Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Biochem J ; 478(4): 685-701, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33599745

ABSTRACT

Rhamnose is an important 6-deoxy sugar present in many natural products, glycoproteins, and structural polysaccharides. Whilst predominantly found as the l-enantiomer, instances of d-rhamnose are also found in nature, particularly in the Pseudomonads bacteria. Interestingly, rhamnose is notably absent from humans and other animals, which poses unique opportunities for drug discovery targeted towards rhamnose utilizing enzymes from pathogenic bacteria. Whilst the biosynthesis of nucleotide-activated rhamnose (NDP-rhamnose) is well studied, the study of rhamnosyltransferases that synthesize rhamnose-containing glycoconjugates is the current focus amongst the scientific community. In this review, we describe where rhamnose has been found in nature, as well as what is known about TDP-ß-l-rhamnose, UDP-ß-l-rhamnose, and GDP-α-d-rhamnose biosynthesis. We then focus on examples of rhamnosyltransferases that have been characterized using both in vivo and in vitro approaches from plants and bacteria, highlighting enzymes where 3D structures have been obtained. The ongoing study of rhamnose and rhamnosyltransferases, in particular in pathogenic organisms, is important to inform future drug discovery projects and vaccine development.


Subject(s)
Glycoconjugates/biosynthesis , Hexosyltransferases/physiology , Rhamnose/biosynthesis , Uridine Diphosphate Sugars/biosynthesis , Arabidopsis Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Capsid/metabolism , Eukaryotic Cells/metabolism , Flavonoids/metabolism , Glycoconjugates/chemistry , Glycolipids/biosynthesis , Glycosylation , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/metabolism , Gram-Positive Bacteria/pathogenicity , Hexosyltransferases/chemistry , Hexosyltransferases/genetics , Models, Molecular , O Antigens/metabolism , Plant Proteins/metabolism , Polysaccharides, Bacterial/metabolism , Prokaryotic Cells/metabolism , Protein Conformation , Protein Processing, Post-Translational , Viral Proteins/metabolism , Virulence
2.
J Am Chem Soc ; 140(23): 7092-7100, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29771506

ABSTRACT

Metabolic chemical reporters of glycosylation in combination with bioorthogonal reactions have been known for two decades and have been used by many different research laboratories for the identification and visualization of glycoconjugates. More recently, however, they have begun to see utility for the investigation of cellular metabolism and the tolerance of biosynthetic enzymes and glycosyltransferases to different sugars. Here, we take this concept one step further by using the metabolic chemical reporter 6-azido-6-deoxy-glucose (6AzGlc). We show that treatment of mammalian cells with the per- O-acetylated version of 6AzGlc results in robust labeling of a variety of proteins. Notably, the pattern of this labeling was consistent with O-GlcNAc modifications, suggesting that the enzyme O-GlcNAc transferase is quite promiscuous for its donor sugar substrates. To confirm this possibility, we show that 6AzGlc-treatment results in the labeling of known O-GlcNAcylated proteins, that the UDP-6AzGlc donor sugar is indeed produced in living cells, and that recombinant OGT will accept UDP-6AzGlc as a substrate in vitro. Finally, we use proteomics to first identify several bona fide 6AzGlc-modifications in mammalian cells and then an endogenous O-glucose modification on host cell factor. These results support the conclusion that OGT can endogenously modify proteins with both N-acetyl-glucosamine and glucose, raising the possibility that intracellular O-glucose modification may be a widespread modification under certain conditions or in particular tissues.


Subject(s)
Azides/metabolism , Deoxyglucose/analogs & derivatives , Deoxyglucose/metabolism , N-Acetylglucosaminyltransferases/metabolism , Proteins/metabolism , Animals , Azides/chemical synthesis , Azides/chemistry , Cell Line, Tumor , Chlorocebus aethiops , Deoxyglucose/chemical synthesis , Glycosylation , Humans , Mice , Protein Processing, Post-Translational , Substrate Specificity , Uridine Diphosphate Sugars/biosynthesis , beta-N-Acetylhexosaminidases/metabolism
3.
J Biol Chem ; 291(41): 21434-21447, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27551039

ABSTRACT

Apiose is a branched monosaccharide that is present in the cell wall pectic polysaccharides rhamnogalacturonan II and apiogalacturonan and in numerous plant secondary metabolites. These apiose-containing glycans are synthesized using UDP-apiose as the donor. UDP-apiose (UDP-Api) together with UDP-xylose is formed from UDP-glucuronic acid (UDP-GlcA) by UDP-Api synthase (UAS). It was hypothesized that the ability to form Api distinguishes vascular plants from the avascular plants and green algae. UAS from several dicotyledonous plants has been characterized; however, it is not known if avascular plants or green algae produce this enzyme. Here we report the identification and functional characterization of UAS homologs from avascular plants (mosses, liverwort, and hornwort), from streptophyte green algae, and from a monocot (duckweed). The recombinant UAS homologs all form UDP-Api from UDP-glucuronic acid albeit in different amounts. Apiose was detected in aqueous methanolic extracts of these plants. Apiose was detected in duckweed cell walls but not in the walls of the avascular plants and algae. Overexpressing duckweed UAS in the moss Physcomitrella patens led to an increase in the amounts of aqueous methanol-acetonitrile-soluble apiose but did not result in discernible amounts of cell wall-associated apiose. Thus, bryophytes and algae likely lack the glycosyltransferase machinery required to synthesize apiose-containing cell wall glycans. Nevertheless, these plants may have the ability to form apiosylated secondary metabolites. Our data are the first to provide evidence that the ability to form apiose existed prior to the appearance of rhamnogalacturonan II and apiogalacturonan and provide new insights into the evolution of apiose-containing glycans.


Subject(s)
Bryopsida/metabolism , Carboxy-Lyases/metabolism , Chlorophyta/metabolism , Evolution, Molecular , Plant Proteins/metabolism , Uridine Diphosphate Sugars/biosynthesis , Bryopsida/genetics , Carboxy-Lyases/genetics , Cell Wall/genetics , Cell Wall/metabolism , Chlorophyta/genetics , Plant Proteins/genetics , Polysaccharides/biosynthesis , Polysaccharides/genetics , Uridine Diphosphate Sugars/genetics
4.
J Org Chem ; 82(4): 2243-2248, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28128958

ABSTRACT

Unnatural uridine diphosphate (UDP)-sugar donors, UDP-4-deoxy-4-fluoro-N-acetylglucosamine (4FGlcNAc) and UDP-4-deoxy-4-fluoro-N-acetylgalactosamine (4FGalNAc), were prepared using both chemical and chemoenzymatic syntheses relying on N-acetylglucosamine-1-phosphate uridylyltransferase (GlmU). The resulting unnatural UDP-sugar donors were then tested as substrates in glycosaminoglycan synthesis catalyzed by various synthases. UDP-4FGlcNAc was transferred onto an acceptor by Pastuerella multocida heparosan synthase 1 and subsequently served as a chain terminator.


Subject(s)
Glycosaminoglycans/biosynthesis , Nucleotidyltransferases/metabolism , Biocatalysis , Carbohydrate Conformation , Glycosaminoglycans/chemistry , Nucleotidyltransferases/chemistry , Uridine Diphosphate Sugars/biosynthesis , Uridine Diphosphate Sugars/chemistry
5.
Proc Natl Acad Sci U S A ; 111(50): 18019-24, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25472841

ABSTRACT

Human cytomegalovirus (HCMV) induces numerous changes to the host metabolic network that are critical for high-titer viral replication. We find that HCMV infection substantially induces de novo pyrimidine biosynthetic flux. This activation is important for HCMV replication because inhibition of pyrimidine biosynthetic enzymes substantially decreases the production of infectious virus, which can be rescued through medium supplementation with pyrimidine biosynthetic intermediates. Metabolomic analysis revealed that pyrimidine biosynthetic inhibition considerably reduces the levels of various UDP-sugar metabolites in HCMV-infected, but not mock-infected, cells. Further, UDP-sugar biosynthesis, which provides the sugar substrates required for glycosylation reactions, was found to be induced during HCMV infection. Pyrimidine biosynthetic inhibition also attenuated the glycosylation of the envelope glycoprotein B (gB). Both glycosylation of gB and viral growth were restored by medium supplementation with either UDP-sugar metabolites or pyrimidine precursors. These results indicate that HCMV drives de novo-synthesized pyrimidines to UDP-sugar biosynthesis to support virion protein glycosylation. The importance of this link between pyrimidine biosynthesis and UDP-sugars appears to be partially shared among diverse virus families, because UDP-sugar metabolites rescued the growth attenuation associated with pyrimidine biosynthetic inhibition during influenza A and vesicular stomatitis virus infection, but not murine hepatitis virus infection. In total, our results indicate that viruses can specifically modulate pyrimidine metabolic flux to provide the glycosyl subunits required for protein glycosylation and production of high titers of infectious progeny.


Subject(s)
Cytomegalovirus/physiology , Pyrimidines/biosynthesis , Uridine Diphosphate Sugars/biosynthesis , Viral Envelope Proteins/metabolism , Virus Replication/physiology , Analysis of Variance , Chromatography, Liquid , DNA Primers/genetics , Glycosylation , Humans , Metabolic Flux Analysis , RNA Interference , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry , Viral Envelope Proteins/biosynthesis
6.
J Biol Chem ; 289(51): 35620-32, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25368324

ABSTRACT

Surface glycan switching is often observed when micro-organisms transition between different biotic and abiotic niches, including biofilms, although the advantages of this switching to the organism are not well understood. Bacillus cereus grown in a biofilm-inducing medium has been shown to synthesize an unusual cell wall polysaccharide composed of the repeating subunit →6)Gal(α1-2)(2-R-hydroxyglutar-5-ylamido)Fuc2NAc4N(α1-6)GlcNAc(ß1→, where galactose is linked to the hydroxyglutarate moiety of FucNAc-4-amido-(2)-hydroxyglutarate. The molecular mechanism involved in attaching 2-hydroxyglutarate to 4-amino-FucNAc has not been determined. Here, we show two genes in B. cereus ATCC 14579 encoding enzymes involved in the synthesis of UDP-FucNAc-4-amido-(2)-oxoglutarate (UDP-Yelosamine), a modified UDP-sugar not previously reported to exist. Using mass spectrometry and real time NMR spectroscopy, we show that Bc5273 encodes a C4″-aminotransferase (herein referred to as Pat) that, in the presence of pyridoxal phosphate, transfers the primary amino group of l-Glu to C-4″ of UDP-4-keto-6-deoxy-d-GlcNAc to form UDP-4-amino-FucNAc and 2-oxoglutarate. Pat also converts 4-keto-xylose, 4-keto-glucose, and 4-keto-2-acetamido-altrose to their corresponding UDP-4-amino-sugars. Bc5272 encodes a carboxylate-amine ligase (herein referred as Pyl) that, in the presence of ATP and Mg(II), adds 2-oxoglutarate to the 4-amino moiety of UDP-4-amino-FucNAc to form UDP-Yelosamine and ADP. Pyl is also able to ligate 2-oxoglutarate to other 4-amino-sugar derivatives to form UDP-Yelose, UDP-Solosamine, and UDP-Aravonose. Characterizing the metabolic pathways involved in the formation of modified nucleotide sugars provides a basis for understanding some of the mechanisms used by bacteria to modify or alter their cell surface polysaccharides in response to changing growth and environmental challenges.


Subject(s)
Bacillus cereus/metabolism , Bacterial Proteins/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Transaminases/metabolism , Uridine Diphosphate Sugars/biosynthesis , Bacillus cereus/genetics , Bacterial Proteins/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbohydrate Sequence , Chromatography, High Pressure Liquid/methods , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Hydrogen-Ion Concentration , Kinetics , Mass Spectrometry/methods , Molecular Sequence Data , Proton Magnetic Resonance Spectroscopy , Recombinant Proteins/metabolism , Substrate Specificity , Transaminases/genetics
7.
J Biol Chem ; 288(45): 32248-32260, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24064219

ABSTRACT

UDP-N,N'-diacetylbacillosamine (UDP-diNAcBac) is a unique carbohydrate produced by a number of bacterial species and has been implicated in pathogenesis. The terminal step in the formation of this important bacterial sugar is catalyzed by an acetyl-CoA (AcCoA)-dependent acetyltransferase in both N- and O-linked protein glycosylation pathways. This bacterial acetyltransferase is a member of the left-handed ß-helix family and forms a homotrimer as the functional unit. Whereas previous endeavors have focused on the Campylobacter jejuni acetyltransferase (PglD) from the N-linked glycosylation pathway, structural characterization of the homologous enzymes in the O-linked glycosylation pathways is lacking. Herein, we present the apo-crystal structures of the acetyltransferase domain (ATD) from the bifunctional enzyme PglB (Neisseria gonorrhoeae) and the full-length acetyltransferase WeeI (Acinetobacter baumannii). Additionally, a PglB-ATD structure was solved in complex with AcCoA. Surprisingly, this structure reveals a contrasting binding mechanism for this substrate when compared with the AcCoA-bound PglD structure. A comparison between these findings and the previously solved PglD crystal structures illustrates a dichotomy among N- and O-linked glycosylation pathway enzymes. Based upon these structures, key residues in the UDP-4-amino and AcCoA binding pockets were mutated to determine their effect on binding and catalysis in PglD, PglB-ATD, and WeeI. Last, a phylogenetic analysis of the aforementioned acetyltransferases was employed to illuminate the diversity among N- and O-linked glycosylation pathway enzymes.


Subject(s)
Acetylglucosamine/analogs & derivatives , Acetyltransferases/chemistry , Acinetobacter baumannii/enzymology , Bacterial Proteins/chemistry , Neisseria gonorrhoeae/enzymology , Uridine Diphosphate Sugars/biosynthesis , Acetyl Coenzyme A , Acetylglucosamine/biosynthesis , Acetylglucosamine/chemistry , Acetylglucosamine/genetics , Acetyltransferases/genetics , Acetyltransferases/metabolism , Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Glycosylation , Mutation , Neisseria gonorrhoeae/genetics , Protein Structure, Tertiary , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Sugars/genetics
8.
J Biol Chem ; 287(2): 879-92, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22102281

ABSTRACT

There is increasing evidence that in several fungi, rhamnose-containing glycans are involved in processes that affect host-pathogen interactions, including adhesion, recognition, virulence, and biofilm formation. Nevertheless, little is known about the pathways for the synthesis of these glycans. We show that rhamnose is present in glycans isolated from the rice pathogen Magnaporthe grisea and from the plant pathogen Botryotinia fuckeliana. We also provide evidence that these fungi produce UDP-rhamnose. This is in contrast to bacteria where dTDP-rhamnose is the activated form of this sugar. In bacteria, formation of dTDP-rhamnose requires three enzymes. Here, we demonstrate that in fungi only two genes are required for UDP-Rha synthesis. The first gene encodes a UDP-glucose-4,6-dehydratase that converts UDP-glucose to UDP-4-keto-6-deoxyglucose. The product was shown by time-resolved (1)H NMR spectroscopy to exist in solution predominantly as a hydrated form along with minor amounts of a keto form. The second gene encodes a bifunctional UDP-4-keto-6-deoxyglucose-3,5-epimerase/-4-reductase that converts UDP-4-keto-6-deoxyglucose to UDP-rhamnose. Sugar composition analysis and gene expression studies at different stages of growth indicate that the synthesis of rhamnose-containing glycans is under tissue-specific regulation. Together, our results provide new insight into the formation of rhamnose-containing glycans during the fungal life cycle. The role of these glycans in the interactions between fungal pathogens and their hosts is discussed. Knowledge of the metabolic pathways involved in the formation of rhamnose-containing glycans may facilitate the development of drugs to combat fungal diseases in humans, as to the best of our knowledge mammals do not make these types of glycans.


Subject(s)
Genes, Fungal/physiology , Glucose/analogs & derivatives , Magnaporthe/metabolism , Uridine Diphosphate Sugars/biosynthesis , Uridine Diphosphate/analogs & derivatives , Bacteria/genetics , Bacteria/metabolism , Base Sequence , Glucose/biosynthesis , Glucose/genetics , Magnaporthe/genetics , Magnaporthe/pathogenicity , Molecular Sequence Data , Plant Diseases/genetics , Plant Diseases/microbiology , Uridine Diphosphate/biosynthesis , Uridine Diphosphate/genetics , Uridine Diphosphate Sugars/genetics
9.
Bioorg Med Chem Lett ; 23(13): 3764-8, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23707255

ABSTRACT

Nucleotide sugars are activated forms of monosaccharides and key intermediates of carbohydrate metabolism in all organisms. The availability of structurally diverse nucleotide sugars is particularly important for the characterization of glycosyltransferases. Given that limited methods are available for preparation of nucleotide sugars, especially their useful non-natural derivatives, we introduced herein an efficient one-step three-enzyme catalytic system for the synthesis of nucleotide sugars from monosaccharides. In this study, a promiscuous UDP-sugar pyrophosphorylase (USP) from Arabidopsis thaliana (AtUSP) was used with a galactokinase from Streptococcus pneumoniae TIGR4 (SpGalK) and an inorganic pyrophosphatase (PPase) to effectively synthesize four UDP-sugars. AtUSP has better tolerance for C4-derivatives of Gal-1-P compared to UDP-glucose pyrophosphorylase from S. pneumoniae TIGR4 (SpGalU). Besides, the nucleotide substrate specificity and kinetic parameters of AtUSP were systematically studied. AtUSP exhibited considerable activity toward UTP, dUTP and dTTP, the yield of which was 87%, 85% and 84%, respectively. These results provide abundant information for better understanding of the relationship between substrate specificity and structural features of AtUSP.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Nucleotidyltransferases/metabolism , Uridine Diphosphate Sugars/biosynthesis , Arabidopsis/metabolism , Carbohydrate Conformation , Galactokinase/metabolism , Pyrophosphatases/metabolism , Streptococcus pneumoniae/enzymology , Uridine Diphosphate Sugars/chemistry
10.
Biochemistry ; 51(44): 8844-55, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23072385

ABSTRACT

Human UDP-α-D-xylose synthase (hUXS) is a member of the short-chain dehydrogenase/reductase family of nucleotide-sugar modifying enzymes. hUXS contains a bound NAD(+) cofactor that it recycles by first oxidizing UDP-α-D-glucuronic acid (UGA), and then reducing the UDP-α-D-4-keto-xylose (UX4O) to produce UDP-α-D-xylose (UDX). Despite the observation that purified hUXS contains a bound cofactor, it has been reported that exogenous NAD(+) will stimulate enzyme activity. Here we show that a small fraction of hUXS releases the NADH and UX4O intermediates as products during turnover. The resulting apoenzyme can be rescued by exogenous NAD(+), explaining the apparent stimulatory effect of added cofactor. The slow release of NADH and UX4O as side products by hUXS is reminiscent of the Escherichia coli UGA decarboxylase (ArnA), a related enzyme that produces NADH and UX4O as products. We report that ArnA can rebind NADH and UX4O to slowly make UDX. This means that both enzymes share the same catalytic machinery, but differ in the preferred final product. We present a bifurcated rate equation that explains how the substrate is shunted to the distinct final products. Using a new crystal structure of hUXS, we identify the structural elements of the shunt and propose that the local unfolding of the active site directs reactants toward the preferred products. Finally, we present evidence that the release of NADH and UX4O involves a cooperative conformational change that is conserved in both enzymes.


Subject(s)
Carboxy-Lyases/metabolism , Escherichia coli Proteins/metabolism , Xylose/biosynthesis , Escherichia coli/metabolism , Humans , Kinetics , Metabolic Networks and Pathways , Models, Molecular , NAD/metabolism , Uridine Diphosphate Sugars/biosynthesis
11.
Biochem J ; 439(3): 375-9, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21992098

ABSTRACT

Plant pyrophosphorylases that are capable of producing UDP-sugars, key precursors for glycosylation reactions, include UDP-glucose pyrophosphorylases (A- and B-type), UDP-sugar pyrophosphorylase and UDP-N-acetylglucosamine pyrophosphorylase. Although not sharing significant homology at the amino acid sequence level, the proteins share a common structural blueprint. Their structures are characterized by the presence of the Rossmann fold in the central (catalytic) domain linked to enzyme-specific N-terminal and C-terminal domains, which may play regulatory functions. Molecular mobility between these domains plays an important role in substrate binding and catalysis. Evolutionary relationships and the role of (de)oligomerization as a regulatory mechanism are discussed.


Subject(s)
Nucleotidyltransferases/biosynthesis , Nucleotidyltransferases/chemistry , Plant Extracts/chemistry , Plant Proteins/biosynthesis , Plant Proteins/chemistry , Structural Homology, Protein , Uridine Diphosphate Sugars/biosynthesis , Uridine Diphosphate Sugars/chemistry , Animals , Humans , Nucleotidyltransferases/physiology , Phylogeny , Plant Extracts/metabolism , Plant Proteins/physiology , UTP-Glucose-1-Phosphate Uridylyltransferase/biosynthesis , UTP-Glucose-1-Phosphate Uridylyltransferase/chemistry , UTP-Glucose-1-Phosphate Uridylyltransferase/physiology , Uridine Diphosphate Sugars/physiology
12.
Biochemistry ; 50(22): 4936-48, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21542610

ABSTRACT

The O-linked protein glycosylation pathway in Neisseria gonorrhoeae is responsible for the synthesis of a complex oligosaccharide on undecaprenyl diphosphate and subsequent en bloc transfer of the glycan to serine residues of select periplasmic proteins. Protein glycosylation (pgl) genes have been annotated on the basis of bioinformatics and top-down mass spectrometry analysis of protein modifications in pgl-null strains [Aas, F. E., et al. (2007) Mol. Microbiol. 65, 607-624; Vik, A., et al. (2009) Proc. Natl. Acad. Sci. U.S.A. 106, 4447-4452], but relatively little biochemical analysis has been performed to date. In this report, we present the expression, purification, and functional characterization of seven Pgl enzymes. Specifically, the enzymes studied are responsible for synthesis of an uncommon uridine diphosphate (UDP)-sugar (PglD, PglC, and PglB-acetyltransferase domain), glycan assembly (PglB-phospho-glycosyltransferase domain, PglA, PglE, and PglH), and final oligosaccharide transfer (PglO). UDP-2,4-diacetamido-2,4,6-trideoxy-α-d-hexose (DATDH), which is the first sugar in glycan biosynthesis, was produced enzymatically, and the stereochemistry was assigned as uridine diphosphate N'-diacetylbacillosamine (UDP-diNAcBac) by nuclear magnetic resonance characterization. In addition, the substrate specificities of the phospho-glycosyltransferase, glycosyltransferases, and oligosaccharyltransferase (OTase) were analyzed in vitro, and in most cases, these enzymes exhibited strong preferences for the native substrates relative to closely related glycans. In particular, PglO, the O-linked OTase, and PglB(Cj), the N-linked OTase from Campylobacter jejuni, preferred the native N. gonorrhoeae and C. jejuni substrates, respectively. This study represents the first comprehensive biochemical characterization of this important O-linked glycosylation pathway and provides the basis for further investigations of these enzymes as antibacterial targets.


Subject(s)
Acetylglucosamine/analogs & derivatives , Bacterial Proteins/chemistry , Neisseria gonorrhoeae/metabolism , Polysaccharides/biosynthesis , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Hexosyltransferases/genetics , Hexosyltransferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neisseria gonorrhoeae/enzymology , Protein Biosynthesis , Substrate Specificity , Uridine Diphosphate Sugars/biosynthesis , Uridine Diphosphate Sugars/metabolism
13.
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
14.
Microbiology (Reading) ; 157(Pt 1): 260-269, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20847005

ABSTRACT

Sinorhizobium meliloti is a soil bacterium that fixes nitrogen after being established inside nodules that can form on the roots of several legumes, including Medicago truncatula. A mutation in an S. meliloti gene (lpsB) required for lipopolysaccharide synthesis has been reported to result in defective nodulation and an increase in the synthesis of a xylose-containing glycan. Glycans containing xylose as well as arabinose are also formed by other rhizobial species, but little is known about their structures and the biosynthetic pathways leading to their formation. To gain insight into the biosynthesis of these glycans and their biological roles, we report the identification of an operon in S. meliloti 1021 that contains two genes encoding activities not previously described in bacteria. One gene encodes a UDP-xylose synthase (Uxs) that converts UDP-glucuronic acid to UDP-xylose, and the second encodes a UDP-xylose 4-epimerase (Uxe) that interconverts UDP-xylose and UDP-arabinose. Similar genes were also identified in other rhizobial species, including Rhizobium leguminosarum, suggesting that they have important roles in the life cycle of this agronomically important class of bacteria. Functional studies established that recombinant SmUxs1 is likely to be active as a dimer and is inhibited by NADH and UDP-arabinose. SmUxe is inhibited by UDP-galactose, even though this nucleotide sugar is not a substrate for the 4-epimerase. Unambiguous evidence for the conversions of UDP-glucuronic acid to UDP-α-D-xylose and then to UDP-ß-L-arabinose (UDP-arabinopyranose) was obtained using real-time (1)H-NMR spectroscopy. Our results provide new information about the ability of rhizobia to form UDP-xylose and UDP-arabinose, which are then used for the synthesis of xylose- and arabinose-containing glycans.


Subject(s)
Carbohydrate Epimerases/metabolism , Carboxy-Lyases/metabolism , Sinorhizobium meliloti/metabolism , Uridine Diphosphate Sugars/biosynthesis , Uridine Diphosphate Xylose/biosynthesis , Carbohydrate Epimerases/genetics , Carboxy-Lyases/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Dimerization , Enzyme Inhibitors/metabolism , Magnetic Resonance Spectroscopy , Medicago truncatula/microbiology , Molecular Sequence Data , NAD/metabolism , Operon , Rhizobium leguminosarum/genetics , Sequence Analysis, DNA , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate Glucuronic Acid/metabolism
15.
ACS Chem Biol ; 16(10): 1961-1967, 2021 10 15.
Article in English | MEDLINE | ID: mdl-33835779

ABSTRACT

Metabolic oligosaccharide engineering (MOE) has fundamentally contributed to our understanding of protein glycosylation. Efficient MOE reagents are activated into nucleotide-sugars by cellular biosynthetic machineries, introduced into glycoproteins and traceable by bioorthogonal chemistry. Despite their widespread use, the metabolic fate of many MOE reagents is only beginning to be mapped. While metabolic interconnectivity can affect probe specificity, poor uptake by biosynthetic salvage pathways may impact probe sensitivity and trigger side reactions. Here, we use metabolic engineering to turn the weak alkyne-tagged MOE reagents Ac4GalNAlk and Ac4GlcNAlk into efficient chemical tools to probe protein glycosylation. We find that bypassing a metabolic bottleneck with an engineered version of the pyrophosphorylase AGX1 boosts nucleotide-sugar biosynthesis and increases bioorthogonal cell surface labeling by up to two orders of magnitude. A comparison with known azide-tagged MOE reagents reveals major differences in glycoprotein labeling, substantially expanding the toolbox of chemical glycobiology.


Subject(s)
Galactosamine/analogs & derivatives , Galactosamine/metabolism , Galactosyltransferases/metabolism , Glucosamine/analogs & derivatives , Glucosamine/metabolism , Alkynes/chemistry , Amino Acid Sequence , Animals , Azides/chemistry , Cell Line, Tumor , Click Chemistry , Fluorescent Dyes/chemistry , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycosylation , Humans , Metabolic Engineering/methods , Mice , Molecular Probes/chemistry , Oligosaccharides/biosynthesis , Polysaccharides/biosynthesis , Uridine Diphosphate Sugars/biosynthesis , Uridine Diphosphate Sugars/metabolism
16.
Metab Eng ; 12(1): 62-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19782148

ABSTRACT

Hyaluronic acid is a biopolymer with valuable applications in the pharmaceutical and cosmetic industries. Streptococcus equi subspecies zooepidemicus cells transformed with a nisin-inducible, empty plasmid control displayed higher molecular weight. This increase in molecular weight is independent of the nisin promoter or antibiotic resistance. Using 2D DIGE followed by mass spectrometry, we identified up-regulation of the last step in UDP-N-acetyl-glucosamine biosynthesis (GlmU) and down-regulation of the first step in peptidoglycan biosynthesis (MurA) as possible mechanism for the plasmid effect. Over-expression of GlmU to further increase activity had no effect on UDP-N-acetyl-glucosamine levels or molecular weight, while over-expression of MurA reduced UDP-N-acetyl-glucosamine levels and molecular weight. Global transcriptional analysis revealed that differential regulation of GlmU and MurA activity was not reflected in transcription levels. This results, suggest that regulation is at a translational or post-translational level. Differential expression of two clp proteases may explain this effect as well as the small but significant changes in transcription levels of nearly 300 genes.


Subject(s)
Hyaluronic Acid/biosynthesis , Plasmids/metabolism , Streptococcus equi/metabolism , Uridine Diphosphate Sugars/biosynthesis , Alkyl and Aryl Transferases/biosynthesis , Alkyl and Aryl Transferases/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Hyaluronic Acid/genetics , Nisin/pharmacology , Plasmids/genetics , Streptococcus equi/genetics , Uridine Diphosphate Sugars/genetics
17.
Bioprocess Biosyst Eng ; 33(1): 71-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19714366

ABSTRACT

Five recombinant Escherichia coli extracts harboring overexpressed galactokinase, galactose-1-phosphate uridyltransferase, UDP-glucose pyrophophorylase, UMP kinase, and acetate kinase (AK) were utilized for the production of UDP-galactose (UDP-Gal). We analyzed the parameters which limit the yield of UDP-Gal in the reaction, and the reaction was optimized by increasing the concentration of AK. AK was used for the ATP regeneration as well as the conversion of UDP to UTP. The activities of four overexpressed enzymes were identically fixed, and then we increased the activity of AK to 20 times higher than others. The extracts catalyzed the production of UDP-Gal from UMP (10 mM), galactose (12 mM), ATP (1 mM), and acetyl phosphate (40 mM). As the result of the reaction, the conversion yield of UDP-Gal reached to 95% from 10 mMUMP.


Subject(s)
Enzymes/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Uridine Diphosphate Sugars/biosynthesis , Cell-Free System/enzymology , Uridine Diphosphate Sugars/chemistry
18.
Cell Tissue Res ; 337(1): 103-11, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19440735

ABSTRACT

Low oxygen tension may change the dependence of chondrocytes on exogenous carbohydrate sources. In this study, we have investigated whether glucosamine sulphate (GS) stimulates proteoglycan synthesis, the mRNA expression of aggrecan and of type II collagen, and UDP-sugar levels in bovine primary chondrocytes under a low oxygen (O(2)) atmosphere. Chondrocytes from bovine femoral condyles were cultivated with or without GS or sulphate at various concentrations in low- (5.5 mM) or high-glucose (25 mM) DMEM under either a 5% or 20% O(2) atmosphere for 2 or 8 days after isolation. The mRNA expression of aggrecan and type II collagen and the synthesis of glycosaminoglycan (GAG) were determined by quantitative real-time reverse transcription with polymerase chain reaction and a [(35)S]-sulphate incorporation assay, respectively. Aggrecan promoter activity was analysed by a dual-luciferase reporter gene assay. Intracellular UDP-N-acetylhexosamines (UDP-HexN), UDP-glucuronic acid and UDP-hexoses were analysed by reversed-phase high-performance liquid chromatography electrospray ionization mass spectrometry. A low (5%) O(2) atmosphere significantly increased GAG synthesis, mRNA expression of aggrecan and of type II collagen and aggrecan promoter activity in bovine primary chondrocytes. A high (1 mM) concentration of GS was required to increase the level of UDP-HexN. However, GS did not increase GAG synthesis, aggrecan promoter activity or mRNA expression of aggrecan and of type II collagen. Interestingly, a 5% O(2) atmosphere increased the level of UDP-HexN in 8-day cultures without GS treatment. Thus, exogenous GS does not change chondrocyte metabolism, whereas a 5% O(2) atmosphere stimulates extracellular matrix production in bovine primary chondrocytes. The balance of UDP-sugars is changed under a 5% O(2) atmosphere for longer culture periods.


Subject(s)
Extracellular Matrix , Glucosamine/pharmacology , Oxygen/metabolism , Aggrecans/biosynthesis , Aggrecans/genetics , Animals , Cattle , Cell Culture Techniques , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen Type II/biosynthesis , Collagen Type II/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression/drug effects , Glucose/metabolism , Glycosaminoglycans/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Uridine Diphosphate Sugars/biosynthesis
19.
J Cell Biol ; 55(1): 179-85, 1972 Oct.
Article in English | MEDLINE | ID: mdl-4347250

ABSTRACT

Cytochalasin B competitively inhibits the transport of uridine and thymidine by Novikoff rat hepatoma cells growing in suspension culture with apparent K(i)'s of 2 and 6 microM, respectively, but has no effect on the intracellular phosphorylation of the nucleosides. Choline transport is not affected by cytochalasin B. Results from pulse-chase experiments indicate that cytochalasin B has no direct effect on the synthesis of RNA, DNA, or uridine diphosphate-sugars. The inhibition of uridine and thymidine incorporation into nucleic acids by cytochalasin B is solely the consequence of the inhibition of nucleoside transport.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Indoles/pharmacology , Liver Neoplasms/metabolism , Thymidine/metabolism , Uridine/metabolism , Animals , Biological Transport/drug effects , Cells, Cultured , Choline/metabolism , DNA, Neoplasm/biosynthesis , Neoplasms, Experimental/metabolism , RNA, Neoplasm/biosynthesis , Rats , Tritium , Uridine Diphosphate Sugars/biosynthesis
20.
Biotechnol J ; 14(4)2019 Apr.
Article in English | MEDLINE | ID: mdl-30367549

ABSTRACT

The availability of nucleotide sugars is considered as bottleneck for Leloir-glycosyltransferases mediated glycan synthesis. A breakthrough for the synthesis of nucleotide sugars is the development of salvage pathway like enzyme cascades with high product yields from affordable monosaccharide substrates. In this regard, the authors aim at high enzyme productivities of these cascades by a repetitive batch approach. The authors report here for the first time that the exceptional high enzyme cascade stability facilitates the synthesis of Uridine-5'-diphospho-α-d-galactose (UDP-Gal), Uridine-5'-diphospho-N-acetylglucosamine (UDP-GlcNAc), and Uridine-5'-diphospho-N-acetylgalactosamine (UDP-GalNAc) in a multi-gram scale by repetitive batch mode. The authors obtained 12.8 g UDP-Gal through a high mass based total turnover number (TTNmass ) of 494 [gproduct /genzyme ] and space-time-yield (STY) of 10.7 [g/L*h]. Synthesis of UDP-GlcNAc in repetitive batch mode gave 11.9 g product with a TTNmass of 522 [gproduct /genzyme ] and a STY of 9.9 [g/L*h]. Furthermore, the scale-up to a 200 mL scale using a pressure operated concentrator was demonstrated for a UDP-GalNAc producing enzyme cascade resulting in an exceptional high STY of 19.4 [g/L*h] and 23.3 g product. In conclusion, the authors demonstrate that repetitive batch mode is a versatile strategy for the multi-gram scale synthesis of nucleotide sugars by stable enzyme cascades.


Subject(s)
Polysaccharides/chemistry , Uridine Diphosphate Galactose/biosynthesis , Uridine Diphosphate N-Acetylglucosamine/biosynthesis , Uridine Diphosphate Sugars/biosynthesis , Glycosyltransferases/chemistry , Nucleotides/biosynthesis , Nucleotides/chemistry , Transferases (Other Substituted Phosphate Groups)/chemistry , Uridine Diphosphate Galactose/chemistry , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate Sugars/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL