Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 488
Filtrar
1.
Invest Ophthalmol Vis Sci ; 63(2): 24, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35147658

RESUMO

Purpose: We investigated the intraocular pressure (IOP)-lowering effect of topical sepetaprost (SPT), a dual agonist of the FP and EP3 receptors. We explored whether certain receptors mediated the hypotensive effect of SPT and outflow facility changes in C57BL/6 mice (wild-type [WT]) and FP and EP3 receptor-deficient mice (FPKO and EP3KO mice, respectively). Methods: IOP was measured using a microneedle. Outflow facility was measured using a two-level, constant-pressure perfusion method. Results: SPT significantly reduced IOP for 8 hours after administration to WT mice. The 2-hour IOP reductions afforded by latanoprost were 15.3 ± 2.5, 1.8 ± 2.0, and 12.3 ± 2.4% in WT, FPKO, and EP3KO mice, respectively; the SPT figures were 13.6 ± 2.1, 5.9 ± 2.7, and 6.6 ± 2.6%, respectively. Latanoprost-mediated IOP reduction was significantly decreased in FPKO mice, and SPT-mediated IOP reduction was reduced in both FPKO and EP3KO mice. At 6 hours after administration, latanoprost did not significantly reduce the IOP in any tested mouse strain. SPT-mediated IOP reduction was reduced in both FPKO and EP3KO mice. IOP reduction at 6 hours was significantly higher after simultaneous administration of selective FP and EP3 receptor agonists, but IOP did not fall on administration of (only) a selective EP3 receptor agonist. SPT significantly increased outflow facility in WT mice, but less so in FPKO and EP3KO mice. Conclusions: The IOP-lowering effect of SPT lasted longer than that of latanoprost. Our data imply that this may be attributable to augmented outflow facility mediated by the FP and EP3 receptors.


Assuntos
Anti-Hipertensivos/uso terapêutico , Pressão Intraocular/efeitos dos fármacos , Oxepinas/uso terapêutico , Receptores de Prostaglandina E Subtipo EP3/fisiologia , Receptores de Prostaglandina/fisiologia , Administração Oftálmica , Animais , Humor Aquoso/fisiologia , Dinoprostona/análogos & derivados , Dinoprostona/uso terapêutico , Pressão Intraocular/fisiologia , Latanoprosta/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Soluções Oftálmicas , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina E Subtipo EP3/agonistas , Fatores de Tempo , Tonometria Ocular
2.
Clin Sci (Lond) ; 136(5): 309-321, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35132998

RESUMO

Prostaglandin D2 (PGD2) released from immune cells or other cell types activates its receptors, D prostanoid receptor (DP)1 and 2 (DP1 and DP2), to promote inflammatory responses in allergic and lung diseases. Prostaglandin-mediated inflammation may also contribute to vascular diseases such as abdominal aortic aneurysm (AAA). However, the role of DP receptors in the pathogenesis of AAA has not been systematically investigated. In the present study, DP1-deficient mice and pharmacological inhibitors of either DP1 or DP2 were tested in two distinct mouse models of AAA formation: angiotensin II (AngII) infusion and calcium chloride (CaCl2) application. DP1-deficient mice [both heterozygous (DP1+/-) and homozygous (DP1-/-)] were protected against CaCl2-induced AAA formation, in conjunction with decreased matrix metallopeptidase (MMP) activity and adventitial inflammatory cell infiltration. In the AngII infusion model, DP1+/- mice, but not DP1-/- mice, exhibited reduced AAA formation. Interestingly, compensatory up-regulation of the DP2 receptor was detected in DP1-/- mice in response to AngII infusion, suggesting a potential role for DP2 receptors in AAA. Treatment with selective antagonists of DP1 (laropiprant) or DP2 (fevipiprant) protected against AAA formation, in conjunction with reduced elastin degradation and aortic inflammatory responses. In conclusion, PGD2 signaling contributes to AAA formation in mice, suggesting that antagonists of DP receptors, which have been extensively tested in allergic and lung diseases, may be promising candidates to ameliorate AAA.


Assuntos
Aneurisma da Aorta Abdominal/etiologia , Receptores Imunológicos/fisiologia , Receptores de Prostaglandina/fisiologia , Angiotensina II/farmacologia , Animais , Aneurisma da Aorta Abdominal/prevenção & controle , Masculino , Camundongos , Receptores Imunológicos/antagonistas & inibidores , Receptores de Prostaglandina/antagonistas & inibidores
3.
FASEB J ; 35(6): e21616, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33978990

RESUMO

IgE-dependent/independent activation of mast cell (MC) has been assumed to play a host defensive role against venom injection in skin. However, its detailed mechanisms remain unknown. We aimed to investigate the contribution of MC-derived prostaglandin D2 (PGD2 )-mediated signaling in host defense against bee venom (BV). To achieve this, we utilized gene-deficient mice of a PGD2 receptor, chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2). We first confirmed that subcutaneous injection of BV produced PGD2 equally in wild-type (WT) and CRTH2-deficient (Crth2-/- ) mice skins. The BV injection dropped body temperature and impaired kidney equally in both lines of mice. In WT mice, pre-injection of BV (3 weeks) significantly inhibited the hypothermia and kidney impairment caused by second BV injection. In contrast, this pre-injection was not effective for the second BV injection in Crth2-/- mice. We also found that BV injections increased serum BV-specific IgE levels in WT mice, and its serum transfused mice improved the BV-induced hypothermia in naïve WT mice. In contrast, serum BV-specific IgE level was significantly lower in Crth2-/- mice. FACS analysis showed the BV injection stimulate migration of dendritic cells (DCs) into regional lymph nodes in WT mice. In Crth2-/- mice, its number was significantly smaller than that of WT mice. In conclusion, PGD2 /CRTH2 signaling plays defensive role against second BV injection. This signaling promotes BV-specific IgE production at least partially by promoting DCs migration into regional lymph node.


Assuntos
Imunidade Adaptativa/genética , Venenos de Abelha/toxicidade , Mastócitos/imunologia , Prostaglandina D2/metabolismo , Receptores Imunológicos/fisiologia , Receptores de Prostaglandina/fisiologia , Células Th2/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Feminino , Imunoglobulina E/metabolismo , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Células Th2/efeitos dos fármacos , Células Th2/metabolismo
4.
Yakugaku Zasshi ; 141(4): 473-479, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-33790113

RESUMO

Prostanoids [prostaglandins (PGs) and thromboxanes (TXs)] are a series of bioactive lipid metabolites that function in an autacoid manner via activation of cognate G protein-coupled receptors (GPCRs). The nine subtypes of prostanoid receptors (DP1, DP2, EP1, EP2, EP3, EP4, FP, IP, TP) are involved in a wide range of functions, including inflammation, immune response, reproduction, and homeostasis of the intestinal mucosa and cardiovascular system. Among the prostanoid receptors, the structure of antagonist-bound DP2, which belongs to the chemoattractant receptor family, was previously determined. However, the mechanisms of prostanoid recognition and receptor activation remained elusive. To address this issue, we determined the crystal structures of antagonist-bound EP4 and PGE2-bound EP3. The EP3-PGE2 complex exhibits an active-like conformation, including outward movement of the cytoplasmic end of transmembrane (TM) 6 relative to the cytoplasmic end of TM6 of the EP4 complex. The carboxyl moiety of PGE2 is recognized through three hydrogen bonds formed by highly conserved residues: Y1142.65, T206Extracelluar loop 2 (ECL2), and R3337.40 (superscripts denote Ballesteros-Weinstein numbering). In addition, the ω-chain of PGE2 orients toward TM6, which appears to contribute to receptor activation. The structure reveals important insights into the activation mechanism of prostanoid receptors and provides a molecular basis for the binding modes of endogenous ligands. These findings should facilitate the development of subtype-selective and non-PG-like ligands.


Assuntos
Receptores de Prostaglandina/química , Receptores de Prostaglandina/metabolismo , Cristalografia por Raios X , Dinoprostona/química , Dinoprostona/metabolismo , Ligantes , Conformação Molecular , Ligação Proteica , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Prostaglandina/fisiologia , Receptores de Prostaglandina E Subtipo EP3/química , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Prostaglandina E Subtipo EP4/química , Receptores de Prostaglandina E Subtipo EP4/metabolismo
5.
Med Hypotheses ; 143: 110122, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32759007

RESUMO

A characteristic feature of COVID-19 disease is lymphopenia. Lymphopenia occurs early in the clinical course and is a predictor of disease severity and outcomes. The mechanism of lymphopenia in COVID-19 is uncertain. It has been variously attributed to the release of inflammatory cytokines including IL-6 and TNF-α; direct infection of the lymphocytes by the virus; and rapid sequestration of lymphocytes in the tissues. Additionally, we postulate that prostaglandin D2 (PGD2) is a key meditator of lymphopenia in COVID-19. First, SARS-CoV infection is known to stimulate the production of PGD2 in the airways, which inhibits the host dendritic cell response via the DP1 receptor signaling. Second, PGD2 is known to upregulate monocytic myeloid-derived suppressor cells (MDSC) via the DP2 receptor signaling in group 2 innate lymphoid cells (ILC2). We propose targeting PGD2/DP2 signaling using a receptor antagonist such as ramatroban as an immunotherapy for immune dysfunction and lymphopenia in COVID-19 disease.


Assuntos
Betacoronavirus , Infecções por Coronavirus/fisiopatologia , Linfopenia/fisiopatologia , Modelos Imunológicos , Terapia de Alvo Molecular , Pandemias , Pneumonia Viral/fisiopatologia , Prostaglandina D2/fisiologia , Sistema Respiratório/metabolismo , Adulto , COVID-19 , Carbazóis/farmacologia , Carbazóis/uso terapêutico , Criança , Infecções por Coronavirus/complicações , Infecções por Coronavirus/imunologia , Células Dendríticas/imunologia , Humanos , Linfopenia/etiologia , Células Mieloides/imunologia , Pneumonia Viral/complicações , Pneumonia Viral/imunologia , Prostaglandina D2/biossíntese , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina/fisiologia , SARS-CoV-2 , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Linfócitos T/imunologia , Tromboxano A2/antagonistas & inibidores
6.
PLoS One ; 15(5): e0232591, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32365129

RESUMO

Prostaglandin F2 alpha (PGF2α) analogues such as latanoprost are common first-line intraocular pressure (IOP) lowering medications. However, their clinical use is limited in some patient populations due to minimal or no IOP lowering response or side effects. In searching for a more targeted approach for IOP reduction, our lab recently identified Stanniocalcin-1 (STC-1) as a molecule that was required for latanoprost-mediated IOP reduction and also acted as a stand-alone IOP lowering agent. In order to determine whether latanoprost and STC-1 were equivalent and/or additive for IOP reduction, we treated C57BL/6J mice with one or a combination of these agents and measured IOP. Importance of the FP receptor for latanoprost- and STC-1-mediated IOP reduction was examined in C57BL/6J mice utilizing the pharmacologic FP receptor inhibitor AL-8810 as well as FP receptor knockout mice generated in our laboratory. Latanoprost-free acid (LFA) and STC-1 reduced IOP to a similar degree and were non-additive in C57BL/6J mice. As expected, the IOP lowering effects of LFA were abrogated by pharmacologic inhibition of the FP receptor with AL-8810 and in FP receptor knockout mice. In contrast, STC-1 maintained IOP-lowering effects in the presence of AL-8810 and also in FP receptor knockout mice. These results suggest that LFA and STC-1 show equivalent and non-additive IOP reduction in C57BL/6J mice and that unlike LFA, STC-1-mediated IOP reduction occurs independent of the FP receptor.


Assuntos
Glicoproteínas/fisiologia , Pressão Intraocular/efeitos dos fármacos , Latanoprosta/metabolismo , Receptores de Prostaglandina/fisiologia , Animais , Dinoprosta/análogos & derivados , Dinoprosta/farmacologia , Feminino , Genótipo , Homozigoto , Latanoprosta/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Prostaglandina/genética , Transdução de Sinais
7.
Naunyn Schmiedebergs Arch Pharmacol ; 392(4): 437-450, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30552456

RESUMO

Various studies have confirmed that prostaglandins (PG) alter the bladder motor activity and micturition reflex in both human and animals. However, no sufficient data is reported about the effect of cyclooxygenase (COX) inhibitors neither in normal bladder physiology nor in pathological conditions. This study aims to compare the potential effects of some COX inhibitors with varying COX-1/COX-2 selectivities (indomethacin, ketoprofen, and diclofenac) with that of the selective COX-2 inhibitor (DFU) on bladder function. The role played by some PGs and their receptors in controlling detrusor muscle function in normal condition and in cystitis is also studied. Organ bath experiments were performed using isolated rat detrusor muscle. Direct and neurogenic contractions were induced using ACh and electric stimulation (EFS), respectively. A model of hemorrhagic cystitis was induced by single injection of cyclophosphamide (300 mg/kg) in rats, and confirmed by histophathological examination. Results are expressed as mean ± SEM of 5-9 rats. Alprostadil and iloprost (1 nM- 10 µM) concentration-dependently potentiated ACh (100 µM)- and EFS (4 Hz)-induced contraction, with maximum potentiation of 40.01 ± 5.29 and 27.59 ± 6.64%, respectively, in case of ACh contractions. In contrast, ONO-AE1-259 (selective EP2 agonist, 1 nM-10 µM) inhibited muscle contraction. SC51322 (EP1-antagonist, 10 µM) and RO1138452 (IP antagonist, 10 µM) inhibited both direct and neurogenic responses. Hemorrhagic cystitis reduced both ACh and EFS responses as well as the potentiatory effect of iloprost and the inhibitory effect of RO1138452 on ACh contractions. ONO-AE3-237 (DP1 antagonist, 1 µM) significantly potentiated contractions in cystitis but showed no effect in normal bladder. A significant inhibition of contractile response was observed in presence of indomethacin, ketoprofen, and diclofenac at all tested concentrations (20, 50, and 100 µM). Highest effect was induced by diclofenac. The effect of these COX inhibitors on EFS contractions was intensified in case of cystitis, indomethacin being the most potent. Atropine (1 nM) significantly reduced indomethacin effect on ACh contraction only in normal rats. On the other hand, DFU (10-6 M) significantly potentiated the contractile effect of ACh in case of cystitis although it showed no effect in normal rats. EP1 receptors seem to play an important role in rat bladder contractility. DP1 receptors as COX-2, on the other hand, gain an important role only in case of cystitis. The use of non-selective COX inhibitors in cystitis may be associated with bladder hypoactivity; selective COX-2 inhibitors may be a safer option.


Assuntos
Inibidores de Ciclo-Oxigenase/efeitos adversos , Cistite/patologia , Músculo Liso/efeitos dos fármacos , Receptores de Prostaglandina/antagonistas & inibidores , Bexiga Urinária/efeitos dos fármacos , Animais , Ciclo-Oxigenase 1/fisiologia , Ciclo-Oxigenase 2/fisiologia , Ciclofosfamida , Cistite/induzido quimicamente , Cistite/fisiopatologia , Modelos Animais de Doenças , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso/patologia , Músculo Liso/fisiologia , Ratos Wistar , Receptores de Prostaglandina/fisiologia , Bexiga Urinária/patologia , Bexiga Urinária/fisiologia
8.
Circulation ; 134(4): 328-38, 2016 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-27440004

RESUMO

BACKGROUND: Inhibitors of cyclooxygenase-2 alleviate pain and reduce fever and inflammation by suppressing the biosynthesis of prostacyclin (PGI2) and prostaglandin E2. However, suppression of these prostaglandins, particularly PGI2, by cyclooxygenase-2 inhibition or deletion of its I prostanoid receptor also predisposes to accelerated atherogenesis and thrombosis in mice. By contrast, deletion of microsomal prostaglandin E synthase 1 (mPGES-1) confers analgesia, attenuates atherogenesis, and fails to accelerate thrombogenesis, while suppressing prostaglandin E2, but increasing biosynthesis of PGI2. METHODS: To address the cardioprotective contribution of PGI2, we generated mice lacking the I prostanoid receptor together with mPges-1 on a hyperlipidemic background (low-density lipoprotein receptor knockouts). RESULTS: mPges-1 depletion modestly increased thrombogenesis, but this response was markedly further augmented by coincident deletion of the I prostanoid receptor (n=10-18). By contrast, deletion of the I prostanoid receptor had no effect on the attenuation of atherogenesis by mPGES-1 deletion in the low-density lipoprotein receptor knockout mice (n=17-21). CONCLUSIONS: Although suppression of prostaglandin E2 accounts for the protective effect of mPGES-1 deletion in atherosclerosis, augmentation of PGI2 is the dominant contributor to its favorable thrombogenic profile. The divergent effects on these prostaglandins suggest that inhibitors of mPGES-1 may be less likely to cause cardiovascular adverse effects than nonsteroidal anti-inflammatory drugs specific for inhibition of cyclooxygenase-2.


Assuntos
Aterosclerose/enzimologia , Epoprostenol/fisiologia , Hiperlipidemias/genética , Prostaglandina-E Sintases/deficiência , Receptores de Prostaglandina/deficiência , Animais , Doenças da Aorta/enzimologia , Doenças da Aorta/genética , Aterosclerose/genética , Artéria Carótida Primitiva/efeitos da radiação , Estenose das Carótidas/etiologia , Hiperlipidemias/enzimologia , Lasers/efeitos adversos , Camundongos , Camundongos Knockout , Microssomos/enzimologia , Polimorfismo de Nucleotídeo Único , Prostaglandina-E Sintases/genética , Prostaglandina-E Sintases/fisiologia , Receptores de Epoprostenol , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/fisiologia
9.
Eur J Pharmacol ; 765: 15-23, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26277322

RESUMO

Prostaglandin (PG) D2 elicits responses through either the DP1 and/or DP2 receptor. Experimental evidence suggests that stimulation of the DP1 receptor contributes to allergic responses, such that antagonists are considered to be directed therapies for allergic diseases. In this study, we demonstrate the activity of a novel synthetic DP1 receptor antagonist termed asapiprant (S-555739) for the DP1 receptor and other receptors in vitro, and assess the efficacy of asapiprant in several animal models of allergic diseases. We determined the affinity and selectivity of asapiprant for the DP1 receptor in binding assays. In the animal models of allergic rhinitis, changes in nasal resistance, nasal secretion, and cell infiltration in nasal mucosa were assessed after antigen challenge with and without asapiprant. Similarly, in the animal models of asthma, the effect of antigen challenge with and without asapiprant on antigen-induced bronchoconstriction, airway hyper-responsiveness, mucin production, and cell infiltration in lung were assessed. In binding studies, asapiprant exhibited high affinity and selectivity for the DP1 receptor. Significant suppression of antigen-induced nasal resistance, nasal secretion, and cell infiltration in nasal mucosa was observed with asapiprant treatment. In addition, treatment with asapiprant suppressed antigen-induced asthmatic responses, airway hyper-responsiveness, and cell infiltration and mucin production in lung. These results show that asapiprant is a potent and selective DP1 receptor antagonist, and exerts suppressive effects in the animal models of allergic diseases. Thus, asapiprant has potential as a novel therapy for allergic airway diseases.


Assuntos
Asma/tratamento farmacológico , Modelos Animais de Doenças , Receptores de Prostaglandina/antagonistas & inibidores , Rinite Alérgica/tratamento farmacológico , Tiofenos/uso terapêutico , Animais , Asma/imunologia , Asma/metabolismo , Cães , Feminino , Cobaias , Humanos , Masculino , Prostaglandinas/química , Prostaglandinas/farmacologia , Prostaglandinas/uso terapêutico , Ratos , Ratos Endogâmicos BN , Receptores de Prostaglandina/fisiologia , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/metabolismo , Rinite Alérgica/imunologia , Rinite Alérgica/metabolismo , Ovinos , Tiofenos/química , Tiofenos/farmacologia , Resultado do Tratamento
10.
Eur J Pharmacol ; 764: 249-255, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26151307

RESUMO

The soluble guanylyl cyclase/cGMP system plays an important role in the vasodilator response to nitric oxide (NO) in various vascular beds. However, in rat retinal arterioles, the cyclooxygenase-1/cAMP-mediated pathway contributes to the vasodilator effects of NO, although the specific prostanoid involved remains to be elucidated. In the present study, we investigated the role of prostaglandin I2 and its receptor (prostanoid IP receptor) system in NO-induced vasodilation of rat retinal arterioles in vivo. Fundus images were captured using a digital camera that was equipped with a special objective lens. Changes in diameter of retinal arterioles were assessed. The NO donor (±)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide (NOR3) increased the diameter of retinal arterioles but decreased systemic blood pressure in a dose-dependent manner. Treatment of rats with indomethacin, a non-selective cyclooxygenase inhibitor, markedly attenuated the retinal vasodilator, but not depressor responses to NOR3. The prostanoid IP receptor antagonist 4,5-dihydro-N-[4-[[4-(1-methylethoxy)phenyl]methyl]phenyl]-1H-imadazol-2-amine (CAY10441), and the prostaglandin I2 synthase inhibitor 9α,11α-azoprosta-5Z,13E-dien-1-oic acid (U-51605), both showed similar preventive effects against the NOR3-induced retinal vasodilator response. Neither CAY10441 nor U-51605 showed any significant effects on the depressor response to NOR3. NOR3 enhanced the release of prostaglandin I2 from cultured human retinal microvascular endothelial cells and the NOR3-induced prostaglandin I2 release was almost completely abolished by the cyclooxygenase-1 inhibitor SC-560, but not by the cyclooxygenase-2 inhibitor NS-398. However, NOR3 did not increase the release of prostaglandin I2 from human intestinal microvascular endothelial cells. These results suggest that NO exerts its dilatory effect via cyclooxygenase-1/prostaglandin I2/prostanoid IP receptor signaling mechanisms in the retinal vasculature.


Assuntos
Arteríolas/fisiologia , Epoprostenol/fisiologia , Vasos Retinianos/fisiologia , Animais , Arteríolas/efeitos dos fármacos , Compostos de Benzil/farmacologia , Células Cultivadas , Ciclo-Oxigenase 1/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Células Endoteliais/metabolismo , Humanos , Hidroxilaminas/farmacologia , Imidazóis/farmacologia , Masculino , Óxido Nítrico/fisiologia , Doadores de Óxido Nítrico/farmacologia , Nitrocompostos , Nitrobenzenos/farmacologia , Prostaglandinas H/farmacologia , Pirazóis/farmacologia , Ratos Wistar , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/fisiologia , Vasos Retinianos/efeitos dos fármacos , Sulfonamidas/farmacologia , Vasodilatação/efeitos dos fármacos
11.
Biochem Pharmacol ; 96(4): 306-14, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26067757

RESUMO

Metastasis is the major cause of death in cancer patients. Elevated expression of cyclooxygenase-2 (COX-2) is observed in many human cancers and over-production of downstream prostaglandins (PGs) has been shown to stimulate metastasis. A role for increased PGE2 production has been proposed, but whether other PGs contribute is currently unclear. In this study the pro-migratory actions of individual PGs were evaluated in MDA-MB-468 breast cancer cells that stably over-expressed COX-2 (MDA-COX-2 cells); cell migration was quantified using 3D-matrigel droplet assays. Inhibition of the prostacyclin and PGE synthases, but not alternate prostanoid synthases, prevented the increase in MDA-COX-2 cell migration produced by arachidonic acid (AA); direct treatment of cells with the stable prostacyclin analogue cicaprost also promoted migration. Pharmacological antagonism and knockdown of the IP receptor decreased cell migration, while antagonists of the alternate DP, EP2, FP, and TP prostanoid receptors were inactive. In support of these findings, activation of the IP receptor also enhanced migration in the MDA-MB-468, MDA-MB-231 and A549 cell lines, and IP receptor knock-down in MDA-COX-2 cells decreased the expression of a number of pro-migratory genes. In further studies, the prostacyclin/IP receptor and PGE2/EP4 receptor pathways were found to be functionally independent and the inhibition of phosphatidylinositol 3-kinase (PI3K) and p38 mitogen-activated protein kinase (MAPK) selectively impaired the IP-receptor-dependent migration in MDA-COX-2 cells. Taken together, the prostacyclin/IP/PI3K-p38 MAPK axis has emerged as a novel pro-migratory pathway in breast cancer cells that over-express COX-2. This information could be utilized in novel treatment strategies to minimize tumor metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Ciclo-Oxigenase 2/metabolismo , Receptores de Prostaglandina/fisiologia , Ácido Araquidônico/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Técnicas de Inativação de Genes , Humanos , Masculino , Metástase Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Receptores de Epoprostenol , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
J Physiol Biochem ; 71(3): 351-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25940857

RESUMO

Several lines of evidence suggest that cyclooxygenase-2 (COX-2) activity can have a beneficial role in the maintenance of vascular tone of the blood vessels in diabetes. Specifically, the increased production of prostacyclin (PGI2) and prostaglandin E2 (PGE2), mediated by COX-2, has been suggested to compensate for decreased synthesis of nitric oxide (NO). The study investigates whether inhibition of COX-2 may reduce the coronary flow in diabetic animals and may also lead to decreased synthesis of prostaglandins. Mice aged 18-20 weeks were used for the study: those with leptin receptor deficiency (db/db) served as a model of diabetes while heterozygous (db/+) mice served as controls. Coronary flow was measured by the Langendorff method, and prostaglandin synthesis by myocardia was assayed in heart perfusates. COX-2 inhibition was found to reduce basal coronary flow in db/db mice but had no effect in db/+ mice. Secretion of PGE2 was found to be higher in db/db mice, while prostacyclin synthesis did not differ. COX-2 inhibition decreased production of both prostaglandins to similar levels in both groups. The use of ONO-1301, a specific agonist for the prostacyclin receptor revealed that vasodilating responses mediated by the receptor were impaired in db/db mice. The expression levels of the receptor in cardiac tissue did not differ between the groups. It is concluded that the increased COX-2 contribution to vasodilation in diabetic animals appears to be partially a result of increased COX-2-dependent synthesis of PGE2 and also may be caused by impaired vasodilation mediated by the prostacyclin receptor.


Assuntos
Vasos Coronários/fisiopatologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Diabetes Mellitus Tipo 2/fisiopatologia , Dinoprostona/fisiologia , Nitrobenzenos/farmacologia , Receptores de Prostaglandina/fisiologia , Sulfonamidas/farmacologia , Animais , Ciclo-Oxigenase 2/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/enzimologia , Epoprostenol/fisiologia , Masculino , Camundongos Obesos , Receptores de Epoprostenol , Fluxo Sanguíneo Regional/efeitos dos fármacos , Vasodilatação
13.
Br J Pharmacol ; 172(16): 4024-37, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25917171

RESUMO

BACKGROUND AND PURPOSE: We have described a urothelium-dependent release of PGD2 -like activity which had inhibitory effects on the motility of guinea pig urinary bladder. Here, we have pharmacologically characterized the receptors involved and localized the sites of PGD2 formation and of its receptors. EXPERIMENTAL APPROACH: In the presence of selective DP and TP receptor antagonists alone or combined, PGD2 was applied to urothelium-denuded diclofenac-treated urinary bladder strips mounted in organ baths. Antibodies against PGD2 synthase and DP1 receptors were used with Western blots and for histochemistry. KEY RESULTS: PGD2 inhibited nerve stimulation -induced contractions in strips of guinea pig urinary bladder with estimated pIC50 of 7.55 ± 0.15 (n = 13), an effect blocked by the DP1 receptor antagonist BW-A868C. After blockade of DP1 receptors, PGD2 enhanced the contractions, an effect abolished by the TP receptor antagonist SQ-29548. Histochemistry revealed strong immunoreactivity for PGD synthase in the urothelium/suburothelium with strongest reaction in the suburothelium. Immunoreactive DP1 receptors were found in the smooth muscle of the bladder wall with a dominant localization to smooth muscle membranes. CONCLUSIONS AND IMPLICATIONS: In guinea pig urinary bladder, the main effect of PGD2 is an inhibitory action via DP1 receptors localized to the smooth muscle, but an excitatory effect via TP receptors can also be evoked. The urothelium with its suburothelium might signal to the smooth muscle which is rich in PGD2 receptors of the DP1 type. The results are important for our understanding of regulation of bladder motility.


Assuntos
Prostaglandina D2/farmacologia , Receptores de Prostaglandina/fisiologia , Receptores de Tromboxanos/fisiologia , Bexiga Urinária/efeitos dos fármacos , Animais , Compostos Bicíclicos Heterocíclicos com Pontes , Ácidos Graxos Insaturados , Feminino , Cobaias , Hidantoínas/farmacologia , Hidrazinas/farmacologia , Técnicas In Vitro , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Tromboxanos/antagonistas & inibidores , Bexiga Urinária/fisiologia
14.
Diabetes ; 63(9): 2911-23, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24722246

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs), including acetylsalicylic acid (ASA), improve glucose metabolism in diabetic subjects, although the underlying mechanisms remain unclear. In this study, we observed dysregulated expression of cyclooxygenase-2, prostacyclin biosynthesis, and the I prostanoid receptor (IP) in the liver's response to diabetic stresses. High doses of ASA reduced hepatic prostaglandin generation and suppressed hepatic gluconeogenesis in mice during fasting, and the hypoglycemic effect of ASA could be restored by IP agonist treatment. IP deficiency inhibited starvation-induced hepatic gluconeogenesis, thus inhibiting the progression of diabetes, whereas hepatic overexpression of IP increased gluconeogenesis. IP deletion depressed cAMP-dependent CREB phosphorylation and elevated AKT phosphorylation by suppressing PI3K-γ/PKC-ζ-mediated TRB3 expression, which subsequently downregulated the gluconeogenic genes for glucose-6-phosphatase (G6Pase) and phosphoenol pyruvate carboxykinase 1 in hepatocytes. We therefore conclude that suppression of IP modulation of hepatic gluconeogenesis through the PKA/CREB and PI3K-γ/PKC-ζ/TRB3/AKT pathways contributes to the effects of NSAIDs in diabetes.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Gluconeogênese/efeitos dos fármacos , Fígado/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptores de Prostaglandina/fisiologia , Animais , Aspirina/farmacologia , Proteínas de Ciclo Celular/fisiologia , Diabetes Mellitus Experimental/prevenção & controle , Dieta Hiperlipídica , Epoprostenol/fisiologia , Jejum , Glucose-6-Fosfatase/metabolismo , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Epoprostenol , Receptores de Prostaglandina/deficiência
15.
J Mol Med (Berl) ; 92(6): 629-40, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24500109

RESUMO

UNLABELLED: Prostaglandin F2(α)-F-prostanoid (PGF2(α)-FP) receptor is closely related to insulin resistance, which plays a causal role in the pathogenesis of diabetic cardiomyopathy (DCM). We sought to reveal whether PGF2(α)-FP receptor plays an important part in modulating DCM and the mechanisms involved. We established the type 2 diabetes rat model by high-fat diet and low-dose streptozotocin (STZ) and then evaluated its characteristics by metabolite tests, Western blot analysis for FP-receptor expression, histopathologic analyses of cardiomyocyte density and fibrosis area. Next, we used gene silencing to investigate the role of FP receptor in the pathophysiologic features of DCM. Our study showed elevated cholesterol, triglyceride, glucose, and insulin levels, severe insulin resistance, and FP-receptor overexpression in diabetic rats. The collagen volume fraction (CVF) and perivascular collagen area/luminal area (PVCA/LA) were higher in the diabetic group than the control group (CVF% 10.99 ± 0.99 vs 1.59 ± 0.18, P < 0.05; PVCA/LA% 17.07 ± 2.61 vs 2.86 ± 0.69, P < 0.05). We found that the silencing of FP receptor decreased cholesterol, triglyceride, glucose, and insulin levels and ameliorated insulin resistance. The CVF and PVCF/LA were significantly downregulated in FP-receptor short hairpin RNA (shRNA) treatment group (FP-receptor shRNA group vs vehicle group: CVF% 5.59 ± 0.92 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 4.74 ± 1.57 vs 14.79 ± 2.22, P < 0.05; FP-receptor shRNA + PGF2(α) group vs vehicle group : CVF% 5.19 ± 0.79 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 5.96 ± 1.15 vs 14.79 ± 2.22, P < 0.05, respectively). Furthermore, with FP-receptor gene silencing, the activated protein kinase C (PKC) and Rho kinase were significantly decreased, and the blunted phosphorylation of Akt was restored. FP-receptor gene silencing may exert a protective effect on DCM by improving myocardial fibrosis, suggesting a new therapeutic approach for human DCM. KEY MESSAGES: FP-receptor gene silencing improves glucose tolerance and insulin resistance in type 2 diabetes (T2D). FP-receptor gene silencing modulates the activities of PKC/Rho and Akt signaling pathways in T2D. FP-receptor gene silencing decreases collagen expression and ameliorates myocardial fibrosis in T2D. FP-receptor gene silencing protects from diabetic cardiomyopathy in T2D.


Assuntos
Cardiomiopatias Diabéticas/prevenção & controle , Inativação Gênica/fisiologia , Miocárdio/patologia , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina/fisiologia , Animais , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/terapia , Cardiomiopatias Diabéticas/sangue , Dieta Hiperlipídica , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina/genética
16.
Reproduction ; 147(5): 703-17, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24481956

RESUMO

Although there is no acute luteolytic mechanism in the absence of pregnancy in the bitch, a precise and well-timed embryo-maternal interaction seems to be required for the initiation and maintenance of gestation. As only limited information is available about these processes in dogs, in this study, the uterine expression of possible decidualization markers was investigated during the pre-implantation stage (days 10-12) of pregnancy and in the corresponding nonpregnant controls. In addition, the expression of selected genes associated with blastocyst development and/or implantation was investigated in embryos flushed from the uteri of bitches used for this study (unhatched and hatched blastocysts). There was an upregulated expression of prolactin receptor (PRLR) and IGF2 observed pre-implantation. The expression of PRL and of IGF1 was unaffected, and neither was the expression of progesterone- or estrogen receptor ß (ESR2). In contrast, (ESR1) levels were elevated during early pregnancy. Prostaglandin (PG)-system revealed upregulated expression of PGE2-synthase and its receptors, PTGER2 and PTGER4, and of the PG-transporter. Elevated levels of AKR1C3 mRNA, but not the protein itself, were noted. Expression of prostaglandin-endoperoxide synthase 2 (PTGS2) remained unaffected. Most of the transcripts were predominantly localized to the uterine epithelial cells, myometrium and, to a lesser extent, to the uterine stroma. PGES (PTGES) mRNA was abundantly expressed in both groups of embryos and appeared higher in the hatched ones. The expression level of IGF2 mRNA appeared higher than that of IGF1 mRNA in hatched embryos. In unhatched embryos IGF1, IGF2, and PTGS2 mRNA levels were below the detection limit.


Assuntos
Cães/fisiologia , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Prenhez/fisiologia , Útero/fisiologia , Animais , Cães/genética , Desenvolvimento Embrionário/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/fisiologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/fisiologia , Gravidez , Proteínas da Gravidez/genética , Proteínas da Gravidez/fisiologia , Prenhez/genética , RNA Mensageiro/genética , RNA Mensageiro/fisiologia , Receptores da Prolactina/genética , Receptores da Prolactina/fisiologia , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/fisiologia
17.
Psychiatry Clin Neurosci ; 68(1): 50-60, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23992456

RESUMO

The aim of this review was to clarify the role of prostaglandins and prostaglandin receptors in the immunopathology of Alzheimer's disease. A PubMed search was done using the key word, 'Alzheimer's disease' in combination with the term 'prostaglandins'. Articles from the past 10 years were preferentially selected but important ones from the past 20 years were also included according to the authors' judgment. Alzheimer's disease is characterized by pathological hallmarks such as extracellular deposition of the amyloid ß-peptide, the appearance of intracellular neurofibrillary tangles, extensive neuronal loss and synaptic changes in the cerebral cortex and hippocampus. These processes induce inflammatory pathways by activating microglia, astrocytes and infiltrating leukocytes that produce inflammatory mediators including cytokines and prostaglandins.Prostaglandins are small lipid mediators derived from arachidonic acid by multi-enzymatic pathways in which cyclooxygenases and phospholipases are the rate-limiting enzymes. In the central nervous system, prostaglandins exhibit either neurotoxic or neuroprotective effects by acting on specific G-protein-coupled receptors that have different subfamilies and differences in their selective agonists, tissue distribution and signal transduction cascades. Further studies on the role of prostaglandins in Alzheimer's disease may contribute to clarification of their neuroprotective actions, which may lead to the development of successful therapeutic strategies.


Assuntos
Doença de Alzheimer/fisiopatologia , Fármacos Neuroprotetores/metabolismo , Prostaglandinas/metabolismo , Doença de Alzheimer/complicações , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Humanos , Inflamação/complicações , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Receptores de Prostaglandina/fisiologia
18.
J Pharmacol Sci ; 123(4): 392-401, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24292413

RESUMO

The purpose of this study was to evaluate the angiogenic potency of ONO-1301, a novel prostacyclin agonist, using a murine sponge model. Solutions of ONO-1301 or hepatocyte growth factor (HGF), as a positive control, were injected into sponges in the backs of mice, daily for 14 days. Hemoglobin and HGF levels in the sponge were increased for up to 14 days on daily treatment with ONO-1301 while on HGF treatment, they peaked on day 7 and had decreased again by day 14. ONO-1301 also upregulated c-Met expression for 14 days in a dose-dependent manner. When the mice were pretreated with an antibody to HGF or the prostaglandin I (IP)-receptor antagonist CAY10441, the angiogenic effect of ONO-1301 was dramatically reduced. Plasma concentrations of cyclic adenosine monophosphate (cAMP) were increased in a dose-dependent manner by once daily treatment with ONO-1301 for 14 days. This effect was reduced by pretreatment with the IP-receptor antagonist. In conclusion, hemoglobin level was increased by repeated treatment with ONO-1301 for 14 days. It is suggested that ONO-1301 induced angiogenesis by promoting the synthesis of HGF and upregulated c-Met expression, followed by an increase in cAMP concentrations mediated by IP-receptor signaling.


Assuntos
AMP Cíclico/sangue , Epoprostenol/agonistas , Fator de Crescimento de Hepatócito/biossíntese , Neovascularização Fisiológica/efeitos dos fármacos , Piridinas/farmacologia , Receptores de Prostaglandina/fisiologia , Transdução de Sinais/fisiologia , Animais , Relação Dose-Resposta a Droga , Hemoglobinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores de Epoprostenol , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
19.
Neuropeptides ; 46(6): 353-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23141054

RESUMO

Prostaglandin (PG) D(2), the most abundant PG in the central nervous system (CNS), is a bioactive lipid having various central actions including sleep induction, hypothermia and modulation of the pain response. We found that centrally administered PGD(2) stimulates food intake via the DP(1) among the two receptor subtypes for PGD(2) in mice. Hypothalamic mRNA expression of lipocalin-type PGD synthase (L-PGDS), which catalyzes production of PGD(2) from arachidonic acid via PGH(2) in the CNS, was increased after fasting. Central administration of antagonist and antisense ODN for the DP(1) receptor remarkably decreased food intake, body weight and fat mass. The orexigenic activity of PGD(2) was also blocked by an antagonist of Y(1) receptor for NPY, the most potent orexigenic peptide in the hypothalamus. Thus, the central PGD(2)-NPY system may play a critical role in food intake regulation under normal physiological conditions. We also found that orally active orexigenic peptide derived from food protein activates the PGD(2)-NPY system, downstream of δ opioid receptor. We revealed that the δ agonist peptide, rubiscolin-6-induced orexigenic activity was mediated by L-PGDS in the leptomeninges but not parenchyma using conditional knockout mice. In this review, we discuss the PGD(2)-NPY system itself, and orexigenic signals to activate it.


Assuntos
Apetite/efeitos dos fármacos , Apetite/fisiologia , Neuropeptídeo Y/farmacologia , Neuropeptídeo Y/fisiologia , Prostaglandina D2/farmacologia , Prostaglandina D2/fisiologia , Receptores Opioides delta/fisiologia , Animais , Humanos , Oxirredutases Intramoleculares/metabolismo , Lipocalinas/metabolismo , Meninges/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores Imunológicos/efeitos dos fármacos , Receptores Imunológicos/fisiologia , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Prostaglandina/fisiologia , Ribulose-Bifosfato Carboxilase/farmacologia
20.
PLoS One ; 7(9): e45273, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984630

RESUMO

Although prostanoids are known to be involved in regulation of the spontaneous beating rate of cultured neonatal rat cardiomyocytes, the various subtypes of prostanoid receptors have not been investigated in detail. In our experiments, prostaglandin (PG)F(2α) and prostanoid FP receptor agonists (fluprostenol, latanoprost and cloprostenol) produced a decrease in the beating rate. Two prostanoid IP receptor agonists (iloprost and beraprost) induced first a marked drop in the beating rate and then definitive abrogation of beating. In contrast, the prostanoid DP receptor agonists (PGD(2) and BW245C) and TP receptor agonists (U-46619) produced increases in the beating rate. Sulprostone (a prostanoid EP(1) and EP(3) receptor agonist) induced marked increases in the beating rate, which were suppressed by SC-19220 (a selective prostanoid EP(1) antagonist). Butaprost (a selective prostanoid EP(2) receptor agonist), misoprostol (a prostanoid EP(2) and EP(3) receptor agonist), 11-deoxy-PGE(1) (a prostanoid EP(2), EP(3) and EP(4) receptor agonist) did not alter the beating rate. Our results strongly suggest that prostanoid EP(1) receptors are involved in positive regulation of the beating rate. Prostanoid EP(1) receptor expression was confirmed by western blotting with a selective antibody. Hence, neonatal rat cardiomyocytes express both prostanoid IP and FP receptors (which negatively regulate the spontaneous beating rate) and prostanoid TP, DP(1) and EP(1) receptors (which positively regulate the spontaneous beating rate).


Assuntos
Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Animais Recém-Nascidos , Western Blotting , Células Cultivadas , Cloprostenol/farmacologia , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilidrazida/farmacologia , Dinoprostona/análogos & derivados , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Hidantoínas/farmacologia , Iloprosta/farmacologia , Latanoprosta , Miócitos Cardíacos/metabolismo , Prostaglandina D2/farmacologia , Prostaglandinas F Sintéticas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP1/agonistas , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP1/fisiologia , Receptores de Tromboxanos/agonistas , Receptores de Tromboxanos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA