Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
Add more filters

Publication year range
1.
Nature ; 596(7870): 143-147, 2021 08.
Article in English | MEDLINE | ID: mdl-34234349

ABSTRACT

The neuronal-type (N-type) voltage-gated calcium (Cav) channels, which are designated Cav2.2, have an important role in the release of neurotransmitters1-3. Ziconotide is a Cav2.2-specific peptide pore blocker that has been clinically used for treating intractable pain4-6. Here we present cryo-electron microscopy structures of human Cav2.2 (comprising the core α1 and the ancillary α2δ-1 and ß3 subunits) in the presence or absence of ziconotide. Ziconotide is thoroughly coordinated by helices P1 and P2, which support the selectivity filter, and the extracellular loops (ECLs) in repeats II, III and IV of α1. To accommodate ziconotide, the ECL of repeat III and α2δ-1 have to tilt upward concertedly. Three of the voltage-sensing domains (VSDs) are in a depolarized state, whereas the VSD of repeat II exhibits a down conformation that is stabilized by Cav2-unique intracellular segments and a phosphatidylinositol 4,5-bisphosphate molecule. Our studies reveal the molecular basis for Cav2.2-specific pore blocking by ziconotide and establish the framework for investigating electromechanical coupling in Cav channels.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Cryoelectron Microscopy , omega-Conotoxins/pharmacology , Calcium Channels, N-Type/ultrastructure , Humans , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Protein Conformation/drug effects , Protein Stability/drug effects
2.
Cell ; 133(7): 1228-40, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18585356

ABSTRACT

Calmodulin (CaM) in complex with Ca(2+) channels constitutes a prototype for Ca(2+) sensors that are intimately colocalized with Ca(2+) sources. The C-lobe of CaM senses local, large Ca(2+) oscillations due to Ca(2+) influx from the host channel, and the N-lobe senses global, albeit diminutive Ca(2+) changes arising from distant sources. Though biologically essential, the mechanism underlying global Ca(2+) sensing has remained unknown. Here, we advance a theory of how global selectivity arises, and we experimentally validate this proposal with methodologies enabling millisecond control of Ca(2+) oscillations seen by the CaM/channel complex. We find that global selectivity arises from rapid Ca(2+) release from CaM combined with greater affinity of the channel for Ca(2+)-free versus Ca(2+)-bound CaM. The emergence of complex decoding properties from the juxtaposition of common elements, and the techniques developed herein, promise generalization to numerous molecules residing near Ca(2+) sources.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Calmodulin/metabolism , Animals , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Calcium Signaling , Calmodulin/genetics , Cell Line , Electrophysiology , Humans , Mutagenesis , Point Mutation , Protein Structure, Tertiary , Rats
3.
Mol Cell Neurosci ; 112: 103609, 2021 04.
Article in English | MEDLINE | ID: mdl-33662542

ABSTRACT

Sound information encoding within the initial synapses in the auditory brainstem requires reliable and precise synaptic transmission in response to rapid and large fluctuations in action potential (AP) firing rates. The magnitude and location of Ca2+ entry through voltage-gated Ca2+ channels (CaV) in the presynaptic terminal are key determinants in triggering AP-mediated release. In the mammalian central nervous system (CNS), the CaV2.1 subtype is the critical subtype for CNS function, since it is the most efficient CaV2 subtype in triggering AP-mediated synaptic vesicle (SV) release. Auditory brainstem synapses utilize CaV2.1 to sustain fast and repetitive SV release to encode sound information. Therefore, understanding the presynaptic mechanisms that control CaV2.1 localization, organization and biophysical properties are integral to understanding auditory processing. Here, we review our current knowledge about the control of presynaptic CaV2 abundance and organization in the auditory brainstem and impact on the regulation of auditory processing.


Subject(s)
Brain Stem/physiology , Calcium Channels, N-Type/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Ion Channel Gating/physiology , Nerve Tissue Proteins/physiology , Presynaptic Terminals/physiology , Animals , Auditory Pathways/physiology , Calcium/metabolism , Calcium Channels, N-Type/chemistry , Humans , Ion Transport , Mammals/physiology , Nerve Tissue Proteins/chemistry , Protein Domains , Protein Subunits , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism
4.
J Biol Chem ; 295(52): 18553-18578, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33097592

ABSTRACT

The dominant role of CaV2 voltage-gated calcium channels for driving neurotransmitter release is broadly conserved. Given the overlapping functional properties of CaV2 and CaV1 channels, and less so CaV3 channels, it is unclear why there have not been major shifts toward dependence on other CaV channels for synaptic transmission. Here, we provide a structural and functional profile of the CaV2 channel cloned from the early-diverging animal Trichoplax adhaerens, which lacks a nervous system but possesses single gene homologues for CaV1-CaV3 channels. Remarkably, the highly divergent channel possesses similar features as human CaV2.1 and other CaV2 channels, including high voltage-activated currents that are larger in external Ba2+ than in Ca2+; voltage-dependent kinetics of activation, inactivation, and deactivation; and bimodal recovery from inactivation. Altogether, the functional profile of Trichoplax CaV2 suggests that the core features of presynaptic CaV2 channels were established early during animal evolution, after CaV1 and CaV2 channels emerged via proposed gene duplication from an ancestral CaV1/2 type channel. The Trichoplax channel was relatively insensitive to mammalian CaV2 channel blockers ω-agatoxin-IVA and ω-conotoxin-GVIA and to metal cation blockers Cd2+ and Ni2+ Also absent was the capacity for voltage-dependent G-protein inhibition by co-expressed Trichoplax Gßγ subunits, which nevertheless inhibited the human CaV2.1 channel, suggesting that this modulatory capacity evolved via changes in channel sequence/structure, and not G proteins. Last, the Trichoplax channel was immunolocalized in cells that express an endomorphin-like peptide implicated in cell signaling and locomotive behavior and other likely secretory cells, suggesting contributions to regulated exocytosis.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Calcium Signaling , Calcium/metabolism , Ion Channel Gating , Synaptic Transmission , Amino Acid Sequence , Animals , Cadmium/pharmacology , Nickel/pharmacology , Phylogeny , Placozoa , Sequence Homology, Amino Acid
5.
Int J Mol Sci ; 22(5)2021 Mar 07.
Article in English | MEDLINE | ID: mdl-33799975

ABSTRACT

Migraine is a common neurological disease that affects about 11% of the adult population. The disease is divided into two main clinical subtypes: migraine with aura and migraine without aura. According to the neurovascular theory of migraine, the activation of the trigeminovascular system (TGVS) and the release of numerous neuropeptides, including calcitonin gene-related peptide (CGRP) are involved in headache pathogenesis. TGVS can be activated by cortical spreading depression (CSD), a phenomenon responsible for the aura. The mechanism of CSD, stemming in part from aberrant interactions between neurons and glia have been studied in models of familial hemiplegic migraine (FHM), a rare monogenic form of migraine with aura. The present review focuses on those interactions, especially as seen in FHM type 1, a variant of the disease caused by a mutation in CACNA1A, which encodes the α1A subunit of the P/Q-type voltage-gated calcium channel.


Subject(s)
Calcium Channels/metabolism , Migraine Disorders/etiology , Neuroglia/pathology , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Humans , Migraine Disorders/drug therapy , Migraine Disorders/physiopathology , Mutation , Neuroglia/metabolism
6.
Biochemistry ; 57(44): 6349-6355, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30281282

ABSTRACT

As an ω-conopeptide originally discovered from Conus striatus, SO-3 contains 25 amino acid residues and three disulfide bridges. Our previous study has shown that this peptide possesses potent analgesic activity in rodent pain models (mouse and rat), and it specifically inhibits an N-type calcium ion channel (Cav2.2). In the study presented here, we investigated the key amino acid residues for their inhibitory activity against Cav2.2 expressed in HEK 293 cells and analgesic activity in mice. To improve the inhibitory activity of SO-3, we also evaluated the effects of some amino acid residues derived from the corresponding residues of ω-peptide MVIIA, CVID, or GVIA. Our data reveal that Lys6, Ile11, and Asn14 are the important functional amino acid residues for SO-3. The replacement of some amino acid residues of SO-3 in loop 1 with the corresponding residues of CVID and GVIA improved the inhibitory activity of SO-3. The binding mode of Cav2.2 with SO-3 amino acids in loop 1 and loop 2 may be somewhat different from that of MVIIA. This study expanded our knowledge of the structure-activity relationship of ω-peptides and provided a new strategy for improving the potency of Cav2.2 inhibitors.


Subject(s)
Analgesics/pharmacology , Behavior, Animal/drug effects , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Pain/drug therapy , Peptides/pharmacology , Analgesics/chemistry , Animals , HEK293 Cells , Humans , Mice , Models, Molecular , Pain/metabolism , Peptides/chemistry , Protein Conformation , Rats , Structure-Activity Relationship
7.
Proteins ; 86(4): 414-422, 2018 04.
Article in English | MEDLINE | ID: mdl-29322546

ABSTRACT

CaV channels are transmembrane proteins that mediate and regulate ion fluxes across cell membranes, and they are activated in response to action potentials to allow Ca2+ influx. Since ion channels are composed of charge or polar groups, an external alternating electric field may affect the ion-selective membrane transport and the performance of the channel. In this article, we have investigated the effect of an external GHz electric field on the dynamics of calcium ions in the selectivity filter of the CaV Ab channel. Molecular dynamics (MD) simulations and the potential of mean force (PMF) calculations were carried out, via the umbrella sampling method, to determine the free energy profile of Ca2+ ions in the CaV Ab channels in presence and absence of an external field. Exposing CaV Ab channel to 1, 2, 3, 4, and 5 GHz electric fields increases the depth of the potential energy well and this may result in an increase in the affinity and strength of Ca2+ ions to binding sites in the selectivity filter the channel. This increase of strength of Ca2+ ions binding in the selectivity filter may interrupt the mechanism of Ca2+ ion conduction, and leads to a reduction of Ca2+ ion permeation through the CaV Ab channel.


Subject(s)
Arcobacter/metabolism , Bacterial Proteins/metabolism , Calcium Channels, N-Type/metabolism , Calcium/metabolism , Arcobacter/chemistry , Bacterial Proteins/chemistry , Calcium/chemistry , Calcium Channels, N-Type/chemistry , Cations, Divalent/chemistry , Cations, Divalent/metabolism , Electricity , Ion Transport , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Thermodynamics
8.
Bioorg Med Chem ; 26(11): 3046-3059, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29622412

ABSTRACT

Both N- and T-type calcium ion channels have been implicated in pain transmission and the N-type channel is a well-validated target for the treatment of neuropathic pain. An SAR investigation of a series of substituted aminobenzothiazoles identified a subset of five compounds with comparable activity to the positive control Z160 in a FLIPR-based intracellular calcium response assay measuring potency at both CaV2.2 and CaV3.2 channels. These compounds may form the basis for the development of drug leads and tool compounds for assessing in vivo effects of variable modulation of CaV2.2 and CaV3.2 channels.


Subject(s)
Benzimidazoles/chemical synthesis , Benzothiazoles/chemical synthesis , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/chemistry , Calcium Channels, T-Type/chemistry , Cyclopropanes/chemical synthesis , Naphthalenes/chemical synthesis , Piperidines/chemical synthesis , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/chemistry , Calcium Channels, N-Type/drug effects , Calcium Channels, T-Type/drug effects , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Molecular Structure , Naphthalenes/chemistry , Naphthalenes/pharmacology , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Structure-Activity Relationship
9.
Biochem Biophys Res Commun ; 482(1): 170-175, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27838299

ABSTRACT

To analyze structural features of ω-Aga IVA, a gating modifier toxin from spider venom, we here investigated the NMR solution structure of ω-Aga IVA within DPC micelles. Under those conditions, the Cys-rich central region of ω-Aga IVA still retains the inhibitor Cys knot motif with three short antiparallel ß-strands seen in water. However, 15N HSQC spectra of ω-Aga IVA within micelles revealed that there are radical changes to the toxin's C-terminal tail and several loops upon binding to micelles. The C-terminal tail of ω-Aga IVA appears to assume a ß-turn like conformation within micelles, though it is disordered in water. Whole-cell patch clamp studies with several ω-Aga IVA analogs indicate that both the hydrophobic C-terminal tail and an Arg patch in the core region of ω-Aga IVA are critical for Cav2.1 blockade. These results suggest that the membrane environment stabilizes the structure of the toxin, enabling it to act in a manner similar to other gating modifier toxins, though its mode of interaction with the membrane and the channel is unique.


Subject(s)
Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/ultrastructure , Cell Membrane/chemistry , Lipid Bilayers/chemistry , Purkinje Cells/chemistry , omega-Agatoxin IVA/chemistry , Animals , Binding Sites , Molecular Conformation , Protein Binding , Rats , Rats, Wistar , Structure-Activity Relationship
10.
Nat Rev Neurosci ; 13(8): 542-55, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22805911

ABSTRACT

The voltage-gated calcium channel α(2)δ and ß subunits are traditionally considered to be auxiliary subunits that enhance channel trafficking, increase the expression of functional calcium channels at the plasma membrane and influence the channels' biophysical properties. Accumulating evidence indicates that these subunits may also have roles in the nervous system that are not directly linked to calcium channel function. For example, ß subunits may act as transcriptional regulators, and certain α(2)δ subunits may function in synaptogenesis. The aim of this Review is to examine both the classic and novel roles for these auxiliary subunits in voltage-gated calcium channel function and beyond.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , Animals , Humans , Protein Subunits/chemistry , Protein Subunits/metabolism , Protein Transport/physiology
11.
Arch Biochem Biophys ; 621: 24-30, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28389298

ABSTRACT

Protein kinase C (PKC) isozymes modulate voltage-gated calcium (Cav) currents through Cav2.2 and Cav2.3 channels by targeting serine/threonine (Ser/Thr) phosphorylation sites of Cavα1 subunits. Stimulatory (Thr-422, Ser-2108 and Ser-2132) and inhibitory (Ser-425) sites were identified in the Cav2.2α1 subunits to PKCs ßII and ε. In the current study, we investigated if the homologous sites of Cav2.3α1 subunits (stimulatory: Thr-365, Ser-1995 and Ser-2011; inhibitory: Ser-369) behaved in similar manner. Several Ala and Asp mutants were constructed in Cav2.3α1 subunits in such a way that the Ser/Thr sites can be examined in isolation. These mutants or WT Cav2.3α1 along with auxiliary ß1b and α2/δ subunits were expressed in Xenopus oocytes and the effects of PKCs ßII and ε studied on the barium current (IBa). Among these sites, stimulatory Thr-365 and Ser-1995 and inhibitory Ser-369 behaved similar to their homologs in Cav2.2α1 subunits. Furthermore PKCs produced neither stimulation nor inhibition when stimulatory Thr-365 or Ser-1995 and inhibitory Ser-369 were present together. However, the PKCs potentiated the IBa when two stimulatory sites, Thr-365 and Ser-1995 were present together, thus overcoming the inhibitory effect of Ser-369. Taken together net PKC effect may be the difference between the responses of the stimulatory and inhibitory sites.


Subject(s)
Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Membrane Potentials/physiology , Oocytes/physiology , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Animals , Binding Sites , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Mutagenesis, Site-Directed , Protein Binding , Protein Subunits , Serine/chemistry , Serine/metabolism , Structure-Activity Relationship , Substrate Specificity , Threonine/chemistry , Threonine/metabolism , Xenopus laevis
12.
FASEB J ; 29(5): 1794-804, 2015 May.
Article in English | MEDLINE | ID: mdl-25563298

ABSTRACT

Rem, Rad, Kir/Gem (RGK) proteins, including Rem2, mediate profound inhibition of high-voltage activated Ca(2+) channels containing intracellular regulatory ß subunits. All RGK proteins bind to voltage-gated Ca(2+) channel ß subunit (Cavß) subunits in vitro, but the necessity of the interaction for current inhibition remains controversial. This study applies NMR and calorimetric techniques to map the binding site for Rem2 on human Cavß4a and measure its binding affinity. Our experiments revealed 2 binding surfaces on the ß4 guanylate kinase domain contributing to a 156 ± 18 µM Kd interaction: a hydrophobic pocket lined by 4 critical residues (L173, N261, H262, and V303), mutation of any of which completely disrupted binding, and a nearby surface containing 3 residues (D206, L209, and D258) that when individually mutated decreased affinity. Voltage-gated Ca(2+) channel α1A subunit (Cav2.1) Ca(2+) currents were completely inhibited by Rem2 when co-expressed with wild-type Cavß4a, but were unaffected by Rem2 when coexpressed with a Cavß4a site 1 (L173A/V303A) or site 2 (D258A) mutant. These results provide direct evidence for a low-affinity Rem2/Cavß4 interaction and show definitively that the interaction is required for Cav2.1 inhibition.


Subject(s)
Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Calorimetry/methods , Ion Channels/physiology , Magnetic Resonance Spectroscopy/methods , Monomeric GTP-Binding Proteins/metabolism , Calcium Channels, N-Type/genetics , Electrophysiology , HEK293 Cells , Humans , Protein Conformation , Protein Interaction Domains and Motifs
13.
J Biol Chem ; 289(8): 5285-95, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24394417

ABSTRACT

N-type and P/Q-type calcium channels are documented players in the regulation of synaptic function; however, the mechanisms underlying their expression and cellular targeting are poorly understood. Ankyrin polypeptides are essential for normal integral membrane protein expression in a number of cell types, including neurons, cardiomyocytes, epithelia, secretory cells, and erythrocytes. Ankyrin dysfunction has been linked to defects in integral protein expression, abnormal cellular function, and disease. Here, we demonstrate that ankyrin-B associates with Cav2.1 and Cav2.2 in cortex, cerebellum, and brain stem. Additionally, using in vitro and in vivo techniques, we demonstrate that ankyrin-B, via its membrane-binding domain, associates with a highly conserved motif in the DII/III loop domain of Cav2.1 and Cav2.2. Further, we demonstrate that this domain is necessary for proper targeting of Cav2.1 and Cav2.2 in a heterologous system. Finally, we demonstrate that mutation of a single conserved tyrosine residue in the ankyrin-binding motif of both Cav2.1 (Y797E) and Cav2.2 (Y788E) results in loss of association with ankyrin-B in vitro and in vivo. Collectively, our findings identify an interaction between ankyrin-B and both Cav2.1 and Cav2.2 at the amino acid level that is necessary for proper Cav2.1 and Cav2.2 targeting in vivo.


Subject(s)
Ankyrins/metabolism , Calcium Channels, N-Type/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Brain , Calcium Channels, N-Type/chemistry , Conserved Sequence , HEK293 Cells , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Protein Binding , Purkinje Cells/metabolism , Rats , Tyrosine/metabolism
14.
Bioorg Med Chem ; 23(18): 6166-72, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26296911

ABSTRACT

T- and N-type calcium channels have known for relating to therapy of neuropathic pain which is chronic, debilitating pain state. Neuropathic pain is caused by damage of the somatosensory system. It may be associated with abnormal sensations and pain produced by normally non-painful stimuli (allodynia). Neuropathic pain is very difficult to treat, and only some 40-60% of patients achieve partial relief. For a neuropathic pain therapy, anticonvulsant like Lamotrigine, Carbamazepine and a topical anesthetic such as Lidocaine are used. We synthesized 15 novel amine derivatives and evaluated their activities against T-type and N-type calcium channels by whole-cell patch clamp recording on HEK293 cells. Among the tested compounds, compound 10 showed good inhibitory activity for both T-type and N-type calcium channels with the IC50 value of 1.9 µM and 4.3 µM, respectively. Compound 10 also showed good analgesic activity on rat spinal cord injury model.


Subject(s)
Amines/chemistry , Calcium Channel Blockers/chemistry , Amines/pharmacology , Amines/therapeutic use , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Male , Motor Activity/drug effects , Neuralgia/drug therapy , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy , Structure-Activity Relationship
15.
Mol Cell Neurosci ; 63: 124-31, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25447945

ABSTRACT

Facilitation and inactivation of P/Q-type Ca2+ currents mediated by Ca2+/calmodulin binding to Ca(V)2.1 channels contribute to facilitation and rapid depression of synaptic transmission, respectively. Other calcium sensor proteins displace calmodulin from its binding site and differentially modulate P/Q-type Ca2 + currents, resulting in diverse patterns of short-term synaptic plasticity. Neuronal calcium sensor-1 (NCS-1, frequenin) has been shown to enhance synaptic facilitation, but the underlying mechanism is unclear. We report here that NCS-1 directly interacts with IQ-like motif and calmodulin-binding domain in the C-terminal domain of Ca(V)2.1 channel. NCS-1 reduces Ca2 +-dependent inactivation of P/Q-type Ca2+ current through interaction with the IQ-like motif and calmodulin-binding domain without affecting peak current or activation kinetics. Expression of NCS-1 in presynaptic superior cervical ganglion neurons has no effect on synaptic transmission, eliminating effects of this calcium sensor protein on endogenous N-type Ca2+ currents and the endogenous neurotransmitter release machinery. However, in superior cervical ganglion neurons expressing wild-type Ca(V)2.1 channels, co-expression of NCS-1 induces facilitation of synaptic transmission in response to paired pulses and trains of depolarizing stimuli, and this effect is lost in Ca(V)2.1 channels with mutations in the IQ-like motif and calmodulin-binding domain. These results reveal that NCS-1 directly modulates Ca(V)2.1 channels to induce short-term synaptic facilitation and further demonstrate that CaS proteins are crucial in fine-tuning short-term synaptic plasticity.


Subject(s)
Calcium Channels, N-Type/metabolism , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/metabolism , Synapses/metabolism , Synaptic Transmission , Amino Acid Motifs , Animals , Binding Sites , Calcium Channels, N-Type/chemistry , Cells, Cultured , HEK293 Cells , Humans , Mice , Neuronal Calcium-Sensor Proteins/genetics , Neuropeptides/genetics , Protein Binding , Rats , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/physiology , Synapses/physiology
16.
Mar Drugs ; 13(4): 2030-45, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25871286

ABSTRACT

A set of fluorophenoxyanilides, designed to be simplified analogues of previously reported ω-conotoxin GVIA mimetics, were prepared and tested for N-type calcium channel inhibition in a SH-SY5Y neuroblastoma FLIPR assay. N-type or Cav2.2 channel is a validated target for the treatment of refractory chronic pain. Despite being significantly less complex than the originally designed mimetics, up to a seven-fold improvement in activity was observed.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Anilides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Drug Design , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/drug effects , Analgesics, Non-Narcotic/chemical synthesis , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/metabolism , Anilides/chemical synthesis , Anilides/chemistry , Anilides/metabolism , Binding, Competitive , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/metabolism , Calcium Channels, N-Type/chemistry , Calcium Signaling/drug effects , Cell Line, Tumor , Fluorobenzenes/chemical synthesis , Fluorobenzenes/chemistry , Fluorobenzenes/metabolism , Fluorobenzenes/pharmacology , High-Throughput Screening Assays , Humans , Molecular Structure , Molecular Targeted Therapy , Nerve Tissue Proteins/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Neurons/metabolism , Neurotoxins/chemistry , Pain, Intractable/drug therapy , Pain, Intractable/metabolism , Structure-Activity Relationship , omega-Conotoxin GVIA/chemistry , omega-Conotoxin GVIA/metabolism , omega-Conotoxin GVIA/pharmacology
18.
Biophys J ; 107(12): 2797-2814, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25517147

ABSTRACT

BKCa-channel activity often affects the firing properties of neurons, the shapes of neuronal action potentials (APs), and in some cases the extent of neurotransmitter release. It has become clear that BKCa channels often form complexes with voltage-gated Ca(2+) channels (CaV channels) such that when a CaV channel is activated, the ensuing influx of Ca(2+) activates its closely associated BKCa channel. Thus, in modeling the electrical properties of neurons, it would be useful to have quantitative models of CaV/BKCa complexes. Furthermore, in a population of CaV/BKCa complexes, all BKCa channels are not exposed to the same Ca(2+) concentration at the same time. Thus, stochastic rather than deterministic models are required. To date, however, no such models have been described. Here, however, I present a stochastic model of a CaV2.1/BKCa(α-only) complex, as might be found in a central nerve terminal. The CaV2.1/BKCa model is based on kinetic modeling of its two component channels at physiological temperature. Surprisingly, The CaV2.1/BKCa model predicts that although the CaV channel will open nearly every time during a typical cortical AP, its associated BKCa channel is expected to open in only 30% of trials, and this percentage is very sensitive to the duration of the AP, the distance between the two channels in the complex, and the presence of fast internal Ca(2+) buffers. Also, the model predicts that the kinetics of the BKCa currents of a population of CaV2.1/BKCa complexes will not be limited by the kinetics of the CaV2.1 channel, and during a train of APs, the current response of the complex is expected to faithfully follow even very rapid trains. Aside from providing insight into how these complexes are likely to behave in vivo, the models presented here could also be of use more generally as components of higher-level models of neural function.


Subject(s)
Calcium Channels, N-Type/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Protein Multimerization , Animals , Calcium Channels, N-Type/chemistry , Humans , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Mice , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats
19.
Biochemistry ; 53(38): 6052-62, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25188201

ABSTRACT

In neurons, entry of extracellular calcium (Ca(2+)) into synaptic terminals through Cav2.1 (P/Q-type) Ca(2+) channels is the driving force for exocytosis of neurotransmitter-containing synaptic vesicles. This class of Ca(2+) channel is, therefore, pivotal during normal neurotransmission in higher organisms. In response to channel opening and Ca(2+) influx, specific Ca(2+)-binding proteins associate with cytoplasmic regulatory domains of the P/Q channel to modulate subsequent channel opening. Channel modulation in this way influences synaptic plasticity with consequences for higher-level processes such as learning and memory acquisition. The ubiquitous Ca(2+)-sensing protein calmodulin (CaM) regulates the activity of all types of mammalian voltage-gated Ca(2+) channels, including the P/Q class, by direct binding to specific regulatory motifs. More recently, experimental evidence has highlighted a role for additional Ca(2+)-binding proteins, particularly of the CaBP and NCS families in the regulation of P/Q channels. NCS-1 is a protein found from yeast to humans and that regulates a diverse number of cellular functions. Physiological and genetic evidence indicates that NCS-1 regulates P/Q channel activity, including calcium-dependent facilitation, although a direct physical association between the proteins has yet to be demonstrated. In this study, we aimed to determine if there is a direct interaction between NCS-1 and the C-terminal cytoplasmic tail of the Cav2.1 α-subunit. Using distinct but complementary approaches, including in vitro binding of bacterially expressed recombinant proteins, fluorescence spectrophotometry, isothermal titration calorimetry, nuclear magnetic resonance, and expression of fluorescently tagged proteins in mammalian cells, we show direct binding and demonstrate that CaM can compete for it. We speculate about how NCS-1/Cav2.1 association might add to the complexity of calcium channel regulation mediated by other known calcium-sensing proteins and how this might help to fine-tune neurotransmission in the mammalian central nervous system.


Subject(s)
Calcium Channels, N-Type/metabolism , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/metabolism , Calcium/metabolism , Calcium Channels, N-Type/chemistry , Cloning, Molecular , Humans , Neuronal Calcium-Sensor Proteins/chemistry , Neuropeptides/chemistry , Protein Binding
20.
Mol Pharmacol ; 86(6): 760-72, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25267719

ABSTRACT

4-Aminopyridine (4-AP, fampridine) is used clinically to improve neuromuscular function in patients with multiple sclerosis, spinal cord injury, and myasthenia gravis. 4-AP can increase neuromuscular and synaptic transmission by directly stimulating high voltage-activated (HVA) Ca(2+) channels independent of its blocking effect on voltage-activated K(+) channels. Here we provide new evidence that the potentiating effect of 4-AP on HVA Ca(2+) channels depends on the specific combination of voltage-activated calcium channel α1 (Cavα1) and voltage-activated calcium channel ß (Cavß) subunits. Among the four Cavß subunits examined, Cavß3 was the most significant subunit involved in the 4-AP-induced potentiation of both L-type and N-type currents. Of particular note, 4-AP at micromolar concentrations selectively potentiated L-type currents reconstituted with Cav1.2, α2δ1, and Cavß3. In contrast, 4-AP potentiated N-type currents only at much higher concentrations and had little effect on P/Q-type currents. In a phrenic nerve-diaphragm preparation, blocking L-type Ca(2+) channels eliminated the potentiating effect of low concentrations of 4-AP on end-plate potentials. Furthermore, 4-AP enhanced the physical interaction of Cav1.2 and Cav2.2 subunits to Cavß3 and also increased their trafficking to the plasma membrane. Site-directed mutagenesis identified specific regions in the guanylate kinase, HOOK, and C-terminus domains of the Cavß3 subunit crucial to the ability of 4-AP to potentiate L-type and N-type currents. Our findings indicate that 4-AP potentiates HVA Ca(2+) channels by enhancing reciprocal Cav1.2-Cavß3 and Cav2.2-Cavß3 interactions. The therapeutic effect of 4-AP on neuromuscular function is probably mediated by its actions on Cavß3-containing L-type Ca(2+) channels.


Subject(s)
4-Aminopyridine/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, N-Type/drug effects , Amino Acid Sequence , Calcium Channels, L-Type/chemistry , Calcium Channels, N-Type/chemistry , HEK293 Cells , Humans , Molecular Sequence Data , Neuromuscular Junction/physiology , Protein Subunits , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL