Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Neurosci ; 38(43): 9186-9201, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30181139

ABSTRACT

The putative cache (Ca2+ channel and chemotaxis receptor) domain containing 1 (CACHD1) protein has predicted structural similarities to members of the α2δ voltage-gated Ca2+ channel auxiliary subunit family. CACHD1 mRNA and protein were highly expressed in the male mammalian CNS, in particular in the thalamus, hippocampus, and cerebellum, with a broadly similar tissue distribution to CaV3 subunits, in particular CaV3.1. In expression studies, CACHD1 increased cell-surface localization of CaV3.1, and these proteins were in close proximity at the cell surface, consistent with the formation of CACHD1-CaV3.1 complexes. In functional electrophysiological studies, coexpression of human CACHD1 with CaV3.1, CaV3.2, and CaV3.3 caused a significant increase in peak current density and corresponding increases in maximal conductance. By contrast, α2δ-1 had no effect on peak current density or maximal conductance in CaV3.1, CaV3.2, or CaV3.3. A comparison of CACHD1-mediated increases in CaV3.1 current density and gating currents revealed an increase in channel open probability. In hippocampal neurons from male and female embryonic day 19 rats, CACHD1 overexpression increased CaV3-mediated action potential firing frequency and neuronal excitability. These data suggest that CACHD1 is structurally an α2δ-like protein that functionally modulates CaV3 voltage-gated calcium channel activity.SIGNIFICANCE STATEMENT This is the first study to characterize the Ca2+ channel and chemotaxis receptor domain containing 1 (CACHD1) protein. CACHD1 is widely expressed in the CNS, in particular in the thalamus, hippocampus, and cerebellum. CACHD1 distribution is similar to that of low voltage-activated (CaV3, T-type) calcium channels, in particular to CaV3.1, a protein that regulates neuronal excitability and is a potential therapeutic target in conditions such as epilepsy and pain. CACHD1 is structurally an α2δ-like protein that functionally increases CaV3 calcium current. CACHD1 increases the presence of CaV3.1 at the cell surface, forms complexes with CaV3.1 at the cell surface, and causes an increase in channel open probability. In hippocampal neurons, CACHD1 causes increases in neuronal firing. Thus, CACHD1 represents a novel protein that modulates CaV3 activity.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/biosynthesis , Hippocampus/metabolism , Membrane Proteins/metabolism , Animals , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Female , HEK293 Cells , Humans , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Rats , Rats, Wistar
8.
PLoS Comput Biol ; 14(11): e1006485, 2018 11.
Article in English | MEDLINE | ID: mdl-30383745

ABSTRACT

Microdomains that emerge from spatially constricted spread of biochemical signaling components play a central role in several neuronal computations. Although dendrites, endowed with several voltage-gated ion channels, form a prominent structural substrate for microdomain physiology, it is not known if these channels regulate the spatiotemporal spread of signaling microdomains. Here, we employed a multiscale, morphologically realistic, conductance-based model of the hippocampal pyramidal neuron that accounted for experimental details of electrical and calcium-dependent biochemical signaling. We activated synaptic N-Methyl-d-Aspartate receptors through theta-burst stimulation (TBS) or pairing (TBP) and assessed microdomain propagation along a signaling pathway that included calmodulin, calcium/calmodulin-dependent protein kinase II (CaMKII) and protein phosphatase 1. We found that the spatiotemporal spread of the TBS-evoked microdomain in phosphorylated CaMKII (pCaMKII) was amplified in comparison to that of the corresponding calcium microdomain. Next, we assessed the role of two dendritically expressed inactivating channels, one restorative (A-type potassium) and another regenerative (T-type calcium), by systematically varying their conductances. Whereas A-type potassium channels suppressed the spread of pCaMKII microdomains by altering the voltage response to TBS, T-type calcium channels enhanced this spread by modulating TBS-induced calcium influx without changing the voltage. Finally, we explored cross-dependencies of these channels with other model components, and demonstrated the heavy mutual interdependence of several biophysical and biochemical properties in regulating microdomains and their spread. Our conclusions unveil a pivotal role for dendritic voltage-gated ion channels in actively amplifying or suppressing biochemical signals and their spatiotemporal spread, with critical implications for clustered synaptic plasticity, robust information transfer and efficient neural coding.


Subject(s)
Dendrites/physiology , Protein Domains , Signal Transduction/physiology , Action Potentials/physiology , Animals , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Humans , Ion Channel Gating , Phosphorylation , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Phosphatase 1/metabolism , Pyramidal Cells/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism
9.
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
10.
Bioorg Med Chem ; 26(15): 4410-4427, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30031654

ABSTRACT

Since 6-prenylnaringenin (6-PNG) was recently identified as a novel T-type calcium channel blocker with the IC50 value around 1 µM, a series of flavanone derivatives were designed, synthesized and subsequently evaluated for T-channel-blocking activity in HEK293 cells transfected with Cav3.2 T-type channels using a patch-clamp technique. As a result, several new flavanones blocked Cav3.2-dependent T-currents more potently than 6-PNG. In the synthesized compounds, 6-(3-ethylpent-2-enyl)-5,7-dihydroxy-2-(2-hydroxyphenyl)chroman-4-one 8j, 6-(3-ethylpent-2-enyl)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one 11b, 6-(2-cyclopentylideneethyl)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one 11d, and 6-(2-Cyclopentylethyl)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one 12c were more potent blocker than 6-PNG with the IC50 value of 0.39, 0.26, 0.46, and 0.50 µM, respectively. Among the above four derivatives, the compound 8j provided the best result in the in vivo experiments; i.e. systemic administration of 8j at the minimum dose completely restored neuropathic pain induced by partial sciatic nerve ligation in mice.


Subject(s)
Analgesics/chemical synthesis , Calcium Channel Blockers/chemical synthesis , Calcium Channels, T-Type/chemistry , Drug Design , Flavonoids/chemistry , Action Potentials/drug effects , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/metabolism , Disease Models, Animal , Flavonoids/pharmacology , Flavonoids/therapeutic use , HEK293 Cells , Humans , Inhibitory Concentration 50 , Male , Mice , Neuralgia/drug therapy , Patch-Clamp Techniques , Structure-Activity Relationship
11.
Bioorg Med Chem ; 26(11): 3046-3059, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29622412

ABSTRACT

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


Subject(s)
Benzimidazoles/chemical synthesis , Benzothiazoles/chemical synthesis , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/chemistry , Calcium Channels, T-Type/chemistry , Cyclopropanes/chemical synthesis , Naphthalenes/chemical synthesis , Piperidines/chemical synthesis , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/chemistry , Calcium Channels, N-Type/drug effects , Calcium Channels, T-Type/drug effects , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Molecular Structure , Naphthalenes/chemistry , Naphthalenes/pharmacology , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Structure-Activity Relationship
12.
Mol Pharmacol ; 92(3): 347-357, 2017 09.
Article in English | MEDLINE | ID: mdl-28696213

ABSTRACT

Calcium-dependent inactivation of high voltage-activated Ca2+ channels plays a crucial role in limiting rises in intracellular calcium (Ca2+i). A key mediator of these effects is calmodulin, which has been found to bind the C-terminus of the pore-forming α subunit. In contrast, little is known about how Ca2+i can regulate low voltage-activated T-type Ca2+ channels. Using whole cell patch clamp, we examined the biophysical properties of Ca2+ current through the three T-type Ca2+ channel isoforms, Cav3.1, Cav3.2, or Cav3.3, comparing internal solutions containing 27 nM and l µM free Ca2+ Both activation and inactivation kinetics of Cav3.3 current in l µM Ca2+i solution were more rapid than those in 27 nM Ca2+i solution. In addition, both activation and steady-state inactivation curves of Cav3.3 were negatively shifted in the higher Ca2+i solution. In contrast, the biophysical properties of Cav3.1 and Cav3.2 isoforms were not significantly different between the two internal solutions. Overexpression of CaM1234 (a calmodulin mutant that doesn't bind Ca2+) occluded the effects of l µM Ca2+i on Cav3.3, implying that CaM is involved in the Ca2+i regulation effects on Cav3.3. Yeast two-hybrid screening and co-immunoprecipitation experiments revealed a direct interaction of CaM with the carboxyl terminus of Cav3.3. Taken together, our results suggest that Cav3.3 T-type channel is potently regulated by Ca2+i via interaction of Ca2+/CaM with the carboxyl terminus of Cav3.3.


Subject(s)
Calcium Channels, T-Type/physiology , Calcium/physiology , Calmodulin/physiology , Animals , Calcium Channels, T-Type/chemistry , HEK293 Cells , Humans , Immunoprecipitation , Rats
13.
Bioorg Med Chem ; 25(17): 4677-4685, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28720324

ABSTRACT

A novel series of fused-benzensulfonamide 2-N-(pyrazol-3-yl)methylbenzo[d]isothiazole-1,1-dioxide derivatives was designed and synthesized as metabolically stable T-type calcium channel inhibitors. Several compounds, 9, 10, and 17, displayed potent T-type channel inhibitory activity. Among them, compounds 10 and 17 showed good metabolic stability in human liver microsomes, and low hERG channel and CYP450 inhibition. Compound 10 exhibited diabetic neuropathic pain-alleviating effects in a streptozotocin-induced peripheral diabetic neuropathy (PDN) model. The maximum efficacy of compound 10, which was 3-fold more potent than gabapentin, was observed at 1h after administration, and co-administration of compound 10 with gabapentin showed a considerable synergic effect.


Subject(s)
Calcium Channel Blockers/chemistry , Thiazoles/chemistry , Animals , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Diabetic Neuropathies/chemically induced , Diabetic Neuropathies/complications , Diabetic Neuropathies/drug therapy , Disease Models, Animal , Half-Life , Humans , Inhibitory Concentration 50 , Male , Microsomes, Liver/metabolism , Neuralgia/etiology , Neuralgia/prevention & control , Pyrazoles/chemistry , Rats , Structure-Activity Relationship , Thiazoles/pharmacokinetics , Thiazoles/therapeutic use
14.
Bioorg Med Chem ; 25(17): 4656-4664, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28720332

ABSTRACT

As a bioisosteric strategy to overcome the poor metabolic stability of lead compound KYS05090S, a series of new fluoro-substituted 3,4-dihydroquinazoline derivatives was prepared and evaluated for T-type calcium channel (Cav3.2) block, cytotoxic effects and liver microsomal stability. Among them, compound 8h (KCP10068F) containing 4-fluorobenzyl amide and 4-cyclohexylphenyl ring potently blocked Cav3.2 currents (>90% inhibition) at 10µM concentration and exhibited cytotoxic effect (IC50=5.9µM) in A549 non-small cell lung cancer cells that was comparable to KYS05090S. Furthermore, 8h showed approximately a 2-fold increase in liver metabolic stability in rat and human species compared to KYS05090S. Based on these overall results, 8h (KCP10068F) may therefore represent a good backup compound for KYS05090S for further biological investigations as novel cytotoxic agent. In addition, compound 8g (KCP10067F) was found to partially protect from inflammatory pain via a blockade of Cav3.2 channels.


Subject(s)
Analgesics/chemical synthesis , Calcium Channel Blockers/chemical synthesis , Quinazolines/chemistry , Quinidine/analogs & derivatives , A549 Cells , Analgesics/chemistry , Analgesics/toxicity , Animals , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/toxicity , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Cell Survival/drug effects , Drug Stability , Fluorine/chemistry , HEK293 Cells , Humans , Inhibitory Concentration 50 , Microsomes, Liver/metabolism , Patch-Clamp Techniques , Quinazolines/chemical synthesis , Quinazolines/toxicity , Quinidine/chemical synthesis , Quinidine/chemistry , Quinidine/toxicity , Rats
15.
J Biol Chem ; 290(26): 16168-76, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25931121

ABSTRACT

Voltage-dependent calcium channels (Cav) of the T-type family (Cav3.1, Cav3.2, and Cav3.3) are activated by low threshold membrane depolarization and contribute greatly to neuronal network excitability. Enhanced T-type channel activity, especially Cav3.2, contributes to disease states, including absence epilepsy. Interestingly, the intracellular loop connecting domains I and II (I-II loop) of Cav3.2 channels is implicated in the control of both surface expression and channel gating, indicating that this I-II loop plays an important regulatory role in T-type current. Here we describe that co-expression of this I-II loop or its proximal region (Δ1-Cav3.2; Ser(423)-Pro(542)) together with recombinant full-length Cav3.2 channel inhibited T-type current without affecting channel expression and membrane incorporation. Similar T-type current inhibition was obtained in NG 108-15 neuroblastoma cells that constitutively express Cav3.2 channels. Of interest, Δ1-Cav3.2 inhibited both Cav3.2 and Cav3.1 but not Cav3.3 currents. Efficacy of Δ1-Cav3.2 to inhibit native T-type channels was assessed in thalamic neurons using viral transduction. We describe that T-type current was significantly inhibited in the ventrobasal neurons that express Cav3.1, whereas in nucleus reticularis thalami neurons that express Cav3.2 and Cav3.3 channels, only the fast inactivating T-type current (Cav3.2 component) was significantly inhibited. Altogether, these data describe a new strategy to differentially inhibit Cav3 isoforms of the T-type calcium channels.


Subject(s)
Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Animals , Brain/metabolism , Calcium Channels, T-Type/genetics , Humans , Neurons/metabolism , Protein Structure, Secondary , Rats , Rats, Wistar
16.
Rev Physiol Biochem Pharmacol ; 167: 115-39, 2014.
Article in English | MEDLINE | ID: mdl-25280639

ABSTRACT

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


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Channels, R-Type/physiology , Calcium Channels, T-Type/physiology , Cation Transport Proteins/physiology , Death, Sudden/etiology , Epilepsy/physiopathology , Heart/physiology , Animals , Calcium Channels, L-Type/chemistry , Calcium Channels, R-Type/chemistry , Calcium Channels, T-Type/chemistry , Cation Transport Proteins/chemistry , Epilepsy/complications , Humans
17.
Bioorg Med Chem Lett ; 26(3): 1073-1079, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26739776

ABSTRACT

As a result of our continuous research, new 3,4-dihydroquinazoline derivative containing ureido group, KCP10043F was synthesized and evaluated for T-type Ca(2+) channel (Cav3.1) blockade, cytotoxicity, and cell cycle arrest against human non-small cell lung (A549) cells. KCP10043F showed both weaker T-type Ca(2+) channel blocking activity and less cytotoxicity against A549 cells than parent compound KYS05090S [4-(benzylcarbamoylmethyl)-3-(4-biphenylyl)-2-(N,N',N'-trimethyl-1,5-pentanediamino)-3,4-dihydroquinazoline 2 hydrochloride], but it exhibited more potent G1-phase arrest than KYS05090S in A549 cells. This was found to be accompanied by the downregulations of cyclin-dependent kinase (CDK) 2, CDK4, CDK6, cyclin D2, cyclin D3, and cyclin E at the protein levels. However, p27(KIP1) as a CDK inhibitor was gradually upregulated at the protein levels and increased recruitment to CDK2, CDK4 and CDK6 after KCP10043F treatment. Based on the strong G1-phase cell cycle arrest of KCP10043F in A549 cells, the combination of KCP10043F with etoposide (or cisplatin) resulted in a synergistic cell death (combination index=0.2-0.8) via the induction of apoptosis compared with either agent alone. Taken together with these overall results and the favorable in vitro ADME (absorption, distribution, metabolism, and excretion) profiles of KCP10043F, therefore, it could be used as a potential agent for the combination therapy on human lung cancer.


Subject(s)
Antineoplastic Agents/chemistry , Calcium Channel Blockers/chemistry , Calcium Channels, T-Type/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Dogs , Down-Regulation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Half-Life , Humans , Mice , Microsomes, Liver/metabolism , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Rats , Up-Regulation/drug effects
18.
Pflugers Arch ; 467(12): 2519-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26306541

ABSTRACT

Low-voltage-activated CaV3 channels are distinguished among other voltage-activated calcium channels by the most negative voltage activation threshold. The voltage dependence of current activation is virtually identical in all three CaV3 channels while the current kinetics of the CaV3.3 current is one order slower than that of the CaV3.1 and CaV3.2 channels. We have analyzed the voltage dependence and kinetics of charge (Q) movement in human recombinant CaV3.3 and CaV3.1 channels. The voltage dependence of voltage sensor activation (Qon-V) of the CaV3.3 channel was significantly shifted with respect to that of the CaV3.1 channel by +18.6 mV and the kinetic of Qon activation in the CaV3.3 channel was significantly slower than that of the CaV3.1 channel. Removal of the gating brake in the intracellular loop connecting repeats I and II in the CaV3.3 channel in the ID12 mutant channel shifted the Qon-V relation to a value even more negative than that for the CaV3.1 channel. The kinetic of Qon activation was not significantly different between ID12 and CaV3.1 channels. Deletion of the gating brake in the CaV3.1 channel resulted in a GD12 channel with the voltage dependence of the gating current activation significantly shifted toward more negative potentials. The Qon kinetic was not significantly altered. ID12 and GD12 mutants did not differ significantly in voltage dependence nor in the kinetic of voltage sensor activation. In conclusion, the putative gating brake in the intracellular loop connecting repeats I and II controls the gating current of the CaV3 channels. We suggest that activation of the voltage sensor in domain I is limiting both the voltage dependence and the kinetics of CaV3 channel activation.


Subject(s)
Calcium Channels, T-Type/metabolism , Ion Channel Gating , Calcium Channels, T-Type/chemistry , HEK293 Cells , Humans , Protein Structure, Tertiary
19.
Bioorg Med Chem ; 23(18): 6166-72, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26296911

ABSTRACT

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


Subject(s)
Amines/chemistry , Calcium Channel Blockers/chemistry , Amines/pharmacology , Amines/therapeutic use , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Male , Motor Activity/drug effects , Neuralgia/drug therapy , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy , Structure-Activity Relationship
20.
J Physiol ; 592(4): 795-809, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24277868

ABSTRACT

T-type calcium channels play essential roles in regulating neuronal excitability and network oscillations in the brain. Mutations in the gene encoding Cav3.2 T-type Ca(2+) channels, CACNA1H, have been found in association with various forms of idiopathic generalized epilepsy. We and others have found that these mutations may influence neuronal excitability either by altering the biophysical properties of the channels or by increasing their surface expression. The goals of the present study were to investigate the excitability of neurons expressing Cav3.2 with the epilepsy mutation, C456S, and to elucidate the mechanisms by which it influences neuronal properties. We found that expression of the recombinant C456S channels substantially increased the excitability of cultured neurons by increasing the spontaneous firing rate and reducing the threshold for rebound burst firing. Additionally, we found that molecular determinants in the I-II loop (the region in which most childhood absence epilepsy-associated mutations are found) substantially increase the surface expression of T-channels but do not alter the relative distribution of channels into dendrites of cultured hippocampal neurons. Finally, we discovered that expression of C456S channels promoted dendritic growth and arborization. These effects were reversed to normal by either the absence epilepsy drug ethosuximide or a novel T-channel blocker, TTA-P2. As Ca(2+)-regulated transcription factors also increase dendritic development, we tested a transactivator trap assay and found that the C456S variant can induce changes in gene transcription. Taken together, our findings suggest that gain-of-function mutations in Cav3.2 T-type Ca(2+) channels increase seizure susceptibility by directly altering neuronal electrical properties and indirectly by changing gene expression.


Subject(s)
Action Potentials , Calcium Channels, T-Type/metabolism , Hippocampus/physiopathology , Mutation, Missense , Neurons/physiology , Seizures/genetics , Animals , Anticonvulsants/pharmacology , Benzamides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Cells, Cultured , Ethosuximide/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Humans , Neurons/drug effects , Neurons/metabolism , Piperidines/pharmacology , Protein Structure, Tertiary , Protein Transport , Rats , Rats, Sprague-Dawley , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL