13
Physical and Functional Mapping of the Replication Protein A Interaction Domain of the Werner and Bloom Syndrome Helicases* S Received for publication, January 18, 2005, and in revised form, June 10, 2005 Published, JBC Papers in Press, June 17, 2005, DOI 10.1074/jbc.M500653200 Kevin M. Doherty‡, Joshua A. Sommers‡, Matthew D. Gray§, Jae Wan Lee‡, Cayetano von Kobbe‡, Nicolas H. Thoma, Raichal P. Kureekattil**, Mark K. Kenny**, and Robert M. Brosh, Jr.‡ ‡‡ From the Laboratory of Molecular Gerontology, NIA, National Institutes of Health, Baltimore, Maryland 21224, the §MDG Associates, Seattle, Washington 98126, the Structural Biology Program, Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, and the **Department of Emergency Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York 10467 The single-stranded DNA-binding protein replication protein A (RPA) interacts with several human RecQ DNA helicases that have important roles in maintaining genomic stability; however, the mechanism for RPA stimulation of DNA unwinding is not well understood. To map regions of Werner syndrome helicase (WRN) that interact with RPA, yeast two-hybrid studies, WRN affinity pull-down experiments and enzyme-linked im- munosorbent assays with purified recombinant WRN protein fragments were performed. The results indi- cated that WRN has two RPA binding sites, a high affin- ity N-terminal site, and a lower affinity C-terminal site. Based on results from mapping studies, we sought to determine if the WRN N-terminal region harboring the high affinity RPA interaction site was important for RPA stimulation of WRN helicase activity. To accom- plish this, we tested a catalytically active WRN helicase domain fragment (WRN H-R ) that lacked the N-terminal RPA interaction site for its ability to unwind long DNA duplex substrates, which the wild-type enzyme can effi- ciently unwind only in the presence of RPA. WRN H-R helicase activity was significantly reduced on RPA- dependent partial duplex substrates compared with full-length WRN despite the presence of RPA. These re- sults clearly demonstrate that, although WRN H-R had comparable helicase activity to full-length WRN on short duplex substrates, its ability to unwind RPA- dependent WRN helicase substrates was significantly impaired. Similarly, a Bloom syndrome helicase (BLM) domain fragment, BLM 642–1290 , that lacked its N-termi- nal RPA interaction site also unwound short DNA du- plex substrates similar to wild-type BLM, but was se- verely compromised in its ability to unwind long DNA substrates that full-length BLM helicase could unwind in the presence of RPA. These results suggest that the physical interaction between RPA and WRN or BLM helicases plays an important role in the mechanism for RPA stimulation of helicase-catalyzed DNA unwinding. Within the last decade, several genetic disorders with pre- mature aging and/or cancer have been identified in which a gene member of the RecQ helicase family is mutated (1, 2). RecQ helicases share a centrally located domain of 450 residues that contains the seven conserved helicase motifs (for review, see Ref. 3). The founding member of the RecQ family, Escherichia coli RecQ helicase, has been extensively studied biochemically and has been genetically implicated in DNA recombination. A single yeast RecQ helicase, Sgs1 or Rqh1, is found in the budding yeast Saccharomyces cerevisiae and fission yeast Schizosaccharomyces pombe, respectively, and these helicases are thought to be important in the cellu- lar response to DNA-damaging agents and maintenance of genome stability. RecQ helicases have also been identified in a number of higher eukaryotes, including Xenopus laevis (focus forming activity 1 (FFA-1) 1 ), Drosophila melanogaster (DmBLM and DmRecQ5), and Caenorhabditis elegans (WRN-1, Ce-RCQ5, HIM-6, and RECQL/Q1). These helicases have proposed functions in DNA replication or repair; how- ever, the precise details of their roles in cellular pathways of DNA metabolism are still under investigation. There are five human RecQ helicases: 1) WRN, defective in Werner syndrome (WS); 2) BLM, defective in Bloom syndrome (BS); 3) RECQL4, defective in Rothmund-Thomson syndrome and RAPADILINO; 4) RECQ1; and 5) RECQ5. The human RecQ helicase disorders have distinctly different clinical phe- notypes; however, WS, BS, and Rothmund-Thomson syndrome are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu- man diseases have not yet been genetically linked to mutations in RECQ1 or RECQ5. The prominent roles of RecQ helicases in the maintenance of genome stability suggest that RECQ1 and RECQ5 helicases are also likely to be important in vivo. In addition to the conserved helicase motifs, the majority of RecQ helicases have a second conserved region of 80 amino acids located C-terminal to the helicase domain designated RecQ-Ct (RQC) (4). Although the conserved helicase domain is responsible for coupling nucleotide hydrolysis to DNA unwind- ing, recent studies suggest that the RQC motif plays an impor- tant role in protein interactions (5), nucleolar localization (6), and/or DNA binding (7). The WRN protein is unique among the * The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. S The on-line version of this article (available at http://www.jbc.org) contains supplemental Figs. S1–S4. Current address: Centro Nacional de Investigaciones Oncologicas (CNIO), Telomeres and Telomerase Group, Madrid 28029, Spain. ‡‡ To whom correspondence should be addressed: Laboratory of Mo- lecular Gerontology, NIA, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224. Tel.: 410-558-8578; Fax: 410-558- 8157; E-mail: [email protected]. 1 The abbreviations used are: FFA-1, focus forming activity 1; WRN, Werner syndrome helicase; BLM, Bloom syndrome helicase; WS, Werner syndrome; BS, Bloom syndrome; RQC, helicase domain desig- nated RecQ-Ct; HRD, helicase-related domain; RPA, replication protein A; 3-AT, 3-amino-1,2,4-triazole; GST, glutathione S-transferase; MBP, maltose-binding protein; ELISA, enzyme-linked immunosorbent assay; ssDNA, single-stranded DNA; BSA, bovine serum albumin. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 280, No. 33, Issue of August 19, pp. 29494 –29505, 2005 © 2005 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. This paper is available on line at http://www.jbc.org 29494 by guest on July 23, 2020 http://www.jbc.org/ Downloaded from

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

Physical and Functional Mapping of the Replication Protein AInteraction Domain of the Werner and Bloom Syndrome Helicases*□S

Received for publication, January 18, 2005, and in revised form, June 10, 2005Published, JBC Papers in Press, June 17, 2005, DOI 10.1074/jbc.M500653200

Kevin M. Doherty‡, Joshua A. Sommers‡, Matthew D. Gray§, Jae Wan Lee‡,Cayetano von Kobbe‡¶, Nicolas H. Thoma�, Raichal P. Kureekattil**, Mark K. Kenny**,and Robert M. Brosh, Jr.‡ ‡‡

From the ‡Laboratory of Molecular Gerontology, NIA, National Institutes of Health, Baltimore, Maryland 21224, the§MDG Associates, Seattle, Washington 98126, the � Structural Biology Program, Howard Hughes Medical Institute,Memorial Sloan-Kettering Cancer Center, New York, New York 10021, and the **Department of Emergency Medicine,Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York 10467

The single-stranded DNA-binding protein replicationprotein A (RPA) interacts with several human RecQDNA helicases that have important roles in maintaininggenomic stability; however, the mechanism for RPAstimulation of DNA unwinding is not well understood.To map regions of Werner syndrome helicase (WRN)that interact with RPA, yeast two-hybrid studies, WRNaffinity pull-down experiments and enzyme-linked im-munosorbent assays with purified recombinant WRNprotein fragments were performed. The results indi-cated that WRN has two RPA binding sites, a high affin-ity N-terminal site, and a lower affinity C-terminal site.Based on results from mapping studies, we sought todetermine if the WRN N-terminal region harboring thehigh affinity RPA interaction site was important forRPA stimulation of WRN helicase activity. To accom-plish this, we tested a catalytically active WRN helicasedomain fragment (WRNH-R) that lacked the N-terminalRPA interaction site for its ability to unwind long DNAduplex substrates, which the wild-type enzyme can effi-ciently unwind only in the presence of RPA. WRNH-Rhelicase activity was significantly reduced on RPA-dependent partial duplex substrates compared withfull-length WRN despite the presence of RPA. These re-sults clearly demonstrate that, although WRNH-R hadcomparable helicase activity to full-length WRN onshort duplex substrates, its ability to unwind RPA-dependent WRN helicase substrates was significantlyimpaired. Similarly, a Bloom syndrome helicase (BLM)domain fragment, BLM642–1290, that lacked its N-termi-nal RPA interaction site also unwound short DNA du-plex substrates similar to wild-type BLM, but was se-verely compromised in its ability to unwind long DNAsubstrates that full-length BLM helicase could unwindin the presence of RPA. These results suggest that thephysical interaction between RPA and WRN or BLMhelicases plays an important role in the mechanism forRPA stimulation of helicase-catalyzed DNA unwinding.

Within the last decade, several genetic disorders with pre-mature aging and/or cancer have been identified in which agene member of the RecQ helicase family is mutated (1, 2).RecQ helicases share a centrally located domain of �450residues that contains the seven conserved helicase motifs(for review, see Ref. 3). The founding member of the RecQfamily, Escherichia coli RecQ helicase, has been extensivelystudied biochemically and has been genetically implicated inDNA recombination. A single yeast RecQ helicase, Sgs1 orRqh1, is found in the budding yeast Saccharomyces cerevisiaeand fission yeast Schizosaccharomyces pombe, respectively,and these helicases are thought to be important in the cellu-lar response to DNA-damaging agents and maintenance ofgenome stability. RecQ helicases have also been identified ina number of higher eukaryotes, including Xenopus laevis(focus forming activity 1 (FFA-1)1), Drosophila melanogaster(DmBLM and DmRecQ5), and Caenorhabditis elegans(WRN-1, Ce-RCQ5, HIM-6, and RECQL/Q1). These helicaseshave proposed functions in DNA replication or repair; how-ever, the precise details of their roles in cellular pathways ofDNA metabolism are still under investigation.

There are five human RecQ helicases: 1) WRN, defective inWerner syndrome (WS); 2) BLM, defective in Bloom syndrome(BS); 3) RECQL4, defective in Rothmund-Thomson syndromeand RAPADILINO; 4) RECQ1; and 5) RECQ5. The humanRecQ helicase disorders have distinctly different clinical phe-notypes; however, WS, BS, and Rothmund-Thomson syndromeare all characterized by genomic instability, an elevated cancerincidence, and/or particular aspects of premature aging. Hu-man diseases have not yet been genetically linked to mutationsin RECQ1 or RECQ5. The prominent roles of RecQ helicases inthe maintenance of genome stability suggest that RECQ1 andRECQ5 helicases are also likely to be important in vivo.

In addition to the conserved helicase motifs, the majority ofRecQ helicases have a second conserved region of �80 aminoacids located C-terminal to the helicase domain designatedRecQ-Ct (RQC) (4). Although the conserved helicase domain isresponsible for coupling nucleotide hydrolysis to DNA unwind-ing, recent studies suggest that the RQC motif plays an impor-tant role in protein interactions (5), nucleolar localization (6),and/or DNA binding (7). The WRN protein is unique among the

* The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby marked“advertisement” in accordance with 18 U.S.C. Section 1734 solely toindicate this fact.

□S The on-line version of this article (available at http://www.jbc.org)contains supplemental Figs. S1–S4.

¶ Current address: Centro Nacional de Investigaciones Oncologicas(CNIO), Telomeres and Telomerase Group, Madrid 28029, Spain.

‡‡ To whom correspondence should be addressed: Laboratory of Mo-lecular Gerontology, NIA, National Institutes of Health, 5600 NathanShock Drive, Baltimore, MD 21224. Tel.: 410-558-8578; Fax: 410-558-8157; E-mail: [email protected].

1 The abbreviations used are: FFA-1, focus forming activity 1; WRN,Werner syndrome helicase; BLM, Bloom syndrome helicase; WS,Werner syndrome; BS, Bloom syndrome; RQC, helicase domain desig-nated RecQ-Ct; HRD, helicase-related domain; RPA, replication proteinA; 3-AT, 3-amino-1,2,4-triazole; GST, glutathione S-transferase; MBP,maltose-binding protein; ELISA, enzyme-linked immunosorbent assay;ssDNA, single-stranded DNA; BSA, bovine serum albumin.

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 280, No. 33, Issue of August 19, pp. 29494–29505, 2005© 2005 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

This paper is available on line at http://www.jbc.org29494

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 2: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

human RecQ helicases in that it also contains a region ofhomology to the 3� to 5� exonuclease domain of proofreadingenzymes (8). Interestingly, the WRN protein has a direct repeatof a highly acidic 27-amino acid sequence located in the Nterminus between the exonuclease and helicase domains (8).Although the exact functional significance of this repeatedsequence is not known, and only one copy of the sequence isfound in Xenopus FFA-1, this acidic repeat has been implicatedin studies of the human WRN protein to play a role in tran-scriptional activation (9). A C-terminal conserved motif desig-nated helicase-related domain (HRD), found in a number ofDNA-metabolizing proteins, is present in many RecQ helicases(4), and recent evidence suggests that the domain is importantin DNA binding (7).

Biochemical analyses of the purified recombinant WRN pro-tein from a number of laboratories, including ours, have char-acterized the catalytic activities and protein interactions ofWRN (for review, see Refs. 5 and 10). WRN protein has beenshown to unwind relatively short DNA duplexes of 20–30 bp inthe absence of any auxiliary factors with a 3� to 5� polarity(11–13). However, WRN is a relatively poor helicase on longerDNA duplexes such as a 34-bp forked duplex (14) or 69-bp M13partial duplex DNA substrate (15), suggesting that the enzymehas limited processivity in the unwinding reaction. WRN heli-case can catalyze unwinding of longer DNA duplexes up to 851bp in a reaction dependent on replication protein A (RPA) (15),a single-stranded DNA-binding protein that is implicated inthe processes of DNA replication, recombination and repair,and transcription (16). Consistent with the functional interac-tion between WRN and RPA, the two proteins physically inter-act (15, 17); however, the mechanism for stimulation of WRN-catalyzed DNA unwinding is not well understood.

To address the mechanism for RPA stimulation of WRNhelicase activity, we have begun to evaluate the importance ofthe physical interaction between WRN and RPA for the stim-ulation of helicase activity. In this work, we have mapped theRPA binding sites on WRN protein using multiple approachesand identified a high affinity RPA interaction site in the Nterminus. Biochemical analyses of a purified recombinantWRN protein fragment, WRNH-R, that contains the helicasecore domain and RQC motif, but lacks the N-terminal regionand C-terminal regions of WRN, retains its ability to unwindshort (28 bp) M13 partial duplexes nearly as efficiently asfull-length WRN helicase; however, WRNH-R is severely com-promised in its ability to unwind longer (50–850 bp) DNAsubstrates that the full-length WRN protein can efficientlyunwind in the presence of RPA. These results demonstrate thatWRN helicase binds tightly to RPA via its N-terminal domainand suggest that this physical protein interaction with RPA islikely to be important in the mechanism of RPA-dependentstimulation of WRN helicase activity.

In addition to WRN, human BLM (18), RECQ1 (19), andRECQ5� (20) helicases also physically and functionally interactwith RPA, but the details of how RPA stimulates the unwindingactivities of these DNA helicases are also largely unknown. Sim-ilar to our findings with WRN, we report here that BLM helicasecontains a high affinity N-terminal RPA interaction site. A BLMrecombinant fragment lacking this RPA interaction site, but con-taining the RQC and HRD motifs, was shown to be defective inunwinding long DNA duplex substrates despite the presence ofRPA. Our studies indicate that both WRN and BLM helicasescontain an N-terminal RPA binding domain that is important forthe stimulation of helicase activity by RPA. The important cellu-lar roles of RPA in DNA metabolism are likely to involve specificinteractions with human RecQ helicases in pathways that main-tain genome stability.

MATERIALS AND METHODS

Yeast Two-hybrid Reporter Assay—A two-hybrid screen was per-formed as previously described (9). Briefly, L40 S. cerevisiae reporterstrain, pBTM116 (LexA DNA-binding domain) and pVP16 (herpes sim-plex virus VP16 transcriptional activation domain) fusion vectors werekindly provided by Dr. Stanley Hollenberg. Yeast media were pur-chased from BIO 101 (La Jolla, CA), and 3-amino-1,2,4-triazole (3-AT)was from Sigma. LexA-WRN fusion protein constructs were createdusing the expression vector pBTM116 (21). This plasmid carries theTRP1 gene for selection in yeast and a multiple cloning site insertedafter the 202-amino acid open reading frame of the E. coli lexA gene,which encodes an SOS function regulatory protein and is driven by aconstitutive yeast ADH promoter in this vector. In-frame fusions be-tween the C terminus of the LexA protein and specified domains of thehuman WRN protein were created by insertion of PCR-generated WRNcDNA domains into the multiple cloning sites of pBTM116 as previouslydescribed (6). Briefly, the full-length WRN cDNA fusion, constructWRN�, was created by insertion of the unique BamHI-SspI WRN cDNAfragment into the BamHI and SalI (filled in) sites of WRN domainwithin pBTM116. To create WRN�1/2, a single copy of the acidic 27-amino acid repeat was removed from construct WRN� by deletion of theunique AflII-AflII fragment of the WRN cDNA, which occurs within therepeated sequence. To create the full deletion of the repeated sequence,WRN�R, a PCR fragment was generated from WRN cDNA using prim-ers TH1 (5�-GCC AGA TCT TGG AAA CAA CTG CAC-3�) and TH�R(5�-CGG CTT AAG CTC AGT AGA TTT A-3�), which was digested withBamHI and AflII and then substituted for the BamHI-AflII WRN frag-ment of construct WRN�1/2. This PCR product results in the addition ofa novel AflII site located just before the acidic repeat sequence, whichbecomes fused upon ligation to the distal AflII site within the endoge-nous WRN message. To create the WRN�, WRN�1/2, and WRN�R fusionprotein constructs, regions of the wild-type or deleted repeat domainswere amplified directly from the corresponding full-length constructs aspreviously described (9). These PCR products were then inserted intothe pBTM116 vector at the chosen restriction sites. VP16-WRN fusionprotein constructs were created using the expression vector pVP16 (21).This plasmid carries the LEU2 gene for selection in yeast and a multi-ple cloning site inserted after the herpes simplex virus VP16 transcrip-tional activation domain. In-frame fusions between the C terminus ofthe VP16 protein, and specified domains of the human WRN proteinwere created by insertion of PCR-generated WRN cDNA domains intothe multiple cloning sites of pVP16. RPA fusion protein constructs werecreated by PCR amplification of individual full-length RPA proteinsubunits (cDNAs were kindly provided by Dr. Marc Wold) using primerscontaining restriction sites compatible with pBTM116 (for LexA fu-sions) or pVP16 (for VP16 fusions) multiple cloning sites. For C-termi-nal truncated versions of the RPA70 subunit, stop codons were intro-duced within PCR reverse primers directly after the specified aminoacid codon or, in the case of the N-terminal truncation, the forwardprimer initiated at the indicated internal amino acid position within thecDNA sequence. All fusion protein constructs were confirmed by man-ual sequencing to be in-frame and devoid of mutations.

For two-hybrid assays, S. cerevisiae LexA reporter strain L40(MATa his3�200 trp1–901 leu2–3112 ade2 LYS2::(lexAop)4-HIS3URA3::(lexAop)8-LacZ GAL4 gal80) was co-transformed with the in-dicated pairs of LexA- and VP16 fusion protein constructs or controlvectors. To assess the relative reporter activities of each pair, positiveco-transformants were selected on yeast media plates lacking trypto-phan and leucine, and multiple colonies of each pairing were re-streaked to templates for even growth (2–3 days on the same media).Co-transformant templates were then replica-plated onto yeast me-dia plates lacking tryptophan, leucine, and histidine and containing0–500 mM 3-AT. Replica plating was performed in reverse order(i.e. from high 3-AT concentration to low) to ensure efficient transferthroughout the series. Relative growth on 3-AT-containing plates wasassessed after 10 days of incubation at 30 °C. All reporter assayswere repeated in at least three different experiments. Reporterstrength was essentially invariable within multiple co-transformants(original colonies isolated) of each construct pair in all 3-AT growthexperiments.

Proteins—Recombinant hexahistidine-tagged WRN protein wasoverexpressed using a baculovirus/Sf9 insect cell system and purified asdescribed previously (22). Glutathione S-Transferase (GST) fusion pro-teins of WRN were subcloned into the bacterial expression plasmidpGEXCS (Amersham Biosciences) as described elsewhere (23). In-frame fusions of GST-2R (WRN residues 406–525) or GST-0R (WRNresidues 406–525 with intervening residues 424–475 deleted) were

Mapping of RPA Interaction Domain on WRN and BLM Helicases 29495

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 3: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

created by inserting the PCR products, generated in the same way asfor the two-hybrid system, into the BamHI site within the multiplecloning site of the pGEXCS vector. GST-WRN fragment fusion proteinswere expressed and purified as described previously (23). A recombi-nant WRN protein fragment (exact boundaries to be described else-where),2 designated WRNH-R, was overexpressed using a baculovirusinsect cell system and purified using immobilized glutathione beads.WRNH-R contains the entire helicase domain (motifs I–VI) and theconserved RQC region but lacks the N-terminal sequence, including theacidic repeats as well as the C-terminal region after the RQC motif(Fig. 5A).

Hexahistidine-tagged recombinant BLM protein, was overexpressedin S. cerevisiae and purified as previously described (24). Expressionconstructs for maltose-binding protein (MBP) fusion proteins of BLMfragments (25) and the BLM642–1290 protein fragment were kindly pro-vided by Dr. Ian Hickson (Cancer Research UK Laboratories). MBP-BLM fusion proteins were purified as previously described (25). Briefly,MBP-BLM fusion proteins were expressed in BL21(DE3) cells (NewEngland Biolabs) that had been transformed with the pMAL-C2 expres-sion plasmids containing various portions of the BLM cDNA (MBP-BLM1–447 and MBP-BLM966–1417). BL21(DE3) cells transformed withplasmid encoding MBP were used for control experiments. Overnighttransformed bacterial cultures were used to inoculate 1 liter of LuriaBertani broth supplemented with 2% glucose and 100 �g/ml ampicillinat a 1:100 dilution, and the cultures were grown at 37 °C to an A600 of�0.5. Isopropyl-1-thio-�-D-galactopyranoside was then added to a finalconcentration of 0.4 mM, and the cultures were allowed to grow for anadditional 3 h before being chilled on ice for 30 min. After centrifugationat 10,000 rpm in a Beckman JA10 rotor, the cell pellet was resuspendedin 50 ml of column buffer (20 mM Tris-HCl, pH 7.4, 200 mM NaCl, 1 mM

EDTA, 10 mM �-mercaptoethanol, 1 mM dithiothreitol) supplementedwith complete protease inhibitor mixture (Roche Applied Science) at themanufacturer’s recommended concentration. Cells were lysed by soni-cation, and the lysate was clarified by centrifugation at 9,000 � g in aBeckman Ti-70 rotor for 30 min at 4 °C. The crude extract was diluted1:5 with column buffer and then bound to amylose resin (New EnglandBiolabs) that had been pre-washed with column buffer. The resin waswashed with 12 volumes of column buffer before the recombinant pro-tein was eluted from the resin with column buffer supplemented with10 mM maltose. RPA containing all three subunits (RPA70, RPA32, andRPA14) was purified as previously described (26).

GST-WRN-Sepharose Pull-down Experiments—GST-WRN pull-down experiments were performed as previously described (23). Briefly,GST-WRN fusion proteins were overexpressed in BL21(DE3)pLysS by 1mM isopropyl-1-thio-�-D-galactopyranoside induction for 8 h at 23 °C.The bacterial cell pellet was lysed by sonication in lysis buffer (phos-phate-buffered saline, 10% glycerol, 0.4% Triton X-100). The lysate wasclarified by centrifugation at 35,000 rpm (Ti-60 rotor; Beckman) for 1 hat 4 °C. One milliliter of the resulting supernatant was incubated with100 �l of GS beads (50% v/v) for 1 h at 4 °C. The beads were washedthree times with 1 ml of lysis buffer, and split into two aliquots, one forbinding experiments and one for determination of background signal inWestern blot analysis. For binding experiments, protein-bound beadswere incubated for 1 h at 4 °C with 1.2 �g of purified recombinant RPAin 250 �l of buffer D (50 mM HEPES, pH 7.1, 100 mM KCl, 10% glycerol).The beads were subsequently washed three times with 500 �l of bufferD and eluted by boiling treatment in 40 �l of Laemmli buffer. Proteinswere electrophoresed on 10% polyacrylamide SDS gels and transferredto polyvinylidene difluoride membranes. Control membranes werestained with Amido Black reagent to demonstrate equal loading ofprotein samples. Membranes were probed with mouse monoclonal anti-RPA antibody (Ab-1, 1:20, Oncogene Research Products), followed byhorse anti-mouse IgG-horseradish peroxidase (Vector) and visualizedusing ECL-Plus (Amersham Pharmacia).

ELISA Assays—The indicated purified recombinant WRN or BLMproteins (full-length or fragment) were diluted to a concentration of 18nM in carbonate buffer (0.016 M Na2CO3, 0.034 M NaHCO3, pH 9.6) andwere added to individual wells of a 96-well microtiter plate (50 �l/well),which was then incubated at 4 °C overnight. Bovine serum albumin(BSA) was used in the coating step for control reactions. After the initialbinding step, samples were aspirated and the wells were blocked for 2 hat 30 °C with blocking buffer (phosphate-buffered saline, 0.5% Tween20, and 3% BSA). After washing, RPA was diluted to 144 nM in blockingbuffer and was added to the appropriate wells of the ELISA plate (50�l/well), which was then incubated for 1 h at 30 °C. The samples were

aspirated, and the wells were washed five times before addition ofanti-RPA (Ab-1) mouse monoclonal antibody (Oncogene Research Prod-ucts) diluted 1:100 in blocking buffer, followed by incubation at 30 °Cfor 1 h. Following three washes, horseradish peroxidase-conjugatedanti-mouse secondary antibody (1:2500) diluted in blocking buffer wasadded to the wells, and the samples were incubated for 30 min at 30 °C.After washing five times, RPA bound to the proteins was detected usingO-phenylenediamine substrate (Sigma). The reaction was terminatedafter 5 min with 3 N H2SO4, and absorbance readings were taken at 490nm. Data analysis for determination of the apparent dissociation con-stant (Kd) was performed as previously described (18).

Helicase Substrate Preparation—M13mp18 single stranded circularDNA was from New England Biolabs. The oligonucleotides used for the28-, 50-, 69-, and 100-bp M13 partial duplex helicase substrates werepurchased from Midland Certified Reagents Co. (Midland, TX). The 28-,50-, 69-, 100-, and 849-bp M13mp18 partial duplex substrate wereconstructed using oligonucleotides complementary to positions 6296–6323, 6272–6322, 6254–6322, and 6039–6138, respectively, in theM13mp18 ssDNA circle. The 849-bp fragment, in which one strand wascomplementary to positions 1396–2244 of the M13mp18 ssDNA circlewas amplified by PCR with the primers 5�-CCTTTAACTCCCTG-CAAGCC-3� and 5�-AAACAAACACTTATAGTTCC-3�. The M13mp18partial duplex substrates were radioactively labeled and constructed aspreviously described (15). The 19-bp forked duplex substrate was ra-dioactively labeled and constructed as previously described (27).

Helicase Assays—Helicase assay reaction mixtures (20 �l) using theM13 partial duplex substrates contained 40 mM Tris (pH 7.4), 4 mM

MgCl2, 5 mM dithiothreitol, 2 mM ATP, 0.5 mg/ml tRNA, and theindicated concentration of WRN or BLM protein. For assays using the19-bp forked duplex substrate, reaction mixtures (20 �l) contained 30mM HEPES (pH 7.5), 40 mM KCl, 8 mM MgCl2, 100 ng/�l BSA, 5%glycerol, and DNA substrate concentrations indicated in the figurelegends. Reaction mixtures were initiated by the addition of WRN orBLM protein. Helicase reactions were incubated at 37 °C and wereterminated by the addition of 10 �l of 50 mM EDTA-40% glycerol-0.9%SDS-0.1% bromphenol blue-0.1% xylene cyanol at time points indicatedin the figure legends for M13 partial duplex substrates or 37 °C for 15min using the 19-bp forked duplex substrate. The products of helicasereactions were resolved on nondenaturing 10% or 12% polyacrylamidegels for the reactions with M13 partial duplex (28, 50, 69, and 100 bp)or oligonucleotide based forked duplex substrates, respectively. Heli-case reaction products using the 849-bp M13 partial duplex substratewere resolved on nondenaturing 8% polyacrylamide gels. RadiolabeledDNA species in polyacrylamide gels were visualized using a Phosphor-Imager and quantitated using the ImageQuaNT software (MolecularDynamics). The percent helicase substrate unwound, defined as thepercentage of the labeled oligonucleotide released from the intact du-plex substrate, was calculated as previously described (27).

RESULTS

We have utilized both in vivo and in vitro assays to map thedomain of the WRN protein important for binding to RPA andto further characterize the physical and functional interactionsbetween the single-stranded DNA-binding protein and the he-licase. Our results have been interpreted to provide a betterunderstanding of how the protein interaction between WRNand RPA might be important in the mechanism for RPA stim-ulation of WRN helicase activity.

Mapping of Interaction Sites between WRN and RPA by aTwo-hybrid Screen—To investigate the protein domains impor-tant for the interaction of WRN with RPA, a two-hybrid re-porter assay was employed. Various regions of the WRN pro-tein coding sequence (Fig. 1) were inserted into the DNAbinding domain plasmid (pBTM116), which was then trans-formed into the yeast strain L40, which contains a transcrip-tional reporter for the expression of HIS3, and the fragmentswere expressed as LexA-WRN fusion proteins. The originalcharacterization of these LexA-WRN fusions revealed that thedirectly repeated sequence of 27 amino acids, located betweenthe exonuclease and helicase domains of WRN, is capable ofstrongly activating transcription itself when present in anyLexA-WRN fusion protein (9) (data not shown). LexA-WRNfusion protein constructs containing this highly acidic sequencecould not, therefore, be used in this study. The three subunits2 Nicolas H. Thoma, submitted for publication.

Mapping of RPA Interaction Domain on WRN and BLM Helicases29496

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 4: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

of RPA (RPA70, RPA32, and RPA14) were fused to the herpessimplex virus transcriptional activation protein VP16, in theplasmid pVP16. In addition, two N-terminal fragments ofRPA70, designated RPA701–168 and RPA701–308, and a C-ter-minal fragment of RPA70, designated RPA70308–616, werefused to VP16. The yeast strain L40 was then used to screen forinteraction sites between the expressed WRN and RPA fusionproteins in vivo. Every combination of constructs was examinedby individual co-transfections (crosses) of construct pairs, andRPA32 was used in both orientations as a control for intra-RPAsubunit interactions. Co-transformed yeast cells were firstplated on a media lacking tryptophan and leucine to select forcolonies containing both LexA and VP16 fusion protein con-structs. Colonies were then restreaked and replica-plated to aseries of plates containing media lacking tryptophan, leucine,and histidine. 3-AT, an inhibitor of histidine biosynthesis, wasused at various concentrations to titrate the strength of thehistidine reporter (data not shown).

The results of the initial crosses between the WRN and RPAfusion protein construct pairs revealed that there were nosignificant interactions observed in crosses between any of theRPA subunits and the WRN exonuclease, helicase, or C-termi-nal domains examined (data not shown). The RPA32 subunitshowed strong interaction with the smaller RPA14 subunit andmodest, but significant, interaction with both constructs con-taining the C-terminal region of RPA70. The intra-RPA sub-unit interactions we observed were exactly as previously re-ported (28) and, hence, confirm the integrity of these constructsin the absence of any WRN interactions.

To further investigate the physical interaction betweenWRN and RPA, the same RPA domains were made as LexAfusion proteins and VP16-WRN fusions constructs were madecontaining the full-length protein both with a single copy of thedirectly repeated sequence of 27 amino acids and the wild-typeversion containing the full repeat (VP�WRN�1/2 and VP�WRN�,respectively, Fig. 1). VP16�WRN fusions were also made corre-sponding to a small fragment containing one-half of the re-peated acidic sequence (VP�WR�1/2), the full repeat (VP�WR�),and the flanking regions containing none of the repeated acidicsequence (VP�WR�). The results of crosses between these WRN

and RPA fusion protein construct pairs are shown in Table Iand summarized in Fig. 2.

These two-hybrid studies revealed a strong interaction be-tween the N-terminal half of RPA70 (RPA701–308) and all WRNconstructs containing the acidic region (Table I). Those WRNproteins containing the full acidic repeated sequence(VP�WRN�, VP�WR�), revealed the greatest ability to transacti-vate the reporter gene when co-expressed with the RPA701–308

fusion protein. The reporter strength observed was decreased by�33% when full-length WRN containing one-half of the repeatedsequence was expressed with RPA701–308. Similarly, the reporterstrength observed was reduced by �29% when the WRNfragment containing only one-half of the repeated sequence(VP�WR�1/2) was co-expressed with RPA701–308. Because allthese fragments contain short sequences that flank the acidicdirect repeat (residues 406–423 and 478–525), the flanking se-quence (VP�WR�) was also tested for interaction with the indi-vidual RPA subunits and the fragments of the RPA70 subunit.No significant interactions were observed with this small(65 residues) fragment for any of the protein pairs examined(Table I). These results demonstrate that WRN, and more spe-cifically the acidic repeat of WRN, is associated with the RPA70subunit in vivo and suggest that the WRN-RPA interaction isoptimized when both of the repeated 27 amino acids were presentin either the full-length WRN protein or the short WRN fragmentharboring the repeat itself and the flanking sequences.

Two-hybrid screens using two additional regions of RPA70(RPA701–168 and RPA70308–616) did not reveal an interactionwith any of the WRN regions tested; however, the C-terminalhalf of RPA70 (RPA70308–616) did interact with RPA32(Table I), as also observed in the opposite orientation in thisstudy (data not shown), and as expected from previous re-ports (17, 29). These results indicate a requirement of theregion between residues 168–308 in the RPA 70-kDa subunitfor the interaction with WRN.

The full-length RPA70 subunit did not interact with any ofthe WRN protein fragments. This may be due to the poorsolubility or improper folding of the RPA70 subunit expressedin the absence of RPA32 and RPA14 (30–32). It was demon-

FIG. 1. RPA and WRN two-hybridfusion protein constructs. Various re-gions of interest of the three RPA sub-units and WRN were fused to either theDNA binding domain of LexA or the her-pes simplex virus VP16 transcriptionalactivation domain.

Mapping of RPA Interaction Domain on WRN and BLM Helicases 29497

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 5: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

strated that expression of full-length RPA70 or RPA701–507

resulted in poor solubility of either the full-length or truncatedRPA70 subunit, whereas most of the expressed RPA701–442

was present in the soluble fraction (32). The authors suggestedthat in the absence of RPA32 and RPA14, full-length RPA70 hasa different conformation than the more soluble RPA70 C-terminal deletion mutants, including RPA701–442. These resultssuggest an explanation for why RPA701–308, but not full-lengthRPA70, interacted strongly with full-length WRN as demon-strated by our yeast two-hybrid studies. Recently, the WRNinteraction domain was mapped to residues 100–300 of RPA70(17), a result consistent with our findings. In the work from theLoeb laboratory, a direct interaction between WRN and the pu-rified soluble recombinant RPA701–442 (lacking the C-terminal174 amino acids) was reported, enabling the authors to concludethat RPA stimulates WRN helicase activity by its interactionwith RPA70 (17). In this study, we have focused our efforts ondetermining the RPA interaction site on WRN protein.

No interactions were detected whatsoever between any RPAsubunit and full-length WRN without the direct repeat(WRN�R) or WRN fragments harboring the helicase domainand/or the entire C-terminal sequence (WRN522–1432 andWRN861–1432) or a portion of the WRN C-terminal sequence

(1162–1432, WRN1162–1432, data not shown). Neither theRPA14 nor RPA32 subunits interacted with any region of WRNprotein, but they did show a very strong interaction with eachother (Table I), as expected from previous reports (28, 29).

FIG. 3. WRN direct repeats mediate a protein interaction withRPA as demonstrated by WRN affinity pull-down experiments.Recombinant GST-0R (lanes 3 and 4) or GST-2R (lanes 5 and 6) WRN-fragment fusion proteins overexpressed in E. coli were bound to gluta-thione beads and incubated with DNase I (2 �g/ml) in the presence(lanes 3 and 5) or absence (lanes 4 and 6) of RPA heterotrimer (1.2 �g),as described under “Materials and Methods.” After washes, bound pro-tein was eluted and resolved on 10% polyacrylamide SDS gels. Thepresence of RPA was detected by Western blot analyses using an anti-body against RPA70 (Oncogene Research Products). Control for equalprotein loading is indicated by Amido Black staining of the membrane.RPA heterotrimer (lane 1, 6000 ng; lane 6, 100 ng) was loaded toindicate purity by Amido Black staining of the membrane and as acontrol for the immunoblot, respectively. For immunoblotting analysis,lane 1 was removed.

TABLE IDetermination of regions of WRN and RPA required for interaction by two-hybrid reporter assay

Additional regions of the WRN protein and RPA subunits were used in a two-hybrid screen to map the sites required for WRN-RPA interactions.Numbers indicate the highest concentration of 3-AT (in media lacking histidine) at which yeast cells containing each particular combination ofconstructs shown could grow.

FIG. 2. Summary of WRN-RPA mapping results from the yeasttwo-hybrid studies. All WRN constructs containing the directly re-peated sequence were found to interact with the N-terminal region ofthe RPA70 truncated protein.

Mapping of RPA Interaction Domain on WRN and BLM Helicases29498

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 6: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

WRN Affinity Pull-down Experiments to Map the RPA Inter-action Domain—The yeast two-hybrid data suggest that theacidic direct repeat domain of WRN may mediate the physicalinteraction between WRN and the RPA 70-kDa subunit. Toverify the yeast two-hybrid data and further map the physicalinteraction site, we performed WRN affinity pull-down exper-iments. Various WRN-GST fusion proteins were expressed andbound to glutathione beads and were subsequently used inpull-down assays with purified RPA heterotrimer, shown byAmido Black staining of the RPA70, RPA32, and RPA14 sub-units immobilized on the membrane (Fig. 3). The GST-2R fu-sion contains the region of WRN406–525, which contains entireacidic direct repeat and the flanking sequences present. TheGST-0R is a fusion of GST to the region of WRN406–525 flankingthe direct repeat sequences but without the direct repeat in themiddle (residues 424–475). After a binding incubation in thepresence of DNase I (2 �g/ml) to eliminate the potential forindirect interactions mediated through co-binding to DNA, theprotein samples were resolved by SDS-PAGE and transferredto membranes for Amido Black staining to verify equal proteinloading followed by Western blot analyses to probe for boundRPA. As shown in Fig. 3, approximately equal amounts ofGST-0R and GST-2R were used in the affinity pull-down ex-periments. RPA, as detected with an antibody against theRPA70 subunit, was identified in the eluted samples from theGST-2R pull-down experiment (Fig. 3, lane 5). Little to no RPAwas detected when the pull-down was performed using theGST-0R-bound glutathione beads (Fig. 4, lane 3) or when RPAwas omitted from the binding incubation (Fig. 3, lane 4). Sim-ilar results were obtained in the presence of ethidium bromide(10 �g/ml), indicating that the interaction between RPA andGST-2R was not DNA mediated (data not shown).

The results from the yeast two-hybrid studies and theWRN affinity pull-down experiments suggest that the directrepeat elements acting alone can mediate an interaction withRPA70; however, we wanted to ascertain whether the inter-action would be retained by a WRN protein fragment thatcontained the direct repeat but in the context of a signifi-cantly greater length of flanking sequence. To address this,we performed RPA binding experiments using glutathionebeads with GST-WRN239–499 (Fig. 4A) bound as the affinityreagent. As shown in Fig. 4B, �25% of the RPA input wasco-precipitated with GST-WRN239–499 (lane 4) by comparisonwith the input (lanes 1 or 8). GST was unable to co-precipitateRPA (lane 2). The results from the WRN affinity pull-downexperiments support the findings from the yeast two-hybridstudies that the WRN direct repeat elements mediate a phys-ical interaction with the RPA70 subunit.

Although it was evident that the N-terminal region ofWRN, residues 239–499, mediated an interaction withRPA70, it was conceivable that other regions of WRN mightalso be associated with RPA70. To address this possibility,we examined the ability of other recombinant GST-WRNfragments bound to glutathione beads to pull-down RPA.Whereas glutathione beads coated with GST-WRN1–120

(Fig. 4B, lane 3) or GST-WRN1072–1432 (Fig. 4B, lane 7) boundRPA weakly, a significant fraction of the RPA70 inputwas bound not only by GST-WRN239–499, but also byGST-WRN949–1432 (lane 5) and GST-WRN949–1092 (lane 6),suggesting that the RQC region, which overlaps withWRN949–1092 (Fig. 4A) and mediates other WRN proteininteractions (2, 5), might also bind RPA70. In controlexperiments, pull-downs were performed in the presence ofDNase I (2 �g/ml) to confirm that the interaction between

FIG. 4. Mapping of the RPA bindingdomains on WRN. A, scheme of WRNfull-length and fragment proteins used forthese studies. B, a series of recombinantGST-WRN fragment fusion proteins over-expressed in E. coli were bound to gluta-thione beads and incubated with RPAheterotrimer (1.2 �g). After washes,bound protein was eluted and resolved on10% polyacrylamide SDS gels. The pres-ence of RPA was detected by Western blotanalyses using an antibody againstRPA70. Control for equal protein loadingis indicated by Amido Black staining ofthe membrane. RPA heterotrimer (58 or116 ng) was loaded for controls in lanes 1and 8, respectively.

Mapping of RPA Interaction Domain on WRN and BLM Helicases 29499

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 7: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

GST-WRN239–499, GST-WRN949–1092, or GST-WRN949–1432

and RPA was not mediated by DNA (Supplemental Fig. S1).Similar results were obtained in the presence of ethidiumbromide (10 �g/ml), also indicating that the interaction be-tween RPA and the WRN protein fragments was direct andnot DNA-mediated (data not shown).

Physical Analysis of the WRN-RPA Protein Interaction—Re-sults from the affinity pull-down experiments and two-hybridstudies suggested that RPA physically interacts with certaindiscrete domains of the WRN protein. ELISA studies wereperformed to explore the specificity and affinity of the WRN-RPA interaction. Increasing amounts of RPA (0–96 nM, hetero-trimer) were incubated in the presence of 3% BSA with WRN orthe specified WRN protein fragment (18 nM) that had beenpreviously immobilized on microtiter wells. Bound RPA proteinwas detected by a mouse monoclonal antibody against RPA70.As shown in Fig. 5, the colorimetric signal was both dose-de-pendent and saturable with full-length WRN protein. The spec-ificity of this interaction was demonstrated by the very lowabsorbance values (0.062 A490) for wells that had been pre-coated with BSA compared with the intense signal obtainedwith WRN. A purified recombinant GST-WRN239–499 fragment(Fig. 4A) that contained the N-terminal acidic repeated ele-ment also displayed a dose-dependent and saturable signal forinteraction with the bound RPA. Although the WRN239–499-RPA signal was �2-fold less than that obtained using full-length WRN, the plateau was approached at the same concen-tration of WRN239–499 and full-length WRN, 22 nM (Fig. 5). Thecolorimetric signal obtained from either WRN-RPA orWRN239–499-RPA interaction was resistant to the presence ofethidium bromide (10 �g/ml, data not shown), indicating that acontaminating DNA bridge was not responsible for the positivesignal. In comparison to WRN or WRN239–499, only a very weaksignal was detected when WRN1072–1432 was incubated withthe RPA-bound wells (Fig. 5). A detectable signal was observedfor the RPA interaction with a recombinant WRN protein frag-ment, WRNH-R, that contains the entire helicase domain (mo-tifs I–VI) and the conserved RQC region but lacks the N-terminal sequence, including the acidic repeats as well as theC-terminal region after the RQC motif; however, the A490 val-ues were significantly lower than that obtained for eitherWRN239–499 or full-length WRN and failed to reach a plateauat the highest RPA concentration tested, 96 nM (Fig. 5), sug-gesting that the WRNH-R-RPA interaction was of considerablylower affinity than that of WRN239–499-RPA or WRN-RPA.

Specific binding of RPA to WRN- or WRN239–499-coated wellswas analyzed according to Scatchard binding theory. The data

were analyzed by Hill plots as described under “Materials andMethods.” The transformed data were linear, indicating a sin-gle site on RPA for binding to either WRN or WRN239–499. Theapparent dissociation constants (Kd) for WRN-RPA andWRN239–499-RPA were 7.2 nM and 11.7 nM, respectively (TableII). These results, together with yeast two-hybrid and affinitypull-down studies, demonstrate a direct physical interactionbetween WRN and RPA mediated by the N-terminal domain ofWRN harboring the direct repeat elements. WRN239–499 bindsto RPA with similar affinity to that observed with the WRN-RPA interaction.

Analysis of DNA Unwinding Reactions Catalyzed by WRNand a WRN Helicase Domain Fragment—The results fromyeast two-hybrid studies, WRN affinity pull-down experiments,and ELISA measurements together suggest that the N-termi-nal region of WRN harboring the acidic repeat is important forthe physical interaction with the RPA 70-kDa subunit. Tobetter understand the importance of the WRN-RPA physicalinteraction for their functional interaction, we set out to test aWRN protein that lacked the acidic direct repeats in functionalhelicase assays on long DNA duplex substrates in the presenceof RPA. A baculovirus construct encoding full-length WRNprotein lacking one or both of the direct repeats was con-structed; however, the engineered recombinant WRN proteinswere insoluble under a number of viral infection and/or celllysis conditions, preventing us from being able to purify them.We next sought to test a catalytically active WRN helicasedomain fragment, WRNH-R, which lacks the N-terminal regioncontaining the acidic direct repeat sequence (Fig. 4A). AlthoughWRNH-R retains the RQC domain, the recombinant fragmentlacks a C-terminal region containing the HRD motif.

TABLE IIApparent dissociation constants (Kd) for interactions between

WRN or BLM with RPASpecific binding of RPA to wells coated with WRN, WRN239–499,

WRNH-R, WRN1072–1443, BLM, BLM1–477, or BLM966–1470 was analyzedby ELISA. Hill plots were used to analyze data.

RecQ protein/fragment Kd with RPA

nM

WRN 7.20WRN239–499 8.05WRNH-R NDa

WRN1072–1443 NDBLM 6.51BLM1–477 5.80BLM966–1470 ND

a ND, Kd was not determined.

FIG. 5. High affinity and low affin-ity RPA binding sites reside in theN-terminal and C-terminal regions ofWRN protein, respectively. Full-length and various truncated WRN pro-teins were used to coat microtiter wells(18 nM application) and were subse-quently incubated with increasing con-centrations (0–96 nM) of RPA heterotri-mer for 1 h at 30 °C. Following washing,bound RPA was detected by ELISA usinga mouse monoclonal antibody againstRPA70. Absorbance readings at eachpoint were corrected by subtracting abackground A490 reading obtained fromBSA-coated wells. The values representthe mean of three independent experi-ments performed in duplicate with stand-ard deviation (S.D.) indicated by errorbars. Filled circle, WRN; open circle,WRN239–499; filled square, WRNH-R; opensquare, WRN1072–1432.

Mapping of RPA Interaction Domain on WRN and BLM Helicases29500

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 8: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

We first investigated the DNA unwinding activity ofWRNH-R on a 19-bp forked duplex substrate. The forked duplexwas previously demonstrated to be a preferred helicase sub-strate of full-length WRN (27). The substrate was efficientlyunwound by full-length WRN (20 and 40 fmol) in the 15-minreaction (Supplemental Fig. S2). At lower protein levels (2.5and 5 fmol), WRN and WRNH-R unwound similar percentagesof the DNA substrate. Slightly more substrate was unwound byWRN compared with WRNH-R at the 10-fmol protein amount.At 20-fmol protein, full-length WRN protein unwound �1.53-fold more of the substrate than WRNH-R. At 40 fmol of protein,�88% of the forked duplex was unwound by WRN, whereas74% of the substrate was unwound by WRNH-R. Overall, theresults suggest that WRNH-R is only modestly compromised inits helicase activity on a 19-bp forked duplex compared with thefull-length recombinant WRN protein.

We next wanted to compare the helicase activity of WRNH-R

with full-length WRN on a series of increasing length M13partial duplex substrates, because we have previously foundthat WRN requires the presence of RPA to efficiently unwindthe longer duplex tracts. Because WRNH-R was slightly lessactive than full-length WRN in its ability to unwind the 19-bpforked duplex, a series of WRN or WRNH-R protein concentra-tions were tested on a 28-bp M13 partial duplex substrate todetermine protein concentrations that would yield comparablelevels of unwinding for full-length WRN and the WRNH-R he-licase domain fragment. In a 15-min reaction, it was deter-mined that a similar percentage of the 28-bp M13 partial du-plex substrate was unwound by 20 nM WRNH-R compared with15 nM WRN (Supplemental Fig. S3A). We then examined theunwinding kinetics for the M13 28-bp partial duplex usingthese protein levels over a 32-min time course (SupplementalFig. S3B). Increasing levels of DNA unwinding by WRNH-R

were observed throughout the early time points; helicase activ-ity achieved a plateau of �90% substrate unwound at 15 min.A quantitative comparison demonstrated similar kinetics ofunwinding by WRN (15 nM) and WRNH-R (20 nM) throughoutthe 15-min time course on the 28-bp partial duplex substrate.

Having normalized the helicase activity of WRN andWRNH-R on a short 28-bp M13 partial duplex substrate, wenext compared the helicase activities of full-length WRN andthe WRNH-R fragment on longer M13 duplex substrates of 50,70, and 100 bp. As expected from our previous characterizationof WRN helicase activity on longer duplex DNA substrates (14,15), WRN alone, in the absence of RPA, did not displace theannealed labeled strand complementary to the M13 sequencefor these longer M13 partial duplex substrates (data notshown). RPA alone also did not denature any of these partialduplex substrates (data not shown). In the presence of RPA (87nM heterotrimer), WRN (15 nM) efficiently unwound a 50-bpM13 partial duplex substrate, resulting in 94% of the substrateunwound in 2 min (Fig. 6A). In contrast, WRNH-R unwoundonly 7% of the substrate by 2 min. Although a linear increase inunwinding of the 50-bp partial duplex substrate by WRNH-R

was observed up to 8 min, only approximately one-third of thesubstrate was unwound compared with the amount of sub-strate unwound by wild-type WRN within 2 min. We nextcompared the helicase activities of WRN and WRNH-R on alonger 70-bp M13 partial duplex (Fig. 6B). Full-length WRNhad unwound 88% of this substrate by 2 min, compared withonly 3.7% of the substrate being unwound by WRNH-R duringthe same time period. By the end of the 32-min time course,WRNH-R had unwound 48.9% of the 70-bp substrate, which wassignificantly less (�2-fold) than the helicase activity catalyzedby full-length WRN after only 2 min. On a 100-bp partialduplex, WRN unwound �30% of the substrate in 2 min; in

contrast, hardly any detectable unwinding of the 100-bp partialduplex was observed by WRNH-R in the same 2-min period (Fig.6C). A kinetic analysis of the helicase activity data over thefirst 4 min of the reaction demonstrated that WRN unwound8.9% of the 100-bp partial duplex per minute. In contrast,WRNH-R poorly unwound the 100-bp substrate over the entiretime course, resulting in only 9.7% of the duplex DNA unwoundafter 32 min (Fig. 6D). Thus, WRNH-R unwound 0.6% of the100-bp substrate per min, a 14.3-fold lower rate compared withthe reaction catalyzed by full-length WRN.

In the presence of RPA, WRN can unwind very long DNAduplexes, including an 849-bp M13 partial duplex substrate(15). As previously published (15), we found that neither WRNnor RPA alone can denature this duplex DNA (data not shown).However, in the presence of RPA, WRN was able to unwind asignificant fraction of the 849-bp partial duplex substrate bythe end of the 32-min incubation (Supplemental Fig. S4A). Incontrast, WRNH-R failed to unwind the 849-bp DNA duplex ineither the presence or absence of RPA (Supplemental Fig. S4B).From these results, we conclude that, although WRNH-R canefficiently unwind a short 28-bp M13 partial duplex substrate,this WRN helicase domain fragment is severely compromisedin its ability to unwind longer DNA duplex substrates in whichthe unwinding activity of the full-length WRN helicase is de-pendent upon the presence of RPA.

N-terminal Region of Bloom Syndrome Helicase Binds RPAand Is Important for RPA Stimulation of BLM Helicase Activ-ity—The compromised ability of WRNH-R to unwind long RPA-dependent DNA substrates suggested that the N-terminal do-main of WRN, which mediates the high affinity physicalinteraction with RPA, is likely to be important for the func-tional interaction between RPA and WRN. Although the N-terminal sequence of BLM protein does not display as great anextent of sequence homology to WRN as found in the helicaseand RQC domains, BLM, like WRN, contains acidic regionslocated before the conserved helicase domain that may be im-portant in mediating a similar physical interaction with RPA.To address this possibility, we tested a purified recombinantMBP-BLM protein fragment that contains three highly acidicregions of the BLM protein, BLM1–447 (Fig. 7A), for directbinding to RPA using the ELISA assay. As observed for full-length BLM protein, the MBP-BLM1–447 fragment exhibited adose-dependent and saturable signal for its interaction withthe immobilized RPA. Although the BLM1–447-RPA signal was�2-fold less than that obtained using full-length BLM, theplateau was approached at the same concentration of BLM1–447

and BLM, 45 nM (Fig. 7B). The colorimetric signal obtainedfrom either BLM-RPA or BLM1–447-RPA interaction was re-sistant to the presence of ethidium bromide (10 �g/ml, data notshown), indicating that a contaminating DNA bridge was notresponsible for the positive signal. In comparison to BLM orBLM1–447, a considerably weaker signal was detected when aC-terminal BLM fragment harboring the RQC and HRD do-mains was incubated with the RPA-bound wells (Fig. 7B),suggesting that the BLM966–1470-RPA interaction was of con-siderably lower affinity than that of BLM1–447-RPA or BLM-RPA. Specific binding of RPA to BLM- or BLM1–447-coatedwells was analyzed according to Scatchard binding theory. Thetransformed data were linear, indicating a single site on RPAfor binding to either BLM or BLM1–447. The apparent dissoci-ation constants (Kd) for BLM-RPA and BLM1–447-RPA were6.51 nM and 5.80 nM, respectively (Table II).

Previously, we determined that RPA can stimulate BLMhelicase to efficiently unwind M13 partial duplex substrates aslong as 259 bp, and to a lesser extent a long 849-bp partial

Mapping of RPA Interaction Domain on WRN and BLM Helicases 29501

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 9: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

duplex (18). In the current study, under slightly modified re-action conditions, we observed that BLM helicase was able toefficiently unwind a 849-bp M13 partial duplex in the presenceof RPA, resulting in 93% of the substrate unwound, whereas nodetectable BLM unwinding was observed in the absence of RPA(Fig. 8). To address the potential functional importance of theN-terminal region of BLM protein, which physically interactswith RPA, for BLM helicase activity on a long DNA duplexsubstrate, we tested a catalytically active MBP-BLM recombi-nant protein, BLM642–1290, for unwinding of the 849-bp M13substrate. This particular recombinant BLM helicase domainfragment retains DNA helicase activity on short linear DNAduplexes (33) and an M13 69-bp partial duplex substrate sim-

ilar to that of full-length BLM.3 As shown in Fig. 8, very little(�1%) unwinding of the 849-bp partial duplex substrate byBLM642–1290 was detected in the presence of RPA. These re-sults suggest that the N-terminal domain of the BLM protein,which physically binds to RPA, is important for unwinding oflong DNA duplexes in which the BLM helicase reaction isdependent upon the presence of RPA.

3 K. M. Doherty, J. A. Sommers, M. D. Gray, J. W. Lee, C. von Kobbe,N. H. Thoma, R. P. Kureekattil, M. K. Kenny, and R. M. Brosh, Jr.,unpublished data.

FIG. 6. Stimulation of WRN or WRNH-R helicase activities by RPA on increasing lengths of M13 partial duplex DNA substrate. WRNprotein (15 nM) or WRNH-R (20 nM) was incubated with RPA (87 nM) and the 50-bp (A), 69-bp (B), or the 100-bp (C) M13 partial duplex substrates(0.125 nM) under the standard helicase reaction conditions as described under “Materials and Methods.” Reactions were quenched at the indicatedtime points. Helicase reaction products were resolved on native 10% polyacrylamide gels. Phosphorimaging images of representative gels areshown. A heat-denatured DNA substrate control is indicated by the filled triangle. D, percent displacement of the radiolabeled DNA strand fromthe M13 partial duplex substrate was plotted versus time for helicase reactions containing WRN (left) and WRNH-R (right) and the 50-bp (filledcircle), 69-bp (open circle), or 100-bp (filled square) M13 partial duplex substrates. Helicase data represent the mean from at least threeindependent experiments with S.D. indicated by error bars.

Mapping of RPA Interaction Domain on WRN and BLM Helicases29502

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 10: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

DISCUSSION

RPA is an essential protein in the cellular processes ofDNA replication, repair and recombination, and RNA tran-scription (16). In addition to its ability to bind ssDNA withhigh affinity and a defined polarity, the heterotrimeric pro-tein physically interacts with a number of nuclear DNA-metabolizing proteins and modulates their functional or cat-alytic activities in a specific manner (for review, see Ref. 16).We have been interested in how the DNA-unwinding reac-tions catalyzed by human RecQ helicases are stimulated byRPA, because heterologous ssDNA-binding proteins either

poorly or completely fail to stimulate DNA unwinding byhuman WRN (15), BLM (18), RECQ1 (19), or RECQ5� (20)depending on the duplex length or helicase in question. Ofthe human RecQ DNA helicases examined, WRN, BLM, andRECQ1 all physically interact with RPA, further supportingthe notion of a specific functional interaction between thehelicase and the single-stranded DNA-binding protein. Inthis work, we sought to define the physical interaction site(s)between RPA and WRN and to investigate whether the WRN-RPA physical interaction is important for RPA-dependentstimulation of WRN-catalyzed DNA unwinding.

The results from our mapping studies reveal that WRN hasa high affinity RPA interaction site located within its N termi-nus, and a second site of lower affinity in its C terminus. Inaddition to its high affinity, the N-terminal RPA interactionsite is of particular interest, because it harbors a perfectly anddirectly repeated acidic sequence element that was shown togive a strong signal for interaction with RPA70 in vivo by theyeast two-hybrid assay. Up to this point, although a number ofthe conserved domains or motifs of the WRN protein have beenextensively characterized, the importance of the N-terminalregion harboring the acidic direct repeats for WRN catalyticfunction or protein interactions had not been characterized.

Interestingly, the Xenopus homolog of WRN, FFA-1, is uniqueamong the eukaryotic RecQ homologs in that it contains one-halfof the identical direct repeat found in WRN (8). Molecular andcellular studies in the Xenopus system have provided some in-sight into the biological importance of the interaction betweenRPA and the eukaryotic RecQ helicase FFA-1. The assembly of

FIG. 8. Comparison of BLM and BLM642–1290 to unwind a 849-bppartial duplex. BLM (15 nM), BLM642–1290 (15 nM), or WRN (15 nM)was incubated with RPA (87 nM) and the 849-bp M13 partial duplexsubstrate (0.125 nM) under the standard helicase reaction conditions asdescribed under “Materials and Methods.” Reactions were quenchedafter 15 min, and helicase reaction products were resolved on native10% polyacrylamide gels. A phosphorimaging image of a representativegel is shown. The band above the released 849-nucleotide oligonucleo-tide represents the unannealed radiolabeled oligomer present in the849-bp helicase substrate preparation. A heat-denatured DNA sub-strate control is indicated by the filled triangle.

FIG. 7. N-terminal region of Bloomsyndrome helicase directly bindsRPA with high affinity. A, scheme ofBLM full-length and fragment proteinsused for these studies. B, full-length andvarious truncated BLM proteins wereused to coat microtiter wells (18 nM appli-cation) and were subsequently incubatedwith increasing concentrations (0–96 nM)of RPA heterotrimer for 1 h at 30 °C. Fol-lowing washing, bound RPA was detectedby ELISA using a mouse monoclonal an-tibody against RPA70. Absorbance read-ings at each point were corrected by sub-tracting a background A490 readingobtained from BSA-coated wells. The val-ues represent the mean of three inde-pendent experiments performed in dupli-cate with standard deviation (S.D.)indicated by error bars. Filled circle,BLM; open circle, BLM1–447; filled square,BLM966–1417.

Mapping of RPA Interaction Domain on WRN and BLM Helicases 29503

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 11: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

RPA-containing replication foci in X. laevis egg extracts requiresFFA-1 (34). FFA-1 binds to nuclear chromatin at an early stage offormation of functional replication origin complexes and enablesRPA to subsequently bind to the foci. RPA was shown to physi-cally interact with FFA-1 and stimulate its helicase activity in aspecific manner, because the T4 gene 32 ssDNA-binding proteindid not stimulate FFA-1 helicase activity (35). The region ofFFA-1 required for the RPA interaction was found to residewithin the N-terminal 291 amino acids, a region that containsthe single acidic repeat element of FFA-1 (35). The notion thatRPA stimulates FFA-1 helicase activity through a direct proteininteraction was further supported by the observation that addi-tion of a GST-FFA-1 fragment, that mediates the physical inter-action with RPA to a reaction mixture containing RPA andFFA-1, inhibited FFA-1-catalyzed unwinding of a DNA duplexsubstrate in a dose-dependent manner but did not affect bindingof RPA to ssDNA (35). Presumably, the inhibitory GST-FFA-1fusion protein can prevent RPA stimulation of FFA-1 helicaseactivity by competing against the full-length FFA-1 for interac-tion with RPA. Consistent with this idea, the FFA-1 fragmentharboring the RPA interaction site can also exert a dominantnegative effect on DNA replication in reconstituted Xenopus nu-clei, suggesting a biological importance for the FFA-1-RPA inter-action (35).

To gain some insight into the WRN-RPA functional interac-tion, we examined the helicase activity of a recombinant heli-case core domain fragment, WRNH-R, that contains the con-served RecQ helicase motifs and RQC region but lacks the highaffinity RPA interaction site in the N terminus as well as aC-terminal portion, on a series of partial duplex DNA sub-strates that varied in their number of base pairs. These studiesled to the conclusion that, although this WRN helicase domainfragment is only mildly reduced in its helicase activity on shortDNA duplex substrates, its ability to unwind DNA duplexsubstrates of 50 bp or greater is seriously impaired despite thepresence of the WRN auxiliary factor RPA in the reactionmixtures. Although it is tempting to conclude that the absenceof the high affinity RPA interaction site in the WRN helicasecore domain fragment is solely responsible for the poor unwind-ing of the longer duplex substrates, other factors may contrib-ute. The slightly reduced helicase activity of WRNH-R on shortduplex substrates compared with the wild-type enzyme sug-gests that regions outside the WRN helicase domain may affectthe intrinsic catalytic activity of the helicase, particularly forlonger duplexes. For example, in addition to the high affinityRPA interaction site, WRNH-R also lacks the HRD motif in theC-terminal region of the full-length WRN protein. Recent evi-dence demonstrates that a C-terminal WRN fragment harbor-ing this motif has the ability to bind various oligonucleotide-based duplex DNA substrates (7). It is conceivable that theHRD motif may contribute to the interaction of full-lengthWRN with the DNA substrate by conferring processivity to theenzyme when RPA is present in the helicase reaction. Anargument that can be made against this explanation for thedefective helicase activity of WRNH-R on RPA-dependent heli-case substrates is that the BLM helicase domain fragmentBLM642–1290, which retains the HRD motif but lacks the highaffinity RPA interaction site, is also defective in unwindingRPA-dependent DNA duplex substrates that the full-lengthBLM enzyme can unwind efficiently in the presence of RPA.However, further molecular studies will be necessary to ad-dress the functional importance of the HRD motif in the cata-lytic activities of WRN or BLM proteins.

It was recently reported that the RPA70 subunit stimulatesWRN helicase activity to the same extent as the RPA hetero-trimer (17). Further mapping demonstrated that the N-termi-

nal half of RPA70 (residues 1–441) was sufficient for stimula-tion of WRN helicase activity. A mutant heterotrimer lackingresidues 1–168 of RPA70 stimulated WRN helicase activity asefficiently as wild-type RPA heterotrimer, suggesting that theWRN-RPA functional interaction is mediated by residues 169–441 (17). By ELISA assays, it was determined that the WRNbinding domain for RPA70 resides within residues 100–300and overlaps with the ssDNA binding domain, residues 150–450 (17). Thus the WRN interacting domain and ssDNA bind-ing domain of RPA overlap with each other, suggesting that thessDNA and WRN-protein binding activities of RPA70 are func-tionally intertwined.

Structural studies of RPA have revealed that the N-terminalregion of the human RPA70 subunit folds into a five-strandedanti-parallel �-barrel (36). Two loops on the side of the struc-ture (residues 31–42 and 87–92) form a large basic cleft con-taining five arginines and a lysine. This cleft has been thoughtto be a region that may bind acidic motifs on interacting pro-teins. Although the minimal physical interaction site for WRNwas mapped to residues 100–300 of RPA70, it was also shownthat mutant RPA heterotrimers containing N-terminal trunca-tions of RPA70, including deletion of the first 112 amino acids,impaired the physical interaction between RPA and WRN (17).In summary, the results of the Loeb study (17) are consistentwith ours in that the N-terminal half of RPA70 mediates WRNbinding. We have extended these observations by mapping theRPA interaction sites on the WRN protein, showing that anN-terminal region of WRN mediates a high affinity interactionwith RPA.

We had previously characterized a physical and functionalinteraction between BLM helicase and RPA (18). BLM andRPA were shown to interact via the RPA70 subunit, similar toWRN. Here we report that the domain for RPA70 bindingresides within the N-terminal region of BLM and that a heli-case domain fragment of BLM lacking the RPA70 interactionsite is unable to unwind long DNA duplexes that the full-lengthBLM helicase can efficiently unwind if RPA is present. Ourstudies suggest that human RecQ helicases, which physicallyand functionally interact with RPA, may do so via domains ofthe helicase that are functionally conserved but do not displayextensive sequence homology. The fact that helicase domainfragments of both BLM and WRN that lack the RPA interactiondomain can efficiently unwind short duplex tracts suggeststhat the RPA interaction domain is folded independently of thecatalytic helicase domain. Although BLM does not have thesame acidic repeat elements that WRN does, the RPA70 inter-action site of BLM contains three highly acidic regions thatmay be important in the RPA interaction. Importantly, theresults from the BLM/WRN helicase and RPA interaction stud-ies suggest that the physical interaction with RPA is an impor-tant component of the mechanism for the stimulation of DNAhelicase activity.

The biological significance of the WRN-RPA or BLM-RPA in-teraction may be highly relevant to our understanding of theDNA metabolic defects in WS and BS. WS cells have a prolongedS phase (37), asymmetry of DNA replication fork progression(38), slower rate of repair associated with DNA damage inducedin S-phase, reduced induction of RAD51 foci, and a higher level ofstrand breaks (39). The in vivo evidence implicating WRN in therecovery of DNA synthesis after replication arrest poses thequestion of whether WRN functions together with RPA in acritical step to resolve a key replication or recombinational inter-mediate that arises from fork stalling or collapse. RPA has beenfound to co-localize with WRN upon replication arrest (40) andDNA damage (41), suggesting that the two proteins may indeedcollaborate to perform certain cellular function(s).

Mapping of RPA Interaction Domain on WRN and BLM Helicases29504

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 12: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

The most characteristic features of BS cells are an elevatedfrequency of chromosome breaks and exchanges (42, 43) aswell as an increase in the level of reciprocal exchanges be-tween sister chromatids (44). Like WS cells, BS cells displayaberrant recombination and abnormalities in DNA replica-tion that include an extended S phase and the accumulationof abnormal replication intermediates compared with normalcells (45, 46). It is conceivable that BLM helicase and RPAmay also act together to facilitate efficient DNA replicationand/or recombination.

S. cerevisiae has a sole RecQ homologue, Sgs1. sgs1 loss offunction mutations result in the development of age-associatedphenotypes, including a 60% reduction in life span and nucle-olar fragmentation (47). An sgs1 deletion mutant exhibitsgenomic instability characterized by elevated homologous andhomeologous recombination throughout the genome, particu-larly within the ribosomal DNA array (47, 48). It was reportedthat expression of either human BLM or WRN can suppress thehyper recombination phenotype of the sgs1 mutant (49), sug-gesting that the human RecQ helicases have a role in main-taining genomic stability by preventing aberrant recombina-tion. DNA polymerase stabilization at stalled replication forksrequires the ATM-related kinase Mec1 and Sgs1 (50). A modelwas proposed whereby Sgs1 helicase resolves aberrantly pairedstructures at stalled forks to maintain single-stranded DNAthat allows RPA and Mec1 to promote DNA polymerase asso-ciation. The observation that Sgs1 is present at replicationforks and binds RPA is suggested to be important for polymer-ase assembly/stabilization. In future studies, insight into thebiological importance of the WRN-RPA or BLM-RPA interac-tions at stalled forks may be gained by genetic complementa-tion studies using WRN or BLM mutants that are specificallydefective in their interaction with RPA.

Acknowledgments—We thank Dr. I. Hickson (Cancer ResearchUK Laboratories) for kindly providing recombinant BLM protein,BLM642–1290, and expression constructs for MBP fusion proteins ofBLM. We thank Drs. S. Sharma, R. Gupta, and V. Bohr (Laboratory ofMolecular Gerontology, NIA, National Institutes of Health) and Dr. I.Hickson (Cancer Research UK Laboratories) for critical reading ofthe manuscript.

REFERENCES

1. Bachrati, C. Z., and Hickson, I. D. (2003) Biochem. J. 374, 577–6062. Harrigan, J. A., and Bohr, V. A. (2003) Biochimie (Paris) 85, 1185–11933. Hickson, I. D. (2003) Nat. Rev. Cancer 3, 169–1784. Morozov, V., Mushegian, A. R., Koonin, E. V., and Bork, P. (1997) Trends.

Biochem. Sci. 22, 417–4185. Brosh, R. M., Jr., and Bohr, V. A. (2002) Exp. Gerontol. 37, 491–5066. von Kobbe, C., and Bohr, V. A. (2002) J. Cell Sci. 115, 3901–39077. von Kobbe, C., Thoma, N. H., Czyzewski, B. K., Pavletich, N. P., and Bohr,

V. A. (2003) J. Biol. Chem. 278, 52997–530068. Yu, C. E., Oshima, J., Fu, Y. H., Wijsman, E. M., Hisama, F., Alisch, R.,

Matthews, S., Nakura, J., Miki, T., Ouais, S., Martin, G. M., Mulligan, J.,and Schellenberg, G. D. (1996) Science 272, 258–262

9. Balajee, A. S., Machwe, A., May, A., Gray, M. D., Oshima, J., Martin, G. M.,Nehlin, J. O., Brosh, R. M., Jr., Orren, D. K., and Bohr, V. A. (1999) Mol.Biol. Cell 10, 2655–2668

10. Opresko, P. L., Cheng, W. H., von Kobbe, C., Harrigan, J. A., and Bohr, V. A.(2003) Carcinogenesis 24, 791–802

11. Gray, M. D., Shen, J. C., Kamath-Loeb, A. S., Blank, A., Sopher, B. L., Martin,

G. M., Oshima, J., and Loeb, L. A. (1997) Nat. Genet. 17, 100–10312. Shen, J. C., Gray, M. D., Oshima, J., and Loeb, L. A. (1998) Nucleic Acids Res.

26, 2879–288513. Suzuki, N., Shimamoto, A., Imamura, O., Kuromitsu, J., Kitao, S., Goto, M.,

and Furuichi, Y. (1997) Nucleic Acids Res. 25, 2973–297814. Opresko, P. L., Laine, J. P., Brosh, R. M., Jr., Seidman, M. M., and Bohr, V. A.

(2001) J. Biol. Chem. 276, 44677–4468715. Brosh, R. M., Jr., Orren, D. K., Nehlin, J. O., Ravn, P. H., Kenny, M. K.,

Machwe, A., and Bohr, V. A. (1999) J. Biol. Chem. 274, 18341–1835016. Wold, M. S. (1997) Annu. Rev. Biochem. 66, 61–9217. Shen, J. C., Lao, Y., Kamath-Loeb, A., Wold, M. S., and Loeb, L. A. (2003)

Mech. Ageing Dev. 124, 921–93018. Brosh, R. M., Jr., Li, J. L., Kenny, M. K., Karow, J. K., Cooper, M. P.,

Kureekattil, R. P., Hickson, I. D., and Bohr, V. A. (2000) J. Biol. Chem. 275,23500–23508

19. Cui, S., Arosio, D., Doherty, K. M., Brosh, R. M., Jr., Falaschi, A., and Vindigni,A. (2004) Nucleic Acids Res. 32, 2158–2170

20. Garcia, P. L., Liu, Y., Jiricny, J., West, S. C., and Janscak, P. (2004) EMBO J.23, 2882–2891

21. Vojtek, A. B., Hollenberg, S. M., and Cooper, J. A. (1993) Cell 74, 205–21422. Sharma, S., Otterlei, M., Sommers, J. A., Driscoll, H. C., Dianov, G. L., Kao,

H. I., Bambara, R. A., and Brosh, R. M., Jr. (2004) Mol. Biol. Cell 15,734–750

23. Brosh, R. M., Jr., von Kobbe, C., Sommers, J. A., Karmakar, P., Opresko, P. L.,Piotrowski, J., Dianova, I., Dianov, G. L., and Bohr, V. A. (2001) EMBO J.20, 5791–5801

24. Karow, J. K., Newman, R. H., Freemont, P. S., and Hickson, I. D. (1999) Curr.Biol. 9, 597–600

25. Wu, L., Davies, S. L., North, P. S., Goulaouic, H., Riou, J. F., Turley, H., Gatter,K. C., and Hickson, I. D. (2000) J. Biol. Chem. 275, 9636–9644

26. Kenny, M. K., Schlegel, U., Furneaux, H., and Hurwitz, J. (1990) J. Biol.Chem. 265, 7693–7700

27. Brosh, R. M., Jr., Waheed, J., and Sommers, J. A. (2002) J. Biol. Chem. 277,23236–23245

28. Lin, Y. L., Chen, C., Keshav, K. F., Winchester, E., and Dutta, A. (1996) J. Biol.Chem. 271, 17190–17198

29. Bochkareva, E., Korolev, S., Lees-Miller, S. P., and Bochkarev, A. (2002)EMBO J. 21, 1855–1863

30. Erdile, L. F., Heyer, W. D., Kolodner, R., and Kelly, T. J. (1991) J. Biol. Chem.266, 12090–12098

31. Gomes, X. V., and Wold, M. S. (1995) J. Biol. Chem. 270, 4534–454332. Henricksen, L. A., Umbricht, C. B., and Wold, M. S. (1994) J. Biol. Chem. 269,

11121–1113233. Janscak, P., Garcia, P. L., Hamburger, F., Makuta, Y., Shiraishi, K., Imai, Y.,

Ikeda, H., and Bickle, T. A. (2003) J. Mol. Biol. 330, 29–4234. Yan, H., Chen, C. Y., Kobayashi, R., and Newport, J. (1998) Nat. Genet. 19,

375–37835. Chen, C. Y., Graham, J., and Yan, H. (2001) J. Cell Biol. 152, 985–99636. Jacobs, D. M., Lipton, A. S., Isern, N. G., Daughdrill, G. W., Lowry, D. F.,

Gomes, X., and Wold, M. S. (1999) J. Biomol. NMR 14, 321–33137. Poot, M., Hoehn, H., Runger, T. M., and Martin, G. M. (1992) Exp. Cell Res.

202, 267–27338. Rodriguez-Lopez, A. M., Jackson, D. A., Iborra, F., and Cox, L. S. (2002) Aging

Cell 1, 30–3939. Pichierri, P., Franchitto, A., Mosesso, P., and Palitti, F. (2001) Mol. Biol. Cell

12, 2412–242140. Constantinou, A., Tarsounas, M., Karow, J. K., Brosh, R. M., Jr., Bohr, V. A.,

Hickson, I. D., and West, S. C. (2000) EMBO Reports 1, 80–8441. Sakamoto, S., Nishikawa, K., Heo, S. J., Goto, M., Furuichi, Y., and Shi-

mamoto, A. (2001) Genes Cells 6, 421–43042. Ellis, N. A., and German, J. (1996) Hum. Mol. Genet. 5, 1457–146343. German, J. (1993) Medicine (Baltimore) 72, 393–40644. Langlois, R. G., Bigbee, W. L., Jensen, R. H., and German, J. (1989) Proc. Natl.

Acad. Sci. U. S. A. 86, 670–67445. Gianneli, F., Benson, P. F., Pawsey, S. A., and Polani, P. E. (1977) Nature 265,

466–46946. Lonn, U., Lonn, S., Nylen, U., Winblad, G., and German, J. (1990) Cancer Res.

50, 3141–314547. Sinclair, D. A., and Guarente, L. (1997) Cell 91, 1033–104248. Myung, K., Chen, C., and Kolodner, R. D. (2001) Nature 411, 1073–107649. Yamagata, K., Kato, J., Shimamoto, A., Goto, M., Furuichi, Y., and Ikeda, H.

(1998) Proc. Natl. Acad. Sci. U. S. A. 95, 8733–873850. Cobb, J. A., Bjergbaek, L., Shimada, K., Frei, C., and Gasser, S. M. (2003)

EMBO J. 22, 4325–4336

Mapping of RPA Interaction Domain on WRN and BLM Helicases 29505

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 13: THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. …are all characterized by genomic instability, an elevated cancer incidence, and/or particular aspects of premature aging. Hu-man diseases

Jr.Kobbe, Nicolas H. Thoma, Raichal P. Kureekattil, Mark K. Kenny and Robert M. Brosh, Kevin M. Doherty, Joshua A. Sommers, Matthew D. Gray, Jae Wan Lee, Cayetano von

of the Werner and Bloom Syndrome HelicasesPhysical and Functional Mapping of the Replication Protein A Interaction Domain

doi: 10.1074/jbc.M500653200 originally published online June 17, 20052005, 280:29494-29505.J. Biol. Chem. 

  10.1074/jbc.M500653200Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

Supplemental material:

  http://www.jbc.org/content/suppl/2005/06/30/M500653200.DC1

  http://www.jbc.org/content/280/33/29494.full.html#ref-list-1

This article cites 50 references, 25 of which can be accessed free at

by guest on July 23, 2020http://w

ww

.jbc.org/D

ownloaded from