Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Plant Cell ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38835286

ABSTRACT

N 6-methyladenosine (m6A) is the most abundant mRNA modification and plays diverse roles in eukaryotes, including plants. It regulates various processes, including plant growth, development, and responses to external or internal stress responses. However, the mechanisms underlying how m6A is related to environmental stresses in both mammals and plants remain elusive. Here, we identified EVOLUTIONARILY CONSERVED C-TERMINAL REGION 8 (ECT8) as an m6A reader protein and showed that its m6A-binding capability is required for salt stress responses in Arabidopsis (Arabidopsis thaliana). ECT8 accelerates the degradation of its target transcripts through direct interaction with the decapping protein DECAPPING 5 within processing bodies. We observed a significant increase in the ECT8 expression level under various environmental stresses. Using salt stress as a representative stressor, we found that the transcript and protein levels of ECT8 rise in response to salt stress. The increased abundance of ECT8 protein results in the enhanced binding capability to m6A-modified mRNAs, thereby accelerating their degradation, especially those of negative regulators of salt stress responses. Our results demonstrated that ECT8 acts as an abiotic stress sensor, facilitating mRNA decay, which is vital for maintaining transcriptome homeostasis and enhancing stress tolerance in plants. Our findings not only advance the understanding of epitranscriptomic gene regulation but also offer potential applications for breeding more resilient crops in the face of rapidly changing environmental conditions.

2.
Proc Natl Acad Sci U S A ; 121(28): e2405100121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38950372

ABSTRACT

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.


Subject(s)
Adenosine , Fruit , Gene Expression Regulation, Plant , Plant Proteins , Protein Biosynthesis , Solanum lycopersicum , Solanum lycopersicum/genetics , Solanum lycopersicum/metabolism , Solanum lycopersicum/growth & development , Fruit/metabolism , Fruit/genetics , Adenosine/metabolism , Adenosine/analogs & derivatives , Plant Proteins/metabolism , Plant Proteins/genetics , Odorants/analysis
3.
RNA ; 30(5): 491-499, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38531642

ABSTRACT

Over the past decade, N 6-methyladenosine (m6A) has emerged as a prevalent and dynamically regulated modification across the transcriptome; it has been reversibly installed, removed, and interpreted by specific binding proteins, and has played crucial roles in molecular and biological processes. Within this scope, we consolidate recent advancements of m6A research in plants regarding gene expression regulation, diverse physiologic and pathogenic processes, as well as crop trial implications, to guide discussions on challenges associated with and leveraging epitranscriptome editing for crop improvement.


Subject(s)
Gene Expression Regulation , Plants , Plants/genetics , Transcriptome
4.
Mol Cell ; 71(6): 973-985.e5, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30197295

ABSTRACT

FTO, the first RNA demethylase discovered, mediates the demethylation of internal N6-methyladenosine (m6A) and N6, 2-O-dimethyladenosine (m6Am) at the +1 position from the 5' cap in mRNA. Here we demonstrate that the cellular distribution of FTO is distinct among different cell lines, affecting the access of FTO to different RNA substrates. We find that FTO binds multiple RNA species, including mRNA, snRNA, and tRNA, and can demethylate internal m6A and cap m6Am in mRNA, internal m6A in U6 RNA, internal and cap m6Am in snRNAs, and N1-methyladenosine (m1A) in tRNA. FTO-mediated demethylation has a greater effect on the transcript levels of mRNAs possessing internal m6A than the ones with cap m6Am in the tested cells. We also show that FTO can directly repress translation by catalyzing m1A tRNA demethylation. Collectively, FTO-mediated RNA demethylation occurs to m6A and m6Am in mRNA and snRNA as well as m1A in tRNA.


Subject(s)
Adenosine/analogs & derivatives , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/physiology , 3T3-L1 Cells , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Cell Nucleus , Cytoplasm , Demethylation , Gene Expression/genetics , HEK293 Cells , HeLa Cells , Humans , Methylation , Mice , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , RNA, Transfer/metabolism
5.
Nucleic Acids Res ; 51(D1): D106-D116, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36382409

ABSTRACT

With advanced technologies to map RNA modifications, our understanding of them has been revolutionized, and they are seen to be far more widespread and important than previously thought. Current next-generation sequencing (NGS)-based modification profiling methods are blind to RNA modifications and thus require selective chemical treatment or antibody immunoprecipitation methods for particular modification types. They also face the problem of short read length, isoform ambiguities, biases and artifacts. Direct RNA sequencing (DRS) technologies, commercialized by Oxford Nanopore Technologies (ONT), enable the direct interrogation of any given modification present in individual transcripts and promise to address the limitations of previous NGS-based methods. Here, we present the first ONT-based database of quantitative RNA modification profiles, DirectRMDB, which includes 16 types of modification and a total of 904,712 modification sites in 25 species identified from 39 independent studies. In addition to standard functions adopted by existing databases, such as gene annotations and post-transcriptional association analysis, we provide a fresh view of RNA modifications, which enables exploration of the epitranscriptome in an isoform-specific manner. The DirectRMDB database is freely available at: http://www.rnamd.org/directRMDB/.


Subject(s)
High-Throughput Nucleotide Sequencing , RNA Processing, Post-Transcriptional , Sequence Analysis, RNA , High-Throughput Nucleotide Sequencing/methods , Molecular Sequence Annotation , Protein Isoforms , RNA/genetics , Sequence Analysis, RNA/methods , Databases, Nucleic Acid
6.
PLoS Genet ; 18(9): e1010386, 2022 09.
Article in English | MEDLINE | ID: mdl-36166469

ABSTRACT

Adenosine bases of RNA can be transiently modified by the deposition of a methyl-group to form N6-methyladenosine (m6A). This adenosine-methylation is an ancient process and the enzymes involved are evolutionary highly conserved. A genetic screen designed to identify suppressors of late flowering transgenic Arabidopsis plants overexpressing the miP1a microProtein yielded a new allele of the FIONA1 (FIO1) m6A-methyltransferase. To characterize the early flowering phenotype of fio1 mutant plants we employed an integrative approach of mRNA-seq, Nanopore direct RNA-sequencing and meRIP-seq to identify differentially expressed transcripts as well as differentially methylated RNAs. We provide evidence that FIO1 is the elusive methyltransferase responsible for the 3'-end methylation of the FLOWERING LOCUS C (FLC) transcript. Furthermore, our genetic and biochemical data suggest that 3'-methylation stabilizes FLC mRNAs and non-methylated FLC is a target for rapid degradation.


Subject(s)
Arabidopsis Proteins , Arabidopsis , 3' Untranslated Regions/genetics , Adenosine/genetics , Adenosine/metabolism , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Flowers/genetics , Flowers/metabolism , Gene Expression Regulation, Plant , Histones/genetics , MADS Domain Proteins/genetics , MADS Domain Proteins/metabolism , Methylation , Methyltransferases/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Nat Methods ; 18(10): 1213-1222, 2021 10.
Article in English | MEDLINE | ID: mdl-34594034

ABSTRACT

Recent years have witnessed rapid progress in the field of epitranscriptomics. Functional interpretation of the epitranscriptome relies on sequencing technologies that determine the location and stoichiometry of various RNA modifications. However, contradictory results have been reported among studies, bringing the biological impacts of certain RNA modifications into doubt. Here, we develop a synthetic RNA library resembling the endogenous transcriptome but without any RNA modification. By incorporating this modification-free RNA library into established mapping techniques as a negative control, we reveal abundant false positives resulting from sequence bias or RNA structure. After calibration, precise and quantitative mapping expands the understanding of two representative modification types, N6-methyladenosine (m6A) and 5-methylcytosine (m5C). We propose that this approach provides a systematic solution for the calibration of various RNA-modification mappings and holds great promise in epitranscriptomic studies.


Subject(s)
Epigenesis, Genetic , Gene Library , High-Throughput Nucleotide Sequencing/methods , RNA/genetics , Transcriptome , Calibration , Gene Expression Regulation , HeLa Cells , Humans
8.
Plant Biotechnol J ; 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38634166

ABSTRACT

The N6-methyladenosine (m6A) mRNA modification is crucial for plant development and stress responses. In rice, the male sterility resulting from the deficiency of OsFIP37, a core component of m6A methyltransferase complex, emphasizes the significant role of m6A in male fertility. m6A is reversible and can be removed by m6A demethylases. However, whether mRNA m6A demethylase regulates male fertility in rice has remained unknown. Here, we identify the mRNA m6A demethylase OsALKBH9 and demonstrate its involvement in male fertility regulation. Knockout of OsALKBH9 causes male sterility, dependent on its m6A demethylation activity. Cytological analysis reveals defective tapetal programmed cell death (PCD) and excessive accumulation of microspores exine in Osalkbh9-1. Transcriptome analysis of anthers shows up-regulation of genes involved in tapetum development, sporopollenin synthesis, and transport pathways in Osalkbh9-1. Additionally, we demonstrate that OsALKBH9 demethylates the m6A modification in TDR and GAMYB transcripts, which affects the stability of these mRNAs and ultimately leads to excessive accumulation of pollen exine. Our findings highlight the precise control of mRNA m6A modification and reveal the pivotal roles played by OsALKBH9-mediated m6A demethylation in tapetal PCD and pollen exine accumulation in rice.

9.
Angew Chem Int Ed Engl ; 63(7): e202313900, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38158383

ABSTRACT

N1 -methyladenosine (m1 A) is a prevalent post-transcriptional RNA modification, and the distribution and dynamics of the modification play key epitranscriptomic roles in cell development. At present, the human AlkB Fe(II)/α-ketoglutarate-dependent dioxygenase family member ALKBH3 is the only known mRNA m1 A demethylase, but its catalytic mechanism remains unclear. Here, we present the structures of ALKBH3-oligo crosslinked complexes obtained with the assistance of a synthetic antibody crystallization chaperone. Structural and biochemical results showed that ALKBH3 utilized two ß-hairpins (ß4-loop-ß5 and ß'-loop-ß'') and the α2 helix to facilitate single-stranded substrate binding. Moreover, a bubble-like region around Asp194 and a key residue inside the active pocket (Thr133) enabled specific recognition and demethylation of m1 A- and 3-methylcytidine (m3 C)-modified substrates. Mutation of Thr133 to the corresponding residue in the AlkB Fe(II)/α-ketoglutarate-dependent dioxygenase family members FTO or ALKBH5 converted ALKBH3 substrate selectivity from m1 A to N6 -methyladenosine (m6 A), as did Asp194 deletion. Our findings provide a molecular basis for understanding the mechanisms of substrate recognition and m1 A demethylation by ALKBH3. This study is expected to aid structure-guided design of chemical probes for further functional studies and therapeutic applications.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , RNA , Humans , RNA/chemistry , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , RNA, Messenger/metabolism , Demethylation , Ferrous Compounds , AlkB Homolog 3, Alpha-Ketoglutarate-Dependent Dioxygenase/metabolism
10.
J Biol Chem ; 297(2): 100973, 2021 08.
Article in English | MEDLINE | ID: mdl-34280435

ABSTRACT

N6-methyladenosine (m6A) is the most frequent chemical modification in eukaryotic mRNA and is known to participate in a variety of physiological processes, including cancer progression and viral infection. The reversible and dynamic m6A modification is installed by m6A methyltransferase (writer) enzymes and erased by m6A demethylase (eraser) enzymes. m6A modification recognized by m6A binding proteins (readers) regulates RNA processing and metabolism, leading to downstream biological effects such as promotion of stability and translation or increased degradation. The m6A writers and erasers determine the abundance of m6A modifications and play decisive roles in its distribution and function. In this review, we focused on m6A writers and erasers and present an overview on their known functions and enzymatic molecular mechanisms, showing how they recognize substrates and install or remove m6A modifications. We also summarize the current applications of m6A writers and erasers for m6A detection and highlight the merits and drawbacks of these available methods. Lastly, we describe the biological functions of m6A in cancers and viral infection based on research of m6A writers and erasers and introduce new assays for m6A functionality via programmable m6A editing tools.


Subject(s)
Adenosine/analogs & derivatives , Eukaryotic Cells/metabolism , Methyltransferases/metabolism , Neoplasms/pathology , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , Adenosine/chemistry , Adenosine/genetics , Adenosine/metabolism , Humans , Neoplasms/genetics , Neoplasms/metabolism
11.
Nat Chem Biol ; 16(8): 896-903, 2020 08.
Article in English | MEDLINE | ID: mdl-32341502

ABSTRACT

The inert chemical property of RNA modification N6-methyladenosine (m6A) makes it very challenging to detect. Most m6A sequencing methods rely on m6A-antibody immunoprecipitation and cannot distinguish m6A and N6,2'-O-dimethyladenosine modification at the cap +1 position (cap m6Am). Although the two antibody-free methods (m6A-REF-seq/MAZTER-seq and DART-seq) have been developed recently, they are dependent on m6A sequence or cellular transfection. Here, we present an antibody-free, FTO-assisted chemical labeling method termed m6A-SEAL for specific m6A detection. We applied m6A-SEAL to profile m6A landscapes in humans and plants, which displayed the known m6A distribution features in transcriptome. By doing a comparison with all available m6A sequencing methods and specific m6A sites validation by SELECT, we demonstrated that m6A-SEAL has good sensitivity, specificity and reliability for transcriptome-wide detection of m6A. Given its tagging ability and FTO's oxidation property, m6A-SEAL enables many applications such as enrichment, imaging and sequencing to drive future functional studies of m6A and other modifications.


Subject(s)
Adenosine/analogs & derivatives , Affinity Labels/chemistry , Adenosine/analysis , Adenosine/chemistry , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/chemistry , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Humans , Immunoprecipitation/methods , Methylation , RNA/chemistry , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Reproducibility of Results , Sensitivity and Specificity , Transcriptome
12.
Proc Natl Acad Sci U S A ; 116(8): 2919-2924, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718435

ABSTRACT

FTO demethylates internal N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am; at the cap +1 position) in mRNA, m6A and m6Am in snRNA, and N1-methyladenosine (m1A) in tRNA in vivo, and in vitro evidence supports that it can also demethylate N6-methyldeoxyadenosine (6mA), 3-methylthymine (3mT), and 3-methyluracil (m3U). However, it remains unclear how FTO variously recognizes and catalyzes these diverse substrates. Here we demonstrate-in vitro and in vivo-that FTO has extensive demethylation enzymatic activity on both internal m6A and cap m6Am Considering that 6mA, m6A, and m6Am all share the same nucleobase, we present a crystal structure of human FTO bound to 6mA-modified ssDNA, revealing the molecular basis of the catalytic demethylation of FTO toward multiple RNA substrates. We discovered that (i) N6-methyladenine is the most favorable nucleobase substrate of FTO, (ii) FTO displays the same demethylation activity toward internal m6A and m6Am in the same RNA sequence, suggesting that the substrate specificity of FTO primarily results from the interaction of residues in the catalytic pocket with the nucleobase (rather than the ribose ring), and (iii) the sequence and the tertiary structure of RNA can affect the catalytic activity of FTO. Our findings provide a structural basis for understanding the catalytic mechanism through which FTO demethylates its multiple substrates and pave the way forward for the structure-guided design of selective chemicals for functional studies and potential therapeutic applications.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/chemistry , Epigenesis, Genetic , RNA, Messenger/chemistry , RNA/chemistry , Adenosine/chemistry , Adenosine/metabolism , AlkB Homolog 5, RNA Demethylase/chemistry , Catalysis , DNA, Single-Stranded/chemistry , Demethylation , Deoxyadenosines/chemistry , Humans , Nucleic Acid Conformation , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity , Thymine/analogs & derivatives , Thymine/chemistry , Uracil/analogs & derivatives , Uracil/chemistry
13.
J Integr Plant Biol ; 64(12): 2361-2373, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36263999

ABSTRACT

The mRNA modification N6 -methyladenosine (m6 A) plays vital roles in plant development and biotic and abiotic stress responses. The RNA m6 A demethylase ALKBH9B can remove m6 A in alfalfa mosaic virus RNA and plays roles in alfalfa mosaic virus infection in Arabidopsis. However, it is unknown whether ALKBH9B also exhibits demethylation activity and has a biological role in endogenous plant mRNA. We demonstrated here that mRNA m6 A modification is induced by the phytohormone abscisic acid (ABA) and that ALKBH9B has m6 A demethylation activity on endogenous mRNA. Knocking out ALKBH9B led to hypersensitivity to ABA treatment during seed germination and early seedling development. We further showed that ALKBH9B removes the m6 A modification in the ABA INSENSITIVE 1 (ABI1) and BRI1-EMS-SUPPRESSOR 1 (BES1) transcripts following ABA treatment, affecting the stability of these mRNAs. Furthermore, we determined that ALKBH9B acts genetically upstream of the transcription factors ABI3 and ABI5, and its regulatory function in ABA responses depended on ABI3 and ABI5. Our findings reveal the important roles of the m6 A modification in ABA responses and highlight the role of ALKBH9B-mediated m6 A demethylation in regulating ABA responses post-transcriptionally.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/physiology , Abscisic Acid/pharmacology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , RNA , Germination/genetics , RNA, Messenger , Gene Expression Regulation, Plant , Seeds/genetics
14.
Plant Physiol ; 182(1): 332-344, 2020 01.
Article in English | MEDLINE | ID: mdl-31591151

ABSTRACT

N 6 -methyladenosine (m6A) is the most abundant modification of eukaryotic mRNA. Although m6A has been demonstrated to affect almost all aspects of RNA metabolism, its global contribution to the post-transcriptional balancing of translational efficiency remains elusive in plants. In this study, we performed a parallel analysis of the transcriptome-wide mRNA m6A distribution and polysome profiling in two maize (Zea mays) inbred lines to assess the global correlation of m6A modification with translational status. m6A sites are widely distributed in thousands of protein-coding genes, confined to a consensus motif and primarily enriched in the 3' untranslated regions, and highly coordinated with alternative polyadenylation usage, suggesting a role of m6A modification in regulating alternative polyadenylation site choice. More importantly, we identified that the m6A modification shows multifaceted correlations with the translational status depending on its strength and genic location. Moreover, we observed a substantial intraspecies variation in m6A modification, and this natural variation was shown to be partly driven by gene-specific expression and alternative splicing. Together, these findings provide an invaluable resource for ascertaining transcripts that are subject to m6A modification in maize and pave the way to a better understanding of natural m6A variation in mediating gene expression regulation.


Subject(s)
RNA, Messenger/metabolism , Zea mays/genetics , Alternative Splicing/genetics , Alternative Splicing/physiology , Gene Expression Profiling , RNA, Messenger/genetics , Transcriptome/genetics
15.
J Exp Bot ; 72(8): 2933-2946, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33606877

ABSTRACT

Heterosis has been extensively utilized to increase productivity in crops, yet the underlying molecular mechanisms remain largely elusive. Here, we generated transcriptome-wide profiles of mRNA abundance, m6A methylation, and translational efficiency from the maize F1 hybrid B73×Mo17 and its two parental lines to ascertain the contribution of each regulatory layer to heterosis at the seedling stage. We documented that although the global abundance and distribution of m6A remained unchanged, a greater number of genes had gained an m6A modification in the hybrid. Superior variations were observed at the m6A modification and translational efficiency levels when compared with mRNA abundance between the hybrid and parents. In the hybrid, the vast majority of genes with m6A modification exhibited a non-additive expression pattern, the percentage of which was much higher than that at levels of mRNA abundance and translational efficiency. Non-additive genes involved in different biological processes were hierarchically coordinated by discrete combinations of three regulatory layers. These findings suggest that transcriptional and post-transcriptional regulation of gene expression make distinct contributions to heterosis in hybrid maize. Overall, this integrated multi-omics analysis provides a valuable portfolio for interpreting transcriptional and post-transcriptional regulation of gene expression in hybrid maize, and paves the way for exploring molecular mechanisms underlying hybrid vigor.


Subject(s)
Hybrid Vigor , Zea mays , Gene Expression Profiling , Gene Expression Regulation, Plant , Hybrid Vigor/genetics , Hybridization, Genetic , Transcriptome , Zea mays/genetics
16.
Plant Cell ; 30(5): 968-985, 2018 05.
Article in English | MEDLINE | ID: mdl-29716990

ABSTRACT

The epitranscriptomic mark N6-methyladenosine (m6A) can be written, read, and erased via the action of a complex network of proteins. m6A binding proteins read m6A marks and transduce their downstream regulatory effects by altering RNA metabolic processes. The characterization of m6A readers is an essential prerequisite for understanding the roles of m6A in plants, but the identities of m6A readers have been unclear. Here, we characterized the YTH-domain family protein ECT2 as an Arabidopsis thaliana m6A reader whose m6A binding function is required for normal trichome morphology. We developed the formaldehyde cross-linking and immunoprecipitation method to identify ECT2-RNA interaction sites at the transcriptome-wide level. This analysis demonstrated that ECT2 binding sites are strongly enriched in the 3' untranslated regions (3' UTRs) of target genes and led to the identification of a plant-specific m6A motif. Sequencing analysis suggested that ECT2 plays dual roles in regulating 3' UTR processing in the nucleus and facilitating mRNA stability in the cytoplasm. Disruption of ECT2 accelerated the degradation of three ECT2 binding transcripts related to trichome morphogenesis, thereby affecting trichome branching. The results shed light on the underlying mechanisms of the roles of m6A in RNA metabolism, as well as plant development and physiology.


Subject(s)
Arabidopsis/metabolism , Trichomes/genetics , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Binding Sites , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Binding , RNA Stability/genetics , RNA Stability/physiology
17.
Cell Biol Toxicol ; 37(6): 811-831, 2021 12.
Article in English | MEDLINE | ID: mdl-34272618

ABSTRACT

Similar to epigenetic DNA and histone modifications, epitranscriptomic modifications (RNA modifications) have emerged as crucial regulators in temporal and spatial gene expression during eukaryotic development. To date, over 170 diverse types of chemical modifications have been identified upon RNA nucleobases. Some of these post-synthesized modifications can be reversibly installed, removed, and decoded by their specific cellular components and play critical roles in different biological processes. Accordingly, dysregulation of RNA modification effectors is tightly orchestrated with developmental processes. Here, we particularly focus on three well-studied RNA modifications, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), and N1-methyladenosine (m1A), and summarize recent knowledge of underlying mechanisms and critical roles of these RNA modifications in stem cell fate determination, embryonic development, and cancer progression, providing a better understanding of the whole association between epitranscriptomic regulation and mammalian development.


Subject(s)
Adenosine , Neoplasms , Adenosine/metabolism , Animals , Cell Differentiation , Methylation , Neoplasms/genetics , RNA/genetics , RNA/metabolism
18.
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
19.
Plant Cell ; 29(12): 2995-3011, 2017 12.
Article in English | MEDLINE | ID: mdl-29180595

ABSTRACT

N6-methyladenosine (m6A) is the most abundant, internal, posttranscriptional modification in mRNA among all higher eukaryotes. In mammals, this modification is reversible and plays broad roles in the regulation of mRNA metabolism and processing. Despite its importance, previous studies on the role and mechanism of m6A methylation in Arabidopsis thaliana have been limited. Here, we report that ALKBH10B is a demethylase that oxidatively reverses m6A methylation in mRNA in vitro and in vivo. Depletion of ALKBH10B in the alkbh10b mutant delays flowering and represses vegetative growth. Complementation with wild-type ALKBH10B, but not a catalytically inactive mutant (ALKBH10B H366A/E368A), rescues these effects in alkbh10b-1 mutant plants, suggesting the observed phenotypes are controlled by the catalytic action of ALKBH10B We show that ALKBH10B-mediated mRNA demethylation affects the stability of target transcripts, thereby influencing floral transition. We identified 1190 m6A hypermethylated transcripts in the alkbh10b-1 mutant involved in plant development. The discovery and characterization of the archetypical RNA demethylase in Arabidopsis sheds light on the occurrence and functional role(s) of reversible mRNA methylation in plants and defines the role of m6A RNA modification in Arabidopsis floral transition.


Subject(s)
Adenosine/analogs & derivatives , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Arabidopsis/physiology , Flowers/enzymology , Flowers/physiology , Oxidoreductases, N-Demethylating/metabolism , RNA, Plant/metabolism , RNA-Binding Proteins/metabolism , Adenosine/chemistry , Adenosine/metabolism , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Demethylation , Flowers/genetics , Gene Expression Regulation, Plant , Genes, Plant , Methylation , Mutation/genetics , Oxidoreductases, N-Demethylating/genetics , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Substrate Specificity , Up-Regulation/genetics
20.
Nature ; 505(7481): 117-20, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24284625

ABSTRACT

N(6)-methyladenosine (m(6)A) is the most prevalent internal (non-cap) modification present in the messenger RNA of all higher eukaryotes. Although essential to cell viability and development, the exact role of m(6)A modification remains to be determined. The recent discovery of two m(6)A demethylases in mammalian cells highlighted the importance of m(6)A in basic biological functions and disease. Here we show that m(6)A is selectively recognized by the human YTH domain family 2 (YTHDF2) 'reader' protein to regulate mRNA degradation. We identified over 3,000 cellular RNA targets of YTHDF2, most of which are mRNAs, but which also include non-coding RNAs, with a conserved core motif of G(m(6)A)C. We further establish the role of YTHDF2 in RNA metabolism, showing that binding of YTHDF2 results in the localization of bound mRNA from the translatable pool to mRNA decay sites, such as processing bodies. The carboxy-terminal domain of YTHDF2 selectively binds to m(6)A-containing mRNA, whereas the amino-terminal domain is responsible for the localization of the YTHDF2-mRNA complex to cellular RNA decay sites. Our results indicate that the dynamic m(6)A modification is recognized by selectively binding proteins to affect the translation status and lifetime of mRNA.


Subject(s)
Adenosine/analogs & derivatives , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Adenosine/metabolism , Adenosine/pharmacology , Base Sequence , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Minor Histocompatibility Antigens , Nucleotide Motifs , Organelles/genetics , Organelles/metabolism , Protein Binding , Protein Biosynthesis , RNA Stability/drug effects , RNA Transport , RNA, Untranslated/chemistry , RNA, Untranslated/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/classification , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL