Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Nucleic Acids Res ; 48(20): 11270-11283, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33068416

RESUMO

Specific elements of viral genomes regulate interactions within host cells. Here, we calculated the secondary structure content of >2000 coronaviruses and computed >100 000 human protein interactions with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The genomic regions display different degrees of conservation. SARS-CoV-2 domain encompassing nucleotides 22 500-23 000 is conserved both at the sequence and structural level. The regions upstream and downstream, however, vary significantly. This part of the viral sequence codes for the Spike S protein that interacts with the human receptor angiotensin-converting enzyme 2 (ACE2). Thus, variability of Spike S is connected to different levels of viral entry in human cells within the population. Our predictions indicate that the 5' end of SARS-CoV-2 is highly structured and interacts with several human proteins. The binding proteins are involved in viral RNA processing, include double-stranded RNA specific editases and ATP-dependent RNA-helicases and have strong propensity to form stress granules and phase-separated assemblies. We propose that these proteins, also implicated in viral infections such as HIV, are selectively recruited by SARS-CoV-2 genome to alter transcriptional and post-transcriptional regulation of host cells and to promote viral replication.


Assuntos
Genoma Viral , Mapas de Interação de Proteínas , SARS-CoV-2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/virologia , Humanos , Ligação Proteica , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Virulência/genética , Internalização do Vírus , Replicação Viral
3.
Methods ; 178: 11-18, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31563541

RESUMO

Given their central role in translation, splicing, localization and stability of transcripts, RNA binding proteins (RBPs) are key regulators of several cellular processes. While experimental efforts have been put to study how RBPs bind to transcripts, very little is known about the RNA contributions to the interaction. Here, we review the most common RNA-centric methods to reveal interactions with RBPs: both in vitro (SELEX, SEQR, RNA-compete and RBNS) and in silico (MEME, SeAMotE, GLAM2, iDeep, MEMERIS, RNA context, RCK, RNApromo and GraphProt). We emphasize the main advantages and disadvantages of each technique and highlight the key physico-chemical features contributing to the identification of RNA motifs involved in RBP recognition. We discuss extrinsic determinants influencing protein-RNA binding, such as post-transcriptional and post-translational modifications as well as expression and location of transcripts.


Assuntos
Conformação de Ácido Nucleico , Proteínas de Ligação a RNA/isolamento & purificação , RNA/isolamento & purificação , Sítios de Ligação/genética , Simulação por Computador , Motivos de Nucleotídeos/genética , Ligação Proteica/genética , RNA/genética , Proteínas de Ligação a RNA/genética
4.
Nucleic Acids Res ; 46(22): 12067-12086, 2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30260431

RESUMO

Syndesmos (SDOS) is a functionally poorly characterized protein that directly interacts with p53 binding protein 1 (53BP1) and regulates its recruitment to chromatin. We show here that SDOS interacts with another important cancer-linked protein, the chaperone TRAP1, associates with actively translating polyribosomes and represses translation. Moreover, we demonstrate that SDOS directly binds RNA in living cells. Combining individual gene expression profiling, nucleotide crosslinking and immunoprecipitation (iCLIP), and ribosome profiling, we discover several crucial pathways regulated post-transcriptionally by SDOS. Among them, we identify a small subset of mRNAs responsible for the biogenesis of primary cilium that have been linked to developmental and degenerative diseases, known as ciliopathies, and cancer. We discover that SDOS binds and regulates the translation of several of these mRNAs, controlling cilia development.


Assuntos
Cílios/genética , Proteínas de Ligação a RNA/fisiologia , Cílios/metabolismo , Ciliopatias/genética , Células HCT116 , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Humanos , Neoplasias/genética , Polirribossomos/metabolismo , Ligação Proteica/genética , Biossíntese de Proteínas/genética , Domínios e Motivos de Interação entre Proteínas/genética , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética
5.
Nucleic Acids Res ; 45(22): 12888-12903, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29149290

RESUMO

Recent evidence indicates a link between Parkinson's Disease (PD) and the expression of a-synuclein (SNCA) isoforms with different 3' untranslated regions (3'UTRs). Yet, the post-transcriptional mechanisms regulating SNCA expression are unknown. Using a large-scale in vitro /in silico screening we identified RNA-binding proteins (RBPs) that interact with SNCA 3' UTRs. We identified two RBPs, ELAVL1 and TIAR, that bind with high affinity to the most abundant and translationally active 3' UTR isoform (575 nt). Knockdown and overexpression experiments indicate that both ELAVL1 and TIAR positively regulate endogenous SNCA in vivo. The mechanism of regulation implies mRNA stabilization as well as enhancement of translation in the case of TIAR. We observed significant alteration of both TIAR and ELAVL1 expression in motor cortex of post-mortem brain donors and primary cultured fibroblast from patients affected by PD and Multiple System Atrophy (MSA). Moreover, trans expression quantitative trait loci (trans-eQTLs) analysis revealed that a group of single nucleotide polymorphisms (SNPs) in TIAR genomic locus influences SNCA expression in two different brain areas, nucleus accumbens and hippocampus. Our study sheds light on the 3' UTR-mediated regulation of SNCA and its link with PD pathogenesis, thus opening up new avenues for investigation of post-transcriptional mechanisms in neurodegeneration.


Assuntos
Regiões 3' não Traduzidas/genética , Regulação da Expressão Gênica , Doença de Parkinson/genética , alfa-Sinucleína/genética , Linhagem Celular Tumoral , Células Cultivadas , Proteína Semelhante a ELAV 1/genética , Proteína Semelhante a ELAV 1/metabolismo , Células HeLa , Hipocampo/metabolismo , Humanos , Núcleo Accumbens/metabolismo , Doença de Parkinson/metabolismo , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , alfa-Sinucleína/metabolismo
6.
RNA Biol ; 13(4): 466-72, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26853560

RESUMO

RBM10 is an RNA binding protein and alternative splicing regulator frequently mutated in lung adenocarcinomas. Recent results indicate that RBM10 inhibits proliferation of lung cancer cells by promoting skipping of exon 9 of the gene NUMB, a frequent alternative splicing change in lung cancer generating a negative regulator of Notch signaling. Complementing these observations, we show that knock down of RBM10 in human cancer cells enhances growth of mouse tumor xenografts, confirming that RBM10 acts as a tumor suppressor, while knock down of an oncogenic mutant version of RBM10 reduces xenograft tumor growth. A RBM10 mutation found in lung cancer cells, V354E, disrupts RBM10-mediated regulation of NUMB alternative splicing, inducing the cell proliferation-promoting isoform. We now show that 2 natural RBM10 isoforms that differ by the presence or absence of V354 in the second RNA Recognition Motif (RRM2), display similar regulatory effects on NUMB alternative splicing, suggesting that V354E actively disrupts RBM10 activity. Structural modeling localizes V354 in the outside surface of one α-helix opposite to the RNA binding surface of RBM10, and we show that the mutation does not compromise binding of the RRM2 domain to NUMB RNA regulatory sequences. We further show that other RBM10 mutations found in lung adenocarcinomas also compromise regulation of NUMB exon 9. Collectively, our previous and current results reveal that RBM10 is a tumor suppressor that represses Notch signaling and cell proliferation through the regulation of NUMB alternative splicing.


Assuntos
Processamento Alternativo , Genes Supressores de Tumor , Proteínas de Ligação a RNA/fisiologia , Animais , Células HEK293 , Células HeLa , Xenoenxertos , Humanos , Camundongos , Mutação
7.
Nucleic Acids Res ; 43(17): 8540-50, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26250109

RESUMO

Fragile X syndrome (FXS), the most common form of inherited intellectual disability, is caused by the silencing of the FMR1 gene encoding an RNA-binding protein (FMRP) mainly involved in translational control. We characterized the interaction between FMRP and the mRNA of GRK4, a member of the guanine nucleotide-binding protein (G protein)-coupled receptor kinase super-family, both in vitro and in vivo. While the mRNA level of GRK4 is unchanged in the absence or in the presence of FMRP in different regions of the brain, GRK4 protein level is increased in Fmr1-null cerebellum, suggesting that FMRP negatively modulates the expression of GRK4 at the translational level in this brain region. The C-terminal region of FMRP interacts with a domain of GRK4 mRNA, that we called G4RIF, that is folded in four stem loops. The SL1 stem loop of G4RIF is protected by FMRP and is part of the S1/S2 sub-domain that directs translation repression of a reporter mRNA by FMRP. These data confirm the role of the G4RIF/FMRP complex in translational regulation. Considering the role of GRK4 in GABAB receptors desensitization, our results suggest that an increased GRK4 levels in FXS might contribute to cerebellum-dependent phenotypes through a deregulated desensitization of GABAB receptors.


Assuntos
Cerebelo/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Quinase 4 de Receptor Acoplado a Proteína G/genética , RNA Mensageiro/metabolismo , Animais , Sítios de Ligação , Proteína do X Frágil da Deficiência Intelectual/química , Proteína do X Frágil da Deficiência Intelectual/genética , Quinase 4 de Receptor Acoplado a Proteína G/metabolismo , Humanos , Camundongos , Camundongos Knockout , Ligação Proteica , Biossíntese de Proteínas , Domínios e Motivos de Interação entre Proteínas , RNA Mensageiro/química
8.
Proc Natl Acad Sci U S A ; 111(44): 15622-9, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25313066

RESUMO

The roles of Argonaute proteins in cytoplasmic microRNA and RNAi pathways are well established. However, their implication in small RNA-mediated transcriptional gene silencing in the mammalian cell nucleus is less understood. We have recently shown that intronic siRNAs cause chromatin modifications that inhibit RNA polymerase II elongation and modulate alternative splicing in an Argonaute-1 (AGO1)-dependent manner. Here we used chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) to investigate the genome-wide distribution of AGO1 nuclear targets. Unexpectedly, we found that about 80% of AGO1 clusters are associated with cell-type-specific transcriptional enhancers, most of them (73%) overlapping active enhancers. This association seems to be mediated by long, rather than short, enhancer RNAs and to be more prominent in intragenic, rather than intergenic, enhancers. Paradoxically, crossing ChIP-seq with RNA-seq data upon AGO1 depletion revealed that enhancer-bound AGO1 is not linked to the global regulation of gene transcription but to the control of constitutive and alternative splicing, which was confirmed by an individual gene analysis explaining how AGO1 controls inclusion levels of the cassette exon 107 in the SYNE2 gene.


Assuntos
Processamento Alternativo/fisiologia , Proteínas Argonautas/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Fatores de Iniciação em Eucariotos/metabolismo , Regulação da Expressão Gênica/fisiologia , RNA/metabolismo , Transcrição Gênica/fisiologia , Proteínas Argonautas/genética , Linhagem Celular , Fatores de Iniciação em Eucariotos/genética , Humanos , RNA/genética , Análise de Sequência de RNA
9.
Cell Rep ; 7(4): 1211-26, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24813895

RESUMO

The Ewing sarcoma protein EWS is an RNA and DNA binding protein implicated in transcription, pre-mRNA splicing, and DNA damage response. Using CLIP-seq, we identified EWS RNA binding sites in exonic regions near 5' splice sites. A prominent target was exon 6 of the FAS/CD95 receptor, which is alternatively spliced to generate isoforms with opposing activities in programmed cell death. Depletion and overexpression experiments showed that EWS promotes exon 6 inclusion and consequently the synthesis of the proapoptotic FAS/CD95 isoform, whereas an EWS-FLI1 fusion protein characteristic of Ewing sarcomas shows decreased activity. Biochemical analyses revealed that EWS binding promotes the recruitment of U1snRNP and U2AF65 to the splice sites flanking exon 6 and therefore exon definition. Consistent with a role for EWS in the regulation of programmed cell death, cells depleted of EWS show decreased sensitivity to FAS-induced apoptosis, and elevated EWS expression enhances apoptosis in EWS-haploinsufficient Ewing sarcoma cells.


Assuntos
Apoptose/genética , Éxons , Sarcoma de Ewing/genética , Sarcoma de Ewing/patologia , Receptor fas/genética , Processamento Alternativo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteína Proto-Oncogênica c-fli-1/genética , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/metabolismo , Sarcoma de Ewing/metabolismo , Transdução de Sinais
10.
Mol Cell ; 52(5): 720-33, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24332178

RESUMO

RBM5, a regulator of alternative splicing of apoptotic genes, and its highly homologous RBM6 and RBM10 are RNA-binding proteins frequently deleted or mutated in lung cancer. We report that RBM5/6 and RBM10 antagonistically regulate the proliferative capacity of cancer cells and display distinct positional effects in alternative splicing regulation. We identify the Notch pathway regulator NUMB as a key target of these factors in the control of cell proliferation. NUMB alternative splicing, which is frequently altered in lung cancer, can regulate colony and xenograft tumor formation, and its modulation recapitulates or antagonizes the effects of RBM5, 6, and 10 in cell colony formation. RBM10 mutations identified in lung cancer cells disrupt NUMB splicing regulation to promote cell growth. Our results reveal a key genetic circuit in the control of cancer cell proliferation.


Assuntos
Processamento Alternativo , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Proteínas de Ligação a RNA/genética , Proteínas Supressoras de Tumor/genética , Animais , Sítios de Ligação , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Células HeLa , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Células MCF-7 , Camundongos , Camundongos Nus , RNA/genética , Receptores Notch/genética , Transcriptoma
11.
Mol Cell ; 51(3): 279-80, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23932711

RESUMO

Is the activity of the complex molecular machineries in charge of gene expression saturated in the cell? In this issue, Munding et al. (2013) report that titration of spliceosomal components by abundant ribosomal protein transcripts controls splicing of other genes and contributes to meiosis-specific splicing in budding yeast.


Assuntos
Meiose/genética , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA , Saccharomyces cerevisiae/genética
12.
Nucleic Acids Res ; 37(4): 1269-79, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19136466

RESUMO

FRAXE is a form of mild to moderate mental retardation due to the silencing of the FMR2 gene. The cellular function of FMR2 protein is presently unknown. By analogy with its homologue AF4, FMR2 was supposed to have a role in transcriptional regulation, but robust evidences supporting this hypothesis are lacking. We observed that FMR2 co-localizes with the splicing factor SC35 in nuclear speckles, the nuclear regions where splicing factors are concentrated, assembled and modified. Similarly to what was reported for splicing factors, blocking splicing or transcription leads to the accumulation of FMR2 in enlarged, rounded speckles. FMR2 is also localized in the nucleolus when splicing is blocked. We show here that FMR2 is able to specifically bind the G-quartet-forming RNA structure with high affinity. Remarkably, in vivo, in the presence of FMR2, the ESE action of the G-quartet situated in mRNA of an alternatively spliced exon of a minigene or of the putative target FMR1 appears reduced. Interestingly, FMR1 is silenced in the fragile X syndrome, another form of mental retardation. All together, our findings strongly suggest that FMR2 is an RNA-binding protein, which might be involved in alternative splicing regulation through an interaction with G-quartet RNA structure.


Assuntos
Processamento Alternativo , Quadruplex G , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA/química , Animais , Linhagem Celular , Estruturas do Núcleo Celular/química , Células Cultivadas , Proteína do X Frágil da Deficiência Intelectual/genética , Humanos , Camundongos , Proteínas Nucleares/análise , Proteínas Nucleares/química , Estrutura Terciária de Proteína , RNA/metabolismo , Proteínas de Ligação a RNA/análise , Proteínas de Ligação a RNA/química
13.
PLoS Biol ; 7(1): e16, 2009 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19166269

RESUMO

Fragile X syndrome, the most frequent form of inherited mental retardation, is due to the absence of Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein involved in several steps of RNA metabolism. To date, two RNA motifs have been found to mediate FMRP/RNA interaction, the G-quartet and the "kissing complex," which both induce translational repression in the presence of FMRP. We show here a new role for FMRP as a positive modulator of translation. FMRP specifically binds Superoxide Dismutase 1 (Sod1) mRNA with high affinity through a novel RNA motif, SoSLIP (Sod1 mRNA Stem Loops Interacting with FMRP), which is folded as three independent stem-loop structures. FMRP induces a structural modification of the SoSLIP motif upon its interaction with it. SoSLIP also behaves as a translational activator whose action is potentiated by the interaction with FMRP. The absence of FMRP results in decreased expression of Sod1. Because it has been observed that brain metabolism of FMR1 null mice is more sensitive to oxidative stress, we propose that the deregulation of Sod1 expression may be at the basis of several traits of the physiopathology of the Fragile X syndrome, such as anxiety, sleep troubles, and autism.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Regulação da Expressão Gênica , RNA Mensageiro/metabolismo , Superóxido Dismutase/genética , Animais , Sítios de Ligação , Encéfalo/enzimologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Humanos , Camundongos , Camundongos Mutantes , Polirribossomos , Biossíntese de Proteínas , RNA Mensageiro/química , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
14.
Nucleic Acids Res ; 35(1): 299-306, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17170008

RESUMO

Fragile X syndrome, the most frequent form of inherited mental retardation, is due to the absence of expression of the Fragile X Mental Retardation Protein (FMRP), an RNA binding protein with high specificity for G-quartet RNA structure. FMRP is involved in several steps of mRNA metabolism: nucleocytoplasmic trafficking, translational control and transport along dendrites in neurons. Fragile X Related Protein 1 (FXR1P), a homologue and interactor of FMRP, has been postulated to have a function similar to FMRP, leading to the hypothesis that it can compensate for the absence of FMRP in Fragile X patients. Here we analyze the ability of three isoforms of FXR1P, expressed in different tissues, to bind G-quartet RNA structure specifically. Only the longest FXR1P isoform was found to be able to bind specifically the G-quartet RNA, albeit with a lower affinity as compared to FMRP, whereas the other two isoforms negatively regulate the affinity of FMRP for G-quartet RNA. This result is important to decipher the molecular basis of fragile X syndrome, through the understanding of FMRP action in the context of its multimolecular complex in different tissues. In addition, we show that the action of FXR1P is synergistic rather than compensatory for FMRP function.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Sequência de Aminoácidos , Cinética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Isoformas de Proteínas/metabolismo , RNA/química , Proteínas de Ligação a RNA/química
15.
Hum Mol Genet ; 15(9): 1525-38, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16571602

RESUMO

The fragile X syndrome, the leading cause of inherited mental retardation, is due to the inactivation of the fragile mental retardation 1 gene (FMR1) and the subsequent absence of its gene product FMRP. This RNA-binding protein is thought to control mRNA translation and its absence in fragile X cells leads to alteration in protein synthesis. In neurons, FMRP is thought to repress specific mRNAs during their transport as silent ribonucleoparticles (mRNPs) from the cell body to the distant synapses which are the sites of local synthesis of neuro-specific proteins. The mechanism by which FMRP sorts out its different mRNAs targets might be tuned by the intervention of different proteins. Using a yeast two-hybrid system, we identified MicroSpherule Protein 58 (MSP58) as a novel FMRP-cellular partner. In cell cultures, we found that MSP58 is predominantly present in the nucleus where it interacts with the nuclear isoform of FMRP. However, in neurons but not in glial cells, MSP58 is also present in the cytoplasmic compartment, as well as in neurites, where it co-localizes with FMRP. Biochemical evidence is given that MSP58 is associated with polyribosomal poly(A)+ mRNPs. We also show that MSP58, similar to FMRP, is present on polyribosomes prepared from synaptoneurosomes and that it behaves as an RNA-binding protein with a high affinity to the G-quartet structure. We propose that this novel cellular partner for FMRP escorts FMRP-containing mRNP from the nucleus and nucleolus to the somato-dendritic compartment where it might participate in neuronal translation regulation.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Nucléolo Celular/metabolismo , Chlorocebus aethiops , Regulação da Expressão Gênica/fisiologia , Células HeLa , Humanos , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley
16.
Hum Mol Genet ; 14(6): 835-44, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15703194

RESUMO

Fragile X syndrome, the most common form of inherited mental retardation, is caused by absence of FMRP, an RNA-binding protein implicated in regulation of mRNA translation and/or transport. We have previously shown that dFMR1, the Drosophila ortholog of FMRP, is genetically linked to the dRac1 GTPase, a key player in actin cytoskeleton remodeling. Here, we demonstrate that FMRP and the Rac1 pathway are connected in a model of murine fibroblasts. We show that Rac1 activation induces relocalization of four FMRP partners to actin ring areas. Moreover, Rac1-induced actin remodeling is altered in fibroblasts lacking FMRP or carrying a point-mutation in the KH1 or in the KH2 RNA-binding domain. In absence of wild-type FMRP, we found that phospho-ADF/Cofilin (P-Cofilin) level, a major mediator of Rac1 signaling, is lowered, whereas the level of protein phosphatase 2A catalytic subunit (PP2Ac), a P-Cofilin phosphatase, is increased. We show that FMRP binds with high affinity to the 5'-UTR of pp2acbeta mRNA and is thus a likely negative regulator of its translation. The molecular mechanism unraveled here points to a role for FMRP in modulation of actin dynamics, which is a key process in morphogenesis of dendritic spines, synaptic structures abnormally developed in Fragile X syndrome patient's brain.


Assuntos
Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...