Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Psychiatry ; 22(10): 1464-1472, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27956739

RESUMEN

ANK3, encoding the adaptor protein Ankyrin-G (AnkG), has been implicated in bipolar disorder by genome-wide association studies. ANK3 has multiple alternative first exons, and a bipolar disorder-associated ANK3 variant has been shown to reduce the expression of exon 1b. Here we identify mechanisms through which reduced ANK3 exon 1b isoform expression disrupts neuronal excitation-inhibition balance. We find that parvalbumin (PV) interneurons and principal cells differentially express ANK3 first exon subtypes. PV interneurons express only isoforms containing exon 1b, whereas excitatory principal cells express exon 1e alone or both 1e and 1b. In transgenic mice deficient for exon 1b, PV interneurons lack voltage-gated sodium channels at their axonal initial segments and have increased firing thresholds and diminished action potential dynamic range. These mice exhibit an Ank3 gene dosage-dependent phenotype including behavior changes modeling bipolar disorder, epilepsy and sudden death. Thus ANK3's important association with human bipolar susceptibility may arise from imbalance between AnkG function in interneurons and principal cells and resultant excessive circuit sensitivity and output. AnkG isoform imbalance is a novel molecular endophenotype and potential therapeutic target.


Asunto(s)
Ancirinas/genética , Ancirinas/metabolismo , Trastorno Bipolar/genética , Trastorno Bipolar/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Adolescente , Empalme Alternativo , Animales , Trastorno Bipolar/patología , Niño , Epilepsia/patología , Exones , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Interneuronas/metabolismo , Interneuronas/patología , Ratones , Ratones Transgénicos , Parvalbúminas/metabolismo , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas
2.
Neurology ; 76(22): 1932-8, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21543734

RESUMEN

Sudden unexpected death in epilepsy (SUDEP) is a devastating complication of epilepsy and is not rare. The NIH and National Institute of Neurological Disorders and Stroke sponsored a 3-day multidisciplinary workshop to advance research into SUDEP and its prevention. Parallel sessions were held: one with a focus on the science of SUDEP, and the other with a focus on issues related to the education of health care practitioners and people with epilepsy. This report summarizes the discussions and recommendations of the workshop, including lessons learned from investigations of sudden infant death syndrome (SIDS), sudden cardiac death, autonomic and respiratory physiology, medical devices, genetics, and animal models. Recommendations include educating all people with epilepsy about SUDEP as part of their general education on the potential harm of seizures, except in extenuating circumstances. Increasing awareness of SUDEP may facilitate improved seizure control, possibly decreasing SUDEP incidence. There have been significant advances in our understanding of the clinical and physiologic features of SIDS, sudden cardiac death, and SUDEP in both people and animals. Research should continue to focus on the cardiac, autonomic, respiratory, and genetic factors that likely contribute to the risk of SUDEP. Multicenter collaborative research should be encouraged, especially investigations with direct implications for the prevention of SUDEP. An ongoing SUDEP Coalition has been established to facilitate this effort. With the expansion of clinical, genetic, and basic science research, there is reasonable hope of advancing our understanding of SUDEP and ultimately our ability to prevent it.


Asunto(s)
Muerte Súbita/etiología , Epilepsia/complicaciones , Epilepsia/fisiopatología , Humanos , National Institute of Neurological Disorders and Stroke (U.S.) , National Institutes of Health (U.S.) , Estados Unidos
3.
Sci Transl Med ; 1(2): 2ra6, 2009 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20368164

RESUMEN

Sudden unexplained death is a catastrophic complication of human idiopathic epilepsy, causing up to 18% of patient deaths. A molecular mechanism and an identified therapy have remained elusive. Here, we find that epilepsy occurs in mouse lines bearing dominant human LQT1 mutations for the most common form of cardiac long QT syndrome, which causes syncopy and sudden death. KCNQ1 encodes the cardiac KvLQT1 delayed rectifier channel, which has not been previously found in the brain. We have shown that, in these mice, this channel is found in forebrain neuronal networks and brainstem nuclei, regions in which a defect in the ability of neurons to repolarize after an action potential, as would be caused by this mutation, can produce seizures and dysregulate autonomic control of the heart. That long QT syndrome mutations in KCNQ1 cause epilepsy reveals the dual arrhythmogenic potential of an ion channelopathy coexpressed in heart and brain and motivates a search for genetic diagnostic strategies to improve risk prediction and prevention of early mortality in persons with seizure disorders of unknown origin.


Asunto(s)
Arritmias Cardíacas/genética , Encéfalo/fisiopatología , Muerte Súbita , Epilepsia/genética , Canal de Potasio KCNQ1/genética , Mutación Puntual , Secuencia de Aminoácidos , Animales , Arritmias Cardíacas/fisiopatología , Encéfalo/metabolismo , Electrocardiografía , Electroencefalografía , Perfilación de la Expresión Génica , Humanos , Canal de Potasio KCNQ1/química , Ratones , Datos de Secuencia Molecular , Canales de Potasio con Entrada de Voltaje/genética , Homología de Secuencia de Aminoácido
4.
J Neurogenet ; 21(4): 253-6, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18161587

RESUMEN

A recessive mutation in the mouse at the mocha locus (mh, chromosome 10) modulates the synchronous synaptic activation of neocortical neurons, resulting in a constant 6-7 Hz (theta) wave pattern in the electrocorticogram. The gene-linked brain rhythm is unaffected by motor behavior and cannot be desynchronized by sensory stimuli. This exemplary neurological mutation affecting cortical excitability is the first to reveal clearly that the predominance of a specific pattern of spontaneous brain wave activity can be inherited as a recessive trait.

5.
Cell Death Differ ; 11(11): 1223-33, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15286683

RESUMEN

Mcl1 is a Bcl2-related antiapoptotic protein originally isolated from human myeloid leukemia cells. Unlike Bcl2, expression has not been reported in CNS neurons. We isolated Mcl1 in a direct screen for candidate modifier genes of neuronal vulnerability by differential display of mRNAs upregulated following prolonged seizures in two mouse strains with contrasting levels of hippocampal cell death. Mcl1 is widely expressed in neurons, and transcription is rapidly induced in both strains. In resistant C57Bl/6J mice, Mcl1 protein levels remain persistently elevated in hippocampal pyramidal neurons after seizures, but fall rapidly in C3H/HeJ hippocampus, coinciding with extensive neuronal apoptosis. DNA damage and caspase-mediated cell death were strikingly increased in Mcl1-deficient mice when compared to +/+ littermates after similar seizures. We identify Mcl1 as a neuronal gene responsive to excitotoxic insult in the brain, and link relative levels of Mcl1 expression to inherited differences in neuronal thresholds for apoptosis.


Asunto(s)
Apoptosis , Sistema Nervioso Central/patología , Proteínas de Neoplasias/biosíntesis , Neuronas/patología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Animales , Western Blotting , Muerte Celular , Daño del ADN , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neuronas/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasas/metabolismo , Regulación hacia Arriba
6.
Cell Death Differ ; 10(12): 1329-35, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12934064

RESUMEN

Apoptosis can be mediated by mechanisms other than the traditional caspase-mediated cleavage cascade. There is growing recognition that alternative proteolytic enzymes such as the lysosomal cathepsin proteases can initiate or propagate proapoptotic signals, but it is currently unclear how cathepsins achieve these actions. Recent in vitro evidence suggests that cathepsins cleave the proapoptotic Bcl-2 family member Bid, thereby activating it and allowing it to induce the mitochondrial release of cytochrome c and subsequent apoptosis. We have tested this hypothesis in vivo by breeding mice that lack cathepsin inhibition (cystatin B-deficient mice) to Bid-deficient mice, to determine whether the apoptosis caused by cathepsins is dependent on Bid signaling. We found that cathepsins are still able to promote apoptosis even in the absence of Bid, indicating that these proteases mediate apoptosis via a different pathway, or that some other molecule can functionally substitute for Bid in this system.


Asunto(s)
Apoptosis , Proteínas Portadoras/metabolismo , Catepsinas/metabolismo , Epilepsias Mioclónicas Progresivas/patología , Transducción de Señal , Factores de Edad , Animales , Ataxia , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Western Blotting , Muerte Celular , Cerebelo/metabolismo , Cistatina B , Cistatinas/genética , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Genotipo , Etiquetado Corte-Fin in Situ , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Epilepsias Mioclónicas Progresivas/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
7.
Neurology ; 58(10): 1559-62, 2002 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-12034802

RESUMEN

Subcortical band heterotopia (SBH) is seen predominantly in females, resulting from mutations in the X-linked doublecortin (DCX) gene, and can present with mild mental retardation and epilepsy. Males carrying DCX mutations usually demonstrate lissencephaly and are clinically much more severely affected. This article reports two cases of males with SBH indistinguishable from the female phenotype, both resulting from somatic mosaicism for DCX mutation.


Asunto(s)
Encéfalo/patología , Coristoma/genética , Epilepsia/genética , Proteínas Asociadas a Microtúbulos , Mosaicismo/genética , Neuropéptidos/genética , Adulto , Niño , Coristoma/patología , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Epilepsia/patología , Humanos , Masculino , Mutación/genética
8.
Neurology ; 57(9): 1536-42, 2001 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-11706087

RESUMEN

The authors propose that epilepsy research embark on a revitalized effort to move from targeting control of symptoms to strategies for prevention and cure. The recent advances that make this a realistic goal include identification of genes mutated in inherited epilepsy syndromes, molecular characterization of brain networks, better imaging of sites of seizure origin, and developments in seizure prediction by quantitative EEG analysis. Research directions include determination of mechanisms of epilepsy development, identification of genes for common epilepsy syndromes through linkage analysis and gene chip technology, and validation of new models of epilepsy and epileptogenesis. Directions for therapeutics include identification of new molecular targets, focal methods of drug delivery tied to EEG activity, gene and cell therapy, and surgical and nonablative therapies. Integrated approaches, such as coupling imaging with electrophysiology, are central to progress in localizing regions of epilepsy development in people at risk and better seizure prediction and treatment for people with epilepsy.


Asunto(s)
Epilepsia/fisiopatología , Neurología/tendencias , Animales , Epilepsia/genética , Epilepsia/terapia , Humanos
9.
Epilepsia ; 42(9): 1103-11, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11580756

RESUMEN

PURPOSE: Clinical features associated with chromosome 1p36 deletion include characteristic craniofacial abnormalities, mental retardation, and epilepsy. The presence and severity of specific phenotypic features are likely to be correlated with loss of a distinct complement of genes in each patient. We hypothesize that hemizygous deletion of one, or a few, critical gene(s) controlling neuronal excitability is associated with the epilepsy phenotype. Because ion channels are important determinants of seizure susceptibility and the voltage-gated K(+) channel beta-subunit gene, KCNAB2, has been localized to 1p36, we propose that deletion of this gene may be associated with the epilepsy phenotype. METHODS: Twenty-four patients were evaluated by fluorescence in situ hybridization with a probe containing KCNAB2. Clinical details were obtained by neurologic examination and EEG. RESULTS: Nine patients are deleted for the KCNAB2 locus, and eight (89%) of these have epilepsy or epileptiform activity on EEG. The majority of patients have a severe seizure phenotype, including infantile spasms. In contrast, of those not deleted for KCNAB2, only 27% have chronic seizures, and none had infantile spasms. CONCLUSIONS: Lack of the beta subunit would be predicted to reduce K(+) channel-mediated membrane repolarization and increase neuronal excitability, suggesting a possible relation between loss of this gene and the development of seizures. Because some patients with seizures were not deleted for KCNAB2, there may be additional genes within 1p36 that contribute to epilepsy in this syndrome. Hemizygosity of this gene in a majority of monosomy 1p36 syndrome patients with epilepsy suggests that haploinsufficiency for KCNAB2 is a significant risk factor for epilepsy.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 1/genética , Epilepsia/genética , Canales de Potasio/genética , Adolescente , Niño , Preescolar , Anomalías Craneofaciales/epidemiología , Anomalías Craneofaciales/genética , Electroencefalografía , Epilepsia/diagnóstico , Epilepsia/epidemiología , Humanos , Hibridación Fluorescente in Situ , Lactante , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/genética , Mutación/genética , Canales de Potasio con Entrada de Voltaje/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Neurosci ; 21(11): 3721-8, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11356859

RESUMEN

Regional variation in synaptic efficacy is an important determinant of associative processing as information flows through major circuits of the brain. The perforant path is the principal route of entry from cortex to the hippocampus and contains the first synapse in the cortical-hippocampal projection pathway. We used optical imaging techniques to analyze presynaptic Ca(2+) entry and neurotransmitter release at synapses in the medial perforant path linking stellate neurons located in layer II of the entorhinal cortex to granule cells in the dentate gyrus. Similar to other excitatory central synapses, the relationship between neurotransmitter release and the amount of Ca(2+) influx can be best described by a Hill equation with a Hill coefficient of 3.5. Our Ca(2+) channel toxin studies indicate that P/Q-type channels are the predominant Ca(2+) source triggering neurotransmitter release in this pathway, as shown by a potent inhibition of Ca(2+) entry and synaptic transmission by the P/Q-type channel blocker omega-agatoxin IVA. However, compared with the downstream hippocampal pyramidal neuron CA3-CA1 synapse, neurotransmitter release was less sensitive to the N-type Ca(2+) channel blocker omega-conotoxin GVIA, although the amount of N-type Ca(2+) current is comparable. The contribution of N-type channels to neurotransmitter release approximates that found at the CA3-CA1 synapse when tested under lower [Ca(2+)](o), which effectively reduces the size of the Ca(2+) microdomain surrounding each channel. These results suggest that P/Q-type channels are more closely associated with release machinery then N-type channels at this synapse and that cooperativity differences for each channel subtype may characterize variations in signaling at central synapses.


Asunto(s)
Canales de Calcio/metabolismo , Corteza Entorrinal/metabolismo , Neuronas/metabolismo , Neurotransmisores/metabolismo , Terminales Presinápticos/metabolismo , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/clasificación , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/metabolismo , Giro Dentado/citología , Giro Dentado/fisiología , Corteza Entorrinal/citología , Corteza Entorrinal/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Colorantes Fluorescentes , Técnicas In Vitro , Ratones , Modelos Neurológicos , Neuronas/efectos de los fármacos , Vía Perforante/citología , Vía Perforante/efectos de los fármacos , Vía Perforante/metabolismo , Terminales Presinápticos/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo
11.
J Neurosci Res ; 64(3): 268-76, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11319771

RESUMEN

Excess neuronal activity upregulates the expression of two neurotrophins, nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) in adult hippocampus. Nerve growth factor has been shown to contribute the induction of aberrant hippocampal mossy fiber sprouting in the inner molecular layer of the dentate gyrus, however the role of prolonged brain-derived neurotrophic factor exposure is uncertain. We examined the distribution and plasticity of mossy fibers in transgenic mice with developmental overexpression of brain-derived neurotrophic factor. Despite 2--3-fold elevated BDNF levels in the hippocampus sufficient to increase the intensity of neuropeptide Y immunoreactivity in interneurons, no visible changes in mossy fiber Timm staining patterns were observed in the inner molecular layer of adult mutant hippocampus compared to wild-type mice. In addition, no changes of the mRNA expression of two growth-associated proteins, GAP-43 and SCG-10 were found. These data suggest that early and persistent elevations of brain-derived neurotrophic factor in granule cells are not sufficient to elicit this pattern of axonal plasticity in the hippocampus.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína GAP-43/metabolismo , Fibras Musgosas del Hipocampo/metabolismo , Neuropéptido Y/metabolismo , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
12.
Genomics ; 71(3): 339-50, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11170751

RESUMEN

The CACNG1 gene on chromosome 17q24 encodes an integral membrane protein that was originally isolated as the regulatory gamma subunit of voltage-dependent Ca2+ channels from skeletal muscle. The existence of an extended family of gamma subunits was subsequently demonstrated upon identification of CACNG2 (22q13), CACNG3 (16p12-p13), and CACNG4 and CACNG5 (17q24). In this study, we describe a cluster of three novel gamma subunit genes, CACNG6, CACNG7, and CACNG8, located in a tandem array on 19q13.4. Phylogenetic analysis indicates that this array is paralogous to the cluster containing CACNG1, CACNG5, and CACNG4, respectively, on chromosome 17q24. We developed sensitive RT-PCR assays and examined the expression profile of each member of the gamma subunit gene family, CACNG1-CACNG8. Analysis of 24 human tissues plus 3 dissected brain regions revealed that CACNG1 through CACNG8 are all coexpressed in fetal and adult brain and differentially transcribed among a wide variety of other tissues. The expression of distinct complements of gamma subunit isoforms in different cell types may be an important mechanism for regulating Ca2+ channel function.


Asunto(s)
Canales de Calcio/genética , Cromosomas Humanos Par 19 , Familia de Multigenes , Secuencia de Aminoácidos , Secuencia de Bases , Encéfalo/embriología , Encéfalo/metabolismo , Línea Celular , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Clonación Molecular , Evolución Molecular , Regulación de la Expresión Génica , Genoma , Hipocampo/metabolismo , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Filogenia , Isoformas de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Distribución Tisular , Transcripción Genética
13.
J Neurosci ; 21(4): 1169-78, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11160387

RESUMEN

Rocker (gene symbol rkr), a new neurological mutant phenotype, was found in descendents of a chemically mutagenized male mouse. Mutant mice display an ataxic, unstable gait accompanied by an intention tremor, typical of cerebellar dysfunction. These mice are fertile and appear to have a normal life span. Segregation analysis reveals rocker to be an autosomal recessive trait. The overall cytoarchitecture of the young adult brain appears normal, including its gross cerebellar morphology. Golgi-Cox staining, however, reveals dendritic abnormalities in the mature cerebellar cortex characterized by a reduction of branching in the Purkinje cell dendritic arbor and a "weeping willow" appearance of the secondary branches. Using simple sequence length polymorphism markers, the rocker locus was mapped to mouse chromosome 8 within 2 centimorgans of the calcium channel alpha1a subunit (Cacna1a, formerly known as tottering) locus. Complementation tests with the leaner mutant allele (Cacna1a(la)) produced mutant animals, thus identifying rocker as a new allele of Cacna1a (Cacna1a(rkr)). Sequence analysis of the cDNA revealed rocker to be a point mutation resulting in an amino acid exchange: T1310K between transmembrane regions 5 and 6 in the third homologous domain. Important distinctions between rocker and the previously characterized alleles of this locus include the absence of aberrant tyrosine hydroxylase expression in Purkinje cells and the separation of the absence seizures (spike/wave type discharges) from the paroxysmal dyskinesia phenotype. Overall these findings point to an important dissociation between the seizure phenotypes and the abnormalities in catecholamine metabolism, and they emphasize the value of allelic series in the study of gene function.


Asunto(s)
Canales de Calcio/genética , Canales de Calcio/metabolismo , Enfermedades Cerebelosas/genética , Alelos , Animales , Ataxia/etiología , Canales de Calcio Tipo N , Canales de Calcio Tipo P , Canales de Calcio Tipo Q , Enfermedades Cerebelosas/patología , Enfermedades Cerebelosas/fisiopatología , Cerebelo/patología , Cerebelo/fisiopatología , Mapeo Cromosómico , Cruzamientos Genéticos , Análisis Mutacional de ADN , Femenino , Genes Recesivos , Prueba de Complementación Genética , Ligamiento Genético/fisiología , Marcadores Genéticos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/genética , Mutación Puntual , Células de Purkinje/patología , Temblor/etiología
14.
Epilepsia ; 42 Suppl 5: 11-5, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11887961

RESUMEN

Two categories of mouse models of human epilepsy are now contributing to the experimental analysis of inherited seizure disorders. The first type includes homologous genetic models arrived at in the classic way; the genes from human inherited epilepsy syndromes are cloned, and mice are recreated with functionally identical mutations. The second category involves the reverse strategy: mutating single genes in mice and determining whether the newly created nervous system develops epilepsy. These "gene-forward" models define specific candidate genes that can then be tested for possible involvement in human epilepsies. Spontaneous mutation of genes in mice and other species is also a source for candidate genes. As each of these genes and their physiologic functions is defined, the focus can shift to (a) fully characterizing the clinical epilepsy phenotype, (b) tracing the steps in the molecular pathogenesis of the disorder, and (c) pinpointing molecular targets for early intervention. Along with providing a unique opportunity to understand the mechanisms of inherited epileptogenesis, the mouse models serve as ideal biological test systems to search for novel therapeutic strategies.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/genética , Síndrome de Angelman/genética , Animales , Anticonvulsivantes/uso terapéutico , Clonación Molecular/métodos , Diseño de Fármacos , Epilepsia/tratamiento farmacológico , Eliminación de Gen , Humanos , Canales Iónicos/genética , Ratones , Ratones Mutantes Neurológicos , Mutación/genética , Fenotipo , Síndrome de Unverricht-Lundborg/genética
15.
J Neurosci ; 20(24): 9071-85, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11124984

RESUMEN

Voltage-gated K(+) channels of the Kv3 subfamily have unusual electrophysiological properties, including activation at very depolarized voltages (positive to -10 mV) and very fast deactivation rates, suggesting special roles in neuronal excitability. In the brain, Kv3 channels are prominently expressed in select neuronal populations, which include fast-spiking (FS) GABAergic interneurons of the neocortex, hippocampus, and caudate, as well as other high-frequency firing neurons. Although evidence points to a key role in high-frequency firing, a definitive understanding of the function of these channels has been hampered by a lack of selective pharmacological tools. We therefore generated mouse lines in which one of the Kv3 genes, Kv3.2, was disrupted by gene-targeting methods. Whole-cell electrophysiological recording showed that the ability to fire spikes at high frequencies was impaired in immunocytochemically identified FS interneurons of deep cortical layers (5-6) in which Kv3.2 proteins are normally prominent. No such impairment was found for FS neurons of superficial layers (2-4) in which Kv3.2 proteins are normally only weakly expressed. These data directly support the hypothesis that Kv3 channels are necessary for high-frequency firing. Moreover, we found that Kv3.2 -/- mice showed specific alterations in their cortical EEG patterns and an increased susceptibility to epileptic seizures consistent with an impairment of cortical inhibitory mechanisms. This implies that, rather than producing hyperexcitability of the inhibitory interneurons, Kv3.2 channel elimination suppresses their activity. These data suggest that normal cortical operations depend on the ability of inhibitory interneurons to generate high-frequency firing.


Asunto(s)
Corteza Cerebral/fisiopatología , Predisposición Genética a la Enfermedad , Inhibición Neural , Neuropéptidos/deficiencia , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/deficiencia , Convulsiones/fisiopatología , Potenciales de Acción/genética , Animales , Conducta Animal , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Clonación Molecular , Electroencefalografía , Femenino , Marcación de Gen , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibición Neural/efectos de los fármacos , Inhibición Neural/genética , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Técnicas de Placa-Clamp , Fenotipo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Convulsiones/genética , Canales de Potasio Shaw , Células Madre , Tetraetilamonio/farmacología
16.
Nat Genet ; 26(2): 191-4, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11017075

RESUMEN

Spinocerebellar ataxia type 10 (SCA10; MIM 603516; refs 1,2) is an autosomal dominant disorder characterized by cerebellar ataxia and seizures. The gene SCA10 maps to a 3.8-cM interval on human chromosome 22q13-qter (refs 1,2). Because several other SCA subtypes show trinucleotide repeat expansions, we examined microsatellites in this region. We found an expansion of a pentanucleotide (ATTCT) repeat in intron 9 of SCA10 in all patients in five Mexican SCA10 families. There was an inverse correlation between the expansion size, up to 22.5 kb larger than the normal allele, and the age of onset (r2=0.34, P=0.018). Analysis of 562 chromosomes from unaffected individuals of various ethnic origins (including 242 chromosomes from Mexican persons) showed a range of 10 to 22 ATTCT repeats with no evidence of expansions. Our data indicate that the new SCA10 intronic ATTCT pentanucleotide repeat in SCA10 patients is unstable and represents the largest microsatellite expansion found so far in the human genome.


Asunto(s)
Cromosomas Humanos Par 22 , ADN/genética , Secuencias Repetitivas de Ácidos Nucleicos , Ataxias Espinocerebelosas/genética , Animales , Pueblo Asiatico/genética , Encéfalo/metabolismo , Encéfalo/patología , Mapeo Cromosómico , ADN/sangre , ADN/química , Epilepsia/genética , Epilepsia/patología , Femenino , Humanos , Masculino , Americanos Mexicanos/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Linaje , Polimorfismo Genético , Ataxias Espinocerebelosas/patología , Estados Unidos , Población Blanca/genética
19.
Curr Opin Genet Dev ; 10(3): 286-91, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10826987

RESUMEN

Inherited disorders of voltage-gated ion channels are a recently recognized etiology of epilepsy in the developing and mature central nervous system. Two human epilepsy syndromes, benign familial neonatal convulsions and generalized epilepsy with febrile seizures plus, represent K+ and Na+ channelopathies, and other newly defined syndromes have now been mapped to chromosomal regions that are rich in ion channel genes. Experimental mouse models promise a resolution of their intriguing pathophysiology, which includes a diverse array of cellular phenotypes consistent with the differential contributions of individual channels to excitability in neural networks.


Asunto(s)
Encéfalo/fisiopatología , Epilepsia/genética , Epilepsia/fisiopatología , Canales Iónicos/genética , Animales , Canales de Calcio/fisiología , Humanos , Canales Iónicos/fisiología , Ratones , Canales de Potasio/genética , Canales de Sodio/genética
20.
Neuroscience ; 95(3): 639-45, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10670432

RESUMEN

The P/Q type voltage-gated Ca2+ channels are involved in membrane excitability and Ca2+-dependent neurotransmitter release within the CNS. Mutations in the CacnalA gene encoding the alpha1A subunit of the P/Q type Ca2+ channel have recently been reported in tottering mice and a more severely affected allele, leaner. Here we show using in vivo cortical microdialysis that evoked increases of extracellular glutamate levels are markedly attenuated in both mutants upon KCl-induced depolarization compared with wild-type mice. Tottering and leaner mice also show a 10-fold resistance to cortical spreading depression induced by cortical electrical stimulation or KCl application to the pial surface. A slower transcortical propagation speed and failure to sustain regenerative spread of the depolarizing wave were more pronounced in leaner neocortex. Both signaling defects appeared unrelated to the developmental history of repeated cortical spike-wave discharges, since neither were observed in the stargazer mouse, a Ca2+ channel gamma2 subunit mutant with a similar seizure phenotype. These data demonstrate two cortical excitability defects revealed by prolonged depolarization in cerebral networks expressing mutant P/Q type Ca2+ channels, and are the first to identify a gene linked to a spreading depression phenotype.


Asunto(s)
Canales de Calcio Tipo P/genética , Depresión de Propagación Cortical/fisiología , Mutación/fisiología , Neurotransmisores/metabolismo , Animales , Umbral Diferencial/fisiología , Electrofisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Potasio/farmacología , Isoformas de Proteínas/genética , Valores de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA