Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Physiol ; 578(Pt 1): 159-71, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17082234

ABSTRACT

Zinc (Zn2+) functions as a signalling molecule in the nervous system and modulates many ionic channels. In this study, we have explored the effects of Zn2+ on recombinant T-type calcium channels (CaV3.1, CaV3.2 and CaV3.3). Using tsA-201 cells, we demonstrate that CaV3.2 current (IC50, 0.8 microm) is significantly more sensitive to Zn2+ than are CaV3.1 and CaV3.3 currents (IC50, 80 microm and approximately 160 microm, respectively). This inhibition of CaV3 currents is associated with a shift to more negative membrane potentials of both steady-state inactivation for CaV3.1, CaV3.2 and CaV3.3 and steady-state activation for CaV3.1 and CaV3.3 currents. We also document changes in kinetics, especially a significant slowing of the inactivation kinetics for CaV3.1 and CaV3.3, but not for CaV3.2 currents. Notably, deactivation kinetics are significantly slowed for CaV3.3 current (approximately 100-fold), but not for CaV3.1 and CaV3.2 currents. Consequently, application of Zn2+ results in a significant increase in CaV3.3 current in action potential clamp experiments, while CaV3.1 and CaV3.2 currents are significantly reduced. In neuroblastoma NG 108-15 cells, the duration of CaV3.3-mediated action potentials is increased upon Zn2+ application, indicating further that Zn2+ behaves as a CaV3.3 channel opener. These results demonstrate that Zn2+ exhibits differential modulatory effects on T-type calcium channels, which may partly explain the complex features of Zn2+ modulation of the neuronal excitability in normal and disease states.


Subject(s)
Calcium Channel Blockers , Calcium Channels, T-Type/drug effects , Zinc/pharmacology , Action Potentials/drug effects , Algorithms , Calcium Channels, T-Type/genetics , Cell Line , Cell Line, Tumor , Electrophysiology , Humans , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/genetics , Patch-Clamp Techniques , Recombinant Proteins , Synaptic Transmission/drug effects , Thalamus/drug effects , Thalamus/physiology , Transfection
2.
Cell Calcium ; 40(2): 121-34, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16797700

ABSTRACT

Low-voltage-activated T-type calcium channels are expressed in various tissues, especially in the brain, where they promote neuronal firing and are involved in slow wave sleep and absence epilepsy. While the transduction pathways by which hormones and neurotransmitters modulate high-voltage-activated calcium channels are beginning to be unraveled, those implicated in T-type calcium channel regulation remain obscure. Several neurotransmitters and hormones regulate native T-type calcium channels, although some contradictory data have been reported depending on the cell type studied. This review focuses on the short-term (minutes range) modulation of T-type calcium channels by neurotransmitters and hormones and on the roles of G proteins and protein kinases in these modulatory effects. Results obtained in different native tissues are discussed and compared with the more recent studies of the three cloned T-type calcium channels CaV3.1, CaV3.2 and CaV3.3 in expression systems.


Subject(s)
Calcium Channels, T-Type/metabolism , Hormones/metabolism , Neurotransmitter Agents/metabolism , Animals , Humans , Ion Channel Gating , Protein Kinases/metabolism
3.
Circ Res ; 98(11): 1422-30, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16690884

ABSTRACT

The generation of the mammalian heartbeat is a complex and vital function requiring multiple and coordinated ionic channel activities. The functional role of low-voltage activated (LVA) T-type calcium channels in the pacemaker activity of the sinoatrial node (SAN) is, to date, unresolved. Here we show that disruption of the gene coding for CaV3.1/alpha1G T-type calcium channels (cacna1g) abolishes T-type calcium current (I(Ca,T)) in isolated cells from the SAN and the atrioventricular node without affecting the L-type Ca2+ current (I(Ca,L)). By using telemetric electrocardiograms on unrestrained mice and intracardiac recordings, we find that cacna1g inactivation causes bradycardia and delays atrioventricular conduction without affecting the excitability of the right atrium. Consistently, no I(Ca,T) was detected in right atrium myocytes in both wild-type and CaV3.1(-/-) mice. Furthermore, inactivation of cacna1g significantly slowed the intrinsic in vivo heart rate, prolonged the SAN recovery time, and slowed pacemaker activity of individual SAN cells through a reduction of the slope of the diastolic depolarization. Our results demonstrate that CaV3.1/T-type Ca2+ channels contribute to SAN pacemaker activity and atrioventricular conduction.


Subject(s)
Atrioventricular Node/physiopathology , Bradycardia/etiology , Bradycardia/physiopathology , Calcium Channels, T-Type/deficiency , Animals , Atrioventricular Node/metabolism , Atrioventricular Node/pathology , Bradycardia/metabolism , Bradycardia/pathology , Electric Conductivity , Electrocardiography , Electrophysiology , Heart Rate , Hypnotics and Sedatives/pharmacology , Mice , Mice, Knockout , Protein Isoforms/deficiency , Sinoatrial Node/physiopathology
4.
Mol Pharmacol ; 69(6): 1963-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16510561

ABSTRACT

Fluoxetine, a widely used antidepressant that primarily acts as a selective serotonin reuptake inhibitor, also inhibits various neuronal ion channels. Using the whole-cell patch-clamp technique, we have examined the effects of fluoxetine and norfluoxetine, its major active metabolite, on cloned low-voltage-activated T-type calcium channels (T channels) expressed in tsA 201 cells. Fluoxetine inhibited the three T channels Ca(V)3.1, Ca(V)3.2, and Ca(V)3.3 in a concentration-dependent manner (IC(50) = 14, 16, and 30 microM, respectively). Norfluoxetine was a more potent inhibitor than fluoxetine, especially on the Ca(V)3.3 T current (IC(50) = 5 microM). The fluoxetine block of T channels was voltage-dependent because it was significantly enhanced for T channels in the inactivated state. Fluoxetine caused a hyperpolarizing shift in steady-state inactivation, with a slower rate of recovery from the inactivated state. These results indicated a tighter binding of fluoxetine to the inactivated state than to the resting state of T channels, suggesting a more potent inhibition of T channels at physiological resting membrane potential. Indeed, fluoxetine and norfluoxetine at 1 microM strongly inhibited cloned T currents (approximately 50 and approximately 75%, respectively) in action potential clamp experiments performed with firing activities of thalamocortical relay neurons. Altogether, these data demonstrate that clinically relevant concentrations of fluoxetine exert a voltage-dependent block of T channels that may contribute to this antidepressant's pharmacological effects.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , Cells, Cultured , Fluoxetine/metabolism , Humans , Inhibitory Concentration 50 , Membrane Transport Proteins/drug effects , Recombinant Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...