Chromatin regulation of transcriptional enhancers and cell fate by the Sotos syndrome gene NSD1.

[1]  Amit Kumar Sharma,et al.  Integrative genetic analysis identifies FLVCR1 as a plasma-membrane choline transporter in mammals. , 2023, Cell metabolism.

[2]  Weiqun Peng,et al.  MLL3/MLL4 methyltransferase activities control early embryonic development and embryonic stem cell differentiation in a lineage-selective manner , 2020, bioRxiv.

[3]  S. Ramachandran,et al.  The pausing zone and control of RNA polymerase II elongation by Spt5: Implications for the pause-release model. , 2022, Molecular cell.

[4]  Shondra M. Pruett-Miller,et al.  NSD1 mediates antagonism between SWI/SNF and polycomb complexes and is required for transcriptional activation upon EZH2 inhibition. , 2022, Molecular cell.

[5]  William A. Pastor,et al.  H3K36 dimethylation shapes the epigenetic interaction landscape by directing repressive chromatin modifications in embryonic stem cells , 2022, Genome research.

[6]  Thomas Vierbuchen,et al.  Rapid and robust directed differentiation of mouse epiblast stem cells into definitive endoderm and forebrain organoids , 2021, bioRxiv.

[7]  I. Wistuba,et al.  NSD2 dimethylation at H3K36 promotes lung adenocarcinoma pathogenesis. , 2021, Molecular cell.

[8]  F. X. Chen,et al.  SPT5 stabilizes RNA polymerase II, orchestrates transcription cycles, and maintains the enhancer landscape. , 2021, Molecular cell.

[9]  P. Cramer,et al.  Two distinct mechanisms of RNA polymerase II elongation stimulation in vivo. , 2021, Molecular cell.

[10]  J. Brickman,et al.  Enhancers are activated by p300/CBP activity-dependent PIC assembly, RNAPII recruitment, and pause release. , 2021, Molecular cell.

[11]  J. Minna,et al.  Elevated NSD3 Histone Methylation Activity Drives Squamous Cell Lung Cancer , 2021, Nature.

[12]  F. Buchholz,et al.  The Paf1 complex positively regulates enhancer activity in mouse embryonic stem cells , 2020, Life Science Alliance.

[13]  Thomas M. Keane,et al.  Twelve years of SAMtools and BCFtools , 2020, GigaScience.

[14]  M. Lorincz,et al.  NSD1-deposited H3K36me2 directs de novo methylation in the mouse male germline and counteracts Polycomb-associated silencing , 2020, Nature Genetics.

[15]  R. Klose,et al.  Live-cell single particle tracking of PRC1 reveals a highly dynamic system with low target site occupancy , 2020, Nature Communications.

[16]  Ingmar B. Schäfer,et al.  Structural basis for PRC2 decoding of active histone methylation marks H3K36me2/3 , 2020, bioRxiv.

[17]  Rushad Pavri,et al.  Spt5-mediated enhancer transcription directly couples enhancer activation with physical promoter interaction , 2020, Nature Genetics.

[18]  Michael J MacCoss,et al.  Skyline for Small Molecules: A Unifying Software Package for Quantitative Metabolomics. , 2020, Journal of proteome research.

[19]  J. Svejstrup,et al.  Using TTchem-seq for profiling nascent transcription and measuring transcript elongation , 2020, Nature Protocols.

[20]  Matthew J. Meiners,et al.  H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape , 2019, Nature.

[21]  Erez Lieberman Aiden,et al.  A Pliable Mediator Acts as a Functional Rather Than an Architectural Bridge between Promoters and Enhancers , 2019, Cell.

[22]  Anshul Kundaje,et al.  The ENCODE Blacklist: Identification of Problematic Regions of the Genome , 2019, Scientific Reports.

[23]  Steven Henikoff,et al.  Improved CUT&RUN chromatin profiling tools , 2019, eLife.

[24]  Y. Dou,et al.  Selective binding of the PHD6 finger of MLL4 to histone H4K16ac links MLL4 and MOF , 2019, Nature Communications.

[25]  Leighton J. Core,et al.  Promoter-proximal pausing of RNA polymerase II: a nexus of gene regulation , 2019, Genes & development.

[26]  Arndt von Haeseler,et al.  Quantification of experimentally induced nucleotide conversions in high-throughput sequencing datasets , 2019, BMC Bioinformatics.

[27]  Michael P. Meers,et al.  Old cogs, new tricks: the evolution of gene expression in a chromatin context , 2019, Nature Reviews Genetics.

[28]  P. Cramer,et al.  MYC Recruits SPT5 to RNA Polymerase II to Promote Processive Transcription Elongation , 2019, Molecular cell.

[29]  Katharine L. Diehl,et al.  Histone H3 tail binds a unique sensing pocket in EZH2 to activate the PRC2 methyltransferase , 2019, Proceedings of the National Academy of Sciences.

[30]  K. Helin,et al.  Molecular Mechanisms Directing PRC2 Recruitment and H3K27 Methylation. , 2019, Molecular cell.

[31]  P. Cramer,et al.  Structure of activated transcription complex Pol II–DSIF–PAF–SPT6 , 2018, Nature.

[32]  A. Shilatifard,et al.  Enhancer Logic and Mechanics in Development and Disease. , 2018, Trends in cell biology.

[33]  J. Brickman,et al.  Time-Resolved Analysis Reveals Rapid Dynamics and Broad Scope of the CBP/p300 Acetylome , 2018, Cell.

[34]  T. Cech,et al.  Live-cell imaging reveals the dynamics of PRC2 and recruitment to chromatin by SUZ12-associated subunits , 2018, Genes & development.

[35]  S. Carr,et al.  Discovery of proteins associated with a predefined genomic locus in living cells via dCAS9-APEX-mediated proximity labeling , 2018, Nature Methods.

[36]  Darren J. Fitzpatrick,et al.  The H3K36me2 Methyltransferase Nsd1 Demarcates PRC2-Mediated H3K27me2 and H3K27me3 Domains in Embryonic Stem Cells. , 2018, Molecular cell.

[37]  James E. Bradner,et al.  The dTAG system for immediate and target-specific protein degradation , 2018, Nature Chemical Biology.

[38]  Christopher A. Lavender,et al.  Widespread transcriptional pausing and elongation control at enhancers , 2018, Genes & development.

[39]  S. Dell’Orso,et al.  The Elongation Factor Spt6 Maintains ESC Pluripotency by Controlling Super-Enhancers and Counteracting Polycomb Proteins. , 2017, Molecular cell.

[40]  Emily C. Dykhuizen,et al.  Sequential Salt Extractions for the Analysis of Bulk Chromatin Binding Properties of Chromatin Modifying Complexes. , 2017, Journal of visualized experiments : JoVE.

[41]  A. Shilatifard,et al.  Histone H3K4 monomethylation catalyzed by Trr and mammalian COMPASS-like proteins at enhancers is dispensable for development and viability , 2017, Nature Genetics.

[42]  Johannes Zuber,et al.  Thiol-linked alkylation of RNA to assess expression dynamics , 2017, Nature Methods.

[43]  Michael Q. Zhang,et al.  PAF1 regulation of promoter-proximal pause release via enhancer activation , 2017, Science.

[44]  Nicholas A. Sinnott-Armstrong,et al.  An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues , 2017, Nature Methods.

[45]  S. Armstrong,et al.  A UTX-MLL4-p300 Transcriptional Regulatory Network Coordinately Shapes Active Enhancer Landscapes for Eliciting Transcription. , 2017, Molecular cell.

[46]  B. Garcia,et al.  Mll3 and Mll4 Facilitate Enhancer RNA Synthesis and Transcription from Promoters Independently of H3K4 Monomethylation. , 2017, Molecular cell.

[47]  P. Park,et al.  Spt5 Plays Vital Roles in the Control of Sense and Antisense Transcription Elongation. , 2017, Molecular cell.

[48]  L. S. Churchman,et al.  The code and beyond: transcription regulation by the RNA polymerase II carboxy-terminal domain , 2017, Nature Reviews Molecular Cell Biology.

[49]  Giulia Basile,et al.  Intragenic DNA methylation prevents spurious transcription initiation , 2017, Nature.

[50]  Dylan M. Marchione,et al.  Impaired H3K36 methylation defines a subset of head and neck squamous cell carcinomas , 2017, Nature Genetics.

[51]  Steven Henikoff,et al.  An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites , 2016, bioRxiv.

[52]  C. Allis,et al.  Selective recognition of histone crotonylation by double PHD fingers of MOZ and DPF2. , 2016, Nature chemical biology.

[53]  J. Wysocka,et al.  Ever-Changing Landscapes: Transcriptional Enhancers in Development and Evolution , 2016, Cell.

[54]  Jonathan M. Cairns,et al.  Lineage-Specific Genome Architecture Links Enhancers and Non-coding Disease Variants to Target Gene Promoters , 2016, Cell.

[55]  Weiqun Peng,et al.  Enhancer priming by H3K4 methyltransferase MLL4 controls cell fate transition , 2016, Proceedings of the National Academy of Sciences.

[56]  C. Allis,et al.  The molecular hallmarks of epigenetic control , 2016, Nature Reviews Genetics.

[57]  S. Thibodeau,et al.  The histone H3.3K36M mutation reprograms the epigenome of chondroblastomas , 2016, Science.

[58]  B. Garcia,et al.  Histone H3K36 mutations promote sarcomagenesis through altered histone methylation landscape , 2016, Science.

[59]  Fidel Ramírez,et al.  deepTools2: a next generation web server for deep-sequencing data analysis , 2016, Nucleic Acids Res..

[60]  G. Tonon,et al.  Structural basis for PHDVC5HCHNSD1–C2HRNizp1 interaction: implications for Sotos syndrome , 2016, Nucleic acids research.

[61]  Andrei L. Turinsky,et al.  NSD1 mutations generate a genome-wide DNA methylation signature , 2015, Nature Communications.

[62]  R. Roeder,et al.  RNA polymerase II–associated factor 1 regulates the release and phosphorylation of paused RNA polymerase II , 2015, Science.

[63]  David A. Scott,et al.  Rationally engineered Cas9 nucleases with improved specificity , 2015, Science.

[64]  Jeroen A. A. Demmers,et al.  Proteins that bind regulatory regions identified by histone modification chromatin immunoprecipitations and mass spectrometry , 2015, Nature Communications.

[65]  Christopher M. Vockley,et al.  Epigenome editing by a CRISPR/Cas9-based acetyltransferase activates genes from promoters and enhancers , 2015, Nature Biotechnology.

[66]  R. Young,et al.  Chromatin proteomic profiling reveals novel proteins associated with histone-marked genomic regions , 2015, Proceedings of the National Academy of Sciences.

[67]  C. Glass,et al.  The selection and function of cell type-specific enhancers , 2015, Nature Reviews Molecular Cell Biology.

[68]  A. Srivastava,et al.  Clinical and Molecular Heterogeneity in Brazilian Patients with Sotos Syndrome , 2015, Molecular Syndromology.

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

[70]  J. Min,et al.  Structure and function of the nucleosome-binding PWWP domain. , 2014, Trends in biochemical sciences.

[71]  B. van Steensel,et al.  Easy quantitative assessment of genome editing by sequence trace decomposition , 2014, Nucleic acids research.

[72]  Eugenia G. Giannopoulou,et al.  Histone Methyltransferase MMSET/NSD2 Alters EZH2 Binding and Reprograms the Myeloma Epigenome through Global and Focal Changes in H3K36 and H3K27 Methylation , 2014, PLoS genetics.

[73]  K. Helin,et al.  Gene silencing triggers polycomb repressive complex 2 recruitment to CpG islands genome wide. , 2014, Molecular cell.

[74]  A. Simeone,et al.  Reorganization of enhancer patterns in transition from naive to primed pluripotency. , 2014, Cell stem cell.

[75]  A. Stark,et al.  Transcriptional enhancers: from properties to genome-wide predictions , 2014, Nature Reviews Genetics.

[76]  M. Rosenfeld,et al.  Brd4 and JMJD6-Associated Anti-Pause Enhancers in Regulation of Transcriptional Pause Release , 2013, Cell.

[77]  Adam D. Wier,et al.  Structural basis for Spt5-mediated recruitment of the Paf1 complex to chromatin , 2013, Proceedings of the National Academy of Sciences.

[78]  Howard Y. Chang,et al.  Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position , 2013, Nature Methods.

[79]  A. Shilatifard,et al.  The MLL3/MLL4 Branches of the COMPASS Family Function as Major Histone H3K4 Monomethylases at Enhancers , 2013, Molecular and Cellular Biology.

[80]  Edward Y. Chen,et al.  Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool , 2013, BMC Bioinformatics.

[81]  David A. Orlando,et al.  Master Transcription Factors and Mediator Establish Super-Enhancers at Key Cell Identity Genes , 2013, Cell.

[82]  David A. Orlando,et al.  Selective Inhibition of Tumor Oncogenes by Disruption of Super-Enhancers , 2013, Cell.

[83]  Jacques Côté,et al.  Perceiving the epigenetic landscape through histone readers , 2012, Nature Structural &Molecular Biology.

[84]  Kevin W Eliceiri,et al.  NIH Image to ImageJ: 25 years of image analysis , 2012, Nature Methods.

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

[86]  E. Wagner,et al.  Understanding the language of Lys36 methylation at histone H3 , 2012, Nature Reviews Molecular Cell Biology.

[87]  B. Garcia,et al.  NSD2 links dimethylation of histone H3 at lysine 36 to oncogenic programming. , 2011, Molecular cell.

[88]  Ming-Ming Zhou,et al.  The PHD finger: a versatile epigenome reader. , 2011, Trends in biochemical sciences.

[89]  A. Bauer,et al.  Histone methylation by PRC2 is inhibited by active chromatin marks. , 2011, Molecular cell.

[90]  Meera Shah,et al.  NSD1 PHD domains bind methylated H3K4 and H3K9 using interactions disrupted by point mutations in human sotos syndrome , 2011, Human mutation.

[91]  Ryan A. Flynn,et al.  A unique chromatin signature uncovers early developmental enhancers in humans , 2011, Nature.

[92]  Li-Rong Yu,et al.  Distinct roles of GCN5/PCAF‐mediated H3K9ac and CBP/p300‐mediated H3K18/27ac in nuclear receptor transactivation , 2011, The EMBO journal.

[93]  B. Zhu,et al.  H3K36 Methylation Antagonizes PRC2-mediated H3K27 Methylation* , 2011, The Journal of Biological Chemistry.

[94]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer , 2011, Nature Biotechnology.

[95]  D. Reinberg,et al.  The Structure of NSD1 Reveals an Autoregulatory Mechanism Underlying Histone H3K36 Methylation* , 2010, The Journal of Biological Chemistry.

[96]  R. Young,et al.  Histone H3K27ac separates active from poised enhancers and predicts developmental state , 2010, Proceedings of the National Academy of Sciences.

[97]  J. Rice,et al.  Role for the nuclear receptor-binding SET domain protein 1 (NSD1) methyltransferase in coordinating lysine 36 methylation at histone 3 with RNA polymerase II function , 2010, Proceedings of the National Academy of Sciences.

[98]  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.

[99]  D. Reinberg,et al.  The Target of the NSD Family of Histone Lysine Methyltransferases Depends on the Nature of the Substrate , 2009, The Journal of Biological Chemistry.

[100]  W. Alkema,et al.  BioVenn – a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams , 2008, BMC Genomics.

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

[102]  A. Afenjar,et al.  Heterogeneity of NSD1 alterations in 116 patients with Sotos syndrome , 2007, Human mutation.

[103]  G. Wang,et al.  NUP98–NSD1 links H3K36 methylation to Hox-A gene activation and leukaemogenesis , 2007, Nature Cell Biology.

[104]  Nathaniel D. Heintzman,et al.  Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome , 2007, Nature Genetics.

[105]  N. Rahman,et al.  Genotype-phenotype associations in Sotos syndrome: an analysis of 266 individuals with NSD1 aberrations. , 2005, American journal of human genetics.

[106]  D. Horn,et al.  Mutations in NSD1 are responsible for Sotos syndrome, but are not a frequent finding in other overgrowth phenotypes , 2003, European Journal of Human Genetics.

[107]  Pierre Chambon,et al.  NSD1 is essential for early post‐implantation development and has a catalytically active SET domain , 2003, The EMBO journal.

[108]  S. Yeh,et al.  Identification and Characterization of a Novel Androgen Receptor Coregulator ARA267-α in Prostate Cancer Cells* , 2001, The Journal of Biological Chemistry.

[109]  J. Vonesch,et al.  Two distinct nuclear receptor interaction domains in NSD1, a novel SET protein that exhibits characteristics of both corepressors and coactivators , 1998, The EMBO journal.

[110]  R. Roeder,et al.  Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. , 1983, Nucleic acids research.

[111]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[112]  Ira M. Hall,et al.  BEDTools: a flexible suite of utilities for comparing genomic features , 2010, Bioinform..

[113]  J. Townsend,et al.  NIH Public Access Author Manuscript , 2006 .

[114]  N. Rahman,et al.  NSD1 mutations are the major cause of Sotos syndrome and occur in some cases of Weaver syndrome but are rare in other overgrowth phenotypes. , 2003, American journal of human genetics.