Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
1.
Cell Rep ; 40(11): 111323, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36103815

RESUMO

Prostaglandin receptors have been implicated in a wide range of functions, including inflammation, immune response, reproduction, and cancer. Our group has previously determined the crystal structure of the active-like EP3 bound to its endogenous agonist, prostaglandin E2. Here, we present the single-particle cryoelectron microscopy (cryo-EM) structure of the human EP3-Gi signaling complex at a resolution of 3.4 Å. The structure reveals the binding mode of Gi to EP3 and the structural changes induced in EP3 by Gi binding. In addition, we compare the structure of the EP3-Gi complex with other subtypes of prostaglandin receptors (EP2 and EP4) bound to Gs that have been previously reported and examine the differences in amino acid composition at the receptor-G protein interface. Mutational analysis reveals that the selectivity of the G protein depends on specific amino acid residues in the second intracellular loop and TM5.


Assuntos
Dinoprostona , Receptores de Prostaglandina E , Aminoácidos , Microscopia Crioeletrônica , Dinoprostona/farmacologia , Humanos , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo
2.
Parasite Immunol ; 42(6): e12713, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32173875

RESUMO

Canine leishmaniasis (CanL) is caused by the intracellular parasite Leishmania infantum. Prostaglandin E2 (PGE2 ) exerts potent regulatory effects on the immune system in experimental model Leishmania infection, but this influence has not yet been studied in CanL. In this study, PGE2 and PGE2 receptor levels and the regulatory effect of PGE2 on arginase activity, NO2 , IL-10, IL-17, IFN-γ, TNF-α and parasite load were evaluated in cultures of splenic leucocytes obtained from dogs with CanL in the presence of agonists and inhibitors. Our results showed that splenic leucocytes from dogs with CanL had lower EP2 receptor levels than those of splenic leucocytes from healthy animals. We observed that NO2 levels decreased when the cells were treated with a PGE2 receptor agonist (EP1/EP2/EP3) or COX-2 inhibitor (NS-398) and that TNF-α, IL-17 and IFN-γ cytokine levels decreased when the cells were treated with a PGE2 receptor agonist (EP2) or PGE2 itself. The parasite load in splenic leucocyte cell cultures from dogs with CanL decreased after stimulation of the cells with PGE2 . We conclude that Leishmania infection of dogs modulates PGE2 receptors and speculate that the binding of PGE2 to its receptors may activate the microbicidal capacity of cells.


Assuntos
Citocinas/imunologia , Dinoprostona/metabolismo , Doenças do Cão/tratamento farmacológico , Leishmania infantum/imunologia , Leishmaniose/veterinária , Receptores de Prostaglandina E/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/agonistas , Dinoprostona/antagonistas & inibidores , Doenças do Cão/imunologia , Doenças do Cão/parasitologia , Cães , Leishmaniose/tratamento farmacológico , Leishmaniose/imunologia , Óxido Nítrico/análise , Nitrobenzenos/farmacologia , Carga Parasitária , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/fisiologia , Sulfonamidas/farmacologia , Fator de Necrose Tumoral alfa/imunologia
3.
Endocrinology ; 159(10): 3482-3491, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30085093

RESUMO

During the periovulatory period, the profile of fibroblast growth factor 2 (FGF2) coincides with elevated prostaglandin E2 (PGE2) levels. We investigated whether PGE2 can directly stimulate FGF2 production in bovine granulosa cells and, if so, which prostaglandin E2 receptor (PTGER) type and signaling cascades are involved. PGE2 temporally stimulated FGF2. Accordingly, endoperoxide-synthase2-silenced cells, exhibiting low endogenous PGE2 levels, had reduced FGF2. Furthermore, elevation of viable granulosa cell numbers by PGE2 was abolished with FGF2 receptor 1 inhibitor, suggesting that FGF2 mediates this action of PGE2. Epiregulin (EREG), a known PGE2-inducible gene, was studied alongside FGF2. PTGER2 agonist elevated cAMP as well as FGF2 and EREG levels. However, a marked difference between cAMP-induced downstream signaling was observed for FGF2 and EREG. Whereas FGF2 upregulated by PGE2, PTGER2 agonist, or forskolin was unaffected by the protein kinase A (PKA) inhibitor H89, EREG was significantly inhibited. FGF2 was dose-dependently stimulated by the exchange protein directly activated by cAMP (EPAC) activator; a similar induction was observed for EREG. However, forskolin-stimulated FGF2, but not EREG, was inhibited in EPAC1-silenced cells. These findings ascribe a novel autocrine role for PGE2, namely, elevating FGF2 production in granulosa cells. This study also reveals that cAMP-activated EPAC1, rather than PKA, mediates the effect of PGE2/PTGER2 on the expression of FGF2. Stimulation of EREG by PGE2 is also mediated by PTGER2 but, in contrast to FGF2, EREG was found to be PKA sensitive. PGE2-stimulated FGF2 can act to maintain granulosa cell survival; it can also act on ovarian endothelial cells to promote angiogenesis.


Assuntos
AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células da Granulosa/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Animais , Bovinos , Células Cultivadas , Epirregulina/genética , Epirregulina/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Expressão Gênica/efeitos dos fármacos , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Modelos Biológicos , Interferência de RNA , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/metabolismo , Transdução de Sinais
4.
Theriogenology ; 95: 8-17, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28460684

RESUMO

In previous work, an EP2 prostanoid receptor (EP2R) agonist in vivo increased mRNA expression of luteal LH receptors (LHR), unoccupied and occupied luteal; LHR, and circulating progesterone, while an EP3R or FPR agonist decreased; mRNA expression of luteal LHR, unoccupied and occupied luteal LHR, and; circulating progesterone. An EP4R and lysophosphatidic acid (LPA) LPA2R and LPA3R agonists were reported to inhibit luteal function and sirtuins have been proposed to increase prostaglandin synthesis. The objectives were to determine; whether an EP4R, LPA2R, or LPA3R agonist affect ovine luteal function in vivo or; in vitro. In addition, whether sirtuin (SIRT)-1, 2, or 3; LPA2R or LPA3R; or EP1R, EP2R, EP3R, or EP4R agonists affect caruncular endometrial PGF2α or PGE (PGE1+PGE2) secretion in vitro. Day-10 nonpregnant ewes received a single injection of Vehicle (N = 5); an LPA2R (N = 5); LPA3R (N = 6); or EP4R (N = 5) agonist given into the interstitial tissue of the ovarian vascular pedicle adjacent to the luteal-containing ovary to determine effects on circulating progesterone, mRNA expression of luteal LHR, and luteal unoccupied and occupied LHR. In addition, agonists for LPA2R, LPA3R, EP1R, EP2R, EP3R, or EP4R or SIRT-1, SIRT-2, or SIRT-3 activators were incubated with caruncular endometrial slices in vitro to determine their effect on caruncular endometrial PGF2α, or PGE secretion. LPA2R, LPA3R, or an EP4R agonist in vivo did not affect (P ≥ 0.05) luteal weight, circulating progesterone, or occupied luteal LHR. However, an LPA2R or EP4R agonist, but; not LPA3R agonist, in vivo increased (P ≤ 0.05) mRNA expression of luteal LHR. An; LPA2R, LPA3R, or EP4R agonist increased (P ≤ 0.05) luteal unoccupied LHR, but; not occupied LHR. An LPA2R, LPA3R, or an EP4R agonist did not affect (P ≥ 0.05); luteal progesterone secretion in vitro. An LPA2R or LPA3R agonist did not affect (P ≥ 0.05) luteal PGF2α, or PGE secretion in vitro. However, an EP4R agonist tended to decrease (P < 0.066) luteal PGF2α secretion and increased (P ≤ 0.05) luteal PGE; secretion in vitro. EP1R, EP2R, EP3R, or an EP4R agonist did not affect (P ≥ 0.05); caruncular endometrial PGF2α secretion in vitro. However, EP1R, EP3R, or an EP4R agonist increased caruncular endometrial PGE secretion in vitro, while two different EP2R agonists did not affect (P ≥ 0.05) caruncular endometrial PGE; secretion. A SIRT-1 activator, but not SIRT-2 or SIRT-3 activators, increased (P ≤ 0.05) caruncular endometrial PGE secretion, while sirtuin 1, 2, or 3 activators did not affect (P ≥ 0.05) caruncular endometrial PGF2α secretion. In conclusion, receptors for EP4, LPA2, and LPA3 do not appear to be involved; in luteolysis, but EP4R and LPA2R might participate in preventing luteolysis by maintaining luteal mRNA expression for LHR and preventing loss of unoccupied luteal LHR. In addition, SIRT-1, EP1R, EP3R, and EP4R might be involved in; regulating caruncular endometrial PGE secretion, but not PGF2α secretion.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Prostaglandinas E/metabolismo , Receptores de Ácidos Lisofosfatídicos/agonistas , Receptores de Prostaglandina E Subtipo EP4/agonistas , Receptores de Prostaglandina E/agonistas , Sirtuínas/farmacologia , Alprostadil/metabolismo , Animais , Corpo Lúteo/fisiologia , Dinoprosta/metabolismo , Endométrio/fisiologia , Feminino , Lisofosfolipídeos/agonistas , Lisofosfolipídeos/antagonistas & inibidores , Progesterona/sangue , Progesterona/metabolismo , RNA Mensageiro/metabolismo , Ovinos
5.
Eur J Pharmacol ; 749: 124-32, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25614334

RESUMO

Extracellular nucleotides act as inflammatory mediators through activation of multiple purinoceptors. Under inflammatory conditions, the purinergic signalling is affected by various inflammatory mediators. We previously showed that prostaglandin (PG) E2 suppressed the elevation of intracellular Ca(2+) concentration ([Ca(2+)]i) stimulated by P2X4, P2Y2, and P2Y6 receptors in J774 murine macrophages. In this study, we examined the mechanism of PGE2 inhibitory effects on P2Y6 receptor-mediated function in J774 cells. The P2Y6 receptor agonist UDP induced a sustained elevation of [Ca(2+)]i by stimulating the phospholipase C (PLC) signalling pathway. PGE2 inhibited [Ca(2+)]i elevation and phosphatidylinositol (PI) hydrolysis in a concentration-dependent manner. J774 cells highly expressed the E-type prostanoid 2 (EP2) receptor subtype, a Gs-coupled receptor. PGE2 and a selective EP2 receptor agonist caused cyclic AMP (cAMP) accumulation in J774 cells. The inhibitory effects of PGE2 on P2Y6 receptor-mediated responses were mimicked by the selective EP2 receptor agonist. Although EP2 receptor is linked to adenylyl cyclase activation, PGE2-induced inhibition of Ca(2+) response and PI hydrolysis could not be mimicked by a lipophilic cAMP derivative, dibutyryl cAMP, or an adenylyl cyclase activator, forskolin. The inhibition of UDP-induced PLC activation by PGE2 was not affected by down-regulation of protein kinase C by phorbol-12-myristate-13-acetate treatment. PGE2 inhibited PLC activation induced by aluminium fluoride, but not by the Ca(2+)-ionophore, ionomycin. Finally, the inhibition of UDP-induced PLC activation by PGE2 was impaired by Gs knockdown using siRNA. These results suggest that EP2 receptor activation in macrophages negatively controls the Gq/11-PLC signalling through a Gs-mediated, but cAMP-independent signalling mechanism.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Dinoprostona/farmacologia , Macrófagos/efeitos dos fármacos , Receptores de Prostaglandina E/metabolismo , Receptores Purinérgicos P2/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Macrófagos/metabolismo , Camundongos , Receptores de Prostaglandina E/agonistas
6.
Mol Med Rep ; 11(6): 4454-62, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25625500

RESUMO

Inflammation is a potent promoter of tumor metastasis. The aim of the present study was to explore the function of systemic inflammation in the formation of lung metastasis of breast cancer cells in a mouse model. BALB/c mice were injected intraperitoneally with lipopolysaccharide (LPS) in order to establish an inflammatory animal model and 4T1 murine breast cancer cells were injected through the tail vein to induce lung metastasis. The levels of proinflammatory cytokines were evaluated by ELISA. Metastases on the surface of the lungs were counted and histologically analyzed by hematoxylin and eosin staining. Angiogenesis in the lungs was examined by CD31 immunofluorescence. Mouse pulmonary endothelial cells (MPVECs) were isolated and used to assay endothelial tube formation and determine the protein expression levels of vascular endothelial growth factor (VEGF) in vitro. Serum levels of VEGF and prostaglandin E2 (PGE2), the number and size of metastatic lesions, and the expression levels of cyclooxygenase­2 were significantly greater in the lungs of LPS­treated mice, as compared with those in control mice threated with phosphate­buffered saline. Blood vessel density was also markedly increased in the LPS­treated mice. These increases were reversed by treatment with celecoxib. In vitro, the protein expression levels of VEGF produced by the PGE2­treated cells were significantly increased in a concentration­dependent manner. In addition, the production of VEGF was increased in response to treatment with the PGE2 receptor (EP2) agonist ONO­AE1­259­01; however, this increase was abrogated by treatment with AH6809, an EP2 receptor antagonist. Treatment with PGE2 or VEGF alone promoted the tube formation of MPVECs and this effect was reversed by treatment with celecoxib. These results demonstrated that PGE2 may regulate the release of VEGF by MPVECs through the EP2 receptor pathway and thereby promoted pulmonary angiogenesis and breast cancer metastasis in a mouse model.


Assuntos
Neoplasias da Mama/patologia , Dinoprostona/metabolismo , Lipopolissacarídeos/toxicidade , Neoplasias Pulmonares/secundário , Pulmão/efeitos dos fármacos , Receptores de Prostaglandina E/metabolismo , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiologia , Neoplasias da Mama/metabolismo , Celecoxib/farmacologia , Celecoxib/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Citocinas/análise , Dinoprostona/sangue , Dinoprostona/farmacologia , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Inflamação/induzido quimicamente , Pulmão/metabolismo , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Xantonas/farmacologia
7.
J Pharmacol Exp Ther ; 351(1): 134-45, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25047516

RESUMO

The excitotoxicity caused by excessive glutamate is a critical element in the neuropathology of acute and chronic brain disorders. Therefore, inhibition of glutamate release is a potentially valuable therapeutic strategy for treating these diseases. In this study, we investigated the effect of celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor that reduces the level of prostaglandin E2 (PGE2), on endogenous glutamate release in rat cerebral cortex nerve terminals (synaptosomes). Celecoxib substantially inhibited the release of glutamate induced by the K(+) channel blocker 4-aminopyridine (4-AP), and this phenomenon was prevented by chelating the extracellular Ca(2+) ions and by the vesicular transporter inhibitor bafilomycin A1. Celecoxib inhibited a 4-AP-induced increase in cytosolic-free Ca(2+) concentration, and the celecoxib-mediated inhibition of glutamate release was prevented by the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC. However, celecoxib did not alter 4-AP-mediated depolarization and Na(+) influx. In addition, this glutamate release-inhibiting effect of celecoxib was mediated through the PGE2 subtype 2 receptor (EP2) because it was not observed in the presence of butaprost (an EP2 agonist) or PF04418948 [1-(4-fluorobenzoyl)-3-[[6-methoxy-2-naphthalenyl)methyl]-3-azetidinecarboxylic acid; an EP2 antagonist]. The celecoxib effect on 4-AP-induced glutamate release was prevented by the inhibition or activation of protein kinase A (PKA), and celecoxib decreased the 4-AP-induced phosphorylation of PKA. We also determined that COX-2 and the EP2 receptor are present in presynaptic terminals because they are colocalized with synaptophysin, a presynaptic marker. These results collectively indicate that celecoxib inhibits glutamate release from nerve terminals by reducing voltage-dependent Ca(2+) entry through a signaling cascade involving EP2 and PKA.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ácido Glutâmico/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Prostaglandinas E/metabolismo , Pirazóis/farmacologia , Receptores de Prostaglandina E/metabolismo , Sulfonamidas/farmacologia , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Celecoxib , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Exocitose , Masculino , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Sódio/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
8.
Eur J Pharmacol ; 681(1-3): 55-9, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22342278

RESUMO

Arterial vascularization of the spinal cord may be mechanically or functionally altered during thoraco-abdominal surgery/intravascular procedures. Increased arterial pressure has been shown to restore spinal perfusion and function probably by increasing the blood flow through the intercostal arteries. The regulation of human intercostal artery (HICA) vascular tone is not well documented. Prostaglandin (PG)E(2) concentration is increased during inflammatory conditions and has been shown to regulate vascular tone in many preparations. In this context, the pharmacological response of HICA to PGE(2) and the characterization of the PGE(2) receptor subtypes (EP(1), EP(2), EP(3) or EP(4)) involved are of importance and that is the aim of this study. Rings of HICA were prepared from 29 patients and suspended in organ baths for isometric recording of tension. Cumulative concentration-response curves were performed in these preparations with various EP receptor agonists in the absence or presence of different receptor antagonists or inhibitors. PGE(2) induced the contraction of HICA (E(max)=7.28 ± 0.16 g; pEC(50) value=0.79 ± 0.18; n=17); contractions were also observed with the EP(3) receptor agonists, sulprostone, 17-phenyl-PGE(2), misoprostol or ONO-AE-248. In conclusion, PGE(2) induced vasoconstriction of HICA via EP(3) receptor subtypes and this result was confirmed by the use of selective EP receptor antagonists (L-826266, ONO-8713, SC-51322) and by a strong detection of EP(3) mRNA. These observations suggest that in the context of perioperative inflammation, increased PGE(2) concentrations could trigger vasoconstriction of HICA and possibly alter spinal vascularization.


Assuntos
Dinoprostona/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Prostaglandina E/metabolismo , Vasoconstritores/metabolismo , Artérias , Dinoprostona/administração & dosagem , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Humanos , Inflamação/fisiopatologia , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/administração & dosagem , Vasoconstritores/farmacologia
9.
Prostaglandins Other Lipid Mediat ; 96(1-4): 10-3, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21801847

RESUMO

Hypertension is a well established risk factor for cardiovascular diseases such as stroke and is the leading cause of chronic kidney failure. Although a number of pharmacologic agents are available for the treatment of hypertension including agents that affect the renin-angiotensin-aldosterone system (RAAS), unmet needs in the treatment of hypertension suggest that identification of novel pharmacological targets would be an important healthcare goal. One potential target is prostaglandin E(2) (PGE(2)), a potent lipid mediator with a diverse and sometimes opposing range of biological effects. PGE(2) signals through four subtypes of G-protein coupled receptors designated EP1 through EP4. PGE(2) functions primarily as a vasodepressor; under certain conditions PGE(2) administration mediates vasopressor activity. This review focuses on the current understanding of the roles of PGE(2) receptors in vascular reactivity, hypertension and end-organ damage.


Assuntos
Doenças Cardiovasculares/metabolismo , Dinoprostona/metabolismo , Hipertensão/metabolismo , Falência Renal Crônica/metabolismo , Rim/metabolismo , Receptores de Prostaglandina E/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Cálcio/metabolismo , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/fisiopatologia , AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Modelos Animais de Doenças , Humanos , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Falência Renal Crônica/tratamento farmacológico , Falência Renal Crônica/etiologia , Falência Renal Crônica/fisiopatologia , Camundongos , Camundongos Knockout , Antagonistas de Prostaglandina/farmacologia , Ratos , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Sistema Renina-Angiotensina/efeitos dos fármacos , Roedores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Vasoconstritores/farmacologia
10.
Eur J Pharmacol ; 659(1): 72-8, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21397595

RESUMO

Increased airway smooth muscle mass due to cell proliferation contributes to airway hyper-responsiveness and remodeling in patients with asthma. Prostaglandin E2 (PGE2) inhibits proliferation of airway smooth muscle cells, but the role of prostanoid EP receptor subtypes in mechanisms involved has not been fully elucidated yet. We investigated the effects of specific prostanoid EP receptor agonists on cell proliferation and intracellular Ca(2+) concentrations ([Ca(2+)]i) in human airway smooth muscle cells. Cell numbers were assessed by mitochondria-dependent reduction of 4-[3-(4-lodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1, 3-benzene disulfonate to formazan (WST-1 assay). RT-PCR data showed that human airway smooth muscle cells express EP2, EP3, and EP4 but not EP1 receptor mRNA. PGE2 (1nM-1µM) inhibited cell proliferation induced by 5% fetal bovine serum (FBS) in a concentration-dependent manner. (16S)-9-deoxy-9ß-chloro-15-deoxy-16-hydroxy-17, 17-trimethylene-19, 20-didehydro PGE2 sodium salt (ONO-AE1-259-01; EP2 receptor agonist) and 16-(3-methoxymethyl)phenyl-ω-tetranor-3,7-dithia PGE2 (ONO-AE1-329; EP4 receptor agonist) inhibited the 5% FBS-induced cell proliferation. ONO-AE1-259-01 and ONO-AE1-329 also significantly increased the cytosolic cAMP levels. In contrast, 11,15-O-dimethyl PGE2 (ONO-AE-248; EP3 receptor agonist) elicited an oscillatory increase in [Ca(2+)]i but did not affect the cell growth or cAMP levels. [(17S)-2,5-ethano-6-oxo-17,20-dimethyl PGE1] (ONO-DI-004; EP1 receptor agonist) did not affect cell growth, cAMP levels, or [Ca(2+)]i. In conclusion, PGE2 inhibits FBS-induced cell proliferation mostly via EP2 and EP4 receptor activation and subsequent cAMP elevation. The EP3 receptor agonist causes an increase in [Ca(2+)]i without affecting cell growth. There is no functional expression of the EP1 receptor. Research on prostanoid EP receptors may lead to novel therapeutic strategies for treatment of asthma.


Assuntos
Brônquios/citologia , Cálcio/metabolismo , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Receptores de Prostaglandina E/agonistas , Proliferação de Células/efeitos dos fármacos , AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Miócitos de Músculo Liso/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E/genética
11.
Br J Pharmacol ; 163(4): 826-34, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21323896

RESUMO

BACKGROUND AND PURPOSE: Human internal mammary arteries (IMA) and saphenous veins (SV) are frequently used for coronary artery bypass graft surgery. Intra- and postoperatively, the bypass grafts are exposed to inflammatory conditions, under which there is a striking increase in the synthesis of prostaglandin E(2) (PGE(2) ). In this context, the physiological response of these vascular grafts to PGE(2) is highly relevant. The aim of this study was thus to characterize the PGE(2) receptor subtypes (EP(1) , EP(2) , EP(3) or EP(4) ) involved in modulation of the vascular tone in these two vessels. EXPERIMENTAL APPROACH: Rings of IMA and SV were prepared from 48 patients. The rings were mounted in organ baths for isometric recording of tension, and a pharmacological study was performed, together with associated reverse transcriptase PCR and immunohistochemistry experiments. KEY RESULTS: PGE(2) induced contractions of IMA (E(max) = 1.43 ± 0.20 g; pEC(50) = 7.50 ± 0.10); contractions were also observed with the EP(3) receptor agonists, sulprostone, 17-phenyl-PGE(2) , misoprostol or ONO-AE-248. In contrast, PGE(2) induced relaxation of the precontracted SV (E(max) =-0.22 ± 0.02 g; pEC(50) = 7.14 ± 0.09), as did the EP(4) receptor agonist, ONO-AE1-329. These results were confirmed by the use of selective EP receptor antagonists (GW627368X, L-826266, ONO-8713, SC-51322) and by molecular biology and immunostaining. CONCLUSIONS AND IMPLICATIONS: PGE(2) induced potent and opposite effects on the human vascular segments used for grafting, namely vasoconstriction of the IMA and vasodilatation of the SV via EP(3) and EP(4) receptors respectively. These observations suggest that EP(3) and EP(4) receptors could constitute therapeutic targets to increase vascular graft patency.


Assuntos
Dinoprostona/metabolismo , Artéria Torácica Interna/efeitos dos fármacos , Artéria Torácica Interna/transplante , Veia Safena/efeitos dos fármacos , Veia Safena/transplante , Acrilamidas/farmacologia , Idoso , Ponte de Artéria Coronária/métodos , Dinoprostona/análogos & derivados , Dinoprostona/farmacologia , Feminino , Humanos , Isoindóis/farmacologia , Masculino , Artéria Torácica Interna/metabolismo , Éteres Metílicos/farmacologia , Misoprostol/farmacologia , Naftalenos/farmacologia , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Receptores de Prostaglandina E/metabolismo , Veia Safena/metabolismo , Sulfonamidas/farmacologia , Enxerto Vascular/métodos
12.
Br J Ophthalmol ; 95(6): 859-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21349943

RESUMO

BACKGROUND: Prostaglandin (PG) E(2) is produced during inflammatory responses and suppresses the production of cytokines induced by lipopolysaccharide stimulation in macrophages and dendritic cells. In this study, we examined the expression of PGE(2) receptors in human conjunctival epithelial cells and investigated whether PGE(2) downregulates polyinosine-polycytidylic acid (polyI:C)-induced cytokine production. METHODS: ELISA and quantitative reverse transcription (RT)-PCR were used to examine the effects of PGE(2) on the polyI:C-induced cytokine expressions by primary human conjunctival epithelial cells (PHCjEC). Reverse transcription-PCR was performed to examine the mRNA expression of the PGE(2) receptors EP1, -2, -3 and -4. RESULTS: PGE(2) significantly attenuated the expressions of chemokine (C-C) motif ligand (CCL) 5, chemokine (C-X-C motif) ligand (CXCL) 10, CXCL11 and interleukin (IL) 6 in PHCjECs. Human conjunctival epithelial cells exhibited expression of EP2, -3 and -4, but not of EP1. EP2 agonist significantly suppressed the polyI:C-induced the expressions of CCL5, CXCL10 and CXCL11 but not of IL-6. EP3 agonist significantly suppressed the expressions of CCL5, CXCL10, CXCL11 and IL-6. On the other hand, EP4 agonist failed to suppress the cytokine production induced by polyI:C stimulation. CONCLUSION: Our results show that PGE(2) attenuated the expression of CCL5, CXCL10 and CXCL11 via both EP2 and EP3, and that the expression of IL-6 was attenuated only by EP3.


Assuntos
Túnica Conjuntiva/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Células Epiteliais/metabolismo , Poli I-C/metabolismo , Receptores de Prostaglandina E/metabolismo , Túnica Conjuntiva/efeitos dos fármacos , AMP Cíclico/metabolismo , Citocinas/genética , Dinoprostona/genética , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/efeitos dos fármacos , Humanos , Receptores de Prostaglandina E/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Gastroenterol Hepatol ; 25 Suppl 1: S67-74, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20586869

RESUMO

BACKGROUND AND AIMS: The effects of an EP4 agonist/antagonist on the healing of lesions produced by indomethacin in the small intestine were examined in rats, especially in relation to the expression of vascular endothelial growth factor (VEGF) and angiogenesis. METHODS: Animals were given indomethacin (10 mg/kg s.c.) and killed at various time points. To impair the healing of these lesions, a small dose of indomethacin (2 mg/kg p.o.) or AE3-208 (EP4 antagonist: 3 mg/kg i.p.) was given once daily for 6 days after the ulceration was induced, with or without the co-administration of AE1-329 (EP4 agonist: 0.1 mg/kg i.p.). RESULTS: Indomethacin (10 mg/kg) caused severe damage in the small intestine, but the lesions healed rapidly decreasing to approximately one-fifth of their initial size within 7 days. The healing process was significantly impaired by indomethacin (2 mg/kg) given once daily for 6 days after the ulceration. This effect of indomethacin was mimicked by the EP4 antagonist and reversed by co-administration of the EP4 agonist. Mucosal VEGF expression was upregulated after the ulceration, reaching a peak on day 3 followed by a decrease. The changes in VEGF expression paralleled those in mucosal cyclooxygenase-2 expression, as well as prostaglandin E(2) (PGE(2)) content. Indomethacin (2 mg/kg) downregulated both VEGF expression and angiogenesis in the mucosa during the healing process, and these effects were significantly reversed by co-treatment with the EP4 agonist. CONCLUSION: The results suggest that endogenous PGE(2) promotes the healing of small intestinal lesions by stimulating angiogenesis through the upregulation of VEGF expression mediated by the activation of EP4 receptors.


Assuntos
Dinoprostona/metabolismo , Indometacina , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/metabolismo , Úlcera Péptica/metabolismo , Receptores de Prostaglandina E/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Naftalenos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Úlcera Péptica/induzido quimicamente , Úlcera Péptica/tratamento farmacológico , Úlcera Péptica/patologia , Fenilbutiratos/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4 , Índice de Gravidade de Doença , Fatores de Tempo , Regulação para Cima , Cicatrização/efeitos dos fármacos
14.
J Pharmacol Exp Ther ; 334(3): 964-72, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20558773

RESUMO

Posttransplant diabetes mellitus is a frequent complication among transplant recipients. Ligation of advanced glycation end products (AGEs) with their receptor on monocytes/macrophages plays a role in diabetes complications. The enhancement of adhesion molecule expression on monocytes/macrophages activates T cells, reducing allograft survival. In previous work, we found that toxic AGEs, AGE-2 and AGE-3, induced the expression of intracellular adhesion molecule-1, B7.1, B7.2, and CD40 on monocytes, production of interferon-gamma and tumor necrosis factor alpha, and lymphocyte proliferation during human mixed lymphocyte reaction. AGE-induced up-regulation of adhesion molecule expression was involved in cytokine production and lymphocyte proliferation. Prostaglandin E2 (PGE2) concentration-dependently inhibited the actions of AGE-2 and AGE-3. The effects of PGE2 were mimicked by an EP2 receptor agonist, ONO-AE1-259-01 (11,15-O-dimethyl PGE2), and an EP4 receptor agonist, ONO-AE1-329 [16-(3-methoxymethyl)phenyl-omega-tetranor-3,7dithia PGE1]. An EP2 receptor antagonist, AH6809 (6-isopropoxy-9-oxaxanthene-2-carboxylic acid), and an EP4 receptor antagonist, AH23848 [(4Z)-7-[(rel-1S,2S,5R)-5-((1,1'-biphenyl-4-yl)methoxy)-2-(4-morpholinyl)-3-oxocyclopentyl]-4-heptenoic acid], inhibited the actions of PGE2. The stimulation of EP2 and EP4 receptors is reported to increase cAMP levels. The effects of PGE2 were reversed by protein kinase A (PKA) inhibitors and mimicked by dibutyryl cAMP and an adenylate cyclase activator, forskolin. These results as a whole indicate that PGE2 inhibited the actions of AGE-2 and AGE-3 via EP2/EP4 receptors and the cAMP/PKA pathway.


Assuntos
Moléculas de Adesão Celular/biossíntese , Proliferação de Células/efeitos dos fármacos , Citocinas/biossíntese , Dinoprostona/farmacologia , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Produtos Finais de Glicação Avançada/biossíntese , Linfócitos/efeitos dos fármacos , Monócitos/metabolismo , Células Cultivadas , Dinoprostona/antagonistas & inibidores , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Interferon gama/biossíntese , Teste de Cultura Mista de Linfócitos , Monócitos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP4 , Fator de Necrose Tumoral alfa/biossíntese
16.
Platelets ; 21(5): 329-42, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20433310

RESUMO

The effects of prostaglandin E(2) (PGE(2)) on platelet function are believed to be the result of opposing mechanisms that lead to both enhancement and inhibition of platelet function. Enhancement of platelet function is known to be via EP3 receptors linked to G(i) and inhibition of adenylyl cyclase. However, the receptors involved in inhibition of platelet function have not been fully defined. Here we have used measurements of platelet aggregation, calcium signaling and P-selectin expression to assess platelet function induced by platelet activating factor (PAF), thrombin receptor activating peptide (TRAP-6) and the thromboxane A(2) mimetic U46619 respectively, to determine the effects of PGE(2) and of selective prostanoid receptor agonists on platelet function. Their effects on vasodilator-stimulated phosphoprotein (VASP) phosphorylation were also determined. We also assessed the ability of selective prostanoid receptor antagonists to modify the effects of PGE(2). The agonists and antagonists used were iloprost (IP agonist), ONO-DI-004 (EP1 agonist), ONO-AE1-259 (EP2 agonist), sulprostone (EP3 agonist), ONO-AE1-329 (EP4 agonist), CAY10441 (IP antagonist), ONO-8713 (EP1 antagonist), DG-041 (EP3 antagonist) and ONO-AE3-208 (EP4 antagonist). Using the agonists available to us we demonstrated that EP3, EP4 and IP receptors elicit functional responses in platelets. The EP3 receptor agonist promoted platelet aggregation, calcium signaling and P-selectin expression and this was associated with a reduction in VASP phosphorylation. Conversely agonists acting at IP and EP4 receptors inhibited platelet function and this was associated with an increase in VASP phosphorylation. The effects on platelet function and VASP phosphorylation of the selective prostanoid receptor antagonists used in conjunction with PGE(2) were consistent with PGE(2) interacting with EP3 receptors to enhance platelet function and with EP4 receptors (but not IP receptors) to inhibit platelet function. This is the first demonstration of the involvement of EP4 receptors in platelet responses to PGE(2).


Assuntos
Plaquetas/efeitos dos fármacos , Dinoprostona/farmacologia , Receptores de Prostaglandina E/sangue , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Plaquetas/metabolismo , Plaquetas/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Moléculas de Adesão Celular/sangue , AMP Cíclico/sangue , Humanos , Proteínas dos Microfilamentos/sangue , Naftalenos/farmacologia , Oligopeptídeos/farmacologia , Selectina-P/biossíntese , Fenilbutiratos/farmacologia , Fosfoproteínas/sangue , Fosforilação/efeitos dos fármacos , Fator de Ativação de Plaquetas/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP3 , Receptores de Prostaglandina E Subtipo EP4
17.
J Immunol ; 184(10): 5595-603, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20400695

RESUMO

PGI(2), which exerts its actions via its specific Gs-coupled I prostanoid receptor (IP), is known to be present in the lymph nodes, but its roles in acquired cutaneous immune responses remain unclear. To investigate the role of PGI(2)-IP signaling in cutaneous immune responses, we applied IP-deficient (Ptgir(-/-)) mice to contact hypersensitivity as a model of acquired immune response and found that Ptgir(-/-) mice exhibited a significantly decreased contact hypersensitivity response. Lymph node cells from sensitized Ptgir(-/-) mice exhibited decreased IFN-gamma production and a smaller T-bet(+) subset compared with control mice. PGI synthase and IP expression were detected in dendritic cells and T cells, respectively, by quantitative real-time PCR analysis, suggesting that PGI(2) produced by dendritic cells acts on IP in T cells. In fact, in vitro Th1 differentiation was enhanced by an IP agonist, and this enhancement was nullified by protein kinase A inhibitor. These results suggest that PGI(2)-IP signaling promotes Th1 differentiation through a cAMP-protein kinase A pathway and thereby initiates acquired cutaneous immune responses.


Assuntos
Diferenciação Celular/imunologia , Dermatite Alérgica de Contato/imunologia , Epoprostenol/metabolismo , Receptores de Prostaglandina E/fisiologia , Transdução de Sinais/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Transferência Adotiva , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Dermatite Alérgica de Contato/metabolismo , Dermatite Alérgica de Contato/patologia , Modelos Animais de Doenças , Feminino , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/metabolismo , Hipersensibilidade Tardia/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidores de Proteínas Quinases/farmacologia , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/deficiência , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP1 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologia , Células Th1/patologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Regulação para Cima/imunologia
18.
Mol Cancer Res ; 8(4): 569-77, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20353998

RESUMO

Many human cancers express elevated levels of cyclooxygenase-2 (COX-2), an enzyme responsible for the biosynthesis of prostaglandins. Available clinical data establish the protective effect of COX-2 inhibition on human cancer progression. However, despite these encouraging outcomes, the appearance of unwanted side effects remains a major hurdle for the general application of COX-2 inhibitors as effective cancer drugs. Hence, a better understanding of the molecular signals downstream of COX-2 is needed for the elucidation of drug targets that may improve cancer therapy. Here, we show that the COX-2 product prostaglandin E(2) (PGE(2)) acts on cognate receptor EP4 to promote the migration of A549 lung cancer cells. Treatment with PGE(2) enhances tyrosine kinase c-Src activation, and blockade of c-Src activity represses the PGE(2)-mediated lung cancer cell migration. PGE(2) affects target cells by activating four receptors named EP1 to EP4. Use of EP subtype-selective ligand agonists suggested that EP4 mediates prostaglandin-induced A549 lung cancer cell migration, and this conclusion was confirmed using a short hairpin RNA approach to specifically knock down EP4 expression. Proximal EP4 effectors include heterotrimeric Gs and betaArrestin proteins. Knockdown of betaArrestin1 expression with shRNA significantly impaired the PGE(2)-induced c-Src activation and cell migration. Together, these results support the idea that increased expression of the COX-2 product PGE(2) in the lung tumor microenvironment may initiate a mitogenic signaling cascade composed of EP4, betaArrestin1, and c-Src which mediates cancer cell migration. Selective targeting of EP4 with a ligand antagonist may provide an efficient approach to better manage patients with advanced lung cancer.


Assuntos
Arrestinas/metabolismo , Carcinoma/metabolismo , Movimento Celular/genética , Dinoprostona/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Prostaglandina E/metabolismo , Arrestinas/efeitos dos fármacos , Arrestinas/genética , Proteína Tirosina Quinase CSK , Carcinoma/genética , Carcinoma/fisiopatologia , Movimento Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Regulação para Baixo/genética , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Pulmonares/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/fisiopatologia , Proteínas Tirosina Quinases/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/farmacologia , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP4 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células Tumorais Cultivadas , beta-Arrestinas , Quinases da Família src
19.
Prostaglandins Other Lipid Mediat ; 92(1-4): 67-72, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20381634

RESUMO

Loss of progesterone secretion at the end of the estrous cycle is via uterine PGF(2alpha) secretion; however, uterine PGF(2alpha) is not decreased during early pregnancy in ewes to prevent luteolysis. Instead the embryo imparts resistance to PGF(2alpha)-induced luteolysis, which is via the 2-fold increase in prostaglandins E(1) and E(2) (PGE(1), PGE(2); PGE) in the endometrium during early pregnancy. Chronic intrauterine infusion of PGE(1) or PGE(2) prevents spontaneous or an estradiol-17beta, IUD, or PGF(2alpha)-induced luteolysis. Four PGE receptor subtypes (EP(1), EP(2), EP(3), and EP(4)) and an FP receptor specific for PGF(2alpha) have been identified. The objective of this experiment was to determine the effects of EP(1), EP(2), EP(3), or FP receptor agonists in vivo on luteal mRNA for LH receptors, occupied and unoccupied LH receptors, and circulating progesterone in ewes. Ewes received a single treatment of 17-phenyl-tri-Nor-PGE(2) (EP(1), EP(3)), butaprost (EP(2)), 19-(R)-OH-PGE(2) (EP(2)), sulprostone (EP(1), EP(3)), or PGF(2alpha) (FP) receptor agonists into the interstitial tissue of the ovarian vascular pedicle adjacent to the luteal-containing ovary. 17-Phenlyl-tri-Nor-PGE(2) had no effect (P> or =0.05) on any parameter analyzed. Butaprost and 19-(R)-OH-PGE(2) increased (P< or =0.05) mRNA for LH receptors, occupied and unoccupied LH receptors, and circulating progesterone. Both sulprostone and PGF(2alpha) decreased (P< or =0.05) mRNA for LH receptors, occupied and unoccupied LH receptors, and circulating progesterone. It is concluded that both EP(3) and FP receptors may be involved in luteolysis. In addition, EP(2) receptors may mediate prevention of luteolysis via regulation of luteal mRNA for LH receptors to prevent loss of occupied and unoccupied LH receptors and therefore to sustaining luteal function.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/fisiologia , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina/agonistas , Ovinos , Animais , Corpo Lúteo/anatomia & histologia , Corpo Lúteo/metabolismo , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/metabolismo , Feminino , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Progesterona/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do LH/genética
20.
Mol Pharmacol ; 77(6): 1025-36, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20335389

RESUMO

The EP1 prostanoid receptor is one of four subtypes whose cognate physiological ligand is prostaglandin-E2 (PGE(2)). It is in the family of G-protein-coupled receptors and is known to activate Ca(2+) signaling, although relatively little is known about other aspects of E-type prostanoid receptor (EP) 1 receptor signaling. In human embryonic kidney (HEK) cells expressing human EP1 receptors, we now show that PGE(2) stimulation of the EP1 receptor up-regulates the expression of hypoxia-inducible factor-1 alpha (HIF-1 alpha), which can be completely blocked by pertussis toxin, indicating coupling to G(i/o). This up-regulation of HIF-1 alpha occurs under normoxic conditions and could be inhibited with wortmannin, Akt inhibitor, and rapamycin, consistent with the activation of a phosphoinositide-3 kinase/Akt/mammalian target of rapamycin (mTOR) signaling pathway, respectively. In contrast to the hypoxia-induced up-regulation of HIF-1 alpha, which involves decreased protein degradation, the up-regulation of HIF-1 alpha by the EP1 receptor was associated with the phosphorylation of ribosomal protein S6 (rpS6), suggesting activation of the ribosomal S6 kinases and increased translation. Stimulation of endogenous EP1 receptors in human HepG2 hepatocellular carcinoma cells recapitulated the normoxic up-regulation of HIF-1 alpha observed in HEK cells, was sensitive to pertussis toxin, and involved the activation of mTOR signaling and phosphorylation of rpS6. In addition, treatment of HepG2 cells with sulprostone, an EP1-selective agonist, up-regulated the mRNA expression of vascular endothelial growth factor-C, a HIF-regulated gene. HIF-1 alpha is known to promote tumor growth and metastasis and is often up-regulated in cancer. Our findings provide a potential mechanism by which increased PGE(2) biosynthesis could up-regulate the expression of HIF-1 alpha and promote tumorigenesis.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Prostaglandina E/metabolismo , Transdução de Sinais , Regulação para Cima , Androstadienos/farmacologia , Linhagem Celular , Dinoprostona/farmacologia , Genes Reporter , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfatidilinositol 3-Quinases/farmacologia , Fosforilação , Reação em Cadeia da Polimerase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E Subtipo EP1 , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Regulação para Cima/efeitos dos fármacos , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA