Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 82(1): 75-89.e9, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34942120

RESUMEN

Circular RNAs (circRNAs) are widely expressed in eukaryotes and are regulated in many biological processes. Although several studies indicate their activity as microRNA (miRNA) and protein sponges, little is known about their ability to directly control mRNA homeostasis. We show that the widely expressed circZNF609 directly interacts with several mRNAs and increases their stability and/or translation by favoring the recruitment of the RNA-binding protein ELAVL1. Particularly, the interaction with CKAP5 mRNA, which interestingly overlaps the back-splicing junction, enhances CKAP5 translation, regulating microtubule function in cancer cells and sustaining cell-cycle progression. Finally, we show that circZNF609 downregulation increases the sensitivity of several cancer cell lines to different microtubule-targeting chemotherapeutic drugs and that locked nucleic acid (LNA) protectors against the pairing region on circZNF609 phenocopy such effects. These data set an example of how the small effects tuned by circZNF609/CKAP5 mRNA interaction might have a potent output in tumor growth and drug response.


Asunto(s)
Carcinogénesis , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neoplasias/metabolismo , ARN Circular/metabolismo , ARN Mensajero/metabolismo , Animales , Antineoplásicos/farmacología , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células K562 , Masculino , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/efectos de los fármacos , Microtúbulos/genética , Microtúbulos/patología , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , ARN Circular/genética , ARN Mensajero/genética , Transducción de Señal , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
2.
EMBO J ; 41(13): e108918, 2022 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-35698802

RESUMEN

The transition from dividing progenitors to postmitotic motor neurons (MNs) is orchestrated by a series of events, which are mainly studied at the transcriptional level by analyzing the activity of specific programming transcription factors. Here, we identify a post-transcriptional role of a MN-specific transcriptional unit (MN2) harboring a lncRNA (lncMN2-203) and two miRNAs (miR-325-3p and miR-384-5p) in this transition. Through the use of in vitro mESC differentiation and single-cell sequencing of CRISPR/Cas9 mutants, we demonstrate that lncMN2-203 affects MN differentiation by sponging miR-466i-5p and upregulating its targets, including several factors involved in neuronal differentiation and function. In parallel, miR-325-3p and miR-384-5p, co-transcribed with lncMN2-203, act by repressing proliferation-related factors. These findings indicate the functional relevance of the MN2 locus and exemplify additional layers of specificity regulation in MN differentiation.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Diferenciación Celular/genética , MicroARNs/genética , Neuronas Motoras , ARN Largo no Codificante/genética
3.
Cell ; 147(2): 358-69, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000014

RESUMEN

Recently, a new regulatory circuitry has been identified in which RNAs can crosstalk with each other by competing for shared microRNAs. Such competing endogenous RNAs (ceRNAs) regulate the distribution of miRNA molecules on their targets and thereby impose an additional level of post-transcriptional regulation. Here we identify a muscle-specific long noncoding RNA, linc-MD1, which governs the time of muscle differentiation by acting as a ceRNA in mouse and human myoblasts. Downregulation or overexpression of linc-MD1 correlate with retardation or anticipation of the muscle differentiation program, respectively. We show that linc-MD1 "sponges" miR-133 and miR-133 [corrected] to regulate the expression of MAML1 and MEF2C, transcription factors that activate muscle-specific gene expression. Finally, we demonstrate that linc-MD1 exerts the same control over differentiation timing in human myoblasts, and that its levels are strongly reduced in Duchenne muscle cells. We conclude that the ceRNA network plays an important role in muscle differentiation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Desarrollo de Músculos , Músculo Esquelético/citología , ARN no Traducido/metabolismo , Animales , Secuencia de Bases , Proteínas de Unión al ADN/genética , Humanos , Proteínas de Dominio MADS/genética , Factores de Transcripción MEF2 , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/embriología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Mioblastos/metabolismo , Factores Reguladores Miogénicos/genética , Proteínas Nucleares/genética , Procesamiento Postranscripcional del ARN , ARN Largo no Codificante , Factores de Transcripción/genética
4.
Nucleic Acids Res ; 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38989616

RESUMEN

The combination of morphogenetic and transcription factors together with the synergic aid of noncoding RNAs and their cognate RNA binding proteins contribute to shape motor neurons (MN) identity. Here, we extend the noncoding perspective of human MN, by detailing the molecular and biological activity of CyCoNP (as Cytoplasmic Coordinator of Neural Progenitors) a highly expressed and MN-enriched human lncRNA. Through in silico prediction, in vivo RNA purification and loss of function experiments followed by RNA-sequencing, we found that CyCoNP sustains a specific neuron differentiation program, required for the physiology of both neuroblastoma cells and hiPSC-derived MN, which mainly involves miR-4492 and NCAM1 mRNA. We propose a novel lncRNA-mediated 'dual mode' of action, in which CyCoNP acts in trans as a classical RNA sponge by sequestering miR-4492 from its pro-neuronal targets, including NCAM1 mRNA, and at the same time it plays an additional role in cis by interacting with NCAM1 mRNA and regulating the availability and localization of the miR-4492 in its proximity. These data highlight novel insights into the noncoding RNA-mediated control of human neuron physiology and point out the importance of lncRNA-mediated interactions for the spatial distribution of regulatory molecules.

5.
Nat Methods ; 19(10): 1208-1220, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35618955

RESUMEN

Circular RNAs (circRNAs) are formed in all domains of life and via different mechanisms. There has been an explosion in the number of circRNA papers in recent years; however, as a relatively young field, circRNA biology has an urgent need for common experimental standards for isolating, analyzing, expressing and depleting circRNAs. Here we propose a set of guidelines for circRNA studies based on the authors' experience. This Perspective will specifically address the major class of circRNAs in Eukarya that are generated by a spliceosome-catalyzed back-splicing event. We hope that the implementation of best practice principles for circRNA research will help move the field forward and allow a better functional understanding of this fascinating group of RNAs.


Asunto(s)
ARN Circular , ARN , ARN/genética , ARN/metabolismo , Empalme del ARN
6.
Mol Cell ; 66(1): 22-37.e9, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28344082

RESUMEN

Circular RNAs (circRNAs) constitute a family of transcripts with unique structures and still largely unknown functions. Their biogenesis, which proceeds via a back-splicing reaction, is fairly well characterized, whereas their role in the modulation of physiologically relevant processes is still unclear. Here we performed expression profiling of circRNAs during in vitro differentiation of murine and human myoblasts, and we identified conserved species regulated in myogenesis and altered in Duchenne muscular dystrophy. A high-content functional genomic screen allowed the study of their functional role in muscle differentiation. One of them, circ-ZNF609, resulted in specifically controlling myoblast proliferation. Circ-ZNF609 contains an open reading frame spanning from the start codon, in common with the linear transcript, and terminating at an in-frame STOP codon, created upon circularization. Circ-ZNF609 is associated with heavy polysomes, and it is translated into a protein in a splicing-dependent and cap-independent manner, providing an example of a protein-coding circRNA in eukaryotes.


Asunto(s)
Proliferación Celular , Desarrollo de Músculos , Proteínas Musculares/biosíntesis , Distrofia Muscular de Duchenne/metabolismo , Mioblastos Esqueléticos/metabolismo , Biosíntesis de Proteínas , ARN/metabolismo , Animales , Genotipo , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Proteínas Musculares/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Mioblastos Esqueléticos/patología , Sistemas de Lectura Abierta , Fenotipo , ARN/genética , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , Interferencia de ARN , Empalme del ARN , ARN Circular , Análisis de Secuencia de ARN/métodos , Transducción de Señal , Transfección
7.
Development ; 147(16)2020 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-32839270

RESUMEN

In recent years, circular RNAs (circRNAs) - a novel class of RNA molecules characterized by their covalently closed circular structure - have emerged as a complex family of eukaryotic transcripts with important biological features. Besides their peculiar structure, which makes them particularly stable molecules, they have attracted much interest because their expression is strongly tissue and cell specific. Moreover, many circRNAs are conserved across eukaryotes, localized in particular subcellular compartments, and can play disparate molecular functions. The discovery of circRNAs has therefore added not only another layer of gene expression regulation but also an additional degree of complexity to our understanding of the structure, function and evolution of eukaryotic genomes. In this Review, we summarize current knowledge of circRNAs and discuss the possible functions of circRNAs in cell differentiation and development.


Asunto(s)
Diferenciación Celular , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Genoma Humano , ARN Circular/biosíntesis , Animales , Humanos , ARN Circular/genética
8.
EMBO J ; 37(18)2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30177572

RESUMEN

Myogenesis is a highly regulated process that involves the conversion of progenitor cells into multinucleated myofibers. Besides proteins and miRNAs, long noncoding RNAs (lncRNAs) have been shown to participate in myogenic regulatory circuitries. Here, we characterize a murine chromatin-associated muscle-specific lncRNA, Charme, which contributes to the robustness of the myogenic program in vitro and in vivo In myocytes, Charme depletion triggers the disassembly of a specific chromosomal domain and the downregulation of myogenic genes contained therein. Notably, several Charme-sensitive genes are associated with human cardiomyopathies and Charme depletion in mice results in a peculiar cardiac remodeling phenotype with changes in size, structure, and shape of the heart. Moreover, the existence of an orthologous transcript in human, regulating the same subset of target genes, suggests an important and evolutionarily conserved function for Charme Altogether, these data describe a new example of a chromatin-associated lncRNA regulating the robustness of skeletal and cardiac myogenesis.


Asunto(s)
Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/metabolismo , Remodelación Ventricular , Animales , Humanos , Ratones , Fibras Musculares Esqueléticas/patología , Miocitos Cardíacos/patología , ARN Largo no Codificante/genética
9.
EMBO Rep ; 21(6): e49942, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32337838

RESUMEN

Guanine-quadruplexes (G4) included in RNA molecules exert several functions in controlling gene expression at post-transcriptional level; however, the molecular mechanisms of G4-mediated regulation are still poorly understood. Here, we describe a regulatory circuitry operating in the early phases of murine muscle differentiation in which a long non-coding RNA (SMaRT) base pairs with a G4-containing mRNA (Mlx-γ) and represses its translation by counteracting the activity of the DHX36 RNA helicase. The time-restricted, specific effect of lnc-SMaRT on the translation of Mlx-γ isoform modulates the general subcellular localization of total MLX proteins, impacting on their transcriptional output and promoting proper myogenesis and mature myotube formation. Therefore, the circuitry made of lnc-SMaRT, Mlx-γ, and DHX36 not only plays an important role in the control of myogenesis but also unravels a molecular mechanism where G4 structures and G4 unwinding activities are regulated in living cells.


Asunto(s)
G-Cuádruplex , ARN Largo no Codificante , Animales , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN Helicasas , Ratones , ARN Largo no Codificante/genética , ARN Mensajero/genética
10.
Nat Rev Genet ; 22(9): 550-551, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34017103
11.
Mol Cell ; 53(3): 506-14, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24440503

RESUMEN

The muscle-specific long noncoding RNA linc-MD1 was shown to be expressed during early phases of muscle differentiation and to trigger the switch to later stages by acting as a sponge for miR-133 and miR-135. Notably, linc-MD1 is also the host transcript of miR-133b, and their biogenesis is mutually exclusive. Here, we describe that this alternative synthesis is controlled by the HuR protein, which favors linc-MD1 accumulation through its ability to bind linc-MD1 and repress Drosha cleavage. We show that HuR is under the repressive control of miR-133 and that the sponging activity of linc-MD1 consolidates HuR expression in a feedforward positive loop. Finally, we show that HuR also acts in the cytoplasm, reinforcing linc-MD1 sponge activity by cooperating for miRNA recruitment. An increase in miR-133 synthesis, mainly from the two unrelated miR-133a coding genomic loci, is likely to trigger the exit from this circuitry and progression to later differentiation stages.


Asunto(s)
Proteínas ELAV/fisiología , Desarrollo de Músculos/genética , ARN Largo no Codificante/fisiología , Animales , Diferenciación Celular , Línea Celular , Citoplasma/metabolismo , Proteínas ELAV/genética , Proteínas ELAV/metabolismo , Retroalimentación Fisiológica , Ratones , MicroARNs/análisis , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
12.
Nucleic Acids Res ; 48(12): 6889-6905, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32479602

RESUMEN

Mutations in the RNA-binding protein FUS cause amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease. FUS plays a role in numerous aspects of RNA metabolism, including mRNA splicing. However, the impact of ALS-causative mutations on splicing has not been fully characterized, as most disease models have been based on overexpressing mutant FUS, which will alter RNA processing due to FUS autoregulation. We and others have recently created knockin models that overcome the overexpression problem, and have generated high depth RNA-sequencing on FUS mutants in parallel to FUS knockout, allowing us to compare mutation-induced changes to genuine loss of function. We find that FUS-ALS mutations induce a widespread loss of function on expression and splicing. Specifically, we find that mutant FUS directly alters intron retention levels in RNA-binding proteins. Moreover, we identify an intron retention event in FUS itself that is associated with its autoregulation. Altered FUS levels have been linked to disease, and we show here that this novel autoregulation mechanism is altered by FUS mutations. Crucially, we also observe this phenomenon in other genetic forms of ALS, including those caused by TDP-43, VCP and SOD1 mutations, supporting the concept that multiple ALS genes interact in a regulatory network.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Homeostasis/genética , Proteína FUS de Unión a ARN/genética , Animales , Citoplasma/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Humanos , Intrones/genética , Mutación con Pérdida de Función , Ratones , Ratones Noqueados , Mutación/genética , Empalme del ARN/genética , Superóxido Dismutasa-1/genética , Proteína que Contiene Valosina/genética
13.
Nat Rev Genet ; 15(1): 7-21, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24296535

RESUMEN

Genomes of multicellular organisms are characterized by the pervasive expression of different types of non-coding RNAs (ncRNAs). Long ncRNAs (lncRNAs) belong to a novel heterogeneous class of ncRNAs that includes thousands of different species. lncRNAs have crucial roles in gene expression control during both developmental and differentiation processes, and the number of lncRNA species increases in genomes of developmentally complex organisms, which highlights the importance of RNA-based levels of control in the evolution of multicellular organisms. In this Review, we describe the function of lncRNAs in developmental processes, such as in dosage compensation, genomic imprinting, cell differentiation and organogenesis, with a particular emphasis on mammalian development.


Asunto(s)
Diferenciación Celular/genética , Compensación de Dosificación (Genética)/genética , Regulación del Desarrollo de la Expresión Génica/genética , Impresión Genómica/genética , Mamíferos/genética , Modelos Genéticos , Organogénesis/genética , ARN Largo no Codificante/genética , Animales , Diferenciación Celular/fisiología , Citoplasma/genética , Mamíferos/crecimiento & desarrollo , Modelos Moleculares , Músculos/fisiología , Especificidad de la Especie
14.
Nucleic Acids Res ; 46(2): 917-928, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29165713

RESUMEN

The human transcriptome contains thousands of long non-coding RNAs (lncRNAs). Characterizing their function is a current challenge. An emerging concept is that lncRNAs serve as protein scaffolds, forming ribonucleoproteins and bringing proteins in proximity. However, only few scaffolding lncRNAs have been characterized and the prevalence of this function is unknown. Here, we propose the first computational approach aimed at predicting scaffolding lncRNAs at large scale. We predicted the largest human lncRNA-protein interaction network to date using the catRAPID omics algorithm. In combination with tissue expression and statistical approaches, we identified 847 lncRNAs (∼5% of the long non-coding transcriptome) predicted to scaffold half of the known protein complexes and network modules. Lastly, we show that the association of certain lncRNAs to disease may involve their scaffolding ability. Overall, our results suggest for the first time that RNA-mediated scaffolding of protein complexes and modules may be a common mechanism in human cells.


Asunto(s)
Biología Computacional/métodos , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Algoritmos , Predisposición Genética a la Enfermedad/genética , Humanos , Unión Proteica , Mapas de Interacción de Proteínas , Proteoma/genética , Proteoma/metabolismo , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/genética , Transcriptoma
15.
Int J Mol Sci ; 21(21)2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153123

RESUMEN

Skeletal muscle atrophy is a pathological condition so far without effective treatment and poorly understood at a molecular level. Emerging evidence suggest a key role for circular RNAs (circRNA) during myogenesis and their deregulation has been reported to be associated with muscle diseases. Spermine oxidase (SMOX), a polyamine catabolic enzyme plays a critical role in muscle differentiation and the existence of a circRNA arising from SMOX gene has been recently identified. In this study, we evaluated the expression profile of circular and linear SMOX in both C2C12 differentiation and dexamethasone-induced myotubes atrophy. To validate our findings in vivo their expression levels were also tested in two murine models of amyotrophic lateral sclerosis: SOD1G93A and hFUS+/+, characterized by progressive muscle atrophy. During C2C12 differentiation, linear and circular SMOX show the same trend of expression. Interestingly, in atrophy circSMOX levels significantly increased compared to the physiological state, in both in vitro and in vivo models. Our study demonstrates that SMOX represents a new player in muscle physiopathology and provides a scientific basis for further investigation on circSMOX RNA as a possible new therapeutic target for the treatment of muscle atrophy.


Asunto(s)
Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , ARN Circular/fisiología , ARN Mensajero/fisiología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Diferenciación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/fisiología , ARN no Traducido/fisiología , Proteína FUS de Unión a ARN/genética , Superóxido Dismutasa-1/genética , Poliamino Oxidasa
16.
Int J Mol Sci ; 20(8)2019 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-31010208

RESUMEN

Circular RNAs (circRNAs) constitute a recently re-discovered class of non-coding RNAs functioning as sponges for miRNAs and proteins, affecting RNA splicing and regulating transcription. CircRNAs are generated by "back-splicing", which is the linking covalently of 3'- and 5'-ends of exons. Thus, circRNA levels might be deregulated in conditions associated with altered RNA-splicing. Significantly, growing evidence indicates their role in human diseases. Specifically, myotonic dystrophy type 1 (DM1) is a multisystemic disorder caused by expanded CTG repeats in the DMPK gene which results in abnormal mRNA-splicing. In this investigation, circRNAs expressed in DM1 skeletal muscles were identified by analyzing RNA-sequencing data-sets followed by qPCR validation. In muscle biopsies, out of nine tested, four transcripts showed an increased circular fraction: CDYL, HIPK3, RTN4_03, and ZNF609. Their circular fraction values correlated with skeletal muscle strength and with splicing biomarkers of disease severity, and displayed higher values in more severely affected patients. Moreover, Receiver-Operating-Characteristics curves of these four circRNAs discriminated DM1 patients from controls. The identified circRNAs were also detectable in peripheral-blood-mononuclear-cells (PBMCs) and the plasma of DM1 patients, but they were not regulated significantly. Finally, increased circular fractions of RTN4_03 and ZNF609 were also observed in differentiated myogenic cell lines derived from DM1 patients. In conclusion, this pilot study identified circRNA dysregulation in DM1 patients.


Asunto(s)
Regulación de la Expresión Génica , Distrofia Miotónica/genética , ARN/genética , Adulto , Empalme Alternativo/genética , Estudios de Casos y Controles , Línea Celular , Femenino , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Miotónica/sangre , Reacción en Cadena de la Polimerasa , ARN/sangre , ARN Circular , Reproducibilidad de los Resultados
17.
J Neurosci ; 37(3): 546-561, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28100738

RESUMEN

MicroRNAs (miRNA) play an important role in post-transcriptional gene regulation of several physiological and pathological processes. In multiple sclerosis (MS), a chronic inflammatory and degenerative disease of the CNS, and in its mouse model, the experimental autoimmune encephalomyelitis (EAE), miRNA dysregulation has been mainly related to immune system dysfunction and white matter (WM) pathology. However, little is known about their role in gray matter pathology. Here, we explored miRNA involvement in the inflammation-driven alterations of synaptic structure and function, collectively known as synaptopathy, a neuropathological process contributing to excitotoxic neurodegeneration in MS/EAE. Particularly, we observed that miR-142-3p is increased in the CSF of patients with active MS and in EAE brains. We propose miR-142-3p as a molecular mediator of the IL-1ß-dependent downregulation of the glial glutamate-aspartate transporter (GLAST), which causes an enhancement of the glutamatergic transmission in the EAE cerebellum. The synaptic abnormalities mediated by IL-1ß and the clinical and neuropathological manifestations of EAE disappeared in miR-142 knock-out mice. Furthermore, we observed that in vivo miR-142-3p inhibition, either by a preventive and local treatment or by a therapeutic and systemic strategy, abolished IL-1ß- and GLAST-dependent synaptopathy in EAE wild-type mice. Consistently, miR-142-3p was responsible for the glutamatergic synaptic alterations caused by CSF of patients with MS, and CSF levels of miR-142-3p correlated with prospective MS disease progression. Our findings highlight miR-142-3p as key molecular player in IL-1ß-mediated synaptic dysfunction, possibly leading to excitotoxic damage in both EAE and MS diseases. Inhibition of miR-142-3p could be neuroprotective in MS. SIGNIFICANCE STATEMENT: Current studies suggest the role of glutamate excitotoxicity in the development and progression of multiple sclerosis (MS) and of its mouse model experimental autoimmune encephalomyelitis (EAE). The molecular mechanisms linking inflammation and synaptic alterations in MS/EAE are still unknown. Here, we identified miR-142-3p as a determinant molecular actor in inflammation-dependent synaptopathy typical of both MS and EAE. miR-142-3p was upregulated in the CSF of MS patients and in EAE cerebellum. Inhibition of miR-142-3p, locally in EAE brain and in a MS chimeric ex vivo model, recovered glutamatergic synaptic enhancement typical of EAE/MS. We proved that miR-142-3p promoted the IL-1ß-dependent glutamate dysfunction by targeting glutamate-aspartate transporter (GLAST), a crucial glial transporter involved in glutamate homeostasis. Finally, we suggest miR-142-3p as a negative prognostic factor in patients with relapsing-remitting multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Interleucina-1beta/biosíntesis , MicroARNs/biosíntesis , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Sinapsis/metabolismo , Adulto , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/patología , Femenino , Técnicas de Sustitución del Gen , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/líquido cefalorraquídeo , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/líquido cefalorraquídeo , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Sinapsis/patología
18.
RNA ; 22(10): 1510-21, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27473170

RESUMEN

Post-transcriptional gene regulation is a fundamental step for coordinating cellular response in a variety of processes. RNA-binding proteins (RBPs) and microRNAs (miRNAs) are the most important factors responsible for this regulation. Here we report that different components of the miR-200 family are involved in c-Jun mRNA regulation with the opposite effect. While miR-200b inhibits c-Jun protein production, miR-200a tends to increase the JUN amount through a stabilization of its mRNA. This action is dependent on the presence of the RBP HuR that binds the 3'UTR of c-Jun mRNA in a region including the mir-200a binding site. The position of the binding site is fundamental; by mutating this site, we demonstrate that the effect is not micro-RNA specific. These results indicate that miR-200a triggers a microRNA-mediated stabilization of c-Jun mRNA, promoting the binding of HuR with c-Jun mRNA. This is the first example of a positive regulation exerted by a microRNA on an important oncogene in proliferating cells.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , MicroARNs/genética , Regiones no Traducidas 3' , Sitios de Unión , Proteína 1 Similar a ELAV/genética , Células HEK293 , Células HeLa , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Células MCF-7 , MicroARNs/metabolismo , Unión Proteica , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
19.
EMBO J ; 31(24): 4502-10, 2012 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-23232809

RESUMEN

microRNA abundance has been shown to depend on the amount of the microprocessor components or, in some cases, on specific auxiliary co-factors. In this paper, we show that the FUS/TLS (fused in sarcoma/translocated in liposarcoma) protein, associated with familial forms of Amyotrophic Lateral Sclerosis (ALS), contributes to the biogenesis of a specific subset of microRNAs. Among them, species with roles in neuronal function, differentiation and synaptogenesis were identified. We also show that FUS/TLS is recruited to chromatin at sites of their transcription and binds the corresponding pri-microRNAs. Moreover, FUS/TLS depletion leads to decreased Drosha level at the same chromatin loci. Limited FUS/TLS depletion leads to a reduced microRNA biogenesis and we suggest a possible link between FUS mutations affecting nuclear/cytoplasmic partitioning of the protein and altered neuronal microRNA biogenesis in ALS pathogenesis.


Asunto(s)
Cromatina/metabolismo , MicroARNs/biosíntesis , Neuronas/citología , Proteína FUS de Unión a ARN/metabolismo , Ribonucleasa III/metabolismo , Sinapsis/fisiología , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoprecipitación , Neuronas/fisiología , Oligonucleótidos/genética , Plásmidos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Sinapsis/genética
20.
Acta Neuropathol ; 132(2): 175-196, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27164932

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating neurological disease with no effective treatment available. An increasing number of genetic causes of ALS are being identified, but how these genetic defects lead to motor neuron degeneration and to which extent they affect common cellular pathways remains incompletely understood. To address these questions, we performed an interactomic analysis to identify binding partners of wild-type (WT) and ALS-associated mutant versions of ATXN2, C9orf72, FUS, OPTN, TDP-43 and UBQLN2 in neuronal cells. This analysis identified several known but also many novel binding partners of these proteins. Interactomes of WT and mutant ALS proteins were very similar except for OPTN and UBQLN2, in which mutations caused loss or gain of protein interactions. Several of the identified interactomes showed a high degree of overlap: shared binding partners of ATXN2, FUS and TDP-43 had roles in RNA metabolism; OPTN- and UBQLN2-interacting proteins were related to protein degradation and protein transport, and C9orf72 interactors function in mitochondria. To confirm that this overlap is important for ALS pathogenesis, we studied fragile X mental retardation protein (FMRP), one of the common interactors of ATXN2, FUS and TDP-43, in more detail in in vitro and in vivo model systems for FUS ALS. FMRP localized to mutant FUS-containing aggregates in spinal motor neurons and bound endogenous FUS in a direct and RNA-sensitive manner. Furthermore, defects in synaptic FMRP mRNA target expression, neuromuscular junction integrity, and motor behavior caused by mutant FUS in zebrafish embryos, could be rescued by exogenous FMRP expression. Together, these results show that interactomics analysis can provide crucial insight into ALS disease mechanisms and they link FMRP to motor neuron dysfunction caused by FUS mutations.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Ataxina-2/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Ataxina-2/genética , Proteínas Relacionadas con la Autofagia , Proteína C9orf72 , Proteínas de Ciclo Celular , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas de Transporte de Membrana , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neuronas/metabolismo , Proteína FUS de Unión a ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA