Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters










Publication year range
1.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 65(Pt 11): 1071-7, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19923721

ABSTRACT

Unlike mammals, bacteria encode enzymes that synthesize branched-chain amino acids. The pyridoxal 50-phosphate-dependent transaminase performs the final biosynthetic step in these pathways, converting keto acid precursors into -amino acids. The branched-chain amino-acid transaminase from Mycobacterium tuberculosis (MtIlvE) has been crystallized and its structure has been solved at 1.9 angstrom resolution. The MtIlvE monomer is composed of two domains that interact to form the active site. The biologically active form of IlvE is a homodimer in which each monomer contributes a substrate-specificity loop to the partner molecule. Additional substrate selectivity may be imparted by a conserved N-terminal Phe30 residue, which has previously been observed to shield the active site in the type IV fold homodimer. The active site of MtIlvE contains density corresponding to bound PMP, which is likely to be a consequence of the presence of tryptone in the crystallization medium. Additionally, two cysteine residues are positioned at the dimer interface for disulfide-bond formation under oxidative conditions. It is unknown whether they are involved in any regulatory activities analogous to those of the human mitochondrial branched-chain amino-acid transaminase.


Subject(s)
Bacterial Proteins/chemistry , Mycobacterium tuberculosis/enzymology , Protein Structure, Quaternary , Protein Structure, Tertiary , Transaminases/chemistry , Amino Acid Sequence , Amino Acids, Branched-Chain/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Sequence Alignment , Transaminases/genetics , Transaminases/metabolism
2.
Biochemistry ; 47(50): 13326-35, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19053270

ABSTRACT

Mycobacterium smegmatis MshC catalyzes the ATP-dependent condensation of GlcN-Ins and l-cysteine to form l-Cys-GlcN-Ins, the penultimate step in mycothiol biosynthesis. Attempts to crystallize the native, full-length MshC have been unsuccessful. However, incubation of the enzyme with the cysteinyl adenylate analogue, 5'-O-[N-(l-cysteinyl)-sulfamonyl]adenosine (CSA), followed by a 24-h limited trypsin proteolysis yielded an enzyme preparation that readily crystallized. The three-dimensional structure of MshC with CSA bound in the active site was solved and refined to 1.6 A. The refined structure exhibited electron density corresponding to the entire 47 kDa MshC molecule, with the exception of the KMSKS loop (residues 285-297), a loop previously implicated in the formation of the adenylate in related tRNA synthases. The overall tertiary fold of MshC is similar to that of cysteinyl-tRNA synthetase, with a Rossmann fold catalytic domain. The interaction of the thiolate of CSA with a zinc ion at the base of the active site suggests that the metal ion participates in amino acid binding and discrimination. A number of active site residues were observed to interact with the ligand, suggesting a role in substrate binding and catalysis. Analysis utilizing modeling of the proteolyzed loop and GlcN-Ins docking, as well as the examination of sequence conservation in the active site suggests similarities and differences between cysteinyl-tRNA synthetases and MshC in recognition of the substrates for their respective reactions.


Subject(s)
Bacterial Proteins/chemistry , Carbon-Nitrogen Ligases/chemistry , Cysteine/biosynthesis , Cysteine/chemistry , Glycopeptides/biosynthesis , Glycopeptides/chemistry , Inositol/biosynthesis , Inositol/chemistry , Mycobacterium smegmatis/enzymology , Bacterial Proteins/biosynthesis , Bacterial Proteins/metabolism , Binding Sites , Carbon-Nitrogen Ligases/biosynthesis , Carbon-Nitrogen Ligases/metabolism , Conserved Sequence , Crystallization , Crystallography, X-Ray , Cysteine/metabolism , Glycopeptides/metabolism , Hydrolysis , Inositol/metabolism , Protein Binding , Protein Structure, Secondary , Substrate Specificity , Trypsin/metabolism
3.
Biochem Soc Trans ; 31(Pt 3): 520-2, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12773148

ABSTRACT

Resistance to antibiotics used in the treatment of bacterial infections is an expanding clinical problem. Aminoglycosides, one of the oldest classes of natural product antibiotics, exert their bactericidal effect as the result of inhibiting bacterial protein synthesis by binding to the acceptor site of the 30 S ribosomal subunit. The most common mechanism of clinical resistance to aminoglycosides results from the expression of enzymes that covalently modify the aminoglycoside. We will discuss the enzymology and structure of two representative chromosomally encoded aminoglycoside N-acetyltransferases, Mycobacterium tuberculosis AAC(2')-Ic and Salmonella enterica AAC(6')-Iy, and speculate about their possible physiological function and substrates.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Acetyltransferases/chemistry , Acetyltransferases/genetics , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Carbohydrate Sequence , Drug Screening Assays, Antitumor , Escherichia coli/enzymology , Escherichia coli/genetics , Molecular Sequence Data , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Protein Conformation
4.
Curr Pharm Biotechnol ; 3(3): 197-225, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12164478

ABSTRACT

Tuberculosis resurged in the late 1980s and now kills more than 2 million people a year. The reemergence of tuberculosis as a potential public health threat, the high susceptibility of human immunodeficiency virus-infected persons to the disease, and the proliferation of multi-drug-resistant (MDR) strains have created much scientific interest in developing new antimycobacterial agents to both treat Mycobacterium tuberculosis strains resistant to existing drugs, and shorten the duration of short-course treatment to improve patient compliance. Bacterial cell-wall biosynthesis is a proven target for new antibacterial drugs. Mycolic acids, which are key components of the mycobacterial cell wall, are alpha-alkyl, beta-hydroxy fatty acids, with a species-dependent saturated "short" arm of 20-26 carbon atoms and a "long" meromycolic acid arm of 50-60 carbon atoms. The latter arm is functionalized at regular intervals by cyclopropyl, alpha-methyl ketone, or alpha-methyl methylethers groups. The mycolic acid biosynthetic pathway has been proposed to involve five distinct stages: (i) synthesis of C20 to C26 straight-chain saturated fatty acids to provide the alpha-alkyl branch; (ii) synthesis of the meromycolic acid chain to provide the main carbon backbone, (iii) modification of this backbone to introduce other functional groups; (iv) the final Claisen-type condensation step followed by reduction; and (v) various mycolyltransferase processes to cellular lipids. The drugs shown to inhibit mycolic acid biosynthesis are isoniazid, ethionamide, isoxyl, thiolactomycin, and triclosan. In addition, pyrazinamide was shown to inhibit fatty acid synthase type I which, in turn, provides precursors for fatty acid elongation to long-chain mycolic acids by fatty acid synthase II. Here we review the biosynthesis of mycolic acids and the mechanism of action of antimicrobial agents that act upon this pathway. In addition, we describe molecular modeling studies on InhA, the bona-fide target for isoniazid, which should improve our understanding of the amino acid residues involved in the enzyme's mechanism of action and, accordingly, provide a rational approach to the design of new drugs.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Mycolic Acids/antagonists & inhibitors , Antitubercular Agents/chemistry , Antitubercular Agents/therapeutic use , Cell Wall/metabolism , Humans , Mycobacterium tuberculosis/metabolism , Mycolic Acids/chemistry , Structure-Activity Relationship , Tuberculosis/drug therapy , Tuberculosis/prevention & control
5.
J Biol Chem ; 276(49): 45876-81, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11590162

ABSTRACT

The chromosomally encoded aminoglycoside N-acetyltransferase, AAC(2')-Ic, of Mycobacterium tuberculosis has a yet unidentified physiological function. The aac(2')-Ic gene was cloned and expressed in Escherichia coli, and AAC(2')-Ic was purified. Recombinant AAC(2')-Ic was a soluble protein of 20,000 Da and acetylated all aminoglycosides substrates tested in vitro, including therapeutically important antibiotics. Acetyl-CoA was the preferred acyl donor. The enzyme, in addition to acetylating aminoglycosides containing 2'-amino substituents, also acetylated kanamycin A and amikacin that contain a 2'-hydroxyl substituent, although with lower activity, indicating the capacity of the enzyme to perform both N-acetyl and O-acetyl transfer. The enzyme exhibited "substrate activation" with many aminoglycoside substrates while exhibiting Michaelis-Menten kinetics with others. Kinetic studies supported a random kinetic mechanism for AAC(2')-Ic. Comparison of the kinetic parameters of different aminoglycosides suggested that their hexopyranosyl residues and, to a lesser extent, the central aminocyclitol residue carry the major determinants of substrate affinity.


Subject(s)
Acetyltransferases/genetics , Chromosomes, Bacterial , Mycobacterium tuberculosis/genetics , Acetyltransferases/isolation & purification , Acetyltransferases/metabolism , Base Sequence , Cloning, Molecular , DNA Primers , Kinetics , Mycobacterium tuberculosis/enzymology , Polymerase Chain Reaction , Substrate Specificity
6.
Biochemistry ; 40(43): 12904-12, 2001 Oct 30.
Article in English | MEDLINE | ID: mdl-11669627

ABSTRACT

Pantothenate synthetase (EC 6.3.2.1), encoded by the panC gene, catalyzes the essential ATP-dependent condensation of D-pantoate and beta-alanine to form pantothenate in bacteria, yeast and plants. Pantothenate synthetase from Mycobacterium tuberculosis was expressed in E. coli, purified to homogeneity, and found to be a homodimer with a subunit molecular mass of 33 kDa. Initial velocity, product, and dead-end inhibition studies showed the kinetic mechanism of pantothenate synthetase to be Bi Uni Uni Bi Ping Pong, with ATP binding followed by D-pantoate binding, release of PP(i), binding of beta-alanine, followed by the release of pantothenate and AMP. Michaelis constants were 0.13, 0.8, and 2.6 mM for D-pantoate, beta-alanine, and ATP, respectively, and the turnover number, k(cat), was 3.4 s(-1). The formation of pantoyl adenylate, suggested as a key intermediate by the kinetic mechanism, was confirmed by (31)P NMR spectroscopy of [(18)O]AMP produced from (18)O transfer using [carboxyl-(18)O]pantoate. Single-turnover reactions for the formation of pyrophosphate and pantothenate were determined using rapid quench techniques, and indicated that the two half-reactions occurred with maximum rates of 1.3 +/- 0.3 and 2.6 +/- 0.3 s(-)(1), respectively, consistent with pantoyl adenylate being a kinetically competent intermediate in the pantothenate synthetase reaction. These data also suggest that both half-reactions are partially rate-limiting. Reverse isotope exchange of [(14)C]-beta-alanine into pantothenate in the presence of AMP was observed, indicating the reversible formation of the pantoyl adenylate intermediate from products.


Subject(s)
Mycobacterium tuberculosis/enzymology , Peptide Synthases/chemistry , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Alanine/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Hydroxybutyrates/chemistry , Hydroxybutyrates/metabolism , Ions/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Models, Chemical , Oxygen/metabolism , Peptide Synthases/genetics , Peptide Synthases/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Temperature , Time Factors
7.
Biochemistry ; 40(38): 11353-63, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11560483

ABSTRACT

The gene encoding dihydrolipoamide dehydrogenase from Mycobacterium tuberculosis, Rv0462, was expressed in Escherichia coli and the protein purified to homogeneity. The 49 kDa polypeptide forms a homodimer containing one tightly bound molecule of FAD/monomer. The results of steady-state kinetic analyses using several reduced pyridine nucleotide analogs and a variety of electron acceptors, and the ability of the enzyme to catalyze the transhydrogenation of NADH and thio-NAD(+) in the absence of D,L-lipoamide, demonstrated that the enzyme uses a ping-pong kinetic mechanism. Primary deuterium kinetic isotope effects on V and V/K at pH 7.5 using NADH deuterated at the C(4)-proS position of the nicotinamide ring are small [(D)(V/K)(NADH) = 1.12 +/- 0.15, (D)V(app) = 1.05 +/- 0.07] when D,L-lipoamide is the oxidant but large and equivalent [(D)(V/K)(NADH) = (D)V = 2.95 +/- 0.03] when 5-hydroxy-1,4-naphthoquinone is the oxidant. Solvent deuterium kinetic isotope effects at pH 5.8, using APADH as the reductant, are inverse with (D)(V/K)(APADH) = 0.73 +/- 0.03, (D)(V/K)(Lip(S))2 = 0.77 +/- 0.03, and (D)V(app) = 0.77 +/- 0.01. Solvent deuterium kinetic isotope effects with 4,4-dithiopyridine (DTP), the 4-thiopyridone product of which requires no protonation, are also inverse with (D)(V/K)(APADH) = 0.75 +/- 0.06, (D)(V/K)(DTP) = 0.71 +/- 0.02, and (D)V(app) = 0.56 +/- 0.15. All proton inventories were linear, indicating that a single proton is being transferred in the solvent isotopically sensitive step. Taken together, these results suggest that (1) the reductive half-reaction (hydride transfer from NADH to FAD) is rate limiting when a quinone is the oxidant, and (2) deprotonation of enzymic thiols, most likely Cys(46) and Cys(41), limits the reductive and oxidative half-reactions, respectively, when D,L-lipoamide is the oxidant.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Dihydrolipoamide Dehydrogenase/genetics , Dihydrolipoamide Dehydrogenase/metabolism , Genes, Bacterial , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Amino Acid Sequence , Cloning, Molecular , Deuterium , Dihydrolipoamide Dehydrogenase/chemistry , Escherichia coli/genetics , Flavin-Adenine Dinucleotide/metabolism , Kinetics , Models, Chemical , NAD/analogs & derivatives , NAD/metabolism , Oxidation-Reduction , Protein Conformation , Pseudomonas putida/enzymology , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Solvents , Spectrophotometry
8.
Biochemistry ; 40(28): 8196-203, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11444965

ABSTRACT

Purine salvage pathways are predicted to be present from the genome sequence of Mycobacterium tuberculosis. The M. tuberculosis deoD gene encodes a presumptive purine nucleoside phosphorylase (PNP). The gene was cloned, expressed, purified, and found to exhibit PNP activity. Purified M. tuberculosis PNP is trimeric, similar to mammalian PNP's but unlike the hexameric Escherichia coli enzyme. Immucillin-H is a rationally designed analogue of the transition state that has been shown to be a potent inhibitor of mammalian PNP's. This inhibitor also exhibits slow-onset inhibition of M. tuberculosis PNP with a rapid, reversible inhibitor binding (K(i) of 2.2 nM) followed by an overall dissociation constant (K(i)) of 28 pM, yielding a K(m)/K(i) value of 10(6). Time-dependent tight binding of the inhibitor occurs with a rate of 0.1 s(-)(1), while relaxation of the complex is slower at 1.4 x 10(-)(3) s(-)(1). The pH dependence of the K(i) value of immucillin-H to the M. tuberculosis PNP suggests that the inhibitor binds as the neutral, unprotonated form that is subsequently protonated to generate the tight-binding species. The M. tuberculosis enzyme demonstrates independent and equivalent binding of immucilin-H at each of the three catalytic sites, unlike mammalian PNP. Analysis of the components of immucillin-H confirms that the inhibition gains most of its binding energy from the 9-deazahypoxanthine group (K(is) of 0.39 microM) while the 1,4-dideoxy-1,4-iminoribitol binds weakly (K(is) of 2.9 mM). Double-inhibition studies demonstrate antagonistic binding of 9-deazahypoxanthine and iminoribitol (beta = 13). However, the covalent attachment of these two components in immucillin-H increases equilibrium binding affinity by a factor of >14 000 (28 pM vs 0.39 microM) compared to 9-deazahypoxanthine alone, and by a factor of >10(8) compared to iminoribitol alone (28 pM vs 2.9 mM), from initial velocity measurements. The structural basis for M. tuberculosis PNP inhibition by immucillin-H and by its component parts is reported in the following paper [Shi, W., Basso, L. A., Santos, D. S., Tyler, P. C., Furneaux, R. H., Blanchard, J. S., Almo, S. C., and Schramm, V. L. (2001) Biochemistry 40, 8204-8215].


Subject(s)
Enzyme Inhibitors/chemistry , Mycobacterium tuberculosis/enzymology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/chemistry , Pyrimidinones/chemistry , Pyrroles/chemistry , Binding, Competitive , Catalysis , Cloning, Molecular , Gene Expression Regulation, Bacterial , Hydrogen-Ion Concentration , Kinetics , Molecular Weight , Purine Nucleosides , Purine-Nucleoside Phosphorylase/biosynthesis , Purine-Nucleoside Phosphorylase/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
9.
Biochemistry ; 40(28): 8204-15, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11444966

ABSTRACT

A structural genomics comparison of purine nucleoside phosphorylases (PNPs) indicated that the enzyme encoded by Mycobacterium tuberculosis (TB-PNP) resembles the mammalian trimeric structure rather than the bacterial hexameric PNPs. The crystal structure of M. tuberculosis PNP in complex with the transition-state analogue immucillin-H (ImmH) and inorganic phosphate was solved at 1.75 A resolution and confirms the trimeric structure. Binding of the inhibitor occurs independently at the three catalytic sites, unlike mammalian PNPs which demonstrate negative cooperativity in ImmH binding. Reduced subunit interface contacts for TB-PNP, compared to the mammalian enzymes, correlate with the loss of the cooperative inhibitor binding. Mammalian and TB-PNPs both exhibit slow-onset inhibition and picomolar dissociation constants for ImmH. The structure supports a catalytic mechanism of reactant destabilization by neighboring group electrostatic interactions, transition-state stabilization, and leaving group activation. Despite an overall amino acid sequence identity of 33% between bovine and TB-PNPs and almost complete conservation in active site residues, one catalytic site difference suggests a strategy for the design of transition-state analogues with specificity for TB-PNP. The structure of TB-PNP was also solved to 2.0 A with 9-deazahypoxanthine (9dHX), iminoribitol (IR), and PO(4) to reconstruct the ImmH complex with its separate components. One subunit of the trimer has 9dHX, IR, and PO(4) bound, while the remaining two subunits contain only 9dHX. In the filled subunit, 9dHX retains the contacts found in the ImmH complex. However, the region of IR that corresponds to the oxocarbenium ion is translocated in the direction of the reaction coordinate, and the nucleophilic phosphate rotates away from the IR group. Loose packing of the pieces of ImmH in the catalytic site establishes that covalent connectivity in ImmH is required to achieve the tightly bound complex.


Subject(s)
Enzyme Inhibitors/chemistry , Mycobacterium tuberculosis/enzymology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/chemistry , Pyrimidinones/chemistry , Pyrroles/chemistry , Actinomycetales/enzymology , Animals , Binding Sites , Catalysis , Cattle , Enzyme Stability , Escherichia coli/enzymology , Macromolecular Substances , Models, Molecular , Phosphates/chemistry , Protein Conformation , Purine Nucleosides
10.
Biochim Biophys Acta ; 1545(1-2): 67-77, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11342032

ABSTRACT

Tuberculosis (TB) remains a leading cause of infectious disease in the world today and therapies developed over the last forty years are becoming increasingly ineffective against resistant strains of Mycobacterium tuberculosis. In an effort to explore new mechanisms for drug development, we have investigated the enzymes of the diaminopimelate biosynthetic pathway as potential targets. Specifically, dihydrodipicolinate reductase, the essential gene product of dapB, was screened for novel inhibitors. Inhibitors were identified both by a molecular modeling approach which utilized the available crystal structure of the enzyme with an inhibitor bound at the active site as well as by more conventional screening strategies. The resulting compounds contain a number of structural motifs and were all found to be competitive with respect to the DHDP substrate. The K(i) values for the inhibitors range from 10 to 90 microM. The molecular modeling approach was very effective in identifying novel inhibitors of the enzyme. These compounds were obtained at a higher frequency based on the number of compounds analyzed than those inhibitors discovered via conventional screening. However, conventional screening proved beneficial in identifying compounds with greater structural diversity.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/antagonists & inhibitors , Binding Sites/drug effects , Diaminopimelic Acid/metabolism , Dihydrodipicolinate Reductase , Drug Design , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Kinetics , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Protein Conformation , Sulfonamides/pharmacology
11.
Biochemistry ; 40(17): 5119-26, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11318633

ABSTRACT

The recent identification of the enzyme in Mycobacterium tuberculosis that catalyzes the NADPH-dependent reduction of the unique low molecular weight disulfide mycothione, mycothione reductase, has led us to examine the mechanism of catalysis in greater detail. The pH dependence of the kinetic parameters V and V/K for NADPH, NADH, and an active analogue of mycothione disulfide, des-myo-inositol mycothione disulfide, has been determined. An analysis of the pH profiles has allowed the tentative assignment of catalytically significant residues crucial to the mechanism of disulfide reduction, namely, the His444-Glu449 ion pair and Cys39. Solvent kinetic isotope effects were observed on V and V/K(DIMSSM), yielding values of 1.7 +/- 0.2 and 1.4 +/- 0.2, respectively, but not on V/K(NADPH). Proton inventory studies (V versus mole fraction of D(2)O) were linear, indicative of a single proton transfer in a solvent isotopically sensitive step. Steady-state primary deuterium kinetic isotope effects on V have been determined using NADPH and NADH, yielding values of 1.27 +/- 0.03 and 1.66 +/- 0.14, respectively. The pre-steady-state primary deuterium kinetic isotope effect on enzyme reduction has values of 1.82 +/- 0.04 and 1.59 +/- 0.06 for NADPH and NADH, respectively. The steady-state primary deuterium kinetic isotope effect using NADH coincide with that obtained under single turnover conditions, suggesting the complete expression of the intrinsic primary kinetic isotope effect. Rapid reaction studies on the reductive half-reaction using NADPH and NADH yielded maximal rates of 129 +/- 2 and 20 +/- 1 s(-1), respectively, while similar studies of the oxidation of the two-electron reduced enzyme by mycothiol disulfide yielded a maximum rate of 190 +/- 10 s(-1). These data suggest a unique flavoprotein disulfide mechanism in which the rate of the oxidative half-reaction is slightly faster than the rate of the reductive half-reaction.


Subject(s)
Deuterium/metabolism , Mycobacterium tuberculosis/enzymology , Oxidoreductases/metabolism , Deuterium/chemistry , Disulfides/chemistry , Hydrogen-Ion Concentration , Kinetics , NAD/chemistry , NADP/chemistry , Oxidation-Reduction , Oxidoreductases/chemistry , Protons , Solvents
12.
Biochemistry ; 40(12): 3700-9, 2001 Mar 27.
Article in English | MEDLINE | ID: mdl-11297438

ABSTRACT

The chromosomally encoded aminoglycoside N-acetyltransferase, AAC(6')-Iy, from Salmonella enterica confers resistance toward a number of aminoglycoside antibiotics. The structural gene was cloned and expressed and the purified enzyme existed in solution as a dimer of ca. 17 000 Da monomers. Acetyl-CoA was the preferred acyl donor, and most therapeutically important aminoglycosides were substrates for acetylation. Exceptions are those aminoglycosides that possess a 6'-hydroxyl substituent (e.g., lividomycin). Thus, the enzyme exhibited regioselective and exclusive acetyltransferase activity to 6'-amine-containing aminoglycosides. The enzyme exhibited Michaelis-Menten kinetics for some aminoglycoside substrates but "substrate activation" with others. Kinetic studies supported a random kinetic mechanism for the enzyme. The enzyme was inactivated by iodoacetamide in a biphasic manner, with half of the activity being lost rapidly and the other half more slowly. Tobramycin, but not acetyl-CoA, protected against inactivation. Each of the three cysteine residues (C70, C109, C145) in the wild-type enzyme were carboxamidomethylated by iodoacetamide. Cysteine 109 in AAC(6')-Iy is conserved in 12 AAC(6') enzyme sequences of the major class I subfamily. Surprisingly, mutation of this residue to alanine neither abolished activity nor altered the biphasic inactivation by iodoacetamide. The maximum velocity and V/K values for a number of aminoglycosides were elevated in this single mutant, and the kinetic behavior of substrates exhibiting linear vs nonlinear kinetics was reversed. Cysteine 70 in AAC(6')-Iy is either a cysteine or a threonine residue in all 12 AAC(6') enzymes of the major class I subfamily. The double mutant, C109A/C70A, was not inactivated by iodoacetamide. The double mutant exhibited large increases in the K(m) values for both acetyl-CoA and aminoglycoside substrates, and all aminoglycoside substrates exhibited Michaelis-Menten kinetics. Solvent kinetic isotope effects on V/K were normal for the WT enzyme and inverse for the double mutant. We discuss a chemical mechanism and the likely rate-limiting steps for both the wild-type and mutant forms of the enzyme.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/genetics , Chromosomes, Bacterial/enzymology , Chromosomes, Bacterial/genetics , Salmonella enterica/enzymology , Salmonella enterica/genetics , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/isolation & purification , Amino Acid Sequence , Aminoglycosides , Anti-Bacterial Agents/pharmacology , Deuterium Oxide/chemistry , Drug Resistance, Microbial/genetics , Enzyme Activation/drug effects , Iodoacetamide/pharmacology , Kinetics , Mass Spectrometry , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Salmonella enterica/drug effects , Solvents , Substrate Specificity/genetics
13.
Org Lett ; 2(24): 3857-60, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11101437

ABSTRACT

[reaction: see text] Vinylogous amides 5 and 6 have been synthesized from L-propargyl glycine and tested against diaminopimelate (DAP) enzymes involved in bacterial lysine biosynthesis. Both are reversible inhibitors of DAP D-dehydrogenase and DAP epimerase with IC(50) values in the 500 microM range. Compound 5 shows competitive inhibition against the L-dihydrodipicolinate (DHDP) reductase with a K(i) value of 32 microM, which is comparable to the planar dipicolinate 16 (K(i) = 26 microM), the best known inhibitor of the enzyme.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Bacteria/chemistry , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Lysine/biosynthesis , Oxidoreductases Acting on CH-CH Group Donors , Amides/chemical synthesis , Amides/chemistry , Amino Acid Isomerases/antagonists & inhibitors , Amino Acid Oxidoreductases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacteria/metabolism , Diaminopimelic Acid/chemistry , Dihydrodipicolinate Reductase , Enzyme Inhibitors/chemistry , Oxidoreductases/antagonists & inhibitors
14.
Protein Sci ; 9(10): 2034-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11106178

ABSTRACT

The three-dimensional (3D) structure of Corynebacterium glutamicum diaminopimelate D-dehydrogenase in a ternary complex with NADPH and L-2-amino-6-methylene-pimelate has been solved and refined to a resolution of 2.1 A. L-2-Amino-6-methylene-pimelate was recently synthesized and shown to be a potent competitive inhibitor (5 microM) vs. meso-diaminopimelate of the Bacillus sphaericus dehydrogenase (Sutherland et al., 1999). Diaminopimelate dehydrogenase catalyzes the reversible NADP+ -dependent oxidation of the D-amino acid stereocenter of mesodiaminopimelate, and is the only enzyme known to catalyze the oxidative deamination of a D-amino acid. The enzyme is involved in the biosynthesis of meso-diaminopimelate and L-lysine from L-aspartate, a biosynthetic pathway of considerable interest because it is essential for growth of certain bacteria. The dehydrogenase is found in a limited number of species of bacteria, as opposed to the alternative succinylase and acetylase pathways that are widely distributed in bacteria and plants. The structure of the ternary complex reported here provides a structural rationale for the nature and potency of the inhibition exhibited by the unsaturated L-2-amino-6-methylene-pimelate against the dehydrogenase. In particular, we compare the present structure with other structures containing either bound substrate, meso-diaminopimelate, or a conformationally restricted isoxazoline inhibitor. We have identified a significant interaction between the alpha-L-amino group of the unsaturated inhibitor and the indole ring of Trp144 that may account for the tight binding of this inhibitor.


Subject(s)
Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/metabolism , Amino Acids/metabolism , Corynebacterium/enzymology , NADP/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Conformation , NADP/chemistry , Protein Structure, Secondary
15.
Biochemistry ; 39(51): 16244-51, 2000 Dec 26.
Article in English | MEDLINE | ID: mdl-11123955

ABSTRACT

Ketopantoate reductase (EC 1.1.1.169) catalyzes the NADPH-dependent reduction of alpha-ketopantoate to D-(-)-pantoate in the biosynthesis of pantothenate. The pH dependence of V and V/K for the E. coli enzyme suggests the involvement of a general acid/base in the catalytic mechanism. To identify residues involved in catalysis and substrate binding, we mutated the following six strictly conserved residues to Ala: Lys72, Lys176, Glu210, Glu240, Asp248, and Glu256. Of these, the K176A and E256A mutant enzymes showed 233- and 42-fold decreases in V(max), and 336- and 63-fold increases in the K(m) value of ketopantoate, respectively, while the other mutants exhibited WT kinetic properties. The V(max) for the K176A and E256A mutant enzymes was markedly increased, up to 25% and 75% of the wild-type level, by exogenously added primary amines and formate, respectively. The rescue efficiencies for the K176A and E256A mutant enzymes were dependent on the molecular volume of rescue agents, as anticipated for a finite active site volume. The protonated form of the amine is responsible for recovery of activity, suggesting that Lys176 functions as a general acid in catalysis of ketopantoate reduction. The rescue efficiencies for the K176A mutant by primary amines were independent of the pK(a) value of the rescue agents (Bronsted coefficient, alpha = -0.004 +/-0.008). Insensitivity to acid strength suggests that the chemical reaction is not rate-limiting, consistent with (a) the catalytic efficiency of the wild-type enzyme (k(cat)/K(m) = 2x10(6) M(-1) s(-1) and (b) the small primary deuterium kinetic isotope effects, (D)V = 1.3 and (D)V/K = 1.5, observed for the wild-type enzyme. Larger primary deuterium isotope effects on V and V/K were observed for the K176A mutant ((D)V = 3.0, (D)V/K = 3.7) but decreased nearly to WT values as the concentration of ethylamine was increased. The nearly WT activity of the E256A mutant in the presence of formate argues for an important role for this residue in substrate binding. The double mutant (K176A/E256A) has no detectable ketopantoate reductase activity. These results indicate that Lys176 and Glu256 of the E. coli ketopantoate reductase are active site residues, and we propose specific roles for each in binding ketopantoate and catalysis.


Subject(s)
Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Bacterial Proteins/metabolism , Escherichia coli/enzymology , Acetic Acid/chemistry , Alanine/genetics , Alcohol Oxidoreductases/biosynthesis , Alcohol Oxidoreductases/isolation & purification , Alkylation , Amino Acid Sequence , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Binding Sites , Cysteine/chemistry , Cysteine/genetics , Deuterium/chemistry , Formates/chemistry , Gene Expression Regulation, Bacterial , Glutamic Acid/genetics , Kinetics , Lysine/genetics , Molecular Sequence Data , Pantothenic Acid/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Sequence Alignment , Substrate Specificity/genetics
16.
Carbohydr Res ; 328(4): 473-80, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-11093703

ABSTRACT

The novel UDP-sugar uridine 5'-(3-deoxy-3-fluoro-D-galactopyranosyl diphosphate) (1) and UDP-(2-deoxy-2-fluoro)-D-galactose (2) have been prepared enzymatically and tested as substrate analogues for the enzyme UDP-galactopyranose mutase (UDP-Galp mutase EC 5.4.99.9). Turnover of both 1 and 2 by UDP-Galp mutase was observed by HPLC and 19F NMR. The HPLC elution profile and 19F chemical shift of the products are consistent with the formation of the predicted furanose forms of 1 and 2. The Km values for compounds 1 and 2 were similar to those of the natural substrate UDP-Galp (0.26 mM for 1, 0.2 mM for 2, and 0.6 mM for UDP-Galp), but the values for kcat were substantially different (1.6/min for 1, 0.02/min for 2, and 1364/min for UDP-Galp). A correlation was also observed between the equilibrium yield of product formed during turnover of UDP-sugar by UDP-Galp mutase (UDP-Galp, compound 1 or compound 2), and the amount of furanose present for the free sugar at thermal equilibrium in aqueous solution, using 1H and 19F NMR spectroscopy. The implications of these results to the mechanism of the unusual enzymatic reaction are discussed.


Subject(s)
Intramolecular Transferases/metabolism , Uridine Diphosphate Galactose/analogs & derivatives , Uridine Diphosphate Galactose/chemical synthesis , Bacterial Proteins/metabolism , Chromatography, High Pressure Liquid , Fluorine , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Substrate Specificity , Uridine Diphosphate Galactose/metabolism
17.
Biochemistry ; 39(31): 9174-87, 2000 Aug 08.
Article in English | MEDLINE | ID: mdl-10924111

ABSTRACT

Phenylalanine dehydrogenase catalyzes the reversible, pyridine nucleotide-dependent oxidative deamination of L-phenylalanine to form phenylpyruvate and ammonia. We have characterized the steady-state kinetic behavior of the enzyme from Rhodococcus sp. M4 and determined the X-ray crystal structures of the recombinant enzyme in the complexes, E.NADH.L-phenylalanine and E.NAD(+). L-3-phenyllactate, to 1.25 and 1.4 A resolution, respectively. Initial velocity, product inhibition, and dead-end inhibition studies indicate the kinetic mechanism is ordered, with NAD(+) binding prior to phenylalanine and the products' being released in the order of ammonia, phenylpyruvate, and NADH. The enzyme shows no activity with NADPH or other 2'-phosphorylated pyridine nucleotides but has broad activity with NADH analogues. Our initial structural analyses of the E.NAD(+).phenylpyruvate and E.NAD(+). 3-phenylpropionate complexes established that Lys78 and Asp118 function as the catalytic residues in the active site [Vanhooke et al. (1999) Biochemistry 38, 2326-2339]. We have studied the ionization behavior of these residues in steady-state turnover and use these findings in conjunction with the structural data described both here and in our first report to modify our previously proposed mechanism for the enzymatic reaction. The structural characterizations also illuminate the mechanism of the redox specificity that precludes alpha-amino acid dehydrogenases from functioning as alpha-hydroxy acid dehydrogenases.


Subject(s)
Amino Acid Oxidoreductases/chemistry , Rhodococcus/enzymology , Amino Acid Oxidoreductases/antagonists & inhibitors , Amino Acid Oxidoreductases/isolation & purification , Catalysis , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Hydrogen/chemistry , Hydrogen-Ion Concentration , Kinetics , Lactates/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , NAD/chemistry , NADP/chemistry , Phenylalanine/chemistry , Phenylpropionates/chemistry , Protein Conformation , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity
18.
Biochemistry ; 39(29): 8556-64, 2000 Jul 25.
Article in English | MEDLINE | ID: mdl-10913262

ABSTRACT

The first unique step in bacterial and plant methionine biosynthesis involves the acylation of the gamma-hydroxyl of homoserine. In Haemophilus influenzae, acylation is accomplished via an acetyl-CoA-dependent acetylation catalyzed by homoserine transacetylase. The activity of this enzyme regulates flux of homoserine into multiple biosynthetic pathways and, therefore, represents a critical control point for cell growth and viability. We have cloned homoserine transacetylase from H. influenzae and present the first detailed enzymatic study of this enzyme. Steady-state kinetic experiments demonstrate that the enzyme utilizes a ping-pong kinetic mechanism in which the acetyl group of acetyl-CoA is initially transferred to an enzyme nucleophile before subsequent transfer to homoserine to form the final product, O-acetylhomoserine. The maximal velocity and V/K(homoserine) were independent of pH over the range of values tested, while V/K(acetyl)(-)(CoA) was dependent upon the ionization state of a single group exhibiting a pK value of 8.6, which was required to be protonated. Solvent kinetic isotope effect studies yielded inverse effects of 0.75 on V and 0.74 on V/K(CoA) on the reverse reaction and effects of 1.2 on V and 1.7 on V/K(homoserine) on the forward reaction. Direct evidence for the formation of an acetyl-enzyme intermediate was obtained using rapid-quench labeling studies. On the basis of these observations, we propose a chemical mechanism for this important member of the acyltransferase family and contrast its mechanism with that of homoserine transsuccinylase.


Subject(s)
Acetyltransferases/genetics , Acetyltransferases/metabolism , Haemophilus influenzae/enzymology , Haemophilus influenzae/genetics , Acetyltransferases/isolation & purification , Acylation , Base Sequence , DNA Primers/genetics , Gene Expression , Genes, Bacterial , Homoserine/chemistry , Homoserine/metabolism , Hydrogen-Ion Concentration , Kinetics , Models, Chemical
19.
Biochemistry ; 39(13): 3708-17, 2000 Apr 04.
Article in English | MEDLINE | ID: mdl-10736170

ABSTRACT

Ketopantoate reductase (EC 1.1.1.169) catalyzes the NADPH-dependent reduction of alpha-ketopantoate to form D-(-)-pantoate in the pantothenate/coenzyme A biosynthetic pathway. The enzyme encoded by the panE gene from E. coli K12 was overexpressed and purified to homogeneity. The native enzyme exists in solution as a monomer with a molecular mass of 34 000 Da. The steady-state initial velocity and product inhibition patterns are consistent with an ordered sequential kinetic mechanism in which NADPH binding is followed by ketopantoate binding, and pantoate release precedes NADP(+) release. The pH dependence of the kinetic parameters V and V/K for substrates in both the forward and reverse reactions suggests the involvement of a single general acid/base in the catalytic mechanism. An enzyme group exhibiting a pK value of 8.4 +/- 0.2 functions as a general acid in the direction of the ketopantoate reduction, while an enzyme group exhibiting a pK value of 7.8 +/- 0.2 serves as a general base in the direction of pantoate oxidation. The stereospecific transfer of the pro-S hydrogen atom of NADPH to the C-2 position of ketopantoate was demonstrated by (1)H NMR spectroscopy. Primary deuterium kinetic isotope effects of 1.3 and 1.5 on V(for) and V/K(NADPH), respectively, and 2.1 and 1.3 on V(rev) and V/K(HP), respectively, suggest that hydride transfer is not rate-limiting in catalysis. Solvent kinetic isotope effects of 1.3 on both V(for) and V/K(KP), and 1.4 and 1.5 on V(rev) and V/K(HP), respectively, support this conclusion. The apparent equilibrium constant, K(eq)', of 676 at pH 7.5 and the standard free energy change, DeltaG, of -14 kcal/mol suggest that ketopantoate reductase reaction is very favorable in the physiologically important direction of pantoate formation.


Subject(s)
Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/metabolism , Escherichia coli/enzymology , Alcohol Oxidoreductases/biosynthesis , Alcohol Oxidoreductases/genetics , Amino Acid Sequence , Deuterium , Escherichia coli/genetics , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Plasmids/chemistry , Plasmids/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Solvents , Stereoisomerism , Thermodynamics
20.
Biochemistry ; 39(6): 1285-93, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10684608

ABSTRACT

The E. coli argE-encoded N-acetyl-L-ornithine deacetylase has been cloned, expressed, and purified in high yield. The substrate specificity of the enzyme is relatively broad, with a number of alpha-N-acyl-L-amino acids exhibiting activity, including both alpha-N-acetyl- and alpha-N-formylmethionine that exhibit higher activity than alpha-N-acetyl-L-ornithine. Sequence homolgy suggests that the enzyme is a member of the metal-dependent aminoacylase family, and the purified enzyme contains a single atom of zinc per monomer. The activity of this enzyme can be increased greater than 2-fold by the addition of zinc, or 8-fold by the addition of cobalt. This suggests that the enzyme can accommodate two metal ions at the active site. The pH dependence of the kinetic parameters has been determined and revealed the presence of two enzymic groups, one functioning as a general base and one functioning as a general acid. Solvent kinetic isotope effects on the hydrolysis of N-acetylornithine have been determined, and a linear proton inventory suggests that a single proton transfer occurs in a partially rate-limiting step. A chemical mechanism is proposed and compared with other mechanisms determined for other members of the aminoacylase family.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/biosynthesis , Amidohydrolases/genetics , Amino Acid Sequence , Cloning, Molecular , Deuterium/chemistry , Escherichia coli/enzymology , Genetic Vectors/chemical synthesis , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemical synthesis , Recombinant Proteins/isolation & purification , Sodium Fluoride/chemistry , Solvents/chemistry , Substrate Specificity/genetics , Zinc/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...