Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Nature ; 483(7387): 96-9, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22367544

RESUMO

Sudden cardiac death exhibits diurnal variation in both acquired and hereditary forms of heart disease, but the molecular basis of this variation is unknown. A common mechanism that underlies susceptibility to ventricular arrhythmias is abnormalities in the duration (for example, short or long QT syndromes and heart failure) or pattern (for example, Brugada's syndrome) of myocardial repolarization. Here we provide molecular evidence that links circadian rhythms to vulnerability in ventricular arrhythmias in mice. Specifically, we show that cardiac ion-channel expression and QT-interval duration (an index of myocardial repolarization) exhibit endogenous circadian rhythmicity under the control of a clock-dependent oscillator, krüppel-like factor 15 (Klf15). Klf15 transcriptionally controls rhythmic expression of Kv channel-interacting protein 2 (KChIP2), a critical subunit required for generating the transient outward potassium current. Deficiency or excess of Klf15 causes loss of rhythmic QT variation, abnormal repolarization and enhanced susceptibility to ventricular arrhythmias. These findings identify circadian transcription of ion channels as a mechanism for cardiac arrhythmogenesis.


Assuntos
Arritmias Cardíacas/fisiopatologia , Ritmo Circadiano/fisiologia , Sistema de Condução Cardíaco/fisiologia , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/genética , Células Cultivadas , Ritmo Circadiano/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Morte Súbita Cardíaca/etiologia , Eletrocardiografia , Regulação da Expressão Gênica , Frequência Cardíaca/fisiologia , Ventrículos do Coração/citologia , Fatores de Transcrição Kruppel-Like , Proteínas Interatuantes com Canais de Kv/biossíntese , Proteínas Interatuantes com Canais de Kv/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Musculares/citologia , Regiões Promotoras Genéticas/genética , Ratos , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
J Cardiovasc Electrophysiol ; 27(1): 110-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26459193

RESUMO

Connexin43 (Cx43) phosphorylation alters gap junction localization and function. In particular, phosphorylation at serine-368 (S368) has been suggested to alter gap junctional conductance, but previous reports have shown inconsistent results for both timing and functional effects of S368 phosphorylation. The objective of this study was to determine the functional effects of isolated S368 phosphorylation. We evaluated wild-type Cx43 (AdCx43) and mutations simulating permanent phosphorylation (Ad368E) or preventing phosphorylation (Ad368A) at S368. Function was assessed by optical mapping of electrical conduction in patterned cultures of neonatal rat ventricular myocytes, under baseline and metabolic stress (MS) conditions. Baseline conduction velocity (CV) was similar for all groups. In the AdCx43 and Ad368E groups, MS moderately decreased CV. Ad368A caused complete conduction block during MS. Triton-X solubility assessment showed no change in Cx43 location during conduction impairment. Western blot analysis showed that Cx43-S368 phosphorylation was present at baseline, and that it decreased during MS. Our data indicate that phosphorylation at S368 does not affect CV under baseline conditions, and that preventing S368 phosphorylation makes Cx43 hypersensitive to MS. These results show the critical role of S368 phosphorylation during stress conditions.


Assuntos
Conexina 43/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Fisiológico , Potenciais de Ação , Animais , Animais Recém-Nascidos , Células Cultivadas , Conexina 43/genética , Mutação , Fosforilação , Ratos Sprague-Dawley , Serina , Transdução de Sinais , Fatores de Tempo , Transfecção , Imagens com Corantes Sensíveis à Voltagem
3.
Proc Natl Acad Sci U S A ; 109(44): 18186-91, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23071315

RESUMO

Nitric oxide (NO) derived from the activity of neuronal nitric oxide synthase (NOS1) is involved in S-nitrosylation of key sarcoplasmic reticulum (SR) Ca(2+) handling proteins. Deficient S-nitrosylation of the cardiac ryanodine receptor (RyR2) has a variable effect on SR Ca(2+) leak/sparks in isolated myocytes, likely dependent on the underlying physiological state. It remains unknown, however, whether such molecular aberrancies are causally related to arrhythmogenesis in the intact heart. Here we show in the intact heart, reduced NOS1 activity increased Ca(2+)-mediated ventricular arrhythmias only in the setting of elevated myocardial [Ca(2+)](i). These arrhythmias arose from increased spontaneous SR Ca(2+) release, resulting from a combination of decreased RyR2 S-nitrosylation (RyR2-SNO) and increased RyR2 oxidation (RyR-SOx) (i.e., increased reactive oxygen species (ROS) from xanthine oxidoreductase activity) and could be suppressed with xanthine oxidoreductase (XOR) inhibition (i.e., allopurinol) or nitric oxide donors (i.e., S-nitrosoglutathione, GSNO). Surprisingly, we found evidence of NOS1 down-regulation of RyR2 phosphorylation at the Ca(2+)/calmodulin-dependent protein kinase (CaMKII) site (S2814), suggesting molecular cross-talk between nitrosylation and phosphorylation of RyR2. Finally, we show that nitroso-redox imbalance due to decreased NOS1 activity sensitizes RyR2 to a severe arrhythmic phenotype by oxidative stress. Our findings suggest that nitroso-redox imbalance is an important mechanism of ventricular arrhythmias in the intact heart under disease conditions (i.e., elevated [Ca(2+)](i) and oxidative stress), and that therapies restoring nitroso-redox balance in the heart could prevent sudden arrhythmic death.


Assuntos
Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , Compostos Nitrosos/metabolismo , Animais , Cobaias , Miocárdio/metabolismo , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Estresse Oxidativo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
4.
Circ Res ; 110(3): 465-70, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22158709

RESUMO

RATIONALE: Abnormal calcium release from sarcoplasmic reticulum (SR) is considered an important trigger of atrial fibrillation (AF). Whereas increased Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activity has been proposed to contribute to SR leak and AF induction, downstream targets of CaMKII remain controversial. OBJECTIVE: To test the hypothesis that inhibition of CaMKII-phosphorylated type-2 ryanodine receptors (RyR2) prevents AF initiation in FKBP12.6-deficient (-/-) mice. METHODS AND RESULTS: Mice lacking RyR2-stabilizing subunit FKBP12.6 had a higher incidence of spontaneous and pacing-induced AF compared with wild-type mice. Atrial myocytes from FKBP12.6-/- mice exhibited spontaneous Ca(2+) waves (SCaWs) leading to Na(+)/Ca(2+)-exchanger activation and delayed afterdepolarizations (DADs). Mutation S2814A in RyR2, which inhibits CaMKII phosphorylation, reduced Ca(2+) spark frequency, SR Ca(2+) leak, and DADs in atrial myocytes from FKBP12.6-/-:S2814A mice compared with FKBP12.6-/- mice. Moreover, FKBP12.6-/-:S2814A mice exhibited a reduced susceptibility to inducible AF, whereas FKBP12.6-/-:S2808A mice were not protected from AF. CONCLUSIONS: FKBP12.6 mice exhibit AF caused by SR Ca(2+) leak, Na(+)/Ca(2+)-exchanger activation, and DADs, which promote triggered activity. Genetic inhibition of RyR2-S2814 phosphorylation prevents AF induction in FKBP12.6-/- mice by suppressing SR Ca(2+) leak and DADs. These results suggest suppression of RyR2-S2814 phosphorylation as a potential anti-AF therapeutic target.


Assuntos
Fibrilação Atrial/prevenção & controle , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Proteínas de Ligação a Tacrolimo/deficiência , Proteínas de Ligação a Tacrolimo/genética , Animais , Fibrilação Atrial/metabolismo , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Átrios do Coração/citologia , Camundongos , Camundongos Knockout , Modelos Animais , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Retículo Sarcoplasmático/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo
5.
J Emerg Med ; 46(1): 1-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24063875

RESUMO

BACKGROUND: Cardiac syncope is associated with poor outcomes and may result in traumatic injuries. In patients presenting to the emergency department (ED) with trauma, recognizing the cause of syncope is particularly challenging. Also, clinical markers to identify cardiac syncope are not well established. STUDY OBJECTIVES: We sought to evaluate clinical markers that could identify cardiac syncope in patients with traumatic falls derived from a large urban trauma database. METHODS: All patients presenting to the ED during a 10-year study period with a traumatic fall were identified retrospectively. The subset of patients with syncope was ascertained by chart review and defined as cardiac syncope (e.g., presence of dysrhythmia, valvular abnormality), non-cardiac syncope (e.g., vasovagal, neurological), or syncope of unknown cause. RESULTS: Of the 5420 patients with traumatic falls, 180 (3.3%) patients with syncope were identified. Among the 180 patients with syncope, the cause was identified as cardiac in 24 (13%), noncardiac in 58 (32%), and unknown in 98 (54%). Three independent predictors (i.e., risk factors) of cardiac syncope were identified: age >65 years, presence of coronary artery disease, and pathological Q waves. Presence of at least one risk factor accurately predicted cardiac syncope in this population, with a sensitivity of 100%, a specificity of 43%, and a negative predictive value of 100% (area under the receiver operating characteristic curve: 0.80 ± 0.04). CONCLUSION: In patients with traumatic falls and syncope, simple clinical and electrocardiographical variables may identify patients with cardiac causes of syncope. Proper identification of cardiac syncope in this population can potentially prevent recurrence of life-threatening traumatic injury.


Assuntos
Acidentes por Quedas , Síncope/diagnóstico , Síncope/etiologia , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Área Sob a Curva , Arritmias Cardíacas/complicações , Arritmias Cardíacas/diagnóstico , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/diagnóstico , Eletromiografia , Serviço Hospitalar de Emergência , Feminino , Doenças das Valvas Cardíacas/complicações , Doenças das Valvas Cardíacas/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Curva ROC , Estudos Retrospectivos , Fatores de Risco , Ferimentos e Lesões/etiologia
6.
Circulation ; 126(17): 2095-104, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23019291

RESUMO

BACKGROUND: Recently, we reported that sarcoplasmic reticulum Ca(2+) ATPase 2a (SERCA2a), the pump responsible for reuptake of cytosolic calcium during diastole, plays a central role in the molecular mechanism of cardiac alternans. Heart failure (HF) is associated with impaired myocardial calcium handling, deficient SERCA2a, and increased susceptibility to cardiac alternans. Therefore, we hypothesized that restoring deficient SERCA2a by gene transfer will significantly reduce arrhythmogenic cardiac alternans in the failing heart. METHODS AND RESULTS: Adult guinea pigs were divided into 3 groups: control, HF, and HF+AAV9.SERCA2a gene transfer. HF resulted in a decrease in left ventricular fractional shortening compared with controls (P<0.001). As expected, isolated HF myocytes demonstrated slower sarcoplasmic reticulum calcium uptake, decreased Ca(2+) release, and increased diastolic Ca(2+) (P<0.05) compared with controls. Moreover, SERCA2a, cardiac ryanodine receptor 2, and sodium-calcium exchanger protein expression was decreased in HF compared with control (P<0.05). As predicted, HF increased susceptibility to cardiac alternans, as evidenced by decreased heart rate thresholds for both V(m) alternans and Ca alternans compared with controls (P<0.01). Interestingly, in vivo gene transfer of AAV9.SERCA2a in the failing heart improved left ventricular contractile function (P<0.01), suppressed cardiac alternans (P<0.01), and reduced ryanodine receptor 2 P(o) secondary to reduction of ryanodine receptor 2-P(S2814) (P<0.01). This ultimately resulted in a decreased incidence of inducible ventricular arrhythmias (P=0.05). CONCLUSIONS: These data show that SERCA2a gene transfer in the failing heart not only improves contractile function but also directly restores electric stability through the amelioration of key arrhythmogenic substrate (ie, cardiac alternans) and triggers (ie, sarcoplasmic reticulum Ca(2+) leak).


Assuntos
Marcação de Genes/métodos , Técnicas de Transferência de Genes , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/terapia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/genética , Animais , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/terapia , Cobaias , Insuficiência Cardíaca/enzimologia , Masculino , Miócitos Cardíacos/enzimologia , Retículo Sarcoplasmático/enzimologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/administração & dosagem
7.
Circulation ; 125(2): 216-25, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22158756

RESUMO

BACKGROUND: Several lines of evidence have suggested that maintenance of atrial fibrillation (AF) depends on reentrant mechanisms. Maintenance of reentry necessitates a sufficiently short refractory period and/or delayed conduction, and AF has been associated with both alterations. Fibrosis, cellular dysfunction, and gap junction protein alterations occur in AF and cause conduction delay. We performed this study to test the hypothesis that gap junction protein overexpression would improve conduction and prevent AF. METHODS AND RESULTS: Thirty Yorkshire swine were randomized into 2 groups (sinus rhythm and AF), and each group into 3 subgroups: sham-operated control, gene therapy with adenovirus expressing connexin (Cx) 40, and gene therapy with adenovirus expressing Cx43 (n=5 per subgroup). All animals had epicardial gene painting; the AF group had burst atrial pacing. All animals underwent terminal study 7 days after gene transfer. Sinus rhythm animals had strong transgene expression but no atrial conduction changes. In AF animals, controls had reduced and lateralized Cx43 expression, and Cx43 gene transfer restored expression and cellular location to sinus rhythm control levels. In the AF group, both Cx40 and Cx43 gene transfer improved conduction and reduced AF relative to controls. CONCLUSIONS: Connexin gene therapy preserved atrial conduction and prevented AF.


Assuntos
Fibrilação Atrial/prevenção & controle , Conexina 43/fisiologia , Conexinas/fisiologia , Sistema de Condução Cardíaco , Animais , Fibrilação Atrial/terapia , Estimulação Cardíaca Artificial , Conexina 43/genética , Conexinas/genética , Técnicas de Transferência de Genes , Terapia Genética , Suínos , Resultado do Tratamento , Proteína alfa-5 de Junções Comunicantes
8.
Am J Physiol Heart Circ Physiol ; 305(3): H410-9, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23709598

RESUMO

Emerging evidence suggests that ventricular electrical remodeling (VER) is triggered by regional myocardial strain via mechanoelectrical feedback mechanisms; however, the ionic mechanisms underlying strain-induced VER are poorly understood. To determine its ionic basis, VER induced by altered electrical activation in dogs undergoing left ventricular pacing (n = 6) were compared with unpaced controls (n = 4). Action potential (AP) durations (APDs), ionic currents, and Ca(2+) transients were measured from canine epicardial myocytes isolated from early-activated (low strain) and late-activated (high strain) left ventricular regions. VER in the early-activated region was characterized by minimal APD prolongation, but marked attenuation of the AP phase 1 notch attributed to reduced transient outward K(+) current. In contrast, VER in the late-activated region was characterized by significant APD prolongation. Despite marked APD prolongation, there was surprisingly minimal change in ion channel densities but a twofold increase in diastolic Ca(2+). Computer simulations demonstrated that changes in sarcolemmal ion channel density could only account for attenuation of the AP notch observed in the early-activated region but failed to account for APD remodeling in the late-activated region. Furthermore, these simulations identified that cytosolic Ca(2+) accounted for APD prolongation in the late-activated region by enhancing forward-mode Na(+)/Ca(2+) exchanger activity, corroborated by increased Na(+)/Ca(2+) exchanger protein expression. Finally, assessment of skinned fibers after VER identified altered myofilament Ca(2+) sensitivity in late-activated regions to be associated with increased diastolic levels of Ca(2+). In conclusion, we identified two distinct ionic mechanisms that underlie VER: 1) strain-independent changes in early-activated regions due to remodeling of sarcolemmal ion channels with no changes in Ca(2+) handling and 2) a novel and unexpected mechanism for strain-induced VER in late-activated regions in the canine arising from remodeling of sarcomeric Ca(2+) handling rather than sarcolemmal ion channels.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio , Cálcio/metabolismo , Sistema de Condução Cardíaco/metabolismo , Ventrículos do Coração/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Potenciais de Ação , Animais , Estimulação Cardíaca Artificial , Simulação por Computador , Cães , Cinética , Masculino , Modelos Cardiovasculares , Potássio/metabolismo , Canais de Potássio/metabolismo , Sarcolema/metabolismo
9.
Crit Care Med ; 40(11): 2954-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22890250

RESUMO

OBJECTIVES: Although the majority of sudden cardiac arrests occur in patients with ischemic heart disease, the effect of therapeutic hypothermia on arrhythmia susceptibility during acute global ischemia is not well understood. While both ischemia and severe hypothermia are arrhythmogenic, patients undergoing therapeutic hypothermia do not have an increase in arrhythmias, despite the fact that most sudden cardiac arrest occur in the setting of ischemia. We hypothesized that mild hypothermia induced prior to myocardial ischemia and reperfusion will have a beneficial effect on ischemia-related arrhythmia substrates. DESIGN: We developed a model of global ischemia and reperfusion in the canine wedge preparation to study the transmural electrophysiologic effects of ischemia at different temperatures. SETTING: Animal study. SUBJECTS: Male mongrel dogs. INTERVENTIONS: Canine left ventricle wedge preparations at 1) control (36°C) or 2) mild hypothermia, to simulate temperatures used in therapeutic hypothermia (32°C), were subjected to 15 mins of no-flow ischemia and subsequently reperfused. MEASUREMENTS AND MAIN RESULTS: Optical action potentials were recorded spanning the transmural wall of left ventricle. Action potential duration for epicardial, mid-myocardial, and epicardial cells was measured. Transmural dispersion of repolarization and conduction velocity were measured at baseline, during ischemia, and during reperfusion. No difference was seen at baseline for conduction velocity or dispersion of repolarization between groups. Conduction velocity decreased from 0.46 ± 0.02 m/sec to 0.23 ± 0.07 m/sec, and dispersion of repolarization increased from 30 ± 5 msecs to 57 ± 4 msecs in the control group at 15 mins of ischemia. Mild hypothermia attenuated both the ischemia-induced conduction velocity slowing (decreasing from 0.44 ± 0.02 m/sec to 0.35 ± 0.03 m/sec; p = .019) and the ischemia-induced increase in dispersion of repolarization (25 ± 3 msecs to 37 ± 7 msecs; p = .037). Epicardial conduction block was observed in six of seven preparations of the control group, but no preparations in the mild hypothermia group developed conduction block (0/6). CONCLUSIONS: Mild hypothermia attenuated ischemia-induced increase in dispersion of repolarization, conduction slowing, and block, which are known mechanisms of arrhythmogenesis in ischemia. These data suggest that therapeutic hypothermia may decrease arrhythmogenesis during myocardial ischemia.


Assuntos
Arritmias Cardíacas/prevenção & controle , Hipotermia Induzida , Modelos Animais , Isquemia Miocárdica/complicações , Animais , Cães , Masculino , Reperfusão Miocárdica , Estados Unidos , Imagens com Corantes Sensíveis à Voltagem
10.
J Cardiovasc Pharmacol ; 60(2): 165-71, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22561361

RESUMO

Torsade de Pointes (TdP) proarrhythmia is a major complication of therapeutic drugs that block the delayed rectifier current. QT interval prolongation, the principal marker used to screen drugs for proarrhythmia, is both insensitive and nonspecific. Consequently, better screening methods are needed. Drug-induced transmural dispersion of repolarization (TDR) is mechanistically linked to TdP. Therefore, we hypothesized that drug-induced enhancement of TDR is more predictive of proarrhythmia than QT interval. High-resolution transmural optical action potential mapping was performed in canine wedge preparations (n = 19) at baseline and after perfusion with 4 different QT prolonging drugs at clinically relevant concentrations. Two proarrhythmic drugs in patients (bepridil and E4031) were compared with 2 nonproarrhythmic drugs (risperidone and verapamil). Both groups prolonged the QT (all P < 0.02), least with the proarrhythmic drug bepridil, reaffirming that QT is a poor predictor of TdP. In contrast, TDR was enhanced only by proarrhythmic drugs (P < 0.03). Increased TDR was due to a preferential prolongation of midmyocardial cell, relative to epicardial cell, APD, whereas nonproarrhythmic drugs similarly prolonged both cell types. In contrast to QT prolongation, augmentation of TDR was induced by proarrhythmic but not nonproarrhythmic drugs, suggesting TDR is a superior preclinical marker of proarrhythmic risk during drug development.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Bepridil/toxicidade , Sistema de Condução Cardíaco/efeitos dos fármacos , Síndrome do QT Longo/induzido quimicamente , Piperidinas/toxicidade , Piridinas/toxicidade , Torsades de Pointes/induzido quimicamente , Testes de Toxicidade , Animais , Cães , Relação Dose-Resposta a Droga , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Técnicas In Vitro , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/fisiopatologia , Bloqueadores dos Canais de Potássio/farmacologia , Medição de Risco , Risperidona/toxicidade , Fatores de Tempo , Torsades de Pointes/metabolismo , Torsades de Pointes/fisiopatologia , Testes de Toxicidade/métodos , Verapamil/toxicidade , Imagens com Corantes Sensíveis à Voltagem
11.
Circulation ; 120(7): 553-9, 2009 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-19652097

RESUMO

BACKGROUND: Cardiac repolarization, the process by which cardiomyocytes return to their resting potential after each beat, is a highly regulated process that is critical for heart rhythm stability. Perturbations of cardiac repolarization increase the risk for life-threatening arrhythmias and sudden cardiac death. Although genetic studies of familial long-QT syndromes have uncovered several key genes in cardiac repolarization, the major heritable contribution to this trait remains unexplained. Identification of additional genes may lead to a better understanding of the underlying biology, aid in identification of patients at risk for sudden death, and potentially enable new treatments for susceptible individuals. METHODS AND RESULTS: We extended and refined a zebrafish model of cardiac repolarization by using fluorescent reporters of transmembrane potential. We then conducted a drug-sensitized genetic screen in zebrafish, identifying 15 genes, including GINS3, that affect cardiac repolarization. Testing these genes for human relevance in 2 concurrently completed genome-wide association studies revealed that the human GINS3 ortholog is located in the 16q21 locus, which is strongly associated with QT interval. CONCLUSIONS: This sensitized zebrafish screen identified 15 novel myocardial repolarization genes. Among these genes is GINS3, the human ortholog of which is a major locus in 2 concurrent human genome-wide association studies of QT interval. These results reveal a novel network of genes that regulate cardiac repolarization.


Assuntos
Perfilação da Expressão Gênica , Genes Reguladores/fisiologia , Sistema de Condução Cardíaco/fisiologia , Coração/fisiologia , Potenciais da Membrana/fisiologia , Animais , Morte Súbita Cardíaca/epidemiologia , Humanos , Síndrome do QT Longo/genética , Modelos Animais , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Fenótipo , Fatores de Risco , Peixe-Zebra
12.
Am J Physiol Heart Circ Physiol ; 298(3): H787-94, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20035026

RESUMO

Gap junctions are critical to maintaining synchronized impulse propagation and repolarization. Heterogeneous expression of the principal ventricular gap junction protein connexin43 (Cx43) is associated with action potential duration (APD) dispersion across the anterior ventricular wall. Little is known about Cx43 expression patterns and their disparate impact on regional electrophysiology throughout the heart. We aimed to determine whether the anterior and posterior regions of the heart are electrophysiologically distinct. Multisegment, high-resolution optical mapping was performed in canine wedge preparations harvested separately from the anterior left ventricle (aLV; n = 8) and posterior left ventricle (pLV; n = 8). Transmural APD dispersion was significantly greater on the aLV than the pLV (45 +/- 13 vs. 26 +/- 8.0 ms; P < 0.05). Conduction velocity dispersion was also significantly higher (P < 0.05) across the aLV (39 +/- 7%) than the pLV (16 +/- 3%). Carbenoxolone perfusion significantly enhanced APD and conduction velocity dispersion on the aLV (by 1.53-fold and 1.36-fold, respectively), but not the pLV (by 1.27-fold and 1.2-fold, respectively), and produced a 4.2-fold increase in susceptibility to inducible arrhythmias in the aLV. Confocal immunofluorescence microscopy revealed significantly (P < 0.05) greater transmural dispersion of Cx43 expression on the aLV (44 +/- 10%) compared with the pLV wall (8.3 +/- 0.7%), suggesting that regional expression of Cx43 expression patterns may account for regional electrophysiological differences. Computer simulations affirmed that localized uncoupling at the epicardial-midmyocardial interface is sufficient to produce APD gradients observed on the aLV. These data demonstrate that the aLV and pLV differ importantly with respect to their electrophysiological properties and Cx43 expression patterns. Furthermore, local underexpression of Cx43 is closely associated with transmural electrophysiological heterogeneity on the aLV. Therefore, regional and transmural heterogeneous Cx43 expression patterns may be an important mechanism underlying arrhythmia susceptibility, particularly in disease states where gap junction expression is altered.


Assuntos
Potenciais de Ação/fisiologia , Junções Comunicantes/fisiologia , Sistema de Condução Cardíaco/fisiologia , Função Ventricular Esquerda/fisiologia , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Simulação por Computador , Conexina 43/genética , Conexina 43/metabolismo , Cães , Técnicas Eletrofisiológicas Cardíacas , Ventrículos do Coração/metabolismo , Masculino , Modelos Animais
13.
Pacing Clin Electrophysiol ; 33(3): 346-52, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20025710

RESUMO

Altering the pattern of activation of the ventricle causes remodeling of the mechanical and electrical properties of the myocardium. The electrical remodeling is evident on the surface electrocardiogram as significant change in T-wave polarity following altered activation; this phenomenon is ascribed to as "T-wave memory" or "cardiac memory." The electrophysiological remodeling following altered activation is characterized by distinct changes in regions proximal (early-activated) versus distal (late-activated) to the site of altered activation. The early-activated region exhibits marked attenuation of epicardial phase 1 notch due to reduced expression of the transient outward potassium current (I(to)). This is attributed to electrotonic changes during altered activation, and angiotensin-mediated regulation of Kv4.3 (the pore-forming alpha subunit responsible for I(to)). The late-activated region exhibits the most significant action potential prolongation due to markedly increased mechanical strain through a mechano-electrical feedback mechanism. Consequently, regionally heterogeneous action potential remodeling occurs following altered activation. This enhances regional repolarization gradients that underlie the electrophysiological basis for T-wave memory. Further, recent clinical studies highlight detrimental consequences of altered activation including worsening mechanical function and increased susceptibility to arrhythmias. Future studies to identify molecular mechanisms that link electrotonic and mechanical strain-induced changes to cellular electrophysiology will provide important insights into the role of altered activation in regulating cardiac repolarization and arrhythmogenesis.


Assuntos
Sistema de Condução Cardíaco/fisiopatologia , Taquicardia Ventricular/fisiopatologia , Remodelação Ventricular/fisiologia , Potenciais de Ação/fisiologia , Animais , Estimulação Cardíaca Artificial , Conexinas/fisiologia , Desfibriladores Implantáveis , Eletrocardiografia , Sistema de Condução Cardíaco/efeitos dos fármacos , Humanos , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Taquicardia Ventricular/terapia , Fatores de Tempo
14.
Prog Biophys Mol Biol ; 97(2-3): 332-47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18395246

RESUMO

Despite the strong association between mechanical dysfunction of the heart and sudden death due to arrhythmias, the causal relationship is not well understood. Cardiac alternans has been linked to arrhythmogenesis and can be mediated by intracellular calcium handling. Given the integral role intracellular calcium plays in contractile function, calcium-mediated alternans may represent an important mechanistic link between mechanical dysfunction and electrical instability. This relationship, however, is not well understood due to complex feedback between membrane currents, intracellular calcium, and contraction. This manuscript describes the cellular mechanisms of cardiac alternans. Through several pathways, calcium transient alternans is coupled to repolarization alternans that can form a substrate for reentrant excitation. Abnormal intracellular calcium cycling, either impaired release or impaired reuptake of sarcoplasmic reticulum calcium, is a cellular mechanism of calcium transient alternans. Thus, cardiac alternans is an important mechanistic link between mechanical dysfunction and sudden cardiac death.


Assuntos
Arritmias Cardíacas/fisiopatologia , Sinalização do Cálcio/fisiologia , Sistema de Condução Cardíaco/fisiopatologia , Mecanotransdução Celular/fisiologia , Miócitos Cardíacos/fisiologia , Animais , Morte Súbita Cardíaca , Humanos
15.
Heart Rhythm ; 16(2): 281-289, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30193854

RESUMO

BACKGROUND: Cardiac alternans is promoted by heart failure (HF)-induced calcium (Ca2+) cycling abnormalities. Late sodium current (INa,L) is enhanced in HF and promotes Ca2+ overload; however, mechanisms underlying an antiarrhythmic effect of INa,L blockade in HF remain unclear. OBJECTIVE: The purpose of this study was to determine whether ranolazine suppresses cardiac alternans in HF by normalizing Ca2+ cycling. METHODS: Transmural dual optical mapping of Ca2+ transients and action potentials was performed in wedge preparations from 8 HF and 8 control (normal) dogs. Susceptibility to action potential duration alternans (APD-ALT) and Ca2+ transient alternans (Ca-ALT) was compared at baseline and with ranolazine (5-10 µM). RESULTS: HF increased APD- and Ca-ALT compared to normal (both P <.05), and ranolazine suppressed APD- and Ca-ALT in both groups (P <.05). The incidence of spatially discordant alternans (DIS-ALT) was increased by HF (8/8) compared to normal (4/8; P <.05), and ranolazine decreased DIS-ALT in HF (4/8; P <.05).Not only did ranolazine mitigate HF-induced Ca2+ overload, it also attenuated APD-ALT to Ca-ALT gain (amount of APD-ALT produced by Ca-ALT). In HF, APD-ALT to Ca-ALT gain was significantly increased (0.55 ± 0.02) compared to normal (0.44 ± 0.02; P <.05) and was normalized by ranolazine (0.36 ± 0.05; P <.05), representing a complementary mechanism by which INa,L blockade suppressed cardiac alternans. CONCLUSION: Ranolazine attenuated arrhythmogenic cardiac alternans in HF, both by suppressing Ca-ALT and decreasing the coupling gain of APD-ALT to Ca-ALT. Blockade of INa,L may reverse impaired Ca2+ cycling to mitigate cardiac alternans, representing a mechanism underlying the antiarrhythmic benefit of INa,L blockade in HF.


Assuntos
Arritmias Cardíacas/tratamento farmacológico , Cálcio/metabolismo , Sistema de Condução Cardíaco/efeitos dos fármacos , Insuficiência Cardíaca/complicações , Miócitos Cardíacos/metabolismo , Ranolazina/uso terapêutico , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/metabolismo , Modelos Animais de Doenças , Cães , Sistema de Condução Cardíaco/fisiopatologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos/patologia , Imagem Óptica/métodos , Bloqueadores dos Canais de Sódio/uso terapêutico
16.
J Mol Cell Cardiol ; 44(1): 31-43, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18061204

RESUMO

The close relationship between life-threatening ventricular arrhythmias and contractile dysfunction in the heart implicates intracellular calcium cycling as an important underlying mechanism of arrhythmogenesis. Despite this close association, however, the mechanisms of arrhythmogenesis attributable to impaired calcium cycling are not fully appreciated or understood. In this report we review some of the current thinking regarding arrhythmia mechanisms associated with either abnormal impulse initiation (i.e. arrhythmia triggers) or impulse propagation (i.e. arrhythmia substrates). In all cases, the mechanisms are primarily related to dysfunction of calcium regulatory proteins associated with the sarcomere. These findings highlight the broad scope of arrhythmias associated with abnormal calcium cycling, and provide a basis for a causal relationship between cardiac electrical instability and contractile dysfunction. Moreover, calcium cycling proteins may provide much needed targets for novel antiarrhythmic therapies.


Assuntos
Arritmias Cardíacas/terapia , Cálcio/metabolismo , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Miocárdio/metabolismo , Animais , Sistema de Condução Cardíaco , Humanos , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia
17.
Circulation ; 115(25): 3145-55, 2007 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-17562957

RESUMO

BACKGROUND: Altered electrical activation of the heart by pacing or disease induces profound ventricular electrical remodeling (VER), manifested electrocardiographically as T-wave memory and ultimately as deleterious mechanical remodeling from heterogeneous strain. Although T-wave memory is associated with altered expression of sarcolemmal ion channels, the biophysical mechanisms responsible for triggering remodeling of cardiac ion channels are unknown. METHODS AND RESULTS: To test the hypothesis that mechanoelectrical feedback triggered by regional strain is a mechanism for VER, dogs (n=6) underwent 4 weeks of ventricular pacing to induce VER. Multisegment transmural optical action potential imaging of left ventricular wedges revealed profound and selective prolongation of action potential duration in late-activated (288+/-29 ms) compared with early-activated (250+/-9 ms) myocardial segments (P<0.05), providing the first experimental evidence that amplification of repolarization gradients between segments of left ventricle is the electrophysiological basis for T-wave memory. In vivo tagged magnetic resonance imaging revealed a 2-fold and preferential increase in circumferential strain in late-activated segments of myocardium, which exactly coincided with segments undergoing VER. VER could not be attributed to structural remodeling because it occurred without any histological evidence of cellular hypertrophy. CONCLUSIONS: The mechanism responsible for triggering remodeling of ion channel function in VER was locally enhanced circumferential strain. These data suggest a novel mechanoelectrical feedback mechanism for inducing physiological and potentially deleterious electrical heterogeneities in the heart.


Assuntos
Eletrocardiografia , Sistema de Condução Cardíaco/fisiopatologia , Ventrículos do Coração/fisiopatologia , Canais Iônicos/fisiologia , Sarcolema/fisiologia , Estresse Mecânico , Potenciais de Ação , Animais , Estimulação Cardíaca Artificial , Cães , Retroalimentação Fisiológica , Fibrose , Ventrículos do Coração/patologia , Hipertrofia , Imageamento por Ressonância Magnética , Masculino , Modelos Cardiovasculares , Miócitos Cardíacos/fisiologia , Necrose , Método Simples-Cego
18.
Europace ; 9 Suppl 6: vi77-82, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17959697

RESUMO

T-wave alternans, a powerful marker for the risk of sudden cardiac death is directly related to alternans of the cellular action potential. When action potential alternans is first initiated, it occurs with identical phase in all cells of a particular region of the heart. However, above a critical heart rate threshold, action potential alternans switches phase in some cells but not in others, such that some cells undergo a prolongation of action potential duration (APD), whereas neighbouring cells undergo APD shortening on the same beat (i.e. discordant alternans). Discordant alternans is linked to a mechanism of arrhythmogenesis because when ventricular action potentials from neighbouring cells are alternating out of phase, repolarization gradients are amplified, producing conduction block and re-entrant excitation. In this review, we discuss potential mechanisms which may underlie discordant alternans in the heart, including (i) conduction velocity restitution, (ii) spatial heterogeneities of calcium cycling and the sarcolemmal ionic currents which govern repolarization, and (iii) intercellular uncoupling.


Assuntos
Potenciais de Ação/fisiologia , Arritmias Cardíacas/complicações , Arritmias Cardíacas/fisiopatologia , Morte Súbita Cardíaca/etiologia , Sistema de Condução Cardíaco/fisiopatologia , Sinalização do Cálcio/fisiologia , Humanos
19.
J Electrocardiol ; 40(6 Suppl): S1-7, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17993305

RESUMO

Heart failure is a progressive clinical syndrome that is characterized by remodeling of the myocardium in response to various stress signals. The past several years has seen remarkable progress in unraveling the molecular and cellular mechanisms of structural and electrical remodeling in HF. Improved understanding of the molecular mechanism of myocardial remodeling has resulted in improved HF therapies and revealed potentially novel therapeutic targets. This review discusses the mechanisms of myocardial remodeling in HF and their clinical manifestations. Current and investigational HF therapies targeting these mechanisms also will be discussed.


Assuntos
Baixo Débito Cardíaco/fisiopatologia , Baixo Débito Cardíaco/terapia , Estimulação Cardíaca Artificial/métodos , Cardiotônicos/uso terapêutico , Terapia Genética/métodos , Sistema de Condução Cardíaco/fisiopatologia , Transplante de Células-Tronco/métodos , Animais , Sistemas de Liberação de Medicamentos/métodos , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/cirurgia , Humanos , Modelos Cardiovasculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA