Biochemical and Cell Biological Characterisation of Sumo E1 Activating Enzyme Aos1/Uba2

Total Page:16

File Type:pdf, Size:1020Kb

Biochemical and Cell Biological Characterisation of Sumo E1 Activating Enzyme Aos1/Uba2 Dissertation zur Erlangung des Doktorgrades der Fakultät für Chemie und Pharmazie der Ludwig-Maximilians-Universität München Biochemical and cell biological characterisation of the SUMO E1 activating enzyme Aos1/Uba2 Katarzyna Chmielarska aus Kielce, Polen 2005 Erklärung Diese Dissertation wurde im Sinne von § 13 Abs. 3 bzw. 4 der Promotionsordnung vom 29. Januar 1998 von Prof. Dr. Frauke Melchior and Prof. Dr. Patrick Cramer betreut. Ehrenwörtliche Versicherung Diese Dissertation wurde selbständig, ohne unerlaubte Hilfe erarbeitet. München, 6.10.2005 Katarzyna Chmielarska Dissertation eingereicht am 6.10.2005 1. Gutachter: Prof. Dr. Patrick Cramer 2. Gutachter: Prof. Dr. Alexander Pfeifer Mündliche Prüfung am 20.12.2005 TABLE OF CONTENTS LIST OF ABBREVIATIONS 1 1 SUMMARY 4 2 INTRODUCTION 4 2.1 Overview 4 2.2 Molecular mechanism of Sumoylation 9 2.2.1 Enzymes mediating the SUMO pathway 9 2.2.2 SUMO-Activating Enzyme (E1) 10 2.2.3 SUMO-Conjugating Enzyme (E2) 13 2.2.4 E3 ligases 14 2.2.5 SUMO processing and deconjugation 18 2.3 Function of SUMO modifications 19 2.3.1 Molecular consequences of sumoylation 20 2.3.2 Pathways in which SUMO was found 21 3 MATERIALS AND METHODS 24 3.1 Materials 24 3.1.1 Chemicals and materials 24 3.1.2 Reagents and reaction kits 25 3.1.3 Buffers and solutions 26 3.1.4 Cell culture 28 3.1.5 Antibodies 29 3.1.6 Recombinant proteins 31 3.1.7 Plasmids 31 3.1.8 Important oligonucleotides 32 3.1.9 Bacterial strains 33 3.2 Molecular biology methods 34 3.2.1 Plasmid DNA purification (miniprep) 34 3.2.2 Plasmid DNA (midi, maxiprep) 34 3.2.3 Preparation of chemically competent cells 34 3.2.4 Estimation of DNA concentration 35 3.2.5 Digestion of plasmid DNA with restriction endonucleases 35 3.2.6 DNA purification 36 3.2.7 Ligation of DNA insert into vector DNA 36 3.2.8 Transformation into DH5 α 36 3.2.9 Polymerase Chain Reaction (PCR) 37 3.2.10 Mutagenesis 37 3.2.11 Sequencing 40 3.2.12 Organ extracts 40 3.2.13 RNA isolation 40 3.2.14 RT-PCR 41 3.3 Cell biology methods 41 3.3.1 Cell lines 41 3.3.2. Transient transfection 42 3.3.3 Fluorescence microscopy 43 3.3.4 Reporter luciferase assay 43 3.4 Biochemistry methods 44 3.4.1 SDS-PAGE 44 3.4.2 Coomassie staining 44 3.4.3 Silver staining 44 3.4.4 Immunoblotting 45 3.4.5 Expression and purification of recombinant proteins 46 3.4.6 Measurement of protein concentration 48 3.4.7 In vitro SUMOylation 48 3.4.8 Fluorescence Resonance Energy Transfer (FRET) 48 3.4.9 Generation and purification of rabbit or goat polyclonal antibodies 50 3.4.10 Immunoprecipitation 50 3.4.11 His-SUMO pull down 52 3.4.12 Mass Spectrometry 52 4 RESULTS 4.1 PART 1: Characterisation of an Uba2 splice variant 54 4.1.1 Identification of Uba2 splice variant 54 4.1.2 Generation and characterisation of Uba2 polyclonal antibody in goat 56 4.1.3 In vivo and in vitro analysis of Uba2 splice variant 59 4.1.4 Finding new binding partners for Uba2 full length and Uba2 splice variant 64 4.2 PART 2: Identification of a novel SUMO target 66 4.2.1 Affinity purification and characterisation of Aos1 polyclonal antibody 66 4.2.2 α Aos1 antibodies decorate the nucleus and Golgi structures 70 4.2.3 A small fraction of Aos1 and Uba2 are present in unsoluble pellet fractions 70 4.2.4 Aos1/Uba2 is present in Golgi fraction 73 4.2.5 Identification of a new binding partner (ELKS) for Aos1 73 4.2.6 Published functions of ELKS 76 4.2.7 Generation of recombinant ELKS 78 4.2.8 Generation and characterisation of ELKS polyclonal antibodies 81 4.2.9 Conformation of ELKS-Aos1 interaction 83 4.2.10 Subcellular localisation of ELKS 85 4.2.11 Recombinant of ELKS and Aos1 do not bind 88 4.2.12 Rab6 is not the binding factor between ELKS and Aos1 88 4.2.13 ELKS does not interact directly with SUMO in vitro 90 4.2.14 Identification of binding partner candidates for Aos1 and ELKS 91 4.2.15 ELKS interacts with RanBP2 in interphase cell extract 93 4.2.16 ELKS does not interact directly with BP2 ∆FG in vitro 94 4.2.17 ELKS does not inhibit sumoylation 95 4.2.18 ELKS is SUMO modified on 3 distinct lysine residues 98 4.2.19 ELKS sumoylation decreases its NF κΒ stimulating activity 106 5 APPENDIX 110 6 DISCUSSION 112 6.1 Characterisation of variant Uba2 112 6.2 Identification of a novel SUMO target 113 5.2.1 Identification of a new Aos1 binding partner 113 5.2.2 ELKS is a protein implicated in NF κB signaling and vesicle transport 114 6.3 Identification of additional ELKS and Aos1 binding partners suggest possible functions in vesicle transport and NF κκκB signaling 117 6.4 ELKS is a new SUMO substrate 118 6.5 SUMO does not change ELKS localisation and Rab6 interaction 119 6.6 ELKS negatively regulates the NF κκκB signaling pathway 120 6.7 RanBP2-an E3 ligase for ELKS? 122 7 REFERENCES 124 8 ACKNOWLEGMENTS 139 9 PUBLICATIONS 140 10 CURRICULUM VITAE 141 Abbreviations 1 ABBREVIATIONS aa amino acid Ac acetat AP aprotinin ATP adenosine-5‘-triphosphate bp base pair BSA bovine serum albumin cDNA complementary DNA Cys cysteine Da dalton dNTP 2‘-Didesoxynucleosid-5‘-triphospate ddH20 double-destilled water DEPC diethyl pyrocarbonate DMEM Dulbecco‘s modified Eagle‘s medium DMSO dimethyl sulfoxide DTT dithiothreitol E.coli Escherichia coli ECL enhanced chemoluminescence EDTA ethylenediaminetetraacetic EGTA ethyleneglycoltetra acetic acid FBS fetal bovine serum FCS fetal calf serum FITC fluorescein isothiocyanate FPLC fast protein liquid chromatography g centrifugation force g gramm GFP green fluorescent protein GTP guanosine-5‘-triphosphate h hour(s) HA Hemagglutinin Abbreviations 2 7EPES N-(2-Hydroxyethyl)piperazine-N‘-(2-ethanesulfonic acid) His Histidine HPLC high-performance liquid chromatography IF immunofluorescence IgG immunoglobulin G IP immunoprecipitation IPTG isopropylthiogalactoside kb kilobase(s) l liter L/P leupeptin/pepstatin LB Luria-Bertani-Medium m mili M molar (mol/l) min minute(s) mRNA messenger ribonucleic acid MW molecular weight µ micro n nano nc nitrocellulose NCS newborn calf serum NEM N-ethylmaleimide NF κκκB nuclear factor kappa B NPC nuclear pore complex OD optical density units ORF open reading frame p pico PAGE polyacrylamide gel electrophoresis pH potential hydrogen PBS phosphate buffered saline PCR polymerase chain reaction Abbreviations 3 2T-PCR reverse transcription-polymerase chain reaction pfu Pyrococcus furiosus (DNA polymerase) PMSF phenylmethanesulfonyl fluoride RIPA radio-immunoprecipitation assay RNase ribonuclease rpm revolutions per minute RT room temperature RT-PCR reverse transcription followed by PCR S. cerevisiae Saccharomyces cerevisiae SDS sodium dodecyl sulfate s second(s) TB transport buffer TEMED N, N, N‘,N‘-Tetramethylethylenediamine TNF ααα Gumor necrosis factor alpha Tris Tris-”(hydroxymethyl)-aminomethane Triton X-100 t-octylphenoxypolyethoxyethanol Tween 20 polyoxyethylenesorbitan monolaurate ub ubiquitin UV ultraviolet light v/v volume per volume w/v weight per volume V volt Vol volume WB Western Blot Summary 4 1 SUMMARY Small ubiquitin-related modifier (SUMO) is a protein that is attached to lysine residues in a variety of target proteins. Sumoylation of proteins can alter their intracellular localisation, stability, activity and interaction with other proteins. The pathway of sumoylation is analogous to that of ubiquitination. The reaction is ATP dependent and requires the E1-activating enzyme (Aos1/Uba2), the E2-conjugation enzyme (Ubc9) and for most target proteins SUMO E3 ligases. The aim of this study was to characterise the SUMO E1 enzyme, a heterodimer consisting of the subunits Aos1 and Uba2. On one hand I characterised an Uba2 splice variant, which lacks one exon encoding 50 amino acids. Using RT-PCR I could determine the tissue specific distribution of the Uba2 splice variant. I furthermore showed that Uba2 variant protein is still able to form an active E1 enzyme complex with Aos1. I could demonstrate that variant Aos1/Uba2 complex is fully active in RanGAP1 sumoylation with SUMO1 or SUMO2. This finding was surprising in light of the missing amino acids, and will have implications for the understanding of E1 function. A large part of my work was dedicated to the identification and characterization of a novel SUMO substrate called ELKS. According to literature, ELKS proteins have been linked to intracellular membrane traffic and NF κB signaling pathways. I identified ELKS in membrane fractions as a binding partner for the Aos1 subunit of the SUMO E1 enzyme and confirmed in vivo interaction with ELKS antibodies that I generated. Because recombinant proteins did not interact directly, I searched for potential bridging factors. Neither SUMO nor Ubc9 or Rab6 (one ELKS partner) mediated interaction between ELKS and Aos1. Performing a large scale immunoprecipitation and analysis by mass spectrometry, I could find several candidates, including nucleoporin RanBP2, a SUMO E3 ligase. This suggested that ELKS may be a target for sumoylation. Indeed, I could show that ELKS was SUMO-modified in vivo and in vitro . Moreover, RanBP2 enhanced ELKS sumoylation. By mass spectrometry I identified two SUMO acceptor sites in ELKS. Mutation of these two residues had no effect on ELKS localisation, but strongly inhibited ELKS induced NF κB activation. In conclusion, work described in this thesis implicates sumoylation as an important mechanism for ELKS function in NF κB signaling. Introduction 0 2 INTRODUCTION 2.1 Overview Posttranslational protein modifications Posttranslational modification of proteins is a fundamental mechanism of modulating their function, activity or localisation after their synthesis has been completed.
Recommended publications
  • SUMO-1 Regulates the Conformational Dynamics of Thymine-DNA
    Smet-Nocca et al. BMC Biochemistry 2011, 12:4 http://www.biomedcentral.com/1471-2091/12/4 RESEARCHARTICLE Open Access SUMO-1 regulates the conformational dynamics of Thymine-DNA Glycosylase regulatory domain and competes with its DNA binding activity Caroline Smet-Nocca1, Jean-Michel Wieruszeski2, Hélène Léger1,3, Sebastian Eilebrecht1,3, Arndt Benecke1,3* Abstract Background: The human thymine-DNA glycosylase (TDG) plays a dual role in base excision repair of G:U/T mismatches and in transcription. Regulation of TDG activity by SUMO-1 conjugation was shown to act on both functions. Furthermore, TDG can interact with SUMO-1 in a non-covalent manner. Results: Using NMR spectroscopy we have determined distinct conformational changes in TDG upon either covalent sumoylation on lysine 330 or intermolecular SUMO-1 binding through a unique SUMO-binding motif (SBM) localized in the C-terminal region of TDG. The non-covalent SUMO-1 binding induces a conformational change of the TDG amino-terminal regulatory domain (RD). Such conformational dynamics do not exist with covalent SUMO-1 attachment and could potentially play a broader role in the regulation of TDG functions for instance during transcription. Both covalent and non-covalent processes activate TDG G:U repair similarly. Surprisingly, despite a dissociation of the SBM/SUMO-1 complex in presence of a DNA substrate, SUMO-1 preserves its ability to stimulate TDG activity indicating that the non-covalent interactions are not directly involved in the regulation of TDG activity. SUMO-1 instead acts, as demonstrated here, indirectly by competing with the regulatory domain of TDG for DNA binding.
    [Show full text]
  • Yeast Genome Gazetteer P35-65
    gazetteer Metabolism 35 tRNA modification mitochondrial transport amino-acid metabolism other tRNA-transcription activities vesicular transport (Golgi network, etc.) nitrogen and sulphur metabolism mRNA synthesis peroxisomal transport nucleotide metabolism mRNA processing (splicing) vacuolar transport phosphate metabolism mRNA processing (5’-end, 3’-end processing extracellular transport carbohydrate metabolism and mRNA degradation) cellular import lipid, fatty-acid and sterol metabolism other mRNA-transcription activities other intracellular-transport activities biosynthesis of vitamins, cofactors and RNA transport prosthetic groups other transcription activities Cellular organization and biogenesis 54 ionic homeostasis organization and biogenesis of cell wall and Protein synthesis 48 plasma membrane Energy 40 ribosomal proteins organization and biogenesis of glycolysis translation (initiation,elongation and cytoskeleton gluconeogenesis termination) organization and biogenesis of endoplasmic pentose-phosphate pathway translational control reticulum and Golgi tricarboxylic-acid pathway tRNA synthetases organization and biogenesis of chromosome respiration other protein-synthesis activities structure fermentation mitochondrial organization and biogenesis metabolism of energy reserves (glycogen Protein destination 49 peroxisomal organization and biogenesis and trehalose) protein folding and stabilization endosomal organization and biogenesis other energy-generation activities protein targeting, sorting and translocation vacuolar and lysosomal
    [Show full text]
  • T-Cell Receptor (TCR) Signaling Promotes the Assembly of Ranbp2
    RESEARCH ARTICLE T-cell receptor (TCR) signaling promotes the assembly of RanBP2/RanGAP1- SUMO1/Ubc9 nuclear pore subcomplex via PKC--mediated phosphorylation of RanGAP1 Yujiao He1, Zhiguo Yang1†, Chen-si Zhao1†, Zhihui Xiao1†, Yu Gong1, Yun-Yi Li1, Yiqi Chen1, Yunting Du1, Dianying Feng1, Amnon Altman2, Yingqiu Li1* 1MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China; 2Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, United States Abstract The nuclear pore complex (NPC) is the sole and selective gateway for nuclear transport, and its dysfunction has been associated with many diseases. The metazoan NPC subcomplex RanBP2, which consists of RanBP2 (Nup358), RanGAP1-SUMO1, and Ubc9, regulates the assembly and function of the NPC. The roles of immune signaling in regulation of NPC remain poorly understood. Here, we show that in human and murine T cells, following T-cell receptor (TCR) stimulation, protein kinase C-q (PKC-q) directly phosphorylates RanGAP1 to facilitate RanBP2 subcomplex assembly and nuclear import and, thus, the nuclear translocation of AP-1 transcription *For correspondence: factor. Mechanistically, TCR stimulation induces the translocation of activated PKC-q to the NPC, 504 506 [email protected] where it interacts with and phosphorylates RanGAP1 on Ser and Ser . RanGAP1 phosphorylation increases its binding affinity for Ubc9, thereby promoting sumoylation of RanGAP1 †These authors contributed and, finally, assembly of the RanBP2 subcomplex. Our findings reveal an unexpected role of PKC-q equally to this work as a direct regulator of nuclear import and uncover a phosphorylation-dependent sumoylation of Competing interests: The RanGAP1, delineating a novel link between TCR signaling and assembly of the RanBP2 NPC authors declare that no subcomplex.
    [Show full text]
  • Identifying SUMO Protease Targets and Investigating E3 Ligase Interactions
    W&M ScholarWorks Dissertations, Theses, and Masters Projects Theses, Dissertations, & Master Projects 2014 Identifying SUMO Protease Targets and Investigating E3 Ligase Interactions Mark Guillotte College of William & Mary - Arts & Sciences Follow this and additional works at: https://scholarworks.wm.edu/etd Part of the Molecular Biology Commons Recommended Citation Guillotte, Mark, "Identifying SUMO Protease Targets and Investigating E3 Ligase Interactions" (2014). Dissertations, Theses, and Masters Projects. Paper 1539626956. https://dx.doi.org/doi:10.21220/s2-wrgj-tz43 This Thesis is brought to you for free and open access by the Theses, Dissertations, & Master Projects at W&M ScholarWorks. It has been accepted for inclusion in Dissertations, Theses, and Masters Projects by an authorized administrator of W&M ScholarWorks. For more information, please contact [email protected]. Identifying SUMO protease targets and investigating E3 ligase interactions Mark Guillotte Baton Rouge, Louisiana Bachelors of Science, Louisiana State University, 2010 A Thesis presented to the Graduate Faculty of the College of William and Mary in Candidacy for the Degree of Master of Science Department of Biology The College of William and Mary January 2014 APPROVAL PAGE This Thesis is submitted in partial fulfillment of the requirements for the degree of Master of Science Mark Guillotte Approved by^he Committee, January 2014 (be Chair Associate Professor Oliver Kerseher, Biology The College of William and Mary ' oVY wG ..G S l1m>. rofessor Lizabeth Allison, Biology The College of William and Mary Professor Diane Shakes, Biology The College of William and Mary ---------- Assistant Professor Shanta Hinton,Biology The College of William and Mary COMPLIANCE PAGE Research approved by Steve Kaattari Protocol number(s): IBC-2012-10-08-8156-opkers Date(s) of approval: 2013-11-02 ABSTRACT Posttranslational modification by the Small Ubiquitin-like Modifier (SUMO) is a pervasive mechanism for controlling protein function.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • UBA2 Activates Wnt/ Β-Catenin Signaling Pathway
    UBA2 Activates Wnt/ β-catenin Signaling Pathway during Protection of R28 Retinal Precursor Cells from Hypoxia by Extracellular Vesicles derived from Placental Mesenchymal Stem Cells Kyungmin Koh CHA University Mira Park CHA University Eun Soo Bae CHA University Van-An Duong Gachon University Jong-Moon Park Gachon University Hookeun Lee Gachon University Helen Lew ( [email protected] ) CHA university https://orcid.org/0000-0003-0121-3000 Research Keywords: Exosome, hPMSCs, Optic nerve injury, Retinal precursor cells, UBA2, Wnt/β-catenin DOI: https://doi.org/10.21203/rs.3.rs-33694/v3 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/23 Abstract Background: Stem cell transplantation has been proposed as an alternative treatment for intractable optic nerve disorders characterized by irrecoverable loss of cells. Mesenchymal stem cells, with varying tissue regeneration and recovery capabilities, are being considered for potential cell therapies. To overcome the limitations of cell therapy, we isolated exosomes from human placenta–derived mesenchymal stem cells (hPMSCs), and investigated their therapeutic effects in R28 cells (retinal precursor cells) exposed to CoCl2. Method: After nine hours of exposure to CoCl2, the hypoxic damaged R28 cells were divided into non treatment group (CoCl2+R28 cells) and treatment group (CoCl2+R28 cells treated with exosome). Immunoblot analysis was performed for Pcna, Hif-1α, Vegf, Vimentin, Thy-1, Gap43, Ermn, Neuro≈ament, Wnt3a, β-catenin, phospo-GSK3β, Lef-1, UBA2, Skp1, βTrcp, and ubiquitin. The proteomes of each group were analyzed by liquid chromatography/tandem mass (LC-MS/MS) spectrometry. Differentially expressed proteins (DEPs) were detected by label-free quantiƒcation and the interactions of the proteins were examined through signal transduction pathway and gene ontology analysis.
    [Show full text]
  • The E3 Ligase PIAS1 Regulates P53 Sumoylation to Control Stress-Induced Apoptosis of Lens
    fcell-09-660494 June 14, 2021 Time: 13:5 # 1 ORIGINAL RESEARCH published: 14 June 2021 doi: 10.3389/fcell.2021.660494 The E3 Ligase PIAS1 Regulates p53 Sumoylation to Control Stress-Induced Apoptosis of Lens Edited by: Wolfgang Knabe, Epithelial Cells Through the Universität Münster, Germany Proapoptotic Regulator Bax Reviewed by: Guillaume Bossis, † † † † † Centre National de la Recherche Qian Nie , Huimin Chen , Ming Zou , Ling Wang , Min Hou , Jia-Wen Xiang, Scientifique (CNRS), France Zhongwen Luo, Xiao-Dong Gong, Jia-Ling Fu, Yan Wang, Shu-Yu Zheng, Yuan Xiao, Stefan Müller, Yu-Wen Gan, Qian Gao, Yue-Yue Bai, Jing-Miao Wang, Lan Zhang, Xiang-Cheng Tang, Goethe University Frankfurt, Germany Xuebin Hu, Lili Gong, Yizhi Liu* and David Wan-Cheng Li*‡ Leena Latonen, University of Eastern Finland, Finland State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China *Correspondence: David Wan-Cheng Li Protein sumoylation is one of the most important post-translational modifications [email protected] Yizhi Liu regulating many biological processes (Flotho A & Melchior F. 2013. Ann Rev. Biochem. [email protected] 82:357–85). Our previous studies have shown that sumoylation plays a fundamental †These authors have contributed role in regulating lens differentiation (Yan et al., 2010. PNAS, 107(49):21034-9.; equally to this work Gong et al., 2014. PNAS. 111(15):5574–9). Whether sumoylation is implicated in lens ‡ ORCID: David Wan-Cheng Li pathogenesis remains elusive. Here, we present evidence to show that the protein orcid.org/0000-0002-7398-7630 inhibitor of activated STAT-1 (PIAS1), a E3 ligase for sumoylation, is implicated in regulating stress-induced lens pathogenesis.
    [Show full text]
  • The Role of Sumoylation of DNA Topoisomerase Iiα C-Terminal Domain in the Regulation of Mitotic Kinases In
    SUMOylation at the centromere: The role of SUMOylation of DNA topoisomerase IIα C-terminal domain in the regulation of mitotic kinases in cell cycle progression. By Makoto Michael Yoshida Submitted to the graduate degree program in the Department of Molecular Biosciences and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. ________________________________________ Chairperson: Yoshiaki Azuma, Ph.D. ________________________________________ Roberto De Guzman, Ph.D. ________________________________________ Kristi Neufeld, Ph.D. _________________________________________ Berl Oakley, Ph.D. _________________________________________ Blake Peterson, Ph.D. Date Defended: July 12, 2016 The Dissertation Committee for Makoto Michael Yoshida certifies that this is the approved version of the following dissertation: SUMOylation at the centromere: The role of SUMOylation of DNA topoisomerase IIα C-terminal domain in the regulation of mitotic kinases in cell cycle progression. ________________________________________ Chairperson: Yoshiaki Azuma, Ph.D. Date approved: July 12, 2016 ii ABSTRACT In many model systems, SUMOylation is required for proper mitosis; in particular, chromosome segregation during anaphase. It was previously shown that interruption of SUMOylation through the addition of the dominant negative E2 SUMO conjugating enzyme Ubc9 in mitosis causes abnormal chromosome segregation in Xenopus laevis egg extract (XEE) cell-free assays, and DNA topoisomerase IIα (TOP2A) was identified as a substrate for SUMOylation at the mitotic centromeres. TOP2A is SUMOylated at K660 and multiple sites in the C-terminal domain (CTD). We sought to understand the role of TOP2A SUMOylation at the mitotic centromeres by identifying specific binding proteins for SUMOylated TOP2A CTD. Through affinity isolation, we have identified Haspin, a histone H3 threonine 3 (H3T3) kinase, as a SUMOylated TOP2A CTD binding protein.
    [Show full text]
  • Quantitative SUMO Proteomics Reveals the Modulation of Several
    www.nature.com/scientificreports OPEN Quantitative SUMO proteomics reveals the modulation of several PML nuclear body associated Received: 10 October 2017 Accepted: 28 March 2018 proteins and an anti-senescence Published: xx xx xxxx function of UBC9 Francis P. McManus1, Véronique Bourdeau2, Mariana Acevedo2, Stéphane Lopes-Paciencia2, Lian Mignacca2, Frédéric Lamoliatte1,3, John W. Rojas Pino2, Gerardo Ferbeyre2 & Pierre Thibault1,3 Several regulators of SUMOylation have been previously linked to senescence but most targets of this modifcation in senescent cells remain unidentifed. Using a two-step purifcation of a modifed SUMO3, we profled the SUMO proteome of senescent cells in a site-specifc manner. We identifed 25 SUMO sites on 23 proteins that were signifcantly regulated during senescence. Of note, most of these proteins were PML nuclear body (PML-NB) associated, which correlates with the increased number and size of PML-NBs observed in senescent cells. Interestingly, the sole SUMO E2 enzyme, UBC9, was more SUMOylated during senescence on its Lys-49. Functional studies of a UBC9 mutant at Lys-49 showed a decreased association to PML-NBs and the loss of UBC9’s ability to delay senescence. We thus propose both pro- and anti-senescence functions of protein SUMOylation. Many cellular mechanisms of defense have evolved to reduce the onset of tumors and potential cancer develop- ment. One such mechanism is cellular senescence where cells undergo cell cycle arrest in response to various stressors1,2. Multiple triggers for the onset of senescence have been documented. While replicative senescence is primarily caused in response to telomere shortening3,4, senescence can also be triggered early by a number of exogenous factors including DNA damage, elevated levels of reactive oxygen species (ROS), high cytokine signa- ling, and constitutively-active oncogenes (such as H-RAS-G12V)5,6.
    [Show full text]
  • Insights Into the Role and Mechanism of the AAA+ Adaptor Clps
    Insights into the role and mechanism of the AAA+ adaptor ClpS by Jennifer Yuan Hou Sc.B. Biochemistry Brown University, 2002 SUBMITTED TO THE DEPARTMENT OF BIOLOGY IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY AT THE MASSACHUSETTS INSTITUTE OF TECHNOLOGY JUNE 2009 © 2009 Jennifer Yuan Hou. All Rights Reserved. The author hereby grants to MIT permission to reproduce and to distribute publicly paper and electronic copies of this thesis document in whole or in part in any medium now known or hereafter created. Signature of Author:_______________________________________________________ Department of Biology May 22, 2009 Certified by:_____________________________________________________________ Tania A. Baker E. C. Whitehead Professor of Biology Thesis Supervisor Accepted by:_____________________________________________________________ Stephen P. Bell Professor of Biology Co-Chair, Graduate Committee 1 2 Insights into the role and mechanism of the AAA+ adaptor ClpS by Jennifer Yuan Hou Submitted to the Department of Biology on May 22, 2009 in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy at the Massachusetts Institute of Technology ABSTRACT Protein degradation is a vital process in cells for quality control and participation in regulatory pathways. Intracellular ATP-dependent proteases are responsible for regulated degradation and are highly controlled in their function, especially with respect to substrate selectivity. Adaptor proteins that can associate with the proteases add an additional layer of control to substrate selection. Thus, understanding the mechanism and role of adaptor proteins is a critical component to understanding how proteases choose their substrates. In this thesis, I examine the role of the intracellular protease ClpAP and its adaptor ClpS in Escherichia coli.
    [Show full text]
  • UBA2 Promotes Proliferation of Colorectal Cancer
    5552 MOLECULAR MEDICINE REPORTS 18: 5552-5562, 2018 UBA2 promotes proliferation of colorectal cancer PING HE1, XUN SUN2, HONG-JING CHENG1, YA‑BIN ZOU2, QUAN WANG3, CHANG‑LI ZHOU1, WAN-QI LIU1, YUE-MING HAO1 and XIANG-WEI MENG1 Departments of 1Gastroenterology, 2Pathology and 3Gastrointestinal Surgery, Bethune First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China Received December 18, 2017; Accepted August 13, 2018 DOI: 10.3892/mmr.2018.9613 Abstract. Small ubiquitin‑like modifier proteins are involved Introduction in tumorigenesis; however, the potential effects and func- tions of the family member ubiquitin‑like modifier‑activating Colorectal cancer is the third most common cancer world- enzyme 2 (UBA2) on colorectal cancer are not clear. The wide (1); the lifetime risk of developing colorectal cancer is present study aimed to examine the effects of UBA2 on the 4.7% for men and 4.4% for women. Although the mortality proliferation of colorectal cancer cells in vitro and in vivo. The rate from colorectal cancer has been declining for several mRNA and protein expression levels of UBA2 in patients with decades owing to the early diagnosis and improved treatment, colorectal cancer were measured by reverse transcription‑quan- >1 million novel cases are diagnosed each year. Therefore, it titative polymerase chain reaction and immunohistochemistry, is crucial to identify novel biomarkers and therapeutic targets respectively. UBA2 expression levels in colorectal cancer for colorectal cancer to improve the prognosis of the disease. tissues were significantly increased compared with the para- Sumoylation is a transient post‑translational modifica- cancerous normal tissues. The expression of UBA2 was also tion process that is highly regulated by the balance between associated with higher stage colorectal cancer and poor prog- enzyme‑mediated conjugating and deconjugating activities.
    [Show full text]
  • Proteomic Analysis of SUMO1-Sumoylome Changes During Defense 4 Elicitation in Arabidopsis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.02.233544; this version posted August 3, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Running Title: SUMO1-substrate identification in plant immunity 2 3 Proteomic analysis of SUMO1-SUMOylome changes during defense 4 elicitation in Arabidopsis 5 6 Kishor D. Ingole1,2 Shraddha K. Dahale1 and Saikat Bhattacharjee1* 7 8 1Laboratory of Signal Transduction and Plant Resistance, UNESCO-Regional Centre for Biotechnology 9 (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad- 121 10 001, Haryana, India. 11 12 2Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar- 751 024, Odisha, India. 13 14 Author for Correspondence: 15 Saikat Bhattacharjee 16 Tel: (+91) 0129-2848837 17 email: [email protected] 18 19 ORCID 20 Saikat Bhattacharjee: https://orcid.org/0000-0003-1369-7700 21 Kishor D. Ingole: https://orcid.org/0000-0003-1946-6639 22 Shraddha K. Dahale: https://orcid.org/0000-0001-8458-3984 23 24 25 26 27 28 bioRxiv preprint doi: https://doi.org/10.1101/2020.08.02.233544; this version posted August 3, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license.
    [Show full text]