FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA...

11
Fluorescence Resonance Energy Transfer Studies of DNA Polymerase THE CRITICAL ROLE OF FINGERS DOMAIN MOVEMENTS AND A NOVEL NON-COVALENT STEP DURING NUCLEOTIDE SELECTION * Received for publication, February 28, 2014, and in revised form, April 23, 2014 Published, JBC Papers in Press, April 24, 2014, DOI 10.1074/jbc.M114.561878 Jamie B. Towle-Weicksel , Shibani Dalal , Christal D. Sohl § , Sylvie Doublie ´ , Karen S. Anderson § , and Joann B. Sweasy ‡1 From the Departments of Therapeutic Radiology and § Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520 and the Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont 05405 Background: DNA Pol participates in base excision repair by choosing correct dNTP to fill single-nucleotide gaps in DNA. Results: Pol experiences a non-covalent step with correct dNTP selection. Conclusion: Correct and incorrect dNTP incorporation by Pol are different. Significance: FRET-based system of Pol elucidates a mechanism of substrate choice necessary for understanding the molec- ular basis of human disease. During DNA repair, DNA polymerase (Pol ) is a highly dynamic enzyme that is able to select the correct nucleotide oppo- site a templating base from a pool of four different deoxynucleoside triphosphates (dNTPs). To gain insight into nucleotide selection, we use a fluorescence resonance energy transfer (FRET)-based system to monitor movement of the Pol fingers domain during catalysis in the presence of either correct or incorrect dNTPs. By labeling the fingers domain with ((((2-iodoacetyl)- amino)ethyl)amino)naphthalene-1-sulfonic acid (IAEDANS) and the DNA substrate with Dabcyl, we are able to observe rapid fingers clos- ing in the presence of correct dNTPs as the IAEDANS comes into contact with a Dabcyl-labeled, one-base gapped DNA. Our findings show that not only do the fingers close after binding to the correct dNTP, but that there is a second conformational change associated with a non-covalent step not previously reported for Pol . Further analyses suggest that this conformational change corresponds to the binding of the catalytic metal into the polymerase active site. FRET studies with incorrect dNTP result in no changes in fluorescence, indi- cating that the fingers do not close in the presence of incorrect dNTP. Together, our results show that nucleotide selection initially occurs in an open fingers conformation and that the catalytic pathways of cor- rect and incorrect dNTPs differ from each other. Overall, this study provides new insight into the mechanism of substrate choice by a polymerase that plays a critical role in maintaining genome stability. Approximately 20,000 DNA lesions are removed per human cell per day via the base excision repair (BER) 2 pathway (1). Short patch BER functions in the removal of single-base dam- age from DNA. BER is initiated by removal of a damaged base by a DNA glycosylase (2). A monofunctional glycosylase gener- ates an abasic (AP) site that is processed by apurinic/apyrimi- dinic AP endonuclease 1 (APE1), which nicks the sugar-phos- phate backbone to generate a 3 OH and a 5 deoxyribose phosphate, which is removed by Pol . Bifunctional glyco- sylases catalyze excision of the damaged base and AP site removal, leaving a 3 deoxyribose phosphate and a 5 phos- phate. APE1 catalyzes removal of the 3 deoxyribose phosphate to generate a 3 OH. In both cases, Pol fills in the gap and XRCC1-Ligase III or Ligase I seals the remaining nick in the DNA (3). Pol plays an important role in ensuring the fidelity of DNA synthesis during repair. The 39-kDa protein is a member of the X-family of DNA polymerases and has four domains: the 8-kDa lyase, thumb, palm, and fingers domains. Whereas the other three domains are responsible for DNA binding and catalytic activity, the fingers domain (residues 262–335) forms the nucleotide binding pocket and thus plays a central role in nucle- otide selection (4 –7). A series of crystal structures of Pol sug- gest that binding of dNTPs induces a conformational change whereby helix N of the fingers domain moves closer to the DNA (8). This conformational change occurs rapidly to form the active site, which catalyzes phosphodiester bond formation (also known as the chemistry step). Product release is consid- ered to be the rate-limiting step (Scheme 1) (9). Pol is known to misincorporate an incorrect nucleotide once in every 10,000 base insertions (10). The efficiency of incorrect incorporation is increased in several Pol variants that exhibit a mutator phenotype (11–15). Previous fluorescence studies with Pol performed by us (16) and others (17–19) observed move- ments of Pol associated with DNA synthesis using a 2-amino- purine reporter in the DNA along with the single Trp residue in the fingers domain. These studies, which did not monitor FRET, suggest that there is a step prior to chemistry that is not rate-limiting for DNA synthesis by Pol , which is likely asso- * This work was supported, in whole or in part, by National Institutes of Health Grants CA 080830 (to J. B. S.), GM49551 (to K. S. A.), and GM099289 (to C. D. S.). 1 To whom correspondence should be addressed: 333 Cedar St., P. O. Box 208040, New Haven, CT 06520. Tel.: 203-737-2626; Fax: 203-785-6309; E-mail: [email protected]. 2 The abbreviations used are: BER, base excision repair; Pol, DNA polymerase; IAEDANS-5, ((((2-iodoacetyl)amino)ethyl)amino)naphthalene-1-sulfonic acid; AP, abasic; APE1, AP endonuclease 1. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 23, pp. 16541–16550, June 6, 2014 © 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. JUNE 6, 2014 • VOLUME 289 • NUMBER 23 JOURNAL OF BIOLOGICAL CHEMISTRY 16541 by guest on June 22, 2020 http://www.jbc.org/ Downloaded from

Transcript of FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA...

Page 1: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

Fluorescence Resonance Energy Transfer Studies of DNAPolymerase �THE CRITICAL ROLE OF FINGERS DOMAIN MOVEMENTS AND A NOVEL NON-COVALENTSTEP DURING NUCLEOTIDE SELECTION*

Received for publication, February 28, 2014, and in revised form, April 23, 2014 Published, JBC Papers in Press, April 24, 2014, DOI 10.1074/jbc.M114.561878

Jamie B. Towle-Weicksel‡, Shibani Dalal‡, Christal D. Sohl§, Sylvie Doublie¶, Karen S. Anderson§,and Joann B. Sweasy‡1

From the Departments of ‡Therapeutic Radiology and §Pharmacology, Yale University School of Medicine, New Haven,Connecticut 06520 and the ¶Department of Microbiology and Molecular Genetics, University of Vermont,Burlington, Vermont 05405

Background: DNA Pol � participates in base excision repair by choosing correct dNTP to fill single-nucleotide gaps in DNA.Results: Pol � experiences a non-covalent step with correct dNTP selection.Conclusion: Correct and incorrect dNTP incorporation by Pol � are different.Significance: FRET-based system of Pol � elucidates a mechanism of substrate choice necessary for understanding the molec-ular basis of human disease.

During DNA repair, DNA polymerase � (Pol �) is a highlydynamic enzyme that is able to select the correct nucleotide oppo-site a templating base from a pool of four different deoxynucleosidetriphosphates (dNTPs). To gain insight into nucleotide selection,we use a fluorescence resonance energy transfer (FRET)-basedsystem to monitor movement of the Pol � fingers domainduring catalysis in the presence of either correct or incorrectdNTPs. By labeling the fingers domain with ((((2-iodoacetyl)-amino)ethyl)amino)naphthalene-1-sulfonicacid(IAEDANS)and theDNA substrate with Dabcyl, we are able to observe rapid fingers clos-ing in the presence of correct dNTPs as the IAEDANS comes intocontact with a Dabcyl-labeled, one-base gapped DNA. Our findingsshow that not only do the fingers close after binding to the correctdNTP, but that there is a second conformational change associatedwith a non-covalent step not previously reported for Pol �. Furtheranalyses suggest that this conformational change corresponds to thebinding of the catalytic metal into the polymerase active site. FRETstudieswithincorrectdNTPresultinnochangesinfluorescence,indi-cating that the fingers do not close in the presence of incorrect dNTP.Together, our results show that nucleotide selection initially occurs inan open fingers conformation and that the catalytic pathways of cor-rect and incorrect dNTPs differ from each other. Overall, this studyprovides new insight into the mechanism of substrate choice by apolymerase that plays a critical role in maintaining genome stability.

Approximately 20,000 DNA lesions are removed per humancell per day via the base excision repair (BER)2 pathway (1).Short patch BER functions in the removal of single-base dam-

age from DNA. BER is initiated by removal of a damaged baseby a DNA glycosylase (2). A monofunctional glycosylase gener-ates an abasic (AP) site that is processed by apurinic/apyrimi-dinic AP endonuclease 1 (APE1), which nicks the sugar-phos-phate backbone to generate a 3� OH and a 5� deoxyribosephosphate, which is removed by Pol �. Bifunctional glyco-sylases catalyze excision of the damaged base and AP siteremoval, leaving a 3� deoxyribose phosphate and a 5� phos-phate. APE1 catalyzes removal of the 3� deoxyribose phosphateto generate a 3� OH. In both cases, Pol � fills in the gap andXRCC1-Ligase III� or Ligase I seals the remaining nick in theDNA (3).

Pol � plays an important role in ensuring the fidelity of DNAsynthesis during repair. The 39-kDa protein is a member of theX-family of DNA polymerases and has four domains: the 8-kDalyase, thumb, palm, and fingers domains. Whereas the otherthree domains are responsible for DNA binding and catalyticactivity, the fingers domain (residues 262–335) forms thenucleotide binding pocket and thus plays a central role in nucle-otide selection (4 –7). A series of crystal structures of Pol � sug-gest that binding of dNTPs induces a conformational changewhereby helix N of the fingers domain moves closer to the DNA(8). This conformational change occurs rapidly to form theactive site, which catalyzes phosphodiester bond formation(also known as the chemistry step). Product release is consid-ered to be the rate-limiting step (Scheme 1) (9).

Pol � is known to misincorporate an incorrect nucleotide oncein every 10,000 base insertions (10). The efficiency of incorrectincorporation is increased in several Pol � variants that exhibit amutator phenotype (11–15). Previous fluorescence studies withPol � performed by us (16) and others (17–19) observed move-ments of Pol � associated with DNA synthesis using a 2-amino-purine reporter in the DNA along with the single Trp residue inthe fingers domain. These studies, which did not monitorFRET, suggest that there is a step prior to chemistry that is notrate-limiting for DNA synthesis by Pol �, which is likely asso-

* Thisworkwassupported, inwholeor inpart,byNational InstitutesofHealthGrantsCA 080830 (to J. B. S.), GM49551 (to K. S. A.), and GM099289 (to C. D. S.).

1 To whom correspondence should be addressed: 333 Cedar St., P. O. Box208040, New Haven, CT 06520. Tel.: 203-737-2626; Fax: 203-785-6309;E-mail: [email protected].

2 The abbreviations used are: BER, base excision repair; Pol, DNA polymerase;IAEDANS-5, ((((2-iodoacetyl)amino)ethyl)amino)naphthalene-1-sulfonicacid; AP, abasic; APE1, AP endonuclease 1.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 23, pp. 16541–16550, June 6, 2014© 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

JUNE 6, 2014 • VOLUME 289 • NUMBER 23 JOURNAL OF BIOLOGICAL CHEMISTRY 16541

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 2: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

ciated with fingers closing. Many of these studies were con-ducted on a recessed DNA substrate, which is not the physio-logically relevant substrate for Pol � (20, 21), as Pol � preferssingle nucleotide-gapped DNA. Given these limitations, wedeveloped a FRET-based approach to monitor the movementsof the fingers domain during DNA synthesis by Pol �, using asingle nucleotide-gapped DNA substrate.

Our results reveal that there is a rapid closing of the fingersdomain in the presence of the correct nucleotide, followed byan additional and not previously identified non-covalent stepthat precedes chemistry and product release. The conforma-tional changes we observe with correct dNTP incorporation areabsent in the presence of the incorrect dNTP, suggesting thatPol � catalysis is different for incorporation of correct versusincorrect nucleotides.

EXPERIMENTAL PROCEDURES

Deoxyoligonucleotides and DNA sequences used in thispaper are described (Table 1).

Generation of C239S/C267C/V303C Pol �—Wild-type (WT)human DNA polymerase � contains three endogenous cysteineresidues. The mutations of Cys-239 and Cys-267 to serineswere introduced into a tagless, human WT Pol � (modifiedpET28a) by site-directed mutagenesis (Stratagene) and did notaffect Pol � function (Table 2). An additional residue change bysite-directed mutagenesis converted Val-303 to Cys to facilitatethiol-reactive labeling with ((((2-iodoacetyl)amino)ethyl)amino)naphthalene-1-sulfonic acid (IAEDANS).

Expression and Purification of Modified Pol �—Plasmid con-taining the modified Pol � gene was expressed in bacteria aspreviously described (22). Protein was stored at 4 °C until label-ing. Pol � protein may be stored in 15% glycerol at �80 °C forseveral months with a subsequent thaw and labeling with noeffect on Pol � activity.

Labeling of Modified Pol �—The selection of the fluoro-phore/quencher pair as well as the procedure for labeling wasadapted from Joyce et al. (23). The protein was kept at 4 °Cduring the labeling process. Approximately 100 �M Pol � wasreduced with 1 mM dithiothreitol (DTT) for 45 min. The DTTwas removed from the sample by gel filtration using a PD-10desalting column (GE Healthcare Life Sciences) and exchangedinto Buffer C (50 mM Tris-HCl, pH 7.6, 100 mM NaCl, 150 �M

Tris(2-carboxyethyl)phosphine-HCl (Thermo Scientific)) fol-lowing the manufacturer’s spin protocol. A 10 mM stock ofIAEDANS was prepared in Buffer D (100 mM Tris-HCl, pH 8,and 100 mM NaCl). A total of 500 �M IAEDANS solution wasslowly added to 100 �M Pol � and allowed to react for 16 –18 hat 4 °C. The reaction was stopped with the addition of 0.5 mM

DTT and incubation for 20 min. The protein mixture was sep-arated from free IAEDANS by a second PD-10 column (follow-ing gravity protocol) and exchanged into Buffer C. Fractions of500 �l were collected, UV-visible analyzed at 336 and 280 nm,and concentrated to a final concentration of �60 –100 �M.Samples were analyzed by 10% SDS-polyacrylamide gel electro-phoresis (PAGE) and stained with either Coomassie or viewedunder UV light (Fig. 2, A and B). A protein preparation contain-ing the C239S/C267S mutation was labeled in conjunction withV303C to ensure that Cys-178 was not labeled. Fig. 2, A and B,demonstrates that the thiol reaction occurred only at position303. The labeling was determined to be greater than 89%. Forsimplicity, the modified, labeled WT is referred to as Pol�-AEDANS.

DNA Substrates—Deoxyoligonucleotides for the one-basegapped DNA, including the Dabcyl-T modified DNA, werepurchased from the Keck Oligo Synthesis Resource (Yale Uni-versity) and purified by PAGE (Table 1). Deoxyoligonucleotideswere annealed together to form the preferred DNA substrate ofPol �, as previously described (24). For consistency, nomencla-ture for the DNA substrates is as described by Joyce et al. (23).

Rapid Chemical Quench Experiments—Chemical quenchexperiments were carried out on a RQF-3 KinTek ChemicalQuench Flow apparatus. A solution of 100 nM protein and 300

SCHEME 1. Biochemical pathway of nucleotide incorporation of Pol �.DNA binding creates a binary complex with an equilibrium dissociation con-stant of KD(DNA) (step 1). The binary Pol � binding to dNTP forms the ternarycomplex with an equilibrium dissociation constant of Kd(dNTP) (step 2). Aglobal rearrangement of the ternary structure in which the fingers domainmoves closer to the DNA aligns the active site for polymerization at a rate ofkpol (step 3). This rearranged species of Pol � is denoted as �*. The fingersdomain reopens and releases the extended DNA product at a rate of kss,which is the rate-limiting step (step 4) (36).

TABLE 1One base pair gap DNA substrate used in this studyThe bold, underlined base is the templating base. The X marks the Dabcyl residue.

DNA sequence Sub strate

5�-GCCTCGCAGCCGGCTGATGCGCGTCGGTCGATCCAATGCCGTCC T:3�OHa

CGGAGCGTCGGCCGACTACGCGGCAGCCAGCTAGGTTACGGCAGG-5�5�-GCCTCGCAGCCGGCAGATGCGC GTCGGTCGATCCAATGCCGTCC T(�8)D:3�OHb

CGGAGCGTCGGCCGXCTACGCGGCAGCCAGCTAGGTTACGGCAGG-5�5�-GCCTCGCAGCCGGCAGATGCGCH GTCGGTCGATCCAATGCCGTCC T:3�Hc

CGGAGCGTCGGCCGTCTACGCGGCAGCCAGCTAGGTTACGGCAGG-5�5�-GCCTCGCAGCCGGCAGATGCGCH GTCGGTCGATCCAATGCCGTCC T(�8)D:3�Hd

CGGAGCGTCGGCCGXCTACGCGGCAGCCAGCTAGGTTACGGCAGG-5�a T:3�OH is the extendable DNA substrate without a Dabcyl residue.b T(�8)D:3�OH is extendable DNA substrate with a Dabcyl residue at the �8 position from the templating base.c T:3�H is non-extendable DNA substrate without a Dabcyl residue.d T(�8)D:3�H is non-extendable DNA substrate with a Dabcyl residue at the �8 position from the templating base.

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

16542 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 23 • JUNE 6, 2014

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 3: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

nM 32P-labeled, one-base gapped DNA (Table 1) were mixedwith 100 �M dCTP and 10 mM MgCl2. All mixtures containedreaction buffer E (50 mM Tris-HCl, pH 8, 100 mM NaCl, and10% glycerol) and were allowed to react for 0.2 to 3 s at 37 °C.Reactions were quenched with 0.5 M EDTA and 90% form-amide sequencing dye. Radioactive products were separatedon a 20% denaturing polyacrylamide gel, observed on aStorm 860 PhosphorImager, and quantified based on n andn�1 products using ImageQuant software. The data werefitted to the following biphasic burst equation using Prism 6GraphPad software.

�Product� � �E�app� kobs2

�kobs � kss2

� �1 � e�(kobs � kss)t ��kobskss)

�kobs � ksst� (Eq. 1)

Unlabeled WT Pol � was assayed DNA substrate T:3�OH.IAEDANS-labeled Pol � (Pol �-AEDANS) was studied onT(�8)D:3�OH (Table 1).

Enzyme Titration—To determine optimal single turnoverconditions, various concentrations of Pol �-AEDANS (500 to2000 nM) were reacted with 100 nM T(�8)D:3�OH and 100 �M

dCTP for 0.03 to 10 s. Reactions were quenched with 0.5 M

EDTA and 90% formamide sequencing dye. Radioactive prod-ucts were separated on a 20% denaturing polyacrylamide gel asdescribed above. The data were fitted to the following singleexponential equation using Prism 6 GraphPad software.

�Product� � A�1 � e�kobst (Eq. 2)

Here, kobs is the observed rate at each concentration of enzyme.

Single-turnover Experiments—Pol �-AEDANS and radiola-beled T(�6)D:3�OH were assayed on the KinTek apparatus todetermine kpol and Kd(dNTP) at 37 °C. Correct nucleotide(dCTP) was titrated from 0.5 to 200 �M over a range of 0.03 to10 s with 500 nM Pol � and 50 nM DNA. Radioactive productswere separated on a denaturing polyacrylamide gel and quanti-fied as described above. Using Prism 6, the data were fitted to asingle exponential equation, where kobs is the observed rateconstant at each concentration of dNTP. The rates (kobs) areplotted against concentration of dNTP and fitted to the hyper-bolic equation,

kobs �kpol�dNTP�

Kd�dNTP � �dNTP�(Eq. 3)

kpol is the maximum rate of polymerization and Kd(dNTP) is theequilibrium dissociation constant for the incoming dNTP.

Circular Dichroism—The secondary structural characteris-tics of unlabeled Pol �, Pol �-AEDANS, and Cys-178 were ana-lyzed by circular dichroism as previously described (24). Theellipicity of 3 �M labeled protein solution was normalized to 3�M unlabeled Pol �.

Fluorescence Emission Scan—The fluorescence of Pol �-AEDANS (600 nM) was observed at room temperature on thePhoton Technology International spectrofluorometer in BufferF (50 mM Tris-HCl, pH 7, 10 mM MgCl2, and 1 mM EDTA). Thesample was excited at 336 nm and an emission scan was per-formed from 380 to 650 nM. The DNA substrate T(�8)D:3�Hwas added to the protein mixture and scanned, followed by theaddition of 100 �M dCTP (correct) or 500 �M dATP (incorrect).

Stopped-flow Fluorescence—In all experiments, the samplewas excited at 336 nm, the emissions were filtered with a400-nm filter, and the voltage was fixed at 400 V. Pol �-AEDANS was assayed along with Dabcyl-labeled DNA sub-strate (see figure legends for specific DNA used) in the stopped-flow SX-19 (Applied Photophysics) at 37 °C. Under single turn-over conditions, 500 nM Pol � was co-incubated with 200 nM

T(�8)D:3�H or 100 nM T(�8)D:3�OH in buffer E and then rap-idly mixed in the stopped-flow instrument with various con-centrations of dNTP as detailed in the Figure legends. In someexperiments, MgCl2 was exchanged for CaCl2. Data collectionbegan as soon as mixing occurred (pre-trigger setting). Fluores-cent signal artifacts from the flow and mixing of the solutionswere removed from the traces (0 – 0.0308 s) based on the results

FIGURE 1. Crystal structure of human DNA polymerase � (Protein Data Bank codes 3ISB and 4KLE). A, Pol � contains four domains (3ISB): the 8-kDa domain(yellow), thumb domain (blue), palm domain (green), and the fingers domain (red). B, a rotation of 180° of the crystal structure reveals the distance between theIAEDANS and Dabcyl to be 43.1 Å in the open, binary complex (3ISB). Residue 303 on the fingers domain is labeled with IAEDANS and the DNA is labeled with Dabcyl-T8 bases away from the gap (�8). C, in the closed, ternary complex, IAEDANS and Dabcyl-T have a calculated distance (The PyMOL Molecular Graphics System, Version1.2r3pre, Schrodinger, LLC.) of 33.7 Å (4KLE).

TABLE 2Activity of modified Pol �

Pol �Residue

178Residue

239Residue

267 kobsa

s�1

WT C C C 13 2178S/239S Ser Ser C Highly unstable178S/267S Ser C Ser No burst239S/267S C Ser Ser 12 2178S/239S/267S Ser Ser Ser No burst

a Reactions were carried out with 100 �M Pol � and 300 �M T:3�OH using 100 �MdCTP as described under �Experimental Procedures.�

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

JUNE 6, 2014 • VOLUME 289 • NUMBER 23 JOURNAL OF BIOLOGICAL CHEMISTRY 16543

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 4: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

from standardized reactions (25). A logarithmic data collectionof 500 time points over 10 s was evaluated for each mixing eventwith five traces per dNTP concentration that were averagedtogether.

KinTek Explorer Fitting—Rate constants were determined byKinTek Explorer Professional Version 3 (26, 27). Raw data wereimported into the program as a concentration series. The datawere plotted a logarithmic time scale and plot residuals wereused to ensure a precise fit. In addition, concentration seriesoffset (addition/subtraction) was added to the simulation. Theobservables were noted as,

a � �ED � b � EDN � c � END. . . � offset_1a

(Eq. 4)

where the observables reflect the presence of protein [E]. TheKd(dNTP) was confirmed using the following equation,

Kd�dnTP �k �

k �

(Eq. 5)

where k� and k� correspond to the rate constants determinedby KinTek Explorer.

A nonlinear regression equation was used to initially esti-mate the fit of the raw stopped-flow data. In instances where thenon-covalent step was absent (Fig. 8A), the analytic functionf(t) � a1 exp(�b1 t) � c, was used. When the non-covalentstep was present (Figs. 5, A and B, and 8B), the function f(t) �a1 exp(�b1 t) � a2 exp(�b2 t) � c was used. Data forextendable DNA (Fig. 7A) was estimated to a quadruple expo-nential equation. The data were fit by KinTek Explorer to thebest possible fit and constrained by standard error and chemicalquench data (Fig. 3) (26). The non-covalent step was furtherdefined by one-dimensional FitSpace calculation of 5.5–25.5s�1 (27).

RESULTS

IAEDANS-labeled Pol � and Dabcyl-labeled DNA—Wedesigned a FRET-based approach for monitoring fingers move-ment of Pol �, based on previous work with DNA polymerase I(23). We chose to label Pol � with the thiol reactive IAEDANSin the fingers domain, and to label the DNA template with Dab-cyl. Quenching of fluorescence will occur upon finger closure,as the fingers move toward the Dabcyl in the DNA, according tocrystal structures of the open binary and closed ternary com-plexes. Pol � contains three endogenous Cys residues at posi-tions 178, 239, and 267; none of which would be appropriate foraccurately reporting the fingers movement. An initial charac-terization of Pol � C239S and Pol � C267S, in which eitherCys-239 or Cys-267 were altered to Ser, or of the double mutant(C239S,C267S) indicated that altering these residues to Serwould not affect the overall Pol � function (Table 2). However,alteration of Cys-178 to Ser affected activity (Table 2), and wethus elected not to alter this residue. Val-303, located on a smallloop in the fingers domain, was mutated to a Cys residue thatcould react with the IAEDANS (Fig. 1A). The placement of theDabcyl quencher on the DNA was carefully selected to main-tain an appropriate Forster distance of about 40 Å. A Dabcyl-Twas inserted eight bases downstream (�8) from the singlenucleotide gap, which provided an optimal distance for observ-ing FRET (Table 1 and Fig. 1, A and B).

FIGURE 2. Labeling protein constructs with IAEDANS. Fluorescence image isshown in A and Coomassie staining of the same 10% PAGE gel in B. Pol � V303C-AEDANS is shown in lanes 1 and 3; the Cys-178 Pol � protein is in lanes 2 and 4.Cys-178 Pol � protein contains a single Cys at position 178. Note that Cys-178 isnot fluorescent and is therefore not labeled with IAEDANS. C, circular dichroismspectra of unlabeled Pol �, Pol � V303C-AEDANS, and Cys-178. Data were nor-malized to unlabeled Pol �.

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

16544 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 23 • JUNE 6, 2014

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 5: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

The Labeled V303C Pol � Variant Has Kinetics PropertiesSimilar to WT Pol �—The variant of Pol � bearing three muta-tions (C239S, C267S, and V303C) termed V303C, was sub-cloned into a tagless pET28a expression vector and expressedin Escherichia coli. The purified protein was labeled withIAEDANS similar to previously published protocols (23, 28)and is outlined under “Experimental Procedures.” A controlprotein termed Cys-178, which harbors only the C239S andC267S alterations with the endogenous Cys-178 maintained,was expressed, purified, and labeled in the same manner. Anal-ysis of a 10% SDS-PAGE gel containing samples of both theIAEDANS-treated V303C and Cys-178 protein preparationsdetected either by UV or Coomassie staining suggests that thethiol reaction occurs at the Cys-303 position and that Cys-178is not labeled with IAEDANS (Fig. 2, A and B) presumablybecause it is buried. To further confirm protein structure, cir-cular dichroism spectroscopy suggests labeled protein prepara-tions retained similar secondary structure characteristics ofunlabeled WT Pol � (Fig. 2C).

Pol � V303C-AEDANS was assessed for pre-steady-stateburst activity using the extendable T(�8)D:3�OH substrate.The burst rate observed for Pol � V303C-AEDANS (14 s�1) issimilar to the burst rate of unlabeled Pol � (13 s�1) on non-labeled DNA (Fig. 3A and Table 2). This rapid rate of 14 s�1

suggests that alteration of Cys-239 and Cys-267 to Ser, as well asthe labeling with IAEDANS at position 303, did not affect theoverall rate of DNA synthesis catalyzed by Pol � and that theoverall rate-limiting step was still product release. We also per-formed experiments under single turnover conditions using aratio of 10:1 protein:DNA. These conditions were determinedempirically by incubating various concentrations of protein

with 100 nM DNA and 100 �M dNTP for various times (11). Asshown in Table 3 and Fig. 3, B and C, the kobs, kpol, and Kd(dNTP)

for Pol � V303C-AEDANS are similar to unlabeled Pol �(29). In combination, our results show that the Pol � V303C-AEDANS has kinetic properties similar to WT Pol � that areunaffected by the IAEDANS label.

FRET Is Observed in Steady-state with Pol � V303C-AEDANSand Dabcyl-labeled DNA—To determine whether we couldobserve a FRET signal with the Pol � V303C-AEDANS andDabcyl-labeled DNA, we characterized steady-state fluores-cence. Upon excitation of the Pol � V303C-AEDANS alone at336 nm, an emission scan reveals a peak at �490 nm (Fig. 4A,red). Upon addition of the non-extendable T(�8)D:3�H Dab-cyl-labeled DNA substrate we observed a quench in fluores-cence intensity, indicating the formation of a binary complex(Fig. 4A, blue). Upon addition of the correct dNTP (dCTP) weobserved greater fluorescence quenching, suggesting that Pol �is in a ternary complex in which the fingers domain movescloser to the Dabcyl in the DNA (Fig. 4A, green). Finally, whenan incorrect dNTP (dATP) is added to binary Pol � we observeless of a fluorescence quench compared with that of correctdNTP (Fig. 4A, black). The same experiment was repeated withnon-Dabcyl-labeled DNA T:3�H (Fig. 4B). There was littledecrease in the fluorescence intensity of Pol �-AEDANS uponaddition of T:3�H and both correct or incorrect dNTP, indicat-ing that the quenching seen in Fig. 4A is due to the IAEDANS/Dabcyl FRET pair coming within the Forster radii. These resultsdemonstrate that we are able to observe a FRET signal between thepolymerase and DNA in the presence of dNTPs.

Stopped-flow Fluorescence Reveals a Novel Non-covalent Stepin the Pol � Catalytic Pathway—To characterize movements ofthe fingers domain in the absence of phosphoryl transfer weperformed stopped-flow fluorescence experiments in whichthe correct dNTP was mixed with Pol � V303C-AEDANS pre-bound to the non-extendable T(�8)D:3�H DNA substrate (Fig.5A). In the presence of the correct nucleotide (dCTP), there is arapid decrease in fluorescence suggesting that during the first0.1 s, the fingers domain comes within close proximity of theDabcyl-labeled DNA, such that a quench is observed. Similartraces were observed with extendable T(�8)D:3�OH DNA in

FIGURE 3. Pol � V303C-AEDANS displays similar biphasic burst activity and single turnover kinetics as unlabeled Pol �. A, pre-steady-state kinetics of WT(Pol �) unlabeled Pol � (circles) and labeled Pol � V303C-AEDANS (squares) with one-base gapped DNA. Both unlabeled and labeled enzymes experience a fastpre-steady-state burst rate of 13 2 and 14 2 s�1, respectively. Labeled Pol � retains its fast polymerization rate. B, a single turnover plot of products formedversus time to determine the kobs for each dNTP concentration shown on the right. C, the kobs were plotted versus correct (dCTP) concentration to determine thekpol rate of 12 s�1 and a Kd(dNTP) of 1.4 �M. Inset shows concentrations up to 25 �M.

TABLE 3Determination of incorporation efficiency for Pol � using rapid chem-ical methodology

Protein Reactiona kobsb kpol

c Kd(dNTP)

s�1 �M

Pol � G-dCTP 13 2 8.9 0.3 (29) 1.7 0.3 (29)Pol � V303C-AEDANS G-dCTP 14 2 12.1 0.4 1.4 0.3

a All reactions used T(-8)D:3�OH and are reported with mean S.D.b Pre-steady-state reaction rate with 100 �M dCTP.c Reactions carried out under single turnover conditions.

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

JUNE 6, 2014 • VOLUME 289 • NUMBER 23 JOURNAL OF BIOLOGICAL CHEMISTRY 16545

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 6: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

the presence of a dCpCpp, a non-hydrolyzable analog of dCTP(Fig. 5B).

The data obtained with the T(�8)D:3�H DNA substrate andcorrect dNTP were initially fitted with Prism 6 to both single-and double-exponential equations. The residuals indicated thata double-exponential equation was the better fit (Fig. 6, A andB), suggesting that upon binding correct dNTP, these twophases indicate that there are at least two movements. We thenemployed KinTek Explorer to globally fit all of the stopped-flowfluorescence data (26). In Fig. 6C, we display an example of thefit of a trace using non-extendable DNA and 50 �M dCTP(residuals shown in red). The fit of the entire 10-s trace is shownin logarithmic scale along with an inset of data points collectedfrom 0 to 0.03 s, plotted in linear scale. These data fit to thesame kinetic scheme with similar rates as the 10-s trace. Ourdata are best described by a kinetic scheme in which ED � NºEDN represents the initial binding of the dCTP (Fig. 6C). Asubsequent step of EDN º END is a conformational change,followed by ENDº NDE, a proposed second conformationalchange.

Nucleotide Incorporation follows a Different Pathway for Cor-rect Versus Incorrect Nucleotide—To examine the role of thefingers domain in nucleotide selection, stopped-flow experi-ments were performed under single turnover conditions withT(�8)D:3�OH and either correct (dCTP) or incorrect (dATP ordTTP) substrates. For correct incorporation, we observed ini-tial quenching of fluorescence followed by a gradual recovery offluorescence (Fig. 7A). Chemistry is occurring due to the pres-ence of the 3�-OH and dCTP and the increase in fluorescence

could reflect chemistry or steps subsequent to chemistry.Stopped-flow traces for incorrect dNTP incorporation signifi-cantly differ from those with correct dNTP. There was nochange in fluorescence with an incorrect purine or pyrimidine,even when titrating up to 900 �M incorrect dNTPs (Fig. 7, Band C).

KinTek Explorer simulations suggest that the kinetics of cor-rect incorporation are best described by a 5-step kinetic modelfor Pol �-AEDANS with extendable T(�8)D:3�OH DNA (Fig.7A) in which Pol � proceeds through: step 2, dNTP binding(ED � Nº EDN); step 3, a conformational change after recog-nition of the correct dNTP (EDNº END); step 3.1, a secondconformational change (END º NDE); step 3.2, chemistry(NDEº EP); step 4, DNA dissociation and product release (EPº E � P). Due to the linear nature of the stopped-flow traceobtained with an incorrect dNTP (Fig. 7, B and C), we wereunable to fit the data to an exponential equation or to model thedata with KinTek Explorer.

Table 4 summarizes the estimated relative rates of the reac-tions in the presence of non-extendable and extendable DNAsubstrates. Parameters used to constrain our model includechemical quench data as well as 2-AP and stopped-flow ratesderived using intrinsic tryptophan residues for Pol � (16, 30,31). The rate constant k1 represents the dNTP binding event.Importantly, calculation of the Kd(dNTP), as described under“Experimental Procedures” (k�1/k�1), yielded a Kd(dNTP) of 1.9to 3 �M, which is within the range of previously publishedKd(dNTP) for Pol � (29, 32). We suggest that the k�2 rate con-stant represents the fingers movement toward the DNA. We

FIGURE 4. Steady-state fluorescence of Pol �-AEDANS in the presence of Dabcyl-DNA and dNTP. A, the fluorescence intensity of 600 �M Pol � V303C-AEDANS (red) is reduced when bound to 100 �M Dabcyl-DNA substrate T(�8)D:3�H (binary complex, blue). The addition of 100 �M correct dNTP (dCTP) oppositetemplate G results in a further reduction in fluorescence intensity (green) suggesting a reduction in distance between the IAEDANS/Dabcyl pair. Less of areduction in intensity was observed with incorporation of 500 �M incorrect dATP (black). B, steady-state fluorescence of Pol �-AEDANS (red) was repeated asdescribed above with non-Dabcyl-labeled substrate T:3�H (blue) and correct (green) or incorrect (black) dNTP. The minor quenching that is observed is likely dueto the presence of dNTP.

FIGURE 5. Stopped-flow fluorescence of Pol � V303C-AEDANS in the absence of chemistry demonstrates fingers closing. A, a solution of Pol � V303C-AEDANS and non-extendable Dabcyl-T DNA (T(�8)D:3�H) in a 2.5:1 protein to DNA ratio was rapidly mixed with increasing correct dCTP to give the finalconcentrations at the right. B, similar to the experiment in A, the dNTP substrate was replaced with the non-hydrolyzable dCpCpp analog and T(�8)D:3�OH wasused for stopped-flow fluorescence experiments. In all experiments, n � 5. KinTek Explorer fits are represented by the dashed line.

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

16546 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 23 • JUNE 6, 2014

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 7: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

base our suggestion on the observation of fingers domainmovement in the binary versus the ternary Pol � crystal struc-tures (8, 33), our previous fluorescence characterization of Pol

� as well as characterization of Pol �, and additional DNA poly-merases by others (16, 23, 30, 35). We propose that the k�3 rateconstant represents a second non-covalent step following clos-ing of the fingers, mainly because it occurs with both theextendable and non-extendable DNA substrates. The k�4 rateconstant is most likely phosphodiester bond formation,because it corresponds to the rate of chemistry, kpol, that wemeasure in rapid chemical quench flow assays. Furthermore,this rate constant is not present when the primer terminus isnot extendable (Table 3). The k�5 rate constant is the overallrate-limiting step of the kinetic pathway and is similar to thesteady-state rate previously measured by us and others (36, 37).This likely represents dissociation of Pol � from the DNA.Scheme 2 summarizes these proposed kinetic steps for Pol �.

Characterization of the Non-covalent Step with CatalyticallyInactive Ca2�—To further explore the non-covalent step (k�3),CaCl2 was used in place of MgCl2 in the stopped-flow experi-ments. Calcium has a larger ionic radius than Mg2� and doesnot support phosphoryl transfer (38, 39). In the presence ofnon-extendable DNA and CaCl2, we observed a fluorescencedecrease upon binding to the correct dCTP (Fig. 8A) that couldbe described by a two-step model using KinTek Explorer (steps2 and 3 of Scheme 2). In contrast, the fluorescence traceobserved with extendable DNA (Fig. 8B) was best described bya three-step model (steps 2, 3, and 3.1 of Scheme 2). Theseresults indicate that in the presence of extendable DNA, theinitial fingers domain movement and the non-covalent step areintact in the presence of CaCl2, but that the chemistry step andsubsequent product release did not occur due to the inability ofCaCl2 to support phosphodiester bond formation. However,the non-covalent step (step 3.1 of Scheme 2) was not observedwith CaCl2 and non-extendable DNA. This suggests that thenon-covalent step 3.1 requires the presence of a 3�-OH.

DISCUSSION

We have developed a FRET-based system for Pol �, whichallows monitoring of movements of the fingers domain duringDNA synthesis. Our results provide valuable insight into themovements of the fingers domain during catalysis. We observethat Pol � experiences at least two conformational changes thatprecede phosphodiester bond formation.

Incorporation of Correct Versus Incorrect Nucleotide by Pol �Is Different—In agreement with published work for Pol � as wellas the A-family polymerases (unlike Y-family polymerases (9,19, 23, 40, 41, 43– 45), we observe that the initial global move-ment of the fingers domain is not rate-limiting and this initialdomain movement likely plays a critical role in facilitatingpolymerization. Single molecule experiments have also sug-gested this to be the case for Sulfolobus solfataricus DNApolymerase B1 (46). Importantly, incorrect dNTP binding doesnot induce any conformational changes with either a pyrimi-dine or purine within the first 10 to 20 s of the reaction, usingFRET to monitor the fingers domain movements. Our results,therefore, suggest that dNTP discrimination occurs at groundstate dNTP binding at a point before the fingers have closed.Future fluorescence experiments with mutator variants of Pol �that map to a number of different regions of the protein (see forexample, Refs. 47–50) will likely provide mechanistic insights

FIGURE 6. Double-exponential equation analysis of stopped-flow datasuggests a complex kinetic pathway and a non-covalent step. A stopped-flow reaction containing 500 nM Pol � V303C-AEDANS along with 200 nM

T(�8)D:3�H were mixed with 50 �M correct dNTP (dCTP). The stopped-flowtrace (blue) was fitted to a single-exponential equation (A; green, residuals,inset) or a double-exponential equation (B; black, residuals, inset) using Prism6 (GraphPad, Inc.). C, a representative image of stopped-flow fluorescencetraces of Pol � V303C-AEDANS with T(�8)D:3�H mixed with 50 �M correctdCTP for 10 s globally fit to a three-step kinetic scheme using KinTek Explorer(red, residuals below) and plotted on a logarithmic time scale. The inset high-lights the stopped-flow trace from 0 to 0.03 s and plotted on a linear timescale. The kinetic scheme below the graph defines ED � Nº EDN as initialbinding of the dCTP. A subsequent step of EDNº END is a conformationalchange associated with the fingers closing and the 10 and 0.03 s (inset) tracesuggest a value of �100 s�1.

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

JUNE 6, 2014 • VOLUME 289 • NUMBER 23 JOURNAL OF BIOLOGICAL CHEMISTRY 16547

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 8: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

into the how fidelity is governed before closure of the fingers.Our previous work suggests that the hinge region plays a role insubstrate discrimination (51–53) suggesting that the hinge mayin some way “sense” the presence of correct versus incorrectdNTP during its initial contact with the fingers domain, androtate toward a closed complex only if correct dNTP is bound.

A Novel Non-covalent Step in the Pol � Kinetic Pathway—The determination of rates from stopped-flow experiments wassupported by chemical quench analysis. Our data, with all DNAsubstrates and metals, report a Kd(dNTP) of 1.9 –3 �M dCTP. Wemeasured the Kd(dNTP) for the labeled protein under singleturnover conditions and found it to be 1.4 �M (Table 3), whichis similar to k�1/k�1 determined with KinTek Explorer and isconfirmed by our own previously published chemical quenchdata of human Pol � on this particular substrate (T:3�OH-,Table 3, and Scheme 2, step 2) (29, 32). Step 3 corresponds to

the rapid fingers closing, which is �100 s�1 in the presence ofMg2�. We have constrained this step using previously pub-lished fast rate constants (16, 30, 31). Due to the complexity ofthe stopped-flow traces, we used KinTek Explorer to reveal anadditional step that immediately precedes chemistry (step 3.1)that occurs with an upper limit of 25 s�1 that has not beenpreviously characterized. This step is remarkably absent withthe non-extendable T(�8)D:3�H DNA substrate in the pres-ence of Ca2� (Fig. 8A, Table 4). The new results emerging fromthis work call for an expansion of Scheme 1 for the pathway ofnucleotide incorporation by Pol �, including a non-covalentstep that may facilitate chemistry as shown in Scheme 3.

A non-covalent step has not been identified in previous stud-ies with Pol �, although there is precedence for a non-covalentstep in the kinetic pathways of the Klenow fragment andKlentaq1 (23, 54, 55) as well as the Y-family DNA polymerase,Dpo4 (40). Step 3.1 (step 3.1, Scheme 2) is of particular interestas it suggests a further rearrangement after fingers closing, butjust prior to chemistry, which was not previously observed forPol � using 2-AP and Trp.

We further explored the non-covalent step 3.1 by replacingMg2� with Ca2�. Calcium is larger than Mg2� and generallycannot support phosphoryl transfer (38, 56). Pol � uses a two-metal ion mechanism (57, 58) for catalysis in which two metalions participate in the reaction and coordinate the catalytictriad of three aspartate residues, Asp-190, Asp-192, and Asp-256. The catalytic metal A (MgA) lowers the pKa of the primerterminus, leading to attack on the �-phosphate of the incomingdNTP substrate. Metal B coordinates the non-bridging oxygens

FIGURE 7. The fingers domain closes in the presence of correct dNTP, but not in the presence of incorrect dNTP. A, Pol � V303C-AEDANS and extendableDabcyl-T DNA (T(�8)D:3�OH) were mixed with increasing concentrations of correct dCTP in single turnover conditions. The stopped-flow fluorescence traceswere globally fit to a five-step model. Misincorporation of Pol � V303C-AEDANS was analyzed using stopped-flow fluorescence, similar to A with the exceptionof increasing incorrect concentrations of dATP (B) and dTTP (C). In all experiments, n � 5. KinTek Explorer fits are represented by the dashed line.

SCHEME 2. Biochemical pathway model for KinTek Explorer simulations.The binary complex of Pol � (E/DNA:ED) is in equilibrium (k�1/k�1) with thenucleotide (N) to form the ternary complex (EDN) and is represented by step2 in the overall biochemical pathway suggested in Scheme 3. After formationof the ternary complex (EDN), the fingers domain moves closer to the DNA(END), signified by the rate constant k2 (step 3). A second conformationalchange occurs in which there is a rearrangement of the active site (NDE),indicated by k3 (a novel step 3.1). This rearrangement facilitates chemistry(EP) k4 (step 3.2), followed by product release (step 4) k5.

TABLE 4Kinetic rate constants for the Pol � V303C-AEDANS catalytic cycle during nucleotide incorporationBased on Scheme 2 analyzed by KinTek Explorer.

Rateconstanta Modelb

T(�8)D:3�H �Mg2� c T(�8)D:3�OH �

dCpCppT(�8)D:3�OH �

Mg2�T(�8)D:3�H �

Ca2�T(�8)D:3�OH �

Ca2�

k�1‡ ED � N3 EDN 11.8 0.64 5.79 0.903 12.1 0.885 35.6 3.07 48.2 3.58k�1 EDN3 ED � N 29.5 132 43.7 36.4 67.7 91.6k�2 EDN3 END 97.8 18.8 166 22.2 121 27.0 116 11.4 224 26.9k�2 END3 EDN (0.01–3.5)d �4.02 10�10 (0.000730–2.84) (0.999–4.2) �7.94 10�7

k�3 END3 NDE 8.1 5.5 7.89 0.342 17.0 0.179 11.9 0.797k�3 NDE3 END (0.06–6.2)d 1.1 10�7 63 11.1 �4.50 10�7

k�4 NDE3 EP 8.92k�4 EP3 NDE 0k�5 EPE � P 1.28 0.165k�5‡ E�P3 EP 21.6 4.15

a Rate constants reported in s�1, except where indicated by ‡ (�M�1s�1).

b Model used in KinTek Explorer program where E represents enzyme; D represents DNA; N represents nucleotide.c The deviations from the rate constant shown are S.E. (26).d Values represent the range.

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

16548 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 23 • JUNE 6, 2014

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 9: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

on the dNTP substrate and as a result neutralizes the negativecharge that develops during chemistry (58, 59). The dataobserved with extendable DNA and CaCl2 were modeled to athree-step pathway whereby step 3.1 was unaffected. However,the stopped-flow data with non-extendable DNA and Ca2�

resulted in a poor fit to the same three-step model indicatingthat the non-covalent step is abolished under these conditions.This observation of the non-covalent step (step 3.1), takentogether with the non-hydrolyzable analog data and the obser-vation that step 3 (fingers closing) was similar with Ca2� andMg2�, may be a result of a rearrangement of the active site(Asp-256, Asp-190, and Asp-192) during the binding of metalA, as recently suggested (23, 34, 39, 42, 55). Other possibilitiesinclude local rearrangement of active site side chains as recentlysuggested (44).

In summary, we have developed a FRET system to study themovements of the fingers domain of Pol �. We have demon-strated that the fingers domain closes rapidly following correctdNTP selection, and observed a non-covalent step that occursjust prior to chemistry. We provide further evidence that thisconformational change is associated with binding of the cata-lytic metal into the polymerase active site. Moreover, our datashow that Pol � discriminates between correct and incorrectnucleotides in a different manner. Our work provides novelinsight into the mechanism of selection and fidelity by thehuman repair DNA polymerase �.

Acknowledgment—We thank Dr. Catherine Joyce for assistance withthe initial setup of this project.

REFERENCES1. Barnes, D. E., and Lindahl, T. (2004) Repair and genetic consequences of

endogenous DNA base damage in mammalian cells. Annu. Rev. Genet. 38,445– 476

2. Wiederhold, L., Leppard, J. B., Kedar, P., Karimi-Busheri, F., Rasouli-Nia,A., Weinfeld, M., Tomkinson, A. E., Izumi, T., Prasad, R., Wilson, S. H.,Mitra, S., and Hazra, T. K. (2004) AP endonuclease-independent DNAbase excision repair in human cells. Mol. Cell 15, 209 –220

3. Yamtich, J., and Sweasy, J. B. (2010) DNA polymerase family X: function,structure, and cellular roles. Biochim. Biophys. Acta 1804, 1136 –1150

4. Prasad, R., Beard, W. A., Strauss, P. R., and Wilson, S. H. (1998) HumanDNA polymerase � deoxyribose phosphate lyase. Substrate specificity andcatalytic mechanism. J. Biol. Chem. 273, 15263–15270

5. Matsumoto, Y., and Kim, K. (1995) Excision of deoxyribose phosphateresidues by DNA polymerase � during DNA repair. Science 269, 699 –702

6. Pelletier, H., Sawaya, M. R., Wolfle, W., Wilson, S. H., and Kraut, J. (1996)Crystal structures of human DNA polymerase beta complexed with DNA:implications for catalytic mechanism, processivity, and fidelity. Biochem-istry 35, 12742–12761

7. Sawaya, M. R., Pelletier, H., Kumar, A., Wilson, S. H., and Kraut, J. (1994)Crystal structure of rat DNA polymerase �: evidence for a common po-lymerase mechanism. Science 264, 1930 –1935

8. Sawaya, M. R., Prasad, R., Wilson, S. H., Kraut, J., and Pelletier, H. (1997)Crystal Structures of human DNA polymerase � complexed with gappedand nicked DNA: evidence for an induced fit mechanism. Biochemistry 36,11205–11215

9. Vande Berg, B. J., Beard, W. A., and Wilson, S. H. (2001) DNA structureand aspartate 276 influence nucleotide binding to human DNA polymer-ase �: implication for the identity of the rate-limiting conformationalchange. J. Biol. Chem. 276, 3408 –3416

10. Kunkel, T. A. (1985) The mutational specificity of DNA polymerase-�during in vitro DNA synthesis: production of frameshift, base substitution,and deletion mutations. J. Biol. Chem. 260, 5787–5796

11. Li, S.-X., Vaccaro, J. A., and Sweasy, J. B. (1999) Involvement of phenyl-alanine 272 of DNA polymerase � in discriminating between correct andincorrect deoxynucleoside triphosphates. Biochemistry 38, 4800 – 4808

12. Kosa, J. L., and Sweasy, J. B. (1999) The E249K mutator mutant of DNApolymerase � extends mispaired termini. J. Biol. Chem. 274, 35866 –35872

13. Shah, A. M., Conn, D. A., Li, S.-X., Capaldi, A., Jager, J., and Sweasy, J. B. (2001)A DNA polymerase � mutator mutant with reduced nucleotide discrimina-tion and increased protein stability. Biochemistry 40, 11372–11381

14. Murphy, D. L., Donigan, K. A., Jaeger, J., and Sweasy, J. B. (2012) TheE288K colon tumor variant of DNA polymerase � is a sequence specificmutator. Biochemistry 51, 5269 –5275

15. Opresko, P. L., Sweasy, J. B., and Eckert, K. A. (1998) The mutator form ofpolymerase � with amino acid substitution at tyrosine 265 in the hingeregion displays an increase in both base substitution and frame shift er-rors. Biochemistry 37, 2111–2119

16. Shah, A. M., Li, S.-X., Anderson, K. S., and Sweasy, J. B. (2001) Y265Hmutator mutant of DNA polymerase �: proper teometric alignment iscritical for fidelity. J. Biol. Chem. 276, 10824 –10831

FIGURE 8. The non-covalent step is dependent on the presence of a free primer 3�OH group and the catalytic Mg2�. A, Pol � V303C-AEDANS andnon-extendable Dabcyl-T DNA (T(�8)D:3�H) were mixed with increasing concentrations of dNTP (correct, dCTP) and CaCl2 was substituted for MgCl2. B, Pol �V303C-AEDANS was analyzed using stopped-flow fluorescence, similar to A with the exception of T(�8)D:3�OH, extendable DNA. In all experiments, n � 5.KinTek Explorer fits are represented by the dashed line.

SCHEME 3. Revised biochemical pathway of nucleotide incorporation ofPol � based on FRET data. The overall formation of the binary and ternarystates of Pol � are the same as described in Scheme 1. Additional steps occurafter the formation of the ternary complex to include the fingers domainclosing (�*, step 3), followed by a rearrangement at the active site (�‡, step 3.1)to set up for polymerization (step 3.2). The rate-limiting step remains theproduct release rate of kss (step 4) (36).

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

JUNE 6, 2014 • VOLUME 289 • NUMBER 23 JOURNAL OF BIOLOGICAL CHEMISTRY 16549

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 10: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

17. Balbo, P. B., Wang, E. C., and Tsai, M.-D. (2011) Kinetic mechanism ofactive site assembly and chemical catalysis of DNA polymerase �. Bio-chemistry 50, 9865–9875

18. Bakhtina, M., Lee, S., Wang, Y., Dunlap, C., Lamarche, B., and Tsai, M.-D.(2005) Use of viscogens, dNTP�S, and rhodium(III) as probes in stopped-flow experiments to obtain new evidence for the mechanism of catalysisby DNA polymerase. Biochemistry 44, 5177–5187

19. Zhong, X., Patel, S. S., Werneburg, B. G., and Tsai, M.-D. (1997) DNApolymerase �: multiple conformational changes in the mechanism of ca-talysis. Biochemistry 36, 11891–11900

20. Chagovetz, A. M., Sweasy, J. B., and Preston, B. D. (1997) Increased activityand fidelity of DNA polymerase � on single-nucleotide gapped DNA.J. Biol. Chem. 272, 27501–27504

21. Singhal, R. K., and Wilson, S. H. (1993) Short gap-filling synthesis by DNApolymerase � is processive. J. Biol. Chem. 268, 15906 –15911

22. Eckenroth, B. E., Towle-Weicksel, J. B., Sweasy, J. B., and Doublie, S. (2013)The E295K cancer variant of human polymerase � favors the mismatchconformational pathway during nucleotide selection. J. Biol. Chem. 288,34850 –34860

23. Joyce, C. M., Potapova, O., Delucia, A. M., Huang, X., Basu, V. P., andGrindley, N. D. (2008) Fingers-closing and other rapid conformationalchanges in DNA polymerase I (Klenow fragment) and their role in nucle-otide selectivity. Biochemistry 47, 6103– 6116

24. Murphy, D. L., Kosa, J., Jaeger, J., and Sweasy, J. B. (2008) The Asp285variant of DNA polymerase � extends mispaired primer termini via in-creased nucleotide binding. Biochemistry 47, 8048 – 8057

25. Peterman, B. F. (1979) Measurement of the dead time of a fluorescencestopped-flow instrument. Anal. Biochem. 93, 442– 444

26. Johnson, K. A., Simpson, Z. B., and Blom, T. (2009) Global kinetic ex-plorer: a new computer program for dynamic simulation and fitting ofkinetic data. Anal. Biochem. 387, 20 –29

27. Johnson, K. A., Simpson, Z. B., and Blom, T. (2009) FitSpace explorer: analgorithm to evaluate multidimensional parameter space in fitting kineticdata. Anal. Biochem. 387, 30 – 41

28. Perumal, S. K., Ren, W., Lee, T. H., and Benkovic, S. J. (2013) How aholoenzyme for DNA replication is formed. Proc. Natl. Acad. Sci. U.S.A.110, 99 –104

29. Klvana, M., Murphy, D. L., Jerabek, P., Goodman, M. F., Warshel, A.,Sweasy, J. B., and Florian, J. (2012) Catalytic effects of mutations of distantprotein residues in human DNA polymerase �: theory and experiment.Biochemistry 51, 8829 – 8843

30. Dunlap, C. A., and Tsai, M.-D. (2002) Use of 2-aminopurine and trypto-phan fluorescence as probes in kinetic analyses of DNA polymerase. Bio-chemistry 41, 11226 –11235

31. Bakhtina, M., Roettger, M. P., Kumar, S., and Tsai, M.-D. (2007) A unifiedkinetic mechanism applicable to multiple DNA polymerases. Biochemis-try 46, 5463–5472

32. Nemec, A. A., Murphy, D. L., Donigan, K. A., and Sweasy, J. B. (2014) TheS229L colon tumor-associated variant of DNA polymerase � induces cel-lular transformation as a result of decreased polymerization efficiency.J. Biol. Chem. 289, 13708 –13716

33. Pelletier, H., Sawaya, M. R., Kumar, A., Wilson, S. H., and Kraut, J. (1994)Structures of ternary complexes of rat DNA polymerase �, a DNA tem-plate-primer, and ddCTP. Science 264, 1891–1903

34. Nakamura, T., Zhao, Y., Yamagata, Y., Hua, Y. J., and Yang, W. (2012) Watch-ing DNA polymerase make a phosphodiester bond. Nature 487, 196–201

35. Li, Y., Korolev, S., and Waksman, G. (1998) Crystal structures of open andclosed forms of binary and ternary complexes of the large fragment ofThermus aquaticus DNA polymerase I: structural basis for nucleotideincorporation. EMBO J. 17, 7514 –7525

36. Murphy, D. L., Jaeger, J., and Sweasy, J. B. (2011) A triad interaction in thefingers subdomain of DNA polymerase � controls polymerase activity.J. Am. Chem. Soc. 133, 6279 – 6287

37. Ahn, J., Werneburg, B. G., and Tsai, M.-D. (1997) DNA polymerase �:structure-fidelity relationship from pre-steady-state kinetic analyses of allpossible correct and incorrect base pairs for wild type and R283A mutant.Biochemistry 36, 1100 –1107

38. Xia, S., Wang, M., Blaha, G., Konigsberg, W. H., and Wang, J. (2011) Structural

insights intocompletemetal ioncoordinationfromternarycomplexesofBfamilyRB69 DNA polymerase. Biochemistry 50, 9114–9124

39. Freudenthal,B.D.,Beard,W.A.,Shock,D.D.,andWilson,S.H. (2013)Observinga DNA polymerase choose right from wrong. Cell 154, 157–168

40. Xu, C., Maxwell, B. A., Brown, J. A., Zhang, L., and Suo, Z. (2009) Globalconformational dynamics of a Y-family DNA polymerase during catalysis.PLoS Biol. 7, e1000225

41. Yang, W. (2003) Damage repair DNA polymerases Y. Curr. Opin. Struct.Biol. 13, 23–30

42. Basu, R. S., and Murakami, K. S. (2013) Watching the bacteriophage N4RNA polymerase transcription by time-dependent soak-trigger-freeze x-ray crystallography. J. Biol. Chem. 288, 3305–3311

43. Sucato, C. A., Upton, T. G., Kashemirov, B. A., Osuna, J., Oertell, K., Beard,W. A., Wilson, S. H., Florian, J., Warshel, A., McKenna, C. E., and Good-man, M. F. (2008) DNA polymerase � fidelity: halomethylene-modifiedleaving groups in pre-steady-state kinetic analysis reveal differences at thechemical transition state. Biochemistry 47, 870 – 879

44. Rothwell, P. J., Mitaksov, V., and Waksman, G. (2005) Motions of the fingerssubdomain of Klentaq1 are fast and not rate limiting: implications for the molec-ular basis of fidelity in DNA polymerases. Mol. Cell 19, 345–355

45. Joyce, C. M., and Benkovic, S. J. (2004) DNA polymerase fidelity: kinetics,structure, and checkpoints. Biochemistry 43, 14317–14324

46. Maxwell, B. A., and Suo, Z. (2013) Single-molecule investigation of sub-strate binding kinetics and protein conformational dynamics of a B-familyreplicative DNA polymerase. J. Biol. Chem. 288, 11590 –11600

47. Starcevic, D., Dalal, S., and Sweasy, J. (2005) Hinge residue Ile260 of DNApolymerase � is important for enzyme activity and fidelity. Biochemistry44, 3775–3784

48. Maitra, M., Gudzelak, A., Jr., Li, S. X., Matsumoto, Y., Eckert, K. A., Jager,J., and Sweasy, J. B. (2002) Threonine 79 is a hinge residue that governs thefidelity of DNA polymerase � by helping to position the DNA within theactive site. J. Biol. Chem. 277, 35550 –35560

49. Lin, G. C., Jaeger, J., and Sweasy, J. B. (2007) Loop II of DNA polymerase �

is important for polymerization activity and fidelity. Nucleic Acids Res. 35,2924 –2935

50. Yamtich, J., Starcevic, D., Lauper, J., Smith, E., Shi, I., Rangarajan, S., Jaeger,J., and Sweasy, J. B. (2010) Hinge residue I174 is critical for proper dNTPselection by DNA polymerase �. Biochemistry 49, 2326 –2334

51. Lin, G. C., Jaeger, J., Eckert, K. A., and Sweasy, J. B. (2009) Loop II of DNApolymerase � is important for discrimination during substrate binding.DNA Repair 8, 182–189

52. Gridley, C. L., Rangarajan, S., Firbank, S., Dalal, S., Sweasy, J. B., and Jaeger,J. (2013) Structural changes in the hydrophobic hinge region adverselyaffect the activity and fidelity of the I260Q mutator DNA polymerase �.Biochemistry 52, 4422– 4432

53. Starcevic, D., Dalal, S., Jaeger, J., and Sweasy, J. B. (2005) The hydrophobichinge region of rat DNA polymerase � is critical for substrate bindingpocket geometry. J. Biol. Chem. 280, 28388 –28393

54. Rothwell, P. J., and Waksman, G. (2007) A pre-equilibrium before nucle-otide binding limits fingers subdomain closure by Klentaq1. J. Biol. Chem.282, 28884 –28892

55. Bermek, O., Grindley, N. D., and Joyce, C. M. (2011) Distinct roles of theactive-site Mg2� ligands, Asp-882 and Asp-705, of DNA polymerase I(Klenow fragment) during the prechemistry conformational transitions.J. Biol. Chem. 286, 3755–3766

56. Pelletier, H., and Sawaya, M. R. (1996) Characterization of the metal ionbinding helix-hairpin-helix motifs in human DNA polymerase � by x-raystructural analysis. Biochemistry 35, 12778 –12787

57. Brautigam, C. A., and Steitz, T. A. (1998) Structural and functional in-sights provided by crystal structures of DNA polymerases and their sub-strate complexes. Curr. Opin. Struct. Biol. 8, 54 – 63

58. Beese, L. S., and Steitz, T. A. (1991) Structural basis for the 3�-5� exonu-clease activity of Escherichia coli DNA polymerase I: a two metal ionmechanism. EMBO J. 10, 25–33

59. Batra, V. K., Beard, W. A., Shock, D. D., Krahn, J. M., Pedersen, L. C., andWilson, S. H. (2006) Magnesium-induced assembly of a complete DNApolymerase catalytic complex. Structure 14, 757–766

Pol � FRET Studies of Fingers Movement during Nucleotide Selection

16550 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 23 • JUNE 6, 2014

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 11: FluorescenceResonanceEnergyTransferStudiesofDNA Polymerase · Unlabeled WT Pol was assayed DNA substrate T:3 OH. IAEDANS-labeled Pol (Pol -AEDANS) was studied on T( 8)D:3 OH (Table

Anderson and Joann B. SweasyJamie B. Towle-Weicksel, Shibani Dalal, Christal D. Sohl, Sylvie Doublié, Karen S.

NON-COVALENT STEP DURING NUCLEOTIDE SELECTIONCRITICAL ROLE OF FINGERS DOMAIN MOVEMENTS AND A NOVEL

: THEβFluorescence Resonance Energy Transfer Studies of DNA Polymerase

doi: 10.1074/jbc.M114.561878 originally published online April 24, 20142014, 289:16541-16550.J. Biol. Chem. 

  10.1074/jbc.M114.561878Access 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

  http://www.jbc.org/content/289/23/16541.full.html#ref-list-1

This article cites 59 references, 20 of which can be accessed free at

by guest on June 22, 2020http://w

ww

.jbc.org/D

ownloaded from