Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
Sci Rep ; 14(1): 15683, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38977794

RESUMO

We explored physiological effects of the sodium-glucose co-transporter-2 inhibitor empagliflozin on intact experimentally hypertrophic murine hearts following transverse aortic constriction (TAC). Postoperative drug (2-6 weeks) challenge resulted in reduced late Na+ currents, and increased phosphorylated (p-)CaMK-II and Nav1.5 but not total (t)-CaMK-II, and Na+/Ca2+ exchanger expression, confirming previous cardiomyocyte-level reports. It rescued TAC-induced reductions in echocardiographic ejection fraction and fractional shortening, and diastolic anterior and posterior wall thickening. Dual voltage- and Ca2+-optical mapping of Langendorff-perfused hearts demonstrated that empagliflozin rescued TAC-induced increases in action potential durations at 80% recovery (APD80), Ca2+ transient peak signals and durations at 80% recovery (CaTD80), times to peak Ca2+ (TTP100) and Ca2+ decay constants (Decay30-90) during regular 10-Hz stimulation, and Ca2+ transient alternans with shortening cycle length. Isoproterenol shortened APD80 in sham-operated and TAC-only hearts, shortening CaTD80 and Decay30-90 but sparing TTP100 and Ca2+ transient alternans in all groups. All groups showed similar APD80, and TAC-only hearts showed greater CaTD80, heterogeneities following isoproterenol challenge. Empagliflozin abolished or reduced ventricular tachycardia and premature ventricular contractions and associated re-entrant conduction patterns, in isoproterenol-challenged TAC-operated hearts following successive burst pacing episodes. Empagliflozin thus rescues TAC-induced ventricular hypertrophy and systolic functional, Ca2+ homeostatic, and pro-arrhythmogenic changes in intact hearts.


Assuntos
Compostos Benzidrílicos , Cálcio , Glucosídeos , Homeostase , Animais , Compostos Benzidrílicos/farmacologia , Glucosídeos/farmacologia , Camundongos , Cálcio/metabolismo , Homeostase/efeitos dos fármacos , Masculino , Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/tratamento farmacológico , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/cirurgia , Camundongos Endogâmicos C57BL , Isoproterenol/farmacologia , Modelos Animais de Doenças
2.
Int J Toxicol ; 43(4): 357-367, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38477622

RESUMO

In silico modeling offers an opportunity to supplement and accelerate cardiac safety testing. With in silico modeling, computational simulation methods are used to predict electrophysiological interactions and pharmacological effects of novel drugs on critical physiological processes. The O'Hara-Rudy's model was developed to predict the response to different ion channel inhibition levels on cardiac action potential duration (APD) which is known to directly correlate with the QT interval. APD data at 30% 60% and 90% inhibition were derived from the model to delineate possible ventricular arrhythmia scenarios and the marginal contribution of each ion channel to the model. Action potential values were calculated for epicardial, myocardial, and endocardial cells, with action potential curve modeling. This study assessed cardiac ion channel inhibition data combinations to consider when undertaking in silico modeling of proarrhythmic effects as stipulated in the Comprehensive in Vitro Proarrhythmia Assay (CiPA). As expected, our data highlight the importance of the delayed rectifier potassium channel (IKr) as the most impactful channel for APD prolongation. The impact of the transient outward potassium channel (Ito) inhibition on APD was minimal while the inward rectifier (IK1) and slow component of the delayed rectifier potassium channel (IKs) also had limited APD effects. In contrast, the contribution of fast sodium channel (INa) and/or L-type calcium channel (ICa) inhibition resulted in substantial APD alterations supporting the pharmacological relevance of in silico modeling using input from a limited number of cardiac ion channels including IKr, INa, and ICa, at least at an early stage of drug development.


Assuntos
Potenciais de Ação , Simulação por Computador , Canais Iônicos , Miócitos Cardíacos , Potenciais de Ação/efeitos dos fármacos , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia
3.
J Biol Chem ; 300(3): 105759, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38367666

RESUMO

Genome-wide association studies have reported a correlation between a SNP of the RING finger E3 ubiquitin protein ligase rififylin (RFFL) and QT interval variability in humans (Newton-Cheh et al., 2009). Previously, we have shown that RFFL downregulates expression and function of the human-like ether-a-go-go-related gene potassium channel and corresponding rapidly activating delayed rectifier potassium current (IKr) in adult rabbit ventricular cardiomyocytes. Here, we report that RFFL also affects the transient outward current (Ito), but in a peculiar way. RFFL overexpression in adult rabbit ventricular cardiomyocytes significantly decreases the contribution of its fast component (Ito,f) from 35% to 21% and increases the contribution of its slow component (Ito,s) from 65% to 79%. Since Ito,f in rabbits is mainly conducted by Kv4.3, we investigated the effect of RFFL on Kv4.3 expressed in HEK293A cells. We found that RFFL overexpression reduced Kv4.3 expression and corresponding Ito,f in a RING domain-dependent manner in the presence or absence of its accessory subunit Kv channel-interacting protein 2. On the other hand, RFFL overexpression in Kv1.4-expressing HEK cells leads to an increase in both Kv1.4 expression level and Ito,s, similarly in a RING domain-dependent manner. Our physiologically detailed rabbit ventricular myocyte computational model shows that these yin and yang effects of RFFL overexpression on Ito,f, and Ito,s affect phase 1 of the action potential waveform and slightly decrease its duration in addition to suppressing IKr. Thus, RFFL modifies cardiac repolarization reserve via ubiquitination of multiple proteins that differently affect various potassium channels and cardiac action potential duration.


Assuntos
Miócitos Cardíacos , Canais de Potássio Shal , Ubiquitina-Proteína Ligases , Animais , Humanos , Coelhos , Potenciais de Ação/fisiologia , Estudo de Associação Genômica Ampla , Miócitos Cardíacos/metabolismo , Potássio/metabolismo , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Células HEK293
4.
Cardiovasc Res ; 120(5): 490-505, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38261726

RESUMO

AIMS: Short QT syndrome type 3 (SQTS3) is a rare arrhythmogenic disease caused by gain-of-function mutations in KCNJ2, the gene coding the inward rectifier potassium channel Kir2.1. We used a multidisciplinary approach and investigated arrhythmogenic mechanisms in an in-vivo model of de-novo mutation Kir2.1E299V identified in a patient presenting an extremely abbreviated QT interval and paroxysmal atrial fibrillation. METHODS AND RESULTS: We used intravenous adeno-associated virus-mediated gene transfer to generate mouse models, and confirmed cardiac-specific expression of Kir2.1WT or Kir2.1E299V. On ECG, the Kir2.1E299V mouse recapitulated the QT interval shortening and the atrial-specific arrhythmia of the patient. The PR interval was also significantly shorter in Kir2.1E299V mice. Patch-clamping showed extremely abbreviated action potentials in both atrial and ventricular Kir2.1E299V cardiomyocytes due to a lack of inward-going rectification and increased IK1 at voltages positive to -80 mV. Relative to Kir2.1WT, atrial Kir2.1E299V cardiomyocytes had a significantly reduced slope conductance at voltages negative to -80 mV. After confirming a higher proportion of heterotetrameric Kir2.x channels containing Kir2.2 subunits in the atria, in-silico 3D simulations predicted an atrial-specific impairment of polyamine block and reduced pore diameter in the Kir2.1E299V-Kir2.2WT channel. In ventricular cardiomyocytes, the mutation increased excitability by shifting INa activation and inactivation in the hyperpolarizing direction, which protected the ventricle against arrhythmia. Moreover, Purkinje myocytes from Kir2.1E299V mice manifested substantially higher INa density than Kir2.1WT, explaining the abbreviation in the PR interval. CONCLUSION: The first in-vivo mouse model of cardiac-specific SQTS3 recapitulates the electrophysiological phenotype of a patient with the Kir2.1E299V mutation. Kir2.1E299V eliminates rectification in both cardiac chambers but protects against ventricular arrhythmias by increasing excitability in both Purkinje-fiber network and ventricles. Consequently, the predominant arrhythmias are supraventricular likely due to the lack of inward rectification and atrial-specific reduced pore diameter of the Kir2.1E299V-Kir2.2WT heterotetramer.


Assuntos
Fibrilação Atrial , Modelos Animais de Doenças , Miócitos Cardíacos , Canais de Potássio Corretores do Fluxo de Internalização , Animais , Humanos , Camundongos , Potenciais de Ação , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/metabolismo , Fibrilação Atrial/genética , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/metabolismo , Predisposição Genética para Doença , Frequência Cardíaca/genética , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
5.
Acta Pharmacol Sin ; 45(3): 517-530, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37880339

RESUMO

Malignant ventricular arrhythmia (VA) after myocardial infarction (MI) is mainly caused by myocardial electrophysiological remodeling. Brahma-related gene 1 (BRG1) is an ATPase catalytic subunit that belongs to a family of chromatin remodeling complexes called Switch/Sucrose Non-Fermentable Chromatin (SWI/SNF). BRG1 has been reported as a molecular chaperone, interacting with various transcription factors or proteins to regulate transcription in cardiac diseases. In this study, we investigated the potential role of BRG1 in ion channel remodeling and VA after ischemic infarction. Myocardial infarction (MI) mice were established by ligating the left anterior descending (LAD) coronary artery, and electrocardiogram (ECG) was monitored. Epicardial conduction of MI mouse heart was characterized in Langendorff-perfused hearts using epicardial optical voltage mapping. Patch-clamping analysis was conducted in single ventricular cardiomyocytes isolated from the mice. We showed that BRG1 expression in the border zone was progressively increased in the first week following MI. Cardiac-specific deletion of BRG1 by tail vein injection of AAV9-BRG1-shRNA significantly ameliorated susceptibility to electrical-induced VA and shortened QTc intervals in MI mice. BRG1 knockdown significantly enhanced conduction velocity (CV) and reversed the prolonged action potential duration in MI mouse heart. Moreover, BRG1 knockdown improved the decreased densities of Na+ current (INa) and transient outward potassium current (Ito), as well as the expression of Nav1.5 and Kv4.3 in the border zone of MI mouse hearts and in hypoxia-treated neonatal mouse ventricular cardiomyocytes. We revealed that MI increased the binding among BRG1, T-cell factor 4 (TCF4) and ß-catenin, forming a transcription complex, which suppressed the transcription activity of SCN5A and KCND3, thereby influencing the incidence of VA post-MI.


Assuntos
Infarto do Miocárdio , Camundongos , Animais , Infarto do Miocárdio/metabolismo , Arritmias Cardíacas/genética , Miocárdio/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Miócitos Cardíacos/metabolismo
6.
Can J Physiol Pharmacol ; 102(5): 331-341, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38118123

RESUMO

Extracellular potassium concentration might modify electrophysiological properties in the border zone of ischemic myocardium. We evaluated the depolarization and repolarization characteristics across the ischemic-normal border under [K+] variation. Sixty-four-lead epicardial mapping was performed in 26 rats ([K+] 2.3-6.4 mM) in a model of acute ischemia/reperfusion. The animals with [K+] < 4.7 mM (low-normal potassium) had an ischemic zone with ST-segment elevation and activation delay, a border zone with ST-segment elevation and no activation delay, and a normal zone without electrophysiological abnormalities. The animals with [K+] >4.7 mM (normal-high potassium) had only the ischemic and normal zones and no transitional area. Activation-repolarization intervals and local conduction velocities were inversely associated with [K+] in linear regression analysis with adjustment for the zone of myocardium. The reperfusion extrasystolic burden (ESB) was greater in the low-normal as compared to normal-high potassium animals. Ventricular tachycardia/fibrillation incidence did not differ between the groups. In patch-clamp experiments, hypoxia shortened action potential duration at 5.4 mM but not at 1.3 mM of [K+]. IK(ATP) current was lower at 1.3 mM than at 5.4 mM of [K+]. We conclude that the border zone formation in low-normal [K+] was associated with attenuation of IK(ATP) response to hypoxia and increased reperfusion ESB.


Assuntos
Potenciais de Ação , Isquemia Miocárdica , Potássio , Animais , Potássio/sangue , Potássio/metabolismo , Masculino , Ratos , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/sangue , Isquemia Miocárdica/metabolismo , Potenciais de Ação/fisiologia , Traumatismo por Reperfusão Miocárdica/sangue , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Ratos Wistar
8.
JACC Clin Electrophysiol ; 9(12): 2507-2519, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37804259

RESUMO

BACKGROUND: Electrophysiological mapping of ventricular tachycardia (VT) is tedious and poorly reproducible. Substrate analysis on imaging cannot explicitly display VT circuits. OBJECTIVES: This study sought to introduce a computed tomography-based model personalization approach, allowing for the simulation of postinfarction VT in a clinically compatible time frame. METHODS: In 10 patients (age 65 ± 11 years, 9 male) referred for post-VT ablation, computed tomography-derived wall thickness maps were registered to 25 electroanatomical maps (sinus rhythm, paced, and VT). The relationship between wall thickness and electrophysiological characteristics (activation-recovery interval) was analyzed. Wall thickness was then employed to parameterize a fast and tractable organ-scale wave propagation model. Pacing protocols were simulated from multiple sites to test VT induction in silico. In silico VTs were compared to VT circuits mapped clinically. RESULTS: Clinically, 6 different VTs could be induced with detailed maps in 9 patients. The proposed model allowed for fast simulation (median: 6 min/pacing site). Simulations of steady pacing (600 milliseconds) from 100 different sites/patient never triggered any arrhythmia. Applying S1-S2 or S1-S2-S3 induction schemes allowed for the induction of in silico VTs in the 9 of 10 patients who were clinically inducible. The patient who was not inducible clinically was also noninducible in silico. A total of 42 different VTs were simulated (4.2 ± 2 per patient). Six in silico VTs matched a VT circuit mapped clinically. CONCLUSIONS: The proposed framework allows for personalized simulations in a matter of hours. In 6 of 9 patients, simulations show re-entrant patterns matching intracardiac recordings.


Assuntos
Taquicardia Ventricular , Humanos , Masculino , Pessoa de Meia-Idade , Idoso , Taquicardia Ventricular/diagnóstico por imagem , Taquicardia Ventricular/cirurgia , Tomografia Computadorizada por Raios X
9.
J Electrocardiol ; 81: 237-243, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37844373

RESUMO

BACKGROUND: Assessing T-wave symmetry in addition to QT subintervals measurements can provide novel independent data about ventricular repolarization abnormalities linked with arrhythmogenesis. However, the causes of the changes of T-wave symmetry are not completely understood. In silico studies showed that the more symmetrical T-waves were associated with shorter action potential duration (APD) and larger dispersion of ventricular repolarization (DOR). The aim of present simulation was to study the association between T-wave symmetry and action potential (AP) shape. METHODS: ECGs were simulated using a cellular automata model shaped as a ventricular wall segment, and two biophysically-detailed models of ventricular AP - the rabbit and the human. The symmetry ratio (SR) was calculated as a T-wave onset-peak to peak-end area ratio. The individual and combined effects of APD, DOR and AP shape on SR were simulated. To study the effect of AP shape, different APs from triangulated to rectangular were simulated. RESULTS: The simulations showed that AP shape along with APD and DOR contributes much to T-wave symmetry. APs with a flat phase 3 (triangulated) produced asymmetrical T-waves (SR ≥ 1.5) in all simulations, except the shortest APD range. APs with a rapid phase 3 (rectangular) were associated with more symmetrical T-waves (SR ≤ =1) both at the short and the long APDs. CONCLUSION: SR marker in combination with the standard ECG parameters (QT interval, Tpeak-Tend interval) may be useful to identify the proarrhythmic triangulated AP shape.


Assuntos
Eletrocardiografia , Síndrome do QT Longo , Animais , Humanos , Coelhos , Potenciais de Ação/fisiologia , Miocárdio , Arritmias Cardíacas/diagnóstico
10.
Physiol Rep ; 11(11): e15670, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37291479

RESUMO

Ventricular repolarization shows notable sex-specificity, with female sex being associated with longer QT-intervals in electrocardiography irrespective of the species studied. From a clinical point of view, women are at a greater risk for drug-induced torsade de pointes and symptomatic long-QT syndrome. Here, we present an optical mapping (OM) approach to reveal sex-specific action potential (AP) heterogeneity in a slice preparation of mouse hearts. Left ventricular epicardial repolarization in female versus male mice shows longer and, interindividually, more variable AP duration (APD), yielding a less prominent transmural APD gradient. By combining OM with mathematical modeling, we suggest a significant role of IKto,f and IKur in AP broadening in females. Other transmembrane currents, including INaL , only marginally affect basal APD. As in many cardiac pathophysiologies, increasing [Ca2+ ]i poses a risk for arrhythmia, the response of AP morphology to enhanced activation of L-type calcium channels (LTCC) was assessed in a sex-selective manner. Both APD and its variation increased significantly more in female versus male mice after pharmacological LTCC activation, which we hypothesize to be due to sex-specific INaL expression based on mathematical modeling. Altogether, we demonstrate a more delayed repolarization of LV epicardium, a leveled LV transmural APD gradient, and a more pronounced epicardial APD response to Ca2+ influx in females versus males. Mathematical modeling quantifies the relative contributions of selected ionic currents to sex-specific AP morphology under normal and pathophysiological conditions.


Assuntos
Eletrocardiografia , Ventrículos do Coração , Feminino , Masculino , Animais , Camundongos , Ventrículos do Coração/metabolismo , Arritmias Cardíacas/metabolismo , Pericárdio , Potenciais de Ação
11.
Heliyon ; 9(3): e14440, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36967904

RESUMO

The action potential forms the basis of cardiac pacemaking, conduction, and contraction. Action potentials can be recorded from numerous preparation types, including ventricular or atrial trabecules, Purkinje fibers, isolated cardiac myocytes. Numerous techniques are also available as well, such as the conventional microelectrode and the single-cell current clamp techniques, optical mapping, or in silico modeling. With such a vast array of electrophysiological methods comes an array of available hardware and software solutions. In this work, we present a software with an intuitive graphical user interface, ActionPytential, that enables the analysis of any type of cardiac action potential, regardless of acquisition method or tissue type. In most available software tools, the analysis of continuous (gap-free) recordings often requires manual user interaction to segment the individual action potentials. We provide an automated solution for this, both for slow-response and for externally paced action potentials. As of now, ActionPytential calculates 34 parameters from each action potential. The most often utilized ones, including amplitude, maximal rate of depolarization, and action potential duration values, were validated on 1200 action potentials from human, dog, rabbit, guinea pig, and rat cardiac preparations. We also provide new parameters that were previously only measurable manually, including the position and the depth of the notch in potentials showing a spike-and-dome morphology. Further notable features include a Butterworth-type low-pass filter, the averaging of multiple potentials, automated corrections for baseline drifting, aided manual analysis, high-quality plots, and batch processing for any number of potentials. ActionPytential is available for all major platforms (Windows, MacOS, GNU + Linux, BSD).

12.
J Mol Cell Cardiol ; 177: 50-61, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36898499

RESUMO

Genetic testing for inherited arrhythmias and discriminating pathogenic or benign variants from variants of unknown significance (VUS) is essential for gene-based medicine. KCNQ1 is a causative gene of type 1 long QT syndrome (LQTS), and approximately 30% of the variants found in type 1 LQTS are classified as VUS. We studied the role of zebrafish cardiac arrhythmia model in determining the clinical significance of KCNQ1 variants. We generated homozygous kcnq1 deletion zebrafish (kcnq1del/del) using the CRISPR/Cas9 and expressed human Kv7.1/MinK channels in kcnq1del/del embryos. We dissected the hearts from the thorax at 48 h post-fertilization and measured the transmembrane potential of the ventricle in the zebrafish heart. Action potential duration was calculated as the time interval between peak maximum upstroke velocity and 90% repolarization (APD90). The APD90 of kcnq1del/del embryos was 280 ± 47 ms, which was significantly shortened by injecting KCNQ1 wild-type (WT) cRNA and KCNE1 cRNA (168 ± 26 ms, P < 0.01 vs. kcnq1del/del). A study of two pathogenic variants (S277L and T587M) and one VUS (R451Q) associated with clinically definite LQTS showed that the APD90 of kcnq1del/del embryos with these mutant Kv7.1/MinK channels was significantly longer than that of Kv7.1 WT/MinK channels. Given the functional results of the zebrafish model, R451Q could be reevaluated physiologically from VUS to likely pathogenic. In conclusion, functional analysis using in vivo zebrafish cardiac arrhythmia model can be useful for determining the pathogenicity of loss-of-function variants in patients with LQTS.


Assuntos
Síndrome do QT Longo , Peixe-Zebra , Animais , Humanos , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/genética , Mutação , RNA Complementar , Virulência , Peixe-Zebra/genética
13.
J Physiol ; 601(8): 1353-1370, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36866700

RESUMO

Optical mapping is a widely used tool to record and visualize the electrophysiological properties in a variety of myocardial preparations such as Langendorff-perfused isolated hearts, coronary-perfused wedge preparations, and cell culture monolayers. Motion artifact originating from the mechanical contraction of the myocardium creates a significant challenge to performing optical mapping of contracting hearts. Hence, to minimize the motion artifact, cardiac optical mapping studies are mostly performed on non-contracting hearts, where the mechanical contraction is removed using pharmacological excitation-contraction uncouplers. However, such experimental preparations eliminate the possibility of electromechanical interaction, and effects such as mechano-electric feedback cannot be studied. Recent developments in computer vision algorithms and ratiometric techniques have opened the possibility of performing optical mapping studies on isolated contracting hearts. In this review, we discuss the existing techniques and challenges of optical mapping of contracting hearts.


Assuntos
Coração , Miocárdio , Potenciais de Ação/fisiologia , Coração/diagnóstico por imagem , Coração/fisiologia
14.
J Cardiovasc Dev Dis ; 10(2)2023 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-36826585

RESUMO

Wilson assumed that the ventricular gradient (VG) is independent of the ventricular activation order. This paradigm has often been refuted and was never convincingly corroborated. We sought to validate Wilson's concept by intra-individual comparison of the VG of sinus beats and ectopic beats, thus assessing the effects of both altered ventricular conduction (caused by the ectopic focus) and restitution (caused by ectopic prematurity). We studied standard diagnostic ECGs of 118 patients with accidental extrasystoles: normally conducted supraventricular ectopic beats (SN, N = 6) and aberrantly conducted supraventricular ectopic beats (SA, N = 20) or ventricular ectopic beats (V, N = 92). In each patient, we computed the VG vectors of the predominant beat, VGp→, of the ectopic beat, VGe→, and of the VG difference vector, ΔVGep→, and compared their sizes. VGe→ of the SA and V ectopic beats were significantly larger than VGp→ (53.7 ± 25.0 vs. 47.8 ± 24.6 mV∙ms, respectively; p < 0.001). ΔVGep→ were three times larger than the difference of VGe→ and VGp→ (19.94 ± 9.76 vs. 5.94 mV∙ms, respectively), demonstrating differences in the VGp→ and VGe→ spatial directions. The amount of ectopic prematurity was not correlated with ΔVGep→, although the larger VG difference vectors were observed for the more premature (<80%) extrasystoles. Electrical restitution properties and electrotonic interactions likely explain our findings. We conclude that the concept of a conduction-independent VG should be tested at equal heart rates and without including premature extrasystoles.

15.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36835338

RESUMO

Mechanisms for the α-adrenoceptor-mediated positive inotropy in neonatal mouse ventricular myocardium were studied with isolated myocardial preparations. The phenylephrine-induced positive inotropy was suppressed by prazosin, nifedipine, and chelerythrine, a protein kinase C inhibitor, but not by SEA0400, a selective Na+/Ca2+ exchanger inhibitor. Phenylephrine increased the L-type Ca2+ channel current and prolonged the action potential duration, while the voltage-dependent K+ channel current was not influenced. In the presence of cromakalim, an ATP-sensitive K+ channel opener, the phenylephrine-induced prolongation of action potential duration, as well as the positive inotropy, were smaller than in the absence of cromakalim. These results suggest that the α-adrenoceptor-mediated positive inotropy is mediated by an increase in Ca2+ influx through the L-type Ca2+ channel, and the concomitant increase in action potential duration acts as an enhancing factor.


Assuntos
Contração Miocárdica , Miocárdio , Camundongos , Animais , Potenciais de Ação , Cromakalim/farmacologia , Contração Miocárdica/fisiologia , Fenilefrina/farmacologia , Receptores Adrenérgicos
16.
Heart Rhythm ; 20(5): 744-753, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36804540

RESUMO

BACKGROUND: Therapeutic hypothermia (TH) increases the susceptibility to ventricular arrhythmias (VAs) by prolonging action potential duration (APD) and facilitating arrhythmogenic spatially discordant alternans (SDA). Levosimendan, a calcium sensitizer, has been reported to shorten APD by enhancing the adenosine triphosphate (ATP)-sensitive K current. OBJECTIVE: The purpose of this study was to test the hypothesis that, during TH, levosimendan shortens the already prolonged APD, attenuates SDA, and prevents VA. METHODS: Langendorff-perfused isolated rabbit hearts were subjected to TH (30°C) for 15 minutes, followed by treatment with either levosimendan 0.5 µM (n = 9) or vehicle (n = 8) for an additional 30 minutes under TH. Using an optical mapping system, epicardial APD was evaluated by S1 pacing. SDA threshold was defined as the longest pacing cycle length (PCL) that induces the phenomenon of SDA. Ventricular fibrillation (VF) inducibility was evaluated by burst pacing for 30 seconds at the shortest PCL that achieved 1:1 ventricular capture. RESULTS: During TH, levosimendan shortened ventricular APD (PCL 400 ms; from 259 ± 8 ms to 241 ± 18 ms; P = .036) and decreased SDA threshold (from 327 ± 88 ms to 311 ± 68 ms; P = .011). VF inducibility was lowered from 39% ± 30% to 14% ± 12% with levosimendan (P = .018), whereas APD at PCL 400 ms (P = .161), SDA threshold (P = 1), and VF inducibility (P = .173) were not changed by vehicle. CONCLUSION: During TH, levosimendan could protect hearts against VA by shortening APD and decreasing SDA threshold. Enhancing ATP-sensitive K current with levosimendan might be a novel approach to preventing VA during TH.


Assuntos
Arritmias Cardíacas , Hipotermia Induzida , Animais , Coelhos , Simendana , Coração , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/prevenção & controle , Potenciais de Ação/fisiologia
17.
Math Med Biol ; 40(2): 175-198, 2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-36689769

RESUMO

Current understanding of arrhythmia mechanisms and design of anti-arrhythmic drug therapies hinges on the assumption that myocytes from the same region of a single heart have similar, if not identical, action potential waveforms and drug responses. On the contrary, recent experiments reveal significant heterogeneity in uncoupled healthy myocytes both from different hearts as well as from identical regions within a single heart. In this work, a methodology is developed for quantifying the individual electrophysiological properties of large numbers of uncoupled cardiomyocytes under ion channel block in terms of the parameters values of a conceptual fast-slow model of electrical excitability. The approach is applied to a population of nearly 500 rabbit ventricular myocytes for which action potential duration (APD) before and after the application of the drug nifedipine was experimentally measured (Lachaud et al., 2022, Cardiovasc. Res.). To this end, drug action is represented by a multiplicative factor to an effective ion conductance, a closed form asymptotic expression for APD is derived and inverted to determine model parameters as functions of APD and $\varDelta $APD (drug-induced change in APD) for each myocyte. Two free protocol-related quantities are calibrated to experiment using an adaptive-domain procedure based on an original assumption of optimal excitability. The explicit APD expression and the resulting set of model parameter values allow (a) direct evaluation of conditions necessary to maintain fixed APD or $\varDelta $APD, (b) predictions of the proportion of cells remaining excitable after drug application, (c) predictions of stimulus period dependency and (d) predictions of dose-response curves, the latter being in agreement with additional experimental data.


Assuntos
Arritmias Cardíacas , Miócitos Cardíacos , Animais , Coelhos , Miócitos Cardíacos/fisiologia , Arritmias Cardíacas/tratamento farmacológico , Potenciais de Ação/fisiologia , Canais Iônicos , Fenômenos Eletrofisiológicos , Ventrículos do Coração
18.
Am J Physiol Heart Circ Physiol ; 324(2): H179-H197, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36487185

RESUMO

Many cardiac diseases are characterized by an increased late sodium current, including heart failure, hypertrophic cardiomyopathy, and inherited long QT syndrome type 3 (LQT3). The late sodium current in LQT3 is caused by a gain-of-function mutation in the voltage-gated sodium channel Nav1.5. Despite a well-defined genetic cause of LQT3, treatment remains inconsistent because of incomplete penetrance of the mutation and variability of antiarrhythmic efficacy. Here, we investigate the relationship between LQT3-associated mutation incomplete penetrance and variability in ion channel expression, simulating a population of 1,000 individuals with the O'Hara-Rudy model of the human ventricular myocyte. We first simulate healthy electrical activity (i.e., in the absence of a mutation) and then incorporate heterozygous expression for three LQT3-associated mutations (Y1795C, I1768V, and ΔKPQ), to directly compare the effects of each mutation on individuals across a diverse population. For all mutations, we find that susceptibility, defined by either the presence of an early afterdepolarization (EAD) or prolonged action potential duration (APD), primarily depends on the balance between the conductance of IKr and INa, for which individuals with a higher IKr-to-INa ratio are less susceptible. Furthermore, we find distinct differences across the population, observing individuals susceptible to zero, one, two, or all three mutations. Individuals tend to be less susceptible with an appropriate balance of repolarizing currents, typically via increased IKs or IK1. Interestingly, the more critical repolarizing current is mutation specific. We conclude that the balance between key currents plays a significant role in mutant-specific presentation of the disease phenotype in LQT3.NEW & NOTEWORTHY An in silico population approach investigates the relationship between variability in ion channel expression and gain-of-function mutations in the voltage-gated sodium channel associated with the congenital disorder long QT syndrome type 3 (LQT3). We find that ion channel variability can contribute to incomplete penetrance of the mutation, with mutant-specific differences in ion channel conductances leading to susceptibility to proarrhythmic action potential duration prolongation or early afterdepolarizations.


Assuntos
Síndrome do QT Longo , Humanos , Potenciais de Ação , Canais Iônicos/genética , Síndrome do QT Longo/genética , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Penetrância , Sódio/metabolismo , Simulação por Computador
19.
JACC Basic Transl Sci ; 7(10): 1001-1017, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36337924

RESUMO

Glycogen synthase kinase 3 (GSK-3) inhibition has emerged as a potential therapeutic target for several diseases, including cancer. However, the role for GSK-3 regulation of human cardiac electrophysiology remains ill-defined. We demonstrate that SB216763, a GSK-3 inhibitor, can acutely reduce conduction velocity in human cardiac slices. Combined computational modeling and experimental approaches provided mechanistic insight into GSK-3 inhibition-mediated changes, revealing that decreased sodium-channel conductance and tissue conductivity may underlie the observed phenotypes. Our study demonstrates that GSK-3 inhibition in human myocardium alters electrophysiology and may predispose to an arrhythmogenic substrate; therefore, monitoring for adverse arrhythmogenic events could be considered.

20.
Toxicol Appl Pharmacol ; 457: 116312, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36343672

RESUMO

Drug-induced torsades de pointes (TdP) risks are responsible for the withdrawal of many drugs from the market. Nowadays, assessments of drug-induced TdP risks are mainly based on maximum effective free therapeutic plasma concentration (EFTPCmax) and cardiac ionic current inhibitions using the human ventricular myocytes model (Tor-ORd model). Myocytes are targets of drug-induced TdP. The TdP risks may be directly linked to myocyte drug concentrations. We aimed to develop a semi-physiologically based pharmacokinetic (Semi-PBPK) model linked to cardiac ionic current inhibition (pharmacodynamics, PD) (Semi-PBPK-PD) to simultaneously predict myocyte drug concentrations and their TdP risks in humans. Alterations in action potential duration (ΔAPD90) were simulated using the Tor-ORd model and ionic current inhibition parameters based on myocyte or plasma drug concentrations. The predicted ΔAPD90 values were translated into in vivo alterations in QT interval(ΔQTc) induced by moxifloxacin, dofetilide, or sotalol. Myocyte drug concentrations of moxifloxacin, dofetilide, and sotalol gave better predictions of ΔQTc than plasma. Following validating the developed semi-PBPK-PD model, TdP risks of 37 drugs were assessed using ΔAPD90 and early afterdepolarization occurrence, which were estimated based on 10 × EFTPCmax and 10 × EFTMCmax (maximum effective free therapeutic myocyte concentration). 10 × EFTMCmax gave more sensitive and accurate predictions of pro-arrhythmic cardiotoxicity and the predicted TdP risks were also closer to clinic practice than 10 × EFTPCmax. In conclusion, pharmacokinetics and TdP risks of 37 drugs were successfully predicted using the semi-PBPK-PD model. Myocyte drug concentrations gave better predictions of ΔQTc and TdP risks than plasma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...