Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Neurobiol Dis ; 167: 105672, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35219855

RESUMO

Thalamocortical network dysfunction contributes to seizures and sleep deficits in Dravet syndrome (DS), an infantile epileptic encephalopathy, but the underlying molecular and cellular mechanisms remain elusive. DS is primarily caused by mutations in the SCN1A gene encoding the voltage-gated sodium channel NaV1.1, which is highly expressed in GABAergic reticular thalamus (nRT) neurons as well as glutamatergic thalamocortical neurons. We hypothesized that NaV1.1 haploinsufficiency alters somatosensory corticothalamic circuit function through both intrinsic and synaptic mechanisms in nRT and thalamocortical neurons. Using Scn1a heterozygous mice of both sexes aged P25-P30, we discovered reduced excitability of nRT neurons and thalamocortical neurons in the ventral posterolateral (VPL) thalamus, while thalamocortical ventral posteromedial (VPM) neurons exhibited enhanced excitability. NaV1.1 haploinsufficiency enhanced GABAergic synaptic input and reduced glutamatergic input to VPL neurons, but not VPM neurons. In addition, glutamatergic input to nRT neurons was reduced in Scn1a heterozygous mice. These findings introduce alterations in glutamatergic synapse function and aberrant glutamatergic neuron excitability in the thalamus as disease mechanisms in DS, which has been widely considered a disease of GABAergic neurons. This work reveals additional complexity that expands current models of thalamic dysfunction in DS and identifies new components of corticothalamic circuitry as potential therapeutic targets.


Assuntos
Epilepsias Mioclônicas , Neurônios GABAérgicos , Animais , Modelos Animais de Doenças , Epilepsias Mioclônicas/genética , Feminino , Neurônios GABAérgicos/fisiologia , Haploinsuficiência , Masculino , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Tálamo
2.
Bioorg Med Chem Lett ; 75: 128946, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35985458

RESUMO

The voltage-gated sodium (Nav) channel is one of most important targets for treatment of epilepsy, and rufinamide is an approved third-generation anti-seizure drug as Nav1.1 channel blocker. Herein, by triazenylation of rufinamide, we reported the triazenyl triazoles as new Nav1.1 channel blocker for treatment of epilepsy. Through the electrophysiological activity assay, compound 6a and 6e were found to modulate the inactivation voltage of Nav 1.1 channel with shift of -10.07 mv and -11.28 mV, respectively. In the pentylenetetrazole (PTZ) mouse model, 6a and 6e reduced the seizure level, prolonged seizure latency and improved the survival rate of epileptic mice at an intragastric administration of 50 mg/kg dosage. In addition, 6a also exhibited promising effectiveness in the maximal electroshock (MES) mouse model and possessed moderate pharmacokinetic profiles. These results demonstrated that 6a was a novel Nav1.1 channel blocker for treatment of epilepsy.


Assuntos
Epilepsia , Pentilenotetrazol , Animais , Modelos Animais de Doenças , Epilepsia/tratamento farmacológico , Camundongos , Sódio , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores dos Canais de Sódio/uso terapêutico , Triazóis/farmacologia , Triazóis/uso terapêutico , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/uso terapêutico
3.
Epilepsy Behav ; 131(Pt B): 108043, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34053869

RESUMO

Dravet syndrome is a well-established electro-clinical condition first described in 1978. A main genetic cause was identified with the discovery of a loss-of-function SCN1A variant in 2001. Mechanisms underlying the phenotypic variations have subsequently been a main topic of research. Various genetic modifiers of clinical severities have been elucidated through many rigorous studies on genotype-phenotype correlations and the recent advances in next generation sequencing technology. Furthermore, a deeper understanding of the regulation of gene expression and remarkable progress on genome-editing technology using the CRISPR-Cas9 system provide significant opportunities to overcome hurdles of gene therapy, such as enhancing NaV1.1 expression. This article reviews the current understanding of genetic pathology and the status of research toward the development of gene therapy for Dravet syndrome. This article is part of the Special Issue "Severe Infantile Epilepsies".


Assuntos
Epilepsias Mioclônicas , Epilepsia , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/terapia , Epilepsia/genética , Síndromes Epilépticas , Terapia Genética , Humanos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Espasmos Infantis
4.
Neurobiol Dis ; 132: 104583, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31445158

RESUMO

Dravet syndrome (DS) is an early onset refractory epilepsy typically caused by de novo heterozygous variants in SCN1A encoding the α-subunit of the neuronal sodium channel Nav1.1. The syndrome is characterized by age-related progression of seizures, cognitive decline and movement disorders. We hypothesized that the distinct neurodevelopmental features in DS are caused by the disruption of molecular pathways in Nav1.1 haploinsufficient cells resulting in perturbed neural differentiation and maturation. Here, we established DS-patient and control induced pluripotent stem cell derived neural progenitor cells (iPSC NPC) and GABAergic inter-neuronal (iPSC GABA) cells. The DS-patient iPSC GABA cells showed a shift in sodium current activation and a perturbed response to induced oxidative stress. Transcriptome analysis revealed specific dysregulations of genes for chromatin structure, mitotic progression, neural plasticity and excitability in DS-patient iPSC NPCs and DS-patient iPSC GABA cells versus controls. The transcription factors FOXM1 and E2F1, positive regulators of the disrupted pathways for histone modification and cell cycle regulation, were markedly up-regulated in DS-iPSC GABA lines. Our study highlights transcriptional changes and disrupted pathways of chromatin remodeling in Nav1.1 haploinsufficient GABAergic cells, providing a molecular framework that overlaps with that of neurodevelopmental disorders and other epilepsies.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Epilepsias Mioclônicas/genética , Neurônios GABAérgicos/metabolismo , Neurogênese/genética , Células Cultivadas , Epilepsias Mioclônicas/metabolismo , Haploinsuficiência , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Células-Tronco Neurais/metabolismo , Plasticidade Neuronal/fisiologia , Estresse Oxidativo/fisiologia , Transcriptoma
5.
Bioorg Med Chem Lett ; 29(6): 815-820, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30704812

RESUMO

The voltage-gated sodium channel, Nav1.1, is predominantly expressed in parvalbumin-positive fast spiking interneurons and has been genetically linked to Dravet syndrome. Starting from a high throughput screening hit isoxazole derivative 5, modifications of 5 via combinations of IonWorks and Q-patch assays successfully identified the nicotinamide derivative 4. Its increasing decay time constant (tau) of Nav1.1 currents at 0.03 µM along with significant selectivity against Nav1.2, Nav1.5, and Nav1.6 and acceptable brain exposure in mice was observed. Compound 4 is a promising Nav1.1 activator that can be used to analyze pathophysiological functions of the Nav1.1 channel towards treating various central nervous system diseases.


Assuntos
Descoberta de Drogas , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Pirrolidinas/farmacologia , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Células CHO , Cricetulus , Camundongos , Estrutura Molecular , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Niacinamida/síntese química , Pirrolidinas/síntese química , Pirrolidinas/química , Relação Estrutura-Atividade , Agonistas do Canal de Sódio Disparado por Voltagem/síntese química , Agonistas do Canal de Sódio Disparado por Voltagem/química
6.
Neurobiol Dis ; 73: 106-17, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25281316

RESUMO

Dominant loss-of-function mutations in voltage-gated sodium channel NaV1.1 cause Dravet Syndrome, an intractable childhood-onset epilepsy. NaV1.1(+/-) Dravet Syndrome mice in C57BL/6 genetic background exhibit severe seizures, cognitive and social impairments, and premature death. Here we show that Dravet Syndrome mice in pure 129/SvJ genetic background have many fewer seizures and much less premature death than in pure C57BL/6 background. These mice also have a higher threshold for thermally induced seizures, fewer myoclonic seizures, and no cognitive impairment, similar to patients with Genetic Epilepsy with Febrile Seizures Plus. Consistent with this mild phenotype, mutation of NaV1.1 channels has much less physiological effect on neuronal excitability in 129/SvJ mice. In hippocampal slices, the excitability of CA1 Stratum Oriens interneurons is selectively impaired, while the excitability of CA1 pyramidal cells is unaffected. NaV1.1 haploinsufficiency results in increased rheobase and threshold for action potential firing and impaired ability to sustain high-frequency firing. Moreover, deletion of NaV1.1 markedly reduces the amplification and integration of synaptic events, further contributing to reduced excitability of interneurons. Excitability is less impaired in inhibitory neurons of Dravet Syndrome mice in 129/SvJ genetic background. Because specific deletion of NaV1.1 in forebrain GABAergic interneuons is sufficient to cause the symptoms of Dravet Syndrome in mice, our results support the conclusion that the milder phenotype in 129/SvJ mice is caused by lesser impairment of sodium channel function and electrical excitability in their forebrain interneurons. This mild impairment of excitability of interneurons leads to a milder disease phenotype in 129/SvJ mice, similar to Genetic Epilepsy with Febrile Seizures Plus in humans.


Assuntos
Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Inibição Neural/genética , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Fenômenos Biofísicos/genética , Condicionamento Psicológico/fisiologia , Modelos Animais de Doenças , Epilepsias Mioclônicas/etiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Medo/psicologia , Hipocampo/citologia , Hipertermia Induzida/efeitos adversos , Técnicas In Vitro , Lidocaína/análogos & derivados , Lidocaína/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia
7.
Neurosci Lett ; 724: 134853, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32114117

RESUMO

Voltage-gated sodium channels (VGSCs) are responsible for the initiation and propagation of action potentials in neurons. The human genome includes ten human VGSC α-subunit genes, SCN(X)A, encoding Nav1.1-1.9 plus Nax. To understand the unique role that each VGSC plays in normal and pathophysiological function in neural networks, compounds with high affinity and selectivity for specific VGSC subtypes are required. Toward that goal, a structural analog of the VGSC pore blocker tetrodotoxin, 4,9-anhydrotetrodotoxin (4,9-ah-TTX), has been reported to be more selective in blocking Na+ current mediated by Nav1.6 than other TTX-sensitive VGSCs, including Nav1.2, Nav1.3, Nav1.4, and Nav1.7. While SCN1A, encoding Nav1.1, has been implicated in several neurological diseases, the effects of 4,9-ah-TTX on Nav1.1-mediated Na+ current have not been tested. Here, we compared the binding of 4,9-ah-TTX for human and mouse brain preparations, and the effects of 4,9-ah-TTX on human Nav1.1-, Nav1.3- and Nav1.6-mediated Na+ currents using the whole-cell patch clamp technique in heterologous cells. We show that, while 4,9-ah-TTX administration results in significant blockade of Nav1.6-mediated Na+ current in the nanomolar range, it also has significant effects on Nav1.1-mediated Na+ current. Thus, 4,9-ah-TTX is not a useful tool in identifying Nav1.6-specific effects in human brain networks.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.1/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Tetrodotoxina/análogos & derivados , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Especificidade da Espécie , Tetrodotoxina/farmacologia
8.
Biochem Pharmacol ; 181: 113991, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32335140

RESUMO

Dravet syndrome (DS) is a catastrophic epileptic encephalopathy characterised by childhood-onset polymorphic seizures, multiple neuropsychiatric comorbidities, and increased risk of sudden death. Heterozygous loss-of-function mutations in one allele of SCN1A, the gene encoding the voltage-gated sodium channel 1.1 (NaV1.1), lead to DS. NaV1.1 is primarily found in the axon initial segment of fast-spiking GABAergic inhibitory interneurons in the brain, and the principle mechanism proposed to underlie seizure genesis in DS is loss of inhibitory input due to dysfunctional firing of GABAergic interneurons. We hypothesised that DS symptoms could be ameliorated by a drug that activates the reduced population of functional NaV1.1 channels in DS interneurons. We recently identified two homologous disulfide-rich spider-venom peptides (Hm1a and Hm1b) that selectively potentiate NaV1.1, and showed that selective activation of NaV1.1 by Hm1a restores the function of inhibitory interneurons in a mouse model of DS. Here we produced recombinant Hm1b (rHm1b) using an E. coli periplasmic expression system, and examined its selectivity against a panel of human NaV subtypes using whole-cell patch-clamp recordings. rHm1b is a potent and highly selective agonist of NaV1.1 and NaV1.3 (EC50 ~12 nM for both). rHm1b is a gating modifier that shifts the voltage dependence of channel activation and inactivation to hyperpolarised and depolarised potentials respectively, presumably by interacting with the channel's voltage-sensor domains. Like Hm1a, the structure of rHm1b determined by using NMR revealed a classical inhibitor cystine knot (ICK) motif. However, we show that rHm1b is an order of magnitude more stable than Hm1a in human cerebrospinal fluid. Overall, our data suggest that rHm1b is an exciting lead for a precision therapeutic targeted against DS.


Assuntos
Epilepsias Mioclônicas/tratamento farmacológico , Interneurônios/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Peptídeos/farmacologia , Agonistas de Canais de Sódio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Epilepsias Mioclônicas/metabolismo , Células HEK293 , Humanos , Interneurônios/metabolismo , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Técnicas de Patch-Clamp , Peptídeos/química , Peptídeos/genética , Homologia de Sequência de Aminoácidos , Agonistas de Canais de Sódio/química , Venenos de Aranha/metabolismo
9.
J Neurosci Methods ; 325: 108315, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31265868

RESUMO

BACKGROUND: Dravet Syndrome (DS) is an epileptic disorder characterized by spontaneous and thermally-induced seizures, hyperactivity, cognitive deficits, autistic-like behaviors, and Sudden Unexpected Death in Epilepsy (SUDEP). DS is caused by de novo loss-of-function mutations in the SCN1A gene. Selective loss of GABAergic interneuron excitability is the primary cause of the disease. Up to 60% of Scn1a+/- mice die from SUDEP before sexual maturity. NEW METHOD: We used Cre-Lox technology to conditionally delete Scn1a in all epiblast-derived somatic cells by crossing a floxed Scn1a mouse with a mouse expressing Cre under the Meox2 promoter. RESULTS: Parental Scn1a flox (F) mice, parental Meox2 Cre+ mice, and their F/+:Meox2-Cre- offspring were phenotypically normal and did not prematurely die. In contrast, F/+:Meox2-Cre+ offspring recapitulated DS seizure and behavioral phenotypes. Unexpectedly, male F/+:Meox2-Cre+ mice demonstrated impaired social interaction, while females did not. COMPARISON WITH EXISTING METHOD: In the previous models, colony maintenance required breeding SUDEP survivors, which greatly increased colony size required to sustain experimental animal production, and raised the concern that surviving breeders have epigenetic traits that impart new phenotypes to their offspring. Our method greatly facilitates breeding, recapitulates DS phenotypes, eliminates concerns about parents that are survivors, and provides initial evidence for unexpected sex-dependent social interaction impairment. CONCLUSIONS: We introduce a more efficient mouse model of human DS that demonstrates an efficient breeding strategy free from potential inherited epigenetic changes and reveals an unexpected sex-specific impairment of social interaction in DS. This new model should have great value to investigators of DS.


Assuntos
Comportamento Animal/fisiologia , Epigênese Genética/fisiologia , Epilepsias Mioclônicas/fisiopatologia , Relações Interpessoais , Animais , Modelos Animais de Doenças , Eletroencefalografia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1 , Caracteres Sexuais
10.
Neuron ; 99(5): 905-913.e7, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30146301

RESUMO

Channelopathies are disorders caused by abnormal ion channel function in differentiated excitable tissues. We discovered a unique neurodevelopmental channelopathy resulting from pathogenic variants in SCN3A, a gene encoding the voltage-gated sodium channel NaV1.3. Pathogenic NaV1.3 channels showed altered biophysical properties including increased persistent current. Remarkably, affected individuals showed disrupted folding (polymicrogyria) of the perisylvian cortex of the brain but did not typically exhibit epilepsy; they presented with prominent speech and oral motor dysfunction, implicating SCN3A in prenatal development of human cortical language areas. The development of this disorder parallels SCN3A expression, which we observed to be highest early in fetal cortical development in progenitor cells of the outer subventricular zone and cortical plate neurons and decreased postnatally, when SCN1A (NaV1.1) expression increased. Disrupted cerebral cortical folding and neuronal migration were recapitulated in ferrets expressing the mutant channel, underscoring the unexpected role of SCN3A in progenitor cells and migrating neurons.


Assuntos
Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/crescimento & desenvolvimento , Desenvolvimento da Linguagem , Canal de Sódio Disparado por Voltagem NAV1.3/genética , Canais de Sódio/genética , Adolescente , Adulto , Animais , Movimento Celular/fisiologia , Células Cultivadas , Córtex Cerebral/patologia , Criança , Pré-Escolar , Feminino , Furões , Células HEK293 , Humanos , Lactente , Masculino , Megalencefalia/diagnóstico por imagem , Megalencefalia/genética , Megalencefalia/patologia , Pessoa de Meia-Idade , Linhagem , Polimicrogiria/diagnóstico por imagem , Polimicrogiria/genética , Polimicrogiria/patologia
11.
Exp Neurol ; 293: 159-171, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28373025

RESUMO

Mutations in the voltage-gated sodium channel (VGSC) gene SCN1A, encoding the Nav1.1 channel, are responsible for a number of epilepsy disorders including genetic epilepsy with febrile seizures plus (GEFS+) and Dravet syndrome (DS). Patients with SCN1A mutations often experience prolonged early-life febrile seizures (FSs), raising the possibility that these events may influence epileptogenesis and lead to more severe adult phenotypes. To test this hypothesis, we subjected 21-23-day-old mice expressing the human SCN1A GEFS+ mutation R1648H to prolonged hyperthermia, and then examined seizure and behavioral phenotypes during adulthood. We found that early-life FSs resulted in lower latencies to induced seizures, increased severity of spontaneous seizures, hyperactivity, and impairments in social behavior and recognition memory during adulthood. Biophysical analysis of brain slice preparations revealed an increase in epileptiform activity in CA3 pyramidal neurons along with increased action potential firing, providing a mechanistic basis for the observed worsening of adult phenotypes. These findings demonstrate the long-term negative impact of early-life FSs on disease outcomes. This has important implications for the clinical management of this patient population and highlights the need for therapeutic interventions that could ameliorate disease progression.


Assuntos
Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Convulsões Febris/complicações , Convulsões Febris/genética , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Arginina/genética , Convulsivantes/toxicidade , Modelos Animais de Doenças , Progressão da Doença , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , Flurotila/toxicidade , Hipocampo/patologia , Histidina/genética , Humanos , Hipertermia Induzida/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Convulsões Febris/etiologia , Convulsões Febris/patologia
12.
Seizure ; 44: 58-64, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27817982

RESUMO

Dravet syndrome combines clonic generalized, focal or unilateral seizures, beginning within the first year of life, often triggered by hyperthermia whatever its cause, including pertussis vaccination. Long-lasting febrile seizures are frequent in infancy and repeat status epilepticus (SE) has negative prognostic value. Massive myoclonus, rare absences, complex partial seizures and generalized spikes may appear several years later. Myoclonic status may occur in childhood, but acute encephalopathy with febrile SE followed by ischemic lesions and psychomotor impairment, the most severe condition, occurs mainly within the first five years of life. Generalized tonic-clonic and tonic seizures in sleep predominate in adulthood. Non epileptic manifestations appear with age, including intellectual disability, ataxia and crouching gait. Incidence of SUDEP is high, whatever the age. SCN1A haploinsufficiency producing NaV1.1 dysfunction mainly affects GABAergic neurons. In cortical interneurons it explains epilepsy, in cerebellum the ataxia, in basal ganglia and motor neurons the crouching gait, in hypothalamus the thermodysregulation and sleep troubles, and dysfunction in all these structures contributes to psychomotor delay. Valproate, stiripentol, topiramate and bromide are the basis of antiepileptic treatment, whereas inhibitors of sodium channel worsen the condition. Benzodiazepines seem to facilitate acute encephalopathy when given chronically, and they should be restricted to SE. Ketogenic diet is useful in both chronic and acute conditions. Only targeting SCN1A haploinsufficiency and NaV1.1 dysfunction could improve non epileptic manifestations of this condition that deserves being considered as a disease, not only as an epilepsy syndrome.


Assuntos
Epilepsias Mioclônicas , Genótipo , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Fenótipo , Anticonvulsivantes/uso terapêutico , Transtornos Cognitivos/etiologia , Morte Súbita , Diagnóstico Diferencial , Epilepsias Mioclônicas/complicações , Epilepsias Mioclônicas/tratamento farmacológico , Epilepsias Mioclônicas/genética , Humanos , Transtornos dos Movimentos/etiologia
13.
Exp Neurol ; 275 Pt 1: 46-58, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26410685

RESUMO

Understanding the role of SCN8A in epilepsy and behavior is critical in light of recently identified human SCN8A epilepsy mutations. We have previously demonstrated that Scn8a(med) and Scn8a(med-jo) mice carrying mutations in the Scn8a gene display increased resistance to flurothyl and kainic acid-induced seizures; however, they also exhibit spontaneous absence seizures. To further investigate the relationship between altered SCN8A function and epilepsy, we introduced the SCN1A-R1648H mutation, identified in a family with generalized epilepsy with febrile seizures plus (GEFS+), into the corresponding position (R1627H) of the mouse Scn8a gene. Heterozygous R1627H mice exhibited increased resistance to some forms of pharmacologically and electrically induced seizures and the mutant Scn8a allele ameliorated the phenotype of Scn1a-R1648H mutants. Hippocampal slices from heterozygous R1627H mice displayed decreased bursting behavior compared to wild-type littermates. Paradoxically, at the homozygous level, R1627H mice did not display increased seizure resistance and were susceptible to audiogenic seizures. We furthermore observed increased hippocampal pyramidal cell excitability in heterozygous and homozygous Scn8a-R1627H mutants, and decreased interneuron excitability in heterozygous Scn8a-R1627H mutants. These results expand the phenotypes associated with disruption of the Scn8a gene and demonstrate that an Scn8a mutation can both confer seizure protection and increase seizure susceptibility.


Assuntos
Hipocampo/fisiopatologia , Interneurônios/metabolismo , Mutação , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Células Piramidais/metabolismo , Convulsões/genética , Estimulação Acústica , Animais , Tronco Encefálico/metabolismo , Tronco Encefálico/fisiopatologia , Suscetibilidade a Doenças , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Convulsões/metabolismo , Convulsões/fisiopatologia
14.
Artigo em Chinês | WPRIM | ID: wpr-383687

RESUMO

Objective To observe the effect of electro-acupuncture therapy (ET) on the expression of sodium channel Na(v) 1.1 in rats after acute cerebral ischemia and the mechanism of any protective function of ET.Methods A model of focal acute cerebral ischemia was established by occluding the right middle cerebral artery.One hundred and eighty healthy SD rats were randomly divided into a sham operation control (SC) group, an ischemia control (IC) group, a real ET group and a false ET group, with 45 in each group. Immunohistochemistry and real-time polymerase chain reaction (PGR) methods were used to detect Na(v)1. 1 expression. 2,3,5-triphenyl tetrazolium chloride (TTC) staining was used to detect infarct volume. Neurological examination and grading was carried out at 6 hours and then 1, 2, 3 and 7 days after inducing ischemia. Results The gradings and infarction volume ratios of the rats in the IC group were the most serious, while in the real ET group the severity was much less at each time point. Compared with the SC group, the expression of Na(v) 1.1 was significantly up-regulated in the IC group. The expression of Na(v) 1.1 was increased at the 6th hour, then down-regulated to the lowest level at day 1,then from the 2nd to the 7th day was up-regulated again. The expression of Na(v) 1.1 in the real ET group was significantly lower than in the IC group. Although the expression of Na(v)1.1 in the false ET group was low compared with the IC group, the difference was not significant. The difference between the real ET group and the false ET group was significant, however. Conclusions ET can reduce damage from cerebral ischemia and benefit the recovery of neural function. ET can also could regulate the expression of Na(v)1.1 after acute cerebral ischemia, which may be an important mechanism for neural function recovery.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa