Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
J Neurosci ; 44(17)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38443186

RESUMO

Dravet syndrome (DS) is a neurodevelopmental disorder characterized by epilepsy, developmental delay/intellectual disability, and features of autism spectrum disorder, caused by heterozygous loss-of-function variants in SCN1A encoding the voltage-gated sodium channel α subunit Nav1.1. The dominant model of DS pathogenesis is the "interneuron hypothesis," whereby GABAergic interneurons (INs) express and preferentially rely on Nav1.1-containing sodium channels for action potential (AP) generation. This has been shown for three of the major subclasses of cerebral cortex GABAergic INs: those expressing parvalbumin (PV), somatostatin, and vasoactive intestinal peptide. Here, we define the function of a fourth major subclass of INs expressing neuron-derived neurotrophic factor (Ndnf) in male and female DS (Scn1a+/-) mice. Patch-clamp electrophysiological recordings of Ndnf-INs in brain slices from Scn1a+/â mice and WT controls reveal normal intrinsic membrane properties, properties of AP generation and repetitive firing, and synaptic transmission across development. Immunohistochemistry shows that Nav1.1 is strongly expressed at the axon initial segment (AIS) of PV-expressing INs but is absent at the Ndnf-IN AIS. In vivo two-photon calcium imaging demonstrates that Ndnf-INs in Scn1a+/â mice are recruited similarly to WT controls during arousal. These results suggest that Ndnf-INs are the only major IN subclass that does not prominently rely on Nav1.1 for AP generation and thus retain their excitability in DS. The discovery of a major IN subclass with preserved function in the Scn1a+/â mouse model adds further complexity to the "interneuron hypothesis" and highlights the importance of considering cell-type heterogeneity when investigating mechanisms underlying neurodevelopmental disorders.


Assuntos
Modelos Animais de Doenças , Epilepsias Mioclônicas , Interneurônios , Canal de Sódio Disparado por Voltagem NAV1.1 , Animais , Interneurônios/metabolismo , Interneurônios/fisiologia , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/fisiopatologia , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Feminino , Masculino , Potenciais de Ação/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
2.
Hippocampus ; 34(1): 7-13, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37933097

RESUMO

There are limited therapeutic options for patients with Dravet syndrome (DS). The equilibrative nucleoside transporters 1 (ENT1) mediate both the influx and efflux of adenosine across the cell membrane exerted beneficial effects in the treatment of epilepsy. This study aimed to evaluate the anticonvulsant effect of the ENT1 inhibitor in an animal model of DS (Scn1aE1099X/+ mice). J7 (5 mg/kg) treatment was efficacious in elevating seizure threshold in Scn1aE1099X/+ mice after hyperthermia exposure. Moreover, the J7 treatment significantly reduced the frequency of spontaneous excitatory post-synaptic currents (sEPSCs, ~35% reduction) without affecting the amplitude in dentate gyrus (DG) granule cells. Pretreatment with the adenosine A1 receptor (A1R) antagonist, DPCPX, abolished the J7 effects on sEPSCs. These observations suggest that the J7 shows an anticonvulsant effect in hyperthermia-induced seizures in Scn1aE1099X/+ mice. This effect possibly acts on presynaptic A1R-mediated signaling modulation in granule cells.


Assuntos
Epilepsias Mioclônicas , Epilepsia , Humanos , Camundongos , Animais , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Nucleosídeos/uso terapêutico , Epilepsias Mioclônicas/tratamento farmacológico , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Neurônios/metabolismo , Modelos Animais de Doenças , Canal de Sódio Disparado por Voltagem NAV1.1/genética
3.
Neurobiol Dis ; 179: 106059, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36868483

RESUMO

SCN1A gain-of-function variants are associated with early onset developmental and epileptic encephalopathies (DEEs) that possess distinct clinical features compared to Dravet syndrome caused by SCN1A loss-of-function. However, it is unclear how SCN1A gain-of-function may predispose to cortical hyper-excitability and seizures. Here, we first report the clinical features of a patient carrying a de novo SCN1A variant (T162I) associated with neonatal-onset DEE, and then characterize the biophysical properties of T162I and three other SCN1A variants associated with neonatal-onset DEE (I236V) and early infantile DEE (P1345S, R1636Q). In voltage clamp experiments, three variants (T162I, P1345S and R1636Q) exhibited changes in activation and inactivation properties that enhanced window current, consistent with gain-of-function. Dynamic action potential clamp experiments utilising model neurons incorporating Nav1.1. channels supported a gain-of-function mechanism for all four variants. Here, the T162I, I236V, P1345S, and R1636Q variants exhibited higher peak firing rates relative to wild type and the T162I and R1636Q variants produced a hyperpolarized threshold and reduced neuronal rheobase. To explore the impact of these variants upon cortical excitability, we used a spiking network model containing an excitatory pyramidal cell (PC) and parvalbumin positive (PV) interneuron population. SCN1A gain-of-function was modelled by enhancing the excitability of PV interneurons and then incorporating three simple forms of homeostatic plasticity that restored pyramidal cell firing rates. We found that homeostatic plasticity mechanisms exerted differential impact upon network function, with changes to PV-to-PC and PC-to-PC synaptic strength predisposing to network instability. Overall, our findings support a role for SCN1A gain-of-function and inhibitory interneuron hyperexcitability in early onset DEE. We propose a mechanism through which homeostatic plasticity pathways can predispose to pathological excitatory activity and contribute to phenotypic variability in SCN1A disorders.


Assuntos
Epilepsias Mioclônicas , Canal de Sódio Disparado por Voltagem NAV1.1 , Recém-Nascido , Humanos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Mutação com Ganho de Função , Interneurônios/metabolismo , Epilepsias Mioclônicas/metabolismo , Neurônios/patologia
4.
PLoS One ; 18(1): e0280842, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36701411

RESUMO

A purified preparation of cannabidiol (CBD), a cannabis constituent, has been approved for the treatment of intractable childhood epilepsies such as Dravet syndrome. Extensive pharmacological characterization of CBD shows activity at numerous molecular targets but its anticonvulsant mechanism(s) of action is yet to be delineated. Many suggest that the anticonvulsant action of CBD is the result of G protein-coupled receptor 55 (GPR55) inhibition. Here we assessed whether Gpr55 contributes to the strain-dependent seizure phenotypes of the Scn1a+/- mouse model of Dravet syndrome. The Scn1a+/- mice on a 129S6/SvEvTac (129) genetic background have no overt phenotype, while those on a [129 x C57BL/6J] F1 background exhibit a severe phenotype that includes hyperthermia-induced seizures, spontaneous seizures and reduced survival. We observed greater Gpr55 transcript expression in the cortex and hippocampus of mice on the seizure-susceptible F1 background compared to those on the seizure-resistant 129 genetic background, suggesting that Gpr55 might be a genetic modifier of Scn1a+/- mice. We examined the effect of heterozygous genetic deletion of Gpr55 and pharmacological inhibition of GPR55 on the seizure phenotypes of F1.Scn1a+/- mice. Heterozygous Gpr55 deletion and inhibition of GPR55 with CID2921524 did not affect the temperature threshold of a thermally-induced seizure in F1.Scn1a+/- mice. Neither was there an effect of heterozygous Gpr55 deletion observed on spontaneous seizure frequency or survival of F1.Scn1a+/- mice. Our results suggest that GPR55 antagonism may not be a suitable anticonvulsant target for Dravet syndrome drug development programs, although future research is needed to provide more definitive conclusions.


Assuntos
Canabidiol , Epilepsias Mioclônicas , Hipertermia Induzida , Convulsões Febris , Camundongos , Animais , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Camundongos Endogâmicos C57BL , Epilepsias Mioclônicas/tratamento farmacológico , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Convulsões/tratamento farmacológico , Convulsões/genética , Canabidiol/farmacologia , Canabidiol/uso terapêutico , Convulsões Febris/tratamento farmacológico , Convulsões Febris/genética , Receptores de Canabinoides/metabolismo
5.
Int J Mol Sci ; 24(2)2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36674476

RESUMO

In this article, we identified a novel epileptogenic variant (G307R) of the gene SLC6A1, which encodes the GABA transporter GAT-1. Our main goal was to investigate the pathogenic mechanisms of this variant, located near the neurotransmitter permeation pathway, and compare it with other variants located either in the permeation pathway or close to the lipid bilayer. The mutants G307R and A334P, close to the gates of the transporter, could be glycosylated with variable efficiency and reached the membrane, albeit inactive. Mutants located in the center of the permeation pathway (G297R) or close to the lipid bilayer (A128V, G550R) were retained in the endoplasmic reticulum. Applying an Elastic Network Model, to these and to other previously characterized variants, we found that G307R and A334P significantly perturb the structure and dynamics of the intracellular gate, which can explain their reduced activity, while for A228V and G362R, the reduced translocation to the membrane quantitatively accounts for the reduced activity. The addition of a chemical chaperone (4-phenylbutyric acid, PBA), which improves protein folding, increased the activity of GAT-1WT, as well as most of the assayed variants, including G307R, suggesting that PBA might also assist the conformational changes occurring during the alternative access transport cycle.


Assuntos
Epilepsias Mioclônicas , Proteínas da Membrana Plasmática de Transporte de GABA , Bicamadas Lipídicas , Humanos , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia
6.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35165147

RESUMO

Dravet syndrome (DS) is one of the most severe childhood epilepsies, characterized by intractable seizures and comorbidities including cognitive and social dysfunction and high premature mortality. DS is mainly caused by loss-of-function mutations in the Scn1a gene encoding Nav1.1 that is predominantly expressed in inhibitory parvalbumin-containing (PV) interneurons. Decreased Nav1.1 impairs PV cell function, contributing to DS phenotypes. Effective pharmacological therapy that targets defective PV interneurons is not available. The known role of brain-derived neurotrophic factor (BDNF) in the development and maintenance of interneurons, together with our previous results showing improved PV interneuronal function and antiepileptogenic effects of a TrkB receptor agonist in a posttraumatic epilepsy model, led to the hypothesis that early treatment with a TrkB receptor agonist might prevent or reduce seizure activity in DS mice. To test this hypothesis, we treated DS mice with LM22A-4 (LM), a partial agonist at the BDNF TrkB receptor, for 7 d starting at postnatal day 13 (P13), before the onset of spontaneous seizures. Results from immunohistochemistry, Western blot, whole-cell patch-clamp recording, and in vivo seizure monitoring showed that LM treatment increased the number of perisomatic PV interneuronal synapses around cortical pyramidal cells in layer V, upregulated Nav1.1 in PV neurons, increased inhibitory synaptic transmission, and decreased seizures and the mortality rate in DS mice. The results suggest that early treatment with a partial TrkB receptor agonist may be a promising therapeutic approach to enhance PV interneuron function and reduce epileptogenesis and premature death in DS.


Assuntos
Benzamidas/uso terapêutico , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/mortalidade , Receptor trkB/agonistas , Receptor trkB/metabolismo , Convulsões/etiologia , Convulsões/genética , Animais , Epilepsias Mioclônicas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Neocórtex/citologia , Células Piramidais/metabolismo , Receptor trkB/genética
7.
Nat Commun ; 13(1): 161, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013317

RESUMO

Dravet syndrome is a severe epileptic encephalopathy caused primarily by haploinsufficiency of the SCN1A gene. Repetitive seizures can lead to endurable and untreatable neurological deficits. Whether this severe pathology is reversible after symptom onset remains unknown. To address this question, we generated a Scn1a conditional knock-in mouse model (Scn1a Stop/+) in which Scn1a expression can be re-activated on-demand during the mouse lifetime. Scn1a gene disruption leads to the development of seizures, often associated with sudden unexpected death in epilepsy (SUDEP) and behavioral alterations including hyperactivity, social interaction deficits and cognitive impairment starting from the second/third week of age. However, we showed that Scn1a gene re-activation when symptoms were already manifested (P30) led to a complete rescue of both spontaneous and thermic inducible seizures, marked amelioration of behavioral abnormalities and normalization of hippocampal fast-spiking interneuron firing. We also identified dramatic gene expression alterations, including those associated with astrogliosis in Dravet syndrome mice, that, accordingly, were rescued by Scn1a gene expression normalization at P30. Interestingly, regaining of Nav1.1 physiological level rescued seizures also in adult Dravet syndrome mice (P90) after months of repetitive attacks. Overall, these findings represent a solid proof-of-concept highlighting that disease phenotype reversibility can be achieved when Scn1a gene activity is efficiently reconstituted in brain cells.


Assuntos
Disfunção Cognitiva/genética , Epilepsias Mioclônicas/genética , Hipocampo/metabolismo , Interneurônios/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Morte Súbita Inesperada na Epilepsia/prevenção & controle , Potenciais de Ação/fisiologia , Animais , Cerebelo/metabolismo , Cerebelo/fisiopatologia , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/prevenção & controle , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/fisiopatologia , Epilepsias Mioclônicas/prevenção & controle , Técnicas de Introdução de Genes , Terapia Genética/métodos , Hipocampo/fisiopatologia , Humanos , Interneurônios/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1/deficiência , Morte Súbita Inesperada na Epilepsia/patologia
8.
Mol Neurobiol ; 59(2): 748-761, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34766239

RESUMO

Dravet syndrome (DS) is a form of severe childhood-onset refractory epilepsy typically caused by a heterozygous loss-of-function mutation. DS patient-derived induced pluripotent stem cells (iPSCs) are appropriate human cells for exploring disease mechanisms and testing new therapeutic strategies in vitro. Repeated spontaneous seizures can cause neuroinflammatory reactions and oxidative stress, resulting in neuronal toxicity, neuronal dysfunction, blood-brain barrier disruption, and hippocampal inflammation. Antiepileptic drug therapy does not delay the development of chronic epilepsy. The application of mesenchymal stem cells (MSCs) is one therapeutic strategy for thwarting epilepsy development. This study evaluated the effects of human umbilical cord mesenchymal stem cell-conditioned medium (HUMSC-CM) in a new in vitro model of neurons differentiated from DS patient-derived iPSCs. In the presence of HUMSC-CM, increases in superoxide dismutase 1 (SOD1), superoxide dismutase 2 (SOD2), glutathione peroxidase (GPX), and glutathione (GSH) levels were found to contribute to a reduction in reactive oxygen species (ROS) levels. In parallel, inflammation was rescued in DS patient-derived neuronal cells via increased expression of anti-inflammatory cytokines (TGF-ß, IL-6, and IL-10) and significant downregulation of tumor necrosis factor-α and interleukin-1ß expression. The intracellular calcium concentration ([Ca2+]i) and malondialdehyde (MDA) and ROS levels were decreased in DS patient-derived cells. In addition, action potential (AP) firing ability was enhanced by HUMSC-CM. In conclusion, HUMSC-CM can effectively eliminate ROS, affect migration and neurogenesis, and promote neurons to enter a highly functional state. Therefore, HUMSC-CM is a promising therapeutic strategy for the clinical treatment of refractory epilepsy such as DS.


Assuntos
Epilepsias Mioclônicas , Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Diferenciação Celular , Criança , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Cordão Umbilical/metabolismo
9.
Neuropharmacology ; 205: 108897, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34822817

RESUMO

Dravet syndrome is a catastrophic childhood epilepsy with multiple seizure types that are refractory to treatment. The endocannabinoid system regulates neuronal excitability so a deficit in endocannabinoid signaling could lead to hyperexcitability and seizures. Thus, we sought to determine whether a deficiency in the endocannabinoid system might contribute to seizure phenotypes in a mouse model of Dravet syndrome and whether enhancing endocannabinoid tone is anticonvulsant. Scn1a+/- mice model the clinical features of Dravet syndrome: hyperthermia-induced seizures, spontaneous seizures and reduced survival. We examined whether Scn1a+/- mice exhibit deficits in the endocannabinoid system by measuring brain cannabinoid receptor expression and endocannabinoid concentrations. Next, we determined whether pharmacologically enhanced endocannabinoid tone was anticonvulsant in Scn1a+/- mice. We used GAT229, a positive allosteric modulator of the cannabinoid (CB1) receptor, and ABX-1431, a compound that inhibits the degradation of the endocannabinoid 2-arachidonoylglycerol (2-AG). The Scn1a+/- phenotype is strain-dependent with mice on a 129S6/SvEvTac (129) genetic background having no overt phenotype and those on an F1 (129S6/SvEvTac x C57BL/6J) background exhibiting a severe epilepsy phenotype. We observed lower brain cannabinoid CB1 receptor expression in the seizure-susceptible F1 compared to seizure-resistant 129 strain, suggesting an endocannabinoid deficiency might contribute to seizure susceptibility. GAT229 and ABX-1431 were anticonvulsant against hyperthermia-induced seizures. However, subchronic ABX1431 treatment increased spontaneous seizure frequency despite reducing seizure severity. Cnr1 is a putative genetic modifier of epilepsy in the Scn1a+/- mouse model of Dravet syndrome. Compounds that increase endocannabinoid tone could be developed as novel treatments for Dravet syndrome.


Assuntos
Anticonvulsivantes/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , Endocanabinoides/antagonistas & inibidores , Endocanabinoides/metabolismo , Epilepsias Mioclônicas/tratamento farmacológico , Epilepsias Mioclônicas/metabolismo , Receptor CB1 de Canabinoide/agonistas , Animais , Modelos Animais de Doenças , Endocanabinoides/deficiência , Indóis/farmacologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Piperazinas/farmacologia , Pirrolidinas/farmacologia
10.
J Cereb Blood Flow Metab ; 42(1): 197-212, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34515548

RESUMO

To assess if magnetic resonance spectroscopy (MRS)-measured Glutamate (Glu) and GABA reflect excitatory and inhibitory neural activities, respectively, we conducted MRS measurements along with two-photon mesoscopic imaging of calcium signals in excitatory and inhibitory neurons of living, unanesthetized mice. For monitoring stimulus-driven activations of a brain region, MRS signals and mesoscopic neural activities were measured during two consecutive sessions of 15-min prolonged sensory stimulations. In the first session, putative excitatory neuronal activities were increased, while inhibitory neuronal activities remained at the baseline level. In the second half, while excitatory neuronal activities remained elevated, inhibitory neuronal activities were significantly enhanced. We assessed regional neurochemical statuses by measuring MRS signals, which were overall in accordance with the neural activities, and neuronal activities and neurochemical statuses in a mouse model of Dravet syndrome under resting condition. Mesoscopic assessments showed that activities of inhibitory neurons in the cortex were diminished relative to wild-type mice in contrast to spared activities of excitatory neurons. Consistent with these observations, the Dravet model exhibited lower concentrations of GABA than wild-type controls. Collectively, the current investigations demonstrate that MRS-measured Glu and GABA can reflect spontaneous and stimulated activities of neurons producing and releasing these neurotransmitters in an awake condition.


Assuntos
Epilepsias Mioclônicas/metabolismo , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Vigília , Ácido gama-Aminobutírico/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Espectroscopia de Ressonância Magnética , Masculino , Camundongos
11.
Cell Mol Life Sci ; 78(19-20): 6409-6430, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34405255

RESUMO

Neuroserpin is a serine protease inhibitor identified in a search for proteins implicated in neuronal axon growth and synapse formation. Since its discovery over 30 years ago, it has been the focus of active research. Many efforts have concentrated in elucidating its neuroprotective role in brain ischemic lesions, the structural bases of neuroserpin conformational change and the effects of neuroserpin polymers that underlie the neurodegenerative disease FENIB (familial encephalopathy with neuroserpin inclusion bodies), but the investigation of the physiological roles of neuroserpin has increased over the last years. In this review, we present an updated and critical revision of the current literature dealing with neuroserpin, covering all aspects of research including the expression and physiological roles of neuroserpin, both inside and outside the nervous system; its inhibitory and non-inhibitory mechanisms of action; the molecular structure of the monomeric and polymeric conformations of neuroserpin, including a detailed description of the polymerisation mechanism; and the involvement of neuroserpin in human disease, with particular emphasis on FENIB. Finally, we briefly discuss the identification by genome-wide screening of novel neuroserpin variants and their possible pathogenicity.


Assuntos
Neuropeptídeos/metabolismo , Serpinas/metabolismo , Animais , Axônios/metabolismo , Epilepsias Mioclônicas/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Humanos , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Polimerização
12.
Neurobiol Dis ; 157: 105423, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34144125

RESUMO

BACKGROUND: Dravet syndrome is a rare, severe pediatric epileptic encephalopathy associated with intellectual and motor disabilities. Proteomic profiling in a mouse model of Dravet syndrome can provide information about the molecular consequences of the genetic deficiency and about pathophysiological mechanisms developing during the disease course. METHODS: A knock-in mouse model of Dravet syndrome with Scn1a haploinsufficiency was used for whole proteome, seizure, and behavioral analysis. Hippocampal tissue was dissected from two- (prior to epilepsy manifestation) and four- (following epilepsy manifestation) week-old male mice and analyzed using LC-MS/MS with label-free quantification. Proteomic data sets were subjected to bioinformatic analysis including pathway enrichment analysis. The differential expression of selected proteins was confirmed by immunohistochemical staining. RESULTS: The findings confirmed an increased susceptibility to hyperthermia-associated seizures, the development of spontaneous seizures, and behavioral alterations in the novel Scn1a-A1873V mouse model of Dravet syndrome. As expected, proteomic analysis demonstrated more pronounced alterations following epilepsy manifestation. In particular, proteins involved in neurotransmitter dynamics, receptor and ion channel function, synaptic plasticity, astrogliosis, neoangiogenesis, and nitric oxide signaling showed a pronounced regulation in Dravet mice. Pathway enrichment analysis identified several significantly regulated pathways at the later time point, with pathways linked to synaptic transmission and glutamatergic signaling dominating the list. CONCLUSION: In conclusion, the whole proteome analysis in a mouse model of Dravet syndrome demonstrated complex molecular alterations in the hippocampus. Some of these alterations may have an impact on excitability or may serve a compensatory function, which, however, needs to be further confirmed by future investigations. The proteomic data indicate that, due to the molecular consequences of the genetic deficiency, the pathophysiological mechanisms may become more complex during the course of the disease. As a result, the management of Dravet syndrome may need to consider further molecular and cellular alterations. Ensuing functional follow-up studies, this data set may provide valuable guidance for the future development of novel therapeutic approaches.


Assuntos
Epilepsias Mioclônicas/metabolismo , Hipocampo/metabolismo , Proteômica , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Animais , Comportamento Animal , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Carbono-Nitrogênio Ligases/metabolismo , Cromatografia Líquida , Modelos Animais de Doenças , Progressão da Doença , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Teste de Labirinto em Cruz Elevado , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/fisiopatologia , Feminino , Técnicas de Introdução de Genes , Gliose , Haploinsuficiência , Hipertermia/fisiopatologia , Imuno-Histoquímica , Masculino , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Neovascularização Fisiológica , Plasticidade Neuronal , Óxido Nítrico , Teste de Campo Aberto , Teste de Desempenho do Rota-Rod , Transdução de Sinais , Comportamento Social , Transmissão Sináptica , Espectrometria de Massas em Tandem , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , ras-GRF1/metabolismo
13.
Neurochem Res ; 46(9): 2249-2261, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34041623

RESUMO

Dravet syndrome (DS) is a rare genetic encephalopathy that is characterized by severe seizures and highly resistant to commonly used antiepileptic drugs (AEDs). In 2020, FDA has approved fenfluramine (FFA) for treatment of seizures associated with DS. However, the clinically used FFA is a racemic mixture (i.e. (±)-FFA), that is substantially metabolized to norfenfluramine (norFFA), and it is presently not known whether the efficacy of FFA is due to a single enantiomer of FFA, or to both, and whether the norFFA enantiomers also contribute significantly. In this study, the antiepileptic activity of enantiomers of FFA (i.e. (+)-FFA and (-)-FFA) and norFFA (i.e. (+)-norFFA and (-)-norFFA) was explored using the zebrafish scn1Lab-/- mutant model of DS. To validate the experimental conditions used, we assessed the activity of various AEDs typically used in the fight against DS, including combination therapy. Overall, our results are highly consistent with the treatment algorithm proposed by the updated current practice in the clinical management of DS. Our results show that (+)-FFA, (-)-FFA and (+)-norFFA displayed significant antiepileptic effects in the preclinical model, and thus can be considered as compounds actively contributing to the clinical efficacy of FFA. In case of (-)-norFFA, the results were less conclusive. We also investigated the uptake kinetics of the enantiomers of FFA and norFFA in larval zebrafish heads. The data show that the total uptake of each compound increased in a time-dependent fashion. A somewhat similar uptake was observed for the (+)-norFFA and (-)-norFFA, implying that the levo/dextrotation of the structure did not dramatically affect the uptake. Significantly, when comparing (+)-FFA with the less lipophilic (+)-norFFA, the data clearly show that the nor-metabolite of FFA is taken up less than the parent compound.


Assuntos
Anticonvulsivantes/uso terapêutico , Epilepsias Mioclônicas/tratamento farmacológico , Fenfluramina/uso terapêutico , Norfenfluramina/uso terapêutico , Animais , Anticonvulsivantes/química , Anticonvulsivantes/metabolismo , Anticonvulsivantes/farmacocinética , Epilepsias Mioclônicas/metabolismo , Fenfluramina/química , Fenfluramina/metabolismo , Fenfluramina/farmacocinética , Cabeça/fisiologia , Norfenfluramina/química , Norfenfluramina/metabolismo , Norfenfluramina/farmacocinética , Estereoisomerismo , Peixe-Zebra
14.
Elife ; 102021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-34013884

RESUMO

Glutamatergic neurons in the retrotrapezoid nucleus (RTN) function as respiratory chemoreceptors by regulating breathing in response to tissue CO2/H+. The RTN and greater parafacial region may also function as a chemosensing network composed of CO2/H+-sensitive excitatory and inhibitory synaptic interactions. In the context of disease, we showed that loss of inhibitory neural activity in a mouse model of Dravet syndrome disinhibited RTN chemoreceptors and destabilized breathing (Kuo et al., 2019). Despite this, contributions of parafacial inhibitory neurons to control of breathing are unknown, and synaptic properties of RTN neurons have not been characterized. Here, we show the parafacial region contains a limited diversity of inhibitory neurons including somatostatin (Sst)-, parvalbumin (Pvalb)-, and cholecystokinin (Cck)-expressing neurons. Of these, Sst-expressing interneurons appear uniquely inhibited by CO2/H+. We also show RTN chemoreceptors receive inhibitory input that is withdrawn in a CO2/H+-dependent manner, and chemogenetic suppression of Sst+ parafacial neurons, but not Pvalb+ or Cck+ neurons, increases baseline breathing. These results suggest Sst-expressing parafacial neurons contribute to RTN chemoreception and respiratory activity.


Assuntos
Dióxido de Carbono/metabolismo , Células Quimiorreceptoras/metabolismo , Epilepsias Mioclônicas/metabolismo , Hidrogênio/metabolismo , Núcleos Intralaminares do Tálamo/metabolismo , Pulmão/inervação , Respiração , Somatostatina/metabolismo , Animais , Modelos Animais de Doenças , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/fisiopatologia , Feminino , Ácido Glutâmico/metabolismo , Núcleos Intralaminares do Tálamo/fisiopatologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Inibição Neural , Somatostatina/genética , Transmissão Sináptica
15.
Cell Calcium ; 96: 102380, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33676317

RESUMO

Epilepsy is a severe neurological disorder defined by spontaneous seizures. Current treatment options fail in a large proportion of patients, while questions as to the basic mechanisms of seizure initiation and propagation remain. Advances in imaging of seizures in experimental model systems could lead to a better understanding of mechanisms of seizures and epilepsy. Recent studies have used two-photon calcium imaging (2 P imaging) in awake, behaving mice in head-fixed preparations to image seizures in vivo at high speed and cellular-level resolution to identify key seizure-related cell classes. Here, we discuss such advances and present 2 P imaging data of excitatory neurons and defined subsets of cerebral cortex GABAergic inhibitory interneurons during naturalistic seizures in a mouse model of Dravet syndrome (Scn1a+/- mice) along with other behavioral measures. Results demonstrate differential recruitment of discrete interneuron subclasses, which could inform mechanisms of seizure generation and propagation in Dravet syndrome and other epilepsies.


Assuntos
Cálcio/metabolismo , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Convulsões/metabolismo , Técnicas Estereotáxicas , Vigília/fisiologia , Animais , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Convulsões/genética
16.
Brain Res Bull ; 170: 81-89, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33581311

RESUMO

Aberrant expression or dysfunction of a number of genes in the brain contributes to epilepsy, a common neurological disorder characterized by recurrent seizures. Local overexpression of arachidonate lipoxygenase 3 (ALOXE3), a key enzyme for arachidonic acid (AA) metabolic pathway, alleviates seizure severities. However, the relationship between the ALOXE3 gene mutation and epilepsy has not been reported until now. Here we firstly characterized the promoter of human ALOXE3 gene and found that the ALOXE3 promoter could drive luciferase gene expression in the human HEK-293 and SH-SY5Y cells. We then screened the ALOXE3 promoter region and all coding exons from those patients with Dravet syndrome and identified 5 variants c.-163T > C, c.-50C > G, c.-37G > A, c. + 228G > A and c. + 290G > T in the promoter region and one missense variant c.1939A > G (p.I647 V) in the exon. Of these variants in the promoter region, only -50C > G was a novel variant located on the transcriptional factor NFII-I binding element. Luciferase reporter gene analyses indicated that the c.-50C > G could decrease gene expression by preventing the TFII-I's binding. In addition, the variant p.I647 V was conserved among all analyzed species and located within the ALOXE3 functional domain for catalyzing its substrate. In cultured cell lines, overexpression of ALOXE3 significantly decreased the cellular AA levels and overexpression of ALOXE3-I647 V could restore the AA levels, suggesting that the p.I647 V mutant led to a decrease in enzyme activity. Taken together, the present study proposes that the identified ALOXE3 variants potentially contribute to the AA-pathway-mediated epileptogenesis, which should provide a novel avenue for clinical diagnosis of epilepsy.


Assuntos
Encéfalo/metabolismo , Epilepsias Mioclônicas/genética , Lipoxigenase/genética , Mutação , Alelos , Epilepsias Mioclônicas/metabolismo , Células HEK293 , Humanos , Lipoxigenase/metabolismo , Fenótipo , Regiões Promotoras Genéticas
17.
Brain Res ; 1751: 147157, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33069731

RESUMO

Dravet Syndrome (DS) is a genetic neurodevelopmental disease. Recurrent severe seizures begin in infancy and co-morbidities follow, including developmental delay, cognitive and behavioral dysfunction. A majority of DS patients have an SCN1A heterozygous gene mutation. This mutation causes a loss-of-function in inhibitory neurons, initiating seizure onset. We have investigated whether the sodium channelopathy may result in structural changes in the DS model independent of seizures. Morphometric analyses of axons within the corpus callosum were completed at P16 and P50 in Scn1a heterozygote KO male mice and their age-matched wild-type littermates. Trainable machine learning algorithms were used to examine electron microscopy images of ~400 myelinated axons per animal, per genotype, including myelinated axon cross-section area, frequency distribution and g-ratios. Pilot data for Scn1a heterozygote KO mice demonstrate the average axon caliber was reduced in developing and adult mice. Qualitative analysis also shows micro-features marking altered myelination at P16 in the DS model, with myelin out-folding and myelin debris within phagocytic cells. The data has indicated, in the absence of behavioral seizures, factors that governed a shift toward small calibre axons at P16 have persisted in adult Scn1a heterozygote KO corpus callosum. The pilot study provides a basis for future meta-analysis that will enable robust estimates of the effects of the sodium channelopathy on axon architecture. We propose that early therapeutic strategies in DS could help minimize the effect of sodium channelopathies, beyond the impact of overt seizures, and therefore achieve better long-term treatment outcomes.


Assuntos
Epilepsias Mioclônicas/genética , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Animais , Axônios/metabolismo , Axônios/fisiologia , Encéfalo/metabolismo , Corpo Caloso/metabolismo , Corpo Caloso/fisiopatologia , Modelos Animais de Doenças , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/fisiopatologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Microscopia Eletrônica/métodos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Neurogênese , Projetos Piloto , Convulsões/fisiopatologia , Canais de Sódio/genética , Canais de Sódio/metabolismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-33049405

RESUMO

MARCHF6 is a large multi-pass E3 ubiquitin ligase embedded in the membranes of the endoplasmic reticulum. It participates in endoplasmic reticulum associated degradation, including autoubiquitination, and many of its identified substrates are involved in sterol and lipid metabolism. Post-translationally, MARCHF6 expression is attuned to cholesterol status, with high cholesterol preventing its degradation and hence boosting MARCHF6 levels. By modulating MARCHF6 activity, cholesterol may regulate other aspects of cell metabolism beyond the known repertoire. Whilst we have learnt much about MARCHF6 in the past decade, there are still many more mysteries to be unravelled to fully understand its regulation, substrates, and role in human health and disease.


Assuntos
Colesterol/metabolismo , Síndrome de Cri-du-Chat/genética , Degradação Associada com o Retículo Endoplasmático , Epilepsias Mioclônicas/genética , Proteínas de Membrana/genética , Obesidade/genética , Processamento de Proteína Pós-Traducional , Ubiquitina-Proteína Ligases/genética , Animais , Índice de Massa Corporal , Síndrome de Cri-du-Chat/metabolismo , Síndrome de Cri-du-Chat/patologia , Retículo Endoplasmático/metabolismo , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia , Humanos , Metabolismo dos Lipídeos/genética , Proteínas de Membrana/deficiência , Obesidade/metabolismo , Obesidade/patologia , Polimorfismo de Nucleotídeo Único , Proteólise , Ubiquitina-Proteína Ligases/deficiência , Ubiquitinação
19.
Neurobiol Dis ; 148: 105209, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271326

RESUMO

Dravet syndrome (Dravet) is a rare, severe childhood-onset epilepsy, caused by heterozygous de novo mutations in the SCN1A gene, encoding for the alpha subunit of the voltage-gated sodium channel, NaV1.1. The neuronal basis of Dravet is debated, with evidence favoring reduced function of inhibitory neurons, that might be transient, or enhanced activity of excitatory cells. Here, we utilized Dravet mice to trace developmental changes in the hippocampal CA1 circuit, examining the properties of CA1 horizontal stratum-oriens (SO) interneurons and pyramidal neurons, through the pre-epileptic, severe and stabilization stages of Dravet. Our data indicate that reduced function of SO interneurons persists from the pre-epileptic through the stabilization stages, with the greatest functional impairment observed during the severe stage. In contrast, opposing changes were detected in CA1 excitatory neurons, with a transient increase in their excitability during the pre-epileptic stage, followed by reduced excitability at the severe stage. Interestingly, alterations in the function of both inhibitory and excitatory neurons were more pronounced when the firing was evoked by synaptic stimulation, implying that loss of function of NaV1.1 may also affect somatodendritic functions. These results suggest a complex pathophysiological mechanism and indicate that the developmental trajectory of this disease is governed by reciprocal functional changes in both excitatory and inhibitory neurons.


Assuntos
Potenciais de Ação/fisiologia , Região CA1 Hipocampal/metabolismo , Epilepsias Mioclônicas/metabolismo , Interneurônios/metabolismo , Células Piramidais/metabolismo , Animais , Região CA1 Hipocampal/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/fisiopatologia , Interneurônios/fisiologia , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Neurônios , Células Piramidais/fisiologia , Convulsões/genética , Convulsões/metabolismo , Convulsões/fisiopatologia
20.
Genetics ; 215(4): 1055-1066, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32554600

RESUMO

Dravet syndrome is a developmental epileptic encephalopathy caused by pathogenic variation in SCN1A To characterize the pathogenic substitution (p.H939R) of a local individual with Dravet syndrome, fibroblast cells from the individual were reprogrammed to pluripotent stem cells and differentiated into neurons. Sodium currents of these neurons were compared with healthy control induced neurons. A novel Scn1aH939R/+ mouse model was generated with the p.H939R substitution. Immunohistochemistry and electrophysiological experiments were performed on hippocampal slices of Scn1aH939R/+ mice. We found that the sodium currents recorded in the proband-induced neurons were significantly smaller and slower compared to wild type (WT). The resting membrane potential and spike amplitude were significantly depolarized in the proband-induced neurons. Similar differences in resting membrane potential and spike amplitude were observed in the interneurons of the hippocampus of Scn1aH939R/+ mice. The Scn1aH939R/+ mice showed the characteristic features of a Dravet-like phenotype: increased mortality and both spontaneous and heat-induced seizures. Immunohistochemistry showed a reduction in amount of parvalbumin and vesicular acetylcholine transporter in the hippocampus of Scn1aH939R/+ compared to WT mice. Overall, these results underline hyper-excitability of the hippocampal CA1 circuit of this novel mouse model of Dravet syndrome which, under certain conditions, such as temperature, can trigger seizure activity. This hyper-excitability is due to the altered electrophysiological properties of pyramidal neurons and interneurons which are caused by the dysfunction of the sodium channel bearing the p.H939R substitution. This novel Dravet syndrome model also highlights the reduction in acetylcholine and the contribution of pyramidal cells, in addition to interneurons, to network hyper-excitability.


Assuntos
Região CA1 Hipocampal/patologia , Modelos Animais de Doenças , Epilepsias Mioclônicas/patologia , Fibroblastos/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Interneurônios/patologia , Células Piramidais/patologia , Animais , Região CA1 Hipocampal/metabolismo , Eletrofisiologia , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Interneurônios/metabolismo , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Células Piramidais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...