Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(13)2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37446290

RESUMO

Multiple sclerosis (MS) is a heterogeneous disease of the central nervous system that is governed by neural tissue loss and dystrophy during its progressive phase, with complex reactive pathological cellular changes. The immune-mediated mechanisms that promulgate the demyelinating lesions during relapses of acute episodes are not characteristic of chronic lesions during progressive MS. This has limited our capacity to target the disease effectively as it evolves within the central nervous system white and gray matter, thereby leaving neurologists without effective options to manage individuals as they transition to a secondary progressive phase. The current review highlights the molecular and cellular sequelae that have been identified as cooperating with and/or contributing to neurodegeneration that characterizes individuals with progressive forms of MS. We emphasize the need for appropriate monitoring via known and novel molecular and imaging biomarkers that can accurately detect and predict progression for the purposes of newly designed clinical trials that can demonstrate the efficacy of neuroprotection and potentially neurorepair. To achieve neurorepair, we focus on the modifications required in the reactive cellular and extracellular milieu in order to enable endogenous cell growth as well as transplanted cells that can integrate and/or renew the degenerative MS plaque.


Assuntos
Esclerose Múltipla Crônica Progressiva , Esclerose Múltipla , Humanos , Esclerose Múltipla/patologia , Recidiva Local de Neoplasia , Esclerose Múltipla Crônica Progressiva/patologia , Sistema Nervoso Central/patologia , Substância Cinzenta/diagnóstico por imagem , Substância Cinzenta/patologia
2.
Front Cell Neurosci ; 17: 1197492, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37361998

RESUMO

Multiple sclerosis (MS) can progress with neurodegeneration as a consequence of chronic inflammatory mechanisms that drive neural cell loss and/or neuroaxonal dystrophy in the central nervous system. Immune-mediated mechanisms can accumulate myelin debris in the disease extracellular milieu during chronic-active demyelination that can limit neurorepair/plasticity and experimental evidence suggests that potentiated removal of myelin debris can promote neurorepair in models of MS. The myelin-associated inhibitory factors (MAIFs) are integral contributors to neurodegenerative processes in models of trauma and experimental MS-like disease that can be targeted to promote neurorepair. This review highlights the molecular and cellular mechanisms that drive neurodegeneration as a consequence of chronic-active inflammation and outlines plausible therapeutic approaches to antagonize the MAIFs during the evolution of neuroinflammatory lesions. Moreover, investigative lines for translation of targeted therapies against these myelin inhibitors are defined with an emphasis on the chief MAIF, Nogo-A, that may demonstrate clinical efficacy of neurorepair during progressive MS.

3.
Epilepsia ; 64(6): 1684-1693, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36916834

RESUMO

OBJECTIVE: Stress is one of the most commonly reported triggers for seizures in patients with epilepsy, although the mechanisms that mediate this effect are not established. The clinical evidence supporting this is derived from patients' subjective experience of stress, and how this influences their own seizures. Animal models can be used to explore this phenomenon in controlled environments, free from subjective bias. Here, we used genetic absence epilepsy rats from Strasbourg (GAERS), a genetic rat model of absence epilepsy, to explore the influence of stress and stress hormones on spontaneous seizures. METHODS: Adult male GAERS (n = 38) and nonepileptic control (NEC) rats (n = 4) were used. First, rats were subjected to 30-min restraint stress to assess hypothalamic-pituitary-adrenal axis function. Next, we assessed the effects of 30-min noise stress, and cage tilt stress, on spike-wave discharge seizures in GAERS. We then performed pharmacological experiments to assess the direct effects of stress hormones on seizures, including corticosterone, metyrapone, and deoxycorticosterone. RESULTS: GAERS exhibited elevated baseline corticosterone levels, compared to NEC rats. Noise stress and cage tilt stress significantly enhanced seizure incidence (p < .05), but only during stress periods. Exogenous corticosterone administration also significantly increased seizure occurrence (p < .05). Metyrapone, an inhibitor of corticosterone synthesis, completely abolished seizures in GAERS, and seizures remained suppressed for >2 h. However, deoxycorticosterone, the precursor of corticosterone, increased seizures. SIGNIFICANCE: These results suggest that GAERS exhibit elevations in stress hormones, and this may contribute to seizures. Inhibiting corticosterone synthesis with metyrapone prevents seizures in GAERS, and shows potential for repurposing this drug as a future antiseizure medication.


Assuntos
Epilepsia Tipo Ausência , Humanos , Ratos , Masculino , Animais , Epilepsia Tipo Ausência/genética , Metirapona/farmacologia , Corticosterona , Sistema Hipotálamo-Hipofisário , Alta do Paciente , Eletroencefalografia , Sistema Hipófise-Suprarrenal , Convulsões , Desoxicorticosterona , Modelos Animais de Doenças
4.
Neurol Sci ; 44(7): 2509-2516, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36856905

RESUMO

BACKGROUND: Despite the primary myelin-related pathophysiology, small fiber neuropathy (SFN) and axonal degeneration are also considered to be involved and associated with disabling symptoms and impaired quality of life in chronic inflammatory demyelinating polyneuropathy (CIDP). Demonstration of SFN usually requires complex or invasive investigations. OBJECTS: In vivo corneal confocal microscopy (IVCCM) has evolved as a non-invasive, easily applied method for quantification of small fiber involvement in peripheral nerve disorders. We aimed to investigate the potential role of IVCCM in CIDP. METHODS: In this cross-sectional study, 15 patients with CIDP underwent assessment with clinical disability scales, neuropathic pain (NP) and autonomic symptom questionnaires, nerve conduction studies, and IVCCM. IVCCM parameters were analyzed and compared to those from 32 healthy controls. RESULTS: Corneal nerve fiber density (CNFD) and corneal nerve fiber length (CNFL) were significantly decreased in the CIDP group, compared to those in controls (p = 0.03 and p = 0.024, respectively). Langerhans cells and fiber tortuosity were increased in CIDP patients (p = 0.005 and p = 0.001, respectively). IVCCM parameters were significantly lower in patients with NP compared to those in patients without NP. CONCLUSION: IVCCM shows promise as a non-invasive complementary biomarker in the assessment of demyelinating polyneuropathies, providing insights into the potential pathophysiology of these non-length-dependent neuropathies.


Assuntos
Polirradiculoneuropatia Desmielinizante Inflamatória Crônica , Humanos , Estudos Transversais , Qualidade de Vida , Fibras Nervosas , Córnea/diagnóstico por imagem , Córnea/inervação , Microscopia Confocal/métodos
6.
Cells ; 11(23)2022 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-36497029

RESUMO

Current therapeutics targeting chronic phases of multiple sclerosis (MS) are considerably limited in reversing the neural damage resulting from repeated inflammation and demyelination insults in the multi-focal lesions. This inflammation is propagated by the activation of microglia, the endogenous immune cell aiding in the central nervous system homeostasis. Activated microglia may transition into polarized phenotypes; namely, the classically activated proinflammatory phenotype (previously categorized as M1) and the alternatively activated anti-inflammatory phenotype (previously, M2). These transitional microglial phenotypes are dynamic states, existing as a continuum. Shifting microglial polarization to an anti-inflammatory status may be a potential therapeutic strategy that can be harnessed to limit neuroinflammation and further neurodegeneration in MS. Our research has observed that the obstruction of signaling by inhibitory myelin proteins such as myelin-associated inhibitory factor, Nogo-A, with its receptor (NgR), can regulate microglial cell function and activity in pre-clinical animal studies. Our review explores the microglial role and polarization in MS pathology. Additionally, the potential therapeutics of targeting Nogo-A/NgR cellular mechanisms on microglia migration, polarization and phagocytosis for neurorepair in MS and other demyelination diseases will be discussed.


Assuntos
Microglia , Esclerose Múltipla , Animais , Microglia/metabolismo , Proteínas Nogo/metabolismo , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Transdução de Sinais , Proteínas da Mielina/metabolismo , Inflamação/patologia
7.
Exp Neurol ; 354: 114088, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35461829

RESUMO

OBJECTIVE: Early life stressors are well-established risk factors for psychiatric disorders, and evidence also suggests that these promote vulnerability to epilepsy. Given the high prevalence of psychiatric disorders in epilepsy, early life stress may represent a common driver for these comorbidities. We used animal modelling to investigate the effects of early life stress on epileptogenesis and depressive behaviors, also exploring HPA axis programming as a potential associative mechanism. METHODS: From post-natal day 2-9, Wistar rat dams (n = 3) and their offspring were exposed to the Limited Bedding and Nesting (LBN) model of early life adversity. Control dams (n = 3) were undisturbed. Maternal care was video-recorded, and behavior scored. As adults, rats (n = 7/group) underwent kainic acid-induced status epilepticus (SE), to trigger epilepsy development. Spontaneous seizures, depression-like behavior and HPA axis function were quantified. RESULTS: LBN significantly altered aspects of maternal care, including markedly reducing the consistency of care (p < 0.05), compared to control conditions. Following SE, LBN rats exhibited significantly accelerated epileptogenesis (p = 0.01) and greater disease severity (p = 0.001), compared to control rats. Anhedonia and behavioral despair were observed in epileptic rats exposed to LBN. LBN rats showed significantly dampened HPA axis responsivity, but epileptic rats showed greater corticosterone responses to CRH administration (all p < 0.05). SIGNIFICANCE: Early life adversity promotes a vulnerability to experimental epileptogenesis. These two 'hits' (early life stress and epilepsy) interact to create a depressive-like phenotype, but effects on HPA axis are complex and contrasting. This has implications for the mechanisms underpinning the increased prevalence of psychiatric disorders observed in people with epilepsy.


Assuntos
Experiências Adversas da Infância , Epilepsia , Animais , Corticosterona , Depressão/etiologia , Modelos Animais de Doenças , Humanos , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Ratos , Ratos Wistar , Estresse Psicológico/complicações
8.
Epilepsia ; 60(8): 1650-1660, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31335966

RESUMO

OBJECTIVE: Cognitive deficits are commonly observed in people with epilepsy, but the biologic causation of these is challenging to identify. Animal models of epilepsy can be used to explore pathophysiologic mechanisms leading to cognitive problems, as well as to test novel therapeutics. We utilized a well-validated animal model of epilepsy to explore cognitive deficits using novel translational assessment tools/automated rodent touchscreen assays. METHODS: To induce epilepsy, adult Wistar rats were subjected to kainic acid-induced status epilepticus or sham control (n = 12/group). Two months following induction, animals underwent the Pairwise Discrimination and Reversal learning touchscreen tasks, novel object recognition, and the Y maze test of spatial memory. RESULTS: In the Pairwise Discrimination paradigm, only 40% of epilepsy animals acquired the discrimination learning criterion, compared to 100% of sham animals (P = 0.003). Epilepsy and sham animals that successfully acquired the discrimination progressed onto the reversal phase, which measures cognitive flexibility. Of interest, there were no differences in the rate of reversal learning; however, on the first reversal session, epilepsy rats committed more perseverative errors than shams (mean ± SEM: 6.3 ± 0.9 vs 1.8 ± 0.5, P < 0.0001). Additional behavioral analysis revealed that epilepsy rats were significantly impaired in novel object recognition and short-term spatial learning and memory. SIGNIFICANCE: Using translationally relevant behavioral tools in combination with traditional assays to measure cognition in animal models, here we identify impairments in learning and memory, and enhanced perseverative behaviors in rats with epilepsy. These tools can be used in future research to explore biologic mechanisms and treatments for cognitive deficits associated with epilepsy.


Assuntos
Disfunção Cognitiva/etiologia , Epilepsia do Lobo Temporal/complicações , Animais , Disfunção Cognitiva/diagnóstico , Disfunção Cognitiva/psicologia , Aprendizagem por Discriminação , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/psicologia , Ácido Caínico/farmacologia , Masculino , Aprendizagem em Labirinto , Ratos , Ratos Wistar , Reconhecimento Psicológico , Reversão de Aprendizagem , Memória Espacial
9.
Exp Neurol ; 320: 112979, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31229637

RESUMO

High mobility group box protein-1 (HMGB1) has been implicated as a key mediator of neuroinflammation and neurodegeneration in a range of neurological conditions including traumatic brain injury (TBI) and epilepsy. To date, however, most studies have examined only acute outcomes, and the adult brain. We have recently demonstrated HMGB1 release after experimental TBI in the pediatric mouse. This study therefore examined the chronic consequences of acute HMGB1 inhibition in the same model, to test the hypothesis that HMGB1 is a pivotal mediator of neuropathological, neurobehavioral, and epilepsy outcomes in pediatric TBI. HMGB1 was inhibited by treatment with 50 mg/kg i.p. Glycyrrhizin (Gly), compared to vehicle controls, commencing 1 h prior to moderate TBI or sham surgery in post-natal day 21 mice. We first demonstrated that Gly reduced brain HMGB1 levels and brain edema at an acute time point of 3 days post-injury. Subsequent analysis over a chronic time course found that pediatric TBI resulted in short-term spatial memory and motor learning deficits alongside an apparent increase in hippocampal microglial reactivity, which was prevented in Gly-treated TBI mice. In contrast, Gly treatment did not reduce the severity of evoked seizures, the proportion of animals exhibiting chronic spontaneous seizure activity, or cortical tissue loss. Together, our findings contribute to a growing appreciation for HMGB1's role in neuropathology and associated behavioral outcomes after TBI. However, further work is needed to fully elucidate the contribution of HMGB1 to epileptogenesis in this context.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Proteína HMGB1/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/patologia , Ácido Glicirrízico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Convulsões/etiologia
10.
Epilepsia ; 59(7): e114-e119, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29858497

RESUMO

Patients with epilepsy often have mood disorders, and these are commonly treated with antidepressant drugs. Although these drugs are often successful in mitigating depressive symptoms, how they affect the epileptogenic processes has been little studied. Recent evidence has demonstrated that treatment with selective serotonin reuptake inhibitor (SSRI) antidepressant drugs adversely promotes epileptogenesis, which may be of great concern considering the number of patients exposed to these drugs. This study investigated 5-HT2A receptor signaling as a potential mechanism driving the pro-epileptogenic effects of the prototypical SSRI fluoxetine. Male homozygous 5-HT2A receptor knockout mice or wild-type littermates (n = 9-14/group) were treated with continuous fluoxetine (10 mg kg-1 d-1 , sc) or vehicle and subjected to electrical kindling of the amygdala. Compared to vehicle, fluoxetine treatment accelerated kindling epileptogenesis (P < .001), but there was no effect of genotype (P = .75), or any treatment x genotype interaction observed (P = .90). Of interest, fluoxetine treatment increased afterdischarge thresholds in both genotypes (P = .007). We conclude that treatment with fluoxetine promotes epileptogenesis in mice, but this effect is not mediated by 5-HT2A receptors. This suggests that antidepressants may accelerate the onset of acquired epilepsy in patients who have experienced epileptogenic cerebral insults.


Assuntos
Fluoxetina/farmacologia , Excitação Neurológica/efeitos dos fármacos , Receptor 5-HT2A de Serotonina/efeitos dos fármacos , Convulsões/induzido quimicamente , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Genótipo , Homozigoto , Assistência de Longa Duração , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 5-HT2A de Serotonina/genética , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos
11.
Neurol Genet ; 4(3): e236, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29725622

RESUMO

OBJECTIVE: To determine whether the GNAQ R183Q mutation is present in the forme fruste cases of Sturge-Weber syndrome (SWS) to establish a definitive molecular diagnosis. METHODS: We used sensitive droplet digital PCR (ddPCR) to detect and quantify the GNAQ mutation in tissues from epilepsy surgery in 4 patients with leptomeningeal angiomatosis; none had ocular or cutaneous manifestations. RESULTS: Low levels of the GNAQ mutation were detected in the brain tissue of all 4 cases-ranging from 0.42% to 7.1% frequency-but not in blood-derived DNA. Molecular evaluation confirmed the diagnosis in 1 case in which the radiologic and pathologic data were equivocal. CONCLUSIONS: We detected the mutation at low levels, consistent with mosaicism in the brain or skin (1.0%-18.1%) of classic cases. Our data confirm that the forme fruste is part of the spectrum of SWS, with the same molecular mechanism as the classic disease and that ddPCR is helpful where conventional diagnosis is uncertain.

12.
Epileptic Disord ; 19(4): 450-455, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29258966

RESUMO

Somatic mutation of the lissencephaly-1 gene is a cause of subcortical band heterotopia ("double cortex"). The severity of the phenotype depends on the level of mutation in brain tissue. Detecting and quantifying low-level somatic mosaic mutations is challenging. Here, we utilized droplet digital PCR, a sensitive method to detect low-level mutation. Droplet digital PCR was used in concert with classic genotyping techniques (SNaPshot assays and pyrosequencing) to detect and characterize the tissue mosaicism of a somatic mutation (LIS1 c.190A>T; p.K64X) in a patient with posterior bilateral SBH and refractory epilepsy. The high sensitivity of droplet digital PCR and the ability to target individual DNA molecules allowed us to detect the mutation at low level in the brain, despite the low quality of the DNA sample derived from formalin-fixed paraffin-embedded tissue. This low mutation frequency in the brain was consistent with the relatively subtle malformation resolved by magnetic resonance imaging. The presence of the mutation in other tissues from the patient permitted us to predict the timing of mutagenesis. This sensitive methodology will have utility for a variety of other brain malformation syndromes associated with epilepsy for which historical pathological specimens are available and specific somatic mosaic mutations are predicted.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda/genética , Epilepsia Resistente a Medicamentos/genética , Epilepsias Parciais/genética , Proteínas Associadas aos Microtúbulos/genética , Mutação , Adulto , Feminino , Humanos , Deficiência Intelectual/genética , Transtornos do Desenvolvimento da Linguagem/genética , Reação em Cadeia da Polimerase em Tempo Real
13.
Neurobiol Dis ; 105: 245-256, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28602636

RESUMO

The Aristaless-related homeobox gene (ARX) is a known intellectual disability (ID) gene that frequently presents with X-linked infantile spasm syndrome as a comorbidity. ID with epilepsy in children is a chronic and devastating disorder that has poor treatment options and disease outcomes. To gain a better understanding of the role that mutations in ARX play in ID and epilepsy, we investigate ARX patient mutations modelled in mice. Over half of all ARX mutations result from expansions of the first two polyalanine (PA1 and PA2 respectively) tracts. However, phenotypic data for the mouse modelling the more frequent ARX PA2 dup24 mutation in patients has not been reported and constitutes a barrier to understanding the molecular mechanisms involved. Here we report the first comprehensive analysis of postnatal outcomes for mice modelling disease-causing expansions to both PA1 and PA2 tracts. Both strains were found to have impaired learning and memory, reduced activity, increased anxiety and reduced sociability; with PA1 mice generally displaying greater behavioural deficits in keeping with the more severe phenotype reported in patients. In agreement with previous reports, 70% of PA1 males exhibit myoclonic seizures by two months of age, with the first observed at P18. In this report, we show 80% of PA2 males also display myoclonic seizures, with the first observed at P19. Consistent with patient phenotypes, we observe large variations in seizure progression and severity for both PA1 and PA2 individual mice. The generation of this comprehensive baseline data is a necessary step on the path to the development of therapies to improve patient outcomes.


Assuntos
Epilepsia/genética , Epilepsia/fisiopatologia , Proteínas de Homeodomínio/metabolismo , Deficiência Intelectual/genética , Deficiência Intelectual/fisiopatologia , Peptídeos/genética , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Modelos Animais de Doenças , Eletroencefalografia , Feminino , Lateralidade Funcional , Genótipo , Proteínas de Homeodomínio/genética , Masculino , Transtornos Mentais/etiologia , Transtornos Mentais/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Comportamento Social , Estatísticas não Paramétricas , Fatores de Transcrição/genética , Gravação em Vídeo
14.
Sci Rep ; 7: 40127, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28054653

RESUMO

Despite their abundance, the molecular functions of long non-coding RNAs in mammalian nervous systems remain poorly understood. Here we show that the long non-coding RNA, NEAT1, directly modulates neuronal excitability and is associated with pathological seizure states. Specifically, NEAT1 is dynamically regulated by neuronal activity in vitro and in vivo, binds epilepsy-associated potassium channel-interacting proteins including KCNAB2 and KCNIP1, and induces a neuronal hyper-potentiation phenotype in iPSC-derived human cortical neurons following antisense oligonucleotide knockdown. Next generation sequencing reveals a strong association of NEAT1 with increased ion channel gene expression upon activation of iPSC-derived neurons following NEAT1 knockdown. Furthermore, we show that while NEAT1 is acutely down-regulated in response to neuronal activity, repeated stimulation results in NEAT1 becoming chronically unresponsive in independent in vivo rat model systems relevant to temporal lobe epilepsy. We extended previous studies showing increased NEAT1 expression in resected cortical tissue from high spiking regions of patients suffering from intractable seizures. Our results indicate a role for NEAT1 in modulating human neuronal activity and suggest a novel mechanistic link between an activity-dependent long non-coding RNA and epilepsy.


Assuntos
Encéfalo/fisiologia , Excitabilidade Cortical , Neurônios/fisiologia , RNA Longo não Codificante/metabolismo , Convulsões/patologia , Animais , Células Cultivadas , Humanos , Proteínas Interatuantes com Canais de Kv/metabolismo , Células-Tronco Pluripotentes/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ligação Proteica , Ratos , Superfamília Shaker de Canais de Potássio
15.
Am J Hum Genet ; 99(2): 423-9, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27453577

RESUMO

Hypothalamic hamartoma (HH) with gelastic epilepsy is a well-recognized drug-resistant epilepsy syndrome of early life.(1) Surgical resection allows limited access to the small deep-seated lesions that cause the disease. Here, we report the results of a search for somatic mutations in paired hamartoma- and leukocyte-derived DNA samples from 38 individuals which we conducted by using whole-exome sequencing (WES), chromosomal microarray (CMA), and targeted resequencing (TRS) of candidate genes. Somatic mutations were identified in genes involving regulation of the sonic hedgehog (Shh) pathway in 14/38 individuals (37%). Three individuals had somatic mutations in PRKACA, which encodes a cAMP-dependent protein kinase that acts as a repressor protein in the Shh pathway, and four subjects had somatic mutations in GLI3, an Shh pathway gene associated with HH. In seven other individuals, we identified two recurrent and three single brain-tissue-specific, large copy-number or loss-of-heterozygosity (LOH) variants involving multiple Shh genes, as well as other genes without an obvious biological link to the Shh pathway. The Shh pathway genes in these large somatic lesions include the ligand itself (SHH and IHH), the receptor SMO, and several other Shh downstream pathway members, including CREBBP and GLI2. Taken together, our data implicate perturbation of the Shh pathway in at least 37% of individuals with the HH epilepsy syndrome, consistent with the concept of a developmental pathway brain disease.


Assuntos
Epilepsias Parciais/genética , Hamartoma/genética , Proteínas Hedgehog/metabolismo , Doenças Hipotalâmicas/genética , Mutação/genética , Transdução de Sinais/genética , Proteína de Ligação a CREB/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Exoma/genética , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/genética , Perda de Heterozigosidade , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteína Gli2 com Dedos de Zinco , Proteína Gli3 com Dedos de Zinco
16.
Neurobiol Dis ; 93: 129-36, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27185593

RESUMO

INTRODUCTION: The absence epilepsies are presumed to be caused by genetic factors, but the influence of environmental exposures on epilepsy development and severity, and whether this influence is transmitted to subsequent generations, is not well known. We assessed the effects of environmental enrichment on epilepsy and anxiety outcomes in multiple generations of GAERS - a genetic rat model of absence epilepsy that manifests comorbid elevated anxiety-like behaviour. METHODS: GAERS were exposed to environmental enrichment or standard housing beginning either prior to, or after epilepsy onset, and underwent EEG recordings and anxiety testing. Then, we exposed male GAERS to early enrichment or standard housing and generated F1 progeny, which also underwent EEG recordings. Hippocampal CRH mRNA expression and DNA methylation were assessed using RT-PCR and pyrosequencing, respectively. RESULTS: Early environmental enrichment delayed the onset of epilepsy in GAERS, and resulted in fewer seizures in adulthood, compared with standard housed GAERS. Enrichment also reduced the frequency of seizures when initiated in adulthood. Anxiety levels were reduced by enrichment, and these anti-epileptogenic and anxiolytic effects were heritable into the next generation. We also found reduced expression of CRH mRNA in GAERS exposed to enrichment, but this was not due to changes in DNA methylation. CONCLUSIONS: Environmental enrichment produces disease-modifying effects on genetically determined absence epilepsy and anxiety, and these beneficial effects are transferable to the subsequent generation. Reduced CRH expression was associated with these phenotypic improvements. Environmental stimulation holds promise as a naturalistic therapy for genetically determined epilepsy which may benefit subsequent generations.


Assuntos
Ansiedade/genética , Encéfalo/fisiopatologia , Epilepsia Tipo Ausência/genética , Convulsões/complicações , Animais , Transtornos de Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Meio Ambiente , Ratos
17.
Epilepsia ; 57(3): 484-94, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26786278

RESUMO

OBJECTIVE: Environmental exposures impart powerful effects on vulnerability to many brain diseases, including epilepsy. Mesial temporal lobe epilepsy (MTLE) is a common form of epilepsy, and it is often accompanied by neuropsychiatric comorbidities. This study tests the hypothesis that environmental enrichment (EE) confers antiepileptogenic, psychoprotective, and neuroprotective effects in the amygdala kindling model of MTLE, and explores potential neurobiologic mechanisms. METHODS: At weaning, male Wistar rats were allocated into either EE (large cages containing running wheels and toys; n = 43) or standard housing (SH; standard laboratory cages; n = 39) conditions. At P56, a bipolar electrode was implanted into the left amygdala, and rats underwent rapid amygdala kindling until experiencing five class V seizures (Racine scale, fully kindled). The elevated plus maze was used to assess anxiety. Postmortem histologic and molecular analyses investigated potential biologic mediators of effects. RESULTS: EE significantly delayed kindling epileptogenesis, with EE rats requiring a significantly greater number of kindling stimulations to reach a fully kindled state compared to SH rats (p < 0.05). EE and kindling both reduced anxiety (p < 0.05). Timm's staining revealed significant reductions in aberrant mossy fiber sprouting in EE rats (p < 0.05), and these effects of EE were accompanied by reduced expression of TrkB and CRH genes. SIGNIFICANCE: We identify beneficial effects of EE on vulnerability to limbic epileptogenesis and anxiety, and identify reduced pathologic neuroplasticity and plasticity-related gene expression as potential underlying mechanisms. Enhanced environmental stimulation represents a potential antiepileptogenic strategy that might also mitigate the common psychiatric comorbidities of MTLE.


Assuntos
Meio Ambiente , Epilepsia do Lobo Temporal/fisiopatologia , Abrigo para Animais , Plasticidade Neuronal , Tonsila do Cerebelo/fisiopatologia , Animais , Epilepsia do Lobo Temporal/patologia , Masculino , Plasticidade Neuronal/fisiologia , Ratos , Ratos Wistar , Fatores de Tempo
18.
J Neuroinflammation ; 12: 238, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26683475

RESUMO

BACKGROUND: Repeated mild traumatic brain injuries, such as concussions, may result in cumulative brain damage, neurodegeneration and other chronic neurological impairments. There are currently no clinically available treatment options known to prevent these consequences. However, growing evidence implicates neuroinflammation and oxidative stress in the pathogenesis of repetitive mild brain injuries; thus, these may represent potential therapeutic targets. Progesterone has been demonstrated to have potent anti-inflammatory and anti-oxidant properties after brain insult; therefore, here, we examined progesterone treatment in rats given repetitive mild brain injuries via the repeated mild fluid percussion injury model. METHODS: Male Long-Evans rats were assigned into four groups: sham injury + vehicle treatment, sham injury + progesterone treatment (8 mg/kg/day), repeated mild fluid percussion injuries + vehicle treatment, and repeated mild fluid percussion injuries + progesterone treatment. Rats were administered a total of three injuries, with each injury separated by 5 days. Treatment was initiated 1 h after the first injury, then administered daily for a total of 15 days. Rats underwent behavioural testing at 12-weeks post-treatment to assess cognition, motor function, anxiety and depression. Brains were then dissected for analysis of markers for neuroinflammation and oxidative stress. Ex vivo MRI was conducted in order to examine structural brain damage and white matter integrity. RESULTS: Repeated mild fluid percussion injuries + progesterone treatment rats showed significantly reduced cognitive and sensorimotor deficits compared to their vehicle-treated counterparts at 12-weeks post-treatment. Progesterone treatment significantly attenuated markers of neuroinflammation and oxidative stress in rats given repeated mild fluid percussion injuries, with concomitant reductions in grey and white matter damage as indicated by MRI. CONCLUSIONS: These findings implicate neuroinflammation and oxidative stress in the pathophysiological aftermath of mild brain injuries and suggest that progesterone may be a viable treatment option to mitigate these effects and their detrimental consequences.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças , Estresse Oxidativo/efeitos dos fármacos , Progesterona/uso terapêutico , Animais , Concussão Encefálica/tratamento farmacológico , Concussão Encefálica/metabolismo , Concussão Encefálica/patologia , Lesões Encefálicas/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Destreza Motora/efeitos dos fármacos , Destreza Motora/fisiologia , Estresse Oxidativo/fisiologia , Progesterona/farmacologia , Ratos , Ratos Long-Evans , Fatores de Tempo , Resultado do Tratamento
19.
Epilepsia ; 55(4): 609-20, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24592881

RESUMO

OBJECTIVE: Evidence from animal and human studies indicates that epilepsy can affect cardiac function, although the molecular basis of this remains poorly understood. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels generate pacemaker activity and modulate cellular excitability in the brain and heart, with altered expression and function associated with epilepsy and cardiomyopathies. Whether HCN expression is altered in the heart in association with epilepsy has not been investigated previously. We studied cardiac electrophysiologic properties and HCN channel subunit expression in rat models of genetic generalized epilepsy (Genetic Absence Epilepsy Rats from Strasbourg, GAERS) and acquired temporal lobe epilepsy (post-status epilepticus SE). We hypothesized that the development of epilepsy is associated with altered cardiac electrophysiologic function and altered cardiac HCN channel expression. METHODS: Electrocardiography studies were recorded in vivo in rats and in vitro in isolated hearts. Cardiac HCN channel messenger RNA (mRNA) and protein expression were measured using quantitative PCR and Western blotting respectively. RESULTS: Cardiac electrophysiology was significantly altered in adult GAERS, with slower heart rate, shorter QRS duration, longer QTc interval, and greater standard deviation of RR intervals compared to control rats. In the post-SE model, we observed similar interictal changes in several of these parameters, and we also observed consistent and striking bradycardia associated with the onset of ictal activity. Molecular analysis demonstrated significant reductions in cardiac HCN2 mRNA and protein expression in both models, providing a molecular correlate of these electrophysiologic abnormalities. SIGNIFICANCE: These results demonstrate that ion channelopathies and cardiac dysfunction can develop as a secondary consequence of chronic epilepsy, which may have relevance for the pathophysiology of cardiac dysfunction in patients with epilepsy.


Assuntos
Canalopatias/genética , Técnicas Eletrofisiológicas Cardíacas , Epilepsia Tipo Ausência/genética , Epilepsia do Lobo Temporal/genética , Frequência Cardíaca/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais de Potássio/genética , Animais , Canalopatias/fisiopatologia , Técnicas Eletrofisiológicas Cardíacas/métodos , Epilepsia Tipo Ausência/fisiopatologia , Epilepsia do Lobo Temporal/fisiopatologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/biossíntese , Masculino , Canais de Potássio/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar
20.
Neurobiol Dis ; 63: 194-200, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24321434

RESUMO

OBJECTIVES: Due to the high comorbidity of epilepsy and depression, antidepressant treatment is commonly indicated for patients with epilepsy. Studies in humans and animal models suggest that selective serotonin reuptake inhibitors (SSRIs) may reduce seizure frequency and severity, and these drugs are generally considered safe for use in epilepsy. No studies have investigated the effects of SSRIs on epileptogenesis, the neurobiological process underlying the development of the epileptic state. METHODS: The effect of continuous infusion of the SSRI, fluoxetine (10mg/kg/day sc), versus vehicle control on amygdala kindling was examined in adult male Wistar rats. Seizure threshold and kindling rates were compared between SSRI-treated rats and controls. The study was then repeated examining the effect of a different SSRI, citalopram (10mg/kg/day sc), versus vehicle control. Hippocampal mRNA expression of the serotonin transporter (SERT) and the 5-HT1A receptor was examined in the brains of the rats post-mortem. RESULTS: Treatment with either fluoxetine or citalopram significantly accelerated kindling epileptogenesis, as evidenced by fewer stimulations to reach Class V seizures compared to their respective vehicle-treated group (p<0.01 for both drugs). Seizure duration was also increased in fluoxetine-treated rats. No differences in seizure threshold were observed between treatments (p>0.05). mRNA analysis did not reveal any molecular changes which were common to both treatments. CONCLUSIONS: The rate of epileptogenesis in rats is enhanced by chronic treatment with SSRIs. This could potentially have implications regarding the effect of SSRIs on the development or progression of human epilepsy.


Assuntos
Antidepressivos/uso terapêutico , Epilepsia/tratamento farmacológico , Epilepsia/etiologia , Fluoxetina/uso terapêutico , Excitação Neurológica , Tonsila do Cerebelo/fisiopatologia , Análise de Variância , Animais , Sistemas de Liberação de Medicamentos , Estimulação Elétrica/efeitos adversos , Epilepsia/sangue , Regulação da Expressão Gênica/efeitos dos fármacos , Vínculo Humano-Animal , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor 5-HT1A de Serotonina/genética , Receptor 5-HT1A de Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...