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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Europace ; 24(12): 2015-2027, 2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-35726875

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Canal de Sodio Activado por Voltaje NAV1.5 , Humanos , Potenciales de Acción/fisiología , Arritmias Cardíacas/genética , Flecainida/farmacología , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Fenotipo , Sodio/metabolismo
2.
Int J Mol Sci ; 22(13)2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34281161

RESUMEN

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are used for genetic models of cardiac diseases. We report an arrhythmia syndrome consisting of Early Repolarization Syndrome (ERS) and Short QT Syndrome (SQTS). The index patient (MMRL1215) developed arrhythmia-mediated syncope after electrocution and was found to carry six mutations. Functional alterations resulting from these mutations were examined in patient-derived hiPSC-CMs. Electrophysiological recordings were made in hiPSC-CMs from MMRL1215 and healthy controls. ECG analysis of the index patient showed slurring of the QRS complex and QTc = 326 ms. Action potential (AP) recordings from MMRL1215 myocytes showed slower spontaneous activity and AP duration was shorter. Field potential recordings from MMRL1215 hiPSC-CMs lack a "pseudo" QRS complex suggesting reduced inward current(s). Voltage clamp analysis of ICa showed no difference in the magnitude of current. Measurements of INa reveal a 60% reduction in INa density in MMRL1215 hiPSC-CMs. Steady inactivation and recovery of INa was unaffected. mRNA analysis revealed ANK2 and SCN5A are significantly reduced in hiPSC-CM derived from MMRL1215, consistent with electrophysiological recordings. The polygenic cause of ERS/SQTS phenotype is likely due to a loss of INa due to a mutation in PKP2 coupled with and a gain of function in IK,ATP due to a mutation in ABCC9.


Asunto(s)
Arritmias Cardíacas/genética , Miocitos Cardíacos/metabolismo , Potenciales de Acción/genética , Adenosina Trifosfato/metabolismo , Ancirinas/genética , Ancirinas/metabolismo , Arritmias Cardíacas/fisiopatología , Fenómenos Electrofisiológicos , Variación Genética/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp/métodos , Placofilinas/genética , Potasio/metabolismo , Sodio/metabolismo , Receptores de Sulfonilureas/genética
3.
Pflugers Arch ; 469(5-6): 703-712, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28243733

RESUMEN

The collar of the pulmonary vein (PV) is the focal point for the initiation of atrial arrhythmias, but the mechanisms underlying how PV cells differ from neighboring left atrial tissue are unclear. We examined the biophysical and molecular properties of INa in cells isolated from the canine pulmonary sleeve and compared the properties to left atrial tissue. PV and left atrial myocytes were isolated and patch clamp techniques were used to record INa. Action potential recordings from either tissue type were made using high-resistance electrodes. mRNA was determined using quantitative RT-PCR and proteins were determined by Western blot. Analysis of the action potential characteristics showed that PV tissue had a lower Vmax compared with left atrial tissue. Fast INa showed that current density was slightly lower in PV cells compared with LA cells (-96 ± 18.7 pA/pF vs. -120 ± 6.7 pA/pF, respectively, p < 0.05). The recovery from inactivation of INa in PV cells was slightly slower but no marked difference in steady-state inactivation was noted. Analysis of late INa during a 225-ms pulse showed that late INa was significantly smaller in PV cells compared to LA cells at all measured time points into the pulse. These results suggest PV cells have lower density of both peak and late INa. Molecular analysis of Nav1.5 and the four beta subunits showed lower levels of Nav1.5 as well as Navß1 subunits, confirming the biophysical findings. These data show that a lower density of INa may lead to depression of excitability and predispose the PV collar to re-entrant circuits under pathophysiological conditions.


Asunto(s)
Potenciales de Acción , Atrios Cardíacos/citología , Miocitos Cardíacos/fisiología , Miocitos del Músculo Liso/fisiología , Venas Pulmonares/citología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Células Cultivadas , Perros , Femenino , Masculino , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/metabolismo , Sodio/metabolismo
4.
J Mol Cell Cardiol ; 84: 52-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25889894

RESUMEN

The inward rectifier potassium current, IK1, contributes to the terminal phase of repolarization of the action potential (AP), as well as the value and stability of the resting membrane potential. Regional variation in IK1 has been noted in the canine heart, but the biophysical properties have not been directly compared. We examined the properties and functional contribution of IK1 in isolated myocytes from ventricular, atrial and Purkinje tissue. APs were recorded from canine left ventricular midmyocardium, left atrial and Purkinje tissue. The terminal rate of repolarization of the AP in ventricle, but not in Purkinje, depended on changes in external K(+) ([K(+)]o). Isolated ventricular myocytes had the greatest density of IK1 while atrial myocytes had the lowest. Furthermore, the outward component of IK1 in ventricular cells exhibited a prominent outward component and steep negative slope conductance, which was also enhanced in 10 mM [K(+)]o. In contrast, both Purkinje and atrial cells exhibited little outward IK1, even in the presence of 10 mM [K(+)]o, and both cell types showed more persistent current at positive potentials. Expression of Kir2.1 in the ventricle was 76.9-fold higher than that of atria and 5.8-fold higher than that of Purkinje, whereas the expression of Kir2.2 and Kir2.3 subunits was more evenly distributed in Purkinje and atria. Finally, AP clamp data showed distinct contributions of IK1 for each cell type. IK1 and Kir2 subunit expression varies dramatically in regions of the canine heart and these regional differences in Kir2 expression likely underlie regional distinctions in IK1 characteristics, contributing to variations in repolarization in response to in [K(+)]o changes.


Asunto(s)
Potenciales de Acción/fisiología , Corazón/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Perros , Femenino , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Activación del Canal Iónico , Cinética , Masculino , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Poliaminas/metabolismo , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Células de Purkinje/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Europace ; 16 Suppl 4: iv46-iv55, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25362170

RESUMEN

AIMS: The study investigates how increased Ito, as mediated by the activator NS5806, affects excitation-contraction coupling in chronic heart failure (HF). We hypothesized that restoring spike-and-dome morphology of the action potential (AP) to a healthy phenotype would be insufficient to restore the intracellular Ca(2) (+) transient (CaT), due to HF-induced remodelling of Ca(2+) handling. METHODS AND RESULTS: An existing mathematical model of the canine ventricular myocyte was modified to incorporate recent experimental data from healthy and failing myocytes, resulting in models of both healthy and HF epicardial, midmyocardial, and endocardial cell variants. Affects of NS5806 were also included in HF models through its direct interaction with Kv4.3 and Kv1.4. Single-cell simulations performed in all models (control, HF, and HF + drug) and variants (epi, mid, and endo) assessed AP morphology and underlying ionic processes with a focus on calcium transients (CaT), how these were altered in HF across the ventricular wall, and the subsequent effects of varying compound concentration in HF. Heart failure model variants recapitulated a characteristic increase in AP duration (APD) in the disease. The qualitative effects of application of half-maximal effective concentration (EC50) of NS5806 on APs and CaT are heterogeneous and non-linear. Deepening in the AP notch with drug is a direct effect of the activation of Ito; both Ito and consequent alteration of IK1 kinetics cause decrease in AP plateau potential. Decreased APD50 and APD90 are both due to altered IK1. Analysis revealed that drug effects depend on transmurality. Ca(2+) transient morphology changes-increased amplitude and shorter time to peak-are due to direct increase in ICa,L and indirect larger SR Ca(2+) release subsequent to Ito activation. CONCLUSIONS: Downstream effects of a compound acting exclusively on sarcolemmal ion channels are difficult to predict. Remediation of APD to pre-failing state does not ameliorate dysfunction in CaT; however, restoration of notch depth appears to impart modest benefit and a likelihood of therapeutic value in modulating early repolarization.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Simulación por Computador , Insuficiencia Cardíaca/tratamiento farmacológico , Modelos Cardiovasculares , Miocitos Cardíacos/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Tetrazoles/farmacología , Potenciales de Acción , Animales , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Acoplamiento Excitación-Contracción/efectos de los fármacos , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Cinética , Canal de Potasio Kv1.4/agonistas , Canal de Potasio Kv1.4/metabolismo , Miocitos Cardíacos/metabolismo , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Canales de Potasio Shal/agonistas , Canales de Potasio Shal/metabolismo
6.
PLoS One ; 19(1): e0287206, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38181028

RESUMEN

We identified two different inherited mutations in KCNH2 gene, or human ether-a-go-go related gene (hERG), which are linked to Long QT Syndrome. The first mutation was in a 1-day-old infant, whereas the second was in a 14-year-old girl. The two KCNH2 mutations were transiently transfected into either human embryonic kidney (HEK) cells or human induced pluripotent stem-cell derived cardiomyocytes. We performed associated multiscale computer simulations to elucidate the arrhythmogenic potentials of the KCNH2 mutations. Genetic screening of the first and second index patients revealed a heterozygous missense mutation in KCNH2, resulting in an amino acid change (P632L) in the outer loop of the channel and substitution at position 428 from serine to proline (S428P), respectively. Heterologous expression of P632L and S428P into HEK cells produced no hERG current compared to the wild type (WT). Moreover, the co-transfection of WT and P632L yielded no hERG current; however, the co-transfection of WT and S428P yielded partial hERG current. Action potentials were prolonged in a complete or partial blockade of hERG current from computer simulations which was more severe in Purkinje than ventricular myocytes. Three dimensional simulations revealed a higher susceptibility to reentry in the presence of hERG current blockade. Our experimental findings suggest that both P632L and S428P mutations may impair the KCNH2 gene. The Purkinje cells exhibit a more severe phenotype than ventricular myocytes, and the hERG current blockade renders the ventricles an arrhythmogenic substrate from computer modeling.


Asunto(s)
Canal de Potasio ERG1 , Síndrome de QT Prolongado , Adolescente , Femenino , Humanos , Lactante , Potenciales de Acción , Simulación por Computador , Células Epiteliales , Canal de Potasio ERG1/genética , Síndrome de QT Prolongado/genética , Mutación
7.
J Mol Cell Cardiol ; 64: 79-89, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24035801

RESUMEN

BACKGROUND: Developmental changes in the electrical characteristics of the ventricular myocardium are not well defined. This study examines the contribution of inwardly rectifying K(+) current (IK1), transient outward K(+) current (Ito), delayed rectifier K(+) currents (IKr and IKs) and sodium channel current (INa) to repolarization in the canine neonate myocardium. METHODS: Single myocytes isolated from the left ventricle of 2-3week old canine neonate hearts were studied using patch-clamp techniques. RESULTS: Neonate cells were ~6-fold smaller than those of adults (28.8±8.8 vs. 176±6.7pF). IK1 was larger in neonate myocytes and displayed a substantial inward component and an outward component with negative slope conductance, peaking at -60mV (4.13 pA/pF). IKr tail currents (at -40mV), were small (<20pA). IKs could not be detected, even after exposure to isoproterenol (100nM). Ito was also absent in the neonate, consistent with the absence of a phase 1 in the action potential. Peak INa, late INa and ICa were smaller in the neonate compared with adults. KCND3, KCNIP2 and KCNQ1 mRNA expression was half, while KCNH2 was equal and KCNJ2 was greater in the neonate when compared with adults. CONCLUSIONS: Two major repolarizing K(+) currents (IKs and Ito) present in adult ventricular cells are absent in the 2week old neonate. Peak and late INa are significantly smaller in the neonate. Our results suggest that the absence of these two currents in the neonate heart may increase the susceptibility to arrhythmias under certain long QT conditions.


Asunto(s)
Canales Iónicos/genética , Canales Iónicos/metabolismo , Función Ventricular/fisiología , Potenciales de Acción , Animales , Animales Recién Nacidos , Antiarrítmicos/farmacología , Calcio/metabolismo , Perros , Femenino , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Humanos , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Piperidinas/farmacología , Potasio/metabolismo , Canales de Potasio/fisiología , Piridinas/farmacología , Sodio/metabolismo , Función Ventricular/efectos de los fármacos
8.
J Mol Cell Cardiol ; 60: 36-46, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23542310

RESUMEN

BACKGROUND: The ability to recapitulate mature adult phenotypes is critical to the development of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) as models of disease. The present study examines the characteristics of the transient outward current (Ito) and its contribution to the hiPSC-CM action potential (AP). METHOD: Embryoid bodies were made from a hiPS cell line reprogrammed with Oct4, Nanog, Lin28 and Sox2. Sharp microelectrodes were used to record APs from beating-clusters (BC) and patch-clamp techniques were used to record Ito in single hiPSC-CM. mRNA levels of Kv1.4, KChIP2 and Kv4.3 were quantified from BCs. RESULTS: BCs exhibited spontaneous beating (60.5±2.6 bpm) and maximum-diastolic-potential (MDP) of 67.8±0.8 mV (n=155). A small 4-aminopyridine-sensitive phase-1-repolarization was observed in only 6/155 BCs. A robust Ito was recorded in the majority of cells (13.7±1.9 pA/pF at +40 mV; n=14). Recovery of Ito from inactivation (at -80 mV) showed slow kinetics (τ1=200±110 ms (12%) and τ2=2380±240 ms (80%)) accounting for its minimal contribution to the AP. Transcript data revealed relatively high expression of Kv1.4 and low expression of KChIP2 compared to human native ventricular tissues. Mathematical modeling predicted that restoration of IK1 to normal levels would result in a more negative MDP and a prominent phase-1-repolarization. CONCLUSION: The slow recovery kinetics of Ito coupled with a depolarized MDP account for the lack of an AP notch in the majority of hiPSC-CM. These characteristics reveal a deficiency for the development of in vitro models of inherited cardiac arrhythmia syndromes in which Ito-induced AP notch is central to the disease phenotype.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Potenciales de la Membrana/fisiología , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Proteínas de Interacción con los Canales Kv/metabolismo , Canal de Potasio Kv1.4/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Miocitos Cardíacos/citología , Canales de Potasio Shal/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 305(1): H66-75, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23645460

RESUMEN

Acetylcholine (ACh) release from the vagus nerve slows heart rate and atrioventricular conduction. ACh stimulates a variety of receptors and channels, including an inward rectifying current [ACh-dependent K⁺ current (IK,ACh)]. The effect of ACh in the ventricle is still debated. We compared the effect of ACh on action potentials in canine atria, Purkinje, and ventricular tissue as well as on ionic currents in isolated cells. Action potentials were recorded from ventricular slices, Purkinje fibers, and arterially perfused atrial preparations. Whole cell currents were recorded under voltage-clamp conditions, and unloaded cell shortening was determined on isolated cells. The effect of ACh (1-10 µM) as well as ACh plus tertiapin, an IK,ACh-specific toxin, was tested. In atrial tissue, ACh hyperpolarized the membrane potential and shortened the action potential duration (APD). In Purkinje and ventricular tissues, no significant effect of ACh was observed. Addition of ACh to atrial cells activated a large inward rectifying current (from -3.5 ± 0.7 to -23.7 ± 4.7 pA/pF) that was abolished by tertiapin. This current was not observed in other cell types. A small inhibition of Ca²âº current (ICa) was observed in the atria, endocardium, and epicardium after ACh. ICa inhibition increased at faster pacing rates. At a basic cycle length of 400 ms, ACh (1 µM) reduced ICa to 68% of control. In conclusion, IK,ACh is highly expressed in atria and is negligible/absent in Purkinje, endocardial, and epicardial cells. In all cardiac tissues, ACh caused rate-dependent inhibition of ICa.


Asunto(s)
Acetilcolina/farmacología , Potenciales de Acción/efectos de los fármacos , Corazón/fisiología , Potenciales de Acción/fisiología , Animales , Perros , Corazón/anatomía & histología , Miocitos Cardíacos/fisiología , Especificidad de Órganos , Potasio/metabolismo
10.
Bull Math Biol ; 75(5): 752-73, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23595789

RESUMEN

We present an inversion of the Hodgkin-Huxley formalism to estimate initial conditions and model parameters, including functions of voltage, from the solutions of the underlying ordinary differential equation (ODE) subjected to multiple voltage step stimulations. As such, the procedure constitutes a means to estimate the parameters including functions of voltage of an Hodgkin-Huxley formalism from experimental data.The basic idea was developed in a previous communication (SIAM J. Appl. Math. 64:1264-1274, 2009). The inversion in question applies to currents exhibiting activation and inactivation, but the version, as published previously, cannot estimate the unknowns for channels that rapidly inactivate just after a brief opening. In such cases, the amplitude of the current, in a given voltage range, is too small to be detectable by the instrumentation using previously applied experimental protocols. This is, for example, the case for the sodium channels in a number of excitable tissue for potential in the vicinity of the cell resting potential. The current communication extends the inversion procedure in a manner to overcome this limitation.Furthermore, within the inversion framework, we can determine whether the data at the basis of the estimation sufficiently constrains the estimation problem, i.e., whether it is complete. We exploit this element of our method to document a set of stimulation protocols that constitute a complete data set for the purpose of inverting the Hodgkin-Huxley formalism.


Asunto(s)
Activación del Canal Iónico , Modelos Biológicos , Algoritmos , Animales , Perros , Conceptos Matemáticos , Miocitos Cardíacos/metabolismo , Dinámicas no Lineales , Técnicas de Placa-Clamp , Canales de Sodio/metabolismo
11.
J Mol Cell Cardiol ; 52(6): 1291-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22434032

RESUMEN

BACKGROUND: Remodeling of ion channel expression is well established in heart failure (HF). We determined the extent to which I(to) is reduced in tachypacing-induced HF and assessed the ability of an I(to) activator (NS5806) to recover this current. METHOD AND RESULTS: Whole-cell patch clamp was used to record I(to) in epicardial (Epi) ventricular myocytes. Epi- and endocardial action potentials were recorded from left ventricular wedge preparations. Right ventricular tachypacing-induced heart failure reduced I(to) density in Epi myocytes (Control=22.1±1.9pA/pF vs 16.1±1.4 after 2weeks and 10.7±1.4pA/pF after 5 weeks, +50mV). Current decay as well as recovery of I(to) from inactivation progressively slowed with the development of heart failure. Reduction of I(to) density was paralleled by a reduction in phase 1 magnitude, epicardial action potential notch and J wave amplitude recorded from coronary-perfused left ventricular wedge preparations. NS5806 increased I(to) (at +50mV) from 16.1±1.4 to 23.9±2.1pA/pF (p<0.05) at 2weeks and from 10.7±1.4 to 14.4±1.9pA/pF (p<0.05) in 5 weeks tachypaced dogs. NS5806 increased both fast and slow phases of I(to) recovery in 2 and 5-week HF cells and restored the action potential notch and J wave in wedge preparations from HF dogs. CONCLUSIONS: The I(to) agonist NS5806 increases the rate of recovery and density of I(to), thus reversing the HF-induced reduction in these parameters. In wedge preparations from HF dogs, NS5806 restored the spike-and-dome morphology of the Epi action potential providing proof of principal that some aspects of electrical remodelling during HF can be pharmacologically reversed.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Canales de Potasio/metabolismo , Potasio/metabolismo , Animales , Gasto Cardíaco/efectos de los fármacos , Modelos Animales de Enfermedad , Perros , Ventrículos Cardíacos/efectos de los fármacos , Hemodinámica , Pericardio/efectos de los fármacos , Pericardio/metabolismo , Pericardio/fisiopatología , Compuestos de Fenilurea/farmacología , Canales de Potasio/agonistas , Tetrazoles/farmacología
12.
Am J Physiol Heart Circ Physiol ; 301(3): H936-44, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21685271

RESUMEN

Cardiac ischemia reduces excitability in ventricular tissue. Acidosis (one component of ischemia) affects a number of ion currents. We examined the effects of extracellular acidosis (pH 6.6) on peak and late Na(+) current (I(Na)) in canine ventricular cells. Epicardial and endocardial myocytes were isolated, and patch-clamp techniques were used to record I(Na). Action potential recordings from left ventricular wedges exposed to acidic Tyrode solution showed a widening of the QRS complex, indicating slowing of transmural conduction. In myocytes, exposure to acidic conditions resulted in a 17.3 ± 0.9% reduction in upstroke velocity. Analysis of fast I(Na) showed that current density was similar in epicardial and endocardial cells at normal pH (68.1 ± 7.0 vs. 63.2 ± 7.1 pA/pF, respectively). Extracellular acidosis reduced the fast I(Na) magnitude by 22.7% in epicardial cells and 23.1% in endocardial cells. In addition, a significant slowing of the decay (time constant) of fast I(Na) was observed at pH 6.6. Acidosis did not affect steady-state inactivation of I(Na) or recovery from inactivation. Analysis of late I(Na) during a 500-ms pulse showed that the acidosis significantly reduced late I(Na) at 250 and 500 ms into the pulse. Using action potential clamp techniques, application of an epicardial waveform resulted in a larger late I(Na) compared with when an endocardial waveform was applied to the same cell. Acidosis caused a greater decrease in late I(Na) when an epicardial waveform was applied. These results suggest acidosis reduces both peak and late I(Na) in both cell types and contributes to the depression in cardiac excitability observed under ischemic conditions.


Asunto(s)
Acidosis/metabolismo , Ventrículos Cardíacos/metabolismo , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Sodio/metabolismo , Potenciales de Acción , Análisis de Varianza , Animales , Perros , Endocardio/metabolismo , Femenino , Concentración de Iones de Hidrógeno , Cinética , Masculino , Técnicas de Placa-Clamp , Pericardio/metabolismo
13.
J Cardiovasc Electrophysiol ; 22(6): 698-705, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21159010

RESUMEN

INTRODUCTION: Angiotensin-converting enzyme (ACE) inhibitors and angiotensin II-receptor blockers (ARBs) are prototypes of "upstream" therapy for the management of atrial fibrillation (AF). Ectopic activity arising from the PV sleeves plays a prominent role in the development of AF. METHODS: Transmembrane action potentials were recorded from canine superfused left superior or inferior PV sleeves using standard microelectrode techniques. Acetylcholine (ACh, 1 µM), isoproterenol (1 µM), high calcium ([Ca(2+)](o) = 5.4 mM) or a combination was used to induce early or delayed afterdepolarizations (EADs or DADs) and triggered activity. RESULTS: The ARB losartan (1 µM, n = 5) and the ACE inhibitor enalapril (10 µM, n = 5) produced no significant change in action potential duration, maximum rate of rise of action potential upstroke (V(max)), action potential amplitude or take-off potential at basic cycle lengths of 200 to 2000 ms. Losartan (1 µM) and enalapril (10-20 µM) markedly attenuated or suppressed EADs and DAD-induced triggered activity elicited by exposure of the PV sleeves to ACh, isoproterenol or high calcium following rapid pacing in 6 of 6 (losartan) and 4 of 5 (enalapril) PV sleeve preparations. Neither losartan nor enalapril altered Ca(2+) or K(+) channel currents in enzymatically-dissociated atrial myocytes at these concentrations. CONCLUSIONS: Our data suggest that in addition to their "upstream" effects to reduce atrial structural remodeling, ACE inhibitors and ARBs exert a "direct" antiarrhythmic effect by suppressing triggers responsible for the genesis of AF and other atrial arrhythmias.


Asunto(s)
Enalapril/administración & dosificación , Sistema de Conducción Cardíaco/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Losartán/administración & dosificación , Venas Pulmonares/fisiopatología , Animales , Antiarrítmicos/administración & dosificación , Perros , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Sistema de Conducción Cardíaco/efectos de los fármacos , Venas Pulmonares/efectos de los fármacos
14.
J Cardiovasc Electrophysiol ; 22(9): 1057-66, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21457383

RESUMEN

INTRODUCTION: NS5806 activates the transient outward potassium current (I(to) ) in canine ventricular cells. We compared the effects of NS5806 on canine atrial versus ventricular tissues and myocytes. METHODS AND RESULTS: NS5806 (10 µM) was evaluated in arterially perfused canine right atrial and right ventricular wedge preparations. In ventricular wedges NS5806 (10 µM) accentuated phase 1 in epicardium (Epi), with little effect in endocardium (Endo), resulting in augmented J-waves on the ECG. In contrast, application of NS5806 (10 µM) to atrial preparations had no effect on phase 1 repolarization but significantly decreased upstroke velocity (dV/dt) and depressed excitability, consistent with sodium channel block. Current and voltage-clamp recordings were made in the absence and presence of NS5806 in (10 µM) enzymatically dissociated atrial and ventricular myocytes. In ventricular myocytes, NS5806 increased I(to) magnitude by 80% and 16% in Epi and Endo, respectively (at +40 mV). In atrial myocytes, NS5806 increased peak I(to) by 25% and had no effect on the sustained current, I(Kur) . Under control conditions, I(Na) density in atrial myocytes was nearly double that in ventricular myocytes. NS5806 caused a shift in steady-state mid-inactivation (V(1/2)) from -73.9 ± 0.27 to -77.3 ± 0.21 mV in ventricular and from -82.6 ± 0.12 to -85.1 ± 0.11 mV in atrial cells, resulting in reduction of I(Na) in both cell types. Expression of mRNA encoding putative I(Na) and I(to) channel subunits was evaluated by qPCR. CONCLUSION: NS5806 produces a prominent augmentation of I(to) with little effect on I(Na) in the ventricles, but a potent inhibition of I(Na) with little augmentation of I(to) in atria.


Asunto(s)
Potenciales de Acción/fisiología , Miocitos Cardíacos/fisiología , Compuestos de Fenilurea/farmacología , Canales de Potasio/agonistas , Canales de Potasio/fisiología , Tetrazoles/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Perros , Femenino , Atrios Cardíacos/citología , Atrios Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Masculino , Miocitos Cardíacos/efectos de los fármacos
15.
Can J Physiol Pharmacol ; 89(10): 723-36, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21895525

RESUMEN

BACKGROUND: Mutations in the SCN5A gene have been linked to Brugada syndrome (BrS), conduction disease, Long QT syndrome (LQT3), atrial fibrillation (AF), and to pre- and neonatal ventricular arrhythmias. OBJECTIVE: The objective of this study is to characterize a novel mutation in Na(v)1.5 found in a newborn with fetal chaotic atrial tachycardia, post-partum intraventricular conduction delay, and QT interval prolongation. METHODS: Genomic DNA was isolated and all exons and intron borders of 15 ion-channel genes were sequenced, revealing a novel missense mutation (Q270K) in SCN5A. Na(v)1.5 wild type (WT) and Q270K were expressed in CHO-K1 with and without the Na(v)ß1 subunit. Results. Patch-clamp analysis showed ∼40% reduction in peak sodium channel current (I(Na)) density for Q270K compared with WT. Fast and slow decay of I(Na) were significantly slower in Q270K. Steady-state activation and inactivation of Q270K channels were shifted to positive potentials, and window current was increased. The tetrodotoxin-sensitive late I(Na) was increased almost 3-fold compared with WT channels. Ranolazine reduced late I(Na) in WT and Q270K channels, while exerting minimal effects on peak I(Na). CONCLUSION: The Q270K mutation in SCN5A reduces peak I(Na) while augmenting late I(Na), and may thus underlie the development of atrial tachycardia, intraventricular conduction delay, and QT interval prolongation in an infant.


Asunto(s)
Arritmias Cardíacas/genética , Potenciales de la Membrana/genética , Mutación Missense/genética , Canales de Sodio/genética , Animales , Células CHO , Línea Celular Transformada , Cricetinae , Análisis Mutacional de ADN , Femenino , Humanos , Recién Nacido , Canales Iónicos/genética , Potenciales de la Membrana/fisiología , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp/métodos , Síndrome
16.
Front Physiol ; 12: 675867, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220540

RESUMEN

The formulation of in silico biophysical models generally requires optimization strategies for reproducing experimentally observed phenomena. In electrophysiological modeling, robust nonlinear regressive methods are often crucial for guaranteeing high fidelity models. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), though nascent, have proven to be useful in cardiac safety pharmacology, regenerative medicine, and in the implementation of patient-specific test benches for investigating inherited cardiac disorders. This study demonstrates the potency of heuristic techniques at formulating biophysical models, with emphasis on a hiPSC-CM model using a novel genetic algorithm (GA) recipe we proposed. The proposed GA protocol was used to develop a hiPSC-CM biophysical computer model by fitting mathematical formulations to experimental data for five ionic currents recorded in hiPSC-CMs. The maximum conductances of the remaining ionic channels were scaled based on recommendations from literature to accurately reproduce the experimentally observed hiPSC-CM action potential (AP) metrics. Near-optimal parameter fitting was achieved for the GA-fitted ionic currents. The resulting model recapitulated experimental AP parameters such as AP durations (APD50, APD75, and APD90), maximum diastolic potential, and frequency of automaticity. The outcome of this work has implications for validating the biophysics of hiPSC-CMs in their use as viable substitutes for human cardiomyocytes, particularly in cardiac safety pharmacology and in the study of inherited cardiac disorders. This study presents a novel GA protocol useful for formulating robust numerical biophysical models. The proposed protocol is used to develop a hiPSC-CM model with implications for cardiac safety pharmacology.

17.
J Mol Cell Cardiol ; 48(1): 191-200, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19632239

RESUMEN

To examine the electrophysiological and molecular properties of the transient outward current (I(to)) in canine left ventricle using a novel I(to) activator, NS5806, I(to) was measured in isolated epicardial (Epi), midmyocardial (Mid) and endocardial (Endo) cells using whole-cell patch-clamp techniques. NS5806 activation of K(v)4.3 current was also studied in CHO-K1 cells and Xenopus laevis oocytes. In CHO-K1 cells co-transfected with K(v)4.3 and KChIP2, NS5806 (10 microM) caused a 35% increase in current amplitude and a marked slowing of current decay with tau increasing from 7.0+/-0.4 to 10.2+/-0.3 ms. In the absence of KChIP2, current decay was unaffected by NS5806. In ventricular myocytes, NS5806 increased I(to) density by 80%, 82%, and 16% in Epi, Mid, and Endo myocytes, respectively (at +40 mV) and shifted steady-state inactivation to negative potentials. NS5806 also significantly slowed decay of I(to), increasing total charge to 227%, 192% and 83% of control in Epi, Mid and Endo cells, respectively (+40 mV, p<0.05). Quantification of K(v)4.3 and KChIP2 mRNA in the 3 ventricular cell types revealed that levels of K(v)4.3 message was uniform but those of KChIP2 were significantly greater in Epi and Mid cells. The KChIP2 gradient was confirmed at the protein level by Western blot. Our results suggest that NS5806 augments I(to) by increasing current density and slowing decay and that both depend on the presence of KChIP2. I(to) and its augmentation by NS5806 are greatest in Epi and Mid cells because KChIP2 levels are highest in these cell types.


Asunto(s)
Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Compuestos de Fenilurea/farmacología , Potasio/metabolismo , Tetrazoles/farmacología , Animales , Western Blotting , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Perros , Electrofisiología , Humanos , Proteínas de Interacción con los Canales Kv/metabolismo , Reacción en Cadena de la Polimerasa , Canales de Potasio Shal/metabolismo , Xenopus laevis
18.
Circ Res ; 103(4): 396-404, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18599870

RESUMEN

Brugada syndrome has been linked to mutations in SCN5A. Agents that dissociate slowly from the sodium channel such as flecainide and ajmaline unmask the Brugada syndrome electrocardiogram and precipitate ventricular tachycardia/fibrillation. Lidocaine, an agent with rapid dissociation kinetics, has previously been shown to exert no effect in patients with Brugada syndrome. We characterized a novel double mutation of SCN5A (V232I in DI-S4+L1308F in DIII-S4) identified in a rare case of lidocaine (1 mg/kg)-induced Brugada syndrome. We studied lidocaine blockade of I(Na) generated by wild-type and V232I+L1308F mutant cardiac sodium channels expressed in mammalian TSA201 cells using patch clamp techniques. Despite no significant difference in steady-state gating parameters between V232I+L1308F and wild-type sodium currents at baseline, use-dependent inhibition of I(Na) by lidocaine was more pronounced in V232I+L1308F versus wild-type (73.0+/-0.1% versus 18.23+/-0.04% at 10 micromol/L measured at 10 Hz, respectively). A dose of 10 micromol/L lidocaine also caused a more negative shift of steady-state inactivation in V232I+L1308F versus wild-type (-14.1+/-0.3 mV and -4.8+/-0.3 mV, respectively). The individual mutations produced a much less accentuated effect. We report the first case of lidocaine-induced Brugada electrocardiogram phenotype. The double mutation in SCN5A, V232I, and L1308F alters the affinity of the cardiac sodium channel for lidocaine such that the drug assumes Class IC characteristics with potent use-dependent block of the sodium channel. Our results demonstrate an additive effect of the 2 missense mutations to sensitize the sodium channel to lidocaine. These findings suggest caution when treating patients carrying such genetic variations with Class I antiarrhythmic drugs.


Asunto(s)
Anestésicos Locales/efectos adversos , Síndrome de Brugada/genética , Lidocaína/efectos adversos , Proteínas Musculares/genética , Mutación Missense/genética , Canales de Sodio/genética , Síndrome de Brugada/fisiopatología , Electrocardiografía , Humanos , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5 , Fenotipo
19.
Pacing Clin Electrophysiol ; 33(3): 274-85, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20025708

RESUMEN

BACKGROUND: Inherited loss of function mutations in SCN5A have been linked to overlapping syndromes including cardiac conduction disease and Brugada syndrome (BrS). The mechanisms responsible for the development of one without the other are poorly understood. METHODS: Direct sequencing was performed in a family with cardiac conduction disease. Wild-type (WT) and mutant channels were expressed in TSA201 cells for electrophysiological study. Green fluorescent protein (GFP)-fused WT or mutant genes were used to assess channel trafficking. RESULTS: A novel SCN5A mutation, P1008S, was identified in all family members displaying first-degree atrioventricular block, but not in unaffected family members nor in 430 reference alleles. Peak P1008S current was 11.77% of WT (P < 0.001). Confocal microscopy showed that WT channels tagged with GFP were localized on the cell surface, whereas GFP-tagged P1008S channels remained trapped in intracellular organelles. Trafficking could be rescued by incubation at room temperature, but not by incubation with mexiletine (300 muM) at 37 degrees C. We also identified a novel polymorphism (D601E) in CACNB2b that slowed inactivation of L-type calcium current (I(Ca,L)), significantly increased total charge. Using the Luo-Rudy action potential (AP) model, we show that the reduction in sodium current (I(Na)) can cause loss of the right ventricular epicardial AP dome in the absence but not in the presence of the slowed inactivation of I(Ca,L). Slowed conduction was present in both cases. CONCLUSIONS: Our results suggest genetic variations leading to a loss-of-function in I(Na) coupled with a gain of function in I(Ca,L) may underlie the development of cardiac conduction disease without BrS.


Asunto(s)
Bradicardia/genética , Canales de Calcio Tipo L/genética , Bloqueo Cardíaco/genética , Sistema de Conducción Cardíaco/fisiopatología , Proteínas Musculares/genética , Mutación , Polimorfismo de Nucleótido Simple , Canales de Sodio/genética , Adolescente , Alelos , Análisis de Varianza , Bradicardia/fisiopatología , Síndrome de Brugada/genética , Síndrome de Brugada/fisiopatología , Técnicas Electrofisiológicas Cardíacas , Femenino , Bloqueo Cardíaco/fisiopatología , Humanos , Masculino , Microscopía Confocal , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5 , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa
20.
Can J Physiol Pharmacol ; 88(12): 1181-90, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21164565

RESUMEN

Long QT syndrome (LQTS) is an inherited disorder characterized by prolonged QT intervals and potentially life-threatening arrhythmias. Mutations in 12 different genes have been associated with LQTS. Here we describe a patient with LQTS who has a mutation in KCNQ1 as well as a polymorphism in KCNH2. The proband (MMRL0362), a 32-year-old female, exhibited multiple ventricular extrasystoles and one syncope. Her ECG (QT interval corrected for heart rate (QTc) = 518ms) showed an LQT2 morphology in leads V4-V6 and LQT1 morphology in leads V1-V2. Genomic DNA was isolated from lymphocytes. All exons and intron borders of 7 LQTS susceptibility genes were amplified and sequenced. Variations were detected predicting a novel missense mutation (V110I) in KCNQ1, as well as a common polymorphism in KCNH2 (K897T). We expressed wild-type (WT) or V110I Kv7.1 channels in CHO-K1 cells cotransfected with KCNE1 and performed patch-clamp analysis. In addition, WT or K897T Kv11.1 were also studied by patch clamp. Current-voltage (I-V) relations for V110I showed a significant reduction in both developing and tail current densities compared with WT at potentials >+20 mV (p < 0.05; n = 8 cells, each group), suggesting a reduction in IKs currents. K897T- Kv11.1 channels displayed a significantly reduced tail current density compared with WT-Kv11.1 at potentials >+10 mV. Interestingly, channel availability assessed using a triple-pulse protocol was slightly greater for K897T compared with WT (V0.5 = -53.1 ± 1.13 mV and -60.7 ± 1.15 mV for K897T and WT, respectively; p < 0.05). Comparison of the fully activated I-V revealed no difference in the rectification properties between WT and K897T channels. We report a patient with a loss-of-function mutation in KCNQ1 and a loss-of-function polymorphism in KCNH2. Our results suggest that a reduction of both IKr and IKs underlies the combined LQT1 and LQT2 phenotype observed in this patient.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Canal de Potasio ERG1 , Electrocardiografía , Femenino , Variación Genética , Humanos , Datos de Secuencia Molecular , Mutación , Fenotipo , Polimorfismo Genético
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA