Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Head Neck ; 32(11): 1479-84, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20175196

ABSTRACT

BACKGROUND: Opinions differ regarding the usefulness of accurate, but costly, frozen sections. Most physicians believe that negative margins are essential for the prognosis of patients with oral and pharyngeal cancer. We examined whether immediate repeated resections in patients with positive margins, based on findings from frozen sections, resulted in improved patient survival. METHODS: Data from 417 patients identified with cancer of the pharynx and floor of the mouth were analyzed retrospectively. RESULTS: The 5-year survival rate for R0 and R1-R0 groups was 72% to 76% and was significantly better (p ≤ .034) than that for R1 and R2 groups (58%, 40%). Despite clear margins, large tumors had a poorer prognosis than that of small tumors. CONCLUSIONS: Patients receiving repeated resection had the same survival rate as patients who had the tumor resected immediately with negative margins. The use of frozen sections yields a benefit for 15.6% of the operated patients and increases the overall 5-year survival rate by 2% to 3%.


Subject(s)
Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Pharyngeal Neoplasms/mortality , Pharyngeal Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma/mortality , Carcinoma/pathology , Carcinoma/surgery , Female , Frozen Sections , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mouth Neoplasms/surgery , Pharyngeal Neoplasms/surgery , Retrospective Studies , Survival Rate
2.
J Biol Chem ; 283(9): 5344-54, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18086676

ABSTRACT

The gene rv0853c from Mycobacterium tuberculosis strain H37Rv codes for a thiamine diphosphate-dependent alpha-keto acid decarboxylase (MtKDC), an enzyme involved in the amino acid degradation via the Ehrlich pathway. Steady state kinetic experiments were performed to determine the substrate specificity of MtKDC. The mycobacterial enzyme was found to convert a broad spectrum of branched-chain and aromatic alpha-keto acids. Stopped-flow kinetics showed that MtKDC is allosterically activated by alpha-keto acids. Even more, we demonstrate that also amino acids are potent activators of this thiamine diphosphate-dependent enzyme. Thus, metabolic flow through the Ehrlich pathway can be directly regulated at the decarboxylation step. The influence of amino acids on MtKDC catalysis was investigated, and implications for other thiamine diphosphate-dependent enzymes are discussed.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Thiamine Pyrophosphate/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Allosteric Regulation/physiology , Bacterial Proteins/genetics , Enzyme Activation/physiology , Keto Acids/metabolism , Kinetics , Mycobacterium tuberculosis/genetics
3.
Biochem Biophys Res Commun ; 366(3): 692-7, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18070592

ABSTRACT

Transketolase from Saccharomyces cerevisiae exhibits a rarely reported activity with a methylated analogue of the native cofactor, 4'-methylamino-thiamin diphosphate. We demonstrated the kinetic stability of the dihydroxyethyl carbanion/enamine intermediate to be dependent on the functionality of the 4'-aminopyrimidine moiety of thiamin diphosphate [R. Golbik, L.E. Meshalkina, T. Sandalova, K. Tittmann, E. Fiedler, H. Neef, S. König, R. Kluger, G.A. Kochetov, G. Schneider, G. Hübner, Effect of coenzyme modification on the structural and catalytic properties of wild-type transketolase and of the variant E418A from Saccharomyces cerevisae, FEBS J. (2005) 272 1326-1342]. This paper extends these investigations of the function of the coenzyme's aminopyrimidine in transketolase catalysis exemplified for the 4'-monomethylamino-thiamin diphosphate analogue. Here, we report near UV circular dichroism data and NMR-based analysis of reaction intermediates that give evidence for a strong destabilisation of the carbanion/enamine of DHE-4'-monomethylamino-thiamin diphosphate on the enzyme. A new negative band in near UV circular dichroism arising during turnover is attributed to the conjugate acid of the carbanion/enamine intermediate, an assignment additionally corroborated by (1)H NMR-based intermediate analysis. As opposed to the kinetically stabilized carbanion/enamine intermediate in transketolase when reconstituted with the native cofactor, DHE-4'-monomethylamino-thiamin diphosphate is rapidly released from the active centers during turnover and accumulates in the medium on a preparative scale.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Thiamine/chemistry , Transketolase/chemistry , Binding Sites , Catalysis , Enzyme Activation , Isoenzymes/chemistry , Protein Binding
4.
Biochemistry ; 46(43): 12037-52, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17914867

ABSTRACT

Transketolase is a prominent thiamin diphosphate-dependent enzyme in sugar metabolism that catalyzes the reversible transfer of a 2-carbon dihydroxyethyl fragment between a donor ketose and an acceptor aldose. The X-ray structures of transketolase from E. coli in a covalent complex with donor ketoses d-xylulose 5-phosphate (X5P) and d-fructose 6-phosphate (F6P) at 1.47 A and 1.65 A resolution reveal significant strain in the tetrahedral cofactor-sugar adducts with a 25-30 degrees out-of-plane distortion of the C2-Calpha bond connecting the substrates' carbonyl with the C2 of the cofactor's thiazolium part. Both intermediates adopt very similar extended conformations in the active site with a perpendicular orientation of the scissile C2-C3 sugar bond relative to the thiazolium ring. The sugar-derived hydroxyl groups of the intermediates form conserved hydrogen bonds with one Asp side chain, with a cluster of His residues and with the N4' of the aminopyrimidine ring of the cofactor. The phosphate moiety is held in place by electrostatic and hydrogen-bonding interactions with Arg, His, and Ser side chains. With the exception of the thiazolium part of the cofactor, no structural changes are observable during intermediate formation indicating that the active site is poised for catalysis. DFT calculations on both X5P-thiamin and X5P-thiazolium models demonstrate that an out-of-plane distortion of the C2-Calpha bond is energetically more favorable than a coplanar bond. The X-ray structure with the acceptor aldose d-ribose 5-phosphate (R5P) noncovalently bound in the active site suggests that the sugar is present in multiple forms: in a strained ring-closed beta-d-furanose form in C2-exo conformation as well as in an extended acyclic aldehyde form, with the reactive C1 aldo function held close to Calpha of the presumably planar carbanion/enamine intermediate. The latter form of R5P may be viewed as a near attack conformation. The R5P binding site overlaps with those of the leaving group moieties of the covalent donor-cofactor adducts, demonstrating that R5P directly competes with the donor-derived products glyceraldehyde 3-phosphate and erythrose 4-phosphate, which are substrates of the reverse reaction, for the same docking site at the active site and reaction with the DHEThDP enamine.


Subject(s)
Escherichia coli/enzymology , Fructosephosphates/chemistry , Pentosephosphates/chemistry , Thiamine/metabolism , Transketolase/chemistry , Base Sequence , Catalysis , Crystallography, X-Ray , DNA Primers , Hydrogen Bonding , Models, Molecular , Molecular Conformation , Nuclear Magnetic Resonance, Biomolecular , Transketolase/metabolism
5.
FEBS J ; 273(18): 4199-209, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16939618

ABSTRACT

The crystal structure of pyruvate decarboxylase from Kluyveromyces lactis has been determined to 2.26 A resolution. Like other yeast enzymes, Kluyveromyces lactis pyruvate decarboxylase is subject to allosteric substrate activation. Binding of substrate at a regulatory site induces catalytic activity. This process is accompanied by conformational changes and subunit rearrangements. In the nonactivated form of the corresponding enzyme from Saccharomyces cerevisiae, all active sites are solvent accessible due to the high flexibility of loop regions 106-113 and 292-301. The binding of the activator pyruvamide arrests these loops. Consequently, two of four active sites become closed. In Kluyveromyces lactis pyruvate decarboxylase, this half-side closed tetramer is present even without any activator. However, one of the loops (residues 105-113), which are flexible in nonactivated Saccharomyces cerevisiae pyruvate decarboxylase, remains flexible. Even though the tetramer assemblies of both enzyme species are different in the absence of activating agents, their substrate activation kinetics are similar. This implies an equilibrium between the open and the half-side closed state of yeast pyruvate decarboxylase tetramers. The completely open enzyme state is favoured for Saccharomyces cerevisiae pyruvate decarboxylase, whereas the half-side closed form is predominant for Kluyveromyces lactis pyruvate decarboxylase. Consequently, the structuring of the flexible loop region 105-113 seems to be the crucial step during the substrate activation process of Kluyveromyces lactis pyruvate decarboxylase.


Subject(s)
Kluyveromyces/enzymology , Pyruvate Decarboxylase/chemistry , Pyruvate Decarboxylase/metabolism , Allosteric Regulation , Amino Acid Substitution , Binding Sites , Crystallography, X-Ray , Enzyme Activation , Kluyveromyces/chemistry , Kluyveromyces/metabolism , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Pyruvate Decarboxylase/genetics , Saccharomyces cerevisiae/enzymology
6.
Biochemistry ; 44(40): 13291-303, 2005 Oct 11.
Article in English | MEDLINE | ID: mdl-16201755

ABSTRACT

The thiamin diphosphate (ThDP)- and flavin adenine dinucleotide (FAD)-dependent pyruvate oxidase from Lactobacillus plantarum catalyses the conversion of pyruvate, inorganic phosphate, and oxygen to acetyl-phosphate, carbon dioxide, and hydrogen peroxide. Central to the catalytic sequence, two reducing equivalents are transferred from the resonant carbanion/enamine forms of alpha-hydroxyethyl-ThDP to the adjacent flavin cofactor over a distance of approximately 7 A, followed by the phosphorolysis of the thereby formed acetyl-ThDP. Pre-steady-state and steady-state kinetics using time-resolved spectroscopy and a 1H NMR-based intermediate analysis indicate that both processes are kinetically coupled. In the presence of phosphate, intercofactor electron-transfer (ET) proceeds with an apparent first-order rate constant of 78 s(-1) and is kinetically gated by the preceding formation of the tetrahedral substrate-ThDP adduct 2-lactyl-ThDP and its decarboxylation. No transient flavin radicals are detectable in the reductive half-reaction. In contrast, when phosphate is absent, ET occurs in two discrete steps with apparent rate constants of 81 and 3 s(-1) and transient formation of a flavin semiquinone/hydroxyethyl-ThDP radical pair. Temperature dependence analysis according to the Marcus theory identifies the second step, the slow radical decay to be a true ET reaction. The redox potentials of the FAD(ox)/FAD(sq) (E1 = -37 mV) and FAD(sq)/FAD(red) (E2 = -87 mV) redox couples in the absence and presence of phosphate are identical. Both the Marcus analysis and fluorescence resonance energy-transfer studies using the fluorescent N3'-pyridyl-ThDP indicate the same cofactor distance in the presence or absence of phosphate. We deduce that the exclusive 10(2)-10(3)-fold rate enhancement of the second ET step is rather due to the nucleophilic attack of phosphate on the kinetically stabilized hydroxyethyl-ThDP radical resulting in a low-potential anion radical adduct than phosphate in a docking site being part of a through-bonded ET pathway in a stepwise mechanism of ET and phosphorolysis. Thus, LpPOX would constitute the first example of a radical-based phosphorolysis mechanism in biochemistry.


Subject(s)
Flavin-Adenine Dinucleotide/chemistry , Lactobacillus plantarum/enzymology , Pyruvate Oxidase/chemistry , Thiamine Pyrophosphate/analogs & derivatives , Thiamine Pyrophosphate/chemistry , Catalysis , Electrons , Flavins/chemistry , Fluorescence Resonance Energy Transfer , Free Radicals , Hydrogen-Ion Concentration , Kinetics , Magnetic Resonance Spectroscopy , Models, Chemical , Models, Statistical , Oxidation-Reduction , Oxygen/chemistry , Phosphates/chemistry , Pyruvic Acid/chemistry , Solvents , Spectrophotometry , Temperature , Thermodynamics , Time Factors
7.
Biochemistry ; 44(24): 8697-700, 2005 Jun 21.
Article in English | MEDLINE | ID: mdl-15952776

ABSTRACT

Proton-nitrogen correlated NMR studies were performed on thiamin diphosphate, which has been specifically labeled with (15)N at the 4'-amino group. After reconstitution of the labeled coenzyme with the apoenzymes of both wild-type pyruvate decarboxylase from Zymomonas mobilis and the E50Q variant, a high-field shift of the (15)N signal of approximately 4 ppm is observed at pH 5.9 when compared to that of the free coenzyme, indicating a higher electron density at the 4'-amino nitrogen in the enzyme-bound state. The pH dependence of the chemical shift of the (15)N signals in the (1)H-(15)N heteronuclear single-quantum coherence NMR spectra reveals typical titration curves for the free as well as the reconstituted coenzyme with nearly identical chemical shift end points. The midpoints of the transitions are at pH 5.3 and 5.0 for the free and enzyme-bound coenzyme, respectively. We conclude that the tremendous rate acceleration of C2-H deprotonation in ThDP enzymes is mainly the result of the enforced V conformation of the cofactor in the active site being perfectly suited to allowing intramolecular acid-base catalysis.


Subject(s)
Pyruvate Decarboxylase/metabolism , Thiamine Pyrophosphate/chemistry , Thiamine Pyrophosphate/metabolism , Transketolase/metabolism , Apoenzymes/chemistry , Apoenzymes/metabolism , Enzyme Activation , Kinetics , Magnetic Resonance Spectroscopy , Nitrogen Isotopes , Pyruvate Decarboxylase/chemistry , Transketolase/chemistry
8.
Anal Biochem ; 342(1): 126-33, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15958189

ABSTRACT

Acetohydroxy acid synthase (AHAS) and related enzymes catalyze the production of chiral compounds [(S)-acetolactate, (S)-acetohydroxybutyrate, or (R)-phenylacetylcarbinol] from achiral substrates (pyruvate, 2-ketobutyrate, or benzaldehyde). The common methods for the determination of AHAS activity have shortcomings. The colorimetric method for detection of acyloins formed from the products is tedious and does not allow time-resolved measurements. The continuous assay for consumption of pyruvate based on its absorbance at 333 nm, though convenient, is limited by the extremely small extinction coefficient of pyruvate, which results in a low signal-to-noise ratio and sensitivity to interfering absorbing compounds. Here, we report the use of circular dichroism spectroscopy for monitoring AHAS activity. This method, which exploits the optical activity of reaction products, displays a high signal-to-noise ratio and is easy to perform both in time-resolved and in commercial modes. In addition to AHAS, we examined the determination of activity of glyoxylate carboligase. This enzyme catalyzes the condensation of two molecules of glyoxylate to chiral tartronic acid semialdehyde. The use of circular dichroism also identifies the product of glyoxylate carboligase as being in the (R) configuration.


Subject(s)
Acetolactate Synthase/analysis , Acetolactate Synthase/antagonists & inhibitors , Acetolactate Synthase/metabolism , Carboxy-Lyases/metabolism , Circular Dichroism/methods , Escherichia coli/enzymology , Glyoxylates/pharmacology , Lactates/metabolism , Pyruvic Acid/metabolism , Stereoisomerism , Valine/pharmacology
9.
Biochemistry ; 44(16): 6164-79, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15835904

ABSTRACT

The thiamin diphosphate (ThDP)-dependent enzyme indolepyruvate decarboxylase (IPDC) is involved in the biosynthetic pathway of the phytohormone 3-indoleacetic acid and catalyzes the nonoxidative decarboxylation of 3-indolepyruvate to 3-indoleacetaldehyde and carbon dioxide. The steady-state distribution of covalent ThDP intermediates of IPDC reacting with 3-indolepyruvate and the alternative substrates benzoylformate and pyruvate has been analyzed by (1)H NMR spectroscopy. For the first time, we are able to isolate and directly assign covalent intermediates of ThDP with aromatic substrates. The intermediate analysis of IPDC variants is used to infer the involvement of active site side chains and functional groups of the cofactor in distinct catalytic steps during turnover of the different substrates. As a result, three residues (glutamate 468, aspartate 29, and histidine 115) positioned perpendicular to the thiazolium moiety of ThDP are involved in binding of all substrates and decarboxylation of the respective tetrahedral ThDP-substrate adducts. Most likely, interactions of these side chains with the substrate-derived carboxylate account for an optimal orientation of the substrate and/or intermediate in the course of carbon-carbon ligation and decarboxylation supporting the suggested least-motion, maximum overlap mechanism. The active site residue glutamine 383, which is located at the opposite site of the thiazolium nucleus as the "carboxylate pocket" (formed by the Glu-Asp-His triad), is central to the substrate specificity of IPDC, probably through orbital alignment. The Glu51-cofactor proton shuttle is, conjointly with the Glu-Asp-His triad, involved in multiple proton transfer steps, including ylide generation, substrate binding, and product release. Studies with para-substituted benzoylformate substrates demonstrate that the electronic properties of the substrate affect the stabilization or destabilization of the carbanion intermediate or carbanion-like transition state and in that way alter the rate dependence on decarboxylation. In conclusion, general mechanistic principles of catalysis of ThDP-dependent enzymes are discussed.


Subject(s)
Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , Thiamine Pyrophosphate/metabolism , Amino Acid Substitution , Base Sequence , Carboxy-Lyases/genetics , Catalytic Domain/genetics , DNA, Bacterial/genetics , Genetic Variation , Glyoxylates/metabolism , Kinetics , Mandelic Acids , Models, Molecular , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Pyruvates/chemistry , Pyruvates/metabolism , Substrate Specificity , Zymomonas/enzymology , Zymomonas/genetics
10.
J Biol Chem ; 280(22): 21473-82, 2005 Jun 03.
Article in English | MEDLINE | ID: mdl-15802265

ABSTRACT

The residue Glu636 is located near the thiamine diphosphate (ThDP) binding site of the Escherichia coli pyruvate dehydrogenase complex E1 subunit (PDHc-E1), and to probe its function two variants, E636A and E636Q were created with specific activities of 2.5 and 26% compared with parental PDHc-E1. According to both fluorescence binding and kinetic assays, the E636A variant behaved according to half-of-the-sites mechanism with respect to ThDP. In contrast, with the E636Q variant a K(d,ThDP) = 4.34 microM and K(m,ThDP) = 11 microM were obtained with behavior more reminiscent of the parental enzyme. The CD spectra of both variants gave evidence for formation of the 1',4'-iminopyrimidine tautomer on binding of phosphonolactylthiamine diphosphate, a stable analog of the substrate-ThDP covalent complex. Rapid formation of optically active (R)-acetolactate by both variants, but not by the parental enzyme, was observed by CD and NMR spectroscopy. The acetolactate configuration produced by the Glu636 variants is opposite that produced by the enzyme acetolactate synthase and the Asp28-substituted variants of yeast pyruvate decarboxylase, suggesting that the active centers of the two sets of enzymes exhibit different facial selectivity (re or si) vis à vis pyruvate. The tryptic peptide map (mass spectral analysis) revealed that the Glu636 substitution changed the mobility of a loop comprising amino acid residues from the ThDP binding fold. Apparently, the residue Glu636 has important functions both in active center communication and in protecting the active center from undesirable "carboligase" side reactions.


Subject(s)
Acetolactate Synthase/physiology , Escherichia coli/enzymology , Glutamic Acid/chemistry , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Acetolactate Synthase/chemistry , Aspartic Acid/chemistry , Binding Sites , Catalysis , Circular Dichroism , Dose-Response Relationship, Drug , Genetic Variation , Kinetics , Lactates/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Chemical , Models, Molecular , Mutagenesis, Site-Directed , Oxygen/chemistry , Oxygen/metabolism , Peptides/chemistry , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Pyrimidine Nucleosides/chemistry , Spectrometry, Fluorescence , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spectroscopy, Fourier Transform Infrared , Stereoisomerism , Temperature , Thiamine Pyrophosphate/chemistry , Trypsin/pharmacology , Ultraviolet Rays
11.
Biochemistry ; 44(13): 5086-94, 2005 Apr 05.
Article in English | MEDLINE | ID: mdl-15794646

ABSTRACT

In pyruvate oxidase (POX) from Lactobacillus plantarum, valine 265 participates in binding the cofactor FAD and is responsible for the strained conformation of its isoalloxazine moiety that is visible in the crystal structure of POX. The contrasting effects of the conservative amino acid exchange V265A on the enzyme's catalytic properties, cofactor affinity, and protein structure were investigated. The most prominent effect of the exchange was observed in the 2.2 A crystal structure of the mutant POX. While the overall structures of the wild-type and the variant are similar, flavin binding in particular is clearly different. Local disorder at the isoalloxazine binding site prevents modeling of the complete FAD cofactor and two protein loops of the binding site. Only the ADP moiety shows well-defined electron density, indicating an "anchor" function for this part of the molecule. This notion is corroborated by competition experiments where ADP was used to displace FAD from the variant enzyme. Despite the fact that the affinity of FAD binding in the variant is reduced, the catalytic properties are very similar to the wild-type, and the redox potential of the bound flavin is the same for both proteins. The rate of electron transfer toward the flavin during turnover is reduced to one-third compared to the wild-type, but k(cat) remains unchanged. Redox-triggered FTIR difference spectroscopy of free FAD shows the nu(C(10a)=N(1)) band at 1548 cm(-)(1). In POX-V265A, this band is found at 1538 cm(-)(1) and thus shifted less strongly than in wild-type POX where it is found at 1534 cm(-)(1). Taking these observations together, the conservative exchange V265A in POX has a surprisingly small effect on the catalytic properties of the enzyme, whereas the effect on the three-dimensional structure is rather big.


Subject(s)
Flavin-Adenine Dinucleotide/metabolism , Pyruvate Oxidase/chemistry , Pyruvate Oxidase/metabolism , Base Sequence , Catalytic Domain , Crystallography, X-Ray , DNA, Bacterial/genetics , Genetic Variation , Kinetics , Lactobacillus plantarum/enzymology , Lactobacillus plantarum/genetics , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Pyruvate Oxidase/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrophotometry , Spectroscopy, Fourier Transform Infrared , Valine/chemistry
12.
FEBS J ; 272(6): 1326-42, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15752351

ABSTRACT

Transketolase from baker's yeast is a thiamin diphosphate-dependent enzyme in sugar metabolism that reconstitutes with various analogues of the coenzyme. The methylated analogues (4'-methylamino-thiamin diphosphate and N1'-methylated thiamin diphosphate) of the native cofactor were used to investigate the function of the aminopyrimidine moiety of the coenzyme in transketolase catalysis. For the wild-type transketolase complex with the 4'-methylamino analogue, no electron density was found for the methyl group in the X-ray structure, whereas in the complex with the N1'-methylated coenzyme the entire aminopyrimidine ring was disordered. This indicates a high flexibility of the respective parts of the enzyme-bound thiamin diphosphate analogues. In the E418A variant of transketolase reconstituted with N1'-methylated thiamin diphosphate, the electron density of the analogue was well defined and showed the typical V-conformation found in the wild-type holoenzyme [Lindqvist Y, Schneider G, Ermler U, Sundstrom M (1992) EMBO J11, 2373-2379]. The near-UV CD spectrum of the variant E418A reconstituted with N1'-methylated thiamin diphosphate was identical to that of the wild-type holoenzyme, while the CD spectrum of the variant combined with the unmodified cofactor did not overlap with that of the native protein. The activation of the analogues was measured by the H/D-exchange at C2. Methylation at the N1' position of the cofactor activated the enzyme-bound cofactor analogue (as shown by a fast H/D-exchange rate constant). The absorbance changes in the course of substrate turnover of the different complexes investigated (transient kinetics) revealed the stability of the alpha-carbanion/enamine as the key intermediate in cofactor action to be dependent on the functionality of the 4-aminopyrimidine moiety of thiamin diphosphate.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Transketolase/genetics , Transketolase/metabolism , Amino Acid Substitution , Binding Sites , Crystallography, X-Ray , Kinetics , Mutation, Missense , Protein Conformation , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Spectrophotometry, Ultraviolet , Substrate Specificity , Transketolase/chemistry
13.
Proc Natl Acad Sci U S A ; 102(3): 553-8, 2005 Jan 18.
Article in English | MEDLINE | ID: mdl-15640355

ABSTRACT

The thiamin diphosphate (ThDP)-dependent enzyme acetohydroxyacid synthase (AHAS) catalyzes the first common step in branched-chain amino acid biosynthesis. By specific ligation of pyruvate with the alternative acceptor substrates 2-ketobutyrate and pyruvate, AHAS controls the flux through this branch point and determines the relative rates of synthesis of isoleucine, valine, and leucine, respectively. We used detailed NMR analysis to determine microscopic rate constants for elementary steps in the reactions of AHAS II and mutants altered at conserved residues Arg-276, Trp-464, and Met-250. In Arg276Lys, both the condensation of the enzyme-bound hydroxyethyl-ThDP carbanion/enamine (HEThDP) with the acceptor substrates and acetohydroxyacid release are slowed several orders of magnitude relative to the wild-type enzyme. We propose that the interaction of the guanidinium moiety of Arg-264 with the carboxylate of the acceptor ketoacid provides an optimal alignment of substrate and HEThDP orbitals in the reaction trajectory for acceptor ligation, whereas its interaction with the carboxylate of the covalent HEThDP-acceptor adduct plays a similar role in product release. Both Trp-464 and Met-250 affect the acceptor specificity. The high preference for ketobutyrate in the wild-type enzyme is lost in Trp464Leu as a consequence of similar forward rate constants of carboligation and product release for the alternative acceptors. In Met250Ala, the turnover rate is determined by the condensation of HEThDP with pyruvate and release of the acetolactate product, whereas the parallel steps with 2-ketobutyrate are considerably faster. We speculate that the specificity of carboligation and product liberation may be cumulative if the former is not completely committed.


Subject(s)
Acetolactate Synthase/metabolism , Carboxylic Acids/metabolism , Acetolactate Synthase/chemistry , Acetolactate Synthase/genetics , Amino Acids, Branched-Chain/biosynthesis , Binding Sites , Carboxylic Acids/chemistry , Conserved Sequence , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Kinetics , Mutation , Nuclear Magnetic Resonance, Biomolecular , Substrate Specificity
14.
Eur J Biochem ; 271(21): 4189-94, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15511224

ABSTRACT

The influence of substrates on the interaction of apotransketolase with thiamin diphosphate was investigated in the presence of magnesium ions. It was shown that the donor substrates, but not the acceptor substrates, enhance the affinity of the coenzyme either to only one active center of transketolase or to both active centers, but to different degrees in each, resulting in a negative cooperativity for coenzyme binding. In the absence of donor substrate, negative cooperativity is not observed. The donor substrate did not affect the interaction of the apoenzyme with the inactive coenzyme analogue, N3'-pyridyl-thiamin diphosphate. The influence of the donor substrate on the coenzyme-apotransketolase interaction was predicted as a result of formation of the transketolase reaction intermediate 2-(alpha,beta-dihydroxyethyl)-thiamin diphosphate, which exhibited a higher affinity to the enzyme than thiamin diphosphate. The enhancement of thiamin diphosphate's affinity to apotransketolase in the presence of donor substrate is probably one of the mechanisms underlying the substrate-affected transketolase regulation at low coenzyme concentrations.


Subject(s)
Gene Expression Regulation, Enzymologic , Transketolase/chemistry , Transketolase/metabolism , Binding Sites , Buffers , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Ions , Kinetics , Magnesium/chemistry , Magnesium Chloride/chemistry , Models, Chemical , Protein Binding , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Spectrophotometry , Temperature , Thiamine Pyrophosphate/chemistry , Time Factors
15.
Biochemistry ; 43(27): 8652-61, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15236573

ABSTRACT

Acetohydroxy acid synthases (AHAS) are thiamin diphosphate- (ThDP-) and FAD-dependent enzymes that catalyze the first common step of branched-chain amino acid biosynthesis in plants, bacteria, and fungi. Although the flavin cofactor is not chemically involved in the physiological reaction of AHAS, it has been shown to be essential for the structural integrity and activity of the enzyme. Here, we report that the enzyme-bound FAD in AHAS is reduced in the course of catalysis in a side reaction. The reduction of the enzyme-bound flavin during turnover of different substrates under aerobic and anaerobic conditions was characterized by stopped-flow kinetics using the intrinsic FAD absorbance. Reduction of enzyme-bound FAD proceeds with a net rate constant of k' = 0.2 s(-1) in the presence of oxygen and approximately 1 s(-1) under anaerobic conditions. No transient flavin radicals are detectable during the reduction process while time-resolved absorbance spectra are recorded. Reconstitution of the binary enzyme-FAD complex with the chemically synthesized intermediate 2-(hydroxyethyl)-ThDP also results in a reduction of the flavin. These data provide evidence for the first time that the key catalytic intermediate 2-(hydroxyethyl)-ThDP in the carbanionic/enamine form is not only subject to covalent addition of 2-keto acids and an oxygenase side reaction but also transfers electrons to the adjacent FAD in an intramolecular redox reaction yielding 2-acetyl-ThDP and reduced FAD. The detection of the electron transfer supports the idea of a common ancestor of acetohydroxy acid synthase and pyruvate oxidase, a homologous ThDP- and FAD-dependent enzyme that, in contrast to AHASs, catalyzes a reaction that relies on intercofactor electron transfer.


Subject(s)
Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Thiamine Pyrophosphate/metabolism , Catalysis , Electron Transport , Flavin-Adenine Dinucleotide/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Oxygen/metabolism , Pyruvic Acid/metabolism , Spectrum Analysis , Thiamine Pyrophosphate/analogs & derivatives , Thiamine Pyrophosphate/chemistry
16.
Biochemistry ; 42(50): 14814-21, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14674755

ABSTRACT

Flavin adenine dinucleotide (FAD) and three different flavoproteins in aqueous solution were subjected to redox-triggered Fourier transform infrared difference spectroscopy. The acquired vibrational spectra show a great number of positive and negative peaks, pertaining to the oxidized and reduced state of the molecule, respectively. Density functional theory calculations on the B3LYP/6-31G(d) level were employed to assign several of the observed bands to vibrational modes of the isoalloxazine moiety of the flavin cofactor in both its oxidized and, for the first time, its reduced state. Prominent modes measured for oxidized FAD include nu(C(4)=O) and nu(C(2)=O) at 1716 and 1674 cm(-1), respectively, nu(C(4a)=N(5)) at 1580 cm(-1), and nu(C(10a)=N(1)) at 1548 cm(-1). Measured modes of the reduced form of FAD include nu(C(2)=O) at 1692 cm(-1), nu(C(4)=O) at 1634 cm(-1), and nu(C(4a)=C(10a)) at 1600 cm(-1). While the overall shape of the enzyme spectra is similar to the shape of the spectrum of free FAD, there are numerous differences in detail. In particular, the nu(C=N) modes of the flavin exhibit frequency shifts in the protein-bound form, most prominently for pyruvate oxidase where nu(C(10a)=N(1)) downshifts by 14 cm(-1) to 1534 cm(-1). The significance of this shift and a possible explanation in connection with the bent conformation of the flavin cofactor in this enzyme are discussed.


Subject(s)
Flavin-Adenine Dinucleotide/chemistry , Flavoproteins/chemistry , Animals , Aspergillus niger/enzymology , Binding Sites , D-Amino-Acid Oxidase/chemistry , D-Amino-Acid Oxidase/metabolism , Electrochemistry , Flavin-Adenine Dinucleotide/metabolism , Flavoproteins/metabolism , Glucose Oxidase/chemistry , Glucose Oxidase/metabolism , Oxidation-Reduction , Pyruvate Oxidase/chemistry , Pyruvate Oxidase/metabolism , Solutions , Spectroscopy, Fourier Transform Infrared/methods , Swine , Water/chemistry
17.
J Biol Chem ; 278(41): 39280-6, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-12882981

ABSTRACT

The Crabtree-negative yeast Kluyveromyces lactis is capable of adjusting its glycolytic flux to the requirements of respiration by tightly regulating glucose uptake. RAG5 encoding the only glucose and fructose phosphorylating enzyme present in K. lactis is required for the up-regulation of glucose transport and also for glucose repression. To understand the significance of the molecular identity and specific function(s) of the corresponding kinase to glucose signaling, RAG5 was overexpressed and its gene product KlHxk1 (Rag5p) isolated and characterized. Stopped-flow kinetics and sedimentation analysis indicated a monomer-homodimer equilibrium of KlHxk1 in a condition of catalysis, i.e. in the presence of substrates and products. The kinetic constants of ATP-dependent glucose phosphorylation identified a 53-kDa monomer as the high affinity/high activity form of the novel enzyme for both glycolytic substrates suggesting a control of glucose phosphorylation at the level of dimer formation and dissociation. In contrast to the highly homologous hexokinase isoenzyme 2 of Saccharomyces cerevisiae (ScHxk2), KlHxk1 was not inhibited by free ATP in a physiological range of nucleotide concentration. Mass spectrometric sequencing of tryptic peptides of KlHxk1 identified unmodified serine at amino acid position 156. The corresponding amino acid in ScHxk2 is serine 157, which represents the autophosphorylation-inactivation site. KlHxk1 did not display, however, the typical pattern of inactivation under the respective in vitro conditions and maintained a high residual glucose phosphorylating activity. The biophysical and functional data are discussed with respect to a possible regulatory role of KlHxk1 in glucose metabolism and signaling in K. lactis.


Subject(s)
Hexokinase/genetics , Hexokinase/metabolism , Kluyveromyces/enzymology , Kluyveromyces/genetics , Amino Acid Sequence , Catalytic Domain , Genes, Fungal , Glucose/metabolism , Hexokinase/chemistry , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Molecular Sequence Data , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Signal Transduction
18.
Biochemistry ; 42(26): 7885-91, 2003 Jul 08.
Article in English | MEDLINE | ID: mdl-12834340

ABSTRACT

Enzymic catalysis proceeds via intermediates formed in the course of substrate conversion. Here, we directly detect key intermediates in thiamin diphosphate (ThDP)-dependent enzymes during catalysis using (1)H NMR spectroscopy. The quantitative analysis of the relative intermediate concentrations allows the determination of the microscopic rate constants of individual catalytic steps. As demonstrated for pyruvate decarboxylase (PDC), this method, in combination with site-directed mutagenesis, enables the assignment of individual side chains to single steps in catalysis. In PDC, two independent proton relay systems and the stereochemical control of the enzymic environment account for proficient catalysis proceeding via intermediates at carbon 2 of the enzyme-bound cofactor. The application of this method to other ThDP-dependent enzymes provides insight into their specific chemical pathways.


Subject(s)
Pyruvate Decarboxylase/chemistry , Thiamine Pyrophosphate/chemistry , Zymomonas/enzymology , Binding Sites/genetics , Catalysis , Magnetic Resonance Spectroscopy , Models, Molecular , Mutagenesis, Site-Directed , Protons , Pyruvate Decarboxylase/genetics , Pyruvate Decarboxylase/metabolism
19.
Eur J Biochem ; 270(10): 2312-21, 2003 May.
Article in English | MEDLINE | ID: mdl-12752451

ABSTRACT

The thiamin diphosphate-dependent enzyme indolepyruvate decarboxylase catalyses the formation of indoleacetaldehyde from indolepyruvate, one step in the indolepyruvate pathway of biosynthesis of the plant hormone indole-3-acetic acid. The crystal structure of this enzyme from Enterobacter cloacae has been determined at 2.65 A resolution and refined to a crystallographic R-factor of 20.5% (Rfree 23.6%). The subunit of indolepyruvate decarboxylase contains three domains of open alpha/beta topology, which are similar in structure to that of pyruvate decarboxylase. The tetramer has pseudo 222 symmetry and can be described as a dimer of dimers. It resembles the tetramer of pyruvate decarboxylase from Zymomonas mobilis, but with a relative difference of 20 degrees in the angle between the two dimers. Active site residues are highly conserved in indolepyruvate/pyruvate decarboxylase, suggesting that the interactions with the cofactor thiamin diphosphate and the catalytic mechanisms are very similar. The substrate binding site in indolepyruvate decarboxylase contains a large hydrophobic pocket which can accommodate the bulky indole moiety of the substrate. In pyruvate decarboxylases this pocket is smaller in size and allows discrimination of larger vs. smaller substrates. In most pyruvate decarboxylases, restriction of cavity size is due to replacement of residues at three positions by large, hydrophobic amino acids such as tyrosine or tryptophan.


Subject(s)
Carboxy-Lyases/chemistry , Enterobacter/enzymology , Indoleacetic Acids/metabolism , Thiamine Pyrophosphate/pharmacology , Amino Acid Sequence , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Escherichia coli/metabolism , Indoleacetic Acids/chemistry , Kinetics , Magnesium/metabolism , Models, Chemical , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity , Thiamine Pyrophosphate/chemistry
20.
Eur J Biochem ; 270(10): 2322-31, 2003 May.
Article in English | MEDLINE | ID: mdl-12752452

ABSTRACT

Enterobacter cloacae, isolated from the rhizosphere of cucumbers, produces large amounts of indole-3-acetic acid. Indolepyruvate decarboxylase, the key enzyme in the biosynthetic pathway of indole-3-acetic acid, catalyses the formation of indole-3-acetaldehyde and carbon dioxide from indole-3-pyruvic acid. The enzyme requires the cofactors thiamine diphosphate and magnesium ions for catalytic activity. Recombinant indolepyruvate decarboxylase was purified from the host Escherichia coli strain JM109. Specificity of the enzyme for the substrates indole-3-pyruvic acid, pyruvic acid, benzoylformic acid, and seven benzoylformic acid analogues was investigated using a continuous optical assay. Stopped-flow kinetic data showed no indication for substrate activation in the decarboxylation reaction of indole-3-pyruvic acid, pyruvic acid or benzoylformic acid. Size exclusion chromatography and small angle X-ray solution scattering experiments suggested the tetramer as the catalytically active state and a pH-dependent subunit association equilibrium. Analysis of the kinetic constants of the benzoylformic acid analogues according to Hansch et al. [Hansch, C., Leo, A., Unger, S.H., Kim, K.H., Nikaitani, D & Lien, E.J. (1973) J. Med. Chem.16, 1207-1216] and comparison with indole-3-pyruvic acid conversion by pyruvate decarboxylases from Saccharomyces cerevisiae and Zymomonas mobilis provided some insight into the catalytic mechanism of indolepyruvate decarboxylase.


Subject(s)
Carboxy-Lyases/chemistry , Enterobacter/enzymology , Indoleacetic Acids/chemistry , Thiamine Pyrophosphate/pharmacology , Binding Sites , Catalysis , Chromatography , Crystallography, X-Ray , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Kinetics , Magnetic Resonance Spectroscopy , Recombinant Proteins/metabolism , Scattering, Radiation , Structure-Activity Relationship , Substrate Specificity , Thiamine Pyrophosphate/chemistry , Time Factors , Tryptophan/chemistry , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...