Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Genome Biol ; 25(1): 81, 2024 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-38553769

RESUMEN

The use of single-cell technologies for clinical applications requires disconnecting sampling from downstream processing steps. Early sample preservation can further increase robustness and reproducibility by avoiding artifacts introduced during specimen handling. We present FixNCut, a methodology for the reversible fixation of tissue followed by dissociation that overcomes current limitations. We applied FixNCut to human and mouse tissues to demonstrate the preservation of RNA integrity, sequencing library complexity, and cellular composition, while diminishing stress-related artifacts. Besides single-cell RNA sequencing, FixNCut is compatible with multiple single-cell and spatial technologies, making it a versatile tool for robust and flexible study designs.


Asunto(s)
Genómica , ARN , Humanos , Animales , Ratones , Fijación del Tejido/métodos , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN/métodos , ARN/genética , Genómica/métodos , Análisis de la Célula Individual/métodos
2.
Nucleic Acids Res ; 49(18): e105, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34255842

RESUMEN

Translation of eukaryotic mRNAs begins with binding of their m7G cap to eIF4E, followed by recruitment of other translation initiation factor proteins. We describe capCLIP, a novel method to comprehensively capture and quantify the eIF4E (eukaryotic initiation factor 4E) 'cap-ome' and apply it to examine the biological consequences of eIF4E-cap binding in distinct cellular contexts. First, we use capCLIP to identify the eIF4E cap-omes in human cells with/without the mTORC1 (mechanistic target of rapamycin, complex 1) inhibitor rapamycin, there being an emerging consensus that rapamycin inhibits translation of TOP (terminal oligopyrimidine) mRNAs by displacing eIF4E from their caps. capCLIP reveals that the representation of TOP mRNAs in the cap-ome is indeed systematically reduced by rapamycin, thus validating our new methodology. capCLIP also refines the requirements for a functional TOP sequence. Second, we apply capCLIP to probe the consequences of phosphorylation of eIF4E. We show eIF4E phosphorylation reduces overall eIF4E-mRNA association and, strikingly, causes preferential dissociation of mRNAs with short 5'-UTRs. capCLIP is a valuable new tool to probe the function of eIF4E and of other cap-binding proteins such as eIF4E2/eIF4E3.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Células HeLa , Humanos , Unión Proteica , Biosíntesis de Proteínas
3.
Cell Mol Life Sci ; 78(1): 249-270, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32170339

RESUMEN

eIF4E plays key roles in protein synthesis and tumorigenesis. It is phosphorylated by the kinases MNK1 and MNK2. Binding of MNKs to eIF4G enhances their ability to phosphorylate eIF4E. Here, we show that mTORC1, a key regulator of mRNA translation and oncogenesis, directly phosphorylates MNK2 on Ser74. This suppresses MNK2 activity and impairs binding of MNK2 to eIF4G. These effects provide a novel mechanism by which mTORC1 signaling impairs the function of MNK2 and thereby decreases eIF4E phosphorylation. MNK2[S74A] knock-in cells show enhanced phosphorylation of eIF4E and S6K1 (i.e., increased mTORC1 signaling), enlarged cell size, and increased invasive and transformative capacities. MNK2[Ser74] phosphorylation was inversely correlated with disease progression in human prostate tumors. MNK inhibition exerted anti-proliferative effects in prostate cancer cells in vitro. These findings define a novel feedback loop whereby mTORC1 represses MNK2 activity and oncogenic signaling through eIF4E phosphorylation, allowing reciprocal regulation of these two oncogenic pathways.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/antagonistas & inhibidores , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Morfolinas/farmacología , Mutagénesis Sitio-Dirigida , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/efectos de los fármacos , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
4.
J Neurochem ; 157(4): 1086-1101, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32892352

RESUMEN

The regulation of protein synthesis is a vital and finely tuned process in cellular physiology. In neurons, this process is very precisely regulated, as which mRNAs undergo translation is highly dependent on context. One of the most prominent regulators of protein synthesis is the enzyme eukaryotic elongation factor kinase 2 (eEF2K) that regulates the elongation stage of protein synthesis. This kinase and its substrate, eukaryotic elongation factor 2 (eEF2) are important in processes such as neuronal development and synaptic plasticity. eEF2K is regulated by multiple mechanisms including Ca2+ -ions and the mTORC1 signaling pathway, both of which play key roles in neurological processes such as learning and memory. In such settings, the localized control of protein synthesis is of crucial importance. In this work, we sought to investigate how the localization of eEF2K is controlled and the impact of this on protein synthesis in neuronal cells. In this study, we used both SH-SY5Y neuroblastoma cells and mouse cortical neurons, and pharmacologically and/or genetic approaches to modify eEF2K function. We show that eEF2K activity and localization can be regulated by its binding partner Homer1b/c, a scaffolding protein known for its participation in calcium-regulated signaling pathways. Furthermore, our results indicate that this interaction is regulated by the mTORC1 pathway, through a known phosphorylation site in eEF2K (S396), and that it affects rates of localized protein synthesis at synapses depending on the presence or absence of this scaffolding protein.


Asunto(s)
Quinasa del Factor 2 de Elongación/metabolismo , Proteínas de Andamiaje Homer/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neuronas/metabolismo , Biosíntesis de Proteínas/fisiología , Animales , Bicuculina/farmacología , Células Cultivadas , Antagonistas de Receptores de GABA-A/farmacología , Humanos , Ratones , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
Mol Metab ; 42: 101054, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32712434

RESUMEN

OBJECTIVES: Diet-driven obesity is increasingly widespread. Its consequences pose major challenges to human health and health care systems. There are MAP kinase-interacting kinases (MNKs) in mice, MNK1 and MNK2. Studies have demonstrated that mice lacking either MNK1 or MNK2 were partially protected against high-fat diet (HFD)-induced weight gain and insulin resistance. The aims of this study were to evaluate the phenotype of mice lacking both MNKs when given an HFD, to assess whether pharmacological inhibition of MNK function also protects against diet-induced obesity (DIO) and its consequences and to probe the mechanisms underlying such protection. METHODS: Male wild-type (WT) C57Bl6 mice or mice lacking both MNK1 and MNK2 (double knockout, DKO) were fed an HFD or control diet (CD) for up to 16 weeks. In a separate study, WT mice were also given an HFD for 6 weeks, after which half were treated with the recently-developed MNK inhibitor ETC-206 daily for 10 more weeks while continuing an HFD. Metabolites and other parameters were measured, and the expression of selected mRNAs and proteins was assessed. RESULTS: MNK-DKO mice were almost completely protected from HFD-induced obesity. Higher energy expenditure (EE) in MNK-DKO mice was observed, which probably reflects the changes in a number of genes or proteins linked to lipolysis, mitochondrial function/biogenesis, oxidative metabolism, and/or ATP consumption. The MNK inhibitor ETC-206 also prevented HFD-induced weight gain, confirming that the activity of the MNKs facilitates weight gain due to excessive caloric consumption. CONCLUSIONS: Disabling MNKs in mice, either genetically or pharmacologically, strongly prevents weight gain on a calorie-rich diet. This finding likely results from increased energy utilisation, involving greater ATP consumption, mitochondrial oxidative metabolism, and other processes.


Asunto(s)
Proteínas Serina-Treonina Quinasas/genética , Aumento de Peso/efectos de los fármacos , Animales , Respiración de la Célula , Dieta Alta en Grasa , Metabolismo Energético , Insulina/metabolismo , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Obesidad/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Aumento de Peso/genética , Aumento de Peso/fisiología
6.
Curr Biol ; 29(5): 737-749.e5, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30773367

RESUMEN

Maintaining accuracy during protein synthesis is crucial to avoid producing misfolded and/or non-functional proteins. The target of rapamycin complex 1 (TORC1) pathway and the activity of the protein synthesis machinery are known to negatively regulate lifespan in many organisms, although the precise mechanisms involved remain unclear. Mammalian TORC1 signaling accelerates the elongation stage of protein synthesis by inactivating eukaryotic elongation factor 2 kinase (eEF2K), which, when active, phosphorylates and inhibits eEF2, which mediates the movement of ribosomes along mRNAs, thereby slowing down the rate of elongation. We show that eEF2K enhances the accuracy of protein synthesis under a range of conditions and in several cell types. For example, our data reveal it links mammalian (m)TORC1 signaling to the accuracy of translation. Activation of eEF2K decreases misreading or termination readthrough errors during elongation, whereas knocking down or knocking out eEF2K increases their frequency. eEF2K also promotes the correct recognition of start codons in mRNAs. Reduced translational fidelity is known to correlate with shorter lifespan. Consistent with this, deletion of the eEF2K ortholog or other factors implicated in translation fidelity in Caenorhabditis elegans decreases lifespan, and eEF2K is required for lifespan extension induced by nutrient restriction. Our data uncover a novel mechanism linking nutrient supply, mTORC1 signaling, and the elongation stage of protein synthesis, which enhances the accuracy of protein synthesis. Our data also indicate that modulating translation elongation and its fidelity affects lifespan.


Asunto(s)
Caenorhabditis elegans/fisiología , Quinasa del Factor 2 de Elongación/genética , Longevidad/genética , Biosíntesis de Proteínas/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans , Factores de Transcripción E2F , Quinasa del Factor 2 de Elongación/metabolismo
7.
Expert Opin Ther Targets ; 23(3): 187-199, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30650992

RESUMEN

INTRODUCTION: The mitogen-activated protein kinase (MAPK)-interacting kinases (MNKs) are switched on by the oncogenic MAPK (ERK) signalling pathway. They phosphorylate eukaryotic initiation factor (eIF) 4E, a protein which recruits ribosomes to mRNAs and thereby mediates their translation. Importantly, overexpression of eIF4E can transform cells, and its function is controlled by a second oncogenic pathway, mechanistic target of rapamycin complex 1. Areas covered: We have evaluated the literature related to the role of the MNKs in human cancers, including their control by oncogenic signalling pathways; their expression and regulation in cancer cells and preclinical cancer models; and their roles in the proliferation, survival and migration/invasion of cancer cells. We also discuss progress towards generating specific and potent inhibitors of the MNKs and data obtained using such compounds. Expert opinion: The available data indicate that MNKs and/or eIF4E phosphorylation play a role in oncogenic transformation, the progression of at least some tumours and especially in processes related to tumour metastasis. MNKs are clearly druggable targets and, as they are not essential, significant 'side effects' of inhibiting the MNKs are likely to be limited. Further work is required to assess the efficacy of MNK inhibition in tackling tumour development, progression and metastasis.


Asunto(s)
Antineoplásicos/farmacología , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Animales , Factor 4E Eucariótico de Iniciación/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Trends Biochem Sci ; 41(10): 847-858, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27527252

RESUMEN

The eukaryotic translation initiation factor (eIF) 4E, which binds to the 5'-cap of mRNA, undergoes phosphorylation on a single conserved serine, executed by the mitogen-activated protein kinase (MAPK)-interacting kinases (MNKs). However, the functional consequences and physiological roles of MNK signalling have remained obscure. Now, new pharmacological and genetic tools have provided unprecedented insights into the function of MNKs and eIF4E phosphorylation. The studies suggest that MNKs control the translation of specific mRNAs in cancer metastasis and neuronal synaptic plasticity by a novel mechanism involving the regulation of the translational repressor, cytoplasmic fragile-X protein-interacting protein 1 (CYFIP1). These recent breakthroughs go a long way to resolving the longstanding enigma and controversy surrounding the function of the MNK-eIF4E axis in cancer cell biology and neurobiology.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Factor 4E Eucariótico de Iniciación/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Biosíntesis de Proteínas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Humanos , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Plasticidad Neuronal , Fosforilación , Unión Proteica , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , Transducción de Señal , Sinapsis/genética , Sinapsis/metabolismo
9.
EMBO J ; 33(18): 2040-56, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25069772

RESUMEN

The microRNAs of the miR-200 family maintain the central characteristics of epithelia and inhibit tumor cell motility and invasiveness. Using the Ago-HITS-CLIP technology for transcriptome-wide identification of direct microRNA targets in living cells, along with extensive validation to verify the reliability of the approach, we have identified hundreds of miR-200a and miR-200b targets, providing insights into general features of miRNA target site selection. Gene ontology analysis revealed a predominant effect of miR-200 targets in widespread coordinate control of actin cytoskeleton dynamics. Functional characterization of the miR-200 targets indicates that they constitute subnetworks that underlie the ability of cancer cells to migrate and invade, including coordinate effects on Rho-ROCK signaling, invadopodia formation, MMP activity, and focal adhesions. Thus, the miR-200 family maintains the central characteristics of the epithelial phenotype by acting on numerous targets at multiple levels, encompassing both cytoskeletal effectors that control actin filament organization and dynamics, and upstream signals that locally regulate the cytoskeleton to maintain cell morphology and prevent cell migration.


Asunto(s)
Movimiento Celular , Proliferación Celular , Células Epiteliales/fisiología , Regulación de la Expresión Génica , Redes Reguladoras de Genes , MicroARNs/genética , MicroARNs/metabolismo , Línea Celular , Citoesqueleto/metabolismo , Humanos
10.
Neuron ; 75(6): 1067-80, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22998874

RESUMEN

The paraneoplastic neurologic disorders target several families of neuron-specific RNA binding proteins (RNABPs), revealing that there are unique aspects of gene expression regulation in the mammalian brain. Here, we used HITS-CLIP to determine robust binding sites targeted by the neuronal Elav-like (nElavl) RNABPs. Surprisingly, nElav protein binds preferentially to GU-rich sequences in vivo and in vitro, with secondary binding to AU-rich sequences. nElavl null mice were used to validate the consequence of these binding events in the brain, demonstrating that they bind intronic sequences in a position dependent manner to regulate alternative splicing and to 3'UTR sequences to regulate mRNA levels. These controls converge on the glutamate synthesis pathway in neurons; nElavl proteins are required to maintain neurotransmitter glutamate levels, and the lack of nElavl leads to spontaneous epileptic seizure activity. The genome-wide analysis of nElavl targets reveals that one function of neuron-specific RNABPs is to control excitation-inhibition balance in the brain.


Asunto(s)
Encéfalo/citología , Proteínas ELAV/metabolismo , Regulación de la Expresión Génica/fisiología , Ácido Glutámico/metabolismo , Neuronas/fisiología , Empalme del ARN/genética , Regiones no Traducidas 3'/fisiología , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Biología Computacional , Modelos Animales de Enfermedad , Proteínas ELAV/deficiencia , Electroencefalografía , Epilepsia/genética , Epilepsia/fisiopatología , Regulación de la Expresión Génica/genética , Glutaminasa/genética , Glutaminasa/metabolismo , Ratones , Ratones Noqueados , Análisis por Micromatrices , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
11.
PLoS One ; 6(6): e21585, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21747913

RESUMEN

Anti-NeuN (Neuronal Nuclei) is a monoclonal antibody used extensively to specifically detect post-mitotic neurons. Anti-NeuN reactivity is predominantly nuclear; by western it detects multiple bands ranging in molecular weight from 45 kDa to >75 kDa. Expression screening putatively identified R3hdm2 as NeuN; however immunoprecipitation and mass spectrometry of the two major NeuN species at 45-50 kDa identified both as the RNA binding protein Rbfox3 (a member of the Fox family of alternative splicing factors), confirming and extending the identification of the 45 kDa band as Rbfox3 by Kim et al. Mapping of the anti-NeuN reactive epitopes in both R3hdm2 and Rbfox3 reveals a common proline- and glutamine-rich domain that lies at the N-terminus of the Rbfox3 protein. Our data suggests that alternative splicing of the Rbfox3 pre-mRNA itself leads to the production of four protein isoforms that migrate in the 45-50 kDa range, and that one of these splicing choices regulates Rbfox3/NeuN sub-cellular steady-state distribution, through the addition or removal of a short C-terminal extension containing the second half of a bipartite hydrophobic proline-tyrosine nuclear localization signal. Rbfox3 regulates alternative splicing of the Rbfox2 pre-mRNA, producing a message encoding a dominant negative form of the Rbfox2 protein. We show here that nuclear Rbfox3 isoforms can also enhance the inclusion of cryptic exons in the Rbfox2 mRNA, resulting in nonsense-mediated decay of the message, thereby contributing to the negative regulation of Rbfox2 by Rbfox3 through a novel mechanism.


Asunto(s)
Empalme Alternativo , Núcleo Celular/metabolismo , Codón sin Sentido/genética , Citoplasma/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de Unión al ADN , Mapeo Epitopo , Células HEK293 , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Factores de Empalme de ARN , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo
12.
PLoS One ; 6(6): e20773, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695151

RESUMEN

The ability of germ cells to carry out a gene regulatory program distinct from the surrounding somatic tissue, and their capacity to specify an entire new organism has made them a focus of many studies that seek to understand how specific regulatory mechanisms, particularly post-transcriptional mechanisms, contribute to cell fate. In zebrafish, germ cells are specified through the inheritance of cytoplasmic determinants, termed the germ plasm, which contains a number of maternal mRNAs and proteins. Investigation of several of these messages has revealed that the restricted localisation of these mRNAs to the germ plasm and subsequent germ cells is due to cis-acting sequence elements present in their 3'UTRs. Here we show that a member of the Hu family of RNA-binding proteins, HuB, is maternally provided in the zebrafish embryo and exhibits germ cell specific expression during embryogenesis. Restriction of HuB mRNA to the germ cells is dependent on a number of sequence elements in its 3'UTR, which act to degrade the mRNA in the soma and stabilise it in the germ cells. In addition, we show that the germ cell specific RNA-binding protein DAZL is able to promote HuB mRNA stability and translation in germ cells, and further demonstrate that these activities require a 30 nucleotide element in the 3'UTR. Our study suggests that DAZL specifically binds the HuB 3'UTR and protects the message from degradation and/or enhances HuB translation, leading to the germ cell specific expression of HuB protein.


Asunto(s)
Proteínas ELAV/genética , Células Germinativas/metabolismo , Estabilidad del ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Regiones no Traducidas 3'/genética , Animales , Proteínas ELAV/metabolismo , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Masculino , Especificidad de Órganos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Tiempo , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo
13.
Methods Mol Biol ; 488: 85-98, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18982285

RESUMEN

We present a newly developed method for fixing RNA-protein complexes in situ in living cells and the subsequent purification of the RNA targets. Using this approach, complex tissue such as mouse brain can be ultraviolet (UV) irradiated to covalently crosslink RNA-protein complexes. Once covalently bound, RNA-protein complexes can be purified under stringent conditions, allowing a highly specific purification scheme to be employed. After UV irradiation, the tissue is solubilized and the RNA partially digested, allowing a small fragment to remain attached to protein. RNA-protein complexes of interest are partially purified by immunoprecipitation and noncovalently associated RNA removed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). These purified RNA-protein complexes are isolated and treated with proteinase K, which removes protein but leaves intact RNA. This RNA is abundant enough, and competent for, RNA linker ligation, reverse transcriptase polymerase chain reaction (RT-PCR) amplification, and sequencing. Database matching of these short 70- to 100-nt RNA CLIP (crosslinking and immunoprecipitation of RNA-protein complexes) "tags," which mark the native binding sites of RNA binding proteins, potentially allows the entire target repertoire of an RNA binding protein to be determined.


Asunto(s)
Inmunoprecipitación/métodos , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/aislamiento & purificación , Animales , Reactivos de Enlaces Cruzados/química , Ratones , Unión Proteica/efectos de la radiación , Proteínas de Unión al ARN/metabolismo , Rayos Ultravioleta
14.
Science ; 302(5648): 1212-5, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-14615540

RESUMEN

Nova proteins are neuron-specific antigens targeted in paraneoplastic opsoclonus myoclonus ataxia (POMA), an autoimmune neurologic disease characterized by abnormal motor inhibition. Nova proteins regulate neuronal pre-messenger RNA splicing by directly binding to RNA. To identify Nova RNA targets, we developed a method to purify protein-RNA complexes from mouse brain with the use of ultraviolet cross-linking and immunoprecipitation (CLIP).Thirty-four transcripts were identified multiple times by Nova CLIP.Three-quarters of these encode proteins that function at the neuronal synapse, and one-third are involved in neuronal inhibition.Splicing targets confirmed in Nova-/- mice include c-Jun N-terminal kinase 2, neogenin, and gephyrin; the latter encodes a protein that clusters inhibitory gamma-aminobutyric acid and glycine receptors, two previously identified Nova splicing targets.Thus, CLIP reveals that Nova coordinately regulates a biologically coherent set of RNAs encoding multiple components of the inhibitory synapse, an observation that may relate to the cause of abnormal motor inhibition in POMA.


Asunto(s)
Antígenos de Neoplasias , Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas 3' , Empalme Alternativo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Exones , Intrones , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteína Quinasa 9 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibición Neural , Antígeno Ventral Neuro-Oncológico , Pruebas de Precipitina , Precursores del ARN/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA