Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Am Coll Cardiol ; 61(11): 1192-201, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23395072

ABSTRACT

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.


Subject(s)
Gene Transfer Techniques , Heart Conduction System , Heart Rate/physiology , Ion Channels/genetics , Muscle Proteins/genetics , Animals , Dogs , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels
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
5.
Cardiovasc Res ; 94(3): 450-9, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22374989

ABSTRACT

AIMS: Reentry accounts for most life-threatening arrhythmias, complicating myocardial infarction, and therapies that consistently prevent reentry from occurring are lacking. In this study, we compare antiarrhythmic effects of gene transfer of green fluorescent protein (GFP; sham), the skeletal muscle sodium channel (SkM1), the liver-specific connexin (Cx32), and SkM1/Cx32 in the subacute canine infarct. METHODS AND RESULTS: Immediately after ligation of the left anterior descending artery, viral constructs were implanted in the epicardial border zone (EBZ). Five to 7 days later, efficient restoration of impulse propagation (narrow QRS and local electrogram duration) occurred in SkM1, Cx32, and SkM1/Cx32 groups (P< 0.05 vs. GFP). Programmed electrical stimulation from the EBZ induced sustained ventricular tachycardia (VT)/ventricular fibrillation (VF) in 15/22 GFP dogs vs. 2/12 SkM1, 6/14 Cx32, and 8/10 SkM1/Cx32 (P< 0.05 SkM1 vs. GFP). GFP, SkM1, and SkM1/Cx32 had predominantly polymorphic VT/VF, whereas in Cx32 dogs, monomorphic VT predominated (P< 0.05 for Cx32 vs. GFP). Tetrazolium red staining showed significantly larger infarcts in Cx32- vs. GFP-treated animals (P< 0.05). CONCLUSION: Whereas SkM1 gene transfer reduces the incidence of inducible VT/VF, Cx32 therapy to improve gap junctional conductance results in larger infarct size, a different VT morphology, and no antiarrhythmic efficacy.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Connexins/metabolism , Gap Junctions/drug effects , Muscle Proteins/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Sodium Channels/metabolism , Ventricular Fibrillation/drug therapy , Animals , Anti-Arrhythmia Agents/therapeutic use , Connexins/genetics , Dogs , Electric Stimulation , Electrocardiography , Male , Mice , Muscle Proteins/genetics , Rats , Sodium Channels/genetics , Ventricular Fibrillation/physiopathology , Gap Junction beta-1 Protein
6.
Cell Transplant ; 20(11-12): 1907-14, 2011.
Article in English | MEDLINE | ID: mdl-21429290

ABSTRACT

Biological pacing has been proposed as a physiologic counterpart to electronic pacing, and the sinoatrial node (SAN) is the general standard for biological pacemakers. We tested the expression of SAN pacemaker cell activity when implanted autologously in the right ventricle (RV). We induced complete heart block and implanted electronic pacemakers in the RV of adult mongrel dogs. Autologous SAN cells isolated enzymatically were studied by patch clamp to confirm SAN identity. SAN cells (400,000) were injected into the RV subepicardial free wall and dogs were monitored for 2 weeks. Pacemaker function was assessed by overdrive pacing and IV epinephrine challenge. SAN cells expressed a time-dependent inward current (I(f)) activating on hyperpolarization: density = 4.3 ± 0.6 pA/pF at -105 mV. Four of the six dogs demonstrated >50% of beats originating from the implant site at 24 h. Biological pacemaker rates on days 7-14 = 45-55 bpm and post-overdrive escape times = 1.5-2.5 s. Brisk catecholamine responsiveness occurred. Dogs implanted with autologous SAN cells manifest biological pacing properties dissimilar from those of the anatomic SAN. This highlights the importance of cell and substrate interaction in generating biological pacemaker function.


Subject(s)
Biological Clocks/physiology , Sinoatrial Node/transplantation , Action Potentials/drug effects , Animals , Biological Clocks/drug effects , Cells, Cultured , Dogs , Electrocardiography , Epinephrine/pharmacology , Heart Block/physiopathology , Heart Ventricles/physiopathology , Male , Patch-Clamp Techniques , Sinoatrial Node/cytology , Transplantation, Autologous
7.
Heart Rhythm ; 7(12): 1835-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20708103

ABSTRACT

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.


Subject(s)
Arousal/physiology , Biological Clocks/physiology , Emotions/physiology , Adenoviridae/genetics , Animals , Atrioventricular Block/physiopathology , Atrioventricular Block/therapy , Dogs , Electrocardiography , Genetic Therapy , Green Fluorescent Proteins , Heart Rate/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/physiology , Luminescent Agents , Pacemaker, Artificial
8.
Heart Rhythm ; 7(8): 1104-10, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20385252

ABSTRACT

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.


Subject(s)
Heart Conduction System/physiopathology , Muscle, Skeletal/physiology , Myocardial Infarction/physiopathology , Sodium Channels/biosynthesis , Action Potentials , Animals , Disease Models, Animal , Dogs
9.
Heart Rhythm ; 7(7): 964-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20346417

ABSTRACT

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.


Subject(s)
Cardiac Pacing, Artificial , Cyclic AMP Response Element-Binding Protein/metabolism , Heart Conduction System/metabolism , Kv Channel-Interacting Proteins/analysis , Ventricular Function, Left/physiology , Action Potentials/physiology , Angiotensin II/physiology , Animals , Arrhythmias, Cardiac/metabolism , Blotting, Western , Cells, Cultured , Dogs , Ion Channels/physiology , Lipid Peroxidation , Male , Models, Animal , Models, Cardiovascular , Myocardium/metabolism , Myocytes, Cardiac/cytology , Oxidative Stress/physiology , Proteasome Endopeptidase Complex/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin/physiology , Ubiquitination , Ventricular Remodeling/physiology
10.
Circulation ; 119(1): 19-27, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19103989

ABSTRACT

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.


Subject(s)
Genetic Therapy/methods , Heart Conduction System/physiology , Models, Cardiovascular , Sodium Channels/genetics , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/therapy , Action Potentials/physiology , Adenoviridae/genetics , Animals , Cell Line , Computer Simulation , Disease Models, Animal , Dogs , Gene Expression , Green Fluorescent Proteins/genetics , Humans , In Vitro Techniques , Kidney/cytology , Male , Muscle, Skeletal/physiology , Myocardial Contraction/physiology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , NAV1.5 Voltage-Gated Sodium Channel , Penicillin G/metabolism , Pericardium/physiology , Sodium Channels/metabolism , Sodium Channels/physiology , Tachycardia, Ventricular/pathology
11.
Heart Rhythm ; 4(3): 341-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17341400

ABSTRACT

BACKGROUND: The concept that the interval between the peak (T(peak)) and the end (T(end)) of the T wave (T(p-e)) is a measure of transmural dispersion of repolarization time is widely accepted but has not been tested rigorously by transmural mapping of the intact heart. OBJECTIVES: The purpose of this study was to test the relationship of T(p-e) to transmural dispersion of repolarization by correlating local repolarization times at endocardial, midmural, and epicardial sites in the left and right ventricles with the T wave of the ECG. METHODS: Local activation times, activation-recovery intervals, and repolarization times were measured at 98 epicardial sites and up to 120 midmural and endocardial sites in eight open-chest dogs. In four of the dogs, long-term cardiac memory was induced by 3 weeks of ventricular pacing at 130 bpm because previous data suggest that, in this setting, delayed epicardial repolarization increases transmural dispersion. The other four dogs were sham operated. RESULTS: In sham dogs, T(p-e) was 41 +/- 2.2 ms (X +/- SEM), whereas the transmural dispersion of repolarization time was 2.7 +/- 4.2 ms (not significant between endocardium and epicardium). Cardiac memory was associated with evolution of a transmural gradient of 14.5 +/- 1.9 ms (P <.02), with epicardium repolarizing later than endocardium. The corresponding T(p-e) was 43 +/- 2.3 ms (not different from sham). In combined sham and memory dogs, T(p-e) intervals did not correlate with transmural dispersion of repolarization times. In contrast, dispersion of repolarization of the whole heart (measured as the difference between the earliest and the latest moment of repolarization from all left and right ventricular, endocardial, intramural, and epicardial recording sites) did correlate with T(p-e) (P <.0005, r = 0.98), although the latter underestimated total repolarization time by approximately 35%. The explanation for this finding is that parts of the heart fully repolarize before the moment of T(peak). CONCLUSION: T(p-e) does not correlate with transmural dispersion of repolarization but is an index of total dispersion of repolarization.


Subject(s)
Electrocardiography , Heart Conduction System/physiology , Action Potentials , Analysis of Variance , Animals , Cardiac Pacing, Artificial , Dogs , Electrodes, Implanted , Electrophysiologic Techniques, Cardiac , Endocardium/physiology , Female , Image Processing, Computer-Assisted , Linear Models , Male , Models, Animal , Models, Cardiovascular , Pericardium/physiology , Research Design , Ventricular Function
12.
Cardiovasc Res ; 74(3): 416-25, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17391659

ABSTRACT

OBJECTIVE: The contribution of regional electrophysiologic heterogeneity to the T-wave changes of long-term cardiac memory (CM) is not known. We mapped activation and repolarization in dogs after induction of CM and in sham animals. METHODS AND RESULTS: CM was induced by three weeks of AV-sequential pacing at the anterior free wall of the left ventricle (LV), midway between apex and base in 5 dogs. In 4 sham controls a pacemaker was implanted but ventricular pacing was not performed. At 3 weeks, unipolar electrograms were recorded (98 epicardial, 120 intramural and endocardial electrodes) during atrial stimulation (cycle length 450 ms). Activation times (AT) and repolarization times (RT) were measured and activation recovery intervals (ARIs) calculated. CM was associated with 1) deeper T waves on ECG, with no change in QT interval; 2) longer activation time at the site of stimulation in CM (29.7+/-1.0, X+/-SEM) than sham (23.9+/-1.3 ms p<0.01); 3) an LV transmural gradient in repolarization time such that repolarization at the epicardium terminated 12.4+/-2.4 ms later than at the endocardium p<0.01), in contrast to no gradient in shams (2.7+/-4.2 ms); in memory dogs, the repolarization time gradient was greatest at sites around the pacing electrode varying from 13.1+/-2.3 ms to 25.5+/-3.8 ms; 4) more negative left ventricular potentials at the peak of the body surface T wave (-4.9+/-0.8 vs -2.2+/-0.4 mV; p<0.05) but no altered right ventricular epicardial T-wave potentials. ARIs did not differ between groups. Right ventricular activation was delayed but was not associated with altered repolarization because of compensatory shortening of the right ventricular ARIs. CONCLUSION: CM-induced T-wave changes are caused by evolution of transmural repolarization gradients manifested during atrial stimulation that are maximal near the site of ventricular pacing.


Subject(s)
Cardiac Pacing, Artificial , Electrocardiography , Heart Conduction System/physiology , Action Potentials , Animals , Dogs , Endocardium/physiology , Male , Pericardium/physiology , Time Factors , Ventricular Function
13.
Circulation ; 112(12): 1711-8, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16157774

ABSTRACT

BACKGROUND: Questions remain about the contributions of transmural versus apicobasal repolarization gradients to the configuration of the T wave in control settings and after the induction of short-term cardiac memory. METHODS AND RESULTS: Short-term cardiac memory is seen as T-wave changes induced by altered ventricular activation that persists after restoration of sinus rhythm. We studied cardiac memory in anesthetized, open-chest dogs paced from the ventricle for 2 hours. Unipolar electrograms were recorded from as many as 98 epicardial and 144 intramural sites, and activation times and activation-recovery intervals (ARIs) were measured. In separate experiments, epicardial monophasic action potentials were recorded. We found no appreciable left ventricular intramural gradients in repolarization times (activation time+ARI) in either control conditions or after the induction of memory. In controls, there was a left ventricular apicobasal gradient, with the shortest repolarization times in anterobasal regions and longest repolarization times posteroapically. After induction of memory, repolarization times shortened uniformly throughout the ventricular wall. Monophasic action potential duration at 90% repolarization decreased by approximately 10 ms after induction of memory. CONCLUSIONS: In the intact canine left ventricle at physiological rates, there is no transmural gradient in repolarization. Apicobasal gradients in repolarization time, with shortest repolarization times in anterobasal areas and longest repolarization times in posteroapical regions, are important in the genesis of the T wave. Repolarization times and monophasic action potentials at the 90% repolarization level shorten after the induction of memory. The deeper T wave in the ECG after induction of memory may be explained by the more rapid phase 3 of the action potential.


Subject(s)
Action Potentials/physiology , Heart Conduction System/physiology , Ventricular Function, Left/physiology , Animals , Blood Pressure , Dogs , Electrocardiography , Electroshock , Male , Membrane Potentials/physiology , Models, Animal
14.
Circulation ; 110(5): 489-95, 2004 Aug 03.
Article in English | MEDLINE | ID: mdl-15262840

ABSTRACT

BACKGROUND: Calcium-insensitive transient outward current (I(to)) is important to the development of cardiac memory (CM), which itself reflects the capacity of the heart to remodel electrophysiologically. We used cardiac pacing to test the hypothesis that CM evolution can be explained by developmental maturation of I(to). METHODS AND RESULTS: Acutely anesthetized dogs from 1 day old to adult were paced from the left ventricle (VP, n=29) or left atrial appendage (AP, n=12) to induce CM. T-wave vector displacement (TVD) obtained during VP was greater than with AP (adults, 0.39+/-0.06 mV; neonates, 0.04+/-0.01 mV; P<0.05). TVD began to increase at approximately 40 days of age, reaching adult levels by approximately 200 days. Microelectrode studies performed in 18 dogs (ages 3 to 94 days) after completing the CM protocol and 20 additional dogs (1 day old to adult) revealed that the epicardial action potential notch was absent in neonates, became apparent in the young, and was deepest in adults. The relationship between TVD and epicardial notch was such that as notch magnitude increased, TVD increased (r=-0.65, P<0.05). KChIP2 and Kv4.3 mRNA (measured via reverse transcription-polymerase chain reaction) also increased with age. CONCLUSIONS: The inducibility of CM gradually increases with age in association with evolution of the epicardial action potential notch and mRNA expression for KChIP2 and Kv4.3. This suggests that the capacity of the heart to remodel electrophysiologically and to manifest memory during development depends in part on evolution of the determinants of I(to).


Subject(s)
Aging/physiology , Calcium-Binding Proteins/biosynthesis , Electrocardiography , Heart Conduction System/growth & development , Potassium Channels, Voltage-Gated/biosynthesis , Action Potentials , Animals , Animals, Newborn , Calcium/metabolism , Calcium-Binding Proteins/genetics , Cardiac Pacing, Artificial , Dogs , Female , Gene Expression Regulation, Developmental , Heart Conduction System/physiology , Ion Transport , Kv Channel-Interacting Proteins , Male , Potassium Channels, Voltage-Gated/genetics , RNA, Messenger/biosynthesis , Shal Potassium Channels
15.
Circ Res ; 94(7): 952-9, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14988226

ABSTRACT

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.


Subject(s)
Genetic Therapy , Ion Channels/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Muscle Proteins/physiology , Myocytes, Cardiac/physiology , Acetylcholine/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Cells, Cultured/drug effects , Cells, Cultured/physiology , Cells, Cultured/transplantation , Cesium/pharmacology , Cholinergic Agents/pharmacology , Coculture Techniques , Dogs , Electroporation , Gap Junctions/physiology , Gap Junctions/ultrastructure , Heart Rate , Heart Ventricles/cytology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Transport/drug effects , Isoproterenol/pharmacology , Mesenchymal Stem Cells/drug effects , Mice , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Potassium Channels , Rats , Recombinant Fusion Proteins/physiology , Transfection
16.
Circulation ; 109(4): 506-12, 2004 Feb 03.
Article in English | MEDLINE | ID: mdl-14734518

ABSTRACT

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.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Heart Conduction System , Heart Ventricles/physiopathology , Ion Channels/genetics , Muscle Proteins/genetics , Action Potentials , Adenoviridae/genetics , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/therapy , Dogs , Electric Stimulation , Electrocardiography , Female , Genetic Therapy , Genetic Vectors , Green Fluorescent Proteins , Hematoma/etiology , Hematoma/pathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/analysis , Luminescent Proteins/genetics , Male , Muscle Proteins/analysis , Myocytes, Cardiac/physiology , Pacemaker, Artificial , Patch-Clamp Techniques , Periodicity , Purkinje Fibers/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...