Collateral lethality between HDAC1 and HDAC2 exploits cancer-specific NuRD complex vulnerabilities
暂无分享,去创建一个
Christopher J. Ott | B. Cravatt | M. Janiszewska | Adam D. Durbin | Michael A. Erb | David Remillard | S. Nance | Yuxiang Zhang | U. Onubogu | Barbara Karakyriakou | Joshua N. Asiaban | Anissa R. Ramos | Kirsten Bowland | Paige Barta | T. Bishop
[1] John A. Tallarico,et al. Rational Chemical Design of Molecular Glue Degraders , 2022, bioRxiv.
[2] S. Cowley,et al. A ‘click’ chemistry approach to novel entinostat (MS-275) based class I histone deacetylase proteolysis targeting chimeras , 2022, RSC medicinal chemistry.
[3] S. Cowley,et al. Comprehensive Transcriptomic Analysis of Novel Class I HDAC Proteolysis Targeting Chimeras (PROTACs) , 2022, Biochemistry.
[4] J. Mackay,et al. A NuRD for all seasons. , 2022, Trends in biochemical sciences.
[5] A. Gopalsamy. Selectivity through Targeted Protein Degradation (TPD). , 2022, Journal of medicinal chemistry.
[6] S. Cowley,et al. Optimization of Class I Histone Deacetylase PROTACs Reveals that HDAC1/2 Degradation is Critical to Induce Apoptosis and Cell Arrest in Cancer Cells , 2022, Journal of medicinal chemistry.
[7] X. Chen,et al. Selective PROTAC-mediated degradation of SMARCA2 is efficacious in SMARCA4 mutant cancers , 2022, Nature Communications.
[8] John G Doench,et al. Paralog knockout profiling identifies DUSP4 and DUSP6 as a digenic dependence in MAPK pathway-driven cancers , 2021, Nature Genetics.
[9] Michael A. Erb,et al. Enabling cancer target validation with genetically encoded systems for ligand-induced protein degradation , 2021, Current Research in Chemical Biology.
[10] A. Berger,et al. Discovery of synthetic lethal and tumor suppressor paralog pairs in the human genome , 2021, Cell reports.
[11] A. Razov,et al. Chemo-proteomics exploration of HDAC degradability by small molecule degraders. , 2021, Cell chemical biology.
[12] Joshua M. Dempster,et al. Selective modulation of a pan-essential protein as a therapeutic strategy in cancer. , 2021, Cancer discovery.
[13] Natalie S. Scholes,et al. Identification and selectivity profiling of small-molecule degraders via multi-omics approaches. , 2021, Cell chemical biology.
[14] Joshua M. Dempster,et al. A First-Generation Pediatric Cancer Dependency Map , 2021, Nature Genetics.
[15] G. Winter,et al. Fast-acting chemical tools to delineate causality in transcriptional control. , 2021, Molecular cell.
[16] André F. Rendeiro,et al. Acute BAF perturbation causes immediate changes in chromatin accessibility , 2021, Nature Genetics.
[17] W. Sellers,et al. Targeting pan-essential genes in cancer: challenges and opportunities. , 2020, Cancer cell.
[18] P. Gaulard,et al. Final Analysis of the Ro-CHOP Phase III Study (Conducted by LYSA): Romidepsin Plus CHOP in Patients with Peripheral T-Cell Lymphoma , 2020 .
[19] J. Khan,et al. NuRD subunit CHD4 regulates super-enhancer accessibility in rhabdomyosarcoma and represents a general tumor dependency , 2020, eLife.
[20] S. Schreiber,et al. An Activity-Guided Map of Electrophile-Cysteine Interactions in Primary Human T Cells , 2020, Cell.
[21] A. Look,et al. ARID1A loss in neuroblastoma promotes the adrenergic-to-mesenchymal transition by regulating enhancer-mediated gene expression , 2020, Science Advances.
[22] S. Cowley,et al. PROTAC-mediated degradation of class I histone deacetylase enzymes in corepressor complexes. , 2020, Chemical communications.
[23] J. Mackay,et al. The Nucleosome Remodeling and Deacetylase Complex Has an Asymmetric, Dynamic, and Modular Architecture , 2020, bioRxiv.
[24] P. Northcott,et al. Large 1p36 Deletions Affecting Arid1a Locus Facilitate Mycn-Driven Oncogenesis in Neuroblastoma , 2020, Cell reports.
[25] David M. Wilson,et al. EED-Targeted PROTACs Degrade EED, EZH2, and SUZ12 in the PRC2 Complex. , 2019, Cell chemical biology.
[26] A. Ashworth,et al. Synthetic lethality as an engine for cancer drug target discovery , 2019, Nature Reviews Drug Discovery.
[27] J. Khan,et al. Histone hyperacetylation disrupts core gene regulatory architecture in rhabdomyosarcoma , 2019, Nature Genetics.
[28] Teng Teng,et al. Optimization of AsCas12a for combinatorial genetic screens in human cells , 2019, bioRxiv.
[29] J. Khan,et al. Chemical genomics reveals histone deacetylases are required for core regulatory transcription , 2019, Nature Communications.
[30] Matthew C. Canver,et al. Rational targeting of a NuRD subcomplex guided by comprehensive in situ mutagenesis , 2019, Nature Genetics.
[31] Arndt von Haeseler,et al. Quantification of experimentally induced nucleotide conversions in high-throughput sequencing datasets , 2019, BMC Bioinformatics.
[32] M. Koegl,et al. BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design , 2019 .
[33] G. Parmigiani,et al. Deciphering the chronology of copy number alterations in Multiple Myeloma , 2019, Blood Cancer Journal.
[34] Dorte B. Bekker-Jensen,et al. Integrated proximal proteomics reveals IRS2 as a determinant of cell survival in ALK-driven neuroblastoma , 2018, Science Signaling.
[35] 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.
[36] Paul Bertone,et al. The Nucleosome Remodeling and Deacetylation Complex Modulates Chromatin Structure at Sites of Active Transcription to Fine-Tune Gene Expression , 2018, Molecular cell.
[37] R. Young,et al. Selective gene dependencies in MYCN-amplified neuroblastoma include the core transcriptional regulatory circuitry , 2018, Nature Genetics.
[38] Aviad Tsherniak,et al. Interrogation of Mammalian Protein Complex Structure, Function, and Membership Using Genome-Scale Fitness Screens. , 2018, Cell systems.
[39] James E. Bradner,et al. The dTAG system for immediate and target-specific protein degradation , 2018, Nature Chemical Biology.
[40] Donald R. Polaski,et al. Enhancer invasion shapes MYCN-dependent transcriptional amplification in neuroblastoma , 2018, Nature Genetics.
[41] T. Golub,et al. CRISPR-Cas9 screen reveals a MYCN-amplified neuroblastoma dependency on EZH2 , 2017, The Journal of clinical investigation.
[42] R. Young,et al. Pharmacological perturbation of CDK9 using selective CDK9 inhibition or degradation. , 2017, Nature chemical biology.
[43] Johannes Zuber,et al. Thiol-linked alkylation of RNA to assess expression dynamics , 2017, Nature Methods.
[44] S. Henikoff,et al. Targeted in situ genome-wide profiling with high efficiency for low cell numbers , 2018, Nature Protocols.
[45] Nicholas A. Sinnott-Armstrong,et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues , 2017, Nature Methods.
[46] Phillip G. Montgomery,et al. Defining a Cancer Dependency Map , 2017, Cell.
[47] Howard Y. Chang,et al. Chromatin Accessibility Landscape of Cutaneous T Cell Lymphoma and Dynamic Response to HDAC Inhibitors. , 2017, Cancer cell.
[48] Ann E. Sizemore,et al. Computational correction of copy-number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells , 2017, Nature Genetics.
[49] Catarina P. Santos,et al. Synthetic lethality between the cohesin subunits STAG1 and STAG2 in diverse cancer contexts , 2017, bioRxiv.
[50] S. Armstrong,et al. Degradation of the BAF Complex Factor BRD9 by Heterobifunctional Ligands. , 2017, Angewandte Chemie.
[51] J. Schwabe,et al. Targeting Class I Histone Deacetylases in a "Complex" Environment. , 2017, Trends in pharmacological sciences.
[52] James E. Bradner,et al. A chemical probe toolbox for dissecting the cancer epigenome , 2017, Nature Reviews Cancer.
[53] Neville E. Sanjana,et al. Transcription control by the ENL YEATS domain in acute leukemia , 2016, Nature.
[54] E. Seto,et al. HDACs and HDAC Inhibitors in Cancer Development and Therapy. , 2016, Cold Spring Harbor perspectives in medicine.
[55] Kyle L. Morris,et al. The structure of the core NuRD repression complex provides insights into its interaction with chromatin , 2016, eLife.
[56] J. Shearstone,et al. Chemical Inhibition of Histone Deacetylases 1 and 2 Induces Fetal Hemoglobin through Activation of GATA2 , 2016, PloS one.
[57] Mariko Sasaki,et al. Targeting p300 Addiction in CBP-Deficient Cancers Causes Synthetic Lethality by Apoptotic Cell Death due to Abrogation of MYC Expression. , 2016, Cancer discovery.
[58] Tetsushi Sakuma,et al. MMEJ-assisted gene knock-in using TALENs and CRISPR-Cas9 with the PITCh systems , 2015, Nature Protocols.
[59] Meagan E. Sullender,et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9 , 2015, Nature Biotechnology.
[60] J. Zuber,et al. Functional-genetic dissection of HDAC dependencies in mouse lymphoid and myeloid malignancies. , 2015, Blood.
[61] F. Speleman,et al. Inhibition of CDK4/6 as a novel therapeutic option for neuroblastoma , 2015, Cancer Cell International.
[62] J. Kinney,et al. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains , 2015, Nature Biotechnology.
[63] Sebastian A. Wagner,et al. Acetylation site specificities of lysine deacetylase inhibitors in human cells , 2015, Nature Biotechnology.
[64] Tina N. Davis,et al. Selective Inhibition of HDAC1 and HDAC2 as a Potential Therapeutic Option for B-ALL , 2015, Clinical Cancer Research.
[65] Howard Y. Chang,et al. ATAC‐seq: A Method for Assaying Chromatin Accessibility Genome‐Wide , 2015, Current protocols in molecular biology.
[66] B. van Steensel,et al. Easy quantitative assessment of genome editing by sequence trace decomposition , 2014, Nucleic acids research.
[67] R. Johnstone,et al. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders , 2014, Nature Reviews Drug Discovery.
[68] 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.
[69] E. Seto,et al. Erasers of histone acetylation: the histone deacetylase enzymes. , 2014, Cold Spring Harbor perspectives in biology.
[70] C. Roberts,et al. Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers , 2014, Proceedings of the National Academy of Sciences.
[71] W. Hahn,et al. Residual Complexes Containing SMARCA2 (BRM) Underlie the Oncogenic Drive of SMARCA4 (BRG1) Mutation , 2014, Molecular and Cellular Biology.
[72] J. Yokota,et al. A synthetic lethality-based strategy to treat cancers harboring a genetic deficiency in the chromatin remodeling factor BRG1. , 2013, Cancer research.
[73] O. Dovey,et al. Histone deacetylase (HDAC) 1 and 2 are essential for normal T cell development and genomic stability in mice , 2013, Clinical Epigenetics.
[74] S. Cowley,et al. The physiological roles of histone deacetylase (HDAC) 1 and 2: complex co-stars with multiple leading parts. , 2013, Biochemical Society transactions.
[75] Giovanni Roti,et al. Selective HDAC1/HDAC2 inhibitors induce neuroblastoma differentiation. , 2013, Chemistry & biology.
[76] David A. Orlando,et al. Master Transcription Factors and Mediator Establish Super-Enhancers at Key Cell Identity Genes , 2013, Cell.
[77] L. Wessels,et al. Dosage-dependent tumor suppression by histone deacetylases 1 and 2 through regulation of c-Myc collaborating genes and p53 function. , 2013, Blood.
[78] Benjamin E. Gross,et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. , 2012, Cancer discovery.
[79] P. Wade,et al. Cancer biology and NuRD: a multifaceted chromatin remodelling complex , 2011, Nature Reviews Cancer.
[80] J. Epstein,et al. Hdac1 and Hdac2 act redundantly to control p63 and p53 functions in epidermal progenitor cells. , 2010, Developmental cell.
[81] R. DePinho,et al. Overlapping functions of Hdac1 and Hdac2 in cell cycle regulation and haematopoiesis , 2010, The EMBO journal.
[82] O. Dovey,et al. Histone deacetylase 1 (HDAC1), but not HDAC2, controls embryonic stem cell differentiation , 2010, Proceedings of the National Academy of Sciences.
[83] H. Kohler,et al. Histone deacetylases 1 and 2 act in concert to promote the G1-to-S progression. , 2010, Genes & development.
[84] V. P. Collins,et al. Characterization of 6q deletions in mature B cell lymphomas and childhood acute lymphoblastic leukemia , 2008, Leukemia & lymphoma.
[85] R. Starke,et al. Chromosome 1p and 11q Deletions and Outcome in Neuroblastoma—A Critical Review , 2008, Clinical medicine. Oncology.
[86] S. Denslow,et al. The human Mi-2/NuRD complex and gene regulation , 2007, Oncogene.
[87] F. Bertoni,et al. Chromosome band 6q deletion pattern in malignant lymphomas. , 2006, Cancer genetics and cytogenetics.
[88] Pablo Tamayo,et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.
[89] Thomas Helleday,et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase , 2005, Nature.
[90] Gudrun Schleiermacher,et al. Gene expression profiling of 1p35–36 genes in neuroblastoma , 2004, Oncogene.
[91] M. Daly,et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes , 2003, Nature Genetics.
[92] D. O’Carroll,et al. Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression , 2002, The EMBO journal.
[93] G. Gahrton,et al. 6q deletions in acute lymphoblastic leukemia and non-Hodgkin's lymphomas. , 1998, Blood.
[94] S. Schreiber,et al. A Mammalian Histone Deacetylase Related to the Yeast Transcriptional Regulator Rpd3p , 1996, Science.
[95] A. Look,et al. Significance of chromosome 1p loss of heterozygosity in neuroblastoma. , 1995, Cancer research.
[96] Rogier Versteeg,et al. Allelic loss of chromosome 1p36 in neuroblastoma is of preferential maternal origin and correlates with N–myc amplification , 1993, Nature Genetics.
[97] Jacob D. Jaffe,et al. Next-generation characterization of the Cancer Cell Line Encyclopedia , 2019, Nature.