Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell Endocrinol ; 474: 105-118, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29496567

RESUMEN

Islet transplantation is a valid therapeutic option for type 1 diabetes treatment. However, in this procedure one of the major problems is the oxidative stress produced during pancreatic islet isolation. The aim of our study was to evaluate potential protective effects of L-carnosine and its isomer D-carnosine against oxidative stress. We evaluated the carnosine effect on cell growth, cell death, insulin production, and the main markers of oxidative stress in rat and murine stressed beta cell lines as well as in human pancreatic islets. Both isomers clearly inhibited hydrogen peroxide induced cytotoxicity, with a decrease in intracellular reactive oxygen and nitrogen species, prevented hydrogen peroxide induced apoptosis/necrosis, nitrite production, and reduced glucose-induced insulin secretion. In addition, NF-κB expression/translocation and nitrated protein induced in stressed cells was significantly reduced. Furthermore, both isomers improved survival and function, and decreased reactive oxygen and nitrogen species, and nitrite and nitrotyrosine production in human islets cultured for 1, 3, and 7 days. These results seem to indicate that both L and D-carnosine have a significant cytoprotective effect by reducing oxidative stress in beta cell lines and human islets, suggesting their potential use to improve islet survival during the islet transplantation procedure.


Asunto(s)
Carnosina/farmacología , Citoprotección/efectos de los fármacos , Células Secretoras de Insulina/patología , Estrés Oxidativo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biomarcadores/metabolismo , Carnosina/química , Línea Celular , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Humanos , Peróxido de Hidrógeno/toxicidad , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Óxido Nitroso/metabolismo , Sustancias Protectoras/farmacología , Ratas , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción ReIA/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
2.
Am J Physiol Heart Circ Physiol ; 306(2): H197-205, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24213612

RESUMEN

Pulmonary arterial hypertension is a severe progressive disease with marked morbidity and high mortality in which right ventricular (RV) failure is the major cause of death. Thus knowledge of the mechanisms underlying RV failure is an area of active interest. Previous studies suggest a role of NADPH oxidase in cardiomyocyte dysfunction in the left heart. Here we postulate that acute pressure overload induced by pulmonary artery banding (PAB) leads to a Nox4-initiated increase in reactive oxygen species (ROS) in mouse RV that may lead to feed-forward induction of Nox2. To test our hypothesis, ROS production was measured in RV and left ventricle homogenates. The data show that hydrogen peroxide (H2O2), but not superoxide anion (O2(·-)), was increased in the early phases (within 6 h) of PAB in RV and that this increase was diminished by catalase and diphenyleneiodonium chloride but not by SOD, N(ω)-nitro-l-arginin methyl ester, febuxostat, or indomethacin. H2O2 production in RV was not attenuated in Nox2 null mice subjected to 6 h PAB. Moreover, we observed an upregulation of Nox4 mRNA after 1 h of PAB and an increase in mitochondrial Nox4 protein 6 h post-PAB. In contrast, we observed an increase in Nox2 mRNA 1 day post-PAB. Expression of antioxidant enzymes SOD, catalase, and glutathione peroxidase did not change, but catalase activity increased 6 h post-PAB. Taken together, these findings show a role of mitochondria-localized Nox4 in the early phase of PAB and suggest an involvement of this isozyme in early ROS generation possibly contributing to progression of RV dysfunction and failure.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Catalasa/genética , Catalasa/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba , Disfunción Ventricular
3.
Cardiovasc Res ; 97(1): 134-42, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22997161

RESUMEN

AIMS: Reactive oxygen species (ROS)-mediated intracellular signalling is well described in the vasculature, yet the precise roles of ROS in paracrine signalling are not known. Studies implicate interstitial ROS hydrogen peroxide (H(2)O(2)) in vascular disease, and plasma H(2)O(2) levels in the micromolar range are detectable in animal models and humans with hypertension. Recently, H(2)O(2) was shown to cross biological membranes of non-vascular cells via aquaporin (Aqp) water channels. Previous findings suggest that H(2)O(2) activates NADPH oxidase (Nox) enzymes in vascular cells and apoptosis signal-regulating kinase 1 (Ask1) in non-vascular cells. We hypothesized that extracellular H(2)O(2) induces smooth muscle cell (SMC) hypertrophy by a mechanism involving Aqp1, Nox1, and Ask1. METHODS AND RESULTS: Treatment of rat aortic SMCs (rASMC) with exogenous H(2)O(2) resulted in a concentration-dependent increase in Nox-derived superoxide (O(2)(•-)), determined by L-012 chemiluminescence, cytochrome c and electron paramagnetic resonance. Nox1 was verified as the source of O(2)(·-) by siRNA. Aqp1 siRNA attenuated H(2)O(2) cellular entry and H(2)O(2)-induced O(2)(•-) production. H(2)O(2) treatment increased Ask1 activation and induced rASMC hypertrophy in a Nox1-dependent mechanism. Adenoviral-dominant-negative Ask1 attenuated H(2)O(2)-induced rASMC hypertrophy and adenoviral overexpression of Ask1 augmented it. CONCLUSION: Our results demonstrate for the first time that extracellular H(2)O(2), at pathophysiological concentrations, stimulates rASMC Nox1-derived O(2)(•-), subsequent Ask1 activation and SMC hypertrophy. The data demonstrate a novel pathway by which H(2)O(2) enters vascular cells via aquaporins and activates Nox, leading to hypertrophy, and provide multiple novel targets for combinatorial therapeutics development targeting hypertrophy and vascular disease.


Asunto(s)
Acuaporina 1/metabolismo , Peróxido de Hidrógeno/farmacología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , NADH NADPH Oxidorreductasas/metabolismo , Oxidantes/farmacología , Animales , Acuaporina 1/genética , Células Cultivadas , Relación Dosis-Respuesta a Droga , Espectroscopía de Resonancia por Spin del Electrón , Activación Enzimática , Citometría de Flujo , Hipertrofia , MAP Quinasa Quinasa Quinasa 5/genética , Microscopía Confocal , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , NADPH Oxidasa 1 , Fosforilación , Interferencia de ARN , Ratas , Transducción de Señal/efectos de los fármacos , Superóxidos/metabolismo , Factores de Tiempo , Transfección
4.
Arterioscler Thromb Vasc Biol ; 32(12): 2966-73, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23087362

RESUMEN

OBJECTIVE: Although the matricellular protein thrombospondin-1 (TSP1) is highly expressed in the vessel wall in response to injury, its pathophysiological role in the development of vascular disease is poorly understood. This study was designed to test the hypothesis that TSP1 stimulates reactive oxygen species production in vascular smooth muscle cells and induces vascular dysfunction by promoting oxidative stress. METHODS AND RESULTS: Nanomolar concentrations of TSP1 found in human vascular disease robustly stimulated superoxide (O(2)(•-)) levels in vascular smooth muscle cells at both cellular and tissue level as measured by cytochrome c and electron paramagnetic resonance. A peptide mimicking the C terminus of TSP1 known to specifically bind CD47 recapitulated this response. Transcriptional knockdown of CD47 and a monoclonal inhibitory CD47 antibody abrogated TSP1-triggered O(2)(•-) in vitro and ex vivo. TSP1 treatment of vascular smooth muscle cells activated phospholipase C and protein kinase C, resulting in phosphorylation of the NADPH oxidase organizer subunit p47(phox) and subsequent Nox1 activation, leading to impairment of arterial vasodilatation ex vivo. Further, we observed that blockade of CD47 and NADPH oxidase 1 gene silencing in vivo in rats improves TSP1-induced impairment of tissue blood flow after ischemia reperfusion. CONCLUSIONS: Our data suggest a highly regulated process of reactive oxygen species stimulation and blood flow regulation promoted through a direct TSP1/CD47-mediated activation of Nox1. This is the first report, to our knowledge, of a matricellular protein acting as a ligand for NADPH oxidase activation and through specific engagement of integrin-associated protein CD47.


Asunto(s)
Antígeno CD47/fisiología , Músculo Liso Vascular/fisiología , NADH NADPH Oxidorreductasas/fisiología , Flujo Sanguíneo Regional/fisiología , Trombospondina 1/fisiología , Animales , Antígeno CD47/genética , Silenciador del Gen , Masculino , Ratones , Músculo Liso Vascular/efectos de los fármacos , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Proteína Quinasa C/fisiología , Ratas , Flujo Sanguíneo Regional/efectos de los fármacos , Transducción de Señal/fisiología , Superóxido Dismutasa/fisiología , Superóxido Dismutasa-1 , Trombospondina 1/farmacología , Fosfolipasas de Tipo C/fisiología
5.
J Cell Physiol ; 226(10): 2633-40, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21792922

RESUMEN

The molecular mechanisms underlying hypoxic pulmonary vasoconstriction (HPV) are not yet properly understood. Mitochondrial electron transport chain (ETC) and NADPH oxidase have been proposed as possible oxygen sensors, with derived reactive oxygen species (ROS) playing key roles in coupling the sensor(s) to the contractile machinery. We have recently reported that activation of neutral sphingomyelinase (nSMase) and protein kinase C ζ (PKCζ) participate in the signalling cascade of HPV. Herein, we studied the significance of nSMase in controlling ROS production rate in rat pulmonary artery (PA) smooth muscle cells and thereby HPV in rat PA. ROS production (analyzed by dichlorofluorescein and dihydroethidium fluorescence) was increased by hypoxia in endothelium-denuded PA segments and their inhibition prevented hypoxia-induced voltage-gated potassium channel (K(V) ) inhibition and pulmonary vasoconstriction. Consistently, H(2) O(2) , or its analogue t-BHP, decreased K(V) currents and induced a contractile response, mimicking the effects of hypoxia. Inhibitors of mitochondrial ETC (rotenone) and NADPH oxidase (apocynin) prevented hypoxia-induced ROS production, K(V) channel inhibition and vasoconstriction. Hypoxia induced p47(phox) phosphorylation and its interaction with caveolin-1. Inhibition of nSMase (GW4869) or PKCζ prevented p47(phox) phosphorylation and ROS production. The increase in ceramide induced by hypoxia (analyzed by immunocytochemistry) was inhibited by rotenone. Exogenous ceramide increased ROS production in a PKCζ sensitive manner. We propose an integrated signalling pathway for HPV which includes nSMase-PKCζ-NADPH oxidase as a necessary step required for ROS production and vasoconstriction.


Asunto(s)
Hipoxia/metabolismo , Hipoxia/fisiopatología , NADPH Oxidasas/metabolismo , Arteria Pulmonar/fisiología , Especies Reactivas de Oxígeno/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Vasoconstricción/fisiología , Enfermedad Aguda , Animales , Ceramidas/antagonistas & inhibidores , Ceramidas/metabolismo , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Hipoxia/enzimología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Fosforilación/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Proteína Quinasa C/metabolismo , Arteria Pulmonar/enzimología , Ratas , Ratas Wistar , Rotenona/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Desacopladores/farmacología , Vasoconstricción/efectos de los fármacos
6.
J Pharmacol Exp Ther ; 338(1): 400-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21521772

RESUMEN

Recent epidemiological data suggest that diabetes is a risk factor for pulmonary arterial hypertension. The aim of the present study was to analyze the link between type 1 diabetes and pulmonary arterial dysfunction in rats. Male Sprague-Dawley rats were randomly divided into a control group (saline) and a diabetic group (70 mg/kg streptozotocin). After 6 weeks, diabetic animals showed a down-regulation of the lung bone morphogenetic protein receptor type 2, up-regulation of 5-hydroxytryptamine (5-HT) 2A receptors and cyclooxygenase-2 (COX-2) proteins as measured by Western blot analysis, and increased contractile responses to 5-HT in isolated intrapulmonary arteries. The hyper-responsiveness to 5-HT was endothelium-independent and unaffected by inhibition of nitric-oxide synthase but prevented by indomethacin, the selective COX-2 inhibitor N-[2-(cyclohexyloxyl)-4-nitrophenyl]-methane sulfonamide (NS-398), superoxide dismutase, and the NADPH oxidase inhibitor apocynin or chronic treatment with insulin. However, diabetic rats at 6 weeks did not develop elevated right ventricular pressure or pulmonary artery muscularization, whereas a longer exposure (4 months) to diabetes induced a modest, but significant, increase in right ventricular systolic pressure. In conclusion, type 1 diabetes mellitus in rats induces a number of changes in lung protein expression and pulmonary vascular reactivity characteristic of clinical and experimental pulmonary arterial hypertension but insufficient to elevate pulmonary pressure. Our results further strengthen the link between diabetes and pulmonary arterial hypertension.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Diabetes Mellitus Tipo 1/metabolismo , Endotelio Vascular/metabolismo , Estrés Oxidativo/fisiología , Arteria Pulmonar/metabolismo , Serotonina/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Diabetes Mellitus Tipo 1/enzimología , Endotelio Vascular/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Masculino , Estrés Oxidativo/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Serotonina/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología
7.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L619-30, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19617310

RESUMEN

The increase in O(2) tension after birth is a major factor stimulating ductus arteriosus (DA) constriction and closure. Here we studied the role of the mitochondrial electron transport chain (ETC) as sensor, H(2)O(2) as mediator, and voltage-gated potassium (K(V)) channels and Rho kinase as effectors of O(2)-induced contraction in the chicken DA during fetal development. Switching from 0% to 21% O(2) contracted the pulmonary side of the mature DA (mature pDA) but had no effect in immature pDA and relaxed the aortic side of the mature DA (mature aDA). This contraction of the pDA was attenuated by inhibitors of the mitochondrial ETC and by the H(2)O(2) scavenger polyethylene glycol (PEG)-catalase. Moreover, O(2) increased reactive oxygen species (ROS) production, measured with the fluorescent probes dihydroethidium and 2',7'-dichlorofluorescein, only in mature pDA. The H(2)O(2) analog t-butyl-hydroperoxide mimicked the responses to O(2) in the three vessels. In contrast to immature pDA cells, mature pDA cells exhibited high-amplitude O(2)-sensitive potassium currents. The K(V) channel blocker 4-aminopyridine prevented the current inhibition elicited by O(2). The L-type Ca(2+) (Ca(L)) channel blocker nifedipine and the Rho kinase inhibitors Y-27632 and hydroxyfasudil induced a similar relaxation when mature pDA were stimulated with O(2) or H(2)O(2). Moreover, the sensitivity to these drugs increased with maturation. Our results indicate the presence of a common mechanism for O(2) sensing/signaling in mammalian and nonmammalian DA and favor the idea that, rather than a single mechanism, a parallel maturation of the sensor and effectors is critical for O(2) sensitivity appearance during development.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Conducto Arterial/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Oxígeno/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Western Blotting , Embrión de Pollo , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Electrofisiología , Peróxido de Hidrógeno/toxicidad , Oxidantes/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Quinasas Asociadas a rho/antagonistas & inhibidores
8.
Cardiovasc Res ; 82(2): 296-302, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19088082

RESUMEN

AIMS: The mechanisms involved in hypoxic pulmonary vasoconstriction (HPV) are not yet fully defined. The aim of the study was to determine the role of protein kinase C zeta (PKCzeta) and neutral sphingomyelinase (nSMase) in HPV. METHODS AND RESULTS: Ceramide content was measured by immunocytochemistry and voltage-gated potassium channel (KV) currents were recorded by the patch clamp technique in isolated rat pulmonary artery smooth muscle cells (PASMC). Contractile responses were analysed in rat pulmonary arteries mounted in a wire myograph. Pulmonary pressure was recorded in anesthetized open-chest rats. Protein and mRNA expression were measured by western blot and RT-PCR, respectively. We found that hypoxia increased ceramide content in PASMC which was abrogated by inhibition of nSMase, but not acid sphingomyelinase (aSMase). The hypoxia-induced vasoconstrictor response in isolated pulmonary arteries and the inhibition of KV currents were strongly reduced by inhibition of PKCzeta or nSMase but not aSMase. The nSMase inhibitor GW4869 prevented HPV in vivo. The vasoconstrictor response to hypoxia was mimicked by exogenous addition of bacterial Smase and ceramide. nSMase2 mRNA expression was approximately 10-fold higher in pulmonary compared with mesenteric arteries. In mesenteric arteries, hypoxia failed to increase ceramide but exogenous SMase induced a contractile response. CONCLUSION: nSMase-derived ceramide production and the activation of PKCzeta are early and necessary events in the signalling cascade of acute HPV.


Asunto(s)
Hipoxia/fisiopatología , Arteria Pulmonar/fisiopatología , Esfingomielina Fosfodiesterasa/fisiología , Vasoconstricción/fisiología , Enfermedad Aguda , Animales , Ceramidas/metabolismo , Modelos Animales de Enfermedad , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Técnicas de Placa-Clamp , Proteína Quinasa C/fisiología , Arteria Pulmonar/citología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Esfingomielina Fosfodiesterasa/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 295(5): L727-32, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18723759

RESUMEN

Recent data suggest that diabetes is a risk factor for pulmonary hypertension. The aim of the present study was to analyze whether diabetes induces endothelial dysfunction in pulmonary arteries and the mechanisms involved. Male Sprague-Dawley rats were randomly divided into a control (saline) and a diabetic group (70 mg/kg(-1) streptozotocin). After 6 wk, intrapulmonary arteries were mounted for isometric tension recording, and endothelial function was tested by the relaxant response to acetylcholine. Protein expression and localization were measured by Western blot and immunohistochemistry and superoxide production by dihydroethidium staining. Pulmonary arteries from diabetic rats showed impaired relaxant response to acetylcholine and reduced vasoconstrictor response to the nitric oxide (NO) synthase inhibitor L-NAME, whereas the response to nitroprusside and the expression of endothelial NO synthase remained unchanged. Endothelial dysfunction was reversed by addition of superoxide dismutase or the NADPH oxidase inhibitor apocynin. An increase in superoxide production and increased expression of the NADPH oxidase regulatory subunit p47(phox) were also found in pulmonary arteries from diabetic rats. In conclusion, the pulmonary circulation is a target for diabetes-induced endothelial dysfunction via enhanced NADPH oxidase-derived superoxide production.


Asunto(s)
Diabetes Mellitus/enzimología , Diabetes Mellitus/fisiopatología , Endotelio/enzimología , Endotelio/fisiopatología , NADPH Oxidasas/biosíntesis , Arteria Pulmonar/enzimología , Arteria Pulmonar/fisiopatología , Acetilcolina/farmacología , Animales , Diabetes Mellitus/patología , Endotelio/efectos de los fármacos , Endotelio/patología , Inducción Enzimática/efectos de los fármacos , Técnicas In Vitro , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Superóxidos/metabolismo , Vasodilatación/efectos de los fármacos
10.
Mol Pharmacol ; 72(5): 1301-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17699685

RESUMEN

Voltage-gated potassium (K(V)) channels play an essential role in regulating pulmonary artery function, and they underpin the phenomenon of hypoxic pulmonary vasoconstriction. Pulmonary hypertension is characterized by inappropriate vasoconstriction, vascular remodeling, and dysfunctional K(V) channels. In the current study, we aimed to elucidate the role of PKCzeta and its adaptor protein p62 in the modulation of K(V) channels. We report that the thromboxane A(2) analog 9,11-dideoxy-11alpha,9alpha-epoxymethano-prostaglandin F(2alpha) methyl acetate (U46619) inhibited K(V) currents in isolated mice pulmonary artery myocytes and the K(V) current carried by human cloned K(V)1.5 channels expressed in Ltk(-) cells. Using protein kinase C (PKC)zeta(-/-) and p62(-/-) mice, we demonstrate that these two proteins are involved in the K(V) channel inhibition. PKCzeta coimmunoprecipitated with K(V)1.5, and this interaction was markedly reduced in p62(-/-) mice. Pulmonary arteries from PKCzeta(-/-) mice also showed a diminished [Ca(2+)](i) and contractile response, whereas genetic inactivation of p62(-/-) resulted in an absent [Ca(2+)](i) response, but it preserved contractile response to U46619. These data demonstrate that PKCzeta and its adaptor protein p62 play a key role in the modulation of K(V) channel function in pulmonary arteries. These observations identify PKCzeta and/or p62 as potential therapeutic targets for the treatment of pulmonary hypertension.


Asunto(s)
Canal de Potasio Kv1.5/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Arteria Pulmonar/fisiología , Factores de Transcripción/metabolismo , Vasoconstricción , Animales , Señalización del Calcio , Canal de Potasio Kv1.5/metabolismo , Ratones , Ratones Mutantes , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Tromboxano A2/farmacología , Factor de Transcripción TFIIH , Factores de Transcripción/genética , Vasoconstricción/genética
11.
Cardiovasc Res ; 73(2): 424-31, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17055466

RESUMEN

OBJECTIVE: Large conductance Ca(2+)-activated K(+) channels (BKCa) regulate coronary artery tone in vivo, play a key role in blood pressure regulation, and have been suggested as novel potential drug targets in hypertension. Quercetin exerts systemic and coronary vasodilator effects in vitro and reduces blood pressure in several rat models of hypertension, and its consumption is associated with a lower mortality rate from coronary heart disease in epidemiological studies. We hypothesized that quercetin might activate BKCa channel in isolated myocytes from rat coronary arteries and that this mechanism might be involved in its coronary artery relaxant effects. METHODS: Membrane currents were measured using the whole-cell configuration of the patch-clamp technique. Contractile tension was recorded in rat coronary artery rings mounted in a myograph. RESULTS: Quercetin (>0.1 muM) increased the outward currents in the whole range of test potentials, hyperpolarized cell membranes, and increased the frequency of spontaneous transient outward currents (STOCs) carried by BKCa channels. These effects were abolished by the selective BKCa blocker iberiotoxin and by catalase. Quercetin increased dichlorofluorescein fluorescence in coronary arteries in a polyethylenglycol-catalase-sensitive manner, indicating that it increased cytosolic H(2)O(2). The membrane-permeable analogue of H(2)O(2)t-butylhydroperoxide mimicked the effects of quercetin on outward currents. The vasodilator effect of quercetin in isolated rat coronary arteries was partially inhibited by iberiotoxin. CONCLUSION: Quercetin increased BKCa currents via production of intracellular H(2)O(2). This effect is involved, at least partly, in the coronary vasodilator effects of quercetin.


Asunto(s)
Vasos Coronarios , Peróxido de Hidrógeno/metabolismo , Músculo Liso Vascular/metabolismo , Canales de Potasio/metabolismo , Quercetina/farmacología , Vasodilatadores/farmacología , 4-Aminopiridina/farmacología , Animales , Biomarcadores/análisis , Catalasa/farmacología , Frutas , Peróxido de Hidrógeno/análisis , Masculino , Músculo Liso Vascular/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Técnicas de Placa-Clamp , Péptidos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Ratas , Ratas Wistar , Verduras
12.
Circ Res ; 98(7): 931-8, 2006 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-16527989

RESUMEN

Multiple lines of evidence indicate that serotonin (5-hydroxytryptamine [5-HT]) and voltage-gated K+ (KV) channels play a central role in the pathogenesis of pulmonary hypertension (PH). We hypothesized that 5-HT might modulate the activity of KV channels, therefore establishing a link between these pathogenetic factors in PH. Here, we studied the effects of 5-HT on KV channels present in rat pulmonary artery smooth muscle cells (PASMC) and on hKV1.5 channels stably expressed in Ltk- cells. 5-HT reduced native KV and hKV1.5 currents, depolarized cell membrane, and caused a contraction of isolated pulmonary arteries. The effects of 5-HT on KV currents and contraction were markedly prevented by the 5-HT2A receptor antagonist ketanserin. Incubation with inhibitors of phospholipase C (U73122), classic protein kinase Cs (Gö6976), or tyrosine kinases (genistein and tyrphostin 23), the cholesterol depletion agent beta-cyclodextrin or concanavalin A, an inhibitor of endocytotic processes, also prevented the effects of 5-HT. In homogenates from pulmonary arteries, 5-HT2A receptors and caveolin-1 coimmunoprecipitated with KV1.5 channels, and this was increased on stimulation with 5-HT. Moreover, KV1.5 channels were internalized when cells were stimulated with 5-HT, and this was prevented by concanavalin A. These findings indicate that activation of 5-HT2A receptors inhibits native KV and hKV1.5 currents via phospholipase C, protein kinase C, tyrosine kinase, and a caveolae pathway. KV channel inhibition accounts, at least partly, for 5-HT-induced pulmonary vasoconstriction and might play a role in PH.


Asunto(s)
Caveolina 1/fisiología , Canal de Potasio Kv1.5/fisiología , Músculo Liso Vascular/fisiología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Arteria Pulmonar/fisiología , Receptor de Serotonina 5-HT2A/fisiología , Serotonina/farmacología , Animales , Caveolina 1/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Ketanserina/farmacología , Canal de Potasio Kv1.5/efectos de los fármacos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Ratas , Ratas Wistar , Receptor de Serotonina 5-HT2A/efectos de los fármacos
13.
Ann N Y Acad Sci ; 1091: 41-51, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17341601

RESUMEN

Voltage-gated potassium channels (Kv) and thromboxane A(2) (TXA(2)) have been involved in several forms of human and experimental pulmonary hypertension. We have reported that the TXA(2) analog U46619, via activation of TP receptors and PKCzeta, inhibited Kv currents in rat pulmonary artery smooth muscle cells (PASMC), increased cytosolic calcium, and induced a contractile response in isolated rat and piglet pulmonary arteries (PA). Herein, we have analyzed the role of reactive oxygen species (ROS) in this signaling pathway. In rat PA, U46619 increased dichlorofluorescein fluorescence, an indicator of intracellular hydrogen peroxide, and this effect was prevented by the NADPH oxidase inhibitor apocynin and by polyethyleneglycol-catalase (PEG-catalase, a membrane-permeable form of catalase). U46619 inhibited Kv currents in native PASMC and these effects were strongly inhibited by apocynin. The contractile responses to U46619 in isolated PA were inhibited by PEG-catalase and the NADPH oxidase inhibitors diphenylene iodonium (DPI) and apocynin. A membrane permeable of hydrogen peroxide, t-butyl hydroperoxide, also inhibited Kv currents and induced a contractile response. Activation of NADPH oxidase and the subsequent production of hydrogen peroxide are involved in the Kv channel inhibition and the contractile response induced by TP receptor activation in rat PA.


Asunto(s)
Bloqueadores de los Canales de Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Arteria Pulmonar/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Vasoconstricción/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Masculino , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Arteria Pulmonar/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/farmacología , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...