Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 377(2): 293-304, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33602875

RESUMO

In failing hearts, Na+/Ca2+ exchanger (NCX) overactivity contributes to Ca2+ depletion, leading to contractile dysfunction. Inhibition of NCX is expected to normalize Ca2+ mishandling, to limit afterdepolarization-related arrhythmias, and to improve cardiac function in heart failure (HF). SAR340835/SAR296968 is a selective NCX inhibitor for all NCX isoforms across species, including human, with no effect on the native voltage-dependent calcium and sodium currents in vitro. Additionally, it showed in vitro and in vivo antiarrhythmic properties in several models of early and delayed afterdepolarization-related arrhythmias. Its effect on cardiac function was studied under intravenous infusion at 250,750 or 1500 µg/kg per hour in dogs, which were either normal or submitted to chronic ventricular pacing at 240 bpm (HF dogs). HF dogs were infused with the reference inotrope dobutamine (10 µg/kg per minute, i.v.). In normal dogs, NCX inhibitor increased cardiac contractility (dP/dtmax) and stroke volume (SV) and tended to reduce heart rate (HR). In HF dogs, NCX inhibitor significantly and dose-dependently increased SV from the first dose (+28.5%, +48.8%, and +62% at 250, 750, and 1500 µg/kg per hour, respectively) while significantly increasing dP/dtmax only at 1500 (+33%). Furthermore, NCX inhibitor significantly restored sympathovagal balance and spontaneous baroreflex sensitivity (BRS) from the first dose and reduced HR at the highest dose. In HF dogs, dobutamine significantly increased dP/dtmax and SV (+68.8%) but did not change HR, sympathovagal balance, or BRS. Overall, SAR340835, a selective potent NCX inhibitor, displayed a unique therapeutic profile, combining antiarrhythmic properties, capacity to restore systolic function, sympathovagal balance, and BRS in HF dogs. NCX inhibitors may offer new therapeutic options for acute HF treatment. SIGNIFICANCE STATEMENT: HF is facing growing health and economic burden. Moreover, patients hospitalized for acute heart failure are at high risk of decompensation recurrence, and no current acute decompensated HF therapy definitively improved outcomes. A new potent, Na+/Ca2+ exchanger inhibitor SAR340835 with antiarrhythmic properties improved systolic function of failing hearts without creating hypotension, while reducing heart rate and restoring sympathovagal balance. SAR340835 may offer a unique and attractive pharmacological profile for patients with acute heart failure as compared with current inotrope, such as dobutamine.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Moduladores de Transporte de Membrana/uso terapêutico , Trocador de Sódio e Cálcio/antagonistas & inibidores , Nervo Vago/efeitos dos fármacos , Animais , Barorreflexo , Cães , Coração/efeitos dos fármacos , Frequência Cardíaca , Moduladores de Transporte de Membrana/administração & dosagem , Moduladores de Transporte de Membrana/farmacologia , Contração Miocárdica , Miocárdio/metabolismo , Suínos
2.
Naunyn Schmiedebergs Arch Pharmacol ; 385(7): 717-27, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526470

RESUMO

ADP-ribosyl cyclases (ADPRCs) catalyse the conversion of nicotinamide adenine dinucleotide to cyclic adenosine diphosphoribose (cADPR) which is a second messenger involved in Ca(2+) mobilisation from intracellular stores. Via its interaction with the ryanodine receptor Ca(2+) channel in the heart, cADPR may exert arrhythmogenic activity. To test this hypothesis, we have studied the effect of novel cardiac ADPRC inhibitors in vitro and in vivo in models of ventricular arrhythmias. Using a high-throughput screening approach on cardiac sarcoplasmic reticulum membranes isolated from pig and rat and nicotinamide hypoxanthine dinuleotide as a surrogate substrate, we have identified potent and selective inhibitors of an intracellular, membrane-bound cardiac ADPRC that are different from the two known mammalian ADPRCs, CD38 and CD157/Bst1. We show that two structurally distinct cardiac ADPRC inhibitors, SAN2589 and SAN4825, prevent the formation of spontaneous action potentials in guinea pig papillary muscle in vitro and that compound SAN4825 is active in vivo in delaying ventricular fibrillation and cardiac arrest in a guinea pig model of Ca(2+) overload-induced arrhythmia. Inhibition of cardiac ADPRC prevents Ca(2+) overload-induced spontaneous depolarizations and ventricular fibrillation and may thus provide a novel therapeutic principle for the treatment of cardiac arrhythmias.


Assuntos
ADP-Ribosil Ciclase/antagonistas & inibidores , Antiarrítmicos/farmacologia , Inibidores Enzimáticos/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , ADP-Ribosil Ciclase/metabolismo , Animais , Cálcio/fisiologia , Cobaias , Coração/fisiologia , Técnicas In Vitro , Miocárdio/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Músculos Papilares/efeitos dos fármacos , Músculos Papilares/fisiologia , Ratos , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/fisiologia , Suínos , Fibrilação Ventricular/tratamento farmacológico , Fibrilação Ventricular/fisiopatologia
3.
Naunyn Schmiedebergs Arch Pharmacol ; 385(4): 373-84, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22234329

RESUMO

Amiodarone is one of the most effective antiarrhythmic drugs. However, poor solubility of this compound has limited its intravenous application. SAR11464A is a water-soluble amiodarone-like drug that lacks iodine and inhibits multiple cardiac ion channels in vitro. This study evaluated the antiarrhythmic efficacy of this drug in vivo. In porcine studies, atrial effective refractory period (AERP) was measured in pentobarbital-anesthetized thoracotomized pigs and atrial fibrillation (AF) was induced by a premature beat. Ventricular fibrillation (VF) was induced via either burst pacing or programmed electrical stimulation (a series of progressively shorter beats, S1-S5). In canine studies, VF was induced by a 2-min occlusion of the left circumflex coronary artery during the last minute of exercise in dogs with healed myocardial infarctions (n = 8). One week later, this test was repeated after pretreatment with SAR114646A (3.0 mg/kg, i.v., slow bolus). SAR114646A produced a significant dose-dependent prolongation of AERP, inhibited AF induced by a premature stimulus, and electrically induced VF in anesthetized pigs. At 1.0 and 3.0 mg/kg, i.v., it was superior to amiodarone, dofetilide, and flecainide. In dogs, SAR114646A did not alter any ECG parameter including QTc (control, 236.9 ± 8.5 ms vs. SAR, 237.2 ± 3.5 ms) but significantly reduced the incidence of VF, protecting six of eight animals (Fisher's exact test, P = 0.01). SAR114646A was effective against both atrial and ventricular arrhythmias without altering ventricular repolarization. These data suggest that the amiodarone-like drug SAR114646A may be an effective antiarrhythmic intervention that does not adversely prolong ventricular repolarization.


Assuntos
Amiodarona/análogos & derivados , Antiarrítmicos/uso terapêutico , Fibrilação Atrial/tratamento farmacológico , Átrios do Coração/efeitos dos fármacos , Fibrilação Ventricular/tratamento farmacológico , Amiodarona/uso terapêutico , Animais , Fibrilação Atrial/fisiopatologia , Modelos Animais de Doenças , Cães , Estimulação Elétrica , Eletrocardiografia , Feminino , Átrios do Coração/fisiopatologia , Masculino , Suínos , Fibrilação Ventricular/fisiopatologia
4.
J Cardiovasc Pharmacol ; 51(4): 380-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18427281

RESUMO

OBJECTIVE: Atrial-selective drug therapy represents a novel therapeutic approach for atrial fibrillation management. The aim of the present study was to investigate the mechanism of hKv1.5 channel inhibition by the atrial-selective compound AVE1231. METHODS: Ionic currents were recorded from CHO cells transfected with KCNA5 cDNA with whole-cell patch-clamp technique. The effect of AVE1231 on human atrial cell action potentials was explored with a computer model. RESULTS: KCNA5 expression resulted in typical K currents that activated and inactivated voltage dependently. Ascending concentrations of AVE1231 (0.1-100 microM) led to concentration- and voltage-dependent current inhibition (IC50 at +40 mV: 2.0 +/- 0.5 microM, Hill coefficient 0.69 +/- 0.12). Acceleration of hKv1.5 current inactivation occurred with increasing AVE1231 concentrations, indicating channel inhibition in the open state (eg, taufast at +40 mV: 318 +/- 92 milliseconds under control; 14 +/- 1 milliseconds with 3 microM, P < 0.05). Using 1/taufast as an approximation of the time course of drug-channel interaction, association rate (K+1) and dissociation rate (K-1) constants were 8.18 x 10 M/s and 45.95 seconds, respectively (KD = 5.62 microM). The onset of current inhibition occurred more rapidly with higher concentrations along with a prominent tail current crossover phenomenon after AVE1231 application. Drug inhibition remained effective through a range of stimulation frequencies. Computer modeling suggested more pronounced prolongation of action potential duration under conditions of atrial remodeling. CONCLUSION: AVE1231 is an inhibitor of hKv1.5 currents with predominant action on channels in their open state; thus, it may be suitable for the treatment of AF.


Assuntos
Antiarrítmicos/farmacologia , Canal de Potássio Kv1.5/antagonistas & inibidores , Potenciais de Ação , Animais , Antiarrítmicos/uso terapêutico , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Humanos , Modelos Cardiovasculares , Técnicas de Patch-Clamp
5.
J Cardiovasc Pharmacol ; 49(4): 197-206, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17438404

RESUMO

The novel compound AVE1231 was investigated in order to elucidate its potential against atrial fibrillation. In CHO cells, the current generated by hKv1.5 or hKv4.3 + KChIP2.2b channels was blocked with IC50 values of 3.6 microM and 5.9 microM, respectively. In pig left atrial myocytes, a voltage-dependent outward current was blocked with an IC50 of 1.1 microM, mainly by accelerating the time constant of decay. Carbachol-activated IKACh was blocked by AVE1231 with an IC50 of 8.4 microM. Other ionic currents, like the IKr, IKs, IKATP, ICa, and INa were only mildly affected by 10 microM AVE1231. In guinea pig papillary muscle the APD90 and the upstroke velocity were not significantly altered by 30 microM AVE1231. In anesthetized pigs, oral doses of 0.3, 1, and 3 mg/kg AVE1231 caused a dose-dependent increase in left atrial refractoriness (LAERP), associated by inhibition of left atrial vulnerability to arrhythmia. There were no effects on the ECG intervals, ventricular monophasic action potentials, or ventricular refractory periods at 3 mg/kg AVE1231 applied intravenously. In conscious goats, both AVE1231 (3 mg/kg/h iv) and dofetilide (10 microg/kg/h iv) significantly prolonged LAERP. After 72 hours of tachypacing, when LAERP was shortened significantly (electrical remodelling), the prolongation of LAERP induced by AVE1231 was even more pronounced than in sinus rhythm. In contrast, the effect of dofetilide was strongly decreased. The present data demonstrate that AVE1231 blocks early atrial K channels and prolongs atrial refractoriness with no effects on ECG intervals and ventricular repolarisation, suggesting that it is suited for the prevention of atrial fibrillation in patients.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Arritmias Cardíacas/fisiopatologia , Função Atrial/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Células CHO , Carbacol/farmacologia , Cardiotônicos/farmacologia , Cricetinae , Cricetulus , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Técnicas Eletrofisiológicas Cardíacas , Feminino , Cabras , Cobaias , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/fisiopatologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Humanos , Concentração Inibidora 50 , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/metabolismo , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Músculos Papilares/efeitos dos fármacos , Músculos Papilares/fisiopatologia , Técnicas de Patch-Clamp , Fenetilaminas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Período Refratário Eletrofisiológico/efeitos dos fármacos , Sulfonamidas/farmacologia , Suínos , Fatores de Tempo
6.
J Pharmacol Exp Ther ; 319(2): 898-906, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16916995

RESUMO

Cardiac electrical activity is modulated by potassium currents. Pigs have been used for antiarrhythmic drug testing, but only sparse data exist regarding porcine atrial ionic electrophysiology. Here, we used electrophysiological, molecular, and pharmacological tools to characterize a prominent porcine outward K(+) current (I(K,PO)) in atrial cardiomyocytes isolated from adult pigs. I(K,PO) activated rapidly (time to peak at +60 mV; 2.1 +/- 0.2 ms), inactivated slowly (tau(f) = 45 +/- 10; tau(s) = 215 +/- 28 ms), and showed very slow recovery (tau(f) = 1.54 +/- 0.73 s; tau(s) = 7.91 +/- 1.78 s; n = 9; 36 degrees C). Activation and inactivation were voltage-dependent, and current properties were consistent with predominant K(+) conductance. Neurotoxins (heteropodatoxin, hongatoxin, and blood depressing substance) that block K(v)4.x, K(v)1.1, -1.2, -1.3, and -3.4 in a highly selective manner as well as H(2)O(2) and tetraethylammonium, did not affect the current. Drugs with K(v)1.5-blocking properties (flecainide, perhexiline, and the novel atrial-selective antiarrhythmic 2'-{2-(4-methoxyphenyl)-acetylamino-methyl}-biphenyl-2-carboxylic acid (2-pyridin-3-yl-ethyl)-amide; AVE0118) inhibited I(K,PO) (IC(50) of 132 +/- 47, 17 +/- 10, and 1.25 +/- 0.62 microM, respectively). 4-Aminopyridine suppressed the current and accelerated its decay, reducing charge carriage with an IC(50) of 39 +/- 15 microM. Porcine-specific K(v) channel subunit sequences were cloned to permit real-time quantitative reverse transcription-polymerase chain reaction on RNA extracted from isolated cardiomyocytes, which showed much greater abundance of K(v)1.5 mRNA compared with K(v)1.4, K(v)4.2, and K(v)4.3. Action potential recordings showed that I(K,PO) inhibition with 0.1 mM 4-AP delayed repolarization (e.g., action potential duration at -50 mV increased from 45 +/- 9 to 69 +/- 5 ms at 3 Hz; P < 0.05). In conclusion, porcine atrium displays a current that is involved in repolarization, inactivates more slowly than classic transient outward current, is associated with strong K(v)1.5 expression, and shows a pharmacological profile typical of K(v)1.5-dependent currents.


Assuntos
Átrios do Coração/metabolismo , Canal de Potássio Kv1.5/fisiologia , 4-Aminopiridina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Cloretos/farmacologia , Flecainida/farmacologia , Canal de Potássio Kv1.5/efeitos dos fármacos , Masculino , Neurotoxinas/farmacologia , Perexilina , Potássio/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Fatores de Tempo
7.
Naunyn Schmiedebergs Arch Pharmacol ; 370(3): 183-92, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15340774

RESUMO

Previous studies in pigs and goats have demonstrated that AVE0118 prolongs atrial refractoriness without any effect on the QT-interval. The purpose of the present study was to investigate the effect of the compound on various cardiac ion channels. AVE0118 blocked the pig Kv1.5 and the human Kv1.5 expressed in Xenopus oocytes with IC(50) values of 5.4+/-0.7 microM and 6.2+/-0.4 microM respectively. In Chinese hamster ovary (CHO) cells, AVE0118 decreased the steady-state hKv1.5 current with an IC(50) of 1.1+/-0.2 microM. The hKv4.3/KChIP2.2 current in CHO cells was blocked by AVE0118 by accelerating the apparent time-constant of inactivation ( tau(inact)), and the integral current was inhibited with an IC(50) of 3.4+/-0.5 microM. At 10 microM AVE0118 tau(inact) decreased from 9.3+/-0.6 ms ( n=8, control) to 3.0+/-0.3 ms ( n=8). The K(ACh) current was investigated in isolated pig atrial myocytes by application of 10 microM carbachol. At a clamp potential of -100 mV the I(KACh) was half-maximally blocked by 4.5+/-1.6 microM AVE0118. In the absence of carbachol, AVE0118 had no effect on the inward current recorded at -100 mV. Effects on the I(Kr) current were investigated on HERG channels expressed in CHO cells. AVE0118 blocked this current half-maximally at approximately 10 microM. Comparable results were obtained in isolated guinea pig ventricular myocytes, where half-maximal inhibition of the I(Kr) tail current occurred at a similar concentration of AVE0118. Other ionic currents, like the I(Ks), I(KATP) (recorded in guinea pig ventricular myocytes), and L-type Ca(2+) (recorded in pig atrial myocytes) were blocked by 10 microM AVE0118 by 10+/-3% ( n=6), 28+/-7% ( n=4), and 22+/-13% ( n=5) respectively. In summary, AVE0118 preferentially inhibits the atrial K(+) channels I(Kur), I(to) and I(KACH). This profile may explain the selective prolongation of atrial refractoriness described previously in pigs and goats.


Assuntos
Antiarrítmicos/farmacologia , Carbacol/farmacologia , Cardiotônicos/farmacologia , Canais Iônicos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Animais , Células CHO , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Células Cultivadas , Cricetinae , Cricetulus , Eletrofisiologia , Humanos , Proteínas Interatuantes com Canais de Kv , Canal de Potássio Kv1.5 , Biologia Molecular , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio Shal , Suínos , Xenopus
8.
Cardiovasc Drugs Ther ; 18(1): 23-30, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15115900

RESUMO

PURPOSE: The progressive shortening of the atrial effective refractory period (ERP) during atrial fibrillation (AF) might be due to the activation of the KATP channels by rapid atrial rates. We tested the hypothesis that the cardioselective, sarcolemmal KATP channel blocker HMR 1098 would prevent atrial ERP shortening during AF. METHODS AND RESULTS: Nine dogs were treated with HMR 1098 (3 mg/kg bolus injection followed by a continuous intravenous (i.v.) infusion at 17 microg/kg/min rate maintained throughout the study) and 7 dogs served as controls receiving i.v. saline. Pharmacological autonomic blockade was induced by i.v. administration of atropine (0.04 mg/kg) and propranolol (0.2 mg/kg) and maintained throughout the study by continuous i.v. infusion of atropine (0.007 mg/kg/h) and propranolol (0.04 mg/kg/h). Rapid right atrial pacing at 50 msec cycle length (CL) was initiated and maintained for 6 hours. High right atrial ERP (HRA-ERP) and corrected sinus node recovery time (HRA-cSNRT), coronary sinus ERP (CS-ERP) and corrected SNRT (CS-cSNRT) at three (400, 300, 200 msec) CLs were measured before and after pacing at different time points. Baseline values were not different between control and treated dogs. In the control group the HRA-ERP progressively shortened (from 179 +/- 21 msec at baseline to 161 +/- 23 msec at 360 min at 400 msec CL) ( p = 0.002), with a gradual decrease, loss or inversion of ERP rate adaptation at shorter (300, 200 msec) CLs. HMR 1098 treatment did not prevent the shortening of HRA-ERP during the first 2 to 3 hours of rapid atrial pacing. However, beginning at 180-240 min, HMR 1098 increased the HRA-ERP ( p = 0.01) to baseline by 360 min. HMR 1098 treatment did not prevent another feature of atrial electrical remodeling, the flattening or inversion of ERP rate adaptation. In neither group did CS-ERP shortening occur. The maximum cSNRT at 360 min prolonged significantly in both groups during HRA and CS pacing as well compared with baseline. CONCLUSIONS: HMR 1098 treatment did not prevent the shortening of HRA-ERP, the salient feature of atrial electrical remodeling in the first 2 to 3 hours of rapid atrial rates, but did prevent it thereafter. Another characteristic feature of atrial electrical remodeling, the flattening or inversion of physiological ERP rate adaptation was not prevented by HMR 1098 treatment. Sinus node depression was detectable after short-term (6 hours) rapid atrial pacing and was not affected by HMR 1098.


Assuntos
Fibrilação Atrial/fisiopatologia , Função do Átrio Direito/efeitos dos fármacos , Benzamidas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Período Refratário Eletrofisiológico/efeitos dos fármacos , Trifosfato de Adenosina/fisiologia , Animais , Cães , Feminino , Masculino , Sarcolema/metabolismo
9.
J Pharmacol Exp Ther ; 309(1): 182-92, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14722326

RESUMO

In the present study, a novel sulfonylthiourea, 1-[[5-[2-(5-chloro-o-anisamido)ethyl]-beta-methoxyethoxyphenyl]sulfonyl]-3-methylthiourea, sodium salt (HMR 1402), was investigated using in vitro and in vivo systems. HMR 1402 inhibited rilmakalim-induced currents in rat and guinea pig myocytes (IC(50) = 60 and 509 nM, respectively). Hypoxia-induced shortening of action potential duration at 90% repolarization was also significantly attenuated by HMR 1402 (68.1 +/- 3.9% of control at 0.3 microM). In contrast, HMR 1402 had a smaller effect on pancreatic beta-cells (rat insuloma cells, RINm5F) hyperpolarized with 100 microM diazoxide (IC(50) = 3.9 microM, compared with glibenclamide IC(50) = 9 nM). In a similar manner, hypoxia induced increases in coronary flow in isolated guinea pig hearts were only slightly reduced by HMR 1402. These data strongly suggest that HMR 1402 has pharmacological selectivity for cardiac myocytes and, therefore, may protect against ischemically induced ventricular fibrillation (VF) without the untoward effects of nonselective compounds. To test this hypothesis, VF was induced in 8 dogs with healed myocardial infarctions by a 2-min coronary occlusion during the last minute of exercise. On a subsequent day, the exercise plus ischemia test was repeated after HMR 1402 (3.0 mg/kg i.v., n = 4, infusion 4 microg/kg/min for 1 h before exercise, n = 4). This drug significantly reduced the incidence of VF protecting seven of eight animals (p = 0.0007) without altering plasma insulin, blood glucose, or the increases in mean coronary blood flow induced by either exercise or 15-s coronary occlusions. Thus, the ATP-sensitive potassium channel antagonist HMR 1402 can prevent ischemically induced VF without altering coronary blood flow or blood glucose.


Assuntos
Coração/efeitos dos fármacos , Isquemia Miocárdica/complicações , Músculos Papilares/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Tioureia/farmacologia , Fibrilação Ventricular/prevenção & controle , Transportadores de Cassetes de Ligação de ATP , Animais , Cães , Feminino , Cobaias , Hiperemia/prevenção & controle , Ilhotas Pancreáticas/efeitos dos fármacos , Canais KATP , Masculino , Músculos Papilares/fisiologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/uso terapêutico , Canais de Potássio Corretores do Fluxo de Internalização , Ratos , Ratos Sprague-Dawley , Tioureia/análogos & derivados , Fibrilação Ventricular/etiologia
10.
Naunyn Schmiedebergs Arch Pharmacol ; 367(3): 281-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12644901

RESUMO

PURPOSE: The chromanol HMR 1556 is a potent blocker of KvLQT1/minK potassium channels expressed in Xenopus oocytes. The compound is therefore a new class III antiarrhythmic drug with a distinct mechanism of action. However, the effect of HMR 1556 on atrial ion channels and the selectivity of block in the human heart has not been investigated. We tested the effects of HMR 1556 on repolarizing potassium currents in human and guinea pig atrial myocytes. METHODS AND RESULTS: Single atrial myocytes were isolated by enzymatic dissociation. Atrial potassium currents (I(Ks), I(Kr), in guinea pig, I(to), I(Kur), I(K1) in humans) were recorded at 36 degrees C in the whole cell mode of the patch clamp technique. HMR 1556 produced a concentration-dependent and reversible block of I(Ks) with a half maximal concentration (EC(50)) of 6.8 nmol/l. 10 micromol/l HMR 1556 almost completely inhibited I(Ks) (97.2+/-3.2%, n=6). Steady-state activation as well as kinetic properties of the current were not altered by HMR 1556. I(Kr) currents were not affected up to concentrations of 10 micromol/l. HMR 1556 did not inhibit other potassium currents in human atrium: I(to), I(Kur) and the classical inward rectifier potassium current I(K1) were not significantly affected up to concentrations that completely blocked I(Ks) (10 micromol/l). CONCLUSIONS: HMR 1556 is a highly-potent blocker of I(Ks) channels without exerting effects on other potassium currents involved in atrial repolarization. Given the potential advantages of I(Ks) vs. I(Kr) blockade, the drug's new mechanism of action warrants further investigation to clarify its role as an antiarrhythmic agent.


Assuntos
Cromanos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/efeitos dos fármacos , Sulfonamidas/farmacologia , Idoso , Animais , Células Cultivadas , Canais de Potássio de Retificação Tardia , Feminino , Cobaias , Átrios do Coração/citologia , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos
11.
Cardiovasc Drugs Ther ; 17(5-6): 435-41, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15107598

RESUMO

PURPOSE: Activation of the myocardial, ATP-dependent potassium current (IK(ATP)) during ischemia causes shortening of the action potential duration thereby increasing dispersion of repolarization between ischemic and non-ischemic myocardium and predisposing to reentrant arrhythmias. The IK(ATP) inhibitor HMR1098 allows selective block of the sarcolemmal myocardial K(ATP)-channel in various animal species. Therefore, we studied the concentration and pH-dependence of HMR1098 in human ventricular myocytes. METHODS: Human ventricular cardiomyocytes were isolated enzymatically. IK(ATP) was measured with the patch-clamp technique in whole cell configuration at 35 degrees C. Action potentials were recorded using Amphotericine B in perforated patch conditions. In voltage clamp experiments, the K(ATP)-channel was activated by application of 1 microM rilmakalim, a K(ATP)-channel opener. In action potential recordings, 0.1 microM rilmakalim was used. RESULTS: At physiological pH (pH = 7.3) half-maximal block of the rilmakalim-induced current occurred at 0.42 +/- 0.008 microM HMR1098 (at 0 mV membrane potential); under acidic conditions as can be expected to be present under ischemic conditions (pH = 6.5), half-maximal block was achieved at markedly lower concentrations (IC(50) = 0.24 +/- 0.009 microM). In current clamp experiments, block of IK(ATP) by HMR1098 was capable of reversing the action potential shortening induced by rilmakalim, and restored the action potential plateau. CONCLUSIONS: HMR1098 appears to be useful to prevent IK(ATP)-induced shortening of the action potential in human ventricular myocardium. More acidic conditions, as observed in ischemia, increase the sensitivity to HMR1098, indicating a more potent effect in ischemic myocardium. Thus, HMR1098 may be a useful agent to prevent action potential shortening and dispersion of repolarization during ischemia, which may protect against ischemia induced ventricular arrhythmias.


Assuntos
Trifosfato de Adenosina/fisiologia , Antiarrítmicos/farmacologia , Benzamidas/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Sarcolema/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Glibureto/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Sarcolema/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...