DNA-PK Phosphorylation Sites on Oct-1 Promote Cell Survival Following DNA Damage

Total Page:16

File Type:pdf, Size:1020Kb

DNA-PK Phosphorylation Sites on Oct-1 Promote Cell Survival Following DNA Damage Oncogene (2007) 26, 3980–3988 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc ORIGINAL ARTICLE DNA-PK phosphorylation sites on Oct-1 promote cell survival following DNA damage C Schild-Poulter1,4, A Shih1, D Tantin3,5, NC Yarymowich1, S Soubeyrand1, PA Sharp3 and RJG Hache´ 1,2 1Department of Medicine, The Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada; 2Department of Biochemistry, Microbiology and Immunology, The Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada and 3Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA Octamer transcription factor-1 (Oct-1) has recently been Introduction shown to function as a stress sensor that promotes cell survival subsequent to DNA damage. Here, we show that Cells that are subjected to double-stranded breaks the survival signal imparted by Oct-1 following exposure (DSBs) in their DNA activate cell cycle checkpoints to ionizing radiation (IR) is dependent upon DNA- and alter specific gene transcription patterns to allow the dependent protein kinase (DNA-PK)-dependent phospho- DNA repair machinery to correct the damage and rylation ofa cluster of13 specific ser/thr residues within preserve the integrity of the genome (Valerie and Povirk, the N-terminal transcriptional regulatory domain of 2003). The predominant DSBs repair pathway in higher Oct-1. Although IR treatment did not affect the recruit- eukaryotic cells is the non-homologous DNA end- ment ofOct-1 to the histone H2B promoter, the joining pathway (NHEJ) (Lees-Miller and Meek, 2003; recruitment ofRNA polymerase II, TATA-binding Lieber et al., 2003). Defects in DNA-break repair are a protein and histone H4 acetylation were strongly reduced, major cause of genomic instability that leads to the consistent with a decrease in Oct-1 transcriptional development of cancer. regulatory potential following IR exposure. Ser/Thr-Ala A key NHEJ factor is the DNA-dependent protein substitution of13 sites present in Oct-1 transcriptional kinase (DNA-PK). DNA-PK is comprised of a large regulatory domain eliminated Oct-1 phosphorylation catalytic subunit, DNA-PKcs, and the Ku antigen subsequent to IR exposure. Further, these substitutions heterodimer (Ku70/Ku80) (Smith and Jackson, 1999). prevented Oct-1 from rescuing the survival of IR-treated DNA-PK is essential in maintaining genomic integrity, Oct-12/2 murine embryonic fibroblasts, providing a direct as mice lacking either DNA-PKcs or Ku suffer from link between DNA-PK-dependent phosphorylation and the profound radiosensitivity, immunodeficiency and age contribution ofOct-1 to cell survival. These results prematurely (Ferguson and Alt, 2001; Lieber et al., implicate Oct-1 as a primary effector in a DNA-PK- 2003). At the cellular level, DNA-PK deficiency results dependent cell survival pathway that is activated by in defective repair of DSBs and defects in telomere double-stranded DNA breaks. maintenance that lead to telomeric fusions and translo- Oncogene (2007) 26, 3980–3988; doi:10.1038/sj.onc.1210165; cations (Lieber et al., 2003; Bailey and Goodwin, 2004; published online 8 January 2007 Collis et al., 2005). DNA-PK exhibits Ser/Thr kinase activity and dis- Keywords: Ku; DNA-dependent protein kinase; Oct-1; plays a strong preference for S/T-Q motifs, although S/ DNA damage; phosphorylation T’s flanked by residues other than Q are also recognized on occasion (Chan et al., 1999; Kim et al., 1999). Suggested substrates for DNA-PK include several factors of the NHEJ pathway (Meek et al., 2004; Collis et al., 2005). DNA-PK is also known to autophos- Correspondence: Dr RJG Hache´ , Hormones, Growth and Develop- phorylate (Chan et al., 2002; Douglas et al., 2002; ment, The Ottawa Health Research Institute, 725 Parkdale Avenue, Soubeyrand et al., 2003). A role for DNA-PK in Ottawa, Ontario, Canada K1Y 4E9. Dr C Schild-Poulter, Robarts signaling to apoptosis machinery has been documented Research Institute, P.O. Box 5015, 100, Perth Drive, London, Ontario, (Bharti et al., 1998; Wang et al., 2000; Woo et al., 2002; Canada N6A 5K8. Sawada et al., 2003). However, the potential for DNA- E-mail: [email protected] or [email protected] 4Current address: Cell Biology Research Group, Robarts Research PK to act as a signaling molecule in regulating other Institute, PO Box 5015, 100, Perth Drive, London, Ontario, Canada cellular responses to DSBs remains to be elucidated. N6A 5K8. Octamer transcription factor-1 (Oct-1) has recently 5Current address: Department of Pathology, University of Utah emerged as an important contributor to cell survival in School of Medicine, EEJB-565, Room 5700B, 15 No. Medical Drive East, Salt Lake City, UT 84112-5650, USA. response to various types of DNA damage (Tantin et al., Received 10 April 2006; revised 29 August 2006; accepted 1 September 2005). Somewhat paradoxically, we have determined 2006; published online 8 January 2007 that Oct-1 protein levels increase subsequent to ionizing Modulation of cell survival through Oct-1 phosphorylation C Schild-Poulter et al 3981 radiation (IR) exposure, whereas at the same time H2B a IR and U2 expression is down-regulated (Zhao et al., C 1.5h 5h 2000a; Jin et al., 2001; Schild-Poulter et al., 2003). Oct-1 is a ubiquitous transcription factor of the Input POU (Pit/Oct-1/2-Unc) subfamily of homeodomain Oct-1 proteins (Herr and Cleary, 1995; Ryan and Rosenfeld, 1997) and is a key regulator of housekeeping genes TBP such as H2B and U2 (Sive and Roeder, 1986; Fletcher Ac H4 et al., 1987). Oct-1 also participates in the transcrip- tional regulation of many signal-regulated and tissue- Pol II specific genes and in the control of viral replication Gal4 (Herr and Cleary, 1995; Ryan and Rosenfeld, 1997; de Jong and van der Vliet, 1999; Phillips and Luisi, b 2000). Oct-1 is essential for embryonic development H2B as Oct-1-deficient embryos die during gestation (Wang 18S et al., 2004). Recently, we determined that the stabilization of c Total Oct-1 in response to IRcorrelated with an overall molar Ac H4 increase in Oct-1 phosphorylation and physical interac- tion with Ku (Schild-Poulter et al., 2003). In vitro Figure 1 Oct-1’s presence on the H2B promoter is unaffected by experiments showed that the association of Oct-1 with DNA damage. (a) ChIP analysis of the H2B promoter in MCF-7 cells untreated (C) and harvested 1.5 h or 5 h after 6 Gy irradiation. Ku facilitated the phosphorylation of Oct-1 by DNA- Cross-linked protein–DNA complexes were immunoprecipitated PK (Schild-Poulter et al., 2001, 2003). with antibodies to the proteins indicated and the purified DNA was Here, we have identified 15 specific phosphorylation analysed by polymerase chain reaction with primers amplifying the sites for DNA-PK on Oct-1, 13 of which occur within H2B promoter. Input corresponds to 0.5–1% of equivalent ChIP samples. (b) Northern blot analysis of histone H2B mRNA after IR the N-terminal glutamine-rich Oct-1 transcriptional treatment. MCF-7 cells were irradiated and harvested as above. regulatory domain. Substitution of these 13 residues The RNA was analysed by Northern blot with a H2B cDNA probe with alanine incrementally prevented Oct-1 stabilization and 18S RNA was visualized by SYBR gold staining. (c) Total in response to IRand abrogated the IR-induced Oct-1 acetylated histone H4 levels were analysed by Western blot using phosphorylation previously shown to be dependent on extracts from cells treated as described in panel a. DNA-PK. Notably, the alanine-substituted Oct-1 failed to rescue the radiation sensitivity of Oct-1À/À mouse embryonic fibroblasts (MEFs) seen upon re-expression not a general effect of DNA damage as total acetylated of wild-type (WT) Oct-1. These results provide the first histone H4 levels remain unchanged (Figure 1c). evidence of a role for DNA-PK as a signaling molecule By contrast, the down-regulation of H2B transcrip- in promoting cell survival in response to DSBs through tion in response to IRwas accomplished without modulation of the activity of the transcription factor affecting the promoter occupancy of Oct-1 (Figure 1a). Oct-1. Thus, the IR-dependent modification of Oct-1 that leads to its stabilization did not appear to affect its targeting to chromatin. This result was consistent with previous observations that Oct-1 DNA-binding to an octamer Results motif was unaffected in extracts prepared from cells subjected to IRtreatment (Meighan-Mantha et al., Oct-1 binding to the H2B promoter is unaffected by 1999; Zhao et al., 2000a). This suggested that the salient DNA damage phosphorylation sites for the IRresponse were outside Oct-1 is readily visualized on the H2B promoter in of the POU DNA-binding domain of Oct-1 that proliferating cells (Zhao et al., 2000b). In response to IR interacts with Ku. treatment, H2B transcription decreases (Meighan- Mantha et al., 1999; Schild-Poulter et al., 2003; Su et al., 2004), suggesting that phosphorylation of Oct-1 in DNA-PKphosphorylates Oct-1 at multiple sites in vitro response to IRcould influence the recruitment of The Oct-1 POU domain is flanked by a N-terminal Oct-1 to the histone H2B promoter. To assess how IR region exhibiting a strong Q-rich character and a exposure affected Oct-1-dependent transcription in vivo, C-terminus containing a S/T-rich region (Figure 2a). we performed a chromatin immunoprecipitation (ChIP) To investigate the hypothesis that the DNA-PK- analysis of the H2B promoter (Figure 1a). The decrease dependent changes in Oct-1 subsequent to treatment in histone H2B transcription upon IRexposure with DNA-damaging agents resulted from direct phos- (Figure 1b) correlated with a rapid decrease in the phorylation, we initiated a characterization of the Oct-1 presence of RNA polymerase II and TATA box-binding residues phosphorylated by purified preparations of protein (Figure 1a).
Recommended publications
  • Introduction of Human Telomerase Reverse Transcriptase to Normal Human Fibroblasts Enhances DNA Repair Capacity
    Vol. 10, 2551–2560, April 1, 2004 Clinical Cancer Research 2551 Introduction of Human Telomerase Reverse Transcriptase to Normal Human Fibroblasts Enhances DNA Repair Capacity Ki-Hyuk Shin,1 Mo K. Kang,1 Erica Dicterow,1 INTRODUCTION Ayako Kameta,1 Marcel A. Baluda,1 and Telomerase, which consists of the catalytic protein subunit, No-Hee Park1,2 human telomerase reverse transcriptase (hTERT), the RNA component of telomerase (hTR), and several associated pro- 1School of Dentistry and 2Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California teins, has been primarily associated with maintaining the integ- rity of cellular DNA telomeres in normal cells (1, 2). Telomer- ase activity is correlated with the expression of hTERT, but not ABSTRACT with that of hTR (3, 4). Purpose: From numerous reports on proteins involved The involvement of DNA repair proteins in telomere main- in DNA repair and telomere maintenance that physically tenance has been well documented (5–8). In eukaryotic cells, associate with human telomerase reverse transcriptase nonhomologous end-joining requires a DNA ligase and the (hTERT), we inferred that hTERT/telomerase might play a DNA-activated protein kinase, which is recruited to the DNA role in DNA repair. We investigated this possibility in nor- ends by the DNA-binding protein Ku. Ku binds to hTERT mal human oral fibroblasts (NHOF) with and without ec- without the need for telomeric DNA or hTR (9), binds the topic expression of hTERT/telomerase. telomere repeat-binding proteins TRF1 (10) and TRF2 (11), and Experimental Design: To study the effect of hTERT/ is thought to regulate the access of telomerase to telomere DNA telomerase on DNA repair, we examined the mutation fre- ends (12, 13).
    [Show full text]
  • Novel Roles of Replication Protein a Phosphorylation in Cellular Response to DNA Damage Moises A
    East Tennessee State University Digital Commons @ East Tennessee State University Electronic Theses and Dissertations Student Works 8-2013 Novel Roles of Replication Protein A Phosphorylation in Cellular Response to DNA Damage Moises A. Serrano East Tennessee State University Follow this and additional works at: https://dc.etsu.edu/etd Part of the Biochemistry, Biophysics, and Structural Biology Commons, and the Laboratory and Basic Science Research Commons Recommended Citation Serrano, Moises A., "Novel Roles of Replication Protein A Phosphorylation in Cellular Response to DNA Damage" (2013). Electronic Theses and Dissertations. Paper 1206. https://dc.etsu.edu/etd/1206 This Dissertation - Open Access is brought to you for free and open access by the Student Works at Digital Commons @ East Tennessee State University. It has been accepted for inclusion in Electronic Theses and Dissertations by an authorized administrator of Digital Commons @ East Tennessee State University. For more information, please contact [email protected]. Novel Roles of Replication Protein A Phosphorylation in the Cellular Response to DNA Damage _____________________________ A dissertation presented to the faculty of the Department of Biomedical Science East Tennessee State University In partial fulfillment of the requirements for the degree Doctor of Philosophy in Biomedical Science _____________________________ by Moises Alejandro Serrano August 2013 _____________________________ Yue Zou, Ph.D., Chair Phillip R. Musich, Ph.D. Antonio E. Rusiñol, Ph.D. Michelle M. Duffourc, Ph.D. William L. Stone, Ph.D. Keywords: DNA Repair, DNA Damage Responses, RPA, p53, Apoptosis ABSTRACT Novel Roles of Replication Protein A Phosphorylation in Cellular Response to DNA Damage by Moises Alejandro Serrano Human replication protein A (RPA) is an eukaryotic single-stranded DNA binding protein directly involved in a variety of DNA metabolic pathways including replication, recombination, DNA damage checkpoints and signaling, as well as all DNA repair pathways.
    [Show full text]
  • HOXC4 Rabbit Pab
    Leader in Biomolecular Solutions for Life Science HOXC4 Rabbit pAb Catalog No.: A13856 Basic Information Background Catalog No. This gene belongs to the homeobox family of genes. The homeobox genes encode a A13856 highly conserved family of transcription factors that play an important role in morphogenesis in all multicellular organisms. Mammals possess four similar homeobox Observed MW gene clusters, HOXA, HOXB, HOXC and HOXD, which are located on different 30kDa chromosomes and consist of 9 to 11 genes arranged in tandem. This gene, HOXC4, is one of several homeobox HOXC genes located in a cluster on chromosome 12. Three Calculated MW genes, HOXC5, HOXC4 and HOXC6, share a 5' non-coding exon. Transcripts may include 29kDa the shared exon spliced to the gene-specific exons, or they may include only the gene- specific exons. Two alternatively spliced variants that encode the same protein have Category been described for HOXC4. Transcript variant one includes the shared exon, and transcript variant two includes only gene-specific exons. Primary antibody Applications WB Cross-Reactivity Mouse, Rat Recommended Dilutions Immunogen Information WB 1:500 - 1:2000 Gene ID Swiss Prot 3221 P09017 Immunogen Recombinant fusion protein containing a sequence corresponding to amino acids 30-130 of human HOXC4 (NP_055435.2). Synonyms HOXC4;HOX3;HOX3E;cp19 Contact Product Information www.abclonal.com Source Isotype Purification Rabbit IgG Affinity purification Storage Store at -20℃. Avoid freeze / thaw cycles. Buffer: PBS with 0.02% sodium azide,50% glycerol,pH7.3. Validation Data Western blot analysis of extracts of various cell lines, using HOXC4 antibody (A13856) at 1:3000 dilution.
    [Show full text]
  • Prospective Isolation of NKX2-1–Expressing Human Lung Progenitors Derived from Pluripotent Stem Cells
    The Journal of Clinical Investigation RESEARCH ARTICLE Prospective isolation of NKX2-1–expressing human lung progenitors derived from pluripotent stem cells Finn Hawkins,1,2 Philipp Kramer,3 Anjali Jacob,1,2 Ian Driver,4 Dylan C. Thomas,1 Katherine B. McCauley,1,2 Nicholas Skvir,1 Ana M. Crane,3 Anita A. Kurmann,1,5 Anthony N. Hollenberg,5 Sinead Nguyen,1 Brandon G. Wong,6 Ahmad S. Khalil,6,7 Sarah X.L. Huang,3,8 Susan Guttentag,9 Jason R. Rock,4 John M. Shannon,10 Brian R. Davis,3 and Darrell N. Kotton1,2 2 1Center for Regenerative Medicine, and The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA. 3Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA. 4Department of Anatomy, UCSF, San Francisco, California, USA. 5Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA. 6Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA. 7Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA. 8Columbia Center for Translational Immunology & Columbia Center for Human Development, Columbia University Medical Center, New York, New York, USA. 9Department of Pediatrics, Monroe Carell Jr. Children’s Hospital, Vanderbilt University, Nashville, Tennessee, USA. 10Division of Pulmonary Biology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA. It has been postulated that during human fetal development, all cells of the lung epithelium derive from embryonic, endodermal, NK2 homeobox 1–expressing (NKX2-1+) precursor cells.
    [Show full text]
  • Effect of Testosterone and Hypoxia on the Expansion of Umbilical Cord Blood CD34+ Cells in Vitro
    EXPERIMENTAL AND THERAPEUTIC MEDICINE 14: 4467-4475, 2017 Effect of testosterone and hypoxia on the expansion of umbilical cord blood CD34+ cells in vitro LIPING ZHOU1,2, XIAOWEI ZHANG2, PANPAN ZHOU1, XUE LI1, XUEJING XU1, QING SHI1, DONG LI1 and XIULI JU1 1Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, Shandong 250012; 2Department of Pediatrics, The Sixth People's Hospital of Jinan, Jinan, Shandong 250200, P.R. China Received October 12, 2016; Accepted June 15, 2017 DOI: 10.3892/etm.2017.5026 Abstract. Successfully expanding hematopoietic stem cells Therefore, the results of the current study indicate that a combina- (HSCs) is advantageous for clinical HSC transplantation. The tion of hypoxia and testosterone may be a promising cultivation present study investigated the influence of testosterone on the condition for HSC/hemopoietic progenitor cell expansion ex vivo. proliferation, antigen phenotype and expression of hemato- poiesis-related genes in umbilical cord blood-derived cluster Introduction of differentiation (CD)34+ cells under normoxic or hypoxia conditions. Cord blood (CB) CD34+ cells were separated using Hematopoietic stem cell (HSC) transplantation is a poten- magnetic activated cell sorting. A cytokine cocktail and feeder tially life-saving procedure used to treat a broad spectrum cells were used to stimulate the expansion of CD34+ cells under of disorders, including hematological, immune and genetic normoxic (20% O2) and hypoxic (1% O2) conditions for 7 days diseases (1). It has been demonstrated that bone marrow and testosterone was added accordingly. Cells were identified reconstituting HSCs reside within a small subpopulation using flow cytometry and reconstruction capacity was deter- of bone marrow or blood-derived mononuclear cells that mined using a colony-forming unit (CFU) assay.
    [Show full text]
  • Homeobox Gene Expression Profile in Human Hematopoietic Multipotent
    Leukemia (2003) 17, 1157–1163 & 2003 Nature Publishing Group All rights reserved 0887-6924/03 $25.00 www.nature.com/leu Homeobox gene expression profile in human hematopoietic multipotent stem cells and T-cell progenitors: implications for human T-cell development T Taghon1, K Thys1, M De Smedt1, F Weerkamp2, FJT Staal2, J Plum1 and G Leclercq1 1Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent, Belgium; and 2Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands Class I homeobox (HOX) genes comprise a large family of implicated in this transformation proces.14 The HOX-C locus transcription factors that have been implicated in normal and has been primarily implicated in lymphomas.15 malignant hematopoiesis. However, data on their expression or function during T-cell development is limited. Using degener- Hematopoietic cells are derived from stem cells that reside in ated RT-PCR and Affymetrix microarray analysis, we analyzed fetal liver (FL) in the embryo and in the adult bone marrow the expression pattern of this gene family in human multipotent (ABM), which have the unique ability to self-renew and thereby stem cells from fetal liver (FL) and adult bone marrow (ABM), provide a life-long supply of blood cells. T lymphocytes are a and in T-cell progenitors from child thymus. We show that FL specific type of hematopoietic cells that play a major role in the and ABM stem cells are similar in terms of HOX gene immune system. They develop through a well-defined order of expression, but significant differences were observed between differentiation steps in the thymus.16 Several transcription these two cell types and child thymocytes.
    [Show full text]
  • Genome-Wide DNA Methylation Profiling Identifies Differential Methylation in Uninvolved Psoriatic Epidermis
    Genome-Wide DNA Methylation Profiling Identifies Differential Methylation in Uninvolved Psoriatic Epidermis Deepti Verma, Anna-Karin Ekman, Cecilia Bivik Eding and Charlotta Enerbäck The self-archived postprint version of this journal article is available at Linköping University Institutional Repository (DiVA): http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-147791 N.B.: When citing this work, cite the original publication. Verma, D., Ekman, A., Bivik Eding, C., Enerbäck, C., (2018), Genome-Wide DNA Methylation Profiling Identifies Differential Methylation in Uninvolved Psoriatic Epidermis, Journal of Investigative Dermatology, 138(5), 1088-1093. https://doi.org/10.1016/j.jid.2017.11.036 Original publication available at: https://doi.org/10.1016/j.jid.2017.11.036 Copyright: Elsevier http://www.elsevier.com/ Genome-Wide DNA Methylation Profiling Identifies Differential Methylation in Uninvolved Psoriatic Epidermis Deepti Verma*a, Anna-Karin Ekman*a, Cecilia Bivik Edinga and Charlotta Enerbäcka *Authors contributed equally aIngrid Asp Psoriasis Research Center, Department of Clinical and Experimental Medicine, Division of Dermatology, Linköping University, Linköping, Sweden Corresponding author: Charlotta Enerbäck Ingrid Asp Psoriasis Research Center, Department of Clinical and Experimental Medicine, Linköping University SE-581 85 Linköping, Sweden Phone: +46 10 103 7429 E-mail: [email protected] Short title Differential methylation in psoriasis Abbreviations CGI, CpG island; DMS, differentially methylated site; RRBS, reduced representation bisulphite sequencing Keywords (max 6) psoriasis, epidermis, methylation, Wnt, susceptibility, expression 1 ABSTRACT Psoriasis is a chronic inflammatory skin disease with both local and systemic components. Genome-wide approaches have identified more than 60 psoriasis-susceptibility loci, but genes are estimated to explain only one third of the heritability in psoriasis, suggesting additional, yet unidentified, sources of heritability.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • CREB-Binding Protein Regulates Ku70 Acetylation in Response to Ionization Radiation in Neuroblastoma
    Published OnlineFirst December 5, 2012; DOI: 10.1158/1541-7786.MCR-12-0065 Molecular Cancer DNA Damage and Repair Research CREB-Binding Protein Regulates Ku70 Acetylation in Response to Ionization Radiation in Neuroblastoma Chitra Subramanian1, Manila Hada2,3, Anthony W. Opipari Jr2, Valerie P. Castle1, and Roland P.S. Kwok2,3 Abstract Ku70 was originally described as an autoantigen, but it also functions as a DNA repair protein in the nucleus and as an antiapoptotic protein by binding to Bax in the cytoplasm, blocking Bax-mediated cell death. In neuroblastoma (NB) cells, Ku70's binding with Bax is regulated by Ku70 acetylation such that increasing Ku70 acetylation results in Bax release, triggering cell death. Although regulating cytoplasmic Ku70 acetylation is important for cell survival, the role of nuclear Ku70 acetylation in DNA repair is unclear. Here, we showed that Ku70 acetylation in the nucleus is regulated by the CREB-binding protein (CBP), and that Ku70 acetylation plays an important role in DNA repair in NB cells. We treated NB cells with ionization radiation and measured DNA repair activity as well as Ku70 acetylation status. Cytoplasmic and nuclear Ku70 were acetylated after ionization radiation in NB cells. Interestingly, cytoplasmic Ku70 was redistributed to the nucleus following irradiation. Depleting CBP in NB cells results in reducing Ku70 acetylation and enhancing DNA repair activity in NB cells, suggesting nuclear Ku70 acetylation may have an inhibitory role in DNA repair. These results provide support for the hypothesis that enhancing Ku70 acetylation, through deacetylase inhibition, may potentiate the effect of ionization radiation in NB cells.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Ku80 Antibody A
    Revision 1 C 0 2 - t Ku80 Antibody a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 3 5 Web: [email protected] 7 www.cellsignal.com 2 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source: UniProt ID: Entrez-Gene Id: WB, IP, IHC-P, IF-IC, F H Mk Endogenous 86 Rabbit P13010 7520 Product Usage Information cell cycle regulation, DNA replication and repair, telomere maintenance, recombination, and transcriptional activation. Application Dilution 1. Tuteja, R. and Tuteja, N. (2000) Crit. Rev. Biochem. Mol. Biol. 35, 1-33. 2. Blier, P.R. et al. (1993) J. Biol. Chem. 268, 7594-7601. Western Blotting 1:1000 3. Jin, S. and Weaver, D.T. (1997) EMBO J. 16, 6874-6885. Immunoprecipitation 1:25 4. Boulton, S.J. and Jackson, S.P. (1998) EMBO J. 17, 1819-1828. 5. Gravel, S. et al. (1998) Science 280, 741-744. Immunohistochemistry (Paraffin) 1:150 - 1:600 6. Cao, Q.P. et al. (1994) Biochemistry 33, 8548-8557. Immunofluorescence (Immunocytochemistry) 1:100 - 1:400 7. Lees-Miller, S.P. et al. (1990) Mol. Cell Biol. 10, 6472-6481. Flow Cytometry 1:50 - 1:100 8. Collis, S.J. et al. (2005) Oncogene 24, 949-961. Storage Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA and 50% glycerol. Store at –20°C. Do not aliquot the antibody. Specificity / Sensitivity Ku80 antibody detects endogenous levels of total Ku80 protein.
    [Show full text]
  • A Werner Syndrome Protein Homolog Affects C. Elegans Development
    Research article 2565 A Werner syndrome protein homolog affects C. elegans development, growth rate, life span and sensitivity to DNA damage by acting at a DNA damage checkpoint Se-Jin Lee, Jong-Sung Yook, Sung Min Han and Hyeon-Sook Koo* Department of Biochemistry, College of Science, Yonsei University, Seoul 120-749, Korea *Author for correspondence (e-mail: [email protected]) Accepted 18 February 2004 Development 131, 2565-2575 Published by The Company of Biologists 2004 doi:10.1242/dev.01136 Summary A Werner syndrome protein homolog in C. elegans (WRN- irrespective of γ-irradiation, and pre-meiotic germ cells had 1) was immunolocalized to the nuclei of germ cells, an abnormal checkpoint response to DNA replication embryonic cells, and many other cells of larval and adult blockage. These observations suggest that WRN-1 acts as worms. When wrn-1 expression was inhibited by RNA a checkpoint protein for DNA damage and replication interference (RNAi), a slight reduction in C. elegans life blockage. This idea is also supported by an accelerated S span was observed, with accompanying signs of premature phase in wrn-1(RNAi) embryonic cells. wrn-1(RNAi) aging, such as earlier accumulation of lipofuscin and phenotypes similar to those of Werner syndrome, such as tissue deterioration in the head. In addition, various premature aging and short stature, suggest wrn-1-deficient developmental defects, including small, dumpy, ruptured, C. elegans as a useful model organism for Werner transparent body, growth arrest and bag of worms, were syndrome. induced by RNAi. The frequency of these defects was accentuated by γ-irradiation, implying that they were derived from spontaneous or induced DNA damage.
    [Show full text]