Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Curr Pharm Des ; 23(37): 5569-5576, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28950818

RESUMO

BACKGROUND: The lack of treatments which can prevent epilepsy development or improve disease prognosis represents an unmet and urgent clinical need. The development of such drugs requires a deep understanding of the mechanisms underlying disease pathogenesis. In the last decade, preclinical studies in models of acute seizures and of chronic epilepsy highlighted that neuroinflammation arising in brain areas of seizure onset and generalization is a key contributor to neuronal hyper-excitability underlying seizure generation. Microglia and astrocytes are pivotal cells involved in both the induction and perpetuation of the inflammatory response to epileptogenic injuries or seizures; other cell contributors are neurons, cell components of the blood brain barrier and leukocytes. METHODS: From the clinical standpoint, neuroinflammation is now considered an hallmark of epileptogenic foci in various forms of focal onset pharmacoresistant epilepsies. Moreover, pharmacological studies in animal model with drugs targeting specific inflammatory molecules, and changes in intrinsic seizure susceptibility of transgenic mice with perturbed neuroinflammatory mechanisms, have demonstrated that neuroinflammation is not a bystander phenomenon but has a pathogenic role in seizures, cell loss and neurological co-morbidities. Understanding the role of neuroinflammation in seizure pathogenesis is instrumental for a mechanism-based discovery of selective therapies targeting the epilepsy causes rather than its symptoms, thereby allowing the development of novel disease-modifying treatments. Notably, clinical translation of laboratory findings may take advantage of anti-inflammatory drugs already in medical use for peripheral autoinflammatory or autoimmune disorders. CONCLUSION: This review reports key preclinical and clinical findings supporting a role for brain inflammation in the pathogenesis of seizures. It also highlights the emerging proof-of-concept studies showing signs of clinical efficacy of target-specific anti-inflammatory interventions in epilepsies of differing etiologies. We will discuss the need for biomarkers and novel clinical trial designs for anti-inflammatory therapies that have a mechanism of action very different than standard antiepileptic drugs.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticonvulsivantes/uso terapêutico , Encefalite/complicações , Epilepsia/tratamento farmacológico , Epilepsia/patologia , Animais , Avaliação Pré-Clínica de Medicamentos , Epilepsia/etiologia , Humanos
3.
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
4.
J Clin Invest ; 127(6): 2118-2132, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28504645

RESUMO

Approximately 30% of epilepsy patients do not respond to antiepileptic drugs, representing an unmet medical need. There is evidence that neuroinflammation plays a pathogenic role in drug-resistant epilepsy. The high-mobility group box 1 (HMGB1)/TLR4 axis is a key initiator of neuroinflammation following epileptogenic injuries, and its activation contributes to seizure generation in animal models. However, further work is required to understand the role of HMGB1 and its isoforms in epileptogenesis and drug resistance. Using a combination of animal models and sera from clinically well-characterized patients, we have demonstrated that there are dynamic changes in HMGB1 isoforms in the brain and blood of animals undergoing epileptogenesis. The pathologic disulfide HMGB1 isoform progressively increased in blood before epilepsy onset and prospectively identified animals that developed the disease. Consistent with animal data, we observed early expression of disulfide HMGB1 in patients with newly diagnosed epilepsy, and its persistence was associated with subsequent seizures. In contrast with patients with well-controlled epilepsy, patients with chronic, drug-refractory epilepsy persistently expressed the acetylated, disulfide HMGB1 isoforms. Moreover, treatment of animals with antiinflammatory drugs during epileptogenesis prevented both disease progression and blood increase in HMGB1 isoforms. Our data suggest that HMGB1 isoforms are mechanistic biomarkers for epileptogenesis and drug-resistant epilepsy in humans, necessitating evaluation in larger-scale prospective studies.


Assuntos
Epilepsia/sangue , Proteína HMGB1/metabolismo , Adolescente , Adulto , Idoso , Animais , Anti-Inflamatórios/farmacologia , Anticonvulsivantes/farmacologia , Biomarcadores/sangue , Encéfalo/metabolismo , Avaliação Pré-Clínica de Medicamentos , Resistência a Medicamentos , Epilepsia/tratamento farmacológico , Feminino , Proteína HMGB1/genética , Humanos , Masculino , Pessoa de Meia-Idade , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Curva ROC , Ratos Sprague-Dawley , Adulto Jovem
5.
Neuropharmacology ; 96(Pt A): 70-82, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25445483

RESUMO

Increasing evidence underlines that prototypical inflammatory cytokines (IL-1ß, TNF-α and IL-6) either synthesized in the central (CNS) or peripheral nervous system (PNS) by resident cells, or imported by immune blood cells, are involved in several pathophysiological functions, including an unexpected impact on synaptic transmission and neuronal excitability. This review describes these unconventional neuromodulatory properties of cytokines, that are distinct from their classical action as effector molecules of the immune system. In addition to the role of cytokines in brain physiology, we report evidence that dysregulation of their biosynthesis and cellular release, or alterations in receptor-mediated intracellular pathways in target cells, leads to neuronal cell dysfunction and modifications in neuronal network excitability. As a consequence, targeting of these cytokines, and related signalling molecules, is considered a novel option for the development of therapies in various CNS or PNS disorders associated with an inflammatory component. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.


Assuntos
Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Neuroglia/fisiologia , Neurônios/fisiologia , Transmissão Sináptica , Animais , Canais de Cálcio/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Neuroglia/metabolismo , Plasticidade Neuronal , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo
6.
Epilepsia ; 54 Suppl 4: 61-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23909854

RESUMO

A biomarker is defined as an objectively measured characteristic of a normal or pathologic biologic process. Identification and proper validation of biomarkers of epileptogenesis (the development of epilepsy) and ictogenesis (the propensity to generate spontaneous seizures) might predict the development of an epilepsy condition; identify the presence and severity of tissue capable of generating spontaneous seizures; measure progression after the condition is established; and determine pharmacoresistance. Such biomarkers could be used to create animal models for more cost-effective screening of potential antiepileptogenic and antiseizure drugs and devices, and to reduce the cost of clinical trials by enriching the trial population, and acting as surrogate markers to shorten trial duration. The objectives of the biomarker subgroup for the London Workshop were to define approaches for identifying possible biomarkers for these purposes. Research to identify reliable biomarkers may also reveal underlying mechanisms that could serve as therapeutic targets for the development of new antiepileptogenic and antiseizure compounds.


Assuntos
Anticonvulsivantes/uso terapêutico , Biomarcadores/sangue , Descoberta de Drogas , Drogas em Investigação/uso terapêutico , Epilepsia/tratamento farmacológico , Epilepsia/fisiopatologia , Animais , Anticonvulsivantes/efeitos adversos , Anticonvulsivantes/economia , Encéfalo/fisiopatologia , Ensaios Clínicos como Assunto/economia , Análise Custo-Benefício , Modelos Animais de Doenças , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos/economia , Resistência a Medicamentos , Drogas em Investigação/efeitos adversos , Drogas em Investigação/economia , Eletroencefalografia/efeitos dos fármacos , Epilepsia/etiologia , Epilepsia/prevenção & controle , Humanos , Fatores Desencadeantes
7.
J Neurosci ; 25(8): 1943-51, 2005 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-15728834

RESUMO

Experimental studies suggest that the delivery of antiepileptic agents into the seizure focus might be of potential utility for the treatment of focal-onset epilepsies. Botulinum neurotoxin E (BoNT/E) causes a prolonged inhibition of neurotransmitter release after its specific cleavage of the synaptic protein synaptosomal-associated protein of 25 kDa (SNAP-25). Here, we show that BoNT/E injected into the rat hippocampus inhibits glutamate release and blocks spike activity of pyramidal neurons. BoNT/E effects persist for at least 3 weeks, as determined by immunodetection of cleaved SNAP-25 and loss of intact SNAP-25. The delivery of BoNT/E to the rat hippocampus dramatically reduces both focal and generalized kainic acid-induced seizures as documented by behavioral and electrographic analysis. BoNT/E treatment also prevents neuronal loss and long-term cognitive deficits associated with kainic acid seizures. Moreover, BoNT/E-injected rats require 50% more electrical stimulations to reach stage 5 of kindling, thus indicating a delayed epileptogenesis. We conclude that BoNT/E delivery to the hippocampus is both antiictal and antiepileptogenic in experimental models of epilepsy.


Assuntos
Anticonvulsivantes/uso terapêutico , Toxinas Botulínicas/uso terapêutico , Epilepsias Parciais/tratamento farmacológico , Epilepsia Generalizada/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Animais , Anticonvulsivantes/administração & dosagem , Toxinas Botulínicas/administração & dosagem , Morte Celular/efeitos dos fármacos , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/prevenção & controle , Convulsivantes/toxicidade , Avaliação Pré-Clínica de Medicamentos , Estimulação Elétrica , Eletroencefalografia , Epilepsias Parciais/fisiopatologia , Epilepsia Generalizada/induzido quimicamente , Epilepsia Generalizada/complicações , Epilepsia Generalizada/fisiopatologia , Ácido Glutâmico/metabolismo , Hipocampo/fisiopatologia , Injeções Intralesionais , Ácido Caínico/toxicidade , Excitação Neurológica/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Células Piramidais/fisiologia , Distribuição Aleatória , Ratos , Ratos Long-Evans , Técnicas Estereotáxicas , Proteína 25 Associada a Sinaptossoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA