Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters










Publication year range
1.
J Cardiovasc Pharmacol ; 46(6): 746-53, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16306797

ABSTRACT

The effects of the sodium-hydrogen (Na/H) exchange inhibitor cariporide (HOE642), on insulin sensitivity and vascular function were studied in the JCR:LA-cp rat and the db/db mouse. In the insulin-resistant rat, cariporide reduced fasting insulin levels (42%, P < 0.02) and insulin response in a meal tolerance test (50%, P < 0.01), indicating increased insulin sensitivity. The ACE inhibitor, ramipril, used as a reference agent, reduced the insulin response to the meal, but not fasting levels. The EC50 for acetylcholine-mediated relaxation of phenylephrine-precontracted aortic rings was significantly lower in cariporide-treated rats (P < 0.002), but not in ramipril-treated rats. Flow response of the coronary circulation to bradykinin was significantly greater in both cariporide- and ramipril-treated rats, (3-fold decrease in the EC50, P < 0.05). Cariporide-treated hearts were smaller, slower beating, with greater developed LVP. In the obese db/db mouse, chronic treatment with cariporide obviated vascular hypercontractility and improved endothelial function. Thus, cariporide had beneficial effects on the abnormal insulin metabolism and associated vascular dysfunction in the JCR:LA-cp insulin-resistant rat, which develops advanced cardiovascular disease and ischemic myocardial lesions. It also improved vascular function in a similar mouse model of insulin resistance. These effects were markedly greater than those of ramipril.


Subject(s)
Aorta/drug effects , Guanidines/pharmacology , Heart/drug effects , Insulin Resistance , Obesity/physiopathology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology , Acetylcholine/pharmacology , Animals , Aorta/physiology , Body Weight/drug effects , Coronary Circulation/drug effects , Eating/drug effects , Heart/physiology , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Nitroprusside/pharmacology , Ramipril/pharmacology , Rats , Receptors, Cell Surface/genetics , Receptors, Leptin , Vasoconstriction/drug effects
2.
Diabetologia ; 48(8): 1645-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16010524

ABSTRACT

AIMS/HYPOTHESIS: Renal accumulation of AGEs may contribute to the progression of diabetic nephropathy. We evaluated the effect of ramipril (a pure ACE inhibitor) and AVE7688 (a dual inhibitor of ACE and neutral endopeptidase) on renal accumulation of the advanced glycation end-product (AGE) 3-deoxyglucosone-imidazolone, carboxymethyllysine (CML) and pentosidine, and on clearance of CML in type 2 diabetes. METHODS: Male Zucker diabetic fatty rats (ZDF, Gmi-fa/fa) rats were treated from age 10 to 37 weeks with ramipril (1 mg.kg(-1).day(-1)), AVE7688 (45 mg.kg(-1).day(-1)) or without drug. Ramipril and AVE7688 reduced albuminuria by 30 and 90%, respectively. RESULTS: ZDF rats showed increased renal accumulation of the AGE subtypes 3-deoxyglucosone-imidazolone, pentosidine and CML by about 40, 55 and 55%, respectively compared with heterozygous, non-diabetic control animals at the age of 37 weeks. AVE7688 but not ramipril attenuated the renal accumulation of 3-deoxyglucosone-imidazolone, pentosidine and CML and improved CML clearance in ZDF rats. During glycation reactions in vitro, AVE7688 also demonstrated potent chelating activity and inhibited metal-catalysed formation of pentosidine and CML. CONCLUSIONS/INTERPRETATION: Improved AGE clearance and direct inhibition of AGE formation by chelation may contribute to reduced accumulation of renal AGEs and to the nephroprotective effects of vasopeptidase inhibition in type 2 diabetes.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Glycation End Products, Advanced/metabolism , Kidney/metabolism , Protease Inhibitors/pharmacology , Animals , Ascorbic Acid/metabolism , Blood Glucose/metabolism , Chromatography, High Pressure Liquid , Creatine/metabolism , Deoxyglucose/analogs & derivatives , Deoxyglucose/metabolism , Glycated Hemoglobin/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Kidney/drug effects , Lysine/analogs & derivatives , Lysine/metabolism , Male , Oxidation-Reduction , Ramipril/pharmacology , Rats , Rats, Zucker , Spectrometry, Fluorescence
3.
Naunyn Schmiedebergs Arch Pharmacol ; 370(3): 183-92, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15340774

ABSTRACT

Previous studies in pigs and goats have demonstrated that AVE0118 prolongs atrial refractoriness without any effect on the QT-interval. The purpose of the present study was to investigate the effect of the compound on various cardiac ion channels. AVE0118 blocked the pig Kv1.5 and the human Kv1.5 expressed in Xenopus oocytes with IC(50) values of 5.4+/-0.7 microM and 6.2+/-0.4 microM respectively. In Chinese hamster ovary (CHO) cells, AVE0118 decreased the steady-state hKv1.5 current with an IC(50) of 1.1+/-0.2 microM. The hKv4.3/KChIP2.2 current in CHO cells was blocked by AVE0118 by accelerating the apparent time-constant of inactivation ( tau(inact)), and the integral current was inhibited with an IC(50) of 3.4+/-0.5 microM. At 10 microM AVE0118 tau(inact) decreased from 9.3+/-0.6 ms ( n=8, control) to 3.0+/-0.3 ms ( n=8). The K(ACh) current was investigated in isolated pig atrial myocytes by application of 10 microM carbachol. At a clamp potential of -100 mV the I(KACh) was half-maximally blocked by 4.5+/-1.6 microM AVE0118. In the absence of carbachol, AVE0118 had no effect on the inward current recorded at -100 mV. Effects on the I(Kr) current were investigated on HERG channels expressed in CHO cells. AVE0118 blocked this current half-maximally at approximately 10 microM. Comparable results were obtained in isolated guinea pig ventricular myocytes, where half-maximal inhibition of the I(Kr) tail current occurred at a similar concentration of AVE0118. Other ionic currents, like the I(Ks), I(KATP) (recorded in guinea pig ventricular myocytes), and L-type Ca(2+) (recorded in pig atrial myocytes) were blocked by 10 microM AVE0118 by 10+/-3% ( n=6), 28+/-7% ( n=4), and 22+/-13% ( n=5) respectively. In summary, AVE0118 preferentially inhibits the atrial K(+) channels I(Kur), I(to) and I(KACH). This profile may explain the selective prolongation of atrial refractoriness described previously in pigs and goats.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Carbachol/pharmacology , Cardiotonic Agents/pharmacology , Ion Channels/drug effects , Myocytes, Cardiac/drug effects , Animals , CHO Cells , Calcium-Binding Proteins/antagonists & inhibitors , Cells, Cultured , Cricetinae , Cricetulus , Electrophysiology , Humans , Kv Channel-Interacting Proteins , Kv1.5 Potassium Channel , Molecular Biology , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Shal Potassium Channels , Swine , Xenopus
4.
Naunyn Schmiedebergs Arch Pharmacol ; 369(4): 374-81, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15024553

ABSTRACT

During heart ischemia, ATP-sensitive potassium channels in the sarcolemmal membrane (sarcK(ATP)) open and cause shortening of the action potential duration. This creates heterogeneity of repolarization, being responsible for the development of re-entry arrhythmias and sudden cardiac death. Therefore, the aim is to develop selective blockers of the cardiac sarcK(ATP) channel. In the present study we established an in vitro model and classified 5 K(ATP) channel inhibitors with respect to their potency and selectivity between cardiomyocytes and the coronary vasculature and compared the results with inhibition of Kir6.2/SUR2A channels expressed in HEK293 cells, recorded with the Rb(+)-efflux methods. We used Langendorff-perfused guinea pig hearts, where low-flow ischemia plus hypoxia was performed by reducing the coronary flow (CF) to 1.2 ml/min and by gassing the perfusion solution with N(2) instead of O(2). Throughout the experiment, the monophasic action potential duration at 90% repolarization (MAPD(90)) was recorded. In separate experiments, high-flow hypoxia was produced by oxygen reduction in the perfusate from 95% to 20%, which caused an increase in the coronary flow. Under normoxic conditions, the substances glibenclamide, repaglinide, meglitinide, HMR 1402 and HMR 1098 (1 microM each) reduced the CF by 34%, 38%, 19%, 12% and 5%, respectively. The hypoxia-induced increase in CF was inhibited by the compounds half-maximally at 25 nM, approximately 200 nM, 600 nM, approximately 9 microM and >100 microM, respectively. In control experiments after 5 min low-flow ischemia plus hypoxia, the MAPD(90) shortened from 121+/-2 to 99+/-2 ms ( n=29). This shortening was half-maximally inhibited by the substances at concentrations of 95 nM, 74 nM, 400 nM, 110 nM and 550 nM, respectively. In HEK293 cells the Rb(+)-efflux through KIR6.2/SUR2A channels was inhibited by the compounds with IC(50) values of 21 nM, 67 nM, 205 nM, 60 nM and 181 nM, respectively. In summary, the present data demonstrate that the sulfonylurea glibenclamide, and the carbamoylbenzoic acid derivatives repaglinide and meglitinide are unselective blockers of K(ATP) channels in cardiac cells and in the cardiac vascular system, whereas the sulfonylthioureas HMR 1402, and especially HMR 1098 selectively blocked the cardiac sarcK(ATP) channel. Blockade of Kir6.2/SUR2A channels in HEK293 cells occurred with comparable efficacy as in the cardiac tissue, indicating that the expression system is suited for screening for novel inhibitors.


Subject(s)
Adenosine Triphosphate/physiology , Heart/drug effects , Potassium Channel Blockers/adverse effects , Potassium Channels/physiology , ATP-Binding Cassette Transporters/physiology , Action Potentials/drug effects , Animals , Cell Line , Coronary Circulation/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , Heart/physiology , Humans , Hypoxia/complications , Hypoxia/physiopathology , In Vitro Techniques , Male , Myocardial Ischemia/complications , Myocardial Ischemia/physiopathology , Potassium Channel Blockers/administration & dosage , Potassium Channels, Inwardly Rectifying/physiology , Receptors, Drug/physiology , Sulfonylurea Receptors
5.
Diabetologia ; 47(1): 98-103, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14618238

ABSTRACT

AIM/HYPOTHESIS: Pharmacological inhibition of the renin angiotensin system has proven clinical efficacy in nephropathies of various origins, including diabetic nephropathy. We tested the effects of the dual inhibition of both angiotensin converting enzyme and neutral endopeptidase by the vasopeptidase inhibitor AVE7688 in an animal model of Type 2 diabetic nephropathy. METHODS: We treated 56 obese Zucker diabetic fatty (ZDF, Gmi-fa/fa) rats aged 34-weeks with either placebo ( n=9) or the vasopeptidase inhibitor AVE7688 in four different doses (each n=9; 3, 10, 30, or 60 mg/kg/d in chow). We used 11 heterozygous (+/fa) rats which received placebo and served as non-diabetic, lean controls. Urinary albumin/creatinine ratio was assessed as a marker of nephropathy at baseline (age 34-weeks) and after 10 weeks of chronic treatment. RESULTS: All obese animals had established diabetes mellitus that was not influenced by AVE7688 (HbA(1c) >12%, stable in all dose groups). There was massive albuminuria in the homozygous ZDF rats (albumine/creatinine ratio >20 mg/mg vs minimal albuminuria in lean controls) that was decreased by AVE7688 in a dose dependent manner (Placebo 2.0+/-4.4 vs 11.9+/-1.8, 13.4+/-0.7, 13.6+/-2.8, and 19.8+/-2.8 mg/mg in the 3, 10, 30, and 60 mg/kg/d groups, respectively; all treatment groups p<0.05 vs Placebo). CONCLUSION/INTERPRETATION: AVE7688 ameliorates proteinuria in Zucker diabetic fatty rats with established diabetes mellitus. Vasopeptidase inhibition represents an effective novel therapeutic principle for intervention in Type 2 diabetic nephropathy independent of metabolic control.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/drug therapy , Heterocyclic Compounds, 3-Ring/therapeutic use , Albuminuria/prevention & control , Animals , Body Weight , Diabetes Mellitus/drug therapy , Drinking Behavior , Energy Intake , Glycated Hemoglobin/metabolism , Male , Obesity , Prodrugs/therapeutic use , Protease Inhibitors/therapeutic use , Rats , Rats, Zucker
6.
Br J Pharmacol ; 134(8): 1647-54, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739240

ABSTRACT

1. Recently we and others have demonstrated a stereoselective inhibition of slowly activating human I(Ks) (KCNQ1/MinK) and homomeric KCNQ1 potassium channels by the enantiomers of the chromanol 293B. Here, we further characterized the mechanism of the 293B block and studied the influence of the 293B enantiomers on the gating kinetics of both channels after their heterologous expression in Xenopus oocytes. 2. Kinetic analysis of currents partially blocked with 10 microM of each 293B enantiomer revealed that only 3R,4S-293B but not 3S,4R-293B exhibited a time-dependent block of I(Ks) and KCNQ1 currents, indicating preferential open channel block activity. 3. Inhibition of both KCNQ1 and I(Ks) channels by 3R,4S-293B but not by 3S,4R-293B increased during a 2 Hz train of stimuli. 4. At high extracellular potassium concentrations the inhibition of KCNQ1 by 3R,4S-293B and 3S,4R-293B was unaffected. Drug inhibition of KCNQ1 and I(Ks) by both enantiomers also did not display a significant voltage-dependence, indicating that 293B does not strongly interact with permeant ions in the pore. 5. The inhibitory properties of 3R,4S-293B on I(Ks)-channels but not those of 3S,4R-293B fulfill the theoretical requirements for a novel class III antiarrhythmic drug, i.e. positive use-dependency. This enantiomer therefore represents a valuable pharmacological tool to evaluate the therapeutic efficiency of I(Ks)blockade.


Subject(s)
Chromans/pharmacology , Potassium Channel Blockers , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated , Sulfonamides/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Binding Sites , Chromans/metabolism , Electrophysiology , Female , Humans , Inhibitory Concentration 50 , Ion Channel Gating , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Oocytes/metabolism , Patch-Clamp Techniques , Perfusion , Potassium Channel Blockers/metabolism , Potassium Channels/metabolism , Stereoisomerism , Sulfonamides/metabolism , Thermodynamics , Xenopus
7.
Naunyn Schmiedebergs Arch Pharmacol ; 364(5): 472-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11692231

ABSTRACT

The inhibitory effects of the novel Kv1.5 channel blocker, S9947 (2'-(benzyloxycarbonylaminomethyl)biphenyl-2-carboxylic acid 2-(2-pyridyl)ethylamide), on cloned human Kv1.5 (hKv1.5), expressed in both Xenopus oocytes and Chinese hamster ovary (CHO) cells, and on native cardiac ultrarapid delayed rectifier potassium currents (IKur) in rat (ventricle myocytes) and human (atrial myocytes) were investigated. The influence of S9947 on the action potential was examined in rat ventricular myocytes. Using the two-electrode voltage-clamp technique in Xenopus oocytes and the patch-clamp technique (whole cell configuration) in CHO cells, hKv1.5 was inhibited by S9947 with IC50 values of 0.65 microM and 0.42 microM, respectively. In addition, inhibition of human Kv4.3 (hKv4.3) and HERG by 10 microM S9947 was low (approximately 20%) and absent, respectively. Using the patch-clamp technique in the whole cell configuration, IKur currents in rat ventricular (rIKur) cardiomyocytes and human atrial (hIKur) cardiomyocytes were inhibited by S9947 with IC50 values of 0.96 microM and 0.07 microM, respectively. In contrast, rat cardiac inward rectifier current (rIK1) and rat (rIto) and human (hIto) cardiac transient outward currents were only inhibited by approximately 20% with 10 microM S9947. In rat cardiomyocytes, using the patch-clamp technique, action potential duration was increased by S9947 in a concentration-dependent (0.3-10 microM) and rate-independent manner. The data show that S9947 suppresses both cloned (Kv1.5) and native (IKur) cardiac potassium currents. Furthermore, S9947 prolongs rat action potential in a rate-independent manner.


Subject(s)
Action Potentials/drug effects , Biphenyl Compounds/pharmacology , Heart/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels , Pyridines/pharmacology , Animals , CHO Cells , Cricetinae , Electric Stimulation , Female , Heart/physiology , Humans , Kv1.5 Potassium Channel , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Xenopus
8.
J Med Chem ; 44(23): 3831-7, 2001 Nov 08.
Article in English | MEDLINE | ID: mdl-11689069

ABSTRACT

Since the discovery of the I(Ks)-potassium channel as the slowly activating component of the delayed rectifier current (I(k)) in cardiac tissue, the search for blockers of this current has been intense. During the screening of K(ATP)-channel openers of the chromanol type we found that chromanol 293B was able to block I(Ks). Chromanol 293B is a sulfonamide analogue of the K(ATP)-channel openers but had no activity on this target. Experiments were initiated to improve the activity and properties based on this lead compound. As a screening model we used Xenopus oocytes injected with human minK (KCNE1). Variations of the aromatic substituent and the sulfonamide group were prepared, and their activity was evaluated. We found that the greatest influence on activity was found in the aromatic substituents. The most active compounds were alkoxy substituted. We chose HMR1556 ((3R, 4S)-(+)-N-[-3-hydroxy-2,2-dimethyl-6-(4,4,4-trifluorobutoxy)chroman-4-yl]-N-methyl-ethanesulfonamide) 10a for development as an antiarrhythmic drug. The absolute configuration, resulting from an X-ray single-crystal structure analysis, was determined.


Subject(s)
Chromans/chemical synthesis , Potassium Channel Blockers , Potassium Channel Blockers/chemical synthesis , Potassium Channels, Voltage-Gated , Sulfonamides/chemical synthesis , Animals , Chromans/chemistry , Chromans/pharmacology , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Xenopus laevis
9.
Br J Pharmacol ; 134(7): 1467-79, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11724753

ABSTRACT

1. We identified the ethacrynic-acid derivative DCPIB as a potent inhibitor of I(Cl,swell), which blocks native I(Cl,swell) of calf bovine pulmonary artery endothelial (CPAE) cells with an IC(50) of 4.1 microM. Similarly, 10 microM DCPIB almost completely inhibited the swelling-induced chloride conductance in Xenopus oocytes and in guinea-pig atrial cardiomyocytes. Block of I(Cl,swell) by DCPIB was fully reversible and voltage independent. 2. DCPIB (10 microM) showed selectivity for I(Cl,swell) and had no significant inhibitory effects on I(Cl,Ca) in CPAE cells, on chloride currents elicited by several members of the CLC-chloride channel family or on the human cystic fibrosis transmembrane conductance regulator (hCFTR) after heterologous expression in Xenopus oocytes. DCPIB (10 microM) also showed no significant inhibition of several native anion and cation currents of guinea pig heart like I(Cl,PKA), I(Kr), I(Ks), I(K1), I(Na) and I(Ca). 3. In all atrial cardiomyocytes (n=7), osmotic swelling produced an increase in chloride current and a strong shortening of the action potential duration (APD). Both swelling-induced chloride conductance and AP shortening were inhibited by treatment of swollen cells with DCPIB (10 microM). In agreement with the selectivity for I(Cl,swell), DCPIB did not affect atrial APD under isoosmotic conditions. 4. Preincubation of atrial cardiomyocytes with DCPIB (10 microM) completely prevented both the swelling-induced chloride currents and the AP shortening but not the hypotonic cell swelling. 5. We conclude that swelling-induced AP shortening in isolated atrial cells is mainly caused by activation of I(Cl,swell). DCPIB therefore is a valuable pharmacological tool to study the role of I(Cl,swell) in cardiac excitability under pathophysiological conditions leading to cell swelling.


Subject(s)
Action Potentials/drug effects , Chloride Channels/antagonists & inhibitors , Cyclopentanes/pharmacology , Heart Atria/drug effects , Indans/pharmacology , Potassium Channels, Voltage-Gated , Animals , Atrial Function , Cell Size/physiology , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Guinea Pigs , Heart Atria/cytology , Membrane Potentials/drug effects , Oocytes , Potassium Channels/drug effects , Potassium Channels/genetics , Potassium Channels/physiology , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Sensitivity and Specificity , Shal Potassium Channels , Time Factors , Xenopus
10.
Cardiovasc Res ; 52(2): 255-64, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11684073

ABSTRACT

OBJECTIVE: The Ca(2+) independent transient outward K(+) current (I(to1)) in the heart is responsible for the initial phase of repolarization. The hKv4.3 K(+) channel alpha-subunit contributes to the I(to1) current in many regions of the human heart. Consistently, downregulation of hKv4.3 transcripts in heart failure and atrial fibrillation is linked to reduction in I(to1) conductance. The recently cloned KChIP family of calcium sensors has been shown to modulate A-type potassium channels of the Kv4 K(+) channel subfamily. METHODS AND RESULTS: We describe the cloning and tissue distribution of hKChIP2, as well as its functional interaction with hKv4.3 after expression in Xenopus oocytes. Furthermore, we isolated a short splice variant of the hKChIP2 gene (hKCNIP2), which represents the major hKChIP2 transcript. Northern blot analyses revealed that hKChIP2 is expressed in the human heart and occurs in the adult atria and ventricles but not in the fetal heart. Upon coexpression with hKv4.3 both hKChIP2 isoforms increased the current amplitude, slowed the inactivation and increased the recovery from inactivation of hKv4.3 currents. For the first time we analyzed the influence of a KChIP protein on the voltage of half-maximal inactivation of Kv4 channels. We demonstrate that the hKChIP2 isoforms shifted the half-maximal inactivation to more positive potentials, but to a different extent. By elucidating the genomic structure, we provide important information for future analysis of the hKCNIP2 gene in candidate disorders. In the course of this work we mapped the hKCNIP2 gene to chromosome 10q24. CONCLUSIONS: Heteromeric hKv4.3/hKChIP2 currents more closely resemble native epicardial I(to1), suggesting that hKChIP2 is a true beta-subunit of human cardiac I(to1). As a result hKChIP2 might play a role in cardiac diseases, where a contribution of I(to1) has been shown.


Subject(s)
Alternative Splicing , Calcium-Binding Proteins/genetics , Chromosomes, Human, Pair 10 , Myocardium/chemistry , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Animals , Blotting, Northern/methods , Chromosome Mapping , Cloning, Molecular , Female , Gene Expression , Gene Transfer Techniques , Humans , Introns , Kv Channel-Interacting Proteins , Myocardium/metabolism , Oocytes/metabolism , Patch-Clamp Techniques , Polymerase Chain Reaction/methods , Potassium Channels/analysis , Protein Isoforms/analysis , Protein Isoforms/genetics , Sequence Analysis, DNA , Shal Potassium Channels , Sodium-Potassium-Exchanging ATPase , Xenopus laevis
11.
Pflugers Arch ; 442(6): 891-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11680622

ABSTRACT

The delayed rectifier potassium current I(Ks) is important for repolarization of the cardiac action potential. In heart I(Ks) is a heteromeric channel composed of KCNQ1 (KvLQT1) and minK (KCNE1, IsK). Here we show that the KCNQ1/minK interaction is influenced by the expression system. Co-expression of KCNQ1 and minK in Xenopus oocytes resulted in potassium currents comparable to endogenous guinea pig cardiac I(Ks) in terms of temperature dependency and activation kinetics. In contrast, heterologous expression of I(Ks) in CHO cells revealed currents with a markedly different biophysical behavior. The sensitivity to the extracellular potassium concentration, temperature dependency and kinetics differ qualitatively. Potentially there is an endogenous component that affects I(Ks) which does not appear in all expression systems.


Subject(s)
Gene Expression , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium Channels/physiology , Action Potentials , Animals , CHO Cells , Cricetinae , Electric Conductivity , Female , Guinea Pigs , Heart/physiology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Kinetics , Oocytes , Potassium/pharmacology , Recombinant Proteins , Temperature , Transfection , Xenopus
12.
Naunyn Schmiedebergs Arch Pharmacol ; 364(1): 33-41, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11485036

ABSTRACT

It has been argued that activation of KATP channels in the sarcolemmal membrane of heart muscle cells during ischemia provides an endogenous cardioprotective mechanism. In order to test whether the novel cardioselective KATP channel blocker HMR 1098 affects cardiac function during ischemia, experiments were performed in rat hearts during ischemia and reperfusion. Isolated perfused working rat hearts were subjected to 30 min of low-flow ischemia in which the coronary flow was reduced to 10% of its control value, followed by 30-min reperfusion. In the first set of experiments the hearts were electrically paced at 5 Hz throughout the entire protocol. At the end of the 30-min ischemic period the aortic flow had fallen to 44 +/- 2% (n=8) of its nonischemic value in vehicle-treated hearts, whereas in the presence of 0.3 micromol/l and 3 micromol/l HMR 1098 it had fallen to 29 +/- 7% (n=5, not significant) and 8 +/- 2% (n=12, P<0.05), respectively. Glibenclamide (3 micromol/l) reduced the aortic flow to 9.5 +/- 7% (n=4, P<0.05). In control hearts the QT interval in the electrocardiogram shortened from 63 +/- 6 ms to 36 +/- 4 ms (n=10, P<0.05) within 4-6 min of low-flow ischemia. This shortening was completely prevented by 3 micromol/l HMR 1098 (60 +/- 5 ms before ischemia, 67 +/- 6 ms during ischemia, n=9, not significant). When rat hearts were not paced, the heart rate fell spontaneously during ischemia, and HMR 1,098 (3 micromol/l) caused only a slight, statistically non-significant reduction in aortic flow during the ischemic period. In order to investigate whether HMR 1098 shows cardiodepressant effects in a more pathophysiological model, the left descending coronary artery was occluded for 30 min followed by reperfusion for 60 min in anesthetized rats. Treatment with HMR 1098 (10 mg/kg i.v.) had no statistically significant effects on mean arterial blood pressure and heart rate during the control, ischemia and reperfusion periods. At the end of the reperfusion period, aortic blood flow was slightly reduced by HMR 1098, without reaching statistical significance (two-way analysis of ANOVA, P=0.15). Myocardial infarct size as a percentage of area at risk was not affected by HMR 1098 (vehicle: 75 +/- 3%, HMR 1098: 72 +/- 2%, n=7 in each group). In conclusion, cardiodepressant effects of HMR 1098 were observed only in isolated perfused working rat hearts which were continuously paced during global low-flow ischemia. In the model of anesthetized rats subjected to regional ischemia, HMR 1098 had no significant effect on cardiac function or infarct size.


Subject(s)
Benzamides/pharmacology , Heart/drug effects , Myocardial Ischemia/physiopathology , Potassium Channel Blockers/pharmacology , Potassium/metabolism , Thiourea/analogs & derivatives , Anesthesia , Animals , Anti-Arrhythmia Agents/pharmacology , Benzamides/administration & dosage , Glyburide/pharmacology , Heart/physiopathology , Hemodynamics/drug effects , In Vitro Techniques , Male , Models, Animal , Myocardial Contraction/drug effects , Myocardial Infarction , Myocardial Reperfusion , Rats , Rats, Wistar , Sulfonamides/pharmacology , Thiourea/pharmacology
13.
Basic Res Cardiol ; 96(2): 192-7, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11327338

ABSTRACT

During myocardial ischemia intracellular acid load increases as a consequence of anaerobic metabolism. Exchange of excessive protons for sodium via the sodium proton exchanger type 1 (NHE1) is supposed to cause intracellular sodium accumulation. The NHE1 inhibitor cariporide has been shown to inhibit ischemia and reperfusion-induced ventricular fibrillation (VF) but the mechanisms are not fully understood. During early reperfusion transient shortening of the action potential has been reported, which renders the heart susceptible to reentrant arrhythmias. In anesthetized pigs subjected to 10 min of left circumflex coronary artery (LCX) occlusion and reperfusion we have investigated whether NHE1 is involved in reperfusion-induced shortening of the monophasic action potential (MAP) taken with an epicardial probe over the ischemic area. In control pigs (n = 7) a moderate decrease in the duration of the MAP at 50 % repolarization (MAPD50) occurred during ischemia reaching 78.8 +/- 5.0% of the pre-ischemic duration at 5 min (p < 0.01) and 87.3 +/- 7.6 % after 10 min. An additional, transient but marked shortening occurred during the first 2 min of reperfusion, which fully recovered after 4 min. At 50 sec of reperfusion MAPD50 fell to 53.1 +/- 8.2 % of the pre-ischemic value corresponding to 90.1 +/- 20.2 msec of reperfusion-induced shortening. Cariporide, 3 mg/kg i.v. 5 min before occlusion (n = 6), totally prevented reperfusion-induced MAP shortening while having no effect on MAPD50 during ischemia. In conclusion, our data suggest that the immediate, transient, but strong action potential shortening during early reperfusion after 10 min of coronary ischemia is due to the activity of the NHE1.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Guanidines/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology , Action Potentials/drug effects , Animals , Blood Pressure/drug effects , Heart Rate/drug effects , Male , Swine , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/metabolism
14.
FEBS Lett ; 492(1-2): 84-9, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11248242

ABSTRACT

We report the primary sequence of TASK-4, a novel member of the acid-sensitive subfamily of tandem pore K(+) channels. TASK-4 transcripts are widely expressed in humans, with highest levels in liver, lung, pancreas, placenta, aorta and heart. In Xenopus oocytes TASK-4 generated K(+) currents displaying a marked outward rectification which was lost by elevation of extracellular K(+). TASK-4 currents were efficiently blocked by barium (83% inhibition at 2 mM), only weakly inhibited by 1 mM concentrations of quinine, bupivacaine and lidocaine, but not blocked by tetraethylammonium, 4-aminopyridine and Cs(+). TASK-4 was sensitive to extracellular pH, but in contrast to other TASK channels, pH sensitivity was shifted to more alkaline pH. Thus, TASK-4 in concert with other TASK channels might regulate cellular membrane potential over a wide range of extracellular pH.


Subject(s)
Potassium Channels, Tandem Pore Domain , Potassium Channels/genetics , Amino Acid Sequence , Animals , Atrioventricular Node/metabolism , Barium/pharmacology , Cloning, Molecular , Electrophysiology , Heart Atria/metabolism , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Oocytes , Phylogeny , Potassium Channel Blockers , Potassium Channels/metabolism , Protein Conformation , Sequence Homology, Amino Acid , Tissue Distribution , Xenopus laevis
15.
J Biol Chem ; 276(17): 13600-5, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11278406

ABSTRACT

KCNQ1 inactivation bears electrophysiological characteristics different from classical N- and C-type inactivation in Shaker-like potassium channels. However, the molecular site of KCNQ1 inactivation has not yet been determined. KCNQ2 channels do not exert a fast inactivation in contrast to KCNQ1 channels. By expressing functional chimeras between KCNQ1 and KCNQ2 in Xenopus oocytes, we mapped the region of this inactivation to transmembrane domain S5 and the pore loop H5 and finally narrowed down the site to positions Gly(272) and Val(307) in KCNQ1. Exchanging these two amino acids individually with the analogous KCNQ2 residue abolished inactivation. Furthermore, a KCNQ1-like inactivation was introduced into KCNQ2 by mutagenesis in the corresponding region, confirming its relevance for the inactivation process. As KCNQ1 inactivation involves the regions S5 and H5, it exhibits a geography distinct from N- or C-type inactivation. Native cardiac I(Ks) channels comprising KCNQ1 and accessory MinK subunits do not inactivate because of the functional interaction of KCNQ1 with MinK. Mutations in KCNQ1 can lead to long QT1 syndrome, an inherited form of arrhythmia. The long QT1 mutant KCNQ1(L273F) displays a pronounced KCNQ1 inactivation. Here we show that when expressing mutant I(Ks) channels formed from KCNQ1(L273F) and MinK, MinK association no longer eliminates KCNQ1 inactivation. This results in smaller repolarizing currents in the heart and therefore represents a novel mechanism leading to long QT syndrome.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/chemistry , Amino Acid Sequence , Animals , Arrhythmias, Cardiac/genetics , Electrophysiology , Glycine/chemistry , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , KCNQ2 Potassium Channel , Long QT Syndrome/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oocytes/metabolism , Patch-Clamp Techniques , Point Mutation , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Structure, Tertiary , RNA, Complementary/metabolism , Sequence Homology, Amino Acid , Time Factors , Valine/chemistry , Xenopus/embryology
16.
Cell Physiol Biochem ; 11(6): 321-30, 2001.
Article in English | MEDLINE | ID: mdl-11832657

ABSTRACT

Here for the first time we investigated the potential involvement of the CLC chloride channel family at the transcriptional level in different cardiovascular diseases. Northern blot and semiquantitative RT-PCR analyses were used to study the gene expression profiles of all CLC genes present in the heart and kidney; namely, CLC-2, CLC-3, CLC-4, CLC-5, CLC-6, CLC-7, CLC-K1, and CLC-K2. Rat models with distinctive cardiovascular diseases were studied: These included spontaneously hypertensive rats, nutritionally- and surgically-induced hypertensive rats with cardiac hypertrophy, as well as rats suffering from chronic heart failure due to myocardial infarction. The present data show that it was not possible to detect apparent differences in the CLC mRNA expression between the hearts and kidneys of diseased and control animals. Our data strongly suggest that altered transcript regulation of CLC chloride channels does not contribute to the cardiac and renal pathology in the examined cardiovascular diseases.


Subject(s)
Anion Transport Proteins , Cardiovascular Diseases/genetics , Chloride Channels/genetics , Disease Models, Animal , Gene Expression Profiling , Membrane Proteins , Animals , CLC-2 Chloride Channels , Male , Rats , Rats, Inbred Dahl/genetics , Rats, Inbred SHR/genetics , Rats, Sprague-Dawley , Rats, Wistar
17.
Naunyn Schmiedebergs Arch Pharmacol ; 361(4): 445-51, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10763861

ABSTRACT

Previous experimental studies showed that the benefit of ischemic preconditioning (IPC) is abolished by K(ATP) channel blockade with glibenclamide. However, the newly discovered K(ATP) channel blocker HMR 1883 (1-[[5-[2-(5-chloro-o-anisamido)ethyl]-methoxyphenyl]sulfonyl]-3-m ethylthiourea) shows marked antifibrillatory activity in the dose range of 3 mg/kg to 10 mg/kg i.v. in various experimental models without affecting blood glucose levels. In order to investigate in a head to head comparison glibenclamide and HMR 1883 with respect to their influence on IPC, experiments were performed in rabbits with ischemia-reperfusion using myocardial infarct mass as final read out. Male New Zealand White rabbits (2.6-3.0 kg) were subjected to 30-min occlusion of a branch of the left descending coronary artery (LAD) followed by 2-h reperfusion. For IPC experiments the LAD was additionally occluded for two periods of 5 min, each followed by 10-min reperfusion, before the long-term ischemia. Infarct mass was evaluated by TTC staining and expressed as a percentage of area at risk. Rabbits (n=7/group) were randomly selected to receive (i.v.) saline vehicle 5 min prior to the 30-min occlusion period in infarct studies without IPC or to receive glibenclamide (0.3 mg/kg) or HMR 1883 (3 mg/kg) in IPC experiments, these substances being given 5 min prior to the first preconditioning or 5 min prior to the long-term ischemia of 30 min. Myocardial risk mass as a percentage of left ventricular mass did not differ between groups. The same was true for the ratio of left ventricular mass to 100 g body weight. Myocardial infarct mass as a percentage of the area at risk in the saline vehicle group without IPC was 41+/-3%. Whereas glibenclamide significantly increased infarct mass (from 41+/-3% to 55+/-4%), HMR 1883 did not affect it. IPC reduced infarct mass from 41+/-3% to 21+/-4% (P<0.05 vs. control without IPC). Glibenclamide given prior to IPC or prior to the long-term ischemia totally abolished the IPC effect (42+/-2% and 55+/-4%, respectively; P<0.05 vs. control). In contrast, HMR 1883 under the same conditions did not affect infarct size when given prior to IPC or prior to the long-term ischemia (21+/-3% and 26+/-2%, respectively). The monophasic action potential duration (MAP50) was reduced from 103+/-3 ms under normoxic conditions to 82+/-2 ms, 5 min after ischemia in the absence of drugs. This ischemia-induced shortening of the MAP was prevented by both HMR 1883 (MAP50 103+/-3 ms) and glibenclamide (MAP50 106+/-3 ms). In conclusion, although both K(ATP) channel blockers prevented ischemia-induced shortening of MAP, HMR 1883 did not abolish the beneficial effects of IPC on myocardial infarct mass in rabbits, whereas glibenclamide totally reversed this cardioprotective effect of IPC. This suggests that the sarcolemmal ATP-sensitive potassium channels are not involved in the mechanism of IPC.


Subject(s)
Adenosine Triphosphate/metabolism , Ischemic Preconditioning, Myocardial , Myocardial Infarction/pathology , Potassium Channel Blockers , Sulfonamides/pharmacology , Thiourea/analogs & derivatives , Action Potentials , Animals , Blood Glucose/metabolism , Glyburide/pharmacology , Heart/physiopathology , Male , Myocardial Infarction/blood , Myocardial Infarction/physiopathology , Rabbits , Thiourea/pharmacology
18.
J Biol Chem ; 275(29): 22395-400, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10787416

ABSTRACT

We have isolated KCNQ5, a novel human member of the KCNQ potassium channel gene family that is differentially expressed in subregions of the brain and in skeletal muscle. When expressed in Xenopus oocytes, KCNQ5 generated voltage-dependent, slowly activating K(+)-selective currents that displayed a marked inward rectification at positive membrane voltages. KCNQ5 currents were insensitive to the K(+) channel blocker tetraethylammonium but were strongly inhibited by the selective M-current blocker linopirdine. Upon coexpression with the structurally related KCNQ3 channel subunit, current amplitudes increased 4-5-fold. Compared with homomeric KCNQ5 currents, KCNQ3/KCNQ5 currents also displayed slower activation kinetics and less inward rectification, indicating that KCNQ5 combined with KCNQ3 to form functional heteromeric channel proteins. This functional interaction between KCNQ5 and KCNQ3, a component of the M-channel, suggests that KCNQ5 may contribute to a diversity of heteromeric channels underlying native neuronal M-currents.


Subject(s)
Neurons/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Genetic Variation , Humans , Ion Transport , KCNQ Potassium Channels , Molecular Sequence Data , Potassium/metabolism , Potassium Channels/metabolism , Sequence Alignment , Xenopus
19.
Naunyn Schmiedebergs Arch Pharmacol ; 361(2): 155-60, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10685870

ABSTRACT

ATP-sensitive potassium channels (KATP) open during myocardial ischemia. The ensuing repolarising potassium efflux shortens the action potential. Accumulation of extracellular potassium is able to partially depolarise the membrane, reducing the upstroke velocity of the action potential and thereby impairing impulse conduction. Both mechanisms are believed to be involved in the development of reentrant arrhythmias during cardiac ischemia. The sulfonylthiourea HMR 1883 (1-[[5-[2-(5-chloro-O-anisamido)ethyl]-methoxyphenyl]sulfonyl]-3-m ethylthiourea) was designed as a cardioselective KATP channel blocker for the prevention of arrhythmic sudden death in patients with ischemic heart disease. The aim of this study was to show that this compound, which has already shown antifibrillatory efficacy in dogs and rats, is able to inhibit ischemic changes of the action potential induced by coronary artery occlusion in anesthetised pigs. Action potentials were taken in situ with the technique of monophasic action potential (MAP) recording. In a control group (n=7), three consecutive occlusions of a small branch of the left circumflex coronary artery resulted in reproducible reductions in MAP duration and a decrease in upstroke velocity. In a separate group (n=7), HMR 1883 (3 mg/kg i.v.) significantly (P<0.05) reduced the ischemia-induced shortening of the MAP: during the first and second control occlusion of the coronary artery in the HMR 1883-group, MAP50 duration shortened from 218.5 +/- 3.0 ms to 166.7 +/- 3.3 ms and from 219.7 +/- 4.5 ms to 164.9 +/- 1.8 ms, respectively. After HMR 1883, during the third occlusion, MAP duration decreased from 226.9 +/- 3.6 ms to 205.3 +/- 4.3 ms only corresponding to 59% inhibition. HMR 1883 also improved the upstroke velocity of the MAP, which was depressed by ischemia: in the two preceding control occlusions ischemia prolonged the time to peak of the MAP, an index for upstroke velocity, from 10.83 +/- 0.43 ms to 39.42 +/- 1.60 ms and from 12.97 +/- 0.40 ms to 37.17 +/- 2.98 ms, respectively. With HMR 1883, time to peak during ischemia rose from 12.42 +/- 0.51 ms to 25.53+/-2.51 ms only, corresponding to an average inhibitory effect of 53.4%. The irregular repolarisation contour of the ischemic MAP was also improved. In conclusion, the present results indicate that HMR 1883 effectively blocks myocardial KATP channels during coronary ischemia in anesthetised pigs, preventing an excessive shortening of the action potential and improving excitation propagation.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Heart/drug effects , Myocardial Ischemia/physiopathology , Potassium Channel Blockers , Sulfonamides/pharmacology , Thiourea/analogs & derivatives , Action Potentials/drug effects , Anesthesia , Animals , Glyburide/pharmacology , Guinea Pigs , Heart/physiology , Male , Myocardial Reperfusion , Swine , Thiourea/pharmacology
20.
Naunyn Schmiedebergs Arch Pharmacol ; 362(6): 480-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11138839

ABSTRACT

Chromanol HMR 1556 [(3R,4S)-(+)-N-[3-hydroxy-2,2-dimethyl-6-(4,4,4-trifluorobutoxy)chroman-4-yl]-N-methylmethanesulfonamide], a novel inhibitor of the slow component of the delayed outward current in heart muscle cells (IKs), has been characterized in several in-vitro systems. mRNA encoding for the human protein minK was injected into Xenopus oocytes, leading to the expression of IKs channels. HMR 1556 inhibited this current half-maximally at a concentration of 120 nmol/l (IC50). Expression of the K+ channels Herg, Kv 1.5, Kv 1.3 and Kir2.1, and also the cationic current HCN2, were blocked little or not at all by 10 micromol/l HMR 1556. In isolated ventricular myocytes from the guinea pig the whole-cell patch-clamp method revealed inhibition of the IKs current with an IC50, of 34 nmol/l. Other current components, like IKr and IK1. were only slightly blocked at an HMR 1556 concentration of 10 micromol/l, whereas 10 micromol/l HMR 1556 inhibited the transient outward current I(to) and the sustained outward current I(sus) in rat ventricular myocytes by 25% and 36%, respectively. The L-type Ca2+ channel in guinea pig cardiomyocytes was blocked by 10 micromol/l HMR 1556 by 31%. Guinea pig right papillary muscles were investigated by the micropuncture technique at various pacing rates. In the frequency range of 0.5-7 Hz HMR 1556 (1 micromol/l) caused a prolongation of the action potential duration at 90% repolarization (APD90) by 19%-27%. In the presence of isoproterenol (10 micromol/l) the prolongation of the APD90 was more pronounced at low pacing rates (47% at 0.5 Hz and 35% at 1 Hz, compared with 25% at 7 Hz). The monophasic action potential was recorded in Langendorff-perfused guinea pig hearts. In spontaneously beating preparations, HMR 1556, at 0.1 micromol/l and 1 micromol/l, prolonged the MAPD90 by 3% and 10%, respectively, with no further prolongation at 10 micromol/l. The prolongation was much greater at low pacing rates [25% at 100 beats per min (bpm) and 13% at 150 bpm] than at fast pacing rates (9% at 350 bpm). The left ventricular pressure LVPmax was not affected at 1 micromol/l HMR 1556, but it decreased by 15% at 10 micromol/l. Other parameters, like the heart rate and coronary flow, were only slightly decreased at 1 micromol/l HMR 1556. In conclusion, HMR 1556 is a potent and selective inhibitor of the IKs current in guinea pig ventricular myocytes. The prolongation of the action potential duration is maintained at fast pacing rates.


Subject(s)
Chromans/pharmacology , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Sulfonamides/pharmacology , Animals , Atrial Function , Guinea Pigs , Heart/drug effects , Heart/physiology , Heart Atria/drug effects , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Myocardium/cytology , Oocytes/drug effects , Oocytes/physiology , Papillary Muscles/drug effects , Papillary Muscles/physiology , Patch-Clamp Techniques , Perfusion , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels/physiology , Purkinje Fibers/drug effects , Purkinje Fibers/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...