The SMAD2/3 interactome reveals that TGFβ controls m6A mRNA methylation in pluripotency

The TGFβ pathway has essential roles in embryonic development, organ homeostasis, tissue repair and disease. These diverse effects are mediated through the intracellular effectors SMAD2 and SMAD3 (hereafter SMAD2/3), whose canonical function is to control the activity of target genes by interacting with transcriptional regulators. Therefore, a complete description of the factors that interact with SMAD2/3 in a given cell type would have broad implications for many areas of cell biology. Here we describe the interactome of SMAD2/3 in human pluripotent stem cells. This analysis reveals that SMAD2/3 is involved in multiple molecular processes in addition to its role in transcription. In particular, we identify a functional interaction with the METTL3–METTL14–WTAP complex, which mediates the conversion of adenosine to N6-methyladenosine (m6A) on RNA. We show that SMAD2/3 promotes binding of the m6A methyltransferase complex to a subset of transcripts involved in early cell fate decisions. This mechanism destabilizes specific SMAD2/3 transcriptional targets, including the pluripotency factor gene NANOG, priming them for rapid downregulation upon differentiation to enable timely exit from pluripotency. Collectively, these findings reveal the mechanism by which extracellular signalling can induce rapid cellular responses through regulation of the epitranscriptome. These aspects of TGFβ signalling could have far-reaching implications in many other cell types and in diseases such as cancer.

[1]  Gordon K Smyth,et al.  Statistical Applications in Genetics and Molecular Biology Linear Models and Empirical Bayes Methods for Assessing Differential Expression in Microarray Experiments , 2011 .

[2]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[3]  Thomas D. Schmittgen,et al.  Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. , 2001, Methods.

[4]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[5]  Johnathon R. Walls,et al.  Foxh1 is essential for development of the anterior heart field. , 2004, Developmental cell.

[6]  R. Derynck,et al.  Smad2 Is Essential for Maintenance of the Human and Mouse Primed Pluripotent Stem Cell State* , 2013, The Journal of Biological Chemistry.

[7]  Uwe Ohler,et al.  RCAS: an RNA centric annotation system for transcriptome-wide regions of interest , 2017, Nucleic acids research.

[8]  Antonella Galli,et al.  Activin/Nodal signaling and NANOG orchestrate human embryonic stem cell fate decisions by controlling the H3K4me3 chromatin mark , 2015, Genes & development.

[9]  M. Greaves,et al.  The TEL-AML1 leukemia fusion gene dysregulates the TGF-beta pathway in early B lineage progenitor cells. , 2009, The Journal of clinical investigation.

[10]  David Newsome,et al.  Gene Dosage–Dependent Embryonic Development and Proliferation Defects in Mice Lacking the Transcriptional Integrator p300 , 1998, Cell.

[11]  Stepanka Vanacova,et al.  N6-methyladenosine demethylase FTO targets pre-mRNAs and regulates alternative splicing and 3′-end processing , 2017, Nucleic acids research.

[12]  Caleb Webber,et al.  GAT: a simulation framework for testing the association of genomic intervals , 2013, Bioinform..

[13]  Gordon Keller,et al.  Development of definitive endoderm from embryonic stem cells in culture , 2004, Development.

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

[15]  R. Pedersen,et al.  Nodal inhibits differentiation of human embryonic stem cells along the neuroectodermal default pathway. , 2004, Developmental biology.

[16]  L. Vallier Serum-free and feeder-free culture conditions for human embryonic stem cells. , 2011, Methods in molecular biology.

[17]  M. Trotter,et al.  Activin/Nodal signalling maintains pluripotency by controlling Nanog expression , 2009, Development.

[18]  Pedro Madrigal fCCAC: functional canonical correlation analysis to evaluate covariance between nucleic acid sequencing datasets , 2017, Bioinform..

[19]  T. Suda,et al.  Mice homozygous for a truncated form of CREB-binding protein exhibit defects in hematopoiesis and vasculo-angiogenesis. , 1999, Blood.

[20]  Daniel J. Gaffney,et al.  A survey of best practices for RNA-seq data analysis , 2016, Genome Biology.

[21]  R. Pedersen,et al.  Inducible and Deterministic Forward Programming of Human Pluripotent Stem Cells into Neurons, Skeletal Myocytes, and Oligodendrocytes , 2017, Stem cell reports.

[22]  S. Morrison,et al.  A comparison of apparent mRNA half-life using kinetic labeling techniques vs decay following administration of transcriptional inhibitors. , 1991, Analytical biochemistry.

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

[24]  V. Harley,et al.  SOX13 is up-regulated in the developing mouse neuroepithelium and identifies a sub-population of differentiating neurons. , 2005, Brain research. Developmental brain research.

[25]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[26]  J. Aramburu,et al.  NFAT5 induction by the pre–T-cell receptor serves as a selective survival signal in T-lymphocyte development , 2013, Proceedings of the National Academy of Sciences.

[27]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[28]  Jonas Korlach,et al.  The birth of the Epitranscriptome: deciphering the function of RNA modifications , 2012, Genome Biology.

[29]  C. Hill,et al.  Tgf-beta superfamily signaling in embryonic development and homeostasis. , 2009, Developmental cell.

[30]  Chengqi Yi,et al.  Epitranscriptome sequencing technologies: decoding RNA modifications , 2016, Nature Methods.

[31]  R. Pedersen,et al.  Activin/Nodal and FGF pathways cooperate to maintain pluripotency of human embryonic stem cells , 2005, Journal of Cell Science.

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

[33]  Y. Saijoh,et al.  The transcription factor FoxH1 (FAST) mediates Nodal signaling during anterior-posterior patterning and node formation in the mouse. , 2001, Genes & development.

[34]  M. Mann,et al.  Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips , 2007, Nature Protocols.

[35]  E. Kroon,et al.  Efficient differentiation of human embryonic stem cells to definitive endoderm , 2005, Nature Biotechnology.

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

[37]  N. Shikama,et al.  Essential function of p300 acetyltransferase activity in heart, lung and small intestine formation , 2003, The EMBO journal.

[38]  David W. Russell,et al.  Identification of associated proteins by coimmunoprecipitation. , 2006, CSH protocols.

[39]  R. Pedersen,et al.  Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development , 2010, Hepatology.

[40]  James D. Cooper,et al.  Optimized inducible shRNA and CRISPR/Cas9 platforms for in vitro studies of human development using hPSCs , 2016, Development.

[41]  B. Lim,et al.  Activin/Nodal Signaling Controls Divergent Transcriptional Networks in Human Embryonic Stem Cells and in Endoderm Progenitors , 2011, Stem cells.

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

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

[44]  Timothy L. Bailey,et al.  Gene expression Advance Access publication May 4, 2011 DREME: motif discovery in transcription factor ChIP-seq data , 2011 .

[45]  C. Hill,et al.  TGF-β signaling to chromatin: how Smads regulate transcription during self-renewal and differentiation. , 2014, Seminars in cell & developmental biology.

[46]  Xiaodong Cui,et al.  MeTDiff: A Novel Differential RNA Methylation Analysis for MeRIP-Seq Data , 2018, IEEE/ACM Transactions on Computational Biology and Bioinformatics.

[47]  M. Mann,et al.  MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification , 2008, Nature Biotechnology.

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

[49]  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.

[50]  Avi Ma'ayan,et al.  Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool , 2013, BMC Bioinformatics.

[51]  Junwei Shi,et al.  Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control , 2017, Nature.

[52]  Ludovic Vallier,et al.  Signaling Pathways Controlling Pluripotency and Early Cell Fate Decisions of Human Induced Pluripotent Stem Cells , 2009, Stem cells.

[53]  Joonsoo Kang,et al.  Regulation of gammadelta versus alphabeta T lymphocyte differentiation by the transcription factor SOX13. , 2007, Science.

[54]  Francine E. Garrett-Bakelman,et al.  The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal and leukemia cells , 2017, Nature Medicine.

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

[56]  Wei Li,et al.  RSeQC: quality control of RNA-seq experiments , 2012, Bioinform..

[57]  E. Fuchs,et al.  The harmonies played by TGF-β in stem cell biology. , 2012, Cell stem cell.

[58]  Ariel J. Levine,et al.  TGFβ/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells , 2005 .

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

[60]  H. Moses,et al.  The roles of TGFβ in the tumour microenvironment , 2013, Nature Reviews Cancer.

[61]  J. Rossant,et al.  FoxH1 (Fast) functions to specify the anterior primitive streak in the mouse. , 2001, Genes & development.

[62]  H. Stunnenberg,et al.  A quantitative proteomics tool to identify DNA-protein interactions in primary cells or blood. , 2015, Journal of proteome research.

[63]  Gideon Rechavi,et al.  Transcriptome-wide mapping of N6-methyladenosine by m6A-seq based on immunocapturing and massively parallel sequencing , 2013, Nature Protocols.

[64]  M. Mann,et al.  Extracting gene function from protein-protein interactions using Quantitative BAC InteraCtomics (QUBIC). , 2011, Methods.

[65]  Roger A. Pedersen,et al.  NANOG and CDX2 pattern distinct subtypes of human mesoderm during exit from pluripotency. , 2014, Cell stem cell.

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

[67]  A. Bradley,et al.  Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells , 2011, Nature.

[68]  W. Harris,et al.  Inhibition of Activin/Nodal signaling promotes specification of human embryonic stem cells into neuroectoderm. , 2008, Developmental biology.

[69]  Reinout Raijmakers,et al.  Multiplex peptide stable isotope dimethyl labeling for quantitative proteomics , 2009, Nature Protocols.

[70]  A. Hyman,et al.  Quantitative proteomics combined with BAC TransgeneOmics reveals in vivo protein interactions , 2010, The Journal of cell biology.

[71]  Cole Trapnell,et al.  TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions , 2013, Genome Biology.

[72]  Roger A. Pedersen,et al.  Early Cell Fate Decisions of Human Embryonic Stem Cells and Mouse Epiblast Stem Cells Are Controlled by the Same Signalling Pathways , 2009, PloS one.

[73]  Tim J. P. Hubbard,et al.  Dalliance: interactive genome viewing on the web , 2011, Bioinform..

[74]  D. Livingston,et al.  Gene dose-dependent control of hematopoiesis and hematologic tumor suppression by CBP. , 2000, Genes & development.

[75]  Douglas L Black,et al.  m6A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover , 2017, Genes & development.

[76]  M. Esteller,et al.  An Adenine Code for DNA: A Second Life for N6-Methyladenine , 2015, Cell.

[77]  Cole Trapnell,et al.  Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. , 2010, Nature biotechnology.

[78]  M. Trotter,et al.  Pluripotency factors regulate definitive endoderm specification through eomesodermin. , 2011, Genes & development.

[79]  Anne E Carpenter,et al.  A Lentiviral RNAi Library for Human and Mouse Genes Applied to an Arrayed Viral High-Content Screen , 2006, Cell.

[80]  Edward Chuong,et al.  Chromatin and transcriptional signatures for Nodal signaling during endoderm formation in hESCs. , 2011, Developmental biology.

[81]  J. Massagué,et al.  Smad transcription factors. , 2005, Genes & development.

[82]  Xiaodong Cui,et al.  Exome-based analysis for RNA epigenome sequencing data , 2013, Bioinform..

[83]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.