Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 16451, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385485

ABSTRACT

Trans-2,3-dihydro-3-hydroxyanthranilic acid (DHHA) is a cyclic ß-amino acid used for the synthesis of non-natural peptides and chiral materials. And it is an intermediate product of phenazine production in Pseudomonas spp. Lzh-T5 is a P. chlororaphis strain isolated from tomato rhizosphere found in China. It can synthesize three antifungal phenazine compounds. Disruption the phzF gene of P. chlororaphis Lzh-T5 results in DHHA accumulation. Several strategies were used to improve production of DHHA: enhancing the shikimate pathway by overexpression, knocking out negative regulatory genes, and adding metal ions to the medium. In this study, three regulatory genes (psrA, pykF, and rpeA) were disrupted in the genome of P. chlororaphis Lzh-T5, yielding 5.52 g/L of DHHA. When six key genes selected from the shikimate, pentose phosphate, and gluconeogenesis pathways were overexpressed, the yield of DHHA increased to 7.89 g/L. Lastly, a different concentration of Fe3+ was added to the medium for DHHA fermentation. This genetically engineered strain increased the DHHA production to 10.45 g/L. According to our result, P. chlororaphis Lzh-T5 could be modified as a microbial factory to produce DHHA. This study laid a good foundation for the future industrial production and application of DHHA.


Subject(s)
3-Hydroxyanthranilic Acid/metabolism , Pseudomonas chlororaphis/genetics , 3-Hydroxyanthranilic Acid/chemistry , Culture Media , Fermentation , Ferric Compounds/metabolism , Gene Knockdown Techniques , Genes, Bacterial , Genes, Regulator , Phenazines/metabolism
2.
Biochem Biophys Res Commun ; 543: 8-14, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33493986

ABSTRACT

Paulomycins (PAUs) refer to a group of glycosylated antibiotics with attractive antibacterial activities against Gram-positive bacteria. They contain a special ring A moiety that is prone to dehydrate between C-4 and C-5 to a quinone-type form at acidic condition, which will reduce the antibacterial activities of PAUs significantly. Elucidation of the biosynthetic mechanism of the ring A moiety may facilitate its structure modifications by combinatorial biosynthesis to generate PAU analogues with enhanced bioactivity or stability. Previous studies showed that the ring A moiety is derived from chorismate, which is converted to 3-hydroxyanthranilic acid (3-HAA) by a 2-amino-2-deoxyisochorismate (ADIC) synthase, a 2,3-dihydro-3-hydroxyanthranilic acid (DHHA) synthase, and a DHHA dehydrogenase. Unfortunately, little is known about the conversion process from 3-HAA to the highly decorated ring A moiety of PAUs. In this work, we characterized Pau17 as an unprecedented 3-HAA 6-hydroxylase responsible for the conversion of 3-HAA to 3,6-DHAA by in vivo and in vitro studies, pushing one step forward toward elucidating the biosynthetic mechanism of the ring A moiety of PAUs.


Subject(s)
3-Hydroxyanthranilic Acid/metabolism , Anti-Bacterial Agents/biosynthesis , Cyclohexenes/metabolism , Disaccharides/biosynthesis , Mixed Function Oxygenases/metabolism , Streptomyces/enzymology , 3-Hydroxyanthranilic Acid/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cyclohexenes/chemistry , Cyclohexenes/pharmacology , Disaccharides/chemistry , Disaccharides/pharmacology , Gram-Positive Bacteria/drug effects , Mixed Function Oxygenases/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Streptomyces/chemistry , Streptomyces/genetics
3.
Article in English | MEDLINE | ID: mdl-31586884

ABSTRACT

A sensitive, rapid and reliable ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed and validated to assay tryptophan (TRP) and its nine metabolites, including kynurenine (KYN), kynurenic acid (KYNA), 3-hydroxykynurenine (3-HK), 3-hydroxyanthranilic acid (3-HAA), xanthurenic acid (XA), 5-hydroxytryptamine (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), 3-indolepropionic acid (IPA) and 3-indoleacetic acid (IAA) in human plasma. Tryptophan-d5 (TRP-d5) and carbamazepine (CAR) were applied to the method quantification, where TRP-d5 was the corresponding internal standard (IS) for TRP and KYN, and CAR was the corresponding IS for the other analytes. Plasma samples were processed by deproteinisation with acetonitrile, followed by separation on an Acquity UPLC HSS T3 column by using gradient elution with 0.1% (v/v) formic acid in water and acetonitrile and detection by electrospray ionisation tandem mass spectrometry in positive ion multiple reaction monitoring (MRM) within a total run time of 5 min. The calibration ranges were 3-600 ng/mL for 3-HK, 1.5-300 ng/mL for 5-HT, 25-5000 ng/mL for KYN, 1-200 ng/mL for XA, 100-20,000 ng/mL for TRP, 5-1000 ng/mL for KYNA, 2-400 ng/mL for 3-HAA, 2.5-500 ng/mL for 5-HIAA and 10-2000 ng/mL for IAA and IPA. All intra- and inter-day analytical variations were acceptable. Matrix effect and recovery evaluation proved that matrix effect can be negligible, and sample preparation approach was effective. The newly developed method can simultaneously determine a panel of TRP metabolites and was successfully applied in the clinical study characterising TRP metabolism in healthy volunteers.


Subject(s)
Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Tryptophan/blood , Tryptophan/metabolism , 3-Hydroxyanthranilic Acid/analysis , 3-Hydroxyanthranilic Acid/chemistry , 3-Hydroxyanthranilic Acid/metabolism , Adult , Female , Humans , Kynurenic Acid/blood , Kynurenic Acid/chemistry , Kynurenic Acid/metabolism , Linear Models , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity , Tryptophan/chemistry
4.
Article in English | MEDLINE | ID: mdl-31067822

ABSTRACT

The fungal metabolite 3-hydroxyanthranilic acid (3-HAA) was used as a redox mediatorwith the aim of increasing dye degradation by Fenton oxidative processes (Fe2+/H2O2, Fe3+/H2O2). ItsFe3+-reducing activity can enhance the generation of reactive oxygen species as HO● radicals.Initially, the influence of 3-HAA on decolorization kinetics of five dyes (methylene blue,chromotrope 2R, methyl orange, phenol red, and safranin T) was investigated using decolorizationdata from a previous work conducted by the present research group. Fe3+-containing reaction datawere well fitted with first-order and mainly second-order kinetic models, whereas the BMG(Behnajady, Modirshahla and Ghanbary) model obtained optimal fit to Fe2+. Improvements inkinetic parameters (i.e., apparent rate constants and maximum oxidation capacity) were observedwith the addition of 3-HAA. In another set of experiments, a decrease in apparent activation energywas observed due to introducing 3-HAA into reactions containing either Fe2+ or Fe3+ in order todecolorize phenol red at different temperatures. This indicates that the redox mediator decreasesthe energy barrier so as to allow reactions to occur. Thus, based on recent experiments and thereaction kinetics models evaluated herein, pro-oxidant properties have been observed for 3-HAAin Fenton processes.


Subject(s)
3-Hydroxyanthranilic Acid/chemistry , Coloring Agents/chemistry , Hydrogen Peroxide/chemistry , Iron/chemistry , Water Pollutants, Chemical/chemistry , Azo Compounds/chemistry , Color , Kinetics , Methylene Blue/chemistry , Naphthalenesulfonates/chemistry , Oxidation-Reduction , Phenazines/chemistry , Phenolsulfonphthalein/chemistry
5.
Int J Biol Macromol ; 122: 210-215, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30365991

ABSTRACT

3-Hydroxyanthranilic acid (3-HAA), as a potential natural laccase mediator, was shown to mediate the oxidation of non-phenolic lignin subunits. The problem of cost and toxicity of artificial mediators could be solved to some extent by a further study about the detailed changes of lignin chemistry structures in laccase 3-HAA system (LHS). In this work, wheat straw auto-hydrolysis lignin (AL) was prepared. Oxidations of AL by LHS and laccase 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid) (ABTS) system were then investigated, respectively. Various structural changes of AL during the oxidation were characterized by different methods including phenolic hydroxyl group determination, nitrobenzene oxidation, ozonation, gel permeation chromatography, ultraviolet-visible spectroscopy, Fourier transform infrared spectroscopy and two-dimensional nuclear magnetic resonance (2D NMR) spectroscopy. The changes in AL chemical structures were found in LHS, including unit removal, bond cleavage and biopolymerization. Compared to laccase ABTS system, a selective removal of guaiacyl-type lignin in LHS was observed, based on the results of nitrobenzene oxidation and 2D NMR analysis. The selective removal of guaiacyl-type lignin was due to improved aromatic ring cleavage and weaken lignin biopolymerization. The selectivity of guaiacyl-type lignin removal in LHS plays an important role, especially for improving bioconversion efficiency of laccase for guaiacyl-rich lignocellulosic biomass.


Subject(s)
3-Hydroxyanthranilic Acid/chemistry , 3-Hydroxyanthranilic Acid/pharmacology , Laccase/metabolism , Lignin/chemistry , Triticum/chemistry , Hydrolysis , Molecular Weight , Nitrobenzenes/chemistry , Oxidation-Reduction , Ozone/chemistry , Phenols/chemistry , Trametes/enzymology
6.
J Biol Chem ; 293(27): 10415-10424, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29784877

ABSTRACT

3-Hydroxyanthranilate 3,4-dioxygenase (HAO) is an iron-dependent protein that activates O2 and inserts both oxygen atoms into 3-hydroxyanthranilate (3-HAA). An intriguing question is how HAO can rapidly bind O2, even though local O2 concentrations and diffusion rates are relatively low. Here, a close inspection of the HAO structures revealed that substrate- and inhibitor-bound structures exhibit a closed conformation with three hydrophobic loop regions moving toward the catalytic iron center, whereas the ligand-free structure is open. We hypothesized that these loop movements enhance O2 binding to the binary complex of HAO and 3-HAA. We found that the carboxyl end of 3-HAA triggers changes in two loop regions and that the third loop movement appears to be driven by an H-bond interaction between Asn27 and Ile142 Mutational analyses revealed that N27A, I142A, and I142P variants cannot form a closed conformation, and steady-state kinetic assays indicated that these variants have a substantially higher Km for O2 than WT HAO. This observation suggested enhanced hydrophobicity at the iron center resulting from the concerted loop movements after the binding of the primary substrate, which is hydrophilic. Given that O2 is nonpolar, the increased hydrophobicity at the iron center of the binary complex appears to be essential for rapid O2 binding and activation, explaining the reason for the 3-HAA-induced loop movements. Because substrate binding-induced open-to-closed conformational changes are common, the results reported here may help further our understanding of how oxygen is enriched in nonheme iron-dependent dioxygenases.


Subject(s)
3-Hydroxyanthranilic Acid/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cupriavidus/enzymology , Dioxygenases/chemistry , Dioxygenases/metabolism , Oxygen/metabolism , 3-Hydroxyanthranilic Acid/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Catalysis , Catalytic Domain , Crystallography, X-Ray , Dioxygenases/genetics , Ligands , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Sequence Homology , Substrate Specificity
7.
Appl Microbiol Biotechnol ; 101(17): 6607-6613, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28702795

ABSTRACT

Trans-2,3-dihydro-3-hydroxyanthranilic acid (DHHA) is a cyclic ß-amino acid that can be used for the synthesis of chiral materials and nonnatural peptides. The aim of this study was to accumulate DHHA by engineering Pseudomonas chlororaphis GP72, a nonpathogenic strain that produces phenazine-1-carboxylic acid and 2-hydroxyphenazine. First, the phzF deletion mutant DA1 was constructed, which produced 1.91 g/L DHHA. Moreover, rpeA and pykF were disrupted and then ppsA and tktA were co-expressed in strain DA1. The resulting strain DA4 increased DHHA concentration to 4.98 g/L, which is 2.6-fold than that of DA1. The effects of the addition of glucose, glycerol, L-tryptophan, and Fe3+on DHHA production were also investigated. Strain DA4 produced 7.48 g/L of DHHA in the culture medium in the presence of 12 g/L glucose and 3 mM Fe3+, which was 1.5-fold higher than the strain in the original fermentation conditions. These results indicate the potential of P. chlororaphis GP72 as a DHHA producer.


Subject(s)
3-Hydroxyanthranilic Acid/analysis , 3-Hydroxyanthranilic Acid/metabolism , Pseudomonas chlororaphis/genetics , Pseudomonas chlororaphis/metabolism , 3-Hydroxyanthranilic Acid/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Culture Media/chemistry , Glucose/pharmacology , Metabolic Engineering/methods , Nitrogenous Group Transferases/genetics , Phenazines/metabolism , Pseudomonas chlororaphis/drug effects , Sequence Deletion
8.
Biopolymers ; 91(12): 1189-95, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19226621

ABSTRACT

3-Hydroxyanthranilate 3,4-dioxygenase, the enzyme that catalyzes the conversion of 3-hydroxyanthranilate to quinolinic acid, has been extracted and purified from bovine kidney, crystallized and its structure determined at 2.5 A resolution. The enzyme, which crystallizes in the triclinic P1 space group, is a monomer, characterized by the so-called cupin fold. The monomer of the bovine enzyme mimics the dimer present in lower species, such as bacteria and yeast, since it is composed of two domains: one of them is equivalent to one monomer, whilst the second domain corresponds to only a portion of it. The active site consists of an iron ion coordinated by two histidine residues, one glutamate and an external ligand, which has been interpreted as a solvent molecule. It is contained in the N-terminal domain, whilst the function of the C-terminal domain is possibly structural. The catalytic mechanism very likely has been conserved through all species, since the positions of all residues considered relevant for the reaction are present from bacteria to humans.


Subject(s)
3-Hydroxyanthranilate 3,4-Dioxygenase/chemistry , Protein Structure, Tertiary , 3-Hydroxyanthranilate 3,4-Dioxygenase/genetics , 3-Hydroxyanthranilate 3,4-Dioxygenase/metabolism , 3-Hydroxyanthranilic Acid/chemistry , 3-Hydroxyanthranilic Acid/metabolism , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Cattle , Crystallization , Crystallography, X-Ray , Glutamic Acid/chemistry , Histidine/chemistry , Iron/chemistry , Kidney/enzymology , Models, Chemical , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Folding , Protein Structure, Secondary , Quinolinic Acid/chemistry , Quinolinic Acid/metabolism , Sequence Homology, Amino Acid
9.
J Cell Biochem ; 103(1): 42-53, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17476692

ABSTRACT

3-Hydroxyanthranilic acid (3HAA) is one of the tryptophan metabolites along the kynurenine pathway and induces apoptosis in T cells. We investigated the mechanism of 3HAA-induced apoptosis in mouse thymocytes. The optimal concentration of 3HAA for apoptosis induction was 300-500 microM. The induction of apoptosis by a suboptimal concentration (100 microM) of 3HAA was enhanced by superoxide dismutase (SOD) as well as MnCl2 and further promoted in the presence of catalase. The 3HAA-mediated generation of intracellular reactive oxygen species (ROS) was enhanced by SOD or MnCl2 and inhibited by catalase. Corresponding to apoptosis induction, the generation of cinnabarinic acid (CA) through the oxidation of 3HAA was enhanced by SOD or MnCl2 in the presence of catalase. The synthesized CA possessed more than 10 times higher apoptosis-inducing activity than 3HAA. The intracellular ROS generation was induced by CA within 15 min and decreased to the control levels within 4 h, whereas the 3HAA-induced ROS generation increased gradually up to 4 h. Corresponding to ROS generation, the mitochondrial membrane potential was downregulated within 15 min and retained by the CA treatment. Apoptosis induction by 3HAA or CA was dependent on caspases, and caspase-3 was much more strongly activated by CA than 3HAA. In conclusion, the CA generated from 3HAA possesses a strong apoptosis-inducing activity in thymocytes through ROS generation, the loss of mitochondrial membrane potential, and caspase activation.


Subject(s)
3-Hydroxyanthranilic Acid/chemistry , Apoptosis/drug effects , Caspase 3/metabolism , Oxazines/pharmacology , Reactive Oxygen Species/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , 3-Hydroxyanthranilic Acid/pharmacology , Animals , Catalase/metabolism , Cells, Cultured , Chlorides/pharmacology , Down-Regulation/drug effects , Manganese Compounds/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Molecular Structure , Oxazines/chemical synthesis , Oxazines/chemistry , Oxidation-Reduction/drug effects , Superoxide Dismutase/metabolism , Thymus Gland/drug effects , Time Factors
10.
Clin Exp Pharmacol Physiol ; 33(11): 1078-87, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17042918

ABSTRACT

1. Metabolism of tryptophan along the oxidative pathway via kynurenine results in the production of quinolinic acid and kynurenic acid, which can act on glutamate receptors in peripheral tissues. We have now measured the concentrations of kynurenine pathway metabolites in the plasma of patients with osteoporosis before treatment with drugs, throughout and after 2 years of treatment with the drugs raloxifene or etidronate. Oxidative stress was assessed by measuring levels of the lipid peroxidation products malondialdehyde and 4-hydroxynonenal. Kynurenines were analysed by HPLC. Bone density was measured using dual-energy X-ray absorptiometry scans. 2. Patients with osteoporosis showed significantly lower baseline levels of 3-hydroxyanthranilic acid compared with healthy controls, but significantly higher levels of anthranilic acid and lipid peroxidation products. After 2 years treatment with etidronate and calcium, we observed significant therapeutic responses quantified by bone densitometric scanning. Significant improvements were not seen in patients treated with raloxifene. 3. In parallel, the levels of 3-hydroxyanthranilic acid, anthranilic acid and lipid peroxidation products were restored to control values by both drug treatments studied and tryptophan levels were increased significantly compared with baseline values. 4. The results suggest that tryptophan metabolism is altered in osteoporosis in a manner that could contribute to the oxidative stress and, thus, to progress of the disease. The oxidative metabolism of tryptophan (the kynurenine pathway) could represent a novel target for the development of new drugs for the treatment of osteoporosis. In addition, we noted that etidronate is a more effective drug than raloxifene, but that the simultaneous use of non-steroidal anti-inflammatory drugs may reduce the efficacy of etidronate.


Subject(s)
Indomethacin/therapeutic use , Kynurenine/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Sodium Salicylate/therapeutic use , 3-Hydroxyanthranilic Acid/chemistry , 3-Hydroxyanthranilic Acid/metabolism , Aged , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Female , Humans , Kynurenic Acid/chemistry , Kynurenic Acid/metabolism , Kynurenine/blood , Kynurenine/chemistry , Lipid Peroxidation , Male , Middle Aged , Molecular Structure , Neopterin/blood , Tryptophan/chemistry , Tryptophan/metabolism , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/metabolism
11.
Biochemistry ; 44(21): 7623-31, 2005 May 31.
Article in English | MEDLINE | ID: mdl-15909977

ABSTRACT

3-Hydroxyanthranilate-3,4-dioxygenase (HAD) is a non-heme Fe(II) dependent enzyme that catalyzes the oxidative ring-opening of 3-hydroxyanthranilate to 2-amino-3-carboxymuconic semialdehyde. The enzymatic product subsequently cyclizes to quinolinate, an intermediate in the biosynthesis of nicotinamide adenine dinucleotide. Quinolinate has also been implicated in important neurological disorders. Here, we describe the mechanism by which 4-chloro-3-hydroxyanthranilate inhibits the HAD catalyzed reaction. Using overexpressed and purified bacterial HAD, we demonstrate that 4-chloro-3-hydroxyanthranilate functions as a mechanism-based inactivating agent. The inactivation results in the consumption of 2 +/- 0.8 equiv of oxygen and the production of superoxide. EPR analysis of the inactivation reaction demonstrated that the inhibitor stimulated the oxidation of the active site Fe(II) to the catalytically inactive Fe(III) oxidation state. The inactivated enzyme can be reactivated by treatment with DTT and Fe(II). High resolution ESI-FTMS analysis of the inactivated enzyme demonstrated that the inhibitor did not form an adduct with the enzyme and that four conserved cysteines were oxidized to two disulfides (Cys125-Cys128 and Cys162-Cys165) during the inactivation reaction. These results are consistent with a mechanism in which the enzyme, complexed to the inhibitor and O2, generates superoxide which subsequently dissociates, leaving the inhibitor and the oxidized iron center at the active site.


Subject(s)
3-Hydroxyanthranilic Acid/analogs & derivatives , 3-Hydroxyanthranilic Acid/chemistry , Dioxygenases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Ralstonia/enzymology , 3-Hydroxyanthranilate 3,4-Dioxygenase , Amino Acid Sequence , Dioxygenases/biosynthesis , Dioxygenases/genetics , Dioxygenases/isolation & purification , Disulfides/chemistry , Dithionitrobenzoic Acid , Enzyme Activation/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Mass Spectrometry , Molecular Sequence Data , Oxygen Consumption/genetics , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity/genetics , Superoxides/metabolism
12.
Biochemistry ; 44(21): 7632-43, 2005 May 31.
Article in English | MEDLINE | ID: mdl-15909978

ABSTRACT

3-Hydroxyanthranilate-3,4-dioxygenase (HAD) catalyzes the oxidative ring opening of 3-hydroxyanthranilate in the final enzymatic step of the biosynthetic pathway from tryptophan to quinolinate, the universal de novo precursor to the pyridine ring of nicotinamide adenine dinucleotide. The enzyme requires Fe2+ as a cofactor and is inactivated by 4-chloro-3-hydroxyanthranilate. HAD from Ralstonia metallidurans was crystallized, and the structure was determined at 1.9 A resolution. The structures of HAD complexed with the inhibitor 4-chloro-3-hydroxyanthranilic acid and either molecular oxygen or nitric oxide were determined at 2.0 A resolution, and the structure of HAD complexed with 3-hydroxyanthranilate was determined at 3.2 A resolution. HAD is a homodimer with a subunit topology that is characteristic of the cupin barrel fold. Each monomer contains two iron binding sites. The catalytic iron is buried deep inside the beta-barrel with His51, Glu57, and His95 serving as ligands. The other iron site forms an FeS4 center close to the solvent surface in which the sulfur atoms are provided by Cys125, Cys128, Cys162, and Cys165. The two iron sites are separated by 24 A. On the basis of the crystal structures of HAD, mutagenesis studies were carried out in order to elucidate the enzyme mechanism. In addition, a new mechanism for the enzyme inactivation by 4-chloro-3-hydroxyanthranilate is proposed.


Subject(s)
3-Hydroxyanthranilic Acid/analogs & derivatives , Dioxygenases/chemistry , Dioxygenases/metabolism , NAD/biosynthesis , Ralstonia/enzymology , 3-Hydroxyanthranilate 3,4-Dioxygenase , 3-Hydroxyanthranilic Acid/chemistry , Binding Sites , Catalysis , Crystallization , Crystallography, X-Ray , Dioxygenases/genetics , Enzyme Inhibitors/chemistry , Ferric Compounds/chemistry , Ligands , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Ralstonia/genetics , Substrate Specificity/genetics
13.
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
14.
Biotechnol Bioeng ; 82(4): 395-8, 2003 May 20.
Article in English | MEDLINE | ID: mdl-12632395

ABSTRACT

Spectrophotometric determination of laccase activity with ABTS acting as chromogen yields exceedingly low values whenever conducted in a water-organic mixed solvent. Nevertheless, there is firm evidence that laccase is able to oxidize substrates such as phenols and amines quantitatively in these mixed solvents. We show that the apparently small rate of ABTS oxidation by laccase in a mixed solvent, such as buffered water-dioxane 1:1, is not amenable to the denaturation of laccase but rather to the decreased stability of ABTS(.+). We propose HAA as a more reliable chromogen for the determination of laccase activity in mixed solvents.


Subject(s)
3-Hydroxyanthranilic Acid/chemistry , Chromogenic Compounds/chemistry , Oxidoreductases/chemistry , Solvents/chemistry , Spectrophotometry, Ultraviolet/methods , Sulfonic Acids/chemistry , Benzothiazoles , Enzyme Activation , Laccase , Solutions/chemistry , Substrate Specificity , Water/chemistry
15.
J Chromatogr B Biomed Sci Appl ; 736(1-2): 237-45, 1999 Dec 24.
Article in English | MEDLINE | ID: mdl-10677004

ABSTRACT

3-Hydroxyanthranilic acid (3-HAA)-derived oxidation products were analyzed using high-performance liquid chromatography with an electrochemical reactor and diode array detection and high-performance liquid chromatography with an electrochemical reactor and UV detection coupled with mass spectrometry. In addition to 3-HAA dimers such as cinnabarinic acid (CA), 6-amino-3-[(2-carboxy-6-hydroxyphenyl)amino]-2,5-dioxo-1,3-cyclohexadien e-1-carboxylic acid and 4,7-diamino-8-hydroxy-6H-dibenzo[a,d]pyran-6-one-3-carboxylic acid, a 3-HAA trimer and a 3-HAA tetramer were also detected and identified based on their electrospray ionization mass spectra and their UV-visible spectra. These five oxidation products were also detected on the elution profiles of high-performance liquid chromatography-diode array detection analyses for the reaction mixtures of the auto-oxidation of 3-HAA, of 3-HAA with potassium ferricyanide, of 3-HAA with horseradish peroxidase and hydrogen peroxide, and of 3-HAA with superoxide dismutase (SOD). 4,7-Diamino-8-hydroxy-6H-dibenzo[a,d]pyran-6-one-3-carboxylic acid was predominant in the auto-oxidation, in the reaction of 3-HAA with horseradish peroxidase and hydrogen peroxide, and in the electrochemical oxidation of 3-HAA at an applied potential of 0.0 V. On the other hand, CA, the 3-HAA trimer and the 3-HAA tetramer were predominant in the reaction of 3-HAA with K3[Fe(CN)6] and in the electrochemical oxidation of 3-HAA at an applied potential of 1.0 V.


Subject(s)
3-Hydroxyanthranilic Acid/chemistry , Chromatography, High Pressure Liquid/methods , Electrochemistry , Oxidation-Reduction , Dimerization , Ferricyanides/chemistry , Horseradish Peroxidase/metabolism , Hydrogen Peroxide/chemistry , Kinetics , Mass Spectrometry , Oxazines/chemistry , Spectrophotometry, Ultraviolet , Superoxide Dismutase/metabolism
16.
J Bacteriol ; 179(17): 5340-6, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9286986

ABSTRACT

Melanin is a fungal extracellular redox buffer which, in principle, can neutralize antimicrobial oxidants generated by immunologic effector cells, but its source of reducing equivalents is not known. We wondered whether Fe(II) generated by the external ferric reductase of fungi might have the physiologic function of reducing fungal melanin and thereby promoting pathogenesis. We observed that exposure of a melanin film electrode to reductants decreased the open-circuit potential (OCP) and reduced the area of a cyclic voltammetric reduction wave whereas exposure to oxidants produced the opposite effects. Exposure to 10, 100, 1,000 or 10,000 microM Fe(II) decreased the OCP of melanin by 0.015, 0.038, 0.100, and 0.120 V, respectively, relative to a silver-silver chloride standard, and decreased the area of the cyclic voltammetric reduction wave by 27, 35, 50, and 83%, respectively. Moreover, exposure to Fe(II) increased the buffering capacity by 44%, while exposure to millimolar dithionite did not increase the buffering capacity. The ratio of the amount of bound iron to the amount of the incremental increase in the following oxidation wave was approximately 1.0, suggesting that bound iron participates in buffering. Light absorption by melanin suspensions was decreased 14% by treatment with Fe(II), consistent with reduction of melanin. Light absorption by suspensions of melanized Cryptococcus neoformans was decreased 1.3% by treatment with Fe(II) (P < 0.05). Cultures of C. neoformans generated between 2 and 160 microM Fe(II) in culture supernatant, depending upon the strain and the conditions [the higher values were achieved by a constitutive ferric reductase mutant in high concentrations of Fe(III)]. We infer that Fe(II) can reduce melanin under physiologic conditions; moreover, it binds to melanin and cooperatively increases redox buffering. The data support a model for physiologic redox cycling of fungal melanin, whereby electrons exported by the yeast to form extracellular Fe(II) maintain the reducing capacity of the extracellular redox buffer.


Subject(s)
Cryptococcus neoformans/chemistry , Ferrous Compounds/chemistry , Melanins/chemistry , 3-Hydroxyanthranilic Acid/chemistry , Dithionite , Electrochemistry , Ferric Compounds/chemistry , Ketoglutaric Acids/chemistry , Oxidants/pharmacology , Oxidation-Reduction , Reducing Agents/pharmacology
17.
Arch Biochem Biophys ; 290(2): 451-7, 1991 Nov 01.
Article in English | MEDLINE | ID: mdl-1929412

ABSTRACT

The mechanism of reaction of proteins with 3-hydroxyanthranilic acid (3OHA) under oxidizing conditions has been examined. A range of proteins were found to tan when exposed to oxidized 3OHA. One exception was lysozyme which tanned only after being denatured by reduction and carboxymethylation. Chemical modification experiments using bovine serum albumin (BSA) suggested that lysine was the primary site of reaction in 3OHA-mediated protein tanning. This reactivity of 3OHA toward lysine was confirmed by autoxidizing 3OHA in the presence of amino acid homopolymers. The rate of modification of both BSA and polylysine was pH dependent. At neutral pH, a component of the coloration of the protein was found to be due to the formation of a lysyl-p-quinone adduct. Other products appear to arise through addition to the 3OHA quinone imine. Poly-(Glu,Lys) was tanned by 3OHA at a greatly reduced rate, suggesting that electrostatic interactions may influence the reaction with lysine residues and may provide an explanation for the lack of tanning of lysozyme. Despite the reaction between 3OHA and lysine, amino acid analysis revealed little quantitative change in the lysine content of proteins even after exposure to 3OHA for a period of 24 h. These results support the proposal that reaction with lysine residues is the major route of protein tanning by 3-hydroxyanthranilic acid.


Subject(s)
3-Hydroxyanthranilic Acid/pharmacology , Lysine/physiology , Serum Albumin, Bovine/drug effects , 3-Hydroxyanthranilic Acid/chemistry , Chromatography, High Pressure Liquid , Hydrogen-Ion Concentration , Lysine/chemistry , Protein Denaturation , Serum Albumin, Bovine/chemistry , Spectrophotometry, Ultraviolet , Structure-Activity Relationship
18.
J Chromatogr ; 566(2): 369-75, 1991 May 31.
Article in English | MEDLINE | ID: mdl-1939450

ABSTRACT

Human lymphocyte kynureninase activity was assessed in homogenized cells by determination of 3-hydroxyanthranilic acid production as a function of time after addition of the substrate, 3-hydroxykynurenine. The product, 3-hydroxyanthranilic acid, was determined by isocratic high-performance liquid chromatography and fluorescence detection. Mean (+/- S.D.) lymphocyte kynureninase activity in a group (n = 12) of vitamin B6-deficient men was 5.04 +/- 0.81 pmol 3-hydroxyanthranilic acid formed per mg protein per min, which was significantly (p = 0.005) lower than the 6.69 +/- 1.70 pmol 3-hydroxyanthranilic acid formed per mg protein per min in men with a normal vitamin B6 status. This indicates that lymphocyte kynureninase activity is depressed during a vitamin B6 deficiency.


Subject(s)
Chromatography, High Pressure Liquid , Hydrolases/blood , Lymphocytes/enzymology , 3-Hydroxyanthranilic Acid/chemistry , Humans , Kynurenine/analogs & derivatives , Kynurenine/chemistry , Male , Nutritional Status , Pyridoxine/blood , Substrate Specificity , Vitamin B 6 Deficiency/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...