TOBF1 modulates mouse embryonic stem cell fate through co-transcriptional regulation of alternative splicing

Embryonic stem (ES) cells retain the ability to undergo lineage-specific differentiation that can eventually give rise to different cell types that constitute an organism. Although stem cell specific biological networks of transcription factors and epigenetic modifiers are well established, how the ES cell specific transcriptional and alternative splicing (AS) machinery regulate their expression has not been sufficiently explored. In this study, we show that the lncRNA associated protein TOBF1 regulates the co-transcriptional alternative splicing of transcripts necessary for maintaining stem cell identity in mouse ES cells. Overlaying information derived from TOBF1 chromatin occupancy, the distribution of its pluripotency-associated OCT-SOX binding motifs, and transcripts undergoing differential expression and alternative splicing upon its disruption unmasked local nuclear territories where these distinct events converge, ultimately leading to the maintenance of mouse ES cell identity.

[1]  X. Roca,et al.  SRRM2 organizes splicing condensates to regulate alternative splicing , 2022, bioRxiv.

[2]  Satyaprakash Pandey,et al.  Alternative splicing modulation mediated by G-quadruplex structures in MALAT1 lncRNA , 2021, Nucleic acids research.

[3]  D. Tillo,et al.  CTCF is a barrier for 2C-like reprogramming , 2021, Nature Communications.

[4]  M. Sang,et al.  Long noncoding RNA DGCR5 involves in tumorigenesis of esophageal squamous cell carcinoma via SRSF1-mediated alternative splicing of Mcl-1 , 2021, Cell Death & Disease.

[5]  N. Barker,et al.  A genome-scale CRISPR screen reveals factors regulating Wnt-dependent renewal of mouse gastric epithelial cells , 2021, Proceedings of the National Academy of Sciences.

[6]  Yongfei Hu,et al.  hnRNPLL controls pluripotency exit of embryonic stem cells by modulating alternative splicing of Tbx3 and Bptf , 2020, The EMBO journal.

[7]  D. Meierhofer,et al.  SON and SRRM2 are essential for nuclear speckle formation , 2020, eLife.

[8]  M. Taipale,et al.  An efficient KRAB domain for CRISPRi applications in human cells , 2020, Nature methods.

[9]  Michael Weber,et al.  Genome-wide analysis in the mouse embryo reveals the importance of DNA methylation for transcription integrity , 2020, Nature Communications.

[10]  Jianlong Wang,et al.  Ribosome heterogeneity in stem cells and development , 2020, The Journal of cell biology.

[11]  F. Tang,et al.  Activin A and BMP4 Signaling Expands Potency of Mouse Embryonic Stem Cells in Serum-Free Media , 2020, Stem cell reports.

[12]  M. Gabut,et al.  Ribosome and Translational Control in Stem Cells , 2020, Cells.

[13]  Meghali Aich,et al.  Role of lncRNAs in stem cell maintenance and differentiation. , 2020, Current topics in developmental biology.

[14]  H. Schöler,et al.  hnRNP‐K Targets Open Chromatin in Mouse Embryonic Stem Cells in Concert with Multiple Regulators , 2019, Stem cells.

[15]  F. Fuller-Pace,et al.  DDX5 plays essential transcriptional and post-transcriptional roles in the maintenance and function of spermatogonia , 2019, Nature Communications.

[16]  Y. Mo,et al.  Emerging roles of lncRNAs in the post-transcriptional regulation in cancer , 2019, Genes & diseases.

[17]  M. Cosma,et al.  Wnt/β-catenin signaling pathway safeguards epigenetic stability and homeostasis of mouse embryonic stem cells , 2019, Scientific Reports.

[18]  M. Muratani,et al.  Klf5 suppresses ERK signaling in mouse pluripotent stem cells , 2018, PloS one.

[19]  R. Kierzek,et al.  Modified RNA triplexes: Thermodynamics, structure and biological potential , 2018, Scientific Reports.

[20]  J. Manley,et al.  TCF3 alternative splicing controlled by hnRNP H/F regulates E-cadherin expression and hESC pluripotency , 2018, Genes & development.

[21]  Lingyi Chen,et al.  COPS2 Antagonizes OCT4 to Accelerate the G2/M Transition of Mouse Embryonic Stem Cells , 2018, Stem cell reports.

[22]  Lei S. Qi,et al.  DNMT3A and TET1 cooperate to regulate promoter epigenetic landscapes in mouse embryonic stem cells , 2018, Genome Biology.

[23]  Yongjie Xu,et al.  The multifunctional RNA-binding protein hnRNPK is critical for the proliferation and differentiation of myoblasts , 2018, BMB reports.

[24]  Maximilian Haeussler,et al.  CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens , 2018, Nucleic Acids Res..

[25]  P. Navarro,et al.  Esrrb, an estrogen‐related receptor involved in early development, pluripotency, and reprogramming , 2018, FEBS letters.

[26]  Michael T. McManus,et al.  The Transcriptionally Permissive Chromatin State of Embryonic Stem Cells Is Acutely Tuned to Translational Output. , 2018, Cell stem cell.

[27]  Guangdun Peng,et al.  TGF-β signaling pathway in early mouse development and embryonic stem cells. , 2018, Acta biochimica et biophysica Sinica.

[28]  A. Stewart,et al.  lncRNA Panct1 Maintains Mouse Embryonic Stem Cell Identity by Regulating TOBF1 Recruitment to Oct-Sox Sequences in Early G1. , 2017, Cell reports.

[29]  Lorenzo Manti,et al.  The RNA processing factors THRAP3 and BCLAF1 promote the DNA damage response through selective mRNA splicing and nuclear export , 2017, Nucleic acids research.

[30]  G. Hu,et al.  Post-transcriptional regulation of the pluripotent state. , 2017, Current opinion in genetics & development.

[31]  R. Padgett,et al.  The hnRNP RALY regulates transcription and cell proliferation by modulating the expression of specific factors including the proliferation marker E2F1 , 2017, The Journal of Biological Chemistry.

[32]  E. Li,et al.  Natural antisense transcript TPM1-AS regulates the alternative splicing of tropomyosin I through an interaction with RNA-binding motif protein 4. , 2017, The international journal of biochemistry & cell biology.

[33]  M. Passos-Bueno,et al.  EIF4A3 deficient human iPSCs and mouse models demonstrate neural crest defects that underlie Richieri-Costa-Pereira syndrome , 2017, Human molecular genetics.

[34]  Robert J. Weatheritt,et al.  Multilayered Control of Alternative Splicing Regulatory Networks by Transcription Factors. , 2017, Molecular cell.

[35]  Lin Liu,et al.  Epigenetic Modifiers Facilitate Induction and Pluripotency of Porcine iPSCs , 2016, Stem cell reports.

[36]  Austin G Smith,et al.  NODAL Secures Pluripotency upon Embryonic Stem Cell Progression from the Ground State , 2016, bioRxiv.

[37]  E. Lander,et al.  Local regulation of gene expression by lncRNA promoters, transcription and splicing , 2016, Nature.

[38]  T. Mikkelsen,et al.  Dynamics of lineage commitment revealed by single-cell transcriptomics of differentiating embryonic stem cells , 2016, Nature Communications.

[39]  J. Ko,et al.  Novel Function of Sprouty4 as a Regulator of Stemness and Differentiation of Embryonic Stem Cells , 2016, Development & reproduction.

[40]  B. Cieply,et al.  Multiphasic and Dynamic Changes in Alternative Splicing during Induction of Pluripotency Are Coordinated by Numerous RNA-Binding Proteins. , 2016, Cell reports.

[41]  A. Fusco,et al.  Hmga2 is necessary for Otx2-dependent exit of embryonic stem cells from the pluripotent ground state , 2016, BMC Biology.

[42]  J Downward,et al.  Tumour-suppression function of KLF12 through regulation of anoikis , 2015, Oncogene.

[43]  Aleksander Tworak,et al.  Overview of the RNA G-quadruplex structures. , 2016, Acta biochimica Polonica.

[44]  D. Oxley,et al.  Fgf and Esrrb integrate epigenetic and transcriptional networks that regulate self-renewal of trophoblast stem cells , 2015, Nature Communications.

[45]  Jennifer Tsialikas,et al.  LIN28: roles and regulation in development and beyond , 2015, Development.

[46]  P. Neveu,et al.  MXS-Chaining: A Highly Efficient Cloning Platform for Imaging and Flow Cytometry Approaches in Mammalian Systems , 2015, PloS one.

[47]  Shun Liu,et al.  Discovery of Protein–lncRNA Interactions by Integrating Large-Scale CLIP-Seq and RNA-Seq Datasets , 2015, Front. Bioeng. Biotechnol..

[48]  Feng Zhang,et al.  Genome engineering using CRISPR-Cas9 system. , 2015, Methods in molecular biology.

[49]  Kate S. Carroll,et al.  Regulation of Alternative Splicing Through Coupling with Transcription and Chromatin Structure , 2015 .

[50]  Jesper V Olsen,et al.  Rapid and deep proteomes by faster sequencing on a benchtop quadrupole ultra-high-field Orbitrap mass spectrometer. , 2014, Journal of proteome research.

[51]  S. Morrison,et al.  Cellular Differences in Protein Synthesis Regulate Tissue Homeostasis , 2014, Cell.

[52]  Joana A. Vidigal,et al.  In vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 system , 2014, Nature.

[53]  Max A. Horlbeck,et al.  Genome-Scale CRISPR-Mediated Control of Gene Repression and Activation , 2014, Cell.

[54]  Robert Blelloch,et al.  Regulation of pluripotency by RNA binding proteins. , 2014, Cell stem cell.

[55]  Jianlong Wang,et al.  RNA-binding proteins in pluripotency, differentiation, and reprogramming , 2014, Frontiers in Biology.

[56]  Jignesh R. Parikh,et al.  Alternative splicing of MBD2 supports self-renewal in human pluripotent stem cells. , 2014, Cell stem cell.

[57]  O. Dovey,et al.  Histone deacetylase (HDAC) 1 and 2 are essential for accurate cell division and the pluripotency of embryonic stem cells , 2014, Proceedings of the National Academy of Sciences.

[58]  H. Stunnenberg,et al.  Otx2 and Oct4 Drive Early Enhancer Activation during Embryonic Stem Cell Transition from Naive Pluripotency , 2014, Cell reports.

[59]  Howard Y. Chang,et al.  Long noncoding RNAs in cell-fate programming and reprogramming. , 2014, Cell stem cell.

[60]  Gwangil Kim,et al.  Lefty1 and lefty2 control the balance between self-renewal and pluripotent differentiation of mouse embryonic stem cells. , 2014, Stem cells and development.

[61]  R. Maehr,et al.  Cas9 effector-mediated regulation of transcription and differentiation in human pluripotent stem cells , 2014, Development.

[62]  J. Tazi,et al.  MBNL1 and RBFOX2 cooperate to establish a splicing programme involved in pluripotent stem cell differentiation , 2013, Nature Communications.

[63]  Pierre-Étienne Jacques,et al.  SON connects the splicing-regulatory network with pluripotency in human embryonic stem cells , 2013, Nature Cell Biology.

[64]  A. Martinez Arias,et al.  Single cell lineage analysis of mouse embryonic stem cells at the exit from pluripotency , 2013, Biology Open.

[65]  M. Alló,et al.  Alternative splicing: a pivotal step between eukaryotic transcription and translation , 2013, Nature Reviews Molecular Cell Biology.

[66]  Zhaoyu Li,et al.  Foxa2 and H2A.Z Mediate Nucleosome Depletion during Embryonic Stem Cell Differentiation , 2012, Cell.

[67]  Joana A. Vidigal,et al.  Embryonic stem cell miRNAs and their roles in development and disease. , 2012, Seminars in cancer biology.

[68]  S. K. Balakrishnan,et al.  Functional and Molecular Characterization of the Role of CTCF in Human Embryonic Stem Cell Biology , 2012, PloS one.

[69]  Wenxiu Zhao,et al.  Embryonic Stem Cell Markers , 2012, Molecules.

[70]  Chunaram Choudhary,et al.  Proteomic investigations reveal a role for RNA processing factor THRAP3 in the DNA damage response. , 2012, Molecular cell.

[71]  T. Down,et al.  Genomic Targets of Brachyury (T) in Differentiating Mouse Embryonic Stem Cells , 2012, PloS one.

[72]  Herbert Schulz,et al.  Combined RNAi and localization for functionally dissecting long noncoding RNAs , 2012, Nature Methods.

[73]  C. Mummery,et al.  Proteomic analysis of stem cell differentiation and early development. , 2012, Cold Spring Harbor perspectives in biology.

[74]  Benjamin L. Kidder,et al.  HDAC1 regulates pluripotency and lineage specific transcriptional networks in embryonic and trophoblast stem cells , 2011, Nucleic acids research.

[75]  Nicholas T. Ingolia,et al.  Ribosome Profiling of Mouse Embryonic Stem Cells Reveals the Complexity and Dynamics of Mammalian Proteomes , 2011, Cell.

[76]  Richard A Young,et al.  Control of the Embryonic Stem Cell State , 2011, Cell.

[77]  T. Russo,et al.  Regulatory role of Klf5 in early mouse development and in embryonic stem cells. , 2011, Vitamins and hormones.

[78]  M. Asashima,et al.  Inhibitory Smad proteins promote the differentiation of mouse embryonic stem cells into ependymal-like ciliated cells. , 2010, Biochemical and biophysical research communications.

[79]  B. Blencowe,et al.  The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. , 2010, Molecular cell.

[80]  Luca Mazzarella,et al.  Jarid2 is a PRC2 component in embryonic stem cells required for multi-lineage differentiation and recruitment of PRC1 and RNA Polymerase II to developmental regulators , 2010, Nature Cell Biology.

[81]  S. Murray,et al.  myc maintains embryonic stem cell pluripotency and self-renewal. , 2010, Differentiation; research in biological diversity.

[82]  Alexander R. Pico,et al.  Alternative splicing regulates mouse embryonic stem cell pluripotency and differentiation , 2010, Proceedings of the National Academy of Sciences.

[83]  Kit T. Rodolfa,et al.  Sox17 promotes differentiation in mouse embryonic stem cells by directly regulating extraembryonic gene expression and indirectly antagonizing self-renewal. , 2010, Genes & development.

[84]  W. Tarn,et al.  TRAP150 activates pre-mRNA splicing and promotes nuclear mRNA degradation , 2010, Nucleic acids research.

[85]  Yang Yang,et al.  Kruppel-like Factor 4 (Klf4) Prevents Embryonic Stem (ES) Cell Differentiation by Regulating Nanog Gene Expression* , 2010, The Journal of Biological Chemistry.

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

[87]  Jungho Kim,et al.  Oct-4 controls cell-cycle progression of embryonic stem cells , 2009, The Biochemical journal.

[88]  Donny D. Licatalosi,et al.  RNA processing and its regulation: global insights into biological networks , 2010, Nature Reviews Genetics.

[89]  Arend Sidow,et al.  Jarid2/Jumonji Coordinates Control of PRC2 Enzymatic Activity and Target Gene Occupancy in Pluripotent Cells , 2009, Cell.

[90]  Stephen Dalton,et al.  The cell cycle and Myc intersect with mechanisms that regulate pluripotency and reprogramming. , 2009, Cell stem cell.

[91]  Gang Pei,et al.  Yamanaka factors critically regulate the developmental signaling network in mouse embryonic stem cells , 2008, Cell Research.

[92]  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.

[93]  Xiang-Dong Fu,et al.  The splicing factor SC35 has an active role in transcriptional elongation , 2008, Nature Structural &Molecular Biology.

[94]  Daniel J. Hoeppner,et al.  Global transcription in pluripotent embryonic stem cells. , 2008, Cell stem cell.

[95]  F. Watt,et al.  Role of the Notch ligand Delta1 in embryonic and adult mouse epidermis. , 2008, The Journal of investigative dermatology.

[96]  K. Sekiguchi,et al.  Regulation of Mesodermal Differentiation of Mouse Embryonic Stem Cells by Basement Membranes* , 2007, Journal of Biological Chemistry.

[97]  Alexei A. Sharov,et al.  Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells , 2007, Nature Cell Biology.

[98]  S. Yamanaka,et al.  Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors , 2006, Cell.

[99]  X. Chen,et al.  The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells , 2006, Nature Genetics.

[100]  T. Misteli,et al.  Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. , 2006, Developmental cell.

[101]  Megan F. Cole,et al.  Core Transcriptional Regulatory Circuitry in Human Embryonic Stem Cells , 2005, Cell.

[102]  K. Boheler,et al.  Embryonic stem cells: prospects for developmental biology and cell therapy. , 2005, Physiological reviews.

[103]  D. Srivastava,et al.  The Hand1 and Hand2 transcription factors regulate expansion of the embryonic cardiac ventricles in a gene dosage-dependent manner , 2004, Development.

[104]  A. Kornblihtt,et al.  Multiple links between transcription and splicing. , 2004, RNA.

[105]  Jing Wang,et al.  BMP4 supports self-renewal of embryonic stem cells by inhibiting mitogen-activated protein kinase pathways. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[106]  M. Richards,et al.  The Transcriptome Profile of Human Embryonic Stem Cells as Defined by SAGE , 2004, Stem cells.

[107]  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.

[108]  Andrius Kazlauskas,et al.  Growth-factor-dependent mitogenesis requires two distinct phases of signalling , 2001, Nature Cell Biology.

[109]  W. Talbot,et al.  Mouse Lefty2 and zebrafish antivin are feedback inhibitors of nodal signaling during vertebrate gastrulation. , 1999, Molecular cell.

[110]  Y. Saijoh,et al.  Left–right asymmetric expression of the TGFβ-family member lefty in mouse embryos , 1996, Nature.

[111]  T. Maniatis,et al.  Factor required for mammalian spliceosome assembly is localized to discrete regions in the nucleus , 1990, Nature.

[112]  A. Bradley,et al.  Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines , 1984, Nature.

[113]  G. Martin,et al.  Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. , 1981, Proceedings of the National Academy of Sciences of the United States of America.

[114]  M. Kaufman,et al.  Establishment in culture of pluripotential cells from mouse embryos , 1981, Nature.