Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Neurol ; 94(1): 106-122, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36935347

RESUMEN

OBJECTIVE: Temporal lobe epilepsy (TLE) is a progressive disorder mediated by pathological changes in molecular cascades and hippocampal neural circuit remodeling that results in spontaneous seizures and cognitive dysfunction. Targeting these cascades may provide disease-modifying treatments for TLE patients. Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) inhibitors have emerged as potential disease-modifying therapies; a more detailed understanding of JAK/STAT participation in epileptogenic responses is required, however, to increase the therapeutic efficacy and reduce adverse effects associated with global inhibition. METHODS: We developed a mouse line in which tamoxifen treatment conditionally abolishes STAT3 signaling from forebrain excitatory neurons (nSTAT3KO). Seizure frequency (continuous in vivo electroencephalography) and memory (contextual fear conditioning and motor learning) were analyzed in wild-type and nSTAT3KO mice after intrahippocampal kainate (IHKA) injection as a model of TLE. Hippocampal RNA was obtained 24 h after IHKA and subjected to deep sequencing. RESULTS: Selective STAT3 knock-out in excitatory neurons reduced seizure progression and hippocampal memory deficits without reducing the extent of cell death or mossy fiber sprouting induced by IHKA injection. Gene expression was rescued in major networks associated with response to brain injury, neuronal plasticity, and learning and memory. We also provide the first evidence that neuronal STAT3 may directly influence brain inflammation. INTERPRETATION: Inhibiting neuronal STAT3 signaling improved outcomes in an animal model of TLE, prevented progression of seizures and cognitive co-morbidities while rescuing pathogenic changes in gene expression of major networks associated with epileptogenesis. Specifically targeting neuronal STAT3 may be an effective disease-modifying strategy for TLE. ANN NEUROL 2023;94:106-122.


Asunto(s)
Epilepsia del Lóbulo Temporal , Epilepsia , Ratones , Animales , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Redes Reguladoras de Genes , Ratones Noqueados , Convulsiones , Hipocampo/patología , Neuronas/metabolismo , Cognición , Modelos Animales de Enfermedad
2.
Epilepsy Behav ; 124: 108320, 2021 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-34592633

RESUMEN

Epilepsy is a brain disorder characterized by the occurrence of recurrent spontaneous seizures. Behavioral disorders and altered cognition are frequent comorbidities affecting the quality of life of people with epilepsy. These impairments are undoubtedly multifactorial and the specific mechanisms underlying these comorbidities are largely unknown. Long-lasting alterations in synaptic strength due to changes in expression, phosphorylation, or function of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors (AMPARs) have been associated with alterations in neuronal synaptic plasticity. In particular, alterations in hippocampal long-term potentiation (LTP), a well-accepted model of learning and memory, have been associated with altered cognition in epilepsy. Here, we analyzed the effects of pilocarpine-induced status epilepticus (SE) on AMPARs to determine if alterations in AMPAR signaling might be one of the mechanisms contributing to altered cognition during epilepsy. We found alterations in the phosphorylation and plasma membrane expression of AMPARs. In addition, we detected altered expression of GRIP, a key scaffolding protein involved in the proper distribution of AMPARs at the neuronal cell surface. Interestingly, a functional analysis revealed that these molecular changes are linked to impaired LTP. Together, these observations suggest that seizure-induced alterations in the molecular machinery regulating AMPARs likely impact the neuron's ability to support synaptic plasticity that is required for learning and memory.

3.
BMC Genomics ; 20(1): 677, 2019 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-31455240

RESUMEN

BACKGROUND: Brain-derived neurotrophic factor (BDNF) is a major signaling molecule that the brain uses to control a vast network of intracellular cascades fundamental to properties of learning and memory, and cognition. While much is known about BDNF signaling in the healthy nervous system where it controls the mitogen activated protein kinase (MAPK) and cyclic-AMP pathways, less is known about its role in multiple brain disorders where it contributes to the dysregulated neuroplasticity seen in epilepsy and traumatic brain injury (TBI). We previously found that neurons respond to prolonged BDNF exposure (both in vivo (in models of epilepsy and TBI) and in vitro (in BDNF treated primary neuronal cultures)) by activating the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling pathway. This pathway is best known for its association with inflammatory cytokines in non-neuronal cells. RESULTS: Here, using deep RNA-sequencing of neurons exposed to BDNF in the presence and absence of well characterized JAK/STAT inhibitors, and without non-neuronal cells, we determine the BDNF transcriptome that is specifically regulated by agents that inhibit JAK/STAT signaling. Surprisingly, the BDNF-induced JAK/STAT transcriptome contains ion channels and neurotransmitter receptors coming from all the major classes expressed in the brain, along with key modulators of synaptic plasticity, neurogenesis, and axonal remodeling. Analysis of this dataset has revealed a unique non-canonical mechanism of JAK/STATs in neurons as differential gene expression mediated by STAT3 is not solely dependent upon phosphorylation at residue 705 and may involve a BDNF-induced interaction of STAT3 with Heterochromatin Protein 1 alpha (HP1α). CONCLUSIONS: These findings suggest that the neuronal BDNF-induced JAK/STAT pathway involves more than STAT3 phosphorylation at 705, providing the first evidence for a non-canonical mechanism that may involve HP1α. Our analysis reveals that JAK/STAT signaling regulates many of the genes associated with epilepsy syndromes where BDNF levels are markedly elevated. Uncovering the mechanism of this novel form of BDNF signaling in the brain may provide a new direction for epilepsy therapeutics and open a window into the complex mechanisms of STAT3 transcriptional regulation in neurological disease.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Encéfalo/metabolismo , Quinasas Janus/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Encéfalo/enzimología , Células Cultivadas , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/metabolismo , RNA-Seq , Ratas , Ratas Sprague-Dawley , Receptores de Neurotransmisores/biosíntesis , Receptores de Neurotransmisores/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal , Transcriptoma
4.
Epilepsia ; 59(1): 37-66, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29247482

RESUMEN

The most common forms of acquired epilepsies arise following acute brain insults such as traumatic brain injury, stroke, or central nervous system infections. Treatment is effective for only 60%-70% of patients and remains symptomatic despite decades of effort to develop epilepsy prevention therapies. Recent preclinical efforts are focused on likely primary drivers of epileptogenesis, namely inflammation, neuron loss, plasticity, and circuit reorganization. This review suggests a path to identify neuronal and molecular targets for clinical testing of specific hypotheses about epileptogenesis and its prevention or modification. Acquired human epilepsies with different etiologies share some features with animal models. We identify these commonalities and discuss their relevance to the development of successful epilepsy prevention or disease modification strategies. Risk factors for developing epilepsy that appear common to multiple acute injury etiologies include intracranial bleeding, disruption of the blood-brain barrier, more severe injury, and early seizures within 1 week of injury. In diverse human epilepsies and animal models, seizures appear to propagate within a limbic or thalamocortical/corticocortical network. Common histopathologic features of epilepsy of diverse and mostly focal origin are microglial activation and astrogliosis, heterotopic neurons in the white matter, loss of neurons, and the presence of inflammatory cellular infiltrates. Astrocytes exhibit smaller K+ conductances and lose gap junction coupling in many animal models as well as in sclerotic hippocampi from temporal lobe epilepsy patients. There is increasing evidence that epilepsy can be prevented or aborted in preclinical animal models of acquired epilepsy by interfering with processes that appear common to multiple acute injury etiologies, for example, in post-status epilepticus models of focal epilepsy by transient treatment with a trkB/PLCγ1 inhibitor, isoflurane, or HMGB1 antibodies and by topical administration of adenosine, in the cortical fluid percussion injury model by focal cooling, and in the albumin posttraumatic epilepsy model by losartan. Preclinical studies further highlight the roles of mTOR1 pathways, JAK-STAT3, IL-1R/TLR4 signaling, and other inflammatory pathways in the genesis or modulation of epilepsy after brain injury. The wealth of commonalities, diversity of molecular targets identified preclinically, and likely multidimensional nature of epileptogenesis argue for a combinatorial strategy in prevention therapy. Going forward, the identification of impending epilepsy biomarkers to allow better patient selection, together with better alignment with multisite preclinical trials in animal models, should guide the clinical testing of new hypotheses for epileptogenesis and its prevention.


Asunto(s)
Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Epilepsia/etiología , Investigación Biomédica Traslacional , Animales , Lesiones Encefálicas/clasificación , Humanos
5.
Epilepsia ; 58(3): 331-342, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28035782

RESUMEN

Neurologic and psychiatric comorbidities are common in patients with epilepsy. Diagnostic, predictive, and pharmacodynamic biomarkers of such comorbidities do not exist. They may share pathogenetic mechanisms with epileptogenesis/ictogenesis, and as such are an unmet clinical need. The objectives of the subgroup on biomarkers of comorbidities at the XIII Workshop on the Neurobiology of Epilepsy (WONOEP) were to present the state-of-the-art recent research findings in the field that highlighting potential biomarkers for comorbidities in epilepsy. We review recent progress in the field, including molecular, imaging, and genetic biomarkers of comorbidities as discussed during the WONOEP meeting on August 31-September 4, 2015, in Heybeliada Island (Istanbul, Turkey). We further highlight new directions and concepts from studies on comorbidities and potential new biomarkers for the prediction, diagnosis, and treatment of epilepsy-associated comorbidities. The activation of various molecular signaling pathways such as the "Janus Kinase/Signal Transducer and Activator of Transcription," "mammalian Target of Rapamycin," and oxidative stress have been shown to correlate with the presence and severity of subsequent cognitive abnormalities. Furthermore, dysfunction in serotonergic transmission, hyperactivity of the hypothalamic-pituitary-adrenocortical axis, the role of the inflammatory cytokines, and the contributions of genetic factors have all recently been regarded as relevant for understanding epilepsy-associated depression and cognitive deficits. Recent evidence supports the utility of imaging studies as potential biomarkers. The role of such biomarker may be far beyond the diagnosis of comorbidities, as accumulating clinical data indicate that comorbidities can predict epilepsy outcomes. Future research is required to reveal whether molecular changes in specific signaling pathways or advanced imaging techniques could be detected in the clinical settings and correlate with epilepsy-associated comorbidities. A reliable biomarker will allow a more accurate diagnosis and improved treatment of epilepsy-associated comorbidities.


Asunto(s)
Biomarcadores , Epilepsia/epidemiología , Trastornos Mentales/epidemiología , Enfermedades del Sistema Nervioso/epidemiología , Animales , Comorbilidad , Humanos , Neurobiología
6.
Epilepsia ; 58 Suppl 4: 68-77, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29105071

RESUMEN

Current antiseizure therapy is ineffective in approximately one third of people with epilepsy and is often associated with substantial side effects. In addition, most current therapeutic paradigms offer treatment, but not cure, and no therapies are able to modify the underlying disease, that is, can prevent or halt the process of epileptogenesis or alleviate the cognitive and psychiatric comorbidities. Preclinical research in the field of epilepsy has been extensive, but unfortunately, not all the animal models being used have been validated for their predictive value. The overall goal of TASK2 of the AES/ILAE Translational Task Force is to organize and coordinate systematic reviews on selected topics regarding animal research in epilepsy. Herein we describe our strategy. In the first part of the paper we provide an overview of the usefulness of systematic reviews and meta-analysis for preclinical research and explain the essentials for their conduct. Then we describe in detail the protocol for a first systematic review, which will focus on the identification and characterization of outcome measures reported in animal models of epilepsy. The specific goals of this study are to define systematically the phenotypic characteristics of the most commonly used animal models, and to effectively compare these with the manifestations of human epilepsy. This will provide epilepsy researchers with detailed information on the strengths and weaknesses of epilepsy models, facilitating their refinement and future research. Ultimately, this could lead to a refined use of relevant models for understanding the mechanism(s) of the epilepsies and developing novel therapies.


Asunto(s)
Comités Consultivos , Epilepsia/diagnóstico , Epilepsia/terapia , Evaluación de Resultado en la Atención de Salud , Investigación Biomédica Traslacional , Animales , Modelos Animales de Enfermedad , Humanos , Revisiones Sistemáticas como Asunto
7.
Neurobiol Dis ; 77: 246-56, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25769812

RESUMEN

In this study, we analyzed the impact that spontaneous seizures might have on the plasma membrane expression, composition and function of GABAA receptors (GABAARs). For this, the tissue of chronically epileptic rats was collected within 3h of seizure occurrence (≤3h group) or at least 24h after seizure occurrence (≥24h group). A retrospective analysis of seizure frequency revealed that selecting animals on the bases of seizure proximity also grouped animals in terms of overall seizure burden with a higher seizure burden observed in the ≤3h group. A biochemical analysis showed that although animals with more frequent/recent seizures (≤3h group) had similar levels of GABAAR at the plasma membrane they showed deficits in inhibitory neurotransmission. By contrast, the tissue obtained from animals experiencing infrequent seizures (≥24h group) had increased plasma membrane levels of GABAAR and showed no deficit in inhibitory function. Together, our findings offer an initial insight into the molecular changes that might help to explain how alterations in GABAAR function can be associated with differential seizure burden. Our findings also suggest that increased plasma membrane levels of GABAAR might act as a compensatory mechanism to more effectively maintain inhibitory function, repress hyperexcitability and reduce seizure burden. This study is an initial step towards a fuller characterization of the molecular events that trigger alterations in GABAergic neurotransmission during chronic epilepsy.


Asunto(s)
Receptores de GABA-A/metabolismo , Estado Epiléptico/metabolismo , Animales , Biotinilación , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Agonistas del GABA/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/patología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Isoxazoles/farmacología , Masculino , Agonistas Muscarínicos/toxicidad , Neuronas/efectos de los fármacos , Pilocarpina/toxicidad , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/patología , Valina/análogos & derivados , Valina/farmacología
8.
Epilepsia ; 55(11): 1826-33, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25223733

RESUMEN

OBJECTIVE: Temporal lobe epilepsy (TLE) is frequently medically intractable and often progressive. Compromised inhibitory neurotransmission due to altered γ-aminobutyric acid (GABA)A receptor α4 subunit (GABAA Rα4) expression has been emphasized as a potential contributor to the initial development of epilepsy following a brain insult (primary epileptogenesis), but the regulation of GABAA Rα4 during chronic epilepsy, specifically, how expression is altered following spontaneous seizures, is less well understood. METHODS: Continuous video-electroencephalography (EEG) recordings from rats with pilocarpine-induced TLE were used to capture epileptic animals within 3 h of a spontaneous seizure (SS), or >24 h after the last SS, to determine whether recent occurrence of a seizure was associated with altered levels of GABAA Rα4 expression. We further evaluated whether this GABAA Rα4 plasticity is regulated by signaling mechanisms active in primary epileptogenesis, specifically, increases in brain-derived neurotrophic factor (BDNF) and early growth response factor 3 (Egr3). RESULTS: Elevated levels of GABAA Rα4 messenger RNA (mRNA) and protein were observed following spontaneous seizures, and were associated with higher levels of BDNF and Egr3 mRNA. SIGNIFICANCE: These data suggest that spontaneous, recurrent seizures that define chronic epilepsy may influence changes in GABAA Rα4 expression, and that signaling pathways known to regulate GABAA Rα4 expression after status epilepticus may also be activated after spontaneous seizures in chronically epileptic animals.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Receptores de GABA-A/metabolismo , Convulsiones/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/inducido químicamente , Pilocarpina/farmacología , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
9.
Adv Exp Med Biol ; 813: 133-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25012373

RESUMEN

Numerous changes in GABAergic neurons, receptors, and inhibitory mechanisms have been described in temporal lobe epilepsy (TLE), either in humans or in animal models. Nevertheless, there remains a common assumption that epilepsy can be explained by simply an insufficiency of GABAergic inhibition. Alternatively, investigators have suggested that there is hyperinhibition that masks an underlying hyperexcitability. Here we examine the status epilepticus (SE) models of TLE and focus on the dentate gyrus of the hippocampus, where a great deal of data have been collected. The types of GABAergic neurons and GABAA receptors are summarized under normal conditions and after SE. The role of GABA in development and in adult neurogenesis is discussed. We suggest that instead of "too little or too much" GABA there is a complexity of changes after SE that makes the emergence of chronic seizures (epileptogenesis) difficult to understand mechanistically, and difficult to treat. We also suggest that this complexity arises, at least in part, because of the remarkable plasticity of GABAergic neurons and GABAA receptors in response to insult or injury.


Asunto(s)
Epilepsia/fisiopatología , Plasticidad Neuronal , Ácido gamma-Aminobutírico/fisiología , Adulto , Humanos
10.
Neoreviews ; 25(6): e338-e349, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38821905

RESUMEN

Neonatal seizures are common among patients with acute brain injury or critical illness and can be difficult to diagnose and treat. The most common etiology of neonatal seizures is hypoxic-ischemic encephalopathy, with other common causes including ischemic stroke and intracranial hemorrhage. Neonatal clinicians can use a standardized approach to patients with suspected or confirmed neonatal seizures that entails laboratory testing, neuromonitoring, and brain imaging. The primary goals of management of neonatal seizures are to identify the underlying cause, correct it if possible, and prevent further brain injury. This article reviews recent evidence-based guidelines for the treatment of neonatal seizures and discusses the long-term outcomes of patients with neonatal seizures.


Asunto(s)
Convulsiones , Humanos , Recién Nacido , Convulsiones/diagnóstico , Convulsiones/etiología , Convulsiones/terapia , Hipoxia-Isquemia Encefálica/diagnóstico , Hipoxia-Isquemia Encefálica/terapia
11.
Epilepsia ; 54(4): 616-24, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23294024

RESUMEN

PURPOSE: Epileptogenesis is the process by which a brain becomes hyperexcitable and capable of generating recurrent spontaneous seizures. In humans, it has been hypothesized that following a brain insult there are a number of molecular and cellular changes that underlie the development of spontaneous seizures. Studies in animal models have shown that an injured brain may develop epileptiform activity before appearance of epileptic seizures and that the pathophysiology accompanying spontaneous seizures is associated with a dysfunction of γ-aminobutyric acid (GABA)ergic neurotransmission. Here, we analyzed the effects of status epilepticus on the expression of GABAA receptors (GABAA Rs) and scaffolding proteins involved in the regulation of GABAA R trafficking and anchoring. METHODS: Western blot analysis was used to determine the levels of proteins involved in GABAA R trafficking and anchoring in adult rats subjected to pilocarpine-induced status epilepticus (SE) and controls. Cell surface biotinylation using a cell membrane-impermeable reagent was used to assay for changes in the expression of receptors at the plasma membrane. Finally, immunoprecipitation experiments were used to evaluate the composition of GABAA Rs. We examined for a correlation between total GABAA R subunit expression, plasma membrane expression, and receptor composition. KEY FINDINGS: Analysis of tissue samples from the CA1 region of hippocampus show that SE promotes a loss of GABAA R subunits and of the scaffolding proteins associated with them. We also found a decrease in the levels of receptors located at the plasma membrane and alterations in GABAA R composition. SIGNIFICANCE: The changes in protein expression of GABAA Rs and scaffolding proteins detected in these studies provide a potential mechanism to explain the deficits in GABAergic neurotransmission observed during the epileptogenic period. Our current observations represent an additional step toward the elucidation of the molecular mechanisms underlying GABAA R dysfunction during epileptogenesis.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Proteínas Portadoras/biosíntesis , Epilepsia/metabolismo , Proteínas de la Membrana/biosíntesis , Receptores de GABA-A/biosíntesis , Animales , Biotinilación , Western Blotting , Proteínas Portadoras/genética , Membrana Celular/metabolismo , Regulación hacia Abajo , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Inmunoprecipitación , Masculino , Proteínas de la Membrana/genética , Microscopía Confocal , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/biosíntesis , Receptores de GABA-A/genética , Estado Epiléptico/inducido químicamente , Estado Epiléptico/metabolismo , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/fisiología
12.
Epilepsia ; 54 Suppl 4: 35-43, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23909852

RESUMEN

Several preclinical proof-of-concept studies have provided evidence for positive treatment effects on epileptogenesis. However, none of these hypothetical treatments has advanced to the clinic. The experience in other fields of neurology such as stroke, Alzheimer's disease, or amyotrophic lateral sclerosis has indicated several problems in the design of preclinical studies, which likely contribute to failures in translating the positive preclinical data to the clinic. The Working Group on "Issues related to development of antiepileptogenic therapies" of the International League Against Epilepsy (ILAE) and the American Epilepsy Society (AES) has considered the possible problems that arise when moving from proof-of-concept antiepileptogenesis (AEG) studies to preclinical AEG trials, and eventually to clinical AEG trials. This article summarizes the discussions and provides recommendations on how to design a preclinical AEG monotherapy trial in adult animals. We specifically address study design, animal and model selection, number of studies needed, issues related to administration of the treatment, outcome measures, statistics, and reporting. In addition, we give recommendations for future actions to advance the preclinical AEG testing.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Drogas en Investigación/uso terapéutico , Adulto , Animales , Anticonvulsivantes/efectos adversos , Niño , Enfermedad Crónica , Ensayos Clínicos Controlados como Asunto , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Aprobación de Drogas , Resistencia a Medicamentos , Drogas en Investigación/efectos adversos , Medicina Basada en la Evidencia , Humanos , National Institute of Neurological Disorders and Stroke (U.S.) , Estados Unidos
13.
Epilepsia ; 54 Suppl 4: 44-60, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23909853

RESUMEN

Many symptoms of neurologic or psychiatric illness--such as cognitive impairment, depression, anxiety, attention deficits, and migraine--occur more frequently in people with epilepsy than in the general population. These diverse comorbidities present an underappreciated problem for people with epilepsy and their caregivers because they decrease quality of life, complicate treatment, and increase mortality. In fact, it has been suggested that comorbidities can have a greater effect on quality of life in people with epilepsy than the seizures themselves. There is increasing recognition of the frequency and impact of cognitive and behavioral comorbidities of epilepsy, highlighted in the 2012 Institute of Medicine report on epilepsy. Comorbidities have also been acknowledged, as a National Institutes of Health (NIH) Benchmark area for research in epilepsy. However, relatively little progress has been made in developing new therapies directed specifically at comorbidities. On the other hand, there have been many advances in understanding underlying mechanisms. These advances have made it possible to identify novel targets for therapy and prevention. As part of the International League Against Epilepsy/American Epilepsy Society workshop on preclinical therapy development for epilepsy, our working group considered the current state of understanding related to terminology, models, and strategies for therapy development for the comorbidities of epilepsy. Herein we summarize our findings and suggest ways to accelerate development of new therapies. We also consider important issues to improve research including those related to methodology, nonpharmacologic therapies, biomarkers, and infrastructure.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Descubrimiento de Drogas , Drogas en Investigación/uso terapéutico , Epilepsia/tratamiento farmacológico , Trastornos Neurocognitivos/tratamiento farmacológico , Animales , Trastornos de Ansiedad/inducido químicamente , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/psicología , Trastorno por Déficit de Atención con Hiperactividad/inducido químicamente , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Trastorno por Déficit de Atención con Hiperactividad/psicología , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/psicología , Comorbilidad , Trastorno Depresivo/inducido químicamente , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/psicología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Drogas en Investigación/efectos adversos , Epilepsia/diagnóstico , Epilepsia/psicología , Humanos , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/psicología , Trastornos Neurocognitivos/inducido químicamente , Trastornos Neurocognitivos/diagnóstico , Trastornos Neurocognitivos/psicología , Calidad de Vida/psicología , Investigación Biomédica Traslacional
14.
Epilepsia ; 54(10): 1780-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24032982

RESUMEN

PURPOSE: Traumatic brain injury (TBI) is an important cause of morbidity and mortality in children, and early posttraumatic seizures (EPTS) are a contributing factor to ongoing acute damage. Continuous video-EEG monitoring (cEEG) was utilized to assess the burden of clinical and electrographic EPTS. METHODS: Eighty-seven consecutive, unselected (mild - severe), acute TBI patients requiring pediatric intensive care unit (PICU) admission at two academic centers were monitored prospectively with cEEG per established clinical TBI protocols. Clinical and subclinical seizures and status epilepticus (SE, clinical and subclinical) were assessed for their relation to clinical risk factors and short-term outcome measures. KEY FINDINGS: Of all patients, 42.5% (37/87) had seizures. Younger age (p = 0.002) and injury mechanism (abusive head trauma - AHT, p < 0.001) were significant risk factors. Subclinical seizures occurred in 16.1% (14/87), while 6.9% (6/87) had only subclinical seizures. Risk factors for subclinical seizures included younger age (p < 0.001), AHT (p < 0.001), and intraaxial bleed (p < 0.001). SE occurred in 18.4% (16/87) with risk factors including younger age (p < 0.001), AHT (p < 0.001), and intraaxial bleed (p = 0.002). Subclinical SE was detected in 13.8% (12/87) with significant risk factors including younger age (p < 0.001), AHT (p = 0.001), and intraaxial bleed (p = 0.004). Subclinical seizures were associated with lower discharge King's Outcome Scale for Childhood Head Injury (KOSCHI) score (p = 0.002). SE and subclinical SE were associated with increased hospital length of stay (p = 0.017 and p = 0.041, respectively) and lower hospital discharge KOSCHI (p = 0.007 and p = 0.040, respectively). SIGNIFICANCE: cEEG monitoring significantly improves detection of seizures/SE and is the only way to detect subclinical seizures/SE. cEEG may be indicated after pediatric TBI, particularly in younger children, AHT cases, and those with intraaxial blood on computerized tomography (CT).


Asunto(s)
Lesiones Encefálicas/complicaciones , Electroencefalografía/métodos , Epilepsias Parciales/diagnóstico , Adolescente , Anticonvulsivantes/uso terapéutico , Lesiones Encefálicas/fisiopatología , Niño , Preescolar , Epilepsias Parciales/tratamiento farmacológico , Epilepsias Parciales/etiología , Epilepsias Parciales/fisiopatología , Femenino , Escala de Coma de Glasgow , Humanos , Lactante , Masculino , Monitoreo Fisiológico/métodos , Estudios Prospectivos , Factores de Riesgo , Convulsiones/diagnóstico , Convulsiones/etiología , Convulsiones/fisiopatología , Estado Epiléptico/diagnóstico , Estado Epiléptico/etiología , Estado Epiléptico/fisiopatología
15.
J Neurochem ; 120(2): 210-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22035109

RESUMEN

Regulation of gene expression via brain-derived neurotrophic factor (BDNF) is critical to the development of the nervous system and may well underlie cognitive performance throughout life. We now describe a mechanism by which BDNF can exert its effects on postsynaptic receptor populations that may have relevance to both the normal and diseased brain where BDNF levels either rise or fall in association with changes in excitatory neurotransmission. Increased levels of NMDA receptors (NMDARs) occur in rat cortical neurons via synthesis of new NMDA receptor 1 (NR1) subunits. The majority of synthesis is controlled by binding of cAMP response element binding protein (CREB) and early growth response factor 3 (Egr3) to the core NR1 promoter (NR1-p) region. BDNF-mediated NR1 transcription depends upon induction of the mitogen-activated protein kinase (MAPK) pathway through activation of the TrK-B receptor. Taken together with the fact that NMDAR activation stimulates BDNF synthesis, our results uncover a feed-forward gene regulatory network that may enhance excitatory neurotransmission to change neuronal behavior over time.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Proteína de Unión a CREB/metabolismo , Corteza Cerebral/citología , Canales de Potasio Éter-A-Go-Go/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Canales de Potasio Éter-A-Go-Go/genética , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas Luminiscentes/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Receptor trkB/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Transfección , Dedos de Zinc/genética
16.
Eur J Neurosci ; 35(12): 1857-65, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22708596

RESUMEN

Neonatal seizures have unique properties that have proved challenging for both clinicians and basic science researchers. Clinical therapies aimed at neonatal seizures have proven only partially effective and new therapies are slow to develop. This article will discuss neonatal seizures within the framework of the barriers that exist to the development of new therapies, and the challenges inherent in bringing new therapies from the bench to the bedside. With the European Union and USA creating national collaborative project infrastructure, improved collaborative resources should advance clinical research on urgently needed new therapies for this disorder.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/provisión & distribución , Anticonvulsivantes/uso terapéutico , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Descubrimiento de Drogas/economía , Descubrimiento de Drogas/tendencias , Humanos , Recién Nacido , Ratones , Terapia Molecular Dirigida , Ratas , Convulsiones/etiología
17.
Epilepsia ; 53 Suppl 9: 71-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23216580

RESUMEN

Epilepsy is a disease of complex etiology, and multiple molecular mechanisms contribute to its development. Temporal lobe epilepsy (TLE) may result from an initial precipitating event such as hypoxia, head injury, or prolonged seizure (i.e., status epilepticus [SE]), that is followed by a latent period of months to years before spontaneous seizures occur. γ-Aminobutyric acid (GABA)(A) receptor (GABA(A) R) subunit changes occur during this latent period and may persist following the onset of spontaneous seizures. Research into the molecular mechanisms regulating these changes and potential targets for intervention to reverse GABA(A) R subunit alterations have uncovered seizure-induced pathways that contribute to epileptogenesis. Several growth or transcription factors are known to be activated by SE, including (but not limited to): brain-derived neurotrophic factor (BDNF), cAMP response element binding protein (CREB), inducible cAMP early repressor (ICER), and early growth response factors (Egrs). Results of multiple studies suggest that these factors transcriptionally regulate GABA(A) R subunit gene expression in a way that is pertinent to the development of epilepsy. This article focuses on these signaling elements and describes their possible roles in gene regulatory pathways that may be critical in the development of chronic epilepsy.


Asunto(s)
Epilepsia/metabolismo , Redes y Vías Metabólicas , Receptores de GABA-A/biosíntesis , Transducción de Señal , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Epilepsia/fisiopatología , Epilepsia del Lóbulo Temporal/metabolismo , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Factores de Crecimiento Nervioso/metabolismo , Neurotransmisores/metabolismo , Receptores de GABA-A/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Epilepsia ; 52 Suppl 1: 13-20, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21214535

RESUMEN

Intellectual and developmental disabilities (IDDs) such as autistic spectrum disorders (ASDs) and epilepsies are heterogeneous disorders that have diverse etiologies and pathophysiologies. The high rate of co-occurrence of these disorders, however, suggests potentially shared underlying mechanisms. A number of well-known genetic disorders share epilepsy, intellectual disability, and autism as prominent phenotypic features, including tuberous sclerosis complex, Rett syndrome, and fragile X syndrome. In addition, mutations of several genes involved in neurodevelopment, including ARX, DCX, neuroligins, and neuropilin 2 have been identified in children with epilepsy, IDDs, ASDs, or a combination of thereof. Finally, in animal models, early life seizures can result in cellular and molecular changes that could contribute to learning and behavioral disabilities. Increased understanding of the common genetic, molecular, and cellular mechanisms of IDDs, ASDs, and epilepsy may provide insight into their underlying pathophysiology and elucidate new therapeutic approaches for these conditions.


Asunto(s)
Trastornos de la Conducta Infantil/psicología , Trastornos del Conocimiento/psicología , Epilepsia/psicología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Niño , Trastornos de la Conducta Infantil/etiología , Trastornos de la Conducta Infantil/genética , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Epilepsia/complicaciones , Epilepsia/genética , Síndrome del Cromosoma X Frágil/complicaciones , Síndrome del Cromosoma X Frágil/psicología , Humanos , Interneuronas/patología , Síndrome de Rett/complicaciones , Síndrome de Rett/psicología , Esclerosis Tuberosa/complicaciones , Esclerosis Tuberosa/psicología
19.
Front Neurol ; 12: 654606, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34122302

RESUMEN

Epilepsy is characterized by recurrent, spontaneous seizures and is a major contributor to the global burden of neurological disease. Although epilepsy can result from a variety of brain insults, in many cases the cause is unknown and, in a significant proportion of cases, seizures cannot be controlled by available treatments. Understanding the molecular alterations that underlie or are triggered by epileptogenesis would help to identify therapeutics to prevent or control progression to epilepsy. To this end, the moderate throughput technique of Reverse Phase Protein Arrays (RPPA) was used to profile changes in protein expression in a pilocarpine mouse model of acquired epilepsy. Levels of 54 proteins, comprising phosphorylation-dependent and phosphorylation-independent components of major signaling pathways and cellular complexes, were measured in hippocampus, cortex and cerebellum of mice at six time points, spanning 15 min to 2 weeks after induction of status epilepticus. Results illustrate the time dependence of levels of the commonly studied MTOR pathway component, pS6, and show, for the first time, detailed responses during epileptogenesis of multiple components of the MTOR, MAPK, JAK/STAT and apoptosis pathways, NMDA receptors, and additional cellular complexes. Also noted are time- and brain region- specific changes in correlations among levels of functionally related proteins affecting both neurons and glia. While hippocampus and cortex are primary areas studied in pilocarpine-induced epilepsy, cerebellum also shows significant time-dependent molecular responses.

20.
Nat Rev Neurol ; 17(4): 231-242, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33594276

RESUMEN

Onset of many forms of epilepsy occurs after an initial epileptogenic insult or as a result of an identified genetic defect. Given that the precipitating insult is known, these epilepsies are, in principle, amenable to secondary prevention. However, development of preventive treatments is difficult because only a subset of individuals will develop epilepsy and we cannot currently predict which individuals are at the highest risk. Biomarkers that enable identification of these individuals would facilitate clinical trials of potential anti-epileptogenic treatments, but no such prognostic biomarkers currently exist. Several putative molecular, imaging, electroencephalographic and behavioural biomarkers of epileptogenesis have been identified, but clinical translation has been hampered by fragmented and poorly coordinated efforts, issues with inter-model reproducibility, study design and statistical approaches, and difficulties with validation in patients. These challenges demand a strategic roadmap to facilitate the identification, characterization and clinical validation of biomarkers for epileptogenesis. In this Review, we summarize the state of the art with respect to biomarker research in epileptogenesis and propose a five-phase roadmap, adapted from those developed for cancer and Alzheimer disease, that provides a conceptual structure for biomarker research.


Asunto(s)
Biomarcadores , Electroencefalografía , Epilepsia/diagnóstico , MicroARNs , Neuroimagen , Animales , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Epilepsia/sangre , Epilepsia/líquido cefalorraquídeo , Epilepsia/fisiopatología , Humanos , MicroARNs/sangre , MicroARNs/líquido cefalorraquídeo , Guías de Práctica Clínica como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA