Matches in SemOpenAlex for { <https://semopenalex.org/work/W2071915265> ?p ?o ?g. }
- W2071915265 endingPage "36549" @default.
- W2071915265 startingPage "36544" @default.
- W2071915265 abstract "The rad17 gene ofSchizosaccharomyces pombe plays an important role as a checkpoint protein following DNA damage and during DNA replication. The human homologue of S. pombe rad17,Hrad17, was recently identified, but its function has not yet been established. Using the yeast two-hybrid system, we determined that HRad17 can interact with a nucleolar protein, NHP2L1. This interaction was also demonstrated biochemically, in human cells. Immunofluorescence studies revealed that HRad17 and NHP2L1 colocalize to the nucleolus, and immunogold labeling further resolved the location of NHP2L1 to the dense fibrillar component of the nucleolus. Interestingly, the localization of HRad17 in the nucleolus was altered in response to UV irradiation. These results provide some insight into the DNA damage and replication checkpoint mechanisms of HRad17. The rad17 gene ofSchizosaccharomyces pombe plays an important role as a checkpoint protein following DNA damage and during DNA replication. The human homologue of S. pombe rad17,Hrad17, was recently identified, but its function has not yet been established. Using the yeast two-hybrid system, we determined that HRad17 can interact with a nucleolar protein, NHP2L1. This interaction was also demonstrated biochemically, in human cells. Immunofluorescence studies revealed that HRad17 and NHP2L1 colocalize to the nucleolus, and immunogold labeling further resolved the location of NHP2L1 to the dense fibrillar component of the nucleolus. Interestingly, the localization of HRad17 in the nucleolus was altered in response to UV irradiation. These results provide some insight into the DNA damage and replication checkpoint mechanisms of HRad17. replication factor C open reading frame polymerase chain reaction phosphate-buffered saline monoclonal antibody high mobility group fibrillar center dense fibrillar component Cell cycle checkpoint proteins ensure that events of the cell cycle progress in an orderly fashion. Whenever DNA is damaged or incompletely replicated, cell cycle checkpoint proteins act to delay cell cycle progression until the aberrant DNA is repaired or replication is completed. Without this protective delay, cell division would occur in the presence of damaged or unreplicated DNA, which would result in genetic mutations or cell death (1Hartwell L.H. Weinert T.A. Science. 1989; 246: 629-634Crossref PubMed Scopus (2431) Google Scholar, 2Carr A.M. Semin. Cell Biol. 1995; 6: 65-72Crossref PubMed Scopus (51) Google Scholar, 3Murray A.W. Curr. Opin. Genet. Dev. 1995; 5: 5-11Crossref PubMed Scopus (138) Google Scholar). In Schizosaccharomyces pombe, therad1 +, rad3 +,rad9 +, rad17 +,rad26 +, and hus1 + genes are involved in DNA damage and replication checkpoints (3Murray A.W. Curr. Opin. Genet. Dev. 1995; 5: 5-11Crossref PubMed Scopus (138) Google Scholar, 4Kitazono A. Matsumoto T. Bioessays. 1998; 20: 391-399Crossref PubMed Scopus (13) Google Scholar). Similarly, in Saccharomyces cerevisiae, the G2-M DNA damage checkpoint is dependent on RAD9,RAD17, RAD24, MEC1/ESR1,RAD53, MEC3, and PDS1/ESP2(3Murray A.W. Curr. Opin. Genet. Dev. 1995; 5: 5-11Crossref PubMed Scopus (138) Google Scholar, 5Paulovich A.G. Margulies R.U. Garvik B.M. Hartwell L.H. Genetics. 1997; 145: 45-62Crossref PubMed Google Scholar). The function and structure of many of these checkpoint genes share a high degree of conservation between fission and budding yeast, because some of them are orthologous. For example, the S. pombe rad1 + gene is structurally related to RAD17of S. cerevisiae (6Siede W. Nusspaumer G. Portillo V. Rodriguez R. Friedberg E.C. Nucleic Acids Res. 1996; 24: 1669-1675Crossref PubMed Scopus (44) Google Scholar); the S. pombe rad3 + gene is a member of the phosphatidylinositol 3-kinase group and is a homologue of the S. cerevisiae RAD53gene (7Hoekstra M.F. Curr. Opin. Genet. Dev. 1997; 7: 170-175Crossref PubMed Scopus (147) Google Scholar); and the S. pombe rad17 + gene, which shares some sequence similarity with replication factor C (RF-C),1 is the homologue of the budding yeast RAD24 gene (8Griffiths D.J. Barbet N.C. McCready S. Lehmann A.R. Carr A.M. EMBO J. 1995; 14: 5812-5823Crossref PubMed Scopus (180) Google Scholar). Moreover, human homologues of S. pombe rad1 +, rad9 +,rad17 +, and hus1 + have been identified as Hrad1, Hrad9,Hrad17, and Hhus1, respectively (9Parker A.E. Van de Weyer I. Laus M.C. Oostveen I. Yon J. Verhasselt P. Luyten W.H. J. Biol. Chem. 1998; 273: 18332-18339Abstract Full Text Full Text PDF PubMed Scopus (93) Google Scholar, 10Lieberman H.B. Hopkins K.M. Nass M. Demetrick D. Davey S. Proc. Natl. Acad. Sci. U. S. A. 1996; 93: 13890-13895Crossref PubMed Scopus (107) Google Scholar, 11Parker A.E. Van de Weyer I. Laus M.C. Verhasselt P. Luyten W.H. J. Biol. Chem. 1998; 273: 18340-18346Abstract Full Text Full Text PDF PubMed Scopus (73) Google Scholar, 12Kostrub C.F. Knudsen K. Subramani S. Enoch T. EMBO J. 1998; 17: 2055-2066Crossref PubMed Scopus (100) Google Scholar), and theS. pombe rad3 + gene has two human homologues, ATM (ataxia telangiectasia mutated) and ATR (ataxia telangiectasia andRad3-related) (13Savitsky K. Bar-Shira A. Gilad S. Rotman G. Ziv Y. Vanagaite L. Tagle D.A. Smith S. Uziel T. Sfez S. et al.Science. 1995; 268: 1749-1753Crossref PubMed Scopus (2377) Google Scholar, 14Bentley N.J. Holtzman D.A. FLAGgs G. Keegan K.S. DeMaggio A. Ford J.C. Hoekstra M. Carr A.M. EMBO J. 1996; 15: 6641-6651Crossref PubMed Scopus (323) Google Scholar, 15Cimprich K.A. Shin T.B. Keith C.T. Schreiber S.L. Proc. Natl. Acad. Sci. U. S. A. 1996; 93: 2850-2855Crossref PubMed Scopus (226) Google Scholar). Genetic data in yeast and recent biochemical studies in yeast and human suggest that an early step in the DNA damage checkpoint response may involve the activation of Rad3p/ATM/ATR and Chk1 kinases. Phosphorylation of Chk1 does not occur when checkpoint radgenes are inactive, placing chk1 downstream ofrad genes in the cascade of events (16Walworth N.C. Bernards R. Science. 1996; 271: 353-356Crossref PubMed Scopus (350) Google Scholar). Furthermore, the Chk1 kinase has been shown to phosphorylate Cdc25c, and it is known that when phosphorylated on Ser216, Cdc25c is sequestered, and thus inhibited, by 14-3-3 (17Furnari B. Rhind N. Russell P. Science. 1997; 277: 1495-1497Crossref PubMed Scopus (475) Google Scholar, 18Sanchez Y. Wong C. Thoma R.S. Richman R. Wu Z. Piwnica-Worms H. Elledge S.J. Science. 1997; 277: 1497-1501Crossref PubMed Scopus (1126) Google Scholar, 19Peng C.Y. Graves P.R. Thoma R.S. Wu Z. Shaw A.S. Piwnica-Worms H. Science. 1997; 277: 1501-1505Crossref PubMed Scopus (1190) Google Scholar). This scenario reveals a potential link between the checkpoint Rad proteins and Cdk regulation through Cdc25 (20Weinert T. Science. 1997; 277: 1450-1451Crossref PubMed Scopus (116) Google Scholar, 21Wang J.Y. Curr Opin Cell Biol. 1998; 10: 240-247Crossref PubMed Scopus (76) Google Scholar). In the last few months, there have been three independent reports of the cloning of Hrad17, the human homologue of theRad17 gene from S. pombe (11Parker A.E. Van de Weyer I. Laus M.C. Verhasselt P. Luyten W.H. J. Biol. Chem. 1998; 273: 18340-18346Abstract Full Text Full Text PDF PubMed Scopus (73) Google Scholar, 22Bao S. Chang M.S. Auclair D. Sun Y. Wang Y. Wong W.K. Zhang J. Liu Y. Qian X. Sutherland R. Magi-Galluzi C. Weisberg E. Cheng E.Y. Hao L. Sasaki H. Campbell M.S. Kraeft S.K. Loda M. Lo K.M. Chen L.B. Cancer Res. 1999; 59: 2023-2028PubMed Google Scholar, 23Li L. Peterson C.A. Kanter-Smoler G. Wei Y.F. Ramagli L.S. Sunnerhagen P. Siciliano M.J. Legerski R.J. Oncogene. 1999; 18: 1689-1699Crossref PubMed Scopus (25) Google Scholar). The HRad17 protein has a significant amino acid identity with S. pombeRad17p and has been demonstrated to interact physically with HRad1 but not with HRad9 (11Parker A.E. Van de Weyer I. Laus M.C. Verhasselt P. Luyten W.H. J. Biol. Chem. 1998; 273: 18340-18346Abstract Full Text Full Text PDF PubMed Scopus (73) Google Scholar). More recently, we showed that Hrad17 is highly expressed in human testis and tumor tissues (22Bao S. Chang M.S. Auclair D. Sun Y. Wang Y. Wong W.K. Zhang J. Liu Y. Qian X. Sutherland R. Magi-Galluzi C. Weisberg E. Cheng E.Y. Hao L. Sasaki H. Campbell M.S. Kraeft S.K. Loda M. Lo K.M. Chen L.B. Cancer Res. 1999; 59: 2023-2028PubMed Google Scholar), and Liet al. (23Li L. Peterson C.A. Kanter-Smoler G. Wei Y.F. Ramagli L.S. Sunnerhagen P. Siciliano M.J. Legerski R.J. Oncogene. 1999; 18: 1689-1699Crossref PubMed Scopus (25) Google Scholar) confirmed the cell-cycle checkpoint functionality of Hrad17, using a complementation assay in S. pombe rad17 mutants. In the present work, we investigated the intracellular localization of HRad17, and we report that it predominantly localizes to the nucleolus. To further investigate the possible biological roles of Hrad17, yeast two-hybrid screening was performed to identify potential HRad17 binding partners. We found that HRad17 interacts and colocalizes with NHP2L1, the human homologue of an essential S. cerevisiae nuclear protein, NHP2 (24Kolodrubetz D. Burgum A. Yeast. 1991; 7: 79-90Crossref PubMed Scopus (26) Google Scholar). In addition, the localization of HRad17 was found to be sensitive to UV irradiation, as the protein then translocated out of the nucleoli and assumed a more diffuse nuclear distribution. These results suggest a functional link between HRad17 and NHP2L1 and implicate the nucleolus as an important site in the DNA damage checkpoint response. The entire open reading frame (ORF) of HRad17 was amplified by PCR with a pair of primers, 5′-CTGGATCCGCATGAATCAGGTAACAGACT-3′ and 5′-GAAGTCGACTATGTCCCATCACTCTCGT-3′. The PCR products were digested byBamHI and SalI and cloned into vector pAS-2 (CLONTECH, Palo Atlo, CA). The ORF sequence of human Chk1 was retrieved from the GenBankTM data base (accession number AF016582), amplified by PCR, and cloned into vector pACT-2 (CLONTECH). The nucleotide sequence of these constructs was confirmed by automated sequencing. The human testis matchmaker cDNA library was purchased fromCLONTECH. The procedures for yeast two-hybrid screening and elimination of false positives were performed exactly according to the manufacturer's instructions. Briefly, six million yeast transformants were screened, and 20 positive clones were selected based on His3 and lacZ reporter gene expression. After cycloheximide counterselection and yeast mating to eliminate false positives, a total of six clones remained positive. Plasmids of these six positive clones were isolated from yeast strain CG1945 and transformed into Escherichia coli strain KC8. Automated sequencing allowed for the identification of two of these six clones as NHP2L1 and two others as KIP. We then proceeded with yeast cotransformation experiments to confirm the two-hybrid interaction. Hrad17 was cut from the pAS-2-Hrad17 construct by BamHI and SalI and then cloned into pQE32 (Qiagen, Hilden, Germany). The ORF region of NHP2L1 was amplified by PCR and cloned into pQE30 (Qiagen). For cloning NHP2L1 and KIP coding sequences in frame into vector pFLAG-CMV2 (Kodak, New Haven, CT), PCR was used to amplify the ORFs corresponding to each gene. The amplified fragments were cloned into the NotI andSalI sites of pFLAG-CMV2. The primers were as follows: for NHP2L1, 5′-TATGCGGCCGCGATGACTGAGGCTGATGTGAAT-3′ and 5′-GCGGTCGACTTAGACTAAGAGCCTTTCAAT-3′, and for KIP, 5′-AATGCGGCCGCGATGGGGGGCTCGGGCAGTCGC-3′ and 5′-TGCGTCGACTCACAGGACAATCTTAAAGGA-3′. The nucleotide sequence of these constructs was confirmed by automated DNA sequencing. The pQE constructs were transformed into E. coli. strain M15 (pREP4). The expression of His tag fusion protein was induced with 1 mmisopropyl-1-thio-d-galactopyranoside at 37 °C for 3–5 h. The bacterial pellets were sonicated in PBS with protease inhibitors (50 mm Tris-HCl, pH 8.0, 150 mm NaCl, 0.5% Nonidet P-40, 10 mm NaF, 1 mm sodium orthovanadate, 1 mm dithiothreitol, 10 μg/ml leupeptin, 10 μg/ml aprotinin, 10 μg/ml trypsin/chymotrypsin inhibitor, 5 μg/ml pepstatin A, and 1 mm phenylmethylsulfonyl fluoride) and then lysed with 1% Triton X-100. The purification of His tag fusion proteins was carried out by His.Bind Quick columns (Novagen, Madison, WI). Monoclonal antibodies against HRad17 (31E9) were developed by immunizing mice with His6-HRad17 and screening the culture supernatants with recombinant HRad17 protein using ELISA. NHP2L1 polyclonal antiserum (R86) was raised in rabbits against two synthetic peptides derived from the amino acid sequences, KQLRKGANEATKTLNRG and SQLKQQIQSIQQSIERLLV from NHP2L1. The polyclonal antiserum was further purified by Affi-10 gel (Bio-Rad) column coupled with His-tagged NHP2L1. After extensive washing with 10 mmTris (pH 7.5) and 10 mm Tris (pH 7.5), 500 mmNaCl, the antibodies were eluted with 100 mm glycine (pH 2.5) and 100 mm triethylamine (pH 11) and dialyzed with PBS for two days. Human HeLa cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 50 units/ml penicillin, and 50 μg/ml streptomycin (Life Technologies, Inc.) HeLa cell lysates were prepared from cultured cells grown to 75–90% confluence on 10-cm dishes. Following two washes with cold PBS, the cells were scraped and solubilized for 1 h in 0.5 ml of lysis buffer with protease inhibitors (50 mm Tris-HCl, pH 8.0, 150 mm NaCl, 0.5% Nonidet P-40, 10 mmNaF, 1 mm sodium orthovanadate, 1 mmdithiothreitol, 10 μg/ml leupeptin, 10 μg/ml aprotinin, 10 μg/ml trypsin/chymotrypsin inhibitor, 5 μg/ml pepstatin A, and 1 mm phenylmethylsulfonyl fluoride), and spun to collect cell extracts (supernatants). For immunoprecipitation assays, the whole cell extract was diluted with the same volume of double distilled H2O to reduce ionic strength interference. Antibodies were incubated with cell extract at 4 °C for 2 h. A protein A-Sepharose and protein G-Sepharose mixture (20 μl 1:1) (Amersham Pharmacia Biotech) was added and incubated overnight at 4 °C. The immunoprecipitate complexes were washed three times with lysis buffer (1% Triton X-100, 1% bovine hemoglobin, 1 mm iodoacetamide, 10 μg/ml aprotinin, 10 μg/ml trypsin inhibitor, 1 mm phenylmethylsulfonyl fluoride, prepared in 10 mm Tris-HCl, pH 8.0, 140 mm NaCl, 0.025% NaN3), solubilized in SDS sample buffer (0.2 m Tris-HCl, pH 6.8, 2% SDS, 10% glycerol, 0.6% β-mercaptoethanol), separated by SDS-polyacrylamide gel electrophoresis, and electrophoretically transferred onto nitrocellulose membranes. The membranes were blocked in PBS with 0.1% Tween 20 and 5% dried milk, probed with anti-HRad17 antibody (1:500 dilution), and processed with the ECL Western blotting detection system (Amersham Pharmacia Biotech). For cultures used in immunoprecipitation assays, HeLa cells were grown to approximately 60–80% confluence on 10-cm dishes. 20 μg of DNA in 40 μl of Superfect reagent (Qiagen) were added to the cells. For cultures used for indirect immunofluorescence detection experiments, HeLa cells were seeded onto glass coverslips, placed in 3-cm dishes, and transfected with 1–2 μg of DNA in 5 μl of Superfect reagent. Following incubation at 37 °C for 2–3 h, the DNA-Superfect complex was replaced with Dulbecco's modified Eagle's medium containing 10% fetal bovine serum. 48 h after transfection, the cells were either fixed for microscopic analysis (immunofluorescence) or lysed, and cell extracts were prepared for immunoprecipitation. For indirect immunofluorescence, cells grown on coverslips were fixed in cold methanol (−20 °C) for 10 min, washed in PBS, and incubated with polyclonal R86 antiserum or monoclonal 31E9 antibody at 37 °C for 1 h. After rinsing with PBS, anti-rabbit or anti-mouse antibodies conjugated with rhodamine or fluorescein isothiocyanate (Jackson Immuno Research, West Grove, PA) were applied at 37 °C for 30 min. Subsequently, DNA was labeled with 4′,6-diamidino-2-phenylindole, and coverslips were mounted in antifade solution (Molecular Probes Inc., Eugene, OR). For double immunofluorescence labeling, both primary or secondary antibodies were mixed and applied together. Confocal images were recorded using a LSM 410 confocal laser scanning microscope (Carl Zeiss, Germany) and printed with the Fujix Pictrography 3000 color printer (Fujifilm, Japan) using Adobe PhotoShop software (Adobe Systems, Mountain View, CA). For immunogold labeling, HeLa cells were fixed for 30 min with 0.1% glutaraldehyde in Zamboni fixative (2% paraformadehyde and 1:6 dilution of a saturated picric acid solution in 0.1 m PBS). Samples were then cryoprotected and frozen in liquid nitrogen as described (25Xu Y. Slayter H.S. J. Histochem. Cytochem. 1994; 42: 1365-1376Crossref PubMed Scopus (28) Google Scholar). Ultrathin cryosections were prepared and incubated with the primary antiserum for 30 min, followed by incubation for 30 min with goat anti-rabbit IgG and gold conjugates of 10 nm gold particle size (10 μg/ml). Sections were thoroughly washed with PBS and stained for 2 min in 1% neutral uranyl acetate and then 2 min in 4% unbuffered uranyl acetate. Samples were mounted in a thin film of 1.25% methylcellulose (Fluka Chemical Co., Donkokomo, NY) and examined with a JEOL 100-cx transmission electron microscope (JEOL USA, Peabody, MA). UV doses were delivered with a single pulse using the UV Stratalinker 2400 (Stratagene, La Jolla, CA). Culture medium was removed prior to UV irradiation and replaced immediately after treatment. In most of these experiments, a dose of 40 J/m2 was used, and the cells were observed after an additional 2 h of culture. To address the subcellular distribution of HRad17, monoclonal antibodies (mAbs) against HRad17 were generated. The mAb 31E9 was found to recognize a protein of 75 kDa by Western blotting of HeLa total cell lysates (Fig. 1 A, lane 1) and immunoprecipitation (lanes 2 and 3). This corresponds to the predicted molecular mass of HRad17. The two major bands below Hrad17 come from the IgG used for immunoprecipitation, as confirmed by an experiment where a control antibody (25G10) was used (not shown). The 31E9 antibody also recognizes a single 75-kDa band in extracts of insect cells expressing HRad17 (data not shown). Immunostaining analysis of HeLa cells with the 31E9 antibody showed that HRad17 localized to the nucleus, exhibiting a punctate pattern with concentration mostly in the nucleoli (Fig. 1 B). Using the yeast two-hybrid method with HRad17 as the bait protein, approximately six million human testis cDNA library transformants were screened, and a total of six colonies were found that wereHIS3 and lacZ positive. Two of them were identified by sequencing as NHP2L1, representing two different mRNA transcripts, 0.7 and 1.3 kilobases, as described previously by others (26Saito H. Fujiwara T. Shin S. Okui K. Nakamura Y. Cytogenet. Cell Genet. 1996; 72: 191-193Crossref PubMed Scopus (16) Google Scholar). Another two colonies were identified asCIB/KIP (27Naik U.P. Patel P.M. Parise L.V. J. Biol. Chem. 1997; 272: 4651-4654Abstract Full Text Full Text PDF PubMed Scopus (241) Google Scholar, 28Wu X. Lieber M.R. Mutat. Res. 1997; 385: 13-20Crossref PubMed Scopus (63) Google Scholar). The specificity of the interaction between KIP/NHP2L1 and HRad17 was further confirmed by cotransformation of HRad17 with NHP2L1 and KIP into yeast strain CG1945. In addition, pACT-2-PCNA and pACT-2-Chk1 were constructed as controls. The result showed that NHP2L1 and KIP can specifically interact with HRad17 using the yeast two-hybrid method (Fig. 2 A). To investigate whether NHP2L1 and KIP interact with HRad17 in mammalian cells, immunoprecipitation experiments were carried out. HeLa cells were transfected with N-terminal FLAG-tagged NHP2L1 or KIP. Cell lysates were immunoprecipitated with either 31E9 or anti-FLAG mAbs and analyzed by Western blotting with corresponding antibodies. Immunoprecipitation with the 31E9 anti-HRad17 antibody followed by Western blotting with anti-FLAG antibody revealed Hrad17 and NHP2L1 in the same immunocomplex (Fig. 2 C). The opposite experiment, immunoprecipitation with anti-FLAG antibody and Western blotting with 31E9, also showed an association between HRad17 and NHP2L1 (Fig. 2 D). As was the case for Fig. 1, the two major bands seen in Fig. 2 (C and D) come from the IgG used for immunoprecipitation, as confirmed by a set of experiments where lysates were substituted for lysis buffer alone (not shown). These results demonstrate that HRad17 and NHP2L1 can interact in HeLa cells. However, we were unable to detect co-immunoprecipitation of HRad17 and KIP (Fig. 2, C and D). Although NHP2 shares some similarity withNHP2L1, a search of the GenBankTM data base revealed an open reading frame from S. cerevisiae even more closely related to NHP2L1. S. cerevisiae NHP2 and human NHP2L1 have only 33% identity (Fig. 3), but this novel S. cerevisiae ORF (GenBankTMaccession number S15037) is 69% identical to human NHP2L1. Therefore, the homology of NHP2L1 to this new gene is likely to be much more relevant than its homology to NHP2. Similarly, we found a C. elegans sequence (Swiss-Prot number Q21568) that is 77% identical to NHP2L1, suggesting that this is a highly conserved protein. Because HRad17 and NHP2L1 could interact in HeLa cells, we set out to examine whether NHP2L1 localized in the nucleolus. First, HeLa cells were transfected with FLAG-tagged NHP2L1. Immunofluorescent labeling with the anti-FLAG antibody showed that NHP2L1 indeed located to the nucleoli (Fig. 4 A). Second, a GFP-NHP2L1 fusion construct was made to transfected into HeLa cells. Again, the NHP2L1 green signal was found to reside in the nucleoli (data not shown). Third, a rabbit polyclonal antiserum, R86, was raised against NHP2L1 using two NHP2L1 synthetic peptides, and an affinity-purified polyclonal antibody was prepared. The affinity-purified antibody specifically recognized one band in the nuclear preparation of HeLa cells, whose size corresponds to that of NHP2L1 (Fig. 4 E). Staining of HeLa cells with this affinity-purified antibody predominantly localized to the nucleoli, with only fainter, punctate labeling in other nuclear regions (Fig. 4 B). Control experiments using preimmunized rabbit serum and synthetic peptides for immunocompetition did not show a nucleolar signal (Fig. 4, Cand D). These results demonstrate that NHP2L1 is a nucleolar protein. To more precisely determine where HRad17 and NHP2L1 resided within the nucleolus, immunogold labeling of ultrathin cryosections prepared from HeLa cells was performed. The anti-HRad17 monoclonal antibody 31E9 was incompatible with our electron microscopy fixation procedures. However, immunogold labeling with polyclonal anti-NHP2L1 antiserum R86 showed that NHP2L1 is largely concentrated in the dense fibrillar component of the nucleolus (Fig. 5). No signal was found in the same region when preimmunized serum was used. To test whether HRad17 and NHP2L1 colocalized in the nucleolus, double staining experiments were performed. Using simultaneous labeling of HeLa cells with anti-HRad17 and anti-NHP2L1 antibodies followed by rhodamine and fluorescein isothiocyanate-conjugated secondary antibodies, large overlapping areas of staining could be visualized in the nucleoli. These are shown inyellow in the superimposed confocal images (Fig. 6). To investigate whether the localization of HRad17 may be responsive to DNA damage, HeLa cells were irradiated with UV light (40 J/m 2M.-S. Chang, H. Sasaki, M. S. Campbell, S.-K. Kraeft, R. Sutherland, C.-Y. Yang, Y. Liu, D. Auclair, L. Hao, H. Sonoda, L. H. Ferland, and L. B. Chen, unpublished observations. ) and cultured for another 2 h. Interestingly, HRad17 staining was no longer seen in the nucleoli, although the nucleolar structures could still be detected by phase contrast microscopy. Instead, we observed only the punctate nuclear labeling pattern after UV treatment (Fig. 7 B). The total amount of HRad17 protein appeared unchanged (Fig. 7 A), however, suggesting that the disappearance of HRad17 nucleolar labeling was caused by protein redistribution rather than degradation. This effect was also observed with lower UV doses, but complete disappearance of Hrad17 nucleolar labeling occurred only after longer post-irradiation incubation periods (e.g. 16 h at 20 J/m2). In control experiments, MPP10, a human U3 small nucleolar ribonucleoprotein component (29Westendorf J.M. Konstantinov K.N. Wormsley S. Shu M.D. Matsumoto-Taniura N. Pirollet F. Klier F.G. Gerace L. Baserga S.J. Mol. Biol. Cell. 1998; 9: 437-449Crossref PubMed Scopus (50) Google Scholar) still localized to the nucleolus after UV irradiation (Fig. 7, C and D). Finally, redistribution of HRad17 was also be observed when the cells were treated with methyl methanesulfonate, mitomycin C, adriamycin, or a combination of γ irradiation andcis-platinum, 2M.-S. Chang, H. Sasaki, M. S. Campbell, S.-K. Kraeft, R. Sutherland, C.-Y. Yang, Y. Liu, D. Auclair, L. Hao, H. Sonoda, L. H. Ferland, and L. B. Chen, unpublished observations.suggesting that this translocation of HRad17 out of the nucleolus indeed accompanies DNA damage. In contrast, the localization of NHP2L1 remained nucleolar when these DNA damage stimuli were used (data not shown). We recently isolated an Hrad17 cDNA clone on the basis of its high levels of expression in human testis and tumor tissues (22Bao S. Chang M.S. Auclair D. Sun Y. Wang Y. Wong W.K. Zhang J. Liu Y. Qian X. Sutherland R. Magi-Galluzi C. Weisberg E. Cheng E.Y. Hao L. Sasaki H. Campbell M.S. Kraeft S.K. Loda M. Lo K.M. Chen L.B. Cancer Res. 1999; 59: 2023-2028PubMed Google Scholar). In the present work, we used the yeast two-hybrid method to identify interacting proteins. Our screen revealed two HRad17 interacting proteins, NHP2L1 and KIP. KIP is related to the phosphatase calcineurin B and was reported to bind to DNA-dependent protein kinase (28Wu X. Lieber M.R. Mutat. Res. 1997; 385: 13-20Crossref PubMed Scopus (63) Google Scholar), which is a relative of Rad3p/ATM/ATR kinases, with a C-terminal domain homologous to phosphatidylinositol 3-kinase and is required for V(D)J rearrangements of immunoglobulin genes, recombination, and repair of radiation-induced double-stranded breaks (30Hartley K.O. Gell D. Smith G.C. Zhang H. Divecha N. Connelly M.A. Admon A. Lees-Miller S.P. Anderson C.W. Jackson S.P. Cell. 1995; 82: 849-856Abstract Full Text PDF PubMed Scopus (672) Google Scholar). Therefore, KIP may participate in the regulation of DNA double-strand break repair via the regulation of phosphorylation and dephosphorylation processes (28Wu X. Lieber M.R. Mutat. Res. 1997; 385: 13-20Crossref PubMed Scopus (63) Google Scholar). Although HRad17 and KIP could clearly interact within the confines of the yeast two-hybrid system, we could not detect such an association in human cells by co-immunoprecipitation. Although this does not rule out that anin vivo interaction may occur in human cells, we concentrated our efforts on the other Hrad17-binding protein, NHP2L1. NHP2L1 is a putative human homologue of the NHP2gene in S. cerevisiae (26Saito H. Fujiwara T. Shin S. Okui K. Nakamura Y. Cytogenet. Cell Genet. 1996; 72: 191-193Crossref PubMed Scopus (16) Google Scholar). NHP2 is related to the high mobility group (HMG) proteins based on the contents of basic and acidic amino acids, and is essential in yeast (24Kolodrubetz D. Burgum A. Yeast. 1991; 7: 79-90Crossref PubMed Scopus (26) Google Scholar). Typically, HMG proteins have a molecular mass that is <30 kDa and are highly charged. HMG domains can bind to a variety of non-B-DNA structures, such as B-Z DNA junction and platinated DNA (31Grosschedl R. Giese K. Pagel J. Trends Genet. 1994; 10: 94-100Abstract Full Text PDF PubMed Scopus (736) Google Scholar, 32Baxevanis A.D. Landsman D. Nucleic Acids Res. 1995; 23: 1604-1613Crossref PubMed Scopus (189) Google Scholar). An RNA binding motif that was hypothesized to deliver additional activity to the ribosome was also found in NHP2 (33Koonin E.V. Bork P. Sander C. Nucleic Acids Res. 1994; 22: 2166-2167Crossref PubMed Scopus (102) Google Scholar). No obvious HMG domain or RNA-binding motif can be identified in human NHP2L1 protein. Two other homologues of humanNHP2L1 were identified in C. elegans and S. cerevisiae (Fig. 3). To assess the importance of the new yeastNHP2L1, we used the single step method (34Baudin A. Ozier-Kalogeropoulos O. Denouel A. Lacroute F. Cullin C. Nucleic Acids Res. 1993; 21: 3329-3330Crossref PubMed Scopus (1127) Google Scholar) to create a null mutant of NHP2L1 in S. cerevisiae. After screening more than 200 transformants containing HIS3 orTRP1 marker genes, however, we were unable to obtain a null mutant of NHP2L1 in S. cerevisiae,2 suggesting that NHP2L1 may be an essential gene in yeast. HRad17 and NHP2L1 were found to localize to the nucleolus. The nucleolus is the site of rDNA transcription, processing of rDNA transcripts, and formation of preribosomal particles (35Shaw P.J. Jordan E.G. Annu. Rev. Cell Dev. Biol. 1995; 11: 93-121Crossref PubMed Scopus (409) Google Scholar). It can be divided into three compartments, the fibrillar centers (FCs), the dense fibrillar component (DFC), and the granular component. Early work showed that the FC is a reservoir for rDNA and RNA polymerase I (36Thiry M. Thiry-Blaise L. Eur J. Cell Biol. 1989; 50: 235-243PubMed Google Scholar,37Scheer U. Rose K.M. Proc. Natl. Acad. Sci. U. S. A. 1984; 81: 1431-1435Crossref PubMed Google Scholar). The DFC is thought to be the likely site of rRNA transcription, because nascent Br-UTP-labeled rRNA was detected at the boundary of FC and DFC (38Dundr M. Raska I. Exp. Cell Res. 1993; 208: 275-281Crossref PubMed Scopus (107) Google Scholar, 39Hozak P. Cook P.R. Schofer C. Mosgoller W. Wachtler F. J. Cell Sci. 1994; 107: 639-648PubMed Google Scholar). However, another line of evidence attributed the site of rRNA transcription to the FC region (40Thiry M. J. Cell Sci. 1993; 105: 33-39PubMed Google Scholar). Later rRNA processing and preribosome assembly take place in the granular component region (35Shaw P.J. Jordan E.G. Annu. Rev. Cell Dev. Biol. 1995; 11: 93-121Crossref PubMed Scopus (409) Google Scholar). Immunogold labeling of NHP2L1 showed that it resides primarily in the DFC region of nucleolus (Fig. 4), suggesting that the function of NHP2L1 may be related to rRNA transcription or processing. We were unable to determine the precise location of Hrad17 within the nucleolus. However, its localization in the nucleolus suggests it may play a role in one or more nucleolar processes. Recently, it has been reported that nucleolar function may be involved in the aging process. In aging S. cerevisiae cells, the Sir complex, consisting of Sir2p, Sir3p, and Sir4p, is found to relocalize from the telomere to the nucleolus, and the number of the extrachromosomal rDNA circles is dramatically increased (41Guarente L. Genes Dev. 1997; 11: 2449-2455Crossref PubMed Scopus (88) Google Scholar, 42Johnson F.B. Marciniak R.A. Guarente L. Curr. Opin. Cell Biol. 1998; 10: 332-338Crossref PubMed Scopus (64) Google Scholar, 43Sinclair D.A. Guarente L. Cell. 1997; 91: 1033-1042Abstract Full Text Full Text PDF PubMed Scopus (1187) Google Scholar, 44Sinclair D.A. Mills K. Guarente L. Science. 1997; 277: 1313-1316Crossref PubMed Scopus (328) Google Scholar). In humans, the Werner syndrome is an autosomal recessive disorder characterized by premature aging. The Werner syndrome protein, which encodes an ATP-dependent DNA helicase, was found to localize within the nucleolus in human cells (45Marciniak R.A. Lombard D.B. Johnson F.B. Guarente L. Proc. Natl. Acad. Sci. U. S. A. 1998; 95: 6887-6892Crossref PubMed Scopus (180) Google Scholar). The S. pombe Rad17p protein is required for DNA damage and replication checkpoints. If HRad17 plays a similar checkpoint function, its localization raises the possibility of a heretofore unsuspected role for the nucleolus in the DNA damage response. DNA damage induced by UV light has been well studied, and a variety of DNA repair mechanisms are triggered to restore the genomic integrity in most organisms (46Park M.S. Knauf J.A. Pendergrass S.H. Coulon C.H. Strniste G.F. Marrone B.L. MacInnes M.A. Proc. Natl. Acad. Sci. U. S. A. 1996; 93: 8368-8373Crossref PubMed Scopus (48) Google Scholar). Given the rapidity of the cellular response to genome damage one would expect that both the damage sensing mechanisms and the immediate downstream elements of the signal transduction pathway would be constitutively present within the nucleus. Indeed, both ATM and Hrad1, two human proteins with known cell cycle checkpoint functionalities (7Hoekstra M.F. Curr. Opin. Genet. Dev. 1997; 7: 170-175Crossref PubMed Scopus (147) Google Scholar, 47Freire R. Murguia J.R. Tarsounas M. Lowndes N.F. Moens P.B. Jackson S.P. Genes Dev. 1998; 12: 2560-2573Crossref PubMed Scopus (102) Google Scholar), display constant protein levels after such DNA damaging stresses as UV or γ irradiation or treatment with the radiomimetic drug neocarzinostatin (47Freire R. Murguia J.R. Tarsounas M. Lowndes N.F. Moens P.B. Jackson S.P. Genes Dev. 1998; 12: 2560-2573Crossref PubMed Scopus (102) Google Scholar, 48Brown K.D. Ziv Y. Sadanandan S.N. Chessa L. Collins F.S. Shiloh Y. Tagle D.A. Proc. Natl. Acad. Sci. U. S. A. 1997; 94: 1840-1845Crossref PubMed Scopus (150) Google Scholar). Our finding of a stable level of Hrad17 protein upon UV irradiation fits well with these observations and suggests that perhaps the entire apparatus of DNA damage sensing and repair is constitutively present in the nucleus, circumventing the need for time-consuming protein synthesis in the response to DNA damage. However, ATM and Hrad1 display punctate nuclear labeling patterns excluding nucleoli in the basal state, and this pattern did not change when the cells were treated with DNA damaging agents (49Watters D. Khanna K.K. Beamish H. Birrell G. Spring K. Kedar P. Gatei M. Stenzel D. Hobson K. Kozlov S. Zhang N. Farrell A. Ramsay J. Gatti R. Lavin M. Oncogene. 1997; 14: 1911-1921Crossref PubMed Scopus (169) Google Scholar).2 This contrasts with the observations reported here of a strong Hrad17 staining in the nucleoli, in addition to fainter, punctate nuclear labeling. Most striking is the disappearance of Hrad17 nucleolar labeling following DNA damage, apparently because of a redistribution of Hrad17 protein to its extranucleolar pool. The significance of this redistribution remains to be determined, but it may be noteworthy that other proteins involved in DNA repair have been shown to redistribute in response to UV irradiation, including theXeroderma pigmentosum type G protein and BRCA1 (46Park M.S. Knauf J.A. Pendergrass S.H. Coulon C.H. Strniste G.F. Marrone B.L. MacInnes M.A. Proc. Natl. Acad. Sci. U. S. A. 1996; 93: 8368-8373Crossref PubMed Scopus (48) Google Scholar, 50Scully R. Chen J. Ochs R.L. Keegan K. Hoekstra M. Feunteun J. Livingston D.M. Cell. 1997; 90: 425-435Abstract Full Text Full Text PDF PubMed Scopus (809) Google Scholar). One plausible function of HRad17 in the cell cycle checkpoint process is suggested by its similarity to RF-C components. Human RF-C is a complex composed of five proteins of molecular masses 36, 37, 38, 40, and 140 kDa. During DNA replication, the RF-C complex recognizes primed DNA at the 3′-OH of the pre-Okazaki fragment and recruits trimeric PCNA molecules onto DNA in the presence of ATP. Subsequent ATP hydrolysis catalyzed by RF-C is required for polymerase δ to join the complex and initiate chain elongation (51Stillman B. Cell. 1994; 78: 725-728Abstract Full Text PDF PubMed Scopus (223) Google Scholar). Therefore, HRad17 may interact with the RF-C complex and interfere with its replication activity, causing cell cycle arrest. In fact, we have found HRad17 and RFC140 in the same immunocomplex generated with Hrad17 antibody 31E9 and RFC140 monoclonal antibodies from Dr. Bruce Stillman, Cold Spring Harbor.2 In summary, we characterized the nuclear localization of the putative checkpoint protein Hrad17 as an overall dotted pattern with prominent staining in the nucleoli. The nucleolar portion of Hrad17 redistributed upon treatment with DNA damaging agents indicating a possible role of this organelle and of Hrad17 in the DNA damage and replication checkpoint pathways. We are grateful to Drs. Tso-Pang Yao, Kin-Ming Lo, and Edmond Cheng for valuable discussions and helpful comments on this article. We also thank Dr. Yuhui Xu for assistance with electron microscopy procedures. Anti-MPP10 antiserum was a gift from Dr. Westendorf." @default.
- W2071915265 created "2016-06-24" @default.
- W2071915265 creator A5007475082 @default.
- W2071915265 creator A5024212886 @default.
- W2071915265 creator A5025927931 @default.
- W2071915265 creator A5029799513 @default.
- W2071915265 creator A5035377886 @default.
- W2071915265 creator A5044201137 @default.
- W2071915265 creator A5046214153 @default.
- W2071915265 creator A5055000089 @default.
- W2071915265 creator A5061491770 @default.
- W2071915265 creator A5074397518 @default.
- W2071915265 creator A5081462168 @default.
- W2071915265 creator A5083550013 @default.
- W2071915265 date "1999-12-01" @default.
- W2071915265 modified "2023-10-16" @default.
- W2071915265 title "HRad17 Colocalizes with NHP2L1 in the Nucleolus and Redistributes after UV Irradiation" @default.
- W2071915265 cites W1498525243 @default.
- W2071915265 cites W1607596288 @default.
- W2071915265 cites W1961693204 @default.
- W2071915265 cites W1963578989 @default.
- W2071915265 cites W1967264277 @default.
- W2071915265 cites W1969308341 @default.
- W2071915265 cites W1974010399 @default.
- W2071915265 cites W1975414139 @default.
- W2071915265 cites W1979402156 @default.
- W2071915265 cites W1984430482 @default.
- W2071915265 cites W1988840026 @default.
- W2071915265 cites W1994909028 @default.
- W2071915265 cites W2004008931 @default.
- W2071915265 cites W2008164539 @default.
- W2071915265 cites W2011962432 @default.
- W2071915265 cites W2016236490 @default.
- W2071915265 cites W2030720512 @default.
- W2071915265 cites W2033126648 @default.
- W2071915265 cites W2033613599 @default.
- W2071915265 cites W2034306661 @default.
- W2071915265 cites W2034761258 @default.
- W2071915265 cites W2041586068 @default.
- W2071915265 cites W2042760624 @default.
- W2071915265 cites W2052598236 @default.
- W2071915265 cites W2054342610 @default.
- W2071915265 cites W2056777272 @default.
- W2071915265 cites W2058094663 @default.
- W2071915265 cites W2064316240 @default.
- W2071915265 cites W2066108933 @default.
- W2071915265 cites W2076314401 @default.
- W2071915265 cites W2078030859 @default.
- W2071915265 cites W2080408951 @default.
- W2071915265 cites W2081543364 @default.
- W2071915265 cites W2089367841 @default.
- W2071915265 cites W2101118374 @default.
- W2071915265 cites W2105592350 @default.
- W2071915265 cites W2112782276 @default.
- W2071915265 cites W2123005075 @default.
- W2071915265 cites W2123847436 @default.
- W2071915265 cites W2132623938 @default.
- W2071915265 cites W2136989853 @default.
- W2071915265 cites W2159168998 @default.
- W2071915265 cites W2160954038 @default.
- W2071915265 cites W2165189712 @default.
- W2071915265 cites W2166571354 @default.
- W2071915265 cites W2166742500 @default.
- W2071915265 cites W2178835627 @default.
- W2071915265 cites W2327471215 @default.
- W2071915265 cites W4251695548 @default.
- W2071915265 doi "https://doi.org/10.1074/jbc.274.51.36544" @default.
- W2071915265 hasPubMedId "https://pubmed.ncbi.nlm.nih.gov/10593953" @default.
- W2071915265 hasPublicationYear "1999" @default.
- W2071915265 type Work @default.
- W2071915265 sameAs 2071915265 @default.
- W2071915265 citedByCount "38" @default.
- W2071915265 countsByYear W20719152652012 @default.
- W2071915265 countsByYear W20719152652013 @default.
- W2071915265 countsByYear W20719152652014 @default.
- W2071915265 countsByYear W20719152652015 @default.
- W2071915265 countsByYear W20719152652016 @default.
- W2071915265 countsByYear W20719152652020 @default.
- W2071915265 countsByYear W20719152652022 @default.
- W2071915265 crossrefType "journal-article" @default.
- W2071915265 hasAuthorship W2071915265A5007475082 @default.
- W2071915265 hasAuthorship W2071915265A5024212886 @default.
- W2071915265 hasAuthorship W2071915265A5025927931 @default.
- W2071915265 hasAuthorship W2071915265A5029799513 @default.
- W2071915265 hasAuthorship W2071915265A5035377886 @default.
- W2071915265 hasAuthorship W2071915265A5044201137 @default.
- W2071915265 hasAuthorship W2071915265A5046214153 @default.
- W2071915265 hasAuthorship W2071915265A5055000089 @default.
- W2071915265 hasAuthorship W2071915265A5061491770 @default.
- W2071915265 hasAuthorship W2071915265A5074397518 @default.
- W2071915265 hasAuthorship W2071915265A5081462168 @default.
- W2071915265 hasAuthorship W2071915265A5083550013 @default.
- W2071915265 hasBestOaLocation W20719152651 @default.
- W2071915265 hasConcept C111337013 @default.
- W2071915265 hasConcept C121332964 @default.
- W2071915265 hasConcept C183873130 @default.
- W2071915265 hasConcept C185544564 @default.
- W2071915265 hasConcept C185592680 @default.