Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Pharmacol ; 926: 175034, 2022 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-35588871

RESUMEN

Cyclosporin A (CsA) is an immunosuppressant used in transplantation patients and inflammatory diseases. CsA-induced local vasoconstriction can lead to serious side effects including nephrotoxicity and hypertension. However, the underlying mechanisms are not fully understood. Mesenteric artery rings of rats were cultured with CsA and specific inhibitors for mitogen-activating protein kinases (MAPK) and nuclear factor-κB (NF-κB) signaling pathways. A sensitive myograph recorded thromboxane (TP) receptor-mediated vasoconstriction. Protein levels of key signaling molecules were assessed by Western blotting. The results show that CsA up-regulated the TP receptor expression with the enhanced vasoconstriction in a dose- and time-dependent manner. Furthermore, the blockage of MAPKs or NF-κB activation markedly attenuated CsA-enhanced vasoconstriction and the TP receptor protein expression. Rats subcutaneously injected with CsA for three weeks showed increased blood pressure in vivo and increased contractile responses to a TP agonist ex vivo. CsA also enhanced TP receptor, as well as p-ERK1/2, p-p38, p- IκBα, p-NF-κB P65 protein levels and decreased IκBα protein expression, demonstrating that CsA induced TP receptor enhanced-vasoconstriction via activation of MAPK and NF-κB pathways. In conclusion, CsA up-regulated the expression of TP receptors via activation of MAPK and NF-κB pathways. The results may provide novel options for prevention of CsA-associated hypertension.


Asunto(s)
Ciclosporina , Hipertensión , FN-kappa B , Receptores de Tromboxano A2 y Prostaglandina H2 , Animales , Ciclosporina/farmacología , Humanos , Arterias Mesentéricas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mitógenos , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Ratas , Receptores de Tromboxanos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Tromboxanos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Toxicology ; 390: 10-21, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28826906

RESUMEN

Aluminum (Al) is a non-essential metal and a significant environmental contaminant and is associated with a number of human diseases including cardiovascular disease. We investigated the effects of Al exposure at doses similar to human dietary levels on the cardiovascular system over a 60day period. Wistar male rats were divided into two major groups and received orally: 1) Low aluminum level - rats were subdivided and treated for 60days as follows: a) Untreated - ultrapure water; b) AlCl3 at a dose of 8.3mg/kg bw for 60days, representing human Al exposure by diet; and 2) High aluminum level - rats were subdivided and treated for 42days as follows: C) Untreated - ultrapure water; d) AlCl3 at 100mg/kg bw for 42days, representing a high level of human exposure to Al. Effects on systolic blood pressure (SBP) and vascular function of aortic and mesenteric resistance arteries (MRA) were studied. Endothelium and smooth muscle integrity were evaluated by concentration-response curves to acetylcholine (ACh) and sodium nitroprusside. Vasoconstrictor responses to phenylephrine (Phe) in the presence and absence of endothelium and in the presence of the NOS inhibitor L-NAME, the potassium channels blocker TEA, the NAD(P)H oxidase inhibitor apocynin, superoxide dismutase (SOD), the non-selective COX inhibitor indomethacin and the selective COX-2 inhibitor NS 398 were analyzed. Vascular reactive oxygen species (ROS), lipid peroxidation and total antioxidant capacity, were measured. The mRNA expressions of eNOS, NAD(P)H oxidase 1 and 2, SOD1, COX-2 and thromboxane A2 receptor (TXA-2 R) were also investigated. Al exposure at human dietary levels impaired the cardiovascular system and these effects were almost the same as Al exposure at much higher levels. Al increased SBP, decreased ACh-induced relaxation, increased response to Phe, decreased endothelial modulation of vasoconstrictor responses, the bioavailability of nitric oxide (NO), the involvement of potassium channels on vascular responses, as well as increased ROS production from NAD(P)H oxidase and contractile prostanoids mainly from COX-2 in both aorta and mesenteric arteries. Al exposure increased vascular ROS production and lipid peroxidation as well as altered the antioxidant status in aorta and MRA. Al decreased vascular eNOS and SOD1 mRNA levels and increased the NAD(P)H oxidase 1, COX-2 and TXA-2 R mRNA levels. Our results point to an excess of ROS mainly from NAD(P)H oxidase after Al exposure and the increased vascular prostanoids from COX-2 acting in concert to decrease NO bioavailability, thus inducing vascular dysfunction and increasing blood pressure. Therefore, 60-day chronic exposure to Al, which reflects common human dietary Al intake, appears to pose a risk for the cardiovascular system.


Asunto(s)
Compuestos de Aluminio/toxicidad , Presión Sanguínea/efectos de los fármacos , Cloruros/toxicidad , Ciclooxigenasa 2/metabolismo , Dieta , Endotelio Vascular/efectos de los fármacos , Hipertensión/inducido químicamente , NADH NADPH Oxidorreductasas/metabolismo , Vasoconstricción/efectos de los fármacos , Cloruro de Aluminio , Animales , Inhibidores de la Ciclooxigenasa 2/farmacología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Humanos , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Masculino , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Medición de Riesgo , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Factores de Tiempo
3.
Pharmacol Rep ; 68(3): 620-6, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26991376

RESUMEN

BACKGROUND: The aim of the present study was to observe the concomitant activation of nitric oxide (NO) and endothelium-derived hyperpolarizing factor (EDHF) pathways by TRPV4 channel agonist GSK1016790A in the rat pulmonary artery and explore the mechanism by which NO synthase inhibition attenuates EDHF-mediated relaxation in endothelium-intact rat pulmonary artery. METHODS: Tension experiments were conducted on the pulmonary artery from male Wistar rats. RESULTS: TRPV4 channel agonist GSK1016790A (GSK) caused concentration-dependent relaxation (Emax 86.9±4.6%; pD2 8.7±0.24) of the endothelium-intact rat pulmonary artery. Combined presence of apamin and TRAM-34 significantly attenuated the relaxation (Emax 61.1±6.0%) to GSK. l-NAME (100µM) significantly attenuated (8.2±2.9%) the relaxation response to GSK that was resistant to apamin plus TRAM-34. However, presence of ICI192605 or furegrelate alongwith l-NAME revealed the GSK-mediated EDHF-response (Emax of 28.5±5.2%; Emax 24.5±4.3%) in this vessel, respectively. Further, these two TxA2 modulators (ICI/furegrelate) alongwith l-NAME had no effect on SNP-induced endothelium-independent relaxation in comparison to l-NAME alone. This EDHF-mediated relaxation was sensitive to inhibition by K(+) channel blockers apamin and TRAM-34 or 60mMK(+) depolarizing solution. Further, combined presence of apamin and TRAM-34 in U46619 pre-contracted pulmonary arterial rings significantly reduced the maximal relaxation (Emax 71.6±6.9%) elicited by GSK, but had no effect on the pD2 (8.1±0.03) of the TRPV4 channel agonist in comparison to controls (Emax, 92.4±4.3% and pD2, 8.3±0.06). CONCLUSION: The present study suggests that NO and EDHF are released concomitantly and NO synthase inhibition attenuates GSK-induced EDHF response through thromboxane pathway in the rat pulmonary artery.


Asunto(s)
Factores Biológicos/fisiología , Leucina/análogos & derivados , Arteria Pulmonar/fisiología , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología , Sulfonamidas/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Apamina/farmacología , Benzofuranos/farmacología , Dioxanos/farmacología , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Leucina/antagonistas & inhibidores , Leucina/farmacología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Potasio/farmacología , Pirazoles/farmacología , Ratas , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Sulfonamidas/antagonistas & inhibidores , Canales Catiónicos TRPV/agonistas , Vasodilatación/efectos de los fármacos
4.
J Invest Dermatol ; 135(12): 3153-3162, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26288351

RESUMEN

The pathogenesis of scleroderma (SSc) includes components of autoimmunity, vascular dysfunction, and accumulation of extracellular matrix. 8-isoprostane, an oxidized lipid created by oxidative stress, activates the thromboxane A2 receptor (TXAR) and the Rho-associated kinase (ROCK) pathway. In this study, we determined whether the TXAR was activated by 8-isoprostane in SSc endothelial cells (ECs) and whether this pathway inhibited VEGF-induced angiogenesis. Elevated 8-isoprostane was observed in plasma and conditioned media from SSc patients. SSc-conditioned media inhibited EC tube formation, whereas addition of vitamin E, by reducing 8-isoprostane, increased tube formation. VEGF did not induce angiogenesis in SSc ECs, but vitamin E or TXAR inhibition restored its effect. The expression of TXAR, RhoA, and ROCK1/2 was elevated in SSc ECs. ROCK activity and 8-isoprostane-induced ROCK activation were significantly higher in SSc ECs, whereas VEGF had no effect. The hyper-activation of the TXAR leads to inhibition of VEGF-induced angiogenesis, as inhibition of the TXAR pathway results in a blockade of 8-isoprostane-induced ROCK activation and restoration of VEGF activity. These results suggest that the TXAR pathway has a crucial role in angiogenesis and that 8-isoprostane is not just a by-product of oxidative stress but also has a significant role in the impaired angiogenesis that characterizes SSc.


Asunto(s)
Dinoprost/análogos & derivados , Neovascularización Fisiológica , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología , Esclerodermia Sistémica/fisiopatología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adulto , Colágeno , Dinoprost/farmacología , Combinación de Medicamentos , Células Endoteliales/fisiología , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Laminina , Masculino , Persona de Mediana Edad , Estrés Oxidativo , Fosforilación , Proteoglicanos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Quinasas Asociadas a rho/fisiología
5.
Cardiovasc Res ; 107(4): 509-21, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25935870

RESUMEN

AIMS: Thromboxane A2 (TXA2) induces platelet adhesion through thromboxane prostanoid (TP) receptor. Platelets contain many pro-angiogenic factors and are recruited to the site of vascular injury. However, the cellular and molecular mechanisms of platelet-dependent angiogenesis, especially the involvement of TP signalling, have not been fully elucidated. The present study hypothesized that TP-dependent platelet adhesion would contribute to angiogenesis in a mouse hindlimb ischaemic model. METHODS AND RESULTS: Blood flow recovery was suppressed by the TXA2 receptor antagonist (S-1452) and the TXA2 synthase inhibitor (OKY-046) compared with control mice. TP knockout mice (TP(-/-)) showed delayed blood flow recovery from ischaemia and impaired angiogenesis compared with wild-type (WT) mice and prostacyclin receptor knockout mice (IP(-/-)). Selective platelet adhesion to ischaemic endothelial cells (ECs) via P-selectin was identified in WT and IP(-/-), but not in TP(-/-), via in vivo microscopy. IF analysis showed that P-selectin glycoprotein ligand-1 (PSGL-1) co-localized with endothelial CD31 in ischaemic muscle in WT and IP(-/-) but not diminished in TP(-/-). Platelet-rich plasma levels of stromal cell-derived factor-1 and VEGF were increased after ischaemia in WT, and suppressed by antibody against P-selectin in WT but not in TP(-/-). Furthermore, the blood flow recovery was suppressed by neutralizing antibodies against VEGF or C-X-C chemokine receptor type 4 in WT and IP(-/-) but not in TP(-/-). CONCLUSION: These results indicated that TP signalling facilitates ischaemia-induced angiogenesis via P-selectin-mediated platelet adhesion to PSGL-1 on the ECs at ischaemic sites and the supply of pro-angiogenic factors by the adherent platelets.


Asunto(s)
Plaquetas/efectos de los fármacos , Células Endoteliales/metabolismo , Isquemia Miocárdica/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Tromboxano A2/farmacología , Animales , Plaquetas/citología , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones Noqueados , Adhesividad Plaquetaria/genética , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
6.
Vascul Pharmacol ; 73: 78-85, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25921923

RESUMEN

Uridine adenosine tetraphosphate (Up4A), a novel endothelium-derived vasoactive agent, is proposed to play a role in cardiovascular disorders and induces aortic contraction through activation of cyclooxygenases (COXs). We and others demonstrated that activation of A1 or A3 adenosine receptors (ARs) results in vascular contraction via thromboxane (TX) A2 production. However, the mechanisms of Up4A-induced vascular contraction in mouse aorta are not understood. We hypothesize that Up4A-induced aortic contraction is through COX-derived TXA2 production, which requires activation of A1 and/or A3AR. Concentration responses to Up4A were conducted in isolated aorta. The TXB2 production, a metabolite of TXA2, was also measured. Up4A (10(-9)-10(-5)M) produced a concentration-dependent contraction >70%, which was markedly attenuated by COX and COX1 but not by COX2 inhibition. Notably, Up4A-induced aortic contraction was blunted by both TX synthase inhibitor ozagrel and TXA2 receptor (TP) antagonist SQ29548. Surprisingly, A3AR deletion had no effect on Up4A-induced contraction. Moreover, A1AR deletion or antagonism as well as A1/A3AR deletion potentiated Up4A-induced aortic contraction, suggesting a vasodilator influence of A1AR. In contrast, non-selective purinergic P2 receptor antagonist PPADS significantly blunted Up4A-induced aortic contraction to a similar extent as selective P2X1R antagonist MRS2159, the latter of which was further reduced by addition of ozagrel. Endothelial denudation almost fully attenuated Up4A-induced contraction. Furthermore, Up4A (3µM) increased TXB2 formation, which was inhibited by either MRS2159 or ozagrel. In conclusion, Up4A-induced aortic contraction depends on activation of TX synthase and TP, which partially requires the activation of P2X1R but not A1 or A3AR through an endothelium-dependent mechanism.


Asunto(s)
Aorta/efectos de los fármacos , Fosfatos de Dinucleósidos/farmacología , Agonistas Purinérgicos/farmacología , Receptores Purinérgicos P2X1/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Animales , Aorta/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Adenosina A1/genética , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A3/genética , Receptor de Adenosina A3/metabolismo , Receptores Purinérgicos P2X1/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Transducción de Señal/efectos de los fármacos , Tromboxano A2/metabolismo , Tromboxano B2/metabolismo , Tromboxano-A Sintasa/antagonistas & inhibidores , Tromboxano-A Sintasa/metabolismo
7.
Chin J Nat Med ; 13(1): 30-40, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25660286

RESUMEN

The present study was designed to target fish for potential bioactive components contained in a Huang Lian Jie Du decoction (HLJDD) and identify the underlying mechanisms of action for the treatment of sepsis at the molecular level. he bioactive components database of HLJDD was constructed and the sepsis-associated targets were comprehensively investigated. The 3D structures of the PAFR and TXA2R proteins were established using the homology modelling (HM) method, and the molecular effects for sepsis treatment were analysed by comparing the bioactive components database and the sepsis targets using computational biology methods. The results of the screening were validated with biological testing against the human oral epidermal carcinoma cell line KB in vitro. We found that multiple bioactive compounds contained in the HLJDD interacted with multiple targets. We also predicted the promising compound leads for sepsis treatment, and the first 28 compounds were characterized. Several compounds, such as berberine, berberrubine and epiberberine, dose-dependently inhibited PGE2 production in human KB cells, and the effects were similar in the presence or absence of TPA. This study demonstrates a novel approach to identifying natural chemical compounds as new leads for the treatment of sepsis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Berberina/farmacocinética , Medicamentos Herbarios Chinos/farmacocinética , Sepsis/tratamiento farmacológico , Berberina/análogos & derivados , Dinoprostona/biosíntesis , Medicamentos Herbarios Chinos/química , Humanos , Células KB , Glicoproteínas de Membrana Plaquetaria/efectos de los fármacos , Transporte de Proteínas , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Sepsis/metabolismo , Acetato de Tetradecanoilforbol/farmacocinética
8.
Hypertension ; 64(3): 551-6, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24914192

RESUMEN

Cyclooxygenase inhibitors decrease renal blood flow in settings with decreased effective circulating volume. The present study examined the hypothesis that prostaglandins, prostaglandin E2 (PGE2) and prostacyclin (PGI2), induce relaxation of human intrarenal arteries through PGE2-EP and PGI2-IP receptors. Intrarenal arteries were microdissected from human nephrectomy samples (n=53, median diameter ≈362 µm, 88% viable, 76% relaxed in response to acetylcholine). Rings were suspended in myographs to record force development. In vessels with K(+)-induced tension (EC70: -log [mol/L]=1.36±0.03), PGE2 and PGI2 induced concentration-dependent relaxation (-log EC50: PGE2=7.1±0.3 and PGI2=7.7). The response to PGE2 displayed endothelium dependence and desensitization. Relaxation by PGE2 was mimicked by an EP4 receptor agonist (CAY10598, EC50=6.7±0.2). The relaxation after PGI2 was abolished by an IP receptor antagonist (BR5064, 10(-8) mol/L). Pretreatment of quiescent arteries with PGE2 for 5 minutes (10(-6) mol/L) led to a significant right shift of the concentration-response to norepinephrine (EC50 from 6.6±0.1-5.9±0.1). In intrarenal arteries with K(+)-induced tone, PGE2 and PGI2 at 10(-5) mol/L elicited increased tension. This was abolished by thromboxane receptor (TP) antagonist (S18886, 10(-6) mol/L). A TP agonist (U46619, n=6) evoked tension (EC50=8.1±0.2) that was inhibited by S18886. Polymerase chain reaction and immunoblotting showed EP4, IP, and TP receptors in intrarenal arteries. In conclusion, PGE2 and PGI2 may protect renal perfusion by activating cognate IP and EP4 receptors associated with smooth muscle cells and endothelium in human intrarenal arteries and contribute to increased renal vascular resistance at high pathological concentrations mediated by noncognate TP receptor.


Asunto(s)
Dinoprostona/farmacología , Epoprostenol/farmacología , Receptores de Epoprostenol/fisiología , Subtipo EP4 de Receptores de Prostaglandina E/fisiología , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología , Arteria Renal/fisiología , Vasoconstricción/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Norepinefrina/farmacología , Cloruro de Potasio/farmacología , Receptores de Epoprostenol/efectos de los fármacos , Subtipo EP4 de Receptores de Prostaglandina E/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Arteria Renal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Resistencia Vascular/fisiología , Vasoconstricción/fisiología , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos
9.
Am J Physiol Heart Circ Physiol ; 304(3): H382-92, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23203967

RESUMEN

Arachidonic acid (AA) metabolites mediate endothelium-dependent relaxation in many vascular beds. Previously, we identified the major AA 12/15-lipoxygenase (12/15-LO) metabolite of mouse arteries as 12-hydroxyeicosatetraenoic acid (12-HETE). The goal was to determine the stereospecific configuration of mouse vascular 12-HETE and characterize the role of 12-HETE stereoisomers in the regulation of vascular tone. Using normal, reverse phase, and chiral HPLC, the stereospecific configuration was identified as 12(S)-HETE. 12(S)-HETE relaxed U46619-, carbocyclic thromboxane A(2)-, PGF(2α)-, and 8-iso PGF(2α)-preconstricted mesenteric arteries, but not phenylephrine-preconstricted arteries. 12(R)-HETE was more potent than 12(S)-HETE in relaxing U46619-preconstricted mouse arteries (maximum relaxations = 91.4 ± 2.7% and 71.8 ± 5.9%, respectively). Neither 12-HETE isomer caused constriction. Pretreatment with 12(S)- or 12(R)-HETE (1 µM) inhibited constrictions to U46619 but not phenylephrine. To investigate the role of thromboxane A(2) (TP) receptors in 12-HETE vascular actions, [(3)H]SQ29548 radioligand binding studies were performed in mouse platelets. U46619, 12(R)-HETE, and 12(S)-HETE displaced [(3)H]SQ29548 binding with IC(50)s of 0.07, 0.32, and 1.73 µM, respectively. Both 12(S)- and 12(R)-HETE inhibited intracellular calcium increases induced by U46619 (10 nM) in HEK293 cells overexpressing TP(α) receptor (65.5% and 45.1%, respectively) and coexpressing prostacyclin (IP) and TP(α) receptors (58.0% and 27.1%, respectively). The LO inhibitor NDGA (10 µM) reduced AA relaxations in arteries preconstricted with U46619 but not phenylephrine. These results indicate that exogenous and endogenous 12(S)-HETE relax mouse mesenteric arteries that are preconstricted with thromboxane agonists. These 12(S)-HETE relaxations are mediated by TP receptor competitive inhibition and inhibition of TP agonist-induced increases in intracellular calcium.


Asunto(s)
Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacología , Endotelio Vascular/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Receptores de Tromboxanos/antagonistas & inhibidores , Vasodilatadores/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Ácido Araquidónico/metabolismo , Plaquetas/efectos de los fármacos , Western Blotting , Compuestos Bicíclicos Heterocíclicos con Puentes , Señalización del Calcio/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Ácidos Grasos Insaturados , Células HEK293 , Humanos , Hidrazinas/farmacología , Contracción Isométrica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Leucotrieno B4/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Vasoconstrictores/farmacología
10.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L140-51, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21984570

RESUMEN

Actions of thromboxane (TXA(2)) to alter airway resistance were first identified over 25 years ago. However, the mechanism underlying this physiological response has remained largely undefined. Here we address this question using a novel panel of mice in which expression of the thromboxane receptor (TP) has been genetically manipulated. We show that the response of the airways to TXA(2) is complex: it depends on expression of other G protein-coupled receptors but also on the physiological context of the signal. In the healthy airway, TXA(2)-mediated airway constriction depends on expression of TP receptors by smooth muscle cells. In contrast, in the inflamed lung, the direct actions of TXA(2) on smooth muscle cell TP receptors no longer contribute to bronchoconstriction. Instead, in allergic lung disease, TXA(2)-mediated airway constriction depends on neuronal TP receptors. Furthermore, this mechanistic switch persists long after resolution of pulmonary inflammation. Our findings demonstrate the powerful ability of lung inflammation to modify pathways leading to airway constriction, resulting in persistent changes in mechanisms of airway reactivity to key bronchoconstrictors. Such alterations are likely to shape the pathogenesis of asthmatic lung disease.


Asunto(s)
Bronquios/inervación , Broncoconstricción , Miocitos del Músculo Liso , Neuronas Aferentes , Receptores de Tromboxanos/metabolismo , Tromboxano A2/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Resistencia de las Vías Respiratorias/efectos de los fármacos , Animales , Asma/patología , Bronquios/metabolismo , Broncoconstricción/efectos de los fármacos , Broncoconstricción/fisiología , Células Cultivadas , Hipersensibilidad/patología , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Neumonía/fisiopatología , Receptores de Tromboxanos/genética , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Sistema Respiratorio/metabolismo , Sistema Respiratorio/patología , Tromboxano A2/análogos & derivados , Vasoconstrictores/farmacología
11.
Naunyn Schmiedebergs Arch Pharmacol ; 381(4): 349-60, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20198363

RESUMEN

Intravenous (i.v.) injection of the endocannabinoid anandamide induces triphasic cardiovascular responses, including a pressor effect mediated via unknown central and peripheral mechanism(s). The aim of the present study was to determine the central mechanism(s) responsible for the pressor response to anandamide. For this purpose, the influence of antagonists at thromboxane A(2) TP (sulotroban, daltroban, SQ 29548), NMDA (MK-801) and beta(2)-adrenergic receptors (ICI 118551) on the pressor effect induced by i.v. and intracerebroventricularly (i.c.v.) administered anandamide was examined in urethane-anaesthetized rats. Anandamide (1.5-3 micromol/kg, i.v.) or its stable analogue methanandamide (0.75 micromol/kg, i.v.) increased blood pressure by 25%. Anandamide (0.03 mumol per animal i.c.v.) caused a pure pressor effect (by 20%) but only in the presence of antagonists of CB(1) and TRPV1 receptors. The effects of cannabinoids (i.v. or i.c.v.) were diminished by i.v. daltroban, sulotroban (10 mumol/kg each), and/or SQ 29548 (1 mumol/kg). The effect of anandamide i.v. was reduced by SQ 29548 (0.02 mumol per animal i.c.v.) and by the thromboxane A(2) synthesis inhibitor furegrelate i.c.v. (1.8 micromol per animal). ICI 118551, MK-801 (1 micromol/kg i.v. each), and bilateral adrenalectomy diminished the effect of anandamide i.c.v. Sulotroban (i.v.) failed to affect the response to anandamide (i.v.) in pithed rats, and anandamide and methanandamide did not bind to TP receptors in rat platelets. The present study suggests that central beta(2)-adrenergic, NMDA and thromboxane A(2) receptors are involved in the anandamide-induced adrenal secretion of catecholamines and their pressor effect in urethane-anaesthetized rats.


Asunto(s)
Ácidos Araquidónicos/farmacología , Alcamidas Poliinsaturadas/farmacología , Receptores Adrenérgicos beta 2/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/metabolismo , Animales , Ácidos Araquidónicos/administración & dosificación , Ácidos Araquidónicos/metabolismo , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Catecolaminas/metabolismo , Relación Dosis-Respuesta a Droga , Endocannabinoides , Inyecciones Intraventriculares , Masculino , Alcamidas Poliinsaturadas/administración & dosificación , Alcamidas Poliinsaturadas/metabolismo , Ratas , Ratas Wistar , Receptores Adrenérgicos beta 2/metabolismo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos
12.
Acta Pharmacol Sin ; 31(2): 150-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20139897

RESUMEN

AIM: To investigate the potential antagonistic activity of the antidiabetic agent glybenclamide for the human platelet thromboxane A(2) receptor (abbreviated as TPR). METHODS: Platelets were obtained from healthy donors. Aggregation studies were performed in a model 700 aggregometry system. Radioactivity was counted in a Beckman LS 6000 liquid scintillation counter and calcium imaging was performed using an LS50B PerkinElmer Fluorescence Spectrometer. RESULTS: It was found that glybenclamide: 1) inhibited aggregation induced by the TPR agonist U46619 (IC(50)=2.3+/-0.31 micromol/L) and by the thromboxane A(2) precursor arachidonic acid (IC(50)=2.6+/-0.24 micromol/L); 2) displaced SQ29,548 from its binding sites on platelets; 3) lacked any detectable effects on aggregation stimulated by ADP, or the thrombin receptor activating-peptide 4; 4) blocked calcium mobilization induced by U46619, but not by ADP; and 5) failed to raise cAMP levels. CONCLUSION: The findings indicate that glybenclamide exerts inhibitory effects on platelets by interacting with TPR. Thus, glybenclamide or a rationally designed derivative has the potential to serve as an antithrombotic agent.


Asunto(s)
Gliburida/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Adenosina Difosfato/farmacología , Sitios de Unión , Plaquetas/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes , Calcio/metabolismo , AMP Cíclico/sangre , Ácidos Grasos Insaturados , Humanos , Hidrazinas/farmacología , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Conteo por Cintilación , Espectrometría de Fluorescencia
13.
Reprod Sci ; 16(4): 364-72, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19336695

RESUMEN

OBJECTIVE: The purpose of this study was to investigate the mechanisms of tension and intracellular calcium regulation following stimulation with the thromboxane A(2) receptor agonist U46619 in the left anterior descending coronary artery of fetal sheep exposed to long-term hypoxia. We hypothesized that there would be a reduction in intracellular calcium responses in long-term hypoxic left anterior descending coronary artery accompanied by an increase in calcium sensitivity of the contractile mechanism. METHODS: Pregnant sheep were kept at altitude (3820 m) from day 30 of gestation until day 140. Fetal hearts from long-term hypoxic and from a control, normoxic group were obtained and the left anterior descending coronary artery of the fetus was dissected, cleaned, and mounted in a bath (Jasco) in which tension and intracellular calcium [Ca(2+)](i), using Fura-2, could be measured simultaneously following stimulation of the thromboxane A(2) receptor with U46619. The role of intracellular calcium and the Rho kinase and protein kinase C pathways in the tension responses were investigated by maintaining intracellular calcium constant or by using the Rho kinase blocker, Y27632, or the protein kinase C blocker, GF109203-X. RESULTS: There was no difference in the tension dose-response to U46619 between the normoxic fetal and hypoxic fetal left anterior descending, although [Ca(2+)](i) was lower in the hypoxic fetal than normoxic fetal at the highest doses. When [Ca(2+)]( i) was maintained constant at baseline levels, U46619 produced the same tension dose-response in both normoxic fetal and hypoxic fetal left anterior descending as when [Ca(2+)](i) was allowed to rise. The tension response was abolished in both groups when the Rho kinase inhibitor, Y27632, was given either during or before stimulation with U46619. The protein kinase C blocker, GF109203-X, had no effect on the tension response in either group. CONCLUSIONS: Long-term hypoxia did not alter the tension response to thromboxane A(2) receptor stimulation in fetal left anterior descending. The contractions in response to U46619 were produced apparently completely by changes in calcium sensitivity through the Rho kinase pathway.


Asunto(s)
Calcio/análisis , Vasos Coronarios/embriología , Hipoxia Fetal/fisiopatología , Contracción Muscular/fisiología , Músculo Liso Vascular/fisiopatología , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Altitud , Animales , Calcio/fisiología , Vasos Coronarios/química , Vasos Coronarios/fisiopatología , Femenino , Hipoxia Fetal/etiología , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Embarazo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Receptores de Tromboxano A2 y Prostaglandina H2/agonistas , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Ovinos/embriología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/fisiología
15.
Circ Res ; 103(9): 1037-46, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18802021

RESUMEN

Isoprostanes are endogenously formed end products of lipid peroxidation. Furthermore, they are markers of oxidative stress and independent risk markers of coronary heart disease. In patients experiencing coronary heart disease, impaired angiogenesis may exacerbate insufficient blood supply of ischemic myocardium. We therefore hypothesized that isoprostanes may exert detrimental cardiovascular effects by inhibiting angiogenesis. We studied the effect of isoprostanes on vascular endothelial growth factor (VEGF)-induced migration and tube formation of human endothelial cells (ECs), and cardiac angiogenesis in vitro as well as on VEGF-induced angiogenesis in the chorioallantoic membrane assay in vivo. The isoprostanes 8-iso-PGF(2alpha), 8-iso-PGE(2), and 8-iso-PGA(2) inhibited VEGF-induced migration, tube formation of ECs, and cardiac angiogenesis in vitro, as well as VEGF-induced angiogenesis in vivo via activation of the thromboxane A(2) receptor (TBXA2R): the specific TBXA2R antagonists SQ-29548, BM 567, and ICI 192,605 but not the thromboxane A(2) synthase inhibitor ozagrel blocked the effect of isoprostanes. The isoprostane 8-iso-PGA(2) degraded into 2 biologically active derivatives in vitro, which also inhibited EC tube formation via the TBXA2R. Moreover, short hairpin RNA-mediated knockdown of the TBXA2R antagonized isoprostane-induced effects. In addition, Rho kinase inhibitor Y-27632 reversed the inhibitory effect of isoprostanes and the thromboxane A(2) mimetic U-46619 on EC migration and tube formation. Finally, the various isoprostanes exerted a synergistic inhibitory effect on EC tube formation. We demonstrate for the first time that isoprostanes inhibit angiogenesis via activation of the TBXA2R. By this mechanism, isoprostanes may contribute directly to exacerbation of coronary heart disease and to capillary rarefaction in disease states of increased oxidative stress.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Movimiento Celular , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Isoprostanos/metabolismo , Neovascularización Fisiológica , Estrés Oxidativo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Actinas/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Vasos Coronarios/efectos de los fármacos , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Dioxanos/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Grasos Insaturados , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Hidrazinas/farmacología , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Prostaglandinas A/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Fibras de Estrés/metabolismo , Compuestos de Sulfonilurea/farmacología , Técnicas de Cultivo de Tejidos
16.
Eur J Pharmacol ; 591(1-3): 237-42, 2008 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-18586024

RESUMEN

Increased intrahepatic resistance is one of the major characteristics of cirrhotic liver, in which extravascular cells including liver myofibroblasts (MFs) abnormally contract. Although several studies provided evidence that various prostaglandins (PG) are involved in liver cirrhosis, the role of PGD(2) remains unknown. In this study, we investigated the effect of PGD(2) on the contractile properties of liver MFs. Cultured rat liver MFs were used at passages 4-7. A collagen gel contraction assay was used for the evaluation of the MFs contraction. mRNA expression was assessed by semi-quantitative RT-PCR. Intracellular Ca(2+) concentrations ([Ca(2+)](i)) were measured by monitoring the fluorescence intensity of fura-2. PGD(2) (1-10 microM) induced liver MF contraction in a dose-dependent manner with [Ca(2+)](i) elevation. Pretreatment with 300 nM LaCl(3), a nonselective Ca(2+) channel blocker abolished the 10 microM PGD(2)-induced MFs contraction. RT-PCR revealed that three distinct PGD(2) responsive receptors, prostanoid DP receptor, chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) and thromboxane A(2) receptor (prostanoid TP receptor), were expressed in liver MFs. While prostanoid DP receptor agonist and CRTH2 agonist didn't induce contraction, 0.01-1 microM U46619 (11alpha, 9alpha-epoxymethano-PGH(2), prostanoid TP receptor agonist) caused robust contraction with [Ca(2+)](i) elevation. Furthermore, pretreatment with prostanoid TP receptor antagonists ramatroban (1 microM) or SQ29548 ([1S-[1alpha, 2alpha(Z), 3alpha, 4alpha]]-7-[3-[[2-[(phenyl amino)carbonyl]hydrazino]methyl]-7-oxabicyclo[2.2.1]hept-2-yl]-5-heptenoic acid, 1 microM) completely suppressed PGD(2)-induced contraction and [Ca(2+)](i) elevation. Additionally, we observed that BW245C (1-10 microM) decreased basal MF contraction. These results suggest that PGD(2) induces rat liver MF contraction with an increase in [Ca(2+)](i) through prostanoid TP receptor.


Asunto(s)
Contracción Muscular/efectos de los fármacos , Prostaglandina D2/farmacología , Receptores Inmunológicos/efectos de los fármacos , Receptores de Prostaglandina/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Animales , Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Prostaglandina D2/administración & dosificación , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Cancer Res ; 68(1): 115-21, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18172303

RESUMEN

Thromboxane A(2) (TxA(2)) is a prostanoid formed by thromboxane synthase using the cyclooxygenase product prostaglandin H(2) as the substrate. Previously, increased expression of thromboxane synthase was found in prostate tumors, and tumor cell motility was attenuated by inhibitors of thromboxane synthase. This study was undertaken to elucidate how tumor motility is regulated by TxA(2). Here, we report that human prostate cancer cells express functional receptors for TxA(2) (TP). Ligand binding assay found that PC-3 cells binded to SQ29548, a high-affinity TP antagonist, in a saturable manner with K(d) of 3.64 nmol/L and B(max) of 120.4 fmol per million cells. Treatment of PC-3 cells by U46619, a TP agonist, induced PC-3 cell contraction, which was blocked by pretreatment with the TP antagonist SQ29548 or pinane TxA(2). The migration of prostate cancer cells was significantly inhibited either by sustained activation of TP or by blockade of TP activation, suggesting that TP activation must be tightly controlled during cell migration. Further studies found that small GTPase RhoA was activated by TP activation, and pretreatment of PC-3 cells with Y27632, a Rho kinase (ROCK) inhibitor, blocked U46619-induced cell contraction. A dominant-negative mutant of RhoA also blocked U46619-induced cell contraction. Taken together, the data suggest that TPs are expressed in prostate cancer and activation of TPs regulates prostate cancer cell motility and cytoskeleton reorganization through activation of Rho.


Asunto(s)
Carcinoma/patología , Movimiento Celular , Neoplasias de la Próstata/patología , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Amidas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes , Carcinoma/química , Carcinoma/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Ácidos Grasos Insaturados , Humanos , Hidrazinas/farmacología , Ligandos , Masculino , Neoplasias de la Próstata/química , Neoplasias de la Próstata/metabolismo , Piridinas/farmacología , Receptores de Tromboxano A2 y Prostaglandina H2/análisis , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Proteína de Unión al GTP rhoA/análisis , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
18.
Am J Physiol Renal Physiol ; 294(1): F120-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17942572

RESUMEN

Early manifestations of kidney disease occur in atherosclerosis and activation of TP (thromboxane A(2)) receptors is implicated in atherosclerotic, diabetes, and renal diseases. The purpose of the present study was to analyze, in isolated, perfused mouse kidneys, the participation of TP receptors in renal vasoconstrictions and vasodilatations. In kidneys, taken from wild-type C57BL6, apolipoprotein E-deficient (ApoE-KO) and diabetic ApoE-KO mice, changes in perfusion pressure were recorded. Constrictions to TP receptor ligands U 46619, arachidonic acid, PGH(2), and 8-iso-PGF(2alpha), but not those to angiotensin II, endothelin, or norepinephrine, were inhibited by the selective TP receptor antagonist Triplion (S 18886; 10 nM). Acetylcholine and prostacyclin evoked biphasic responses during methoxamine constrictions; the constrictor part was blocked by Triplion. In ApoE-KO mouse kidneys, compared with C57BL6, a specific decrease in norepinephrine response and no modification in dilator responses were observed. In diabetic ApoE-KO mouse kidneys, constrictions to U 46619 and those to 8-iso-PGF(2alpha) were significantly and selectively augmented, without modification in the expression of the TP receptor, and again without any significant change in vasodilator activity. Thus TP receptors are functional, and their activation is not involved in norepinephrine, endothelin, and angiotensin II vasoconstrictions but is implicated in the unusual vasoconstrictions to acetylcholine and prostacyclin. Increased responsiveness of TP receptors occurs in diabetic ApoE-KO mouse kidneys. Thus early changes in TP receptor-mediated vasoconstrictor activity may participate in the development of kidney disease in atherosclerosis and diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Riñón/irrigación sanguínea , Riñón/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Vasoconstricción/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Acetilcolina/farmacología , Animales , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Diabetes Mellitus Experimental/fisiopatología , Dinoprost/análogos & derivados , Dinoprost/farmacología , Modelos Animales de Enfermedad , Epoprostenol/farmacología , Masculino , Metoxamina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Naftalenos/farmacología , Propionatos/farmacología , Receptores de Tromboxano A2 y Prostaglandina H2/antagonistas & inhibidores , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Estreptozocina , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
19.
J Hypertens ; 25(4): 861-70, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17351380

RESUMEN

BACKGROUND: Inflammatory processes may play a pivotal role in the pathogenesis of cerebrovascular injury in salt-loaded, stroke-prone, spontaneously hypertensive rats (SHRSP). Thromboxane A2 (TP) receptor stimulation by 8-iso-prostaglandin F2alpha (8-iso-PGF2alpha) is involved in the process of vascular inflammation. OBJECTIVE: In the present study, we examined the involvement of TP receptor in the development of cerebrovascular damage in salt-loaded SHRSP. METHODS: Nine-week-old SHRSP were fed a 0.4% NaCl or a 4% NaCl diet with or without ONO-8809 treatment (a TP receptor antagonist) for 5 weeks. Blood pressure, mortality, and the parameters of cerebrovascular inflammation and damage were compared between the groups. Moreover, we examined the effect of 8-iso-PGF2alpha infusion on cerebrovascular injury of SHRSP. RESULTS: High salt intake in SHRSP significantly increased blood-brain barrier impairment and early mortality, which were suppressed by ONO-8809 treatment independent of changes in blood pressure. Salt loading also significantly increased superoxide production in basilar arteries of SHRSP, which was suppressed by ONO-8809 treatment. Macrophage accumulation and matrix metalloproteinase-9 (MMP-9) activity in the stroke-negative area in the contralateral cerebral cortex to the stroke lesion of salt-loaded SHRSP and 8-iso-PGF2alpha-treated SHRSP were significantly reduced by ONO-8809 treatment. The ONO-8809 treatment prevented thinning of the vessel layer in cerebral arterioles of salt-loaded SHRSP and 8-iso-PGF2alpha-treated SHRSP. CONCLUSIONS: These results suggest that TP receptor stimulation by 8-iso-PGF2alpha may involve salt loading-induced stroke through activation of cerebrovascular inflammation and damage.


Asunto(s)
Dinoprost/análogos & derivados , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Vasoconstrictores/farmacología , Análisis de Varianza , Animales , Arteria Basilar/efectos de los fármacos , Arteria Basilar/metabolismo , Biomarcadores/sangre , Presión Sanguínea/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Compuestos Bicíclicos con Puentes/farmacología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Quimiocina CCL2/sangre , Dinoprost/farmacología , Modelos Animales de Enfermedad , Ácidos Grasos Monoinsaturados/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratas , Ratas Endogámicas SHR , Receptores de Tromboxano A2 y Prostaglandina H2/antagonistas & inhibidores , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Accidente Cerebrovascular/mortalidad , Accidente Cerebrovascular/fisiopatología , Superóxidos/metabolismo , Factores de Tiempo , Túnica Media/efectos de los fármacos , Túnica Media/metabolismo , Túnica Media/fisiopatología
20.
Eur J Obstet Gynecol Reprod Biol ; 130(1): 51-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16386349

RESUMEN

OBJECTIVE: To study if spontaneous contractions augmented by proteinase-activated receptor-2 (PAR-2)-activating peptide serine-leucine-isoleucine-glycine-arginine-leucine (SLIGRL) involve coactivation of membrane chemoceptors and are associated with expression of PAR-2 mRNA in non-pregnant and pregnant rat myometrium. MATERIALS AND METHODS: Non-pregnant, mid-pregnant, and late pregnant rat uterine horn and small intestine segments were snap-frozen in liquid nitrogen to determine PAR-2 mRNA levels by real time polymerase chain reaction (PCR). Uterine rings were used for isometric tension recording. Effect of SLIGRL (0.1 mM) on spontaneous contractions before and after exposure to ibuprofen (cyclooxygenase inhibitor, 1.0 microM), SQ-29548 (thromboxane A(2) receptor inhibitor, 1.0 microM), ketotifen (histamine 1 receptor inhibitor, 10 microM), WEB-2170BS (platelet-activating factor (PAF) receptor inhibitor, 10 microM), atropine (muscarinic receptor inhibitor, 0.1 microM), or ketanserin (serotonin receptor inhibitor, 10 microM) were compared. Paired t-test and one-way ANOVA followed by Dunnett's or Newman-Keuls post hoc tests were used for statistical analysis when appropriate. SIGNIFICANCE: P<0.05. RESULTS: The agents did not significantly affect time-associated decay in spontaneous contractile activity in any group of the tissues. Activation of spontaneous contractions induced by SLIGRL in non-pregnant rat myometrium did not involve coactivation of membrane chemoceptors, while in mid-pregnant rat myometrium coactivation of prostanoid, histamine, and serotonin receptors and in late pregnant rat myometrium coactivation of thromboxane receptors was noted. Expression of PAR-2 mRNA was similar in non-pregnant, mid-pregnant, and late pregnant rat myometrium. CONCLUSIONS: Expression of PAR-2 in rat myometrium is not dependent on gestational age. Stimulation of PAR-2 is associated with production/release of cyclooxygenase pathway product(s) activating thromboxane/prostaglandin H2 receptors, partial involvement of histamine H1 receptors and serotonin receptors in midpregnancy and thromboxane A2/prostaglandin H2 receptors in late pregnancy.


Asunto(s)
Redes y Vías Metabólicas/fisiología , Miometrio/efectos de los fármacos , Oligopéptidos/fisiología , Receptor PAR-2/efectos de los fármacos , Contracción Uterina/efectos de los fármacos , Animales , Atropina/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes , Ácidos Grasos Insaturados , Femenino , Hidrazinas/farmacología , Ibuprofeno/farmacología , Técnicas In Vitro , Ketanserina/farmacología , Cetotifen/farmacología , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Embarazo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor PAR-2/antagonistas & inhibidores , Receptor PAR-2/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Histamínicos H1/efectos de los fármacos , Receptores Histamínicos H1/fisiología , Receptores de Serotonina/efectos de los fármacos , Receptores de Serotonina/fisiología , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...