Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 315(2): H318-H326, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29522372

RESUMEN

The action mechanism of stimulation toward spiral waves (SWs) owing to the complex excitation patterns that occur just after point stimulation has not yet been experimentally clarified. This study sought to test our hypothesis that the effect of capturing excitable gap of SWs by stimulation can also be explained as the interaction of original phase singularity (PS) and PSs induced by the stimulation on the wave tail (WT) of the original SW. Phase variance analysis was used to quantitatively analyze the postshock PS trajectories. In a two-dimensional subepicardial layer of Langendorff-perfused rabbit hearts, optical mapping was used to record the excitation pattern during stimulation. After a SW was induced by S1-S2 shock, single biphasic point stimulation S3 was applied. In 70 of the S1-S2-S3 stimulation episodes applied on 6 hearts, the original PS was clearly observed just before the S3 point stimulation in 37 episodes. Pairwise PSs were newly induced by the S3 in 20 episodes. The original PS collided with the newly induced PSs in 16 episodes; otherwise, they did not interact with the original PS. SW shift occurred most efficiently when the S3 shock was applied at the relative refractory period, and PS shifted in the direction of the WT. In conclusion, quantitative tracking of PS clarified that stimulation in desirable conditions induces pairwise PSs on WT and that the collision of PSs causes SW shift along the WT. The results of this study indicate the importance of the interaction of shock-induced excitation with the WT for effective stimulation. NEW & NOTEWORTHY The quantitative analysis of spiral wave dynamics during stimulation clarified the action mechanism of capturing the excitable gap, i.e., the induction of pairwise phase singularities on the wave tail and spiral wave shift along the wave tail as a result of these interactions. The importance of the wave tail for effective stimulation was revealed.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Corazón/fisiología , Modelos Cardiovasculares , Animales , Conejos
2.
Biochem Biophys Rep ; 9: 245-256, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28956012

RESUMEN

Andersen-Tawil syndrome (ATS) is a rare inherited channelopathy. The cardiac phenotype in ATS is typified by a prominent U wave and ventricular arrhythmia. An effective treatment for this disease remains to be established. We reprogrammed somatic cells from three ATS patients to generate induced pluripotent stem cells (iPSCs). Multi-electrode arrays (MEAs) were used to record extracellular electrograms of iPSC-derived cardiomyocytes, revealing strong arrhythmic events in the ATS-iPSC-derived cardiomyocytes. Ca2+ imaging of cells loaded with the Ca2+ indicator Fluo-4 enabled us to examine intracellular Ca2+ handling properties, and we found a significantly higher incidence of irregular Ca2+ release in the ATS-iPSC-derived cardiomyocytes than in control-iPSC-derived cardiomyocytes. Drug testing using ATS-iPSC-derived cardiomyocytes further revealed that antiarrhythmic agent, flecainide, but not the sodium channel blocker, pilsicainide, significantly suppressed these irregular Ca2+ release and arrhythmic events, suggesting that flecainide's effect in these cardiac cells was not via sodium channels blocking. A reverse-mode Na+/Ca2+exchanger (NCX) inhibitor, KB-R7943, was also found to suppress the irregular Ca2+ release, and whole-cell voltage clamping of isolated guinea-pig cardiac ventricular myocytes confirmed that flecainide could directly affect the NCX current (INCX). ATS-iPSC-derived cardiomyocytes recapitulate abnormal electrophysiological phenotypes and flecainide suppresses the arrhythmic events through the modulation of INCX.

3.
Circ J ; 82(1): 19-27, 2017 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-28781288

RESUMEN

BACKGROUND: A recent study suggested that midkine (MK), a heparin-binding growth factor, is associated with atherosclerosis progression in patients with artery disease. It has previously been reported that MK plays a critical role in neointima formation in a restenosis model, whereas the role of MK in the development of atherosclerosis has not been investigated. The present study assessed the effect of MK administration on the process of atherosclerotic plaque formation in apolipoprotein E-knockout (ApoE-/-) mice.Methods and Results:Using an osmotic pump, human recombinant MK protein was intraperitoneally administered for 12 weeks in C57BL/6 ApoE-/-(ApoE-/--MK) and ApoE+/+mice fed a high-fat diet. Saline was administered to the control groups of ApoE-/-(ApoE-/--saline) and ApoE+/+mice. The atherosclerotic lesion areas in longitudinal aortic sections were significantly larger in ApoE-/--MK mice than in ApoE-/--saline mice. The aortic mRNA levels of pro-inflammatory and angiogenic factors, and the percentage of macrophages in aortic root lesions, were significantly higher in ApoE-/--MK mice than in ApoE-/--saline mice, whereas the percentage of apoptotic cells was significantly lower in ApoE-/--MK mice than in ApoE-/--saline mice. CONCLUSIONS: The systemic administration of MK in ApoE-/-mice promoted atherosclerotic plaque formation through pro-inflammatory, angiogenic, and anti-apoptotic effects. MK may serve as a potential therapeutic target for the prevention of atherosclerosis under atherogenic conditions.


Asunto(s)
Apoptosis/efectos de los fármacos , Inflamación/inducido químicamente , Midkina/farmacología , Neovascularización Patológica/inducido químicamente , Placa Aterosclerótica/patología , Animales , Aorta/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Placa Aterosclerótica/etiología , Proteínas Recombinantes/farmacología
4.
Circ J ; 81(10): 1411-1428, 2017 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-28552884

RESUMEN

BACKGROUND: Ventricular tachycardia/fibrillation (VT/VF) associated with acute myocardial ischemia is the most common cause of sudden cardiac death, but its underlying mechanisms are incompletely understood. It is hypothesized that late Na+current (INa) contributes to arrhythmogenic activity in ischemic myocardium.Methods and Results:Langendorff-perfused rabbit hearts with regional ischemia in ventricles were optically mapped. Perfusion with ranolazine (10 µmol/L), a selective inhibitor of lateINa, significantly reduced excitation frequency and facilitated termination of VT/VF induced after occlusion of the left main coronary trunk. The activation pattern during ischemic VT/VF was characterized by breakthrough-type excitations (BEs) from multiple origins, predominantly in the ischemic border zone (BZ) and occasional short-lived rotors. Ranolazine perfusion significantly reduced the incidence of BEs in the BZ. Rotors tended to decrease with progression of ischemia and disappeared after ranolazine perfusion. During constant pacing, ranolazine attenuated ischemia-induced shortening of action potentials in the BZ without affecting conduction velocity, probably due toIKrinhibition. In intact hearts without coronary occlusion, ranolazine (10 µmol/L) terminated aconitine-induced VT by inhibiting focal arrhythmogenic activity in the injection site. CONCLUSIONS: LateINa-mediated focal arrhythmogenic activity plays important roles in the maintenance of ischemic VT/VF in isolated rabbit hearts. Suppression of lateINaby ranolazine may be a promising therapeutic strategy to reduce arrhythmic death during the acute phase of myocardial infarction.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Isquemia Miocárdica , Ranolazina/farmacología , Taquicardia Ventricular/tratamiento farmacológico , Animales , Muerte Súbita Cardíaca , Sistema de Conducción Cardíaco/efectos de los fármacos , Preparación de Corazón Aislado , Conejos , Ranolazina/uso terapéutico , Bloqueadores de los Canales de Sodio/farmacología , Bloqueadores de los Canales de Sodio/uso terapéutico , Taquicardia Ventricular/fisiopatología
5.
Sci Rep ; 6: 34198, 2016 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-27677334

RESUMEN

SCN5A is abundant in heart and has a major role in INa. Loss-of-function mutation in SCN5A results in Brugada syndrome (BrS), which causes sudden death in adults. It remains unclear why disease phenotype does not manifest in the young even though mutated SCN5A is expressed in the young. The aim of the present study is to elucidate the timing of the disease manifestation in BrS. A gain-of-function mutation in SCN5A also results in Long QT syndrome type 3 (LQTS3), leading to sudden death in the young. Induced pluripotent stem cells (iPSCs) were generated from a patient with a mixed phenotype of LQTS3 and BrS with the E1784K SCN5A mutation. Here we show that electrophysiological analysis revealed that LQTS3/BrS iPSC-derived cardiomyocytes recapitulate the phenotype of LQTS3 but not BrS. Each ß-subunit of the sodium channel is differentially expressed in embryonic and adult hearts. SCN3B is highly expressed in embryonic hearts and iPSC-derived cardiomyocytes. A heterologous expression system revealed that INa of mutated SCN5A is decreased and SCN3B augmented INa of mutated SCN5A. Knockdown of SCN3B in LQTS3/BrS iPSC-derived cardiomyocytes successfully unmasked the phenotype of BrS. Isogenic control of LQTS3/BrS (corrected-LQTS3/BrS) iPSC-derived cardiomyocytes gained the normal electrophysiological properties.

6.
Am J Physiol Heart Circ Physiol ; 311(3): H750-8, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27422985

RESUMEN

It has been reported that blockade of the inward rectifier K(+) current (IK1) facilitates termination of ventricular fibrillation. We hypothesized that partial IK1 blockade destabilizes spiral wave (SW) re-entry, leading to its termination. Optical action potential (AP) signals were recorded from left ventricles of Langendorff-perfused rabbit hearts with endocardial cryoablation. The dynamics of SW re-entry were analyzed during ventricular tachycardia (VT), induced by cross-field stimulation. Intercellular electrical coupling in the myocardial tissue was evaluated by the space constant. In separate experiments, AP recordings were made using the microelectrode technique from right ventricular papillary muscles of rabbit hearts. Ba(2+) (10-50 µM) caused a dose-dependent prolongation of VT cycle length and facilitated termination of VT in perfused hearts. Baseline VT was maintained by a stable rotor, where an SW rotated around an I-shaped functional block line (FBL). Ba(2+) at 10 µM prolonged I-shaped FBL and phase-singularity trajectory, whereas Ba(2+) at 50 µM transformed the SW rotation dynamics from a stable linear pattern to unstable circular/cycloidal meandering. The SW destabilization was not accompanied by SW breakup. Under constant pacing, Ba(2+) caused a dose-dependent prolongation of APs, and Ba(2+) at 50 µM decreased conduction velocity. In papillary muscles, Ba(2+) at 50 µM depolarized the resting membrane potential. The space constant was increased by 50 µM Ba(2+) Partial IK1 blockade destabilizes SW rotation dynamics through a combination of prolongation of the wave length, reduction of excitability, and enhancement of electrotonic interactions, which facilitates termination of ventricular tachyarrhythmias.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Bario/farmacología , Corazón/efectos de los fármacos , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Taquicardia Ventricular/metabolismo , Fibrilación Ventricular/metabolismo , Animales , Arritmias Cardíacas , Criocirugía , Corazón/fisiopatología , Preparación de Corazón Aislado , Imagen Óptica , Canales de Potasio de Rectificación Interna/metabolismo , Conejos , Taquicardia Ventricular/fisiopatología , Fibrilación Ventricular/fisiopatología
7.
Cardiovasc Res ; 111(4): 410-21, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27357638

RESUMEN

AIM: In healthy hearts, ventricular gap junctions are mainly composed by connexin43 (Cx43) and localize in the intercalated disc, enabling appropriate electrical coupling. In diseased hearts, Cx43 is heterogeneously down-regulated, whereas activity of calmodulin/calcium-calmodulin protein kinase II (CaM/CaMKII) signalling increases. It is unclear if CaM/CaMKII affects Cx43 expression/localization or impulse propagation. We analysed different models to assess this. METHODS AND RESULTS: AC3-I mice with CaMKII genetically inhibited were subjected to pressure overload (16 weeks, TAC vs. sham). Optical and epicardial mapping was performed on Langendorff-perfused rabbit and AC3-I hearts, respectively. Cx43 subcellular distribution from rabbit/mouse ventricles was evaluated by immunoblot after Triton X-100-based fractionation. In mice with constitutively reduced CaMKII activity (AC3-I), conduction velocity (CV) was augmented (n = 11, P < 0.01 vs. WT); in AC3-I, CV was preserved after TAC, in contrast to a reduction seen in TAC-WT mice (-20%). Cx43 expression was preserved after TAC in AC3-I mice, though arrhythmias and fibrosis were still present. In rabbits, W7 (CaM inhibitor, 10 µM) increased CV (6-13%, n= 6, P< 0.05), while susceptibility to arrhythmias decreased. Immunoconfocal microscopy revealed enlarged Cx43 cluster sizes at intercalated discs of those hearts. Total Cx43 did not change by W7 (n= 4), whereas Triton X-100 insoluble Cx43 increased (+21%, n= 4, P< 0.01). Similar findings were obtained in AC3-I mouse hearts when compared with control, and in cultured dog cardiomyocytes. Functional implication was shown through increased intercellular coupling in cultured neonatal rat cardiomyocytes. CONCLUSION: Both acute and chronic CaM/CaMKII inhibition improves conduction characteristics and enhances localization of Cx43 in the intercalated disc. In the absence of fibrosis, this reduced the susceptibility for arrhythmias.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calmodulina/metabolismo , Comunicación Celular/efectos de los fármacos , Corazón/fisiopatología , Miocardio/metabolismo , Animales , Antiarrítmicos/metabolismo , Conexina 43/metabolismo , Perros , Uniones Comunicantes/metabolismo , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/metabolismo , Ratones , Modelos Animales , Conejos , Ratas
8.
Heart Vessels ; 31(1): 96-104, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25155308

RESUMEN

Midkine (MK), a heparin-binding growth factor, has been shown to prevent cardiac remodeling after ischemic injury through its anti-apoptotic effect. Cell apoptosis is central to the pathophysiology of cardiac remodeling in congestive heart failure (CHF) of ischemic as well as non-ischemic origin. We hypothesized that MK exerts the anti-apoptotic cardioprotective effect in CHF of non-ischemic etiology. MK protein or vehicle (normal saline) was subcutaneously administered in tachycardia-induced CHF rabbits (right ventricular pacing, 350 beats/min, 4 weeks). The vehicle-treated rabbits (n = 19, control) demonstrated severe CHF and high mortality rate, whereas MK (n = 16) demonstrated a well-compensated state and a lower mortality rate. In echocardiography, left ventricular (LV) end-diastolic dimension decreased in MK versus control, whereas LV systolic function increased. In histological analysis (picrosirius red staining), MK decreased collagen deposition area compared with control. TUNEL staining showed that MK prevented cell apoptosis and minimized myocyte loss in the CHF rabbit ventricle, associated with activation of PI3-K/Akt signaling, producing a parallel decrease of Bax/Bcl-2 ratio. MK prevented progression of cardiac remodeling in the CHF rabbit, likely by activation of anti-apoptotic signaling. Exogenous MK application might be a novel therapeutic strategy for CHF due to non-ischemic origin.


Asunto(s)
Estimulación Cardíaca Artificial/efectos adversos , Citocinas/administración & dosificación , Insuficiencia Cardíaca/tratamiento farmacológico , Miocardio/patología , Remodelación Ventricular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Ecocardiografía , Ventrículos Cardíacos/fisiopatología , Masculino , Midkina , Contracción Miocárdica/efectos de los fármacos , Conejos , Taquicardia
9.
Cardiovasc Res ; 109(1): 162-73, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26531129

RESUMEN

AIMS: The progression of pathological left ventricular remodelling leads to cardiac dysfunction and contributes to the occurrence of malignant arrhythmias and sudden cardiac death. The underlying molecular mechanisms remain unclear, however. Our aim was to examine the role of the renin-angiotensin system (RAS) in the mechanism underlying arrhythmogenic cardiac remodelling using a transgenic mouse expressing a cardiac-specific dominant-negative form of neuron-restrictive silencer factor (dnNRSF-Tg). This mouse model exhibits progressive cardiac dysfunction leading to lethal arrhythmias. METHODS AND RESULTS: Subcutaneous administration of aliskiren, a direct renin inhibitor, significantly suppressed the progression of pathological cardiac remodelling and improved survival among dnNRSF-Tg mice while reducing arrhythmogenicity. Genetic deletion of the angiotensin type 1a receptor (AT1aR) similarly suppressed cardiac remodelling and sudden death. In optical mapping analyses, spontaneous ventricular tachycardia (VT) and fibrillation (VF) initiated by breakthrough-type excitations originating from focal activation sites and maintained by functional re-entry were observed in dnNRSF-Tg hearts. Under constant pacing, dnNRSF-Tg hearts exhibited markedly slowed conduction velocity, which likely contributes to the arrhythmogenic substrate. Aliskiren treatment increased conduction velocity and reduced the incidence of sustained VT. These effects were associated with suppression of cardiac fibrosis and restoration of connexin 43 expression in dnNRSF-Tg ventricles. CONCLUSION: Renin inhibition or genetic deletion of AT1aR suppresses pathological cardiac remodelling that leads to the generation of substrates maintaining VT/VF and reduces the occurrence of sudden death in dnNRSF-Tg mice. These findings demonstrate the significant contribution of RAS activation to the progression of arrhythmogenic substrates.


Asunto(s)
Arritmias Cardíacas/etiología , Cardiomiopatías/complicaciones , Sistema Renina-Angiotensina/fisiología , Animales , Conexina 43/análisis , Fibrosis , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Receptor de Angiotensina Tipo 1/fisiología , Renina/antagonistas & inhibidores , Remodelación Ventricular
10.
Cardiovasc Res ; 104(2): 364-70, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25183791

RESUMEN

AIMS: The aim of this study was to investigate the role of gap junctions in atrial fibrillation (AF) by analysing the effects of a gap junction enhancer and blocker on AF vulnerability and electrophysiological properties of isolated hearts. METHODS AND RESULTS: The acute atrial stretch model of AF in the isolated rabbit heart was used. Sustained AF (SAF) was induced by a burst of high-frequency stimulation of the Bachmann's bundle. The effective refractory period (ERP) was measured, and the total conduction time (TCT) and the pattern of conduction of the anterior surface of the left atrium were monitored by using an optical mapping system. The effect of enhancing gap junction function by 100-1000 nM rotigaptide (ZP123) and block by 30 µM carbenoxolone on these parameters was measured. SAF inducibility was increased with an elevation of intra-atrial pressure. Enhanced gap junction conductance induced by treatment with 100-1000 nM rotigaptide reduced SAF inducibility, and the gap junction blocker carbenoxolone increased SAF inducibility. In the absence of gap junction enhancer or blocker, normal conduction was observed at 0 cmH2O. When intra-atrial pressure was raised to 12 cmH2O, the conduction pattern was changed to a heterogeneous zig-zag pattern and TCT was prolonged. Conduction pattern was not affected by either agent. Rotigaptide shortened TCT, whereas carbenoxolone prolonged TCT. ERP was significantly shortened with an increase in intra-atrial pressure, but ERP was unaffected by either agent. CONCLUSION: Gap junction modulators changed AF inducibility through their effects on atrial conduction, not by altering ERP.


Asunto(s)
Fibrilación Atrial/metabolismo , Presión Atrial , Uniones Comunicantes/metabolismo , Atrios Cardíacos/metabolismo , Frecuencia Cardíaca , Mecanotransducción Celular , Husos Musculares/metabolismo , Potenciales de Acción , Animales , Antiarrítmicos/farmacología , Fibrilación Atrial/etiología , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/prevención & control , Carbenoxolona/farmacología , Uniones Comunicantes/efectos de los fármacos , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Preparación de Corazón Aislado , Masculino , Mecanotransducción Celular/efectos de los fármacos , Husos Musculares/efectos de los fármacos , Husos Musculares/fisiopatología , Oligopéptidos/farmacología , Conejos , Periodo Refractario Electrofisiológico , Factores de Tiempo
11.
Biochem Biophys Res Commun ; 452(3): 701-7, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25193703

RESUMEN

Ras-related small G-protein Rad plays a critical role in generating arrhythmias via regulation of the L-type Ca(2+) channel (LTCC). The aim was to demonstrate the role of Rad in intracellular calcium homeostasis by cardiac-Specific dominant-negative suppression of Rad. Transgenic (TG) mice overexpressing dominant-negative mutant Rad (S105N Rad TG) were generated. To measure intracellular Ca(2+) concentration ([Ca(2+)]i), we recorded [Ca(2+)]i transients and Ca(2+) sparks from isolated cardiomyocytes using confocal microscopy. The mean [Ca(2+)]i transient amplitude was significantly increased in S105N Rad TG cardiomyocytes, compared with control littermate mouse cells. The frequency of Ca(2+) sparks was also significantly higher in TG cells than in control cells, although there were no significant differences in amplitude. The sarcoplasmic reticulum Ca(2+) content was not altered in the S105N Rad TG cells, as assessed by measuring caffeine-induced [Ca(2+)]i transient. In contrast, phosphorylation of Ser(2809) on the cardiac ryanodine receptor (RyR2) was significantly enhanced in TG mouse hearts compared with controls. Additionally, the Rad-mediated RyR2 phosphorylation was regulated via a direct interaction of Rad with protein kinase A (PKA).


Asunto(s)
Arritmias Cardíacas/genética , Calcio/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Proteínas ras/genética , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Cafeína/farmacología , Señalización del Calcio , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Mutación , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación , Cultivo Primario de Células , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Proteínas ras/deficiencia
12.
PLoS One ; 9(4): e94565, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24759974

RESUMEN

The sinoatrial node (SAN) is heterogeneous in terms of cell size, ion channels, current densities, connexins and electrical coupling. For example, Nav1.5 (responsible for INa) and Cx43 (responsible for electrical coupling) are absent from the centre of the SAN (normally the leading pacemaker site), but present in the periphery (at SAN-atrial muscle junction). To test whether the heterogeneity is important for the functioning of the SAN, one- and two-dimensional models of the SAN and surrounding atrial muscle were created. Normal functioning of the SAN (in terms of cycle length, position of leading pacemaker site, conduction times, activation and repolarization sequences and space constants) was observed when, from the centre to the periphery, (i) cell characteristics (cell size and ionic current densities) were changed in a gradient fashion from a central-type (lacking INa) to a peripheral-type (possessing INa) and (ii) coupling conductance was increased in a gradient fashion. We conclude that the heterogeneous nature of the node is important for its normal functioning. The presence of Nav1.5 and Cx43 in the periphery may be essential for the node to be able to drive the atrial muscle: Nav1.5 provides the necessary depolarizing current and Cx43 delivers it to the atrial muscle.


Asunto(s)
Nodo Sinoatrial/fisiología , Potenciales de Acción/fisiología , Animales , Conexina 43/metabolismo , Atrios Cardíacos/metabolismo , Canales Iónicos/metabolismo , Conejos , Nodo Sinoatrial/metabolismo
13.
Circ J ; 77(11): 2728-35, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23986028

RESUMEN

BACKGROUND: To establish a simple and accurate method for the automated identification of the end of a T wave, we approximated electrocardiograph (ECG) traces using a Gaussian mixture model in conjunction with a split-and-merge expectation-maximization algorithm. METHODS AND RESULTS: A total of 286 ECG traces of heart beats of 50 healthy men were used as control data and ECGs from 15 subjects recorded before and after 400mg oral moxifloxacin as positive controls. An experienced cardiologist determined the reference points by visual inspection of the original ECGs. The primary estimated point for the end of the T wave was selected as the point 2 ms before the point at which the gradient of the approximated wave was not steeper than the common threshold value. This point was then adjusted by applying modification rules proposed by an experienced cardiologist. The absolute value of the average interval between the resulting final estimated point and the manually selected reference point was 1.8±7.7 ms for the control data. After treatment with moxifloxacin, the average QT interval, corrected by Bazett's formula, showed a 17.2±27.1 ms prolongation with a lower bound of the 95% confidence interval of 4.9 ms. CONCLUSIONS: When the modification rules were applied, the accuracy of QT measurement was improved, and the present system was capable of detecting QT prolongation correctly.


Asunto(s)
Electrocardiografía/métodos , Frecuencia Cardíaca/fisiología , Modelos Cardiovasculares , Adulto , Antibacterianos/farmacología , Compuestos Aza/farmacología , Femenino , Fluoroquinolonas , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Moxifloxacino , Quinolinas/farmacología
14.
Proc Natl Acad Sci U S A ; 110(31): 12667-72, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23861494

RESUMEN

Heart disease remains a leading cause of death worldwide. Owing to the limited regenerative capacity of heart tissue, cardiac regenerative therapy has emerged as an attractive approach. Direct reprogramming of human cardiac fibroblasts (HCFs) into cardiomyocytes may hold great potential for this purpose. We reported previously that induced cardiomyocyte-like cells (iCMs) can be directly generated from mouse cardiac fibroblasts in vitro and vivo by transduction of three transcription factors: Gata4, Mef2c, and Tbx5, collectively termed GMT. In the present study, we sought to determine whether human fibroblasts also could be converted to iCMs by defined factors. Our initial finding that GMT was not sufficient for cardiac induction in HCFs prompted us to screen for additional factors to promote cardiac reprogramming by analyzing multiple cardiac-specific gene induction with quantitative RT-PCR. The addition of Mesp1 and Myocd to GMT up-regulated a broader spectrum of cardiac genes in HCFs more efficiently compared with GMT alone. The HCFs and human dermal fibroblasts transduced with GMT, Mesp1, and Myocd (GMTMM) changed the cell morphology from a spindle shape to a rod-like or polygonal shape, expressed multiple cardiac-specific proteins, increased a broad range of cardiac genes and concomitantly suppressed fibroblast genes, and exhibited spontaneous Ca(2+) oscillations. Moreover, the cells matured to exhibit action potentials and contract synchronously in coculture with murine cardiomyocytes. A 5-ethynyl-2'-deoxyuridine assay revealed that the iCMs thus generated do not pass through a mitotic cell state. These findings demonstrate that human fibroblasts can be directly converted to iCMs by defined factors, which may facilitate future applications in regenerative medicine.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica , Proteínas Musculares/biosíntesis , Miocitos Cardíacos/metabolismo , Factores de Transcripción/biosíntesis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Niño , Preescolar , Femenino , Fibroblastos/citología , Humanos , Lactante , Masculino , Ratones , Persona de Mediana Edad , Proteínas Musculares/genética , Miocitos Cardíacos/citología , Factores de Transcripción/genética
15.
Heart Rhythm ; 10(6): 901-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23454487

RESUMEN

BACKGROUND: The mechanisms by which acute left atrial ischemia (LAI) leads to atrial fibrillation (AF) initiation and perpetuation remain unclear. OBJECTIVE: To investigate the electrophysiological mechanisms of AF perpetuation in the presence of regional atrial ischemia. METHODS: LAI (90-minute ischemia) was obtained in isolated sheep hearts by selectively perfusing microspheres into the left anterior atrial artery. Two charge-coupled device cameras and several bipolar electrodes enabled recording from multiple atrial locations: with a dual-camera setup (protocol 1, n = 10, and protocol 1', n = 4, for biatrial or atrioventricular camera setups, respectively), in the presence of propranolol/atropine (1 µM) added to the perfusate after LAI (protocol 2, n = 3) and after a pretreatment with glibenclamide (10 µM; protocol 3, n = 4). RESULTS: Spontaneous AF occurred in 41.2% (7 of 17) of the hearts that were in sinus rhythm before LAI. LAI caused action potential duration shortening in both the ischemic (IZ) and nonischemic (NIZ) zones by 21% ± 8% and 34% ± 13%, respectively (pacing, 5 Hz; P<.05 compared to baseline). Apparent impulse velocity was significantly reduced in the IZ but not in the NIZ (-65% ± 19% and 9% ± 18%; P = .001 and NS, respectively). During LAI-related AF, a significant NIZ maximal dominant frequency increase from 7.4 ± 2.5 to 14.0 ± 5.5 Hz (P<.05) was observed. Glibenclamide, an ATP-sensitive potassium current (IKATP) channel blocker, averted LAI-related maximal dominant frequency increase (NIZ: LAI vs glibenclamide 14.0 ± 5.5 Hz vs 5.9 ± 1.3 Hz; P<.05). An interplay between spontaneous focal discharges and rotors, locating at the IZ-NIZ border zone, maintained LAI-related AF. CONCLUSIONS: LAI leads to an IKATP conductance-dependent action potential duration shortening and spontaneous AF maintained by both spontaneous focal discharges and reentrant circuits locating at the IZ border zone.


Asunto(s)
Fibrilación Atrial/fisiopatología , Atrios Cardíacos/fisiopatología , Isquemia Miocárdica/fisiopatología , Animales , Modelos Animales de Enfermedad , Técnicas Electrofisiológicas Cardíacas , Gliburida/farmacología , Técnicas In Vitro , Ovinos , Imagen de Colorante Sensible al Voltaje
16.
Cardiovasc Res ; 98(1): 136-44, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23341577

RESUMEN

AIMS: Na(+) channel blockers are often used to treat atrial fibrillation (AF), but may sometimes cause ventricular contractile dysfunction. However, amiodarone, a multi-channel blocker with Na(+) channel block, causes less contractile dysfunction. In this study, we tested the hypothesis that Na(+) channel block by amiodarone is selective in atrial myocytes (AM) compared with ventricular myocytes (VM). METHODS AND RESULTS: Na(+) currents (INa) were measured using whole-cell patch-clamp technique in isolated rabbit AM and VM. Amiodarone inhibited INa in AM (IC50: 1.8 ± 1.1 µM; n = 8) much more than in VM (40.4 ± 11.9 µM; n = 7, P < 0.01). Amiodarone at 10 µM shifted the steady-state inactivation relationship in AM (-16.2 ± 1.7 mV shift, n = 12) compared with VM (-5.9 ± 0.7 mV shift; n = 13; P < 0.01). For mexiletine, the inhibition of INa and inactivation curve shifts were comparable for AM and VM. The effects of amiodarone and mexiletine on conduction velocity (CV) in Langendorff-perfused rabbit hearts were evaluated using an optical mapping system. The decrease of CV by 3 µM amiodarone was significantly larger in the atrium (-18.9 ± 3.8% change; n = 5) compared with the ventricle (-3.7 ± 3.7%; n = 5; P < 0.01). In contrast, mexiletine reduced CV equally in the atrium and the ventricle. CONCLUSION: Amiodarone preferentially inhibits INa of AM compared with VM. Atrial selective Na(+) channel block by amiodarone may contribute to treating AF with less effect on ventricular contractility than other Na(+) channel blockers.


Asunto(s)
Amiodarona/farmacología , Atrios Cardíacos/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiología , Masculino , Mexiletine/farmacología , Miocitos Cardíacos/efectos de los fármacos , Conejos
17.
Circ J ; 76(12): 2799-806, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22972199

RESUMEN

BACKGROUND: Right atrial (RA) appendage (RAA) pacing is reported to impair hemodynamic benefits of cardiac resynchronization therapy (CRT) through a considerable delay of left atrial (LA) contraction, which compromises appropriate balance of atrioventricular (AV) and left ventricular (LV) synchrony. Potential usefulness of Bachmann's bundle (BB) pacing to solve the problem remains to be confirmed. METHODS AND RESULTS: Atrial synchrony and LV performance was investigated by echocardiography in 25 patients undergoing pacemaker implantation with preserved AV conduction and LV function (Group I), and 15 patients receiving CRT (Group II). In Group I, RAA pacing (AAI mode, n=10) increased P-wave duration (PWD) and RA-to-LA contraction delay (IAMD) compared with sinus rhythm (132±14 and 35±12 ms vs. 108±16 and 13±13 ms, P<0.001). The delayed LA contraction was associated with early interruption of LV filling, leading to an impairment of LV performance (Tei index: 0.43±0.12 vs. 0.34±0.09, P<0.01). BB pacing (AAI, n=15) did not cause such undesirable effects. In Group II, RA (BB)-paced biventricular pacing (DDD) reduced PWD and IAMD compared with RA-sensed biventricular pacing (VDD) (102±14 and -3±13 ms vs. 117±10 and 21±18 ms, P<0.001). This restoration of atrial synchrony was associated with significant improvement of LV performance (Tei index: 0.56±0.18 vs. 0.62±0.16, P<0.05). CONCLUSIONS: BB pacing preserves atrial synchrony, and might be more favorable than RAA pacing for maximizing hemodynamic efficacy of CRT.


Asunto(s)
Estimulación Cardíaca Artificial/métodos , Terapia de Resincronización Cardíaca , Sistema de Conducción Cardíaco/fisiopatología , Insuficiencia Cardíaca/terapia , Función Ventricular Izquierda , Adulto , Anciano , Anciano de 80 o más Años , Apéndice Atrial/fisiopatología , Ecocardiografía Doppler , Electrocardiografía , Femenino , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/fisiopatología , Hemodinámica , Humanos , Masculino , Persona de Mediana Edad , Recuperación de la Función , Volumen Sistólico , Factores de Tiempo , Resultado del Tratamiento
18.
Circulation ; 126(17): 2051-64, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-22992321

RESUMEN

BACKGROUND: Fibroblast proliferation and differentiation are central in atrial fibrillation (AF)-promoting remodeling. Here, we investigated fibroblast regulation by Ca(2+)-permeable transient receptor potential canonical-3 (TRPC3) channels. METHODS AND RESULTS: Freshly isolated rat cardiac fibroblasts abundantly expressed TRPC3 and had appreciable nonselective cation currents (I(NSC)) sensitive to a selective TPRC3 channel blocker, pyrazole-3 (3 µmol/L). Pyrazole-3 suppressed angiotensin II-induced Ca(2+) influx, proliferation, and α-smooth muscle actin protein expression in fibroblasts. Ca(2+) removal and TRPC3 blockade suppressed extracellular signal-regulated kinase phosphorylation, and extracellular signal-regulated kinase phosphorylation inhibition reduced fibroblast proliferation. TRPC3 expression was upregulated in atria from AF patients, goats with electrically maintained AF, and dogs with tachypacing-induced heart failure. TRPC3 knockdown (based on short hairpin RNA [shRNA]) decreased canine atrial fibroblast proliferation. In left atrial fibroblasts freshly isolated from dogs kept in AF for 1 week by atrial tachypacing, TRPC3 protein expression, currents, extracellular signal-regulated kinase phosphorylation, and extracellular matrix gene expression were all significantly increased. In cultured left atrial fibroblasts from AF dogs, proliferation rates, α-smooth muscle actin expression, and extracellular signal-regulated kinase phosphorylation were increased and were suppressed by pyrazole-3. MicroRNA-26 was downregulated in canine AF atria; experimental microRNA-26 knockdown reproduced AF-induced TRPC3 upregulation and fibroblast activation. MicroRNA-26 has NFAT (nuclear factor of activated T cells) binding sites in the 5' promoter region. NFAT activation increased in AF fibroblasts, and NFAT negatively regulated microRNA-26 transcription. In vivo pyrazole-3 administration suppressed AF while decreasing fibroblast proliferation and extracellular matrix gene expression. CONCLUSIONS: TRPC3 channels regulate cardiac fibroblast proliferation and differentiation, likely by controlling the Ca(2+) influx that activates extracellular signal-regulated kinase signaling. AF increases TRPC3 channel expression by causing NFAT-mediated downregulation of microRNA-26 and causes TRPC3-dependent enhancement of fibroblast proliferation and differentiation. In vivo, TRPC3 blockade prevents AF substrate development in a dog model of electrically maintained AF. TRPC3 likely plays an important role in AF by promoting fibroblast pathophysiology and is a novel potential therapeutic target.


Asunto(s)
Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Fibroblastos/metabolismo , Canales Catiónicos TRPC/fisiología , Animales , Fibrilación Atrial/genética , Función del Atrio Derecho/genética , Proliferación Celular , Células Cultivadas , Perros , Regulación hacia Abajo/genética , Fibroblastos/patología , Técnicas de Silenciamiento del Gen/métodos , Cabras , Células HEK293 , Humanos , Ratas , Canales Catiónicos TRPC/genética
19.
Cardiovasc Res ; 95(4): 419-29, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22739119

RESUMEN

AIMS: Long QT syndrome (LQTS) is an inheritable and life-threatening disease; however, it is often difficult to determine disease characteristics in sporadic cases with novel mutations, and more precise analysis is necessary for the successful development of evidence-based clinical therapies. This study thus sought to better characterize ion channel cardiac disorders using induced pluripotent stem cells (iPSCs). METHODS AND RESULTS: We reprogrammed somatic cells from a patient with sporadic LQTS and from controls, and differentiated them into cardiomyocytes through embryoid body (EB) formation. Electrophysiological analysis of the LQTS-iPSC-derived EBs using a multi-electrode array (MEA) system revealed a markedly prolonged field potential duration (FPD). The IKr blocker E4031 significantly prolonged FPD in control- and LQTS-iPSC-derived EBs and induced frequent severe arrhythmia only in LQTS-iPSC-derived EBs. The IKs blocker chromanol 293B did not prolong FPD in the LQTS-iPSC-derived EBs, but significantly prolonged FPD in the control EBs, suggesting the involvement of IKs disturbance in the patient. Patch-clamp analysis and immunostaining confirmed a dominant-negative role for 1893delC in IKs channels due to a trafficking deficiency in iPSC-derived cardiomyocytes and human embryonic kidney (HEK) cells. CONCLUSIONS: This study demonstrated that iPSCs could be useful to characterize LQTS disease as well as drug responses in the LQTS patient with a novel mutation. Such analyses may in turn lead to future progress in personalized medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Síndrome de Romano-Ward/metabolismo , Potenciales de Acción , Adolescente , Animales , Diferenciación Celular , Reprogramación Celular , Técnicas de Cocultivo , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/trasplante , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Masculino , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/trasplante , Técnicas de Placa-Clamp , Fenotipo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Síndrome de Romano-Ward/diagnóstico , Síndrome de Romano-Ward/genética , Síndrome de Romano-Ward/patología , Teratoma/metabolismo , Teratoma/patología , Factores de Tiempo , Transfección
20.
Am J Physiol Heart Circ Physiol ; 303(5): H578-86, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22707561

RESUMEN

Spiral-wave (SW) reentry is a major organizing principle of ventricular tachycardia/fibrillation (VT/VF). We tested a hypothesis that pharmacological modification of gap junction (GJ) conductance affects the stability of SW reentry in a two-dimensional (2D) epicardial ventricular muscle layer prepared by endocardial cryoablation of Langendorff-perfused rabbit hearts. Action potential signals were recorded and analyzed by high-resolution optical mapping. Carbenoxolone (CBX; 30 µM) and rotigaptide (RG, 0.1 µM) were used to inhibit and enhance GJ coupling, respectively. CBX decreased the space constant (λ) by 36%, whereas RG increased it by 22-24% (n = 5; P < 0.01). During centrifugal propagation, there was a linear relationship between the wavefront curvature (κ) and local conduction velocity (LCV): LCV = LCV(0) - D·κ (D, diffusion coefficient; LCV(0), LCV at κ = 0). CBX decreased LCV(0) and D by 27 ± 3 and 57 ± 3%, respectively (n = 5; P < 0.01). RG increased LCV(0) and D by 18 ± 3 and 54 ± 5%, respectively (n = 5, P < 0.01). The regression lines with and without RG crossed, resulting in a paradoxical decrease of LCV with RG at κ > ~60 cm(-1). SW reentry induced after CBX was stable, and the incidence of sustained VTs (>30 s) increased from 38 ± 4 to 85 ± 4% after CBX (n = 18; P < 0.01). SW reentry induced after RG was characterized by decremental conduction near the rotation center, prominent drift and self-termination by collision with the anatomical boundaries, and the incidence of sustained VTs decreased from 40 ± 5 to 17 ± 6% after RG (n = 13; P < 0.05). These results suggest that decreased intercellular coupling stabilizes SW reentry in 2D cardiac muscle, whereas increased coupling facilitates its early self-termination.


Asunto(s)
Antiarrítmicos/farmacología , Carbenoxolona/farmacología , Comunicación Celular/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Sistema de Conducción Cardíaco/efectos de los fármacos , Oligopéptidos/farmacología , Taquicardia Ventricular/prevención & control , Fibrilación Ventricular/prevención & control , Potenciales de Acción , Animales , Modelos Animales de Enfermedad , Técnicas Electrofisiológicas Cardíacas , Uniones Comunicantes/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Perfusión , Conejos , Taquicardia Ventricular/etiología , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Fibrilación Ventricular/etiología , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/fisiopatología , Imagen de Colorante Sensible al Voltaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...