Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
PLoS One ; 19(5): e0303202, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38753641

RESUMO

PURPOSE: This study seeks to investigate the impact of co-administering either a Prostaglandin EP2 receptor agonist or an EP1 receptor antagonist alone with a low dose BMP7 on in vitro healing process, collagen content and maturation of human osteoblasts. METHODOLOGY: Human osteoblast cells were used in this study. These cells were cultured and subjected to different concentrations of Prostaglandin EP2 receptor agonist, EP1 receptor antagonist, BMP7, Control (Ct) (Vehicle alone), and various combinations treatments. Cell viability at 24, 48 and 72 hours (h) was evaluated using the XTT assay. A wound healing assay was conducted to observe the migration ability of human osteoblast cells. Additionally, Sirius red staining and Fourier-Transform Infrared Spectroscopy Imaging (FT-IR) was employed to analyze various parameters, including total protein concentration, collagen production, mature collagen concentration, and mineral content. RESULTS: The combination of low dose BMP7 and Prostaglandin EP2 receptor agonist resulted to the lowest cell viability when compared to both the Ct and individual treatments. In contrast, the Prostaglandin EP1 receptor antagonist alone showed the highest cellular viability at 72 h. In the wound healing assay, the combined treatment of low dose BMP7 with the Prostaglandin EP2 receptor agonist and EP1 receptor antagonist showed a decrease in human osteoblast healing after 24 h. Analysis of FT-IR data indicated a reduction in total protein content, collagen maturity, collagen concentration and mineral content in combination treatment compared to the single or Ct treatments. CONCLUSION: The combination of a Prostaglandin EP2 receptor agonist or an EP1 receptor antagonist when combined with low dose BMP7 significantly hinders both human osteoblast healing and collagen maturity/concentration in comparison to low dose BMP7 treatment alone.


Assuntos
Proteína Morfogenética Óssea 7 , Colágeno , Osteoblastos , Humanos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Colágeno/metabolismo , Proteína Morfogenética Óssea 7/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP2/agonistas , Cicatrização/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Linhagem Celular
2.
J Cancer Res Clin Oncol ; 149(8): 5369-5376, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36436093

RESUMO

PURPOSE: In recent years, incidence of vulvar cancer has been on the rise, whereas therapeutic options are still restricted. Therefore, new prognosticators and therapeutic targets are essential. Chronic inflammation plays an important role in carcinogenesis and COX-2, and its product prostaglandin E2 and its receptors EP1-4 are known to be important mediators in cancer initiation and progression. METHODS: EP1 expression in vulvar cancer specimens (n = 129) was investigated via immunohistochemistry and evaluated using the well-established immunoreactive score (IRS). Subsequently, the values were correlated with clinicopathological parameters. RESULTS: Our analysis did not reveal EP1 expression as a negative prognostic factor in overall and disease-free survival. However, in the subgroup of patients with lymph-node metastasis, overall survival was significantly shorter in tumors with high EP1 expression. Moreover, EP1 expression correlated positively with good differentiation of the tumor, but not with p16 status or COX-2 expression. CONCLUSIONS: This study shed first light on EP1 expression in vulvar carcinoma. EP1 expression correlated significantly with the grading of the tumor, suggesting that it influences cell differentiation. Further research on EP1 signaling may lead to a deeper understanding of the molecular mechanisms of carcinogenesis.


Assuntos
Dinoprostona , Neoplasias Vulvares , Feminino , Humanos , Ciclo-Oxigenase 2/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Carcinogênese
3.
Biochem Pharmacol ; 195: 114858, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34822808

RESUMO

This review article provides an update for the role of prostaglandin E2 receptors (EP1, EP2, EP3 and EP4) in cardiovascular disease. Where possible we have reported citations from the last decade although this was not possible for all of the topics covered due to the paucity of publications. The authors have attempted to cover the subjects of ischemia-reperfusion injury, arrhythmias, hypertension, novel protein binding partners of the EP receptors and their pathophysiological significance, and cardiac regeneration. These latter two topics bring studies of the EP receptors into new and exciting areas of research that are just beginning to be explored. Where there is peer-reviewed literature, the authors have placed particular emphasis on clinical studies although these are limited in number.


Assuntos
Doenças Cardiovasculares/metabolismo , Dinoprostona/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Animais , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Humanos , Modelos Cardiovasculares , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Regeneração
4.
Sci Rep ; 10(1): 11186, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32636414

RESUMO

A close association between pericytes and endothelial cells (ECs) is crucial to the stability and function of capillary blood vessels and microvessels. The loss or dysfunction of pericytes results in significant disruption of these blood vessels as observed in pathological conditions, including cancer, diabetes, stroke, and Alzheimer's disease. Prostaglandin E2 (PGE2) is a lipid mediator of inflammation, and its tissue concentration is elevated in cancer and neurological disorders. Here, we show that the exposure to PGE2 switches pericytes to a fast-migrating, loosely adhered phenotype that fails to intimately interact with ECs. N-cadherin and connexin-43 in adherens junction and gap junction between pericytes and ECs are downregulated by EP-4 and EP-1-dependent mechanisms, leading to breakdown of the pericyte-EC interaction. Furthermore, R-Ras, a small GTPase important for vascular normalization and vessel stability, is transcriptionally repressed by PGE2 in an EP4-dependent manner. Mouse dermal capillary vessels lose pericyte coverage substantially upon PGE2 injection into the skin. Our results suggest that EP-mediated direct disruption of pericytes by PGE2 is a key process for vascular destabilization. Restoring pericyte-EC interaction using inhibitors of PGE2 signaling may offer a therapeutic strategy in cancer and neurological disorders, in which pericyte dysfunction contributes to the disease progression.


Assuntos
Dinoprostona/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Junções Aderentes/metabolismo , Animais , Caderinas/metabolismo , Movimento Celular , Células Cultivadas , Conexina 43/metabolismo , Regulação para Baixo , Junções Comunicantes/metabolismo , Células HEK293 , Células HT29 , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pericitos/metabolismo , Pericitos/fisiologia , Proteínas ras/metabolismo
5.
Ecotoxicol Environ Saf ; 201: 110712, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32502905

RESUMO

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.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/efeitos dos fármacos , Manganês/toxicidade , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Reprodução/efeitos dos fármacos , Animais , Hipotálamo/metabolismo , Masculino , Manganês/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores
6.
Lab Invest ; 100(3): 414-425, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31527829

RESUMO

Prostaglandin E2 receptor EP1 (PGE2/EP1) promotes diabetic renal injury, and EP1 receptor deletion improves hyperfiltration, albuminuria, and fibrosis. The role of EP1 receptors in hypertensive kidney disease (HKD) remains controversial. We examined the contribution of EP1 receptors to HKD. EP1 null (EP1-/-) mice were bred with hypertensive TTRhRen mice (Htn) to evaluate kidney function and injury at 24 weeks. EP1 deletion had no effect on elevation of systolic blood pressure in Htn mice (HtnEP1-/-) but resulted in pronounced albuminuria and reduced FITC-inulin clearance, compared with Htn or wild-type (WT) mice. Ultrastructural injury to podocytes and glomerular endothelium was prominent in HtnEP1-/- mice; including widened subendothelial space, subendothelial lucent zones and focal lifting of endothelium from basement membrane, with focal subendothelial cell debris. Cortex COX2 mRNA was increased by EP1 deletion. Glomerular EP3 mRNA was reduced by EP1 deletion, and EP4 by Htn and EP1 deletion. In WT mice, PGE2 increased chloride reabsorption via EP1 in isolated perfused thick ascending limb (TAL), but PGE2 or EP1 deletion did not affect vasopressin-mediated chloride reabsorption. In WT and Htn mouse inner medullary collecting duct (IMCD), PGE2 inhibited vasopressin-water transport, but not in EP1-/- or HtnEP1-/- mice. Overall, EP1 mediated TAL and IMCD transport in response to PGE2 is unaltered in Htn, and EP1 is protective in HKD.


Assuntos
Hipertensão Renal , Podócitos , Receptores de Prostaglandina E Subtipo EP1 , Animais , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Deleção de Genes , Taxa de Filtração Glomerular/genética , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Transgênicos , Podócitos/citologia , Podócitos/metabolismo , Podócitos/patologia , Receptores de Prostaglandina E Subtipo EP1/genética , Receptores de Prostaglandina E Subtipo EP1/metabolismo
7.
J Microbiol Biotechnol ; 29(10): 1675-1681, 2019 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-31474101

RESUMO

Clostridium difficile toxin A is known to cause colonic epithelial cell apoptosis, which is considered the main causative event that triggers inflammatory responses in the colon, reflecting the concept that the essential role of epithelial cells in the colon is to form a physical barrier in the gut. We previously showed that toxin A-induced colonocyte apoptosis and subsequent inflammation were dependent on prostaglandin E2 (PGE2) produced in response to toxin A stimulation. However, the molecular mechanism by which PGE2 mediates cell apoptosis in toxin A-exposed colonocytes has remained unclear. Here, we sought to identify the signaling pathway involved in toxin A-induced, PGE2-mediated colonocyte apoptosis. In non-transformed NCM460 human colonocytes, toxin A exposure strongly upregulated expression of Bak, which is known to form mitochondrial outer membrane pores, resulting in apoptosis. RT-PCR analyses revealed that this increase in Bak expression was attributable to toxin A-induced transcriptional upregulation. We also found that toxin A upregulation of Bak expression was dependent on PGE2 production, and further showed that this effect was recapitulated by an EP1 receptor agonist, but not by agonists of other EP receptors. Collectively, these results suggest that toxin A-induced cell apoptosis involves PGE2-upregulation of Bak through the EP1 receptor.


Assuntos
Toxinas Bacterianas/farmacologia , Clostridioides difficile/metabolismo , Dinoprostona/metabolismo , Enterotoxinas/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Transformada , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Receptores de Prostaglandina E Subtipo EP1/agonistas , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Transdução de Sinais , Ativação Transcricional/efeitos dos fármacos , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
8.
PLoS One ; 14(8): e0220987, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31393950

RESUMO

We showed previously an epinephrine-induced inhibition of the Na+/K+ ATPase in Caco-2 cells mediated via PGE2. This work is an attempt to further elucidate mediators downstream of PGE2 and involved in the observed inhibitory effect. The activity of the Na+/K+ ATPase was assayed by measuring the amount of inorganic phosphate liberated in presence and absence of ouabain, a specific inhibitor of the enzyme. Changes in the protein expression of the Na+/K+ ATPase were investigated by western blot analysis which revealed a significant decrease in the abundance of the ATPase in plasma membranes. Treating the cells with epinephrine or PGE2 in presence of SC19220, a blocker of EP1 receptors abolished completely the effect of the hormone and the prostaglandin while the effect was maintained unaltered in presence of antagonists to all other receptors. Treatment with calphostin C, PTIO, ODQ or KT5823, respective inhibitors of PKC, NO, soluble guanylate cyclase and PKG, abrogated completely the effect of epinephrine and PGE2, suggesting an involvement of these mediators. A significant inhibition of the ATPase was observed when cells were treated with PMA, an activator of PKC or with 8-Br-cGMP, a cell permeable cGMP analogue. PMA did reduce the protein expression of IκB, as shown by western blot analysis, and its effect on the ATPase was not manifested in presence of an inhibitor of NF-κB while that of SNAP, a nitric oxide donor, was not affected. The results infer that NF-κB is downstream PKC and upstream NO. The data support a pathway in which epinephrine induces the production of PGE2 which binds to EP1 receptors and activates PKC and NF-κB leading to NO synthesis. The latter activates soluble guanylate cyclase resulting in cGMP production and activation of PKG which through direct or indirect phosphorylation inhibits the Na+/K+ ATPase by inducing its internalization.


Assuntos
Dinoprostona/farmacologia , Epinefrina/farmacologia , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Proteína Quinase C/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Células CACO-2 , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Guanilil Ciclase Solúvel/metabolismo
9.
Pharmacology ; 104(5-6): 267-275, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31434088

RESUMO

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.


Assuntos
Alprostadil/análogos & derivados , Carcinoma Hepatocelular/tratamento farmacológico , Catequina/análogos & derivados , Neoplasias Hepáticas/tratamento farmacológico , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Alprostadil/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Catequina/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dinoprostona/metabolismo , Sinergismo Farmacológico , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Cicatrização/efeitos dos fármacos
10.
Bosn J Basic Med Sci ; 19(3): 265-273, 2019 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-30995899

RESUMO

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.


Assuntos
Neoplasias Ósseas/patologia , Dinoprostona/farmacologia , Osteossarcoma/patologia , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inativação Gênica , Humanos , Hidrazinas/farmacologia , Camundongos , Camundongos Nus , Oxazepinas/farmacologia , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP1/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Pharm Pharm Sci ; 21(1): 326-339, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30075829

RESUMO

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.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Dinoprostona/farmacologia , Fibroblastos/efeitos dos fármacos , Fibrose/induzido quimicamente , Animais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Fibrose/metabolismo , Injeções Intraperitoneais , Masculino , Interferência de RNA/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP1/metabolismo
12.
Mol Pharm ; 15(9): 4121-4131, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30109938

RESUMO

Diabetics often face greater risk of cognitive impairment than nondiabetics. However, how to prevent this disease is still unconfirmed. In this study, we investigated the potential protection and mechanism of meloxicam on cognitive impairment in diabetic rats. The diabetic rat model was established with a high-fat diet and a small dose of streptozotocin (40 mg/kg). The changes of spatial learning and memory, histopathology, and the protein expressions of amyloid protein precursor (APP) and ß-amyloid (Aß) indicated that diabetic rats had neuronal injury and cognitive impairment. Tumor necrosis factor α (TNFα), interleukin 6 (IL-6), C reactive protein (CRP) and prostaglandin E2 (PGE2) levels, and microglial cell number were significantly increased in the diabetic rat brain. Meanwhile, the protein expressions of APP, Aß, cyclooxygenases2 (COX2), E-type prostanoid recptors 1 (EP1) and EP2, and the level of cyclic adenosine monophosphate (cAMP) were significantly increased, while the protein expressions of EP3 and phosphorylated protein kinase A (pPKA) were significantly decreased in the diabetic rat hippocampus and cortex. However, the EP4 protein expression had no significant changes. Meloxicam significantly improved neuronal injury and cognitive impairment, and significantly decreased inflammatory cytokines levels. Meloxicam also significantly decreased the protein expressions of APP, Aß, COX2, EP1 and EP2, and the level of cAMP and significantly increased the EP3 and pPKA protein expressions in rat hippocampus and cortex. However, meloxicam did not significantly influence the levels of blood glucose, lipids, and insulin of rats. Our results suggest that meloxicam could significantly protect diabetic rats from cognitive impairment via a mechanism that may be associated with rebalancing the COX2-PGE2-EPs-cAMP/PKA pathway.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/metabolismo , Meloxicam/uso terapêutico , Animais , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Disfunção Cognitiva/sangue , Imuno-Histoquímica , Inflamação/sangue , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Insulina/sangue , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Bioorg Med Chem Lett ; 28(14): 2408-2412, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29934246

RESUMO

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.


Assuntos
Descoberta de Drogas , Consolidação da Fratura/efeitos dos fármacos , Fraturas Ósseas/tratamento farmacológico , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Tiazóis/farmacologia , Animais , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Fraturas Ósseas/metabolismo , Células Madin Darby de Rim Canino/efeitos dos fármacos , Células Madin Darby de Rim Canino/metabolismo , Células Madin Darby de Rim Canino/patologia , Camundongos , Camundongos Knockout , Estrutura Molecular , Receptores de Prostaglandina E Subtipo EP1/deficiência , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Relação Estrutura-Atividade , Tiazóis/química
14.
Biochem Biophys Res Commun ; 500(2): 435-442, 2018 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-29660334

RESUMO

BACKGROUND: Effects of lutein (L) and fatty acids [linoleic acid (LA), eicosapentaenoic acid (EPA)+docosahexaenoic acid (DHA) and oleic acid (OA)] on oxidative stress and inflammation in cataract were assessed. METHODS: Cataract was induced in male Wistar rat pups (11 days old) by giving a single dose of sodium selenite (25 µM/kg body weight) by IP. Lutein (1.3 µmol/kg body weight) was given one day before and five days after selenite injection as a micelle with 7.5 mM LA, or 7.5 mM EPA + DHA or 7.5 mM OA. Serum and lens oxidative stress and inflammatory parameters having a bearing cataract were assessed. RESULTS: Serum and lens nitric oxide, MDA and protein carbonyls were significantly (p < 0.05) increased in cataract compared to control and experimental groups. Catalase, SOD, glutathione peroxidase and glutathione transferase activity and glutathione level in serum and lens of cataract group were significantly (p < 0.05) decreased. Serum eicosanoids (PGE2, LTB4, and LTC4) and cytokines (CRP, TNF-α, IL1-ß, and MCP-1) were significantly (p < 0.05) increased in cataract. The activity of cPLA2 and Cox-2 in cataract lens was higher (p < 0.05) compared to other groups. EP-1, NOS-2 and NF-kB expression were higher (p < 0.05) in cataract. The ratio of water insoluble to water soluble protein was increased in cataract lens. Group administered with L + EPA + DHA exhibited highest cataract prevention compared to L + LA and L + OA. Pups given lutein with EPA + DHA had the highest amount of lutein in the lens. CONCLUSIONS: The anti-cataract activity of lutein was influenced by fatty acids and found to be highest with EPA + DHA compared to LA or OA.


Assuntos
Catarata/tratamento farmacológico , Catarata/prevenção & controle , Ácidos Graxos/uso terapêutico , Inflamação/tratamento farmacológico , Luteína/uso terapêutico , Estresse Oxidativo , Animais , Antioxidantes/metabolismo , Biomarcadores/sangue , Catarata/sangue , Ciclo-Oxigenase 2/metabolismo , Citocinas/sangue , Eicosanoides/sangue , Proteínas do Olho/metabolismo , Ácidos Graxos/farmacologia , Glutationa/sangue , Glutationa/metabolismo , Inflamação/patologia , Cristalino/metabolismo , Cristalino/patologia , Luteína/farmacologia , Masculino , Modelos Biológicos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfolipases A2 Citosólicas/metabolismo , Ratos , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Solubilidade , Água
16.
Lab Invest ; 98(3): 360-370, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29251736

RESUMO

PGE2 regulates glomerular hemodynamics, renin secretion, and tubular transport. This study examined the contribution of PGE2 EP1 receptors to sodium and water homeostasis. Male EP1-/- mice were bred with hypertensive TTRhRen mice (Htn) to evaluate blood pressure and kidney function at 8 weeks of age in four groups: wildtype (WT), EP1-/-, Htn, HtnEP1-/-. Blood pressure and water balance were unaffected by EP1 deletion. COX1 and mPGE2 synthase were increased and COX2 was decreased in mice lacking EP1, with increases in EP3 and reductions in EP2 and EP4 mRNA throughout the nephron. Microdissected proximal tubule sglt1, NHE3, and AQP1 were increased in HtnEP1-/-, but sglt2 was increased in EP1-/- mice. Thick ascending limb NKCC2 was reduced in the cortex but increased in the medulla. Inner medullary collecting duct (IMCD) AQP1 and ENaC were increased, but AVP V2 receptors and urea transporter-1 were reduced in all mice compared to WT. In WT and Htn mice, PGE2 inhibited AVP-water transport and increased calcium in the IMCD, and inhibited sodium transport in cortical collecting ducts, but not in EP1-/- or HtnEP1-/- mice. Amiloride (ENaC) and hydrochlorothiazide (pendrin inhibitor) equally attenuated the effect of PGE2 on sodium transport. Taken together, the data suggest that EP1 regulates renal aquaporins and sodium transporters, attenuates AVP-water transport and inhibits sodium transport in the mouse collecting duct, which is mediated by both ENaC and pendrin-dependent pathways.


Assuntos
Dinoprostona/metabolismo , Hipertensão/metabolismo , Túbulos Renais Coletores/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Sódio/metabolismo , Animais , Aquaporinas/metabolismo , Pressão Sanguínea , Cálcio/metabolismo , Taxa de Filtração Glomerular , Masculino , Camundongos , Prostaglandina-E Sintases/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Trocador 3 de Sódio-Hidrogênio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
17.
Histochem Cell Biol ; 149(2): 153-160, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29134301

RESUMO

Prostaglandin E2 (PGE2) is well described to be associated with both endometrial functions and disorders. The primary aim of this study was to explore the underlying mechanisms that affect the growth and function of endometrial epithelium and stroma by assessing the staining intensity of PGE2 receptors (EP) in healthy endometrium across the menstrual cycle and in pathological endometrium, such as ovarian endometriosis and endometrial cancer. We retrospectively analyzed the EPs staining intensity in human nonpregnant endometrium throughout the menstrual cycle by immunohistochemistry and further focused on EP1 (n = 42). The variation of EP1 was compared among healthy endometrium, ovarian endometriosis (n = 14), and endometrial cancer (n = 140) crosswise. EP1 presented cyclical changes with increased intensity in both epithelium and stroma during the proliferative phase. EP1 staining in the epithelium was increased in endometriotic tissue compared to healthy endometrium and tumor tissue, while in the stroma, the staining in the tumor was lower than that in both normal tissue and endometriosis. No significant differences in EP1 intensity were detected for histological, stage, grading, metastatic and recurrent subtypes in endometrial cancer. EP1 was also correlated with neither progression-free survival nor overall survival of patients with cancer. EP1 staining in progesterone receptor B (PRB)-positive tumor was stronger compared to PRB-negative tumor. EP1 may play a role in human endometrial physiology and pathology. Further studies on the effect of EP1 on human endometrium are needed.


Assuntos
Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Adulto , Idoso , Neoplasias do Endométrio/química , Neoplasias do Endométrio/patologia , Endométrio/química , Endométrio/diagnóstico por imagem , Feminino , Humanos , Pessoa de Meia-Idade , Receptores de Prostaglandina E Subtipo EP1/análise
18.
Am J Physiol Renal Physiol ; 313(4): F1038-F1049, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28701311

RESUMO

During the early phase of ANG II-dependent hypertension, tubular PGE2 is increased. Renin synthesis and secretion in the collecting duct (CD) are upregulated by ANG II, contributing to further intratubular ANG II formation. However, what happens first and whether the triggering mechanism is independent of tubular ANG II remain unknown. PGE2 stimulates renin synthesis in juxtaglomerular cells via E-prostanoid (EP) receptors through the cAMP/cAMP-responsive element-binding (CREB) pathway. EP receptors are also expressed in the CD. Here, we tested the hypothesis that renin is upregulated by PGE2 in CD cells. The M-1 CD cell line expressed EP1, EP3, and EP4 but not EP2. Dose-response experiments, in the presence of ANG II type 1 receptor blockade with candesartan, demonstrated that 10-6 M PGE2 maximally increases renin mRNA (approximately 4-fold) and prorenin/renin protein levels (approximately 2-fold). This response was prevented by micromolar doses of SC-19220 (EP1 antagonist), attenuated by the EP4 antagonist, L-161982, and exacerbated by the highly selective EP3 antagonist, L-798106 (~10-fold increase). To evaluate further the signaling pathway involved, we used the PKC inhibitor calphostin C and transfections with PKCα dominant negative. Both strategies blunted the PGE2-induced increases in cAMP levels, CREB phosphorylation, and augmentation of renin. Knockdown of the EP1 receptor and CREB also prevented renin upregulation. These results indicate that PGE2 increases CD renin expression through the EP1 receptor via the PKC/cAMP/CREB pathway. Therefore, we conclude that during the early stages of ANG II-dependent hypertension, there is augmentation of PGE2 that stimulates renin in the CD, resulting in increased tubular ANG II formation and further stimulation of renin.


Assuntos
Proteína de Ligação a CREB/metabolismo , AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Proteína Quinase C/metabolismo , Receptores de Prostaglandina E Subtipo EP1/agonistas , Sistema Renina-Angiotensina/efeitos dos fármacos , Renina/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Proteína de Ligação a CREB/genética , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Túbulos Renais Coletores/enzimologia , Camundongos , Simulação de Acoplamento Molecular , Fosforilação , Antagonistas de Prostaglandina/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Receptores de Prostaglandina E Subtipo EP1/genética , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Renina/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Regulação para Cima
19.
J Pept Sci ; 23(7-8): 650-658, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28626925

RESUMO

G-protein-coupled receptors (GPCRs) form the largest family of transmembrane receptors, and their oligomerization has been suggested to be related to their functions. Despite extensive studies, their oligomeric states are highly controversial. One of the reasons is the overestimation of oligomerization by conventional methods. We recently established a stoichiometric analysis method for precisely determining the oligomeric state of membrane proteins on living cells with the combined use of the coiled-coil labeling method and a spectral imaging technique and showed that the prototypical class-A GPCR ß2 -adrenergic receptor (ß2 AR) did not form functional oligomers. In this study, we expanded our study to three well-studied class-A GPCRs: C-X-C chemokine receptor of stromal cell-derived factor-1α (CXCR4), dopamine receptor D2 short isotype (D2R), and prostaglandin E receptor subtype 1 (EP1R). We found that these receptors did not form constitutive homooligomers. The receptors exhibited calcium signaling upon agonist stimulation as monomers, although CXCR4 and EP1R gradually clustered after fast signaling. We conclude that homooligomerization is not necessary for the signal transductions of these four class-A GPCRs. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.


Assuntos
Receptores CXCR4/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células CHO , Cricetulus , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Microscopia Confocal , Receptores CXCR4/genética , Receptores de Dopamina D2/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Prostaglandina E Subtipo EP1/genética , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Transdução de Sinais
20.
J Cell Biochem ; 118(12): 4383-4393, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28444901

RESUMO

Mesenchymal stromal cells (MSCs) are multipotent progenitors capable of differentiation into osteoblasts and can potentially serve as a source for cell-based therapies for bone repair. Many factors have been shown to regulate MSC differentiation into the osteogenic lineage such as the Cyclooxygenase-2 (COX2)/Prostaglandin E2 (PGE2) signaling pathway that is critical for bone repair. PGE2 binds four different receptors EP1-4. While most studies focus on the role PGE2 receptors EP2 and EP4 in MSC differentiation, our study focuses on the less studied, receptor subtype 1 (EP1) in MSC function. Recent work from our laboratory showed that EP1-/- mice have enhanced fracture healing, stronger cortical bones, higher trabecular bone volume and increased in vivo bone formation, suggesting that EP1 is a negative regulator of bone formation. In this study, the regulation of MSC osteogenic differentiation by EP1 receptor was investigated using EP1 genetic deletion in EP1-/- mice. The data suggest that EP1 receptor functions to maintain MSCs in an undifferentiated state. Loss of the EP1 receptor changes MSC characteristics and permits stem cells to undergo more rapid osteogenic differentiation. Notably, our studies suggest that EP1 receptor regulates MSC differentiation by modulating MSC bioenergetics, preventing the shift to mitochondrial oxidative phosphorylation by maintaining high Hif1α activity. Loss of EP1 results in inactivation of Hif1α, increased oxygen consumption rate and thus increased osteoblast differentiation. J. Cell. Biochem. 118: 4383-4393, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular , Metabolismo Energético , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteogênese , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Animais , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Knockout , Consumo de Oxigênio , Receptores de Prostaglandina E Subtipo EP1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA