Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 186(11): 2438-2455.e22, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37178687

RESUMEN

The generation of distinct messenger RNA isoforms through alternative RNA processing modulates the expression and function of genes, often in a cell-type-specific manner. Here, we assess the regulatory relationships between transcription initiation, alternative splicing, and 3' end site selection. Applying long-read sequencing to accurately represent even the longest transcripts from end to end, we quantify mRNA isoforms in Drosophila tissues, including the transcriptionally complex nervous system. We find that in Drosophila heads, as well as in human cerebral organoids, 3' end site choice is globally influenced by the site of transcription initiation (TSS). "Dominant promoters," characterized by specific epigenetic signatures including p300/CBP binding, impose a transcriptional constraint to define splice and polyadenylation variants. In vivo deletion or overexpression of dominant promoters as well as p300/CBP loss disrupted the 3' end expression landscape. Our study demonstrates the crucial impact of TSS choice on the regulation of transcript diversity and tissue identity.


Asunto(s)
Empalme Alternativo , Isoformas de ARN , Sitio de Iniciación de la Transcripción , Humanos , Poliadenilación , Regiones Promotoras Genéticas , Isoformas de ARN/metabolismo , ARN Mensajero/metabolismo
2.
Nat Methods ; 20(10): 1544-1552, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37735569

RESUMEN

Organoids derived from stem cells have become an increasingly important tool for studying human development and modeling disease. However, methods are still needed to control and study spatiotemporal patterns of gene expression in organoids. Here we combined optogenetics and gene perturbation technologies to activate or knock-down RNA of target genes in programmable spatiotemporal patterns. To illustrate the usefulness of our approach, we locally activated Sonic Hedgehog (SHH) signaling in an organoid model for human neurodevelopment. Spatial and single-cell transcriptomic analyses showed that this local induction was sufficient to generate stereotypically patterned organoids and revealed new insights into SHH's contribution to gene regulation in neurodevelopment. With this study, we propose optogenetic perturbations in combination with spatial transcriptomics as a powerful technology to reprogram and study cell fates and tissue patterning in organoids.


Asunto(s)
Proteínas Hedgehog , Optogenética , Humanos , Proteínas Hedgehog/metabolismo , Organoides/metabolismo , Diferenciación Celular , Expresión Génica
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.
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
5.
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
6.
Nat Methods ; 16(9): 879-886, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31384046

RESUMEN

Although messenger RNAs are key molecules for understanding life, until now, no method has existed to determine the full-length sequence of endogenous mRNAs including their poly(A) tails. Moreover, although non-A nucleotides can be incorporated in poly(A) tails, there also exists no method to accurately sequence them. Here, we present full-length poly(A) and mRNA sequencing (FLAM-seq), a rapid and simple method for high-quality sequencing of entire mRNAs. We report a complementary DNA library preparation method coupled to single-molecule sequencing to perform FLAM-seq. Using human cell lines, brain organoids and Caenorhabditis elegans we show that FLAM-seq delivers high-quality full-length mRNA sequences for thousands of different genes per sample. We find that 3' untranslated region length is correlated with poly(A) tail length, that alternative polyadenylation sites and alternative promoters for the same gene are linked to different tail lengths, and that tails contain a substantial number of cytosines.


Asunto(s)
Encéfalo/metabolismo , Organoides/metabolismo , Poli A/química , Poli A/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN/métodos , Animales , Caenorhabditis elegans , Regulación de la Expresión Génica , Células HeLa , Humanos , Poli A/genética , Poliadenilación , Regiones Promotoras Genéticas , ARN Mensajero/genética
7.
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
8.
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
9.
Biochem Soc Trans ; 41(4): 844-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23863142

RESUMEN

It is now becoming largely accepted that the non-coding portion of the genome, rather than its coding counterpart, is likely to account for the greater complexity of higher eukaryotes. Moreover, non-coding RNAs have been demonstrated to participate in regulatory circuitries that are crucial for development and differentiation. Whereas the biogenesis and function of small non-coding RNAs, particularly miRNAs (microRNAs), has been extensively clarified in many eukaryotic systems, very little is known about the long non-coding counterpart of the transcriptome. In the present review, we revise the current knowledge of how small non-coding RNAs and lncRNAs (long non-coding RNAs) impinge on circuitries controlling proper muscle differentiation and homoeostasis and how their biogenesis is regulated. Moreover, we provide new insights into an additional mechanism of post-transcriptional regulation mediated by lncRNAs, which, acting as miRNA 'sponges', have an impact on the distribution of miRNA molecules on their targets with features similar to those described for ceRNAs (competing endogenous RNAs).


Asunto(s)
Diferenciación Celular/genética , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , ARN no Traducido/genética , Humanos , MicroARNs/fisiología , Distrofia Muscular de Duchenne/patología
10.
iScience ; 26(1): 105878, 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36691625

RESUMEN

Poly(A) tails protect RNAs from degradation and their deadenylation rates determine RNA stability. Although poly(A) tails are generated in the nucleus, deadenylation of tails has mostly been investigated within the cytoplasm. Here, we combined long-read sequencing with metabolic labeling, splicing inhibition and cell fractionation experiments to quantify, separately, the genesis and trimming of nuclear and cytoplasmic tails in vitro and in vivo. We present evidence for genome-wide, nuclear synthesis of tails longer than 200 nt, which are rapidly shortened after transcription. Our data suggests that rapid deadenylation is a nuclear process, and that different classes of transcripts and even transcript isoforms have distinct nuclear tail lengths. For example, many long-noncoding RNAs retain long poly(A) tails. Modeling deadenylation dynamics predicts nuclear deadenylation about 10 times faster than cytoplasmic deadenylation. In summary, our data suggests that nuclear deadenylation might be a key mechanism for regulating mRNA stability, abundance, and subcellular localization.

11.
Nat Microbiol ; 8(7): 1252-1266, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37349587

RESUMEN

Herpes simplex encephalitis is a life-threatening disease of the central nervous system caused by herpes simplex viruses (HSVs). Following standard of care with antiviral acyclovir treatment, most patients still experience various neurological sequelae. Here we characterize HSV-1 infection of human brain organoids by combining single-cell RNA sequencing, electrophysiology and immunostaining. We observed strong perturbations of tissue integrity, neuronal function and cellular transcriptomes. Under acyclovir treatment viral replication was stopped, but did not prevent HSV-1-driven defects such as damage of neuronal processes and neuroepithelium. Unbiased analysis of pathways deregulated upon infection revealed tumour necrosis factor activation as a potential causal factor. Combination of anti-inflammatory drugs such as necrostatin-1 or bardoxolone methyl with antiviral treatment prevented the damages caused by infection, indicating that tuning the inflammatory response in acute infection may improve current therapeutic strategies.


Asunto(s)
Encefalitis Viral , Herpes Simple , Herpesvirus Humano 1 , Humanos , Herpesvirus Humano 1/genética , Herpes Simple/complicaciones , Herpes Simple/tratamiento farmacológico , Aciclovir/farmacología , Aciclovir/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Encefalitis Viral/tratamiento farmacológico , Organoides
12.
Sci Adv ; 8(47): eadd9938, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36427315

RESUMEN

Soft-bodied cephalopods such as octopuses are exceptionally intelligent invertebrates with a highly complex nervous system that evolved independently from vertebrates. Because of elevated RNA editing in their nervous tissues, we hypothesized that RNA regulation may play a major role in the cognitive success of this group. We thus profiled messenger RNAs and small RNAs in three cephalopod species including 18 tissues of the Octopus vulgaris. We show that the major RNA innovation of soft-bodied cephalopods is an expansion of the microRNA (miRNA) gene repertoire. These evolutionarily novel miRNAs were primarily expressed in adult neuronal tissues and during the development and had conserved and thus likely functional target sites. The only comparable miRNA expansions happened, notably, in vertebrates. Thus, we propose that miRNAs are intimately linked to the evolution of complex animal brains.


Asunto(s)
MicroARNs , Octopodiformes , Animales , Octopodiformes/genética , MicroARNs/genética , Encéfalo , Alimentos Marinos , ARN Mensajero
13.
Front Mol Neurosci ; 15: 832133, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35310884

RESUMEN

Mesial temporal lobe epilepsy (mTLE) is a chronic disease characterized by recurrent seizures that originate in the temporal lobes of the brain. Anti-epileptic drugs (AEDs) are the standard treatment for managing seizures in mTLE patients, but are frequently ineffective. Resective surgery is an option for some patients, but does not guarantee a postoperative seizure-free period. Therefore, further insight is needed into the pathogenesis of mTLE to enable the design of new therapeutic strategies. Circular RNAs (circRNAs) have been identified as important regulators of neuronal function and have been implicated in epilepsy. However, the mechanisms through which circRNAs contribute to epileptogenesis remain unknown. Here, we determine the circRNA transcriptome of the hippocampus and cortex of mTLE patients by using RNA-seq. We report 333 differentially expressed (DE) circRNAs between healthy individuals and mTLE patients, of which 23 circRNAs displayed significant adjusted p-values following multiple testing correction. Interestingly, hippocampal expression of circ_Satb1, a circRNA derived from special AT-rich sequence binding protein 1 (SATB1), is decreased in both mTLE patients and in experimental epilepsy. Our work shows that circ_Satb1 displays dynamic patterns of neuronal expression in vitro and in vivo. Further, circ_Satb1-specific knockdown using CRISPR/CasRx approaches in hippocampal cultures leads to defects in dendritic spine morphology, a cellular hallmark of mTLE. Overall, our results identify a novel epilepsy-associated circRNA with disease-specific expression and previously unidentified cellular effects that are relevant for epileptogenesis.

14.
Mol Ther ; 18(9): 1675-82, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20551908

RESUMEN

One promising approach for the gene therapy of Duchenne muscular dystrophy (DMD) is exon skipping. When thinking of possible intervention on human, it is very crucial to identify the most appropriate antisense sequences able to provide the highest possible skipping efficiency. In this article, we compared the exon 51 skipping activity of 10 different antisense molecules, raised against splice junctions and/or exonic splicing enhancers (ESEs), expressed as part of the U1 small nuclear RNA (snRNA). The effectiveness of each construct was tested in human DMD myoblasts carrying the deletion of exons 48-50, which can be treated with skipping of exon 51. Our results show that the highest skipping activity and dystrophin rescue is achieved upon expression of a U1 snRNA-derived antisense molecule targeting exon 51 splice sites in combination with an internal exon sequence. The efficacy of this molecule was further proven on an exon 45-50 deletion background, utilizing patient's fibroblasts transdifferentiated into myoblasts. In this system, we showed that the selected antisense was able to produce 50% skipping of exon 51.


Asunto(s)
Distrofina/genética , Exones/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , ARN sin Sentido/genética , ARN Nuclear Pequeño/genética , Animales , Northern Blotting , Western Blotting , Línea Celular , Células Cultivadas , Distrofina/metabolismo , Humanos , Ratones , Reacción en Cadena de la Polimerasa
15.
iScience ; 24(3): 102151, 2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33585804

RESUMEN

Detailed knowledge of the molecular biology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is crucial for understanding of viral replication, host responses, and disease progression. Here, we report gene expression profiles of three SARS-CoV- and SARS-CoV-2-infected human cell lines. SARS-CoV-2 elicited an approximately two-fold higher stimulation of the innate immune response compared to SARS-CoV in the human epithelial cell line Calu-3, including induction of miRNA-155. Single-cell RNA sequencing of infected cells showed that genes induced by virus infections were broadly upregulated, whereas interferon beta/lambda genes, a pro-inflammatory cytokines such as IL-6, were expressed only in small subsets of infected cells. Temporal analysis suggested that transcriptional activities of interferon regulatory factors precede those of nuclear factor κB. Lastly, we identified heat shock protein 90 (HSP90) as a protein relevant for the infection. Inhibition of the HSP90 activity resulted in a reduction of viral replication and pro-inflammatory cytokine expression in primary human airway epithelial cells.

16.
Mol Neurobiol ; 57(5): 2301-2313, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32020500

RESUMEN

Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition with unknown etiology. Recent experimental evidences suggest the contribution of non-coding RNAs (ncRNAs) in the pathophysiology of ASD. In this work, we aimed to investigate the expression profile of the ncRNA class of circular RNAs (circRNAs) in the hippocampus of the BTBR T + tf/J (BTBR) mouse model and age-matched C57BL/6J (B6) mice. Alongside, we analyzed BTBR hippocampal gene expression profile to evaluate possible correlations between the differential abundance of circular and linear gene products. From RNA sequencing data, we identified circRNAs highly modulated in BTBR mice. Thirteen circRNAs and their corresponding linear isoforms were validated by RT-qPCR analysis. The BTBR-regulated circCdh9 was better characterized in terms of molecular structure and expression, highlighting altered levels not only in the hippocampus, but also in the cerebellum, prefrontal cortex, and amygdala. Finally, gene expression analysis of the BTBR hippocampus pinpointed altered biological and molecular pathways relevant for the ASD phenotype. By comparison of circRNA and gene expression profiles, we identified 6 genes significantly regulated at either circRNA or mRNA gene products, suggesting low overall correlation between circRNA and host gene expression. In conclusion, our results indicate a consistent deregulation of circRNA expression in the hippocampus of BTBR mice. ASD-related circRNAs should be considered in functional studies to identify their contribution to the etiology of the disorder. In addition, as abundant and highly stable molecules, circRNAs represent interesting potential biomarkers for autism.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones Endogámicos/metabolismo , Ratones Mutantes/metabolismo , ARN Circular/biosíntesis , ARN Mensajero/biosíntesis , Animales , Trastorno del Espectro Autista/genética , Química Encefálica , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos/genética , Ratones Mutantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie
17.
Oncogene ; 38(20): 3843-3854, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30670781

RESUMEN

Circular RNAs (circRNAs) represent a class of covalently closed RNAs, derived from non-canonical splicing events, which are expressed in all eukaryotes and often conserved among different species. We previously showed that the circRNA originating from the ZNF609 locus (circ-ZNF609) acts as a crucial regulator of human primary myoblast growth: indeed, the downregulation of the circRNA, and not of its linear counterpart, strongly reduced the proliferation rate of in vitro cultured myoblasts. To deepen our knowledge about circ-ZNF609 role in cell cycle regulation, we studied its expression and function in rhabdomyosarcoma (RMS), a pediatric skeletal muscle malignancy. We found that circ-ZNF609 is upregulated in biopsies from the two major RMS subtypes, embryonal (ERMS) and alveolar (ARMS). Moreover, we discovered that in an ERMS-derived cell line circ-ZNF609 knock-down induced a specific block at the G1-S transition, a strong decrease of p-Akt protein level and an alteration of the pRb/Rb ratio. Regarding p-Akt, we were able to show that circ-ZNF609 acts by counteracting p-Akt proteasome-dependent degradation, thus working as a new regulator of cell proliferation-related pathways. As opposed to ERMS-derived cells, the circRNA depletion had no cell cycle effects in ARMS-derived cells. Since in these cells the p53 gene resulted downregulated, with a concomitant upregulation of its cell cycle-related target genes, we suggest that this could account for the lack of circ-ZNF609 effect in ARMS.


Asunto(s)
ARN/genética , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , ARN Circular , Rabdomiosarcoma/inmunología , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/inmunología , Rabdomiosarcoma Alveolar/patología , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/inmunología , Rabdomiosarcoma Embrionario/patología , Regulación hacia Arriba
18.
Nat Commun ; 8: 14741, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28358055

RESUMEN

The RNA-binding protein FUS participates in several RNA biosynthetic processes and has been linked to the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. Here we report that FUS controls back-splicing reactions leading to circular RNA (circRNA) production. We identified circRNAs expressed in in vitro-derived mouse motor neurons (MNs) and determined that the production of a considerable number of these circRNAs is regulated by FUS. Using RNAi and overexpression of wild-type and ALS-associated FUS mutants, we directly correlate the modulation of circRNA biogenesis with alteration of FUS nuclear levels and with putative toxic gain of function activities. We also demonstrate that FUS regulates circRNA biogenesis by binding the introns flanking the back-splicing junctions and that this control can be reproduced with artificial constructs. Most circRNAs are conserved in humans and specific ones are deregulated in human-induced pluripotent stem cell-derived MNs carrying the FUSP525L mutation associated with ALS.


Asunto(s)
Neuronas Motoras/metabolismo , Células Madre Embrionarias de Ratones/citología , Proteína FUS de Unión a ARN/metabolismo , ARN/genética , Animales , Diferenciación Celular , Exones/genética , Eliminación de Gen , Regulación de la Expresión Génica , Intrones/genética , Ratones , Mutación/genética , Unión Proteica/genética , ARN/biosíntesis , ARN/metabolismo , Empalme del ARN/genética , ARN Circular , Análisis de Secuencia de ARN , Médula Espinal/citología
19.
Oncotarget ; 8(19): 31003-31015, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28415684

RESUMEN

Long noncoding RNAs (lncRNAs) are major regulators of physiological and disease-related gene expression, particularly in the central nervous system. Dysregulated lncRNA expression has been documented in several human cancers, and their tissue-specificity makes them attractive candidates as diagnostic/prognostic biomarkers and/or therapeutic agents. Here we show that linc-NeD125, which we previously characterized as a neuronal-induced lncRNA, is significantly overexpressed in Group 4 medulloblastomas (G4 MBs), the largest and least well characterized molecular MB subgroup. Mechanistically, linc-NeD125 is able to recruit the miRNA-induced silencing complex (miRISC) and to directly bind the microRNAs miR-19a-3p, miR-19b-3p and miR-106a-5p. Functionally, linc-NeD125 acts as a competing endogenous RNA (ceRNA) that, sequestering the three miRNAs, leads to de-repression of their targets CDK6, MYCN, SNCAIP, and KDM6A, which are major driver genes of G4 MB. Accordingly, linc-NeD125 downregulation reduces G4 cell proliferation. Moreover, we also provide evidence that linc-NeD125 ectopic expression in the aggressive Group 3 MB cells attenuates their proliferation, migration and invasion.This study unveils the first lncRNA-based ceRNA network in central nervous system tumours and provides a novel molecular circuit underlying the enigmatic Group 4 medulloblastoma.


Asunto(s)
Neoplasias Cerebelosas/genética , Regulación Neoplásica de la Expresión Génica , Meduloblastoma/genética , MicroARNs/genética , Interferencia de ARN , ARN Largo no Codificante/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Cerebelosas/patología , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Meduloblastoma/patología
20.
Oncotarget ; 7(37): 60155-60168, 2016 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-27517498

RESUMEN

Alterations in genetic programs required for terminal myeloid differentiation and aberrant proliferation characterize acute myeloid leukemia (AML) cells. Here, we identify the host transcript of miR-223, linc-223, as a novel functional long non-coding RNA (lncRNA) in AML. We show that from the primary nuclear transcript, the alternative production of miR-223 and linc-223 is finely regulated during monocytic differentiation. Moreover, linc-223 expression inhibits cell cycle progression and promotes monocytic differentiation of AML cells. We also demonstrate that endogenous linc-223 localizes in the cytoplasm and acts as a competing endogenous RNA for miR-125-5p, an oncogenic microRNA in leukemia. In particular, we show that linc-223 directly binds to miR-125-5p and that its knockdown increases the repressing activity of miR-125-5p resulting in the downregulation of its target interferon regulatory factor 4 (IRF4), which it was previously shown to inhibit the oncogenic activity of miR-125-5p in vivo. Furthermore, data from primary AML samples show significant downregulation of linc-223 in different AML subtypes. Therein, these findings indicate that the newly identified lncRNA linc-223 may have an important role in myeloid differentiation and leukemogenesis, at least in part, by cross-talking with IRF4 mRNA.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Factores Reguladores del Interferón/genética , Leucemia Mieloide Aguda/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Adulto , Anciano , Diferenciación Celular/genética , Femenino , Perfilación de la Expresión Génica/métodos , Células HL-60 , Humanos , Factores Reguladores del Interferón/metabolismo , Células K562 , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA