Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(7)2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-32244818

RESUMO

Many epilepsy patients are refractory to conventional antiepileptic drugs. Resurgent and persistent currents can be enhanced by epilepsy mutations in the Nav1.2 channel, but conventional antiepileptic drugs inhibit normal transient currents through these channels, along with aberrant resurgent and persistent currents that are enhanced by Nav1.2 epilepsy mutations. Pharmacotherapies that specifically target aberrant resurgent and/or persistent currents would likely have fewer unwanted side effects and be effective in many patients with refractory epilepsy. This study investigated the effects of cannbidiol (CBD) and GS967 (each at 1 µM) on transient, resurgent, and persistent currents in human embryonic kidney (HEK) cells stably expressing wild-type hNav1.2 channels. We found that CBD preferentially inhibits resurgent currents over transient currents in this paradigm; and that GS967 preferentially inhibits persistent currents over transient currents. Therefore, CBD and GS967 may represent a new class of more targeted and effective antiepileptic drugs.


Assuntos
Canabidiol/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Piridinas/farmacologia , Triazóis/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Anticonvulsivantes/farmacologia , Epilepsia/tratamento farmacológico , Epilepsia/genética , Epilepsia/fisiopatologia , Células HEK293 , Humanos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Camundongos , Mutação , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia
2.
Mol Autism ; 11(1): 23, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32264956

RESUMO

Efforts to identify the causes of autism spectrum disorders have highlighted the importance of both genetics and environment, but the lack of human models for many of these disorders limits researchers' attempts to understand the mechanisms of disease and to develop new treatments. Induced pluripotent stem cells offer the opportunity to study specific genetic and environmental risk factors, but the heterogeneity of donor genetics may obscure important findings. Diseases associated with unusually high rates of autism, such as SCN2A syndromes, provide an opportunity to study specific mutations with high effect sizes in a human genetic context and may reveal biological insights applicable to more common forms of autism. Loss-of-function mutations in the SCN2A gene, which encodes the voltage-gated sodium channel NaV1.2, are associated with autism rates up to 50%. Here, we review the findings from experimental models of SCN2A syndromes, including mouse and human cell studies, highlighting the potential role for patient-derived induced pluripotent stem cell technology to identify the molecular and cellular substrates of autism.


Assuntos
Transtorno do Espectro Autista/fisiopatologia , Canalopatias/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Células-Tronco Pluripotentes , Animais , Humanos
3.
Neuron ; 103(4): 673-685.e5, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31230762

RESUMO

Autism spectrum disorder (ASD) is strongly associated with de novo gene mutations. One of the most commonly affected genes is SCN2A. ASD-associated SCN2A mutations impair the encoded protein NaV1.2, a sodium channel important for action potential initiation and propagation in developing excitatory cortical neurons. The link between an axonal sodium channel and ASD, a disorder typically attributed to synaptic or transcriptional dysfunction, is unclear. Here we show that NaV1.2 is unexpectedly critical for dendritic excitability and synaptic function in mature pyramidal neurons in addition to regulating early developmental axonal excitability. NaV1.2 loss reduced action potential backpropagation into dendrites, impairing synaptic plasticity and synaptic strength, even when NaV1.2 expression was disrupted in a cell-autonomous fashion late in development. These results reveal a novel dendritic function for NaV1.2, providing insight into cellular mechanisms probably underlying circuit and behavioral dysfunction in ASD.


Assuntos
Transtorno do Espectro Autista/genética , Dendritos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Potenciais de Ação , Animais , Sinalização do Cálcio , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Potenciais Pós-Sinápticos em Miniatura/fisiologia , N-Metilaspartato/análise , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Neocórtex/citologia , Neocórtex/crescimento & desenvolvimento , Neocórtex/fisiologia , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Engenharia de Proteínas , Comportamento Social , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/análise
4.
Mol Med ; 25(1): 6, 2019 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-30813884

RESUMO

BACKGROUND: Deleterious variants in the voltage-gated sodium channel type 2 (Nav1.2) lead to a broad spectrum of phenotypes ranging from benign familial neonatal-infantile epilepsy (BFNIE), severe developmental and epileptic encephalopathy (DEE) and intellectual disability (ID) to autism spectrum disorders (ASD). Yet, the underlying mechanisms are still incompletely understood. METHODS: To further elucidate the genotype-phenotype correlation of SCN2A variants we investigated the functional effects of six variants representing the phenotypic spectrum by whole-cell patch-clamp studies in transfected HEK293T cells and in-silico structural modeling. RESULTS: The two variants p.L1342P and p.E1803G detected in patients with early onset epileptic encephalopathy (EE) showed profound and complex changes in channel gating, whereas the BFNIE variant p.L1563V exhibited only a small gain of channel function. The three variants identified in ID patients without seizures, p.R937C, p.L611Vfs*35 and p.W1716*, did not produce measurable currents. Homology modeling of the missense variants predicted structural impairments consistent with the electrophysiological findings. CONCLUSIONS: Our findings support the hypothesis that complete loss-of-function variants lead to ID without seizures, small gain-of-function variants cause BFNIE and EE variants exhibit variable but profound Nav1.2 gating changes. Moreover, structural modeling was able to predict the severity of the variant impact, supporting a potential role of structural modeling as a prognostic tool. Our study on the functional consequences of SCN2A variants causing the distinct phenotypes of EE, BFNIE and ID contributes to the elucidation of mechanisms underlying the broad phenotypic variability reported for SCN2A variants.


Assuntos
Epilepsia Neonatal Benigna/genética , Síndromes Epilépticas/genética , Deficiência Intelectual/genética , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Adolescente , Criança , Epilepsia Neonatal Benigna/fisiopatologia , Síndromes Epilépticas/fisiopatologia , Estudos de Associação Genética , Células HEK293 , Humanos , Deficiência Intelectual/fisiopatologia , Fenótipo , Adulto Jovem
5.
Psychiatr Genet ; 29(3): 91-94, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30741786

RESUMO

The sodium voltage-gated channel α subunit 2 (SCN2A) gene encodes a subunit of sodium voltage-gated channels expressed primarily in the central nervous system that are responsible for action potential initiation and propagation in excitable cells. SCN2A mutations underlie a spectrum of distinct phenotypes, including seizure disorders, neurodevelopmental disorders, and rarer instances of episodic ataxia and schizophrenia. We report on a 38-year-old patient with adult-onset psychotic symptoms on a background of infantile-onset seizures, autistic features and episodic ataxia. Whole-exome sequencing revealed a de-novo novel SCN2A mutation (c.4966T > C, p.Ser1656Pro). This and other SCN2A mutations associated with the schizophrenia phenotype overlap those seen in neurodevelopmental disorders, suggesting a common underlying mechanism. This is the first report of a patient with the entire known SCN2A phenotypic spectrum. We highlight the importance of recognizing the psychiatric phenotypes associated with SCN2A mutations and that the phenotypic spectrum is more fluid, and less categorical, than previously thought.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.2/genética , Esquizofrenia/genética , Esquizofrenia/fisiopatologia , Adulto , Ataxia/genética , Epilepsia/genética , Humanos , Masculino , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Transtornos do Neurodesenvolvimento/genética , Fenótipo , Psicologia do Esquizofrênico , Sequenciamento do Exoma
6.
Brain Dev ; 41(4): 389-391, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30415926

RESUMO

SCN2A mutations have been described in a very broad spectrum of clinical phenotypes including benign (familial) neonatal/infantile seizures and early infantile epileptic encephalopathies (EIEE) as Ohtahara syndrome (OS), Dravet syndrome (DS), epilepsy of infancy with migrating focal seizures and West syndrome (WS). Treatment modalities for epilepsy caused by SCN2A mutations mainly consist of sodium channel blockers but ketogenic diet (KD) is also considered as an option of treatment for intractible seizures caused by SCN2A mutations. Because of the wide nature of the heterogeneity of mutations related to SCN2A gene, the clinical phenotypes vary in severity and treatment response to KD has been reported to be controversial. We present a patient diagnosed with OS associated with a novel SCN2A mutation (c.408G > A, p.Met136lle; OMIM®: 182390) who had a complete resolution of seizures and EEG abnormalities with KD commenced at 39 days of age. As far as we are aware our case is the youngest patient with SCN2A mutation treated with KD with complete resolution of epilepsy at an early age and has been seizure free of antiepileptic medications for a long duration.


Assuntos
Dieta Cetogênica/métodos , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Convulsões/terapia , Anticonvulsivantes/uso terapêutico , Encéfalo/fisiopatologia , Eletroencefalografia , Epilepsia/genética , Síndromes Epilépticas/genética , Feminino , Humanos , Lactente , Mutação , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Fenótipo , Convulsões/genética , Bloqueadores dos Canais de Sódio/uso terapêutico , Espasmos Infantis/genética , Espasmos Infantis/terapia
7.
Biol Pharm Bull ; 41(9): 1471-1474, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30175781

RESUMO

Escitalopram, a selective serotonin reuptake inhibitor (SSRI), may induce seizures, particularly in epileptic patients. In this study, we investigated the effect of escitalopram in Nav1.2 voltage-gated sodium channels (VGSCs) transfected HEK293 cells. Nav1.2 VGSCs current decreased by approximately 50.7±8.3% under treatment with 100 µM escitalopram. The IC50 of escitalopram against Nav1.2 VGSCs was 114.17 µM. Moreover, the treatment with 100 µM escitalopram changed the voltage-dependence of inactivation and the voltage at half-maximal inactivation shifted significantly from -50.3±3.7 to -56.7±6.0 mV toward negative potential under treatment with 100 µM escitalopram. Surprisingly, the treatment with 100 µM escitalopram also changed the voltage-dependence of activation and the voltage at half-maximal activation shifted significantly from -13.8±4.6 to -21.5±3.9 mV toward negative potential under treatment with 100 µM escitalopram. These findings suggested that escitalopram might be able to inhibit Nav1.2 VGSCs current and affects both activation and inactivation states of Nav1.2 VGSCs.


Assuntos
Citalopram/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Células HEK293 , Humanos
8.
Brain ; 140(5): 1316-1336, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28379373

RESUMO

Mutations in SCN2A, a gene encoding the voltage-gated sodium channel Nav1.2, have been associated with a spectrum of epilepsies and neurodevelopmental disorders. Here, we report the phenotypes of 71 patients and review 130 previously reported patients. We found that (i) encephalopathies with infantile/childhood onset epilepsies (≥3 months of age) occur almost as often as those with an early infantile onset (<3 months), and are thus more frequent than previously reported; (ii) distinct phenotypes can be seen within the late onset group, including myoclonic-atonic epilepsy (two patients), Lennox-Gastaut not emerging from West syndrome (two patients), and focal epilepsies with an electrical status epilepticus during slow sleep-like EEG pattern (six patients); and (iii) West syndrome constitutes a common phenotype with a major recurring mutation (p.Arg853Gln: two new and four previously reported children). Other known phenotypes include Ohtahara syndrome, epilepsy of infancy with migrating focal seizures, and intellectual disability or autism without epilepsy. To assess the response to antiepileptic therapy, we retrospectively reviewed the treatment regimen and the course of the epilepsy in 66 patients for which well-documented medical information was available. We find that the use of sodium channel blockers was often associated with clinically relevant seizure reduction or seizure freedom in children with early infantile epilepsies (<3 months), whereas other antiepileptic drugs were less effective. In contrast, sodium channel blockers were rarely effective in epilepsies with later onset (≥3 months) and sometimes induced seizure worsening. Regarding the genetic findings, truncating mutations were exclusively seen in patients with late onset epilepsies and lack of response to sodium channel blockers. Functional characterization of four selected missense mutations using whole cell patch-clamping in tsA201 cells-together with data from the literature-suggest that mutations associated with early infantile epilepsy result in increased sodium channel activity with gain-of-function, characterized by slowing of fast inactivation, acceleration of its recovery or increased persistent sodium current. Further, a good response to sodium channel blockers clinically was found to be associated with a relatively small gain-of-function. In contrast, mutations in patients with late-onset forms and an insufficient response to sodium channel blockers were associated with loss-of-function effects, including a depolarizing shift of voltage-dependent activation or a hyperpolarizing shift of channel availability (steady-state inactivation). Our clinical and experimental data suggest a correlation between age at disease onset, response to sodium channel blockers and the functional properties of mutations in children with SCN2A-related epilepsy.


Assuntos
Epilepsia/tratamento farmacológico , Epilepsia/genética , Epilepsia/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Transtornos do Neurodesenvolvimento/genética , Bloqueadores dos Canais de Sódio/uso terapêutico , Adolescente , Adulto , Idade de Início , Criança , Pré-Escolar , Dinamarca/epidemiologia , Epilepsia/epidemiologia , Feminino , Humanos , Lactente , Masculino , Mutação , Fenótipo , Adulto Jovem
9.
Biochim Biophys Acta ; 1850(4): 832-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25615535

RESUMO

BACKGROUND: Phosphorylation plays an essential role in regulating voltage-gated sodium (Na(v)) channels and excitability. Yet, a surprisingly limited number of kinases have been identified as regulators of Na(v) channels. We posited that glycogen synthase kinase 3 (GSK3), a critical kinase found associated with numerous brain disorders, might directly regulate neuronal Na(v) channels. METHODS: We used patch-clamp electrophysiology to record sodium currents from Na(v)1.2 channels stably expressed in HEK-293 cells. mRNA and protein levels were quantified with RT-PCR, Western blot, or confocal microscopy, and in vitro phosphorylation and mass spectrometry to identify phosphorylated residues. RESULTS: We found that exposure of cells to GSK3 inhibitor XIII significantly potentiates the peak current density of Na(v)1.2, a phenotype reproduced by silencing GSK3 with siRNA. Contrarily, overexpression of GSK3ß suppressed Na(v)1.2-encoded currents. Neither mRNA nor total protein expression was changed upon GSK3 inhibition. Cell surface labeling of CD4-chimeric constructs expressing intracellular domains of the Na(v)1.2 channel indicates that cell surface expression of CD4-Na(v)1.2 C-tail was up-regulated upon pharmacological inhibition of GSK3, resulting in an increase of surface puncta at the plasma membrane. Finally, using in vitro phosphorylation in combination with high resolution mass spectrometry, we further demonstrate that GSK3ß phosphorylates T(1966) at the C-terminal tail of Na(v)1.2. CONCLUSION: These findings provide evidence for a new mechanism by which GSK3 modulates Na(v) channel function via its C-terminal tail. GENERAL SIGNIFICANCE: These findings provide fundamental knowledge in understanding signaling dysfunction common in several neuropsychiatric disorders.


Assuntos
Quinase 3 da Glicogênio Sintase/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Sequência de Aminoácidos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Células HEK293 , Humanos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.2/química , Fosforilação
10.
PLoS One ; 9(11): e113272, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25420080

RESUMO

Cinobufagin and resibufogenin are two major effective bufadienolides of Chan su (toad venom), which is a Chinese medicine obtained from the skin venom gland of toads and is used as a cardiotonic and central nervous system (CNS) respiratory agent, an analgesic and anesthetic, and as a remedy for ulcers. Many clinical cases showed that Chan su has severe side-effects on the CNS, causing shortness of breath, breathlessness, seizure, coma and cardiac arrhythmia. We used whole-cell recordings from brain slices to determine the effects of bufadienolides on excitability of a principal neuron in main olfactory bulb (MOB), mitral cells (MCs), and the cellular mechanism underlying the excitation. At higher concentrations, cinobufagin and resibufogenin induced irreversible over-excitation of MCs indicating a toxic effect. At lower concentrations, they concentration-dependently increased spontaneous firing rate, depolarized the membrane potential of MCs, and elicited inward currents. The excitatory effects were due to a direct action on MCs rather than an indirect phasic action. Bufadienolides and ouabain had similar effects on firing of MCs which suggested that bufadienolides activated neuron through a ouabain-like effect, most likely by inhibiting Na+/K+-ATPase. The direct action of bufadienolide on brain Na+ channels was tested by recordings from stably Nav1.2-transfected cells. Bufadienolides failed to make significant changes of the main properties of Nav1.2 channels in current amplitude, current-voltage (I-V) relationships, activation and inactivation. Our results suggest that inhibition of Na+/K+-ATPase may be involved in both the pharmacological and toxic effects of bufadienolide-evoked CNS excitation.


Assuntos
Bufanolídeos/farmacologia , Neurônios/fisiologia , Venenos de Anfíbios/química , Animais , Bufanolídeos/química , Células CHO , Cardiotônicos/farmacologia , Sistema Nervoso Central/citologia , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Neurônios/metabolismo , Bulbo Olfatório/citologia , Ouabaína/farmacologia , Técnicas de Patch-Clamp , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/metabolismo
11.
Naunyn Schmiedebergs Arch Pharmacol ; 387(10): 991-1000, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25012093

RESUMO

Amylmetacresol and dichloro-benzylalcohol are ingredients of lozenges used for the treatment of sore throat. In a former in vitro study, a local anaesthetic-like effect of these substances has been described. Since amylmetacresol and dichloro-benzylalcohol are co-administered in over-the-counter lozenges, the intention of this study is to evaluate the in vitro effects of the combination of these compounds on the voltage-gated sodium channel. We analysed the block of inward sodium currents induced by the combination of amylmetacresol, dichloro-benzylalcohol and the local anaesthetic lidocaine. Tonic and use-dependent block and effects on the inactivated channel state of the neuronal sodium channel were examined. Therefore, the α-subunit of the voltage-gated NaV1.2 sodium channel was heterologously expressed in HEK 293 cells in vitro. Inward sodium currents were investigated in the whole-cell configuration of the patch-clamp technique. The combination of amylmetacresol and dichloro-benzylalcohol and the combination of amylmetacresol and lidocaine induced a block of resting and inactivated sodium channels both displaying a pronounced block at the inactivated channel state. In addition, the combination of all three compounds also resulted in a voltage-dependent block of inward sodium currents. While use-dependent block by co-application of amylmetacresol and dichloro-benzylalcohol was moderate (<20 %), lidocaine and amylmetacresol induced a robust use-dependent block (up to 50 %). This study demonstrates local anaesthetic-like effects of a combination of amylmetacresol and dichloro-benzylalcohol as established ingredients of lozenges. In the presence of amylmetacresol, dichloro-benzylalcohol and lidocaine, a prominent block of inward sodium currents is apparent.


Assuntos
Anestésicos Locais/administração & dosagem , Anti-Infecciosos Locais/administração & dosagem , Lidocaína/administração & dosagem , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Faringite , Bloqueadores dos Canais de Sódio/administração & dosagem , Administração Tópica , Álcoois Benzílicos , Cresóis/administração & dosagem , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Células HEK293 , Humanos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Faringite/tratamento farmacológico
12.
Mol Pharmacol ; 85(1): 162-74, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24202911

RESUMO

Ranolazine is an approved drug for chronic stable angina that acts by suppressing a noninactivating current conducted by the cardiac sodium channel [persistent sodium ion current (INa)]. Ranolazine has also been shown to inhibit the increased persistent INa carried by NaV1.1 channels encoding epilepsy- and migraine-associated mutations. Here, we investigate the antiepileptic properties of ranolazine exhibited through the reduction of hippocampal neuronal excitability. At therapeutically relevant concentrations, ranolazine reduced action potential firing frequency of hippocampal neurons in response to repetitive depolarizing current injections. Similarly, using a single current injection paradigm, ranolazine required a long depolarization (4 seconds) to produce significant inhibition of excitability, which was similar to that observed for the anticonvulsants phenytoin (slowly binds to the fast-inactivated state) and lacosamide (binds to the slow-inactivated state). Ranolazine enhanced the development of fast and slow inactivation assessed with conditioning prepulses of 100, 1000, or 10,000 milliseconds. Recovery of channels from inactivated states was also slowed in the presence of ranolazine. Interestingly, the use-dependent inhibition of hippocampal neurons was dependent on the duration of the voltage step, suggesting ranolazine does not selectively affect the open state and may also interact with inactivated states. NEURON (Yale University, New Haven, CT) computational simulations predict equal inhibition of action potential generation for binding to either fast-inactivated or slow-inactivated states. Binding of ranolazine to either preopen or open states did not affect the excitability of the simulation. Ranolazine was able to significantly reduce the epileptiform activity of the neuronal cultures, suggesting possible antiepileptic activity.


Assuntos
Acetanilidas/farmacologia , Anticonvulsivantes/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Piperazinas/farmacologia , Canais de Sódio Disparados por Voltagem/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Células Cultivadas , Simulação por Computador , Epilepsia/fisiopatologia , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Cadeias de Markov , N-Metilaspartato/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.1/química , Canal de Sódio Disparado por Voltagem NAV1.1/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.2/química , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Ligação Proteica , Conformação Proteica , Ranolazina , Ratos , Canais de Sódio Disparados por Voltagem/química
13.
J Pharmacol Sci ; 120(1): 54-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22878600

RESUMO

Neurosteroids are known as allosteric modulators of the ligand-gated ion channel superfamily. Voltage-gated sodium channels (Na(v)) play an important role in mediating excitotoxic damages. Here we report the effects of neurosteroids on the function of Na(v), using voltage-clamp techniques in Xenopus oocytes expressed with the Na(v)1.2 α subunit. Pregnenolone sulphate, but not pregnenolone, inhibited sodium currents (I(Na)) at 3 - 100 µmol/L. The suppression of I(Na) by pregnenolone sulphate was due to increased inactivation with little change in activation. These findings suggest that pregnenolone sulphate, a metabolite of pregnenolone, suppresses the function of Na(v) via increased inactivation, which may contribute to the neuroprotection.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Pregnenolona/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Células Cultivadas , Desidroepiandrosterona/farmacologia , Sulfato de Desidroepiandrosterona/farmacologia , Feminino , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Pregnenolona/metabolismo , Xenopus laevis
14.
Br J Pharmacol ; 166(7): 2148-60, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22452751

RESUMO

BACKGROUND AND PURPOSE: Voltage-gated sodium channels are expressed primarily in excitable cells and play a pivotal role in the initiation and propagation of action potentials. Nine subtypes of the pore-forming α-subunit have been identified, each with a distinct tissue distribution, biophysical properties and sensitivity to tetrodotoxin (TTX). Na(v) 1.8, a TTX-resistant (TTX-R) subtype, is selectively expressed in sensory neurons and plays a pathophysiological role in neuropathic pain. In comparison with TTX-sensitive (TTX-S) Na(v) α-subtypes in neurons, Na(v) 1.8 is most strongly inhibited by the µO-conotoxin MrVIB from Conus marmoreus. To determine which domain confers Na(v) 1.8 α-subunit its biophysical properties and MrVIB binding, we constructed various chimeric channels incorporating sequence from Na(v) 1.8 and the TTX-S Na(v) 1.2 using a domain exchange strategy. EXPERIMENTAL APPROACH: Wild-type and chimeric Na(v) channels were expressed in Xenopus oocytes, and depolarization-activated Na⁺ currents were recorded using the two-electrode voltage clamp technique. KEY RESULTS: MrVIB (1 µM) reduced Na(v) 1.2 current amplitude to 69 ± 12%, whereas Na(v) 1.8 current was reduced to 31 ± 3%, confirming that MrVIB has a binding preference for Na(v) 1.8. A similar reduction in Na⁺ current amplitude was observed when MrVIB was applied to chimeras containing the region extending from S6 segment of domain I through the S5-S6 linker of domain II of Na(v) 1.8. In contrast, MrVIB had only a small effect on Na⁺ current for chimeras containing the corresponding region of Na(v) 1.2. CONCLUSIONS AND IMPLICATIONS: Taken together, these results suggest that domain II of Na(v) 1.8 is an important determinant of MrVIB affinity, highlighting a region of the α-subunit that may allow further nociceptor-specific ligand targeting.


Assuntos
Conotoxinas/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.8/fisiologia , Animais , Células Cultivadas , Feminino , Oócitos , Subunidades Proteicas , Tetrodotoxina/farmacologia , Xenopus laevis
15.
Sheng Li Xue Bao ; 63(1): 1-8, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21340428

RESUMO

A new method of axon recording through axon bleb has boosted the studies on the functional role of central nervous system (CNS) axons. Using this method, we have revealed the mechanisms underlying the initiation and propagation of the digital-mode signal, all-or-none action potentials (APs), in neocortical pyramidal neurons. Accumulation of the low-threshold Na(+) channel subtype Na(v)1.6 at the distal end of the axon initial segment (AIS) determines the lowest threshold for AP initiation, whereas accumulation of the high-threshold subtype Na(v)1.2 at the proximal region of the AIS promotes AP backpropagation to the soma and dendrites. Through dual recording from the soma and the axon, we have showed that subthreshold membrane potential (V(m)) fluctuations in the soma propagate along the axon to a long distance and probably reach the axon terminals. Paired recording from cortical neurons has revealed that these V(m) changes in the soma modulate AP-triggered synaptic transmission. This new V(m)-dependent mode of synaptic transmission is called analog communication. Unique properties of axonal K(+) channels (K(v)1 channels) may contribute to shaping the AP waveform, particularly its duration, and thus controlling synaptic strength at different levels of presynaptic V(m). The level of background Ca(2+) may also participate in mediating the analog signaling. Together, these findings enrich our knowledge on the principles of neuronal signaling in the CNS and help understand how the brain works.


Assuntos
Potenciais de Ação/fisiologia , Axônios/fisiologia , Sistema Nervoso Central/fisiologia , Células Piramidais/fisiologia , Animais , Sistema Nervoso Central/citologia , Humanos , Potenciais da Membrana/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.2/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Neocórtex/citologia , Neocórtex/fisiologia , Técnicas de Patch-Clamp , Canais de Sódio/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA