Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 725: 150271, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38901222

ABSTRACT

The R-type voltage-gated calcium channel CaV2.3 is predominantly located in the presynapse and is implicated in distinct types of epileptic seizures. It has consequently emerged as a molecular target in seizure treatment. Here, we determined the cryo-EM structure of the CaV2.3-α2δ1-ß1 complex in the topiramate-bound state at a 3.0 Å resolution. We provide a snapshot of the binding site of topiramate, a widely prescribed antiepileptic drug, on a voltage-gated ion channel. The binding site is located at an intracellular juxtamembrane hydrophilic cavity. Further structural analysis revealed that topiramate may allosterically facilitate channel inactivation. These findings provide fundamental insights into the mechanism underlying the inhibitory effect of topiramate on CaV and NaV channels, elucidating a previously unseen modulator binding site and thus pointing toward a route for the development of new drugs.


Subject(s)
Anticonvulsants , Calcium Channels, R-Type , Cryoelectron Microscopy , Topiramate , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Topiramate/chemistry , Topiramate/pharmacology , Humans , Allosteric Regulation/drug effects , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/metabolism , Binding Sites , Models, Molecular , HEK293 Cells , Protein Conformation , Fructose/chemistry , Fructose/analogs & derivatives , Fructose/metabolism , Animals , Cation Transport Proteins
2.
Biochim Biophys Acta Biomembr ; 1862(11): 183439, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32814116

ABSTRACT

Eukaryote voltage-gated Ca2+ channels of the CaV2 channel family are hetero-oligomers formed by the pore-forming CaVα1 protein assembled with auxiliary CaVα2δ and CaVß subunits. CaVß subunits are formed by a Src homology 3 (SH3) domain and a guanylate kinase (GK) domain connected through a HOOK domain. The GK domain binds a conserved cytoplasmic region of the pore-forming CaVα1 subunit referred as the "AID". Herein we explored the phylogenetic and functional relationship between CaV channel subunits in distant eukaryotic organisms by investigating the function of a MAGUK protein (XM_004990081) cloned from the choanoflagellate Salpingoeca rosetta (Sro). This MAGUK protein (Sroß) features SH3 and GK structural domains with a 25% primary sequence identity to mammalian CaVß. Recombinant expression of its cDNA with mammalian high-voltage activated Ca2+ channel CaV2.3 in mammalian HEK cells produced robust voltage-gated inward Ca2+ currents with typical activation and inactivation properties. Like CaVß, Sroß prevents fast degradation of total CaV2.3 proteins in cycloheximide assays. The three-dimensional homology model predicts an interaction between the GK domain of Sroß and the AID motif of the pore-forming CaVα1 protein. Substitution of AID residues Trp (W386A) and Tyr (Y383A) significantly impaired co-immunoprecipitation of CaV2.3 with Sroß and functional upregulation of CaV2.3 currents. Likewise, a 6-residue deletion within the GK domain of Sroß, similar to the locus found in mammalian CaVß, significantly reduced peak current density. Altogether our data demonstrate that an ancestor MAGUK protein reconstitutes the biophysical and molecular features responsible for channel upregulation by mammalian CaVß through a minimally conserved molecular interface.


Subject(s)
Calcium Channels, R-Type/chemistry , Cation Transport Proteins/chemistry , Guanylate Kinases/chemistry , Protozoan Proteins/chemistry , Amino Acid Substitution , Calcium Channels, R-Type/genetics , Calcium Channels, R-Type/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , HEK293 Cells , Humans , Mutation, Missense , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
3.
Channels (Austin) ; 12(1): 326-334, 2018.
Article in English | MEDLINE | ID: mdl-30165790

ABSTRACT

During the recording of whole cell currents from stably transfected HEK-293 cells, the decline of currents carried by the recombinant human Cav2.3+ß3 channel subunits is related to adenosine triphosphate (ATP) depletion after rupture of the cells. It reduces the number of functional channels and leads to a progressive shift of voltage-dependent gating to more negative potentials (Neumaier F., et al., 2018). Both effects can be counteracted by hydrolysable ATP, whose protective action is almost completely prevented by inhibition of serine/threonine but not tyrosine or lipid kinases. These findings indicate that ATP promotes phosphorylation of either the channel or an associated protein, whereas dephosphorylation during cell dialysis results in run-down. Protein phosphorylation is required for Cav2.3 channel function and could directly influence the normal features of current carried by these channels. Therefore, results from in vitro and in vivo phosphorylation of Cav2.3 are summarized to come closer to a functional analysis of structural variations in Cav2.3 splice variants.


Subject(s)
Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/metabolism , Animals , Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Humans , Phosphorylation
4.
Can J Physiol Pharmacol ; 93(4): 291-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25741585

ABSTRACT

The objective of this work was to verify whether, as in the case of the plasma membrane of human vascular smooth muscle cells (hVSMCs), cytosolic ET-1-induced increase of nuclear calcium is mediated via the activation of calcium influx through the steady-state R-type calcium channel. Pharmacological tools to identify the R-type calcium channels, as well as real 3-D confocal microscopy imaging techniques coupled to calcium fluorescent probes, were used to study the effect of cytosolic ET-1 on nuclear calcium in isolated nuclei of human hepatocytes and plasma membrane perforated hVSMCs. Our results showed that pre-treatment with pertussis toxin (PTX) or cholera toxin (CTX) prevented cytosolic ET-1 (10(-9) mol/L) from inducing a sustained increase in nuclear calcium. Furthermore, the L-type calcium channel blocker nifedipine did not prevent cytosolic ET-1 from inducing an increase in nuclear calcium, as opposed to the dual L- and R-type calcium channel blocker isradipine (PN200-110) (in the presence of nifedipine). In conclusion, the preventative effect with PTX and CTX, and the absence of an effect with nifedipine, as well as the blockade by isradipine on cytosolic ET-1-induced increase in nuclear calcium, suggest that this nuclear calcium influx in hVSMCs is due to activation of the steady-state R-type calcium channel. The sarcolemmal and nuclear membrane R-type calcium channels in hVSMCs are involved in ET-1 modulation of vascular tone in physiology and pathology.


Subject(s)
Calcium Channels, R-Type/metabolism , Calcium Signaling , Cation Transport Proteins/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Endothelin-1/metabolism , Muscle, Smooth, Vascular/metabolism , Nuclear Envelope/metabolism , Aorta , Calcium Channel Blockers/pharmacology , Calcium Channels, R-Type/chemistry , Calcium Signaling/drug effects , Cation Transport Proteins/chemistry , Cell Membrane Permeability/drug effects , Cell Nucleus/drug effects , Cells, Cultured , Cholera Toxin/pharmacology , Cytosol/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Imaging, Three-Dimensional , Membrane Fusion/drug effects , Membrane Transport Modulators/pharmacology , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nuclear Envelope/drug effects , Pertussis Toxin/pharmacology , Sarcolemma/drug effects , Sarcolemma/metabolism
5.
Biophys J ; 108(4): 975-985, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25692602

ABSTRACT

We present a simplified reaction network in a single well-mixed volume that captures the general features of CaMKII dynamics observed during both synaptic input and spine depolarization. Our model can also account for the greater-than-control CaMKII activation observed with added EGTA during depolarization. Calcium input currents are modeled after experimental observations, and existing models of calmodulin and CaMKII autophosphorylation are used. After calibration against CaMKII activation data in the absence of chelators, CaMKII activation dynamics due to synaptic input via n-methyl-d-aspartate receptors are qualitatively accounted for in the presence of the chelators EGTA and BAPTA without additional adjustments to the model. To account for CaMKII activation dynamics during spine depolarization with added EGTA or BAPTA, the model invokes the modulation of CaV2.3 (R-type) voltage-dependent calcium channel (VDCC) currents observed in the presence of EGTA or BAPTA. To our knowledge, this is a novel explanation for the increased CaMKII activation seen in dendritic spines with added EGTA, and suggests that differential modulation of VDCCs by EGTA and BAPTA offers an alternative or complementary explanation for other experimental results in which addition of EGTA or BAPTA produces different effects. Our results also show that a simplified reaction network in a single, well-mixed compartment is sufficient to account for the general features of observed CaMKII dynamics.


Subject(s)
Calcium Chelating Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendrites/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Animals , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/metabolism , Calcium Chelating Agents/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Dendrites/drug effects , Egtazic Acid/chemistry , Models, Biological
6.
Rev Physiol Biochem Pharmacol ; 167: 115-39, 2014.
Article in English | MEDLINE | ID: mdl-25280639

ABSTRACT

Voltage-gated Ca(2+) channels (VGCCs) are ubiquitous in excitable cells. These channels play key roles in many physiological events like cardiac regulation/pacemaker activity due to intracellular Ca(2+) transients. In the myocardium, the Cav1 subfamily (L-type: Cav1.2 and Cav1.3) is the main contributor to excitation-contraction coupling and/or pacemaking, whereas the Cav3 subfamily (T-type: Cav3.1 and Cav3.2) is important in rhythmically firing of the cardiac nodal cells. No established cardiac function has been attributed to the Cav2 family (E-/R-type: Cav2.3) despite accumulating evidence of cardiac dysregulation observed upon deletion of the Cav2.3 gene, the only member of this family so far detected in cardiomyocytes. In this review, we summarize the pathophysiological changes observed after ablation of the E-/R-type VGCC and propose a cardiac mechanism of action for this channel. Also, considering the role played by this channel in epilepsy and its reported sensitivity to antiepileptic drugs, a putative involvement of this channel in the cardiac mechanism of sudden unexpected death in epilepsy is also discussed.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Channels, R-Type/physiology , Calcium Channels, T-Type/physiology , Cation Transport Proteins/physiology , Death, Sudden/etiology , Epilepsy/physiopathology , Heart/physiology , Animals , Calcium Channels, L-Type/chemistry , Calcium Channels, R-Type/chemistry , Calcium Channels, T-Type/chemistry , Cation Transport Proteins/chemistry , Epilepsy/complications , Humans
7.
J Neurosci ; 32(39): 13555-67, 2012 Sep 26.
Article in English | MEDLINE | ID: mdl-23015445

ABSTRACT

R-type calcium channels (RTCCs) are well known for their role in synaptic plasticity, but little is known about their subcellular distribution across various neuronal compartments. Using subtype-specific antibodies, we characterized the regional and subcellular localization of Ca(v)2.3 in mice and rats at both light and electron microscopic levels. Ca(v)2.3 immunogold particles were found to be predominantly presynaptic in the interpeduncular nucleus, but postsynaptic in other brain regions. Serial section analysis of electron microscopic images from the hippocampal CA1 revealed a higher density of immunogold particles in the dendritic shaft plasma membrane compared with the pyramidal cell somata. However, the labeling densities were not significantly different among the apical, oblique, or basal dendrites. Immunogold particles were also observed over the plasma membrane of dendritic spines, including both synaptic and extrasynaptic sites. Individual spine heads contained <20 immunogold particles, with an average density of ∼260 immunoparticles per µm(3) spine head volume, in accordance with the density of RTCCs estimated using calcium imaging (Sabatini and Svoboda, 2000). The Ca(v)2.3 density was variable among similar-sized spine heads and did not correlate with the density in the parent dendrite, implying that spines are individual calcium compartments operating autonomously from their parent dendrites.


Subject(s)
Calcium Channels, R-Type/metabolism , Calcium Channels, R-Type/ultrastructure , Cation Transport Proteins/metabolism , Cation Transport Proteins/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Analysis of Variance , Animals , Animals, Newborn , Brain/cytology , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/deficiency , Cation Transport Proteins/chemistry , Cation Transport Proteins/deficiency , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Epitopes/metabolism , Female , Guinea Pigs , Imaging, Three-Dimensional , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Peptides/metabolism , Post-Synaptic Density/metabolism , Post-Synaptic Density/ultrastructure , Rats , Statistics as Topic , Statistics, Nonparametric , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
8.
Biochim Biophys Acta ; 1824(9): 1045-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22633975

ABSTRACT

Ca(v)2.3 containing voltage-activated Ca(2+) channels are expressed in excitable cells and trigger neurotransmitter and peptide-hormone release. Their expression remote from the fast release sites leads to the accumulation of presynaptic Ca(2+) which can both, facilitate and inhibit the influx of Ca(2+) ions through Ca(v)2.3. The facilitated Ca(2+) influx was recently related to hippocampal postsynaptic facilitation and long term potentiation. To analyze Ca(2+) mediated modulation of cellular processes more in detail, protein partners of the carboxy terminal tail of Ca(v)2.3 were identified by yeast-2-hybrid screening, leading in two human cell lines to the detection of a novel, extended and rarely occurring splice variant of calmodulin-2 (CaM-2), called CaM-2-extended (CaM-2-ext). CaM-2-ext interacts biochemically with the C-terminus of Ca(v)2.3 similar to the classical CaM-2 as shown by co-immunoprecipitation. Functionally, only CaM-2-ext reduces whole cell inward currents significantly. The insertion of the novel 46 nts long exon and the consecutive expression of CaM-2-ext must be dependent on a new upstream translation initiation site which is only rarely used in the tested human cell lines. The structure of the N-terminal extension is predicted to be more hydrophobic than the remaining CaM-2-ext protein, suggesting that it may help to dock it to the lipophilic membrane surrounding.


Subject(s)
Alternative Splicing , Calcium Channels, R-Type/metabolism , Calmodulin/metabolism , Cation Transport Proteins/metabolism , Amino Acid Sequence , Base Sequence , Calcium/metabolism , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/genetics , Calmodulin/chemistry , Calmodulin/genetics , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cell Line , HEK293 Cells , Humans , Immunoprecipitation , Molecular Sequence Data
9.
J Gen Physiol ; 139(3): 219-34, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22371363

ABSTRACT

Here, we describe a new mechanism by which glutamate (Glu) and trace metals reciprocally modulate activity of the Ca(v)2.3 channel by profoundly shifting its voltage-dependent gating. We show that zinc and copper, at physiologically relevant concentrations, occupy an extracellular binding site on the surface of Ca(v)2.3 and hold the threshold for activation of these channels in a depolarized voltage range. Abolishing this binding by chelation or the substitution of key amino acid residues in IS1-IS2 (H111) and IS2-IS3 (H179 and H183) loops potentiates Ca(v)2.3 by shifting the voltage dependence of activation toward more negative membrane potentials. We demonstrate that copper regulates the voltage dependence of Ca(v)2.3 by affecting gating charge movements. Thus, in the presence of copper, gating charges transition into the "ON" position slower, delaying activation and reducing the voltage sensitivity of the channel. Overall, our results suggest a new mechanism by which Glu and trace metals transiently modulate voltage-dependent gating of Ca(v)2.3, potentially affecting synaptic transmission and plasticity in the brain.


Subject(s)
Calcium Channels, R-Type/metabolism , Cation Transport Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Biophysical Phenomena , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/genetics , Cation Transport Proteins/agonists , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Copper/pharmacology , Glutamic Acid/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , HEK293 Cells , Humans , In Vitro Techniques , Ion Channel Gating/drug effects , Membrane Potentials , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Rats , Rats, Transgenic , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trace Elements/pharmacology
10.
Trends Endocrinol Metab ; 23(1): 41-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22134088

ABSTRACT

Maintaining blood glucose homeostasis is a complex process that depends on pancreatic islet hormone secretion. Hormone secretion from islets is coupled to calcium entry which results from regenerative islet cell electrical activity. Therefore, the ionic mechanisms that regulate calcium entry into islet cells are crucial for maintaining normal glucose homeostasis. Genome-wide association studies (GWAS) have identified single-nucleotide polymorphisms (SNPs), including five located in or near ion-channel or associated subunit genes, which show an association with human diseases characterized by dysglycemia. This review focuses on polymorphisms and mutations in ion-channel genes that are associated with perturbations in human glucose homeostasis and discusses their potential roles in modulating pancreatic islet hormone secretion.


Subject(s)
Blood Glucose/metabolism , Homeostasis , Insulin/metabolism , Ion Channels/genetics , Islets of Langerhans/metabolism , Animals , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/genetics , Calcium Channels, R-Type/metabolism , Glucose Metabolism Disorders/blood , Glucose Metabolism Disorders/genetics , Glucose Metabolism Disorders/metabolism , Humans , Insulin Secretion , Ion Channels/chemistry , Ion Channels/metabolism , KATP Channels/chemistry , KATP Channels/genetics , KATP Channels/metabolism , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Mutation , Polymorphism, Single Nucleotide , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism
11.
Cell Physiol Biochem ; 28(4): 603-12, 2011.
Article in English | MEDLINE | ID: mdl-22178872

ABSTRACT

BACKGROUND: Voltage gated calcium channels (VGCCs) regulate cellular activity in response to membrane depolarization by altering calcium homeostasis. Because calcium is the most versatile second messenger, regulation of the amount of VGCCs at the plasma membrane is highly critical for several essential cellular processes. Among the different types of VGCCs, the Ca(v)2.3 calcium channel and its regulation mechanisms are least understood due to Ca(v)2.3's resistance to most pharmacological agents. METHODS: In order to study regulation and surface expression of Ca(v)2.3, a yeast two hybrid (Y2H) screen with the II-III loop of human Ca(v)2.3 as bait, was performed. APLP1, a member of the APP gene family and Rab5A, an endocytotic catalyst were identified as putative interaction partners. The interaction were confirmed by immunoprecipitation. To study the functional importance of the interaction, patch-clamp recordings in Ca(v)2.3 stably transfected HEK-293 cells (2C6) and surface biotin endocytosis assays were performed. RESULTS: We are able to show that the II-III loop of the Ca(v)2.3 calcium channel binds APLP1 and that this binding promotes internalization of the channel. In addition, Rab5A also binds to the same loop of the channel and exerts an inhibitory effect on APLP1 mediated channel internalization. CONCLUSIONS: This study identifies a regulation mechanism of Ca(v)2.3's surface expression, which implicates APLP1 as a regulator of calcium homeostasis. Thus APLP1 may play a crucial role in neuropathological mechanisms, which involve modulation of surface expression of voltage-gated Ca(2+) channels.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Calcium Channels, R-Type/metabolism , Cation Transport Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Biotin/genetics , Biotin/metabolism , Calcium Channels, R-Type/chemistry , Cation Transport Proteins/chemistry , Endocytosis , HEK293 Cells , Humans , Immunoprecipitation , Patch-Clamp Techniques , Protein Binding , Protein Structure, Tertiary , Transfection , rab5 GTP-Binding Proteins/genetics
12.
J Biol Chem ; 286(31): 27197-205, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21652722

ABSTRACT

Mutations in distal S6 were shown to significantly alter the stability of the open state of Ca(V)2.3 (Raybaud, A., Baspinar, E. E., Dionne, F., Dodier, Y., Sauvé, R., and Parent, L. (2007) J. Biol. Chem. 282, 27944-27952). By analogy with K(V) channels, we tested the hypothesis that channel activation involves electromechanical coupling between S6 and the S4S5 linker in Ca(V)2.3. Among the 11 positions tested in the S4S5 linker of domain II, mutations of the leucine residue at position 596 were found to destabilize significantly the closed state with a -50 mV shift in the activation potential and a -20 mV shift in its charge-voltage relationship as compared with Ca(V)2.3 wt. A double mutant cycle analysis was performed by introducing pairs of glycine residues between S4S5 and S6 of Domain II. Strong coupling energies (ΔΔG(interact) > 2 kcal mol(-1)) were measured for the activation gating of 12 of 39 pairs of mutants. Leu-596 (IIS4S5) was strongly coupled with distal residues in IIS6 from Leu-699 to Asp-704. In particular, the double mutant L596G/I701G showed strong cooperativity with a ΔΔG(interact) ≈6 kcal mol(-1) suggesting that both positions contribute to the activation gating of the channel. Altogether, our results highlight the role of a leucine residue in S4S5 and provide the first series of evidence that the IIS4S5 and IIS6 regions are energetically coupled during the activation of a voltage-gated Ca(V) channel.


Subject(s)
Calcium Channels, R-Type/metabolism , Cation Transport Proteins/metabolism , Leucine/metabolism , Point Mutation , Amino Acid Sequence , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/genetics , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , DNA, Recombinant/genetics , Humans , Molecular Sequence Data , Patch-Clamp Techniques
13.
Cell Physiol Biochem ; 27(5): 421-32, 2011.
Article in English | MEDLINE | ID: mdl-21691059

ABSTRACT

BACKGROUND: Calcium channels are essential in coupling action potential to signal transduction in cells. There are several types of calcium channels, which can be pharmacologically classified as L-, N-, P/Q-, R- and T-type. But molecular basis of R-type channels is less clearly understood compared the other channel types. Therefore the current study aims at understanding the molecular functions of R-type calcium channels by identifying interaction partners of the channel. METHODS: In order to do so, a yeast two hybrid (Y2H) screen, with carboxy terminus of α1 subunit of the channel, as the bait, was performed. G1 subunit of v-ATPase was identified as a putative interaction partner of human Ca(v)2.3 by using the Y2H screening. The interaction was confirmed by immunoprecipitation. To study the functional importance of the interaction, bafilomycin A(1), a potent and specific inhibitor of v-ATPase was used in patch-clamp recordings in Ca(v)2.3 stably-transfected HEK-293 cells (2C6) as well as in electroretinography of the isolated bovine retina expressing R-type Ca(2+) channels. RESULTS: G1 subunit of v-ATPase interacts with C-terminal tail of Ca(v)2.3 and bafilomycin A(1) reduces Ca(v)2.3 mediated calcium currents. Additionally peak I(Ca) is inhibited in retinal signal transduction when recorded as ERG b-wave. CONCLUSIONS: The results suggest that v-ATPase interacts physically and also functionally with Ca(v)2.3. This is the first demonstration of association of Ca(v)2.3 C-terminus with a protein complex which is involved in transmembrane signalling.


Subject(s)
Adenosine Triphosphatases/metabolism , Calcium Channels, R-Type/metabolism , Calcium/metabolism , Cation Transport Proteins/metabolism , Ion Channel Gating/physiology , Protein Subunits/metabolism , Recombinant Proteins/metabolism , Retina/physiology , Signal Transduction/physiology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Animals , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/genetics , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cattle , Electroretinography , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Immunoprecipitation , Ion Channel Gating/drug effects , Macrolides/pharmacology , Membrane Potentials/physiology , Patch-Clamp Techniques , Plasmids , Protein Subunits/chemistry , Protein Subunits/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Signal Transduction/drug effects , Tissue Culture Techniques , Transfection , Two-Hybrid System Techniques
14.
Pflugers Arch ; 462(2): 303-14, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21611731

ABSTRACT

Extracellular acidification decreases Ca(2+) current amplitude and produces a depolarizing shift in the activation potential (Va) of voltage-gated Ca(2+) channels (VGCC). These effects are common to all VGCC, but differences exist between Ca(2+) channel types and the underlying molecular mechanisms remain largely unknown. We report here that the changes in current amplitude induced by extracellular acidification or alkalinisation are more important for Cav2.3 R type than for Cav2.1 P/Q-type Ca(2+) channels. This difference results from a higher shift of Va combined with a modification of channel conductance. Although involved in the sensitivity of channel conductance to extracellular protons, neither the EEEE locus nor the divalent cation selectivity locus could explain the specificity of the pH effects. We show that this specificity involves two separate sets of amino acids within domain I of the Cavα subunit. Residues of the voltage sensor domain and residues in the pore domain mediate the effects of extracellular protons on Va and on channel conductance, respectively. These new insights are important for elucidating the molecular mechanisms that control VGCC gating and conductance and for understanding the role of extracellular protons in other channels or membrane-tethered enzymes with similar pore and/or voltage sensor domains.


Subject(s)
Amino Acids/metabolism , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/metabolism , Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Ion Channel Gating/physiology , Protons , Amino Acid Sequence , Amino Acids/chemistry , Animals , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Calcium Channels, R-Type/genetics , Cation Transport Proteins/genetics , Molecular Sequence Data , Patch-Clamp Techniques , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Sequence Alignment , Xenopus
15.
Cell Physiol Biochem ; 26(2): 197-208, 2010.
Article in English | MEDLINE | ID: mdl-20798503

ABSTRACT

UNLABELLED: Cell swelling-induced insulin secretion represents an alternative pathway of stimulation of insulin secretion. INS-1E rat tumor beta cells do not release insulin in response to cell swelling in presence of Ca(2+) despite a good response to glucose challenge and appropriate increase in cell volume. Surprisingly, perifusion with Ca(2+)-depleted medium showed distinct secretory response of INS-1E cells to hypotonicity. Objective of this study was further characterization of the role of Ca(2+) in secretory process in INS-1 and INS-1E cell lines. Ca(2+) depleted hypotonic medium with 10 muM BAPTA/AM (intracellular chelator) induced insulin secretion from both types of cells. We demonstrated expression of L-type Ca(2+) channel Ca(v)1.2 and non-L-type Ca(2+) channels Ca(v)2.1 (P/Q-type), Ca(v)2.2 (N-type), and Ca(v)3.1 (T-type) in both cell lines. Inhibition of L type channel with nifedipine and/or P/Q type with omega-agatoxin IVA enabled distinct response to hypotonic medium also in INS-1E cells. Tetanus toxin (TeTx) in medium containing Ca(2+) and a group of calcium channel blockers inhibited hypotonicity-induced insulin secretion from INS-1 cells but not from INS-1E cells. CONCLUSION: Hypotonicity-induced insulin secretion from INS-1E cells is inhibited by extracellular Ca(2+), does not require intracellular Ca(2+) and is TeTx resistant.


Subject(s)
Calcium/pharmacology , Insulin/metabolism , Tetanus Toxin/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Calcium Channels, P-Type/chemistry , Calcium Channels, P-Type/genetics , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/chemistry , Calcium Channels, Q-Type/genetics , Calcium Channels, Q-Type/metabolism , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/genetics , Calcium Channels, R-Type/metabolism , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Cell Line, Tumor , Cell Size , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Hypotonic Solutions/chemistry , Insulin Secretion , Male , Nifedipine/pharmacology , Rats , SNARE Proteins/metabolism , SNARE Proteins/physiology , omega-Agatoxin IVA/pharmacology
16.
Channels (Austin) ; 4(1): 42-50, 2010.
Article in English | MEDLINE | ID: mdl-20026913

ABSTRACT

Calcium entry through voltage-gated calcium channels (VGCC) initiates diverse cellular functions. VGCC pore-forming subunit (Ca(V)alpha(1)) contains four homology repeats, each encompassing a voltage sensor and a pore domain. Three main classes of Ca(V)alpha(1) subunits have been described, Ca(V)1, Ca(V)2 and Ca(V)3 that differ in their voltage-dependence of activation and in the extent in which this process is modulated by the auxiliary beta-subunit (Ca(V)beta). Association of Ca(V)beta induces a coil-to-helix conformation of the I-II intracellular linker joining the first and second repeat of Ca(V)alpha(1) that is thought to be crucial for modulation of channel function. When expressed in Xenopus laevis oocytes in the absence of Ca(V)beta, the voltage to reach 50% activation (V(0.5)) for Ca(V)1.2 and Ca(V)2.3 differs by more than 60 mV and the channel current-carrying capacity by more than thirty-fold. Here we report that the difference in V(0.5) is reduced to about 30 mV and the current-carrying capacity becomes virtually identical when the I-II linkers of Ca(V)1.2 and Ca(V)2.3 are swapped. Co-expression with Ca(V)beta increases the current-carrying capacity of chimeric channels by the same extent, while the difference in V(0.5) with respect to their corresponding parental channels vanishes. Our findings indicate that Ca(V)beta modulatory potency is determined by both, the nature of the I-II linker and the pore-forming subunit background. Moreover, they demonstrate that the I-II linker encodes self-reliant molecular determinants for channel activation and suggest that besides the secondary structure adopted by this segment upon Ca(V)beta association, its chemical nature is as well relevant.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, R-Type/metabolism , Calcium/metabolism , Cation Transport Proteins/metabolism , Ion Channel Gating , Amino Acid Sequence , Animals , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/genetics , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Membrane Potentials , Molecular Sequence Data , Oocytes , Protein Conformation , Protein Structure, Tertiary , Rabbits , Rats , Structure-Activity Relationship , Time Factors , Xenopus laevis
17.
Structure ; 16(4): 607-20, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18400181

ABSTRACT

Calmodulin (CaM) regulation of Ca(2+) channels is central to Ca(2+) signaling. Ca(V)1 versus Ca(V)2 classes of these channels exhibit divergent forms of regulation, potentially relating to customized CaM/IQ interactions among different channels. Here we report the crystal structures for the Ca(2+)/CaM IQ domains of both Ca(V)2.1 and Ca(V)2.3 channels. These highly similar structures emphasize that major CaM contacts with the IQ domain extend well upstream of traditional consensus residues. Surprisingly, upstream mutations strongly diminished Ca(V)2.1 regulation, whereas downstream perturbations had limited effects. Furthermore, our Ca(V)2 structures closely resemble published Ca(2+)/CaM-Ca(V)1.2 IQ structures, arguing against Ca(V)1/2 regulatory differences based solely on contrasting CaM/IQ conformations. Instead, alanine scanning of the Ca(V)2.1 IQ domain, combined with structure-based molecular simulation of corresponding CaM/IQ binding energy perturbations, suggests that the C lobe of CaM partially dislodges from the IQ element during channel regulation, allowing exposed IQ residues to trigger regulation via isoform-specific interactions with alternative channel regions.


Subject(s)
Calcium Channels, N-Type/chemistry , Calcium Channels, R-Type/chemistry , Calcium/chemistry , Calmodulin/chemistry , Models, Molecular , Alanine/genetics , Amino Acid Sequence , Calcium Channels, N-Type/genetics , Crystallography, X-Ray , Molecular Sequence Data , Mutagenesis , Protein Structure, Tertiary , Sequence Homology, Amino Acid
18.
FEBS Lett ; 581(30): 5774-80, 2007 Dec 22.
Article in English | MEDLINE | ID: mdl-18037383

ABSTRACT

We recently reported that a histidine (H191) in the S3-S4 loop of domain I is critical for nickel inhibition of the Cav3.2 T-type Ca2+ channel. As in Cav3.2, two histidine residues are commonly found in the IS3-IS4 loops of mammalian Cav2.3 Ca2+ channels, which are also blocked by low micromolar concentrations of nickel. We show here by site-directed mutagenesis and electrophysiology that both residues contribute to the nickel sensitivity of Cav2.3, with H183 being more critical than H179. These findings strongly suggest that both H179 and H183 in the IS3-IS4 loop are essential structural determinants required for nickel sensitive inhibition of the Cav2.3.


Subject(s)
Calcium Channels, R-Type/chemistry , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/chemistry , Histidine/metabolism , Nickel/pharmacology , Amino Acid Sequence , Animals , Calcium Channels, R-Type/metabolism , Cation Transport Proteins/metabolism , Dose-Response Relationship, Drug , Female , Humans , Ion Channel Gating/drug effects , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Structure, Secondary , Sequence Alignment , Structure-Activity Relationship , Xenopus
19.
J Biol Chem ; 282(38): 27944-52, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17660294

ABSTRACT

The hydrophobic locus VAVIM is conserved in the S6 transmembrane segment of domain IV (IVS6) in Ca(V)1 and Ca(V)2 families. Herein we show that glycine substitution of the VAVIM motif in Ca(V)2.3 produced whole cell currents with inactivation kinetics that were either slower (A1719G approximately V1720G), similar (V1718G), or faster (I1721G approximately M1722G) than the wild-type channel. The fast kinetics of I1721G were observed with a approximately +10 mV shift in its voltage dependence of activation (E(0.5,act)). In contrast, the slow kinetics of A1719G and V1720G were accompanied by a significant shift of approximately -20 mV in their E(0.5,act) indicating that the relative stability of the channel closed state was decreased in these mutants. Glycine scan performed with Val (349) in IS6, Ile(701) in IIS6, and Leu(1420) in IIIS6 at positions predicted to face Val(1720) in IVS6 also produced slow inactivating currents with hyperpolarizing shifts in the activation and inactivation potentials, again pointing out a decrease in the stability of the channel closed state. Mutations to other hydrophobic residues at these positions nearly restored the channel gating. Altogether these data indicate that residues at positions equivalent to 1720 exert a critical control upon the relative stability of the channel closed and open states and more specifically, that hydrophobic residues at these positions promote the channel closed state. We discuss a three-dimensional homology model of Ca(V)2.3 based upon Kv1.2 where hydrophobic residues at positions facing Val(1720) in IS6, IIS6, and IIIS6 play a critical role in stabilizing the closed state in Ca(V)2.3.


Subject(s)
Calcium Channels, R-Type/chemistry , Cation Transport Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , DNA Mutational Analysis , Humans , Kinetics , Kv1.2 Potassium Channel/chemistry , Molecular Conformation , Molecular Sequence Data , Oocytes/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Xenopus
20.
Neuron ; 53(2): 249-60, 2007 Jan 18.
Article in English | MEDLINE | ID: mdl-17224406

ABSTRACT

The roles of voltage-sensitive sodium (Na) and calcium (Ca) channels located on dendrites and spines in regulating synaptic signals are largely unknown. Here we use 2-photon glutamate uncaging to stimulate individual spines while monitoring uncaging-evoked excitatory postsynaptic potentials (uEPSPs) and Ca transients. We find that, in CA1 pyramidal neurons in acute mouse hippocampal slices, CaV(2.3) voltage-sensitive Ca channels (VSCCs) are found selectively on spines and act locally to dampen uncaging-evoked Ca transients and somatic potentials. These effects are mediated by a regulatory loop that requires opening of CaV(2.3) channels, voltage-gated Na channels, small conductance Ca-activated potassium (SK) channels, and NMDA receptors. Ca influx through CaV(2.3) VSCCs selectively activates SK channels, revealing the presence of functional Ca microdomains within the spine. Our results suggest that synaptic strength can be modulated by mechanisms that regulate voltage-gated conductances within the spine but do not alter the properties or numbers of synaptic glutamate receptors.


Subject(s)
Calcium Channels, R-Type/physiology , Cation Transport Proteins/physiology , Dendritic Spines/metabolism , Hippocampus/physiology , Pyramidal Cells/physiology , Synapses/physiology , Animals , Calcium/metabolism , Calcium Channels, R-Type/chemistry , Cation Transport Proteins/chemistry , Electrophysiology , Excitatory Postsynaptic Potentials , Hippocampus/metabolism , In Vitro Techniques , Ion Channel Gating , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Sodium Channels/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...