Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 20(7): 406-420, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30992545

RESUMEN

Nonsense-mediated mRNA decay (NMD) is one of the best characterized and most evolutionarily conserved cellular quality control mechanisms. Although NMD was first found to target one-third of mutated, disease-causing mRNAs, it is now known to also target ~10% of unmutated mammalian mRNAs to facilitate appropriate cellular responses - adaptation, differentiation or death - to environmental changes. Mutations in NMD genes in humans are associated with intellectual disability and cancer. In this Review, we discuss how NMD serves multiple purposes in human cells by degrading both mutated mRNAs to protect the integrity of the transcriptome and normal mRNAs to control the quantities of unmutated transcripts.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Discapacidad Intelectual/metabolismo , Mutación , Neoplasias/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Transcriptoma , Animales , Humanos , Discapacidad Intelectual/genética , Neoplasias/genética , ARN Mensajero/genética , ARN Neoplásico/genética
2.
Nat Rev Mol Cell Biol ; 20(6): 384, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31028366

RESUMEN

The HTML version of the article displayed the wrong Figure 3 (while the PDF version was correct); the HTML has now been corrected and we apologize for any confusion it may have created.

3.
Mol Cell ; 82(23): 4564-4581.e11, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36356584

RESUMEN

How fragile X syndrome protein (FMRP) binds mRNAs and regulates mRNA metabolism remains unclear. Our previous work using human neuronal cells focused on mRNAs targeted for nonsense-mediated mRNA decay (NMD), which we showed are generally bound by FMRP and destabilized upon FMRP loss. Here, we identify >400 high-confidence FMRP-bound mRNAs, only ∼35% of which are NMD targets. Integrative transcriptomics together with SILAC-LC-MS/MS reveal that FMRP loss generally results in mRNA destabilization and more protein produced per FMRP target. We use our established RIP-seq technology to show that FMRP footprints are independent of protein-coding potential, target GC-rich and structured sequences, and are densest in 5' UTRs. Regardless of where within an mRNA FMRP binds, we find that FMRP protects mRNAs from deadenylation and directly binds the cytoplasmic poly(A)-binding protein. Our results reveal how FMRP sequesters polyadenylated mRNAs into stabilized and translationally repressed complexes, whose regulation is critical for neurogenesis and synaptic plasticity.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Humanos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem , Síndrome del Cromosoma X Frágil/genética
4.
Genes Dev ; 28(17): 1900-16, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25184677

RESUMEN

Nonsense-mediated mRNA decay (NMD) controls the quality of eukaryotic gene expression and also degrades physiologic mRNAs. How NMD targets are identified is incompletely understood. A central NMD factor is the ATP-dependent RNA helicase upframeshift 1 (UPF1). Neither the distance in space between the termination codon and the poly(A) tail nor the binding of steady-state, largely hypophosphorylated UPF1 is a discriminating marker of cellular NMD targets, unlike for premature termination codon (PTC)-containing reporter mRNAs when compared with their PTC-free counterparts. Here, we map phosphorylated UPF1 (p-UPF1)-binding sites using transcriptome-wide footprinting or DNA oligonucleotide-directed mRNA cleavage to report that p-UPF1 provides the first reliable cellular NMD target marker. p-UPF1 is enriched on NMD target 3' untranslated regions (UTRs) along with suppressor with morphogenic effect on genitalia 5 (SMG5) and SMG7 but not SMG1 or SMG6. Immunoprecipitations of UPF1 variants deficient in various aspects of the NMD process in parallel with Förster resonance energy transfer (FRET) experiments reveal that ATPase/helicase-deficient UPF1 manifests high levels of RNA binding and disregulated hyperphosphorylation, whereas wild-type UPF1 releases from nonspecific RNA interactions in an ATP hydrolysis-dependent mechanism until an NMD target is identified. 3' UTR-associated UPF1 undergoes regulated phosphorylation on NMD targets, providing a binding platform for mRNA degradative activities. p-UPF1 binding to NMD target 3' UTRs is stabilized by SMG5 and SMG7. Our results help to explain why steady-state UPF1 binding is not a marker for cellular NMD substrates and how this binding is transformed to induce mRNA decay.


Asunto(s)
Estabilidad del ARN/genética , Transactivadores/genética , Transactivadores/metabolismo , Adenosina Trifosfatasas/metabolismo , Sitios de Unión , Células HEK293 , Células HeLa , Humanos , Unión Proteica , Proteína Fosfatasa 2/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , ARN Helicasas/metabolismo , Transcriptoma , Regulación hacia Arriba
5.
Methods ; 155: 68-76, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30576707

RESUMEN

Nonsense-mediated mRNA decay (NMD) is a cellular mRNA degradation mechanism that inhibits the expression of aberrant mRNAs harboring premature termination codons (PTCs). Recent progress in transcriptome-wide sequencing techniques has revealed that NMD also degrades approximately 5-30% of non-mutated cellular mRNAs in a way that can be regulated in response to various cellular signals. In mammals, NMD is governed by the central NMD factor UPF1, which is activated by phosphorylation after translation terminates at a nonsense codon that triggers NMD. We have found that immunoprecipitation using an antibody that is specific for phosphorylated UPF1 is a useful tool to define not only cellular NMD targets but also the nature of NMD decay intermediates and, thus, the process of NMD. To this end, we describe here a detailed protocol for what we call "NMD degradome sequencing" using high-throughput technology.


Asunto(s)
ADN Complementario/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Degradación de ARNm Mediada por Codón sin Sentido , ARN Mensajero/genética , Codón sin Sentido , ADN Complementario/metabolismo , Regulación de la Expresión Génica , Biblioteca de Genes , Células HEK293 , Humanos , Inmunoprecipitación/métodos , Ácido Ocadaico/farmacología , Fosforilación/efectos de los fármacos , ARN Helicasas/genética , ARN Helicasas/metabolismo , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
6.
J Cell Sci ; 129(3): 461-7, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26787741

RESUMEN

Nonsense-mediated mRNA decay (NMD) is an mRNA quality-control mechanism that typifies all eukaryotes examined to date. NMD surveys newly synthesized mRNAs and degrades those that harbor a premature termination codon (PTC), thereby preventing the production of truncated proteins that could result in disease in humans. This is evident from dominantly inherited diseases that are due to PTC-containing mRNAs that escape NMD. Although many cellular NMD targets derive from mistakes made during, for example, pre-mRNA splicing and, possibly, transcription initiation, NMD also targets ∼10% of normal physiological mRNAs so as to promote an appropriate cellular response to changing environmental milieus, including those that induce apoptosis, maturation or differentiation. Over the past ∼35 years, a central goal in the NMD field has been to understand how cells discriminate mRNAs that are targeted by NMD from those that are not. In this Cell Science at a Glance and the accompanying poster, we review progress made towards this goal, focusing on human studies and the role of the key NMD factor up-frameshift protein 1 (UPF1).


Asunto(s)
Degradación de ARNm Mediada por Codón sin Sentido/genética , ARN Mensajero/genética , Apoptosis/genética , Diferenciación Celular/genética , Codón sin Sentido/genética , Humanos , ARN Helicasas , Transactivadores/genética
7.
Proc Natl Acad Sci U S A ; 110(9): 3357-62, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23404710

RESUMEN

Nonsense-mediated mRNA decay (NMD), which degrades transcripts harboring a premature termination codon (PTC), depends on the helicase up-frameshift 1 (UPF1). However, mRNAs that are not NMD targets also bind UPF1. What governs the timing, position, and function of UPF1 binding to mRNAs remains unclear. We provide evidence that (i) multiple UPF1 molecules accumulate on the 3'-untranslated region (3' UTR) of PTC-containing mRNAs and to an extent that is greater per unit 3' UTR length if the mRNA is an NMD target; (ii) UPF1 binding begins ≥35 nt downstream of the PTC; (iii) enhanced UPF1 binding to the 3' UTR of PTC-containing mRNA relative to its PTC-free counterpart depends on translation; and (iv) the presence of a 3' UTR exon-junction complex (EJC) further enhances UPF1 binding and/or affinity. Our data suggest that NMD involves UPF1 binding along a 3' UTR whether the 3' UTR contains an EJC. This binding explains how mRNAs without a 3' UTR EJC but with an abnormally long 3' UTR can be NMD targets, albeit not as efficiently as their counterparts that contain a 3' UTR EJC.


Asunto(s)
Regiones no Traducidas 3'/genética , Transactivadores/metabolismo , Codón sin Sentido/genética , Exones/genética , Humanos , Nucleótidos/metabolismo , Factores de Terminación de Péptidos/metabolismo , Unión Proteica/genética , Biosíntesis de Proteínas/genética , ARN Helicasas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Globinas beta/genética , Globinas beta/metabolismo
8.
Neurobiol Dis ; 69: 200-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24878509

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by transcription of CUG repeat RNA, which causes sequestration of muscleblind-like 1 (MBNL1) and upregulation of CUG triplet repeat RNA-binding protein (CUG-BP1). In DM1, dysregulation of these proteins contributes to many aberrant splicing events, causing various symptoms of the disorder. Here, we demonstrate the occurrence of aberrant splicing of LIM domain binding 3 (LDB3) exon 11 in DM1 skeletal muscle. Exon array surveys, RT-PCR, and western blotting studies demonstrated that exon 11 inclusion was DM1 specific and could be reproduced by transfection of a minigene containing the CTG repeat expansion. Moreover, we found that the LDB3 exon 11-positive isoform had reduced affinity for PKC compared to the exon 11-negative isoform. Since PKC exhibits hyperactivation in DM1 and stabilizes CUG-BP1 by phosphorylation, aberrant splicing of LDB3 may contribute to CUG-BP1 upregulation through changes in its affinity for PKC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Músculo Esquelético/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Proteína Quinasa C/metabolismo , Adulto , Anciano , Niño , Estudios de Cohortes , Exones , Femenino , Humanos , Lactante , Isoenzimas , Masculino , Persona de Mediana Edad , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Isoformas de Proteínas , Empalme del ARN , Transfección , Expansión de Repetición de Trinucleótido , Adulto Joven
9.
Genome Biol ; 25(1): 31, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38263082

RESUMEN

In non-polarized cells, nonsense-mediated mRNA decay (NMD) generally begins during the translation of newly synthesized mRNAs after the mRNAs are exported to the cytoplasm. Binding of the FMRP translational repressor to UPF1 on NMD targets mainly inhibits NMD. However, in polarized cells like neurons, FMRP additionally localizes mRNAs to cellular projections. Here, we review the literature and evaluate available transcriptomic data to conclude that, in neurons, the translation of physiologic NMD targets bound by FMRP is partially inhibited until the mRNAs localize to projections. There, FMRP displacement in response to signaling induces a burst in protein synthesis followed by rapid mRNA decay.


Asunto(s)
Neuronas , Degradación de ARNm Mediada por Codón sin Sentido , Citoplasma , Transducción de Señal , Perfilación de la Expresión Génica , ARN Mensajero
10.
bioRxiv ; 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38352429

RESUMEN

UPF1-mediated decay entails several mRNA surveillance pathways that play a crucial role in cellular homeostasis. However, the precise role of UPF1 in postmitotic neurons remains unresolved, as does its activity in amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease characterized by TDP-43 pathology and disrupted mRNA metabolism. Here, we used human iPSC-derived spinal motor neurons (MNs) to identify mRNAs subject to UPF1 degradation by integrating RNA-seq before and after UPF1 knockdown with RIP-seq to identify RNAs that co-immunoprecipitate with the active form of phosphorylated UPF1. We define a stringent set of bona fide UPF1 targets in MNs that are functionally enriched for autophagy and structurally enriched for GC-rich and long 3' UTRs but not for premature termination codon (PTC)-containing transcripts. TDP-43 depletion in iPSC-derived MNs reduces UPF1 phosphorylation and consequently post-transcriptional upregulation of UPF1 targets, suggesting that TDP-43 dysfunction compromises UPF1-mediated mRNA surveillance. Intriguingly, our datasets reveal that UPF1 and TDP-43 regulate alternative polyadenylation and 3'UTR length of mRNAs associated with synaptic and axonal function, a process that we find to be compromised in ALS models in vitro and ALS patient tissue. Our study provides a comprehensive description of UPF1-mediated mRNA decay activity in neurons, reveals overlapping roles between UPF1 and TDP-43 in regulating 3'UTR length, and offers novel insight into the intricate interplay between RNA metabolism and neurodegeneration in ALS.

11.
Biochem Genet ; 50(3-4): 309-17, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21964520

RESUMEN

Human PQBP-1 is known to interact with triplet repeat disease gene products such as ataxin and huntingtin through their poly-glutamine (poly-Q) tracts. The poly-Q tracts show extensive variation in both the number and the configuration of repeats among species. A surface plasmon resonance assay showed clear interaction between human PQBP-1 and Q(11), representative of the poly-Q tract of the ataxin-1 of Old World monkeys. No response was observed using Q(2)PQ(2)P(4)Q(2), representative of the poly-Q tract of the ataxin-1 of New World monkeys. This implies that the interaction of human PQBP-1 with ataxin-1 is limited to humans and closely related species. Comparison of the human and mouse PQBP-1 sequences showed an elevated amino acid substitution rate in the polar amino acid-rich domain of PQBP-1 that is responsible for binding to poly-Q tracts. This could have been advantageous to the new biological function of human PQBP-1 through poly-Q tracts.


Asunto(s)
Proteínas Portadoras/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Ataxina-1 , Ataxinas , Proteínas Portadoras/química , Proteínas de Unión al ADN , Evolución Molecular , Variación Genética , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/química , Primates/genética , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
12.
Front Genet ; 13: 936869, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36199580

RESUMEN

Spinocerebellar ataxia type 10 (SCA10) is characterized by progressive cerebellar neurodegeneration and, in many patients, epilepsy. This disease mainly occurs in individuals with Indigenous American or East Asian ancestry, with strong evidence supporting a founder effect. The mutation causing SCA10 is a large expansion in an ATTCT pentanucleotide repeat in intron 9 of the ATXN10 gene. The ATTCT repeat is highly unstable, expanding to 280-4,500 repeats in affected patients compared with the 9-32 repeats in normal individuals, one of the largest repeat expansions causing neurological disorders identified to date. However, the underlying molecular basis of how this huge repeat expansion evolves and contributes to the SCA10 phenotype remains largely unknown. Recent progress in next-generation DNA sequencing technologies has established that the SCA10 repeat sequence has a highly heterogeneous structure. Here we summarize what is known about the structure and origin of SCA10 repeats, discuss the potential contribution of variant repeats to the SCA10 disease phenotype, and explore how this information can be exploited for therapeutic benefit.

13.
Genome Biol ; 22(1): 317, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34784943

RESUMEN

BACKGROUND: Fragile X syndrome (FXS) is an intellectual disability attributable to loss of fragile X protein (FMRP). We previously demonstrated that FMRP binds mRNAs targeted for nonsense-mediated mRNA decay (NMD) and that FMRP loss results in hyperactivated NMD and inhibition of neuronal differentiation in human stem cells. RESULTS: We show here that NMD is hyperactivated during the development of the cerebral cortex, hippocampus, and cerebellum in the Fmr1-knockout (KO) mouse during embryonic and early postnatal periods. Our findings demonstrate that NMD regulates many neuronal mRNAs that are important for mouse brain development. CONCLUSIONS: We reveal the abnormal regulation of these mRNAs in the Fmr1-KO mouse, a model of FXS, and highlight the importance of early intervention.


Asunto(s)
Encefalopatías/genética , Encéfalo/crecimiento & desarrollo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Degradación de ARNm Mediada por Codón sin Sentido/genética , Animales , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo
14.
Nat Cell Biol ; 23(1): 40-48, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33420492

RESUMEN

Loss of the fragile X protein FMRP is a leading cause of intellectual disability and autism1,2, but the underlying mechanism remains poorly understood. We report that FMRP deficiency results in hyperactivated nonsense-mediated mRNA decay (NMD)3,4 in human SH-SY5Y neuroblastoma cells and fragile X syndrome (FXS) fibroblast-derived induced pluripotent stem cells (iPSCs). We examined the underlying mechanism and found that the key NMD factor UPF1 binds directly to FMRP, promoting FMRP binding to NMD targets. Our data indicate that FMRP acts as an NMD repressor. In the absence of FMRP, NMD targets are relieved from FMRP-mediated translational repression so that their half-lives are decreased and, for those NMD targets encoding NMD factors, increased translation produces abnormally high factor levels despite their hyperactivated NMD. Transcriptome-wide alterations caused by NMD hyperactivation have a role in the FXS phenotype. Consistent with this, small-molecule-mediated inhibition of hyperactivated NMD, which typifies iPSCs derived from patients with FXS, restores a number of neurodifferentiation markers, including those not deriving from NMD targets. Our mechanistic studies reveal that many molecular abnormalities in FMRP-deficient cells are attributable-either directly or indirectly-to misregulated NMD.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Eliminación de Gen , Neuroblastoma/patología , Degradación de ARNm Mediada por Codón sin Sentido , Transcriptoma , Estudios de Casos y Controles , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuronas/metabolismo , Neuronas/patología , RNA-Seq , Transactivadores
15.
Mol Biol Evol ; 26(11): 2573-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19651850

RESUMEN

Spinocerebellar ataxia type 10 is caused by ATTCT repeat expansion in the ATXN10 gene in humans. We studied the evolutionary history of the human genome to determine the time and mechanism of the acquisition of unstable ATTCT repeats in the genome. We found that long interspersed element-1 (LINE-1) was inserted into ATXN10 intron 9; Alu was then inserted in the middle of LINE-1; and endogenous retrovilcus K was lastly retrotransposed in the middle of Alu. The ATTCT repeat was located on the boundary between the 3'-end of the Alu element and the direct repeat arising from LINE-1. We determined nucleotide sequences of the orthologous region of 50 individuals representing 33 primate species and compared them with the human sequence. The analysis revealed that the ATTCT repeat is present only in human and apes. Old World monkeys also possess pentanucleotide repeats, but their motifs are TGTCT and GGTCT. New World monkeys and prosimians are not informative because they lack the corresponding region in ATXN10 intron 9. Our studies dictate two parsimonious scenarios of evolution. First, a TTTCT motif arose from a TTTTT motif at the junction of Alu and LINE-1, which was followed by introduction of A to make an ATTCT motif in hominoids. Second, an ATTCT motif was directly generated from an ancestral ATTTT motif in the common ancestor of catarrhines. We also demonstrate that orangutan uniquely introduced G to make a GTTCT motif and later C to make a GTTCC motif, where newly introduced nucleotides are underlined. Our studies reveal that nucleotide substitutions in a poly(A) tail of the Alu element and the following amplification of pentanucleotides occurred in the lineages of Old World monkeys and hominoids and that unstable ATTCT pentanucleotide repeats originated in the common ancestor of hominoids. These findings also highlight a new aspect of the role of retrotransposons in human disease and evolution, which might be useful in investigating the mystery of human uniqueness.


Asunto(s)
Repeticiones de Microsatélite/genética , Proteínas del Tejido Nervioso/genética , Ataxina-10 , Secuencia de Bases , Evolución Molecular , Humanos , Intrones/genética , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Retroelementos/genética , Análisis de Secuencia de ADN , Ataxias Espinocerebelosas/genética
17.
Methods Mol Biol ; 1720: 175-186, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29236259

RESUMEN

Recent progress in the technology of transcriptome-wide high-throughput sequencing has revealed that nonsense-mediated mRNA decay (NMD) targets ~10% of physiologic transcripts for the purpose of tuning gene expression in response to various environmental conditions. Regardless of the eukaryote studied, NMD requires the ATP-dependent RNA helicase upframeshift 1 (UPF1). It was initially thought that cellular NMD targets could be defined by their binding to steady-state UPF1, which is largely hypophosphorylated. However, the propensity for steady-state UPF1 to bind RNA nonspecifically, coupled with regulated phosphorylation of UPF1 on an NMD target serving as the trigger for NMD, made it clear that it is phosphorylated UPF1 (p-UPF1), rather than steady-state UPF1, that can be used to distinguish cellular NMD targets from cellular RNAs that are not. Here, we describe the immunoprecipitation of p-UPF1 followed by RNA sequencing (p-UPF1 RIP-seq) as a transcriptome-wide approach to define physiologic NMD targets.


Asunto(s)
Inmunoprecipitación/métodos , Degradación de ARNm Mediada por Codón sin Sentido/fisiología , ARN Helicasas/metabolismo , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN/métodos , Transactivadores/metabolismo , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Fosforilación/inmunología , ARN Helicasas/inmunología , ARN Mensajero/genética , Transactivadores/inmunología , Transcriptoma/fisiología
18.
Nat Struct Mol Biol ; 25(10): 940-950, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30275517

RESUMEN

Nonsense-mediated messenger RNA decay (NMD) controls mRNA quality and degrades physiologic mRNAs to fine-tune gene expression in changing developmental or environmental milieus. NMD requires that its targets are removed from the translating pool of mRNAs. Since the decay steps of mammalian NMD remain unknown, we developed assays to isolate and sequence direct NMD decay intermediates transcriptome-wide based on their co-immunoprecipitation with phosphorylated UPF1, which is the active form of this essential NMD factor. We show that, unlike steady-state UPF1, phosphorylated UPF1 binds predominantly deadenylated mRNA decay intermediates and activates NMD cooperatively from 5'- and 3'-ends. We leverage method modifications to characterize the 3'-ends of NMD decay intermediates, show that they are ribosome-bound, and reveal that some are subject to the addition of non-templated nucleotide. Uridines are added by TUT4 and TUT7 terminal uridylyl transferases and removed by the Perlman syndrome-associated exonuclease DIS3L2. The addition of other non-templated nucleotides appears to inhibit decay.


Asunto(s)
Regulación de la Expresión Génica , Estabilidad del ARN , ARN Mensajero/metabolismo , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Exorribonucleasas/fisiología , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/fisiología , Células HEK293 , Humanos , Modelos Moleculares , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/fisiología
19.
Nat Struct Mol Biol ; 25(11): 1059, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30353197

RESUMEN

In the version of this paper originally published, in the PDF references 48-55 appeared in the reference list for the Methods section although they should have been in the reference list for the main text. The error has been corrected in the PDF now available.

20.
Gene ; 373: 23-7, 2006 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-16497448

RESUMEN

We investigated the CAG repeat sequence of the spinocerebellar ataxia type 1 (SCA1) gene in various species of primates to reveal how human has acquired the repeat structure with interruptions. Our results demonstrate no repetitive structure in the region corresponding to the human CAG repeats in prosimians and New World monkeys like in rodents, perfect (uninterrupted) CAG repeats in Old World monkeys, and interrupted CAG repeats in hominoids. Comparative analysis on the secondary structures of the primate SCA1 transcripts suggests the human prototype was built in the common ancestor of simians. We show an evolutionary scenario for acquisition of CAG repeats with interruptions in the human SCA1 gene.


Asunto(s)
Evolución Molecular , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Repeticiones de Trinucleótidos , Secuencia de Aminoácidos , Animales , Ataxina-1 , Ataxinas , Cercopithecidae/genética , Glutamina , Hominidae/genética , Humanos , Datos de Secuencia Molecular , Strepsirhini/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA