Human Origin Recognition Complex Is Essential for HP1 Binding to Chromatin and Heterochromatin Organization

Total Page:16

File Type:pdf, Size:1020Kb

Human Origin Recognition Complex Is Essential for HP1 Binding to Chromatin and Heterochromatin Organization Human origin recognition complex is essential for HP1 binding to chromatin and heterochromatin organization Supriya G. Prasantha,b,1, Zhen Shenb, Kannanganattu V. Prasanthb, and Bruce Stillmana,1 aCold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724; and bDepartment of Cell and Developmental Biology, University of Illinois, Urbana- Champaign, IL 61801 Contributed by Bruce Stillman, July 9, 2010 (sent for review April 10, 2010) The origin recognition complex (ORC) is a DNA replication initiator (15, 22, 23), cytokinesis in human cells and Drosophila (24, 25), protein also known to be involved in diverse cellular functions regulation of dendrite development in postmitotic neurons (26), including gene silencing, sister chromatid cohesion, telomere bi- and neural transmission and synaptic function in Drosophila (27) ology, heterochromatin localization, centromere and centrosome (see reviews in refs. 28, 29). activity, and cytokinesis. We show that, in human cells, multiple ORC The human Orc2 subunit is associated with heterochromatin in subunits associate with hetereochromatin protein 1 (HP1) α-and a cell cycle-dependent manner, being present only at the cen- HP1β-containing heterochromatic foci. Fluorescent bleaching studies tromeres during late G2 and mitosis (15, 30). Furthermore, Orc2 indicate that multiple subcomplexes of ORC exist at heterochroma- biochemically interacts indirectly with the heterochromatin protein tin, with Orc1 stably associating with heterochromatin in G1 phase, 1 (HP1) in mammalian, Drosophila,andXenopus cells (14, 15, 31, whereas other ORC subunits have transient interactions throughout 32). Depletion of Orc2 or Orc3 in HeLa cells results in a mitotic the cell-division cycle. Both Orc1 and Orc3 directly bind to HP1α,and arrest with abnormally condensed chromosomes, lack of chromo- two domains of Orc3, a coiled-coil domain and a mod-interacting some alignment at the metaphase plate, and spindle defects (15). region domain, can independently bind to HP1α; however, both The interaction between ORC and heterochromatic proteins are essential for in vivo localization of Orc3 to heterochromatic foci. α β CELL BIOLOGY Direct binding of both Orc1 and Orc3 to HP1 suggests that, after the like HP1 and and ORC binding to centromeres suggests a di- rect role for ORC subunits in chromosome organization and for degradation of Orc1 at the G1/S boundary, Orc3 facilitates assembly – of ORC/HP1 proteins to chromatin. Although depletion of Orc2 and chromosome condensation and segregation during mitosis (33 Orc3 subunits by siRNA caused loss of HP1α association to hetero- 35). HP1 is a heterochromatin-associated protein that localizes to chromatin, loss of Orc1 and Orc5 caused aberrant HP1α distribution pericentromeres and telomeres, has a dose-dependent effect on only to pericentric heterochromatin-surrounding nucleoli. Depletion gene silencing, and has a critical role in heterochromatin forma- of HP1α from human cells also shows loss of Orc2 binding to het- tion and maintenance (36). To address the role of ORC/HP1 in erochromatin, suggesting that ORC and HP1 proteins are mutually heterochromatin organization and mitotic progression, we sys- required for each other to bind to heterochromatin. Similar to tematically analyzed the role of individual ORC subunits in HP1 HP1α-depleted cells, Orc2 and Orc3 siRNA-treated cells also show binding and recruitment to heterochromatic foci in human cells. loss of compaction at satellite repeats, suggesting that ORC to- We find that ORC and HP1 are required for maintenance of each gether with HP1 proteins may be involved in organizing higher- other’s association with heterochromatin and identify a role for order chromatin structure and centromere function. ORC in maintenance of higher-order chromatin structure at centromeres. Interestingly, different ORC subunits play differ- centromere | origin recognition complex | pericentric heterochromatin | ential roles in heterochromatin structure and function. chromatin structure Results he initiation of DNA replication in eukaryotic cells is a highly Multiple ORC Subunits Localize to Heterochromatin. Immunofluo- Tcoordinated process that is intimately linked to chromatin or- rescence studies showed that human and mouse Orc2 localizes in ganization. The accurate duplication of DNA is governed by many a cell cycle-regulated manner to many diffuse foci in the nucleus, proteins including the origin recognition complex (ORC) (1, 2). HP1-containing heterochromatic loci, and centrosomes and Binding of ORC to replication origins is a key step in initiation centromeres (Fig. 1 B’–D’) (11, 15, 20, 21, 30). In addition to Orc2, of DNA replication, leading to the recruitment of other prerep- other ORC subunits including Orc1, Orc3, and Orc5 localize at lication complex (pre-RC) proteins including Cdc6, Cdt1, and the Orc2- and HP1-associated heterochromatin in MCF7 cells 2-7 the MCM . Unlike yeast ORC, human ORC binds to chromatin (Fig. 1). In human cells, Orc2 coimmunoprecipitates with HP1α fi – in a nonsequence-speci c manner (3). In human cells, Orc2 5 and β (15), and in Drosophila embryonic nuclei, HP1 colocalizes forms the core ORC, and Orc1 is transiently associated with this – with Orc2 at heterochromatin (14). Because other ORC subunits complex (4 7). During the G1 to S phase transition, human Orc1 also colocalize with HP1α in human cells, it suggests an evolu- dissociates from chromatin, is ubiquitylated and degraded by tionary conserved function for ORC at heterochromatin. ubiquitin-mediated proteolysis, and is reloaded to the chromatin at the M-G1 transition when new pre-RCs are formed (5, 7–9). The remaining ORC subunits are dynamically associated with chro- Author contributions: S.G.P. and B.S. designed research; S.G.P., Z.S., and K.V.P. performed matin throughout the remainder of the cell cycle (5, 6). research; S.G.P. and B.S. contributed new reagents/analytic tools; S.G.P., Z.S., K.V.P., and In addition to their role in DNA replication, ORC subunits have B.S. analyzed data; and S.G.P. and B.S. wrote the paper. also been implicated in transcriptional gene silencing in Saccha- The authors declare no conflict of interest. romyces cerevisiae (10), heterochromatin formation in Drosophila, Freely available online through the PNAS open access option. – mouse, and humans (11 15), sister chromatid cohesion in S. cer- 1To whom correspondence may be addressed. E-mail: [email protected] or stillman@ evisiae, Schizosaccharomyces pombe, and Xenopus (16–18), ribo- cshl.edu. some biogenesis (19), centrosome biology in human and mouse This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. (15, 20, 21), mitotic chromosome function in human and Xenopus 1073/pnas.1009945107/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1009945107 PNAS Early Edition | 1of6 Downloaded by guest on October 2, 2021 A A’ A’’ A Orc3-YFP HP1 DAPI B B’ B’’ Orc3-YFP Orc2 pAb DAPI C C’ C’’ YFP-Orc1 Orc2 pAb DAPI D D’ D’’ D’’’ YFP-Orc5 Orc2 pAb HP1 DAPI Fig. 1. Multiple ORC subunits localize with HP1 to heterochromatic foci. YFP fusion constructs were transfected into MCF7 cells, and endogenous B Orc1 Orc2 Orc3 Orc4 Orc5 proteins were detected by indirect immunofluorescence. Orc3-YFP coloc- alizes with endogenous HP1α at heterochromatic foci (A, A’, and A”). En- Input dogenous Orc2 and Orc3-YFP colocalize at the heterochromatic foci (B, B’, Orc1 Orc2 Orc3 Orc4 Orc5 GST-HP1 GST-HP1 GST-HP1 GST-HP1 GST-HP1 and B”). YFP-Orc1 colocalizes with endogenous Orc2 at the heterochromatic GST GST GST GST GST foci (C, C’, and C”). YFP-Orc5 showed colocalization with both endogenous Orc2 and HP1α at the heterochromatic foci in MCF7 cells (D, D’, D”, and D”’). Chromatin was stained with DAPI in blue. (Scale bar, 5 μm.) Differential Binding of ORC Subunits to Heterochromatin. A large proportion of the HP1 protein pool displays a remarkably dynamic exchange at heterochromatin (37–39). Because the human Orc1, Orc2, Orc3, and Orc5 subunits localized to heterochromatin, we compared the mobility of YFP-tagged ORC subunits with YFP- α fl Fig. 2. ORC dynamics at the heterochromatin. (A) FRAP kinetics of various HP1 at heterochromatin by uorescence recovery after photo- ORC subunits at the heterochromatic foci in MCF7 cells. Similar FRAP kinetics bleaching (FRAP) analysis (Fig. 2A). A defined heterochromatic of ∼3–4 s t1/2 for YFP-HP1α, YFP-Orc2, and Orc3-YFP were observed at region in the nucleus was irreversibly bleached of the YFP signal, heterochromatic foci, whereas YFP-Orc1 showed >40 s t1/2 at heterochro- and the recovery kinetics of fluorescence intensity into the bleached matin. (B) GST pull-down assays using GST or GST-HP1α beads and incubating region were assessed. Orc2 and Orc3 had very similar and complete with in vitro transcribed and translated mRNA from individual ORC subunits kinetics of fluorescence recovery, as observed for HP1α. However, shows direct binding of both human Orc1 and Orc3 to HP1α. The input is Orc1 showed very different and incomplete recovery kinetics, shown on Right. suggesting that Orc1 chromatin association is unique among ORC fl subunits tested, perhaps re ecting different chromatin interaction (aa151-269) (32). To map the region of Orc3 that interacts with (Fig. 2A). Thus, by FRAP analysis, it is clear that ORC proteins, HP1α, various mutants of Orc3 were generated in a coupled in especially Orc2 and Orc3, assemble and disassemble from the vitro transcription-translation vector and tested for binding to GST heterochromatic foci in a dynamic fashion like the bulk of HP1α or GST-HP1α (Fig. 3 A and B and Fig. S2). Two independent protein, but Orc1 seems to exchange less frequently, if at all. domains in Orc3, a coiled-coil domain at the N terminus and α a second region containing a MOD1-interacting region (MIR; 213- Orc1 and Orc3 Directly Bind to HP1 . In Drosophila and Xenopus, fi α HP1 was found to interact with the N-terminal domain of the Orc1 218aa) (42), were identi ed, both of which directly bound to HP1 .
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • ORC3 (1D6): Sc-23888
    SANTA CRUZ BIOTECHNOLOGY, INC. ORC3 (1D6): sc-23888 BACKGROUND STORAGE The initiation of DNA replication is a multi-step process that depends on the Store at 4° C, **DO NOT FREEZE**. Stable for one year from the date of formation of pre-replication complexes, which trigger initiation. Among the shipment. Non-hazardous. No MSDS required. proteins required for establishing these complexes are the origin recognition complex (ORC) proteins. ORC proteins bind specifically to origins of replication DATA where they serve as scaffold for the assembly of additional initiation factors. A B Human ORC subunits 1-6 are expressed in the nucleus of proliferating cells AB C DE and tissues, such as the testis. ORC1 and ORC2 are both expressed at equiv- 113 K – 89 K – alent concentrations throughout the cell cycle; however, only ORC2 remains < ORC3 stably bound to chromatin. ORC4 and ORC6 are also expressed constantly throughout the cell cycle. ORC2, ORC3, ORC4 and ORC5 form a core complex 49 K – upon which ORC6 and ORC1 assemble. The formation of this core complex suggests that ORC proteins play a crucial role in the G1-S transition in mam- malian cells. ORC3 (1D6): sc-23888. Western blot analysis of ORC3 ORC3 (1D6): sc-23888. Immunoperoxidase staining of expression in HeLa (A), Raji (B), Jurkat (C), WI-38 (D) formalin fixed, paraffin-embedded human ovary tissue CHROMOSOMAL LOCATION and A-549 (E) whole cell lysates. showing nuclear localization (A,B). Genetic locus: ORC3L (human) mapping to 6q15. SELECT PRODUCT CITATIONS SOURCE 1. Braden, W.A., et al. 2006. Distinct action of the retinoblastoma pathway ORC3 (1D6) is a rat monoclonal antibody raised against partially purified on the DNA replication machinery defines specific roles for cyclin-depen- His-tagged bacterially expressed fusion protein corresponding to human ORC3.
    [Show full text]
  • Systemic Analysis of the DNA Replication Regulators Origin Recognition Complex in Lung Adenocarcinomas Identifes Prognostic and Expression Signifcance
    Systemic Analysis of the DNA Replication Regulators Origin Recognition Complex in Lung Adenocarcinomas Identies Prognostic and Expression Signicance Juan Chen University of South China Juan Zou University of South China Juan Zeng University of South China Tian Zeng University of South China Qi-hao Hu University of South China Jun-hui Bai University of South China Min Tang University of South China Yu-kun Li ( [email protected] ) University of South China https://orcid.org/0000-0002-8517-9075 Primary research Keywords: lung adenocarcinomas, ORC complex, public databases, prognostic value, comprehensive bioinformatics Posted Date: May 11th, 2021 DOI: https://doi.org/10.21203/rs.3.rs-487176/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/24 Abstract Background: Origin recognition complex (ORC) 1, ORC2, ORC3, ORC4, ORC5 and ORC6, form a replication- initiator complex to mediate DNA replication, which play a key role in carcinogenesis, while their role in lung adenocarcinomas (LUAD) remains poorly understood. Methods: We conrmed the transcriptional and post-transcriptional levels, DNA alteration, DNA methylation, miRNA network, protein structure, PPI network, functional enrichment, immune inltration and prognostic value of ORCs in LUAD based on Oncomine, GEPIA, HPA, cBioportal, TCGA, GeneMANIA, Metascape, KM-plot, GENT2, and TIMER database. Results: ORC mRNA and protein were both enhanced obviously based on Oncomine, Ualcan, GEPIA, TCGA and HPA database. Furthermore, ORC1 and ORC6 have signicant prognostic values for LUAD patients based on GEPIA database. Protein structure, PPI network, functional enrichment and immune inltration analysis indicated that ORC complex cooperatively accelerate the LUAD development by promoting DNA replication, cellular senescence and metabolic process.
    [Show full text]
  • Chromosome Integrity in Saccharomyces Cerevisiae: the Interplay of DNA Replication Initiation Factors, Elongation Factors, and Origins
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press Chromosome integrity in Saccharomyces cerevisiae: the interplay of DNA replication initiation factors, elongation factors, and origins Dongli Huang1,2 and Douglas Koshland1,3 1Howard Hughes Medical Institute, Carnegie Institution of Washington, Department of Embryology, Baltimore, Maryland 21210, USA; 2Johns Hopkins University, Department of Biology, Baltimore, Maryland 21218, USA The integrity of chromosomes during cell division is ensured by both trans-acting factors and cis-acting chromosomal sites. Failure of either these chromosome integrity determinants (CIDs) can cause chromosomes to be broken and subsequently misrepaired to form gross chromosomal rearrangements (GCRs). We developed a simple and rapid assay for GCRs, exploiting yeast artificial chromosomes (YACs) in Saccharomyces cerevisiae. We used this assay to screen a genome-wide pool of mutants for elevated rates of GCR. The analyses of these mutants define new CIDs (Orc3p, Orc5p, and Ycs4p) and new pathways required for chromosome integrity in DNA replication elongation (Dpb11p), DNA replication initiation (Orc3p and Orc5p), and mitotic condensation (Ycs4p). We show that the chromosome integrity function of Orc5p is associated with its ATP-binding motif and is distinct from its function in controlling the efficiency of initiation of DNA replication. Finally, we used our YAC assay to assess the interplay of trans and cis factors in chromosome integrity. Increasing the number of origins on a YAC suppresses GCR formation in our dpb11 mutant but enhances it in our orc mutants. This result provides potential insights into the counterbalancing selective pressures necessary for the evolution of origin density on chromosomes.
    [Show full text]
  • Genome-Wide Analysis of the Core DNA Replication Machinery in the Higher Plants Arabidopsis and Rice1[W][OA]
    Genome Analysis Genome-Wide Analysis of the Core DNA Replication Machinery in the Higher Plants Arabidopsis and Rice1[W][OA] Randall W. Shultz2, Vinaya M. Tatineni, Linda Hanley-Bowdoin*, and William F. Thompson Department of Plant Biology (R.W.S., W.F.T.), Department of Statistical Genetics and Bioinformatics (V.M.T.), and Department of Molecular and Structural Biochemistry (L.H.-B.), North Carolina State University, Raleigh, North Carolina 27695 Core DNA replication proteins mediate the initiation, elongation, and Okazaki fragment maturation functions of DNA replication. Although this process is generally conserved in eukaryotes, important differences in the molecular architecture of the DNA replication machine and the function of individual subunits have been reported in various model systems. We have combined genome-wide bioinformatic analyses of Arabidopsis (Arabidopsis thaliana)andrice(Oryza sativa)with published experimental data to provide a comprehensive view of the core DNA replication machinery in plants. Many components identified in this analysis have not been studied previously in plant systems, including the GINS (go ichi ni san) complex (PSF1, PSF2, PSF3, and SLD5), MCM8, MCM9, MCM10, NOC3, POLA2, POLA3, POLA4, POLD3, POLD4, and RNASEH2. Our results indicate that the core DNA replication machinery from plants is more similar to vertebrates than single-celled yeasts (Saccharomyces cerevisiae), suggesting that animal models may be more relevant to plant systems. However, we also uncovered some important differences between plants and vertebrate machinery. For example, we did not identify geminin or RNASEH1 genes in plants. Our analyses also indicate that plants may be unique among eukaryotes in that they have multiple copies of numerous core DNA replication genes.
    [Show full text]
  • Signature of Progression and Negative Outcome in Colorectal Cancer
    Oncogene (2010) 29, 876–887 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc ORIGINAL ARTICLE A ‘DNA replication’ signature of progression and negative outcome in colorectal cancer M-J Pillaire1,7, J Selves2,7, K Gordien2, P-A Gouraud3, C Gentil3, M Danjoux2,CDo3, V Negre4, A Bieth1, R Guimbaud2, D Trouche5, P Pasero6,MMe´chali6, J-S Hoffmann1 and C Cazaux1 1Genetic Instability and Cancer Group, Department Biology of Cancer, Institute of Pharmacology and Structural Biology, UMR5089 CNRS, University of Toulouse, University Paul Sabatier, Toulouse, France; 2INSERM U563, Federation of Digestive Cancerology and Department of Anatomo-pathology, University of Toulouse, University Paul Sabatier, Toulouse, France; 3Service of Epidemiology, INSERM U558, Faculty of Medicine, University of Toulouse, University Paul Sabatier, Alle´es Jules Guesde, Toulouse, France; 4aCGH GSO Canceropole Platform, INSERM U868, Val d’Aurelle, Montpellier, France; 5Laboratory of Cellular and Molecular Biology of Cell Proliferation Control, UMR 5099 CNRS, University of Toulouse, University Paul Sabatier, Toulouse, France and 6Institute of Human Genetics UPR1142 CNRS, Montpellier, France Colorectal cancer is one of the most frequent cancers Introduction worldwide. As the tumor-node-metastasis (TNM) staging classification does not allow to predict the survival of DNA replication in normal cells is regulated by an patients in many cases, additional prognostic factors are ‘origin licensing’ mechanism that ensures that it occurs needed to better forecast their outcome. Genes involved in just once per cycle. Once cells enter the S-phase, the DNA replication may represent an underexplored source stability of DNA replication forks must be preserved to of such prognostic markers.
    [Show full text]
  • A Free-Living Protist That Lacks Canonical Eukaryotic DNA Replication and Segregation Systems
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.14.435266; this version posted March 15, 2021. 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 A free-living protist that lacks canonical eukaryotic DNA replication and segregation systems 2 Dayana E. Salas-Leiva1, Eelco C. Tromer2,3, Bruce A. Curtis1, Jon Jerlström-Hultqvist1, Martin 3 Kolisko4, Zhenzhen Yi5, Joan S. Salas-Leiva6, Lucie Gallot-Lavallée1, Geert J. P. L. Kops3, John M. 4 Archibald1, Alastair G. B. Simpson7 and Andrew J. Roger1* 5 1Centre for Comparative Genomics and Evolutionary Bioinformatics (CGEB), Department of 6 Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada, B3H 4R2 2 7 Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom 8 3Oncode Institute, Hubrecht Institute – KNAW (Royal Netherlands Academy of Arts and Sciences) 9 and University Medical Centre Utrecht, Utrecht, The Netherlands 10 4Institute of Parasitology Biology Centre, Czech Acad. Sci, České Budějovice, Czech Republic 11 5Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, 12 South China Normal University, Guangzhou 510631, China 13 6CONACyT-Centro de Investigación en Materiales Avanzados, Departamento de medio ambiente y 14 energía, Miguel de Cervantes 120, Complejo Industrial Chihuahua, 31136 Chihuahua, Chih., México 15 7Centre for Comparative Genomics and Evolutionary Bioinformatics (CGEB), Department of 16 Biology, Dalhousie University, Halifax, NS, Canada, B3H 4R2 17 *corresponding author: [email protected] 18 D.E.S-L ORCID iD: 0000-0003-2356-3351 19 E.C.T.
    [Show full text]
  • The Architecture of the DNA Replication Origin Recognition Complex in Saccharomyces Cerevisiae
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Spiral - Imperial College Digital Repository The architecture of the DNA replication origin recognition complex in Saccharomyces cerevisiae Zhiqiang Chen, Christian Speck, Patricia Wendel, Chunyan Tang, Bruce Stillman, and Huilin Li ABSTRACT The origin recognition complex (ORC) is conserved in all eukaryotes. The six proteins of the Saccharomyces cerevisiae ORC that form a stable complex bind to origins of DNA replication and recruit prereplicative complex (pre-RC) proteins, one of which is Cdc6. To further understand the function of ORC we recently determined by single-particle reconstruction of electron micrographs a low-resolution, 3D structure of S. cerevisiae ORC and the ORC–Cdc6 complex. In this article, the spatial arrangement of the ORC subunits within the ORC structure is described. In one approach, a maltose binding protein (MBP) was systematically fused to the N or the C termini of the five largest ORC subunits, one subunit at a time, generating 10 MBP-fused ORCs, and the MBP density was localized in the averaged, 2D EM images of the MBP-fused ORC particles. Determining the Orc1–5 structure and comparing it with the native ORC structure localized the Orc6 subunit near Orc2 and Orc3. Finally, subunit–subunit interactions were determined by immunoprecipitation of ORC subunits synthesized in vitro. Based on the derived ORC architecture and existing structures of archaeal Orc1–DNA structures, we propose a model for ORC and suggest how ORC interacts with origin DNA and Cdc6. The studies provide a basis for understanding the overall structure of the pre- RC.
    [Show full text]
  • Multifunctionality of the Telomere-Capping Shelterin Complex Explained by Variations in Its Protein Composition
    cells Review Multifunctionality of the Telomere-Capping Shelterin Complex Explained by Variations in Its Protein Composition Claire Ghilain 1, Eric Gilson 1,2,3,* and Marie-Josèphe Giraud-Panis 1,* 1 Université Côte d’Azur, CNRS, INSERM, IRCAN, 06000 Nice, France; [email protected] 2 International Research Laboratory for Cancer, Aging and Hematology, Shanghai Ruijin Hospital, Shanghai Jiaotong University and Côte-d’Azur University, Shanghai 200025, China 3 Department of Genetics, CHU Nice, 06000 Nice, France * Correspondence: [email protected] (E.G.); [email protected] (M.-J.G.-P.) Abstract: Protecting telomere from the DNA damage response is essential to avoid the entry into cellular senescence and organismal aging. The progressive telomere DNA shortening in dividing somatic cells, programmed during development, leads to critically short telomeres that trigger replicative senescence and thereby contribute to aging. In several organisms, including mammals, telomeres are protected by a protein complex named Shelterin that counteract at various levels the DNA damage response at chromosome ends through the specific function of each of its subunits. The changes in Shelterin structure and function during development and aging is thus an intense area of research. Here, we review our knowledge on the existence of several Shelterin subcomplexes and the functional independence between them. This leads us to discuss the possibility that the multifunctionality of the Shelterin complex is determined by the formation of different subcomplexes Citation: Ghilain, C.; Gilson, E.; whose composition may change during aging. Giraud-Panis, M.-J. Multifunctionality of the Keywords: telomere; aging; Shelterin; senescence; DNA damage response Telomere-Capping Shelterin Complex Explained by Variations in Its Protein Composition.
    [Show full text]
  • A Conserved but Plant-Specific CDK-Mediated Regulation of DNA Replication Protein A2 in the Precise Control of Stomatal Terminal Division
    A conserved but plant-specific CDK-mediated regulation of DNA replication protein A2 in the precise control of stomatal terminal division Kezhen Yanga, Lingling Zhua,b, Hongzhe Wanga, Min Jianga, Chunwang Xiaoc,d, Xiangyang Hue, Steffen Vannestef,g,h, Juan Dongi, and Jie Lea,b,1 aKey Laboratory of Plant Molecular Physiology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Botany, Chinese Academy of Sciences, 100093 Beijing, China; bUniversity of Chinese Academy of Sciences, 100049 Beijing, China; cCollege of Life and Environmental Sciences, Minzu University of China, 100081 Beijing, China; dHulun Lake Reserve Grassland Ecology Research Station, Minzu University of China, 100081 Beijing, China; eShanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, 200444 Shanghai, China; fCenter for Plant Systems Biology, VIB, 9052 Ghent, Belgium; gDepartment of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; hLaboratory of Plant Growth Analysis, Ghent University Global Campus, 21985 Incheon, Republic of Korea; and iWaksman Institute of Microbiology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 Edited by David C. Baulcombe, University of Cambridge, Cambridge, United Kingdom, and approved July 29, 2019 (received for review November 11, 2018) The R2R3-MYB transcription factor FOUR LIPS (FLP) controls the condition, RPA2 is hyperphosphorylated by the PIKK-family stomatal terminal division through transcriptional repression of kinases (ATM, ATR, and DNA-PK) that facilitates mitotic exit the cell cycle genes CYCLIN-DEPENDENT KINASE (CDK) B1s (CDKB1s), and the initiation of DNA repair (13–15). All known RPA2 CDKA;1,andCYCLIN A2s (CYCA2s). We mutagenized the weak mu- homologs have a conserved N-terminal phosphorylation domain, tant allele flp-1 seeds with ethylmethane sulfonate and screened although the specific residues may be not conserved in different out a flp-1 suppressor 1 (fsp1) that suppressed the flp-1 stomatal species (11).
    [Show full text]
  • Cshperspect-REP-A015727 Table3 1..10
    Table 3. Nomenclature for proteins and protein complexes in different organisms Mammals Budding yeast Fission yeast Flies Plants Archaea Bacteria Prereplication complex assembly H. sapiens S. cerevisiae S. pombe D. melanogaster A. thaliana S. solfataricus E. coli Hs Sc Sp Dm At Sso Eco ORC ORC ORC ORC ORC [Orc1/Cdc6]-1, 2, 3 DnaA Orc1/p97 Orc1/p104 Orc1/Orp1/p81 Orc1/p103 Orc1a, Orc1b Orc2/p82 Orc2/p71 Orc2/Orp2/p61 Orc2/p69 Orc2 Orc3/p66 Orc3/p72 Orc3/Orp3/p80 Orc3/Lat/p82 Orc3 Orc4/p50 Orc4/p61 Orc4/Orp4/p109 Orc4/p52 Orc4 Orc5L/p50 Orc5/p55 Orc5/Orp5/p52 Orc5/p52 Orc5 Orc6/p28 Orc6/p50 Orc6/Orp6/p31 Orc6/p29 Orc6 Cdc6 Cdc6 Cdc18 Cdc6 Cdc6a, Cdc6b [Orc1/Cdc6]-1, 2, 3 DnaC Cdt1/Rlf-B Tah11/Sid2/Cdt1 Cdt1 Dup/Cdt1 Cdt1a, Cdt1b Whip g MCM helicase MCM helicase MCM helicase MCM helicase MCM helicase Mcm DnaB Mcm2 Mcm2 Mcm2/Nda1/Cdc19 Mcm2 Mcm2 Mcm3 Mcm3 Mcm3 Mcm3 Mcm3 Mcm4 Mcm4/Cdc54 Mcm4/Cdc21 Mcm4/Dpa Mcm4 Mcm5 Mcm5/Cdc46/Bob1 Mcm5/Nda4 Mcm5 Mcm5 Mcm6 Mcm6 Mcm6/Mis5 Mcm6 Mcm6 Mcm7 Mcm7/Cdc47 Mcm7 Mcm7 Mcm7/Prolifera Gmnn/Geminin Geminin Mcm9 Mcm9 Hbo1 Chm/Hat1 Ham1 Ham2 DiaA Ihfa Ihfb Fis SeqA Replication fork assembly Hs Sc Sp Dm At Sso Eco Mcm8 Rec/Mcm8 Mcm8 Mcm10 Mcm10/Dna43 Mcm10/Cdc23 Mcm10 Mcm10 DDK complex DDK complex DDK complex DDK complex Cdc7 Cdc7 Hsk1 l(1)G0148 Hsk1-like 1 Dbf4/Ask Dbf4 Dfp1/Him1/Rad35 Chif/chiffon Drf1 Continued 2 Replication fork assembly (Continued ) Hs Sc Sp Dm At Sso Eco CDK complex CDK complex CDK complex CDK complex CDK complex Cdk1 Cdc28/Cdk1 Cdc2/Cdk1 Cdc2 CdkA Cdk2 Cdc2c CcnA1, A2 CycA CycA1, A2,
    [Show full text]
  • The Human Origin Recognition Complex Is Essential for Pre-RC Assembly
    RESEARCH ARTICLE The human origin recognition complex is essential for pre-RC assembly, mitosis, and maintenance of nuclear structure Hsiang-Chen Chou1,2†, Kuhulika Bhalla1†, Osama EL Demerdesh1, Olaf Klingbeil1, Kaarina Hanington1, Sergey Aganezov3, Peter Andrews1, Habeeb Alsudani1, Kenneth Chang1, Christopher R Vakoc1, Michael C Schatz3, W Richard McCombie1, Bruce Stillman1* 1Cold Spring Harbor Laboratory, Cold Spring Harbor, United States; 2Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, United States; 3Department of Computer Science, Whiting School of Engineering, Johns Hopkins University, Baltimore, United States Abstract The origin recognition complex (ORC) cooperates with CDC6, MCM2-7, and CDT1 to form pre-RC complexes at origins of DNA replication. Here, using tiling-sgRNA CRISPR screens, we report that each subunit of ORC and CDC6 is essential in human cells. Using an auxin-inducible degradation system, we created stable cell lines capable of ablating ORC2 rapidly, revealing multiple cell division cycle phenotypes. The primary defects in the absence of ORC2 were cells encountering difficulty in initiating DNA replication or progressing through the cell division cycle due to reduced MCM2-7 loading onto chromatin in G1 phase. The nuclei of ORC2-deficient cells were also large, with decompacted heterochromatin. Some ORC2-deficient cells that completed DNA replication entered into, but never exited mitosis. ORC1 knockout cells also demonstrated extremely slow cell proliferation and abnormal cell and nuclear morphology. Thus, ORC proteins and CDC6 are indispensable for normal cellular proliferation and contribute to nuclear organization. *For correspondence: [email protected] †These authors contributed equally to this work Introduction Cell division requires the entire genome to be duplicated once and only once during S-phase of the Competing interest: See cell cycle, followed by segregation of the sister chromatids into two daughter cells.
    [Show full text]