Chromatin Accessibility Programs CD8 T Cell Memory Christopher D

Total Page:16

File Type:pdf, Size:1020Kb

Chromatin Accessibility Programs CD8 T Cell Memory Christopher D Cutting Edge: Chromatin Accessibility Programs CD8 T Cell Memory Christopher D. Scharer, Alexander P. R. Bally, Bhanu Gandham and Jeremy M. Boss This information is current as of October 1, 2021. J Immunol 2017; 198:2238-2243; Prepublished online 8 February 2017; doi: 10.4049/jimmunol.1602086 http://www.jimmunol.org/content/198/6/2238 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2017/02/07/jimmunol.160208 Material 6.DCSupplemental References This article cites 30 articles, 8 of which you can access for free at: http://www.jimmunol.org/content/198/6/2238.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on October 1, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2017 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Th eJournal of Cutting Edge Immunology Cutting Edge: Chromatin Accessibility Programs CD8 T Cell Memory Christopher D. Scharer,1 Alexander P. R. Bally,1 Bhanu Gandham, and Jeremy M. Boss CD8 T cell memory is characterized by rapid recall of been studied (1–4), it is still unclear how these properties are effector function, increased proliferation, and reduced molecularly programmed following Ag encounter and main- activation requirements. Despite the extensive func- tained during homeostasis. The functional fate of T cells in- tional characterization, the molecular mechanisms that volves the integration of external signals through distinct facilitate these enhanced properties are not well charac- transcription factor complexes, such as Nfatc1 (5), at cis- terized. In this study, the assay for transposase- regulatory sites; however, the catalog of cis-regulatory sites that participate in CD8 T cell differentiation is not known. accessible chromatin sequencing was employed to Downloaded from map the cis-regulatory elements in CD8 T cells The dynamic alterations in transcriptional and epigenetic responding to acute and chronic lymphocytic chorio- programs, such as DNA methylation and histone modifica- tions, that are enacted during the different stages of the CD8 meningitis virus infections. Integration of chromatin T cell response to infection have been profiled in multiple accessibility profiles with gene expression data identi- systems (6–12); however, these datasets have not been inte- fied unique regulatory modules that were enriched for grated. The consequences of epigenetic mechanisms and http://www.jimmunol.org/ distinct combinations of transcription factor–binding programs can be observed through the changes in chromatin motifs. Memory CD8 T cells displayed a chromatin accessibility. In this study, the assay for transposase-accessible accessibility structure that was absent from other acute chromatin sequencing (ATAC-seq) (13) was used to define and exhausted cells types and included key effector and distinct epigenetic profiles in T cells during different stages of proliferative genes. Stimulation of memory cells acute and chronic viral infections. These profiles reveal a revealed enhanced transcription of “memory-primed” unique epigenetic state of memory CD8 T cells that may genes compared with naive cells. Thus, memory CD8 provide a mechanism for enhanced function. T cells display a preprogrammed chromatin accessibil- ity profile and maintain a molecular history of cis- Materials and Methods by guest on October 1, 2021 element usage, thereby reducing the steps necessary to Mice and lymphocytic choriomeningitis virus infection revive effector functions. The Journal of Immunology, Wild-type C57BL/6J mice were obtained from The Jackson Laboratory and 2017, 198: 2238–2243. bred on-site. Thy1.1+ P14 mice and viral stocks of lymphocytic choriomen- ingitis virus (LCMV) strains Armstrong and clone 13 were provided by Dr. Rafi Ahmed (Emory University). Infections were performed on 6- to 8-wk- old mice as previously described (14), and viral titers were confirmed by pon recognition of cognate Ag and given the proper plaque assay (15). All animal protocols were approved by the Emory Uni- costimulatory signals, CD8 T cells proliferate and versity Institutional Animal Care and Use Committee. U differentiate into short-lived effector cells capable of killing infected cells. Following Ag clearance, a small pop- Cell isolation and analysis ulation of long-lived memory cells emerges that has unique CD8 T cells were enriched from splenocytes by MACS (Miltenyi Biotec) in functional properties from their naive precursors, including accordance with the manufacturer’s instructions, stained in FACS buffer lower activation requirements, enhanced proliferative capacity, (PBS, 1% BSA, 1 mM EDTA), and sorted on a BD FACSAria II. Abs used for sorting were from Tonbo Biosciences (CD8 FITC, CD4 PerCP-Cy5.5, and more rapid induction of effector genes (1–4). Although CD44 allophycocyanin-Cy7) or BioLegend (CD62L Alexa Fluor 700, the functional enhancements of memory CD8 T cells have Thy1.1 Pacific Blue, and PD-1 PE). Mouse MHC class I (H-2Db) tetramers Department of Microbiology and Immunology, Emory University School of Medicine, Address correspondence and reprint requests to Dr. Jeremy M. Boss, Emory University, Atlanta, GA 30322; and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Road, Room 3001, Atlanta, GA 30322. E-mail address: jmboss@emory. Atlanta, GA 30322 edu 1C.D.S. and A.P.R.B. contributed equally to this work. The online version of this article contains supplemental material. ORCIDs: 0000-0001-7716-8504 (C.D.S.); 0000-0003-4494-5033 (A.P.R.B.); 0000- Abbreviations used in this article: aD, LCMV Armstrong on day; ATAC-seq, assay for 0002-4027-1062 (B.G.); 0000-0002-2432-1840 (J.M.B.). transposase-accessible chromatin sequencing; clD, LCMV clone 13 on day; DAR, dif- ferentially accessible region; DEG, differentially expressed gene; GO, Gene Ontology; Received for publication December 21, 2016. Accepted for publication January 18, LCMV, lymphocytic choriomeningitis virus; PC, principal component; PCA, principal 2017. component analysis. This work was supported by National Institutes of Health Grant R01 AI113021 (to J.M.B.). A.P.R.B. was supported by National Institutes of Health Grant T32 AI007610. Copyright Ó 2017 by The American Association of Immunologists, Inc. 0022-1767/17/$30.00 The ATAC-sequencing data presented in this article have been submitted to the National Center for Biotechnology Information Gene Expression Omnibus database (http://www. ncbi.nlm.nih.gov/geo/) under accession number GSE83081. www.jimmunol.org/cgi/doi/10.4049/jimmunol.1602086 The Journal of Immunology 2239 corresponding to LCMV peptides gp33 var C41M (KAVYNFATM), gp276 Chromatin accessibility and gene expression patterns are linked to (SGVENPGGYCL), and np396 (FQPQNGQFI) were obtained from the T cell function National Institutes of Health Tetramer Core facility at Emory University. For ex vivo experiments, 104 P14 CD8 T cells were adoptively transferred Chromatin accessibility data from naive, day 8, and day 30 into wild-type hosts. After 1 d, mice were infected with LCMV Armstrong. At were integrated with their respective gene expression data (6). day 28, splenocytes from immune mice or naive P14 mice were stimulated with 4 mM LCMV gp33 var C41M peptide for the times indicated. Cells DARs (n = 8239) were mapped to a differentially expressed were fixed in 1% paraformaldehyde, and virus-specific P14 cells were sorted gene (DEG), and a normalized Euclidean distance metric by FACS using markers described above. RNA was prepared using the Pin- followed by k-means clustering was used to categorize the point slide RNA isolation system (Zymo Research). patterns of gene expression and accessibility changes at all loci relative to each other (23). Seven distinct k-means mod- ATAC-seq and data analyses ules were identified (Fig. 2A, Supplemental Fig. 1D, ATAC-seq libraries were generated from 104 cells for each sample, sequenced, Supplemental Table III). Some modules demonstrated coor- and data processed as detailed previously (16). Differential accessibility was dinate increases (1, 7) or decreases (2, 6) in accessibility and . determined using edgeR (17), and loci with accessibility changes 2-fold and gene expression from naive to day 30 CD8 T cells. In module a false discovery rate of ,0.05 were called significant. Gene Ontology (GO) term enrichment was determined using DAVID (18) and motifs identified 4, both accessibility and gene expression increased to day 8, with HOMER (19) using all significant acute time point peaks as back- followed by a subsequent decrease in both parameters at ground. For transcription factor footprinting analysis, a bed file of motif aD30. With the exception of module 4, progressive/linear occurrences in the differentially accessible region (DAR) was used as input for the HOMER (19) annotatePeaks.pl script with the following options: “-norm
Recommended publications
  • Supplementary Materials: Evaluation of Cytotoxicity and Α-Glucosidase Inhibitory Activity of Amide and Polyamino-Derivatives of Lupane Triterpenoids
    Supplementary Materials: Evaluation of cytotoxicity and α-glucosidase inhibitory activity of amide and polyamino-derivatives of lupane triterpenoids Oxana B. Kazakova1*, Gul'nara V. Giniyatullina1, Akhat G. Mustafin1, Denis A. Babkov2, Elena V. Sokolova2, Alexander A. Spasov2* 1Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences, 71, pr. Oktyabrya, 450054 Ufa, Russian Federation 2Scientific Center for Innovative Drugs, Volgograd State Medical University, Novorossiyskaya st. 39, Volgograd 400087, Russian Federation Correspondence Prof. Dr. Oxana B. Kazakova Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences 71 Prospeсt Oktyabrya Ufa, 450054 Russian Federation E-mail: [email protected] Prof. Dr. Alexander A. Spasov Scientific Center for Innovative Drugs of the Volgograd State Medical University 39 Novorossiyskaya st. Volgograd, 400087 Russian Federation E-mail: [email protected] Figure S1. 1H and 13C of compound 2. H NH N H O H O H 2 2 Figure S2. 1H and 13C of compound 4. NH2 O H O H CH3 O O H H3C O H 4 3 Figure S3. Anticancer screening data of compound 2 at single dose assay 4 Figure S4. Anticancer screening data of compound 7 at single dose assay 5 Figure S5. Anticancer screening data of compound 8 at single dose assay 6 Figure S6. Anticancer screening data of compound 9 at single dose assay 7 Figure S7. Anticancer screening data of compound 12 at single dose assay 8 Figure S8. Anticancer screening data of compound 13 at single dose assay 9 Figure S9. Anticancer screening data of compound 14 at single dose assay 10 Figure S10.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • 140503 IPF Signatures Supplement Withfigs Thorax
    Supplementary material for Heterogeneous gene expression signatures correspond to distinct lung pathologies and biomarkers of disease severity in idiopathic pulmonary fibrosis Daryle J. DePianto1*, Sanjay Chandriani1⌘*, Alexander R. Abbas1, Guiquan Jia1, Elsa N. N’Diaye1, Patrick Caplazi1, Steven E. Kauder1, Sabyasachi Biswas1, Satyajit K. Karnik1#, Connie Ha1, Zora Modrusan1, Michael A. Matthay2, Jasleen Kukreja3, Harold R. Collard2, Jackson G. Egen1, Paul J. Wolters2§, and Joseph R. Arron1§ 1Genentech Research and Early Development, South San Francisco, CA 2Department of Medicine, University of California, San Francisco, CA 3Department of Surgery, University of California, San Francisco, CA ⌘Current address: Novartis Institutes for Biomedical Research, Emeryville, CA. #Current address: Gilead Sciences, Foster City, CA. *DJD and SC contributed equally to this manuscript §PJW and JRA co-directed this project Address correspondence to Paul J. Wolters, MD University of California, San Francisco Department of Medicine Box 0111 San Francisco, CA 94143-0111 [email protected] or Joseph R. Arron, MD, PhD Genentech, Inc. MS 231C 1 DNA Way South San Francisco, CA 94080 [email protected] 1 METHODS Human lung tissue samples Tissues were obtained at UCSF from clinical samples from IPF patients at the time of biopsy or lung transplantation. All patients were seen at UCSF and the diagnosis of IPF was established through multidisciplinary review of clinical, radiological, and pathological data according to criteria established by the consensus classification of the American Thoracic Society (ATS) and European Respiratory Society (ERS), Japanese Respiratory Society (JRS), and the Latin American Thoracic Association (ALAT) (ref. 5 in main text). Non-diseased normal lung tissues were procured from lungs not used by the Northern California Transplant Donor Network.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • DCIR2 Cdc2 Dcs and Zbtb32 Restore CD4 T-Cell Tolerance
    Diabetes Volume 64, October 2015 3521 Jeffrey D. Price, Chie Hotta-Iwamura, Yongge Zhao, Nicole M. Beauchamp, and Kristin V. Tarbell DCIR2+ cDC2 DCs and Zbtb32 Restore CD4+ T-Cell Tolerance and Inhibit Diabetes Diabetes 2015;64:3521–3531 | DOI: 10.2337/db14-1880 During autoimmunity, the normal ability of dendritic cells clinical efficacy has been observed for this approach (1,2). (DCs) to induce T-cell tolerance is disrupted; therefore, Autoimmune individuals elicit immune responses in an autoimmune disease therapies based on cell types and inflammatory context and are therefore refractory to tol- IMMUNOLOGY AND TRANSPLANTATION molecular pathways that elicit tolerance in the steady erance induction, yet most studies of T-cell tolerance have state may not be effective. To determine which DC been performed in either a steady-state context or in subsets induce tolerance in the context of chronic models of autoimmunity requiring immunization with fi autoimmunity, we used chimeric antibodies speci cfor autoantigen that best model the effector phase (3). There- DC inhibitory receptor 2 (DCIR2) or DEC-205 to target fi + + fore, to move beyond therapies that nonspeci cally block self-antigen to CD11b (cDC2) DCs and CD8 (cDC1) DCs, effector functions, it is important to learn what condi- respectively, in autoimmune-prone nonobese diabetic tions are needed to enable antigen-specific T-cell tolerance (NOD) mice. Antigen presentation by DCIR2+ DCs but induction in a chronic inflammatory autoimmune envi- not DEC-205+ DCs elicited tolerogenic CD4+ T-cell ronment, which can be modeled using autoimmune-prone responses in NOD mice. b-Cell antigen delivered to DCIR2+ DCs delayed diabetes induction and induced in- nonobese diabetic (NOD) mice that show spontaneous creased T-cell apoptosis without interferon-g (IFN-g)or loss of self-tolerance due to genetic and environmental sustained expansion of autoreactive CD4+ T cells.
    [Show full text]
  • XPO7 Is a Tumor Suppressor Regulating P21cip1-Dependent Senescence
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press XPO7 is a tumor suppressor regulating p21CIP1-dependent senescence Andrew J. Innes,1,2,3 Bin Sun,1,2 Verena Wagner,1,2 Sharon Brookes,1,2 Domhnall McHugh,1,2 Joaquim Pombo,1,2 Rosa María Porreca,1,2 Gopuraja Dharmalingam,1,2 Santiago Vernia,1,2 Johannes Zuber,4 Jean-Baptiste Vannier,1,2 Ramón García-Escudero,5,6,7 and Jesús Gil1,2 1MRC London Institute of Medical Sciences (LMS), London W12 0NN, United Kingdom; 2Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom; 3Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom; 4Research Institute of Molecular Pathology (IMP), 1030 Vienna, Austria; 5Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; 6Research Institute 12 de Octubre (i+12), 28041 Madrid, Spain; 7Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain Senescence is a key barrier to neoplastic transformation. To identify senescence regulators relevant to cancer, we screened a genome-wide shRNA library. Here, we describe exportin 7 (XPO7) as a novel regulator of senescence and validate its function in telomere-induced, replicative, and oncogene-induced senescence (OIS). XPO7 is a bidirec- tional transporter that regulates the nuclear-cytoplasmic shuttling of a broad range of substrates. Depletion of XPO7 results in reduced levels of TCF3 and an impaired induction of the cyclin-dependent kinase inhibitor p21CIP1 during OIS. Deletion of XPO7 correlates with poorer overall survival in several cancer types.
    [Show full text]
  • Potential Genotoxicity from Integration Sites in CLAD Dogs Treated Successfully with Gammaretroviral Vector-Mediated Gene Therapy
    Gene Therapy (2008) 15, 1067–1071 & 2008 Nature Publishing Group All rights reserved 0969-7128/08 $30.00 www.nature.com/gt SHORT COMMUNICATION Potential genotoxicity from integration sites in CLAD dogs treated successfully with gammaretroviral vector-mediated gene therapy M Hai1,3, RL Adler1,3, TR Bauer Jr1,3, LM Tuschong1, Y-C Gu1,XWu2 and DD Hickstein1 1Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA and 2Laboratory of Molecular Technology, Scientific Applications International Corporation-Frederick, National Cancer Institute-Frederick, Frederick, Maryland, USA Integration site analysis was performed on six dogs with in hematopoietic stem cells. Integrations clustered around canine leukocyte adhesion deficiency (CLAD) that survived common insertion sites more frequently than random. greater than 1 year after infusion of autologous CD34+ bone Despite potential genotoxicity from RIS, to date there has marrow cells transduced with a gammaretroviral vector been no progression to oligoclonal hematopoiesis and no expressing canine CD18. A total of 387 retroviral insertion evidence that vector integration sites influenced cell survival sites (RIS) were identified in the peripheral blood leukocytes or proliferation. Continued follow-up in disease-specific from the six dogs at 1 year postinfusion. A total of 129 RIS animal models such as CLAD will be required to provide an were identified in CD3+ T-lymphocytes and 102 RIS in accurate estimate
    [Show full text]
  • Figure S17 Figure S16
    immune responseregulatingcellsurfacereceptorsignalingpathway ventricular cardiacmuscletissuedevelopment t cellactivationinvolvedinimmuneresponse intrinsic apoptoticsignalingpathway single organismalcelladhesion cholesterol biosyntheticprocess myeloid leukocytedifferentiation determination ofadultlifespan response tointerferongamma muscle organmorphogenesis endothelial celldifferentiation brown fatcelldifferentiation mitochondrion organization myoblast differentiation response toprotozoan amino acidtransport leukocyte migration cytokine production t celldifferentiation protein secretion response tovirus angiogenesis Scrt1 Tcf25 Dpf1 Sap30 Ing2 Zfp654 Sp9 Zfp263 Mxi1 Hes6 Zfp395 Rlf Ppp1r13l Snapc1 C030039L03Rik Hif1a Arrb1 Glis3 Rcor2 Hif3a Fbxo21 Dnajc21 Rest Sirt6 Foxj1 Kdm5b Ankzf1 Sos2 Plscr1 Jdp2 Rbbp8 Etv4 Msh5 Mafg Tsc22d3 Nupr1 Ddit3 Cebpg Zfp790 Atf5 Cebpb Atf3 Trim21 Irf9 Irf2 Tbx21 Stat2 Stat1 Zbp1 Irf1 aGOslimPos Ikzf3 Oasl1 Irf7 Trim30a Dhx58 Txk Rorc Rora Nr1d2 Setdb2 Vdr Vax2 Nr1d1 Foxs1 Eno1 Thap3 Nfkbil1 Edf1 Srebf1 Lbr Tead1 Zfp608 Pcx Ift57 Ssbp4 Stat3 Mxd1 Pml Ssh2 Chd7 Maf Cic Bcl3 Prkdc Mbd5 Ppfibp2 Foxp2 Tal2 Mlf1 Bcl6b Zfp827 Ikzf2 Phtf2 Bmyc Plagl2 Nfkb2 Nfkb1 Tox Nrip1 Utf1 Klf3 Plagl1 Nfkbib Spib Nfkbie Akna Rbpj Ncoa3 Id1 Tnp2 Gata3 Gata1 Pparg Id2 Epas1 Zfp280b Commons Pathway Erg GO MSigDB KEGG Hhex WikiPathways SetGene Databases Batf Aff3 Zfp266 gene modules other (hypergeometric TF, from Figure Trim24 Zbtb5 Foxo3 Aebp2 XPodNet -protein-proteininteractionsinthepodocyteexpandedbySTRING Ppp1r10 Dffb Trp53 Enrichment
    [Show full text]
  • Lineage Transcription Factors Co-Regulate Subtype-Specific Genes Providing a Roadmap For
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.13.249029; this version posted August 14, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Lineage transcription factors co-regulate subtype-specific genes providing a roadmap for systematic identification of small cell lung cancer vulnerabilities Karine Pozo1,2, Rahul K. Kollipara3, Demetra P. Kelenis1, Kathia E. Rodarte1, Xiaoyang Zhang4, John D. Minna5,6,7,8 and Jane E. Johnson1,6,8 1Department of Neuroscience, 2Department of Surgery, 3McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX 75390, USA 4Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 5Hamon Center for Therapeutic Oncology Research, 6Simmons Comprehensive Cancer Center, 7Department of Internal Medicine, 8Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA JDM receives licensing fees for lung cancer lines from the NIH and UT Southwestern. 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.08.13.249029; this version posted August 14, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. ABSTRACT Lineage-defining transcription factors (LTFs) play key roles in tumor cell growth, making them highly attractive, but currently “undruggable”, small cell lung cancer (SCLC) vulnerabilities.
    [Show full text]
  • MDM4 Is Targeted by 1Q Gain and Drives Disease in Burkitt Lymphoma
    Published OnlineFirst April 18, 2019; DOI: 10.1158/0008-5472.CAN-18-3438 Cancer Translational Science Research MDM4 Is Targeted by 1q Gain and Drives Disease in Burkitt Lymphoma Jennifer Hullein€ 1,2, Mikołaj Słabicki1, Maciej Rosolowski3, Alexander Jethwa1,2, Stefan Habringer4, Katarzyna Tomska1, Roma Kurilov5, Junyan Lu6, Sebastian Scheinost1, Rabea Wagener7,8, Zhiqin Huang9, Marina Lukas1, Olena Yavorska6, Hanne Helfrich10,Rene Scholtysik11, Kyle Bonneau12, Donato Tedesco12,RalfKuppers€ 11, Wolfram Klapper13, Christiane Pott14, Stephan Stilgenbauer10, Birgit Burkhardt15, Markus Lof€ fler3, Lorenz H. Trumper€ 16, Michael Hummel17, Benedikt Brors5, Marc Zapatka9, Reiner Siebert7,8, Markus Kreuz3, Ulrich Keller4,18, Wolfgang Huber6, and Thorsten Zenz1,19 Abstract Oncogenic MYC activation promotes proliferation in growth in a xenograft model in a p53-dependent manner. Burkitt lymphoma, but also induces cell-cycle arrest and Small molecule inhibition of the MDM4–p53 interaction apoptosis mediated by p53, a tumor suppressor that is was effective only in TP53wt Burkitt lymphoma cell lines. mutated in 40% of Burkitt lymphoma cases. To identify Moreover, primary TP53wt Burkitt lymphoma samples fre- molecular dependencies in Burkitt lymphoma, we per- quently acquired gains of chromosome 1q, which includes formed RNAi-based, loss-of-function screening in eight the MDM4 locus, and showed elevated MDM4 mRNA levels. Burkitt lymphoma cell lines and integrated non-Burkitt 1q gain was associated with TP53wt across 789 cancer cell lymphoma RNAi screens and genetic data. We identified lines and MDM4 was essential in the TP53wt-context in 216 76 genes essential to Burkitt lymphoma, including genes cell lines representing 19 cancer entities from the Achilles associated with hematopoietic cell differentiation (FLI1, Project.
    [Show full text]
  • Subtype-Specific Addiction of the Activated B-Cell Subset of Diffuse
    Subtype-specific addiction of the activated B-cell PNAS PLUS subset of diffuse large B-cell lymphoma to FOXP1 Joseph D. Dekkera, Daechan Parka,b, Arthur L. Shaffer IIIc, Holger Kohlhammerc, Wei Denga, Bum-Kyu Leea, Gregory C. Ippolitoa,b, George Georgioua,b, Vishwanath R. Iyera, Louis M. Staudtc,1, and Haley O. Tuckera,1 aDepartment of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712; bDepartment of Chemical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712; and cLymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 Contributed by Louis M. Staudt, December 21, 2015 (sent for review October 8, 2015; reviewed by George Q. Daley and Edward E. Morrisey) High expression of the forkhead box P1 (FOXP1) transcription fac- (FOXP1) (9). Implicated in development, proliferation, and differ- tor distinguishes the aggressive activated B cell (ABC) diffuse large entiation in multiple contexts (12–16), FOXP1 is highly expressed in B-cell lymphoma (DLBCL) subtype from the better prognosis ger- ABC-DLBCL relative to its levels in normal PB or in GCB-DLBCL minal center B-cell (GCB)-DLBCL subtype and is highly correlated (2, 3, 9, 17). Chromosomal loss, gain, or fusion of FOXP1 is associ- with poor outcomes. A genetic or functional role for FOXP1 in ated with several B-cell malignancies, including a modest frequency lymphomagenesis, however, remains unknown. Here, we report in ABC-DLBCL (18–20). However, ABC-DLBCL tumors without that sustained FOXP1 expression is vital for ABC-DLBCL cell-line such aberrations still show increased FOXP1 levels relative to GCB- survival.
    [Show full text]
  • Characterization of TCF3 Rearrangements in Pediatric B-Lymphoblastic Leukemia/Lymphoma by Mate-Pair Sequencing
    Rowsey et al. Blood Cancer Journal (2019) 9:81 https://doi.org/10.1038/s41408-019-0239-z Blood Cancer Journal ARTICLE Open Access Characterization of TCF3 rearrangements in pediatric B-lymphoblastic leukemia/lymphoma by mate-pair sequencing (MPseq) identifies complex genomic rearrangements and a novel TCF3/TEF gene fusion Ross A. Rowsey1, Stephanie A. Smoley1, Cynthia M. Williamson1,GeorgeVasmatzis2, James B. Smadbeck2,YiNing3, Patricia T. Greipp1, Nicole L. Hoppman1, Linda B. Baughn1, Rhett P. Ketterling1 and Jess F. Peterson1 Abstract The TCF3/PBX1 gene fusion is a recurrent genetic abnormality in pediatric B-lymphoblastic leukemia/lymphoma (B-ALL/LBL). While dual-color, dual-fusion fluorescence in situ hybridization (D-FISH) probes can detect TCF3/PBX1 fusions, further characterization of atypical TCF3 FISH patterns as indicated by additional or diminished TCF3 signals is currently limited. Herein we describe the use of a next-generation sequencing assay, mate-pair sequencing (MPseq), to characterize typical and cryptic TCF3/PBX1 fusions and to identify TCF3 translocation partners based on results obtained from our laboratory-developed TCF3/PBX1 D-FISH probe set. MPseq was performed on 21 cases of pediatric B-ALL/LBL with either TCF3/PBX1 fusion, or no TCF3/PBX1 fusion but with additional or diminished TCF3 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; signals obtained by our PBX1/TCF3 D-FISH probe set. In addition, MPseq was performed on one pediatric B-ALL/LBL case with an apparently normal karyotype and abnormal TCF3 break-apart probe results. Of 22 specimens successfully evaluated by MPseq, 13 cases (59%) demonstrated TCF3/PBX1 fusion, including three cases with previously undescribed insertional rearrangements.
    [Show full text]