Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Med Rep ; 22(4): 2887-2895, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32700746

RESUMEN

Prostaglandin E2 (PGE2) is involved in numerous physiological and pathological processes of the kidney via its four receptors. A previous study has suggested that a defect in the PGE2 receptor 1 (EP1) gene markedly suppressed the transforming growth factor­ß1 (TGF­ß1)­induced mesangial cell (MC) proliferation and extracellular matrix aggregation. Therefore, the present study aimed to adopt a pharmacological method of specifically suppressing or activating the EP1 receptor to further verify and demonstrate these results. The EP1 receptor antagonist SC­19220 and EP1 receptor agonist 17­phenyl­trinor­PGE2 ethyl amide (17­pt­PGE2) were selectively used to treat five­sixths nephrectomy renal fibrosis model mice and TGF­ß1­stimulated MCs. An Alpha screen PGE2 assay kit, flow cytometry, western blotting and immunohistochemical techniques were adopted to perform in vivo and in vitro experiments. The present results suggested that compared with the control group, the selective EP1 receptor antagonist SC­19220 improved renal function, markedly reduced the plasma blood urea nitrogen and creatinine levels (P<0.05) and alleviated glomerulosclerosis (P<0.05). By contrast, the EP1 receptor agonist 17­pt­PGE2 aggravated renal dysfunction and glomerulosclerosis (P<0.05). To verify the renal protection mechanisms mediated by suppression of the EP1 receptor, the expression levels of endoplasmic reticulum stress (ERS)­related proteins, including chaperone glucose­regulated protein 78 (GRP78), transient receptor potential channel 1 (TRPC1) and protein kinase R­like endoplasmic reticulum kinase (PERK), were further evaluated histologically. The expression of GRP78, TRPC1 and PERK in the antagonist treatment group were markedly downregulated (P<0.05), whereas those in the agonist treatment group were upregulated (P<0.05). The present in vitro experiments demonstrated that, compared with the control group, the EP1 receptor antagonist suppressed the expression of GRP78, TRPC1 and PERK (P<0.05), reduced the production of PGE2 (P<0.05) and decreased the MC apoptosis rate (P<0.05), thus alleviating TGF­ß1­stimulated MC injury. Consequently, consistent with previous results, selectively antagonizing the EP1 receptor improved renal function and mitigated glomerulosclerosis, and its potential mechanism might be associated with the suppression of ERS.


Asunto(s)
Dinoprostona/metabolismo , Glomerulonefritis/tratamiento farmacológico , Subtipo EP1 de Receptores de Prostaglandina E/agonistas , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/análogos & derivados , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glomerulonefritis/etiología , Glomerulonefritis/fisiopatología , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Ratones , Ratones Endogámicos C57BL , Nefrectomía/efectos adversos , Antagonistas de Prostaglandina/farmacología , Canales Catiónicos TRPC/efectos de los fármacos , Canales Catiónicos TRPC/metabolismo , Factor de Crecimiento Transformador beta1/toxicidad , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo
2.
Br J Pharmacol ; 173(9): 1541-55, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26856544

RESUMEN

BACKGROUND AND PURPOSE: Prostanoids derived from COX-2 and EP receptors are involved in vascular remodelling in different cardiovascular pathologies. This study evaluates the contribution of COX-2 and EP1 receptors to vascular remodelling and function in hypertension. EXPERIMENTAL APPROACH: Spontaneously hypertensive rats (SHR) and angiotensin II (AngII)-infused (1.44 mg · kg(-1) · day(-1), 2 weeks) mice were treated with the COX-2 inhibitor celecoxib (25 mg · kg(-1) · day(-1) i.p) or with the EP1 receptor antagonist SC19220 (10 mg · kg(-1) · day(-1) i.p.). COX-2(-/-) mice with or without AngII infusion were also used. KEY RESULTS: Celecoxib and SC19220 treatment did not modify the altered lumen diameter and wall : lumen ratio in mesenteric resistance arteries from SHR-infused and/or AngII-infused animals. However, both treatments and COX-2 deficiency decreased the augmented vascular stiffness in vessels from hypertensive animals. This was accompanied by diminished vascular collagen deposition, normalization of altered elastin structure and decreased connective tissue growth factor and plasminogen activator inhibitor-1 gene expression. COX-2 deficiency and SC19220 treatment diminished the increased vasoconstrictor responses and endothelial dysfunction induced by AngII infusion. Hypertensive animals showed increased mPGES-1 expression and PGE2 production in vascular tissue, normalized by celecoxib. Celecoxib treatment also decreased AngII-induced macrophage infiltration and TNF-α expression. Macrophage conditioned media (MCM) increased COX-2 and collagen type I expression in vascular smooth muscle cells; the latter was reduced by celecoxib treatment. CONCLUSIONS AND IMPLICATIONS: COX-2 and EP1 receptors participate in the increased extracellular matrix deposition and vascular stiffness, the impaired vascular function and inflammation in hypertension. Targeting PGE2 receptors might have benefits in hypertension-associated vascular damage.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/metabolismo , Hipertensión/tratamiento farmacológico , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Rigidez Vascular/efectos de los fármacos , Animales , Celecoxib/administración & dosificación , Celecoxib/química , Celecoxib/farmacología , Células Cultivadas , Ciclooxigenasa 2/deficiencia , Inhibidores de la Ciclooxigenasa 2/farmacología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/administración & dosificación , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/química , Relación Dosis-Respuesta a Droga , Humanos , Hipertensión/metabolismo , Masculino , Ratones , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Relación Estructura-Actividad
3.
Brain Res ; 1625: 29-38, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26320551

RESUMEN

The spinal trigeminal subnucleus caudalis (Vc) receives preferentially nociceptive afferent signals from the orofacial area. Nociceptive stimuli to the orofacial area induce cyclooxygenase both peripherally and centrally, which can synthesize a major prostanoid prostaglandin E2 (PGE2) that implicates in diverse physiological functions. To clarify the roles of centrally-synthesized PGE2 in nociception, effects of exogenous PGE2 on synaptic transmission in the Vc neurons were investigated in the rat brainstem slice. Spontaneously occurring excitatory and inhibitory postsynaptic currents (sEPSCs and sIPSCs) were recorded, respectively, under pharmacological blockade of inhibitory and excitatory transmission by whole-cell patch-clamp mode. Perfusion of PGE2 (1-5 µM) increased the frequency of sIPSCs in a concentration-dependent manner but had no significant effect on the amplitude. Similarly to the effects on sIPSCs, PGE2 increased the sEPSC frequency without any effect on the amplitude. These facilitatory effects of PGE2 on spontaneous synaptic transmissions were blocked by an EP1 antagonist SC19220 but not by an EP4 antagonist AH23848. Electrical stimulation of the trigeminal tract evoked short latency EPSCs (eEPSCs) in the Vc neurons. PGE2 (5 µM) was ineffective on the eEPSCs. The present study demonstrated that PGE2 facilitated spontaneous synaptic transmissions in the Vc neurons through activating the presynaptic EP1 receptors but had no effect on the trigeminal tract-mediated excitatory transmission. These results suggest that centrally-synthesized PGE2 modifies the synaptic transmission in the Vc region, thereby contributing to the processing of nociceptive signals originated from the orofacial area.


Asunto(s)
Dinoprostona/farmacología , Neuronas/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Núcleo Espinal del Trigémino/citología , Animales , Compuestos de Bifenilo/farmacología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Antagonistas del GABA/farmacología , Técnicas In Vitro , Masculino , Técnicas de Placa-Clamp , Picrotoxina/farmacología , Antagonistas de Prostaglandina/farmacología , Quinoxalinas/farmacología , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos
4.
PLoS One ; 7(9): e45273, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22984630

RESUMEN

Although prostanoids are known to be involved in regulation of the spontaneous beating rate of cultured neonatal rat cardiomyocytes, the various subtypes of prostanoid receptors have not been investigated in detail. In our experiments, prostaglandin (PG)F(2α) and prostanoid FP receptor agonists (fluprostenol, latanoprost and cloprostenol) produced a decrease in the beating rate. Two prostanoid IP receptor agonists (iloprost and beraprost) induced first a marked drop in the beating rate and then definitive abrogation of beating. In contrast, the prostanoid DP receptor agonists (PGD(2) and BW245C) and TP receptor agonists (U-46619) produced increases in the beating rate. Sulprostone (a prostanoid EP(1) and EP(3) receptor agonist) induced marked increases in the beating rate, which were suppressed by SC-19220 (a selective prostanoid EP(1) antagonist). Butaprost (a selective prostanoid EP(2) receptor agonist), misoprostol (a prostanoid EP(2) and EP(3) receptor agonist), 11-deoxy-PGE(1) (a prostanoid EP(2), EP(3) and EP(4) receptor agonist) did not alter the beating rate. Our results strongly suggest that prostanoid EP(1) receptors are involved in positive regulation of the beating rate. Prostanoid EP(1) receptor expression was confirmed by western blotting with a selective antibody. Hence, neonatal rat cardiomyocytes express both prostanoid IP and FP receptors (which negatively regulate the spontaneous beating rate) and prostanoid TP, DP(1) and EP(1) receptors (which positively regulate the spontaneous beating rate).


Asunto(s)
Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Células Cultivadas , Cloprostenol/farmacología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/análogos & derivados , Dinoprostona/farmacología , Relación Dosis-Respuesta a Droga , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Hidantoínas/farmacología , Iloprost/farmacología , Latanoprost , Miocitos Cardíacos/metabolismo , Prostaglandina D2/farmacología , Prostaglandinas F Sintéticas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E/agonistas , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E/fisiología , Receptores de Tromboxanos/agonistas , Receptores de Tromboxanos/fisiología
5.
Artículo en Inglés | MEDLINE | ID: mdl-22858445

RESUMEN

The chick chorioallantoic membrane (CAM) subserves gas exchange in the developing embryo and shell-less culture affords a unique opportunity for direct observations over time of individual blood vessels to pharmacologic interventions. We tested a number of lipids including prostaglandins PGE(1&2) for vascular effects and signaling in the CAM. Application of PGE(1&2) induced a decrease in the diameter of large blood vessels and a concentration-dependent, localized, reversible loss of blood flow through small vessels. The loss of flow was also mimicked by misoprostol, an agonist for 3 of 4 known PGE receptors, EP(2-4), and by U46619, a thromboxane mimetic. Selective receptor antagonists for EP(3) and thromboxane each partially blocked the response. This is a first report of the effects of prostaglandins on vasoreactivity in the CAM. Our model allows the unique ability to examine simultaneous responses of large and small vessels in real time and in vivo.


Asunto(s)
Alprostadil/farmacología , Membrana Corioalantoides/efectos de los fármacos , Dinoprostona/farmacología , Vasoconstrictores/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Alprostadil/antagonistas & inhibidores , Animales , Compuestos de Bifenilo/farmacología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/fisiología , Compuestos Bicíclicos Heterocíclicos con Puentes , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/metabolismo , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Estradiol/farmacología , Estrógenos/farmacología , Ácidos Grasos Insaturados , Hidrazinas/farmacología , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Misoprostol/farmacología , Antagonistas de Prostaglandina/farmacología , Ratas , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxanos/agonistas , Receptores de Tromboxanos/antagonistas & inhibidores , Receptores de Tromboxanos/metabolismo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/antagonistas & inhibidores , Xantonas/farmacología , Ácido alfa-Linolénico/farmacología
6.
J Neurochem ; 115(2): 363-72, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20666934

RESUMEN

Prostaglandin E2 (PGE2) is a well known pain and pro-inflammatory mediator abundantly produced in inflamed tissue. It causes pain by directly exciting nociceptive primary sensory neurons (nociceptors) and indirectly stimulating the release of pain-related peptide substance P (SP) and calcitonin gene-related peptide (CGRP). In an ex vivo culture of sensory ganglion explants, we tested the hypothesis that PGE2 could induce the synthesis of SP and CGRP in nociceptors. A stabilized PGE2 analog, 16,16-dimethyl PGE2, in a concentration- and time-dependent manner, significantly increased mRNA and peptide levels of SP and CGRP. The agonists of EP1 and EP4 receptors also significantly increased SP and CGRP levels. Moreover, 16,16-dimethyl PGE2-induced SP and CGRP were blocked by EP1 and EP4 antagonists as well as the inhibitors of both protein kinase A and protein kinase C. Nerve growth factor was partially involved in PGE2-induced SP and CGRP synthesis. Taken together, these results indicate that PGE2 contributes to the synthesis of SP and CGRP in nociceptors, an event mediated by EP1 and EP4 receptors, nerve growth factor and protein kinase A and protein kinase C signalling pathways. We thus conclude that facilitating the synthesis of pain-related peptides in nociceptors is a novel mechanism underlying the role of PGE2 in nociception and chronic pain states.


Asunto(s)
16,16-Dimetilprostaglandina E2/farmacología , Antiulcerosos/farmacología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Ganglios Espinales/efectos de los fármacos , Sustancia P/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Factor de Crecimiento Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Antagonistas de Prostaglandina/farmacología , Prostaglandinas Sintéticas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Sustancia P/genética , Factores de Tiempo
7.
Neurochem Res ; 34(12): 2170-80, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19513831

RESUMEN

The role of enzymes and receptors of the prostanoid pathway in the inhibitory effect of 8-isoprostaglandin E2 (8-isoPGE2) on endogenous amino acid neurotransmitter levels was examined, ex vivo. Freshly isolated bovine eyeballs were injected intravitreally with IsoPs, incubated in Krebs buffer for 30 min and retina prepared for HPLC-ECD detection of amino acids. 8-isoPGE2 attenuated retinal glutamate and its metabolite, glutamine and glycine in a concentration-dependent manner. The nonselective cyclooxygenase (COX)-inhibitor, flurbiprofen, COX-2 selective inhibitor, NS-398 and thromboxane (Tx) synthase inhibitor, furegrelate had no effect on both basal amino acid levels and the inhibitory effects of 8-isoPGE2 (1-100 µM) on the retinal amino acids. Whereas the TP-receptor antagonist SQ-29548(10 µM) exhibited no effect, SC-19220(EP1; 30 µM), AH-6809(EP(1-3); 30 µM) and AH-23848(EP4; 30 µM) reversed the inhibitory effects of 8-isoPGE2 (0.01-100 µM) on glutamate, glutamine and glycine levels. We conclude that prostanoid EP-receptors regulate the inhibitory effect of 8-isoPGE2 on basal levels of endogenous amino acids in bovine retina, ex vivo.


Asunto(s)
Dinoprostona/análogos & derivados , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Glicina/metabolismo , Isoprostanos/farmacología , Neurotransmisores/metabolismo , Prostaglandinas/metabolismo , Receptores de Prostaglandina/fisiología , Retina/metabolismo , Animales , Benzofuranos/farmacología , Compuestos de Bifenilo/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes , Bovinos , Inhibidores de la Ciclooxigenasa/farmacología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/farmacología , Ácidos Grasos Insaturados , Flurbiprofeno/farmacología , Hidrazinas/farmacología , Nitrobencenos/farmacología , Receptores de Prostaglandina/efectos de los fármacos , Retina/efectos de los fármacos , Sulfonamidas/farmacología , Xantonas/farmacología
8.
Am J Physiol Gastrointest Liver Physiol ; 296(6): G1180-90, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19359421

RESUMEN

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.


Asunto(s)
Peristaltismo/efectos de los fármacos , Peristaltismo/fisiología , Prostaglandinas/farmacología , Estómago/efectos de los fármacos , Estómago/fisiología , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/fisiología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/análogos & derivados , Dinoprostona/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiología , Prostaglandinas/agonistas , Prostaglandinas E Sintéticas/farmacología , Antro Pilórico/citología , Antro Pilórico/efectos de los fármacos , Antro Pilórico/fisiología , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP3 de Receptores de Prostaglandina E , Estómago/citología
9.
Stroke ; 40(1): 261-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18948615

RESUMEN

BACKGROUND AND PURPOSE: Prostaglandin E(2) (PGE(2)) modulates autonomic transmission in the peripheral circulation. We investigated the role of endogenous PGE(2) and its presynaptic EP(1) receptor subtype in modulating the autonomic neurotransmission in cerebral vasculature. METHODS: The standard in vitro tissue-bath technique was used for measuring changes in isolated porcine basilar arterial tone. Calcium imaging and nitric oxide estimation along with immunohistochemical analysis for cyclo-oxygenase-1, cyclo-oxygenase-2, EP(1) receptor, PGE synthase, and neuronal nitric oxide synthase were done in cultured sphenopalatine ganglia and basilar artery. RESULTS: Selective EP(1) receptor antagonists (SC-19220 and SC-51322) inhibited relaxation of endothelium-denuded basilar arterial rings elicited by transmural nerve stimulation (2 and 8 Hz) without affecting that induced by nicotine or sodium nitroprusside (a nitric oxide donor). The SC-19220 inhibition of transmural nerve stimulation-elicited relaxation was blocked by cyclo-oxygenase inhibitors (salicylic acid and naproxen) but was not affected by guanethidine (a sympathetic neuronal blocker) or atropine. Perivascular cyclo-oxygenase-1- and cyclo-oxygenase-2-immunoreactive fibers were observed in basilar arteries. PGE synthase and EP(1) receptor immunoreactivities were coincident with neuronal nitric oxide synthase immunoreactivities in perivascular nerves of the basilar arteries and the sphenopalatine ganglia. omega-conotoxin (an N-type calcium channel blocker) significantly blocked transmural nerve stimulation-induced relaxation, which was further attenuated by SC-19220. In cultured sphenopalatine ganglia neurons, exogenous PGE(2) significantly increased calcium influx and diaminofluorescein fluorescence indicative of nitric oxide synthesis. Both responses were blocked by SC-19220. CONCLUSIONS: These results suggest that neuronal PGE(2) facilitates nitric oxide release from the cerebral perivascular parasympathetic nitrergic nerve terminals by increasing neuronal calcium influx through activation of presynaptic EP(1) receptors. PGE(2) may play an important role in regulating the nitrergic neurovascular transmission in the cerebral circulation.


Asunto(s)
Vías Autónomas/metabolismo , Arterias Cerebrales/inervación , Dinoprostona/metabolismo , Neuronas Nitrérgicas/metabolismo , Receptores de Prostaglandina E/metabolismo , Vasodilatación/fisiología , Animales , Vías Autónomas/efectos de los fármacos , Vías Autónomas/ultraestructura , Arteria Basilar/inervación , Arteria Basilar/fisiología , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Células Cultivadas , Arterias Cerebrales/fisiología , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Ganglios Parasimpáticos/efectos de los fármacos , Ganglios Parasimpáticos/metabolismo , Ganglios Parasimpáticos/ultraestructura , Inmunohistoquímica , Oxidorreductasas Intramoleculares/metabolismo , Neuronas Nitrérgicas/efectos de los fármacos , Neuronas Nitrérgicas/ultraestructura , Óxido Nítrico Sintasa de Tipo I/metabolismo , Antagonistas de Prostaglandina/farmacología , Prostaglandina-E Sintasas , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E , Sus scrofa , Vasodilatación/efectos de los fármacos
10.
Eur J Med Res ; 13(4): 154-62, 2008 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-18504170

RESUMEN

AIM: We have investigated CsA induced liver hyperplasia to explore the potential effects on the immunogenicity of the regenerating liver within the clinical context of rejection after transplantation. MATERIALS AND METHODS: Flow cytometry analysis of hepatocytes, isolated 48 hours after 2/3 partial hepatectomy (PH2/3) or sham operation in rats, was performed to determine the effect of CsA on DNA synthesis and MHC molecule expression. The possible role of PGE2 was evaluated by the administration of SC-19220, an EP1-PGE2 receptor antagonist. RESULTS: CsA augmented liver regeneration and this was partially attenuated by SC-19220. The moderate expression of class I MHC expression, as well as the very low class II MHC expression detected in normal hepatocytes by flow cytometry was augmented after PH2/3 and reduced by CsA. The CsA-mediated decrease of hepatocyte immunogenicity was not SC-19220 dependent. CONCLUSIONS: It is proposed that the enhancing effect of CsA on hepatocyte proliferation is by means of an indirect mechanism that can be attributed to a) reduced immunogenicity of the regenerating liver as a result of inhibition of class I and II MHC hepatocyte expression and b) increased PGE2 synthesis in the liver mediated by its action on EP1 receptor.


Asunto(s)
Ciclosporina/farmacología , Dinoprostona/metabolismo , Inmunosupresores/farmacología , Regeneración Hepática/efectos de los fármacos , Regeneración Hepática/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Animales , División Celular/efectos de los fármacos , División Celular/fisiología , ADN/biosíntesis , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Citometría de Flujo , Hepatectomía , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Masculino , Antagonistas de Prostaglandina/farmacología , Ratas , Ratas Wistar , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E
11.
Cardiovasc Res ; 78(1): 130-8, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18093985

RESUMEN

AIMS: The present study examined the hypothesis that prostaglandin E2 (PGE2) through activation of prostaglandin E (EP) receptor contributes to endothelium-dependent contractions. METHODS AND RESULTS: Western blotting revealed that the protein expression of EP1 receptor was significantly down-regulated in the aorta of the spontaneously hypertensive rat (SHR), but there was no significant difference in the expression of EP2, EP4, and total EP3 receptors between preparations of Wistar Kyoto rats (WKY) and SHR. Isometric tension studies showed that low concentrations of PGE2 caused endothelium-dependent relaxations in WKY but not in aortas of the SHR. High concentrations of PGE2 evoked contractions predominately through the activation of thromboxane-prostanoid (TP) receptors in the WKY, but involves the dual activation EP and TP receptors in the SHR. SQ29,548, BAYu3405 and Terutroban (TP receptor antagonists), and AH6809 (non-selective EP receptor antagonist) abolished, while SC19220 (preferential EP1 receptor antagonist) did not inhibit endothelium-dependent contractions. Both SC19220 and AH6809 significantly inhibited contractions to U46619 (TP receptor agonist). CONCLUSION: The present study demonstrates that the contraction caused by PGE2 in the SHR aorta is dependent on the activation of EP1 and TP receptors, but that endothelium-dependent contractions do not require the former. Thus, PGE2 is unlikely to be an endothelium-derived contracting factor in this artery. The ability of AH6809 to inhibit endothelium-dependent contractions can be attributed to its partial antagonism at TP receptors. Nevertheless, the impairment of PGE2-mediated relaxation may contribute to endothelial dysfunction in the aorta of the SHR.


Asunto(s)
Aorta Torácica/metabolismo , Dinoprostona/metabolismo , Endotelio Vascular/metabolismo , Hipertensión/metabolismo , Receptores de Prostaglandina E/metabolismo , Receptores de Tromboxanos/metabolismo , Vasoconstricción , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiopatología , Western Blotting , Compuestos Bicíclicos Heterocíclicos con Puentes , Carbazoles/farmacología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/análogos & derivados , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Ácidos Grasos Insaturados , Hidrazinas/farmacología , Hipertensión/fisiopatología , Inmunohistoquímica , Naftalenos/farmacología , Fenilefrina/farmacología , Cloruro de Potasio/farmacología , Propionatos/farmacología , Antagonistas de Prostaglandina/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E , Receptores de Tromboxanos/antagonistas & inhibidores , Sulfonamidas/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación , Xantonas/farmacología
12.
J Endocrinol ; 194(3): 595-602, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17761898

RESUMEN

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.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Dinoprostona/farmacología , Células Lúteas/metabolismo , Progesterona/biosíntesis , Receptores de Prostaglandina E/metabolismo , Alprostadil/análogos & derivados , Alprostadil/farmacología , Células Cultivadas , Cortisona/metabolismo , AMP Cíclico/metabolismo , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hidrocortisona/metabolismo , Células Lúteas/efectos de los fármacos , Antagonistas de Prostaglandina/farmacología , Prostaglandinas E Sintéticas/farmacología , Subtipo EP1 de Receptores de Prostaglandina E , Subtipo EP2 de Receptores de Prostaglandina E , Xantonas/farmacología
13.
Am J Physiol Regul Integr Comp Physiol ; 293(1): R528-37, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17363677

RESUMEN

PGE(2) is a well-known inhibitor of the antidiuretic hormone-induced increase of osmotic water permeability (OWP) in different osmoregulatory epithelia; however, the mechanisms underlying this effect of PGE(2) are not completely understood. Here, we report that, in the frog Rana temporaria urinary bladder, EP(1)-receptor-mediated inhibition of arginine-vasotocin (AVT)-induced OWP by PGE(2) is attributed to increased generation of nitric oxide (NO) in epithelial cells. It was shown that the inhibitory effect of 17-phenyl-trinor-PGE(2) (17-ph-PGE(2)), an EP(1) agonist, on AVT-induced OWP was significantly reduced in the presence of 7-nitroindazole (7-NI), a neuronal NO synthase (nNOS) inhibitor. NO synthase (NOS) activity in both lysed and intact epithelial cells measured as a rate of conversion of l-[(3)H]arginine to l-[(3)H]citrulline was Ca(2+) dependent and inhibited by 7-NI. PGE(2) and 17-ph-PGE(2), but not M&B-28767 (EP(3) agonist) or butaprost (EP(2) agonist), stimulated NOS activity in epithelial cells. The above effect of PGE(2) was abolished in the presence of SC-19220, an EP(1) antagonist. 7-NI reduced the stimulatory effect of 17-ph-PGE(2) on NOS activity. 17-ph-PGE(2) increased intracellular Ca(2+) concentration and cGMP in epithelial cells. Western blot analysis revealed an nNOS expression in epithelial cells. These results show that the inhibitory effect of PGE(2) on AVT-induced OWP in the frog urinary bladder is based at least partly on EP(1)-receptor-mediated activation of the NO/cGMP pathway, suggesting a novel cross talk between AVT, PGE(2), and nNOS that may be important in the regulation of water transport.


Asunto(s)
AMP Cíclico/fisiología , Dinoprostona/farmacología , Óxido Nítrico/fisiología , Oxitócicos/farmacología , Receptores de Prostaglandina E/fisiología , Transducción de Señal/efectos de los fármacos , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vasotocina/antagonistas & inhibidores , Vasotocina/farmacología , Equilibrio Hidroelectrolítico/efectos de los fármacos , Animales , Western Blotting , Química Encefálica/efectos de los fármacos , Calcio/metabolismo , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/análogos & derivados , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Técnicas In Vitro , Indazoles/farmacología , Masculino , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/metabolismo , Permeabilidad/efectos de los fármacos , Rana temporaria , Subtipo EP1 de Receptores de Prostaglandina E
14.
Eur J Pharmacol ; 517(1-2): 17-27, 2005 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-15964567

RESUMEN

Cyclooxygenase-2 is harmful in models of cerebral ischemia yet plays a protective role in preconditioning-induced ischemic tolerance in the heart. This study examined the mechanisms underlying cyclooxygenase-2-mediated neurotoxicity and preconditioning-induced neuroprotection in an in vitro model of cerebral ischemia. Inhibition of cyclooxygenase-2 protects cortical neuronal cultures from death induced by oxygen-glucose deprivation and reduces oxygen-glucose deprivation-induced increases in intracellular Ca(2+) ([Ca(2+)](i)). In the present study, we determined if prostaglandin E(2) (PGE(2)) is responsible for this cyclooxygenase-2-mediated effect. Rat cortical cultures expressed mRNA for the prostanoid EP(1)-EP(4) receptors. PGE(2) reversed the attenuation in [Ca(2+)](i) and the protection offered by cyclooxygenase-2 inhibition during oxygen-glucose deprivation. These effects likely occur via activation of the prostanoid EP(1) receptor since blocking this receptor during oxygen-glucose deprivation reduced [Ca(2+)](i) and neurotoxicity. Next, we considered if the moderate activation of this pathway, by preconditioning cultures with sub-lethal oxygen-glucose deprivation, influenced the development of tolerance to an otherwise lethal oxygen-glucose deprivation insult, 48 h later. Inhibition of cyclooxygenase-2 during oxygen-glucose deprivation-preconditioning abolished preconditioning-induced protection. Furthermore, cultures were rendered tolerant to oxygen-glucose deprivation by the transient exposure to exogenous PGE(2) 24 h prior to the insult, indicating that this product of the cyclooxygenase-2 pathway is sufficient to induce ischemic tolerance. This study shows that cyclooxygenase-2 and PGE(2) are involved in both oxygen-glucose deprivation-induced neurotoxicity and preconditioning-induced neuroprotection. While neurotoxic in the context of lethal oxygen-glucose deprivation, the moderate activation of this signalling pathway confers ischemic tolerance.


Asunto(s)
Dinoprostona/fisiología , Neuronas/metabolismo , Animales , Encéfalo , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/fisiología , Células Cultivadas , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Feto , Expresión Génica/efectos de los fármacos , Glucosa/deficiencia , Masculino , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Embarazo , Antagonistas de Prostaglandina/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Pirazoles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/genética , Subtipo EP1 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E
15.
J Bone Miner Res ; 20(1): 15-22, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15619665

RESUMEN

UNLABELLED: We examined the direct effect of SC-19220, an EP1 prostaglandin (PG) E2 receptor antagonist, on osteoclastogenesis induced by RANK/RANKL signaling in mouse cell cultures. We found that SC-19220 inhibited RANKL-induced osteoclastogenesis by suppression of the RANK/RANKL signaling pathway in osteoclast precursors. INTRODUCTION: Bone growth is accomplished by a dynamic equilibrium between formation by osteoblasts and resorption by osteoclasts, which are regulated by many systemic and local osteotropic factors that induce osteoclast formation from hematopoietic precursors through RANK/RANKL signaling. There are four subtypes of prostaglandin E (PGE) receptors, EP1, EP2, EP3, and EP4, and PGE2 facilitates bone resorption by a mechanism mediated by EP2/EP4. It is well known that SC-19220 is an EP1-specific antagonist. We previously found that SC-19220 inhibited osteoclastogenesis induced by osteotropic factors, including PGE2; however, the inhibitory mechanism is not clear. In this study, we investigated the inhibitory effects of SC-19220 on osteoclastogenesis induced by RANK/RANKL signaling in mouse cell cultures and analyzed the mechanism involved. MATERIALS AND METHODS: A bone marrow culture system and bone marrow macrophages were used to examine the effects of SC-19220 on PGE2-, 11-deoxy-PGE1-, and RANKL-induced osteoclastogenesis. We analyzed RANKL expression in osteoblasts induced by PGE2 using RT-PCR. We also examined the effects of SC-19220 on the macrophage-colony-stimulating factor (M-CSF) receptor (c-Fms) and RANK expression in osteoclast precursors as well as RANK/RANKL signaling using RT-PCR and Western blotting analyses. RESULTS AND CONCLUSION: SC-19220 dose-dependently inhibited osteoclast formation induced by PGE2, 11-deoxy-PGE1, and RANKL in the mouse culture system; however, it had no influence on RANKL expression in osteoblasts induced by PGE2. Furthermore, the expression of RANK and c-Fms in osteoclast precursors was decreased by SC-19220 at the mRNA and protein levels. In RANK signaling networks, SC-19220 inhibited c-Src and NFAT2 expression. Our findings indicated that SC-19220 inhibits RANKL-induced osteoclastogenesis through the suppression of RANK, c-Fms, c-Src, and NFAT2, suggesting that this EP1-specific antagonist inhibits osteoclast formation induced by RANKL from the early stage of osteoclastogenesis.


Asunto(s)
Alprostadil/análogos & derivados , Proteínas Portadoras/antagonistas & inhibidores , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Glicoproteínas de Membrana/antagonistas & inhibidores , Osteoclastos/citología , Antagonistas de Prostaglandina/farmacología , Receptores de Prostaglandina E/antagonistas & inhibidores , Alprostadil/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Proteína Tirosina Quinasa CSK , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Proteínas de Unión al ADN/metabolismo , Dinoprostona/agonistas , Dinoprostona/antagonistas & inhibidores , Dinoprostona/farmacología , Regulación hacia Abajo , Femenino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos , Factores de Transcripción NFATC , Proteínas Nucleares/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores de Prostaglandina E/agonistas , Subtipo EP1 de Receptores de Prostaglandina E , Factores de Transcripción/metabolismo , Familia-src Quinasas
16.
Osteoarthritis Cartilage ; 12(11): 895-903, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15501405

RESUMEN

OBJECTIVE: Calpains are known as Ca(2+)-dependent intracellular neutral cysteine proteases. However, m-calpain is detected in synovial fluid of arthritic joints and is shown to possess the proteoglycanase activity in vitro. The mechanism of m-calpain release into the extracellular spaces during arthritis has not yet been well characterized. In the present study, we have analyzed m-calpain release from cultured chondrocytes stimulated by a proinflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha). The effects of non-steroidal anti-inflammatory drugs (NSAIDs) on m-calpain release were also examined. METHODS: Human chondrocytic HCS-2/8 cells were stimulated by TNF-alpha in the presence or absence of an NSAID. m-Calpain in the cells and culture medium was quantified by Western blot analysis using an anti-m-calpain antibody. Western blots were subjected to densitometric analysis and band intensities were determined. RESULTS: TNF-alpha (10 ng/ml) stimulated m-calpain release with transient increase in cellular m-calpain in HCS-2/8 cells. NSAIDs examined (aspirin, loxoprofen-SRS, diclofenac sodium, indomethacin and NS398) inhibited m-calpain release and production of prostaglandin E(2) (PGE(2)) induced by 10 ng/ml TNF-alpha. Exogenously added PGE(2) accelerated the release of m-calpain in response to a lower concentration of TNF-alpha (1 ng/ml). AH6809, an EP1/2 antagonist, but not SC19220 (an EP1 antagonist), effectively inhibited TNF-alpha-induced m-calpain release. In contrast, butaprost, an EP2 agonist, accelerated release of m-calpain by 1 ng/ml TNF-alpha. CONCLUSIONS: These results suggest that TNF-alpha stimulates upregulation and release of m-calpain in chondrocytic HCS-2/8 cells, and that stimulation of EP2-PGE(2) receptor by produced PGE(2) is deeply involved in this process.


Asunto(s)
Alprostadil/análogos & derivados , Antiinflamatorios no Esteroideos/farmacología , Calpaína/metabolismo , Condrocitos/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Alprostadil/farmacología , Apoptosis/fisiología , Aspirina/farmacología , Calpaína/antagonistas & inhibidores , Línea Celular Tumoral , Condrocitos/efectos de los fármacos , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Diclofenaco/farmacología , Dinoprostona/biosíntesis , Dinoprostona/farmacología , Humanos , Nitrobencenos/farmacología , Fenilpropionatos/farmacología , Antagonistas de Prostaglandina/farmacología , Sulfonamidas/farmacología , Regulación hacia Arriba , Xantonas/farmacología
17.
Br J Pharmacol ; 142(4): 788-96, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15172959

RESUMEN

1. In human pulmonary vascular preparations, precontracted arteries were more sensitive to the relaxant effect of acetylcholine (ACh) than veins (pD(2) values: 7.25+/-0.08 (n=23) and 5.92+/-0.09 (n=25), respectively). Therefore, the role of prostacyclin (PGI(2)) was explored to examine whether this mediator may be responsible for the difference in relaxation. 2. In the presence of the cyclooxygenase (COX) inhibitor, indomethacin (INDO), the ACh relaxations were reduced in arteries but not in veins. On the contrary, an inhibitor (l-NOARG) of the nitric oxide synthase blocked preferentially the relaxation in veins. 3. A greater release of 6-keto-PGF(1alpha), the stable metabolite of PGI(2), was observed in arterial preparations than in venous preparations when stimulated with either ACh or arachidonic acid (AA). 4. Exogenous PGI(2) produced a reduced relaxant effect in the precontracted vein when compared with the artery. In the presence of the EP(1)-receptor antagonist AH6809, the PGI(2) relaxation of veins was similar to arteries. 5. In veins, AA (0.1 mm) produced a biphasic response, namely, a contraction peak (0.4-0.5 g) followed by a relaxation. These contractions in venous preparations were abolished either in the absence of endothelium or in the presence of INDO or an EP(1)-receptor antagonist (AH6809, SC19220). In the arterial preparations AA induced only relaxations. 6. In both vascular preparations, COX-1 but not the COX-2 protein was detected in microsomal preparations derived from homogenized tissues or freshly isolated endothelial cells. 7. The differential vasorelaxations induced by ACh may be explained, in part, by a more pronounced production and release of PGI(2) in human pulmonary arteries than in the veins. In addition, while PGI(2) induced relaxation by activation of IP-receptors in both types of vessels, a PGI(2) constrictor effect was responsible for masking the relaxation in the veins by activation of the EP(1)-receptor.


Asunto(s)
Epoprostenol/fisiología , Arteria Pulmonar/fisiología , Venas Pulmonares/fisiología , Receptores de Prostaglandina/fisiología , 6-Cetoprostaglandina F1 alfa/química , 6-Cetoprostaglandina F1 alfa/metabolismo , Acetilcolina/antagonistas & inhibidores , Acetilcolina/farmacología , Animales , Ácido Araquidónico/farmacología , Western Blotting/métodos , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Francia , Humanos , Indometacina/farmacología , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiología , Nitroarginina/farmacología , Arteria Pulmonar/química , Arteria Pulmonar/efectos de los fármacos , Venas Pulmonares/química , Venas Pulmonares/efectos de los fármacos , Receptores de Prostaglandina/efectos de los fármacos , Receptores de Prostaglandina E/antagonistas & inhibidores , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Xantonas/farmacología
18.
J Physiol ; 553(Pt 2): 407-14, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500777

RESUMEN

The synaptic release of glutamate evokes in astrocytes periodic increases in [Ca2+]i, due to the activation of metabotropic glutamate receptors (mGluRs). The frequency of these [Ca2+]i oscillations is controlled by the level of neuronal activity, indicating that they represent a specific, frequency-coded signalling system of neuron-to-astrocyte communication. We recently found that neuronal activity-dependent [Ca2+]i oscillations in astrocytes are the main signal that regulates the coupling between neuronal activity and blood flow, the so-called functional hyperaemia. Prostaglandins play a major role in this fundamental phenomenon in brain function, but little is known about a possible link between [Ca2+]i oscillations and prostaglandin release from astrocytes. To investigate whether [Ca2+]i oscillations regulate the release of vasoactive prostaglandins, such as the potent vasodilator prostaglandin E2 (PGE2), from astrocytes, we plated wild-type human embryonic kidney (HEK)293 cells, which respond constitutively to PGE2 with [Ca2+]i elevations, onto cultured astrocytes, and used them as biosensors of prostaglandin release. After loading the astrocyte-HEK cell co-cultures with the calcium indicator Indo-1, confocal microscopy revealed that mGluR-mediated [Ca2+]i oscillations triggered spatially and temporally coordinated [Ca2+]i increases in the sensor cells. This response was absent in a clone of HEK cells that are unresponsive to PGE2, and recovered after transfection with the InsP3-linked prostanoid receptor EP1. We conclude that [Ca2+]i oscillations in astrocytes regulate prostaglandin releases that retain the oscillatory behaviour of the [Ca2+]i changes. This finely tuned release of PGE2 from astrocytes provides a coherent mechanistic background for the role of these glial cells in functional hyperaemia.


Asunto(s)
Astrocitos/fisiología , Señalización del Calcio/fisiología , Ácido Glutámico/fisiología , Prostaglandinas/fisiología , Valina/análogos & derivados , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Calcio/metabolismo , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Cicloleucina/análogos & derivados , Cicloleucina/farmacología , Citosol/metabolismo , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Proteínas Fluorescentes Verdes , Humanos , Indometacina/farmacología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Técnicas de Placa-Clamp , Prostaglandinas/metabolismo , Ácido Quiscuálico/farmacología , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/fisiología , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/fisiología , Subtipo EP1 de Receptores de Prostaglandina E , Transfección , Valina/farmacología , Xantonas/farmacología
19.
Exp Cell Res ; 289(2): 265-74, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14499627

RESUMEN

Aberrant upregulation of COX-2 enzyme resulting in accumulation of PGE2 in a cancer cell environment is a marker for progression of many cancers, including breast cancer. Four subtypes of cell surface receptors (EP1, EP2, EP3, and EP4), which are coupled with different G-proteins, mediate PGE2 actions. Since migration is an essential step in invasion and metastasis, in the present study we defined the expression of EP receptors and their roles in migratory function of breast cancer cells of murine (C3L5) and human (MDA-MB-231 and MCF-7) origin. Highly metastatic C3L5 and MDA-MB-231 cells, found to be highly migratory in a Transwell migration assay, were shown to accumulate much higher levels of PGE2 in culture media in comparison with nonmetastatic and poorly migrating MCF-7 cells; the levels of PGF2alpha and 6-keto-PGF1alpha were low in all cases. The elevated PGE2 production by metastatic cancer cells was due to COX-2 activity since dual COX-1/2 inhibitor indomethacin and selective COX-2 inhibitor NS-398 equally suppressed both basal and inducible (by IFN-gamma/LPS or Ca2+-ionophores) PGE2 accumulation. RT-PCR analysis revealed that murine C3L5 cells expressed mRNA of EP1, EP3, and EP4 but not EP2 receptors. On the other hand, human MDA-MB-231 and MCF-7 cells expressed all the above receptors. High levels of expression of functional EP4 receptors coupled with Gs-protein was confirmed in C3L5 cells by biochemical assay showing a dose-dependent increase of intracellular cAMP synthesis in response to PGE2. EP receptor antagonists SC-19220, AH-6809, and AH-23848B, having highest affinity for EP1, EP1/EP2/DP, and EP4 receptors, respectively, variably inhibited migration of metastatic breast cancer cells. An autocrine PGE2-mediated migratory activity of these cells appeared to be associated predominantly with EP4 receptor-mediated signaling pathway, which uses cAMP as a second messenger. This conclusion is based on several observations: (1) selective EP4 antagonist AH-23848B effectively inhibited migration of both C3L5 and MDA-MB-231 cells in a dose-dependent manner; (2) exogenous PGE2 and EP4 agonist PGE1 alcohol increased migration of C3L5 cells; (3) forskolin, a potent activator of adenylate cyclase, as well as membrane-permeable analogues of cAMP (8-bromo-cAMP, dibutyryl-cAMP) stimulated migration of C3L5 cells; and (4) Rp-cAMPS, a selective protein kinase A inhibitor, reduced migration of C3L5 cells. Migration of poorly migratory MCF-7 cells remained unaffected with either PGE2 or EP4 antagonist. These findings are relevant for designing therapeutic strategies against breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Movimiento Celular/fisiología , Dinoprostona/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Receptores de Prostaglandina E/metabolismo , Xantonas , Alprostadil/metabolismo , Alprostadil/farmacología , Animales , Neoplasias de la Mama/genética , Carcinoma/genética , Movimiento Celular/efectos de los fármacos , Colforsina/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Indoles/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Neoplasias Mamarias Experimentales/genética , Proteínas de la Membrana , Ratones , Invasividad Neoplásica , Prostaglandina-Endoperóxido Sintasas/metabolismo , Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Xantenos/farmacología
20.
J Cell Sci ; 116(Pt 16): 3285-94, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12829746

RESUMEN

Arginine-vasopressin (AVP) facilitates water reabsorption in renal collecting duct principal cells by activation of vasopressin V2 receptors and the subsequent translocation of water channels (aquaporin-2, AQP2) from intracellular vesicles into the plasma membrane. Prostaglandin E2 (PGE2) antagonizes AVP-induced water reabsorption; the signaling pathway underlying the diuretic response is not known. Using primary rat inner medullary collecting duct (IMCD) cells, we show that stimulation of prostaglandin EP3 receptors induced Rho activation and actin polymerization in resting IMCD cells, but did not modify the intracellular localization of AQP2. However, AVP-, dibutyryl cAMP- and forskolin-induced AQP2 translocation was strongly inhibited. This inhibitory effect was independent of increases in cAMP and cytosolic Ca2+. In addition, stimulation of EP3 receptors inhibited the AVP-induced Rho inactivation and the AVP-induced F-actin depolymerization. The data suggest that the signaling pathway underlying the diuretic effects of PGE2 and probably those of other diuretic agents include cAMP- and Ca2+-independent Rho activation and F-actin formation.


Asunto(s)
AMP Cíclico/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Dinoprostona/farmacología , Médula Renal/metabolismo , Receptores de Prostaglandina E/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Actinas/metabolismo , Animales , Acuaporina 2 , Acuaporinas/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Membrana Celular/metabolismo , Células Cultivadas , Colforsina/farmacología , Vesículas Citoplasmáticas/metabolismo , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Microscopía Fluorescente , Modelos Moleculares , Neurofisinas/metabolismo , Antagonistas de Prostaglandina/farmacología , Precursores de Proteínas/metabolismo , Ratas , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Vasopresinas/metabolismo , Transducción de Señal , Vasopresinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA