Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Heart Vessels ; 38(4): 606-613, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36522555

ABSTRACT

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.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Infarction , Myocardial Reperfusion Injury , Mice , Animals , Proto-Oncogene Proteins c-akt/therapeutic use , Cyclooxygenase 2 , Prostaglandins/therapeutic use
2.
Inflamm Regen ; 42(1): 1, 2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34983695

ABSTRACT

BACKGROUND: Microsomal prostaglandin E synthase-1 (mPGES-1) is a key enzyme that acts downstream of cyclooxygenase and plays a major role in inflammation by converting prostaglandin (PG) H2 to PGE2. The present study investigated the effect of genetic deletion of mPGES-1 on the development of immunologic responses to experimental colitis induced by dextran sodium sulfate (DSS), a well-established model of inflammatory bowel disease (IBD). METHODS: Colitis was induced in mice lacking mPGES-1 (mPGES-1-/- mice) and wild-type (WT) mice by administering DSS for 7 days. Colitis was assessed by body weight loss, diarrhea, fecal bleeding, and histological features. The colonic expression of mPGES-1 was determined by real-time PCR, western blotting, and immunohistochemistry. The impact of mPGES-1 deficiency on T cell immunity was determined by flow cytometry and T cell depletion in vivo. RESULTS: After administration of DSS, mPGES-1-/- mice exhibited more severe weight loss, diarrhea, and fecal bleeding than WT mice. Histological analysis further showed significant exacerbation of colonic inflammation in mPGES-1-/- mice. In WT mice, the colonic expression of mPGES-1 was highly induced on both mRNA and protein levels and colonic PGE2 increased significantly after DSS administration. Additionally, mPGES-1 protein was localized in the colonic mucosal epithelium and infiltrated inflammatory cells in underlying connective tissues and the lamina propria. The abnormalities consistent with colitis in mPGES-1-/- mice were associated with higher expression of colonic T-helper (Th)17 and Th1 cytokines, including interleukin 17A and interferon-γ. Furthermore, lack of mPGES-1 increased the numbers of Th17 and Th1 cells in the lamina propria mononuclear cells within the colon, even though the number of suppressive regulatory T cells also increased. CD4+ T cell depletion effectively reduced symptoms of colitis as well as colonic expression of Th17 and Th1 cytokines in mPGES-1-/- mice, suggesting the requirement of CD4+ T cells in the exacerbation of DSS-induced colitis under mPGES-1 deficiency. CONCLUSIONS: These results demonstrate that mPGES-1 is the main enzyme responsible for colonic PGE2 production and deficiency of mPGES-1 facilitates the development of colitis by affecting the development of colonic T cell-mediated immunity. mPGES-1 might therefore impact both the intestinal inflammation and T cell-mediated immunity associated with IBD.

3.
Thromb Haemost ; 119(8): 1311-1320, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31129913

ABSTRACT

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.


Subject(s)
Platelet Activation , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Animals , Bleeding Time , Blood Platelets/metabolism , Cyclic AMP/metabolism , Dinoprost , Female , Hemostasis , Humans , Inositol Phosphates/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Platelet Adhesiveness , Platelet Aggregation , Thromboembolism/blood
4.
Mol Reprod Dev ; 86(7): 786-797, 2019 07.
Article in English | MEDLINE | ID: mdl-31087493

ABSTRACT

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 .


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Chorionic Gonadotropin/pharmacology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Leydig Cells/metabolism , Reproductive Control Agents/pharmacology , Animals , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclooxygenase 2/genetics , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Signal Transduction/drug effects , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription, Genetic , Transfection
5.
FASEB J ; 32(5): 2354-2365, 2018 05.
Article in English | MEDLINE | ID: mdl-29247122

ABSTRACT

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.


Subject(s)
Epoprostenol/pharmacology , Food, Formulated/adverse effects , Hepatocytes/metabolism , Kupffer Cells/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Epoprostenol/analogs & derivatives , Gene Expression Regulation/drug effects , Hepatocytes/pathology , Kupffer Cells/pathology , Liver/pathology , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Receptors, Epoprostenol/agonists , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/metabolism , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
6.
Sci Rep ; 7(1): 8374, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827713

ABSTRACT

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.


Subject(s)
Androgens/blood , Aromatase/genetics , Diethylstilbestrol/administration & dosage , Estrogens, Non-Steroidal/administration & dosage , Estrogens/blood , Granulosa Cells/drug effects , Ovary/drug effects , Theca Cells/drug effects , Animals , Female , Gene Expression Profiling , Gonadotropins/blood , Granulosa Cells/metabolism , Ovary/metabolism , Rats , Steroid 17-alpha-Hydroxylase/analysis , Steroid 17-alpha-Hydroxylase/genetics , Theca Cells/metabolism
7.
TH Open ; 1(2): e122-e129, 2017 Jul.
Article in English | MEDLINE | ID: mdl-31249917

ABSTRACT

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.

8.
J Clin Endocrinol Metab ; 101(10): 3582-3591, 2016 10.
Article in English | MEDLINE | ID: mdl-27428878

ABSTRACT

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.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Cytochrome P-450 Enzyme System/metabolism , Leydig Cells/metabolism , Testosterone/analogs & derivatives , Theca Cells/metabolism , Female , Humans , Male , Testosterone/metabolism , Tumor Cells, Cultured
9.
Acta Derm Venereol ; 96(7): 899-904, 2016 Nov 02.
Article in English | MEDLINE | ID: mdl-27066774

ABSTRACT

To investigate the mechanisms underlying itching in atopic dermatitis, we examined whether thromboxane (TX) A2, an arachidonic acid metabolite, is involved in spontaneous scratching, an itch-related response, in NC mice with atopic dermatitis-like skin lesions. The TXA2 receptor (TP) antagonist ONO-3708 inhibited the spontaneous scratching. The mRNA expression of TX synthase (TXSyn) distributed mainly in epidermis and the concentration of TXB2, a metabolite of TXA2, were increased in lesional skin. Scratching caused by the PAR2 agonist SLIGRL-NH2 was suppressed by ONO-3708. SLIGRL-NH2-induced scratching decreased approximately 75% in TP-deficient mice, compared to wild-type mice. In primary cultures of mouse keratinocytes, SLIGRL-NH2 induced the production of TXA2, as evidenced by the increased TXB2, which was inhibited by the TXSyn inhibitor sodium ozagrel and a PAR2-neutralizing antibody. Taken together, these results suggest that epidermal TXA2, which may be produced via PAR2 activation, is involved in itching in atopic dermatitis.


Subject(s)
Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Pruritus/drug therapy , Pruritus/metabolism , Thromboxane A2/analogs & derivatives , Thromboxane A2/metabolism , Animals , Keratinocytes/metabolism , Male , Methacrylates/pharmacology , Mice , Oligopeptides/adverse effects , Oligopeptides/pharmacology , RNA, Messenger/metabolism , Receptor, PAR-2/antagonists & inhibitors , Thromboxane A2/pharmacology
10.
J Pharmacol Exp Ther ; 353(2): 269-78, 2015 May.
Article in English | MEDLINE | ID: mdl-25740898

ABSTRACT

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.


Subject(s)
Platelet Aggregation Inhibitors/pharmacology , Pyridines/pharmacology , Thromboxane A2/biosynthesis , Administration, Oral , Animals , Blood Pressure/drug effects , Cyclic AMP/biosynthesis , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Male , Mice , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Pyridines/administration & dosage , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Signal Transduction/drug effects , Thromboxane A2/metabolism , Thromboxane-A Synthase/antagonists & inhibitors
11.
Circ J ; 77(4): 1053-62, 2013.
Article in English | MEDLINE | ID: mdl-23257313

ABSTRACT

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.


Subject(s)
Bone Marrow Transplantation , Endothelial Cells/metabolism , Epoprostenol/metabolism , Ischemia/therapy , Microcirculation , Neovascularization, Physiologic , Stem Cells/metabolism , Animals , Cell Adhesion , Disease Models, Animal , Endothelial Cells/pathology , Epoprostenol/genetics , Hindlimb/blood supply , Hindlimb/metabolism , Hindlimb/pathology , Ischemia/genetics , Ischemia/metabolism , Male , Mice , Mice, Knockout , Pericytes/metabolism , Pericytes/pathology , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Stem Cells/pathology
12.
Mol Cell Endocrinol ; 362(1-2): 176-82, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22713853

ABSTRACT

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.


Subject(s)
Dinoprostone/biosynthesis , Estradiol/physiology , Ovary/physiology , Ovulation , 46, XX Disorders of Sex Development/genetics , Animals , Aromatase/deficiency , Aromatase/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Gynecomastia/genetics , Infertility, Male/genetics , Metabolism, Inborn Errors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Transcription, Genetic
13.
Kidney Int ; 82(2): 158-71, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22513820

ABSTRACT

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.


Subject(s)
Dinoprostone/metabolism , Epithelial Cells/metabolism , Kidney Diseases/prevention & control , Kidney Tubules/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Animals , Cell Proliferation , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fibrosis , Folic Acid , Gene Expression Regulation , Heptanoates/pharmacology , Kidney Diseases/etiology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/metabolism , Myofibroblasts/pathology , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/deficiency , Receptors, Prostaglandin E, EP4 Subtype/genetics , Signal Transduction , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/complications
16.
Pharmacol Ther ; 129(2): 195-205, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20920529

ABSTRACT

Prostanoids consisting of prostaglandins (PGs) and thromboxanes (TXs) are produced from arachidonic acids, representative fatty acids contained in cell membrane, by the sequential actions of phospholipase A(2), cyclooxygenases and respective prostanoid synthases. Prostanoids are released outside of the cells immediately after biosynthesis and exert a wide range of actions in the body. These actions are mediated by their respective G protein-coupled receptors expressed in the target cells, which receptors include the DP, EP, FP, IP and TP receptors for PGD(2), PGE(2), PGF(2)α, PGI(2) and TXA(2), respectively. In addition, there are four subtypes of the EP receptors: EP(1), EP(2), EP(3) and EP(4). Recently, roles of prostanoids in the pathogenesis of cardiovascular diseases have been widely examined using mice lacking each prostanoid receptor individually or enzyme participating in prostanoid biosynthesis. These studies have revealed important and novel roles of prostanoids in the development of cardiovascular diseases, such as acute myocardial infarction, cardiac hypertrophy, atherosclerosis, vascular remodeling, hypertension and cerebral thrombosis. Roles of prostanoids in the generation of inflammatory tachycardia and the regulation of platelet function have also been clarified. In this review, we summarize these roles of prostanoids revealed from knockout mouse studies.


Subject(s)
Atherosclerosis/physiopathology , Cardiomegaly/physiopathology , Cardiovascular Diseases/physiopathology , Myocardial Infarction/physiopathology , Prostaglandins/physiology , Animals , Atherosclerosis/metabolism , Cardiomegaly/metabolism , Cardiovascular Diseases/metabolism , Mice , Mice, Knockout , Myocardial Infarction/metabolism , Receptors, Prostaglandin/genetics
19.
Thromb Haemost ; 104(4): 796-803, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20664890

ABSTRACT

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.


Subject(s)
Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/agonists , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Cells, Cultured , Dinoprostone/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Thromboxane/agonists
20.
Arterioscler Thromb Vasc Biol ; 30(3): 464-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007911

ABSTRACT

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.


Subject(s)
Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic/physiology , Animals , Cell Adhesion , Cell Movement , Cell Proliferation , Endothelium, Vascular/pathology , Fibronectins/metabolism , Hyperplasia/metabolism , Hyperplasia/pathology , Mesenchymal Stem Cells/pathology , Mice , Mice, Knockout , Models, Animal , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/metabolism , Signal Transduction/physiology , Tunica Intima/injuries , Tunica Intima/metabolism , Tunica Intima/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...