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








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 7024, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34857733

RESUMO

The sugar fucose is expressed on mammalian cell membranes as part of glycoconjugates and mediates essential physiological processes. The aberrant expression of fucosylated glycans has been linked to pathologies such as cancer, inflammation, infection, and genetic disorders. Tools to modulate fucose expression on living cells are needed to elucidate the biological role of fucose sugars and the development of potential therapeutics. Herein, we report a class of fucosylation inhibitors directly targeting de novo GDP-fucose biosynthesis via competitive GMDS inhibition. We demonstrate that cell permeable fluorinated rhamnose 1-phosphate derivatives (Fucotrim I & II) are metabolic prodrugs that are metabolized to their respective GDP-mannose derivatives and efficiently inhibit cellular fucosylation.


Assuntos
Inibidores Enzimáticos/farmacologia , Fucose/química , Guanosina Difosfato Fucose/antagonistas & inibidores , Hidroliases/antagonistas & inibidores , Pró-Fármacos/farmacologia , Animais , Sequência de Carboidratos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Expressão Gênica , Glicosilação/efeitos dos fármacos , Guanosina Difosfato Fucose/biossíntese , Halogenação , Humanos , Hidroliases/genética , Hidroliases/metabolismo , Células Jurkat , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Pró-Fármacos/síntese química , Relação Estrutura-Atividade , Células THP-1
2.
J Agric Food Chem ; 68(20): 5668-5675, 2020 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-32336091

RESUMO

Guanosine 5'-diphosphate (GDP)-l-fucose is an important nucleotide sugar involved in the synthesis of fucosylated oligosaccharides, such as fucosylated human milk oligosaccharides, which play important roles in physiological and pathological processes. Here, a combinatorial modular pathway engineering strategy was implemented to efficiently increase the intracellular titers of GDP-l-fucose in engineered Escherichia coli. The de novo GDP-l-fucose synthesis pathway was partitioned into two modules and fine-tuned at both transcriptional and translational levels, which remarkably improved the GDP-l-fucose production. In addition, the gene encoding the UDP-glucose lipid carrier transferase (WcaJ) was inactivated to eliminate the competing metabolite pathway from GDP-l-fucose to colanic acid. Furthermore, cofactors were regenerated to promote biocatalysis. Taken together, the final engineered strain EWL37, which could achieve a titer of 18.33 mg/L in shake-flask cultivation, showed 106.21 mg/L intracellular GDP-l-fucose accumulation and a DCW-specific GDP-l-fucose content of 4.28 mg/g through fed-batch cultivation. In general, this study demonstrated that the utilization of combinatorial modular pathway engineering significantly improved the de novo synthesis of GDP-l-fucose in engineered E. coli.


Assuntos
Escherichia coli/genética , Escherichia coli/metabolismo , Guanosina Difosfato Fucose/biossíntese , Vias Biossintéticas , Fucose/metabolismo , Guanosina Difosfato/metabolismo , Engenharia Metabólica
3.
Metab Eng ; 52: 232-242, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30557615

RESUMO

Oligosaccharides present in human breast milk have been linked to beneficial effects on infant health. Inclusion of these human milk oligosaccharides (HMOs) in infant formula can recapitulate these health benefits. As a result, there is substantial commercial interest in a cost-effective source of HMOs as infant formula ingredients. Here we demonstrate that the yeast species Saccharomyces cerevisiae and Yarrowia lipolytica both can be engineered to produce 2'-fucosyllactose (2'FL), which is the most abundant oligosaccharide in human breast milk, at high titer and productivity. Both yeast species were modified to enable uptake of lactose and synthesis of GDP-fucose - the two precursors of 2'FL - by installing a lactose transporter and enzymes that convert GDP-mannose to GDP-fucose. Production of 2'FL was then enabled by expression of α-1,2-fucosyltransferases from various organisms. By screening candidate transporters from a variety of sources, we identified transporters capable of exporting 2'FL from yeast, which is a key consideration for any biocatalyst for 2'FL production. In particular, we identified CDT2 from Neurospora crassa as a promising target for further engineering to improve 2'FL efflux. Finally, we demonstrated production of 2'FL in fermenters at rates and titers that indicate the potential of engineered S. cerevisiae and Y. lipolytica strains for commercial 2'FL production.


Assuntos
Engenharia Metabólica/métodos , Leite Humano/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Trissacarídeos/biossíntese , Yarrowia/genética , Yarrowia/metabolismo , Feminino , Fermentação , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/biossíntese , Humanos , Lactose/biossíntese , Neurospora crassa/genética , Neurospora crassa/metabolismo , Galactosídeo 2-alfa-L-Fucosiltransferase
4.
Cell Chem Biol ; 24(12): 1467-1478.e5, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29033318

RESUMO

Fucosylation is a glycan modification critically involved in cancer and inflammation. Although potent fucosylation inhibitors are useful for basic and clinical research, only a few inhibitors have been developed. Here, we focus on a fucose analog with an alkyne group, 6-alkynyl-fucose (6-Alk-Fuc), which is used widely as a detection probe for fucosylated glycans, but is also suggested for use as a fucosylation inhibitor. Our glycan analysis using lectin and mass spectrometry demonstrated that 6-Alk-Fuc is a potent and general inhibitor of cellular fucosylation, with much higher potency than the existing inhibitor, 2-fluoro-fucose (2-F-Fuc). The action mechanism was shown to deplete cellular GDP-Fuc, and the direct target of 6-Alk-Fuc is FX (encoded by TSTA3), the bifunctional GDP-Fuc synthase. We also show that 6-Alk-Fuc halts hepatoma invasion. These results highlight the unappreciated role of 6-Alk-Fuc as a fucosylation inhibitor and its potential use for basic and clinical science.


Assuntos
Alcinos/farmacologia , Antineoplásicos/farmacologia , Carboidratos Epimerases/antagonistas & inibidores , Carcinoma Hepatocelular/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Fucose/farmacologia , Guanosina Difosfato Fucose/biossíntese , Cetona Oxirredutases/antagonistas & inibidores , Neoplasias Hepáticas/tratamento farmacológico , Alcinos/química , Antineoplásicos/química , Carboidratos Epimerases/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/química , Fucose/química , Células HEK293 , Células HeLa , Humanos , Cetona Oxirredutases/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia
5.
Gastroenterology ; 152(1): 193-205.e10, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27639802

RESUMO

BACKGROUND & AIMS: De novo synthesis of guanosine diphosphate (GDP)-fucose, a substrate for fucosylglycans, requires sequential reactions mediated by GDP-mannose 4,6-dehydratase (GMDS) and GDP-4-keto-6-deoxymannose 3,5-epimerase-4-reductase (FX or tissue specific transplantation antigen P35B [TSTA3]). GMDS deletions and mutations are found in 6%-13% of colorectal cancers; these mostly affect the ascending and transverse colon. We investigated whether a lack of fucosylation consequent to loss of GDP-fucose synthesis contributes to colon carcinogenesis. METHODS: FX deficiency and GMDS deletion produce the same biochemical phenotype of GDP-fucose deficiency. We studied a mouse model of fucosylation deficiency (Fx-/- mice) and mice with the full-length Fx gene (controls). Mice were placed on standard chow or fucose-containing diet (equivalent to a control fucosylglycan phenotype). Colon tissues were collected and analyzed histologically or by enzyme-linked immunosorbent assays to measure cytokine levels; T cells also were collected and analyzed. Fecal samples were analyzed by 16s ribosomal RNA sequencing. Mucosal barrier function was measured by uptake of fluorescent dextran. We transplanted bone marrow cells from Fx-/- or control mice (Ly5.2) into irradiated 8-week-old Fx-/- or control mice (Ly5.1). We performed immunohistochemical analyses for expression of Notch and the hes family bHLH transcription factor (HES1) in colon tissues from mice and a panel of 60 human colorectal cancer specimens (27 left-sided, 33 right-sided). RESULTS: Fx-/- mice developed colitis and serrated-like lesions. The intestinal pathology of Fx-/- mice was reversed by addition of fucose to the diet, which restored fucosylation via a salvage pathway. In the absence of fucosylation, dysplasia appeared and progressed to adenocarcinoma in up to 40% of mice, affecting mainly the right colon and cecum. Notch was not activated in Fx-/- mice fed standard chow, leading to decreased expression of its target Hes1. Fucosylation deficiency altered the composition of the fecal microbiota, reduced mucosal barrier function, and altered epithelial proliferation marked by Ki67. Fx-/- mice receiving control bone marrow cells had intestinal inflammation and dysplasia, and reduced expression of cytokines produced by cytotoxic T cells. Human sessile serrated adenomas and right-sided colorectal tumors with epigenetic loss of MutL homolog 1 (MLH1) had lost or had lower levels of HES1 than other colorectal tumor types or nontumor tissues. CONCLUSIONS: In mice, fucosylation deficiency leads to colitis and adenocarcinoma, loss of Notch activation, and down-regulation of Hes1. HES1 loss correlates with the development of human right-sided colorectal tumors with epigenetic loss of MLH1. These findings indicate that carcinogenesis in a subset of colon cancer is consequent to a molecular mechanism driven by fucosylation deficiency and/or HES1-loss.


Assuntos
Adenocarcinoma/etiologia , Carboidratos Epimerases/deficiência , Colite/etiologia , Colite/metabolismo , Colo/metabolismo , Neoplasias do Colo/etiologia , Mucosa Intestinal/metabolismo , Cetona Oxirredutases/deficiência , Adenocarcinoma/química , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Transplante de Medula Óssea , Carboidratos Epimerases/genética , Carcinogênese , Ceco/patologia , Proliferação de Células , Colite/patologia , Colite/prevenção & controle , Colo/patologia , Neoplasias do Colo/química , Neoplasias do Colo/patologia , Citocinas/genética , Citocinas/metabolismo , Fezes/microbiologia , Feminino , Fucose/administração & dosagem , Microbioma Gastrointestinal , Guanosina Difosfato Fucose/biossíntese , Guanosina Difosfato Fucose/deficiência , Humanos , Cetona Oxirredutases/genética , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Permeabilidade , RNA Mensageiro/metabolismo , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1/análise , Fatores de Transcrição HES-1/metabolismo , Adulto Jovem
6.
Sci Rep ; 6: 37230, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27849032

RESUMO

Glycosylation is an important posttranslational protein modification in all eukaryotes. Besides glycosylphosphatidylinositol (GPI) anchors and N-glycosylation, O-fucosylation has been recently reported in key sporozoite proteins of the malaria parasite. Previous analyses showed the presence of GDP-fucose (GDP-Fuc), the precursor for all fucosylation reactions, in the blood stages of Plasmodium falciparum. The GDP-Fuc de novo pathway, which requires the action of GDP-mannose 4,6-dehydratase (GMD) and GDP-L-fucose synthase (FS), is conserved in the parasite genome, but the importance of fucose metabolism for the parasite is unknown. To functionally characterize the pathway we generated a PfGMD mutant and analyzed its phenotype. Although the labelling by the fucose-binding Ulex europaeus agglutinin I (UEA-I) was completely abrogated, GDP-Fuc was still detected in the mutant. This unexpected result suggests the presence of an alternative mechanism for maintaining GDP-Fuc in the parasite. Furthermore, PfGMD null mutant exhibited normal growth and invasion rates, revealing that the GDP-Fuc de novo metabolic pathway is not essential for the development in culture of the malaria parasite during the asexual blood stages. Nonetheless, the function of this metabolic route and the GDP-Fuc pool that is generated during this stage may be important for gametocytogenesis and sporogonic development in the mosquito.


Assuntos
Fucose/metabolismo , Glicoconjugados/metabolismo , Guanosina Difosfato Fucose/biossíntese , Plasmodium falciparum/metabolismo , Vias Biossintéticas/genética , Genoma de Protozoário/genética , Hidroliases/genética , Hidroliases/metabolismo , Microscopia de Fluorescência , Mutação , Fenótipo , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
7.
Biotechnol Lett ; 38(10): 1761-8, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27395065

RESUMO

OBJECTIVE: To clarify the molecular mechanism of GDP-L-fucose biosynthesis in Mortierella alpina. RESULTS: Analysis of the M. alpina genome suggests that there were two isofunctional GDP-D-mannose-4,6-dehydratase genes (GMD1 and GMD2) that have never been found in a microorganism before. GMD2 was expressed heterologously in Escherichia coli and purified to homogeneity. The addition of exogenous NAD(+) or NADP(+) was not essential for GMD2 activity. GMD2 may have considerable importance for GDP-L-fucose biosynthesis under nitrogen starvation. The transcriptional regulation of GMD1 may be more susceptible to GDP and GTP than that of GMD2. Significant changes were observed in the concentration of GDP-L-fucose (30 and 36 % inhibition respectively) and total fatty acids (18 and 12 % inhibition respectively) in M. alpina grown on GMD inhibitors medium, which suggests that GDP-L-fucose is functionally significant in lipid metabolism. CONCLUSIONS: This is the first time that an isofunctional GDP-D-mannose-4,6-dehydratase has been characterized in a microorganism.


Assuntos
Clonagem Molecular/métodos , Guanosina Difosfato Fucose/biossíntese , Hidroliases/genética , Hidroliases/metabolismo , Mortierella/enzimologia , Escherichia coli/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/isolamento & purificação , Proteínas Fúngicas/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Fúngica da Expressão Gênica , Genoma Fúngico , Hidroliases/isolamento & purificação , Isoenzimas/genética , Isoenzimas/isolamento & purificação , Isoenzimas/metabolismo , Metabolismo dos Lipídeos , Mortierella/genética , Nitrogênio/metabolismo
8.
Glycobiology ; 26(8): 880-887, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26957583

RESUMO

GDP-l-fucose functions as a biological donor for fucosyltransferases, which are required for the catalysis of l-fucose to various acceptor molecules including oligosaccharides, glycoproteins and glycolipids. Mortierella alpina is one of the highest lipid-producing fungi and can biosynthesis GDP-l-fucose in the de novo pathway. Analysis of the M. alpina genome suggests that there is a gene encoding l-fucokinase (FUK) for the conversion of fucose to l-fucose-1-phosphate in the GDP-l-fucose salvage pathway, which has never been found in fungi before. This gene was characterized to explore its role in GDP-l-fucose synthesis. The yield of GDP-l-fucose is relatively higher in lipid accumulation phase (0.096 mg per g cell) than that in cell multiplication phase (0.074 mg per g cell) of M. alpina Additionally, the transcript level of FUK is up regulated by nitrogen exhaustion when M. alpina starts to accumulate lipid, highlights the functional significance of FUK in the GDP-l-fucose biosynthesis in M. alpina Gene encoding FUK was expressed heterologously in Escherichia coli and the resulting protein was purified to homogeneity. The product of FUK reaction was analyzed by liquid chromatography and mass spectrometry. Kinetic parameters and other properties of FUK were investigated. Comparative analyses between the FUK protein and other homologous proteins were performed. To our knowledge, this study is the first to report a comprehensive characterization of FUK in a fungus. Mortierella alpina could be used as an alternative source for the production of GDP-l-fucose.


Assuntos
Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Guanosina Difosfato Fucose/biossíntese , Mortierella/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , RNA Mensageiro/metabolismo , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Fucose/metabolismo , Proteínas Fúngicas/genética , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Cinética , Redes e Vias Metabólicas/genética , Mortierella/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , RNA Mensageiro/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
Enzyme Microb Technol ; 69: 38-45, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25640723

RESUMO

Guanosine 5'-diphosphate (GDP)-fucose is the indispensible donor substrate for fucosyltransferase-catalyzed synthesis of fucose-containing biomolecules, which have been found involving in various biological functions. In this work, the salvage pathway for GDP-fucose biosynthesis from Bacterioides fragilis was introduced into Escherichia coli. Besides, the biosynthesis of guanosine 5'-triphosphate (GTP), an essential substrate for GDP-fucose biosynthesis, was enhanced via overexpression of enzymes involved in the salvage pathway of GTP biosynthesis. The production capacities of metabolically engineered strains bearing different combinations of recombinant enzymes were compared. The shake flask fermentation of the strain expressing Fkp, Gpt, Gmk and Ndk obtained the maximum GDP-fucose content of 4.6 ± 0.22 µmol/g (dry cell mass), which is 4.2 fold that of the strain only expressing Fkp. Through fed-batch fermentation, the GDP-fucose content further rose to 6.6 ± 0.14 µmol/g (dry cell mass). In addition to a better productivity than previous fermentation processes based on the de novo pathway for GDP-fucose biosynthesis, the established schemes in this work also have the advantage to be a potential avenue to GDP-fucose analogs encompassing chemical modification on the fucose residue.


Assuntos
Guanosina Difosfato Fucose/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteroides fragilis/enzimologia , Bacteroides fragilis/genética , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Fermentação , Engenharia Genética , Engenharia Metabólica , Redes e Vias Metabólicas , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
J Biol Chem ; 288(23): 16506-16517, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23615908

RESUMO

Carbohydrate structures play important roles in many biological processes, including cell adhesion, cell-cell communication, and host-pathogen interactions. Sugar nucleotides are activated forms of sugars used by the cell as donors for most glycosylation reactions. Using a liquid chromatography-tandem mass spectrometry-based method, we identified and quantified the pools of UDP-glucose, UDP-galactose, UDP-N-acetylglucosamine, GDP-mannose, and GDP-fucose in Plasmodium falciparum intraerythrocytic life stages. We assembled these data with the in silico functional reconstruction of the parasite metabolic pathways obtained from the P. falciparum annotated genome, exposing new active biosynthetic routes crucial for further glycosylation reactions. Fucose is a sugar present in glycoconjugates often associated with recognition and adhesion events. Thus, the GDP-fucose precursor is essential in a wide variety of organisms. P. falciparum presents homologues of GDP-mannose 4,6-dehydratase and GDP-L-fucose synthase enzymes that are active in vitro, indicating that most GDP-fucose is formed by a de novo pathway that involves the bioconversion of GDP-mannose. Homologues for enzymes involved in a fucose salvage pathway are apparently absent in the P. falciparum genome. This is in agreement with in vivo metabolic labeling experiments showing that fucose is not significantly incorporated by the parasite. Fluorescence microscopy of epitope-tagged versions of P. falciparum GDP-mannose 4,6-dehydratase and GDP-L-fucose synthase expressed in transgenic 3D7 parasites shows that these enzymes localize in the cytoplasm of P. falciparum during the intraerythrocytic developmental cycle. Although the function of fucose in the parasite is not known, the presence of GDP-fucose suggests that the metabolite may be used for further fucosylation reactions.


Assuntos
Guanosina Difosfato Fucose/biossíntese , Guanosina Difosfato Manose/biossíntese , Plasmodium falciparum/metabolismo , Carboidratos Epimerases/genética , Carboidratos Epimerases/metabolismo , Genoma/fisiologia , Guanosina Difosfato Fucose/genética , Guanosina Difosfato Manose/genética , Humanos , Hidroliases/genética , Hidroliases/metabolismo , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
11.
Bioprocess Biosyst Eng ; 36(6): 749-56, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23404100

RESUMO

Wild-type Corynebacterium glutamicum was metabolically engineered to convert glucose and mannose into guanosine 5'-diphosphate (GDP)-L-fucose, a precursor of fucosyl-oligosaccharides, which are involved in various biological and pathological functions. This was done by introducing the gmd and wcaG genes of Escherichia coli encoding GDP-D-mannose-4,6-dehydratase and GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase-4-reductase, respectively, which are known as key enzymes in the production of GDP-L-fucose from GDP-D-mannose. Coexpression of the genes allowed the recombinant C. glutamicum cells to produce GDP-L-fucose in a minimal medium containing glucose and mannose as carbon sources. The specific product formation rate was much higher during growth on mannose than on glucose. In addition, the specific product formation rate was further increased by coexpressing the endogenous phosphomanno-mutase gene (manB) and GTP-mannose-1-phosphate guanylyl-transferase gene (manC), which are involved in the conversion of mannose-6-phosphate into GDP-D-mannose. However, the overexpression of manA encoding mannose-6-phosphate isomerase, catalyzing interconversion of mannose-6-phosphate and fructose-6-phosphate showed a negative effect on formation of the target product. Overall, coexpression of gmd, wcaG, manB and manC in C. glutamicum enabled production of GDP-L-fucose at the specific rate of 0.11 mg g cell(-1) h(-1). The specific GDP-L-fucose content reached 5.5 mg g cell(-1), which is a 2.4-fold higher than that of the recombinant E. coli overexpressing gmd, wcaG, manB and manC under comparable conditions. Well-established metabolic engineering tools may permit optimization of the carbon and cofactor metabolisms of C. glutamicum to further improve their production capacity.


Assuntos
Carboidratos Epimerases , Corynebacterium glutamicum , Proteínas de Escherichia coli , Escherichia coli , Glucose/metabolismo , Guanosina Difosfato Fucose/biossíntese , Hidroliases , Cetona Oxirredutases , Manose/metabolismo , Complexos Multienzimáticos , Carboidratos Epimerases/biossíntese , Carboidratos Epimerases/genética , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Expressão Gênica , Glucose/farmacologia , Guanosina Difosfato Fucose/genética , Hidroliases/biossíntese , Hidroliases/genética , Cetona Oxirredutases/biossíntese , Cetona Oxirredutases/genética , Manose/farmacologia , Engenharia Metabólica/métodos , Complexos Multienzimáticos/biossíntese , Complexos Multienzimáticos/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Edulcorantes/metabolismo , Edulcorantes/farmacologia
12.
Microb Cell Fact ; 11: 48, 2012 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-22545760

RESUMO

BACKGROUND: 2'-Fucosyllactose (2-FL) is a functional oligosaccharide present in human milk which protects against the infection of enteric pathogens. Because 2-FL can be synthesized through the enzymatic fucosylation of lactose with guanosine 5'-diphosphate (GDP)-l-fucose by α-1,2-fucosyltransferase (FucT2), an 2-FL producing Escherichia coli can be constructed through overexpressing genes coding for endogenous GDP- l-fucose biosynthetic enzymes and heterologous fucosyltransferase. RESULTS: The gene for FucT2 from Helicobacter pylori was introduced to the GDP-l-fucose producing recombinant E. coli BL21 star(DE3) strain. However, only small amount of 2-FL was produced in a batch fermentation because the E. coli BL21star(DE3) strain assimilated lactose instead of converting to 2-FL. As an alternative host, the E. coli JM109(DE3) strain which is incapable of assimilating lactose was chosen as a 2-FL producer. Whole cell biosynthesis of 2-FL from lactose was investigated in a series of batch fermentations using various concentrations of lactose. The results of batch fermentations showed that lactose was slowly assimilated by the engineered E. coli JM109(DE3) strain and 2-FL was synthesized without supplementation of another auxiliary sugar for cell growth. A maximum 2-FL concentration of 1.23 g/l was obtained from a batch fermentation with 14.5 g/l lactose. The experimentally obtained yield (g 2-FL/g lactose) corresponded to 20% of the theoretical maximum yield estimated by the elementary flux mode (EFM) analysis. CONCLUSIONS: The experimental 2-FL yield in this study corresponded to about 20% of the theoretical maximum yield, which suggests further modifications via metabolic engineering of a host strain or optimization of fermentation processes might be carried out for improving 2-FL yield. Improvement of microbial production of 2-FL from lactose by engineered E. coli would increase the feasibility of utilizing 2-FL as a prebiotic in various foods.


Assuntos
Escherichia coli/genética , Escherichia coli/metabolismo , Microbiologia Industrial/métodos , Oligossacarídeos/biossíntese , Trissacarídeos/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fermentação , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Engenharia Genética , Guanosina Difosfato Fucose/biossíntese , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Lactose/metabolismo
13.
Expert Opin Ther Pat ; 21(9): 1497-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21548846

RESUMO

BACKGROUND: The application (WO2010141855) is in the field of glycobiology, and involves the control of the rate of fucosylation of proteins by exogenous factors. OBJECTIVE: It aims at controlling the rate of protein fucosylation with inhibitors (drugs or nucleic acid antagonists) of enzymes involved in the synthesis of GDP-fucose. METHODS: Mammalian cell lines were cultured in the presence of inhibitors, for example, siRNA. The rates of GDP-fucose in cells and during protein fucosylation were characterized. RESULTS: The level of protein fucosylation decreases rapidly in response to a decrease in GDP-fucose level. CONCLUSION: The relationship between the rate of fucosylation of proteins and the level of GDP-fucose in a cell is non-linear. Reduction in the rate of protein fucosylation can be achieved with a minimal reduction of the level of GDP-fucose in cells. The paradigm may be used to synthesize proteins and antibodies, with a reduced rate of fucosylation. The application claims that the use of drugs or nucleic acid antagonists that inhibit the enzymes involved in GDP-fucose biosynthesis optimizes the level of GDP-fucose present in cells, and reduces the rate of fucosylation of glycoproteins.


Assuntos
Fucose/metabolismo , Glicoproteínas/biossíntese , Guanosina Difosfato Fucose/biossíntese , Proteínas/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Inibidores Enzimáticos/farmacologia , Fucosiltransferases/antagonistas & inibidores , Cinética , Patentes como Assunto , RNA Interferente Pequeno/farmacologia
14.
Nat Protoc ; 5(4): 636-46, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20224564

RESUMO

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


Assuntos
Guanosina Difosfato Fucose/biossíntese , Uridina Difosfato N-Acetilgalactosamina/biossíntese , Uridina Difosfato N-Acetilglicosamina/biossíntese , Glicosilação , Glicosiltransferases/metabolismo , Oligossacarídeos/biossíntese , Oligossacarídeos/química , Fenômenos de Química Orgânica , Fosforilação , Polissacarídeos/biossíntese , Polissacarídeos/química , Uridina Difosfato N-Acetilgalactosamina/análogos & derivados , Uridina Difosfato N-Acetilglicosamina/análogos & derivados
15.
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
16.
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
17.
Bioresour Technol ; 100(24): 6143-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19692232

RESUMO

Guanosine 5'-diphosphate (GDP)-L-fucose, an activated form of a nucleotide sugar, plays an important role in a wide range of biological functions. In this study, the enhancement of GDP-L-fucose production was attempted by supplementation of mannose, which is a potentially better carbon source to be converted into GDP-L-fucose than glucose, and combinatorial overexpression of the genes involved in the biosynthesis of GDP-D-mannose, a precursor of GDP-L-fucose. Supply of a mannose and glucose led to a 1.3-fold-increase in GDP-L-fucose concentration (52.5+/-0.8 mg l(-1)) in a fed-batch fermentation of recombinant E. coli BL21star(DE3) overexpressing the gmd and wcaG genes, compared with the case using glucose as a sole carbon source. A maximum GDP-L-fucose concentration of 170.3+/-2.3 mg l(-1), corresponding to a 4.4-fold enhancement compared with the control strain overexpressing gmd and wcaG genes only, was achieved in a glucose-limited fed-batch fermentation of a recombinant E. coli BL21star(DE3) strain overexpressing manB, manC, gmd and wcaG genes. Further improvement of GDP-L-fucose production was not obtained by additional overexpression of the manA gene.


Assuntos
Escherichia coli/metabolismo , Guanosina Difosfato Fucose/biossíntese , Guanosina Difosfato Manose/metabolismo , Carbono/farmacologia , Eletroforese em Gel de Poliacrilamida , Escherichia coli/efeitos dos fármacos , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fermentação/efeitos dos fármacos , Genes Bacterianos , Engenharia Genética , Glucose/metabolismo , Glucose/farmacologia , Manose/metabolismo , Manose/farmacologia
18.
J Biol Chem ; 282(39): 28853-28863, 2007 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-17640865

RESUMO

The protozoan parasite Trypanosoma brucei causes human African sleeping sickness in sub-Saharan Africa. The parasite makes several essential glycoproteins, which has led to the investigation of the sugar nucleotides and glycosyltransferases required to synthesize these structures. Fucose is a common sugar in glycoconjugates from many organisms; however, the sugar nucleotide donor GDP-fucose was only recently detected in T. brucei, and the importance of fucose metabolism in this organism is not known. In this paper, we identified the genes encoding functional GDP-fucose biosynthesis enzymes in T. brucei and created conditional null mutants of TbGMD, the gene encoding the first enzyme in the pathway from GDP-mannose to GDP-fucose, in both bloodstream form and procyclic form parasites. Under nonpermissive conditions, both life cycle forms of the parasite became depleted in GDP-fucose and suffered growth arrest, demonstrating that fucose metabolism is essential to both life cycle stages. In procyclic form parasites, flagellar detachment from the cell body was also observed under nonpermissive conditions, suggesting that fucose plays a significant role in flagellar adhesion. Fluorescence microscopy of epitope-tagged TbGMD revealed that this enzyme is localized in glycosomes, despite the absence of PTS-1 or PTS-2 target sequences.


Assuntos
Desidrogenases de Carboidrato/metabolismo , Flagelos/enzimologia , Guanosina Difosfato Fucose/biossíntese , Guanosina Difosfato Manose/metabolismo , Proteínas de Protozoários/metabolismo , Trypanosoma brucei brucei/enzimologia , Animais , Desidrogenases de Carboidrato/genética , Flagelos/genética , Flagelos/ultraestrutura , Glicoconjugados/genética , Glicoconjugados/metabolismo , Glicosilação , Guanosina Difosfato Fucose/genética , Guanosina Difosfato Manose/genética , Humanos , Proteínas de Protozoários/genética , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/ultraestrutura , Tripanossomíase Africana/enzimologia , Tripanossomíase Africana/genética
19.
Appl Microbiol Biotechnol ; 74(4): 768-75, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17111133

RESUMO

A recombinant Escherichia coli strain was developed to produce guanosine 5'-diphosphate (GDP)-L-fucose, donor of L-fucose, which is an essential substrate for the synthesis of fucosyloligosaccharides. GDP-D: -mannose-4, 6-dehydratase (GMD) and GDP-4-keto-6-deoxymannose 3, 5-epimerase 4-reductase (WcaG), the two crucial enzymes for the de novo GDP-L-fucose biosynthesis, were overexpressed in recombinant E. coli by constructing inducible overexpression vectors. Optimum expression conditions for GMD and WcaG in recombinant E. coli BL21(DE3) were 25 degrees C and 0.1 mM isopropyl-beta-D-thioglucopyranoside. Maximum GDP-L-fucose concentration of 38.9 +/- 0.6 mg l(-1) was obtained in a glucose-limited fed-batch cultivation, and it was enhanced further by co-expression of NADPH-regenerating glucose-6-phosphate dehydrogenase encoded by the zwf gene to achieve 55.2 +/- 0.5 mg l(-1) GDP-L-fucose under the same cultivation condition.


Assuntos
Escherichia coli/metabolismo , Guanosina Difosfato Fucose/biossíntese , Carboidratos Epimerases/biossíntese , Carboidratos Epimerases/genética , Escherichia coli/genética , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Expressão Gênica , Glucosefosfato Desidrogenase/biossíntese , Glucosefosfato Desidrogenase/genética , Hidroliases/genética , Microbiologia Industrial , Cetona Oxirredutases/biossíntese , Cetona Oxirredutases/genética , Complexos Multienzimáticos/biossíntese , Complexos Multienzimáticos/genética
20.
APMIS ; 114(7-8): 539-48, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16907860

RESUMO

L-fucose is a fundamental monosaccharide component of many mammalian glycoproteins and glycolipids. Fucosylation requires GDP-L-fucose as a donor of fucose and a specific fucosyltransferase (Fuc-T) to catalyze the transfer of L-fucose to various lactosamine acceptor molecules. The biosynthesis of GDP-L-fucose consists of two pathways. The constitutively active de novo pathway involves conversion of cellular GDP-D-mannose to GDP-L-fucose by GDP-D-mannose-4,6-dehydratase (GMD) and GDP-4-keto-6-deoxy-D-mannose-3,5-epimerase-4-reductase (FX). In the alternative biosynthetic pathway, in the salvage metabolism, L-fucokinase (Fuk) synthesizes L-fucose-1-phosphate from free fucose. L-fucose-1-phosphate is further catalyzed to GDP-L-fucose by GDP-L-fucose pyrophosphorylase (Fpgt). GDP-L-fucose, synthesized in the cytosol, is translocated to the Golgi for fucosylation by a specific GDP-fucose transporter (FUCT1). Glycans that contain alpha(1,3)-fucosylated modifications, e.g. sialyl Lewis X-type glycans, have an important role in inflammation and in tumorigenesis. We studied the mRNA expression levels of GDP-L-fucose-synthesizing enzymes, GDP-fucose transporter and fucosyltransferase VII by quantitative real-time PCR in mouse endothelial cells, macrophages and lymphoid tumor cells. Moreover, the expression of the same transcripts was detected in acute inflammation using rat kidney allograft as model system. Our results indicate the simultaneous upregulation of the GDP-L-fucose synthesizing enzymes of the de novo pathway, GDP-fucose transporter and fucosyltransferase VII in inflammation and in tumorigenesis.


Assuntos
Fucosiltransferases/genética , Expressão Gênica/fisiologia , Guanosina Difosfato Fucose/biossíntese , Proteínas de Transporte de Monossacarídeos/genética , Animais , Linhagem Celular , Fucosiltransferases/biossíntese , Rejeição de Enxerto/metabolismo , Transplante de Rim , Camundongos , Proteínas de Transporte de Monossacarídeos/biossíntese , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA