Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Mol Brain ; 14(1): 18, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33478561

ABSTRACT

Writer's cramp (WC) is a task-specific focal dystonia that occurs selectively in the hand and arm during writing. Previous studies have shown a role for genetics in the pathology of task-specific focal dystonia. However, to date, no causal gene has been reported for task-specific focal dystonia, including WC. In this study, we investigated the genetic background of a large Dutch family with autosomal dominant‒inherited WC that was negative for mutations in known dystonia genes. Whole exome sequencing identified 4 rare variants of unknown significance that segregated in the family. One candidate gene was selected for follow-up, Calcium Voltage-Gated Channel Subunit Alpha1 H, CACNA1H, due to its links with the known dystonia gene Potassium Channel Tetramerization Domain Containing 17, KCTD17, and with paroxysmal movement disorders. Targeted resequencing of CACNA1H in 82 WC cases identified another rare, putative damaging variant in a familial WC case that did not segregate. Using structural modelling and functional studies in vitro, we show that both the segregating p.Arg481Cys variant and the non-segregating p.Glu1881Lys variant very likely cause structural changes to the Cav3.2 protein and lead to similar gains of function, as seen in an accelerated recovery from inactivation. Both mutant channels are thus available for re-activation earlier, which may lead to an increase in intracellular calcium and increased neuronal excitability. Overall, we conclude that rare functional variants in CACNA1H need to be interpreted very carefully, and additional studies are needed to prove that the p.Arg481Cys variant is the cause of WC in the large Dutch family.


Subject(s)
Calcium Channels, T-Type/genetics , Dystonic Disorders/genetics , Genetic Predisposition to Disease , Mutation, Missense/genetics , Chromosome Segregation , Female , Humans , Male , Pedigree , Phenotype
2.
Nat Commun ; 11(1): 2755, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488011

ABSTRACT

Fragile X Syndrome results from a loss of Fragile X Mental Retardation Protein (FMRP). We now show that FMRP is a member of a Cav3-Kv4 ion channel complex that is known to regulate A-type potassium current in cerebellar granule cells to produce mossy fiber LTP. Mossy fiber LTP is absent in Fmr1 knockout (KO) mice but is restored by FMRP(1-297)-tat peptide. This peptide further rapidly permeates the blood-brain barrier to enter cells across the cerebellar-cortical axis that restores the balance of protein translation for at least 24 h and transiently reduces elevated levels of activity of adult Fmr1 KO mice in the Open Field Test. These data reveal that FMRP(1-297)-tat can improve function from the levels of protein translation to synaptic efficacy and behaviour in a model of Fragile X syndrome, identifying a potential therapeutic strategy for this genetic disorder.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Ion Channels/metabolism , Animals , Brain/pathology , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Male , Mice , Mice, Knockout , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurons/metabolism , Protein Biosynthesis
3.
Mol Brain ; 12(1): 86, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31651342

ABSTRACT

Two paternally-inherited missense variants in CACNA1H were identified and characterized in a 6-year-old child with generalized epilepsy. Febrile and unprovoked seizures were present in this child. Both variants were expressed in cis or isolation using human recombinant Cav3.2 calcium channels in tsA-201 cells. Whole-cell patch-clamp recordings indicated that one variant (c.3844C > T; p.R1282W) caused a significant increase in current density consistent with a pathogenic gain-of-function phenotype; while the other cis-related variant (c.5294C > T; p.A1765V) had a benign profile.


Subject(s)
Calcium Channels, T-Type/genetics , Epilepsy, Generalized/genetics , Mutation/genetics , Biophysical Phenomena , Child , Female , Humans , Infant , Infant, Newborn
4.
Mol Brain ; 12(1): 73, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455361

ABSTRACT

Cav3.2 calcium channels play a key role in nociceptive signaling in the primary afferent pain pathway. We have previously reported the regulation of Cav3.2 calcium channels by the deubiquitinase USP5 and its importance for regulating peripheral transmission of pain signals. Here we describe the regulation of the Cav3.2-USP5 interaction by SUMOylation. We show that endogenous USP5 protein expressed in dorsal root ganglia undergoes SUMOylation, and the level of USP5 SUMOylation is reduced following peripheral nerve injury. SUMO prediction software identified several putative lysines that have the propensity to be targets for SUMO conjugation. A series of single lysine substitutions in an mCherry tagged USP5 construct followed by expression in tsA-201 cells identified lysine K113 as a key target for USP5 SUMO2/3 modification. Finally, Cav3.2 calcium channel immunoprecipitates revealed a stronger interaction of Cav3.2 with a SUMO2/3 resistant USP5-K113R mutant, indicating that SUMO2/3 modification of USP5 reduces its affinity for the calcium channel Cav3.2. Collectively, our data suggest that dysregulation of USP5 SUMOylation after peripheral nerve injury may contribute to the well described alteration in Cav3.2 channel activity during neuropathic pain states.


Subject(s)
Calcium Channels, T-Type/metabolism , Endopeptidases/metabolism , Sumoylation , Ubiquitin-Specific Proteases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Endopeptidases/chemistry , Humans , Mice, Inbred C57BL , Protein Binding , Sciatic Nerve/injuries , Sciatic Nerve/metabolism
5.
Bioorg Chem ; 88: 102915, 2019 07.
Article in English | MEDLINE | ID: mdl-31005784

ABSTRACT

Drugs targeting different calcium channel subtypes have strong therapeutic potential for future drug development for cardiovascular disorders, neuropsychiatric diseases and cancer. This study aims to design and synthesize a new series of C2 substituted dihydropyrimidines to mimic the structure features of third generation long acting dihydropyridine calcium channel blockers and dihydropyrimidines analogues. The target compounds have been evaluated as blockers for CaV1.2 and CaV3.2 utilizing the whole-cell patch clamp technique. Among the tested compounds, compound 7a showed moderate calcium channel blockade activity against CaV3.2. Moreover, the predicted physicochemical properties and pharmacokinetic profiles of the target compounds recommend that they can be considered as drug-like candidates. The results highlight some significant information for the future design of lead compounds as calcium channel blockers.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Pyrimidines/pharmacology , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacokinetics , Cell Line , Computer Simulation , Drug Design , Electrophysiology/methods , Humans , Patch-Clamp Techniques , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics
6.
Mol Brain ; 12(1): 117, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31888677

ABSTRACT

Clinical and preclinical studies have shown that patients with Diabetic Neuropathy Pain (DNP) present with increased tumor necrosis factor alpha (TNF-α) serum concentration, whereas studies with diabetic animals have shown that TNF-α induces an increase in NaV1.7 sodium channel expression. This is expected to result in sensitization of nociceptor neuron terminals, and therefore the development of DNP. For further study of this mechanism, dissociated dorsal root ganglion (DRG) neurons were exposed to TNF-α for 6 h, at a concentration equivalent to that measured in STZ-induced diabetic rats that developed hyperalgesia. Tetrodotoxin sensitive (TTXs), resistant (TTXr) and total sodium current was studied in these DRG neurons. Total sodium current was also studied in DRG neurons expressing the collapsin response mediator protein 2 (CRMP2) SUMO-incompetent mutant protein (CRMP2-K374A), which causes a significant reduction in NaV1.7 membrane cell expression levels. Our results show that TNF-α exposure increased the density of the total, TTXs and TTXr sodium current in DRG neurons. Furthermore, TNF-α shifted the steady state activation and inactivation curves of the total and TTXs sodium current. DRG neurons expressing the CRMP2-K374A mutant also exhibited total sodium current increases after exposure to TNF-α, indicating that these effects were independent of SUMOylation of CRMP2. In conclusion, TNF-α sensitizes DRG neurons via augmentation of whole cell sodium current. This may underlie the pronociceptive effects of TNF-α and suggests a molecular mechanism responsible for pain hypersensitivity in diabetic neuropathy patients.


Subject(s)
Ganglia, Spinal/cytology , Intercellular Signaling Peptides and Proteins/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Sumoylation , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Animals , Behavior, Animal , Cell Membrane/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Hyperalgesia/blood , Hyperalgesia/complications , Ion Channel Gating , Male , Mutant Proteins/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Tumor Necrosis Factor-alpha/blood
7.
Bioorg Chem ; 83: 354-366, 2019 03.
Article in English | MEDLINE | ID: mdl-30408648

ABSTRACT

New dihydropyrimidines bearing various lipophilic pharmacophores and functionalities at position 3 were designed and synthesized. The basic framework of the new compounds was designed to maintain the main structural requirements for calcium channel blocking activity of the known dihydropyridines and dihydropyrimidines calcium channel blockers. The newly synthesized compounds were evaluated as antagonists for CaV1.2 and CaV3.2 using the whole-cell patch clamp technique. Seven compounds (4b, 4c, 6c, 9, 13c, 13e and 17b) showed promising dual calcium channel blocking activity and three compounds (13b, 14b and 17a) were selective against Cav3.2. Their drug-likeness has been assessed using Molinspiration and Molsoft softwares. Their physicochemical properties and pharmacokinetic profiles recommend that they can be considered as drug-like candidates.


Subject(s)
Calcium Channel Blockers/pharmacology , Pyrimidines/pharmacology , Animals , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacokinetics , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Cell Line , Drug Design , Humans , Molecular Structure , Patch-Clamp Techniques , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Rats , Solubility , Structure-Activity Relationship
8.
Cell Rep ; 24(9): 2356-2369.e5, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30157429

ABSTRACT

The NLRP3 inflammasome senses a range of cellular disturbances, although no consensus exists regarding a common mechanism. Canonical NLRP3 activation is blocked by high extracellular K+, regardless of the activating signal. We report here that canonical NLRP3 activation leads to Ca2+ flux and increased calpain activity. Activated calpain releases a pool of Caspase-1 sequestered by the cytoskeleton to regulate NLRP3 activation. Using electrophysiological recording, we found that resting-state eukaryotic membrane potential (MP) is required for this calpain activity, and depolarization by high extracellular K+ or artificial hyperpolarization results in the inhibition of calpain. Therefore, the MP/Ca2+/calpain/Caspase-1 axis acts as an independent regulatory mechanism for NLRP3 activity. This finding provides mechanistic insight into high K+-mediated inhibition of NLRP3 activation, and it offers an alternative model of NLRP3 inflammasome activation that does not involve K+ efflux.


Subject(s)
Calpain/metabolism , Caspase 1/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Calcium/metabolism , Female , HEK293 Cells , Humans , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Potassium/metabolism , THP-1 Cells
9.
Mol Brain ; 11(1): 24, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720258

ABSTRACT

This study describes the functional interaction between the Cav3.1 and Cav3.2 T-type calcium channels and cytoskeletal spectrin (α/ß) and ankyrin B proteins. The interactions were identified utilizing a proteomic approach to identify proteins that interact with a conserved negatively charged cytosolic region present in the carboxy-terminus of T-type calcium channels. Deletion of this stretch of amino acids decreased binding of Cav3.1 and Cav3.2 calcium channels to spectrin (α/ß) and ankyrin B and notably also reduced T-type whole cell current densities in expression systems. Furthermore, fluorescence recovery after photobleaching analysis of mutant channels lacking the proximal C-terminus region revealed reduced recovery of both Cav3.1 and Cav3.2 mutant channels in hippocampal neurons. Knockdown of spectrin α and ankyrin B decreased the density of endogenous Cav3.2 in hippocampal neurons. These findings reveal spectrin (α/ß) / ankyrin B cytoskeletal and signaling proteins as key regulators of T-type calcium channels expressed in the nervous system.


Subject(s)
Ankyrins/metabolism , Calcium Channels, T-Type/metabolism , Spectrin/metabolism , Amino Acid Sequence , Animals , Calcium Channels, T-Type/chemistry , Caveolin 3/chemistry , Caveolin 3/metabolism , Cytoskeleton/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Humans , Ion Channel Gating , Mice , Mutant Proteins/metabolism , Protein Binding , Protein Domains , Rats
10.
Cell Rep ; 22(8): 1956-1964, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466724

ABSTRACT

Cavα2δ subunits contribute to the cell-surface expression of Cav2 calcium channels. Upregulation of Cavα2δ-1 in dorsal root ganglion neurons occurs after nerve injury and results in an increased synaptic abundance of Cav2.2 channels in the spinal dorsal horn, thus enhancing the transmission of pain signals. Here, we report that large conductance calcium-activated potassium (BK) channels interact with the Cavα2δ subunit. Coexpression of BK channels with the Cav2 calcium channels reduces their cell-surface expression and whole-cell current density by competing the Cavα2δ subunit away from the Cav2 complex. Biochemical analysis reveals that the extracellular N terminus region of the BK channel is the key molecular determinant of this effect. Intrathecally delivered virus constructs encoding a membrane-anchored BK channel N terminus peptide produces long-lasting analgesia in mouse models of inflammatory and neuropathic pain. Collectively, our data reveal an endogenous ligand of the Cavα2δ subunit with analgesic properties.


Subject(s)
Calcium Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Neuralgia/metabolism , Neuralgia/pathology , Protein Subunits/metabolism , Amino Acid Sequence , Analgesia , Animals , Cell Membrane/metabolism , Inflammation/pathology , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Male , Mice, Inbred C57BL , Protein Subunits/chemistry
11.
J Neurosci ; 37(46): 11255-11270, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29038242

ABSTRACT

CaV1 L-type calcium channels are key to regulating neuronal excitability, with the range of functional roles enhanced by interactions with calmodulin, accessory proteins, or CaMKII that modulate channel activity. In hippocampal pyramidal cells, a prominent elevation of CaV1 activity is apparent in late channel openings that can last for seconds following a depolarizing stimulus train. The current study tested the hypothesis that a reported interaction among CaV1.3 channels, the scaffolding protein densin, and CaMKII could generate a facilitation of channel activity that outlasts a depolarizing stimulus. We found that CaV1.3 but not CaV1.2 channels exhibit a long-duration calcium-dependent facilitation (L-CDF) that lasts up to 8 s following a brief 50 Hz stimulus train, but only when coexpressed with densin and CaMKII. To test the physiological role for CaV1.3 L-CDF, we coexpressed the intermediate-conductance KCa3.1 potassium channel, revealing a strong functional coupling to CaV1.3 channel activity that was accentuated by densin and CaMKII. Moreover, the CaV1.3-densin-CaMKII interaction gave rise to an outward tail current of up to 8 s duration following a depolarizing stimulus in both tsA-201 cells and male rat CA1 pyramidal cells. A slow afterhyperpolarization in pyramidal cells was reduced by a selective block of CaV1 channels by isradipine, a CaMKII blocker, and siRNA knockdown of densin, and spike frequency increased upon selective block of CaV1 channel conductance. The results are important in revealing a CaV1.3-densin-CaMKII interaction that extends the contribution of CaV1.3 calcium influx to a time frame well beyond a brief input train.SIGNIFICANCE STATEMENT CaV1 L-type calcium channels play a key role in regulating the output of central neurons by providing calcium influx during repetitive inputs. This study identifies a long-duration calcium-dependent facilitation (L-CDF) of CaV1.3 channels that depends on the scaffolding protein densin and CaMKII and that outlasts a depolarizing stimulus by seconds. We further show a tight functional coupling between CaV1.3 calcium influx and the intermediate-conductance KCa3.1 potassium channel that promotes an outward tail current of up to 8 s following a depolarizing stimulus. Tests in CA1 hippocampal pyramidal cells reveal that a slow AHP is reduced by blocking different components of the CaV1.3-densin-CaMKII interaction, identifying an important role for CaV1.3 L-CDF in regulating neuronal excitability.


Subject(s)
Action Potentials/physiology , Calcium Channels/metabolism , Hippocampus/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Neurons/metabolism , Action Potentials/drug effects , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Hippocampus/drug effects , Male , Neurons/drug effects , Organ Culture Techniques , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/metabolism
12.
Sci Rep ; 7(1): 11513, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912545

ABSTRACT

Low-voltage-activated T-type calcium channels are essential contributors to the functioning of thalamocortical neurons by supporting burst-firing mode of action potentials. Enhanced T-type calcium conductance has been reported in the Genetic Absence Epilepsy Rat from Strasbourg (GAERS) and proposed to be causally related to the overall development of absence seizure activity. Here, we show that calnexin, an endoplasmic reticulum integral membrane protein, interacts with the III-IV linker region of the Cav3.2 channel to modulate the sorting of the channel to the cell surface. We demonstrate that the GAERS missense mutation located in the Cav3.2 III-IV linker alters the Cav3.2/calnexin interaction, resulting in an increased surface expression of the channel and a concomitant elevation in calcium influx. Our study reveals a novel mechanism that controls the expression of T-type channels, and provides a molecular explanation for the enhancement of T-type calcium conductance in GAERS.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Calnexin/metabolism , Epilepsy, Absence/genetics , Mutation, Missense , Animals , Disease Models, Animal , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Transport , Rats
13.
Mol Brain ; 10(1): 37, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28800734

ABSTRACT

Calmodulin (CaM) is an important signaling molecule that regulates a vast array of cellular functions by activating second messengers involved in cell function and plasticity. Low voltage-activated calcium channels of the Cav3 family have the important role of mediating low threshold calcium influx, but were not believed to interact with CaM. We find a constitutive association between CaM and the Cav3.1 channel at rest that is lost through an activity-dependent and Cav3.1 calcium-dependent CaM dissociation. Moreover, Cav3 calcium influx is sufficient to activate αCaMKII in the cytoplasm in a manner that depends on an intact Cav3.1 C-terminus needed to support the CaM interaction. Our findings thus establish that T-type channel calcium influx invokes a novel dynamic interaction between CaM and Cav3.1 channels to trigger a signaling cascade that leads to αCaMKII activation.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Animals , Calcium/metabolism , Enzyme Activation , Fluorescence Resonance Energy Transfer , Humans , Immunoprecipitation , Mice, Inbred C57BL , Neurons/metabolism , Phosphorylation , Protein Aggregates , Rats, Sprague-Dawley
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.
Channels (Austin) ; 11(5): 434-443, 2017 Sep 03.
Article in English | MEDLINE | ID: mdl-28467171

ABSTRACT

Formation of complexes between ion channels is important for signal processing in the brain. Here we investigate the biochemical and biophysical interactions between HCN1 channels and Cav3.2 T-type channels. We found that HCN1 co-immunoprecipitated with Cav3.2 from lysates of either mouse brain or tsA-201 cells, with the HCN1 N-terminus associating with the Cav3.2 N-terminus. Cav3.2 channel activity appeared to be functionally regulated by HCN1. The expression of HCN1 induced a decrease in Cav3.2 Ba2+ influx (IBa2+) along with altered channel kinetics and a depolarizing shift in activation gating. However, a reciprocal regulation of HCN1 by Cav3.2 was not observed. This study highlights a regulatory role of HCN1 on Cav3.2 voltage-dependent properties, which are expected to affect physiologic functions such as synaptic transmission and cellular excitability.


Subject(s)
Calcium Channels, T-Type/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Potassium Channels/metabolism , Animals , Brain/metabolism , Calcium Channels, T-Type/genetics , Cell Line , Down-Regulation , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Mice , Potassium Channels/genetics , Signal Transduction
16.
Eur J Med Chem ; 134: 52-61, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28399450

ABSTRACT

Cardiovascular diseases (CVDs) are the main cause of deaths worldwide. Up-to-date, hypertension is the most significant contributing factor to CVDs. Recent clinical studies recommend calcium channel blockers (CCBs) as effective treatment alone or in combination with other medications. Being the most clinically useful CCBs, 1,4-dihydropyridines (DHPs) attracted great interest in improving potency and selectivity. However, the short plasma half-life which may be attributed to the metabolic oxidation to the pyridine-counterparts is considered as a major limitation for this class. Among the most efficient modifications of the DHP scaffold, is the introduction of biologically active N3-substituted dihydropyrimidine mimics (DHPMs). Again, some potent DHPMs showed only in vitro activity due to first pass effect through hydrolysis and removal of the N3-substitutions. Herein, the synthesis of new N3-substituted DHPMs with various functionalities linked to the DHPM core via two-carbon spacer to guard against possible metabolic inactivation is described. It was designed to keep close structural similarities to clinically efficient DHPs and the reported lead DHPMs analogues, while attempting to improve the pharmacokinetic properties through better metabolic stability. Applying whole batch clamp technique, five compounds showed promising L- and T- type calcium channel blocking activity and were identified as lead compounds. Structure requirements for selectivity against Cav1.2 as well against Cav3.2 are described.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Dihydropyridines/chemistry , Dihydropyridines/pharmacology , Calcium Channel Blockers/chemical synthesis , Crystallography, X-Ray , Dihydropyridines/chemical synthesis , HEK293 Cells , Humans , Hypertension/drug therapy , Models, Molecular
17.
Epilepsia ; 58(4): 617-626, 2017 04.
Article in English | MEDLINE | ID: mdl-28230232

ABSTRACT

OBJECTIVES: Carisbamate (CRS) is a novel monocarbamate compound that possesses antiseizure and neuroprotective properties. However, the mechanisms underlying these actions remain unclear. Here, we tested both direct and indirect effects of CRS on several cellular systems that regulate intracellular calcium concentration [Ca2+ ]i . METHODS: We used a combination of cellular electrophysiologic techniques, as well as cell viability, Store Overload-Induced Calcium Release (SOICR), and mitochondrial functional assays to determine whether CRS might affect [Ca2+ ]i levels through actions on the endoplasmic reticulum (ER), mitochondria, and/or T-type voltage-gated Ca2+ channels. RESULTS: In CA3 pyramidal neurons, kainic acid induced significant elevations in [Ca2+ ]i and long-lasting neuronal hyperexcitability, both of which were reversed in a dose-dependent manner by CRS. Similarly, CRS suppressed spontaneous rhythmic epileptiform activity in hippocampal slices exposed to zero-Mg2+ or 4-aminopyridine. Treatment with CRS also protected murine hippocampal HT-22 cells against excitotoxic injury with glutamate, and this was accompanied by a reduction in [Ca2+ ]i . Neither kainic acid nor CRS alone altered the mitochondrial membrane potential (ΔΨ) in intact, acutely isolated mitochondria. In addition, CRS did not affect mitochondrial respiratory chain activity, Ca2+ -induced mitochondrial permeability transition, and Ca2+ release from the ER. However, CRS significantly decreased Ca2+ flux in human embryonic kidney tsA-201 cells transfected with Cav 3.1 (voltage-dependent T-type Ca2+ ) channels. SIGNIFICANCE: Our data indicate that the neuroprotective and antiseizure activity of CRS likely results in part from decreased [Ca2+ ]i accumulation through blockade of T-type Ca2+ channels.


Subject(s)
Anticonvulsants/pharmacology , Calcium Channels, T-Type/metabolism , Carbamates/pharmacology , Neurons/drug effects , Animals , Calcium/metabolism , Calcium Channels, T-Type/genetics , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Glutamic Acid/pharmacology , HEK293 Cells , Hippocampus/cytology , Humans , In Vitro Techniques , Kainic Acid/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium Channel Blockers/pharmacology , Spectrometry, Fluorescence , Transfection
18.
Bioorg Med Chem ; 25(6): 1926-1938, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28233679

ABSTRACT

Low-voltage-activated calcium channels are important regulators of neurotransmission and membrane ion conductance. A plethora of intracellular events rely on their modulation. Accordingly, they are implicated in many disorders including epilepsy, Parkinson's disease, pain and other neurological diseases. Among different subfamilies, T-type calcium channels, and in particular the CaV3.2 isoform, were shown to be involved in nociceptive neurotransmission. The role of CaV3.2 in pain modulation was supported by demonstrating selective antisense oligonucleotide-mediated CaV3.2 knockdown, in vivo antinociceptive effects of T-type blockers, and pain attenuation in CaV3.2 knockout formalin-induced pain model. These Emerging investigations have provided new insights into targeting T-type calcium channels for pain management. Within this scope, various T-type calcium channel blockers have been developed such as mibefradil and ethosuximide. Although being active, most of these molecules interact with other receptors as well. This addresses the need for T-selectivity. Few selective T-type channel blockers of diverse chemical classes were developed such as ABT-639 and TTA-P2. Interestingly, R(-) efonidipine which is a dihydropyridine (DHP) showed T-channel selectivity. Systematic modification of 1,4-dihydropyridine scaffold introduced novel derivatives with 40-fold T-type selectivity over L-type calcium channels. Along these lines, substitution of the DHP core with various analogues favored T-selectivity and may serve as novel pharmacophores. Several dihydropyrimidine (DHPM) mimics were introduced by Squibb as potential candidates. As a continuation of this approach, the current study describes the synthesis of Novel N3 substituted DHPMs with structure similarities to the active DHPs. Different functional groups were introduced to the N3 position through a spacer to gain more information about activity and selectivity. Furthermore, the spacer aims at improving the metabolic stability of the molecules. Initial screening data by whole patch clamp technique showed a robust inhibition of Cav3.2 T-type channels by eleven compounds. Interestingly, four compounds of these were efficient selective T-type blockers. Based on selectivity and efficiency, two compounds were selected for in vivo evaluation in mouse models of inflammatory pain. Results showed effective attenuation of nociception and mechanical hypersensitivity.


Subject(s)
Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacology , Disease Models, Animal , Inflammation/drug therapy , Pain/drug therapy , Animals , Calcium Channel Blockers/chemistry , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Proton Magnetic Resonance Spectroscopy
19.
Pflugers Arch ; 468(11-12): 1837-1851, 2016 11.
Article in English | MEDLINE | ID: mdl-27659162

ABSTRACT

T-type calcium channels are key contributors to neuronal physiology where they shape electrical activity of nerve cells and contribute to the release of neurotransmitters. Enhanced T-type channel expression has been causally linked to a number of pathological conditions including peripheral painful diabetic neuropathy. Recently, it was demonstrated that asparagine-linked glycosylation not only plays an essential role in regulating cell surface expression of Cav3.2 channels, but may also support glucose-dependent potentiation of T-type currents. However, the underlying mechanisms by which N-glycosylation and glucose levels modulate the expression of T-type channels remain elusive. In the present study, we show that site-specific N-glycosylation of Cav3.2 is essential to stabilize expression of the channel at the plasma membrane. In contrast, elevated external glucose concentration appears to potentiate intracellular forward trafficking of the channel to the cell surface, resulting in an increased steady-state expression of the channel protein at the plasma membrane. Collectively, our study indicates that glucose and N-glycosylation act in concert to control the expression of Cav3.2 channels, and that alteration of these mechanisms may contribute to the altered expression of T-type channels in pathological conditions.


Subject(s)
Calcium Channels, T-Type/metabolism , Glucose/pharmacology , Protein Processing, Post-Translational , Asparagine/metabolism , Calcium Channels, T-Type/genetics , Cell Membrane/metabolism , Glycosylation , HEK293 Cells , Humans , Protein Transport/drug effects
20.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27053601

ABSTRACT

BACKGROUND: T-type calcium channels are important contributors to signaling in the primary afferent pain pathway and are thus important targets for the development of analgesics. It has been previously reported that certain piperazine-based compounds such as flunarizine are able to inhibit T-type calcium channels. Thus, we hypothesized that novel piperazine compounds could potentially act as analgesics. RESULTS: Here, we have created a series of 14 compound derivatives around a diphenyl methyl-piperazine core pharmacophore. Testing their effects on transiently expressed Cav3.2 calcium channels revealed one derivative (3-((4-(bis(4-fluorophenyl)methyl)piperazin-1-yl)methyl)-4-(2-methoxyphenyl)-1,2,5-oxadiazole 2-oxide, compound 10e) as a potent blocker. 10e mediate tonic block of these channels with an IC50 of around 4 micromolar. 10e also blocked Cav3.1 and Cav3.3 channels, but only weakly affected high-voltage-activated Cav1.2 and Cav2.2 channels. Intrathecal delivery of 10e mediated relief from formalin and complete Freund's adjuvant induced inflammatory pain that was ablated by genetic knockout of Cav3.2 channels. CONCLUSIONS: Altogether, our data identify a novel T-type calcium channel blocker with tight structure activity relationship (SAR) and relevant in vivo efficacy in inflammatory pain conditions.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Piperazines/chemical synthesis , Piperazines/pharmacology , Acute Pain/drug therapy , Acute Pain/physiopathology , Analgesics/therapeutic use , Animals , Calcium Channel Blockers/therapeutic use , Electrophysiological Phenomena/drug effects , HEK293 Cells , Humans , Injections, Spinal , Male , Mice, Inbred C57BL , Piperazines/therapeutic use , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...