Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 451
Filtrar
1.
Lancet Neurol ; 21(5): 417-427, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35429480

RESUMO

BACKGROUND: CDKL5 deficiency disorder (CDD) is a rare, X-linked, developmental and epileptic encephalopathy characterised by severe global developmental impairment and seizures that can begin in the first few months after birth and are often treatment refractory. Ganaxolone, an investigational neuroactive steroid, reduced seizure frequency in an open-label, phase 2 trial that included patients with CDD. We aimed to further assess the efficacy and safety of ganaxolone in patients with CDD-associated refractory epilepsy. METHODS: In the double-blind phase of this randomised, placebo-controlled, phase 3 trial, done at 39 outpatient clinics in eight countries (Australia, France, Israel, Italy, Poland, Russia, the UK, and the USA), patients were eligible if they were aged 2-21 years with a pathogenic or probably pathogenic CDKL5 variant and at least 16 major motor seizures (defined as bilateral tonic, generalised tonic-clonic, bilateral clonic, atonic, or focal to bilateral tonic-clonic) per 28 days in each 4-week period of an 8-week historical period. After a 6-week prospective baseline period, patients were randomly assigned (1:1) via an interactive web response system to receive either enteral adjunctive ganaxolone or matching enteral adjunctive placebo (maximum dose 63 mg/kg per day for patients weighing ≤28 kg or 1800 mg/day for patients weighing >28 kg) for 17 weeks. Patients, caregivers, investigators (including those analysing data), trial staff, and the sponsor (other than the investigational product manager) were masked to treatment allocation. The primary efficacy endpoint was percentage change in median 28-day major motor seizure frequency from the baseline period to the 17-week double-blind phase and was analysed (using a Wilcoxon-rank sum test) in all patients who received at least one dose of trial treatment and for whom baseline data were available. Safety (compared descriptively across groups) was analysed in all patients who received at least one dose of trial treatment. This study is registered with ClinicalTrials.gov, NCT03572933, and the open-label extension phase is ongoing. FINDINGS: Between June 25, 2018, and July 2, 2020, 114 patients were screened for eligibility, of whom 101 (median age 6 years [IQR 3 to 10]) were randomly assigned to receive either ganaxolone (n=50) or placebo (n=51). All patients received at least one dose of a study drug, but seizure frequency for one patient in the ganaxolone group was not recorded at baseline and so the primary endpoint was analysed in a population of 100 patients. There was a median percentage change in 28-day major motor seizure frequency of -30·7% (IQR -49·5 to -1·9) in the ganaxolone group and of -6·9% (-24·1 to 39·7) in the placebo group (p=0·0036). The Hodges-Lehmann estimate of median difference in responses to ganaxolone versus placebo was -27·1% (95% CI -47·9 to - 9·6). Treatment-emergent adverse events occurred in 43 (86%) of 50 patients in the ganaxolone group and in 45 (88%) of 51 patients in the placebo group. Somnolence, pyrexia, and upper respiratory tract infections occurred in at least 10% of patients in the ganaxolone group and more frequently than in the placebo group. Serious adverse events occurred in six (12%) patients in the ganaxolone group and in five (10%) patients in the placebo group. Two (4%) patients in the ganaxolone group and four (8%) patients in the placebo group discontinued the trial. There were no deaths in the double-blind phase. INTERPRETATION: Ganaxolone significantly reduced the frequency of CDD-associated seizures compared with placebo and was generally well tolerated. Results from what is, to our knowledge, the first controlled trial in CDD suggest a potential treatment benefit for ganaxolone. Long-term treatment is being assessed in the ongoing open-label extension phase of this trial. FUNDING: Marinus Pharmaceuticals.


Assuntos
Síndromes Epilépticas , Pregnanolona , Espasmos Infantis , Criança , Pré-Escolar , Método Duplo-Cego , Síndromes Epilépticas/tratamento farmacológico , Síndromes Epilépticas/enzimologia , Humanos , Lactente , Pregnanolona/análogos & derivados , Estudos Prospectivos , Proteínas Serina-Treonina Quinases/deficiência , Convulsões/tratamento farmacológico , Convulsões/enzimologia , Espasmos Infantis/tratamento farmacológico , Espasmos Infantis/enzimologia , Resultado do Tratamento
2.
Nucleic Acids Res ; 49(22): 13108-13121, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34878141

RESUMO

Mutations in genes encoding mitochondrial aminoacyl-tRNA synthetases are linked to diverse diseases. However, the precise mechanisms by which these mutations affect mitochondrial function and disease development are not fully understood. Here, we develop a Drosophila model to study the function of dFARS2, the Drosophila homologue of the mitochondrial phenylalanyl-tRNA synthetase, and further characterize human disease-associated FARS2 variants. Inactivation of dFARS2 in Drosophila leads to developmental delay and seizure. Biochemical studies reveal that dFARS2 is required for mitochondrial tRNA aminoacylation, mitochondrial protein stability, and assembly and enzyme activities of OXPHOS complexes. Interestingly, by modeling FARS2 mutations associated with human disease in Drosophila, we provide evidence that expression of two human FARS2 variants, p.G309S and p.D142Y, induces seizure behaviors and locomotion defects, respectively. Together, our results not only show the relationship between dysfunction of mitochondrial aminoacylation system and pathologies, but also illustrate the application of Drosophila model for functional analysis of human disease-causing variants.


Assuntos
Deficiências do Desenvolvimento/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Mitocondriais/genética , Mutação , Fenilalanina-tRNA Ligase/genética , RNA de Transferência/genética , Convulsões/genética , Animais , Linhagem Celular , Deficiências do Desenvolvimento/enzimologia , Modelos Animais de Doenças , Proteínas de Drosophila/deficiência , Drosophila melanogaster/enzimologia , Técnicas de Silenciamento de Genes , Humanos , Microscopia Eletrônica de Transmissão , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/deficiência , Fosforilação Oxidativa , Fenilalanina-tRNA Ligase/deficiência , RNA de Transferência/metabolismo , Convulsões/enzimologia , Aminoacilação de RNA de Transferência
3.
Blood ; 137(26): 3660-3669, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33763700

RESUMO

Glycosylphosphatidylinositol (GPI) is a glycolipid that anchors >150 proteins to the cell surface. Pathogenic variants in several genes that participate in GPI biosynthesis cause inherited GPI deficiency disorders. Here, we reported that homozygous null alleles of PIGG, a gene involved in GPI modification, are responsible for the rare Emm-negative blood phenotype. Using a panel of K562 cells defective in both the GPI-transamidase and GPI remodeling pathways, we show that the Emm antigen, whose molecular basis has remained unknown for decades, is carried only by free GPI and that its epitope is composed of the second and third ethanolamine of the GPI backbone. Importantly, we show that the decrease in Emm expression in several inherited GPI deficiency patients is indicative of GPI defects. Overall, our findings establish Emm as a novel blood group system, and they have important implications for understanding the biological function of human free GPI.


Assuntos
Antígenos de Grupos Sanguíneos , Deficiências do Desenvolvimento , Glicosilfosfatidilinositóis/deficiência , Glicosilfosfatidilinositóis/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool) , Convulsões , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Deficiências do Desenvolvimento/enzimologia , Deficiências do Desenvolvimento/genética , Glicosilfosfatidilinositóis/genética , Humanos , Células K562 , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Convulsões/enzimologia , Convulsões/genética
4.
Eur J Pharmacol ; 898: 173959, 2021 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-33617826

RESUMO

Epilepsy is a serious neurological disorder. Available antiepileptic drugs are still lacking. Hydrogen sulfide (H2S), a neuron-protective endogenous gasotransmitter, is reported to have effect on epilepsy. But it remains to be determined for its mechanism. In the present study, we found that a novel carbazole-based H2S donor could effectively suppress pentylenetetrazol-induced seizures in rats. The H2S donor could alleviate not only the epileptic behavior of animals but also the hippocampal EEG activity of seizures. The H2S donor down-regulated the expression of aquaporin 4 in the hippocampus of epilepsy rats. The H2S donor also decreased the seizure-induced release of inflammatory cytokines including IL-1ß, IL-6 and TNF-α. In addition, the H2S donor increased protein kinase C (PKC) expression in the hippocampus of epilepsy rats. These effects of the H2S donor on epilepsy rats were attenuated after blockade of PKC signaling by Go6983, suggesting that PKC signaling participated in the antiepileptic process of H2S donor. Taken together, the H2S donor has a beneficial effect on epilepsy control in a PKC-dependent manner.


Assuntos
Anticonvulsivantes/farmacologia , Ondas Encefálicas/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Proteína Quinase C/metabolismo , Convulsões/prevenção & controle , Animais , Aquaporina 4/genética , Aquaporina 4/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Hipocampo/enzimologia , Hipocampo/fisiopatologia , Mediadores da Inflamação/metabolismo , Masculino , Pentilenotetrazol , Proteína Quinase C/genética , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/enzimologia , Convulsões/fisiopatologia , Transdução de Sinais
5.
Neuropharmacology ; 184: 108405, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33212114

RESUMO

Sudden unexpected death in epilepsy (SUDEP) is a leading cause of death in patients with refractory epilepsy. Centrally-mediated respiratory dysfunction has been identified as one of the principal mechanisms responsible for SUDEP. Seizures generate a surge in adenosine release. Elevated adenosine levels suppress breathing. Insufficient metabolic clearance of a seizure-induced adenosine surge might be a precipitating factor in SUDEP. In order to deliver targeted therapies to prevent SUDEP, reliable biomarkers must be identified to enable prompt intervention. Because of the integral role of the phrenic nerve in breathing, we hypothesized that suppression of phrenic nerve activity could be utilized as predictive biomarker for imminent SUDEP. We used a rat model of kainic acid-induced seizures in combination with pharmacological suppression of metabolic adenosine clearance to trigger seizure-induced death in tracheostomized rats. Recordings of EEG, blood pressure, and phrenic nerve activity were made concomitant to the seizure. We found suppression of phrenic nerve burst frequency to 58.9% of baseline (p < 0.001, one-way ANOVA) which preceded seizure-induced death; importantly, irregularities of phrenic nerve activity were partly reversible by the adenosine receptor antagonist caffeine. Suppression of phrenic nerve activity may be a useful biomarker for imminent SUDEP. The ability to reliably detect the onset of SUDEP may be instrumental in the timely administration of potentially lifesaving interventions.


Assuntos
Adenosina Quinase/antagonistas & inibidores , Nervo Frênico/enzimologia , Nervo Frênico/fisiopatologia , Convulsões/enzimologia , Convulsões/fisiopatologia , Morte Súbita Inesperada na Epilepsia , Adenosina Quinase/metabolismo , Animais , Ácido Caínico/toxicidade , Masculino , Nervo Frênico/efeitos dos fármacos , Valor Preditivo dos Testes , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Tubercidina/análogos & derivados , Tubercidina/farmacologia
6.
Biomolecules ; 10(12)2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33302551

RESUMO

Tissue-nonspecific alkaline phosphatase (TNAP) is a ubiquitously expressed enzyme that is best known for its role during mineralization processes in bones and skeleton. The enzyme metabolizes phosphate compounds like inorganic pyrophosphate and pyridoxal-5'-phosphate to provide, among others, inorganic phosphate for the mineralization and transportable vitamin B6 molecules. Patients with inherited loss of function mutations in the ALPL gene and consequently altered TNAP activity are suffering from the rare metabolic disease hypophosphatasia (HPP). This systemic disease is mainly characterized by impaired bone and dental mineralization but may also be accompanied by neurological symptoms, like anxiety disorders, seizures, and depression. HPP characteristically affects all ages and shows a wide range of clinical symptoms and disease severity, which results in the classification into different clinical subtypes. This review describes the molecular function of TNAP during the mineralization of bones and teeth, further discusses the current knowledge on the enzyme's role in the nervous system and in sensory perception. An additional focus is set on the molecular role of TNAP in health and on functional observations reported in common laboratory vertebrate disease models, like rodents and zebrafish.


Assuntos
Fosfatase Alcalina/genética , Ansiedade/genética , Osso e Ossos/enzimologia , Depressão/genética , Hipofosfatasia/genética , Convulsões/genética , Dente/enzimologia , Fosfatase Alcalina/deficiência , Animais , Ansiedade/enzimologia , Ansiedade/patologia , Osso e Ossos/patologia , Calcificação Fisiológica/genética , Depressão/enzimologia , Depressão/patologia , Difosfatos/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Humanos , Hipofosfatasia/enzimologia , Hipofosfatasia/patologia , Mutação , Convulsões/enzimologia , Convulsões/patologia , Índice de Gravidade de Doença , Dente/crescimento & desenvolvimento , Vitamina B 6/metabolismo
7.
ASN Neuro ; 12: 1759091420938175, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32664815

RESUMO

Ganglioside GM3 synthase (α-2,3-sialyltransferase, ST3GAL5, GM3S) is a key enzyme involved in the biosynthesis of gangliosides. ST3GAL5 deficiency causes an absence of GM3 and all downstream biosynthetic derivatives. The affected individuals manifest deafness, severe irritability, intractable seizures, and profound intellectual disability. To investigate whether deficiency of GM3 is involved in seizure susceptibility, we induced seizures with different chemoconvulsants in ST3GAL5 knockout mice. We report here that ST3GAL5 knockout mice are hyperactive and more susceptible to seizures induced by chemoconvulsants, including kainate and pilocarpine, compared with normal controls. In the hippocampal dentate gyrus, loss of GM3 aggravates seizure-induced aberrant neurogenesis. These data indicate that GM3 and gangliosides derived from GM3 may serve as important regulators of epilepsy and may play an important role in aberrant neurogenesis associated with seizures.


Assuntos
Pilocarpina/toxicidade , Convulsões/induzido quimicamente , Convulsões/enzimologia , Sialiltransferases/deficiência , Animais , Giro Denteado/efeitos dos fármacos , Giro Denteado/enzimologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Convulsões/genética , Sialiltransferases/genética
8.
Arch Toxicol ; 94(6): 2149-2162, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32303805

RESUMO

Organophosphate (OP) threat agents can trigger seizures that progress to status epilepticus, resulting in persistent neuropathology and cognitive deficits in humans and preclinical models. However, it remains unclear whether patients who do not show overt seizure behavior develop neurological consequences. Therefore, this study compared two subpopulations of rats with a low versus high seizure response to diisopropylfluorophosphate (DFP) to evaluate whether acute OP intoxication causes persistent neuropathology in non-seizing individuals. Adult male Sprague Dawley rats administered DFP (4 mg/kg, sc), atropine sulfate (2 mg/kg, im), and pralidoxime (25 mg/kg, im) were monitored for seizure activity for 4 h post-exposure. Animals were separated into groups with low versus high seizure response based on behavioral criteria and electroencephalogram (EEG) recordings. Cholinesterase activity was evaluated by Ellman assay, and neuropathology was evaluated at 1, 2, 4, and 60 days post-exposure by Fluoro-Jade C (FJC) staining and micro-CT imaging. DFP significantly inhibited cholinesterase activity in the cortex, hippocampus, and amygdala to the same extent in low and high responders. FJC staining revealed significant neurodegeneration in DFP low responders albeit this response was delayed, less persistent, and decreased in magnitude compared to DFP high responders. Micro-CT scans at 60 days revealed extensive mineralization that was not significantly different between low versus high DFP responders. These findings highlight the importance of considering non-seizing patients for medical care in the event of acute OP intoxication. They also suggest that OP intoxication may induce neurological damage via seizure-independent mechanisms, which if identified, might provide insight into novel therapeutic targets.


Assuntos
Ondas Encefálicas/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Inibidores da Colinesterase/toxicidade , Convulsivantes/toxicidade , Isoflurofato/toxicidade , Degeneração Neural , Síndromes Neurotóxicas/etiologia , Convulsões/induzido quimicamente , Acetilcolinesterase/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Encéfalo/enzimologia , Encéfalo/fisiopatologia , Proteínas Ligadas por GPI/metabolismo , Masculino , Síndromes Neurotóxicas/diagnóstico por imagem , Síndromes Neurotóxicas/enzimologia , Síndromes Neurotóxicas/fisiopatologia , Ratos Sprague-Dawley , Convulsões/diagnóstico por imagem , Convulsões/enzimologia , Convulsões/fisiopatologia , Fatores de Tempo , Microtomografia por Raio-X
9.
Int J Mol Sci ; 21(5)2020 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-32121250

RESUMO

Recent evidence suggests that autophagy impairment is implicated in the epileptogenic mechanisms downstream of mTOR hyperactivation. This holds true for a variety of genetic and acquired epileptic syndromes besides malformations of cortical development which are classically known as mTORopathies. Autophagy suppression is sufficient to induce epilepsy in experimental models, while rescuing autophagy prevents epileptogenesis, improves behavioral alterations, and provides neuroprotection in seizure-induced neuronal damage. The implication of autophagy in epileptogenesis and maturation phenomena related to seizure activity is supported by evidence indicating that autophagy is involved in the molecular mechanisms which are implicated in epilepsy. In general, mTOR-dependent autophagy regulates the proliferation and migration of inter-/neuronal cortical progenitors, synapse development, vesicular release, synaptic plasticity, and importantly, synaptic clustering of GABAA receptors and subsequent excitatory/inhibitory balance in the brain. Similar to autophagy, the ubiquitin-proteasome system is regulated downstream of mTOR, and it is implicated in epileptogenesis. Thus, mTOR-dependent cell-clearing systems are now taking center stage in the field of epilepsy. In the present review, we discuss such evidence in a variety of seizure-related disorders and models. This is expected to provide a deeper insight into the molecular mechanisms underlying seizure activity.


Assuntos
Convulsões/enzimologia , Convulsões/patologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia , Modelos Animais de Doenças , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo
10.
Acta Neuropathol Commun ; 8(1): 6, 2020 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-32000863

RESUMO

Human WWOX gene resides in the chromosomal common fragile site FRA16D and encodes a tumor suppressor WW domain-containing oxidoreductase. Loss-of-function mutations in both alleles of WWOX gene lead to autosomal recessive abnormalities in pediatric patients from consanguineous families, including microcephaly, cerebellar ataxia with epilepsy, mental retardation, retinal degeneration, developmental delay and early death. Here, we report that targeted disruption of Wwox gene in mice causes neurodevelopmental disorders, encompassing abnormal neuronal differentiation and migration in the brain. Cerebral malformations, such as microcephaly and incomplete separation of the hemispheres by a partial interhemispheric fissure, neuronal disorganization and heterotopia, and defective cerebellar midline fusion are observed in Wwox-/- mice. Degenerative alterations including severe hypomyelination in the central nervous system, optic nerve atrophy, Purkinje cell loss and granular cell apoptosis in the cerebellum, and peripheral nerve demyelination due to Schwann cell apoptosis correspond to reduced amplitudes and a latency prolongation of transcranial motor evoked potentials, motor deficits and gait ataxia in Wwox-/- mice. Wwox gene ablation leads to the occurrence of spontaneous epilepsy and increased susceptibility to pilocarpine- and pentylenetetrazol (PTZ)-induced seizures in preweaning mice. We determined that a significantly increased activation of glycogen synthase kinase 3ß (GSK3ß) occurs in Wwox-/- mouse cerebral cortex, hippocampus and cerebellum. Inhibition of GSK3ß by lithium ion significantly abolishes the onset of PTZ-induced seizure in Wwox-/- mice. Together, our findings reveal that the neurodevelopmental and neurodegenerative deficits in Wwox knockout mice strikingly recapitulate the key features of human neuropathies, and that targeting GSK3ß with lithium ion ameliorates epilepsy.


Assuntos
Encéfalo/enzimologia , Encéfalo/patologia , Epilepsia/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Transtornos do Neurodesenvolvimento/genética , Convulsões/genética , Oxidorredutase com Domínios WW/genética , Animais , Movimento Celular , Epilepsia/enzimologia , Camundongos Knockout , Transtornos do Neurodesenvolvimento/enzimologia , Neurônios/patologia , Nervos Periféricos/ultraestrutura , Tratos Piramidais/fisiopatologia , Células de Schwann/patologia , Convulsões/enzimologia
11.
Amino Acids ; 52(2): 129-139, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31197571

RESUMO

Several studies have demonstrated high polyamine levels in brain diseases such as epilepsy. Epilepsy is the fourth most common neurological disorder and affects people of all ages. Excitotoxic stress has been associated with epilepsy and it is considered one of the main causes of neuronal degeneration and death. The transgenic mouse line Dach-SMOX, with CD1 background, specifically overexpressing spermine oxidase in brain cortex, has been proven to be highly susceptible to epileptic seizures and excitotoxic stress induced by kainic acid. In this study, we analysed the effect of spermine oxidase over-expression in a different epileptic model, pentylenetetrazole. Behavioural evaluations of transgenic mice compared to controls showed a higher susceptibility towards pentylentetrazole. High-performance liquid chromatography analysis of transgenic brain from treated mice revealed altered polyamine content. Immunoistochemical analysis indicated a rise of 8-oxo-7,8-dihydro-2'-deoxyguanosine, demonstrating an increase in oxidative damage, and an augmentation of system xc- as a defence mechanism. This cascade of events can be initially linked to an increase in protein kinase C alpha, as shown by Western blot. This research points out the role of spermine oxidase, as a hydrogen peroxide producer, in the oxidative stress during epilepsy. Moreover, Dach-SMOX susceptibility demonstrated by two different epileptic models strongly indicates this transgenic mouse line as a potential animal model to study epilepsy.


Assuntos
Córtex Cerebral/enzimologia , Estresse Oxidativo , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Convulsões/enzimologia , Animais , Comportamento Animal , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Poliaminas/metabolismo , Convulsões/genética , Convulsões/metabolismo , Convulsões/psicologia , Poliamina Oxidase
13.
Epilepsy Behav ; 102: 106632, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31747631

RESUMO

The aim of the present study was to investigate the effect of rutin administration (100 mg/kg/day) to pentylenetetrazol (PTZ)-treated Balb-c mice (60 mg/kg/day), with respect to anxiety-like behavior using both open-field and elevated plus-maze (EPM) tests, and acetylcholinesterase (AChE) activity in salt-soluble (SS) fraction and detergent-soluble (DS) fraction of the cerebral cortex, hippocampus, striatum, midbrain, and diencephalon. Our results demonstrated that the administration of PTZ in 3 doses and the induction of seizures increased significantly anxiety behavior of mice and reduced significantly DS-AChE activity in all brain regions examined, while the reduction in the SS fraction was brain region-specific. Rutin administration to normal mice did not affect their behavior, while it induced a brain region-specific reduction in SS-AChE and a significant decrease in DS-AChE in all brain regions. We demonstrated for the first time that pretreatment of PTZ-mice with rutin (PTZ + Rutin group) prevented the manifestation of anxiety and induced interestingly a further significant reduction on the SS- and DS-AChE activities only in the cerebral cortex and striatum, in comparison with PTZ group. Our results show that rutin exhibits an important anxiolytic effect and an anticholinesterase activity in specific brain areas in the seizure model of PTZ.


Assuntos
Acetilcolinesterase/metabolismo , Ansiedade/tratamento farmacológico , Ansiedade/enzimologia , Encéfalo/enzimologia , Pentilenotetrazol/toxicidade , Rutina/uso terapêutico , Convulsões/tratamento farmacológico , Convulsões/enzimologia , Animais , Encéfalo/efeitos dos fármacos , Isoenzimas/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Rutina/farmacologia , Convulsões/induzido quimicamente , Resultado do Tratamento
14.
Ann Neurol ; 86(6): 939-950, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31525273

RESUMO

OBJECTIVE: Temporal lobe epilepsy (TLE) is a devastating disease in which seizures persist in 35% of patients despite optimal use of antiseizure drugs. Clinical and preclinical evidence implicates seizures themselves as one factor promoting epilepsy progression. What is the molecular consequence of a seizure that promotes progression? Evidence from preclinical studies led us to hypothesize that activation of tropomyosin kinase B (TrkB)-phospholipase-C-gamma-1 (PLCγ1) signaling induced by a seizure promotes epileptogenesis. METHODS: To examine the effects of inhibiting TrkB signaling on epileptogenesis following an isolated seizure, we implemented a modified kindling model in which we induced a seizure through amygdala stimulation and then used either a chemical-genetic strategy or pharmacologic methods to disrupt signaling for 2 days following the seizure. The severity of a subsequent seizure was assessed by behavioral and electrographic measures. RESULTS: Transient inhibition of TrkB-PLCγ1 signaling initiated after an isolated seizure limited progression of epileptogenesis, evidenced by the reduced severity and duration of subsequent seizures. Unexpectedly, transient inhibition of TrkB-PLCγ1 signaling initiated following a seizure also reverted a subset of animals to an earlier state of epileptogenesis. Remarkably, inhibition of TrkB-PLCγ1 signaling in the absence of a recent seizure did not reduce severity of subsequent seizures. INTERPRETATION: These results suggest a novel strategy for limiting progression or potentially ameliorating severity of TLE whereby transient inhibition of TrkB-PLCγ1 signaling is initiated following a seizure. ANN NEUROL 2019;86:939-950.


Assuntos
Excitação Neurológica/fisiologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Quinases/metabolismo , Convulsões/tratamento farmacológico , Convulsões/enzimologia , Transdução de Sinais/fisiologia , Animais , Eletroencefalografia/efeitos dos fármacos , Eletroencefalografia/métodos , Feminino , Excitação Neurológica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos
15.
Mol Neurobiol ; 56(12): 8392-8407, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31243719

RESUMO

Pharmacoresistance is a major clinical challenge for approximately 30% of patients with epilepsy. Previous studies indicate nuclear receptors (NRs), drug efflux transporters, and cytochrome P450 enzymes (CYPs) control drug passage across the blood-brain barrier (BBB) in drug-resistant epilepsy. Here, we (1) evaluate BBB changes, neurovascular nuclear receptors, and drug transporters in lesional/epileptic (EPI) and non-lesional/non-epileptic (NON-EPI) regions of the same brain, (2) examine regional CYP expression and activity, and (3) investigate the association among CYP brain expression, seizure frequency, duration of epilepsy, and antiepileptic drug (AED) combination. We used surgically resected brain specimens from patients with medically intractable epilepsy (n = 22) where the epileptogenic loci were well-characterized by invasive and non-invasive methods; histology confirmed distinction of small NON-EPI regions from EPI tissues. NRs, transporters, CYPs, and tight-junction proteins were assessed by western blots/immunohistochemistry, and CYP metabolic activity was determined and compared. The relationship of CYP expression with seizure frequency, duration of epilepsy, and prescribed AEDs was evaluated. Decreased BBB tight-junction proteins accompanied IgG leakage in EPI regions and correlated with upregulated NR and efflux transporter levels. CYP expression and activity significantly increased in EPI compared to NON-EPI tissues. Change in EPI and NON-EPI CYP3A4 expression increased in patients taking AEDs that were CYP substrates, was downregulated when CYP- and non-CYP-substrate AEDs were given together, and correlated with seizure frequency. Our studies suggest focal neurovascular CYP-NR-transporter alterations, as demonstrated by the relationship of seizure frequency and AED combination to brain CYP3A4, might together impact biotransformation machinery of human pharmacoresistant epilepsy.


Assuntos
Anticonvulsivantes/uso terapêutico , Encéfalo/enzimologia , Epilepsia/tratamento farmacológico , Epilepsia/enzimologia , Convulsões/tratamento farmacológico , Convulsões/enzimologia , Adolescente , Adulto , Idoso , Anticonvulsivantes/farmacologia , Biotransformação , Barreira Hematoencefálica/patologia , Encéfalo/patologia , Criança , Pré-Escolar , Citocromo P-450 CYP3A/metabolismo , Feminino , Humanos , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Pessoa de Meia-Idade , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas de Junções Íntimas/metabolismo , Adulto Jovem
16.
Ann Neurol ; 85(6): 907-920, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30937971

RESUMO

OBJECTIVE: Despite decades of epilepsy research, 30% of focal epilepsies remain resistant to antiseizure drugs, with effective drug development impeded by lack of understanding on how seizures are initiated. Here, we report the mechanism of seizure onset relevant to most seizures that are characteristic of focal epilepsies. METHODS: Electric and metabolic network parameters were measured using several seizure models in mouse hippocampal slices and acutely induced seizures in rats in vivo to determine metabolic events occurring at seizure onset. RESULTS: We show that seizure onset is associated with a rapid release of H2 O2 resulting from N-methyl-D-aspartate (NMDA) receptor-mediated activation of nicotinamide adenine dinucleotide phosphate oxidase (NOX). NOX blockade prevented the fast H2 O2 release as well as the direct current shift and seizurelike event induction in slices. Similarly, intracerebroventricular injection of NOX antagonists prevented acutely induced seizures in rats. INTERPRETATION: Our results show that seizures are initiated by NMDA receptor-mediated NOX-induced oxidative stress and can be arrested by NOX inhibition. We introduce a novel use for blood-brain barrier-permeable NOX inhibitor with a significant potential to become the first seizure-specific medication. Thus, targeting NOX may provide a breakthrough treatment for focal epilepsies. ANN NEUROL 2019;85:907-920.


Assuntos
Modelos Animais de Doenças , NADPH Oxidases/metabolismo , Convulsões/enzimologia , Convulsões/fisiopatologia , Animais , Ativação Enzimática/fisiologia , Hipocampo/enzimologia , Hipocampo/fisiopatologia , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/metabolismo
17.
Oxid Med Cell Longev ; 2019: 1327986, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31019649

RESUMO

Epilepsy is a neurological disorder characterized by recurrent spontaneous seizures due to an imbalance between cerebral excitability and inhibition, with a tendency towards uncontrolled excitability. Epilepsy has been associated with oxidative and nitrosative stress due to prolonged neuronal hyperexcitation and loss neurons during seizures. The experimental animal models report level of ATP diminished and increase in lipid peroxidation, catalase, and glutathione altered activity in the brain. We studied the immunohistochemical expression and localization of antioxidant enzymes GPx, SOD, and CAT in the rat brains treated with KA and PTZ. A significant decrease was observed in the number of immunoreactive cells to GPx, without significant changes for SOD and CAT in KA-treated rats, and decrease in the number of immunoreactive cells to SOD, without significant changes for GPx and only CAT in PTZ-treated rats. Evident immunoreactivity of GPx, SOD, and CAT was observed mainly in astrocytes and neurons of the hippocampal brain region in rats exposed at KA; similar results were observed in rats treated with PTZ at the first hours. These results provide evidence supporting the role of activation of the Nrf2 antioxidant system pathway against oxidative stress effects in the experimental models of epileptic seizures.


Assuntos
Imuno-Histoquímica/métodos , Fator 2 Relacionado a NF-E2/metabolismo , Convulsões/enzimologia , Convulsões/patologia , Animais , Antioxidantes/metabolismo , Comportamento Animal , Hipocampo/patologia , Ácido Caínico , Masculino , Modelos Biológicos , Pentilenotetrazol , Ratos Wistar
18.
Neurotoxicology ; 73: 81-84, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30853371

RESUMO

Acute intoxication with organophosphate cholinesterase inhibitors (OPs) is a significant human health threat, and current medical countermeasures for OP poisoning are of limited therapeutic efficacy. The rat model of acute intoxication with diisopropylfluorophosphate (DFP) is increasingly being used to test candidate compounds for efficacy in protecting against the immediate and long-term consequences of acute OP toxicity. In this model, rats are typically pretreated with pyridostigmine bromide (PB), a reversible cholinesterase inhibitor, to enhance survival. However, PB pretreatment is not likely in most scenarios of civilian exposure to acutely neurotoxic levels of OPs. Therefore, the goal of this study was to determine whether PB pretreatment significantly increases survival in DFP-intoxicated rats. Adult male Sprague Dawley rats were injected with DFP (4 mg/kg, s.c.) or vehicle (VEH) followed 1 min later by combined i.m. injection of atropine sulfate (2 mg/kg) and 2-pralidoxime (25 mg/kg). Animals were pretreated 30 min prior to these injections with PB (0.1 mg/kg, i.m.) or an equal volume of saline. DFP triggered rapid and sustained seizure behavior irrespective of PB pretreatment, and there was no significant difference in average seizure behavior score during the first 4 h following injection between DFP animals pretreated with PB or not. PB pretreatment also had no significant effect on survival or brain AChE activity at 24 h post-DFP exposure. In summary, PB pretreatment is not necessary to ensure survival of rats acutely intoxicated with DFP, and eliminating PB pretreatment in the rat model of acute DFP intoxication would increase its relevance to acute OP intoxication in civilians.


Assuntos
Encéfalo/efeitos dos fármacos , Inibidores da Colinesterase/farmacologia , Isoflurofato , Síndromes Neurotóxicas/etiologia , Intoxicação por Organofosfatos/etiologia , Brometo de Piridostigmina/farmacologia , Convulsões/etiologia , Acetilcolinesterase/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Proteínas Ligadas por GPI/metabolismo , Masculino , Síndromes Neurotóxicas/enzimologia , Síndromes Neurotóxicas/fisiopatologia , Intoxicação por Organofosfatos/enzimologia , Intoxicação por Organofosfatos/fisiopatologia , Ratos Sprague-Dawley , Convulsões/enzimologia , Convulsões/fisiopatologia , Fatores de Tempo
19.
Epilepsia ; 60(4): 626-635, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30770561

RESUMO

OBJECTIVE: Viral encephalitis increases the risk for developing seizures and epilepsy. Indoleamine 2,3-dioxygenase 1 (Ido1) is induced by inflammatory cytokines and functions to metabolize tryptophan to kynurenine. Kynurenine can be further metabolized to produce kynurenic acid and the N-methyl-d-aspartate receptor agonist quinolinic acid (QuinA). In the present study, we sought to determine the role of Ido1 in promoting seizures in an animal model of viral encephalitis. METHODS: C57BL/6J and Ido1 knockout mice (Ido1-KO) were infected with Theiler's murine encephalomyelitis virus (TMEV). Quantitative real-time polymerase chain reaction was used to evaluate hippocampal expression of proinflammatory cytokines, Ido1, and viral RNA. Body weights and seizure scores were recorded daily. Elevated zero maze was used to assess differences in behavior, and hippocampal pathology was determined by immunohistochemistry. RESULTS: Infected C57BL/6J mice up-regulated proinflammatory cytokines, Ido1, and genes encoding the enzymatic cascade responsible for QuinA production in the kynurenine pathway prior to the onset of seizures. Seizure incidence was elevated in Ido1-KO compared to C57BL/6J mice. Infection increased locomotor activity in Ido1-KO compared to C57BL/6J mice. Furthermore, the occurrence of seizures was associated with hyperexcitability. Neither expression of proinflammatory cytokines nor viral RNA was altered as a result of genotype. Immunohistochemical analysis revealed increased hippocampal pathology in Ido1-KO mice. SIGNIFICANCE: Our findings suggest that Ido1 deletion promotes seizures and neuropathogenesis during acute TMEV encephalitis.


Assuntos
Encefalite Viral/complicações , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Convulsões/enzimologia , Animais , Infecções por Cardiovirus/complicações , Modelos Animais de Doenças , Encefalite Viral/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Convulsões/virologia , Theilovirus
20.
Am J Hum Genet ; 104(2): 287-298, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661771

RESUMO

Hypusine is formed post-translationally from lysine and is found in a single cellular protein, eukaryotic translation initiation factor-5A (eIF5A), and its homolog eIF5A2. Biosynthesis of hypusine is a two-step reaction involving the enzymes deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH). eIF5A is highly conserved throughout eukaryotic evolution and plays a role in mRNA translation, cellular proliferation, cellular differentiation, and inflammation. DHPS is also highly conserved and is essential for life, as Dhps-null mice are embryonic lethal. Using exome sequencing, we identified rare biallelic, recurrent, predicted likely pathogenic variants in DHPS segregating with disease in five affected individuals from four unrelated families. These individuals have similar neurodevelopmental features that include global developmental delay and seizures. Two of four affected females have short stature. All five affected individuals share a recurrent missense variant (c.518A>G [p.Asn173Ser]) in trans with a likely gene disrupting variant (c.1014+1G>A, c.912_917delTTACAT [p.Tyr305_Ile306del], or c.1A>G [p.Met1?]). cDNA studies demonstrated that the c.1014+1G>A variant causes aberrant splicing. Recombinant DHPS enzyme harboring either the p.Asn173Ser or p.Tyr305_Ile306del variant showed reduced (20%) or absent in vitro activity, respectively. We co-transfected constructs overexpressing HA-tagged DHPS (wild-type or mutant) and GFP-tagged eIF5A into HEK293T cells to determine the effect of these variants on hypusine biosynthesis and observed that the p.Tyr305_Ile306del and p.Asn173Ser variants resulted in reduced hypusination of eIF5A compared to wild-type DHPS enzyme. Our data suggest that rare biallelic variants in DHPS result in reduced enzyme activity that limits the hypusination of eIF5A and are associated with a neurodevelopmental disorder.


Assuntos
Genes Recessivos/genética , Lisina/análogos & derivados , Mutação , Transtornos do Neurodesenvolvimento/enzimologia , Transtornos do Neurodesenvolvimento/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Alelos , Sequência de Aminoácidos , Criança , Pré-Escolar , Deficiências do Desenvolvimento/enzimologia , Deficiências do Desenvolvimento/genética , Feminino , Haplótipos , Humanos , Lisina/biossíntese , Masculino , Erros Inatos do Metabolismo/enzimologia , Erros Inatos do Metabolismo/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/química , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Linhagem , Fatores de Iniciação de Peptídeos/química , Fatores de Iniciação de Peptídeos/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Convulsões/enzimologia , Convulsões/genética , Adulto Jovem , Fator de Iniciação de Tradução Eucariótico 5A
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA