Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
iScience ; 27(6): 109973, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38827405

RESUMO

N- and P/Q-type voltage-gated Ca2+ channels are critical for synaptic transmission. While their expression is increased in the dorsal root ganglion (DRG) neuron cell bodies during neuropathic pain conditions, less is known about their synaptic remodeling. Here, we combined genetic tools with 2-photon Ca2+ imaging to explore the functional remodeling that occurs in central presynaptic terminals of DRG neurons during neuropathic pain. We imaged GCaMP6s fluorescence responses in an ex vivo spinal cord preparation from mice expressing GCaMP6s in Trpv1-Cre lineage nociceptors. We show that Ca2+ transient amplitude is increased in central terminals of these neurons after spared nerve injury, and that this increase is mediated by both N- and P/Q-type channels. We found that GABA-B receptor-dependent inhibition of Ca2+ transients was potentiated in the superficial layer of the dorsal horn. Our results provide direct evidence toward nerve injury-induced functional remodeling of presynaptic Ca2+ channels in Trpv1-lineage nociceptor terminals.

2.
Biomed Pharmacother ; 174: 116472, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38531121

RESUMO

The Voltage-Gated Calcium Channel (VGCC) auxiliary subunit Cavα2δ-1 (CACNA2D1) is the target/receptor of gabapentinoids which are known therapeutics in epilepsy and neuropathic pain. Following damage to the peripheral sensory nervous system, Cavα2δ-1 is upregulated in dorsal root ganglion (DRG) neurons in several animal models of chronic neuropathic pain. Gabapentinoids, such as gabapentin and pregabalin, engage with Cavα2δ-1 via binding an arginine residue (R241) within an RRR motif located at the N-terminus of human Cavα2δ-1. A novel, next generation gabapentinoid, engineered not to penetrate the brain, was able to generate a strong analgesic response in Chronic Constriction Injury animal model of chronic neuropathic pain and showed binding specificity for Cavα2δ-1 versus the Cavα2δ-2 subunit. This novel non-brain penetrant gabapentinoid, binds to R241 and a novel binding site on Cavα2δ-1, which is located within the VGCC_α2 domain, identified as a lysine residue within an IKAK amino acid motif (K634). The overall whole cell current amplitudes were diminished by the compound, with these inhibitory effects being diminished in R241A mutant Cavα2δ-1 subunits. The functional effects occurred at lower concentrations than those needed for inhibition by gabapentin or pregabalin, which apparently bound the Cavα2δ-1 subunit only on the R241 and not on the K634 residue. Our work sets the stage for the identification and characterisation of novel compounds with therapeutic properties in neuropathic pain and possibly in other disorders and conditions which require engagement of the Cavα2δ-1 target.


Assuntos
Canais de Cálcio Tipo L , Neuralgia , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Animais , Ligantes , Humanos , Masculino , Canais de Cálcio/metabolismo , Canais de Cálcio/genética , Gabapentina/farmacologia , Ratos Sprague-Dawley , Gânglios Espinais/metabolismo , Gânglios Espinais/efeitos dos fármacos , Ratos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio Tipo N/genética , Analgésicos/farmacologia , Modelos Animais de Doenças , Pregabalina/farmacologia
3.
Cell Rep Med ; 5(2): 101425, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38382469

RESUMO

Progressive myoclonus epilepsy type 7, a debilitating neurological disorder, is caused by a loss-of-function mutation in the KV3.1 channel. Exciting work by Feng et al.1 utilizes a new knockin mouse model to identify a potential therapeutic intervention.


Assuntos
Epilepsias Mioclônicas Progressivas , Animais , Camundongos , Epilepsias Mioclônicas Progressivas/genética , Mutação
5.
Br J Pharmacol ; 180(12): 1616-1633, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36647671

RESUMO

BACKGROUND AND PURPOSE: Cannabinoids are a promising therapeutic avenue for chronic pain. However, clinical trials often fail to report analgesic efficacy of cannabinoids. Inhibition of voltage gate calcium (Cav ) channels is one mechanism through which cannabinoids may produce analgesia. We hypothesized that cannabinoids and cannabinoid receptor agonists target different types of Cav channels through distinct mechanisms. EXPERIMENTAL APPROACH: Electrophysiological recordings from tsA-201 cells expressing either Cav 3.2 or Cav 2.2 were used to assess inhibition by HU-210 or cannabidiol (CBD) in the absence and presence of the CB1 receptor. Homology modelling assessed potential interaction sites for CBD in both Cav 2.2 and Cav 3.2. Analgesic effects of CBD were assessed in mouse models of inflammatory and neuropathic pain. KEY RESULTS: HU-210 (1 µM) inhibited Cav 2.2 function in the presence of CB1 receptor but had no effect on Cav 3.2 regardless of co-expression of CB1 receptor. By contrast, CBD (3 µM) produced no inhibition of Cav 2.2 and instead inhibited Cav 3.2 independently of CB1 receptors. Homology modelling supported these findings, indicating that CBD binds to and occludes the pore of Cav 3.2, but not Cav 2.2. Intrathecal CBD alleviated thermal and mechanical hypersensitivity in both male and female mice, and this effect was absent in Cav 3.2 null mice. CONCLUSION AND IMPLICATIONS: Our findings reveal differential modulation of Cav 2.2 and Cav 3.2 channels by CB1 receptors and CBD. This advances our understanding of how different cannabinoids produce analgesia through action at different voltage-gated calcium channels and could influence the development of novel cannabinoid-based therapeutics for treatment of chronic pain.


Assuntos
Canabidiol , Canabinoides , Dor Crônica , Masculino , Feminino , Camundongos , Animais , Canabidiol/farmacologia , Canais de Cálcio , Dor Crônica/tratamento farmacológico , Analgésicos/farmacologia , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo
6.
Pain ; 163(12): 2315-2325, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-35467587

RESUMO

ABSTRACT: Trigeminal neuralgia (TN) is a rare but debilitating disorder characterized by excruciating facial pain, with a higher incidence in women. Recent studies demonstrated that TN patients present mutations in the gene encoding the Ca V 3.2 T-type calcium channel, an important player in peripheral pain pathways. We characterize the role of Ca V 3.2 channels in TN at 2 levels. First, we examined the biophysical properties of CACNA1H variants found in TN patients. Second, we investigated the role of Ca V 3.2 in an animal model of trigeminal neuropathic pain. Whole-cell patch-clamp recordings from 4 different mutants expressed in tsA-201 cells (E286K in the pore loop of domain I, H526Y, G563R, and P566T in the domain I-II linker) identified a loss of function in activation in the E286K mutation and gain of function in the G563R and P566T mutations. Moreover, a loss of function in inactivation was observed with the E286K and H526Y mutations. Cell surface biotinylation revealed no difference in channel trafficking among the variants. The G563R mutant also caused a gain of function in the firing properties of transfected trigeminal ganglion neurons. In female and male mice, constriction of the infraorbital nerve induced facial thermal heat hyperalgesia. Block of T-type channels with Z944 resulted in antihyperalgesia. The effect of Z944 was absent in Ca V 3.2 -/- mice, indicating that Ca V 3.2 is the molecular target of the antihyperalgesic Z944 effect. Finally, enzyme-linked immunosorbent assay analysis revealed increased Ca V 3.2 channel expression in the spinal trigeminal subnucleus caudalis. Altogether, the present study demonstrates an important role of Ca V 3.2 channels in trigeminal pain.


Assuntos
Canais de Cálcio Tipo T , Neuralgia do Trigêmeo , Animais , Feminino , Masculino , Camundongos , Dor Facial , Hiperalgesia , Gânglio Trigeminal/metabolismo , Neuralgia do Trigêmeo/genética , Canais de Cálcio Tipo T/metabolismo
7.
FEBS J ; 289(3): 614-633, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33576127

RESUMO

Voltage-gated calcium (CaV ) channels and their regulation by proteins at the synaptic cleft play a critical role in neurotransmission. These interactions fine-tune the synaptic response through the regulation of Ca2+ entry into the presynaptic terminal and trigger the fusion of vesicles filled with neurotransmitters and peptides. Regulation of CaV channel intrinsic properties and their numbers at the active zones shape the timing and strength of synaptic function. Here, we provide an overview of a number of proteins reported to be part of CaV channel nanodomains at the synaptic cleft and the repercussions of these interactions for CaV channel trafficking, tethering at the active zone, and regulation of their biophysical properties. We summarize the current state of knowledge by which CaV channels are regulated at presynaptic sites.


Assuntos
Canais de Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Sinapses/genética , Transmissão Sináptica/genética , Cálcio/metabolismo , Sinalização do Cálcio/genética , Humanos , Neurônios/metabolismo , Terminações Pré-Sinápticas/metabolismo
8.
Pflugers Arch ; 474(4): 447-454, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34623515

RESUMO

This study describes the interaction between CaV3.2 calcium channels and the receptor for activated C kinase 1 (Rack-1), a scaffold protein which has recently been implicated in neuropathic pain. The coexpression of CaV3.2 and Rack-1 in tsA-201 cells led to a reduction in the magnitude of whole-cell CaV3.2 currents and CaV3.2 channel expression at the plasma membrane. Co-immunoprecipitations from transfected cells show the formation of a molecular protein complex between Cav3.2 channels and Rack-1. We determined that the interaction of Rack-1 occurs at the intracellular II-III loop and the C-terminus of the channel. Finally, the coexpression of PKCßII abolished the effect of Rack-1 on current densities. Altogether, our findings show that Rack-1 regulates CaV3.2-mediated calcium entry in a PKC-dependent manner.


Assuntos
Canais de Cálcio Tipo T , Neuralgia , Canais de Cálcio Tipo T/metabolismo , Membrana Celular/metabolismo , Humanos , Receptores de Quinase C Ativada/metabolismo
10.
Mol Brain ; 14(1): 145, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34544471

RESUMO

The CACNA1H gene encodes the α1 subunit of the low voltage-activated Cav3.2 T-type calcium channel, an important regulator of neuronal excitability. Alternative mRNA splicing can generate multiple channel variants with distinct biophysical properties and expression patterns. Two major splice variants, containing or lacking exon 26 (± 26) have been found in different human tissues. In this study, we report splice variant specific effects of a Cav3.2 mutation found in patients with autosomal dominant writer's cramp, a specific type of focal dystonia. We had previously reported that the R481C missense mutation caused a gain of function effect when expressed in Cav3.2 (+ 26) by accelerating its recovery from inactivation. Here, we show that when the mutation is expressed in the short variant of the channel (- 26), we observe a significant increase in current density when compared to wild-type Cav3.2 (- 26) but the effect on the recovery from inactivation is lost. Our data add to growing evidence that the functional expression of calcium channel mutations depends on which splice variant is being examined.


Assuntos
Canais de Cálcio Tipo T/genética , Distúrbios Distônicos/genética , Potenciais de Ação , Processamento Alternativo , Bário/metabolismo , Canais de Cálcio Tipo T/fisiologia , Distúrbios Distônicos/fisiopatologia , Éxons/genética , Mutação com Ganho de Função , Humanos , Transporte de Íons , Modelos Moleculares , Mutação de Sentido Incorreto , Mutação Puntual , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia
11.
Mol Brain ; 14(1): 27, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33557884

RESUMO

CACNA1A pathogenic variants have been linked to several neurological disorders including familial hemiplegic migraine and cerebellar conditions. More recently, de novo variants have been associated with severe early onset developmental encephalopathies. CACNA1A is highly expressed in the central nervous system and encodes the pore-forming CaVα1 subunit of P/Q-type (Cav2.1) calcium channels. We have previously identified a patient with a de novo missense mutation in CACNA1A (p.Y1384C), characterized by hemiplegic migraine, cerebellar atrophy and developmental delay. The mutation is located at the transmembrane S5 segment of the third domain. Functional analysis in two predominant splice variants of the neuronal Cav2.1 channel showed a significant loss of function in current density and changes in gating properties. Moreover, Y1384 variants exhibit differential splice variant-specific effects on recovery from inactivation. Finally, structural analysis revealed structural damage caused by the tyrosine substitution and changes in electrostatic potentials.


Assuntos
Canais de Cálcio Tipo N/genética , Cerebelo/patologia , Deficiências do Desenvolvimento/genética , Predisposição Genética para Doença , Enxaqueca com Aura/genética , Mutação/genética , Adolescente , Adulto , Processamento Alternativo/genética , Atrofia , Fenômenos Biofísicos , Canais de Cálcio Tipo N/química , Canais de Cálcio Tipo N/metabolismo , Linhagem Celular , Pré-Escolar , Deficiências do Desenvolvimento/complicações , Feminino , Humanos , Recém-Nascido , Ativação do Canal Iônico , Masculino , Enxaqueca com Aura/complicações , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Isoformas de Proteínas/genética , Estrutura Secundária de Proteína , Homologia Estrutural de Proteína
12.
Mol Brain ; 14(1): 18, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33478561

RESUMO

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.


Assuntos
Canais de Cálcio Tipo T/genética , Distúrbios Distônicos/genética , Predisposição Genética para Doença , Mutação de Sentido Incorreto/genética , Segregação de Cromossomos , Feminino , Humanos , Masculino , Linhagem , Fenótipo
13.
Mol Brain ; 14(1): 4, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33413531

RESUMO

A novel missense mutation in the CACNA1A gene that encodes the pore forming α1 subunit of the CaV2.1 voltage-gated calcium channel was identified in a patient with trigeminal neuralgia. This mutation leads to a substitution of proline 2455 by histidine (P2455H) in the distal C-terminus region of the channel. Due to the well characterized role of this channel in neurotransmitter release, our aim was to characterize the biophysical properties of the P2455H variant in heterologously expressed CaV2.1 channels. Whole-cell patch clamp recordings of wild type and mutant CaV2.1 channels expressed in tsA-201 cells reveal that the mutation mediates a depolarizing shift in the voltage-dependence of activation and inactivation. Moreover, the P2455H mutant strongly reduced calcium-dependent inactivation of the channel that is consistent with an overall gain of function. Hence, the P2455H CaV2.1 missense mutation alters the gating properties of the channel, suggesting that associated changes in CaV2.1-dependent synaptic communication in the trigeminal system may contribute to the development of trigeminal neuralgia.


Assuntos
Canais de Cálcio/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Variação Genética , Ativação do Canal Iônico/genética , Neuralgia do Trigêmeo/genética , Linhagem Celular , Humanos , Mutação/genética
14.
Mol Brain ; 13(1): 149, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33176830

RESUMO

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.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Motivos de Aminoácidos , Asparagina/metabolismo , Canais de Cálcio Tipo T/química , Glicosilação , Humanos , Relação Estrutura-Atividade
15.
Cell Rep ; 33(1): 108233, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33027646

RESUMO

The innate immune system responds to infections that give rise to pain. How the innate immune system interacts with the sensory nervous system and contributes to pain is poorly understood. Here we report that hyperactivity of innate immunity primes and initiates pain states via the TLR2-interleukin-33 (IL-33) axis. Toll-like receptors (TLRs) are upregulated in the complete Freund's adjuvant (CFA) pain model, and knockout of TLR2 abolishes CFA-induced pain. Selective activation of TLR2/6 triggers acute pain via upregulation of IL-33 in the hindpaw, dorsal root ganglia (DRG), and spinal cord in an NLRP3-dependent manner. The IL-33 increase further initiates priming of nociceptive neurons and pain states. Finally, blocking IL-33 receptors at the spinal level mediates analgesia during acute and chronic inflammatory pain, underscoring an important function of IL-33 in pain signaling. Collectively, our data reveal a critical role of the TLR2-IL-33 axis in innate immune activation for pain initiation and maintenance.


Assuntos
Imunidade Inata/genética , Interleucina-33/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Humanos , Camundongos
16.
Bioorg Med Chem ; 28(11): 115491, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32327350

RESUMO

In our previous work, a series of 2-amino-3,4-dihydroquinazoline derivativesusing an electron acceptor group was reported to be potent T-type calcium channel blockers and exhibit strong cytotoxic effects against various cancerous cell lines. To investigate the role of the guanidine moiety in the 2-amino-3,4-dihydroquinazoline scaffold as a pharmacophore for dual biological activity, a new series of 2-thio-3,4-dihydroquniazoline derivatives using an electron donor group at the C2-position was synthesized and evaluated for T-type calcium channel blocking activity and cytotoxic effects against two human cancerous cell lines (lung cancer A549 and colon cancer HCT-116). Among them, compound 6g showed potent inhibition of Cav3.2 currents (83% inhibition) at 10 µM concentrations. The compound also exhibited IC50 values of 5.0 and 6.4 µM against A549 and HCT-116 cell lines, respectively, which are comparable to the parental lead compound KYS05090. These results indicate that the isothiourea moiety similar to the guanidine moiety of 2-amino-3,4-dihydroquinazoline derivatives may be an essential pharmacophore for the desired biological activities. Therefore, our preliminary work can provide the opportunity to expand a chemical repertoire to improve affinity and selectivity for T-type calcium channels.


Assuntos
Antineoplásicos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/metabolismo , Quinazolinas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Bloqueadores dos Canais de Cálcio/síntese química , Bloqueadores dos Canais de Cálcio/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Quinazolinas/síntese química , Quinazolinas/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
17.
Mol Brain ; 12(1): 98, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31775826

RESUMO

We have examined the regulation of mutually exclusive Cav2.2 exon 37a and b variants by the mouse µ-opioid receptor (mMOR) C-terminal splice variants 1, 1C and 1O in tsA-201 cells. Electrophysiological analyses revealed that both channel isoforms exhibit DAMGO-induced voltage-dependent (Gßγ-mediated) inhibition and its recovery by voltage pre-pulses, as well as a voltage-independent component. However, the two channel isoforms differ in their relative extent of voltage-dependent and independent inhibition, with Cav2.2-37b showing significantly more voltage-dependent inhibition upon activation of the three mMOR receptors studied. In addition, coexpression of either mMOR1 or mMOR1C results in an agonist-independent reduction in the peak current density of Cav2.2-37a channels, whereas the peak current density of Cav2.2-37b does not appear to be affected. Interestingly, this decrease is not due to an effect on channel expression at the plasma membrane, as demonstrated by biotinylation experiments. We further examined the mechanism underlying the agonist-independent modulation of Cav2.2-37a by mMOR1C. Incubation of cells with pertussis toxin did not affect the mMOR1C mediated inhibition of Cav2.2-37a currents, indicating a lack of involvement of Gi/o signaling. However, when a Src tyrosine kinase inhibitor was applied, the effect of mMOR1C was lost. Moreover, when we recorded currents using a Cav2.2-37a mutant in which tyrosine 1747 was replaced with phenylalanine (Y1747F), the agonist independent effects of mMOR1C were abolished. Altogether our findings show that Cav2.2-37a and Cav2.2-37b isoforms are subject to differential regulation by C-terminal splice variants of mMORs, and that constitutive mMOR1C activity and downstream tyrosine kinase activity exert a selective inhibition of the Cav2.2-37a splice variant, an N-type channel isoform that is highly enriched in nociceptors. Our study provides new insights into the roles of the MOR full-length C-terminal variants in modulating Cav2.2 channel isoform activities.


Assuntos
Processamento Alternativo/genética , Canais de Cálcio Tipo N/genética , Éxons/genética , Receptores Opioides mu/genética , Animais , Canais de Cálcio Tipo N/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Ativação do Canal Iônico/genética , Camundongos , Proteínas Tirosina Quinases/metabolismo , Ratos , Receptores Opioides mu/metabolismo
18.
Mol Brain ; 12(1): 86, 2019 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-31651342

RESUMO

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.


Assuntos
Canais de Cálcio Tipo T/genética , Epilepsia Generalizada/genética , Mutação/genética , Fenômenos Biofísicos , Criança , Feminino , Humanos , Lactente , Recém-Nascido
19.
Bioorg Chem ; 91: 103187, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31419643

RESUMO

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.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo T/química , Di-Hidropiridinas/farmacologia , Descoberta de Drogas , Glutationa/metabolismo , Sítios de Ligação , Cálcio/metabolismo , Cisteína/química , Cisteína/metabolismo , Humanos , Modelos Moleculares , Conformação Proteica
20.
Mol Brain ; 12(1): 62, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31253182

RESUMO

This study describes the interaction between Cav2 calcium channels and Rabconnectin-3, a di-subunit protein that is associated with synaptic vesicles. Immunostaining reveals that both Rabconnectin-3α (RB-3α) and Rabconnectin-3ß (RB-3ß) are colocalized in mouse hippocampal neurons. Co-immunoprecipitations from brain tissue is consistent with the formation of a protein complex between RB-3α and RB-3ß and both Cav2.2 and the related Cav2.1 calcium channel. The coexpression of either RB-3α or RB-3ß with Cav2.2 calcium channels in tsA-201 cells led to a reduction in Cav2.2 current density without any effects on the voltage-dependence of activation or inactivation. Coexpression of both Rabconnectin-3 subunits did not cause an additive effect on current densities. Finally, the presence of Rabconnectin-3 did not interfere with µ-opioid receptor mediated Gßγ modulation of Cav2.2 channels. Altogether, our findings show that Rabconnectin-3 has the propensity to regulate calcium entry mediated by Cav2.2 channels.


Assuntos
Caveolina 2/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Hipocampo/citologia , Humanos , Camundongos , Neurônios/metabolismo , Ligação Proteica , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA