Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 15(2): e0228818, 2020.
Article in English | MEDLINE | ID: mdl-32084145

ABSTRACT

INTRODUCTION: High beat-to-beat morphological variation (divergence) on the ventricular electrogram during programmed ventricular stimulation (PVS) is associated with increased risk of ventricular fibrillation (VF), with unclear mechanisms. We hypothesized that ventricular divergence is associated with epicardial wavebreaks during PVS, and that it predicts VF occurrence. METHOD AND RESULTS: Langendorff-perfused rabbit hearts (n = 10) underwent 30-min therapeutic hypothermia (TH, 30°C), followed by a 20-min treatment with rotigaptide (300 nM), a gap junction modifier. VF inducibility was tested using burst ventricular pacing at the shortest pacing cycle length achieving 1:1 ventricular capture. Pseudo-ECG (p-ECG) and epicardial activation maps were simultaneously recorded for divergence and wavebreaks analysis, respectively. A total of 112 optical and p-ECG recordings (62 at TH, 50 at TH treated with rotigaptide) were analyzed. Adding rotigaptide reduced ventricular divergence, from 0.13±0.10 at TH to 0.09±0.07 (p = 0.018). Similarly, rotigaptide reduced the number of epicardial wavebreaks, from 0.59±0.73 at TH to 0.30±0.49 (p = 0.036). VF inducibility decreased, from 48±31% at TH to 22±32% after rotigaptide infusion (p = 0.032). Linear regression models showed that ventricular divergence correlated with epicardial wavebreaks during TH (p<0.001). CONCLUSION: Ventricular divergence correlated with, and might be predictive of epicardial wavebreaks during PVS at TH. Rotigaptide decreased both the ventricular divergence and epicardial wavebreaks, and reduced the probability of pacing-induced VF during TH.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Heart Ventricles/physiopathology , Hypothermia, Induced/adverse effects , Pericardium/physiology , Animals , Electrocardiography , Heart Ventricles/drug effects , Oligopeptides/pharmacology , Pericardium/drug effects , Pericardium/physiopathology , Rabbits
2.
Heart Rhythm ; 13(1): 251-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26188250

ABSTRACT

BACKGROUND: Therapeutic hypothermia (TH) may increase the susceptibility to ventricular arrhythmias by decreasing ventricular conduction velocity (CV) and facilitating arrhythmogenic spatially discordant alternans (SDA). OBJECTIVE: The purpose of this study was to test the hypothesis that rotigaptide, a gap junction enhancer, can increase ventricular CV, delay the onset of SDA, and decrease the susceptibility to pacing-induced ventricular fibrillation (PIVF) during TH. METHODS: Langendorff-perfused isolated rabbit hearts were subjected to 30-minute moderate hypothermia (33°C) followed by 20-minute treatment with rotigaptide (300 nM, n = 8) or vehicle (n = 5). The same protocol was also performed at severe hypothermia (30°C; n = 8 for rotigaptide, n = 5 for vehicle). Using an optical mapping system, epicardial CV and SDA threshold were evaluated by S1 pacing. Ventricular fibrillation inducibility was evaluated by burst pacing for 30 seconds at the shortest pacing cycle length (PCL) that achieved 1:1 ventricular capture. RESULTS: Rotigaptide increased ventricular CV during 33°C (PCL 300 ms, from 76 ± 6 cm/s to 84 ± 7 cm/s, P = .039) and 30°C (PCL 300 ms, from 62 ± 6 cm/s to 68 ± 4 cm/s, P = .008). Rotigaptide decreased action potential duration dispersion at 33°C (P = .01) and 30°C (P = .035). During 30°C, SDA thresholds (P = .042) and incidence of premature ventricular complexes (P = .025) were decreased by rotigaptide. PIVF inducibility was decreased by rotigaptide at 33°C (P = .039) and 30°C (P = .042). Rotigaptide did not change connexin43 expressions and distributions during hypothermia. CONCLUSION: Rotigaptide protects the hearts against ventricular arrhythmias by increasing ventricular CV, delaying the onset of SDA, and reducing repolarization heterogeneity during TH. Enhancing cell-to-cell coupling by rotigaptide might be a novel approach to prevent ventricular arrhythmias during TH.


Subject(s)
Connexin 43/metabolism , Gap Junctions , Heart Ventricles , Hypothermia, Induced/adverse effects , Oligopeptides/pharmacology , Ventricular Fibrillation , Animals , Anti-Arrhythmia Agents/pharmacology , Gap Junctions/drug effects , Gap Junctions/metabolism , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Models, Cardiovascular , Rabbits , Treatment Outcome , Ventricular Fibrillation/etiology , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/physiopathology , Ventricular Fibrillation/prevention & control , Voltage-Sensitive Dye Imaging/methods
3.
Biochem J ; 441(1): 511-22, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21880012

ABSTRACT

The peptides encoded by the VGF gene are gaining biomedical interest and are increasingly being scrutinized as biomarkers for human disease. An endocrine/neuromodulatory role for VGF peptides has been suggested but never demonstrated. Furthermore, no study has demonstrated so far the existence of a receptor-mediated mechanism for any VGF peptide. In the present study, we provide a comprehensive in vitro, ex vivo and in vivo identification of a novel pro-lipolytic pathway mediated by the TLQP-21 peptide. We show for the first time that VGF-immunoreactivity is present within sympathetic fibres in the WAT (white adipose tissue) but not in the adipocytes. Furthermore, we identified a saturable receptor-binding activity for the TLQP-21 peptide. The maximum binding capacity for TLQP-21 was higher in the WAT as compared with other tissues, and selectively up-regulated in the adipose tissue of obese mice. TLQP-21 increases lipolysis in murine adipocytes via a mechanism encompassing the activation of noradrenaline/ß-adrenergic receptors pathways and dose-dependently decreases adipocytes diameters in two models of obesity. In conclusion, we demonstrated a novel and previously uncharacterized peripheral lipolytic pathway encompassing the VGF peptide TLQP-21. Targeting the sympathetic nerve-adipocytes interaction might prove to be a novel approach for the treatment of obesity-associated metabolic complications.


Subject(s)
Neuropeptides/metabolism , Peptide Fragments/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Animals , Body Composition , Dietary Fats/adverse effects , Dietary Fats/metabolism , Male , Mice , NIH 3T3 Cells , Nerve Growth Factors , Obesity/chemically induced , Obesity/metabolism , Protein Binding , Protein Transport , Receptors, Cell Surface
4.
Bioorg Med Chem Lett ; 19(16): 4551-4, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19616941

ABSTRACT

In an effort to discover potent, orally bioavailable compounds for the treatment of atrial fibrillation (AF) and ventricular tachycardia (VT), we developed a class of gap-junction modifiers typified by GAP-134 (1, R(1)=OH, R(2)=NH(2)), a compound currently under clinical evaluation. Selected compounds with the desired in-vitro profile demonstrated positive in vivo results in the mouse CaCl(2) arrhythmia model upon oral administration.


Subject(s)
Anti-Arrhythmia Agents/chemistry , Benzamides/chemistry , Gap Junctions/drug effects , Proline/analogs & derivatives , Administration, Oral , Animals , Anti-Arrhythmia Agents/pharmacokinetics , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Atrial Fibrillation/drug therapy , Benzamides/pharmacokinetics , Benzamides/pharmacology , Disease Models, Animal , Dogs , Drug Discovery , Mice , Proline/chemistry , Proline/pharmacokinetics , Proline/pharmacology , Rats , Structure-Activity Relationship , Tachycardia, Ventricular/drug therapy
5.
J Pharmacol Exp Ther ; 317(1): 236-43, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16344331

ABSTRACT

The antiarrhythmic and cardioprotective effect of increasing gap junction intercellular communication during ischemia/reperfusion injury has not been studied. The antiarrhythmic peptide rotigaptide (previously ZP123), which maintains gap junction intercellular communication, was tested in dogs subjected to a 60-min coronary artery occlusion and 4 h of reperfusion. Rotigaptide was administered i.v. 10 min before reperfusion as a bolus + i.v. infusion at doses of 1 ng/kg bolus + 10 ng/kg/h infusion (n = 6), 10 ng/kg bolus + 100 ng/kg/h infusion (n = 5), 100 ng/kg bolus + 1000 ng/kg/h infusion (n = 8), 1000 ng/kg bolus + 10 mug/kg/h infusion (n = 6), and vehicle control (n = 5). Premature ventricular complexes (PVCs) were quantified during reperfusion. A series of four or more consecutive PVCs was defined as ventricular tachycardia (VT). The total incidence of VT was reduced significantly with the two highest doses of rotigaptide (20.3 +/- 10.9 and 4.3 +/- 4.1 events; p < 0.05) compared with controls (48.7 +/- 6.0). Total PVCs were reduced significantly from 25.1 +/- 4.2% in control animals to 11.0 +/- 4.4 and 1.7 +/- 1.3% after the two highest doses of rotigaptide. Infarct size, expressed as a percentage of the left ventricle, was reduced significantly from 13.2 +/- 1.9 in controls to 7.1 +/- 1.0 (p < 0.05) at the highest dose of rotigaptide. Ultrastructural evaluation revealed no differences in myocardial injury in the infarct area, area at risk, border zone, or normal zone in vehicle and rotigaptide-treated animals. However, rotigaptide did increase the presence of gap junctions in the area at risk (p = 0.022, Fisher's exact test). Rotigaptide had no effect on heart rate, blood pressure, heart rate-corrected QT interval, or left ventricular end-diastolic pressure. In conclusion, these results demonstrate that rotigaptide is a potent antiarrhythmic compound with cardioprotective effects and desirable safety.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/complications , Oligopeptides/therapeutic use , Ventricular Premature Complexes/prevention & control , Animals , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/pharmacokinetics , Dogs , Gap Junctions/ultrastructure , Hemodynamics/drug effects , Microscopy, Electron, Transmission , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/ultrastructure , Oligopeptides/adverse effects , Oligopeptides/pharmacokinetics , Treatment Outcome , Ventricular Premature Complexes/etiology , Ventricular Premature Complexes/pathology , Ventricular Premature Complexes/physiopathology
6.
Cell Commun Adhes ; 10(4-6): 371-8, 2003.
Article in English | MEDLINE | ID: mdl-14681044

ABSTRACT

Antiarrhythmic peptides such as AAP10 (Gly-Ala-Gly-4Hyp-Pro-Tyr-CONH(2)) have antiarrhythmic properties related to their stimulatory effect on gap junctional coupling. However, most of these peptides are not stable in enzymatic environment which limits studies with these compounds in vivo. ZP123 is a new antiarrhythmic peptide constructed using a retro-all-D-amino acid design of the AAP10 template (Ac-D-Tyr-D-Pro-D-4Hyp-Gly-D-Ala-Gly-NH(2)). The aim of this study was to compare the effects of AAP10 and ZP123 on epicardial activation and repolarization patterns in isolated perfused rabbit hearts. In addition, we tested the effect of these compounds on PKC activation in cultured HeLa-Cx43 cells. Rabbit hearts were perfused according to the Langendorff technique with Tyrode solution at constant pressure (70 cm H(2)O). After 45 min equilibration, either AAP10 (n = 7) or ZP123 (n = 7) was infused intracoronarily in concentrations of 0.1, 1, 10, 100, and 1000 nM (15 min for each concentration) in the presence of 0.05% bovine serum albumine. 256 AgCl electrodes were attached to the hearts surface and connected to the inputs of a 256 channel mapping system in a unipolar circuit (4 kHz/channel, 0.04 mV vertical resolution, 1 mm spatial resolution). For each electrode the activation and repolarization timepoint were determined. We found that both peptides significantly reduced epicardial dispersion by a maximum of about 20% thereby enhancing the homogeneity of epicardial action potential duration, while the action potential duration itself was not affected. The beat-to-beat variability of the epicardial activation pattern was stabilized by both peptides as compared to an untreated time-control series. Other parameters such as LVP, CF, heart rate, or total activation time were not effected by either of the peptides. In a second protocol, rectangular pulses were delivered to the back wall and the propagation velocity was determined longitudinal and transversal to the fiber axis. We found an increase in both longitudinal and transversal conduction velocity. Using a commercial PKC assay on HeLa-Cx43 cells we found that 50 nM AAP10 and 50 nM ZP123 increased activity by 99 +/- 6% and 146 +/- 54%, respectively. The PKC activation induced by either of these compounds was completely blocked using the selective PKCalpha inhibitor GCP54345. We conclude that AAP10 and ZP123 have similar effects in vitro, but the superior enzymatic stability of ZP123 makes this compound the preferred substance for in vivo studies of antiarrhythmic peptides.


Subject(s)
Heart Conduction System/drug effects , Heart/physiopathology , Myocardial Contraction/drug effects , Oligopeptides/pharmacology , Protein Kinase C/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Connexin 43/metabolism , Electrocardiography , Enzyme Inhibitors/pharmacology , Gap Junctions/physiology , HeLa Cells , Heart/drug effects , Humans , Male , Protein Denaturation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha , Rabbits , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...