Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Ultrasound Med Biol ; 40(1): 177-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24239363

ABSTRACT

Electromechanical wave imaging (EWI) has been show capable of directly and entirely non-invasively mapping the trans mural electromechanical activation in all four cardiac chambers in vivo. In this study, we assessed EWI repeatability and reproducibility, as well as its capability of localizing electronic and, for the first time, biological pacing locations in closed-chest, conscious canines. Electromechanical activation was obtained in six conscious animals during normal sinus rhythm (NSR) and idioventricular rhythms occurring in dogs with complete heart block instrumented with electronic and biologic pacemakers (EPM and BPM respectively). After atrioventricular node ablation, dogs were implanted with an EPM in the right ventricular (RV) endocardial apex (n = 4) and two additionally received a BPM at the left ventricular (LV) epicardial base (n = 2). EWI was performed trans thoracically during NSR, BPM and EPM pacing, in conscious dogs, using an unfocused transmit sequence at 2000 frames/s. During NSR, the EW originated at the right atrium (RA), propagated to the left atrium (LA) and emerged from multiple sources in both ventricles. During EPM, the EW originated at the RV apex and propagated throughout both ventricles. During BPM, the EW originated from the LV basal lateral wall and subsequently propagated throughout the ventricles. EWI differentiated BPM from EPM and NSR and identified the distinct pacing origins. Isochrone comparison indicated that EWI was repeatable and reliable. These findings thus indicate the potential for EWI to serve as a simple, non-invasive and direct imaging technology for mapping and characterizing arrhythmias as well as the treatments thereof.


Subject(s)
Body Surface Potential Mapping/methods , Cardiac Pacing, Artificial , Echocardiography/methods , Excitation Contraction Coupling/physiology , Heart Conduction System/physiology , Heart/physiology , Myocardial Contraction/physiology , Animals , Dogs , Image Interpretation, Computer-Assisted/methods , Male
2.
Circulation ; 126(5): 528-36, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22753192

ABSTRACT

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.


Subject(s)
Adenylyl Cyclases/genetics , Adenylyl Cyclases/physiology , Atrioventricular Block/therapy , Genetic Therapy , Heart Conduction System/physiology , Ion Channels/genetics , Ion Channels/physiology , Adenoviridae/genetics , Animals , Atrioventricular Block/etiology , Benzazepines/pharmacology , Catheter Ablation/adverse effects , Circadian Rhythm/physiology , Dogs , Electrocardiography , Gene Transfer Techniques , Heart Rate/drug effects , Heart Rate/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ivabradine , Models, Animal , Potassium Channel Blockers/pharmacology
3.
Heart Rhythm ; 9(11): 1865-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22820054

ABSTRACT

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.


Subject(s)
Cardiac Pacing, Artificial , Heart Conduction System/physiology , Losartan/pharmacology , Microtubules/metabolism , Potassium Channels/physiology , Receptors, Angiotensin/metabolism , Adaptation, Physiological/physiology , Analysis of Variance , Animals , Biopsy , Blotting, Western , Colchicine/pharmacology , Dogs , Heart Conduction System/drug effects , Kv Channel-Interacting Proteins/metabolism , Male , Patch-Clamp Techniques , Potassium Channels/drug effects
4.
Circ Arrhythm Electrophysiol ; 5(4): 831-40, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22722661

ABSTRACT

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.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Muscle Proteins/metabolism , Myocardial Infarction/surgery , Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Sodium/metabolism , Tachycardia, Ventricular/prevention & control , Ventricular Fibrillation/prevention & control , Action Potentials , Animals , Animals, Newborn , Cardiac Pacing, Artificial , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Dogs , Electrophysiologic Techniques, Cardiac , Humans , Muscle Proteins/genetics , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , NAV1.5 Voltage-Gated Sodium Channel , Rats , Rats, Sprague-Dawley , Sodium Channels/genetics , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Transfection , Ventricular Fibrillation/etiology , Ventricular Fibrillation/genetics , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...