Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 912
Filter
1.
Invest Ophthalmol Vis Sci ; 61(8): 44, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32725213

ABSTRACT

Purpose: Cyclic adenosine monophosphate (cAMP) and peroxisome proliferator-activated receptor alpha (PPARα) levels mediate extracellular matrix (ECM) changes by altering the levels of hypoxia-inducible factor 1-alpha (HIF-1α) in various tissues. We aimed to determine, in the sclera of guinea pigs, whether a prostanoid receptor (EP2)-linked cAMP modulation affects PPARα and HIF-1α signaling during myopia. Methods: Three-week-old guinea pigs (n = 20 in each group), were monocularly injected with either an EP2 agonist (butaprost 1 µmol/L/10 µmol/L), an antagonist (AH6809 10 µmol/L/30 µmol/L) or a vehicle solution for two weeks during normal ocular growth. Separate sets of animals received these injections and underwent form deprivation (FD) simultaneously. Refraction and axial length (AL) were measured at two weeks, followed by scleral tissue isolation for quantitative PCR (qPCR) analysis (n = 10) and cAMP detection (n = 10) using a radioimmunoassay. Results: Butaprost induced myopia development during normal ocular growth, with proportional increases in AL and cAMP levels. FD did not augment the magnitude of myopia or cAMP elevations in these agonist-injected eyes. AH6809 suppressed cAMP increases and myopia progression during FD, but had no effect in a normal visual environment. Of the diverse set of 27 genes related to cAMP, PPARα and HIF-1α signaling and ECM remodeling, butaprost differentially regulated 15 of them during myopia development. AH6809 injections during FD negated such differential gene expressions. Conclusion: EP2 agonism increased cAMP and HIF-1α signaling subsequent to declines in PPARα and RXR mRNA levels, which in turn decreased scleral fibrosis and promoted myopia. EP2 antagonism instead inhibited each of these responses. Our data suggest that EP2 suppression may sustain scleral ECM structure and inhibit myopia development.


Subject(s)
Alprostadil/analogs & derivatives , Extracellular Matrix , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myopia, Degenerative , PPAR alpha/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Xanthones/pharmacology , Alprostadil/pharmacology , Animals , Cyclic AMP/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Guinea Pigs , Myopia, Degenerative/etiology , Myopia, Degenerative/metabolism , Myopia, Degenerative/prevention & control , Prostaglandin Antagonists/pharmacology , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Signal Transduction
2.
Dig Dis Sci ; 64(10): 2806-2814, 2019 10.
Article in English | MEDLINE | ID: mdl-30989466

ABSTRACT

BACKGROUND: The prostaglandin D2 receptor DP2 has been implicated in eosinophil infiltration and the development of eosinophilic esophagitis (EoE). AIMS AND METHODS: In this study, we investigated an involvement of PGE2 (EP1-EP4) and PGD2 (DP1) receptors in EoE by measuring their expression in peripheral blood eosinophils and esophageal mucosal biopsies of EoE patients and by performing migration and adhesion assays with eosinophils from healthy donors. RESULTS: Expression of EP2 and EP4, but not EP1 and EP3, was decreased in blood eosinophils of patients with EoE vs. control subjects. Adhesion of eosinophils to esophageal epithelial cells was decreased by EP2 receptor agonist butaprost and EP4 agonist ONO-AE1-329, whereas DP1 agonist BW245C increased adhesion. In chemotaxis assays with supernatant from human esophageal epithelial cells, only ONO-AE1-329 but not butaprost or BW245C inhibited the migration of eosinophils. Expression of EP and DP receptors in epithelial cells and eosinophils was detected in sections of esophageal biopsies from EoE patients by immunohistochemistry. qPCR of biopsies from EoE patients revealed that gene expression of EP4 and DP1 was the highest among PGE2 and PGD2 receptors. Esophageal epithelial cells in culture showed high gene expression for EP2 and EP4. Activation of EP2 and EP4 receptors decreased barrier integrity of esophageal epithelial cells in impedance assays. CONCLUSIONS: Activation of EP2 and EP4 receptors may inhibit eosinophil recruitment to the esophageal mucosa. However, their activation could negatively affect esophageal barrier integrity suggesting that eosinophilic rather than epithelial EP2 and EP4 have a protective role in EoE.


Subject(s)
Eosinophilic Esophagitis , Eosinophils , Esophageal Mucosa , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Cell Adhesion , Cell Migration Assays/methods , Cells, Cultured , Eosinophilic Esophagitis/blood , Eosinophilic Esophagitis/metabolism , Eosinophilic Esophagitis/pathology , Eosinophils/drug effects , Eosinophils/metabolism , Esophageal Mucosa/drug effects , Esophageal Mucosa/metabolism , Esophageal Mucosa/pathology , Humans , Immunohistochemistry , Methyl Ethers/pharmacology , Pilot Projects , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/analysis , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/analysis
3.
Int J Dev Neurosci ; 71: 46-51, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30157453

ABSTRACT

Activation of peroxisome proliferator-activated receptor alpha (PPARα) has been reported to modulate cell proliferation, migration, and differentiation in astrocytes. In this study, we used a retinoic acid (RA)-induced differentiation model of NTERA-2/clone D1 (NT2) cells to explore the functional significance of PPARα in neuronal differentiation. We found that activating PPARα by Wy14643 accelerated neuronal differentiation via regulating the expression of neuronal markers. RT-PCR assays showed a significant increase in NeuroD expression and a decrease in nestin expression in cells treated concomitantly with RA and Wy14643 for 2 days compared to the levels in cells treated with RA alone. Expression of MAP2 protein, a mature neuronal marker, was markedly upregulated at day 10 of Wy14643 treatment, which was maintained after 21 days of neuronal formation. Corresponding to the changes in MAP2 expression, the expression of Cdk5 was upregulated with Wy14643 exposure from day 10 to day 21. Moreover, cells treated with Wy14643 displayed higher expression levels of phospho-ERK and phospho-p38 in the differentiation process than cell treated with RA alone. These results indicated that activation of PPARα accelerated neuronal differentiation through upregulating the expression of NeuroD, MAP2, and Cdk5 and downregulating the expression of nestin. MAPK signals, ERK and p38, might contribute to the accelerated differentiation process. These findings suggest that PPARα plays a role in regulating neuronal differentiation and may be beneficial for functional recovery from neurological disorders.


Subject(s)
Cell Differentiation/drug effects , Neurons/drug effects , PPAR alpha/pharmacology , Signal Transduction/drug effects , Analysis of Variance , Cell Line, Tumor , Cell Movement/drug effects , Cyclin-Dependent Kinase 5/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Microtubule-Associated Proteins/metabolism , Nestin/metabolism , Prostaglandins E, Synthetic/pharmacology , Teratocarcinoma/pathology , Time Factors , Tretinoin/pharmacology
4.
Curr Drug Deliv ; 14(4): 473-482, 2017.
Article in English | MEDLINE | ID: mdl-27834148

ABSTRACT

BACKGROUND: There is a general consensus that sleep-wake cycle is controlled by neuroanatomical, neurochemical and molecular systems as well as by homeostatic and circadian complex networks. The research has shown that a molecular element that could be displaying a relevant role in the modulation of sleep is the peroxisome proliferator-activated receptor alpha (PPARα), which belongs to the family of nuclear receptor ligand-activated transcription factors that includes PPARß/δ and PPARγ. A growing body of evidence supports the notion that PPARα is activated by natural ligands such as the anorexic lipid mediator oleoylethanolamide (OEA) or synthetic compounds including Wy14643 whereas antagonists like MK-886 block the neurobiological outcomes of PPARα. More recently, studies have reported the permissive role of PPARα by modulating diverse neurobiological functions such as inflammation, metabolic disorders, learning, degenerative diseases and sleep. Remarkably, this nuclear receptor has been described in sleep-related brain regions leading to the hypothesis that PPARα might be involved in sleep modulation inasmuch as activation of this protein promotes a robust enhancement of wakefulness while reduces sleep. OBJECTIVE: In this mini review, the emerging evidence of the putative role of PPARα in sleep control is highlighted. Even though the data are derived from new areas of research, there are many reasons to believe that understanding and appreciation of PPARα functions may provide knowledge of possible mechanisms of action activated by this nuclear receptor in sleep modulation. CONCLUSION: Novel insights of therapeutic intervention for sleep disorders might be visualized targeting the function of PPARα in sleep abnormalities.


Subject(s)
PPAR alpha/physiology , Sleep Wake Disorders/physiopathology , Endocannabinoids/pharmacology , Humans , Indoles/pharmacology , Oleic Acids/pharmacology , Prostaglandins E, Synthetic/pharmacology
5.
Bioorg Med Chem ; 20(11): 3502-22, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22546206

ABSTRACT

To identify topically effective EP4 agonists and EP2/EP4 dual agonists with excellent subtype selectivity, further optimization of the 16-phenyl ω-chain moiety of the γ-lactam 5-thia prostaglandin E analog and the 2-mercaptothiazole-4-carboxylic acid analog were undertaken. Rat in vivo evaluation of these newly identified compounds as their poly (lactide-co-glycolide) microsphere formulation, from which sustained release of the test compound is possible, led us to discover compounds that showed efficacy in a rat bone fracture healing model after its topical administration without serious influence on blood pressure and heart rate. A structure-activity relationship study is also presented.


Subject(s)
Lactams/chemical synthesis , Lactams/pharmacology , Prostaglandins E, Synthetic/chemical synthesis , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/agonists , Administration, Topical , Animals , Blood Pressure/drug effects , CHO Cells , Cricetinae , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Dinoprostone/chemistry , Drug Evaluation, Preclinical/methods , Fracture Healing/drug effects , Heart Rate/drug effects , Lactams/administration & dosage , Male , Mice , Microspheres , Molecular Structure , Polyglactin 910/administration & dosage , Polyglactin 910/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thiazolidines/chemistry
6.
Chem Pharm Bull (Tokyo) ; 59(12): 1494-508, 2011.
Article in English | MEDLINE | ID: mdl-22130372

ABSTRACT

For the purpose of discovering an orally available EP4 subtype-selective agonist, a series of 8-aza prostaglandin E(1) (PGE(1)) analogs were synthesized and evaluated for their affinity for PGE(2) receptor subtypes. Additionally, the structure-activity relationships of these compounds were studied. Among the tested compounds, the 8-aza PGE(1) analog 6 and 8-aza-5-thiaPGE(1) analog 12 had highly potent EP4 receptor affinity, good functional activity, and excellent subtype-selectivity. Furthermore, these analogs demonstrated good stability in human liver microsomes. As a result, we concluded that these two series of 8-aza PGE(1) analogs could be promising chemical leads for an orally available EP4 subtype-selective agonist.


Subject(s)
Alprostadil/analogs & derivatives , Prostaglandins E, Synthetic/chemistry , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/agonists , Alprostadil/chemical synthesis , Alprostadil/chemistry , Alprostadil/metabolism , Alprostadil/pharmacology , Humans , Microsomes, Liver/metabolism , Prostaglandins E, Synthetic/chemical synthesis , Prostaglandins E, Synthetic/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism
7.
Chem Pharm Bull (Tokyo) ; 59(12): 1523-34, 2011.
Article in English | MEDLINE | ID: mdl-22130375

ABSTRACT

Analogs 8-aza-16-aryl prostaglandin E(1) (PGE(1)) and 8-aza-5-thia-16-arylPGE(1) were synthesized and evaluated with respect to their subtype receptor affinity and EP4 agonist activity for the purposes of identifying subtype-selective EP4 agonists that demonstrate oral efficacy. Using an inhibition assay of lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-α production in rats, representative compounds were evaluated for their pharmacokinetic profiles and in vivo efficacy. Structure-activity relationships (SARs) were characterized and presented. Of the compounds tested, several demonstrated better oral exposure and/or in vivo efficacy compared with the previously reported analog 2a.


Subject(s)
Alprostadil/analogs & derivatives , Prostaglandins E, Synthetic/chemistry , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/agonists , Administration, Oral , Alprostadil/administration & dosage , Alprostadil/chemical synthesis , Alprostadil/chemistry , Alprostadil/pharmacokinetics , Alprostadil/pharmacology , Animals , Humans , Lipopolysaccharides/immunology , Prostaglandins E, Synthetic/administration & dosage , Prostaglandins E, Synthetic/chemical synthesis , Prostaglandins E, Synthetic/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP4 Subtype/immunology , Tumor Necrosis Factor-alpha/immunology
8.
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
9.
J Recept Signal Transduct Res ; 29(5): 246-56, 2009.
Article in English | MEDLINE | ID: mdl-19627252

ABSTRACT

Interleukin-1 (IL-1alpha) induced inflammatory and pro-fibrotic responses in human lung fibroblasts are mediated by activation of MAPK and NFkappaB pathways. The purpose of the present study was to broadly profile the activity of a variety of compounds which function as inhibitors of these key signaling pathways that may affect IL-1alpha mediated gene changes. A reference set of genes was derived from microarray analysis of IL-1alpha stimulated cells. The genes were chosen to provide a range of expression profiles which serve to represent the actions of the underlying signaling network. We show that G(s)-coupled receptor agonists have a unique pattern of activity as represented by their impact on IL-1alpha dependent gene changes. These effects were not mimicked by direct inhibitors of p38, JNK, MEK or IKK but were mimicked by forskolin and cAMP analogs. These findings indicate that cAMP/PKA serves as a point of convergence for regulation of IL-1alpha responses by multiple G(s)-coupled receptors and regulates IL-1alpha responses by a distinct mechanism that does not solely involve direct inhibition of p38, JNK, MEK or IKK. The data also point to a potentially useful paradigm wherein monitoring of a small subset of genes is sufficient to identify pathway activity of novel compounds.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Profiling , Interleukin-1alpha/pharmacology , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Anti-Ulcer Agents/pharmacology , Calcium/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Humans , Hydantoins/pharmacology , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Iloprost/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/cytology , Lung/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Misoprostol/pharmacology , Oligonucleotide Array Sequence Analysis , Platelet Aggregation Inhibitors/pharmacology , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin/agonists , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Br J Pharmacol ; 158(1): 372-81, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19486006

ABSTRACT

BACKGROUND AND PURPOSE: Cyclooxygenase inhibitors function to reduce levels of prostaglandin E(2) (PGE(2)) and are broadly efficacious in models of bladder overactivity. We therefore investigated a regulation of urinary bladder function in conscious rats by modulation of the EP(3) receptor for PGE(2). EXPERIMENTAL APPROACH: The activity of the EP(3) receptor agonist GR63799X, and EP(3) receptor antagonists, CM9 and DG041, at recombinant EP(3) receptors was evaluated in vitro. In vivo, intraduodenal dosing during conscious, continuous-filling cystometry of spontaneously hypertensive rats was utilized to determine the urodynamic effect of EP(3) receptor modulation. KEY RESULTS: GR63799X dose-dependently (0.001-1 mg x kg(-1)) reduced bladder capacity, as indicated by a reduction in both the micturition interval and volume of urine per void. In contrast, CM9 (10 and 30 mg x kg(-1)) and DG041 (30 mg x kg(-1)) enhanced bladder capacity, as indicated by significantly longer micturition intervals and larger void volumes. CM9 and DG041 inhibited the responses to GR63799X supporting the in vivo activity of these pharmacological agents at the EP(3) receptor. In addition to its effect on bladder capacity, GR63799X increased endogenous urine production. Intra-arterial infusion of saline mimicked the enhancement of urine flow observed with GR63799X, and the response was inhibited by CM9. CONCLUSIONS AND IMPLICATIONS: These data support the EP(3) receptor as a modulator of urinary bladder activity in the conscious rat, and in addition, indicate a role for EP(3) receptor activity in regulating urine flow.


Subject(s)
Consciousness , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/physiology , Urinary Bladder/physiology , Urination/physiology , Animals , Cell Line , Consciousness/physiology , Female , Humans , Prostaglandins E, Synthetic/chemical synthesis , Prostaglandins E, Synthetic/pharmacology , Rats , Rats, Inbred SHR , Receptors, Prostaglandin E, EP3 Subtype , Urinary Bladder/drug effects , Urination/drug effects
11.
Am J Physiol Gastrointest Liver Physiol ; 296(6): G1180-90, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19359421

ABSTRACT

Gastric emptying depends on functional coupling of slow waves between the corpus and antrum, to allow slow waves initiated in the gastric corpus to propagate to the pyloric sphincter and generate gastric peristalsis. Functional coupling depends on a frequency gradient where slow waves are generated at higher frequency in the corpus and drive the activity of distal pacemakers. Simultaneous intracellular recording from corpus and antrum was used to characterize the effects of PGE(2) on slow waves in the murine stomach. PGE(2) increased slow-wave frequency, and this effect was mimicked by EP(3), but not by EP(2), receptor agonists. Chronotropic effects were due to EP(3) receptors expressed by intramuscular interstitial cells of Cajal because these effects were not observed in W/W(V) mice. Although the integrated chronotropic effects of EP(3) receptor agonists were deduced from electrophysiological experiments, no clear evidence of functional uncoupling was observed with two-point electrical recording. Gastric peristalsis was also monitored by video imaging and spatiotemporal maps to study the impact of chronotropic agonists on propagating contractions. EP(3) receptor agonists increased the frequency of peristaltic contractions and caused ectopic sites of origin and collisions of peristaltic waves. The impact of selective regional application of chronotropic agonists was investigated by use of a partitioned bath. Antral slow waves followed enhanced frequencies induced by stimulation of the corpus, and corpus slow waves followed when slow-wave frequency was elevated in the antrum. This demonstrated reversal of slow-wave propagation with selective antral chronotropic stimulation. These studies demonstrate the impact of chronotropic agonists on regional intrinsic pacemaker frequency and integrated gastric peristalsis.


Subject(s)
Peristalsis/drug effects , Peristalsis/physiology , Prostaglandins/pharmacology , Stomach/drug effects , Stomach/physiology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Biological Clocks/drug effects , Biological Clocks/physiology , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Prostaglandins/agonists , Prostaglandins E, Synthetic/pharmacology , Pyloric Antrum/cytology , Pyloric Antrum/drug effects , Pyloric Antrum/physiology , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Stomach/cytology
12.
Am J Physiol Renal Physiol ; 295(2): F507-14, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18508878

ABSTRACT

Nonsteroidal anti-inflammatory cyclooxygenase inhibitors that function to reduce prostaglandin E2 (PGE2) production have been widely reported as effective agents in models of urinary bladder overactivity. We therefore investigated a potential role for the PGE2 receptor, EP3, in urinary bladder function by performing conscious, freely moving cystometry on EP3 receptor knockout (KO) mice. EP3 KO mice demonstrated an enhanced bladder capacity compared with wild-type (WT) mice ( approximately 185% of WT) under control conditions, based on larger voided and infused bladder volumes. Infusion of the EP3 receptor agonist GR63799X into the bladder of WT mice reduced the bladder capacity. This was ineffective in EP3 KO mice that demonstrated a time-dependent increase in bladder capacity with GR63799X, an effect similar to that observed with vehicle in both genotypes. In addition, infusion of PGE2 into WT mice induced bladder overactivity, an effect that was significantly blunted in the EP3 KO mice. The data reported here provide the first evidence supporting a functional role for EP3 receptors in normal urinary bladder function and implicate EP3 as a contributor to bladder overactivity during pathological conditions of enhanced PGE2 production, as reported previously in overactive bladder patients.


Subject(s)
Dinoprostone/metabolism , Receptors, Prostaglandin E/metabolism , Urinary Bladder, Overactive/metabolism , Urinary Bladder/pathology , Urinary Bladder/physiopathology , Acetic Acid , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostaglandins E, Synthetic/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Urinary Bladder, Overactive/chemically induced
13.
J Dent Res ; 87(3): 262-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18296611

ABSTRACT

Orofacial inflammation is associated with prostaglandin release and the sensitization of nociceptive receptors such as the transient receptor potential subtype V(1) (TRPV(1)). We hypothesized that certain PGE(2) receptor subtypes (EP1-EP4) are co-expressed with TRPV(1) in trigeminal nociceptors and sensitize responses to a TRPV(1) agonist, capsaicin. Accordingly, combined in situ hybridization was performed with immunohistochemistry on rat trigeminal ganglia. We next evaluated the effects of specific EP2 and EP3 agonists (butaprost and sulprostone) in cultured trigeminal ganglia neurons. The results showed that EP2 and EP3 are expressed in trigeminal neurons (58% and 53% of total neurons, respectively) and are co-expressed in TRPV(1)-positive neurons (64% and 67 % of TRPV(1)-positive neurons, respectively). Moreover, most of the cells expressing EP2 or EP3 mRNA were of small to medium diameter (< 30 microm). The application of butaprost and sulprostone triggered neuropeptide exocytosis, and butaprost sensitized capsaicin responses. Analysis of these data, collectively, supports the hypothesis that prostaglandins regulate trigeminal TRPV(1) nociceptors via activation of the EP2 and EP3 receptors.


Subject(s)
Nociceptors/metabolism , Receptors, Prostaglandin E/metabolism , Trigeminal Ganglion/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Capsaicin/pharmacology , Cell Size , Cells, Cultured , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Exocytosis/drug effects , Immunohistochemistry , In Situ Hybridization , Male , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , Nociceptors/drug effects , Prostaglandins E, Synthetic/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , TRPV Cation Channels/agonists , Trigeminal Ganglion/drug effects
14.
Int J Gynaecol Obstet ; 99 Suppl 2: S168-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17961571

ABSTRACT

Cervical priming with misoprostol has shown to facilitate transcervical procedures and to reduce side-effects. Cervical priming is recommended by several evidence-based guidelines prior to surgical abortion, dilatation and curettage, hysteroscopy and intrauterine device insertion. It is effective in pregnant as well as in non-pregnant women while the results in post-menopausal women are conflicting. Misoprostol is the best suited prostaglandin for a number of reasons: it has a short half-life, few side effects, it is stable at room temperature, it is relatively cheap and the dosage can easily be adjusted according to the clinical need. Various doses, routes, and time intervals between misoprostol application and the intervention have been evaluated. A single dose of 400 microg given sublingually or vaginally 3h before the intervention has given the best efficacy with the least side effects. Higher doses or longer intervals do not improve the effect on the cervix. Pain is a frequent side effect, but usually responds well to NSAIDs. Other side effects are rare.


Subject(s)
Cervix Uteri/drug effects , Misoprostol , Prostaglandins E, Synthetic , Abortion, Therapeutic/methods , Administration, Intravaginal , Administration, Oral , Cervical Ripening/drug effects , Dilatation and Curettage/methods , Female , Humans , Hysteroscopy/methods , Misoprostol/administration & dosage , Misoprostol/adverse effects , Misoprostol/pharmacology , Pregnancy , Prostaglandins E, Synthetic/administration & dosage , Prostaglandins E, Synthetic/adverse effects , Prostaglandins E, Synthetic/pharmacology
15.
J Endocrinol ; 194(3): 595-602, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17761898

ABSTRACT

In luteinizing granulosa cells, prostaglandin E(2) (PGE(2)) can exert luteotrophic actions, apparently via the cAMP signalling pathway. In addition to stimulating progesterone synthesis, PGE(2) can also stimulate oxidation of the physiological glucocorticoid, cortisol, to its inactive metabolite, cortisone, by the type 1 11beta-hydroxysteroid dehydrogenase (11betaHSD1) enzyme in human granulosa-lutein cells. Having previously shown these human ovarian cells to express functional G-protein coupled, E-series prostaglandin (PTGER)1, PTGER2 and PTGER4 receptors, the aim of this study was to delineate the roles of PTGER1 and PTGER2 receptors in mediating the effects of PGE(2) on steroidogenesis and cortisol metabolism in human granulosa-lutein cells. PGE(2)-stimulated concentration-dependent increases in both progesterone production and cAMP accumulation (by 1.9 +/- 0.1- and 18.7 +/- 6.8-fold respectively at 3000 nM PGE(2)). While a selective PTGER1 antagonist, SC19220, could partially inhibit the steroidogenic response to PGE(2) (by 55.9 +/- 4.1% at 1000 nM PGE(2)), co-treatment with AH6809, a mixed PTGER1/PTGER2 receptor antagonist, completely abolished the stimulation of progesterone synthesis at all tested concentrations of PGE(2) and suppressed the stimulation of cAMP accumulation. Both PGE(2) and butaprost (a preferential PTGER2 receptor agonist) stimulated concentration-dependent increases in cortisol oxidation by 11betaHSD1 (by 42.5 +/- 3.1 and 40.0 +/- 3.0% respectively, at PGE(2) and butaprost concentrations of 1000 nM). Co-treatment with SC19220 enhanced the ability of both PGE(2) and butaprost to stimulate 11betaHSD1 activity (by 30.2 +/- 0.2 and 30.5 +/- 0.6% respectively), whereas co-treatment with AH6809 completely abolished the 11betaHSD1 responses to PGE(2) and butaprost. These findings implicate the PTGER2 receptor-cAMP signalling pathway in the stimulation of progesterone production and 11betaHSD1 activity by PGE(2) in human granulosa-lutein cells.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Dinoprostone/pharmacology , Luteal Cells/metabolism , Progesterone/biosynthesis , Receptors, Prostaglandin E/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Cells, Cultured , Cortisone/metabolism , Cyclic AMP/metabolism , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Hydrocortisone/metabolism , Luteal Cells/drug effects , Prostaglandin Antagonists/pharmacology , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Xanthones/pharmacology
16.
Arterioscler Thromb Vasc Biol ; 26(11): 2454-61, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16931794

ABSTRACT

OBJECTIVE: To understand the mechanism by which oxidants are linked to insulin resistance, bovine aortic endothelial cells were exposed to oxidized low-density lipoproteins (oxLDL) or peroxynitrite. METHODS AND RESULTS: OxLDL transiently increased phosphorylation of Erk and Akt within 5 minutes, but 60 minutes later, resulted in decreased insulin-induced Akt phosphorylation. OxLDL promoted a 2- to 5-fold increase in oxidant generation as measured by dihydrorhodamine or dihydroethidium oxidation that was ascribed to peroxynitrite. Exogenous peroxynitrite (25 to 100 micromol/L) or oxidized glutathione mimicked the effects of oxLDL. OxLDL increased the S-glutathiolation of p21ras, and adenoviral transfection with either a mutant p21ras (C118S) lacking the predominant site of S-glutathiolation or a dominant-negative mutant restored insulin-induced Akt phosphorylation. The requirement for oxidant-mediated S-glutathiolation and activation of p21ras in mediating insulin resistance was further implicated by showing that insulin signaling was restored by Mek inhibitors or by overexpression of glutaredoxin-1. Furthermore, oxLDL increased Erk-dependent phosphorylation of insulin receptor substrate-1 serine-616 that was prevented by inhibiting oxidant generation, Erk activation, or by the p21ras C118S mutant. CONCLUSIONS: This study provides direct evidence for a novel molecular mechanism by which oxidants can induce insulin resistance via S-glutathiolation of p21ras and Erk-dependent inhibition of insulin signaling.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Glutathione/metabolism , Insulin Resistance/physiology , Lipoproteins, LDL/pharmacology , Oncogene Protein p21(ras)/metabolism , Peroxynitrous Acid/pharmacology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Aorta/cytology , Cattle , Dinoprostone/agonists , Endothelial Cells/drug effects , Endothelial Cells/physiology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione Disulfide/pharmacology , Insulin/metabolism , Insulin Receptor Substrate Proteins , Lysophosphatidylcholines/pharmacology , Oncogene Protein p21(ras)/drug effects , Oxidants/pharmacology , Phosphoproteins/metabolism , Phosphorylation/drug effects , Prostaglandins E, Synthetic/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
17.
Invest Ophthalmol Vis Sci ; 47(9): 4042-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936121

ABSTRACT

PURPOSE: To investigate the ocular hypotensive effect of the prostanoid EP2 receptor agonist butaprost and to establish its mechanism of action. METHODS: All experiments were performed in cynomolgus monkeys after topical application of butaprost (0.1%). The effects of butaprost on aqueous humor flow were determined by fluorophotometry. Total outflow facility was measured by the two-level, constant-pressure perfusion method, and uveoscleral outflow was determined by perfusion of FITC-labeled dextran through the anterior chamber. Effects on ocular morphology were studied after tissue fixation with transcardial perfusion by paraformaldehyde and immersion fixation of the globe, in animals subjected to long-term treatment with butaprost. Conscious ocular normotensive monkeys and monkeys with unilateral ocular hypertension were used for intraocular pressure (IOP) studies. RESULTS: Butaprost had no significant effect on aqueous humor flow or total outflow facility in ocular normotensive monkeys. Uveoscleral outflow was significantly higher in the butaprost treated eyes than in vehicle treated eyes, 1.03 +/- 0.20 vs. 0.53 +/- 0.18 microL.min(-1). After a 1-year treatment with butaprost, the morphology of the ciliary muscle was changed, showing increased spaces between ciliary muscle bundles and the apparent formation of new outflow channels. In many instances, changes were observed in the trabecular meshwork as well. Butaprost, in a single 0.1% dose, decreased IOP significantly in ocular normotensive monkeys and reduced IOP in laser-induced glaucomatous monkey eyes to the same level as that in the ocular normotensive contralateral eyes. CONCLUSIONS: The prostanoid EP2 receptor agonist butaprost appears to lower IOP by increasing uveoscleral outflow, according to both physiological and morphologic findings. Although the prostanoid EP2 receptor is structurally and functionally distinct from the FP receptor, the effects of EP2 and FP receptor stimulation on aqueous humor outflow are similar.


Subject(s)
Alprostadil/analogs & derivatives , Aqueous Humor/metabolism , Intraocular Pressure/drug effects , Ocular Hypertension/drug therapy , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E/agonists , Sclera/drug effects , Uvea/drug effects , Administration, Topical , Alprostadil/pharmacology , Animals , Ciliary Body/drug effects , Ciliary Body/pathology , Dextrans/metabolism , Disease Models, Animal , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Fluorophotometry , Macaca fascicularis , Muscle, Smooth/drug effects , Muscle, Smooth/pathology , Ocular Hypertension/pathology , Receptors, Prostaglandin E, EP2 Subtype , Sclera/metabolism , Trabecular Meshwork , Uvea/metabolism
18.
Eur J Pharmacol ; 529(1-3): 16-23, 2006 Jan 04.
Article in English | MEDLINE | ID: mdl-16316649

ABSTRACT

The importance of arachidonic acid metabolites on the control of cell growth, particularly those derived from cyclooxygenase pathway has long been recognized. Recently, we observed that prostaglandin E(2) (PGE(2)) interaction with EP(1) and EP(4) receptors is involved in serum-induced 3T6 fibroblast growth due to their effect at various levels of the cell cycle machinery. This study shows that prostanoid EP(3) receptor was expressed in 3T6 fibroblast. We studied the role of EP(3) receptor agonist GR 63799X in serum-induced 3T6 cell proliferation. This was concentration-dependent inhibit (IC(50) approximately 10 microM) to a complete inhibition without any cytotoxic or proapoptotic effect. The prostanoid EP(3) receptor agonist treatment decreased the G(0)/G(1) and G(2)/M populations whereas cells were accumulated in S phase. This arrest in S phase was associated with a decrease in cyclin B levels and the enhancement of p21 expression. Our data show that EP(3) agonist decreases cAMP levels in our experimental conditions. Interestingly, the S arrest caused by prostanoid EP(3) receptor agonist seems to be cAMP dependent, at least in part, because forskolin treatment allowed S-arrested cells to progress through cell cycle and consequently growth. Thus, our results suggest that PGE(2) EP(3) receptor interaction may be involved in serum-induced 3T6 fibroblast growth due to their effects on cAMP levels and on cell cycle machinery of the S phase.


Subject(s)
Cell Proliferation/drug effects , Fibroblasts/drug effects , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E/agonists , S Phase/drug effects , Animals , Cell Line , Cyclic AMP/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Receptors, Prostaglandin E, EP3 Subtype
19.
Life Sci ; 78(16): 1878-83, 2006 Mar 13.
Article in English | MEDLINE | ID: mdl-16309709

ABSTRACT

The activation of glutamate receptors, particularly N-methyl-D-aspartate (NMDA) receptors, initiates ischemic cascade in the early stages of cerebral ischemia. Postischemia, cerebral ischemia is also associated with an inflammatory reaction that contributes to tissue damage. The up-regulation of neuronal cyclooxygenase-2 (COX-2) and elevation of prostaglandin E2 (PGE2) have been reported to occur after cerebral ischemic insult. We therefore studied whether the COX-2 reaction product PGE2 affects glutamate receptor-mediated cell death in cultured rat cortical cells. PGE2 was found to augment NMDA-mediated cell death. The transcription of EP1, EP2, EP3 and EP4 PGE2 receptor genes was investigated using reverse transcriptase-polymerase chain reaction (RT-PCR). EP1, EP2 and EP3 receptor genes were found in cortical cells. Butaprost (an EP2 agonist) markedly enhanced NMDA-mediated cell death, whereas 17-phenyl trinor-PGE2 (an EP1 agonist) and sulprostone (an EP3 agonist) had little effect. Both PGE2 and butaprost elevated cAMP intracellular levels in the cortical cells; moreover, forskolin, an activator of adenylate cyclase, enhanced NMDA-mediated cell death. These results suggest that PGE2, acting via EP2 receptors, aggravates excitotoxic neurodegeneration by a cAMP-dependent mechanism.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/drug effects , Dinoprostone/pharmacology , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Prostaglandin E/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cyclic AMP/metabolism , Female , Fetus/drug effects , Neurons/metabolism , Neurons/pathology , Pregnancy , Prostaglandins E, Synthetic/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Reverse Transcriptase Polymerase Chain Reaction
20.
J Neurosci ; 25(43): 9858-70, 2005 Oct 26.
Article in English | MEDLINE | ID: mdl-16251433

ABSTRACT

Increasing evidence suggests that cyclooxygenase-2 (COX-2) is involved in synaptic transmission and plasticity, and prostaglandin E2 (PGE2) is a key molecule in COX-2-meduated synaptic modification. However, the precise mechanisms, in particular, which subtypes of PGE2 receptors (EPs) mediate the PGE2-induced synaptic response, are not clear. Recently, we demonstrated that EPs are expressed heterogeneously in the hippocampus, and EP2/4 are mainly expressed in presynaptic terminals. Here, we report that PGE2 increased synaptic stimulus-evoked amplitudes of EPSPs in hippocampal slices and frequency of miniature EPSCs (mEPSCs) in hippocampal neurons in culture. These actions were mimicked by an EP2 agonist and attenuated by protein kinase A inhibitors. Decrease of EP2 expression through silencing the EP2 gene eliminated PGE2-induced increase of the frequency of mEPSCs. COX-2 and microsomal PGE synthase-1 (mPGES-1) and mPGES-2 are present in postsynaptic dendritic spines, because they are colocalized with PSD-95 (postsynaptic density-95), a postsynaptic marker. In addition, the frequency of mEPSCs was enhanced in neurons pretreated with interleukin-1beta or lipopolysaccharide, which elevated expression of COX-2 and mPGES-1 and produced PGE2, and this enhancement was inhibited by a COX-2 inhibitor that inhibited production of PGE2. Our results suggest that PGE2 synthesized by postsynaptically localized COX-2 functions as a retrograde messenger in hippocampal synaptic signaling via a presynaptic EP2 receptor.


Subject(s)
Hippocampus/cytology , Neurons/physiology , Presynaptic Terminals/metabolism , Prostaglandins E, Synthetic/pharmacology , Receptors, Prostaglandin E/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cyclooxygenase 2/metabolism , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Fluorescent Antibody Technique/methods , In Vitro Techniques , Interleukin-1/pharmacology , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Membrane Proteins , Mice , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques/methods , Placenta Growth Factor , Pregnancy Proteins/metabolism , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Receptors, Prostaglandin E, EP2 Subtype , Reverse Transcriptase Polymerase Chain Reaction/methods , Synaptic Transmission/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...