Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Am Coll Cardiol ; 61(11): 1192-201, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23395072

RESUMEN

OBJECTIVES: This study sought to test the hypothesis that hyperpolarization-activated cyclic nucleotide-gated (HCN)-based biological pacing might be improved significantly by hyperpolarizing the action potential (AP) threshold via coexpression of the skeletal muscle sodium channel 1 (SkM1). BACKGROUND: Gene-based biological pacemakers display effective in vivo pacemaker function. However, approaches used to date have failed to manifest optimal pacemaker properties, defined as basal beating rates of 60 to 90 beats/min, a brisk autonomic response achieving maximal rates of 130 to 160 beats/min, and low to absent electronic backup pacing. METHODS: We implanted adenoviral SkM1, HCN2, or HCN2/SkM1 constructs into left bundle branches (LBB) or left ventricular (LV) epicardium of atrioventricular-blocked dogs. RESULTS: During stable peak gene expression on days 5 to 7, HCN2/SkM1 LBB-injected dogs showed highly stable in vivo pacemaker activity superior to SkM1 or HCN2 alone and superior to LV-implanted dogs with regard to beating rates (resting approximately 80 beats/min; maximum approximately 130 beats/min), no dependence on electronic backup pacing, and enhanced modulation of pacemaker function during circadian rhythm or epinephrine infusion. In vitro isolated LV of dogs overexpressing SkM1 manifested a significantly more negative AP threshold. CONCLUSIONS: LBB-injected HCN2/SkM1 potentially provides a more clinically suitable biological pacemaker strategy than other reported constructs. This superiority is attributable to the more negative AP threshold and injection into the LBB.


Asunto(s)
Técnicas de Transferencia de Gen , Sistema de Conducción Cardíaco , Frecuencia Cardíaca/fisiología , Canales Iónicos/genética , Proteínas Musculares/genética , Animales , Perros , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización
2.
Heart Rhythm ; 9(11): 1865-72, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22820054

RESUMEN

BACKGROUND: Left ventricular pacing (LVP) in canine heart alters ventricular activation, leading to reduced transient outward potassium current (I(to)), loss of the epicardial action potential notch, and T-wave vector displacement. These repolarization changes, referred to as cardiac memory, are initiated by locally increased angiotensin II (AngII) levels. In HEK293 cells in which Kv4.3 and KChIP2, the channel subunits contributing to I(to), are overexpressed with the AngII receptor 1 (AT1R), AngII induces a decrease in I(to) as the result of internalization of a Kv4.3/KChIP2/AT1R macromolecular complex. OBJECTIVE: To test the hypothesis that in canine heart in situ, 2h LVP-induced decreases in membrane KChIP2, AT1R, and I(to) are prevented by blocking subunit trafficking. METHODS: We used standard electrophysiological, biophysical, and biochemical methods to study 4 groups of dogs: (1) Sham, (2) 2h LVP, (3) LVP + colchicine (microtubule-disrupting agent), and (4) LVP + losartan (AT1R blocker). RESULTS: The T-wave vector displacement was significantly greater in LVP than in Sham and was inhibited by colchicine or losartan. Epicardial biopsies showed significant decreases in KChIP2 and AT1R proteins in the membrane fraction after LVP but not after sham treatment, and these decreases were prevented by colchicine or losartan. Colchicine but not losartan significantly reduced microtubular polymerization. In isolated ventricular myocytes, AngII-induced I(to) reduction and loss of action potential notch were blocked by colchicine. CONCLUSIONS: LVP-induced reduction of KChIP2 in plasma light membranes depends on an AngII-mediated pathway and intact microtubular status. Loss of I(to) and the action potential notch appear to derive from AngII-initiated trafficking of channel subunits.


Asunto(s)
Estimulación Cardíaca Artificial , Sistema de Conducción Cardíaco/fisiología , Losartán/farmacología , Microtúbulos/metabolismo , Canales de Potasio/fisiología , Receptores de Angiotensina/metabolismo , Adaptación Fisiológica/fisiología , Análisis de Varianza , Animales , Biopsia , Western Blotting , Colchicina/farmacología , Perros , Sistema de Conducción Cardíaco/efectos de los fármacos , Proteínas de Interacción con los Canales Kv/metabolismo , Masculino , Técnicas de Placa-Clamp , Canales de Potasio/efectos de los fármacos
3.
Circulation ; 126(5): 528-36, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22753192

RESUMEN

BACKGROUND: Biological pacing performed solely via HCN2 gene transfer in vivo results in relatively slow idioventricular rates and only moderate autonomic responsiveness. We induced biological pacing using the Ca(2+)-stimulated adenylyl cyclase AC1 gene expressed alone or in combination with HCN2 and compared outcomes with those with single-gene HCN2 transfer. METHODS AND RESULTS: We implanted adenoviral HCN2, AC1, or HCN2/AC1 constructs into the left bundle branches of atrioventricular-blocked dogs. During steady-state gene expression (days 5-7), differences between AC1, HCN2/AC1, and HCN2 alone were evident in basal beating rate, escape time, and dependence on electronic backup pacing. In HCN2, AC1, and HCN2/AC1, these parameters were as follows: basal beating rate: 50±1.5, 60±5.0, and 129±28.9 bpm (P<0.05 for HCN2/AC1 versus HCN2 or AC1 alone), respectively; escape time: 2.4±0.2, 1.3±0.2, and 1.1±.0.4 seconds (P<0.05 for AC1 and HCN2/AC1 versus HCN2); and percent electronic beats: 34±8%, 2±1%, and 6±2% (P<0.05 for AC1 and HCN2/AC1 versus HCN2). Instantaneous (SD1) and long-term (SD2) heart rate variability and circadian rhythm analyzed via 24-hour Holter recordings showed a shift toward greater sensitivity to parasympathetic modulation in animals injected with AC1 and a high degree of sympathetic modulation in animals injected with HCN2/AC1. CONCLUSION: AC1 or HCN2/AC1 overexpression in left bundle branches provides highly efficient biological pacing and greater sensitivity to autonomic modulation than HCN2 alone.


Asunto(s)
Adenilil Ciclasas/genética , Adenilil Ciclasas/fisiología , Bloqueo Atrioventricular/terapia , Terapia Genética , Sistema de Conducción Cardíaco/fisiología , Canales Iónicos/genética , Canales Iónicos/fisiología , Adenoviridae/genética , Animales , Bloqueo Atrioventricular/etiología , Benzazepinas/farmacología , Ablación por Catéter/efectos adversos , Ritmo Circadiano/fisiología , Perros , Electrocardiografía , Técnicas de Transferencia de Gen , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ivabradina , Modelos Animales , Bloqueadores de los Canales de Potasio/farmacología
4.
Circ Arrhythm Electrophysiol ; 5(4): 831-40, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22722661

RESUMEN

BACKGROUND: In depolarized myocardial infarct epicardial border zones, the cardiac sodium channel is largely inactivated, contributing to slow conduction and reentry. We have demonstrated that adenoviral delivery of the skeletal muscle Na(+) channel (SkM1) to epicardial border zones normalizes conduction and reduces induction of ventricular tachycardia/ventricular fibrillation. We now studied the impact of canine mesenchymal stem cells (cMSCs) in delivering SkM1. METHODS AND RESULTS: cMSCs were isolated and transfected with SkM1. Coculture experiments showed cMSC/SkM1 but not cMSC alone and maintained fast conduction at depolarized potentials. We studied 3 groups in the canine 7d infarct: sham, cMSC, and cMSC/SkM1. In vivo epicardial border zones electrograms were broad and fragmented in sham, narrower in cMSCs, and narrow and unfragmented in cMSC/SkM1 (P<0.05). During programmed electrical stimulation of epicardial border zones, QRS duration in cMSC/SkM1 was shorter than in cMSC and sham (P<0.05). Programmed electrical stimulation-induced ventricular tachycardia/ventricular fibrillation was equivalent in all groups (P>0.05). CONCLUSION: cMSCs provide efficient delivery of SkM1 current. The interventions performed (cMSCs or cMSC/SkM1) were neither antiarrhythmic nor proarrhythmic. Comparing outcomes with cMSC/SkM1 and viral gene delivery highlights the criticality of the delivery platform to SkM1 antiarrhythmic efficacy.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Proteínas Musculares/metabolismo , Infarto del Miocardio/cirugía , Miocitos Cardíacos/metabolismo , Canales de Sodio/metabolismo , Sodio/metabolismo , Taquicardia Ventricular/prevención & control , Fibrilación Ventricular/prevención & control , Potenciales de Acción , Animales , Animales Recién Nacidos , Estimulación Cardíaca Artificial , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Perros , Técnicas Electrofisiológicas Cardíacas , Humanos , Proteínas Musculares/genética , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.5 , Ratas , Ratas Sprague-Dawley , Canales de Sodio/genética , Taquicardia Ventricular/etiología , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Transfección , Fibrilación Ventricular/etiología , Fibrilación Ventricular/genética , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/fisiopatología
5.
Heart Rhythm ; 7(12): 1835-40, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20708103

RESUMEN

BACKGROUND: Biological pacemakers based on the HCN2 channel isoform respond to beta-adrenergic and muscarinic stimulation, suggesting a capacity to respond to autonomic input. OBJECTIVE: The purpose of this study was to investigate autonomic response to emotional arousal in canines implanted with murine HCN2-based biological pacemakers using gene therapy. METHODS: An electronic pacemaker was implanted with its lead in the right ventricular apical endocardium (VVI 35 bpm). An adenoviral HCN2/GFP construct (Ad-HCN2, n = 7) or saline (control, n = 5) was injected into the left bundle branch on day 2 after radiofrequency ablation of the atrioventricular node to induce complete atrioventricular block. Emotional arousal was achieved by presenting food following an overnight fast. Autonomic control was evaluated with Poincaré plots of R-R(N) against R-R(N+1) intervals to characterize heart rate variability (HRV) and with continuous RR interval assessment via 24-hour ambulatory ECG. The 24-hour ECG and Poincaré plot shape were analyzed. RESULTS: During day 1 after biological pacemaker implantation, Poincaré HRV parameters and RR intervals were unchanged with food presentation. However, on day 7, food presentation was accompanied by an increase in HRV (SD1, p < 0.07, and SD2, p < 0.05) and shortening of RR interval (P < .05) in dogs with Ad-HCN2 but not in controls. CONCLUSION: This is the first demonstration that biological pacemakers are capable of responding to natural arousal stimuli to elicit appropriate chronotropic responses, a potential advantage over electronic pacemakers.


Asunto(s)
Nivel de Alerta/fisiología , Relojes Biológicos/fisiología , Emociones/fisiología , Adenoviridae/genética , Animales , Bloqueo Atrioventricular/fisiopatología , Bloqueo Atrioventricular/terapia , Perros , Electrocardiografía , Terapia Genética , Proteínas Fluorescentes Verdes , Frecuencia Cardíaca/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/fisiología , Sustancias Luminiscentes , Marcapaso Artificial
6.
Heart Rhythm ; 7(8): 1104-10, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20385252

RESUMEN

BACKGROUND: Skeletal muscle sodium channel (Nav1.4) expression in border zone myocardium increases action potential upstroke velocity in depolarized isolated tissue. Because resting membrane potential in the 1-week canine infarct is reduced, we hypothesized that conduction velocity (CV) is greater in Nav1.4 dogs compared with in control dogs. OBJECTIVE: The purpose of this study was to measure CV in the infarct border zone border in dogs with and without Nav1.4 expression. METHODS: Adenovirus was injected in the infarct border zone in 34 dogs. The adenovirus incorporated the Nav1.4- and a green fluorescent protein (GFP) gene (Nav1.4 group, n = 16) or only GFP (n = 18). After 1 week, upstroke velocity and CV were measured by sequential microelectrode recordings at 4 and 7 mM [K(+)] in superfused epicardial slabs. High-density in vivo epicardial activation mapping was performed in a subgroup (8 Nav1.4, 6 GFP) at three to four locations in the border zone. Microscopy and antibody staining confirmed GFP or Nav1.4 expression. RESULTS: Infarct sizes were similar between groups (30.6% +/- 3% of left ventricle mass, mean +/- standard error of the mean). Longitudinal CV was greater in Nav1.4 than in GFP sites (58.5 +/- 1.8 vs. 53.3 +/- 1.2 cm/s, 20 and 15 sites, respectively; P <.05). Transverse CV was not different between the groups. In tissue slabs, dV/dt(max) was higher and CV was greater in Nav1.4 than in control at 7 mM [K(+)] (P <.05). Immunohistochemical Nav1.4 staining was seen at the longitudinal ends of the myocytes. CONCLUSION: Nav1.4 channels in myocardium surviving 1 week infarction increases longitudinal but not transverse CV, consistent with the increased dV/dt(max) and with the cellular localization of Nav1.4.


Asunto(s)
Sistema de Conducción Cardíaco/fisiopatología , Músculo Esquelético/fisiología , Infarto del Miocardio/fisiopatología , Canales de Sodio/biosíntesis , Potenciales de Acción , Animales , Modelos Animales de Enfermedad , Perros
7.
Heart Rhythm ; 7(7): 964-70, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20346417

RESUMEN

BACKGROUND: Left ventricular pacing (LVP) to induce cardiac memory (CM) in dogs results in a decreased transient outward K current (I(to)) and reduced mRNA and protein of the I(to) channel accessory subunit, KChIP2. The KChIP2 decrease is attributed to a decrease in its transcription factor, cyclic adenosine monophosphate response element binding protein (CREB). OBJECTIVE: This study sought to determine the mechanisms responsible for the CREB decrease that is initiated by LVP. METHODS: CM was quantified as T-wave vector displacement in 18 LVP dogs. In 5 dogs, angiotensin II receptor blocker, saralasin, was infused before and during pacing. In 3 dogs, proteasomal inhibitor, lactacystin, was injected into the left anterior descending artery before LVP. Epicardial biopsy samples were taken before and after LVP. Neonatal rat cardiomyocytes (NRCM) were incubated with H(2)O(2) (50 micromol/l) for 1 hour with or without lactacystin. RESULTS: LVP significantly displaced the T-wave vector and was associated with increased lipid peroxidation and increased tissue angiotensin II levels. Saralasin prevented T-vector displacement and lipid peroxidation. CREB was significantly decreased after 2 hours of LVP and was comparably decreased in H(2)O(2)-treated NRCM. Lactacystin inhibited the CREB decrease in LVP dogs and H(2)O(2)-treated NRCM. LVP and H(2)O(2) both induced CREB ubiquitination, and the H(2)O(2)-induced CREB decrease was prevented by knocking down ubiquitin. CONCLUSION: LVP initiates myocardial angiotensin II production and reactive oxygen species synthesis, leading to CREB ubiquitination and its proteasomal degradation. This sequence of events would explain the pacing-induced reduction in KChIP2, and contribute to altered repolarization and the T-wave changes of cardiac memory.


Asunto(s)
Estimulación Cardíaca Artificial , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Proteínas de Interacción con los Canales Kv/análisis , Función Ventricular Izquierda/fisiología , Potenciales de Acción/fisiología , Angiotensina II/fisiología , Animales , Arritmias Cardíacas/metabolismo , Western Blotting , Células Cultivadas , Perros , Canales Iónicos/fisiología , Peroxidación de Lípido , Masculino , Modelos Animales , Modelos Cardiovasculares , Miocardio/metabolismo , Miocitos Cardíacos/citología , Estrés Oxidativo/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ubiquitina/fisiología , Ubiquitinación , Remodelación Ventricular/fisiología
8.
Circulation ; 119(1): 19-27, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19103989

RESUMEN

BACKGROUND: In depolarized myocardial infarct epicardial border zones, the cardiac sodium channel (SCN5A) is largely inactivated, contributing to low action potential upstroke velocity (V(max)), slow conduction, and reentry. We hypothesized that a fast inward current such as the skeletal muscle sodium channel (SkM1) operating more effectively at depolarized membrane potentials might restore fast conduction in epicardial border zones and be antiarrhythmic. METHODS AND RESULTS: Computer simulations were done with a modified Hund-Rudy model. Canine myocardial infarcts were created by coronary ligation. Adenovirus expressing SkM1 and green fluorescent protein or green fluorescent protein alone (sham) was injected into epicardial border zones. After 5 to 7 days, dogs were studied with epicardial mapping, programmed premature stimulation in vivo, and cellular electrophysiology in vitro. Infarct size was determined, and tissues were immunostained for SkM1 and green fluorescent protein. In the computational model, modest SkM1 expression preserved fast conduction at potentials as positive as -60 mV; overexpression of SCN5A did not. In vivo epicardial border zone electrograms were broad and fragmented in shams (31.5 +/- 2.3 ms) and narrower in SkM1 (22.6 +/- 2.8 ms; P=0.03). Premature stimulation induced ventricular tachyarrhythmia/fibrillation >60 seconds in 6 of 8 shams versus 2 of 12 SkM1 (P=0.02). Microelectrode studies of epicardial border zones from SkM1 showed membrane potentials equal to that of shams and V(max) greater than that of shams as membrane potential depolarized (P<0.01). Infarct sizes were similar (sham, 30 +/- 2.8%; SkM1, 30 +/- 2.6%; P=0.86). SkM1 expression in injected epicardium was confirmed immunohistochemically. CONCLUSIONS: SkM1 increases V(max) of depolarized myocardium and reduces the incidence of inducible sustained ventricular tachyarrhythmia/fibrillation in canine infarcts. Gene therapy to normalize activation by increasing V(max) at depolarized potentials may be a promising antiarrhythmic strategy.


Asunto(s)
Terapia Genética/métodos , Sistema de Conducción Cardíaco/fisiología , Modelos Cardiovasculares , Canales de Sodio/genética , Taquicardia Ventricular/fisiopatología , Taquicardia Ventricular/terapia , Potenciales de Acción/fisiología , Adenoviridae/genética , Animales , Línea Celular , Simulación por Computador , Modelos Animales de Enfermedad , Perros , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Humanos , Técnicas In Vitro , Riñón/citología , Masculino , Músculo Esquelético/fisiología , Contracción Miocárdica/fisiología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Canal de Sodio Activado por Voltaje NAV1.5 , Penicilina G/metabolismo , Pericardio/fisiología , Canales de Sodio/metabolismo , Canales de Sodio/fisiología , Taquicardia Ventricular/patología
9.
Heart Rhythm ; 5(2): 282-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18242555

RESUMEN

BACKGROUND: A potential concern about biological pacemakers is their possible malfunction, which might create ventricular tachycardias (VTs). OBJECTIVE: The purpose of this study was to test our hypothesis that should VTs complicate implantation of HCN-channel-based biological pacemakers, they would be suppressed by inhibitors of the pacemaker current, I(f). METHODS: We created a chimeric channel (HCN212) containing the N- and C-termini of mouse HCN2 and the transmembrane region of mouse HCN1 and implanted it in HEK293 cells. Forty-eight hours later, in whole-cell patch clamp recordings, mean steady state block induced by 3 microM ivabradine (IVB) showed HCN1 = HCN212 > HCN2 currents. The HCN212 adenoviral construct was then implanted into the canine left bundle branch in 11 dogs. Complete AV block was created via radiofrequency ablation, and a ventricular demand electronic pacemaker was implanted (VVI 45 bpm). Electrocardiogram, 24-hour Holter monitoring, and pacemaker log record check were performed for 11 days. RESULTS: All dogs developed rapid VT (>120 bpm, maximum rate = 285 +/- 37 bpm) at 0.9 +/- 0.3 days after implantation that persisted through 5 +/- 1 days. IVB, 1 mg/kg over 5 minutes, was administered during rapid VT, and three dogs received a second dose 24 hours later. While VT terminated with IBV in all instances within 3.4 +/- 0.6 minutes, no effect of IVB on sinus rate was noted. CONCLUSION: We conclude that (1) I(f)-associated tachyarrhythmias-if they occur with HCN-based biological pacemakers-can be controlled with I(f)-inhibiting drugs such as IVB; (2) in vitro, IVB appears to have a greater steady state inhibiting effect on HCN1 and HCN212 isoforms than on HCN4; and (3) VT originating from the HCN212 injection site is suppressed more readily than sinus rhythm. This suggests a selectivity of IVB at the concentration attained for ectopic over HCN4-based pacemaker function. This might confer a therapeutic benefit.


Asunto(s)
Benzazepinas/farmacología , Canales de Calcio , Estimulación Cardíaca Artificial , Fármacos Cardiovasculares/farmacología , Desfibriladores Implantables , Taquicardia Ventricular/fisiopatología , Taquicardia Ventricular/terapia , Animales , Ablación por Catéter , Perros , Electrofisiología , Ivabradina , Masculino , Células Musculares , Ratas , Factores de Riesgo , Taquicardia Ventricular/tratamiento farmacológico
10.
Circulation ; 116(7): 706-13, 2007 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-17646577

RESUMEN

BACKGROUND: Biological pacemaking has been performed with viral vectors, human embryonic stem cells, and adult human mesenchymal stem cells (hMSCs) as delivery systems. Only with human embryonic stem cells are data available regarding stability for >2 to 3 weeks, and here, immunosuppression has been used to facilitate survival of xenografts. The purpose of the present study was to determine whether hMSCs provide stable impulse initiation over 6 weeks without the use of immunosuppression, the "dose" of hMSCs that ensures function over this period, and the catecholamine responsiveness of hMSC-packaged pacemakers. METHODS AND RESULTS: A full-length mHCN2 cDNA subcloned in a pIRES2-EGFP vector was electroporated into hMSCs. Transfection efficiency was estimated by GFP expression. I(HCN2) was measured with patch clamp, and cells were administered into the left ventricular anterior wall of adult dogs in complete heart block and with backup electronic pacemakers. Studies encompassed 6 weeks. I(HCN2) for all cells was 32.1+/-1.3 pA/pF (mean+/-SE) at -150 mV. Pacemaker function in intact dogs required 10 to 12 days to fully stabilize and persisted consistently through day 42 in dogs receiving > or =700,000 hMSCs (approximately 40% of which carried current). Rhythms were catecholamine responsive. Tissues from animals killed at 42 days manifested neither apoptosis nor humoral or cellular rejection. CONCLUSIONS: hMSCs provide a means for administering catecholamine-responsive biological pacemakers that function stably for 6 weeks and manifest no cellular or humoral rejection at that time. Cell doses >700,000 are sufficient for pacemaking when administered to left ventricular myocardium.


Asunto(s)
Corazón/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Adulto , Animales , Células Cultivadas , Perros , Conductividad Eléctrica , Electrocardiografía , Epinefrina/farmacología , Bloqueo Cardíaco/fisiopatología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Técnicas de Placa-Clamp , Canales de Potasio , Transfección , Trasplante Heterólogo
11.
Cardiovasc Res ; 68(2): 259-67, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16054122

RESUMEN

OBJECTIVE: Long-term cardiac memory (LTCM), expressed as a specific pattern of T-wave change on ECG, is associated with 1) reduced transient outward potassium current (I(to)), 2) reduced mRNA for the pore-forming protein of I(to), Kv4.3, 3) reduced cAMP response element binding protein (CREB), and 4) diminished binding to its docking site on the DNA, the cAMP response element (CRE). We hypothesized a causal link between the decrease of the transcription factor CREB and down-regulation of I(to) and one of its channel subunits, KChIP2, in LTCM. METHODS: After three weeks of left ventricular pacing to induce LTCM (8 paced, 7 sham control dogs), epicardial KChIP2 mRNA and protein levels were assessed by real-time PCR and Western blotting. Mimicking the CREB down-regulation in LTCM, CREB was knocked down in situ in other dogs using adenoviral anti-sense. Effects on the action potential notch, reflecting I(to), were investigated in situ using monophasic action potential (MAP) recordings and at the cellular level by the whole-cell patch clamp technique. CREB binding in the KChIP2 promoter region was ascertained by electrophoretic mobility-shift assays. RESULTS: In LTCM, epicardial KChIP2 mRNA and protein were reduced by 62% and 76%, respectively, compared to shams (p < 0.05). CREB binding by the canine KChIP2 promoter region was demonstrated. CREB knockdown led to disappearance of the phase1 notch in MAP and ablation of I(to). CONCLUSIONS: These results strengthen the hypothesis that down-regulation of CREB-mediated transcription underlies the attenuation of epicardial I(to) in LTCM. They also emphasize that ventricular pacing exerts effects at a subcellular level contributing to memory and conceivably to other forms of cardiac remodeling.


Asunto(s)
Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Miocardio/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Potenciales de Acción , Animales , Western Blotting/métodos , Proteínas de Unión al Calcio/metabolismo , Estimulación Cardíaca Artificial , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Perros , Regulación hacia Abajo , Electrocardiografía , Ensayo de Cambio de Movilidad Electroforética , Modelos Animales , Oligonucleótidos Antisentido/farmacología , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/análisis , Canales de Potasio con Entrada de Voltaje/genética , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Remodelación Ventricular
12.
Circ Res ; 94(7): 952-9, 2004 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-14988226

RESUMEN

We tested the ability of human mesenchymal stem cells (hMSCs) to deliver a biological pacemaker to the heart. hMSCs transfected with a cardiac pacemaker gene, mHCN2, by electroporation expressed high levels of Cs+-sensitive current (31.1+/-3.8 pA/pF at -150 mV) activating in the diastolic potential range with reversal potential of -37.5+/-1.0 mV, confirming the expressed current as I(f)-like. The expressed current responded to isoproterenol with an 11-mV positive shift in activation. Acetylcholine had no direct effect, but in the presence of isoproterenol, shifted activation 15 mV negative. Transfected hMSCs influenced beating rate in vitro when plated onto a localized region of a coverslip and overlaid with neonatal rat ventricular myocytes. The coculture beating rate was 93+/-16 bpm when hMSCs were transfected with control plasmid (expressing only EGFP) and 161+/-4 bpm when hMSCs were expressing both EGFP+mHCN2 (P<0.05). We next injected 10(6) hMSCs transfected with either control plasmid or mHCN2 gene construct subepicardially in the canine left ventricular wall in situ. During sinus arrest, all control (EGFP) hearts had spontaneous rhythms (45+/-1 bpm, 2 of right-sided origin and 2 of left). In the EGFP+mHCN2 group, 5 of 6 animals developed spontaneous rhythms of left-sided origin (rate=61+/-5 bpm; P<0.05). Moreover, immunostaining of the injected regions demonstrated the presence of hMSCs forming gap junctions with adjacent myocytes. These findings demonstrate that genetically modified hMSCs can express functional HCN2 channels in vitro and in vivo, mimicking overexpression of HCN2 genes in cardiac myocytes, and represent a novel delivery system for pacemaker genes into the heart or other electrical syncytia.


Asunto(s)
Terapia Genética , Canales Iónicos/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Proteínas Musculares/fisiología , Miocitos Cardíacos/fisiología , Acetilcolina/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas/efectos de los fármacos , Células Cultivadas/fisiología , Células Cultivadas/trasplante , Cesio/farmacología , Colinérgicos/farmacología , Técnicas de Cocultivo , Perros , Electroporación , Uniones Comunicantes/fisiología , Uniones Comunicantes/ultraestructura , Frecuencia Cardíaca , Ventrículos Cardíacos/citología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Transporte Iónico/efectos de los fármacos , Isoproterenol/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp , Canales de Potasio , Ratas , Proteínas Recombinantes de Fusión/fisiología , Transfección
13.
Circulation ; 109(4): 506-12, 2004 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-14734518

RESUMEN

BACKGROUND: We hypothesized that administration of the HCN2 gene to the left bundle-branch (LBB) system of intact dogs would provide pacemaker function in the physiological range of heart rates. METHODS AND RESULTS: An adenoviral construct incorporating HCN2 and green fluorescent protein (GFP) as a marker was injected via catheter under fluoroscopic control into the posterior division of the LBB. Controls were injected with an adenoviral construct of GFP alone or saline. Animals were monitored electrocardiographically for up to 7 days after surgery, at which time they were anesthetized and subjected to vagal stimulation to permit emergence of escape pacemakers. Hearts were then removed and injection sites visually identified and removed for microelectrode study of action potentials, patch clamp studies of pacemaker current, and/or immunohistochemical studies of HCN2. For 48 hours postoperatively, 7 of 7 animals subjected to 24-hour ECG monitoring showed multiple ventricular premature depolarizations and/or ventricular tachycardia attributable to injection-induced injury. Thereafter, sinus rhythm prevailed. During vagal stimulation, HCN2-injected dogs showed rhythms originating from the left ventricle, the rate of which was significantly more rapid than in the controls. Excised posterior divisions of the LBB from HCN2-injected animals manifested automatic rates significantly greater than the controls. Isolated tissues showed immunohistochemical and biophysical evidence of overexpressed HCN2. CONCLUSIONS: A gene-therapy approach for induction of biological pacemaker activity within the LBB system provides ventricular escape rhythms that have physiologically acceptable rates. Long-term stability and feasibility of the approach remain to be tested.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Sistema de Conducción Cardíaco , Ventrículos Cardíacos/fisiopatología , Canales Iónicos/genética , Proteínas Musculares/genética , Potenciales de Acción , Adenoviridae/genética , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/terapia , Perros , Estimulación Eléctrica , Electrocardiografía , Femenino , Terapia Genética , Vectores Genéticos , Proteínas Fluorescentes Verdes , Hematoma/etiología , Hematoma/patología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/análisis , Proteínas Luminiscentes/genética , Masculino , Proteínas Musculares/análisis , Miocitos Cardíacos/fisiología , Marcapaso Artificial , Técnicas de Placa-Clamp , Periodicidad , Ramos Subendocárdicos/citología
14.
Circulation ; 107(8): 1106-9, 2003 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-12615786

RESUMEN

BACKGROUND: We hypothesized that localized overexpression of the hyperpolarization-activated, cyclic nucleotide-gated (HCN2) pacemaker current isoform in canine left atrium (LA) would constitute a novel biological pacemaker. METHODS AND RESULTS: Adenoviral constructs of mouse HCN2 and green fluorescent protein (GFP) or GFP alone were injected into LA, terminal studies performed 3 to 4 days later, hearts removed, and myocytes examined for native and expressed pacemaker current (I(f)). Spontaneous LA rhythms occurred after vagal stimulation-induced sinus arrest in 4 of 4 HCN2+GFP dogs and 0 of 3 GFP dogs (P<0.05). Native I(f) in nonexpressed atrial myocytes was 7+/-4 pA at -130 mV (n=5), whereas HCN2+GFP LA had expressed pacemaker current (I(HCN2)) of 3823+/-713 pA at -125 mV (n=10) and 768+/-365 pA at -85 mV. CONCLUSIONS: HCN2 overexpression provides an I(f)-based pacemaker sufficient to drive the heart when injected into a localized region of atrium, offering a promising gene therapy for pacemaker disease.


Asunto(s)
Función Atrial , Atrios Cardíacos/metabolismo , Canales Iónicos/metabolismo , Canales Iónicos/fisiología , Proteínas Musculares/metabolismo , Proteínas Musculares/fisiología , Adenoviridae/genética , Animales , Relojes Biológicos , Células Cultivadas , Perros , Conductividad Eléctrica , Electrocardiografía , Femenino , Vectores Genéticos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/genética , Masculino , Proteínas Musculares/genética , Canales de Potasio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA