Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 142(7): 3506-3512, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31986016

ABSTRACT

A highly efficient di-C-glycosyltransferase GgCGT was discovered from the medicinal plant Glycyrrhiza glabra. GgCGT catalyzes a two-step di-C-glycosylation of flopropione-containing substrates with conversion rates of >98%. To elucidate the catalytic mechanisms of GgCGT, we solved its crystal structures in complex with UDP-Glc, UDP-Gal, UDP/phloretin, and UDP/nothofagin, respectively. Structural analysis revealed that the sugar donor selectivity was controlled by the hydrogen-bond interactions of sugar hydroxyl groups with D390 and other key residues. The di-C-glycosylation capability of GgCGT was attributed to a spacious substrate-binding tunnel, and the G389K mutation could switch di- to mono-C-glycosylation. GgCGT is the first di-C-glycosyltransferase with a crystal structure, and the first C-glycosyltransferase with a complex structure containing a sugar acceptor. This work could benefit the development of efficient biocatalysts to synthesize C-glycosides with medicinal potential.


Subject(s)
Glycosyltransferases/chemistry , Glycosyltransferases/metabolism , Glycyrrhiza/enzymology , Cloning, Molecular , Crystallography, X-Ray , Glycosylation , Glycosyltransferases/genetics , Glycyrrhiza/genetics , Ligands , Models, Molecular , Phloretin/chemistry , Phloretin/metabolism , Substrate Specificity , Transcriptome , Uridine Diphosphate Galactose/chemistry , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate Glucuronic Acid/chemistry , Uridine Diphosphate Glucuronic Acid/metabolism , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism , Uridine Diphosphate Xylose/chemistry , Uridine Diphosphate Xylose/metabolism
2.
Plant Cell Rep ; 35(11): 2403-2421, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27591771

ABSTRACT

KEY MESSAGE: The present study first identified the involvement of OcUAXS2 and OcUXS1-3 in anticancer polysaccharides biosynthesis in O. caudatum. UDP-xylose synthase (UXS) and UDP-D-apiose/UDP-D-xylose synthase (UAXS), both capable of converting UDP-D-glucuronic acid to UDP-D-xylose, are believed to transfer xylosyl residue to anticancer polysaccharides biosynthesis in Ornithogalum caudatum Ait. However, the cDNA isolation and functional characterization of genes encoding the two enzymes from O. caudatum has never been documented. Previously, the transcriptome sequencing of O. caudatum was performed in our laboratory. In this study, a total of six and two unigenes encoding UXS and UAXS were first retrieved based on RNA-Seq data. The eight putative genes were then successfully isolated from transcriptome of O. caudatum by reverse transcription polymerase chain reaction (RT-PCR). Phylogenetic analysis revealed the six putative UXS isoforms can be classified into three types, one soluble and two distinct putative membrane-bound. Moreover, the two UAXS isoenzymes were predicted to be soluble forms. Subsequently, these candidate cDNAs were characterized to be bona fide genes by functional expression in Escherichia coli individually. Although UXS and UAXS catalyzed the same reaction, their biochemical properties varied significantly. It is worth noting that a ratio switch of UDP-D-xylose/UDP-D-apiose for UAXS was established, which is assumed to be helpful for its biotechnological application. Furthermore, a series of mutants were generated to test the function of NAD+ binding motif GxxGxxG. Most importantly, the present study determined the involvement of OcUAXS2 and OcUXS1-3 in xylose-containing polysaccharides biosynthesis in O. caudatum. These data provide a comprehensive knowledge for UXS and UAXS families in plants.


Subject(s)
Carboxy-Lyases/genetics , Genes, Plant , Multigene Family , Ornithogalum/enzymology , Ornithogalum/genetics , Transcriptome/genetics , Uridine Diphosphate Sugars/metabolism , Uridine Diphosphate Xylose/metabolism , Amino Acid Motifs , Amino Acid Sequence , Ammonium Compounds/pharmacology , Biocatalysis/drug effects , Buffers , Calcium/pharmacology , Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , Chromatography, High Pressure Liquid , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Hydrogen-Ion Concentration , Kinetics , Organ Specificity/drug effects , Organ Specificity/genetics , Ornithogalum/drug effects , Proton Magnetic Resonance Spectroscopy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA , Temperature , Transcriptome/drug effects , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Xylose/chemistry
3.
Biochemistry ; 54(3): 807-19, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25521717

ABSTRACT

The man o' war (mow) phenotype in zebrafish is characterized by severe craniofacial defects due to a missense mutation in UDP-α-d-xylose synthase (UXS), an essential enzyme in proteoglycan biosynthesis. The mow mutation is located in the UXS dimer interface ∼16 Å away from the active site, suggesting an indirect effect on the enzyme mechanism. We have examined the structural and catalytic consequences of the mow mutation (R236H) in the soluble fragment of human UXS (hUXS), which shares 93% sequence identity with the zebrafish enzyme. In solution, hUXS dimers undergo a concentration-dependent association to form a tetramer. Sedimentation velocity studies show that the R236H substitution induces the formation of a new hexameric species. Using two new crystal structures of the hexamer, we show that R236H and R236A substitutions cause a local unfolding of the active site that allows for a rotation of the dimer interface necessary to form the hexamer. The disordered active sites in the R236H and R236A mutant constructs displace Y231, the essential acid/base catalyst in the UXS reaction mechanism. The loss of Y231 favors an abortive catalytic cycle in which the reaction intermediate, UDP-α-d-4-keto-xylose, is not reduced to the final product, UDP-α-d-xylose. Surprisingly, the mow-induced hexamer is almost identical to the hexamers formed by the deeply divergent UXS homologues from Staphylococcus aureus and Helicobacter pylori (21% and 16% sequence identity, respectively). The persistence of a latent hexamer-building interface in the human enzyme suggests that the ancestral UXS may have been a hexamer.


Subject(s)
Biocatalysis , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Mutation/genetics , Protein Multimerization , Animals , Carboxy-Lyases/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Mutant Proteins/chemistry , Nucleotides/metabolism , Phenotype , Protein Folding , Protein Structure, Quaternary , Proteoglycans/biosynthesis , Solutions , Uridine Diphosphate Xylose/chemistry , Uridine Diphosphate Xylose/metabolism , Zebrafish
4.
J Am Chem Soc ; 134(34): 13946-9, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-22830643

ABSTRACT

UDP-D-apiose/UDP-D-xylose synthase (AXS) catalyzes the conversion of UDP-D-glucuronic acid to UDP-D-apiose and UDP-D-xylose. An acetyl-protected phosphonate analogue of UDP-D-apiose was synthesized and used in an in situ HPLC assay to demonstrate for the first time the ability of AXS to interconvert the two reaction products. Density functional theory calculations provided insight into the energetics of this process and the apparent inability of AXS to catalyze the conversion of UDP-D-xylose to UDP-D-apiose. The data suggest that this observation is unlikely to be due to an unfavorable equilibrium but rather results from substrate inhibition by the most stable chair conformation of UDP-D-xylose. The detection of xylose cyclic phosphonate as the turnover product reveals significant new details about the AXS-catalyzed reaction and supports the proposed retroaldol-aldol mechanism of catalysis.


Subject(s)
Carboxy-Lyases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Organophosphonates/metabolism , Uridine Diphosphate Sugars/metabolism , Uridine Diphosphate Xylose/metabolism , Escherichia coli/metabolism , Kinetics , Models, Molecular , Organophosphonates/chemistry , Quantum Theory , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Xylose/chemistry
5.
J Biol Chem ; 285(32): 24825-33, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20529859

ABSTRACT

We have identified an operon and characterized the functions of two genes from the severe food-poisoning bacterium, Bacillus cereus subsp. cytotoxis NVH 391-98, that are involved in the synthesis of a unique UDP-sugar, UDP-2-acetamido-2-deoxyxylose (UDP-N-acetyl-xylosamine, UDP-XylNAc). UGlcNAcDH encodes a UDP-N-acetyl-glucosamine 6-dehydrogenase, converting UDP-N-acetylglucosamine (UDP-GlcNAc) to UDP-N-acetyl-glucosaminuronic acid (UDP-GlcNAcA). The second gene in the operon, UXNAcS, encodes a distinct decarboxylase not previously described in the literature, which catalyzes the formation of UDP-XylNAc from UDP-GlcNAcA in the presence of exogenous NAD(+). UXNAcS is specific and cannot utilize UDP-glucuronic acid and UDP-galacturonic acid as substrates. UXNAcS is active as a dimer with catalytic efficiency of 7 mM(-1) s(-1). The activity of UXNAcS is completely abolished by NADH but unaffected by UDP-xylose. A real-time NMR-based assay showed unambiguously the dual enzymatic conversions of UDP-GlcNAc to UDP-GlcNAcA and subsequently to UDP-XylNAc. From the analyses of all publicly available sequenced genomes, it appears that UXNAcS is restricted to pathogenic Bacillus species, including Bacillus anthracis and Bacillus thuringiensis. The identification of UXNAcS provides insight into the formation of UDP-XylNAc. Understanding the metabolic pathways involved in the utilization of this amino-sugar may allow the development of drugs to combat and eradicate the disease.


Subject(s)
Bacillus cereus/metabolism , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Uridine Diphosphate Sugars/biosynthesis , Uridine Diphosphate Xylose/chemistry , Carbohydrate Sequence , Cloning, Molecular , Dimerization , Extracellular Matrix/metabolism , Glycosaminoglycans/chemistry , Humans , Magnetic Resonance Spectroscopy , Models, Biological , Models, Chemical , Molecular Sequence Data , Recombinant Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Uridine Diphosphate Sugars/chemistry
6.
J Biol Chem ; 285(12): 9030-40, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20118241

ABSTRACT

The UDP-sugar interconverting enzymes involved in UDP-GlcA metabolism are well described in eukaryotes but less is known in prokaryotes. Here we identify and characterize a gene (RsU4kpxs) from Ralstonia solanacearum str. GMI1000, which encodes a dual function enzyme not previously described. One activity is to decarboxylate UDP-glucuronic acid to UDP-beta-l-threo-pentopyranosyl-4''-ulose in the presence of NAD(+). The second activity converts UDP-beta-l-threo-pentopyranosyl-4''-ulose and NADH to UDP-xylose and NAD(+), albeit at a lower rate. Our data also suggest that following decarboxylation, there is stereospecific protonation at the C5 pro-R position. The identification of the R. solanacearum enzyme enables us to propose that the ancestral enzyme of UDP-xylose synthase and UDP-apiose/UDP-xylose synthase was diverged to two distinct enzymatic activities in early bacteria. This separation gave rise to the current UDP-xylose synthase in animal, fungus, and plant as well as to the plant Uaxs and bacterial ArnA and U4kpxs homologs.


Subject(s)
Alcohol Oxidoreductases/chemistry , Carboxy-Lyases/chemistry , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Multienzyme Complexes/chemistry , Plants/microbiology , Ralstonia solanacearum/metabolism , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Xylose/chemistry , Alcohol Oxidoreductases/physiology , Amino Acid Sequence , Carboxy-Lyases/physiology , Cloning, Molecular , Escherichia coli/metabolism , Magnetic Resonance Spectroscopy , Models, Biological , Models, Chemical , Molecular Sequence Data , Multienzyme Complexes/physiology , Phylogeny , Sequence Homology, Amino Acid
7.
J Biol Chem ; 282(8): 5389-403, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17190829

ABSTRACT

UDP-L-rhamnose is required for the biosynthesis of cell wall rhamnogalacturonan-I, rhamnogalacturonan-II, and natural compounds in plants. It has been suggested that the RHM2/MUM4 gene is involved in conversion of UDP-D-glucose to UDP-L-rhamnose on the basis of its effect on rhamnogalacturonan-I-directed development in Arabidopsis thaliana. RHM2/MUM4-related genes, RHM1 and RHM3, can be found in the A. thaliana genome. Here we present direct evidence that all three RHM proteins have UDP-D-glucose 4,6-dehydratase, UDP-4-keto-6-deoxy-D-glucose 3,5-epimerase, and UDP-4-keto-L-rhamnose 4-keto-reductase activities in the cytoplasm when expressed in the yeast Saccharomyces cerevisiae. Functional domain analysis revealed that the N-terminal region of RHM2 (RHM2-N; amino acids 1-370) has the first activity and the C-terminal region of RHM2 (RHM2-C; amino acids 371-667) has the two following activities. This suggests that RHM2 converts UDP-d-glucose to UDP-L-rhamnose via an UDP-4-keto-6-deoxy-D-glucose intermediate. Site-directed mutagenesis of RHM2 revealed that mucilage defects in MUM4-1 and MUM4-2 mutant seeds of A. thaliana are caused by abolishment of RHM2 enzymatic activity in the mutant strains and furthermore, that the GXXGXX(G/A) and YXXXK motifs are important for enzymatic activity. Moreover, a kinetic analysis of purified His(6)-tagged RHM2-N protein revealed 5.9-fold higher affinity of RHM2 for UDP-D-glucose than for dTDP-D-glucose, the preferred substrate for dTDP-D-glucose 4,6-dehydratase from bacteria. RHM2-N activity is strongly inhibited by UDP-L-rhamnose, UDP-D-xylose, and UDP but not by other sugar nucleotides, suggesting that RHM2 maintains cytoplasmic levels of UDP-D-glucose and UDP-L-rhamnose via feedback inhibition by UDP-L-rhamnose and UDP-D-xylose.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Cell Wall/metabolism , Genome, Plant/physiology , Multienzyme Complexes/metabolism , Rhamnose/metabolism , Uridine Diphosphate Glucose/metabolism , Amino Acid Motifs/genetics , Arabidopsis/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutagenesis, Site-Directed , Pectins/biosynthesis , Pectins/chemistry , Pectins/genetics , Rhamnose/chemistry , Rhamnose/genetics , Saccharomyces cerevisiae/genetics , Seeds/chemistry , Seeds/enzymology , Seeds/genetics , Uridine Diphosphate Glucose/chemistry , Uridine Diphosphate Glucose/genetics , Uridine Diphosphate Xylose/chemistry , Uridine Diphosphate Xylose/genetics , Uridine Diphosphate Xylose/metabolism
8.
J Biol Chem ; 280(52): 42774-84, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16258169

ABSTRACT

Expression of decorin using the vaccinia virus/T7 expression system resulted in secretion of two distinct glycoforms: a proteoglycan substituted with a single chondroitin sulfate chain and N-linked oligosaccharides and a core protein glycoform substituted with N-linked glycans but without a glycosaminoglycan chain. In this report, we have addressed two distinct questions. What is the rate-limiting step in glycosaminoglycan synthesis? Is glycosylation with either N-linked oligosaccharides or glycosaminoglycan required for secretion of decorin? N-terminal sequencing of the core protein glycoform, the addition of benzyl-beta-d-xyloside, and a UDP-xylose: core protein beta-d-xylosyltransferase activity assay show that xylosylation is a rate-limiting step in chondroitin sulfate biosynthesis. Decorin can be efficiently secreted with N-linked oligosaccharides alone or with a single chondroitin sulfate chain alone; however, there is severely impaired secretion of core protein devoid of any glycosylation. A decorin core protein mutant devoid of N-linked oligosaccharide attachment sites will not be secreted by Chinese hamster ovary cells deficient in xylosyltransferase or by parental Chinese hamster ovary wild type cells if the xylosyltransferase recognition sequence is disrupted. This finding suggests that quality control mechanisms sensitive to an absence of N-linked oligosaccharides can be abrogated by interaction of the core protein with the glycosaminoglycan synthetic machinery. We propose a model of regulation of decorin secretion that has several components, including appropriate substitution with N-linked oligosaccharides and factors involved in glycosaminoglycan synthesis.


Subject(s)
Proteoglycans/chemistry , Animals , Blotting, Northern , CHO Cells , Cell Line , Chondroitin Sulfates/chemistry , Cricetinae , DNA, Complementary/metabolism , Decorin , Escherichia coli/metabolism , Extracellular Matrix Proteins , Glycosaminoglycans/chemistry , Glycosides/chemistry , Glycosylation , HeLa Cells , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Genetic , Mutagenesis, Site-Directed , Mutation , Oligosaccharides/chemistry , Protein Isoforms , Protein Structure, Tertiary , Proteoglycans/genetics , Proteoglycans/metabolism , Recombinant Proteins/chemistry , Temperature , Time Factors , Tunicamycin/pharmacology , Uridine Diphosphate Xylose/chemistry , Vaccinia virus/metabolism , Xylose/chemistry
9.
J Biol Chem ; 279(49): 51669-76, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15383535

ABSTRACT

Cryptococcus neoformans is a pathogenic fungus surrounded by an elaborate polysaccharide capsule that is strictly required for its virulence in humans and other mammals. Nearly half of the sugar residues in the capsule are derived from UDP-glucuronic acid or its metabolites. To examine the role of these nucleotide sugars in C. neoformans, the gene encoding UDP-glucose dehydrogenase was disrupted. Mass spectrometry analysis of nucleotide sugar pools showed that the resulting mutant lacked both UDP-glucuronic acid and its downstream product, UDP-xylose, thus confirming the effect of the knockout and indicating that an alternate pathway for UDP-glucuronic acid production was not used. The mutant was dramatically affected by the lack of specific sugar donors, demonstrating altered cell integrity, temperature sensitivity, lack of growth in an animal model of cryptococcosis, and morphological defects. Additionally, the polysaccharide capsule could not be detected on the mutant cells, although the possibility remains that abbreviated forms of capsule components are made, possibly without proper surface display. The capsule defect is largely independent of the other observed changes, as cells that are acapsular because of mutations in other genes show lack of virulence but do not exhibit alterations in cell integrity, temperature sensitivity, or cellular morphology. All of the observed alterations were reversed by correction of the gene disruption.


Subject(s)
Cryptococcus neoformans/physiology , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/physiology , Antibodies, Monoclonal/chemistry , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Coloring Agents/pharmacology , DNA/chemistry , Genetic Complementation Test , Glucuronic Acid/chemistry , Immunoblotting , Mass Spectrometry , Microscopy, Electron , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microscopy, Interference , Models, Genetic , Mutation , Nucleotides/chemistry , Polysaccharides/chemistry , Protein Binding , Temperature , Uridine Diphosphate Xylose/chemistry
10.
Plant Physiol ; 130(4): 2188-98, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12481102

ABSTRACT

UDP-xylose (Xyl) is an important sugar donor for the synthesis of glycoproteins, polysaccharides, various metabolites, and oligosaccharides in animals, plants, fungi, and bacteria. UDP-Xyl also feedback inhibits upstream enzymes (UDP-glucose [Glc] dehydrogenase, UDP-Glc pyrophosphorylase, and UDP-GlcA decarboxylase) and is involved in its own synthesis and the synthesis of UDP-arabinose. In plants, biosynthesis of UDP-Xyl is catalyzed by different membrane-bound and soluble UDP-GlcA decarboxylase (UDP-GlcA-DC) isozymes, all of which convert UDP-GlcA to UDP-Xyl. Because synthesis of UDP-Xyl occurs both in the cytosol and in membranes, it is not known which source of UDP-Xyl the different Golgi-localized xylosyltransferases are utilizing. Here, we describe the identification of several distinct Arabidopsis genes (named AtUXS for UDP-Xyl synthase) that encode functional UDP-GlcA-DC isoforms. The Arabidopsis genome contains five UXS genes and their protein products can be subdivided into three isozyme classes (A-C), one soluble and two distinct putative membrane bound. AtUxs from each class, when expressed in Escherichia coli, generate active UDP-GlcA-DC that converts UDP-GlcA to UDP-Xyl. Members of this gene family have a large conserved C-terminal catalytic domain (approximately 300 amino acids long) and an N-terminal variable domain differing in sequence and size (30-120 amino acids long). Isoforms of class A and B appear to encode putative type II membrane proteins with their catalytic domains facing the lumen (like Golgi-glycosyltransferases) and their N-terminal variable domain facing the cytosol. Uxs class C is likely a cytosolic isoform. The characteristics of the plant Uxs support the hypothesis that unique UDP-GlcA-DCs with distinct subcellular localizations are required for specific xylosylation events.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Carboxy-Lyases/genetics , Uridine Diphosphate Xylose/biosynthesis , Amino Acid Sequence , Arabidopsis/enzymology , Arabidopsis Proteins/metabolism , Carboxy-Lyases/metabolism , Cloning, Molecular , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Plant , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Multigene Family/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Uridine Diphosphate Glucose/chemistry , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Xylose/chemistry
11.
Proc Natl Acad Sci U S A ; 98(21): 12003-8, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11593010

ABSTRACT

UDP-xylose is a sugar donor required for the synthesis of diverse and important glycan structures in animals, plants, fungi, and bacteria. Xylose-containing glycans are particularly abundant in plants and in the polysaccharide capsule that is the major virulence factor of the pathogenic fungus Cryptococcus neoformans. Biosynthesis of UDP-xylose is mediated by UDP-glucuronic acid decarboxylase, which converts UDP-glucuronic acid to UDP-xylose. Although this enzymatic activity was described over 40 years ago it has never been fully purified, and the gene encoding it has not been identified. We used homology to a bacterial gene, hypothesized to encode a related function, to identify a cryptococcal sequence as putatively encoding a UDP-glucuronic acid decarboxylase. A soluble 47-kDa protein derived from bacteria expressing the C. neoformans gene catalyzed conversion of UDP-glucuronic acid to UDP-xylose, as confirmed by NMR analysis. NADH, UDP, and UDP-xylose inhibit the activity. Close homologs of the cryptococcal gene, which we termed UXS1, appear in genome sequence data from organisms ranging from bacteria to humans.


Subject(s)
Carboxy-Lyases/physiology , Cryptococcus neoformans/enzymology , Uridine Diphosphate Xylose/biosynthesis , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Cations, Divalent , Cloning, Molecular , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , DNA, Fungal , Hydrogen-Ion Concentration , Molecular Sequence Data , Molecular Structure , NAD/metabolism , Nuclear Magnetic Resonance, Biomolecular , Open Reading Frames , Substrate Specificity , Temperature , Uridine Diphosphate Glucuronic Acid/metabolism , Uridine Diphosphate Xylose/chemistry
12.
Biochemistry ; 39(23): 7012-23, 2000 Jun 13.
Article in English | MEDLINE | ID: mdl-10841783

ABSTRACT

Bacterial UDP-glucose dehydrogenase (UDPGlcDH) is essential for formation of the antiphagocytic capsule that protects many virulent bacteria such as Streptococcus pyogenes andStreptococcus pneumoniae type 3 from the host's immune system. We have determined the X-ray structures of both native and Cys260Ser UDPGlcDH from S. pyogenes (74% similarity to S. pneumoniae) in ternary complexes with UDP-xylose/NAD(+) and UDP-glucuronic acid/NAD(H), respectively. The 402 residue homodimeric UDPGlcDH is composed of an N-terminal NAD(+) dinucleotide binding domain and a C-terminal UDP-sugar binding domain connected by a long (48 A) central alpha-helix. The first 290 residues of UDPGlcDH share structural homology with 6-phosphogluconate dehydrogenase, including conservation of an active site lysine and asparagine that are implicated in the enzyme mechanism. Also proposed to participate in the catalytic mechanism are a threonine and a glutamate that hydrogen bond to a conserved active site water molecule suitably positioned for general acid/base catalysis.


Subject(s)
Bacterial Proteins/chemistry , Streptococcus pyogenes/enzymology , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Dimerization , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , NAD/chemistry , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Uridine Diphosphate Glucuronic Acid/chemistry , Uridine Diphosphate Xylose/chemistry
13.
Anal Biochem ; 225(2): 296-304, 1995 Mar 01.
Article in English | MEDLINE | ID: mdl-7762795

ABSTRACT

Pectins are complex polysaccharides that contain 1,4-linked alpha-D-galactosyluronic acid residues found in the primary wall of all higher plant cells. The pectic polysaccharides play critical roles in cell wall structure and in plant growth and development. As a first step in studying pectin biosynthesis a method was developed to routinely generate and purify UDP-[U-14C]galacturonic acid (UDP-[14C]GalA), the nucleotide sugar substrate for homogalacturonan biosynthesis. UDP-[14C]GalA was enzymatically synthesized by 4-epimerization of commercially available UDP-[U-14C]glucuronic acid (UDP-[14C]GlcA) using a particulate preparation from radish roots. The resulting mixture of UDP-[14C]GalA and UDP-[14C]GlcA was separated by high-performance anion-exchange chromatography using a Dionex CarboPac PA1 anion-exchange column. The UDP-sugars were detected by their absorbance at 262 nm or by pulsed amperometric detection following postcolumn addition of NaOH. The yield of UDP-[14C]GalA obtained using this procedure was 16% of the starting UDP-[14C]GlcA. Establishment of a reliable method to synthesize and purify UDP-[14C]GalA will facilitate the identification and purification of the galacturonosyltransferase(s) involved in pectin biosynthesis.


Subject(s)
Pectins/biosynthesis , Uridine Diphosphate Sugars/chemical synthesis , Uridine Diphosphate Sugars/isolation & purification , Anions , Carbon Radioisotopes , Chromatography, High Pressure Liquid/methods , Chromatography, Ion Exchange/methods , Plant Extracts , Racemases and Epimerases/chemistry , Racemases and Epimerases/metabolism , Uridine Diphosphate Glucuronic Acid/chemistry , Uridine Diphosphate Glucuronic Acid/metabolism , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate Sugars/metabolism , Uridine Diphosphate Xylose/chemistry , Vegetables/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...