Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
J Chem Inf Model ; 62(3): 632-646, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35043627

RESUMEN

Uridine diphosphate (UDP)-apiose/UDP-xylose synthase (UAXS) is a member of the short-chain dehydrogenase/reductase superfamily (SDR), which catalyzes the ring contraction and closure of UDP-d-glucuronic acid (UDP-GlcA), affording UDP-apiose and UDP-xylose. UAXS is a special enzyme that integrates ring-opening, decarboxylation, rearrangement, and ring closure/contraction in a single active site. Recently, the ternary complex structure of UAXS was crystallized from Arabidopsis thaliana. In this work, to gain insights into the detailed formation mechanism of UDP-apiose and UDP-xylose, an enzyme-substrate reactant model has been constructed and quantum mechanical/molecular mechanical (QM/MM) calculations have been performed. Our calculation results reveal that the reaction starts from the C4-OH oxidation, which is accompanied by the conformational transformation of the sugar ring from chair type to boat type. The sugar ring-opening is prior to decarboxylation, and the deprotonation of the C2-OH group is the prerequisite for sugar ring-opening. Moreover, the keto-enol tautomerization of the decarboxylated intermediate is a necessary step for ring closure/contraction. Based on our calculation results, more UDP-apiose product was expected, which is in line with the experimental observation. Three titratable residues, Tyr185, Cys100, and Cys140, steer the reaction by proton transfer from or to UDP-GlcA, and Arg182, Glu141, and D337 constitute a proton conduit for sugar C2-OH deprotonation. Although Thr139 and Tyr105 are not directly involved in the enzymatic reaction, they are responsible for promoting the catalysis by forming hydrogen-bonding interactions with GlcA. Our calculations may provide useful information for understanding the catalysis of the SDR family.


Asunto(s)
Carboxiliasas , Xilosa , Carboxiliasas/química , Catálisis , Pentosas , Azúcares , Azúcares de Uridina Difosfato/química
2.
J Biol Chem ; 295(35): 12461-12473, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32661196

RESUMEN

UDP-glucuronic acid is converted to UDP-galacturonic acid en route to a variety of sugar-containing metabolites. This reaction is performed by a NAD+-dependent epimerase belonging to the short-chain dehydrogenase/reductase family. We present several high-resolution crystal structures of the UDP-glucuronic acid epimerase from Bacillus cereus The geometry of the substrate-NAD+ interactions is finely arranged to promote hydride transfer. The exquisite complementarity between glucuronic acid and its binding site is highlighted by the observation that the unligated cavity is occupied by a cluster of ordered waters whose positions overlap the polar groups of the sugar substrate. Co-crystallization experiments led to a structure where substrate- and product-bound enzymes coexist within the same crystal. This equilibrium structure reveals the basis for a "swing and flip" rotation of the pro-chiral 4-keto-hexose-uronic acid intermediate that results from glucuronic acid oxidation, placing the C4' atom in position for receiving a hydride ion on the opposite side of the sugar ring. The product-bound active site is almost identical to that of the substrate-bound structure and satisfies all hydrogen-bonding requirements of the ligand. The structure of the apoenzyme together with the kinetic isotope effect and mutagenesis experiments further outlines a few flexible loops that exist in discrete conformations, imparting structural malleability required for ligand rotation while avoiding leakage of the catalytic intermediate and/or side reactions. These data highlight the double nature of the enzymatic mechanism: the active site features a high degree of precision in substrate recognition combined with the flexibility required for intermediate rotation.


Asunto(s)
Bacillus cereus/enzimología , Proteínas Bacterianas/química , Carbohidrato Epimerasas/química , Cristalografía por Rayos X , Ligandos , NAD/química , Oxidación-Reducción , Rotación , Azúcares de Uridina Difosfato/química
3.
Glycobiology ; 29(12): 839-846, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31679023

RESUMEN

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


Asunto(s)
Transferasas Intramoleculares/química , Transferasas Intramoleculares/metabolismo , Oryza/enzimología , Azúcares de Uridina Difosfato/química , Azúcares de Uridina Difosfato/metabolismo , Productos Biológicos/química , Productos Biológicos/metabolismo , Pared Celular/química , Pared Celular/metabolismo , Oryza/citología
4.
Molecules ; 24(17)2019 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-31443364

RESUMEN

Isorhamnetin-3-O-rhamnoside was synthesized by a highly efficient three-enzyme (rhamnosyltransferase, glycine max sucrose synthase and uridine diphosphate (UDP)-rhamnose synthase) cascade using a UDP-rhamnose regeneration system. The rhamnosyltransferase gene (78D1) from Arabidopsis thaliana was cloned, expressed, and characterized in Escherichia coli. The optimal activity was at pH 7.0 and 45 °C. The enzyme was stable over the pH range of 6.5 to 8.5 and had a 1.5-h half-life at 45 °C. The Vmax and Km for isorhamnetin were 0.646 U/mg and 181 µM, respectively. The optimal pH and temperature for synergistic catalysis were 7.5 and 25 °C, and the optimal concentration of substrates were assayed, respectively. The highest titer of isorhamnetin-3-O-rhamnoside production reached 231 mg/L with a corresponding molar conversion of 100%. Isorhamnetin-3-O-rhamnoside was purified and the cytotoxicity against HepG2, MCF-7, and A549 cells were evaluated. Therefore, an efficient method for isorhamnetin-3-O-rhamnoside production described herein could be widely used for the rhamnosylation of flavonoids.


Asunto(s)
Carbohidrato Epimerasas/química , Técnicas de Química Sintética , Flavonoles/síntesis química , Glucosiltransferasas/química , Hexosiltransferasas/química , Azúcares de Uridina Difosfato/química , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Catálisis , Línea Celular Tumoral , Flavonoles/farmacología , Humanos
5.
J Biol Chem ; 292(27): 11499-11507, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28490633

RESUMEN

The enzyme UDP-glucose:glycoprotein glucosyltransferase (UGGT) mediates quality control of glycoproteins in the endoplasmic reticulum by attaching glucose to N-linked glycan of misfolded proteins. As a sensor, UGGT ensures that misfolded proteins are recognized by the lectin chaperones and do not leave the secretory pathway. The structure of UGGT and the mechanism of its selectivity for misfolded proteins have been unknown for 25 years. Here, we used negative-stain electron microscopy and small-angle X-ray scattering to determine the structure of UGGT from Drosophila melanogaster at 18-Å resolution. Three-dimensional reconstructions revealed a cage-like structure with a large central cavity. Particle classification revealed flexibility that precluded determination of a high-resolution structure. Introduction of biotinylation sites into a fungal UGGT expressed in Escherichia coli allowed identification of the catalytic and first thioredoxin-like domains. We also used hydrogen-deuterium exchange mass spectrometry to map the binding site of an accessory protein, Sep15, to the first thioredoxin-like domain. The UGGT structural features identified suggest that the central cavity contains the catalytic site and is lined with hydrophobic surfaces. This enhances the binding of misfolded substrates with exposed hydrophobic residues and excludes folded proteins with hydrophilic surfaces. In conclusion, we have determined the UGGT structure, which enabled us to develop a plausible functional model of the mechanism for UGGT's selectivity for misfolded glycoproteins.


Asunto(s)
Glucosiltransferasas/química , Pliegue de Proteína , Azúcares de Uridina Difosfato/química , Animales , Medición de Intercambio de Deuterio , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Dominios Proteicos , Selenoproteínas/química , Selenoproteínas/genética , Selenoproteínas/metabolismo , Azúcares de Uridina Difosfato/genética , Azúcares de Uridina Difosfato/metabolismo
6.
Biochim Biophys Acta Proteins Proteom ; 1865(5): 510-519, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28192204

RESUMEN

UDP-arabinopyranose mutase (UAM) is a plant enzyme which interconverts UDP-arabinopyranose (UDP-Arap; a six-membered sugar) to UDP-arabinofuranose (UDP-Araf; a five-membered sugar). Plant mutases belong to a small gene family called Reversibly Glycosylated Proteins (RGPs). So far, UAM has been identified in Oryza sativa (Rice), Arabidopsis thaliana and Hordeum vulgare (Barley). The enzyme requires divalent metal ions for catalytic activity. Here, the divalent metal ion dependency of UAMs from O. sativa (rice) and A. thaliana have been studied using HPLC-based kinetic assays. It was determined that UAM from these species had the highest relative activity in a range of 40-80µM Mn2+. Excess Mn2+ ion concentration decreased the enzyme activity. This trend was observed when other divalent metal ions were used to test activity. To gain a perspective of the role played by the metal ion in activity, an ab initio structural model was generated based on the UAM amino acid sequence and a potential metal binding region was identified. Based on our results, we propose that the probable role of the metal in UAM is stabilizing the diphosphate of the substrate, UDP-Arap.


Asunto(s)
Arabidopsis/enzimología , Transferasas Intramoleculares/química , Oryza/enzimología , Azúcares de Uridina Difosfato/química , Sitios de Unión , Catálisis , Pared Celular/enzimología , Regulación de la Expresión Génica de las Plantas , Transferasas Intramoleculares/genética , Transferasas Intramoleculares/metabolismo , Iones/química , Cinética , Metales/química , Unión Proteica , Azúcares de Uridina Difosfato/metabolismo
7.
J Org Chem ; 82(4): 2243-2248, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28128958

RESUMEN

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


Asunto(s)
Glicosaminoglicanos/biosíntesis , Nucleotidiltransferasas/metabolismo , Biocatálisis , Conformación de Carbohidratos , Glicosaminoglicanos/química , Nucleotidiltransferasas/química , Azúcares de Uridina Difosfato/biosíntesis , Azúcares de Uridina Difosfato/química
8.
Glycobiology ; 26(10): 1059-1071, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27233805

RESUMEN

P-selectin glycoprotein ligand-1 (PSGL-1, CD162) is a cell-surface glycoprotein that is expressed, either constitutively or inducibly, on all myeloid and lymphoid cell lineages. PSGL-1 is implicated in cell-cell interactions between platelets, leukocytes and endothelial cells, and a key mediator of inflammatory cell recruitment and transmigration into tissues. Here, we have investigated the effects of the ß-1,4-galactosyltransferase inhibitor 5-(5-formylthien-2-yl) UDP-Gal (5-FT UDP-Gal, compound 1: ) and two close derivatives on the cell surface levels of PSGL-1 on human peripheral blood mononuclear cells (hPBMCs). PSGL-1 levels were studied both under basal conditions, and upon stimulation of hPBMCs with interleukin-1ß (IL-1ß). Between 1 and 24 hours after IL-1ß stimulation, we observed initial PSGL-1 shedding, followed by an increase in PSGL-1 levels on the cell surface, with a maximal window between IL-1ß-induced and basal levels after 72 h. All three inhibitors reduce PSGL-1 levels on IL-1ß-stimulated cells in a concentration-dependent manner, but show no such effect in resting cells. Compound 1: also affects the cell surface levels of adhesion molecule CD11b in IL-1ß-stimulated hPBMCs, but not of glycoproteins CD14 and CCR2. This activity profile may be linked to the inhibition of global Sialyl Lewis presentation on hPBMCs by compound 1: , which we have also observed. Although this mechanistic explanation remains hypothetical at present, our results show, for the first time, that small molecules can discriminate between IL-1ß-induced and basal levels of cell surface PSGL-1. These findings open new avenues for intervention with PSGL-1 presentation on the cell surface of primed hPBMCs and may have implications for anti-inflammatory drug development.


Asunto(s)
Interleucina-1beta/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Glicoproteínas de Membrana/antagonistas & inhibidores , Azúcares de Uridina Difosfato/farmacología , Conformación de Carbohidratos , Relación Dosis-Respuesta a Droga , Humanos , Leucocitos Mononucleares/metabolismo , Glicoproteínas de Membrana/metabolismo , Relación Estructura-Actividad , Azúcares de Uridina Difosfato/química
9.
Arch Biochem Biophys ; 597: 21-9, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27036853

RESUMEN

The storage polymer glycogen normally contains small amounts of covalently attached phosphate as phosphomonoesters at C2, C3 and C6 atoms of glucose residues. In the absence of the laforin phosphatase, as in the rare childhood epilepsy Lafora disease, the phosphorylation level is elevated and is associated with abnormal glycogen structure that contributes to the pathology. Laforin therefore likely functions in vivo as a glycogen phosphatase. The mechanism of glycogen phosphorylation is less well-understood. We have reported that glycogen synthase incorporates phosphate into glycogen via a rare side reaction in which glucose-phosphate rather than glucose is transferred to a growing polyglucose chain (Tagliabracci et al. (2011) Cell Metab13, 274-282). We proposed a mechanism to account for phosphorylation at C2 and possibly at C3. Our results have since been challenged (Nitschke et al. (2013) Cell Metab17, 756-767). Here we extend the evidence supporting our conclusion, validating the assay used for the detection of glycogen phosphorylation, measurement of the transfer of (32)P from [ß-(32)P]UDP-glucose to glycogen by glycogen synthase. The (32)P associated with the glycogen fraction was stable to ethanol precipitation, SDS-PAGE and gel filtration on Sephadex G50. The (32)P-signal was not affected by inclusion of excess unlabeled UDP before analysis or by treatment with a UDPase, arguing against the signal being due to contaminating [ß-(32)P]UDP generated in the reaction. Furthermore, [(32)P]UDP did not bind non-covalently to glycogen. The (32)P associated with glycogen was released by laforin treatment, suggesting that it was present as a phosphomonoester. The conclusion is that glycogen synthase can mediate the introduction of phosphate into glycogen, thereby providing a possible mechanism for C2, and perhaps C3, phosphorylation.


Asunto(s)
Glucógeno Sintasa/química , Glucógeno/química , Fosfatos/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Glucógeno/biosíntesis , Glucógeno Sintasa/metabolismo , Humanos , Fosfatos/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Azúcares de Uridina Difosfato/química , Azúcares de Uridina Difosfato/metabolismo
10.
Plant Cell Rep ; 35(11): 2403-2421, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27591771

RESUMEN

KEY MESSAGE: The present study first identified the involvement of OcUAXS2 and OcUXS1-3 in anticancer polysaccharides biosynthesis in O. caudatum. UDP-xylose synthase (UXS) and UDP-D-apiose/UDP-D-xylose synthase (UAXS), both capable of converting UDP-D-glucuronic acid to UDP-D-xylose, are believed to transfer xylosyl residue to anticancer polysaccharides biosynthesis in Ornithogalum caudatum Ait. However, the cDNA isolation and functional characterization of genes encoding the two enzymes from O. caudatum has never been documented. Previously, the transcriptome sequencing of O. caudatum was performed in our laboratory. In this study, a total of six and two unigenes encoding UXS and UAXS were first retrieved based on RNA-Seq data. The eight putative genes were then successfully isolated from transcriptome of O. caudatum by reverse transcription polymerase chain reaction (RT-PCR). Phylogenetic analysis revealed the six putative UXS isoforms can be classified into three types, one soluble and two distinct putative membrane-bound. Moreover, the two UAXS isoenzymes were predicted to be soluble forms. Subsequently, these candidate cDNAs were characterized to be bona fide genes by functional expression in Escherichia coli individually. Although UXS and UAXS catalyzed the same reaction, their biochemical properties varied significantly. It is worth noting that a ratio switch of UDP-D-xylose/UDP-D-apiose for UAXS was established, which is assumed to be helpful for its biotechnological application. Furthermore, a series of mutants were generated to test the function of NAD+ binding motif GxxGxxG. Most importantly, the present study determined the involvement of OcUAXS2 and OcUXS1-3 in xylose-containing polysaccharides biosynthesis in O. caudatum. These data provide a comprehensive knowledge for UXS and UAXS families in plants.


Asunto(s)
Carboxiliasas/genética , Genes de Plantas , Familia de Multigenes , Ornithogalum/enzimología , Ornithogalum/genética , Transcriptoma/genética , Azúcares de Uridina Difosfato/metabolismo , Uridina Difosfato Xilosa/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Compuestos de Amonio/farmacología , Biocatálisis/efectos de los fármacos , Tampones (Química) , Calcio/farmacología , Carboxiliasas/química , Carboxiliasas/metabolismo , Cromatografía Líquida de Alta Presión , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Concentración de Iones de Hidrógeno , Cinética , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Ornithogalum/efectos de los fármacos , Espectroscopía de Protones por Resonancia Magnética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Temperatura , Transcriptoma/efectos de los fármacos , Azúcares de Uridina Difosfato/química , Uridina Difosfato Xilosa/química
11.
Proc Natl Acad Sci U S A ; 110(19): 7648-53, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610417

RESUMEN

We described the integration of the general reversibility of glycosyltransferase-catalyzed reactions, artificial glycosyl donors, and a high throughput colorimetric screen to enable the engineering of glycosyltransferases for combinatorial sugar nucleotide synthesis. The best engineered catalyst from this study, the OleD Loki variant, contained the mutations P67T/I112P/T113M/S132F/A242I compared with the OleD wild-type sequence. Evaluated against the parental sequence OleD TDP16 variant used for screening, the OleD Loki variant displayed maximum improvements in k(cat)/K(m) of >400-fold and >15-fold for formation of NDP-glucoses and UDP-sugars, respectively. This OleD Loki variant also demonstrated efficient turnover with five variant NDP acceptors and six variant 2-chloro-4-nitrophenyl glycoside donors to produce 30 distinct NDP-sugars. This study highlights a convenient strategy to rapidly optimize glycosyltransferase catalysts for the synthesis of complex sugar nucleotides and the practical synthesis of a unique set of sugar nucleotides.


Asunto(s)
Glicosiltransferasas/química , Azúcares de Nucleósido Difosfato/química , Ingeniería de Proteínas/métodos , Azúcares de Uridina Difosfato/química , Catálisis , Variación Genética , Glicómica/métodos , Glicosiltransferasas/genética , Ensayos Analíticos de Alto Rendimiento , Conformación Molecular , Mutación , Nitrofenoles/química , Recombinación Genética
12.
Drug Metab Rev ; 47(3): 335-45, 2015 08.
Artículo en Inglés | MEDLINE | ID: mdl-26289097

RESUMEN

Enzymes of the human uridine diphosphate (UDP)-glycosyltransferase (UGT) superfamily typically catalyze the covalent addition of a sugar from UDP-sugar cofactors to relatively small lipophilic compounds. The sugar conjugates are often biologically less active with improved water-solubility, facilitating more effective elimination from the body. Experimental data indicate that UGT proteins exhibit differing selectivities toward various UDP-sugars. Although, three-dimensional (3D) structures of UGT proteins are required to provide insights into the UDP-sugar selectivities observed for the various UGT proteins, there are currently, no experimental structures available for human UGTs bound to UDP-sugar(s). Thus, the absence of 3D structures poses a major challenge for analyzing UDP-sugar selectivity at an atomic level. In this commentary, we highlight the application of comparative homology modeling for understanding the UDP-sugar selectivities of UGT proteins. Homology models of the C-terminal (CT) domain indicate a highly conserved structural fold across the UGT family with backbone root mean-squared deviations (rmsds) between 0.066 and 0.079 Å with respect to the UGT2B7-CT X-ray crystal structure. The models show that four residues in the terminal portion of the CT signature sequence play an important role in UDP-sugar selectivity. The N-terminal domain is less likely to be associated with UDP-sugar selectivity, although, a conserved residue, Arg-259 (UGT2B7 numbering) in the UGT 1 and 2 families may influence UDP-sugar selectivity. Overall, the models demonstrate excellent agreement with experimental observations in predicting the key residues that influence the selectivity of UDP-sugar binding.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas de Plantas/metabolismo , Azúcares de Uridina Difosfato/metabolismo , Conformación de Carbohidratos , Glucuronosiltransferasa/química , Humanos , Proteínas de Plantas/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Especificidad por Sustrato , Azúcares de Uridina Difosfato/química
13.
Biochemistry ; 53(4): 796-805, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24460375

RESUMEN

Cationic Antimicrobial Peptides (CAMPs) represent a first line of defense against bacterial colonization. When fighting Gram-negative bacteria, CAMPs initially interact electrostatically with the negatively charged phosphate groups in lipid A and are thought to kill bacteria by disrupting their membrane integrity. However, many human pathogens, including Salmonella and Pseudomonas , have evolved lipid A modification mechanisms that result in resistance to CAMPs and related antibiotics such as Colistin. The addition of 4-amino-4-deoxy-l-Arabinose (Ara4N) to a phosphate group in lipid A is one such modification, frequently found in Pseudomonas isolated from cystic fibrosis patients. The pathway for biosynthesis of Ara4N-lipid A requires conversion of UDP-Glucuronic acid into UDP-Ara4N and subsequent transfer of the amino-sugar to lipid A. ArnB is a pyridoxal-phosphate (PLP) dependent transaminase that catalyzes a crucial step in the pathway: synthesis of UDP-Ara4N from UDP-4-keto-pentose. Here we present the 2.3 Å resolution crystal structure of an active site mutant of ArnB (K188A) in complex with the reaction intermediate aldimine formed by UDP-Ara4N and PLP. The sugar-nucleotide binding site is in a cleft between the subunits of the ArnB dimer with the uracil buried at the interface and the UDP ribose and phosphate groups exposed to the solvent. The Ara4N moiety is found in the (4)C1 conformation and its positioning, stabilized by interactions with both the protein and cofactor, is compatible with catalysis. The structure suggests strategies for the development of specific inhibitors that may prove useful in the treatment of resistant bacteria such as Pseudomonas found in cystic fibrosis patients.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/química , Farmacorresistencia Bacteriana , Polimixinas/farmacología , Salmonella typhimurium/enzimología , Transaminasas/química , Dominio Catalítico , Cristalografía por Rayos X , Modelos Moleculares , Mutación , Conformación Proteica , Fosfato de Piridoxal/química , Salmonella typhimurium/genética , Especificidad por Sustrato , Azúcares de Uridina Difosfato/química
14.
J Biol Chem ; 288(45): 32248-32260, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24064219

RESUMEN

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


Asunto(s)
Acetilglucosamina/análogos & derivados , Acetiltransferasas/química , Acinetobacter baumannii/enzimología , Proteínas Bacterianas/química , Neisseria gonorrhoeae/enzimología , Azúcares de Uridina Difosfato/biosíntesis , Acetilcoenzima A , Acetilglucosamina/biosíntesis , Acetilglucosamina/química , Acetilglucosamina/genética , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Acinetobacter baumannii/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Glicosilación , Mutación , Neisseria gonorrhoeae/genética , Estructura Terciaria de Proteína , Azúcares de Uridina Difosfato/química , Azúcares de Uridina Difosfato/genética
15.
J Biol Chem ; 288(15): 10578-87, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23460642

RESUMEN

Oligosaccharyltransferases (OTases) are enzymes that catalyze the transfer of an oligosaccharide from a lipid carrier to an acceptor molecule, commonly a protein. OTases are classified as N-OTases and O-OTases, depending on the nature of the glycosylation reaction. The N-OTases catalyze the glycan transfer to amide groups in asparagines in a reaction named N-linked glycosylation. The O-OTases are responsible for protein O-linked glycosylation, which involves the attachment of glycans to hydroxyl groups of serine or threonine residues. These enzymes exhibit a relaxed specificity and are able to transfer a variety of glycan structures to different protein acceptors. This property confers OTases with great biotechnological potential as these enzymes can produce glycoconjugates relevant to the pharmaceutical industry. Furthermore, OTases are thought to be involved in pathogenesis mechanisms. Several aspects of the functionality of OTases are not fully understood. In this work, we developed a novel approach to perform kinetic studies on PglL, the O-OTase from Neisseria meningitidis. We investigated the importance of the acyl moiety of the lipid glycan donor substrate on the functionality of PglL by testing the efficiency of glycosylation reactions using synthetic substrates carrying the same glycan structure but different acyl moieties. We found that PglL can function with many lipids as glycan donors, although the length and the conformation of the lipid moiety significantly influenced the catalytic efficiency. Interestingly, PglL was also able to transfer a monosaccharide employing its nucleotide-activated form, acting as a Leloir glycosyltransferase. These results provide new insights on the function and the evolution of oligosaccharyltransferases.


Asunto(s)
Hexosiltransferasas/química , Lípidos de la Membrana/química , Proteínas de la Membrana/química , Neisseria meningitidis/enzimología , Azúcares de Uridina Difosfato/química , Glicosilación , Hexosiltransferasas/genética , Hexosiltransferasas/metabolismo , Metabolismo de los Lípidos/fisiología , Lípidos de la Membrana/genética , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neisseria meningitidis/genética , Especificidad por Sustrato/fisiología , Azúcares de Uridina Difosfato/genética , Azúcares de Uridina Difosfato/metabolismo
16.
Bioorg Med Chem Lett ; 23(13): 3764-8, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707255

RESUMEN

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


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Nucleotidiltransferasas/metabolismo , Azúcares de Uridina Difosfato/biosíntesis , Arabidopsis/metabolismo , Conformación de Carbohidratos , Galactoquinasa/metabolismo , Pirofosfatasas/metabolismo , Streptococcus pneumoniae/enzimología , Azúcares de Uridina Difosfato/química
17.
J Biol Chem ; 286(3): 1777-85, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21084307

RESUMEN

Heparosan synthase catalyzes the polymerization of heparosan (-4GlcUAß1-4GlcNAcα1-)(n) by transferring alternatively the monosaccharide units from UDP-GlcUA and UDP-GlcNAc to an acceptor molecule. Details on the heparosan chain initiation by Pasteurella multocida heparosan synthase PmHS2 and its influence on the polymerization process have not been reported yet. By site-directed mutagenesis of PmHS2, the single action transferases PmHS2-GlcUA(+) and PmHS2-GlcNAc(+) were obtained. When incubated together in the standard polymerization conditions, the PmHS2-GlcUA(+)/PmHS2-GlcNAc(+) showed comparable polymerization properties as determined for PmHS2. We investigated the first step occurring in heparosan chain initiation by the use of the single action transferases and by studying the PmHS2 polymerization process in the presence of heparosan templates and various UDP-sugar concentrations. We observed that PmHS2 favored the initiation of the heparosan chains when incubated in the presence of an excess of UDP-GlcNAc. It resulted in a higher number of heparosan chains with a lower average molecular weight or in the synthesis of two distinct groups of heparosan chain length, in the absence or in the presence of heparosan templates, respectively. These data suggest that PmHS2 transfers GlcUA from UDP-GlcUA moiety to a UDP-GlcNAc acceptor molecule to initiate the heparosan polymerization; as a consequence, not only the UDP-sugar concentration but also the amount of each UDP-sugar is influencing the PmHS2 polymerization process. In addition, it was shown that PmHS2 hydrolyzes the UDP-sugars, UDP-GlcUA being more degraded than UDP-GlcNAc. However, PmHS2 incubated in the presence of both UDP-sugars favors the synthesis of heparosan polymers over the hydrolysis of UDP-sugars.


Asunto(s)
Proteínas Bacterianas/química , Glicosiltransferasas/química , Hidrolasas/química , Pasteurella multocida/enzimología , Azúcares de Uridina Difosfato/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Hidrolasas/genética , Hidrolasas/metabolismo , Hidrólisis , Pasteurella multocida/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Azúcares de Uridina Difosfato/genética , Azúcares de Uridina Difosfato/metabolismo
18.
J Am Chem Soc ; 134(34): 13946-9, 2012 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-22830643

RESUMEN

UDP-D-apiose/UDP-D-xylose synthase (AXS) catalyzes the conversion of UDP-D-glucuronic acid to UDP-D-apiose and UDP-D-xylose. An acetyl-protected phosphonate analogue of UDP-D-apiose was synthesized and used in an in situ HPLC assay to demonstrate for the first time the ability of AXS to interconvert the two reaction products. Density functional theory calculations provided insight into the energetics of this process and the apparent inability of AXS to catalyze the conversion of UDP-D-xylose to UDP-D-apiose. The data suggest that this observation is unlikely to be due to an unfavorable equilibrium but rather results from substrate inhibition by the most stable chair conformation of UDP-D-xylose. The detection of xylose cyclic phosphonate as the turnover product reveals significant new details about the AXS-catalyzed reaction and supports the proposed retroaldol-aldol mechanism of catalysis.


Asunto(s)
Carboxiliasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Organofosfonatos/metabolismo , Azúcares de Uridina Difosfato/metabolismo , Uridina Difosfato Xilosa/metabolismo , Escherichia coli/metabolismo , Cinética , Modelos Moleculares , Organofosfonatos/química , Teoría Cuántica , Azúcares de Uridina Difosfato/química , Uridina Difosfato Xilosa/química
19.
Glycoconj J ; 29(7): 491-502, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22711644

RESUMEN

The α-1,3-glucosyltransferase WaaG is involved in the synthesis of the core region of lipopolysaccharides in E. coli. A fragment-based screening for inhibitors of the WaaG glycosyltrasferase donor site has been performed using NMR spectroscopy. Docking simulations were performed for three of the compounds of the fragment library that had shown binding activity towards WaaG and yielded 3D models for the respective complexes. The three ligands share a hetero-bicyclic ring system as a common structural motif and they compete with UDP-Glc for binding. Interestingly, one of the compounds promoted binding of uridine to WaaG, as seen from STD NMR titrations, suggesting a different binding mode for this ligand. We propose these compounds as scaffolds for the design of selective high-affinity inhibitors of WaaG. Binding of natural substrates, enzymatic activity and donor substrate selectivity were also investigated by NMR spectroscopy. Molecular dynamics simulations of WaaG were carried out with and without bound UDP and revealed structural changes compared to the crystal structure and also variations in flexibility for some amino acid residues between the two WaaG systems studied.


Asunto(s)
Inhibidores Enzimáticos/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Glucosiltransferasas/química , Azúcares de Uridina Difosfato/química , Inhibidores Enzimáticos/metabolismo , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/metabolismo , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/metabolismo , Glucolípidos/biosíntesis , Ligandos , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Azúcares de Uridina Difosfato/metabolismo
20.
PLoS Comput Biol ; 7(10): e1002224, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22046111

RESUMEN

Combinatorial sensor arrays, such as the olfactory system, can detect a large number of analytes using a relatively small number of receptors. However, the complex pattern of receptor responses to even a single analyte, coupled with the non-linearity of responses to mixtures of analytes, makes quantitative prediction of compound concentrations in a mixture a challenging task. Here we develop a physical model that explicitly takes receptor-ligand interactions into account, and apply it to infer concentrations of highly related sugar nucleotides from the output of four engineered G-protein-coupled receptors. We also derive design principles that enable accurate mixture discrimination with cross-specific sensor arrays. The optimal sensor parameters exhibit relatively weak dependence on component concentrations, making a single designed array useful for analyzing a sizable range of mixtures. The maximum number of mixture components that can be successfully discriminated is twice the number of sensors in the array. Finally, antagonistic receptor responses, well-known to play an important role in natural olfactory systems, prove to be essential for the accurate prediction of component concentrations.


Asunto(s)
Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/métodos , Mezclas Complejas/análisis , Modelos Biológicos , Receptores Acoplados a Proteínas G/metabolismo , Azúcares de Uridina Difosfato/análisis , Algoritmos , Teorema de Bayes , Mezclas Complejas/química , Biología Computacional , Simulación por Computador , Humanos , Unión Proteica , Receptores Acoplados a Proteínas G/genética , Olfato/fisiología , Termodinámica , Azúcares de Uridina Difosfato/química , Azúcares de Uridina Difosfato/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda