Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nucleosides Nucleotides Nucleic Acids ; 20(4-7): 1239-41, 2001.
Article in English | MEDLINE | ID: mdl-11562993

ABSTRACT

A DNA-binding peptide was selected from a random peptide phage display library. For competitive elution using the DNA methyltransferase M.TaqI in the selection step, a biotin-labeled duplex oligodeoxyribonucleotide containing the 5'-TCGA-3' recognition sequence of M.TaqI was employed. Nine of ten phages selected were found to have the same deduced amino acid sequence SVSVGMKPSPRP. The selected phage binds to DNA, as demonstrated in an ELISA.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Oligopeptides/metabolism , Peptide Library , Amino Acid Sequence , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism
2.
J Biol Chem ; 276(24): 20924-34, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11283006

ABSTRACT

Kinetic and binding studies involving a model DNA cytosine-5-methyltransferase, M.HhaI, and a 37-mer DNA duplex containing a single hemimethylated target site were applied to characterize intermediates on the reaction pathway. Stopped-flow fluorescence studies reveal that cofactor S-adenosyl-l-methionine (AdoMet) and product S-adenosyl-l-homocysteine (AdoHcy) form similar rapidly reversible binary complexes with the enzyme in solution. The M.HhaI.AdoMet complex (k(off) = 22 s(-)1, K(D) = 6 microm) is partially converted into products during isotope-partitioning experiments, suggesting that it is catalytically competent. Chemical formation of the product M.HhaI.(Me)DNA.AdoHcy (k(chem) = 0.26 s(-)1) is followed by a slower decay step (k(off) = 0.045 s(-)1), which is the rate-limiting step in the catalytic cycle (k(cat) = 0.04 s(-)1). Analysis of reaction products shows that the hemimethylated substrate undergoes complete (>95%) conversion into fully methylated product during the initial burst phase, indicating that M.HhaI exerts high binding selectivity toward the target strand. The T250N, T250D, and T250H mutations, which introduce moderate perturbation in the catalytic site, lead to substantially increased K(D)(DNA(ternary)), k(off)(DNA(ternary)), K(M)(AdoMet(ternary)) values but small changes in K(D)(DNA(binary)), K(D)(AdoMet(binary)), k(chem), and k(cat). When the target cytosine is replaced with 5-fluorocytosine, the chemistry step leading to an irreversible covalent M.HhaI.DNA complex is inhibited 400-fold (k(chem)(5FC) = 0.7 x 10(-)3 s(-)1), and the Thr-250 mutations confer further dramatic decrease of the rate of the covalent methylation k(chem). We suggest that activation of the pyrimidine ring via covalent addition at C-6 is a major contributor to the rate of the chemistry step (k(chem)) in the case of cytosine but not 5-fluorocytosine. In contrast to previous reports, our results imply a random substrate binding order mechanism for M.HhaI.


Subject(s)
DNA Methylation , DNA-Cytosine Methylases/metabolism , Amino Acid Substitution , Base Sequence , DNA-Cytosine Methylases/chemistry , DNA-Cytosine Methylases/genetics , Kinetics , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Restriction Mapping , S-Adenosylmethionine/metabolism
3.
Nat Struct Biol ; 8(2): 121-5, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11175899

ABSTRACT

The 2.0 A crystal structure of the N6-adenine DNA methyltransferase M.TaqI in complex with specific DNA and a nonreactive cofactor analog reveals a previously unrecognized stabilization of the extrahelical target base. To catalyze the transfer of the methyl group from the cofactor S-adenosyl-l-methionine to the 6-amino group of adenine within the double-stranded DNA sequence 5'-TCGA-3', the target nucleoside is rotated out of the DNA helix. Stabilization of the extrahelical conformation is achieved by DNA compression perpendicular to the DNA helix axis at the target base pair position and relocation of the partner base thymine in an interstrand pi-stacked position, where it would sterically overlap with an innerhelical target adenine. The extrahelical target adenine is specifically recognized in the active site, and the 6-amino group of adenine donates two hydrogen bonds to Asn 105 and Pro 106, which both belong to the conserved catalytic motif IV of N6-adenine DNA methyltransferases. These hydrogen bonds appear to increase the partial negative charge of the N6 atom of adenine and activate it for direct nucleophilic attack on the methyl group of the cofactor.


Subject(s)
DNA/metabolism , S-Adenosylmethionine/analogs & derivatives , S-Adenosylmethionine/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)/chemistry , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Thermus/enzymology , Adenine/chemistry , Adenine/metabolism , Amino Acid Sequence , Base Pairing , Binding Sites , Catalysis , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , DNA Methylation , Enzyme Stability , Hydrogen Bonding , Models, Molecular , Protein Conformation , Rotation , S-Adenosylmethionine/chemistry
4.
J Biol Chem ; 275(49): 38722-30, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11102456

ABSTRACT

DNA cytosine-5-methyltransferase HhaI recognizes the GCGC sequence and flips the inner cytosine out of DNA helix and into the catalytic site for methylation. The 5'-phosphate of the flipped out cytosine is in contact with the conserved Thr-250 from the target recognition domain. We have produced 12 mutants of Thr-250 and examined their methylation potential in vivo. Six active mutants were subjected to detailed biochemical and structural studies. Mutants with similar or smaller side chains (Ser, Cys, and Gly) are very similar to wild-type enzyme in terms of steady-state kinetic parameters k(cat), K(m)(DNA), K(m)(AdoMet). In contrast, the mutants with bulkier side chains (Asn, Asp, and His) show increased K(m) values for both substrates. Fluorescence titrations and stopped-flow kinetic analysis of interactions with duplex oligonucleotides containing 2-aminopurine at the target base position indicate that the T250G mutation leads to a more polar but less solvent-accessible position of the flipped out target base. The x-ray structure of the ternary M. HhaI(T250G).DNA.AdoHcy complex shows that the target cytosine is locked in the catalytic center of enzyme. The space created by the mutation is filled by water molecules and the adjacent DNA backbone atoms dislocate slightly toward the missing side chain. In aggregate, our results suggest that the side chain of Thr-250 is involved in constraining the conformation the DNA backbone and the target base during its rotation into the catalytic site of enzyme.


Subject(s)
DNA-Cytosine Methylases/chemistry , DNA-Cytosine Methylases/metabolism , Threonine , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Binding Sites , Conserved Sequence , DNA Methylation , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Insertional , Nucleic Acid Conformation , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
5.
Biochemistry ; 39(47): 14611-6, 2000 Nov 28.
Article in English | MEDLINE | ID: mdl-11087417

ABSTRACT

The enzymes that transfer a methyl group to C5 of cytosine within specific sequences (C5 Mtases) deaminate the target cytosine to uracil if the methyl donor S-adenosylmethionine (SAM) is omitted from the reaction. Recently, it was shown that cytosine deamination caused by C5 Mtases M.HpaII, M.SssI and M.MspI is enhanced in the presence of several analogues of SAM, and a mechanism for this analogue-promoted deamination was proposed. According to this mechanism, the analogues protonate C5 of the target cytosine, creating a dihydrocytosine intermediate that is susceptible to deamination. We show here that one of these analogues, 5'-aminoadenosine (AA), enhances cytosine deamination by the Mtase M. EcoRII, but it does so without enhancing protonation of C5. Further, we show that uracil is an intermediate in the mutational pathway and propose an alternate mechanism for the analogue-promoted deamination. The new mechanism involves a facilitated water attack at C4 but does not require attack at C6 by the enzyme. The latter feature of the mechanism was tested by using M.EcoRII mutants defective in the nucleophilic attack at C6 in the deamination assay. We find that although these proteins are defective in methyl transfer and cytosine deamination, they cause cytosine deaminations in the presence of AA in the reaction. Our results point to a possible connection between the catalytic mechanism of C5 Mtases and of enzymes that transfer methyl groups to N(4) of cytosine. Further, they provide an unusual example where a coenzyme activates an otherwise "dead" enzyme to perform catalysis by a new reaction pathway.


Subject(s)
Cytosine/chemistry , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/chemistry , S-Adenosylmethionine/chemistry , Adenosine/analogs & derivatives , Adenosine/chemistry , Alanine/genetics , Cysteine/genetics , DNA-Cytosine Methylases/chemistry , DNA-Cytosine Methylases/genetics , Deamination , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Mutagenesis, Site-Directed , Serine/genetics , Uracil/chemistry
6.
J Biol Chem ; 274(21): 15066-72, 1999 May 21.
Article in English | MEDLINE | ID: mdl-10329711

ABSTRACT

DNA methyltransferases flip their target bases out of the DNA double helix for catalysis. Base flipping of C5-cytosine DNA methyltransferases was directly observed in the protein-DNA cocrystal structures of M.HhaI and M.HaeIII. Indirect structural evidence for base flipping of N6-adenine and N4-cytosine DNA methyltransferases was obtained by modeling DNA into the three-dimensional structures of M.TaqI and M.PvuII in complex with the cofactor. In addition, biochemical evidence of base flipping was reported for different N6-adenine DNA methyltransferases. As no protein-DNA cocrystal structure for the related N6-adenine and N4-cytosine DNA methyltransferases is available, we used light-induced photochemical cross-linking to identify the binding site of the extrahelical target bases. The N6-adenine DNA methyltransferases M.TaqI and M.CviBIII, which both methylate adenine within the double-stranded 5'-TCGA-3' DNA sequence, were photo-cross-linked to duplex oligodeoxyribonucleotides containing 5-iodouracil at the target position in 50-60% and almost quantitative yield, respectively. Proteolytic fragmentation of the M. CviBIII-DNA complex followed by Edman degradation and electrospray ionization mass spectrometry indicates photo-cross-linking to tyrosine 122. In addition, the mutant methyltransferases M. TaqI/Y108A and M.TaqI/F196A were photo-cross-linked with 6-fold and 2-fold reduced efficiency, respectively, which suggests that tyrosine 108 is the primary site of modification in M.TaqI. Our results indicate a close proximity between the extrahelical target base and tyrosine 122 in M.CviBIII or tyrosine 108 in M.TaqI. As both residues belong to the conserved motif IV ((N/D/S)(P/I)P(Y/F/W)) found in all N6-adenine and N4-cytosine DNA as well as in N6-adenine RNA methyltransferases, a similar spatial relationship between the target bases and the aromatic amino acid residue within motif IV is expected for all these methyltransferases.


Subject(s)
Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/radiation effects , Site-Specific DNA-Methyltransferase (Adenine-Specific)/chemistry , Site-Specific DNA-Methyltransferase (Adenine-Specific)/radiation effects , Uracil/analogs & derivatives , Amino Acid Sequence , Binding Sites , Protein Structure, Tertiary , Site-Specific DNA-Methyltransferase (Adenine-Specific)/isolation & purification , Ultraviolet Rays , Uracil/radiation effects
7.
Biochemistry ; 38(5): 1426-34, 1999 Feb 02.
Article in English | MEDLINE | ID: mdl-9931007

ABSTRACT

The DNA methyltransferase (Mtase) from Thermus aquaticus (M.TaqI) catalyzes the transfer of the activated methyl group of S-adenosyl-L-methionine to the N6 position of adenine within the double-stranded DNA sequence 5'-TCGA-3'. To achieve catalysis M.TaqI flips the target adenine out of the DNA helix. On the basis of the three-dimensional structure of M.TaqI in complex with the cofactor and its structural homology to the C5-cytosine DNA Mtase from Haemophilus haemolyticus, Tyr 108 and Phe 196 were suggested to interact with the extrahelical adenine. The functional roles of these two aromatic amino acid residues in M.TaqI were investigated by mutational analysis. The obtained mutant Mtases were analyzed in an improved kinetic assay, and their ability to flip the target base was studied in a fluorescence-based assay using a duplex oligodeoxynucleotide containing the fluorescent base analogue 2-aminopurine at the target position. While the mutant Mtases containing the aromatic amino acid Trp at position 108 or 196 (Y108W and F196W) showed almost wild-type catalytic activity, the mutant Mtases with the nonaromatic amino acid Ala (Y108A and F196A) had a strongly reduced catalytic constant. Y108A was still able to flip the target base, whereas F196A was strongly impaired in base flipping. These results indicate that Phe 196 is important for stabilizing the extrahelical target adenine and suggest that Tyr 108 is involved in placing the extrahelical target base in an optimal position for methyl group transfer. Since both aromatic amino acids belong to the conserved motifs IV and XIII found in N6-adenine and N4-cytosine DNA Mtases as well as in N6-adenine RNA Mtases, a similar function of aromatic amino acid residues within these motifs is expected for the different Mtases.


Subject(s)
Conserved Sequence , Site-Specific DNA-Methyltransferase (Adenine-Specific)/chemistry , Thermus/enzymology , Tryptophan/physiology , Amino Acid Sequence , Base Pair Mismatch , Catalysis , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Phenylalanine/chemistry , Phenylalanine/genetics , Phenylalanine/physiology , Polymerase Chain Reaction , Site-Specific DNA-Methyltransferase (Adenine-Specific)/genetics , Taq Polymerase/genetics , Tryptophan/chemistry , Tryptophan/genetics
8.
Nucleic Acids Res ; 26(15): 3473-9, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9671807

ABSTRACT

The DNA cytosine-C5 methyltransferase M. Hha I flips its target base out of the DNA helix during interaction with the substrate sequence GCGC. Binary and ternary complexes between M. Hha I and hemimethylated DNA duplexes were used to examine the suitability of four chemical methods to detect flipped-out bases in protein-DNA complexes. These methods probe the structural peculiarities of pyrimidine bases in DNA. We find that in cases when the target cytosine is replaced with thymine (GTGC), KMnO4proved an efficient probe for positive display of flipped-out thymines. The generality of this procedure was further verified by examining a DNA adenine-N6 methyltransferase, M. Taq I, in which case an enhanced reactivity of thymine replacing the target adenine (TCGT) in the recognition sequence TCGA was also observed. Our results support the proposed base-flipping mechanism for adenine methyltransferases, and offer a convenient laboratory tool for detection of flipped-out thymines in protein-DNA complexes.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Cytosine Methylases/metabolism , Thymine/metabolism , Nucleic Acid Heteroduplexes/metabolism , Oligodeoxyribonucleotides/metabolism , S-Adenosylmethionine/analogs & derivatives , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism
9.
Nucleic Acids Res ; 26(4): 1076-83, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9461471

ABSTRACT

DNA base flipping, which was first observed for the C5-cytosine DNA methyltransferase M. Hha I, results in a complete removal of the stacking interactions between the target base and its neighbouring bases. We have investigated whether duplex oligodeoxynucleotides containing the fluorescent base analogue 2-aminopurine can be used to sense DNA base flipping. Using M. Hha I as a paradigm for a base flipping enzyme, we find that the fluorescence intensity of duplex oligodeoxynucleotides containing 2-aminopurine at the target site is dramatically enhanced (54-fold) in the presence of M. Hha I. Duplex oligodeoxynucleotides containing 2-aminopurine adjacent to the target cytosine show little fluorescence increase upon addition of M. Hha I. These results clearly demonstrate that duplex oligodeoxynucleotides containing 2-aminopurine at the target site can serve as fluorescence probes for base flipping. Another enzyme hypothesized to use a base flipping mechanism is the N6-adenine DNA methyltransferase M. Taq I. Addition of M. Taq I to duplex oligodeoxynucleotides bearing 2-aminopurine at the target position, also results in a strongly enhanced fluorescence (13-fold), whereas addition to duplex oligodeoxynucleotides containing 2-aminopurine at the 3'- or 5'-neighbouring position leads only to small fluorescence increases. These results give the first experimental evidence that the adenine-specific DNA methyltransferase M. Taq I also flips its target base.


Subject(s)
2-Aminopurine , DNA-Cytosine Methylases , DNA/chemistry , DNA/metabolism , Fluorescent Dyes , 2-Aminopurine/chemistry , Base Sequence , DNA-Cytosine Methylases/metabolism , Fluorescent Dyes/chemistry , Oligodeoxyribonucleotides/chemical synthesis , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Spectrometry, Fluorescence
10.
Nucleic Acids Res ; 25(6): 1303-4, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9092643

ABSTRACT

A quick in vitro mutagenesis method for the construction of nested deletion libraries was developed. Many deletions can be obtained in a single inverse PCR (IPCR) by replacing one of the two primers with a mixture of 5'-truncated oligodeoxynucleotides. Since chemical DNA synthesis proceeds from the 3'to the 5'end, such a mixture of 5'-truncated oligodeoxynucleotides can easily be obtained in a single automated DNA synthesis under reduced coupling efficiency. This deletion mutagenesis method yields many different deletions in a defined short DNA segment and is, therefore, best suited for a deletion analysis at base pair level. Applications might include functional analysis of regulatory DNA segments and protein engineering work that requires libraries for the expression of N-terminal, C-terminal or internal truncated proteins as well as fusion proteins having different splice sites.


Subject(s)
Gene Library , Mutagenesis, Site-Directed , Polymerase Chain Reaction/methods , Sequence Deletion , Base Sequence , DNA/chemical synthesis , DNA Primers , Deoxyribonucleases, Type II Site-Specific/biosynthesis , Deoxyribonucleases, Type II Site-Specific/genetics , Genetic Variation , Molecular Sequence Data , Oligodeoxyribonucleotides/chemical synthesis , Protein Engineering , Regulatory Sequences, Nucleic Acid , Thermus/enzymology , Thermus/genetics
11.
J Mol Biol ; 265(1): 56-67, 1997 Jan 10.
Article in English | MEDLINE | ID: mdl-8995524

ABSTRACT

The crystal structures of the binary complexes of the DNA methyltransferase M.TaqI with the inhibitor Sinefungin and the reaction product S-adenosyl-L-homocysteine were determined, both at 2.6 A resolution. Structural comparison of these binary complexes with the complex formed by M.TaqI and the cofactor S-adenosyl-L-methionine suggests that the key element for molecular recognition of these ligands is the binding of their adenosine part in a pocket, and discrimination between cofactor, reaction product and inhibitor is mediated by different conformations of these molecules; the methionine part of S-adenosyl-L-methionine is located in the binding cleft, whereas the amino acid moieties of Sinefungin and S-adenosyl-L-homocysteine are in a different orientation and interact with the active site amino acid residues 105NPPY108. Dissociation constants for the complexes of M.TaqI with the three ligands were determined spectrofluorometrically. Sinefungin binds more strongly than S-adenosyl-L-homocysteine or S-adenosyl-L-methionine, with KD=0.34 microM, 2.4 microM and 2.0 microM, respectively.


Subject(s)
Adenosine/analogs & derivatives , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Adenosine/chemistry , Adenosine/metabolism , Binding Sites , Catalysis , Crystallography, X-Ray , Electrochemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli/enzymology , Hydrogen Bonding , Kinetics , Ligands , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Molecular Structure , Protein Conformation , S-Adenosylhomocysteine/chemistry , S-Adenosylmethionine/chemistry , Site-Specific DNA-Methyltransferase (Adenine-Specific)/antagonists & inhibitors , Site-Specific DNA-Methyltransferase (Adenine-Specific)/chemistry , Spectrometry, Fluorescence
12.
Acta Chem Scand (Cph) ; 50(3): 243-8, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8901176

ABSTRACT

Paleomolecular biochemistry is a new field of science that seeks to understand how life emerged and developed in interaction with its geophysical surroundings. It is an experimental science, involving reconstruction of extinct biomolecules in the laboratory, studying their properties in the laboratory, and inferring details of their behavior and function in the context of geological data. An outline is provided of some tools of this field, together with its application to the study of two specific systems, ribonuclease and alcohol dehydrogenase.


Subject(s)
Biological Evolution , Catalysis , Enzymes/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data
13.
Protein Eng ; 8(5): 457-61, 1995 May.
Article in English | MEDLINE | ID: mdl-8532667

ABSTRACT

Analysis of a crystal structure of alcohol dehydrogenase (Adh) from horse liver suggests that Trp54 in the homologous yeast alcohol dehydrogenase prevents the yeast enzyme from efficiently catalysing the oxidation of long-chain primary alcohols with branching at the 4 position (e.g. 4-methyl-1-pentanol, cinnamyl alcohol). This residue has been altered to Leu by site-directed mutagenesis. The alteration yields an enzyme that serves as an effective catalyst for both longer straight-chain primary alcohols and branched chain alcohols.


Subject(s)
Alcohol Dehydrogenase/metabolism , Protein Engineering , Saccharomyces cerevisiae/enzymology , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/genetics , Alcohols/metabolism , Binding Sites , Kinetics , Leucine/chemistry , Mutagenesis, Site-Directed , Saccharomyces cerevisiae/genetics , Substrate Specificity , Tryptophan/chemistry
14.
Biochem J ; 281 ( Pt 1): 211-8, 1992 Jan 01.
Article in English | MEDLINE | ID: mdl-1731758

ABSTRACT

D(--)-Mandelate dehydrogenase, the first enzyme of the mandelate pathway in the yeast Rhodotorula graminis, catalyses the NAD(+)-dependent oxidation of D(--)-mandelate to phenylglyoxylate. D(--)-2-(Bromoethanoyloxy)-2-phenylethanoic acid ['D(--)-bromoacetylmandelic acid'], an analogue of the natural substrate, was synthesized as a probe for reactive and accessible nucleophilic groups within the active site of the enzyme. D(--)-Mandelate dehydrogenase was inactivated by D(--)-bromoacetylmandelate in a psuedo-first-order process. D(--)-Mandelate protected against inactivation, suggesting that the residue that reacts with the inhibitor is located at or near the active site. Complete inactivation of the enzyme resulted in the incorporation of approx. 1 mol of label/mol of enzyme subunit. D(--)-Mandelate dehydrogenase that had been inactivated with 14C-labelled D(--)-bromoacetylmandelate was digested with trypsin; there was substantial incorporation of 14C into two tryptic-digest peptides, and this was lowered in the presence of substrate. One of the tryptic peptides had the sequence Val-Xaa-Leu-Glu-Ile-Gly-Lys, with the residue at the second position being the site of radiolabel incorporation. The complete sequence of the second peptide was not determined, but it was probably an N-terminally extended version of the first peptide. High-voltage electrophoresis of the products of hydrolysis of modified protein showed that the major peak of radioactivity co-migrated with N tau-carboxymethylhistidine, indicating that a histidine residue at the active site of the enzyme is the most likely nucleophile with which D(--)-bromoacetylmandelate reacts. D(--)-Mandelate dehydrogenase was incubated with phenylglyoxylate and either (4S)-[4-3H]NADH or (4R)-[4-3H]NADH and then the resulting D(--)-mandelate and NAD+ were isolated. The enzyme transferred the pro-R-hydrogen atom from NADH during the reduction of phenylglyoxylate. The results are discussed with particular reference to the possibility that this enzyme evolved by the recruitment of a 2-hydroxy acid dehydrogenase from another metabolic pathway.


Subject(s)
Alcohol Oxidoreductases/metabolism , Rhodotorula/enzymology , Affinity Labels/chemical synthesis , Affinity Labels/metabolism , Amino Acid Sequence , Binding Sites , Carbon Radioisotopes , Mandelic Acids/chemical synthesis , Mandelic Acids/metabolism , Molecular Sequence Data , NAD/metabolism , Peptide Fragments/isolation & purification , Radioisotope Dilution Technique , Tritium
15.
Proc Natl Acad Sci U S A ; 88(19): 8420-4, 1991 Oct 01.
Article in English | MEDLINE | ID: mdl-1924300

ABSTRACT

Replacing Leu-182 by Ala in yeast alcohol dehydrogenase (YADH; alcohol:NAD+ oxidoreductase, EC 1.1.1.1) yields a mutant that retains 34% of its kcat value and makes one stereochemical "mistake" every 850,000 turnovers (instead of approximately 1 error every 7,000,000,000 turnovers in native YADH) in its selection of the 4-Re hydrogen of NADH. Half of the decrease in stereochemical fidelity comes from an increase in the rate of transfer of the 4-Si hydrogen of NADH. The mutant also accepts 5-methylnicotinamide adenine dinucleotide, a cofactor analog not accepted by native YADH. The stereospecificity of the mutant is lower still with analogs of NADH where the carboxamide group of the nicotinamide ring is replaced by groups with weaker hydrogen bonding potential. For example, with thio-NADH, the mutant enzyme makes 1 stereochemical "mistake" every 450 turnovers. Finally, the double mutant T157S/L182A, in which Thr-157 is replaced by Ser and Leu-182 is replaced by Ala, also shows decreased stereochemical fidelity. These results suggest that Si transfer in the mutant enzymes arises from NADH bound in a syn conformation in the active site and that this binding is not obstructed in native YADH by side chains essential for catalysis.


Subject(s)
Alcohol Dehydrogenase/metabolism , Saccharomyces cerevisiae/enzymology , DNA Mutational Analysis , Ethanol/metabolism , Kinetics , NAD/metabolism , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...