Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Physiol ; 100(4): 399-409, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25641541

RESUMO

NEW FINDINGS: What is the central question of this study? What are the effects of protein kinase C (PKC) and Ca(2+) -calmodulin-dependent protein kinase II (CaMKII) on late sodium current (INaL ), reverse Na(+) -Ca(2+) exchange current (reverse INCX ) or intracellular Ca(2+) levels changed by ouabain? What is the main finding and its importance? Ouabain, even at low concentrations (0.5-8.0 µm), can increase INaL and reverse INCX , and these effects may contribute to the effect of the glycoside to increase Ca(2+) transients and contractility. Both PKC and CaMKII activities may mediate or modulate these processes. It has been reported that the cardiac glycoside ouabain can increase the late sodium current (INaL ), as well as the diastolic intracellular calcium concentration and contractile shortening. Whether an increase of INaL participates in a pathway that can mediate the positive inotropic response to ouabain is unknown. We therefore determined the effects of ouabain on INaL , reverse Na(+) -Ca(2+) exchange current (reverse INCX ), intracellular Ca(2+) ([Ca(2+) ]i ) levels and contractile shortening in rabbit isolated ventricular myocytes. Ouabain (0.1-8 µm) markedly increased INaL and reverse INCX in a concentration-dependent manner, with significant effects at concentrations as low as 0.5 and 1 µm. These effects of ouabain were suppressed by the INaL inhibitors TTX and ranolazine, the protein kinase C inhibitor bisindolylmaleimide and the Ca(2+) -calmodulin-dependent protein kinase II inhibitor KN-93. The enhancement by 0.5 µm ouabain of ventricular myocyte contractility and intracellular Ca(2+) transients was suppressed by 2.0 µm TTX. We conclude that ouabain, even at low concentrations (0.5-8.0 µm), can increase INaL and reverse INCX , and these effects may contribute to the effect of the glycoside to increase Ca(2+) transients and contractility. Both protein kinase C and Ca(2+) -calmodulin-dependent protein kinase II activities may mediate or modulate these processes.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Ouabaína/administração & dosagem , Proteína Quinase C/metabolismo , Sódio/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Cardiotônicos/administração & dosagem , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/administração & dosagem , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Coelhos
2.
Heart Rhythm ; 12(2): 440-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25460862

RESUMO

Myocyte sodium channel current that persists throughout the plateau of the cardiac action potential is referred to as late sodium current (I(Na-L)). The magnitude of I(Na-L) is normally small, but can increase significantly in common acute and chronic pathological settings as a result of inherited and/or acquired Na(+) channelopathies that alter channel opening and closing (ie, gating), location (trafficking), or anchoring and interactions with cytoskeletal proteins. An increase in I(Na-L) reduces repolarization reserve in atrial and ventricular myocytes and prolongs the action potential duration and the QT interval. An enhanced I(Na-L) is a cause of long QT syndrome 3. I(Na-L) may be a cause of afterdepolarizations, triggered arrhythmias, and spontaneous diastolic depolarization-induced automaticity. In addition, enhancement of I(Na-L) increases both the temporal and the spatial dispersion of repolarization in the myocardium and may lead to spatially discordant action potential duration alternans, wavebreak, and reentrant arrhythmias. Positive feedback loops between increases in I(Na-L) and the activity of Ca(2+)/calmodulin-dependent protein kinase II appear to contribute to the genesis of arrhythmias and to certain abnormalities of the ischemic heart. In this review, we discuss some of the more relevant experimental results, clinical findings, and insights from cellular and animal models that highlight the role of I(Na-L) in the genesis of arrhythmias, long QT syndromes, and intracellular Ca(2+) homeostasis.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Síndrome do QT Longo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Sódio/metabolismo , Animais , Humanos , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/patologia , Síndrome do QT Longo/fisiopatologia , Miócitos Cardíacos/patologia
3.
J Mol Cell Cardiol ; 76: 247-56, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25252177

RESUMO

An increase of late Na(+) current (INaL) in cardiac myocytes can raise the cytosolic Na(+) concentration and is associated with activation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and alterations of mitochondrial metabolism and Ca(2+) handling by sarcoplasmic reticulum (SR). We tested the hypothesis that augmentation of INaL can increase mitochondrial reactive oxygen species (ROS) production and oxidation of CaMKII, resulting in spontaneous SR Ca(2+) release and increased diastolic Ca(2+) in myocytes. Increases of INaL and/or of the cytosolic Na(+) concentration led to mitochondrial ROS production and oxidation of CaMKII to cause dysregulation of Ca(2+) handling in rabbit cardiac myocytes.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Miócitos Cardíacos/enzimologia , Sódio/metabolismo , Potenciais de Ação , Animais , Sinalização do Cálcio , Feminino , Ventrículos do Coração/citologia , Ventrículos do Coração/enzimologia , Espaço Intracelular/metabolismo , Oxirredução , Estresse Oxidativo , Coelhos , Espécies Reativas de Oxigênio/metabolismo
4.
J Cardiovasc Pharmacol ; 64(1): 60-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24705174

RESUMO

Ranolazine attenuates cardiac arrhythmic activity associated with hypoxia and hydrogen peroxide (H2O2) by inhibition of late sodium current (late INa). The mechanism of ranolazine's action on Na channels was investigated using whole-cell and single-channel recording from guinea pig isolated ventricular myocytes. Hypoxia increased whole-cell late INa from -0.48 ± 0.02 to -3.99 ± 0.07 pA/pF. Ranolazine at 3 and 9 µmol/L reduced the hypoxia-induced late INa by 16% ± 3% and 55% ± 3%, respectively. Hypoxia increased the mean open probability and open time of Na-channel late openings from 0.016 ± 0.001 to 0.064 ± 0.007 milliseconds and from 0.693 ± 0.043 to 1.081 ± 0.098 milliseconds, respectively. Ranolazine at 3 and 9 µmol/L attenuated the hypoxia-induced increase of open probability by 19% ± 7% and 61% ± 1%, and increase of open time by 26% ± 19% and 74 ± 21%, respectively. H2O2 increased the mean open probability and open time of Na-channel late openings from 0.013 ± 0.002 to 0.107 ± 0.015 milliseconds and from 0.689 ± 0.075 to 1.487 ± 0.072 milliseconds, respectively. Ranolazine at 3 and 6 µmol/L reduced the H2O2-induced increase of mean open probability by 60% ± 7% and 95% ± 2%, and the increase of mean open time by 31% ± 21% and 82% ± 8%. In conclusion, the inhibition by ranolazine of hypoxia- and H2O2-stimulated late INa is due to reduction of both the open probability and open time of Na-channel late openings.


Assuntos
Acetanilidas/farmacologia , Antiarrítmicos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Piperazinas/farmacologia , Canais de Sódio/efeitos dos fármacos , Acetanilidas/administração & dosagem , Animais , Antiarrítmicos/administração & dosagem , Hipóxia Celular , Relação Dose-Resposta a Droga , Feminino , Cobaias , Peróxido de Hidrogênio/farmacologia , Masculino , Miócitos Cardíacos/metabolismo , Piperazinas/administração & dosagem , Ranolazina , Canais de Sódio/metabolismo , Fatores de Tempo
5.
Handb Exp Pharmacol ; 221: 137-68, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24737235

RESUMO

Late I Na is an integral part of the sodium current, which persists long after the fast-inactivating component. The magnitude of the late I Na is relatively small in all species and in all types of cardiomyocytes as compared with the amplitude of the fast sodium current, but it contributes significantly to the shape and duration of the action potential. This late component had been shown to increase in several acquired or congenital conditions, including hypoxia, oxidative stress, and heart failure, or due to mutations in SCN5A, which encodes the α-subunit of the sodium channel, as well as in channel-interacting proteins, including multiple ß subunits and anchoring proteins. Patients with enhanced late I Na exhibit the type-3 long QT syndrome (LQT3) characterized by high propensity for the life-threatening ventricular arrhythmias, such as Torsade de Pointes (TdP), as well as for atrial fibrillation. There are several distinct mechanisms of arrhythmogenesis due to abnormal late I Na, including abnormal automaticity, early and delayed after depolarization-induced triggered activity, and dramatic increase of ventricular dispersion of repolarization. Many local anesthetic and antiarrhythmic agents have a higher potency to block late I Na as compared with fast I Na. Several novel compounds, including ranolazine, GS-458967, and F15845, appear to be the most selective inhibitors of cardiac late I Na reported to date. Selective inhibition of late I Na is expected to be an effective strategy for correcting these acquired and congenital channelopathies.


Assuntos
Arritmias Cardíacas/metabolismo , Frequência Cardíaca , Miócitos Cardíacos/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Animais , Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Predisposição Genética para Doença , Frequência Cardíaca/efeitos dos fármacos , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Fenótipo , Transdução de Sinais , Bloqueadores dos Canais de Sódio/uso terapêutico , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/genética
6.
Am J Physiol Heart Circ Physiol ; 306(3): H455-61, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24322614

RESUMO

An increase of cardiac late sodium current (INa.L) is arrhythmogenic in atrial and ventricular tissues, but the densities of INa.L and thus the potential relative contributions of this current to sodium ion (Na(+)) influx and arrhythmogenesis in atria and ventricles are unclear. In this study, whole-cell and cell-attached patch-clamp techniques were used to measure INa.L in rabbit left atrial and ventricular myocytes under identical conditions. The density of INa.L was 67% greater in left atrial (0.50 ± 0.09 pA/pF, n = 20) than in left ventricular cells (0.30 ± 0.07 pA/pF, n = 27, P < 0.01) when elicited by step pulses from -120 to -20 mV at a rate of 0.2 Hz. Similar results were obtained using step pulses from -90 to -20 mV. Anemone toxin II (ATX II) increased INa.L with an EC50 value of 14 ± 2 nM and a Hill slope of 1.4 ± 0.1 (n = 9) in atrial myocytes and with an EC50 of 21 ± 5 nM and a Hill slope of 1.2 ± 0.1 (n = 12) in ventricular myocytes. Na(+) channel open probability (but not mean open time) was greater in atrial than in ventricular cells in the absence and presence of ATX II. The INa.L inhibitor ranolazine (3, 6, and 9 µM) reduced INa.L more in atrial than ventricular myocytes in the presence of 40 nM ATX II. In summary, rabbit left atrial myocytes have a greater density of INa.L and higher sensitivities to ATX II and ranolazine than rabbit left ventricular myocytes.


Assuntos
Acetanilidas/farmacologia , Venenos de Cnidários/farmacologia , Átrios do Coração/citologia , Ventrículos do Coração/citologia , Miócitos Cardíacos/fisiologia , Piperazinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Sódio/metabolismo , Potenciais de Ação , Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Especificidade de Órgãos , Coelhos , Ranolazina
7.
Am J Physiol Heart Circ Physiol ; 305(7): H1068-79, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23873796

RESUMO

The treatment of heart failure (HF) is challenging and morbidity and mortality are high. The goal of this study was to determine if inhibition of the late Na(+) current with ranolazine during early hypertensive heart disease might slow or stop disease progression. Spontaneously hypertensive rats (aged 7 mo) were subjected to echocardiographic study and then fed either control chow (CON) or chow containing 0.5% ranolazine (RAN) for 3 mo. Animals were then restudied, and each heart was removed for measurements of t-tubule organization and Ca(2+) transients using confocal microscopy of the intact heart. RAN halted left ventricular hypertrophy as determined from both echocardiographic and cell dimension (length but not width) measurements. RAN reduced the number of myocytes with t-tubule disruption and the proportion of myocytes with defects in intracellular Ca(2+) cycling. RAN also prevented the slowing of the rate of restitution of Ca(2+) release and the increased vulnerability to rate-induced Ca(2+) alternans. Differences between CON- and RAN-treated animals were not a result of different expression levels of voltage-dependent Ca(2+) channel 1.2, sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a, ryanodine receptor type 2, Na(+)/Ca(2+) exchanger-1, or voltage-gated Na(+) channel 1.5. Furthermore, myocytes with defective Ca(2+) transients in CON rats showed improved Ca(2+) cycling immediately upon acute exposure to RAN. Increased late Na(+) current likely plays a role in the progression of cardiac hypertrophy, a key pathological step in the development of HF. Early, chronic inhibition of this current slows both hypertrophy and development of ultrastructural and physiological defects associated with the progression to HF.


Assuntos
Acetanilidas/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Piperazinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Sódio/metabolismo , Animais , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Hipertensão/complicações , Hipertensão/diagnóstico por imagem , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Masculino , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Ranolazina , Ratos , Ratos Endogâmicos SHR , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Canais de Sódio/metabolismo , Trocador de Sódio e Cálcio/efeitos dos fármacos , Trocador de Sódio e Cálcio/metabolismo , Fatores de Tempo , Ultrassonografia
8.
Cardiovasc Res ; 99(4): 600-11, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23752976

RESUMO

This review presents the roles of cardiac sodium channel NaV1.5 late current (late INa) in generation of arrhythmic activity. The assumption of the authors is that proper Na(+) channel function is necessary to the maintenance of the transmembrane electrochemical gradient of Na(+) and regulation of cardiac electrical activity. Myocyte Na(+) channels' openings during the brief action potential upstroke contribute to peak INa and initiate excitation-contraction coupling. Openings of Na(+) channels outside the upstroke contribute to late INa, a depolarizing current that persists throughout the action potential plateau. The small, physiological late INa does not appear to be critical for normal electrical or contractile function in the heart. Late INa does, however, reduce the net repolarizing current, prolongs action potential duration, and increases cellular Na(+) loading. An increase of late INa, due to acquired conditions (e.g. heart failure) or inherited Na(+) channelopathies, facilitates the formation of early and delayed afterpolarizations and triggered arrhythmias, spontaneous diastolic depolarization, and cellular Ca(2+) loading. These in turn increase the spatial and temporal dispersion of repolarization time and may lead to reentrant arrhythmias.


Assuntos
Arritmias Cardíacas/etiologia , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Potenciais de Ação , Animais , Cálcio/metabolismo , Homeostase , Humanos , Sódio/metabolismo , Tetrodotoxina/farmacologia
9.
J Pharmacol Exp Ther ; 344(1): 23-32, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23010360

RESUMO

Inhibition of cardiac late sodium current (late I(Na)) is a strategy to suppress arrhythmias and sodium-dependent calcium overload associated with myocardial ischemia and heart failure. Current inhibitors of late I(Na) are unselective and can be proarrhythmic. This study introduces GS967 (6-[4-(trifluoromethoxy)phenyl]-3-(trifluoromethyl)-[1,2,4]triazolo[4,3-a]pyridine), a potent and selective inhibitor of late I(Na), and demonstrates its effectiveness to suppress ventricular arrhythmias. The effects of GS967 on rabbit ventricular myocyte ion channel currents and action potentials were determined. Anti-arrhythmic actions of GS967 were characterized in ex vivo and in vivo rabbit models of reduced repolarization reserve and ischemia. GS967 inhibited Anemonia sulcata toxin II (ATX-II)-induced late I(Na) in ventricular myocytes and isolated hearts with IC(50) values of 0.13 and 0.21 µM, respectively. Reduction of peak I(Na) by GS967 was minimal at a holding potential of -120 mV but increased at -80 mV. GS967 did not prolong action potential duration or the QRS interval. GS967 prevented and reversed proarrhythmic effects (afterdepolarizations and torsades de pointes) of the late I(Na) enhancer ATX-II and the I(Kr) inhibitor E-4031 in isolated ventricular myocytes and hearts. GS967 significantly attenuated the proarrhythmic effects of methoxamine+clofilium and suppressed ischemia-induced arrhythmias. GS967 was more potent and effective to reduce late I(Na) and arrhythmias than either flecainide or ranolazine. Results of all studies and assays of binding and activity of GS967 at numerous receptors, transporters, and enzymes indicated that GS967 selectively inhibited late I(Na). In summary, GS967 selectively suppressed late I(Na) and prevented and/or reduced the incidence of experimentally induced arrhythmias in rabbit myocytes and hearts.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Cardiotônicos/farmacologia , Piridinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Triazóis/farmacologia , Acetanilidas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Venenos de Cnidários/farmacologia , Feminino , Flecainida/farmacologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Mutação/fisiologia , Isquemia Miocárdica/complicações , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Piperazinas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Compostos de Amônio Quaternário/farmacologia , Coelhos , Ranolazina
10.
J Cardiovasc Pharmacol ; 60(3): 276-82, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22635076

RESUMO

PURPOSES: We determined whether a small molecule inhibitor of apoptosis signal-regulating kinase 1 (ASK1-i) could reduce myocardial infarct size in a rat ischemia/reperfusion model. METHODS AND RESULTS: Sprague-Dawley rats were randomized to 3 groups: ASK1-i infusion (n = 16), vehicle infusion (n = 16), or ischemic preconditioning (IPC; n = 15). Infusion of ASK1-i (10 mg/kg, iv) or vehicle commenced 45 minutes before myocardial ischemia. IPC consisted of 3 cycles of 3 minutes of coronary occlusion followed by 5 minutes of reperfusion immediately before index myocardial ischemia, which consisted of 30-minute left coronary occlusion followed by 180 minutes of reperfusion. Pathologic analysis revealed no significant difference in the ischemic risk size among the 3 groups. ASK1-I and IPC significantly reduced myocardial infarct size (27.7% ± 3.3%, 16.5% ± 3.4%, and 41.5% ± 4.8% in the ASK1-i group, the IPC group, and the vehicle group, respectively; P = 0.0002) and apoptosis (the percentage of apoptotic nuclei averaged 11.6% ± 1.0%, 10.2% ± 1.7%, and 17.7% ± 2.0% in the ASK1-i group, IPC group, and vehicle group, respectively, P = 0.0055). CONCLUSIONS: A small molecule inhibitor of ASK1 was shown for the first time to reduce apoptosis and myocardial infarct size in a rat model of ischemia/reperfusion.


Assuntos
Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , MAP Quinase Quinase Quinase 5/antagonistas & inibidores , Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Apoptose/fisiologia , MAP Quinase Quinase Quinase 5/metabolismo , Masculino , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley
11.
Hypertension ; 59(3): 705-11, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22311911

RESUMO

Endothelin receptor antagonists and phosphodiesterase type 5 inhibitors are used to treat pulmonary arterial hypertension. We tested the hypothesis that a selective endothelin type A receptor antagonist (ambrisentan) and a phosphodiesterase type 5 inhibitor (tadalafil) may act synergistically to relax endothelin-constricted pulmonary arteries. Rat isolated intrapulmonary arterial rings contracted with 8 nmol/L endothelin-1 were relaxed by 10 nmol/L ambrisentan and 30 nmol/L tadalafil alone by 26±3% and 21±1%, respectively, whereas both drugs in combination acted synergistically to relax arterial rings by 83±6%. The nonselective endothelin type A and B receptor antagonists bosentan (100 nmol/L) and macitentan (30 nmol/L) alone relaxed endothelin-contracted rings by 30±5% and 24±3%, respectively. Combinations of 30 nmol/L tadalafil with 100 nmol/L bosentan or 30 nmol/L macitentan relaxed endothelin-contracted rings by 53±5% or 46±7%, respectively; these values are similar to the calculated sums of the individual effects of these compounds. Denudation of endothelium from pulmonary arterial rings abolished the vasodilator response to 30 nmol/L tadalafil and the synergistic vasorelaxant effect of tadalafil with ambrisentan. In the presence of 1 µmol/L BQ-788, a selective endothelin type B receptor antagonist, the vasorelaxant effects of 10 nmol/L ambrisentan and 30 nmol/L tadalafil were additive but not synergistic. These data can be interpreted to suggest that ambrisentan and tadalafil synergistically inhibit endothelin-1-induced constriction of rat intrapulmonary arteries and that endothelin type B receptors in endothelium are necessary to enable a synergistic vasorelaxant effect of the drug combination.


Assuntos
Carbolinas/farmacologia , Endotelina-1/toxicidade , Hipertensão Pulmonar/tratamento farmacológico , Fenilpropionatos/farmacologia , Artéria Pulmonar/fisiopatologia , Piridazinas/farmacologia , Vasoconstrição/efeitos dos fármacos , Animais , Western Blotting , Modelos Animais de Doenças , Sinergismo Farmacológico , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/fisiopatologia , Masculino , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/patologia , Ratos , Ratos Sprague-Dawley , Tadalafila , Vasodilatadores/farmacologia
12.
Neuropharmacology ; 62(7): 2251-60, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22313527

RESUMO

Ranolazine, an anti-anginal drug, reduces neuropathic and inflammatory-induced allodynia in rats. However, the mechanism of ranolazin's anti-allodynic effect is not known. We hypothesized that ranolazine would reduce dorsal root ganglion (DRG) Na(+) current (I(Na)) and neuronal firing by stabilizing Na(+) channels in inactivated states to cause voltage- and frequency-dependent block. Therefore, we investigated the effects of ranolazine on tetrodotoxin-sensitive (TTXs) and tetrodotoxin-resistant (TTXr) I(Na) and action potential parameters of small diameter DRG neurons from embryonic rats. Ranolazine (10 and 30 µM) significantly reduced the firing frequency of evoked action potentials in DRG neurons from 19.2 ± 1.4 to 9.8 ± 2.7 (10 µM) and 5.7 ± 1.3 (30 µM) Hz at a resting membrane potential of -40 mV. Ranolazine blocked TTXs and TTXr in a voltage- and frequency-dependent manner. Furthermore, ranolazine (10 µM) blocked hNa(v)1.3 (expressed in HEK293 cells) and caused a hyperpolarizing shift in the voltage dependence of steady-state intermediate and slow inactivation Na(v)1.3 current. Taken together, the results suggest that ranolazine suppresses the hyperexcitability of DRG neurons by interacting with the inactivated states of Na(+) channels and these actions may contribute to its anti-allodynic effect in animal models of neuropathic pain.


Assuntos
Acetanilidas/farmacologia , Potenciais de Ação/fisiologia , Gânglios Espinais/embriologia , Gânglios Espinais/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.3/fisiologia , Piperazinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Células HEK293 , Humanos , Ranolazina , Ratos
13.
J Am Heart Assoc ; 1(5): e002360, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23316291

RESUMO

BACKGROUND: Despite the clear advantages of reperfusion in acute myocardial infarction, part of the myocardium is injured during reperfusion by reactive oxygen species. Reactive oxygen species activate apoptosis signal-regulating kinase-1, a key mediator in cell death. We hypothesized that inhibition of apoptosis signal-regulating kinase-1 at the time of reperfusion would protect the heart from ischemia-reperfusion injury. METHODS AND RESULTS: Male CD1 mice underwent transient coronary artery ligation (30 minutes) followed by reperfusion or underwent sham surgery (n=10 to 12 per group). A selective small-molecule inhibitor of apoptosis signal-regulating kinase-1 (GS-459679) was given immediately after reperfusion (10 or 30 mg/kg IP). Infarct size was measured early (at 24 hours, in a subgroup of mice) by triphenyl tetrazolium chloride staining and late (at 7 days) by Masson's trichrome staining for fibrosis. Apoptosis was assessed by measurement of caspase-3 activity and by determination of DNA fragmentation in cardiomyocytes bordering the infarct. Transthoracic echocardiography was performed before surgery and then at 24 hours and 7 days later. Treatment with GS-459679 at reperfusion led to a significant dose-related reduction in infarct size (31% for 10 mg/kg [P<0.001 versus vehicle] and 60% for 30 mg/kg [P<0.001 versus vehicle]), inhibition of apoptotic cell death, and preservation of left ventricular dimension and systolic function at both 24 hours and 7 days. CONCLUSIONS: Inhibition of apoptosis signal-regulating kinase-1 at the time of reperfusion limits infarct size and preserves left ventricular function in a model of acute myocardial infarction in the mouse.


Assuntos
MAP Quinase Quinase Quinase 5/antagonistas & inibidores , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Fragmentação do DNA/efeitos dos fármacos , Ecocardiografia , Masculino , Camundongos , Modelos Animais , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia
14.
Bioorg Med Chem ; 20(2): 996-1007, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22182575

RESUMO

In a preliminary article, we reported the potent allosteric enhancer activity at the A(1) adenosine receptor of a small series of 2-amino-3-(4-chlorobenzoyl)-4-[4-(aryl)piperazin-1-yl)methyl]thiophene derivatives bearing electron-withdrawing or electron-releasing groups at the para-position of the phenylpiperazine moiety. In the present study, we report the development of the compounds previously studied by modifying both the number and position of substituents on the phenylpiperazine moiety, aimed at establishing a structure-activity relationship identifying additional compounds with improved activity. The nature and the position of substituents on the phenyl ring tethered to the piperazine seemed to exert a fundamental influence on the allosteric enhancer activity, with the 3,4-difluoro 4i, 3-chloro-4-fluoro 4o, and 4-trifluoromethoxy 4ak derivatives being the most active compounds in binding (saturation and competition experiments) and functional cAMP studies. This study shows that it is also possible to obtain a good separation between allosteric enhancement and antagonistic activity at the A(1) adenosine receptor.


Assuntos
Antagonistas do Receptor A1 de Adenosina/química , Piperazinas/química , Receptor A1 de Adenosina/química , Tiofenos/química , Antagonistas do Receptor A1 de Adenosina/síntese química , Antagonistas do Receptor A1 de Adenosina/farmacologia , Regulação Alostérica , Animais , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Humanos , Ligação Proteica/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Relação Estrutura-Atividade , Tiofenos/síntese química , Tiofenos/farmacologia
15.
Am J Physiol Cell Physiol ; 302(8): C1141-51, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22189558

RESUMO

An increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) augments late sodium current (I(Na.L)) in cardiomyocytes. This study tests the hypothesis that both Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) and protein kinase C (PKC) mediate the effect of increased [Ca(2+)](i) to increase I(Na.L). Whole cell and open cell-attached patch clamp techniques were used to record I(Na.L) in rabbit ventricular myocytes dialyzed with solutions containing various concentrations of [Ca(2+)](i). Dialysis of cells with [Ca(2+)](i) from 0.1 to 0.3, 0.6, and 1.0 µM increased I(Na.L) in a concentration-dependent manner from 0.221 ± 0.038 to 0.554 ± 0.045 pA/pF (n = 10, P < 0.01) and was associated with an increase in mean Na(+) channel open probability and prolongation of channel mean open-time (n = 7, P < 0.01). In the presence of 0.6 µM [Ca(2+)](i), KN-93 (10 µM) and bisindolylmaleimide (BIM, 2 µM) decreased I(Na.L) by 45.2 and 54.8%, respectively. The effects of KN-93 and autocamtide-2-related inhibitory peptide II (2 µM) were not different. A combination of KN-93 and BIM completely reversed the increase in I(Na.L) as well as the Ca(2+)-induced changes in Na(+) channel mean open probability and mean open-time induced by 0.6 µM [Ca(2+)](i). Phorbol myristoyl acetate increased I(Na.L) in myocytes dialyzed with 0.1 µM [Ca(2+)](i); the effect was abolished by Gö-6976. In summary, both CaMKII and PKC are involved in [Ca(2+)](i)-mediated augmentation of I(Na.L) in ventricular myocytes. Inhibition of CaMKII and/or PKC pathways may be a therapeutic target to reduce myocardial dysfunction and cardiac arrhythmias caused by calcium overload.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Quinase C/metabolismo , Sódio/metabolismo , Animais , Benzilaminas/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Feminino , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/enzimologia , Peptídeos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Coelhos , Canais de Sódio/metabolismo , Sulfonamidas/farmacologia
16.
J Nucl Cardiol ; 19(1): 109-17, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22135160

RESUMO

BACKGROUND: Regadenoson is a coronary vasodilator that causes tachycardia via activation of the sympathetic nervous system. We determined whether ß(1)-adrenergic blockade can attenuate tachycardia without significantly reducing coronary vasodilation induced by regadenoson. METHODS AND RESULTS: Hemodynamics and coronary blood flow (CBF) were measured in conscious dogs. Baseline CBF and heart rate (HR) were 42 ± 2 mL/min and 87 ± 8 bpm (mean ± SEM), respectively. Regadenoson (1, 2.5, and 5 µg/kg) increased peak CBF by 129 ± 10, 149 ± 7, and 174 ± 10 mL/min and HR by 48 ± 6, 67 ± 5, and 85 ± 11 bpm, respectively, (all P < .05 vs baseline). In the presence of metoprolol (1.5 mg/kg), the peak increases in CBF caused by these three doses of regadenoson were reduced by only 11 ± 7%, 10 ± 4%, and 21 ± 2% (P = NS, <.05, and <.05 vs regadenoson alone), respectively, whereas the regadenoson-induced tachycardia was significantly reduced by 55 ± 8%, 55 ± 4%, and 52 ± 5% (all P < .05). In the presence of metoprolol, the duration of the regadenoson-induced increase in CBF was reduced, but the duration of the 2-fold increase in CBF caused by 5 µg/kg regadenoson was still nearly 6 minutes. CONCLUSION: ß(1)-Adrenergic blockade with metoprolol attenuated the regadenoson-induced increase in HR more than the increase in CBF.


Assuntos
Metoprolol/administração & dosagem , Purinas/efeitos adversos , Pirazóis/efeitos adversos , Taquicardia/induzido quimicamente , Taquicardia/prevenção & controle , Animais , Antiarrítmicos/administração & dosagem , Cães , Frequência Cardíaca/efeitos dos fármacos , Masculino , Taquicardia/fisiopatologia , Resultado do Tratamento , Vasodilatadores/efeitos adversos
17.
J Cardiovasc Pharmacol ; 57(6): 639-47, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21633249

RESUMO

Effects of ranolazine alone and in the presence of phenylephrine (PE) or isoproterenol (ISO) on hemodynamics, coronary blood flow and heart rate (HR) in the absence and presence of hexamethonium (a ganglionic blocker) were studied in conscious dogs. Ranolazine (0.4, 1.2, 3.6, and 6 mg/kg, intravenous) alone caused transient (<1 minute) and reversible hemodynamic changes. PE (0.3-10 µg/kg) caused a dose-dependent increase in blood pressure and decrease in HR. ISO (0.01-0.3 µg/kg) caused a dose-dependent decrease in blood pressure and an increase in HR. Ranolazine at high (11-13 mM), but not at moderate (4-5 mM) concentrations partially attenuated changes in mean arterial blood pressure and HR caused by either PE or ISO in normal conscious dogs. However, in dogs treated with hexamethonium (20 mg/kg) to cause autonomic blockade, ranolazine (both 4-5 and 11-13 µM) significantly attenuated both the PE- and ISO-induced changes in mean arterial blood pressure. The results suggest that a potential antiadrenergic effect of ranolazine was masked by autonomic control mechanisms in conscious dogs but could be observed when these mechanisms were inhibited (eg, in the hexamethonium-treated dog). Ranolazine, at plasma concentrations <10 µM and in conscious dogs with intact autonomic regulation, had minimal antiadrenergic (α and ß) effects.


Assuntos
Acetanilidas/uso terapêutico , Antagonistas Adrenérgicos/uso terapêutico , Hemodinâmica/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Hipotensão/tratamento farmacológico , Piperazinas/uso terapêutico , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Acetanilidas/administração & dosagem , Acetanilidas/sangue , Acetanilidas/metabolismo , Antagonistas Adrenérgicos/administração & dosagem , Antagonistas Adrenérgicos/sangue , Antagonistas Adrenérgicos/metabolismo , Animais , Fármacos do Sistema Nervoso Autônomo/administração & dosagem , Fármacos do Sistema Nervoso Autônomo/uso terapêutico , Circulação Coronária/efeitos dos fármacos , Cães , Relação Dose-Resposta a Droga , Bloqueadores Ganglionares/farmacologia , Cobaias , Frequência Cardíaca/efeitos dos fármacos , Hexametônio/farmacologia , Hipertensão/induzido quimicamente , Hipotensão/induzido quimicamente , Isoproterenol/administração & dosagem , Isoproterenol/toxicidade , Cinética , Fenilefrina/administração & dosagem , Fenilefrina/toxicidade , Piperazinas/administração & dosagem , Piperazinas/sangue , Piperazinas/metabolismo , Ranolazina , Ratos , Receptores Adrenérgicos alfa/química , Receptores Adrenérgicos beta/química , Vasoconstritores/administração & dosagem , Vasoconstritores/toxicidade , Vasodilatadores/administração & dosagem , Vasodilatadores/toxicidade
18.
Am J Physiol Cell Physiol ; 301(3): C577-86, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21677263

RESUMO

Late Na(+) current (I(NaL)) and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) are both increased in the diseased heart. Recently, CaMKII was found to phosphorylate the Na(+) channel 1.5 (Na(v)1.5), resulting in enhanced I(NaL). Conversely, an increase of I(NaL) would be expected to cause elevation of intracellular Ca(2+) and activation of CaMKII. However, a relationship between enhancement of I(NaL) and activation of CaMKII has yet to be demonstrated. We investigated whether Na(+) influx via Na(v)1.5 leads to CaMKII activation and explored the functional significance of this pathway. In neonatal rat ventricular myocytes (NRVM), treatment with the I(NaL) activators anemone toxin II (ATX-II) or veratridine increased CaMKII autophosphorylation and increased phosphorylation of CaMKII substrates phospholamban and ryanodine receptor 2. Knockdown of Na(v)1.5 (but not Na(v)1.1 or Na(v)1.2) prevented ATX-II-induced CaMKII phosphorylation, providing evidence for a specific role of Na(v)1.5 in CaMKII activation. In support of this view, CaMKII activity was also increased in hearts of transgenic mice overexpressing a gain-of-function Na(v)1.5 mutant (N(1325)S). The effects of both ATX-II and the N(1325)S mutation were reversed by either I(NaL) inhibition (with ranolazine or tetrodotoxin) or CaMKII inhibition (with KN93 or autocamtide 2-related inhibitory peptide). Furthermore, ATX-II treatment also induced CaMKII-Na(v)1.5 coimmunoprecipitation. The same association between CaMKII and Na(v)1.5 was also found in N(1325)S mice, suggesting a direct protein-protein interaction. Pharmacological inhibitions of either CaMKII or I(NaL) also prevented ATX-II-induced cell death in NRVM and reduced the incidence of polymorphic ventricular tachycardia induced by ATX-II in rat perfused hearts. Taken together, these results suggest that a Na(v)1.5-dependent increase in Na(+) influx leads to activation of CaMKII, which in turn phosphorylates Na(v)1.5, further promoting Na(+) influx. Pharmacological inhibition of either CaMKII or Na(v)1.5 can ameliorate cardiac dysfunction caused by excessive Na(+) influx.


Assuntos
Substituição de Aminoácidos/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Acetanilidas/farmacologia , Acetanilidas/uso terapêutico , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Venenos de Cnidários/farmacologia , Relação Dose-Resposta a Droga , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Feminino , Expressão Gênica/efeitos dos fármacos , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5 , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Perfusão , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , RNA Interferente Pequeno/genética , Coelhos , Ranolazina , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canais de Sódio/genética , Trocador de Sódio e Cálcio/antagonistas & inibidores , Trocador de Sódio e Cálcio/metabolismo , Taquicardia Ventricular/induzido quimicamente , Taquicardia Ventricular/prevenção & controle , Tetrodotoxina/farmacologia , Veratridina/farmacologia
19.
Circulation ; 123(16): 1713-20, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21482963

RESUMO

BACKGROUND: The reverse rate dependence (RRD) of actions of I(Kr)-blocking drugs to increase the action potential duration (APD) and beat-to-beat variability of repolarization (BVR) of APD is proarrhythmic. We determined whether inhibition of endogenous, physiological late Na(+) current (late I(Na)) attenuates the RRD and proarrhythmic effect of I(Kr) inhibition. METHODS AND RESULTS: Duration of the monophasic APD (MAPD) was measured from female rabbit hearts paced at cycle lengths from 400 to 2000 milliseconds, and BVR was calculated. In the absence of a drug, duration of monophasic action potential at 90% completion of repolarization (MAPD(90)) and BVR increased as the cycle length was increased from 400 to 2000 milliseconds (n=36 and 26; P<0.01). Both E-4031 (20 nmol/L) and d-sotalol (10 µmol/L) increased MAPD(90) and BVR at all stimulation rates, and the increase was greater at slower than at faster pacing rates (n=19, 11, 12 and 7, respectively; P<0.01). Tetrodotoxin (1 µmol/L) and ranolazine significantly attenuated the RRD of MAPD(90,) reduced BVR (P<0.01), and abolished torsade de pointes in hearts treated with either 20 nmol/L E-4031 or 10 µmol/L d-sotalol. Endogenous late I(Na) in cardiomyocytes stimulated at cycle lengths from 500 to 4000 milliseconds was greater at slower than at faster stimulation rates, and rapidly decreased during the first several beats at faster but not at slower rates (n=8; P<0.01). In a computational model, simulated RRD of APD caused by E-4031 and d-sotalol was attenuated when late I(Na) was inhibited. CONCLUSION: Endogenous late I(Na) contributes to the RRD of I(Kr) inhibitor-induced increases in APD and BVR and to bradycardia-related ventricular arrhythmias.


Assuntos
Potenciais de Ação/fisiologia , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Canais de Sódio/fisiologia , Torsades de Pointes/fisiopatologia , Acetanilidas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Antiarrítmicos/farmacologia , Bradicardia/tratamento farmacológico , Bradicardia/fisiopatologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/fisiopatologia , Modelos Cardiovasculares , Contração Miocárdica/efeitos dos fármacos , Técnicas de Patch-Clamp , Piperazinas/farmacologia , Piperidinas/farmacologia , Piridinas/farmacologia , Coelhos , Ranolazina , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Torsades de Pointes/tratamento farmacológico
20.
J Pharmacol Exp Ther ; 337(2): 513-23, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21325441

RESUMO

Inhibition by cardiac glycosides of Na(+), K(+)-ATPase reduces sodium efflux from myocytes and may lead to Na(+) and Ca(2+) overload and detrimental effects on mechanical function, energy metabolism, and electrical activity. We hypothesized that inhibition of sodium persistent inward current (late I(Na)) would reduce ouabain's effect to cause cellular Na(+) loading and its detrimental metabolic (decrease of ATP) and functional (arrhythmias, contracture) effects. Therefore, we determined effects of ouabain on concentrations of intracellular sodium (Na(+)(i)) and high-energy phosphates using (23)Na and (31)P NMR, the amplitude of late I(Na) using the whole-cell patch-clamp technique, and contractility and electrical activity of guinea pig isolated hearts, papillary muscles, and ventricular myocytes in the absence and presence of inhibitors of late I(Na). Ouabain (1-1.3 µM) increased Na(+)(i) and late I(Na) of guinea pig isolated hearts and myocytes by 3.7- and 4.2-fold, respectively. The late I(Na) inhibitors ranolazine and tetrodotoxin significantly reduced ouabain-stimulated increases in Na(+)(i) and late I(Na). Reductions of ATP and phosphocreatine contents and increased diastolic tension in ouabain-treated hearts were also markedly attenuated by ranolazine. Furthermore, the ouabain-induced increase of late I(Na) was also attenuated by the Ca(2+)-calmodulin-dependent kinase I inhibitors KN-93 [N-[2-[[[3-(4-chlorophenyl)-2-propenyl]methylamino]methyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulphonamide] and autocamide-2 related inhibitory peptide, but not by KN-92 [2-[N-(4'-methoxybenzenesulfonyl)]amino-N-(4'-chlorophenyl)-2-propenyl-N-methylbenzylamine phosphate]. We conclude that ouabain-induced Na(+) and Ca(2+) overload is ameliorated by the inhibition of late I(Na).


Assuntos
Inibidores Enzimáticos/farmacologia , Coração/fisiologia , Ouabaína/farmacologia , Canais de Sódio/fisiologia , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Acetanilidas/administração & dosagem , Acetanilidas/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fenômenos Eletrofisiológicos , Metabolismo Energético/efeitos dos fármacos , Feminino , Cobaias , Testes de Função Cardíaca , Espectroscopia de Ressonância Magnética , Masculino , Contração Miocárdica/efeitos dos fármacos , Miocárdio/química , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Músculos Papilares/efeitos dos fármacos , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Ranolazina , Sódio/análise , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/administração & dosagem , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/administração & dosagem , Tetrodotoxina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...