Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biomolecules ; 14(3)2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38540798

RESUMEN

Although granule cell dispersion (GCD) in the hippocampus is known to be an important feature associated with epileptic seizures in temporal lobe epilepsy (TLE), the endogenous molecules that regulate GCD are largely unknown. In the present study, we have examined whether there is any change in AEG-1 expression in the hippocampus of a kainic acid (KA)-induced mouse model of TLE. In addition, we have investigated whether the modulation of astrocyte elevated gene-1 (AEG-1) expression in the dentate gyrus (DG) by intracranial injection of adeno-associated virus 1 (AAV1) influences pathological phenotypes such as GCD formation and seizure susceptibility in a KA-treated mouse. We have identified that the protein expression of AEG-1 is upregulated in the DG of a KA-induced mouse model of TLE. We further demonstrated that AEG-1 upregulation by AAV1 delivery in the DG-induced anticonvulsant activities such as the delay of seizure onset and inhibition of spontaneous recurrent seizures (SRS) through GCD suppression in the mouse model of TLE, while the inhibition of AEG-1 expression increased susceptibility to seizures. The present observations suggest that AEG-1 is a potent regulator of GCD formation and seizure development associated with TLE, and the significant induction of AEG-1 in the DG may have therapeutic potential against epilepsy.


Asunto(s)
Epilepsia del Lóbulo Temporal , Epilepsia , Animales , Ratones , Astrocitos/metabolismo , Giro Dentado/metabolismo , Epilepsia/metabolismo , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Hipocampo/metabolismo , Ácido Kaínico/efectos adversos , Ácido Kaínico/metabolismo , Convulsiones/inducido químicamente , Convulsiones/genética , Convulsiones/metabolismo
2.
Eur Surg Res ; 64(1): 89-107, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35073547

RESUMEN

INTRODUCTION: Chronic epilepsy models require neurosurgical procedures including depth electrode implants. The intrahippocampal kainate model is a frequently used chronic paradigm, which is based on chemoconvulsant administration and status epilepticus induction during the surgical procedure. This experimental approach raises the question of the extent to which this approach affects postsurgical recovery. In addition to the short- and long-term impact of the surgical intervention, a potential impact of highly frequent electrographic seizure events needs to be considered in the context of severity assessment. METHODS: Various behavioral, biochemical, and telemetric parameters were analyzed in four experimental groups of mice: 1st naive, 2nd with transmitter implants, 3rd with transmitter and electrode implants, and 4th with transmitter implants, electrode implants, and kainate-induced status epilepticus. RESULTS: During the early postsurgical phase, transmitter implants caused a transient impact on Mouse Grimace scores and intragroup increase of fecal corticosterone metabolites. Additional craniotomy was associated with an influence on total heart rate variability and fecal corticosterone metabolites. Heart rate and Irwin score increases as well as a prolonged increase in Mouse Grimace scores pointed to an added burden related to the induction of a nonconvulsive status epilepticus. Data from the chronic phase argued against a relevant influence of frequent electrographic seizures on behavioral patterns, fecal corticosterone metabolites, heart rate, and its variability. However, Irwin scores indicated long-term changes in some animals with increased reactivity, body tone, and Straub tail. Interestingly, selected behavioral and telemetric data from the early post-status epilepticus phase correlated with the frequency of electrographic seizure events in the chronic phase. CONCLUSION: In conclusion, our findings argue against the pronounced impact of highly frequent electrographic seizures on the well-being of mice. However, an increased level of nervousness in a subgroup of animals should be considered for handling procedures and refinement measures. In the early postsurgical phase, several parameters indicate an influence of the interventions with evidence that the nonconvulsive status epilepticus can negatively affect the recovery. Thus, the development and validation of refinement efforts should focus on this experimental phase. Finally, the datasets suggest that simple readout parameters may predict the long-term consequences of the epileptogenic insult. Respective biomarker candidates require further validation in the follow-up studies in models with subgroups of animals with or without epilepsy development.


Asunto(s)
Epilepsia , Estado Epiléptico , Ratones , Animales , Ácido Kaínico/efectos adversos , Corticosterona , Convulsiones/inducido químicamente , Estado Epiléptico/inducido químicamente , Modelos Animales de Enfermedad
3.
Food Funct ; 13(20): 10401-10414, 2022 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-36148811

RESUMEN

Rutin, a naturally derived flavonoid molecule with known neuroprotective properties, has been demonstrated to have anticonvulsive potential, but the mechanism of this effect is still unclear. The current study aimed to investigate the probable antiseizure mechanisms of rutin in rats using the kainic acid (KA) seizure model. Rutin (50 and 100 mg kg-1) and carbamazepine (100 mg kg-1) were administered daily by oral gavage for 7 days before KA (15 mg kg-1) intraperitoneal (i.p.) injection. Seizure behavior, neuronal cell death, glutamate concentration, excitatory amino acid transporters (EAATs), glutamine synthetase (GS), glutaminase, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits GluA1 and GluA2, N-methyl-D-aspartate (NMDA) receptor subunits GluN2A and GluN2B, activated astrocytes, and inflammatory and anti-inflammatory molecules in the hippocampus were evaluated. Supplementation with rutin attenuated seizure severity in KA-treated rats and reversed KA-induced neuronal loss and glutamate elevation in the hippocampus. Decreased glutaminase and GluN2B, and increased EAATs, GS, GluA1, GluA2 and GluN2A were observed with rutin administration. Rutin pretreatment also suppressed activated astrocytes, downregulated the protein levels of inflammatory molecules [interleukin-1ß (IL-1ß), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), high mobility group Box 1 (HMGB1), interleukin-1 receptor 1 (IL-1R1), and Toll-like receptor-4 (TLR-4)] and upregulated anti-inflammatory molecule interleukin-10 (IL-10) protein expression. Taken together, the results indicate that the preventive treatment of rats with rutin attenuated KA-induced seizures and neuronal loss by decreasing glutamatergic hyperactivity and suppressing the IL-1R1/TLR4-related neuroinflammatory cascade.


Asunto(s)
Proteína HMGB1 , Ácido Kaínico , Sistemas de Transporte de Aminoácidos , Animales , Antiinflamatorios/farmacología , Carbamazepina , Glutamato-Amoníaco Ligasa/metabolismo , Glutamato-Amoníaco Ligasa/farmacología , Ácido Glutámico/metabolismo , Glutaminasa/genética , Glutaminasa/metabolismo , Glutaminasa/farmacología , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Hipocampo/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ácido Kaínico/efectos adversos , N-Metilaspartato/efectos adversos , N-Metilaspartato/metabolismo , Ratas , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/uso terapéutico , Rutina/metabolismo , Rutina/farmacología , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/efectos adversos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
4.
Int J Mol Sci ; 23(8)2022 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-35456948

RESUMEN

Current anti-seizure drugs fail to control approximately 30% of epilepsies. Therefore, there is a need to develop more effective anti-seizure drugs, and medicinal plants provide an attractive source for new compounds. This study aimed to evaluate the possible anti-seizure and neuroprotective effects of neferine, an alkaloid from the lotus seed embryos of Nelumbo nucifera, in a kainic acid (KA)-induced seizure rat model and its underlying mechanisms. Rats were intraperitoneally (i.p.) administrated neferine (10 and 50 mg/kg) 30 min before KA injection (15 mg/kg, i.p.). Neferine pretreatment increased seizure latency and reduced seizure scores, prevented glutamate elevation and neuronal loss, and increased presynaptic protein synaptophysin and postsynaptic density protein 95 expression in the hippocampi of rats with KA. Neferine pretreatment also decreased glial cell activation and proinflammatory cytokine (interleukin-1ß, interleukin-6, tumor necrosis factor-α) expression in the hippocampi of rats with KA. In addition, NOD-like receptor 3 (NLRP3) inflammasome, caspase-1, and interleukin-18 expression levels were decreased in the hippocampi of seizure rats pretreated with neferine. These results indicated that neferine reduced seizure severity, exerted neuroprotective effects, and ameliorated neuroinflammation in the hippocampi of KA-treated rats, possibly by inhibiting NLRP3 inflammasome activation and decreasing inflammatory cytokine secretion. Our findings highlight the potential of neferine as a therapeutic option in the treatment of epilepsy.


Asunto(s)
Alcaloides , Antineoplásicos , Bencilisoquinolinas , Fármacos Neuroprotectores , Alcaloides/farmacología , Alcaloides/uso terapéutico , Animales , Bencilisoquinolinas/farmacología , Bencilisoquinolinas/uso terapéutico , Citocinas/metabolismo , Inflamasomas/metabolismo , Ácido Kaínico/efectos adversos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Semillas/metabolismo , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico
5.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34948040

RESUMEN

Epilepsy is a complex disorder affecting the central nervous system and is characterised by spontaneously recurring seizures (SRSs). Epileptic patients undergo symptomatic pharmacological treatments, however, in 30% of cases, they are ineffective, mostly in patients with temporal lobe epilepsy. Therefore, there is a need for developing novel treatment strategies. Transplantation of cells releasing γ-aminobutyric acid (GABA) could be used to counteract the imbalance between excitation and inhibition within epileptic neuronal networks. We generated GABAergic interneuron precursors from human embryonic stem cells (hESCs) and grafted them in the hippocampi of rats developing chronic SRSs after kainic acid-induced status epilepticus. Using whole-cell patch-clamp recordings, we characterised the maturation of the grafted cells into functional GABAergic interneurons in the host brain, and we confirmed the presence of functional inhibitory synaptic connections from grafted cells onto the host neurons. Moreover, optogenetic stimulation of grafted hESC-derived interneurons reduced the rate of epileptiform discharges in vitro. We also observed decreased SRS frequency and total time spent in SRSs in these animals in vivo as compared to non-grafted controls. These data represent a proof-of-concept that hESC-derived GABAergic neurons can exert a therapeutic effect on epileptic animals presumably through establishing inhibitory synapses with host neurons.


Asunto(s)
Interneuronas/citología , Ácido Kaínico/efectos adversos , Convulsiones/terapia , Estado Epiléptico/terapia , Trasplante de Células Madre/métodos , Ácido gamma-Aminobutírico/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Interneuronas/metabolismo , Masculino , Ratas , Recurrencia , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Convulsiones/patología , Estado Epiléptico/inducido químicamente , Estado Epiléptico/metabolismo , Estado Epiléptico/patología , Células Madre/citología , Células Madre/metabolismo
6.
Sci Rep ; 10(1): 3760, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111960

RESUMEN

Epilepsy is a widespread neurological disease characterized by abnormal neuronal activity resulting in recurrent seizures. There is mounting evidence that a circadian system disruption, involving clock genes and their downstream transcriptional regulators, is associated with epilepsy. In this study, we characterized the hippocampal expression of clock genes and PAR bZIP transcription factors (TFs) in a mouse model of temporal lobe epilepsy induced by intrahippocampal injection of kainic acid (KA). The expression of PAR bZIP TFs was significantly altered following KA injection as well as in other rodent models of acquired epilepsy. Although the PAR bZIP TFs are regulated by proinflammatory cytokines in peripheral tissues, we discovered that the regulation of their expression is inflammation-independent in hippocampal tissue and rather mediated by clock genes and hyperexcitability. Furthermore, we report that hepatic leukemia factor (Hlf), a member of PAR bZIP TFs family, is invariably downregulated in animal models of acquired epilepsy, regulates neuronal activity in vitro and its overexpression in dentate gyrus neurons in vivo leads to altered expression of genes associated with seizures and epilepsy. Overall, our study provides further evidence of PAR bZIP TFs involvement in epileptogenesis and points to Hlf as the key player.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Giro Dentado/metabolismo , Epilepsia/metabolismo , Regulación de la Expresión Génica , Animales , Giro Dentado/patología , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Ácido Kaínico/efectos adversos , Ácido Kaínico/farmacología , Masculino , Ratones
7.
J Cell Biochem ; 121(10): 4261-4270, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31909503

RESUMEN

This article aimed to reveal the mechanism of long noncoding RNA (lncRNA) urothelial cancer-associated 1 (UCA1) regulated astrocyte activation in temporal lobe epilepsy (TLE) rats via mediating the activation of the JAK/STAT signaling pathway. A model of TLE was established based on rats via kainic acid (KA) injection. All rats were divided into the Sham group (without any treatments), KA group, normal control (NC; injection with empty vector) + KA group, and UCA1 + KA group. The Morris water maze was used to test the learning and memory ability of rats, and the expression of UCA1 in the hippocampus was determined by quantitative real time polymerase chain reaction (qRT-PCR). Surviving neurons were counted by Nissl staining, and expression levels of glial cells glial fibrillary acidic protein (GFAP), p-JAK1, and p-STAT3 and glutamate/aspartate transporter (GLAST) were analyzed by immunofluorescence and Western blot analysis. A rat model of TLE was established by intraperitoneal injection of KA. qRT-PCR and fluorescence analyses showed that UCA1 inhibited astrocyte activation in the hippocampus of epileptic rats. Meanwhile, the Morris water maze analysis indicated that UCA1 improved the learning and memory in epilepsy rats. Moreover, the Nissl staining showed that UCA1 might have a protective effect on neuronal injury induced by KA injection. Furthermore, the immunofluorescence and Western blot analysis revealed that the overexpression of UCA1 inhibited KA-induced abnormal elevation of GLAST, astrocyte activation of the JAK/STAT signaling pathway, as well as hippocampus of epilepsy rats. UCA1 inhibited hippocampal astrocyte activation and JAK/STAT/GLAST expression in TLE rats and improved the adverse reactions caused by epilepsy.


Asunto(s)
Astrocitos/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Janus Quinasa 1/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Animales , Conducta Animal , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/inducido químicamente , Vectores Genéticos/administración & dosificación , Hipocampo/metabolismo , Ácido Kaínico/efectos adversos , Masculino , Memoria , Prueba del Laberinto Acuático de Morris , Neuroglía/metabolismo , Neuronas/metabolismo , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/genética , Ratas , Ratas Sprague-Dawley
8.
PLoS One ; 14(12): e0226575, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31887157

RESUMEN

BACKGROUND AND AIMS: Mesial Temporal Lobe Epilepsy is characterized by progressive changes of both neurons and glia, also referred to as epileptogenesis. No curative treatment options, apart from surgery, are available. DNA methylation (DNAm) is a potential upstream mechanism in epileptogenesis and may serve as a novel therapeutic target. To our knowledge, this is the first study to investigate epilepsy-related DNAm, gene expression (GE) and their relationship, in neurons and glia. METHODS: We used the intracortical kainic acid injection model to elicit status epilepticus. At 24 hours post injection, hippocampi from eight kainic acid- (KA) and eight saline-injected (SH) mice were extracted and shock frozen. Separation into neurons and glial nuclei was performed by flow cytometry. Changes in DNAm and gene expression were measured with reduced representation bisulfite sequencing (RRBS) and mRNA-sequencing (mRNAseq). Statistical analyses were performed in R with the edgeR package. RESULTS: We observed fulminant DNAm- and GE changes in both neurons and glia at 24 hours after initiation of status epilepticus. The vast majority of these changes were specific for either neurons or glia. At several epilepsy-related genes, like HDAC11, SPP1, GAL, DRD1 and SV2C, significant differential methylation and differential gene expression coincided. CONCLUSION: We found neuron- and glia-specific changes in DNAm and gene expression in early epileptogenesis. We detected single genetic loci in several epilepsy-related genes, where DNAm and GE changes coincide, worth further investigation. Further, our results may serve as an information source for neuronal and glial alterations in both DNAm and GE in early epileptogenesis.


Asunto(s)
Metilación de ADN , Epilepsia del Lóbulo Temporal/genética , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Neuroglía/química , Neuronas/química , Animales , Modelos Animales de Enfermedad , Epigénesis Genética , Epilepsia del Lóbulo Temporal/inducido químicamente , Galanina/genética , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Histona Desacetilasas/genética , Ácido Kaínico/efectos adversos , Masculino , Ratones , Osteopontina/genética , Receptores de Dopamina D1/genética , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
9.
EBioMedicine ; 47: 470-483, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31474551

RESUMEN

BACKGROUND: NACHT and WD repeat domain-containing protein 1 (Nwd1) is a member of the innate immune protein subfamily. Nwd1 contributes to the androgen receptor signaling pathway and is involved in axonal growth. However, the mechanisms that underlie pathophysiological dysfunction in seizures remain unclear. METHODS: Biochemical methods were used to assess Nwd1 expression and localization in a mouse model of kainic acid (KA)-induced acute seizures and temporal lobe epilepsy (TLE) patients. Electrophysiological recordings were used to measure the role of Nwd1 in regulating synaptic transmission and neuronal hyperexcitability in a model of magnesium-free-induced seizure in vitro. Behavioral experiments were performed, and seizure-induced pathological changes were evaluated in a KA-induced seizure model in vivo. GluN2B expression was measured and its correlation with Tyr1472-GluN2B phosphorylation was analyzed in primary hippocampal neurons. FINDINGS: We demonstrated high protein levels of Nwd1 in brain tissues obtained from mice with acute seizures and TLE patients. Silencing Nwd1 in mice using an adeno-associated virus (AAV) profoundly suppressed neuronal hyperexcitability and the occurrence of acute seizures, which may have been caused by reducing GluN2B-containing NMDA receptor-dependent glutamatergic synaptic transmission. Moreover, the decreased activation of Nwd1 reduced GluN2B expression and the phosphorylation of the GluN2B subunit at Tyr1472. INTERPRETATION: Here, we report a previously unrecognized but important role of Nwd1 in seizure models in vitro and in vivo, i.e., modulating the phosphorylation of the GluN2B subunit at Tyr1472 and regulating neuronal hyperexcitability. Meanwhile, our findings may provide a therapeutic strategy for the treatment of epilepsy or other hyperexcitability-related neurological disorders. FUND: The funders have not participated in the study design, data collection, data analysis, interpretation, or writing of the report.


Asunto(s)
Potenciales Evocados/efectos de los fármacos , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ácido Kaínico/efectos adversos , Ratones , Fosforilación , Convulsiones/etiología , Convulsiones/metabolismo , Convulsiones/fisiopatología , Sinapsis/genética , Sinapsis/metabolismo , Transmisión Sináptica
10.
Food Chem Toxicol ; 133: 110765, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31430510

RESUMEN

Oridonin (ORI) is a natural diterpenoid presented in some medicinal plants. The effects of pre-treatments from ORI against MPP+- or kainic acid (KA)-induced damage in nerve growth factor (NGF)-differentiated PC12 cells were investigated. Results showed that pre-treatments of ORI at 0.25-2 µM enhanced the viability and plasma membrane integrity of NGF-differentiated PC12 cells. MPP+ or KA exposure down-regulated Bcl-2 mRNA expression, up-regulated Bax mRNA expression, increased caspase-3 activity and decreased Na+-K+ ATPase activity. ORI pre-treatments at test concentrations reversed these changes. ORI pre-treatments decreased reactive oxygen species production, raised glutathione level, and increased glutathione peroxidase, glutathione reductase and catalase activities in MPP+ or KA treated cells. ORI pre-treatments lowered tumor necrosis factor-alpha, interleukin (IL)-1beta, IL-6 and prostaglandin E2 levels in MPP+ or KA treated cells. ORI also diminished MPP+ or KA induced increase in nuclear factor-κB binding activity. MPP+ exposure suppressed tyrosine hydroxylase (TH) mRNA expression and decreased dopamine content. KA exposure reduced glutamine synthetase (GS) mRNA expression, raised glutamate level and lowered glutamine level. ORI pre-treatments at 0.5-2 µM up-regulated mRNA expression of TH and GS, restored DA and glutamine content. These findings suggested that oridonin was a potent neuro-protective agent against Parkinson's disease and seizure.


Asunto(s)
1-Metil-4-fenilpiridinio/efectos adversos , Diterpenos de Tipo Kaurano/farmacología , Ácido Kaínico/efectos adversos , Fármacos Neuroprotectores/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Diterpenos de Tipo Kaurano/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/metabolismo , Fármacos Neuroprotectores/toxicidad , Estrés Oxidativo/efectos de los fármacos , Células PC12 , ARN Mensajero/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
EBioMedicine ; 39: 377-387, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30502054

RESUMEN

BACKGROUND: Glycogen synthase kinase-3ß (GSK3ß) is a key regulator of cellular homeostasis. In neurons, GSK3ß contributes to the control of neuronal transmission and plasticity, but its role in epilepsy remains to be defined. METHODS: Biochemical and electrophysiological methods were used to assess the role of GSK3ß in regulating neuronal transmission and epileptogenesis. GSK3ß activity was increased genetically in GSK3ß[S9A] mice. Its effects on neuronal transmission and epileptogenesis induced by kainic acid were assessed by field potential recordings in mice brain slices and video electroencephalography in vivo. The ion channel expression was measured in brain samples from mice and followed by analysis in samples from patients with temporal lobe epilepsy or focal cortical dysplasia in correlation to GSK3ß phosphorylation. FINDINGS: Higher GSK3ß activity decreased the progression of kainic acid induced epileptogenesis. At the biochemical level, higher GSK3ß activity increased the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel 4 under basal conditions and in the epileptic mouse brain and decreased phosphorylation of the glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 at Serine 831 under basal conditions. Moreover, we found a significant correlation between higher inhibitory GSK3ß phosphorylation at Serine 9 and higher activating GluA1 phosphorylation at Serine 845 in brain samples from epileptic patients. INTERPRETATION: Our data imply GSK3ß activity in the protection of neuronal networks from hyper-activation in response to epileptogenic stimuli and indicate that the anti-epileptogenic function of GSK3ß involves modulation of HCN4 level and the synaptic AMPA receptors pool.


Asunto(s)
Epilepsia/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ácido Kaínico/efectos adversos , Proteínas Musculares/metabolismo , Canales de Potasio/metabolismo , Receptores AMPA/metabolismo , Adolescente , Adulto , Animales , Células Cultivadas , Niño , Preescolar , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/inducido químicamente , Epilepsia/genética , Femenino , Glucógeno Sintasa Quinasa 3 beta/química , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Fosforilación , Receptores AMPA/química , Transducción de Señal , Transmisión Sináptica , Grabación en Video
12.
Mol Med Rep ; 17(6): 8357-8362, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29693706

RESUMEN

The purpose of the present study was to determine whether triptolide (T10) had any effect on major histocompatibility complex class II (MHC II) expression in kainic acid (KA)­activated microglia, and to investigate the underlying molecular mechanism. BV­2 microglia were pretreated with T10 prior to activation with KA. The expression level of MHC II and class II transactivator (CIITA) mRNA was determined via reverse transcription­polymerase chain reaction. The expression of MHC II, CIITA and the phosphorylation level of c­Jun and proto­oncogene c­Fos (c­Fos) was determined by western blotting. The protein expression level of MHC II was determined by immunocytochemistry. It was observed that the mRNA and protein levels of MHC II and CIITA were increased in KA­activated BV­2 microglia, and that this increase was almost completely eliminated by T10. AP­1 is a family of homodimers or heterodimers, composed of Jun family and Fos family proteins. Sequence analysis revealed an AP­1 DNA binding site in the promoter of CIITA. The phosphorylation of c­Jun and c­Fos was increased in KA­activated microglia, while T10 was able to suppress the phosphorylation of c­Jun and c­Fos in KA­activated microglia. These data suggested that T10 may exert suppressive effects on MHC II expression in KA­activated microglia, and that the mechanism may involve the regulation of AP­1 activity.


Asunto(s)
Diterpenos/farmacología , Epilepsia/etiología , Expresión Génica , Antígenos de Histocompatibilidad Clase II/genética , Ácido Kaínico/efectos adversos , Microglía/metabolismo , Fenantrenos/farmacología , Animales , Epilepsia/metabolismo , Compuestos Epoxi/farmacología , Inmunohistoquímica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Mensajero/genética , Ratas , Transactivadores/genética , Transactivadores/metabolismo
13.
Epilepsy Behav ; 79: 154-161, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29289903

RESUMEN

Tobacco smoking is considered the greatest risk factor for early death caused by noncommunicable diseases. Currently, there are more than one billion tobacco smokers in the world predisposed to many diseases including heart attack, stroke, cancer, and premature birth or birth defects related to the consumption of cigarettes. However, studies on the association between tobacco smoking and seizures or epilepsy are insufficient and not well documented. In the present study, the authors examined the convulsive effects of the intracerebroventricular administration of cigarette smoke condensate (CSC, 2µl/Rat) in rats and compared it with the intensity of seizures in the kainic acid (KA)-induced seizure model of epilepsy. The role of the cholinergic system was also investigated by testing the effect of the muscarinic acetylcholine receptors (mAChRs) antagonist atropine (2ml/kg) on CSC-induced seizures. The results indicate that a central injection of CSC produces an epileptic behavior similar to that induced by KA, the similarities include the following parameters: time latency of seizures, latency and duration of tonic-clonic seizures, duration of seizures, survival, and tonic-clonic rate. However, a pretreatment with atropine reduced seizures and all their parameters.


Asunto(s)
Convulsivantes , Epilepsia/inducido químicamente , Antagonistas Muscarínicos/farmacología , Convulsiones/inducido químicamente , Fumar/efectos adversos , Animales , Atropina/farmacología , Femenino , Ácido Kaínico/efectos adversos , Ácido Kaínico/metabolismo , Ácido Kaínico/farmacología , Masculino , Embarazo , Ratas , Receptores Muscarínicos , Convulsiones/epidemiología
14.
Pathobiology ; 84(4): 184-191, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28095381

RESUMEN

BACKGROUND: Previous studies demonstrated the cytoprotective effect of geranylgeranylacetone (GGA), a heat shock protein inducer, against ischemic insult or kainic acid (KA)-induced neuronal cell death. Phosphatidylinositol-3 kinase (PI3K)/Akt is thought to be an important factor that mediates neuroprotection. However, the signaling pathways in the brain in vivo after oral GGA administration remain unclear. METHODS: We measured and compared hippocampal neuron density to investigate the effect of GGA on KA-induced cell death in rats. We evaluated the effects of pretreatment with wortmannin (Wort), a specific PI3K inhibitor, on GGA-induced neuroprotection against KA-induced cell death. To clarify the relationship between PI3K/Akt activation and neuroprotection, we used immunoblot analysis to determine the amounts of p-Akt and vascular endothelial growth factor (VEGF) proteins present after GGA administration with or without Wort treatment. RESULTS: Neuroprotective effects of GGA (pretreatment with a single oral dose of GGA, 800 mg/kg, 48 h before KA injection) were prevented by Wort pretreatment, which indicates that the selective PI3K/Akt pathway may mediate the GGA-dependent protection. Oral GGA-induced p-Akt and VEGF, and GGA pretreatment enhanced KA-induced VEGF, both of which were prevented by Wort pretreatment. CONCLUSION: These results suggest that a single oral dose of GGA induces p-Akt and that GGA plays an important role in neuroprotection against KA-induced neuronal cell death through VEGF induction.


Asunto(s)
Muerte Celular/efectos de los fármacos , Diterpenos/administración & dosificación , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Administración Oral , Androstadienos/farmacología , Animales , Encéfalo/metabolismo , Hipocampo/metabolismo , Ácido Kaínico/efectos adversos , Masculino , Neuronas/fisiología , Fosfatidilinositol 3-Quinasa/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Ratas , Ratas Wistar , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Wortmanina
15.
Indian J Pharmacol ; 48(1): 11-4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26997715

RESUMEN

OBJECTIVE: Since temporal lobe epilepsy (TLE) is associated with learning and memory impairment, we investigated the beneficial effect of Vitamin E on the impaired learning and memory in the intrahippocampal kainate model of TLE in rats. MATERIALS AND METHODS: Rats were divided into sham, Vitamin E-treated sham, kainate, and Vitamin E-treated kainate. Intrahippocampal kainate was used for induction of epilepsy. Vitamin E was injected intraperitoneal (i.p.) at a dose of 200 mg/kg/day started 1 week before surgery until 1 h presurgery. Initial and step-through latencies in the passive avoidance test and alternation behavior percentage in Y-maze were finally determined in addition to measurement of some oxidative stress markers. RESULTS: Kainate injection caused a higher severity and rate of seizures and deteriorated learning and memory performance in passive avoidance paradigm and spontaneous alternation as an index of spatial recognition memory in Y-maze task. Intrahippocampal kainate also led to the elevation of malondialdehyde (MDA) and nitrite and reduced activity of superoxide dismutase (SOD). Vitamin E pretreatment significantly attenuated severity and incidence rate of seizures, significantly improved retrieval and recall in passive avoidance, did not ameliorate spatial memory deficit in Y-maze, and lowered MDA and enhanced SOD activity. CONCLUSION: Vitamin E improves passive avoidance learning and memory and part of its beneficial effect is due to its potential to mitigate hippocampal oxidative stress.


Asunto(s)
Epilepsia del Lóbulo Temporal/prevención & control , Hipocampo/efectos de los fármacos , Ácido Kaínico/efectos adversos , Aprendizaje/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Vitamina E/farmacología , Animales , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/fisiopatología , Masculino , Ratas , Ratas Wistar
16.
Epilepsy Behav ; 50: 40-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26103532

RESUMEN

Maternal immune activation (MIA) results in the development of autism in the offspring via hyperactivation of IL-6 signaling. Furthermore, experimental studies showed that the MIA-associated activation of interleukin-1ß (IL-1ß) concurrently with IL-6 increases the rate and the severity of hippocampal kindling in mice, thus, offering an explanation for autism-epilepsy comorbidity. We examined whether epileptic phenotype triggered by prenatal exposure to IL-6 and IL-1ß combination is restricted to kindling or whether it is reproducible in another model of epilepsy, whereby spontaneous seizures develop following kainic acid (KA)-induced status epilepticus. We also examined whether in mice prenatally exposed to IL-6 and IL-6+IL-1ß, the presence of spontaneous seizures would exacerbate autism-like features. Between days 12 and 16 of pregnancy, C57BL/6J mice received daily injections of IL-6, IL-1ß, or IL-6+IL-1ß combination. At postnatal day 40, male offspring were examined for the presence of social behavioral deficit, and status epilepticus was induced by intrahippocampal KA injection. After 6weeks of monitoring for spontaneous seizures, sociability was tested again. Both IL-6 and IL-6+IL-1ß offspring presented with social behavioral deficit. Prenatal exposure to IL-6 alleviated, while such exposure to IL-6+IL-1ß exacerbated, the severity of KA-induced epilepsy. Increased severity of epilepsy in the IL-6+IL-1ß mice correlated with the improvement of autism-like behavior. We conclude that complex and not necessarily agonistic relationships exist between epileptic and autism-like phenotypes in an animal model of MIA coupled with KA-induced epilepsy and that the nature of these relationships depends on components of MIA involved.


Asunto(s)
Trastorno Autístico/fisiopatología , Citocinas/fisiología , Epilepsia/fisiopatología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/fisiopatología , Epilepsia/inducido químicamente , Femenino , Hipocampo/metabolismo , Interleucina-1beta/fisiología , Interleucina-6/fisiología , Ácido Kaínico/efectos adversos , Excitación Neurológica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Convulsiones/inducido químicamente , Estado Epiléptico/fisiopatología
17.
Cytotherapy ; 16(12): 1760-70, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25442789

RESUMEN

BACKGROUND AIMS: Hippocampal neurodegeneration is one of the hallmarks in neurological and neurodegenerative diseases such as temporal lobe epilepsy and Alzheimer disease. Human embryonic kidney (HEK) cells are a mixed population of cells, including neurons, and their conditioned medium is enriched with erythropoietin (EPO). Because EPO is a known neuroprotectant, we hypothesized that infusion of HEK cells or HEK-conditioned medium (HEK-CM) may provide neuroprotection against kainic acid (KA)-induced hippocampal damage in mice. METHODS: Adult CF1 mice were treated with KA to induce hippocampal damage. On 3rd and 5th days after KA treatment, HEK cells or HEK-CM was infused intravenously through the tail vein. On the 7th and 8th days after KA treatment, all groups of mice were subjected to cognitive and depression assessment by use of a novel object recognition test and a forced swim test, respectively. Subsequent to this assessment, mice were killed and the brain samples were used to assess the histopathology and messenger RNA expression for EPO and B-cell lymphoma-2 (Bcl-2). RESULTS: We found that infusion of HEK cells/HEK-CM improves cognitive function and alleviates symptoms of depression. Histological assessment demonstrates complete neuroprotection against KA-mediated excitotoxicity, and the hippocampal cytoarchitecture of HEK cells/HEK-CM treated mice was comparable to normal control mice. HEK cells/HEK-CM treatment could provide neuroprotection by upregulating the endogenous EPO and Bcl-2 in KA-treated mice. CONCLUSIONS: Our present data demonstrate for the first time that infusion of HEK cells/HEK-CM can prevent excitotoxic hippocampal damage and alleviate consequent behavioral abnormalities.


Asunto(s)
Lesiones Encefálicas , Medios de Cultivo Condicionados , Agonistas de Aminoácidos Excitadores/efectos adversos , Hipocampo/lesiones , Ácido Kaínico/efectos adversos , Animales , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Células HEK293 , Hipocampo/metabolismo , Humanos , Ácido Kaínico/farmacología , Masculino , Ratones , Factores de Tiempo
18.
Gene Ther ; 21(12): 1029-40, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25231174

RESUMEN

Astrocytes are an attractive cell target for gene therapy, but the validation of new therapeutic candidates is needed. We determined whether adeno-associated viral (AAV) vector-mediated overexpression of glutamine synthetase (GS) or excitatory amino-acid transporter 2 (EAAT2), or expression of microRNA targeting adenosine kinase (miR-ADK) in hippocampal astrocytes in the rat brain could modulate susceptibility to kainate-induced seizures and neuronal cell loss. Transgene expression was found predominantly in astrocytes following direct injection of glial-targeting AAV9 vectors by 3 weeks postinjection. ADK expression in miR-ADK vector-injected rats was reduced by 94-96% and was associated with an ~50% reduction in the duration of kainate-induced seizures and greater protection of dentate hilar neurons but not CA3 neurons compared with miR-control vector-injected rats. In contrast, infusion of AAV-GS and EAAT2 vectors did not afford any protection against seizures or neuronal damage as the level of transcriptional activity of the glial fibrillary acidic promoter was too low to drive any significant increase in transgenic GS or EAAT2 relative to the high endogenous levels of these proteins. Our findings support ADK as a prime therapeutic target for gene therapy of temporal lobe epilepsy and suggest that alternative approaches including the use of stronger glial promoters are needed to increase transgenic GS and EAAT2 expression to levels that may be required to affect seizure induction and propagation.


Asunto(s)
Adenosina Quinasa/genética , Epilepsia del Lóbulo Temporal/terapia , Transportador 2 de Aminoácidos Excitadores/genética , Marcación de Gen , Terapia Genética/métodos , Glutamato-Amoníaco Ligasa/genética , Adenosina Quinasa/metabolismo , Animales , Astrocitos/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos , Glutamato-Amoníaco Ligasa/metabolismo , Hipocampo/metabolismo , Ácido Kaínico/efectos adversos , Masculino , Neuroglía/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Transgenes/genética
19.
Zhonghua Yi Xue Za Zhi ; 93(7): 550-3, 2013 Feb 19.
Artículo en Chino | MEDLINE | ID: mdl-23660329

RESUMEN

OBJECTIVE: To explore the effects of low-frequency electric stimulus (LFS) on hippocampal of α5 subunit of extrasynaptic GABAA receptor in kainic acid-induced epilepsy rats and explore the possible mechanism of LFS on hippocampus to treat epilepsy. METHODS: A total of 32 male Sprague-Dawley rats were divided randomly into 4 groups:epileptic, pseudo-surgery, LFS treatment and control. The expression of α5 subunit of extrasynaptic GABAA receptor and the association with the curative effects of LFS were evaluated by behavioristics, real-time fluorogenic quantitative polymerase chain reaction (PCR) and Western blot. RESULT: After treatment, the number of seizures in LFS group were significantly lower than that in the epileptic and pseudo-surgery groups (2.88 ± 0.83 vs 8.50 ± 0.93 and 8.88 ± 0.83) (P < 0.01). The results of real-time PCR demonstrated that the expression of mRNA of α5 subunits in LFS treatment group was obviously higher than that in epileptic and pseudo-surgery groups (0.74 ± 0.20 vs 0.30 ± 0.16 and 0.31 ± 0.16) (P < 0.01), but was lower than 1.10 ± 0.23 in control group. Western blot demonstrated that the protein expression of α5 subunits in LFS (0.75 ± 0.09) and control (0.88 ± 0.09) groups were dramatically higher than that in epileptic (0.22 ± 0.08) and pseudo-surgery (0.26 ± 0.08) groups (P < 0.01). CONCLUSION: Low-frequency electric stimulus on hippocampus is effective in controlling epileptic seizures. And the mechanism may be related with the expression level of α5 subunits of extrasynaptic GABAA receptor.


Asunto(s)
Estimulación Eléctrica/métodos , Epilepsia/metabolismo , Hipocampo/metabolismo , Receptores de GABA-A/metabolismo , Animales , Epilepsia/inducido químicamente , Ácido Kaínico/efectos adversos , Masculino , Ratas , Ratas Sprague-Dawley
20.
J Immunol ; 190(7): 3466-79, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23420885

RESUMEN

The C/EBP ß is a basic leucine zipper transcription factor that regulates a variety of biological processes, including metabolism, cell proliferation and differentiation, and immune response. Recent findings show that C/EBP ß-induced inflammatory responses mediate kainic acid-triggered excitotoxic brain injury. In this article, we show that protein kinase C ζ enhances K-ras expression and subsequently activates the Raf/MEK/ERK1/2 pathway in the hippocampus of domoic acid (DA)-treated mice, which promotes C/EBP ß expression and induces inflammatory responses. Elevated production of TNF-α impairs mitochondrial function and increases the levels of reactive oxygen species by IκB kinase ß/NF-κB signaling. The aforementioned inflammation and oxidative stress lead to memory deficits in DA-treated mice. However, troxerutin inhibits cyclin-dependent kinase 1 expression, enhances type 1 protein phosphatase α dephosphorylation, and abolishes MEK/ERK1/2/C/EBP ß activation, which subsequently reverses the memory impairment observed in the DA-treated mice. Thus, troxerutin is recommended as a potential candidate for the prevention and therapeutic treatment of cognitive deficits resulting from excitotoxic brain damage and other brain disorders.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Hidroxietilrutósido/análogos & derivados , Inflamación/metabolismo , Trastornos de la Memoria/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Butadienos/farmacología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína Quinasa CDC2/metabolismo , Técnicas de Silenciamiento del Gen , Genes ras , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hidroxietilrutósido/administración & dosificación , Hidroxietilrutósido/farmacología , Mediadores de Inflamación/metabolismo , Ácido Kaínico/efectos adversos , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/genética , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , NADPH Oxidasas/genética , Nitrilos/farmacología , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA