Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Front Biosci (Landmark Ed) ; 29(5): 200, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38812311

RESUMEN

AIMS: Changes in myocardial mitochondrial morphology and function in premature ventricular contractions (PVCs)-induced cardiomyopathy (PVCCM) remain poorly studied. Here, we investigated the effects of PVCs with different coupling intervals (CIs) on myocardial mitochondrial remodelling in a canine model of PVCCM. METHODS AND RESULTS: Twenty-one beagles underwent pacemaker implantation and were randomised into the sham (n = 7), short-coupled PVCs (SCP, n = 7), and long-coupled PVCs (LCP, n = 7) groups. Right ventricular (RV) apical bigeminy was produced for 12-week to induce PVCCM in the SCP (CI, 250 ms) and LCP (CI, 350 ms) groups. Echocardiography was performed at baseline and biweekly thereafter to evaluate cardiac function. Masson's trichrome staining measured ventricular interstitial fibrosis. The ultrastructural morphology of the myocardial mitochondria was analysed using transmission electron microscopy. Mitochondrial Ca2+ concentration, reactive oxygen species (ROS) levels, adenosine triphosphate (ATP) content, membrane potential, and electron transport chain (ETC) complex activity were measured to assess myocardial mitochondrial function. Twelve-week-PVCs led to left ventricular (LV) enlargement with systolic dysfunction, disrupted mitochondrial morphology, increased mitochondrial Ca2+ concentration and ROS levels, decreased mitochondrial ATP content and membrane potential, and impaired ETC complex activity in both the SCP and LCP groups (all p < 0.01 vs the sham group). Ventricular fibrosis was observed only in canines with LCP. Worse cardiac function and more pronounced abnormalities in mitochondrial morphology and function were observed in the LCP group than to the SCP group (all p < 0.05). CONCLUSION: We demonstrated myocardial mitochondrial abnormalities in dogs with PVCCM, characterised by abnormal mitochondrial morphology, mitochondrial Ca2+ overload, oxidative stress, and impaired mitochondrial energy metabolism. Compared to SCP, long-term LCP exposure resulted in more severe mitochondrial remodelling and cardiac dysfunction in dogs.


Asunto(s)
Calcio , Cardiomiopatías , Modelos Animales de Enfermedad , Mitocondrias Cardíacas , Especies Reactivas de Oxígeno , Complejos Prematuros Ventriculares , Animales , Perros , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/ultraestructura , Mitocondrias Cardíacas/patología , Cardiomiopatías/fisiopatología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/etiología , Complejos Prematuros Ventriculares/fisiopatología , Complejos Prematuros Ventriculares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Calcio/metabolismo , Masculino , Adenosina Trifosfato/metabolismo , Potencial de la Membrana Mitocondrial , Ecocardiografía
2.
Mol Cell Biochem ; 478(7): 1447-1456, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36350464

RESUMEN

Premature ventricular contractions (PVCs) are the most frequent ventricular arrhythmias in the overall population. PVCs are known to acutely enhance contractility by the post-extrasystolic potentiation phenomenon, but over time persistent PVCs promote PVC-induced cardiomyopathy (PVC-CM), characterized by a reduction of the left ventricular (LV) ejection fraction. Ca2+ cycling in myocytes commands muscle contraction and in this process, SERCA2 leads the Ca2+ reuptake into the sarcoplasmic reticulum (SR) shaping cytosolic Ca2+ signal decay and muscle relaxation. Altered Ca2+ reuptake can contribute to the contractile dysfunction observed in PVC-CM. To better understand Ca2+ handling using our PVC-CM model (canines with 50% PVC burden for 12 weeks), SR-Ca2+ reuptake was investigated by measuring Ca2+ dynamics and analyzing protein expression. Kinetic analysis of Ca2+ reuptake in electrically paced myocytes showed a ~ 21 ms delay in PVC-CM compared to Sham in intact isolated myocytes, along with a ~ 13% reduction in SERCA2 activity assessed in permeabilized myocytes. Although these trends were not statistically significant between groups using hierarchical statistics, relaxation of myocytes following contraction was significantly slower in PVC-CM vs Sham myocytes. Western blot analyses indicate a 22% reduction in SERCA2 expression, a 23% increase in phospholamban (PLN) expression, and a 50% reduction in PLN phosphorylation in PVC-CM samples vs Sham. Computational analysis simulating a 20% decrease in SR-Ca2+ reuptake resulted in a ~ 22 ms delay in Ca2+ signal decay, consistent with the experimental result described above. In conclusion, SERCA2 and PLB alterations described above have a modest contribution to functional adaptations observed in PVC-CM.


Asunto(s)
Cardiomiopatías , Complejos Prematuros Ventriculares , Animales , Perros , Complejos Prematuros Ventriculares/metabolismo , Retículo Sarcoplasmático/metabolismo , Cinética , Cardiomiopatías/metabolismo , Células Musculares , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Calcio/metabolismo , Miocitos Cardíacos/metabolismo
3.
FASEB J ; 36(8): e22455, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35899468

RESUMEN

Hypokalemia causes ectopic heartbeats, but the mechanisms underlying such cardiac arrhythmias are not understood. In reduced serum K+ concentrations that occur under hypokalemia, K2P1 two-pore domain K+ channels change ion selectivity and switch to conduct inward leak cation currents, which cause aberrant depolarization of resting potential and induce spontaneous action potential of human cardiomyocytes. K2P1 is expressed in the human heart but not in mouse hearts. We test the hypothesis that K2P1 leak cation channels contribute to ectopic heartbeats under hypokalemia, by analysis of transgenic mice, which conditionally express induced K2P1 specifically in hearts, mimicking K2P1 channels in the human heart. Conditional expression of induced K2P1 specifically in the heart of hypokalemic mice results in multiple types of ventricular ectopic beats including single and multiple ventricular premature beats as well as ventricular tachycardia and causes sudden death. In isolated mouse hearts that express induced K2P1, sustained ventricular fibrillation occurs rapidly after perfusion with low K+ concentration solutions that mimic hypokalemic conditions. These observed phenotypes occur rarely in control mice or in the hearts that lack K2P1 expression. K2P1-expressing mouse cardiomyocytes of transgenic mice much more frequently fire abnormal single and/or rhythmic spontaneous action potential in hypokalemic conditions, compared to wild type mouse cardiomyocytes without K2P1 expression. These findings confirm that K2P1 leak cation channels induce ventricular ectopic beats and sudden death of transgenic mice with hypokalemia and imply that K2P1 leak cation channels may play a critical role in human ectopic heartbeats under hypokalemia.


Asunto(s)
Hipopotasemia , Complejos Prematuros Ventriculares , Potenciales de Acción , Animales , Cationes/metabolismo , Muerte Súbita , Humanos , Hipopotasemia/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Complejos Prematuros Ventriculares/metabolismo
4.
Cells ; 11(6)2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35326403

RESUMEN

Background: Cellular transplantation has emerged as promising approach for treating cardiac diseases. However, a poor engraftment rate limits our understanding on how transplanted cardiomyocytes contribute to cardiac function in the recipient's heart. Methods: The CRISPR/Cas9 technique was employed for stable and constitutive gene expression in human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs). Myocardial infarction was induced in adult immunodeficient mice, followed by intramyocardial injection of hiPSC-CMs expressing either CCND2/channelrhodopsin 2 (hiPSC-CCND2OE/ChR2OECMs) or CCND2/luciferase (hiPSC-CCND2OE/LuciOECMs). Six months later, hemodynamics and intramural electrocardiogram were recorded upon blue light illuminations in anesthetized, open-chest mice. Results: Blue light resets automaticity of spontaneously beating hiPSC-CCND2OE/ChR2OECMs in culture, but not that of hiPSC-CCND2OE/LuciOECMs. Response to blue light was also observed in mice carrying large (>106 cells) intracardiac grafts of hiPSC-CCND2OE/ChR2OECM but not in mice carrying hiPSC-CCND2OE/LuciOECMs. The former exhibited single premature ventricular contractions upon light illumination or ventricular quadrigeminy upon second-long illuminations. At the onset of premature ventricular contractions, maximal systolic ventricular pressure decreased while ventricular volume rose concomitantly. Light-induced changes reversed upon resumption of sinus rhythm. Conclusions: We established an in vivo model for optogenetic-based modulation of the excitability of donor cardiomyocytes in a functional, reversible, and localized manner. This approach holds unique value for studying electromechanical coupling and molecular interactions between donor cardiomyocytes and recipient hearts in live animals.


Asunto(s)
Células Madre Pluripotentes Inducidas , Infarto del Miocardio , Complejos Prematuros Ventriculares , Animales , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Optogenética , Complejos Prematuros Ventriculares/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 320(6): H2283-H2294, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33929896

RESUMEN

Fibroblast growth factor 23 (FGF23) is a phosphate regulating protein hormone released by osteocytes. FGF23 becomes markedly elevated in chronic kidney disease (CKD), for which the leading cause of death is cardiovascular disease, particularly sudden cardiac death. Previously, we found that FGF23 increases intracellular Ca2+ in cardiomyocytes and alters contractility in mouse ventricles ex vivo via FGF receptor 4 (FGFR4). In the present study, we demonstrate that FGF23 induces cardiac arrhythmias and prolongs QTc interval in mice, and we tested whether these effects are mediated through FGFR4. In isolated Langendorff perfused hearts, FGF23 perfusion increased mechanical arrhythmias in the form of premature ventricular beats (PVBs), and induced runs of ventricular tachycardia in 6 of 11 animals, which were attenuated with pretreatment of an anti-FGFR4 blocking antibody. Ex vivo ECG analysis of isolated intact hearts showed increased ventricular arrhythmias and QTc prolongation after FGF23 infusion compared with vehicle. In vivo, injection of FGF23 into the jugular vein led to the emergence of premature ventricular contractions (PVCs) in 5 out of 11 experiments. FGF23 also produced a significant lengthening effect upon QTc interval in vivo. In vivo FGFR4 blockade ameliorated the arrhythmogenic and QTc prolonging effects of FGF23. Finally, FGF23 increased cardiomyocyte Ca2+ levels in intact left ventricular muscle which was inhibited by FGR4 blockade. We conclude that FGF23/FGFR4 signaling in the heart may contribute to ventricular arrhythmogenesis and repolarization disturbances commonly observed in patients with CKD via Ca2+ overload and may be an important therapeutic target to reduce cardiac mortality in CKD.NEW & NOTEWORTHY Here we provide direct evidence that fibroblast growth factor 23 (FGF23), a phosphaturic hormone elevated in chronic kidney disease, is proarrhythmic. FGF23 acutely triggered ventricular arrhythmias and prolonged corrected QT interval (QTc) in isolated mouse hearts and in vivo. FGF23 also increased Ca2+ levels in ventricular muscle tissue. Blockade of the FGF receptor 4 signaling pathway using a monoclonal antibody ameliorated ventricular arrhythmias, QTc prolongation, and elevated ventricular Ca2+ induced by FGF23, and may represent a potential therapeutic target in chronic kidney disease.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Síndrome de QT Prolongado/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Taquicardia Ventricular/metabolismo , Complejos Prematuros Ventriculares/metabolismo , Animales , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Electrocardiografía , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/farmacología , Corazón/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Preparación de Corazón Aislado , Ratones , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Transducción de Señal
6.
BMC Endocr Disord ; 20(1): 61, 2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32393234

RESUMEN

BACKGROUND: Cardiac damage triggered by severe hypocalcemia is well known. However, the role of chronic hypoparathyroidism (HP) and pseudohypoparathyroidism (PHP) in cardiac health is still unclear. We investigated the effect of chronic HP and PHP on cardiac structure and conductive function in patients compiling with treatment. METHODS: The study included 18 patients with HP and eight with PHP aged 45.4 ± 15.4 and 22.1 ± 6.4 years, respectively with a previously regular follow-up. In addition, 26 age- and sex-matched healthy controls were included. General characteristics and biochemical indices were recorded. Cardiac function and structure were assessed by estimation of myocardial enzymes, B-type natriuretic peptide (BNP), and echocardiography. The 12-lead electrocardiogram and 24-h Holter electrocardiography were performed to evaluate the conductive function. RESULTS: Levels of serum calcium in HP and PHP were 2.05 ± 0.16 mmol/L and 2.25 ± 0.19 mmol/L, respectively. The levels of myocardial enzyme and BNP were within the normal range. Adjusting for age at evaluation and body mass index, all M-mode measurements, left ventricular mass (LVM), LVM index (LVMI) and relative wall thickness (RWT) were comparable between patients and controls. Prolongation of corrected QT (QTc) intervals occurred in 52.6% (10/19) of patients, and 6.7% (1/15) of patients manifested more than 100 episodes of supraventricular and ventricular extrasystoles, as well as supraventricular tachycardia. None of the above arrhythmias was related to a severe clinical event. CONCLUSIONS: From this pilot study, patients diagnosed with HP and PHP and well-controlled serum calcium levels manifested normal cardiac morphology and ventricular function, except for prolonged QTc intervals, and a small percentage of mild arrhythmias needing further investigation.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Hipoparatiroidismo/fisiopatología , Seudohipoparatiroidismo/fisiopatología , Disfunción Ventricular Izquierda/fisiopatología , Adolescente , Adulto , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Complejos Atriales Prematuros/etiología , Complejos Atriales Prematuros/metabolismo , Complejos Atriales Prematuros/fisiopatología , Calcio/metabolismo , Estudios de Casos y Controles , Enfermedad Crónica , Ecocardiografía , Electrocardiografía , Electrocardiografía Ambulatoria , Femenino , Humanos , Hipoparatiroidismo/complicaciones , Hipoparatiroidismo/metabolismo , Síndrome de QT Prolongado/etiología , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/fisiopatología , Masculino , Persona de Mediana Edad , Péptido Natriurético Encefálico/metabolismo , Proyectos Piloto , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/metabolismo , Taquicardia Supraventricular/etiología , Taquicardia Supraventricular/metabolismo , Taquicardia Supraventricular/fisiopatología , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/metabolismo , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología , Adulto Joven
7.
J Cardiovasc Pharmacol ; 75(5): 432-438, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32079857

RESUMEN

BACKGROUND: Low-intensity focused ultrasound (LIFU) has been shown to be a beneficial tool for autonomic nervous system modulation, but its effect on the left stellate ganglion (LSG) remains unknown. OBJECTIVE: To seek the effect of LIFU on myocardial infarction (MI)-induced LSG activation and ventricular arrhythmias (VAs). METHODS: In this study, 20 dogs were included and randomly divided into the LIFU (LIFU & MI, n = 8), Sham (sham LIFU & MI, n = 8), and Control group (sham LIFU & sham MI, n = 4). For each LIFU intervention (1.0-2.0 W, 10 minutes) of the LSG, the LSG function, ventricular effective refractory period (ERP), and temperature were tested pre-intervention and postintervention. Thereafter, MI was induced by left anterior artery ligation and VAs were recorded for 1 hour. At the end, both the LSG and the heart were extracted for biomedical and histological analysis. RESULTS: In the Sham group, no significant change was shown in ventricular ERP or LSG function for any intensity settings of sham LIFU intervention when compared with the group baseline. In the LIFU group, however, both 1.5 and 2.0 W LIFU modulation of LSG resulted in significant prolongation of ERP and attenuation of LSG function. Furthermore, the incidence of VAs was significantly attenuated in the LIFU group compared with the Sham group. Moreover, histological analysis showed that no damage or apoptosis was observed in LSG although a statistically significant increase was shown in temperature (maximal increase <1°C) with 1.5 and 2.0 W LIFU intervention. CONCLUSION: LIFU stimulation may be a safe and beneficial tool for LSG attenuation and VA prevention in the MI canine model.


Asunto(s)
Corazón/inervación , Infarto del Miocardio/terapia , Ganglio Estrellado/fisiopatología , Taquicardia Ventricular/prevención & control , Terapia por Ultrasonido , Fibrilación Ventricular/prevención & control , Complejos Prematuros Ventriculares/prevención & control , Potenciales de Acción , Animales , Modelos Animales de Enfermedad , Perros , Frecuencia Cardíaca , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Periodo Refractario Electrofisiológico , Taquicardia Ventricular/etiología , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Fibrilación Ventricular/etiología , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/fisiopatología , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología
8.
Physiol Res ; 68(5): 867-871, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31424250

RESUMEN

Endothelin alters central sympathetic responses, but the resultant effects on arrhythmogenesis are unknown. We examined ventricular tachyarrhythmias after endothelin receptor-A blockade in the brain of Wistar rats with acute myocardial infarction. For this aim, BQ-123 (n=6) or phosphate-buffered saline (n=6) were injected intracerebroventricularly. After 10 min, the left coronary artery was ligated, followed by implantation of telemetry transmitters. Electrocardiography and voluntary activity (as a surrogate of acute left ventricular failure) were continuously monitored for 24 h. Infarct-size was similar in the two groups. There were fewer episodes of ventricular tachyarrhythmias of shorter average duration in treated rats, leading to markedly shorter total duration (12.3+/-8.9 s), when compared to controls (546.2+/-130.3 s). Voluntary activity increased in treated rats during the last hours of recording, but bradyarrhythmic episodes were comparable between the two groups. Endothelin receptor-A blockade in the brain of rats decreases the incidence of ventricular tachyarrhythmias post-ligation, without affecting bradyarrhythmic episodes. These findings call for further research on the pathophysiologic role of endothelin during acute myocardial infarction.


Asunto(s)
Ventrículos Cerebrales/efectos de los fármacos , Antagonistas de los Receptores de la Endotelina A/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Péptidos Cíclicos/administración & dosificación , Receptor de Endotelina A/efectos de los fármacos , Taquicardia Ventricular/prevención & control , Complejos Prematuros Ventriculares/prevención & control , Animales , Ventrículos Cerebrales/metabolismo , Ventrículos Cerebrales/fisiopatología , Modelos Animales de Enfermedad , Inyecciones Intraventriculares , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Ratas Wistar , Receptor de Endotelina A/metabolismo , Taquicardia Ventricular/etiología , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología
9.
Am J Physiol Heart Circ Physiol ; 317(3): H607-H616, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31322427

RESUMEN

Mechanisms behind development of premature ventricular contraction (PVC)-induced cardiomyopathy remain unclear. PVCs may adversely modulate the autonomic nervous system to promote development of heart failure. Afferent neurons in the inferior vagal (nodose) ganglia transduce cardiac activity and modulate parasympathetic output. Effects of PVCs on cardiac parasympathetic efferent and vagal afferent neurotransmission are unknown. The purpose of this study was to evaluate effects of PVCs on vagal afferent neurotransmission and compare these effects with a known powerful autonomic modulator, myocardial ischemia. In 16 pigs, effects of variably coupled PVCs on heart rate variability (HRV) and vagal afferent neurotransmission were evaluated. Direct nodose neuronal recordings were obtained in vivo, and cardiac-related afferent neurons were identified based on their response to cardiovascular interventions, including ventricular chemical and mechanical stimuli, left anterior descending (LAD) coronary artery occlusion, and variably coupled PVCs. On HRV analysis before versus after PVCs, parasympathetic tone decreased (normalized high frequency: 83.6 ± 2.8 to 72.5 ± 5.3; P < 0.05). PVCs had a powerful impact on activity of cardiac-related afferent neurons, altering activity of 51% of neurons versus 31% for LAD occlusion (P < 0.05 vs. LAD occlusion and all other cardiac interventions). Both chemosensitive and mechanosensitive neurons were activated by PVCs, and their activity remained elevated even after cessation of PVCs. Cardiac afferent neural responses to PVCs were greater than any other intervention, including ischemia of similar duration. These data suggest that even brief periods of PVCs powerfully modulate vagal afferent neurotransmission, reflexly decreasing parasympathetic efferent tone.NEW & NOTEWORTHY Premature ventricular contractions (PVCs) are common in many patients and, at an increased burden, are known to cause heart failure. This study determined that PVCs powerfully modulate cardiac vagal afferent neurotransmission (exerting even greater effects than ventricular ischemia) and reduce parasympathetic efferent outflow to the heart. PVCs activated both mechano- and chemosensory neurons in the nodose ganglia. These peripheral neurons demonstrated adaptation in response to PVCs. This study provides additional data on the potential role of the autonomic nervous system in PVC-induced cardiomyopathy.


Asunto(s)
Cardiomiopatías/etiología , Frecuencia Cardíaca , Corazón/inervación , Contracción Miocárdica , Nervio Vago/fisiopatología , Complejos Prematuros Ventriculares/complicaciones , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Células Quimiorreceptoras/metabolismo , Modelos Animales de Enfermedad , Mecanorreceptores/metabolismo , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Ganglio Nudoso/metabolismo , Ganglio Nudoso/fisiopatología , Sus scrofa , Transmisión Sináptica , Factores de Tiempo , Nervio Vago/metabolismo , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología
10.
J Cardiovasc Electrophysiol ; 30(7): 1138-1147, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31104349

RESUMEN

BACKGROUND: Sympathetic neural activation plays a key role in the incidence and maintenance of acute myocardial infarction (AMI) induced ventricular arrhythmia (VA). Furthermore, previous studies showed that AMI might induce microglia and sympathetic activation and that microglial activation might contribute to sympathetic activation. Recently, studies showed that light emitting diode (LED) therapy might attenuate microglial activation. Therefore, we hypothesized that LED therapy might reduce AMI-induced VA by attenuating microglia and sympathetic activation. METHODS: Thirty anesthetized rats were randomly divided into three groups: the Control group (n = 6), AMI group (n = 12), and AMI + LED group (n = 12). Electrocardiogram (ECG) and left stellate ganglion (LSG) neural activity were continuously recorded. The incidence of VAs was recorded during the first hour after AMI. Furthermore, we sampled the brain and myocardium tissue of the different groups to examine the microglial activation and expression of nerve growth factor (NGF), interleukin-18 (IL-18), and IL-1ß, respectively. RESULTS: Compared to the AMI group, LED therapy significantly reduced the incidence of AMI-induced VAs (ventricular premature beats [VPB] number: 85.08 ± 13.91 vs 27.5 ± 9.168, P < .01; nonsustained ventricular tachycardia (nSVT) duration: 34.39 ± 8.562 vs 9.005 ± 3.442, P < .05; nSVT number: 18.92 ± 4.52 vs 7.583 ± 3.019, P < .05; incidence rate of SVT/VF: 58.33% vs. 8.33%, P < .05) and reduced the LSG neural activity (P < .01) in the AMI + LED group. Furthermore, LED significantly attenuated microglial activation and reduced IL-18, IL-1ß, and NGF expression in the peri-infarct myocardium. CONCLUSION: LED therapy may protect against AMI-induced VAs by suppressing sympathetic neural activity and the inflammatory response.


Asunto(s)
Corazón/inervación , Láseres de Semiconductores , Terapia por Luz de Baja Intensidad/instrumentación , Infarto del Miocardio/radioterapia , Neuroinmunomodulación , Núcleo Hipotalámico Paraventricular/fisiopatología , Ganglio Estrellado/fisiopatología , Taquicardia Supraventricular/prevención & control , Taquicardia Ventricular/prevención & control , Complejos Prematuros Ventriculares/prevención & control , Potenciales de Acción , Animales , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Masculino , Microglía/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Sprague-Dawley , Taquicardia Supraventricular/etiología , Taquicardia Supraventricular/metabolismo , Taquicardia Supraventricular/fisiopatología , Taquicardia Ventricular/etiología , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología
11.
Heart Rhythm ; 16(5): 765-772, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30414461

RESUMEN

BACKGROUND: Type 2 diabetes (T2D) increases arrhythmia risk through incompletely elucidated mechanisms. Ventricular arrhythmias could be initiated by delayed afterdepolarizations (DADs) resulting from elevated spontaneous sarcoplasmic reticulum (SR) Ca2+ release (SR Ca2+ leak). OBJECTIVE: The purpose of this study was to test the role of DADs and SR Ca2+ leak in triggering arrhythmias in T2D hearts. METHODS: We compared rats with late-onset T2D that display pancreatic and cardiac phenotypes similar to those in humans with T2D (HIP rats) and their nondiabetic littermates (wild type [WT]). RESULTS: HIP rats showed higher propensity for premature ventricular complexes and ventricular tachyarrhythmias, whereas HIP myocytes displayed more frequent DADs and had lower SR Ca2+ content than WT. However, the threshold SR Ca2+ at which depolarizing transient inward currents (Itis) are generated was also significantly decreased in HIP myocytes and was below the actual SR Ca2+ load, which explains the increased DAD incidence despite reduced Ca2+ in SR. In agreement with these findings, Ca2+ spark frequency was augmented in myocytes from HIP vs WT rats, which suggests activation of ryanodine receptors (RyRs) in HIP hearts. Indeed, RyR phosphorylation (by CaMKII and protein kinase A) and oxidation are enhanced in HIP hearts, whereas there is no RyR O-GlcNAcylation in either HIP or control hearts. CaMKII inhibition dissipated the difference in Ca2+ spark frequency between HIP and WT myocytes. CONCLUSION: The threshold SR Ca2+ for generating depolarizing Itis is lower in T2D because of RyR activation after hyperphosphorylation and oxidation, which favors the occurrence of DADs despite low SR Ca2+ loads.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Taquicardia Ventricular/metabolismo , Complejos Prematuros Ventriculares/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Fenómenos Electrofisiológicos/fisiología , Miocitos Cardíacos/metabolismo , Ratas , Taquicardia Ventricular/etiología , Taquicardia Ventricular/fisiopatología , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/fisiopatología
12.
J Cardiovasc Pharmacol Ther ; 24(3): 262-268, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30474396

RESUMEN

Myocardial ischemia-reperfusion (IR) injury is associated with high disability and mortality worldwide. This study was to explore the roles of dioscin in the myocardial IR rats and discover the related molecular mechanisms. Rats were divided into 5 groups: sham, IR, IR + 15 mg/kg dioscin, IR + 30 mg/kg dioscin, and IR + 60 mg/kg dioscin. Heart rate (HR), mean arterial blood pressure (MAP), and rate pressure product (RPP) were evaluated at 10 minutes before ischemia, immediately after ischemia, and at the beginning, middle, and end of reperfusion. Arrhythmia score and myocardial infarct size were examined in rats of all groups. The serum creatine kinase-muscle/brain (CKMB) and cardiac troponin I (cTnI) levels were analyzed via enzyme-linked immunosorbent assay. Protein amount of total connexin 43 (T-Cx43) and phosphorylated connexin 43 (P-Cx43) was evaluated by Western blot. Ischemia reperfusion significantly decreased HR, MAP, and RPP of rats compared to the sham group. However, dioscin significantly attenuated the above phenomena in a dose-dependent manner. Dioscin markedly inhibited IR-induced increase in arrhythmias score, infarct size, and serum CKMB and cTnI levels. In addition, dioscin strikingly induced IR-repressed expression of T-Cx43 and P-Cx43. Our results suggested that dioscin pretreatment exhibited protective effects against myocardial IR injury. Moreover, we found that dioscin attenuated myocardial IR-induced ventricular arrhythmias via upregulating Cx43 expression and activation.


Asunto(s)
Arritmias Cardíacas/prevención & control , Conexina 43/metabolismo , Diosgenina/análogos & derivados , Frecuencia Cardíaca/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Sustancias Protectoras/farmacología , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Biomarcadores/sangre , Forma MB de la Creatina-Quinasa/sangre , Diosgenina/farmacología , Modelos Animales de Enfermedad , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Fosforilación , Ratas Sprague-Dawley , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Taquicardia Ventricular/prevención & control , Troponina I/sangre , Regulación hacia Arriba , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/fisiopatología , Fibrilación Ventricular/prevención & control , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología , Complejos Prematuros Ventriculares/prevención & control
13.
J Am Heart Assoc ; 7(2)2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29335317

RESUMEN

BACKGROUND: Attenuated cardiac vagal activity is associated with ventricular arrhythmogenesis and related mortality in patients with chronic heart failure. Our recent study has shown that expression of N-type Ca2+ channel α-subunits (Cav2.2-α) and N-type Ca2+ currents are reduced in intracardiac ganglion neurons from rats with chronic heart failure. Rat intracardiac ganglia are divided into the atrioventricular ganglion (AVG) and sinoatrial ganglion. Ventricular myocardium receives projection of neuronal terminals only from the AVG. In this study we tested whether a decrease in N-type Ca2+ channels in AVG neurons contributes to ventricular arrhythmogenesis. METHODS AND RESULTS: Lentiviral Cav2.2-α shRNA (2 µL, 2×107 pfu/mL) or scrambled shRNA was in vivo transfected into rat AVG neurons. Nontransfected sham rats served as controls. Using real-time single-cell polymerase chain reaction and reverse-phase protein array, we found that in vivo transfection of Cav2.2-α shRNA decreased expression of Cav2.2-α mRNA and protein in rat AVG neurons. Whole-cell patch-clamp data showed that Cav2.2-α shRNA reduced N-type Ca2+ currents and cell excitability in AVG neurons. The data from telemetry electrocardiographic recording demonstrated that 83% (5 out of 6) of conscious rats with Cav2.2-α shRNA transfection had premature ventricular contractions (P<0.05 versus 0% of nontransfected sham rats or scrambled shRNA-transfected rats). Additionally, an index of susceptibility to ventricular arrhythmias, inducibility of ventricular arrhythmias evoked by programmed electrical stimulation, was higher in rats with Cav2.2-α shRNA transfection compared with nontransfected sham rats and scrambled shRNA-transfected rats. CONCLUSIONS: A decrease in N-type Ca2+ channels in AVG neurons attenuates vagal control of ventricular myocardium, thereby initiating ventricular arrhythmias.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Ganglios Parasimpáticos/metabolismo , Frecuencia Cardíaca , Ventrículos Cardíacos/inervación , Neuronas/metabolismo , Nervio Vago/metabolismo , Complejos Prematuros Ventriculares/metabolismo , Potenciales de Acción , Animales , Canales de Calcio Tipo N/genética , Estimulación Cardíaca Artificial , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ganglios Parasimpáticos/fisiopatología , Masculino , Ratas Sprague-Dawley , Periodo Refractario Electrofisiológico , Factores de Tiempo , Nervio Vago/fisiopatología , Función Ventricular Izquierda , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/genética , Complejos Prematuros Ventriculares/fisiopatología
14.
Rev Cardiovasc Med ; 19(3): 97-101, 2018 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-31054558

RESUMEN

Although ischemic heart disease is invariably associated with marked activation of sympathetic nervous system, elevated levels of circulating catecholamines and lethal ventricular arrhythmias, the mechanisms of arrhythmogenesis due to myocardial ischemia are not fully understood. Since catecholamines are known to produce stimulatory effects in the heart mainly by acting on ß1-adrenoceptors, this study was undertaken to test the involvement of these receptors in the development of arrhythmias due to myocardial infarction (MI) induced upon occluding the left coronary artery in rats for a period of 2 h. The animals were treated with or without atenolol (20 mg/kg; daily), a selective ß1-adrenoceptors blocker, for 14 days before inducing MI. No alterations in the number of MIinduced episodes and incidence or duration of different types of arrhythmias were observed. In fact, the incidence of trigemines and reversible ventricular fibrillation due to MI were significantly increased in the atenolol-treated animals. These observations support the view that the activation of ß;1-adrenoceptors may not be exclusively involved in the development of arrhythmias during the occurrence of ischemic heart disease and other mechanisms can underlie the electric instability of such damaged heart.


Asunto(s)
Frecuencia Cardíaca , Infarto del Miocardio/complicaciones , Miocardio/metabolismo , Miocardio/patología , Receptores Adrenérgicos beta 1/metabolismo , Taquicardia Ventricular/etiología , Fibrilación Ventricular/etiología , Complejos Prematuros Ventriculares/etiología , Antagonistas de Receptores Adrenérgicos beta 1/farmacología , Animales , Atenolol/farmacología , Modelos Animales de Enfermedad , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Ratas Sprague-Dawley , Transducción de Señal , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología , Taquicardia Ventricular/fisiopatología , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/patología , Fibrilación Ventricular/fisiopatología , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/patología , Complejos Prematuros Ventriculares/fisiopatología
15.
Chin Med J (Engl) ; 130(14): 1639-1647, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28685712

RESUMEN

BACKGROUND: Pharmacological therapy for congestive heart failure (CHF) with ventricular arrhythmia is limited. In the study, our aim was to evaluate the effects of Chinese traditional medicine Shensong Yangxin capsules (SSYX) on heart rhythm and function in CHF patients with frequent ventricular premature complexes (VPCs). METHODS: This double-blind, placebo-controlled, multicenter study randomized 465 CHF patients with frequent VPCs to the SSYX (n = 232) and placebo groups (n = 233) for 12 weeks of treatment. The primary endpoint was the VPCs monitored by a 24-h ambulatory electrocardiogram. The secondary endpoints included the left ventricular ejection fraction (LVEF), left ventricular end-diastolic diameter, N-terminal pro-brain natriuretic peptide (NT-proBNP), New York Heart Association (NYHA) classification, 6-min walking distance (6MWD), Minnesota Living with Heart Failure Questionnaire (MLHFQ) scores, and composite cardiac events (CCEs). RESULTS: The clinical characteristics were similar at baseline. SSYX caused a significantly greater decline in the total number of VPCs than the placebo did (-2145 ± 2848 vs. -841 ± 3411, P < 0.05). The secondary endpoints of the LVEF, NYHA classification, NT-proBNP, 6MWD, and MLHFQ scores showed a greater improvements in the SSYX group than in the placebo group (ΔLVEF at 12th week: 4.75 ± 7.13 vs. 3.30 ± 6.53; NYHA improvement rate at the 8th and 12th week: 32.6% vs. 21.8%, 40.5% vs. 25.7%; mean level of NT-proBNP in patients with NT-proBNP ≥125 pg/ml at 12th week: -122 [Q1, Q3: -524, 0] vs. -75 [Q1, Q3: -245, 0]; Δ6MWD at 12th week: 35.1 ± 38.6 vs. 17.2 ± 45.6; ΔMLHFQ at the 4th, 8th, and 12th week: -4.24 ± 6.15 vs. -2.31 ± 6.96, -8.19 ± 8.41 vs. -3.25 ± 9.40, -10.60 ± 9.41 vs. -4.83 ± 11.23, all P < 0.05). CCEs were not different between the groups during the study period. CONCLUSIONS: In this 12-week pilot study, SSYX was demonstrated to have the benefits of VPCs suppression and cardiac function improvement with good compliance on a background of standard treatment for CHF. TRIAL REGISTRATION: www.chictr.org.cn, ChiCTR-TRC-12002061 (http://www.chictr.org.cn/showproj.aspx?proj=7487) and Clinicaltrials.gov, NCT01612260 (https://clinicaltrials.gov/ct2/show/NCT01612260).


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Medicina Tradicional China/métodos , Complejos Prematuros Ventriculares/tratamiento farmacológico , Adolescente , Adulto , Anciano , Método Doble Ciego , Medicamentos Herbarios Chinos/uso terapéutico , Femenino , Insuficiencia Cardíaca/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Función Ventricular Izquierda/efectos de los fármacos , Complejos Prematuros Ventriculares/metabolismo , Adulto Joven
16.
Am J Physiol Heart Circ Physiol ; 313(3): H620-H630, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28646025

RESUMEN

Hypertrophic cardiomyopathy (HCM) is one of the most common genetic cardiac diseases and among the leading causes of sudden cardiac death (SCD) in the young. The cellular mechanisms leading to SCD in HCM are not well known. Prolongation of the action potential (AP) duration (APD) is a common feature predisposing hypertrophied hearts to SCD. Previous studies have explored the roles of inward Na+ and Ca2+ in the development of HCM, but the role of repolarizing K+ currents has not been defined. The objective of this study was to characterize the arrhythmogenic phenotype and cellular electrophysiological properties of mice with HCM, induced by myosin-binding protein C (MyBPC) knockout (KO), and to test the hypothesis that remodeling of repolarizing K+ currents causes APD prolongation in MyBPC KO myocytes. We demonstrated that MyBPC KO mice developed severe hypertrophy and cardiac dysfunction compared with wild-type (WT) control mice. Telemetric electrocardiographic recordings of awake mice revealed prolongation of the corrected QT interval in the KO compared with WT control mice, with overt ventricular arrhythmias. Whole cell current- and voltage-clamp experiments comparing KO with WT mice demonstrated ventricular myocyte hypertrophy, AP prolongation, and decreased repolarizing K+ currents. Quantitative RT-PCR analysis revealed decreased mRNA levels of several key K+ channel subunits. In conclusion, decrease in repolarizing K+ currents in MyBPC KO ventricular myocytes contributes to AP and corrected QT interval prolongation and could account for the arrhythmia susceptibility.NEW & NOTEWORTHY Ventricular myocytes isolated from the myosin-binding protein C knockout hypertrophic cardiomyopathy mouse model demonstrate decreased repolarizing K+ currents and action potential and QT interval prolongation, linking cellular repolarization abnormalities with arrhythmia susceptibility and the risk for sudden cardiac death in hypertrophic cardiomyopathy.


Asunto(s)
Proteínas Portadoras/metabolismo , Frecuencia Cardíaca , Miocitos Cardíacos/metabolismo , Canales de Potasio/metabolismo , Potasio/metabolismo , Taquicardia Ventricular/metabolismo , Complejos Prematuros Ventriculares/metabolismo , Potenciales de Acción , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Electrocardiografía Ambulatoria , Fibrosis , Predisposición Genética a la Enfermedad , Cinética , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , Contracción Miocárdica , Miocitos Cardíacos/patología , Técnicas de Placa-Clamp , Fenotipo , Canales de Potasio/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/patología , Taquicardia Ventricular/fisiopatología , Telemetría , Complejos Prematuros Ventriculares/genética , Complejos Prematuros Ventriculares/patología , Complejos Prematuros Ventriculares/fisiopatología
17.
Am J Physiol Heart Circ Physiol ; 311(6): H1470-H1484, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27765749

RESUMEN

The occurrence of early afterdepolarizations (EADs) and increased dispersion of repolarization are two known factors for arrhythmogenesis in long QT syndrome. However, increased dispersion of repolarization tends to suppress EADs due to the source-sink effect, and thus how the two competing factors cause initiation of arrhythmias remains incompletely understood. Here we used optical mapping and computer simulation to investigate the mechanisms underlying spontaneous initiation of arrhythmias in type 2 long QT (LQT2) syndrome. In optical mapping experiments of transgenic LQT2 rabbit hearts under isoproterenol, premature ventricular complexes (PVCs) were observed to originate from the steep spatial repolarization gradient (RG) regions and propagated unidirectionally. The same PVC behaviors were demonstrated in computer simulations of tissue models of rabbits. Depending on the heterogeneities, these PVCs could lead to either repetitive focal excitations or reentry without requiring an additional vulnerable substrate. Systematic simulations showed that cellular phase 2 EADs were either suppressed or confined to the long action potential region due to the source-sink effect. Tissue-scale phase 3 EADs and PVCs occurred due to tissue-scale dynamical instabilities caused by RG and enhanced L-type calcium current (ICa,L), occurring under both large and small RG. Presence of cellular EADs was not required but potentiated PVCs when RG was small. We also investigated how other factors affect the dynamical instabilities causing PVCs. Our main conclusion is that tissue-scale dynamical instabilities caused by RG and enhanced ICa,L give rise to both the trigger and the vulnerable substrate simultaneously for spontaneous initiation of arrhythmias in LQT2 syndrome.


Asunto(s)
Sistema de Conducción Cardíaco/fisiopatología , Síndrome de QT Prolongado/fisiopatología , Complejos Prematuros Ventriculares/fisiopatología , Agonistas Adrenérgicos beta/toxicidad , Animales , Animales Modificados Genéticamente , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Calcio/metabolismo , Simulación por Computador , Sistema de Conducción Cardíaco/metabolismo , Isoproterenol/toxicidad , Síndrome de QT Prolongado/metabolismo , Miocardio/metabolismo , Conejos , Complejos Prematuros Ventriculares/inducido químicamente , Complejos Prematuros Ventriculares/metabolismo
18.
Heart Rhythm ; 13(3): 743-52, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26538326

RESUMEN

BACKGROUND: In a canine model of premature ventricular contraction-induced cardiomyopathy (PVC-CM), Cav1.2 is downregulated and misplaced from transverse tubules (T tubules). Junctophilin-2 (JPH-2) is also downregulated. OBJECTIVES: The objectives of this study were to understand the role of JPH-2 in PVC-CM and to probe changes in other proteins involved in dyad structure and function. METHODS: We quantify T-tubule contents (di-8-ANEPPS fluorescence in live myocytes), examine myocyte ultrastructures (electron microscopy), probe JPH-2-interacting proteins (co-immunoprecipitation), quantify dyad and nondyad protein levels (immunoblotting), and examine subcellular distributions of dyad proteins (immunofluorescence/confocal microscopy). We also test direct JPH-2 modulation of channel function (vs indirect modulation through dyad formation) using heterologous expression. RESULTS: PVC myocytes have reduced T-tubule contents but otherwise normal ultrastructures. Among 19 proteins examined, only JPH-2, bridging integrator-1 (BIN-1), and Cav1.2 are highly downregulated in PVC hearts. However, statistical analysis indicates a general reduction in dyad protein levels when JPH-2 is downregulated. Furthermore, several dyad proteins, including Na/Ca exchanger, are missing or shifted from dyads to the peripheral surface in PVC myocytes. JPH-2 directly or indirectly interacts with Cai-handling proteins, Cav1.2 and KCNQ1, although not BIN-1 or other scaffolding proteins tested. Expression in mammalian cells that do not have dyads confirms direct JPH-2 modulation of the L-type Ca channel current (Cav1.2/voltage-gated Ca channel ß subunit 2) and slow delayed rectifier current (KCNQ1/KCNE1). CONCLUSION: JPH-2 is more than a "dyad glue": it can modulate Cai handling and ion channel function in the dyad region. Downregulation of JPH-2, BIN-1, and Cav1.2 plays a deterministic role in PVC-CM. Dissecting the hierarchical relationship among the three is necessary for the design of therapeutic interventions to prevent the progression of PVC-CM.


Asunto(s)
Cardiomiopatías/metabolismo , Proteínas de la Membrana/biosíntesis , Miocardio/metabolismo , Intercambiador de Sodio-Calcio/biosíntesis , Complejos Prematuros Ventriculares/metabolismo , Animales , Cardiomiopatías/etiología , Cardiomiopatías/patología , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Immunoblotting , Microscopía Confocal , Microscopía Electrónica , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Complejos Prematuros Ventriculares/complicaciones , Complejos Prematuros Ventriculares/patología
19.
Ter Arkh ; 86(1): 10-7, 2014.
Artículo en Ruso | MEDLINE | ID: mdl-24754063

RESUMEN

AIM: To study the specific features of an immune response and the role of infectious pathogens in the occurrence, development, and maintenance of ventricular ectopic activity in patients without signs of organic disease of the cardiovascular system (CVS). SUBJECTS AND METHODS: The investigation enrolled 91 patients (27 men and 64 women with a mean age of 36.5 +/- 11.5 years) with premature ventricular contractions (PVC) without signs of organic CVS pathology. A control group comprised 31 healthy volunteers. In addition to standard physical examination, a study of N-terminal pro-brain natriuretic peptide (NT-proBNP) and cellular and humoral immune parameters was performed and the serological and molecular genetic markers of infections were determined in all the patients. RESULTS: The persons with PVC, as compared to the control group, were recorded to have a higher NT-proBNP level (49.2 pg/ml versus 25.4 pg/ml; p = 0.001) accompanied by an increase in both the total number of PVC and the number of coupled PVC and the episodes of ventricular tachycardia. They were also found to have elevated CD3(+)HLA-DR+ (2.4% versus 1.56%) and CD3(+)CD95+ (27.6% versus 18.8%) counts (p = 0.001). In patients with a C-reactive protein level of more than 2 mg/l, the total number of PVC was larger than that in the other patients (p = 0.065). The patients with PVC did not show a statistically significant difference from the controls in the levels of antiviral and antibacterial antibodies. The people with PVC displayed a number of relationships between the infectious pathogen antibody titers and the ECG Holter monitoring and echocardiography readings. CONCLUSION: In the patients with PVC without signs of organic CVS pathology, the parameters of an immune response were not different from those obtained in the control group, which was likely to be associated with the involvement of the immune system in the development and maintenance of ventricular arrhythmias.


Asunto(s)
Anticuerpos/inmunología , Proteína C-Reactiva/metabolismo , Electrocardiografía , Inmunidad Celular , Péptido Natriurético Encefálico/metabolismo , Fragmentos de Péptidos/metabolismo , Complejos Prematuros Ventriculares/inmunología , Adulto , Femenino , Humanos , Masculino , Índice de Severidad de la Enfermedad , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología
20.
J Cardiovasc Pharmacol ; 64(2): 127-33, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24662492

RESUMEN

OBJECTIVES: To investigate the effects and mechanisms of Nardostachys chinensis (NC) on spontaneous ventricular arrhythmias in rats with hyper-acute myocardial infarction (AMI). METHODS: Seventy-two rats were randomly divided into the control group (n = 24), metoprolol group (n = 24), and the NC group (n = 24). Premature ventricular contractions (PVCs), ventricular tachycardias (VTs), ventricular fibrillations (VFs), and blood pressure were monitored for 4 hours after coronary artery ligation. The connexin 43 (Cx43) expression in ventricular myocardium was measured by immunohistochemistry, Western blot, and real-time RT-PCR. RESULTS: Compared with the control, metoprolol and NC decreased the VF incidence (50% vs. 4.2%, P < 0.001, and 50% vs. 12.5%, P = 0.005, respectively). There was a steady decrease in the cumulative number of PVCs and VTs within 4 hours from ligating in 3 groups. Compared with the control, metoprolol and NC reduced the cumulative number of VTs and PVCs. Compared with control, metoprolol and NC decreased the infarct size of the left ventricular tissue (55.98% ± 6.20% vs. 39.13% ± 4.53%, P < 0.001, and 55.98% ± 6.20% vs. 42.39% ± 3.44%, P < 0.001, respectively). The results from immunohistochemistry, Western blot, and real-time RT-PCR showed that the protein expression of Cx43 in the control group was significantly lower than that in the metoprolol and NC groups in the infarcted zone. CONCLUSIONS: NC decreased the incidence of spontaneous ventricular arrhythmias (especially VF), reduced Cx43 degradation, and improved Cx43 redistribution in myocardial infarcted zone in rats with hyper-AMI. The data of the present study indicated that NC may be a promising drug in the future to prevent patients with AMI from lethal ventricular arrhythmias in prehospital setting.


Asunto(s)
Antiarrítmicos/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Nardostachys/química , Taquicardia Ventricular/prevención & control , Fibrilación Ventricular/prevención & control , Complejos Prematuros Ventriculares/prevención & control , Animales , Antiarrítmicos/administración & dosificación , Presión Sanguínea/efectos de los fármacos , Western Blotting , Conexina 43/metabolismo , Medicamentos Herbarios Chinos/administración & dosificación , Electrocardiografía , Femenino , Inmunohistoquímica , Masculino , Metoprolol/administración & dosificación , Metoprolol/uso terapéutico , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Rizoma/química , Taquicardia Ventricular/etiología , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología , Fibrilación Ventricular/etiología , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/patología , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...