Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 695: 149481, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38211534

RESUMEN

Spinocerebellar ataxia type 6 (SCA6) is a polyglutamine (polyQ) disease, which is caused by the elongation of CAG repeats encoding polyQ in the CACNA1A gene. The CACNA1A gene encodes two proteins, namely, α1A (a subunit of the plasma membrane calcium channel), which is translated in its entire length, and α1ACT, which is translated from the second cistron, and both proteins have a polyQ tract. The α1A-polyQ and α1ACT-polyQ proteins with an elongated polyQ stretch have been reported to form aggregates in cells and induce neuronal cell death, but the subcellular localization of these proteins and their cytotoxic properties remain unclear. In this study, we first analyzed SCA6 model mice and found that α1A-polyQlong localized mainly to the Golgi apparatus, whereas a portion of α1ACT-polyQlong localized to the nucleus. Analysis using Neuro2a cells also showed similar subcellular localizations of these proteins, and a proportion of both proteins localized to the endoplasmic reticulum (ER). Cytotoxic studies demonstrated that both proteins induce both the ER stress response and apoptosis, indicating that they are able to induce ER stress-induced apoptosis.


Asunto(s)
Canales de Calcio Tipo N , Ataxias Espinocerebelosas , Animales , Ratones , Canales de Calcio/metabolismo , Canales de Calcio Tipo N/metabolismo , Retículo Endoplásmico/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo
2.
Genome Res ; 33(3): 435-447, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37307504

RESUMEN

Tandem repeats (TRs) are one of the largest sources of polymorphism, and their length is associated with gene regulation. Although previous studies reported several tandem repeats regulating gene splicing in cis (spl-TRs), no large-scale study has been conducted. In this study, we established a genome-wide catalog of 9537 spl-TRs with a total of 58,290 significant TR-splicing associations across 49 tissues (false discovery rate 5%) by using Genotype-Tissue expression (GTex) Project data. Regression models explaining splicing variation by using spl-TRs and other flanking variants suggest that at least some of the spl-TRs directly modulate splicing. In our catalog, two spl-TRs are known loci for repeat expansion diseases, spinocerebellar ataxia 6 (SCA6) and 12 (SCA12). Splicing alterations by these spl-TRs were compatible with those observed in SCA6 and SCA12. Thus, our comprehensive spl-TR catalog may help elucidate the pathomechanism of genetic diseases.


Asunto(s)
Ingeniería Genética , Empalme del ARN , Humanos , Polimorfismo Genético , Secuencias Repetidas en Tándem
3.
Brain ; 143(6): 1811-1825, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32436573

RESUMEN

The polyglutamine (polyQ) diseases are a group of inherited neurodegenerative diseases that include Huntington's disease, various spinocerebellar ataxias, spinal and bulbar muscular atrophy, and dentatorubral pallidoluysian atrophy. They are caused by the abnormal expansion of a CAG repeat coding for the polyQ stretch in the causative gene of each disease. The expanded polyQ stretches trigger abnormal ß-sheet conformational transition and oligomerization followed by aggregation of the polyQ proteins in the affected neurons, leading to neuronal toxicity and neurodegeneration. Disease-modifying therapies that attenuate both symptoms and molecular pathogenesis of polyQ diseases remain an unmet clinical need. Here we identified arginine, a chemical chaperone that facilitates proper protein folding, as a novel compound that targets the upstream processes of polyQ protein aggregation by stabilizing the polyQ protein conformation. We first screened representative chemical chaperones using an in vitro polyQ aggregation assay, and identified arginine as a potent polyQ aggregation inhibitor. Our in vitro and cellular assays revealed that arginine exerts its anti-aggregation property by inhibiting the toxic ß-sheet conformational transition and oligomerization of polyQ proteins before the formation of insoluble aggregates. Arginine exhibited therapeutic effects on neurological symptoms and protein aggregation pathology in Caenorhabditis elegans, Drosophila, and two different mouse models of polyQ diseases. Arginine was also effective in a polyQ mouse model when administered after symptom onset. As arginine has been safely used for urea cycle defects and for mitochondrial myopathy, encephalopathy, lactic acid and stroke syndrome patients, and efficiently crosses the blood-brain barrier, a drug-repositioning approach for arginine would enable prompt clinical application as a promising disease-modifier drug for the polyQ diseases.


Asunto(s)
Arginina/metabolismo , Arginina/farmacología , Péptidos/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Drosophila/metabolismo , Femenino , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Endogámicos , Chaperonas Moleculares/genética , Péptidos/genética , Agregación Patológica de Proteínas , Conformación Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Ataxias Espinocerebelosas/genética
4.
Nat Commun ; 8(1): 1864, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29192206

RESUMEN

YAP and its neuronal isoform YAPdeltaC are implicated in various cellular functions. We found that expression of YAPdeltaC during development, but not adulthood, rescued neurodegeneration phenotypes of mutant ataxin-1 knock-in (Atxn1-KI) mice. YAP/YAPdeltaC interacted with RORα via the second WW domain and served as co-activators of its transcriptional activity. YAP/YAPdeltaC formed a transcriptional complex with RORα on cis-elements of target genes and regulated their expression. Both normal and mutant Atxn1 interacted with YAP/YAPdeltaC, but only mutant Atxn1 depleted YAP/YAPdeltaC from the RORα complex to suppress transcription on short timescales. Over longer periods, mutant Atxn1 also decreased RORα in vivo. Genetic supplementation of YAPdeltaC restored the RORα and YAP/YAPdeltaC levels, recovered YAP/YAPdeltaC in the RORα complex and normalized target gene transcription in Atxn1-KI mice in vivo. Collectively, our data suggest that functional impairment of YAP/YAPdeltaC by mutant Atxn1 during development determines the adult pathology of SCA1 by suppressing RORα-mediated transcription.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ataxina-1/genética , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Neuronas/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fosfoproteínas/metabolismo , Ataxias Espinocerebelosas/genética , Animales , Proteínas de Ciclo Celular , Cerebelo/citología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Masculino , Ratones , Fenotipo , Isoformas de Proteínas , Prueba de Desempeño de Rotación con Aceleración Constante , Ataxias Espinocerebelosas/fisiopatología , Proteínas Señalizadoras YAP
5.
Hum Mol Genet ; 26(16): 3094-3104, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28510727

RESUMEN

Alternative splicing (AS) that occurs at the final coding exon (exon 47) of the Cav2.1 voltage-gated calcium channel (VGCC) gene produces two major isoforms in the brain, MPI and MPc. These isoforms differ in their splice acceptor sites; human MPI is translated into a polyglutamine tract associated with spinocerebellar ataxia type 6 (SCA6), whereas MPc splices to an immediate stop codon, resulting in a shorter cytoplasmic tail. To gain insight into the functional role of the AS in vivo and whether modulating the splice patterns at this locus can be a potential therapeutic strategy for SCA6, here we created knockin mice that exclusively express MPc by inserting the splice-site mutation. The resultant Cacna1aCtmKO/CtmKO mice developed non-progressive neurological phenotypes, featuring early-onset ataxia and absence seizure without significant alterations in the basic properties of the channel. Interactions of Cav2.1 with Cavß4 and Rimbp2 were significantly reduced while those with GABAB2 were enhanced in the cerebellum of Cacna1aCtmKO/CtmKO mice. Treatment with the GABAB antagonist CGP35348 partially rescued the motor impairments seen in Cacna1aCtmKO/CtmKO mice. These results suggest that the carboxyl-terminal domain of Cav2.1 is not essential for maintaining the basic properties of the channel in the cerebellar Purkinje neurons but is involved in multiple interactions of Cav2.1 with other proteins, and plays an essential role in preventing a complex neurological disease.


Asunto(s)
Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Empalme Alternativo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Cerebelo/metabolismo , Exones , Técnicas de Sustitución del Gen , Humanos , Ratones , Células de Purkinje/metabolismo , Isoformas de ARN , Sitios de Empalme de ARN , Ataxias Espinocerebelosas/genética
6.
Hum Mol Genet ; 25(20): 4507-4517, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28173004

RESUMEN

Point mutations in the vacuolar protein sorting 35 gene (VPS35) have been associated with an autosomal dominant form of late-onset Parkinson disease (PARK17), but there has been considerable debate over whether it is caused by a loss- or gain-of-function mechanism and over the intracellular target site of neurotoxicity. To investigate the pathogenesis of PARK17 in vivo, we generated Vps35 D620N knock-in (KI) mice, expressing the homologous mutant protein with endogenous patterns of expression, simultaneously with Vps35 deletion 1 (Del1) mice, which carry 1bp deletion in the exon15 of Vps35, by CRISPR/Cas9-mediated genome engineering. Neither homozygous nor heterozygous Vps35 D620N KI mice suffered from premature death or developed clear neurodegeneration up to 70 weeks of age. Vps35 Del1 allele appeared to be a null or at least severely hypomorphic allele and homozygous Vps35 Del1 showed early embryonic lethality. Heterozygous crossings between Del1 and D620N knock-in mice revealed that the D620N/Del1 compound heterozygous mice, but not heterozygous Del1 mice, suffered from survival disadvantage. In vivo microdialysis showed that DA release evoked by 120 mM potassium chloride was significantly reduced in the caudate putamen of adult homozygous Vps35 D620N KI mice. Taken together, these results suggest that Vps35 D620N allele is a partial-loss-of-function allele and that such a genetic predisposition and age-related alterations in the nigrostriatal dopamine system cooperatively influence the pathogenesis of PARK17.


Asunto(s)
Modelos Animales de Enfermedad , Dopamina/metabolismo , Mutación , Enfermedad de Parkinson/metabolismo , Proteínas de Transporte Vesicular/genética , Animales , Técnicas de Sustitución del Gen , Homocigoto , Ratones , Neostriado/metabolismo , Neostriado/fisiopatología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/fisiopatología
7.
Hum Mol Genet ; 25(20): 4432-4447, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28173122

RESUMEN

DNA damage and repair is a critical domain of many neurodegenerative diseases. In this study, we focused on RpA1, a candidate key molecule in polyQ disease pathologies, and tested the therapeutic effect of adeno-associated virus (AAV) vector expressing RpA1 on mutant Ataxin-1 knock-in (Atxn1-KI) mice. We found significant effects on motor functions, normalized DNA damage markers (γH2AX and 53BP1), and improved Purkinje cell morphology; effects that lasted for 50 weeks following AAV-RpA1 infection. In addition, we confirmed that AAV-RpA1 indirectly recovered multiple cellular functions such as RNA splicing, transcription and cell cycle as well as abnormal morphology of dendrite and dendritic spine of Purkinje cells in Atxn1-KI mice. All these results suggested a possibility of gene therapy with RpA1 for SCA1.


Asunto(s)
Ataxina-1/genética , Reparación del ADN , Mutación , Proteína de Replicación A/metabolismo , Ataxias Espinocerebelosas/metabolismo , Animales , Ciclo Celular , ADN/metabolismo , Daño del ADN , Dependovirus , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Terapia Genética , Ratones , Células de Purkinje/metabolismo , Células de Purkinje/patología , Células de Purkinje/fisiología , ARN/metabolismo , Empalme del ARN , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/fisiopatología , Transcripción Genética
8.
Sci Rep ; 5: 16102, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26531852

RESUMEN

Late-onset neurodegenerative diseases are characterized by neurological symptoms and progressive neuronal death. Accumulating evidence suggests that neuronal dysfunction, rather than neuronal death, causes the symptoms of neurodegenerative diseases. However, the mechanisms underlying the dysfunction that occurs prior to cell death remain unclear. To investigate the synaptic basis of this dysfunction, we employed in vivo two-photon imaging to analyse excitatory postsynaptic dendritic protrusions. We used Sca1(154Q/2Q) mice, an established knock-in mouse model of the polyglutamine disease spinocerebellar ataxia type 1 (SCA1), which replicates human SCA1 features including ataxia, cognitive impairment, and neuronal death. We found that Sca1(154Q/2Q) mice exhibited greater synaptic instability than controls, without synaptic loss, in the cerebral cortex, where obvious neuronal death is not observed, even before the onset of distinct symptoms. Interestingly, this abnormal synaptic instability was evident in Sca1(154Q/2Q) mice from the synaptic developmental stage, and persisted into adulthood. Expression of synaptic scaffolding proteins was also lower in Sca1(154Q/2Q) mice than controls before synaptic maturation. As symptoms progressed, synaptic loss became evident. These results indicate that aberrant synaptic instability, accompanied by decreased expression of scaffolding proteins during synaptic development, is a very early pathology that precedes distinct neurological symptoms and neuronal cell death in SCA1.


Asunto(s)
Ataxina-1/genética , Ataxias Espinocerebelosas/patología , Sinapsis/metabolismo , Animales , Ataxina-1/metabolismo , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Corteza Cerebral/metabolismo , Dendritas/metabolismo , Dendritas/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Microscopía de Fluorescencia por Excitación Multifotónica , Proteínas Nucleares/metabolismo , Ataxias Espinocerebelosas/metabolismo , Factores de Tiempo
9.
Hum Mol Genet ; 24(17): 4780-91, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26034136

RESUMEN

Spinocerebellar ataxia type 6 (SCA6) is dominantly inherited neurodegenerative disease, caused by an expansion of CAG repeat encoding a polyglutamine (PolyQ) tract in the Cav2.1 voltage-gated calcium channel. Its key pathological features include selective degeneration of the cerebellar Purkinje cells (PCs), a common target for PolyQ-induced toxicity in various SCAs. Mutant Cav2.1 confers toxicity primarily through a toxic gain-of-function mechanism; however, its molecular basis remains elusive. Here, we studied the cerebellar gene expression patterns of young Sca6-MPI(118Q/118Q) knockin (KI) mice, which expressed mutant Cav2.1 from an endogenous locus and recapitulated many phenotypic features of human SCA6. Transcriptional signatures in the MPI(118Q/118Q) mice were distinct from those in the Sca1(154Q/2Q) mice, a faithful SCA1 KI mouse model. Temporal expression profiles of the candidate genes revealed that the up-regulation of genes associated with microglial activation was initiated before PC degeneration and was augmented as the disease progressed. Histological analysis of the MPI(118Q/118Q) cerebellum showed the predominance of M1-like pro-inflammatory microglia and it was concomitant with elevated expression levels of tumor necrosis factor, interleukin-6, Toll-like receptor (TLR) 2 and 7. Genetic ablation of MyD88, a major adaptor protein conveying TLR signaling, altered expression patterns of M1/M2 microglial phenotypic markers in the MPI(118Q/118Q) cerebellum. More importantly, it ameliorated PC loss and partially rescued motor impairments in the early disease phase. These results suggest that early neuroinflammatory response may play an important role in the pathogenesis of SCA6 and its modulation could pave the way for slowing the disease progression during the early stage of the disease.


Asunto(s)
Eliminación de Gen , Factor 88 de Diferenciación Mieloide/genética , Células de Purkinje/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología , Animales , Biomarcadores , Cerebelo/metabolismo , Cerebelo/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Microglía/metabolismo , Actividad Motora , Factor 88 de Diferenciación Mieloide/deficiencia , ARN Mensajero/genética
10.
Proc Natl Acad Sci U S A ; 109(43): 17693-8, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23054835

RESUMEN

Spinocerebellar ataxia type 6 (SCA6) is a neurodegenerative disease caused by the expansion of a polyglutamine tract in the Ca(v)2.1 voltage-gated calcium channel. To elucidate how the expanded polyglutamine tract in this plasma membrane protein causes the disease, we created a unique knockin mouse model that modestly overexpressed the mutant transcripts under the control of an endogenous promoter (MPI-118Q). MPI-118Q mice faithfully recapitulated many features of SCA6, including selective Purkinje cell degeneration. Surprisingly, analysis of inclusion formation in the mutant Purkinje cells indicated the lysosomal localization of accumulated mutant Ca(v)2.1 channels in the absence of autophagic response. The lack of cathepsin B, a major lysosomal cysteine proteinase, exacerbated the loss of Purkinje cells and was accompanied by an acceleration of inclusion formation in this model. Thus, the pathogenic mechanism of SCA6 involves the endolysosomal degradation pathway, and unique pathological features of this model further illustrate the pivotal role of protein context in the pathogenesis of polyglutamine diseases.


Asunto(s)
Modelos Animales de Enfermedad , Lisosomas/fisiología , Células de Purkinje/patología , Ataxias Espinocerebelosas/patología , Animales , Autofagia , Ratones , Ratones Transgénicos
12.
EMBO J ; 29(14): 2446-60, 2010 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-20531390

RESUMEN

Non-cell-autonomous effect of mutant proteins expressed in glia has been implicated in several neurodegenerative disorders, whereas molecules mediating the toxicity are currently not known. We identified a novel molecule named multiple alpha-helix protein located at ER (Maxer) downregulated by mutant ataxin-1 (Atx1) in Bergmann glia. Maxer is an endoplasmic reticulum (ER) membrane protein interacting with CDK5RAP3. Maxer anchors CDK5RAP3 to the ER and inhibits its function of Cyclin D1 transcription repression in the nucleus. The loss of Maxer eventually induces cell accumulation at G1 phase. It was also shown that mutant Atx1 represses Maxer and inhibits proliferation of Bergmann glia in vitro. Consistently, Bergmann glia are reduced in the cerebellum of mutant Atx1 knockin mice before onset. Glutamate-aspartate transporter reduction in Bergmann glia by mutant Atx1 and vulnerability of Purkinje cell to glutamate are both strengthened by Maxer knockdown in Bergmann glia, whereas Maxer overexpression rescues them. Collectively, these results suggest that the reduction of Maxer mediates functional deficiency of Bergmann glia, and might contribute to the non-cell-autonomous pathology of SCA1.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso , Neuroglía/metabolismo , Proteínas Nucleares , Secuencia de Aminoácidos , Animales , Ataxina-1 , Ataxinas , Proteínas de Ciclo Celular , Proliferación Celular , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuroglía/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Distribución Tisular , Proteínas Supresoras de Tumor
13.
Rinsho Shinkeigaku ; 50(11): 858-60, 2010 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-21921472

RESUMEN

Spinocerebellar ataxia type 6 (SCA6) is one of the common dominantly inherited ataxias in Japan, featuring late-onset ataxia and selective Purkinje cell (PC) degeneration. Molecular pathogenesis of SCA6 has been attracting considerable attention since it is caused by small CAG repeat expansions within the Ca(v)2.1 voltage-gated Ca(++) channel gene (CACNA1A). During the past 9 years, efforts have been made to generate and analyze a precise SCA6 model in order to disclose its molecular pathogenesis in vivo. Evidence indicates that the SCA6 mutation does not directly change the basic properties of the channel but rather exerts neurotoxicity through a mechanism associated with age-dependent accumulation of the expanded polyglutamine protein. We envisage further analysis on a knockin model developing PC degeneration at their young age will lead to elucidation of the molecular pathways involved in SCA6 and thus be useful for developing therapeutic strategies against the disease.


Asunto(s)
Canales de Calcio/genética , Ataxias Espinocerebelosas/genética , Animales , Canales de Calcio Tipo N , Modelos Animales de Enfermedad , Humanos , Ratones , Mutación , Péptidos , Ataxias Espinocerebelosas/patología , Expansión de Repetición de Trinucleótido
14.
Proc Natl Acad Sci U S A ; 105(33): 11987-92, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18687887

RESUMEN

Spinocerebellar ataxia type 6 (SCA6) is a neurodegenerative disorder caused by CAG repeat expansions within the voltage-gated calcium (Ca(V)) 2.1 channel gene. It remains controversial whether the mutation exerts neurotoxicity by changing the function of Ca(V)2.1 channel or through a gain-of-function mechanism associated with accumulation of the expanded polyglutamine protein. We generated three strains of knockin (KI) mice carrying normal, expanded, or hyperexpanded CAG repeat tracts in the Cacna1a locus. The mice expressing hyperexpanded polyglutamine (Sca6(84Q)) developed progressive motor impairment and aggregation of mutant Ca(V)2.1 channels. Electrophysiological analysis of cerebellar Purkinje cells revealed similar Ca(2+) channel current density among the three KI models. Neither voltage sensitivity of activation nor inactivation was altered in the Sca6(84Q) neurons, suggesting that expanded CAG repeat per se does not affect the intrinsic electrophysiological properties of the channels. The pathogenesis of SCA6 is apparently linked to an age-dependent process accompanied by accumulation of mutant Ca(V)2.1 channels.


Asunto(s)
Envejecimiento/fisiología , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Proteínas Mutantes/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Ataxias Espinocerebelosas/fisiopatología , Empalme Alternativo/genética , Animales , Progresión de la Enfermedad , Electrofisiología , Exones/genética , Expresión Génica , Ratones , Ratones Transgénicos , Proteínas Mutantes/genética , Mutación/genética , Enfermedades del Sistema Nervioso/genética , Fenotipo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Transgenes/genética
15.
Proc Natl Acad Sci U S A ; 105(4): 1291-6, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18216249

RESUMEN

Polyglutamine diseases are inherited neurodegenerative disorders caused by expansion of CAG repeats encoding a glutamine tract in the disease-causing proteins. There are nine disorders, each having distinct features but also clinical and pathological similarities. In particular, spinocerebellar ataxia type 1 and 7 (SCA1 and SCA7) patients manifest cerebellar ataxia with degeneration of Purkinje cells. To determine whether the disorders share molecular pathogenic events, we studied two mouse models of SCA1 and SCA7 that express the glutamine-expanded protein from the respective endogenous loci. We found common transcriptional changes, with down-regulation of insulin-like growth factor binding protein 5 (Igfbp5) representing one of the most robust changes. Igfbp5 down-regulation occurred in granule neurons through a non-cell-autonomous mechanism and was concomitant with activation of the insulin-like growth factor (IGF) pathway and the type I IGF receptor on Purkinje cells. These data define one common pathogenic response in SCA1 and SCA7 and reveal the importance of intercellular mechanisms in their pathogenesis.


Asunto(s)
Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Transducción de Señal/genética , Somatomedinas/fisiología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Animales , Ataxina-1 , Ataxina-7 , Ataxinas , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación de la Expresión Génica/fisiología , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/antagonistas & inhibidores , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Transducción de Señal/fisiología , Somatomedinas/metabolismo , Ataxias Espinocerebelosas/etiología
16.
Dev Biol ; 311(1): 238-50, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17915208

RESUMEN

The Bergmann glia is a unipolar astrocyte in the cerebellar cortex, displaying a tight association with Purkinje cells. The cell bodies of Bergmann glia are located in a row around Purkinje cell somata; they extend radially arranged Bergmann fibers which enwrap the synapses on the Purkinje cell dendrites. It is well known that Bergmann glial somata migrate from the ventricular zone through the mantle zone, forming an epithelium-like lining in the Purkinje cell layer during development. However, the mechanism of the monolayer formation of Bergmann glia is poorly understood. Several reports have suggested that Notch signaling plays instructive roles in promoting the identities of several types of glial cells, including Bergmann glia. Moreover, Notch receptors are expressed in Bergmann glia during development. Here, we have deleted the Notch1, Notch2 and RBP-J genes in the Bergmann glia by GFAP-driven, Cre-mediated recombination, to study the role of Notch-RBP-J-signaling in the monolayer formation of Bergmann glia. Notch1/2- and RBP-J-conditional mutant mice showed disorganization of Bergmann fibers, irregularities of the Bergmann glial lining and aberrant localization of Bergmann glia in the molecular layer. Thus, Notch-RBP-J signaling plays crucial roles in the monolayer formation and morphogenesis of Bergmann glia.


Asunto(s)
Astrocitos/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Cerebelo/embriología , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Transducción de Señal , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Cerebelo/citología , Eliminación de Gen , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptor Notch1/genética , Receptor Notch2/genética
17.
PLoS Med ; 4(5): e182, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17535104

RESUMEN

BACKGROUND: Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disorder characterized by progressive motor and cognitive dysfunction. Caused by an expanded polyglutamine tract in ataxin 1 (ATXN1), SCA1 pathogenesis involves a multifactorial process that likely begins with misfolding of ATXN1, which has functional consequences on its interactions, leading to transcriptional dysregulation. Because lithium has been shown to exert neuroprotective effects in a variety of conditions, possibly by affecting gene expression, we tested the efficacy of lithium treatment in a knock-in mouse model of SCA1 (Sca1(154Q/2Q) mice) that replicates many features of the human disease. METHODS AND FINDINGS: Sca1(154Q/2Q) mice and their wild-type littermates were fed either regular chow or chow that contained 0.2% lithium carbonate. Dietary lithium carbonate supplementation resulted in improvement of motor coordination, learning, and memory in Sca1(154Q/2Q) mice. Importantly, motor improvement was seen when treatment was initiated both presymptomatically and after symptom onset. Neuropathologically, lithium treatment attenuated the reduction of dendritic branching in mutant hippocampal pyramidal neurons. We also report that lithium treatment restored the levels of isoprenylcysteine carboxyl methyltransferase (Icmt; alternatively, Pccmt), down-regulation of which is an early marker of mutant ATXN1 toxicity. CONCLUSIONS: The effect of lithium on a marker altered early in the course of SCA1 pathogenesis, coupled with its positive effect on multiple behavioral measures and hippocampal neuropathology in an authentic disease model, make it an excellent candidate treatment for human SCA1 patients.


Asunto(s)
Antimaníacos/farmacología , Carbonato de Litio/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Ataxias Espinocerebelosas/tratamiento farmacológico , Ataxias Espinocerebelosas/patología , Animales , Ataxina-1 , Ataxinas , Dendritas/enzimología , Dendritas/patología , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Actividad Motora/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Ataxias Espinocerebelosas/genética
18.
Nat Cell Biol ; 9(4): 402-14, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17384639

RESUMEN

Nuclear dysfunction is a key feature of the pathology of polyglutamine (polyQ) diseases. It has been suggested that mutant polyQ proteins impair functions of nuclear factors by interacting with them directly in the nucleus. However, a systematic analysis of quantitative changes in soluble nuclear proteins in neurons expressing mutant polyQ proteins has not been performed. Here, we perform a proteome analysis of soluble nuclear proteins prepared from neurons expressing huntingtin (Htt) or ataxin-1 (AT1) protein, and show that mutant AT1 and Htt similarly reduce the concentration of soluble high mobility group B1/2 (HMGB1/2) proteins. Immunoprecipitation and pulldown assays indicate that HMGBs interact with mutant AT1 and Htt. Immunohistochemistry showed that these proteins were reduced in the nuclear region outside of inclusion bodies in affected neurons. Compensatory expression of HMGBs ameliorated polyQ-induced pathology in primary neurons and in Drosophila polyQ models. Furthermore, HMGBs repressed genotoxic stress signals induced by mutant Htt or transcriptional repression. Thus, HMGBs may be critical regulators of polyQ disease pathology and could be targets for therapy development.


Asunto(s)
Proteína HMGB1/fisiología , Proteína HMGB2/fisiología , Enfermedades Neurodegenerativas/metabolismo , Proteínas Nucleares/fisiología , Proteómica/métodos , Animales , Western Blotting , Muerte Celular , Células Cultivadas , Drosophila , Electroforesis en Gel Bidimensional , Proteína HMGB1/análisis , Proteína HMGB1/metabolismo , Proteína HMGB2/análisis , Proteína HMGB2/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Modelos Biológicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/análisis , Proteínas Nucleares/metabolismo , Péptidos/genética , Péptidos/metabolismo , Unión Proteica , Células de Purkinje/citología , Células de Purkinje/metabolismo , ARN Interferente Pequeño , Ratas , Ratas Wistar , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
19.
J Neurosci ; 25(38): 8788-93, 2005 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-16177048

RESUMEN

Glutamate transporters are essential for terminating excitatory neurotransmission. Two distinct glutamate transporters, glutamate-aspartate transporter (GLAST) and excitatory amino acid transporter 4 (EAAT4), are expressed most abundantly in the molecular layer of the cerebellar cortex. GLAST is expressed in Bergmann glial processes surrounding excitatory synapses on Purkinje cell dendritic spines, whereas EAAT4 is concentrated on the extrasynaptic regions of Purkinje cell spine membranes. To clarify the functional significance of the coexistence of these transporters, we analyzed the kinetics of EPSCs in Purkinje cells of mice lacking either GLAST or EAAT4. There was no difference in the amplitude or the kinetics of the rising and initial decay phase of EPSCs evoked by stimulations of climbing fibers and parallel fibers between wild-type and EAAT4-deficient mice. However, long-lasting tail currents of the EPSCs appeared age dependently in most of Purkinje cells in EAAT4-deficient mice. These tail currents were never seen in mice lacking GLAST. In the GLAST-deficient mice, however, the application of cyclothiazide that reduces desensitization of AMPA receptors increased the peak amplitude of the EPSC and prolonged its decay more markedly than in both wild-type and EAAT4-deficient mice. The results indicate that these transporters play differential roles in the removal of synaptically released glutamate. GLAST contributes mainly to uptake of glutamate that floods out of the synaptic cleft at early times after transmitter release. In contrast, the main role of EAAT4 is to remove low concentrations of glutamate that escape from the uptake by glial transporters at late times and thus prevents the transmitter from spilling over to neighboring synapses.


Asunto(s)
Transportador 1 de Aminoácidos Excitadores/fisiología , Transportador 4 de Aminoácidos Excitadores/fisiología , Neuroglía/fisiología , Células de Purkinje/fisiología , Sinapsis/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología
20.
Brain Res ; 998(1): 122-9, 2004 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-14725975

RESUMEN

Neurotensin (NT) is a neuropeptide that induces a wide range of biological activities including hypothermia and analgesia. Such effects are mediated by the NT receptors Ntsr1, Ntsr2 and Ntsr3, although the involvement of each receptor in specific NT functions remains unknown. To address nociceptive function in vivo, we generated both Ntsr1-deficient and Ntsr2-deficient mice. In addition, histochemical analyses of both Ntsr1 and Ntsr2 mRNAs were performed in the mouse brain regions involved in NT-related nociception. The expression of Ntsr2 mRNA was greater than that of Ntsr1 in the periaqueductal gray (PAG) and the rostral ventral medulla (RVM). The mutant and control mice were subjected to the examination of thermal nociception, and in the hot plate test, a significant alteration in jump latency was observed in Ntsr2-deficient mice compared to Ntsr1-deficient or wild-type control mice. Latencies of tail flick and hind paw licking of the mutant mice were not affected compared to control mice. These results suggest that Ntsr2 has an important role in thermal nociception compared to Ntsr1, and that these mutant mice may represent a useful tool for the development of analgesic drugs.


Asunto(s)
Dolor/metabolismo , Receptores de Neurotensina/metabolismo , Animales , Conducta Animal , Clonación Molecular/métodos , Regulación de la Expresión Génica , Calor/efectos adversos , Hibridación in Situ/métodos , Bulbo Raquídeo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurotensina/metabolismo , Dolor/fisiopatología , Dimensión del Dolor , Umbral del Dolor/fisiología , Sustancia Gris Periacueductal/metabolismo , ARN Mensajero/biosíntesis , Tiempo de Reacción , Receptores de Neurotensina/deficiencia , Receptores de Neurotensina/genética , Receptores de Neurotensina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...