140 results on '"Calcium Channels, N-Type chemistry"'
Search Results
2. Comparison of quinazoline and benzoylpyrazoline chemotypes targeting the CaVα-β interaction as antagonists of the N-type CaV2.2 channel.
- Author
-
Ran D, Gomez K, Moutal A, Patek M, Perez-Miller S, and Khanna R
- Subjects
- Animals, Rats, Humans, Calcium Channel Blockers pharmacology, Calcium Channel Blockers chemistry, Pyrazoles pharmacology, Pyrazoles chemistry, Calcium Channels, N-Type metabolism, Calcium Channels, N-Type chemistry, Quinazolines pharmacology, Quinazolines chemistry
- Abstract
Structural studies with an α subunit fragment of voltage-gated calcium (CaV) channels in complex with the CaVβ subunits revealed a high homology between the various CaVα-β subunits, predicting that targeting of this interface would result in nonselective compounds. Despite this likelihood, my laboratory initiated a rational structure-based screening campaign focusing on "hot spots" on the alpha interacting domain (AID) of the CaVβ2a subunits and identified the small molecule 2-(3,5-dimethylisoxazol-4-yl)-N-((4-((3-phenylpropyl)amino)quinazolin-2-yl)methyl)acetamide ( IPPQ ) which selectively targeted the interface between the N-type calcium (CaV2.2) channel and CaVβ. IPPQ (i) specifically bound to CaVβ2a; (ii) inhibited CaVβ2 's interaction with CaV.2-AID; (iii) inhibited CaV2.2 currents in sensory neurons; (iv) inhibited pre-synaptic localization of CaV2.2 in vivo ; and (v) inhibited spinal neurotransmission, which resulted in decreased neurotransmitter release. IPPQ was anti-nociceptive in naïve rats and reversed mechanical allodynia and thermal hyperalgesia in rodent models of acute, neuropathic, and genetic pain. In structure-activity relationship (SAR) studies focused on improving binding affinity of IPPQ , another compound (BTT-369), a benzoyl-3,4-dihydro-1'H,2 H-3,4'-bipyrazole class of compounds, was reported by Chen and colleagues, based on work conducted in my laboratory beginning in 2008. BTT-369 contains tetraaryldihydrobipyrazole scaffold - a chemotype featuring phenyl groups known to be significantly metabolized, lower the systemic half-life, and increase the potential for toxicity. Furthermore, the benzoylpyrazoline skeleton in BTT-369 is patented across multiple therapeutic indications. Prior to embarking on an extensive optimization campaign of IPPQ , we performed a head-to-head comparison of the two compounds. We conclude that IPPQ is superior to BTT-369 for on-target efficacy, setting the stage for SAR studies to improve on IPPQ for the development of novel pain therapeutics.
- Published
- 2021
- Full Text
- View/download PDF
3. Structure of human Ca v 2.2 channel blocked by the painkiller ziconotide.
- Author
-
Gao S, Yao X, and Yan N
- Subjects
- 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, Analgesics, Non-Narcotic pharmacology, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Cryoelectron Microscopy, omega-Conotoxins pharmacology
- Abstract
The neuronal-type (N-type) voltage-gated calcium (Ca
v ) channels, which are designated Cav 2.2, have an important role in the release of neurotransmitters1-3 . Ziconotide is a Cav 2.2-specific peptide pore blocker that has been clinically used for treating intractable pain4-6 . Here we present cryo-electron microscopy structures of human Cav 2.2 (comprising the core α1 and the ancillary α2δ-1 and β3 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 Cav 2-unique intracellular segments and a phosphatidylinositol 4,5-bisphosphate molecule. Our studies reveal the molecular basis for Cav 2.2-specific pore blocking by ziconotide and establish the framework for investigating electromechanical coupling in Cav channels., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
4. Presynaptic voltage-gated calcium channels in the auditory brainstem.
- Author
-
Young SM Jr and Veeraraghavan P
- Subjects
- 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, 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
- 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 Ca
2+ 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 CaV 2.1 subtype is the critical subtype for CNS function, since it is the most efficient CaV 2 subtype in triggering AP-mediated synaptic vesicle (SV) release. Auditory brainstem synapses utilize CaV 2.1 to sustain fast and repetitive SV release to encode sound information. Therefore, understanding the presynaptic mechanisms that control CaV 2.1 localization, organization and biophysical properties are integral to understanding auditory processing. Here, we review our current knowledge about the control of presynaptic CaV 2 abundance and organization in the auditory brainstem and impact on the regulation of auditory processing., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
5. Migraine: Calcium Channels and Glia.
- Author
-
Kowalska M, Prendecki M, Piekut T, Kozubski W, and Dorszewska J
- Subjects
- 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, Calcium Channels metabolism, Migraine Disorders etiology, Neuroglia pathology
- 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.
- Published
- 2021
- Full Text
- View/download PDF
6. The de novo CACNA1A pathogenic variant Y1384C associated with hemiplegic migraine, early onset cerebellar atrophy and developmental delay leads to a loss of Cav2.1 channel function.
- Author
-
Gandini MA, Souza IA, Ferron L, Innes AM, and Zamponi GW
- Subjects
- Adolescent, Adult, Alternative Splicing genetics, Atrophy, Biophysical Phenomena, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Cell Line, Child, Preschool, Developmental Disabilities complications, Female, Humans, Infant, Newborn, Ion Channel Gating, Male, Migraine with Aura complications, Models, Molecular, Mutant Proteins chemistry, Mutant Proteins metabolism, Protein Isoforms genetics, Protein Structure, Secondary, Structural Homology, Protein, Calcium Channels, N-Type genetics, Cerebellum pathology, Developmental Disabilities genetics, Genetic Predisposition to Disease, Migraine with Aura genetics, Mutation genetics
- Abstract
CACNA1A pathogenic variants have been linked to several neurological disorders including familial hemiplegic migraine and cerebellar conditions. More recently, de novo variants have been associated with severe early onset developmental encephalopathies. CACNA1A is highly expressed in the central nervous system and encodes the pore-forming Ca
V α1 subunit of P/Q-type (Cav2.1) calcium channels. We have previously identified a patient with a de novo missense mutation in CACNA1A (p.Y1384C), characterized by hemiplegic migraine, cerebellar atrophy and developmental delay. The mutation is located at the transmembrane S5 segment of the third domain. Functional analysis in two predominant splice variants of the neuronal Cav2.1 channel showed a significant loss of function in current density and changes in gating properties. Moreover, Y1384 variants exhibit differential splice variant-specific effects on recovery from inactivation. Finally, structural analysis revealed structural damage caused by the tyrosine substitution and changes in electrostatic potentials.- Published
- 2021
- Full Text
- View/download PDF
7. Conserved biophysical features of the Ca V 2 presynaptic Ca 2+ channel homologue from the early-diverging animal Trichoplax adhaerens .
- Author
-
Gauberg J, Abdallah S, Elkhatib W, Harracksingh AN, Piekut T, Stanley EF, and Senatore A
- Subjects
- Amino Acid Sequence, Animals, Cadmium pharmacology, Nickel pharmacology, Phylogeny, Placozoa, Sequence Homology, Amino Acid, Calcium metabolism, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Calcium Signaling, Ion Channel Gating, Synaptic Transmission
- Abstract
The dominant role of Ca
V 2 voltage-gated calcium channels for driving neurotransmitter release is broadly conserved. Given the overlapping functional properties of CaV 2 and CaV 1 channels, and less so CaV 3 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 CaV 2 channel cloned from the early-diverging animal Trichoplax adhaerens , which lacks a nervous system but possesses single gene homologues for CaV 1-CaV 3 channels. Remarkably, the highly divergent channel possesses similar features as human CaV 2.1 and other CaV 2 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 CaV 2 suggests that the core features of presynaptic CaV 2 channels were established early during animal evolution, after CaV 1 and CaV 2 channels emerged via proposed gene duplication from an ancestral CaV 1/2 type channel. The Trichoplax channel was relatively insensitive to mammalian CaV 2 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 Gβγ subunits, which nevertheless inhibited the human CaV 2.1 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., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Gauberg et al.)- Published
- 2020
- Full Text
- View/download PDF
8. Expression and functions of N-type Cav2.2 and T-type Cav3.1 channels in rat vasopressin neurons under normotonic conditions.
- Author
-
Sato-Numata K, Numata T, Ueta Y, and Okada Y
- Subjects
- Action Potentials, Animals, Animals, Genetically Modified, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, Calcium Channels, T-Type chemistry, Calcium Channels, T-Type genetics, Calcium Signaling, Male, Rats, Rats, Wistar, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type metabolism, Calcium Channels, T-Type metabolism, Neurons metabolism, Vasopressins metabolism
- Abstract
Arginine vasopressin (AVP) neurons play essential roles in sensing the change in systemic osmolarity and regulating AVP release from their neuronal terminals to maintain the plasma osmolarity. AVP exocytosis depends on the Ca
2+ entry via voltage-gated Ca2+ channels (VGCCs) in AVP neurons. In this study, suppression by siRNA-mediated knockdown and pharmacological sensitivity of VGCC currents evidenced molecular and functional expression of N-type Cav2.2 and T-type Cav3.1 in AVP neurons under normotonic conditions. Also, both the Cav2.2 and Cav3.1 currents were found to be sensitive to flufenamic acid (FFA). TTX-insensitive spontaneous action potentials were suppressed by FFA and T-type VGCC blocker Ni2+ . However, Cav2.2-selective ω-conotoxin GVIA failed to suppress the firing activity. Taken together, it is concluded that Cav2.2 and Cav3.1 are molecularly and functionally expressed and both are sensitive to FFA in unstimulated rat AVP neurons. Also, it is suggested that Cav3.1 is primarily involved in their action potential generation.- Published
- 2020
- Full Text
- View/download PDF
9. Tissue- and cell-specific expression of a splice variant in the II-III cytoplasmic loop of Cacna1b.
- Author
-
Bunda A, LaCarubba B, Akiki M, and Andrade A
- Subjects
- Alternative Splicing genetics, Animals, Central Nervous System metabolism, Female, Male, Mice, Mice, Inbred C57BL, Models, Genetic, Organ Specificity, RNA, Messenger genetics, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, Cytoplasm genetics
- Abstract
Presynaptic Ca
V 2.2 (N-type) channels are fundamental for transmitter release across the nervous system. The gene encoding CaV 2.2 channels, Cacna1b, contains alternatively spliced exons that result in functionally distinct splice variants (e18a, e24a, e31a, and 37a/37b). Alternative splicing of the cassette exon 18a generates two mRNA transcripts (+e18a-Cacna1b and ∆e18a-Cacna1b). In this study, using novel mouse genetic models and in situ hybridization (BaseScope™), we confirmed that +e18a-Cacna1b splice variants are expressed in monoaminergic regions of the midbrain. We expanded these studies and identified +e18a-Cacna1b mRNA in deep cerebellar cells and spinal cord motor neurons. Furthermore, we determined that +e18a-Cacna1b is enriched in cholecystokinin-expressing interneurons. Our results provide key information to understand cell-specific functions of CaV 2.2 channels., (© 2019 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)- Published
- 2019
- Full Text
- View/download PDF
10. Structure-Activity Analysis of N-Type Calcium Channel Inhibitor SO-3.
- Author
-
Dong M, Wang F, Yan Z, Yu S, Wei J, Wu Q, Liu Z, Tang Y, Ding J, and Dai Q
- Subjects
- Analgesics chemistry, Animals, HEK293 Cells, Humans, Mice, Models, Molecular, Pain metabolism, Peptides chemistry, Protein Conformation, Rats, Structure-Activity Relationship, Analgesics pharmacology, Behavior, Animal drug effects, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Pain drug therapy, Peptides pharmacology
- 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.
- Published
- 2018
- Full Text
- View/download PDF
11. Synthesis and evaluation of aminobenzothiazoles as blockers of N- and T-type calcium channels.
- Author
-
Sairaman A, Cardoso FC, Bispat A, Lewis RJ, Duggan PJ, and Tuck KL
- Subjects
- 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, 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
- 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 Ca
V 2.2 and CaV 3.2 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 CaV 2.2 and CaV 3.2 channels., (Crown Copyright © 2018. Published by Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
12. Simulation of the effect of an external GHz electric field on the potential energy profile of Ca 2+ ions in the selectivity filter of the Ca V Ab channel.
- Author
-
Adiban J, Jamali Y, and Rafii-Tabar H
- Subjects
- 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, Arcobacter metabolism, Bacterial Proteins metabolism, Calcium metabolism, Calcium Channels, N-Type metabolism
- Abstract
Ca
V 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., (© 2018 Wiley Periodicals, Inc.)- Published
- 2018
- Full Text
- View/download PDF
13. Molecular moieties masking Ca 2+ -dependent facilitation of voltage-gated Ca v 2.2 Ca 2+ channels.
- Author
-
Thomas JR, Hagen J, Soh D, and Lee A
- Subjects
- Alternative Splicing, Animals, Binding Sites, Calcium metabolism, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, HEK293 Cells, Humans, Membrane Potentials, Protein Binding, Rats, Calcium Channels, N-Type metabolism, Calmodulin metabolism
- Abstract
Voltage-gated Ca
v 2.1 (P/Q-type) Ca2+ channels undergo Ca2+ -dependent inactivation (CDI) and facilitation (CDF), both of which contribute to short-term synaptic plasticity. Both CDI and CDF are mediated by calmodulin (CaM) binding to sites in the C-terminal domain of the Cav 2.1 α1 subunit, most notably to a consensus CaM-binding IQ-like (IQ) domain. Closely related Cav 2.2 (N-type) channels display CDI but not CDF, despite overall conservation of the IQ and additional sites (pre-IQ, EF-hand-like [EF] domain, and CaM-binding domain) that regulate CDF of Cav 2.1. Here we investigate the molecular determinants that prevent Cav 2.2 channels from undergoing CDF. Although alternative splicing of C-terminal exons regulates CDF of Cav 2.1, the splicing of analogous exons in Cav 2.2 does not reveal CDF. Transfer of sequences encoding the Cav 2.1 EF, pre-IQ, and IQ together (EF-pre-IQ-IQ), but not individually, are sufficient to support CDF in chimeric Cav 2.2 channels; Cav 2.1 chimeras containing the corresponding domains of Cav 2.2, either alone or together, fail to undergo CDF. In contrast to the weak binding of CaM to just the pre-IQ and IQ of Cav 2.2, CaM binds to the EF-pre-IQ-IQ of Cav 2.2 as well as to the corresponding domains of Cav 2.1. Therefore, the lack of CDF in Cav 2.2 likely arises from an inability of its EF-pre-IQ-IQ to transduce the effects of CaM rather than weak binding to CaM per se. Our results reveal a functional divergence in the CDF regulatory domains of Cav 2 channels, which may help to diversify the modes by which Cav 2.1 and Cav 2.2 can modify synaptic transmission., (© 2018 Thomas et al.)- Published
- 2018
- Full Text
- View/download PDF
14. Ca V 2 channel subtype expression in rat sympathetic neurons is selectively regulated by α 2 δ subunits.
- Author
-
Scott MB and Kammermeier PJ
- Subjects
- Animals, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, Calcium Channels, N-Type metabolism, Cells, Cultured, HEK293 Cells, Humans, Male, Protein Subunits biosynthesis, Protein Subunits genetics, Rats, Rats, Wistar, Calcium Channels, N-Type biosynthesis, Neurons metabolism, Protein Subunits metabolism
- Abstract
Type two voltage gated calcium (Ca
V 2) channels are the primary mediators of neurotransmission at neuronal presynapses, but their function at neural soma is also important in regulating excitability.1 Mechanisms that regulate CaV 2 channel expression at synapses have been studied extensively, which motivated us to perform similar studies in the soma. Rat sympathetic neurons from the superior cervical ganglion (SCG) natively express CaV 2.2 and CaV 2.3.2 We noted previously that heterologous expression of CaV 2.1 but not CaV 2.2 results in increased calcium current in SCG neurons.3 In the present study, we extended these observations to show that both CaV 2.1 and CaV 2.3 expression resulted in increased calcium currents while CaV 2.2 expression did not. Further, CaV 2.1 could displace native CaV 2.2 channels, but CaV 2.3 expression could not. Heterologous expression of the individual accessory subunits α2 δ-1, α2 δ-2, α2 δ-3, or β4 alone failed to increase current density, suggesting that the calcium current ceiling when CaV 2.2 was over-expressed was not due to lack of these subunits. Interestingly, introduction of recombinant α2δ subunits produced surprising effects on displacement of native CaV 2.2 by recombinant channels. Both α2 δ-1 and α2 δ-2 seemed to promote CaV 2.2 displacement by recombinant channel expression, while α2 δ-3 appeared to protect CaV 2.2 from displacement. Thus, we observe a selective prioritization of CaV channel functional expression in neurons by specific α2 δ subunits. These data highlight a new function for α2 δ subtypes that could shed light on subtype selectivity of CaV 2 membrane expression.- Published
- 2017
- Full Text
- View/download PDF
15. Stimulatory and inhibitory effects of PKC isozymes are mediated by serine/threonine PKC sites of the Ca v 2.3α 1 subunits.
- Author
-
Rajagopal S, Burton BK, Fields BL, El IO, and Kamatchi GL
- Subjects
- 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, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Membrane Potentials physiology, Oocytes physiology, Protein Kinase C chemistry, Protein Kinase C metabolism
- Abstract
Protein kinase C (PKC) isozymes modulate voltage-gated calcium (Ca
v ) currents through Cav 2.2 and Cav 2.3 channels by targeting serine/threonine (Ser/Thr) phosphorylation sites of Cav α1 subunits. Stimulatory (Thr-422, Ser-2108 and Ser-2132) and inhibitory (Ser-425) sites were identified in the Cav 2.2α1 subunits to PKCs βII and ε. In the current study, we investigated if the homologous sites of Cav 2.3α1 subunits (stimulatory: Thr-365, Ser-1995 and Ser-2011; inhibitory: Ser-369) behaved in similar manner. Several Ala and Asp mutants were constructed in Cav 2.3α1 subunits in such a way that the Ser/Thr sites can be examined in isolation. These mutants or WT Cav 2.3α1 along with auxiliary β1b and α2 /δ subunits were expressed in Xenopus oocytes and the effects of PKCs βII 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 Cav 2.2α1 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., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
16. The HOOK region of voltage-gated Ca2+ channel β subunits senses and transmits PIP2 signals to the gate.
- Author
-
Park CG, Park Y, and Suh BC
- Subjects
- Animals, Binding Sites, Calcium Channels, N-Type chemistry, HEK293 Cells, Humans, Mice, Protein Binding, Protein Subunits chemistry, Protein Subunits metabolism, Rats, Calcium Channels, N-Type metabolism, Ion Channel Gating, Phosphatidylinositol 4,5-Diphosphate metabolism
- Abstract
The β subunit of voltage-gated Ca
2+ (CaV ) channels plays an important role in regulating gating of the α1 pore-forming subunit and its regulation by phosphatidylinositol 4,5-bisphosphate (PIP2 ). Subcellular localization of the CaV β subunit is critical for this effect; N-terminal-dependent membrane targeting of the β subunit slows inactivation and decreases PIP2 sensitivity. Here, we provide evidence that the HOOK region of the β subunit plays an important role in the regulation of CaV biophysics. Based on amino acid composition, we broadly divide the HOOK region into three domains: S (polyserine), A (polyacidic), and B (polybasic). We show that a β subunit containing only its A domain in the HOOK region increases inactivation kinetics and channel inhibition by PIP2 depletion, whereas a β subunit with only a B domain decreases these responses. When both the A and B domains are deleted, or when the entire HOOK region is deleted, the responses are elevated. Using a peptide-to-liposome binding assay and confocal microscopy, we find that the B domain of the HOOK region directly interacts with anionic phospholipids via polybasic and two hydrophobic Phe residues. The β2c-short subunit, which lacks an A domain and contains fewer basic amino acids and no Phe residues in the B domain, neither associates with phospholipids nor affects channel gating dynamically. Together, our data suggest that the flexible HOOK region of the β subunit acts as an important regulator of CaV channel gating via dynamic electrostatic and hydrophobic interaction with the plasma membrane., (© 2017 Park et al.)- Published
- 2017
- Full Text
- View/download PDF
17. Structure-activity relationships of ω-Agatoxin IVA in lipid membranes.
- Author
-
Ryu JH, Jung HJ, Konishi S, Kim HH, Park ZY, and Kim JI
- Subjects
- Animals, Binding Sites, Molecular Conformation, Protein Binding, Rats, Rats, Wistar, Structure-Activity Relationship, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type ultrastructure, Cell Membrane chemistry, Lipid Bilayers chemistry, Purkinje Cells chemistry, omega-Agatoxin IVA chemistry
- 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 β-strands seen in water. However,
15 N 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 β-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., (Copyright © 2016 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
18. Vulnerability of Purkinje Cells Generated from Spinocerebellar Ataxia Type 6 Patient-Derived iPSCs.
- Author
-
Ishida Y, Kawakami H, Kitajima H, Nishiyama A, Sasai Y, Inoue H, and Muguruma K
- Subjects
- Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Cell Differentiation drug effects, Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells metabolism, Protein Domains, Purkinje Cells drug effects, Purkinje Cells metabolism, Riluzole pharmacology, Thyrotropin pharmacology, Up-Regulation drug effects, Induced Pluripotent Stem Cells pathology, Purkinje Cells pathology, Spinocerebellar Ataxias pathology
- Abstract
Spinocerebellar ataxia type 6 (SCA6) is a dominantly inherited neurodegenerative disease characterized by loss of Purkinje cells in the cerebellum. SCA6 is caused by CAG trinucleotide repeat expansion in CACNA1A, which encodes Cav2.1, α1A subunit of P/Q-type calcium channel. However, the pathogenic mechanism and effective therapeutic treatments are still unknown. Here, we have succeeded in generating differentiated Purkinje cells that carry patient genes by combining disease-specific iPSCs and self-organizing culture technologies. Patient-derived Purkinje cells exhibit increased levels of full-length Cav2.1 protein but decreased levels of its C-terminal fragment and downregulation of the transcriptional targets TAF1 and BTG1. We further demonstrate that SCA6 Purkinje cells exhibit thyroid hormone depletion-dependent degeneration, which can be suppressed by two compounds, thyroid releasing hormone and Riluzole. Thus, we have constructed an in vitro disease model recapitulating both ontogenesis and pathogenesis. This model may be useful for pathogenic investigation and drug screening., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
19. 3-Benzamides and 3,4,5-trimethoxyphenyl amines as calcium channel blockers.
- Author
-
Kang B, Oh JA, Lee JY, Rhim H, Yune TY, and Park Choo HY
- Subjects
- 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, Amines chemistry, Calcium Channel Blockers chemistry
- 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., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
20. "Slow" Voltage-Dependent Inactivation of CaV2.2 Calcium Channels Is Modulated by the PKC Activator Phorbol 12-Myristate 13-Acetate (PMA).
- Author
-
Zhu L, McDavid S, and Currie KP
- Subjects
- Animals, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, Calcium Signaling drug effects, Cattle, Cells, Cultured, Chromaffin Cells drug effects, Chromaffin Cells metabolism, Enzyme Activation drug effects, Guanosine Diphosphate analogs & derivatives, Guanosine Diphosphate metabolism, HEK293 Cells, Humans, Kinetics, Patch-Clamp Techniques, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Thionucleotides metabolism, Calcium Channels, N-Type metabolism, Protein Kinase C metabolism, Tetradecanoylphorbol Acetate pharmacology
- Abstract
CaV2.2 (N-type) voltage-gated calcium channels (Ca2+ channels) play key roles in neurons and neuroendocrine cells including the control of cellular excitability, neurotransmitter / hormone secretion, and gene expression. Calcium entry is precisely controlled by channel gating properties including multiple forms of inactivation. "Fast" voltage-dependent inactivation is relatively well-characterized and occurs over the tens-to- hundreds of milliseconds timeframe. Superimposed on this is the molecularly distinct, but poorly understood process of "slow" voltage-dependent inactivation, which develops / recovers over seconds-to-minutes. Protein kinases can modulate "slow" inactivation of sodium channels, but little is known about if/how second messengers control "slow" inactivation of Ca2+ channels. We investigated this using recombinant CaV2.2 channels expressed in HEK293 cells and native CaV2 channels endogenously expressed in adrenal chromaffin cells. The PKC activator phorbol 12-myristate 13-acetate (PMA) dramatically prolonged recovery from "slow" inactivation, but an inactive control (4α-PMA) had no effect. This effect of PMA was prevented by calphostin C, which targets the C1-domain on PKC, but only partially reduced by inhibitors that target the catalytic domain of PKC. The subtype of the channel β-subunit altered the kinetics of inactivation but not the magnitude of slowing produced by PMA. Intracellular GDP-β-S reduced the effect of PMA suggesting a role for G proteins in modulating "slow" inactivation. We postulate that the kinetics of recovery from "slow" inactivation could provide a molecular memory of recent cellular activity and help control CaV2 channel availability, electrical excitability, and neurotransmission in the seconds-to-minutes timeframe.
- Published
- 2015
- Full Text
- View/download PDF
21. The first disease connection for Cav2.2 channels.
- Author
-
Weiss N
- Subjects
- Amino Acid Sequence, Genetic Predisposition to Disease genetics, Humans, Molecular Sequence Data, Mutation genetics, Structure-Activity Relationship, Arrhythmias, Cardiac genetics, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, Dystonic Disorders genetics, Ion Channel Gating genetics
- Abstract
Commentary to: CACNA1B mutation is linked to unique myoclonus-dystonia syndrome. (Hum. Mol. Genet. 2015, pp. 987-993).
- Published
- 2015
- Full Text
- View/download PDF
22. Two New Classes of T-Type Calcium Channel Inhibitors with New Chemical Scaffolds from Ganoderma cochlear.
- Author
-
Zhou FJ, Nian Y, Yan Y, Gong Y, Luo Q, Zhang Y, Hou B, Zuo ZL, Wang SM, Jiang HH, Yang J, and Cheng YX
- Subjects
- Calcium Channel Blockers chemistry, Calcium Channels, N-Type metabolism, Calcium Channels, T-Type metabolism, Humans, Molecular Structure, Neural Inhibition physiology, Terpenes chemistry, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Calcium Channels, T-Type chemistry, Ganoderma chemistry, Neural Inhibition drug effects, Terpenes chemical synthesis, Terpenes pharmacology
- Abstract
T-type calcium channel (TTCC) inhibitors hold great potential for the treatment of a variety of neurological disorders. Cochlearoids A-E (1-5), five pairs of dimeric meroterpenoid enantiomers, and cochlearines A (6) and B (7), two pairs of enantiomeric hybrid metabolites, were isolated and characterized from Ganoderma cochlear. Biological evaluation found that compounds (+)-1, (-)-3, and (±)-6 significantly inhibited Cav3.1 TTCC and showed noticeable selectivity against Cav1.2, Cav2.1, Cav2.2, and Kv11.1 (hERG) channels.
- Published
- 2015
- Full Text
- View/download PDF
23. Solution NMR and calorimetric analysis of Rem2 binding to the Ca2+ channel β4 subunit: a low affinity interaction is required for inhibition of Cav2.1 Ca2+ currents.
- Author
-
Xu X, Zhang F, Zamponi GW, and Horne WA
- Subjects
- Calcium Channels, N-Type genetics, Electrophysiology, HEK293 Cells, Humans, Protein Conformation, Protein Interaction Domains and Motifs, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Calorimetry methods, Ion Channels physiology, Magnetic Resonance Spectroscopy methods, Monomeric GTP-Binding Proteins metabolism
- Abstract
Rem, Rad, Kir/Gem (RGK) proteins, including Rem2, mediate profound inhibition of high-voltage activated Ca(2+) channels containing intracellular regulatory β subunits. All RGK proteins bind to voltage-gated Ca(2+) channel β subunit (Cavβ) 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 Cavβ4a and measure its binding affinity. Our experiments revealed 2 binding surfaces on the β4 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 Cavβ4a, but were unaffected by Rem2 when coexpressed with a Cavβ4a site 1 (L173A/V303A) or site 2 (D258A) mutant. These results provide direct evidence for a low-affinity Rem2/Cavβ4 interaction and show definitively that the interaction is required for Cav2.1 inhibition., (© FASEB.)
- Published
- 2015
- Full Text
- View/download PDF
24. Inhibition of N-type calcium channels by fluorophenoxyanilide derivatives.
- Author
-
Gleeson EC, Graham JE, Spiller S, Vetter I, Lewis RJ, Duggan PJ, and Tuck KL
- Subjects
- 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, 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
- 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.
- Published
- 2015
- Full Text
- View/download PDF
25. Molecular characterization and functional expression of the Apis mellifera voltage-dependent Ca2+ channels.
- Author
-
Cens T, Rousset M, Collet C, Charreton M, Garnery L, Le Conte Y, Chahine M, Sandoz JC, and Charnet P
- Subjects
- Amino Acid Sequence, Animals, Bees chemistry, Bees genetics, Calcium metabolism, Calcium Channels chemistry, Calcium Channels genetics, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, Calcium Channels, N-Type metabolism, Exons, Insect Proteins metabolism, Membrane Potentials, Molecular Sequence Data, Protein Structure, Tertiary, Sequence Alignment, Xenopus, Bees metabolism, Calcium Channels metabolism
- Abstract
Voltage-gated Ca(2+) channels allow the influx of Ca(2+) ions from the extracellular space upon membrane depolarization and thus serve as a transducer between membrane potential and cellular events initiated by Ca(2+) transients. Most insects are predicted to possess three genes encoding Cavα, the main subunit of Ca(2+) channels, and several genes encoding the two auxiliary subunits, Cavβ and Cavα2δ; however very few of these genes have been cloned so far. Here, we cloned three full-length cDNAs encoding the three Cavα subunits (AmelCav1a, AmelCav2a and AmelCav3a), a cDNA encoding a novel variant of the Cavβ subunit (AmelCavβc), and three full-length cDNAs encoding three Cavα2δ subunits (AmelCavα2δ1 to 3) of the honeybee Apis mellifera. We identified several alternative or mutually exclusive exons in the sequence of the AmelCav2 and AmelCav3 genes. Moreover, we detected a stretch of glutamine residues in the C-terminus of the AmelCav1 subunit that is reminiscent of the motif found in the human Cav2.1 subunit of patients with Spinocerebellar Ataxia type 6. All these subunits contain structural domains that have been identified as functionally important in their mammalian homologues. For the first time, we could express three insect Cavα subunits in Xenopus oocytes and we show that AmelCav1a, 2a and 3a form Ca(2+) channels with distinctive properties. Notably, the co-expression of AmelCav1a or AmelCav2a with AmelCavβc and AmCavα2δ1 produces High Voltage-Activated Ca(2+) channels. On the other hand, expression of AmelCav3a alone leads to Low Voltage-Activated Ca(2+) channels., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
26. Single calcium channel domain gating of synaptic vesicle fusion at fast synapses; analysis by graphic modeling.
- Author
-
Stanley EF
- Subjects
- Animals, Calcium Channels, N-Type chemistry, Mice, Models, Neurological, Protein Multimerization, Protein Structure, Tertiary, Rats, Xenopus, Calcium Channels, N-Type metabolism, Ion Channel Gating, Neuromuscular Junction metabolism, Synaptic Vesicles metabolism
- Abstract
At fast-transmitting presynaptic terminals Ca(2+) enter through voltage gated calcium channels (CaVs) and bind to a synaptic vesicle (SV) -associated calcium sensor (SV-sensor) to gate fusion and discharge. An open CaV generates a high-concentration plume, or nanodomain of Ca(2+) that dissipates precipitously with distance from the pore. At most fast synapses, such as the frog neuromuscular junction (NMJ), the SV sensors are located sufficiently close to individual CaVs to be gated by single nanodomains. However, at others, such as the mature rodent calyx of Held (calyx of Held), the physiology is more complex with evidence that CaVs that are both close and distant from the SV sensor and it is argued that release is gated primarily by the overlapping Ca(2+) nanodomains from many CaVs. We devised a 'graphic modeling' method to sum Ca(2+) from individual CaVs located at varying distances from the SV-sensor to determine the SV release probability and also the fraction of that probability that can be attributed to single domain gating. This method was applied first to simplified, low and high CaV density model release sites and then to published data on the contrasting frog NMJ and the rodent calyx of Held native synapses. We report 3 main predictions: the SV-sensor is positioned very close to the point at which the SV fuses with the membrane; single domain-release gating predominates even at synapses where the SV abuts a large cluster of CaVs, and even relatively remote CaVs can contribute significantly to single domain-based gating.
- Published
- 2015
- Full Text
- View/download PDF
27. Modeling a Ca(2+) channel/BKCa channel complex at the single-complex level.
- Author
-
Cox DH
- Subjects
- Animals, Calcium Channels, N-Type chemistry, Humans, Large-Conductance Calcium-Activated Potassium Channels chemistry, Mice, Protein Binding, Protein Subunits chemistry, Protein Subunits metabolism, Rats, Calcium Channels, N-Type metabolism, Large-Conductance Calcium-Activated Potassium Channels metabolism, Protein Multimerization
- 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., (Copyright © 2014 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
28. Discovery of novel tetrahydroisoquinoline derivatives as orally active N-type calcium channel blockers with high selectivity for hERG potassium channels.
- Author
-
Ogiyama T, Inoue M, Honda S, Yamada H, Watanabe T, Gotoh T, Kiso T, Koakutsu A, Kakimoto S, and Shishikura J
- Subjects
- Animals, Calcium Channel Blockers metabolism, Calcium Channel Blockers therapeutic use, Calcium Channels, N-Type chemistry, Cell Line, Tumor, Cytochrome P-450 Enzyme System chemistry, Cytochrome P-450 Enzyme System metabolism, Drug Evaluation, Preclinical, Ether-A-Go-Go Potassium Channels chemistry, Humans, Male, Neuralgia drug therapy, Protein Binding, Rats, Rats, Sprague-Dawley, Structure-Activity Relationship, Tetrahydroisoquinolines metabolism, Tetrahydroisoquinolines therapeutic use, Calcium Channel Blockers chemistry, Calcium Channels, N-Type metabolism, Ether-A-Go-Go Potassium Channels metabolism, Tetrahydroisoquinolines chemistry
- Abstract
N-type calcium channels represent a promising target for the treatment of neuropathic pain. The selective N-type calcium channel blocker ziconotide ameliorates severe chronic pain but has a narrow therapeutic window and requires intrathecal administration. We identified tetrahydroisoquinoline derivative 1a as a novel potent N-type calcium channel blocker. However, this compound also exhibited potent inhibitory activity against hERG channels. Structural optimizations led to identification of (1S)-(1-cyclohexyl-3,4-dihydroisoquinolin-2(1H)-yl)-2-{[(1-hydroxycyclohexyl)methyl]amino}ethanone ((S)-1h), which exhibited high selectivity for hERG channels while retaining potency for N-type calcium channel inhibition. (S)-1h went on to demonstrate in vivo efficacy as an orally available N-type calcium channel blocker in a rat spinal nerve ligation model of neuropathic pain., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
29. Potentiation of high voltage-activated calcium channels by 4-aminopyridine depends on subunit composition.
- Author
-
Li L, Li DP, Chen SR, Chen J, Hu H, and Pan HL
- Subjects
- 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, 4-Aminopyridine pharmacology, Calcium Channels, L-Type drug effects, Calcium Channels, N-Type drug effects
- 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 β (Cavβ) subunits. Among the four Cavβ subunits examined, Cavβ3 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 Cavβ3. 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 Cavβ3 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 Cavβ3 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-Cavβ3 and Cav2.2-Cavβ3 interactions. The therapeutic effect of 4-AP on neuromuscular function is probably mediated by its actions on Cavβ3-containing L-type Ca(2+) channels., (Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2014
- Full Text
- View/download PDF
30. Modulation of CaV2.1 channels by neuronal calcium sensor-1 induces short-term synaptic facilitation.
- Author
-
Yan J, Leal K, Magupalli VG, Nanou E, Martinez GQ, Scheuer T, and Catterall WA
- Subjects
- 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, Calcium Channels, N-Type metabolism, Neuronal Calcium-Sensor Proteins metabolism, Neuropeptides metabolism, Synapses metabolism, Synaptic Transmission
- 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.
- Published
- 2014
- Full Text
- View/download PDF
31. Demonstration of binding of neuronal calcium sensor-1 to the cav2.1 p/q-type calcium channel.
- Author
-
Lian LY, Pandalaneni SR, Todd PA, Martin VM, Burgoyne RD, and Haynes LP
- Subjects
- Calcium metabolism, Calcium Channels, N-Type chemistry, Cloning, Molecular, Humans, Neuronal Calcium-Sensor Proteins chemistry, Neuropeptides chemistry, Protein Binding, Calcium Channels, N-Type metabolism, Neuronal Calcium-Sensor Proteins metabolism, Neuropeptides metabolism
- 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.
- Published
- 2014
- Full Text
- View/download PDF
32. Voltage control of Ca²⁺ permeation through N-type calcium (Ca(V)2.2) channels.
- Author
-
Buraei Z, Liang H, and Elmslie KS
- Subjects
- Animals, Barium pharmacology, Binding Sites, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Cells, Cultured, Ion Transport, Neurons drug effects, Neurons metabolism, Neurons physiology, Purines pharmacology, Rana catesbeiana, Roscovitine, Calcium metabolism, Calcium Channels, N-Type metabolism, Membrane Potentials
- Abstract
Voltage-gated calcium (Ca(V)) channels deliver Ca(2+) to trigger cellular functions ranging from cardiac muscle contraction to neurotransmitter release. The mechanism by which these channels select for Ca(2+) over other cations is thought to involve multiple Ca(2+)-binding sites within the pore. Although the Ca(2+) affinity and cation preference of these sites have been extensively investigated, the effect of voltage on these sites has not received the same attention. We used a neuronal preparation enriched for N-type calcium (Ca(V)2.2) channels to investigate the effect of voltage on Ca(2+) flux. We found that the EC50 for Ca(2+) permeation increases from 13 mM at 0 mV to 240 mM at 60 mV, indicating that, during permeation, Ca(2+) ions sense the electric field. These data were nicely reproduced using a three-binding-site step model. Using roscovitine to slow Ca(V)2.2 channel deactivation, we extended these measurements to voltages <0 mV. Permeation was minimally affected at these hyperpolarized voltages, as was predicted by the model. As an independent test of voltage effects on permeation, we examined the Ca(2+)-Ba(2+) anomalous mole fraction (MF) effect, which was both concentration and voltage dependent. However, the Ca(2+)-Ba(2+) anomalous MF data could not be reproduced unless we added a fourth site to our model. Thus, Ca(2+) permeation through Ca(V)2.2 channels may require at least four Ca(2+)-binding sites. Finally, our results suggest that the high affinity of Ca(2+) for the channel helps to enhance Ca(2+) influx at depolarized voltages relative to other ions (e.g., Ba(2+) or Na(+)), whereas the absence of voltage effects at negative potentials prevents Ca(2+) from becoming a channel blocker. Both effects are needed to maximize Ca(2+) influx over the voltages spanned by action potentials., (© 2014 Buraei et al.)
- Published
- 2014
- Full Text
- View/download PDF
33. Crotoxin from Crotalus durissus terrificus snake venom induces the release of glutamate from cerebrocortical synaptosomes via N and P/Q calcium channels.
- Author
-
Lomeo Rda S, Gonçalves AP, da Silva CN, de Paula AT, Costa Santos DO, Fortes-Dias CL, Gomes DA, and de Lima ME
- Subjects
- Animals, Calcium Channel Agonists metabolism, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Calcium Signaling drug effects, Cells, Cultured, Cerebral Cortex metabolism, Crotalid Venoms enzymology, Crotoxin antagonists & inhibitors, Crotoxin metabolism, Glutamic Acid metabolism, Group II Phospholipases A2 antagonists & inhibitors, Group II Phospholipases A2 metabolism, Group II Phospholipases A2 pharmacology, Male, Nerve Tissue Proteins agonists, Nerve Tissue Proteins antagonists & inhibitors, Nerve Tissue Proteins metabolism, Neurons drug effects, Neurons metabolism, Neurotoxins antagonists & inhibitors, Neurotoxins metabolism, Neurotoxins pharmacology, Protein Subunits antagonists & inhibitors, Protein Subunits metabolism, Protein Subunits pharmacology, Protein Transport drug effects, Rats, Wistar, Reptilian Proteins antagonists & inhibitors, Reptilian Proteins metabolism, Reptilian Proteins pharmacology, Synaptic Transmission drug effects, Synaptosomes metabolism, Calcium Channel Agonists pharmacology, Calcium Channels, N-Type metabolism, Cerebral Cortex drug effects, Crotalid Venoms chemistry, Crotalus, Crotoxin pharmacology, Synaptosomes drug effects
- Abstract
Crotoxin (Crtx), the main toxin in the venom of Crotalus durissus terrificus snake, is a heterodimer with a basic subunit, CB, and an acidic subunit, CA. CB is a phospholipase A2 that depends on CA to specifically bind to the cell membrane. This toxin acts in the central nervous system (CNS) causing chronic seizure effects and other cytotoxic effects. Here, we report its action on glutamate release in rat cerebral cortex synaptosomes. Aiming at a better understanding of the mechanism of action of Crtx, calcium channel blockers were used and internalization studies were performed in cerebellar granule neurons. Our results show that Crtx induces calcium-dependent glutamate release via N and P/Q calcium channels. In addition, the CB subunit of Crtx is shown to be internalized. This internalization does not depend on the presence of CA subunit neither on the PLA2 activity of CB. A correlation between CB internalization and glutamate release remains to be established., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
34. Ankyrin-B regulates Cav2.1 and Cav2.2 channel expression and targeting.
- Author
-
Kline CF, Scott J, Curran J, Hund TJ, and Mohler PJ
- Subjects
- 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, Ankyrins metabolism, Calcium Channels, N-Type metabolism
- 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.
- Published
- 2014
- Full Text
- View/download PDF
35. Identifying key amino acid residues that affect α-conotoxin AuIB inhibition of α3β4 nicotinic acetylcholine receptors.
- Author
-
Grishin AA, Cuny H, Hung A, Clark RJ, Brust A, Akondi K, Alewood PF, Craik DJ, and Adams DJ
- Subjects
- Alanine chemistry, Alanine genetics, Amino Acids chemistry, Amino Acids genetics, Animals, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism, Conotoxins chemistry, Conotoxins genetics, Gene Expression Regulation, Humans, Magnetic Resonance Spectroscopy, Models, Molecular, Molecular Docking Simulation, Molecular Dynamics Simulation, Mutagenesis, Site-Directed, Nociception, Oocytes metabolism, Protein Binding, Protein Interaction Domains and Motifs, Rats, Receptors, Nicotinic chemistry, Receptors, Nicotinic genetics, Xenopus laevis, Amino Acids metabolism, Conotoxins metabolism, Receptors, Nicotinic metabolism
- Abstract
α-Conotoxin AuIB is a selective α3β4 nicotinic acetylcholine receptor (nAChR) subtype inhibitor. Its analgesic properties are believed to result from it activating GABAB receptors and subsequently inhibiting CaV2.2 voltage-gated calcium channels. The structural determinants that mediate diverging AuIB activity at these targets are unknown. We performed alanine scanning mutagenesis of AuIB and α3β4 nAChR, homology modeling, and molecular dynamics simulations to identify the structural determinants of the AuIB·α3β4 nAChR interaction. Two alanine-substituted AuIB analogues, [P6A]AuIB and [F9A]AuIB, did not inhibit the α3β4 nAChR. NMR and CD spectroscopy studies demonstrated that [F9A]AuIB retains its native globular structure, so its activity loss is probably due to loss of specific toxin-receptor residue pairwise contacts. Compared with AuIB, the concentration-response curve for inhibition of α3β4 by [F9A]AuIB shifted rightward more than 10-fold, and its subtype selectivity profile changed. Homology modeling and molecular dynamics simulations suggest that Phe-9 of AuIB interacts with a two-residue binding pocket on the β4 nAChR subunit. This hypothesis was confirmed by site-directed mutagenesis of the β4-Trp-59 and β4-Lys-61 residues of loop D, which form a putative binding pocket. AuIB analogues with Phe-9 substitutions corroborated the finding of a binding pocket on the β4 subunit and gave further insight into how AuIB Phe-9 interacts with the β4 subunit. In summary, we identified critical residues that mediate interactions between AuIB and its cognate nAChR subtype. These findings might help improve the design of analgesic conopeptides that selectively "avoid" nAChR receptors while targeting receptors involved with nociception.
- Published
- 2013
- Full Text
- View/download PDF
36. Mechanisms of conotoxin inhibition of N-type (Ca(v)2.2) calcium channels.
- Author
-
Adams DJ and Berecki G
- Subjects
- Animals, Calcium Channels, N-Type metabolism, Humans, Mice, Pain metabolism, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Conotoxins pharmacology, Pain drug therapy
- Abstract
N-type (Ca(v)2.2) voltage-gated calcium channels (VGCC) transduce electrical activity into other cellular functions, regulate calcium homeostasis and play a major role in processing pain information. Although the distribution and function of these channels vary widely among different classes of neurons, they are predominantly expressed in nerve terminals, where they control neurotransmitter release. To date, genetic and pharmacological studies have identified that high-threshold, N-type VGCCs are important for pain sensation in disease models. This suggests that N-type VGCC inhibitors or modulators could be developed into useful drugs to treat neuropathic pain. This review discusses the role of N-type (Ca(v)2.2) VGCCs in nociception and pain transmission through primary sensory dorsal root ganglion (DRG) neurons (nociceptors). It also outlines the potent and selective inhibition of N-type VGCCs by conotoxins, small disulfide-rich peptides isolated from the venom of marine cone snails. Of these conotoxins, ω-conotoxins are selective N-type VGCC antagonists that preferentially block nociception in inflammatory pain models, and allodynia and/or hyperalgesia in neuropathic pain models. Another conotoxin family, α-conotoxins, were initially proposed as competitive antagonists of muscle and neuronal nicotinic acetylcholine receptors (nAChR). Surprisingly, however, α-conotoxins Vc1.1 and RgIA, also potently inhibit N-type VGCC currents in the sensory DRG neurons of rodents and α9 nAChR knockout mice, via intracellular signaling mediated by G protein-coupled GABAB receptors. Understanding how conotoxins inhibit VGCCs is critical for developing these peptides into analgesics and may result in better pain management. This article is part of a Special Issue entitled: Calcium channels., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. Structural flexibility of CaV1.2 and CaV2.2 I-II proximal linker fragments in solution.
- Author
-
Almagor L, Avinery R, Hirsch JA, and Beck R
- Subjects
- Animals, Guanylate Kinases chemistry, Models, Molecular, Protein Structure, Secondary, Protein Structure, Tertiary, Rabbits, Solutions, Calcium Channels, L-Type chemistry, Calcium Channels, N-Type chemistry
- Abstract
Voltage-dependent calcium channels (CaV) enable the inward flow of calcium currents for a wide range of cells. CaV1 and CaV2 subtype α1 subunits form the conducting pore using four repeated membrane domains connected by intracellular linkers. The domain I-II linker connects to the membrane gate (IS6), forming an α-helix, and is bound to the CaVβ subunit. Previous studies indicated that this region may or may not form a continuous helix depending on the CaV subtype, thereby modulating channel activation and inactivation properties. Here, we used small-angle x-ray scattering and ensemble modeling analysis to investigate the solution structure of these linkers, extending from the membrane domain and including the CaVβ-binding site, called the proximal linker (PL). The results demonstrate that the CaV1.2 PL is more flexible than the CaV2.2 PL, the flexibility is intrinsic and not dependent on CaVβ binding, and the flexibility can be most easily explained by the presence of conserved glycines. Our analysis also provides a robust example of investigating protein domains in which flexibility plays an essential role., (Copyright © 2013 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
38. Modular, efficient synthesis of asymmetrically substituted piperazine scaffolds as potent calcium channel blockers.
- Author
-
Borzenko A, Pajouhesh H, Morrison JL, Tringham E, Snutch TP, and Schafer LL
- Subjects
- Animals, Calcium Channel Blockers chemistry, Calcium Channel Blockers metabolism, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type genetics, Calcium Channels, L-Type metabolism, Calcium Channels, N-Type genetics, Calcium Channels, N-Type metabolism, Catalysis, Cell Line, Humans, Piperazine, Piperazines chemical synthesis, Piperazines metabolism, Protein Binding, Rats, Stereoisomerism, Structure-Activity Relationship, Titanium chemistry, Zirconium chemistry, Calcium Channel Blockers chemical synthesis, Calcium Channels, N-Type chemistry, Piperazines chemistry
- Abstract
A novel approach to the synthesis of substituted piperazines and their investigation as N-type calcium channel blockers is presented. A common scaffold exhibiting high activity as N-type blockers is N-substituted piperazine. Using recently developed titanium and zirconium catalysts, we describe the efficient and modular synthesis of 2,5-asymmetrically disubstituted piperazines from simple amines and alkynes. The method requires only three isolation/purification protocols and no protection/deprotection steps for the diastereoselective synthesis of 2,5-dialkylated piperazines in moderate to high yield. Screening of the synthesized piperazines for N-type channel blocking activity and selectivity shows the highest activity for a compound with a benzhydryl group on the nitrogen (position 1) and an unprotected alcohol-functionalized side chain., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
39. Complex structures between the N-type calcium channel (CaV2.2) and ω-conotoxin GVIA predicted via molecular dynamics.
- Author
-
Chen R and Chung SH
- Subjects
- Amino Acid Sequence, Molecular Sequence Data, Protein Conformation, Sequence Homology, Amino Acid, Calcium Channels, N-Type chemistry, Molecular Dynamics Simulation, omega-Conotoxin GVIA chemistry
- Abstract
The N-type voltage-gated Ca(2+) channel CaV2.2 is one of the important targets for pain management. ω-Conotoxins isolated from venoms of cone snails, which specifically inhibit CaV2.2, are promising scaffolds for novel analgesics. The inhibitory action of ω-conotoxins on CaV2.2 has been examined experimentally, but the modes of binding of the toxins to this and other related subfamilies of Ca(2+) channels are not understood in detail. Here molecular dynamics simulations are used to construct models of ω-conotoxin GVIA in complex with a homology model of the pore domain of CaV2.2. Three different binding modes in which the side chain of Lys2, Arg17, or Lys24 from the toxin protrudes into the selectivity filter of CaV2.2 are considered. In all the modes, the toxin forms a salt bridge with an aspartate residue of subunit II just above the EEEE ring of the selectivity filter. Using the umbrella sampling technique and potential of mean force calculations, the half-maximal inhibitory concentration (IC50) values are calculated to be 1.5 and 0.7 nM for the modes in which Lys2 and Arg17 occlude the ion conduction pathway, respectively. Both IC50 values compare favorably with the values of 0.04-1.0 nM determined experimentally. The similar IC50 values calculated for the different binding modes demonstrate that GVIA can inhibit CaV2.2 with alternative binding modes. Such a multiple-binding mode mechanism may be common for ω-conotoxins.
- Published
- 2013
- Full Text
- View/download PDF
40. Ca2+-independent activation of Ca2+/calmodulin-dependent protein kinase II bound to the C-terminal domain of CaV2.1 calcium channels.
- Author
-
Magupalli VG, Mochida S, Yan J, Jiang X, Westenbroek RE, Nairn AC, Scheuer T, and Catterall WA
- Subjects
- Electrophysiology methods, Humans, Models, Biological, Neuronal Plasticity, Neurotransmitter Agents metabolism, Phosphorylation, Presynaptic Terminals metabolism, Protein Binding, Protein Structure, Tertiary, Recombinant Proteins metabolism, Signal Transduction, Synapses metabolism, Transfection, Calcium Channels, N-Type chemistry, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Gene Expression Regulation
- Abstract
Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) forms a major component of the postsynaptic density where its functions in synaptic plasticity are well established, but its presynaptic actions are poorly defined. Here we show that CaMKII binds directly to the C-terminal domain of Ca(V)2.1 channels. Binding is enhanced by autophosphorylation, and the kinase-channel signaling complex persists after dephosphorylation and removal of the Ca(2+)/CaM stimulus. Autophosphorylated CaMKII can bind the Ca(V)2.1 channel and synapsin-1 simultaneously. CaMKII binding to Ca(V)2.1 channels induces Ca(2+)-independent activity of the kinase, which phosphorylates the enzyme itself as well as the neuronal substrate synapsin-1. Facilitation and inactivation of Ca(V)2.1 channels by binding of Ca(2+)/CaM mediates short term synaptic plasticity in transfected superior cervical ganglion neurons, and these regulatory effects are prevented by a competing peptide and the endogenous brain inhibitor CaMKIIN, which blocks binding of CaMKII to Ca(V)2.1 channels. These results define the functional properties of a signaling complex of CaMKII and Ca(V)2.1 channels in which both binding partners are persistently activated by their association, and they further suggest that this complex is important in presynaptic terminals in regulating protein phosphorylation and short term synaptic plasticity.
- Published
- 2013
- Full Text
- View/download PDF
41. Protein kinase Cα and P-type Ca channel CaV2.1 in red blood cell calcium signalling.
- Author
-
Wagner-Britz L, Wang J, Kaestner L, and Bernhardt I
- Subjects
- Agatoxins pharmacology, Aniline Compounds chemistry, Calcium metabolism, Calcium Channels, N-Type chemistry, Calcium Signaling drug effects, Cell Membrane drug effects, Erythrocytes cytology, Erythrocytes drug effects, Flow Cytometry, Hemolysis, Humans, Indoles pharmacology, Kinetics, Lysophospholipids pharmacology, Maleimides pharmacology, Phosphatidylserines pharmacology, Protein Kinase C-alpha antagonists & inhibitors, Tetradecanoylphorbol Acetate analogs & derivatives, Tetradecanoylphorbol Acetate pharmacology, Xanthenes chemistry, Calcium Channels, N-Type metabolism, Erythrocytes metabolism, Protein Kinase C-alpha metabolism
- Abstract
Background/aims: Protein kinase Cα (PKCα) is activated by an increase in cytosolic Ca(2+) in red blood cells (RBCs). Previous work has suggested that PKCα directly stimulates the CaV2.1 channel, whereas other studies revealed that CaV2.1 is insensitive to activation by PKC. The aim of this study was to resolve this discrepancy., Methods: We performed experiments based on a single cell read-out of the intracellular Ca(2+) concentration in terms of Fluo-4 fluorescence intensity and phosphatidylserine exposure to the external membrane leaflet. Measurement modalities included flow cytometry and live cell imaging., Results: Treatment of RBCs with phorbol 12-myristate 13-acetate (PMA) led to two distinct populations of cells with an increase in intracellular Ca(2+): a weak-responding and a strong-responding population. The EC50 of PMA for the number of cells with Ca(2+) elevation was 2.7±1.2 µM; for phosphatidylserine exposure to the external membrane surface, it was 2.8±0.5 µM; and for RBC haemolysis, it was 2.9±0.5 µM. Using pharmacological manipulation with the CaV2.1 inhibitor ω-agatoxin TK and the broad protein kinase C inhibitor Gö6983, we are able to show that there are two independent PMA-activated Ca(2+) entry processes: the first is independent of CaV2.1 and directly PKCα-activated, while the second is associated with a likely indirect activation of CaV2.1. Further studies using lysophosphatidic acid (LPA) as a stimulation agent have provided additional evidence that PKCα and CaV2.1 are not directly interconnected in a signalling chain., Conclusion: Although we provide evidence for a lack of interaction between PKCα and CaV2.1 in RBCs, further studies are required to decipher the signalling relationship between LPA, PKCα and CaV2.1., (Copyright © 2013 S. Karger AG, Basel.)
- Published
- 2013
- Full Text
- View/download PDF
42. Aminopiperidine sulfonamide Cav2.2 channel inhibitors for the treatment of chronic pain.
- Author
-
Shao PP, Ye F, Chakravarty PK, Varughese DJ, Herrington JB, Dai G, Bugianesi RM, Haedo RJ, Swensen AM, Warren VA, Smith MM, Garcia ML, McManus OB, Lyons KA, Li X, Green M, Jochnowitz N, McGowan E, Mistry S, Sun SY, Abbadie C, Kaczorowski GJ, and Duffy JL
- Subjects
- Animals, Calcium Channel Blockers chemical synthesis, Calcium Channel Blockers pharmacokinetics, Calcium Channels, N-Type metabolism, Cells, Cultured, Dogs, Humans, Mice, Mice, Knockout, Microsomes, Liver drug effects, Patch-Clamp Techniques, Piperidines chemical synthesis, Piperidines pharmacokinetics, Rats, Rats, Sprague-Dawley, Sulfonamides chemical synthesis, Sulfonamides pharmacokinetics, Tissue Distribution, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type physiology, Chronic Pain drug therapy, Hyperalgesia drug therapy, Inflammation drug therapy, Neuralgia drug therapy, Piperidines pharmacology, Sulfonamides pharmacology
- Abstract
The voltage-gated calcium channel Ca(v)2.2 (N-type calcium channel) is a critical regulator of synaptic transmission and has emerged as an attractive target for the treatment of chronic pain. We report here the discovery of sulfonamide-derived, state-dependent inhibitors of Ca(v)2.2. In particular, 19 is an inhibitor of Ca(v)2.2 that is selective over cardiac ion channels, with a good preclinical PK and biodistribution profile. This compound exhibits dose-dependent efficacy in preclinical models of inflammatory hyperalgesia and neuropathic allodynia and is devoid of ancillary cardiovascular or CNS pharmacology at the doses tested. Importantly, 19 exhibited no efficacy in Ca(v)2.2 gene-deleted mice. The discovery of metabolite 26 confounds further development of members of this aminopiperidine sulfonamide series. This discovery also suggests specific structural liabilities of this class of compounds that must be addressed.
- Published
- 2012
- Full Text
- View/download PDF
43. Calcium currents are enhanced by α2δ-1 lacking its membrane anchor.
- Author
-
Kadurin I, Alvarez-Laviada A, Ng SF, Walker-Gray R, D'Arco M, Fadel MG, Pratt WS, and Dolphin AC
- Subjects
- Animals, Cell Membrane metabolism, DNA, Complementary metabolism, Electrophysiology methods, Ganglia, Spinal metabolism, Hydrogen-Ion Concentration, Immunohistochemistry methods, Protein Binding, Protein Structure, Tertiary, Protein Subunits chemistry, Rabbits, Rats, Rats, Sprague-Dawley, Sequence Analysis, DNA, Calcium metabolism, Calcium Channels, N-Type chemistry
- Abstract
The accessory α(2)δ subunits of voltage-gated calcium channels are membrane-anchored proteins, which are highly glycosylated, possess multiple disulfide bonds, and are post-translationally cleaved into α(2) and δ. All α(2)δ subunits have a C-terminal hydrophobic, potentially trans-membrane domain and were described as type I transmembrane proteins, but we found evidence that they can be glycosylphosphatidylinositol-anchored. To probe further the function of membrane anchoring in α(2)δ subunits, we have now examined the properties of α(2)δ-1 constructs truncated at their putative glycosylphosphatidylinositol anchor site, located before the C-terminal hydrophobic domain (α(2)δ-1ΔC-term). We find that the majority of α(2)δ-1ΔC-term is soluble and secreted into the medium, but unexpectedly, some of the protein remains associated with detergent-resistant membranes, also termed lipid rafts, and is extrinsically bound to the plasma membrane. Furthermore, heterologous co-expression of α(2)δ-1ΔC-term with Ca(V)2.1/β1b results in a substantial enhancement of the calcium channel currents, albeit less than that produced by wild-type α(2)δ-1. These results call into question the role of membrane anchoring of α(2)δ subunits for calcium current enhancement.
- Published
- 2012
- Full Text
- View/download PDF
44. Calcium channel auxiliary α2δ and β subunits: trafficking and one step beyond.
- Author
-
Dolphin AC
- Subjects
- Animals, Humans, Protein Subunits chemistry, Protein Subunits metabolism, Protein Transport physiology, Calcium Channels chemistry, Calcium Channels metabolism, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type metabolism, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type metabolism
- Abstract
The voltage-gated calcium channel α(2)δ and β 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, β 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.
- Published
- 2012
- Full Text
- View/download PDF
45. Synthesis and SAR of 4-aminocyclopentapyrrolidines as N-type Ca²⁺ channel blockers with analgesic activity.
- Author
-
Beebe X, Darczak D, Henry RF, Vortherms T, Janis R, Namovic M, Donnelly-Roberts D, Kage KL, Surowy C, Milicic I, Niforatos W, Swensen A, Marsh KC, Wetter JM, Franklin P, Baker S, Zhong C, Simler G, Gomez E, Boyce-Rustay JM, Zhu CZ, Stewart AO, Jarvis MF, and Scott VE
- Subjects
- Acetamides pharmacology, Acetamides therapeutic use, Analgesics pharmacology, Analgesics therapeutic use, Animals, Behavior, Animal drug effects, Calcium Channel Blockers pharmacology, Calcium Channel Blockers therapeutic use, Calcium Channels, N-Type metabolism, Disease Models, Animal, Male, Pain drug therapy, Pyrrolidines pharmacology, Pyrrolidines therapeutic use, Rats, Rats, Sprague-Dawley, Structure-Activity Relationship, Acetamides chemical synthesis, Analgesics chemical synthesis, Calcium Channel Blockers chemical synthesis, Calcium Channels, N-Type chemistry, Pyrrolidines chemical synthesis, Pyrrolidines chemistry
- Abstract
A novel 4-aminocyclopentapyrrolidine series of N-type Ca(2+) channel blockers have been discovered. Enantioselective synthesis of the 4-aminocyclopentapyrrolidines was enabled using N-tert-butyl sulfinamide chemistry. SAR studies demonstrate selectivity over L-type Ca(2+) channels. N-type Ca(2+) channel blockade was confirmed using electrophysiological recording techniques. Compound 25 is an N-type Ca(2+) channel blocker that produces antinociception in inflammatory and nociceptive pain models without exhibiting cardiovascular or motor liabilities., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
46. Molecular determinants of Gem protein inhibition of P/Q-type Ca2+ channels.
- Author
-
Fan M, Zhang WK, Buraei Z, and Yang J
- Subjects
- Amino Acid Motifs physiology, Amino Acid Sequence, Animals, Binding Sites physiology, Calcium Channels, N-Type chemistry, Calmodulin metabolism, Cell Membrane physiology, Escherichia coli genetics, HEK293 Cells, Humans, Molecular Sequence Data, Monomeric GTP-Binding Proteins chemistry, Oocytes physiology, Patch-Clamp Techniques, Peptide Mapping, Protein Structure, Tertiary physiology, Rabbits, Rats, Signal Transduction physiology, Xenopus laevis, Calcium Channels, N-Type genetics, Calcium Channels, N-Type physiology, Monomeric GTP-Binding Proteins genetics, Monomeric GTP-Binding Proteins physiology
- Abstract
The RGK family of monomeric GTP-binding proteins potently inhibits high voltage-activated Ca(2+) channels. The molecular mechanisms of this inhibition are largely unclear. In Xenopus oocytes, Gem suppresses the activity of P/Q-type Ca(2+) channels on the plasma membrane. This is presumed to occur through direct interactions of one or more Gem inhibitory sites and the pore-forming Ca(v)2.1 subunit in a manner dependent on the Ca(2+) channel subunit β (Ca(v)β). In this study we investigated the molecular determinants in Gem that are critical for this inhibition. Like other RGK proteins, Gem contains a conserved Ras-like core and extended N and C termini. A 12-amino acid fragment in the C terminus was found to be crucial for and sufficient to produce Ca(v)β-dependent inhibition, suggesting that this region forms an inhibitory site. A three-amino acid motif in the core was also found to be critical, possibly forming another inhibitory site. Mutating either site individually did not hamper Gem inhibition, but mutating both sites together completely abolished Gem inhibition without affecting Gem protein expression level or disrupting Gem interaction with Ca(v)2.1 or Ca(v)β. Mutating Gem residues that are crucial for interactions with previously demonstrated RGK modulators such as calmodulin, 14-3-3, and phosphatidylinositol lipids did not significantly affect Gem inhibition. These results suggest that Gem contains two candidate inhibitory sites, each capable of producing full inhibition of P/Q-type Ca(2+) channels.
- Published
- 2012
- Full Text
- View/download PDF
47. Discovery and evaluation of selective N-type calcium channel blockers: 6-unsubstituted-1,4-dihydropyridine-5-carboxylic acid derivatives.
- Author
-
Yamamoto T, Niwa S, Tokumasu M, Onishi T, Ohno S, Hagihara M, Koganei H, Fujita S, Takeda T, Saitou Y, Iwayama S, Takahara A, Iwata S, and Shoji M
- Subjects
- Administration, Oral, Analgesics chemical synthesis, Analgesics pharmacology, Animals, Calcium Channel Blockers chemical synthesis, Calcium Channel Blockers pharmacology, Calcium Channels, N-Type metabolism, Carboxylic Acids chemical synthesis, Carboxylic Acids pharmacology, Drug Evaluation, Preclinical, Formaldehyde toxicity, Pain Measurement drug effects, Rats, Structure-Activity Relationship, Analgesics chemistry, Calcium Channel Blockers chemistry, Calcium Channels, N-Type chemistry, Carboxylic Acids chemistry, Dihydropyridines chemistry
- Abstract
A structure-activity relationship study of 6-unsubstituted-1,4-dihydropyridine and 2,6-unsubstituted-1,4-dihydropyridine derivatives was conducted in an attempt to discover N-type calcium channel blockers that were highly selective over L-type calcium channel blockers. Among the tested compounds, (+)-4-(3,5-dichloro-4-methoxy-phenyl)-1,4-dihydro-pyridine-3,5-dicarboxylic acid 3-cinnamyl ester was found to be an effective and selective N-type calcium channel blocker with oral analgesic potential., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
48. Nanodomain Ca²⁺ of Ca²⁺ channels detected by a tethered genetically encoded Ca²⁺ sensor.
- Author
-
Tay LH, Dick IE, Yang W, Mank M, Griesbeck O, and Yue DT
- Subjects
- Cytoplasm chemistry, Cytoplasm metabolism, Electrophysiology, Fluorescence Resonance Energy Transfer, Genetic Engineering, HEK293 Cells, Humans, Kinetics, Calcium metabolism, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type genetics, Calcium Channels, N-Type metabolism
- Abstract
Coupling of excitation to secretion, contraction and transcription often relies on Ca(2+) computations within the nanodomain-a conceptual region extending tens of nanometers from the cytoplasmic mouth of Ca(2+) channels. Theory predicts that nanodomain Ca(2+) signals differ vastly from the slow submicromolar signals routinely observed in bulk cytoplasm. However, direct visualization of nanodomain Ca(2+) far exceeds optical resolution of spatially distributed Ca(2+) indicators. Here we couple an optical, genetically encoded Ca(2+) indicator (TN-XL) to the carboxy tail of Ca(V)2.2 Ca(2+) channels, enabling near-field imaging of the nanodomain. Under total internal reflection fluorescence microscopy, we detect Ca(2+) responses indicative of large-amplitude pulses. Single-channel electrophysiology reveals a corresponding Ca(2+) influx of only 0.085 pA, and fluorescence resonance energy transfer measurements estimate TN-XL distance to the cytoplasmic mouth at ~55 Å. Altogether, these findings raise the possibility that Ca(2+) exits the channel through the analogue of molecular portals, mirroring the crystallographic images of side windows in voltage-gated K channels.
- Published
- 2012
- Full Text
- View/download PDF
49. Domain III regulates N-type (CaV2.2) calcium channel closing kinetics.
- Author
-
Yarotskyy V, Gao G, Peterson BZ, and Elmslie KS
- Subjects
- Animals, Biophysical Phenomena drug effects, Biophysical Phenomena genetics, Calcium Channels, N-Type genetics, Electric Stimulation, HEK293 Cells, Humans, Ion Channel Gating drug effects, Ion Channel Gating genetics, Membrane Potentials drug effects, Membrane Potentials genetics, Mutant Chimeric Proteins genetics, Patch-Clamp Techniques, Protein Kinase Inhibitors pharmacology, Protein Structure, Tertiary, Protein Subunits genetics, Protein Subunits metabolism, Purines pharmacology, Rabbits, Roscovitine, Transfection, Biophysical Phenomena physiology, Calcium Channels, N-Type chemistry, Calcium Channels, N-Type physiology, Ion Channel Gating physiology
- Abstract
Ca(V)2.2 (N-type) and Ca(V)1.2 (L-type) calcium channels gate differently in response to membrane depolarization, which is critical to the unique physiological functions mediated by these channels. We wondered if the source for these differences could be identified. As a first step, we examined the effect of domain exchange between N-type and L-type channels on activation-deactivation kinetics, which were significantly different between these channels. Kinetic analysis of chimeric channels revealed N-channel-like deactivation for all chimeric channels containing N-channel domain III, while activation appeared to be a more distributed function across domains. This led us to hypothesize that domain III was an important regulator of N-channel closing. This idea was further examined with R-roscovitine, which is a trisubstituted purine that slows N-channel deactivation by exclusively binding to activated N-channels. L-channels lack this response to roscovitine, which allowed us to use N-L chimeras to test the role of domain III in roscovitine modulation of N-channel deactivation. In support of our hypothesis, all chimeric channels containing the N-channel domain III responded to roscovitine with slowed deactivation, while those chimeric channels with L-channel domain III did not. Thus a combination of kinetic and pharmacological evidence supports the hypothesis that domain III is an important regulator of N-channel closing. Our results support specialization of gating functions among calcium channel domains.
- Published
- 2012
- Full Text
- View/download PDF
50. Molecular and biophysical basis of glutamate and trace metal modulation of voltage-gated Ca(v)2.3 calcium channels.
- Author
-
Shcheglovitov A, Vitko I, Lazarenko RM, Orestes P, Todorovic SM, and Perez-Reyes E
- Subjects
- 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, Calcium Channels, R-Type metabolism, Cation Transport Proteins metabolism
- 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., (© 2012 Shcheglovitov et al.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.