Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 828
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(4): 877-889.e17, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28965759

RESUMEN

N6-methyladenosine (m6A), installed by the Mettl3/Mettl14 methyltransferase complex, is the most prevalent internal mRNA modification. Whether m6A regulates mammalian brain development is unknown. Here, we show that m6A depletion by Mettl14 knockout in embryonic mouse brains prolongs the cell cycle of radial glia cells and extends cortical neurogenesis into postnatal stages. m6A depletion by Mettl3 knockdown also leads to a prolonged cell cycle and maintenance of radial glia cells. m6A sequencing of embryonic mouse cortex reveals enrichment of mRNAs related to transcription factors, neurogenesis, the cell cycle, and neuronal differentiation, and m6A tagging promotes their decay. Further analysis uncovers previously unappreciated transcriptional prepatterning in cortical neural stem cells. m6A signaling also regulates human cortical neurogenesis in forebrain organoids. Comparison of m6A-mRNA landscapes between mouse and human cortical neurogenesis reveals enrichment of human-specific m6A tagging of transcripts related to brain-disorder risk genes. Our study identifies an epitranscriptomic mechanism in heightened transcriptional coordination during mammalian cortical neurogenesis.


Asunto(s)
Neurogénesis , Prosencéfalo/embriología , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Animales , Ciclo Celular , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Organoides/metabolismo , Prosencéfalo/citología , Prosencéfalo/metabolismo , Estabilidad del ARN
2.
Cell ; 169(5): 824-835.e14, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28525753

RESUMEN

Maintenance of proper levels of the methyl donor S-adenosylmethionine (SAM) is critical for a wide variety of biological processes. We demonstrate that the N6-adenosine methyltransferase METTL16 regulates expression of human MAT2A, which encodes the SAM synthetase expressed in most cells. Upon SAM depletion by methionine starvation, cells induce MAT2A expression by enhanced splicing of a retained intron. Induction requires METTL16 and its methylation substrate, a vertebrate conserved hairpin (hp1) in the MAT2A 3' UTR. Increasing METTL16 occupancy on the MAT2A 3' UTR is sufficient to induce efficient splicing. We propose that, under SAM-limiting conditions, METTL16 occupancy on hp1 increases due to inefficient enzymatic turnover, which promotes MAT2A splicing. We further show that METTL16 is the long-unknown methyltransferase for the U6 spliceosomal small nuclear RNA (snRNA). These observations suggest that the conserved U6 snRNA methyltransferase evolved an additional function in vertebrates to regulate SAM homeostasis.


Asunto(s)
Intrones , Metionina Adenosiltransferasa/genética , Metiltransferasas/metabolismo , Empalme del ARN , S-Adenosilmetionina/metabolismo , Animales , Secuencia de Bases , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , Secuencias Invertidas Repetidas , Metionina Adenosiltransferasa/química , Metilación , Metiltransferasas/química , Schizosaccharomyces/metabolismo
3.
Mol Cell ; 83(18): 3303-3313.e6, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37683648

RESUMEN

Cells respond to intrinsic and extrinsic stresses by reducing global protein synthesis and activating gene programs necessary for survival. Here, we show that the integrated stress response (ISR) is driven by the non-canonical cap-binding protein eIF3d that acts as a critical effector to control core stress response orchestrators, the translation factor eIF2α and the transcription factor ATF4. We find that during persistent stress, eIF3d activates the translation of the kinase GCN2, inducing eIF2α phosphorylation and inhibiting general protein synthesis. In parallel, eIF3d upregulates the m6A demethylase ALKBH5 to drive 5' UTR-specific demethylation of stress response genes, including ATF4. Ultimately, this cascade converges on ATF4 expression by increasing mRNA engagement of translation machinery and enhancing ribosome bypass of upstream open reading frames (uORFs). Our results reveal that eIF3d acts in a life-or-death decision point during chronic stress and uncover a synergistic signaling mechanism in which translational cascades complement transcriptional amplification to control essential cellular processes.


Asunto(s)
Estrés del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación , Regiones no Traducidas 5' , Factor 2 Eucariótico de Iniciación/genética , Sistemas de Lectura Abierta , Fosforilación , Proteínas de Unión a Caperuzas de ARN , Humanos
4.
Mol Cell ; 83(23): 4239-4254.e10, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38065062

RESUMEN

A common mRNA modification is 5-methylcytosine (m5C), whose role in gene-transcript processing and cancer remains unclear. Here, we identify serine/arginine-rich splicing factor 2 (SRSF2) as a reader of m5C and impaired SRSF2 m5C binding as a potential contributor to leukemogenesis. Structurally, we identify residues involved in m5C recognition and the impact of the prevalent leukemia-associated mutation SRSF2P95H. We show that SRSF2 binding and m5C colocalize within transcripts. Furthermore, knocking down the m5C writer NSUN2 decreases mRNA m5C, reduces SRSF2 binding, and alters RNA splicing. We also show that the SRSF2P95H mutation impairs the ability of the protein to read m5C-marked mRNA, notably reducing its binding to key leukemia-related transcripts in leukemic cells. In leukemia patients, low NSUN2 expression leads to mRNA m5C hypomethylation and, combined with SRSF2P95H, predicts poor outcomes. Altogether, we highlight an unrecognized mechanistic link between epitranscriptomics and a key oncogenesis driver.


Asunto(s)
Leucemia , Síndromes Mielodisplásicos , Neoplasias , Metilación de ARN , Factores de Empalme Serina-Arginina , Humanos , Leucemia/genética , Síndromes Mielodisplásicos/genética , Neoplasias/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Factores de Empalme Serina-Arginina/genética , Metilación de ARN/genética
5.
Mol Cell ; 81(20): 4228-4242.e8, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34686315

RESUMEN

Central to genotoxic responses is their ability to sense highly specific signals to activate the appropriate repair response. We previously reported that the activation of the ASCC-ALKBH3 repair pathway is exquisitely specific to alkylation damage in human cells. Yet the mechanistic basis for the selectivity of this pathway was not immediately obvious. Here, we demonstrate that RNA but not DNA alkylation is the initiating signal for this process. Aberrantly methylated RNA is sufficient to recruit ASCC, while an RNA dealkylase suppresses ASCC recruitment during chemical alkylation. In turn, recruitment of ASCC during alkylation damage, which is mediated by the E3 ubiquitin ligase RNF113A, suppresses transcription and R-loop formation. We further show that alkylated pre-mRNA is sufficient to activate RNF113A E3 ligase in vitro in a manner dependent on its RNA binding Zn-finger domain. Together, our work identifies an unexpected role for RNA damage in eliciting a specific response to genotoxins.


Asunto(s)
Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 3 de AlkB/metabolismo , Núcleo Celular/enzimología , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias/enzimología , Proteínas Nucleares/metabolismo , Procesamiento Postranscripcional del ARN , ARN Neoplásico/metabolismo , Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 3 de AlkB/genética , Núcleo Celular/genética , ADN Helicasas/genética , Metilación de ADN , Proteínas de Unión al ADN/genética , Células HEK293 , Células HeLa , Humanos , Metilación , Neoplasias/genética , Proteínas Nucleares/genética , Estructuras R-Loop , ARN Neoplásico/genética , Empalmosomas/genética , Empalmosomas/metabolismo , Transcripción Genética , Ubiquitinación
6.
Mol Cell ; 81(20): 4209-4227.e12, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34453888

RESUMEN

The microtubule-associated protein tau oligomerizes, but the actions of oligomeric tau (oTau) are unknown. We have used Cry2-based optogenetics to induce tau oligomers (oTau-c). Optical induction of oTau-c elicits tau phosphorylation, aggregation, and a translational stress response that includes stress granules and reduced protein synthesis. Proteomic analysis identifies HNRNPA2B1 as a principle target of oTau-c. The association of HNRNPA2B1 with endogenous oTau was verified in neurons, animal models, and human Alzheimer brain tissues. Mechanistic studies demonstrate that HNRNPA2B1 functions as a linker, connecting oTau with N6-methyladenosine (m6A) modified RNA transcripts. Knockdown of HNRNPA2B1 prevents oTau or oTau-c from associating with m6A or from reducing protein synthesis and reduces oTau-induced neurodegeneration. Levels of m6A and the m6A-oTau-HNRNPA2B1 complex are increased up to 5-fold in the brains of Alzheimer subjects and P301S tau mice. These results reveal a complex containing oTau, HNRNPA2B1, and m6A that contributes to the integrated stress response of oTau.


Asunto(s)
Adenosina/análogos & derivados , Enfermedad de Alzheimer/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Procesamiento Postranscripcional del ARN , ARN/metabolismo , Proteínas tau/metabolismo , Adenosina/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Masculino , Metilación , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Agregado de Proteínas , Agregación Patológica de Proteínas , ARN/genética , Índice de Severidad de la Enfermedad , Proteínas tau/genética
7.
Trends Biochem Sci ; 49(3): 199-207, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38071089

RESUMEN

Gene expression is a complex process requiring many control mechanisms to achieve a desired phenotype. DNA accessibility within chromatin is well established as an important determinant of gene expression. By contrast, while mRNA also associates with a complement of proteins, the exact nature of messenger ribonucleoprotein (mRNP) packaging and its functional relevance is not as clear. Recent reports indicate that exon junction complex (EJC)-mediated mRNP packaging renders exon junction-proximal regions inaccessible for m6A methylation, and that EJCs reside within the inaccessible interior of globular transcription and export (TREX) complex-associated nuclear mRNPs. We propose that 'mRNA accessibility' within mRNPs is an important determinant of gene expression that may modulate the specificity of a broad array of regulatory processes including but not limited to m6A methylation.


Asunto(s)
Núcleo Celular , Ribonucleoproteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Núcleo Celular/metabolismo , Expresión Génica
8.
Immunity ; 50(3): 600-615.e15, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30824325

RESUMEN

CCR7 chemokine receptor stimulation induces rapid but transient dendritic cell (DC) migration toward draining lymph nodes, which is critical for the initiation of protective immunity and maintenance of immune homeostasis. The mechanisms for terminating CCR7-mediated DC migration remain incompletely understood. Here we have identified a long non-coding RNA lnc-Dpf3 whose feedback restrained CCR7-mediated DC migration. CCR7 stimulation upregulated lnc-Dpf3 via removing N6-methyladenosine (m6A) modification to prevent RNA degradation. DC-specific lnc-Dpf3 deficiency increased CCR7-mediated DC migration, leading to exaggerated adaptive immune responses and inflammatory injuries. Mechanistically, CCR7 stimulation activated the HIF-1α transcription factor pathway in DCs, leading to metabolic reprogramming toward glycolysis for DC migration. lnc-Dpf3 directly bound to HIF-1α and suppressed HIF-1α-dependent transcription of the glycolytic gene Ldha, thus inhibiting DC glycolytic metabolism and migratory capacity. We demonstrate a critical role for CCR7-inducible lnc-Dpf3 in coupling epigenetic and metabolic pathways to feedback-control DC migration and inflammatory responses.


Asunto(s)
Movimiento Celular/genética , Proteínas de Unión al ADN/genética , Glucólisis/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Receptores CCR7/genética , Factores de Transcripción/genética , Inmunidad Adaptativa/genética , Animales , Línea Celular , Células Dendríticas/patología , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/patología , Ganglios Linfáticos/patología , Redes y Vías Metabólicas/genética , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/genética , Regulación hacia Arriba/genética
9.
Genes Dev ; 34(19-20): 1373-1391, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32943573

RESUMEN

The N6-methyladenosine (m6A) modification is the most prevalent post-transcriptional mRNA modification, regulating mRNA decay and splicing. It plays a major role during normal development, differentiation, and disease progression. The modification is regulated by a set of writer, eraser, and reader proteins. The YTH domain family of proteins consists of three homologous m6A-binding proteins, Ythdf1, Ythdf2, and Ythdf3, which were suggested to have different cellular functions. However, their sequence similarity and their tendency to bind the same targets suggest that they may have overlapping roles. We systematically knocked out (KO) the Mettl3 writer, each of the Ythdf readers, and the three readers together (triple-KO). We then estimated the effect in vivo in mouse gametogenesis, postnatal viability, and in vitro in mouse embryonic stem cells (mESCs). In gametogenesis, Mettl3-KO severity is increased as the deletion occurs earlier in the process, and Ythdf2 has a dominant role that cannot be compensated by Ythdf1 or Ythdf3, due to differences in readers' expression pattern across different cell types, both in quantity and in spatial location. Knocking out the three readers together and systematically testing viable offspring genotypes revealed a redundancy in the readers' role during early development that is Ythdf1/2/3 gene dosage-dependent. Finally, in mESCs there is compensation between the three Ythdf reader proteins, since the resistance to differentiate and the significant effect on mRNA decay occur only in the triple-KO cells and not in the single KOs. Thus, we suggest a new model for the Ythdf readers function, in which there is profound dosage-dependent redundancy when all three readers are equivalently coexpressed in the same cell types.


Asunto(s)
Compensación de Dosificación (Genética) , Gametogénesis/genética , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular , Células Madre Embrionarias , Fertilidad/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados
10.
Genes Dev ; 34(11-12): 733-744, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32482714

RESUMEN

The X inactive-specific transcript (Xist) gene is the master regulator of X chromosome inactivation in mammals. Xist produces a long noncoding (lnc)RNA that accumulates over the entire length of the chromosome from which it is transcribed, recruiting factors to modify underlying chromatin and silence X-linked genes in cis Recent years have seen significant progress in identifying important functional elements in Xist RNA, their associated RNA-binding proteins (RBPs), and the downstream pathways for chromatin modification and gene silencing. In this review, we summarize progress in understanding both how these pathways function in Xist-mediated silencing and the complex interplay between them.


Asunto(s)
Proteínas/metabolismo , ARN Largo no Codificante/metabolismo , Inactivación del Cromosoma X/genética , Proteínas de Unión al ADN/metabolismo , Silenciador del Gen/fisiología , Metiltransferasas/metabolismo , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptor de Lamina B
11.
EMBO J ; 42(18): e113378, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37431920

RESUMEN

In virus-host interactions, nucleic acid-directed first lines of defense that allow viral clearance without compromising growth are of paramount importance. Plants use the RNA interference pathway as a basal antiviral immune system, but additional RNA-based mechanisms of defense also exist. The infectivity of a plant positive-strand RNA virus, alfalfa mosaic virus (AMV), relies on the demethylation of viral RNA by the recruitment of the cellular N6-methyladenosine (m6 A) demethylase ALKBH9B, but how demethylation of viral RNA promotes AMV infection remains unknown. Here, we show that inactivation of the Arabidopsis cytoplasmic YT521-B homology domain (YTH)-containing m6 A-binding proteins ECT2, ECT3, and ECT5 is sufficient to restore AMV infectivity in partially resistant alkbh9b mutants. We further show that the antiviral function of ECT2 is distinct from its previously demonstrated function in the promotion of primordial cell proliferation: an ect2 mutant carrying a small deletion in its intrinsically disordered region is partially compromised for antiviral defense but not for developmental functions. These results indicate that the m6 A-YTHDF axis constitutes a novel branch of basal antiviral immunity in plants.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Virus ARN , Antivirales , Proteínas de Plantas/metabolismo , Proteínas de Unión al ARN/metabolismo , Arabidopsis/metabolismo , ARN Viral/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo
12.
Mol Cell ; 75(6): 1188-1202.e11, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31399345

RESUMEN

The maternal-to-zygotic transition (MZT) is a conserved and fundamental process during which the maternal environment is converted to an environment of embryonic-driven development through dramatic reprogramming. However, how maternally supplied transcripts are dynamically regulated during MZT remains largely unknown. Herein, through genome-wide profiling of RNA 5-methylcytosine (m5C) modification in zebrafish early embryos, we found that m5C-modified maternal mRNAs display higher stability than non-m5C-modified mRNAs during MZT. We discovered that Y-box binding protein 1 (Ybx1) preferentially recognizes m5C-modified mRNAs through π-π interactions with a key residue, Trp45, in Ybx1's cold shock domain (CSD), which plays essential roles in maternal mRNA stability and early embryogenesis of zebrafish. Together with the mRNA stabilizer Pabpc1a, Ybx1 promotes the stability of its target mRNAs in an m5C-dependent manner. Our study demonstrates an unexpected mechanism of RNA m5C-regulated maternal mRNA stabilization during zebrafish MZT, highlighting the critical role of m5C mRNA modification in early development.


Asunto(s)
5-Metilcitosina/metabolismo , Embrión no Mamífero/embriología , Desarrollo Embrionario/fisiología , Estabilidad del ARN/fisiología , ARN Mensajero Almacenado/metabolismo , Pez Cebra/embriología , Animales , Células HeLa , Humanos , Ratones , ARN Mensajero Almacenado/genética , Pez Cebra/genética
13.
Mol Cell ; 74(6): 1278-1290.e9, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31031083

RESUMEN

7-methylguanosine (m7G) is present at mRNA caps and at defined internal positions within tRNAs and rRNAs. However, its detection within low-abundance mRNAs and microRNAs (miRNAs) has been hampered by a lack of sensitive detection strategies. Here, we adapt a chemical reactivity assay to detect internal m7G in miRNAs. Using this technique (Borohydride Reduction sequencing [BoRed-seq]) alongside RNA immunoprecipitation, we identify m7G within a subset of miRNAs that inhibit cell migration. We show that the METTL1 methyltransferase mediates m7G methylation within miRNAs and that this enzyme regulates cell migration via its catalytic activity. Using refined mass spectrometry methods, we map m7G to a single guanosine within the let-7e-5p miRNA. We show that METTL1-mediated methylation augments let-7 miRNA processing by disrupting an inhibitory secondary structure within the primary miRNA transcript (pri-miRNA). These results identify METTL1-dependent N7-methylation of guanosine as a new RNA modification pathway that regulates miRNA structure, biogenesis, and cell migration.


Asunto(s)
Guanosina/análogos & derivados , Metiltransferasas/genética , MicroARNs/genética , Procesamiento Postranscripcional del ARN , Células A549 , Secuencia de Bases , Bioensayo , Células CACO-2 , Movimiento Celular , Proliferación Celular , Guanosina/metabolismo , Células HEK293 , Humanos , Metilación , Metiltransferasas/metabolismo , MicroARNs/metabolismo , Conformación de Ácido Nucleico
14.
Proc Natl Acad Sci U S A ; 121(28): e2405100121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38950372

RESUMEN

N6-methyladenosine (m6A) is a fundamentally important RNA modification for gene regulation, whose function is achieved through m6A readers. However, whether and how m6A readers play regulatory roles during fruit ripening and quality formation remains unclear. Here, we characterized SlYTH2 as a tomato m6A reader protein and profiled the binding sites of SlYTH2 at the transcriptome-wide level. SlYTH2 undergoes liquid-liquid phase separation and promotes RNA-protein condensate formation. The target mRNAs of SlYTH2, namely m6A-modified SlHPL and SlCCD1B associated with volatile synthesis, are enriched in SlYTH2-induced condensates. Through polysome profiling assays and proteomic analysis, we demonstrate that knockout of SlYTH2 expedites the translation process of SlHPL and SlCCD1B, resulting in augmented production of aroma-associated volatiles. This aroma enrichment significantly increased consumer preferences for CRISPR-edited fruit over wild type. These findings shed light on the underlying mechanisms of m6A in plant RNA metabolism and provided a promising strategy to generate fruits that are more attractive to consumers.


Asunto(s)
Adenosina , Frutas , Regulación de la Expresión Génica de las Plantas , Proteínas de Plantas , Biosíntesis de Proteínas , Solanum lycopersicum , Solanum lycopersicum/genética , Solanum lycopersicum/metabolismo , Solanum lycopersicum/crecimiento & desarrollo , Frutas/metabolismo , Frutas/genética , Adenosina/metabolismo , Adenosina/análogos & derivados , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Odorantes/análisis
15.
Proc Natl Acad Sci U S A ; 121(14): e2321611121, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38547058

RESUMEN

Malignant glioma exhibits immune evasion characterized by highly expressing the immune checkpoint CD47. RNA 5-methylcytosine(m5C) modification plays a pivotal role in tumor pathogenesis. However, the mechanism underlying m5C-modified RNA metabolism remains unclear, as does the contribution of m5C-modified RNA to the glioma immune microenvironment. In this study, we demonstrate that the canonical 28SrRNA methyltransferase NSUN5 down-regulates ß-catenin by promoting the degradation of its mRNA, leading to enhanced phagocytosis of tumor-associated macrophages (TAMs). Specifically, the NSUN5-induced suppression of ß-catenin relies on its methyltransferase activity mediated by cysteine 359 (C359) and is not influenced by its localization in the nucleolus. Intriguingly, NSUN5 directly interacts with and deposits m5C on CTNNB1 caRNA (chromatin-associated RNA). NSUN5-induced recruitment of TET2 to chromatin is independent of its methyltransferase activity. The m5C modification on caRNA is subsequently oxidized into 5-hydroxymethylcytosine (5hmC) by TET2, which is dependent on its binding affinity for Fe2+ and α-KG. Furthermore, NSUN5 enhances the chromatin recruitment of RBFOX2 which acts as a 5hmC-specific reader to recognize and facilitate the degradation of 5hmC caRNA. Notably, hmeRIP-seq analysis reveals numerous mRNA substrates of NSUN5 that potentially undergo this mode of metabolism. In addition, NSUN5 is epigenetically suppressed by DNA methylation and is negatively correlated with IDH1-R132H mutation in glioma patients. Importantly, pharmacological blockage of DNA methylation or IDH1-R132H mutant and CD47/SIRPα signaling synergistically enhances TAM-based phagocytosis and glioma elimination in vivo. Our findings unveil a general mechanism by which NSUN5/TET2/RBFOX2 signaling regulates RNA metabolism and highlight NSUN5 targeting as a potential strategy for glioma immune therapy.


Asunto(s)
5-Metilcitosina , 5-Metilcitosina/análogos & derivados , Proteínas de Unión al ADN , Dioxigenasas , Glioma , Proteínas Musculares , Humanos , 5-Metilcitosina/metabolismo , beta Catenina/metabolismo , Cromatina , Antígeno CD47/genética , ARN , Evasión Inmune , Glioma/patología , ARN Mensajero/metabolismo , Metiltransferasas/metabolismo , ARN Nuclear Pequeño , Microambiente Tumoral , Factores de Empalme de ARN/genética , Proteínas Represoras/metabolismo
16.
Hum Mol Genet ; 33(11): 969-980, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38483349

RESUMEN

RNA methylation of N6-methyladenosine (m6A) is emerging as a fundamental regulator of every aspect of RNA biology. RNA methylation directly impacts protein production to achieve quick modulation of dynamic biological processes. However, whether RNA methylation regulates mitochondrial function is not known, especially in neuronal cells which require a high energy supply and quick reactive responses. Here we show that m6A RNA methylation regulates mitochondrial function through promoting nuclear-encoded mitochondrial complex subunit RNA translation. Conditional genetic knockout of m6A RNA methyltransferase Mettl14 (Methyltransferase like 14) by Nestin-Cre together with metabolomic analysis reveals that Mettl14 knockout-induced m6A depletion significantly downregulates metabolites related to energy metabolism. Furthermore, transcriptome-wide RNA methylation profiling of wild type and Mettl14 knockout mouse brains by m6A-Seq shows enrichment of methylation on mitochondria-related RNA. Importantly, loss of m6A leads to a significant reduction in mitochondrial respiratory capacity and membrane potential. These functional defects are paralleled by the reduced expression of mitochondrial electron transport chain complexes, as well as decreased mitochondrial super-complex assembly and activity. Mechanistically, m6A depletion decreases the translational efficiency of methylated RNA encoding mitochondrial complex subunits through reducing their association with polysomes, while not affecting RNA stability. Together, these findings reveal a novel role for RNA methylation in regulating mitochondrial function. Given that mitochondrial dysfunction and RNA methylation have been increasingly implicate in neurodegenerative disorders, our findings not only provide insights into fundamental mechanisms regulating mitochondrial function, but also open up new avenues for understanding the pathogenesis of neurological diseases.


Asunto(s)
Adenosina , Metiltransferasas , Ratones Noqueados , Mitocondrias , Animales , Mitocondrias/metabolismo , Mitocondrias/genética , Ratones , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/genética , ARN/genética , ARN/metabolismo , Humanos , Biosíntesis de Proteínas , Metabolismo Energético/genética , Neuronas/metabolismo , Metilación de ARN
17.
Mol Cell ; 71(2): 244-255.e5, 2018 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-29983320

RESUMEN

tRNAs are subject to numerous modifications, including methylation. Mutations in the human N7-methylguanosine (m7G) methyltransferase complex METTL1/WDR4 cause primordial dwarfism and brain malformation, yet the molecular and cellular function in mammals is not well understood. We developed m7G methylated tRNA immunoprecipitation sequencing (MeRIP-seq) and tRNA reduction and cleavage sequencing (TRAC-seq) to reveal the m7G tRNA methylome in mouse embryonic stem cells (mESCs). A subset of 22 tRNAs is modified at a "RAGGU" motif within the variable loop. We observe increased ribosome occupancy at the corresponding codons in Mettl1 knockout mESCs, implying widespread effects on tRNA function, ribosome pausing, and mRNA translation. Translation of cell cycle genes and those associated with brain abnormalities is particularly affected. Mettl1 or Wdr4 knockout mESCs display defective self-renewal and neural differentiation. Our study uncovers the complexity of the mammalian m7G tRNA methylome and highlights its essential role in ESCs with links to human disease.


Asunto(s)
Proteínas de Unión al GTP/genética , Guanosina/análogos & derivados , Metiltransferasas/genética , ARN de Transferencia/genética , Animales , Secuencia de Bases , Diferenciación Celular/genética , Línea Celular , Autorrenovación de las Células/genética , Células Madre Embrionarias , Proteínas de Unión al GTP/metabolismo , Guanosina/genética , Guanosina/metabolismo , Humanos , Metilación , Metiltransferasas/metabolismo , Ratones , Células Madre Embrionarias de Ratones , Procesamiento Postranscripcional del ARN , ARN de Transferencia/metabolismo
18.
Proc Natl Acad Sci U S A ; 120(9): e2204933120, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36812208

RESUMEN

N6-methyladenosine (m6A) regulates mRNA metabolism. While it has been implicated in the development of the mammalian brain and in cognition, the role of m6A in synaptic plasticity, especially during cognitive decline, is not fully understood. In this study, we employed methylated RNA immunoprecipitation sequencing to obtain the m6A epitranscriptome of the hippocampal subregions CA1, CA3, and the dentate gyrus and the anterior cingulate cortex (ACC) in young and aged mice. We observed a decrease in m6A levels in aged animals. Comparative analysis of cingulate cortex (CC) brain tissue from cognitively intact human subjects and Alzheimer's disease (AD) patients showed decreased m6A RNA methylation in AD patients. m6A changes common to brains of aged mice and AD patients were found in transcripts linked to synaptic function including calcium/calmodulin-dependent protein kinase 2 (CAMKII) and AMPA-selective glutamate receptor 1 (Glua1). We used proximity ligation assays to show that reduced m6A levels result in decreased synaptic protein synthesis as exemplified by CAMKII and GLUA1. Moreover, reduced m6A levels impaired synaptic function. Our results suggest that m6A RNA methylation controls synaptic protein synthesis and may play a role in cognitive decline associated with aging and AD.


Asunto(s)
Enfermedad de Alzheimer , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Humanos , Ratones , Animales , Anciano , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hipocampo/metabolismo , Enfermedad de Alzheimer/metabolismo , Envejecimiento/metabolismo , ARN/metabolismo , Mamíferos/genética
19.
J Biol Chem ; 300(1): 105548, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38092148

RESUMEN

Therapeutic mRNAs are generated using modified nucleotides, namely N1-methylpseudouridine (m1Ψ) triphosphate, so that the mRNA evades detection by the immune system. RNA modifications, even at a single-nucleotide position, perturb RNA structure, although it is not well understood how structure and function is impacted by globally modified RNAs. Therefore, we examined the metastasis-associated lung adenocarcinoma transcript 1 triple helix, a highly structured stability element that includes single-, double-, and triple-stranded RNA, globally modified with N6-methyladenosine (m6A), pseudouridine (Ψ), or m1Ψ. UV thermal denaturation assays showed that m6A destabilizes both the Hoogsteen and Watson-Crick faces of the RNA by ∼20 °C, Ψ stabilizes the Hoogsteen and Watson-Crick faces of the RNA by ∼12 °C, and m1Ψ has minimal effect on the stability of the Hoogsteen face of the RNA but increases the stability of the Watson-Crick face by ∼9 °C. Native gel-shift assays revealed that binding of the methyltransferase-like protein 16 to the metastasis-associated lung adenocarcinoma transcript 1 triple helix was weakened by at least 8-, 99-, and 23-fold, respectively, when RNA is globally modified with m6A, Ψ, or m1Ψ. These results demonstrate that a more thermostable RNA structure does not lead to tighter RNA-protein interactions, thereby highlighting the regulatory power of RNA modifications by multiple means.


Asunto(s)
ARN Largo no Codificante , ARN , Metiltransferasas/genética , Metiltransferasas/metabolismo , Conformación de Ácido Nucleico , Nucleótidos , Seudouridina , ARN/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
20.
J Biol Chem ; 300(8): 107488, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908752

RESUMEN

Transfer RNAs (tRNAs) are the most highly modified cellular RNAs, both with respect to the proportion of nucleotides that are modified within the tRNA sequence and with respect to the extraordinary diversity in tRNA modification chemistry. However, the functions of many different tRNA modifications are only beginning to emerge. tRNAs have two general clusters of modifications. The first cluster is within the anticodon stem-loop including several modifications essential for protein translation. The second cluster of modifications is within the tRNA elbow, and roles for these modifications are less clear. In general, tRNA elbow modifications are typically not essential for cell growth, but nonetheless several tRNA elbow modifications have been highly conserved throughout all domains of life. In addition to forming modifications, many tRNA modifying enzymes have been demonstrated or hypothesized to also play an important role in folding tRNA acting as tRNA chaperones. In this review, we summarize the known functions of tRNA modifying enzymes throughout the lifecycle of a tRNA molecule, from transcription to degradation. Thereby, we describe how tRNA modification and folding by tRNA modifying enzymes enhance tRNA maturation, tRNA aminoacylation, and tRNA function during protein synthesis, ultimately impacting cellular phenotypes and disease.


Asunto(s)
Procesamiento Postranscripcional del ARN , ARN de Transferencia , ARN de Transferencia/metabolismo , ARN de Transferencia/genética , Humanos , Biosíntesis de Proteínas , Animales , Anticodón/metabolismo , Anticodón/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA