Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Commun ; 11(1): 3071, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32555162

ABSTRACT

Unlimited access to calorie-dense, palatable food is a hallmark of Western societies and substantially contributes to the worldwide rise of metabolic disorders. In addition to promoting overconsumption, palatable diets dampen daily intake patterns, further augmenting metabolic disruption. We developed a paradigm to reveal differential timing in the regulation of food intake behavior in mice. While homeostatic intake peaks in the active phase, conditioned place preference and choice experiments show an increased sensitivity to overeating on palatable food during the rest phase. This hedonic appetite rhythm is driven by endogenous circadian clocks in dopaminergic neurons of the ventral tegmental area (VTA). Mice with disrupted clock function in the VTA lose their hedonic overconsumption rhythms without affecting homeostatic intake. These findings assign a functional role of VTA clocks in modulating palatable feeding behaviors and identify a potential therapeutic route to counteract hyperphagy in an obesogenic environment.


Subject(s)
Circadian Rhythm , Dopaminergic Neurons/physiology , Feeding Behavior , Ventral Tegmental Area/physiology , Animals , Appetite , Behavior, Animal , Choice Behavior , Homeostasis , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Oscillometry
2.
J Endocrinol ; 231(3): 209-221, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27660201

ABSTRACT

In modern societies, the risk of developing a whole array of affective and somatic disorders is associated with the prevalence of frequent psychosocial stress. Therefore, a better understanding of adaptive stress responses and their underlying molecular mechanisms is of high clinical interest. In response to an acute stressor, each organism can either show passive freezing or active fight-or-flight behaviour, with activation of sympathetic nervous system and the hypothalamus-pituitary-adrenal (HPA) axis providing the necessary energy for the latter by releasing catecholamines and glucocorticoids (GC). Recent data suggest that stress responses are also regulated by the endogenous circadian clock. In consequence, the timing of stress may critically affect adaptive responses to and/or pathological effects of repetitive stressor exposure. In this article, we characterize the impact of predictable social defeat stress during daytime versus nighttime on bodyweight development and HPA axis activity in mice. While 19 days of social daytime stress led to a transient reduction in bodyweight without altering HPA axis activity at the predicted time of stressor exposure, more detrimental effects were seen in anticipation of nighttime stress. Repeated nighttime stressor exposure led to alterations in food metabolization and reduced HPA axis activity with lower circulating adrenocorticotropic hormone (ACTH) and GC concentrations at the time of predicted stressor exposure. Our data reveal a circadian gating of stress adaptation to predictable social defeat stress at the level of the HPA axis with impact on metabolic homeostasis underpinning the importance of timing for the body's adaptability to repetitive stress.


Subject(s)
Circadian Rhythm/physiology , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Stress, Psychological/physiopathology , Adaptation, Physiological , Adrenocorticotropic Hormone/physiology , Animals , Arginine Vasopressin/genetics , Arginine Vasopressin/physiology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/physiology , Energy Metabolism , Glucocorticoids/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
Curr Pharm Des ; 12(18): 2271-83, 2006.
Article in English | MEDLINE | ID: mdl-16787254

ABSTRACT

Human ether-a-go-go-related gene (hERG) potassium channels conduct the rapid component of the delayed rectifier potassium current, IKr, which is crucial for repolarization of cardiac action potentials. Moderate hERG blockade may produce a beneficial class III antiarrhythmic effect. In contrast, a reduction in hERG currents due to either genetic defects or adverse drug effects can lead to hereditary or acquired long QT syndromes characterized by action potential prolongation, lengthening of the QT interval on the surface ECG, and an increased risk for "torsade de pointes" arrhythmias and sudden death. This undesirable side effect of non-antiarrhythmic compounds has prompted the withdrawal of several blockbuster drugs from the market. Studies on mechanisms of hERG channel inhibition provide significant insights into the molecular factors that determine state-, voltage-, and use-dependency of hERG current block. In addition, crucial properties of the high-affinity drug binding site in hERG and its interaction with drug molecules have been identified, providing the basis for more refined approaches in drug design, safety pharmacology and in silico modeling. Recently, mutations in hERG have been shown to cause current increase and hereditary short QT syndrome with a high risk for life-threatening arrhythmias. Finally, the discovery of adrenergic mechanisms of hERG channel regulation as well as the development of strategies to enhance hERG currents and to modify intracellular hERG protein processing may provide novel antiarrhythmic options in repolarization disorders. In conclusion, the increasing understanding of hERG channel function and molecular mechanisms of hERG current regulation could improve prevention and treatment of hERG-associated cardiac repolarization disorders.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Antidepressive Agents, Tricyclic/adverse effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Heart/drug effects , Long QT Syndrome/etiology , Potassium Channel Blockers/adverse effects , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Animals , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Heart/innervation , Humans , Ion Channel Gating , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Models, Molecular , Myocardium/metabolism , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/therapeutic use , Protein Conformation , Receptors, Adrenergic/drug effects , Receptors, Adrenergic/metabolism , Stress, Physiological/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
4.
Herzschrittmacherther Elektrophysiol ; 16(4): 229-38, 2005 Dec.
Article in German | MEDLINE | ID: mdl-16362729

ABSTRACT

The last decade has seen rapid progress in our understanding of the molecular basis of arrhythmias, particularly concerning hereditary arrhythmia syndromes. This has led to significant improvement regarding differentiation, risk stratification and therapy in these patients and their families. However, there is mounting evidence that the knowledge obtained by studying these rare monogenic disorders will also enable us to dissect the molecular mechanisms underlying polygenetic and multi-factorial arrhythmias that are by far more common in clinical practice. The goal of this review is to give a brief overview of current knowledge on the molecular basis of primary electrical heart diseases. A focus is on the long QT syndrome.


Subject(s)
Action Potentials , Arrhythmias, Cardiac/physiopathology , Heart Conduction System/physiopathology , Models, Cardiovascular , Potassium Channels/metabolism , Animals , Humans , Molecular Biology/methods
5.
Biochim Biophys Acta ; 1690(3): 185-92, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15511625

ABSTRACT

Romano-Ward syndrome (RWS), the autosomal dominant form of the congenital long QT syndrome, is characterised by prolongation of the cardiac repolarisation process associated with ventricular tachyarrhythmias of the torsades de pointes type. Genetic studies have identified mutations in six ion channel genes, KCNQ1, KCNH2, SCN5A, KCNE1 and KCNE2 and the accessory protein Ankyrin-B gene, to be responsible for this disorder. Single-strand conformation polymorphism (SSCP) analysis and subsequent DNA sequence analysis have identified a KCNQ1 mutation in a family that were clinically conspicuous due to several syncopes and prolonged QTc intervals in the ECG. The mutant subunit was expressed and functionally characterised in the Xenopus oocyte expression system. A novel heterozygous missense mutation with a C to T transition at the first position of codon 343 (CCA) of the KCNQ1 gene was identified in three concerned family members (QTc intervals: 500, 510 and 530 ms, respectively). As a result, proline 343 localised within the highly conserved transmembrane segment S6 of the KCNQ1 channel is replaced by a serine. Co-expression of mutant (KCNQ1-P343S) and wild-type (KCNQ1) cRNA in Xenopus oocytes produced potassium currents reduced by approximately 92%, while IKs reconstitution experiments with a combination of KCNQ1 mutant, wild-type and KCNE1 subunits yielded currents reduced by approximately 60%. A novel mutation (P343S) identified in the KCNQ1 subunit gene of three members of a RWS family showed a dominant-negative effect on native IKs currents leading to prolongation of the heart repolarisation and possibly increases the risk of malign arrhythmias with sudden cardiac death.


Subject(s)
Mutation/genetics , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Romano-Ward Syndrome/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Mutational Analysis , Electrophysiology , Female , Gene Expression , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Male , Molecular Sequence Data , Oocytes/metabolism , Patch-Clamp Techniques , Pedigree , Phenotype , Polymorphism, Single-Stranded Conformational , Potassium/metabolism , Potassium Channels, Voltage-Gated/chemistry , Proline/genetics , Sequence Alignment , Xenopus laevis
6.
J Pharmacol Exp Ther ; 297(2): 753-61, 2001 May.
Article in English | MEDLINE | ID: mdl-11303067

ABSTRACT

Human ether-a-go-go-related gene (HERG) potassium channels are one primary target for the pharmacological treatment of cardiac arrhythmias by class III antiarrhythmic drugs. These drugs are characterized by high antiarrhythmic efficacy, but they can also initiate life-threatening "torsade de pointes" tachyarrhythmias. Recently, it has been suggested that combining potassium and calcium channel blocking mechanisms reduces the proarrhythmic potential of selective class III antiarrhythmic agents. BRL-32872 is a novel antiarrhythmic drug that inhibits potassium and calcium currents in isolated cardiomyocytes. In our study, we investigated the effects of BRL-32872 on cloned HERG channels heterologously expressed in Xenopus oocytes. Using the two-microelectrode voltage clamp technique, we found that BRL-32872 caused a high-affinity, state-dependent block of open HERG channels (IC(50) = 241 nM) in a frequency-dependent manner with slow unbinding kinetics. Inactivated channels mainly had to open to be blocked by BRL-32872. The HERG S620T mutant channel, which has a strongly reduced degree of inactivation, was 51-fold less sensitive to BRL-32872 block, indicating that BRL-32872 binding was enhanced by the inactivation process. In an additional approach, we studied HERG channels expressed in a human cell line (HEK 293) using the whole-cell patch-clamp technique. BRL-32872 inhibited HERG currents in HEK 293 cells in a dose-dependent manner, with an IC(50) value of 19.8 nM. We conclude that BRL-32872 is a potent blocker of HERG potassium channels, which accounts for the class III antiarrhythmic action of BRL-32872.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzamides/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Myocardium/metabolism , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Trans-Activators , Animals , Cloning, Molecular , ERG1 Potassium Channel , Electrophysiology , Ether-A-Go-Go Potassium Channels , Humans , Mutation , Oocytes/drug effects , Oocytes/metabolism , Potassium Channels/genetics , Transcriptional Regulator ERG , Xenopus laevis
7.
Cardiovasc Res ; 49(2): 361-70, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11164846

ABSTRACT

OBJECTIVE: The aryloxypropanolamine carvedilol is a multiple action cardiovascular drug with blocking effects on alpha-receptors, beta-receptors, Ca(2+)-channels, Na(+)-channels and various native cardiac K(+) channels, thereby prolonging the cardiac action potential. In a number of clinical trials with patients suffering from congestive heart failure, carvedilol appeared to be superior to other beta-blocking agents in reducing total mortality. Given the multiple pharmacological actions of carvedilol, this may be due to specific channel blockade rather than beta-antagonistic activity. Since human ether-a-go-go related gene (HERG) K(+)channels play a critical role in the pathogenesis of cardiac arrhythmias and sudden cardiac death, the effects of carvedilol on HERG K(+)channels were investigated. METHODS: Double-electrode voltage-clamp experiments were performed on HERG potassium channels which were expressed heterologously in Xenopus oocytes. RESULTS: Carvedilol at a concentration of 10 microM blocked HERG potassium tail currents by 47%. The electrophysiological characteristics of HERG, i.e. activation, steady-state inactivation and recovery from inactivation were not affected by carvedilol. Inhibition of current gradually increased from 0% immediately after the test pulse to about 80% at 600 ms with subsequent marginal changes of current kinetics during the resting 29 s, indicating a very fast open channel block by carvedilol as the major blocking mechanism. CONCLUSION: This is the first study demonstrating that carvedilol blocks HERG potassium channels. The biophysical data presented in this study with a potentially antiarrhythmic effect may contribute to the positive outcome of clinical trials with carvedilol.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Carbazoles/pharmacology , Cation Transport Proteins , Oocytes/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Propanolamines/pharmacology , Animals , Carvedilol , Depression, Chemical , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels , Female , In Vitro Techniques , Ion Channel Gating/drug effects , Patch-Clamp Techniques , Xenopus
8.
J Biol Chem ; 275(35): 26743-7, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10823835

ABSTRACT

The slow component of the delayed rectifier potassium current (IKs) plays an important role during repolarization in the human heart. Life-threatening arrhythmias can be triggered by sympathetic stimulation, presumably acting on IKs. The ion channel responsible for the IKs current is made of two proteins, the KvLQT1 protein and the MinK protein. In this study, we investigated the effects of adrenergic stimulation on the KvLQT1/MinK channel by coexpressing KvLQT1/MinK channels with the human beta(3)-adrenoreceptor subunit heterologously in Xenopus oocytes. Western blot experiments revealed that beta(3)-adrenoreceptor proteins appear in the cell membrane of Xenopus oocytes, when the corresponding cRNA was injected. In electrophysiological measurements we found that stimulation with the beta-adrenergic agonist isoproterenol increased the current amplitude of the beta(3)/KvLQT1/MinK complex up to 237% with an ED(50) of 8 nm, a value similar to that found on IKs in guinea pig cardiomyocytes. When oocytes with beta(3)/KvLQT1/MinK were preincubated with cholera toxin (2 microg/ml), an activator of G(S) proteins, the basal current amplitude of the beta(3)/KvLQT1/MinK complex was increased 3.1-fold, and the current amplitude increase by isoproterenol was drastically reduced, indicating that the signal transduction cascade was mediated via G(s) proteins. The knowledge about functional coupling of the human beta(3)-adrenoreceptor to KvLQT1/MinK channels reveals interesting aspects about the genesis and therapy of arrhythmias.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Receptors, Adrenergic, beta/metabolism , Animals , GTP-Binding Proteins/metabolism , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Long QT Syndrome/metabolism , Protein Binding , Receptors, Adrenergic, beta-3 , Xenopus laevis
9.
Trends Cardiovasc Med ; 10(5): 205-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-11282296

ABSTRACT

Protein kinase A is an enzyme that regulates many cellular processes and is activated in many pathological conditions such as stress and various types of heart failure. Recently it has been shown that protein kinase A couples functionally to the HERG cardiac potassium channel, thereby altering repolarization in the heart. This link between a repolarizing potassium channel and the protein kinase system of cardiac cells may contribute to arrhythmogenesis and may become a target for future approaches to antiarrhythmic therapy.


Subject(s)
Cation Transport Proteins , Cyclic AMP-Dependent Protein Kinases/physiology , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Trans-Activators , Animals , ERG1 Potassium Channel , Electrophysiology , Ether-A-Go-Go Potassium Channels , Heart/physiology , Humans , Transcriptional Regulator ERG
10.
J Biol Chem ; 274(39): 27457-62, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488078

ABSTRACT

We investigated the role of protein kinase A (PKA) in regulation of the human ether-a-go-go-related gene (HERG) potassium channel activation. HERG clones with single mutations destroying one of four consensus PKA phosphorylation sites (S283A, S890A, T895A, S1137A), as well as one clone carrying all mutations with no PKA phosphorylation sites (HERG 4M) were constructed. These clones were expressed heterologously in Xenopus oocytes, and HERG potassium currents were measured with the two microelectrode voltage clamp technique. Application of the cAMP-specific phosphodiesterase (PDE IV) inhibitor Ro-20-1724 (100 microM), which results in an increased cAMP level and PKA stimulation, induced a reduction of HERG wild type outward currents by 19.1% due to a shift in the activation curve of 12.4 mV. When 100 microM Ro-20-1724 was applied to the HERG 4M channel, missing all PKA sites, there was no significant shift in the activation curve, and the current amplitude was not reduced. Furthermore, the adenylate cyclase activator forskolin that leads to PKA activation (400 microM, 60 min), shifted HERG wild type channel activation by 14.1 mV and reduced currents by 39.9%, whereas HERG 4M channels showed only a small shift of 4.3 mV and a weaker current reduction of 22.3%. We conclude that PKA regulates HERG channel activation, and direct phosphorylation of the HERG channel protein has a functional role that may be important in regulation of cardiac repolarization.


Subject(s)
Cation Transport Proteins , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Trans-Activators , 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Colforsin/pharmacology , Consensus Sequence , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Female , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Molecular , Mutagenesis, Site-Directed , Oocytes/physiology , Phosphorylation , Potassium Channels/chemistry , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Transcriptional Regulator ERG , Xenopus laevis
11.
Am J Physiol ; 277(1): H199-210, 1999 07.
Article in English | MEDLINE | ID: mdl-10409198

ABSTRACT

The rapid, repolarizing K(+) current in cardiomyocytes (I(Kr)) has unique inwardly rectifying properties that contribute importantly to the downstroke of the cardiac action potential. The human ether-à-go-go-related gene (HERG) expresses a macroscopic current virtually identical to I(Kr), but a description of the single-channel properties that cause rectification is lacking. For this reason we measured single-channel and macropatch currents heterologously expressed by HERG in Xenopus oocytes. Our experiments had two main findings. First, the single-channel current-voltage relation showed inward rectification, and conductance was 9.7 pS at -100 mV and 3.9 pS at 100 mV when measured in symmetrical 100 mM K(+) solutions. Second, single channels frequently showed no openings during depolarization but nevertheless revealed bursts of openings during repolarization. This type of gating may explain the inward rectification of HERG currents. To test this hypothesis, we used a three-closed state kinetics model and obtained rate constants from fits to macropatch data. Results from the model are consistent with rapid inactivation from closed states as a significant source of HERG rectification.


Subject(s)
Action Potentials , Cation Transport Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Animals , Electrophysiology , Ether-A-Go-Go Potassium Channels , Oocytes , Xenopus
12.
Naunyn Schmiedebergs Arch Pharmacol ; 359(3): 212-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10208308

ABSTRACT

The human ether-a-go-go-related gene (HERG) encodes a K+ channel with biophysical properties nearly identical to the rapid component of the cardiac-delayed rectifier K+ current (I(Kr)). HERG channels are one primary target for the pharmacological management of arrhythmias. In this study, we investigated the acute effects of the class III antiarrhythmic drug amiodarone on HERG channels expressed heterologously in Xenopus oocytes by use of the two-microelectrode voltage clamp technique. Amiodarone blocked HERG channels with an IC50 of 9.8 microM with a maximum outward tail current reduction of 62.8%. The block consisted of two main components, a closed channel block that could not be reversed within the time of experiments and an open channel block with a slow unblock, having a recovery time constant of 73 s at -80 mV. Inactivation of the HERG channel at very positive potentials could not prevent amiodarone block. These results indicate that HERG channels can be blocked by amiodarone in closed, open and inactivated states. The block of open channels was cumulative, use-dependent and voltage-dependent. In summary, our data suggest that the strong class III antiarrhythmic action of amiodarone is at least partially based upon its acute inhibitory effects on HERG potassium channels.


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Oocytes/drug effects , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Trans-Activators , Xenopus/genetics , Animals , Cloning, Molecular , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , In Vitro Techniques , Patch-Clamp Techniques , Potassium Channels/drug effects , Time Factors , Transcriptional Regulator ERG
13.
J Biol Chem ; 273(39): 25285-91, 1998 Sep 25.
Article in English | MEDLINE | ID: mdl-9737994

ABSTRACT

We investigated the effects of the phorbol ester phorbol 12-myristate 13-acetate (PMA) on the rapid component of the delayed rectifier potassium current, IKr, in guinea pig cardiomyocytes and found that the IKr current amplitude was reduced by 20% with 10 nM PMA and 44% with 100 nM PMA. The ether-a-go-go-related gene (HERG) encodes IKr in human heart. We expressed HERG heterologously in Xenopus oocytes and investigated the effects of PMA on the delayed rectifier potassium current. Upon application of PMA in a concentration of 100 nM, we found a similar reduction of HERG outward current amplitude by 59%. This reduction was due to a shift in the HERG activation curve by 37 mV. The ED50 for the PMA-induced shift was 9.0 nM. The inactive 4alpha-phorbol 12-myristate 13-acetate (4alpha-PMA) had no effect. PMA is known to act by stimulating distinct protein kinase cascades. Additional application of the specific protein kinase C inhibitors chelerythrine (10 microM) or bisindolylmaleimide (1 microM) could not attenuate the PMA-induced shift. In contrast, the shift by PMA was reduced significantly when the specific protein kinase A (PKA) inhibitors H89 (50 microM) or KT5720 (2.5 microM) were applied. Forskolin (400 microM), an activator of the adenylate cyclase that results in PKA activation, shifted the HERG activation curve by 14 mV. Moreover the specific protein kinase C activator 1-stearoyl-2-arachidonylglycerol (10 microM) showed no effect. Our data suggest that mainly PKA is mediating the shift of the HERG activation kinetics.


Subject(s)
Cation Transport Proteins , Cyclic AMP-Dependent Protein Kinases/metabolism , Long QT Syndrome/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Alkaloids , Animals , Benzophenanthridines , Enzyme Activation , Enzyme Inhibitors/pharmacology , Ether-A-Go-Go Potassium Channels , Guinea Pigs , Indoles/pharmacology , Maleimides/pharmacology , Membrane Potentials , Myocardium/metabolism , Myocardium/pathology , Patch-Clamp Techniques , Phenanthridines/pharmacology , Potassium Channels/drug effects , Protein Kinase C/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
14.
Circ Res ; 82(3): 386-95, 1998 Feb 23.
Article in English | MEDLINE | ID: mdl-9486667

ABSTRACT

The human ether-a-go-go-related gene (HERG) encodes a K+ channel with biophysical properties nearly identical to the rapid component of the cardiac delayed rectifier K+ current (IKr). HERG/IKr channels are a prime target for the pharmacological management of arrhythmias and are selectively blocked by class III antiarrhythmic methanesulfonanilide drugs, such as dofetilide, E4031, and MK-499, at submicromolar concentrations. By contrast, the closely related bovine ether-a-go-go channel (BEAG) is 100-fold less sensitive to dofetilide. To identify the molecular determinants for dofetilide block, we first engineered chimeras between HERG and BEAG and then used site-directed mutagenesis to localize single amino acid residues responsible for block. Using constructs heterologously expressed in Xenopus oocytes, we found that transplantation of the S5-S6 linker from BEAG into HERG removed high-affinity block by dofetilide. A point mutation in the S5-S6 linker region, HERG S620T, abolished high-affinity block and interfered with C-type inactivation. Thus, our results indicate that important determinants of dofetilide binding are localized to the pore region of HERG. Since the loss of high-affinity drug binding was always correlated with a loss of C-type inactivation, it is possible that the changes observed in drug binding are due to indirect allosteric modifications in the structure of the channel protein and not to the direct interaction of dofetilide with the respective mutated site chains. However, the chimeric approach was not able to identify domains outside the S5-S6 linker region of the HERG channel as putative candidates involved in drug binding. Moreover, the reverse mutation BEAG T432S increased the affinity of BEAG K+ channels for dofetilide, whereas C-type inactivation could not be recovered. Thus, the serine in position HERG 620 may participate directly in dofetilide binding; however, an intact C-type inactivation process seems to be crucial for high-affinity drug binding.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Phenethylamines/pharmacology , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Sulfonamides/pharmacology , Trans-Activators , Amino Acid Sequence , Animals , Cattle , Chimera , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Female , Humans , Mutagenesis, Site-Directed , Oocytes , Patch-Clamp Techniques , Potassium Channels/physiology , Transcriptional Regulator ERG , Xenopus laevis
15.
J Tongji Med Univ ; 18(1): 13-7, 1998.
Article in English | MEDLINE | ID: mdl-10806794

ABSTRACT

The aim of this study was to compare the effects of d, l-sotalol and d-Sotalol on the delayed rectifier K+ outward current in the presence of isoproterenol at different concentrations. Time-dependent delayed rectifier K+ outward currents were measured in isolated guinea pig single myocytes using the whole-cell configuration of the patch-clamp technique. Currents were measured in response to 300 ms depolarizing pulses from a holding potential of -40 mV in three experimental protocols [control, isoproterenol (10(-9) mol/L-10(-6) mol/L), and isoproterenol (10(-9) mol/L-10(-6) mol/L) plus either d, l-Sotalol (10(-4) mol/L) or d-Sotalol (10(-4) mol/L)]. IK tail currents were measured upon repolarization to -40 mV. It was found that IK was significantly amplified in the presence of isoproterenol (10(-9) mol/L-10(-6) mol/L) plus d-Sotalol. At 10(-8) mol/L isoproterenol, IK was increased by 92.7% +/- 17.1% (P < 0.05) and 54.3% +/- 13.4% after d-Sotalol addition (P < 0.05). In contrast, d, l-Sotalol completely conteracted the increase of Ik by isoproterenol (< 10(-8) mol/L), and compared to control, IK was decreased by 35.6% +/- 8.1% at 10(-8) mol/L isoproterenol plus d, l-Sotalol (P < 0.05). It is concluded that the beta-adrenergic blocking property of d, l-Sotalol but not that of d-Sotalol maintains the delayed rectifier K+ outward current blockade in the presence of isoproterenol in guinea pig myocytes. This might contribute to a superior antiarrhythmic efficacy as compared to d-Sotalol.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Isoproterenol/pharmacology , Myocardium/cytology , Sotalol/pharmacology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Guinea Pigs , Membrane Potentials/drug effects , Potassium Channels/drug effects , Refractory Period, Electrophysiological , Stereoisomerism
16.
J Biol Chem ; 272(41): 25824-31, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9325312

ABSTRACT

Kvbeta subunits have been shown to affect kinetic properties of voltage-gated K+ channel Kv1alpha subunits and increase the number of cell surface dendrotoxin-binding sites when coexpressed with Kv1. 2. Here, we show that Kvbeta1.2 alters both current expression and gating of Kvalpha1 channels and that each effect is mediated by a distinct Kvbeta1.2 domain. The Kvbeta1.2 N terminus or Kvalpha1-blocking domain introduced steady state current block, an apparent negative shift in steady state activation, and a slowing of deactivation along with a dramatic reduction in single channel open probability. N-terminal deletions of Kvbeta1.2 no longer altered channel kinetics but promoted dramatic increases in Kv1.2 current. The conserved Kvbeta1 C terminus or Kvalpha1 expression domain alone was sufficient to increase the number of functional channels. The same effect was observed with the normally noninactivating subunit, Kvbeta2. By contrast, Kv1.5 currents were reduced when coexpressed with either the Kvbeta1 C terminus or Kvbeta2, indicating that the Kvalpha1 expression domain has Kvalpha1 isoform-specific effects. Our results demonstrate that Kvbeta subunits consist of two domains that are separable on the basis of both primary structure and functional modulation of voltage-gated K+ channels.


Subject(s)
Ion Channel Gating , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Animals , Binding Sites , Elapid Venoms/metabolism , Kinetics , Kv1.2 Potassium Channel , Kv1.5 Potassium Channel , Oocytes/metabolism , Protein Conformation , Surface Properties , Xenopus
17.
J Biol Chem ; 272(45): 28232-6, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9353274

ABSTRACT

Experiments were carried out to determine whether coinjection of Kvalpha1.2 with inactivating and noninactivating Kvbeta subunits would produce currents with intermediate kinetics and channel complexes containing a mixture of these subunits. Upon coexpression with a saturating amount of Kvbeta1.2 and increasing levels of a noninactivating deletion mutant of Kvbeta1.2, we show that macroscopic Kvalpha1.2 currents have levels of fractional inactivation and inactivation time constants that are intermediate between those obtained with either the inactivating Kvbeta1.2 or the noninactivating Kvbeta1.2 mutant. We also find that coexpression of Kvalpha1.2 with saturating amounts of Kvbeta1.2 and the deletion mutant produces a population of single channels with properties intermediate to either the inactivating or noninactivating parental phenotype. Our data can best be explained by the presence of an intermediate population of heterooligomeric channels consisting of Kvalpha1.2 with different combinations of both types of subunits. Since Kvalpha1.2 subunits coexist in cells with inactivating and noninactivating Kvbeta subunits, our findings suggest that heterooligomeric assembly of these subunits occurs to increase the range of K+ current kinetics and expression levels.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Potassium/metabolism , Animals , Kinetics , Kv1.2 Potassium Channel , Mutation , Oocytes/metabolism , Potassium Channels/genetics , Protein Conformation , Sequence Deletion , Xenopus
18.
Support Care Cancer ; 5(5): 387-95, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9322351

ABSTRACT

There is still controversy as to what constitutes the optimal therapy for acute and delayed chemotherapy-induced emesis and nausea. We conducted a three-armed randomized multi-centre study in 193 chemotherapy-naive patients receiving highly emetogenic chemotherapy inducing both acute and delayed symptoms (cisplatin > or = 50 mg/m2, carboplatin > or = 300 mg/m2, cyclophosphamide > or = 750 mg/m2, ifosfamide > or = 1.5 g/m2 on day 1). Group A: 1 x 5 mg tropisetron i.v. on day 1 + 2, then 10 mg p.o. (oral dose now recommended: 5 mg); group B: tropisetron as for A+dexamethasone, 20 mg i.v., on days 1 + 2, then 4 mg i.v./p.o.; group C: tropisetron as for A+metoclopramide, 20 mg i.v. +2 x 10 mg p.o. on day 1, then 3 x 10 mg p.o. Treatment was continued for at least 2 days after the end of chemotherapy. Tropisetron+dexamethasone was significantly superior to tropisetron alone both for acute (P = 0.0064) and delayed (P = 0.0053) emesis. Complete control of acute and delayed emesis (nausea) was achieved in 80% (75%) and 53% (46%) in group A, 97% (90%) and 80% (58%) in group B, and 86% (80%) and 49% (45%) in group C. Patients completely asymptomatic during the whole cycle accounted for 26% of those in group A, 49% in group B and 28% in group C. The most frequent adverse events were constipation (16.6%), headache (7.3%) and tiredness (7.3%). Once-daily tropisetron+dexamethasone over several days is well tolerated and is a simple means of achieving further significant improvement in the efficacy of tropisetron against acute and delayed symptoms.


Subject(s)
Antiemetics/administration & dosage , Antineoplastic Agents/adverse effects , Dexamethasone/administration & dosage , Indoles/administration & dosage , Metoclopramide/administration & dosage , Nausea/drug therapy , Vomiting/prevention & control , Acute Disease , Adult , Aged , Antineoplastic Agents/therapeutic use , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Nausea/chemically induced , Quality of Life , Treatment Outcome , Tropisetron , Vomiting/chemically induced
19.
J Biol Chem ; 271(45): 28311-7, 1996 Nov 08.
Article in English | MEDLINE | ID: mdl-8910452

ABSTRACT

Voltage-gated K+ (Kv) channels consist of alpha subunits complexed with cytoplasmic Kvbeta subunits. Kvbeta1 subunits enhance the inactivation of currents expressed by the Kv1 alpha subunit subfamily. Binding has been demonstrated between the C terminus of Kvbeta1.1 and a conserved segment of the N terminus of Kv1.4, Kv1.5, and Shaker alpha subunits. Here we have examined the interaction and functional properties of two alternatively spliced human Kvbeta subunits, 1.2 and 1.3, with Kvalpha subunits 1.1, 1.2, 1.4, and 1.5. In the yeast two-hybrid assay, we found that both Kvbeta subunits interact specifically through their conserved C-terminal domains with the N termini of each Kvalpha subunit. In functional experiments, we found differences in modulation of Kv1alpha subunit currents that we attribute to the unique N-terminal domains of the two Kvbeta subunits. Both Kvbeta subunits act as open channel blockers at physiological membrane potentials, but hKvbeta1.2 is a more potent blocker than hKvbeta1.3 of Kv1.1, Kv1.2, Kv1.4, and Kv1. 5. Moreover, hKvbeta1.2 is sensitive to redox conditions, whereas hKvbeta1.3 is not. We suggest that different Kvbeta subunits extend the range over which distinct Kv1alpha subunits are modulated and may provide a variable mechanism for adjusting K+ currents in response to alterations in cellular conditions.


Subject(s)
Alternative Splicing , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Base Sequence , Brain Chemistry , Humans , Kv1.3 Potassium Channel , Molecular Sequence Data , Myocardium/chemistry , Potassium Channels/genetics , Ribonucleases/metabolism
20.
Circulation ; 94(10): 2572-9, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8921803

ABSTRACT

BACKGROUND: The human ether-a-go-go-related gene (HERG) is one locus for the hereditary long-QT syndrome. A hypothesis is that HERG produces the repolarizing cardiac potassium current IKr with the consequence that mutations in HERG prolong the QT interval by reducing IKr. The elementary properties of HERG are unknown, and as a test of the hypothesis that HERG produces IKr, we compared their elementary properties. METHODS AND RESULTS: We injected HERG cRNA into Xenopus oocytes and measured currents from single channels or current variance from the noise produced by ensembles of channels recorded from macro patches. Single-channel conductance was dependent on the extracellular potassium concentration ([K]o). At physiological [K]o, it was 2 picosiemens (pS), and at 100 mmol/L [K]o, it was 10 pS. Openings occurred in bursts with a mean duration of 26 ms at -100 mV. Mean open time was 3.2 ms and closed times were 1.0 and 26 ms. In excised macro patches, HERG currents were blocked by the class III antiarrhythmic drug dofetilide, with an IC50 of 35 nmol/L. Dofetilide block was slow and greatly attenuated at positive potentials at which HERG rectifies. CONCLUSIONS: The microscopic physiology of HERG and IKr is similar, consistent with HERG being an important component of IKr. The pharmacology is also similar; dofetilide appears to primarily block activated channels and has a much lower affinity for closed and inactivated channels.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Ion Channels/antagonists & inhibitors , Ion Channels/physiology , Phenethylamines/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Sulfonamides/pharmacology , Trans-Activators , Animals , Artifacts , ERG1 Potassium Channel , Electric Conductivity , Electrophysiology , Ether-A-Go-Go Potassium Channels , Female , Humans , Molecular Biology/methods , Oocytes , Potassium/physiology , Transcriptional Regulator ERG , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...