Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Ann Neurol ; 92(1): 45-60, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35467038

RESUMO

OBJECTIVE: Infantile spasms are associated with a wide variety of clinical conditions, including perinatal brain injuries. We have created a model in which prolonged infusion of tetrodotoxin (TTX) into the neocortex, beginning in infancy, produces a localized lesion and reproduces the behavioral spasms, electroencephalogram (EEG) abnormalities, and drug responsiveness seen clinically. Here, we undertook experiments to explore the possibility that the growth factor IGF-1 plays a role in generating epileptic spasms. METHODS: We combined long-term video EEG recordings with quantitative immunohistochemical and biochemical analyses to unravel IGF-1's role in spasm generation. Immunohistochemistry was undertaken in surgically resected tissue from infantile spasms patients. We used viral injections in neonatal conditional IGF-1R knock-out mice to show that an IGF-1-derived tripeptide (1-3)IGF-1, acts through the IGF-1 receptor to abolish spasms. RESULTS: Immunohistochemical methods revealed widespread loss of IGF-1 from cortical neurons, but an increase in IGF-1 in the reactive astrocytes in the TTX-induced lesion. Very similar changes were observed in the neocortex from patients with spasms. In animals, we observed reduced signaling through the IGF-1 growth pathways in areas remote from the lesion. To show the reduction in IGF-1 expression plays a role in spasm generation, epileptic rats were treated with (1-3)IGF-1. We provide 3 lines of evidence that (1-3)IGF-1 activates the IGF-1 signaling pathway by acting through the receptor for IGF-1. Treatment with (1-3)IGF-1 abolished spasms and hypsarrhythmia-like activity in the majority of animals. INTERPRETATION: Results implicate IGF-1 in the pathogenesis of infantile spasms and IGF-1 analogues as potential novel therapies for this neurodevelopmental disorder. ANN NEUROL 2022;92:45-60.


Assuntos
Espasmos Infantis , Animais , Modelos Animais de Doenças , Eletroencefalografia/métodos , Humanos , Lactente , Fator de Crescimento Insulin-Like I , Camundongos , Ratos , Espasmo/induzido quimicamente , Espasmos Infantis/induzido quimicamente , Espasmos Infantis/tratamento farmacológico , Tetrodotoxina/farmacologia
2.
Am J Med Genet A ; 191(6): 1619-1625, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36905087

RESUMO

The p-21-activated kinase 1 (PAK1) protein, encoded by the PAK1 gene, is an evolutionarily conserved serine/threonine-protein kinase that regulates key cellular developmental processes. To date, seven de novo PAK1 variants have been reported to cause the Intellectual Developmental Disorder with Macrocephaly, Seizures, and Speech Delay (IDDMSSD). In addition to the namesake features, other common characteristics include structural brain anomalies, delayed development, hypotonia, and dysmorphic features. Here, we report a de novo PAK1 NM_002576.5: c.1409 T > A variant (p.Leu470Gln) identified by trio genome sequencing (GS) in a 13-year-old boy with postnatal macrocephaly, obstructive hydrocephalus, medically refractory epilepsy, spastic quadriplegia, white matter hyperintensities, profound developmental disabilities, and a horseshoe kidney. This is the first recurrently affected residue identified in the protein kinase domain. Combined assessment of the eight pathogenic PAK1 missense variants reveal that the variants cluster in either the protein kinase or autoregulatory domains. Although interpretation of the phenotypic spectrum is limited by the sample size, neuroanatomical alterations were found more often in individuals with PAK1 variants in the autoregulatory domain. In contrast, non-neurological comorbidities were found more often in individuals with PAK1 variants in the protein kinase domain. Together, these findings expand the clinical spectrum of PAK1-associated IDDMSSD and reveal potential correlations with the affected protein domains.


Assuntos
Epilepsia , Hidrocefalia , Deficiência Intelectual , Megalencefalia , Masculino , Humanos , Adolescente , Domínios Proteicos , Proteínas Quinases , Epilepsia/diagnóstico , Epilepsia/genética , Megalencefalia/diagnóstico , Megalencefalia/genética , Deficiência Intelectual/genética , Hidrocefalia/diagnóstico , Hidrocefalia/genética , Quadriplegia/diagnóstico , Quadriplegia/genética , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/química
3.
Ann Neurol ; 89(2): 226-241, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33068018

RESUMO

OBJECTIVE: Epileptic spasms are a hallmark of severe seizure disorders. The neurophysiological mechanisms and the neuronal circuit(s) that generate these seizures are unresolved and are the focus of studies reported here. METHODS: In the tetrodotoxin model, we used 16-channel microarrays and microwires to record electrophysiological activity in neocortex and thalamus during spasms. Chemogenetic activation was used to examine the role of neocortical pyramidal cells in generating spasms. Comparisons were made to recordings from infantile spasm patients. RESULTS: Current source density and simultaneous multiunit activity analyses indicate that the ictal events of spasms are initiated in infragranular cortical layers. A dramatic pause of neuronal activity was recorded immediately prior to the onset of spasms. This preictal pause is shown to share many features with the down states of slow wave sleep. In addition, the ensuing interictal up states of slow wave rhythms are more intense in epileptic than control animals and occasionally appear sufficient to initiate spasms. Chemogenetic activation of neocortical pyramidal cells supported these observations, as it increased slow oscillations and spasm numbers and clustering. Recordings also revealed a ramp-up in the number of neocortical slow oscillations preceding spasms, which was also observed in infantile spasm patients. INTERPRETATION: Our findings provide evidence that epileptic spasms can arise from the neocortex and reveal a previously unappreciated interplay between brain state physiology and spasm generation. The identification of neocortical up states as a mechanism capable of initiating epileptic spasms will likely provide new targets for interventional therapies. ANN NEUROL 2021;89:226-241.


Assuntos
Ondas Encefálicas/fisiologia , Neocórtex/fisiopatologia , Células Piramidais/fisiologia , Espasmos Infantis/fisiopatologia , Tálamo/fisiopatologia , Animais , Modelos Animais de Doenças , Eletrocorticografia , Feminino , Humanos , Lactente , Masculino , Neocórtex/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Bloqueadores dos Canais de Sódio/toxicidade , Espasmo/induzido quimicamente , Espasmo/fisiopatologia , Espasmos Infantis/induzido quimicamente , Tetrodotoxina/toxicidade , Tálamo/efeitos dos fármacos
4.
Epilepsia ; 63(12): 3078-3089, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36179064

RESUMO

OBJECTIVE: Our goal was to determine whether animals with a history of epileptic spasms have learning and memory deficits. We also used continuous (24/7) long-term electroencephalographic (EEG) recordings to evaluate the evolution of epileptiform activity in the same animals over time. METHODS: Object recognition memory and object location memory tests were undertaken, as well as a matching to place water maze test that evaluated working memory. A retrospective analysis was undertaken of long-term video/EEG recordings from rats with epileptic spasms. The frequency and duration of the ictal events of spasms were quantified. RESULTS: Rats with a history of epileptic spasms showed impairment on the three behavioral tests, and their scores on the object recognition memory and matching to place water maze tests indicated neocortical involvement in the observed impaired cognition. Analysis of EEG recordings unexpectedly showed that the ictal events of spasms and their accompanying behaviors progressively increased in duration over a 2-week period soon after onset, after which spasm duration plateaued. At the same time, spasm frequency remained unchanged. Soon after spasm onset, ictal events were variable in wave form but became more stereotyped as the syndrome evolved. SIGNIFICANCE: Our EEG findings are the first to demonstrate progressive ictogenesis for epileptic spasms. Furthermore, in demonstrating cognitive deficits in the tetrodotoxin model, we have met a criterion for an animal model of West syndrome. Animal models will allow in-depth studies of spasm progression's potential role in cognitive regression and may elucidate why early treatment is considered essential for improved neurodevelopmental outcomes in children.


Assuntos
Espasmos Infantis , Ratos , Animais , Estudos Retrospectivos
5.
Epilepsia ; 62(9): 2263-2273, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34258765

RESUMO

OBJECTIVE: Epileptic spasms are a hallmark of a severe epileptic state. A previous study showed neocortical up and down states defined by unit activity play a role in the generation of spasms. However, recording unit activity is challenging in clinical settings, and more accessible neurophysiological signals are needed for the analysis of these brain states. METHODS: In the tetrodotoxin model, we used 16-channel microarrays to record electrophysiological activity in the neocortex during interictal periods and spasms. High-frequency activity (HFA) in the frequency range of fast ripples (200-500 Hz) was analyzed, as were slow wave oscillations (1-8 Hz), and correlated with the neocortical up and down states defined by multiunit activity (MUA). RESULTS: HFA and MUA had high temporal correlation during interictal and ictal periods. Both increased strikingly during interictal up states and ictal events but were silenced during interictal down states and preictal pauses, and their distributions were clustered at the peak of slow oscillations in local field potential recordings. In addition, both HFA power and MUA firing rates were increased to a greater extent during spasms than interictal up states. During non-rapid eye movement sleep, the HFA rhythmicity faithfully followed the MUA up and down states, but during rapid eye movement sleep when MUA up and down states disappeared the HFA rhythmicity was largely absent. We also observed an increase in the number of HFA down state minutes prior to ictal onset, consistent with the results from analyses of MUA down states. SIGNIFICANCE: This study provides evidence that HFA may serve as a biomarker for the pathological up states of epileptic spasms. The availability of HFA recordings makes this a clinically practical technique. These findings will likely provide a novel approach for localizing and studying epileptogenic neocortical networks not only in spasms patients but also in other types of epilepsy.


Assuntos
Epilepsia , Neocórtex , Espasmos Infantis , Animais , Biomarcadores , Modelos Animais de Doenças , Eletroencefalografia , Humanos , Lactente , Espasmo
6.
Epilepsy Behav ; 116: 107786, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33548914

RESUMO

Studies were undertaken to evaluate the effectiveness of Acthar® Gel (repository corticotropin injection [RCI]) in the tetrodotoxin (TTX) model of early-life-induced epileptic spasms. Repository corticotropin injection (RCI) is widely used in the United States to treat infantile spasms. A major component of RCI is N25 deamidated ACTH. Additionally, we hoped to provide some insight into the possible role circulating corticosteroids play in spasm cessation by comparing the RCI dose-response relationships for spasm suppression to RCI-induced corticosterone release from the adrenal gland. Spasms were induced by chronic TTX infusion into the neocortex beginning on postnatal day 11. Repository corticotropin injection (RCI) dosages were between 8 and 32 IU/kg/day. Drug titration protocols were used, and comparisons were made to injections of a vehicle gel. Video/EEG recordings (24/7) monitored the drug's effects continuously for up to 2 months. Tetrodotoxin (TTX)-infused control rats were monitored for the same period of time. In separate experiments, the same dosages of RCI were given to rats and 1 h later plasma was collected and assayed for corticosterone. A parallel study compared the effects of 1-day and 10-day RCI treatments on circulating corticosterone. Results showed that RCI was ineffective at dosages of 8, 12, and 16 IU/kg/day but eliminated spasms in 66% of animals treated with 24 or 32 IU/kg/day. Treating animals with 32 IU/kg/day alone produced the same degree of spasms suppression as observed during the titration protocols. In rats that had hypsarrhythmia-like activity, RCI eliminated this abnormal interictal EEG pattern in all rats that became seizure-free. In terms of plasma corticosterone, 1- and 10-day treatments with RCI produced similar increases in this hormone and the levels increased linearly with increasing dosages of RCI. This stood in sharp contrast to the sigmoid-like dose-response curve for decreases in spasm counts. Our results further validate the TTX model as relevant for the study of infantile spasms. The model should be useful for investigating how RCI acts to eliminate seizures and hypsarrhythmia. Dose-response results suggest that either very high concentrations of circulating corticosteroids are required to abolish spasms or RCI acts through a different mechanism.


Assuntos
Neocórtex , Espasmos Infantis , Hormônio Adrenocorticotrópico , Animais , Modelos Animais de Doenças , Eletroencefalografia , Ratos , Espasmo , Espasmos Infantis/induzido quimicamente , Espasmos Infantis/tratamento farmacológico
7.
Nature ; 528(7580): 123-6, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26605526

RESUMO

Copy number variations have been frequently associated with developmental delay, intellectual disability and autism spectrum disorders. MECP2 duplication syndrome is one of the most common genomic rearrangements in males and is characterized by autism, intellectual disability, motor dysfunction, anxiety, epilepsy, recurrent respiratory tract infections and early death. The broad range of deficits caused by methyl-CpG-binding protein 2 (MeCP2) overexpression poses a daunting challenge to traditional biochemical-pathway-based therapeutic approaches. Accordingly, we sought strategies that directly target MeCP2 and are amenable to translation into clinical therapy. The first question that we addressed was whether the neurological dysfunction is reversible after symptoms set in. Reversal of phenotypes in adult symptomatic mice has been demonstrated in some models of monogenic loss-of-function neurological disorders, including loss of MeCP2 in Rett syndrome, indicating that, at least in some cases, the neuroanatomy may remain sufficiently intact so that correction of the molecular dysfunction underlying these disorders can restore healthy physiology. Given the absence of neurodegeneration in MECP2 duplication syndrome, we propose that restoration of normal MeCP2 levels in MECP2 duplication adult mice would rescue their phenotype. By generating and characterizing a conditional Mecp2-overexpressing mouse model, here we show that correction of MeCP2 levels largely reverses the behavioural, molecular and electrophysiological deficits. We also reduced MeCP2 using an antisense oligonucleotide strategy, which has greater translational potential. Antisense oligonucleotides are small, modified nucleic acids that can selectively hybridize with messenger RNA transcribed from a target gene and silence it, and have been successfully used to correct deficits in different mouse models. We find that antisense oligonucleotide treatment induces a broad phenotypic rescue in adult symptomatic transgenic MECP2 duplication mice (MECP2-TG), and corrected MECP2 levels in lymphoblastoid cells from MECP2 duplication patients in a dose-dependent manner.


Assuntos
Dosagem de Genes/genética , Técnicas de Silenciamento de Genes , Genes Duplicados/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteína 2 de Ligação a Metil-CpG/genética , Oligonucleotídeos Antissenso/genética , Fenótipo , Animais , Sítios de Ligação Microbiológicos/genética , Células Cultivadas , Modelos Animais de Doenças , Eletroencefalografia , Duplicação Gênica/genética , Humanos , Integrases/genética , Integrases/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/fisiopatologia , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Transgênicos
8.
J Neurosci ; 37(36): 8595-8611, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28751459

RESUMO

Changes in synaptic strength and connectivity are thought to be a major mechanism through which many gene variants cause neurological disease. Hyperactivation of the PI3K-mTOR signaling network, via loss of function of repressors such as PTEN, causes epilepsy in humans and animal models, and altered mTOR signaling may contribute to a broad range of neurological diseases. Changes in synaptic transmission have been reported in animal models of PTEN loss; however, the full extent of these changes, and their effect on network function, is still unknown. To better understand the scope of these changes, we recorded from pairs of mouse hippocampal neurons cultured in a two-neuron microcircuit configuration that allowed us to characterize all four major connection types within the hippocampus. Loss of PTEN caused changes in excitatory and inhibitory connectivity, and these changes were postsynaptic, presynaptic, and transynaptic, suggesting that disruption of PTEN has the potential to affect most connection types in the hippocampal circuit. Given the complexity of the changes at the synaptic level, we measured changes in network behavior after deleting Pten from neurons in an organotypic hippocampal slice network. Slices containing Pten-deleted neurons showed increased recruitment of neurons into network bursts. Importantly, these changes were not confined to Pten-deleted neurons, but involved the entire network, suggesting that the extensive changes in synaptic connectivity rewire the entire network in such a way that promotes a widespread increase in functional connectivity.SIGNIFICANCE STATEMENT Homozygous deletion of the Pten gene in neuronal subpopulations in the mouse serves as a valuable model of epilepsy caused by mTOR hyperactivation. To better understand how gene deletions lead to altered neuronal activity, we investigated the synaptic and network effects that occur 1 week after Pten deletion. PTEN loss increased the connectivity of all four types of hippocampal synaptic connections, including two forms of increased inhibition of inhibition, and increased network functional connectivity. These data suggest that single gene mutations that cause neurological diseases such as epilepsy may affect a surprising range of connection types. Moreover, given the robustness of homeostatic plasticity, these diverse effects on connection types may be necessary to cause network phenotypes such as increased synchrony.


Assuntos
Potenciais de Ação/fisiologia , Hipocampo/crescimento & desenvolvimento , Rede Nervosa/crescimento & desenvolvimento , Neurônios/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Transmissão Sináptica/fisiologia , Animais , Feminino , Hipocampo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/citologia , Vias Neurais/citologia , Vias Neurais/fisiologia , Neurônios/citologia , PTEN Fosfo-Hidrolase/genética
9.
Neuropathology ; 36(2): 146-56, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26381727

RESUMO

Neuropathology of resected brain tissue has revealed an association of focal cortical dysplasia (FCD) with drug-resistant epilepsy (DRE). Recent studies have shown that the mechanistic target of rapamycin (mTOR) pathway is hyperactivated in FCD as evidenced by increased phosphorylation of the ribosomal protein S6 (S6) at serine 240/244 (S(240/244) ), a downstream target of mTOR. Moreover, extracellular regulated kinase (ERK) has been shown to phosphorylate S6 at serine 235/236 (S(235/236) ) and tuberous sclerosis complex 2 (TSC2) at serine 664 (S(664) ) leading to hyperactive mTOR signaling. We evaluated ERK phosphorylation of S6 and TSC2 in two types of FCD (FCD I and FCD II) as a candidate mechanism contributing to mTOR pathway dysregulation. Tissue samples from patients with tuberous sclerosis (TS) served as a positive control. Immunostaining for phospho-S6 (pS6(240/244) and pS6(235/236) ), phospho-ERK (pERK), and phospho-TSC2 (pTSC2) was performed on resected brain tissue with FCD and TS. We found increased pS6(240/244) and pS6(235/236) staining in FCD I, FCD II and TS compared to normal-appearing tissue, while pERK and pTSC2 staining was increased only in FCD IIb and TS tissue. Our results suggest that both the ERK and mTOR pathways are dysregulated in FCD and TS; however, the signaling alterations are different for FCD I as compared to FCD II and TS.


Assuntos
Epilepsia/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Malformações do Desenvolvimento Cortical do Grupo I/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Adolescente , Criança , Pré-Escolar , Ativação Enzimática , Epilepsia/patologia , Feminino , Humanos , Imuno-Histoquímica , Lactente , Sistema de Sinalização das MAP Quinases/fisiologia , Imageamento por Ressonância Magnética , Masculino , Malformações do Desenvolvimento Cortical do Grupo I/patologia , Fosforilação , Proteína S6 Ribossômica/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
10.
Neurobiol Dis ; 82: 1-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26026423

RESUMO

Abnormal high frequency oscillations (HFOs) in EEG recordings are thought to be reflections of mechanisms responsible for focal seizure generation in the temporal lobe and neocortex. HFOs have also been recorded in patients and animal models of infantile spasms. If HFOs are important contributors to infantile spasms then anticonvulsant drugs that suppress these seizures should decrease the occurrence of HFOs. In experiments reported here, we used long-term video/EEG recordings with digital sampling rates capable of capturing HFOs. We tested the effectiveness of vigabatrin (VGB) in the TTX animal model of infantile spasms. VGB was found to be quite effective in suppressing spasms. In 3 of 5 animals, spasms ceased after a daily two week treatment. In the other 2 rats, spasm frequency dramatically decreased but gradually increased following treatment cessation. In all animals, hypsarrhythmia was abolished by the last treatment day. As VGB suppressed the frequency of spasms, there was a decrease in the intensity of the behavioral spasms and the duration of the ictal EEG event. Analysis showed that there was a burst of high frequency activity at ictal onset, followed by a later burst of HFOs. VGB was found to selectively suppress the late HFOs of ictal complexes. VGB also suppressed abnormal HFOs recorded during the interictal periods. Thus VGB was found to be effective in suppressing both the generation of spasms and hypsarrhythmia in the TTX model. Vigabatrin also appears to preferentially suppress the generation of abnormal HFOs, thus implicating neocortical HFOs in the infantile spasms disease state.


Assuntos
Anticonvulsivantes/uso terapêutico , Neocórtex/efeitos dos fármacos , Espasmos Infantis/tratamento farmacológico , Vigabatrina/uso terapêutico , Animais , Anticonvulsivantes/farmacologia , Mapeamento Encefálico , Modelos Animais de Doenças , Eletroencefalografia , Humanos , Lactente , Masculino , Neocórtex/fisiopatologia , Ratos , Espasmos Infantis/fisiopatologia , Vigabatrina/farmacologia
11.
Adv Exp Med Biol ; 813: 123-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25012372

RESUMO

Maintaining physiological variables within narrow operating limits by homeostatic mechanisms is a fundamental property of most if not all living cells and organisms. In recent years, research from many laboratories has shown that the activity of neurons and neural circuits are also homeostatically regulated. Here, we attempt to apply concepts of homeostasis in general, and more specifically synaptic homeostatic plasticity, to the study of epilepsy. We hypothesize that homeostatic mechanisms are actively engaged in the epileptic brain. These processes attempt to re-establish normal neuronal and network activity, but are opposed by the concurrent mechanisms underlying epileptogenesis. In forms of intractable epilepsy, seizures are so frequent and intense that homeostatic mechanisms are unable to restore normal levels of neuronal activity. In such cases, we contend that homeostatic plasticity mechanisms nevertheless remain active. However, their continuing attempts to reset neuronal activity become maladaptive and results in dyshomeostasis with neurobehavioral consequences. Using the developing hippocampus as a model system, we briefly review experimental results and present a series of arguments to propose that the cognitive neurobehavioral comorbidities of childhood epilepsy result, at least in part, from unchecked homeostatic mechanisms.


Assuntos
Epilepsia/fisiopatologia , Homeostase , Plasticidade Neuronal , Humanos
12.
J Neurosci ; 32(33): 11441-52, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22895726

RESUMO

The mammalian target of rapamycin (mTOR) signaling pathway in neurons integrates a variety of extracellular signals to produce appropriate translational responses. mTOR signaling is hyperactive in neurological syndromes in both humans and mouse models that are characterized by epilepsy, autism, and cognitive disturbances. In addition, rapamycin, a clinically important immunosuppressant, is a specific and potent inhibitor of mTOR signaling. While mTOR is known to regulate growth and synaptic plasticity of glutamatergic neurons, its effects on basic parameters of synaptic transmission are less well studied, and its role in regulating GABAergic transmission is unexplored. We therefore performed an electrophysiological and morphological comparison of glutamatergic and GABAergic neurons in which mTOR signaling was either increased by loss of the repressor Pten or decreased by treatment with rapamycin. We found that hyperactive mTOR signaling increased evoked synaptic responses in both glutamatergic and GABAergic neurons by ∼50%, due to an increase in the number of synaptic vesicles available for release, the number of synapses formed, and the miniature event size. Prolonged (72 h) rapamycin treatment prevented these abnormalities and also decreased synaptic transmission in wild-type glutamatergic, but not GABAergic, neurons. Further analyses suggested that hyperactivation of the mTOR pathway also impairs presynaptic function, possibly by interfering with vesicle fusion. Despite this presynaptic impairment, the net effect of Pten loss is enhanced synaptic transmission in both GABAergic and glutamatergic neurons, which has numerous implications, depending on where in the brain mutations of an mTOR suppressor gene occur.


Assuntos
Glutamatos/metabolismo , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Ácido gama-Aminobutírico/metabolismo , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Biofísica , Corpo Estriado/citologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Estimulação Elétrica , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Glutamatos/farmacologia , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , PTEN Fosfo-Hidrolase/deficiência , Técnicas de Patch-Clamp , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sirolimo/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/genética , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Serina-Treonina Quinases TOR/efeitos dos fármacos , Tetrodotoxina/farmacologia , Fatores de Tempo , Transfecção , Ubiquitina-Proteína Ligases , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/farmacologia
13.
Neurobiol Dis ; 46(2): 377-88, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22342513

RESUMO

While infantile spasms is the most common catastrophic epilepsy of infancy and early-childhood, very little is known about the basic mechanisms responsible for this devastating disorder. In experiments reported here, spasms were induced in rats by the chronic infusion of TTX into the neocortex beginning on postnatal days 10-12. Studies of focal epilepsy suggest that high frequency EEG oscillations (HFOs) occur interictally at sites that are most likely responsible for seizure generation. Thus, our goal was to determine if HFOs occurred and where they occurred in cortex in the TTX model. We also undertook multiunit recordings to begin to analyze the basic mechanisms responsible for HFOs. Our results show that HFOs occur most frequently during hypsarrhythmia and NREM sleep and are most prominent contralateral to the TTX infusion site in the homotopic cortex and anterior to this region in frontal cortex. While HFOs were largest and most frequent in these contralateral regions, they were also commonly recorded synchronously across multiple and widely-spaced recordings sites. The amplitude and spatial distribution of interictal HFOs were found to be very similar to the high frequency bursts seen at seizure onset. However, the latter differed from the interictal events in that the high frequency activity was more intense at seizure onset. Microwire recordings showed that neuronal unit firing increased abruptly with the generation of HFOs. A similar increase in neuronal firing occurred at the onset of the ictal events. Taken together, results suggest that neocortical networks are abnormally excitable, particularly contralateral to TTX infusion, and that these abnormalities are not restricted to small areas of cortex. Multiunit firing coincident with HFOs is fully consistent with a neocortical hyperexcitability hypothesis particularly since they both occur at seizure onset.


Assuntos
Modelos Animais de Doenças , Eletroencefalografia , Epilepsia/induzido quimicamente , Epilepsia/fisiopatologia , Fatores Etários , Animais , Animais Recém-Nascidos , Eletroencefalografia/métodos , Neocórtex/efeitos dos fármacos , Ratos , Espasmo/induzido quimicamente , Espasmo/fisiopatologia , Tetrodotoxina/toxicidade
14.
Epilepsia ; 53 Suppl 1: 116-24, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22612816

RESUMO

Childhood epilepsy can be severe and even catastrophic. In these instances, cognition can be impaired-leading to long-term intellectual disabilities. One factor that could potentially cause cognitive deficits is the frequent seizures that characterize intractable epilepsy. However, it has been difficult to separate the effects seizures may have from those of preexisting neuropathologies and/or the effects of ongoing anticonvulsant therapies. Therefore, important questions are: Do early life seizures produce the learning deficits? And if they do, how do they do it? Results from recent animal models studies reviewed here show that recurrent seizures in infancy stop the growth of CA1 hippocampal dendrites. We speculate that the molecular mechanisms responsible for seizure-induced growth suppression are homeostatic/neuroprotective, used by the developing nervous system in an attempt to limit neuronal and network excitability and prevent the continued generation of seizures. However, by preventing the normal growth of dendrites, there is a reduction in CA1 glutamatergic synapses that supports long-lasting forms of synaptic plasticity thought to be the cellular basis of learning and memory. Therefore, dendrite growth suppression would reduce the neuroanatomic substrates for learning and memory, and in so doing could contribute in important ways to spatial learning and memory deficits that may be relevant to the cognitive deficits associated with childhood epilepsy.


Assuntos
Dendritos/patologia , Deficiências do Desenvolvimento/patologia , Deficiência Intelectual/patologia , Convulsões/patologia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Epilepsia/patologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/patologia , Hipocampo/fisiologia , Humanos , Aprendizagem/fisiologia , Memória/fisiologia , Fármacos Neuroprotetores , Transdução de Sinais/fisiologia
15.
Neurobiol Dis ; 44(2): 205-14, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21777677

RESUMO

Impaired learning and memory are common in epilepsy syndromes of childhood. Clinical investigations suggest that the developing brain may be particularly vulnerable to the effects of intractable seizure disorders. Magnetic resonance imaging (MRI) studies have demonstrated reduced volumes in brain regions involved in learning and memory. The earlier the onset of an epilepsy the larger the effects seem to be on both brain anatomy and cognition. Thus, childhood epilepsy has been proposed to interfere in some unknown way with brain development. Experiments reported here explore these ideas by examining the effects of seizures in infant mice on learning and memory and on the growth of CA1 hippocampal pyramidal cell dendrites. Fifteen brief seizures were induced by flurothyl between postnatal days 7 and 11 in mice that express green fluorescent protein (GFP) in hippocampal pyramidal cells. One to 44days later, dendritic arbors were reconstructed to measure growth. Spatial learning and memory were also assessed in a water maze. Our results show that recurrent seizures produced marked deficits in learning and memory. Seizures also dramatically slowed the growth of basilar dendrites while neurons in littermate control mice continued to add new dendritic branches and lengthen existing branches. When experiments were performed in older mice, seizures had no measureable effects on either dendrite arbor complexity or spatial learning and memory. Our results suggest that the recurring seizures of intractable childhood epilepsy contribute to associated learning and memory deficits by suppressing dendrite growth.


Assuntos
Dendritos/patologia , Epilepsia/fisiopatologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/patologia , Deficiências da Aprendizagem/fisiopatologia , Transtornos da Memória/fisiopatologia , Inibição Neural/fisiologia , Fatores Etários , Animais , Modelos Animais de Doenças , Epilepsia/complicações , Epilepsia/patologia , Feminino , Hipocampo/fisiopatologia , Deficiências da Aprendizagem/etiologia , Deficiências da Aprendizagem/patologia , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
Epilepsia ; 52(1): 53-62, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21204817

RESUMO

PURPOSE: To describe high frequency (HF) electrographic activity accompanying ictal discharges in the tetrodotoxin (TTX) model of infantile spasms. Previous studies of HF oscillations in humans and animals suggest that they arise at sites of seizure onset. We compared HF oscillations at several cortical sites to determine regional differences. METHODS: TTX was infused for 4 weeks into the neocortex of rats beginning on postnatal days 11 or 12. Electroencephalography (EEG) electrodes were implanted 2 weeks later and video-EEG recordings were analyzed between postnatal days 31 and 47. EEG recordings were digitally sampled at 2,048 Hz. HF EEG activity (20-900 Hz) was quantified using compressed spectral arrays and band-pass filtering. KEY FINDINGS: Multiple seizures were analyzed in 10 rats. Ictal onset was associated with multiple bands of rhythmic HF activity that could extend to 700 Hz. The earliest and most intense discharging typically occurred contralaterally to where TTX was infused. HF activity continued to occur throughout the seizure (even during the electrodecrement that is recorded with more traditional filter settings), although there was a gradual decrease of the intensity of the highest frequency components as the amplitude of lower frequency oscillations increased. Higher frequencies sometimes reappeared in association with spike/sharp-waves at seizure termination. SIGNIFICANCE: The findings show that HF EEG activity accompanies ictal events in the TTX model. Results also suggest that the seizures in this model do not originate from the TTX infusion site. Instead HF discharges are usually most intense and occur earliest contralaterally, suggesting that these homologous regions may be involved in seizure generation.


Assuntos
Modelos Animais de Doenças , Eletroencefalografia/métodos , Neocórtex/fisiopatologia , Espasmos Infantis/fisiopatologia , Animais , Animais Recém-Nascidos , Eletroencefalografia/efeitos dos fármacos , Humanos , Recém-Nascido , Neocórtex/efeitos dos fármacos , Ratos , Espasmos Infantis/induzido quimicamente , Espasmos Infantis/diagnóstico , Tetrodotoxina/toxicidade
17.
Elife ; 92020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32073399

RESUMO

Mutations in genes encoding synaptic proteins cause many neurodevelopmental disorders, with the majority affecting postsynaptic apparatuses and much fewer in presynaptic proteins. Syntaxin-binding protein 1 (STXBP1, also known as MUNC18-1) is an essential component of the presynaptic neurotransmitter release machinery. De novo heterozygous pathogenic variants in STXBP1 are among the most frequent causes of neurodevelopmental disorders including intellectual disabilities and epilepsies. These disorders, collectively referred to as STXBP1 encephalopathy, encompass a broad spectrum of neurologic and psychiatric features, but the pathogenesis remains elusive. Here we modeled STXBP1 encephalopathy in mice and found that Stxbp1 haploinsufficiency caused cognitive, psychiatric, and motor dysfunctions, as well as cortical hyperexcitability and seizures. Furthermore, Stxbp1 haploinsufficiency reduced cortical inhibitory neurotransmission via distinct mechanisms from parvalbumin-expressing and somatostatin-expressing interneurons. These results demonstrate that Stxbp1 haploinsufficient mice recapitulate cardinal features of STXBP1 encephalopathy and indicate that GABAergic synaptic dysfunction is likely a crucial contributor to disease pathogenesis.


Assuntos
Encefalopatias/genética , Encefalopatias/patologia , Haploinsuficiência , Proteínas Munc18/genética , Animais , Ansiedade/genética , Comportamento Animal , Peso Corporal/genética , Transtornos Cognitivos/genética , Modelos Animais de Doenças , Genes Letais , Heterozigoto , Membro Posterior/fisiopatologia , Homozigoto , Humanos , Camundongos , Camundongos Knockout , Taxa de Sobrevida , Transmissão Sináptica/genética
18.
Epilepsy Behav ; 14(3): 438-45, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19341977

RESUMO

During the past decade, substantial progress has been made in delineating clinical features of the epilepsies and the basic mechanisms responsible for these disorders. Eleven human epilepsy genes have been identified and many more are now known from animal models. Candidate targets for cures are now based upon newly identified cellular and molecular mechanisms that underlie epileptogenesis. However, epilepsy is increasingly recognized as a group of heterogeneous syndromes characterized by other conditions that co-exist with seizures. Cognitive, emotional and behavioral co-morbidities are common and offer fruitful areas for study. These advances in understanding mechanisms are being matched by the rapid development of new diagnostic methods and therapeutic approaches. This article reviews these areas of progress and suggests specific goals that once accomplished promise to lead to cures for epilepsy.


Assuntos
Epilepsia/diagnóstico , Epilepsia/terapia , Anticonvulsivantes/uso terapêutico , Comorbidade , Epilepsia/complicações , Epilepsia/epidemiologia , Epilepsia/etiologia , Epilepsia/genética , Humanos , Procedimentos Neurocirúrgicos
19.
Brain Res ; 1140: 75-83, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-16996039

RESUMO

The majority of cortical and hippocampal interneurons originate in the subcortical telencephalon and migrate tangentially into pallial regions before settling in various cortical layers. The molecular cues that regulate final positioning of specific interneurons in cortical structures have not yet been identified. The positioning of radially migrating principal neurons of the cortex and hippocampus depends upon Reelin, an extracellular protein expressed near the pial surface during embryonic development that is absent in reeler mutant mice. To determine whether the layer specification of interneurons, like that of principal neurons, requires Reelin, we crossed reeler with transgenic mice that contain Green Fluorescent Protein (GFP)-expressing Inhibitory Neurons (GINs). These neurons express basal forebrain markers Dlx1/2 in normal and reeler mice. In normal mice, GINs express Reelin and are localized to specific layers of the cortex and hippocampus. In reeler mutant mice, we show that GINs migrate normally into the pallium, but fail to acquire proper layer position. Double labeling experiments indicate that the neurochemical profile of these interneurons is not generally altered in reeler mice. However, the extension of their cellular processes is abnormal. Quantitative analysis of GINs in the cortex revealed that they are hypertrophic, bearing longer neuritic branches than normal. Thus, the lack of Reelin signaling results in abnormal positioning and altered morphology of forebrain interneurons.


Assuntos
Dendritos/fisiologia , Interneurônios/citologia , Camundongos Mutantes Neurológicos/anatomia & histologia , Prosencéfalo/anormalidades , Prosencéfalo/citologia , Animais , Animais Recém-Nascidos , Padronização Corporal/fisiologia , Contagem de Células , Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Hipocampo/fisiologia , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Proteína Reelina , Fatores de Transcrição/metabolismo
20.
eNeuro ; 4(2)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28462391

RESUMO

Neurobehavioral abnormalities are commonly associated with intractable childhood epilepsy. Studies from numerous labs have demonstrated cognitive and socialization deficits in rats and mice that have experienced early-life seizures. However, the cellular and molecular mechanisms underlying these effects are unknown. Previously, experiments have shown that recurrent seizures in infancy suppress the growth of hippocampal dendrites at the same time they impair learning and memory. Experiments in slice cultures have also demonstrated dendrite growth suppression. Here, we crossed calcineurin B1 (CaNB1) floxed and Thy1GFP-M mice to produce mice that were homozygous for the both the floxed CaNB1 and the Thy1GFP-M transgene. Littermates that were homozygous for wild-type CaNB1 and Thy1GFP-M served as controls. Hippocampal slice cultures from these mice were transfected with an AAV/hSyn-mCherry-Cre virus to eliminate CaNB1 from neurons. Immunohistochemical results showed that CaNB1 was eliminated from at least 90% of the transfected CA1 pyramidal cells. Moreover, the CaN-dependent nuclear translocation of the CREB transcription coactivator, CREB-regulated transcriptional coactivator 1 (CRTC1), was blocked in transfected neurons. Cell attach patch recordings combined with live multiphoton imaging demonstrated that the loss of CaNB1 did not prevent neurons from fully participating in electrographic seizure activity. Finally, dendrite reconstruction showed that the elimination of CaNB1 prevented seizure-induced decreases in both dendrite length and branch number. Results suggest that CaN plays a key role in seizure-induced dendrite growth suppression and may contribute to the neurobehavioral comorbidities of childhood epilepsy.


Assuntos
Calcineurina/metabolismo , Dendritos/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Fosfoproteínas/metabolismo , Convulsões/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular , Aprendizagem/fisiologia , Memória/fisiologia , Camundongos , Células Piramidais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA