Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 396(1): 1-9, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11716455

ABSTRACT

The recognition and removal of damaged bases in the genome is the province of a highly specialized assemblage of enzymes known as DNA glycosylases. In recent years, structural and mechanistic studies have rapidly moved forward such that in some cases, the high-resolution structures of all stable complexes along the reaction pathway are available. In parallel, advances in isotopic labeling of DNA have allowed the determination of a transition state structure of a DNA repair glycosylase using kinetic isotope effect methods. The use of stable substrate analogs and fluorescent probes have provided methods for real time measurement of the critical step of damaged base flipping. Taken together, these synergistic structural and chemical approaches have elevated our understanding of DNA repair enzymology to the level previously attained in only a select few enzymatic systems. This review summarizes recent studies of the paradigm enzyme, uracil DNA glycosylase, and discusses future areas for investigation in this field.


Subject(s)
DNA Glycosylases , DNA Repair , N-Glycosyl Hydrolases/metabolism , Uracil/metabolism , Catalysis , DNA/chemistry , DNA/metabolism , DNA Damage , Deoxyribonucleosides/chemistry , Models, Genetic , Uracil-DNA Glycosidase
2.
J Biol Chem ; 276(45): 42347-54, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11551943

ABSTRACT

Base flipping is a highly conserved process by which enzymes swivel an entire nucleotide from the DNA base stack into their active site pockets. Uracil DNA glycosylase (UDG) is a paradigm enzyme that uses a base flipping mechanism to catalyze the hydrolysis of the N-glycosidic bond of 2'-deoxyuridine (2'-dUrd) in DNA as the first step in uracil base excision repair. Flipping of 2'-dUrd by UDG has been proposed to follow a "pushing" mechanism in which a completely conserved leucine side chain (Leu-191) is inserted into the DNA minor groove to expel the uracil. Here we report a novel implementation of the "chemical rescue" approach to show that the weak binding affinity and low catalytic activity of L191A or L191G can be completely or partially restored by substitution of a pyrene (Y) nucleotide wedge on the DNA strand opposite to the uracil base (U/A to U/Y). These results indicate that pyrene acts both as a wedge to push the uracil from the base stack in the free DNA and as a "plug" to hinder its reinsertion after base flipping. Pyrene rescue should serve as a useful and novel tool to diagnose the functional roles of other amino acid side chains involved in base flipping.


Subject(s)
DNA Glycosylases , DNA/chemistry , N-Glycosyl Hydrolases/chemistry , Pyrenes/pharmacology , Catalysis , DNA/metabolism , N-Glycosyl Hydrolases/metabolism , Oxidation-Reduction , Peroxides/pharmacology , Uracil/chemistry , Uracil-DNA Glycosidase
3.
Biochemistry ; 40(25): 7710-9, 2001 Jun 26.
Article in English | MEDLINE | ID: mdl-11412125

ABSTRACT

The DNA repair enzyme uracil DNA glycosylase (UDG) is a powerful N-glycohydrolase that cleaves the glycosidic bond of deoxyuridine in DNA. We have investigated the role of substrate binding energy in catalysis by systematically dismantling the optimal substrate Ap(+1)UpA(-1)pA(-2) by replacing the nucleotides at the +1, -1, or -2 position with a tetrahydrofuran abasic site nucleotide (D), a 3-hydroxypropyl phosphodiester spacer (S), a phosphate monoester (p), or a hydroxyl group (h). Contrary to previous reports, the minimal substrate for UDG is 2'-deoxyuridine (hUh). UDG has a significant catalytic efficiency (CE) for hUh of 4 x 10(7) M(-1) [CE = (k(cat)/K(m))(1/k(non)), where k(non) is the rate of the spontaneous hydrolysis reaction of hUh at 25 degrees C]. Addition of +1 and -1 phosphate monoanions to form pUp increases k(cat)/K(m) by 45-fold compared to that of hUh. The k(cat)/K(m) for pUp, but not pU or Up, is found to decrease by 20-fold over the pH range of 6-9 with a pK(a) of 7.1, which is identical to the pK(a) values for deprotonation of the +1 and -1 phosphate groups determined by the pH dependence of the (31)P NMR chemical shifts. This pH dependence indicates that binding of the pUp tetraanion is disfavored, possibly due to unfavorable desolvation or electrostatic properties of the highly charged +1 and -1 phosphate groups. Addition of flexible hydroxypropyl groups to the +1 and -1 positions to make SpUpS increases k(cat)/K(m) by more than 10(5)-fold compared to that of hUh, which is a 20-fold greater effect than observed with rigid D substituents in these positions (i.e., DpUpD). The -2 phosphoester or nucleotide is found to increase the reactivity of trimer substrates with rigid furanose rings or nucleotides in the +1 and -1 positions by 1300-270000-fold (i.e., DpUpD --> DpUpDpA or ApUpA --> ApUpApA). In contrast, the -2 nucleotide provides only an 8-fold rate enhancement when appended to the substrate containing the more flexible +1 and -1 S substituents (SpUpS --> SpUpSpA). These context-dependent effects of a -2 nucleotide are interpreted in terms of a mechanism in which the binding energy of this "handle" is used drive the rigid +1 and -1 A or D substituents into their binding pockets, resulting in a net catalytic benefit of -4.3 to -7.5 kcal/mol. Taken together, these results systematically track how UDG uses distant site binding interactions to produce an overall four billion-fold increase in CE compared to that of the minimal substrate hUh.


Subject(s)
DNA Glycosylases , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/metabolism , Anions , Binding Sites , Catalysis , Deoxyuridine/chemistry , Deoxyuridine/metabolism , Energy Transfer , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Organophosphates/chemistry , Organophosphates/metabolism , Protein Binding , Substrate Specificity , Uracil-DNA Glycosidase
4.
Biochemistry ; 40(4): 957-67, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11170417

ABSTRACT

Abasic sites are highly mutagenic lesions in DNA that arise as intermediates in the excision repair of modified bases. These sites are generated by the action of damage-specific DNA glycosylases and are converted into downstream intermediates by the specific activity of apurinic/apyrimidinic endonucleases. Enzymes in both families have been observed in crystal structures to impose deformations on the abasic-site DNA, including DNA kinking and base flipping. On the basis of these apparent protein-induced deformations, we propose that altered conformation and dynamics of abasic sites may contribute to the specificity of these repair enzymes. Previously, measurements of the steady-state fluorescence of the adenine analogue 2-aminopurine (2AP) opposite an abasic site demonstrated that binding of divalent cations could induce a conformational change that increased the accessibility of 2AP to solute quenching [Stivers, J. T. (1998) Nucleic Acids Res. 26, 3837-44]. We have performed time-resolved fluorescence experiments to characterize the states involved in this conformational change. Interpretation of these studies is based on a recently developed model attributing the static and dynamic fluorescence quenching of 2AP in DNA to aromatic stacking and collisional interactions with neighboring bases, respectively (see the preceding paper in this issue). The time-resolved fluorescence results indicate that divalent cation binding shifts the equilibrium of the abasic site between two conformations: a "closed" state, characterized by short average fluorescence lifetime and complex decay kinetics, and an "open" state, characterized by monoexponential decay with lifetime approximately that of the free nucleoside. Because the lifetime and intensity decay kinetics of 2AP incorporated into DNA are sensitive primarily to collisional interactions with the neighboring bases, the absence of dynamic quenching in the open state strongly suggests that the fluorescent base is extrahelical in this conformation. Consistent with this interpretation, time-resolved quenching studies reveal that the open state is accessible to solute quenching by potassium iodide, but the closed state is not. Greater static quenching is observed in the abasic site when the fluorescent base is flanked by 5'- and 3'-thymines than in the context of 5'- and 3'-adenines, indicating that 2AP is more stacked with the neighboring bases in the former sequence. These results imply that the conformation of the abasic site varies in a sequence-dependent manner. Undamaged sequences in which the abasic site is replaced by thymine do not exhibit an open state and have different levels of both static and dynamic quenching than their damaged homologues. These differences in structure and dynamics may be significant determinants of the high specific affinity of repair enzymes for the abasic site.


Subject(s)
2-Aminopurine/chemistry , DNA Glycosylases , DNA Repair , DNA/chemistry , Escherichia coli Proteins , Nucleic Acid Conformation , Calcium/chemistry , Carbon-Oxygen Lyases/chemistry , Cations, Divalent/chemistry , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase , Deoxyribonuclease IV (Phage T4-Induced) , Escherichia coli/enzymology , Kinetics , N-Glycosyl Hydrolases/chemistry , Oligonucleotides/chemistry , Solutions , Spectrometry, Fluorescence/methods , Thermodynamics , Titrimetry , Uracil-DNA Glycosidase
5.
Biochemistry ; 39(46): 14054-64, 2000 Nov 21.
Article in English | MEDLINE | ID: mdl-11087352

ABSTRACT

The DNA repair enzyme uracil DNA glycosylase catalyzes the first step in the uracil base excision repair pathway, the hydrolytic cleavage of the N-glycosidic bond of deoxyuridine in DNA. Here we report kinetic isotope effect (KIE) measurements that have allowed the determination of the transition-state structure for this important reaction. The small primary (13)C KIE (=1.010 +/- 0.009) and the large secondary alpha-deuterium KIE (=1.201 +/- 0.021) indicate that (i) the glycosidic bond is essentially completely broken in the transition state and (ii) there is significant sp(2) character at the anomeric carbon. Large secondary beta-deuterium KIEs were observed when [2'R-(2)H] = 1.102 +/- 0.011 and [2'S-(2)H] = 1.106 +/- 0.010. The nearly equal and large magnitudes of the two stereospecific beta-deuterium KIEs indicate strong hyperconjugation between the elongated glycosidic bond and both of the C2'-H2' bonds. Geometric interpretation of these beta-deuterium KIEs indicates that the furanose ring adopts a mild 3'-exo sugar pucker in the transition state, as would be expected for maximal stabilization of an oxocarbenium ion. Taken together, these results strongly indicate that the reaction proceeds through a dissociative transition state, with complete dissociation of the uracil anion followed by addition of water. To our knowledge, this is the first transition-state structure determined for enzymatic cleavage of the glycosidic linkage in a pyrimidine deoxyribonucleotide.


Subject(s)
DNA Glycosylases , N-Glycosyl Hydrolases/chemistry , Uracil/chemistry , Anions , Binding Sites , Catalysis , Chromatography, High Pressure Liquid , DNA Repair , DNA, Single-Stranded/chemical synthesis , Deoxyuridine/chemistry , Deuterium , Glycosides/chemistry , Kinetics , Static Electricity , Substrate Specificity , Tritium/chemistry , Uracil-DNA Glycosidase
6.
Biochemistry ; 39(41): 12585-94, 2000 Oct 17.
Article in English | MEDLINE | ID: mdl-11027138

ABSTRACT

The DNA repair enzyme uracil DNA glycosylase (UDG) pinches the phosphodiester backbone of damaged DNA using the hydroxyl side chains of a conserved trio of serine residues, resulting in flipping of the deoxyuridine from the DNA helix into the enzyme active site. We have investigated the energetic role of these serine-phosphodiester interactions using the complementary approaches of crystallography, directed mutagenesis, and stereospecific phosphorothioate substitutions. A new crystal structure of UDG bound to 5'-HO-dUAAp-3' (which lacks the 5' phosphodiester group that interacts with the Ser88 pinching finger) shows that the glycosidic bond of dU has been cleaved, and that the enzyme has undergone the same specific clamping motion that brings key active site groups into position as previously observed in the structures of human UDG bound to large duplex DNA substrates. From this structure, it may be concluded that glycosidic bond cleavage and the induced fit conformational change in UDG can occur without the 5' pinching interaction. The S88A, S189A, and S192G "pinching" mutations exhibit 360-, 80-, and 21-fold damaging effects on k(cat)/K(m), respectively, while the S88A/S189A double mutant exhibits an 8200-fold damaging effect. A free energy analysis of the combined effects of nonbridging phosphorothioate substitution and mutation at these positions reveals the presence of a modest amount of strain energy between the compressed 5' and 3' phosphodiester groups flanking the bound uridine. Overall, these results indicate a role for these serine-phosphodiester interactions in uracil flipping and preorganization of the sugar ring into a reactive conformation. However, in contrast to a recent proposal [Parikh, S. S., et al. (2000) Proc Natl. Acad. Sci. 94, 5083], there is no evidence that conformational strain of the glycosidic bond induced by serine pinching plays a major role in the 10(12)-fold rate enhancement brought about by UDG.


Subject(s)
DNA Damage , DNA Glycosylases , DNA/chemistry , N-Glycosyl Hydrolases/chemistry , Organophosphates/chemistry , Serine/chemistry , Catalysis , Conserved Sequence/genetics , Crystallography, X-Ray , DNA Repair , Escherichia coli/enzymology , Humans , Kinetics , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/genetics , Serine/genetics , Stereoisomerism , Structure-Activity Relationship , Thionucleotides/chemistry , Uracil/chemistry , Uracil-DNA Glycosidase
7.
Biochemistry ; 39(43): 13241-50, 2000 Oct 31.
Article in English | MEDLINE | ID: mdl-11052677

ABSTRACT

Using off-resonance Raman spectroscopy, we have examined each complex along the catalytic pathway of the DNA repair enzyme uracil DNA glycosylase (UDG). The binding of undamaged DNA to UDG results in decreased intensity of the DNA Raman bands, which can be attributed to an increased level of base stacking, with little perturbation in the vibrational modes of the DNA backbone. A specific complex between UDG and duplex DNA containing 2'-beta-fluorodeoxyuridine shows similar increases in the level of DNA base stacking, but also a substrate-directed conformational change in UDG that is not observed with undamaged DNA, consistent with an induced-fit mechanism for damage site recognition. The similar increases in the level of DNA base stacking for the nonspecific and specific complexes suggest a common enzyme-induced distortion in the DNA, potentially DNA bending. The difference spectrum of the extrahelical uracil base in the substrate-analogue complexes reveals only a small electron density reorganization in the uracil ring for the ground state complex, but large 34 cm(-)(1) downshifts in the carbonyl normal modes. Thus, UDG activates the uracil ring in the ground state mainly through H bonds to its C=O groups, without destroying its quasi-aromaticity. This result is at variance with the conclusion from a recent crystal structure, in which the UDG active site significantly distorts the flipped-out pseudouridine analogue such that a change in hybridization at C1 occurs [Parikh, S. S., et al. (2000) Proc. Natl. Acad. Sci. USA 97, 5083]. The Raman vibrational signature of the bound uracil product differs significantly from that of free uracil at neutral pH, and indicates that the uracil is anionic. This is consistent with recent NMR results, which established that the enzyme stabilizes the uracil anion leaving group by 3.4 pK(a) units compared to aqueous solution, contributing significantly to catalysis. These observations are generally not apparent from the high-resolution crystal structures of UDG and its complexes with DNA; thus, Raman spectroscopy can provide unique and valuable insights into the nature of enzyme-DNA interactions.


Subject(s)
DNA Damage , DNA Glycosylases , DNA, Bacterial/chemistry , N-Glycosyl Hydrolases/chemistry , Base Composition , Catalysis , DNA Repair , DNA-Binding Proteins/chemistry , Escherichia coli/chemistry , Escherichia coli/genetics , Floxuridine/chemistry , Furans , Glycosides/chemistry , Hydrogen Bonding , Hydrogen-Ion Concentration , Macromolecular Substances , Nucleic Acid Conformation , Solvents , Spectrum Analysis, Raman , Uracil/chemistry , Uracil-DNA Glycosidase
8.
Biochemistry ; 39(39): 11865-75, 2000 Oct 03.
Article in English | MEDLINE | ID: mdl-11009598

ABSTRACT

Uracil DNA glycosylase (UDG) cleaves the glycosidic bond of deoxyuridine in DNA using a hydrolytic mechanism, with an overall catalytic rate enhancement of 10(12)-fold over the solution reaction. The nature of the enzyme-substrate interactions that lead to this large rate enhancement are key to understanding enzymatic DNA repair. Using (1)H and heteronuclear NMR spectroscopy, we have characterized one such interaction in the ternary product complex of Escherichia coli UDG, the short (2.7 A) H bond between His187 N(epsilon)(2) and uracil O2. The H bond proton is highly deshielded at 15.6 ppm, indicating a short N-O distance and exhibits a solvent exchange rate that is 400- and 10(5)-fold slower than free imidazole at pH 7.5 and pH 10, respectively. Heteronuclear NMR experiments at neutral pH show that this H bond involves the neutral imidazole form of His187 and the N1-O2 imidate form of uracil. The excellent correspondence of the pK(a) for the disappearance of the H bond (pK(a) = 6.3 +/- 0.1) with the previously determined pK(a) = 6.4 for the N1 proton of enzyme-bound uracil indicates that the H bond requires negative charge on uracil O2 [Drohat, A. C., and Stivers, J. T. (2000) J. Am. Chem. Soc. 122, 1840-1841]. Although the above characteristics suggest a short strong H bond, the D/H fractionation factor of phi = 1.0 is more typical of a normal H bond. This unexpected observation may reflect a large donor-acceptor pK(a) mismatch or the net result of two opposing effects on vibrational frequencies: decreased N-H bond stretching frequencies (phi < 1) and increased bending frequencies (phi > 1) relative to the O-H bonds of water. The role of this H bond in catalysis by UDG and several approaches to quantify the H bond energy are discussed.


Subject(s)
DNA Glycosylases , Escherichia coli/enzymology , Histidine/chemistry , N-Glycosyl Hydrolases/chemistry , Uracil/chemistry , Anions , Binding Sites , DNA Repair , Deuterium Oxide/chemistry , Hydrogen Bonding , Hydrogen-Ion Concentration , Imidazoles/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protons , Solvents , Uracil-DNA Glycosidase
9.
Biochemistry ; 39(18): 5561-72, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10820030

ABSTRACT

To probe the mechanism of the reversible DNA phosphodiester bond cleavage and religation mechanism of the type I topoisomerase from vaccinia virus, we have synthesized DNA substrates carrying a single nonbridging Rp- or Sp-phosphorothioate (Ps) modification at the scissile phosphodiester (Pd) bond. Analysis of the stereochemical outcome of the net cleavage and rejoining reaction established that the reaction proceeds with retention of configuration, as expected for a double-displacement mechanism. Single-turnover kinetic studies on irreversible strand cleavage using 18/24 mer suicide substrates showed thio effects (k(Pd)/k(Ps)) of 340- and 30-fold for the Rp-Ps and Sp-Ps stereoisomers, respectively, but approximately 10-fold smaller thio effects for the reverse single-turnover religation reaction (Rp-Ps = 30 and Sp-Ps = 3). As compared to the smaller suicide cleavage substrates, approach-to-equilibrium cleavage studies using 32/32 mer substrates showed 7-9-fold smaller thio effects on cleavage, similar effects on religation, and the same ratio of the Rp to Sp thio effect as the suicide cleavage reaction ( approximately 10). In general, thio effects of 2.4-7.2-fold on the cleavage equilibrium are observed for the wild-type and H265A enzymes, suggesting differences in the interactions of the enzyme with the nonbridging sulfur in the noncovalent and covalent complexes. Studies of the cleavage, religation, and approach-to-equilibrium reactions catalyzed by the H265A active site mutant revealed a stereoselective, 11-fold decrease in the Rp-thio effect on cleavage and religation as compared to the wild-type enzyme. This result suggests that His-265 interacts with the nonbridging pro-Rp oxygen in the transition state for cleavage and religation, consistent with the arrangement of this conserved residue in the crystal structure of the human topoisomerase-DNA complex. In general, the greatest effect of thio substitution and the H265A mutation is to destabilize the transition state, with smaller effects on substrate binding. The interaction of His-265 with the pro-Rp nonbridging oxygen is inconsistent with the proposal that this conserved residue acts as a general acid in the strand cleavage reaction.


Subject(s)
DNA Topoisomerases, Type I/chemistry , Stereoisomerism , Vaccinia virus/enzymology , DNA Topoisomerases, Type I/genetics , Humans , Kinetics , Mutation , Oligodeoxyribonucleotides/chemistry , Spectrometry, Fluorescence
10.
Biochemistry ; 39(19): 5630-41, 2000 May 16.
Article in English | MEDLINE | ID: mdl-10801313

ABSTRACT

Rev is an essential HIV-1 regulatory protein that binds the Rev responsive element (RRE) within the env gene of the HIV-1 RNA genome and is involved in transport of unspliced or partially spliced viral mRNA from the cell nucleus to the cytoplasm. Previous studies have shown that a short alpha-helical peptide derived from Rev (Rev 34-50), and a truncated form of the RRE sequence provide a useful in vitro system to study this interaction while still preserving the essential aspects of the native complex. We have selectively incorporated the fluorescent probe 2-aminopurine 2'-O-methylriboside (2-AP) into the RRE sequence in nonperturbing positions (A68 and U72) such that the binding of both Rev peptide and aminoglycoside ligands could be characterized directly by fluorescence methods. Rev peptide binding to the RRE-72AP variant resulted in a 2-fold fluorescence increase that provided a useful signal to monitor this binding interaction (K(D) = 20 +/- 7 nM). Using stopped-flow kinetic measurements, we have shown that specific Rev peptide binding occurs by a two-step process involving diffusion-controlled encounter, followed by isomerization of the RNA. Using the RRE-68AP and -72AP constructs, three classes of binding sites for the aminoglycoside neomycin were unambiguously detected. The first site is noninhibitory to Rev binding (K(D) = 0.24 +/- 0.040 microM), the second site inhibited Rev binding in a competitive fashion (K(D) = 1. 8 +/- 0.8 microM), and the third much weaker site (or sites) is attributed to nonspecific binding (K(D) >/= 40 microM). Complementary NMR measurements have shown that neomycin forms both a specific binary complex with RRE and a specific ternary complex with RRE and Rev. NMR data further suggest that neomycin occupies a similar high-affinity binding site in both the binary and ternary complexes, and that this site is located in the lower stem region of RRE.


Subject(s)
Framycetin/metabolism , Gene Products, rev/genetics , Gene Products, rev/metabolism , HIV-1/genetics , HIV-1/metabolism , Peptide Fragments/metabolism , Response Elements , 2-Aminopurine/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/metabolism , Binding, Competitive/genetics , Gene Products, rev/antagonists & inhibitors , Kinetics , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Protein Binding/genetics , Spectrometry, Fluorescence , Thermodynamics , rev Gene Products, Human Immunodeficiency Virus
11.
Biochemistry ; 38(37): 11876-86, 1999 Sep 14.
Article in English | MEDLINE | ID: mdl-10508390

ABSTRACT

The nature of the putative general acid His187 in the reaction catalyzed by Escherichia coli uracil DNA glycosylase (UDG) was investigated using X-ray crystallography and NMR spectroscopy. The crystal structures of H187Q UDG, and its complex with uracil, have been solved at 1.40 and 1.60 A resolution, respectively. The structures are essentially identical to those of the wild-type enzyme, except that the side chain of Gln187 is turned away from the uracil base and cannot interact with uracil O2. This result provides a structural basis for the similar kinetic properties of the H187Q and H187A enzymes. The ionization state of His187 was directly addressed with (1)H-(15)N NMR experiments optimized for histidine ring spin systems, which established that His187 is neutral in the catalytically active state of the enzyme (pK(a) <5.5). These NMR experiments also show that His187 is held in the N(epsilon)()2-H tautomeric form, consistent with the crystallographic observation of a 2.9 A hydrogen bond from the backbone nitrogen of Ser189 to the ring N(delta)()1 of His187. The energetic cost of breaking this hydrogen bond may contribute significantly to the low pK(a) of His187. Thus, the traditional view that a cationic His187 donates a proton to uracil O2 is incorrect. Rather, we propose a concerted mechanism involving general base catalysis by Asp64 and electrophilic stabilization of the developing enolate on uracil O2 by a neutral His187.


Subject(s)
DNA Glycosylases , Escherichia coli/enzymology , Histidine/chemistry , N-Glycosyl Hydrolases/chemistry , Uracil/chemistry , Binding Sites/genetics , Carbon Isotopes , Catalysis , Crystallography, X-Ray , Enzyme Stability , Glutamine/genetics , Histidine/genetics , Histidine/metabolism , Hydrogen Bonding , Hydrogen-Ion Concentration , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Protons , Substrate Specificity , Uracil/metabolism , Uracil-DNA Glycosidase
12.
Biochemistry ; 38(37): 11866-75, 1999 Sep 14.
Article in English | MEDLINE | ID: mdl-10508389

ABSTRACT

Escherichia coli uracil DNA glycosylase (UDG) catalyzes the hydrolysis of premutagenic uracil bases in DNA by flipping the deoxyuridine from the DNA helix [Stivers, J. T., et al. (1999) Biochemistry 38, 952]. A general acid-base mechanism has been proposed whereby His187 facilitates leaving group departure by protonating the O2 of uracil and Asp64 activates a water molecule for nucleophilic attack at C1' of the deoxyribose. Detailed kinetic studies on the H187Q, H187A, and D64N mutant enzymes indicate that Asp64 and His187 stabilize the chemical transition state by 5.3 and 4.8 kcal/mol, respectively, with little effect on substrate or product binding. The pH dependence of k(cat) for wild-type and H187Q UDG indicates that an unprotonated group in the enzyme-substrate complex (pK(a) = 6.2 +/- 0.2) is required for catalysis. This unprotonated group has a small DeltaH of ionization (-0.4 +/- 1.7 kcal/mol) and is absent in the pH profile for D64N UDG, suggesting that it corresponds to the general base Asp64. The pH dependence of k(cat) for wild-type, H187Q, and D64N UDG shows no evidence for an essential protonated group over the pH range of 5.5-10. Hence, the pK(a) of His187 must be outside this pH range if it serves as an electrophilic catalyst. These results support a mechanism in which Asp64 serves as the general base and His187 acts as a neutral electrophile, stabilizing a developing negative charge on uracil O2 in the transition state. In the following paper of this issue we establish by crystallography and heteronuclear NMR spectroscopy that the imidazole of His187 is neutral during the catalytic cycle of UDG.


Subject(s)
DNA Glycosylases , Escherichia coli/enzymology , N-Glycosyl Hydrolases/chemistry , Asparagine/genetics , Aspartic Acid/genetics , Binding Sites/genetics , Catalysis , Deuterium Oxide/chemistry , Glutamine/genetics , Histidine/genetics , Hydrogen Bonding , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Solvents , Substrate Specificity/genetics , Uracil/metabolism , Uracil-DNA Glycosidase
13.
Biochemistry ; 38(38): 12343-57, 1999 Sep 21.
Article in English | MEDLINE | ID: mdl-10493802

ABSTRACT

Three arginine residues (Arg-11, Arg-39, Arg-61) are found at the active site of 4-oxalocrotonate tautomerase in the X-ray structure of the affinity-labeled enzyme [Taylor, A. B., Czerwinski, R. M., Johnson, R. M., Jr., Whitman, C. P., and Hackert, M. L. (1998) Biochemistry 37, 14692-14700]. The catalytic roles of these arginines were examined by mutagenesis, kinetic, and heteronuclear NMR studies. With a 1,6-dicarboxylate substrate (2-hydroxymuconate), the R61A mutation showed no kinetic effects, while the R11A mutation decreased k(cat) 88-fold and increased K(m) 8.6-fold, suggesting both binding and catalytic roles for Arg-11. With a 1-monocarboxylate substrate (2-hydroxy-2,4-pentadienoate), no kinetic effects of the R11A mutation were found, indicating that Arg-11 interacts with the 6-carboxylate of the substrate. The stereoselectivity of the R11A-catalyzed protonation at C-5 of the dicarboxylate substrate decreased, while the stereoselectivity of protonation at C-3 of the monocarboxylate substrate increased in comparison with wild-type 4-OT, indicating the importance of Arg-11 in properly orienting the dicarboxylate substrate by interacting with the charged 6-carboxylate group. With 2-hydroxymuconate, the R39A and R39Q mutations decreased k(cat) by 125- and 389-fold and increased K(m) by 1.5- and 2.6-fold, respectively, suggesting a largely catalytic role for Arg-39. The activity of the R11A/R39A double mutant was at least 10(4)-fold lower than that of the wild-type enzyme, indicating approximate additivity of the effects of the two arginine mutants on k(cat). For both R11A and R39Q, 2D (1)H-(15)N HSQC and 3D (1)H-(15)N NOESY-HSQC spectra showed chemical shift changes mainly near the mutated residues, indicating otherwise intact protein structures. The changes in the R39Q mutant were mainly in the beta-hairpin from residues 50 to 57 which covers the active site. HSQC titration of R11A with the substrate analogue cis, cis-muconate yielded a K(d) of 22 mM, 37-fold greater than the K(d) found with wild-type 4-OT (0.6 mM). With the R39Q mutant, cis, cis-muconate showed negative cooperativity in active site binding with two K(d) values, 3.5 and 29 mM. This observation together with the low K(m) of 2-hydroxymuconate (0.47 mM) suggests that only the tight binding sites function catalytically in the R39Q mutant. The (15)Nepsilon resonances of all six Arg residues of 4-OT were assigned, and the assignments of Arg-11, -39, and -61 were confirmed by mutagenesis. The binding of cis,cis-muconate to wild-type 4-OT upshifts Arg-11 Nepsilon (by 0.05 ppm) and downshifts Arg-39 Nepsilon (by 1.19 ppm), indicating differing electronic delocalizations in the guanidinium groups. A mechanism is proposed in which Arg-11 interacts with the 6-carboxylate of the substrate to facilitate both substrate binding and catalysis and Arg-39 interacts with the 1-carboxylate and the 2-keto group of the substrate to promote carbonyl polarization and catalysis, while Pro-1 transfers protons from C-3 to C-5. This mechanism, together with the effects of mutations of catalytic residues on k(cat), provides a quantitative explanation of the 10(7)-fold catalytic power of 4-OT. Despite its presence in the active site in the crystal structure of the affinity-labeled enzyme, Arg-61 does not play a significant role in either substrate binding or catalysis.


Subject(s)
Arginine/genetics , Isomerases/chemistry , Isomerases/genetics , Mutagenesis, Site-Directed , Alanine/genetics , Binding Sites/genetics , Catalysis , Glutamine/genetics , Isomerases/biosynthesis , Isomerases/metabolism , Kinetics , Models, Molecular , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemical synthesis , Recombinant Proteins/chemistry , Sorbic Acid/analogs & derivatives , Sorbic Acid/metabolism , Stereoisomerism , Titrimetry
14.
Biochemistry ; 38(38): 12358-66, 1999 Sep 21.
Article in English | MEDLINE | ID: mdl-10493803

ABSTRACT

The unusually low pK(a) value of the general base catalyst Pro-1 (pK(a) = 6.4) in 4-oxalocrotonate tautomerase (4-OT) has been ascribed to both a low dielectric constant at the active site and the proximity of the cationic residues Arg-11 and Arg-39 [Stivers, J. T., Abeygunawardana, C., Mildvan, A. S., Hajipour, G., and Whitman, C. P. (1996) Biochemistry 35, 814-823]. In addition, the pH-rate profiles in that study showed an unidentified protonated group essential for catalysis with a pK(a) of 9.0. To address these issues, the pK(a) values of the active site Pro-1 and lower limit pK(a) values of arginine residues were determined by direct (15)N NMR pH titrations. The pK(a) values of Pro-1 and of the essential acid group were determined independently from pH-rate profiles of the kinetic parameters of 4-OT in arginine mutants of 4-OT and compared with those of wild type. The chemical shifts of all of the Arg Nepsilon resonances in wild-type 4-OT and in the R11A and R39Q mutants were found to be independent of pH over the range 4.9-9.7, indicating that no arginine is responsible for the kinetically determined pK(a) of 9.0 for an acidic group in free 4-OT. With the R11A mutant, where k(cat)/K(m) was reduced by a factor of 10(2.9), the pK(a) of Pro-1 was not significantly altered from that of the wild-type enzyme (pK(a) = 6.4 +/- 0.2) as revealed by both direct (15)N NMR titration (pK(a) = 6.3 +/- 0.1) and the pH dependence of k(cat)/K(m) (pK(a) = 6.4 +/- 0.2). The pH-rate profiles of both k(cat)/K(m) and k(cat) for the reaction of the R11A mutant with the dicarboxylate substrate, 2-hydroxymuconate, showed humps, i.e., sharply defined maxima followed by nonzero plateaus. The humps disappeared in the reaction with the monocarboxylate substrate, 2-hydroxy-2,4-pentadienoate, indicating that, unlike the wild-type enzyme which reacts only with the dianionic form of the dicarboxylic substrate, the R11A mutant reacts with both the 6-COOH and 6-COO(-) forms, with the 6-COOH form being 12-fold more active. This reversal in the preferred ionization state of the 6-carboxyl group of the substrate that occurs upon mutation of Arg-11 to Ala provides strong evidence that Arg-11 interacts with the 6-carboxylate of the substrate. In the R39Q mutant, where k(cat)/K(m) was reduced by a factor of 10(3), the kinetically determined pK(a) value for Pro-1 was 4.6 +/- 0.2, while the ionization of Pro-1 showed negative cooperativity with an apparent pK(a) of 7.1 +/- 0.1 determined by 1D (15)N NMR. From the Hill coefficient of 0.54, it can be shown that the apparent pK(a) value of 7.1 could result most simply from the averaging of two limiting pK(a) values of 4.6 and 8.2. Mutation of Arg-39, by altering the structure of the beta-hairpin which covers the active site, could result in an increase in the solvent exposure of Pro-1, raising its upper limit pK(a) value to 8.2. In the R39A mutant, the kinetically determined pK(a) of Pro-1 was also low, 5.0 +/- 0.2, indicating that in both the R39Q and R39A mutants, only the sites with low pK(a) values were kinetically operative. With the fully active R61A mutant, the kinetically determined pK(a) of Pro-1 (pK(a) = 6.5 +/- 0.2) agreed with that of wild-type 4-OT. It is concluded that the unusually low pK(a) of Pro-1 shows little contribution from electrostatic effects of the nearby cationic Arg-11, Arg-39, and Arg-61 residues but results primarily from a site of low local dielectric constant.


Subject(s)
Arginine/genetics , Arginine/metabolism , Isomerases/genetics , Isomerases/metabolism , Mutagenesis, Site-Directed , Alanine/genetics , Binding Sites/genetics , Catalysis , Glutamine/metabolism , Hydrogen-Ion Concentration , Kinetics , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Proline/metabolism , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Substrate Specificity , Titrimetry
15.
Proteins ; 35(1): 13-24, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10090282

ABSTRACT

The DNA repair enzyme uracil DNA glycosylase (UDG) catalyzes the hydrolysis of premutagenic uracil residues from single-stranded or duplex DNA, producing free uracil and abasic DNA. Here we report the high-resolution crystal structures of free UDG from Escherichia coli strain B (1.60 A), its complex with uracil (1.50 A), and a second active-site complex with glycerol (1.43 A). These represent the first high-resolution structures of a prokaryotic UDG to be reported. The overall structure of the E. coli enzyme is more similar to the human UDG than the herpes virus enzyme. Significant differences between the bacterial and viral structures are seen in the side-chain positions of the putative general-acid (His187) and base (Asp64), similar to differences previously observed between the viral and human enzymes. In general, the active-site loop that contains His187 appears preorganized in comparison with the viral and human enzymes, requiring smaller substrate-induced conformational changes to bring active-site groups into catalytic position. These structural differences may be related to the large differences in the mechanism of uracil recognition used by the E. coli and viral enzymes. The pH dependence of k(cat) for wild-type UDG and the D64N and H187Q mutant enzymes is consistent with general-base catalysis by Asp64, but provides no evidence for a general-acid catalyst. The catalytic mechanism of UDG is critically discussed with respect to these results.


Subject(s)
DNA Glycosylases , Escherichia coli/enzymology , Glycerol/chemistry , N-Glycosyl Hydrolases/chemistry , Uracil/chemistry , Amino Acid Sequence , Catalysis , Crystallography, X-Ray , Humans , Hydrolysis , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Uracil-DNA Glycosidase
16.
Biochemistry ; 38(3): 952-63, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9893991

ABSTRACT

The DNA repair enzyme uracil DNA glycosylase (UDG) catalyzes hydrolytic cleavage of the N-glycosidic bond of premutagenic uracil residues in DNA by flipping the uracil base from the DNA helix. The mechanism of base flipping and the role this step plays in site-specific DNA binding and catalysis by enzymes are largely unknown. The thermodynamics and kinetics of DNA binding and uracil flipping by UDG have been studied in the absence of glycosidic bond cleavage using substrate analogues containing the 2'-alpha and 2'-beta fluorine isomers of 2'-fluoro-2'-deoxyuridine (Ubeta, Ualpha) positioned adjacent to a fluorescent nucleotide reporter group 2-aminopurine (2-AP). Activity measurements show that DNA containing a Ubeta or Ualpha nucleotide is a 10(7)-fold slower substrate for UDG (t1/2 approximately 20 h), which allows measurements of DNA binding and base flipping in the absence of glycosidic bond cleavage. When UDG binds these analogues, but not other DNA molecules, a 4-8-fold 2-AP fluorescence enhancement is observed, as expected for a decrease in 2-AP base stacking resulting from enzymatic flipping of the adjacent uracil. Thermodynamic measurements show that UDG forms weak nonspecific complexes with dsDNA (KDns = 1.5 microM) and binds approximately 25-fold more tightly to Ubeta containing dsDNA (KDapp approximately 50 nM). Thus, base flipping contributes less than approximately 2 kcal/mol to the free energy of binding and is not a major component of the >10(6)-fold catalytic specificity of UDG. Kinetic studies at 25 degrees C show that site-specific binding occurs by a two-step mechanism. The first step (E + S left and right arrow ES) involves the diffusion-controlled binding of UDG to form a weak nonspecific complex with the DNA (KD approximately 1.5-3 microM). The second step (ES left and right arrow E'F) involves a rapid step leading to reversible uracil flipping (kmax approximately 1200 s-1). This step is followed closely by a conformational change in UDG that was monitored by the quenching of tryptophan fluorescence. The results provide evidence for an enzyme-assisted mechanism for uracil flipping and exclude a passive mechanism in which the enzyme traps a transient extrahelical base in the free substrate. The data suggest that the duplex structure of the DNA is locally destabilized before the base-flipping step, thereby facilitating extrusion of the uracil. Thus, base flipping contributes little to the free energy of DNA binding but contributes greatly to specificity through an induced-fit mechanism.


Subject(s)
DNA Damage , DNA Glycosylases , DNA Repair , Escherichia coli/enzymology , N-Glycosyl Hydrolases/chemistry , Uracil/chemistry , Binding Sites , Escherichia coli/genetics , Floxuridine/analogs & derivatives , Floxuridine/chemistry , Kinetics , Macromolecular Substances , Spectrometry, Fluorescence , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity , Thermodynamics , Uracil-DNA Glycosidase
17.
Nucleic Acids Res ; 26(16): 3837-44, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9685503

ABSTRACT

Metal-ion and sequence dependent changes in the stacking interactions of bases surrounding abasic (AB) sites in 10 different DNA duplexes were examined by incorporating the fluorescent nucleotide probe 2-aminopurine (2-AP), opposite to the site (AB-APopp) or adjacent to the site (AB-APadj) on either strand. A detailed study of the fluorescence emission and excitation spectra of these AB duplexes and their corresponding parent duplexes indicates that AB-APoppis significantly less stacked than 2-AP in the corresponding normal duplex. In general, AB-APadjon the AB strand is stacked, but AB-APadjon the opposite strand shows destabilized stacking interactions. The results also indicate that divalent cation binding to the AB duplexes contributes to destabilizaton of the base stacking interactions of AB-APopp, but has little or no effect on the stacking interactions of AB-APadj. Consistent with these results, the fluorescence of AB-APoppis 18-30-fold more sensitive to an externally added quenching agent than the parent normal duplex. When uracil DNA glycosylase binds to AB-APoppin the presence of 2.5 mM MgCl2, a 3-fold decrease in fluorescence is observed ( K d = 400 +/- 90 nM) indicating that the unstacked 2-APoppbecomes more stacked upon binding. On the basis of these fluorescence studies a model for the local base stacking interactions at these AB sites is proposed.


Subject(s)
2-Aminopurine , DNA Glycosylases , DNA/chemistry , DNA/metabolism , Fluorescent Dyes , Acrylamide , Acrylamides , Base Sequence , Binding Sites , Cations, Divalent , Kinetics , Magnesium Chloride , N-Glycosyl Hydrolases/metabolism , Nucleic Acid Conformation , Spectrometry, Fluorescence , Temperature , Uracil-DNA Glycosidase
18.
Biochemistry ; 36(17): 5212-22, 1997 Apr 29.
Article in English | MEDLINE | ID: mdl-9136883

ABSTRACT

The Vaccinia type I topoisomerase catalyzes site-specific DNA strand cleavage and religation by forming a transient phosphotyrosyl linkage between the DNA and Tyr-274, resulting in the release of DNA supercoils. For type I topoisomerases, two mechanisms have been proposed for supercoil release: (I) a coupled mechanism termed strand passage, in which a single supercoil is removed per cleavage/religation cycle, resulting in multiple topoisomer intermediates and late product formation, or (2) an uncoupled mechanism termed free rotation, where multiple supercoils are removed per cleavage/religation cycle, resulting in few intermediates and early product formation. To determine the mechanism, single-turnover experiments were done with supercoiled plasmid DNA under conditions in which the topoisomerase cleaves predominantly at a single site per DNA molecule. The concentrations of supercoiled substrate, intermediate topoisomers, and relaxed product vs time were measured by fluorescence imaging, and the rate constants for their interconversion were determined by kinetic simulation. Few intermediates and early product formation were observed. From these data, the rate constants for cleavage (0.3 s(-1)), religation (4 s(-1)), and the cleavage equilibrium constant on the enzyme (0.075) at 22 degrees C are in reasonable agreement with those obtained with small oligonucleotide substrates, while the rotation rate of the cleaved DNA strand is fast (approximately 20 rotations/s). Thus, the average number of supercoils removed for each cleavage event greatly exceeds unity (delta n = 5) and depends on kinetic competition between religation and supercoil release, establishing a free rotation mechanism. This free rotation mechanism for a type I topoisomerase differs from the strand passage mechanism proposed for the type II enzymes.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA, Superhelical/metabolism , Nucleic Acid Conformation , Vaccinia/enzymology , Kinetics , Models, Chemical , Osmolar Concentration
19.
Biochemistry ; 35(50): 16036-47, 1996 Dec 17.
Article in English | MEDLINE | ID: mdl-8973173

ABSTRACT

The solution secondary structure of 4-oxalocrotonate tautomerase (4-OT), a 41 kDa homohexamer with 62 residues per subunit, consists of an alpha-helix, two beta-strands, a beta-hairpin, two loops, two turns, and a C-terminal coil [Stivers et al. (1996) Protein Sci. 5, 729-741]. The general base, proline-1, as well as the two loops and the beta-hairpin have been shown to comprise the active site [Stivers et al. (1996) Biochemistry 35, 814-823]. The backbone dynamics of both the free enzyme and its complex with a substrate analog have been studied by 1H-detected 15N relaxation rates and NOE determinations at 500 and 600 MHz. Analysis of the data using the model-free formalism showed that the nanosecond to picosecond motion of 53 of the 60 backbone 15N-H vectors was highly restricted with a mean order parameter mean value of S2 = 0.87 +/- 0.03. The lowest backbone mobility (S2 > 0.90) is found in the beta 1-strand, loop 2, and turn 2. Greater backbone mobility is found in the active site (0.5 < or = S2 < or = 0.83) and at C-terminal residues 58-62 (0.03 < or = S2 < or = 0.70). A tau m value for the free hexamer of 13.7 ns at 42 degrees C was determined, consistent with a compact globular molecule of 41 kDa. Saturation of 4-OT with the analog of the dienolic intermediate and linear competitive inhibitor cis, cis-muconate (4) (KD = 0.59 mM) increased the backbone S2 of seven residues and decreased the backbone S2 of another eight residues, both at the active site and at the antiparallel beta 1-beta 1 interface. The S2 values of the other 44 detectable NH vectors were not altered by the binding of 4. The increases in S2, resulting from the "freezing" of the backbone NH vectors of seven residues upon the binding of 4, correspond to an unfavorable entropic contribution to delta Gbinding of 3.2 +/- 1.1 kcal/mol. This freezing is partially compensated for by the mobilization of the other eight residues, since the decreases in S2 for these residues correspond to an entropic contribution to binding of -1.9 +/- 0.1 kcal/mol. These entropy changes, resulting solely from alterations in high-frequency motion, are significant compared to the overall delta Gbinding = -4.6 kcal/mol for 4. Other effects of the binding of 4 include (1) changes in 15N and NH chemical shifts localized to the active site and (2) increases in the exchange contributions (R(ex)) to 1/T2 of backbone 15N resonances at the active site and at the subunit interface, reflecting microsecond to millisecond motions which may play a role in substrate binding (k(on) > or = 4 x 10(6) M-1 s-1) and/or catalysis (kcat = 10(3) s-1).


Subject(s)
Enzyme Inhibitors/metabolism , Isomerases/antagonists & inhibitors , Isomerases/chemistry , Protein Structure, Secondary , Sorbic Acid/analogs & derivatives , Binding Sites , Entropy , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Magnetic Resonance Spectroscopy , Models, Chemical , Models, Structural , Nitrogen Isotopes , Proline , Sorbic Acid/chemistry , Sorbic Acid/metabolism , Sorbic Acid/pharmacology , Stereoisomerism
20.
Protein Sci ; 5(4): 729-41, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8845763

ABSTRACT

4-Oxalocrotonate tautomerase (4-OT), a homohexamer consisting of 62 residues per subunit, catalyzes the isomerization of unsaturated alpha-keto acids using Pro-1 as a general base (Stivers et al., 1996a, 1996b). We report the backbone and side-chain 1H, 15N, and 13C NMR assignments and the solution secondary structure for 4-OT using 2D and 3D homonuclear and heteronuclear NMR methods. The subunit secondary structure consists of an alpha-helix (residues 13-30), two beta-strands (beta 1, residues 2-8; beta 2, residues 39-45), a beta-hairpin (residues 50-57), two loops (I, residues 9-12; II, 34-38), and two turns (I, residues 30-33; II, 47-50). The remaining residues form coils. The beta 1 strand is parallel to the beta 2 strand of the same subunit on the basis of cross stand NH(i)-NH(j) NOEs in a 2D 15N-edited 1H-NOESY spectrum of hexameric 4-OT containing two 15N-labeled subunits/hexamer. The beta 1 strand is also antiparallel to another beta 1 strand from an adjacent subunit forming a subunit interface. Because only three such pairwise interactions are possible, the hexamer is a trimer of dimers. The diffusion constant, determined by dynamic light scattering, and the rotational correlation time (14.5 ns) estimated from 15N T1/T2 measurements, are consistent with the hexameric molecular weight of 41 kDa. Residue Phe-50 is in the active site on the basis of transferred NOEs to the bound partial substrate 2-oxo-1,6-hexanedioate. Modification of the general base, Pro-1, with the active site-directed irreversible inhibitor, 3-bromopyruvate, significantly alters the amide 15N and NH chemical shifts of residues in the beta-hairpin and in loop II, providing evidence that these regions change conformation when the active site is occupied.


Subject(s)
Isomerases/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...