Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Am J Hum Genet ; 103(5): 666-678, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343943

RESUMO

Developmental and epileptic encephalopathies (DEEs) are severe neurodevelopmental disorders often beginning in infancy or early childhood that are characterized by intractable seizures, abundant epileptiform activity on EEG, and developmental impairment or regression. CACNA1E is highly expressed in the central nervous system and encodes the α1-subunit of the voltage-gated CaV2.3 channel, which conducts high voltage-activated R-type calcium currents that initiate synaptic transmission. Using next-generation sequencing techniques, we identified de novo CACNA1E variants in 30 individuals with DEE, characterized by refractory infantile-onset seizures, severe hypotonia, and profound developmental impairment, often with congenital contractures, macrocephaly, hyperkinetic movement disorders, and early death. Most of the 14, partially recurring, variants cluster within the cytoplasmic ends of all four S6 segments, which form the presumed CaV2.3 channel activation gate. Functional analysis of several S6 variants revealed consistent gain-of-function effects comprising facilitated voltage-dependent activation and slowed inactivation. Another variant located in the domain II S4-S5 linker results in facilitated activation and increased current density. Five participants achieved seizure freedom on the anti-epileptic drug topiramate, which blocks R-type calcium channels. We establish pathogenic variants in CACNA1E as a cause of DEEs and suggest facilitated R-type calcium currents as a disease mechanism for human epilepsy and developmental disorders.


Assuntos
Canais de Cálcio Tipo R/genética , Proteínas de Transporte de Cátions/genética , Contratura/genética , Discinesias/genética , Epilepsia/genética , Variação Genética/genética , Megalencefalia/genética , Espasmos Infantis/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Transtornos do Neurodesenvolvimento/genética
4.
Mol Pain ; 13: 1744806917724698, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28741432

RESUMO

Abstract: We recently reported that nerve injury or peripheral inflammation triggers an upregulation of the deubiquitinase, USP5 in mouse dorsal root ganglion and spinal dorsal horn. This leads to dysregulated ubiquitination of Cav3.2 T-type calcium channels, thus increasing Cav3.2 channel plasma membrane expression and nociceptive signaling in the primary afferent pain pathway. This phenomenon could be recapitulated by noninvasive, optogenetic activation of transient receptor potential vanilloid-1­expressing nociceptors, indicating that neuronal activity is a key player in this process. Given the relevance of the pro-inflammatory cytokine interleukin-1 beta in many forms of pathological pain, we hypothesized that interleukin-1 beta may be a critical cofactor required to drive upregulation of interactions between USP5 and Cav3.2 channels. Here, we report that gene expression, as well as protein levels for interleukin-1 beta and the endogenous interleukin-1 receptor-I antagonist, IL-1Ra are unaltered following conditioning stimulation of optogenetically targeted cutaneous nociceptors, indicating that neuronal activity is not a driver of interleukin-1 beta signaling. In contrast, co-immunoprecipitation experiments revealed that intrathecal administration of interleukin-1 beta in wild-type mice led to an increase in the interaction between USP5 and Cav3.2 in the spinal dorsal horn. Moreover, disruption of the interaction between USP5 and Cav3.2 with TAT peptides suppressed acute nocifensive responses produced by interleukin-1 beta, which was similar to that achieved by elimination of T-type channel activity with the channel blockers, mibefradil, or TTA-A2. Finally, this upregulation could be maintained in dorsal root ganglion neuron cultures exposed overnight to interleukin-1 beta, while the copresence of interleukin-1 receptor antagonist or the dampening of neuronal cell activity with tetrodotoxin attenuated this response. Altogether, our findings identify interleukin-1 beta as an upstream trigger for the upregulation of interactions between USP5 and Cav3.2 channels in the pain pathway, presumably by triggering increased firing activity in afferent fibers.


Assuntos
Canais de Cálcio Tipo T/genética , Interleucina-1beta/metabolismo , Dor/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Animais , Canais de Cálcio Tipo T/metabolismo , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuralgia/metabolismo , Neurônios/metabolismo , Nociceptores/metabolismo , Regulação para Cima
6.
Pflugers Arch ; 468(4): 635-42, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26706850

RESUMO

We report expression system-dependent effects of heterozygous mutations (P769L and A1059S) in the Cav3.2 CACNA1H gene identified in a pediatric patient with chronic pain and absence seizures. The mutations were introduced individually into recombinant channels and then analyzed by means of electrophysiology. When both mutants were co-expressed in tsA-201 cells, we observed a loss of channel function, with significantly smaller current densities across a wide range of voltages (-40 to +20 mV). In addition, when both mutant channels were co-expressed, the channels opened at a more depolarizing potential with a ~5-mV right shift in the half-activation potential, with no changes in half-inactivation potential and the rate of recovery from inactivation. Interestingly, when both mutants were co-expressed in the neuronal-derived CAD cells in a different extracellular milieu, the effect was remarkably different. Although not statistically significant (p < 0.07), current densities appeared augmented compared to wild-type channels and the difference in the half-activation potential was lost. This could be attributed to the replacement of extracellular sodium and potassium with tetraethylammonium chloride. Our results show that experimental conditions can be a confounding factor in the biophysical effects of T-type calcium channel mutations found in certain neurological disorders.


Assuntos
Canais de Cálcio Tipo T/genética , Dor Crônica/genética , Epilepsia Tipo Ausência/genética , Ativação do Canal Iônico , Mutação de Sentido Incorreto , Potenciais de Ação , Adolescente , Canais de Cálcio Tipo T/metabolismo , Dor Crônica/complicações , Dor Crônica/metabolismo , Epilepsia Tipo Ausência/complicações , Epilepsia Tipo Ausência/metabolismo , Feminino , Células HEK293 , Humanos
7.
Pflugers Arch ; 467(4): 677-86, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24862738

RESUMO

The L-type voltage-gated calcium channel Cav1.2 and the calcium-activated CaM kinase cascade both regulate excitation transcription coupling in the brain. CaM kinase is known to associate with the C terminus of Cav1.2 in a region called the PreIQ-IQ domain, which also binds multiple calmodulin molecules. Here we identify and characterize a second CaMKII binding site in the N terminus of Cav1.2 that is formed by a stretch of four amino residues (cysteine-isoleucine-serine-isoleucine) and which regulates channel expression and function. By using live cell imaging of tsA-201 cells we show that GFP fusion constructs of the CaMKII binding region, termed N2B-II co-localize with mCherry-CaMKII. Mutating CISI to AAAA ablates binding to and colocalization with CaMKII. Cav1.2-AAAA channels show reduced cell surface expression in tsA-201 cells, but interestingly, display an increase in channel function that offsets the trafficking deficit. Altogether our data reveal that the proximal N terminus of Cav1.2 contains a CaMKII binding region which contributes to channel surface expression and function.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Potenciais de Ação , Sequência de Aminoácidos , Animais , Sítios de Ligação , Canais de Cálcio Tipo L/química , Membrana Celular/metabolismo , Células HEK293 , Humanos , Dados de Sequência Molecular , Ligação Proteica , Transporte Proteico , Ratos
8.
PLoS Biol ; 9(11): e1001194, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22087075

RESUMO

Cholinergic neurons in the striatum are thought to play major regulatory functions in motor behaviour and reward. These neurons express two vesicular transporters that can load either acetylcholine or glutamate into synaptic vesicles. Consequently cholinergic neurons can release both neurotransmitters, making it difficult to discern their individual contributions for the regulation of striatal functions. Here we have dissected the specific roles of acetylcholine release for striatal-dependent behaviour in mice by selective elimination of the vesicular acetylcholine transporter (VAChT) from striatal cholinergic neurons. Analysis of several behavioural parameters indicates that elimination of VAChT had only marginal consequences in striatum-related tasks and did not affect spontaneous locomotion, cocaine-induced hyperactivity, or its reward properties. However, dopaminergic sensitivity of medium spiny neurons (MSN) and the behavioural outputs in response to direct dopaminergic agonists were enhanced, likely due to increased expression/function of dopamine receptors in the striatum. These observations indicate that previous functions attributed to striatal cholinergic neurons in spontaneous locomotor activity and in the rewarding responses to cocaine are mediated by glutamate and not by acetylcholine release. Our experiments demonstrate how one population of neurons can use two distinct neurotransmitters to differentially regulate a given circuitry. The data also raise the possibility of using VAChT as a target to boost dopaminergic function and decrease high striatal cholinergic activity, common neurochemical alterations in individuals affected with Parkinson's disease.


Assuntos
Acetilcolina/metabolismo , Ácido Glutâmico/metabolismo , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo , Animais , Western Blotting , Encéfalo , Catecolaminas/metabolismo , Imunofluorescência , Técnicas In Vitro , Imageamento por Ressonância Magnética , Masculino , Camundongos , Atividade Motora/genética , Atividade Motora/fisiologia , Reação em Cadeia da Polimerase , Sinaptossomos/metabolismo , Proteínas Vesiculares de Transporte de Acetilcolina/genética
9.
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
10.
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
11.
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
12.
Mol Brain ; 15(1): 91, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36397158

RESUMO

Trigeminal neuralgia (TN) is a rare form of chronic neuropathic pain characterized by spontaneous or elicited paroxysms of electric shock-like or stabbing pain in a region of the face. While most cases occur in a sporadic manner and are accompanied by intracranial vascular compression of the trigeminal nerve root, alteration of ion channels has emerged as a potential exacerbating factor. Recently, whole exome sequencing analysis of familial TN patients identified 19 rare variants in the gene CACNA1H encoding for Cav3.2T-type calcium channels. An initial analysis of 4 of these variants pointed to a pathogenic role. In this study, we assessed the electrophysiological properties of 13 additional TN-associated Cav3.2 variants expressed in tsA-201 cells. Our data indicate that 6 out of the 13 variants analyzed display alteration of their gating properties as evidenced by a hyperpolarizing shift of their voltage dependence of activation and/or inactivation resulting in an enhanced window current supported by Cav3.2 channels. An additional variant enhanced the recovery from inactivation. Simulation of neuronal electrical membrane potential using a computational model of reticular thalamic neuron suggests that TN-associated Cav3.2 variants could enhance neuronal excitability. Altogether, the present study adds to the notion that ion channel polymorphisms could contribute to the etiology of some cases of TN and further support a role for Cav3.2 channels.


Assuntos
Neuralgia do Trigêmeo , Humanos , Canais de Cálcio , Potenciais da Membrana , Neurônios , Neuralgia do Trigêmeo/genética , Fenômenos Eletrofisiológicos
13.
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
14.
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
15.
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
16.
Mol Brain ; 14(1): 126, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34399820

RESUMO

Developmental and epileptic encephalopathies (DEEs) are a group of severe epilepsies that are characterized by seizures and developmental delay. DEEs are primarily attributed to genetic causes and an increasing number of cases have been correlated with variants in ion channel genes. In this study, we report a child with an early severe DEE. Whole exome sequencing showed a de novo heterozygous variant (c.4873-4881 duplication) in the SCN8A gene and an inherited heterozygous variant (c.952G > A) in the CACNA1H gene encoding for Nav1.6 voltage-gated sodium and Cav3.2 voltage-gated calcium channels, respectively. In vitro functional analysis of human Nav1.6 and Cav3.2 channel variants revealed mild but significant alterations of their gating properties that were in general consistent with a gain- and loss-of-channel function, respectively. Although additional studies will be required to confirm the actual pathogenic involvement of SCN8A and CACNA1H, these findings add to the notion that rare ion channel variants may contribute to the etiology of DEEs.


Assuntos
Deficiências do Desenvolvimento/genética , Epilepsia Resistente a Medicamentos/genética , Epilepsia Tônico-Clônica/genética , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Anormalidades Múltiplas/genética , Canais de Cálcio Tipo T/genética , Canais de Cálcio Tipo T/fisiologia , Feminino , Mutação com Ganho de Função , Duplicação Gênica , Predisposição Genética para Doença , Humanos , Recém-Nascido , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Linhagem , Mutação Puntual , Escoliose/genética
17.
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
18.
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
19.
Mol Brain ; 13(1): 33, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32143681

RESUMO

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


Assuntos
Esclerose Lateral Amiotrófica/genética , Canais de Cálcio Tipo T/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Mutação/genética , Sequência de Aminoácidos , Animais , Canais de Cálcio Tipo T/química , Genes Dominantes , Heterozigoto , Masculino , Ratos , Homologia Estrutural de Proteína , Sequenciamento Completo do Genoma
20.
Mol Brain ; 12(1): 75, 2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31477143

RESUMO

Ankyrin B (AnkB) is an adaptor and scaffold for motor proteins and various ion channels that is ubiquitously expressed, including in the brain. AnkB has been associated with neurological disorders such as epilepsy and autism spectrum disorder, but understanding of the underlying mechanisms is limited. Cav2.1, the pore-forming subunit of P/Q type voltage gated calcium channels, is a known interactor of AnkB and plays a crucial role in neuronal function. Here we report that wildtype AnkB increased overall Cav2.1 levels without impacting surface Cav2.1 levels in HEK293T cells. An AnkB variant, p.S646F, which we recently discovered to be associated with seizures, further increased overall Cav2.1 levels, again with no impact on surface Cav2.1 levels. AnkB p.Q879R, on the other hand, increased surface Cav2.1 levels in the presence of accessory subunits α2δ1 and ß4. Additionally, AnkB p.E1458G decreased surface Cav2.1 irrespective of the presence of accessory subunits. In addition, we found that partial deletion of AnkB in cortex resulted in a decrease in overall Cav2.1 levels, with no change to the levels of Cav2.1 detected in synaptosome fractions. Our work suggests that depending on the particular variant, AnkB regulates intracellular and surface Cav2.1. Notably, expression of the AnkB variant associated with seizure (AnkB p.S646F) caused further increase in intracellular Cav2.1 levels above that of even wildtype AnkB. These novel findings have important implications for understanding the role of AnkB and Cav2.1 in the regulation of neuronal function in health and disease.


Assuntos
Anquirinas/metabolismo , Canais de Cálcio Tipo N/metabolismo , Membrana Celular/metabolismo , Espaço Intracelular/metabolismo , Proteínas Mutantes/metabolismo , Animais , Anquirinas/genética , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mutação/genética , Subunidades Proteicas/metabolismo , Sinapses/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA