Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Heart Vessels ; 38(4): 606-613, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36522555

RESUMO

Ischemic preconditioning (IPC) describes a phenomenon wherein brief ischemia of the heart induces a potent cardioprotective mechanism against succeeding ischemic insult. Cyclooxygenase-2 (COX-2), a rate-limiting enzyme in prostanoid biosynthesis, is upregulated in the ischemic heart and contributes to IPC. Prostaglandin E2 (PGE2) protects the heart from ischemia-reperfusion (I/R) injury via its receptor subtype EP4. We sought to clarify the role of the PGE2/EP4 system in the late phase of IPC. Mice were subjected to four IPC treatment cycles, consisting of 5 min of occlusion of the left anterior descending coronary artery (LAD). We found that COX-2 mRNA was significantly upregulated in wild-type hearts at 6 h after IPC treatment. Cardiac PGE2 levels at 24 h after IPC treatment were significantly increased in both wild-type mice and mice lacking EP4 (EP4-/-). At 24 h after IPC treatment, I/R injury was induced by 30 min of LAD occlusion followed by 2 h of reperfusion and the cardiac infarct size was determined. The infarct size was significantly reduced by IPC treatment in wild-type mice; a reduction was not observed in EP4-/- mice. AE1-329, an EP4 agonist, significantly reduced infarct size and significantly ameliorated deterioration of cardiac function in wild-type mice subjected to I/R without IPC treatment. Furthermore, AE1-329 significantly enhanced the I/R-induced activation of Akt, a pro-survival kinase. We demonstrated that the PGE2/EP4 system in the heart plays a critical role in the late phase of IPC, partly by augmenting Akt-mediated signaling. These findings clarify the mechanism of IPC and may contribute to the development of therapeutic strategies for ischemic heart disease.


Assuntos
Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt/uso terapêutico , Ciclo-Oxigenase 2 , Prostaglandinas/uso terapêutico
2.
Mol Reprod Dev ; 86(7): 786-797, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31087493

RESUMO

Cyclooxygenase 2 (COX-2) is an inducible rate-limiting enzyme for prostanoid production. Because COX-2 represents one of the inducible genes in mouse mesenchymal stem cells upon differentiation into Leydig cells, we investigated COX-2 expression and production of prostaglandin (PG) in Leydig cells. Although COX-2 was undetectable in mouse testis, it was transiently induced in Leydig cells by human chorionic gonadotropin (hCG) administration. Consistent with the finding that Leydig cells expressed aldo-keto reductase 1B7 (PGF synthase) and PGE synthase 2, induction of COX-2 by hCG caused a marked increase in testicular PGF 2α and PGE 2 levels. Using mouse Leydig cell tumor-derived MA-10 cells as a model, it was indicated by reporter assays and electron mobility shift assays that transcription of the COX-2 gene was activated by CCAAT/enhancer-binding protein ß (C/EBPß) with cAMP-stimulation. C/EBPß expression was induced by cAMP-stimulation, whereas expression of C/EBP homolog protein (CHOP) was robustly downregulated. Transfection of CHOP expression plasmid inhibited cAMP-induced COX-2 promoter activity. In addition, CHOP reduced constitutive COX-2 expression in other mouse Leydig cell tumor-derived TM3 cells. These results indicate that COX-2 is induced in Leydig cells by activation of C/EBPß via reduction of CHOP expression upon gonadotropin-stimulation to produce PGF 2α and PGE 2 .


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Gonadotropina Coriônica/farmacologia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Células Intersticiais do Testículo/metabolismo , Substâncias para o Controle da Reprodução/farmacologia , Animais , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Transcrição Gênica , Transfecção
3.
Thromb Haemost ; 119(8): 1311-1320, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31129913

RESUMO

Platelets play an important role in both physiological hemostasis and pathological thrombosis. Thromboxane (TX) A2 and prostaglandin (PG) I2 are well known as a potent stimulator and an inhibitor of platelet function, respectively. Recently, PGE2 has also been reported to regulate platelet function via PGE2 receptor subtypes. However, the effect of PGF2α on platelet function remains to be determined. The aim of the present study was to clarify the effect of PGF2α on murine platelet function both in vitro and in vivo. Platelets prepared from wild-type mice (WT platelets) expressed several types of prostanoid receptors, including the PGE2 receptor subtype EP3 and the TXA2 receptor TP, while expression of the PGF2α receptor FP was not detected. In WT platelets, PGF2α potentiated adenosine diphosphate-induced aggregation in a concentration-dependent manner, while PGF2α alone did not induce aggregation. In platelets prepared from mice lacking FP, however, PGF2α-induced potentiation was not significantly different from that in WT platelets. Interestingly, the potentiation was significantly blunted in platelets lacking EP3 or TP and disappeared completely in platelets lacking both EP3 and TP. Accordingly, PGF2α decreased the cyclic adenosine monophosphate level via EP3 and increased the inositol triphosphate level via TP in WT platelets. Intravenously administered PGF2α significantly shortened the bleeding time and aggravated arachidonic acid-induced acute thromboembolism in WT mice, suggesting that PGF2α works as a platelet stimulator also in vivo. In conclusion, PGF2α potentiates platelet aggregation in vitro via EP3 and TP but not FP. Accordingly, PGF2α facilitates hemostasis and thromboembolism in vivo.


Assuntos
Ativação Plaquetária , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Animais , Tempo de Sangramento , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Dinoprosta , Feminino , Hemostasia , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Adesividade Plaquetária , Agregação Plaquetária , Tromboembolia/sangue
4.
FASEB J ; 32(5): 2354-2365, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29247122

RESUMO

Nonalcoholic steatohepatitis (NASH) is a hepatic manifestation of metabolic syndrome. Although the prostaglandin (PG)I2 receptor IP is expressed broadly in the liver, the role of PGI2-IP signaling in the development of NASH remains to be determined. Here, we investigated the role of the PGI2-IP system in the development of steatohepatitis using mice lacking the PGI2 receptor IP [IP-knockout (IP-KO) mice] and beraprost (BPS), a specific IP agonist. IP-KO and wild-type (WT) mice were fed a methionine- and choline-deficient diet (MCDD) for 2, 5, or 10 wk. BPS was administered orally to mice every day during the experimental periods. The effect of BPS on the expression of chemokine and inflammatory cytokines was examined also in cultured Kupffer cells. WT mice fed MCDD developed steatohepatitis at 10 wk. IP-KO mice developed steatohepatitis at 5 wk with augmented histologic derangements accompanied by increased hepatic monocyte chemoattractant protein-1 (MCP-1) and TNF-α concentrations. After 10 wk of MCDD, IP-KO mice had greater hepatic iron deposition with prominent oxidative stress, resulting in hepatocyte damage. In WT mice, BPS improved histologic and biochemical parameters of steatohepatitis, accompanied by reduced hepatic concentration of MCP-1 and TNF-α. Accordingly, BPS suppressed the LPS-stimulated Mcp-1 and Tnf-α mRNA expression in cultured Kupffer cells prepared from WT mice. PGI2-IP signaling plays a crucial role in the development and progression of steatohepatitis by modulating the inflammatory response, leading to augmented oxidative stress. We suggest that the PGI2-IP system is an attractive therapeutic target for treating patients with NASH.-Kumei, S., Yuhki, K.-I., Kojima, F., Kashiwagi, H., Imamichi, Y., Okumura, T., Narumiya, S., Ushikubi, F. Prostaglandin I2 suppresses the development of diet-induced nonalcoholic steatohepatitis in mice.


Assuntos
Epoprostenol/farmacologia , Alimentos Formulados/efeitos adversos , Hepatócitos/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Animais , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Epoprostenol/análogos & derivados , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/patologia , Células de Kupffer/patologia , Fígado/patologia , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Receptores de Epoprostenol/agonistas , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
5.
Sci Rep ; 7(1): 8374, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28827713

RESUMO

Diethylstilbestrol (DES), a strong estrogenic compound, is well-known to affect the reproductive system. In this study, we investigated the effects of DES administration on gonadotropin levels and ovarian steroidogenesis in prepubertal rats. DES treatment acutely reduced serum LH levels, followed by a reduction in the expression of various steroidogenesis-related genes in theca cells. Serum FSH levels were almost unaffected by DES-treatment, even though Cyp19a1 expression was markedly reduced. Serum progesterone, testosterone and estradiol levels were also declined at this time. LH levels recovered from 12 h after DES-treatment and gradually increased until 96 h with a reduction of ERα expression observed in the pituitary. Steroidogenesis-related genes were also up-regulated during this time, except for Cyp17a1 and Cyp19a1. Consistent with observed gene expression pattern, serum testosterone and estradiol concentrations were maintained at lower levels, even though progesterone levels recovered. DES-treatment induced the inducible nitric oxide synthase (iNOS) in granulosa cells, and a nitric oxide generator markedly repressed Cyp19a1 expression in cultured granulosa cells. These results indicate that DES inhibits thecal androgen production via suppression of pituitary LH secretion and ovarian Cyp17a1 expression. In addition, DES represses Cyp19a1 expression by inducing iNOS gene expression for continuous inhibition of estrogen production in granulosa cells.


Assuntos
Androgênios/sangue , Aromatase/genética , Dietilestilbestrol/administração & dosagem , Estrogênios não Esteroides/administração & dosagem , Estrogênios/sangue , Células da Granulosa/efeitos dos fármacos , Ovário/efeitos dos fármacos , Células Tecais/efeitos dos fármacos , Animais , Feminino , Perfilação da Expressão Gênica , Gonadotropinas/sangue , Células da Granulosa/metabolismo , Ovário/metabolismo , Ratos , Esteroide 17-alfa-Hidroxilase/análise , Esteroide 17-alfa-Hidroxilase/genética , Células Tecais/metabolismo
6.
TH Open ; 1(2): e122-e129, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31249917

RESUMO

The results of studies that were performed to determine whether cigarette smoking affects platelet function have been controversial, and the effects of nicotine- and tar-free cigarette smoke extract (CSE) on platelet function remain to be determined. The aim of this study was to determine the effect of CSE on platelet aggregation and to clarify the mechanism by which CSE affects platelet function. CSE inhibited murine platelet aggregation induced by 9,11-dideoxy-9α,11α-methanoepoxy-prosta-5Z,13E-dien-1-oic acid (U-46619), a thromboxane (TX) A 2 receptor agonist, and that induced by collagen with respective IC 50 values of 1.05 ± 0.14% and 1.34 ± 0.19%. A similar inhibitory action of CSE was also observed in human platelets. CSE inhibited arachidonic acid-induced TXA 2 production in murine platelets with an IC 50 value of 7.32 ± 2.00%. Accordingly, the inhibitory effect of CSE on collagen-induced aggregation was significantly blunted in platelets lacking the TXA 2 receptor compared with the inhibitory effect in control platelets. In contrast, the antiplatelet effects of CSE in platelets lacking each inhibitory prostanoid receptor, prostaglandin (PG) I 2 receptor and PGE 2 receptor subtypes EP 2 and EP 4 , were not significantly different from the effects in respective control platelets. Among the enzymes responsible for TXA 2 production in platelets, the activity of cyclooxygenase (COX)-1 was inhibited by CSE with an IC 50 value of 1.07 ± 0.15% in an uncompetitive manner. In contrast, the activity of TX synthase was enhanced by CSE. The results indicate that CSE inhibits COX-1 activity and thereby decreases TXA 2 production in platelets, leading to inhibition of platelet aggregation.

7.
J Clin Endocrinol Metab ; 101(10): 3582-3591, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27428878

RESUMO

CONTEXT: 11-ketotestosterone (11-KT) is a novel class of active androgen. However, the detail of its synthesis remains unknown for humans. OBJECTIVE: The objective of this study was to clarify the production and properties of 11-KT in human. Design, Participants, and Methods: Expression of cytochrome P450 and 11ß-hydroxysteroid dehydrogenase types 1 and 2 (key enzymes involved in the synthesis of 11-KT) were investigated in human gonads. The production of 11-KT was investigated in Leydig cells. Plasma concentrations of testosterone and 11-KT were measured in 10 women and 10 men of reproductive age. Investigation of its properties was performed using breast cancer-derived MCF-7 cells. RESULTS: Cytochrome P450 and 11ß-hydroxysteroid dehydrogenase types 1 and 2 were detected in Leydig cells and theca cells. Leydig cells produced 11-KT, and relatively high levels of plasma 11-KT were measured in both men and women. There was no sexual dimorphism in the plasma levels of 11-KT, even though testosterone levels were more than 20 times higher in men than in women. It is noteworthy that the levels of testosterone and 11-KT were similar in women. In a luciferase reporter system, 11-KT activated human androgen receptor-mediated transactivation. Conversely, 11-KT did not activate estrogen receptor-mediated transactivation in aromatase-expressed MCF-7 cells, whereas testosterone did following conversion to estrogen. 11-KT did not affect the estrogen/estrogen receptor -mediated cell proliferation of MCF-7 cells. Furthermore, it significantly inhibited cell proliferation when androgen receptor was transfected into MCF-7 cells. CONCLUSIONS: The current study indicates that 11-KT is produced in the gonads and represents a major androgen in human. It can potentially serve as a nonaromatizable androgen.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Células Intersticiais do Testículo/metabolismo , Testosterona/análogos & derivados , Células Tecais/metabolismo , Feminino , Humanos , Masculino , Testosterona/metabolismo , Células Tumorais Cultivadas
8.
J Pharmacol Exp Ther ; 353(2): 269-78, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740898

RESUMO

ONO-1301 [(E)-[5-[2-[1-phenyl-1-(3-pyridyl)methylidene-aminooxy]ethyl]-7,8-dihydronaphthalene-1-yloxy]acetic acid] is a novel prostaglandin (PG) I2 mimetic with inhibitory activity on the thromboxane (TX) A2 synthase. Interestingly, ONO-1301 retains its inhibitory effect on platelet aggregation after repeated administration, while beraprost, a representative agonist for the PGI2 receptor (IP), loses its inhibitory effect after repeated administration. In the present study, we intended to clarify the mechanism by which ONO-1301 escapes desensitization of an antiplatelet effect. In platelets prepared from wild-type mice, ONO-1301 inhibited collagen-induced aggregation and stimulated cAMP production in an IP-dependent manner. In addition, ONO-1301 inhibited arachidonic acid-induced TXA2 production in platelets lacking IP. Despite the decrease in stimulatory action on cAMP production, the antiplatelet effect of ONO-1301 hardly changed after repeated administration for 10 days in wild-type mice. Noteworthy, beraprost could retain its antiplatelet effect after repeated administration in combination with a low dose of ozagrel, a TXA2 synthase inhibitor. Therefore, we hypothesized that chronic IP stimulation by beraprost induces an increase in TXA2 production, leading to reduction in the antiplatelet effect. As expected, repeated administration of beraprost increased the plasma and urinary levels of a TXA2 metabolite, while ONO-1301 did not increase them significantly. In addition, beraprost could retain the ability to inhibit platelet aggregation after repeated administration in mice lacking the TXA2 receptor (TP). These results indicate that TP-mediated signaling participates in platelet desensitization against IP agonists and that simultaneous inhibition of TXA2 production confers resistance against desensitization on IP agonists.


Assuntos
Inibidores da Agregação Plaquetária/farmacologia , Piridinas/farmacologia , Tromboxano A2/biossíntese , Administração Oral , Animais , Pressão Sanguínea/efeitos dos fármacos , AMP Cíclico/biossíntese , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Masculino , Camundongos , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/administração & dosagem , Piridinas/administração & dosagem , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tromboxano A2/metabolismo , Tromboxano-A Sintase/antagonistas & inibidores
9.
J Gastroenterol ; 48(8): 885-96, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23242454

RESUMO

BACKGROUND: Muscarinic acetylcholine receptors (mAChRs) are major regulators of gut epithelial functions. However, the precise subtype composition has not been clarified. METHODS: We characterized the pharmacological profile of mAChRs on mouse colonic crypts, employing [(3)H]-N-methyl scopolamine chloride as a radioligand and several subtype-selective chemicals, and the functional aspect by measuring short-circuit current (I sc) in Ussing chambers and by evaluating MAP kinase phosphorylation in mouse colonic mucosal sheets. RESULTS: The mAChRs were detected on the crypts (K d = 163.2 ± 32.3 pM, B max = 47.3 ± 2.6 fmol/mg of total cell protein). Muscarinic toxin 7 (MT-7, M1 subtype selective) gave a displacement curve with high affinity, but there was a part insensitive to MT-7 (18.8 ± 0.4 % of the total specific binding). The MT-7-insensitive component was displaced completely by darifenacin (M3 selective) with high affinity. ACh induced an increase in I sc, which was significantly enhanced by MT-7 but was completely inhibited by darifenacin or atropine. Colitis induction resulted in a significant decrease in the density of mAChRs, which occurred mainly in the MT-7-sensitive component (M1 subtype). Immunological experiments exhibited a reduction of M1 but not of M3 signal after colitis induction. Muscarinic stimulation induced an increase in MAP kinase phosphorylation, which was completely suppressed by MT-7 and was attenuated by inflammation, in mouse colonic epithelium. CONCLUSIONS: These results suggest that mAChRs in mouse colonic epithelial cells consist of two subtypes, M1 (80 %) and M3 (20 %). The major M1 subtype was likely to regulate epithelial chloride secretion negatively and was susceptible to inflammation and may be relevant to inflammatory gut dysfunction.


Assuntos
Colo/metabolismo , Mucosa Intestinal/metabolismo , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/metabolismo , Animais , Atropina/farmacologia , Benzofuranos/metabolismo , Colite/fisiopatologia , Colo/citologia , Colo/fisiopatologia , Venenos Elapídicos/metabolismo , Células Epiteliais/metabolismo , Inflamação/fisiopatologia , Mucosa Intestinal/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , N-Metilescopolamina/metabolismo , Parassimpatolíticos/metabolismo , Pirrolidinas/metabolismo , Ensaio Radioligante
10.
Circ J ; 77(4): 1053-62, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23257313

RESUMO

BACKGROUND: Prostacyclin (PGI2) enhances angiogenesis, especially in cooperation with bone marrow (BM)-derived endothelial progenitor cells (EPCs). However, the mechanisms of PGI2 in EPC-mediated angiogenesis in vivo remain unclear. The purpose of this study was to clarify the role of PGI2 in EPC-mediated angiogenesis using BM-specific IP deletion mice. METHODS AND RESULTS: Hind limb ischemia (HLI) was induced in wild-type (WT) mice transplanted with IP-deleted BM (WT/BM(IP(-/-)). Recovery of blood flow (RBF) in WT/BM(IP(-/-)) was impaired for 28 days after HLI, whereas RBF in IP(-/-)/BM(WT) was attenuated for up to 7 days compared with WT/BM(WT). The impaired RBF in WT/BM(IP(-/-)) was completely recovered by intramuscular injection of WT EPCs but not IP(-/-) EPCs. The impaired effects of IP(-/-) EPCs were in accordance with reduced formation of capillary and arterioles in ischemic muscle. An ex vivo aortic ring assay revealed that microvessel formation was enhanced by accumulation/adhesion of EPCs to perivascular sites as pericytes. IP(-/-)EPCs, in which expression of integrins was decreased, had impaired production of angiogenic cytokines, adhesion to neovessels and their angiogenic effects. The small-interfering RNA (siRNA)-mediated knockdown of integrin ß1 in WT EPCs attenuated adhesion to microvessels and their in vivo and in vitro angiogenic effects. CONCLUSIONS: PGI2 may induce persistent angiogenic effects in HLI through adhesion of EPCs to perivascular sites of neovessels via integrins in addition to paracrine effects.


Assuntos
Transplante de Medula Óssea , Células Endoteliais/metabolismo , Epoprostenol/metabolismo , Isquemia/terapia , Microcirculação , Neovascularização Fisiológica , Células-Tronco/metabolismo , Animais , Adesão Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Epoprostenol/genética , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Membro Posterior/patologia , Isquemia/genética , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pericitos/metabolismo , Pericitos/patologia , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Células-Tronco/patologia
11.
Mol Cell Endocrinol ; 362(1-2): 176-82, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22713853

RESUMO

Aromatase-deficient (ArKO) mice are totally anovulatory due to insufficient estrogen production. However, sequential administrations of high doses of 17ß-estradiol (E2) and gonadotropins were found to induce ovulation in these mice. Here, we examined how the ovulatory stimulation for ArKO mice alters the expressions of genes related to prostaglandin (PG) E(2) metabolism and ovarian contents of PGE(2), as PGE(2) is one of the critical mediators of ovulatory induction. The ovulatory stimulation significantly increased mRNA expressions of prostaglandin-endoperoxide synthase 2, PGE(2) receptor type 4 and sulfotransferase family 1E, member 1, in preovulatory ArKO ovaries. In contrast, it suppressed the mRNA expression of 15-hydroxyprostaglandin dehydrogenase. Furthermore, significant elevation in the PGE(2) contents was detected in the preovulatory ovaries of ArKO mice after stimulation with E2 plus ovulatory doses of gonadotropins. Thus, these analyses demonstrate a requirement of E2 for the preovulatory enhancement of PGE(2) synthesis, leading to future success in ovulation.


Assuntos
Dinoprostona/biossíntese , Estradiol/fisiologia , Ovário/fisiologia , Ovulação , Transtornos 46, XX do Desenvolvimento Sexual/genética , Animais , Aromatase/deficiência , Aromatase/genética , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Feminino , Ginecomastia/genética , Infertilidade Masculina/genética , Erros Inatos do Metabolismo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovário/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Transcrição Gênica
12.
Kidney Int ; 82(2): 158-71, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22513820

RESUMO

Inflammatory responses in the kidney lead to tubulointerstitial fibrosis, a common feature of chronic kidney diseases. Here we examined the role of prostaglandin E(2) (PGE(2)) in the development of tubulointerstitial fibrosis. In the kidneys of wild-type mice, unilateral ureteral obstruction leads to progressive tubulointerstitial fibrosis with macrophage infiltration and myofibroblast proliferation. This was accompanied by an upregulation of COX-2 and PGE(2) receptor subtype EP(4) mRNAs. In the kidneys of EP(4) gene knockout mice, however, obstruction-induced histological alterations were significantly augmented. In contrast, an EP(4)-specific agonist significantly attenuated these alterations in the kidneys of wild-type mice. The mRNAs for macrophage chemokines and profibrotic growth factors were upregulated in the kidneys of wild-type mice after ureteral obstruction. This was significantly augmented in the kidneys of EP(4)-knockout mice and suppressed by the EP(4) agonist but only in the kidneys of wild-type mice. Notably, COX-2 and MCP-1 proteins, as well as EP(4) mRNA, were localized in renal tubular epithelial cells after ureteral obstruction. In cultured renal fibroblasts, another EP(4)-specific agonist significantly inhibited PDGF-induced proliferation and profibrotic connective tissue growth factor production. Hence, an endogenous PGE(2)-EP(4) system in the tubular epithelium limits the development of tubulointerstitial fibrosis by suppressing inflammatory responses.


Assuntos
Dinoprostona/metabolismo , Células Epiteliais/metabolismo , Nefropatias/prevenção & controle , Túbulos Renais/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Fibrose , Ácido Fólico , Regulação da Expressão Gênica , Heptanoatos/farmacologia , Nefropatias/etiologia , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miofibroblastos/metabolismo , Miofibroblastos/patologia , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E Subtipo EP4/agonistas , Receptores de Prostaglandina E Subtipo EP4/deficiência , Receptores de Prostaglandina E Subtipo EP4/genética , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/complicações
13.
Thromb Haemost ; 104(4): 796-803, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20664890

RESUMO

The effect of selective activation of platelet prostaglandin (PG) E2 receptor subtype EP2 or EP4 on platelet aggregation remains to be determined. In platelets prepared from wild-type mice (WT platelets), high concentrations of PGE2 inhibited platelet aggregation induced by U-46619, a thromboxane receptor agonist. However, there was no significant change in the inhibitory effect of PGE2 on platelets lacking EP2 (EP2-/- platelets) and EP4 (EP4-/- platelets) compared with the inhibitory effect on WT platelets. On the other hand, AE1-259 and AE1-329, agonists for EP2 and EP4, respectively, potently inhibited U-46619 -induced aggregation with respective IC50 values of 590 ± 14 and 100 ± 4.9 nM in WT platelets, while the inhibition was significantly blunted in EP2-/- and EP4-/- platelets. In human platelets, AE1-259 and AE1-329 inhibited U-46619-induced aggregation with respective IC50 values of 640 ± 16 and 2.3 ± 0.3 nM. Notably, the inhibitory potency of AE1-329 in human platelets was much higher than that in murine platelets, while such a difference was not observed in the inhibitory potency of AE1-259. AE1-329 also inhibited adenosine diphosphate-induced platelet aggregation, and the inhibition was almost completely blocked by AE3-208, an EP4 antagonist. In addition, AE1-329 increased intracellular cAMP concentrations in a concentration- and EP4-dependent manner in human platelets. These results indicate that selective activation of EP2 or EP4 can inhibit platelet aggregation and that EP4 agonists are particularly promising as novel anti-platelet agents.


Assuntos
Plaquetas/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP2/agonistas , Receptores de Prostaglandina E Subtipo EP4/agonistas , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Células Cultivadas , Dinoprostona/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Prostaglandina E Subtipo EP2/genética , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Tromboxanos/agonistas
14.
Arterioscler Thromb Vasc Biol ; 30(3): 464-70, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20007911

RESUMO

OBJECTIVE: Endothelial progenitor cells (EPCs) play an important role in the self-healing of a vascular injury by participating in the reendothelialization that limits vascular remodeling. We evaluated whether prostaglandin I(2) plays a role in the regulation of the function of EPCs to limit vascular remodeling. METHODS AND RESULTS: EPCs (Lin(-)cKit(+)Flk-1(+) cells) were isolated from the bone marrow (BM) of wild-type (WT) mice or mice lacking the prostaglandin I(2) receptor IP (IP(-/-) mice). Reverse transcription-polymerase chain reaction analysis showed that EPCs among BM cells specifically express IP. The cellular properties of EPCs, adhesion, migration, and proliferation on fibronectin were significantly attenuated in IP-deficient EPCs compared with WT EPCs. In contrast, IP agonists facilitated these functions in WT EPCs, but not in IP-deficient EPCs. The specific deletion of IP in BM cells, which was performed by transplanting BM cells of IP(-/-) mice to WT mice, accelerated wire injury-mediated neointimal hyperplasia in the femoral artery. Notably, transfused WT EPCs, but not IP-deficient EPCs, were recruited to the injured vessels, participated in reendothelialization, and efficiently rescued the accelerated vascular remodeling. CONCLUSIONS: These findings clearly indicate that the prostaglandin I(2)-IP system is essential for EPCs to accomplish their function and plays a critical role in the regulation of vascular remodeling.


Assuntos
Endotélio Vascular/metabolismo , Epoprostenol/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/fisiologia , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Endotélio Vascular/patologia , Fibronectinas/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Knockout , Modelos Animais , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Transdução de Sinais/fisiologia , Túnica Íntima/lesões , Túnica Íntima/metabolismo , Túnica Íntima/patologia
15.
Hypertens Res ; 29(11): 905-13, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17345791

RESUMO

Reactive oxygen species (ROS) enhance myocardial ischemia-reperfusion (I/R) injury. Ebselen, a seleno-organic glutathione peroxidase (GPx) mimetic, has a protective effect against tissue injury induced by ROS. However, the cardio-protective effect of orally administered ebselen has never been investigated in cardiac I/R injury. We investigated the effects and mechanisms of orally administered ebselen on experimental myocardial infarction. Isolated perfused rabbit hearts underwent 30 min of global ischemia and 60 min of reperfusion, with or without oral administration of ebselen 24 h before I/R, with or without enhanced oxidative stress by H202 infusion for the first 1 min of reperfusion. The recovery of left ventricular developed pressure (LVDP) was significantly improved, and the myocardial infarct size was significantly reduced by ebselen. The recovery of LVDP and the myocardial infarct size were markedly aggravated by H202 infusion. These enhancements by H202 were dose-dependently suppressed by ebselen, along with a reduction in myocardial 8-hydroxydeoxyguanosine levels, a marker for oxidative DNA damage. The myocardial reduced glutathione (GSH) level was preserved by ebselen. Ebselen markedly enhanced myocardial heat shock protein (HSP) 72 expression. The cardioprotective effect of ebselen-induced HSP72 was confirmed by MTT assay in isolated cardiomyocytes using KNK437, a novel HSP inhibitor. In conclusion, an oral administration of ebselen 24 h before I/R provided excellent cardioprotective effects, at least in part through HSP72 induction and GSH preservation.


Assuntos
Antioxidantes/uso terapêutico , Azóis/uso terapêutico , Proteínas de Choque Térmico HSP72/efeitos dos fármacos , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Compostos Organosselênicos/uso terapêutico , 8-Hidroxi-2'-Desoxiguanosina , Administração Oral , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Azóis/administração & dosagem , Azóis/farmacologia , Compostos Benzidrílicos/farmacologia , Células Cultivadas , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Expressão Gênica/efeitos dos fármacos , Glutationa/efeitos dos fármacos , Proteínas de Choque Térmico HSP72/antagonistas & inibidores , Peróxido de Hidrogênio/farmacologia , Técnicas In Vitro , Isoindóis , Masculino , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Compostos Organosselênicos/administração & dosagem , Compostos Organosselênicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Pirrolidinonas/farmacologia , Coelhos , Ratos , Ratos Sprague-Dawley , Selênio/metabolismo , Sais de Tetrazólio , Tiazóis
16.
Circulation ; 112(1): 84-92, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15983244

RESUMO

BACKGROUND: In the heart, the expressions of several types of prostanoid receptors have been reported. However, their roles in cardiac hypertrophy in vivo remain unknown. We intended to clarify the roles of these receptors in pressure overload-induced cardiac hypertrophy using mice lacking each of their receptors. METHODS AND RESULTS: We used a model of pressure overload-induced cardiac hypertrophy produced by banding of the transverse aorta in female mice. In wild-type mice subjected to the banding, cardiac hypertrophy developed during the observation period of 8 weeks. In mice lacking the prostaglandin (PG) I2 receptor (IP(-/-)), however, cardiac hypertrophy and cardiomyocyte hypertrophy were significantly greater than in wild-type mice at 2 and 4 weeks but not at 8 weeks, whereas there was no such augmentation in mice lacking the prostanoid receptors other than IP. In addition, cardiac fibrosis observed in wild-type hearts was augmented in IP(-/-) hearts, which persisted for up to 8 weeks. In IP(-/-) hearts, the expression level of mRNA for atrial natriuretic peptide, a representative marker of cardiac hypertrophy, was significantly higher than in wild-type hearts. In vitro, cicaprost, an IP agonist, reduced platelet-derived growth factor-induced proliferation of wild-type noncardiomyocytes, although it could not inhibit cardiotrophin-1-induced hypertrophy of cardiomyocytes. Accordingly, cicaprost increased cAMP concentration efficiently in noncardiomyocytes. CONCLUSIONS: IP plays a suppressive role in the development of pressure overload-induced cardiac hypertrophy via the inhibition of both cardiomyocyte hypertrophy and cardiac fibrosis. Both effects have been suggested as originating from the action on noncardiomyocytes rather than cardiomyocytes.


Assuntos
Cardiomegalia/etiologia , Hipertensão/complicações , Receptores de Epoprostenol/fisiologia , Animais , Biomarcadores/análise , Cardiomegalia/patologia , Crescimento Celular , AMP Cíclico/sangue , Modelos Animais de Doenças , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Feminino , Fibrose , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , RNA Mensageiro/análise , Receptores de Epoprostenol/deficiência , Receptores de Epoprostenol/genética
17.
Circulation ; 109(20): 2462-8, 2004 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-15123528

RESUMO

BACKGROUND: In the heart with acute myocardial infarction, production of prostaglandin (PG) E2 increases significantly. In addition, several subtypes of PGE2 receptors (EPs) have been reported to be expressed in the heart. The role of PGE2 in cardiac ischemia-reperfusion (I/R) injury, however, remains unknown. We intended to clarify the role of PGE2 via EP4, an EP subtype, in I/R injury using mice lacking EP4 (EP4-/- mice). METHODS AND RESULTS: In murine cardiac ventricle, competitive reverse transcription-polymerase chain reaction revealed the highest expression level of EP4 mRNA among EP mRNAs. EP4-/- mice had larger infarct size than wild-type mice in a model of I/R; the left anterior descending coronary artery was occluded for 1 hour, followed by 24 hours of reperfusion. In addition, isolated EP4-/- hearts perfused according to the Langendorff technique had greater functional and biochemical derangements in response to I/R than wild-type hearts. In vitro, AE1-329, an EP4 agonist, raised cAMP concentration remarkably in noncardiomyocytes, whereas the action was weak in cardiomyocytes. When 4819-CD, another EP4 agonist, was administered 1 hour before coronary occlusion, it reduced infarct size significantly in wild-type mice. Notably, a similar cardioprotective effect was observed even when it was administered 50 minutes after coronary occlusion. CONCLUSIONS: Both endogenous PGE2 and an exogenous EP4 agonist protect the heart from I/R injury via EP4. The potent cardioprotective effects of 4819-CD suggest that the compound would be useful for treatment of acute myocardial infarction.


Assuntos
Dinoprostona/fisiologia , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Receptores de Prostaglandina E/agonistas , Animais , Cardiotônicos/uso terapêutico , AMP Cíclico/metabolismo , Ventrículos do Coração/metabolismo , Masculino , Éteres Metílicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP4
18.
Proc Natl Acad Sci U S A ; 100(7): 4132-7, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12642666

RESUMO

Sickness evokes various neural responses, one of which is activation of the hypothalamo-pituitary-adrenal (HPA) axis. This response can be induced experimentally by injection of bacterial lipopolysaccharide (LPS) or inflammatory cytokines such as IL-1. Although prostaglandins (PGs) long have been implicated in LPS-induced HPA axis activation, the mechanism downstream of PGs remained unsettled. By using mice lacking each of the four PGE receptors (EP1-EP4) and an EP1-selective antagonist, ONO-8713, we showed that both EP1 and EP3 are required for adrenocorticotropic hormone release in response to LPS. Analysis of c-Fos expression as a marker for neuronal activity indicated that both EP1 and EP3 contribute to activation of neurons in the paraventricular nucleus of the hypothalamus (PVN). This analysis also revealed that EP1, but not EP3, is involved in LPS-induced activation of the central nucleus of the amygdala. EP1 immunostaining in the PVN revealed its localization at synapses on corticotropin-releasing hormone-containing neurons. These findings suggest that EP1- and EP3-mediated neuronal pathways converge at corticotropin-releasing hormone-containing neurons in the PVN to induce HPA axis activation upon sickness.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Endotoxinas/toxicidade , Lipopolissacarídeos/toxicidade , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptores de Prostaglandina E/deficiência , Receptores de Prostaglandina E/fisiologia , Sinapses/fisiologia , Animais , Infecções Bacterianas , Hormônio Liberador da Corticotropina/análise , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Regulação da Expressão Gênica/efeitos dos fármacos , Genes fos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiologia , Isoenzimas/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/fisiologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP1 , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP3 , Receptores de Prostaglandina E Subtipo EP4 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA