Selective Inhibition of Human Tankyrases | 2014 230226 789521 9 ISBN 978-952-12-3022-6 Mohit Narwal

Total Page:16

File Type:pdf, Size:1020Kb

Selective Inhibition of Human Tankyrases | 2014 230226 789521 9 ISBN 978-952-12-3022-6 Mohit Narwal Mohit Narwal | Selective Inhibition of Human Tankyrases | 2014 Mohit NarwalTankyrases | Selective Inhibition of Human Mohit Narwal Selective Inhibition of Human Tankyrases Pharmaceutical Sciences Department of Biosciences Åbo Akademi University Turku, Finland 9 789521 230226 2014 ISBN 978-952-12-3022-6 Mohit Narwal Mohit Narwal was born on January 20, 1984 in Nahan, H.P., India. He did his Bachelor and Master of Science in Biotechnology from Panjab University and IIT Roorkee, respectively in India. This PhD thesis work has taken place under the supervision of Dr. Lari Lehtiö at Phar- maceutical Sciences, Åbo Akademi University and Faculty of Biochemistry and Molecular Medicine, University of Oulu during 2009-2014. Åbo Akademi University Press Tavastgatan 13, FI-20500 Åbo, Finland Tel. +358 (0)2 215 3478 E-mail: [email protected] Sales and distribution: Åbo Akademi University Library Domkyrkogatan 2–4, FI-20500 Åbo, Finland Tel. +358 (0)2 -215 4190 E-mail: [email protected] Selective Inhibition of Human Tankyrases Mohit Narwal Pharmaceutical Sciences Department of Biosciences Åbo Akademi University Turku, Finland 2014 Supervisor Dr. Lari Lehtiö Docent Biocenter Oulu Faculty of Biochemistry and Molecular Medicine University of Oulu Finland Reviewer Dr. Tommi Kajander Docent, Institute of Biotechnology University of Helsinki Finland And Dr. Jens Preben Morth Center for Molecular Medicine University of Oslo Norway Opponent Dr. Herwig Schüler PI Structural Biochemistry Department of Medical Biochemistry and Biophysics Karolinska Institutet Sweden Cover: Crystal structure of tankyrase 2 in complex with IWR-1 My photo: Picture by Bhargav Prabhakar ISBN 978-952-12-3022-6 Painosalama Oy – Turku, Finland 2014 Table of Contents ORIGINAL PUBLICATIONS ............................................................................... I CONTRIBUTIONS OF THE AUTHOR ............................................................... II ACKNOWLEDGEMENTS .................................................................................. III ABSTRACT......................................................................................................... VI ABBREVIATIONS ........................................................................................... VIII 1 REVIEW OF THE LITERATURE ................................................................... 1 1.1 Introduction ................................................................................................ 1 1.1.1 ARTD activation, poly(ADP-ribosyl)ation reaction and ARTD catabolism ............................................................................. 2 1.2 ARTDs and their functions ......................................................................... 3 1.2.1 ARTD1 ............................................................................................. 3 1.2.2 ARTD2 ............................................................................................. 4 1.2.3 ARTD3 ............................................................................................. 5 1.2.4 ARTD4 ............................................................................................. 5 1.2.5 Mono (ADP-ribosyl) transferases .................................................... 6 1.3 Tankyrases .................................................................................................. 6 1.3.1 Crystal Structure ............................................................................... 7 1.3.2 Tankyrase modifications .................................................................. 8 1.3.3 The SAM domain ............................................................................. 9 1.3.4 The ankyrin repeats and the target specificity .................................. 9 1.4 Functions of tankyrases ............................................................................ 10 1.4.1 Telomere maintenance ................................................................... 11 1.4.2 Role in Wnt signaling pathway ...................................................... 13 1.4.3 GLUT4 vesicle translocation ......................................................... 14 1.4.4 Mitosis ............................................................................................ 15 1.4.5 Proteasome regulation .................................................................... 16 1.5 Role of tankyrases in cancer ..................................................................... 17 1.5.1 Role of tankyrases in viral infections ............................................. 18 1.5.2 Role of tankyrases in obesity and other diseases ........................... 18 1.6 Drug development and available ARTD inhibitors .................................. 19 1.6.1 Properties of a drug candidate and screening strategy ................... 19 1.6.2 Pharmacodynamics and pharmacokinetics ..................................... 20 1.6.3 Clinical phases ............................................................................... 20 1.6.4 PARP inhibitors and selective tankyrase inhibitors ....................... 21 1.6.5 Tankyrase inhibitors binding to the nicotinamide site ................... 21 1.6.6 Adenosine site and dual-site binding inhibitors ............................. 22 2 AIMS OF THE STUDY .................................................................................. 24 3 MATERIALS AND METHODS .................................................................... 25 3.1 Expression vectors (I-V) ........................................................................... 25 3.2 Protein expression and purification (I-V) ................................................. 25 3.3 Activity assay (I-V) .................................................................................. 26 3.4 Optimization of assay conditions and assay repeatability (I, III) ............. 26 3.5 Screening and potency measurements (I, III, IV, V) ................................ 27 3.6 Western blot method (I, II) ....................................................................... 27 3.7 Crystallization and crystallography (II-V) ............................................... 28 4 RESULTS ........................................................................................................ 29 4.1 Adaptation of an assay for screening the compounds .............................. 29 4.1.1 Protein expression and purification (I-V) ....................................... 29 4.1.2 Assay optimization (I, III) .............................................................. 29 4.1.3 Assay validation (I, III) .................................................................. 30 4.1.4 Validatory Screening and potency measurements (I) ..................... 30 4.2 Identifications of flavones as potent and selective tankyrase inhibitors (III, IV) ..................................................................................... 31 4.2.1 Screening of the flavonoids library and binding mode of flavones (III) ................................................................................... 31 4.2.2 Screening of the flavones with single substitutions and co- crystal structures (IV) ..................................................................... 32 4.2.3 Inhibition of Wnt signaling and profiling of the inhibitors (IV) ................................................................................................. 37 4.3 Inhibitor binding to adenosine site (II) ..................................................... 38 4.3.1 Co-crystal structure of TNKS2 catalytic domain in complex with IWR-1 ..................................................................................... 38 4.3.2 Structure-activity relationship studies of IWR-1 analogues ........... 38 4.3.3 IWR-1 selectivity ........................................................................... 39 4.4 Characterization of known ARTD inhibitors (V) ..................................... 40 4.4.1 Screening of the inhibitors and potency measurements ................. 40 4.4.2 Binding modes of the compounds .................................................. 41 4.4.2.1 Binding modes of Phenanthridinone (16) and TIQ- A (18) .............................................................................. 42 4.4.2.2 Binding mode of PJ-34 (17) ............................................ 42 4.4.2.3 Binding mode of Rucaparib (29) .................................... 42 4.5 Substrate Binding (II, V) .......................................................................... 43 4.5.1 Co-crystal structure of TNKS2 catalytic domain in complex with nicotinamide (II)..................................................................... 43 4.5.2 Co-crystal structure of TNKS2 catalytic domain in complex with EB-47 (V) ............................................................................... 43 5 DISCUSSION .................................................................................................. 46 5.1 The screening assay (I, III, IV) ................................................................. 46 5.2 Flavones as tankyrase inhibitors (III, IV) ................................................. 47 5.3 Novel binding mode of IWR-1 (II)........................................................... 50 5.4 Inhibition of tankyrases by ARTD inhibitors (V)..................................... 50 5.5 Substrate binding (II, V) ........................................................................... 51 6 CONCLUSIONS AND FUTURE PERSPECTIVES .....................................
Recommended publications
  • Analysis of Trans Esnps Infers Regulatory Network Architecture
    Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Anat Kreimer All rights reserved ABSTRACT Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer eSNPs are genetic variants associated with transcript expression levels. The characteristics of such variants highlight their importance and present a unique opportunity for studying gene regulation. eSNPs affect most genes and their cell type specificity can shed light on different processes that are activated in each cell. They can identify functional variants by connecting SNPs that are implicated in disease to a molecular mechanism. Examining eSNPs that are associated with distal genes can provide insights regarding the inference of regulatory networks but also presents challenges due to the high statistical burden of multiple testing. Such association studies allow: simultaneous investigation of many gene expression phenotypes without assuming any prior knowledge and identification of unknown regulators of gene expression while uncovering directionality. This thesis will focus on such distal eSNPs to map regulatory interactions between different loci and expose the architecture of the regulatory network defined by such interactions. We develop novel computational approaches and apply them to genetics-genomics data in human. We go beyond pairwise interactions to define network motifs, including regulatory modules and bi-fan structures, showing them to be prevalent in real data and exposing distinct attributes of such arrangements. We project eSNP associations onto a protein-protein interaction network to expose topological properties of eSNPs and their targets and highlight different modes of distal regulation.
    [Show full text]
  • Chromosomal Aberrations in Head and Neck Squamous Cell Carcinomas in Norwegian and Sudanese Populations by Array Comparative Genomic Hybridization
    825-843 12/9/08 15:31 Page 825 ONCOLOGY REPORTS 20: 825-843, 2008 825 Chromosomal aberrations in head and neck squamous cell carcinomas in Norwegian and Sudanese populations by array comparative genomic hybridization ERIC ROMAN1,2, LEONARDO A. MEZA-ZEPEDA3, STINE H. KRESSE3, OLA MYKLEBOST3,4, ENDRE N. VASSTRAND2 and SALAH O. IBRAHIM1,2 1Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 91; 2Department of Oral Sciences - Periodontology, Faculty of Medicine and Dentistry, University of Bergen, Årstadveien 17, 5009 Bergen; 3Department of Tumor Biology, Institute for Cancer Research, Rikshospitalet-Radiumhospitalet Medical Center, Montebello, 0310 Oslo; 4Department of Molecular Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway Received January 30, 2008; Accepted April 29, 2008 DOI: 10.3892/or_00000080 Abstract. We used microarray-based comparative genomic logical parameters showed little correlation, suggesting an hybridization to explore genome-wide profiles of chromosomal occurrence of gains/losses regardless of ethnic differences and aberrations in 26 samples of head and neck cancers compared clinicopathological status between the patients from the two to their pair-wise normal controls. The samples were obtained countries. Our findings indicate the existence of common from Sudanese (n=11) and Norwegian (n=15) patients. The gene-specific amplifications/deletions in these tumors, findings were correlated with clinicopathological variables. regardless of the source of the samples or attributed We identified the amplification of 41 common chromosomal carcinogenic risk factors. regions (harboring 149 candidate genes) and the deletion of 22 (28 candidate genes). Predominant chromosomal alterations Introduction that were observed included high-level amplification at 1q21 (harboring the S100A gene family) and 11q22 (including Head and neck squamous cell carcinoma (HNSCC), including several MMP family members).
    [Show full text]
  • Essential Genes and Their Role in Autism Spectrum Disorder
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2017 Essential Genes And Their Role In Autism Spectrum Disorder Xiao Ji University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Bioinformatics Commons, and the Genetics Commons Recommended Citation Ji, Xiao, "Essential Genes And Their Role In Autism Spectrum Disorder" (2017). Publicly Accessible Penn Dissertations. 2369. https://repository.upenn.edu/edissertations/2369 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/2369 For more information, please contact [email protected]. Essential Genes And Their Role In Autism Spectrum Disorder Abstract Essential genes (EGs) play central roles in fundamental cellular processes and are required for the survival of an organism. EGs are enriched for human disease genes and are under strong purifying selection. This intolerance to deleterious mutations, commonly observed haploinsufficiency and the importance of EGs in pre- and postnatal development suggests a possible cumulative effect of deleterious variants in EGs on complex neurodevelopmental disorders. Autism spectrum disorder (ASD) is a heterogeneous, highly heritable neurodevelopmental syndrome characterized by impaired social interaction, communication and repetitive behavior. More and more genetic evidence points to a polygenic model of ASD and it is estimated that hundreds of genes contribute to ASD. The central question addressed in this dissertation is whether genes with a strong effect on survival and fitness (i.e. EGs) play a specific oler in ASD risk. I compiled a comprehensive catalog of 3,915 mammalian EGs by combining human orthologs of lethal genes in knockout mice and genes responsible for cell-based essentiality.
    [Show full text]
  • Rat Anti-TERF1 Monoclonal Antibody, Clone 683D (CABT-RM172) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Rat Anti-TERF1 monoclonal antibody, clone 683D (CABT-RM172) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Specificity Specifically detects murine Telomeric repeat-binding factor 1 (TRF1). Target TERF1 Immunogen His-tagged full-length recombinant mouse Telomeric repeat-binding factor 1 (TRF1). Isotype IgG1, κ Source/Host Rat Species Reactivity Mouse Clone 683D Purification Protein G purified Conjugate unconjugated Applications FC, ICC, IF, WB Molecular Weight ~51 kDa observed; 48.22 kDa calculated. Uncharacterized bands may be observed in some lysate(s). Format Liquid Size 100 μg, 25 μg Buffer 0.1 M Tris-Glycine (pH 7.4), 150 mM NaCl Preservative 0.05% sodium azide Storage Stable for 1 year at 2-8°C from date of receipt. Warnings Unless otherwise stated in our catalog or other company documentation accompanying the product(s), our products are intended for research use only and are not to be used for any other purpose, which includes but is not limited to, unauthorized commercial uses, in vitro diagnostic uses, ex vivo or in vivo therapeutic uses or any type of consumption or application to humans or animals. 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved BACKGROUND Introduction Telomeric repeat-binding factor 1 is encoded by the Terf1 gene in murine species. TRF1 is a component of the shelterin complex that is involved in the regulation of telomere length and protection. It binds to telomeric DNA as a homodimer and protects telomeres.
    [Show full text]
  • Whole Proteome Analysis of Human Tankyrase Knockout Cells Reveals Targets of Tankyrase- Mediated Degradation
    ARTICLE DOI: 10.1038/s41467-017-02363-w OPEN Whole proteome analysis of human tankyrase knockout cells reveals targets of tankyrase- mediated degradation Amit Bhardwaj1, Yanling Yang2, Beatrix Ueberheide2 & Susan Smith1 Tankyrase 1 and 2 are poly(ADP-ribose) polymerases that function in pathways critical to cancer cell growth. Tankyrase-mediated PARylation marks protein targets for proteasomal 1234567890 degradation. Here, we generate human knockout cell lines to examine cell function and interrogate the proteome. We show that either tankyrase 1 or 2 is sufficient to maintain telomere length, but both are required to resolve telomere cohesion and maintain mitotic spindle integrity. Quantitative analysis of the proteome of tankyrase double knockout cells using isobaric tandem mass tags reveals targets of degradation, including antagonists of the Wnt/β-catenin signaling pathway (NKD1, NKD2, and HectD1) and three (Notch 1, 2, and 3) of the four Notch receptors. We show that tankyrases are required for Notch2 to exit the plasma membrane and enter the nucleus to activate transcription. Considering that Notch signaling is commonly activated in cancer, tankyrase inhibitors may have therapeutic potential in targeting this pathway. 1 Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY 10016, USA. 2 Proteomics Laboratory, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA. Correspondence and requests for materials should be addressed to S.S. (email: [email protected]) NATURE COMMUNICATIONS | 8: 2214 | DOI: 10.1038/s41467-017-02363-w | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02363-w ankyrases function in cellular pathways that are critical to function in human cells will provide insights into the clinical cancer cell growth including telomere cohesion and length utility of tankyrase inhibitors.
    [Show full text]
  • Integrating Protein Copy Numbers with Interaction Networks to Quantify Stoichiometry in Mammalian Endocytosis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.10.29.361196; this version posted October 29, 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-ND 4.0 International license. Integrating protein copy numbers with interaction networks to quantify stoichiometry in mammalian endocytosis Daisy Duan1, Meretta Hanson1, David O. Holland2, Margaret E Johnson1* 1TC Jenkins Department of Biophysics, Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218. 2NIH, Bethesda, MD, 20892. *Corresponding Author: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.10.29.361196; this version posted October 29, 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-ND 4.0 International license. Abstract Proteins that drive processes like clathrin-mediated endocytosis (CME) are expressed at various copy numbers within a cell, from hundreds (e.g. auxilin) to millions (e.g. clathrin). Between cell types with identical genomes, copy numbers further vary significantly both in absolute and relative abundance. These variations contain essential information about each protein’s function, but how significant are these variations and how can they be quantified to infer useful functional behavior? Here, we address this by quantifying the stoichiometry of proteins involved in the CME network. We find robust trends across three cell types in proteins that are sub- vs super-stoichiometric in terms of protein function, network topology (e.g.
    [Show full text]
  • Mayer-Rokitansky-Küster- Hauser Syndrome
    Morcel et al. Orphanet Journal of Rare Diseases 2011, 6:9 http://www.ojrd.com/content/6/1/9 RESEARCH Open Access Utero-vaginal aplasia (Mayer-Rokitansky-Küster- Hauser syndrome) associated with deletions in known DiGeorge or DiGeorge-like loci Karine Morcel1,2*†, Tanguy Watrin1†, Laurent Pasquier1,3, Lucie Rochard1, Cédric Le Caignec4,5, Christèle Dubourg1,6, Philippe Loget7, Bernard-Jean Paniel8, Sylvie Odent1,3, Véronique David1,6, Isabelle Pellerin1, Claude Bendavid1,6 and Daniel Guerrier1 Abstract Background: Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is characterized by congenital aplasia of the uterus and the upper part of the vagina in women showing normal development of secondary sexual characteristics and a normal 46, XX karyotype. The uterovaginal aplasia is either isolated (type I) or more frequently associated with other malformations (type II or Müllerian Renal Cervico-thoracic Somite (MURCS) association), some of which belong to the malformation spectrum of DiGeorge phenotype (DGS). Its etiology remains poorly understood. Thus the phenotypic manifestations of MRKH and DGS overlap suggesting a possible genetic link. This would potentially have clinical consequences. Methods: We searched DiGeorge critical chromosomal regions for chromosomal anomalies in a cohort of 57 subjects with uterovaginal aplasia (55 women and 2 aborted fetuses). For this candidate locus approach, we used a multiplex ligation-dependent probe amplification (MLPA) assay based on a kit designed for investigation of the chromosomal regions known to be involved in DGS. The deletions detected were validated by Duplex PCR/liquid chromatography (DP/LC) and/or array-CGH analysis. Results: We found deletions in four probands within the four chromosomal loci 4q34-qter, 8p23.1, 10p14 and 22q11.2 implicated in almost all cases of DGS syndrome.
    [Show full text]
  • APC Mutations As a Potential Biomarker for Sensitivity To
    Published OnlineFirst February 8, 2017; DOI: 10.1158/1535-7163.MCT-16-0578 Companion Diagnostics and Cancer Biomarkers Molecular Cancer Therapeutics APC Mutations as a Potential Biomarker for Sensitivity to Tankyrase Inhibitors in Colorectal Cancer Noritaka Tanaka1,2, Tetsuo Mashima1, Anna Mizutani1, Ayana Sato1,3, Aki Aoyama3,4, Bo Gong3,4, Haruka Yoshida1, Yukiko Muramatsu1, Kento Nakata1,5, Masaaki Matsuura6, Ryohei Katayama4, Satoshi Nagayama7, Naoya Fujita3,4,5, Yoshikazu Sugimoto2, and Hiroyuki Seimiya1,3,5 Abstract In most colorectal cancers, Wnt/b-catenin signaling is acti- "short" truncated APCs lacking all seven b-catenin-binding vated by loss-of-function mutations in the adenomatous polyposis 20-amino acid repeats (20-AARs). In contrast, the drug-resistant coli (APC) gene and plays a critical role in tumorigenesis. cells possessed "long" APC retaining two or more 20-AARs. Knock- Tankyrases poly(ADP-ribosyl)ate and destabilize Axins, a neg- down of the long APCs with two 20-AARs increased b-catenin, ative regulator of b-catenin, and upregulate b-catenin signaling. Tcf/LEF transcriptional activity and its target gene AXIN2 expres- Tankyrase inhibitors downregulate b-catenin and are expected sion. Under these conditions, tankyrase inhibitors were able to to be promising therapeutics for colorectal cancer. However, downregulate b-catenin in the resistant cells. These results indicate colorectal cancer cells are not always sensitive to tankyrase that the long APCs are hypomorphic mutants, whereas they exert inhibitors, and predictive biomarkers for the drug sensitivity a dominant-negative effect on Axin-dependent b-catenin degra- remain elusive. Here we demonstrate that the short-form APC dation caused by tankyrase inhibitors.
    [Show full text]
  • Atlas Journal
    Atlas of Genetics and Cytogenetics in Oncology and Haematology Home Genes Leukemias Solid Tumours Cancer-Prone Deep Insight Portal Teaching X Y 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 NA Atlas Journal Atlas Journal versus Atlas Database: the accumulation of the issues of the Journal constitutes the body of the Database/Text-Book. TABLE OF CONTENTS Volume 5, Number 2, Apr-Jun 2001 Previous Issue / Next Issue Genes COL1A1 (collagen, type I, alpha 1) (17q21.31-q22). Marie-Pierre Simon, Georges Maire, Florence Pedeutour. Atlas Genet Cytogenet Oncol Haematol 2001; 5 (2): 169-177. [Full Text] [PDF] URL : http://AtlasGeneticsOncology.org/Genes/COL1A1ID186.html NF2 (neurofibromatosis type 2) (22q12.1-12.2) - updated. James F Gusella. Atlas Genet Cytogenet Oncol Haematol 2001; 5 (2): 178-184. [Full Text] [PDF] URL : http://AtlasGeneticsOncology.org/Genes/NF2117.html PDGFB (22q12.3-q13.1). Marie-Pierre Simon, Georges Maire, Florence Pedeutour. Atlas Genet Cytogenet Oncol Haematol 2001; 5 (2): 185-194. [Full Text] [PDF] URL : http://AtlasGeneticsOncology.org/Genes/PDGFBID155.html XPA (9q22.3-9q22.3). Anne Stary, Alain Sarasin. Atlas Genet Cytogenet Oncol Haematol 2001; 5 (2): 195-204. [Full Text] [PDF] Atlas Genet Cytogenet Oncol Haematol 2001; 2 I URL : http://AtlasGeneticsOncology.org/Genes/XPAID104.html ERCC-3 (Excision repair cross-complementing rodent repair deficiency, complementation group 3) (2q21). Anne Stary, Alain Sarasin. Atlas Genet Cytogenet Oncol Haematol 2001; 5 (2): 205-214. [Full Text] [PDF] URL : http://AtlasGeneticsOncology.org/Genes/XPBID296.html XPC (3p25.1).
    [Show full text]
  • * Supplementary Table 3B. Complete List of Lymphocytic-Associated Genes
    Supplementary Table 3b. Complete list of lymphocytic-associated genes for the HER2+I subtype based on the SNR score (p-val <= 0.005 and FDR<=0.05). A positive score represents genes up-regulated in HER2+I and a negative score represents genes up-regulated in HER2+NI. The first 9 genes, marked with *, are chemokines near the HER2+ amplicon at chr17q12. The remaining are sorted based by chromosomal location (from chr 1 to chr X) and signal-to-noise ratio (SNR). Gene Name Description SNR Score Feature P value FDR(BH) Q Value * CCL5 chemokine (C-C motif) ligand 5 1.395 0.002 0.036 0.025 * CCR7 chemokine (C-C motif) receptor 7 1.362 0.002 0.036 0.025 * CD79B CD79B antigen (immunoglobulin-associated beta) 1.248 0.002 0.036 0.025 * CCL13 chemokine (C-C motif) ligand 13 1.003 0.002 0.036 0.025 * CCL2 chemokine (C-C motif) ligand 2 0.737 0.004 0.056 0.037 * CCL8 chemokine (C-C motif) ligand 8 0.724 0.002 0.036 0.025 * CCL18 chemokine (C-C motif) ligand 18 (pulmonary and activ 0.703 0.002 0.036 0.025 * CCL23 chemokine (C-C motif) ligand 23 0.701 0.002 0.036 0.025 * CCR6 chemokine (C-C motif) receptor 6 0.715 0.002 0.036 0.025 PTPN7 protein tyrosine phosphatase, non-receptor type 7 1.861 0.002 0.036 0.025 CD3Z CD3Z antigen, zeta polypeptide (TiT3 complex) 1.811 0.002 0.036 0.025 CD48 CD48 antigen (B-cell membrane protein) 1.809 0.002 0.036 0.025 IL10RA interleukin 10 receptor, alpha 1.806 0.002 0.036 0.025 SELL selectin L (lymphocyte adhesion molecule 1) 1.773 0.002 0.036 0.025 LCK lymphocyte-specific protein tyrosine kinase 1.745 0.002 0.036 0.025
    [Show full text]
  • Common Differentially Expressed Genes and Pathways Correlating Both Coronary Artery Disease and Atrial Fibrillation
    EXCLI Journal 2021;20:126-141– ISSN 1611-2156 Received: December 08, 2020, accepted: January 11, 2021, published: January 18, 2021 Supplementary material to: Original article: COMMON DIFFERENTIALLY EXPRESSED GENES AND PATHWAYS CORRELATING BOTH CORONARY ARTERY DISEASE AND ATRIAL FIBRILLATION Youjing Zheng, Jia-Qiang He* Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA * Corresponding author: Jia-Qiang He, Department of Biomedical Sciences and Pathobiology, Virginia Tech, Phase II, Room 252B, Blacksburg, VA 24061, USA. Tel: 1-540-231-2032. E-mail: [email protected] https://orcid.org/0000-0002-4825-7046 Youjing Zheng https://orcid.org/0000-0002-0640-5960 Jia-Qiang He http://dx.doi.org/10.17179/excli2020-3262 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/). Supplemental Table 1: Abbreviations used in the paper Abbreviation Full name ABCA5 ATP binding cassette subfamily A member 5 ABCB6 ATP binding cassette subfamily B member 6 (Langereis blood group) ABCB9 ATP binding cassette subfamily B member 9 ABCC10 ATP binding cassette subfamily C member 10 ABCC13 ATP binding cassette subfamily C member 13 (pseudogene) ABCC5 ATP binding cassette subfamily C member 5 ABCD3 ATP binding cassette subfamily D member 3 ABCE1 ATP binding cassette subfamily E member 1 ABCG1 ATP binding cassette subfamily G member 1 ABCG4 ATP binding cassette subfamily G member 4 ABHD18 Abhydrolase domain
    [Show full text]
  • Genetic Studies of Leptin Concentrations Implicate Leptin in the Regulation of Early
    Page 1 of 93 Diabetes Genetic studies of leptin concentrations implicate leptin in the regulation of early adiposity Hanieh Yaghootkar1,2,3*&, Yiying Zhang4&, Cassandra N Spracklen5, Tugce Karaderi6,7,8,9, Lam Opal Huang10, Jonathan Bradfield11,12, Claudia Schurmann13, Rebecca S Fine14,15,16, Michael H Preuss13, Zoltan Kutalik1,17,18, Laura BL Wittemans6,19, Yingchang Lu13,20, Sophia Metz10, Sara M Willems19, Ruifang Li-Gao21, Niels Grarup10, Shuai Wang22, Sophie Molnos23,24, América A Sandoval-Zárate25, Mike A Nalls26,27, Leslie A Lange28, Jeffrey Haesser29, Xiuqing Guo30, Leo-Pekka Lyytikäinen31,32, Mary F Feitosa33, Colleen M Sitlani34, Cristina Venturini35, Anubha Mahajan6,36, Tim Kacprowski37,38, Carol A Wang22, Daniel I Chasman39,40, Najaf Amin41, Linda Broer42, Neil Robertson6,36, Kristin L Young43, Matthew Allison44, Paul L Auer45, Matthias Blüher46, Judith B Borja47,48, Jette Bork-Jensen10, Germán D Carrasquilla10, Paraskevi Christofidou35, Ayse Demirkan41, Claudia A Doege49, Melissa E Garcia50, Mariaelisa Graff43,51, Kaiying Guo4, Hakon Hakonarson11,52, Jaeyoung Hong22, Yii-Der Ida Chen30, Rebecca Jackson53, Hermina Jakupović10, Pekka Jousilahti54, Anne E Justice55, Mika Kähönen56,57, Jorge R Kizer58,59, Jennifer Kriebel23,24, Charles A LeDuc4, Jin Li60, Lars Lind61, Jian’an Luan19, David Mackey62, Massimo Mangino35,63, Satu Männistö54, Jayne F Martin Carli4, Carolina Medina-Gomez41,42, Dennis O Mook-Kanamori21,64, Andrew P Morris65,6,66, Renée de Mutsert21, Matthias Nauck67,38, Ivana Nedeljkovic41, Craig E Pennell22, Arund D Pradhan39,40, Bruce M Psaty68,69, Olli T Raitakari70,71,72, Robert A Scott19, Tea Skaaby73, Konstantin Strauch74,75, Kent D Taylor30, Alexander Teumer76,38, Andre G Uitterlinden41,42, Ying Wu5, Jie Yao30, Mark Walker77, Kari E North43, Peter Kovacs46, M.
    [Show full text]