Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.655
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39305902

ABSTRACT

m6A modification is best known for its critical role in controlling multiple post-transcriptional processes of the mRNAs. Here, we discovered elevated levels of m6A modification on centromeric RNA (cenRNA) in cancerous cells compared with non-cancerous cells. We then identified CENPA, an H3 variant, as an m6A reader of cenRNA. CENPA is localized at centromeres and is essential in preserving centromere integrity and function during mitosis. The m6A-modified cenRNA stabilizes centromeric localization of CENPA in cancer cells during the S phase of the cell cycle. Mutations of CENPA at the Leu61 and the Arg63 or removal of cenRNA m6A modification lead to loss of centromere-bound CENPA during S phase. This in turn results in compromised centromere integrity and abnormal chromosome separation and hinders cancer cell proliferation and tumor growth. Our findings unveil an m6A reading mechanism by CENPA that epigenetically governs centromere integrity in cancer cells, providing potential targets for cancer therapy.

2.
Annu Rev Biochem ; 92: 145-173, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37068770

ABSTRACT

Over the past decade, mRNA modifications have emerged as important regulators of gene expression control in cells. Fueled in large part by the development of tools for detecting RNA modifications transcriptome wide, researchers have uncovered a diverse epitranscriptome that serves as an additional layer of gene regulation beyond simple RNA sequence. Here, we review the proteins that write, read, and erase these marks, with a particular focus on the most abundant internal modification, N6-methyladenosine (m6A). We first describe the discovery of the key enzymes that deposit and remove m6A and other modifications and discuss how our understanding of these proteins has shaped our views of modification dynamics. We then review current models for the function of m6A reader proteins and how our knowledge of these proteins has evolved. Finally, we highlight important future directions for the field and discuss key questions that remain unanswered.


Subject(s)
Adenosine , Gene Expression Regulation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Adenosine/genetics , Adenosine/metabolism , Proteins/genetics , Proteins/metabolism , Transcriptome
3.
Cell ; 184(12): 3125-3142.e25, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33930289

ABSTRACT

The N6-methyladenosine (m6A) RNA modification is used widely to alter the fate of mRNAs. Here we demonstrate that the C. elegans writer METT-10 (the ortholog of mouse METTL16) deposits an m6A mark on the 3' splice site (AG) of the S-adenosylmethionine (SAM) synthetase pre-mRNA, which inhibits its proper splicing and protein production. The mechanism is triggered by a rich diet and acts as an m6A-mediated switch to stop SAM production and regulate its homeostasis. Although the mammalian SAM synthetase pre-mRNA is not regulated via this mechanism, we show that splicing inhibition by 3' splice site m6A is conserved in mammals. The modification functions by physically preventing the essential splicing factor U2AF35 from recognizing the 3' splice site. We propose that use of splice-site m6A is an ancient mechanism for splicing regulation.


Subject(s)
Adenosine/analogs & derivatives , RNA Splice Sites/genetics , RNA Splicing/genetics , Splicing Factor U2AF/metabolism , Adenosine/metabolism , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans/genetics , Conserved Sequence/genetics , Diet , HeLa Cells , Humans , Introns/genetics , Methionine Adenosyltransferase , Methylation , Methyltransferases/chemistry , Mice , Mutation/genetics , Nucleic Acid Conformation , Protein Binding , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear , S-Adenosylmethionine , Transcriptome/genetics
4.
Cell ; 181(7): 1582-1595.e18, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32492408

ABSTRACT

N6-methyladenosine (m6A) is the most abundant mRNA nucleotide modification and regulates critical aspects of cellular physiology and differentiation. m6A is thought to mediate its effects through a complex network of interactions between different m6A sites and three functionally distinct cytoplasmic YTHDF m6A-binding proteins (DF1, DF2, and DF3). In contrast to the prevailing model, we show that DF proteins bind the same m6A-modified mRNAs rather than different mRNAs. Furthermore, we find that DF proteins do not induce translation in HeLa cells. Instead, the DF paralogs act redundantly to mediate mRNA degradation and cellular differentiation. The ability of DF proteins to regulate stability and differentiation becomes evident only when all three DF paralogs are depleted simultaneously. Our study reveals a unified model of m6A function in which all m6A-modified mRNAs are subjected to the combined action of YTHDF proteins in proportion to the number of m6A sites.


Subject(s)
Adenosine/analogs & derivatives , RNA-Binding Proteins/metabolism , Adenosine/genetics , Adenosine/metabolism , Cell Differentiation , HeLa Cells , Humans , Methylation , Methyltransferases/metabolism , Protein Biosynthesis , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
5.
Cell ; 172(1-2): 90-105.e23, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29249359

ABSTRACT

R-2-hydroxyglutarate (R-2HG), produced at high levels by mutant isocitrate dehydrogenase 1/2 (IDH1/2) enzymes, was reported as an oncometabolite. We show here that R-2HG also exerts a broad anti-leukemic activity in vitro and in vivo by inhibiting leukemia cell proliferation/viability and by promoting cell-cycle arrest and apoptosis. Mechanistically, R-2HG inhibits fat mass and obesity-associated protein (FTO) activity, thereby increasing global N6-methyladenosine (m6A) RNA modification in R-2HG-sensitive leukemia cells, which in turn decreases the stability of MYC/CEBPA transcripts, leading to the suppression of relevant pathways. Ectopically expressed mutant IDH1 and S-2HG recapitulate the effects of R-2HG. High levels of FTO sensitize leukemic cells to R-2HG, whereas hyperactivation of MYC signaling confers resistance that can be reversed by the inhibition of MYC signaling. R-2HG also displays anti-tumor activity in glioma. Collectively, while R-2HG accumulated in IDH1/2 mutant cancers contributes to cancer initiation, our work demonstrates anti-tumor effects of 2HG in inhibiting proliferation/survival of FTO-high cancer cells via targeting FTO/m6A/MYC/CEBPA signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Glutarates/pharmacology , Leukemia/drug therapy , Signal Transduction/drug effects , Adenosine/analogs & derivatives , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Antineoplastic Agents/therapeutic use , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Glutarates/therapeutic use , HEK293 Cells , Humans , Jurkat Cells , Mice , Proto-Oncogene Proteins c-myc/metabolism , RNA Processing, Post-Transcriptional
6.
Cell ; 169(2): 326-337.e12, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388414

ABSTRACT

Transcription and translation are two main pillars of gene expression. Due to the different timings, spots of action, and mechanisms of regulation, these processes are mainly regarded as distinct and generally uncoupled, despite serving a common purpose. Here, we sought for a possible connection between transcription and translation. Employing an unbiased screen of multiple human promoters, we identified a positive effect of TATA box on translation and a general coupling between mRNA expression and translational efficiency. Using a CRISPR-Cas9-mediated approach, genome-wide analyses, and in vitro experiments, we show that the rate of transcription regulates the efficiency of translation. Furthermore, we demonstrate that m6A modification of mRNAs is co-transcriptional and depends upon the dynamics of the transcribing RNAPII. Suboptimal transcription rates lead to elevated m6A content, which may result in reduced translation. This study uncovers a general and widespread link between transcription and translation that is governed by epigenetic modification of mRNAs.


Subject(s)
Adenosine/analogs & derivatives , Gene Expression Regulation , Protein Biosynthesis , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Transcription, Genetic , Adenosine/metabolism , Humans , Methylation , Peptide Chain Initiation, Translational , RNA Polymerase II/metabolism , TATA Box
7.
Cell ; 171(4): 877-889.e17, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28965759

ABSTRACT

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.


Subject(s)
Neurogenesis , Prosencephalon/embryology , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Animals , Cell Cycle , Gene Expression Regulation , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Organoids/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , RNA Stability
8.
Cell ; 169(5): 824-835.e14, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525753

ABSTRACT

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.


Subject(s)
Introns , Methionine Adenosyltransferase/genetics , Methyltransferases/metabolism , RNA Splicing , S-Adenosylmethionine/metabolism , Animals , Base Sequence , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Inverted Repeat Sequences , Methionine Adenosyltransferase/chemistry , Methylation , Methyltransferases/chemistry , Schizosaccharomyces/metabolism
9.
Mol Cell ; 84(19): 3810-3825.e10, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39303720

ABSTRACT

Cys2-His2 zinc-finger proteins (C2H2-ZNFs) constitute the largest class of DNA-binding transcription factors (TFs) yet remain largely uncharacterized. Although certain family members, e.g., GTF3A, have been shown to bind both DNA and RNA, the extent to which C2H2-ZNFs interact with-and regulate-RNA-associated processes is not known. Using UV crosslinking and immunoprecipitation (CLIP), we observe that 148 of 150 analyzed C2H2-ZNFs bind directly to RNA in human cells. By integrating CLIP sequencing (CLIP-seq) RNA-binding maps for 50 of these C2H2-ZNFs with data from chromatin immunoprecipitation sequencing (ChIP-seq), protein-protein interaction assays, and transcriptome profiling experiments, we observe that the RNA-binding profiles of C2H2-ZNFs are generally distinct from their DNA-binding preferences and that they regulate a variety of post-transcriptional processes, including pre-mRNA splicing, cleavage and polyadenylation, and m6A modification of mRNA. Our results thus define a substantially expanded repertoire of C2H2-ZNFs that bind RNA and provide an important resource for elucidating post-transcriptional regulatory programs.


Subject(s)
Protein Binding , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , CYS2-HIS2 Zinc Fingers/genetics , RNA Processing, Post-Transcriptional , RNA Splicing , Binding Sites , RNA/metabolism , RNA/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , HEK293 Cells , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Chromatin Immunoprecipitation Sequencing , Polyadenylation , Gene Expression Regulation
10.
Mol Cell ; 84(19): 3758-3774.e10, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39127036

ABSTRACT

N6-methyladenosine (m6A) modification is deemed to be co-transcriptionally installed on pre-mRNAs, thereby influencing various downstream RNA metabolism events. However, the causal relationship between m6A modification and RNA processing is often unclear, resulting in premature or even misleading generalizations on the function of m6A modification. Here, we develop 4sU-coupled m6A-level and isoform-characterization sequencing (4sU-m6A-LAIC-seq) and 4sU-GLORI to quantify the m6A levels for both newly synthesized and steady-state RNAs at transcript and single-base-resolution levels, respectively, which enable dissecting the relationship between m6A modification and alternative RNA polyadenylation. Unexpectedly, our results show that many m6A addition events occur post-transcriptionally, especially on transcripts with high m6A levels. Importantly, we find higher m6A levels on shorter 3' UTR isoforms, which likely result from sequential polyadenylation of longer 3' UTR isoforms with prolonged nuclear dwelling time. Therefore, m6A modification can also take place post-transcriptionally to intimately couple with other key RNA metabolism processes to establish and dynamically regulate epi-transcriptomics in mammalian cells.


Subject(s)
Adenosine , Cell Nucleus , Polyadenylation , RNA Processing, Post-Transcriptional , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/genetics , Humans , Cell Nucleus/metabolism , Cell Nucleus/genetics , 3' Untranslated Regions , RNA, Messenger/metabolism , RNA, Messenger/genetics , HEK293 Cells , Methyltransferases/metabolism , Methyltransferases/genetics , HeLa Cells , Animals
11.
Mol Cell ; 84(3): 584-595.e6, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38244546

ABSTRACT

The most abundant N6-methyladenosine (m6A) modification on mRNAs is installed non-stoichiometrically across transcripts, with 5' untranslated regions (5' UTRs) being the least conductive. 5' UTRs are essential for translation initiation, yet the molecular mechanisms orchestrated by m6A remain poorly understood. Here, we combined structural, biochemical, and single-molecule approaches and show that at the most common position, a single m6A does not affect translation yields, the kinetics of translation initiation complex assembly, or start codon recognition both under permissive growth and following exposure to oxidative stress. Cryoelectron microscopy (cryo-EM) structures of the late preinitiation complex reveal that m6A purine ring established stacking interactions with an arginine side chain of the initiation factor eIF2α, although with only a marginal energy contribution, as estimated computationally. These findings provide molecular insights into m6A interactions with the initiation complex and suggest that the subtle stabilization is unlikely to affect the translation dynamics under homeostatic conditions or stress.


Subject(s)
Adenosine/analogs & derivatives , Peptide Chain Initiation, Translational , Protein Biosynthesis , 5' Untranslated Regions , Cryoelectron Microscopy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Codon, Initiator/genetics
12.
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
13.
Mol Cell ; 84(19): 3610-3626, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39366350

ABSTRACT

Complex pathways involving the DNA damage response (DDR) contend with cell-intrinsic and -extrinsic sources of DNA damage. DDR mis-regulation results in genome instability that can contribute to aging and diseases including cancer and neurodegeneration. Recent studies have highlighted key roles for several RNA species in the DDR, including short RNAs and RNA/DNA hybrids (R-loops) at DNA break sites, all contributing to efficient DNA repair. RNAs can undergo more than 170 distinct chemical modifications. These RNA modifications have emerged as key orchestrators of the DDR. Here, we highlight the function of enzyme- and non-enzyme-induced RNA modifications in the DDR, with particular emphasis on m6A, m5C, and RNA editing. We also discuss stress-induced RNA damage, including RNA alkylation/oxidation, RNA-protein crosslinks, and UV-induced RNA damage. Uncovering molecular mechanisms that underpin the contribution of RNA modifications to DDR and genome stability will have direct application to disease and approaches for therapeutic intervention.


Subject(s)
DNA Damage , DNA Repair , Epigenesis, Genetic , RNA , Humans , Animals , RNA/metabolism , RNA/genetics , Transcriptome , RNA Processing, Post-Transcriptional , Genomic Instability , RNA Editing , Adenosine/metabolism , Adenosine/analogs & derivatives , Adenosine/genetics
14.
Annu Rev Cell Dev Biol ; 33: 319-342, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28759256

ABSTRACT

In recent years, m6A has emerged as an abundant and dynamically regulated modification throughout the transcriptome. Recent technological advances have enabled the transcriptome-wide identification of m6A residues, which in turn has provided important insights into the biology and regulation of this pervasive regulatory mark. Also central to our current understanding of m6A are the discovery and characterization of m6A readers, writers, and erasers. Over the last few years, studies into the function of these proteins have led to important discoveries about the regulation and function of m6A. However, during this time our understanding of these proteins has also evolved considerably, sometimes leading to the reversal of early concepts regarding the reading, writing and erasing of m6A. In this review, we summarize recent advances in m6A research, and we highlight how these new findings have reshaped our understanding of how m6A is regulated in the transcriptome.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , DNA Methylation/genetics , Humans , RNA/metabolism
15.
Mol Cell ; 83(2): 219-236.e7, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36634675

ABSTRACT

RNA methylation at adenosine N6 (m6A) is one of the most common RNA modifications, impacting RNA stability, transport, and translation. Previous studies uncovered RNA destabilization in amyotrophic lateral sclerosis (ALS) models in association with accumulation of the RNA-binding protein TDP43. Here, we show that TDP43 recognizes m6A RNA and that RNA methylation is critical for both TDP43 binding and autoregulation. We also observed extensive RNA hypermethylation in ALS spinal cord, corresponding to methylated TDP43 substrates. Emphasizing the importance of m6A for TDP43 binding and function, we identified several m6A factors that enhance or suppress TDP43-mediated toxicity via single-cell CRISPR-Cas9 in primary neurons. The most promising modifier-the canonical m6A reader YTHDF2-accumulated within ALS spinal neurons, and its knockdown prolonged the survival of human neurons carrying ALS-associated mutations. Collectively, these data show that m6A modifications modulate RNA binding by TDP43 and that m6A is pivotal for TDP43-related neurodegeneration in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Humans , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Methylation , Neurons/metabolism , RNA/genetics , RNA/metabolism
16.
Mol Cell ; 83(2): 237-251.e7, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36599352

ABSTRACT

N6-methyladenosine (m6A), a widespread destabilizing mark on mRNA, is non-uniformly distributed across the transcriptome, yet the basis for its selective deposition is unknown. Here, we propose that m6A deposition is not selective. Instead, it is exclusion based: m6A consensus motifs are methylated by default, unless they are within a window of ∼100 nt from a splice junction. A simple model which we extensively validate, relying exclusively on presence of m6A motifs and exon-intron architecture, allows in silico recapitulation of experimentally measured m6A profiles. We provide evidence that exclusion from splice junctions is mediated by the exon junction complex (EJC), potentially via physical occlusion, and that previously observed associations between exon-intron architecture and mRNA decay are mechanistically mediated via m6A. Our findings establish a mechanism coupling nuclear mRNA splicing and packaging with the covalent installation of m6A, in turn controlling cytoplasmic decay.


Subject(s)
RNA Splicing , Transcriptome , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA Stability , Exons/genetics
17.
Mol Cell ; 83(3): 428-441, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36736310

ABSTRACT

Since the early days of foundational studies of nucleic acids, many chemical moieties have been discovered to decorate RNA and DNA in diverse organisms. In mammalian cells, one of these chemical modifications, N6-methyl adenosine (m6A), is unique in a way that it is highly abundant not only on RNA polymerase II (RNAPII) transcribed, protein-coding transcripts but also on non-coding RNAs, such as ribosomal RNAs and snRNAs, mediated by distinct, evolutionarily conserved enzymes. Here, we review RNA m6A modification in the light of the recent appreciation of nuclear roles for m6A in regulating chromatin states and gene expression, as well as the recent discoveries of the evolutionarily conserved methyltransferases, which catalyze methylation of adenosine on diverse sets of RNAs. Considering that the substrates of these enzymes are involved in many important biological processes, this modification warrants further research to understand the molecular mechanisms and functions of m6A in health and disease.


Subject(s)
Methyltransferases , Transcriptome , Animals , Methylation , Methyltransferases/metabolism , Adenosine/metabolism , RNA/metabolism , Mammals/metabolism
18.
Mol Cell ; 83(12): 2003-2019.e6, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37257451

ABSTRACT

Regulation of RNA substrate selectivity of m6A demethylase ALKBH5 remains elusive. Here, we identify RNA-binding motif protein 33 (RBM33) as a previously unrecognized m6A-binding protein that plays a critical role in ALKBH5-mediated mRNA m6A demethylation of a subset of mRNA transcripts by forming a complex with ALKBH5. RBM33 recruits ALKBH5 to its m6A-marked substrate and activates ALKBH5 demethylase activity through the removal of its SUMOylation. We further demonstrate that RBM33 is critical for the tumorigenesis of head-neck squamous cell carcinoma (HNSCC). RBM33 promotes autophagy by recruiting ALKBH5 to demethylate and stabilize DDIT4 mRNA, which is responsible for the oncogenic function of RBM33 in HNSCC cells. Altogether, our study uncovers the mechanism of selectively demethylate m6A methylation of a subset of transcripts during tumorigenesis that may explain demethylation selectivity in other cellular processes, and we showed its importance in the maintenance of tumorigenesis of HNSCC.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Carcinogenesis
19.
Mol Cell ; 83(18): 3303-3313.e6, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37683648

ABSTRACT

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.


Subject(s)
Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2 , 5' Untranslated Regions , Eukaryotic Initiation Factor-2/genetics , Open Reading Frames , Phosphorylation , RNA Cap-Binding Proteins , Humans
20.
Mol Cell ; 83(21): 3818-3834.e7, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37820733

ABSTRACT

N6-methyladenosine (m6A) modifications play crucial roles in RNA metabolism. How m6A regulates RNA polymerase II (RNA Pol II) transcription remains unclear. We find that 7SK small nuclear RNA (snRNA), a regulator of RNA Pol II promoter-proximal pausing, is highly m6A-modified in non-small cell lung cancer (NSCLC) cells. In A549 cells, we identified eight m6A sites on 7SK and discovered methyltransferase-like 3 (METTL3) and alkB homolog 5 (ALKBH5) as the responsible writer and eraser. When the m6A-7SK is specifically erased by a dCasRx-ALKBH5 fusion protein, A549 cell growth is attenuated due to reduction of RNA Pol II transcription. Mechanistically, removal of m6A leads to 7SK structural rearrangements that facilitate sequestration of the positive transcription elongation factor b (P-TEFb) complex, which results in reduction of serine 2 phosphorylation (Ser2P) in the RNA Pol II C-terminal domain and accumulation of RNA Pol II in the promoter-proximal region. Taken together, we uncover that m6A modifications of a non-coding RNA regulate RNA Pol II transcription and NSCLC tumorigenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Positive Transcriptional Elongation Factor B/genetics , Lung Neoplasms/genetics , RNA, Small Nuclear/genetics , Transcription, Genetic , HeLa Cells , Methyltransferases/genetics , Methyltransferases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL