Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Biotechnol Bioeng ; 120(2): 524-535, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36326175

RESUMO

As one of the most abundant components in human milk oligosaccharides, 2'-fucosyllactose (2'-FL) possesses versatile beneficial health effects. Although most studies focused on overexpressing or fine-tuning the expression of pathway enzymes and achieved a striking increase of 2'-FL production, directly facilitating the metabolic flux toward the key intermediate GDP-l-fucose seems to be ignored. Here, multienzyme complexes consisting of sequential pathway enzymes were constructed by using specific peptide interaction motifs in recombinant Escherichia coli to achieve a higher titer of 2'-FL. Specifically, we first fine-tuned the expression level of pathway enzymes and balanced the metabolic flux toward 2'-FL synthesis. Then, two key enzymes (GDP-mannose 4,6-dehydratase and GDP- l-fucose synthase) were self-assembled into enzyme complexes in vivo via a short peptide interaction pair RIAD-RIDD (RI anchoring disruptor-RI dimer D/D domains), resulting in noticeable improvement of 2'-FL production. Next, to further strengthen the metabolic flux toward 2'-FL, three pathway enzymes were further aggregated into multienzyme assemblies by using another orthogonal protein interaction motif (Spycatcher-SpyTag or PDZ-PDZlig). Intracellular multienzyme assemblies remarkably enlarged the flux toward 2'-FL biosynthesis and showed a 2.1-fold increase of 2'-FL production compared with a strain expressing free-floating and unassembled enzymes. The optimally engineered strain EZJ23 accumulated 4.8 g/L 2'-FL in shake flask fermentation and was capable of producing 25.1 g/L 2'-FL by fed-batch cultivation. This work provides novel approaches for further improvement and large-scale production of 2'-FL and demonstrates the effectiveness of spatial assembly of pathway enzymes to improve the production of valuable products in the engineered host strain.


Assuntos
Escherichia coli , Fucose , Trissacarídeos , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Fucose/metabolismo , Guanosina Difosfato Fucose/metabolismo , Engenharia Metabólica/métodos , Complexos Multienzimáticos/metabolismo , Peptídeos/metabolismo , Trissacarídeos/biossíntese
2.
Bioengineered ; 13(3): 5551-5563, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35184647

RESUMO

Eriodictyol is a natural flavonoid with many pharmacological effects, such as anti-oxidation, anti-inflammation, anti-tumor, and neuroprotection. Besides, it has been reported that flavonoids play an important role in protein glycosylation. The fucosylation structure is closely associated with processes of various tumor metastases. TSTA3 is involved in the de novo synthesis and can convert cellular GDP-D-mannose into GDP-L-fucose. It was predicted on the STITCH database that eriodictyol interacted with TSTA3. In addition, literature has confirmed that TSTA3 is upregulated in CRC and can regulate the proliferation and migration of breast cancer cells. Herein, the precise effects of eriodictyol on the clone-forming, proliferative, migratory and invasive abilities of CRC cells as well as EMT process were assessed. Moreover, the correlation among eriodictyol, TSTA3, and fucosylation in these malignant behaviors of CRC cells was evaluated, in order to elucidate the underlying mechanism. The current work discovered that eriodictyol inhibited the viability, clone-formation, proliferation, migration, invasion, and EMT of CRC cells, and that these inhibitory effects of eriodictyol on the malignant behavior of CRC cells were reversed by TSTA3 overexpression. Additionally, eriodictyol suppresses fucosylation by downregulating the TSTA3 expression. Results confirmed that fucosylation inhibitor (2-F-Fuc) inhibited clone formation, proliferation, migration, invasion, as well as EMT of CRC cells and eriodictyol treatment further reinforced the suppressing effects of 2-F-Fuc on the malignant behavior of CRC cells. We conclude that eriodictyol suppresses the clone-forming, proliferative, migrative and invasive abilities of CRC cells as well as represses the EMT process by downregulating TSTA3 expression to restrain fucosylation.


Assuntos
Carboidratos Epimerases , Neoplasias Colorretais , Cetona Oxirredutases , Carboidratos Epimerases/antagonistas & inibidores , Carboidratos Epimerases/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/tratamento farmacológico , Transição Epitelial-Mesenquimal , Flavanonas , Glicosilação , Guanosina Difosfato Fucose/metabolismo , Guanosina Difosfato Fucose/farmacologia , Humanos , Cetona Oxirredutases/antagonistas & inibidores , Cetona Oxirredutases/metabolismo
3.
Theranostics ; 10(24): 11339-11358, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042286

RESUMO

Background: TSTA3 gene encodes an enzyme responsible for synthesis of GDP-L-fucose as the only donor in fucosylation. This study was designed to explore clinical value, function and underlying mechanism of TSTA3 in the development of esophageal squamous cell carcinoma (ESCC). Methods: Whole genomic sequencing data from 663 ESCC patients and RNA sequencing data from 155 ESCC patients were used to analyze the copy number variation and mRNA expression of TSTA3 respectively. Immunohistochemistry based or not based on the tissue microarrays was used to detect its protein expression. Transwell assay and in vivo metastasis assay were used to study the effect of TSTA3 on invasion and metastasis of ESCC. Immunofluorescence was used to analyze fucosylation level. N-glycoproteomics and proteomics analysis, Lens Culinaris Agglutinin (LCA) and Ulex Europaeus Agglutinin I (UEA-I) affinity chromatography, immunoprecipitation, glycosyltransferase activity kit and rescue assay were used to explore the mechanism of TSTA3. Results: TSTA3 was frequently amplified and overexpressed in ESCC. TSTA3 amplification and protein overexpression were significantly associated with malignant progression and poor prognosis of ESCC patients. TSTA3 knockdown significantly suppressed ESCC cells invasion and tumor dissemination by decreasing fucosylation level. Conversely, exogenous overexpression of TSTA3 led to increased invasion and tumor metastasis in vitro and in vivo by increasing fucosylation level. Moreover, core fucosylated LAMP2 and terminal fucosylated ERBB2 might be mediators of TSTA3-induced pro-invasion in ESCC and had a synergistic effect on the process. Peracetylated 2-F-Fuc, a fucosyltransferase activity inhibitor, reduced TSTA3 expression and fucosylation modification of LAMP2 and ERBB2, thereby inhibiting ESCC cell invasion. Conclusion: Our results indicate that TSTA3 may be a driver of ESCC metastasis through regulating fucosylation of LAMP2 and ERBB2. Fucosylation inhibitor may have prospect to suppress ESCC metastasis by blocking aberrant fucosylation.


Assuntos
Carboidratos Epimerases/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/secundário , Cetona Oxirredutases/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Receptor ErbB-2/metabolismo , Idoso , Animais , Carboidratos Epimerases/genética , Linhagem Celular Tumoral , Proliferação de Células , Variações do Número de Cópias de DNA , Progressão da Doença , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidade , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/mortalidade , Esôfago/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glicosilação , Guanosina Difosfato Fucose/metabolismo , Humanos , Cetona Oxirredutases/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Prognóstico , Sequenciamento Completo do Genoma , Ensaios Antitumorais Modelo de Xenoenxerto
4.
World J Gastroenterol ; 25(23): 2947-2960, 2019 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-31249452

RESUMO

BACKGROUND: Changes in N-linked glycosylation have been observed in the circulation of individuals with hepatocellular carcinoma. In particular, an elevation in the level of core fucosylation has been observed. However, the mechanisms through which core fucose is increased are not well understood. We hypothesized that a review of the literature and related bioinformatic review regarding six genes known to be involved in the attachment of core fucosylation, the synthesis of the fucosylation substrate guanosine diphosphate (GDP)-fucose, or the transport of the substrate into the Golgi might offer mechanistic insight into the regulation of core fucose levels. AIM: To survey the literature to capture the involvement of genes regulating core N-linked fucosylation in hepatocellular carcinoma. METHODS: The PubMed biomedical literature database was searched for the association of hepatocellular carcinoma and each of the core fucose-related genes and their protein products. We also queried The Cancer Genome Atlas Liver hepatocellular carcinoma (LIHC) dataset for genetic, epigenetic and gene expression changes for the set of six genes using the tools at cBioportal. RESULTS: A total of 27 citations involving one or more of the core fucosylation-related genes (FPGT, FUK, FUT8, GMDS, SLC35C1, TSTA3) and hepatocellular carcinoma were identified. The same set of gene symbols was used to query the 371 patients with liver cancer in the LIHC dataset to identify the frequency of mRNA over or under expression, as well as non-synonymous mutations, copy number variation and methylation level. Although all six genes trended to more samples displaying over expression relative to under-expression, it was noted that a number of tumor samples had undergone amplification of the genes of the de novo synthesis pathway, GMDS (27 samples) and TSTA3 (78 samples). In contrast, the other four genes had undergone amplification in 2 or fewer samples. CONCLUSION: Amplification of genes involved in the de novo pathway for generation of GDP-fucose, GMDS and TSTA3, likely contributes to the elevated core fucose observed in hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Redes e Vias Metabólicas/genética , Carboidratos Epimerases/metabolismo , Carcinoma Hepatocelular/patologia , Variações do Número de Cópias de DNA , Metilação de DNA , Glicoproteínas/metabolismo , Glicosilação , Guanosina Difosfato Fucose/metabolismo , Humanos , Hidroliases/metabolismo , Cetona Oxirredutases/metabolismo , Neoplasias Hepáticas/patologia , Mutação
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.
Histopathology ; 63(2): 174-86, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23730929

RESUMO

AIMS: Fucosylation is regulated by fucosyltransferases, the guanosine diphosphate-L-fucose (GDP-L-Fuc) synthetic pathway, and the GDP-L-fucose transporter (GDP-L-Fuc Tr). We have reported previously an increased level of α(1,6)fucosyltransferase activity and expression in colorectal cancer (CRC). The present study aimed to analyse the expression profiles of the FX enzyme and GDP-L-Fuc Tr in a cohort of operated CRC patients to elucidate their role in α(1,6)fucosylation in this neoplasm. METHODS AND RESULTS: We assessed the immunohistochemical expression of FX and GDP-L-Fuc Tr in a series of tumour samples and healthy tissues from CRC specimens. FX expression was observed in 58 of 91 (63.7%) tumours and 23 of 28 (82.1%) corresponding healthy samples. GDP-L-Fuc Tr expression was detected in 86 of 102 (84.3%) colorectal tumours, and 13 of 27 (48.1%) healthy tissue specimens. The expression of GDP-L-Fuc Tr was statistically higher in tumours than in healthy tissues (P < 0.001). A correlation was found between FX and GDP-L-Fuc Tr expression in tumour samples (P = 0.003). CONCLUSION: GDP-L-Fuc Tr overexpression in the tumour tissue of CRC patients suggests that GDP-L-Fuc transport to the Golgi apparatus may be an important factor associated with increased α(1,6)fucosylation in CRC.


Assuntos
Carboidratos Epimerases/metabolismo , Neoplasias Colorretais/metabolismo , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/metabolismo , Cetona Oxirredutases/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Idoso , Neoplasias Colorretais/patologia , Intervalo Livre de Doença , Feminino , Glicosilação , Complexo de Golgi/metabolismo , Humanos , Hidroliases/metabolismo , Imuno-Histoquímica , Masculino , Prognóstico
7.
Proc Natl Acad Sci U S A ; 110(14): 5404-9, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23493549

RESUMO

The key role played by fucose in glycoprotein and cellular function has prompted significant research toward identifying recombinant and biochemical strategies for blocking its incorporation into proteins and membrane structures. Technologies surrounding engineered cell lines have evolved for the inhibition of in vitro fucosylation, but they are not applicable for in vivo use and drug development. To address this, we screened a panel of fucose analogues and identified 2-fluorofucose and 5-alkynylfucose derivatives that depleted cells of GDP-fucose, the substrate used by fucosyltransferases to incorporate fucose into protein and cellular glycans. The inhibitors were used in vitro to generate fucose-deficient antibodies with enhanced antibody-dependent cellular cytotoxicity activities. When given orally to mice, 2-fluorofucose inhibited fucosylation of endogenously produced antibodies, tumor xenograft membranes, and neutrophil adhesion glycans. We show that oral 2-fluorofucose treatment afforded complete protection from tumor engraftment in a syngeneic tumor vaccine model, inhibited neutrophil extravasation, and delayed the outgrowth of tumor xenografts in immune-deficient mice. The results point to several potential therapeutic applications for molecules that selectively block the endogenous generation of fucosylated glycan structures.


Assuntos
Anticorpos Monoclonais/metabolismo , Vacinas Anticâncer/farmacologia , Fucose/farmacologia , Fucosiltransferases/antagonistas & inibidores , Guanosina Difosfato Fucose/metabolismo , Polissacarídeos/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Cromatografia Líquida , Cricetinae , Cricetulus , Desenho de Fármacos , Feminino , Fucose/química , Humanos , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Neutrófilos/metabolismo
8.
Biochim Biophys Acta ; 1820(12): 1915-25, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22982178

RESUMO

BACKGROUND: Fucosylation is essential for various biological processes including tumorigenesis, inflammation, cell-cell recognition and host-pathogen interactions. Biosynthesis of fucosylated glycans is accomplished by fucosyltransferases. The enzymatic product of core α1,6-fucosyltransferase (FUT8) plays a major role in a plethora of pathological conditions, e.g. in prognosis of hepatocellular carcinoma and in colon cancer. Detailed knowledge of the binding mode of its substrates is required for the design of molecules that can modulate the activity of the enzyme. METHODS: We provide a detailed description of binding interactions of human FUT8 with its natural donor substrate GDP-fucose and related compounds. GDP-Fuc was placed in FUT8 by structural analogy to the structure of protein-O-fucosyltransferase (cePOFUT) co-crystallized with GDP-Fuc. The epitope of the donor substrate bound to FUT8 was determined by STD NMR. The in silico model is further supported by experimental data from SPR binding assays. The complex was optimized by molecular dynamics simulations. RESULTS: Guanine is specifically recognized by His363 and Asp453. Furthermore, the pyrophosphate is tightly bound via numerous hydrogen bonds and contributes affinity to a major part. Arg365 was found to bind both the ß-phosphate and the fucose moiety at the same time. CONCLUSIONS: Discovery of a novel structural analogy between cePOFUT and FUT8 allows the placement of the donor substrate GDP-Fuc. The positioning was confirmed by various experimental and computational techniques. GENERAL SIGNIFICANCE: The model illustrates details of the molecular basis of substrate recognition for a human fucosyltransferase for the first time and, thus, provides a basis for structure-based design of inhibitors.


Assuntos
Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/metabolismo , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Catálise , Fucosiltransferases/química , Fucosiltransferases/genética , Fucosiltransferases/isolamento & purificação , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica , Ressonância de Plasmônio de Superfície
9.
Biol. Res ; 44(1): 25-34, 2011. ilus
Artigo em Inglês | LILACS | ID: lil-591861

RESUMO

The Notch signaling pathway plays an important role in development and physiology. In Drosophila, Notch is activated by its Delta or Serrate ligands, depending in part on the sugar modifications present in its extracellular domain. O-fucosyltransferase-1 (OFUT1) performs the first glycosylation step in this process, O-fucosylating various EGF repeats at the Notch extracellular domain. Besides its O-fucosyltransferase activity, OFUT1 also behaves as a chaperone during Notch synthesis and is able to down regulate Notch by enhancing its endocytosis and degradation. We have reevaluated the roles that O-fucosylation and the synthesis of GDP-fucose play in the regulation of Notch protein stability. Using mutants and the UAS/Gal4 system, we modified in developing tissues the amount of GDP-mannose-deshydratase (GMD), the first enzyme in the synthesis of GDP-fucose. Our results show that GMD activity, and likely the levels of GDP-fucose and O-fucosylation, are essential to stabilize the Notch protein. Notch degradation observed under low GMD expression is absolutely dependent on OFUT1 and this is also observed in Notch Abruptex mutants, which have mutations in some potential O-fucosylated EGF domains. We propose that the GDP-fucose/OFUT1 balance determines the ability of OFUT1 to endocytose and degrade Notch in a manner that is independent of the residues affected by Abruptex mutations in Notch EGF domains.


Assuntos
Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Fucosiltransferases/metabolismo , Guanosina Difosfato Fucose/metabolismo , Guanosina Difosfato Manose/metabolismo , Receptores Notch/metabolismo , Asas de Animais/metabolismo , Alelos , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/metabolismo , Endocitose/genética , Fucosiltransferases/genética , Guanosina Difosfato Fucose/genética , Guanosina Difosfato Manose/genética , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação/genética , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Notch/genética , Transdução de Sinais , Asas de Animais/anatomia & histologia
10.
Proc Natl Acad Sci U S A ; 102(51): 18532-7, 2005 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-16344471

RESUMO

Congenital disorder of glycosylation IIc (CDG IIc), also termed leukocyte adhesion deficiency II, is a recessive syndrome characterized by slowed growth, mental retardation, and severe immunodeficiency. Recently, the gene responsible for CDG IIc was found to encode a GDP-fucose transporter. Here, we investigated the possible cause of the developmental defects in CDG IIc patients by using a Drosophila model. Biochemically, we demonstrated that a Drosophila homolog of the GDP-fucose transporter, the Golgi GDP-fucose transporter (Gfr), specifically transports GDP-fucose in vitro. To understand the function of the Gfr gene, we generated null mutants of Gfr in Drosophila. The phenotypes of the Drosophila Gfr mutants were rescued by the human GDP-fucose transporter transgene. Our phenotype analyses revealed that Notch (N) signaling was deficient in these Gfr mutants. GDP-fucose is known to be essential for the fucosylation of N-linked glycans and for O-fucosylation, and both fucose modifications are present on N. Our results suggest that Gfr is involved in the fucosylation of N-linked glycans on N and its O-fucosylation, as well as those of bulk proteins. However, despite the essential role of N O-fucosylation during development, the Gfr homozygote was viable. Thus, our results also indicate that the Drosophila genome encodes at least another GDP-fucose transporter that is involved in the O-fucosylation of N. Finally, we found that mammalian Gfr is required for N signaling in mammalian cultured cells. Therefore, our results implicate reduced N signaling in the pathology of CDG IIc.


Assuntos
Proteínas de Drosophila/deficiência , Drosophila melanogaster/metabolismo , Síndrome da Aderência Leucocítica Deficitária/metabolismo , Receptores Notch/deficiência , Animais , Linhagem Celular Tumoral , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Fucose/metabolismo , Glicosilação , Complexo de Golgi/metabolismo , Guanosina Difosfato Fucose/metabolismo , Humanos , Síndrome da Aderência Leucocítica Deficitária/genética , Ligantes , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Mutação/genética , Fenótipo , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais
11.
Glycobiology ; 14(12): 1285-94, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15269183

RESUMO

The de novo synthesis and expression of sulfo sLex glycan on vascular endothelial glycoproteins has a central role in the initiation of inflammatory reactions, serving as a putative ZIP code for organ-specific trafficking of leukocytes into sites of inflammation. The synthesis of sulfo sLex requires energy carrying donors, CMP-sialic acid (CMP-SA), GDP-fucose (GDP-Fuc), and adenosine 3'-phosphate 5'-phosphosulphate (PAPS) for donation of SA, Fuc, and sulfate, respectively. These donors are synthesized in the nucleus or cytosol and translocated into Golgi by specific transporters where corresponding transferase and proteins as well as enzymatic activities increase on inflammatory stimuli. Here we analyze the transcriptional coregulation of CMP-SA, GDP-Fuc, and PAPS transporters with in situ hybridization and real-time PCR in acute inflammation using kidney and heart allografts as model systems. Our results indicate that these three transporters display coordinated transcriptional regulation during the induction of the sulfo sLex glycan biosynthesis. With in silico analysis, the data generated with 230 human Affymetrix U133A gene chips indicated that the coregulated expression of CMP-SA and GDP-Fuc transporters was not common. Taken together our results suggest that inflammation-induced transcriptional regulation exists for Golgi membrane transporters required for the synthesis of the inflammation-inducible ZIP code sulfo sLex glycans.


Assuntos
Proteínas de Transporte/metabolismo , Epitopos/biossíntese , Complexo de Golgi/metabolismo , Inflamação , Oligossacarídeos/biossíntese , Transcrição Gênica/fisiologia , Animais , Ácido N-Acetilneuramínico do Monofosfato de Citidina/metabolismo , Epitopos/genética , Guanosina Difosfato Fucose/metabolismo , Humanos , Hibridização In Situ , Antígenos CD15/análogos & derivados , Oligossacarídeos/genética , Fosfoadenosina Fosfossulfato/metabolismo , Ratos , Ratos Endogâmicos , Antígeno Sialil Lewis X/análogos & derivados
12.
Anal Biochem ; 310(1): 100-6, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12413479

RESUMO

To investigate the biological significance of GDP-L-fucose, we established a unique method for the determination of GDP-L-fucose levels in microsomal fractions, using an HPLC assay of alpha 1-6-fucosyltransferase (alpha1-6-FucT), an enzyme that catalyzes the synthesis of core fucosylation in N-glycans. A microsomal protein and a large excess of fluorescence-labeled synthetic oligosaccharide (a substrate) were incubated with a large excess of alpha1-6-FucT. The fluorescent intensity of the fucosylated reaction product, which was analyzed by isocratic reverse phase HPLC, was proportional to the level of GDP-L-fucose in the microsomal fractions over the range 0.20-10 pmol. This assay is applicable to the determination of the GDP-L-fucose content in various cancer cell lines as well as rat liver and would be useful in developing a better understanding of the fucosylation potential of such cells and tissues.


Assuntos
Guanosina Difosfato Fucose/análise , Guanosina Difosfato Fucose/metabolismo , Aminopiridinas/química , Aminopiridinas/metabolismo , Animais , Sequência de Carboidratos , Cromatografia Líquida de Alta Pressão/métodos , Neoplasias do Colo/metabolismo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Fucosiltransferases/metabolismo , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Microssomos/química , Microssomos/metabolismo , Dados de Sequência Molecular , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Ratos , Espectrometria de Fluorescência , Fatores de Tempo , Células Tumorais Cultivadas
13.
J Biol Chem ; 273(46): 30165-74, 1998 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-9804772

RESUMO

The enzyme that catalyzes the formation of GDP-L-fucose from GTP and beta-L-fucose-1-phosphate (i.e. GDP-beta-L-fucose pyrophosphorylase, GFPP) was purified about 560-fold from the cytosolic fraction of pig kidney. At this stage, there were still a number of protein bands on SDS gels, but only the 61-kDa band became specifically labeled with the photoaffinity substrate, azido-GDP-L-[32P]fucose. Several peptides from this 61-kDa band were sequenced and these sequences were used for cloning the gene. The cDNA clone yielded high levels of GFPP activity when expressed in myeloma cells and in a baculovirus system, demonstrating that the 61-kDa band is the authentic GFPP. The porcine tissue with highest specific activity for GFPP was kidney, with lung, liver, and pancreas being somewhat lower. GFPP was also found in Chinese hamster ovary, but not Madin-Darby canine kidney cells. Northern analysis showed the mRNA in human spleen, prostate, testis, ovary, small intestine, and colon. GFPP was stable at 4 (o)C in buffer containing 50 mM sucrose, with little loss of activity over a 9-day period. GTP was the best nucleoside triphosphate substrate but significant activity was also observed with ITP and to a lesser extent with ATP. The enzyme was reasonably specific for beta-L-fucose-1-P, but could also utilize alpha-D-arabinose-1-P to produce GDP-alpha-D-arabinose. The product of the reaction with GTP and alpha-L-fucose-1-P was characterized as GDP-beta-L-fucose by a variety of chemical and chromatographic methods.


Assuntos
Nucleotidiltransferases/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Catálise , Cricetinae , Citosol/enzimologia , Bases de Dados Factuais , Cães , Fucose/análogos & derivados , Fucose/metabolismo , Guanosina Difosfato Fucose/metabolismo , Guanosina Trifosfato/metabolismo , Hexosefosfatos/metabolismo , Humanos , Rim/enzimologia , Dados de Sequência Molecular , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Regiões Promotoras Genéticas , Especificidade por Substrato , Suínos
14.
Biochimie ; 80(11): 923-31, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9893952

RESUMO

L-fucose and L-rhamnose are two 6-deoxyhexoses naturally occurring in several complex carbohydrates. In prokaryotes both of them are found in polysaccharides of the cell wall, while in animals only L-fucose has been described, which mainly participates to the structure of glycoconjugates, either in the cell membrane or secreted in biological fluids, such as ABH blood groups and Lewis system antigens. L-fucose and L-rhamnose are synthesized by two de novo biosynthetic pathways starting from GDP-D-mannose and dTDP-D-glucose, respectively, which share several common features. The first step for both pathways is a dehydration reaction catalyzed by specific nucleotide-sugar dehydratases. This leads to the formation of unstable 4-keto-6-deoxy intermediates, which undergo a subsequent epimerization reaction responsible for the change from D- to L-conformation, and then a NADPH-dependent reduction of the 4-keto group, with the consequent formation of either GDP-L-fucose or dTDP-L-rhamnose. These compounds are then the substrates of specific glycosyltransferases which are responsible for insertion of either L-fucose or L-rhamnose in the corresponding glycoconjugates. The enzyme involved in the first step of GDP-L-fucose biosynthesis in E. coli, i.e., GDP-D-mannose 4,6 dehydratase, has been recently expressed as recombinant protein and characterized in our laboratory. We have also cloned and fully characterized a human protein, formerly named FX, and an E. coli protein, WcaG, which display both the epimerase and the reductase activities, thus indicating that only two enzymes are required for GDP-L-fucose production. Fucosylated complex glycoconjugates at the cell surface can then be recognized by specific counter-receptors in interacting cells, these mechanisms initiating important processes including inflammation and metastasis. The second pathway starting from dTDP-D-glucose leads to the synthesis of antibiotic glycosides or, alternatively, to the production of dTDP-L-rhamnose. While several sets of data are available on the first enzyme of the pathway, i.e., dTDP-D-glucose dehydratase, the enzymes involved in the following steps still need to be identified and characterized.


Assuntos
Fucose/metabolismo , Ramnose/metabolismo , Animais , Carboidratos Epimerases/metabolismo , Fucose/biossíntese , Guanosina Difosfato Fucose/metabolismo , Humanos , Hidroliases , Modelos Químicos , Ramnose/biossíntese
15.
J Biol Chem ; 272(35): 21994-8, 1997 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-9268337

RESUMO

The Lewis alpha(1,3/1,4)-fucosyltransferase, Fuc-TIII, encoded by the FUT3 gene is responsible for the final synthesis of Lea and Leb antigens. Various point mutations have been described explaining the Lewis negative phenotype, Le(a-b-), on erythrocytes and secretions. Two of these, T202C and C314T originally described in a Swedish population, have not been found as single isolated point mutations so far. To define the relative contribution of each of these two mutations to the Lewis negative phenotype, we cloned and made chimeric FUT3 constructs separating the T202C mutation responsible for the amino acid change Trp68 --> Arg, from the C314T mutation leading to the Thr105 --> Met shift. COS-7 cells were transfected and the expression of Fuc-TIII enzyme activity and the presence of Lewis antigens were determined. There was no decrease in enzyme activity nor of immunofluorescence staining on cells transfected with the construct containing the isolated C314T mutation compared with cells transfected with a wild type FUT3 allele control. No enzyme activity nor immunoreactivity for Lewis antigens was detected in FUT3 constructs containing both mutations in combination. The T202C mutation alone decreased the enzyme activity to less than 1% of the activity of the wild type FUT3 allele. These results demonstrate, that the Trp68 --> Arg substitution in human Fuc-TIII is the capital amino acid change responsible for the appearance of the Le(a-b-) phenotype on human erythrocytes in individuals homozygous for both the T202C and C314T mutations.


Assuntos
Fucosiltransferases/genética , Antígenos do Grupo Sanguíneo de Lewis/genética , Mutação Puntual , Alelos , Animais , Arginina/genética , Células COS , Clonagem Molecular , Fucosiltransferases/metabolismo , Expressão Gênica , Guanosina Difosfato Fucose/metabolismo , Humanos , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Triptofano/genética
16.
Carbohydr Res ; 305(3-4): 511-5, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9648268

RESUMO

The Lewis (alpha 1-3/4) fucosyltransferase isolated from human milk could be used for preparative fucosylations of the disaccharide acceptors Gal(beta 1-3)GlcNAc(beta 1-O)R (at position OH-4) and Gal(beta 1-4)GlcNAc(beta 1-O)R (at position OH-3) [R = (CH2)8COOMe]. As donors GDP-L-Gal and deoxygenated derivatives were used to lead to a series of novel modified trisaccharides of the Lewis(a) and the Lewis(x) type, respectively.


Assuntos
Fucosiltransferases/metabolismo , Galactosídeos/síntese química , Antígenos do Grupo Sanguíneo de Lewis/química , Trissacarídeos/síntese química , Animais , Sequência de Carboidratos , Dissacarídeos/metabolismo , Guanosina Difosfato Fucose/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Leite/enzimologia , Dados de Sequência Molecular , Especificidade por Substrato
17.
J Biol Chem ; 271(44): 27274-9, 1996 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8910301

RESUMO

FX is a homodimeric NADP(H)-binding protein of 68 kDa, first identified in human erythrocytes, from which it was purified to homogeneity. Its function has been unrecognized despite partial structural and genetic characterization. Recently, on the basis of partial amino acid sequence, it proved to be the human homolog of the murine protein P35B, a tumor rejection antigen. In order to address the biochemical role of FX, its primary structure was completed by cDNA sequencing. This sequence revealed a significant homology with many proteins from different organisms. Specifically, FX showed a remarkable similarity with a putative Escherichia coli protein, named Yefb, whose gene maps in a region of E. coli chromosome coding for enzymes involved in synthesis and utilization of GDP-D-mannose. Accordingly, a possible role of FX in this metabolism was investigated. The data obtained indicate FX as the enzyme responsible for the last step of the major metabolic pathway resulting in GDP-L-fucose synthesis from GDP-D-mannose in procaryotic and eucaryotic cells. Specifically, purified FX apparently catalyzes a combined epimerase and NADPH-dependent reductase reaction, converting GDP-4-keto-6-D-deoxymannose to GDP-L-fucose. This is the substrate of several fucosyltranferases involved in the correct expression of many glyconjugates, including blood groups and developmental antigens.


Assuntos
Carboidratos Epimerases , Proteínas de Transporte/metabolismo , Eritrócitos/metabolismo , Guanosina Difosfato Fucose/metabolismo , Cetona Oxirredutases , Sequência de Aminoácidos , Animais , Antígenos de Superfície/química , Sequência de Bases , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Caenorhabditis elegans , Carcinoma Hepatocelular , Proteínas de Transporte/biossíntese , Proteínas de Transporte/química , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Citosol/metabolismo , DNA Complementar , Escherichia coli , Guanosina Difosfato Manose/metabolismo , Humanos , Neoplasias Hepáticas , Camundongos , Dados de Sequência Molecular , Estrutura Molecular , Proteínas do Tecido Nervoso/química , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos , Sintaxina 1
18.
FEBS Lett ; 395(1): 68-72, 1996 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-8849691

RESUMO

A fucosyltransferase activity has been detected using lacto-N-biose I as acceptor in the lower eukaryote Dictyostelium discoideum. This transferase requires divalent cations and is inhibited by N-ethylmaleimide and detergent treatment. Apparent calculated Km values for GDP-Fuc and lacto-N-biose I are 1.27 microM and 2.80 mM, respectively. The activity is quantitatively recovered in the supernatant after centrifugation at 100000 x g for 1 h. The reaction product, as determined by gel permeation chromatography, sensitivity to fucosidases, and analysis of partially methylated derivatives, is Fucalpha1-2Galbeta1-3GlcNAc (H type 1 trisaccharide).


Assuntos
Citosol/enzimologia , Dictyostelium/enzimologia , Fucosiltransferases/metabolismo , Trissacarídeos/biossíntese , Acetilglucosamina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Sequência de Carboidratos , Cátions Bivalentes , Fracionamento Celular , Quelantes/farmacologia , Ácido Edético/farmacologia , Inibidores Enzimáticos/farmacologia , Etilmaleimida/farmacologia , Fucose/farmacologia , Fucosiltransferases/análise , Fucosiltransferases/antagonistas & inibidores , Guanosina Difosfato Fucose/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Magnésio/farmacologia , Manganês/farmacologia , Metilação , Dados de Sequência Molecular , Temperatura , Galactosídeo 2-alfa-L-Fucosiltransferase
19.
Arch Biochem Biophys ; 296(2): 562-8, 1992 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-1321590

RESUMO

The NCI-H69 cell alpha 1----3fucosyltransferase has been purified from a 0.2% Triton X-100R solubilized enzyme fraction by GDP-hexanolamine-Sepharose affinity chromatography and Superose 12 gel filtration. Photoaffinity labeling experiments with 125I-GDP-hexanolaminyl-4-azidosalicylic acid present in concentrations equivalent to 0.5 and 1 times Ki of the inhibitor for the enzyme indicated that labeling of the 45-kDa protein band could be inhibited by addition of 400 microM GDP-fucose but was not effected by similar concentrations of either GDP-mannose or GDP-glucose. The purified enzyme was applied to studies intended to define catalytically essential amino acid residues of the protein. Incubation of the enzyme in the presence of increasing concentrations of pyridoxal 5'-phosphate was found to result in irreversible inactivation of the enzyme after NaBH4 reduction. The donor substrate, GDP-fucose, was found to protect the enzyme from inactivation. Little or no protection was found for either GDP-mannose or the acceptor substrate nLc4. Pyridoxal 5'-phosphate was shown to behave as a competitive inhibitor with respect to GDP-fucose with a Ki of 105 microM. Labeling with 3H-pyridoxal 5'-phosphate resulted in the incorporation of approximately 8 mol pyridoxal 5'-phosphate per mole subunit. Parallel experiments containing GDP-fucose indicated protection of one site per subunit correlated with GDP-fucose binding. Acid hydrolysis and chromatographic analysis of the 3H-pyridoxylated protein indicated greater than 95% of the 3H label was recovered as pyridoxyl-lysine irrespective of whether GDP-fucose was present or not during labeling. These studies indicate the presence of a catalytically essential lysine residue associated with GDP-fucose binding to this enzyme. This information will be of value in further studies of this and other alpha 1----3fucosyltransferases and may suggest a practical basis for modulation of enzyme activity in the cell.


Assuntos
Carcinoma de Células Pequenas/enzimologia , Fucosiltransferases/antagonistas & inibidores , Guanosina Difosfato Fucose/farmacologia , Neoplasias Pulmonares/enzimologia , Lisina/química , Fosfato de Piridoxal/farmacologia , Sítios de Ligação , Ligação Competitiva , Boroidretos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Fucosiltransferases/química , Guanosina Difosfato Fucose/metabolismo , Humanos , Oxirredução , Fosfato de Piridoxal/metabolismo , Células Tumorais Cultivadas
20.
Proc Natl Acad Sci U S A ; 81(22): 7051-5, 1984 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-6095266

RESUMO

The mechanism of translocation in vitro of sugar nucleotides and adenosine 3'-phosphate 5'-phosphosulfate (PAPS) into the lumen of rat liver Golgi apparatus vesicles has been studied. It has been previously shown that the Golgi apparatus membrane has specific carrier proteins for PAPS and sugar nucleotides. We now report that translocation of the above nucleotide derivatives across Golgi membranes occurs via a coupled equimolar exchange with the corresponding nucleoside monophosphates. An initial incubation of Golgi vesicles with GDP-fucose radiolabeled in the guanidine ring resulted in accumulation within the lumen of radiolabeled GMP. Exit of GMP from these vesicles was specifically dependent on the entry of (additional) GDP-fucose into the vesicles (GDP-mannose and other sugar nucleotides had no effect). GDP-fucose-stimulated exit of GMP was temperature dependent, was blocked by inhibitors of GDP-fucose transport, such as 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid, and appeared to be equimolar with GDP-fucose entry. Preliminary evidence for specific, equimolar exchange of CMP-N-acetylneuraminic acid with CMP, PAPS with 3'-AMP, and UDP-galactose and UDP-N-acetylglucosamine with UMP was also obtained. These results strongly suggest the existence of different antiport proteins within the Golgi membrane that mediate the 1:1 exchange of sugar nucleotides or PAPS with the corresponding nucleoside monophosphate. Such proteins may have a regulatory role in glycosylation and sulfation reactions within the Golgi apparatus.


Assuntos
Nucleotídeos de Adenina/metabolismo , Ácido N-Acetilneuramínico do Monofosfato de Citidina/metabolismo , Complexo de Golgi/metabolismo , Guanosina Difosfato Fucose/metabolismo , Açúcares de Nucleosídeo Difosfato/metabolismo , Fosfoadenosina Fosfossulfato/metabolismo , Ácidos Siálicos/metabolismo , Monofosfato de Adenosina/metabolismo , Animais , Transporte Biológico , Monofosfato de Citidina/metabolismo , Guanosina Monofosfato/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA