Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Med Sci Monit ; 25: 8499-8508, 2019 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-31710596

RESUMO

BACKGROUND This study aimed to discover the effect and mechanism of microRNA-27a-3p (miR-27a-3p) in epilepsy. MATERIAL AND METHODS To perform our investigation, in vivo and in vitro models of epilepsy were induced using kainic acid (KA). Expression of miR-27a-3p in the hippocampus of epileptic rats or normal rats or neuronal cells was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Racine score was used to assess seizures in epileptic rats. Cell viability and cell apoptosis were analyzed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay and flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was performed to detect inflammatory factors expression. RESULTS Significantly higher expression of miR-27a-3p in the hippocampus of epileptic rats and in KA-induced neurons was observed. We found that miR-27a-3p inhibitor alleviated seizures in epileptic rats. miR-27a-3p inhibitor also inhibited apoptosis of hippocampal neurons in epileptic rats, promoted Bcl2 expression, and decreased Bax and Caspase3 expression. The results showed that miR-27a-3p inhibitor effectively reduced the expression levels of interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor-alpha (TNF-alpha) in hippocampal tissues of epileptic rats. Dual luciferase reporter assay showed that mitogen-activated protein kinase 4 (MAP2K4) was a direct target of miR-27a-3p. miR-27a-3p inhibitor significantly promoted the cell viability of KA-induced neurons, inhibited cell apoptosis, promoted the expression of Bcl-2, and decreased Bax and Caspase3 expression, and all these changes were abolished by MAP2K4-siRNA co-transfection. CONCLUSIONS Our preliminary findings indicated that miR-27a-3p inhibitor protected against epilepsy-induced inflammatory response and hippocampal neuronal apoptosis by targeting MAP2K4.


Assuntos
Epilepsia/metabolismo , Hipocampo/metabolismo , MAP Quinase Quinase 4/biossíntese , MicroRNAs/metabolismo , Neurônios/metabolismo , Animais , Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Regulação para Baixo , Epilepsia/genética , Epilepsia/patologia , Regulação da Expressão Gênica , Células HEK293 , Hipocampo/patologia , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Ácido Caínico/administração & dosagem , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , MicroRNAs/genética , Neurônios/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Ativação Transcricional , Fator de Necrose Tumoral alfa/metabolismo
2.
Epilepsia ; 60(11): 2314-2324, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31608439

RESUMO

OBJECTIVE: More than one-third of patients with temporal lobe epilepsy (TLE) continue to have seizures despite treatment with antiepileptic drugs, and many experience severe drug-related side effects, illustrating the need for novel therapies. Selective expression of inhibitory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) allows cell-type-specific reduction of neuronal excitability. In this study, we evaluated the effect of chemogenetic suppression of excitatory pyramidal and granule cell neurons of the sclerotic hippocampus in the intrahippocampal mouse model (IHKA) for temporal lobe epilepsy. METHODS: Intrahippocampal IHKA mice were injected with an adeno-associated viral vector carrying the genes for an inhibitory DREADD hM4Di in the sclerotic hippocampus or control vector. Next, animals were treated systemically with different single doses of clozapine-N-oxide (CNO) (1, 3, and 10 mg/kg) and clozapine (0.03 and 0.1 mg/kg) and the effect on spontaneous hippocampal seizures, hippocampal electroencephalography (EEG) power, fast ripples (FRs) and behavior in the open field test was evaluated. Finally, animals received prolonged treatment with clozapine for 3 days and the effect on seizures was monitored. RESULTS: Treatment with both CNO and clozapine resulted in a robust suppression of hippocampal seizures for at least 15 hours only in DREADD-expressing animals. Moreover, total EEG power and the number of FRs were significantly reduced. CNO and/or clozapine had no effects on interictal hippocampal EEG, seizures, or locomotion/anxiety in the open field test in non-DREADD epileptic IHKA mice. Repeated clozapine treatment every 8 hours for 3 days resulted in almost complete seizure suppression in DREADD animals. SIGNIFICANCE: This study shows the potency of chemogenetics to robustly and sustainably suppress spontaneous epileptic seizures and pave the way for an epilepsy therapy in which a systemically administered exogenous drug selectively modulates specific cell types in a seizure network, leading to a potent seizure suppression devoid of the typical drug-related side effects.


Assuntos
Anticonvulsivantes/administração & dosagem , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/prevenção & controle , Convulsões/genética , Convulsões/prevenção & controle , Animais , Clozapina/administração & dosagem , Clozapina/análogos & derivados , Eletroencefalografia/efeitos dos fármacos , Eletroencefalografia/métodos , Epilepsia do Lobo Temporal/fisiopatologia , Vetores Genéticos/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Ácido Caínico/administração & dosagem , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Convulsões/fisiopatologia
3.
FASEB J ; 33(12): 13998-14009, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31618599

RESUMO

Immune changes occur in experimental and clinical epilepsy. Here, we tested the hypothesis that during epileptogenesis and spontaneous recurrent seizures (SRS) an impairment of the endogenous anti-inflammatory pathway glucocorticoid receptor (GR)-annexin A1 (ANXA1) occurs. By administrating exogenous ANXA1, we studied whether pharmacological potentiation of the anti-inflammatory response modifies seizure activity and pathophysiology. We used an in vivo model of temporal lobe epilepsy based on intrahippocampal kainic acid (KA) injection. Video-electroencephalography, molecular biology analyses on brain and peripheral blood samples, and pharmacological investigations were performed in this model. Human epileptic cortices presenting type II focal cortical dysplasia (IIa and b), hippocampi with or without hippocampal sclerosis (HS), and available controls were used to study ANXA1 expression. A decrease of phosphorylated (phospho-) GR and phospho-GR/tot-GR protein expression occurred in the hippocampus during epileptogenesis. Downstream to GR, the anti-inflammatory protein ANXA1 remained at baseline levels while inflammation installed and endured. In peripheral blood, ANXA1 and corticosterone levels showed no significant modifications during disease progression except for an early and transient increase poststatus epilepticus. These results indicate inadequate ANXA1 engagement over time and in these experimental conditions. By analyzing human brain specimens, we found that where significant inflammation exists, the pattern of ANXA1 immunoreactivity was abnormal because the typical perivascular ANXA1 immunoreactivity was reduced. We next asked whether potentiation of the endogenous anti-inflammatory mechanism by ANXA1 administration modifies the disease pathophysiology. Although with varying efficacy, administration of exogenous ANXA1 somewhat reduced the time spent in seizure activity as compared to saline. These results indicate that the anti-inflammatory GR-ANXA1 pathway is defective during experimental seizure progression. The prospect of pharmacologically restoring or potentiating this endogenous anti-inflammatory mechanism as an add-on therapeutic strategy for specific forms of epilepsy is proposed.-Zub, E., Canet, G., Garbelli, R., Blaquiere, M., Rossini, L., Pastori, C., Sheikh, M., Reutelingsperger, C., Klement, W., de Bock, F., Audinat, E., Givalois, L., Solito, E., Marchi, N. The GR-ANXA1 pathway is a pathological player and a candidate target in epilepsy.


Assuntos
Anexina A1/metabolismo , Epilepsia , Receptores de Glucocorticoides/metabolismo , Animais , Anexina A1/genética , Contagem de Células Sanguíneas , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Corticosterona/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipocampo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Ácido Caínico/administração & dosagem , Ácido Caínico/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Glucocorticoides/genética
4.
J Neuroendocrinol ; 31(10): e12783, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31433881

RESUMO

Parental experience imposes neuroplasticity in the hippocampus of females and males. In lactating rat dams, the hippocampus is protected against excitotoxic damage by kainic acid lesioning, although it is still unknown whether paternity can provide such protection to male rodents. To evaluate the protective effects of fatherhood against excitotoxic lesions, we paired male mice with females and co-housed them until the day of parturition (PPD0), when we randomly assigned them to two groups: (i) the pregnancy group (males housed individually overnight and injected i.c.v. with 100 ng per 1 µL of kainic acid or vehicle on PPD1) and (ii) the sire group (males housed with the dam and pups until PPD8, when injected i.c.v. after evaluation of parental behaviour). Individually housed virgin adult male mice formed the control group. Markers of neurodegeneration (NeuN, Fluoro-Jade C) and astrogliosis (glial fibrillary acidic protein) were evaluated in fixed cerebral tissue containing the dorsal CA1, CA3 and CA4 hippocampal subfields. The CA1 subfield did not suffer damage in any of the experimental groups. The sire group exhibited less neurodegeneration and astrogliosis in the CA3 and CA4 subfields compared to their respective controls, independently of the expression of parental behaviour. Western blot analysis was conducted for prolactin (PRL), PRL receptor and related intracellular pathways. Monomeric PRL was lower in the hippocampus of sires in the first week postpartum with a parallel rise of a 48-kDa dimerised isoform compared to virgin controls. The long isoform of PRL receptor did not change, and signal transducer and activator of transcription 5 (STAT5) was not detected in the hippocampus. However, a sustained rise in pAkt, a signalling molecule that participates in cell survival, was observed in the sire group. These results indicate that the hippocampus of sires housed with the dam and pups is less sensitive to neurotoxic injury, which might not be primarily regulated by PRL-STAT5-modulated mechanisms.


Assuntos
Gliose/patologia , Hipocampo/patologia , Ácido Caínico/toxicidade , Degeneração Neural/patologia , Paternidade , Animais , Feminino , Hipocampo/metabolismo , Infusões Intraventriculares , Ácido Caínico/administração & dosagem , Masculino , Camundongos , Degeneração Neural/induzido quimicamente , Comportamento de Nidação , Fosforilação , Prolactina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores da Prolactina/metabolismo , Fator de Transcrição STAT5/metabolismo
5.
Neuropeptides ; 76: 101932, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31227312

RESUMO

Inflammation is an important factor in the pathology of epilepsy with the hallmarks of resident microglia activation and infiltration of circulating monocytes in the damaged area. In the case of recovery and tissue repair, some monocytes change to macrophages (mo-MΦ) to enhance tissue repair. 2-deoxyglucose (2DG) is an analog of glucose capable of protecting the brain, and progranulin is a neurotrophic factor produced mainly by microglia and has an inflammation modulator effect. This study attempted to evaluate if one of the neuroprotective mechanisms of 2-DG is comprised of increasing monocyte-derived macrophages (mo-MΦ) and progranulin production. Status epilepticus (SE) was induced by i.c.v. injection of kainic acid (KA).2DG (125/mg/kg/day) was administered intraperitoneally. Four days later, animals were sacrificed. Their brain sections were then stained with Cresyl violet and Fluoro-Jade B to count the number of necrotic and degenerating neurons in CA3 and Hilus of dentate gyrus of the hippocampus. Lastly, immunohistochemistry was used to detect CD11b + monocyte, macrophage cells, and Progranulin level was evaluated by Western blotting. The histological analysis showed that 2DG can reduce the number of necrotic and degenerating neurons in CA3 and Hilar areas. Following KA administration, a great number of cD11b+ cells with monocyte morphology were observed in the hippocampus. 2DG not only reduced cD11b+ monocyte cells but was able to convert them to cells with the morphology of macrophages (mo-MΦ). 2DG also caused a significant increase in progranulin level in the hippocampus. Because macrophages and microglia are the most important sources of progranulin, it appears that 2DG caused the derivation of monocytes to macrophages and these cells produced progranulin with a subsequent anti-inflammation effect. In summary, it was concluded that 2DG is neuroprotective and probably one of its neuroprotective mechanisms is by modulating monocyte-derived macrophages by progranulin production.


Assuntos
Desoxiglucose/administração & dosagem , Epilepsia do Lobo Temporal/patologia , Epilepsia do Lobo Temporal/prevenção & controle , Hipocampo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Progranulinas/metabolismo , Animais , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/induzido quimicamente , Hipocampo/patologia , Ácido Caínico/administração & dosagem , Macrófagos/patologia , Masculino , Neurônios/patologia , Ratos Wistar
6.
J Neurosci Res ; 97(11): 1378-1392, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31090233

RESUMO

Antiepileptogenic agents that prevent the development of epilepsy following a brain insult remain the holy grail of epilepsy therapeutics. We have employed a label-free proteomic approach that allows quantification of large numbers of brain-expressed proteins in a single analysis in the mouse (male C57BL/6J) kainate (KA) model of epileptogenesis. In addition, we have incorporated two putative antiepileptogenic drugs, postsynaptic density protein-95 blocking peptide (PSD95BP or Tat-NR2B9c) and a highly selective inducible nitric oxide synthase inhibitor, 1400W, to give an insight into how such agents might ameliorate epileptogenesis. The test drugs were administered after the induction of status epilepticus (SE) and the animals were euthanized at 7 days, their hippocampi removed, and subjected to LC-MS/MS analysis. A total of 2,579 proteins were identified; their normalized abundance was compared between treatment groups using ANOVA, with correction for multiple testing by false discovery rate. Significantly altered proteins were subjected to gene ontology and KEGG pathway enrichment analyses. KA-induced SE was most robustly associated with an alteration in the abundance of proteins involved in neuroinflammation, including heat shock protein beta-1 (HSP27), glial fibrillary acidic protein, and CD44 antigen. Treatment with PSD95BP or 1400W moderated the abundance of several of these proteins plus that of secretogranin and Src substrate cortactin. Pathway analysis identified the glutamatergic synapse as a key target for both drugs. Our observations require validation in a larger-scale investigation, with candidate proteins explored in more detail. Nevertheless, this study has identified several mechanisms by which epilepsy might develop and several targets for novel drug development. OPEN PRACTICES: This article has been awarded Open Data. All materials and data are publicly accessible as supporting information. Learn more about the Open Practices badges from the Center for Open Science: https://osf.io/tvyxz/wiki.


Assuntos
Amidinas/administração & dosagem , Anticonvulsivantes/administração & dosagem , Benzilaminas/administração & dosagem , Epilepsia/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Peptídeos/administração & dosagem , Animais , Epilepsia/induzido quimicamente , Ácido Caínico/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL , Proteômica , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/metabolismo
7.
Eur J Neurosci ; 49(2): 215-231, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30362615

RESUMO

The secondary phase of spinal cord injury arising after the primary lesion largely extends the damage severity with delayed negative consequences for sensory-motor pathways. It is, therefore, important to find out if enhancing intrinsic mechanisms of neuroprotection can spare motoneurons that are very vulnerable cells. This issue was investigated with an in vitro model of rat spinal cord excitotoxicity monitored for up to 24 hr after the primary injury evoked by kainate. This study sought to pharmacologically boost the expression of heat shock proteins (HSP) to protect spinal motoneurons using celastrol to investigate if the rat spinal cord can upregulate HSP as neuroprotective mechanism. Despite its narrow range of drug safety in vitro, celastrol was not toxic to the rat spinal cord at 0.75 µM concentration and enhanced the expression of HSP70 by motoneurons. When celastrol was applied either before or after kainate, the number of dead motoneurons was significantly decreased and the nuclear localization of the cell death biomarker AIF strongly inhibited. Nevertheless, electrophysiological recording showed that protection of lumbar motor networks by celastrol was rather limited as reflex activity was impaired and fictive locomotion largely depressed, suggesting that functional deficit persisted, though the networks could express slow rhythmic oscillations. While our data do not exclude further recovery at later times beyond the experimental observations, the present results indicate that the upregulated expression of HSP in the aftermath of acute injury may be an interesting avenue for early protection of spinal motoneurons.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Neurônios Motores/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Triterpenos/administração & dosagem , Animais , Animais Recém-Nascidos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Ácido Caínico/administração & dosagem , Locomoção/efeitos dos fármacos , Masculino , Triterpenos Pentacíclicos , Ratos Wistar , Medula Espinal/efeitos dos fármacos , Traumatismos da Medula Espinal/induzido quimicamente
8.
Neurosci Res ; 130: 47-55, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28807642

RESUMO

Current studies have indicated that apoptotic and autophagic signaling pathways are triggered by epileptic seizures, but the precise roles of these processes in epilepsy-induced neuronal loss remain unclear. Identifying a concrete molecular mechanism may help researchers develop relevant epilepsy therapies that are more effective than existing treatments. Autophagy is a type of conserved degradation that contributes to cellular homeostasis. The involved signaling pathways allow us to observe alterations in autophagy and apoptosis during epileptic seizures over time. This study investigated the time-dependent changes in autophagy, apoptosis and neuronal morphology in developing brain of epilepsy model rats. At 48h after epileptic seizure onset, the number of neurons in neocortex decreased, and the number of apoptotic cells in neocortex increased. The ratio of microtubule-associated protein 1 light chain 3 (LC3) II to LC3 I and Beclin-1 protein levels increased from 12h to 48h after epileptic seizure onset. P62 protein and Bcl-2 protein levels decreased from 24h to 48h after epileptic seizure onset. The changes in the levels of these autophagy and apoptosis markers indicate that autophagy starts before apoptosis in rats with epilepsy, demonstrating a potential role of autophagy in epilepsy-induced neuronal loss in developing brain.


Assuntos
Apoptose , Autofagia , Epilepsia/metabolismo , Neocórtex/metabolismo , Transdução de Sinais , Animais , Proteína Beclina-1/metabolismo , Epilepsia/induzido quimicamente , Epilepsia/patologia , Ácido Caínico/administração & dosagem , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Neocórtex/crescimento & desenvolvimento , Neocórtex/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/metabolismo
9.
Behav Brain Res ; 326: 103-111, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28274653

RESUMO

The lateral wings subnucleus of the dorsal raphe nucleus (lwDR) has been implicated in the modulation of panic-like behaviors, such as escape. Infusion of non- excitotoxic doses of the excitatory amino acid kainic acid into this subnucleus promptly evokes a vigorous escape response. In addition, rats exposed to panic-inducing situations show an increase in Fos protein expression in neurons within the lwDR. In the present study, we first investigated whether key structures associated with the mediation of escape behavior are recruited after chemical stimulation of the lwDR with kainic acid. We next investigated whether the infusion of the GABAA receptor antagonist bicuculline into the lwDR also evoked escape responses measured both in a circular arena and in the rat elevated T-maze. The effects of bicuculline in the circular arena were compared to those caused by the infusion of this antagonist into the ventrolateral periaqueductal gray (vlPAG), an area in close vicinity to the lwDR. The results showed that kainic acid infusion into the lwDR increased Fos protein immunostaining in brain structures deeply involved in panic-like defensive behaviors, such as the periaqueductal gray and hypothalamus, but not the amygdala. As observed with kainic acid, bicuculline evoked a pronounced escape response in the circular arena when microinjected in the lwDR, but not in the vlPAG. The escape-promoting effect of bicuculline in the lwDR was also evidenced in the elevated T-maze. These findings strength the view that dysfunction in mechanisms controlling escape in the lwDR is critically implicated in the pathophysiology of panic disorder.


Assuntos
Comportamento Animal/efeitos dos fármacos , Bicuculina/farmacologia , Núcleo Dorsal da Rafe/efeitos dos fármacos , Reação de Fuga/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Ácido Caínico/farmacologia , Pânico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Animais , Bicuculina/administração & dosagem , Agonistas de Aminoácidos Excitatórios/administração & dosagem , Antagonistas de Receptores de GABA-A/administração & dosagem , Imuno-Histoquímica , Ácido Caínico/administração & dosagem , Masculino , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Ratos , Ratos Wistar , Estimulação Química
10.
Neuroreport ; 28(4): 200-207, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28169952

RESUMO

Recent studies suggest that epileptic seizures might be facilitated by immune reactions in the brain. However, it remains unclear whether inflammatory reactions could result in acute and chronic epileptic seizures in vivo. In the present study, we investigated whether a hippocampal infusion of lipopolysaccharide (LPS) can induce epileptic seizures in rats. We also aimed to elucidate the molecular mechanisms that underpin this phenomenon. Male Wistar rats received a 1.5 µl infusion of either LPS (5 µg/µl), normal saline, or kainate (KA) (0.4 µg/µl) into the right dorsal hippocampus. No seizures were observed in the normal saline group and associated electroencephalograms showed basic rhythms. Various grades of seizures were observed in rats from the LPS and KA groups, and electroencephalograms were characterized by seizure activities (sharp waves or spikes). In the chronic phase (8 weeks after injection), astrocytes proliferated and the number of neurons decreased in the hippocampus of the rats in the LPS and KA groups. When assessed at the acute phase (within 6 h after injection), the expression of interleukin 1ß, tumor necrosis factor α, and neuronal nitric oxide synthase was significantly increased in the hippocampus of the LPS groups. The results of the present study indicate that hippocampal infusion of LPS can activate innate immune responses of glial cells, thereby inducing epileptic seizures and hippocampal sclerosis in rats.


Assuntos
Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Lipopolissacarídeos/administração & dosagem , Convulsões/imunologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Agonistas de Aminoácidos Excitatórios/administração & dosagem , Hipocampo/patologia , Hipocampo/fisiopatologia , Imunidade Inata , Ácido Caínico/administração & dosagem , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos Wistar , Convulsões/induzido quimicamente
11.
Neurosci Lett ; 622: 30-6, 2016 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-27095588

RESUMO

In rodent models of epilepsy, EEG implantation surgery is an essential modality to evaluate electrographic seizures. The inflammatory consequences of EEG electrode-implantation and their resultant effects on seizure susceptibility are unclear. We evaluated electrode-implantation in a two-hit model of epileptogenesis in C57BL/6 mice that included brief, recurrent febrile seizures (FS) at P14 and kainic acid induced seizures (KA-SZ) at P28. During KA-SZ, latencies to first electrographic and behavioral seizures, seizure severity, and KA dose sensitivity were measured. Mice that received subdural screw electrode implants at P25 for EEG monitoring at P28 had significantly shorter latencies to seizures than sham mice, regardless of early life seizure experience. Electrode-implanted mice were sensitive to low dose KA as shown by high mortality rate at KA doses above 10mg/kg. We then directly compared electrode-implantation and KA-SZ in seizure naive CX3CR1(GFP/+) transgenic C57BL/6 mice, wherein microglia express green fluorescent protein (GFP), to determine if microglia activation related to surgery was associated with the increased seizure susceptibility in electrode-implanted mice from the two-hit model. Hippocampal microglia activation, as demonstrated by percent area GFP signal and GFP positive cell counts, prior to seizures was indistinguishable between electrode-implanted mice and controls, but was significantly greater in electrode-implanted mice following seizures. Electrode-implantation had a confounding priming effect on the inflammatory response to subsequent seizures.


Assuntos
Epilepsia do Lobo Temporal/cirurgia , Animais , Relação Dose-Resposta a Droga , Eletrodos Implantados/efeitos adversos , Eletroencefalografia , Epilepsia do Lobo Temporal/etiologia , Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/patologia , Hipocampo/cirurgia , Hipertermia Induzida , Inflamação/etiologia , Inflamação/patologia , Ácido Caínico/administração & dosagem , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia
12.
Elife ; 52016 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-27083045

RESUMO

Status epilepticus (SE) is a common neurological emergency for which new treatments are needed. In vitro studies suggest a novel approach to controlling seizures in SE: acute inhibition of estrogen synthesis in the brain. Here, we show in rats that systemic administration of an aromatase (estrogen synthase) inhibitor after seizure onset strongly suppresses both electrographic and behavioral seizures induced by kainic acid (KA). We found that KA-induced SE stimulates synthesis of estradiol (E2) in the hippocampus, a brain region commonly involved in seizures and where E2 is known to acutely promote neural activity. Hippocampal E2 levels were higher in rats experiencing more severe seizures. Consistent with a seizure-promoting effect of hippocampal estrogen synthesis, intra-hippocampal aromatase inhibition also suppressed seizures. These results reveal neurosteroid estrogen synthesis as a previously unknown factor in the escalation of seizures and suggest that acute administration of aromatase inhibitors may be an effective treatment for SE.


Assuntos
Inibidores da Aromatase/administração & dosagem , Estradiol/metabolismo , Neurotransmissores/metabolismo , Estado Epiléptico/tratamento farmacológico , Animais , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Inibição Psicológica , Ácido Caínico/administração & dosagem , Ratos , Estado Epiléptico/induzido quimicamente , Resultado do Tratamento
13.
eNeuro ; 3(1)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27022627

RESUMO

Epilepsy is a neurological disorder defined by the presence of seizure activity, manifest both behaviorally and as abnormal activity in neuronal networks. An established model to study the disorder in rodents is the systemic injection of kainic acid, an excitatory neurotoxin that at low doses quickly induces behavioral and electrophysiological seizures. Although the CA3 region of the hippocampus has been suggested to be crucial for kainic acid-induced seizure, because of its strong expression of kainate glutamate receptors and its high degree of recurrent connectivity, the precise role of excitatory transmission in CA3 in the generation of seizure and the accompanying increase in neuronal oscillations remains largely untested. Here we use transgenic mice in which CA3 pyramidal cell synaptic transmission can be inducibly silenced in the adult to demonstrate CA3 excitatory output is required for both the generation of epileptiform oscillatory activity and the progression of behavioral seizures.


Assuntos
Região CA3 Hipocampal/fisiopatologia , Modelos Animais de Doenças , Ácido Caínico/administração & dosagem , Células Piramidais/fisiologia , Convulsões/fisiopatologia , Animais , Ondas Encefálicas/efeitos dos fármacos , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Células Piramidais/efeitos dos fármacos , Convulsões/induzido quimicamente , Transmissão Sináptica/efeitos dos fármacos , Toxina Tetânica/genética
14.
Arch Pharm Res ; 39(5): 660-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26987339

RESUMO

In the present study, we characterized the expression and role of forkhead box O (FoxO3a) in kainic acid (KA)-induced hippocampal neuronal cell death. FoxO3a and pFoxO3a expression in the CA1, CA2, and dentate gyrus regions in the hippocampus increased 0.5 and 1 h after intracerebroventricular administration of KA. In addition, both FoxO3a and pFoxO3a expression in the hippocampal CA3 region increased significantly and equally for 1 h but decreased gradually for 24 h after KA administration. In particular, the KA-induced increases in FoxO3a and pFoxO3a expression in the hippocampal CA3 region were inhibited by pretreatment with the N-methyl-D-aspartate (NMDA) receptor antagonist (MK-801, dizocilpine, 1 µg/5 µl) or a non-NMDA receptor antagonist (CNQX, 6-cyano-7-nitroquinoxaline-2,3-dione, 0.5 µg/5 µl). Furthermore, dizocilpine and CNQX produced a neuroprotective effect against KA-induced neuronal death in the CA3 region of the hippocampus. Our results suggest that FoxO3a and pFoxO3 expression is upregulated by KA. Both FoxO3a and pFoxO3a expression appear to be responsible for KA-induced neuronal death in the CA3 region of the hippocampus.


Assuntos
Proteína Forkhead Box O3/biossíntese , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ácido Caínico/toxicidade , Neurônios/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/metabolismo , Região CA3 Hipocampal/patologia , Morte Celular/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Imuno-Histoquímica , Injeções Intraventriculares , Ácido Caínico/administração & dosagem , Masculino , Camundongos Endogâmicos ICR , Neurônios/metabolismo , Neurônios/ultraestrutura , Fosforilação , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Fatores de Tempo , Regulação para Cima
15.
Neuroreport ; 27(7): 501-7, 2016 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-26999361

RESUMO

Lidocaine, the most commonly used local anesthetic, inhibits glutamate release from nerve terminals. Given the involvement of glutamate neurotoxicity in the pathogenesis of various neurological disorders, this study investigated the role of lidocaine in hippocampal neuronal death and inflammatory events induced by an i.p. injection of kainic acid (KA) (15 mg/kg), a glutamate analog. The results showed that KA significantly led to neuronal death in the CA3 pyramidal layers of the hippocampus and this effect was attenuated by the systemic administration of lidocaine (0.8 or 4 mg/kg, i.p.) 30 min before KA injection. Moreover, KA-induced microglia activation and gene expression of proinflammatory cytokines, namely, interleukin-1ß, interleukin-6, and tumor necrosis factor-α, in the hippocampus were reduced by the lidocaine pretreatment. Altogether, the results suggest that lidocaine can effectively treat glutamate excitotoxicity-related brain disorders.


Assuntos
Anti-Inflamatórios/administração & dosagem , Região CA3 Hipocampal/efeitos dos fármacos , Ácido Caínico/administração & dosagem , Lidocaína/administração & dosagem , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Animais , Região CA3 Hipocampal/metabolismo , Morte Celular/efeitos dos fármacos , Encefalite/metabolismo , Masculino , Microglia/efeitos dos fármacos , Microglia/fisiologia , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
16.
Brain Struct Funct ; 221(4): 2061-74, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-25772509

RESUMO

The cannabinoid type 1 receptor (Cnr1, CB1R) mediates a plethora of physiological functions in the central nervous system as a presynaptic modulator of neurotransmitter release. The recently identified cannabinoid receptor-interacting protein 1a (Cnrip1a, CRIP1a) binds to the C-terminal domain of CB1R, a region known to be important for receptor desensitization and internalization. Evidence that CRIP1a and CB1R interact in vivo has been reported, but the neuroanatomical distribution of CRIP1a is unknown. Moreover, while alterations of hippocampal CRIP1a levels following limbic seizures indicate a role in controlling excessive neuronal activity, the physiological function of CRIP1a in vivo has not been investigated. In this study, we analyzed the spatial distribution of CRIP1a in the hippocampus and examined CRIP1a as a potential modulator of CB1R signaling. We found that Cnrip1a mRNA is co-expressed with Cnr1 mRNA in pyramidal neurons and interneurons of the hippocampal formation. CRIP1a protein profiles were largely segregated from CB1R profiles in mossy cell terminals but not in hippocampal CA1 region. CB1R activation induced relocalization to close proximity with CRIP1a. Adeno-associated virus-mediated overexpression of CRIP1a specifically in the hippocampus revealed that CRIP1a modulates CB1R activity by enhancing cannabinoid-induced G protein activation. CRIP1a overexpression extended the depression of excitatory currents by cannabinoids in pyramidal neurons of the hippocampus and diminished the severity of chemically induced acute epileptiform seizures. Collectively, our data indicate that CRIP1a enhances hippocampal CB1R signaling in vivo.


Assuntos
Proteínas de Transporte/metabolismo , Hipocampo/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais , Animais , Proteínas de Transporte/fisiologia , Dronabinol/administração & dosagem , Dronabinol/análogos & derivados , Potenciais Pós-Sinápticos Excitadores , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hipocampo/fisiologia , Ácido Caínico/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Piramidais/fisiologia , RNA Mensageiro/metabolismo , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/fisiologia , Convulsões/induzido quimicamente , Convulsões/fisiopatologia
17.
Cereb Cortex ; 26(4): 1619-1633, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25595182

RESUMO

The RNase Dicer is essential for the maturation of most microRNAs, a molecular system that plays an essential role in fine-tuning gene expression. To gain molecular insight into the role of Dicer and the microRNA system in brain function, we conducted 2 complementary RNA-seq screens in the hippocampus of inducible forebrain-restricted Dicer1 mutants aimed at identifying the microRNAs primarily affected by Dicer loss and their targets, respectively. Functional genomics analyses predicted the main biological processes and phenotypes associated with impaired microRNA maturation, including categories related to microRNA biology, signal transduction, seizures, and synaptic transmission and plasticity. Consistent with these predictions, we found that, soon after recombination, Dicer-deficient mice exhibited an exaggerated seizure response, enhanced induction of immediate early genes in response to different stimuli, stronger and more stable fear memory, hyperphagia, and increased excitability of CA1 pyramidal neurons. In the long term, we also observed slow and progressive excitotoxic neurodegeneration. Overall, our results indicate that interfering with microRNA biogenesis causes an increase in neuronal responsiveness and disrupts homeostatic mechanisms that protect the neuron against overactivation, which may explain both the initial and late phenotypes associated with the loss of Dicer in excitatory neurons.


Assuntos
RNA Helicases DEAD-box/genética , Memória/fisiologia , MicroRNAs/biossíntese , Neurônios/fisiologia , Prosencéfalo/fisiopatologia , Ribonuclease III/genética , Convulsões/metabolismo , Potenciais de Ação/genética , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiopatologia , Condicionamento Clássico , Medo/fisiologia , Feminino , Hiperfagia/genética , Hiperfagia/metabolismo , Ácido Caínico/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , Plasticidade Neuronal , Neurônios/metabolismo , Fenótipo , Prosencéfalo/metabolismo , Convulsões/induzido quimicamente , Convulsões/genética , Análise de Sequência de RNA
18.
Neurobiol Dis ; 86: 52-61, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26607785

RESUMO

Although novel treatment strategies based on the gene therapy approach for epilepsy has been encouraging, there is still a gap in demonstrating a proof-of-concept in a clinically relevant animal model and study design. In the present study, a conceptually novel framework reflecting a plausible clinical trial for gene therapy of temporal lobe epilepsy was explored: We investigated (i) whether the post intrahippocampal kainate-induced status epilepticus (SE) model of chronic epilepsy in rats could be clinically relevant; and (ii) whether a translationally designed neuropeptide Y (NPY)/Y2 receptor-based gene therapy approach targeting only the seizure-generating focus unilaterally can decrease seizure frequency in this chronic model of epilepsy. Our data suggest that the intrahippocampal kainate model resembles the disease development of human chronic mesial temporal lobe epilepsy (mTLE): (i) spontaneous seizures originate in the sclerotic hippocampus; (ii) only a part of the animals develops chronic epilepsy; (iii) animals show largely variable seizure frequency that (iv) tends to progressively increase over time. Despite significant hippocampal degeneration caused by the kainate injection, the use of MRI allowed targeting the recombinant adeno-associated viral (rAAV) vectors encoding NPY and Y2 receptor genes to the remaining dorsal and ventral hippocampal areas ipsilateral to the kainate injection. Continuous video-EEG monitoring demonstrated not only prevention of the progressive increase in seizure frequency in rAAV-NPY/Y2 treated animals as compared to the controls, but even 45% decrease of seizure frequency in 80% of the epileptic animals. This translationally designed study in a clinically relevant model of epilepsy suggests that simultaneous overexpression of NPY and Y2 receptors unilaterally in the seizure focus is a relevant and promising approach that can be further validated in more extensive preclinical studies to develop a future treatment strategy for severe, often pharmacoresistant focal epilepsy cases that cannot be offered alternative therapeutic options.


Assuntos
Córtex Cerebral/fisiopatologia , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/terapia , Terapia Genética/métodos , Receptores de Neuropeptídeo Y/genética , Animais , Córtex Cerebral/efeitos dos fármacos , Dependovirus/genética , Eletroencefalografia , Epilepsia do Lobo Temporal/induzido quimicamente , Vetores Genéticos/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Ácido Caínico/administração & dosagem , Masculino , Ratos , Ratos Wistar , Pesquisa Translacional Biomédica
19.
J Neurol Sci ; 358(1-2): 390-7, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26453128

RESUMO

Duloxetine (DXT), a potent serotonin/norepinephrine reuptake inhibitor, is widely used in the treatment of major depressive disorder. In the present study, we examined the effects of DXT treatment on seizure behavior and excitotoxic neuronal damage in the mouse hippocampal CA3 region following intraperitoneal kainic acid (KA) injection. DXT treatment showed no effect on KA-induced behavioral seizure activity. However, treatment with 10mg/kg DXT reduced KA-induced neuronal death in the hippocampal CA3 region at 72h after KA administration, and treatment with 20 and 40mg/kg DXT showed a noticeable neuroprotection in the hippocampal CA3 region after KA injection. In addition, KA-induced activations of microglia and astrocytes as well as KA-induced increases of TNF-α and IL-1ß levels were also suppressed by DXT treatment. These results indicate that DXT displays the neuroprotective effect against KA-induced excitotoxic neuronal death through anti-inflammatory action.


Assuntos
Astrócitos/efeitos dos fármacos , Região CA3 Hipocampal/efeitos dos fármacos , Cloridrato de Duloxetina/farmacologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Interleucina-1beta/efeitos dos fármacos , Ácido Caínico/toxicidade , Microglia/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Inibidores da Recaptação de Serotonina e Norepinefrina/farmacologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Cloridrato de Duloxetina/administração & dosagem , Agonistas de Aminoácidos Excitatórios/administração & dosagem , Ácido Caínico/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos ICR , Inibidores da Recaptação de Serotonina e Norepinefrina/administração & dosagem
20.
Brain Struct Funct ; 220(3): 1759-76, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24706067

RESUMO

Excitotoxicity underlies neuronal death in many neuropathological disorders, such as Alzheimer's disease and multiple sclerosis. In murine models of these diseases, disruption of CX3CR1 signaling has thus far generated data either in favor or against a neuroprotective role of this crucial regulator of microglia and monocyte functions. In this study, we investigated the recruitment of circulating PU.1-expressing cells following sterile excitotoxicity and delineated the CX3CR1-dependent neuroprotective functions of circulating monocytes versus that of microglia in this context. WT, Cx3cr1-deficient and chimeric mice were subjected to a sterile excitotoxic insult via an intrastriatal injection of kainic acid (KA), a conformational analog of glutamate. Following KA administration, circulating monocytes physiologically engrafted the brain and selectively accumulated in the vicinity of excitotoxic lesions where they gave rise to activated macrophages depicting strong Iba1 and CD68 immunoreactivity 7 days post-injury. Monocyte-derived macrophages completely vanished upon recovery and did thus not permanently seed the brain. Furthermore, Cx3cr1 deletion significantly exacerbated neuronal death, behavioral deficits and activation of microglia cells following sterile excitotoxicity. Cx3cr1 disruption also markedly altered the blood levels of patrolling monocytes 24 h after KA administration. The specific elimination of patrolling monocytes using Nr4a1(-/-) chimeric mice conditioned with chemotherapy provided direct evidence that these circulating monocytes are essential for neuroprotection. Taken together, these data support a beneficial role of CX3CR1 signaling during excitotoxicity and highlight a novel and pivotal role of patrolling monocytes in neuroprotection. These findings open new research and therapeutic avenues for neuropathological disorders implicating excitotoxicity.


Assuntos
Corpo Estriado/efeitos dos fármacos , Ácido Caínico/toxicidade , Microglia/citologia , Monócitos/citologia , Neuroproteção/efeitos dos fármacos , Receptores de Quimiocinas/deficiência , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Receptor 1 de Quimiocina CX3C , Proteínas de Ligação ao Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Ácido Caínico/administração & dosagem , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microinjeções/métodos , Monócitos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/deficiência , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Receptores de Quimiocinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA