Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 185: 106251, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37536383

RESUMO

The latency between traumatic brain injury (TBI) and the onset of epilepsy (PTE) represents an opportunity for counteracting epileptogenesis. Antiepileptogenesis trials are hampered by the lack of sensitive biomarkers that allow to enrich patient's population at-risk for PTE. We aimed to assess whether specific ECoG signals predict PTE in a clinically relevant mouse model with ∼60% epilepsy incidence. TBI was provoked in adult CD1 male mice by controlled cortical impact on the left parieto-temporal cortex, then mice were implanted with two perilesional cortical screw electrodes and two similar electrodes in the hemisphere contralateral to the lesion site. Acute seizures and spikes/sharp waves were ECoG-recorded during 1 week post-TBI. These early ECoG events were analyzed according to PTE incidence as assessed by measuring spontaneous recurrent seizures (SRS) at 5 months post-TBI. We found that incidence, number and duration of acute seizures during 3 days post-TBI were similar in PTE mice and mice not developing epilepsy (No SRS mice). Control mice with cortical electrodes (naïve, n = 5) or with electrodes and craniotomy (sham, n = 5) exhibited acute seizures but did not develop epilepsy. The daily number of spikes/sharp waves at the perilesional electrodes was increased similarly in PTE (n = 15) and No SRS (n = 8) mice vs controls (p < 0.05, n = 10) from day 2 post-injury. Differently, the daily number of spikes/sharp waves at both contralateral electrodes showed a progressive increase in PTE mice vs No SRS and control mice. In particular, spikes number was higher in PTE vs No SRS mice (p < 0.05) at 6 and 7 days post-TBI, and this measure predicted epilepsy development with high accuracy (AUC = 0.77, p = 0.03; CI 0.5830-0.9670). The cut-off value was validated in an independent cohort of TBI mice (n = 12). The daily spike number at the contralateral electrodes showed a circadian distribution in PTE mice which was not observed in No SRS mice. Analysis of non-linear dynamics at each electrode site showed changes in dimensionality during 4 days post-TBI. This measure yielded the best discrimination between PTE and No SRS mice (p < 0.01) at the cortical electrodes contralateral to injury. Data show that epileptiform activity contralateral to the lesion site has the the highest predictive value for PTE in this model reinforcing the hypothesis that the hemisphere contralateral to the lesion core may drive epileptogenic networks after TBI.


Assuntos
Lesões Encefálicas Traumáticas , Epilepsia Pós-Traumática , Epilepsia , Masculino , Camundongos , Animais , Epilepsia Pós-Traumática/complicações , Lesões Encefálicas Traumáticas/complicações , Convulsões/complicações , Epilepsia/etiologia , Eletrocorticografia
2.
Neurobiol Dis ; 173: 105835, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35932989

RESUMO

Therapies for epilepsy mainly provide symptomatic control of seizures since most of the available drugs do not target disease mechanisms. Moreover, about one-third of patients fail to achieve seizure control. To address the clinical need for disease-modifying therapies, research should focus on targets which permit interventions finely balanced between optimal efficacy and safety. One potential candidate is the brain-specific enzyme cholesterol 24-hydroxylase. This enzyme converts cholesterol to 24S-hydroxycholesterol, a metabolite which among its biological roles modulates neuronal functions relevant for hyperexcitability underlying seizures. To study the role of cholesterol 24-hydroxylase in epileptogenesis, we administered soticlestat (TAK-935/OV935), a potent and selective brain-penetrant inhibitor of the enzyme, during the early disease phase in a mouse model of acquired epilepsy using a clinically relevant dose. During soticlestat treatment, the onset of epilepsy was delayed and the number of ensuing seizures was decreased by about 3-fold compared to vehicle-treated mice, as assessed by EEG monitoring. Notably, the therapeutic effect was maintained 6.5 weeks after drug wash-out when seizure number was reduced by about 4-fold and their duration by 2-fold. Soticlestat-treated mice showed neuroprotection of hippocampal CA1 neurons and hilar mossy cells as assessed by post-mortem brain histology. High throughput RNA-sequencing of hippocampal neurons and glia in mice treated with soticlestat during epileptogenesis showed that inhibition of cholesterol 24-hydroxylase did not directly affect the epileptogenic transcriptional network, but rather modulated a non-overlapping set of genes that might oppose the pathogenic mechanisms of the disease. In human temporal lobe epileptic foci, we determined that cholesterol 24-hydroxylase expression trends higher in neurons, similarly to epileptic mice, while the enzyme is ectopically induced in astrocytes compared to control specimens. Soticlestat reduced significantly the number of spontaneous seizures in chronic epileptic mice when was administered during established epilepsy. Data show that cholesterol 24-hydroxylase contributes to spontaneous seizures and is involved in disease progression, thus it represents a novel target for chronic seizures inhibition and disease-modification therapy in epilepsy.


Assuntos
Epilepsia do Lobo Temporal , Epilepsia , Animais , Colesterol/metabolismo , Colesterol 24-Hidroxilase/metabolismo , Modelos Animais de Doenças , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Hipocampo/metabolismo , Humanos , Camundongos , Piperidinas , Piridinas , RNA/metabolismo , Convulsões/metabolismo
3.
Brain ; 142(7): e39, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31145451

RESUMO

Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.


Assuntos
Acetilcisteína/farmacologia , Epilepsia/prevenção & controle , Glutationa/metabolismo , Isotiocianatos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Biomarcadores/metabolismo , Estudos de Casos e Controles , Contagem de Células , Disfunção Cognitiva/complicações , Disfunção Cognitiva/prevenção & controle , Modelos Animais de Doenças , Estimulação Elétrica , Epilepsia/complicações , Proteína HMGB1/sangue , Hipocampo/metabolismo , Humanos , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos , Estado Epiléptico/complicações , Estado Epiléptico/metabolismo , Estado Epiléptico/prevenção & controle , Sulfóxidos
4.
Neurobiol Dis ; 124: 373-378, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30590177

RESUMO

The lack of early biomarkers of epileptogenesis precludes a sound prediction of epilepsy development after acute brain injuries and of the natural course of the disease thus impairing the development of antiepileptogenic treatments. We investigated whether the dimensional changes of nonlinear dynamics in EEG/ECoG signals, that were recorded in the early aftermath of different epileptogenic injuries, provide a measure to be exploited as a sensitive prognostic and predictive biomarker for epilepsy. Using three different models of epilepsy in two rodent species, we report a common and significant decrease of nonlinear dynamics dimension in EEG/ECoG tracings during early epileptogenesis. In particular, the magnitude of this dimensional decrease predicts the severity of ensuing epilepsy, and this measure is modulated by disease-modifying or antiepileptogenic treatments. The broad application of EEG/ECoG monitoring in epilepsy underlines the translational value of these findings for enriching the population of patients at risk for developing epilepsy in clinical investigations.


Assuntos
Lesões Encefálicas/fisiopatologia , Eletrocorticografia/métodos , Eletroencefalografia/métodos , Epilepsia/fisiopatologia , Animais , Lesões Encefálicas/complicações , Epilepsia/etiologia , Camundongos , Dinâmica não Linear , Ratos , Processamento de Sinais Assistido por Computador
5.
Epilepsia ; 59(1): 79-91, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29171003

RESUMO

OBJECTIVE: Status epilepticus (SE) is a life-threatening and commonly drug-refractory condition. Novel therapies are needed to rapidly terminate seizures to prevent mortality and morbidity. Monoacylglycerol lipase (MAGL) is the key enzyme responsible for the hydrolysis of the endocannabinoid 2-arachidonoylglycerol (2-AG) and a major contributor to the brain pool of arachidonic acid (AA). Inhibiting of monoacylglycerol lipase modulates synaptic activity and neuroinflammation, 2 mediators of excessive neuronal activation underlying seizures. We studied the effect of a potent and selective irreversible MAGL inhibitor, CPD-4645, on SE that was refractory to diazepam, its neuropathologic sequelae, and the mechanism underlying the drug's effects. METHODS: Diazepam-resistant SE was induced in adult mice fed with standard or ketogenic diet or in cannabinoid receptor type 1 (CB1) receptor knock-out mice. CPD-4645 (10 mg/kg, subcutaneously) or vehicle was dosed 1 and 7 h after status epilepticus onset in video-electroencephalography (EEG) recorded mice. At the end of SE, mice were examined in the novel object recognition test followed by neuronal cellloss analysis. RESULTS: CPD-4645 maximal plasma and brain concentrations were attained 0.5 h postinjection (half-life = 3.7 h) and elevated brain 2-AG levels by approximately 4-fold. CPD-4645 administered to standard diet-fed mice progressively reduced spike frequency during 3 h postinjection, thereby shortening SE duration by 47%. The drug immediately abrogated SE in ketogenic diet-fed mice. CPD-4645 rescued neuronal cell loss and cognitive deficit and reduced interleukin (IL)-1ß and cyclooxygenase 2 (COX-2) brain expression resulting from SE. The CPD-4645 effect on SE was similar in mice lacking CB1 receptors. SIGNIFICANCE: MAGL represents a novel therapeutic target for treating status epilepticus and improving its sequelae. CPD-4645 therapeutic effects appear to be predominantly mediated by modulation of neuroinflammation.


Assuntos
Carbamatos/uso terapêutico , Monoacilglicerol Lipases/antagonistas & inibidores , Piperidinas/uso terapêutico , Estado Epiléptico , Sulfonamidas/uso terapêutico , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Ondas Encefálicas/efeitos dos fármacos , Ondas Encefálicas/fisiologia , Carbamatos/química , Carbamatos/farmacocinética , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Diazepam/efeitos adversos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epilepsia Resistente a Medicamentos/induzido quimicamente , Epilepsia Resistente a Medicamentos/enzimologia , Epilepsia Resistente a Medicamentos/terapia , Eletroencefalografia , Agonistas de Aminoácidos Excitatórios/toxicidade , Fluoresceínas/metabolismo , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monoacilglicerol Lipases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Piperidinas/química , Piperidinas/farmacologia , Distribuição Aleatória , Receptor CB1 de Canabinoide/deficiência , Receptor CB1 de Canabinoide/genética , Reconhecimento Psicológico/efeitos dos fármacos , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/complicações , Estado Epiléptico/enzimologia , Estado Epiléptico/terapia , Sulfonamidas/química , Sulfonamidas/farmacocinética , Fatores de Tempo
6.
Brain ; 140(7): 1885-1899, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28575153

RESUMO

Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of disulfide high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented disulfide HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.


Assuntos
Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Epilepsia/tratamento farmacológico , Domínios HMG-Box/efeitos dos fármacos , Proteína HMGB1/sangue , Proteína HMGB1/metabolismo , Isotiocianatos/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Biomarcadores/sangue , Biomarcadores/metabolismo , Disfunção Cognitiva/complicações , Disfunção Cognitiva/tratamento farmacológico , Modelos Animais de Doenças , Quimioterapia Combinada , Epilepsia/metabolismo , Proteína HMGB1/biossíntese , Hipocampo/metabolismo , Isotiocianatos/farmacologia , Masculino , Degeneração Neural/dietoterapia , Neurônios/metabolismo , Ratos , Sulfóxidos
7.
J Neurosci ; 37(17): 4450-4461, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28330876

RESUMO

Postinjury epilepsy (PIE) is a devastating sequela of various brain insults. While recent studies offer novel insights into the mechanisms underlying epileptogenesis and discover potential preventive treatments, the lack of PIE biomarkers hinders the clinical implementation of such treatments. Here we explored the biomarker potential of different electrographic features in five models of PIE. Electrocorticographic or intrahippocampal recordings of epileptogenesis (from the insult to the first spontaneous seizure) from two laboratories were analyzed in three mouse and two rat PIE models. Time, frequency, and fractal and nonlinear properties of the signals were examined, in addition to the daily rate of epileptiform spikes, the relative power of five frequency bands (theta, alpha, beta, low gamma, and high gamma) and the dynamics of these features over time. During the latent pre-seizure period, epileptiform spikes were more frequent in epileptic compared with nonepileptic rodents; however, this feature showed limited predictive power due to high inter- and intra-animal variability. While nondynamic rhythmic representation failed to predict epilepsy, the dynamics of the theta band were found to predict PIE with a sensitivity and specificity of >90%. Moreover, theta dynamics were found to be inversely correlated with the latency period (and thus predict the onset of seizures) and with the power change of the high-gamma rhythm. In addition, changes in theta band power during epileptogenesis were associated with altered locomotor activity and distorted circadian rhythm. These results suggest that changes in theta band during the epileptogenic period may serve as a diagnostic biomarker for epileptogenesis, able to predict the future onset of spontaneous seizures.SIGNIFICANCE STATEMENT Postinjury epilepsy is an unpreventable and devastating disorder that develops following brain injuries, such as traumatic brain injury and stroke, and is often associated with neuropsychiatric comorbidities. As PIE affects as many as 20% of brain-injured patients, reliable biomarkers are imperative before any preclinical therapeutics can find clinical translation. We demonstrate the capacity to predict the epileptic outcome in five different models of PIE, highlighting theta rhythm dynamics as a promising biomarker for epilepsy. Our findings prompt the exploration of theta dynamics (using repeated electroencephalographic recordings) as an epilepsy biomarker in brain injury patients.


Assuntos
Biomarcadores , Eletrocorticografia , Epilepsia/fisiopatologia , Animais , Lesões Encefálicas/complicações , Ritmo Circadiano , Convulsivantes/administração & dosagem , Epilepsia/induzido quimicamente , Hipocampo/fisiopatologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Atividade Motora , Ratos , Convulsões/fisiopatologia
9.
Sci Rep ; 6: 31129, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27488140

RESUMO

The lack of a marker of epileptogenesis is an unmet medical need, not only from the clinical perspective but also from the point of view of the pre-clinical research. Indeed, the lack of this kind of marker affects the investigations on the mechanisms of epileptogenesis as well as the development of novel therapeutic approaches aimed to prevent or to mitigate the severity of the incoming epilepsy in humans. In this work, we provide evidence that in an experimental model of epileptogenesis that mimics the alteration of the blood-brain barrier permeability, a key-mechanism that contributes to the development of epilepsy in humans and in animals, the prolonged occurrence in the electrocorticograms (ECoG) of high rates of a nonlinear dynamical regimes known as intermittency univocally characterizes the population of experimental animals which develop epilepsy, hence it can be considered as the first biophysical marker of epileptogenesis.


Assuntos
Epilepsia/patologia , Animais , Biomarcadores/metabolismo , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Eletrocorticografia/métodos , Epilepsia/metabolismo , Camundongos , Dinâmica não Linear , Permeabilidade
10.
Neurobiol Dis ; 93: 146-55, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27173096

RESUMO

One major unmet clinical need in epilepsy is the identification of therapies to prevent or arrest epilepsy development in patients exposed to a potential epileptogenic insult. The development of such treatments has been hampered by the lack of non-invasive biomarkers that could be used to identify the patients at-risk, thereby allowing to design affordable clinical studies. Our goal was to test the predictive value of cognitive deficits and brain astrocyte activation for the development of epilepsy following a potential epileptogenic injury. We used a model of epilepsy induced by pilocarpine-evoked status epilepticus (SE) in 21-day old rats where 60-70% of animals develop spontaneous seizures after around 70days, although SE is similar in all rats. Learning was evaluated in the Morris water-maze at days 15 and 65 post-SE, each time followed by proton magnetic resonance spectroscopy for measuring hippocampal myo-Inositol levels, a marker of astrocyte activation. Rats were video-EEG monitored for two weeks at seven months post-SE to detect spontaneous seizures, then brain histology was done. Behavioral and imaging data were retrospectively analysed in epileptic rats and compared with non-epileptic and control animals. Rats displayed spatial learning deficits within three weeks from SE. However, only epilepsy-prone rats showed accelerated forgetting and reduced learning rate compared to both rats not developing epilepsy and controls. These deficits were associated with reduced hippocampal neurogenesis. myo-Inositol levels increased transiently in the hippocampus of SE-rats not developing epilepsy while this increase persisted until spontaneous seizures onset in epilepsy-prone rats, being associated with a local increase in S100ß-positive astrocytes. Neuronal cell loss was similar in all SE-rats. Our data show that behavioral deficits, together with a non-invasive marker of astrocyte activation, predict which rats develop epilepsy after an acute injury. These measures have potential clinical relevance for identifying individuals at-risk for developing epilepsy following exposure to epileptogenic insults, and consequently, for designing adequately powered antiepileptogenesis trials.


Assuntos
Encéfalo/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Estado Epiléptico/fisiopatologia , Animais , Astrócitos/metabolismo , Comportamento Animal/fisiologia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Eletroencefalografia/métodos , Masculino , Neurogênese/fisiologia , Neurônios/metabolismo , Ratos Sprague-Dawley , Estado Epiléptico/complicações
11.
Neurobiol Dis ; 58: 102-14, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23523633

RESUMO

Toll-like receptor 4 (TLR4) activation in neuron and astrocytes by High Mobility Group Box 1 (HMGB1) protein is a key mechanism of seizure generation. HMGB1 also activates the Receptor for Advanced Glycation Endproducts (RAGE), but it was unknown whether RAGE activation contributes to seizures or to HMGB1 proictogenic effects. We found that acute EEG seizures induced by 7ng intrahippocampal kainic acid (KA) were significantly reduced in Rage-/- mice relative to wild type (Wt) mice. The proictogenic effect of HMGB1 was decreased in Rage-/- mice, but less so, than in Tlr4-/- mice. In a mouse mesial temporal lobe epilepsy (mTLE) model, status epilepticus induced by 200ng intrahippocampal KA and the onset of the spontaneous epileptic activity were similar in Rage-/-, Tlr4-/- and Wt mice. However, the number of hippocampal paroxysmal episodes and their duration were both decreased in epileptic Rage-/- and Tlr4-/- mice vs Wt mice. All strains of epileptic mice displayed similar cognitive deficits in the novel object recognition test vs the corresponding control mice. CA1 neuronal cell loss was increased in epileptic Rage-/- vs epileptic Wt mice, while granule cell dispersion and doublecortin (DCX)-positive neurons were similarly affected. Notably, DCX neurons were preserved in epileptic Tlr4-/- mice. We did not find compensatory changes in HMGB1-related inflammatory signaling nor in glutamate receptor subunits in Rage-/- and Tlr4-/- naïve mice, except for ~20% NR2B subunit reduction in Rage-/- mice. RAGE was induced in neurons, astrocytes and microvessels in human and experimental mTLE hippocampi. We conclude that RAGE contributes to hyperexcitability underlying acute and chronic seizures, as well as to the proictogenic effects of HMGB1. RAGE and TLR4 play different roles in the neuropathologic sequelae developing after status epilepticus. These findings reveal new molecular mechanisms underlying seizures, cell loss and neurogenesis which involve inflammatory pathways upregulated in human epilepsy.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , Regulação da Expressão Gênica/genética , Receptores Imunológicos/metabolismo , Convulsões/metabolismo , Regulação para Cima/fisiologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Estimulação Elétrica/efeitos adversos , Eletroencefalografia , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/etiologia , Epilepsia do Lobo Temporal/patologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína HMGB1/administração & dosagem , Proteína HMGB1/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/deficiência , Convulsões/induzido quimicamente , Convulsões/etiologia , Receptor 4 Toll-Like/deficiência , Regulação para Cima/genética
12.
Brain ; 131(Pt 6): 1506-15, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18477594

RESUMO

Temporal lobe epilepsy remains amongst the most common and drug refractory of neurological disorders. Gene therapy may provide a realistic therapeutic approach alternative to surgery for intractable focal epilepsies. To test this hypothesis, we applied here a gene therapy approach, using a recombinant adeno-associated viral (rAAV) vector expressing the human neuropeptide Y (NPY) gene, to a progressive and spontaneous seizure model of temporal lobe epilepsy induced by electrical stimulation of the temporal pole of the hippocampus, which replicates many features of the human condition. rAAV-NPY or a control vector lacking the expression cassette (rAAV-Empty) was delivered into the epileptic rat hippocampi at an early progressive stage of the disease. Chronic epileptic rats were video-EEG monitored to establish pre-injection baseline recordings of spontaneous seizures and the effect of rAAV-NPY versus rAAV-Empty vector injection. Both non-injected stimulated controls and rAAV-empty injected rats showed a similar progressive increase of spontaneous seizure frequency consistent with epileptogenesis. The delivery of rAAV-NPY in epileptic rat brain leads to a remarkable decrease in the progression of seizures as compared to both control groups and this effect was correlated with the NPY over-expression in the hippocampus. Moreover, spontaneous seizure frequency was significantly reduced in 40% of treated animals as compared to their pre-injection baseline. Our data show that this gene therapy strategy decreases spontaneous seizures and suppresses their progression in chronic epileptic rats, thus representing a promising new therapeutic strategy.


Assuntos
Epilepsia do Lobo Temporal/terapia , Terapia Genética/métodos , Neuropeptídeo Y/genética , Animais , Doença Crônica , Dependovirus/genética , Eletroencefalografia , Epilepsia do Lobo Temporal/metabolismo , Expressão Gênica , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Hipocampo/química , Hipocampo/metabolismo , Injeções , Masculino , Neurônios/química , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Neuropeptídeo Y/uso terapêutico , Ratos , Ratos Sprague-Dawley , Transdução Genética/métodos , Gravação em Vídeo
13.
Neurobiol Dis ; 24(3): 429-42, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17027274

RESUMO

We examined the blood-brain barrier (BBB) function in methylazoxymethanol acetate (MAM)-treated rats, a model of human developmental brain malformations. We found aberrant vessels morphology and serum albumin leakage in the heterotopic (malformed) hippocampus; these changes were associated with a significant increase in endothelial P-glycoprotein (P-gp) expression. Seizures exacerbated BBB leakage and greatly augmented P-gp expression in vessels and additionally in perivascular/parenchymal astrocytes. The effects of seizures were observed to a much larger extent in malformed than in normal brain tissue. The intrinsic changes in BBB function in MAM-exposed rats were associated with increased blood-to-brain penetration of ondansetron, a P-gp substrate. However, a marked reduction in drug brain levels was provoked by seizures, and this effect was reversed by selective blockade of P-gp activity with tariquidar. Changes in BBB function may critically contribute to determine the brain uptake and distribution of P-gp substrates in epileptic tissue associated with developmental malformations.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Epilepsia/metabolismo , Hipocampo , Convulsões/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Anticonvulsivantes/farmacocinética , Astrócitos/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/anormalidades , Encéfalo/irrigação sanguínea , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Coristoma/induzido quimicamente , Modelos Animais de Doenças , Resistência a Medicamentos/fisiologia , Células Endoteliais/efeitos dos fármacos , Epilepsia/patologia , Feminino , Masculino , Análise por Pareamento , Acetato de Metilazoximetanol , Ondansetron/farmacocinética , Gravidez , RNA Mensageiro/análise , Ratos , Convulsões/patologia , Albumina Sérica/metabolismo
14.
J Neuroimmunol ; 172(1-2): 27-37, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16337691

RESUMO

Erythropoietin (EPO) mediates a wide range of neuroprotective activities, including amelioration of disease and neuroinflammation in rat models of EAE. However, optimum dosing parameters are currently unknown. In the present study, we used a chronic EAE model induced in mice by immunization with the myelin oligodendrocyte glycoprotein peptide (MOG35-55) to compare the effect of EPO given with different treatment schedules. EPO was administered intraperitoneally at 0.5, 5.0 or 50 microg/kg three times weekly starting from day 3 after immunization (preventive schedule), at the onset of clinical disease (therapeutic schedule) or 15 days after the onset of symptoms (late therapeutic schedule). The results show that EPO is effective even when given after the appearance of clinical signs of EAE, but with a reduced efficacy compared to the preventative schedule. To determine whether this effect requires the homodimeric EPO receptor (EPOR2)-mediated hematopoietic effect of EPO, we studied the effect of carbamylated EPO (CEPO) that does not bind EPOR2. CEPO, ameliorated EAE without changing the hemoglobin concentration. Another non-erythropoietic derivative, asialoEPO was also effective. Both EPO and CEPO equivalently decreased the EAE-associated production of TNF-alpha, IL-1beta and IL-1Ra in the spinal cord, and IFN-gamma by peripheral lymphocytes, indicating that their action involves targeting neuroinflammation. The lowest dosage tested appeared fully effective. The possibility to dissociate the anti-neuroinflammatory action of EPO from its hematopoietic action, which may cause undesired side effects in non-anemic patients, present new avenues to the therapy of multiple sclerosis.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/prevenção & controle , Eritropoetina/análogos & derivados , Eritropoetina/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Doença Crônica , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Encefalomielite Autoimune Experimental/sangue , Encefalomielite Autoimune Experimental/induzido quimicamente , Feminino , Glicoproteínas , Hematócrito/métodos , Humanos , Imuno-Histoquímica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos , RNA Mensageiro/metabolismo , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Índice de Gravidade de Doença , Medula Espinal/efeitos dos fármacos , Baço/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
15.
Epilepsia ; 46(10): 1613-9, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16190932

RESUMO

PURPOSE: We measured the brain-to-plasma partition of 10,11-dihydro-10-hydroxy-5H-dibenzo(b,f)azepine-5-carboxamide (10-OHCBZ) in epilepsy patients undergoing surgery to alleviate drug-resistant seizures and administered with different oral doses of oxcarbazepine (OXC). We addressed the possible contribution of the multidrug transporter P-glycoprotein (P-gp or MDR1) in determining 10-OHCBZ brain levels by measuring whether this active metabolite is a substrate of P-gp and the relation between the level of expression of MDR1 and the drug concentration in the same brain tissue specimens. METHODS: Steady-state plasma and brain concentrations (C(ss)) of 10-OHCBZ were determined intraoperatively in 11 patients by high-performance liquid chromatography (HPLC) with UV detection. The level of expression of MDR1 mRNA was measured in surgically resected brain tissue by reverse transcriptase polymerase chain reaction (RT-PCR). The ability of 10-OHCBZ to act as substate of P-gp was evaluated by measuring its uptake in cell lines expressing different levels of P-gp, in the presence or absence of a selective P-gp inhibitor. RESULTS: OXC was converted to 10-OHCBZ and to Di-OHCBZ, the two main metabolites measured in plasma. The brain concentrations of the active metabolite 10-OHCBZ did not reflect plasma C(ss). A significant inverse linear correlation was found between 10-OHCBZ brain-to-plasma concentration ratio and the level of brain expression of MDR1 mRNA. In vitro uptake studies demonstrated lower intracellular 10-OHCBZ levels in cells with higher P-gp expression. Intracellular drug concentration was increased by XR9576, a specific P-gp blocker. CONCLUSIONS: Pharmacologic failure of OXC in pharmacoresistant epilepsy is unlikely to be due to alterations in drug metabolism. 10-OHCBZ does not appear to cross the blood-brain barrier by simple diffusion, and it acts as a substrate of P-gp. The level of expression of MDR1 is inversely correlated with 10-OHCBZ concentration in the epileptic tissue. P-gp may play a role in the pharmacoresistance to OXC by determining the attainment of insufficient concentrations of its active metabolite at neuronal targets.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Anticonvulsivantes/uso terapêutico , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Carbamazepina/análogos & derivados , Epilepsia/tratamento farmacológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/sangue , Adulto , Idoso , Anticonvulsivantes/sangue , Anticonvulsivantes/metabolismo , Carbamazepina/sangue , Carbamazepina/metabolismo , Carbamazepina/uso terapêutico , Criança , Resistência a Medicamentos , Resistência a Múltiplos Medicamentos , Epilepsia/sangue , Epilepsia/metabolismo , Feminino , Humanos , Técnicas In Vitro , Masculino , Oxcarbazepina , Projetos Piloto , RNA Mensageiro/metabolismo
16.
Epilepsia ; 46 Suppl 5: 113-7, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15987264

RESUMO

PURPOSE: We investigated the activation of microglia and astrocytes, induction of cytokines, and hippocampal neuronal damage, 4 and 24 h after kainic acid-induced status epilepticus (SE) in postnatal day (PN) 9, 15, and 21 rats. METHODS: Limbic seizures were induced by systemic injection of kainic acid. Glia activation and neuronal cell loss were studied by using immunocytochemistry and Western blot. Cytokine expression was analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR) followed by Southern blot quantification. RESULTS: After SE onset, hippocampal glia activation, cytokine expression, and neuronal damage are all age-dependent phenomena. In the hippocampus, neuronal injury occurs only when cytokines are induced in glia, and cytokine synthesis precedes the appearance of degenerating neurons. Neuronal injury is more pronounced when interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) are produced in addition to IL-1beta. CONCLUSIONS: This study shows that cytokine induction in rat brain after sustained seizures is age dependent, and it is associated with the appearance of cell injury.


Assuntos
Hipocampo/crescimento & desenvolvimento , Hipocampo/imunologia , Inflamação/fisiopatologia , Neuroglia/imunologia , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/imunologia , Animais , Animais Recém-Nascidos , Astrócitos/imunologia , Astrócitos/fisiologia , Western Blotting , Citocinas/imunologia , Citocinas/fisiologia , Modelos Animais de Doenças , Gliose/imunologia , Gliose/fisiopatologia , Hipocampo/fisiopatologia , Imuno-Histoquímica , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/fisiologia , Interleucina-6/imunologia , Ácido Caínico , Masculino , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Neuroglia/fisiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estado Epiléptico/fisiopatologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/fisiologia
17.
Epilepsy Res ; 60(2-3): 203-13, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15380564

RESUMO

There is recent evidence that increased expression of multidrug transporters, such as P-glycoprotein (P-gp), may lead to reduced antiepileptic drug (AED) concentrations in the brain, shortly after status epilepticus (SE), thereby suggesting a possible mechanism for drug-resistance. To get insights on whether increased P-gp expression is a consequence of the initial insult, or evolves more gradually as a result of recurrent spontaneous seizures, we used a rat model of temporal lobe epilepsy in which spontaneous seizures develop after an electrically induced SE. We investigated the temporal and region-specific expression of two isoforms of the multidrug resistance gene (mdr1a and mdr1b, both encoding for P-gp) in two regions within the temporal lobe (the dentate gyrus (DG) and the parahippocampal cortex (PHC)). Using real-time PCR, we found that the mdr1b isoform was increased in the temporal lobe, 1 week after SE; however, this increase was reversible in dentate gyrus while it persisted in the parahippocampal cortex of chronic epileptic rats. Mdr1b upregulation was related to the occurrence of spontaneous seizures, since this isoform was unchanged in rats that were stimulated, but that did not develop SE (non-SE). The mdr1a isoform was transiently upregulated in the dentate gyrus. P-gp immunostaining was enhanced in endothelial and glia-like cells, 1 week after SE. In chronic epileptic rats, the number of strongly P-gp positive glia-like cells was much lower than 1 week after SE, and it was mainly present in the most ventral part of the temporal lobe. These cells were in close apposition to strongly stained blood vessels. These findings show that both mdr1a and mdr1b are induced by SE, although the increase in mdr1b isoform was more persistent. More importantly, increased P-gp expression is still present in chronic epileptic rats.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/metabolismo , Giro Para-Hipocampal/metabolismo , RNA Mensageiro/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Doença Crônica , Primers do DNA , DNA Complementar/biossíntese , DNA Complementar/genética , Giro Denteado/metabolismo , Estimulação Elétrica , Eletrodos Implantados , Eletroencefalografia , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Recidiva , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Convulsões/metabolismo , Regulação para Cima , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
18.
Neurobiol Dis ; 14(3): 494-503, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14678765

RESUMO

In adult rats, status epilepticus (SE) induces cytokine production by glia especially when seizures are associated with neuronal injury. This suggests that cytokines may play a role in seizure-induced neuronal damage. As SE-induced injury is age-specific, we used rats of different ages (with distinct susceptibilities to seizure-induced neuronal injury) to elucidate the role of cytokines in this process. Thus, we investigated the activation of microglia and astrocytes, induction of cytokines, and hippocampal neuronal injury 4 and 24 h following kainic acid-induced SE in postnatal day (PN) 9, 15, and 21 rats. At PN9, there was little activation of microglia and astrocytes at any time point studied. Interleukin-1beta (IL), tumor necrosis factor-alpha (TNF), and IL-6 or the naturally occurring IL-1 receptor antagonist (Ra) mRNA expression did not increase. No evidence of cell injury has been detected. At PN15, immunostaining of microglia and astrocytes was enhanced, but only IL-1beta mRNA expression was increased. These changes were observed 4 h after SE. Scattered injured neurons in CA3 and subiculum, but not in any other region, were present 24 h following SE. At PN21, immunostaining of microglia and astrocytes and the mRNA expression of all cytokines studied was significantly increased already 4 h after SE. At 24 h, many injured neurons were present in CA1 and CA3 regions and in 40% of rats in other forebrain areas. These data show that (i) the pattern of glia activation and cytokine gene transcription induced by SE is age-dependent and (ii) neuronal injury in the hippocampus occurs only when cytokines are induced and their synthesis precedes the appearance of neuronal damage. Thus, cytokine expression in immature brain is associated specifically with cell injury rather than with seizures per se, suggesting that proinflammatory cytokines may contribute to the occurence of SE-induced hippocampal damage.


Assuntos
Envelhecimento/metabolismo , Citocinas/metabolismo , Gliose/fisiopatologia , Hipocampo/fisiopatologia , Degeneração Neural/fisiopatologia , Estado Epiléptico/metabolismo , Animais , Animais Recém-Nascidos , Citocinas/genética , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/fisiopatologia , Epilepsia/imunologia , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Feminino , Gliose/imunologia , Gliose/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Mediadores da Inflamação/metabolismo , Proteína Antagonista do Receptor de Interleucina 1 , Ácido Caínico , Masculino , Degeneração Neural/imunologia , Degeneração Neural/metabolismo , Neuroglia/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Sialoglicoproteínas/metabolismo , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/imunologia , Regulação para Cima/fisiologia
19.
J Neurosci ; 22(14): 5833-9, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12122045

RESUMO

The causes and mechanisms underlying multidrug resistance (MDR) in epilepsy are still elusive and may depend on inadequate drug concentration in crucial brain areas. We studied whether limbic seizures or anticonvulsant drug treatments in rodents enhance the brain expression of the MDR gene (mdr) encoding a permeability glycoprotein (P-gp) involved in MDR to various cancer chemotherapeutic agents. We also investigated whether changes in P-gp levels affect anticonvulsant drug concentrations in the brain. Mdr mRNA measured by RT-PCR increased by 85% on average in the mouse hippocampus 3-24 hr after kainic acid-induced limbic seizures, returning to control levels by 72 hr. Treatment with therapeutic doses of phenytoin or carbamazepine for 7 d did not change mdr mRNA expression in the mouse hippocampus 1-72 hr after the last drug administration. Six hours after seizures, the brain/plasma ratio of phenytoin was reduced by 30% and its extracellular concentration estimated by microdialysis was increased by twofold compared with control mice. Knock-out mice (mdr1a/b -/-) lacking P-gp protein showed a 46% increase in phenytoin concentrations in the hippocampus 1 and 4 hr after injection compared with wild-type mice. A significant 23% increase was found in the cerebellum at 1 hr and in the cortex at 4 hr. Carbamazepine concentrations were measurable in the hippocampus at 3 hr in mdr1a/b -/- mice, whereas they were undetectable at the same time interval in wild-type mice. In rats having spontaneous seizures 3 months after electrically induced status epilepticus, mdr1 mRNA levels were enhanced by 1.8-fold and fivefold on average in the hippocampus and entorhinal cortex, respectively. Thus, changes in P-gp mRNA levels occur in limbic areas after both acute and chronic epileptic activity. P-gp alterations significantly affect antiepileptic drugs concentrations in the brain, suggesting that seizure-induced mdr mRNA expression contributes to MDR in epilepsy.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Encéfalo/fisiopatologia , Resistência a Medicamentos , Convulsões/fisiopatologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Anticonvulsivantes/farmacocinética , Anticonvulsivantes/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Resistência a Medicamentos/fisiologia , Estimulação Elétrica , Eletroencefalografia , Córtex Entorrinal/efeitos dos fármacos , Córtex Entorrinal/metabolismo , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Homozigoto , Sistema Límbico/efeitos dos fármacos , Sistema Límbico/metabolismo , Sistema Límbico/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...