGE2­treated cells were significantly increased in a concentration­dependent manner. In addition, the production of VEGF was increased in response to treatment with the PGE2 receptor (EP2) agonist ONO­AE1­259­01; however, this increase was abrogated by treatment with AH6809, an EP2 receptor antagonist. Treatment with PGE2 or VEGF alone promoted the tube formation of MPVECs and this effect was reversed by treatment with celecoxib. These results demonstrated that PGE2 may regulate the release of VEGF by MPVECs through the EP2 receptor pathway and thereby promoted pulmonary angiogenesis and breast cancer metastasis in a mouse model.


Subject(s)
Breast Neoplasms/pathology , Dinoprostone/metabolism , Lipopolysaccharides/toxicity , Lung Neoplasms/secondary , Lung/drug effects , Receptors, Prostaglandin E/metabolism , Animals , Blood Vessels/drug effects , Blood Vessels/physiology , Breast Neoplasms/metabolism , Celecoxib/pharmacology , Celecoxib/therapeutic use , Cell Proliferation/drug effects , Cells, Cultured , Cyclooxygenase 2/metabolism , Cytokines/analysis , Dinoprostone/blood , Dinoprostone/pharmacology , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Inflammation/chemically induced , Lung/metabolism , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Xanthones/pharmacology
10.
Eur J Pharmacol ; 749: 124-32, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25614334

ABSTRACT

Extracellular nucleotides act as inflammatory mediators through activation of multiple purinoceptors. Under inflammatory conditions, the purinergic signalling is affected by various inflammatory mediators. We previously showed that prostaglandin (PG) E2 suppressed the elevation of intracellular Ca(2+) concentration ([Ca(2+)]i) stimulated by P2X4, P2Y2, and P2Y6 receptors in J774 murine macrophages. In this study, we examined the mechanism of PGE2 inhibitory effects on P2Y6 receptor-mediated function in J774 cells. The P2Y6 receptor agonist UDP induced a sustained elevation of [Ca(2+)]i by stimulating the phospholipase C (PLC) signalling pathway. PGE2 inhibited [Ca(2+)]i elevation and phosphatidylinositol (PI) hydrolysis in a concentration-dependent manner. J774 cells highly expressed the E-type prostanoid 2 (EP2) receptor subtype, a Gs-coupled receptor. PGE2 and a selective EP2 receptor agonist caused cyclic AMP (cAMP) accumulation in J774 cells. The inhibitory effects of PGE2 on P2Y6 receptor-mediated responses were mimicked by the selective EP2 receptor agonist. Although EP2 receptor is linked to adenylyl cyclase activation, PGE2-induced inhibition of Ca(2+) response and PI hydrolysis could not be mimicked by a lipophilic cAMP derivative, dibutyryl cAMP, or an adenylyl cyclase activator, forskolin. The inhibition of UDP-induced PLC activation by PGE2 was not affected by down-regulation of protein kinase C by phorbol-12-myristate-13-acetate treatment. PGE2 inhibited PLC activation induced by aluminium fluoride, but not by the Ca(2+)-ionophore, ionomycin. Finally, the inhibition of UDP-induced PLC activation by PGE2 was impaired by Gs knockdown using siRNA. These results suggest that EP2 receptor activation in macrophages negatively controls the Gq/11-PLC signalling through a Gs-mediated, but cAMP-independent signalling mechanism.


Subject(s)
Calcium Signaling/drug effects , Dinoprostone/pharmacology , Macrophages/drug effects , Receptors, Prostaglandin E/metabolism , Receptors, Purinergic P2/metabolism , Type C Phospholipases/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Macrophages/metabolism , Mice , Receptors, Prostaglandin E/agonists
11.
J Pharmacol Exp Ther ; 351(1): 134-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25047516

ABSTRACT

The excitotoxicity caused by excessive glutamate is a critical element in the neuropathology of acute and chronic brain disorders. Therefore, inhibition of glutamate release is a potentially valuable therapeutic strategy for treating these diseases. In this study, we investigated the effect of celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor that reduces the level of prostaglandin E2 (PGE2), on endogenous glutamate release in rat cerebral cortex nerve terminals (synaptosomes). Celecoxib substantially inhibited the release of glutamate induced by the K(+) channel blocker 4-aminopyridine (4-AP), and this phenomenon was prevented by chelating the extracellular Ca(2+) ions and by the vesicular transporter inhibitor bafilomycin A1. Celecoxib inhibited a 4-AP-induced increase in cytosolic-free Ca(2+) concentration, and the celecoxib-mediated inhibition of glutamate release was prevented by the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC. However, celecoxib did not alter 4-AP-mediated depolarization and Na(+) influx. In addition, this glutamate release-inhibiting effect of celecoxib was mediated through the PGE2 subtype 2 receptor (EP2) because it was not observed in the presence of butaprost (an EP2 agonist) or PF04418948 [1-(4-fluorobenzoyl)-3-[[6-methoxy-2-naphthalenyl)methyl]-3-azetidinecarboxylic acid; an EP2 antagonist]. The celecoxib effect on 4-AP-induced glutamate release was prevented by the inhibition or activation of protein kinase A (PKA), and celecoxib decreased the 4-AP-induced phosphorylation of PKA. We also determined that COX-2 and the EP2 receptor are present in presynaptic terminals because they are colocalized with synaptophysin, a presynaptic marker. These results collectively indicate that celecoxib inhibits glutamate release from nerve terminals by reducing voltage-dependent Ca(2+) entry through a signaling cascade involving EP2 and PKA.


Subject(s)
Cerebral Cortex/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Glutamic Acid/metabolism , Presynaptic Terminals/drug effects , Prostaglandins E/metabolism , Pyrazoles/pharmacology , Receptors, Prostaglandin E/metabolism , Sulfonamides/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Celecoxib , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Exocytosis , Male , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Sodium/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
12.
Heart Vessels ; 29(3): 390-5, 2014 May.
Article in English | MEDLINE | ID: mdl-23748433

ABSTRACT

Prostaglandin E2 (PGE2) is a major prostanoid produced under inflammatory situations. There have been controversial reports showing contractile or relaxant effect of PGE2 on vascular tone in various types of blood vessels. Thus, it is still elusive whether and how PGE2 modulates vascular tone. We here assessed the effects of PGE2 on vascular contractility using different types of vasculatures isolated form rat. In endothelium-denuded aortas and mesenteric arteries, PGE2 (1 nM-10 µM) concentration-dependently enhanced the contraction elicited by K(+) (35.4 mM) or norepinephrine (10 nM). In pulmonary arteries, PGE2 did not alter the both-induced contraction. Tail arteries were relaxed by a low dose of PGE2 (1-100 nM), but this response shifted to contraction by the higher dose of PGE2 (300 nM-10 µM). There are four types of PGE2 receptors EP1-4. RT-PCR showed that aortas and mesenteric arteries abundantly expressed EP3, while tail arteries abundantly expressed EP4. We next revealed that selective EP3 agonism enhanced the contraction in mesenteric arteries, whereas EP4 agonism induced relaxation in tail arteries. Taken together, PGE2 causes different contractile responses depending on the type of vascular bed. This phenomenon may be due to the difference in expression pattern and activity of EP receptors.


Subject(s)
Dinoprostone/pharmacology , Muscle, Smooth, Vascular/drug effects , Tail/blood supply , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Dose-Response Relationship, Drug , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism
13.
Parasite Immunol ; 35(1): 51-4, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23075034

ABSTRACT

Through their receptors, prostaglandins play crucial roles in various infections. Although prostaglandin E2 (PGE2) is implicated as a susceptibility factor in Leishmania infection, the relative contributions of its four receptors--EP1, EP2, EP3 and EP4--to this infection remain unknown. We report that Leishmania major infection of BALB/c-derived peritoneal macrophages up-regulated EP1 and EP3 expressions but down-regulated EP2 and EP4 expressions. EP2 and EP4 agonists reduced parasite load, but EP1 and EP3 agonists increased parasite load in macrophages in vitro. Agonists of EP2 and EP4, antagonists of EP1 and EP3, or lentivirally expressed EP1-shRNA and EP3-shRNA significantly reduced parasite burden in susceptible BALB/c mice. These novel data suggest differential regulation and counteractive functions of EP receptor subsets.


Subject(s)
Leishmania major/physiology , Leishmaniasis, Cutaneous/immunology , Macrophages, Peritoneal/immunology , Receptors, Prostaglandin E/immunology , Animals , Leishmaniasis, Cutaneous/parasitology , Mice , Mice, Inbred BALB C , Parasite Load , RNA, Small Interfering , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/classification
14.
J Trace Elem Med Biol ; 27(1): 31-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22748695

ABSTRACT

PROJECT: Selenium deficiency has been associated with enhanced propensity of seizures in man and laboratory animals. Therefore, the present study has been designed to investigate the anti-convulsant effect of sodium selenite and seleno-dl-methionine on pentylenetetrazole induced seizures in mice and the role of prostaglandin receptor activation in the proposed anticonvulsant effect of sodium selenite. PROCEDURE: Sodium selenite (1, 3 and 10 mg kg(-1), i.p.) and seleno-dl-methionine (0.3, 1 and 3 mg kg(-1), i.p.) was used to evaluate the potential effect on pentylenetetrazole induced seizures in mice. Pentylenetetrazole induced seizures were assessed in terms of onset time of straub's tail phenomenon, jerky movements of the whole body and convulsions. Additionally, an isobolographic study design was used to examine the interaction between sodium selenite and celecoxib (a cyclooxygenase-2 inhibitor). Sodium selenite and seleno-dl-methionine significantly attenuated pentylenetetrazole induced seizures in mice. RESULTS: Prior administration of misoprostol (a selective agonist of prostaglandin E(1) receptors) markedly attenuated the anticonvulsant effect of sodium selenite as well as seleno-dl-methionine in mice. However, the administration of misoprostol per se did not produce any behavioral changes. Further, sodium selenite was observed to exert a synergistic interaction with celecoxib. CONCLUSIONS: Selenium induced reduction in seizure like behavior might be ascribed to the activation of a prostaglandin E(1) receptor activation linked mechanism. It is further proposed that sodium selenite exerts a synergistic anti-convulsant effect with celecoxib indicating the therapeutic usefulness of combining the two agents to treat epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Receptors, Prostaglandin E/agonists , Seizures/drug therapy , Selenomethionine/pharmacology , Sodium Selenite/pharmacology , Animals , Anticonvulsants/administration & dosage , Dose-Response Relationship, Drug , Male , Mice , Misoprostol , Pentylenetetrazole , Receptors, Prostaglandin E/metabolism , Seizures/chemically induced , Seizures/metabolism , Selenomethionine/administration & dosage , Sodium Selenite/administration & dosage , Structure-Activity Relationship
15.
Eur J Pharmacol ; 681(1-3): 55-9, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22342278

ABSTRACT

Arterial vascularization of the spinal cord may be mechanically or functionally altered during thoraco-abdominal surgery/intravascular procedures. Increased arterial pressure has been shown to restore spinal perfusion and function probably by increasing the blood flow through the intercostal arteries. The regulation of human intercostal artery (HICA) vascular tone is not well documented. Prostaglandin (PG)E(2) concentration is increased during inflammatory conditions and has been shown to regulate vascular tone in many preparations. In this context, the pharmacological response of HICA to PGE(2) and the characterization of the PGE(2) receptor subtypes (EP(1), EP(2), EP(3) or EP(4)) involved are of importance and that is the aim of this study. Rings of HICA were prepared from 29 patients and suspended in organ baths for isometric recording of tension. Cumulative concentration-response curves were performed in these preparations with various EP receptor agonists in the absence or presence of different receptor antagonists or inhibitors. PGE(2) induced the contraction of HICA (E(max)=7.28 ± 0.16 g; pEC(50) value=0.79 ± 0.18; n=17); contractions were also observed with the EP(3) receptor agonists, sulprostone, 17-phenyl-PGE(2), misoprostol or ONO-AE-248. In conclusion, PGE(2) induced vasoconstriction of HICA via EP(3) receptor subtypes and this result was confirmed by the use of selective EP receptor antagonists (L-826266, ONO-8713, SC-51322) and by a strong detection of EP(3) mRNA. These observations suggest that in the context of perioperative inflammation, increased PGE(2) concentrations could trigger vasoconstriction of HICA and possibly alter spinal vascularization.


Subject(s)
Dinoprostone/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E/metabolism , Vasoconstrictor Agents/metabolism , Arteries , Dinoprostone/administration & dosage , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Humans , Inflammation/physiopathology , RNA, Messenger/metabolism , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Vasoconstriction/drug effects , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacology
16.
Int Heart J ; 52(5): 266-9, 2011.
Article in English | MEDLINE | ID: mdl-22008433

ABSTRACT

Prostaglandin E2 (PGE(2)) is produced in inflammatory responses and regulates a variety of immunological reactions through 4 different receptor subtypes; EP1, 2, 3 and 4. However, the precise role of each receptor in cardiovascular disease has not yet been elucidated. Enhanced expression of some EPs has been observed in clinical and experimental cardiovascular diseases. EP agonists have been developed to clarify the role of each receptor. Recently, we developed a novel selective agonist to examine the effects of EP4 on cardiac transplantation, myocardial ischemia, and myocarditis. Of note, a selective EP4 agonist attenuated inflammatory cytokines and chemokines via attenuation of macrophage activation in inflammatory heart diseases. In this review article, we discuss the effects of PGE(2) receptor agonists on the development of cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/immunology , Dinoprostone/physiology , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/physiology , Animals , Anti-Inflammatory Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Chemokines/blood , Cytokines/blood , Graft Rejection/immunology , Heart Transplantation/immunology , Humans , Inflammation Mediators/blood , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/physiology , Structure-Activity Relationship
17.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L675-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21821730

ABSTRACT

Epoxyeicosatrienoic acid (EET) and thromboxane A(2) are arachidonic acid derivatives. The former has initially been defined as an epithelium-derived hyperpolarizing factor displaying broncho-relaxing and anti-inflammatory properties, as recently demonstrated, whereas thromboxane A(2) induces vaso- and bronchoconstriction upon binding to thromboxane-prostanoid (TP)-receptor. EETs, however, are quickly degraded by the soluble epoxide hydrolase (sEH) into inactive diol compounds. The aim of this study was to investigate the effects of 14,15-EET on TP-receptor activation in human bronchi. Tension measurements performed on native bronchi from various species, acutely treated with increasing 14,15-EET concentrations, revealed specific and concentration-dependent relationships as well as a decrease in the tension induced by 30 nM U-46619, used as a synthetic TP-receptor agonist. Interestingly, acute treatments with 3 µM N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide, an epoxygenase inhibitor, which minimizes endogenous production of EET, resulted in an increased reactivity to U-46619. Furthermore, we demonstrated that chronic treatments with trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB), a sEH inhibitor, reduced human bronchi reactivity to U-46619. During our tension measurements, we also observed that human bronchi generated small-amplitude contractions; these spontaneous activities were reduced upon acute 14,15-EET treatments in the presence of t-AUCB. Altogether, these data demonstrate that endogenous and exogenous 14,15-EET could interfere with the activation of TP-receptors as well as with spontaneous oscillations in human airway smooth muscle tissues.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Bronchi/drug effects , Muscle Tonus/drug effects , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, Prostaglandin E/metabolism , Receptors, Thromboxane/metabolism , Signal Transduction , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Arachidonic Acid/metabolism , Benzoates/pharmacology , Blotting, Western , Bronchi/cytology , Bronchi/physiology , Cells, Cultured , Eicosapentaenoic Acid/metabolism , Electrophoresis, Polyacrylamide Gel , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/metabolism , Epoxy Compounds , Fluorescent Antibody Technique , Guinea Pigs , Humans , Mice , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Rats , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Thromboxane/agonists , Receptors, Thromboxane/antagonists & inhibitors , Thromboxane A2/metabolism , Urea/analogs & derivatives , Urea/pharmacology
18.
Prostaglandins Other Lipid Mediat ; 96(1-4): 10-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21801847

ABSTRACT

Hypertension is a well established risk factor for cardiovascular diseases such as stroke and is the leading cause of chronic kidney failure. Although a number of pharmacologic agents are available for the treatment of hypertension including agents that affect the renin-angiotensin-aldosterone system (RAAS), unmet needs in the treatment of hypertension suggest that identification of novel pharmacological targets would be an important healthcare goal. One potential target is prostaglandin E(2) (PGE(2)), a potent lipid mediator with a diverse and sometimes opposing range of biological effects. PGE(2) signals through four subtypes of G-protein coupled receptors designated EP1 through EP4. PGE(2) functions primarily as a vasodepressor; under certain conditions PGE(2) administration mediates vasopressor activity. This review focuses on the current understanding of the roles of PGE(2) receptors in vascular reactivity, hypertension and end-organ damage.


Subject(s)
Cardiovascular Diseases/metabolism , Dinoprostone/metabolism , Hypertension/metabolism , Kidney Failure, Chronic/metabolism , Kidney/metabolism , Receptors, Prostaglandin E/metabolism , Animals , Blood Pressure/drug effects , Calcium/metabolism , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Cyclic AMP/metabolism , Dinoprostone/pharmacology , Disease Models, Animal , Humans , Hypertension/complications , Hypertension/drug therapy , Hypertension/physiopathology , Kidney/drug effects , Kidney/physiopathology , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/physiopathology , Mice , Mice, Knockout , Prostaglandin Antagonists/pharmacology , Rats , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Renin-Angiotensin System/drug effects , Rodentia , Signal Transduction/drug effects , Signal Transduction/physiology , Vasoconstrictor Agents/pharmacology
19.
Neurogastroenterol Motil ; 23(8): 782-e336, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21605283

ABSTRACT

BACKGROUND: Prostaglandin E(2) (PGE(2) ) is an inflammatory mediator implicated in several gastrointestinal pathologies that affect normal intestinal transit. The aim was to establish the contribution of the four EP receptor types (EP(1-4) ), in human colon, that mediate PGE(2) -induced longitudinal smooth muscle contraction. METHODS: Changes in isometric muscle tension of human colon, mouse colon and mouse ileum were measured in organ baths in response to receptor-specific agonists and antagonists. In addition, lidocaine was used to block neurogenic activity to investigate whether EP receptors were pre- or post-junctional. KEY RESULTS: PGE(2) contracted longitudinal muscle from human and mouse colon and mouse ileum. These contractions were inhibited by the EP(1) receptor antagonist, EP(1) A in human colon, whereas a combination of EP(1) A and the EP(3) antagonist, L798106 inhibited agonist responses in both mouse preparations. The EP(3) agonist, sulprostone also increased muscle tension in both mouse tissues, and these responses were inhibited by lidocaine in the colon but not in the ileum. Although PGE(2) consistently contracted all three muscle preparations, butaprost decreased tension by activating smooth muscle EP(2) receptors in both colonic tissues. Alternatively, in mouse ileum, butaprost responses were lidocaine-sensitive, suggesting that it was activating prejunctional EP(2) receptors on inhibitory motor neurons. Conversely, EP(4) receptors were not functional in all the intestinal muscle preparations tested. CONCLUSIONS & INFERENCES: PGE(2) -induced contraction of longitudinal smooth muscle is mediated by EP(1) receptors in human colon and by a combination of EP(1) and EP(3) receptors in mouse intestine, whereas EP(2) receptors modulate relaxation in all three preparations.


Subject(s)
Colon/physiology , Ileum/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Protein Isoforms/metabolism , Receptors, Prostaglandin E/metabolism , Aged , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Dinoprostone/metabolism , Female , Humans , Isometric Contraction/physiology , Male , Mice , Mice, Inbred C57BL , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Neurons/metabolism , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors
20.
Thorax ; 66(12): 1029-35, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21606476

ABSTRACT

BACKGROUND: Asthma and chronic obstructive pulmonary disease are airway inflammatory diseases characterised by airflow obstruction. Currently approved bronchodilators such as long-acting ß(2) adrenoceptor agonists are the mainstay treatments but often fail to relieve symptoms of chronic obstructive pulmonary disease and severe asthma and safety concerns have been raised over long-term use. The aim of the study was to identify the receptor involved in prostaglandin E(2) (PGE(2))-induced relaxation in guinea pig, murine, monkey, rat and human airways in vitro. METHODS: Using an extensive range of pharmacological tools, the relaxant potential of PGE(2) and selective agonists for the EP(1-4) receptors in the presence and absence of selective antagonists in guinea pig, murine, monkey, rat and human isolated airways was investigated. RESULTS: In agreement with previous studies, it was found that the EP(2) receptor mediates PGE(2)-induced relaxation of guinea pig, murine and monkey trachea and that the EP(4) receptor mediates PGE(2)-induced relaxation of the rat trachea. These data have been confirmed in murine airways from EP(2) receptor-deficient mice (Ptger2). In contrast to previous publications, a role for the EP(4) receptor in relaxant responses in human airways in vitro was found. Relaxant activity of AH13205 (EP(2) agonist) was also demonstrated in guinea pig but not human airway tissue, which may explain its failure in clinical studies. CONCLUSION: Identification of the receptor mediating PGE(2)-induced relaxation represents a key step in developing a novel bronchodilator therapy. These data explain the lack of bronchodilator activity observed with selective EP(2) receptor agonists in clinical studies.


Subject(s)
Asthma/drug therapy , Bronchodilator Agents/pharmacology , Dinoprostone/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , Receptors, Prostaglandin E/agonists , Trachea/drug effects , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Asthma/physiopathology , Bridged Bicyclo Compounds, Heterocyclic , Dinoprostone/analogs & derivatives , Fatty Acids, Unsaturated , Guinea Pigs , Humans , Hydrazines/pharmacology , Macaca fascicularis , Methyl Ethers/pharmacology , Mice , Mice, Inbred C57BL , Naphthalenes/pharmacology , Phenylbutyrates/pharmacology , Prostanoic Acids/pharmacology , Pulmonary Disease, Chronic Obstructive/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/physiology , Regression Analysis , Species Specificity , Trachea/physiology , Xanthones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL