HSP90 and Its R2TP/Prefoldin-Like Cochaperone Are Involved in the Cytoplasmic Assembly of RNA Polymerase II

Total Page:16

File Type:pdf, Size:1020Kb

HSP90 and Its R2TP/Prefoldin-Like Cochaperone Are Involved in the Cytoplasmic Assembly of RNA Polymerase II Molecular Cell Article HSP90 and Its R2TP/Prefoldin-like Cochaperone Are Involved in the Cytoplasmic Assembly of RNA Polymerase II Se´ verine Boulon,1,5,6 Be´ renge` re Pradet-Balade,2,3,4,5 Ce´ line Verheggen,2,3,4,5 Dorothe´ e Molle,2,3,4,5 Ste´ phanie Boireau,2,3,4 Marya Georgieva,2,3,4 Karim Azzag,2,3,4 Marie-Ce´ cile Robert,2,3,4 Yasmeen Ahmad,1 Henry Neel,2,3 Angus I. Lamond,1 and Edouard Bertrand2,3,4,* 1Wellcome Trust Centre for Gene Regulation and Expression, University of Dundee, Dundee DD1 5EH, UK 2Institut de Ge´ ne´ tique Mole´ culaire de Montpellier - UMR 5535 CNRS, Universite´ Montpellier 2, 34293 Montpellier Cedex 5, France 3Universite´ Montpellier 2, 34095 Montpellier Cedex 5, France 4Universite´ Montpellier 1, 34060 Montpellier Cedex 2, France 5These authors contributed equally to this work 6Present address: Centre de Recherche en Biochimie Macromole´ culaire - UMR 5237, 34294 Montpellier Cedex 5, France *Correspondence: [email protected] DOI 10.1016/j.molcel.2010.08.023 SUMMARY The activities, structures, and subunit composition of the three major RNA polymerases have been characterized in detail RNA polymerases are key multisubunit cellular (Cramer et al., 2008). RNA polymerases I, II, and III are enzymes. Microscopy studies indicated that RNA composed of 14, 12, and 17 subunits, respectively. The two polymerase I assembles near its promoter. However, largest subunits form the catalytic core of the enzyme, while the mechanism by which RNA polymerase II is the others are smaller and generally bind on their surface. The assembled from its 12 subunits remains unclear. three RNA polymerases are related to each other, and this struc- We show here that RNA polymerase II subunits tural similarity is also highlighted by the fact that some subunits are shared by several polymerases, with a few being present in Rpb1 and Rpb3 accumulate in the cytoplasm when all three enzymes. Despite intensive studies on the structure assembly is prevented and that nuclear import of and regulation of RNA polymerases, relatively little is known Rpb1 requires the presence of all subunits. Using about the location and mechanism of their assembly. To date, MS-based quantitative proteomics, we character- this question has been principally addressed by live-cell micros- ized assembly intermediates. These included a cyto- copy techniques that used GFP-tagged subunits in mammalian plasmic complex containing subunits Rpb1 and cells. FRAP (fluorescence recovery after photobleaching) Rpb8 associated with the HSP90 cochaperone studies on RNA polymerase I have indicated that some subunits hSpagh (RPAP3) and the R2TP/Prefoldin-like com- can either assemble or exchange directly at promoters (Dundr plex. Remarkably, HSP90 activity stabilized incom- et al., 2002). Furthermore, the exchange/assembly rate depends pletely assembled Rpb1 in the cytoplasm. Our data on the phase of the cell cycle, indicating that assembly of RNA indicate that RNA polymerase II is built in the cyto- polymerase I in the nucleolus is a way to regulate gene expres- sion (Gorski et al., 2008). Live-cell studies on RNA polymerase plasm and reveal quality-control mechanisms that II promoters have also revealed a rapid exchange of basal and link HSP90 to the nuclear import of fully assembled sequence-specific transcription factors at promoters. In yeast, enzymes. hSpagh also bound the free RPA194 the TATA binding factor TBP and the transcriptional activator subunit of RNA polymerase I, suggesting a general Ace1p were shown to rapidly come on and off the chromatin, role in assembling RNA polymerases. with a residency time in the range of seconds (Karpova et al., 2008; Sprouse et al., 2008). Moreover, the rapidly cycling Ace1p molecules were shown to be responsible for transcrip- INTRODUCTION tional activation. In mammalian cells, glucocorticoid and estra- diol receptors were shown to have similarly high exchange rates RNA polymerases play fundamental roles in the cell. RNA poly- with their target sites on DNA (McNally et al., 2000; Stenoien merases I and III synthesize the noncoding RNAs that form the et al., 2001). More recently, the use of Drosophila salivary glands translational apparatus, and their activity is intimately linked to with polytene chromosomes and the generation of human cell the growth state of the cell (White, 2005). RNA polymerase II lines carrying artificial arrays of reporter genes have allowed synthesizes capped noncoding RNAs as well as all mRNAs. detailed studies of transcription in living cells (Janicki et al., This enzyme is at the heart of gene regulation and is subjected 2004; Yao et al., 2006). In particular, FRAP studies of GFP- to many controls, including at the level of initiation, elongation, tagged subunits of RNA polymerase II were used to define the and termination (Fuda et al., 2009). dynamics of their binding to promoters and the transcription 912 Molecular Cell 39, 912–924, September 24, 2010 ª2010 Elsevier Inc. Molecular Cell HSP90, R2TP, and RNA Polymerase II Assembly kinetics of this key enzyme. In two studies that used HIV-1 a specific inhibitor of the exportin CRM1 (Fornerod et al., 1997). reporters or natural Drosophila genes, an efficient initiation entry This resulted in the retention of GFP-Rpb3 in the nucleus mode was found (Boireau et al., 2007; Yao et al., 2007). In (Figure 1A), indicating that an active export mechanism removes contrast, a study that used a Tet-inducible promoter in human GFP-Rpb3 from nuclei when Rpb1 is degraded. cells found that a large fraction of the polymerase exchanges Surprisingly, LMB prevented nuclear import of Rpb1 rapidly at promoters, with only a few percent that go into produc- (Figure 1A) and made this subunit insensitive to a-amanitin tive elongation (Darzacq et al., 2007). Given the precedent (Figures 1A and S1). Because a-amanitin specifically promotes example of RNA polymerase I assembly at its promoter, these degradation of the elongating form of RNA polymerase II data led to the suggestion that RNA polymerase II may also (Nguyen et al., 1996), the cytoplasmic accumulation of Rpb1 assemble at its promoter and that this could allow a gene- most likely represents newly synthesized subunits that were specific regulation of its assembly (Darzacq and Singer, 2008; never engaged in transcription elongation. These data suggest Hager et al., 2009). that factors required for Rpb1 nuclear import may be retained Recently, affinity purification of soluble human RNA poly- in the nucleus upon LMB treatment. Since Rpb3 remains nuclear merase II with TAP-tagged subunits identified a number of poly- in these conditions, an intriguing possibility would be that Rpb1 merase-associated factors of unknown function (Jeronimo et al., needs to be incorporated in fully assembled enzymes before 2004, 2007). Four of these factors are homologous to the yeast being imported into the nucleus. R2TP complex (Boulon et al., 2008; Zhao et al., 2005), and further To test this hypothesis, RNA polymerase II assembly was pre- studies suggested that they indeed form a complex that contains vented by inhibiting de novo synthesis of individual subunits with the R2TP factors plus five prefoldin-like proteins (referred to as siRNAs. Depletion of Rpb2, the second largest subunit, pre- R2TP/Prefoldin-like) (Boulon et al., 2008; Cloutier et al., 2009; vented nuclear import of Rpb1 (Figure 1B). The pool of Rpb1 Sardiu et al., 2008). The yeast R2TP protein Tah1 was initially accumulated in the cytoplasm was resistant to a-amanitin, indi- characterized as a cochaperone for HSP90 (Zhao et al., 2005) cating that it corresponds to newly synthesized proteins that and, in agreement, we recently showed that the human Tah1 were never engaged in elongation. Furthermore, analysis with homolog hSpagh (also called RPAP3 or FLJ21908) is an a panel of antibodies that recognize various phosphoisoforms HSP90 cofactor (Boulon et al., 2008). Furthermore, HSP90 and of the CTD heptad-repeat indicated that the cytoplasmic Rpb1 R2TP proteins, including hSpagh, play a key role during the is mostly unphosphorylated (see Supplemental Experimental assembly of snoRNPs (Boulon et al., 2008; Gonzales et al., Procedures and Figure S1). This effect on Rpb1 caused by 2005; King et al., 2001; Zhao et al., 2008), raising the possibility siRNA-mediated depletion of Rpb2 was not an indirect conse- that the association of hSpagh with RNA polymerase II subunits quence of transcription inhibition. Indeed, the transcription inhib- could be involved in the assembly of this enzyme. itors DRB and actinomycin D, which do not trigger subunit In this study, we used a combination of quantitative MS-based degradation (Nguyen et al., 1996), did not induce a cytoplasmic proteomics and fluorescence microscopy to characterize the accumulation of Rpb1 (Figures 1C and S1). These results were mechanism of RNA polymerase II assembly in human cells and extended by examining siRNA-mediated removal of other RNA the role of hSpagh and Hsp90 in this process. polymerase II subunits. Remarkably, depletion of any subunit, including Rpb3, resulted in a similar accumulation of Rpb1 in RESULTS the cytoplasm (Figures 1D and S1, and see below for quantitative measurements of Rpb1 localization). Together with the fact that Unassembled RNA Polymerase II Subunits Accumulate degradation of elongating Rpb1 by a-amanitin triggered nuclear in the Cytoplasm export of Rpb3, these data suggested that assembly of RNA To study assembly of RNA polymerase II, we took advantage of polymerase II occurs in the cytoplasm and is an obligatory a-amanitin. This drug binds the large Rpb1 subunit of human step prior to nuclear import. RNA polymerase II with high affinity. In vitro, this reduces the elongation rate more than 100-fold and makes the polymerase GFP-Tagged Subunits Rpb1 and Rpb3 Have Similar prone to essentially irreversible stalling (Rudd and Luse, 1996). Dynamics at the Transcription Site of an HIV-1 Reporter In vivo, this results in transcriptional arrest and concomitant Gene destruction of subunit Rpb1.
Recommended publications
  • Structure of the Molecular Chaperone Prefoldin
    CORE Metadata, citation and similar papers at core.ac.uk Provided by Elsevier - Publisher Connector Cell, Vol. 103, 621±632, November 10, 2000, Copyright 2000 by Cell Press Structure of the Molecular Chaperone Prefoldin: Unique Interaction of Multiple Coiled Coil Tentacles with Unfolded Proteins of newly synthesized bacterial %10ف ,Ralf Siegert,² Michel R. Leroux,²³ 1999). In addition Clemens Scheufler, F. Ulrich Hartl,* proteins complete their folding in the sequestered envi- and Ismail Moarefi* ronment provided by GroEL/GroES (Horwich et al., 1993; Max-Planck Institut fuÈ r Biochemie Ewalt et al., 1997; Houry et al., 1999). The eukaryotic Am Klopferspitz 18a Hsp70 chaperone machine also binds nascent chains D82152 Martinsried (Beckmann et al., 1990; Nelson et al., 1992; Eggers et Germany al., 1997; Thulasiraman et al., 1999), and some proteins, including actins and tubulins, depend on the Group II cytosolic chaperonin TRiC (TCP-1 ring Complex; also Summary termed CCT) for folding (Frydman et al., 1992; Gao et al., 1992; Yaffe et al., 1992; Kubota et al., 1995; Siegers Prefoldin (GimC) is a hexameric molecular chaperone et al., 1999). complex built from two related classes of subunits The archaeal Group II chaperonin (thermosome) is and present in all eukaryotes and archaea. Prefoldin closely related to its eukaryotic homologue TRiC (Gutsche et al., 1999). In contrast, Hsp70 proteins and interacts with nascent polypeptide chains and, in vitro, TF are generally missing from the archaeal kingdom can functionally substitute for the Hsp70 chaperone though some archaea have acquired Hsp70, presumably system in stabilizing non-native proteins for subse- by lateral gene transfer (Gribaldo et al., 1999).
    [Show full text]
  • Chaperonin-Assisted Protein Folding: a Chronologue
    Quarterly Reviews of Chaperonin-assisted protein folding: Biophysics a chronologue cambridge.org/qrb Arthur L. Horwich1,2 and Wayne A. Fenton2 1Howard Hughes Medical Institute, Yale School of Medicine, Boyer Center, 295 Congress Avenue, New Haven, CT 06510, USA and 2Department of Genetics, Yale School of Medicine, Boyer Center, 295 Congress Avenue, New Invited Review Haven, CT 06510, USA Cite this article: Horwich AL, Fenton WA (2020). Chaperonin-assisted protein folding: a Abstract chronologue. Quarterly Reviews of Biophysics This chronologue seeks to document the discovery and development of an understanding of – 53, e4, 1 127. https://doi.org/10.1017/ oligomeric ring protein assemblies known as chaperonins that assist protein folding in the cell. S0033583519000143 It provides detail regarding genetic, physiologic, biochemical, and biophysical studies of these Received: 16 August 2019 ATP-utilizing machines from both in vivo and in vitro observations. The chronologue is orga- Revised: 21 November 2019 nized into various topics of physiology and mechanism, for each of which a chronologic order Accepted: 26 November 2019 is generally followed. The text is liberally illustrated to provide firsthand inspection of the key Key words: pieces of experimental data that propelled this field. Because of the length and depth of this Chaperonin; GroEL; GroES; Hsp60; protein piece, the use of the outline as a guide for selected reading is encouraged, but it should also be folding of help in pursuing the text in direct order. Author for correspondence: Arthur L. Horwich, E-mail: [email protected] Table of contents I. Foundational discovery of Anfinsen and coworkers – the amino acid sequence of a polypeptide contains all of the information required for folding to the native state 7 II.
    [Show full text]
  • Structural and Functional Dissection of Reovirus Capsid Folding and Assembly by the Prefoldin-Tric/CCT Chaperone Network
    Structural and functional dissection of reovirus capsid folding and assembly by the prefoldin-TRiC/CCT chaperone network Jonathan J. Knowltona,b,1, Daniel Gestautc,1, Boxue Mad,e,f,2, Gwen Taylora,g,2, Alpay Burak Sevenh,i, Alexander Leitnerj, Gregory J. Wilsonk, Sreejesh Shankerl, Nathan A. Yatesm, B. V. Venkataram Prasadl, Ruedi Aebersoldj,n, Wah Chiud,e,f, Judith Frydmanc,3, and Terence S. Dermodya,g,o,3 aDepartment of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224; bDepartment of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232; cDepartment of Biology, Stanford University, Stanford, CA 94305; dDepartment of Bioengineering, Stanford University, Stanford, CA 94305; eDepartment of Microbiology and Immunology, Stanford University, Stanford, CA 94305; fDepartment of Photon Science, Stanford University, Stanford, CA 94305; gCenter for Microbial Pathogenesis, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224; hDepartment of Structural Biology, Stanford University, Stanford, CA 94305; iDepartment of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94305; jDepartment of Biology, Institute of Molecular Systems Biology, ETH Zürich, 8093 Zürich, Switzerland; kDepartment of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232; lVerna and Marrs Mclean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030; mDepartment of Cell Biology, University of Pittsburgh School of Medicine,
    [Show full text]
  • The Role of Stress Proteins in Haloarchaea and Their Adaptive Response to Environmental Shifts
    biomolecules Review The Role of Stress Proteins in Haloarchaea and Their Adaptive Response to Environmental Shifts Laura Matarredona ,Mónica Camacho, Basilio Zafrilla , María-José Bonete and Julia Esclapez * Agrochemistry and Biochemistry Department, Biochemistry and Molecular Biology Area, Faculty of Science, University of Alicante, Ap 99, 03080 Alicante, Spain; [email protected] (L.M.); [email protected] (M.C.); [email protected] (B.Z.); [email protected] (M.-J.B.) * Correspondence: [email protected]; Tel.: +34-965-903-880 Received: 31 July 2020; Accepted: 24 September 2020; Published: 29 September 2020 Abstract: Over the years, in order to survive in their natural environment, microbial communities have acquired adaptations to nonoptimal growth conditions. These shifts are usually related to stress conditions such as low/high solar radiation, extreme temperatures, oxidative stress, pH variations, changes in salinity, or a high concentration of heavy metals. In addition, climate change is resulting in these stress conditions becoming more significant due to the frequency and intensity of extreme weather events. The most relevant damaging effect of these stressors is protein denaturation. To cope with this effect, organisms have developed different mechanisms, wherein the stress genes play an important role in deciding which of them survive. Each organism has different responses that involve the activation of many genes and molecules as well as downregulation of other genes and pathways. Focused on salinity stress, the archaeal domain encompasses the most significant extremophiles living in high-salinity environments. To have the capacity to withstand this high salinity without losing protein structure and function, the microorganisms have distinct adaptations.
    [Show full text]
  • Structure-Function Relationship and Their Role in Protein Folding
    Chapter 8 1 Molecular Chaperones: Structure-Function 2 Relationship and their Role in Protein Folding 3 Bhaskar K. Chatterjee, Sarita Puri, Ashima Sharma, Ashutosh Pastor, 4 and Tapan K. Chaudhuri 5 Abstract During heat shock conditions a plethora of proteins are found to play a 6 role in maintaining cellular homeostasis. They play diverse roles from folding of 7 non-native proteins to the proteasomal degradation of harmful aggregates. A few 8 out of these heat shock proteins (Hsp) help in the folding of non-native substrate 9 proteins and are termed as molecular chaperones. Various structural and functional 10 adaptations make them work efficiently under both normal and stress conditions. 11 These adaptations involve transitions to oligomeric structures, thermal stability, 12 efficient binding affinity for substrates and co-chaperones, elevated synthesis during 13 shock conditions, switching between ‘holding’ and ‘folding’ functions etc. Their 14 ability to function under various kinds of stress conditions like heat shock, cancers, 15 neurodegenerative diseases, and in burdened cells due to recombinant protein pro- 16 duction makes them therapeutically and industrially important biomolecules. 17 Keywords Chaperone assisted folding · Heat shock · Molecular chaperones · 18 Protein folding · Structure-function of chaperones 19 Abbreviations 20 ACD α-crystallin domain 21 ADP Adenosine di-phosphate 22 ATP Adenosine tri-phosphate 23 CCT Chaperonin containing TCP-1 24 CIRCE Controlling inverted repeat of chaperone expression 25 Bhaskar K. Chatterjee, Sarita Puri, Ashima Sharma, and Ashutosh Pastor authors are equally contributed. B. K. Chatterjee · S. Puri · A. Sharma · A. Pastor · T. K. Chaudhuri (*) Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, HauzKhas, New Delhi, India e-mail: [email protected] © Springer International Publishing AG 2018 181 A.
    [Show full text]
  • Human Prefoldin Modulates Co-Transcriptional Pre-Mrna Splicing
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.14.150466; this version posted July 22, 2020. 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. BIOLOGICAL SCIENCES: Biochemistry Human prefoldin modulates co-transcriptional pre-mRNA splicing Payán-Bravo L 1,2, Peñate X 1,2 *, Cases I 3, Pareja-Sánchez Y 1, Fontalva S 1,2, Odriozola Y 1,2, Lara E 1, Jimeno-González S 2,5, Suñé C 4, Reyes JC 5, Chávez S 1,2. 1 Instituto de Biomedicina de Sevilla, Universidad de Sevilla-CSIC-Hospital Universitario V. del Rocío, Seville, Spain. 2 Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain. 3 Centro Andaluz de Biología del Desarrollo, CSIC-Universidad Pablo de Olavide, Seville, Spain. 4 Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" IPBLN-CSIC, PTS, Granada, Spain. 5 Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain. Correspondence: Sebastián Chávez, IBiS, campus HUVR, Avda. Manuel Siurot s/n, Sevilla, 41013, Spain. Phone: +34-955923127: e-mail: [email protected]. * Co- corresponding; [email protected]. bioRxiv preprint doi: https://doi.org/10.1101/2020.06.14.150466; this version posted July 22, 2020. 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. Abstract Prefoldin is a heterohexameric complex conserved from archaea to humans that plays a cochaperone role during the cotranslational folding of actin and tubulin monomers.
    [Show full text]
  • Eukaryotic Chaperonins: Lubricating the Folding of WD-Repeat Proteins
    Current Biology, Vol. 13, R904–R905, December 2, 2003, ©2003 Elsevier Science Ltd. All rights reserved. DOI 10.1016/j.cub.2003.11.009 Eukaryotic Chaperonins: Lubricating Dispatch the Folding of WD-repeat Proteins Elizabeth A. Craig components during anaphase. Specifically, Cdc20 is required for activating the ubiquitin ligase called anaphase promoting complex or cyclosome (APC/C), Recent work has shown that the eukaryotic presumably by recruiting substrates for the ligase [7]. chaperonin CCT/TRiC facilitates folding of WD-repeat By exploiting the well-established physical interaction proteins, vastly enlarging the known clientele for this of Cdc20 with APC/C and checkpoint proteins such as chaperone beyond actin and tubulin. While the Mad2 to monitor its functional conformation, Camasses cytoskeletal proteins transit through the cochaperone et al. [1] were able to show that CCT is required for GimC/prefoldin, an Hsp70 conveys WD-repeat pro- Cdc20 to fold properly in cells. teins to CCT. The interaction between CCT and Cdc20 occurs within the region of Cdc20 that contains WD repeats. WD repeats are generally found in multiple copies in For most proteins of the eukaryotic cytosol, the folding proteins where they form four β strands and typically pathways are still largely a mystery, even though many have a tryptophan (W)-aspartic acid (D) dipeptide at cytosolic chaperones have been identified, including their carboxyl terminus. The WD-repeats fold into a multiple members of the heat shock protein (Hsp) 70 propeller structure with blades, each consisting of family, Hsp90 and the chaperonin, CCT/TRiC. Particu- four-stranded β sheets [8]. Experiments with a series larly puzzling has been the role of CCT, a distant cousin of deletion mutants expressed in yeast showed that of the prokaryotic chaperonin GroEL, found in eukary- the region of Cdc20 required for interaction with CCT otes and archea.
    [Show full text]
  • Prefoldin 6 Mediates Longevity Response from Heat Shock Factor 1 to FOXO in C
    Downloaded from genesdev.cshlp.org on September 30, 2021 - Published by Cold Spring Harbor Laboratory Press Prefoldin 6 mediates longevity response from heat shock factor 1 to FOXO in C. elegans Heehwa G. Son,1,12 Keunhee Seo,1,12 Mihwa Seo,1,2,3,4 Sangsoon Park,1 Seokjin Ham,1 Seon Woo A. An,1 Eun-Seok Choi,1,10,11 Yujin Lee,1 Haeshim Baek,1 Eunju Kim,5 Youngjae Ryu,6 Chang Man Ha,6 Ao-Lin Hsu,5,7,8 Tae-Young Roh,1,9 Sung Key Jang,1 and Seung-Jae V. Lee1,2 1Department of Life Sciences, 2School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, South Korea; 3Center for plant Aging Research, Institute for Basic Science, 4Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, South Korea; 5Department of Internal Medicine, Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA, 6Research Division, Korea Brain Research Institute, Daegu 41068, South Korea; 7Research Center for Healthy Aging, 8Institute of New Drug Development, China Medical University, Taichung 404, Taiwan; 9Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, South Korea Heat shock factor 1 (HSF-1) and forkhead box O (FOXO) are key transcription factors that protect cells from various stresses. In Caenorhabditis elegans, HSF-1 and FOXO together promote a long life span when insulin/IGF-1 signaling (IIS) is reduced. However, it remains poorly understood how HSF-1 and FOXO cooperate to confer IIS- mediated longevity.
    [Show full text]
  • 4Ac6f0708394e259036cf237a15
    International Journal of Molecular Sciences Article Expression, Functional Characterization, and Preliminary Crystallization of the Cochaperone Prefoldin from the Thermophilic Fungus Chaetomium thermophilum Kento Morita 1, Yohei Y. Yamamoto 1, Ayaka Hori 1, Tomohiro Obata 1, Yuko Uno 1, Kyosuke Shinohara 1, Keiichi Noguchi 2, Kentaro Noi 3,4, Teru Ogura 3,4, Kentaro Ishii 5, Koichi Kato 5 ID , Mahito Kikumoto 6, Rocio Arranz 7, Jose M. Valpuesta 7 ID and Masafumi Yohda 1,* 1 Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Naka, Koganei, Tokyo 184-8588, Japan; [email protected] (K.M.); [email protected] (Y.Y.Y.); [email protected] (A.H.); [email protected] (T.O.); [email protected] (Y.U.); [email protected] (K.S.) 2 Instrumentation Analysis Center, Tokyo University of Agriculture and Technology, Naka, Koganei, Tokyo 184-8588, Japan; [email protected] 3 Department of Molecular Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan.; [email protected] (K.N.); [email protected] (T.O.) 4 Core Research for Evolutional Science and Technology (CREST), JST, Kawaguchi, Saitama 332-0012, Japan 5 Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Myodaiji, Okazaki 444-8787, Japan; [email protected] (K.I.); [email protected] (K.K.) 6 Structural Biology Research Center, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601, Japan; [email protected] 7 Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Madrid 28049, Spain; [email protected] (R.A.); [email protected] (J.M.V.) * Correspondence: [email protected]; Tel.: +81-42-388-7479 Received: 4 June 2018; Accepted: 15 August 2018; Published: 19 August 2018 Abstract: Prefoldin is a hexameric molecular chaperone found in the cytosol of archaea and eukaryotes.
    [Show full text]
  • Function of Host Protein Staufen1 in Rabies Virus Replication
    viruses Article Function of Host Protein Staufen1 in Rabies Virus Replication Gaowen Liu †, Congjie Chen †, Ruixian Xu, Ming Yang, Qinqin Han, Binghui Wang, Yuzhu Song *, Xueshan Xia * and Jinyang Zhang * Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; [email protected] (G.L.); [email protected] (C.C.); [email protected] (R.X.); [email protected] (M.Y.); [email protected] (Q.H.); [email protected] (B.W.) * Correspondence: [email protected] (Y.S.); [email protected] (X.X.); [email protected] (J.Z.); Tel.: +86-871-65939528 (Y.S. & J.Z.) † These authors contributed equally to this work. Abstract: Rabies virus is a highly neurophilic negative-strand RNA virus with high lethality and remains a huge public health problem in developing countries to date. The double-stranded RNA- binding protein Staufen1 (STAU1) has multiple functions in RNA virus replication, transcription, and translation. However, its function in RABV infection and its mechanism of action are not clear. In this study, we investigated the role of host factor STAU1 in RABV infection of SH-SY-5Y cells. Immunofluorescence, TCID50 titers, confocal microscopy, quantitative real-time PCR and Western blotting were carried out to determine the molecular function and subcellular distribution of STAU1 in these cell lines. Expression of STAU1 in SH-SY-5Y cells was down-regulated by RNA interference or up-regulated by transfection of eukaryotic expression vectors. The results showed that N proficiently colocalized with STAU1 in SH-SY-5Y at 36 h post-infection, and the expression level of STAU1 was also proportional to the time of infection.
    [Show full text]
  • It's Not Just a Phase; Ubiquitination in Cytosolic Protein Quality Control
    Biochemical Society Transactions (2021) 49 365–377 https://doi.org/10.1042/BST20200694 Review Article It’s not just a phase; ubiquitination in cytosolic protein quality control Heather A. Baker1 and Jonathan P. Bernardini1,2,3 1Michael Smith Laboratories, University of British Columbia, Vancouver, Canada; 2Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; 3Department of Medical Biology, The University of Melbourne, Melbourne, Australia Correspondence: Jonathan P. Bernardini ([email protected]) Downloaded from http://portlandpress.com/biochemsoctrans/article-pdf/49/1/365/905078/bst-2020-0694c.pdf by guest on 27 September 2021 The accumulation of misfolded proteins is associated with numerous degenerative condi- tions, cancers and genetic diseases. These pathological imbalances in protein homeosta- sis (termed proteostasis), result from the improper triage and disposal of damaged and defective proteins from the cell. The ubiquitin-proteasome system is a key pathway for the molecular control of misfolded cytosolic proteins, co-opting a cascade of ubiquitin ligases to direct terminally damaged proteins to the proteasome via modification with chains of the small protein, ubiquitin. Despite the evidence for ubiquitination in this crit- ical pathway, the precise complement of ubiquitin ligases and deubiquitinases that modu- late this process remains under investigation. Whilst chaperones act as the first line of defence against protein misfolding, the ubiquitination machinery has a pivotal role in tar- geting terminally defunct cytosolic proteins for destruction. Recent work points to a complex assemblage of chaperones, ubiquitination machinery and subcellular quarantine as components of the cellular arsenal against proteinopathies. In this review, we examine the contribution of these pathways and cellular compartments to the maintenance of the cytosolic proteome.
    [Show full text]
  • Bio-Functional Analysis of Ubiquitin Ligase UBE3D
    Bio-Functional Analysis of Ubiquitin Ligase UBE3D by Adam William Penn A thesis submitted in conformity with the requirements for the degree of Masters of Science Department of Medical Biophysics University of Toronto © Copyright by Adam William Penn 2020 ii Bio-Functional Analysis of Ubiquitin Ligase UBE3D Adam William Penn Masters of Science Department of Medical Biophysics University of Toronto 2020 Abstract The ubiquitin system is comprised of a reversible three step process: E1 activating enzyme, E2 conjugating enzyme and an E3 ligase, leading to ubiquitin molecules being post-translationally modified onto substrate proteins leading to a plethora of downstream effects (localization, function and half-life). UBE3D, a HECT (homologous to E6-AP carboxylic terminus) E3 ligase, has a relatively elusive regulatory role within the cell. Here, we systematically analyze and characterize UBE3D as well as its highest confidence interactor, Dynein axonemal assembly factor (DNAAF2) through: Autoubiquitylation assay; intracellular localization with immunofluorescence; interaction network using proximity-dependent biotin identification (BioID) to better understand the relationship of these two proteins. DNAAF2 protein interaction mapping allowed for insight into PIH domain. In summary, I have used multiple approaches to gain novel knowledge and insight into the potential functional role of UBE3D within the cell, and its putative partner protein, DNAAF2. ii iii Table of Contents 1 Introduction 1 1.1 The Ubiquitin System 1 1.1.1 E1 Ubiquitin Activating
    [Show full text]