Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 501
Filter
1.
Chem Commun (Camb) ; 60(70): 9368-9371, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39135501

ABSTRACT

Chemical syntheses of UDP-rhamnose and UDP-arabinofuranose and respective azido-modified analogues are reported. The prepared substrates are useful for the glycan array-based analysis of glycosyltransferases, as exemplified with the plant cell wall-biosynthetic enzymes PvXAT3, AtRRT4 and PtRRT5.


Subject(s)
Glycosyltransferases , Polysaccharides , Uridine Diphosphate Sugars , Glycosyltransferases/metabolism , Glycosyltransferases/chemistry , Polysaccharides/chemistry , Polysaccharides/chemical synthesis , Polysaccharides/metabolism , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Sugars/metabolism , Azides/chemistry , Arabinose/chemistry , Arabinose/analogs & derivatives , Plants/chemistry
2.
Plant J ; 119(1): 508-524, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38678521

ABSTRACT

L-Arabinose (L-Ara) is a plant-specific sugar found in cell wall polysaccharides, proteoglycans, glycoproteins, and small glycoconjugates, which play physiologically important roles in cell proliferation and other essential cellular processes. L-Ara is synthesized as UDP-L-arabinose (UDP-L-Ara) from UDP-xylose (UDP-Xyl) by UDP-Xyl 4-epimerases (UXEs), a type of de novo synthesis of L-Ara unique to plants. In Arabidopsis, the Golgi-localized UXE AtMUR4 is the main contributor to UDP-L-Ara synthesis. However, cytosolic bifunctional UDP-glucose 4-epimerases (UGEs) with UXE activity, AtUGE1, and AtUGE3 also catalyze this reaction. For the present study, we first examined the physiological importance of bifunctional UGEs in Arabidopsis. The uge1 and uge3 mutants enhanced the dwarf phenotype of mur4 and further reduced the L-Ara content in cell walls, suggesting that bifunctional UGEs contribute to UDP-L-Ara synthesis. Through the introduction of point mutations exchanging corresponding amino acid residues between AtUGE1 with high UXE activity and AtUGE2 with low UXE activity, two mutations that increase relative UXE activity of AtUGE2 were identified. The crystal structures of AtUGE2 in complex forms with NAD+ and NAD+/UDP revealed that the UDP-binding domain of AtUGE2 has a more closed conformation and smaller sugar-binding site than bacterial and mammalian UGEs, suggesting that plant UGEs have the appropriate size and shape for binding UDP-Xyl and UDP-L-Ara to exhibit UXE activity. The presented results suggest that the capacity for cytosolic synthesis of UDP-L-Ara was acquired by the small sugar-binding site and several mutations of UGEs, enabling diversified utilization of L-Ara in seed plants.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Wall , Cytosol , UDPglucose 4-Epimerase , Uridine Diphosphate Sugars , Arabidopsis/genetics , Arabidopsis/enzymology , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Cytosol/metabolism , Cytosol/enzymology , Uridine Diphosphate Sugars/metabolism , Cell Wall/metabolism , UDPglucose 4-Epimerase/genetics , UDPglucose 4-Epimerase/metabolism , Mutation , Uridine Diphosphate Xylose/metabolism , Uridine Diphosphate Xylose/genetics
3.
ACS Synth Biol ; 13(4): 1215-1224, 2024 04 19.
Article in English | MEDLINE | ID: mdl-38467016

ABSTRACT

Glycosylation of biomolecules can greatly alter their physicochemical properties, cellular recognition, subcellular localization, and immunogenicity. Glycosylation reactions rely on the stepwise addition of sugars using nucleotide diphosphate (NDP)-sugars. Making these substrates readily available will greatly accelerate the characterization of new glycosylation reactions, elucidation of their underlying regulation mechanisms, and production of glycosylated molecules. In this work, we engineered Saccharomyces cerevisiae to heterologously express nucleotide sugar synthases to access a wide variety of uridine diphosphate (UDP)-sugars from simple starting materials (i.e., glucose and galactose). Specifically, activated glucose, uridine diphosphate d-glucose (UDP-d-Glc), can be converted to UDP-d-glucuronic acid (UDP-d-GlcA), UDP-d-xylose (UDP-d-Xyl), UDP-d-apiose (UDP-d-Api), UDP-d-fucose (UDP-d-Fuc), UDP-l-rhamnose (UDP-l-Rha), UDP-l-arabinopyranose (UDP-l-Arap), and UDP-l-arabinofuranose (UDP-l-Araf) using the corresponding nucleotide sugar synthases of plant and microbial origins. We also expressed genes encoding the salvage pathway to directly activate free sugars to achieve the biosynthesis of UDP-l-Arap and UDP-l-Araf. We observed strong inhibition of UDP-d-Glc 6-dehydrogenase (UGD) by the downstream product UDP-d-Xyl, which we circumvented using an induction system (Tet-On) to delay the production of UDP-d-Xyl to maintain the upstream UDP-sugar pool. Finally, we performed a time-course study using strains containing the biosynthetic pathways to produce five non-native UDP-sugars to elucidate their time-dependent interconversion and the role of UDP-d-Xyl in regulating UDP-sugar metabolism. These engineered yeast strains are a robust platform to (i) functionally characterize sugar synthases in vivo, (ii) biosynthesize a diverse selection of UDP-sugars, (iii) examine the regulation of intracellular UDP-sugar interconversions, and (iv) produce glycosylated secondary metabolites and proteins.


Subject(s)
Nucleotides , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sugars , Uridine Diphosphate Sugars/genetics , Uridine Diphosphate Sugars/metabolism , Xylose
4.
J Biol Chem ; 300(2): 105616, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38159854

ABSTRACT

O-linked ß-N-acetylglucosamine (O-GlcNAcylation) is a dynamic post-translational modification that regulates thousands of proteins and almost all cellular processes. Aberrant O-GlcNAcylation has been associated with numerous diseases, including cancer, neurodegenerative diseases, cardiovascular diseases, and type 2 diabetes. O-GlcNAcylation is highly nutrient-sensitive since it is dependent on UDP-GlcNAc, the end product of the hexosamine biosynthetic pathway (HBP). We previously observed daily rhythmicity of protein O-GlcNAcylation in a Drosophila model that is sensitive to the timing of food consumption. We showed that the circadian clock is pivotal in regulating daily O-GlcNAcylation rhythms given its control of the feeding-fasting cycle and hence nutrient availability. Interestingly, we reported that the circadian clock also modulates daily O-GlcNAcylation rhythm by regulating molecular mechanisms beyond the regulation of food consumption time. A large body of work now indicates that O-GlcNAcylation is likely a generalized cellular status effector as it responds to various cellular signals and conditions, such as ER stress, apoptosis, and infection. In this review, we summarize the metabolic regulation of protein O-GlcNAcylation through nutrient availability, HBP enzymes, and O-GlcNAc processing enzymes. We discuss the emerging roles of circadian clocks in regulating daily O-GlcNAcylation rhythm. Finally, we provide an overview of other cellular signals or conditions that impact O-GlcNAcylation. Many of these cellular pathways are themselves regulated by the clock and/or metabolism. Our review highlights the importance of maintaining optimal O-GlcNAc rhythm by restricting eating activity to the active period under physiological conditions and provides insights into potential therapeutic targets of O-GlcNAc homeostasis under pathological conditions.


Subject(s)
Circadian Clocks , Protein Processing, Post-Translational , Signal Transduction , Animals , Acetylglucosamine/metabolism , Circadian Clocks/physiology , Uridine Diphosphate Sugars/metabolism , Humans
5.
Plant Physiol Biochem ; 197: 107643, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36989989

ABSTRACT

Rhamnosyltransferase (RT) and rhamnose synthase (Rhs) are the key enzymes that are responsible for the biosynthesis of rhamnosides and UDP-l-rhamnose (UDP-Rha) in plants, respectively. How to discover such enzymes efficiently for use is still a problem to be solved. Here, we identified HmF3RT, HmRhs1, and HmRhs2 from Hypericum monogynum, which is abundant in flavonol rhamnosides, with the help of a full-length and high throughput transcriptome sequencing platform. HmF3RT could regiospecifically transfer the rhamnose moiety of UDP-Rha onto the 3-OH position of flavonols and has weakly catalytic for UDP-xylose (UDP-Xyl) and UDP-glucose (UDP-Glc). HmF3RT showed well quercetin substrate affinity and high catalytic efficiency with Km of 5.14 µM and kcat/Km of 2.21 × 105 S-1 M-1, respectively. Docking, dynamic simulation, and mutagenesis studies revealed that V129, D372, and N373 are critical residues for the activity and sugar donor recognition of HmF3RT, mutant V129A, and V129T greatly enhance the conversion rate of catalytic flavonol glucosides. HmRhs1 and HmRhs2 convert UDP-Glc to UDP-Rha, which could be further used by HmF3RT. The HmF3RT and HmRhs1 co-expressed strain RTS1 could produce quercetin 3-O-rhamnoside (quercitrin), kaempferol 3-O-rhamnoside (afzelin), and myricetin 3-O-rhamnoside (myricitrin) at yields of 85.1, 110.7, and 77.6 mg L-1, respectively. It would provide a valuable reference for establishing a better and more efficient biocatalyst for preparing bioactive flavonol rhamnosides by identifying HmF3RT and HmRhs.


Subject(s)
Hypericum , Transferases , Flavonols/metabolism , Hypericum/enzymology , Rhamnose/metabolism , Uridine Diphosphate Sugars/metabolism , Transferases/chemistry , Transferases/metabolism
6.
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
7.
FEBS J ; 288(4): 1163-1178, 2021 02.
Article in English | MEDLINE | ID: mdl-32645249

ABSTRACT

UDP-glucuronic acid (UDP-GlcA) is a central precursor in sugar nucleotide biosynthesis and common substrate for C4-epimerases and decarboxylases releasing UDP-galacturonic acid (UDP-GalA) and UDP-pentose products, respectively. Despite the different reactions catalyzed, the enzymes are believed to share mechanistic analogy rooted in their joint membership to the short-chain dehydrogenase/reductase (SDR) protein superfamily: Oxidation at the substrate C4 by enzyme-bound NAD+ initiates the catalytic pathway. Here, we present mechanistic characterization of the C4-epimerization of UDP-GlcA, which in comparison with the corresponding decarboxylation has been largely unexplored. The UDP-GlcA 4-epimerase from Bacillus cereus functions as a homodimer and contains one NAD+ /subunit (kcat  = 0.25 ± 0.01 s-1 ). The epimerization of UDP-GlcA proceeds via hydride transfer from and to the substrate's C4 while retaining the enzyme-bound cofactor in its oxidized form (≥ 97%) at steady state and without trace of decarboxylation. The kcat for UDP-GlcA conversion shows a kinetic isotope effect of 2.0 (±0.1) derived from substrate deuteration at C4. The proposed enzymatic mechanism involves a transient UDP-4-keto-hexose-uronic acid intermediate whose formation is rate-limiting overall, and is governed by a conformational step before hydride abstraction from UDP-GlcA. Precise positioning of the substrate in a kinetically slow binding step may be important for the epimerase to establish stereo-electronic constraints in which decarboxylation of the labile ß-keto acid species is prevented effectively. Mutagenesis and pH studies implicate the conserved Tyr149 as the catalytic base for substrate oxidation and show its involvement in the substrate positioning step. Collectively, this study suggests that based on overall mechanistic analogy, stereo-electronic control may be a distinguishing feature of catalysis by SDR-type epimerases and decarboxylases active on UDP-GlcA.


Subject(s)
Bacillus cereus/enzymology , Bacterial Proteins/metabolism , Racemases and Epimerases/metabolism , Recombinant Proteins/metabolism , Uridine Diphosphate Sugars/metabolism , Bacterial Proteins/genetics , Biocatalysis , Carbohydrate Sequence , Catalytic Domain , Chromatography, High Pressure Liquid , Escherichia coli/genetics , Hydrogen-Ion Concentration , Kinetics , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation , Racemases and Epimerases/genetics , Recombinant Proteins/genetics , Uridine Diphosphate Sugars/chemistry
8.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 11): 557-567, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33135674

ABSTRACT

Bacterial nonhydrolyzing UDP-N-acetylglucosamine 2-epimerases catalyze the reversible interconversion of UDP-N-acetylglucosamine (UDP-GlcNAc) and UDP-N-acetylmannosamine (UDP-ManNAc). UDP-ManNAc is an important intermediate in the biosynthesis of certain cell-surface polysaccharides, including those in some pathogenic bacteria, such as Neisseria meningitidis and Streptococcus pneumoniae. Many of these epimerases are allosterically regulated by UDP-GlcNAc, which binds adjacent to the active site and is required to initiate UDP-ManNAc epimerization. Here, two crystal structures of UDP-N-acetylglucosamine 2-epimerase from Neisseria meningitidis serogroup A (NmSacA) are presented. One crystal structure is of the substrate-free enzyme, while the other structure contains UDP-GlcNAc substrate bound to the active site. Both structures form dimers as seen in similar epimerases, and substrate binding to the active site induces a large conformational change in which two Rossmann-like domains clamp down on the substrate. Unlike other epimerases, NmSacA does not require UDP-GlcNAc to instigate the epimerization of UDP-ManNAc, although UDP-GlcNAc was found to enhance the rate of epimerization. In spite of the conservation of residues involved in binding the allosteric UDP-GlcNAc observed in similar UDP-GlcNAc 2-epimerases, the structures presented here do not contain UDP-GlcNAc bound in the allosteric site. These structural results provide additional insight into the mechanism and regulation of this critical enzyme and improve the structural understanding of the ability of NmSacA to epimerize modified substrates.


Subject(s)
Neisseria meningitidis, Serogroup A/enzymology , Allosteric Site , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/metabolism , Catalytic Domain , Crystallography, X-Ray , Hydrolysis , Models, Molecular , Protein Conformation , Sodium/chemistry , Sodium/metabolism , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Sugars/metabolism
9.
Plant J ; 104(1): 252-267, 2020 09.
Article in English | MEDLINE | ID: mdl-32662159

ABSTRACT

Rhamnogalacturonan-II (RG-II) is structurally the most complex glycan in higher plants, containing 13 different sugars and 21 distinct glycosidic linkages. Two monomeric RG-II molecules can form an RG-II-borate diester dimer through the two apiosyl (Api) residues of side chain A to regulate cross-linking of pectin in the cell wall. But the relationship of Api biosynthesis and RG-II dimer is still unclear. In this study we investigated the two homologous UDP-D-apiose/UDP-D-xylose synthases (AXSs) in Arabidopsis thaliana that synthesize UDP-D-apiose (UDP-Api). Both AXSs are ubiquitously expressed, while AXS2 has higher overall expression than AXS1 in the tissues analyzed. The homozygous axs double mutant is lethal, while heterozygous axs1/+ axs2 and axs1 axs2/+ mutants display intermediate phenotypes. The axs1/+ axs2 mutant plants are unable to set seed and die. By contrast, the axs1 axs2/+ mutant plants exhibit loss of shoot and root apical dominance. UDP-Api content in axs1 axs2/+ mutants is decreased by 83%. The cell wall of axs1 axs2/+ mutant plants is thicker and contains less RG-II-borate complex than wild-type Col-0 plants. Taken together, these results provide direct evidence of the importance of AXSs for UDP-Api and RG-II-borate complex formation in plant growth and development.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/growth & development , Pectins/metabolism , Uridine Diphosphate Sugars/metabolism , Arabidopsis/metabolism , Arabidopsis Proteins/physiology , Pollen/metabolism
10.
J Agric Food Chem ; 68(35): 9513-9523, 2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32693583

ABSTRACT

UDP-rhamnose is the main type of sugar donor and endows flavonoids with special activity, selectivity, and pharmacological properties by glycosylation. In this study, several UDP-glucose synthesis pathways and UDP-rhamnose synthases were screened to develop an efficient UDP-rhamnose biosynthesis pathway in Escherichia coli. Maximal UDP-rhamnose production reached 82.2 mg/L in the recombinant strain by introducing the cellobiose phosphorolysis pathway and Arabidopsis thaliana UDP-rhamnose synthase (AtRHM). Quercitrin production of 3522 mg/L was achieved in the recombinant strain by coupling the UDP-rhamnose generation system with A. thaliana rhamnosyltransferase (AtUGT78D1) to recycle UDP-rhamnose. To further increase UDP-rhamnose supply, an NADPH-independent fusion enzyme was constructed, the UTP supply was improved, and NADPH regenerators were overexpressed in vivo. Finally, by optimizing the bioconversion conditions, the highest quercitrin production reached 7627 mg/L with the average productivity of 141 mg/(L h), which is the highest yield of quercitrin and efficiency of UDP-rhamnose supply reported to date in E. coli. Therefore, the method described herein for the regeneration of UDP-rhamnose from cellobiose may be widely used for the rhamnosylation of flavonoids and other bioactive substances.


Subject(s)
Escherichia coli/metabolism , Flavonoids/metabolism , NADP/metabolism , Uridine Diphosphate Sugars/metabolism , Cellobiose/metabolism , Escherichia coli/genetics , Flavonoids/chemistry , Glycosylation , Uridine Diphosphate Glucose/metabolism
11.
Metab Eng ; 61: 131-140, 2020 09.
Article in English | MEDLINE | ID: mdl-32454222

ABSTRACT

UDP-glycosyltransferase (UGT)-mediated glycosylation is a widespread modification of plant natural products (PNPs), which exhibit a wide range of bioactivities, and are of great pharmaceutical, ecological and agricultural significance. However, functional annotation is available for less than 2% of the family 1 UGTs, which currently has 20,000 members that are known to glycosylate several classes of PNPs. This low percentage illustrates the difficulty of experimental study and accurate prediction of their function. Here, a synthetic biology platform for elucidating the UGT-mediated glycosylation process of PNPs was established, including glycosyltransferases dependent on UDP-glucose and UDP-xylose. This platform is based on reconstructing the specific PNPs biosynthetic pathways in dedicated microbial yeast chassis by the simple method of plug-and-play. Five UGT enzymes were identified as responsible for the biosynthesis of the main glycosylation products of triterpenes in Panax notoginseng, including a novel UDP-xylose dependent glycosyltransferase enzyme for notoginsenoside R1 biosynthesis. Additionally, we constructed a yeast cell factory that yields >1 g/L of ginsenoside compound K. This platform for functional gene identification and strain engineering can serve as the basis for creating alternative sources of important natural products and thereby protecting natural plant resources.


Subject(s)
Panax notoginseng , Synthetic Biology , Triterpenes/metabolism , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Panax notoginseng/genetics , Panax notoginseng/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Uridine Diphosphate Sugars/genetics , Uridine Diphosphate Sugars/metabolism
12.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326617

ABSTRACT

The purinergic (P2) receptor P2Y14 is the only P2 receptor that is stimulated by uridine diphosphate (UDP)-sugars and its role in bone formation is unknown. We confirmed P2Y14 expression in primary murine osteoblasts (CB-Ob) and the C2C12-BMP2 osteoblastic cell line (C2-Ob). UDP-glucose (UDPG) had undiscernible effects on cAMP levels, however, induced dose-dependent elevations in the cytosolic free calcium concentration ([Ca2+]i) in CB-Ob, but not C2-Ob cells. To antagonize the P2Y14 function, we used the P2Y14 inhibitor PPTN or generated CRISPR-Cas9-mediated P2Y14 knockout C2-Ob clones (Y14KO). P2Y14 inhibition facilitated calcium signalling and altered basal cAMP levels in both models of osteoblasts. Importantly, P2Y14 inhibition augmented Ca2+ signalling in response to ATP, ADP and mechanical stimulation. P2Y14 knockout or inhibition reduced osteoblast proliferation and decreased ERK1/2 phosphorylation and increased AMPKα phosphorylation. During in vitro osteogenic differentiation, P2Y14 inhibition modulated the timing of osteogenic gene expression, collagen deposition, and mineralization, but did not significantly affect differentiation status by day 28. Of interest, while P2ry14-/- mice from the International Mouse Phenotyping Consortium were similar to wild-type controls in bone mineral density, their tibia length was significantly increased. We conclude that P2Y14 in osteoblasts reduces cell responsiveness to mechanical stimulation and mechanotransductive signalling and modulates osteoblast differentiation.


Subject(s)
Cell Proliferation/genetics , Osteoblasts/metabolism , Osteogenesis/genetics , Purinergic Antagonists/pharmacology , Receptors, Purinergic P2Y/metabolism , Signal Transduction/genetics , Uridine Diphosphate Sugars/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Bone Density/genetics , CRISPR-Cas Systems , Calcium/metabolism , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteogenesis/drug effects , Phosphorylation , Purinergic Antagonists/metabolism , Receptors, Purinergic P2Y/genetics , Signal Transduction/drug effects , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Glucose/pharmacology , Uridine Diphosphate Sugars/pharmacology
13.
Glycobiology ; 29(12): 839-846, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31679023

ABSTRACT

l-arabinofuranose is a ubiquitous component of the cell wall and various natural products in plants, where it is synthesized from cytosolic UDP-arabinopyranose (UDP-Arap). The biosynthetic machinery long remained enigmatic in terms of responsible enzymes and subcellular localization. With the discovery of UDP-Arap mutase in plant cytosol, the demonstration of its role in cell-wall arabinose incorporation and the identification of UDP-arabinofuranose transporters in the Golgi membrane, it is clear that the cytosolic UDP-Arap mutases are the key enzymes converting UDP-Arap to UDP-arabinofuranose for cell wall and natural product biosynthesis. This has recently been confirmed by several genotype/phenotype studies. In contrast to the solid evidence pertaining to UDP-Arap mutase function in vivo, the molecular features, including enzymatic mechanism and oligomeric state, remain unknown. However, these enzymes belong to the small family of proteins originally identified as reversibly glycosylated polypeptides (RGPs), which has been studied for >20 years. Here, we review the UDP-Arap mutase and RGP literature together, to summarize and systemize reported molecular characteristics and relations to other proteins.


Subject(s)
Intramolecular Transferases/chemistry , Intramolecular Transferases/metabolism , Oryza/enzymology , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Sugars/metabolism , Biological Products/chemistry , Biological Products/metabolism , Cell Wall/chemistry , Cell Wall/metabolism , Oryza/cytology
14.
Carbohydr Res ; 477: 20-25, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30933787

ABSTRACT

UDP-apiose, a donor substrate of apiosyltransferases, is labile because of its intramolecular self-cyclization ability, resulting in the formation of apiofuranosyl-1,2-cyclic phosphate. Therefore, stabilization of UDP-apiose is indispensable for its availability and identifying and characterizing the apiosyltransferases involved in the biosynthesis of apiosylated sugar chains and glycosides. Here, we established a method for stabilizing UDP-apiose using bulky cations as counter ions. Bulky cations such as triethylamine effectively suppressed the degradation of UDP-apiose in solution. The half-life of UDP-apiose was increased to 48.1 ±â€¯2.4 h at pH 6.0 and 25 °C using triethylamine as a counter cation. UDP-apiose coordinated with a counter cation enabled long-term storage under freezing conditions. UDP-apiose was utilized as a donor substrate for apigenin 7-O-ß-D-glucoside apiosyltransferase to produce the apiosylated glycoside apiin. This apiosyltransferase assay will be useful for identifying genes encoding apiosyltransferases.


Subject(s)
Enzyme Assays/methods , Pentosyltransferases/metabolism , Uridine Diphosphate Sugars/chemical synthesis , Uridine Diphosphate Sugars/metabolism , Carbohydrate Conformation , Pentosyltransferases/genetics , Uridine Diphosphate Sugars/chemistry
15.
J Bacteriol ; 201(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30833352

ABSTRACT

Sinorhizobium meliloti produces multiple extracellular glycans, including among others, lipopolysaccharides (LPS), and the exopolysaccharides (EPS) succinoglycan (SG) and galactoglucan (GG). These polysaccharides serve cell protective roles. Furthermore, SG and GG promote the interaction of S. meliloti with its host Medicago sativa in root nodule symbiosis. ExoB has been suggested to be the sole enzyme catalyzing synthesis of UDP-galactose in S. meliloti (A. M. Buendia, B. Enenkel, R. Köplin, K. Niehaus, et al. Mol Microbiol 5:1519-1530, 1991, https://doi.org/10.1111/j.1365-2958.1991.tb00799.x). Accordingly, exoB mutants were previously found to be affected in the synthesis of the galactose-containing glycans LPS, SG, and GG and consequently, in symbiosis. Here, we report that the S. meliloti Rm2011 uxs1-uxe-apsS-apsH1-apsE-apsH2 (SMb20458-63) gene cluster directs biosynthesis of an arabinose-containing polysaccharide (APS), which contributes to biofilm formation, and is solely or mainly composed of arabinose. Uxe has previously been identified as UDP-xylose 4-epimerase. Collectively, our data from mutational and overexpression analyses of the APS biosynthesis genes and in vitro enzymatic assays indicate that Uxe functions as UDP-xylose 4- and UDP-glucose 4-epimerase catalyzing UDP-xylose/UDP-arabinose and UDP-glucose/UDP-galactose interconversions, respectively. Overexpression of uxe suppressed the phenotypes of an exoB mutant, evidencing that Uxe can functionally replace ExoB. We suggest that under conditions stimulating expression of the APS biosynthesis operon, Uxe contributes to the synthesis of multiple glycans and thereby to cell protection, biofilm formation, and symbiosis. Furthermore, we show that the C2H2 zinc finger transcriptional regulator MucR counteracts the previously reported CuxR-c-di-GMP-mediated activation of the APS biosynthesis operon. This integrates the c-di-GMP-dependent control of APS production into the opposing regulation of EPS biosynthesis and swimming motility in S. melilotiIMPORTANCE Bacterial extracellular polysaccharides serve important cell protective, structural, and signaling roles. They have particularly attracted attention as adhesives and matrix components promoting biofilm formation, which significantly contributes to resistance against antibiotics. In the root nodule symbiosis between rhizobia and leguminous plants, extracellular polysaccharides have a signaling function. UDP-sugar 4-epimerases are important enzymes in the synthesis of the activated sugar substrates, which are frequently shared between multiple polysaccharide biosynthesis pathways. Thus, these enzymes are potential targets to interfere with these pathways. Our finding of a bifunctional UDP-sugar 4-epimerase in Sinorhizobium meliloti generally advances the knowledge of substrate promiscuity of such enzymes and specifically of the biosynthesis of extracellular polysaccharides involved in biofilm formation and symbiosis in this alphaproteobacterium.


Subject(s)
Carbohydrate Epimerases/metabolism , Polysaccharides, Bacterial/biosynthesis , Sinorhizobium meliloti/enzymology , Sinorhizobium meliloti/metabolism , Carbohydrate Epimerases/genetics , Sinorhizobium meliloti/genetics , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate Sugars/metabolism , Uridine Diphosphate Xylose/metabolism
16.
Protein Expr Purif ; 156: 44-49, 2019 04.
Article in English | MEDLINE | ID: mdl-30597216

ABSTRACT

The uridine diphosphate glycosyltransferase (UGT) plays the central role in glycosylation of small molecules by transferring sugars to various acceptors including bioactive natural products in plants. UGT89C1 from Arabidopsis thaliana is a novel UGT, a rhamnosyltransferase, specifically recognizes UDP-l-rhamnose as donor. To provide an insight into the sugar specificity for UDP-l-rhamnose and interactions between UGT89C1 and its substrates, the UGT89C1 was expressed in Escherichia coli and purified toward biochemical and structural studies. Enzyme activity assay was performed, and the recombinant UGT89C1 recognized UDP-l-rhamnose and rhamnosylated kaempferol. Crystals of AtUGT89C1 were obtained, they diffracted to 2.7 Šresolution and belonged to space group I41. AtUGT89C1 was also co-crystallized with UDP. Interestingly, two crystal forms were obtained in the same crystallization condition, including the previous I41 crystal form, and the new crystal form that diffracted to 3.0 Šresolution and belonged to space group P21.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis Proteins/isolation & purification , Arabidopsis/enzymology , Hexosyltransferases/genetics , Hexosyltransferases/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Crystallography, X-Ray , Escherichia coli/enzymology , Hexosyltransferases/chemistry , Hexosyltransferases/metabolism , Kaempferols/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Uridine Diphosphate Sugars/metabolism
17.
Int J Mol Sci ; 20(2)2019 Jan 11.
Article in English | MEDLINE | ID: mdl-30641943

ABSTRACT

Solute carrier family 35 member A5 (SLC35A5) is a member of the SLC35A protein subfamily comprising nucleotide sugar transporters. However, the function of SLC35A5 is yet to be experimentally determined. In this study, we inactivated the SLC35A5 gene in the HepG2 cell line to study a potential role of this protein in glycosylation. Introduced modification affected neither N- nor O-glycans. There was also no influence of the gene knock-out on glycolipid synthesis. However, inactivation of the SLC35A5 gene caused a slight increase in the level of chondroitin sulfate proteoglycans. Moreover, inactivation of the SLC35A5 gene resulted in the decrease of the uridine diphosphate (UDP)-glucuronic acid, UDP-N-acetylglucosamine, and UDP-N-acetylgalactosamine Golgi uptake, with no influence on the UDP-galactose transport activity. Further studies demonstrated that SLC35A5 localized exclusively to the Golgi apparatus. Careful insight into the protein sequence revealed that the C-terminus of this protein is extremely acidic and contains distinctive motifs, namely DXEE, DXD, and DXXD. Our studies show that the C-terminus is directed toward the cytosol. We also demonstrated that SLC35A5 formed homomers, as well as heteromers with other members of the SLC35A protein subfamily. In conclusion, the SLC35A5 protein might be a Golgi-resident multiprotein complex member engaged in nucleotide sugar transport.


Subject(s)
Golgi Apparatus/metabolism , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Solute Carrier Proteins/genetics , Solute Carrier Proteins/metabolism , Uridine Diphosphate Sugars/metabolism , Amino Acid Motifs , Chondroitin Sulfate Proteoglycans/metabolism , Cytosol/metabolism , Gene Knockout Techniques , Glycosylation , Hep G2 Cells , Humans , Nucleotide Transport Proteins/chemistry , Uridine Diphosphate Glucuronic Acid/metabolism , Uridine Diphosphate N-Acetylglucosamine/metabolism
18.
Plant Cell ; 30(12): 3038-3057, 2018 12.
Article in English | MEDLINE | ID: mdl-30429223

ABSTRACT

Glycosylation of small molecules is critical for numerous biological processes in plants, including hormone homeostasis, neutralization of xenobiotics, and synthesis and storage of specialized metabolites. Glycosylation of plant natural products is usually performed by uridine diphosphate-dependent glycosyltransferases (UGTs). Triterpene glycosides (saponins) are a large family of plant natural products that determine important agronomic traits such as disease resistance and flavor and have numerous pharmaceutical applications. Most characterized plant natural product UGTs are glucosyltransferases, and little is known about enzymes that add other sugars. Here we report the discovery and characterization of AsAAT1 (UGT99D1), which is required for biosynthesis of the antifungal saponin avenacin A-1 in oat (Avena strigosa). This enzyme adds l-Ara to the triterpene scaffold at the C-3 position, a modification critical for disease resistance. The only previously reported plant natural product arabinosyltransferase is a flavonoid arabinosyltransferase from Arabidopsis (Arabidopsis thaliana). We show that AsAAT1 has high specificity for UDP-ß-l-arabinopyranose, identify two amino acids required for sugar donor specificity, and through targeted mutagenesis convert AsAAT1 into a glucosyltransferase. We further identify a second arabinosyltransferase potentially implicated in the biosynthesis of saponins that determine bitterness in soybean (Glycine max). Our investigations suggest independent evolution of UDP-Ara sugar donor specificity in arabinosyltransferases in monocots and eudicots.


Subject(s)
Glycosyltransferases/metabolism , Pentosyltransferases/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Avena/genetics , Avena/metabolism , Glycosyltransferases/genetics , Pentosyltransferases/genetics , Saponins/metabolism , Triterpenes/metabolism , Uridine Diphosphate Sugars/genetics , Uridine Diphosphate Sugars/metabolism
19.
J Biol Chem ; 293(49): 19047-19063, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30327429

ABSTRACT

Homogalacturonan (HG) is a pectic glycan in the plant cell wall that contributes to plant growth and development and cell wall structure and function, and interacts with other glycans and proteoglycans in the wall. HG is synthesized by the galacturonosyltransferase (GAUT) gene family. Two members of this family, GAUT1 and GAUT7, form a heteromeric enzyme complex in Arabidopsis thaliana Here, we established a heterologous GAUT expression system in HEK293 cells and show that co-expression of recombinant GAUT1 with GAUT7 results in the production of a soluble GAUT1:GAUT7 complex that catalyzes elongation of HG products in vitro The reaction rates, progress curves, and product distributions exhibited major differences dependent upon small changes in the degree of polymerization (DP) of the oligosaccharide acceptor. GAUT1:GAUT7 displayed >45-fold increased catalytic efficiency with DP11 acceptors relative to DP7 acceptors. Although GAUT1:GAUT7 synthesized high-molecular-weight polymeric HG (>100 kDa) in a substrate concentration-dependent manner typical of distributive (nonprocessive) glycosyltransferases with DP11 acceptors, reactions primed with short-chain acceptors resulted in a bimodal product distribution of glycan products that has previously been reported as evidence for a processive model of GT elongation. As an alternative to the processive glycosyltransfer model, a two-phase distributive elongation model is proposed in which a slow phase, which includes the de novo initiation of HG and elongation of short-chain acceptors, is distinguished from a phase of rapid elongation of intermediate- and long-chain acceptors. Upon reaching a critical chain length of DP11, GAUT1:GAUT7 elongates HG to high-molecular-weight products.


Subject(s)
Arabidopsis Proteins/metabolism , Glucuronosyltransferase/metabolism , Pectins/biosynthesis , Arabidopsis/enzymology , Arabidopsis Proteins/chemistry , Glucuronosyltransferase/chemistry , HEK293 Cells , Humans , Models, Biological , Molecular Structure , Pectins/chemistry , Static Electricity , Substrate Specificity , Uridine Diphosphate Sugars/metabolism
20.
Plant Signal Behav ; 13(8): e1507406, 2018.
Article in English | MEDLINE | ID: mdl-30125142

ABSTRACT

UDP-sugars are key precursors for biomass production in nature (synthesis of cellulose, hemicellulose, etc.). They are produced de novo by distinct UDP-sugar producing pyrophosphorylases. Studies on the roles of these enzymes using genetic knockouts were hampered by sterility of the mutants and by functional-complementation from related enzyme(s), hindering clear interpretation of the results. In an attempt to override these difficulties, we turned to the reverse chemical genetics approaches to identify compounds which interfere with the activity of those enzymes in vivo. Hit expansion on one of such compounds, a salicylimide derivative, allowed us to identify several inhibitors with a range of activities. The present study provides a structure-activity relationship for these compounds.


Subject(s)
Nucleotidyltransferases/metabolism , Uridine Diphosphate Sugars/metabolism , Biomass , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL