Cancer-specific CTCF binding facilitates oncogenic transcriptional dysregulation
暂无分享,去创建一个
A. Ratan | M. Figueroa | C. Zang | Kyle P. Eagen | P. Ntziachristos | Alexandre Gaspar-Maia | B. Singer | K. Helmin | Stephanie L. Safgren | Zhenjia Wang | Benjamin D. Singer | Celestia Fang | Cuijuan Han | Emmalee R Adelman | Aakrosh Ratan | A. Gaspar-Maia | Alexandre Gaspar-Maia
[1] A. Ratan,et al. Cancer-specific CTCF binding facilitates oncogenic transcriptional dysregulation , 2020, Genome Biology.
[2] Qiang Wu,et al. Tandem CTCF sites function as insulators to balance spatial chromatin contacts and topological enhancer-promoter selection , 2020, Genome Biology.
[3] A. Tsirigos,et al. Three-dimensional chromatin landscapes in T cell acute lymphoblastic leukemia , 2020, Nature Genetics.
[4] Alexander P. Wu,et al. Deciphering essential cistromes using genome-wide CRISPR screens , 2019, Proceedings of the National Academy of Sciences.
[5] B. Singer,et al. A Practical Guide to the Measurement and Analysis of DNA Methylation. , 2019, American journal of respiratory cell and molecular biology.
[6] William A. Flavahan,et al. Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs , 2019, Nature.
[7] A. Tsirigos,et al. Dynamic 3D chromosomal landscapes in acute leukemia , 2019, bioRxiv.
[8] P. Fraser,et al. Long-range enhancer–promoter contacts in gene expression control , 2019, Nature Reviews Genetics.
[9] S. Mundlos,et al. Functional dissection of the Sox9–Kcnj2 locus identifies nonessential and instructive roles of TAD architecture , 2019, Nature Genetics.
[10] Bianca J. Diaz,et al. Identification of Cancer Drivers at CTCF Insulators in 1,962 Whole Genomes. , 2019, Cell systems.
[11] Lovelace J. Luquette,et al. Linked-read analysis identifies mutations in single-cell DNA-sequencing data , 2019, Nature Genetics.
[12] Shawn C. Little,et al. Oncogenic Notch promotes long-range regulatory interactions within hyperconnected 3D cliques , 2019, bioRxiv.
[13] N. Hannett,et al. Enhancer features that drive formation of transcriptional condensates , 2018, bioRxiv.
[14] N. Hannett,et al. Transcription Factors Activate Genes through the Phase-Separation Capacity of Their Activation Domains , 2018, Cell.
[15] A. Chambery,et al. Interactome mapping defines BRG1, a component of the SWI/SNF chromatin remodeling complex, as a new partner of the transcriptional regulator CTCF , 2018, The Journal of Biological Chemistry.
[16] Mauro A. A. Castro,et al. The chromatin accessibility landscape of primary human cancers , 2018, Science.
[17] V. Corces,et al. Organizational principles of 3D genome architecture , 2018, Nature Reviews Genetics.
[18] C. Lareau,et al. A non-canonical SWI/SNF complex is a synthetic lethal target in cancers driven by BAF complex perturbation , 2018, Nature Cell Biology.
[19] F. Alt,et al. CTCF-Binding Elements Mediate Accessibility of RAG Substrates During Chromatin Scanning , 2018, Cell.
[20] Hans Christian Wittich,et al. Recommending plant taxa for supporting on-site species identification , 2018, BMC Bioinformatics.
[21] P. Tan,et al. Mutation hotspots at CTCF binding sites coupled to chromosomal instability in gastrointestinal cancers , 2018, Nature Communications.
[22] Daniel S. Day,et al. Transcriptional Dysregulation of MYC Reveals Common Enhancer-Docking Mechanism , 2018, Cell reports.
[23] Nathan C. Sheffield,et al. BART: a transcription factor prediction tool with query gene sets or epigenomic profiles , 2018, bioRxiv.
[24] Aristotelis Tsirigos,et al. Stratification of TAD boundaries reveals preferential insulation of super-enhancers by strong boundaries , 2018, Nature Communications.
[25] David J. Arenillas,et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework , 2017, Nucleic Acids Res..
[26] J. Michael Cherry,et al. The Encyclopedia of DNA elements (ENCODE): data portal update , 2017, Nucleic Acids Res..
[27] Erez Lieberman Aiden,et al. Cohesin Loss Eliminates All Loop Domains , 2017, Cell.
[28] Nuno A. Fonseca,et al. Two independent modes of chromatin organization revealed by cohesin removal , 2017, Nature.
[29] Liping Yang,et al. CTCF prevents genomic instability by promoting homologous recombination-directed DNA double-strand break repair , 2017, Proceedings of the National Academy of Sciences.
[30] Xiaodong Cheng,et al. Structural Basis for the Versatile and Methylation-Dependent Binding of CTCF to DNA. , 2017, Molecular cell.
[31] L. Mirny,et al. Targeted Degradation of CTCF Decouples Local Insulation of Chromosome Domains from Genomic Compartmentalization , 2017, Cell.
[32] Geet Duggal,et al. Salmon: fast and bias-aware quantification of transcript expression using dual-phase inference , 2017, Nature Methods.
[33] R. Young,et al. A Phase Separation Model for Transcriptional Control , 2017, Cell.
[34] G. Stein,et al. The connection between BRG1, CTCF and topoisomerases at TAD boundaries , 2017, Nucleus.
[35] Anthony D. Schmitt,et al. A Compendium of Chromatin Contact Maps Reveals Spatially Active Regions in the Human Genome. , 2016, Cell reports.
[36] Aristotelis Tsirigos,et al. HiC-bench: comprehensive and reproducible Hi-C data analysis designed for parameter exploration and benchmarking , 2016, BMC Genomics.
[37] Tao Liu,et al. Cistrome Data Browser: a data portal for ChIP-Seq and chromatin accessibility data in human and mouse , 2016, Nucleic Acids Res..
[38] S. Mundlos,et al. Formation of new chromatin domains determines pathogenicity of genomic duplications , 2016, Nature.
[39] Tao Liu,et al. ChiLin: a comprehensive ChIP-seq and DNase-seq quality control and analysis pipeline , 2016, BMC Bioinformatics.
[40] E. Nora,et al. CTCF and Cohesin in Genome Folding and Transcriptional Gene Regulation. , 2016, Annual review of genomics and human genetics.
[41] Iannis Aifantis,et al. Emerging concepts of epigenetic dysregulation in hematological malignancies , 2016, Nature Immunology.
[42] Shenglin Mei,et al. Modeling cis-regulation with a compendium of genome-wide histone H3K27ac profiles , 2016, Genome research.
[43] Harvard T. H. Chan,et al. NF-E2, FLI1 and RUNX1 collaborate at areas of dynamic chromatin to activate transcription in mature mouse megakaryocytes , 2016, Scientific Reports.
[44] F. Cunningham,et al. The Ensembl Variant Effect Predictor , 2016, bioRxiv.
[45] P. Ntziachristos,et al. Histone demethylases in physiology and cancer: a tale of two enzymes, JMJD3 and UTX. , 2016, Current opinion in genetics & development.
[46] C. Tyler-Smith,et al. Ancient DNA and the rewriting of human history: be sparing with Occam’s razor , 2016, Genome Biology.
[47] M. Robinson,et al. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. , 2015, F1000Research.
[48] Shawn M. Gillespie,et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas , 2015, Nature.
[49] Jean-Philippe Vert,et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing , 2015, Genome Biology.
[50] Daniel S. Day,et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods , 2015, Science.
[51] Michael Q. Zhang,et al. CRISPR Inversion of CTCF Sites Alters Genome Topology and Enhancer/Promoter Function , 2015, Cell.
[52] O. Hofmann,et al. VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research , 2016, Nucleic acids research.
[53] Matthew T. Maurano,et al. Role of DNA Methylation in Modulating Transcription Factor Occupancy. , 2015, Cell reports.
[54] Niko Välimäki,et al. CTCF/cohesin-binding sites are frequently mutated in cancer , 2015, Nature Genetics.
[55] A. Visel,et al. Disruptions of Topological Chromatin Domains Cause Pathogenic Rewiring of Gene-Enhancer Interactions , 2015, Cell.
[56] Philip A. Ewels,et al. Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C , 2015, Nature Genetics.
[57] Pedro P. Rocha,et al. CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation , 2015, Science.
[58] Edwin Cuppen,et al. Sambamba: fast processing of NGS alignment formats , 2015, Bioinform..
[59] Jing Liang,et al. Chromatin architecture reorganization during stem cell differentiation , 2015, Nature.
[60] Neva C. Durand,et al. A 3D Map of the Human Genome at Kilobase Resolution Reveals Principles of Chromatin Looping , 2014, Cell.
[61] W. Huber,et al. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.
[62] R. Young,et al. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element , 2014, Science.
[63] Martin J. Aryee,et al. Coverage recommendations for methylation analysis by whole genome bisulfite sequencing , 2014, Nature Methods.
[64] Teresa Palomero,et al. A NOTCH1-driven MYC enhancer promotes T cell development, transformation and acute lymphoblastic leukemia , 2014, Nature Medicine.
[65] Aristotelis Tsirigos,et al. Genome-wide Mapping and Characterization of Notch-Regulated Long Noncoding RNAs in Acute Leukemia , 2014, Cell.
[66] R. Jaenisch,et al. Contrasting roles for histone 3 lysine 27 demethylases in acute lymphoblastic leukemia , 2014, Nature.
[67] B. Ren,et al. The 3D genome in transcriptional regulation and pluripotency. , 2014, Cell stem cell.
[68] I. Shmulevich,et al. CTCF haploinsufficiency destabilizes DNA methylation and predisposes to cancer. , 2014, Cell reports.
[69] Ira M. Hall,et al. SAMBLASTER: fast duplicate marking and structural variant read extraction , 2014, Bioinform..
[70] J. Sage,et al. From fly wings to targeted cancer therapies: a centennial for notch signaling. , 2014, Cancer cell.
[71] V. Corces,et al. CTCF: an architectural protein bridging genome topology and function , 2014, Nature Reviews Genetics.
[72] Jon C. Aster,et al. NOTCH1–RBPJ complexes drive target gene expression through dynamic interactions with superenhancers , 2013, Proceedings of the National Academy of Sciences.
[73] 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.
[74] A. Tanay,et al. Single cell Hi-C reveals cell-to-cell variability in chromosome structure , 2013, Nature.
[75] Jennifer E. Phillips-Cremins,et al. Architectural Protein Subclasses Shape 3D Organization of Genomes during Lineage Commitment , 2013, Cell.
[76] Doris Berger,et al. International Cancer Genome Consortium , 2013, Im Focus Onkologie.
[77] Victor V Lobanenkov,et al. A genome-wide map of CTCF multivalency redefines the CTCF code. , 2013, Cell reports.
[78] Stein Aerts,et al. Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia , 2012, Nature Genetics.
[79] Sean R. Davis,et al. NCBI GEO: archive for functional genomics data sets—update , 2012, Nucleic Acids Res..
[80] B. Sobhian,et al. NOTCH1 nuclear interactome reveals key regulators of its transcriptional activity and oncogenic function. , 2012, Molecular cell.
[81] Matthew T. Maurano,et al. Widespread plasticity in CTCF occupancy linked to DNA methylation , 2012, Genome research.
[82] Richard A Young,et al. Core transcriptional regulatory circuit controlled by the TAL1 complex in human T cell acute lymphoblastic leukemia. , 2012, Cancer cell.
[83] Jesse R. Dixon,et al. Topological Domains in Mammalian Genomes Identified by Analysis of Chromatin Interactions , 2012, Nature.
[84] Steven L Salzberg,et al. Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.
[85] A. Ferrando,et al. Genetic Inactivation of the PRC2 Complex in T-Cell Acute Lymphoblastic Leukemia , 2011, Nature Medicine.
[86] Clifford A. Meyer,et al. Cistrome: an integrative platform for transcriptional regulation studies , 2011, Genome Biology.
[87] Felix Krueger,et al. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications , 2011, Bioinform..
[88] Kenneth H. Buetow,et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia , 2011, Nature.
[89] William Stafford Noble,et al. FIMO: scanning for occurrences of a given motif , 2011, Bioinform..
[90] W. J. Kent,et al. BigWig and BigBed: enabling browsing of large distributed datasets. , 2010, Bioinformatics.
[91] 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.
[92] Gary D Bader,et al. International network of cancer genome projects , 2010, Nature.
[93] A. Ferrando,et al. Therapeutic targeting of NOTCH1 signaling in T-cell acute lymphoblastic leukemia. , 2009, Clinical lymphoma & myeloma.
[94] Gonçalo R. Abecasis,et al. The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..
[95] Clifford A. Meyer,et al. Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.
[96] A. Ferrando,et al. NOTCH1 extracellular juxtamembrane expansion mutations in T-ALL. , 2008, Blood.
[97] Richard A Young,et al. Chromatin immunoprecipitation and microarray-based analysis of protein location , 2006, Nature Protocols.
[98] A. Ferrando,et al. CUTLL1, a novel human T-cell lymphoma cell line with t(7;9) rearrangement, aberrant NOTCH1 activation and high sensitivity to γ-secretase inhibitors , 2006, Leukemia.
[99] A. Gnirke,et al. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis , 2005, Nucleic acids research.
[100] J. Watson,et al. Biochemical and biological characterization of lymphocyte regulatory molecules. V. Identification of an interleukin 2-producing human leukemia T cell line , 1980, The Journal of experimental medicine.
[101] J. Henzen. Publisher's note , 1979, Brain Research.
[102] David J. Arenillas,et al. update of the open-access database of transcription factor binding profiles and its web framework , 2017 .
[103] Lipika R. Pal,et al. Genetic basis of common human disease: insight into the role of nonsynonymous SNPs from genome-wide association studies , 2011, Genome Biology.
[104] Ira M. Hall,et al. BEDTools: a flexible suite of utilities for comparing genomic features , 2010, Bioinform..
[105] I. Amit,et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. , 2009, Science.
[106] Claude-Alain H. Roten,et al. Fast and accurate short read alignment with Burrows–Wheeler transform , 2009, Bioinform..
[107] Y. Benjamini,et al. Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .
[108] Charles Elkan,et al. Fitting a Mixture Model By Expectation Maximization To Discover Motifs In Biopolymer , 1994, ISMB.