Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 60(51): 26555-26560, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34661966

RESUMEN

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.


Asunto(s)
Galactosiltransferasas/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Conformación de Carbohidratos , Helicobacter pylori/enzimología , Neisseria meningitidis/enzimología , Uridina Difosfato N-Acetilgalactosamina/biosíntesis , Uridina Difosfato N-Acetilgalactosamina/química
2.
Biochem Biophys Res Commun ; 529(4): 984-990, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819609

RESUMEN

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.


Asunto(s)
Dextranos/farmacología , Ficoll/farmacología , Mucina 5AC/metabolismo , Polietilenglicoles/farmacología , Procesamiento Proteico-Postraduccional , Albúmina Sérica Bovina/farmacología , Secuencia de Aminoácidos , Animales , Bovinos , Clonación Molecular , Dextranos/química , Escherichia coli/genética , Escherichia coli/metabolismo , Ficoll/química , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glicosilación/efectos de los fármacos , Células HEK293 , Humanos , Cinética , Mucina 5AC/química , Péptidos/síntesis química , Péptidos/metabolismo , Polietilenglicoles/química , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Albúmina Sérica Bovina/química , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/química , Uridina Difosfato N-Acetilgalactosamina/metabolismo
3.
Anal Chem ; 91(4): 2620-2625, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30657688

RESUMEN

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.


Asunto(s)
Acetilglucosamina/análisis , Proteínas Adaptadoras Transductoras de Señales/química , Péptidos/química , Espectrometría de Masas en Tándem/métodos , alfa-Cristalinas/química , Acetilglucosamina/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Alquinos/química , Azidas/química , Química Clic , Reacción de Cicloadición , Glicosilación , Células HEK293 , Humanos , Oxidación-Reducción , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/química , alfa-Cristalinas/metabolismo
4.
Proteomics ; 17(11)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28394504

RESUMEN

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.


Asunto(s)
Azidas/análisis , Pruebas de Enzimas/métodos , N-Acetilgalactosaminiltransferasas/análisis , Análisis por Matrices de Proteínas/métodos , Proteoma/análisis , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Azidas/metabolismo , Células HEK293 , Humanos , N-Acetilgalactosaminiltransferasas/metabolismo , Isoformas de Proteínas , Especificidad por Sustrato , Uridina Difosfato N-Acetilgalactosamina/análisis , Uridina Difosfato N-Acetilgalactosamina/metabolismo , Polipéptido N-Acetilgalactosaminiltransferasa
5.
Chembiochem ; 18(7): 623-628, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28130882

RESUMEN

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.


Asunto(s)
Óxido de Aluminio/química , Azidas/química , Hexosaminas/química , Sondas Moleculares/química , Nanoestructuras/química , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Animales , Células CHO , Carbocianinas/química , Permeabilidad de la Membrana Celular , Cricetulus , Sistemas de Liberación de Medicamentos , Colorantes Fluorescentes/química , Glicosilación , Procesamiento Proteico-Postraduccional , Rodaminas/química , Uridina Difosfato N-Acetilgalactosamina/química
6.
PLoS One ; 10(3): e0115634, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781966

RESUMEN

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.


Asunto(s)
Acetiltransferasas/química , Acetiltransferasas/metabolismo , Biocatálisis , Helicobacter pylori/metabolismo , Azúcares Ácidos/metabolismo , Acetilcoenzima A/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Helicobacter pylori/enzimología , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Especificidad por Sustrato , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/metabolismo
7.
J Biol Chem ; 288(36): 26201-26208, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23836908

RESUMEN

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.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/química , Inhibidores Enzimáticos/química , N-Acetilgalactosaminiltransferasas/antagonistas & inhibidores , N-Acetilgalactosaminiltransferasas/química , Sistema del Grupo Sanguíneo ABO/metabolismo , Secuencias de Aminoácidos , Catálisis , Humanos , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/metabolismo , Uridina Difosfato Galactosa/análogos & derivados , Uridina Difosfato Galactosa/química , Uridina Difosfato Galactosa/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/química , Uridina Difosfato N-Acetilgalactosamina/metabolismo
8.
Biochem Biophys Res Commun ; 434(3): 473-8, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23583405

RESUMEN

The role of UDP-galactose transporter (UGT; SLC35A2) and UDP-N-acetylglucosamine transporter (NGT; SLC35A3) in glycosylation of macromolecules may be coupled and either of the transporters may partially replace the function played by its partner. The aim of this study was to construct chimeric transporters composed of the N-terminal portion of human NGT and the C-terminal portion of human UGT1 or UGT2 (NGT-UGT1 or NGT-UGT2, respectively), as well as of the N-terminal portion of UGT and C-terminal portion of NGT (UGT-NGT), overexpress them stably in UGT-deficient CHO-Lec8 and MDCK-RCA(r) cells, and characterize their involvement in glycosylation. Two chimeric proteins, NGT-UGT1 and NGT-UGT2, did not overexpress properly. In contrast, UGT-NGT chimeric protein was successfully overexpressed and localized properly to the Golgi apparatus. UGT-NGT chimeric transporter delivered UDP-Gal to the Golgi vesicles of UGT-deficient cells, which resulted in the increased content of galactosylated structures to such an extent that the wild-type phenotypes were completely restored. Our data further support our hypothesis that UGT and NGT cooperate in the UDP-Gal delivery for glycosyltransferases located in the Golgi apparatus. In a wider context, the results gained in this study add to our understanding of glycosylation, one of the basic posttranslational modifications, which affects the majority of macromolecules.


Asunto(s)
Proteínas de Transporte de Monosacáridos/metabolismo , Mutación , Uridina Difosfato Galactosa/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cartilla de ADN , Glicosilación , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/genética , Mutagénesis Sitio-Dirigida , Homología de Secuencia de Aminoácido , Uridina Difosfato N-Acetilgalactosamina/metabolismo
9.
Nat Protoc ; 5(4): 636-46, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20224564

RESUMEN

Enzymatic synthesis using glycosyltransferases is a powerful approach to building polysaccharides with high efficiency and selectivity. Sugar nucleotides are fundamental donor molecules in enzymatic glycosylation reactions by Leloir-type glycosyltransferases. The applications of these donors are restricted by their limited availability. In this protocol, N-acetylglucosamine (GlcNAc)/N-acetylgalactosamine (GalNAc) are phosphorylated by N-acetylhexosamine 1-kinase (NahK) and subsequently pyrophosphorylated by N-acetylglucosamine uridyltransferase (GlmU) to give UDP-GlcNAc/GalNAc. Other UDP-GlcNAc/GalNAc analogues can also be prepared depending on the tolerance of these enzymes to the modified sugar substrates. Starting from L-fucose, GDP-fucose is constructed by one bifunctional enzyme L-fucose pyrophosphorylase (FKP) via two reactions.


Asunto(s)
Guanosina Difosfato Fucosa/biosíntesis , Uridina Difosfato N-Acetilgalactosamina/biosíntesis , Uridina Difosfato N-Acetilglucosamina/biosíntesis , Glicosilación , Glicosiltransferasas/metabolismo , Oligosacáridos/biosíntesis , Oligosacáridos/química , Fenómenos Químicos Orgánicos , Fosforilación , Polisacáridos/biosíntesis , Polisacáridos/química , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilglucosamina/análogos & derivados
10.
J Biol Chem ; 280(21): 20902-8, 2005 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15790564

RESUMEN

Campylobacter jejuni produces multiple glycoproteins whose glycans contain 4-amino 6-deoxy sugars or their derivatives, such as diacetamidobacillosamine or pseudaminic acid. Because the proteoglycans contribute to bacterial virulence and their constitutive sugars are not commonly found in humans, inhibitors developed against the enzymes that are responsible for their biosynthesis could be novel therapeutic targets to fight this important food-borne pathogen. The biosynthesis of diacetamidobacillosamine is anticipated to involve a sugar nucleotide C6 dehydratase, a C4 aminotransferase and an acetyltransferase. We have identified a set of genes (cj1293, cj1294, and cj1298) potentially encoding one of each enzymatic activity, and demonstrated earlier that Cj1293 was a UDP-GlcNAc-specific C6 dehydratase. Others have shown that Cj1293 was involved in protein glycosylation. Here, we report on our investigation of the potential activity of Cj1294 as a sugar nucleotide C4 aminotransferase. Our biochemical characterization of overexpressed and purified protein shows that Cj1294 is a pyridoxal phosphate-dependent aminotransferase specific for UDP-4-keto-6-deoxy-GlcNAc that uses preferentially glutamic acid as an amino donor. A detailed physicokinetic study of Cj1294 was performed to determine the K(m) of 1.28 +/- 0.2 mm and k(cat) of 11.5 +/- 1.3 min(-1). Also, two residues essential for protein stability and activity, Arg(228) and Lys(181), respectively, were identified by site-directed mutagenesis. Finally, we demonstrated by NMR analysis of purified reaction product that Cj1294 produces UDP-4-amino-4,6-dideoxy-GalNAc. These results indicate that Cj1294 is involved in the biosynthesis of diacetamidofucosamine, a C4 epimer of diacetamidobacillosamine not yet described in C. jejuni proteoglycans, suggesting that the composition of C. jejuni proteoglycans is more variable than anticipated.


Asunto(s)
Acetilglucosamina/análogos & derivados , Campylobacter jejuni/enzimología , Transaminasas/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/metabolismo , Acetilglucosamina/biosíntesis , Arginina , Proteínas Bacterianas , Clonación Molecular , Dimerización , Estabilidad de Enzimas , Escherichia coli/genética , Expresión Génica , Concentración de Iones de Hidrógeno , Lisina , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosfato de Piridoxal/farmacología , Proteínas Recombinantes , Relación Estructura-Actividad , Especificidad por Sustrato , Transaminasas/química , Transaminasas/genética
11.
J Am Chem Soc ; 126(1): 6-7, 2004 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-14709032

RESUMEN

The development of rapid screening methods for probing glycosyltransferase activities is essential for advancing the field of glycobiology. While assays for specific glycosyltransferases exist, there is no generalizable method that can be applied across the enzyme superfamily. Herein we describe a novel glycosyltransferase assay that exploits their unnatural substrate tolerance and the unique chemical reactivity of the azide. We applied this "azido-ELISA" to the family of polypeptide alpha-N-acetylgalactosaminyltransferases (ppGalNAcTs), all of which were able to transfer N-azidoacetylgalactosamine (GalNAz) from the unnatural nucleotide sugar donor UDP-GalNAz. The azide was detected and quantified by Staudinger ligation with a phosphine probe in a microtiter plate format. This approach should be applicable to any glycosyltransferase or group-transfer enzyme that tolerates unnatural azido substrates.


Asunto(s)
Azidas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , N-Acetilgalactosaminiltransferasas/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Secuencia de Aminoácidos , Datos de Secuencia Molecular , N-Acetilgalactosaminiltransferasas/química , Especificidad por Sustrato , Uridina Difosfato N-Acetilgalactosamina/metabolismo
13.
Bioorg Med Chem Lett ; 13(11): 1853-6, 2003 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-12749883

RESUMEN

A series of three O-methylated UDP-GalNAc analogues have been synthesised using a divergent strategy from a 3,6-di-O-pivaloyl GlcNAc derivative. The biological activity of these probes toward polypeptide-alpha-GalNAc-transferase T1 has been investigated. This study shows that this glycosyltransferase exhibits a very high substrate specificity.


Asunto(s)
N-Acetilgalactosaminiltransferasas/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Glicosilación , Isoenzimas , Metilación , N-Acetilgalactosaminiltransferasas/antagonistas & inhibidores , Especificidad por Sustrato , Uridina Difosfato N-Acetilgalactosamina/síntesis química , Polipéptido N-Acetilgalactosaminiltransferasa
14.
Biochemistry ; 41(51): 15410-4, 2002 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-12484781

RESUMEN

Plesiomonas shigelloides is a ubiquitous waterborne pathogen responsible for diseases such as diarrhea and bacillary dysentery, commonly afflicting infants and children. This bacterium is endowed with an O-antigen gene cluster consisting of 10 consecutive reading frames. One of these, designated wbgU (orf3), has been overexpressed and biochemically characterized to show that it encodes a uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) C4 epimerase, only the second microbial enzyme characterized to have this activity. Epimerization is an equilibrium reaction resulting in a 70:30 ratio of UDP-GlcNAc to uridine diphosphate-N-acetylgalactosamine (UDP-GalNAc), irrespective of the initial substrate. The K(m) values for UDP-GalNAc and UDP-GlcNAc are 131 microM and 137 microM, respectively. WbgU is also capable of converting nonacetylated derivatives but with much lower efficiency. It contains a tightly bound nicotinamide adenine dinucleotide [NAD(H)] molecule and requires no other cofactors for activity. We propose here that this enzyme catalyzes the first of the three transformations in the biosynthetic pathway of 2-acetamino-2-deoxy-L-altruronic acid, an unusual sugar present in the O-specific side chains of lipopolysaccharide of P. shigelloides O17 and its close relative Escherichia coli Sonnei.


Asunto(s)
Carbohidrato Epimerasas/química , Antígenos O/biosíntesis , Plesiomonas/enzimología , Polisacáridos Bacterianos/biosíntesis , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Ácidos Urónicos/metabolismo , Carbohidrato Epimerasas/biosíntesis , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/aislamiento & purificación , Fenómenos Químicos , Química Física , Electroforesis Capilar , Activación Enzimática , Cinética , Lipopolisacáridos/biosíntesis , NAD/química , NAD/aislamiento & purificación , Oligosacáridos/biosíntesis , Plesiomonas/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Especificidad por Sustrato , Uridina Difosfato N-Acetilgalactosamina/química , Uridina Difosfato N-Acetilglucosamina/química
15.
Carbohydr Res ; 337(21-23): 2187-94, 2002 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-12433482

RESUMEN

UDP-GalNAc analogues with slight modifications in the 2-acetamido group of the GalNAc moiety are prepared in order to study their role in the mechanism of the N-acetylgalactosaminyl transferase mediated glycosylation step. The analogues with N-propionyl-, N-butyryl- and N-bromoacetyl-groups were synthesized, utilizing Khorana's morpholidate coupling method starting from D-galactosaminyl-1-phosphate after selective N-acylation of its amino group with the appropriate N-acyloxysuccinimides. Furthermore, in addition to UDP-galactosamine its 2-azido analogue has been efficiently prepared involving a metal catalyzed diazo transfer reaction.


Asunto(s)
N-Acetilgalactosaminiltransferasas/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/síntesis química , Glicosilación , Espectroscopía de Resonancia Magnética , Unión Proteica , Relación Estructura-Actividad
16.
Acta Biochim Pol ; 47(2): 349-53, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11051199

RESUMEN

Galactosamine (GalN), a well-known hepatotoxin that depletes the cellular pool of uracil nucleotides, was previously shown to have greater impact on the inhibition of protein synthesis in hepatocytes of old rats as compared with young animals (Kmiec 1994, Ann. N.Y. Ac. Sci. 717, 216-225). In the present study we compared the effects of GalN on the nucleotide content (measured by ion-exchange HPLC) in the livers of young (4 months), adult (12 months), and old (24-26 months old) rats two hours after its intraperitoneal administration. UTP content of the livers of old control rats was significantly lower (by 28%) than that of young animals. GalN administration decreased the UTP content in the livers of young, adult and old rats by, respectively, 55%, 65% and 89%, and increased the content of UDP-sugars by 189%, 175% and 305%. The hepatic content of ATP, ADP, AMP, NAD, GTP except CTP did not differ significantly among the age groups of rats studied, and was not changed by GalN treatment. The content of CTP was significantly higher in old rats (P < 0.03) upon GalN treatment. The lower hepatic content of UTP may partially explain the increased sensitivity of hepatocytes and livers of old rats to the action of galactosamine, and possibly to other hepatotoxic compounds that decrease transcription in the liver.


Asunto(s)
Galactosamina/farmacología , Hígado/crecimiento & desarrollo , Uridina Trifosfato/metabolismo , Envejecimiento , Alanina Transaminasa/sangre , Animales , Glucosamina/análogos & derivados , Glucosamina/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratas , Ratas Wistar , Uridina Difosfato Ácido Glucurónico/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/metabolismo
17.
Neurochem Res ; 25(1): 145-52, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10685614

RESUMEN

It is known that ceramide (Cer), the precursor of sphingoglycolipids and of sphingomyelin, participates in events leading to activation of the apoptotic pathway, and per se or through conversion to glucosylceramide (GlcCer) modulates formation of neuritic processes in developing neurons. To learn about the fate of de novo synthesized Cer and GlcCer we examined, in Golgi membranes from chicken embryo neural retina cells, the metabolic relationships of endogenous Cer, GlcCer and lactosylceramide (LacCer). Incubation of the membranes with UDP-[3H]Glc revealed a pool of endogenous Cer useful for synthesis of GlcCer. Most of the GlcCer synthesized, however, was not used for synthesis of LacCer, indicating that it was functionally uncoupled from LacCer synthase. On the other hand, incubation with UDP-[3H]Gal revealed a pool of endogenous GlcCer that depending of the integrity of the membranes was functionally coupled to LacCer and ganglioside synthesis. These results indicate that most GlcCer formed in vitro from Cer is topologically segregated from the synthesis of LacCer. However, subfractionation in sucrose gradients of Golgi membranes labeled with both precursors failed to separate membranes enriched in [3H]GlcCer from those enriched in [3H]Gal-labeled LacCer. It is concluded that despite both transfer steps co-localize in the Golgi membranes, coupling of GlcCer synthesis to LacCer synthesis requires conditions not present in our in vitro assay. This suggests that a coupling activity exists that could be relevant for regulation of the cytoplasmic levels of Cer and GlcCer.


Asunto(s)
Antígenos CD , Ceramidas/metabolismo , Glucosilceramidas/biosíntesis , Aparato de Golgi/metabolismo , Lactosilceramidos/biosíntesis , Retina/embriología , Animales , Centrifugación por Gradiente de Densidad , Embrión de Pollo , Gangliósido G(M1)/metabolismo , Gangliósido G(M3)/metabolismo , Gangliósidos/metabolismo , Glucosamina/análogos & derivados , Glucosamina/metabolismo , Membranas Intracelulares/metabolismo , Retina/metabolismo , Retina/ultraestructura , Tritio , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/metabolismo
18.
J Biol Chem ; 274(7): 4246-53, 1999 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-9933624

RESUMEN

The two hyaluronan synthases (HASs) from Streptococcus pyogenes (spHAS) and Streptococcus equisimilis (seHAS) were expressed in Escherichia coli as recombinant proteins containing His6 tails. The accompanying paper has described the purification and lipid dependence of both HASs, their preference for cardiolipin, and their stability during storage (Tlapak-Simmons, V. L., Baggenstoss, B. A., Clyne, T., and Weigel, P. H. (1999) J. Biol. Chem. 274, 4239-4245). Kinetic characterization of the enzymes in isolated membranes gave Km values for UDP-GlcUA of 40 +/- 4 microM for spHAS and 51 +/- 5 microM for seHAS. In both cases, the Vmax profiles at various concentrations of UDP-GlcNAc were hyperbolic, with no evidence of cooperativity. In contrast, membrane-bound spHAS, but not seHAS, showed sigmoidal behavior as the UDP-GlcNAc concentration was increased, with a Hill number of approximately 2, indicating significant cooperativity. The Hill number for UDP-GlcNAc utilization by seHAS was 1, confirming the lack of cooperativity for UDP-GlcNAc in this enzyme. The Km values for UDP-GlcNAc were 60 +/- 7 microM for seHAS and 149 +/- 3 microM for spHAS in the isolated membranes. The kinetic characteristics of the two affinity-purified HAS enzymes were assessed in the presence of cardiolipin after 8-9 days of storage at -80 degreesC without cardiolipin. With increasing storage time, the enzymes showed a gradual increase in their Km values for both substrates and a decrease in Vmax. Even in the presence of cardiolipin, the detergent-solubilized, purified HASs had substantially higher Km values for both substrates than the membrane-bound enzymes. The KUDP-GlcUA for purified spHAS and seHAS increased 2-4-fold. The KUDP-GlcNAc for spHAS and seHAS increased 4- and 5-fold, respectively. Despite the higher Km values, the Vmax values for the purified HASs were only approximately 50% lower than those for the membrane-bound enzymes. Significantly, purified spHAS displayed the same cooperative interaction with UDP-GlcNAc (nH approximately 2), whereas purified seHAS showed no cooperativity.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Glicosiltransferasas , Proteínas de la Membrana , Streptococcus equi/enzimología , Streptococcus pyogenes/enzimología , Transferasas , Proteínas de Xenopus , Sitios de Unión , Detergentes , Escherichia coli/enzimología , Hialuronano Sintasas , Cinética , Modelos Biológicos , Proteínas Recombinantes/metabolismo , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/metabolismo
19.
J Chromatogr B Biomed Sci Appl ; 696(2): 193-202, 1997 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-9323540

RESUMEN

UDP-N-acetylgalactosamine-4-sulfate (UDP-GalNAc-4-S) was isolated from hen oviduct (isthmus) with a yield of 31 mumol per 100 g of wet tissue and used for arylsulfatase B (ASB) activity determination. Two HPLC methods of separation and quantitation of the reaction product were described: (1) an original gradient elution method which makes it possible to determine the reaction product when only partially purified ASB was used and additional uridine derivatives were formed during incubation; (2) an improved, fast isocratic elution method which may be used in the case of purified ASB preparations, devoid of other nucleotide hydrolysing enzymes. For both methods the detection limit was 0.1 nmol of product with standard error of determination < or = 3%. Using the gradient elution method we have found that UDP-GalNAc-4-S was hydrolysed by bovine arylsulfatase B1 most efficiently at pH 5.0 and concentration 0.5 mM with K(m) = 85 microM.


Asunto(s)
Condro-4-Sulfatasa/análisis , Cromatografía Líquida de Alta Presión/métodos , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Animales , Bovinos , Pollos , Condro-4-Sulfatasa/aislamiento & purificación , Hidrólisis , Cinética , Uridina Difosfato N-Acetilgalactosamina/química
20.
Anal Biochem ; 239(1): 99-106, 1996 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-8660631

RESUMEN

The chemical synthesis and utilization of two photoaffinity analogs, 125I-labeled 5-[3-(p-azidosalicylamido)-1-propenyl]-UDP-GlcNAc and -UDP-GalNAc, is described. Starting with either UDP-GlcNAc or UDP-GalNAc, the synthesis involved the preparation of the 5-mercuri-UDP-HexNAc and then attachment of an allylamine to the 5 position to give 5-(3-amino)allyl-UDP-HexNAc. This was followed by acylation with N-hydroxysuccinimide p-aminosalicylic acid to form the final product, i.e., 5-[3-(p-azidosalicylamido)-1-propenyl]-UDP-GlcNAc or UDP-GalNAc. These products could then be iodinated with chloramine T to give the 125I-derivatives. Both the UDP-GlcNAc and the UDP-GalNAc derivatives reacted in a concentration-dependent manner with a highly purified UDP-HexNAc pyrophosphorylase, and both specifically labeled the subunit(s) of this protein. The labeling of the protein by the UDP-GlcNAc derivative was inhibited in dose-dependent fashion by either unlabeled UDP-GlcNAc or unlabeled UDP-GalNAc. Likewise, labeling with the UDP-GalNAc probe was blocked by either UDP-GlcNAc or UDP-GalNAc. The UDP-GlcNAc probe also specifically labeled a partially purified preparation of GlcNAc transferase I.


Asunto(s)
Marcadores de Afinidad/síntesis química , Glicosiltransferasas/análisis , Nucleotidiltransferasas/análisis , Salicilamidas/síntesis química , Uridina Difosfato Galactosa/análogos & derivados , Uridina Difosfato Glucosa/análogos & derivados , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilglucosamina/análogos & derivados , Animales , Azidas , Fotoquímica , Salicilatos , Porcinos , Uridina Difosfato Galactosa/síntesis química , Uridina Difosfato Glucosa/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...