Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Biomed Pharmacother ; 144: 112372, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34794237

RESUMEN

Small- and intermediate-conductance Ca2+-activated K+ channels, KCa2.3 and KCa3.1, are involved in cellular signaling processes associated with inflammation and fibrosis. KCa2.3 and KCa3.1 are upregulated by proinflammatory cytokines and profibrotic growth factors. Cyclic AMP, which downregulates KCa2.3 and KCa3.1, is elevated by modafinil in cells; accordingly, we investigated whether modafinil exerts anti-inflammatory and anti-fibrotic responses via KCa2.3- and KCa3.1-mediated pathways in high-fat diet (HFD)- or thioacetamide-induced liver disease models in mice. Modafinil was administered orally in the form of a racemate, (R)-isomer, or (S)-isomer. We also determined whether the treatment targeted the profibrotic activity of hepatic stellate cells using immortalized human hepatic stellate cells (LX-2 cells). Modafinil improved HFD- or thioacetamide-induced changes compared to the control, leading to a reduced inflammatory response, collagen deposition, and α-smooth muscle actin expression both in vivo and in vitro. However, modafinil did not relieve HFD-induced steatosis. There were no significant differences in the effects of the (R)- and (S)-isomers of modafinil. KCa2.3 and KCa3.1 were upregulated and catalase was downregulated in liver tissues from thioacetamide- or HFD-induced liver disease models or in TGF-ß-treated LX-2 cells. TGF-ß-induced upregulation of KCa2.3, KCa3.1, collagen, and α-smooth muscle actin and downregulation of catalase were reversed by modafinil, polyethylene glycol catalase, N-acetylcysteine, siRNA against KCa2.3 or KCa3.1, and Epac inhibitors. Our investigation revealed that modafinil attenuated inflammatory and fibrotic progression via KCa2.3- and KCa3.1-mediated pathways in nonalcoholic hepatitis, suggesting that inhibiting KCa2.3- and KCa3.1-mediated signaling may serve as a novel therapeutic approach for inflammatory and fibrotic liver diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Cirrosis Hepática/tratamiento farmacológico , Modafinilo/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Actinas/biosíntesis , Animales , Antiinflamatorios no Esteroideos/farmacología , Línea Celular , Colágeno/metabolismo , Dieta Alta en Grasa , Hígado Graso/tratamiento farmacológico , Células Estrelladas Hepáticas , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Estereoisomerismo , Tioacetamida/toxicidad
2.
Annu Rev Pharmacol Toxicol ; 60: 219-240, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31337271

RESUMEN

The three small-conductance calcium-activated potassium (KCa2) channels and the related intermediate-conductance KCa3.1 channel are voltage-independent K+ channels that mediate calcium-induced membrane hyperpolarization. When intracellular calcium increases in the channel vicinity, it calcifies the flexible N lobe of the channel-bound calmodulin, which then swings over to the S4-S5 linker and opens the channel. KCa2 and KCa3.1 channels are highly druggable and offer multiple binding sites for venom peptides and small-molecule blockers as well as for positive- and negative-gating modulators. In this review, we briefly summarize the physiological role of KCa channels and then discuss the pharmacophores and the mechanism of action of the most commonly used peptidic and small-molecule KCa2 and KCa3.1 modulators. Finally, we describe the progress that has been made in advancing KCa3.1 blockers and KCa2.2 negative- and positive-gating modulators toward the clinic for neurological and cardiovascular diseases and discuss the remaining challenges.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Animales , Sitios de Unión , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/fisiopatología , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/fisiopatología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
3.
Basic Clin Pharmacol Toxicol ; 122(6): 570-576, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29278444

RESUMEN

Endothelial IKCa and/or SKCa channels play an important role in the control of vascular tone by participating in endothelium-dependent relaxation. Whether ß-AR antagonists, mainly used in hypertension, affect endothelial KCa channel function is unknown. In this study, we examined the effect of the ß2-AR antagonist and inverse agonist ICI 118,551 on the IKCa /SKCa channel activity by assessing functional relaxation responses to several agonists that stimulate these channels. Mesenteric arterial rings isolated from male Sprague Dawley mounted to organ baths. Acetylcholine elicited IKCa - and SKCa -mediated relaxations that were abolished by TRAM-34 and apamin, respectively. ICI 118,551, which did not dilate the arteries per se, increased the IKCa -mediated relaxations, whereas SKCa -mediated relaxations remained unaltered. Same potentiating effect was also detected on the IKCa -mediated relaxations to carbachol and A23187, but not to NS309. Neither acetylcholine-induced nitric oxide-mediated relaxations nor SNP relaxations changed with ICI 118,551. The PKA inhibitor KT-5720, the selective ß2-AR agonist salbutamol, the selective ß2-AR antagonist butoxamine, the non-selective ß-AR antagonist propranolol, and the inverse agonists carvedilol or nadolol failed to affect the IKCa -mediated relaxations. ICI 118,551-induced increase was not reversed by salbutamol or propranolol as well. Besides, low potassium-induced relaxations in endothelium-removed arteries remained the same in the presence of ICI 118,551. These data demonstrate a previously unrecognized action of ICI 118,551, the ability to potentiate endothelial IKCa channel-mediated vasodilation, through a mechanism independent of ß2-AR antagonistic or inverse agonistic action. Instead, the enhancement of acetylcholine relaxation seems likely to occur by a mechanism secondary to endothelial calcium increase.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 2/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Propanolaminas/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Técnicas In Vitro , Masculino , Potasio/farmacología , Ratas , Ratas Wistar
4.
Am J Physiol Heart Circ Physiol ; 311(1): H229-38, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27208165

RESUMEN

Cardiac t-tubules are critical for efficient excitation-contraction coupling but become significantly remodeled during various stress conditions. However, the mechanisms by which t-tubule remodeling occur are poorly understood. Recently, we demonstrated that recovery of mouse ventricular myocytes after hyposmotic shock is associated with t-tubule sealing. In this study, we found that the application of Small Membrane Permeable Molecules (SMPM) such as DMSO, formamide and acetamide upon washout of hyposmotic solution significantly reduced the amount of extracellular dextran trapped within sealed t-tubules. The SMPM protection displayed sharp biphasic concentration dependence that peaks at ∼140 mM leading to >3- to 4-fold reduction in dextran trapping. Consistent with these data, detailed analysis of the effects of DMSO showed that the magnitude of normalized inward rectifier tail current (IK1,tail), an electrophysiological marker of t-tubular integrity, was increased ∼2-fold when hyposmotic stress was removed in the presence of 1% DMSO (∼140 mM). Analysis of dynamics of cardiomyocytes shrinking during resolution of hyposmotic stress revealed only minor increase in shrinking rate in the presence of 1% DMSO, and cell dimensions returned fully to prestress values in both control and DMSO groups. Application and withdrawal of 10% DMSO in the absence of preceding hyposmotic shock induced classical t-tubule sealing. This suggests that the biphasic concentration dependence originated from an increase in secondary t-tubule sealing when high SMPM concentrations are removed. Overall, the data suggest that SMPM protect against sealing of t-tubules following hyposmotic stress, likely through membrane modification and essentially independent of their osmotic effects.


Asunto(s)
Acetamidas/farmacología , Permeabilidad de la Membrana Celular , Membrana Celular/efectos de los fármacos , Dimetilsulfóxido/farmacología , Formamidas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Presión Osmótica , Acetamidas/química , Acetamidas/metabolismo , Animales , Membrana Celular/metabolismo , Dextranos/metabolismo , Dimetilsulfóxido/química , Dimetilsulfóxido/metabolismo , Relación Dosis-Respuesta a Droga , Acoplamiento Excitación-Contracción/efectos de los fármacos , Femenino , Formamidas/química , Formamidas/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Masculino , Potenciales de la Membrana , Ratones Endogámicos C57BL , Peso Molecular , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo
5.
Biochem Biophys Res Commun ; 470(4): 907-12, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26820527

RESUMEN

The intermediate conductance calcium-activated potassium channel (KCa3.1) mediates proliferation of many cell types including fibroblasts, and is a molecular target for intervention in various cell proliferative diseases. Our previous study showed that reduction of KCa3.1 channel expression by lyso-globotriaosylceramide (lyso-Gb3) inhibits differentiation into myofibroblasts and collagen synthesis, which might lead to development of ascending thoracic aortic aneurysm secondary to Fabry disease. However, how lyso-Gb3 downregulates KCa3.1 channel expression is unknown. Therefore, we aimed to investigate the underlying mechanisms of lyso-Gb3-mediated KCa3.1 channel downregulation, focusing on the cAMP signaling pathway. We found that lyso-Gb3 increased the intracellular cAMP concentration by upregulation of adenylyl cyclase 6 and inhibited ERK 1/2 phosphorylation through the protein kinase A (PKA) pathway, leading to the inhibition of KCa3.1 channel synthesis, not the exchange protein directly activated by cAMP (Epac) pathway. Moreover, lyso-Gb3 suppressed expression of class II phosphatidylinositol 3-kinase C2ß (PI3KC2ß) by PKA activation, which reduces the production of phosphatidylinositol 3-phosphate [PI(3)P], and the reduced membrane surface expression of KCa3.1 channel was recovered by increasing the intracellular levels of PI(3)P. Consequently, our findings that lyso-Gb3 inhibited both KCa3.1 channel synthesis and surface expression by increasing intracellular cAMP, and controlled surface expression through changes in PI3KC2ß-mediated PI(3)P production, suggest that modulation of PKA and PI3KC2ß activity to control of KCa3.1 channel expression can be an alternative important target to attenuate ascending thoracic aortic aneurysms in Fabry disease.


Asunto(s)
Membrana Celular/metabolismo , Fosfatidilinositol 3-Quinasas Clase II/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucolípidos/administración & dosificación , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Fosfatos de Fosfatidilinositol/metabolismo , Esfingolípidos/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Ratones , Células 3T3 NIH
6.
Basic Clin Pharmacol Toxicol ; 119(2): 184-92, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26821335

RESUMEN

Opening of intermediate-conductance calcium-activated potassium channels (KC a 3.1) produces membrane hyperpolarization in the vascular endothelium. Here, we studied the ability of two new KC a 3.1-selective positive-gating modulators, SKA-111 and SKA-121, to (1) evoke porcine endothelial cell KC a 3.1 membrane hyperpolarization, (2) induce endothelium-dependent and, particularly, endothelium-derived hyperpolarization (EDH)-type relaxation in porcine coronary arteries (PCA) and (3) influence coronary artery tone in isolated rat hearts. In whole-cell patch-clamp experiments on endothelial cells of PCA (PCAEC), KC a currents evoked by bradykinin (BK) were potentiated ≈7-fold by either SKA-111 or SKA-121 (both at 1 µM) and were blocked by a KC a 3.1 blocker, TRAM-34. In membrane potential measurements, SKA-111 and SKA-121 augmented bradykinin-induced hyperpolarization. Isometric tension measurements in large- and small-calibre PCA showed that SKA-111 and SKA-121 potentiated endothelium-dependent relaxation with intact NO synthesis and EDH-type relaxation to BK by ≈2-fold. Potentiation of the BK response was prevented by KC a 3.1 inhibition. In Langendorff-perfused rat hearts, SKA-111 potentiated coronary vasodilation elicited by BK. In conclusion, our data show that positive-gating modulation of KC a 3.1 channels improves BK-induced membrane hyperpolarization and endothelium-dependent relaxation in small and large PCA as well as in the coronary circulation of rats. Positive-gating modulators of KC a 3.1 could be therapeutically useful to improve coronary blood flow and counteract impaired coronary endothelial dysfunction in cardiovascular disease.


Asunto(s)
Vasos Coronarios/citología , Células Endoteliales/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Animales , Bradiquinina/farmacología , Células Cultivadas , Circulación Coronaria/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Corazón/efectos de los fármacos , Corazón/fisiología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Masculino , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxazoles/farmacología , Técnicas de Placa-Clamp , Pirazoles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Porcinos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
7.
Am J Physiol Heart Circ Physiol ; 309(12): H2031-41, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26453324

RESUMEN

Cerebral parenchymal arterioles (PA) regulate blood flow between pial arteries on the surface of the brain and the deeper microcirculation. Regulation of PA contractility differs from that of pial arteries and is not completely understood. Here, we investigated the hypothesis that the Ca(2+) permeable vanilloid transient receptor potential (TRPV) channel TRPV3 can mediate endothelium-dependent dilation of cerebral PA. Using total internal reflection fluorescence microscopy (TIRFM), we found that carvacrol, a monoterpenoid compound derived from oregano, increased the frequency of unitary Ca(2+) influx events through TRPV3 channels (TRPV3 sparklets) in endothelial cells from pial arteries and PAs. Carvacrol-induced TRPV3 sparklets were inhibited by the selective TRPV3 blocker isopentenyl pyrophosphate (IPP). TRPV3 sparklets have a greater unitary amplitude (ΔF/F0 = 0.20) than previously characterized TRPV4 (ΔF/F0 = 0.06) or TRPA1 (ΔF/F0 = 0.13) sparklets, suggesting that TRPV3-mediated Ca(2+) influx could have a robust influence on cerebrovascular tone. In pressure myography experiments, carvacrol caused dilation of cerebral PA that was blocked by IPP. Carvacrol-induced dilation was nearly abolished by removal of the endothelium and block of intermediate (IK) and small-conductance Ca(2+)-activated K(+) (SK) channels. Together, these data suggest that TRPV3 sparklets cause dilation of cerebral parenchymal arterioles by activating IK and SK channels in the endothelium.


Asunto(s)
Arteriolas/fisiología , Señalización del Calcio/genética , Señalización del Calcio/fisiología , Calcio/metabolismo , Circulación Cerebrovascular/genética , Circulación Cerebrovascular/fisiología , Endotelio Vascular/fisiología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/fisiología , Animales , Señalización del Calcio/efectos de los fármacos , Cimenos , Electromiografía , Hemiterpenos/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Masculino , Monoterpenos/farmacología , Tono Muscular/efectos de los fármacos , Tono Muscular/genética , Tono Muscular/fisiología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Compuestos Organofosforados/farmacología , Ratas , Ratas Sprague-Dawley , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Canales Catiónicos TRPV/antagonistas & inhibidores , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
8.
Atherosclerosis ; 242(1): 191-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26204495

RESUMEN

OBJECTIVE: It remains incompletely understood how homocysteine impairs endothelial function. Whether mechanisms such as calcium-activated potassium (KCa) channels are involved is uncertain and the significance of endoplasmic reticulum (ER) stress in KCa channel-dependent endothelial function in hyperhomocysteinemia remains unexplored. We investigated the effect of homocysteine on endothelial KCa channels in coronary vasculature with further exploration of the role of ER stress. METHODS: Vasorelaxation mediated by intermediate- and small-conductance KCa (IKCa and SKCa) channels was studied in porcine coronary arteries in a myograph. IKCa and SKCa channel currents were recorded by whole-cell patch-clamp in coronary endothelial cells. Protein levels of endothelial IKCa and SKCa channels were determined for both whole-cell and surface expressions. RESULTS: Homocysteine impaired bradykinin-induced IKCa and SKCa-dependent EDHF-type relaxation and attenuated the vasorelaxant response to the channel activator. IKCa and SKCa currents were suppressed by homocysteine. Inhibition of ER stress during homocysteine exposure enhanced IKCa and SKCa currents, associated with improved EDHF-type response and channel activator-induced relaxation. Homocysteine did not alter whole-cell protein levels of IKCa and SKCa whereas lowered surface expressions of these channels, which were restored by ER stress inhibition. CONCLUSIONS: Homocysteine induces endothelial dysfunction through a mechanism involving ER stress-mediated suppression of IKCa and SKCa channels. Inhibition of cell surface expression of these channels by ER stress is, at least partially, responsible for the suppressive effect of homocysteine on the channel function. This study provides new mechanistic insights into homocysteine-induced endothelial dysfunction and advances our knowledge of the significance of ER stress in vascular disorders.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Homocisteína/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Animales , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Técnicas In Vitro , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Potenciales de la Membrana , Transducción de Señal/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Porcinos , Vasodilatadores/farmacología
9.
Toxicol Sci ; 147(1): 286-95, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26117837

RESUMEN

Evaluation of stem cell-derived cardiomyocytes (SC-CM) using multi-electrode array (MEA) has attracted attention as a novel model to detect drug-induced arrhythmia. An experiment was conducted to determine if MEA recording from human induced pluripotent SC-CM (hiPSC-CM) could assess proarrhythmic risk. Ten hERG blockers, 4 Na(+) blockers, and 1 IKs blocker were evaluated blindly. Eight drugs are associated with Torsades de Pointes (TdP) and 4 are not. Multiple parameters, including field potential duration (FPD), Na(+) slope, Na(+) amplitude, beat rate (BR), and early after-depolarization (EAD) were recorded. Minimum effective concentrations (MEC) that elicited a significant change were calculated. Our results determined that FPD and EAD were unable to distinguish torsadogenic from benign compounds, Na(+) slope and amplitude could not differentiate Na(+) channel blockade from hERG blockade, BR had an inconsistent response to pharmacological treatment, and that hiPSC-CM were, in general, insensitive to IKs inhibition. A ratio was calculated that relates MEC for evoking FPD prolongation, or triggering EAD, to the human therapeutic unbound Cmax (MEC/Cmax). The key finding was that the ratio was sensitive, but specificity was low. Consistently, the ratio had high positive predictive value and low negative predictive value. In conclusion, MEA recordings of hiPSC-CM were sensitive for FPD and EAD detection, but unable to distinguish agents with low- and high-risk for TdPs. Although some published reports suggested great potential for MEA recordings in hSC-CM to assess preclinical cardiac toxicity, the current evaluation implies that this model would have a high false-positive rate in regard to proarrhythmic risk.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Arritmias Cardíacas/patología , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Microelectrodos , Bloqueadores de los Canales de Potasio/toxicidad , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Medición de Riesgo , Bloqueadores de los Canales de Sodio/toxicidad , Torsades de Pointes/inducido químicamente , Torsades de Pointes/fisiopatología
10.
Physiol Res ; 64(1): 39-49, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25194131

RESUMEN

To investigate lisinopril effect on the contribution of nitric oxide (NO) and K(Ca) channels to acetylcholine (ACh)-induced relaxation in isolated mesenteric arteries of spontaneously hypertensive rats (SHRs). Third branch mesenteric arteries isolated from lisinopril treated SHR rats (20 mg/kg/day for ten weeks, SHR-T) or untreated (SHR-UT) or normotensive WKY rats were mounted on tension myograph and ACh concentration-response curves were obtained. Westernblotting of eNOS and K(Ca) channels was performed. ACh-induced relaxations were similar in all groups while L-NMMA and indomethacin caused significant rightward shift only in SHR-T group. Apamin and TRAM-34 (SK(Ca) and IK(Ca) channels blockers, respectively) significantly attenuated ACh-induced maximal relaxation by similar magnitude in vessels from all three groups. In the presence of L-NMMA, indomethacin, apamin and TRAM-34 further attenuated ACh-induced relaxation only in SHR-T. Furthermore, lisinopril treatment increased expression of eNOS, SK(Ca) and BK(Ca) proteins. Lisinopril treatment increased expression of eNOS, SK(Ca), BK(Ca) channel proteins and increased the contribution of NO to ACh-mediated relaxation. This increased role of NO was apparent only when EDHF component was blocked by inhibiting SK(Ca) and IK(Ca) channels. Such may suggest that in mesenteric arteries, non-EDHF component functions act as a reserve system to provide compensatory vasodilatation if (and when) hyperpolarization that is mediated by SK(Ca) and IK(Ca) channels is reduced.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Antihipertensivos/farmacología , Hipertensión/tratamiento farmacológico , Lisinopril/farmacología , Arterias Mesentéricas/efectos de los fármacos , Óxido Nítrico/metabolismo , Canales de Potasio Calcio-Activados/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipertensión/metabolismo , Hipertensión/fisiopatología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/efectos de los fármacos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Masculino , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Calcio-Activados/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Transducción de Señal/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
11.
Reprod Sci ; 22(3): 278-84, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25194151

RESUMEN

Circulating inflammatory factors and endothelial dysfunction have been proposed to contribute to the pathophysiology of hemolysis, elevated liver enzymes, and low platelet count (HELLP) syndrome. To date, the occurrence of neurological complications in these women has been reported, but few studies have examined whether impairment in blood-brain barrier (BBB) permeability or cerebrovascular reactivity is present in women having HELLP syndrome. We hypothesized that plasma from women with HELLP syndrome causes increased BBB permeability and cerebrovascular dysfunction. Posterior cerebral arteries from female nonpregnant rats were perfused with 20% serum from women with normal pregnancies (n = 5) or women with HELLP syndrome (n = 5), and BBB permeability and vascular reactivity were compared. Plasma from women with HELLP syndrome increased BBB permeability while not changing myogenic tone and reactivity to pressure. Addition of the nitric oxide (NO) synthase inhibitor N(ω)-nitro-L-arginine methyl ester caused constriction of arteries that was not different with the different plasmas nor was dilation to the NO donor sodium nitroprusside different between the 2 groups. However, dilation to the small- and intermediate-conductance, calcium-activated potassium channel activator NS309 was decreased in vessels exposed to HELLP plasma. Thus, increased BBB permeability in response to HELLP plasma was associated with selective endothelial dysfunction.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Síndrome HELLP/sangre , Arteria Cerebral Posterior/metabolismo , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Femenino , Síndrome HELLP/fisiopatología , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Arteria Cerebral Posterior/efectos de los fármacos , Arteria Cerebral Posterior/fisiopatología , Embarazo , Ratas Sprague-Dawley , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Adulto Joven
12.
Zhonghua Yi Xue Za Zhi ; 94(3): 182-6, 2014 Jan 21.
Artículo en Chino | MEDLINE | ID: mdl-24731458

RESUMEN

OBJECTIVE: To explore the effects of Telmisartan on IKCa1 potassium channel after T-lymphocyte activation and proliferation in peripheral blood of hypertensive patients in Xinjiang Kazakh. METHODS: Peripheral blood T cells in vitro culture were isolated from 30 Xinjiang Kazakh outpatients without antihypertensive drug therapy. They were randomly selected from our hypertension clinic from August 2012 to December 2012. The proliferated T lymphocytes were divided into control, telmisartan and TRAM-34 groups. After culturing for 0, 24, 48 h after corresponding treatments, the patch-clamp technique was employed to record the electrophysiological changes of IKCa1 potassium channel of T lymphocytes. RESULTS: Under different treatment conditions, the IKCa1 potassium channel showed different electrophysiological changes. Pairwise comparison was made among the groups on the same time. For the telmisartan group, IKCa1 potassium channel peak current, peak current density of intervention 24 h and 48 h were significantly reduced compared with the control group (24 h:(835 ± 117)vs(1 471 ± 255) pA, (213 ± 61) vs (388 ± 129) pA/pF; 48 h:(631 ± 142) vs (1 555 ± 383) pA, (155 ± 54) vs (388 ± 114) pA/pF, all P < 0.01) . And the blocking rates of 0 h, 24 h and 48 h of telmisartan on IKCa1 potassium channel were 6.8%, 45.1% and 60.1% respectively. CONCLUSION: Telmisartan can block the IKCa1 potassium channel of T lymphocytes in peripheral blood of hypertensive patients in Xinjiang Kazakh. It suggests that telmisartan may play an anti-inflammatory effect by blocking the IKCa1 potassium channels of T lymphocyte activation.


Asunto(s)
Bencimidazoles/farmacología , Benzoatos/farmacología , Hipertensión/fisiopatología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Activación de Linfocitos , Células Cultivadas , Femenino , Humanos , Hipertensión/sangre , Hipertensión/etnología , Masculino , Persona de Mediana Edad , Linfocitos T/citología , Telmisartán
13.
Pflugers Arch ; 466(2): 307-17, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23873353

RESUMEN

The present study was designed to investigate the role of advanced glycation end products (AGEs) in intermediate-conductance and small-conductance Ca(2+)-activated potassium channels (KCa3.1 and KCa2.3)-mediated relaxation in rat resistance arteries and the underlying mechanism. The endothelial function of mesenteric arteries was assessed with the use of wire myography. Expression levels of KCa3.1 and KCa2.3 were measured by using Western blot. Reactive oxygen species (ROS) were measured by using dihydroethidium and 2', 7'-dichlorofluorescein diacetate. KCa3.1 and KCa2.3-mediated vasodilatation responses to acetylcholine and NS309 (opener of KCa3.1 and KCa2.3) were impaired by incubation of the third-order mesenteric arteries from normal rats with AGEs (200 µg ml(-1) for 3 h). In cultured human umbilical vein endothelial cells (HUVECs), AGEs increased ROS level and decreased the protein expression of KCa3.1 and KCa2.3. Antioxidant alpha lipoic acid restored the impairment in both vasodilatation function and expression of KCa3.1 and KCa2.3. H2O2 could mimic the effect of AGEs on the protein expression of KCa3.1 and KCa2.3 in cultured HUVECs. These results demonstrate for the first time that AGEs impaired KCa3.1 and KCa2.3-mediated vasodilatation in rat mesenteric arteries via downregulation of both KCa3.1 and KCa2.3, in which the enhanced oxidative stress was involved.


Asunto(s)
Productos Finales de Glicación Avanzada/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Arterias Mesentéricas/fisiología , Estrés Oxidativo/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Alcanos/farmacología , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/biosíntesis , Masculino , Arterias Mesentéricas/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Pirazoles/farmacología , Compuestos de Quinolinio/farmacología , Ratas , Ratas Sprague-Dawley , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/biosíntesis
14.
Br J Pharmacol ; 170(2): 293-303, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23734697

RESUMEN

BACKGROUND AND PURPOSE: The calcium-activated potassium channel KCa3.1 is expressed in the vascular endothelium where its activation causes endothelial hyperpolarization and initiates endothelium-derived hyperpolarization (EDH)-dependent dilatation. Here, we investigated whether pharmacological activation of KCa3.1 dilates skeletal muscle arterioles and whether myoendothelial gap junctions formed by connexin40 (Cx40) are required for EDH-type dilatations and pressure depressor responses in vivo. EXPERIMENTAL APPROACH: We performed intravital microscopy in the cremaster muscle microcirculation and blood pressure telemetry in Cx40-deficient mice. KEY RESULTS: In wild-type mice, the KCa3.1-activator SKA-31 induced pronounced concentration-dependent arteriolar EDH-type dilatations, amounting to ∼40% of maximal dilatation, and enhanced the effects of ACh. These responses were absent in mice devoid of KCa3.1 channels. In contrast, SKA-31-induced dilatations were not attenuated in mice with endothelial cells deficient in Cx40 (Cx40(fl/fl):Tie2-Cre). In isolated endothelial cell clusters, SKA-31 induced hyperpolarizations of similar magnitudes (by ∼38 mV) in Cx40(fl/fl):Tie2-Cre, ubiquitous Cx40-deficient mice (Cx40(-/-)) and controls (Cx40(fl/fl)), which were reversed by the specific KCa3.1-blocker TRAM-34. In normotensive wild-type and Cx40(fl/fl):Tie2-Cre as well as in hypertensive Cx40(-/-) animals, i.p. injections of SKA-31 (30 and 100 mg·kg(-1)) decreased arterial pressure by ∼32 mmHg in all genotypes. The depressor response to 100 mg·kg(-1) SKA-31 was associated with a decrease in heart rate. CONCLUSIONS AND IMPLICATIONS: We conclude that endothelial hyperpolarization evoked by pharmacological activation of KCa3.1 channels induces EDH-type arteriolar dilatations that are independent of endothelial Cx40 and Cx40-containing myoendothelial gap junctions. As SKA-31 reduced blood pressure in hypertensive Cx40-deficient mice, KCa3.1 activators may be useful drugs for severe treatment-resistant hypertension.


Asunto(s)
Benzotiazoles/farmacología , Conexinas/genética , Hipertensión/tratamiento farmacológico , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Animales , Benzotiazoles/administración & dosificación , Presión Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Genotipo , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/fisiopatología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Ratones , Ratones Noqueados , Microcirculación/efectos de los fármacos , Pirazoles/farmacología , Telemetría , Vasodilatación/efectos de los fármacos , Proteína alfa-5 de Unión Comunicante
15.
J Membr Biol ; 246(4): 315-26, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23430221

RESUMEN

DIDS, NPPB, tannic acid (TA) and AO1 are widely used inhibitors of Cl(-) channels. Some Cl(-) channel inhibitors (NPPB, DIDS, niflumic acid) were shown to affect phosphatidylserine (PS) scrambling and, thus, the life span of human red blood cells (hRBCs). Since a number of publications suggest Ca(2+) dependence of PS scrambling, we explored whether inhibitors of Cl(-) channels (DIDS, NPPB) or of Ca(2+)-activated Cl(-) channels (DIDS, NPPB, TA, AO1) modified intracellular free Ca(2+) concentration ([Ca(2+)]i) and activity of Ca(2+)-activated K(+) (Gardos) channel in hRBCs. According to Fluo-3 fluorescence in flow cytometry, a short treatment (15 min, +37 °C) with Cl(-) channels inhibitors decreased [Ca(2+)]i in the following order: TA > AO1 > DIDS > NPPB. According to forward scatter, the decrease of [Ca(2+)]i was accompanied by a slight but significant increase in cell volume following DIDS, NPPB and AO1 treatments. TA treatment resulted in cell shrinkage. According to whole-cell patch-clamp experiments, TA activated and NPPB and AO1 inhibited Gardos channels. The Cl(-) channel blockers further modified the alterations of [Ca(2+)]i following ATP depletion (glucose deprivation, iodoacetic acid, 6-inosine), oxidative stress (1 mM t-BHP) and treatment with Ca(2+) ionophore ionomycin (1 µM). The ability of the Cl(-) channel inhibitors to modulate PS scrambling did not correlate with their influence on [Ca(2+)]i as TA and AO1 had a particularly strong decreasing effect on [Ca(2+)]i but at the same time enhanced PS exposure. In conclusion, Cl(-) channel inhibitors affect Gardos channels, influence Ca(2+) homeostasis and induce PS exposure of hRBCs by Ca(2+)-independent mechanisms.


Asunto(s)
Calcio/metabolismo , Canales de Cloruro/antagonistas & inhibidores , Canales de Cloruro/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Células Cultivadas , Electrofisiología , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Nitrobenzoatos/farmacología , Taninos/farmacología
16.
Diab Vasc Dis Res ; 10(1): 65-71, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22621918

RESUMEN

Fluoroquinolone antibiotics cause rare, but clinically important, adverse events including hyperglycaemia and hypoglycaemia. The present study focuses on the possible effect of levofloxacin and moxifloxacin on the cardiovascular functions of rats with type I diabetes. Both antibiotics caused bradycardia. Levofloxacin but not moxifloxacin caused hypoglycaemia in diabetic rats and an increase in amplitude of the ST segment revealed by electrocardiogram (ECG) analysis of isolated hearts. In pressurized mesenteric arteries, levofloxacin did not affect the endothelium-derived hyperpolarising factor (EDHF) pathway or its main components, the small-conductance Ca(2+) activated potassium (SK(Ca)) and intermediate-conductance Ca(2+) activated potassium (IK(Ca)) channels. In moxifloxacin-treated rats, an increase in the EDHF response was observed, which was largely attributed to SK(Ca)-activation. In conclusion, levofloxacin and moxifloxacin use appeared to vary but with no evidence of impairment of the cardiovascular function. However, it is still possible that these antibiotics may produce different effects if there are co-morbidities and therefore their use must be with care.


Asunto(s)
Antibacterianos/farmacología , Compuestos Aza/farmacología , Sistema Cardiovascular/efectos de los fármacos , Diabetes Mellitus Experimental/fisiopatología , Hipoglucemia/inducido químicamente , Levofloxacino , Ofloxacino/farmacología , Quinolinas/farmacología , Animales , Factores Biológicos , Glucemia/efectos de los fármacos , Bradicardia/inducido químicamente , Diabetes Mellitus Experimental/inducido químicamente , Electrocardiografía , Endotelio Vascular/efectos de los fármacos , Fluoroquinolonas/farmacología , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Masculino , Arterias Mesentéricas/efectos de los fármacos , Moxifloxacino , Ratas , Ratas Wistar , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Estreptozocina
17.
J Neuroimmune Pharmacol ; 8(1): 227-37, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22527636

RESUMEN

Microglia monitor the CNS for 'danger' signals after acute injury, such as stroke and trauma, and then undergo complex activation processes. Classical activation of microglia can produce neurotoxic levels of glutamate and immune mediators (e.g., pro-inflammatory cytokines, reactive oxygen and nitrogen species), while alternative activation up-regulates anti-inflammatory molecules and is thought to resolve inflammation and protect the brain. Thus, pharmacological strategies to decrease classical- and/or promote alternative activation are of interest. Here, we assessed actions of the neuroprotective drug, riluzole, on two Ca(2+)- activated K channels in microglia - SK3 (KCa2.3, KCNN3) and SK4 (KCa3.1, KCNN4) - and on classical versus alternative microglial activation. Riluzole is used to treat amyotrophic lateral sclerosis, and is in clinical trials for several other CNS disorders, where it has been presumed to target neurons and reduce glutamate-mediated toxicity. We show that simply elevating intracellular Ca(2+) to micromolar levels in whole-cell recordings does not activate SK channels in a cell line derived from primary rat microglia (MLS-9). In intact cells, riluzole raised cytoplasmic Ca(2+), but it was marginal (~200 nM) and transient (2 min). Surprisingly then, in whole cell recordings, riluzole rapidly activated SK3 and SK4 channels for as long as it was present, and did not require elevated intracellular Ca(2+). We then used primary rat microglia to analyze expression of several activation markers and inflammatory mediators. Riluzole decreased classical LPS-induced activation, and increased some aspects of IL-4-induced alternative activation. These actions on microglia suggest an additional mechanism underlying the neuroprotective actions of riluzole.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Riluzol/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Línea Celular , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Microglía/metabolismo , Técnicas de Placa-Clamp , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
J Cardiovasc Pharmacol ; 61(2): 102-12, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23107876

RESUMEN

Most cardiovascular researchers are familiar with intermediate-conductance KCa3.1 and small-conductance KCa2.3 channels because of their contribution to endothelium-derived hyperpolarization. However, to immunologists and neuroscientists, these channels are primarily known for their role in lymphocyte activation and neuronal excitability. KCa3.1 is involved in the proliferation and migration of T cells, B cells, mast cells, macrophages, fibroblasts, and dedifferentiated vascular smooth muscle cells and is, therefore, being pursued as a potential target for use in asthma, immunosuppression, and fibroproliferative disorders. In contrast, the 3 KCa2 channels (KCa2.1, KCa2.2, and KCa2.3) contribute to the neuronal medium afterhyperpolarization and, depending on the type of neuron, are involved in determining firing rates and frequencies or in regulating bursting. KCa2 activators are accordingly being studied as potential therapeutics for ataxia and epilepsy, whereas KCa2 channel inhibitors like apamin have long been known to improve learning and memory in rodents. Given this background, we review the recent discoveries of novel KCa3.1 and KCa2.3 modulators and critically assess the potential of KCa activators for the treatment of diabetes and cardiovascular diseases by improving endothelium-derived hyperpolarizations.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Animales , Factores Biológicos/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/fisiopatología , Diseño de Fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Terapia Molecular Dirigida , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-22908201

RESUMEN

Early studies showed that airway cells secrete HCO(3)(-) in response to cAMP-mediated agonists and HCO(3)(-) secretion was impaired in cystic fibrosis (CF). Studies with Calu-3 cells, an airway serous model with high expression of CFTR, also show the secretion of HCO(3)(-) when cells are stimulated with cAMP-mediated agonists. Activation of basolateral membrane hIK-1 K(+) channels inhibits HCO(3)(-) secretion and stimulates Cl(-) secretion. CFTR mediates the exit of both HCO(3)(-) and Cl(-) across the apical membrane. Entry of HCO(3)(-) on a basolateral membrane NBC or Cl(-) on the NKCC determines which anion is secreted. Switching between these two secreted anions is determined by the activity of hIK-1 K(+) channels.


Asunto(s)
Bicarbonatos/metabolismo , Bronquios/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Bencimidazoles/farmacología , Bicarbonatos/antagonistas & inhibidores , Bronquios/citología , Calcio/metabolismo , Agonistas de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Línea Celular , Colforsina/farmacología , AMP Cíclico/antagonistas & inhibidores , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Impedancia Eléctrica , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Potenciales de la Membrana/efectos de los fármacos , Microelectrodos , Membrana Serosa/efectos de los fármacos
20.
Acta Physiol (Oxf) ; 203(1): 117-26, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21362152

RESUMEN

AIM: Endothelial membrane hyperpolarization mediated by KCa3.1 and KCa2.3 channels has been demonstrated to initiate endothelium-derived hyperpolarizing factor (EDHF)-type vasodilations. Moreover, pharmacological potentiation of KCa3.1/KCa2.3 channels has been suggested to improve EDHF-type vasodilations. Herein, we determined whether the KCa3.1/KCa2.3 activator SKA-31 and its derivative SKA-20 improve endothelial dysfunction in KCa3.1-/- and NOS3-/- mice. METHODS: Membrane potentials were measured using patch-clamp electrophysiology on carotid artery (CA) endothelial cells (CAEC) from wild-type (wt) and KCa3.1-/- mice. Endothelium-dependent vasodilations were determined by pressure myography in CA. RESULTS: SKA-31 (1 µm) activated KCa3.1 and KCa2.3 channels and induced membrane hyperpolarization in CAEC of wt (ΔMP -45 mV). These responses were significantly reduced in CAEC of KCa3.1-/- (ΔMP -8 mV). SKA-31 (200 nm, 500 nm) and SKA-20 (300 nm) significantly enhanced EDHF vasodilations in wt. SKA-20 also improved vasodilations during NO synthesis. In KCa3.1-/-, the defective EDHF vasodilations were unchanged at 200 nm SKA-31, but were significantly improved at 500 nm. EDHF vasodilations were slightly enhanced at 300 nm SKA-20, but vasodilations during NO synthesis were unchanged. SKA-31 (500 nm) enhanced the impaired endothelium-dependent vasodilation in NOS3-/- mice twofold. Pharmacological inhibition of the soluble epoxide hydrolase by t-AUCB (1 µm) in contrast did not increase ACh-induced EDHF- or NO-mediated vasodilations in wt and KCa3.1-/-. CONCLUSION: Normal and defective endothelium-dependent vasodilations in murine carotid arteries can be improved by pharmacological enhancement of KCa3.1/KCa2.3 functions. These findings further support the concept that pharmacological activation of endothelial KCa2.3/KCa3.1 could offer a novel endothelium-specific antihypertensive strategy.


Asunto(s)
Benzotiazoles/farmacología , Endotelio Vascular/efectos de los fármacos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Endotelio Vascular/metabolismo , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Vasodilatación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...