Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Genome Biol ; 25(1): 162, 2024 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-38902825

RESUMO

BACKGROUND: The functional coupling between alternative pre-mRNA splicing (AS) and the mRNA quality control mechanism called nonsense-mediated decay (NMD) can modulate transcript abundance. Previous studies have identified several examples of such a regulation in developing neurons. However, the systems-level effects of AS-NMD in this context are poorly understood. RESULTS: We developed an R package, factR2, which offers a comprehensive suite of AS-NMD analysis functions. Using this tool, we conducted a longitudinal analysis of gene expression in pluripotent stem cells undergoing induced neuronal differentiation. Our analysis uncovers hundreds of AS-NMD events with significant potential to regulate gene expression. Notably, this regulation is significantly overrepresented in specific functional groups of developmentally downregulated genes. Particularly strong association with gene downregulation is detected for alternative cassette exons stimulating NMD upon their inclusion into mature mRNA. By combining bioinformatic analyses with CRISPR/Cas9 genome editing and other experimental approaches we show that NMD-stimulating cassette exons regulated by the RNA-binding protein PTBP1 dampen the expression of their genes in developing neurons. We also provided evidence that the inclusion of NMD-stimulating cassette exons into mature mRNAs is temporally coordinated with NMD-independent gene repression mechanisms. CONCLUSIONS: Our study provides an accessible workflow for the discovery and prioritization of AS-NMD targets. It further argues that the AS-NMD pathway plays a widespread role in developing neurons by facilitating the downregulation of functionally related non-neuronal genes.


Assuntos
Processamento Alternativo , Regulação para Baixo , Neurônios , Degradação do RNAm Mediada por Códon sem Sentido , Proteína de Ligação a Regiões Ricas em Polipirimidinas , Animais , Camundongos , Neurônios/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Éxons , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Diferenciação Celular/genética , Neurogênese/genética
2.
Nucleus ; 15(1): 2306777, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38281066

RESUMO

The perinucleolar compartment (PNC) was initially identified as a nuclear structure enriched for the polypyrimidine tract-binding protein. Since then, the PNC has been implicated in carcinogenesis. The prevalence of this compartment is positively correlated with disease progression in various types of cancer, and its expression in primary tumors is linked to worse patient outcomes. Using the PNC as a surrogate marker for anti-cancer drug efficacy has led to the development of a clinical candidate for anti-metastasis therapies. The PNC is a multicomponent nuclear body situated at the periphery of the nucleolus. Thus far, several non-coding RNAs and RNA-binding proteins have been identified as the PNC components. Here, we summarize the current understanding of the structure and function of the PNC, as well as its recurrent links to cancer progression and metastasis.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Núcleo Celular/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Nucléolo Celular/metabolismo , Proteínas de Ligação a RNA/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/metabolismo
3.
Glia ; 72(1): 90-110, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37632136

RESUMO

Brain-derived neurotrophic factor (BDNF) plays a fundamental role in the developing and adult nervous system, contributing to neuronal survival, differentiation, and synaptic plasticity. Dysregulation of BDNF synthesis, secretion or signaling has been associated with many neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Although the transcriptional regulation of the Bdnf gene has been extensively studied in neurons, less is known about the regulation and function of BDNF in non-neuronal cells. The most abundant type of non-neuronal cells in the brain, astrocytes, express BDNF in response to catecholamines. However, genetic elements responsible for this regulation have not been identified. Here, we investigated four potential Bdnf enhancer regions and based on reporter gene assays, CRISPR/Cas9 engineering and CAPTURE-3C-sequencing we conclude that a region 840 kb upstream of the Bdnf gene regulates catecholamine-dependent expression of Bdnf in rodent astrocytes. We also provide evidence that this regulation is mediated by CREB and AP1 family transcription factors. This is the first report of an enhancer coordinating the transcription of Bdnf gene in non-neuronal cells.


Assuntos
Astrócitos , Fator Neurotrófico Derivado do Encéfalo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Astrócitos/metabolismo , Catecolaminas/metabolismo , Fatores de Transcrição/metabolismo , Neurônios/metabolismo , Roedores/metabolismo
4.
Hum Genet ; 2023 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-38153590

RESUMO

Simple repeated sequences (SRSs), defined as tandem iterations of microsatellite- to satellite-sized DNA units, occupy a substantial part of the human genome. Some of these elements are known to be transcribed in the context of repeat expansion disorders. Mounting evidence suggests that the transcription of SRSs may also contribute to normal cellular functions. Here, we used genome-wide bioinformatics approaches to systematically examine SRS transcriptional activity in cells undergoing neuronal differentiation. We identified thousands of long noncoding RNAs containing >200-nucleotide-long SRSs (SRS-lncRNAs), with hundreds of these transcripts significantly upregulated in the neural lineage. We show that SRS-lncRNAs often originate from telomere-proximal regions and that they have a strong potential to form multivalent contacts with a wide range of RNA-binding proteins. Our analyses also uncovered a cluster of neurally upregulated SRS-lncRNAs encoded in a centromere-proximal part of chromosome 9, which underwent an evolutionarily recent segmental duplication. Using a newly established in vitro system for rapid neuronal differentiation of induced pluripotent stem cells, we demonstrate that at least some of the bioinformatically predicted SRS-lncRNAs, including those encoded in the segmentally duplicated part of chromosome 9, indeed increase their expression in developing neurons to readily detectable levels. These and other lines of evidence suggest that many SRSs may be expressed in a cell type and developmental stage-specific manner, providing a valuable resource for further studies focused on the functional consequences of SRS-lncRNAs in the normal development of the human brain, as well as in the context of neurodevelopmental disorders.

5.
STAR Protoc ; 4(4): 102644, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37862173

RESUMO

Inducible degradation of proteins of interest provides a powerful approach for functional studies. Here, we present a protocol for tightly controlled depletion of the RNA-binding protein PTBP1 in mouse embryonic stem cells (ESCs). We describe steps for establishing an ESC line expressing doxycycline-inducible auxin receptor protein OsTIR1 and tagging endogenous Ptbp1 alleles using CRISPR-Cas9 and homology-directed repair reagents. We then detail procedures for assaying the efficiency of inducible PTBP1 knockdown by immunoblotting. This protocol is adaptable for other protein targets. For complete details on the use and execution of this protocol, please refer to Iannone et al.1.


Assuntos
Ácidos Indolacéticos , Células-Tronco Embrionárias Murinas , Animais , Camundongos , Ácidos Indolacéticos/farmacologia , Proteínas de Ligação a RNA , Doxiciclina
6.
Mol Cell ; 83(2): 203-218.e9, 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36626906

RESUMO

Many spliceosomal introns are excised from nascent transcripts emerging from RNA polymerase II (RNA Pol II). The extent of cell-type-specific regulation and possible functions of such co-transcriptional events remain poorly understood. We examined the role of the RNA-binding protein PTBP1 in this process using an acute depletion approach followed by the analysis of chromatin- and RNA Pol II-associated transcripts. We show that PTBP1 activates the co-transcriptional excision of hundreds of introns, a surprising effect given that this protein is known to promote intron retention. Importantly, some co-transcriptionally activated introns fail to complete their splicing without PTBP1. In a striking example, retention of a PTBP1-dependent intron triggers nonsense-mediated decay of transcripts encoding DNA methyltransferase DNMT3B. We provide evidence that this regulation facilitates the natural decline in DNMT3B levels in developing neurons and protects differentiation-specific genes from ectopic methylation. Thus, PTBP1-activated co-transcriptional splicing is a widespread phenomenon mediating epigenetic control of cellular identity.


Assuntos
Células-Tronco Pluripotentes , RNA Polimerase II , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Splicing de RNA/genética , Spliceossomos/metabolismo , Íntrons/genética , Células-Tronco Pluripotentes/metabolismo , Epigênese Genética , Processamento Alternativo
7.
Nat Commun ; 13(1): 6994, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36414621

RESUMO

Loss of SFPQ is a hallmark of motor degeneration in ALS and prevents maturation of motor neurons when occurring during embryogenesis. Here, we show that in zebrafish, developing motor neurons lacking SFPQ exhibit axon extension, branching and synaptogenesis defects, prior to degeneration. Subcellular transcriptomics reveals that loss of SFPQ in neurons produces a complex set of aberrant intron-retaining (IR) transcripts coding for neuron-specific proteins that accumulate in neurites. Some of these local IR mRNAs are prematurely terminated within the retained intron (PreT-IR). PreT-IR mRNAs undergo intronic polyadenylation, nuclear export, and localise to neurites in vitro and in vivo. We find these IR and PreT-IR mRNAs enriched in RNAseq datasets of tissue from patients with familial and sporadic ALS. This shared signature, between SFPQ-depleted neurons and ALS, functionally implicates SFPQ with the disease and suggests that neurite-centred perturbation of alternatively spliced isoforms drives the neurodegenerative process.


Assuntos
Esclerose Lateral Amiotrófica , Animais , Íntrons/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Axônios/metabolismo , Neurônios Motores/metabolismo
8.
Curr Biol ; 32(23): 5099-5115.e8, 2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36384140

RESUMO

Regulation of pre-mRNA splicing and polyadenylation plays a profound role in neurons by diversifying the proteome and modulating gene expression in response to physiological cues. Although most of the pre-mRNA processing is thought to occur in the nucleus, numerous splicing regulators are also found in neurites. Here, we show that U1-70K/SNRNP70, a component of the major spliceosome, localizes in RNA-associated granules in zebrafish axons. We identify the extra-nuclear SNRNP70 as an important regulator of motor axonal growth, nerve-dependent acetylcholine receptor (AChR) clustering, and neuromuscular synaptogenesis. This cytoplasmic pool has a protective role for a limited number of transcripts regulating their abundance and trafficking inside axons. Moreover, non-nuclear SNRNP70 regulates splice variants of transcripts such as agrin, thereby controlling synapse formation. Our results point to an unexpected, yet essential, function of non-nuclear SNRNP70 in axonal development, indicating a role of spliceosome proteins in cytoplasmic RNA metabolism during neuronal connectivity.


Assuntos
Precursores de RNA , Peixe-Zebra , Animais , Peixe-Zebra/genética
9.
STAR Protoc ; 3(1): 101139, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35128480

RESUMO

This protocol describes a hybridization-proximity labeling (HyPro) approach for identification of proteins and RNAs co-localizing with a transcript of interest in genetically unperturbed cells. It outlines steps required for purification of a recombinant HyPro enzyme, hybridization of fixed and permeabilized cells with digoxigenin-labeled probes, HyPro enzyme binding, proximity biotinylation, and downstream analyses of the biotinylated products. Although the protocol is optimized for relatively abundant noncoding transcripts, recommendations are provided for improving the signal-to-noise ratio in case of scarcer RNA "baits." For complete details on the use and execution of this protocol, please refer to Yap et al. (2021).


Assuntos
RNA , Biotinilação , Linhagem Celular , Células Cultivadas , Digoxigenina , Humanos , RNA/genética
10.
Mol Cell ; 82(2): 463-478.e11, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-34741808

RESUMO

The ability of RNAs to form specific contacts with other macromolecules provides an important mechanism for subcellular compartmentalization. Here we describe a suite of hybridization-proximity (HyPro) labeling technologies for unbiased discovery of proteins (HyPro-MS) and transcripts (HyPro-seq) associated with RNAs of interest in genetically unperturbed cells. As a proof of principle, we show that HyPro-MS and HyPro-seq can identify both known and previously unexplored spatial neighbors of the noncoding RNAs 45S, NEAT1, and PNCTR expressed at markedly different levels. Notably, HyPro-seq uncovers an extensive repertoire of incompletely processed, adenosine-to-inosine-edited transcripts accumulating at the interface between their encoding chromosomal regions and the NEAT1-containing paraspeckle compartment. At least some of these targets require NEAT1 for their optimal expression. Overall, this study provides a versatile toolkit for dissecting RNA interactomes in diverse biomedical contexts and expands our understanding of the functional architecture of the mammalian nucleus.


Assuntos
Compartimento Celular , Núcleo Celular/metabolismo , Técnicas Genéticas , RNA Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Núcleo Celular/genética , Células HeLa , Humanos , Espectrometria de Massas , Estudo de Prova de Conceito , Ligação Proteica , Proteoma , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Nuclear/genética , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Proteínas de Ligação a RNA/genética , RNA-Seq , Transcriptoma
11.
Nat Commun ; 12(1): 1918, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33771997

RESUMO

The RNA-binding protein SFPQ plays an important role in neuronal development and has been associated with several neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Alzheimer's disease. Here, we report that loss of sfpq leads to premature termination of multiple transcripts due to widespread activation of previously unannotated cryptic last exons (CLEs). These SFPQ-inhibited CLEs appear preferentially in long introns of genes with neuronal functions and can dampen gene expression outputs and/or give rise to short peptides interfering with the normal gene functions. We show that one such peptide encoded by the CLE-containing epha4b mRNA isoform is responsible for neurodevelopmental defects in the sfpq mutant. The uncovered CLE-repressive activity of SFPQ is conserved in mouse and human, and SFPQ-inhibited CLEs are found expressed across ALS iPSC-derived neurons. These results greatly expand our understanding of SFPQ function and uncover a gene regulation mechanism with wide relevance to human neuropathologies.


Assuntos
Esclerose Lateral Amiotrófica/genética , Códon sem Sentido , Éxons/genética , Fator de Processamento Associado a PTB/genética , Animais , Sequência de Bases , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Humanos , Hibridização In Situ/métodos , Íntrons/genética , Camundongos , Neurônios/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética
12.
Elife ; 102021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33560226

RESUMO

Brain-derived neurotrophic factor (BDNF) controls the survival, growth, and function of neurons both during the development and in the adult nervous system. Bdnf is transcribed from several distinct promoters generating transcripts with alternative 5' exons. Bdnf transcripts initiated at the first cluster of exons have been associated with the regulation of body weight and various aspects of social behavior, but the mechanisms driving the expression of these transcripts have remained poorly understood. Here, we identify an evolutionarily conserved intronic enhancer region inside the Bdnf gene that regulates both basal and stimulus-dependent expression of the Bdnf transcripts starting from the first cluster of 5' exons in mouse and rat neurons. We further uncover a functional E-box element in the enhancer region, linking the expression of Bdnf and various pro-neural basic helix-loop-helix transcription factors. Collectively, our results shed new light on the cell-type- and stimulus-specific regulation of the important neurotrophic factor BDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Regulação da Expressão Gênica , Íntrons , Neurônios/fisiologia , Regiões Promotoras Genéticas , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Feminino , Humanos , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
13.
Dev Cell ; 52(4): 477-491.e8, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-32097653

RESUMO

Most adult neurons and glia originate from radial glial progenitors (RGs), a type of stem cell typically extending from the apical to the basal side of the developing cortex. Precise regulation of the choice between RG self-renewal and differentiation is critical for normal development, but the mechanisms underlying this transition remain elusive. We show that the non-canonical tubulin Tuba8, transiently expressed in cortical progenitors, drives differentiation of RGs into apical intermediate progenitors, a more restricted progenitor type lacking attachment to the basal lamina. This effect depends on the unique C-terminal sequence of Tuba8 that antagonizes tubulin tyrosination and Δ2 cleavage, two post-translational modifications (PTMs) essential for RG fiber maintenance and the switch between direct and indirect neurogenesis and ultimately distinct neuronal lineage outcomes. Our work uncovers an instructive role of a developmentally regulated tubulin isotype in progenitor differentiation and provides new insights into biological functions of the cellular tubulin PTM "code."


Assuntos
Diferenciação Celular , Córtex Cerebral/citologia , Fator 10 de Crescimento de Fibroblastos/fisiologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios/citologia , Tubulina (Proteína)/fisiologia , Animais , Linhagem da Célula , Células Cultivadas , Córtex Cerebral/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Neurogênese , Neuroglia/metabolismo , Neurônios/metabolismo , Tirosina/metabolismo
14.
Nat Commun ; 11(1): 361, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31953406

RESUMO

Eukaryotic gene expression relies on extensive crosstalk between transcription and RNA processing. Changes in this composite regulation network may provide an important means for shaping cell type-specific transcriptomes. Here we show that the RNA-associated protein Srrt/Ars2 sustains embryonic stem cell (ESC) identity by preventing premature termination of numerous transcripts at cryptic cleavage/polyadenylation sites in first introns. Srrt interacts with the nuclear cap-binding complex and facilitates recruitment of the spliceosome component U1 snRNP to cognate intronic positions. At least in some cases, U1 recruited in this manner inhibits downstream cleavage/polyadenylation events through a splicing-independent mechanism called telescripting. We further provide evidence that the naturally high expression of Srrt in ESCs offsets deleterious effects of retrotransposable sequences accumulating in its targets. Our work identifies Srrt as a molecular guardian of the pluripotent cell state.


Assuntos
Células-Tronco Embrionárias/metabolismo , Processamento Pós-Transcricional do RNA , Transcriptoma , Animais , Sequência de Bases , Diferenciação Celular , Biologia Computacional , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Íntrons/genética , Camundongos , Poliadenilação , Proteínas/genética , Proteínas/metabolismo , Splicing de RNA , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Spliceossomos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética
15.
Adv Mater ; 31(28): e1900514, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31081206

RESUMO

Local mechanical cues can affect crucial fate decisions of living cells. Transepithelial stress has been discussed in the context of epithelial monolayers, but the lack of appropriate experimental systems leads current studies to approximate it simply as an in-plane stress. To evaluate possible contribution of force vectors acting in other directions, double epithelium in a 3D-printed "GeminiChip" containing a sessile and a pendant channel is reconstituted. Intriguingly, the sessile epithelia is prone to apoptotic cell extrusion upon crowding, whereas the pendant counterpart favors live cell delamination. Transcriptome analyses show upregulation of RhoA, BMP2, and hypoxia-signaling genes in the pendant epithelium, consistent with the onset of an epithelial-mesenchymal transition program. HepG2 microtumor spheroids also display differential spreading patterns in the sessile and pendant configuration. Using this multilayered GeminiChip, these results uncover a progressive yet critical role of perpendicular force vectors in collective cell behaviors and point at fundamental importance of these forces in the biology of cancer.


Assuntos
Epitélio/metabolismo , Homeostase , Fenômenos Mecânicos , Impressão Tridimensional , Fenômenos Biomecânicos , Estresse Mecânico
16.
Mol Cell ; 72(3): 525-540.e13, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30318443

RESUMO

Functions of many long noncoding RNAs (lncRNAs) depend on their ability to interact with multiple copies of specific RNA-binding proteins (RBPs). Here, we devised a workflow combining bioinformatics and experimental validation steps to systematically identify RNAs capable of multivalent RBP recruitment. This uncovered a number of previously unknown transcripts encoding high-density RBP recognition arrays within genetically normal short tandem repeats. We show that a top-scoring hit in this screen, lncRNA PNCTR, contains hundreds of pyrimidine tract-binding protein (PTBP1)-specific motifs allowing it to sequester a substantial fraction of PTBP1 in a nuclear body called perinucleolar compartment. Importantly, PNCTR is markedly overexpressed in a variety of cancer cells and its downregulation is sufficient to induce programmed cell death at least in part by stimulating PTBP1 splicing regulation activity. This work expands our understanding of the repeat-containing fraction of the human genome and illuminates a novel mechanism driving malignant transformation of cancer cells.


Assuntos
Processamento Alternativo/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/fisiologia , Proteínas de Ligação a RNA/fisiologia , Processamento Alternativo/genética , Linhagem Celular , Movimento Celular , Núcleo Celular , Proliferação de Células , Sobrevivência Celular , Biologia Computacional/métodos , Éxons , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Repetições de Microssatélites/genética , Repetições de Microssatélites/fisiologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Pirimidinas , Splicing de RNA , RNA Longo não Codificante/fisiologia
17.
PLoS Genet ; 13(5): e1006824, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28549066

RESUMO

Cellular homeostasis of the minor spliceosome is regulated by a negative feed-back loop that targets U11-48K and U11/U12-65K mRNAs encoding essential components of the U12-type intron-specific U11/U12 di-snRNP. This involves interaction of the U11 snRNP with an evolutionarily conserved splicing enhancer giving rise to unproductive mRNA isoforms. In the case of U11/U12-65K, this mechanism controls the length of the 3' untranslated region (3'UTR). We show that this process is dynamically regulated in developing neurons and some other cell types, and involves a binary switch between translation-competent mRNAs with a short 3'UTR to non-productive isoforms with a long 3'UTR that are retained in the nucleus or/and spliced to the downstream amylase locus. Importantly, the choice between these alternatives is determined by alternative terminal exon definition events regulated by conserved U12- and U2-type 5' splice sites as well as sequence signals used for pre-mRNA cleavage and polyadenylation. We additionally show that U11 snRNP binding to the U11/U12-65K mRNA species with a long 3'UTR is required for their nuclear retention. Together, our studies uncover an intricate molecular circuitry regulating the abundance of a key spliceosomal protein and shed new light on the mechanisms limiting the export of non-productively spliced mRNAs from the nucleus to the cytoplasm.


Assuntos
Processamento Alternativo , Núcleo Celular/metabolismo , Éxons , Ribonucleoproteínas Nucleares Pequenas/genética , Transporte Ativo do Núcleo Celular , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Spliceossomos/metabolismo
18.
Nucleic Acids Res ; 45(11): 6761-6774, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28379442

RESUMO

RBM10 is an RNA-binding protein that plays an essential role in development and is frequently mutated in the context of human disease. RBM10 recognizes a diverse set of RNA motifs in introns and exons and regulates alternative splicing. However, the molecular mechanisms underlying this seemingly relaxed sequence specificity are not understood and functional studies have focused on 3΄ intronic sites only. Here, we dissect the RNA code recognized by RBM10 and relate it to the splicing regulatory function of this protein. We show that a two-domain RRM1-ZnF unit recognizes a GGA-centered motif enriched in RBM10 exonic sites with high affinity and specificity and test that the interaction with these exonic sequences promotes exon skipping. Importantly, a second RRM domain (RRM2) of RBM10 recognizes a C-rich sequence, which explains its known interaction with the intronic 3΄ site of NUMB exon 9 contributing to regulation of the Notch pathway in cancer. Together, these findings explain RBM10's broad RNA specificity and suggest that RBM10 functions as a splicing regulator using two RNA-binding units with different specificities to promote exon skipping.


Assuntos
Proteínas de Ligação a RNA/fisiologia , Autoantígenos , Sequência de Bases , Sítios de Ligação , Éxons , Células HEK293 , Humanos , Ligação Proteica , Splicing de RNA , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Dedos de Zinco
19.
Nucleic Acids Res ; 45(21): 12455-12468, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30053257

RESUMO

Many RNA-binding proteins including a master regulator of splicing in developing brain and muscle, polypyrimidine tract-binding protein 1 (PTBP1), can either activate or repress alternative exons depending on the pre-mRNA recruitment position. When bound upstream or within regulated exons PTBP1 tends to promote their skipping, whereas binding to downstream sites often stimulates inclusion. How this switch is orchestrated at the molecular level is poorly understood. Using bioinformatics and biochemical approaches we show that interaction of PTBP1 with downstream intronic sequences can activate natural cassette exons by promoting productive docking of the spliceosomal U1 snRNP to a suboptimal 5' splice site. Strikingly, introducing upstream PTBP1 sites to this circuitry leads to a potent splicing repression accompanied by the assembly of an exonic ribonucleoprotein complex with a tightly bound U1 but not U2 snRNP. Our data suggest a molecular mechanism underlying the transition between a better-known repressive function of PTBP1 and its role as a bona fide splicing activator. More generally, we argue that the functional outcome of individual RNA contacts made by an RNA-binding protein is subject to extensive context-specific modulation.


Assuntos
Processamento Alternativo , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Modelos Genéticos , Proteína de Ligação a Regiões Ricas em Polipirimidinas/fisiologia , Processamento Alternativo/genética , Animais , Linhagem Celular Tumoral , Biologia Computacional , Proteínas de Ligação a DNA/genética , Éxons/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Íntrons/genética , Camundongos , Neuroblastoma , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1 , Ubiquitina-Proteína Ligases
20.
Biochem Soc Trans ; 44(4): 1079-85, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27528755

RESUMO

Alternative pre-mRNA splicing provides an effective means for expanding coding capacity of eukaryotic genomes. Recent studies suggest that co-expression of different splice isoforms may increase diversity of RNAs and proteins at a single-cell level. A pertinent question in the field is whether such co-expression is biologically meaningful or, rather, represents insufficiently stringent splicing regulation. Here we argue that isoform co-expression may produce functional outcomes that are difficult and sometimes impossible to achieve using other regulation strategies. Far from being a 'splicing noise', co-expression is often established through co-ordinated activity of specific cis-elements and trans-acting factors. Further work in this area may uncover new biological functions of alternative splicing (AS) and generate important insights into mechanisms allowing different cell types to attain their unique molecular identities.


Assuntos
Processamento Alternativo , Variação Genética , Precursores de RNA/genética , RNA Mensageiro/genética , Animais , Células Eucarióticas/metabolismo , Humanos , Modelos Genéticos , Isoformas de Proteínas/genética , Análise de Célula Única/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA