Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Nucleic Acids Res ; 44(13): 6232-41, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27141962

ABSTRACT

Proliferating cell nuclear antigen (PCNA) forms a trimeric ring that encircles duplex DNA and acts as an anchor for a number of proteins involved in DNA metabolic processes. PCNA has two structurally similar domains (I and II) linked by a long loop (inter-domain connector loop, IDCL) on the outside of each monomer of the trimeric structure that makes up the DNA clamp. All proteins that bind to PCNA do so via a PCNA-interacting peptide (PIP) motif that binds near the IDCL. A small protein, called TIP, binds to PCNA and inhibits PCNA-dependent activities although it does not contain a canonical PIP motif. The X-ray crystal structure of TIP bound to PCNA reveals that TIP binds to the canonical PIP interaction site, but also extends beyond it through a helix that relocates the IDCL. TIP alters the relationship between domains I and II within the PCNA monomer such that the trimeric ring structure is broken, while the individual domains largely retain their native structure. Small angle X-ray scattering (SAXS) confirms the disruption of the PCNA trimer upon addition of the TIP protein in solution and together with the X-ray crystal data, provides a structural basis for the mechanism of PCNA inhibition by TIP.


Subject(s)
DNA/chemistry , Peptides/chemistry , Proliferating Cell Nuclear Antigen/chemistry , Protein Conformation , Crystallography, X-Ray , DNA/metabolism , Nucleic Acid Conformation , Peptides/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Domains , Thermococcus/chemistry , Thermococcus/metabolism
3.
Biochemistry ; 51(51): 10121-3, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23234431

ABSTRACT

The structure of PA5508 from Pseudomonas aeruginosa, a glutamine synthetase (GS) homologue, has been determined at 2.5 Å. Surprisingly, PA5508 forms single hexameric rings rather than the stacked double rings that are characteristic of GS. The C-terminal helical thong motif that links GS rings is present in PA5508; however, it is folded back toward the core of its own polypeptide, preventing it from interacting with a second ring. Interestingly, PA5508 displays a clear preference for aromatic amine substrates. Unique aspects of the structure illustrate how the enzyme is able to catalyze reactions involving bulky amines rather than ammonia.


Subject(s)
Bacterial Proteins/chemistry , Glutamate-Ammonia Ligase/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Glutamate-Ammonia Ligase/metabolism , Models, Molecular , Polyamines/metabolism , Protein Multimerization , Pseudomonas aeruginosa/enzymology , Substrate Specificity
4.
Biochemistry ; 51(51): 10208-17, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23230967

ABSTRACT

PabB, aminodeoxychorismate synthase, is the chorismic acid binding component of the heterodimeric PabA-PabB complex that converts chorismic acid to 4-amino-4-deoxychorismate, a precursor of p-aminobenzoate and folic acid in microorganisms. The second component, a glutamine amidotransferase subunit, PabA, generates ammonia that is channeled to the PabB active site where it attacks C4 of a chorismate-derived intermediate that is covalently bound, through C2, to an active site lysine residue. The presence of a PIKGT motif was, until recently, believed to allow discrimination of PabB enzymes from the closely related enzyme anthranilate synthase, which typically contains a PIAGT active site motif and does not form a covalent enzyme-substrate intermediate with chorismate. A subclass of PabB enzymes that employ an alternative mechanism requiring 2 equiv of ammonia from glutamine and that feature a noncovalently bound 2-amino-2-deoxyisochorismate intermediate was recently identified. Here we report the 2.25 Å crystal structure of PabB from the emerging pathogen Stenotrophomonas maltophilia. It is the first reported structure of a PabB that features the PIAGT motif. Surprisingly, no dedicated pabA is evident in the genome of S. maltophilia, suggesting that another cellular amidotransferase is able to fulfill the role of PabA in this organism. Evaluation of the ammonia-dependent aminodeoxychorismate synthase activity of S. maltophilia PabB alone revealed that it is virtually inactive. However, in the presence of a heterologous PabA surrogate, typical levels of activity were observed using either glutamine or ammonia as the nitrogen source. Additionally, the structure suggests that a key segment of the polypeptide can remodel itself to interact with a nonspecialized or shared amidotransferase partner in vivo. The structure and mass spectral analysis further suggest that S. maltophilia PabB, like Escherichia coli PabB, binds tryptophan in a vestigial regulatory site. The observation that the binding site is unoccupied in the crystal structure, however, suggests the affinity may be low relative to that of E. coli PabB.


Subject(s)
Transaminases/chemistry , Binding Sites , Calorimetry , Carbon-Carbon Lyases/metabolism , Catalytic Domain , Escherichia coli Proteins/metabolism , Kinetics , Sequence Alignment , Stenotrophomonas maltophilia/enzymology , Transaminases/metabolism , Tryptophan/metabolism
5.
Proc Natl Acad Sci U S A ; 108(7): 2711-6, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21270332

ABSTRACT

Proliferating cell nuclear antigen (PCNA) is a ring-shaped protein that encircles duplex DNA and plays an essential role in many DNA metabolic processes in archaea and eukarya. The eukaryotic and euryarchaea genomes contain a single gene encoding for PCNA. Interestingly, the genome of the euryarchaeon Thermococcus kodakaraensis contains two PCNA-encoding genes (TK0535 and TK0582), making it unique among the euryarchaea kingdom. It is shown here that the two T. kodakaraensis PCNA proteins support processive DNA synthesis by the polymerase. Both proteins form trimeric structures with characteristics similar to those of other archaeal and eukaryal PCNA proteins. One of the notable differences between the TK0535 and TK0582 rings is that the interfaces are different, resulting in different stabilities for the two trimers. The possible implications of these observations for PCNA functions are discussed.


Subject(s)
DNA Replication/genetics , Models, Molecular , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/ultrastructure , Thermococcus/chemistry , Amino Acid Sequence , Cloning, Molecular , Crystallization , DNA Primers/genetics , Molecular Sequence Data , Species Specificity
6.
Biochemistry ; 50(7): 1274-81, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21222452

ABSTRACT

The crystal structure (1.50 Å resolution) and biochemical properties of the GSH transferase homologue, YghU, from Escherichia coli reveal that the protein is unusual in that it binds two molecules of GSH in each active site. The crystallographic observation is consistent with biphasic equilibrium binding data that indicate one tight (K(d1) = 0.07 ± 0.03 mM) and one weak (K(d2) = 1.3 ± 0.2 mM) binding site for GSH. YghU exhibits little or no GSH transferase activity with most typical electrophilic substrates but does possess a modest catalytic activity toward several organic hydroperoxides. Most notably, the enzyme also exhibits disulfide-bond reductase activity toward 2-hydroxyethyl disulfide [k(cat) = 74 ± 6 s(-1), and k(cat)/K(M)(GSH) = (6.6 ± 1.3) × 10(4) M(-1) s(-1)] that is comparable to that previously determined for YfcG. A superposition of the structures of the YghU·2GSH and YfcG·GSSG complexes reveals a remarkable structural similarity of the active sites and the 2GSH and GSSG molecules in each. We conclude that the two structures represent reduced and oxidized forms of GSH-dependent disulfide-bond oxidoreductases that are distantly related to glutaredoxin 2. The structures and properties of YghU and YfcG indicate that they are members of the same, but previously unidentified, subfamily of GSH transferase homologues, which we suggest be called the nu-class GSH transferases.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Glutathione Transferase/chemistry , Glutathione Transferase/physiology , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Models, Biological , Models, Molecular , Molecular Dynamics Simulation , Multigene Family , Phylogeny , Protein Structure, Secondary , Sequence Homology
7.
Biochemistry ; 48(28): 6559-61, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19537707

ABSTRACT

YfcG is one of eight glutathione (GSH) transferase homologues encoded in the Escherichia coli genome. The protein exhibits low or no GSH transferase activity toward a panel of electrophilic substrates. In contrast, it has a very robust disulfide-bond reductase activity toward 2-hydroxyethyldisulfide on par with mammalian and bacterial glutaredoxins. The structure of YfcG at 2.3 A-resolution from crystals grown in the presence of GSH reveals a molecule of glutathione disulfide in the active site. The crystallographic results and the lack of functional cysteine residues in the active site of YfcG suggests that the reductase activity is unique in that no sulfhydryl groups in the YfcG protein are covalently involved in the redox chemistry.


Subject(s)
Disulfides/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Oxidoreductases/metabolism , Binding Sites , Fluorescence , Glutathione Disulfide/metabolism , Kinetics , Ligands , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Multimerization , Protein Structure, Secondary , Static Electricity , Substrate Specificity , Sulfhydryl Compounds/metabolism , Temperature , Titrimetry
8.
FEBS J ; 275(19): 4824-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18727669

ABSTRACT

The Rv0948c gene from Mycobacterium tuberculosis H(37)R(v) encodes a 90 amino acid protein as the natural gene product with chorismate mutase (CM) activity. The protein, 90-MtCM, exhibits Michaelis-Menten kinetics with a k(cat) of 5.5+/-0.2s(-1) and a K(m) of 1500+/-100microm at 37 degrees C and pH7.5. The 2.0A X-ray structure shows that 90-MtCM is an all alpha-helical homodimer (Protein Data Bank ID: 2QBV) with the topology of Escherichia coli CM (EcCM), and that both protomers contribute to each catalytic site. Superimposition onto the structure of EcCM and the sequence alignment shows that the C-terminus helix3 is shortened. The absence of two residues in the active site of 90-MtCM corresponding to Ser84 and Gln88 of EcCM appears to be one reason for the low k(cat). Hence, 90-MtCM belongs to a subfamily of alpha-helical AroQ CMs termed AroQ(delta.) The CM gene (y2828) from Yersinia pestis encodes a 186 amino acid protein with an N-terminal signal peptide that directs the protein to the periplasm. The mature protein, *YpCM, exhibits Michaelis-Menten kinetics with a k(cat) of 70+/-5s(-1) and K(m) of 500+/-50microm at 37 degrees C and pH7.5. The 2.1A X-ray structure shows that *YpCM is an all alpha-helical protein, and functions as a homodimer, and that each protomer has an independent catalytic unit (Protein Data Bank ID: 2GBB). *YpCM belongs to the AroQ(gamma) class of CMs, and is similar to the secreted CM (Rv1885c, *MtCM) from M.tuberculosis.


Subject(s)
Chorismate Mutase/chemistry , Chorismate Mutase/classification , Chorismate Mutase/metabolism , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Kinetics , Models, Molecular , Mycobacterium tuberculosis/enzymology , Yersinia pestis/enzymology
9.
Acta Crystallogr D Biol Crystallogr ; 64(Pt 5): 607-10, 2008 May.
Article in English | MEDLINE | ID: mdl-18453696

ABSTRACT

The electron carrier menaquinone is one of many important bacterial metabolites that are derived from the key intermediate chorismic acid. MenF, the first enzyme in the menaquinone pathway, catalyzes the isomerization of chorismate to isochorismate. Here, an improved structure of MenF in a new crystal form is presented. The structure, solved at 2.0 angstroms resolution in complex with magnesium, reveals a well defined closed active site. Existing evidence suggests that the mechanism of the reaction catalyzed by MenF involves nucleophilic attack of a water molecule on the chorismate ring. The structure reveals a well defined water molecule located in an appropriate position for activation by Lys190 and attack on the substrate.


Subject(s)
Escherichia coli Proteins/chemistry , Intramolecular Transferases/chemistry , Magnesium/chemistry , Crystallography, X-Ray/methods , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Intramolecular Transferases/genetics , Intramolecular Transferases/metabolism , Magnesium/metabolism , Models, Molecular , Molecular Structure , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
10.
Biochemistry ; 47(19): 5281-9, 2008 May 13.
Article in English | MEDLINE | ID: mdl-18416536

ABSTRACT

The human pathogen Pseudomonas aeruginosa produces pyocyanin, a blue-pigmented phenazine derivative, which is known to play a role in virulence. Pyocyanin is produced from chorismic acid via the phenazine pathway, nine proteins encoded by a gene cluster. Phenazine-1-carboxylic acid, the initial phenazine formed, is converted to pyocyanin in two steps that are catalyzed by the enzymes PhzM and PhzS. PhzM is an adenosylmethionine dependent methyltransferase, and PhzS is a flavin dependent hydroxylase. It has been shown that PhzM is only active in the physical presence of PhzS, suggesting that a protein-protein interaction is involved in pyocyanin formation. Such a complex would prevent the release of 5-methyl-phenazine-1-carboxylate, the putative intermediate, and an apparently unstable compound. Here, we describe the three-dimensional structure of PhzS, solved by single anomalous dispersion, at a resolution of 2.4 A. The structure reveals that PhzS is a member of the family of aromatic hydroxylases characterized by p-hydroxybenzoate hydroxylase. The flavin cofactor of PhzS is in the solvent exposed out orientation typically seen in unliganded aromatic hydroxylases. The PhzS flavin, however, appears to be held in a strained conformation by a combination of stacking interactions and hydrogen bonds. The structure suggests that access to the active site is gained via a tunnel on the opposite side of the protein from where the flavin is exposed. The C-terminal 23 residues are disordered as no electron density is present for these atoms. The probable location of the C-terminus, near the substrate access tunnel, suggests that it may be involved in substrate binding as has been shown for another structural homologue, RebC. This region also may be an element of a PhzM-PhzS interface. Aromatic hydroxylases have been shown to catalyze electrophilic substitution reactions on activated substrates. The putative PhzS substrate, however, is electron deficient and unlikely to act as a nucleophile, suggesting that PhzS may use a different mechanism than its structural relatives.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/metabolism , Pyocyanine/chemistry , Pyocyanine/metabolism , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , Mass Spectrometry , Mixed Function Oxygenases/genetics , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein , Substrate Specificity
11.
Biochemistry ; 46(23): 6710-22, 2007 Jun 12.
Article in English | MEDLINE | ID: mdl-17508726

ABSTRACT

The enzyme 2-hydroxychromene-2-carboxylic acid (HCCA) isomerase catalyzes the glutathione (GSH)-dependent interconversion (Keq = 1.5) of HCCA and trans-o-hydroxybenzylidene pyruvic acid (tHBPA) in the naphthalene catabolic pathway of Pseudomonas putida. The dimeric protein binds one molecule of GSH very tightly (Kd approximately 5 nM) and a second molecule of GSH with much lower affinity (Kd approximately 2 to 11 microM). The enzyme is unstable in the absence of GSH. The turnover number in the forward direction (47 s(-1) at 25 degrees C) greatly exceeds off rates for GSH (koff approximately 10(-3) to 10(-2) s(-1) at 10 degrees C), suggesting that GSH acts as a tightly bound cofactor in the reaction. The crystal structure of the enzyme at 1.7 A resolution reveals that the isomerase is closely related to class kappa GSH transferases. Diffraction quality crystals could only be obtained in the presence of GSH and HCCA/tHBPA. Clear electron density is seen for GSH. Electron density for the organic substrates is located near the GSH and is best modeled to include both HCCA and tHBPA at occupancies of 0.5 for each. Although there is no electron density connecting the sulfur of GSH to the organic substrates, the sulfur is located very close (2.78 A) to C7 of HCCA. Taken together, the results suggest that the isomerization reaction involves a short-lived covalent adduct between the sulfur of GSH and C7 of the substrate.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Pseudomonas putida/enzymology , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Glutathione/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Intramolecular Oxidoreductases/chemistry , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
12.
Biochemistry ; 46(7): 1821-8, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17253782

ABSTRACT

Pyocyanin is a biologically active phenazine produced by the human pathogen Pseudomonas aeruginosa. It is thought to endow P. aeruginosa with a competitive growth advantage in colonized tissue and is also thought to be a virulence factor in diseases such as cystic fibrosis and AIDS where patients are commonly infected by pathogenic Pseudomonads due to their immunocompromised state. Pyocyanin is also a chemically interesting compound due to its unusual oxidation-reduction activity. Phenazine-1-carboxylic acid, the precursor to the bioactive phenazines, is synthesized from chorismic acid by enzymes encoded in a seven-gene cistron in P. aeruginosa and in other Pseudomonads. Phenzine-1-carboxylic acid is believed to be converted to pyocyanin by the sequential actions of the putative S-adenosylmethionine-dependent N-methyltransferase PhzM and the putative flavin-dependent hydroxylase PhzS. Here we report the 1.8 A crystal structure of PhzM determined by single anomalous dispersion. Unlike many methyltransferases, PhzM is a dimer in solution. The 36 kDa PhzM polypeptide folds into three domains. The C-terminal domain exhibits the alpha/beta-hydrolase fold typical of small molecule methyltransferases. Two smaller N-terminal domains form much of the dimer interface. Structural alignments with known methyltransferases show that PhzM is most similar to the plant O-methyltransferases that are characterized by an unusual intertwined dimer interface. The structure of PhzM contains no ligands, and the active site is open and solvent-exposed when compared to structures of similar enzymes. In vitro experiments using purified PhzM alone demonstrate that it has little or no ability to methylate phenzine-1-carboxylic acid. However, when the putative hydroxylase PhzS is included, pyocyanin is readily produced. This observation suggests that a mechanism has evolved in P. aeruginosa that ensures efficient production of pyocyanin via the prevention of the formation and release of an unstable and potentially deleterious intermediate.


Subject(s)
Bacterial Proteins/chemistry , Methyltransferases/chemistry , Pseudomonas aeruginosa/metabolism , Pyocyanine/biosynthesis , Bacterial Proteins/physiology , Binding Sites , Crystallography, X-Ray , Dimerization , Methyltransferases/physiology , Mixed Function Oxygenases/chemistry , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Pseudomonas aeruginosa/enzymology , Pyocyanine/chemical synthesis , Solutions
13.
J Bacteriol ; 188(24): 8638-48, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17146044

ABSTRACT

The gene Rv1885c from the genome of Mycobacterium tuberculosis H37Rv encodes a monofunctional and secreted chorismate mutase (*MtCM) with a 33-amino-acid cleavable signal sequence; hence, it belongs to the *AroQ class of chorismate mutases. Consistent with the heterologously expressed *MtCM having periplasmic destination in Escherichia coli and the absence of a discrete periplasmic compartment in M. tuberculosis, we show here that *MtCM secretes into the culture filtrate of M. tuberculosis. *MtCM functions as a homodimer and exhibits a dimeric state of the protein at a concentration as low as 5 nM. *MtCM exhibits simple Michaelis-Menten kinetics with a Km of 0.5 +/- 0.05 mM and a k(cat) of 60 s(-1) per active site (at 37 degrees C and pH 7.5). The crystal structure of *MtCM has been determined at 1.7 A resolution (Protein Data Bank identifier 2F6L). The protein has an all alpha-helical structure, and the active site is formed within a single chain without any contribution from the second chain in the dimer. Analysis of the structure shows a novel fold topology for the protein with a topologically rearranged helix containing Arg134. We provide evidence by site-directed mutagenesis that the residues Arg49, Lys60, Arg72, Thr105, Glu109, and Arg134 constitute the catalytic site; the numbering of the residues includes the signal sequence. Our investigation on the effect of phenylalanine, tyrosine, and tryptophan on *MtCM shows that *MtCM is not regulated by the aromatic amino acids. Consistent with this observation, the X-ray structure of *MtCM does not have an allosteric regulatory site.


Subject(s)
Amino Acids, Aromatic/pharmacology , Chorismate Mutase , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Catalytic Domain , Chorismate Mutase/chemistry , Chorismate Mutase/genetics , Chorismate Mutase/metabolism , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/genetics
14.
Article in English | MEDLINE | ID: mdl-16511320

ABSTRACT

Bacteria have evolved elaborate schemes that help them thrive in environments where free iron is severely limited. Siderophores such as yersiniabactin are small iron-scavenging molecules that are deployed by bacteria during iron starvation. Several studies have linked siderophore production and virulence. Yersiniabactin, produced by several Enterobacteriaceae, is derived from the key metabolic intermediate chorismic acid via its conversion to salicylate by salicylate synthase. Crystals of salicylate synthase from the uropathogen Escherichia coli CFT073 have been grown by vapour diffusion using polyethylene glycol as the precipitant. The monoclinic (P2(1)) crystals diffract to 2.5 A. The unit-cell parameters are a = 57.27, b = 164.07, c = 59.04 A, beta = 108.8 degrees. The solvent content of the crystals is 54% and there are two molecules of the 434-amino-acid protein in the asymmetric unit. It is anticipated that the structure will reveal key details about the reaction mechanism and the evolution of salicylate synthase.


Subject(s)
Escherichia coli/enzymology , Lyases/chemistry , Chorismic Acid/metabolism , Crystallization/methods , Crystallography, X-Ray , Siderophores/biosynthesis
15.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 11): 2110-3, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15502343

ABSTRACT

PhzG is a flavin-dependent oxidase that is believed to play a role in phenazine antibiotic synthesis in various bacteria, including Pseudomonas. Phenazines are chorismic acid derivatives that provide the producing organisms, including the opportunistic pathogen P. aeruginosa, with a competitive growth advantage. Here, the crystal structures of PhzG from both P. aeruginosa and P. fluorescens solved in an unliganded state at 1.9 and 1.8 A resolution, respectively, are described. Although the specific reaction in phenazine biosynthesis catalyzed by PhzG is unknown, the structural data indicates that PhzG is closely related to pyridoxine-5'-phosphate oxidase, the Escherichia coli pdxH gene product, which catalyzes the final step in pyridoxal-5'-phosphate (PLP) biosynthesis. A previous proposal suggested that the physiological substrate of PhzG to be 2,3-dihydro-3-hydroxyanthranilic acid (DHHA), a phenazine precursor produced by the sequential actions of the PhzE and PhzD enzymes on chorismate, and that two DHHA molecules dimerized in another enzyme-catalyzed reaction to yield phenazine-1-carboxylate. However, it was not possible to demonstrate any in vitro activity upon incubation of PhzG and DHHA. Interestingly, analysis of the in vitro activities of PhzG in combination with PhzF suggests that PhzF acts on DHHA and that PhzG then reacts with a non-aromatic tricyclic phenazine precusor to catalyze an oxidation/aromatization reaction that yields phenazine-1-carboxylate. It is proposed that phzG arose by duplication of pdxH and that the subtle differences seen between the structures of PhzG and PdxH correlate with the loss of the ability of PhzG to catalyze PLP formation. Sequence alignments and superimpositions of the active sites of PhzG and PdxH reveal that the residues that form a positively charged pocket around the phosphate of PLP in the PdxH-PLP complex are not conserved in PhzG, consistent with the inability of phosphorylated compounds to serve as substrates for PhzG.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Phenazines/metabolism , Pseudomonas aeruginosa/enzymology , Pseudomonas fluorescens/enzymology , Binding Sites , Dimerization , Flavins/pharmacology , Models, Molecular , Molecular Structure , Phenazines/chemistry , Protein Structure, Tertiary , Pyridoxaminephosphate Oxidase/chemistry , Structural Homology, Protein
16.
Biochemistry ; 43(39): 12427-35, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15449932

ABSTRACT

Phenazines, including pyocyanin and iodonin, are biologically active compounds that are believed to confer producing organisms with a competitive growth advantage, and also are thought to be virulence factors in certain diseases including cystic fibrosis. The basic, tricyclic phenazine ring system is synthesized in a series of poorly characterized steps by enzymes encoded in a seven-gene cistron in Pseudomonas and other organisms. Despite the biological importance of these compounds, and our understanding of their mode of action, the biochemistry and mechanisms of phenazine biosynthesis are not well resolved. Here we report the 1.8 A crystal structure of PhzF, a key enzyme in phenazine biosynthesis, solved by molecular replacement. PhzF is structurally similar to the lysine biosynthetic enzyme diaminopimelate epimerase, sharing an unusual fold consisting of two nearly identical domains with the active site located in an occluded cleft between the domains. Unlike diaminopimelate epimerase, PhzF is a dimer in solution. The two apparently independent active sites open toward opposite sides of the dimer and are occupied by sulfate ions in the structure. In vitro experiments using a mixture of purified PhzF, -A, -B, and -G confirm that phenazine-1-carboxylic acid (PCA) is readily produced from trans-2,3-dihydro-3-hydroxyanthranilic acid (DHHA) without aid of other cellular factors. PhzA, -B, and -G have no activity toward DHHA. However, in the presence of PhzF, individually or in combinations, they accelerate the formation of PCA from DHHA and therefore appear to function after the action of PhzF. Surprisingly, PhzF is itself capable of producing PCA, albeit slowly, from DHHA. These observations suggest that PhzF catalyzes the initial step in the conversion of DHHA to PCA, probably via a rearrangement reaction yielding the more reactive 3-oxo analogue of DHHA, and that subsequent steps can occur spontaneously. A hypothetical model for how DHHA binds to the PhzF active site suggests that Glu45 and Asp208 could act as general acid-base catalysts in a rearrangement reaction. Given that four reactions lie between DHHA and PCA, ketone formation, ring formation, decarboxylation, and oxidation, we hypothesize that the similar PhzA and -B proteins catalyze ring formation and thus may be more than noncatalytic accessory proteins. PhzG is almost certainly an oxidase and is predicted to catalyze the final oxidation/aromatization reaction.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Pseudomonas fluorescens/chemistry , Trans-Activators/chemistry , Trans-Activators/physiology , 3-Hydroxyanthranilic Acid/chemistry , Bacterial Proteins/genetics , Binding Sites , Catalysis , Chorismic Acid/chemistry , Crystallization , Crystallography, X-Ray , DNA-Binding Proteins/genetics , Deuterium Exchange Measurement , Dimerization , Magnetic Resonance Spectroscopy , Oxidation-Reduction , Phenazines/chemistry , Phenazines/metabolism , Pseudomonas fluorescens/genetics , Spectrophotometry, Ultraviolet , Structure-Activity Relationship , Substrate Specificity , Surface Properties , Trans-Activators/genetics
17.
Biochemistry ; 43(2): 352-61, 2004 Jan 20.
Article in English | MEDLINE | ID: mdl-14717589

ABSTRACT

The class kappa glutathione (GSH) transferase is an enzyme that resides in the mitochondrial matrix. Its relationship to members of the canonical GSH transferase superfamily has remained an enigma. The three-dimensional structure of the class kappa enzyme from rat (rGSTK1-1) in complex with GSH has been solved by single isomorphous replacement with anomalous scattering at a resolution of 2.5 A. The structure reveals that the enzyme is more closely related to the protein disulfide bond isomerase, dsbA, from Escherichia coli than it is to members of the canonical superfamily. The structures of rGSTK1-1 and the canonical superfamily members indicate that the proteins folds have diverged from a common thioredoxin/glutaredoxin progenitor but did so by different mechanisms. The mitochondrial enzyme, therefore, represents a fourth protein superfamily that supports GSH transferase activity. The thioredoxin domain functions in a manner that is similar to that seen in the canonical enzymes by providing key structural elements for the recognition of GSH. The hydroxyl group of S16 is within hydrogen-bonding distance of the sulfur of bound GSH and is, in part, responsible for the ionization of the thiol in the E*GSH complex (pKa = 6.4 +/- 0.1). Preequilibrium kinetic experiments indicate that the k(on) for GSH is 1 x 10(5) M(-1) s(-1) and k(off) for GS- is approximately 8 s(-1) and relatively slow with respect to turnover with 1-chloro-2, 4-dinitrobenzene (CDNB). As a result, the KM(GSH) (11 mM) is much larger than the apparent Kd(GSH) (90 microM). The active site has a relatively open access channel that is flanked by disordered loops that may explain the relatively high turnover number (280 s(-1) at pH 7.0) toward CDNB. The disordered loops form an extensive contiguous patch on one face of the dimeric enzyme, a fact that suggests that the protein surface may interact with a membrane or other protein partner.


Subject(s)
Evolution, Molecular , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Mitochondria/enzymology , Amino Acid Substitution/genetics , Animals , Binding Sites , Catalysis , Crystallography, X-Ray , Dimerization , Glutathione/chemistry , Glutathione/metabolism , Glutathione Transferase/genetics , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Serine/genetics , Structure-Activity Relationship
18.
BMC Struct Biol ; 3: 7, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-14519207

ABSTRACT

BACKGROUND: The protein encoded by the gene ybgI was chosen as a target for a structural genomics project emphasizing the relation of protein structure to function. RESULTS: The structure of the ybgI protein is a toroid composed of six polypeptide chains forming a trimer of dimers. Each polypeptide chain binds two metal ions on the inside of the toroid. CONCLUSION: The toroidal structure is comparable to that of some proteins that are involved in DNA metabolism. The di-nuclear metal site could imply that the specific function of this protein is as a hydrolase-oxidase enzyme.


Subject(s)
Escherichia coli Proteins/chemistry , Metals/chemistry , Models, Molecular , Binding Sites , Crystallography, X-Ray , Dimerization , Escherichia coli Proteins/metabolism , Metals/metabolism
19.
Biochemistry ; 42(19): 5684-93, 2003 May 20.
Article in English | MEDLINE | ID: mdl-12741825

ABSTRACT

PhzD from Pseudomonas aeruginosa is an isochorismatase involved in phenazine biosynthesis. Phenazines are antimicrobial compounds that provide Pseudomonas with a competitive advantage in certain environments and may be partly responsible for the persistence of Pseudomonas infections. In vivo, PhzD catalyzes the hydrolysis of the vinyl ether functional group of 2-amino-2-deoxyisochorismate, yielding pyruvate and trans-2,3-dihydro-3-hydroxyanthranilic acid, which is then utilized in the phenazine biosynthetic pathway. PhzD also catalyzes hydrolysis of the related vinyl ethers isochorismate, chorismate, and 4-amino-4-deoxychorismate. Here we report the 1.5 A crystal structure of native PhzD, and the 1.6 A structure of the inactive D38A variant in complex with isochorismate. The structures reveal that isochorismate binds to the PhzD active site in a trans-diaxial conformation, and superposition of the structures indicates that the methylene pyruvyl carbon of isochorismate is adjacent to the side chain carboxylate of aspartate 38. The proximity of aspartate 38 to isochorismate and the complete loss of activity resulting from the conversion of aspartate 38 to alanine suggest a mechanism in which the carboxylate acts as a general acid to protonate the substrate, yielding a carbocation/oxocarbonium ion that is then rapidly hydrated to form a hemiketal intermediate, which then decomposes spontaneously to products. The structure of PhzD is remarkably similar to other structures from a subfamily of alpha/beta-hydrolase enzymes that includes pyrazinamidase and N-carbamoylsarcosine amidohydrolase. However, PhzD catalyzes unrelated chemistry and lacks a nucleophilic cysteine found in its close structural relatives. The vinyl ether hydrolysis catalyzed by PhzD represents yet another example of the catalytic diversity seen in the alpha/beta-hydrolase family, whose members are also known to hydrolyze amides, phosphates, phosphonates, epoxides, and C-X bonds.


Subject(s)
Hydrolases/chemistry , Hydrolases/metabolism , Phenazines/metabolism , Pseudomonas aeruginosa/enzymology , Catalytic Domain , Chorismic Acid/chemistry , Chorismic Acid/metabolism , Crystallography, X-Ray , Cyclohexenes , Dimerization , Hydrolases/genetics , Kinetics , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Phenazines/chemistry , Protein Conformation , Protein Structure, Quaternary , Pseudomonas aeruginosa/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Biochemistry ; 41(51): 15161-72, 2002 Dec 24.
Article in English | MEDLINE | ID: mdl-12484753

ABSTRACT

Glutathione transferase rGSTM1-1 catalyzes the addition of glutathione (GSH) to 1-chloro-2,4-dinitrobenzene, a reaction in which the chemical step is 60-fold faster than the physical step of product release. The hydroxyl group of Y115, located in the active site access channel, controls the egress of product from the active site. The Y115F mutant enzyme has a k(cat) (72 s(-)(1)) that is 3.6-fold larger than that of the native enzyme (20 s(-)(1)). Crystallographic observations and evidence from amide proton exchange kinetics are consistent with localized increases in the degree of segmental motion of the Y115F mutant that are coupled to the enhanced rate of product release. The loss of hydrogen bonding interactions involving the hydroxyl group of Y115 is reflected in subtle alterations in the backbone position, an increase in B-factors for structural elements that comprise the channel to the active site, and, most dramatically, a loss of well-defined electron density near the site of mutation. The kinetics of amide proton exchange are also enhanced by a factor between 3 and 12 in these regions, providing direct, quantitative evidence for changes in local protein dynamics affecting product release. The enhanced product release rate is proposed to derive from a small shift in the equilibrium population of protein conformers that permit egress of the product from the active site.


Subject(s)
Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Thermodynamics , Amides/chemistry , Amino Acid Substitution/genetics , Animals , Catalysis , Circular Dichroism , Crystallization , Crystallography, X-Ray , Deuterium Oxide/chemistry , Dinitrochlorobenzene/chemistry , Dinitrochlorobenzene/metabolism , Glutathione Transferase/genetics , Kinetics , Mutagenesis, Site-Directed , Phenylalanine/genetics , Protein Conformation , Protons , Rats , Spectrometry, Fluorescence , Structure-Activity Relationship , Substrate Specificity/genetics , Tyrosine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...