The Role of BRF1 and BRF2 in the Immune Response

Total Page:16

File Type:pdf, Size:1020Kb

The Role of BRF1 and BRF2 in the Immune Response The Role of BRF1 and BRF2 in the Immune Response Rebecca Crawford A thesis submitted for the degree of Doctor of Philosophy to the Imperial College Faculty of Medicine Imperial College London September 2008 The Kennedy Institute for Rheumatology Division Faculty of Medicine Imperial College London Chapter 1 Introduction Declaration This thesis is the result of my own work. Some figures were contributed with the help of other members of the laboratory. These figures are labeled as such. The work was carried out at the Kennedy Institute of Rheumatology Division, Imperial College School of Medicine, London. 2 Chapter 1 Introduction Acknowledgements I would like to thank a number of people for their help, advice and patience over the past four years. First, my supervisor Dr. Andy Clark, who has provided continual support and advice particularly whilst writing this thesis. Secondly I would like to acknowledge the members of the lab who have provided me with ideas and technical assistance throughout my PhD. Finally, I would like to thank Alison, Tom and Liaquat without whose support and encouragement I would have struggled to reach this stage. 3 Chapter 1 Introduction Abstract Tristetraprolin (TTP), butyrate response factor 1 (BRF1) and butyrate response factor 2 (BRF2) are members of a family of zinc finger containing ARE binding proteins known as the Zfp36 family. They all possess a conserved tandem zinc finger domain, which facilitates their binding to mRNAs that contain adenosine/uridine rich elements (ARE) in their 3’ untranslated region (3’UTR). Binding to the target mRNA results in its destabilisation. Several mRNAs containing an ARE in their 3’UTR are stabilised by the mitogen activated protein kinase (MAPK) p38 pathway. Both the expression and the mRNA destabilising function of TTP are controlled by the p38 MAPK pathway. Much less is known about BRF1 and BRF2 functions, and it is not clear whether their expression or function is regulated by the p38 MAPK pathway. So far no difference has been seen in the binding specificities of the three proteins in vitro, however the phenotypes of knockout mice suggest that they have distinct functions, and may have different mRNA targets in vivo. Western blotting and quantitative PCR (qPCR) have been used to investigate the expression of members of the Zfp36 family. RNA interference was used to knock down the expression of BRF1 and BRF2 in HeLa cells, and the effects on p38-regulated inflammatory mediator expression were examined. BRF1-/- mouse embryonic fibroblast (MEF) cell lines were used to investigate the function of this family member. No evidence that BRF1 or BRF2 contributes to the post-transcriptional regulation of inflammatory mediators by the p38 MAPK pathway in HeLa cells or MEFs was found. BRF1 null MEFs over-expressed IL-6, protein, IL-6 mRNA and Cox-2 mRNA but did not over-express KC protein. The hypothesis that BRF1 is regulating IL-6 and Cox-2 by controlling mRNA stability was disproved. As a result investigation of the transcriptional regulation of these genes was researched. Primary transcript qPCR showed that both IL-6 and Cox-2 under-go more rapid transcription in BRF1-/- MEFs. This suggests that IL-6 and Cox-2 are indirect targets of BRF1 and that their regulation is through a transcription factor which is itself a target of BRF1. 4 Chapter 1 Introduction Contents Title page………………………………………………………………………………….....1 Declaration…………………………………………………………………………………..2 Acknowledgements……………………………………………………………………….....3 Abstract…………………………………………………………………………………...…4 Table of contents…………………………………………………………………………….5 List of figures and tables…………………………………………………………………….9 1) INTRODUCTION .....................................................................22 1.1) The immune system.................................................................................................. 22 1.1.1) Introduction........................................................................................................ 22 1.1.2) Innate Immunity and the inflammatory response- the initial response to infection ....................................................................................................................... 22 1.1.3) Leukocytes......................................................................................................... 23 1.1.4) Fibroblasts.......................................................................................................... 24 1.1.5) Cytokines ........................................................................................................... 25 1.1.5.1) Interleukin-1 (IL-1)..................................................................................... 26 1.1.5.2) Interleukin -6 (IL-6) and its receptor .......................................................... 27 1.1.5.3) Cyclooxygenase-1/2 (Cox-1/2)....................................................................... 29 1.1.5.4) Chemokine (CXC motif) family: Interleukin-8 (IL-8), growth-regulated oncogene (GRO)α and keratinocyte chemokine (KC)................................................. 31 1.2) Inflammatory signalling pathways ........................................................................... 33 1.2.1) Introduction........................................................................................................ 33 1.2.2) Interleukin-1 receptor (IL-1R) and the toll-like receptor (TLR) superfamily ... 33 1.2.3) IL-1 Signalling................................................................................................... 34 1.2.4) TLR Signalling .................................................................................................. 34 1.2.5) The mitogen activated protein kinase (MAPK) pathway .................................. 40 1.2.5.1) Extra cellular regulated kinase (ERK) pathway ......................................... 42 1.2.5.2) MAPKp38 Pathway .................................................................................... 43 1.2.5.3) The c- jun amino terminal kinase (JNK) pathway...................................... 45 1.2.6) The Phosphoinositide 3-kinase (PI3K) Pathway ............................................... 46 1.3) Gene Expression ....................................................................................................... 47 1.3.1) Introduction........................................................................................................ 47 1.3.2) Transcription...................................................................................................... 48 1.3.2.1) Chromatin structure .................................................................................... 48 1.3.2.2) Chromatin modifications ............................................................................ 48 1.3.2.3) Transcription activation and repression...................................................... 49 1.3.2.4) Initiation of transcription ............................................................................ 50 1.3.2.5) Elongation................................................................................................... 51 1.3.3) mRNA processing and translation..................................................................... 53 1.3.3.1) Introduction................................................................................................. 53 5 Chapter 1 Introduction 1.3.3.2) mRNA regulation- an important mechanism in the control of gene expression................................................................................................................. 55 1.3.3.3) mRNA decay .............................................................................................. 56 1.3.3.4) Processing bodies (PBs) and stress granules (SGs).................................... 58 1.3.3.5) mRNA surveillance: Nonsensense-mediated decay (NMD) ...................... 60 1.3.3.6) RNA interference (RNAi) pathway ............................................................ 61 1.3.4) Adenosine/ uridine rich element (ARE) mediated decay (AMD) ..................... 62 1.3.4.1) Introduction................................................................................................. 62 1.3.4.2) Classification of AREs ............................................................................... 63 1.3.5) Signalling pathways and mRNA decay ............................................................. 64 1.3.5.1) The p38 pathway and mRNA stabilisation................................................. 65 1.3.5.2) Mechanism of p38 mRNA mediated stabilisation...................................... 66 1.4) Adenosine/uridine rich element binding proteins (AUBPs)..................................... 68 1.4.1) Introduction........................................................................................................ 68 1.4.1.1) Human antigen R (HuR)............................................................................. 68 1.4.1.2) Heterogeneous ribonucleoprotein (hnRNP)................................................ 71 1.4.1.3) T cell –restricted antigen (TIA)-1 and TIA related protein (TIAR) ........... 74 1.4.1.4) The far upstream sequence element binding protein (FBP) family............ 76 1.4.1.5) The zinc finger 36 (Zfp36) family .............................................................. 79 1.4.1.5.1) Introduction.......................................................................................... 79 1.4.1.5.2) The tandem zinc finger (TZF) domain ...............................................
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]
  • The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid
    The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid Leukemia Cells Supported Only by Exogenous Cytokines Is Associated with a Patient Subset with Adverse Outcome Annette K. Brenner, Elise Aasebø, Maria Hernandez-Valladares, Frode Selheim, Frode Berven, Ida-Sofie Grønningsæter, Sushma Bartaula-Brevik and Øystein Bruserud Supplementary Material S2 of S31 Table S1. Detailed information about the 68 AML patients included in the study. # of blasts Viability Proliferation Cytokine Viable cells Change in ID Gender Age Etiology FAB Cytogenetics Mutations CD34 Colonies (109/L) (%) 48 h (cpm) secretion (106) 5 weeks phenotype 1 M 42 de novo 241 M2 normal Flt3 pos 31.0 3848 low 0.24 7 yes 2 M 82 MF 12.4 M2 t(9;22) wt pos 81.6 74,686 low 1.43 969 yes 3 F 49 CML/relapse 149 M2 complex n.d. pos 26.2 3472 low 0.08 n.d. no 4 M 33 de novo 62.0 M2 normal wt pos 67.5 6206 low 0.08 6.5 no 5 M 71 relapse 91.0 M4 normal NPM1 pos 63.5 21,331 low 0.17 n.d. yes 6 M 83 de novo 109 M1 n.d. wt pos 19.1 8764 low 1.65 693 no 7 F 77 MDS 26.4 M1 normal wt pos 89.4 53,799 high 3.43 2746 no 8 M 46 de novo 26.9 M1 normal NPM1 n.d. n.d. 3472 low 1.56 n.d. no 9 M 68 MF 50.8 M4 normal D835 pos 69.4 1640 low 0.08 n.d.
    [Show full text]
  • Role and Regulation of the P53-Homolog P73 in the Transformation of Normal Human Fibroblasts
    Role and regulation of the p53-homolog p73 in the transformation of normal human fibroblasts Dissertation zur Erlangung des naturwissenschaftlichen Doktorgrades der Bayerischen Julius-Maximilians-Universität Würzburg vorgelegt von Lars Hofmann aus Aschaffenburg Würzburg 2007 Eingereicht am Mitglieder der Promotionskommission: Vorsitzender: Prof. Dr. Dr. Martin J. Müller Gutachter: Prof. Dr. Michael P. Schön Gutachter : Prof. Dr. Georg Krohne Tag des Promotionskolloquiums: Doktorurkunde ausgehändigt am Erklärung Hiermit erkläre ich, dass ich die vorliegende Arbeit selbständig angefertigt und keine anderen als die angegebenen Hilfsmittel und Quellen verwendet habe. Diese Arbeit wurde weder in gleicher noch in ähnlicher Form in einem anderen Prüfungsverfahren vorgelegt. Ich habe früher, außer den mit dem Zulassungsgesuch urkundlichen Graden, keine weiteren akademischen Grade erworben und zu erwerben gesucht. Würzburg, Lars Hofmann Content SUMMARY ................................................................................................................ IV ZUSAMMENFASSUNG ............................................................................................. V 1. INTRODUCTION ................................................................................................. 1 1.1. Molecular basics of cancer .......................................................................................... 1 1.2. Early research on tumorigenesis ................................................................................. 3 1.3. Developing
    [Show full text]
  • Cellular Senescence Bypass Screen Identifies New Putative Tumor
    Oncogene (2008) 27, 1961–1970 & 2008 Nature Publishing Group All rights reserved 0950-9232/08 $30.00 www.nature.com/onc ORIGINAL ARTICLE Cellular senescence bypass screen identifies new putative tumor suppressor genes JFM Leal1, J Fominaya1, A Casco´ n2, MV Guijarro1, C Blanco-Aparicio1, M Lleonart3, ME Castro1, SRamon y Cajal 3, M Robledo2, DH Beach4 and A Carnero1 1Experimental Therapeutics Programme, Centro Nacional de Investigaciones Oncolo´gicas (CNIO), Madrid, Spain; 2Molecular Pathology Programme, Centro Nacional de Investigaciones Oncolo´gicas (CNIO), Madrid, Spain; 3Departamento de Patologı´a, Hospital Vall d’Hebron, Barcelona, Spain and 4Institute for Cell and Molecular Sciences, Center for Cutaneous Biology, London, UK Senescence is a mechanism that limits cellular lifespan and Introduction constitutes a barrier against cellular immortalization. To identify new senescence regulatory genes that might play a Replicative senescence is characterized by a progressive role in tumorigenesis, we have designed and performed a loss of proliferative potential with the increase of popu- large-scale antisense-based genetic screen in primary lation doublings, resulting in a permanent and irrever- mouse embryo fibroblasts (MEFs). Out of this screen, sible cell-cycle arrest. Although the process of senescence we have identified five different genes through which loss occurs both in vitro and in vivo (Dimri et al., 1995; of function partially bypasses senescence. These genes Schmitt et al., 2002; Shay and Roninson, 2004; Braig belong to very different biochemical families: csn2 et al., 2005; Collado et al., 2005; Michaloglou et al., (component of the Cop9 signalosome), aldose reductase 2005), the transition to the senescent phenotype is (a metabolic enzyme) and brf1 (subunit of the RNA commonly studied in culture where a cell population polymerase II complex), S-adenosyl homocysteine hydro- can be grown and monitored.
    [Show full text]
  • BRF1 Mutations Alter RNA Polymerase III–Dependent Transcription and Cause Neurodevelopmental Anomalies
    Downloaded from genome.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press Research BRF1 mutations alter RNA polymerase III–dependent transcription and cause neurodevelopmental anomalies Guntram Borck,1,20 Friederike Hog,€ 2,20 Maria Lisa Dentici,3,20 Perciliz L. Tan,4,20 Nadine Sowada,1 Ana Medeira,5 Lucie Gueneau,6 Holger Thiele,7 Maria Kousi,4 Francesca Lepri,3 Larissa Wenzeck,2 Ian Blumenthal,8 Antonio Radicioni,9 Tito Livio Schwarzenberg,10 Barbara Mandriani,11,12 Rita Fischetto,13 Deborah J. Morris-Rosendahl,14 Janine Altmuller,€ 7,15 Alexandre Reymond,6 Peter Nurnberg,€ 7,16,17 Giuseppe Merla,11 Bruno Dallapiccola,3,21 Nicholas Katsanis,4,21 Patrick Cramer,18,21 and Christian Kubisch1,19,21 1Institute of Human Genetics, University of Ulm, 89081 Ulm, Germany; 2Gene Center Munich and Department of Biochemistry, Center for Integrated Protein Science CIPSM, Ludwig-Maximilians-Universitat€ Munchen,€ 81377 Munich, Germany; 3Bambino GesuChildren’s Hospital, IRCCS, 00165 Rome, Italy; 4Center for Human Disease Modeling, Duke University, Durham, North Carolina 27710, USA; 5Servic¸o de Genetica, Departamento de Pediatria, Hospital S. Maria, CHLN, 1649-035 Lisboa, Portugal; 6Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; 7Cologne Center for Genomics (CCG), University of Cologne, 50931 Cologne, Germany; 8Molecular Neurogenetics Unit and Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; 9Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy; 10Department of Neonatology, Sapienza University, 00161 Rome, Italy; 11IRCCS Casa Sollievo Della Sofferenza, Medical Genetics Unit, 71013 San Giovanni Rotondo, Italy; 12PhD Program, Molecular Genetics applied to Medical Sciences, University of Brescia, 25121 Brescia, Italy; 13U.O.
    [Show full text]
  • BRF1 Antibody (Center) Affinity Purified Rabbit Polyclonal Antibody (Pab) Catalog # Ap14801c
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 BRF1 Antibody (Center) Affinity Purified Rabbit Polyclonal Antibody (Pab) Catalog # AP14801c Specification BRF1 Antibody (Center) - Product Information Application WB,E Primary Accession Q92994 Other Accession NP_663718.1, NP_001510.2 Reactivity Human Host Rabbit Clonality Polyclonal Isotype Rabbit Ig Calculated MW 73840 Antigen Region 314-342 BRF1 Antibody (Center) - Additional Information BRF1 Antibody (Center) (Cat. #AP14801c) Gene ID 2972 western blot analysis in Hela cell line lysates (35ug/lane).This demonstrates the BRF1 Other Names antibody detected the BRF1 protein (arrow). Transcription factor IIIB 90 kDa subunit, TFIIIB90, hTFIIIB90, B-related factor 1, BRF-1, hBRF, TAF3B2, TATA box-binding BRF1 Antibody (Center) - Background protein-associated factor, RNA polymerase III, subunit 2, BRF1, BRF, GTF3B, TAF3B2, This gene encodes one of the three subunits TAF3C of the RNA polymerase III transcription factor complex. Target/Specificity This BRF1 antibody is generated from This complex plays a rabbits immunized with a KLH conjugated central role in transcription initiation by RNA synthetic peptide between 314-342 amino polymerase III on acids from the Central region of human genes encoding tRNA, 5S rRNA, and other BRF1. small structural RNAs. The gene product belongs to the TF2B family. Two Dilution alternatively spliced WB~~1:1000 variants encoding different isoforms, that function at different Format promoters transcribed by RNA polymerase III, Purified polyclonal antibody supplied in PBS have been identified. with 0.09% (W/V) sodium azide. This Other transcript variants are possible, but their antibody is purified through a protein A full-length column, followed by peptide affinity natures have not been completely purification.
    [Show full text]
  • Structural Basis of Ty3 Retrotransposon Integration at RNA Polymerase III-Transcribed Genes
    bioRxiv preprint doi: https://doi.org/10.1101/2021.07.27.453949; this version posted July 27, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Structural basis of Ty3 retrotransposon integration at RNA Polymerase III-transcribed genes Guillermo Abascal-Palacios1*, Laura Jochem1, Carlos Pla-Prats2, Fabienne Beuron1 and Alessandro Vannini1,3* Affiliations: 1Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom. 2Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland. 3Fondazione Human Technopole, Structural Biology Research Centre, 20157 Milan, Italy *Corresponding authors ABSTRACT Retrotransposons are endogenous elements that have the ability to mobilise their DNA and integrate at different locations in the host genome. In budding yeast, the Ty3 retrotransposon integrates with an exquisite specificity in a narrow window upstream of RNA Polymerase III-transcribed genes, such as the genes of transfer RNAs, representing a paradigm for specific targeted integration. Here we present the cryo-EM reconstruction at 4.0 Å-resolution of an active Ty3 strand-transfer complex (Ty3 intasome) caught in the act of integrating onto a specific tRNA gene bound to the RNA Polymerase III general transcription factor TFIIIB, which is required for Ty3 specific targeting. The structure unravels the molecular mechanisms underlying Ty3 integration specificity at RNA Polymerase III-transcribed genes and sheds light into the architecture of a retrotransposon integration machinery during the process of strand transfer at a genomic locus. The Ty3 intasome establishes contacts with a region of the TATA-binding protein (TBP), a subunit of TFIIIB, which is blocked by the ubiquitous transcription regulator negative cofactor 2 (NC2) in RNA Pol II-transcribed genes.
    [Show full text]
  • BRF1/2 Antibody A
    C 0 2 - t BRF1/2 Antibody a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 9 1 Web: [email protected] 1 www.cellsignal.com 2 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source: UniProt ID: Entrez-Gene Id: WB H M R Mk Endogenous 40 to 50, 62 Rabbit P47974, Q07352 678, 677 Product Usage Information 7. Nair, A.P. et al. (1994) Nature 369, 239-242. 8. Carballo, E. et al. (1998) Science 281, 1001-1005. Application Dilution 9. Schmidlin, M. et al. (2004) EMBO J. 23, 4760-4769. Western Blotting 1:1000 Storage Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA and 50% glycerol. Store at –20°C. Do not aliquot the antibody. Specificity / Sensitivity This antibody detects endogenous levels of total BRF1 and BRF2 proteins. Species Reactivity: Human, Mouse, Rat, Monkey Species predicted to react based on 100% sequence homology: Chicken, Bovine Source / Purification Polyclonal antibodies are produced by immunizing animals with a synthetic peptide corresponding to the carboxy terminus of human BRF1 protein. Antibodies are purified by protein A and peptide affinity chromatography. Background Butyrate response factor 1 (BRF1; also known as EGF response factor 1 [ERF1], TIS11B, ZFP36L1) and butyrate response factor 2 (BRF2; also known as EGF response factor 2 [ERF2], TIS11D, ZFP36L2) both belong to the TIS11 family of CCCH zinc-finger proteins (1). This family of proteins, which also includes tristetraprolin (TTP), bind to AU- rich elements (ARE) found in the 3'-untranslated regions of mRNAs and promote de- adenylation and rapid degradation by the exosome (2,3).
    [Show full text]
  • RNA Polymerase III Subunit Architecture and Implications for Open Promoter Complex Formation
    RNA polymerase III subunit architecture and implications for open promoter complex formation Chih-Chien Wua,b, Franz Herzogc, Stefan Jennebachd, Yu-Chun Lina, Chih-Yu Paia, Ruedi Aebersoldc,e, Patrick Cramerd, and Hung-Ta Chena,1 aInstitute of Molecular Biology, Academia Sinica, Taipei, Taiwan 115, Republic of China; bDepartment of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan 112, Republic of China; cDepartment of Biology, Institute of Molecular Systems Biology, Eidgenössiche Technische Hochschule Zurich, CH-8093 Zurich, Switzerland; dGene Center and Department of Biochemistry, Center for Integrated Protein Science Munich, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; and eFaculty of Science, University of Zurich, CH-8006 Zurich, Switzerland Edited by E. Peter Geiduschek, University of California at San Diego, La Jolla, CA, and approved October 3, 2012 (received for review July 11, 2012) Transcription initiation by eukaryotic RNA polymerase (Pol) III relies subcomplexes and their contacts with the 12-subunit polymerase on the TFIIE-related subcomplex C82/34/31. Here we combine cross- core. The C37/53 dimerization module was positioned into the linking and hydroxyl radical probing to position the C82/34/31 electron density adjacent to the lobe domain of the C128 subunit subcomplex around the Pol III active center cleft. The extended on one side of the polymerase cleft, similar to the localization of winged helix (WH) domains 1 and 4 of C82 localize to the polymerase the TFIIF dimerization module and the A49/34.5 dimerization domains clamp head and clamp core, respectively, and the two WH module on Pol II and Pol I, respectively (23–25).
    [Show full text]
  • Fusions of Yeast TATA-Binding Protein with Brf1, and the Function of Transcription Factor IIIB
    Reconfiguring the connectivity of a multiprotein complex: Fusions of yeast TATA-binding protein with Brf1, and the function of transcription factor IIIB George A. Kassavetis*, Elisabetta Soragni, Robert Driscoll†, and E. Peter Geiduschek Division of Biological Sciences, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0634 Contributed by E. Peter Geiduschek, September 1, 2005 Transcription factor (TF) IIIB, the central transcription initiation transcription initiation factors bound to a proximal promoter factor of RNA polymerase III (pol III), is composed of three subunits, element and distal enhancer-like site (reviewed in ref. 4). Bdp1, Brf1 and TATA-binding protein (TBP), all essential for normal Brf1 is a hybrid protein: Its N-terminal half is related to TFIIB, function in vivo and in vitro. Brf1 is a modular protein: Its N- whereas the C-terminal half has no counterpart in the pol II or proximal half is related to TFIIB and binds similarly to the C- pol I transcription apparatus. A conserved segment in the terminal stirrup of TBP; its C-proximal one-third provides most of C-terminal one-third of Brf1 contributes most of its affinity for the affinity for TBP by binding along the entire length of the TBP. An additional, weaker site is located in the TFIIB-related convex surface and N-terminal lateral face of TBP. A structure- N-terminal half (5–8). The C-proximal one-third of Brf1 also informed triple fusion protein, with TBP core placed between the provides the primary binding site for Bdp1, but an additional N- and C-proximal domains of Brf1, has been constructed.
    [Show full text]
  • TBP, a Universal Eukaryotic Transcription Factor?
    Downloaded from genesdev.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW TBP, a universal eukaryotic transcription factor? Nouria Hernandez Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11742 In eukaryotes, transcription is carried out by three dif- cDNAs from several other species, including Drosophila ferent RNA polymerases, RNA polymerases I, II, and III, (Hoey et al. 1990; Muhich et al. 1990) and humans (Ho- each of which is dedicated to the transcription of differ- ffman et al. 1990b; Kao et al. 1990; Peterson et al. 1990). ent sets of genes. The genes in each class contain char- Like the yeast protein, the Drosophila and human TBPs acteristic promoters, which often consist of two types of are small proteins of 38 kD, a surprising result because elements: the basal promoter elements and the modula- the molecular mass of the mammalian TATA box-bind- tor promoter elements. The basal promoter elements are ing factor TFIID was known to be -750 kD (Nakajima et sufficient to determine RNA polymerase specificity and al. 1988; Conaway et al. 1990, 1991). In addition, like direct low levels of transcription, whereas the modulator TFIID, these TBPs could mediate basal RNA polymerase elements enhance or reduce the basal levels of transcrip- II transcription, but unlike TFIID, they could not re- tion. None of the RNA polymerases can recognize its spond to transcriptional activators (Hoey et al. 1990; Ho- target promoters directly. Instead, basal promoter ele- ffman et al. 1990b; Peterson et al. 1990; Pugh and Tjian ments are first recognized by specific transcription fac- 1990; Smale et al.
    [Show full text]
  • The Jnks Differentially Regulate RNA Polymerase III Transcription by Coordinately Modulating the Expression of All TFIIIB Subunits
    The JNKs differentially regulate RNA polymerase III transcription by coordinately modulating the expression of all TFIIIB subunits Shuping Zhong and Deborah L. Johnson1 Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, and the Norris Comprehensive Cancer Center, 2011 Zonal Avenue, Los Angeles, CA 90033 Edited by Robert G. Roeder, The Rockefeller University, New York, NY, and approved June 12, 2009 (received for review May 4, 2009) RNA polymerase (pol) III-dependent transcription is subject to strin- III transcription has been extensively observed in transformed and gent regulation by tumor suppressors and oncogenic proteins and tumor cells supporting the idea that it plays a crucial role in enhanced RNA pol III transcription is essential for cellular transfor- tumorigenesis (11). Recent studies have demonstrated that en- mation and tumorigenesis. Since the c-Jun N-terminal kinases (JNKs) hanced RNA pol III transcription is required for promoting onco- display both oncogenic and tumor suppressor properties, the roles of genic transformation (12, 13). Thus, understanding the molecular these proteins in regulating RNA pol III transcription were examined. pathways by which this transcription process is controlled may In both mouse and human cells, loss or reduction in JNK1 expression provide useful therapeutic approaches for cancer. represses RNA pol III transcription. In contrast, loss or reduction in The TATA binding protein (TBP) associates with specific JNK2 expression induces transcription. The JNKs coordinately regu- proteins, TBP-associated factors (TAFs), to form at least 3 late expression of all 3 TFIIIB subunits. While JNK1 positively regulates distinct complexes, SL1, TFIID, and TFIIIB, which specify the TBP expression, the RNA pol III-specific factors, Brf1 and Bdp1, JNK2 role of TBP in the expression of genes directed by RNA pol I, negatively regulates their expression.
    [Show full text]