Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Mol Sci ; 20(14)2019 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-31331039

RESUMO

Psychiatric disorders are mental, behavioral or emotional disorders. These conditions are prevalent, one in four adults suffer from any type of psychiatric disorders world-wide. It has always been observed that psychiatric disorders have a genetic component, however, new methods to sequence full genomes of large cohorts have identified with high precision genetic risk loci for these conditions. Psychiatric disorders include, but are not limited to, bipolar disorder, schizophrenia, autism spectrum disorder, anxiety disorders, major depressive disorder, and attention-deficit and hyperactivity disorder. Several risk loci for psychiatric disorders fall within genes that encode for voltage-gated calcium channels (CaVs). Calcium entering through CaVs is crucial for multiple neuronal processes. In this review, we will summarize recent findings that link CaVs and their auxiliary subunits to psychiatric disorders. First, we will provide a general overview of CaVs structure, classification, function, expression and pharmacology. Next, we will summarize tools to study risk loci associated with psychiatric disorders. We will examine functional studies of risk variations in CaV genes when available. Finally, we will review pharmacological evidence of the use of CaV modulators to treat psychiatric disorders. Our review will be of interest for those studying pathophysiological aspects of CaVs.


Assuntos
Canais de Cálcio/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Transtornos Mentais/genética , Animais , Biomarcadores , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio/química , Canais de Cálcio/metabolismo , Ensaios Clínicos como Assunto , Regulação da Expressão Gênica , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Transtornos Mentais/diagnóstico , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/metabolismo , Terapia de Alvo Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade , Resultado do Tratamento
2.
Hum Mol Genet ; 24(4): 987-93, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25296916

RESUMO

Using exome sequencing and linkage analysis in a three-generation family with a unique dominant myoclonus-dystonia-like syndrome with cardiac arrhythmias, we identified a mutation in the CACNA1B gene, coding for neuronal voltage-gated calcium channels CaV2.2. This mutation (c.4166G>A;p.Arg1389His) is a disruptive missense mutation in the outer region of the ion pore. The functional consequences of the identified mutation were studied using whole-cell and single-channel patch recordings. High-resolution analyses at the single-channel level showed that, when open, R1389H CaV2.2 channels carried less current compared with WT channels. Other biophysical channel properties were unaltered in R1389H channels including ion selectivity, voltage-dependent activation or voltage-dependent inactivation. CaV2.2 channels regulate transmitter release at inhibitory and excitatory synapses. Functional changes could be consistent with a gain-of-function causing the observed hyperexcitability characteristic of this unique myoclonus-dystonia-like syndrome associated with cardiac arrhythmias.


Assuntos
Canais de Cálcio Tipo N/genética , Distúrbios Distônicos/genética , Estudos de Associação Genética , Mutação , Potenciais de Ação , Canais de Cálcio Tipo N/metabolismo , Sinalização do Cálcio , Distúrbios Distônicos/diagnóstico , Exoma , Feminino , Ligação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Técnicas de Patch-Clamp , Linhagem , Fenótipo
3.
Biochim Biophys Acta ; 1828(7): 1522-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23022282

RESUMO

Neuronal voltage-gated calcium channels generate rapid, transient intracellular calcium signals in response to membrane depolarization. Neuronal Ca(V) channels regulate a range of cellular functions and are implicated in a variety of neurological and psychiatric diseases including epilepsy, Parkinson's disease, chronic pain, schizophrenia, and bipolar disorder. Each mammalian Cacna1 gene has the potential to generate tens to thousands of Ca(V) channels by alternative pre-mRNA splicing, a process that adds fine granulation to the pool of Ca(V) channel structures and functions. The precise composition of Ca(V) channel splice isoform mRNAs expressed in each cell are controlled by cell-specific splicing factors. The activity of splicing factors are in turn regulated by molecules that encode various cellular features, including cell-type, activity, metabolic states, developmental state, and other factors. The cellular and behavioral consequences of individual sites of Ca(V) splice isoforms are being elucidated, as are the cell-specific splicing factors that control splice isoform selection. Altered patterns of alternative splicing of Ca(V) pre-mRNAs can alter behavior in subtle but measurable ways, with the potential to influence drug efficacy and disease severity. This article is part of a Special Issue entitled: Calcium channels.


Assuntos
Processamento Alternativo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Ativação do Canal Iônico/fisiologia , Transdução de Sinais , Animais , Humanos
4.
J Neurosci ; 32(30): 10365-9, 2012 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-22836269

RESUMO

Ca(V)2.2 (N-type) calcium channels control the entry of calcium into neurons to regulate essential functions but most notably presynaptic transmitter release. Ca(V)2.2 channel expression levels are precisely controlled, but we know little of the cellular mechanisms involved. The ubiquitin proteasome system (UPS) is known to regulate expression of many synaptic proteins, including presynaptic elements, to optimize synaptic efficiency. However, we have limited information about ubiquitination of Ca(V)2 channels. Here we show that Ca(V)2.2 proteins are ubiquitinated, and that elements in the proximal C terminus of Ca(V)2.2 encoded by exon 37b of the mouse Cacna1b gene predispose cloned and native channels to downregulation by the UPS. Ca(V)2.2 channels containing e37b are expressed throughout the mammalian nervous system, but in some cells, notably nociceptors, sometimes e37a--not e37b--is selected during alternative splicing of Ca(V)2.2 pre-mRNA. By a combination of biochemical and functional analyses we show e37b promotes a form of ubiquitination that is coupled to reduced Ca(V)2.2 current density and increased sensitivity to the UPS. Cell-specific alternative splicing of e37a in nociceptors reduces Ca(V)2.2 channel ubiquitination and sensitivity to the UPS, suggesting a role in pain processing.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitinação/fisiologia , Processamento Alternativo , Animais , Canais de Cálcio Tipo N/genética , Gânglios Espinais/metabolismo , Camundongos , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
Mol Pain ; 9: 67, 2013 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-24369063

RESUMO

Presynaptic voltage-gated calcium Ca(V)2.2 channels play a privileged role in spinal level sensitization following peripheral nerve injury. Direct and indirect inhibitors of Ca(V)2.2 channel activity in spinal dorsal horn are analgesic in chronic pain states. Ca(V)2.2 channels represent a family of splice isoforms that are expressed in different combinations according to cell-type. A pair of mutually exclusive exons in the Ca(V)2.2 encoding Cacna1b gene, e37a and e37b, differentially influence morphine analgesia. In mice that lack exon e37a, which is enriched in nociceptors, the analgesic efficacy of intrathecal morphine against noxious thermal stimuli is reduced. Here we ask if sequences unique to e37a influence: the development of abnormal thermal and mechanical sensitivity associated with peripheral nerve injury; and the actions of two other classes of analgesics that owe part or all of their efficacy to Ca(V)2.2 channel inhibition. We find that: i) the analgesic efficacy of morphine, but not ziconotide or gabapentin, is reduced in mice lacking e37a, ii) the induction and maintenance of behaviors associated with sensitization that accompany peripheral nerve injury, do not require e37a-specific sequence, iii) intrathecal morphine, but not ziconotide or gabapentin analgesia to thermal stimuli is significantly lower in wild-type mice after peripheral nerve injury, iv) the analgesic efficacy of ziconotide and gabapentin to mechanical stimuli is reduced following nerve injury, and iv) intrathecal morphine analgesia to thermal stimuli in mice lacking e37a is not further reduced by peripheral nerve injury. Our findings show that the analgesic action of morphine, but not ziconotide or gabapentin, to thermal stimuli is linked to which Cacna1b exon, e37a or e37b, is selected during alternative pre-mRNA splicing.


Assuntos
Processamento Alternativo/genética , Aminas/uso terapêutico , Canais de Cálcio Tipo N/genética , Ácidos Cicloexanocarboxílicos/uso terapêutico , Morfina/uso terapêutico , Ácido gama-Aminobutírico/uso terapêutico , ômega-Conotoxinas/uso terapêutico , Processamento Alternativo/fisiologia , Analgesia , Animais , Gabapentina , Masculino , Camundongos , Neuralgia/tratamento farmacológico , Precursores de RNA/genética
6.
Biosensors (Basel) ; 13(11)2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-37998158

RESUMO

Serotonin (5-HT) is a critical neurotransmitter involved in many neuronal functions, and 5-HT depletion has been linked to several mental diseases. The fast release and clearance of serotonin in the extracellular space, low analyte concentrations, and a multitude of interfering species make the detection of serotonin challenging. This work presents an electrochemical aptamer-based biosensing platform that can monitor 5-HT continuously with high sensitivity and selectivity. Our electrochemical sensor showed a response time of approximately 1 min to a step change in the serotonin concentration in continuous monitoring using a single-frequency EIS (electrochemical impedance spectroscopy) technique. The developed sensing platform was able to detect 5-HT in the range of 25-150 nM in the continuous sample fluid flow with a detection limit (LOD) of 5.6 nM. The electrochemical sensor showed promising selectivity against other species with similar chemical structures and redox potentials, including dopamine (DA), norepinephrine (NE), L-tryptophan (L-TP), 5-hydroxyindoleacetic acid (5-HIAA), and 5-hydroxytryptophan (5-HTP). The proposed sensing platform is able to achieve high selectivity in the nanomolar range continuously in real-time, demonstrating the potential for monitoring serotonin from neurons in organ-on-a-chip or brain-on-a-chip-based platforms.


Assuntos
Técnicas Biossensoriais , Serotonina , Dopamina , Norepinefrina , Triptofano , Oligonucleotídeos , Técnicas Eletroquímicas
7.
bioRxiv ; 2023 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-37693414

RESUMO

Cell-specific alternative splicing of Cacna1b pre-mRNA generates functionally distinct voltage-gated CaV2.2 channels. CaV2.2 channels mediate the release of glutamate from nociceptor termini in the dorsal horn spinal cord and they are implicated in chronic pain. One alternatively spliced exon in Cacna1b, e37a, is highly expressed in dorsal root ganglia, relative to other regions of the nervous system, and it is particularly important in inflammatory hyperalgesia. Here we studied the effects of two ω-phonetoxins, PnTx3-4 and Phα1ß, derived from the spider Phoneutria nigriventer on CaV2.2 channel isoforms of dorsal root ganglia (CaV2.2 e37a and CaV2.2 e37b). Both PnTx3-4 and Phα1ß are known to have analgesic effects in rodent models of pain and to inhibit CaV2.2 channels. CaV2.2 e37a and CaV2.2 e37b isoforms expressed in a mammalian cell line were inhibited by PnTx3-4 and Phα1ß with similar potency and with similar timecourse, although CaV2.2 e37a currents were slightly, but consistently more sensitive to toxin inhibition compared to CaV2.2 e37b. The inhibitory effects of PnTx3-4 and Phα1ß on CaV2.2-e37a and CaV2.2-e37b channels were voltage-dependent, and both occlude the inhibitory effects of ω-conotoxin GVIA, consistent with a common site of action. The potency of PnTx3-4 and Phα1ß on both major splice isoforms in dorsal root ganglia constribute to understanding the analgesic actions of these ω-phonetoxins.

8.
Methods Mol Biol ; 2537: 185-196, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35895265

RESUMO

Defining the cell-specific alternative splicing landscape in complex tissues is an important goal to gain functional insights. Deep-sequencing techniques coupled to genetic strategies for cell identification has provided important cues on cell-specific exon usage in complex tissues like the nervous system. BaseScope™ has emerged as a powerful and highly sensitive alternative to in situ hybridization to determine exon composition in tissue with spatial and morphological context. In this protocol, we will review how BaseScope was utilized to detect the e37a-Cacna1b splice variant of the presynaptic calcium channel CaV2.2 or N-type. This splice variant arises from a pair of mutually exclusive exons (e37a and e37b). E37a-Cacna1b is heavily underrepresented relative to e37b-Cacna1b and both exons share 60% of their sequence. By using BaseScope™, we were able to discover that e37a-Cacna1b is expressed in excitatory pyramidal neurons of hippocampus and cortex, as well as motor neurons of the ventral horn of the spinal cord.


Assuntos
Processamento Alternativo , Canais de Cálcio Tipo N , Canais de Cálcio Tipo N/genética , Éxons , Hibridização In Situ , RNA Mensageiro/genética
9.
OBM Neurobiol ; 5(1)2021.
Artigo em Inglês | MEDLINE | ID: mdl-33521586

RESUMO

N-type (CaV2.2) calcium channels are key for action potential-evoked transmitter release in the peripheral and central nervous system. Previous studies have highlighted the functional relevance of N-type calcium channels at both the peripheral and central level. In the periphery, the N-type calcium channels regulate nociceptive and sympathetic responses. At the central level, N-type calcium channels have been linked to aggression, hyperlocomotion, and anxiety. Among the areas of the brain that are involved in anxiety are the basolateral amygdala, medial prefrontal cortex, and ventral hippocampus. These three areas share similar characteristics in their neuronal circuitry, where pyramidal projection neurons are under the inhibitory control of a wide array of interneurons including those that express the peptide cholecystokinin. This type of interneuron is well-known to rely on N-type calcium channels to release GABA in the hippocampus, however, whether these channels control GABA release from cholecystokinin-expressing interneurons in the basolateral amygdala and medial prefrontal cortex is not known. Here, using mouse models to genetically label cholecystokinin-expressing interneurons and electrophysiology, we found that in the basolateral amygdala, N-type calcium channels control ~50% of GABA release from these neurons onto pyramidal cells. By contrast, in the medial prefrontal cortex N-type calcium channels are functionally absent in synapses of cholecystokinin-expressing interneurons, but control ~40% of GABA release from other types of interneurons. Our findings provide insights into the precise localization of N-type calcium channels in interneurons of brain areas related to anxiety.

10.
Biol Psychiatry ; 89(3): 256-269, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33032807

RESUMO

BACKGROUND: Neurogranin (Ng), encoded by the schizophrenia risk gene NRGN, is a calmodulin-binding protein enriched in the postsynaptic compartments, and its expression is reduced in the postmortem brains of patients with schizophrenia. Experience-dependent translation of Ng is critical for encoding contextual memory, and Ng regulates developmental plasticity in the primary visual cortex during the critical period. However, the overall impact of Ng on the neuronal signaling that regulates synaptic plasticity is unknown. METHODS: Altered Ng expression was achieved via virus-mediated gene manipulation in mice. The effect on long-term potentiation (LTP) was accessed using spike timing-dependent plasticity protocols. Quantitative phosphoproteomics analyses led to discoveries in significant phosphorylated targets. An identified candidate was examined with high-throughput planar patch clamp and was validated with pharmacological manipulation. RESULTS: Ng bidirectionally modulated LTP in the hippocampus. Decreasing Ng levels significantly affected the phosphorylation pattern of postsynaptic density proteins, including glutamate receptors, GTPases, kinases, RNA binding proteins, selective ion channels, and ionic transporters, some of which highlighted clusters of schizophrenia- and autism-related genes. Hypophosphorylation of NMDA receptor subunit Grin2A, one significant phosphorylated target, resulted in accelerated decay of NMDA receptor currents. Blocking protein phosphatase PP2B activity rescued the accelerated NMDA receptor current decay and the impairment of LTP mediated by Ng knockdown, implicating the requirement of synaptic PP2B activity for the deficits. CONCLUSIONS: Altered Ng levels affect the phosphorylation landscape of neuronal proteins. PP2B activity is required for mediating the deficit in synaptic plasticity caused by decreasing Ng levels, revealing a novel mechanistic link of a schizophrenia risk gene to cognitive deficits.


Assuntos
Neurogranina , Esquizofrenia , Animais , Calmodulina/metabolismo , Hipocampo/metabolismo , Humanos , Potenciação de Longa Duração , Camundongos , Neurogranina/genética , Neurogranina/metabolismo , Plasticidade Neuronal , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/genética , Sinapses/metabolismo
11.
FEBS Open Bio ; 9(9): 1603-1616, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31314171

RESUMO

Presynaptic CaV 2.2 (N-type) channels are fundamental for transmitter release across the nervous system. The gene encoding CaV 2.2 channels, Cacna1b, contains alternatively spliced exons that result in functionally distinct splice variants (e18a, e24a, e31a, and 37a/37b). Alternative splicing of the cassette exon 18a generates two mRNA transcripts (+e18a-Cacna1b and ∆e18a-Cacna1b). In this study, using novel mouse genetic models and in situ hybridization (BaseScope™), we confirmed that +e18a-Cacna1b splice variants are expressed in monoaminergic regions of the midbrain. We expanded these studies and identified +e18a-Cacna1b mRNA in deep cerebellar cells and spinal cord motor neurons. Furthermore, we determined that +e18a-Cacna1b is enriched in cholecystokinin-expressing interneurons. Our results provide key information to understand cell-specific functions of CaV 2.2 channels.


Assuntos
Canais de Cálcio Tipo N/química , Canais de Cálcio Tipo N/genética , Citoplasma/genética , Processamento Alternativo/genética , Animais , Sistema Nervoso Central/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Especificidade de Órgãos , RNA Mensageiro/genética
12.
Mol Brain ; 12(1): 81, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31630675

RESUMO

Presynaptic CaV2.2 channels control calcium entry that triggers neurotransmitter release at both central and peripheral synapses. The Cacna1b gene encodes the α1-pore forming subunit of CaV2.2 channels. Distinct subsets of splice variants of CaV2.2 derived from cell-specific alternative splicing of the Cacna1b pre-mRNA are expressed in specific subpopulations of neurons. Four cell-specific sites of alternative splicing in Cacna1b that alter CaV2.2 channel function have been described in detail: three cassette exons (e18a, e24a, and e31a) and a pair of mutually exclusive exons (e37a/e37b). Cacna1b mRNAs containing e37a are highly enriched in a subpopulation of nociceptors where they influence nociception and morphine analgesia. E37a-Cacna1b mRNAs are also expressed in brain, but their cell-specific expression in this part of the nervous system, their functional consequences in central synapses and their role on complex behavior have not been studied. In this report, we show that e37a-Cacna1b mRNAs are expressed in excitatory projection neurons where CaV2.2 channels are known to influence transmitter release at excitatory inputs from entorhinal cortex (EC) to dentate gyrus (DG). By comparing behaviors of WT mice to those that only express e37b-CaV2.2 channels, we found evidence that e37a-CaV2.2 enhances behavioral responses to aversive stimuli. Our results suggest that alternative splicing of Cacna1b e37a influences excitatory transmitter release and couples to complex behaviors.


Assuntos
Processamento Alternativo/genética , Comportamento Animal , Canais de Cálcio Tipo N/genética , Transmissão Sináptica/genética , Animais , Encéfalo/metabolismo , Canais de Cálcio Tipo N/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Estimulação Física , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sinapses/metabolismo
13.
J Neurosci ; 27(12): 3317-27, 2007 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-17376992

RESUMO

Auxiliary gamma subunits are an important component of high-voltage-activated calcium (Ca(V)) channels, but their precise regulatory role remains to be determined. In the current report, we have used complementary approaches including molecular biology and electrophysiology to investigate the influence of the gamma subunits on neuronal Ca(V) channel activity and expression. We found that coexpression of gamma2 or gamma3 subunits drastically inhibited macroscopic currents through recombinant N-type channels (Ca(V)2.2/beta3/alpha2delta) in HEK-293 cells. Using inhibitors of internalization, we found that removal of functional channels from the plasma membrane is an improbable mechanism of current regulation by gamma. Instead, changes in current amplitude could be attributed to two distinct mechanisms. First, gamma subunit expression altered the voltage dependence of channel activity. Second, gamma subunit expression reduced N-type channel synthesis via activation of the endoplasmic reticulum unfolded protein response. Together, our findings (1) corroborate that neuronal gamma subunits significantly downregulate Ca(V)2.2 channel activity, (2) uncover a role for the gamma2 subunit in Ca(V)2.2 channel expression through early components of the biosynthetic pathway, and (3) suggest that, under certain conditions, channel protein misfolding could be induced by interactions with the gamma subunits, supporting the notion that Ca(V) channels constitute a class of difficult-to-fold proteins.


Assuntos
Canais de Cálcio/fisiologia , Dobramento de Proteína , Subunidades Proteicas/fisiologia , Proteínas Recombinantes/antagonistas & inibidores , Animais , Canais de Cálcio/química , Canais de Cálcio/genética , Linhagem Celular , Retículo Endoplasmático/química , Retículo Endoplasmático/genética , Retículo Endoplasmático/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Subunidades Proteicas/química , Subunidades Proteicas/genética , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
14.
J Neurosci Res ; 86(15): 3456-68, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18627030

RESUMO

Glial and glia-derived cells express a variety of receptors for neurotransmitters and hormones, the majority of which evoke both Ca(2+) release from intracellular stores and Ca(2+) entry across the plasma membrane. We investigated the links between histamine H(1) receptor activation, Ca(2+) release from intracellular stores and Ca(2+) influx in human astrocytoma U373 MG cells. Histamine, through a H(1) receptor-mediated effect, evoked an increase in cytoplasmic free calcium concentration ([Ca(2+)](i)) that occurred in two phases: an initial, transient, increase owing to Ca(2+) mobilization from intracellular pools, and a second, sustained increase dependent on both Ca(2+) influx and continuous receptor occupancy. The characteristics of histamine-induced increases in [Ca(2+)](i) were similar to the capacitative entry evoked by emptying of the Ca(2+) stores with thapsigargine, and different from that observed when Ca(2+) influx was activated with OAG (1-oleoyl-2-acetyl-sn-glycerol), a diacylglycerol (DAG) analog. OAG application or increased endogenous DAG, resulting from DAG kinase inhibition, reduced the histamine-induced response. Furthermore, activation of the DAG target, protein kinase C (PKC), by TPA (12-O-tetradecanoyl 4beta-phorbol 13alpha-acetate) resulted in inhibition of the histamine-induced Ca(2+) response, an action prevented by PKC inhibitors. By using reverse transcriptase-polymerase chain reaction analysis, mRNAs for transient receptor potential channels (TRPCs) 1, 4, and 6 as well as for STIM1 (stromal-interacting molecule) and Orai1 were found to be expressed in the U373 MG cells, and confocal microscopy using specific antibodies revealed the presence of the corresponding proteins. Therefore, TRPCs may be candidate proteins forming store-operated channels in the U373 MG cell line. Further, our results confirm the involvement of PKC in the regulation of H(1) receptor-induced responses and point out to the existence of a feedback mechanism acting via PKC to limit the increase in [Ca(2+)](i).


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Histamina/metabolismo , Neuroglia/metabolismo , Canais de Cátion TRPC/metabolismo , Astrocitoma/metabolismo , Linhagem Celular Tumoral , Diacilglicerol Quinase/metabolismo , Expressão Gênica , Humanos , Imuno-Histoquímica , Proteínas de Membrana/metabolismo , Microscopia Confocal , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Proteína Quinase C/metabolismo , Receptores Histamínicos H1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/genética
15.
FEBS Lett ; 581(23): 4430-8, 2007 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-17719582

RESUMO

We previously reported that expression of myotonic dystrophy (DM1) expanded CUG repeats impedes NGF-induced differentiation in a PC12 clone (CTG90 cells). Here, we present evidence for changes in the fractional contribution of distinct voltage-gated Ca(2+) channels, key elements in neurotrophin-promoted differentiation, to the total Ca(2+) current in the CTG90 cells. Patch-clamp recordings showed that the relative proportion of pharmacologically isolated Ca(2+) channel types differed between control and CTG90 cells. Particularly, the functional expression of N-type channels was significantly reduced. Though quantitative real-time RT-PCR revealed that transcripts for the pore-forming subunit encoding the N-type channels remained unchanged, the protein level analyzed by semi-quantitative Western blotting was down-regulated in the CTG90 cells. These data suggest modifications in the processing of N-type Ca(2+) channels in PC12 cells expressing the DM1 mutation.


Assuntos
Canais de Cálcio/genética , Proteínas Serina-Treonina Quinases/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Western Blotting , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Canais de Cálcio/fisiologia , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio Tipo N/fisiologia , Eletrofisiologia , Potenciais da Membrana/efeitos dos fármacos , Microscopia de Contraste de Fase , Mutação , Miotonina Proteína Quinase , Nifedipino/farmacologia , Células PC12 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Expansão das Repetições de Trinucleotídeos/fisiologia
16.
Cell Calcium ; 66: 1-9, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28807144

RESUMO

cGMP is a second messenger widely used in the nervous system and other tissues. One of the major effectors for cGMP is the serine/threonine protein kinase, cGMP-dependent protein kinase (PKG), which catalyzes the phosphorylation of a variety of proteins including ion channels. Previously, it has been shown that the cGMP-PKG signaling pathway inhibits Ca2+ currents in rat vestibular hair cells and chromaffin cells. This current allegedly flow through voltage-gated CaV1.3L-type Ca2+ channels, and is important for controlling vestibular hair cell sensory function and catecholamine secretion, respectively. Here, we show that native L-type channels in the insulin-secreting RIN-m5F cell line, and recombinant CaV1.3 channels heterologously expressed in HEK-293 cells, are regulatory targets of the cGMP-PKG signaling cascade. Our results indicate that the CaVα1 ion-conducting subunit of the CaV1.3 channels is highly expressed in RIN-m5F cells and that the application of 8-Br-cGMP, a membrane-permeable analogue of cGMP, significantly inhibits Ca2+ macroscopic currents and impair insulin release stimulated with high K+. In addition, KT-5823, a specific inhibitor of PKG, prevents the current inhibition generated by 8-Br-cGMP in the heterologous expression system. Interestingly, mutating the putative phosphorylation sites to residues resistant to phosphorylation showed that the relevant PKG sites for CaV1.3 L-type channel regulation centers on two amino acid residues, Ser793 and Ser860, located in the intracellular loop connecting the II and III repeats of the CaVα1 pore-forming subunit of the channel. These findings unveil a novel mechanism for how the cGMP-PKG signaling pathway may regulate CaV1.3 channels and contribute to regulate insulin secretion.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Insulina/metabolismo , Transdução de Sinais , Animais , Canais de Cálcio Tipo L/genética , Carbazóis/farmacologia , Linhagem Celular , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Células HEK293 , Humanos , Potenciais da Membrana/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Óxido Nítrico/metabolismo , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
17.
eNeuro ; 4(5)2017.
Artigo em Inglês | MEDLINE | ID: mdl-29067356

RESUMO

The majority of multiexon mammalian genes contain alternatively spliced exons that have unique expression patterns in different cell populations and that have important cell functions. The expression profiles of alternative exons are controlled by cell-specific splicing factors that can promote exon inclusion or exon skipping but with few exceptions we do not know which specific splicing factors control the expression of alternatively spliced exons of known biological function. Many ion channel genes undergo extensive alternative splicing including Cacna1b that encodes the voltage-gated CaV2.2 α1 subunit. Alternatively spliced exon 18a in Cacna1b RNA encodes 21 amino acids in the II-III loop of CaV2.2, and its expression differs across the nervous system and over development. Genome-wide, protein-RNA binding analyses coupled to high-throughput RNA sequencing show that RNA binding Fox (Rbfox) proteins associate with CaV2.2 (Cacna1b) pre-mRNAs. Here, we link Rbfox2 to suppression of e18a. We show increased e18a inclusion in CaV2.2 mRNAs: (1) after siRNA knockdown of Rbfox2 in a neuronal cell line and (2) in RNA from sympathetic neurons of adult compared to early postnatal mice. By immunoprecipitation of Rbfox2-RNA complexes followed by qPCR, we demonstrate reduced Rbfox2 binding upstream of e18a in RNA from sympathetic neurons of adult compared to early postnatal mice. CaV2.2 currents in cell lines and in sympathetic neurons expressing only e18a-CaV2.2 are larger compared to currents from those expressing only Δ18a-CaV2.2. We conclude that Rbfox2 represses e18a inclusion during pre-mRNA splicing of CaV2.2, limiting the size of CaV2.2 currents early in development in certain neuronal populations.


Assuntos
Canais de Cálcio Tipo N/genética , Éxons/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Neurônios/fisiologia , Fatores de Processamento de RNA/metabolismo , RNA Mensageiro/metabolismo , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo N/metabolismo , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Fatores de Processamento de RNA/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Gânglio Cervical Superior/citologia
18.
Curr Mol Pharmacol ; 8(1): 22-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25966698

RESUMO

Voltage-gated calcium ion channels are essential for numerous biological functions of excitable cells and there is wide spread appreciation of their importance as drug targets in the treatment of many disorders including those of cardiovascular and nervous systems. Each Cacna1 gene has the potential to generate a number of structurally, functionally, and in some cases pharmacologically unique CaVα1 subunits through alternative pre-mRNA splicing and the use of alternate promoters. Analyses of rapidly emerging deep sequencing data for a range of human tissue transcriptomes contain information to quantify tissue-specific and alternative exon usage patterns for Cacna1 genes. Cellspecific actions of nuclear DNA and RNA binding proteins control the use of alternate promoters and the selection of alternate exons during pre-mRNA splicing, and they determine the spectrum of protein isoforms expressed within different types of cells. Amino acid compositions within discrete protein domains can differ substantially among CaV isoforms expressed in different tissues, and such differences may be greater than those that exist across CaV channel homologs of closely related species. Here we highlight examples of CaV isoforms that have unique expression patterns and that exhibit different pharmacological sensitivities. Knowledge of expression patterns of CaV isoforms in different human tissues, cell populations, ages, and disease states should inform strategies aimed at developing the next generation of CaV channel inhibitors and agonists with improved tissue-specificity.


Assuntos
Processamento Alternativo , Canais de Cálcio/genética , Animais , Canais de Cálcio/efeitos dos fármacos , Humanos
19.
FEBS Lett ; 576(1-2): 21-6, 2004 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-15474003

RESUMO

The CaValpha2delta auxiliary subunit is a glycosylated protein that regulates the trafficking and function of voltage-gated Ca2+ channels. One of the most prominent roles of CaValpha2delta is to increase whole-cell Ca2+ current amplitude. Using N-glycosidase F and truncated forms of CaValpha2delta, earlier studies suggested an important role for N-linked glycosylation in current stimulation. Here, we used site-directed mutagenesis and heterologous expression in HEK-293 cells to examine the impact of individual glycosylation sites within the CaValpha2delta subunit on the regulation of Ba2+ currents through recombinant Ca2+ channels. We found two N-glycosylation consensus sites (NX(S/T)) in the extracellular alpha2 domain of the protein that are functional. Substitution of asparagines for glutamines at amino acid positions 136 and 184 rendered these sites non-functional as shown by patch-clamp experiments. These results corroborate that N-glycosylation is required for the CaValpha2delta subunit-induced current stimulation and suggest that sites N136 and N184 are directly involved in this action. Likewise, N136Q and N184Q mutations prevented whole-cell current stimulation without altering its kinetic properties, suggesting a regulation on the number of functional channels at the plasma membrane.


Assuntos
Asparagina/metabolismo , Canais de Cálcio/química , Canais de Cálcio/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Bário/metabolismo , Canais de Cálcio/genética , Linhagem Celular , Sequência Consenso , Genes Reporter , Glicosilação , Humanos , Ativação do Canal Iônico , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/genética , Mutação Puntual , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Regulação para Cima
20.
Wiley Interdiscip Rev Membr Transp Signal ; 2(5): 207-220, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24949251

RESUMO

Voltage-gated Ca2+ (CaV) channels mediate Ca2+ ions influx into cells in response to depolarization of the plasma membrane. They are responsible for initiation of excitation-contraction and excitation-secretion coupling, and the Ca2+ that enters cells through this pathway is also important in the regulation of protein phosphorylation, gene transcription, and many other intracellular events. Initial electrophysiological studies divided CaV channels into low-voltage-activated (LVA) and high-voltage-activated (HVA) channels. The HVA CaV channels were further subdivided into L, N, P/Q, and R-types which are oligomeric protein complexes composed of an ion-conducting CaVα1 subunit and auxiliary CaVα2δ, CaVß, and CaVγ subunits. The functional consequences of the auxiliary subunits include altered functional and pharmacological properties of the channels as well as increased current densities. The latter observation suggests an important role of the auxiliary subunits in membrane trafficking of the CaVα1 subunit. This includes the mechanisms by which CaV channels are targeted to the plasma membrane and to appropriate regions within a given cell. Likewise, the auxiliary subunits seem to participate in the mechanisms that remove CaV channels from the plasma membrane for recycling and/or degradation. Diverse studies have provided important clues to the molecular mechanisms involved in the regulation of CaV channels by the auxiliary subunits, and the roles that these proteins could possibly play in channel targeting and membrane Stabilization.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA