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










Publication year range
1.
Physiol Res ; 64(4): 495-503, 2015.
Article in English | MEDLINE | ID: mdl-25470517

ABSTRACT

Atrial natriuretic peptide antifibrotic properties are mainly described in cardiac myocytes or in induced cardiac myofibroblasts (Angiotensin II or TGF-beta induced differentiation). In the present work, we investigate the effects of ANP/NPRA/cGMP system in modulating rat cardiac fibroblasts function. Cardiac fibroblasts were isolated from adult Wistar male rats and cultured in the presence of serum in order to induce fibroblasts differentiation. Cultures were then treated with ANP (1 microM), 8-Br-cGMP (100 microM) or IBMX (100 microM), a non-specific phosphodiesterases inhibitor. ANP significantly decreased proliferation rate and collagen secretion. Its effect was mimicked by the cGMP analog, while combining ANP with 8-Br-cGMP did not lead to additional effects. Moreover intracellular cGMP levels were elevated when cells were incubated with ANP confirming that ANP intracellular pathway is mediated by cGMP. Additionally, immunoblotting and immunofluorescence were used to confirm the presence of guanylyl cyclase specific natriuretic peptide receptors A and B. Finally we scanned specific cGMP dependent PDEs via RT-qPCR, and noticed that inhibiting all PDEs led to an important decrease in proliferation rate. Effect of ANP became more prominent after 10 culture days, confirming the importance of ANP in fibroblasts to myofibroblasts differentiation. Uncovering cellular aspects of ANP/NPRA/cGMP signaling system provided more elements to help understand cardiac fibrotic process.


Subject(s)
Atrial Natriuretic Factor/administration & dosage , Cell Differentiation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Heart Ventricles/cytology , Myofibroblasts/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Heart Ventricles/drug effects , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myofibroblasts/drug effects , Rats , Rats, Wistar
2.
Br J Pharmacol ; 159(1): 93-101, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19703166

ABSTRACT

INTRODUCTION: Doxorubicin, an anthracycline widely used in the treatment of a broad range of tumours, causes acute QT prolongation. Dexrazoxane has been shown to prevent the QT prolongation induced by another anthracycline, epirubicin, but has not yet been reported to prevent that induced by doxorubicin. Thus, the present study was designed to test whether the acute QT effects induced by doxorubicin could be blocked by dexrazoxane and to explore the mechanism. Results were compared with those obtained with a reference human ether-a-go-go (hERG) channel blocker, moxifloxacin. METHODS: The effects of moxifloxacin (100 microM) and doxorubicin (30 microM), with or without dexrazoxane (from 3 to 30 microM), have been evaluated on the QTc interval in guinea-pig isolated hearts and on I(Kr) (rapid component of the delayed rectifier current) and I(Ks) (slow component of the delayed rectifier current) currents stably expressed in human embryonic kidney 293 cells. RESULTS: Moxifloxacin (100 microM), a potent hERG blocker, prolonged QTc by 22%, and this effect was not prevented by dexrazoxane. Doxorubicin (30 microM) also prolonged QTc by 13%, did not significantly block hERG channels and specifically inhibited I(Ks) (IC(50): 4.78 microM). Dexrazoxane significantly reduced the doxorubicin-induced QTc prolongation and prevented doxorubicin-induced inhibition of I(Ks). CONCLUSION AND IMPLICATIONS: Doxorubicin acutely prolonged the QT interval in guinea-pig heart by selective I(Ks) blockade. This effect was prevented by dexrazoxane. This result is important because it illustrates the danger of neglecting I(Ks) in favour of hERG screening alone, for early preclinical testing for possible induction of torsade de pointes.


Subject(s)
Cardiovascular Agents/pharmacology , Doxorubicin/adverse effects , Long QT Syndrome/prevention & control , Razoxane/pharmacology , Animals , Antibiotics, Antineoplastic/adverse effects , Aza Compounds/adverse effects , Cardiovascular Agents/administration & dosage , Cell Line , Delayed Rectifier Potassium Channels/drug effects , Delayed Rectifier Potassium Channels/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Fluoroquinolones , Guinea Pigs , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Long QT Syndrome/chemically induced , Moxifloxacin , Quinolines/adverse effects , Razoxane/administration & dosage
3.
Biochem Biophys Res Commun ; 281(2): 452-60, 2001 Feb 23.
Article in English | MEDLINE | ID: mdl-11181069

ABSTRACT

Relative expression pattern of short and long isoforms of hKv4.3 channels was evaluated by RT-PCR and RPA. Electrophysiological studies were performed in HEK293 cells transfected with short or long hKv4.3 cDNA. The long variant L-hKv4.3 was the only form present in lung, pancreas, and small intestine. The short variant S-hKv4.3 was predominant in brain whereas expression levels of the two isoforms were similar in cardiac and skeletal muscles. Properties of the ionic channels encoded by L-hKv4.3 and S-hKv4.3 cDNAs were essentially similar. Cadmium chloride and verapamil inhibited hKv4.3 current (with EC50s of 0.110 +/- 0.004 mM and 492.9 +/- 15.1 microM, respectively). Verapamil also accelerated current inactivation. Another calcium channel antagonist nicardipine was found inactive. In conclusion, this study confirms that both isoforms underlie the transient outward potassium current. Moreover, calcium channel inhibitors markedly affect hKv4.3 current, an effect which must be considered when evaluating transient outward potassium channel properties in native tissues.


Subject(s)
Calcium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Cadmium Chloride/pharmacology , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Membrane Potentials/drug effects , Potassium Channels/genetics , Potassium Channels/physiology , Protein Isoforms/drug effects , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Shal Potassium Channels , Time Factors , Tissue Distribution , Verapamil/pharmacology
4.
Eur J Pharmacol ; 383(2): 215-22, 1999 Oct 27.
Article in English | MEDLINE | ID: mdl-10585537

ABSTRACT

Amongst the different pharmacological approaches to the treatment of cardiac arrhythmias, compounds with multiple electrophysiological activities appear to exhibit a reduced adverse effect profile. BRL-32872 (N-(3,4-dimethoxyphenyl)-N-[3[[2-(3,4-dimethoxyphenyl) ethyl] propyl]-4-nitrobenzamide hydrochloride) is a typical example of an antiarrhythmic agent with combined K(+) and Ca(2+) blocking actions. In this study, we investigated the effects of BRL-32872 on early after-depolarizations and on dispersion of repolarization. Action potentials were recorded either in canine cardiac Purkinje fibers alone or in preparations containing both ventricular muscle and the attached Purkinje fibers. In Purkinje fibers, BRL-32872 (0. 3-10 microM) induced a bell-shaped concentration-dependent increase in action potential duration. At 90% of repolarization, the action potential was prolonged at concentrations up to 1 microM and was shortened when the concentration of BRL-32872 was further increased. In all 17 experiments, BRL-32872 did not cause early after-depolarizations in Purkinje fibers. On the contrary, BRL-32872 (3 microM) systematically suppressed early after-depolarizations induced by clofilium (4-chloro-N, N-diethyl-N-heptylbenzenebutanaminium tosylate, 1 microM), a selective inhibitor of the delayed rectifier K(+) current. A similar effect was observed once with 1 microM BRL-32872, a concentration able to prolong Purkinje fiber action potentials. Simultaneous recording of action potentials in ventricular and Purkinje preparations showed that increasing concentrations of BRL-32872 (0. 3-10 microM) induced a limited increase in the difference of repolarization time between the two tissues. The selective K(+) channel inhibitor E-4031 (N-(4-(1-[2-(6-methyl-2-pyridyl) ethyl]-4-piperidyl)-carbonyl] phenyl) methanesulfonamide dihydrochloride dihydrate) exhibited a significant concentration-dependent increase in dispersion of repolarization. We conclude from the present results that the Ca(2+) blocking activity of BRL-32872 (i) prevents the occurrence of early after-depolarizations associated with action potential prolongation and (ii) limits an excessive increase in action potential duration heterogeneity. These electrophysiological features might represent the basis for antiarrhythmic compounds with reduced proarrhythmic profile.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzamides/pharmacology , Heart/drug effects , Purkinje Fibers/drug effects , Action Potentials/drug effects , Animals , Dogs , Electrophysiology , Female , Heart/innervation , Heart Ventricles/drug effects , Heart Ventricles/innervation , In Vitro Techniques , Kinetics , Male , Piperidines/pharmacology , Potassium Channel Blockers , Pyridines/pharmacology , Quaternary Ammonium Compounds/pharmacology
5.
J Biol Chem ; 274(51): 36592-600, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593960

ABSTRACT

Starting with computational tools that search for tissue-selective expression of assembled expressed sequenced tags, we have identified by focusing on heart libraries a novel small stress protein of 170 amino acids that we named cvHsp. cvHsp was found as being computationally selectively and highly (0.3% of the total RNA) expressed in human heart. The cvHsp gene mapped to 1p36.23-p34.3 between markers D1S434 and D1S507. The expression of cvHsp was analyzed with RNA dot, Northern blots, or reverse transcription-polymerase chain reaction: expression was high in heart, medium in skeletal muscle, and low in aorta or adipose tissues. In the heart of rat models of cardiac pathologies, cvHsp mRNA expression was either unchanged (spontaneous hypertension), up-regulated (right ventricular hypertrophy induced by monocrotaline treatment), or down-regulated (left ventricular hypertrophy following aortic banding). In obese Zucker rats, cvHsp mRNA was increased in skeletal muscle, brown, and white adipose tissues but remained unchanged in the heart. Western blot analysis using antipeptide polyclonal antibodies revealed two specific bands at 23 and 25 kDa for cvHsp in human heart. cvHsp interacted in both yeast two-hybrid and immunoprecipitation experiments with alpha-filamin or actin-binding protein 280. Within cvHsp, amino acid residues 56-119 were shown to be important for its specific interaction with the C-terminal tail of alpha-filamin.


Subject(s)
Cardiovascular System/metabolism , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Insulin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Gene Expression Regulation , Humans , Molecular Sequence Data , Organ Specificity , Rats , Sequence Alignment
6.
Cardiovasc Res ; 41(1): 188-99, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10325966

ABSTRACT

OBJECTIVE: The Shal (or Kv4) gene family has been proposed to be responsible for primary subunits of the transient outward potassium current (Ito). More precisely, Kv4.2 and Kv4.3 have been suggested to be the most likely molecular correlates for Ito in rat cells. The purpose of the present study was to compare the properties of the rat Kv4.3 gene product when expressed in a human cell line (HEK293 cells) with that of Ito recorded from rat ventricular cells. METHODS: The cDNA encoding the rat Kv4.3 potassium channel was cloned into the pHook2 mammalian expression vector and expressed into HEK293. Patch clamp experiments using the whole cell configuration were used to characterise the electrophysiological parameters of the current induced by Kv4.3 in comparison with the rat ventricular myocyte Ito current. RESULTS: The transfection of HEK293 cells with rat Kv4.3 resulted in the expression of a time- and voltage-dependent outward potassium current. The current activated for potentials positive to -40 mV and the steady-state inactivation curve had a midpoint of -47.4 +/- 0.3 mV and a slope of 5.9 +/- 0.2 mV. Rat ventricular Ito current was activated at potentials positive to -20 mV and inactivated with a half-inactivation potential and a Boltzmann factor of -29.1 +/- 0.7 mV and 4.5 +/- 0.5 mV, respectively. The time course of recovery from inactivation of rat Kv4.3 expressed in HEK293 cells and of Ito recorded from native rat ventricular cells were exponentials with time constants of 213.2 +/- 4.1 msec and 23. +/- 1.5 msec, respectively. Pharmacologically, Ito of rat myocytes showed a greater sensitivity to 4-aminopyridine than Kv4.3 since half-maximal effects were obtained with 1.54 +/- 0.13 mM and 0.14 +/- 0.02 mM on Kv4.3 and Ito, respectively. In both Kv4.3 and Ito, 4-aminopyridine appears to bind to the closed state of the channel. Finally, although a higher level of expression was observed in the atria compared to the ventricle, the distribution of the Kv4.3 gene across the ventricles appeared to be homogeneous. CONCLUSION: The results of the present study show that Kv4.3 channel may play a major role in the molecular structure of the rat cardiac Ito current. Furthermore, because the distribution of Kv4.3 across the ventricle is homogeneous, the blockade of this channel by specific drugs may not alter the normal heterogeneity of Ito current.


Subject(s)
Kidney/metabolism , Myocardium/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , 4-Aminopyridine/pharmacology , Analysis of Variance , Animals , Cadmium/pharmacology , Cell Line , Gene Expression , Gene Transfer Techniques , Heart Atria , Heart Ventricles , Humans , Male , Patch-Clamp Techniques , Potassium Channel Blockers , RNA, Messenger/analysis , Rats , Rats, Wistar , Shal Potassium Channels
7.
Cardiovasc Res ; 37(1): 130-40, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9539867

ABSTRACT

OBJECTIVE: The effects of BRL-32872, azimilide and a selective blocker of the delayed rectifier potassium current, E-4031, were measured at two different basic cycle lengths (BCL), 300 and 1000 ms. Calcium channel antagonists of sarcolemmal (verapamil and nitrendipine) and sarcoplasmic reticulum (ryanodine) membranes were used to investigate whether the inhibition of the calcium current or the calcium release from the sarcoplasmic reticulum could alter the reverse-rate dependence of E-4031 on action potential duration (APD). METHODS: Guinea pig isolated papillary muscles were superfused with a Tyrode solution maintained at 37 degrees C and stimulated at a BCL of 300 or 1000 ms. The standard microelectrode technique was used to record action potential parameters and to study the effects of azimilide, BRL-32872 and E-4031. E-4031 was superfused at increasing concentrations (0.01, 0.03, 0.1 and 0.3 microM) in the absence or in the presence of verapamil (0.3 microM), nitrendipine (0.03 microM) or ryanodine (0.1 microM). RESULTS: BRL-32872 and azimilide induced a self-limited concentration-dependent increase in APD. The effect of BRL-32872 was not dependent on the stimulation frequency whereas the effect of azimilide was significantly reduced at the shorter BCL. E-4031 induced a concentration-dependent increase in APD at both stimulation BCL. The increase in APD was significantly more pronounced in fibres stimulated at a BCL of 1000 ms than in fibres stimulated at a BCL of 300 ms, characterising the reverse-frequency dependent effect of class III antiarrhythmic agents. The reverse-frequency dependence in action potential prolongation induced by E-4031 was significantly reduced in the presence of a low concentration of verapamil (0.3 microM), nitrendipine (0.03 microM), or ryanodine (0.1 microM. CONCLUSION: The results show that BRL-32872, in contrast to azimilide, does not induce the reverse-rate dependency of action potential prolongation typically produced by class III antiarrhythmic agents such as E-4031. Our results also show that reverse-rate dependency induced by E-4031 can be reduced by the simultaneous administration of a low concentration of a calcium channel antagonist or an inhibitor of the release of calcium from the sarcoplasmic reticulum. It is thus suggested that compounds with a suitable balance of potassium and calcium antagonistic activities may have less adverse effects than purely selective potassium channel blockers.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzamides/pharmacology , Calcium Channel Blockers/pharmacology , Imidazoles/pharmacology , Imidazolidines , Piperazines/pharmacology , Potassium Channel Blockers , Action Potentials/drug effects , Animals , Electric Stimulation , Guinea Pigs , Hydantoins , In Vitro Techniques , Male , Membrane Potentials/drug effects , Nitrendipine/pharmacology , Papillary Muscles , Piperidines/pharmacology , Pyridines/pharmacology , Random Allocation , Ryanodine/pharmacology , Verapamil/pharmacology
8.
J Cardiovasc Pharmacol ; 31(4): 551-7, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9554804

ABSTRACT

The modulation of the protein kinase A-activated chloride current (PKA-I[Cl]) may lead to modification of the cardiac action potential shape. The purpose of this study was to evaluate the effects of glibenclamide, tedisamil, dofetilide, E-4031, and BRL-32872 on the PKA-I(Cl). Experiments were conducted by using the patch-clamp technique in guinea pig ventricular myocytes. PKA-I(Cl) was activated by application of 1 microM isoproterenol and was inhibited by 1 microM propranolol, 10 microM acetylcholine, or 1 mM 4-acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic acid (SITS). The sulfonylurea receptor inhibitor, glibenclamide, inhibited PKA-I(Cl) at micromolar concentration. Among class III antiarrhythmic agents, tedisamil induced a dose-dependent inhibition of PKA-I(Cl) with a half effective concentration (EC50) of 7.15 microM (Hill coefficient, 0.54). This effect may contribute to action potential widening induced by tedisamil. In contrast, the selective inhibitors of the rapid component of the delayed rectifier K current (I[Kr]), dofetilide, and E-4031, as well as BRL-32872, that blocks I(Kr) and the L-type calcium current, did not significantly affect the amplitude of PKA-I(Cl), even at high concentrations (10-30 microM). These results demonstrate that compounds such as glibenclamide and tedisamil that are known to block the adenosine triphosphate (ATP)-sensitive K current also affect PKA-I(Cl). Furthermore it appears that blockade of PKA-I(Cl) is not a common feature for all class III antiarrhythmic agents.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cardiotonic Agents/pharmacology , Chlorides/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocardium/metabolism , Potassium Channels/drug effects , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , Acetylcholine/pharmacology , Animals , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclopropanes/pharmacology , Glyburide/pharmacology , Guinea Pigs , Heart Ventricles/drug effects , Hypoglycemic Agents/pharmacology , Ion Transport/drug effects , Isoproterenol/pharmacology , Male , Patch-Clamp Techniques , Phenethylamines/pharmacology , Piperidines/pharmacology , Propranolol/pharmacology , Pyridines/pharmacology , Sulfonamides/pharmacology
9.
Bioorg Med Chem ; 6(11): 1993-2011, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9881092

ABSTRACT

Class III antiarrhythmic agents have been shown to prevent reentrant arrhythmias but also to be responsible for initiating arrhythmias characterised by afterdepolarizations and triggered activities. By combining potassium and calcium channel antagonistic actions, as with BRL-32872 (1), it might be possible to reduce the incidence of proarrhythmias albeit retaining antiarrhythmic efficacy. In the present study we synthesised and tested for their electrophysiological activity in guinea pig papillary muscle a wide panel of analogues of BRL-32872. Some qualitative relationships between compound structure and the inhibitory effect on the rapidly activating component of the delayed rectifier potassium current and/or the L-type calcium current will be presented. New derivatives depicting bell-shaped dose-response curves on action potential duration may therefore represent novel agents for improved antiarrhythmic therapy.


Subject(s)
Anti-Arrhythmia Agents/chemical synthesis , Benzamides/chemical synthesis , Benzamides/pharmacology , Calcium Channel Blockers/chemical synthesis , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/pharmacology , Benzamides/chemistry , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Delayed Rectifier Potassium Channels , Electrophysiology , Guinea Pigs , Hydrogen Bonding , Indicators and Reagents , Molecular Structure , Papillary Muscles/drug effects , Papillary Muscles/physiology , Potassium Channels/physiology , Structure-Activity Relationship
10.
Am J Physiol ; 273(3 Pt 2): H1082-9, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9321792

ABSTRACT

Cyclic ADP-ribose (cADPR), an endogenous metabolite of beta-NAD+, activates Ca2+ release from endoplasmic reticulum in sea urchin eggs via the ryanodine receptor (RyR) pathway. A similar role has been proposed in cardiac sarcoplasmic reticulum (SR), although this remains controversial. We therefore investigated the ability of cADPR to induce Ca2+ release from canine cardiac SR microsomes using fluo 3 to monitor extravesicular Ca2+ concentration. We found that cADPR induced Ca2+ release in a concentration-dependent manner, whereas neither its precursor, NAD+, nor its metabolite, ADP-ribose, elicited a consistent effect. In addition, an additive effect on calcium release between cADPR and 9-Me-7-Br-eudistomin-D (MBED), an activator of RyR, was found as well as no cross-desensitization between cADPR and MBED. Specific blockers of the RyR did not abolish the cADPR-induced Ca2+ release. These results provide evidence for cADPR-induced Ca2+ release from dog cardiac SR via a novel mechanism which is independent of RyR activation.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Calcium Channels/physiology , Calcium/metabolism , Microsomes/metabolism , Muscle Proteins/physiology , Myocardium/metabolism , Sarcoplasmic Reticulum/metabolism , Adenosine Diphosphate Ribose/pharmacology , Aniline Compounds , Animals , Calcimycin/pharmacology , Calcium Channel Agonists/pharmacology , Calcium Channels/drug effects , Carbolines/pharmacology , Cyclic ADP-Ribose , Dogs , Drug Synergism , Fluorescent Dyes , Kinetics , Microsomes/drug effects , Models, Cardiovascular , Muscle Proteins/drug effects , NAD/pharmacology , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum/drug effects , Xanthenes
11.
J Pharmacol Exp Ther ; 276(2): 637-46, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8632331

ABSTRACT

In the search for novel antiarrhythmic agents, compounds with a diversity of electrophysiological actions have been suggested to result in treatments with potentially improved efficacy but with reduced proarrhythmic risk. To test this hypothesis, the antiarrhythmic versus proarrhythmic profile of BRL-32872, a novel agent with combined potassium and calcium channel blocking activity, was assessed in two different in vivo models of ventricular arrhythmia. Furthermore, the effects of potassium and calcium channel antagonists given either alone or in combination were assessed in the same models. Dogs with myocardial infarction received intravenously either vehicle, BRL-32872, the class III antiarrhythmic agent, E-4031, verapamil or a combination of E-4031 with verapamil (n = 8 per group). Ventricular tachyarrhythmias were induced by programmed electrical stimulation (PES). BRL-32872 (0.1, 0.3, 1.0 mg/kg) significantly increased QTc interval (from 387 +/- 10 to 462 +/- 19 msec.sec-1/2 at 1.0 mg/kg, P < .01). Ventricular effective refractory periods were increased in normal and infarcted areas (P < .01). Similar effects were observed with E-4031 (0.1, 0.3, 1.0 mg/kg). Verapamil (0.03, 0.1, 0.3 mg/kg) reduced heart rate, mean arterial pressure and, to a lesser extent, (+)dP/dtmax. Verapamil did not change QTc interval and ventricular effective refractory periods, but increased PR interval (P < .001). PES-induced tachyarrhythmias were not changed by vehicle or increasing doses of verapamil. E-4031 reduced the severity of arrhythmias from sustained ventricular tachycardia (VT) to nonsustained VT (7 dogs at 1.0 mg/kg, P = .013 vs. vehicle). BRL-32872 (0.1 and 0.3 mg/kg) suppressed the induction of sustained VT in six dogs (P = .02 vs. vehicle). In the presence of BRL-32872, 1.0 mg/kg, five dogs became noninducible to PES (P = .013 vs. vehicle). Combination of E-4031 (0.1 mg/kg) with verapamil provided a degree of protection that was similar to that observed with BRL-32872. In a second model, the proarrhythmic potential of BRL-32872 was assessed in anesthetized rabbits sensitized to develop torsades de pointes (TdP). BRL-32872 was compared with the class III antiarrhythmic agents, E-4031, dofetilide, clofilium and RP-58866. The pure class III antiarrhythmic agents induced TdP in 50 to 90% of the rabbits, and prolonged QT interval by 20 to 50%. BRL-32872 (10 micrograms/kg/min) increased QT interval by 35 +/- 5%, but did not promote TdP. In additional experiments, verapamil reduced the incidence of TdP induced by E-4031. These results show that BRL-32872 is a potent antiarrhythmic compound in a model of PES-induced arrhythmias and induces fewer proarrhythmic events than typical class III antiarrhythmic agents. The effects observed with BRL-32872 suggest that a compound with a combination of potassium (class III) and calcium (class IV) channel antagonistic properties might constitute a novel antiarrhythmic agent with reduced proarrhythmic risk.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzamides/pharmacology , Calcium Channel Blockers/pharmacology , Potassium Channel Blockers , Animals , Dogs , Electric Stimulation , Electroencephalography , Female , Hemodynamics/drug effects , Male , Myocardial Infarction/physiopathology , Rabbits , Torsades de Pointes/drug therapy
12.
Am J Physiol ; 269(5 Pt 2): H1625-33, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7503258

ABSTRACT

ATP-sensitive K+ (KATP) channels are thought only to open during conditions of metabolic impairment (e.g., myocardial ischemia). However, the regulation of KATP channel opening during ischemia remains poorly understood. We tested whether thiol (SH) group oxidation, which is known to occur during ischemia, may be involved in KATP channel regulation. Inside-out membrane patches were voltage clamped at a constant potential (O mV) in asymmetrical K+ solutions. The effects of compounds that specifically modify SH groups [p-chloromercuri-phenylsulfonic acid (pCMPS), 5-5'-dithio-bis(2-nitrobenzoic acid) [DTNB], and thimerosal] were tested. The membrane-impermeable compound, pCMPS (> or = 5 microM), caused a quick and irreversible inhibition of KATP channel activity. The reducing agent, dl-dithiothreitol (DTT) (3 mM) was able to reverse this inhibition. DTNB (500 microM) caused a rapid, but spontaneously reversible, block of KATP channel activity. After DTNB, no change was observed in single channel conductance. Oxidized glutathione (GSSG, 3 mM) did not block KATP channel activity. Thimerosal (100-500 microM) induced a DTT-reversible block of partially rundown KATP channels, or channels that underwent complete rundown; these channels were reactivated with trypsin (1 mg/ml). Thimerosal did not block KATP channels that had a high degree of activity. However, the ATP sensitivity was decreased; the concentration of ATP needed to half-maximally inhibit the channel (Ki) was increased from 47 +/- 12 to 221 +/- 35 microM (n = 6, P < 0.05). This was not due to a spontaneous change with time.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
4-Chloromercuribenzenesulfonate/pharmacology , Adenosine Triphosphate/physiology , Dithionitrobenzoic Acid/pharmacology , Myocardium/metabolism , Potassium Channels/drug effects , Sulfhydryl Reagents/pharmacology , Thimerosal/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Dithiothreitol/pharmacology , Guinea Pigs , Heart Ventricles , Myocardium/cytology , Potassium Channels/metabolism
13.
J Pharmacol Exp Ther ; 273(3): 1264-72, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7791099

ABSTRACT

The effects of N-(3,4-dimethoxyphenyl)-N-[3[[2-(3,4-dimethoxyphenyl) ethyl] propyl]-4-nitrobenzamide hydrochloride (BRL-32872), a novel antiarrhythmic agent, were studied in guinea pig cardiac preparations using standard microelectrode and patch-clamp techniques. In papillary muscle, BRL-32872 did not change resting membrane potential and maximum rate of depolarization but prolonged action potential duration (APD) by 24% +/- 2% at 1.0 microM. When the concentration was increased to 3.0 and 10.0 microM, the effect on APD was not further enhanced, and a bell-shaped dose-response curve resulted. Patch-clamp experiments in isolated myocytes showed that BRL-32872 inhibited the rapidly activating component of the delayed rectifier potassium current (EC50 = 0.028 microM) and the L-type calcium current (EC50 = 2.8 microM) but had a limited effect on the inward rectifier potassium current. In papillary muscles stimulated at 300, 500, 1000 and 2000 msec, the effect of BRL-32872 in prolonging APD did not vary (P = .717). By contrast, N-(4-(1-[2-(6-methyl-2-pyridyl)ethyl]-4-piperidyl)- carbonyl]phenyl)methanesulfonamide dihydrochloride dihydrate (E-4031), a pure class III antiarrhythmic agent, increased APD more at slower than at faster stimulation rates (P = .001), which illustrated the reverse frequency-dependence of this agent. Among the 35 experiments performed with BRL-32872, only one fiber showed early afterdepolarizations (EADs), and these, which occurred at 1.0 microM, were suppressed at higher concentration (3.0 microM). Moreover, EADs induced by E-4031 were suppressed by BRL-32872 (3.0 microM).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzamides/pharmacology , Calcium Channel Blockers/pharmacology , Heart/drug effects , Potassium Channel Blockers , Action Potentials/drug effects , Animals , Guinea Pigs , Heart/physiology , In Vitro Techniques , Male , Microelectrodes
14.
Pflugers Arch ; 418(3): 261-5, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1649991

ABSTRACT

To examine the effects of the activation of adenosine 5'-triphosphate (ATP)-sensitive K channels in a skeletal muscle we have applied the ATP-sensitive K channel opener SR44866 whilst recording single ion channels, voltage-clamped membrane currents, evoked action potentials and tension in sartorius muscles of the frog. In excised inside-out membrane patches SR44866 opened channels which could be inhibited by internal ATP and glibenclamide. In voltage-clamped individual muscle fibres SR44866 evoked a glibenclamide-sensitive membrane current which reversed at -70 mV. The effect of SR44866 was dose dependent with an effective concentration for 50% maximal effect (EC50) of 67 microM and a slope factor of 2. SR44866 dose dependently reduced the duration of the spike after-potential, spike overshoot, Vmax, tetrodotoxin-sensitive voltage-gated inward membrane currents and muscle twitch tension. From this evidence it can be concluded that the opening of ATP-sensitive K channels may be associated with the inhibition of contraction of skeletal muscle.


Subject(s)
Adenosine Triphosphate/pharmacology , Benzopyrans/pharmacology , Dihydropyridines/pharmacology , Muscles/physiology , Potassium Channels/physiology , Action Potentials/drug effects , Animals , Benzopyrans/administration & dosage , Cell Membrane/physiology , Dihydropyridines/administration & dosage , Dose-Response Relationship, Drug , Glyburide/pharmacology , Membrane Potentials/drug effects , Muscle Contraction/drug effects , Muscles/drug effects , Potassium Channels/drug effects , Rana esculenta , Sodium Channels/drug effects , Sodium Channels/physiology , Tetrodotoxin/pharmacology
15.
FEBS Lett ; 279(1): 95-7, 1991 Feb 11.
Article in English | MEDLINE | ID: mdl-1995351

ABSTRACT

We show that ATP-sensitive K+ channels of excised inside-out membrane patches of rat ventricular myocytes show considerable variation in their sensitivity to ATP. In 102 different membrane patches IC50 values ranged from 9 to 580 microM ATP and Hill coefficients from 1% to 6.41% of patches showed openings of ATP-sensitive K+ channels in the presence of 1 mM ATP. These results considerably widen the range of internal ATP concentrations over which one might expect activation of the ATP-sensitive K+ current in cardiac myocytes.


Subject(s)
Adenosine Triphosphate/metabolism , Muscle, Smooth/metabolism , Potassium Channels/metabolism , Animals , Membrane Potentials , Rats
16.
Biochim Biophys Acta ; 1029(1): 167-72, 1990 Nov 02.
Article in English | MEDLINE | ID: mdl-2223807

ABSTRACT

It is difficult to associate the ATP-sensitive potassium (K-ATP) channel of cardiac muscle with hypoxia/ischemia induced action potential shortening because this occurs before intracellular ATP falls to levels associated in vitro with channel opening. This leaves the cardiac K-ATP channel without any obvious physiological function. We have quantitatively examined the relationship between action potential duration and K-ATP channel activity in enzymatically isolated ventricular myocytes of the guinea-pig. In whole-cell voltage-clamp recording experiments when the K-ATP channel opener SR 44866 (2-10 microM) stimulated an outward membrane current greater than 50 pA at 0 mV membrane potential (the equivalent of 30 open K-ATP channels or 1% of the cell K-ATP channel population) action potential duration was reduced by more than 50%. In the majority of cell-attached membrane patch recordings metabolic inhibition stimulated K-ATP channel open probability of 1-2% which continued for long periods (7-25 min) before cell contracture and coincident major K-ATP channel activation (open probability 65%). Our quantitative analysis thus shows that physiologically relevant activity of K-ATP channels in cardiac muscle is confined to a very small percentage of the possible cell K-ATP current and thus intracellular ATP would not have to fall very far before the opening of K-ATP channels would influence cardiac excitability.


Subject(s)
Adenosine Triphosphate/metabolism , Coronary Disease/metabolism , Myocardium/metabolism , Potassium Channels/metabolism , Animals , Benzopyrans/pharmacology , Dihydropyridines/pharmacology , Guinea Pigs , Heart/drug effects , In Vitro Techniques , Membrane Potentials , Myocardium/cytology , Potassium/metabolism
17.
Toxicon ; 28(8): 925-37, 1990.
Article in English | MEDLINE | ID: mdl-1964254

ABSTRACT

Maitotoxin (MTX) irreversibly suppressed the voltage-dependent calcium current after a variable delay, an effect which was preceded, in 61% of the cells, by a transient increase in calcium current partly attributable to a shift (4-7 mV) of the activation curve towards negative potentials. MTX also induced the development of a voltage-independent background inward current which did not occur in the absence of external calcium and was reduced by removal of external sodium, by calcium channel blockers and by high concentrations of quinidine. MTX-induced single channel activity consisted of long lasting bursts of inward current. Channel activity was voltage-independent, with a unitary conductance of 14 pS and an extrapolated reversal potential of +16 mV. Single-channel current amplitude was not detectably reduced in the absence of external calcium but strongly reduced in the absence of external sodium, in the presence of 2 mM nickel or when external sodium was replaced by 96 mM calcium or 50 mM barium. The channel activity was also inhibited by quinidine. It is concluded that MTX alters, then suppresses the voltage-activated calcium current and induces the development of a voltage-independent inward current, part of which results from the opening of nickel-sensitive cation channels, mostly permeable to sodium ions.


Subject(s)
Calcium Channels/drug effects , Marine Toxins/pharmacology , Myocardium/metabolism , Oxocins , Animals , In Vitro Techniques , Membrane Potentials/drug effects , Myocardium/cytology , Nickel/pharmacology , Quinidine/pharmacology , Rats , Sodium Channels/drug effects
18.
Biochim Biophys Acta ; 984(1): 1-5, 1989 Aug 21.
Article in English | MEDLINE | ID: mdl-2504288

ABSTRACT

Drugs which influence the electrical activity of insulin-secreting B cells of mammalian islets of Langerhans by closing (tolbutamide and glibenclamide) or opening (diazoxide) ATP-sensitive potassium channels were applied to the ventricular muscle of the rat. Action potentials were recorded from ventricular epicardium of perfused intact rat hearts. Tolbutamide (0.5-2.0 mM), glibenclamide (0.01-0.1 mM) and diazoxide (0.5 mM) each evoked a dose-dependent increase (7-33%) in the duration of the ventricular action potential measured at 50% of repolarization. These drugs were without effect upon the resting membrane potential or the peak of the action potential. Single-channel recordings of ATP-sensitive K+ channels were obtained from excised membrane patches of enzymatically isolated rat ventricular myocytes. Tolbutamide and diazoxide inhibited openings of ATP-sensitive K+ channels. Diazoxide inhibited ATP-sensitive K+ channel openings in the presence of ATP. Diazoxide did not evoke opening of ATP-sensitive K+ channels. It is concluded that these drugs could act to increase the duration of the cardiac action potential by inhibiting openings of ATP-sensitive K+ channels.


Subject(s)
Diazoxide/pharmacology , Glyburide/pharmacology , Heart/drug effects , Tolbutamide/pharmacology , Action Potentials/drug effects , Adenosine Triphosphate/pharmacology , Animals , Heart/physiology , In Vitro Techniques , Male , Potassium Channels/drug effects , Rats , Rats, Inbred Strains
19.
Am J Physiol ; 252(1 Pt 2): H142-8, 1987 Jan.
Article in English | MEDLINE | ID: mdl-3028160

ABSTRACT

It has been suggested in a previous article [Escande et al., Am. J. Physiol. 249 (Heart Circ. Physiol. 18): H843-H850, 1985] that transient outward currents may participate in the initial repolarization of human atrial fibers. The present study substantiates the existence of such currents in human myocardium. Membrane currents were recorded in enzymatically dissociated cells using the whole cell patch-clamp technique. Two kinds of transient outward currents were observed: 1) a long-lasting outward current, (ilo), which was suppressed by 4-aminopyridine. The time to peak of ilo was 18.0 +/- 0.7 ms, and its inactivation time constant was 35.7 +/- 2.1 ms at room temperature (test pulses, +20 mV; holding potential, -40 mV); 2) a brief outward current (ibo), which persisted with 3 mM 4-aminopyridine and exhibited a shorter time to peak (5.5 +/- 0.2 ms) and a faster decay (time constant, 9.1 +/- 1.8 ms). ilo was inhibited by Ba but was insensitive to the calcium blocker Co. Co blocked both the slow inward current (isi) and ibo. It is concluded that two different transient outward currents control the repolarization in human atrial cells.


Subject(s)
Heart/physiology , 4-Aminopyridine , Adult , Aminopyridines/pharmacology , Atrial Function , Electric Conductivity , Heart Atria/cytology , Heart Atria/drug effects , Humans , In Vitro Techniques , Membrane Potentials/drug effects
20.
J Mol Cell Cardiol ; 18(5): 547-51, 1986 May.
Article in English | MEDLINE | ID: mdl-3723601

ABSTRACT

The slow inward current isi plays a prominent role in impulse formation and slow conduction in the human heart. For this reason, it may be involved in a great variety of arrhythmias that occur clinically, particularly at the supraventricular level. isi also appears to be the major target of numerous cardioactive drugs daily administered to many patients. Difficulties encountered in voltage clamping human tissues, inevitably dissected from restricted zones of myocardium with inadequate geometry, may explain why direct measurements of this current have never been performed on multicellular human preparations. The recent development of enzymatic techniques allowing the isolation of calcium-tolerant myocytes has provided a solution to these problems. To our knowledge only one abstract has presented until now records of isi in human ventricular myocytes. We describe here the result of experiments carried out to investigate the slow inward current in single cells isolated from adult human atria. The present work provides the first quantitative description of this current in human myocytes.


Subject(s)
Heart/physiology , Atrial Function , Cells, Cultured , Culture Media , Electric Conductivity , Humans , In Vitro Techniques , Kinetics , Membrane Potentials
SELECTION OF CITATIONS
SEARCH DETAIL
...