Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Mol Cell ; 83(2): 203-218.e9, 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36626906

RESUMEN

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.


Asunto(s)
Células Madre Pluripotentes , ARN Polimerasa II , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Empalme del ARN/genética , Empalmosomas/metabolismo , Intrones/genética , Células Madre Pluripotentes/metabolismo , Epigénesis Genética , Empalme Alternativo
2.
Mol Cell ; 82(2): 463-478.e11, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-34741808

RESUMEN

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.


Asunto(s)
Compartimento Celular , Núcleo Celular/metabolismo , Técnicas Genéticas , ARN Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Núcleo Celular/genética , Células HeLa , Humanos , Espectrometría de Masas , Prueba de Estudio Conceptual , Unión Proteica , Proteoma , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Nuclear/genética , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/genética , RNA-Seq , Transcriptoma
3.
Mol Cell ; 72(3): 525-540.e13, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30318443

RESUMEN

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.


Asunto(s)
Empalme Alternativo/fisiología , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Proteína de Unión al Tracto de Polipirimidina/fisiología , Proteínas de Unión al ARN/fisiología , Empalme Alternativo/genética , Línea Celular , Movimiento Celular , Núcleo Celular , Proliferación Celular , Supervivencia Celular , Biología Computacional/métodos , Exones , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Repeticiones de Microsatélite/genética , Repeticiones de Microsatélite/fisiología , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Pirimidinas , Empalme del ARN , ARN Largo no Codificante/fisiología
4.
Glia ; 72(1): 90-110, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37632136

RESUMEN

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.


Asunto(s)
Astrocitos , Factor Neurotrófico Derivado del Encéfalo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Astrocitos/metabolismo , Catecolaminas/metabolismo , Factores de Transcripción/metabolismo , Neuronas/metabolismo , Roedores/metabolismo
5.
Hum Genet ; 143(7): 875-895, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38153590

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas , Neuronas , ARN Largo no Codificante , Humanos , Neuronas/metabolismo , ARN Largo no Codificante/genética , Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Secuencias Repetitivas de Ácidos Nucleicos , Transcripción Genética , Regulación del Desarrollo de la Expresión Génica , Genoma Humano , Neurogénesis/genética , Repeticiones de Microsatélite , Biología Computacional/métodos
6.
PLoS Genet ; 13(5): e1006824, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28549066

RESUMEN

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.


Asunto(s)
Empalme Alternativo , Núcleo Celular/metabolismo , Exones , Ribonucleoproteínas Nucleares Pequeñas/genética , Transporte Activo de Núcleo Celular , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Empalmosomas/metabolismo
7.
Genes Dev ; 26(11): 1209-23, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661231

RESUMEN

Differentiated cells acquire unique structural and functional traits through coordinated expression of lineage-specific genes. An extensive battery of genes encoding components of the synaptic transmission machinery and specialized cytoskeletal proteins is activated during neurogenesis, but the underlying regulation is not well understood. Here we show that genes encoding critical presynaptic proteins are transcribed at a detectable level in both neurons and nonneuronal cells. However, in nonneuronal cells, the splicing of 3'-terminal introns within these genes is repressed by the polypyrimidine tract-binding protein (Ptbp1). This inhibits the export of incompletely spliced mRNAs to the cytoplasm and triggers their nuclear degradation. Clearance of these intron-containing transcripts occurs independently of the nonsense-mediated decay (NMD) pathway but requires components of the nuclear RNA surveillance machinery, including the nuclear pore-associated protein Tpr and the exosome complex. When Ptbp1 expression decreases during neuronal differentiation, the regulated introns are spliced out, thus allowing the accumulation of translation-competent mRNAs in the cytoplasm. We propose that this mechanism counters ectopic and precocious expression of functionally linked neuron-specific genes and ensures their coherent activation in the appropriate developmental context.


Asunto(s)
Diferenciación Celular , Neuronas/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Línea Celular Tumoral , Células Cultivadas , Exosomas/metabolismo , Técnicas de Silenciamiento del Gen , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Intrones , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Precursores del ARN/metabolismo , Toxina Shiga I/genética , Toxina Shiga I/metabolismo , Sintaxina 1/genética , Sintaxina 1/metabolismo
8.
Nucleic Acids Res ; 45(21): 12455-12468, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30053257

RESUMEN

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.


Asunto(s)
Empalme Alternativo , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Modelos Genéticos , Proteína de Unión al Tracto de Polipirimidina/fisiología , Empalme Alternativo/genética , Animales , Línea Celular Tumoral , Biología Computacional , Proteínas de Unión al ADN/genética , Exones/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Intrones/genética , Ratones , Neuroblastoma , Proteína de Unión al Tracto de Polipirimidina/genética , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1 , Ubiquitina-Proteína Ligasas
9.
Nucleic Acids Res ; 45(11): 6761-6774, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28379442

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ARN/fisiología , Autoantígenos , Secuencia de Bases , Sitios de Unión , Exones , Células HEK293 , Humanos , Unión Proteica , Empalme del ARN , ARN Mensajero/química , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Dedos de Zinc
10.
J Mol Cell Cardiol ; 121: 287-296, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30048712

RESUMEN

Cysteine and glycine rich protein 3 (CSRP3) encodes Muscle LIM Protein (MLP), a well-established disease gene for Hypertrophic Cardiomyopathy (HCM). MLP, in contrast to the proteins encoded by the other recognised HCM disease genes, is non-sarcomeric, and has important signalling functions in cardiomyocytes. To gain insight into the disease mechanisms involved, we generated a knock-in mouse (KI) model, carrying the well documented HCM-causing CSRP3 mutation C58G. In vivo phenotyping of homozygous KI/KI mice revealed a robust cardiomyopathy phenotype with diastolic and systolic left ventricular dysfunction, which was supported by increased heart weight measurements. Transcriptome analysis by RNA-seq identified activation of pro-fibrotic signalling, induction of the fetal gene programme and activation of markers of hypertrophic signalling in these hearts. Further ex vivo analyses validated the activation of these pathways at transcript and protein level. Intriguingly, the abundance of MLP decreased in KI/KI mice by 80% and in KI/+ mice by 50%. Protein depletion was also observed in cellular studies for two further HCM-causing CSRP3 mutations (L44P and S54R/E55G). We show that MLP depletion is caused by proteasome action. Moreover, MLP C58G interacts with Bag3 and results in a proteotoxic response in the homozygous knock-in mice, as shown by induction of Bag3 and associated heat shock proteins. In conclusion, the newly generated mouse model provides insights into the underlying disease mechanisms of cardiomyopathy caused by mutations in the non-sarcomeric protein MLP. Furthermore, our cellular experiments suggest that protein depletion and proteasomal overload also play a role in other HCM-causing CSPR3 mutations that we investigated, indicating that reduced levels of functional MLP may be a common mechanism for HCM-causing CSPR3 mutations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Cardiomiopatía Hipertrófica/genética , Corazón/fisiopatología , Proteínas con Dominio LIM/genética , Proteínas Musculares/genética , Animales , Cardiomiopatía Hipertrófica/fisiopatología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Ratones , Mutación , Sarcómeros/genética
11.
Bioessays ; 38(9): 830-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27438915

RESUMEN

Eukaryotic gene expression is extensively controlled at the level of mRNA stability and the mechanisms underlying this regulation are markedly different from their archaeal and bacterial counterparts. We propose that two such mechanisms, nonsense-mediated decay (NMD) and motif-specific transcript destabilization by CCCH-type zinc finger RNA-binding proteins, originated as a part of cellular defense against RNA pathogens. These branches of the mRNA turnover pathway might have been used by primeval eukaryotes alongside RNA interference to distinguish their own messages from those of RNA viruses and retrotransposable elements. We further hypothesize that the subsequent advent of "professional" innate and adaptive immunity systems allowed NMD and the motif-triggered mechanisms to be efficiently repurposed for regulation of endogenous cellular transcripts. This scenario explains the rapid emergence of archetypical mRNA destabilization pathways in eukaryotes and argues that other aspects of post-transcriptional gene regulation in this lineage might have been derived through a similar exaptation route.


Asunto(s)
Eucariontes/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido , Animales , Eucariontes/genética , Humanos
12.
Semin Cell Dev Biol ; 47-48: 32-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26493705

RESUMEN

A majority of messenger RNA precursors (pre-mRNAs) in the higher eukaryotes undergo alternative splicing to generate more than one mature product. By targeting the open reading frame region this process increases diversity of protein isoforms beyond the nominal coding capacity of the genome. However, alternative splicing also frequently controls output levels and spatiotemporal features of cellular and organismal gene expression programs. Here we discuss how these non-coding functions of alternative splicing contribute to development through regulation of mRNA stability, translational efficiency and cellular localization.


Asunto(s)
Empalme Alternativo , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Precursores del ARN/genética , Animales , Evolución Molecular , Humanos , Modelos Genéticos , Biosíntesis de Proteínas/genética , Precursores del ARN/metabolismo , Estabilidad del ARN/genética
13.
J Biol Chem ; 291(17): 9295-309, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26907693

RESUMEN

Many eukaryotic organisms encode more than one RNA-dependent RNA polymerase (RdRP) that probably emerged as a result of gene duplication. Such RdRP paralogs often participate in distinct RNA silencing pathways and show characteristic repertoires of enzymatic activities in vitro However, to what extent members of individual paralogous groups can undergo functional changes during speciation remains an open question. We show that orthologs of QDE-1, an RdRP component of the quelling pathway in Neurospora crassa, have rapidly diverged in evolution at the amino acid sequence level. Analyses of purified QDE-1 polymerases from N. crassa (QDE-1(Ncr)) and related fungi, Thielavia terrestris (QDE-1(Tte)) and Myceliophthora thermophila (QDE-1(Mth)), show that all three enzymes can synthesize RNA, but the precise modes of their action differ considerably. Unlike their QDE-1(Ncr) counterpart favoring processive RNA synthesis, QDE-1(Tte) and QDE-1(Mth) produce predominantly short RNA copies via primer-independent initiation. Surprisingly, a 3.19 Å resolution crystal structure of QDE-1(Tte) reveals a quasisymmetric dimer similar to QDE-1(Ncr) Further electron microscopy analyses confirm that QDE-1(Tte) occurs as a dimer in solution and retains this status upon interaction with a template. We conclude that divergence of orthologous RdRPs can result in functional innovation while retaining overall protein fold and quaternary structure.


Asunto(s)
Evolución Molecular , Proteínas Fúngicas , Neurospora crassa , Multimerización de Proteína/fisiología , ARN Polimerasa Dependiente del ARN , Cristalografía por Rayos X , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Neurospora crassa/enzimología , Neurospora crassa/genética , Estructura Cuaternaria de Proteína , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo
14.
PLoS Genet ; 10(11): e1004771, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375251

RESUMEN

Alternative splicing (AS) provides a potent mechanism for increasing protein diversity and modulating gene expression levels. How alternate splice sites are selected by the splicing machinery and how AS is integrated into gene regulation networks remain important questions of eukaryotic biology. Here we report that polypyrimidine tract-binding protein 1 (Ptbp1/PTB/hnRNP-I) controls alternate 5' and 3' splice site (5'ss and 3'ss) usage in a large set of mammalian transcripts. A top scoring event identified by our analysis was the choice between competing upstream and downstream 5'ss (u5'ss and d5'ss) in the exon 18 of the Hps1 gene. Hps1 is essential for proper biogenesis of lysosome-related organelles and loss of its function leads to a disease called type 1 Hermansky-Pudlak Syndrome (HPS). We show that Ptbp1 promotes preferential utilization of the u5'ss giving rise to stable mRNAs encoding a full-length Hps1 protein, whereas bias towards d5'ss triggered by Ptbp1 down-regulation generates transcripts susceptible to nonsense-mediated decay (NMD). We further demonstrate that Ptbp1 binds to pyrimidine-rich sequences between the u5'ss and d5'ss and activates the former site rather than repressing the latter. Consistent with this mechanism, u5'ss is intrinsically weaker than d5'ss, with a similar tendency observed for other genes with Ptbp1-induced u5'ss bias. Interestingly, the brain-enriched Ptbp1 paralog Ptbp2/nPTB/brPTB stimulated the u5'ss utilization but with a considerably lower efficiency than Ptbp1. This may account for the tight correlation between Hps1 with Ptbp1 expression levels observed across mammalian tissues. More generally, these data expand our understanding of AS regulation and uncover a post-transcriptional strategy ensuring co-expression of a subordinate gene with its master regulator through an AS-NMD tracking mechanism.


Asunto(s)
Empalme Alternativo/genética , Síndrome de Hermanski-Pudlak/genética , Proteína de Unión al Tracto de Polipirimidina/genética , ARN Mensajero/genética , Animales , Exones , Regulación de la Expresión Génica , Células HeLa , Síndrome de Hermanski-Pudlak/patología , Humanos , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Proteína de Unión al Tracto de Polipirimidina/biosíntesis , Sitios de Empalme de ARN/genética
15.
Biochem Soc Trans ; 44(4): 1079-85, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528755

RESUMEN

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.


Asunto(s)
Empalme Alternativo , Variación Genética , Precursores del ARN/genética , ARN Mensajero/genética , Animales , Células Eucariotas/metabolismo , Humanos , Modelos Genéticos , Isoformas de Proteínas/genética , Análisis de la Célula Individual/métodos
16.
J Biol Chem ; 289(30): 20788-801, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24878960

RESUMEN

Polycomb group protein Ezh2 is a histone H3 Lys-27 histone methyltransferase orchestrating an extensive epigenetic regulatory program. Several nervous system-specific genes are known to be repressed by Ezh2 in stem cells and derepressed during neuronal differentiation. However, the molecular mechanisms underlying this regulation remain poorly understood. Here we show that Ezh2 levels are dampened during neuronal differentiation by brain-enriched microRNA miR-124. Expression of miR-124 in a neuroblastoma cells line was sufficient to up-regulate a significant fraction of nervous system-specific Ezh2 target genes. On the other hand, naturally elevated expression of miR-124 in embryonic carcinoma cells undergoing neuronal differentiation correlated with down-regulation of Ezh2 levels. Importantly, overexpression of Ezh2 mRNA with a 3'-untranslated region (3'-UTR) lacking a functional miR-124 binding site, but not with the wild-type Ezh2 3'-UTR, hampered neuronal and promoted astrocyte-specific differentiation in P19 and embryonic mouse neural stem cells. Overall, our results uncover a molecular mechanism that allows miR-124 to balance the choice between alternative differentiation possibilities through fine-tuning the expression of a critical epigenetic regulator.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Neuronas/metabolismo , Complejo Represivo Polycomb 2/biosíntesis , Regiones no Traducidas 3'/fisiología , Animales , Astrocitos/citología , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Células HEK293 , Humanos , Ratones , MicroARNs/genética , Neuronas/citología , Complejo Represivo Polycomb 2/genética
17.
Biochem Soc Trans ; 42(4): 1168-73, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25110020

RESUMEN

Higher eukaryotes rely on AS (alternative splicing) of pre-mRNAs (mRNA precursors) to generate more than one protein product from a single gene and to regulate mRNA stability and translational activity. An important example of the latter function involves an interplay between AS and NMD (nonsense-mediated decay), a cytoplasmic quality control mechanism eliminating mRNAs containing PTCs (premature translation termination codons). Although originally identified as an error surveillance process, AS-NMD additionally provides an efficient strategy for deterministic regulation of gene expression outputs. In this review, we discuss recently published examples of AS-NMD and delineate functional contexts where recurrent use of this mechanism orchestrates expression of important genes.


Asunto(s)
Empalme Alternativo/genética , Precursores del ARN/genética , Empalme Alternativo/fisiología , Animales , Expresión Génica/genética , Expresión Génica/fisiología , Humanos , Degradación de ARNm Mediada por Codón sin Sentido/genética , Estabilidad del ARN/genética , Estabilidad del ARN/fisiología
18.
Nucleic Acids Res ; 40(2): 787-800, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21948791

RESUMEN

RNA-binding protein HuR modulates the stability and translational efficiency of messenger RNAs (mRNAs) encoding essential components of the cellular proliferation, growth and survival pathways. Consistent with these functions, HuR levels are often elevated in cancer cells and reduced in senescent and quiescent cells. However, the molecular mechanisms that control HuR expression are poorly understood. Here we show that HuR protein autoregulates its abundance through a negative feedback loop that involves interaction of the nuclear HuR protein with a GU-rich element (GRE) overlapping with the HuR major polyadenylation signal (PAS2). An increase in the cellular HuR protein levels stimulates the expression of long HuR mRNA species containing an AU-rich element (ARE) that destabilizes the mRNAs and thus reduces the protein production output. The PAS2 read-through occurs due to a reduced recruitment of the CstF-64 subunit of the pre-mRNA cleavage stimulation factor in the presence of the GRE-bound HuR. We propose that this mechanism maintains HuR homeostasis in proliferating cells. Since only the nuclear HuR is expected to contribute to the auto-regulation, our model may explain the longstanding observation that the increase in the total HuR expression in cancer cells often correlates with the accumulation of its substantial fraction in the cytoplasm.


Asunto(s)
Proteínas ELAV/genética , Poliadenilación , Regiones no Traducidas 3' , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Línea Celular , Factor de Estimulación del Desdoblamiento , Citoplasma/metabolismo , Proteínas ELAV/metabolismo , Retroalimentación Fisiológica , Humanos , Ratones , Precursores del ARN/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo
19.
Nucleic Acids Res ; 40(14): 6808-20, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22505576

RESUMEN

MicroRNAs (miRNAs) are 19- to 25-nt-long non-coding RNAs that regulate gene expression by base-pairing with target mRNAs and reducing their stability or translational efficiency. Mammalian miRNAs function in association with four closely related Argonaute proteins, AGO1-4. All four proteins contain the PAZ and the MID domains interacting with the miRNA 3' and 5' termini, respectively, as well as the PIWI domain comprising an mRNA 'slicing' activity in the case of AGO2 but not AGO1, AGO3 and AGO4. However, the slicing mode of the miRNA-programmed AGO2 is rarely realized in vivo and the four Argonautes are thought to play largely overlapping roles in the mammalian miRNA pathway. Here, we show that the average length of many miRNAs is diminished during nervous system development as a result of progressive shortening of the miRNA 3' ends. We link this modification with an increase in the fractional abundance of Ago2 in the adult brain and identify a specific structural motif within the PAZ domain that enables efficient trimming of miRNAs associated with this but not the other three Argonautes. Taken together, our data suggest that mammalian Argonautes may define the length and possibly biological activity of mature mammalian miRNAs in a developmentally controlled manner.


Asunto(s)
Proteínas Argonautas/metabolismo , MicroARNs/metabolismo , Animales , Proteínas Argonautas/química , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/química , Neuronas/citología , Estructura Terciaria de Proteína , Procesamiento Postranscripcional del ARN , Ribonucleasa III/metabolismo
20.
Mol Cell Neurosci ; 56: 420-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23357783

RESUMEN

Eukaryotic gene expression is orchestrated on a genome-wide scale through several post-transcriptional mechanisms. Of these, alternative pre-mRNA splicing expands the proteome diversity and modulates mRNA stability through downstream RNA quality control (QC) pathways including nonsense-mediated decay (NMD) of mRNAs containing premature termination codons and nuclear retention and elimination (NRE) of intron-containing transcripts. Although originally identified as mechanisms for eliminating aberrant transcripts, a growing body of evidence suggests that NMD and NRE coupled with deliberate changes in pre-mRNA splicing patterns are also used in a number of biological contexts for deterministic control of gene expression. Here we review recent studies elucidating molecular mechanisms and biological significance of these gene regulation strategies with a specific focus on their roles in nervous system development and physiology. This article is part of a Special Issue entitled 'RNA and splicing regulation in neurodegeneration'.


Asunto(s)
Empalme Alternativo , Enfermedades Neurodegenerativas/genética , Degradación de ARNm Mediada por Codón sin Sentido , ARN Mensajero/metabolismo , Animales , Humanos , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA