Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 922
1.
Cardiovasc Res ; 118(1): 184-195, 2022 01 07.
Article En | MEDLINE | ID: mdl-33098411

AIMS: Systemic inflammation and increased activity of atrial NOX2-containing NADPH oxidases have been associated with the new onset of atrial fibrillation (AF) after cardiac surgery. In addition to lowering LDL-cholesterol, statins exert rapid anti-inflammatory and antioxidant effects, the clinical significance of which remains controversial. METHODS AND RESULTS: We first assessed the impact of cardiac surgery and cardiopulmonary bypass (CPB) on atrial nitroso-redox balance by measuring NO synthase (NOS) and GTP cyclohydrolase-1 (GCH-1) activity, biopterin content, and superoxide production in paired samples of the right atrial appendage obtained before (PRE) and after CPB and reperfusion (POST) in 116 patients. The effect of perioperative treatment with atorvastatin (80 mg once daily) on these parameters, blood biomarkers, and the post-operative atrial effective refractory period (AERP) was then evaluated in a randomized, double-blind, placebo-controlled study in 80 patients undergoing cardiac surgery on CPB. CPB and reperfusion led to a significant increase in atrial superoxide production (74% CI 71-76%, n = 46 paired samples, P < 0.0001) and a reduction in atrial tetrahydrobiopterin (BH4) (34% CI 33-35%, n = 36 paired samples, P < 0.01), and in GCH-1 (56% CI 55-58%, n = 26 paired samples, P < 0.001) and NOS activity (58% CI 52-67%, n = 20 paired samples, P < 0.001). Perioperative atorvastatin treatment prevented the effect of CPB and reperfusion on all parameters but had no significant effect on the postoperative right AERP, troponin release, or NT-proBNP after cardiac surgery. CONCLUSION: Perioperative statin therapy prevents post-reperfusion atrial nitroso-redox imbalance in patients undergoing on-pump cardiac surgery but has no significant impact on postoperative atrial refractoriness, perioperative myocardial injury, or markers of postoperative LV function. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT01780740.


Atorvastatin/therapeutic use , Atrial Fibrillation/prevention & control , Atrial Function, Right/drug effects , Cardiac Surgical Procedures/adverse effects , Cardiopulmonary Bypass/adverse effects , Heart Atria/drug effects , Nitroso Compounds/metabolism , Refractory Period, Electrophysiological/drug effects , Action Potentials/drug effects , Atorvastatin/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Biopterins/analogs & derivatives , Biopterins/metabolism , Double-Blind Method , England , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Rate/drug effects , Humans , NADPH Oxidases/metabolism , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Superoxides/metabolism , Time Factors , Treatment Outcome
2.
Cardiovasc Res ; 118(2): 503-516, 2022 01 29.
Article En | MEDLINE | ID: mdl-33624748

AIMS: Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare laminopathy caused by expression of progerin, a lamin A variant, also present at low levels in non-HGPS individuals. HGPS patients age and die prematurely, predominantly from cardiovascular complications. Progerin-induced cardiac repolarization defects have been described previously, although the underlying mechanisms are unknown. METHODS AND RESULTS: We conducted studies in heart tissue from progerin-expressing LmnaG609G/G609G (G609G) mice, including microscopy, intracellular calcium dynamics, patch-clamping, in vivo magnetic resonance imaging, and electrocardiography. G609G mouse cardiomyocytes showed tubulin-cytoskeleton disorganization, t-tubular system disruption, sarcomere shortening, altered excitation-contraction coupling, and reductions in ventricular thickening and cardiac index. G609G mice exhibited severe bradycardia, and significant alterations of atrio-ventricular conduction and repolarization. Most importantly, 50% of G609G mice had altered heart rate variability, and sinoatrial block, both significant signs of premature cardiac aging. G609G cardiomyocytes had electrophysiological alterations, which resulted in an elevated action potential plateau and early afterdepolarization bursting, reflecting slower sodium current inactivation and long Ca+2 transient duration, which may also help explain the mild QT prolongation in some HGPS patients. Chronic treatment with low-dose paclitaxel ameliorated structural and functional alterations in G609G hearts. CONCLUSIONS: Our results demonstrate that tubulin-cytoskeleton disorganization in progerin-expressing cardiomyocytes causes structural, cardiac conduction, and excitation-contraction coupling defects, all of which can be partially corrected by chronic treatment with low dose paclitaxel.


Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Cytoskeleton/drug effects , Excitation Contraction Coupling/drug effects , Heart Conduction System/drug effects , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , Paclitaxel/pharmacology , Progeria/drug therapy , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Lamin Type A/genetics , Lamin Type A/metabolism , Male , Mice, Mutant Strains , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Progeria/genetics , Progeria/metabolism , Progeria/physiopathology , Refractory Period, Electrophysiological/drug effects , Swine , Swine, Miniature , Tubulin/metabolism
3.
Int Heart J ; 62(3): 607-615, 2021.
Article En | MEDLINE | ID: mdl-34054001

The aim was to investigate the role of the α7nAChR-mediated cholinergic anti-inflammatory pathway in vagal nerve regulated atrial fibrillation (AF).18 beagles (standard dogs for testing) were used in this study, and the effective refractory period (ERP) of atrium and pulmonary veins and AF inducibility were measured hourly during rapid atrial pacing at 800 beats/minute for 6 hours in all beagles. After cessation of 3 hours of RAP, the low-level vagal nerve stimulation (LL-VNS) group (n = 6) was given LL-VNS and injection of salinne (0.5 mL/GP) into four GPs, the methyllycaconitine (MLA, the antagonist of α7nAChR) group (n = 6) was given LL-VNS and injection of MLA into four GPs, and the Control group (n = 6) was given saline into four GPs and the right cervical vagal nerve was exposed without stimulation. Then, the levels of the tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), acetylcholine (ACh), STAT3, and NF-κB proteins were measured. During the first 3 hours of RAP, the ERPs gradually decreased while the dispersion of ERPs (dERPs) and AF inducibility gradually increased in all three groups. During the last 3 hours of 6 hours' RAP in this study, the ERPs in the LL-VNS group were higher, while the dERPs and AF inducibility were significantly lower when compared with the Control and MLA groups at the same time points. The levels of ACh in the serum and atrium in the LL-VNS and MLA groups were higher than in the Control group, and the levels of TNF-α and IL-6 were higher in the Control and MLA groups than in the LL-VNS group. The concentrations of STAT3 in RA and LA tissues were higher in the LL-VNS group while those of NF-κB were lower.In conclusion, the cholinergic anti-inflammatory pathway mediated by α7nACh plays an important role in low-level vagal nerve-regulated AF.


Aconitine/analogs & derivatives , Atrial Fibrillation/physiopathology , Neuroimmunomodulation/drug effects , Vagus Nerve/drug effects , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Acetylcholine/blood , Aconitine/administration & dosage , Aconitine/pharmacology , Animals , Cardiac Pacing, Artificial/adverse effects , Cardiac Pacing, Artificial/methods , Case-Control Studies , Disease Models, Animal , Dogs , Heart Atria/innervation , Heart Atria/physiopathology , Interleukin-6/blood , NF-kappa B/blood , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/pharmacology , Pulmonary Veins/innervation , Pulmonary Veins/physiopathology , Refractory Period, Electrophysiological/drug effects , STAT3 Transcription Factor/blood , Tumor Necrosis Factor-alpha/blood , Vagus Nerve Stimulation/adverse effects , Vagus Nerve Stimulation/methods
4.
Basic Clin Pharmacol Toxicol ; 128(6): 741-746, 2021 Jun.
Article En | MEDLINE | ID: mdl-33548156

BACKGROUND: Metoclopramide (MCP) is a dopamine D2 -receptor antagonist, mainly used to treat post-operative or chemotherapy-induced nausea. While it is very effective in the cure of gastric symptoms, MCP can cause severe neurologic side effects. Furthermore, there is growing evidence for severe arrhythmic side effects resulting from inhibitory effects on cardiac sodium and potassium channels. METHODS AND RESULTS: Thirteen hearts of New Zealand white rabbits were retrogradely perfused, and electrophysiology studies were performed to obtain action potential duration (APD90 ) and effective refractory period (ERP). After generating baseline data, the hearts were perfused with increasing concentrations of metoclopramide (MCP 10 µM, MCP 50 µM, MCP 100 µM) and the standardized protocol was repeated for each concentration. Perfusion with MCP resulted in a significant prolongation of APD90 and QT interval. In parallel, the incidence of ventricular tachycardias was significantly increased by high doses of MCP. CONCLUSION: This is the first experimental study that investigated the effect of increasing doses of metoclopramide on a sensitive whole-heart model of proarrhythmia. MCP led to a significant increase in action potential duration and QT interval; meanwhile, the number of ventricular tachycardias was significantly increased.


Action Potentials/drug effects , Dopamine D2 Receptor Antagonists/adverse effects , Heart/drug effects , Metoclopramide/adverse effects , Metoclopramide/pharmacology , Animals , Cardiac Electrophysiology , Disease Models, Animal , Female , Postoperative Nausea and Vomiting/drug therapy , Rabbits , Refractory Period, Electrophysiological/drug effects , Tachycardia, Ventricular
5.
Cardiovasc Toxicol ; 21(3): 206-215, 2021 03.
Article En | MEDLINE | ID: mdl-33074476

While vanoxerine (GBR-12909) is a synaptosomal dopamine uptake inhibitor, it also suppresses IKr, INa and ICa,L in vitro. Based on these profiles on ionic currents, vanoxerine has been developed as a candidate compound for treating atrial fibrillation. To investigate electropharmacological profiles, vanoxerine dihydrochloride was intravenously administered at 0.03 and 0.3 mg/kg to halothane-anesthetized dogs (n = 4), possibly providing subtherapeutic and therapeutic concentrations, respectively. The low dose increased the heart rate and cardiac output, whereas it prolonged the ventricular refractoriness. The high dose decreased the heart rate but increased the total peripheral vascular resistance, whereas it delayed the ventricular repolarization and increased the atrial refractoriness in addition to further enhancing the ventricular refractoriness. The extent of increase in the refractoriness in the atrium was 0.8 times of that in the ventricle. The high dose also prolonged the early and late repolarization periods of the ventricle as well as the terminal repolarization period. Meanwhile, no significant change was detected in the mean blood pressure, ventricular contraction, preload to the left ventricle, or the intra-atrial, intra-ventricular or atrioventricular conductions. The high dose can be considered to inhibit IKr, but it may not suppress INa or ICa in the in situ heart, partly explaining its poor atrial selectivity for increasing refractoriness. The prolongation of early repolarization period may reflect enhancement of net inward current, providing potential risk for intracellular Ca2+ overload. Thus, vanoxerine may provide both trigger and substrate toward torsade de pointes, which would make the drug less promising as an anti-atrial fibrillatory drug.


Anti-Arrhythmia Agents/toxicity , Atrial Fibrillation/drug therapy , Dopamine Uptake Inhibitors/toxicity , Heart Conduction System/drug effects , Heart Rate/drug effects , Piperazines/toxicity , Torsades de Pointes/chemically induced , Action Potentials/drug effects , Anesthesia, Inhalation , Anesthetics, Inhalation , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Dogs , Female , Halothane , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Refractory Period, Electrophysiological/drug effects , Risk Assessment , Time Factors , Torsades de Pointes/metabolism , Torsades de Pointes/physiopathology
6.
J Cardiovasc Pharmacol ; 76(5): 492-505, 2020 11.
Article En | MEDLINE | ID: mdl-33165131

Antiarrhythmic drugs remain the mainstay therapy for patients with atrial fibrillation (AF). A major disadvantage of the currently available anti-AF agents is the risk of induction of ventricular proarrhythmias. Aiming to reduce this risk, several atrial-specific or -selective ion channel block approaches have been introduced for AF suppression, but only the atrial-selective inhibition of the sodium channel has been demonstrated to be valid in both experimental and clinical studies. Among the other pharmacological anti-AF approaches, "upstream therapy" has been prominent but largely disappointing, and pulmonary delivery of anti-AF drugs seems to be promising. Major contradictions exist in the literature about the electrophysiological mechanisms of AF (ie, reentry or focal?) and the mechanisms by which anti-AF drugs terminate AF, making the search for novel anti-AF approaches largely empirical. Drug-induced termination of AF may or may not be associated with prolongation of the atrial effective refractory period. Anti-AF drug research has been largely based on the "suppress reentry" ideology; however, results of the AF mapping studies increasingly indicate that nonreentrant mechanism(s) plays an important role in the maintenance of AF. Also, the analysis of anti-AF drug-induced electrophysiological alterations during AF, conducted in the current study, leans toward the focal source as the prime mechanism of AF maintenance. More effort should be placed on the investigation of pharmacological suppression of the focal mechanisms.


Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Heart Conduction System/drug effects , Heart Rate/drug effects , Animals , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Heart Conduction System/physiopathology , Humans , Potassium Channel Blockers/pharmacology , Refractory Period, Electrophysiological/drug effects , Sodium Channel Blockers/pharmacology
7.
J Mol Cell Cardiol ; 147: 18-26, 2020 10.
Article En | MEDLINE | ID: mdl-32768409

INTRODUCTION: GapmeRs are oligonucleotides that bind to a specific RNA sequence and thereby affecting posttranscriptional gene regulation. They therefore hold the potential to manipulate targets where current pharmacological modulators are inefficient or exhibit adverse side effects. Here, we show that a treatment with a GapmeR, mediating knockdown of small conductance Ca2+-activated K+ channels (SK3), has an in vivo protective effect against atrial fibrillation (AF) in rats. MATERIAL AND METHODS: A unique SK3-GapmeR design was selected after thorough in vitro evaluation. 22 rats were randomly assigned to receive either 50 mg/kg SK3-GapmeR or vehicle subcutaneously once a week for two weeks. Langendorff experiments were performed seven days after the last injection, where action potential duration (APD90), effective refractory period (ERP) and AF propensity were investigated. SK3 channel activity was evaluated using the SK channel blocker, ICA (N-(pyridin-2-yl)-4-(pyridine-2-yl)thiazol-2-amine). SK3 protein expression was assessed by Western Blot. RESULTS: The designed GapmeR effectively down-regulate the SK3 protein expression in the heart (48% downregulation, p = 0.0095) and did indeed protect against AF. Duration of AF episodes elicited by burst pacing in the rats treated with SK3-GapmeR was reduced 78% compared to controls (3.7 s vs. 16.8 s, p = 0.0353). The number of spontaneous AF episodes were decreased by 68% in the SK3-GapmeR group (39 episodes versus 123 in the control group, respectively) and were also significantly shorter in duration (7.2 s versus 29.7 s in the control group, p = 0.0327). Refractoriness was not altered at sinus rhythm, but ERP prolongation following ICA application was blunted in the SK3-GapmeR group. CONCLUSION: The selected GapmeR silenced the cardiac SK3 channels, thereby preventing AF in rats. Thus, GapmeR technology can be applied as an experimental tool of downregulation of cardiac proteins and could potentially offer a novel modality for treatment of cardiac diseases.


Atrial Fibrillation/drug therapy , Atrial Fibrillation/prevention & control , Gene Knockdown Techniques , Oligonucleotides, Antisense/therapeutic use , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Atrial Fibrillation/pathology , Cell Line , Down-Regulation/drug effects , Myocardium/metabolism , Myocardium/pathology , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Refractory Period, Electrophysiological/drug effects , Refractory Period, Electrophysiological/physiology , Small-Conductance Calcium-Activated Potassium Channels/genetics
8.
J Am Heart Assoc ; 9(10): e015751, 2020 05 18.
Article En | MEDLINE | ID: mdl-32390491

Background The tandem of P domains in a weak inward rectifying K+ channel (TWIK)-related acid-sensitive K+ channel (TASK-1; hK2P3.1) two-pore-domain potassium channel was recently shown to regulate the atrial action potential duration. In the human heart, TASK-1 channels are specifically expressed in the atria. Furthermore, upregulation of atrial TASK-1 currents was described in patients suffering from atrial fibrillation (AF). We therefore hypothesized that TASK-1 channels represent an ideal target for antiarrhythmic therapy of AF. In the present study, we tested the antiarrhythmic effects of the high-affinity TASK-1 inhibitor A293 on cardioversion in a porcine model of paroxysmal AF. Methods and Results Heterologously expressed human and porcine TASK-1 channels are blocked by A293 to a similar extent. Patch clamp measurements from isolated human and porcine atrial cardiomyocytes showed comparable TASK-1 currents. Computational modeling was used to investigate the conditions under which A293 would be antiarrhythmic. German landrace pigs underwent electrophysiological studies under general anesthesia. Paroxysmal AF was induced by right atrial burst stimulation. After induction of AF episodes, intravenous administration of A293 restored sinus rhythm within cardioversion times of 177±63 seconds. Intravenous administration of A293 resulted in significant prolongation of the atrial effective refractory period, measured at cycle lengths of 300, 400 and 500 ms, whereas the surface ECG parameters and the ventricular effective refractory period lengths remained unchanged. Conclusions Pharmacological inhibition of atrial TASK-1 currents exerts antiarrhythmic effects in vivo as well as in silico, resulting in acute cardioversion of paroxysmal AF. Taken together, these experiments indicate the therapeutic potential of A293 for AF treatment.


Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Sulfonamides/pharmacology , ortho-Aminobenzoates/pharmacology , Animals , Atrial Fibrillation/diagnosis , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Disease Models, Animal , Electrocardiography , Electrophysiologic Techniques, Cardiac , Female , Humans , Male , Membrane Potentials/drug effects , Molecular Docking Simulation , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Proof of Concept Study , Refractory Period, Electrophysiological/drug effects , Sus scrofa , Time Factors , Xenopus laevis
9.
J Cardiovasc Pharmacol ; 76(2): 164-172, 2020 08.
Article En | MEDLINE | ID: mdl-32453071

The mechanisms underlying atrial-selective prolongation of effective refractory period (ERP) and suppression of atrial fibrillation (AF) by NS8593 and UCL1684, small conductance calcium-activated potassium (SK) channel blockers, are poorly defined. The purpose of the study was to confirm the effectiveness of these agents to suppress AF and to probe the underlying mechanisms. Transmembrane action potentials and pseudoelectrocardiograms were recorded from canine isolated coronary-perfused canine atrial and ventricular wedge preparations. Patch clamp techniques were used to record sodium channel current (INa) in atrial and ventricular myocytes and human embryonic kidney cells. In both atria and ventricles, NS8593 (3-10 µM) and UCL1684 (0.5 µM) did not significantly alter action potential duration, suggesting little to no SK channel inhibition. Both agents caused atrial-selective: (1) prolongation of ERP secondary to development of postrepolarization refractoriness, (2) reduction of Vmax, and (3) increase of diastolic threshold of excitation (all are sodium-mediated parameters). NS8593 and UCL1684 significantly reduced INa density in human embryonic kidney cells as well as in atrial but not in ventricular myocytes at physiologically relevant holding potentials. NS8593 caused a shift of steady-state inactivation to negative potentials in atrial but not ventricular cells. NS8593 and UCL1684 prevented induction of acetylcholine-mediated AF in 6/6 and 8/8 preparations, respectively. This anti-AF effect was associated with strong rate-dependent depression of excitability. The SK channel blockers, NS8593 and UCL1684, are effective in preventing the development of AF due to potent atrial-selective inhibition of INa, causing atrial-selective prolongation of ERP secondary to induction of postrepolarization refractoriness.


1-Naphthylamine/analogs & derivatives , Alkanes/pharmacology , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/prevention & control , Heart Atria/drug effects , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/drug effects , Quinolinium Compounds/pharmacology , Sodium Channel Blockers/pharmacology , 1-Naphthylamine/pharmacology , Action Potentials/drug effects , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Dogs , Female , HEK293 Cells , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Humans , Male , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Potassium Channel Blockers/pharmacology , Refractory Period, Electrophysiological/drug effects , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/metabolism
10.
Heart Vessels ; 35(9): 1316-1322, 2020 Sep.
Article En | MEDLINE | ID: mdl-32346771

AVE0118, an inhibitor of IKur, Ito and IK,ACh, was in the drug pipeline for atrial fibrillation. To investigate the limitation of AVE0118 as an anti-atrial fibrillatory drug, we studied its electropharmacological effects particularly focusing on the anti-atrial fibrillatory action as reverse translational research. We adopted the chronic atrioventricular block beagle dogs (n = 4), having a pathophysiology of bradycardia-associated, volume overload-induced chronic heart failure, in which the atrial fibrillation was induced by 10 s of burst pacing on atrial septum. AVE0118 in doses of 0.24 and 1.2 mg/kg, i.v. over 10 min hardly altered electrophysiological variables. Meanwhile, AVE0118 in a dose of 6 mg/kg, i.v. over 10 min delayed the inter-atrial conduction in a frequency-dependent manner and prolonged the atrial effective refractory period in a reverse frequency-dependent manner, whereas it did not significantly alter the duration of atrial fibrillation or its cycle length. The increment of atrial effective refractory period was 3.3 times greater compared with that of ventricular one at a basic cycle length of 400 ms. Torsade de pointes was not induced during the experimental period. Thus, AVE0118 may possess a favorable cardiac safety pharmacological profile, but its weak anti-atrial fibrillatory effect would indicate the limitation of atrial repolarization-delaying agents for suppressing atrial fibrillation.


Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/prevention & control , Atrioventricular Block/drug therapy , Biphenyl Compounds/pharmacology , Heart Atria/drug effects , Heart Rate/drug effects , Potassium Channel Blockers/pharmacology , Refractory Period, Electrophysiological/drug effects , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Remodeling/drug effects , Atrioventricular Block/complications , Atrioventricular Block/metabolism , Atrioventricular Block/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Heart Atria/metabolism , Heart Atria/physiopathology , Male , Time Factors
11.
BMC Cardiovasc Disord ; 20(1): 85, 2020 02 17.
Article En | MEDLINE | ID: mdl-32066388

BACKGROUND: Coronary microembolization (CME) has a poor prognosis, with ventricular arrhythmia being the most serious consequence. Understanding the underlying mechanisms could improve its management. We investigated the effects of granulocyte colony-stimulating factor (G-CSF) on connexin-43 (Cx43) expression and ventricular arrhythmia susceptibility after CME. METHODS: Forty male rabbits were randomized into four groups (n = 10 each): Sham, CME, G-CSF, and AG490 (a JAK2 selective inhibitor). Rabbits in the CME, G-CSF, and AG490 groups underwent left anterior descending (LAD) artery catheterization and CME. Animals in the G-CSF and AG490 groups received intraperitoneal injection of G-CSF and G-CSF + AG490, respectively. The ventricular structure was assessed by echocardiography. Ventricular electrical properties were analyzed using cardiac electrophysiology. The myocardial interstitial collagen content and morphologic characteristics were evaluated using Masson and hematoxylin-eosin staining, respectively. RESULTS: Western blot and immunohistochemistry were employed to analyze the expressions of Cx43, G-CSF receptor (G-CSFR), JAK2, and STAT3. The ventricular effective refractory period (VERP), VERP dispersion, and inducibility and lethality of ventricular tachycardia/fibrillation were lower in the G-CSF than in the CME group (P < 0.01), indicating less severe myocardial damage and arrhythmias. The G-CSF group showed higher phosphorylated-Cx43 expression (P < 0.01 vs. CME). Those G-CSF-induced changes were reversed by A490, indicating the involvement of JAK2. G-CSFR, phosphorylated-JAK2, and phosphorylated-STAT3 protein levels were higher in the G-CSF group than in the AG490 (P < 0.01) and Sham (P < 0.05) groups. CONCLUSION: G-CSF might attenuate myocardial remodeling via JAK2-STAT3 signaling and thereby reduce ventricular arrhythmia susceptibility after CME.


Arrhythmias, Cardiac/prevention & control , Coronary Artery Disease/drug therapy , Granulocyte Colony-Stimulating Factor/pharmacology , Heart Rate/drug effects , Janus Kinase 2/metabolism , Myocardial Infarction/prevention & control , Myocardium/enzymology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/enzymology , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Connexin 43/metabolism , Coronary Artery Disease/enzymology , Coronary Artery Disease/pathology , Coronary Artery Disease/physiopathology , Disease Models, Animal , Fibrosis , Male , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Phosphorylation , Rabbits , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Refractory Period, Electrophysiological/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction
12.
Cardiovasc Toxicol ; 20(4): 339-350, 2020 08.
Article En | MEDLINE | ID: mdl-31898152

Since deuterium replacement has a potential to modulate pharmacodynamics, pharmacokinetics and toxicity, we developed deuterated dronedarone; poyendarone, and assessed its cardiovascular effects. Poyendarone hydrochloride in doses of 0.3 and 3 mg/kg over 30 s was intravenously administered to the halothane-anesthetized dogs (n = 4), which provided peak plasma concentrations of 108 ± 10 and 1120 ± 285 ng/mL, respectively. The 0.3 mg/kg shortened the ventricular repolarization period. The 3 mg/kg transiently increased the heart rate at 5 min but decreased at 45 min, and elevated the total peripheral vascular resistance and left ventricular preload, whereas it reduced the mean blood pressure at 5 min, left ventricular contractility and cardiac output. The transient tachycardic action is considered to be induced by the hypotension-induced, reflex-mediated increase of sympathetic tone. The 3 mg/kg delayed both intra-atrial and intra-ventricular conductions, indicating Na+ channel inhibitory action. Moreover, the 3 mg/kg transiently shortened the ventricular repolarization period at 5 min. No significant change was detected in the late repolarization by poyendarone, indicating it might not hardly significantly alter rapidly activating delayed-rectifier K+ current (IKr). Poyendarone prolonged the atrial effective refractory period greater than the ventricular parameter. When compared with dronedarone, poyendarone showed similar pharmacokinetics of dronedarone, but reduced ß-adrenoceptor blocking activity as well as the cardio-suppressive effect. Poyendarone failed to inhibit IKr and showed higher atrial selectivity in prolonging the effective refractory period of atrium versus ventricle. Thus, the deuteration may be an effective way to improve the cardiovascular profile of dronedarone. Poyendarone is a promising anti-atrial fibrillatory drug candidate.


Action Potentials/drug effects , Anti-Arrhythmia Agents/administration & dosage , Deuterium , Dronedarone/administration & dosage , Heart Conduction System/drug effects , Heart Rate/drug effects , Administration, Intravenous , Animals , Anti-Arrhythmia Agents/pharmacokinetics , Delayed Rectifier Potassium Channels/drug effects , Delayed Rectifier Potassium Channels/metabolism , Dogs , Dronedarone/analogs & derivatives , Dronedarone/pharmacokinetics , Female , Heart Conduction System/metabolism , Refractory Period, Electrophysiological/drug effects
13.
Toxicol Lett ; 318: 57-64, 2020 Jan.
Article En | MEDLINE | ID: mdl-31585160

3-Bromopyruvate (3-BrPA) is a promising agent that has been widely studied in the treatment of cancer and pulmonary hypertension. Rotenone is a pesticide commonly used on farms and was shown to have anti-cancer activity and delay fibrosis progression in chronic kidney disease in a recent study. However, there are few studies showing the toxicity of rotenone and 3-BrPA in the myocardium. To support further medical exploration, it is necessary to clarify the side effects of these compounds on the heart. This study was designed to examine the cardiotoxicity of 3-BrPA and rotenone by investigating electrical and structural cardiac remodeling in rats. Forty male rats were divided into 4 groups (n = 10 in each group) and injected intraperitoneally with 3-BrPA, rotenone or a combination of 3-BrPA and rotenone. The ventricular effective refractory period (VERP), corrected QT interval (QTc), and ventricular tachycardia/ventricular fibrillation (VT/VF) inducibility were measured. The expression of Cx43, Kir2.1, Kir6.2, DHPRα1, KCNH2, caspase3, caspase9, Bax, Bcl2, and P53 was detected. Masson's trichrome, TUNEL, HE, and PAS staining and transmission electron microscopy were used to detect pathological and ultrastructural changes. Our results showed that rotenone alone and rotenone combined with 3-BrPA significantly increased the risk of ventricular arrhythmias. Rotenone combined with 3-BrPA caused myocardial apoptosis, and rotenone alone and rotenone combined with 3-BrPA caused electrical and structural cardiac remodeling in rats.


Antineoplastic Agents/toxicity , Heart Ventricles/drug effects , Insecticides/toxicity , Pyruvates/toxicity , Rotenone/toxicity , Tachycardia, Ventricular/chemically induced , Ventricular Fibrillation/chemically induced , Ventricular Remodeling/drug effects , Action Potentials/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cardiotoxicity , Connexin 43/genetics , Connexin 43/metabolism , Heart Rate/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Heart Ventricles/ultrastructure , Male , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Rats, Wistar , Refractory Period, Electrophysiological/drug effects , Risk Assessment , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/pathology , Ventricular Fibrillation/physiopathology
14.
Cardiovasc Toxicol ; 20(2): 168-175, 2020 04.
Article En | MEDLINE | ID: mdl-31302839

Several studies suggest QT prolongation and torsade de pointes with acetylcholine-esterase inhibitors. We therefore examined the electrophysiologic profile of donepezil, rivastigmine, and galantamine in a sensitive whole-heart model of proarrhythmia. 34 rabbit hearts were isolated and retrogradely perfused employing the Langendorff setup. Hearts were treated either with donepezil, rivastigmine, or galantamine in rising concentrations and electrophysiologic studies were performed. In the presence of donepezil and galantamine, spatial dispersion of repolarization was amplified. Cardiac repolarization (QT interval and action potential duration) was prolonged with donepezil but not with galantamine. Remarkably, both drugs induced triggered activity (early afterdepolarizations and torsade de pointes tachycardia). Despite a pronounced prolongation of repolarization with rivastigmine, no increase in spatial dispersion of repolarization and thus no triggered activity was observed. In the present study, donepezil and galantamine provoked triggered activity, whereas rivastigmine did not have proarrhythmic effects. Spatial dispersion of repolarization but not duration of cardiac repolarization was associated with increased risk of drug-induced proarrhythmia with acetylcholine-esterase inhibitors. Consequently, QT interval duration might be insufficient to estimate the risk of proarrhythmia with acetylcholine-esterase inhibitors. Our findings emphasize the need for further electrocardiographic risk predictors.


Arrhythmias, Cardiac/chemically induced , Cholinesterase Inhibitors/toxicity , Donepezil/toxicity , Galantamine/toxicity , Heart Conduction System/drug effects , Heart Rate/drug effects , Rivastigmine/toxicity , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/physiopathology , Cardiotoxicity , Heart Conduction System/physiopathology , Isolated Heart Preparation , Rabbits , Refractory Period, Electrophysiological/drug effects , Risk Assessment , Time Factors
15.
Cardiovasc Toxicol ; 20(1): 49-57, 2020 02.
Article En | MEDLINE | ID: mdl-31175534

Recent experimental studies showed a protective effect of the renin inhibitor aliskiren regarding atrial structural remodeling. Purpose of this study was to assess acute electrophysiologic effects of aliskiren in a whole-heart model of atrial fibrillation (AF) and to investigate its impact on the ventricle. Twelve rabbit hearts were excised, retrogradely perfused, and paced at different cycle lengths. To enhance atrial vulnerability, a combination of acetylcholine (ACh) and isoproterenol (Iso) was infused and significantly reduced atrial action potential duration (aAPD90) and atrial effective refractory period (aERP). Additional infusion of aliskiren prolonged aAPD90 but did not alter aERP. A triangulation of action potential with ACh/Iso and a further triangulation after treatment with aliskiren were noted. Vulnerability to AF was tested by employing trains of burst pacing. Administration of ACh/Iso provoked more episodes of AF (baseline: 26 episodes, Iso/Ach: 48 episodes). Additional treatment with aliskiren induced AF significantly more often (108 episodes). Another nine hearts were perfused with aliskiren to examine its ventricular effects. Infusion with aliskiren abbreviated ventricular APD90 and ERP. Utilizing programmed ventricular stimulation, a trend towards more ventricular arrhythmias in aliskiren-treated hearts was observed. Though aliskiren did not reduce aAPD90 or aERP, acute treatment with aliskiren promoted AF. Triangulation of atrial action potentials, which is an established risk factor for ventricular proarrhythmia, may contribute to the increased atrial vulnerability. This effect may interfere with its recently demonstrated beneficial properties in atrial remodeling. Of note, aliskiren might have a proarrhythmic effect on the ventricular level.


Action Potentials/drug effects , Amides/toxicity , Atrial Fibrillation/chemically induced , Fumarates/toxicity , Heart Atria/drug effects , Heart Rate/drug effects , Heart Ventricles/drug effects , Acetylcholine , Animals , Atrial Fibrillation/physiopathology , Cardiac Pacing, Artificial , Disease Models, Animal , Female , Heart Atria/physiopathology , Heart Ventricles/physiopathology , Isolated Heart Preparation , Isoproterenol , Rabbits , Refractory Period, Electrophysiological/drug effects , Time Factors
16.
Circ Arrhythm Electrophysiol ; 12(7): e007294, 2019 07.
Article En | MEDLINE | ID: mdl-31248280

BACKGROUND: Phthalates are used as plasticizers in the manufacturing of flexible, plastic medical products. Patients can be subjected to high phthalate exposure through contact with plastic medical devices. We aimed to investigate the cardiac safety and biocompatibility of mono-2-ethylhexyl phthalate (MEHP), a phthalate with documented exposure in intensive care patients. METHODS: Optical mapping of transmembrane voltage and pacing studies were performed on isolated, Langendorff-perfused rat hearts to assess cardiac electrophysiology after MEHP exposure compared with controls. MEHP dose was chosen based on reported blood concentrations after an exchange transfusion procedure. RESULTS: Thirty-minute exposure to MEHP increased the atrioventricular node (147 versus 107 ms) and ventricular (117 versus 77.5 ms) effective refractory periods, compared with controls. Optical mapping revealed prolonged action potential duration at slower pacing cycle lengths, akin to reverse use dependence. The plateau phase of the action potential duration restitution curve steepened and became monophasic in MEHP-exposed hearts (0.18 versus 0.06 slope). Action potential duration lengthening occurred during late-phase repolarization resulting in triangulation (70.3 versus 56.6 ms). MEHP exposure also slowed epicardial conduction velocity (35 versus 60 cm/s), which may be partly explained by inhibition of Nav1.5 (874 and 231 µmol/L half-maximal inhibitory concentration, fast and late sodium current). CONCLUSIONS: This study highlights the impact of acute MEHP exposure, using a clinically relevant dose, on cardiac electrophysiology in the intact heart. Heightened clinical exposure to plasticized medical products may have cardiac safety implications-given that action potential triangulation and electrical restitution modifications are a risk factor for early after depolarizations and cardiac arrhythmias.


Action Potentials/drug effects , Arrhythmias, Cardiac/chemically induced , Diethylhexyl Phthalate/analogs & derivatives , Equipment and Supplies/adverse effects , Heart Conduction System/drug effects , Heart Rate/drug effects , Plasticizers/toxicity , Animals , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Computer Simulation , Diethylhexyl Phthalate/toxicity , Equipment Design , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Humans , Isolated Heart Preparation , Male , Models, Cardiovascular , Rats, Sprague-Dawley , Refractory Period, Electrophysiological/drug effects , Risk Assessment , Sodium Channels/drug effects , Sodium Channels/metabolism , Time Factors , Voltage-Sensitive Dye Imaging
17.
Exp Physiol ; 104(4): 490-504, 2019 04.
Article En | MEDLINE | ID: mdl-30758086

NEW FINDINGS: What is the central question of this study? Are modifications in the restitution of ventricular action potential duration induced by antiarrhythmic drugs the same when assessed with premature extrastimulus application at variable coupling intervals (the standard stimulation protocol) and with steady state pacing at variable rates (the dynamic stimulation protocol)? What is the main finding and its importance? With class I and class III antiarrhythmics, the effects on electrical restitution determined with the standard stimulation protocol dissociate from those obtained during dynamic pacing. These findings indicate a limited value of the electrical restitution assessments based on extrasystolic stimulations alone, as performed in the clinical studies, in estimating the outcomes of antiarrhythmic drug therapies. ABSTRACT: A steep slope of the ventricular action potential duration (APD) to diastolic interval (DI) relationships (the electrical restitution) can precipitate tachyarrhythmia, whereas a flattened slope is antiarrhythmic. The derangements in APD restitution responsible for transition of tachycardia to ventricular fibrillation can be assessed with cardiac pacing at progressively increasing rates (the dynamic stimulation protocol). Nevertheless, this method is not used clinically owing to the risk of inducing myocardial ischaemia. Instead, the restitution kinetics is determined with a premature extrastimulus application at variable coupling intervals (the standard stimulation protocol). Whether the two protocols are equivalent in estimating antiarrhythmic drug effects is uncertain. In this study, dofetilide and quinidine, the agents blocking repolarizing K+ currents, increased epicardial APD in perfused guinea-pig hearts, with effects being greater at long vs. short DIs. These changes were more pronounced during dynamic pacing compared to premature extrastimulations. Accordingly, although both agents markedly steepened the dynamic restitution, there was only a marginal increase in the standard restitution slope with dofetilide, and no effect with quinidine. Lidocaine and mexiletine, selective Na+ channel blockers, prolonged the effective refractory period without changing APD, and increased the minimum DI that enabled ventricular capture during extrastimulations. No change in the minimum DI was noted during dynamic pacing. Consequently, although lidocaine and mexiletine reduced the standard restitution slope, they failed to flatten the dynamic restitution. Overall, these findings imply a limited value of the electrical restitution assessments with premature extrastimulations alone in discriminating arrhythmic vs. antiarrhythmic changes during drug therapies.


Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Heart/drug effects , Refractory Period, Electrophysiological/drug effects , Ventricular Fibrillation/drug therapy , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/metabolism , Cardiac Pacing, Artificial/methods , Electrocardiography/methods , Female , Guinea Pigs , Heart Conduction System/drug effects , Heart Conduction System/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Phenethylamines/pharmacology , Quinidine/pharmacology , Sulfonamides/pharmacology , Ventricular Fibrillation/metabolism
18.
Cardiovasc Toxicol ; 19(3): 237-243, 2019 06.
Article En | MEDLINE | ID: mdl-30515668

A post hoc analysis of the PALLAS trial suggested life-threatening interactions of digitalis and dronedarone. Thus, there is concern about an interplay between digitalis and other drugs that influence cardiac electrophysiology. We therefore investigated the interaction between digitalis and flecainide or ranolazine. Twenty-five rabbit hearts were Langendorff-perfused and treated with flecainide (2 µM, 12 hearts) or ranolazine (10 µM, 13 hearts). Infusion of flecainide prolonged mean action potential duration [APD90, from 153 ms (interquartile range (IQR): 29.7 ms) to 159 ms (IQR: 24.9 ms, p = 0.04)] and effective refractory period [ERP, 170 ms (IQR: 40 ms) vs. 200 ms (IQR: 32.5 ms, p < 0.01)]. Administration of ranolazine prolonged APD90 [144 ms (IQR: 34.3 ms)) vs. 157 ms (IQR: 31.2 ms, p < 0.01)] and ERP [180 ms (IQR: 40 ms) vs. 200 ms (IQR: 30 ms, p < 0.01)]. Additional infusion of the digitalis glycoside ouabain (0.2 µM) abbreviated APD90 and ERP in both groups (flecainide: APD90: to 128 ms (IQR: 19 ms), ERP: to 170 ms (IQR: 20 ms), p < 0.01 each; ranolazine: APD90: to 141 ms (IQR: 40 ms), ERP: to 160 ms (IQR: 30 ms), p < 0.01 each). Ventricular vulnerability was assessed by a pacing protocol employing premature extra stimuli and burst stimulation. No proarrhythmic effect was observed with flecainide (1 vs. 3 episodes at baseline) or ranolazine (3 vs. 11 episodes at baseline). However, further infusion of ouabain had a proarrhythmic effect for both drugs (flecainide: 15 episodes, p = 0.04; ranolazine: 21 episodes, p = 0.09). Concomitant treatment of the sodium channel blockers flecainide or ranolazine with digitalis seems to be proarrhythmic. Abbreviation of repolarization and refractoriness that can facilitate re-entry was found as underlying mechanism.


Anti-Arrhythmia Agents/toxicity , Arrhythmias, Cardiac/chemically induced , Digitalis Glycosides/toxicity , Flecainide/toxicity , Heart Rate/drug effects , Ouabain/toxicity , Ranolazine/toxicity , Voltage-Gated Sodium Channel Blockers/toxicity , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/physiopathology , Cardiotoxicity , Drug Interactions , Isolated Heart Preparation , Rabbits , Refractory Period, Electrophysiological/drug effects , Risk Assessment , Time Factors
19.
Pharmacology ; 103(1-2): 38-49, 2019.
Article En | MEDLINE | ID: mdl-30380560

BACKGROUND/AIMS: In this study, we evaluated the functional impact of facilitatory presynaptic adenosine A2A and muscarinic M1 receptors in the recovery of neuromuscular tetanic depression caused by the blockage of high-affinity choline transporter (HChT) by hemicholinium-3 (HC-3), a condition that mimics a myasthenia-like condition. METHODS: Rat diaphragm preparations were indirectly stimulated via the phrenic nerve trunk with 50-Hz frequency trains, each consisting of 500-750 supramaximal intensity pulses. The tension at the beginning (A) and at the end (B) of the tetanus was recorded and the ratio (R) B/A calculated. RESULTS: Activation of A2A and M1 receptors with CGS21680 (CGS; 2 nmol/L) and McN-A-343c (McN; 3 µmol/L) increased R values. Similar facilitatory effects were obtained with forskolin (FSK; 3 µmol/L) and phorbol 12-myristate 13-acetate (PMA; 10 µmol/L), which activate adenylate cyclase and protein kinase C respectively. HC-3 (4 µmol/L) decreased transmitter exocytosis measured by real-time videomicroscopy with the FM4-64 fluorescent dye and prevented the facilitation of neuromuscular transmission caused by CGS, McN, and FSK, with a minor effect on PMA. The acetylcholinesterase inhibitor, neostigmine (NEO; 0.5 µmol/L), also decreased transmitter exocytosis. The paradoxical neuromuscular tetanic fade caused by NEO (0.5 µmol/L) was also prevented by HC-3 (4 µmol/L) and might result from the rundown of the positive feedback mechanism operated by neuronal nicotinic receptors (blocked by hexamethonium, 120 µmol/L). CONCLUSION: Data suggest that the recovery of tetanic neuromuscular facilitation by adenosine A2A and M1 receptors is highly dependent on HChT activity and may be weakened in myasthenic patients when HChT is inoperative.


Membrane Transport Proteins/physiology , Receptor, Adenosine A2A/physiology , Receptor, Muscarinic M1/physiology , Refractory Period, Electrophysiological/drug effects , (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Colforsin/pharmacology , Diaphragm/drug effects , Diaphragm/physiology , Hemicholinium 3/pharmacology , Neostigmine/pharmacology , Phenethylamines/pharmacology , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Rats , Rats, Wistar , Synaptic Transmission , Tetanus/drug therapy , Tetanus/physiopathology , Tetradecanoylphorbol Acetate/pharmacology
20.
PLoS One ; 13(12): e0209085, 2018.
Article En | MEDLINE | ID: mdl-30562383

A study has been made of the effect of chronic exercise on myocardial electrophysiological heterogeneity and stability, as well as of the role of cholinergic neurons in these changes. Determinations in hearts from untrained and trained rabbits on a treadmill were performed. The hearts were isolated and perfused. A pacing electrode and a recording multielectrode were located in the left ventricle. The parameters determined during induced VF, before and after atropine (1µM), were: fibrillatory cycle length (VV), ventricular functional refractory period (FRPVF), normalized energy (NE) of the fibrillatory signal and its coefficient of variation (CV), and electrical ventricular activation complexity, as an approach to myocardial heterogeneity and stability. The VV interval was longer in the trained group than in the control group both prior to atropine (78±10 vs. 68±10 ms) and after atropine (76±8 vs. 67±10 ms). Likewise, FRPVF was longer in the trained group than in the control group both prior to and after atropine (53±8 vs. 42±7 ms and 50±6 vs. 40±6 ms, respectively), and atropine did not modify FRPVF. The CV of FRPVF was lower in the trained group than in the control group prior to atropine (12.5±1.5% vs. 15.1±3.8%) and, decreased after atropine (15.1±3.8% vs. 12.2±2.4%) in the control group. The trained group showed higher NE values before (0.40±0.04 vs. 0.36±0.05) and after atropine (0.37±0.04 vs. 0.34±0.06; p = 0.08). Training decreased the CV of NE both before (23.3±2% vs. 25.2±4%; p = 0.08) and after parasympathetic blockade (22.6±1% vs. 26.1±5%). Cholinergic blockade did not modify these parameters within the control and trained groups. Activation complexity was lower in the trained than in the control animals before atropine (34±8 vs. 41±5), and increased after atropine in the control group (41±5 vs. 48±9, respectively). Thus, training decreases the intrinsic heterogeneity of the myocardium, increases electrophysiological stability, and prevents some modifications due to muscarinic block.


Heart/physiology , Running/physiology , Animals , Atropine/pharmacology , Heart/drug effects , Male , Muscarinic Antagonists/pharmacology , Parasympatholytics/pharmacology , Rabbits , Refractory Period, Electrophysiological/drug effects , Tissue Culture Techniques , Ventricular Fibrillation/physiopathology
...