Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Biochim Biophys Acta Biomembr ; 1866(6): 184337, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38763272

ABSTRACT

Ca2+ influx through Cav3.3 T-type channel plays crucial roles in neuronal excitability and is subject to regulation by various signaling molecules. However, our understanding of the partners of Cav3.3 and the related regulatory pathways remains largely limited. To address this quest, we employed the rat Cav3.3 C-terminus as bait in yeast-two-hybrid screenings of a cDNA library, identifying rat Gß2 as an interaction partner. Subsequent assays revealed that the interaction of Gß2 subunit was specific to the Cav3.3 C-terminus. Through systematic dissection of the C-terminus, we pinpointed a 22 amino acid sequence (amino acids 1789-1810) as the Gß2 interaction site. Coexpression studies of rat Cav3.3 with various Gßγ compositions were conducted in HEK-293 cells. Patch clamp recordings revealed that coexpression of Gß2γ2 reduced Cav3.3 current density and accelerated inactivation kinetics. Interestingly, the effects were not unique to Gß2γ2, but were mimicked by Gß2 alone as well as other Gßγ dimers, with similar potencies. Deletion of the Gß2 interaction site abolished the effects of Gß2γ2. Importantly, these Gß2 effects were reproduced in human Cav3.3. Overall, our findings provide evidence that Gß(γ) complexes inhibit Cav3.3 channel activity and accelerate the inactivation kinetics through the Gß interaction with the Cav3.3 C-terminus.


Subject(s)
Calcium Channels, T-Type , GTP-Binding Protein beta Subunits , Animals , Humans , Rats , Calcium Channels, R-Type , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/chemistry , Cation Transport Proteins , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/chemistry , HEK293 Cells , Kinetics , Patch-Clamp Techniques , Protein Binding
3.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731963

ABSTRACT

Venom peptides have evolved to target a wide range of membrane proteins through diverse mechanisms of action and structures, providing promising therapeutic leads for diseases, including pain, epilepsy, and cancer, as well as unique probes of ion channel structure-function. In this work, a high-throughput FLIPR window current screening assay on T-type CaV3.2 guided the isolation of a novel peptide named ω-Buthitoxin-Hf1a from scorpion Hottentotta franzwerneri crude venom. At only 10 amino acid residues with one disulfide bond, it is not only the smallest venom peptide known to target T-type CaVs but also the smallest structured scorpion venom peptide yet discovered. Synthetic Hf1a peptides were prepared with C-terminal amidation (Hf1a-NH2) or a free C-terminus (Hf1a-OH). Electrophysiological characterization revealed Hf1a-NH2 to be a concentration-dependent partial inhibitor of CaV3.2 (IC50 = 1.18 µM) and CaV3.3 (IC50 = 0.49 µM) depolarized currents but was ineffective at CaV3.1. Hf1a-OH did not show activity against any of the three T-type subtypes. Additionally, neither form showed activity against N-type CaV2.2 or L-type calcium channels. The three-dimensional structure of Hf1a-NH2 was determined using NMR spectroscopy and used in docking studies to predict its binding site at CaV3.2 and CaV3.3. As both CaV3.2 and CaV3.3 have been implicated in peripheral pain signaling, the analgesic potential of Hf1a-NH2 was explored in vivo in a mouse model of incision-induced acute post-surgical pain. Consistent with this role, Hf1a-NH2 produced antiallodynia in both mechanical and thermal pain.


Subject(s)
Calcium Channels, T-Type , Disease Models, Animal , Hyperalgesia , Pain, Postoperative , Scorpion Venoms , Animals , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/chemistry , Mice , Scorpion Venoms/chemistry , Scorpion Venoms/pharmacology , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Pain, Postoperative/drug therapy , Pain, Postoperative/metabolism , Calcium/metabolism , Male , Humans , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/chemistry
4.
Cell Res ; 34(6): 440-450, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38605177

ABSTRACT

The Cav3.2 subtype of T-type calcium channels has been targeted for developing analgesics and anti-epileptics for its role in pain and epilepsy. Here we present the cryo-EM structures of Cav3.2 alone and in complex with four T-type calcium channel selective antagonists with overall resolutions ranging from 2.8 Å to 3.2 Å. The four compounds display two binding poses. ACT-709478 and TTA-A2 both place their cyclopropylphenyl-containing ends in the central cavity to directly obstruct ion flow, meanwhile extending their polar tails into the IV-I fenestration. TTA-P2 and ML218 project their 3,5-dichlorobenzamide groups into the II-III fenestration and place their hydrophobic tails in the cavity to impede ion permeation. The fenestration-penetrating mode immediately affords an explanation for the state-dependent activities of these antagonists. Structure-guided mutational analysis identifies several key residues that determine the T-type preference of these drugs. The structures also suggest the role of an endogenous lipid in stabilizing drug binding in the central cavity.


Subject(s)
Calcium Channel Blockers , Calcium Channels, T-Type , Cryoelectron Microscopy , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/chemistry , Humans , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Binding Sites , Protein Binding , Models, Molecular , HEK293 Cells
5.
Handb Exp Pharmacol ; 279: 263-288, 2023.
Article in English | MEDLINE | ID: mdl-36592228

ABSTRACT

CaV3.3 is the third member of the low-voltage-activated calcium channel family and the last to be recognized as disease gene. Previously, CACNA1I, the gene encoding CaV3.3, had been described as schizophrenia risk gene. More recently, de novo missense mutations in CACNA1I were identified in patients with variable degrees of neurodevelopmental disease with and without epilepsy. Their functional characterization indicated gain-of-function effects resulting in increased calcium load and hyperexcitability of neurons expressing CaV3.3. The amino acids mutated in the CaV3.3 disease variants are located in the vicinity of the channel's activation gate and thus are classified as gate-modifying channelopathy mutations. A persistent calcium leak during rest and prolonged calcium spikes due to increased voltage sensitivity of activation and slowed kinetics of channel inactivation, respectively, may be causal for the neurodevelopmental defects. The prominent expression of CaV3.3 in thalamic reticular nucleus neurons and its essential role in generating the rhythmic thalamocortical network activity are consistent with a role of the mutated channels in the etiology of epileptic seizures and thus suggest T-type channel blockers as a viable treatment option.


Subject(s)
Calcium Channels, T-Type , Channelopathies , Humans , Channelopathies/genetics , Calcium/metabolism , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Mutation
6.
J Biol Chem ; 298(12): 102621, 2022 12.
Article in English | MEDLINE | ID: mdl-36272643

ABSTRACT

Cav3 T-type calcium channels from great pond snail Lymnaea stagnalis have a selectivity-filter ring of five acidic residues, EE(D)DD. Splice variants with exons 12b or 12a spanning the extracellular loop between the outer helix IIS5 and membrane-descending pore helix IIP1 (IIS5-P1) in Domain II of the pore module possess calcium selectivity or dominant sodium permeability, respectively. Here, we use AlphaFold2 neural network software to predict that a lysine residue in exon 12a is salt-bridged to the aspartate residue immediately C terminal to the second-domain glutamate in the selectivity filter. Exon 12b has a similar folding but with an alanine residue in place of lysine in exon 12a. We express LCav3 channels with mutated exons Ala-12b-Lys and Lys-12a-Ala and demonstrate that they switch the ion preference to high sodium permeability and calcium selectivity, respectively. We propose that in the calcium-selective variants, a calcium ion chelated between Domain II selectivity-filter glutamate and aspartate is knocked-out by the incoming calcium ion in the process of calcium permeation, whereas sodium ions are repelled. The aspartate is neutralized by the lysine residue in the sodium-permeant variants, allowing for sodium permeation through the selectivity-filter ring of four negatively charged residues akin to the prokaryotic sodium channels with four glutamates in the selectivity filter. The evolutionary adaptation in invertebrate LCav3 channels highlight the involvement of a key, ubiquitous aspartate, "a calcium beacon" of sorts in the outer pore of Domain II, as determinative for the calcium ion preference over sodium ions through eukaryotic Cav1, Cav2, and Cav3 channels.


Subject(s)
Calcium Channels, T-Type , Calcium , Lysine , Sodium , Aspartic Acid , Calcium/chemistry , Glutamic Acid , Ions , Lysine/chemistry , Sodium/chemistry , Lymnaea , Animals , Calcium Channels, T-Type/chemistry
7.
Molecules ; 26(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34770935

ABSTRACT

Catharanthus roseus is a well-known traditional herbal medicine for the treatment of cancer, hypertension, scald, and sore in China. Phytochemical investigation on the twigs and leaves of this species led to the isolation of two new monoterpene indole alkaloids, catharanosines A (1) and B (2), and six known analogues (3-8). Structures of 1 and 2 were established by 1H-, 13C- and 2D-NMR, and HREIMS data. The absolute configuration of 1 was confirmed by single-crystal X-ray diffraction analysis. Compound 2 represented an unprecedented aspidosperma-type alkaloid with a 2-piperidinyl moiety at C-10. Compounds 6-8 exhibited remarkable Cav3.1 low voltage-gated calcium channel (LVGCC) inhibitory activity with IC50 values of 11.83 ± 1.02, 14.3 ± 1.20, and 14.54 ± 0.99 µM, respectively.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/chemistry , Catharanthus/chemistry , Indole Alkaloids/pharmacology , Monoterpenes/pharmacology , Plant Extracts/pharmacology , Calcium Channel Blockers/chemistry , Calcium Channels, T-Type/metabolism , Dose-Response Relationship, Drug , Indole Alkaloids/chemistry , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Monoterpenes/chemistry , Plant Extracts/chemistry , Structure-Activity Relationship
8.
Pharmacol Res Perspect ; 9(3): e00783, 2021 05.
Article in English | MEDLINE | ID: mdl-33984189

ABSTRACT

Pharmaceutical features of phenylalkylamine derivatives (PAAs) binding to calcium channels have been studied extensively in the past decades. Only a few PAAs have the binding specificity on calcium channels, for example, NNC 55-0396. Here, we created the homology models of human Cav 3.2, Cav 3.3 and use them as a receptor on the rigid docking tests. The nonspecific calcium channel blocker mibefradil showed inconsistent docking preference across four domains; however, NNC 55-0396 had a unique binding pattern on domain II specifically. The subsequent molecular dynamics (MD) simulations identified that Cav 3.1, Cav 3.2, and Cav 3.3 share domain II when Ca2+ appearing in the neighbor region of selective filters (SFs). Moreover, free-energy perturbation analysis suggests single mutation of lysine at P-loop domain III, or threonine at the P-loop domain II largely reduced the total amount of hydration-free energy in the system. All these findings suggest that P-loop and segment six domain II in the T-type calcium channels (TCCs) are crucial for attracting the PAAs with specificity as the antagonist.


Subject(s)
Benzimidazoles/chemistry , Calcium Channel Blockers/chemistry , Calcium Channels, T-Type/chemistry , Cyclopropanes/chemistry , Mibefradil/chemistry , Models, Molecular , Naphthalenes/chemistry , Humans
9.
Transl Res ; 234: 20-30, 2021 08.
Article in English | MEDLINE | ID: mdl-33422652

ABSTRACT

T-type calcium channels regulate neuronal excitability and are important contributors of pain processing. CaV3.2 channels are the major isoform expressed in nonpeptidergic and peptidergic nociceptive neurons and are emerging as promising targets for pain treatment. Numerous studies have shown that CaV3.2 expression and/or activity are significantly increased in spinal dorsal horn and in dorsal root ganglia neurons in different inflammatory and neuropathic pain models. Pharmacological campaigns to inhibit the functional expression of CaV3.2 for treatment of pain have focused on the development of direct channel blockers, but none have produced lead candidates. Targeting the proteins that regulate the trafficking or transcription, and the ones that modify the channels via post-translational modifications are alternative means to regulate expression and function of CaV3.2 channels and hence to develop new drugs to control pain. Here we synthesize data supporting a role for CaV3.2 in numerous pain modalities and then discuss emerging opportunities for the indirect targeting of CaV3.2 channels.


Subject(s)
Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/physiology , Chronic Pain/drug therapy , Animals , Biophysical Phenomena , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Chronic Pain/physiopathology , Disease Models, Animal , Drug Development , Ganglia, Spinal/physiopathology , Humans , Models, Molecular , Neuralgia/drug therapy , Neuralgia/physiopathology , Protein Processing, Post-Translational/drug effects , Spinal Cord Dorsal Horn/physiopathology , Transcription, Genetic/drug effects , Translational Research, Biomedical
10.
Mol Brain ; 13(1): 149, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33176830

ABSTRACT

Low-voltage-activated T-type calcium channels are important contributors to nervous system function. Post-translational modification of these channels has emerged as an important mechanism to control channel activity. Previous studies have documented the importance of asparagine (N)-linked glycosylation and identified several asparagine residues within the canonical consensus sequence N-X-S/T that is essential for the expression and function of Cav3.2 channels. Here, we explored the functional role of non-canonical N-glycosylation motifs in the conformation N-X-C based on site directed mutagenesis. Using a combination of electrophysiological recordings and surface biotinylation assays, we show that asparagines N345 and N1780 located in the motifs NVC and NPC, respectively, are essential for the expression of the human Cav3.2 channel in the plasma membrane. Therefore, these newly identified asparagine residues within non-canonical motifs add to those previously reported in canonical sites and suggest that N-glycosylation of Cav3.2 may also occur at non-canonical motifs to control expression of the channel in the plasma membrane. It is also the first study to report the functional importance of non-canonical N-glycosylation motifs in an ion channel.


Subject(s)
Calcium Channels, T-Type/metabolism , Amino Acid Motifs , Asparagine/metabolism , Calcium Channels, T-Type/chemistry , Glycosylation , Humans , Structure-Activity Relationship
11.
J Physiol Sci ; 70(1): 49, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33059597

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 Ca2+ 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.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Calcium Channels, T-Type/metabolism , Neurons/metabolism , Vasopressins/metabolism , 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
12.
Bioelectrochemistry ; 136: 107618, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32795940

ABSTRACT

A family of current-time curves of T-type Cav3.1 Ca2+ channels available in the literature is simulated by a kinetic model differing from that used for the interpretation of all salient features of Na+ and Shaker K+ channels by the insertion of a multiplying factor expressing the difference between the working potential ϕ and the reversal potential ϕr. This deterministic model is also used to simulate experimental curves taken from the literature for steady-state 'fast inactivation' and for a gradual passage from fast to 'slow inactivation'. A depolarizing pulse induces fast or slow inactivation depending on whether it lasts 100-500 ms or about 1 min, and is believed to cause a collapse of the central pore near the selectivity filter (SF). A number of features of fast and slow inactivation of Cav3.1 Ca2+ channels are qualitatively interpreted on the basis of a sequence of conformational states. Briefly, the conformation responsible for 'fast inactivation' is assumed to have the activation gate open and the inactivation gate (i.e., the SF) inactive. Immediately after a depolarizing pulse, this conformation is inactive and requires a sufficiently long rest time at a far negative holding potential to recover from inactivation. 'Slow inactivation' is ascribed to a different conformation with the activation gate closed and the SF inactive.


Subject(s)
Calcium Channels, T-Type/chemistry , Models, Molecular , Calcium Channels, T-Type/physiology , Humans , Ion Channel Gating , Protein Conformation
13.
Mol Brain ; 13(1): 95, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32560664

ABSTRACT

Low-voltage-activated Cav3 calcium channels (T-type) play an essential role in the functioning of the nervous system where they support oscillatory activities that relie on several channel molecular determinants that shape their unique gating properties. In a previous study, we documented the important role of the carboxy proximal region in the functioning of Cav3.3 channels. Here, we explore the ability of a TAT-based cell penetrating peptide containing this carboxy proximal region (TAT-C3P) to modulate the activity of Cav3 channels. We show that chronic application of TAT-C3P on tsA-201 cells expressing Cav3 channels selectively inhibits Cav3.3 channels without affecting Cav3.1 and Cav3.2 channels. Therefore, the TAT-C3P peptide described in this study represents a new tool to address the specific physiological role of Cav3.3 channels, and to potentially enhance our understanding of Cav3.3 in disease.


Subject(s)
Calcium Channels, T-Type/metabolism , Neurons/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , Calcium Channels, T-Type/chemistry , Cell Line , Humans , Models, Molecular
14.
Cell Calcium ; 89: 102214, 2020 07.
Article in English | MEDLINE | ID: mdl-32428730

ABSTRACT

There is growing evidence indicating that the pore structure of voltage-gated ion channels (VGICs) influences gating besides their conductance. Regarding low voltage-activated (LVA) Ca2+ channels, it has been demonstrated that substitutions of the pore aspartate (D) by a glutamate (D-to-E substitution) in domains III and IV alter channel gating properties such as a positive shift in the channel activation voltage dependence. In the present report, we evaluated the effects of E-to-D substitution in domains I and II on the CaV3.1 channel gating properties. Our results indicate that substitutions in these two domains differentially modify the gating properties of CaV3.1 channels. The channel with a single mutation in domain I (DEDD) presented slower activation and faster inactivation kinetics and a slower recovery from inactivation, as compared with the WT channel. In contrast, the single mutant in domain II (EDDD) presented a small but significant negative shift of activation voltage dependence with faster activation and slower inactivation kinetics. Finally, the double mutant channel (DDDD) presented somehow intermediate properties with respect to the two single mutants but with fastest deactivation kinetics. Overall, our results indicate that single amino acid modification of the selectivity filter of LVA Ca2+ channels in distinct domains differentially influence their gating properties, supporting a pore pseudo-symmetry.


Subject(s)
Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Mutation/genetics , Glutamic Acid/metabolism , HEK293 Cells , Humans , Ion Channel Gating , Kinetics , Markov Chains , Models, Molecular , Mutant Proteins/chemistry , Probability , Protein Domains
15.
J Biol Chem ; 295(18): 6177-6186, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32188693

ABSTRACT

T-type (Cav3) Ca2+ channels are important regulators of excitability and rhythmic activity of excitable cells. Among other voltage-gated Ca2+ channels, Cav3 channels are uniquely sensitive to oxidation and zinc. Using recombinant protein expression in HEK293 cells, patch clamp electrophysiology, site-directed mutagenesis, and homology modeling, we report here that modulation of Cav3.2 by redox agents and zinc is mediated by a unique extracellular module containing a high-affinity metal-binding site formed by the extracellular IS1-IS2 and IS3-IS4 loops of domain I and a cluster of extracellular cysteines in the IS1-IS2 loop. Patch clamp recording of recombinant Cav3.2 currents revealed that two cysteine-modifying agents, sodium (2-sulfonatoethyl) methanethiosulfonate (MTSES) and N-ethylmaleimide, as well as a reactive oxygen species-producing neuropeptide, substance P (SP), inhibit Cav3.2 current to similar degrees and that this inhibition is reversed by a reducing agent and a zinc chelator. Pre-application of MTSES prevented further SP-mediated current inhibition. Substitution of the zinc-binding residue His191 in Cav3.2 reduced the channel's sensitivity to MTSES, and introduction of the corresponding histidine into Cav3.1 sensitized it to MTSES. Removal of extracellular cysteines from the IS1-IS2 loop of Cav3.2 reduced its sensitivity to MTSES and SP. We hypothesize that oxidative modification of IS1-IS2 loop cysteines induces allosteric changes in the zinc-binding site of Cav3.2 so that it becomes sensitive to ambient zinc.


Subject(s)
Calcium Channels, T-Type/metabolism , Extracellular Space/metabolism , Calcium Channels, T-Type/chemistry , HEK293 Cells , Humans , Models, Molecular , Oxidation-Reduction , Protein Conformation
16.
Mol Brain ; 13(1): 33, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32143681

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the progressive loss of cortical, brain stem and spinal motor neurons that leads to muscle weakness and death. A previous study implicated CACNA1H encoding for Cav3.2 calcium channels as a susceptibility gene in ALS. In the present study, two heterozygous CACNA1H variants were identified by whole genome sequencing in a small cohort of ALS patients. These variants were functionally characterized using patch clamp electrophysiology, biochemistry assays, and molecular modeling. A previously unreported c.454GTAC > G variant produced an inframe deletion of a highly conserved isoleucine residue in Cav3.2 (p.ΔI153) and caused a complete loss-of-function of the channel, with an additional dominant-negative effect on the wild-type channel when expressed in trans. In contrast, the c.3629C > T variant caused a missense substitution of a proline with a leucine (p.P1210L) and produced a comparatively mild alteration of Cav3.2 channel activity. The newly identified ΔI153 variant is the first to be reported to cause a complete loss of Cav3.2 channel function. These findings add to the notion that loss-of-function of Cav3.2 channels associated with rare CACNA1H variants may be risk factors in the complex etiology of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Calcium Channels, T-Type/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation/genetics , Amino Acid Sequence , Animals , Calcium Channels, T-Type/chemistry , Genes, Dominant , Heterozygote , Male , Rats , Structural Homology, Protein , Whole Genome Sequencing
17.
Nature ; 576(7787): 492-497, 2019 12.
Article in English | MEDLINE | ID: mdl-31766050

ABSTRACT

Among the ten subtypes of mammalian voltage-gated calcium (Cav) channels, Cav3.1-Cav3.3 constitute the T-type, or the low-voltage-activated, subfamily, the abnormal activities of which are associated with epilepsy, psychiatric disorders and pain1-5. Here we report the cryo-electron microscopy structures of human Cav3.1 alone and in complex with a highly Cav3-selective blocker, Z9446,7, at resolutions of 3.3 Å and 3.1 Å, respectively. The arch-shaped Z944 molecule reclines in the central cavity of the pore domain, with the wide end inserting into the fenestration on the interface between repeats II and III, and the narrow end hanging above the intracellular gate like a plug. The structures provide the framework for comparative investigation of the distinct channel properties of different Cav subfamilies.


Subject(s)
Apoproteins/chemistry , Apoproteins/ultrastructure , Calcium Channel Blockers/chemistry , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/ultrastructure , Cryoelectron Microscopy , Piperidines/chemistry , Allosteric Regulation/drug effects , Amino Acid Motifs , Amino Acid Sequence , Apoproteins/genetics , Binding Sites , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/genetics , Humans , Models, Molecular , Piperidines/pharmacology , Protein Binding , Protein Conformation , Sequence Deletion
18.
Bioorg Chem ; 91: 103187, 2019 10.
Article in English | MEDLINE | ID: mdl-31419643

ABSTRACT

1,4-Dihydropyridines (DHPs) are an important class of blockers targeting different calcium channel subtypes and have great therapeutic value against cardiovascular and neurophysiologic conditions. Here, we present the design of DHP-based hexahydroquinoline derivatives as either selective or covalent inhibitors of calcium channels. These compounds were synthesized via a modified Hantzsch reaction under microwave irradiation and characterized by IR, 1H NMR, 13C NMR and mass spectra. Additionally, the proposed structure of HM12 was resolved by single crystal X-ray analysis. The abilities of the target compounds to block both L- and T-type calcium channels were evaluated by utilizing the whole-cell patch clamp technique. Our results identified covalent inhibitors of calcium channels for the first time, which could be achieved by introducing a Michael acceptor group into the ester side chain of the compounds. The proposed covalent binding between the compounds and the cysteine amino acid (Cys1492) within the DHP binding pocket of L-type calcium channel was supported by docking and pharmacophore analysis as well as a glutathione reactivity assay.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/chemistry , Calcium Channels, T-Type/chemistry , Dihydropyridines/pharmacology , Drug Discovery , Glutathione/metabolism , Binding Sites , Calcium/metabolism , Cysteine/chemistry , Cysteine/metabolism , Humans , Models, Molecular , Protein Conformation
19.
Channels (Austin) ; 13(1): 153-161, 2019 12.
Article in English | MEDLINE | ID: mdl-31070086

ABSTRACT

Neuromuscular disorders encompass a wide range of conditions often associated with a genetic component. In the present study, we report a patient with severe infantile-onset amyotrophy in whom two compound heterozygous variants in the gene CACNA1H encoding for Cav3.2 T-type calcium channels were identified. Functional analysis of Cav3.2 variants revealed several alterations of the gating properties of the channel that were in general consistent with a loss-of-channel function. Taken together, these findings suggest that severe congenital amyoplasia may be related to CACNA1H and would represent a new phenotype associated with mutations in this gene.


Subject(s)
Brachial Plexus Neuritis/genetics , Calcium Channels, T-Type/genetics , Mutation, Missense , Brachial Plexus Neuritis/physiopathology , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/physiology , Electrophysiology , Female , Heterozygote , Humans , Infant , Phenotype , Exome Sequencing
20.
Trends Mol Med ; 25(7): 571-584, 2019 07.
Article in English | MEDLINE | ID: mdl-31031178

ABSTRACT

T-Type calcium channels (TTCCs) are key regulators of membrane excitability, which is the reason why TTCC pharmacology is subject to intensive research in the neurological and cardiovascular fields. TTCCs also play a role in cancer physiology, and pharmacological blockers such as tetralols and dihydroquinazolines (DHQs) reduce the viability of cancer cells in vitro and slow tumor growth in murine xenografts. However, the available compounds are better suited to blocking TTCCs in excitable membranes rather than TTCCs contributing window currents at steady potentials. Consistently, tetralols and dihydroquinazolines exhibit cytostatic/cytotoxic activities at higher concentrations than those required for TTCC blockade, which may involve off-target effects. Gene silencing experiments highlight the targetability of TTCCs, but further pharmacological research is required for TTCC blockade to become a chemotherapeutic option.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Ion Channel Gating , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/chemistry , Cell Survival/drug effects , Disease Susceptibility , Gene Knockdown Techniques , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...