Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 39(39): 7759-7777, 2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31405929

RESUMEN

The etiology of the autoimmune disorder systemic lupus erythematosus (SLE) remains poorly understood. In neuropsychiatric SLE (NPSLE), autoimmune responses against neural self-antigens find expression in neurological and cognitive alterations. SLE autoantibodies often target nucleic acids, including RNAs and specifically RNA domains with higher-order structural content. We report that autoantibodies directed against neuronal regulatory brain cytoplasmic (BC) RNAs were generated in a subset of SLE patients. By contrast, anti-BC RNA autoantibodies (anti-BC abs) were not detected in sera from patients with autoimmune diseases other than SLE (e.g., rheumatoid arthritis or multiple sclerosis) or in sera from healthy subjects with no evidence of disease. SLE anti-BC abs belong to the IgG class of immunoglobulins and target both primate BC200 RNA and rodent BC1 RNA. They are specifically directed at architectural motifs in BC RNA 5' stem-loop domains that serve as dendritic targeting elements (DTEs). SLE anti-BC abs effectively compete with RNA transport factor heterogeneous nuclear ribonucleoprotein A2 (hnRNP A2) for DTE access and significantly diminish BC RNA delivery to synapto-dendritic sites of function. In vivo experiments with male BALB/c mice indicate that, upon lipopolysaccharide-induced opening of the blood-brain barrier, SLE anti-BC abs are taken up by CNS neurons where they significantly impede localization of endogenous BC1 RNA to synapto-dendritic domains. Lack of BC1 RNA causes phenotypic abnormalities including epileptogenic responses and cognitive dysfunction. The combined data indicate a role for anti-BC RNA autoimmunity in SLE and its neuropsychiatric manifestations.SIGNIFICANCE STATEMENT Although clinical manifestations of neuropsychiatric lupus are well recognized, the underlying molecular-cellular alterations have been difficult to determine. We report that sera of a subset of lupus patients contain autoantibodies directed at regulatory brain cytoplasmic (BC) RNAs. These antibodies, which we call anti-BC abs, target the BC RNA 5' domain noncanonical motif structures that specify dendritic delivery. Lupus anti-BC abs effectively compete with RNA transport factor heterogeneous nuclear ribonucleoprotein A2 (hnRNP A2) for access to BC RNAs. As a result, hnRNP A2 is displaced, and BC RNAs are impaired in their ability to reach synapto-dendritic sites of function. The results reveal an unexpected link between BC RNA autoantibody recognition and dendritic RNA targeting. Cellular RNA dysregulation may thus be a contributing factor in the pathogenesis of neuropsychiatric lupus.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Lupus Eritematoso Sistémico/inmunología , Neuronas/metabolismo , ARN Citoplasmático Pequeño/inmunología , ARN Citoplasmático Pequeño/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Femenino , Humanos , Lupus Eritematoso Sistémico/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Transporte de ARN/fisiología
2.
eNeuro ; 5(2)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29766042

RESUMEN

Fragile X premutation disorder is caused by CGG triplet repeat expansions in the 5' untranslated region of FMR1 mRNA. The question of how expanded CGG repeats cause disease is a subject of continuing debate. Our work indicates that CGG-repeat structures compete with regulatory BC1 RNA for access to RNA transport factor hnRNP A2. As a result, BC1 RNA is mislocalized in vivo, as its synapto-dendritic presence is severely diminished in brains of CGG-repeat knock-in animals (a premutation mouse model). Lack of BC1 RNA is known to cause seizure activity and cognitive dysfunction. Our working hypothesis thus predicted that absence, or significantly reduced presence, of BC1 RNA in synapto-dendritic domains of premutation animal neurons would engender cognate phenotypic alterations. Testing this prediction, we established epileptogenic susceptibility and cognitive impairments as major phenotypic abnormalities of CGG premutation mice. In CA3 hippocampal neurons of such animals, synaptic release of glutamate elicits neuronal hyperexcitability in the form of group I metabotropic glutamate receptor-dependent prolonged epileptiform discharges. CGG-repeat knock-in animals are susceptible to sound-induced seizures and are cognitively impaired as revealed in the Attentional Set Shift Task. These phenotypic disturbances occur in young-adult premutation animals, indicating that a neurodevelopmental deficit is an early-initial manifestation of the disorder. The data are consistent with the notion that RNA mislocalization can contribute to pathogenesis.


Asunto(s)
Disfunción Cognitiva/genética , Síndrome del Cromosoma X Frágil/genética , Transporte de ARN/genética , ARN Citoplasmático Pequeño/genética , Secuencias Reguladoras de Ácido Ribonucleico/genética , Convulsiones/genética , Expansión de Repetición de Trinucleótido/genética , Factores de Edad , Animales , Región CA3 Hipocampal/fisiopatología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/fisiopatología , Modelos Animales de Enfermedad , Síndrome del Cromosoma X Frágil/complicaciones , Síndrome del Cromosoma X Frágil/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Convulsiones/etiología , Convulsiones/fisiopatología
3.
Learn Mem ; 24(7): 267-277, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28620074

RESUMEN

Dendritic regulatory BC1 RNA is a non-protein-coding (npc) RNA that operates in the translational control of gene expression. The absence of BC1 RNA in BC1 knockout (KO) animals causes translational dysregulation that entails neuronal phenotypic alterations including prolonged epileptiform discharges, audiogenic seizure activity in vivo, and excessive cortical oscillations in the γ frequency band. Here we asked whether BC1 RNA control is also required for higher brain functions such as learning, memory, or cognition. To address this question, we used odor/object attentional set shifting tasks in which prefrontal cortical performance was assessed in a series of discrimination and conflict learning sessions. Results obtained in these behavioral trials indicate that BC1 KO animals were significantly impaired in their cognitive flexibility. When faced with conflicting information sources, BC1 KO animals committed regressive errors as they were compromised in their ability to disengage from recently acquired memories even though recall of such memories was in conflict with new situational context. The observed cognitive deficits are reminiscent of those previously described in subtypes of human autism spectrum disorders.


Asunto(s)
Atención/fisiología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Odorantes , ARN Citoplasmático Pequeño/metabolismo , Animales , Conflicto Psicológico , Aprendizaje Discriminativo/fisiología , Aseo Animal/fisiología , Curva de Aprendizaje , Aprendizaje por Laberinto , Recuerdo Mental/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Citoplasmático Pequeño/genética
4.
J Cell Biol ; 207(2): 237-52, 2014 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-25332164

RESUMEN

In neurons, translational regulation of gene expression has been implicated in the activity-dependent management of synapto-dendritic protein repertoires. However, the fundamentals of stimulus-modulated translational control in neurons remain poorly understood. Here we describe a mechanism in which regulatory brain cytoplasmic (BC) RNAs cooperate with eukaryotic initiation factor 4B (eIF4B) to control translation in a manner that is responsive to neuronal activity. eIF4B is required for the translation of mRNAs with structured 5' untranslated regions (UTRs), exemplified here by neuronal protein kinase Mζ (PKMζ) mRNA. Upon neuronal stimulation, synapto-dendritic eIF4B is dephosphorylated at serine 406 in a rapid process that is mediated by protein phosphatase 2A. Such dephosphorylation causes a significant decrease in the binding affinity between eIF4B and BC RNA translational repressors, enabling the factor to engage the 40S small ribosomal subunit for translation initiation. BC RNA translational control, mediated via eIF4B phosphorylation status, couples neuronal activity to translational output, and thus provides a mechanistic basis for long-term plastic changes in nerve cells.


Asunto(s)
Factores Eucarióticos de Iniciación/fisiología , Neuronas/metabolismo , ARN Mensajero/metabolismo , ARN Citoplasmático Pequeño/fisiología , Regiones no Traducidas 5' , Animales , Línea Celular , Factores Eucarióticos de Iniciación/metabolismo , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Modelos Genéticos , Neuronas/citología , Neuronas/ultraestructura , Fosforilación , Biosíntesis de Proteínas , ARN Citoplasmático Pequeño/metabolismo , Ratas Sprague-Dawley , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/fisiología , Células Sf9 , Transducción de Señal
5.
Elife ; 2: e00178, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23359859

RESUMEN

The neuronal RNA binding protein NOVA regulates splicing, shuttles to the cytoplasm, and co-localizes with target transcripts in dendrites, suggesting links between splicing and local translation. Here we identified >200 transcripts showing NOVA-dependent changes in abundance, but, surprisingly, HITS-CLIP revealed NOVA binds these RNAs in introns rather than 3' UTRs. This led us to discover NOVA-regulated splicing of cryptic exons within these introns. These exons triggered nonsense mediated decay (NMD), as UPF1 and protein synthesis were required for NOVA's effect on RNA levels. Their regulation was dynamic and physiologically relevant. The NMD exons were regulated by seizures, which also induced changes in Nova subcellular localization and mediated large changes in synaptic proteins, including proteins implicated in familial epilepsy. Moreover, Nova haploinsufficient mice had spontaneous epilepsy. The data reveal a hidden means of dynamic RNA regulation linking electrical activity to splicing and protein output, and of mediating homeostatic excitation/inhibition balance in neurons.DOI:http://dx.doi.org/10.7554/eLife.00178.001.


Asunto(s)
Antígenos de Neoplasias/fisiología , Exones , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/fisiología , Convulsiones/metabolismo , Sinapsis/metabolismo , Regiones no Traducidas 3' , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Encéfalo/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Antígeno Ventral Neuro-Oncológico , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Fracciones Subcelulares
6.
Mol Cell Biol ; 31(22): 4538-49, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21930783

RESUMEN

In higher eukaryotes, increasing evidence suggests, gene expression is to a large degree controlled by RNA. Regulatory RNAs have been implicated in the management of neuronal function and plasticity in mammalian brains. However, much of the molecular-mechanistic framework that enables neuronal regulatory RNAs to control gene expression remains poorly understood. Here, we establish molecular mechanisms that underlie the regulatory capacity of neuronal BC RNAs in the translational control of gene expression. We report that regulatory BC RNAs employ a two-pronged approach in translational control. One of two distinct repression mechanisms is mediated by C-loop motifs in BC RNA 3' stem-loop domains. These C-loops bind to eIF4B and prevent the factor's interaction with 18S rRNA of the small ribosomal subunit. In the second mechanism, the central A-rich domains of BC RNAs target eIF4A, specifically inhibiting its RNA helicase activity. Thus, BC RNAs repress translation initiation in a bimodal mechanistic approach. As BC RNA functionality has evolved independently in rodent and primate lineages, our data suggest that BC RNA translational control was necessitated and implemented during mammalian phylogenetic development of complex neural systems.


Asunto(s)
Factor 4A Eucariótico de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Biosíntesis de Proteínas , ARN Citoplasmático Pequeño/metabolismo , ARN/metabolismo , Animales , Secuencia de Bases , Regulación de la Expresión Génica , Secuencias Invertidas Repetidas , Ratones , Neuronas/metabolismo , Conformación de Ácido Nucleico , ARN/genética , ARN Ribosómico 18S/genética , ARN Ribosómico 18S/metabolismo , ARN Citoplasmático Pequeño/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Subunidades Ribosómicas Pequeñas/metabolismo , Análisis de Secuencia de ARN
7.
Science ; 329(5990): 439-43, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20558669

RESUMEN

The control of RNA alternative splicing is critical for generating biological diversity. Despite emerging genome-wide technologies to study RNA complexity, reliable and comprehensive RNA-regulatory networks have not been defined. Here, we used Bayesian networks to probabilistically model diverse data sets and predict the target networks of specific regulators. We applied this strategy to identify approximately 700 alternative splicing events directly regulated by the neuron-specific factor Nova in the mouse brain, integrating RNA-binding data, splicing microarray data, Nova-binding motifs, and evolutionary signatures. The resulting integrative network revealed combinatorial regulation by Nova and the neuronal splicing factor Fox, interplay between phosphorylation and splicing, and potential links to neurologic disease. Thus, we have developed a general approach to understanding mammalian RNA regulation at the systems level.


Asunto(s)
Empalme Alternativo , Antígenos de Neoplasias/metabolismo , Encéfalo/metabolismo , Redes Reguladoras de Genes , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Inteligencia Artificial , Teorema de Bayes , Sitios de Unión , Línea Celular , Biología Computacional , Evolución Molecular , Exones , Humanos , Intrones , Ratones , Modelos Genéticos , Modelos Estadísticos , Enfermedades del Sistema Nervioso/genética , Antígeno Ventral Neuro-Oncológico , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , ARN/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-20407637

RESUMEN

Nova proteins are neuron-specific RNA binding proteins targeted by autoantibodies in a disorder manifest by failure of motor inhibition, and they regulate splicing and alternative 3' processing. Nova regulates splicing of RNAs encoding synaptic proteins, including the inhibitory glycine receptor alpha2 subunit (GlyRalpha2), and binds to others, including the GIRK2 channel. We found that Nova harbors functional NES and NLS elements, shuttles between the nucleus and cytoplasm, and that 50% of the protein localizes to the soma-dendritic compartment. Immunofluoresence and EM analysis of spinal cord motor neurons demonstrated that Nova co-localizes beneath synaptic contacts in dendrites with the same RNA, GlyRalpha2, whose splicing it regulates in the nucleus. HITS-CLIP identified intronic and 3' UTR sites where Nova binds to GlyRalpha2 and GIRK2 transcripts in the brain. This led directly to the identification of a 3' UTR localization element that mediates Nova-dependent localization of GIRK2 in primary neurons. These data demonstrate that HITS-CLIP can identify functional RNA localization elements, and they suggest new links between the regulation of nuclear RNA processing and mRNA localization.

9.
J Neurosci ; 23(32): 10433-44, 2003 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-14614102

RESUMEN

The dendritic transport and local translation of mRNA may be an essential mechanism to regulate synaptic growth and plasticity. We investigated the molecular mechanism and function of beta-actin mRNA localization in dendrites of cultured hippocampal neurons. Previous studies have shown that beta-actin mRNA localization to the leading edge of fibroblasts or the growth cones of developing neurites involved a specific interaction between a zipcode sequence in the 3' untranslated region and the mRNA-binding protein zipcode-binding protein-1 (ZBP1). Here, we show that ZBP1 is required for the localization of beta-actin mRNA to dendrites. Knock-down of ZBP1 using morpholino antisense oligonucleotides reduced dendritic levels of ZBP1 and beta-actin mRNA and impaired growth of dendritic filopodia in response to BDNF treatment. Transfection of an enhanced green fluorescent protein (EGFP)-beta-actin construct, which contained the zipcode, increased the density of dendritic filopodia and filopodial synapses. Transfection of an EGFP construct, also with the zipcode, resulted in recruitment of endogenous ZBP1 and beta-actin mRNA into dendrites and similarly increased the density of dendritic filopodia. However, the beta-actin zipcode did not affect filopodial length or the density of mature spines. These results reveal a novel function for an mRNA localization element and its binding protein in the regulation of dendritic morphology and synaptic growth via dendritic filopodia.


Asunto(s)
Actinas/genética , Dendritas/metabolismo , Seudópodos/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Sinapsis/metabolismo , Animales , Encéfalo/citología , Encéfalo/embriología , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Técnicas de Cocultivo , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Sustancias Macromoleculares , Neuronas/metabolismo , Neuronas/ultraestructura , Oligonucleótidos Antisentido/farmacología , Seudópodos/efectos de los fármacos , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...