Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 119: 363-380, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38608741

RESUMO

The gut microbiota is altered in epilepsy and is emerging as a potential target for new therapies. We studied the effects of rifaximin, a gastrointestinal tract-specific antibiotic, on seizures and neuropathology and on alterations in the gut and its microbiota in a mouse model of temporal lobe epilepsy (TLE). Epilepsy was induced by intra-amygdala kainate injection causing status epilepticus (SE) in C57Bl6 adult male mice. Sham mice were injected with vehicle. Two cohorts of SE mice were fed a rifaximin-supplemented diet for 21 days, starting either at 24 h post-SE (early disease stage) or at day 51 post-SE (chronic disease stage). Corresponding groups of SE mice (one each disease stage) were fed a standard (control) diet. Cortical ECoG recording was done at each disease stage (24/7) for 21 days in all SE mice to measure the number and duration of spontaneous seizures during either rifaximin treatment or control diet. Then, epileptic mice ± rifaximin and respective sham mice were sacrificed and brain, gut and feces collected. Biospecimens were used for: (i) quantitative histological analysis of the gut structural and cellular components; (ii) markers of gut inflammation and intestinal barrier integrity by RTqPCR; (iii) 16S rRNA metagenomics analysis in feces. Hippocampal neuronal cell loss was assessed in epileptic mice killed in the early disease phase. Rifaximin administered for 21 days post-SE (early disease stage) reduced seizure duration (p < 0.01) and prevented hilar mossy cells loss in the hippocampus compared to epileptic mice fed a control diet. Epileptic mice fed a control diet showed a reduction of both villus height and villus height/crypt depth ratio (p < 0.01) and a decreased number of goblet cells (p < 0.01) in the duodenum, as well as increased macrophage (Iba1)-immunostaining in the jejunum (p < 0.05), compared to respective sham mice. Rifaximin's effect on seizures was associated with a reversal of gut structural and cellular changes, except for goblet cells which remained reduced. Seizure duration in epileptic mice was negatively correlated with the number of mossy cells (p < 0.01) and with villus height/crypt depth ratio (p < 0.05). Rifaximin-treated epileptic mice also showed increased tight junctions (occludin and ZO-1, p < 0.01) and decreased TNF mRNA expression (p < 0.01) in the duodenum compared to epileptic mice fed a control diet. Rifaximin administered for 21 days in chronic epileptic mice (chronic disease stage) did not change the number or duration of seizures compared to epileptic mice fed a control diet. Chronic epileptic mice fed a control diet showed an increased crypt depth (p < 0.05) and reduced villus height/crypt depth ratio (p < 0.01) compared to respective sham mice. Rifaximin treatment did not affect these intestinal changes. At both disease stages, rifaximin modified α- and ß-diversity in epileptic and sham mice compared to respective mice fed a control diet. The microbiota composition in epileptic mice, as well as the effects of rifaximin at the phylum, family and genus levels, depended on the stage of the disease. During the early disease phase, the abundance of specific taxa was positively correlated with seizure duration in epileptic mice. In conclusion, gut-related alterations reflecting a dysfunctional state, occur during epilepsy development in a TLE mouse model. A short-term treatment with rifaximin during the early phase of the disease, reduced seizure duration and neuropathology, and reversed some intestinal changes, strengthening the therapeutic effects of gut-based therapies in epilepsy.


Assuntos
Modelos Animais de Doenças , Epilepsia do Lobo Temporal , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Rifaximina , Convulsões , Animais , Rifaximina/uso terapêutico , Rifaximina/farmacologia , Camundongos , Masculino , Microbioma Gastrointestinal/efeitos dos fármacos , Convulsões/tratamento farmacológico , Epilepsia do Lobo Temporal/tratamento farmacológico , Estado Epiléptico/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Epilepsia/tratamento farmacológico
2.
Epilepsy Behav ; 140: 109095, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36753859

RESUMO

Status epilepticus (SE) is a very heterogeneous clinical condition often refractory to available treatment options. Evidence in animal models shows that neuroinflammation arises in the brain during SE due to the activation of innate immune mechanisms in brain parenchyma cells. Intervention studies in animal models support the involvement of neuroinflammation in SE onset, duration, and severity, refractoriness to treatments, and long-term neurological consequences. Clinical evidence shows that neuroinflammation occurs in patients with SE of diverse etiologies likely representing a common phenomenon, thus broadening the involvement of the immune system beyond the infective and autoimmune etiologies. There is urgent need for novel therapies for refractory SE that rely upon a better understanding of the basic mechanisms underlying this clinical condition. Preclinical and clinical evidence encourage consideration of specific anti-inflammatory treatments for controlling SE and its consequences in patients.


Assuntos
Doenças Neuroinflamatórias , Estado Epiléptico , Animais , Estado Epiléptico/tratamento farmacológico , Encéfalo , Modelos Animais , Anticonvulsivantes/uso terapêutico
3.
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
4.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-34638900

RESUMO

We assessed the effect of antioxidant therapy using the Food and Drug Administration-approved respiratory drug N-acetylcysteine (NAC) or sulforaphane (SFN) as monotherapies or duotherapy in vitro in neuron-BV2 microglial co-cultures and validated the results in a lateral fluid-percussion model of TBI in rats. As in vitro measures, we assessed neuronal viability by microtubule-associated-protein 2 immunostaining, neuroinflammation by monitoring tumor necrosis factor (TNF) levels, and neurotoxicity by measuring nitrite levels. In vitro, duotherapy with NAC and SFN reduced nitrite levels to 40% (p < 0.001) and neuroinflammation to -29% (p < 0.001) compared with untreated culture. The treatment also improved neuronal viability up to 72% of that in a positive control (p < 0.001). The effect of NAC was negligible, however, compared with SFN. In vivo, antioxidant duotherapy slightly improved performance in the beam walking test. Interestingly, duotherapy treatment decreased the plasma interleukin-6 and TNF levels in sham-operated controls (p < 0.05). After TBI, no treatment effect on HMGB1 or plasma cytokine levels was detected. Also, no treatment effects on the composite neuroscore or cortical lesion area were detected. The robust favorable effect of duotherapy on neuroprotection, neuroinflammation, and oxidative stress in neuron-BV2 microglial co-cultures translated to modest favorable in vivo effects in a severe TBI model.


Assuntos
Acetilcisteína/farmacologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Isotiocianatos/farmacologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Sulfóxidos/farmacologia , Animais , Antioxidantes/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Ratos Sprague-Dawley
5.
Neurobiol Dis ; 158: 105468, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34358616

RESUMO

CXCL1, a functional murine orthologue of the human chemokine CXCL8 (IL-8), and its CXCR1 and CXCR2 receptors were investigated in a murine model of acquired epilepsy developing following status epilepticus (SE) induced by intra-amygdala kainate. CXCL8 and its receptors were also studied in human temporal lobe epilepsy (TLE). The functional involvement of the chemokine in seizure generation and neuronal cell loss was assessed in mice using reparixin (formerly referred to as repertaxin), a non-competitive allosteric inhibitor of CXCR1/2 receptors. We found a significant increase in hippocampal CXCL1 level within 24 h of SE onset that lasted for at least 1 week. No changes were measured in blood. In analogy with human TLE, immunohistochemistry in epileptic mice showed that CXCL1 and its two receptors were increased in hippocampal neuronal cells. Additional expression of these molecules was found in glia in human TLE. Mice were treated with reparixin or vehicle during SE and for additional 6 days thereafter, using subcutaneous osmotic minipumps. Drug-treated mice showed a faster SE decay, a reduced incidence of acute symptomatic seizures during 48 h post-SE, and a delayed time to spontaneous seizures onset compared to vehicle controls. Upon reparixin discontinuation, mice developed spontaneous seizures similar to vehicle mice, as shown by EEG monitoring at 14 days and 2.5 months post-SE. In the same epileptic mice, reparixin reduced neuronal cell loss in the hippocampus vs vehicle-injected mice, as assessed by Nissl staining at completion of EEG monitoring. Reparixin administration for 2 weeks in mice with established chronic seizures, reduced by 2-fold on average seizure number vs pre-treatment baseline, and this effect was reversible upon drug discontinuation. No significant changes in seizure number were measured in vehicle-injected epileptic mice that were EEG monitored in parallel. Data show that CXCL1-IL-8 signaling is activated in experimental and human epilepsy and contributes to acute and chronic seizures in mice, therefore representing a potential new target to attain anti-ictogenic effects.


Assuntos
Quimiocina CXCL1/genética , Epilepsia do Lobo Temporal/genética , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8B/genética , Convulsões/genética , Animais , Quimiocina CXCL1/antagonistas & inibidores , Eletroencefalografia , Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Receptores de Interleucina-8A/antagonistas & inibidores , Receptores de Interleucina-8B/antagonistas & inibidores , Convulsões/fisiopatologia , Estado Epiléptico/genética , Estado Epiléptico/patologia , Sulfonamidas/farmacologia
6.
Neuropharmacology ; 167: 107742, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31421074

RESUMO

Neuroinflammation and reactive oxygen and nitrogen species are rapidly induced in the brain after acute cerebral injuries that are associated with an enhanced risk for epilepsy in humans and related animal models. These phenomena reinforce each others and persist during epileptogenesis as well as during chronic spontaneous seizures. Anti-inflammatory and anti-oxidant drugs transiently administered either before, or shortly after the clinical onset of symptomatic epilepsy, similarly block the progression of spontaneous seizures, and may delay their onset. Moreover, neuroprotection and rescue of cognitive deficits are also observed in the treated animals. Therefore, although these treatments do not prevent epilepsy development, they offer clinically relevant disease-modification effects. These therapeutic effects are mediated by targeting molecular signaling pathways such as the IL-1ß-IL-1 receptor type 1 and TLR4, P2X7 receptors, the transcriptional anti-oxidant factor Nrf2, while the therapeutic impact of COX-2 inhibition for reducing spontaneous seizures remains controversial. Some anti-inflammatory and anti-oxidant drugs that are endowed of disease modification effects in preclinical models are already in medical use and have a safety profile, therefore, they provide potential re-purposed treatments for improving the disease course and for reducing seizure burden. Markers of neuroinflammation and oxidative stress can be measured in blood or by neuroimaging, therefore they represent testable prognostic and predictive biomarkers for selecting the patient's population at high risk for developing epilepsy therefore eligible for novel treatments. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticonvulsivantes/uso terapêutico , Antioxidantes/uso terapêutico , Epilepsia/tratamento farmacológico , Mediadores da Inflamação/antagonistas & inibidores , Animais , Anti-Inflamatórios/farmacologia , Anticonvulsivantes/farmacologia , Epilepsia/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
7.
Nat Rev Neurol ; 15(8): 459-472, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31263255

RESUMO

Epilepsy is a chronic neurological disease characterized by an enduring propensity for generation of seizures. The pathogenic processes of seizure generation and recurrence are the subject of intensive preclinical and clinical investigations as their identification would enable development of novel treatments that prevent epileptic seizures and reduce seizure burden. Such treatments are particularly needed for pharmacoresistant epilepsies, which affect ~30% of patients. Neuroinflammation is commonly activated in epileptogenic brain regions in humans and is clearly involved in animal models of epilepsy. An increased understanding of neuroinflammatory mechanisms in epilepsy has identified cellular and molecular targets for new mechanistic therapies or existing anti-inflammatory drugs that could overcome the limitations of current medications, which provide only symptomatic control of seizures. Moreover, inflammatory mediators in the blood and molecular imaging of neuroinflammation could provide diagnostic, prognostic and predictive biomarkers for epilepsy, which will be instrumental for patient stratification in future clinical studies. In this Review, we focus on our understanding of the IL-1 receptor-Toll-like receptor 4 axis, the arachidonic acid-prostaglandin cascade, oxidative stress and transforming growth factor-ß signalling associated with blood-brain barrier dysfunction, all of which are pathways that are activated in pharmacoresistant epilepsy in humans and that can be modulated in animal models to produce therapeutic effects on seizures, neuronal cell loss and neurological comorbidities.


Assuntos
Encefalite/diagnóstico , Encefalite/metabolismo , Epilepsia/diagnóstico , Epilepsia/terapia , Animais , Ácido Araquidônico/metabolismo , Biomarcadores/sangue , Encefalite/complicações , Epilepsia/complicações , Epilepsia/metabolismo , Humanos , Estresse Oxidativo , Prostaglandinas/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Fator de Crescimento Transformador beta/metabolismo
8.
Epilepsy Behav ; 101(Pt B): 106275, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31171434

RESUMO

Preclinical studies in immature and adult rodents and clinical observations show that neuroinflammation and oxidative stress are rapid onset phenomena occurring in the brain during status epilepticus and persisting thereafter. Notably, both neuroinflammation and oxidative stress contribute to the acute and long-term sequelae of status epilepticus thus representing potential druggable targets. Antiinflammatory drugs that interfere with the IL-1ß pathway, such as anakinra, can control benzodiazepine-refractory status epilepticus in animals, and there is recent proof-of-concept evidence for therapeutic effects in children with Febrile infection related epilepsy syndrome (FIRES). Inhibitors of monoacylglycerol lipase and P2X7 receptor antagonists are also promising antiinflammatory drug candidates for rapidly aborting de novo status epilepticus and provide neuroprotection. Antiinflammatory and antioxidant drugs administered to rodents during status epilepticus and transiently thereafter, prevent long-term sequelae such as cognitive deficits and seizure progression in animals developing epilepsy. Some drugs are already in medical use and are well-tolerated, therefore, they may be considered for treating status epilepticus and its neurological consequences. Finally, markers of neuroinflammation and oxidative stress are measureable in peripheral blood and by neuroimaging, which offers an opportunity for developing prognostic and predictive mechanistic biomarkers in people exposed to status epilepticus. This article is part of the Special Issue "Proceedings of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures.


Assuntos
Anti-Inflamatórios/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Biomarcadores/sangue , Biomarcadores/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Espécies Reativas de Oxigênio/antagonistas & inibidores , Convulsões/tratamento farmacológico , Convulsões/metabolismo
9.
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
10.
Neurotherapeutics ; 15(2): 470-488, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29464573

RESUMO

Understanding the mechanisms of epileptogenesis is essential to develop novel drugs that could prevent or modify the disease. Neuroinflammation has been proposed as a promising target for therapeutic interventions to inhibit the epileptogenic process that evolves from traumatic brain injury. However, it remains unclear whether cytokine-related pathways, particularly TNFα signaling, have a critical role in the development of epilepsy. In this study, we investigated the role of innate inflammation in an in vitro model of post-traumatic epileptogenesis. We combined organotypic hippocampal slice cultures, representing an in vitro model of post-traumatic epilepsy, with multi-electrode array recordings to directly monitor the development of epileptiform activity and to examine the concomitant changes in cytokine release, cell death, and glial cell activation. We report that synchronized ictal- and interictal-like activities spontaneously evolve in this culture. Dynamic changes in the release of the pro-inflammatory cytokines IL-1ß, TNFα, and IL-6 were observed throughout the culture period (3 to 21 days in vitro) with persistent activation of microglia and astrocytes. We found that neutralizing TNFα with a polyclonal antibody significantly reduced ictal discharges, and this effect lasted for 1 week after antibody washout. Neither phenytoin nor an anti-IL-6 polyclonal antibody was efficacious in inhibiting the development of epileptiform activity. Our data show a sustained effect of the anti-TNFα antibody on the ictal progression in organotypic hippocampal slice cultures supporting the critical role of inflammatory mediators in epilepsy and establishing a proof-of-principle evidence for the utility of this preparation to test the therapeutic effects of anti-inflammatory treatments.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Encefalite/metabolismo , Epilepsia/metabolismo , Hipocampo/metabolismo , Animais , Astrócitos/metabolismo , Lesões Encefálicas Traumáticas/complicações , Morte Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/complicações , Epilepsia/etiologia , Feminino , Masculino , Microglia/metabolismo , Neurônios/metabolismo , Ratos Sprague-Dawley , Técnicas de Cultura de Tecidos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/imunologia
11.
Brain Behav Immun ; 72: 14-21, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29031614

RESUMO

Approximately 30% of epilepsy patients experience seizures that are not controlled by the available drugs. Moreover, these drugs provide mainly a symptomatic treatment since they do not interfere with the disease's mechanisms. A mechanistic approach to the discovery of key pathogenic brain modifications causing seizure onset, recurrence and progression is instrumental for designing novel and rationale therapeutic interventions that could modify the disease course or prevent its development. In this regard, increasing evidence shows that neuroinflammation is a pathogenic factor in drug-resistant epilepsies. The High Mobility Group Box 1 (HMGB1)/Toll-like receptor 4 axis is a key initiator of neuroinflammation following brain injuries leading to epilepsy, and its activation contributes to seizure mechanisms in animal models. Recent findings have shown dynamic changes in HMGB1 and its isoforms in the brain and blood of animals exposed to acute brain injuries and undergoing epileptogenesis, and in surgically resected epileptic foci in humans. HMGB1 isoforms reflect different pathophysiological processes, and the disulfide isoform, which is generated in the brain during oxidative stress, is implicated in seizures, cell loss and cognitive dysfunctions. Interfering with disulfide HMGB1-activated cell signaling mediates significant therapeutic effects in epilepsy models. Moreover, both clinical and experimental data suggest that HMGB1 isoforms may serve as mechanistic biomarkers for epileptogenesis and drug-resistant epilepsy. These novel findings suggest that the HMGB1 system could be targeted to prevent seizure generation and may provide clinically useful prognostic biomarkers which may also predict the patient's response to therapy.


Assuntos
Epilepsia/patologia , Proteína HMGB1/metabolismo , Alarminas/metabolismo , Alarminas/fisiologia , Animais , Biomarcadores/sangue , Encéfalo/metabolismo , Disfunção Cognitiva/complicações , Modelos Animais de Doenças , Epilepsia/metabolismo , Proteína HMGB1/fisiologia , Humanos , Convulsões/etiologia , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo
12.
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
13.
Antioxid Redox Signal ; 21(12): 1726-40, 2014 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-24094148

RESUMO

AIMS: Using primary cultures of mouse hippocampal neurons, we studied the molecular and functional interactions between high mobility group box-1 (HMGB1) and the N-methyl-d-aspartate receptor (NMDAR), two proteins playing a key role in neuronal hyperexcitability. By measuring NMDA-induced calcium (Ca(2+)) increase in neuronal somata and neurotoxicity as functional read-out parameters, we explored the role of the redox state of HMGB1, the receptor involved, and the molecular signaling underlying its interactions with postsynaptic NMDAR. We also investigated whether HMGB1 redox state affects its proconvulsive effects in mice. RESULTS: Nonoxidizable HMGB1 with a triple cysteine-to-serine replacement (3S-HMGB1) was ineffective on NMDA response. Conversely, the disulfide-containing form of HMGB1 dose dependently enhanced NMDA-induced Ca(2+) increase in neuronal cell bodies. This effect was prevented by BoxA, a competitive HMGB1 antagonist, and by Rhodobacter sphaeroides lipopolysaccharide (LPS-RS), a toll-like receptor 4 (TLR4) selective antagonist, and it was abrogated in neurons lacking TLR4 while persisting in the absence of receptor for advanced glycation end products (RAGE). TLR4 and NMDAR subunit 1 (NR1) and 2B (NR2B) were colocalized in neurons. Disulfide HMGB1 effect on NMDA-induced Ca(2+) influx was prevented by 3-O-methylsphingomyelin (3-O-MS) and 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyrimidine, (PP2) selective inhibitors of neutral sphingomyelinase and Src-family Tyr kinases, respectively. Disulfide HMGB1, but not 3S-HMGB1, increased Tyr(1472) phosphorylation of the NR2B subunit of the NMDAR, which is known to increase Ca(2+) channel permeability. Similarly, disulfide HMGB1 increased NMDA-induced neuronal cell death in vitro and enhanced kainate-induced seizures in vivo. INNOVATION AND CONCLUSION: We describe a novel molecular neuronal pathway activated by HMGB1 that could be targeted in vivo to prevent neurodegeneration and seizures mediated by excessive NMDARs stimulation.


Assuntos
Dissulfetos/metabolismo , Proteína HMGB1/química , Proteína HMGB1/metabolismo , Hipocampo/citologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução
16.
Brain Behav Immun ; 25(7): 1281-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21473909

RESUMO

Increasing evidence supports the involvement of immune and inflammatory processes in the etiopathogenesis of seizures. In particular, activation of innate immune mechanisms and the subsequent inflammatory responses, that are induced in the brain by infection, febrile seizures, neurotrauma, stroke are well documented conditions associated with acute symptomatic seizures and with a high risk of developing epilepsy. A decade ago, pharmacological experiments showed that elevated brain levels of the anti-inflammatory molecule IL-1 receptor antagonist reduced seizures in epilepsy models. This observation, together with the evidence of in situ induction of inflammatory mediators and their receptors in experimental and human epileptogenic brain tissue, established the proof-of-concept evidence that the activation of innate immunity and inflammation in the brain are intrinsic features of the pathologic hyperexcitable tissue. Recent breakthroughs in understanding the molecular organization of the innate immune system first in macrophages, then in the different cell types of the CNS, together with pharmacological and genetic studies in epilepsy models, showed that the activation of IL-1 receptor/Toll-like receptor (IL-1R/TLR) signaling significantly contributes to seizures. IL-1R/TLR mediated pro-excitatory actions are elicited in the brain either by mimicking bacterial or viral infections and inflammatory responses, or via the action of endogenous ligands. These ligands include proinflammatory cytokines, such as IL-1beta, or danger signals, such as HMGB1, released from activated or injured cells. The IL-1R/TLR signaling mediates rapid post-translational changes in voltage- and ligand-gated ion channels that increase excitability, and transcriptional changes in genes involved in neurotransmission and synaptic plasticity that contribute to lower seizure thresholds chronically. The anticonvulsant effects of inhibitors of the IL-1R/TLR signaling in various seizures models suggest that this system could be targeted to inhibit seizures in presently pharmaco-resistant epilepsies.


Assuntos
Infecções/imunologia , Inflamação/imunologia , Degeneração Neural/imunologia , Receptores de Interleucina-1/metabolismo , Convulsões/imunologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/metabolismo , Humanos , Doenças Neurodegenerativas/imunologia
17.
Brain ; 134(Pt 10): 2828-43, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21482549

RESUMO

We have generated an experimental 'double-hit' model of chronic epilepsy to recapitulate the co-existence of abnormal cortical structure and frequently recurrent seizures as observed in human focal cortical dysplasia. We induced cortical malformations by exposing rats prenatally to methylazoxymethanol acetate and triggered status epilepticus and recurrent seizures in adult methylazoxymethanol acetate rats with pilocarpine. We studied the course of epilepsy and the long-term morphologic and molecular changes induced by the occurrence of status epilepticus and subsequent chronic epilepsy in the malformed methylazoxymethanol acetate exposed brain. Behavioural and electroencephalographic analyses showed that methylazoxymethanol acetate pilocarpine rats develop more severe epilepsy than naïve rats. Morphologic and molecular analyses demonstrated that status epilepticus and subsequent seizures, but not pilocarpine treatment per se, was capable of affecting both cortical architectural and N-methyl-D-aspartate receptor abnormalities induced by methylazoxymethanol acetate. In particular, cortical thickness was further decreased and N-methyl-D-aspartate regulatory subunits were recruited at the postsynaptic membrane. In addition, methylazoxymethanol acetate pilocarpine rats showed abnormally large cortical pyramidal neurons with neurofilament over-expression. These neurons bear similarities to the hypertrophic/dysmorphic pyramidal neurons observed in acquired human focal cortical dysplasia. These data show that status epilepticus sets in motion a pathological process capable of significantly changing the cellular and molecular features of pre-existing experimental cortical malformations. They suggest that seizure recurrence in human focal cortical dysplasia might be an additional factor in establishing a pathological circuitry that favours chronic neuronal hyperexcitability.


Assuntos
Córtex Cerebral/patologia , Malformações do Desenvolvimento Cortical/patologia , Neurônios/patologia , Estado Epiléptico/patologia , Animais , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Malformações do Desenvolvimento Cortical/induzido quimicamente , Malformações do Desenvolvimento Cortical/fisiopatologia , Acetato de Metilazoximetanol , Neurônios/fisiologia , Pilocarpina , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/fisiopatologia
18.
Neurosci Lett ; 497(3): 223-30, 2011 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-21362451

RESUMO

CNS injuries such as trauma, stroke, viral infection, febrile seizures, status epilepticus occurring either in infancy or during a lifetime are considered common risk factors for developing epilepsy. Long term CNS inflammation develops rapidly after these events, suggesting that a pro-inflammatory state in the brain might play a role in the development of the epileptic process. This hypothesis is corroborated by two main lines of evidence: (1) the upregulation of pro-inflammatory signals during epileptogenesis in brain areas of seizure onset/generalization; (2) pharmacological targeting of specific pro-inflammatory pathways after status epilepticus or in kindling shows antiepileptogenic effects. The mechanisms by which pro-inflammatory molecules might favor the establishment of chronic neuronal network hyperexcitability involve both rapid, non-transcriptional effects on glutamate and GABA receptors, and transcriptional activation of genes involved in synaptic plasticity. This emerging evidence predicts that pharmacological interventions targeting brain inflammation might provide a key to new antiepileptic drug design.


Assuntos
Anticonvulsivantes/administração & dosagem , Encéfalo/metabolismo , Encefalite/metabolismo , Encefalite/prevenção & controle , Epilepsia/metabolismo , Epilepsia/prevenção & controle , Animais , Encéfalo/efeitos dos fármacos , Encefalite/complicações , Epilepsia/complicações , Humanos
19.
Neurotherapeutics ; 8(2): 304-15, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21431948

RESUMO

Experimental evidence and clinical observations indicate that brain inflammation is an important factor in epilepsy. In particular, induction of interleukin-converting enzyme (ICE)/caspase-1 and activation of interleukin (IL)-1ß/IL-1 receptor type 1 axis both occur in human epilepsy, and contribute to experimentally induced acute seizures. In this study, the anticonvulsant activity of VX-765 (a selective ICE/caspase-1 inhibitor) was examined in a mouse model of chronic epilepsy with spontaneous recurrent epileptic activity refractory to some common anticonvulsant drugs. Moreover, the effects of this drug were studied in one acute model of seizures in mice, previously shown to involve activation of ICE/caspase-1. Quantitative analysis of electroencephalogram activity was done in mice exposed to acute seizures or those developing chronic epileptic activity after status epilepticus to assess the anticonvulsant effects of systemic administration of VX-765. Histological and immunohistochemical analysis of brain tissue was carried out at the end of pharmacological experiments in epileptic mice to evaluate neuropathology, glia activation and IL-1ß expression, and the effect of treatment. Repeated systemic administration of VX-765 significantly reduced chronic epileptic activity in mice in a dose-dependent fashion (12.5-200 mg/kg). This effect was observed at doses ≥ 50 mg/kg, and was reversible with discontinuation of the drug. Maximal drug effect was associated with inhibition of IL-1ß synthesis in activated astrocytes. The same dose regimen of VX-765 also reduced acute seizures in mice and delayed their onset time. These results support a new target system for anticonvulsant pharmacological intervention to control epileptic activity that does not respond to some common anticonvulsant drugs.


Assuntos
Epilepsia/metabolismo , Interleucina-1beta/metabolismo , Convulsões/metabolismo , Ácido 4-Aminobenzoico/administração & dosagem , Animais , Anticonvulsivantes/administração & dosagem , Convulsivantes/toxicidade , Dipeptídeos/administração & dosagem , Relação Dose-Resposta a Droga , Eletroencefalografia , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , para-Aminobenzoatos
20.
J Neurochem ; 115(6): 1445-54, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21044073

RESUMO

Hippocampal excitability and the metabolic glial-neuronal interactions were investigated in 22-week-old mice with motor neuron degeneration (mnd), a model of progressive epilepsy with mental retardation. Mnd mice developed spontaneous spikes in the hippocampus and were more susceptible to kainate-induced seizures compared with control mice. Neuronal hyperexcitability in their hippocampus was confirmed by the selective increase of c-Fos positive nuclei. Glial activation and pro-inflammatory cytokines over-expression were observed in the hippocampus of mnd mice, even in the absence of marked hippocampal neurodegeneration, as suggested by unchanged amounts of neuroactive amino acids and N-acetyl aspartate. Concentration of other amino acids, including GABA and glutamate, was not changed as well. However, ex vivo(13) C magnetic resonance spectroscopy, after simultaneous injection of [1-(13) C]glucose and [1,2-(13) C]acetate, followed by decapitation, showed decreased [1,2-(13) C]GABA formation from hippocampal astrocytic precursors and a marked reduction in [4,5-(13) C]glutamate derived from glutamine. We suggest that astrocyte dysfunction plays a primary role in the pathology and that mnd mice are of value to investigate early pathogenetic mechanism of progressive epilepsy with mental retardation.


Assuntos
Comunicação Celular/fisiologia , Modelos Animais de Doenças , Epilepsia/patologia , Hipocampo/patologia , Deficiência Intelectual/patologia , Neuroglia/patologia , Neurônios/patologia , Convulsões/patologia , Animais , Epilepsia/complicações , Epilepsia/metabolismo , Feminino , Hipocampo/metabolismo , Deficiência Intelectual/complicações , Deficiência Intelectual/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Neurônios/metabolismo , Convulsões/complicações , Convulsões/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA