Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Acta Biochim Biophys Sin (Shanghai) ; 45(9): 720-5, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23774504

RESUMO

Human GDP-l-fucose synthase, also known as FX protein, synthesizes GDP-l-fucose from its substrate GDP-4-keto-6-deoxy-d-mannose. The reaction involves epimerization at both C-3 and C-5 followed by an NADPH-dependent reduction of the carbonyl at C-4. In this paper, the first crystal structure of human FX protein was determined at 2.37 Å resolution. The asymmetric unit of the crystal structure contains four molecules which form two homodimers. Each molecule consists of two domains, a Rossmann-fold NADPH-binding motif and a carboxyl terminal domain. Compared with the Escherichia coli GDP-l-fucose synthase, the overall structures of these two enzymes have four major differences. There are four loops in the structure of human FX protein corresponding to two α-helices and two ß-sheets in that of the E. coli enzyme. Besides, there are seven different amino acid residues binding with NAPDH comparing human FX protein with that from E. coli. The structure of human FX reveals the key catalytic residues and could be useful for the design of drugs for the treatment of inflammation, auto-immune diseases, and possibly certain types of cancer.


Assuntos
Carboidratos Epimerases/química , Cetona Oxirredutases/química , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Sítios de Ligação , Biocatálise , Carboidratos Epimerases/genética , Carboidratos Epimerases/metabolismo , Cristalografia por Raios X , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Guanosina Difosfato Manose/análogos & derivados , Guanosina Difosfato Manose/metabolismo , Açúcares de Guanosina Difosfato/metabolismo , Humanos , Cetona Oxirredutases/genética , Cetona Oxirredutases/metabolismo , Modelos Moleculares , NADP/química , NADP/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
2.
Biochem J ; 439(2): 235-48, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21711244

RESUMO

The capsule of Campylobacter jejuni strain 81-176 comprises the unusual 6-deoxy-α-D-altro-heptose, whose biosynthesis and function are not known. In the present study, we characterized enzymes of the capsular cluster, WcbK and WcaG, to determine their role in 6-deoxy-altro-heptose synthesis. These enzymes are similar to the Yersinia pseudotuberculosis GDP-manno-heptose dehydratase/reductase DmhA/DmhB that we characterized previously. Capillary electrophoresis and MS analyses showed that WcbK is a GDP-manno-heptose dehydratase whose product can be reduced by WcaG, and that WcbK/WcaG can use the substrate GDP-mannose, although with lower efficiency than heptose. Comparison of kinetic parameters for WcbK and DmhA indicated that the relaxed substrate specificity of WcbK comes at the expense of catalytic performance on GDP-manno-heptose. Moreover, although WcbK/WcaG and DmhA/DmhB are involved in altro- versus manno-heptose synthesis respectively, the enzymes can be used interchangeably in mixed reactions. NMR spectroscopy analyses indicated conservation of the sugar manno configuration during catalysis by WcbK/WcaG. Therefore additional capsular enzymes may perform the C3 epimerization necessary to generate 6-deoxy-altro-heptose. Finally, a conserved residue (Thr(187) in WcbK) potentially involved in substrate specificity was identified by structural modelling of mannose and heptose dehydratases. Site-directed mutagenesis and kinetic analyses demonstrated its importance for enzymatic activity on heptose and mannose substrates.


Assuntos
Campylobacter jejuni/metabolismo , Guanosina Difosfato Manose/análogos & derivados , Hidroliases/metabolismo , Oxirredutases/metabolismo , Sequência de Bases , Campylobacter jejuni/enzimologia , Clonagem Molecular , Primers do DNA , Guanosina Difosfato Manose/biossíntese , Hidroliases/genética , Hidroliases/isolamento & purificação , Espectrometria de Massas , Ressonância Magnética Nuclear Biomolecular , Oxirredutases/genética , Oxirredutases/isolamento & purificação
3.
Biochemistry ; 48(23): 5246-53, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19402712

RESUMO

L-colitose and d-perosamine are unusual sugars found in the O-antigens of some Gram-negative bacteria such as Escherichia coli, Vibrio cholerae, and Salmonella enterica, among others. The biosynthetic pathways for these two sugars begin with the formation of GDP-mannose from d-mannose 1-phosphate and GTP followed by the subsequent dehydration and oxidation of GDP-mannose to yield GDP-4-keto-6-deoxymannose. Following the production of GDP-4-keto-6-deoxymannose, the two pathways diverge. In the case of GDP-perosamine biosynthesis, the next step involves an amination reaction at the C-4' position of the sugar, whereas in GDP-colitose production, the 3'-hydroxyl group is removed. The enzymes catalyzing these reactions are GDP-perosamine synthase and GDP-4-keto-6-deoxymannose-3-dehydratase (ColD), respectively. Both of these enzymes are pyridoxal 5'-phosphate (PLP) dependent, and their three-dimensional structures place them into the well-characterized aspartate aminotransferase superfamily. A comparison of the active site architecture of ColD from E. coli (strain 5a, type O55:H7) to that of GDP-perosamine synthase from Caulobacter crescentus CB15 suggested that only two mutations would be required to convert ColD into an aminotransferase. Here we present a combined structural and functional analysis of the ColD S187N/H188K mutant protein that, indeed, has been converted from a sugar dehydratase into an aminotransferase.


Assuntos
Proteínas de Bactérias/química , Proteínas de Escherichia coli/química , Hidroliases/química , Mutação , Transaminases/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Carboidratos Epimerases/química , Carboidratos Epimerases/metabolismo , Caulobacter crescentus/enzimologia , Caulobacter crescentus/metabolismo , Cristalografia por Raios X , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Guanosina Difosfato Manose/análogos & derivados , Guanosina Difosfato Manose/química , Guanosina Difosfato Manose/metabolismo , Hidroliases/genética , Hidroliases/metabolismo , Cinética , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato , Transaminases/metabolismo
4.
Chem Commun (Camb) ; (36): 4321-3, 2008 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-18802557

RESUMO

GDP-2-, 3-, 4- or 6-azidomannoses can be successfully prepared from the corresponding azidomannose-1-phosphates and GTP using the enzyme GDP-Mannosepyrophosphorylase (GDP-ManPP) from Salmonella enterica and may serve as useful probes for mannosyltransferase activity.


Assuntos
Guanosina Difosfato Manose/análogos & derivados , Guanosina Difosfato Manose/síntese química , Manosiltransferases/química , Sondas Moleculares/química , Configuração de Carboidratos , Ativação Enzimática , Guanosina Difosfato Manose/química , Guanosina Trifosfato/química , Manosiltransferases/análise , Proteínas Recombinantes/química , Salmonella enterica/enzimologia
5.
Protein Sci ; 15(9): 2093-106, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16943443

RESUMO

L-colitose is a 3,6-dideoxysugar found in the O-antigens of some Gram-negative bacteria such as Escherichia coli and in marine bacteria such as Pseudoalteromonas tetraodonis. The focus of this investigation, GDP-4-keto-6-deoxy-D-mannose-3-dehydratase, catalyzes the third step in colitose production, which is the removal of the hydroxyl group at C3' of GDP-4-keto-6-deoxymannose. It is an especially intriguing PLP-dependent enzyme in that it acts as both a transaminase and a dehydratase. Here we present the first X-ray structure of this enzyme isolated from E. coli Strain 5a, type O55:H7. The two subunits of the protein form a tight dimer with a buried surface area of approximately 5000 A2. This is a characteristic feature of the aspartate aminotransferase superfamily. Although the PLP-binding pocket is formed primarily by one subunit, there is a loop, delineated by Phe 240 to Glu 253 in the second subunit, that completes the active site architecture. The hydrated form of PLP was observed in one of the enzyme/cofactor complexes described here. Amino acid residues involved in anchoring the cofactor to the protein include Gly 56, Ser 57, Asp 159, Glu 162, and Ser 183 from one subunit and Asn 248 from the second monomer. In the second enzyme/cofactor complex reported, a glutamate ketimine intermediate was found trapped in the active site. Taken together, these two structures, along with previously reported biochemical data, support the role of His 188 as the active site base required for catalysis.


Assuntos
Proteínas de Escherichia coli/química , Guanosina Difosfato Manose/análogos & derivados , Hidroliases/química , Modelos Químicos , Estrutura Terciária de Proteína , Complexo Vitamínico B/química , Sítios de Ligação , Coenzimas/química , Cristalografia por Raios X , Guanosina Difosfato Manose/química , Ligação de Hidrogênio , Modelos Moleculares , Especificidade por Substrato
6.
FEBS J ; 273(10): 2244-56, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16650000

RESUMO

The deoxyhexose sugar fucose has an important fine-tuning role in regulating the functions of glycoconjugates in disease and development in mammals. The two genetic model organisms Caenorhabditis elegans and Drosophila melanogaster also express a range of fucosylated glycans, and the nematode particularly has a number of novel forms. For the synthesis of such glycans, the formation of GDP-fucose, which is generated from GDP-mannose in three steps catalysed by two enzymes, is required. By homology we have identified and cloned cDNAs encoding these two proteins, GDP-mannose dehydratase (GMD; EC 4.2.1.47) and GDP-keto-6-deoxymannose 3,5-epimerase/4-reductase (GER or FX protein; EC 1.1.1.271), from both Caenorhabditis and Drosophila. Whereas the nematode has two genes encoding forms of GMD (gmd-1 and gmd-2) and one GER-encoding gene (ger-1), the insect has, like mammalian species, only one homologue of each (gmd and gmer). This compares to the presence of two forms of both enzymes in Arabidopsis thaliana. All corresponding cDNAs from Caenorhabditis and Drosophila, as well as the previously uncharacterized Arabidopsis GER2, were separately expressed, and the encoded proteins found to have the predicted activity. The biochemical characterization of these enzymes is complementary to strategies aimed at manipulating the expression of fucosylated glycans in these organisms.


Assuntos
Caenorhabditis elegans/enzimologia , Carboidratos Epimerases/metabolismo , Drosophila melanogaster/enzimologia , Guanosina Difosfato Fucose/metabolismo , Hidroliases/metabolismo , Sequência de Aminoácidos , Animais , Arabidopsis/enzimologia , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Carboidratos Epimerases/genética , Clonagem Molecular , DNA Complementar , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Guanosina Difosfato Fucose/química , Guanosina Difosfato Manose/análogos & derivados , Guanosina Difosfato Manose/genética , Guanosina Difosfato Manose/metabolismo , Humanos , Hidroliases/genética , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
7.
J Mol Biol ; 303(1): 77-91, 2000 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-11021971

RESUMO

GDP-4-keto-6-deoxy-d-mannose epimerase/reductase is a bifunctional enzyme responsible for the last step in the biosynthesis of GDP-l-fucose, the substrate of fucosyl transferases. Several cell-surface antigens, including the leukocyte Lewis system and cell-surface antigens in pathogenic bacteria, depend on the availability of GDP-l-fucose for their expression. Therefore, the enzyme is a potential target for therapy in pathological states depending on selectin-mediated cell-to-cell interactions. Previous crystallographic investigations have shown that GDP-4-keto-6-deoxy-d-mannose epimerase/reductase belongs to the short-chain dehydrogenase/reductase protein homology family. The enzyme active-site region is at the interface of an N-terminal NADPH-binding domain and a C-terminal domain, held to bind the substrate. The design, expression and functional characterization of seven site-specific mutant forms of GDP-4-keto-6-deoxy-d-mannose epimerase/reductase are reported here. In parallel, the crystal structures of the native holoenzyme and of three mutants (Ser107Ala, Tyr136Glu and Lys140Arg) have been investigated and refined at 1. 45-1.60 A resolution, based on synchrotron data (R-factors range between 12.6 % and 13.9 %). The refined protein models show that besides the active-site residues Ser107, Tyr136 and Lys140, whose mutations impair the overall enzymatic activity and may affect the coenzyme binding mode, side-chains capable of proton exchange, located around the expected substrate (GDP-4-keto-6-deoxy-d-mannose) binding pocket, are selectively required during the epimerization and reduction steps. Among these, Cys109 and His179 may play a primary role in proton exchange between the enzyme and the epimerization catalytic intermediates. Finally, the additional role of mutated active-site residues involved in substrate recognition and in enzyme stability has been analyzed.


Assuntos
Carboidratos Epimerases/química , Carboidratos Epimerases/metabolismo , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Guanosina Difosfato Manose/análogos & derivados , Cetona Oxirredutases , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Mutagênese Sítio-Dirigida/genética , Desidrogenase do Álcool de Açúcar/química , Desidrogenase do Álcool de Açúcar/metabolismo , Substituição de Aminoácidos/genética , Sítios de Ligação , Carboidratos Epimerases/genética , Catálise , Cromatografia em Camada Fina , Cristalografia por Raios X , Desoxiaçúcares/análise , Desoxiaçúcares/metabolismo , Estabilidade Enzimática , Fucose/análogos & derivados , Fucose/química , Fucose/metabolismo , Guanosina Difosfato Manose/química , Guanosina Difosfato Manose/metabolismo , Holoenzimas/química , Holoenzimas/genética , Holoenzimas/metabolismo , Ligação de Hidrogênio , Cinética , Modelos Moleculares , Complexos Multienzimáticos/genética , Mutação/genética , NADP/metabolismo , Conformação Proteica , Prótons , Relação Estrutura-Atividade , Especificidade por Substrato , Desidrogenase do Álcool de Açúcar/genética
8.
J Biol Chem ; 263(4): 1693-7, 1988 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-3338988

RESUMO

The first committed enzyme in GDP-L-fucose formation from GDP-D-mannose is GDP-D-mannose 4,6-dehydratase, which forms GDP-4-keto-6-deoxy-D-mannose. The uncertain enzymatic steps beyond this point were examined in this study. Assays were developed for the epimerase and reductase activities which the putative pathway would predict. A protein was isolated exhibiting homogeneity by several criteria. This single protein, which forms GDP-L-fucose from GDP-4-keto-6-deoxy-D-mannose and NADH, appears to possess both epimerase and reductase capabilities and may be termed GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase-4-reductase. Analysis on a molecular sieve column using fast protein liquid chromatography established a molecular weight of 63,100 for the native enzyme, whereas sodium dodecyl sulfate-polyacrylamide gel electrophoresis established a subunit molecular weight of 31,500.


Assuntos
Carboidratos Epimerases/metabolismo , Proteínas de Escherichia coli , Fucose/biossíntese , Guanosina Difosfato Manose/análogos & derivados , Cetona Oxirredutases , Complexos Multienzimáticos , Ramnose/análogos & derivados , Desidrogenase do Álcool de Açúcar/metabolismo , Animais , Guanosina Difosfato Fucose/metabolismo , Cinética , Peso Molecular , Ramnose/isolamento & purificação , Suínos , Glândula Tireoide/enzimologia
9.
J Biol Chem ; 268(24): 17943-50, 1993 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-7688733

RESUMO

Pig liver GDP-mannose pyrophosphorylase was purified 5,000-fold to apparent homogeneity using standard techniques. The native enzyme showed a single band on gels of about 450 kDa and two subunits of 43 and 37 kDa on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The 37-kDa (beta-) subunit had only methionine at its amino terminus and a surprisingly hydrophobic sequence: Met-Lys-Ala-Leu-Ile-Leu-Val-Gly-Gly-Tyr-Gly-Thr-Arg-Leu- Arg-Pro-Leu-Thr-Leu-Ser-Ile-Pro-Lys. The 43-kDa (alpha-) subunit was blocked at the amino terminus, but a 29-kDa CNBr fragment had the following sequence: Leu-Asp-Ala-His-Arg-His-Arg-Pro-His-Pro- Phe-Leu-Leu-. Substrate specificity studies done in the direction of formation of nucleoside triphosphate and sugar-1-P indicated that the enzyme was most effective with GDP-glucose as substrate (100%) followed by IDP-mannose (72%) and then GDP-mannose (61%). That GDP-mannose and GDP-glucose activities were indeed catalyzed by the same enzyme was indicated by the following. (i) Various studies indicated that the enzyme was homogeneous. (ii) A staining procedure for production of GTP stained the same single band on native gels when either GDP-mannose or GDP-glucose was the substrate. (iii). GDP-mannose inhibited the utilization of GDP-glucose by the enzyme, and vice versa. When 8-azido-[32P]GTP was incubated with native enzyme and exposed to UV light, both the 43-kDa and the 37-kDa subunits became labeled, although the 37-kDa subunit reacted more strongly. On the other hand, 8-azido-GDP-[32P]mannose only photolabeled the 43-kDa band. Most importantly, the purified enzyme can be utilized to produce 8-azido-[32P]GDP mannose or 8-azido-[32P]GDP glucose.


Assuntos
Marcadores de Afinidade/síntese química , Azidas/síntese química , Guanosina Difosfato Manose/análogos & derivados , Açúcares de Guanosina Difosfato/síntese química , Guanosina Trifosfato/análogos & derivados , Fígado/enzimologia , Nucleotidiltransferases/isolamento & purificação , Nucleotidiltransferases/metabolismo , Sequência de Aminoácidos , Animais , Radioisótopos de Carbono , Cromatografia , Cromatografia DEAE-Celulose , Cromatografia em Gel , Cromatografia por Troca Iônica , Durapatita , Guanosina Difosfato Manose/síntese química , Hidroxiapatitas , Substâncias Macromoleculares , Dados de Sequência Molecular , Nucleotidiltransferases/química , Radioisótopos de Fósforo , Conformação Proteica , Especificidade por Substrato , Suínos
10.
Arch Biochem Biophys ; 300(2): 694-8, 1993 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-7679567

RESUMO

The first enzyme in the formation of GDP-L-fucose from GDP-D-mannose, which forms a GDP-4-keto sugar intermediate, was purified to homogeneity from cell extracts of Klebsiella pneumoniae. During purification, the enzyme was found to be highly activated by NADP. It was proven that the pyridine nucleotide coenzyme of the enzyme was NADP, not NAD, which differs from previously accepted information. NAD had no effect on enzyme activity. The product of the enzyme reaction with NADP as coenzyme was separated from other nucleotides by high-performance liquid chromatography, and using ion spray liquid chromatography/mass spectrometry the mass was determined for the first time, as 587, which is same as the calculated mass of GDP-4-keto-6-deoxy-D-mannose.


Assuntos
Desidrogenases de Carboidrato/isolamento & purificação , Desidrogenases de Carboidrato/metabolismo , Guanosina Difosfato Manose/metabolismo , Klebsiella pneumoniae/enzimologia , NADP/metabolismo , Cromatografia DEAE-Celulose , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Guanosina Difosfato Manose/análogos & derivados , Guanosina Difosfato Manose/análise , Cinética , Espectrometria de Massas , NAD/metabolismo , Especificidade por Substrato
11.
Arch Biochem Biophys ; 362(2): 339-45, 1999 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9989944

RESUMO

The enzyme that catalyzes the formation of GDP-d-mannose from GTP and alpha-d-mannose-1-P was purified about 2300-fold to near homogeneity from the soluble fraction of Mycobacterium smegmatis. At the final stage of purification, a major protein band of 37 kDa was observed and this band was specifically labeled, and in a concentration-dependent manner, by the photoaffinity probe 8-N3-GDP[32P]-d-mannose. The purified enzyme was stable for several months when kept in the frozen state. The 37-kDa band was subjected to protein sequencing and one peptide sequence of 25 amino acids showed over 80% identity to GDP-mannose pyrophosphorylases of pig liver and Saccharomyces cerevesiae. In contrast to some other bacterial GDP-mannose pyrophosphorylases, the mycobacterial enzyme was not multifunctional and did not have phosphomannose isomerase or phosphoglucose isomerase activity. Also, in contrast to the pig liver enzyme which uses mannose-1-P or glucose-1-P plus GTP to synthesize either GDP-mannose or GDP-glucose, the mycobacterial enzyme was specific for mannose-1-P as the sugar phosphate substrate. The enzyme was also relatively specific for GTP as the nucleoside triphosphate substrate. ITP was about 18% as effective as GTP, but ATP, CTP, and UTP were inactive. The activity of the enzyme was inhibited by GDP-glucose and glucose-1-P, although neither was a substrate for this enzyme. The pH optimum for the enzyme was 8.0, and Mg2+ was the best cation with optimum activity at about 5 mM. This enzyme is important for producing the activated form of mannose for formation of cell wall lipoarabinomannan and various mannose-containing glycolipids and polysaccharides.


Assuntos
Mycobacterium smegmatis/enzimologia , Nucleotidiltransferases/metabolismo , Sequência de Aminoácidos , Animais , Estabilidade Enzimática , Glucofosfatos/farmacologia , Guanosina Difosfato Manose/análogos & derivados , Guanosina Difosfato Manose/metabolismo , Açúcares de Guanosina Difosfato/farmacologia , Guanosina Trifosfato/metabolismo , Concentração de Íons de Hidrogênio , Inosina Trifosfato/metabolismo , Cinética , Magnésio/farmacologia , Manose-6-Fosfato Isomerase/metabolismo , Manosefosfatos/metabolismo , Dados de Sequência Molecular , Peso Molecular , Nucleotidiltransferases/antagonistas & inibidores , Nucleotidiltransferases/química , Nucleotidiltransferases/isolamento & purificação , Marcadores de Fotoafinidade , Análise de Sequência , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
12.
Biochem J ; 248(2): 523-31, 1987 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-2449168

RESUMO

The effects of the guanosine diphosphate esters of 4-deoxy-4-fluoro-D-mannose (GDP-4FMan) and 4-deoxy-D-mannose (GDP-4dMan) on reactions of the dolichol pathway in chick-embryo cell microsomal membranes were investigated by studies with chick-embryo cell microsomal membranes in vitro and in baby-hamster kidney (BHK) cells in vivo. Each nucleotide sugar analogue inhibited lipid-linked oligosaccharide biosynthesis in a concentration-dependent manner. GDP-4FMan blocked in vitro the addition of mannose to Dol-PP-(GlcNAc)2Man from GDP-Man (where Dol represents dolichol), but did not interfere with the formation of Dol-P-Man, Dol-P-Glc and Dol-PP-(GlcNAc)2. Although GDP-4FMan and Dol-P-4FMan were identified as metabolites of 4FMan in BHK cells labelled with [1-14C]4FMan, GDP-4FMan was a very poor substrate for GDP-Man:Dol-P mannosyltransferase and Dol-P-4FMan could only be synthesized in vitro if the chick-embryo cell membranes were primed with Dol-P. It therefore appears that the inhibition of lipid-linked oligosaccharide formation in BHK cells treated with 4FMan [Grier & Rasmussen (1984) J. Biol. Chem. 259, 1027-1030] is due primarily to a blockage in the formation of Dol-PP-(GlcNAc)2Man2 by GDP-4FMan. In contrast, GDP-4dMan was a substrate for those mannosyltransferases that catalyse the transfer of the first five mannose residues to Dol-PP-(GlcNAc)2. In addition, GDP-4dMan was a substrate for GDP-Man:Dol-P mannosyltransferase, which catalysed the formation of Dol-P-4dMan. As a consequence of this, the formation of Dol-P-Man, Dol-P-Glc and Dol-PP-(GlcNAc)2 may be inhibited through competition for Dol-P. In BHK cells treated with 10 mM-4dMan, Dol-PP-(GlcNAc)2Man9 was the major lipid-linked oligosaccharide detected. Nearly normal extents of protein glycosylation were observed, but very little processing to complex oligosaccharides occurred, and the high-mannose structures were smaller than in untreated cells.


Assuntos
Nucleotídeos de Guanina/farmacologia , Guanosina Difosfato Manose/farmacologia , Guanosina Difosfato/farmacologia , Lipopolissacarídeos/biossíntese , Açúcares de Nucleosídeo Difosfato/farmacologia , Proteínas/metabolismo , Ramnose/análogos & derivados , Animais , Embrião de Galinha , Cromatografia em Gel , Cromatografia em Camada Fina , Cricetinae , Dolicol Monofosfato Manose/biossíntese , Glicosilação , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato Manose/análogos & derivados , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Microssomos/efeitos dos fármacos , Microssomos/metabolismo , Monossacarídeos de Poli-Isoprenil Fosfato/biossíntese , Ramnose/farmacologia
13.
Glycobiology ; 10(9): 875-81, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10988249

RESUMO

The 6-deoxyhexose L-fucose is an important and characteristic element in glycoconjugates of bacteria (e.g., lipopolysaccharides), plants (e.g., xyloglucans) and animals (e.g., glycolipids, glycoproteins, and oligosaccharides). The biosynthetic pathway of GDP-L-fucose starts with a dehydration of GDP-D-mannose catalyzed by GDP-D-mannose 4,6-dehydratase (Gmd) creating GDP-4-keto-6-deoxymannose which is subsequently converted by the GDP-4-keto-6-deoxy-D-mannose 3,5-epimerase-4-reductase (WcaG; GDP-beta-L-fucose synthetase) to GDP-beta-L-fucose. Both biosynthetic genes gmd and wcaG were cloned from Escherichia coli K12 and the enzymes overexpressed under control of the T7 promoter in the expression vectors pET11a and pET16b, yielding both native and N-terminal His-tag fusion proteins, respectively. The activities of the Gmd and WcaG were analyzed. The enzymatic conversion from GDP-D-mannose to GDP-beta-L-fucose was optimized and the final product was purified. The formation of GDP-beta-L-fucose by the recombinant enzymes was verified by HPLC and NMR analyses. The His-tag fusion variants of the Gmd and WcaG proteins were purified to near homogeneity. The His-tag Gmd recombinant enzyme was inactive, whereas His-tag WcaG showed very similar enzymatic properties relative to the native GDP-beta-L-fucose synthetase. With the purified His-tag WcaG Km and Vmax values, respectively, of 40 microM and 23 nkat/mg protein for the substrate GDP-4-keto-6-deoxy-D-mannose and of 21 microM and 10 nkat/mg protein for the cosubstrate NADPH were obtained; a pH optimum of 7.5 was determined and the enzyme was stimulated to equal extend by the divalent cations Mg2+ and Ca2+. The Gmd enzyme showed a strong feedback inhibition by GDP-beta-L-fucose.


Assuntos
Proteínas de Escherichia coli , Escherichia coli/enzimologia , Fucose/análogos & derivados , Fucose/biossíntese , Glicosídeos/biossíntese , Guanosina Difosfato Fucose/síntese química , Carboidratos Epimerases/genética , Carboidratos Epimerases/isolamento & purificação , Carboidratos Epimerases/metabolismo , Cátions Bivalentes/farmacologia , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , Escherichia coli/genética , Fucose/isolamento & purificação , Guanosina Difosfato Manose/análogos & derivados , Guanosina Difosfato Manose/metabolismo , Concentração de Íons de Hidrogênio , Cetona Oxirredutases/genética , Cetona Oxirredutases/isolamento & purificação , Cetona Oxirredutases/metabolismo , Cinética , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/isolamento & purificação , Complexos Multienzimáticos/metabolismo , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Desidrogenase do Álcool de Açúcar/genética , Desidrogenase do Álcool de Açúcar/isolamento & purificação , Desidrogenase do Álcool de Açúcar/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA