Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 413
Filter
1.
J Physiol ; 601(18): 4013-4032, 2023 09.
Article in English | MEDLINE | ID: mdl-37475475

ABSTRACT

The best pharmacological treatment for each atrial fibrillation (AF) patient is unclear. We aim to exploit AF simulations in 800 virtual atria to identify key patient characteristics that guide the optimal selection of anti-arrhythmic drugs. The virtual cohort considered variability in electrophysiology and low voltage areas (LVA) and was developed and validated against experimental and clinical data from ionic currents to ECG. AF sustained in 494 (62%) atria, with large inward rectifier K+ current (IK1 ) and Na+ /K+ pump (INaK ) densities (IK1 0.11 ± 0.03 vs. 0.07 ± 0.03 S mF-1 ; INaK 0.68 ± 0.15 vs. 0.38 ± 26 S mF-1 ; sustained vs. un-sustained AF). In severely remodelled left atrium, with LVA extensions of more than 40% in the posterior wall, higher IK1 (median density 0.12 ± 0.02 S mF-1 ) was required for AF maintenance, and rotors localized in healthy right atrium. For lower LVA extensions, rotors could also anchor to LVA, in atria presenting short refractoriness (median L-type Ca2+ current, ICaL , density 0.08 ± 0.03 S mF-1 ). This atrial refractoriness, modulated by ICaL and fast Na+ current (INa ), determined pharmacological treatment success for both small and large LVA. Vernakalant was effective in atria presenting long refractoriness (median ICaL density 0.13 ± 0.05 S mF-1 ). For short refractoriness, atria with high INa (median density 8.92 ± 2.59 S mF-1 ) responded more favourably to amiodarone than flecainide, and the opposite was found in atria with low INa (median density 5.33 ± 1.41 S mF-1 ). In silico drug trials in 800 human atria identify inward currents as critical for optimal stratification of AF patient to pharmacological treatment and, together with the left atrial LVA extension, for accurately phenotyping AF dynamics. KEY POINTS: Atrial fibrillation (AF) maintenance is facilitated by small L-type Ca2+ current (ICaL ) and large inward rectifier K+ current (IK1 ) and Na+ /K+ pump. In severely remodelled left atrium, with low voltage areas (LVA) covering more than 40% of the posterior wall, sustained AF requires higher IK1 and rotors localize in healthy right atrium. For lower LVA extensions, rotors can also anchor to LVA, if the atria present short refractoriness (low ICaL ) Vernakalant is effective in atria presenting long refractoriness (high ICaL ). For short refractoriness, atria with fast Na+ current (INa ) up-regulation respond more favourably to amiodarone than flecainide, and the opposite is found in atria with low INa . The inward currents (ICaL and INa ) are critical for optimal stratification of AF patient to pharmacological treatment and, together with the left atrial LVA extension, for accurately phenotyping AF dynamics.


Subject(s)
Amiodarone , Atrial Fibrillation , Humans , Atrial Fibrillation/drug therapy , Flecainide/pharmacology , Flecainide/therapeutic use , Heart Atria , Amiodarone/pharmacology , Amiodarone/therapeutic use , Action Potentials/physiology
2.
Circ Res ; 128(3): 321-331, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33297863

ABSTRACT

RATIONALE: The class Ic antiarrhythmic drug flecainide prevents ventricular tachyarrhythmia in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT), a disease caused by hyperactive RyR2 (cardiac ryanodine receptor) mediated calcium (Ca) release. Although flecainide inhibits single RyR2 channels in vitro, reports have claimed that RyR2 inhibition by flecainide is not relevant for its mechanism of antiarrhythmic action and concluded that sodium channel block alone is responsible for flecainide's efficacy in CPVT. OBJECTIVE: To determine whether RyR2 block independently contributes to flecainide's efficacy for suppressing spontaneous sarcoplasmic reticulum Ca release and for preventing ventricular tachycardia in vivo. METHODS AND RESULTS: We synthesized N-methylated flecainide analogues (QX-flecainide and N-methyl flecainide) and showed that N-methylation reduces flecainide's inhibitory potency on RyR2 channels incorporated into artificial lipid bilayers. N-methylation did not alter flecainide's inhibitory activity on human cardiac sodium channels expressed in HEK293T cells. Antiarrhythmic efficacy was tested utilizing a Casq2 (cardiac calsequestrin) knockout (Casq2-/-) CPVT mouse model. In membrane-permeabilized Casq2-/- cardiomyocytes-lacking intact sarcolemma and devoid of sodium channel contribution-flecainide, but not its analogues, suppressed RyR2-mediated Ca release at clinically relevant concentrations. In voltage-clamped, intact Casq2-/- cardiomyocytes pretreated with tetrodotoxin to inhibit sodium channels and isolate the effect of flecainide on RyR2, flecainide significantly reduced the frequency of spontaneous sarcoplasmic reticulum Ca release, while QX-flecainide and N-methyl flecainide did not. In vivo, flecainide effectively suppressed catecholamine-induced ventricular tachyarrhythmias in Casq2-/- mice, whereas N-methyl flecainide had no significant effect on arrhythmia burden, despite comparable sodium channel block. CONCLUSIONS: Flecainide remains an effective inhibitor of RyR2-mediated arrhythmogenic Ca release even when cardiac sodium channels are blocked. In mice with CPVT, sodium channel block alone did not prevent ventricular tachycardia. Hence, RyR2 channel inhibition likely constitutes the principal mechanism of antiarrhythmic action of flecainide in CPVT.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Flecainide/pharmacology , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/drug effects , Tachycardia, Ventricular/prevention & control , Action Potentials , Animals , Calcium Signaling , Calsequestrin/genetics , Calsequestrin/metabolism , Disease Models, Animal , Female , HEK293 Cells , Humans , Male , Mice, Knockout , Myocytes, Cardiac/metabolism , Phosphorylation , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Sheep, Domestic , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Voltage-Gated Sodium Channel Blockers/pharmacology
3.
Europace ; 25(3): 1152-1161, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36504385

ABSTRACT

AIMS: Atrial fibrillation (AF) is the most common cardiac arrhythmia. Pathogenic variants in genes encoding ion channels are associated with familial AF. The point mutation M1875T in the SCN5A gene, which encodes the α-subunit of the cardiac sodium channel Nav1.5, has been associated with increased atrial excitability and familial AF in patients. METHODS AND RESULTS: We designed a new murine model carrying the Scn5a-M1875T mutation enabling us to study the effects of the Nav1.5 mutation in detail in vivo and in vitro using patch clamp and microelectrode recording of atrial cardiomyocytes, optical mapping, electrocardiogram, echocardiography, gravimetry, histology, and biochemistry. Atrial cardiomyocytes from newly generated adult Scn5a-M1875T+/- mice showed a selective increase in the early (peak) cardiac sodium current, larger action potential amplitude, and a faster peak upstroke velocity. Conduction slowing caused by the sodium channel blocker flecainide was less pronounced in Scn5a-M1875T+/- compared to wildtype atria. Overt hypertrophy or heart failure in Scn5a-M1875T+/- mice could be excluded. CONCLUSION: The Scn5a-M1875T point mutation causes gain-of-function of the cardiac sodium channel. Our results suggest increased atrial peak sodium current as a potential trigger for increased atrial excitability.


Subject(s)
Atrial Fibrillation , Animals , Mice , Atrial Fibrillation/drug therapy , Atrial Fibrillation/genetics , Flecainide/pharmacology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Mutation , Heart Atria
4.
J Mol Cell Cardiol ; 166: 23-35, 2022 05.
Article in English | MEDLINE | ID: mdl-35114252

ABSTRACT

Atrial fibrillation (AF) affects over 1% of the population and is a leading cause of stroke and heart failure in the elderly. A feared side effect of sodium channel blocker therapy, ventricular pro-arrhythmia, appears to be relatively rare in patients with AF. The biophysical reasons for this relative safety of sodium blockers are not known. Our data demonstrates intrinsic differences between atrial and ventricular cardiac voltage-gated sodium currents (INa), leading to reduced maximum upstroke velocity of action potential and slower conduction, in left atria compared to ventricle. Reduced atrial INa is only detected at physiological membrane potentials and is driven by alterations in sodium channel biophysical properties and not by NaV1.5 protein expression. Flecainide displayed greater inhibition of atrial INa, greater reduction of maximum upstroke velocity of action potential, and slowed conduction in atrial cells and tissue. Our work highlights differences in biophysical properties of sodium channels in left atria and ventricles and their response to flecainide. These differences can explain the relative safety of sodium channel blocker therapy in patients with atrial fibrillation.


Subject(s)
Atrial Fibrillation , Flecainide , Action Potentials , Aged , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/metabolism , Flecainide/metabolism , Flecainide/pharmacology , Flecainide/therapeutic use , Heart Atria/metabolism , Humans , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism
5.
J Cell Physiol ; 237(10): 3900-3911, 2022 10.
Article in English | MEDLINE | ID: mdl-35959596

ABSTRACT

Tachycardia is characterized by high beating rates that can lead to life-threatening fibrillations. Mutations in several ion-channel genes were implicated with tachycardia; however, the complex genetic contributors and their modes of action are still unclear. Here, we investigated the influence of an SCN5A gene variant on tachycardia phenotype by deriving patient-specific iPSCs and cardiomyocytes (iPSC-CM). Two tachycardia patients were genetically analyzed and revealed to inherit a heterozygous p.F1465L variant in the SCN5A gene. Gene expression and immunocytochemical analysis in iPSC-CMs generated from patients did not show any significant changes in mRNA levels of SCN5A or gross NaV1.5 cellular mislocalization, compared to healthy-derived iPSC-CMs. Electrophysiological and contraction imaging analysis in patient iPSC-CMs revealed intermittent fibrillation-like states, occasional arrhythmic events, and sustained high-paced contractions that could be selectively reduced by flecainide treatment. The patch-clamp analysis demonstrated a negative shift in the voltage-dependent activation at the patient-derived iPSC-CMs compared to the healthy control line, suggestive of a gain-of-function activity associated with the SCN5A+/p.F1465L variant. Our patient-derived iPSC-CM model recapitulated the clinically relevant characteristics of tachycardia associated with a novel pathogenic SCN5A+/p.F1465L variant leading to altered Na+ channel kinetics as the likely mechanism underlying high excitability and tachycardia phenotype.


Subject(s)
Induced Pluripotent Stem Cells , Arrhythmias, Cardiac , Flecainide/metabolism , Flecainide/pharmacology , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel , RNA, Messenger/metabolism , Tachycardia/metabolism , Tachycardia/pathology
6.
Europace ; 24(12): 2015-2027, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-35726875

ABSTRACT

AIMS: Variants in SCN5A encoding Nav1.5 are associated with cardiac arrhythmias. We aimed to determine the mechanism by which c.638G>A in SCNA5 resulting in p.Gly213Asp (G213D) in Nav1.5 altered Na+ channel function and how flecainide corrected the defect in a family with multifocal ectopic Purkinje-related premature contractions (MEPPC)-like syndrome. METHODS AND RESULTS: Five patients carrying the G213D variant were treated with flecainide. Gating pore currents were evaluated in Xenopus laevis oocytes. The 638G>A SCN5A variant was introduced to human-induced pluripotent stem cell (hiPSC) by CRISPR-Cas9 gene editing and subsequently differentiated to cardiomyocytes (hiPSC-CM). Action potentials and sodium currents were measured in the absence and presence of flecainide. Ca2+ transients were measured by confocal microscopy. The five patients exhibited premature atrial and ventricular contractions which were suppressed by flecainide treatment. G213D induced gating pore current at potentials negative to -50 mV. Voltage-clamp analysis in hiPSC-CM revealed the activation threshold of INa was shifted in the hyperpolarizing direction resulting in a larger INa window current. The G213D hiPSC-CMs had faster beating rates compared with wild-type and frequently showed Ca2+ waves and alternans. Flecainide applied to G213D hiPSC-CMs decreased window current by shifting the steady-state inactivation curve and slowed the beating rate. CONCLUSION: The G213D variant in Nav1.5 induced gating pore currents and increased window current. The changes in INa resulted in a faster beating rate and Ca2+ transient dysfunction. Flecainide decreased window current and inhibited INa, which is likely responsible for the therapeutic effectiveness of flecainide in MEPPC patients carrying the G213D variant.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , NAV1.5 Voltage-Gated Sodium Channel , Humans , Action Potentials/physiology , Arrhythmias, Cardiac/genetics , Flecainide/pharmacology , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Phenotype , Sodium/metabolism
7.
PLoS Comput Biol ; 16(2): e1007678, 2020 02.
Article in English | MEDLINE | ID: mdl-32097431

ABSTRACT

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and is a major cause of stroke and morbidity. Recent genome-wide association studies have shown that paired-like homeodomain transcription factor 2 (Pitx2) to be strongly associated with AF. However, the mechanisms underlying Pitx2 modulated arrhythmogenesis and variable effectiveness of antiarrhythmic drugs (AADs) in patients in the presence or absence of impaired Pitx2 expression remain unclear. We have developed multi-scale computer models, ranging from a single cell to tissue level, to mimic control and Pitx2-knockout atria by incorporating recent experimental data on Pitx2-induced electrical and structural remodeling in humans, as well as the effects of AADs. The key findings of this study are twofold. We have demonstrated that shortened action potential duration, slow conduction and triggered activity occur due to electrical and structural remodelling under Pitx2 deficiency conditions. Notably, the elevated function of calcium transport ATPase increases sarcoplasmic reticulum Ca2+ concentration, thereby enhancing susceptibility to triggered activity. Furthermore, heterogeneity is further elevated due to Pitx2 deficiency: 1) Electrical heterogeneity between left and right atria increases; and 2) Increased fibrosis and decreased cell-cell coupling due to structural remodelling slow electrical propagation and provide obstacles to attract re-entry, facilitating the initiation of re-entrant circuits. Secondly, our study suggests that flecainide has antiarrhythmic effects on AF due to impaired Pitx2 by preventing spontaneous calcium release and increasing wavelength. Furthermore, our study suggests that Na+ channel effects alone are insufficient to explain the efficacy of flecainide. Our study may provide the mechanisms underlying Pitx2-induced AF and possible explanation behind the AAD effects of flecainide in patients with Pitx2 deficiency.


Subject(s)
Atrial Fibrillation/metabolism , Computer Simulation , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Action Potentials , Animals , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/genetics , Atrial Remodeling , Calcium/metabolism , Electrophysiology , Endoplasmic Reticulum/metabolism , Fibrosis , Flecainide/pharmacology , Gene Expression Regulation , Genome-Wide Association Study , Heart Atria/physiopathology , Homeodomain Proteins/genetics , Humans , Kinetics , Mice , Mice, Knockout , Phenotype , Ryanodine Receptor Calcium Release Channel/pharmacology , Sarcoplasmic Reticulum/metabolism , Sodium/metabolism , Transcription Factors/genetics , Homeobox Protein PITX2
8.
Pacing Clin Electrophysiol ; 43(4): 360-363, 2020 04.
Article in English | MEDLINE | ID: mdl-32010978

ABSTRACT

A 64-year-old man underwent implantation of a permanent His-bundle pacemaker. A marked rise in the selective His-bundle capture threshold was noted 1 month after the patient started flecainide acetate for rhythm control of recurrent, symptomatic atrial flutter and atrial fibrillation. The capture threshold subsequently normalized 4 days after discontinuing flecainide and switching to dofetilide. To our knowledge, this is the first documented case of a rise in selective His-bundle capture threshold associated with flecainide acetate. Further studies are needed to characterize this association which could result in higher capture thresholds, decreased battery longevity, and mimic His-bundle lead failure.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Bundle of His/drug effects , Bundle of His/physiopathology , Cardiac Pacing, Artificial , Flecainide/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Flecainide/therapeutic use , Humans , Male , Middle Aged
9.
Pharmacol Res ; 148: 104443, 2019 10.
Article in English | MEDLINE | ID: mdl-31493514

ABSTRACT

Over the last 35 years, flecainide proved itself one of the most commonly used arrhythmic drugs, expanding its original indication for ventricular arrhythmias and results nowadays as the cornerstone of the rhythm control strategy in atrial fibrillation management of patients without structural heart disease. While the increased mortality associated with flecainide in the Cardiac Arrhythmia Suppression Trial (CAST) still casts his shadow over flecainide clinical profile, this compound has subsequently demonstrated safe and is now used successfully for a plethora of indications, including pharmacological cardioversion of atrial fibrillation, cathecolaminergic polymorphic ventricular tachycardia, supraventricular tachyarrhythmias and ventricular pre-excitation. Moreover, the recent marketing of a controlled release formulation, along with the intravenous and immediate release formulations, increased the armamentarium to the clinician's disposal while improving patients' compliance. In the present paper, we offer a comprehensive review of the anti-arrhythmic effects of flecainide, detailing its electrophysiological properties, its effects on the conduction system, its clinical use and the major side effects and contraindications in clinical practice.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Electrocardiography/drug effects , Flecainide/pharmacology , Flecainide/therapeutic use , Animals , Humans
10.
BMC Cardiovasc Disord ; 19(1): 228, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31638896

ABSTRACT

BACKGROUND: Large animal models are important in atrial fibrillation (AF) research, as they can be used to study the pathophysiology of AF and new therapeutic approaches. Unlike other animal models, horses spontaneously develop AF and could therefore serve as a bona fide model in AF research. We therefore aimed to study the electrical, functional and structural remodelling caused by chronic AF in a horse model. METHOD: Nine female horses were included in the study, with six horses tachypaced into self-sustained AF and three that served as a time-matched sham-operated control group. Acceleration in atrial fibrillatory rate (AFR), changes in electrocardiographic and echocardiographic variables and response to medical treatment (flecainide 2 mg/kg) were recorded over a period of 2 months. At the end of the study, changes in ion channel expression and fibrosis were measured and compared between the two groups. RESULTS: AFR increased from 299 ± 33 fibrillations per minute (fpm) to 376 ± 12 fpm (p < 0.05) and atrial function (active left atrial fractional area change) decreased significantly during the study (p < 0.05). No changes were observed in heart rate or ventricular function. The AF group had more atrial fibrosis compared to the control group (p < 0.05). No differences in ion channel expression were observed. CONCLUSION: Horses with induced AF show signs of atrial remodelling that are similar to humans and other animal models.


Subject(s)
Action Potentials , Atrial Fibrillation/physiopathology , Atrial Function, Left , Atrial Remodeling , Heart Atria/physiopathology , Heart Rate , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Cardiac Pacing, Artificial , Disease Models, Animal , Female , Fibrosis , Flecainide/pharmacology , Heart Atria/drug effects , Heart Atria/metabolism , Heart Rate/drug effects , Horses , Ion Channels/metabolism , Time Factors
11.
Biol Pharm Bull ; 42(7): 1226-1229, 2019.
Article in English | MEDLINE | ID: mdl-31257298

ABSTRACT

Flecainide, an anti-arrhythmic drug, undergoes renal excretion through active renal tubular secretion in addition to passive glomerular filtration. The contribution of renal uptake and efflux transporters in active renal tubular secretion of flecainide remains unclear except that flecainide is a substrate of human multidrug resistance protein 1 (MDR1). To elucidate renal efflux and uptake transporters involved with active renal tubular secretion of flecainide, we conducted in vitro interaction studies of flecainide using organic cation transporter 2 (OCT2), multidrug and toxin extrusion (MATE) 1, and MATE2-K. Uptake transporter inhibition assays using hOCT2-Chinese hamster ovary (CHO), hMATE1-CHO, and hMATE2-K-Madin Darby canine kidney strain II (MDCKII) cells revealed that flecainide (2.5 µM) inhibited hMATE1-mediated transport by 40% with an IC50 value of 6.7 µM; however, it showed no or weak inhibitory effects on hOCT2- and hMATE2-K-mediated transport. For investigating flecainide as a substrate of hMATE1, the accumulation of flecainide in hMATE1-CHO was compared with that in control cells. Uptake transporter substrate assay revealed that flecainide (1 µM) showed 1.11-fold accumulation though the hMATE1-related active transport was significantly decreased in the presence of quinidine (42.0 ± 23.9 vs. 11.8 ± 4.1 pmol/mg in transfected cells; p < 0.05). These results suggest that flecainide is a weak substrate of hMATE1, which is involved in the renal tubular secretion of cationic drugs, and hMATE1 may be less important in the pharmacokinetic drug-drug interaction for renal excretion of flecainide. However, in vivo drug-drug interaction studies of flecainide with substrates of hMATE1 may be needed because flecainide has the potential to inhibit hMATE1.


Subject(s)
Flecainide/pharmacology , Kidney/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , Renal Elimination , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , CHO Cells , Cricetulus , Dogs , Drug Interactions , Madin Darby Canine Kidney Cells , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2/genetics
12.
Am J Physiol Heart Circ Physiol ; 315(4): H1051-H1062, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30028197

ABSTRACT

Sudden cardiac death from ventricular arrhythmias is more common in adult patients with with heart failure compared with pediatric patients with heart failure. We identified age-specific differences in arrhythmogenesis using a guinea pig model of acute ß-adrenergic stimulation. Young and adult guinea pigs were exposed to the ß-adrenergic agonist isoproterenol (ISO; 0.7 mg/kg) for 30 min in the absence or presence of flecainide (20 mg/kg), an antiarrhythmic that blocks Na+ and ryanodine channels. Implanted cardiac monitors (Reveal LINQ, Medtronic) were used to monitor heart rhythm. Alterations in phosphorylation and oxidation of ryanodine receptor 2 (RyR2) were measured in left ventricular tissue. There were age-specific differences in arrhythmogenesis and sudden death associated with acute ß-adrenergic stimulation in guinea pigs. Young and adult guinea pigs developed arrhythmias in response to ISO; however, adult animals developed significantly more premature ventricular contractions and experienced higher arrhythmia-related mortality than young guinea pigs treated with ISO. Although there were no significant differences in the phosphorylation of left ventricular RyR2 between young and adult guinea pigs, adult guinea pigs exposed to acute ISO had significantly more oxidation of RyR2. Flecainide treatment significantly improved survival and decreased the number of premature ventricular contractions in young and adult animals in association with lower RyR2 oxidation. Adult guinea pigs had a greater propensity to develop arrhythmias and suffer sudden death than young guinea pigs when acutely exposed to ISO. This was associated with higher oxidation of RyR2. The incidence of sudden death can be rescued with flecainide treatment, which decreases RyR2 oxidation. NEW & NOTEWORTHY Clinically, adult patients with heart failure are more likely to develop arrhythmias and sudden death than pediatric patients with heart failure. In the present study, older guinea pigs also showed a greater propensity to arrhythmias and sudden death than young guinea pigs when acutely exposed to isoproterenol. Although there are well-described age-related cardiac structural changes that predispose patients to arrhythmogenesis, the present data suggest contributions from dynamic changes in cellular signaling also play an important role in arrhythmogenesis.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Death, Sudden, Cardiac/etiology , Heart Rate , Heart Ventricles/physiopathology , Isoproterenol , Ventricular Function, Left , Action Potentials , Age Factors , Animals , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Death, Sudden, Cardiac/prevention & control , Disease Models, Animal , Female , Flecainide/pharmacology , Guinea Pigs , Heart Rate/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Oxidation-Reduction , Phosphorylation , Ryanodine Receptor Calcium Release Channel/metabolism , Time Factors , Ventricular Function, Left/drug effects
13.
Int J Neuropsychopharmacol ; 21(7): 687-696, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29635319

ABSTRACT

Background: Modafinil, a nonamphetaminic wake-promoting compound, is prescribed as first line therapy in narcolepsy, an invalidating disorder characterized by excessive daytime sleepiness and cataplexy. Although its mode of action remains incompletely known, recent studies indicated that modafinil modulates astroglial connexin-based gap junctional communication as administration of a low dose of flecainide, an astroglial connexin inhibitor, enhanced the wake-promoting and procognitive activity of modafinil in rodents and healthy volunteers. The aim of this study is to investigate changes in glucose cerebral metabolism in rodents, induced by the combination of modafinil+flecainide low dose (called THN102). Methods: The impact of THN102 on brain glucose metabolism was noninvasively investigated using 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography imaging in Sprague-Dawley male rats. Animals were injected with vehicle, flecainide, modafinil, or THN102 and further injected with 18F-2-fluoro-2-deoxy-D-glucose followed by 60-minute Positron Emission Tomography acquisition. 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography images were coregistered to a rat brain template and normalized from the total brain Positron Emission Tomography signal. Voxel-to-voxel analysis was performed using SPM8 software. Comparison of brain glucose metabolism between groups was then performed. Results: THN102 significantly increased regional brain glucose metabolism as it resulted in large clusters of 18F-2-fluoro-2-deoxy-D-glucose uptake localized in the cortex, striatum, and amygdala compared with control or drugs administered alone. These regions, highly involved in the regulation of sleep-wake cycle, emotions, and cognitive functions were hence quantitatively modulated by THN102. Conclusion: Data presented here provide the first evidence of a regional brain activation induced by THN102, currently being tested in a phase II clinical trial in narcoleptic patients.


Subject(s)
Amygdala/drug effects , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Flecainide/pharmacology , Fluorodeoxyglucose F18/pharmacokinetics , Modafinil/pharmacology , Positron-Emission Tomography/methods , Voltage-Gated Sodium Channel Blockers/pharmacology , Wakefulness-Promoting Agents/pharmacology , Amygdala/diagnostic imaging , Amygdala/metabolism , Animals , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Corpus Striatum/diagnostic imaging , Corpus Striatum/metabolism , Drug Combinations , Flecainide/administration & dosage , Male , Modafinil/administration & dosage , Rats , Rats, Sprague-Dawley , Voltage-Gated Sodium Channel Blockers/administration & dosage , Wakefulness-Promoting Agents/administration & dosage
14.
Exp Physiol ; 103(9): 1230-1242, 2018 09.
Article in English | MEDLINE | ID: mdl-29956404

ABSTRACT

NEW FINDINGS: What is the central question of this study? Is the slowed conduction upon premature ventricular activations during clinical electrophysiological testing attributable to the prolonged activation latency, or increased impulse propagation time, or both? What is the main finding and its importance? Prolonged activation latency at the stimulation site is the critical determinant of conduction slowing and associated changes in the ventricular response intervals in premature beats initiated during phase 3 repolarization in perfused guinea-pig heart. These relations are likely to have an effect on arrhythmia induction and termination independently of the presence of ventricular conduction defects or the proximity of the stimulation site to the re-entrant circuit. ABSTRACT: During cardiac electrophysiological testing, slowed conduction upon premature ventricular activation can limit the delivery of the closely coupled impulses from the stimulation site to the region of tachycardia origin. In order to examine the contributing factors, in this study, cardiac conduction intervals and refractory periods were determined from left ventricular (LV) and the right ventricular (RV) monophasic action potential recordings obtained in perfused guinea-pig hearts. A premature activation induced immediately after the termination of the refractory period was associated with conduction slowing. The latter was primarily accounted for by the markedly increased (+54%) activation latency at the LV stimulation site, with only negligible changes (+12%) noted in the LV-to-RV delay. The prolonged activation latency was acting to limit the shortest interval at which two successive action potentials can be induced in the LV and RV chambers. The prolongation of the activation latency in premature beats was accentuated upon an increase in the stimulating current intensity, or during hypokalaemia. This change was related to the reduced ratio of the refractory period to the action potential duration, which allowed extrastimulus capture to occur earlier during phase 3 repolarization. Flecainide, a Na+ channel blocker, prolonged both the activation latency and the LV-to-RV delay, without changing their relative contributions to conduction slowing. In summary, these findings suggest that the activation latency is the critical determinant of conduction slowing and associated changes in the ventricular response intervals upon extrastimulus application during phase 3 of the action potential.


Subject(s)
Heart/physiopathology , Ventricular Premature Complexes/physiopathology , Action Potentials , Animals , Electric Stimulation , Flecainide/pharmacology , Guinea Pigs , Heart Conduction System/physiopathology , Hypokalemia/physiopathology , In Vitro Techniques , Male , Refractory Period, Electrophysiological , Sodium Channel Blockers/pharmacology , Ventricular Function, Left , Ventricular Function, Right
15.
Clin Exp Pharmacol Physiol ; 45(12): 1286-1292, 2018 12.
Article in English | MEDLINE | ID: mdl-30028903

ABSTRACT

This study examined the inhibitory effect of flecainide, a class 1c antiarrhythmic agent (Na+ channel blocker), on voltage-dependent K+ (Kv) channels in smooth muscle cells isolated from coronary arteries. Flecainide decreased the vascular Kv channel current in a dose-dependent manner with an IC50 value of 5.90 ± 0.87 µmol/L and a Hill coefficient of 0.77 ± 0.06. Although the steady-state activation curve was not affected by flecainide, it shifted the steady-state inactivation curves toward a more negative potential. Application of train pulses such as 1 or 2 Hz did not change the flecainide-induced inhibition of Kv channels, indicating that the inhibitory effect of flecainide was not use-dependent. Using perforated-patch clamp experiments, we found that inhibition of Kv channels by flecainide caused membrane depolarization. Together, these results suggest that flecainide inhibits Kv channels in a concentration-dependent, but not use-dependent manner by changing the inactivation gating properties. Furthermore, Kv channel inhibition by flecainide occurs regardless of Na+ channel inhibition.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Coronary Vessels/cytology , Electrophysiological Phenomena/drug effects , Flecainide/pharmacology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Potassium/metabolism , Animals , Dose-Response Relationship, Drug , Inhibitory Concentration 50 , Ion Channel Gating/drug effects , Male , Membrane Potentials/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/metabolism , Rabbits
16.
J Emerg Med ; 54(3): 320-327, 2018 03.
Article in English | MEDLINE | ID: mdl-29269083

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is the most commonly encountered dysrhythmia in the emergency department, and its prevalence is increasing. A substantial proportion of these patients have recent-onset AF (<48 h). The poor prognosis associated with AF is being increasingly recognized, and there is some evidence for better outcomes in younger patients with recent-onset AF when sinus rhythm is restored. Flecainide is recommended in the latest international guidelines for cardioversion of recent-onset AF, but its safety and efficacy relative to other recommended agents are unclear. OBJECTIVE: Our aim was to clarify the Level 1 evidence for the use of i.v. flecainide in acute AF. METHODS: We performed a systematic review and meta-analysis of the literature. Medline, Ovid, Embase, and Cochrane Central databases were searched for relevant studies. Only randomized controlled trials (RCTs) of i.v. flecainide for acute conversion of recent-onset AF were selected for meta-analysis. RESULTS: Four hundred and three studies were screened, of which 11 RCTs were eligible for meta-analysis. Flecainide had high efficacy for cardioversion within 2 h (number needed to treat [NNT] = 1.8). Efficacy was superior to propafenone, amiodarone, procainamide, ibutilide, and sotalol (NNT = 4.3). There was no statistically significant difference in pro-dysrhythmia compared to these anti-dysrhythmics or placebo. CONCLUSIONS: Intravenous flecainide cardioversion could be a safe and effective option for emergency physicians to restore sinus rhythm in selected patients with acute AF.


Subject(s)
Atrial Fibrillation/drug therapy , Flecainide/pharmacology , Administration, Intravenous/methods , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Electric Countershock/methods , Emergency Service, Hospital/organization & administration , Flecainide/therapeutic use , Humans , United Kingdom
17.
Circ Res ; 116(8): 1324-35, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25648700

ABSTRACT

RATIONALE: Flecainide, a class 1c antiarrhythmic, has emerged as an effective therapy in preventing arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT) refractory to ß-adrenergic receptor blockade. It has been proposed that the clinical efficacy of flecainide in CPVT is because of the combined actions of direct blockade of ryanodine receptors (RyR2) and Na(+) channel inhibition. However, there is presently no direct evidence to support the notion that flecainide blocks RyR2 Ca(2+) flux in the physiologically relevant (luminal-to-cytoplasmic) direction. The mechanism of flecainide action remains controversial. OBJECTIVE: To examine, in detail, the effect of flecainide on the human RyR2 channel and to establish whether the direct blockade of physiologically relevant RyR2 ion flow by the drug contributes to its therapeutic efficacy in the clinical management of CPVT. METHODS AND RESULTS: Using single-channel analysis, we show that, even at supraphysiological concentrations, flecainide did not inhibit the physiologically relevant, luminal-to-cytosolic flux of cations through the channel. Moreover, flecainide did not alter RyR2 channel gating and had negligible effect on the mechanisms responsible for the sarcoplasmic reticulum charge-compensating counter current. Using permeabilized cardiac myocytes to eliminate any contribution of plasmalemmal Na(+) channels to the observed actions of the drug at the cellular level, flecainide did not inhibit RyR2-dependent sarcoplasmic reticulum Ca(2+) release. CONCLUSIONS: The principal action of flecainide in CPVT is not via a direct interaction with RyR2. Our data support a model of flecainide action in which Na(+)-dependent modulation of intracellular Ca(2+) handling attenuates RyR2 dysfunction in CPVT.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Flecainide/pharmacology , Myocytes, Cardiac/drug effects , Ryanodine Receptor Calcium Release Channel/drug effects , Tachycardia, Ventricular/drug therapy , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Male , Membrane Potentials , Myocytes, Cardiac/metabolism , Potassium Channels/drug effects , Potassium Channels/metabolism , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Transfection
18.
BMC Cardiovasc Disord ; 17(1): 289, 2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29221440

ABSTRACT

BACKGROUND: Atrial fibrillatory cycle length has been considered one of the indices of atrial electrical remodelling during atrial fibrillation (AF), which can be assessed from surface ECG by computer-assisted calculation of atrial fibrillatory rate (AFR). Horses have been suggested as a bona fide model for AF studies since horses too, develop lone AF, however data on AF characteristics in horses are extremely sparse and non-invasive characterization of AF complexity using surface ECG processing has not been reported. AIM: The aim was to study characteristics of induced AF and its modification by flecainide. METHODS: The study group consisted on 3 horses with spontaneous persistent AF and 13 with pace-induced AF. Seven horses were treated with saline (control) and eight with flecainide (2 mg/kg). ECGs were analysed using spatiotemporal cancellation of QRST complexes and calculation of AFR from the residual atrial signal. RESULTS: At AF onset, AFR was 295 ± 52 fibrillations per minute (fpm) in the horses with induced AF treated with flecainide, 269 ± 36 fpm in the control group (ns), and 364 ± 26 fpm in the horses with spontaneous persistent AF (P < 0.05 compared to the control group). Flecainide caused a decrease in AFR in all animals and restored sinus rhythm in the animals with induced AF. In the control animals, AFR increased from 269 ± 36 fpm to a plateau of 313 ± 14 fpm before decreasing to 288 ± 28 fpm during the last 10% of the AF episodes preceding spontaneous conversion (P < 0.05). CONCLUSION: AFR in horses with induced AF resembles AFR in humans with paroxysmal AF. Flecainide caused a rapid decrease in AFR in all horses, further supporting the method to be a non-invasive technique to study the effect of antiarrhythmic compounds.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/prevention & control , Flecainide/pharmacology , Heart Atria/drug effects , Heart Rate/drug effects , Action Potentials/drug effects , Animals , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Cardiac Pacing, Artificial , Disease Models, Animal , Electrocardiography , Female , Heart Atria/physiopathology , Horses , Male , Time Factors
19.
Eur Heart J ; 37(7): 630-7, 2016 Feb 14.
Article in English | MEDLINE | ID: mdl-26261291

ABSTRACT

AIMS: Published reports regarding inferolateral early repolarization (ER) syndrome (ERS) before 2013 possibly included patients with Brugada-pattern electrocardiogram (BrP-ECG) recorded only in the high intercostal spaces (HICS). We investigated the significance of HICS ECG recording in ERS patients. METHODS AND RESULTS: Fifty-six patients showing inferolateral ER in the standard ECG and spontaneous ventricular fibrillation (VF) not linked to structural heart disease underwent drug provocation tests by sodium channel blockade with right precordial ECG (V1-V3) recording in the 2nd-4th intercostal spaces. The prevalence and long-term outcome of ERS patients with and without BrP-ECG in HICS were investigated. After 18 patients showing type 1 BrP-ECG in the standard ECG were excluded, 38 patients (34 males, mean age; 40.4 ± 13.6 years) were classified into four groups [group A (n = 6;16%):patients with ER and type 1 BrP-ECG only in HICS, group B (n = 5;13%):ERS with non-type 1 BrP-ECG only in HICS, group C (n = 8;21%):ERS with non-type 1 BrP-ECG in the standard ECG, and group D (n = 19;50%):ERS only, spontaneously or after drug provocation test]. During follow-up of 110.0 ± 55.4 months, the rate of VF recurrence including electrical storm was significantly higher in groups A (4/6:67%), B (4/5:80%), and C (4/8:50%) compared with D (2/19:11%) (A, B, and C vs. D, P < 0.05). CONCLUSIONS: Approximately 30% of the patients with ERS who had been diagnosed with the previous criteria showed BrP-ECG only in HICS. Ventricular fibrillation mostly recurred in patients showing BrP-ECG in any precordial lead including HICS; these comprised 50% of the ERS cohort.


Subject(s)
Electrocardiography , Ventricular Fibrillation/etiology , Adult , Anti-Arrhythmia Agents/pharmacology , Brugada Syndrome/complications , Brugada Syndrome/diagnosis , Electrophysiologic Techniques, Cardiac/methods , Female , Flecainide/pharmacology , Humans , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Male , Prognosis , Recurrence , Risk Assessment , Ventricular Fibrillation/diagnosis
20.
J Physiol ; 594(3): 567-93, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26515697

ABSTRACT

KEY POINTS: The mechanism of therapeutic efficacy of flecainide for catecholaminergic polymorphic ventricular tachycardia (CPVT) is unclear. Model predictions suggest that Na(+) channel effects are insufficient to explain flecainide efficacy in CPVT. This study represents a first step toward predicting therapeutic mechanisms of drug efficacy in the setting of CPVT and then using these mechanisms to guide modelling and simulation to predict alternative drug therapies. Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited arrhythmia syndrome characterized by fatal ventricular arrhythmias in structurally normal hearts during ß-adrenergic stimulation. Current treatment strategies include ß-blockade, flecainide and ICD implementation--none of which is fully effective and each comes with associated risk. Recently, flecainide has gained considerable interest in CPVT treatment, but its mechanism of action for therapeutic efficacy is unclear. In this study, we performed in silico mutagenesis to construct a CPVT model and then used a computational modelling and simulation approach to make predictions of drug mechanisms and efficacy in the setting of CPVT. Experiments were carried out to validate model results. Our simulations revealed that Na(+) channel effects are insufficient to explain flecainide efficacy in CPVT. The pure Na(+) channel blocker lidocaine and the antianginal ranolazine were additionally tested and also found to be ineffective. When we tested lower dose combination therapy with flecainide, ß-blockade and CaMKII inhibition, our model predicted superior therapeutic efficacy than with flecainide monotherapy. Simulations indicate a polytherapeutic approach may mitigate side-effects and proarrhythmic potential plaguing CPVT pharmacological management today. Importantly, our prediction of a novel polytherapy for CPVT was confirmed experimentally. Our simulations suggest that flecainide therapeutic efficacy in CPVT is unlikely to derive from primary interactions with the Na(+) channel, and benefit may be gained from an alternative multi-drug regimen.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Flecainide/pharmacology , Models, Cardiovascular , Tachycardia, Ventricular/physiopathology , Animals , Animals, Genetically Modified , Anti-Arrhythmia Agents/therapeutic use , Electrocardiography , Flecainide/therapeutic use , Mice , Rabbits , Ryanodine Receptor Calcium Release Channel/physiology , Sodium Channels/physiology , Tachycardia, Ventricular/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL