Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing.

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.

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

[2]  Zhiqin Xie,et al.  Transcriptome analysis of alternative splicing events regulated by SRSF10 reveals position-dependent splicing modulation , 2014, Nucleic acids research.

[3]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[4]  Junjie Chen,et al.  Functional Interaction between Chfr and Kif22 Controls Genomic Stability* , 2009, Journal of Biological Chemistry.

[5]  Jef Rozenski,et al.  The RNA modification database, RNAMDB: 2011 update , 2010, Nucleic Acids Res..

[6]  Marco Blanchette,et al.  SR proteins control a complex network of RNA-processing events , 2015, RNA.

[7]  Sylvain Foissac,et al.  A General Definition and Nomenclature for Alternative Splicing Events , 2008, PLoS Comput. Biol..

[8]  W. Hong,et al.  A role for sorting nexin 27 in AMPA receptor trafficking , 2014, Nature Communications.

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

[10]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[11]  A. Hartmann,et al.  The interaction and colocalization of Sam68 with the splicing-associated factor YT521-B in nuclear dots is regulated by the Src family kinase p59(fyn). , 1999, Molecular biology of the cell.

[12]  Schraga Schwartz,et al.  High-Resolution Mapping Reveals a Conserved, Widespread, Dynamic mRNA Methylation Program in Yeast Meiosis , 2013, Cell.

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

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

[15]  Uwe Ohler,et al.  PARalyzer: definition of RNA binding sites from PAR-CLIP short-read sequence data , 2011, Genome Biology.

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

[17]  J. Dahlberg,et al.  Molecular biology. , 1977, Science.

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

[19]  Lior Pachter,et al.  Sequence Analysis , 2020, Definitions.

[20]  T. Nilsen Internal mRNA Methylation Finally Finds Functions , 2014, Science.

[21]  R. North,et al.  Calcium dependent regulation of Rab activation and vesicle fusion by an intracellular P2X ion channel , 2013, Nature Cell Biology.

[22]  Zhen Shi,et al.  SRSF1 and SRSF9 RNA binding proteins promote Wnt signalling-mediated tumorigenesis by enhancing β-catenin biosynthesis , 2013, EMBO molecular medicine.

[23]  Peter J. Shepard,et al.  The SR protein family , 2009, Genome Biology.

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

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

[26]  M. Ares,et al.  Context-dependent control of alternative splicing by RNA-binding proteins , 2014, Nature Reviews Genetics.

[27]  Eric T. Wang,et al.  MBNL proteins repress ES-cell-specific alternative splicing and reprogramming , 2013, Nature.

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

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

[30]  A. Krainer,et al.  Regulation of alternative splicing in vivo by overexpression of antagonistic splicing factors. , 1994, Science.

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

[32]  E. Buratti,et al.  SR protein-mediated inhibition of CFTR exon 9 inclusion: molecular characterization of the intronic splicing silencer , 2007, Nucleic acids research.

[33]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

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

[35]  J. Bujnicki,et al.  MODOMICS: a database of RNA modification pathways—2013 update , 2012, Nucleic Acids Res..

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

[37]  Jernej Ule,et al.  The RNA-binding landscapes of two SR proteins reveal unique functions and binding to diverse RNA classes , 2012, Genome Biology.

[38]  T. Nilsen,et al.  Expansion of the eukaryotic proteome by alternative splicing , 2010, Nature.

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

[40]  Y. Li,et al.  Cytostellin distributes to nuclear regions enriched with splicing factors. , 1994, Journal of cell science.

[41]  M. Tohyama,et al.  Cloning of a gene, YT521, for a novel RNA splicing-related protein induced by hypoxia/reoxygenation. , 1998, Brain research. Molecular brain research.

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

[43]  Stefan Stamm,et al.  The ER Repeat Protein Yt521-B Localizes to a Novel Subnuclear Compartment , 2000, The Journal of cell biology.

[44]  Gene W. Yeo,et al.  Integrative genome‐wide analysis reveals cooperative regulation of alternative splicing by hnRNP proteins , 2012, Cell reports.

[45]  Bing Ren,et al.  N6-methyladenosine-dependent regulation of messenger RNA stability , 2013 .

[46]  Scott B. Dewell,et al.  Transcriptome-wide Identification of RNA-Binding Protein and MicroRNA Target Sites by PAR-CLIP , 2010, Cell.

[47]  Dmitri D. Pervouchine,et al.  The human transcriptome across tissues and individuals , 2015, Science.

[48]  S. Jaffrey,et al.  A mRNA methylation directs translational control of heat shock response , 2015 .

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

[50]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[51]  Gene W. Yeo,et al.  Genome-wide analysis reveals SR protein cooperation and competition in regulated splicing. , 2013, Molecular cell.

[52]  Björn Usadel,et al.  Trimmomatic: a flexible trimmer for Illumina sequence data , 2014, Bioinform..

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

[54]  B. Langmead,et al.  Aligning Short Sequencing Reads with Bowtie , 2010, Current protocols in bioinformatics.

[55]  Aaron R. Quinlan,et al.  BIOINFORMATICS APPLICATIONS NOTE , 2022 .

[56]  Clifford A. Meyer,et al.  Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.

[57]  Joonhee Han,et al.  SR Proteins Induce Alternative Exon Skipping through Their Activities on the Flanking Constitutive Exons , 2010, Molecular and Cellular Biology.

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

[59]  T. Maniatis,et al.  Serine/arginine-rich protein-dependent suppression of exon skipping by exonic splicing enhancers. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[60]  Neville E. Sanjana,et al.  Perturbation of m 6 AWriters Reveals Two Distinct Classes of mRNA Methylation at Internal and 5 0 Sites , 2014 .

[61]  S. Stamm,et al.  Rapid generation of splicing reporters with pSpliceExpress. , 2008, Gene.

[62]  Chuan He,et al.  FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis , 2014, Cell Research.

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

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

[65]  Qi Zhou,et al.  m(6)A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. , 2015, Cell stem cell.