Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biotechnol Prog ; 40(3): e3438, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38415431

RESUMO

Fucosylation is an important quality attribute for therapeutic antibodies. Afucosylated antibodies exhibit higher therapeutic efficacies than their fucosylated counterparts through antibody-dependent cellular cytotoxicity (ADCC) mechanism. Since higher potency is beneficial in reducing dose or duration of the treatment, afucosylated antibodies have attracted a great deal of interest in biotherapeutics development. In this study, novel small molecules GDP-D-Rhamnose and its derivatives (Ac-GDP-D-Rhamnose and rhamnose sodium phosphate) were synthesized to inhibit the enzyme in the GDP-fucose synthesis pathway. Addition of these compounds into cell culture increased antibody afucosylation levels in a dose-dependent manner and had no significant impact on other protein quality attributes. A novel and effective mechanism to generate afucosylated antibody is demonstrated for biologics discovery, analytical method development, process development, and other applications.


Assuntos
Cricetulus , Fucose , Fucose/metabolismo , Fucose/química , Animais , Células CHO , Glicosilação , Anticorpos Monoclonais/química , Anticorpos Monoclonais/biossíntese , Ramnose/química , Ramnose/metabolismo , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Humanos , Guanosina Difosfato Fucose/metabolismo , Guanosina Difosfato Fucose/química
2.
Bioorg Med Chem ; 25(11): 2844-2850, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28284868

RESUMO

We developed α1,6-fucosyltransferase (FUT8) inhibitors through a diversity-oriented synthesis. The coupling reaction between the fucose unit containing alkyne and the guanine unit containing sulfonyl azide under various conditions afforded a series of Guanosine 5'-diphospho-ß-l-fucose (GDP-fucose) analogs. The synthesized compounds displayed FUT8 inhibition activity. A docking study revealed that the binding mode of the inhibitor synthesized with FUT8 was similar to that of GDP-fucose.


Assuntos
Alcinos/farmacologia , Azidas/farmacologia , Inibidores Enzimáticos/farmacologia , Fucosiltransferases/antagonistas & inibidores , Guanosina Difosfato Fucose/farmacologia , Alcinos/química , Azidas/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/química , Humanos , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
3.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 7): 564-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27380374

RESUMO

The plant cell wall is a complex network of polysaccharides made up of cellulose, hemicelluloses and pectins. Xyloglucan (XyG), which is the main hemicellulosic component of dicotyledonous plants, has attracted much attention for its role in plant development and for its many industrial applications. The XyG-specific fucosyltransferase (FUT1) adds a fucose residue from GDP-fucose to the 2-O position of the terminal galactosyl residues on XyG side chains. Recombinant FUT1 from Arabidopsis thaliana was crystallized in two different crystal forms, with the best diffracting crystals (up to 1.95 Šresolution) belonging to the monoclinic space group P21, with unit-cell parameters a = 87.6, b = 84.5, c = 150.3 Å, ß = 96.3°. Ab initio phases were determined using a two-wavelength anomalous dispersion experiment on a tantalum bromide-derivatized crystal with data collected at the rising and descending inflection points of the Ta white line. An interpretable electron-density map was obtained after elaborate density modification. Model completion and structural analysis are currently under way.


Assuntos
Proteínas de Arabidopsis/química , Arabidopsis/química , Fucosiltransferases/química , Glucanos/química , Guanosina Difosfato Fucose/química , Xilanos/química , Animais , Arabidopsis/enzimologia , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Baculoviridae/genética , Baculoviridae/metabolismo , Clonagem Molecular , Cristalização , Cristalografia por Raios X , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Expressão Gênica , Glucanos/metabolismo , Guanosina Difosfato Fucose/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Spodoptera , Difração de Raios X , Xilanos/metabolismo , Galactosídeo 2-alfa-L-Fucosiltransferase
4.
Biotechnol Lett ; 38(10): 1747-52, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27314478

RESUMO

OBJECTIVES: To achieve multienzymatic cascade synthesis of fucosyl oligosaccharide from D-mannose by two-step fermentation pathway in Escherichia coli. RESULTS: E. coli BL21(DE3) harboring pET-22b(+) vectors with six genes, i.e., glucokinase (Glk), phosphomannomutase (ManB), mannose-1-phosphate guanylytransferase (ManC), GDP-mannose 4,6-dehydratase (Gmd), GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase/4-reductase (WcaG), and α-1,2-fucosyltransferase (Fuct) were co-inoculated, and the multienzyme synthetic pathway was constructed to produce fucosyloligosaccharide using D-mannose as substrate. The product, analyzed by LC/MS, fucosyloligosaccharide was formed under the catalysis of Fuct using GDP-fucose as donor substrate and lactose as acceptor substrate. Fucosyloligosaccharides reached 22 mM by a two-step fermentation compared to 3.7 mM with a one-pot fermentation. CONCLUSIONS: Fucosyloligosaccharide was produced by a two-step fermentation to avoid the inhibitory effect of GDP-fucose on Gmd. Two-step fermentation is a rational synthetic pathway for accumulating fucosyloligosaccharide.


Assuntos
Escherichia coli/crescimento & desenvolvimento , Fucose/química , Manose/metabolismo , Complexos Multienzimáticos/genética , Oligossacarídeos/biossíntese , Vias Biossintéticas , Desidrogenases de Carboidrato/genética , Carboidratos Epimerases/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Fermentação , Fucosiltransferases/genética , Vetores Genéticos/genética , Glucoquinase/genética , Guanosina Difosfato Fucose/química , Cetona Oxirredutases/genética , Lactose/química , Complexos Multienzimáticos/metabolismo , Nucleotidiltransferases/genética , Oligossacarídeos/química , Oligossacarídeos/isolamento & purificação , Fosfotransferases (Fosfomutases)/genética , Transformação Bacteriana
5.
Bioorg Med Chem ; 22(22): 6430-7, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25438767

RESUMO

Fucosyltransferases catalyze the transfer of l-fucose from an activated GDP-ß-l-fucose to various acceptor molecules such as N-acetyllactosamine. Frequently fucosylation is the final step within the glycosylation machinery, and the resulting glycans are involved in various cellular processes such as cell-cell recognition, adhesion and inflammation or tumor metastasis. The selective blocking of these interactions would thus be a potential promising therapeutic strategy. The syntheses and analyses of various potential α1,3-fucosyltransferase inhibitors derived from GDP-ß-l-fucose containing a triazole linker unit is summarized and the observed inhibitory effect was compared with that of small molecules such as GDP or fucose. To examine their specificity and selectivity, all inhibitors were tested with human α1,3-fucosyltransferase IX and Helicobacter pylori α1,3-fucosyltransferase, which is to date the only α1,3-fucosyltransferase with a known high resolution structure. Specific inhibitors which inhibit either H. pylori α1,3-fucosyltransferase or human fucosyltransferase IX with Ki values in the micromolar range were identified. In that regard, acetylated GDP-galactose derivative Ac-3 turned out to inhibit H. pylori α1,3-fucosyltransferase but not human fucosyltransferase IX, whereas GDP-6-amino-ß-l-fucose 17 showed an appreciably better inhibitory effect on fucosyltransferase IX activity than on that of H. pylori fucosyltransferase.


Assuntos
Inibidores Enzimáticos/síntese química , Fucosiltransferases/antagonistas & inibidores , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/síntese química , Guanosina Difosfato Fucose/química , Guanosina Difosfato Fucose/metabolismo , Helicobacter pylori/enzimologia , Humanos , Cinética , Ligação Proteica , Triazóis/química
6.
Proc Natl Acad Sci U S A ; 109(38): 15318-23, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22949680

RESUMO

Notch (N) is a transmembrane receptor that mediates cell-cell interactions to determine many cell-fate decisions. N contains EGF-like repeats, many of which have an O-fucose glycan modification that regulates N-ligand binding. This modification requires GDP-L-fucose as a donor of fucose. The GDP-L-fucose biosynthetic pathways are well understood, including the de novo pathway, which depends on GDP-mannose 4,6 dehydratase (Gmd) and GDP-4-keto-6-deoxy-D-mannose 3,5-epimerase/4-reductase (Gmer). However, the potential for intercellularly supplied GDP-L-fucose and the molecular basis of such transportation have not been explored in depth. To address these points, we studied the genetic effects of mutating Gmd and Gmer on fucose modifications in Drosophila. We found that these mutants functioned cell-nonautonomously, and that GDP-L-fucose was supplied intercellularly through gap junctions composed of Innexin-2. GDP-L-fucose was not supplied through body fluids from different isolated organs, indicating that the intercellular distribution of GDP-L-fucose is restricted within a given organ. Moreover, the gap junction-mediated supply of GDP-L-fucose was sufficient to support the fucosylation of N-glycans and the O-fucosylation of the N EGF-like repeats. Our results indicate that intercellular delivery is a metabolic pathway for nucleotide sugars in live animals under certain circumstances.


Assuntos
Carboidratos Epimerases/metabolismo , Drosophila/metabolismo , Junções Comunicantes/metabolismo , Guanosina Difosfato Fucose/química , Hidroliases/metabolismo , Receptores Notch/metabolismo , Alelos , Animais , Fator de Crescimento Epidérmico/metabolismo , Fucose/química , Regulação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Homozigoto , Modelos Genéticos , Mutação , Fenótipo , Recombinação Genética
7.
J Vis Exp ; (52)2011 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-21673647

RESUMO

Imaging glycans in vivo has recently been enabled using a bioorthogonal chemical reporter strategy by treating cells or organisms with azide- or alkyne-tagged monosaccharides. The modified monosaccharides, processed by the glycan biosynthetic machinery, are incorporated into cell surface glycoconjugates. The bioorthogonal azide or alkyne tags then allow covalent conjugation with fluorescent probes for visualization, or with affinity probes for enrichment and glycoproteomic analysis. This protocol describes the procedures typically used for noninvasive imaging of fucosylated glycans in zebrafish embryos, including: 1) microinjection of one-cell stage embryos with GDP-5-alkynylfucose (GDP-FucAl), 2) labeling fucosylated glycans in the enveloping layer of zebrafish embryos with azide-conjugated fluorophores via biocompatible Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC), and 3) imaging by confocal microscopy. The method described here can be readily extended to visualize other classes of glycans, e.g. glycans containing sialic acid and N-acetylgalactosamine, in developing zebrafish and in other living organisms.


Assuntos
Química Click/métodos , Polissacarídeos/análise , Peixe-Zebra/metabolismo , Alcinos/química , Animais , Azidas/química , Embrião não Mamífero/química , Embrião não Mamífero/metabolismo , Corantes Fluorescentes/química , Guanosina Difosfato Fucose/química , Microinjeções , Microscopia Confocal , Polissacarídeos/metabolismo
8.
Biochem Biophys Res Commun ; 391(4): 1663-9, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-20035716

RESUMO

Mortierella alpina is a filamentous fungus commonly found in soil, which is able to produce large amount of polyunsaturated fatty acids. L-fucose is an important sugar found in a diverse range of organisms, playing a variety of biological roles. In this study, we characterized the de novo biosynthetic pathway of GDP-L-fucose (the nucleotide-activated form of L-fucose) in M. alpina. Genes encoding GDP-D-mannose 4,6-dehydratase (GMD) and GDP-keto-6-deoxymannose 3,5-epimerase/4-reductase (GMER) were expressed heterologously in Escherichia coli. The recombinant enzymes were produced as His-tagged fusion proteins. Conversion of GDP-mannose to GDP-4-keto-6-deoxy mannose by GMD and GDP-4-keto-6-deoxy mannose to GDP-L-fucose by GMER were analyzed by capillary electrophoresis, electro-spray ionization-mass spectrometry, and nuclear magnetic resonance spectroscopy. The k(m) values of GMD for GDP-mannose and GMER for GDP-4-keto-6-deoxy mannose were determined to be 0.77 mM and 1.047 mM, respectively. Both NADH and NADPH may be used by GMER as the coenzyme. The optimum temperature and pH were determined to be 37 degrees C and pH 9.0 (GMD) or pH 7.0 (GMER). Divalent cations are not required for GMD and GMER activity, and the activities of both enzymes may be enhanced by DTT. To our knowledge this is the first report on the characterization of GDP-L-fucose biosynthetic pathway in fungi.


Assuntos
Guanosina Difosfato Fucose/biossíntese , Mortierella/enzimologia , Carboidratos Epimerases/genética , Carboidratos Epimerases/metabolismo , Cátions Bivalentes/química , Cátions Bivalentes/metabolismo , Guanosina Difosfato Fucose/química , Guanosina Difosfato Fucose/genética , Hidroliases/genética , Hidroliases/metabolismo , Concentração de Íons de Hidrogênio , Mortierella/genética , Espectrometria de Massas por Ionização por Electrospray , Temperatura
9.
Proc Natl Acad Sci U S A ; 106(38): 16096-101, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19805264

RESUMO

Lewis X (Le(x))-containing glycans play important roles in numerous cellular processes. However, the absence of robust, facile, and cost-effective methods for the synthesis of Le(x) and its structurally related analogs has severely hampered the elucidation of the specific functions of these glycan epitopes. Here we demonstrate that chemically defined guanidine 5'-diphosphate-beta-l-fucose (GDP-fucose), the universal fucosyl donor, the Le(x) trisaccharide, and their C-5 substituted derivatives can be synthesized on preparative scales, using a chemoenzymatic approach. This method exploits l-fucokinase/GDP-fucose pyrophosphorylase (FKP), a bifunctional enzyme isolated from Bacteroides fragilis 9343, which converts l-fucose into GDP-fucose via a fucose-1-phosphate (Fuc-1-P) intermediate. Combining the activities of FKP and a Helicobacter pylori alpha1,3 fucosyltransferase, we prepared a library of Le(x) trisaccharide glycans bearing a wide variety of functional groups at the fucose C-5 position. These neoglycoconjugates will be invaluable tools for studying Le(x)-mediated biological processes.


Assuntos
Proteínas de Bactérias/metabolismo , Guanosina Difosfato Fucose/biossíntese , Nucleotidiltransferases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Polissacarídeos/biossíntese , Trifosfato de Adenosina/metabolismo , Bacteroides fragilis/enzimologia , Catálise , Cristalografia por Raios X , Eletroforese em Gel de Poliacrilamida , Guanosina Difosfato Fucose/química , Guanosina Trifosfato/metabolismo , Humanos , Cinética , Antígenos CD15/química , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Polissacarídeos/química , Especificidade por Substrato
10.
J Am Chem Soc ; 130(51): 17593-602, 2008 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19053199

RESUMO

L-fucose, 6-deoxy-L-galactose, is a key component of many important glycoconjugates including the blood group antigens and the Lewis(X) ligands. The biosynthesis of GDP-L-fucose begins with the action of a dehydratase that converts GDP-D-mannose into GDP-4-keto-6-deoxy-mannose. The enzyme GDP-fucose synthase, GFS, (also known as GDP-4-keto-6-deoxy-D-mannose epimerase/reductase, GMER) then converts GDP-4-keto-6-deoxy-D-mannose into GDP-L-fucose. The GFS reaction involves epimerizations at both C-3'' and C-5'' followed by an NADPH-dependent reduction of the carbonyl at C-4. This manuscript describes studies that elucidate the order of the epimerization steps and the roles of the active site acid/base residues responsible for the epimerizations. An active site mutant, Cys109Ser, produces GDP-6-deoxy-D-altrose as its major product indicating that C-3'' epimerization occurs first and premature reduction of the GDP-4-keto-6-deoxy-D-altrose intermediate becomes competitive with GDP-L-fucose production. The same mutation results in the appearance of a kinetic isotope effect when [3''-(2)H]-GDP-6-deoxy-4-keto-mannose is used as a substrate. This indicates that Cys109 is the base responsible for the deprotonation of the substrate at C-3''. The Cys109Ser mutant also catalyzes a rapid wash-in of solvent derived deuterium into the C-5'' position of GDP-fucose in the presence of NADP(+). This confirms the order of epimerizations and the role of Cys109. Finally, the inactive His179Gln mutant readily catalyzes the wash-out of deuterium from the C-3'' position of [3''-(2)H]-GDP-6-deoxy-4-keto-mannose. Together these results strongly implicate an ordered sequence of epimerizations (C-3'' followed by C-5'') and suggest that Cys109 acts as a base and His179 acts as an acid in both epimerization steps.


Assuntos
Carboidratos Epimerases/química , Proteínas de Escherichia coli/química , Guanosina Difosfato Fucose/química , Cetona Oxirredutases/química , Complexos Multienzimáticos/química , Catálise , Domínio Catalítico , Clonagem Molecular , Deutério/química , Escherichia coli/metabolismo , Guanosina Difosfato/química , Cinética , Mutagênese Sítio-Dirigida , Mutação , Proteínas Recombinantes/química , Solventes/química , Espectrometria de Massas por Ionização por Electrospray , Especificidade por Substrato
11.
Carbohydr Res ; 343(5): 970-6, 2008 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-18280461

RESUMO

The 3-sulfated Lewis(a) pentasaccharide was synthesized on multimeric-based polyethylene glycol support. Coupling of O-(2,3,4,6-tetra-O-acetyl-beta-D-galactopyranosyl)-(1-->3)-4,6-di-O-acetyl-2-deoxy-2-phthalimido-beta-D-glucopyranosyl trichloroacetimidate with (2,6-di-O-acetyl-beta-D-galactopyranosyl)-(1-->4)-(2,3,6-tri-O-acetyl-beta-D-glucopyranoside) bound onto the polymer afforded lacto-N-tetraose, which was then regioselectively sulfated at the 3-OH position of the terminal galactose using the stannylene procedure. Fucosylation of the sulfated tetrasaccharide was performed using an immobilized fucosyltransferase FucTIII to give the title compound after cleavage.


Assuntos
Fucosiltransferases/química , Oligossacarídeos/síntese química , Polietilenoglicóis/química , Sequência de Carboidratos , Cromatografia Líquida , Guanosina Difosfato Fucose/química , Antígenos do Grupo Sanguíneo de Lewis , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Modelos Químicos , Dados de Sequência Molecular , Estrutura Molecular , Oligossacarídeos/química , Propilenoglicóis/química , Óxidos de Enxofre/química
12.
Chemistry ; 14(2): 478-87, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17929334

RESUMO

We have developed a facile and versatile protocol for the continuous monitoring of human fucosyltransferases activity by using fluorescence energy resonance transfer (FRET), and have explored the feasibility of its use in an inhibitor screening assay. A convenient sugar nucleotide with a fluorogenic probe, 6-deoxy-6-N-(2-naphalene-2-yl-acetamide)-beta-L-galactopyranos-1-yl-guanosine 5'-diphosphate disodium salt (1), was efficiently synthesized from naturally abundant D-galactopyranose via a key intermediate, 6-azide-1,2,3,4-tetra-O-benzoyl-6-deoxy-beta-L-galactopyranose (10). It was demonstrated that the combined use of the glycosyl donor 1 and a dansylated acceptor substrate, sialyl-alpha2,3-LacNAc derivative (2) allowed us to carry out highly sensitive, direct, and continuous in vitro monitoring of the generation of sialyl Lewis X (SLe x), which is catalyzed by human alpha-1,3-fucosyltransferase VI (FUT-VI). A kinetic analysis revealed that compound 1 was an excellent donor substrate (KM=0.94 microM and Vmax=0.14 microM min(-1)) for detecting human FUT-VI activity. To the best of our knowledge, this synthetic fluorogenic probe is the most sensitive and selective donor substrate for FUT-VI among all of the known GDP-Fuc analogues, including the parent GDP-Fuc. When a dansylated asparagine-linked glycopeptide 20, which is derived from egg yolk was employed as an alternate acceptor substrate, a FRET-based assay with compound 1 could be used to directly monitor the alpha1,6-fucosylation at the reducing terminal GlcNAc residue by human FUT-VIII (KM=175 microM and Vmax=0.06 microM/ min); this indicates that the present method might become a general protocol for the characterization of various mammalian fucosyltransferases in the presence of designated fluorogenic acceptor substrates. The present protocol revealed that compound 23, which was obtained by a 1,3-dipolar cycloaddition between the disodium salt 16 and 1-ethynyl-naphthalene exhibits highly potent inhibitory effects against the FUT-VI-mediated sialyl Lewis X synthesis (IC50=5.4 microM).


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Fucosiltransferases/química , Galactose/química , Guanosina Difosfato Fucose/síntese química , Ativação Enzimática , Guanosina Difosfato Fucose/química , Humanos , Estrutura Molecular , Estereoisomerismo , Fatores de Tempo
13.
J Biol Chem ; 283(1): 184-193, 2008 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17974560

RESUMO

GDP-D-mannose 4,6-dehydratase (GMD) is a key enzyme involved in the synthesis of 6-deoxyhexoses in prokaryotes and eukaryotes. Paramecium bursaria chlorella virus-1 (PBCV-1) encodes a functional GMD, which is unique among characterized GMDs because it also has a strong stereospecific NADPH-dependent reductase activity leading to GDP-D-rhamnose formation (Tonetti, M., Zanardi, D., Gurnon, J., Fruscione, F., Armirotti, A., Damonte, G., Sturla, L., De Flora, A., and Van Etten, J.L. (2003) J. Biol. Chem. 278, 21559-21565). In the present study we characterized a recombinant GMD encoded by another chlorella virus, Acanthocystis turfacea chlorella virus 1 (ATCV-1), demonstrating that it has the expected dehydratase activity. However, it also displayed significant differences when compared with PBCV-1 GMD. In particular, ATCV-1 GMD lacks the reductase activity present in the PBCV-1 enzyme. Using recombinant PBCV-1 and ATCV-1 GMDs, we determined that the enzymatically active proteins contain tightly bound NADPH and that NADPH is essential for maintaining the oligomerization status as well as for the stabilization and function of both enzymes. Phylogenetic analysis indicates that PBCV-1 GMD is the most evolutionary diverged of the GMDs. We conclude that this high degree of divergence was the result of the selection pressures that led to the acquisition of new reductase activity to synthesize GDP-D-rhamnose while maintaining the dehydratase activity in order to continue to synthesize GDP-L-fucose.


Assuntos
Chlorella/virologia , Hidroliases/metabolismo , NADP/metabolismo , Phycodnaviridae/metabolismo , Proteínas Virais/metabolismo , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Guanosina Difosfato Fucose/química , Guanosina Difosfato Fucose/metabolismo , Guanosina Difosfato Manose/química , Guanosina Difosfato Manose/metabolismo , Hidroliases/química , Hidroliases/classificação , Estrutura Molecular , Filogenia , Espectrometria de Fluorescência/métodos , Proteínas Virais/química , Proteínas Virais/classificação
14.
Glycobiology ; 17(9): 955-62, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17602138

RESUMO

L-Fucose for mammalian glycosylation contains an anomeric carbon atom generating alpha- and beta-L-fucoses. Based on sequence comparison of mouse and human homologs with the prokaryotic fucose mutarotases (FucU) characterized previously, we investigated their function in mammalian cells. By nuclear magnetic resonance (NMR) measurement with saturation difference analysis, the purified mammalian mutarotases were demonstrated to be involved in an interconversion between the two anomeric forms with comparable efficiency as that of the Escherichia coli FucU. The mouse gene was widely expressed in various tissues and cell lines, including kidney, liver, and pancreas, although expression was marginal in muscle and testis. By generating stably expressed cell lines for mutarotase genes in HepG2, it was shown that fucose incorporations into cellular proteins were increased as demonstrated by an incorporation of radiolabeled fucose into the cells. Furthermore, intracellular levels of GDP-L-fucose, measured with high performance liquid chromatography (HPLC), were enhanced by an overproduction of cellular mutarotase, which was reversed by gene silencing of mutarotase based on RNA interference. The results suggest that the mammalian mutarotase is functional in facilitated incorporation of fucose through the salvage pathway.


Assuntos
Carboidratos Epimerases/química , Fucose/química , Guanosina Difosfato Fucose/química , Sequência de Aminoácidos , Animais , Carboidratos Epimerases/fisiologia , Linhagem Celular Tumoral , Escherichia coli/metabolismo , Fucose/fisiologia , Cobaias , Humanos , Espectroscopia de Ressonância Magnética/métodos , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
15.
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
16.
Biometals ; 17(1): 35-43, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14977360

RESUMO

The effect of manganese and other divalent cations on the activity of a soluble recombinant form of human alpha3/4 fucosyltransferase III (SFT3) expressed in Spodoptera frugiperda (Sf9) insect cells was studied. SFT3 was active in the absence of divalent cations with an optimum pH of 4.5. In the absence of Mn2+ increasing the pH from 4.5 to 7.0 caused a decrease in the affinity of SFT3 for the acceptor Galbeta3GlcNAcO(CH2)3NHCO(CH2)5NH-biotin, as monitored by the 4-fold increase in the apparent KM value (0.9 to 3.3 mM). At pH 7.0, the addition of Mn2+ activated the enzyme and caused an increase in the affinity of SFT3 for the acceptor, as monitored by the 5-fold decrease of the apparent KM value (3.3 to 0.7 mM). In solution, a complex between GDP-Fuc donor and the divalent cation Mn2+ was observed by electrospray ionization mass spectrometry, in a 1:1 stoichiometry. These results indicated that Mn2+ bound the enzyme and increased its affinity for the acceptor; one possible functional role of manganese in catalysis could be as an electrophilic catalyst, co-ordinating the negative charges of the phosphate groups of the GDP-Fuc donor and promoting Fuc transfer. At low pH values such role would be played by the proton.


Assuntos
Fucosiltransferases/metabolismo , Manganês/farmacologia , Sequência de Aminoácidos , Animais , Catálise/efeitos dos fármacos , Cátions Bivalentes/metabolismo , Cátions Bivalentes/farmacologia , Linhagem Celular , Fucosiltransferases/química , Fucosiltransferases/genética , Expressão Gênica , Guanosina/química , Guanosina/metabolismo , Guanosina Difosfato Fucose/química , Guanosina Difosfato Fucose/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Cinética , Manganês/metabolismo , Dados de Sequência Molecular , Alinhamento de Sequência , Espectrometria de Massas por Ionização por Electrospray , Spodoptera/enzimologia , Spodoptera/genética
17.
Glycobiology ; 10(10): 1041-7, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11030750

RESUMO

Fucosylation of glycans on glycoproteins and -lipids requires the enzymatic activity of relevant fucosyltransferases and GDP-L-fucose as the donor. Due to the biological importance of fucosylated glycans, a readily accessible source of GDP-L-fucose would be required. Here we describe the construction of a stable recombinant S.cerevisiae strain expressing the E.coli genes gmd and wcaG encoding the two enzymes, GDP-mannose-4,6-dehydratase (GMD) and GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase/4-reductase (GMER(FX)) respectively, needed to convert GDP-mannose to GDP-fucose via the de novo pathway. Taking advantage of the rich inherent cytosolic GDP-mannose pool in S.cerevisiae cells we could easily produce 0.2 mg/l of GDP-L-fucose with this recombinant yeast strain without addition of any external GDP-mannose. The GDP-L-fucose product could be used as the fucose donor for alpha1,3fucosyltransferase to synthesize sialyl Lewis x (sLex), a glycan crucial for the selectin-dependent leukocyte traffic.


Assuntos
Proteínas de Escherichia coli , Escherichia coli/enzimologia , Guanosina Difosfato Fucose/biossíntese , Guanosina Difosfato Manose/metabolismo , Cetona Oxirredutases , Complexos Multienzimáticos , Saccharomyces cerevisiae/metabolismo , Carboidratos Epimerases/genética , Carboidratos Epimerases/metabolismo , Genes Bacterianos , Engenharia Genética/métodos , Guanosina Difosfato Fucose/química , Hidroliases/genética , Hidroliases/metabolismo , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Estereoisomerismo , Desidrogenase do Álcool de Açúcar/genética , Desidrogenase do Álcool de Açúcar/metabolismo , Transformação Genética
18.
J Org Chem ; 65(12): 3811-5, 2000 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-10864769

RESUMO

Both five- and six-membered iminocyclitols have proven to be useful transition-state analogue inhibitors of glycosidases. They also mimic the transition-state sugar moiety of the nucleoside phosphate sugar in glycosyltransferase-catalyzed reactions. Described here is the development of a general strategy toward the parallel synthesis of a five-membered iminocyclitol linked to a hydroxamic acid group designed to mimic the transition state of GDP-fucose complexed with Mn(II) in fucosyltransferase reactions. The iminocyclitol 8 containing a protected hydroxylamine unit was prepared from D-mannitol. The hydroxamic acid moiety was introduced via the reaction of 8 with various acid chlorides. The strategy is generally applicable to the construction of libraries for identification of glycosyltransferase inhibitors.


Assuntos
Ácidos Hidroxâmicos/síntese química , Pirrolidinas/síntese química , Fucosiltransferases/química , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/química , Guanosina Difosfato Fucose/metabolismo , Ácidos Hidroxâmicos/química , Indicadores e Reagentes , Manganês/química , Manganês/metabolismo , Manitol , Conformação Molecular , Estrutura Molecular , Pirrolidinas/química , Soluções
19.
Anal Biochem ; 267(1): 227-33, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9918676

RESUMO

For assays involving glycosyltransferases or transporters, several GDP-sugars are either commercially unavailable or expensive. We describe an enzymatic synthesis of GDP-d-[3H]arabinosep and GDP-l-[3H]fucose that yields 66-95% nucleotide-sugar from the appropriate radiolabeled sugar in less than 30 min. The coupled reaction requires Mg2+, ATP, and GTP along with the appropriate radioactive monosaccharide, sugar-1-kinase, and pyrophosphorylase. The latter two activities are present in a cytosolic fraction of Crithidia fasciculata, which is easily grown at room temperature in simple culture medium without serum or added CO2. Addition of commercial yeast inorganic pyrophosphatase shifts the equilibrium of the pyrophosphorylase reaction toward nucleotide-sugar formation. To verify that these nucleotide-sugars are biologically active, we tested their ability to serve as substrates for glycosyltransferases. GDP-l-[3H]fucose functions as the donor substrate for recombinant human fucosyltransferase V, and GDP-d-[3H]arabinosep serves as the donor substrate for the arabinosyltransferase activities present in Leishmania major microsomes.


Assuntos
Proteínas de Arabidopsis , Guanosina Difosfato Fucose/biossíntese , Açúcares de Guanosina Difosfato/biossíntese , Animais , Cromatografia Líquida de Alta Pressão/métodos , Crithidia fasciculata/enzimologia , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/química , Açúcares de Guanosina Difosfato/química , Humanos , Técnicas In Vitro , Cinética , Leishmania major/enzimologia , Nucleotidiltransferases/metabolismo , Pentosiltransferases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Trítio
20.
FEBS Lett ; 409(2): 312-6, 1997 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-9202168

RESUMO

The nolK gene of Azorhizobium caulinodans is essential for the incorporation of a fucosyl group in Nod factors. A NAD(P)-binding site is present in the NolK amino acid sequence and the gene is homologous to Escherichia coli genes, presumably involved in GDP-fucose synthesis. Protein extracts of A. caulinodans, overexpressing nolK, have an enzyme activity that synthesizes GDP-fucose from GDP-mannose. nolK most probably encodes a 4-reductase performing the last step in GDP-fucose synthesis. Wild-type A. caulinodans produces a population of fucosylated and non-fucosylated molecules but the nolK-overexpressing strain produces only fucosylated Nod factors. Thus, the production of activated fucosyl donors is a rate-limiting step in Nod factor fucosylation.


Assuntos
Genes Bacterianos , Guanosina Difosfato Fucose/biossíntese , Guanosina Difosfato Manose/biossíntese , Fixação de Nitrogênio/fisiologia , Rhizobiaceae/genética , Fucose/metabolismo , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/química , Guanosina Difosfato Manose/química , Fixação de Nitrogênio/genética , Rhizobiaceae/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...