Characterization of the Repeat-Tract Instability and Mutator Phenotypes Conferred by a Tn3 Insertion in RFC1, the Large Subunit of the Yeast Clamp Loader

Total Page:16

File Type:pdf, Size:1020Kb

Characterization of the Repeat-Tract Instability and Mutator Phenotypes Conferred by a Tn3 Insertion in RFC1, the Large Subunit of the Yeast Clamp Loader Copyright 1999 by the Genetics Society of America Characterization of the Repeat-Tract Instability and Mutator Phenotypes Conferred by a Tn3 Insertion in RFC1, the Large Subunit of the Yeast Clamp Loader Yali Xie,* Chris Counter² and Eric Alani* *Section of Genetics and Development, Cornell University, Ithaca, New York 14853-2703 and ²Department of Pharmacology and Cancer Biology, Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710 Manuscript received August 3, 1998 Accepted for publication October 22, 1998 ABSTRACT The RFC1 gene encodes the large subunit of the yeast clamp loader (RFC) that is a component of eukaryotic DNA polymerase holoenzymes. We identi®ed a mutant allele of RFC1 (rfc1::Tn3) from a large collection of Saccharomyces cerevisiae mutants that were inviable when present in a rad52 null mutation background. Analysis of rfc1::Tn3 strains indicated that they displayed both a mutator and repeat-tract instability phenotype. Strains bearing this allele were characterized in combination with mismatch repair (msh2D, pms1D), double-strand break repair (rad52), and DNA replication (pol3-01, pol30-52, rth1D/rad27D) mutations in both forward mutation and repeat-tract instability assays. This analysis indicated that the rfc1::Tn3 allele displays synthetic lethality with pol30, pol3, and rad27 mutations. Measurement of forward mutation frequencies in msh2D rfc1:Tn3 and pms1D rfc1:Tn3 strains indicated that the rfc1::Tn3 mutant displayed a mutation frequency that appeared nearly multiplicative with the mutation frequency exhibited by mismatch-repair mutants. In repeat-tract instability assays, however, the rfc1::Tn3 mutant displayed a tract instability phenotype that appeared epistatic to the phenotype displayed by mismatch-repair mutants. From these data we propose that defects in clamp loader function result in DNA replication errors, a subset of which are acted upon by the mismatch-repair system. UTATIONS in genes that are involved in DNA A major type of chromosomal instability that has been M replication and repair often result in chromo- identi®ed in yeast, in bacteria, and in cancer cells is somal instabilities such as base pair substitutions and repeat-tract instability (reviewed in Crouse 1996; Kolod- frameshifts as well as insertion, deletion, and re- ner 1996; Modrich and Lahue 1996; Sia et al. 1997b). arrangement events (i.e., Schaaper and Dunn 1987; This instability is thought to result mainly from the Aguilera and Klein 1988; Strand et al. 1993; McAlear failure to repair DNA slippage events that occur during et al. 1996; Tran et al. 1996; Green and Jinks-Robert- the replication of repetitive DNA sequences such as son 1997). Mutations that confer these phenotypes have those that contain mono-, di-, tri-, and tetranucleotide been directly identi®ed in Escherichia coli and Saccharo- repeats. In both yeast and mammalian cells, mutations myces cerevisiae using a variety of chromosome instability in mutS and mutL homolog mismatch-repair genes result assays (i.e., Feinstein and Low 1986; Aguilera and in a large increase (100- to 10,000-fold) in the rate of Klein 1988; Michaels et al. 1990; Jeyaprakash et al. repeat-tract instability (Strand et al. 1993; Tran et al. 1994). Such basic research approaches have also led to 1997; Umar et al. 1998). This increase is thought to be a better understanding of the underlying chromosome due to the inability of these mutants to repair small stability defects that have been observed in patients who loop insertion/deletion mutations that occur during suffer from particular inherited diseases. For example, DNA slippage (reviewed in Crouse 1996; Kolodner the phenotypes exhibited by mismatch-repair-defective 1996; Modrich and Lahue 1996; Sia et al. 1997b). In E. coli and S. cerevisiae strains provided evidence indi- yeast, mutations in DNA replication genes that encode cating that the underlying cause for a large percentage the polymerase processivity factor PCNA (POL30) and of hereditary nonpolyposis colorectal cancers was a de- the ¯ap endonuclease (RTH1/RAD27) have also been fect in mismatch repair (Levinson and Gutman 1987; shown to confer an increase in repeat-tract instability at Strand et al. 1993; Crouse 1996; reviewed in Kolodner a rate that is similar to that observed in mismatch-repair- 1996; Modrich and Lahue 1996). defective mutants (Johnson et al. 1995, 1996; Umar et al. 1996; Kokoska et al. 1998). Mutations in other DNA replication genes, such as those that encode the Pold (POL3), and Polε (POL2) DNA polymerases, however, Corresponding author: Eric Alani, Section of Genetics and Develop- ment, Cornell University, 459 Biotechnology Bldg., Ithaca, NY 14853- resulted in only modest increases in repeat-tract instabil- 2703. E-mail: [email protected] ity (Johnson et al. 1995; Tran et al. 1997; Kokoska et Genetics 151: 499±509 (February 1999) 500 Y. Xie, C. Counter and E. Alani al. 1998). These data have led to the proposal that some man 1989; Burgers 1991; Fien and Stillman 1992; DNA replication factors function directly in the mis- Stillman 1994). match-repair pathway to repair loop insertion/deletions that result from DNA slippage events while others act In this study we tested strains bearing the rfc1::Tn3 at the level of DNA replication to prevent the formation allele alone or in combination with mismatch repair and of these events (Kokoska et al. 1998; reviewed in Sia et DNA replication mutations for defects in chromosome al. 1997b). stability. As described below, the rfc1::Tn3 mutation con- The above observations, in conjunction with the obser- ferred a mutator phenotype that appeared multipli- vation that many cancer cells display repeat-tract insta- cative with mismatch-repair mutations. In repeat-tract bilities that are unlinked to previously identi®ed repair stability assays the rfc1::Tn3 mutation conferred an z10- and replication genes, encouraged us to initiate screens fold increase in the frequency of dinucleotide repeat- in yeast to identify chromosomal instability mutants tract instability that appeared to be epistatic to the phe- (Liu et al. 1995). We hoped to identify additional factors notype observed in mismatch-repair-defective mutants. that are involved in preventing mutagenic replication Taken together, our data are consistent with the idea errors by preventing DNA slippage events or by facilitat- that defects in clamp loader function result in DNA ing the repair of these slippages through mismatch- replication errors, a subset of which are identi®ed and repaired by the mismatch-repair system. repair mechanisms. In one screen we searched for mu- tants that displayed an increase in the frequency of both repeat-tract insertion/deletion and base pair substitu- MATERIALS AND METHODS tion/frameshift events; unfortunately, this screen only identi®ed mutants that displayed mutations in the pre- Media and chemicals: E. coli strains were grown in Luria- viously characterized mutS (MSH2) and mutL (PMS1, Bertani (LB) broth or on LB agar, which was supplemented m MLH1) homolog genes and the RAD27 gene (Y. Xie, with 100 g/ml ampicillin when required (Miller 1972). Yeast strains were grown in either YPD or minimal selective L. Schvaneveldt and E. Alani, unpublished data). In media (Rose et al. 1990). Selective media contained 0.7% yeast a second screen that is the focus of this article, we nitrogen base, 2% agar, 2% glucose, and 0.09% of a drop-out searched for mutants that were inviable in a double- mix that lacks the amino acid used for selection. Sporulation strand break-repair-de®cient (rad52) background and media (SPM) were prepared as described previously (Detloff also displayed a mutator phenotype (Counter et al. et al. 1991). When required, canavanine was included in mini- mal selective media at 60 mg/liter (Rose et al. 1990). 5-¯uoro- 1997). This screen was conducted on the basis of two orotic acid (5-FOA) was purchased from U.S. Biologicals and observations made in E. coli: used as described previously (Boeke et al. 1984). When re- quired, methyl-methane sulfonate (MMS; Aldrich Chemical, 1. Certain DNA replication mutants display chromo- Milwaukee) was included in YPD media at 0.017% (v/v). some instability defects such as chromosomal break- E. coli strains: DH5a [F9 phi80, dLacZD (lacZYA-argF), U169, 2 1 2 ages that are lethal in recombination-de®cient back- recA1, endA1, hsdr17 (r K, m K), lambda , thi1, gyrA, relA1] and KC8 (gammax14862, MK121, leuB600, trpC9830, pyrF::Tn5, grounds (Michel et al. 1997). hisB463, Dlacx74, StrA, galU, K) were used to amplify and 2. Strains that lack dam methylase, and are thus defec- manipulate all plasmids described in this article. tive in strand discrimination during mismatch re- S. cerevisiae strains: The genotypes of all strains used in pair, display a mutator phenotype and are inviable these studies are shown in Table 1. With the exception of in recombination de®cient (recA2) backgrounds NKY1068, EAY561, and DNR53, all strains were derived from the isogenic FY strain background (Winston et al. 1995). (McGraw and Marinus 1980). This inviability, DNR53 is also an S288C-derived strain that contains the which can be rescued by mutations in the mutS, rfc1::Tn3 allele (Burns et al. 1994; Counter et al. 1997). This mutL, and mutH mismatch-repair genes, is thought strain also contains a rad52::HIS3 mutation and is viable be- to be caused by unrepairable double-strand breaks cause it contains an ARS1, CEN4, URA3, plasmid bearing the in dam2 recA2 strains that form as the result
Recommended publications
  • Regulates Cellular Telomerase Activity by Methylation of TERT Promoter
    www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 5), pp: 7977-7988 Research Paper Tianshengyuan-1 (TSY-1) regulates cellular Telomerase activity by methylation of TERT promoter Weibo Yu1, Xiaotian Qin2, Yusheng Jin1, Yawei Li2, Chintda Santiskulvong3, Victor Vu1, Gang Zeng4,5, Zuofeng Zhang6, Michelle Chow1, Jianyu Rao1,5 1Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA 2Beijing Boyuantaihe Biological Technology Co., Ltd., Beijing, China 3Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA, USA 4Department of Urology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA 5Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, USA 6Department of Epidemiology, School of Public Health, University of California at Los Angeles, Los Angeles, CA, USA Correspondence to: Jianyu Rao, email: [email protected] Keywords: TSY-1, hematopoietic cells, Telomerase, TERT, methylation Received: September 08, 2016 Accepted: November 24, 2016 Published: December 15, 2016 ABSTRACT Telomere and Telomerase have recently been explored as anti-aging and anti- cancer drug targets with only limited success. Previously we showed that the Chinese herbal medicine Tianshengyuan-1 (TSY-1), an agent used to treat bone marrow deficiency, has a profound effect on stimulating Telomerase activity in hematopoietic cells. Here, the mechanism of TSY-1 on cellular Telomerase activity was further investigated using HL60, a promyelocytic leukemia cell line, normal peripheral blood mononuclear cells, and CD34+ hematopoietic stem cells derived from umbilical cord blood. TSY-1 increases Telomerase activity in normal peripheral blood mononuclear cells and CD34+ hematopoietic stem cells with innately low Telomerase activity but decreases Telomerase activity in HL60 cells with high intrinsic Telomerase activity, both in a dose-response manner.
    [Show full text]
  • A New Class of Disordered Elements Controls DNA Replication Through
    RESEARCH ARTICLE A new class of disordered elements controls DNA replication through initiator self-assembly Matthew W Parker1,2, Maren Bell2, Mustafa Mir2, Jonchee A Kao2, Xavier Darzacq2, Michael R Botchan2*, James M Berger1* 1Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, United States; 2Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States Abstract The initiation of DNA replication in metazoans occurs at thousands of chromosomal sites known as origins. At each origin, the Origin Recognition Complex (ORC), Cdc6, and Cdt1 co- assemble to load the Mcm2-7 replicative helicase onto chromatin. Current replication models envisage a linear arrangement of isolated origins functioning autonomously; the extent of inter- origin organization and communication is unknown. Here, we report that the replication initiation machinery of D. melanogaster unexpectedly undergoes liquid-liquid phase separation (LLPS) upon binding DNA in vitro. We find that ORC, Cdc6, and Cdt1 contain intrinsically disordered regions (IDRs) that drive LLPS and constitute a new class of phase separating elements. Initiator IDRs are shown to regulate multiple functions, including chromosome recruitment, initiator-specific co- assembly, and Mcm2-7 loading. These data help explain how CDK activity controls replication initiation and suggest that replication programs are subject to higher-order levels of inter-origin organization. DOI: https://doi.org/10.7554/eLife.48562.001 *For
    [Show full text]
  • Supplementary Table S1. Correlation Between the Mutant P53-Interacting Partners and PTTG3P, PTTG1 and PTTG2, Based on Data from Starbase V3.0 Database
    Supplementary Table S1. Correlation between the mutant p53-interacting partners and PTTG3P, PTTG1 and PTTG2, based on data from StarBase v3.0 database. PTTG3P PTTG1 PTTG2 Gene ID Coefficient-R p-value Coefficient-R p-value Coefficient-R p-value NF-YA ENSG00000001167 −0.077 8.59e-2 −0.210 2.09e-6 −0.122 6.23e-3 NF-YB ENSG00000120837 0.176 7.12e-5 0.227 2.82e-7 0.094 3.59e-2 NF-YC ENSG00000066136 0.124 5.45e-3 0.124 5.40e-3 0.051 2.51e-1 Sp1 ENSG00000185591 −0.014 7.50e-1 −0.201 5.82e-6 −0.072 1.07e-1 Ets-1 ENSG00000134954 −0.096 3.14e-2 −0.257 4.83e-9 0.034 4.46e-1 VDR ENSG00000111424 −0.091 4.10e-2 −0.216 1.03e-6 0.014 7.48e-1 SREBP-2 ENSG00000198911 −0.064 1.53e-1 −0.147 9.27e-4 −0.073 1.01e-1 TopBP1 ENSG00000163781 0.067 1.36e-1 0.051 2.57e-1 −0.020 6.57e-1 Pin1 ENSG00000127445 0.250 1.40e-8 0.571 9.56e-45 0.187 2.52e-5 MRE11 ENSG00000020922 0.063 1.56e-1 −0.007 8.81e-1 −0.024 5.93e-1 PML ENSG00000140464 0.072 1.05e-1 0.217 9.36e-7 0.166 1.85e-4 p63 ENSG00000073282 −0.120 7.04e-3 −0.283 1.08e-10 −0.198 7.71e-6 p73 ENSG00000078900 0.104 2.03e-2 0.258 4.67e-9 0.097 3.02e-2 Supplementary Table S2.
    [Show full text]
  • Arsenic Hexoxide Has Differential Effects on Cell Proliferation And
    www.nature.com/scientificreports OPEN Arsenic hexoxide has diferential efects on cell proliferation and genome‑wide gene expression in human primary mammary epithelial and MCF7 cells Donguk Kim1,7, Na Yeon Park2,7, Keunsoo Kang3, Stuart K. Calderwood4, Dong‑Hyung Cho2, Ill Ju Bae5* & Heeyoun Bunch1,6* Arsenic is reportedly a biphasic inorganic compound for its toxicity and anticancer efects in humans. Recent studies have shown that certain arsenic compounds including arsenic hexoxide (AS4O6; hereafter, AS6) induce programmed cell death and cell cycle arrest in human cancer cells and murine cancer models. However, the mechanisms by which AS6 suppresses cancer cells are incompletely understood. In this study, we report the mechanisms of AS6 through transcriptome analyses. In particular, the cytotoxicity and global gene expression regulation by AS6 were compared in human normal and cancer breast epithelial cells. Using RNA‑sequencing and bioinformatics analyses, diferentially expressed genes in signifcantly afected biological pathways in these cell types were validated by real‑time quantitative polymerase chain reaction and immunoblotting assays. Our data show markedly diferential efects of AS6 on cytotoxicity and gene expression in human mammary epithelial normal cells (HUMEC) and Michigan Cancer Foundation 7 (MCF7), a human mammary epithelial cancer cell line. AS6 selectively arrests cell growth and induces cell death in MCF7 cells without afecting the growth of HUMEC in a dose‑dependent manner. AS6 alters the transcription of a large number of genes in MCF7 cells, but much fewer genes in HUMEC. Importantly, we found that the cell proliferation, cell cycle, and DNA repair pathways are signifcantly suppressed whereas cellular stress response and apoptotic pathways increase in AS6‑treated MCF7 cells.
    [Show full text]
  • RFC1 Rabbit Pab
    Leader in Biomolecular Solutions for Life Science RFC1 Rabbit pAb Catalog No.: A1625 Basic Information Background Catalog No. This gene encodes the large subunit of replication factor C, a five subunit DNA A1625 polymerase accessory protein, which is a DNA-dependent ATPase required for eukaryotic DNA replication and repair. The large subunit acts as an activator of DNA Observed MW polymerases, binds to the 3' end of primers, and promotes coordinated synthesis of 140KDa both strands. It may also have a role in telomere stability. Alternatively spliced transcript variants encoding different isoforms have been noted for this gene. Calculated MW 128kDa Category Primary antibody Applications WB, IF Cross-Reactivity Human, Mouse, Rat Recommended Dilutions Immunogen Information WB 1:500 - 1:2000 Gene ID Swiss Prot 5981 P35251 IF 1:50 - 1:200 Immunogen Recombinant fusion protein containing a sequence corresponding to amino acids 400-700 of human RFC1 (NP_001191676.1). Synonyms RFC1;A1;MHCBFB;PO-GA;RECC1;RFC;RFC140 Contact Product Information www.abclonal.com Source Isotype Purification Rabbit IgG Affinity purification Storage Store at -20℃. Avoid freeze / thaw cycles. Buffer: PBS with 0.02% sodium azide,50% glycerol,pH7.3. Validation Data Western blot analysis of extracts of various cell lines, using RFC1 antibody (A1625) at 1:1000 dilution. Secondary antibody: HRP Goat Anti-Rabbit IgG (H+L) (AS014) at 1:10000 dilution. Lysates/proteins: 25ug per lane. Blocking buffer: 3% nonfat dry milk in TBST. Detection: ECL Basic Kit (RM00020). Exposure time: 180s. Western blot analysis of extracts of HeLa cells, using RFC1 antibody (A1625) at 1:1000 dilution.
    [Show full text]
  • The Second Subunit of DNA Polymerase Delta Is Required for Genomic Stability and Epigenetic Regulation1[OPEN]
    The Second Subunit of DNA Polymerase Delta Is Required for Genomic Stability and Epigenetic Regulation1[OPEN] Jixiang Zhang, Shaojun Xie, Jinkui Cheng, Jinsheng Lai, Jian-Kang Zhu, and Zhizhong Gong* State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (J.Z., J.C., Z.G.); Shanghai Center for Plant Stress Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China (S.X., J.-K.Z.); Department of Horticulture and Landscape Architecture, Purdue University, West Lafayette, Indiana 47906 (S.X., J.-K.Z.); and State Key Laboratory of Agrobiotechnology, China National Maize Improvement Center, Department of Plant Genetics and Breeding, China Agricultural University, Beijing 100193, China (J.L.) ORCID IDs: 0000-0002-1641-8650 (J.Z.); 0000-0002-6719-9814 (S.X.); 0000-0001-5134-731X (J.-K.Z.). DNA polymerase d plays crucial roles in DNA repair and replication as well as maintaining genomic stability. However, the function of POLD2, the second small subunit of DNA polymerase d, has not been characterized yet in Arabidopsis (Arabidopsis thaliana). During a genetic screen for release of transcriptional gene silencing, we identified a mutation in POLD2. Whole-genome bisulfite sequencing indicated that POLD2 is not involved in the regulation of DNA methylation. POLD2 genetically interacts with Ataxia Telangiectasia-mutated and Rad3-related and DNA polymerase a. The pold2-1 mutant exhibits genomic instability with a high frequency of homologous recombination. It also exhibits hypersensitivity to DNA-damaging reagents and short telomere length. Whole-genome chromatin immunoprecipitation sequencing and RNA sequencing analyses suggest that pold2-1 changes H3K27me3 and H3K4me3 modifications, and these changes are correlated with the gene expression levels.
    [Show full text]
  • Control of Genome Integrity by RFC Complexes; Conductors of PCNA Loading Onto and Unloading from Chromatin During DNA Replication
    Review Control of Genome Integrity by RFC Complexes; Conductors of PCNA Loading onto and Unloading from Chromatin during DNA Replication Yasushi Shiomi *and Hideo Nishitani * Graduate School of Life Science, University of Hyogo, Kamigori, Ako‐gun, Hyogo 678‐1297, Japan Correspondence: [email protected]‐hyogo.ac.jp (Y.S.); [email protected]‐hyogo.ac.jp (H.N.) Academic Editor: Eishi Noguchi Received: 28 November 2016; Accepted: 21 January 2017; Published: 26 January 2017 Abstract: During cell division, genome integrity is maintained by faithful DNA replication during S phase, followed by accurate segregation in mitosis. Many DNA metabolic events linked with DNA replication are also regulated throughout the cell cycle. In eukaryotes, the DNA sliding clamp, proliferating cell nuclear antigen (PCNA), acts on chromatin as a processivity factor for DNA polymerases. Since its discovery, many other PCNA binding partners have been identified that function during DNA replication, repair, recombination, chromatin remodeling, cohesion, and proteolysis in cell‐cycle progression. PCNA not only recruits the proteins involved in such events, but it also actively controls their function as chromatin assembles. Therefore, control of PCNA‐loading onto chromatin is fundamental for various replication‐coupled reactions. PCNA is loaded onto chromatin by PCNA‐loading replication factor C (RFC) complexes. Both RFC1‐RFC and Ctf18‐RFC fundamentally function as PCNA loaders. On the other hand, after DNA synthesis, PCNA must be removed from chromatin by Elg1‐RFC. Functional defects in RFC complexes lead to chromosomal abnormalities. In this review, we summarize the structural and functional relationships among RFC complexes, and describe how the regulation of PCNA loading/unloading by RFC complexes contributes to maintaining genome integrity.
    [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]
  • CIC De Novo Loss of Function Variants Contribute to Cerebral Folate Deficiency by Downregulating FOLR1 Expression Xuanye Cao,1 Annika Wolf,2 Sung-­Eun Kim,3 Robert M
    Neurogenetics J Med Genet: first published as 10.1136/jmedgenet-2020-106987 on 20 August 2020. Downloaded from Original research CIC de novo loss of function variants contribute to cerebral folate deficiency by downregulating FOLR1 expression Xuanye Cao,1 Annika Wolf,2 Sung- Eun Kim,3 Robert M. Cabrera,1 Bogdan J. Wlodarczyk,1 Huiping Zhu,4 Margaret Parker,3 Ying Lin,1 John W. Steele,1,5 Xiao Han,1 Vincent Th Ramaekers ,6 Robert Steinfeld,2,7 Richard H. Finnell,3,8,9 Yunping Lei 1 2 ► Additional material is ABSTRact epilepsy, ataxia and pyramidal tract signs. Folate is published online only. To view Background Cerebral folate deficiency (CFD) absorbed into the bloodstream via the gastrointes- please visit the journal online (http:// dx. doi. org/ 10. 1136/ syndrome is characterised by a low concentration of tinal tract mainly through two uptake systems: the jmedgenet- 2020- 106987). 5- methyltetrahydrofolate in cerebrospinal fluid, while reduced folate carrier 1 (RFC1; SLC19A1) and the folate levels in plasma and red blood cells are in the low proton- coupled folate transporter (PCFT).3 From For numbered affiliations see normal range. Mutations in several folate pathway genes, the bloodstream, folate binds to the folate receptor end of article. including FOLR1 (folate receptor alpha, FRα), DHFR alpha (FRα/FOLR1) on the basolateral endothelial (dihydrofolate reductase) and PCFT (proton coupled surface of the choroid plexus. Through receptor- Correspondence to Dr. Yunping Lei, Baylor College folate transporter) have been previously identified in mediated endocytosis and transcytosis, folate is of Medicine, Houston, Texas, patients with CFD. then transported across the blood- CSF- barrier USA; yunpinglei@ gmail.
    [Show full text]
  • Mutations in DNA Repair Genes Are Associated with Increased
    www.nature.com/scientificreports OPEN Mutations in DNA repair genes are associated with increased neoantigen burden and a distinct Received: 19 February 2018 Accepted: 19 December 2018 immunophenotype in lung Published: xx xx xxxx squamous cell carcinoma Young Kwang Chae1,2, Jonathan F. Anker 1, Michael S. Oh 1, Preeti Bais3, Sandeep Namburi3, Sarita Agte1, Francis J. Giles1,2 & Jefrey H. Chuang 3,4 Defciencies in DNA repair pathways, including mismatch repair (MMR), have been linked to higher tumor mutation burden and improved response to immune checkpoint inhibitors. However, the signifcance of MMR mutations in lung cancer has not been well characterized, and the relevance of other processes, including homologous recombination (HR) and polymerase epsilon (POLE) activity, remains unclear. Here, we analyzed a dataset of lung squamous cell carcinoma samples from The Cancer Genome Atlas. Variants in DNA repair genes were associated with increased tumor mutation and neoantigen burden, which in turn were linked with greater tumor infltration by activated T cells. The subset of tumors with DNA repair gene variants but without T cell infltration exhibited upregulation of TGF-β and Wnt pathway genes, and a combined score incorporating these genes and DNA repair status accurately predicted immune cell infltration. Finally, high neoantigen burden was positively associated with genes related to cytolytic activity and immune checkpoints. These fndings provide evidence that DNA repair pathway defects and immunomodulatory genes together lead to specifc immunophenotypes in lung squamous cell carcinoma and could potentially serve as biomarkers for immunotherapy. Immune checkpoint inhibitors have reshaped the landscape of treatment for multiple cancers, including squa- mous cell carcinoma (SCC) of the lung and other types of non-small cell lung cancer (NSCLC)1,2.
    [Show full text]
  • Androgen Receptor Signaling Regulates DNA Repair in Prostate Cancers
    Published OnlineFirst September 11, 2013; DOI: 10.1158/2159-8290.CD-13-0172 RESEARCH BRIEF Androgen Receptor Signaling Regulates DNA Repair in Prostate Cancers William R. Polkinghorn 1 , 4 , Joel S. Parker 10 , 11 , Man X. Lee 1 , Elizabeth M. Kass 2 , Daniel E. Spratt 1 , Phillip J. Iaquinta 1 , Vivek K. Arora 1 , 5 , Wei-Feng Yen 3 , Ling Cai 1 , Deyou Zheng 9 , Brett S. Carver 1 , 6 , Yu Chen 1 , 5 , Philip A. Watson 1 , Neel P. Shah 1 , Sho Fujisawa 8 , Alexander G. Goglia 4 , Anuradha Gopalan 7 , Haley Hieronymus 1 , John Wongvipat 1 , Peter T. Scardino 6 , Michael J. Zelefsky 1 , Maria Jasin 2 , Jayanta Chaudhuri 3 , Simon N. Powell 4 , and Charles L. Sawyers 1 ABSTRACT We demonstrate that the androgen receptor (AR) regulates a transcriptional pro- gram of DNA repair genes that promotes prostate cancer radioresistance, providing a potential mechanism by which androgen deprivation therapy synergizes with ionizing radiation. Using a model of castration-resistant prostate cancer, we show that second-generation antiandrogen therapy results in downregulation of DNA repair genes. Next, we demonstrate that primary prostate cancers dis- play a signifi cant spectrum of AR transcriptional output, which correlates with expression of a set of DNA repair genes. Using RNA-seq and ChIP-seq , we defi ne which of these DNA repair genes are both induced by androgen and represent direct AR targets. We establish that prostate cancer cells treated with ionizing radiation plus androgen demonstrate enhanced DNA repair and decreased DNA damage and furthermore that antiandrogen treatment causes increased DNA damage and decreased clonogenic survival.
    [Show full text]
  • Regulation of PCNA Cycling on Replicating DNA by RFC and RFC-Like Complexes
    ARTICLE https://doi.org/10.1038/s41467-019-10376-w OPEN Regulation of PCNA cycling on replicating DNA by RFC and RFC-like complexes Mi-Sun Kang 1,4, Eunjin Ryu1,2,4, Seung-Won Lee2,4, Jieun Park1, Na Young Ha1, Jae Sun Ra1, Yeong Jae Kim1,2, Jinwoo Kim1,2, Mohamed Abdel-Rahman3, Su Hyung Park1, Kyoo-young Lee1, Hajin Kim1,2, Sukhyun Kang1 & Kyungjae Myung1,2 Replication-Factor-C (RFC) and RFC-like complexes (RLCs) mediate chromatin engagement 1234567890():,; of the proliferating cell nuclear antigen (PCNA). It remains controversial how RFC and RLCs cooperate to regulate PCNA loading and unloading. Here, we show the distinct PCNA loading or unloading activity of each clamp loader. ATAD5-RLC possesses the potent PCNA unloading activity. ATPase motif and collar domain of ATAD5 are crucial for the unloading activity. DNA structures did not affect PCNA unloading activity of ATAD5-RLC. ATAD5-RLC could unload ubiquitinated PCNA. Through single molecule measurements, we reveal that ATAD5-RLC unloaded PCNA through one intermediate state before ATP hydrolysis. RFC loaded PCNA through two intermediate states on DNA, separated by ATP hydrolysis. Replication proteins such as Fen1 could inhibit the PCNA unloading activity of Elg1-RLC, a yeast homolog of ATAD5-RLC in vitro. Our findings provide molecular insights into how PCNA is released from chromatin to finalize DNA replication/repair. 1 Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea. 2 School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea. 3 Department of Ophthalmology, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
    [Show full text]