Targeted Mass Spectrometry Enables Quantification of Novel

Total Page:16

File Type:pdf, Size:1020Kb

Targeted Mass Spectrometry Enables Quantification of Novel cancers Article Targeted Mass Spectrometry Enables Quantification of Novel Pharmacodynamic Biomarkers of ATM Kinase Inhibition Jeffrey R. Whiteaker 1 , Tao Wang 1, Lei Zhao 1, Regine M. Schoenherr 1, Jacob J. Kennedy 1, Ulianna Voytovich 1, Richard G. Ivey 1, Dongqing Huang 1, Chenwei Lin 1, Simona Colantonio 2, Tessa W. Caceres 2, Rhonda R. Roberts 2, Joseph G. Knotts 2, Jan A. Kaczmarczyk 2, Josip Blonder 2, Joshua J. Reading 2, Christopher W. Richardson 2, Stephen M. Hewitt 3, Sandra S. Garcia-Buntley 2, William Bocik 2, Tara Hiltke 4, Henry Rodriguez 4, Elizabeth A. Harrington 5, J. Carl Barrett 5, Benedetta Lombardi 5, Paola Marco-Casanova 5, Andrew J. Pierce 5 and Amanda G. Paulovich 1,* 1 Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA 98109, USA; [email protected] (J.R.W.); [email protected] (T.W.); [email protected] (L.Z.); [email protected] (R.M.S.); [email protected] (J.J.K.); [email protected] (U.V.); [email protected] (R.G.I.); [email protected] (D.H.); [email protected] (C.L.) 2 Cancer Research Technology Program, Antibody Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA; [email protected] (S.C.); [email protected] (T.W.C.); [email protected] (R.R.R.); [email protected] (J.G.K.); [email protected] (J.A.K.); [email protected] (J.B.); [email protected] (J.J.R.); [email protected] (C.W.R.); [email protected] (S.S.G.-B.); [email protected] (W.B.) 3 Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Citation: Whiteaker, J.R.; Wang, T.; Institute, National Institute of Health, Bethesda, MD 20892, USA; [email protected] Zhao, L.; Schoenherr, R.M.; Kennedy, 4 Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA; J.J.; Voytovich, U.; Ivey, R.G.; Huang, [email protected] (T.H.); [email protected] (H.R.) D.; Lin, C.; Colantonio, S.; et al. 5 Translational Sciences, Oncology, AstraZeneca, Cambridge CB4 0WG, UK; Targeted Mass Spectrometry Enables [email protected] (E.A.H.); [email protected] (J.C.B.); Quantification of Novel [email protected] (B.L.); [email protected] (P.M.-C.); Pharmacodynamic Biomarkers of [email protected] (A.J.P.) * Correspondence: [email protected]; Tel.: +1-206-667-1912 ATM Kinase Inhibition. Cancers 2021, 13, 3843. https://doi.org/10.3390/ cancers13153843 Simple Summary: Functionality of the cellular DNA damage response (DDR) network affects risk for developing cancer, and the DDR is also a target of cancer therapies. Thus, it is important that we have Academic Editors: Udayan Guha and reliable laboratory methods for determining the activity of this network. Wedescribe the development and Xu Zhang analytical validation of a targeted mass spectrometry-based 51-plex assay (DDR-2) for measuring proteins and post-translational modifications related to the DDR. The findings demonstrate identification of Received: 18 June 2021 potential novel pharmacodynamic biomarkers. Accepted: 23 July 2021 Published: 30 July 2021 Abstract: The ATM serine/threonine kinase (HGNC: ATM) is involved in initiation of repair of DNA double-stranded breaks, and ATM inhibitors are currently being tested as anti-cancer agents in Publisher’s Note: MDPI stays neutral clinical trials, where pharmacodynamic (PD) assays are crucial to help guide dose and scheduling with regard to jurisdictional claims in and support mechanism of action studies. To identify and quantify PD biomarkers of ATM inhibition, published maps and institutional affil- we developed and analytically validated a 51-plex assay (DDR-2) quantifying protein expression and iations. DNA damage-responsive phosphorylation. The median lower limit of quantification was 1.28 fmol, the linear range was over 3 orders of magnitude, the median inter-assay variability was 11% CV, and 86% of peptides were stable for storage prior to analysis. Use of the assay was demonstrated to quantify signaling following ionizing radiation-induced DNA damage in both immortalized Copyright: © 2021 by the authors. lymphoblast cell lines and primary human peripheral blood mononuclear cells, identifying PD Licensee MDPI, Basel, Switzerland. This article is an open access article biomarkers for ATM inhibition to support preclinical and clinical studies. distributed under the terms and conditions of the Creative Commons Keywords: multiple reaction monitoring; DNA damage response; immuno-MRM; targeted proteomics Attribution (CC BY) license (https:// creativecommons.org/licenses/by/ 4.0/). Cancers 2021, 13, 3843. https://doi.org/10.3390/cancers13153843 https://www.mdpi.com/journal/cancers Cancers 2021, 13, 3843 2 of 25 1. Introduction The DNA damage response (DDR) is a highly regulated signal transduction network that orchestrates the temporal and spatial organization of protein complexes required to detect and repair (or tolerate) DNA damage (e.g., nucleotide excision repair, base excision repair, homologous recombination, non-homologous end joining, post-replication repair) [1]. The DDR is of critical significance in oncology, as genetic instability caused by dysregulation of the DDR is a key feature of cancer cells [2], and chemotherapy and radiation therapy induce DNA damage to promote cell death. Thus, components of the DDR network are potential therapeutic targets, and being able to quantify and understand activity in the DDR network has clinical implications for cancer patients and those at risk [3]. The Ataxia-telangiectasia-mutated (ATM) gene encodes a serine/threonine protein kinase that initiates the DDR to DNA double-stranded breaks (DSB). ATM is activated through dissociation of the ATM homodimer to active monomers via autophosphorylation at several sites [4], including Ser1981 [5], Ser367 [6], and Ser2996 [7]. Activated ATM phosphorylates downstream components to mediate DNA repair and cell cycle regulation. Cells lacking ATM, as seen in patients with ataxia telangiectasia, are sensitive to DNA damage [8]. Three ATM inhibitors developed by KuDOS (Cambridge, UK) and AstraZeneca (Cambridge, UK), KU-55933, AZD0156, AD1390, display potent and exquisite inhibition of ATM with no meaningful capacity to inhibit other phosphatidylinositol 3-kinase-related kinases (PIKKs), such as ATR and DNA-PK [9–11]. AZD0156 is a particularly potent inhibitor of ATM and is in clinical trials (NCT02588105) in combination with DNA damage- inducing agents, where it is expected to enhance cell death in tumor cells by reducing their capacity to resolve DNA damage. Quantifying proteins and protein networks for pharmacokinetic (PK), pharmaco- dynamic (PD), and proof of mechanism (POM) studies is critical for translating novel therapies, such as ATM inhibitors. Indeed, AstraZeneca recently revised its drug devel- opment framework [12,13] with a view to increasing success rates of drugs in pivotal Phase 3 studies. Key components are POM studies and determining PK/PD relation- ships in early phase clinical trials. Conventional immunoassay platforms (e.g., ELISA, immunohistochemistry) are generally used for these studies; however, these platforms are critically dependent on the absolute specificity of individual antibodies, and establishing this specificity is costly in terms of time and resource. As a result, only a handful of fully validated assays can be developed for each drug project, where the choice of which assays to develop is largely done on the basis of “best educated guess” arising from orthogonal preclinical methods such as Western blotting. Additionally, in many cases an assay for a given target cannot be quickly developed because there are no fit-for-purpose antibodies available. In such cases a bespoke reagent antibody generation campaign may be initiated, adding to the time and cost of drug development. Adding to the issue, proteins act as interconnected “networks”, and the effects of cancer driver mutations spread throughout the network; thus, ideally we would have assays to quantify panels of multiple proteins in early phase clinical trials to assess the activity of pathways/networks that determine treatment responses [14], and this developmental effort is not practical using conventional platforms. Thus, new tools are needed for quantifying proteins and post-translational modifications to aid discovery of new pharmacodynamic markers, enhance mechanistic studies, and translate research findings to clinical implementation. To address this need, we applied targeted multiple reaction monitoring (MRM) mass spectrometry (MS) to develop quantitative assays to proteins and post-translational modifi- cations [15,16]. MRM is a targeted mode of MS in which the full analytical capacity of the instrument is focused on a set of specific analytes, maintaining a high degree of specificity and enhancing the sensitivity of detection [17,18]. By using spiked-in isotopically labeled internal standards, precise relative quantification is achieved, and results can be harmo- nized across laboratories [19,20]. To analyze low abundance proteins and post-translation modifications, including phosphorylation, we coupled an antibody enrichment step to MRM [21]. These “immuno-MRM” assays enable precise quantification of low abundance Cancers 2021, 13, 3843 3 of 25 proteins [22], high multiplexing capability while maintaining specificity for individual analytes [23,24], and
Recommended publications
  • Deregulated Gene Expression Pathways in Myelodysplastic Syndrome Hematopoietic Stem Cells
    Leukemia (2010) 24, 756–764 & 2010 Macmillan Publishers Limited All rights reserved 0887-6924/10 $32.00 www.nature.com/leu ORIGINAL ARTICLE Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells A Pellagatti1, M Cazzola2, A Giagounidis3, J Perry1, L Malcovati2, MG Della Porta2,MJa¨dersten4, S Killick5, A Verma6, CJ Norbury7, E Hellstro¨m-Lindberg4, JS Wainscoat1 and J Boultwood1 1LRF Molecular Haematology Unit, NDCLS, John Radcliffe Hospital, Oxford, UK; 2Department of Hematology Oncology, University of Pavia Medical School, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; 3Medizinische Klinik II, St Johannes Hospital, Duisburg, Germany; 4Division of Hematology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; 5Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK; 6Albert Einstein College of Medicine, Bronx, NY, USA and 7Sir William Dunn School of Pathology, University of Oxford, Oxford, UK To gain insight into the molecular pathogenesis of the the World Health Organization.6,7 Patients with refractory myelodysplastic syndromes (MDS), we performed global gene anemia (RA) with or without ringed sideroblasts, according to expression profiling and pathway analysis on the hemato- poietic stem cells (HSC) of 183 MDS patients as compared with the the French–American–British classification, were subdivided HSC of 17 healthy controls. The most significantly deregulated based on the presence or absence of multilineage dysplasia. In pathways in MDS include interferon signaling, thrombopoietin addition, patients with RA with excess blasts (RAEB) were signaling and the Wnt pathways. Among the most signifi- subdivided into two categories, RAEB1 and RAEB2, based on the cantly deregulated gene pathways in early MDS are immuno- percentage of bone marrow blasts.
    [Show full text]
  • CPTC-TP53BP1-1 (CAB079980) Immunohistochemistry Immunofluorescence
    CPTC-TP53BP1-1 (CAB079980) Uniprot ID: Q12888 Protein name: TP53B_HUMAN Full name: TP53-binding protein 1 Function: Double-strand break (DSB) repair protein involved in response to DNA damage, telomere dynamics and class-switch recombination (CSR) during antibody genesis (PubMed:12364621, PubMed:22553214, PubMed:23333306, PubMed:17190600, PubMed:21144835, PubMed:28241136). Plays a key role in the repair of double-strand DNA breaks (DSBs) in response to DNA damage by promoting non-homologous end joining (NHEJ)- mediated repair of DSBs and specifically counteracting the function of the homologous recombination (HR) repair protein BRCA1 (PubMed:22553214, PubMed:23727112, PubMed:23333306). In response to DSBs, phosphorylation by ATM promotes interaction with RIF1 and dissociation from NUDT16L1/TIRR, leading to recruitment to DSBs sites (PubMed:28241136). Recruited to DSBs sites by recognizing and binding histone H2A monoubiquitinated at 'Lys-15' (H2AK15Ub) and histone H4 dimethylated at 'Lys-20' (H4K20me2), two histone marks that are present at DSBs sites (PubMed:23760478, PubMed:28241136, PubMed:17190600). Required for immunoglobulin class-switch recombination (CSR) during antibody genesis, a process that involves the generation of DNA DSBs (PubMed:23345425). Participates in the repair and the orientation of the broken DNA ends during CSR (By similarity). In contrast, it is not required for classic NHEJ and V(D)J recombination (By similarity). Promotes NHEJ of dysfunctional telomeres via interaction with PAXIP1 (PubMed:23727112). Subcellular
    [Show full text]
  • Funkce CDK12 a CDK13 V Regulaci Transkripce Hana Paculová
    MASARYKOVA UNIVERZITA PŘÍRODOVĚDECKÁ FAKULTA ÚSTAV BIOCHEMIE Funkce CDK12 a CDK13 v regulaci transkripce Disertační práce Hana Paculová Školitel: Mgr. Jiří Kohoutek, Ph.D Brno 2018 Bibliogra cký záznam Autorka: Mgr. Hana Paculová Prrodovedecáá aaául,a鏈 Maaarkáova unvverv,a Úa,av bvochemve Název práce: Funáce CDK12 a CDK13 v regulacv ,ranaárvpce Studijní program: Bvochemve Studijní obor: Bvochemve Školitel: Mgr. Jvr Kohou,eá鏈 Ph.D Akademický rok: 2017/2018 Po et stran: 89 Klí ová slova: Ckálvn-dependen,n ávnaaa鏈 CDK12鏈 ,ranaárvpce鏈 RNA polkmeraaa II鏈 raáovvna vaječnáů鏈 CHK1 Bibliographic entry Author: Mgr. Hana Paculová Facul,k oa acvence鏈 Maaarká unvverav,k Department of Biochemistry Title oF dissertation: CDK12 and CDK13 aunc,von vn ,ranacrvp,von regula,von Degree programme: Bvochemva,rk Field oF study: Bvochemva,rk Supervisor: Mgr. Jvr Kohou,eá鏈 Ph.D Academic year: 2017/2018 Number oF pages: 89 Keywords: Ckclvn-dependen ávnaae鏈 CDK12鏈 ,ranacrvp,von鏈 RNA polkmeraae II鏈 ovarvan cancer鏈 CHK1 Abstrakt Ckálvn-dependen,n ávnaaa 12 (CDK12) je ,ranaárvpčn ávnaaa鏈 á,erá rd expreav avých clových genů ,m鏈 že aoaaorkluje RNA polkmeraau II v průbehu elongačn aáe ,ranaárvpce. CDK12 je apojena do neáolváa bunečných preceaů鏈 což ahrnuje odpoveď na pošáoen DNA鏈 vývoj a bunečnou dvaerencvacv a aea,rvh mRNA. CDK12 bkla popaána jaáo jeden genů鏈 á,eré jaou čaa,o mu,ovánk v hvgh-grade aerónm ovarválnm áarcvnomu鏈 nvcméne vlvv ,ech,o mu,ac na aunácv CDK12 a jejvch role v áarcvnogenev dopoaud nebkla a,anovena. Zjva,vlv jame鏈 že ve,švna mu,ac CDK12鏈 á,eré bklk naleenk v nádorech鏈 brán vk,voren áomplexu CDK12 a Ckálvnem K a vnhvbuj ávnaaovou aá,vvv,u CDK12.
    [Show full text]
  • RIDDLE Immunodeficiency Syndrome Is Linked to Defects in 53BP1-Mediated DNA Damage Signaling
    RIDDLE immunodeficiency syndrome is linked to defects in 53BP1-mediated DNA damage signaling Grant S. Stewart*†, Tatjana Stankovic*, Philip J. Byrd*, Thomas Wechsler‡, Edward S. Miller*, Aarn Huissoon§, Mark T. Drayson¶, Stephen C. West‡, Stephen J. Elledge†ʈ, and A. Malcolm R. Taylor* *Cancer Research UK, Institute for Cancer Studies, Birmingham University, Vincent Drive, Edgbaston, Birmingham B15 2TT, United Kingdom; ‡Cancer Research UK, Clare Hall Laboratories, London Research Institute, South Mimms, Hertfordshire EN6 3LD, United Kingdom; §Department of Immunology, Birmingham Heartlands Hospital, Birmingham, B9 5SS, United Kingdom; ¶Division of Immunity and Infection, Birmingham University Medical School, Vincent Drive, Edgbaston, Birmingham B15 2TT, United Kingdom; and ʈHoward Hughes Medical Institute, Department of Genetics, Harvard Partners Center for Genetics and Genomics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115 Contributed by Stephen J. Elledge, September 6, 2007 (sent for review July 6, 2007) Cellular DNA double-strand break-repair pathways have evolved with an alternative constant region (e.g., ␣, ␥, ␧) to generate to protect the integrity of the genome from a continual barrage of different Ig isotypes e.g., IgA, IgG, and IgE (2). potentially detrimental insults. Inherited mutations in genes that During the process of CSR, it has been hypothesized that two control this process result in an inability to properly repair DNA closely positioned single-stranded DNA nicks result in the damage, ultimately
    [Show full text]
  • Anti-EGFR Monoclonal Antibodies and EGFR Tyrosine Kinase Inhibitors As Combination Therapy for Triple-Negative Breast Cancer
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 45 Research Paper Anti-EGFR monoclonal antibodies and EGFR tyrosine kinase inhibitors as combination therapy for triple-negative breast cancer Abderrahim El Guerrab1,2, Mahchid Bamdad2,3, Fabrice Kwiatkowski1, Yves-Jean Bignon1,2,*, Frédérique Penault-Llorca1,2,*, Corinne Aubel1,2 1Centre Jean Perrin - ERTICa-EA4677, BP392, 63011 Clermont-Ferrand Cedex, France 2Clermont Université - Université d’Auvergne - ERTICa-EA4677, Faculté de Médecine, BP38, 63001 Clermont-Ferrand Cedex, France 3Clermont Université - Université d’Auvergne - ERTICa-EA4677, Institut Universitaire de Technologie, Département Génie Biologique, Ensemble Universitaire des Cézeaux, BP86, 63172 Aubière Cedex, France *These authors have contributed equally to this work Correspondence to: Yves-Jean Bignon, email: [email protected] Keywords: triple-negative breast cancer, epidermal growth factor receptor, anti-EGFR targeted therapy, cytotoxicity, cell cycle Received: November 09, 2015 Accepted: August 22, 2016 Published: September 15, 2016 ABSTRACT Triple-negative breast cancer (TNBC) is characterized by overexpression of epidermal growth factor receptor (EGFR) and activation of its downstream signaling pathways. Dual targeting of EGFR using one monoclonal antibody (mAb; cetuximab or panitumumab) and one tyrosine kinase inhibitor (EGFR-TKI; gefitinib or erlotinib) is a potential therapeutic approach. We investigated the effect of these therapies in EGFR-expressing TNBC cell lines that do or do not harbor the main activating mutations of EGFR pathways. Cell lines were sensitive to EGFR-TKIs, whereas mAbs were active only in MDA-MB-468 (EGFR amplification) and SUM-1315 (KRAS and PTEN wild-type) cells. MDA-MB-231 (KRAS mutated) and HCC-1937 (PTEN deletion) cells were resistant to mAbs.
    [Show full text]
  • WO 2019/079361 Al 25 April 2019 (25.04.2019) W 1P O PCT
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization I International Bureau (10) International Publication Number (43) International Publication Date WO 2019/079361 Al 25 April 2019 (25.04.2019) W 1P O PCT (51) International Patent Classification: CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, C12Q 1/68 (2018.01) A61P 31/18 (2006.01) DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, C12Q 1/70 (2006.01) HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KH, KN, KP, KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, (21) International Application Number: MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, PCT/US2018/056167 OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, (22) International Filing Date: SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, 16 October 2018 (16. 10.2018) TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (25) Filing Language: English (84) Designated States (unless otherwise indicated, for every kind of regional protection available): ARIPO (BW, GH, (26) Publication Language: English GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, (30) Priority Data: UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, 62/573,025 16 October 2017 (16. 10.2017) US TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, FI, FR, GB, GR, HR, HU, ΓΕ , IS, IT, LT, LU, LV, (71) Applicant: MASSACHUSETTS INSTITUTE OF MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, TECHNOLOGY [US/US]; 77 Massachusetts Avenue, TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, GW, Cambridge, Massachusetts 02139 (US).
    [Show full text]
  • PRODUCT INFORMATION TP53BP1 BRCT Domains (Human Recombinant) Item No
    PRODUCT INFORMATION TP53BP1 BRCT domains (human recombinant) Item No. 14171 • Batch No. XXXX Overview and Properties Synonyms: Tumor Protein p53 binding Protein 1, Tumor Suppressor p53-binding Protein 1 Source: Recombinant human N-terminal GST-tagged protein expressed in E. coli amino acids 1,717-1,972 (N- and C-terminal truncation) Uniprot No.: Q12888 Batch specific information can be found on the Batch Specific Insert or by contacting Technical Support Molecular Weight: 55.1 kDa Storage: -80°C (as supplied) Stability: ≥6 months Purity: batch specific Supplied in: 50 mM Tris-HCl, pH 8.0, containing 150 mM sodium chloride and 20% glycerol Protein Concentration: batch specific mg/ml Image(s) 1 2 3 4 250 kDa · · · · · · · 150 kDa · · · · · · · 100 kDa · · · · · · · 75 kDa · · · · · · · 50 kDa · · · · · · · 37 kDa · · · · · · · 25 kDa · · · · · · · 20 kDa · · · · · · · Lane 1: MW Markers Lane 2: TP53BP1 BRCT domains (1 µg) Lane 3: TP53BP1 BRCT domains (2 µg) Lane 4: TP53BP1 BRCT domains (5 µg) Representative gel image shown; actual purity may vary between each batch. WARNING CAYMAN CHEMICAL THIS PRODUCT IS FOR RESEARCH ONLY - NOT FOR HUMAN OR VETERINARY DIAGNOSTIC OR THERAPEUTIC USE. 1180 EAST ELLSWORTH RD SAFETY DATA ANN ARBOR, MI 48108 · USA This material should be considered hazardous until further information becomes available. Do not ingest, inhale, get in eyes, on skin, or on clothing. Wash thoroughly after handling. Before use, the user must review the complete Safety Data Sheet, which has been sent via email to your institution. PHONE: [800] 364-9897 WARRANTY AND LIMITATION OF REMEDY [734] 971-3335 Buyer agrees to purchase the material subject to Cayman’s Terms and Conditions.
    [Show full text]
  • Ubch7 Regulates 53BP1 Stability and DSB Repair
    UbcH7 regulates 53BP1 stability and DSB repair Xiangzi Hana,1, Lei Zhanga,1, Jinsil Chunga,1, Franklin Mayca Pozoa, Amanda Trana, Darcie D. Seachrista, James W. Jacobbergerb, Ruth A. Keria, Hannah Gilmorec, and Youwei Zhanga,2 aDepartment of Pharmacology, bDivision of General Medical Sciences, Case Comprehensive Cancer Center, School of Medicine, and cUniversity Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH 44106 Edited by Tony Hunter, The Salk Institute for Biological Studies, La Jolla, CA, and approved November 3, 2014 (received for review May 8, 2014) DNA double-strand break (DSB) repair is not only key to genome and generation of one-ended DSBs. Such one-ended DSBs re- stability but is also an important anticancer target. Through an quire HR, but not NHEJ, to repair (8). In contrast, repair of one- shRNA library-based screening, we identified ubiquitin-conjugat- ended DSBs by NHEJ leads to radial chromosomes and cell death ing enzyme H7 (UbcH7, also known as Ube2L3), a ubiquitin E2 (12–14). Therefore, stabilization of 53BP1 by UbcH7 depletion enzyme, as a critical player in DSB repair. UbcH7 regulates both increased the sensitivity of cancers cells to CPT and other DNA the steady-state and replicative stress-induced ubiquitination damaging agents. Our data suggest a novel strategy in enhancing and proteasome-dependent degradation of the tumor suppressor the anticancer effect of radiotherapy or chemotherapy through p53-binding protein 1 (53BP1). Phosphorylation of 53BP1 at the N terminus is involved in the replicative stress-induced 53BP1 degra- stabilizing or increasing 53BP1. dation. Depletion of UbcH7 stabilizes 53BP1, leading to inhibition Results of DSB end resection.
    [Show full text]
  • Tumor Suppressor and DNA Damage Response Panel 2
    Tumor suppressor and DNA damage response panel 2 (55 analytes) Gene Symbol Target protein name UniProt ID (& link) Modification* *blanks mean the assay detects the ACT Actin; ACTA2 ACTA1 ACTB ACTG1 ACTC1 ACTG2 Q562L2 non‐modified peptide sequence CASC5 cancer susceptibility candidate 5 Q8NG31 pS767 CASC5 cancer susceptibility candidate 5 Q8NG31 CASP3 caspase 3, apoptosis‐related cysteine peptidase P42574 pS26 CDC25B cell division cycle 25 homolog B (S. pombe) P30305 pS16 CDC25B cell division cycle 25 homolog B (S. pombe) P30305 pS323 CDC25B cell division cycle 25 homolog B (S. pombe) P30305 CDC25C cell division cycle 25 homolog B (S. pombe) P30307 pS216 CDC25C cell division cycle 25 homolog B (S. pombe) P30307 CDCA8 cell division cycle associated 8 Q53HL2 pT16 CDK1 cyclin‐dependent kinase 1 P06493 pT161 CDK1 cyclin‐dependent kinase 1 P06493 CDK7 cyclin‐dependent kinase 7 P50613 pT17 CHEK1 CHK1 checkpoint homolog (S. pombe) O14757 pS286 CHEK2 CHK2 checkpoint homolog (S. pombe) O96017 pS379 CHEK2 CHK2 checkpoint homolog (S. pombe) O96017 pT387 CHEK2 CHK2 checkpoint homolog (S. pombe) O96017 GAPDH Glyceraldehyde‐3‐phosphate dehydrogenase P04406 LAT linker for activation of T cells O43561 pS224 LMNB1 lamin B1 P20700 pT2; pS23 LMNB1 lamin B1 P20700 pT2 LMNB1 lamin B1 P20700 pS23 MCM6 minichromosome maintenance complex component 6 Q14566 pS762 MCM6 minichromosome maintenance complex component 6 Q14566 MDM2 Mdm2, transformed 3T3 cell double minute 2, p53 binding protein (mouse) Q00987 pS166 MDM2 Mdm2, transformed 3T3 cell double minute 2, p53 binding protein (mouse) Q00987 MKI67 antigen identified by monoclonal antibody Ki‐67 P46013 pT181 MKI67 antigen identified by monoclonal antibody Ki‐67 P46013 MKI67 antigen identified by monoclonal antibody Ki‐67 P46013 pT246 MRE11A MRE11 meiotic recombination 11 homolog A (S.
    [Show full text]
  • The Biological Effects of Simple Tandem Repeats: Lessons from the Repeat Expansion Diseases
    Downloaded from genome.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press Review The biological effects of simple tandem repeats: Lessons from the repeat expansion diseases Karen Usdin1 Section on Gene Structure and Disease, Laboratory of Molecular and Cellular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0830, USA Tandem repeats are common features of both prokaryote and eukaryote genomes, where they can be found not only in intergenic regions but also in both the noncoding and coding regions of a variety of different genes. The repeat expansion diseases are a group of human genetic disorders caused by long and highly polymorphic tandem repeats. These disorders provide many examples of the effects that such repeats can have on many biological processes. While repeats in the coding sequence can result in the generation of toxic or malfunctioning proteins, noncoding repeats can also have significant effects including the generation of chromosome fragility, the silencing of the genes in which they are located, the modulation of transcription and translation, and the sequestering of proteins involved in processes such as splicing and cell architecture. Tandem repeats are a ubiquitous feature of the genomes of many also known that involve an expanded amino acid tract generated organisms, and the human genome database is replete with ex- by insertion or duplication of an imperfect trinucleotide repeat. amples of repeat tracts of different sequence and repeat number These diseases differ in key ways from “classic” members of the in different genomic locations (Riggins et al.
    [Show full text]
  • Comprehensive Assessment of Indian Variations in the Druggable Kinome Landscape Highlights Distinct Insights at the Sequence, Structure and Pharmacogenomic Stratum
    SUPPLEMENTARY MATERIAL Comprehensive assessment of Indian variations in the druggable kinome landscape highlights distinct insights at the sequence, structure and pharmacogenomic stratum Gayatri Panda1‡, Neha Mishra1‡, Disha Sharma2,3, Rahul C. Bhoyar3, Abhinav Jain2,3, Mohamed Imran2,3, Vigneshwar Senthilvel2,3, Mohit Kumar Divakar2,3, Anushree Mishra3, Priyanka Banerjee4, Sridhar Sivasubbu2,3, Vinod Scaria2,3, Arjun Ray1* 1 Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla, India. 2 Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India. 3 CSIR-Institute of Genomics and Integrative Biology, Mathura Road, Delhi-110020, India. 4 Institute for Physiology, Charite-University of Medicine, Berlin, 10115 Berlin, Germany. ‡These authors contributed equally to this work. * [email protected] TABLE OF CONTENTS Name Title Supplemental_Figure_S1 Fauchere and Pliska hydrophobicity scale for variations in structure data Supplemental_Figure_S2 Phenotypic drug-drug correlogram Supplemental_Table_S1 545 kinase coding genes used in the study Supplemental_Table_S2 Classes and count of kinase coding genes Supplemental_Table_S3 Allele frequency Indian v/s other populations from 1000 genome data(1000g2015). Supplemental_Table_S4 IndiGen Structure Data- consisting of 12 genes and their 22 variants Supplemental_Table_S5 Genes, PDB ids, mutations in IndiGen data and associated drugs (FDA approved) Supplemental_Table_S6 Data used for docking and binding pocket similarity analysis Supplemental_Table_S7
    [Show full text]
  • Small-Molecule Binding Sites to Explore New Targets in the Cancer Proteome
    Electronic Supplementary Material (ESI) for Molecular BioSystems. This journal is © The Royal Society of Chemistry 2016 Small-molecule binding sites to explore new targets in the cancer proteome David Xu, Shadia I. Jalal, George W. Sledge Jr., and Samy O. Meroueh* Supplementary Text Druggable Binding Sites across all 10 Diseases. Using the previously established cutoffs, we identified genes that were overexpressed across multiple cancer types and featured druggable binding sites. We ranked these genes based on the total number of tumors that overexpressed the gene (Fig. S1). Using a simple PubMed query, we then counted the number of articles in which either the gene symbol or gene name was co-mentioned with the term ‘cancer’. Most of the most frequently occurring differentially-expressed genes correspond to proteins of well- established cancer targets. Among them are matrix metalloproteinases (MMPs), including MMP1, MMP9, and MMP12, which are implicated in tumor invasion and metastasis (1). There are several protein kinases, including TTK, AURKA, AURKB, and PLK1, that are involved in cell signaling and well-established oncology targets (2). Some genes among this list that have not been extensively studied nor targeted in cancer. These include the serine/threonine kinase PKMYT1 (MYT1) is a regulator of G2/M transition in the cell cycle, but lacks focused small molecule inhibitors that specifically target the kinase. Recent efforts in developing small molecule inhibitors involve repurposing of available kinase inhibitors to specifically target the kinase (3). A subunit of the GINS complex GINS2 (PSF2) is involved in cell proliferation and survival in cancer cell lines (4,5).
    [Show full text]