Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nat Chem Biol ; 15(1): 8-10, 2019 01.
Article in English | MEDLINE | ID: mdl-30510192

ABSTRACT

Prostanoids are a series of bioactive lipid metabolites that function in an autacoid manner via activation of cognate G-protein-coupled receptors (GPCRs). Here, we report the crystal structure of human prostaglandin (PG) E receptor subtype EP3 bound to endogenous ligand PGE2 at 2.90 Å resolution. The structure reveals important insights into the activation mechanism of prostanoid receptors and provides a molecular basis for the binding modes of endogenous ligands.


Subject(s)
Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/chemistry , Binding Sites , Crystallography, X-Ray , Dinoprostone/chemistry , Dinoprostone/metabolism , Humans , Models, Molecular , Protein Conformation , Receptors, Prostaglandin E, EP3 Subtype/genetics , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Transforming Growth Factor alpha/metabolism
2.
Nat Chem Biol ; 15(1): 11-17, 2019 01.
Article in English | MEDLINE | ID: mdl-30510194

ABSTRACT

Misoprostol is a life-saving drug in many developing countries for women at risk of post-partum hemorrhaging owing to its affordability, stability, ease of administration and clinical efficacy. However, misoprostol lacks receptor and tissue selectivities, and thus its use is accompanied by a number of serious side effects. The development of pharmacological agents combining the advantages of misoprostol with improved selectivity is hindered by the absence of atomic details of misoprostol action in labor induction. Here, we present the 2.5 Å resolution crystal structure of misoprostol free-acid form bound to the myometrium labor-inducing prostaglandin E2 receptor 3 (EP3). The active state structure reveals a completely enclosed binding pocket containing a structured water molecule that coordinates misoprostol's ring structure. Modeling of selective agonists in the EP3 structure reveals rationales for selectivity. These findings will provide the basis for the next generation of uterotonic drugs that will be suitable for administration in low resource settings.


Subject(s)
Misoprostol/chemistry , Receptors, Prostaglandin E, EP3 Subtype/chemistry , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Binding Sites , Crystallography, X-Ray , Dinoprostone/analogs & derivatives , Dinoprostone/chemistry , Dinoprostone/metabolism , Humans , Misoprostol/metabolism , Molecular Docking Simulation , Mutagenesis, Site-Directed , Protein Conformation , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/genetics , Signal Transduction , Water/chemistry
3.
J Cell Mol Med ; 22(12): 6327-6337, 2018 12.
Article in English | MEDLINE | ID: mdl-30338939

ABSTRACT

Recent studies showed that both prostaglandin E2 (PGE2) and transient receptor potential melastatin 7 (TRPM7) play important roles in migration and proliferation of human glioblastoma cells. In this study, we tested the association between PGE2 and TRPM7. We found that PGE2 increased TRPM7 currents in HEK293 and human glioblastoma A172 cells. The PGE2 EP3 receptor antagonist L-798106 abrogated the PGE2 stimulatory effect, while EP3 agonist 17-phenyl trinor prostaglandin E2 (17-pt-PGE2) mimicked the effect of PEG2 on TRPM7. The TRPM7 phosphotransferase activity-deficient mutation, K1646R had no effect on PGE2 induced increase of TRPM7 currents. Inhibition of protein kinase A (PKA) activity by Rp-cAMP increased TRPM7 currents. TRPM7 PKA phosphorylation site mutation S1269A abolished the PGE2 effect on TRPM7 currents. PGE2 increased both mRNA and membrane protein expression of TRPM7 in A172 cells. Knockdown of TRPM7 by shRNA abrogated the PGE2 stimulated migration and proliferation of A172 cells. Blockage of TRPM7 with 2-aminoethoxydiphenyl borate (2-APB) or NS8593 had a similar effect as TRPM7-shRNA. In conclusion, our results demonstrate that PGE2 activates TRPM7 via EP3/PKA signalling pathway, and that PGE2 enhances migration and proliferation of human glioblastoma cells by up-regulation of the TRPM7 channel.


Subject(s)
Dinoprostone/genetics , Glioblastoma/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Prostaglandin E, EP3 Subtype/genetics , TRPM Cation Channels/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , HEK293 Cells , Humans , Mutation/genetics , Phosphorylation/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Prostaglandin E, EP3 Subtype/agonists , Signal Transduction/genetics
4.
Bioorg Med Chem ; 26(1): 200-214, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29203142

ABSTRACT

A highly potent and well-balanced dual agonist for the EP2 and EP3 receptors is described. Optimization of the lead compound was accomplished in consideration of the relative agonist activity against each EP subtype receptor and the pharmacokinetic profile. As the result, 2-[(2-{(1R,2R)-2-[(1E,4S)-5-cyclopentyl-4-hydroxy-4-methyl-1-penten-1-yl]-5-oxocyclopentyl}eth-yl)thio]-1,3-thiazole-4-carboxylic acid (10) showed excellent potency (human EC50 EP2 = 1.1 nM, EP3 = 1.0 nM) with acceptable selectivity over the EP1 and EP4 subtypes (>2000-fold). Further fine-tuning of compound 10 led to identification of ONO-8055 as a clinical candidate. ONO-8055 was effective at an extremely low dose (0.01 mg/kg, po, bid) in rats, and dose-dependently improved voiding dysfunction in a monkey model of underactive bladder (UAB). ONO-8055 is expected to be a novel and highly promising drug for UAB.


Subject(s)
Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/agonists , Thiazoles/pharmacology , Animals , Caco-2 Cells , Cell Membrane Permeability/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Haplorhini , Humans , Male , Models, Molecular , Molecular Structure , Rats , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
5.
Biochem Biophys Res Commun ; 485(2): 535-541, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28131828

ABSTRACT

EP3 is a receptor for prostaglandin E2 (PGE2), and although its effect on bladder excitability has attracted considerable attention, the underlying mechanism remains unclear. To investigate whether the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in the interstitial cells of Cajal (ICCs) of the bladder are involved in the effect of EP3 activation on bladder excitability, wild-type mice, HCN1 knockout (HCN1-/-) mice and rats were used in our study. Double immunofluorescence staining and immunoprecipitation assays demonstrated the interaction between EP3 and the HCN channels. Sulprostone is a selective agonist of EP3. The current density of HCN channels was enhanced by sulprostone or PGE2 using whole-cell patch clamping. Western blot analyses showed that the expression levels of HCN1 and HCN4 were higher in bladders that had undergone intravesical instillation with sulprostone than in bladders treated with normal saline (NS). Both PGE2 and sulprostone increased the calcium concentration of the ICCs, and their effects were inhibited by ZD7288 (antagonist of HCN channels) treatment. In bladder detrusor strip testing, both PGE2 and sulprostone enhanced the amplitude of the bladder detrusor in HCN1-/- mice; however, these effects were less than those in the wild-type mice. Furthermore, the effects of PGE2 and sulprostone were inhibited by ZD7288. Taken together, our results indicate that EP3 is expressed in bladder ICCs and facilitates bladder excitability via HCN channels. This study provides more comprehensive insights into the mechanism between inflammation and bladder excitability and highlights methods that can resolve bladder hyperactivity.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Interstitial Cells of Cajal/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Urinary Bladder/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cells, Cultured , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Female , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Interstitial Cells of Cajal/drug effects , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Muscle Contraction/drug effects , Protein Binding , Pyrimidines/pharmacology , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP3 Subtype/agonists , Urinary Bladder/cytology , Urinary Bladder/drug effects
6.
J Pharmacol Sci ; 135(2): 64-71, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28966102

ABSTRACT

Prostaglandin E2 (PGE2) exerts various biological effects by binding to E-prostanoid receptors (EP1-4). Although recent studies have shown that PGE2 induces cell differentiation in some neuronal cells such as mouse DRG neurons and sensory neuron-like ND7/23 cells, it is unclear whether PGE2 plays a role in differentiation of motor neurons. In the present study, we investigated the mechanism of PGE2-induced differentiation of motor neurons using NSC-34, a mouse motor neuron-like cell line. Exposure of undifferentiated NSC-34 cells to PGE2 and butaprost, an EP2-selective agonist, resulted in a reduction of MTT reduction activity without increase the number of propidium iodide-positive cells and in an increase in the number of neurite-bearing cells. Sulprostone, an EP1/3 agonist, also significantly lowered MTT reduction activity by 20%; however, no increase in the number of neurite-bearing cells was observed within the concentration range tested. PGE2-induced neurite outgrowth was attenuated significantly in the presence of PF-0441848, an EP2-selective antagonist. Treatment of these cells with dibutyryl-cAMP increased the number of neurite-bearing cells with no effect on cell proliferation. These results suggest that PGE2 promotes neurite outgrowth and suppresses cell proliferation by activating the EP2 subtype, and that the cAMP-signaling pathway is involved in PGE2-induced differentiation of NSC-34 cells.


Subject(s)
Dinoprostone/pharmacology , Dinoprostone/physiology , Motor Neurons/cytology , Neurites/physiology , Neuronal Outgrowth/drug effects , Neuronal Outgrowth/genetics , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Bucladesine/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclic AMP/physiology , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Mice , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/agonists , Signal Transduction/physiology
7.
J Urol ; 196(2): 609-16, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26880410

ABSTRACT

PURPOSE: We investigated whether the novel EP (prostaglandin E2) receptor agonist ONO-8055 would improve the lower urinary tract dysfunction of neurogenic underactive bladder in a rat lumbar spinal canal stenosis model. MATERIALS AND METHODS: First, we studied the agonistic effect of ONO-8055 on EP receptors in EP receptor expressing CHO (Chinese hamster ovary) cells using the increase in the intracellular calcium level and intracellular cAMP (cyclic adenosine monophosphate) production as indicators of receptor activation. The effects of ONO-8055 on bladder and urethral strips from normal rats were then investigated. Finally, the effects of ONO-8055 on bladder and urethral function in rats with lumbar spinal canal stenosis were evaluated by awake cystometry and intraurethral perfusion pressure, respectively. The effects of tamsulosin and distigmine on urethral pressure were also evaluated. RESULTS: ONO-8055 is a highly potent and selective agonist for EP2 and EP3 receptors on CHO cells. While this compound contracted bladder strips, it relaxed urethral strips. Awake cystometry showed that ONO-8055 significantly decreased bladder capacity, post-void residual urine and voiding pressure. Compared with vehicle, tamsulosin and ONO-8055 significantly decreased urethral pressure. CONCLUSIONS: ONO-8055 decreased post-void residual urine, probably by decreasing bladder capacity. The decrease in voiding pressure probably resulted from the lowered urethral pressure due to relaxation of the urethra. Thus, the novel EP2 and EP3 receptor dual agonist ONO-8055 has the potential to improve neurogenic underactive bladder.


Subject(s)
Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/agonists , Spinal Stenosis/complications , Thiazoles/therapeutic use , Urinary Bladder, Neurogenic/drug therapy , Urological Agents/therapeutic use , Animals , Biomarkers/metabolism , Male , Rats , Rats, Wistar , Thiazoles/pharmacology , Treatment Outcome , Urethra/drug effects , Urethra/metabolism , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder, Neurogenic/etiology , Urinary Bladder, Neurogenic/metabolism , Urological Agents/pharmacology
9.
Bioorg Med Chem Lett ; 26(3): 1016-1019, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26725951

ABSTRACT

The cyclic carbamate derivatives, 2-{[2-((4S)-4-{(1E,3R)-8-fluoro-3-hydroxy-4,4-dimethyl-1-octenyl}-2-oxo-1,3-oxazolidin-3-yl)ethyl]sulfanyl}-1,3-thiazole-4-carboxylic acid (5) and 2-{[2-((4S)-4-{(1E,3R)-3-[1-(4-fluorobutyl)cyclobutyl]-3-hydroxy-1-propenyl}-2-oxo-1,3-oxazolidin-3-yl)ethyl]sulfanyl}-1,3-thiazole-4-carboxylic acid (7) were identified as the first potent dual EP2 and EP3 agonists with selectivity against the EP1 and EP4 subtypes. Compounds 5 and 7 demonstrated highly potent dual EP2 and EP3 agonist activity with EC50 values of 10nM or less. In addition, these compounds possess structural features distinct from natural prostaglandins, such as a cyclic carbamate moiety, a dimethyl or cyclobutyl group and a terminal fluorine atom.


Subject(s)
Carboxylic Acids/chemistry , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/agonists , Animals , Carboxylic Acids/chemical synthesis , Carboxylic Acids/pharmacokinetics , Half-Life , Humans , Kinetics , Mice , Protein Binding , Rats , Receptors, Prostaglandin E, EP1 Subtype/agonists , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Structure-Activity Relationship
10.
Mediators Inflamm ; 2016: 5079597, 2016.
Article in English | MEDLINE | ID: mdl-27298516

ABSTRACT

Prostaglandin E2 (PGE2), a major metabolite of arachidonic acid produced by cyclooxygenase pathways, exerts its bioactive responses by activating four E-prostanoid receptor subtypes, EP1, EP2, EP3, and EP4. PGE2 enables modulating N-methyl-D-aspartate (NMDA) receptor-mediated responses. However, the effect of E-prostanoid receptor agonists on large-conductance Ca(2+)-activated K(+) (BK) channels, which are functionally coupled with NMDA receptors, remains unclear. Here, we showed that EP2 receptor-mediated signaling pathways increased NMDA-induced outward currents (I NMDA-OUT), which are associated with the BK channel activation. Patch-clamp recordings from the acutely dissociated mouse cortical neurons revealed that an EP2 receptor agonist activated I NMDA-OUT, whereas an EP3 receptor agonist reduced it. Agonists of EP1 or EP4 receptors showed no significant effects on I NMDA-OUT. A direct perfusion of 3,5'-cyclic adenosine monophosphate (cAMP) through the patch pipette facilitated I NMDA-OUT, which was abolished by the presence of protein kinase A (PKA) inhibitor. Furthermore, facilitation of I NMDA-OUT caused by an EP2 receptor agonist was significantly suppressed by PKA inhibitor. Finally, the activation of BK channels through EP2 receptors facilitated the recovery phase of NMDA-induced dendritic beading in the primary cultured cortical neurons. These results suggest that a direct activation of BK channels by EP2 receptor-mediated signaling pathways plays neuroprotective roles in cortical neurons.


Subject(s)
Dinoprostone/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Carbazoles/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , In Vitro Techniques , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Methyl Ethers/pharmacology , Mice , Mice, Inbred C57BL , Pyrroles/pharmacology , Receptors, Prostaglandin E, EP1 Subtype/agonists , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/metabolism
11.
Pharmacology ; 96(3-4): 137-43, 2015.
Article in English | MEDLINE | ID: mdl-26228833

ABSTRACT

Platelets express the EP2, EP3 and EP4 receptors. Prostaglandin (PG) E2 has a biphasic effect on platelets. Low concentrations of PGE2 enhance platelet aggregation through the activation of the EP3 receptors, while at high concentrations it attenuates aggregation via the EP4 receptor. Consequently, EP3 receptor inhibition was shown to inhibit artherothrombosis, but had no influence on bleeding time in vivo. In this study, we investigated the role of the EP3 receptor in adhesion and thrombus formation under flow conditions in vitro. The EP3 agonist sulprostone caused an increase in the adhesion of washed platelets to fibrinogen as well as to collagen under low shear stress, an effect that was blocked by the EP3 antagonist L-798106. In contrast, when whole blood was perfused over collagen-coated surfaces, sulprostone did not enhance binding and thrombus formation of platelets on collagen; at high concentrations it even attenuated this response. We conclude that in more physiological models of thrombus formation, the role for EP3 receptors is limited, indirectly suggesting that the primary action of PGE2 in haemostasis might be an inhibitory one.


Subject(s)
Blood Platelets/metabolism , Dinoprostone/analogs & derivatives , Platelet Adhesiveness/drug effects , Receptors, Prostaglandin E, EP3 Subtype/agonists , Thrombosis/blood , Blood Platelets/drug effects , Collagen/pharmacology , Dinoprostone/pharmacology , Fibrinogen/pharmacology , Humans , In Vitro Techniques , Platelet Aggregation/drug effects , Sulfonamides/pharmacology , Thrombosis/chemically induced
12.
Am J Respir Cell Mol Biol ; 48(3): 322-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23221043

ABSTRACT

Migration of airway smooth muscle (ASM) cells plays an important role in the pathophysiology of airway hyperresponsiveness and remodeling in asthma. It has been reported that prostaglandin (PG)E2 inhibits migration of ASM cells. Although PGE2 regulates cellular functions via binding to distinct prostanoid EP receptors, the role of EP receptor subtypes in mechanisms underlying cell migration has not been fully elucidated. We investigated the role of EP receptors in the inhibitory effects of PGE2 on the migration of human ASM cells. Migration induced by platelet-derived growth factor (PDGF)-BB (10 ng/ml, 6 h) was assessed by a chemotaxis chamber assay. PDGF-BB-induced cell migration was inhibited by PGE2, the specific EP2 agonist ONO-AE1-259-01, the specific EP4 agonist ONO-AE1-329, and cAMP-mobilizing agents. The inhibition of cell migration by PGE2 was significantly reversed by a blockade of EP2 and EP4 receptors using antagonists or transfection with siRNAs. Moreover, PGE2, the EP2 agonist, and the EP4 agonist significantly increased phosphorylation of small heat shock protein 20, one of the protein substrates for protein kinase A (PKA), with depolymerization of actin. In contrast, the EP3 agonist ONO-AE-248 significantly promoted baseline cell migration without affecting PDGF-BB-induced cell migration. In summary, activation of EP2 and EP4 receptors and subsequent activation of the cAMP/PKA pathway are the main mechanisms of inhibition of ASM cell migration by PGE2. HSP20 phosphorylation by PKA is possibly involved in this mechanism. Conversely, EP3 is potent in promoting cell migration. EP receptor subtypes may be novel therapeutic target molecules in airway remodeling and asthma.


Subject(s)
Cell Movement/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Respiratory System/metabolism , Actin Depolymerizing Factors/metabolism , Actins/drug effects , Cell Movement/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/pharmacology , HSP20 Heat-Shock Proteins/metabolism , Humans , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Respiratory System/drug effects , Stress Fibers/drug effects
13.
Mol Pharmacol ; 83(1): 206-16, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23087260

ABSTRACT

Unlike the majority of G protein-coupled receptors, the prostaglandin E(2) (PGE(2)) E-prostanoid 3 (EP3) receptor binds agonist with high affinity that is insensitive to the presence of guanosine 5[prime]-O-(3-thio)triphosphate (GTPγS). We report the identification of mutations that confer GTPγS sensitivity to agonist binding. Seven point mutations were introduced into the conserved motif in the second extracellular loop (ECII) of EP3, resulting in acquisition of GTP-sensitive agonist binding. One receptor mutation W203A was studied in detail. Loss of agonist binding was observed on intact human embryonic kidney 293 cells expressing the W203A receptor, conditions where high GTP levels are present; however, high affinity binding [(3)H]PGE(2) was observed in broken cell preparations washed free of GTP. The [(3)H]PGE(2) binding of W203A in broken cell membrane fractions was inhibited by addition of GTPγS (IC(50) 21 ± 1.8 nM). Taken together, these results suggest that the wild-type EP3 receptor displays unusual characteristics of the complex coupled equilibria between agonist-receptor and receptor-G protein interaction. Moreover, mutation of ECII can alter this coupled equilibrium from GTP-insensitive agonist binding to more conventional GTP-sensitive binding. This suggests that for the mutant receptors, ECII plays a critical role in linking the agonist bound receptor conformation to the G protein nucleotide bound state.


Subject(s)
Dinoprostone/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Receptors, Prostaglandin E, EP3 Subtype/agonists , Animals , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Point Mutation , Protein Conformation , Rabbits , Radioligand Assay , Receptors, Prostaglandin E, EP3 Subtype/genetics , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Signal Transduction
14.
Pflugers Arch ; 463(4): 593-602, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22371141

ABSTRACT

Prostaglandin E(2) (PGE(2)), an important and ubiquitously present vasoactive eicosanoid, may either constrict or dilate systemic vascular beds. However, little is known about the vascular contractile responsiveness to and signaling pathways for PGE(2) at the chronic stage of type 2 diabetes. We hypothesized that PGE(2)-induced arterial contraction is augmented in type 2 diabetic Goto-Kakizaki (GK) rats via the protein kinase Cδ (PKCδ) pathway. Here, we investigated the vasoconstrictor effects of PGE(2) and of sulprostone (EP1-/EP3-receptor agonist) in rings cut from superior mesenteric arteries isolated from GK rats (37-44 weeks old). In arteries from GK rats (vs. those from age-matched Wistar rats), examined in the presence of a nitric oxide synthase inhibitor: 1) the PGE(2)- and sulprostone-induced vasocontractions (which were not blocked by the selective EP1 receptor antagonist sc19220) were enhanced, and these enhancements were suppressed by rottlerin (selective PKCδ inhibitor) but not by Gö6976 (selective PKCα/ß inhibitor); 2) the sulprostone-stimulated phosphorylation of PKCδ (at Thr(505)), which yields an active form, was increased and 3) sulprostone-stimulated caldesmon phosphorylations, which are related to isometric force generation in smooth muscle, were increased. The protein expression of EP3 receptor in superior mesenteric arteries was similar between the two groups of rats. Our data suggest that the diabetes-related enhancement of EP3 receptor-mediated vasocontraction results from activation of the PKCδ pathway. Alterations in EP3 receptor-mediated vasocontraction may be important factors in the pathophysiological influences over arterial tone that are present in diabetic states.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Mesenteric Arteries/physiopathology , Protein Kinase C-delta/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Vasoconstriction , Animals , Calmodulin-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/enzymology , Dinoprostone/pharmacology , Male , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Phosphorylation , Rats , Rats, Wistar , Receptors, Prostaglandin E, EP3 Subtype/agonists , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
15.
Invest Ophthalmol Vis Sci ; 63(2): 24, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35147658

ABSTRACT

Purpose: We investigated the intraocular pressure (IOP)-lowering effect of topical sepetaprost (SPT), a dual agonist of the FP and EP3 receptors. We explored whether certain receptors mediated the hypotensive effect of SPT and outflow facility changes in C57BL/6 mice (wild-type [WT]) and FP and EP3 receptor-deficient mice (FPKO and EP3KO mice, respectively). Methods: IOP was measured using a microneedle. Outflow facility was measured using a two-level, constant-pressure perfusion method. Results: SPT significantly reduced IOP for 8 hours after administration to WT mice. The 2-hour IOP reductions afforded by latanoprost were 15.3 ± 2.5, 1.8 ± 2.0, and 12.3 ± 2.4% in WT, FPKO, and EP3KO mice, respectively; the SPT figures were 13.6 ± 2.1, 5.9 ± 2.7, and 6.6 ± 2.6%, respectively. Latanoprost-mediated IOP reduction was significantly decreased in FPKO mice, and SPT-mediated IOP reduction was reduced in both FPKO and EP3KO mice. At 6 hours after administration, latanoprost did not significantly reduce the IOP in any tested mouse strain. SPT-mediated IOP reduction was reduced in both FPKO and EP3KO mice. IOP reduction at 6 hours was significantly higher after simultaneous administration of selective FP and EP3 receptor agonists, but IOP did not fall on administration of (only) a selective EP3 receptor agonist. SPT significantly increased outflow facility in WT mice, but less so in FPKO and EP3KO mice. Conclusions: The IOP-lowering effect of SPT lasted longer than that of latanoprost. Our data imply that this may be attributable to augmented outflow facility mediated by the FP and EP3 receptors.


Subject(s)
Antihypertensive Agents/therapeutic use , Intraocular Pressure/drug effects , Oxepins/therapeutic use , Receptors, Prostaglandin E, EP3 Subtype/physiology , Receptors, Prostaglandin/physiology , Administration, Ophthalmic , Animals , Aqueous Humor/physiology , Dinoprostone/analogs & derivatives , Dinoprostone/therapeutic use , Intraocular Pressure/physiology , Latanoprost/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ophthalmic Solutions , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin E, EP3 Subtype/agonists , Time Factors , Tonometry, Ocular
16.
J Thromb Thrombolysis ; 32(2): 158-66, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21424266

ABSTRACT

Atherosclerosis has an important inflammatory component. Macrophages accumulating in atherosclerotic arteries produce prostaglandin E(2) (PGE(2)), a main inflammatory mediator. Platelets express inhibitory receptors (EP(2), EP(4)) and a stimulatory receptor (EP(3)) for this prostanoid. Recently, it has been reported in ApoE(-/-) mice that PGE(2) accumulating in inflammatory atherosclerotic lesions might contribute to atherothrombosis after plaque rupture by activating platelet EP(3), and EP(3) blockade has been proposed to be a promising new approach in anti-thrombotic therapy. The aim of our investigation was to study the role of PGE(2) in human atherosclerotic plaques on human platelet function and thrombus formation. Plaque PGE(2) might either activate or inhibit platelets depending on stimulation of either EP(3) or EP(4), respectively. We found that the two EP(3)-antagonists AE5-599 (300 nM) and AE3-240 (300 nM) specifically and completely inhibited the synergistic effect of the EP(3)-agonist sulprostone on U46619-induced platelet aggregation in blood. However, these two EP(3)-antagonists neither inhibited atherosclerotic plaque-induced platelet aggregation, GPIIb/IIIa exposure, dense and alpha granule secretion in blood nor reduced plaque-induced platelet thrombus formation under arterial flow. The EP(4)-antagonist AE3-208 (1-3 µM) potentiated in combination with PGE(2) (1 µM) ADP-induced aggregation, demonstrating that PGE(2) enhances platelet aggregation when the inhibitory EP(4)-receptor is inactivated. However, plaque-induced platelet aggregation was not augmented after platelet pre-treatment with AE3-208, indicating that plaque PGE(2) does not stimulate the EP(4)-receptor. We found that PGE(2) was present in plaques only at very low levels (15 pg PGE(2)/mg plaque). We conclude that PGE(2) in human atherosclerotic lesions does not modulate (i.e. stimulate or inhibit) atherothrombosis in blood after plaque rupture.


Subject(s)
Blood Platelets/metabolism , Carotid Stenosis/metabolism , Dinoprostone/metabolism , Plaque, Atherosclerotic/metabolism , Platelet Aggregation , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Thrombosis/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Abortifacient Agents, Nonsteroidal/pharmacology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Blood Platelets/pathology , Carotid Stenosis/genetics , Carotid Stenosis/pathology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Female , Humans , Male , Mice , Mice, Knockout , Naphthalenes , Phenylbutyrates , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics , Rupture, Spontaneous , Thrombosis/genetics , Vasoconstrictor Agents/pharmacology
17.
J Allergy Clin Immunol ; 126(6): 1218-25.e10, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20947153

ABSTRACT

BACKGROUND: Stevens-Johnson syndrome (SJS) and its severe variant, toxic epidermal necrolysis (TEN), are acute inflammatory vesiculobullous reactions of the skin and mucosa. They often affect the ocular surface and can result in permanent visual dysfunction. OBJECTIVES: We sought to discover genetic markers for SJS/TEN susceptibility. METHODS: We performed a genome-wide association study with 60 patients and 300 control subjects. We applied stringent filter and visual assessments for selecting single nucleotide polymorphisms (SNPs) and a high false discovery rate threshold. We fine-mapped the region where a candidate SNP was found and confirmed the results by means of sequencing. We evaluated the function of agonist-activated prostaglandin E receptor 3 (EP3), the gene for which contained several SNPs, in regulating cytokine production in human conjunctival epithelial (CE) cells. The expression levels of EP3 in the CE cells from patients and control subjects were also compared. RESULTS: We identified 3 SNPs that passed the false discovery rate threshold. One (rs17131450) was close to the EP3 gene. Therefore we analyzed the EP3 region in detail and identified 5 other SNPs. We confirmed the association between SJS/TEN and all 6 SNPs. Activated EP3 was expressed in control CE cells, and it suppressed polyI:C-stimulated cytokine production, suggesting that EP3 might help prevent ocular surface inflammation. Concordantly, the EP3 levels were much lower in the CE cells of the patients than in those of the control subjects. CONCLUSION: We demonstrated, using both genetic and functional analyses, that EP3 could be a key player in the pathogenesis of SJS/TEN accompanied by ocular complications.


Subject(s)
Epithelial Cells/metabolism , Receptors, Prostaglandin E, EP3 Subtype/genetics , Stevens-Johnson Syndrome/genetics , Cell Line , Conjunctiva/pathology , Cytokines/genetics , Cytokines/metabolism , DNA Mutational Analysis , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Inflammation , Polymorphism, Genetic , Receptors, Prostaglandin E, EP3 Subtype/agonists , Stevens-Johnson Syndrome/pathology , Stevens-Johnson Syndrome/physiopathology
18.
Eur J Pharmacol ; 863: 172693, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31560870

ABSTRACT

Corticotropin-releasing factor (CRF) plays an important role in sympathetic regulation. Centrally administered CRF elevates plasma catecholamine levels, resulting in CRF-dependent hypertension and tachycardia. We previously reported that brain thromboxane A2 mediates CRF-induced elevation of plasma adrenaline levels, whereas prostanoids other than thromboxane A2 mediate elevations in plasma noradrenaline levels. However, the mechanism by which CRF induces elevations in plasma noradrenaline levels remains unknown. Previous studies have revealed that brain prostaglandin (PG) E2, but not other PGs, causes sympathetic activation. In this study, we examined the roles of brain PGE2 and its receptors in CRF-induced elevation of plasma noradrenaline levels in rats. Our results showed that intracerebroventricular pretreatment with an antagonist of the PGE2 receptor EP3 subtype, but not other subtypes, suppressed CRF-induced elevations in plasma noradrenaline levels. We also examined the role of PGE2 and EP3 receptors in the paraventricular hypothalamic nucleus (PVN), the major integrative center for sympathetic regulation, in CRF-induced elevation of plasma noradrenaline levels. Centrally administered CRF increased PGE2 levels in PVN microdialysates, and microinjection of an EP3 receptor agonist into the PVN elevated plasma noradrenaline levels. Bilateral blockade of EP3 receptors in the PVN suppressed the elevation of plasma noradrenaline levels evoked by intracerebroventricular administration and PVN-microinjection of CRF. Our results suggest that CRF stimulates PGE2 release into the PVN that activates EP3 receptors in the PVN, resulting in the elevation of plasma noradrenaline levels.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Norepinephrine/blood , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Animals , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Drug Interactions , Male , Rats , Rats, Wistar , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors
19.
Article in English | MEDLINE | ID: mdl-29883889

ABSTRACT

Sulprostone is a potent prostaglandin E2 (PGE2) analogue and one of the first identified selective G-protein-coupled receptor 3 (EP3) agonists. It has been investigated as a potential antiulcer agent and frequently used in the research of EP3 antagonist. To assist pharmacokinetic and pharmacodynamic studies, a rapid and sensitive LC-MS/MS method was developed and qualified for the quantitation of sulprostone in monkey plasma. Using electrospray ionization mass spectrometry, an ammonium adduct in positive mode was chosen for analysis which had seven times of the sensitivity of the depronated ion in negative mode. Latanoprost, a prostaglandin F2α analogue, was used as the internal standard while good sensitivity and chromatography were obtained on a 2.6 µm core-shell column with pentafluorophenyl stationary phase. An assay dynamic range of 2 to 4000 ng/mL was achieved with a sample volume of 25 µL plasma on a Sciex API4000 instrument with simple protein precipitation. Several esterase inhibitors including sodium fluoride (NaF), phenylmethanesulfonyl fluoride (PMSF), diisopropylfluorophosphate (DFP), paraoxon and dichlorvos as well as wet ice conditions were explored for the stabilization of sulprostone in monkey plasma. The developed method was successfully applied for the evaluation of pharmacokinetics of sulprostone after intravenous administration of 0.5 mg/kg to cynomolgus monkey.


Subject(s)
Chromatography, Liquid/methods , Dinoprostone/analogs & derivatives , Tandem Mass Spectrometry/methods , Animals , Dinoprostone/blood , Dinoprostone/chemistry , Dinoprostone/pharmacokinetics , Drug Stability , Linear Models , Macaca fascicularis , Male , Receptors, Prostaglandin E, EP3 Subtype/agonists , Reproducibility of Results , Sensitivity and Specificity
20.
Drug Discov Today ; 22(1): 57-71, 2017 01.
Article in English | MEDLINE | ID: mdl-27506873

ABSTRACT

Prostaglandin E2 is a potent endogenous molecule that binds to four different G-protein-coupled receptors: EP1-4. Each of these receptors is a valuable drug target, with distinct tissue localisation and signalling pathways. We review the structural features of EP modulators required for subtype-selective activity, as well as the structural requirements for improved pharmacokinetic parameters. Novel EP receptor subtype selective agonists and antagonists appear to be valuable drug candidates in the therapy of many pathophysiological states, including ulcerative colitis, glaucoma, bone healing, B cell lymphoma, neurological diseases, among others, which have been studied in vitro, in vivo and in early phase clinical trials.


Subject(s)
Pharmaceutical Preparations/chemistry , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Animals , Dinoprostone/chemistry , Drug Design , Humans , Ligands , Molecular Structure , Molecular Targeted Therapy , Protein Binding , Receptors, Prostaglandin E, EP1 Subtype/agonists , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL