Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Anal Methods ; 16(12): 1821-1825, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38433563

ABSTRACT

Uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) is one of the major nucleotide sugars in living organisms and serves as the key donor substrate for the post-translational modification of protein O-GlcNAcylation. It undergoes interconversion to its epimer uridine diphosphate-N-acetylgalactosamine (UDP-GalNAc), which acts as a sugar donor initiating mucin-type O-linked glycosylation. The intracellular levels of the two differ between the cell lines and largely fluctuate in response to metabolic perturbations, and recent studies have focused on the details of their biosynthesis or turnover. However, due to their similar chemical properties, sufficient resolution for the two epimers required non-volatile mobile phases that cannot be applied directly to a mass spectrometer. In this study, to implement simple liquid chromatography-mass spectrometry for UDP-GlcNAc and UDP-GalNAc, we optimized a condition of hydrophilic interaction liquid chromatography-mass spectrometry. We found that the use of ammonium hydroxide and an amide column with an optimized water-acetonitrile ratio, flow rate, and column temperature, provided complete separation of the two. The method allowed the analysis of intracellular levels, a stable isotope-labeled target, and patterns of product ion spectra in a single run with fewer sample preparation steps. The new method can be widely used for mass spectrometric analysis of UDP-GlcNAc and UDP-GalNAc.


Subject(s)
Liquid Chromatography-Mass Spectrometry , Uridine Diphosphate N-Acetylgalactosamine , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Nucleotides , Uridine Diphosphate N-Acetylglucosamine/metabolism , Hydrophobic and Hydrophilic Interactions
2.
Angew Chem Int Ed Engl ; 60(51): 26555-26560, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34661966

ABSTRACT

Accessing large numbers of structurally diverse glycans and derivatives is essential to functional glycomics. We showed a general tolerance of galactosyltransferases toward uridine-diphosphate-galactosamine (UDP-GalN), which is not a commonly used sugar nucleotide donor. The property was harnessed to develop a two-step chemoenzymatic strategy for facile synthesis of novel and divergent N-acetylgalactosamine (GalNAc)-glycosides and derivatives in preparative scales. The discovery and the application of the new property of existing glycosyltransferases expand their catalytic capabilities in generating novel carbohydrate linkages, thus prompting the synthesis of diverse glycans and glycoconjugates for biological studies.


Subject(s)
Galactosyltransferases/metabolism , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Carbohydrate Conformation , Helicobacter pylori/enzymology , Neisseria meningitidis/enzymology , Uridine Diphosphate N-Acetylgalactosamine/biosynthesis , Uridine Diphosphate N-Acetylgalactosamine/chemistry
3.
Proc Natl Acad Sci U S A ; 117(41): 25293-25301, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32989128

ABSTRACT

Protein glycosylation events that happen early in the secretory pathway are often dysregulated during tumorigenesis. These events can be probed, in principle, by monosaccharides with bioorthogonal tags that would ideally be specific for distinct glycan subtypes. However, metabolic interconversion into other monosaccharides drastically reduces such specificity in the living cell. Here, we use a structure-based design process to develop the monosaccharide probe N-(S)-azidopropionylgalactosamine (GalNAzMe) that is specific for cancer-relevant Ser/Thr(O)-linked N-acetylgalactosamine (GalNAc) glycosylation. By virtue of a branched N-acylamide side chain, GalNAzMe is not interconverted by epimerization to the corresponding N-acetylglucosamine analog by the epimerase N-acetylgalactosamine-4-epimerase (GALE) like conventional GalNAc-based probes. GalNAzMe enters O-GalNAc glycosylation but does not enter other major cell surface glycan types including Asn(N)-linked glycans. We transfect cells with the engineered pyrophosphorylase mut-AGX1 to biosynthesize the nucleotide-sugar donor uridine diphosphate (UDP)-GalNAzMe from a sugar-1-phosphate precursor. Tagged with a bioorthogonal azide group, GalNAzMe serves as an O-glycan-specific reporter in superresolution microscopy, chemical glycoproteomics, a genome-wide CRISPR-knockout (CRISPR-KO) screen, and imaging of intestinal organoids. Additional ectopic expression of an engineered glycosyltransferase, "bump-and-hole" (BH)-GalNAc-T2, boosts labeling in a programmable fashion by increasing incorporation of GalNAzMe into the cell surface glycoproteome. Alleviating the need for GALE-KO cells in metabolic labeling experiments, GalNAzMe is a precision tool that allows a detailed view into the biology of a major type of cancer-relevant protein glycosylation.


Subject(s)
Acetylgalactosamine/metabolism , Glycoproteins/metabolism , Acetylgalactosamine/chemistry , Gene Expression Regulation, Enzymologic , Glycosylation , Humans , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Substrate Specificity , Uridine Diphosphate N-Acetylgalactosamine/chemistry
4.
Biochem Biophys Res Commun ; 529(4): 984-990, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32819609

ABSTRACT

OBJECTIVE: To investigate the effects of macromolecular crowding on the folding and aggregation of MUC5AC with different levels of glycosylation during refolding. METHODS: Part 1:An in vitro catalytic reaction comprising the ppGalNAc T2 enzyme, uridine-5'-diphospho-N-galactosamine (UDP-GalNAc) and an 11-amino acid peptide substrate, was used to assess the enzyme activity of the ppGalNAc T2 enzyme in macromolecular crowding environment respectively with bovine serum albumin (BSA), polyethylene glycol (PEG2000), Dextran70 and Ficoll70 at different concentration and temperature. Part 2: The recombinant MUC5AC was expressed in HEK293 cells and purified by nickel column chromatography. The purified protein was treated with PNGase F, and the degree of glycosylation was analyzed by SDS-PAGE. Macromolecular crowding was simulated using PEG2000 at the concentrations of 50, 100, and 200 g/L. Deglycosylated-MUC5AC (d-MUC5AC) and glycosylated MUC5AC (g-MUC5AC) were denatured by GdnHCl and renatured by dilution in a refolding buffer. Protein aggregation was monitored continuously by absorbance reading at 488 nm using a UV spectrophotometer at 25 °C. The refolded proteins were centrifuged, the protein concentration of the supernatant was measured, and refolding yield in different refolding buffers was determined. RESULTS: Enzyme activityof ppGalNAc T2 was observed to increase with increasing crowding agent concentration, with highest enzyme activity at 200 g/L. Compared with the group in the absence of crowding reagent, the refolding yield of g-MUC5AC and d-MUC5AC were reduced significantly in the presence of different concentrations of PEG2000 (200, 100, and 50 g/L). Compared with the dilute solution, aggregation increased significantly in the presence of PEG2000, especially at 200 g/L. Moreover, in the crowded reagent with the same concentration, the refolding yield of d-MUC5AC was higher than that of g-MUC5AC, whereas the degree of aggregation of d-MUC5AC was lower than that of g-MUC5AC. CONCLUSION: The crowded intracellular environment reduces the refolding rate of MUC5AC and strongly induces the misfolding and aggregation of glycosylated MUC5AC.


Subject(s)
Dextrans/pharmacology , Ficoll/pharmacology , Mucin 5AC/metabolism , Polyethylene Glycols/pharmacology , Protein Processing, Post-Translational , Serum Albumin, Bovine/pharmacology , Amino Acid Sequence , Animals , Cattle , Cloning, Molecular , Dextrans/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Ficoll/chemistry , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycosylation/drug effects , HEK293 Cells , Humans , Kinetics , Mucin 5AC/chemistry , Peptides/chemical synthesis , Peptides/metabolism , Polyethylene Glycols/chemistry , Protein Aggregates/drug effects , Protein Folding/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Albumin, Bovine/chemistry , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Uridine Diphosphate N-Acetylgalactosamine/chemistry , Uridine Diphosphate N-Acetylgalactosamine/metabolism
5.
Neuromolecular Med ; 22(2): 171-193, 2020 06.
Article in English | MEDLINE | ID: mdl-31894464

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia and the number of elderly patients suffering from AD has been steadily increasing. Despite worldwide efforts to cope with this disease, little progress has been achieved with regard to identification of effective therapeutics. Thus, active research focusing on identification of new therapeutic targets of AD is ongoing. Among the new targets, post-translational modifications which modify the properties of mature proteins have gained attention. O-GlcNAcylation, a type of PTM that attaches O-linked ß-N-acetylglucosamine (O-GlcNAc) to a protein, is being sought as a new target to treat AD pathologies. O-GlcNAcylation has been known to modify the two important components of AD pathological hallmarks, amyloid precursor protein, and tau protein. In addition, elevating O-GlcNAcylation levels in AD animal models has been shown to be effective in alleviating AD-associated pathology. Although studies investigating the precise mechanism of reversal of AD pathologies by targeting O-GlcNAcylation are not yet complete, it is clearly important to examine O-GlcNAcylation regulation as a target of AD therapeutics. This review highlights the mechanisms of O-GlcNAcylation and its role as a potential therapeutic target under physiological and pathological AD conditions.


Subject(s)
Acetylglucosamine/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/metabolism , Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational/drug effects , tau Proteins/metabolism , Aged , Alzheimer Disease/metabolism , Animals , Antigens, Neoplasm/metabolism , Brain/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Glucose/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/antagonists & inhibitors , Glycosylation/drug effects , Hexosamines/biosynthesis , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Hyaluronoglucosaminidase/antagonists & inhibitors , Hyaluronoglucosaminidase/metabolism , Insulin Resistance , Molecular Structure , Nerve Tissue Proteins/antagonists & inhibitors , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphorylation , Protein Isoforms/metabolism , Stroke/metabolism , Uridine Diphosphate , Uridine Diphosphate N-Acetylgalactosamine/metabolism
6.
Biochem Biophys Res Commun ; 514(1): 157-165, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31029427

ABSTRACT

FOXA1 functions as a pioneer factor of transcriptional regulation that binds to specific sites in the chromatin and recruits other transcription factors, promoting the initiation of gene transcription and mediating the regulation of downstream target gene expression. FOXA1 was reported to facilitate or reprogram ERα binding, thus playing a key function in breast cancer progression. Our previous results indicated that the O-linked N-acetylgalactosamine (O-GalNAc) modification of FOXA1 plays a potentially significant role in the ERα transcription network. However, further investigations are needed to identify the specific mechanism of modification and the specific glycosylation sites on FOXA1. In this study, we first suggested that FOXA1 could be O-GalNAcylated by ppGalNAc-T2 in vitro. By dividing and expressing recombinant FOXA1 as three segments, two O-GalNAcylation sites were found on FOXA1, both located at the C-terminal of the protein. Then, synthesized peptides, including the predicted O-GalNAc sites in the C-terminus of FOXA1, were used in a vitro reaction, and peptides mutated at the predicted O-GalNAc sites were employed as controls. Through an ESI-MS assay, S354 and S355 were identified as probable O-GalNAcylation sites on FOXA1. Additionally, we performed ESI-ETD-MS/MS analysis of the full-length O-GalNAcylated FOXA1 protein and identified S355 as the O-GalNAc modification site on FOXA1, consistent with the peptide reaction. In conclusion, our results demonstrated that FOXA1 can be O-GalNAcylated by ppGalNAc-T2 at S355 in vitro. These results will provide new insights for studying the role of O-GalNAcylation in the development of breast cancer.


Subject(s)
Hepatocyte Nuclear Factor 3-alpha/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Acetylgalactosamine/metabolism , Acylation , Glycosylation , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Serine/metabolism , Spectrometry, Mass, Electrospray Ionization , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Polypeptide N-acetylgalactosaminyltransferase
7.
Anal Chem ; 91(4): 2620-2625, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30657688

ABSTRACT

As a dynamic post-translational modification, O-linked ß- N-acetylglucosamine ( O-GlcNAc) modification (i.e., O-GlcNAcylation) of proteins regulates many biological processes involving cellular metabolism and signaling. However, O-GlcNAc site mapping, a prerequisite for site-specific functional characterization, has been a challenge since its discovery. Herein we present a novel method for O-GlcNAc enrichment and site mapping. In this method, the O-GlcNAc moiety on peptides was labeled with UDP-GalNAz followed by copper-free azide-alkyne cycloaddition with a multifunctional reagent bearing a terminal cyclooctyne, a disulfide bridge, and a biotin handle. The tagged peptides were then released from NeutrAvidin beads upon reductant treatment, alkylated with (3-acrylamidopropyl)trimethylammonium chloride, and subjected to electron-transfer dissociation mass spectrometry analysis. After validation by using standard synthetic peptide gCTD and model protein α-crystallin, such an approach was applied to the site mapping of overexpressed TGF-ß-activated kinase 1/MAP3K7 binding protein 2 (TAB2), with four O-GlcNAc sites unambiguously identified. Our method provides a promising tool for the site-specific characterization of O-GlcNAcylation of important proteins.


Subject(s)
Acetylglucosamine/analysis , Adaptor Proteins, Signal Transducing/chemistry , Peptides/chemistry , Tandem Mass Spectrometry/methods , alpha-Crystallins/chemistry , Acetylglucosamine/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Alkynes/chemistry , Azides/chemistry , Click Chemistry , Cycloaddition Reaction , Glycosylation , HEK293 Cells , Humans , Oxidation-Reduction , Peptides/metabolism , Protein Processing, Post-Translational , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Uridine Diphosphate N-Acetylgalactosamine/chemistry , alpha-Crystallins/metabolism
8.
Int J Hematol ; 108(2): 139-144, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29619625

ABSTRACT

Von Willebrand factor (VWF) is one of the plasma protein carrying ABO(H) blood group antigens, but the combining process of these antigens is not clear. In the present study, we examined whether plasma glycosyltransferase affects the blood group antigens on VWF. VWF expressing H-antigen (H-VWF) from blood group O and bovine serum albumin conjugated with H-antigen (H-BSA) were incubated with recombinant α1-3-N-acetylgalactosaminyltransferase (rA-transferase) and A-plasma with or without an additional UDP-GalNAc. Transformed antigens were detected by western blotting and ELISA, using an anti-A antibody. Both H-VWF and H-BSA acquired the A-antigen after incubation with rA-transferase and UDP-GalNAc. Incubation with A-plasma very weakly converted the H-antigen on BSA and VWF to A-antigen only in the presence of supplemented UDP-GalNAc. This conversion was enhanced on desialylation of H-VWF. These results indicate that sugar chains of plasma VWF can be modified by the external glycosyltransferase, but that plasma glycosyltransferase has no effect on the blood group antigens of VWF due to its low activity and the lack of donor sugars. Further, sialic acid residues of VWF may exert a protective effect against post-translational glycosylation. Our results clearly exclude the possibility that blood group antigens of VWF are constructed extracellularly in plasma.


Subject(s)
ABO Blood-Group System/immunology , Blood Group Antigens/metabolism , Glycosyltransferases/blood , von Willebrand Factor/immunology , Antigens, Bacterial/metabolism , Enzyme-Linked Immunosorbent Assay , Glycosylation , Humans , N-Acetylgalactosaminyltransferases , N-Acetylneuraminic Acid , Plasma/metabolism , Protein Processing, Post-Translational , Recombinant Proteins , Uridine Diphosphate N-Acetylgalactosamine , von Willebrand Factor/chemistry
9.
Mol Biochem Parasitol ; 221: 56-65, 2018 04.
Article in English | MEDLINE | ID: mdl-29581010

ABSTRACT

Cryptosporidium spp. are the causative agents of diarrheal disease worldwide, but effective treatments are lacking. Cryptosporidium employs mucin-like glycoproteins with O-glycans to attach to and infect host intestinal epithelial cells. The Tn antigen (GalNAcα1-Ser/Thr) is an O-glycan essential for these processes, as Tn-specific lectins and a Tn-specific monoclonal antibody block attachment to and infection of host cells in vitro. The enzymes in Cryptosporidium catalyzing their synthesis, however, have not been studied. Previously, we identified four genes encoding putative UDP N-acetyl-α-d-galactosamine:polypeptide N-acetylgalactosaminyltransferases (ppGalNAc-Ts) in the genomes of three Cryptosporidium spp. Here we report the in silico analysis, cloning, expression, purification, and characterization of one of the four enzymes Cryptosporidium parvum (Cp)-ppGalNAc-T4. This enzyme contains the characteristic domains and motifs conserved in ppGalNAc-Ts and is expressed at multiple time points during in vitro infection. Recombinant soluble Cp-ppGalNAc-T4 was enzymatically active against an unmodified EA2 peptide suggesting that it may function as an "initiating" ppGalNAc-T. Cp-ppGalNAc-T4 also exhibited a strong preference for UDP-GalNAc over other nucleotide sugar donors and was active against unmodified and O-glycosylated versions of the C. parvum gp40-derived peptide, with a preference for the former, suggesting it may play a role in modifying this glycoprotein in vivo. Given the importance of mucin-type O-glycosylation in Cryptosporidium spp., the enzymes that catalyze their synthesis may serve as potential therapeutic targets.


Subject(s)
Cryptosporidium parvum/enzymology , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cloning, Molecular , Cryptosporidium parvum/genetics , Epithelial Cells/metabolism , Gene Expression , Gene Expression Profiling , HEK293 Cells , Humans , Models, Molecular , N-Acetylgalactosaminyltransferases/chemistry , N-Acetylgalactosaminyltransferases/isolation & purification , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity , Polypeptide N-acetylgalactosaminyltransferase
10.
Proteomics ; 17(11)2017 Jun.
Article in English | MEDLINE | ID: mdl-28394504

ABSTRACT

O-GalNAc glycosylation is the initial step of the mucin-type O-glycosylation. In humans, it is catalyzed by a family of 20 homologous UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferases (ppGalNAc-Ts). So far, there is very limited information on their protein substrate specificities. In this study, we developed an on-chip ppGalNAc-Ts assay that could rapidly and systematically identify the protein substrates of each ppGalNAc-T. In detail, we utilized a human proteome microarray as the protein substrates and UDP-GalNAz as the nucleotide sugar donor for click chemistry detection. From a total of 16 368 human proteins, we identified 570 potential substrates of ppGalNAc-T1, T2, and T3. Among them, 128 substrates were overlapped, while the rest were isoform specific. Further cluster analysis of these substrates showed that the substrates of ppGalNAc-T1 had a closer phylogenetic relationship with that of ppGalNAc-T3 compared with ppGalNAc-T2, which was consistent with the topology of the phylogenetic tree of these ppGalNAc-Ts. Taken together, our microarray-based enzymatic assay comprehensively reveals the substrate profile of the ppGalNAc-T1, T2, and T3, which not only provides a plausible explanation for their partial functional redundancy as reported, but clearly implies some specialized roles of each enzyme in different biological processes.


Subject(s)
Azides/analysis , Enzyme Assays/methods , N-Acetylgalactosaminyltransferases/analysis , Protein Array Analysis/methods , Proteome/analysis , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Azides/metabolism , HEK293 Cells , Humans , N-Acetylgalactosaminyltransferases/metabolism , Protein Isoforms , Substrate Specificity , Uridine Diphosphate N-Acetylgalactosamine/analysis , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Polypeptide N-acetylgalactosaminyltransferase
11.
Biochem J ; 474(6): 897-905, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28104756

ABSTRACT

The obligate intracellular lifestyle of Plasmodium falciparum and the difficulties in obtaining sufficient amounts of biological material have hampered the study of specific metabolic pathways in the malaria parasite. Thus, for example, the pools of sugar nucleotides required to fuel glycosylation reactions have never been studied in-depth in well-synchronized asexual parasites or in other stages of its life cycle. These metabolites are of critical importance, especially considering the renewed interest in the presence of N-, O-, and other glycans in key parasite proteins. In this work, we adapted a liquid chromatography tandem mass spectrometry (LC-MS/MS) method based on the use of porous graphitic carbon (PGC) columns and MS-friendly solvents to quantify sugar nucleotides in the malaria parasite. We report the thorough quantification of the pools of these metabolites throughout the intraerythrocytic cycle of P. falciparum The sensitivity of the method enabled, for the first time, the targeted analysis of these glycosylation precursors in gametocytes, the parasite sexual stages that are transmissible to the mosquito vector.


Subject(s)
Guanosine Diphosphate Fucose/metabolism , Guanosine Diphosphate Mannose/metabolism , Guanosine Diphosphate Sugars/metabolism , Plasmodium falciparum/metabolism , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate Glucose/metabolism , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Chromatography, Liquid , Erythrocytes/parasitology , Gametogenesis/physiology , Guanosine Diphosphate Fucose/analysis , Guanosine Diphosphate Mannose/analysis , Guanosine Diphosphate Sugars/analysis , Humans , Life Cycle Stages/physiology , Plasmodium falciparum/growth & development , Tandem Mass Spectrometry , Uridine Diphosphate Galactose/analysis , Uridine Diphosphate Glucose/analysis , Uridine Diphosphate N-Acetylgalactosamine/analysis
12.
Chembiochem ; 18(7): 623-628, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28130882

ABSTRACT

Bioorthogonal chemistry is an effective tool for elucidating metabolic pathways and measuring cellular activity, yet its use is currently limited by the difficulty of getting probes past the cell membrane and into the cytoplasm, especially if more complex probes are desired. Here we present a simple and minimally perturbative technique to deliver functional probes of glycosylation into cells by using a nanostructured "nanostraw" delivery system. Nanostraws provide direct intracellular access to cells through fluid conduits that remain small enough to minimize cell perturbation. First, we demonstrate that our platform can deliver an unmodified azidosugar, N-azidoacetylmannosamine, into cells with similar effectiveness to a chemical modification strategy (peracetylation). We then show that the nanostraw platform enables direct delivery of an azidosugar modified with a charged uridine diphosphate group (UDP) that prevents intracellular penetration, thereby bypassing multiple enzymatic processing steps. By effectively removing the requirement for cell permeability from the probe, the nanostraws expand the toolbox of bioorthogonal probes that can be used to study biological processes on a single, easy-to-use platform.


Subject(s)
Aluminum Oxide/chemistry , Azides/chemistry , Hexosamines/chemistry , Molecular Probes/chemistry , Nanostructures/chemistry , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Animals , CHO Cells , Carbocyanines/chemistry , Cell Membrane Permeability , Cricetulus , Drug Delivery Systems , Fluorescent Dyes/chemistry , Glycosylation , Protein Processing, Post-Translational , Rhodamines/chemistry , Uridine Diphosphate N-Acetylgalactosamine/chemistry
13.
Glycoconj J ; 33(5): 789-96, 2016 10.
Article in English | MEDLINE | ID: mdl-27206414

ABSTRACT

The milk/colostrum of some mammalian species is known to contain sugar nucleotides including uridine diphosphate (UDP) oligosaccharides in addition to lactose and milk oligosaccharides, but the detailed structures of these UDP oligosaccharides have not so far been clarified. In this study we isolated two UDP-sialyl N-acetyllactosamines from ovine colostrum and characterized them using (1)H-NMR and MALDI-TOFMS spectroscopies. Their structures were found to be Neu5Gc(α2-3)Gal(ß1-4)GlcNAcα1-UDP and Neu5Gc(α2-6)Gal(ß1-4)GlcNAcα1-UDP.


Subject(s)
Colostrum/chemistry , Uridine Diphosphate N-Acetylgalactosamine/analysis , Animals , Colostrum/metabolism , Female , Magnetic Resonance Spectroscopy , Sheep , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Uridine Diphosphate N-Acetylgalactosamine/metabolism
14.
FEBS Lett ; 589(21): 3258-62, 2015 Oct 24.
Article in English | MEDLINE | ID: mdl-26408204

ABSTRACT

Uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) donates GlcNAc for various glycans and glycoconjugates. We previously found that GlcNAc supplementation increases the UDP-GlcNAc content in Arabidopsis; however, the metabolic pathway was undefined. Here, we show that the homolog of human GlcNAc kinase (GNK) is conserved in land plants. Enzymatic assays of the Arabidopsis homologous protein (AtGNK) revealed kinase activity that was highly specific for GlcNAc. We also demonstrate the role of AtGNK in plants by using its knockout mutant, which presents lower UDP-GlcNAc contents and is insensitive to GlcNAc supplementation. Moreover, our results demonstrate the presence of a GlcNAc salvage pathway in plants.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Uridine Diphosphate N-Acetylgalactosamine/biosynthesis , Acetylglucosamine/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Gene Knockout Techniques , Humans , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sequence Homology, Amino Acid , Signal Transduction , Substrate Specificity
15.
FASEB J ; 29(7): 2993-3002, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25868729

ABSTRACT

Glycosaminoglycan (GAG) polysaccharides have been implicated in a variety of cellular processes, and alterations in their amount and structure have been associated with diseases such as cancer. In this study, we probed 11 sugar analogs for their capacity to interfere with GAG biosynthesis. One analog, with a modification not directly involved in the glycosidic bond formation, 6F-N-acetyl-d-galactosamine (GalNAc) (Ac3), was selected for further study on its metabolic and biologic effect. Treatment of human ovarian carcinoma cells with 50 µM 6F-GalNAc (Ac3) inhibited biosynthesis of GAGs (chondroitin/dermatan sulfate by ∼50-60%, heparan sulfate by ∼35%), N-acetyl-d-glucosamine (GlcNAc)/GalNAc containing glycans recognized by the lectins Datura stramonium and peanut agglutinin (by ∼74 and ∼43%, respectively), and O-GlcNAc protein modification. With respect to function, 6F-GalNAc (Ac3) treatment inhibited growth factor signaling and reduced in vivo angiogenesis by ∼33%. Although the analog was readily transformed in cells into the uridine 5'-diphosphate (UDP)-activated form, it was not incorporated into GAGs. Rather, it strongly reduced cellular UDP-GalNAc and UDP-GlcNAc pools. Together with data from the literature, these findings indicate that nucleotide sugar depletion without incorporation is a common mechanism of sugar analogs for inhibiting GAG/glycan biosynthesis.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Glycosaminoglycans/biosynthesis , Acetylgalactosamine/chemistry , Acetylgalactosamine/pharmacology , Animals , Cell Line , Chick Embryo , Fibroblast Growth Factor 2/metabolism , Glycosaminoglycans/antagonists & inhibitors , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Physiologic/drug effects , Polysaccharides/antagonists & inhibitors , Polysaccharides/biosynthesis , Signal Transduction/drug effects , Structure-Activity Relationship , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Uridine Diphosphate N-Acetylglucosamine/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
PLoS One ; 10(3): e0115634, 2015.
Article in English | MEDLINE | ID: mdl-25781966

ABSTRACT

Helicobacter pylori infection is the common cause of gastroduodenal diseases linked to a higher risk of the development of gastric cancer. Persistent infection requires functional flagella that are heavily glycosylated with 5,7-diacetamido-3,5,7,9-tetradeoxy-L-glycero-L-manno-nonulosonic acid (pseudaminic acid). Pseudaminic acid biosynthesis protein H (PseH) catalyzes the third step in its biosynthetic pathway, producing UDP-2,4-diacetamido-2,4,6-trideoxy-ß-L-altropyranose. It belongs to the GCN5-related N-acetyltransferase (GNAT) superfamily. The crystal structure of the PseH complex with cofactor acetyl-CoA has been determined at 2.3 Å resolution. This is the first crystal structure of the GNAT superfamily member with specificity to UDP-4-amino-4,6-dideoxy-ß-L-AltNAc. PseH is a homodimer in the crystal, each subunit of which has a central twisted ß-sheet flanked by five α-helices and is structurally homologous to those of other GNAT superfamily enzymes. Interestingly, PseH is more similar to the GNAT enzymes that utilize amino acid sulfamoyl adenosine or protein as a substrate than a different GNAT-superfamily bacterial nucleotide-sugar N-acetyltransferase of the known structure, WecD. Analysis of the complex of PseH with acetyl-CoA revealed the location of the cofactor-binding site between the splayed strands ß4 and ß5. The structure of PseH, together with the conservation of the active-site general acid among GNAT superfamily transferases, are consistent with a common catalytic mechanism for this enzyme that involves direct acetyl transfer from AcCoA without an acetylated enzyme intermediate. Based on structural homology with microcin C7 acetyltransferase MccE and WecD, the Michaelis complex can be modeled. The model suggests that the nucleotide- and 4-amino-4,6-dideoxy-ß-L-AltNAc-binding pockets form extensive interactions with the substrate and are thus the most significant determinants of substrate specificity. A hydrophobic pocket accommodating the 6'-methyl group of the altrose dictates preference to the methyl over the hydroxyl group and thus to contributes to substrate specificity of PseH.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Biocatalysis , Helicobacter pylori/metabolism , Sugar Acids/metabolism , Acetyl Coenzyme A/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Helicobacter pylori/enzymology , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Quaternary , Substrate Specificity , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Uridine Diphosphate N-Acetylgalactosamine/metabolism
17.
Proc Natl Acad Sci U S A ; 112(1): 291-6, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535363

ABSTRACT

The formation of glycoconjugates depends on nucleotide sugars, which serve as donor substrates for glycosyltransferases in the lumen of Golgi vesicles and the endoplasmic reticulum (ER). Import of nucleotide sugars from the cytosol is an important prerequisite for these reactions and is mediated by nucleotide sugar transporters. Here, we report the identification of REPRESSOR OF CYTOKININ DEFICIENCY 1 (ROCK1, At5g65000) as an ER-localized facilitator of UDP-N-acetylglucosamine (UDP-GlcNAc) and UDP-N-acetylgalactosamine (UDP-GalNAc) transport in Arabidopsis thaliana. Mutant alleles of ROCK1 suppress phenotypes inferred by a reduced concentration of the plant hormone cytokinin. This suppression is caused by the loss of activity of cytokinin-degrading enzymes, cytokinin oxidases/dehydrogenases (CKXs). Cytokinin plays an essential role in regulating shoot apical meristem (SAM) activity and shoot architecture. We show that rock1 enhances SAM activity and organ formation rate, demonstrating an important role of ROCK1 in regulating the cytokinin signal in the meristematic cells through modulating activity of CKX proteins. Intriguingly, genetic and molecular analysis indicated that N-glycosylation of CKX1 was not affected by the lack of ROCK1-mediated supply of UDP-GlcNAc. In contrast, we show that CKX1 stability is regulated in a proteasome-dependent manner and that ROCK1 regulates the CKX1 level. The increased unfolded protein response in rock1 plants and suppression of phenotypes caused by the defective brassinosteroid receptor bri1-9 strongly suggest that the ROCK1 activity is an important part of the ER quality control system, which determines the fate of aberrant proteins in the secretory pathway.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Carrier Proteins/metabolism , Cytokinins/metabolism , Endoplasmic Reticulum/metabolism , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Uridine Diphosphate N-Acetylglucosamine/metabolism , Arabidopsis/ultrastructure , Biological Transport , Meristem/metabolism , Meristem/ultrastructure , Phenotype
18.
J Biol Chem ; 289(12): 8041-50, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24459149

ABSTRACT

Mammalian members of glycosyltransferase family 6 (GT6) of the CAZy database have a GT-A fold containing a conserved Asp-X-Asp (DXD) sequence that binds an essential metal cofactor. Bacteroides ovatus GT6a represents a GT6 clade found in more than 30 Gram-negative bacteria that is similar in sequence to the catalytic domains of mammalian GT6, but has an Asn(95)-Ala-Asn(97) (NXN) sequence substituted for the DXD motif and metal-independent catalytic activity. Co-crystals of a low activity mutant of BoGT6a (E192Q) with UDP-GalNAc contained protein complexes with intact UDP-GalNAc and two forms with hydrolysis products (UDP plus GalNAc) representing an initial closed complex and later open form primed for product release. Two cationic residues near the C terminus of BoGT6a, Lys(231) and Arg(243), interact with the diphosphate moiety of UDP-GalNAc, but only Lys(231) interacts with the UDP product and may function in leaving group stabilization. The amide group of Asn(95), the first Asn of the NXN motif, interacts with the ribose moiety of the substrate. This metal-independent GT6 resembles its metal-dependent homologs in undergoing conformational changes on binding UDP-GalNAc that arise from structuring the C terminus to cover this substrate. It appears that in the GT6 family, the metal cofactor functions specifically in binding the UDP moiety in the donor substrate and transition state, actions that can be efficiently performed by components of the polypeptide chain.


Subject(s)
Bacteroides/enzymology , Glycosyltransferases/chemistry , Glycosyltransferases/metabolism , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Bacteroides/chemistry , Bacteroides/metabolism , Crystallography, X-Ray , Hydrolysis , Metals/metabolism , Models, Molecular , Protein Conformation , Uridine Diphosphate N-Acetylgalactosamine/chemistry
19.
Chembiochem ; 15(1): 47-56, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24302429

ABSTRACT

Pyranose-furanose mutases are essential enzymes in the life cycle of a number of microorganisms, but are absent in mammalian systems, and hence represent novel targets for drug development. To date, all such mutases show preferential recognition of a single substrate (e.g., UDP-Gal). We report here the detailed structural characterization of the first bifunctional pyranose-furanose mutase, which recognizes both UDP-Gal and UDP-GalNAc. The enzyme under investigation (cjUNGM) is involved in the biosynthesis of capsular polysaccharides (CPSs) in Campylobacter jejuni 11168. These CPSs are known virulence factors that are required for adhesion and invasion of human epithelial cells. Using a combination of UV/visible spectroscopy, X-ray crystallography, saturation transfer difference NMR spectroscopy, molecular dynamics and CORCEMA-ST calculations, we have characterized the binding of the enzyme to both UDP-Galp and UDP-GalpNAc, and compared these interactions with those of a homologous monofunctional mutase enzyme from E. coli (ecUGM). These studies reveal that two arginines in cjUNGM, Arg59 and Arg168, play critical roles in the catalytic mechanism of the enzyme and in controlling its specificity to ultimately lead to a GalfNAc-containing CPS. In ecUGM, these arginines are replaced with histidine and lysine, respectively, and this results in an enzyme that is selective for UDP-Gal. We propose that these changes in amino acids allow C. jejuni 11168 to produce suitable quantities of the sugar nucleotide substrate required for the assembly of a CPS containing GalfNAc, which is essential for viability.


Subject(s)
Bacterial Proteins/metabolism , Campylobacter Infections/therapy , Campylobacter jejuni/enzymology , Intramolecular Transferases/metabolism , Arginine/metabolism , Bacterial Proteins/chemistry , Binding Sites , Biocatalysis , Campylobacter Infections/metabolism , Campylobacter Infections/pathology , Crystallography, X-Ray , Escherichia coli/enzymology , Humans , Intramolecular Transferases/chemistry , Molecular Dynamics Simulation , Protein Binding , Protein Structure, Tertiary , Substrate Specificity , Uridine Diphosphate Galactose/chemistry , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate N-Acetylgalactosamine/chemistry , Uridine Diphosphate N-Acetylgalactosamine/metabolism
20.
J Biol Chem ; 288(36): 26201-26208, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23836908

ABSTRACT

Glycosyltransferases (GTs) are enzymes that are involved, as Nature's "glycosylation reagents," in many fundamental biological processes including cell adhesion and blood group biosynthesis. Although of similar importance to that of other large enzyme families such as protein kinases and proteases, the undisputed potential of GTs for chemical biology and drug discovery has remained largely unrealized to date. This is due, at least in part, to a relative lack of GT inhibitors and tool compounds for structural, mechanistic, and cellular studies. In this study, we have used a novel class of GT donor analogues to obtain new structural and enzymological information for a representative blood group GT. These analogues interfere with the folding of an internal loop and the C terminus, which are essential for catalysis. Our experiments have led to the discovery of an entirely new active site folding mode for this enzyme family, which can be targeted in inhibitor development, similar to the DFG motif in protein kinases. Taken together, our results provide new insights into substrate binding, dynamics, and utilization in this important enzyme family, which can very likely be harnessed for the rational development of new GT inhibitors and probes.


Subject(s)
ABO Blood-Group System/chemistry , Enzyme Inhibitors/chemistry , N-Acetylgalactosaminyltransferases/antagonists & inhibitors , N-Acetylgalactosaminyltransferases/chemistry , ABO Blood-Group System/metabolism , Amino Acid Motifs , Catalysis , Humans , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Uridine Diphosphate Galactose/analogs & derivatives , Uridine Diphosphate Galactose/chemistry , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Uridine Diphosphate N-Acetylgalactosamine/chemistry , Uridine Diphosphate N-Acetylgalactosamine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...