Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Zhonghua Xin Xue Guan Bing Za Zhi ; 50(11): 1087-1093, 2022 Nov 24.
Artigo em Chinês | MEDLINE | ID: mdl-36418277

RESUMO

Objective: To determine the electrophysiological effects and related mechanisms of late sodium current inhibitors on hearts with short QT intervals. Methods: The electrophysiological study was performed on isolated Langendorff perfused rabbit hearts. A total of 80 New Zealand White rabbits were used and 34 hearts without drug treatment were defined as control group A, these hearts were then treated with IKATP opener pinacidil, defined as pinacidil group A. Then, 27 hearts from pinacidil group A were selected to receive combined perfusion with sodium channel inhibitors or quinidine, a traditional drug used to treat short QT syndrome, including ranolazine combined group (n=9), mexiletine combined group (n=9), and quinidine combined group (n=9). Nineteen out of the remaining 46 New Zealand rabbits were selected as control group B (no drug treatments, n=19), and then treated with pinacidil, defined as pinacidil group B (n=19). The remaining 27 rabbits were treated with sodium inhibitors or quinidine alone, including ranolazine alone group (n=9), mexiletine alone group (n=9), and quinidine alone group (n=9). Electrocardiogram (ECG) physiological parameters of control group A and pinacidil group A were collected. In control group B and pinacidil group B, programmed electrical stimulation was used to induce ventricular arrhythmias and ECG was collected. ECG physiological parameters and ventricular arrhythmia status of various groups were analyzed. The concentrations of pinacidil, ranolazine, mexiletine and quinidine used in this study were 30, 10, 30 and 1 µmol/L, respectively. Results: Compared with control group A, the QT interval, 90% of the repolarization in epicardial and endocardial monophasic action potential duration (MAPD90-Epi, MAPD90-Endo) was shortened, the transmural dispersion of repolarization (TDR) was increased, and the effective refractor period (ERP) and post-repolarization refractoriness (PRR) were reduced in pinacidil group A (all P<0.05). Compared with the pinacidil group A, MAPD90-Epi, MAPD90-Endo, QT interval changes were reversed in quinidine combined group and mexiletine combined group (all P<0.05), but not in ranolazine combined group. All these three drugs reversed the pinacidil-induced increases of TDR and the decreases of ERP and PRR. The induced ventricular arrhythmia rate was 0 in control group B, and increased to 10/19 (χ2=13.6, P<0.05) in pinacidil group B during programmed electrical stimulation. Compared with the pinacidil group B, incidences of ventricular arrhythmia decreased to 11% (1/9), 11% (1/9) and 0 (0/9) (χ2=4.5, 4.5, 7.4, P<0.05) respectively in ranolazine group, mexiletine group and quinidine group. Conclusions: Inhibition of late sodium current does not increase but even decreases the risk of malignant arrhythmia in hearts with a shortened QT interval. The antiarrhythmic mechanism might be associated with the reversal of the increase of TDR and the decrease of refractoriness (including both ERP and PRR) of hearts with shortened QT interval.


Assuntos
Mexiletina , Quinidina , Coelhos , Animais , Quinidina/farmacologia , Quinidina/uso terapêutico , Mexiletina/farmacologia , Mexiletina/uso terapêutico , Pinacidil/farmacologia , Pinacidil/uso terapêutico , Sódio , Ranolazina/farmacologia , Ranolazina/uso terapêutico , Técnicas Eletrofisiológicas Cardíacas , Arritmias Cardíacas/tratamento farmacológico
2.
PLoS Comput Biol ; 17(2): e1008089, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33591962

RESUMO

Short QT (SQT) syndrome is a genetic cardiac disorder characterized by an abbreviated QT interval of the patient's electrocardiogram. The syndrome is associated with increased risk of arrhythmia and sudden cardiac death and can arise from a number of ion channel mutations. Cardiomyocytes derived from induced pluripotent stem cells generated from SQT patients (SQT hiPSC-CMs) provide promising platforms for testing pharmacological treatments directly in human cardiac cells exhibiting mutations specific for the syndrome. However, a difficulty is posed by the relative immaturity of hiPSC-CMs, with the possibility that drug effects observed in SQT hiPSC-CMs could be very different from the corresponding drug effect in vivo. In this paper, we apply a multistep computational procedure for translating measured drug effects from these cells to human QT response. This process first detects drug effects on individual ion channels based on measurements of SQT hiPSC-CMs and then uses these results to estimate the drug effects on ventricular action potentials and QT intervals of adult SQT patients. We find that the procedure is able to identify IC50 values in line with measured values for the four drugs quinidine, ivabradine, ajmaline and mexiletine. In addition, the predicted effect of quinidine on the adult QT interval is in good agreement with measured effects of quinidine for adult patients. Consequently, the computational procedure appears to be a useful tool for helping predicting adult drug responses from pure in vitro measurements of patient derived cell lines.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/fisiopatologia , Avaliação Pré-Clínica de Medicamentos/métodos , Sistema de Condução Cardíaco/anormalidades , Cardiopatias Congênitas/tratamento farmacológico , Cardiopatias Congênitas/fisiopatologia , Modelos Cardiovasculares , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Potenciais de Ação/efeitos dos fármacos , Adulto , Ajmalina/farmacologia , Algoritmos , Arritmias Cardíacas/genética , Linhagem Celular , Biologia Computacional , Avaliação Pré-Clínica de Medicamentos/estatística & dados numéricos , Canal de Potássio ERG1/genética , Eletrocardiografia , Sistema de Condução Cardíaco/fisiopatologia , Cardiopatias Congênitas/genética , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/fisiologia , Ivabradina/farmacologia , Mexiletina/farmacologia , Mutação , Quinidina/farmacologia , Pesquisa Translacional Biomédica
3.
Pharmacology ; 105(11-12): 662-668, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32460288

RESUMO

BACKGROUND: Adrenaline (Adr) and dexmedetomidine (Dex) are commonly used adjuvants of local anesthetics; however, the difference in the improvement of analgesia of local anesthetics between the 2 adjuvants remains unclear. OBJECTIVE: The objective of this experimental research was to evaluate the cutaneous analgesic effect of mexiletine (Mex) by coadministration with Dex or Adr. METHODS: The effect of a nociceptive block was assessed based on the inhibition of the cutaneous trunci muscle reflex in response to skin pinpricks in rats. The analgesic activity of Mex alone and Mex coadministered with Dex or Adr was evaluated after subcutaneous injections. Subcutaneous injections of drugs or combinations include Mex 0.6, 1.8, and 6.0 µmol; Adr 13.66 nmol; Dex 1.05600 nmol; saline; and Mex 1.8 and 6.0 µmol, respectively, combined with Dex 0.01056, 0.10560, and 1.05600 nmol or Adr 0.55, 2.73, and 13.66 nmol, with each injection dose of 0.6 mL. RESULTS: Subcutaneous injections of Mex elicited dose-related cutaneous analgesia. Compared with Mex (1.8 µmol), adding Dex or Adr to Mex (1.8 µmol) solutions for skin nociceptive block potentiated and prolonged the action. Mex (6.0 µmol) combined with Dex or Adr extended the duration of cutaneous analgesia when compared with Mex (6.0 µmol) alone. A high dose of Adr is more effective with Mex 1.8 µmol than that of Dex, whereas medium and low doses were less effective. Mex 6.0 µmol combined with any dose of Adr is superior to that of Dex. CONCLUSIONS: Both Dex and Adr improve the sensory block and enhance the nociceptive block duration of Mex. But in most cases, Adr is superior to Dex. It may be that different mechanisms of action of the 2 adjuvants lead to the differences.


Assuntos
Adjuvantes Farmacêuticos/farmacologia , Anestésicos Locais/farmacologia , Dexmedetomidina/farmacologia , Epinefrina/farmacologia , Mexiletina/farmacologia , Dor Nociceptiva/tratamento farmacológico , Adjuvantes Farmacêuticos/administração & dosagem , Analgesia/métodos , Anestesia Local/métodos , Anestésicos Locais/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Dexmedetomidina/administração & dosagem , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Epinefrina/administração & dosagem , Injeções Subcutâneas , Masculino , Mexiletina/administração & dosagem , Ratos , Ratos Sprague-Dawley , Reflexo/efeitos dos fármacos , Pele/efeitos dos fármacos
4.
J Membr Biol ; 250(5): 471-481, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28766006

RESUMO

Cardiac electrophysiology is regulated by the autonomic nervous system, and this has both pathophysiological, and possibly therapeutic importance. Furthermore, chamber differences in electrophysiology exist between atria and ventricles, yet there have been few direct comparisons. There is substantial literature on ion channel modulation at the single-cell level but less work on how this affects tissue-level parameters. We used a microelectrode array system to explore these issues using murine atrial and ventricular tissue slices. Activation time, conduction velocity and repolarisation were measured, and their modulation by temperature and pharmacological autonomic agonists were assessed. The system recorded reliable measurements under control conditions in the absence of drug/thermal challenge, and significant baseline differences were found in chamber electrophysiology. The sodium channel blocker mexiletine, produced large magnitude changes in all three measured parameters. Carbachol and isoprenaline induced differing effects in atria and ventricles, whereas temperature produced similar effects on activation and repolarisation.


Assuntos
Função Atrial/fisiologia , Técnicas Eletrofisiológicas Cardíacas , Fenômenos Eletrofisiológicos , Miocárdio , Função Ventricular/fisiologia , Animais , Função Atrial/efeitos dos fármacos , Carbacol/farmacologia , Feminino , Átrios do Coração , Ventrículos do Coração , Isoproterenol/farmacologia , Masculino , Mexiletina/farmacologia , Camundongos , Microeletrodos , Função Ventricular/efeitos dos fármacos
5.
Biophys Chem ; 229: 62-67, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28673509

RESUMO

The human Ether-a-go-go Related Gene (hERG) potassium channel plays a central role in the rapid component (IKr) of cardiac action potential repolarization phase. A large number of structurally different compounds block hERG and cause a high risk of arrhythmias. Among the drugs that block hERG channel, a few compounds have been identified as hERG channel activators. Such compounds may be useful, at least in theory, for the treatment of long term QT syndrome. Here we describe a new activator of hERG channel, named MC450. This compound is a symmetric urea, derived from (R)-mexiletine. Using patch-clamp recordings, we found that MC450 increased the activation current of hERG channel, with an EC50 of 41±4µM. Moreover MC450 caused a depolarizing shift in the voltage dependence of inactivation from -64.1±1.2mV (control), to -35.9±1.4mV, whereas it had no effect on the voltage dependence of activation. Furthermore, MC450 slowed current inactivation and the effect of MC450 was attenuated by the inactivation-impaired double mutant G628C/S631C.


Assuntos
Canal de Potássio ERG1/agonistas , Canal de Potássio ERG1/metabolismo , Mexiletina/análogos & derivados , Mexiletina/química , Ureia/análogos & derivados , Potenciais de Ação/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Canal de Potássio ERG1/genética , Células HEK293 , Humanos , Mexiletina/metabolismo , Mexiletina/farmacologia , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Estereoisomerismo , Ureia/química , Ureia/metabolismo , Ureia/farmacologia
6.
J Pharmacol Toxicol Methods ; 63(3): 258-68, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21194571

RESUMO

INTRODUCTION: Nonclinical in vivo models used for cardiovascular safety testing have not previously been studied for their sensitivity for detection of conduction slowing resulting from cardiac sodium channel block. The goal of this study was to examine the sensitivity of in vivo models to cardiac sodium channel block, and translation of the effect from in vitro to in vivo models using sodium channel inhibitors flecainide and mexiletine; flecainide, but not mexiletine is commonly associated with QRS complex prolongation in humans. METHODS: Inhibition of cloned cardiac sodium channels (hNav1.5) was studied using the IonWorks platform. Conduction slowing was measured in vitro in the rabbit isolated ventricular wedge (RVW) and in vivo in the conscious telemetered rat and dog, and anaesthetised dog. RESULTS: Flecainide and mexiletine inhibited hNav1.5 channels with IC50 values of 10.7 and 67.2 µM respectively. In the RVW, QRS was increased by flecainide at 60 bpm, and at 120bpm, there was an increased effect of both drugs. In conscious rats, flecainide significantly increased QRS complex duration; mexiletine had no significant effect, but there was an increase at the highest dose in 4/6 animals. QRS complex was increased by flecainide and mexiletine in anaesthetised dogs but this was not statistically significant; in conscious dog, only flecainide produced a significant increase in QRS complex. DISCUSSION: When compared to clinical data, effects of flecainide and mexiletine in RVW and conscious dog compared well with effects in patients and healthy volunteers in terms of sensitivity. The anaesthetised dog was least sensitive for detection of changes in QRS. All assays showed some differentiation between the expected conduction slowing activity of flecainide and mexiletine. Based on these data, RVW and conscious dog were most predictive for effects of compounds on QRS complex and cardiac conduction.


Assuntos
Flecainida/farmacologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Mexiletina/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Linhagem Celular , Ensaios Clínicos como Assunto , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Eletrocardiografia , Feminino , Flecainida/sangue , Frequência Cardíaca/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Humanos , Masculino , Mexiletina/sangue , Canal de Sódio Disparado por Voltagem NAV1.5 , Ligação Proteica , Coelhos , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/sangue , Canais de Sódio/genética , Transfecção
7.
Neurochem Res ; 34(10): 1816-23, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19504185

RESUMO

Physical or chemical damage to peripheral nerves can result in neuropathic pain which is not easily alleviated by conventional analgesic drugs. Substantial evidence has demonstrated that voltage-gated Na+ channels in the membrane of damaged nerves play a key role in the establishment and maintenance of pathological neuronal excitability not only of these peripheral nerves but also in the second- and third-order neurons in the pain pathway to the cerebral cortex. Na+ channel blocking drugs have been used in treating neuropathic pain with limited success mainly because of a preponderance of side-effects. We have developed an analogue of mexiletine which is approximately 80 times more potent than mexiletine in competing with the radioligand, 3H-batrachotoxinin for binding to Na+ channels in rat brain membranes and also it is much more lipophilic than mexiletine which should enhance its uptake into the brain to block the increased expression of Na+ channels on second- and third-order neurons of the pain pathway. This analogue, HFI-1, has been tested in three different rat models of neuropathic pain (formalin paw model, ligated spinal nerve model and contusive spinal cord injury model) and found to be more effective in reducing pain behaviours than mexiletine.


Assuntos
Modelos Animais de Doenças , Mexiletina/análogos & derivados , Mexiletina/uso terapêutico , Neuralgia/tratamento farmacológico , Bloqueadores dos Canais de Sódio/uso terapêutico , Canais de Sódio/metabolismo , Analgésicos/química , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Masculino , Mexiletina/farmacologia , Neuralgia/fisiopatologia , Medição da Dor/métodos , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/fisiologia
8.
Anticancer Res ; 23(1A): 231-4, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12680218

RESUMO

BACKGROUND: Within the last decade, there has been much interest in the area of tumor angiogenesis, with the advent of many new anti-angiogenic drugs undergoing testing in cancer clinical Phases II and III. Many of the cancer patients also take multiple medications for a variety of chronic illnesses. Because of possible drug-drug interactions, it is important to investigate the effect that commonly prescribed medications may have on angiogenesis. MATERIALS AND METHODS: In this pilot study, we assessed the effect of the following drugs on in vitro angiogenesis: atenolol, diltiazem, enalapril, disopyramide, mexiletine, coumadin, cimetidine and omeprazole. RESULTS & CONCLUSION: We observed that, although some of these drugs at massive doses inhibited endothelial proliferation, they did not affect in vitro angiogenesis at human therapeutic ranges.


Assuntos
Inibidores da Angiogênese/farmacologia , Endotélio Vascular/efeitos dos fármacos , Atenolol/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Cimetidina/farmacologia , Diltiazem/farmacologia , Disopiramida/farmacologia , Avaliação Pré-Clínica de Medicamentos , Enalapril/farmacologia , Endotélio Vascular/citologia , Humanos , Mexiletina/farmacologia , Omeprazol/farmacologia , Projetos Piloto , Varfarina/farmacologia
10.
Proc Natl Acad Sci U S A ; 94(6): 2724-8, 1997 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-9122264

RESUMO

Previous studies have shown that chronic administration of class I antiarrhythmic drugs, which have definite inhibitory action on the fast Na+ channel, result in up-regulation of cardiac Na+ channel expression, and suggest that this effect may contribute to their deleterious effects during chronic administration. Recent studies have shown that the antiarrhythmic effects of free n - 3 polyunsaturated fatty acids (PUFA) are associated with an inhibition of the Na+ channel. Whether the PUFA when used chronically will mimic the effect of the class I drugs on the expression of the Na+ channel is not known. To answer this question, we determined the level of mRNA encoding cardiac Na+ channels and the number of the Na+ channels per cell in cultured neonatal rat cardiac myocytes after supplementation of the cells with the n - 3 PUFA eicosapentaenoic acid (EPA), the class I drug mexiletine, or both EPA and mexiletine for 3-4 days. The number of sodium channels was assessed with a radioligand binding assay using the sodium channel-specific toxin [3H]batrachotoxinin benzoate ([3H]BTXB). The supplementation of myocytes with mexiletine (20 microM) induced a 4-fold increase in [3H]BTXB specific binding to the cells. In contrast, chronic treatment with EPA (20 microM) alone did not significantly affect [3H]BTXB binding. However, the combination of EPA with mexiletine produced a 40-50% reduction in the [3H]BTXB binding, compared with that seen with mexiletine alone. RNA isolated from cardiac myocytes was probed with a 2.5-kb cRNA transcribed with T7 RNA polymerase from the clone Na-8.4, which encodes nucleotides 3361-5868 of the alpha-subunit of the R(IIA) sodium channel subtype. The changes in the level of mRNA encoding sodium channel alpha-subunit were correlated with comparable changes in sodium channel number in the cultured myocytes, indicating that regulation of transcription of mRNA or its processing and stability is primarily responsible for the regulation of sodium channel number. These data demonstrate that chronic EPA treatment not only does not up-regulate the cardiac sodium channel expression but also reduces the mexiletine-induced increase in the cardiac sodium channel expression.


Assuntos
Antiarrítmicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Coração/efeitos dos fármacos , Mexiletina/farmacologia , Miocárdio/metabolismo , Canais de Sódio/biossíntese , Transcrição Gênica/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Batraquiotoxinas/metabolismo , Células Cultivadas , Miocárdio/citologia , RNA Mensageiro/biossíntese , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Trítio
11.
Clin Invest Med ; 14(5): 476-83, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1660369

RESUMO

This article reviews the data which support the use of selected drug combinations to enhance anti-arrhythmic activity. Specifically, we have focused on the mexiletine-quinidine interaction and the relation between anti-arrhythmic efficacy and electrophysiologic effects. In an initial clinical study, we found that combination therapy with mexiletine-quinidine produced enhanced efficacy in suppressing spontaneous ventricular tachycardia with fewer side-effects than high dose monotherapy. This enhanced efficacy has been confirmed in other laboratories. Combination therapy also enhanced suppression of inducible ventricular tachycardia in patients and in animal models. Animal models were used to assess the relation between electrophysiologic effects and anti-arrhythmic efficacy. In the animal studies, combination therapy produced selective prolongation of refractoriness and conduction in the infarct and peri-infarct zones without significant changes in the normal zone. Subsequent studies focused on the relative contribution of sodium channel and potassium channel blocking properties of these drugs to the enhanced activity seen with the combination. Studies using the selective sodium channel blocker tetrodotoxin confirmed that sodium channel blockade was necessary for this interaction. To assess the contribution of prolongation of action potential duration by quinidine to the combined effect we compared the anti-arrhythmic and electrophysiologic effects of the stereoisomers quinidine and quinine given alone and in combination with mexiletine. These experimental data confirm that the property of prolongation of action potential duration by quinidine is essential to the interaction. When comparing quinidine and quinine it is apparent that prolongation of refractoriness in the peri-infarct zone is essential for anti-arrhythmic activity.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Arritmias Cardíacas/tratamento farmacológico , Mexiletina/uso terapêutico , Quinidina/uso terapêutico , Animais , Arritmias Cardíacas/fisiopatologia , Quimioterapia Combinada , Eletrofisiologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Humanos , Mexiletina/administração & dosagem , Mexiletina/farmacologia , Quinidina/administração & dosagem , Quinidina/farmacologia , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/fisiologia
12.
Jpn Heart J ; 28(5): 719-35, 1987 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3430732

RESUMO

We studied the effects of Ca channel blockers and 3 antiarrhythmic drugs on the digitalis-induced oscillatory afterpotential (OAP). The OAP was observed in Purkinje fibers stimulated by pulse trains, with cycle lengths ranging from 1,000 to 300 msec. The Ca channel blockers verapamil, diltiazem and nifedipine (2.0 x 10(-6) M) depressed OAP significantly and abolished triggered activity. Verapamil was more effective than diltiazem. However, nicardipine and nitrendipine (2.0 x 10(-6) M) had no depressant effects on OAP or triggered activity. The antiarrhythmic drugs procainamide (1.0 x 10(-4) M), mexiletine (1.0 x 10(-5) M) and propranolol (1.0 x 10(-4) M) depressed both OAP and triggered activity. There were no significant differences in the depressant effects between the Ca2+ antagonists (except for nitrendipine and nicardipine) and the other antiarrhythmic drugs. The OAP coupling interval was prolonged by verapamil, diltiazem, propranolol, procainamide and mexiletine. Although the APD50 was shortened by verapamil, diltiazem and nifedipine, it was prolonged by propranolol. It is concluded that nifedipine, verapamil, diltiazem, procainamide, mexiletine and propranolol may be effective for digitalis-related arrhythmia.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/fisiopatologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sistema de Condução Cardíaco/fisiopatologia , Ramos Subendocárdicos/fisiopatologia , Animais , Arritmias Cardíacas/tratamento farmacológico , Digitalis , Diltiazem/farmacologia , Cães , Estimulação Elétrica , Feminino , Masculino , Mexiletina/farmacologia , Nicardipino/farmacologia , Nifedipino/farmacologia , Nitrendipino/farmacologia , Plantas Medicinais , Plantas Tóxicas , Procainamida/farmacologia , Propranolol/farmacologia , Ramos Subendocárdicos/efeitos dos fármacos , Verapamil/farmacologia
13.
J Pharmacol Exp Ther ; 241(1): 282-8, 1987 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3572790

RESUMO

The purpose of these experiments was to evaluate the relationship between interval dependent effects of antiarrhythmic drugs on conduction time and Vmax in canine cardiac Purkinje fibers. Standard microelectrode techniques were used to monitor action potential characteristics at two sites along a canine cardiac false tendon and to measure interelectrode conduction time. The maximum rate of voltage rise during phase 0 (Vmax) and conduction time were independent of diastolic interval under control conditions. In the presence of local anesthetic drugs, recovery from drug-induced depression of Vmax and conduction were first order processes with recovery time constants (mean +/- S.D. in seconds) of 0.14 +/- 0.02 (for Vmax) and 0.15 +/- 0.04 (for conduction time) for lidocaine; 0.17 +/- 0.04 and 0.18 +/- 0.05, respectively, for mexiletine; 0.26 +/- 0.05 and 0.27 +/- 0.07 for amitriptyline; and 1.01 +/- 0.31 and 1.00 +/- 0.32 for procainamide. The kinetics of onset of block were studied using a 30-sec pause, followed by a pacing cycle length of 300 msec (for procainamide) or 1 sec (for quinidine). The onset time constants averaged 2.66 +/- 0.53 pulses (for Vmax) and 2.49 +/- 0.42 pulses (for conduction time) in the presence of procainamide; and 4.02 +/- 1.33 pulses (for Vmax) and 3.86 +/- 1.22 pulses (for conduction time) in the presence of quinidine. These experiments show that local anesthetic drugs produce use dependent changes in conduction time in vitro with time constants comparable to simultaneously measured time constants for effects on Vmax. They imply that the use dependence of drug effects on cardiac conduction can be studied quantitatively in vivo by studying the response to changes in activation frequency.


Assuntos
Antiarrítmicos/farmacologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Ramos Subendocárdicos/efeitos dos fármacos , Potenciais de Ação , Amitriptilina/farmacologia , Anestesia Local , Animais , Cães , Relação Dose-Resposta a Droga , Condutividade Elétrica , Eletrofisiologia , Cinética , Lidocaína/farmacologia , Mexiletina/farmacologia , Microeletrodos , Procainamida/farmacologia , Ramos Subendocárdicos/fisiologia
14.
J Cardiovasc Pharmacol ; 6(2): 213-9, 1984.
Artigo em Inglês | MEDLINE | ID: mdl-6200706

RESUMO

Antiarrhythmic effects of mexiletine were examined using three canine ventricular arrhythmia models--digitalis-, adrenaline-, and two-stage coronary ligation-induced arrhythmias. The minimum effective plasma concentration for each arrhythmia model was determined. Mexiletine suppressed all the arrhythmias, and the minimum effective plasma concentrations for arrhythmias induced by digitalis, adrenaline, 24-h coronary ligation, and 48-h coronary ligation were 1.8 +/- 0.6, 3.7 +/- 0.9, 1.9 +/- 0.3, and 2.2 +/- 0.4 micrograms/ml, respectively (mean +/- SD, n = 6-7). The concentration for adrenaline-induced arrhythmias was significantly higher than those for the other types of arrhythmias. Mexiletine had a hypotensive effect when it was given intravenously, but this effect was not observed when it was given orally. Oral mexiletine was also effective in suppressing the 24-h coronary ligation-induced arrhythmia. Mexiletine induced convulsions when higher doses were given. The correlations between the mexiletine plasma concentrations and its antiarrhythmic effects were not very strong, probably indicating individual variations in sensitivity to mexiletine.


Assuntos
Arritmias Cardíacas/tratamento farmacológico , Mexiletina/uso terapêutico , Propilaminas/uso terapêutico , Animais , Doença das Coronárias/complicações , Digitalis , Cães , Relação Dose-Resposta a Droga , Epinefrina , Feminino , Canais Iônicos/efeitos dos fármacos , Masculino , Mexiletina/sangue , Mexiletina/farmacologia , Plantas Medicinais , Plantas Tóxicas
15.
Arch Sci Med (Torino) ; 136(1): 1-15, 1979.
Artigo em Italiano | MEDLINE | ID: mdl-464786

RESUMO

It was shown experimentally that mexilithene possesses anti-arrhythmic activity with slight depression of sino-carotid baroreceptorial and glomo-carotid and gangliar chemoreceptorial activity and catecholamine uptake. Hypotensive and bradycardizing effects were only noted when high per kg doses were used. The drug did not display vascular alpha- and beta-adrenolytic, anti-muscarinic and anti-histaminic activity.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Mexiletina/farmacologia , Propilaminas/farmacologia , Animais , Arritmias Cardíacas/tratamento farmacológico , Pressão Sanguínea/efeitos dos fármacos , Corpo Carotídeo/efeitos dos fármacos , Seio Carotídeo/efeitos dos fármacos , Cães , Avaliação Pré-Clínica de Medicamentos , Frequência Cardíaca/efeitos dos fármacos , Lidocaína/farmacologia , Mexiletina/administração & dosagem , Mexiletina/uso terapêutico , Pressorreceptores/efeitos dos fármacos , Coelhos , Ratos , Respiração/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA