Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters











Publication year range
1.
J Mol Biol ; 319(3): 757-66, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-12054868

ABSTRACT

Indoleglycerol phosphate synthase catalyzes the ring closure of an N-alkylated anthranilate to a 3-alkyl indole derivative, a reaction requiring Lewis acid catalysis in vitro. Here, we investigated the enzymatic reaction mechanism through X-ray crystallography of complexes of the hyperthermostable enzyme from Sulfolobus solfataricus with the substrate 1-(o-carboxyphenylamino) 1-deoxyribulose 5-phosphate, a substrate analogue and the product indole-3-glycerol phosphate. The substrate and the substrate analogue are bound to the active site in a similar, extended conformation between the previously identified phosphate binding site and a hydrophobic pocket for the anthranilate moiety. This binding mode is unproductive, because the carbon atoms that are to be joined are too far apart. The indole ring of the bound product resides in a second hydrophobic pocket adjacent to that of the anthranilate moiety of the substrate. Although the hydrophobic moiety of the substrate moves during catalysis from one hydrophobic pocket to the other, the triosephosphate moiety remains rigidly bound to the same set of hydrogen-bonding residues. Simultaneously, the catalytically important residues Lys53, Lys110 and Glu159 maintain favourable distances to the atoms of the ligand undergoing covalent changes. On the basis of these data, the structures of two putative catalytic intermediates were modelled into the active site. This new structural information and the modelling studies provide further insight into the mechanism of enzyme-catalyzed indole synthesis. The charged epsilon-amino group of Lys110 is the general acid, and the carboxylate group of Glu159 is the general base. Lys53 guides the substrate undergoing conformational transitions during catalysis, by forming a salt-bridge to the carboxylate group of its anthranilate moiety.


Subject(s)
Glycerophosphates/metabolism , Indole-3-Glycerol-Phosphate Synthase/chemistry , Indole-3-Glycerol-Phosphate Synthase/metabolism , Ribulosephosphates/metabolism , Sulfolobus acidocaldarius/enzymology , Binding Sites , Catalysis , Crystallography, X-Ray , Glycerophosphates/chemistry , Hydrogen Bonding , Indoles/metabolism , Ligands , Models, Molecular , Protein Conformation , Ribulosephosphates/chemistry , Static Electricity
2.
Nat Struct Biol ; 8(11): 963-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11685243

ABSTRACT

DOPA decarboxylase (DDC) is responsible for the synthesis of the key neurotransmitters dopamine and serotonin via decarboxylation of L-3,4-dihydroxyphenylalanine (L-DOPA) and L-5-hydroxytryptophan, respectively. DDC has been implicated in a number of clinic disorders, including Parkinson's disease and hypertension. Peripheral inhibitors of DDC are currently used to treat these diseases. We present the crystal structures of ligand-free DDC and its complex with the anti-Parkinson drug carbiDOPA. The inhibitor is bound to the enzyme by forming a hydrazone linkage with the cofactor, and its catechol ring is deeply buried in the active site cleft. The structures provide the molecular basis for the development of new inhibitors of DDC with better pharmacological characteristics.


Subject(s)
Antiparkinson Agents/chemistry , Antiparkinson Agents/metabolism , Aromatic Amino Acid Decarboxylase Inhibitors , Carbidopa/chemistry , Carbidopa/metabolism , Dopa Decarboxylase/chemistry , Parkinson Disease/drug therapy , 5-Hydroxytryptophan/chemistry , 5-Hydroxytryptophan/metabolism , Animals , Antiparkinson Agents/pharmacology , Benserazide/chemistry , Benserazide/pharmacology , Binding Sites , Carbidopa/pharmacology , Crystallography, X-Ray , Dopa Decarboxylase/metabolism , Drug Design , Humans , Kidney/enzymology , Levodopa/chemistry , Levodopa/metabolism , Ligands , Models, Molecular , Parkinson Disease/enzymology , Pliability , Protein Structure, Secondary , Swine
3.
J Mol Biol ; 303(2): 279-86, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11023792

ABSTRACT

A new crystal structure of O-acetylserine sulfhydrylase (OASS) has been solved with chloride bound at an allosteric site and sulfate bound at the active site. The bound anions result in a new "inhibited" conformation, that differs from the "open" native or "closed" external aldimine conformations. The allosteric site is located at the OASS dimer interface. The new inhibited structure involves a change in the position of the "moveable domain" (residues 87-131) to a location that differs from that in the open or closed forms. Formation of the external aldimine with substrate is stabilized by interaction of the alpha-carboxyl group of the substrate with a substrate-binding loop that is part of the moveable domain. The inhibited conformation prevents the substrate-binding loop from interacting with the alpha-carboxyl group, and hinders formation of the external Schiff base and thus subsequent chemistry. Chloride may be an analog of sulfide, the physiological inhibitor. Finally, these results suggest that OASS represents a new class of PLP-dependent enzymes that is regulated by small anions.


Subject(s)
Chlorides/metabolism , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Salmonella typhimurium/enzymology , Allosteric Regulation , Allosteric Site , Anions/metabolism , Anions/pharmacology , Chlorides/pharmacology , Crystallography, X-Ray , Cysteine/biosynthesis , Cysteine/metabolism , Cysteine Synthase/antagonists & inhibitors , Dimerization , Hydrogen Bonding , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , Pyridoxal Phosphate/metabolism , Salmonella typhimurium/metabolism , Structure-Activity Relationship , Sulfates/metabolism , Sulfides/metabolism
4.
J Mol Biol ; 302(1): 135-54, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10964566

ABSTRACT

Thioredoxins are small ubiquitous proteins which act as general protein disulfide reductases in living cells. Chloroplasts contain two distinct thioredoxins ( f and m) with different phylogenetic origin. Both act as enzyme regulatory proteins but have different specificities towards target enzymes. Thioredoxin f (Trx f), which shares only low sequence identity with thioredoxin m (Trx m) and with all other known thioredoxins, activates enzymes of the Calvin cycle and other photosynthetic processes. Trx m shows high sequence similarity with bacterial thioredoxins and activates other chloroplast enzymes. The here described structural studies of the two chloroplast thioredoxins were carried out in order to gain insight into the structure/function relationships of these proteins. Crystal structures were determined for oxidized, recombinant thioredoxin f (Trx f-L) and at the N terminus truncated form of it (Trx f-S), as well as for oxidized and reduced thioredoxin m (at 2.1 and 2.3 A resolution, respectively). Whereas thioredoxin f crystallized as a monomer, both truncated thioredoxin f and thioredoxin m crystallized as non-covalent dimers. The structures of thioredoxins f and m exhibit the typical thioredoxin fold consisting of a central twisted five-stranded beta-sheet surrounded by four alpha-helices. Thioredoxin f contains an additional alpha-helix at the N terminus and an exposed third cysteine close to the active site. The overall three-dimensional structures of the two chloroplast thioredoxins are quite similar. However, the two proteins have a significantly different surface topology and charge distribution around the active site. An interesting feature which might significantly contribute to the specificity of thioredoxin f is an inherent flexibility of its active site, which has expressed itself crystallographically in two different crystal forms.


Subject(s)
Plant Proteins/chemistry , Spinacia oleracea/chemistry , Thioredoxins/chemistry , Amino Acid Sequence , Binding Sites , Chloroplast Thioredoxins , Chloroplasts/chemistry , Crystallography, X-Ray , Cysteine/metabolism , Evolution, Molecular , Models, Molecular , Molecular Sequence Data , Molecular Weight , Oxidation-Reduction , Pliability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Sequence Alignment , Spinacia oleracea/cytology , Structure-Activity Relationship , Substrate Specificity , Surface Properties
5.
J Mol Biol ; 294(3): 745-56, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10610793

ABSTRACT

The 2.4 A crystal structure of the vitamin B6-dependent enzyme 1-aminocyclopropane-1-carboxylate (ACC) synthase is described. This enzyme catalyses the committed step in the biosynthesis of ethylene, a plant hormone that is responsible for the initiation of fruit ripening and for regulating many other developmental processes. ACC synthase has 15 % sequence identity with the well-studied aspartate aminotransferase, and a completely different catalytic activity yet the overall folds and the active sites are very similar. The new structure together with available biochemical data enables a comparative mechanistic analysis that largely explains the catalytic roles of the conserved and non-conserved active site residues. An external aldimine reaction intermediate (external aldimine with ACC, i.e. with the product) has been modeled. The new structure provides a basis for the rational design of inhibitors with broad agricultural applications.


Subject(s)
Ethylenes/biosynthesis , Lyases/chemistry , Lyases/metabolism , Plants/enzymology , Amino Acid Sequence , Aspartate Aminotransferases/chemistry , Binding Sites , Crystallography, X-Ray , Dimerization , Drug Design , Enzyme Inhibitors/chemical synthesis , Models, Molecular , Molecular Sequence Data , Protein Conformation
6.
J Mol Biol ; 294(1): 193-200, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10556038

ABSTRACT

The crystal structures of four inhibitor complexes of dialkylglycine decarboxylase are reported. The enzyme does not undergo a domain closure, as does aspartate aminotransferase, upon inhibitor binding. Two active-site conformations have been observed in previous structures that differ in alkali metal ion content, and two active-site conformations have been shown to coexist in solution when a single type of metal ion is present. There is no indication of coexisting conformers in the structures reported here or in the previously reported structures, and the observed conformation is that expected based on the presence of potassium in the enzyme. Thus, although two active-site conformations coexist in solution, a single conformation, corresponding to the more active enzyme, predominates in the crystal. The structure of 1-aminocyclopropane-1-carboxylate bound in the active site shows the aldimine double bond to the pyridoxal phosphate cofactor to be fully out of the plane of the coenzyme ring, whereas the Calpha-CO2(-) bond lies close to it. This provides an explanation for the observed lack of decarboxylation reactivity with this amino acid. The carboxylate groups of both 1-aminocyclopropane-1-carboxylate and 5'-phosphopyridoxyl-2-methylalanine interact with Ser215 and Arg406 as previously proposed. This demonstrates structurally that alternative binding modes, which constitute substrate inhibition, occur in the decarboxylation half-reaction. The structures of d and l-cycloserine bound to the active-site show that the l-isomer is deprotonated at C(alpha), presumably by Lys272, while the d-isomer is not. This difference explains the approximately 3000-fold greater potency of the l versus the d-isomer as a competitive inhibitor of dialkylglycine decarboxylase.


Subject(s)
Amino Acids, Cyclic , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/chemistry , Alanine/analogs & derivatives , Alanine/chemistry , Amino Acids/chemistry , Crystallography, X-Ray , Cycloserine/chemistry , Ligands , Models, Molecular , Potassium/chemistry , Pyridines/chemistry , Pyridoxal Phosphate/chemistry
7.
J Biol Chem ; 274(44): 31203-8, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10531314

ABSTRACT

The conjoint substitution of three active-site residues in aspartate aminotransferase (AspAT) of Escherichia coli (Y225R/R292K/R386A) increases the ratio of L-aspartate beta-decarboxylase activity to transaminase activity >25 million-fold. This result was achieved by combining an arginine shift mutation (Y225R/R386A) with a conservative substitution of a substrate-binding residue (R292K). In the wild-type enzyme, Arg(386) interacts with the alpha-carboxylate group of the substrate and is one of the four residues that are invariant in all aminotransferases; Tyr(225) is in its vicinity, forming a hydrogen bond with O-3' of the cofactor; and Arg(292) interacts with the distal carboxylate group of the substrate. In the triple-mutant enzyme, k(cat)' for beta-decarboxylation of L-aspartate was 0.08 s(-1), whereas k(cat)' for transamination was decreased to 0.01 s(-1). AspAT was thus converted into an L-aspartate beta-decarboxylase that catalyzes transamination as a side reaction. The major pathway of beta-decarboxylation directly produces L-alanine without intermediary formation of pyruvate. The various single- or double-mutant AspATs corresponding to the triple-mutant enzyme showed, with the exception of AspAT Y225R/R386A, no measurable or only very low beta-decarboxylase activity. The arginine shift mutation Y225R/R386A elicits beta-decarboxylase activity, whereas the R292K substitution suppresses transaminase activity. The reaction specificity of the triple-mutant enzyme is thus achieved in the same way as that of wild-type pyridoxal 5'-phosphate-dependent enzymes in general and possibly of many other enzymes, i.e. by accelerating the specific reaction and suppressing potential side reactions.


Subject(s)
Aspartate Aminotransferases/genetics , Aspartic Acid/metabolism , Carboxy-Lyases/genetics , Catalytic Domain/genetics , Mutation , Aspartate Aminotransferases/metabolism , Carboxy-Lyases/metabolism , Computer Simulation , Escherichia coli/enzymology , Models, Chemical , Models, Molecular , Protein Engineering
8.
Proc Natl Acad Sci U S A ; 96(17): 9479-84, 1999 Aug 17.
Article in English | MEDLINE | ID: mdl-10449718

ABSTRACT

Anthranilate synthase catalyzes the synthesis of anthranilate from chorismate and glutamine and is feedback-inhibited by tryptophan. The enzyme of the hyperthermophile Sulfolobus solfataricus has been crystallized in the absence of physiological ligands, and its three-dimensional structure has been determined at 2.5-A resolution with x-ray crystallography. It is a heterotetramer of anthranilate synthase (TrpE) and glutamine amidotransferase (TrpG) subunits, in which two TrpG:TrpE protomers associate mainly via the TrpG subunits. The small TrpG subunit (195 residues) has the known "triad" glutamine amidotransferase fold. The large TrpE subunit (421 residues) has a novel fold. It displays a cleft between two domains, the tips of which contact the TrpG subunit across its active site. Clusters of catalytically essential residues are located inside the cleft, spatially separated from clustered residues involved in feedback inhibition. The structure suggests a model in which chorismate binding triggers a relative movement of the two domain tips of the TrpE subunit, activating the TrpG subunit and creating a channel for passage of ammonia toward the active site of the TrpE subunit. Tryptophan presumably blocks this rearrangement, thus stabilizing the inactive states of both subunits. The structure of the TrpE subunit is a likely prototype for the related enzymes 4-amino 4-deoxychorismate synthase and isochorismate synthase.


Subject(s)
Anthranilate Synthase/chemistry , Sulfolobus/enzymology , Amino Acid Sequence , Binding Sites , Catalysis , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Folding
9.
J Mol Biol ; 291(4): 941-53, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10452898

ABSTRACT

Covalent binding of L-methionine as an external aldimine to the pyridoxal 5'-phosphate-cofactor in the K41A mutant of O-acetylserine sulfhydrylase from Salmonella typhimurium induces a large conformational change in the protein. Methionine mimics the action of the substrate O-acetyl-L-serine during catalysis. The alpha-carboxylate moiety of L-methionine in external aldimine linkage with the active site pyridoxal 5'-phosphate forms a hydrogen bonding network to the "asparagine-loop" P67-T68-N69-G70 which adopts a different conformation than in the native protein. The side-chain nitrogen of Asn69 moves more than 7 A to make a hydrogen bond to the alpha-carboxylate group of the inhibitor. As the external aldimine is formed, the PLP tilts by 13 degrees along its longitudinal axis such that C4' moves toward the entrance to the active site and the side-chain of the methionine is directed toward the active site entrance. The local rearrangement acts as a trigger to induce a large global conformational change in the protein. A subdomain comprised of beta-strand 4, alpha-helix 3, beta-strand 5 and alpha-helix 4 moves towards the active site by a rotation of 7 degrees. This subdomain movement results in a reduction of the severe twist of its central beta-sheet and reduces the active site entrance to a small hole, giving access only to small molecules like sulfide, the second substrate, or acetate, the first product.


Subject(s)
Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Salmonella typhimurium/enzymology , Aspartate Aminotransferases/chemistry , Aspartate Aminotransferases/metabolism , Catalytic Domain/genetics , Crystallography, X-Ray , Cysteine Synthase/genetics , Dimerization , Hydrogen Bonding , Ligands , Methionine/metabolism , Models, Molecular , Point Mutation , Protein Conformation , Protein Structure, Secondary , Salmonella typhimurium/genetics , Stereoisomerism , Tryptophan Synthase/chemistry , Tryptophan Synthase/metabolism
10.
Biochemistry ; 38(27): 8628-34, 1999 Jul 06.
Article in English | MEDLINE | ID: mdl-10393538

ABSTRACT

gamma-Aminobutyrate aminotransferase (GABA-AT), a pyridoxal phosphate-dependent enzyme, is responsible for the degradation of the inhibitory neurotransmitter GABA and is a target for antiepileptic drugs because its selective inhibition raises GABA concentrations in brain. The X-ray structure of pig GABA-AT has been determined to 3.0 A resolution by molecular replacement with the distantly related enzyme ornithine aminotransferase. Both omega-aminotransferases have the same fold, but exhibit side chain replacements in the closely packed binding site that explain their respective specificities. The aldimines of GABA and the antiepileptic drug vinyl-GABA have been modeled into the active site.


Subject(s)
4-Aminobutyrate Transaminase/chemistry , Anticonvulsants/chemistry , Epilepsy/drug therapy , Epilepsy/enzymology , Amino Acid Sequence , Animals , Binding Sites , Computer Simulation , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Folding , Solutions , Swine , Vigabatrin , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/therapeutic use
11.
J Mol Biol ; 288(4): 753-63, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10329177

ABSTRACT

The recombinantly expressed protein indoleglycerol phosphate synthase from the hyperthermophilic bacterium Thermotoga maritima (tIGPS) was purified and characterized with respect to oligomerization state, catalytic properties and thermostability. This enzyme from the biosynthetic pathway of tryptophan is a monomer in solution. In contrast to IGPS from the hyperthermophilic archaeon Sulfolobus solfataricus, tIGPS shows high catalytic activity at room temperature and only weak product inhibition. In order to test the hypothesis that salt bridges in a critical context contribute to the high thermostability of tIGPS, two solvent-exposed salt bridges were selected, based on its three-dimensional structure, for individual disruption by site-directed mutagenesis. The first salt bridge fixes the N terminus to the core of the protein, and the second serves as a clamp between helices alpha1 and alpha8, which are widely separated in sequence but adjacent in the (betaalpha)8-barrel. Kinetics of irreversible heat inactivation reveal that the salt bridge crosslinking helices alpha1 and alpha8 stabilizes tIGPS more strongly than that tethering the N terminus.


Subject(s)
Thermotoga maritima/enzymology , Tryptophan Synthase/metabolism , Base Sequence , Biopolymers , Catalysis , Cloning, Molecular , DNA Primers , Enzyme Stability , Hot Temperature , Kinetics , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solvents , Tryptophan Synthase/genetics , Tryptophan Synthase/isolation & purification
12.
Acta Crystallogr D Biol Crystallogr ; 55(Pt 2): 568-70, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10089382

ABSTRACT

DOPA decarboxylase is responsible for the synthesis of the key neurotransmitters dopamine and serotonin via decarboxylation of L-3, 4-dihydroxyphenylalanine (L-DOPA) and L-5-hydroxytryptophan, respectively. The crystals of recombinant DOPA decarboxylase differ from those previously reported for the enzyme purified from pig kidney. They belong to space group P622 with unit-cell dimensions a = b = 302.6, c = 178.1 A. Both the self-rotation function and the good diffraction quality of these crystals (2.5 A on a synchrotron source) suggest that there should be at least three protein dimers in the asymmetric unit. Diffraction data sets have been collected for the native enzyme and a heavy-atom derivative.


Subject(s)
Dopa Decarboxylase/chemistry , Kidney/enzymology , Animals , Crystallography, X-Ray , Protein Conformation , Recombinant Proteins/chemistry , Swine
13.
J Mol Biol ; 286(3): 829-50, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10024454

ABSTRACT

Phosphoserine aminotransferase (PSAT; EC 2.6.1.52), a member of subgroup IV of the aminotransferases, catalyses the conversion of 3-phosphohydroxypyruvate to l-phosphoserine. The crystal structure of PSAT from Escherichia coli has been solved in space group P212121 using MIRAS phases in combination with density modification and was refined to an R-factor of 17.5% (Rfree=20.1 %) at 2.3 A resolution. In addition, the structure of PSAT in complex with alpha-methyl-l-glutamate (AMG) has been refined to an R-factor of 18.5% (Rfree=25.1%) at 2.8 A resolution. Each subunit (361 residues) of the PSAT homodimer is composed of a large pyridoxal-5'-phosphate binding domain (residues 16-268), consisting of a seven-stranded mainly parallel beta-sheet, two additional beta-strands and seven alpha-helices, and a small C-terminal domain, which incorporates a five-stranded beta-sheet and two alpha-helices. A three-dimensional structural comparison to four other vitamin B6-dependent enzymes reveals that three alpha-helices of the large domain, as well as an N-terminal domain (subgroup II) or subdomain (subgroup I) are absent in PSAT. Its only 15 N-terminal residues form a single beta-strand, which participates in the beta-sheet of the C-terminal domain. The cofactor is bound through an aldimine linkage to Lys198 in the active site. In the PSAT-AMG complex Ser9 and Arg335 bind the AMG alpha-carboxylate group while His41, Arg42 and His328 are involved in binding the AMG side-chain. Arg77 binds the AMG side-chain indirectly through a solvent molecule and is expected to position itself during catalysis between the PLP phosphate group and the substrate side-chain. Comparison of the active sites of PSAT and aspartate aminotransferase suggests a similar catalytic mechanism, except for the transaldimination step, since in PSAT the Schiff base is protonated. Correlation of the PSAT crystal structure to a published profile sequence analysis of all subgroup IV members allows active site modelling of nifs and the proposal of a likely molecular reaction mechanism.


Subject(s)
Escherichia coli/enzymology , Glutamates/chemistry , Transaminases/chemistry , Alanine Transaminase/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Dimerization , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Folding , Protein Structure, Secondary , Pyridoxal Phosphate/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
14.
Biochemistry ; 38(3): 905-13, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9893985

ABSTRACT

To elucidate the role of the two conserved cis-proline residues of aspartate aminotransferase (AspAT), one double and two single mutants of the enzyme from Escherichia coli (EcAspAT) were prepared: P138A, P195A and P138A/P195A in which the two prolines were replaced by alanine. The crystal structures of P195A and P138A/P195A have been determined at 2.3-2.1 A resolution. The wild-type geometry, including the cis conformation of the 194-195 peptide bond is retained upon substitution of proline 195 by alanine, whereas the trans conformation is adopted at the 137-138 peptide bond. Quite surprisingly, the replacement of each of the two prolines by alanine does not significantly affect either the activity or the stability of the protein. All the three mutants follow the same pathway as the wild type for unfolding equilibrium induced by guanidine hydrochloride [Herold, M., and Kirschner, K. (1990) Biochemistry 29, 1907-1913]. The kinetics of renaturation of P195A, where the alanine retains the wild-type cis conformation, is faster than wild type, whereas renaturation of P138A, which adopts the trans conformation, is slower. We conclude that cis-prolines seem to have been retained throughout the evolution of aspartate aminotransferase to possibly play a subtle role in directing the traffic of intermediates toward the unique structure of the native state, rather than to respond to the needs for a specific catalytic or functional role.


Subject(s)
Aspartate Aminotransferases/genetics , Escherichia coli/enzymology , Mutagenesis, Site-Directed , Proline/genetics , Alanine/genetics , Amino Acid Substitution/genetics , Aspartate Aminotransferases/chemistry , Crystallography, X-Ray , Enzyme Activation/genetics , Escherichia coli/genetics , Guanidine , Hot Temperature , Kinetics , Ligands , Proline/chemistry , Protein Conformation , Protein Denaturation , Protein Folding , Spectrophotometry , Structure-Activity Relationship
15.
J Mol Biol ; 285(1): 297-309, 1999 Jan 08.
Article in English | MEDLINE | ID: mdl-9878407

ABSTRACT

Ornithine aminotransferase (l-ornithine:2-oxoacid delta-aminotransferase; EC 2.6.1.13), a pyridoxal-5'-phosphate-dependent mitochondrial enzyme controls the l-ornithine level in tissues by catalyzing the transfer of the delta-amino group of l-ornithine to 2-oxoglutarate, producing l-glutamate- gamma-semialdehyde and l-glutamate. (2S, 5S)-5-Fluoromethylornithine is the only inhibitor exclusively specific for ornithine aminotransferase known to date. Both in vitro and in vivo, it blocks the enzyme by a suicide reaction leading to a covalent adduct with the cofactor. The crystal structure of the enzyme-inhibitor complex was solved at a resolution of 1.95 A. No significant conformational changes compared with the native enzyme structure were observed. The structure reveals the atomic details of the cofactor-inhibitor adduct and its interactions with the active site of the enzyme. The main residues responsible for specific binding of the inhibitor are Arg180, which forms a strong salt bridge with the alpha-carboxylate and Tyr55, which is involved in a short hydrogen bond with the alpha-amino group. The experimental observation that in the racemic mixture, (2S, 5S)-5-fluoromethylornithine is exclusively responsible for the enzyme inhibition can be explained on the basis of the active site topology. Model building studies strongly suggest that the natural substrate l-ornithine, in its external aldimine adduct with the enzyme, makes use of the same recognition site as the inhibitor. It is proposed that the neutralization of the active site Arg413 by a salt bridge with Glu235 also plays an important role in productive binding of both 5-fluoromethylornithine and l-ornithine. Arg180 and Arg413 are believed to be instrumental in recognition of l-glutamate, by binding its gamma and alpha-carboxylate groups, respectively. This requires a different side-chain conformation of Glu235. Lys292 is the only obvious candidate for catalyzing the rate-limiting proton transfer steps in the transamination reaction.


Subject(s)
Enzyme Inhibitors/chemistry , Ornithine-Oxo-Acid Transaminase/chemistry , Ornithine/analogs & derivatives , Protein Conformation , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Glutamic Acid , Humans , Microspectrophotometry , Models, Molecular , Molecular Sequence Data , Molecular Structure , Ornithine/chemistry , Ornithine/metabolism , Ornithine-Oxo-Acid Transaminase/antagonists & inhibitors , Ornithine-Oxo-Acid Transaminase/metabolism , Solutions , Substrate Specificity
16.
Acta Crystallogr D Biol Crystallogr ; 54(Pt 5): 1020-2, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9757126

ABSTRACT

Glutamate decarboxylase (GAD) is a vitamin B6 enzyme which catalyzes the alpha-decarboxylation of L-glutamate to gamma-aminobutyric acid (GABA). Escherichia coli cells coexpress two highly homologous enzyme isoforms, GADalpha and GADbeta. Well diffracting crystals of GADbeta were obtained by taking advantage of the possibility of expressing each isoform separately. They belong to space group P31 or P32 with the unit-cell dimensions a = b = 115.6 and c = 206.6 A and contain one GAD hexamer in the asymmetric unit. High-resolution synchrotron data were collected at 100 K for the native protein and a potential heavy-atom derivative.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli/enzymology , Glutamate Decarboxylase/chemistry , Isoenzymes/chemistry , Protein Conformation , Bacterial Proteins/isolation & purification , Crystallization , Crystallography, X-Ray , Glutamate Decarboxylase/isolation & purification , Isoenzymes/isolation & purification , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification
17.
J Mol Biol ; 283(1): 121-33, 1998.
Article in English | MEDLINE | ID: mdl-9761678

ABSTRACT

The last step in cysteine biosynthesis in enteric bacteria is catalyzed by the pyridoxal 5'-phosphate-dependent enzyme O-acetylserine sulfhydrylase. Here we report the crystal structure at 2.2 A resolution of the A-isozyme of O-acetylserine sulfhydrylase isolated from Salmonella typhimurium. O-acetylserine sulfhydrylase shares the same fold with tryptophan synthase-beta from Salmonella typhimurium but the sequence identity level is below 20%. There are some major structural differences: the loops providing the interface to the alpha-subunit in tryptophan synthase-beta and two surface helices of tryptophan synthase-beta are missing in O-acetylserine sulfhydrylase. The hydrophobic channel for indole transport from the alpha to the beta active site of tryptophan synthase-beta is, not unexpectedly, also absent in O-acetylserine sulfhydrylase. The dimer interface, on the other hand, is more or less conserved in the two enzymes. The active site cleft of O-acetylserine sulfhydrylase is wider and therefore more exposed to the solvent. A possible binding site for the substrate O-acetylserine is discussed.


Subject(s)
Cysteine Synthase/chemistry , Models, Molecular , Salmonella typhimurium/enzymology , Binding Sites , Crystallography, X-Ray , Dimerization , Indoles , Protein Conformation , Pyridoxal Phosphate/chemistry , Tryptophan Synthase/chemistry
18.
J Mol Biol ; 277(1): 81-102, 1998 Mar 20.
Article in English | MEDLINE | ID: mdl-9514741

ABSTRACT

Ornithine aminotransferase (OAT), a pyridoxal-5'-phosphate dependent enzyme, catalyses the transfer of the delta-amino group of L-ornithine to 2-oxoglutarate, producing L-glutamate-gamma-semialdehyde, which spontaneously cyclizes to pyrroline-5-carboxylate, and L-glutamate. The crystal structure determination of human recombinant OAT is described in this paper. As a first step, the structure was determined at low resolution (6 A) by molecular replacement using the refined structure of dialkylglycine decarboxylase as a search model. Crystallographic phases were then refined and extended in a step-wise fashion to 2.5 A by cyclic averaging of the electron density corresponding to the three monomers within the asymmetric unit. Interpretation of the resulting map was straightforward and refinement of the model resulted in an R-factor of 17.1% (Rfree=24.3%). The success of the procedure demonstrates the power of real-space molecular averaging even with only threefold redundancy. The alpha6-hexameric molecule is a trimer of intimate dimers with a monomer-monomer interface of 5500 A2 per subunit. The three dimers are related by an approximate 3-fold screw axis with a translational component of 18 A. The monomer fold is that of a typical representative of subgroup 2 aminotransferases and very similar to those described for dialkylglycine decarboxylase from Pseudomonas cepacia and glutamate-1-semialdehyde aminomutase from Synechococcus. It consists of a large domain that contributes most to the subunit interface, a C-terminal small domain most distant to the 2-fold axis and an N-terminal region that contains a helix, a loop and a three stranded beta-meander embracing a protrusion in the large domain of the second subunit of the dimer. The large domain contains the characteristic central seven-stranded beta-sheet (agfedbc) covered by eight helices in a typical alpha/beta fold. The cofactor pyridoxal-5'-phosphate is bound through a Schiff base to Lys292, located in the loop between strands f and g. The C-terminal domain includes a four-stranded antiparallel beta-sheet in contact with the large domain and three further helices at the far end of the subunit. The active sites of the dimer lie, about 25 A apart, at the subunit and domain interfaces. The conical entrances are on opposite sides of the dimer. In the active site, R180, E235 and R413 are probable substrate binding residues. Structure-based sequence comparisons with related transaminases in this work support that view. In patients suffering from gyrate atrophy, a recessive hereditary genetic disorder that can cause blindness in humans, ornithine aminotransferase activity is lacking. A large number of frameshift and point mutations in the ornithine aminotransferase gene have been identified in such patients. Possible effects of the various point mutations on the structural stability or the catalytic competence of the enzyme are discussed in light of the three-dimensional structure.


Subject(s)
Ornithine-Oxo-Acid Transaminase/chemistry , Binding Sites , Catalysis , Crystallography, X-Ray , Dimerization , Humans , Ornithine-Oxo-Acid Transaminase/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
19.
Curr Opin Struct Biol ; 8(6): 759-69, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9914259

ABSTRACT

The number of known three-dimensional structures of vitamin B6-dependent enzymes has doubled in the past two years. A fourth type of fold for B6-dependent enzymes, involving a TIM-barrel domain, has been discovered. Alanine racemase is the first known representative of this new fold. Significant progress has been made in understanding the allosteric effects in the tryptophan synthase reaction.


Subject(s)
Alanine Racemase/chemistry , Alanine Transaminase/chemistry , Aspartate Aminotransferases/chemistry , Pyridoxine/metabolism , Tryptophan Synthase/chemistry , Alanine Racemase/metabolism , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , D-Alanine Transaminase , Models, Molecular , Protein Conformation , Tryptophan Synthase/metabolism
20.
Biochemistry ; 36(20): 6009-16, 1997 May 20.
Article in English | MEDLINE | ID: mdl-9166771

ABSTRACT

The structural basis of thermostability of proteins is of great scientific and biotechnological interest. Differences in the X-ray structues of orthologous proteins from hyperthermophilic and mesophilic organisms can indicate crucial stabilizing interactions. To this end the crystal structure of dimeric phosphoribosyl anthranilate isomerase from the hyperthermophile Thermotoga maritima (tPRAI) was determined using phases derived from the isomorphous replacement method and was refined at 2.0 A resolution. The comparison to the known 2.0 A structure of PRAI from Escherichia coli (ePRAI) shows that tPRAI has the complete TIM- or (beta alpha)8-barrel fold, whereas helix alpha5 in ePRAI is replaced by a loop. The subunits of tPRAI associate via the N-terminal faces of their central beta-barrels. Two long, symmetry-related loops that protrude reciprocally into cavities of the other subunit provide for multiple hydrophobic interactions. Moreover, the side chains of the N-terminal methionines and the C-terminal leucines of both subunits are immobilized in a hydrophobic cluster, and the number of salt bridges is increased in tPRAI. These features appear to be mainly responsible for the high thermostability of tPRAI. In contrast to other hyperthermostable enzymes, tPRAI at 25 degrees C is catalytically more efficient than ePRAI, mainly due to its small K(M) value for the substrate [Sterner, R., Kleemann, G. R., Szadkowski, H., Lustig, A., Hennig, M., & Kirschner, K. (1996) Protein Sci. 5, 2000-2008]. The increased number of hydrogen bonds between the phosphate ion and tPRAI compared to ePRAI could be responsible for this effect.


Subject(s)
Aldose-Ketose Isomerases , Carbohydrate Epimerases/chemistry , Enzyme Stability , Gram-Negative Anaerobic Bacteria/enzymology , Protein Structure, Secondary , Protein Structure, Tertiary , Amino Acid Sequence , Binding Sites , Computer Simulation , Crystallography, X-Ray , Dimerization , Escherichia coli/enzymology , Hot Temperature , Models, Molecular , Molecular Sequence Data , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL