Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
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.
Ecotoxicol Environ Saf ; 201: 110712, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32502905

RESUMEN

Exposure to manganese (Mn) can cause male reproductive damage and lead to abnormal secretion of sex hormones. Gonadotropin-releasing hormone (GnRH) plays an important role in the neuromodulation of vertebrate reproduction. Astrocytes can indirectly regulate the secretion of GnRH by binding paracrine prostaglandin E2 (PGE2) specifically to the EP1 and EP2 receptors on GnRH neurons. Prior studies assessed the abnormal secretion of GnRH caused by Mn exposure, but the specific mechanism has not been reported in detail. This study investigated the effects of Mn exposure on the reproductive system of male mice to clarify the role of PGE2 in the abnormal secretion of GnRH in the hypothalamus caused by exposure to Mn. Our data demonstrate that antagonizing the EP1 and EP2 receptors of PGE2 can restore abnormal levels of GnRH caused by Mn exposure. Mn exposure causes reduced sperm count and sperm shape deformities. These findings suggest that EP1 and EP2, the receptors of PGE2, may be the key to abnormal GnRH secretion caused by Mn exposure. Antagonizing the PGE2 receptors may reduce reproductive damage caused by Mn exposure.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Manganeso/toxicidad , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Reproducción/efectos de los fármacos , Animales , Hipotálamo/metabolismo , Masculino , Manganeso/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores
3.
J Alzheimers Dis ; 74(1): 173-187, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31985468

RESUMEN

BACKGROUND: Neuroinflammation has been recognized as an important factor in the pathogenesis of Alzheimer's disease (AD). One of the most recognized pathways in mediating neuroinflammation is the prostaglandin E2-EP1 receptor pathway. OBJECTIVE: Here, we examined the efficacy of the selective EP1 antagonist ONO-8713 in limiting amyloid-ß (Aß), lesion volumes, and behavioral indexes in AD mouse models after ischemic stroke. METHODS: Transgenic APP/PS1, 3xTgAD, and wildtype (WT) mice were subjected to permanent distal middle cerebral artery occlusion (pdMCAO) and sham surgeries. Functional outcomes, memory, anatomical outcomes, and Aß concentrations were assessed 14 days after surgery. RESULTS: pdMCAO resulted in significant deterioration in functional and anatomical outcomes in the transgenic mice compared with the WT mice. No relevant differences were observed in the behavioral tests when comparing the ONO-8713 and vehicle-treated groups. Significantly lower cavitation (p = 0.0373) and percent tissue loss (p = 0.0247) were observed in APP/PS1 + ONO-8713 mice compared with the WT + ONO-8713 mice. However, the percent tissue injury was significantly higher in APP/PS1 + ONO-8713 mice compared with the WT + ONO-8713 group (p = 0.0373). Percent tissue loss was also significantly lower in the 3xTgAD + ONO-8713 mice than in the WT + ONO-8713 mice (p = 0.0185). ONO-8713 treatment also attenuated cortical microgliosis in APP/PS1 mice as compared with the vehicle (p = 0.0079); however, no differences were observed in astrogliosis across the groups. Finally, APP/PS1 mice presented with characteristic Aß load in the cortex while 3xTgAD mice exhibited very low Aß levels. CONCLUSION: In conclusion, under the experimental conditions, EP1 receptor antagonist ONO-8713 showed modest benefits in anatomical outcomes after stroke, mainly in APP/PS1 mice.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Dinoprostona , Accidente Cerebrovascular Isquémico/complicaciones , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Enfermedad de Alzheimer/patología , Animales , Astrocitos/patología , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Proteínas Sanguíneas/genética , Cinamatos/farmacología , Encefalitis/complicaciones , Encefalitis/patología , Gliosis/tratamiento farmacológico , Gliosis/patología , Humanos , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Accidente Cerebrovascular Isquémico/patología , Masculino , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Proteínas de Unión a Poli(A)/genética , Presenilina-1/genética
4.
Digestion ; 101(3): 270-278, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30897584

RESUMEN

BACKGROUND/AIMS: Transient lower esophageal sphincter relaxations (TLESRs) are the major cause of gastroesophageal reflux. Recently, an EP1 receptor antagonist, ONO-8539, showed the reduction of TLESRs in monkeys. However, its effect on TLESRs in humans remains unclear. This study investigated the effect of ONO-8539 on postprandial TLESRs in healthy male subjects. METHODS: Twenty-seven subjects participated in this placebo-controlled, cross-over study. The subjects received either placebo or ONO-8539 (450 mg) after a standardized breakfast. A 30-min basal recording was performed 4 h after drug administration. Subsequently, TLESR recordings were performed after a high-fat test meal for 3 h. The examination was repeated at least 7 days from the first evaluation for washout. RESULTS: Thirteen patients were ultimately analyzed. The basal lower esophageal sphincter pressure was not different between the 2 groups (16.3 and 18.0 mm Hg for placebo and ONO-8539, respectively; p = 0.88). ONO-8539 significantly reduced the number of TLESRs from 15.0 to 12.0 for 3 h (p < 0.05). The proportion of terminating events of TLESRs was significantly different between the 2 groups (p < 0.05). No events and swallowing terminated more TLESRs with ONO-8539 than with placebo. CONCLUSIONS: ONO-8539 suppressed TLESRs mildly. EP1 receptor may be involved with the mechanism of human TLESRs.


Asunto(s)
Benzoatos/administración & dosificación , Esfínter Esofágico Inferior/efectos de los fármacos , Reflujo Gastroesofágico/prevención & control , Indenos/administración & dosificación , Relajación Muscular/efectos de los fármacos , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Tiazoles/administración & dosificación , Adulto , Estudios Cruzados , Método Doble Ciego , Endoscopía del Sistema Digestivo/métodos , Esfínter Esofágico Inferior/diagnóstico por imagen , Esfínter Esofágico Inferior/fisiopatología , Femenino , Reflujo Gastroesofágico/fisiopatología , Humanos , Masculino , Manometría/métodos , Relajación Muscular/fisiología , Periodo Posprandial , Resultado del Tratamiento , Adulto Joven
5.
Ann Biomed Eng ; 48(3): 927-939, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30980293

RESUMEN

Fracture healing is a complex and integrated process that involves mesenchymal progenitor cell (MPC) recruitment, proliferation and differentiation that eventually results in bone regeneration. Prostaglandin E2 (PGE2) is an important regulator of bone metabolism and has an anabolic effect on fracture healing. Prior work from our laboratory showed EP1-/- mice have enhanced fracture healing, stronger cortical bones, higher trabecular bone volume and increased in vivo bone formation. We also showed that bone marrow MSCs from EP1-/- mice exhibit increased osteoblastic differentiation in vitro. In this study we investigate the changes in the periosteal derived MPCs (PDMPCs), which are crucial for fracture repair, upon EP1 deletion. EP1-/- PDMPCs exhibit increased numbers of total (CFU-F) and osteoblastic colonies (CFU-O) as well as enhanced osteoblastic and chondrogenic differentiation. Moreover, we tested the possible therapeutic application of a specific EP1 receptor antagonist to accelerate fracture repair. Our findings showed that EP1 antagonist administration to wild type mice in the early stages of repair similarly resulted in enhanced CFU-F, CFU-O, and osteoblast differentiation in PDMPCs and resulted in enhanced fracture callus formation at 10 days post fracture and increased bone volume and improved biomechanical healing of femur fractures at 21 days post fracture.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Periostio/citología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Diferenciación Celular , Condrogénesis , Femenino , Curación de Fractura , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/fisiología , Osteogénesis , Subtipo EP1 de Receptores de Prostaglandina E/genética , Subtipo EP1 de Receptores de Prostaglandina E/fisiología
6.
Pharmacology ; 104(5-6): 267-275, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31434088

RESUMEN

Epigallocatechin-3-gallate (EGCG), the principal catechin of green tea, modulates different molecular mechanisms underlying hepatocellular carcinoma (HCC). Accumulating studies showed that the activation of prostaglandin (PG) receptor EP1 promotes cell migration and invasion in different cancers, which could be inverted by blocking the EP1 receptor. This study investigated the synergetic effects of EP1-selective antagonist ONO-8711 and EGCG treatment on HCC to better understand the potential strategy to treat HCC. We found that EGCG significantly inhibited PGE2 and EP1-selective agonist induced migration of HCC cells and increased the ratio of Bax/Bcl-2 even in the presence of ONO-DI-004 or PGE2. ONO-8711 significantly inhibited PGE2-induced HCC proliferation while increased the inhibitory effect of EGCG on HCC cell viability and migration ability compared with EGCG alone. These findings suggest that a combination of ONO-8711 and EGCG is a potential treatment for HCC therapy.


Asunto(s)
Alprostadil/análogos & derivados , Carcinoma Hepatocelular/tratamiento farmacológico , Catequina/análogos & derivados , Neoplasias Hepáticas/tratamiento farmacológico , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Alprostadil/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Catequina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dinoprostona/metabolismo , Sinergismo Farmacológico , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Cicatrización de Heridas/efectos de los fármacos
7.
Bosn J Basic Med Sci ; 19(3): 265-273, 2019 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-30995899

RESUMEN

Recent studies showed that the activation of prostaglandin (PG) receptor EP1 promotes cell migration and invasion in different cancers. The aim of this study was to investigate the role of EP1 in the proliferation of osteosarcoma (OS) cells in vitro and in vivo. EP1 mRNA and protein levels were analyzed by real-time RT-PCR and Western blot, respectively in human OS cell lines MG63, OS732, U-2OS, and 143B compared to human fetal osteoblastic hFOB 1.19 cells. MG63 cells were treated with PGE2, EP1 specific agonist 17-PT-PGE2, 17-PT-PGE2 + EP1 specific antagonist SC51089, or DMSO (control). EP1R-siRNA or a non-silencing irrelevant RNA duplex (negative control) were used for the transfection of MG63 cells, followed by PGE2 treatment. Nude mice carrying MG63 xenografts were treated with SC51089 (2 mg/kg/day). MG63 cells/xenografts were analyzed by MTT assay, TUNEL assay, PKC enzyme activity assay, and Western blot (EP1 and apoptotic proteins), and tumor growth/volume was evaluated in mice. EP1 levels were significantly higher in OS cells compared to osteoblasts. PGE2 or 17-PT-PGE2 treatment increased the proliferation and decreased the apoptosis of MG63 cells. Inhibition of EP1 by SC51089 or siRNA markedly decreased the viability of MG63 cells. Similarly, SC51089 treatment significantly inhibited MG63 cell proliferation and promoted apoptosis in vivo. The silencing of EP1 receptor by siRNA or blockade of EP1 signaling by SC51089 activated extrinsic and intrinsic apoptotic pathways both in vivo and in vitro, as evidenced by increased levels of Bax, cyt c, cleaved caspase-3, caspase-8 and caspase-9. EP1 appears to be involved in PGE2-induced proliferative activity of MG63 cells. Antagonizing EP1 may provide a novel therapeutic approach to the treatment of OS.


Asunto(s)
Neoplasias Óseas/patología , Dinoprostona/farmacología , Osteosarcoma/patología , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Silenciador del Gen , Humanos , Hidrazinas/farmacología , Ratones , Ratones Desnudos , Oxazepinas/farmacología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Pharm Pharm Sci ; 21(1): 326-339, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30075829

RESUMEN

Prostaglandin E2 (PGE2), one of the arachidonic acid metabolites synthetized from arachidonic acid through cyclooxygenase (COX) catalysis, demonstrates multiple physiological and pathological actions through different subtypes of EP receptors. PURPOSE: The present study was designed to explore the effects of PGE2 on cardiac fibrosis and the involved mechanism. METHODS: We used western blot analysis, real-time quantitative PCR and immunostaining etc. to testify the mechanism. RESULTS: Our data showed that in cultured adult rat cardiac fibroblasts (CFs), PGE2 effectively promoted the expression of α-smooth muscle actin (α-SMA), connective tissue growth factor (CTGF),fibronectin (FN), Collagen I and induced [Ca2+]i increase. Besides, calcium increase evoked by PGE2 is mediated by virtue of EP1 activation. Instead of EP3 or EP4, inhibition of EP1 attenuated PGE2-stimulated upregulation of α-SMA,CTGF, FN, collagen I and [Ca2+]i, as well as the nuclear factor of activated T cell cytoplasmic 4 protein (NFATc4) translocation. CONCLUSIONS: PGE2 may promote cardiac fibrosis via EP1 receptor and calcium signal pathway.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Dinoprostona/farmacología , Fibroblastos/efectos de los fármacos , Fibrosis/inducido químicamente , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Fibrosis/metabolismo , Inyecciones Intraperitoneales , Masculino , Interferencia de ARN/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo
9.
Bioorg Med Chem Lett ; 28(14): 2408-2412, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29934246

RESUMEN

We describe a medicinal chemistry approach to the discovery of a novel EP1 antagonist exhibiting high potency and good pharmacokinetics. Our starting point is 1, an EP1 receptor antagonist that exhibits pharmacological efficacy in cystometry models following intravenous administration. Despite its good potency in vitro, the high lipophilicity of 1 is a concern in long-term in vivo studies. Further medicinal chemistry efforts identified 4 as an improved lead compound with good in vitro ADME profile applicable to long term in vivo studies. A rat fracture study was conducted with 4 for 4 weeks to validate its utility in bone fracture healing. The results suggest that this EP1 receptor antagonist stimulates callus formation and thus 4 has potential for enhancing fracture healing.


Asunto(s)
Descubrimiento de Drogas , Curación de Fractura/efectos de los fármacos , Fracturas Óseas/tratamiento farmacológico , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Tiazoles/farmacología , Animales , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Fracturas Óseas/metabolismo , Células de Riñón Canino Madin Darby/efectos de los fármacos , Células de Riñón Canino Madin Darby/metabolismo , Células de Riñón Canino Madin Darby/patología , Ratones , Ratones Noqueados , Estructura Molecular , Subtipo EP1 de Receptores de Prostaglandina E/deficiencia , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Relación Estructura-Actividad , Tiazoles/química
10.
Neurourol Urodyn ; 37(1): 132-137, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28608967

RESUMEN

AIMS: We examined the effect of an E-series prostaglandin 1 (EP1) receptor antagonist on bladder activity using a rat model of spinal cord injury (SCI). METHODS: Female Sprague-Dawley rats were used. Six weeks after spinal cord transection, conscious-filling cystometry was performed. We evaluated the urodynamic parameters before and after intravenous (0.1 and 1.0 mg/kg) or intrathecal (0.1 and 1.0 µg) administration of SC51089, an EP1 antagonist. Spinal prostaglandin E2 (PGE2) concentration and EP1 receptor transcripts in the spinal cord and dorsal root ganglia (DRG) were measured by enzyme-linked immunosorbent assay and RT-PCR, respectively. RESULTS: The time to the first non-voiding contraction (NVC) was significantly prolonged after both 0.1 and 1.0 mg/kg of intravenous administration of SC51089 (75% prolongation at 1.0 mg/kg) whereas other parameters were not significantly changed compared to vehicle treatment. In addition, the time to the first NVC was also significantly prolonged after 1.0 µg of intrathecal administration of SC51089 (18% prolongation at 1.0 µg) whereas other parameters were not significantly changed. The spinal PGE2 concentration in SCI rats was significantly higher than that in spinal intact rats. The mRNA expressions of EP1 receptors in the both spinal cord and DRG from SCI rats were significantly higher than those from spinal intact rats. CONCLUSIONS: The PGE2-induced activation of EP1 receptors in the spinal cord contributes to the initiation of detrusor overactivity in SCI. Thus, the EP1 receptor could be a therapeutic target for the treatment of neurogenic detrusor overactivity due to SCI.


Asunto(s)
Hidrazinas/uso terapéutico , Oxazepinas/uso terapéutico , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Traumatismos de la Médula Espinal/complicaciones , Vejiga Urinaria Hiperactiva/tratamiento farmacológico , Urodinámica/efectos de los fármacos , Administración Intravenosa , Animales , Femenino , Hidrazinas/farmacología , Oxazepinas/farmacología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/fisiopatología , Vejiga Urinaria Hiperactiva/etiología , Vejiga Urinaria Hiperactiva/fisiopatología
11.
Bioorg Med Chem Lett ; 27(17): 4044-4050, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28784294

RESUMEN

A scaffold-hopping strategy towards a new pyrazolo[1,5-a]pyridine based core using molecular hybridization of two structurally distinct EP1 antagonists, followed by structure-activity relationship-guided optimization, resulted in the identification of potent EP1 antagonists exemplified by 4c, 4f, and 4j, which were shown to reduce pathological intravesical pressure in rats when administered at 1mg/kg iv.


Asunto(s)
Descubrimiento de Drogas , Pirazoles/farmacología , Piridinas/farmacología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Relación Dosis-Respuesta a Droga , Estructura Molecular , Pirazoles/síntesis química , Pirazoles/química , Piridinas/síntesis química , Piridinas/química , Ratas , Relación Estructura-Actividad
12.
Bioorg Med Chem ; 25(13): 3406-3430, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28483455

RESUMEN

Herein we described the design, synthesis and evaluation of a novel series of benzo[d]thiazole derivatives toward an orally active EP1 antagonist. Lead generation studies provided benzo[d]thiazole core from the four designed scaffolds. Optimization of this scaffold in terms of EP1 antagonist potency and ligand-lipophilicity efficiency (LLE; pIC50-clogP) led to a 1,2,3,6-tetrahydropyridyl-substituted benzo[d]thiazole derivative, 7r (IC50 1.1nM; LLE 4.7), which showed a good pharmacological effect when administered intraduodenally in a 17-phenyl trinor-PGE2 (17-PTP)-induced overactive bladder model in rats.


Asunto(s)
Benzotiazoles/farmacología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Vejiga Urinaria Hiperactiva/tratamiento farmacológico , Administración Oral , Animales , Benzotiazoles/administración & dosificación , Benzotiazoles/química , Dinoprostona/análogos & derivados , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ligandos , Estructura Molecular , Ratas , Relación Estructura-Actividad , Vejiga Urinaria Hiperactiva/inducido químicamente
13.
Bioorg Med Chem ; 25(9): 2635-2642, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28342692

RESUMEN

Novel pyrazolo[1,5-a]pyridine derivatives were designed, synthesized and evaluated as orally active EP1 antagonists for the treatment of overactive bladder. Matched molecular pair analysis (MMPA) allowed the design of a new series of pyrazolo[1,5-a]pyridine derivatives 4-6. Structure-activity relationships (SAR) studies of 4-6 were performed, leading to identification of the nanomolar-level EP1 antagonist 4c, which exhibited good pharmacological effect through intraduodenal (id) administration in a 17-phenyltrinor prostaglandin E2-induced bladder contraction model in rats.


Asunto(s)
Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Línea Celular , Masculino , Pirazoles/síntesis química , Pirazoles/farmacocinética , Piridinas/síntesis química , Piridinas/farmacocinética , Ratas Wistar , Relación Estructura-Actividad , Vejiga Urinaria Hiperactiva/tratamiento farmacológico
14.
J Gastroenterol ; 52(10): 1081-1089, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28120127

RESUMEN

BACKGROUND: Patients with proton pump inhibitor (PPI)-refractory gastroesophageal reflux disease (GERD) have unmet clinical needs. Recently, we reported that esophageal prostaglandin E2 (PGE2) plays a crucial role in the generation of heartburn. In the present study, we focused on the PGE2 receptor, EP1, and investigated the effects of ONO-8539, a novel EP1 receptor antagonist, on heartburn symptoms in healthy male volunteers. METHODS: This prospective, double-blind, placebo-controlled, two-period crossover study was performed in 20 healthy male subjects. The novel prostanoid EP1 receptor antagonist, ONO-8539 (450 mg), was administered once 4 h prior to acid perfusion test. During the test, hydrochloric acid (0.15 mol l-1) was perfused into the lower esophagus for 30 min. Acid perception threshold was quantified by the time to first sensation of heartburn and intensity of GI symptoms determined using a validated categorical rating scale, and the area under the curve (AUC) as the total symptom score. RESULTS: ONO-8539 significantly reduced a total heartburn symptom score, not other upper GI symptom scores, during acid perfusion compared with placebo (AUC for heartburn, 85.0 ± 10.6 for placebo and 56.5 ± 7.2 for ONO-8539; P < 0.01), and significantly extended the time to first sensation of heartburn compared with placebo (5.7 ± 4.3 min for placebo and 9.7 ± 7.2 min for ONO-8539; P < 0.05). CONCLUSIONS: ONO-8539 attenuated acid-induced heartburn in healthy male subjects, suggesting that EP1 receptors play a role in generation of heartburn symptoms. ONO-8539 is a potential novel therapeutic option for controlling heartburn symptoms in GERD patients. Clinical Trials Registry No: UMIN000015753.


Asunto(s)
Benzoatos/uso terapéutico , Reflujo Gastroesofágico/tratamiento farmacológico , Fármacos Gastrointestinales/farmacología , Pirosis/tratamiento farmacológico , Indenos/uso terapéutico , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Tiazoles/uso terapéutico , Adulto , Estudios Cruzados , Método Doble Ciego , Reflujo Gastroesofágico/patología , Pirosis/patología , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
15.
Drug Discov Today ; 22(1): 57-71, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506873

RESUMEN

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


Asunto(s)
Preparaciones Farmacéuticas/química , Subtipo EP1 de Receptores de Prostaglandina E , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP3 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E , Animales , Dinoprostona/química , Diseño de Fármacos , Humanos , Ligandos , Estructura Molecular , Terapia Molecular Dirigida , Unión Proteica , Subtipo EP1 de Receptores de Prostaglandina E/agonistas , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP3 de Receptores de Prostaglandina E/agonistas , Subtipo EP3 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores
16.
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
17.
Sci Rep ; 5: 17956, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26648273

RESUMEN

Cyclooxygenase-2 (COX-2) is activated in response to ischemia and significantly contributes to the neuroinflammatory process. Accumulation of COX-2-derived prostaglandin E2 (PGE2) parallels the substantial increase in stroke-mediated blood-brain barrier (BBB) breakdown. Disruption of the BBB is a serious consequence of ischemic stroke, and is mainly mediated by matrix metalloproteinases (MMPs). This study aimed to investigate the role of PGE2 EP1 receptor in neurovascular injury in stroke. We hypothesized that pharmacological blockade or genetic deletion of EP1 protects against BBB damage and hemorrhagic transformation by decreasing the levels and activity of MMP-3 and MMP-9. We found that post-ischemic treatment with the EP1 antagonist, SC-51089, or EP1 genetic deletion results in a significant reduction in BBB disruption and reduced hemorrhagic transformation in an experimental model of transient focal cerebral ischemia. These neurovascular protective effects of EP1 inactivation are associated with a significant reduction in MMP-9/-3, less peripheral neutrophil infiltration, and a preservation of tight junction proteins (ZO-1 and occludin) composing the BBB. Our study identifies the EP1 signaling pathway as an important link between neuroinflammation and MMP-mediated BBB breakdown in ischemic stroke. Targeting the EP1 receptor could represent a novel approach to diminish the devastating consequences of stroke-induced neurovascular damage.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Infarto Encefálico/metabolismo , Infarto Encefálico/patología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Expresión Génica , Técnicas de Inactivación de Genes , Hidrazinas/farmacología , Masculino , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Infiltración Neutrófila , Oxazepinas/farmacología , Permeabilidad/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ratas , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Proteínas de Uniones Estrechas/metabolismo
18.
Ophthalmic Res ; 54(3): 135-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26376245

RESUMEN

PURPOSE: Adenosine triphosphate (ATP) is involved in the tone regulation of retinal arterioles, and the effect may be direct, through ATP degradation or mediated by cyclo-oxygenase products. However, the relative contribution of these mechanisms and the extent to which the mechanisms are active in the retinal vascular wall or depend on the perivascular retinal tissue are unknown. METHODS: Porcine retinal arterioles with perivascular retinal tissue were mounted in a wire myograph for isometric tone recordings. The relaxing effects of ATP and the non-degradable analogue ATP-x03B3;S were studied in the presence of antagonists to ATP, adenosine and prostaglandin E (EP) receptors. The experiments were repeated after removal of the perivascular retinal tissue. RESULTS: ATP induced a significant concentration-dependent relaxation of retinal arterioles (p < 0.05) which was reduced after removal of perivascular retinal tissue. The effect was due to non-degraded ATP and a degradation product of ATP acting via adenosine receptors. Relaxation was reduced by ibuprofen and blocking of EP1 receptors. CONCLUSION: ATP-induced relaxation of retinal arterioles is mediated by ATP, ATP degradation products and by stimulation of EP1 receptors, involving both the perivascular retina and the vascular wall. The findings emphasize the complexity of purinergic effects in the regulation of retinal vascular tone.


Asunto(s)
Adenosina Trifosfato/farmacología , Músculo Liso Vascular/fisiología , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Arteria Retiniana/fisiología , Adenosina Trifosfato/análogos & derivados , Animales , Arteriolas/fisiología , Relación Dosis-Respuesta a Droga , Miografía , Antagonistas Purinérgicos/farmacología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Sus scrofa , Vasodilatación/efectos de los fármacos
19.
BMC Neurosci ; 16: 48, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26232001

RESUMEN

BACKGROUND: Inflammation following intracerebral hemorrhage (ICH) significantly contributes to secondary brain damage and poor outcomes. Prostaglandin E2 (PGE2) is known to modulate neuroinflammatory responses and is upregulated in response to brain injury as a result of changes in inducible cyclooxygenase 2 (COX-2) and the membrane-bound type of PGE synthase. Inhibition of COX-2 activity has been reported to attenuate ICH-induced brain injury; however, the clinical utility of such drugs is limited due to the potential for severe side effects. Therefore, it is now important to search for downstream targets capable of preferentially modulating PGE2 signaling, and the four E prostanoid receptors, EP1-4, which are the main targets of PGE2, remain a viable therapeutic option. We have previously shown that EP1 receptor deletion aggravates ICH-induced brain injury and impairs functional recovery, thus the current study aimed to elaborate on these results by including a pharmacologic approach targeting the EP1 receptor. RESULTS: Chronic post-treatment with the selective EP1 receptor antagonist, SC-51089, increased lesion volume by 30.1 ± 14.5% (p < 0.05) and treatment with the EP1 agonist, 17-pt-PGE2, improved neuromuscular functional recovery on grip strength (p < 0.01) and hanging wire (p < 0.05) behavioral testing. To begin identifying the mechanisms involved in EP1-mediated neuroprotection after ICH, histology was performed to assess ferric iron content, neuroinflammation, leukocyte transendothelial migratory potential, and peripheral neutrophil and immunoglobulin infiltration. Following ICH, mice treated with the antagonist displayed increased ferric iron (p < 0.05) and cortical microgliosis (p < 0.05), whereas treatment with the agonist decreased cortical (p < 0.01) and striatal (p < 0.001) astrogliosis, leukocyte transendothelial migratory potential (p < 0.01), neutrophil infiltration (p < 0.05), and blood brain barrier breakdown (p < 0.05). CONCLUSIONS: In agreement with our previous results, selective antagonism of the EP1 receptor aggravated ICH-induced brain injury. Furthermore, EP1 receptor agonism improved anatomical outcomes and functional recovery. Thus, the present data continues to reinforce a putative role for EP1 as a new and more selective therapeutic target for the treatment of ICH that could reduce the side effects associated with COX-2 inhibition while still exploiting the beneficial effects.


Asunto(s)
Encéfalo/efectos de los fármacos , Hemorragia Cerebral/tratamiento farmacológico , Subtipo EP1 de Receptores de Prostaglandina E/agonistas , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/patología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Encéfalo/inmunología , Encéfalo/patología , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/patología , Colagenasas , Modelos Animales de Enfermedad , Gliosis/tratamiento farmacológico , Gliosis/inmunología , Gliosis/patología , Hidrazinas/farmacología , Hierro/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/patología , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/patología , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Fármacos Neuroprotectores/farmacología , Oxazepinas/farmacología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Recuperación de la Función/efectos de los fármacos
20.
J Cell Mol Med ; 19(10): 2413-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26176998

RESUMEN

Previous work from both our lab and others have indicated that exposure to 50 Hz magnetic fields (ELF-MF) was able to modify ion channel functions. However, very few studies have investigated the effects of MF on γ-aminobutyric acid (GABA) type A receptors (GABA(A) Rs) channel functioning, which are fundamental to overall neuronal excitability. Here, our major goal is to reveal the potential effects of ELF-MF on GABA(A) Rs activity in rat cerebellar granule neurons (CGNs). Our results indicated that exposing CGNs to 1 mT ELF-MF for 60 min. significantly increased GABA(A) R currents without modifying sensitivity to GABA. However, activation of PKA by db-cAMP failed to do so, but led to a slight decrease instead. On the other hand, PKC activation or inhibition by PMA or Bis and Docosahexaenoic acid (DHA) mimicked or eliminated the field-induced-increase of GABA(A) R currents. Western blot analysis indicated that the intracellular levels of phosphorylated PKC (pPKC) were significantly elevated after 60 min. of ELF-MF exposure, which was subsequently blocked by application of DHA or EP1 receptor-specific (prostaglandin E receptor 1) antagonist (SC19220), but not by EP2-EP4 receptor-specific antagonists. SC19220 also significantly inhibited the ELF-MF-induced elevation on GABA(A) R currents. Together, these data obviously demonstrated for the first time that neuronal GABA(A) currents are significantly increased by ELF-MF exposure, and also suggest that these effects are mediated via an EP1 receptor-mediated PKC pathway. Future work will focus on a more comprehensive analysis of the physiological and/or pathological consequences of these effects.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Activación del Canal Iónico , Campos Magnéticos , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , Receptores de GABA-A/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Modelos Biológicos , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Análisis de Regresión , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...