Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Acta Neuropathol Commun ; 8(1): 206, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33256836

RESUMO

The Ihara epileptic rat (IER) is a mutant model with limbic-like seizures whose pathology and causative gene remain elusive. In this report, via linkage analysis, we identified Down syndrome cell adhesion molecule-like 1(Dscaml1) as the responsible gene for IER. A single base mutation in Dscaml1 causes abnormal splicing, leading to lack of DSCAML1. IERs have enhanced seizure susceptibility and accelerated kindling establishment. Furthermore, GABAergic neurons are severely reduced in the entorhinal cortex (ECx) of these animals. Voltage-sensitive dye imaging that directly presents the excitation status of brain slices revealed abnormally persistent excitability in IER ECx. This suggests that reduced GABAergic neurons may cause weak sustained entorhinal cortex activations, leading to natural kindling via the perforant path that could cause dentate gyrus hypertrophy and epileptogenesis. Furthermore, we identified a single nucleotide substitution in a human epilepsy that would result in one amino acid change in DSCAML1 (A2105T mutation). The mutant DSCAML1A2105T protein is not presented on the cell surface, losing its homophilic cell adhesion ability. We generated knock-in mice (Dscaml1A2105T) carrying the corresponding mutation and observed reduced GABAergic neurons in the ECx as well as spike-and-wave electrocorticogram. We conclude that DSCAML1 is required for GABAergic neuron placement in the ECx and suppression of seizure susceptibility in rodents. Our findings suggest that mutations in DSCAML1 may affect seizure susceptibility in humans.


Assuntos
Moléculas de Adesão Celular/genética , Córtex Entorrinal/patologia , Neurônios GABAérgicos/patologia , Convulsões/genética , Animais , Eletroencefalografia , Predisposição Genética para Doença , Excitação Neurológica/genética , Camundongos , Ratos , Ratos Mutantes
2.
Epilepsia ; 61(10): 2277-2288, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32954490

RESUMO

OBJECTIVE: Development of novel therapies for temporal lobe epilepsy is hindered by a lack of models suitable for drug screening. While testing the hypothesis that "inhibiting inhibitory neurons" was sufficient to induce seizures, it was discovered that a mild electrical kindling protocol of VGAT-Cre mice led to spontaneous motor and electrographic seizures. This study characterizes these seizures and investigates the mechanism. METHODS: Mice were implanted with electroencephalographic (EEG) headsets that included a stimulating electrode in the hippocampus before being electrically kindled. Seizures were evaluated by review of EEG recordings and behavior. γ-Aminobutyric acidergic (GABAergic) neurotransmission was evaluated by quantitative polymerase chain reaction, immunocytochemistry, Western blot, and electrophysiology. RESULTS: Electrical kindling of VGAT-Cre mice induces spontaneous recurring seizures after a short latency (6 days). Seizures occur 1-2 times per day in both male and female mice, with only minimal neuronal death. These mice express Cre recombinase under the control of the vesicular GABA transporter (VGAT), a gene that is specifically expressed in GABAergic inhibitory neurons. The insertion of Cre disrupts the expression of VGAT mRNA and protein, and impairs GABAergic synaptic transmission in the hippocampus. SIGNIFICANCE: Kindled VGAT-Cre mice can be used to study the mechanisms involved in epileptogenesis and may be useful for screening novel therapeutics.


Assuntos
Modelos Animais de Doenças , Epilepsia do Lobo Temporal/metabolismo , Integrases/biossíntese , Excitação Neurológica/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/biossíntese , Animais , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/fisiopatologia , Feminino , Integrases/genética , Excitação Neurológica/genética , Excitação Neurológica/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/antagonistas & inibidores , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
3.
Curr Mol Pharmacol ; 13(3): 251-260, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31894752

RESUMO

BACKGROUND: Epilepsy remains challenging to treat still no etiologic treatment has been identified, however, some antiepileptic drugs (AEDs) are able to modify the pathogenesis of the disease. Lacosamide (LCM) has been shown to possess complex anticonvulsant and neuroprotective actions, being an enhancer of the slow inactivation of voltage-gated sodium channels, and it has the potential to prevent epileptogenesis. Recent evidence has shown that LCM indirectly improves the function of GABAA receptors. Receptors at most GABAergic synapses involve the gamma-2 subunit, which contributes to both phasic and tonic inhibition, and its presence assures benzodiazepine sensitivity. Moreover, mutant gamma-2 subunits were associated with generalized epilepsy syndromes. In animal models, the expression of the gamma-2 subunit of the gamma-aminobutyric acid A receptor (GABAAg2) was shown to be increased in pentylenetetrazole (PTZ)-induced chemical kindling in Wistar rats. OBJECTIVE: This study hypothesized that LCM might affect the kindling process by influencing the expression of GABAA receptors in the hippocampus. METHODS: The gene and protein expression levels of the GABAAg2 were studied using RT-qPCR and immunofluorescent staining. RESULTS: It was found that LCM treatment (10 mg/kg i.p. daily for 57 days) reduced the maximal intensity of the PTZ-induced seizures but did not prevent kindling. On the other hand, LCM treatment reverted the increase of mRNA expression of GABAAg2 in the hippocampus and prevented the decrease of GABAAg2 protein in the hippocampal CA1 region. CONCLUSION: LCM could exhibit modulatory effects on the GABAergic system of the hippocampus that may be independent of the anticonvulsant action.


Assuntos
Anticonvulsivantes/farmacologia , Excitação Neurológica/genética , Lacosamida/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Receptores de GABA-A/biossíntese , Convulsões/prevenção & controle , Animais , Anticonvulsivantes/uso terapêutico , Convulsivantes/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Lacosamida/uso terapêutico , Masculino , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Pentilenotetrazol/toxicidade , Subunidades Proteicas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Receptores de GABA-A/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico
4.
Neurobiol Dis ; 136: 104719, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31862541

RESUMO

Patients with Alzheimer's disease (AD) experience seizures at higher rates than the general population of that age, suggesting an underexplored role of hyperexcitability in AD. Genetic variants in presenilin (PSEN) 1 and 2 genes lead to autosomal dominant early-onset AD (ADAD); patients with PSEN gene variants also report seizures. Pharmacological control of seizures in AD may be disease-modifying. Preclinical efficacy of FDA-approved antiseizure drugs (ASDs) is well defined in young adult rodents; however, the efficacy of ASDs in aged rodents with chronic seizures is less clear. The mechanism by which ADAD genes lead to AD remains unclear, and even less studied is the pathogenesis of epilepsy in AD. PSEN variants generally all result in a biochemical loss of function (De Strooper, 2007). We herein determined whether well-established models of acute and chronic seizure could be used to explore the relationship between AD genes and seizures through investigating whether loss of normal PSEN2 function age-dependently influenced susceptibility to seizures and/or corneal kindling acquisition. PSEN2 knockout (KO) and age-matched wild-type (WT) mice were screened from 2- to 10-months-old to establish age-dependent focal seizure threshold. Additionally, PSEN2 KO and WT mice aged 2- and 8-months-old underwent corneal kindling such that mice were aged 3- and 9-months old at the beginning of ASD efficacy testing. We then defined the dose-dependent efficacy of mechanistically distinct ASDs on kindled seizures of young versus aged mice to better understand the applicability of corneal kindling to real-world use for geriatric patients. PSEN2 KO mice demonstrated early-life reductions in seizure threshold. However, kindling acquisition was delayed in 2-month-old PSEN2 KO versus WT mice. Young male WT mice took 24.3 ± 1.3 (S.E.M.) stimulations to achieve kindling criterion, whereas age-matched PSEN2 KO male mice took 41.2 ± 1.1 stimulations (p < .0001). The rate of kindling acquisition of 8-month-old mice was no longer different from WT. This study demonstrates that loss of normal PSEN2 function is associated with age-dependent changes in the in vivo susceptibility to acute seizures and kindling. Loss of normal PSEN2 function may be an underexplored molecular contributor to seizures. The use of validated models of chronic seizures in aged rodents may uncover age-related changes in susceptibility to epileptogenesis and/or ASD efficacy in mice with AD-associated genotypes, which may benefit the management of seizures in AD.


Assuntos
Predisposição Genética para Doença , Excitação Neurológica/metabolismo , Presenilina-2/deficiência , Convulsões/metabolismo , Animais , Feminino , Predisposição Genética para Doença/genética , Excitação Neurológica/genética , Masculino , Camundongos , Camundongos Knockout , Presenilina-2/genética , Convulsões/genética
5.
Epilepsia ; 60(10): 2023-2036, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31468516

RESUMO

Epidemiological data and gene association studies suggest a genetic predisposition to developing epilepsy after an acquired brain insult, such as traumatic brain injury. An improved understanding of genetic determinants of vulnerability is imperative for early disease diagnosis and prognosis prediction, with flow-on benefits for the development of targeted antiepileptogenic treatments as well as optimal clinical trial design. In the laboratory, one approach to investigate why some individuals are more vulnerable to acquired epilepsy than others is to examine unique rodent models exhibiting either vulnerability or resistance to epileptogenesis. This review focuses on the most well-characterized of these models, the FAST (seizure-prone) and SLOW (seizure-resistant) rat strains, which were derived by selective breeding for differential amygdala electrical kindling rates. We describe how these strains differ in their seizure profiles, neuroanatomy, and neurobehavioral phenotypes, both at baseline and after a brain insult, with this knowledge proving fruitful to identify common pathological abnormalities associated with seizure susceptibility and psychiatric comorbidities. It is important to note that accruing data on strain differences in multiple biological processes provides insight into why some individuals may be more vulnerable to epileptogenesis, although future studies are evidently needed to identify the precise molecular and genetic risk factors. Together, the FAST and SLOW rat strains, and other similar experimental models, are invaluable neurobiological tools to investigate the effect of genetic background on acquired epilepsy risk, as well as the poorly understood relationship between epilepsy development and associated comorbidities.


Assuntos
Modelos Animais de Doenças , Epilepsia/genética , Predisposição Genética para Doença , Convulsões/genética , Tonsila do Cerebelo/fisiopatologia , Animais , Epilepsia/fisiopatologia , Excitação Neurológica/genética , Fenótipo , Ratos , Convulsões/fisiopatologia
6.
Epilepsia ; 60(7): 1378-1386, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31206645

RESUMO

OBJECTIVE: Recent data indicate that amygdala kindling leads to significant changes in interictal neuronal firing patterns of thalamic reticular nucleus (TRN) neurons by decreasing the spontaneous firing rate and increasing burst firing in nonepileptic control (NEC) rats. Genetic Absence Epilepsy Rats From Strasbourg (GAERS) were resistant to these kindling-induced firing changes in TRN neurons, and are also resistant to the progression of kindling. We investigated whether a homozygous, missense, single nucleotide mutation (R1584P) in the Cav 3.2 T-type Ca2+ channel gene, which has been correlated with the expression of absence seizures in GAERS, influenced kindling progression and TRN firing patterns. METHODS: Double-crossed (GAERS vs NEC; F2) rats that were homozygous for the Cav 3.2 mutation (PP) and those negative for the mutation (RR) were implanted with a stimulating electrode in the amygdala. Rats received a total of 30 kindling stimulations at their afterdischarge threshold current twice daily, and kindling progression was evaluated. Thereafter, the extracellular neuronal activity of TRN neurons was recorded in vivo under neuroleptanesthesia to investigate the influence of Cav 3.2 mutation on TRN firing patterns. RESULTS: We found that the R1584P mutation did not affect kindling progression in F2 crosses (P = 0.78). However, it influenced kindling-induced neuronal firing of TRN neurons. After 30 stimulations, RR rats exhibited a lower firing rate and a higher percentage of burst firing compared to PP rats. The decrease in firing frequency was correlated with the increase in the amount of burst firing in RR rats (R2  = 0.497). SIGNIFICANCE: Our findings suggest that mutation in Cav 3.2 T-type Ca2+ channels may play a role in the resistance to kindling-induced changes in TRN neurons to a low-frequency and high-percentage bursting pattern seen in association with the convulsive stages of amygdala kindling, but is not in itself enough to explain the resistance to kindling progression observed in GAERS.


Assuntos
Canais de Cálcio Tipo T/genética , Epilepsia Tipo Ausência/genética , Excitação Neurológica , Núcleos Talâmicos/fisiopatologia , Tonsila do Cerebelo/fisiopatologia , Animais , Eletrodos Implantados , Eletroencefalografia , Epilepsia Tipo Ausência/etiologia , Epilepsia Tipo Ausência/fisiopatologia , Excitação Neurológica/genética , Excitação Neurológica/fisiologia , Masculino , Mutação de Sentido Incorreto/genética , Reação em Cadeia da Polimerase , Ratos
7.
Sci Rep ; 9(1): 5236, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30918286

RESUMO

Epileptogenesis is the gradual process by which the healthy brain develops epilepsy. However, the neuronal circuit changes that underlie epileptogenesis are not well understood. Unfortunately, current chemically or electrically induced epilepsy models suffer from lack of cell specificity, so it is seldom known which cells were activated during epileptogenesis. We therefore sought to develop an optogenetic variant of the classical kindling model of epilepsy in which activatable cells are both genetically defined and fluorescently tagged. We briefly optogenetically activated pyramidal cells (PCs) in awake behaving mice every two days and conducted a series of experiments to validate the effectiveness of the model. Although initially inert, brief optogenetic stimuli eventually elicited seizures that increased in number and severity with additional stimulation sessions. Seizures were associated with long-lasting plasticity, but not with tissue damage or astrocyte reactivity. Once optokindled, mice retained an elevated seizure susceptibility for several weeks in the absence of additional stimulation, indicating a form of long-term sensitization. We conclude that optokindling shares many features with classical kindling, with the added benefit that the role of specific neuronal populations in epileptogenesis can be studied. Links between long-term plasticity and epilepsy can thus be elucidated.


Assuntos
Epilepsia/genética , Epilepsia/fisiopatologia , Excitação Neurológica/genética , Neocórtex/fisiopatologia , Optogenética , Animais , Eletroencefalografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Neurochem Int ; 124: 68-81, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30597180

RESUMO

Previously we demonstrated that p53 mediates dopaminergic neurotoxicity via inducing mitochondrial burdens and proapoptotsis. However, little is known about the role of p53 in the excitotoxicity induced by psychostimulant, such as cocaine. Cocaine-induced kindling (convulsive) behaviors significantly increased p53 expression in the brain. Cocaine-induced p53 expression was more pronounced in hippocampus than in striatum or prefrontal cortex. Genetic depletion of p53 significantly attenuated cocaine-induced convulsive behaviors, followed by c-Fos immunoreactivity, and oxidative burdens in the hippocampus of mice. The antioxidant potentials mediated by genetic depletion of p53 were more pronounced in the mitochondrial-than cytosolic-fraction. Depletion of p53 significantly attenuated the changes in mitochondrial transmembrane potential, intramitochondrial Ca2+ level, and mitochondrial oxidative burdens induced by cocaine. Consistently, depletion of p53 significantly inhibited mitochondrial p53 translocation, and cleaved-PKCδ induced by cocaine. In addition, depletion of p53 protected from cytosolic cytochrome c release, and pro-apoptotic changes induced by cocaine. Importantly, the protective/anticonvulsant potentials by genetic depletion of p53 were comparable to those by pifithrin-µ (PFT), a p53 inhibitor. Our results suggest that depletion of p53 offers anticonvulsive and neuroprotective potentials mainly via attenuating mitochondrial oxidative burdens, mitochondrial dysfunction, and pro-apoptotic signalings against cocaine-induced convulsive neurotoxicity.


Assuntos
Apoptose/fisiologia , Cocaína/toxicidade , Excitação Neurológica/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo/fisiologia , Proteína Supressora de Tumor p53/deficiência , Animais , Apoptose/efeitos dos fármacos , Excitação Neurológica/efeitos dos fármacos , Excitação Neurológica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Distribuição Aleatória , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
9.
PLoS One ; 14(1): e0210670, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30677045

RESUMO

Oxidative stress has been considered as one of pathogenesis of brain damage led by epilepsy. Reducing oxidative stress can ameliorate brain damage during seizures. However, expression levels of important antioxidative enzymes such as thioredoxin-1 (TRX1), thioredoxin-like 1 protein (TXNL1) and thioredoxin reductase 1 (TXNRD1) during seizures have not been investigated. In this study, we examined protein and mRNA expression levels of TRX1, TXNL1 and TXNRD1 in different brain regions in PTZ induced seizure model mice. We found that protein expression levels of TRX1, TXNL1 and TXNRD1 are simultaneously up-regulated by 2- or 3-fold in the cortex of both acute and chronic seizure model mice. But there is no unified expression pattern change of these enzymes in the hippocampus, cerebellum and diencephalon in the seizure model mice. Less extent up-regulation of mRNA expression of these enzymes were also observed in the cortex of seizure mice. These data suggest that antioxidative enzymes may provide a protective effect against oxidative stress in the cortex during seizures.


Assuntos
Excitação Neurológica/metabolismo , Convulsões/metabolismo , Tiorredoxina Redutase 1/metabolismo , Tiorredoxinas/metabolismo , Animais , Modelos Animais de Doenças , Excitação Neurológica/genética , Masculino , Camundongos , Convulsões/genética , Tiorredoxina Redutase 1/genética , Tiorredoxinas/genética
10.
Free Radic Biol Med ; 131: 408-431, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30592974

RESUMO

Compelling evidence indicates that oxidative stress contributes to cocaine neurotoxicity. The present study was performed to elucidate the role of the glutathione peroxidase-1 (GPx-1) in cocaine-induced kindling (convulsive) behaviors in mice. Cocaine-induced convulsive behaviors significantly increased GPx-1, p-IkB, and p-JAK2/STAT3 expression, and oxidative burdens in the hippocampus of mice. There was no significant difference in cocaine-induced p-IkB expression between non-transgenic (non-TG) and GPx-1 overexpressing transgenic (GPx-1 TG) mice, but significant differences were observed in cocaine-induced p-JAK2/STAT3 expression and oxidative stress between non-TG and GPx-1 TG mice. Cocaine-induced glial fibrillary acidic protein (GFAP)-labeled astrocytic level was significantly higher in the hippocampus of GPx-1 TG mice. Triple-labeling immunocytochemistry indicated that GPx-1-, p-STAT3-, and GFAP-immunoreactivities were co-localized in the same cells. AG490, a JAK2/STAT3 inhibitor, but not pyrrolidone dithiocarbamate, an NFκB inhibitor, significantly counteracted GPx-1-mediated protective potentials (i.e., anticonvulsant-, antioxidant-, antiapoptotic-effects). Genetic overexpression of GPx-1 significantly attenuated proliferation of Iba-1-labeled microglia induced by cocaine in mice. However, AG490 or astrocytic inhibition (by GFAP antisense oligonucleotide and α-aminoadipate) significantly increased Iba-1-labeled microglial activity and M1 phenotype microglial mRNA levels, reflecting that proinflammatory potentials were mediated by AG490 or astrocytic inhibition. This microglial activation was less pronounced in GPx-1 TG than in non-TG mice. Furthermore, either AG490 or astrocytic inhibition significantly counteracted GPx-1-mediated protective potentials. Therefore, our results suggest that astrocytic modulation between GPx-1 and JAK2/STAT3 might be one of the underlying mechanisms for protecting against convulsive neurotoxicity induced by cocaine.


Assuntos
Cocaína/toxicidade , Glutationa Peroxidase/genética , Janus Quinase 2/genética , Excitação Neurológica/efeitos dos fármacos , Fator de Transcrição STAT3/genética , Convulsões/prevenção & controle , Ácido 2-Aminoadípico/farmacologia , Animais , Anticonvulsivantes/farmacologia , Antioxidantes/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/antagonistas & inibidores , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Glutationa Peroxidase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Excitação Neurológica/genética , Excitação Neurológica/metabolismo , Excitação Neurológica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Estresse Oxidativo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Convulsões/induzido quimicamente , Convulsões/genética , Convulsões/fisiopatologia , Transdução de Sinais , Tirfostinas/farmacologia , Glutationa Peroxidase GPX1
11.
ACS Chem Neurosci ; 10(3): 1434-1444, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30396268

RESUMO

Inflammatory responses involving Toll-like receptor signaling represent a key factor contributing to epileptogenesis. Thus, it is of particular interest to explore the relevance of toll-like receptor ligands and modulators, such as heat shock protein 70 (HSP70). Motivated by recent findings demonstrating an upregulation of HSP70 in a model of epileptogenesis, we analyzed the consequences of genetic and pharmacological targeting of HSP70 expression in a mouse kindling paradigm. Lack of inducible HSP70 resulted in increased prekindling seizure thresholds. However, at threshold stimulation the deficiency-promoted seizure spread, as indicated by an increased seizure severity. Subsequent kindling stimulations elicited more severe seizures in knockout mice, whereas endogenous termination of seizure activity remained unaffected with duration of behavioral and electrographic seizure activity comparable to that of wild-type mice. Interestingly, HSP70 deficiency resulted in enhanced microglia activation in the CA1 region. Next, we assessed a pharmacological targeting approach aiming to promote HSP70 expression. Celastrol treatment had no impact on kindling progression but reduced postkindling seizure thresholds and enhanced microglia activation in CA1 and CA3. In conclusion, the findings from HSP70-knockout mice support a protective role of HSP70 with an effect on microglia activation and spread of seizure activity. Unexpectedly, celastrol administration resulted in detrimental consequences. These findings should be considered as a warning about the general safety of celastrol as a drug candidate. The impact of pathophysiological mechanisms on the quality of celastrol effects requires comprehensive future studies exploring influencing factors. Moreover, alternate strategies to increase HSP70 expression should be further developed and validated.


Assuntos
Tonsila do Cerebelo/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Marcação de Genes/métodos , Proteínas de Choque Térmico HSP70/biossíntese , Excitação Neurológica/genética , Excitação Neurológica/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Excitação Neurológica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Triterpenos Pentacíclicos , Distribuição Aleatória , Triterpenos/farmacologia
12.
Neuroscience ; 386: 108-120, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-29964156

RESUMO

Strong evidence exists that Toll-like receptor (TLR)-mediated effects on microglia functional states can promote ictogenesis and epileptogenesis. So far, research has focused on the role of high-mobility group box protein 1 as an activator of TLRs. However, the development of targeting strategies might need to consider a role of additional receptor ligands. Considering the fact that heat shock protein A1 (hsp70) has been confirmed as a TLR 2 and 4 ligand, we have explored the consequences of its overexpression in a mouse kindling paradigm. The genetic modulation enhanced seizure susceptibility with lowered seizure thresholds prior to kindling. In contrast to wildtype (WT) mice, HSPA1A transgenic (TG) mice exhibited generalized seizures very early during the kindling paradigm. Along with an increased seizure severity, seizure duration proved to be prolonged in TG mice during this phase. Toward the end of the stimulation phase seizure parameters of WT mice reached comparable levels. However, a difference between genotypes was still evident when comparing seizure parameters during the post-kindling threshold determination. Surprisingly, HSPA1A overexpression did not affect microglia activation in the hippocampus. In conclusion, the findings demonstrate that hsp70 can exert pro-convulsant effects promoting ictogenesis in naïve animals. The pronounced impact on the response to subsequent stimulations gives first evidence that genetic HSPA1A upregulation may also contribute to epileptogenesis. Thus, strategies inhibiting hsp70 or its expression might be of interest for prevention of seizures and epilepsy. However, conclusions about a putative pro-epileptogenic effect of hsp70 require further investigations in models with development of spontaneous recurrent seizures.


Assuntos
Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/genética , Excitação Neurológica/genética , Excitação Neurológica/metabolismo , Convulsões/genética , Convulsões/metabolismo , Animais , Progressão da Doença , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Excitação Neurológica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Distribuição Aleatória , Convulsões/patologia
13.
Epilepsia ; 59(4): 753-764, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29377096

RESUMO

OBJECTIVE: Aberrant myelination and developmental delay have been reported in epilepsy. However, it is unclear whether these are linked to intrinsic mechanisms that support a predisposition toward seizures and the development of epilepsy. Thus, we compared rates of myelination and neurodevelopment in male rats selectively bred for enhanced susceptibility to kindling epileptogenesis (FAST) with male rats bred for resistance (SLOW). METHODS: Myelin-specific gene expression was compared in the brainstem, cerebellum, and cerebral hemisphere of FAST and SLOW rats on postnatal days (PNDs) 5, 11, 17, 23, and 90 to determine strain-specific myelination rates. Myelin protein levels were also compared at PNDs 5 and 23 in the brainstem. Relative rates of neurodevelopment were evaluated between PNDs 5 and 21 using physical growth landmarks and neuromotor tests including righting reflex, cliff avoidance, negative geotaxis, and locomotor activity. RESULTS: Myelin-specific mRNA expression was significantly down-regulated in FAST rats on PNDs 5 and 11 in all 3 brain structures, indicating relatively delayed myelination. Likewise, corresponding protein levels were significantly lower in FAST brainstem on PND 5. Developmental delay was evident in the FAST strain such that only 9% of FAST pups, compared to 81% of SLOW, had open eyes by PND 13, locomotor activity was significantly reduced between PNDs 12 and 16, and neuromotor task acquisition was delayed between PNDs 5 and 10. SIGNIFICANCE: Relative delays in myelination and neurodevelopment co-occurred in the seizure-prone FAST strain in the absence of seizures. These findings suggest these symptoms are not seizure-induced and may be mechanistically linked to an underlying pathophysiology supporting a predisposition toward developing epilepsy.


Assuntos
Bainha de Mielina/genética , Fibras Nervosas Mielinizadas/fisiologia , Transtornos do Neurodesenvolvimento/genética , Convulsões/genética , Animais , Modelos Animais de Doenças , Excitação Neurológica/genética , Excitação Neurológica/metabolismo , Locomoção/fisiologia , Masculino , Bainha de Mielina/metabolismo , Transtornos do Neurodesenvolvimento/fisiopatologia , Ratos , Ratos Long-Evans , Ratos Wistar , Convulsões/metabolismo , Convulsões/fisiopatologia , Especificidade da Espécie
14.
CNS Neurosci Ther ; 24(1): 18-28, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29105300

RESUMO

AIMS: Intracellular calcium plays an important role in neuronal hyperexcitability that leads to seizures. One calcium influx route of interest is the transient receptor potential vanilloid type 1 (TRPV1) channel. Here, we evaluated the effects of capsazepine (CPZ), a potent blocker of TRPV1 channels on acoustically evoked seizures (audiogenic seizures, AGS) in the genetically epilepsy-prone rat (GEPR-3), a model of inherited epilepsy. METHODS: Male and female GEPR-3s were used. For the acute CPZ treatment study, GEPR-3s were tested for AGS susceptibility before and after treatment with various doses of CPZ (0, 1, 3, and 10 mg/kg; ip). For semichronic CPZ treatment study, GEPR-3s were tested for AGS susceptibility before and after 5-day CPZ treatment at the dose of 1 mg/kg (ip). The prevalence, latency, and severity of AGS were recorded and analyzed. RESULTS: We found that acute CPZ pretreatment reduced the seizure severity in male GEPR-3s; the effect was dose-dependent. In female GEPR-3s, however, CPZ treatment completely suppressed the seizure susceptibility. Furthermore, semichronic CPZ treatment suppressed seizure susceptibility in female GEPR-3s, but only reduced the seizure severity in male GEPR-3s. CONCLUSIONS: These findings suggest that the TRPV1 channel is a promising molecular target for seizure suppression, with female GEPR-3s exhibiting higher sensitivity than male GEPR-3s.


Assuntos
Epilepsia/metabolismo , Canais de Cátion TRPV/metabolismo , Estimulação Acústica/efeitos adversos , Análise de Variância , Animais , Anticonvulsivantes/uso terapêutico , Benzoatos/uso terapêutico , Temperatura Corporal , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epilepsia/tratamento farmacológico , Epilepsia/etiologia , Epilepsia/genética , Feminino , Excitação Neurológica/genética , Excitação Neurológica/fisiologia , Masculino , Oxazóis/uso terapêutico , Ratos , Ratos Mutantes , Fatores Sexuais , Fatores de Tempo
15.
G3 (Bethesda) ; 7(8): 2545-2558, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28620084

RESUMO

Epilepsy has many causes and comorbidities affecting as many as 4% of people in their lifetime. Both idiopathic and symptomatic epilepsies are highly heritable, but genetic factors are difficult to characterize among humans due to complex disease etiologies. Rodent genetic studies have been critical to the discovery of seizure susceptibility loci, including Kcnj10 mutations identified in both mouse and human cohorts. However, genetic analyses of epilepsy phenotypes in mice to date have been carried out as acute studies in seizure-naive animals or in Mendelian models of epilepsy, while humans with epilepsy have a history of recurrent seizures that also modify brain physiology. We have applied a repeated seizure model to a genetic reference population, following seizure susceptibility over a 36-d period. Initial differences in generalized seizure threshold among the Hybrid Mouse Diversity Panel (HMDP) were associated with a well-characterized seizure susceptibility locus found in mice: Seizure susceptibility 1 Remarkably, Szs1 influence diminished as subsequent induced seizures had diminishing latencies in certain HMDP strains. Administration of eight seizures, followed by an incubation period and an induced retest seizure, revealed novel associations within the calmodulin-binding transcription activator 1, Camta1 Using systems genetics, we have identified four candidate genes that are differentially expressed between seizure-sensitive and -resistant strains close to our novel Epileptogenesis susceptibility factor 1 (Esf1) locus that may act individually or as a coordinated response to the neuronal stress of seizures.


Assuntos
Epilepsia/genética , Loci Gênicos , Predisposição Genética para Doença , Variação Genética , Convulsões/genética , Alelos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cromossomos de Mamíferos/genética , Cruzamentos Genéticos , Modelos Animais de Doenças , Epistasia Genética , Feminino , Flurotila , Estudo de Associação Genômica Ampla , Excitação Neurológica/genética , Masculino , Camundongos , Polimorfismo de Nucleotídeo Único/genética , Locos de Características Quantitativas/genética , Análise de Regressão
16.
Neurobiol Dis ; 105: 213-220, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28602856

RESUMO

How a seizure spreads from a focal onset zone to other regions of the brain is not well understood, and animal studies suggest that there is a genetic influence. To understand how genetic factors may influence seizure spread, we examined whether the kindling resistance of WAG/Rij rats, which are slow to develop kindled motor seizures, is independent of the site of seizure induction and thus a global phenomenon, or whether it is circuit specific. We compared the kindling rates (number of stimulations to induce kindled motor seizures) of WAG/Rij rats to the rates of kindling in Sprague Dawley rats. Both groups underwent a standard hippocampal kindling protocol and a separate group was kindled from the medial dorsal nucleus of the thalamus, a site that has been previously demonstrated to result in the very rapid development of motor seizures. To examine whether there were differences in the interaction in a circuit involved with the motor seizures, evoked responses were obtained from the prefrontal cortex following stimulation of the subiculum or medial dorsal thalamic nucleus. The WAG/Rij rats once again demonstrated resistance to kindling in the hippocampus, but both strains kindled rapidly from the medial dorsal nucleus. In the WAG/Rij rats there was also a reduction in the duration of the afterdischarge in the frontal cortex during hippocampal stimulation, but there was no reduction during thalamic kindling. The prefrontal cortex evoked responses were reduced following stimulation of the subiculum in the WAG/Rij rats, but the evoked responses to thalamic stimulation were the same in both strains. These findings suggest that there are genetic influences in the strength of the input from the subiculum to the prefrontal cortex in WAG/Rij rats that could explain the resistance to limbic kindling because of reduced excitatory drive onto a key target region.


Assuntos
Ondas Encefálicas/genética , Excitação Neurológica , Sistema Límbico/fisiopatologia , Vias Neurais/fisiopatologia , Convulsões/patologia , Animais , Modelos Animais de Doenças , Estimulação Elétrica/efeitos adversos , Eletroencefalografia , Feminino , Lobo Frontal/fisiopatologia , Hipocampo/fisiopatologia , Excitação Neurológica/genética , Masculino , Ratos , Ratos Endogâmicos , Ratos Sprague-Dawley , Convulsões/etiologia , Convulsões/genética , Tálamo/fisiopatologia
17.
Sci Rep ; 6: 38108, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27905510

RESUMO

Voltage-gated sodium channels (VGSCs) play a vital role in controlling neuronal excitability. Nav1.6 is the most abundantly expressed VGSCs subtype in the adult central nervous system and has been found to contribute to facilitate the hyperexcitability of neurons after electrical induction of status epilepticus (SE). To clarify the exact expression patterns of Nav1.6 during epileptogenesis, we examined the expression of Nav1.6 at protein and mRNA levels in two distinct animal models of temporal lobe epilepsy (TLE) including a post-SE model induced by kainic acid (KA) intrahippocampal injection and a kindling model evoked by pentylenetetrazole (PTZ). A prominent, seizure intensity-dependent increase of Nav1.6 expression in reactive astrocytes was observed in ipsilateral hippocampus of post-SE rats, reaching the peak at 21 days after SE, a time point during the latent stage of epileptogenesis. However, Nav1.6 with low expression level was selectively expressed in the hippocampal neurons rather than astrocytes in PTZ-kindled animals. This seizure-related increase of a VGSCs subtype in reactive astrocytes after SE may represent a new mechanism for signal communication between neuron and glia in the course of epileptogenesis, facilitating the neuronal hyperexcitability.


Assuntos
Astrócitos/metabolismo , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Animais , Convulsivantes/toxicidade , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/etiologia , Hipocampo/metabolismo , Humanos , Ácido Caínico/toxicidade , Excitação Neurológica/genética , Excitação Neurológica/metabolismo , Masculino , Neurônios/metabolismo , Pentilenotetrazol/toxicidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/genética , Convulsões/metabolismo , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/complicações , Fatores de Tempo
18.
Genet Mol Res ; 15(3)2016 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-27706789

RESUMO

Epilepsy refers to a clinical syndrome generated by spontaneous seizures in the central nervous system. Epilepsy triggers a complex pathological process including inflammatory response and aquaporin 4 (AQP4) increase. It has been reported that AQP4 helps to enhance the immunological function of the central nervous system in pathological conditions, but the relationship between AQP4 and inflammatory cytokines is poorly understood in chronic epilepsy processes. As an inhibitor of sulfonamide carbonic anhydrase (CA), acetazolamide (AZA) may inhibit water infiltration through AQP4. In this context, pentylenetetrazole (PTZ) is used to induce the chronic epilepsy model in rats to study the chronic epilepsy effects of AQP4 inhibition on proinflammatory cytokine expression in the hippocampus and proinflammatory cytokine quantification analysis of the plasma. Based on the assumption that AQP4 regulates proinflammatory cytokine expression, this article aims to demonstrate this effect in chronic epilepsy of rats. Rats were divided into four groups and were treated with different drugs: saline (Control), acetazolamide (AZA), pentylenetetrazole (PTZ), and pentylenetetrazole plus acetazolamide (PTZ+AZA). The data showed that seizures increased proinflammatory cytokine expression and that AZA significantly inhibited AQP4 expression. Overall, the results suggested that AQP4 inhibition could weaken excitotoxicity in epileptogenesis by reducing proinflammatory cytokines in the hippocampus. The findings provide a new insight into the involvement of cerebral edema insult and proinflammatory cytokines in the process of chronic epilepsy.


Assuntos
Acetazolamida/farmacologia , Aquaporina 4/antagonistas & inibidores , Citocinas/biossíntese , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Hipocampo/metabolismo , Microvasos/metabolismo , Animais , Aquaporina 4/sangue , Aquaporina 4/genética , Aquaporina 4/metabolismo , Doença Crônica , Epilepsia/sangue , Epilepsia/genética , Hipocampo/irrigação sanguínea , Hipocampo/patologia , Excitação Neurológica/genética , Microvasos/patologia , Pentilenotetrazol , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
19.
J Huazhong Univ Sci Technolog Med Sci ; 35(5): 716-722, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26489628

RESUMO

Synaptic vesicle protein 2A (SV2A) involvement has been reported in the animal models of epilepsy and in human intractable epilepsy. The difference between pharmacosensitive epilepsy and pharmacoresistant epilepsy remains poorly understood. The present study aimed to observe the hippocampus SV2A protein expression in amygdale-kindling pharmacoresistant epileptic rats. The pharmacosensitive epileptic rats served as control. Amygdaloid-kindling model of epilepsy was established in 100 healthy adult male Sprague-Dawley rats. The kindled rat model of epilepsy was used to select pharmacoresistance by testing their seizure response to phenytoin and phenobarbital. The selected pharmacoresistant rats were assigned to a pharmacoresistant epileptic group (PRE group). Another 12 pharmacosensitive epileptic rats (PSE group) served as control. Immunohistochemistry, real-time PCR and Western blotting were used to determine SV2A expression in the hippocampus tissue samples from both the PRE and the PSE rats. Immunohistochemistry staining showed that SV2A was mainly accumulated in the cytoplasm of the neurons, as well as along their dendrites throughout all subfields of the hippocampus. Immunoreactive staining level of SV2A-positive cells was 0.483 ± 0.304 in the PRE group and 0.866 ± 0.090 in the PSE group (P < 0.05). Real-time PCR analysis demonstrated that 2(-ΔΔCt) value of SV2A mRNA was 0.30 ± 0.43 in the PRE group and 0.76 ± 0.18 in the PSE group (P < 0.05). Western blotting analysis obtained the similar findings (0.27 ± 0.21 versus 1.12 ± 0.21, P < 0.05). PRE rats displayed a significant decrease of SV2A in the brain. SV2A may be associated with the pathogenesis of intractable epilepsy of the amygdaloid-kindling rats.


Assuntos
Tonsila do Cerebelo/metabolismo , Epilepsia/genética , Excitação Neurológica/genética , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , Vesículas Sinápticas/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiopatologia , Animais , Anticonvulsivantes/farmacologia , Modelos Animais de Doenças , Resistência a Medicamentos , Estimulação Elétrica , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Epilepsia/patologia , Regulação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Excitação Neurológica/efeitos dos fármacos , Excitação Neurológica/metabolismo , Excitação Neurológica/patologia , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fenobarbital/farmacologia , Fenitoína/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/patologia
20.
Neuroscience ; 310: 114-27, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26391920

RESUMO

High-intensity sound can induce seizures in susceptible animals. After repeated acoustic stimuli changes in behavioural seizure repertoire and epileptic EEG activity might be seen in recruited limbic and forebrain structures, a phenomenon known as audiogenic kindling. It is postulated that audiogenic kindling can produce synaptic plasticity events leading to the spread of epileptogenic activity to the limbic system. In order to test this hypothesis, we investigated if long-term potentiation (LTP) of hippocampal Schaffer-CA1 synapses and spatial navigation memory are altered by a repeated high-intensity sound stimulation (HISS) protocol, consisting of one-minute 120 dB broadband noise applied twice a day for 10 days, in normal Wistar rats and in audiogenic seizure-prone rats (Wistar Audiogenic Rats - WARs). After HISS all WARs exhibited midbrain seizures and 50% of these animals developed limbic recruitment, while only 26% of Wistar rats presented midbrain seizures and none of them had limbic recruitment. In naïve animals, LTP in hippocampal CA1 neurons was induced by 50- or 100-Hz high-frequency stimulation of Schaffer fibres in slices from both Wistar and WAR animals similarly. Surprisingly, HISS suppressed LTP in CA1 neurons in slices from Wistar rats that did not present any seizure, and inhibited LTP in slices from Wistar rats with only midbrain seizures. However HISS had no effect on LTP in CA1 neurons from slices of WARs. Interestingly HISS did not alter spatial navigation and memory in both strains. These findings show that repeated high-intensity sound stimulation prevent LTP of Schaffer-CA1 synapses from Wistar rats, without affecting spatial memory. This effect was not seen in hippocampi from audiogenic seizure-prone WARs. In WARs the link between auditory stimulation and hippocampal LTP seems to be disrupted which could be relevant for the susceptibility to seizures in this strain.


Assuntos
Estimulação Acústica/efeitos adversos , Região CA1 Hipocampal/fisiologia , Potenciação de Longa Duração/fisiologia , Inibição Neural/fisiologia , Vias Neurais/fisiologia , Animais , Modelos Animais de Doenças , Estimulação Elétrica , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas In Vitro , Excitação Neurológica/genética , Excitação Neurológica/patologia , Potenciação de Longa Duração/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Psicoacústica , Ratos , Ratos Wistar , Convulsões/patologia , Convulsões/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA