Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
2.
Addict Biol ; 23(1): 448-460, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28074533

RESUMEN

Netrin G1 is a presynaptic ligand involved in axonal projection. Although molecular mechanisms underlying cocaine addiction are still poorly understood, Netrin G1 might have a role as a regulator of anxiety, fear and spatial memory, behavioural traits impaired in the context of cocaine exposure. In this study, the Netrin G1 (Ntng1) expression was investigated in the nucleus accumbens of mice primarily conditioned to cocaine using a place preference paradigm. A genetic association study was then conducted on 146 multiplex families of the Collaborative study on Genetics of Alcoholism, in which seven single nucleotide polymorphisms located in the NTNG1 gene were genotyped. NTNG1 expression levels were also quantified in BA10, BA46 and the cerebellum of healthy controls (with no Axis 1 psychopathology). Decreased Ntng1 expression was initially observed in the nucleus accumbens of mice conditioned to cocaine. Significant genetic family-based associations were detected between NTNG1 polymorphisms and cocaine dependence. NTNG1 expression in BA10, BA46 and the cerebellum, however, were not significantly associated with any allele or haplotype of this gene. These results confirm that Ntng1 expression is disturbed in the nucleus accumbens of mice, after cocaine conditioning. A haplotype of NTNG1 was found to constitute a vulnerability factor for cocaine use disorder in patients, although none of its single nucleotide polymorphisms were associated with a differential expression pattern in healthy controls. The data suggest that change in the Ntng1 expression is a consequence of cocaine exposure, and that some of its genetic markers are associated with a greater risk for cocaine use disorder.


Asunto(s)
Trastornos Relacionados con Cocaína/genética , Cocaína , Condicionamiento Psicológico , Inhibidores de Captación de Dopamina , Netrinas/genética , Núcleo Accumbens/metabolismo , Adulto , Animales , Estudios de Casos y Controles , Cerebelo/metabolismo , Trastornos Relacionados con Cocaína/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Ratones , Netrinas/metabolismo , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo
3.
Hum Mol Genet ; 19(14): 2841-57, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20457675

RESUMEN

The SMARCA2 gene, which encodes BRM in the SWI/SNF chromatin-remodeling complex, was recently identified as being associated with schizophrenia (SZ) in a genome-wide approach. Polymorphisms in SMARCA2, associated with the disease, produce changes in the expression of the gene and/or in the encoded amino acid sequence. We show here that an SWI/SNF-centered network including the Smarca2 gene is modified by the down-regulation of REST/NRSF in a mouse neuronal cell line. REST/NRSF down-regulation also modifies the levels of Smarce1, Smarcd3 and SWI/SNF interactors (Hdac1, RcoR1 and Mecp2). Smarca2 down-regulation generates an abnormal dendritic spine morphology that is an intermediate phenotype of SZ. We further found that 8 (CSF2RA, HIST1H2BJ, NOTCH4, NRGN, SHOX, SMARCA2, TCF4 and ZNF804A) out of 10 genome-wide supported SZ-associated genes are part of an interacting network (including SMARCA2), 5 members of which encode transcription regulators. The expression of 3 (TCF4, SMARCA2 and CSF2RA) of the 10 genome-wide supported SZ-associated genes is modified when the REST/NRSF-SWI/SNF chromatin-remodeling complex is experimentally manipulated in mouse cell lines and in transgenic mouse models. The REST/NRSF-SWI/SNF deregulation also results in the differential expression of genes that are clustered in chromosomes suggesting the induction of genome-wide epigenetic changes. Finally, we found that SMARCA2 interactors and the genome-wide supported SZ-associated genes are considerably enriched in genes displaying positive selection in primates and in the human lineage which suggests the occurrence of novel protein interactions in primates. Altogether, these data identify the SWI/SNF chromatin-remodeling complex as a key component of the genetic architecture of SZ.


Asunto(s)
Redes Reguladoras de Genes/fisiología , Primates/genética , Proteínas Represoras/genética , Esquizofrenia/genética , Factores de Transcripción/genética , Animales , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , Evolución Molecular , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , Proteínas Represoras/metabolismo , Especificidad de la Especie
4.
Hum Genet ; 131(6): 803-22, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22350797

RESUMEN

Dopamine plays a key role in reward behavior, yet the association of drug dependence as a chronic, relapsing disorder with the genes encoding the various dopaminergic receptor subtypes remains difficult to delineate. In the context of subsequent genome-wide association (GWAS) research and post-GWAS investigations, we summarize the novel data that link genes encoding molecules involved in the dopaminergic system (dopamine receptors, transporter and enzymes in charge of its metabolism) to drug addiction. Recent reports indicate that the heritability of drug addiction should be high enough to allow a significant role for a specific set of genes, and the available genetic studies, which might not be already conclusive because of the heterogeneity of designs, methods and recruited samples, should support the idea of a significant role of at least one gene related to dopaminergic system. Evolutionary changes in primates and non-primate animals of genes coding for molecules involved in dopaminergic system highlight why addictive disorders are mainly limited to humans. Restricting the analyses to more specific intermediate phenotypes (or endophenotypes) such as attention allocation, stress reactivity, novelty seeking, behavioral disinhibition and impulsivity, instead of the broad addictive disorder concept can be instrumental to identify novel genes associated with these traits in the context of genome-wide studies.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Evolución Molecular , Predisposición Genética a la Enfermedad/genética , Variación Genética , Receptores Dopaminérgicos/genética , Trastornos Relacionados con Sustancias/genética , Secuencia de Aminoácidos , Animales , Atención , Estudio de Asociación del Genoma Completo/métodos , Humanos , Conducta Impulsiva , Metaanálisis como Asunto , Ratones , Datos de Secuencia Molecular
5.
Hum Mol Genet ; 18(8): 1405-14, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19218269

RESUMEN

The molecular mechanisms that lead to the cognitive defects characteristic of Down syndrome (DS), the most frequent cause of mental retardation, have remained elusive. Here we use a transgenic DS mouse model (152F7 line) to show that DYRK1A gene dosage imbalance deregulates chromosomal clusters of genes located near neuron-restrictive silencer factor (REST/NRSF) binding sites. We found that Dyrk1a binds the SWI/SNF complex known to interact with REST/NRSF. The mutation of a REST/NRSF binding site in the promoter of the REST/NRSF target gene L1cam modifies the transcriptional effect of Dyrk1a-dosage imbalance on L1cam. Dyrk1a dosage imbalance perturbs Rest/Nrsf levels with decreased Rest/Nrsf expression in embryonic neurons and increased expression in adult neurons. Using transcriptome analysis of embryonic brain subregions of transgenic 152F7 mouse line, we identified a coordinated deregulation of multiple genes that are responsible for dendritic growth impairment present in DS. Similarly, Dyrk1a overexpression in primary mouse cortical neurons induced severe reduction of the dendritic growth and dendritic complexity. We propose that DYRK1A overexpression-related neuronal gene deregulation via disturbance of REST/NRSF levels, and the REST/NRSF-SWI/SNF chromatin remodelling complex, significantly contributes to the neural phenotypic changes that characterize DS.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Síndrome de Down/genética , Síndrome de Down/fisiopatología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Dendritas/fisiología , Ratones , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Transfección , Quinasas DyrK
6.
Hum Mol Genet ; 17(16): 2541-51, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18492799

RESUMEN

Autism spectrum disorders (ASDs) are common, heritable, but genetically heterogeneous neurodevelopmental conditions. We recently defined a susceptibility locus for ASDs on chromosome 1q41-q42. High-resolution single-nucleotide polymorphisms (126 SNPs) genotyping across the chromosome 1q41-q42 region, followed by a MARK1 (microtubule affinity-regulating kinase 1)-tagged-SNP association study in 276 families with autism from the Autism Genetic Research Exchange, showed that several SNPs within the MARK1 gene were significantly associated with ASDs by transmission disequilibrium tests. Haplotype rs12740310*C-rs3737296*G-rs12410279*A was overtransmitted (P(corrected)= 0.0016), with a relative risk for autism of 1.8 in homozygous carriers. Furthermore, ASD-associated SNP rs12410279 modulates the level of transcription of MARK1. We found that MARK1 was overexpressed in the prefrontal cortex (BA46) but not in cerebellar granule cells, on postmortem brain tissues from patients. MARK1 displayed an accelerated evolution along the lineage leading to humans, suggesting possible involvement of this gene in cognition. MARK1 encodes a kinase-regulating microtubule-dependent transport in axons and dendrites. Both overexpression and silencing of MARK1 resulted in significantly shorter dendrite length in mouse neocortical neurons and modified dendritic transport speed. As expected for a gene encoding a key polarity determinant Par-1 protein kinase, MARK1 is involved in axon-dendrite specification. Thus, MARK1 overexpression in humans may be responsible for subtle changes in dendritic functioning.


Asunto(s)
Trastorno Autístico/enzimología , Predisposición Genética a la Enfermedad , Proteínas Serina-Treonina Quinasas/metabolismo , Adolescente , Adulto , Animales , Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Línea Celular Tumoral , Polaridad Celular , Corteza Cerebelosa/enzimología , Corteza Cerebelosa/fisiopatología , Niño , Preescolar , Mapeo Cromosómico , Cromosomas Humanos Par 1/genética , Dendritas/química , Dendritas/enzimología , Dendritas/fisiología , Evolución Molecular , Femenino , Expresión Génica , Haplotipos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas
7.
Behav Brain Res ; 359: 903-909, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29935919

RESUMEN

Autistic spectrum disorders (ASDs) are neurodevelopmental disorders for which genetic components have been well defined. However, specific gene deregulations related to synapse function in the autistic brain have not been as extensively described. Based on a candidate genes approach, we present in this study the expression data of 4 transcripts of interest (BDNF, CAMK2a, NR-CAM and RIMS1) located at the synapse in two regions of interest in the context of the ASDs; the lobule VI of cerebellum and the Brodmann area 46. We have also genotyped in our cohort the coding single nucleotide polymorphism rs6265, located in the BDNF gene. After correction for age and sex, whereas no change was observed in the lobule VI between controls and autistic patients, we found a significant increase of BDNF expression level in the BA46 from autistic patients. No significant interaction between the rs6265 genotype and autism was observed for the BDNF expression. However, "A" allele carriers are more likely to have increased BDNF levels. Finally, we found a significant positive correlation between BDNF and RIMS1 expression levels. Our data suggest that these two molecules which are involved in cell signalling at the synapse, might have coordinated expressions and, that BDNF regulation in the brain has to be investigated further in the context of ASDs.


Asunto(s)
Trastorno Autístico/patología , Factor Neurotrófico Derivado del Encéfalo/genética , Lóbulo Frontal/metabolismo , Regulación de la Expresión Génica/fisiología , ARN Mensajero/metabolismo , Adolescente , Adulto , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Niño , Preescolar , Diagnóstico , Femenino , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Genotipo , Humanos , Captura por Microdisección con Láser , Modelos Lineales , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Adulto Joven
8.
Neuroreport ; 19(7): 751-5, 2008 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-18418251

RESUMEN

Dysfunctions affecting the connections of basal ganglia lead to major neurological and psychiatric disorders. We investigated levels of mRNA for three neurexins (Nrxn) and three neuroligins (Nlgn) in the globus pallidus, subthalamic nucleus, and substantia nigra, in control conditions and after short-term exposure to cocaine. The expression of Nrxn2beta and Nlgn3 in the substantia nigra and Nlgn1 in the subthalamic nucleus depended on genetic background. The development of short-term cocaine appetence induced an increase in Nrxn3beta expression in the globus pallidus. Human NRXN3 has recently been linked to several addictions. Thus, NRXN3 adhesion molecules may play an important role in the synaptic plasticity of neurons involved in the indirect pathways of basal ganglia, in which they regulate reward-related learning.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Globo Pálido/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Animales , Moléculas de Adhesión Celular Neuronal , Cocaína/efectos adversos , Inhibidores de Captación de Dopamina/efectos adversos , Expresión Génica , Globo Pálido/efectos de los fármacos , Rayos Láser , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microdisección , Proteínas del Tejido Nervioso/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Moléculas de Adhesión de Célula Nerviosa/efectos de los fármacos , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
9.
Neuroreport ; 18(5): 441-6, 2007 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-17496800

RESUMEN

Neurite outgrowth involves various molecular mechanisms generating complex brain connections. These mechanisms have been linked to plasticity and learning and are thought to be deregulated in neuropsychiatric diseases. The transcription factor REST/NRSF regulates a subset of genes encoding neurite outgrowth molecules. We demonstrate here the downregulation of Rest/Nrsf expression in a mouse neuroblastoma cell line. This downregulation induced a clear increase in neurite length. Quantitative polymerase chain reaction showed deregulation of the candidate genes L1cam, Elmo2, Ulip1 and Ulip2. These genes are bona fide candidates known to be involved in dendrite and axonal outgrowth. This approach could be adapted to high-throughput techniques for determination of the mammalian neurite outgrowth gene repertoire.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Plasticidad Neuronal/fisiología , ARN Interferente Pequeño/farmacología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuritas/efectos de los fármacos , Neuroblastoma/patología , Plasticidad Neuronal/efectos de los fármacos , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo , Transfección/métodos
10.
Gene Expr Patterns ; 5(4): 561-8, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15749087

RESUMEN

Nogo protein has been identified as the component of central nervous system (CNS) myelin that limits axonal regeneration. We investigated the expression of the genes encoding Nogo and its receptor, NgR, between weeks eight and 23 of human embryonic development, by quantitative radioactive in situ hybridization. At 8 weeks, we detected NOGO and NgR transcripts in developing neuronal and non-neuronal structures. We focused on two different structures: the brain and the dental germs. During this period of development, NOGO and NgR transcripts colocalized in the cortical and ventricular zones of the brain, with expression strongest for these two genes in the postmitotic cells of the cortical plate. In developing dental germs, NgR was more strongly expressed than NOGO at 16 and 21 weeks. NOGO and NgR were expressed in zones of epithelium-mesenchyme interaction, which induce the differentiation of ameloblasts/odontoblasts. These genes were expressed most strongly in differentiated cells.


Asunto(s)
Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Mielina/genética , Receptores de Superficie Celular/genética , Desarrollo Embrionario , Desarrollo Fetal , Proteínas Ligadas a GPI , Humanos , Hibridación in Situ , Mandíbula/embriología , Proteínas Nogo , Receptor Nogo 1 , Odontogénesis , Transcripción Genética
11.
Gene Expr Patterns ; 5(5): 577-85, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15908283

RESUMEN

Neocortical neurons are generated predominantly from the cells that proliferate in the ventricular zone of the telencephalon. In order to understand the nature of these expanding cortical neuronal progenitor cells, we selected by differential display some transcripts that were enriched in the telencephalon as compared to the more caudal regions (diencephalon/mesencephalon). This systematic screening revealed one of the differentially expressed transcripts, namely the Fkbp25 mRNA that encodes a member of the FK506 binding proteins (FKBPs). Northern blot analysis showed that the expression of the single 1.4kb Fkbp25 transcript reached a maximum level on embryonic day 11.5 at the start of cortical neurogenesis in the mouse and was followed by a weak basal expression in the adult brain. In the embryo, Fkbp25 gene was strongly expressed in the telencephalon ventricular zone but also in areas active in myogenesis (walls of the ventricle and the atrium) and chondrogenesis (the cartilage of the rib and the hindlimb). An increase in the transcript levels of the Fkbp25 gene was also observed during the two successive proliferation waves of the cerebellum development. Immunostaining on primary cultures of embryonic day 10.5 telencephalon stem cells showed that the Fkbp25 protein was present in the cytoplasm and nuclei of cells cultured for 6h but exclusively in the nuclei of the Tuj-1 immunoreactive neurons obtained after 3 days of culture (The sequence data reported here have been submitted to GenBank under accession no. AF135595.).


Asunto(s)
Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Unión a Tacrolimus/biosíntesis , Proteínas de Unión a Tacrolimus/genética , Animales , Northern Blotting , Western Blotting , Encéfalo/metabolismo , Células COS , Núcleo Celular/metabolismo , Proliferación Celular , Clonación Molecular , Citoplasma/metabolismo , ADN Complementario/metabolismo , Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , Miocardio/metabolismo , Neuronas/metabolismo , Hibridación de Ácido Nucleico , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Proteínas de Unión a Tacrolimus/química , Telencéfalo/metabolismo , Factores de Tiempo , Transfección
12.
Prenat Diagn ; 27(10): 926-31, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17602442

RESUMEN

OBJECTIVE: To investigate the involvement of the genes encoding for COL6A1, COLA2 and super-oxide dismutase (SOD) in the mechanism for the retention of subcutaneous fluid in fetuses with trisomy 21. METHODS: During a 7-month period (November 2004-May 2005), human fetal skin from the nuchal region was obtained from euploid fetuses and from fetuses with trisomy 21 following abortions and terminations of pregnancy. Cell cultures were performed from nuchal skin. Quantification of COL6A1, COL6A2, COL6A3 and SOD mRNAs were performed using real-time quantitative RT-PCR. RESULTS: Twelve fetuses were studied between 13-15 and 19-20 weeks of gestation including 7 cases of trisomy 21. A significant overexpression of genes of interest was demonstrated in trisomy 21 fetuses when compared with euploid fetuses, in the first and in the second trimester of pregnancy (p < 0.0001). CONCLUSION: This study demonstrates a homogeneous overexpression of the genes encoding for alpha1 and alpha2 chains of Collagen type VI, and SOD in nuchal skin of human trisomy 21 fetuses. Persistence of this overexpression in the second trimester of pregnancy, despite the absence of an enlarged nuchal translucency (NT), may characterize some compensatory mechanisms.


Asunto(s)
Colágeno Tipo VI/genética , Síndrome de Down/genética , Regulación del Desarrollo de la Expresión Génica , Superóxido Dismutasa/genética , Síndrome de Down/diagnóstico por imagen , Femenino , Edad Gestacional , Humanos , Medida de Translucencia Nucal , Embarazo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/química
13.
Eur J Neurosci ; 20(3): 603-10, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15255972

RESUMEN

We carried out a screening of genes that are differentially expressed in normal mice and reeler mutants and are characterized by abnormal neuronal migration and neurite deployment due to defective Reelin signalling. A novel gene, provisionally named C61, was overexpressed in Reelin-deficient embryonic mouse brain RNA. C61 encodes a 3.7 kb mRNA that is brain specific and developmentally regulated, with predominant expression in differentiating neurons. The predicted protein is 664 amino acids long, and contains LAG1 and Ezrin/Radixin/Moesin-Myosin-Filament motifs, suggesting that it may function as an intracellular adaptor. From E14.5 to birth, C61 was highly expressed in all neuronal differentiation fields, with the highest signal in the telencephalic cortical plate and mitral cells in the olfactory bulb. When expressed as a GFP fusion protein in transfected non-neuronal cells and primary neurons, this protein localizes, respectively, to the nuclear membrane or axonal outgrowths, indicating a function in axonal traffic or signalling.


Asunto(s)
Encéfalo/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/fisiología , Proteínas de la Matriz Extracelular/fisiología , Regulación del Desarrollo de la Expresión Génica , Sinapsinas/metabolismo , Secuencias de Aminoácidos/fisiología , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Northern Blotting/métodos , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Caenorhabditis elegans , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular , Clonación Molecular , Drosophila , Embrión de Mamíferos , Embrión no Mamífero , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Proteínas Luminiscentes/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes Neurológicos , Proteínas de Microfilamentos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Neuronas/metabolismo , Especificidad de Órganos , ARN Mensajero/biosíntesis , Proteína Reelina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Serina Endopeptidasas , Transfección , Tubulina (Proteína)/metabolismo , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA