Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nucleic Acids Res ; 51(10): 5193-5209, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37070602

RESUMEN

The long non-coding RNA EPR is expressed in epithelial tissues, binds to chromatin and controls distinct biological activities in mouse mammary gland cells. Because of its high expression in the intestine, in this study we have generated a colon-specific conditional targeted deletion (EPR cKO) to evaluate EPR in vivo functions in mice. EPR cKO mice display epithelium hyperproliferation, impaired mucus production and secretion, as well as inflammatory infiltration in the proximal portion of the large intestine. RNA sequencing analysis reveals a rearrangement of the colon crypt transcriptome with strong reduction of goblet cell-specific factors including those involved in the synthesis, assembly, transport and control of mucus proteins. Further, colon mucosa integrity and permeability are impaired in EPR cKO mice, and this results in higher susceptibility to dextran sodium sulfate (DSS)-induced colitis and tumor formation. Human EPR is down-regulated in human cancer cell lines as well as in human cancers, and overexpression of EPR in a colon cancer cell line results in enhanced expression of pro-apoptotic genes. Mechanistically, we show that EPR directly interacts with select genes involved in mucus metabolism whose expression is reduced in EPR cKO mice and that EPR deletion causes tridimensional chromatin organization changes.


Asunto(s)
Transformación Celular Neoplásica , Inflamación , Moco , ARN Largo no Codificante , Animales , Humanos , Ratones , Transformación Celular Neoplásica/inmunología , Colon/metabolismo , Modelos Animales de Enfermedad , Inflamación/inmunología , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
2.
Nucleic Acids Res ; 50(13): 7608-7622, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35748870

RESUMEN

EPR is a long non-coding RNA (lncRNA) that controls cell proliferation in mammary gland cells by regulating gene transcription. Here, we report on Mettl7a1 as a direct target of EPR. We show that EPR induces Mettl7a1 transcription by rewiring three-dimensional chromatin interactions at the Mettl7a1 locus. Our data indicate that METTL7A1 contributes to EPR-dependent inhibition of TGF-ß signaling. METTL7A1 is absent in tumorigenic murine mammary gland cells and its human ortholog (METTL7A) is downregulated in breast cancers. Importantly, re-expression of METTL7A1 in 4T1 tumorigenic cells attenuates their transformation potential, with the putative methyltransferase activity of METTL7A1 being dispensable for its biological functions. We found that METTL7A1 localizes in the cytoplasm whereby it interacts with factors implicated in the early steps of mRNA translation, associates with ribosomes, and affects the levels of target proteins without altering mRNA abundance. Overall, our data indicates that METTL7A1-a transcriptional target of EPR-modulates translation of select transcripts.


Asunto(s)
Neoplasias de la Mama , Metiltransferasas/metabolismo , ARN Largo no Codificante , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Cromatina/genética , Femenino , Humanos , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ribosomas/metabolismo
3.
Hematol Oncol ; 40(1): 40-47, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34679195

RESUMEN

Long non-coding RNAs are emerging as essential regulators of gene expression, but their role in normal and neoplastic B cells is still largely uncharacterized. Here, we report on the expression pattern of the LINC00152 in normal B cells and Chronic Lymphocytic Leukemia B cell clones. Higher LINC00152 levels were consistently observed in memory B cell populations when compared to naïve B cells in the normal tissues analyzed [peripheral blood (PB), tonsils, and spleen]. In addition, independent stimulation via Immunoglobulins (IG), CD40, or Toll-like Receptor 9 (TLR9) upregulated LINC00152 in PB B cells. The expression of LINC00152 in a cohort of 107 early stage Binet A CLL patients was highly variable and did not correlate with known prognostic markers or clinical evolution. TLR9 stimulation, but not CD40 or IG challenge, was able to upregulate LINC00152 expression in CLL cells. In addition, LINC00152 silencing in CLL cell lines expressing LINC00152 failed to induce significant cell survival or apoptosis changes. These data suggest that, in normal B cells, the expression of LINC00152 is regulated by immunomodulatory signals, which are only partially effective in CLL cells. However, LINC00152 does not appear to contribute to CLL cell expansion and/or survival in a cohort of newly diagnosed CLL patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Tonsila Palatina/metabolismo , ARN Largo no Codificante/metabolismo , Bazo/metabolismo , Biomarcadores de Tumor/genética , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Pronóstico , Estudios Prospectivos , ARN Largo no Codificante/genética , Tasa de Supervivencia
4.
Nucleic Acids Res ; 48(16): 9053-9066, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32756918

RESUMEN

Long non-coding RNAs (lncRNAs) can affect multiple layers of gene expression to control crucial cellular functions. We have previously demonstrated that the lncRNA EPR, by controlling gene expression at different levels, affects cell proliferation and migration in cultured mammary gland cells and impairs breast tumor formation in an orthotopic transplant model in mice. Here, we used ChIRP-Seq to identify EPR binding sites on chromatin of NMuMG mammary gland cells overexpressing EPR and identified its trans binding sites in the genome. Then, with the purpose of relating EPR/chromatin interactions to the reshaping of the epitranscriptome landscape, we profiled histone activation marks at promoter/enhancer regions by ChIP-Seq. Finally, we integrated data derived from ChIRP-Seq, ChIP-Seq as well as RNA-Seq in a comprehensive analysis and we selected a group of bona fide direct transcriptional targets of EPR. Among them, we identified a subset of EPR targets whose expression is controlled by TGF-ß with one of them-Arrdc3-being able to modulate Epithelial to Mesenchymal Transition. This experimental framework allowed us to correlate lncRNA/chromatin interactions with the real outcome of gene expression and to start defining the gene network regulated by EPR as a component of the TGF-ß pathway.


Asunto(s)
Arrestinas/genética , Neoplasias de la Mama/genética , ARN Largo no Codificante/genética , Factor de Crecimiento Transformador beta/genética , Animales , Sitios de Unión/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Cromatina/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Transcriptoma/genética
5.
Proc Natl Acad Sci U S A ; 111(47): E5023-8, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25385579

RESUMEN

Long noncoding RNAs (lncRNAs) interact with protein factors to regulate different layers of gene expression transcriptionally or posttranscriptionally. Here we report on the functional consequences of the unanticipated interaction of the RNA binding protein K homology-type splicing regulatory protein (KSRP) with the H19 lncRNA (H19). KSRP directly binds to H19 in the cytoplasm of undifferentiated multipotent mesenchymal C2C12 cells, and this interaction favors KSRP-mediated destabilization of labile transcripts such as myogenin. AKT activation induces KSRP dismissal from H19 and, as a consequence, myogenin mRNA is stabilized while KSRP is repurposed to promote maturation of myogenic microRNAs, thus favoring myogenic differentiation. Our data indicate that H19 operates as a molecular scaffold that facilitates effective association of KSRP with myogenin and other labile transcripts, and we propose that H19 works with KSRP to optimize an AKT-regulated posttranscriptional switch that controls myogenic differentiation.


Asunto(s)
ARN Largo no Codificante/fisiología , ARN Mensajero/metabolismo , Animales , Línea Celular , Humanos , Unión Proteica , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Transactivadores/metabolismo
6.
Semin Cell Dev Biol ; 34: 2-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24845017

RESUMEN

The single-strand-RNA binding protein KSRP is able to negatively regulate gene expression operating with at least two distinct and integrated postranscriptional mechanisms: (i) by promoting decay of unstable mRNAs and (ii) by favoring maturation from precursors of select microRNAs (miRNAs) including the prototypical tumor suppressor let-7. Studies performed in primary and cultured cells as well as in mice proved that the ability of KSRP to integrate different levels of gene expression is required for proper immune response, lipid metabolism, cell-fate decisions, tissue regeneration, and DNA damage response.


Asunto(s)
Proteínas de Unión al ARN/fisiología , Transactivadores/fisiología , Animales , Diferenciación Celular , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Genes MHC Clase II , Humanos , Metabolismo de los Lípidos , MicroARNs/genética , MicroARNs/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
Nature ; 459(7249): 1010-4, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19458619

RESUMEN

Consistent with the role of microRNAs (miRNAs) in down-regulating gene expression by reducing the translation and/or stability of target messenger RNAs, the levels of specific miRNAs are important for correct embryonic development and have been linked to several forms of cancer. However, the regulatory mechanisms by which primary miRNAs (pri-miRNAs) are processed first to precursor miRNAs (pre-miRNAs) and then to mature miRNAs by the multiprotein Drosha and Dicer complexes, respectively, remain largely unknown. The KH-type splicing regulatory protein (KSRP, also known as KHSRP) interacts with single-strand AU-rich-element-containing mRNAs and is a key mediator of mRNA decay. Here we show in mammalian cells that KSRP also serves as a component of both Drosha and Dicer complexes and regulates the biogenesis of a subset of miRNAs. KSRP binds with high affinity to the terminal loop of the target miRNA precursors and promotes their maturation. This mechanism is required for specific changes in target mRNA expression that affect specific biological programs, including proliferation, apoptosis and differentiation. These findings reveal an unexpected mechanism that links KSRP to the machinery regulating maturation of a cohort of miRNAs that, in addition to its role in promoting mRNA decay, independently serves to integrate specific regulatory programs of protein expression.


Asunto(s)
MicroARNs/biosíntesis , Proteínas de Unión al ARN/metabolismo , Transactivadores/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , Ribonucleasa III/química , Ribonucleasa III/metabolismo
8.
PLoS Genet ; 8(7): e1002823, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22844247

RESUMEN

Gene silencing mediated by either microRNAs (miRNAs) or Adenylate/uridylate-rich elements Mediated mRNA Degradation (AMD) is a powerful way to post-transcriptionally modulate gene expression. We and others have reported that the RNA-binding protein KSRP favors the biogenesis of select miRNAs (including let-7 family) and activates AMD promoting the decay of inherently labile mRNAs. Different layers of interplay between miRNA- and AMD-mediated gene silencing have been proposed in cultured cells, but the relationship between the two pathways in living organisms is still elusive. We conditionally deleted Dicer in mouse pituitary from embryonic day (E) 9.5 through Cre-mediated recombination. In situ hybridization, immunohistochemistry, and quantitative reverse transcriptase-PCR revealed that Dicer is essential for pituitary morphogenesis and correct expression of hormones. Strikingly, αGSU (alpha glycoprotein subunit, common to three pituitary hormones) was absent in Dicer-deleted pituitaries. αGSU mRNA is unstable and its half-life increases during pituitary development. A transcriptome-wide analysis of microdissected E12.5 pituitaries revealed a significant increment of KSRP expression in conditional Dicer-deleted mice. We found that KSRP directly binds to αGSU mRNA, promoting its rapid decay; and, during pituitary development, αGSU expression displays an inverse temporal relationship to KSRP. Further, let-7b/c downregulated KSRP expression, promoting the degradation of its mRNA by directly binding to the 3'UTR. Therefore, we propose a model in which let-7b/c and KSRP operate within a negative feedback loop. Starting from E12.5, KSRP induces the maturation of let-7b/c that, in turn, post-transcriptionally downregulates the expression of KSRP itself. This event leads to stabilization of αGSU mRNA, which ultimately enhances the steady-state expression levels. We have identified a post-transcriptional regulatory network active during mouse pituitary development in which the expression of the hormone αGSU is increased by let7b/c through downregulation of KSRP. Our study unveils a functional crosstalk between miRNA- and AMD-dependent gene regulation during mammalian organogenesis events.


Asunto(s)
MicroARNs/genética , Organogénesis/genética , Hipófisis , ARN Mensajero , Proteínas de Unión al ARN/genética , Transactivadores/genética , Animales , ARN Helicasas DEAD-box/genética , Desarrollo Embrionario/genética , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Hormonas Glicoproteicas de Subunidad alfa/genética , Hormonas Glicoproteicas de Subunidad alfa/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , MicroARNs/metabolismo , Células 3T3 NIH , Hipófisis/embriología , Hipófisis/crecimiento & desarrollo , Hipófisis/metabolismo , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/metabolismo , Estabilidad del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/genética , Transactivadores/metabolismo
9.
Biochim Biophys Acta ; 1829(5): 469-79, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23462617

RESUMEN

Understanding the molecular mechanisms that control the balance between multipotency and differentiation is of great importance to elucidate the genesis of both developmental disorders and cell transformation events. To investigate the role of the RNA binding protein KSRP in controlling neural differentiation, we used the P19 embryonal carcinoma cell line that is able to differentiate into neuron-like cells under appropriate culture conditions. We have recently reported that KSRP controls the differentiative fate of multipotent mesenchymal cells owing to its ability to promote decay of unstable transcripts and to favor maturation of selected micro-RNAs (miRNAs) from precursors. Here we report that KSRP silencing in P19 cells favors neural differentiation increasing the expression of neuronal markers. Further, the expression of two master transcriptional regulators of neurogenesis, ASCL1 and JMJD3, was enhanced while the maturation of miR-200 family members from precursors was impaired in KSRP knockdown cells. These molecular changes can contribute to the reshaping of P19 cells transcriptome that follows KSRP silencing. Our data suggests that KSRP function is required to maintain P19 cells in a multipotent undifferentiated state and that its inactivation can orient cells towards neural differentiation.


Asunto(s)
Silenciador del Gen , Neurogénesis/genética , Proteínas de Unión al ARN/genética , Transactivadores/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , MicroARNs/metabolismo , Precursores del ARN/metabolismo , Estabilidad del ARN , Proteínas de Unión al ARN/metabolismo , Teratocarcinoma , Transactivadores/metabolismo , Transcripción Genética , Transcriptoma
10.
Biochim Biophys Acta ; 1829(6-7): 689-94, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23178464

RESUMEN

KSRP is a single strand nucleic acid binding protein that controls gene expression at multiple levels. In this review we focus on the recent molecular, cellular, and structural insights into the mRNA decay promoting function of KSRP. We discuss also some aspects of KSRP-dependent microRNA maturation from precursors that are related to its mRNA destabilizing function. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Asunto(s)
MicroARNs/genética , Estabilidad del ARN/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Transactivadores/genética , Sitios de Unión , Línea Celular , Regulación de la Expresión Génica , Humanos , Proteínas de Unión al ARN/metabolismo
12.
Nucleic Acids Res ; 40(14): 6873-86, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22547390

RESUMEN

In eukaryotes, RNA-binding proteins that contain multiple K homology (KH) domains play a key role in coordinating the different steps of RNA synthesis, metabolism and localization. Understanding how the different KH modules participate in the recognition of the RNA targets is necessary to dissect the way these proteins operate. We have designed a KH mutant with impaired RNA-binding capability for general use in exploring the role of individual KH domains in the combinatorial functional recognition of RNA targets. A double mutation in the hallmark GxxG loop (GxxG-to-GDDG) impairs nucleic acid binding without compromising the stability of the domain. We analysed the impact of the GDDG mutations in individual KH domains on the functional properties of KSRP as a prototype of multiple KH domain-containing proteins. We show how the GDDG mutant can be used to directly link biophysical information on the sequence specificity of the different KH domains of KSRP and their role in mRNA recognition and decay. This work defines a general molecular biology tool for the investigation of the function of individual KH domains in nucleic acid binding proteins.


Asunto(s)
Dominios y Motivos de Interacción de Proteínas/genética , Proteínas de Unión al ARN/química , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Mutación , ARN/química , ARN/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transactivadores/química , Transactivadores/metabolismo
14.
Nucleic Acids Res ; 39(17): 7576-85, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21705779

RESUMEN

Alternative pre-mRNA splicing (AS) is a major mechanism that allows proteomic variability in eukaryotic cells. However, many AS events result in mRNAs containing a premature termination codon, which are degraded by nonsense-mediated mRNA decay (NMD) pathway. We have previously demonstrated that human rpL3 autoregulates its expression through the association of AS with NMD. In fact, overexpression of rpL3 promotes downregulation of canonical splicing and upregulation of alternative splicing that produces an NMD-targeted mRNA isoform. The result of these events is a decreased production of rpL3. We have also identified heterogeneous nuclear ribonucleoprotein (hnRNP) H1 as a splicing factor involved in the regulation of rpL3 alternative splicing and identified its regulatory cis-elements within intron 3 transcript. Here, we report that NPM and KHSRP are two newly identified proteins involved in the regulation of rpL3 gene expression via AS-NMD. We demonstrate that hnRNP H1, KHSRP and NPM can be found associated, and present also in ribonucleoproteins (RNPs) including rpL3 and intron 3 RNA in vivo, and describe protein-protein and RNA-protein interactions. Moreover, our data provide an insight on the crucial role of hnRNP H1 in the regulation of the alternative splicing of the rpL3 gene.


Asunto(s)
Empalme Alternativo , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Ribosómicas/genética , Transactivadores/metabolismo , Animales , Línea Celular , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/fisiología , Homeostasis , Humanos , Intrones , Proteínas Nucleares/fisiología , Nucleofosmina , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/fisiología , Ratas , Proteína Ribosomal L3 , Proteínas Ribosómicas/metabolismo , Transactivadores/fisiología
15.
Nucleic Acids Res ; 38(15): 5193-205, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20385598

RESUMEN

KSRP is a multi-domain RNA-binding protein that recruits the exosome-containing mRNA degradation complex to mRNAs coding for cellular proliferation and inflammatory response factors. The selectivity of this mRNA degradation mechanism relies on KSRP recognition of AU-rich elements in the mRNA 3'UTR, that is mediated by KSRP's KH domains. Our structural analysis shows that the inter-domain linker orients the two central KH domains of KSRP-and their RNA-binding surfaces-creating a two-domain unit. We also show that this inter-domain arrangement is important to the interaction with KSRP's RNA targets.


Asunto(s)
Regiones no Traducidas 3' , Proteínas de Unión al ARN/química , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , ARN/química , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Transactivadores/química , Transactivadores/metabolismo
16.
Adv Exp Med Biol ; 700: 36-42, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21755471

RESUMEN

microrNNA (mirNAs) are small noncoding RNAs that down-regulate gene expression by reducing stability and/or translation of target mRNAs. In animals, miRNAs arise from sequential processing of hairpin primary transcripts by two rNase III domain-containing enzymes, namely Drosha and Dicer, to generate a mature form of about 22 nucleotides. In this chapter we discuss our latest fndings indicating that KSRP is an integral component of both Drosha and Dicer complexes. KSRP binds to the terminal loop sequence of a subset of miRNA precursors promoting their maturation. our data indicate that the terminal loop is a pivotal structure where activators of miRNA processing as well as repressors of miRNA processing act in a coordinated way to convert cellular signals into changes in miRNA expression processing. This uncovers a new level of complexity of miRNA mechanisms for gene expression regulation.


Asunto(s)
MicroARNs , ARN Mensajero , Animales , Expresión Génica , Regulación de la Expresión Génica , Humanos , MicroARNs/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética
17.
FASEB J ; 23(9): 2898-908, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19423639

RESUMEN

The importance of post-transcriptional mechanisms for the regulation of the homoeostasis of the immune system and the response to challenge by microorganisms is becoming increasingly appreciated. We investigated the contribution of microRNAs (miRNAs) to macrophage activation induced by lipopolysaccharide (LPS). We first observed that Dicer knockout in bone marrow-derived macrophages (BMDMs) increases the LPS-induced expression of some inflammation mediators. miRNA microarray analysis in BMDMs revealed that LPS significantly induces the expression of a single miRNA, miR-155, and this induction depends on enhanced miR-155 maturation from its precursors. The single-strand RNA-binding protein KH-type splicing regulatory protein (KSRP) binds to the terminal loop of miR-155 precursors and promotes their maturation. Both inhibition of miR-155 and KSRP knockdown enhance the LPS-induced expression of select inflammation mediators, and the effect of KSRP knockdown is reverted by mature miR-155. Our studies unveil the existence of an LPS-dependent post-transcriptional regulation of miR-155 biogenesis. Once induced, miR-155 finely tunes the expression of select inflammation mediators in response to LPS.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Transactivadores/metabolismo , Animales , ARN Helicasas DEAD-box/deficiencia , Endorribonucleasas/deficiencia , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Procesamiento Postranscripcional del ARN , Ribonucleasa III , Activación Transcripcional
18.
Adv Exp Med Biol ; 700: 36-42, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21627028

RESUMEN

microRNAs (miRNAs) are small noncodingRNAs that down-regulate gene expression by reducing stability and/or translation of target mRNAs. In animals, miRNAs arise from sequential processing of hairpin primary transcripts by two RNAse III domain-containing enzymes, namely Drosha and Dicer, to generate a mature form of about 22 nucleotides. In this chapter we discuss our latest findings indicating that KSRP is an integral component of both Drosha and Dicer complexes. KSRP binds to the terminal loop sequence of a subset of miRNA precursors promoting their maturation. Our data indicate that the terminal loop is a pivotal structure where activators of miRNA processing as well as repressors of miRNA processing act in a coordinated way to convert cellular signals into changes in miRNA expression processing. This uncovers a new level of complexity of miRNA mechanisms for gene expression regulation.


Asunto(s)
MicroARNs/fisiología , Precursores del ARN/fisiología , Proteínas de Unión al ARN/fisiología , Transactivadores/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Proteínas Co-Represoras/fisiología , Humanos , Neoplasias/etiología
19.
Noncoding RNA ; 6(3)2020 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-32957640

RESUMEN

Although mammals possess roughly the same number of protein-coding genes as worms, it is evident that the non-coding transcriptome content has become far broader and more sophisticated during evolution. Indeed, the vital regulatory importance of both short and long non-coding RNAs (lncRNAs) has been demonstrated during the last two decades. RNA binding proteins (RBPs) represent approximately 7.5% of all proteins and regulate the fate and function of a huge number of transcripts thus contributing to ensure cellular homeostasis. Transcriptomic and proteomic studies revealed that RBP-based complexes often include lncRNAs. This review will describe examples of how lncRNA-RBP networks can virtually control all the post-transcriptional events in the cell.

20.
BMC Cell Biol ; 10: 70, 2009 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-19775426

RESUMEN

BACKGROUND: Parathyroid hormone (PTH) gene expression is regulated post-transcriptionally through the binding of the trans-acting proteins AU rich binding factor 1 (AUF1), Upstream of N-ras (Unr) and KH-type splicing regulatory protein (KSRP) to an AU rich element (ARE) in PTH mRNA 3'-UTR. AUF1 and Unr stabilize PTH mRNA while KSRP, recruiting the exoribonucleolytic complex exosome, promotes PTH mRNA decay. RESULTS: PTH mRNA is cleaved by the endoribonuclease polysomal ribonuclease 1 (PMR1) in an ARE-dependent manner. Moreover, PMR1 co-immunoprecipitates with PTH mRNA, the exosome and KSRP. Knock-down of either exosome components or KSRP by siRNAs prevents PMR1-mediated cleavage of PTH mRNA. CONCLUSION: PTH mRNA is a target for the endonuclease PMR1. The PMR1 mediated decrease in PTH mRNA levels involves the PTH mRNA 3'-UTR ARE, KSRP and the exosome. This represents an unanticipated mechanism by which the decay of an ARE-containing mRNA is facilitated by KSRP and is dependent on both the exosome and an endoribonuclease.


Asunto(s)
ATPasas Transportadoras de Calcio/metabolismo , Exosomas/metabolismo , Hormona Paratiroidea/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transactivadores/metabolismo , Regiones no Traducidas 3' , Línea Celular , Humanos , Hormona Paratiroidea/genética , ARN Mensajero/genética , ARN Interferente Pequeño/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA