Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Neurosci ; 39(46): 9251-9260, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31537705

RESUMEN

Dravet syndrome (DS) is a severe early-onset epilepsy associated with heterozygous loss-of-function mutations in SCN1A Animal models of DS with global Scn1a haploinsufficiency recapitulate the DS phenotype, including seizures, premature death, and impaired spatial memory performance. Spatial memory requires hippocampal sharp-wave ripples (SPW-Rs), which consist of high-frequency field potential oscillations (ripples, 100-260 Hz) superimposed on a slower SPW. Published in vitro electrophysiologic recordings in DS mice demonstrate reduced firing of GABAergic inhibitory neurons, which are essential for the formation of SPW-R complexes. Here, in vivo electrophysiologic recordings of hippocampal local field potential in both male and female mice demonstrate that Scn1a haploinsufficiency slows intrinsic ripple frequency and reduces the rate of SPW-R occurrence. In DS mice, peak ripple-band power is shifted to lower frequencies, average intertrough intervals of individually detected ripples are slower, and the rate of SPW-R generation is reduced, while SPW amplitude remains unaffected. These alterations in SPW-R properties, in combination with published reductions in interneuron function in DS, suggest a direct link between reduced inhibitory neuron excitability and impaired SPW-R function. A simple interconnected, conductance-based in silico interneuron network model was used to determine whether reduced sodium conductance is sufficient to slow ripple frequency, and stimulation with a modeled SPW demonstrates that reduced sodium conductance alone is sufficient to slow oscillatory frequencies. These findings forge a potential mechanistic link between impaired SPW-R generation and Scn1a mutation in DS mice, expanding the set of disorders in which SPW-R dysfunction contributes to impaired memory.SIGNIFICANCE STATEMENT Disruption of sharp-wave ripples, a characteristic hippocampal rhythm coordinated by the precise timing of GABAergic interneurons, impairs spatial learning and memory. Prior in vitro patch-clamp recordings in brain slices from genetic mouse models of Dravet syndrome (DS) reveal reduced sodium current and excitability in GABAergic interneurons but not excitatory cells, suggesting a causal role for impaired interneuron activity in seizures and cognitive impairment. Here, heterozygous Scn1a mutation in DS mice reduces hippocampal sharp-wave ripple occurrence and slows internal ripple frequency in vivo and a simple in silico model demonstrates reduction in interneuron function alone is sufficient to slow model oscillations. Together, these findings provide a plausible pathophysiologic mechanism for Scn1a gene mutation to impair spatial memory.


Asunto(s)
Ondas Encefálicas , Epilepsias Mioclónicas/fisiopatología , Hipocampo/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.1/fisiología , Neuronas/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Neuronas GABAérgicas/fisiología , Haploinsuficiencia , Interneuronas/fisiología , Masculino , Ratones Endogámicos C57BL , Modelos Neurológicos , Canal de Sodio Activado por Voltaje NAV1.1/genética
2.
Nature ; 489(7416): 385-90, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22914087

RESUMEN

Haploinsufficiency of the SCN1A gene encoding voltage-gated sodium channel Na(V)1.1 causes Dravet's syndrome, a childhood neuropsychiatric disorder including recurrent intractable seizures, cognitive deficit and autism-spectrum behaviours. The neural mechanisms responsible for cognitive deficit and autism-spectrum behaviours in Dravet's syndrome are poorly understood. Here we report that mice with Scn1a haploinsufficiency exhibit hyperactivity, stereotyped behaviours, social interaction deficits and impaired context-dependent spatial memory. Olfactory sensitivity is retained, but novel food odours and social odours are aversive to Scn1a(+/-) mice. GABAergic neurotransmission is specifically impaired by this mutation, and selective deletion of Na(V)1.1 channels in forebrain interneurons is sufficient to cause these behavioural and cognitive impairments. Remarkably, treatment with low-dose clonazepam, a positive allosteric modulator of GABA(A) receptors, completely rescued the abnormal social behaviours and deficits in fear memory in the mouse model of Dravet's syndrome, demonstrating that they are caused by impaired GABAergic neurotransmission and not by neuronal damage from recurrent seizures. These results demonstrate a critical role for Na(V)1.1 channels in neuropsychiatric functions and provide a potential therapeutic strategy for cognitive deficit and autism-spectrum behaviours in Dravet's syndrome.


Asunto(s)
Trastorno Autístico/tratamiento farmacológico , Trastorno Autístico/fisiopatología , Moduladores del GABA/uso terapéutico , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Canales de Sodio/genética , Canales de Sodio/metabolismo , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Ansiedad/fisiopatología , Trastorno Autístico/complicaciones , Trastorno Autístico/genética , Clonazepam/farmacología , Clonazepam/uso terapéutico , Epilepsias Mioclónicas/complicaciones , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/fisiopatología , Moduladores del GABA/farmacología , Neuronas GABAérgicas/metabolismo , Haploinsuficiencia/genética , Heterocigoto , Hipocampo/citología , Proteínas de Homeodominio/genética , Hipercinesia/fisiopatología , Interneuronas/metabolismo , Masculino , Memoria , Ratones , Canal de Sodio Activado por Voltaje NAV1.1 , Conducta Social , Percepción Espacial , Trastorno de Movimiento Estereotipado/fisiopatología , Síndrome , Factores de Transcripción/genética
3.
Proc Natl Acad Sci U S A ; 109(36): 14646-51, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22908258

RESUMEN

Heterozygous loss-of-function mutations in the brain sodium channel Na(V)1.1 cause Dravet syndrome (DS), a pharmacoresistant infantile-onset epilepsy syndrome with comorbidities of cognitive impairment and premature death. Previous studies using a mouse model of DS revealed reduced sodium currents and impaired excitability in GABAergic interneurons in the hippocampus, leading to the hypothesis that impaired excitability of GABAergic inhibitory neurons is the cause of epilepsy and premature death in DS. However, other classes of GABAergic interneurons are less impaired, so the direct cause of hyperexcitability, epilepsy, and premature death has remained unresolved. We generated a floxed Scn1a mouse line and used the Cre-Lox method driven by an enhancer from the Dlx1,2 locus for conditional deletion of Scn1a in forebrain GABAergic neurons. Immunocytochemical studies demonstrated selective loss of Na(V)1.1 channels in GABAergic interneurons in cerebral cortex and hippocampus. Mice with this deletion died prematurely following generalized tonic-clonic seizures, and they were equally susceptible to thermal induction of seizures as mice with global deletion of Scn1a. Evidently, loss of Na(V)1.1 channels in forebrain GABAergic neurons is both necessary and sufficient to cause epilepsy and premature death in DS.


Asunto(s)
Epilepsias Mioclónicas/genética , Interneuronas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/deficiencia , Animales , Electrocardiografía , Electroencefalografía , Epilepsias Mioclónicas/patología , Hipocampo/metabolismo , Inmunohistoquímica , Ratones , Ratones Transgénicos , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Plásmidos/genética , Prosencéfalo/metabolismo
4.
J Pharmacol Exp Ther ; 345(2): 215-24, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23424217

RESUMEN

Seizures remain uncontrolled in 30% of patients with epilepsy, even with concurrent use of multiple drugs, and uncontrolled seizures result in increased morbidity and mortality. An extreme example is Dravet syndrome (DS), an infantile-onset severe epilepsy caused by heterozygous loss of function mutations in SCN1A, the gene encoding the brain type-I voltage-gated sodium channel NaV1.1. Studies in Scn1a heterozygous knockout mice demonstrate reduced excitability of GABAergic interneurons, suggesting that enhancement of GABA signaling may improve seizure control and comorbidities. We studied the efficacy of two GABA-enhancing drugs, clonazepam and tiagabine, alone and in combination, against thermally evoked myoclonic and generalized tonic-clonic seizures. Clonazepam, a positive allosteric modulator of GABA-A receptors, protected against myoclonic and generalized tonic-clonic seizures. Tiagabine, a presynaptic GABA reuptake inhibitor, was protective against generalized tonic-clonic seizures but only minimally protective against myoclonic seizures and enhanced myoclonic seizure susceptibility at high doses. Combined therapy with clonazepam and tiagabine was synergistic against generalized tonic-clonic seizures but was additive against myoclonic seizures. Toxicity determined by rotorod testing was additive for combination therapy. The synergistic actions of clonazepam and tiagabine gave enhanced seizure protection and reduced toxicity, suggesting that combination therapy may be well tolerated and effective for seizures in DS.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsias Mioclónicas/tratamiento farmacológico , Agonistas del GABA/uso terapéutico , Convulsiones/tratamiento farmacológico , Ácido gamma-Aminobutírico/fisiología , Animales , Clonazepam/uso terapéutico , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Epilepsias Mioclónicas/complicaciones , Epilepsia Tónico-Clónica/tratamiento farmacológico , Femenino , Moduladores del GABA/uso terapéutico , Calor , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.1/genética , Ácidos Nipecóticos/uso terapéutico , Equilibrio Postural/efectos de los fármacos , Convulsiones/etiología , Transmisión Sináptica/efectos de los fármacos , Tiagabina
5.
Front Cell Neurosci ; 15: 751762, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733140

RESUMEN

Dravet Syndrome (DS) is a genetic, infantile-onset epilepsy with refractory seizures and severe cognitive impairment. While network level pathophysiology is poorly understood, work in genetic mouse models of DS reveals selective reduction of inhibitory interneuron excitability, a likely mechanism of seizures and comorbidities. Consistent with the critical role of interneurons in timing and recruitment of network activity, hippocampal sharp wave ripples (SPW-R)-interneuron dependent compound brain rhythms essential for spatial learning and memory-are less frequent and ripple frequency is slower in DS mice, both likely to impair cognitive performance. Febrile seizures are characteristic of DS, reflecting a temperature-dependent shift in excitation-inhibition balance. DS interneurons are sensitive to depolarization block and may fall silent with increased excitation precipitating epileptic transformation of ripples. To determine the temperature dependence of SWP-R features and relationship of SPW-R to hippocampal interictal activity, we recorded hippocampal local field potentials in a DS mouse model and wildtype littermate controls while increasing core body temperature. In both genotypes, temperature elevation speeds ripple frequency, although DS ripples remain consistently slower. The rate of SPW-R also increases in both genotypes but subsequently falls in DS mice as interictal epileptic activity simultaneously increases preceding a thermally-evoked seizure. Epileptic events occur intermixed with SPW-R, some during SPW-R burst complexes, and transiently suppress SPW-R occurrence suggesting shared network elements. Together these data demonstrate a temperature dependence of SPW-R rate and ripple frequency and suggest a pathophysiologic mechanism by which elevated temperature transforms a normal brain rhythm into epileptic event.

6.
Curr Biol ; 16(3): 321-7, 2006 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-16461287

RESUMEN

In the final stages of ovarian follicular development, the mouse oocyte remains arrested in the first meiotic prophase, and cAMP-stimulated PKA plays an essential role in this arrest. After the LH surge, a decrease in cAMP and PKA activity in the oocyte initiates an irreversible maturation process that culminates in a second arrest at metaphase II prior to fertilization. A-kinase anchoring proteins (AKAPs) mediate the intracellular localization of PKA and control the specificity and kinetics of substrate phosphorylation. Several AKAPs have been identified in oocytes including one at 140 kDa that we now identify as a product of the Akap1 gene. We show that PKA interaction with AKAPs is essential for two sequential steps in the maturation process: the initial maintenance of meiotic arrest and the subsequent irreversible progression to the polar body extruded stage. A peptide inhibitor (HT31) that disrupts AKAP/PKA interactions stimulates oocyte maturation in the continued presence of high cAMP. However, during the early minutes of maturation, type II PKA moves from cytoplasmic sites to the mitochondria, where it associates with AKAP1, and this is shown to be essential for maturation to continue irreversibly.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Profase Meiótica I/fisiología , Oocitos/crecimiento & desarrollo , Proteínas de Anclaje a la Quinasa A , Animales , AMP Cíclico/metabolismo , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Cinética , Ratones , Ratones Noqueados , Microscopía Fluorescente , Mitocondrias/metabolismo , Fosforilación , Transporte de Proteínas/fisiología
7.
Neuron ; 83(2): 361-371, 2014 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-25033180

RESUMEN

The serine hydrolase α/ß-hydrolase domain 6 (ABHD6) hydrolyzes the most abundant endocannabinoid (eCB) in the brain, 2-arachidonoylglycerol (2-AG), and controls its availability at cannabinoid receptors. We show that ABHD6 inhibition decreases pentylenetetrazole (PTZ)-induced generalized tonic-clonic and myoclonic seizure incidence and severity. This effect is retained in Cnr1(-/-) or Cnr2(-/-) mice, but blocked by addition of a subconvulsive dose of picrotoxin, suggesting the involvement of GABAA receptors. ABHD6 inhibition also blocked spontaneous seizures in R6/2 mice, a genetic model of juvenile Huntington's disease known to exhibit dysregulated eCB signaling. ABHD6 blockade retained its antiepileptic activity over chronic dosing and was not associated with psychomotor or cognitive effects. While the etiology of seizures in R6/2 mice remains unsolved, involvement of the hippocampus is suggested by interictal epileptic discharges, increased expression of vGLUT1 but not vGAT, and reduced Neuropeptide Y (NPY) expression. We conclude that ABHD6 inhibition may represent a novel antiepileptic strategy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Encéfalo/efectos de los fármacos , Carbamatos/uso terapéutico , Monoacilglicerol Lipasas/antagonistas & inhibidores , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/fisiopatología , Carbamatos/farmacología , Masculino , Ratones , Ratones Noqueados , Pentilenotetrazol , Receptores de Cannabinoides/genética , Convulsiones/inducido químicamente , Convulsiones/fisiopatología
8.
J Clin Invest ; 123(4): 1798-808, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23524966

RESUMEN

Sudden unexpected death in epilepsy (SUDEP) is the most common cause of death in intractable epilepsies, but physiological mechanisms that lead to SUDEP are unknown. Dravet syndrome (DS) is an infantile-onset intractable epilepsy caused by heterozygous loss-of-function mutations in the SCN1A gene, which encodes brain type-I voltage-gated sodium channel NaV1.1. We studied the mechanism of premature death in Scn1a heterozygous KO mice and conditional brain- and cardiac-specific KOs. Video monitoring demonstrated that SUDEP occurred immediately following generalized tonic-clonic seizures. A history of multiple seizures was a strong risk factor for SUDEP. Combined video-electroencephalography-electrocardiography revealed suppressed interictal resting heart-rate variability and episodes of ictal bradycardia associated with the tonic phases of generalized tonic-clonic seizures. Prolonged atropine-sensitive ictal bradycardia preceded SUDEP. Similar studies in conditional KO mice demonstrated that brain, but not cardiac, KO of Scn1a produced cardiac and SUDEP phenotypes similar to those found in DS mice. Atropine or N-methyl scopolamine treatment reduced the incidence of ictal bradycardia and SUDEP in DS mice. These findings suggest that SUDEP is caused by apparent parasympathetic hyperactivity immediately following tonic-clonic seizures in DS mice, which leads to lethal bradycardia and electrical dysfunction of the ventricle. These results have important implications for prevention of SUDEP in DS patients.


Asunto(s)
Arritmias Cardíacas/mortalidad , Epilepsias Mioclónicas/mortalidad , Animales , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/fisiopatología , Bloqueo Atrioventricular/tratamiento farmacológico , Bloqueo Atrioventricular/mortalidad , Bloqueo Atrioventricular/fisiopatología , Atropina/uso terapéutico , Bradicardia/tratamiento farmacológico , Bradicardia/mortalidad , Bradicardia/fisiopatología , Modelos Animales de Enfermedad , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/fisiopatología , Epilepsia Tónico-Clónica/tratamiento farmacológico , Epilepsia Tónico-Clónica/mortalidad , Epilepsia Tónico-Clónica/fisiopatología , Frecuencia Cardíaca , Humanos , Ratones , Ratones Noqueados , N-Metilescopolamina/uso terapéutico , Canal de Sodio Activado por Voltaje NAV1.1/genética , Parasimpatolíticos/uso terapéutico
9.
Channels (Austin) ; 7(6): 468-72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23965409

RESUMEN

Dravet Syndrome (DS) is an intractable genetic epilepsy caused by loss-of-function mutations in SCN1A, the gene encoding brain sodium channel Nav 1.1. DS is associated with increased frequency of sudden unexpected death in humans and in a mouse genetic model of this disease. Here we correlate the time course of declining expression of the murine embryonic sodium channel Nav 1.3 and the rise in expression of the adult sodium channel Nav 1.1 with susceptibility to epileptic seizures and increased incidence of sudden death in DS mice. Parallel studies with unaffected human brain tissue demonstrate similar decline in Nav 1.3 and increase in Nav 1.1 with age. In light of these results, we introduce the hypothesis that the natural loss Nav 1.3 channel expression in brain development, coupled with the failure of increase in functional Nav 1.1 channels in DS, defines a tipping point that leads to disinhibition of neural circuits, intractable seizures, co-morbidities, and premature death in this disease.


Asunto(s)
Muerte Súbita , Epilepsias Mioclónicas/metabolismo , Regulación de la Expresión Génica , Canales de Sodio/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Humanos , Ratones , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Canal de Sodio Activado por Voltaje NAV1.3/metabolismo , Factores de Tiempo
10.
J Biol Chem ; 283(12): 7390-400, 2008 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-18079112

RESUMEN

The kynurenine pathway of tryptophan degradation is hypothesized to play an important role in Huntington disease, a neurodegenerative disorder caused by a polyglutamine expansion in the protein huntingtin. Neurotoxic metabolites of the kynurenine pathway, generated in microglia and macrophages, are present at increased levels in the brains of patients and mouse models during early stages of disease, but the mechanism by which kynurenine pathway up-regulation occurs in Huntington disease is unknown. Here we report that expression of a mutant huntingtin fragment was sufficient to induce transcription of the kynurenine pathway in yeast and that this induction was abrogated by impairing the activity of the histone deacetylase Rpd3. Moreover, numerous genetic suppressors of mutant huntingtin toxicity that are functionally unrelated converged unexpectedly on the kynurenine pathway, supporting a critical role for the kynurenine pathway in mediating mutant huntingtin toxicity in yeast. Histone deacetylase-dependent regulation of the kynurenine pathway was also observed in a mouse model of Huntington disease, in which treatment with a neuroprotective histone deacetylase inhibitor blocked activation of the kynurenine pathway in microglia expressing a mutant huntingtin fragment in vitro and in vivo. These findings suggest that a mutant huntingtin fragment can perturb transcriptional programs in microglia, and thus implicate these cells as potential modulators of neurodegeneration in Huntington disease that are worthy of further investigation.


Asunto(s)
Histona Desacetilasas/metabolismo , Enfermedad de Huntington/metabolismo , Quinurenina/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Mutación , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transcripción Genética , Animales , Modelos Animales de Enfermedad , Expresión Génica , Histona Desacetilasas/genética , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Quinurenina/genética , Macrófagos/patología , Ratones , Microglía/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA