Rethinking m6A Readers, Writers, and Erasers.

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.

[1]  J. Valcárcel,et al.  Biochemical Function of Female-Lethal (2)D/Wilms' Tumor Suppressor-1-associated Proteins in Alternative Pre-mRNA Splicing* , 2003, The Journal of Biological Chemistry.

[2]  Ram Reddy,et al.  Accurate and efficient N-6-adenosine methylation in spliceosomal U6 small nuclear RNA by HeLa cell extract in vitro , 1995, Nucleic Acids Res..

[3]  Chao Xu,et al.  A novel RNA-binding mode of the YTH domain reveals the mechanism for recognition of determinant of selective removal by Mmi1 , 2015, Nucleic acids research.

[4]  Minoru Yoshida,et al.  RNA-Methylation-Dependent RNA Processing Controls the Speed of the Circadian Clock , 2013, Cell.

[5]  R J Roberts,et al.  Sequence specificity of the human mRNA N6-adenosine methylase in vitro. , 1990, Nucleic acids research.

[6]  A. Aguzzi,et al.  Phase Separation: Linking Cellular Compartmentalization to Disease. , 2016, Trends in cell biology.

[7]  Fei Wang,et al.  Transcriptome-wide distribution and function of RNA hydroxymethylcytosine , 2016, Science.

[8]  Zhike Lu,et al.  m6A RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells , 2017, Cell reports.

[9]  Ping Wang,et al.  Structural Basis for Cooperative Function of Mettl3 and Mettl14 Methyltransferases. , 2016, Molecular cell.

[10]  L. Sánchez,et al.  The gene fl(2)d is needed for the sex-specific splicing of transformer pre-mRNA but not for double-sex pre-mRNA in Drosophila melanogaster , 1996, Molecular and General Genetics MGG.

[11]  Samir Adhikari,et al.  Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing. , 2016, Molecular cell.

[12]  N. Cox,et al.  Obesity-associated variants within FTO form long-range functional connections with IRX3 , 2014, Nature.

[13]  Chuanzhao Zhang,et al.  Hypoxia-inducible factors regulate pluripotency factor expression by ZNF217- and ALKBH5-mediated modulation of RNA methylation in breast cancer cells , 2016, Oncotarget.

[14]  R. Perry,et al.  Existence of Methylated Messenger RNA in Mouse L Cells , 1974 .

[15]  Liang Tong,et al.  Molecular basis for the recognition of methylated adenines in RNA by the eukaryotic YTH domain , 2014, Proceedings of the National Academy of Sciences.

[16]  B. Moss,et al.  N6, O2′-dimethyladenosine a novel methylated ribonucleoside next to the 5′ terminal of animal cell and virus mRNAs , 1975, Nature.

[17]  Samie R. Jaffrey,et al.  Emerging links between m6A and misregulated mRNA methylation in cancer , 2017, Genome Medicine.

[18]  Manolis Kellis,et al.  FTO Obesity Variant Circuitry and Adipocyte Browning in Humans. , 2015, The New England journal of medicine.

[19]  Simon Hess,et al.  The fat mass and obesity associated gene (Fto) regulates activity of the dopaminergic midbrain circuitry , 2013, Nature Neuroscience.

[20]  Chuanzhao Zhang,et al.  Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA , 2016, Proceedings of the National Academy of Sciences.

[21]  E. Nogales,et al.  Two RNA-binding motifs in eIF3 direct HCV IRES-dependent translation , 2013, Nucleic acids research.

[22]  Shu-Bing Qian,et al.  Dynamic m6A mRNA methylation directs translational control of heat shock response , 2015, Nature.

[23]  S. Tavazoie,et al.  N6-methyladenosine marks primary microRNAs for processing , 2015, Nature.

[24]  O. Elemento,et al.  Comprehensive Analysis of mRNA Methylation Reveals Enrichment in 3′ UTRs and near Stop Codons , 2012, Cell.

[25]  Zhike Lu,et al.  m6A-dependent regulation of messenger RNA stability , 2013, Nature.

[26]  Anthony Barsic,et al.  ATPase-Modulated Stress Granules Contain a Diverse Proteome and Substructure , 2016, Cell.

[27]  Jens C. Brüning,et al.  Inactivation of the Fto gene protects from obesity , 2009, Nature.

[28]  Chuan He,et al.  N 6 -methyladenosine Modulates Messenger RNA Translation Efficiency , 2015, Cell.

[29]  N. Sonenberg,et al.  Regulation of Eukaryotic Initiation Factor 4E by Converging Signaling Pathways during Metabotropic Glutamate Receptor-Dependent Long-Term Depression , 2006, The Journal of Neuroscience.

[30]  Kaori Takai,et al.  RNA helicase YTHDC2 promotes cancer metastasis via the enhancement of the efficiency by which HIF-1α mRNA is translated. , 2016, Cancer letters.

[31]  Erez Y. Levanon,et al.  m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation , 2015, Science.

[32]  Chengqi Yi,et al.  Oxidative demethylation of 3‐methylthymine and 3‐methyluracil in single‐stranded DNA and RNA by mouse and human FTO , 2008, FEBS letters.

[33]  Yang Wang,et al.  N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells , 2014, Nature Cell Biology.

[34]  Bryan R. Cullen,et al.  Posttranscriptional m(6)A Editing of HIV-1 mRNAs Enhances Viral Gene Expression. , 2016, Cell host & microbe.

[35]  Jeannie T. Lee Lessons from X-chromosome inactivation: long ncRNA as guides and tethers to the epigenome. , 2009, Genes & development.

[36]  Jie Jin,et al.  FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N6-Methyladenosine RNA Demethylase. , 2017, Cancer cell.

[37]  Chuan He,et al.  N6-methyladenosine of HIV-1 RNA regulates viral infection and HIV-1 Gag protein expression , 2016, eLife.

[38]  Samie R. Jaffrey,et al.  The dynamic epitranscriptome: N6-methyladenosine and gene expression control , 2014, Nature Reviews Molecular Cell Biology.

[39]  Alexa B. R. McIntyre,et al.  N6-Methyladenosine in Flaviviridae Viral RNA Genomes Regulates Infection , 2016, Cell host & microbe.

[40]  Howard Y. Chang,et al.  Structural imprints in vivo decode RNA regulatory mechanisms , 2015, Nature.

[41]  Michel Herzog,et al.  MTA Is an Arabidopsis Messenger RNA Adenosine Methylase and Interacts with a Homolog of a Sex-Specific Splicing Factor[W][OA] , 2008, The Plant Cell Online.

[42]  Ligang Wu,et al.  YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4–NOT deadenylase complex , 2016, Nature Communications.

[43]  J. Darnell,et al.  The absolute frequency of labeled N-6-methyladenosine in HeLa cell messenger RNA decreases with label time. , 1978, Journal of molecular biology.

[44]  D. Storm,et al.  Hypomorphism of Fto and Rpgrip1l causes obesity in mice. , 2016, The Journal of clinical investigation.

[45]  J. Ule,et al.  iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution , 2010, Nature Structural &Molecular Biology.

[46]  Chuan He,et al.  N6-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions , 2015, Nature.

[47]  Qiang Wang,et al.  Structural basis of N6-adenosine methylation by the METTL3–METTL14 complex , 2016, Nature.

[48]  R. Schneiter,et al.  The gene virilizer is required for female-specific splicing controlled by Sxl, the master gene for sexual development in Drosophila. , 1995, Development.

[49]  R. Jackson,et al.  The mechanism of eukaryotic translation initiation and principles of its regulation , 2010, Nature Reviews Molecular Cell Biology.

[50]  P. Brown,et al.  Transcriptome-Wide Mapping of Pseudouridines: Pseudouridine Synthases Modify Specific mRNAs in S. cerevisiae , 2014, PloS one.

[51]  Olivier Elemento,et al.  Reversible methylation of m6Am in the 5′ cap controls mRNA stability , 2016, Nature.

[52]  J. Cate,et al.  eIF3 targets cell proliferation mRNAs for translational activation or repression , 2015, Nature.

[53]  Markus Blatter,et al.  Solution structure of the YTH domain in complex with N6-methyladenosine RNA: a reader of methylated RNA , 2014, Nucleic acids research.

[54]  Olivier Elemento,et al.  5′ UTR m6A Promotes Cap-Independent Translation , 2015, Cell.

[55]  Saeed Tavazoie,et al.  HNRNPA2B1 Is a Mediator of m6A-Dependent Nuclear RNA Processing Events , 2015, Cell.

[56]  Matthieu Schapira,et al.  Structural Basis for the Discriminative Recognition of N6-Methyladenosine RNA by the Human YT521-B Homology Domain Family of Proteins* , 2015, The Journal of Biological Chemistry.

[57]  Arne Klungland,et al.  A majority of m6A residues are in the last exons, allowing the potential for 3′ UTR regulation , 2015, Genes & development.

[58]  W. Gilbert,et al.  Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells , 2014, Nature.

[59]  Nian Liu,et al.  Probing N6-methyladenosine RNA modification status at single nucleotide resolution in mRNA and long noncoding RNA , 2013, RNA.

[60]  S. Stamm,et al.  YTH: a new domain in nuclear proteins. , 2002, Trends in biochemical sciences.

[61]  R. Kierzek,et al.  The thermodynamic stability of RNA duplexes and hairpins containing N6-alkyladenosines and 2-methylthio-N6-alkyladenosines. , 2003, Nucleic acids research.

[62]  Chuan He,et al.  Dynamics of Human and Viral RNA Methylation during Zika Virus Infection. , 2016, Cell host & microbe.

[63]  Samie R. Jaffrey,et al.  m6A RNA methylation promotes XIST-mediated transcriptional repression , 2016, Nature.

[64]  Yi Xing,et al.  m(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells. , 2014, Cell stem cell.

[65]  M. Kupiec,et al.  Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq , 2012, Nature.

[66]  N. Sonenberg,et al.  Regulation of cap-dependent translation by eIF4E inhibitory proteins , 2005, Nature.

[67]  B. Moss,et al.  5'-Terminal and internal methylated nucleotide sequences in HeLa cell mRNA. , 1976, Biochemistry.

[68]  Christopher B. Burge,et al.  Protein-RNA Networks Regulated by Normal and ALS-Associated Mutant HNRNPA2B1 in the Nervous System , 2016, Neuron.

[69]  Miguel A. Andrade-Navarro,et al.  m6A modulates neuronal functions and sex determination in Drosophila , 2016, Nature.

[70]  Chuan He,et al.  m6A Demethylase ALKBH5 Maintains Tumorigenicity of Glioblastoma Stem-like Cells by Sustaining FOXM1 Expression and Cell Proliferation Program. , 2017, Cancer cell.

[71]  Jimin Pei,et al.  Cell-free Formation of RNA Granules: Low Complexity Sequence Domains Form Dynamic Fibers within Hydrogels , 2012, Cell.

[72]  S. Horner,et al.  RNA modifications go viral , 2017, PLoS pathogens.

[73]  Schraga Schwartz,et al.  Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5' sites. , 2014, Cell reports.

[74]  Christopher E. Mason,et al.  Single-nucleotide resolution mapping of m6A and m6Am throughout the transcriptome , 2015, Nature Methods.

[75]  Gerald R. Fink,et al.  RNA Methylation by the MIS Complex Regulates a Cell Fate Decision in Yeast , 2012, PLoS genetics.

[76]  Chengqi Yi,et al.  N6-Methyladenosine in Nuclear RNA is a Major Substrate of the Obesity-Associated FTO , 2011, Nature chemical biology.

[77]  Samir Adhikari,et al.  Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase , 2014, Cell Research.

[78]  M. Jinek,et al.  Structural insights into the molecular mechanism of the m6A writer complex , 2016, eLife.

[79]  Maxwell R. Mumbach,et al.  Transcriptome-wide Mapping Reveals Widespread Dynamic-Regulated Pseudouridylation of ncRNA and mRNA , 2014, Cell.

[80]  F. Rottman,et al.  Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. , 1997, RNA.

[81]  B. Moss,et al.  Nucleotide sequences at the N6-methyladenosine sites of HeLa cell messenger ribonucleic acid. , 1977, Biochemistry.

[82]  Ke Liu,et al.  Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. , 2014, Nature chemical biology.

[83]  C. Niehrs,et al.  RNA fate determination through cotranscriptional adenosine methylation and microprocessor binding , 2017, Nature Structural &Molecular Biology.

[84]  Inês Barroso,et al.  The genetics of obesity: FTO leads the way , 2010, Trends in genetics : TIG.

[85]  F. Rottman,et al.  Characterization and partial purification of mRNA N6-adenosine methyltransferase from HeLa cell nuclei. Internal mRNA methylation requires a multisubunit complex. , 1994, The Journal of biological chemistry.

[86]  R. Desrosiers,et al.  Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. , 1974, Proceedings of the National Academy of Sciences of the United States of America.

[87]  Makoto Naito,et al.  Identification of Wilms' Tumor 1-associating Protein Complex and Its Role in Alternative Splicing and the Cell Cycle* , 2013, The Journal of Biological Chemistry.

[88]  F. Rottman,et al.  An in vitro system for accurate methylation of internal adenosine residues in messenger RNA. , 1988, Science.

[89]  Yang Xie,et al.  The U6 snRNA m6A Methyltransferase METTL16 Regulates SAM Synthetase Intron Retention , 2017, Cell.

[90]  Yingpu Yu,et al.  Factor requirements for translation initiation on the Simian picornavirus internal ribosomal entry site. , 2007, RNA.

[91]  Nathan Archer,et al.  m6A potentiates Sxl alternative pre-mRNA splicing for robust Drosophila sex determination , 2016, Nature.

[92]  M. Schapira Structural Chemistry of Human RNA Methyltransferases. , 2016, ACS chemical biology.

[93]  Arne Klungland,et al.  ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. , 2013, Molecular cell.

[94]  B. Moss,et al.  HeLa cell RNA (2'-O-methyladenosine-N6-)-methyltransferase specific for the capped 5'-end of messenger RNA. , 1978, The Journal of biological chemistry.

[95]  K. Morohashi,et al.  Cyclosporin A Associated Helicase-Like Protein Facilitates the Association of Hepatitis C Virus RNA Polymerase with Its Cellular Cyclophilin B , 2011, PloS one.

[96]  Miao Yu,et al.  A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation , 2013, Nature chemical biology.

[97]  Ran Elkon,et al.  Transcription Impacts the Efficiency of mRNA Translation via Co-transcriptional N6-adenosine Methylation , 2017, Cell.

[98]  Eric C. Lai,et al.  The m6A pathway facilitates sex determination in Drosophila , 2017, Nature Communications.

[99]  Howard Y. Chang,et al.  Structure and thermodynamics of N6-methyladenosine in RNA: a spring-loaded base modification. , 2015, Journal of the American Chemical Society.

[100]  Michael Hiller,et al.  The YTH Domain Is a Novel RNA Binding Domain* , 2010, The Journal of Biological Chemistry.