Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
Add more filters











Publication year range
1.
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
2.
J Card Surg ; 35(8): 2047-2049, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32652625

ABSTRACT

Mediastinal paragangliomas are very uncommon neuroendocrine neoplasms. Due to their tissue of origin (sympathetic ganglia of the great vessels), they tend to arise deep within pericardial space and, more importantly, intimately attached to great vessels, which makes surgical resection, even with cardiopulmonary bypass, very challenging. This commentary accompanies the case report describing complex surgical management of a paraganglioma located in the anterior mediastinum that was initially thought to be a thymoma.


Subject(s)
Mediastinal Neoplasms/surgery , Paraganglioma/surgery , Thoracic Surgical Procedures/methods , Cardiopulmonary Bypass , Diagnosis, Differential , Diagnostic Imaging , Galactosamine/analogs & derivatives , Humans , Imino Pyranoses , Mediastinal Neoplasms/diagnosis , Mediastinum/surgery , Paraganglioma/diagnosis , Thymoma , Thymus Neoplasms
3.
Org Biomol Chem ; 17(17): 4326-4334, 2019 04 24.
Article in English | MEDLINE | ID: mdl-30976765

ABSTRACT

An unnatural monosaccharide with a C6-azide, Ac36AzGalNAc, has been developed as a potent and selective probe for O-GlcNAc-modified proteins. Combined with click chemistry, we demonstrate that Ac36AzGalNAc can robustly label O-GlcNAc glycosylation in a wide range of cell lines. Meanwhile, cell imaging and LC-MS/MS proteomics verify its selective activity on O-GlcNAc. More importantly, the protocol presented here provides a general methodology for tracking, capturing and identifying unnatural monosaccharide modified proteins in cells or cell lysates.


Subject(s)
Galactosamine/chemistry , Molecular Probes/chemistry , N-Acetylglucosaminyltransferases/analysis , beta-N-Acetylhexosaminidases/analysis , Animals , Cells, Cultured , Galactosamine/analogs & derivatives , Galactosamine/chemical synthesis , Humans , Mice , Molecular Probes/chemical synthesis , Molecular Structure , N-Acetylglucosaminyltransferases/metabolism , beta-N-Acetylhexosaminidases/metabolism
4.
Curr Opin Neurobiol ; 57: 71-80, 2019 08.
Article in English | MEDLINE | ID: mdl-30771697

ABSTRACT

Synapse development depends on a dynamic balance between synapse promoters and suppressors. MDGAs, immunoglobulin superfamily proteins, negatively regulate synapse development through blocking neuroligin-neurexin interactions. Recent analyses of MDGA-neuroligin complexes revealed the structural basis of this activity and indicate that MDGAs interact with all neuroligins with differential affinities. Surprisingly, analyses of mouse mutants revealed a functional divergence, with targeted mutation of Mdga1 and Mdga2 elevating inhibitory and excitatory synapses, respectively, on hippocampal pyramidal neurons. Further research is needed to determine the synapse-specific organizing properties of MDGAs in neural circuits, which may depend on relative levels and subcellular distributions of each MDGA, neuroligin and neurexin. Behavioral deficits in Mdga mutant mice support genetic links to schizophrenia and autism spectrum disorders and raise the possibility of harnessing these interactions for therapeutic purposes.


Subject(s)
Synapses , Animals , Cell Adhesion Molecules, Neuronal , Dansyl Compounds , Galactosamine/analogs & derivatives , Mice , Nerve Tissue Proteins
5.
J Bacteriol ; 200(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29507091

ABSTRACT

Most organisms, from Bacteria to Eukarya, synthesize UDP-N-acetylglucosamine (UDP-GlcNAc) from fructose-6-phosphate via a four-step reaction, and UDP-N-acetylgalactosamine (UDP-GalNAc) can only be synthesized from UDP-GlcNAc by UDP-GlcNAc 4-epimerase. In Archaea, the bacterial-type UDP-GlcNAc biosynthetic pathway was reported for Methanococcales. However, the complete biosynthetic pathways for UDP-GlcNAc and UDP-GalNAc present in one archaeal species are unidentified. Previous experimental analyses on enzymatic activities of the ST0452 protein, identified from the thermophilic crenarchaeon Sulfolobus tokodaii, predicted the presence of both a bacterial-type UDP-GlcNAc and an independent UDP-GalNAc biosynthetic pathway in this archaeon. In the present work, functional analyses revealed that the recombinant ST2186 protein possessed an glutamine:fructose-6-phosphate amidotransferase activity and that the recombinant ST0242 protein possessed a phosphoglucosamine-mutase activity. Along with the acetyltransferase and uridyltransferase activities of the ST0452 protein, the activities of the ST2186 and ST0242 proteins confirmed the presence of a bacterial-type UDP-GlcNAc biosynthetic pathway in S. tokodaii In contrast, the UDP-GlcNAc 4-epimerase homologue gene was not detected within the genomic data. Thus, it was expected that galactosamine-1-phosphate or galactosamine-6-phosphate (GalN-6-P) was provided by conversion of glucosamine-1-phosphate or glucosamine-6-phosphate (GlcN-6-P). A novel epimerase converting GlcN-6-P to GalN-6-P was detected in a cell extract of S. tokodaii, and the N-terminal sequence of the purified protein indicated that the novel epimerase was encoded by the ST2245 gene. Along with the ST0242 phosphogalactosamine-mutase activity, this observation confirmed the presence of a novel UDP-GalNAc biosynthetic pathway from GlcN-6-P in S. tokodaii Discovery of the novel pathway provides a new insight into the evolution of nucleotide sugar metabolic pathways.IMPORTANCE In this work, a novel protein capable of directly converting glucosamine-6-phosphate to galactosamine-6-phosphate was successfully purified from a cell extract of the thermophilic crenarchaeon Sulfolobus tokodaii Confirmation of this novel activity using the recombinant protein indicates that S. tokodaii possesses a novel UDP-GalNAc biosynthetic pathway derived from glucosamine-6-phosphate. The distributions of this and related genes indicate the presence of three different types of UDP-GalNAc biosynthetic pathways: a direct pathway using a novel enzyme and two conversion pathways from UDP-GlcNAc using known enzymes. Additionally, Crenarchaeota species lacking all three pathways were found, predicting the presence of one more unknown pathway. Identification of these novel proteins and pathways provides important insights into the evolution of nucleotide sugar biosynthesis, as well as being potentially important industrially.


Subject(s)
Acetylgalactosamine/biosynthesis , Archaeal Proteins/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Phosphoglucomutase/metabolism , Sulfolobus/enzymology , Uridine Diphosphate N-Acetylglucosamine/biosynthesis , Acetyltransferases/genetics , Acetyltransferases/metabolism , Archaeal Proteins/genetics , Biosynthetic Pathways , Galactosamine/analogs & derivatives , Galactosamine/metabolism , Glucosamine/analogs & derivatives , Glucosamine/metabolism , Glucose-6-Phosphate/analogs & derivatives , Glucose-6-Phosphate/metabolism , Glucosephosphates/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Phosphates/metabolism , Phosphoglucomutase/genetics , Sulfolobus/genetics
6.
Plant Physiol ; 177(1): 24-37, 2018 05.
Article in English | MEDLINE | ID: mdl-29555787

ABSTRACT

With nearly 140 α-glycosidases in 14 different families, plants are well equipped with enzymes that can break the α-glucosidic bonds in a large diversity of molecules. Here, we introduce activity-based protein profiling (ABPP) of α-glycosidases in plants using α-configured cyclophellitol aziridine probes carrying various fluorophores or biotin. In Arabidopsis (Arabidopsis thaliana), these probes label members of the GH31 family of glycosyl hydrolases, including endoplasmic reticulum-resident α-glucosidase-II Radial Swelling3/Priority for Sweet Life5 (RSW3/PSL5) and Golgi-resident α-mannosidase-II Hybrid Glycosylation1 (HGL1), both of which trim N-glycans on glycoproteins. We detected the active state of extracellular α-glycosidases such as α-xylosidase XYL1, which acts on xyloglucans in the cell wall to promote cell expansion, and α-glucosidase AGLU1, which acts in starch hydrolysis and can suppress fungal invasion. Labeling of α-glycosidases generates pH-dependent signals that can be suppressed by α-glycosidase inhibitors in a broad range of plant species. To demonstrate its use on a nonmodel plant species, we applied ABPP on saffron crocus (Crocus sativus), a cash crop for the production of saffron spice. Using a combination of biotinylated glycosidase probes, we identified and quantified 67 active glycosidases in saffron crocus stigma, of which 10 are differentially active. We also uncovered massive changes in hydrolase activities in the corms upon infection with Fusarium oxysporum using multiplex fluorescence labeling in combination with probes for serine hydrolases and cysteine proteases. These experiments demonstrate the ease with which active α-glycosidases and other hydrolases can be analyzed through ABPP in model and nonmodel plants.


Subject(s)
Fluorescent Dyes/chemistry , Glycoside Hydrolases/chemistry , Plant Proteins/metabolism , Proteomics/methods , Acarbose/pharmacology , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Biotinylation , Carbocyanines/chemistry , Catalytic Domain , Crocus/enzymology , Enzyme Inhibitors/pharmacology , Fusarium/pathogenicity , Galactosamine/analogs & derivatives , Galactosamine/pharmacology , Glucosidases/antagonists & inhibitors , Glucosidases/chemistry , Glucosidases/metabolism , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/metabolism , Hydrogen-Ion Concentration , Plant Diseases/microbiology , Plant Proteins/analysis , Plant Proteins/chemistry
7.
Neuron ; 95(4): 729-732, 2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28817794

ABSTRACT

Neurexins and neuroligins form trans-synaptic complexes that promote synapse development. In this issue of Neuron, Aricescu and colleagues (Elegheert et al., 2017) complement and strengthen two recent reports by the Kim and Rudenko teams (Kim et al., 2017; Gangwar et al., 2017) to dissect the molecular determinants by which MDGAs challenge the neurexin-neuroligin partnership.


Subject(s)
Dansyl Compounds/metabolism , Galactosamine/analogs & derivatives , Synapses/physiology , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Galactosamine/metabolism , Multiprotein Complexes/metabolism
8.
Neuron ; 95(4): 896-913.e10, 2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28817804

ABSTRACT

Neuroligin-neurexin (NL-NRX) complexes are fundamental synaptic organizers in the central nervous system. An accurate spatial and temporal control of NL-NRX signaling is crucial to balance excitatory and inhibitory neurotransmission, and perturbations are linked with neurodevelopmental and psychiatric disorders. MDGA proteins bind NLs and control their function and interaction with NRXs via unknown mechanisms. Here, we report crystal structures of MDGA1, the NL1-MDGA1 complex, and a spliced NL1 isoform. Two large, multi-domain MDGA molecules fold into rigid triangular structures, cradling a dimeric NL to prevent NRX binding. Structural analyses guided the discovery of a broad, splicing-modulated interaction network between MDGA and NL family members and helped rationalize the impact of autism-linked mutations. We demonstrate that expression levels largely determine whether MDGAs act selectively or suppress the synapse organizing function of multiple NLs. These results illustrate a potentially brain-wide regulatory mechanism for NL-NRX signaling modulation.


Subject(s)
Dansyl Compounds/metabolism , Galactosamine/analogs & derivatives , Neurturin/metabolism , Signal Transduction/physiology , Synapses/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , COS Cells , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Chickens , Coculture Techniques , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Galactosamine/genetics , Galactosamine/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Molecular , Mutagenesis, Site-Directed , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurturin/genetics , Protein Interaction Maps , Receptors, N-Methyl-D-Aspartate/metabolism , Sequence Alignment
9.
Biochim Biophys Acta Proteins Proteom ; 1865(11 Pt A): 1348-1357, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28844747

ABSTRACT

Erwinia amylovora, a Gram-negative plant pathogen, is the causal agent of Fire Blight, a contagious necrotic disease affecting plants belonging to the Rosaceae family, including apple and pear. E. amylovora is highly virulent and capable of rapid dissemination in orchards; effective control methods are still lacking. One of its most important pathogenicity factors is the exopolysaccharide amylovoran. Amylovoran is a branched polymer made by the repetition of units mainly composed of galactose, with some residues of glucose, glucuronic acid and pyruvate. E. amylovora glucose-1-phosphate uridylyltransferase (UDP-glucose pyrophosphorylase, EC 2.7.7.9) has a key role in amylovoran biosynthesis. This enzyme catalyses the production of UDP-glucose from glucose-1-phosphate and UTP, which the epimerase GalE converts into UDP-galactose, the main building block of amylovoran. We determined EaGalU kinetic parameters and substrate specificity with a range of sugar 1-phosphates. At time point 120min the enzyme catalysed conversion of the sugar 1-phosphate into the corresponding UDP-sugar reached 74% for N-acetyl-α-d-glucosamine 1-phosphate, 28% for α-d-galactose 1-phosphate, 0% for α-d-galactosamine 1-phosphate, 100% for α-d-xylose 1-phosphate, 100% for α-d-glucosamine 1-phosphate, 70% for α-d-mannose 1-phosphate, and 0% for α-d-galacturonic acid 1-phosphate. To explain our results we obtained the crystal structure of EaGalU and augmented our study by docking the different sugar 1-phosphates into EaGalU active site, providing both reliable models for substrate binding and enzyme specificity, and a rationale that explains the different activity of EaGalU on the sugar 1-phosphates used. These data demonstrate EaGalU potential as a biocatalyst for biotechnological purposes, as an alternative to the enzyme from Escherichia coli, besides playing an important role in E. amylovora pathogenicity.


Subject(s)
Bacterial Proteins/chemistry , Erwinia amylovora/enzymology , Glucosephosphates/chemistry , UTP-Glucose-1-Phosphate Uridylyltransferase/chemistry , Uridine Diphosphate Glucose/chemistry , Uridine Triphosphate/chemistry , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Erwinia amylovora/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Galactosamine/analogs & derivatives , Galactosamine/chemistry , Galactosamine/metabolism , Galactosephosphates/chemistry , Galactosephosphates/metabolism , Gene Expression , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Glucosamine/metabolism , Glucosephosphates/metabolism , Kinetics , Mannosephosphates/chemistry , Mannosephosphates/metabolism , Models, Molecular , Molecular Docking Simulation , Pentosephosphates/chemistry , Pentosephosphates/metabolism , Polysaccharides, Bacterial/biosynthesis , Polysaccharides, Bacterial/chemistry , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , UTP-Glucose-1-Phosphate Uridylyltransferase/genetics , UTP-Glucose-1-Phosphate Uridylyltransferase/metabolism , Uridine Diphosphate Glucose/metabolism , Uridine Triphosphate/metabolism
10.
Molecules ; 22(5)2017 May 12.
Article in English | MEDLINE | ID: mdl-28498332

ABSTRACT

A series of novel N-substituted-ß-d-glucosamine derivatives that incorporate benzenesulfonamides were designed using a fragment-based drug design strategy. Each derivative was synthesized and evaluated in vitro for its inhibitory activity against human carbonic anhydrase (hCA) IX; several derivatives displayed desirable potency profiles against this enzyme. The molecular docking studies provided the design rationale and predicted potential binding modes for carbonic anhydrase (CA) IX and three target compounds, including the most potent inhibitor, compound 7f (IC50 = 10.01 nM). Moreover, the calculated Log P (cLog P) values showed that all the compounds tended to be hydrophilic. In addition, topological polar surface area (TPSA) value-based predictions highlighted the selectivity of these carbohydrate-based inhibitors for membrane-associated CA IX.


Subject(s)
Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/pharmacology , Galactosamine/analogs & derivatives , Glucosamine/analogs & derivatives , Sulfonamides/chemical synthesis , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/metabolism , Drug Design , Galactosamine/chemical synthesis , Galactosamine/chemistry , Galactosamine/pharmacology , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Benzenesulfonamides
11.
Sci Rep ; 7(1): 1359, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28465509

ABSTRACT

Growing evidence has shown that gut microbiome is a key factor involved in liver health. Therefore, gut microbiota modulation with probiotic bacteria, such as Saccharomyces boulardii, constitutes a promising therapy for hepatosis. In this study, we aimed to investigate the protective effects of S. boulardii on D-Galactosamine-induced liver injury in mice. Liver function test and histopathological analysis both suggested that the liver injury can be effectively attenuated by S. boulardii administration. In the meantime, S. boulardii induced dramatic changes in the gut microbial composition. At the phylum level, we found that S. boulardii significantly increased in the relative abundance of Bacteroidetes, and decreased the relative abundance of Firmicutes and Proteobacteria, which may explain the hepatic protective effects of S. boulardii. Taken together, our results demonstrated that S. boulardii administration could change the gut microbiota in mice and alleviate acute liver failure, indicating a potential protective and therapeutic role of S. boulardii.


Subject(s)
Chemical and Drug Induced Liver Injury/microbiology , Galactosamine/toxicity , Gastrointestinal Microbiome , Saccharomyces boulardii , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , BALB 3T3 Cells , Chemical and Drug Induced Liver Injury/diet therapy , Chemical and Drug Induced Liver Injury/pathology , Galactosamine/analogs & derivatives , Mice , Probiotics
12.
Free Radic Biol Med ; 91: 10-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26675345

ABSTRACT

Uncontrolled inflammation is a leading cause of various chronic diseases. Cinnamaldehyde (CA) is a major bioactive compound isolated from the essential oil of the leaves of Cinnamomum osmophloeum kaneh that exhibits anti-inflammatory activity; however, the use of CA is limited by its cytotoxicity. Here, we synthesized three CA derivatives and identified 4-hydroxycinnamaldehyde-galactosamine (HCAG) as a low toxicity anti-inflammatory compound in vitro (HCAG IC50 ≫ 1600 µM; CA IC50=40 µM) and in vivo. HCAG reduced pro-inflammatory mediator expression in LPS-activated macrophages by inhibiting MAPK and PKC-α/δ phosphorylation, decreasing ROS generation and reducing NF-κB activation. HCAG also reduced NLRP3 inflammasome-derived IL-1ß secretion by inhibiting the ATP-mediated phosphorylation of AKT and PKC-α/δ. In a mouse model of LPS-induced renal inflammation, we observed reduced albuminuria and a mild degree of glomerular proliferation, glomerular sclerosis and periglomerular inflammation in the HCAG-treated mice compared with the vehicle-treated mice. The underlying mechanisms for these renoprotective effects involved: (1) inhibited NLRP3 inflammasome activation; (2) decreased superoxide anion levels and apoptosis; and (3) suppressed activation of NF-κB and related downstream inflammatory mediators.


Subject(s)
Acrolein/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , Galactosamine/analogs & derivatives , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Nephritis/drug therapy , Acrolein/pharmacology , Animals , Cell Survival/drug effects , Chemokine CCL2/metabolism , Drug Evaluation, Preclinical , Female , Galactosamine/pharmacology , Inflammasomes/antagonists & inhibitors , Interleukin-6/metabolism , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nephritis/immunology , Nephritis/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism
13.
Extremophiles ; 19(2): 417-27, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25567746

ABSTRACT

The ST0452 protein from the thermophilic archaean Sulfolobus tokodaii has been identified as an enzyme with multiple sugar-1-phosphate nucleotidylyltransferase and amino-sugar-1-phosphate acetyltransferase (amino-sugar-1-P AcTase) activities. Analysis of the protein showed that in addition to glucosamine-1-phosphate (GlcN-1-P) AcTase activity, it possesses unique galactosamine-1-phosphate (GalN-1-P) AcTase activity not detected in any other proteins. Comparison of the crystal structures of the ST0452 protein and GlmU from Escherichia coli (EcGlmU), which possesses only GlcN-1-P AcTase activity, showed that the overall sequence identity between these two proteins is less than 25 %, but the amino acid residues predicted to comprise the catalytic center of EcGlmU are conserved in the ST0452 protein. To understand the molecular mechanism by which the ST0452 amino-sugar-1-P AcTase activity recognizes two independent substrates, several ST0452 substitution and truncation mutant proteins were constructed and analyzed. We found that His308 is essential for both GalN-1-P and GlcN-1-P AcTase activities, whereas Tyr311 and Asn331 are important only for the GalN-1-P AcTase activity. In addition, deletion of the C-terminal 5 or 11 residues showed that the 11-residue C-terminal region exerts a modest stimulatory effect on GalN-1-P AcTase activity but dramatically suppresses GlcN-1-P AcTase activity. This region also appears to make an important contribution to the thermostability of the entire ST0452 protein. Systematic deletions from the C-terminus also demonstrated that the C-terminal region with the ß-helix structure has an important role mediating the trimerization of the ST0452 protein. This is the first report of an analysis of a thermostable archaeal enzyme exhibiting multiple amino-sugar-1-P AcTase activities.


Subject(s)
Archaeal Proteins/chemistry , Galactosamine/analogs & derivatives , Galactosephosphates/metabolism , Glucosamine 6-Phosphate N-Acetyltransferase/chemistry , Sulfolobus/enzymology , Amino Acid Sequence , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Escherichia coli Proteins/chemistry , Galactosamine/metabolism , Glucosamine 6-Phosphate N-Acetyltransferase/genetics , Glucosamine 6-Phosphate N-Acetyltransferase/metabolism , Molecular Sequence Data , Multienzyme Complexes/chemistry , Protein Binding , Protein Structure, Tertiary
14.
Org Lett ; 16(21): 5512-5, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25330411

ABSTRACT

The synthesis of 1,2-cis-homoiminosugars bearing an NHAc group at the C-2 position is described. The key step to prepare these α-D-GlcNAc and α-D-GalNAc mimics utilizes a ß-amino alcohol skeletal rearrangement applied to an azepane precursor. This strategy also allows access to naturally occurring α-HGJ and α-HNJ. The α-D-GlcNAc-configured iminosugar was coupled to a glucoside acceptor to yield a novel pseudodisaccharide. Preliminary glycosidase inhibition evaluation indicates that the α-D-GalNAc-configured homoiminosugar is a potent and selective α-N-acetylgalactosaminidase inhibitor.


Subject(s)
Amino Alcohols/chemistry , Amino Sugars/chemistry , Enzyme Inhibitors/chemistry , Galactosamine/chemistry , Glucosamine/chemistry , alpha-N-Acetylgalactosaminidase/antagonists & inhibitors , alpha-N-Acetylgalactosaminidase/chemistry , Galactosamine/analogs & derivatives , Glucosamine/analogs & derivatives , Molecular Structure
15.
Org Lett ; 16(21): 5516-9, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25330462

ABSTRACT

The first synthesis of 1,2-trans-homoiminosugars devised as mimics of ß-D-GlcNAc and α-D-ManNAc is described. Key steps include a regioselective azidolysis of a cyclic sulfite and a ß-amino alcohol skeletal rearrangement applied to a polyhydroxylated azepane. The ß-D-GlcNAc derivative has been coupled to serine to deliver an iminosugar C-amino acid. The two homoiminosugars demonstrate moderate glycosidase inhibition.


Subject(s)
Amino Alcohols/chemical synthesis , Enzyme Inhibitors/chemistry , Galactosamine/chemical synthesis , Glucosamine/chemical synthesis , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/chemistry , Heterocyclic Compounds/chemistry , Amino Alcohols/chemistry , Galactosamine/analogs & derivatives , Galactosamine/chemistry , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure
16.
Nucleic Acids Res ; 42(13): 8796-807, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24992960

ABSTRACT

Triantennary N-acetyl galactosamine (GalNAc, GN3: ), a high-affinity ligand for the hepatocyte-specific asialoglycoprotein receptor (ASGPR), enhances the potency of second-generation gapmer antisense oligonucleotides (ASOs) 6-10-fold in mouse liver. When combined with next-generation ASO designs comprised of short S-cEt (S-2'-O-Et-2',4'-bridged nucleic acid) gapmer ASOs, ∼ 60-fold enhancement in potency relative to the parent MOE (2'-O-methoxyethyl RNA) ASO was observed. GN3: -conjugated ASOs showed high affinity for mouse ASGPR, which results in enhanced ASO delivery to hepatocytes versus non-parenchymal cells. After internalization into cells, the GN3: -ASO conjugate is metabolized to liberate the parent ASO in the liver. No metabolism of the GN3: -ASO conjugate was detected in plasma suggesting that GN3: acts as a hepatocyte targeting prodrug that is detached from the ASO by metabolism after internalization into the liver. GalNAc conjugation also enhanced potency and duration of the effect of two ASOs targeting human apolipoprotein C-III and human transthyretin (TTR) in transgenic mice. The unconjugated ASOs are currently in late stage clinical trials for the treatment of familial chylomicronemia and TTR-mediated polyneuropathy. The ability to translate these observations in humans offers the potential to improve therapeutic index, reduce cost of therapy and support a monthly dosing schedule for therapeutic suppression of gene expression in the liver using ASOs.


Subject(s)
Galactosamine/analogs & derivatives , Glycolipids/chemistry , Hepatocytes/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/chemistry , Animals , Apolipoprotein C-III/genetics , Asialoglycoprotein Receptor/metabolism , Factor XI/antagonists & inhibitors , Galactosamine/chemistry , Humans , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotides, Antisense/metabolism , Prealbumin/antagonists & inhibitors , alpha 1-Antitrypsin
17.
J Am Chem Soc ; 135(38): 14189-97, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-23987472

ABSTRACT

Mucin-type O-glycans form one of the most abundant and complex post-translational modifications (PTM) on cell surface proteins that govern adhesion, migration, and trafficking of hematopoietic cells. Development of targeted approaches to probe functions of O-glycans is at an early stage. Among several approaches, small molecules with unique chemical functional groups that could modulate glycan biosynthesis form a critical tool. Herein, we show that metabolism of peracetyl N-acyl-D-galactosamine derivatives carrying an N-thioglycolyl (Ac5GalNTGc, 1) moiety-but not N-glycolyl (Ac5GalNGc, 2) and N-acetyl (Ac4GalNAc, 3)-through the N-acetyl-D-galactosamine (GalNAc) salvage pathway induced abrogation of MAL-II and PNA epitopes in Jurkat cells. Mass spectrometry of permethylated O-glycans from Jurkat cells confirmed the presence of significant amounts of elaborated O-glycans (sialyl-T and disialyl-T) which were inhibited upon treatment with 1. O-Glycosylation of CD43, a cell surface antigen rich in O-glycans, was drastically reduced by 1 in a thiol-dependent manner. By contrast, only mild effects were observed for CD45 glycoforms. Direct metabolic incorporation of 1 was confirmed by thiol-selective Michael addition reaction of immunoprecipitated CD43-myc/FLAG. Mechanistically, CD43 glycoforms were unperturbed by peracetylated N-(3-acetylthiopropanoyl) (4), N-(4-acetylthiobutanoyl) (5), and N-methylthioacetyl (6) galactosamine derivatives, N-thioglycolyl-D-glucosamine (7, C-4 epimer of 1), and α-O-benzyl 2-acetamido-2-deoxy-D-galactopyranoside (8), confirming the critical requirement of both free sulfhydryl and galactosamine moieties for inhibition of mucin-type O-glycans. Similar, yet differential, effects of 1 were observed for CD43 glycoforms in multiple hematopoietic cells. Development of small molecules that could alter glycan patterns in an antigen-selective and cell-type selective manner might provide avenues for understanding biological functions of glycans.


Subject(s)
Acetylgalactosamine/metabolism , Galactosamine/analogs & derivatives , Mucins/metabolism , Galactosamine/metabolism , Glycosylation , Humans , Jurkat Cells , Leukosialin/metabolism
18.
PLoS One ; 8(6): e67646, 2013.
Article in English | MEDLINE | ID: mdl-23799153

ABSTRACT

We have undertaken an extensive survey of a group of epimerases originally named Gne, that were thought to be responsible for inter-conversion of UDP-N-acetylglucosamine (UDP-GlcNAc) and UDP-N-acetylgalactosamine (UDP-GalNAc). The analysis builds on recent work clarifying the specificity of some of these epimerases. We find three well defined clades responsible for inter-conversion of the gluco- and galacto-configuration at C4 of different N-acetylhexosamines. Their major biological roles are the formation of UDP-GalNAc, UDP-N-acetylgalactosaminuronic acid (UDP-GalNAcA) and undecaprenyl pyrophosphate-N-acetylgalactosamine (UndPP-GalNAc) from the corresponding glucose forms. We propose that the clade of UDP-GlcNAcA epimerase genes be named gnaB and the clade of UndPP-GlcNAc epimerase genes be named gnu, while the UDP-GlcNAc epimerase genes retain the name gne. The Gne epimerases, as now defined after exclusion of those to be named GnaB or Gnu, are in the same clade as the GalE 4-epimerases for inter-conversion of UDP-glucose (UDP-Glc) and UDP-galactose (UDP-Gal). This work brings clarity to an area that had become quite confusing. The identification of distinct enzymes for epimerisation of UDP-GlcNAc, UDP-GlcNAcA and UndPP-GlcNAc will greatly facilitate allocation of gene function in polysaccharide gene clusters, including those found in bacterial genome sequences. A table of the accession numbers for the 295 proteins used in the analysis is provided to enable the major tree to be regenerated with the inclusion of additional proteins of interest. This and other suggestions for annotation of 4-epimerase genes will facilitate annotation.


Subject(s)
Carbohydrate Epimerases/genetics , Escherichia coli Proteins/genetics , Galactosamine/analogs & derivatives , Hexuronic Acids/metabolism , Polyisoprenyl Phosphates/biosynthesis , Uridine Diphosphate N-Acetylglucosamine/biosynthesis , Escherichia coli/enzymology , Escherichia coli/genetics , Galactosamine/biosynthesis , Humans , Molecular Sequence Annotation , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/genetics , Sequence Analysis, Protein , Yersinia enterocolitica/enzymology , Yersinia enterocolitica/genetics , Yersinia pseudotuberculosis/enzymology , Yersinia pseudotuberculosis/genetics
19.
Carbohydr Res ; 374: 1-7, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23603240

ABSTRACT

The diastereospecific formation of ß-N-glycosidically linked α,ß-unsaturated δ-amino aldehyde derivatives has been achieved with high yield via a vinylogous Mannich reaction. The reaction was performed by using a O-pivaloylated galactosyl amine as a chiral template and AlCl3 as a promoter in THF. (S)-8-(p-Nitrophenyl) azocan-2-one can be stereoselective synthesized from (S) ethyl 7-galactosylamino-7-(p-nitrophenyl)hepta-2,4-dienoate by sequential hydrogenation of the double bond, cyclic lactam formation, and removal of the N-glycosidic auxiliary under basic conditions.


Subject(s)
Galactosamine/analogs & derivatives , Galactosamine/chemistry , Lactams/chemical synthesis , Lactams/chemistry , Molecular Structure , Stereoisomerism
20.
J Proteome Res ; 11(12): 6175-86, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23140450

ABSTRACT

Chinese hamster ovary cells (CHO) are the most common cell line used in the production of therapeutic proteins. Understanding the complex pattern of secreted host cell proteins (HCP) that are released by CHO cells will facilitate the development of new recombinant protein production processes. In this study, we have adapted the N-azido-galactosamine (GalNAz) metabolic labeling method to enable the mass spectrometry identification and quantification of secreted proteins in cell culture media. CHO DG44 and CHO-S cells were cultured in media containing GalNAz, which was metabolically incorporated into mucin-type O-linked glycans of secreted proteins. These proteins were effectively enriched using click-chemistry from the cell culture media, allowing for the analysis of secreted proteins across multiple days of cell growth. When compared to the standard method for secretome analysis, the GalNAz method not only increased the total number of proteins identified but dramatically improved the quality of data by decreasing the number of background proteins (cytosolic or nuclear) to essentially zero.


Subject(s)
Click Chemistry/methods , Mucins/metabolism , Proteome/analysis , Proteomics/methods , Animals , CHO Cells , Cell Nucleus/metabolism , Cricetinae , Culture Media/metabolism , Cytoplasm/metabolism , Galactosamine/analogs & derivatives , Galactosamine/metabolism , Glycosylation , Nuclear Proteins/metabolism , Polysaccharides/metabolism , Proteome/metabolism , Reproducibility of Results , Secretory Pathway , Staining and Labeling , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL