Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 165(2): 382-95, 2016 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-27040500

RESUMEN

Gene duplication is a major evolutionary force driving adaptation and speciation, as it allows for the acquisition of new functions and can augment or diversify existing functions. Here, we report a gene duplication event that yielded another outcome--the generation of antagonistic functions. One product of this duplication event--UPF3B--is critical for the nonsense-mediated RNA decay (NMD) pathway, while its autosomal counterpart--UPF3A--encodes an enigmatic protein previously shown to have trace NMD activity. Using loss-of-function approaches in vitro and in vivo, we discovered that UPF3A acts primarily as a potent NMD inhibitor that stabilizes hundreds of transcripts. Evidence suggests that UPF3A acquired repressor activity through simple impairment of a critical domain, a rapid mechanism that may have been widely used in evolution. Mice conditionally lacking UPF3A exhibit "hyper" NMD and display defects in embryogenesis and gametogenesis. Our results support a model in which UPF3A serves as a molecular rheostat that directs developmental events.


Asunto(s)
Desarrollo Embrionario , Genes Duplicados , Degradación de ARNm Mediada por Codón sin Sentido , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Evolución Molecular , Gametogénesis , Células HeLa , Humanos , Ratones
2.
Development ; 149(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36255229

RESUMEN

Nonsense-mediated RNA decay (NMD) is a highly conserved RNA turnover pathway that degrades RNAs harboring in-frame stop codons in specific contexts. Loss of NMD factors leads to embryonic lethality in organisms spanning the phylogenetic scale, but the mechanism remains unknown. Here, we report that the core NMD factor, UPF2, is required for expansion of epiblast cells within the inner cell mass of mice in vivo. We identify NMD target mRNAs in mouse blastocysts - both canonical and alternatively processed mRNAs - including those encoding cell cycle arrest and apoptosis factors, raising the possibility that NMD is essential for embryonic cell proliferation and survival. In support, the inner cell mass of Upf2-null blastocysts rapidly regresses with outgrowth and is incompetent for embryonic stem cell derivation in vitro. In addition, we uncovered concordant temporal- and lineage-specific regulation of NMD factors and mRNA targets, indicative of a shift in NMD magnitude during peri-implantation development. Together, our results reveal developmental and molecular functions of the NMD pathway in the early embryo.


Asunto(s)
Degradación de ARNm Mediada por Codón sin Sentido , ARN , Ratones , Animales , ARN/metabolismo , Filogenia , Degradación de ARNm Mediada por Codón sin Sentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estratos Germinativos/metabolismo , Proteínas de Unión al ARN/metabolismo
3.
Nucleic Acids Res ; 50(13): 7310-7325, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35776114

RESUMEN

The nuanced mechanisms driving primordial germ cells (PGC) specification remain incompletely understood since genome-wide transcriptional regulation in developing PGCs has previously only been defined indirectly. Here, using SLAMseq analysis, we determined genome-wide transcription rates during the differentiation of embryonic stem cells (ESCs) to form epiblast-like (EpiLC) cells and ultimately PGC-like cells (PGCLCs). This revealed thousands of genes undergoing bursts of transcriptional induction and rapid shut-off not detectable by RNAseq analysis. Our SLAMseq datasets also allowed us to infer RNA turnover rates, which revealed thousands of mRNAs stabilized and destabilized during PGCLC specification. mRNAs tend to be unstable in ESCs and then are progressively stabilized as they differentiate. For some classes of genes, mRNA turnover regulation collaborates with transcriptional regulation, but these processes oppose each other in a surprisingly high frequency of genes. To test whether regulated mRNA turnover has a physiological role in PGC development, we examined three genes that we found were regulated by RNA turnover: Sox2, Klf2 and Ccne1. Circumvention of their regulated RNA turnover severely impaired the ESC-to-EpiLC and EpiLC-to-PGCLC transitions. Our study demonstrates the functional importance of regulated RNA stability in germline development and provides a roadmap of transcriptional and post-transcriptional regulation during germline specification.


Asunto(s)
Células Germinativas , ARN , Animales , Diferenciación Celular , Células Madre Embrionarias , Células Germinativas/citología , Estratos Germinativos , Ratones , Estabilidad del ARN , ARN Mensajero/genética , Transcripción Genética
4.
Nucleic Acids Res ; 50(20): 11470-11491, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36259644

RESUMEN

Nonsense-mediated RNA decay (NMD) is a highly conserved and selective RNA turnover pathway that depends on the endonuclease SMG6. Here, we show that SMG6 is essential for male germ cell differentiation in mice. Germ-cell conditional knockout (cKO) of Smg6 induces extensive transcriptome misregulation, including a failure to eliminate meiotically expressed transcripts in early haploid cells, and accumulation of NMD target mRNAs with long 3' untranslated regions (UTRs). Loss of SMG6 in the male germline results in complete arrest of spermatogenesis at the early haploid cell stage. We find that SMG6 is strikingly enriched in the chromatoid body (CB), a specialized cytoplasmic granule in male germ cells also harboring PIWI-interacting RNAs (piRNAs) and the piRNA-binding protein PIWIL1. This raises the possibility that SMG6 and the piRNA pathway function together, which is supported by several findings, including that Piwil1-KO mice phenocopy Smg6-cKO mice and that SMG6 and PIWIL1 co-regulate many genes in round spermatids. Together, our results demonstrate that SMG6 is an essential regulator of the male germline transcriptome, and highlight the CB as a molecular platform coordinating RNA regulatory pathways to control sperm production and fertility.


Asunto(s)
Endorribonucleasas , Gránulos de Ribonucleoproteína de Células Germinales , Espermatogénesis , Transcriptoma , Animales , Masculino , Ratones , Células Germinativas/metabolismo , ARN Interferente Pequeño/genética , Espermátides/metabolismo , Espermatogénesis/genética , Endorribonucleasas/metabolismo
5.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34083437

RESUMEN

Transposable elements (TEs) are mobile sequences that engender widespread mutations and thus are a major hazard that must be silenced. The most abundant active class of TEs in mammalian genomes is long interspersed element class 1 (LINE1). Here, we report that LINE1 transposition is suppressed in the male germline by transcription factors encoded by a rapidly evolving X-linked homeobox gene cluster. LINE1 transposition is repressed by many members of this RHOX transcription factor family, including those with different patterns of expression during spermatogenesis. One family member-RHOX10-suppresses LINE1 transposition during fetal development in vivo when the germline would otherwise be susceptible to LINE1 activation because of epigenetic reprogramming. We provide evidence that RHOX10 suppresses LINE transposition by inducing Piwil2, which encodes a key component in the Piwi-interacting RNA pathway that protects against TEs. The ability of RHOX transcription factors to suppress LINE1 is conserved in humans but is lost in RHOXF2 mutants from several infertile human patients, raising the possibility that loss of RHOXF2 causes human infertility by allowing uncontrolled LINE1 expression in the germline. Together, our results support a model in which the Rhox gene cluster is in an evolutionary arms race with TEs, resulting in expansion of the Rhox gene cluster to suppress TEs in different biological contexts.


Asunto(s)
Elementos Transponibles de ADN/genética , Células Germinativas/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Elementos de Nucleótido Esparcido Largo/fisiología , Familia de Multigenes , Animales , Regulación de la Expresión Génica , Genes Ligados a X , Células HEK293 , Proteínas de Homeodominio , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Espermatogénesis/genética , Factores de Transcripción/metabolismo
6.
Development ; 147(3)2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31964773

RESUMEN

Pro-spermatogonia (SG) serve as the gateway to spermatogenesis. Using single-cell RNA sequencing (RNAseq), we studied the development of ProSG, their SG descendants and testicular somatic cells during the perinatal period in mice. We identified both gene and protein markers for three temporally distinct ProSG cell subsets, including a migratory cell population with a transcriptome distinct from the previously defined T1- and T2-ProSG stages. This intermediate (I)-ProSG subset translocates from the center of seminiferous tubules to the spermatogonial stem cell (SSC) 'niche' in its periphery soon after birth. We identified three undifferentiated SG subsets at postnatal day 7, each of which expresses distinct genes, including transcription factor and signaling genes. Two of these subsets have the characteristics of newly emergent SSCs. We also molecularly defined the development of Sertoli, Leydig and peritubular myoid cells during the perinatal period, allowing us to identify candidate signaling pathways acting between somatic and germ cells in a stage-specific manner during the perinatal period. Our study provides a rich resource for those investigating testicular germ and somatic cell developmental during the perinatal period.


Asunto(s)
Células Germinativas/crecimiento & desarrollo , Células Intersticiales del Testículo/metabolismo , RNA-Seq/métodos , Células de Sertoli/metabolismo , Análisis de la Célula Individual/métodos , Espermatogonias/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Vía de Señalización Hippo , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Proteínas Serina-Treonina Quinasas/metabolismo , Espermatogénesis/genética , Nicho de Células Madre/fisiología , Factores de Transcripción/metabolismo , Transcriptoma
7.
Nat Rev Neurosci ; 19(12): 715-728, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30410025

RESUMEN

Steady-state RNA levels are controlled by the balance between RNA synthesis and RNA turnover. A selective RNA turnover mechanism that has received recent attention in neurons is nonsense-mediated RNA decay (NMD). NMD has been shown to influence neural development, neural stem cell differentiation decisions, axon guidance and synaptic plasticity. In humans, NMD factor gene mutations cause some forms of intellectual disability and are associated with neurodevelopmental disorders, including schizophrenia and autism spectrum disorder. Impairments in NMD are linked to neurodegenerative disorders, including amyotrophic lateral sclerosis. We discuss these findings, their clinical implications and challenges for the future.


Asunto(s)
Trastorno del Espectro Autista/patología , Encéfalo/metabolismo , Neurogénesis/fisiología , Degradación de ARNm Mediada por Codón sin Sentido/fisiología , Esquizofrenia/patología , Animales , Diferenciación Celular , Humanos
8.
Proc Natl Acad Sci U S A ; 117(30): 17832-17841, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32661178

RESUMEN

Spermatogonial stem cells (SSCs) are essential for the generation of sperm and have potential therapeutic value for treating male infertility, which afflicts >100 million men world-wide. While much has been learned about rodent SSCs, human SSCs remain poorly understood. Here, we molecularly characterize human SSCs and define conditions favoring their culture. To achieve this, we first identified a cell-surface protein, PLPPR3, that allowed purification of human primitive undifferentiated spermatogonia (uSPG) highly enriched for SSCs. Comparative RNA-sequencing analysis of these enriched SSCs with differentiating SPG (KIT+ cells) revealed the full complement of genes that shift expression during this developmental transition, including genes encoding key components in the TGF-ß, GDNF, AKT, and JAK-STAT signaling pathways. We examined the effect of manipulating these signaling pathways on cultured human SPG using both conventional approaches and single-cell RNA-sequencing analysis. This revealed that GDNF and BMP8B broadly support human SPG culture, while activin A selectively supports more advanced human SPG. One condition-AKT pathway inhibition-had the unique ability to selectively support the culture of primitive human uSPG. This raises the possibility that supplementation with an AKT inhibitor could be used to culture human SSCs in vitro for therapeutic applications.


Asunto(s)
Transducción de Señal , Espermatogonias/citología , Espermatogonias/metabolismo , Transcriptoma , Biomarcadores , Separación Celular , Células Cultivadas , Biología Computacional , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunofenotipificación , Masculino , Fosfatidato Fosfatasa/genética , Fosfatidato Fosfatasa/metabolismo
9.
Hum Mol Genet ; 29(15): 2568-2578, 2020 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-32667670

RESUMEN

Loss-of-function mutations of the X-chromosome gene UPF3B cause male neurodevelopmental disorders (NDDs) via largely unknown mechanisms. We investigated initially by interrogating a novel synonymous UPF3B variant in a male with absent speech. In silico and functional studies using cell lines derived from this individual show altered UPF3B RNA splicing. The resulting mRNA species encodes a frame-shifted protein with a premature termination codon (PTC) predicted to elicit degradation via nonsense-mediated mRNA decay (NMD). UPF3B mRNA was reduced in the cell line, and no UPF3B protein was produced, confirming a loss-of-function allele. UPF3B is itself involved in the NMD mechanism which degrades both PTC-bearing mutant transcripts and also many physiological transcripts. RNAseq analysis showed that ~1.6% of mRNAs exhibited altered expression. These mRNA changes overlapped and correlated with those we identified in additional cell lines obtained from individuals harbouring other UPF3B mutations, permitting us to interrogate pathogenic mechanisms of UPF3B-associated NDDs. We identified 102 genes consistently deregulated across all UPF3B mutant cell lines. Of the 51 upregulated genes, 75% contained an NMD-targeting feature, thus identifying high-confidence direct NMD targets. Intriguingly, 22 of the dysregulated genes encoded known NDD genes, suggesting UPF3B-dependent NMD regulates gene networks critical for cognition and behaviour. Indeed, we show that 78.5% of all NDD genes encode a transcript predicted to be targeted by NMD. These data describe the first synonymous UPF3B mutation in a patient with prominent speech and language disabilities and identify plausible mechanisms of pathology downstream of UPF3B mutations involving the deregulation of NDD-gene networks.


Asunto(s)
Codón sin Sentido/genética , Trastornos del Neurodesarrollo/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Trastornos del Habla/genética , Línea Celular , Preescolar , Redes Reguladoras de Genes/genética , Humanos , Lactante , Mutación con Pérdida de Función/genética , Masculino , Trastornos del Neurodesarrollo/patología , Degradación de ARNm Mediada por Codón sin Sentido/genética , Empalme del ARN/genética , Mutación Silenciosa/genética , Trastornos del Habla/patología
10.
EMBO Rep ; 20(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30573526

RESUMEN

Testis-expressed X-linked genes typically evolve rapidly. Here, we report on a testis-expressed X-linked microRNA (miRNA) cluster that despite rapid alterations in sequence has retained its position in the Fragile-X region of the X chromosome in placental mammals. Surprisingly, the miRNAs encoded by this cluster (Fx-mir) have a predilection for targeting the immediately adjacent gene, Fmr1, an unexpected finding given that miRNAs usually act in trans, not in cis Robust repression of Fmr1 is conferred by combinations of Fx-mir miRNAs induced in Sertoli cells (SCs) during postnatal development when they terminate proliferation. Physiological significance is suggested by the finding that FMRP, the protein product of Fmr1, is downregulated when Fx-mir miRNAs are induced, and that FMRP loss causes SC hyperproliferation and spermatogenic defects. Fx-mir miRNAs not only regulate the expression of FMRP, but also regulate the expression of eIF4E and CYFIP1, which together with FMRP form a translational regulatory complex. Our results support a model in which Fx-mir family members act cooperatively to regulate the translation of batteries of mRNAs in a developmentally regulated manner in SCs.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , MicroARNs/genética , Familia de Multigenes , Interferencia de ARN , ARN Mensajero/genética , Espermatogénesis/genética , Regiones no Traducidas 3' , Animales , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Testículo/metabolismo
11.
Nature ; 568(7751): 179-180, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30962551
12.
Mol Cell ; 45(3): 271-3, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22325347

RESUMEN

Capped and polyadenylated long noncoding RNAs (lncRNAs) are shown to be degraded by a DCP2-mediated turnover mechanism by Geisler et al. (2012); this provides a new level of regulatory control for inducible genes by lncRNAs.


Asunto(s)
Exorribonucleasas/genética , Regulación Fúngica de la Expresión Génica , Caperuzas de ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN no Traducido/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética
13.
Bioessays ; 40(1)2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29236296

RESUMEN

The RNA-binding protein, UPF1, is best known for its central role in the nonsense-mediated RNA decay (NMD) pathway. Feng et al. now report a new function for UPF1-it is an E3 ubiquitin ligase that specifically promotes the decay of a key pro-muscle transcription factor: MYOD. UPF1 achieves this through its RING-like domain, which confers ubiquitin E3 ligase activity. Feng et al. provide evidence that the ability of UPF1 to destabilize MYOD represses myogenesis. In the future, it will be important to define other protein substrates of UPF1-driven ubiquitination and to determine whether this biochemical activity is responsible for some of UPF1's previously defined biological functions, including in development and stress responses. The exciting findings presented by Feng et al. open up the possibility that protein turnover and RNA turnover are coupled processes.


Asunto(s)
Degradación de ARNm Mediada por Codón sin Sentido , ARN Helicasas/metabolismo , Transactivadores/metabolismo , Represión Epigenética , Humanos , Desarrollo de Músculos , Proteína MioD/genética , Proteína MioD/metabolismo , Proteolisis , Proteoma , ARN Helicasas/genética , Transactivadores/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
14.
Mol Cell ; 43(6): 950-61, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21925383

RESUMEN

Nonsense-mediated mRNA decay (NMD) is a conserved RNA decay pathway that degrades aberrant mRNAs and directly regulates many normal mRNAs. This dual role for NMD raises the possibility that its magnitude is buffered to prevent the potentially catastrophic alterations in gene expression that would otherwise occur if NMD were perturbed by environmental or genetic insults. In support of this, here we report the existence of a negative feedback regulatory network that directly acts on seven NMD factors. Feedback regulation is conferred by different branches of the NMD pathway in a cell type-specific and developmentally regulated manner. We identify feedback-regulated NMD factors that are rate limiting for NMD and demonstrate that reversal of feedback regulation in response to NMD perturbation is crucial for maintaining NMD. Together, our results suggest the existence of an intricate feedback network that maintains both RNA surveillance and the homeostasis of normal gene expression in mammalian cells.


Asunto(s)
Estabilidad del ARN , ARN Mensajero/metabolismo , Factor de Transcripción Activador 3/metabolismo , Western Blotting , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Células HeLa , Homeostasis , Humanos , ARN Helicasas , Interferencia de ARN , Transactivadores/antagonistas & inhibidores
15.
Mol Cell ; 42(4): 500-10, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21596314

RESUMEN

Nonsense-mediated decay (NMD) degrades both normal and aberrant transcripts harboring stop codons in particular contexts. Mutations that perturb NMD cause neurological disorders in humans, suggesting that NMD has roles in the brain. Here, we identify a brain-specific microRNA-miR-128-that represses NMD and thereby controls batteries of transcripts in neural cells. miR-128 represses NMD by targeting the RNA helicase UPF1 and the exon-junction complex core component MLN51. The ability of miR-128 to regulate NMD is a conserved response occurring in frogs, chickens, and mammals. miR-128 levels are dramatically increased in differentiating neuronal cells and during brain development, leading to repressed NMD and upregulation of mRNAs normally targeted for decay by NMD; overrepresented are those encoding proteins controlling neuron development and function. Together, these results suggest the existence of a conserved RNA circuit linking the microRNA and NMD pathways that induces cell type-specific transcripts during development.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Estabilidad del ARN , Transactivadores/metabolismo , Activación Transcripcional , Animales , Encéfalo/metabolismo , Embrión de Pollo , Exones , Células HEK293 , Células HeLa , Humanos , Ratones , MicroARNs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Helicasas , Proteínas de Unión al ARN , Ratas , Transactivadores/genética , Xenopus laevis
16.
Hum Mol Genet ; 25(22): 4898-4910, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28171660

RESUMEN

The X-linked reproductive homeobox (RHOX) gene cluster encodes transcription factors preferentially expressed in reproductive tissues. This gene cluster has important roles in male fertility based on phenotypic defects of Rhox-mutant mice and the finding that aberrant RHOX promoter methylation is strongly associated with abnormal human sperm parameters. However, little is known about the molecular mechanism of RHOX function in humans. Using gene expression profiling, we identified genes regulated by members of the human RHOX gene cluster. Some genes were uniquely regulated by RHOXF1 or RHOXF2/2B, while others were regulated by both of these transcription factors. Several of these regulated genes encode proteins involved in processes relevant to spermatogenesis; e.g. stress protection and cell survival. One of the target genes of RHOXF2/2B is RHOXF1, suggesting cross-regulation to enhance transcriptional responses. The potential role of RHOX in human infertility was addressed by sequencing all RHOX exons in a group of 250 patients with severe oligozoospermia. This revealed two mutations in RHOXF1 (c.515G > A and c.522C > T) and four in RHOXF2/2B (-73C > G, c.202G > A, c.411C > T and c.679G > A), of which only one (c.202G > A) was found in a control group of men with normal sperm concentration. Functional analysis demonstrated that c.202G > A and c.679G > A significantly impaired the ability of RHOXF2/2B to regulate downstream genes. Molecular modelling suggested that these mutations alter RHOXF2/F2B protein conformation. By combining clinical data with in vitro functional analysis, we demonstrate how the X-linked RHOX gene cluster may function in normal human spermatogenesis and we provide evidence that it is impaired in human male fertility.


Asunto(s)
Proteínas de Homeodominio/genética , Infertilidad Masculina/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Genes Homeobox , Genes Ligados a X , Células HEK293 , Proteínas de Homeodominio/metabolismo , Humanos , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Familia de Multigenes , Regiones Promotoras Genéticas , Espermatogénesis/genética , Espermatozoides/patología , Factores de Transcripción/genética
18.
EMBO Rep ; 16(5): 599-609, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25807986

RESUMEN

Endoplasmic reticulum (ER) stress induces the unfolded protein response (UPR), an essential adaptive intracellular pathway that relieves the stress. Although the UPR is an evolutionarily conserved and beneficial pathway, its chronic activation contributes to the pathogenesis of a wide variety of human disorders. The fidelity of UPR activation must thus be tightly regulated to prevent inappropriate signaling. The nonsense-mediated RNA decay (NMD) pathway has long been known to function in RNA quality control, rapidly degrading aberrant mRNAs, and has been suggested to regulate subsets of normal mRNAs. Here, we report that the NMD pathway regulates the UPR. NMD increases the threshold for triggering the UPR in vitro and in vivo, thereby preventing UPR activation in response to normally innocuous levels of ER stress. NMD also promotes the timely termination of the UPR. We demonstrate that NMD directly targets the mRNAs encoding several UPR components, including the highly conserved UPR sensor, IRE1α, whose NMD-dependent degradation partly underpins this process. Our work not only sheds light on UPR regulation, but demonstrates the physiological relevance of NMD's ability to regulate normal mRNAs.


Asunto(s)
Degradación de ARNm Mediada por Codón sin Sentido , Respuesta de Proteína Desplegada/genética , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Estrés del Retículo Endoplásmico/genética , Endorribonucleasas/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Orden Génico , Vectores Genéticos/genética , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Transcripción Genética
20.
Nucleic Acids Res ; 43(19): 9314-26, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25908788

RESUMEN

Olfactory receptor (Olfr) genes comprise the largest gene family in mice. Despite their importance in olfaction, how most Olfr mRNAs are regulated remains unexplored. Using RNA-seq analysis coupled with analysis of pre-existing databases, we found that Olfr mRNAs have several atypical features suggesting that post-transcriptional regulation impacts their expression. First, Olfr mRNAs, as a group, have dramatically higher average AU-content and lower predicted secondary structure than do control mRNAs. Second, Olfr mRNAs have a higher density of AU-rich elements (AREs) in their 3'UTR and upstream open reading frames (uORFs) in their 5 UTR than do control mRNAs. Third, Olfr mRNAs have shorter 3' UTR regions and with fewer predicted miRNA-binding sites. All of these novel properties correlated with higher Olfr expression. We also identified striking differences in the post-transcriptional features of the mRNAs from the two major classes of Olfr genes, a finding consistent with their independent evolutionary origin. Together, our results suggest that the Olfr gene family has encountered unusual selective forces in neural cells that have driven them to acquire unique post-transcriptional regulatory features. In support of this possibility, we found that while Olfr mRNAs are degraded by a deadenylation-dependent mechanism, they are largely protected from this decay in neural lineage cells.


Asunto(s)
Mucosa Olfatoria/metabolismo , ARN Mensajero/química , Receptores Odorantes/genética , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Animales , Sitios de Unión , Línea Celular , Femenino , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Familia de Multigenes , Sistemas de Lectura Abierta , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , ARN Mensajero/metabolismo , Receptores Odorantes/metabolismo , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA