Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 84(9): 1711-1726.e11, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38569554

ABSTRACT

N6-methyladenosine (m6A) is a crucial RNA modification that regulates diverse biological processes in human cells, but its co-transcriptional deposition and functions remain poorly understood. Here, we identified the RNA helicase DDX21 with a previously unrecognized role in directing m6A modification on nascent RNA for co-transcriptional regulation. DDX21 interacts with METTL3 for co-recruitment to chromatin through its recognition of R-loops, which can be formed co-transcriptionally as nascent transcripts hybridize onto the template DNA strand. Moreover, DDX21's helicase activity is needed for METTL3-mediated m6A deposition onto nascent RNA following recruitment. At transcription termination regions, this nexus of actions promotes XRN2-mediated termination of RNAPII transcription. Disruption of any of these steps, including the loss of DDX21, METTL3, or their enzymatic activities, leads to defective termination that can induce DNA damage. Therefore, we propose that the R-loop-DDX21-METTL3 nexus forges the missing link for co-transcriptional modification of m6A, coordinating transcription termination and genome stability.


Subject(s)
Adenosine , Adenosine/analogs & derivatives , DEAD-box RNA Helicases , Exoribonucleases , Genomic Instability , Methyltransferases , R-Loop Structures , RNA Polymerase II , Transcription Termination, Genetic , Humans , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Adenosine/metabolism , Adenosine/genetics , Exoribonucleases/metabolism , Exoribonucleases/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , HEK293 Cells , Chromatin/metabolism , Chromatin/genetics , DNA Damage , HeLa Cells , RNA/metabolism , RNA/genetics , Transcription, Genetic , RNA Methylation
2.
Mol Cell ; 83(23): 4239-4254.e10, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38065062

ABSTRACT

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.


Subject(s)
Leukemia , Myelodysplastic Syndromes , Neoplasms , RNA Methylation , Serine-Arginine Splicing Factors , Humans , Leukemia/genetics , Myelodysplastic Syndromes/genetics , Neoplasms/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors/genetics , RNA Methylation/genetics
3.
Mol Cell ; 82(4): 713-715, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35180427

ABSTRACT

In this issue of Molecular Cell, Tegowski et al. established a single-cell DART-seq (scDART-seq) method for m6A site profiling and further revealed the fundamental features of m6A biology in single cells. The findings improve our understating of the intrinsic regulatory and functional mechanisms of m6A in a single-cell basis.

4.
Cell ; 149(1): 101-12, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22445173

ABSTRACT

Eukaryotes have evolved complex mechanisms to repair DNA double-strand breaks (DSBs) through coordinated actions of protein sensors, transducers, and effectors. Here we show that ∼21-nucleotide small RNAs are produced from the sequences in the vicinity of DSB sites in Arabidopsis and in human cells. We refer to these as diRNAs for DSB-induced small RNAs. In Arabidopsis, the biogenesis of diRNAs requires the PI3 kinase ATR, RNA polymerase IV (Pol IV), and Dicer-like proteins. Mutations in these proteins as well as in Pol V cause significant reduction in DSB repair efficiency. In Arabidopsis, diRNAs are recruited by Argonaute 2 (AGO2) to mediate DSB repair. Knock down of Dicer or Ago2 in human cells reduces DSB repair. Our findings reveal a conserved function for small RNAs in the DSB repair pathway. We propose that diRNAs may function as guide molecules directing chromatin modifications or the recruitment of protein complexes to DSB sites to facilitate repair.


Subject(s)
Arabidopsis/metabolism , DNA Breaks, Double-Stranded , DNA Repair , RNA, Plant/metabolism , RNA, Untranslated/metabolism , DEAD-box RNA Helicases/metabolism , Humans , Ribonuclease III/metabolism
5.
Mol Cell ; 75(6): 1188-1202.e11, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31399345

ABSTRACT

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.


Subject(s)
5-Methylcytosine/metabolism , Embryo, Nonmammalian/embryology , Embryonic Development/physiology , RNA Stability/physiology , RNA, Messenger, Stored/metabolism , Zebrafish/embryology , Animals , HeLa Cells , Humans , Mice , RNA, Messenger, Stored/genetics , Zebrafish/genetics
6.
Nucleic Acids Res ; 52(D1): D909-D918, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37870433

ABSTRACT

Diverse individuals age at different rates and display variable susceptibilities to tissue aging, functional decline and aging-related diseases. Centenarians, exemplifying extreme longevity, serve as models for healthy aging. The field of human aging and longevity research is rapidly advancing, garnering significant attention and accumulating substantial data in recent years. Omics technologies, encompassing phenomics, genomics, transcriptomics, proteomics, metabolomics and microbiomics, have provided multidimensional insights and revolutionized cohort-based investigations into human aging and longevity. Accumulated data, covering diverse cells, tissues and cohorts across the lifespan necessitates the establishment of an open and integrated database. Addressing this, we established the Human Aging and Longevity Landscape (HALL), a comprehensive multi-omics repository encompassing a diverse spectrum of human cohorts, spanning from young adults to centenarians. The core objective of HALL is to foster healthy aging by offering an extensive repository of information on biomarkers that gauge the trajectory of human aging. Moreover, the database facilitates the development of diagnostic tools for aging-related conditions and empowers targeted interventions to enhance longevity. HALL is publicly available at https://ngdc.cncb.ac.cn/hall/index.


Subject(s)
Aging , Databases, Factual , Longevity , Multiomics , Aged, 80 and over , Humans , Young Adult , Aging/genetics , Biomarkers , Disease Susceptibility , Genomics , Longevity/genetics
7.
Proc Natl Acad Sci U S A ; 120(7): e2212212120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36745802

ABSTRACT

During vertebrate embryogenesis, hematopoietic stem and progenitor cell (HSPC) production through endothelial-to-hematopoietic transition requires suitable developmental signals, but how these signals are accurately regulated remains incompletely understood. Cytoplasmic polyadenylation, which is one of the posttranscriptional regulations, plays a crucial role in RNA metabolism. Here, we report that Cpeb1b-mediated cytoplasmic polyadenylation is important for HSPC specification by translational control of Hedgehog (Hh) signaling during zebrafish early development. Cpeb1b is highly expressed in notochord and its deficiency results in defective HSPC production. Mechanistically, Cpeb1b regulates hemogenic endothelium specification by the Hedgehog-Vegf-Notch axis. We demonstrate that the cytoplasmic polyadenylation element motif-dependent interaction between Cpeb1b and shha messenger RNA (mRNA) in the liquid-like condensates, which are induced by Pabpc1b phase separation, is required for cytoplasmic polyadenylation of shha mRNA. Intriguingly, the cytoplasmic polyadenylation regulates translation but not stability of shha mRNA, which further enhances the Shha protein level and Hh signal transduction. Taken together, our findings uncover the role of Cpeb1b-mediated cytoplasmic polyadenylation in HSPC development and provide insights into how posttranscriptional regulation can direct developmental signals with high fidelity to translate them into cell fate transition.


Subject(s)
Polyadenylation , Zebrafish , Animals , Zebrafish/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Hedgehog Proteins/metabolism , Hematopoiesis/genetics
8.
J Biol Chem ; 299(6): 104738, 2023 06.
Article in English | MEDLINE | ID: mdl-37086786

ABSTRACT

O-linked GlcNAc (O-GlcNAc) is an emerging post-translation modification that couples metabolism with cellular signal transduction by crosstalk with phosphorylation and ubiquitination to orchestrate various biological processes. The mechanisms underlying the involvement of O-GlcNAc modifications in N6-methyladenosine (m6A) regulation are not fully characterized. Herein, we show that O-GlcNAc modifies the m6A mRNA reader YTH domain family 1 (YTHDF1) and fine-tunes its nuclear translocation by the exportin protein Crm1. First, we present evidence that YTHDF1 interacts with the sole O-GlcNAc transferase (OGT). Second, we verified Ser196/Ser197/Ser198 as the YTHDF1 O-GlcNAcylation sites, as described in numerous chemoproteomic studies. Then we constructed the O-GlcNAc-deficient YTHDF1-S196A/S197F/S198A (AFA) mutant, which significantly attenuated O-GlcNAc signals. Moreover, we revealed that YTHDF1 is a nucleocytoplasmic protein, whose nuclear export is mediated by Crm1. Furthermore, O-GlcNAcylation increases the cytosolic portion of YTHDF1 by enhancing binding with Crm1, thus upregulating downstream target (e.g. c-Myc) expression. Molecular dynamics simulations suggest that O-GlcNAcylation at S197 promotes the binding between the nuclear export signal motif and Crm1 through increasing hydrogen bonding. Mouse xenograft assays further demonstrate that YTHDF1-AFA mutants decreased the colon cancer mass and size via decreasing c-Myc expression. In sum, we found that YTHDF1 is a nucleocytoplasmic protein, whose cytosolic localization is dependent on O-GlcNAc modification. We propose that the OGT-YTHDF1-c-Myc axis underlies colorectal cancer tumorigenesis.


Subject(s)
Colorectal Neoplasms , Protein Processing, Post-Translational , Mice , Animals , Humans , Phosphorylation , Ubiquitination , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , N-Acetylglucosaminyltransferases/metabolism , Acetylglucosamine/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
Breast Cancer Res ; 26(1): 94, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844963

ABSTRACT

BACKGROUND: RNA m5C methylation has been extensively implicated in the occurrence and development of tumors. As the main methyltransferase, NSUN2 plays a crucial regulatory role across diverse tumor types. However, the precise impact of NSUN2-mediated m5C modification on breast cancer (BC) remains unclear. Our study aims to elucidate the molecular mechanism underlying how NSUN2 regulates the target gene HGH1 (also known as FAM203) through m5C modification, thereby promoting BC progression. Additionally, this study targets at preliminarily clarifying the biological roles of NSUN2 and HGH1 in BC. METHODS: Tumor and adjacent tissues from 5 BC patients were collected, and the m5C modification target HGH1 in BC was screened through RNA sequencing (RNA-seq) and single-base resolution m5C methylation sequencing (RNA-BisSeq). Methylation RNA immunoprecipitation-qPCR (MeRIP-qPCR) and RNA-binding protein immunoprecipitation-qPCR (RIP-qPCR) confirmed that the methylation molecules NSUN2 and YBX1 specifically recognized and bound to HGH1 through m5C modification. In addition, proteomics, co-immunoprecipitation (co-IP), and Ribosome sequencing (Ribo-Seq) were used to explore the biological role of HGH1 in BC. RESULTS: As the main m5C methylation molecule, NSUN2 is abnormally overexpressed in BC and increases the overall level of RNA m5C. Knocking down NSUN2 can inhibit BC progression in vitro or in vivo. Combined RNA-seq and RNA-BisSeq analysis identified HGH1 as a potential target of abnormal m5C modifications. We clarified the mechanism by which NSUN2 regulates HGH1 expression through m5C modification, a process that involves interactions with the YBX1 protein, which collectively impacts mRNA stability and protein synthesis. Furthermore, this study is the first to reveal the binding interaction between HGH1 and the translation elongation factor EEF2, providing a comprehensive understanding of its ability to regulate transcript translation efficiency and protein synthesis in BC cells. CONCLUSIONS: This study preliminarily clarifies the regulatory role of the NSUN2-YBX1-m5C-HGH1 axis from post-transcriptional modification to protein translation, revealing the key role of abnormal RNA m5C modification in BC and suggesting that HGH1 may be a new epigenetic biomarker and potential therapeutic target for BC.


Subject(s)
Breast Neoplasms , Disease Progression , Gene Expression Regulation, Neoplastic , Methyltransferases , RNA Stability , Y-Box-Binding Protein 1 , Animals , Female , Humans , Mice , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Methylation , Methyltransferases/metabolism , Methyltransferases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Y-Box-Binding Protein 1/metabolism , Y-Box-Binding Protein 1/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism
10.
Acc Chem Res ; 56(23): 3417-3427, 2023 12 05.
Article in English | MEDLINE | ID: mdl-37965760

ABSTRACT

More than 170 different types of chemical modifications have been identified on diverse types of RNA, collectively known as the epitranscriptome. Among them, N6-methyladenine (m6A), 5-methylcytosine (m5C), N1-methyladenine (m1A), and N7-methylguanosine (m7G) as the ubiquitous post-transcriptional modification are widely involved in regulating the metabolic processes such as RNA degradation, translation, stability, and export, mediating important physiological and pathological processes such as stress regulation, immune response, development, and tumorigenesis. Recently, the regulatory role of RNA modification during developmental processes is getting more attention. Therefore, the development of low-input even single-cell and high-resolution sequencing technologies is crucial for the exploration of the regulatory roles of RNA modifications in these important biological events of trace samples.This account focuses on the roles of RNA modifications in various developmental processes. We describe the distribution characteristics of various RNA modifications, catalytic enzymes, binding proteins, and the development of sequencing technologies. RNA modification is dynamically reversible, which can be catalyzed by methyltransferases and eliminated by demethylases. RNA m6A is the most abundant post-transcriptional modification on eukaryote mRNA, which is mainly concentrated near the stop codon, and involves in RNA metabolism regulation. RNA m5C, another most studied RNA modification, has been identified in a various of organisms and RNA species, mainly enriched in the regions downstream of translation initiation sites and broadly distributes across the whole coding sequence (CDS) in mammalian mRNAs. RNA m1A, with a lower abundance than m6A, is widely distributed in various RNA types, mainly locates in the 5' untranslated region (5'UTR) of mRNA and regulates translation. RNA m7G, one of the most common RNA modifications in eukaryotes, has been identified at cap regions and internal positions of RNAs and recently gained considerable attention.Thanks to the development of sequencing technology, m6A has been found to regulate the tumorigenic process, including tumor proliferation, invasion, and metastasis by modulating oncogenes and tumor suppressor genes, and affect oocyte maturation and embryonic development through regulating maternal and zygotic genes. m5C related proteins have been identified to participate in embryonic development, plant growth, and neural stem cell differentiation in a m5C dependent manner. m1A also has been revealed to be involved in these developmental processes. m7G dysregulation mainly involves in neurodevelopmental disorders and neurodegenerative diseases.Collectively, we summarized the gradually exhibited roles of RNA methylation during development, and discussed the possibility of RNA modifications as candidate biomarkers and potential therapeutic targets. The technological development is anticipated as the major driving force to expand our knowledge in this field.


Subject(s)
Methyltransferases , RNA , Animals , Methylation , RNA/genetics , RNA/metabolism , RNA, Messenger/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Cell Differentiation , RNA Processing, Post-Transcriptional , Mammals/genetics , Mammals/metabolism
11.
Mol Cell ; 61(4): 507-519, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26876937

ABSTRACT

The regulatory role of N(6)-methyladenosine (m(6)A) and its nuclear binding protein YTHDC1 in pre-mRNA splicing remains an enigma. Here we show that YTHDC1 promotes exon inclusion in targeted mRNAs through recruiting pre-mRNA splicing factor SRSF3 (SRp20) while blocking SRSF10 (SRp38) mRNA binding. Transcriptome assay with PAR-CLIP-seq analysis revealed that YTHDC1-regulated exon-inclusion patterns were similar to those of SRSF3 but opposite of SRSF10. In vitro pull-down assay illustrated a competitive binding of SRSF3 and SRSF10 to YTHDC1. Moreover, YTHDC1 facilitates SRSF3 but represses SRSF10 in their nuclear speckle localization, RNA-binding affinity, and associated splicing events, dysregulation of which, as the result of YTHDC1 depletion, can be restored by reconstitution with wild-type, but not m(6)A-binding-defective, YTHDC1. Our findings provide the direct evidence that m(6)A reader YTHDC1 regulates mRNA splicing through recruiting and modulating pre-mRNA splicing factors for their access to the binding regions of targeted mRNAs.


Subject(s)
Cell Cycle Proteins/metabolism , Nerve Tissue Proteins/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Binding Sites , Exons , HeLa Cells , Humans , RNA Splicing Factors , RNA, Messenger/metabolism , Serine-Arginine Splicing Factors
12.
Mol Cancer ; 22(1): 81, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37161388

ABSTRACT

BACKGROUND: RNA 5-methylcytosine (m5C) modification plays critical roles in the pathogenesis of various tumors. However, the function and molecular mechanism of RNA m5C modification in tumor drug resistance remain unclear. METHODS: The correlation between RNA m5C methylation, m5C writer NOP2/Sun RNA methyltransferase family member 2 (NSUN2) and EGFR-TKIs resistance was determined in non-small-cell lung cancer (NSCLC) cell lines and patient samples. The effects of NSUN2 on EGFR-TKIs resistance were investigated by gain- and loss-of-function assays in vitro and in vivo. RNA-sequencing (RNA-seq), RNA bisulfite sequencing (RNA-BisSeq) and m5C methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR) were performed to identify the target gene of NSUN2 involved in EGFR-TKIs resistance. Furthermore, the regulatory mechanism of NSUN2 modulating the target gene expression was investigated by functional rescue and puromycin incorporation assays. RESULTS: RNA m5C hypermethylation and NSUN2 were significantly correlated with intrinsic resistance to EGFR-TKIs. Overexpression of NSUN2 resulted in gefitinib resistance and tumor recurrence, while genetic inhibition of NSUN2 led to tumor regression and overcame intrinsic resistance to gefitinib in vitro and in vivo. Integrated RNA-seq and m5C-BisSeq analyses identified quiescin sulfhydryl oxidase 1 (QSOX1) as a potential target of aberrant m5C modification. NSUN2 methylated QSOX1 coding sequence region, leading to enhanced QSOX1 translation through m5C reader Y-box binding protein 1 (YBX1). CONCLUSIONS: Our study reveals a critical function of aberrant RNA m5C modification via the NSUN2-YBX1-QSOX1 axis in mediating intrinsic resistance to gefitinib in EGFR-mutant NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Gefitinib/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Neoplasm Recurrence, Local , RNA , ErbB Receptors/genetics , Y-Box-Binding Protein 1 , Oxidoreductases Acting on Sulfur Group Donors , Methyltransferases/genetics
13.
EMBO Rep ; 22(3): e51519, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33426808

ABSTRACT

The MYC oncoprotein activates and represses gene expression in a transcription-dependent or transcription-independent manner. Modification of mRNA emerges as a key gene expression regulatory nexus. We sought to determine whether MYC alters mRNA modifications and report here that MYC promotes cancer progression by down-regulating N6-methyladenosine (m6 A) preferentially in transcripts of a subset of MYC-repressed genes (MRGs). We find that MYC activates the expression of ALKBH5 and reduces m6 A levels in the mRNA of the selected MRGs SPI1 and PHF12. We also show that MYC-regulated m6 A controls the translation of MRG mRNA via the specific m6 A reader YTHDF3. Finally, we find that inhibition of ALKBH5, or overexpression of SPI1 or PHF12, effectively suppresses the growth of MYC-deregulated B-cell lymphomas, both in vitro and in vivo. Our findings uncover a novel mechanism by which MYC suppresses gene expression by altering m6 A modifications in selected MRG transcripts promotes cancer progression.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Neoplasms , Adenosine , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , RNA, Messenger/genetics
14.
Nature ; 549(7671): 273-276, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28869969

ABSTRACT

N6-methyladenosine (m6A) has been identified as the most abundant modification on eukaryote messenger RNA (mRNA). Although the rapid development of high-throughput sequencing technologies has enabled insight into the biological functions of m6A modification, the function of m6A during vertebrate embryogenesis remains poorly understood. Here we show that m6A determines cell fate during the endothelial-to-haematopoietic transition (EHT) to specify the earliest haematopoietic stem/progenitor cells (HSPCs) during zebrafish embryogenesis. m6A-specific methylated RNA immunoprecipitation combined with high-throughput sequencing (MeRIP-seq) and m6A individual-nucleotide-resolution cross-linking and immunoprecipitation with sequencing (miCLIP-seq) analyses reveal conserved features on zebrafish m6A methylome and preferential distribution of m6A peaks near the stop codon with a consensus RRACH motif. In mettl3-deficient embryos, levels of m6A are significantly decreased and emergence of HSPCs is blocked. Mechanistically, we identify that the delayed YTHDF2-mediated mRNA decay of the arterial endothelial genes notch1a and rhoca contributes to this deleterious effect. The continuous activation of Notch signalling in arterial endothelial cells of mettl3-deficient embryos blocks EHT, thereby repressing the generation of the earliest HSPCs. Furthermore, knockdown of Mettl3 in mice confers a similar phenotype. Collectively, our findings demonstrate the critical function of m6A modification in the fate determination of HSPCs during vertebrate embryogenesis.


Subject(s)
Adenosine/analogs & derivatives , Cell Differentiation , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , RNA, Messenger/metabolism , Zebrafish/embryology , Adenosine/metabolism , Animals , Cell Differentiation/genetics , Codon, Terminator/genetics , Consensus Sequence , Endothelial Cells/metabolism , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Homeodomain Proteins/genetics , Immunoprecipitation , Methylation , Methyltransferases/deficiency , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Nerve Tissue Proteins/genetics , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/genetics , Receptor, Notch1/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
15.
Semin Cancer Biol ; 75: 97-115, 2021 10.
Article in English | MEDLINE | ID: mdl-33220459

ABSTRACT

RNA methylations, as the prevalent post-transcriptional modifications, are critical in regulating various biological processes, such as RNA transcription, splicing, structure, stability, and translation. Its dysregulation is closely related to the occurrence of human malignancies. The advance of high-throughput sequencing technology facilitates the investigations about how methylation of coding and non-coding RNAs regulates cancer progression through reshaping the transcriptomics. Here, we review the current progress about the regulatory role of several representative RNA modifications in cancers, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A) and 2'-O-methylation (Nm). Meanwhile, we also discuss the potential clinical value of RNA methylation in diagnostic and therapeutic implications of human cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Animals , Biomarkers, Tumor/genetics , Humans , Methylation , Neoplasms/genetics , Neoplasms/metabolism
16.
Nucleic Acids Res ; 48(19): 11083-11096, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33035345

ABSTRACT

N6-Methyladenosine (m6A) messenger RNA methylation is a well-known epitranscriptional regulatory mechanism affecting central biological processes, but its function in human cellular senescence remains uninvestigated. Here, we found that levels of both m6A RNA methylation and the methyltransferase METTL3 were reduced in prematurely senescent human mesenchymal stem cell (hMSC) models of progeroid syndromes. Transcriptional profiling of m6A modifications further identified MIS12, for which m6A modifications were reduced in both prematurely senescent hMSCs and METTL3-deficient hMSCs. Knockout of METTL3 accelerated hMSC senescence whereas overexpression of METTL3 rescued the senescent phenotypes. Mechanistically, loss of m6A modifications accelerated the turnover and decreased the expression of MIS12 mRNA while knockout of MIS12 accelerated cellular senescence. Furthermore, m6A reader IGF2BP2 was identified as a key player in recognizing and stabilizing m6A-modified MIS12 mRNA. Taken together, we discovered that METTL3 alleviates hMSC senescence through m6A modification-dependent stabilization of the MIS12 transcript, representing a novel epitranscriptional mechanism in premature stem cell senescence.


Subject(s)
Adenosine/analogs & derivatives , Methyltransferases/metabolism , Microtubule-Associated Proteins/genetics , Progeria/genetics , RNA, Messenger/metabolism , Werner Syndrome/genetics , Adenosine/genetics , Cells, Cultured , Cellular Senescence , Humans , Mesenchymal Stem Cells , Methylation , RNA-Binding Proteins/metabolism
17.
Nat Methods ; 15(3): 213-220, 2018 03.
Article in English | MEDLINE | ID: mdl-29431736

ABSTRACT

We combine the labeling of newly transcribed RNAs with 5-ethynyluridine with the characterization of bound proteins. This approach, named capture of the newly transcribed RNA interactome using click chemistry (RICK), systematically captures proteins bound to a wide range of RNAs, including nascent RNAs and traditionally neglected nonpolyadenylated RNAs. RICK has identified mitotic regulators amongst other novel RNA-binding proteins with preferential affinity for nonpolyadenylated RNAs, revealed a link between metabolic enzymes/factors and nascent RNAs, and expanded the known RNA-bound proteome of mouse embryonic stem cells. RICK will facilitate an in-depth interrogation of the total RNA-bound proteome in different cells and systems.


Subject(s)
Click Chemistry/methods , Proteome/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , HeLa Cells , High-Throughput Nucleotide Sequencing/methods , Humans , Mass Spectrometry/methods , Mice , Protein Interaction Maps , RNA/genetics , RNA-Binding Proteins/genetics , Uridine/analogs & derivatives , Uridine/chemistry
18.
PLoS Biol ; 16(6): e2004880, 2018 06.
Article in English | MEDLINE | ID: mdl-29879109

ABSTRACT

N6-methyladenosine (m6A) RNA methylation is the most abundant modification on mRNAs and plays important roles in various biological processes. The formation of m6A is catalyzed by a methyltransferase complex including methyltransferase-like 3 (METTL3) as a key factor. However, the in vivo functions of METTL3 and m6A modification in mammalian development remain unclear. Here, we show that specific inactivation of Mettl3 in mouse nervous system causes severe developmental defects in the brain. Mettl3 conditional knockout (cKO) mice manifest cerebellar hypoplasia caused by drastically enhanced apoptosis of newborn cerebellar granule cells (CGCs) in the external granular layer (EGL). METTL3 depletion-induced loss of m6A modification causes extended RNA half-lives and aberrant splicing events, consequently leading to dysregulation of transcriptome-wide gene expression and premature CGC death. Our findings reveal a critical role of METTL3-mediated m6A in regulating the development of mammalian cerebellum.


Subject(s)
Adenosine/analogs & derivatives , Cerebellum/embryology , Methyltransferases/metabolism , RNA, Messenger/genetics , Adenosine/metabolism , Alternative Splicing/genetics , Animals , Apoptosis/genetics , Cells, Cultured , Cerebellum/abnormalities , Cerebellum/pathology , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Gene Expression Regulation/genetics , Methylation , Methyltransferases/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Nervous System Malformations/genetics , Nervous System Malformations/pathology , RNA Stability/genetics , RNA, Messenger/metabolism
19.
RNA Biol ; 18(6): 875-887, 2021 06.
Article in English | MEDLINE | ID: mdl-32991228

ABSTRACT

The pluripotency of embryonic stem cells (ESCs) is controlled by a multilayer regulatory network, of which the key factors include core pluripotency genes Oct4, Sox2 and Nanog, and multiple microRNAs (miRNAs). Recently, long noncoding RNAs (lncRNAs) have been discovered as a class of new regulators for ESCs, and some lncRNAs could function as competing endogenous RNAs (ceRNAs) to regulate mRNAs by competitively binding to miRNAs. Here, we identify mmu-miR-139-5p as a new regulator for Nanog by targeting Nanog 3' untranslated region (UTR) to repress Nanog expression in mouse ESCs and embryos. Such regulation could be released by an ESC-specifically expressed ceRNA named lnc-NAP. The expression of lnc-NAP is activated by OCT4, SOX2, as well as NANOG through promoter binding. Downregulation of lnc-NAP reduces Nanog abundance, which leads to decreased pluripotency of mouse ESCs and embryonic lethality. These results reveal lnc-NAP as a new regulator for Nanog in mouse ESCs, and uncover a feed-forward regulatory loop of Nanog through the participation of lnc-NAP.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Nanog Homeobox Protein/genetics , RNA, Long Noncoding/genetics , 3' Untranslated Regions/genetics , Animals , Cell Differentiation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Stem Cells/cytology , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Mice, SCID , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA-Seq/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
20.
Mol Cell ; 49(1): 18-29, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23177736

ABSTRACT

N(6)-methyladenosine (m(6)A) is the most prevalent internal modification of messenger RNA (mRNA) in higher eukaryotes. Here we report ALKBH5 as another mammalian demethylase that oxidatively reverses m(6)A in mRNA in vitro and in vivo. This demethylation activity of ALKBH5 significantly affects mRNA export and RNA metabolism as well as the assembly of mRNA processing factors in nuclear speckles. Alkbh5-deficient male mice have increased m(6)A in mRNA and are characterized by impaired fertility resulting from apoptosis that affects meiotic metaphase-stage spermatocytes. In accordance with this defect, we have identified in mouse testes 1,551 differentially expressed genes that cover broad functional categories and include spermatogenesis-related mRNAs involved in the p53 functional interaction network. The discovery of this RNA demethylase strongly suggests that the reversible m(6)A modification has fundamental and broad functions in mammalian cells.


Subject(s)
Dioxygenases/metabolism , Membrane Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , AlkB Homolog 5, RNA Demethylase , Animals , Base Sequence , Cell Nucleus/metabolism , Dioxygenases/chemistry , Dioxygenases/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Infertility, Male/enzymology , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Knockout , Organ Size , Oxidoreductases, N-Demethylating/chemistry , Oxidoreductases, N-Demethylating/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport , RNA Interference , RNA, Messenger/chemistry , Spermatogenesis/genetics , Testis/enzymology , Testis/pathology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL