Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Metab Eng ; 74: 61-71, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36152932

RESUMEN

Glycosylation of recombinant therapeutics like monoclonal antibodies (mAbs) is a critical quality attribute. N-glycans in mAbs are known to affect various effector functions, and thereby therapeutic use of such glycoproteins can depend on a particular glycoform profile to achieve desired efficacy. However, there are currently limited options for modulating the glycoform profile, which depend mainly on over-expression or knock-out of glycosyltransferase enzymes that can introduce or eliminate specific glycans but do not allow predictable glycoform modulation over a range of values. In this study, we demonstrate the ability to predictably modulate the glycoform profile of recombinant IgG. Using CRISPR/Cas9, we have engineered nucleotide sugar synthesis pathways in CHO cells expressing recombinant IgG for combinatorial modulation of galactosylation and fucosylation. Knocking out the enzymes UDP-galactose 4'-epimerase (Gale) and GDP-L-fucose synthase (Fx) resulted in ablation of de novo synthesis of UDP-Gal and GDP-Fuc. With Gale knock-out, the array of N-glycans on recombinantly expressed IgG is narrowed to agalactosylated glycans, mainly A2F glycan (89%). In the Gale and Fx double knock-out cell line, agalactosylated and afucosylated A2 glycan is predominant (88%). In the double knock-out cell line, galactosylation and fucosylation was entirely dependent on the salvage pathway, which allowed for modulation of UDP-Gal and GDP-Fuc synthesis and intracellular nucleotide sugar availability by controlling the availability of extracellular galactose and fucose. We demonstrate that the glycoform profile of recombinant IgG can be modulated from containing predominantly agalactosylated and afucosylated glycans to up to 42% and 96% galactosylation and fucosylation, respectively, by extracellular feeding of sugars in a dose-dependent manner. By simply varying the availability of extracellular galactose and/or fucose, galactosylation and fucosylation levels can be simultaneously and independently modulated. In addition to achieving the production of tailored glycoforms, this engineered CHO host platform can cater to the rapid synthesis of variably glycoengineered proteins for evaluation of biological activity.


Asunto(s)
Fucosa , Galactosa , Cricetinae , Animales , Células CHO , Cricetulus , Glicosilación , Fucosa/genética , Fucosa/metabolismo , Galactosa/genética , Galactosa/metabolismo , Polisacáridos/genética , Anticuerpos Monoclonales/genética , Inmunoglobulina G , Nucleótidos/metabolismo , Uridina Difosfato/metabolismo
2.
J Biomed Sci ; 29(1): 105, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36517806

RESUMEN

BACKGROUND: In endothelial cells, phospholipase C (PLC) ß1-activated Ca2+ is a crucial second messenger for the signaling pathways governing angiogenesis. PLCß1 is inactivated by complexing with an intracellular protein called translin-associated factor X (TRAX). This study demonstrates specific interactions between Globo H ceramide (GHCer) and TRAX, which highlight a new angiogenic control through PLCß1 activation. METHODS: Globo-series glycosphingolipids (GSLs), including GHCer and stage-specific embryonic antigen-3 ceramide (SSEA3Cer), were analyzed using enzyme-linked immunosorbent assay (ELISA) and Biacore for their binding with TRAX. Angiogenic activities of GSLs in human umbilical vein endothelial cells (HUVECs) were evaluated. Molecular dynamics (MD) simulation was used to study conformations of GSLs and their molecular interactions with TRAX. Fluorescence resonance energy transfer (FRET) analysis of HUVECs by confocal microscopy was used to validate the release of PLCß1 from TRAX. Furthermore, the in vivo angiogenic activity of extracellular vesicles (EVs) containing GHCer was confirmed using subcutaneous Matrigel plug assay in mice. RESULTS: The results of ELISA and Biacore analysis showed a stable complex between recombinant TRAX and synthetic GHCer with KD of 40.9 nM. In contrast, SSEA3Cer lacking a fucose residue of GHCer at the terminal showed ~ 1000-fold decrease in the binding affinity. These results were consistent with their angiogenic activities in HUVECs. The MD simulation indicated that TRAX interacted with the glycan moiety of GHCer at amino acid Q223, Q219, L142, S141, and E216. At equilibrium the stable complex maintained 4.6 ± 1.3 H-bonds. TRAX containing double mutations with Q223A and Q219A lost its ability to interact with GHCer in both MD simulation and Biacore assays. Removal of the terminal fucose from GHCer to become SSEA3Cer resulted in decreased H-bonding to 1.2 ± 1.0 by the MD simulation. Such specific H-bonding was due to the conformational alteration in the whole glycan which was affected by the presence or absence of the fucose moiety. In addition, ELISA, Biacore, and in-cell FRET assays confirmed the competition between GHCer and PLCß1 for binding to TRAX. Furthermore, the Matrigel plug assay showed robust vessel formation in the plug containing tumor-secreted EVs or synthetic GHCer, but not in the plug with SSEA3Cer. The FRET analysis also indicated the disruption of colocalization of TRAX and PLCß1 in cells by GHCer derived from EVs. CONCLUSIONS: Overall, the fucose residue in GHCer dictated the glycan conformation for its complexing with TRAX to release TRAX-sequestered PLCß1, leading to Ca2+ mobilization in endothelial cells and enhancing angiogenesis in tumor microenvironments.


Asunto(s)
Proteínas de Unión al ADN , Fucosa , Células Endoteliales de la Vena Umbilical Humana , Animales , Humanos , Ratones , Ceramidas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fucosa/genética , Fucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo
3.
Am J Hum Genet ; 102(1): 188-195, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29304374

RESUMEN

Fucosyltransferase 8 (FUT8) encodes a Golgi-localized α1,6 fucosyltransferase that is essential for transferring the monosaccharide fucose into N-linked glycoproteins, a process known as "core fucosylation." Here we describe three unrelated individuals, who presented with intrauterine growth retardation, severe developmental and growth delays with shortened limbs, neurological impairments, and respiratory complications. Each underwent whole-exome sequencing and was found to carry pathogenic variants in FUT8. The first individual (consanguineous family) was homozygous for c.715C>T (p.Arg239∗), while the second (non-consanguineous family) was compound heterozygous for c.1009C>G (p.Arg337Gly) and a splice site variant c.1259+5G>T. The third individual (consanguineous family) was homozygous for a c.943C>T (p.Arg315∗). Splicing analysis confirmed the c.1259+5G>T resulted in expression of an abnormal FUT8 transcript lacking exon 9. Functional studies using primary fibroblasts from two affected individuals revealed a complete lack of FUT8 protein expression that ultimately resulted in substantial deficiencies in total core fucosylated N-glycans. Furthermore, serum samples from all three individuals showed a complete loss of core fucosylation. Here, we show that loss of function mutations in FUT8 cause a congenital disorder of glycosylation (FUT8-CDG) characterized by defective core fucosylation that phenotypically parallels some aspects of the Fut8-/- knockout mouse. Importantly, identification of additional affected individuals can be easily achieved through analysis of core fucosylation of N-glycans.


Asunto(s)
Alelos , Fucosa/genética , Fucosiltransferasas/genética , Mutación/genética , Empalme Alternativo/genética , Células Cultivadas , Niño , Preescolar , Resultado Fatal , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Glicosilación , Humanos , Lectinas/metabolismo , Masculino , Polisacáridos/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Biochem J ; 477(6): 1179-1201, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32141499

RESUMEN

Fucosyltransferase 8 (FUT8) and ß-galactoside α-2,6-sialyltransferase 1 (ST6GAL1) are glycosyltransferases that catalyze α1,6-fucosylation and α2,6-sialylation, respectively, in the mammalian N-glycosylation pathway. They are aberrantly expressed in various human diseases. FUT8 is non-glycosylated but is responsible for the fucosylation of ST6GAL1. However, the mechanism for the interaction between these two enzymes is unknown. In this study, we show that serum levels of α2,6-sialylated N-glycans are increased in Fut8-/- mice, whereas the mRNA and protein levels of ST6GAL1 are unchanged in mouse live tissues. The level of α2,6-sialylation on IgG was also enhanced in Fut8-/- mice along with ST6GAL1 catalytic activity increase in both serum and liver. Moreover, it was observed that ST6GAL1 prefers non-fucosylated substrates. Interestingly, increased core fucosylation accompanied by a reduction in α2,6-sialylation, was detected in rheumatoid arthritis patient serum. These findings provide new insight into the interactions between FUT8 and ST6GAL1.


Asunto(s)
Antígenos CD/genética , Fucosiltransferasas/deficiencia , Fucosiltransferasas/genética , Sialiltransferasas/deficiencia , Sialiltransferasas/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Femenino , Fucosa/genética , Fucosa/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilación , Humanos , Ratones , Ratones Noqueados , Persona de Mediana Edad
5.
Int J Mol Sci ; 22(24)2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34948129

RESUMEN

Fucosylation is an oligosaccharide modification that plays an important role in immune response and malignancy, and specific fucosyltransferases (FUTs) catalyze the three types of fucosylations: core-type, Lewis type, and H type. FUTs regulate cancer proliferation, invasiveness, and resistance to chemotherapy by modifying the glycosylation of signaling receptors. Oligosaccharides on PD-1/PD-L1 proteins are specifically fucosylated, leading to functional modifications. Expression of FUTs is upregulated in renal cell carcinoma, bladder cancer, and prostate cancer. Aberrant fucosylation in prostate-specific antigen (PSA) could be used as a novel biomarker for prostate cancer. Furthermore, elucidation of the biological function of fucosylation could result in the development of novel therapeutic targets. Further studies are needed in the field of fucosylation glycobiology in urological malignancies.


Asunto(s)
Fucosa/metabolismo , Fucosiltransferasas/metabolismo , Proteínas de Neoplasias/metabolismo , Oligosacáridos/metabolismo , Neoplasias Urológicas/metabolismo , Fucosa/genética , Fucosiltransferasas/genética , Glicosilación , Humanos , Proteínas de Neoplasias/genética , Oligosacáridos/genética , Neoplasias Urológicas/genética
6.
J Biol Chem ; 294(6): 1967-1983, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30538131

RESUMEN

Toxoplasma gondii is an intracellular parasite that causes disseminated infections that can produce neurological damage in fetuses and immunocompromised individuals. Microneme protein 2 (MIC2), a member of the thrombospondin-related anonymous protein (TRAP) family, is a secreted protein important for T. gondii motility, host cell attachment, invasion, and egress. MIC2 contains six thrombospondin type I repeats (TSRs) that are modified by C-mannose and O-fucose in Plasmodium spp. and mammals. Here, using MS analysis, we found that the four TSRs in T. gondii MIC2 with protein O-fucosyltransferase 2 (POFUT2) acceptor sites are modified by a dHexHex disaccharide, whereas Trp residues within three TSRs are also modified with C-mannose. Disruption of genes encoding either POFUT2 or the putative GDP-fucose transporter (NST2) resulted in loss of MIC2 O-fucosylation, as detected by an antibody against the GlcFuc disaccharide, and in markedly reduced cellular levels of MIC2. Furthermore, in 10-15% of the Δpofut2 or Δnst2 vacuoles, MIC2 accumulated earlier in the secretory pathway rather than localizing to micronemes. Dissemination of tachyzoites in human foreskin fibroblasts was reduced for these knockouts, which both exhibited defects in attachment to and invasion of host cells comparable with the Δmic2 phenotype. These results, indicating that O-fucosylation of TSRs is required for efficient processing of MIC2 and for normal parasite invasion, are consistent with the recent demonstration that Plasmodium falciparum Δpofut2 strain has decreased virulence and also support a conserved role for this glycosylation pathway in quality control of TSR-containing proteins in eukaryotes.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Fucosiltransferasas/metabolismo , Estadios del Ciclo de Vida , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Moléculas de Adhesión Celular/genética , Fucosa/genética , Fucosa/metabolismo , Fucosiltransferasas/genética , Glicosilación , Humanos , Proteínas Protozoarias/genética , Secuencias Repetitivas de Aminoácido , Toxoplasma/genética , Toxoplasma/crecimiento & desarrollo
7.
Nat Chem Biol ; 14(1): 65-71, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29176671

RESUMEN

Notch is a cell-surface receptor that controls cell-fate decisions and is regulated by O-glycans attached to epidermal growth factor-like (EGF) repeats in its extracellular domain. Protein O-fucosyltransferase 1 (Pofut1) modifies EGF repeats with O-fucose and is essential for Notch signaling. Constitutive activation of Notch signaling has been associated with a variety of human malignancies. Therefore, tools that inhibit Notch activity are being developed as cancer therapeutics. To this end, we screened L-fucose analogs for their effects on Notch signaling. Two analogs, 6-alkynyl and 6-alkenyl fucose, were substrates of Pofut1 and were incorporated directly into Notch EGF repeats in cells. Both analogs were potent inhibitors of binding to and activation of Notch1 by Notch ligands Dll1 and Dll4, but not by Jag1. Mutagenesis and modeling studies suggest that incorporation of the analogs into EGF8 of Notch1 markedly reduces the ability of Delta ligands to bind and activate Notch1.


Asunto(s)
Familia de Proteínas EGF/metabolismo , Fucosa/análogos & derivados , Fucosa/farmacología , Fucosiltransferasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Fucosa/química , Fucosa/genética , Fucosiltransferasas/genética , Células HEK293 , Humanos , Ligandos , Unión Proteica
8.
Biochem Biophys Res Commun ; 511(2): 482-489, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30808544

RESUMEN

Pancreatic cancer is a highly malignant tumor of the digestive system. Previous studies have shown that abnormal cell surface glycosylation is associated with cancer metastasis, which suggests that glycosylation changes may open a new window for discovering metastasis-related pathways. In this study, we used a microarray with 55 lectins to screen for altered glycosylation between two metastatic pancreatic cancer lines (Capan-1 and Su.86.86) and two nonmetastatic pancreatic cancer lines (Panc-1 and MIA PaCa-2), and we further analyzed three lectins with high-binding activities (AAL, UEA-I, and PHA-E) in cell motility assays using these pancreatic cancer cells to detect whether blocking certain forms of cell surface glycosylation affects any processes associated with metastasis. As a result, we found that AAL, a fucose-specific lectin, has different binding patterns between metastatic pancreatic cancer and nonmetastatic pancreatic cancer lines and inhibits cell motility in metastatic pancreatic cancer cells. Furthermore, the N-fucosylation-related genes FUT3, 5, and 6 were found to be responsible for the elevated fucosylation in metastatic pancreatic cells through real-time PCR screening. In summary, our findings that the specific bindings of AAL on cell surfaces and highly expressed FUT3, 5, and 6 in metastatic pancreatic cancer cells, although preliminary, are encouraging, and our established combined method is also suitable for discovering metastasis-related mechanisms in other cancers.


Asunto(s)
Fucosiltransferasas/genética , Neoplasias Pancreáticas/genética , Línea Celular Tumoral , Movimiento Celular , Fucosa/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Regulación hacia Arriba
9.
Biotechnol Bioeng ; 116(9): 2412-2417, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31145478

RESUMEN

2'-Fucosyllactose (2-FL), one of the most abundant oligosaccharides in human milk, has been spotlighted for its neutraceutical and pharmaceutical potentials. Microbial production of 2-FL is promising since it is efficient as compared to other production methods. In 2-FL microbial production via the salvage pathway for biosynthesis of guanosine 5'-diphosphate (GDP)-l-fucose from fucose, the conversion yield from fucose is important because of the high price of fucose. In this study, deletion of the genes (araA and rhaA) coding for arabinose isomerase (AraA) and rhamnose isomerase (RhaA) was attempted in engineered Escherichia coli for improving 2-FL production by using fucose, lactose, and glycerol. The engineered E. coli constructed previously is able to express fucokinase/GDP-l-fucose pyrophosphorylase (Fkp) from Bacteroides fragilis and the α-1,2-fucosyltransferase (FucT2) from Helicobacter pylori and deficient in ß-galactosidase (LacZ), fucose isomerase (FucI), and fuculose kinase (FucK). The additional double-deletion of the araA and rhaA genes in the engineered E. coli enhanced the product yield of 2-FL to 0.52 mole 2-FL/mole fucose, and hence the concentration of 2-FL reached to 47.0 g/L, which are 44% and two-fold higher than those (23.1 g/L and 0.36 mole 2-FL/mole fucose) of the control strain in fed-batch fermentation. Elimination of sugar isomerases exhibiting promiscuous activities with fucose might be critical in the microbial production of 2-FL through the salvage pathway of GDP-l-fucose.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Fucosa/metabolismo , Eliminación de Gen , Ingeniería Metabólica , Trisacáridos/biosíntesis , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fucosa/genética , Trisacáridos/genética
10.
Biotechnol Bioeng ; 116(4): 904-911, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30597526

RESUMEN

L-Fucose (6-deoxy-L-galactose) is a major constituent of glycans and glycolipids in mammals. Fucosylation of glycans can confer unique functional properties and may be an economical way to manufacture L-fucose. Research can extract L-fucose directly from brown algae, or by enzymatic hydrolysis of L-fucose-rich microbial exopolysaccharides. However, these L-fucose production methods are not economical or scalable for various applications. We engineered an Escherichia coli strain to produce L-fucose. Specifically, we modified the strain genome to eliminate endogenous L-fucose and lactose metabolism, produce 2'-fucosyllactose (2'-FL), and to liberate L-fucose from 2'-FL. This E. coli strain produced 16.7 g/L of L-fucose with productivity of 0.1 g·L-1 ·h-1 in a fed-batch fermentation. This study presents an efficient one-pot biosynthesis strategy to produce a monomeric form of L-fucose by microbial fermentation, making large-scale industrial production of L-fucose feasible.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Escherichia coli/metabolismo , Fucosa/metabolismo , Reactores Biológicos , Vías Biosintéticas , Escherichia coli/genética , Fermentación , Fucosa/genética , Microbiología Industrial/métodos , Ingeniería Metabólica/métodos
11.
J Biol Chem ; 291(26): 13743-52, 2016 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-27129198

RESUMEN

Notch is a transmembrane receptor that mediates cell-cell interactions and controls various cell-fate specifications in metazoans. The extracellular domain of Notch contains multiple epidermal growth factor (EGF)-like repeats. At least five different glycans are found in distinct sites within these EGF-like repeats. The function of these individual glycans in Notch signaling has been investigated, primarily by disrupting their individual glycosyltransferases. However, we are just beginning to understand the potential functional interactions between these glycans. Monosaccharide O-fucose and O-glucose trisaccharide (O-glucose-xylose-xylose) are added to many of the Notch EGF-like repeats. In Drosophila, Shams adds a xylose specifically to the monosaccharide O-glucose. We found that loss of the terminal dixylose of O-glucose-linked saccharides had little effect on Notch signaling. However, our analyses of double mutants of shams and other genes required for glycan modifications revealed that both the monosaccharide O-glucose and the terminal dixylose of O-glucose-linked saccharides function redundantly with the monosaccharide O-fucose in Notch activation and trafficking. The terminal dixylose of O-glucose-linked saccharides and the monosaccharide O-glucose were required in distinct Notch trafficking processes: Notch transport from the apical plasma membrane to adherens junctions, and Notch export from the endoplasmic reticulum, respectively. Therefore, the monosaccharide O-glucose and terminal dixylose of O-glucose-linked saccharides have distinct activities in Notch trafficking, although a loss of these activities is compensated for by the presence of monosaccharide O-fucose. Given that various glycans attached to a protein motif may have redundant functions, our results suggest that these potential redundancies may lead to a serious underestimation of glycan functions.


Asunto(s)
Proteínas de Drosophila/metabolismo , Retículo Endoplásmico/metabolismo , Fucosa/metabolismo , Receptores Notch/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Retículo Endoplásmico/genética , Fucosa/genética , Glucosa/genética , Glucosa/metabolismo , Glicosilación , Transporte de Proteínas/fisiología , Receptores Notch/genética , Secuencias Repetitivas de Aminoácido , Xilosa/genética , Xilosa/metabolismo
12.
J Biol Chem ; 291(31): 16348-60, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27268051

RESUMEN

Glycosylation of the Notch receptor is essential for its activity and serves as an important modulator of signaling. Three major forms of O-glycosylation are predicted to occur at consensus sites within the epidermal growth factor-like repeats in the extracellular domain of the receptor: O-fucosylation, O-glucosylation, and O-GlcNAcylation. We have performed comprehensive mass spectral analyses of these three types of O-glycosylation on Drosophila Notch produced in S2 cells and identified peptides containing all 22 predicted O-fucose sites, all 18 predicted O-glucose sites, and all 18 putative O-GlcNAc sites. Using semiquantitative mass spectral methods, we have evaluated the occupancy and relative amounts of glycans at each site. The majority of the O-fucose sites were modified to high stoichiometries. Upon expression of the ß3-N-acetylglucosaminyltransferase Fringe with Notch, we observed varying degrees of elongation beyond O-fucose monosaccharide, indicating that Fringe preferentially modifies certain sites more than others. Rumi modified O-glucose sites to high stoichiometries, although elongation of the O-glucose was site-specific. Although the current putative consensus sequence for O-GlcNAcylation predicts 18 O-GlcNAc sites on Notch, we only observed apparent O-GlcNAc modification at five sites. In addition, we performed mass spectral analysis on endogenous Notch purified from Drosophila embryos and found that the glycosylation states were similar to those found on Notch from S2 cells. These data provide foundational information for future studies investigating the mechanisms of how O-glycosylation regulates Notch activity.


Asunto(s)
Proteínas de Drosophila , N-Acetilglucosaminiltransferasas , Mapeo Peptídico , Péptidos , Receptores Notch , Animales , Línea Celular , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Fucosa/química , Fucosa/genética , Fucosa/metabolismo , Glicosilación , N-Acetilglucosaminiltransferasas/biosíntesis , N-Acetilglucosaminiltransferasas/química , N-Acetilglucosaminiltransferasas/genética , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Receptores Notch/química , Receptores Notch/genética , Receptores Notch/metabolismo
13.
Breast Cancer Res ; 19(1): 111, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28982386

RESUMEN

BACKGROUND: Core fucosylation (addition of fucose in α-1,6-linkage to core N-acetylglucosamine of N-glycans) catalyzed by fucosyltransferase 8 (FUT8) is critical for signaling receptors involved in many physiological and pathological processes such as cell growth, adhesion, and tumor metastasis. Transforming growth factor-ß (TGF-ß)-induced epithelial-mesenchymal transition (EMT) regulates the invasion and metastasis of breast tumors. However, whether receptor core fucosylation affects TGF-ß signaling during breast cancer progression remains largely unknown. METHOD: In this study, gene expression profiling and western blot were used to validate the EMT-associated expression of FUT8. Lentivirus-mediated gain-of-function study, short hairpin RNA (shRNA) or CRISPR/Cas9-mediated loss-of-function studies and pharmacological inhibition of FUT8 were used to elucidate the molecular function of FUT8 during TGF-ß-induced EMT in breast carcinoma cells. In addition, lectin blot, luciferase assay, and in vitro ligand binding assay were employed to demonstrate the involvement of FUT8 in the TGF-ß1 signaling pathway. The role of FUT8 in breast cancer migration, invasion, and metastasis was confirmed using an in vitro transwell assay and mammary fat pad xenograft in vivo tumor model. RESULTS: Gene expression profiling analysis revealed that FUT8 is upregulated in TGF-ß-induced EMT; the process was associated with the migratory and invasive abilities of several breast carcinoma cell lines. Gain-of-function and loss-of-function studies demonstrated that FUT8 overexpression stimulated the EMT process, whereas FUT8 knockdown suppressed the invasiveness of highly aggressive breast carcinoma cells. Furthermore, TGF-ß receptor complexes might be core fucosylated by FUT8 to facilitate TGF-ß binding and enhance downstream signaling. Importantly, FUT8 inhibition suppressed the invasive ability of highly metastatic breast cancer cells and impaired their lung metastasis. CONCLUSIONS: Our results reveal a positive feedback mechanism of FUT8-mediated receptor core fucosylation that promotes TGF-ß signaling and EMT, thus stimulating breast cancer cell invasion and metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Fucosiltransferasas/genética , Invasividad Neoplásica/genética , Factor de Crecimiento Transformador beta1/genética , Neoplasias de la Mama/patología , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Fucosa/genética , Fucosa/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Lentivirus/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Fosforilación , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal/genética
14.
Mol Microbiol ; 101(4): 575-89, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27145048

RESUMEN

Campylobacter jejuni and Campylobacter coli are zoonotic pathogens once considered asaccharolytic, but are now known to encode pathways for glucose and fucose uptake/metabolism. For C. jejuni, strains with the fuc locus possess a competitive advantage in animal colonization models. We demonstrate that this locus is present in > 50% of genome-sequenced strains and is prevalent in livestock-associated isolates of both species. To better understand how these campylobacters sense nutrient availability, we examined biofilm formation and chemotaxis to fucose. C. jejuni NCTC11168 forms less biofilms in the presence of fucose, although its fucose permease mutant (fucP) shows no change. In a newly developed chemotaxis assay, both wild-type and the fucP mutant are chemotactic towards fucose. C. jejuni 81-176 naturally lacks the fuc locus and is unable to swim towards fucose. Transfer of the NCTC11168 locus into 81-176 activated fucose uptake and chemotaxis. Fucose chemotaxis also correlated with possession of the pathway for C. jejuni RM1221 (fuc+) and 81116 (fuc-). Systematic mutation of the NCTC11168 locus revealed that Cj0485 is necessary for fucose metabolism and chemotaxis. This study suggests that components for fucose chemotaxis are encoded within the fuc locus, but downstream signals only in fuc + strains, are involved in coordinating fucose availability with biofilm development.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Campylobacter jejuni/fisiología , Fucosa/metabolismo , Campylobacter coli/genética , Campylobacter coli/metabolismo , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Metabolismo de los Hidratos de Carbono , Quimiotaxis/fisiología , Fucosa/genética , Genotipo
15.
EMBO J ; 31(14): 3183-97, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22588082

RESUMEN

Protein O-fucosylation is a post-translational modification found on serine/threonine residues of thrombospondin type 1 repeats (TSR). The fucose transfer is catalysed by the protein O-fucosyltransferase 2 (POFUT2) and >40 human proteins contain the TSR consensus sequence for POFUT2-dependent fucosylation. To better understand O-fucosylation on TSR, we carried out a structural and functional analysis of human POFUT2 and its TSR substrate. Crystal structures of POFUT2 reveal a variation of the classical GT-B fold and identify sugar donor and TSR acceptor binding sites. Structural findings are correlated with steady-state kinetic measurements of wild-type and mutant POFUT2 and TSR and give insight into the catalytic mechanism and substrate specificity. By using an artificial mini-TSR substrate, we show that specificity is not primarily encoded in the TSR protein sequence but rather in the unusual 3D structure of a small part of the TSR. Our findings uncover that recognition of distinct conserved 3D fold motifs can be used as a mechanism to achieve substrate specificity by enzymes modifying completely folded proteins of very wide sequence diversity and biological function.


Asunto(s)
Fucosiltransferasas/química , Pliegue de Proteína , Cristalografía por Rayos X , Fucosa/química , Fucosa/genética , Fucosa/metabolismo , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Glicosilación , Humanos , Estructura Terciaria de Proteína , Secuencias Repetitivas de Aminoácido , Relación Estructura-Actividad
16.
J Proteome Res ; 14(12): 5291-305, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26538210

RESUMEN

Recent studies have shown a remarkable degree of plasticity in the N-glycome of the model nematode Caenorhabditis elegans; ablation of glycosylation-relevant genes can result in radically altered N-glycan profiles despite only minor biological phenotypic effects. Up to four fucose residues and five different linkages of fucose are known on the N-glycans of C. elegans. Due to the complexity in the wild type, we established three mutant strains defective in two core fucosyltransferases each (fut-1;fut-6, fut-1;fut-8, and fut-6;fut-8). Enzymatically released N-glycans were subject to HPLC and MALDI-TOF MS/MS, in combination with various treatments, to verify structural details. The N-glycome of the fut-1;fut-6 mutant was the most complex of the three double-mutant strains due to the extension of the core α1,6-fucose as well as the presence of fucose on the bisecting galactose. In contrast, maximally two fucoses were found on N-glycans of the fut-1;fut-8 and fut-6;fut-8 strains. The different locations and capping of fucose meant that up to 13 isomeric structures, many highly galactosylated, were determined for some single masses. These data not only show the high variability of the N-glycomic capacity of a "simple" nematode but also exemplify the need for multiple approaches to reveal individual glycan structures within complex invertebrate glycomes.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Fucosiltransferasas/química , Fucosiltransferasas/genética , Animales , Secuencia de Carbohidratos , Cromatografía Líquida de Alta Presión , Fucosa/química , Fucosa/genética , Genes de Helminto , Glicómica , Glicosilación , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación , Polisacáridos/química , Polisacáridos/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
17.
J Biol Chem ; 288(23): 16839-16847, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23609441

RESUMEN

Erythropoiesis results from a complex combination of the expression of several transcription factor genes and cytokine signaling. However, the overall view of erythroid differentiation remains unclear. First, we screened for erythroid differentiation-related genes by comparing the expression profiles of high differentiation-inducible and low differentiation-inducible murine erythroleukemia cells. We identified that overexpression of α-1,6-fucosyltransferase (Fut8) inhibits hemoglobin production. FUT8 catalyzes the transfer of a fucose residue to N-linked oligosaccharides on glycoproteins via an α-1,6 linkage, leading to core fucosylation in mammals. Expression of Fut8 was down-regulated during chemically induced differentiation of murine erythroleukemia cells. Additionally, expression of Fut8 was positively regulated by c-Myc and c-Myb, which are known as suppressors of erythroid differentiation. Second, we found that FUT8 is the only fucosyltransferase family member that inhibits hemoglobin production. Functional analysis of FUT8 revealed that the donor substrate-binding domain and a flexible loop play essential roles in inhibition of hemoglobin production. This result clearly demonstrates that core fucosylation inhibits hemoglobin production. Third, FUT8 also inhibited hemoglobin production of human erythroleukemia K562 cells. Finally, a short hairpin RNA study showed that FUT8 down-regulation induced hemoglobin production and increase of transferrin receptor/glycophorin A-positive cells in human erythroleukemia K562 cells. Our findings define FUT8 as a novel factor for hemoglobin production and demonstrate that core fucosylation plays an important role in erythroid differentiation.


Asunto(s)
Diferenciación Celular , Fucosiltransferasas/metabolismo , Hemoglobinas/biosíntesis , Leucemia Eritroblástica Aguda/enzimología , Animales , Transporte Biológico Activo/genética , Fucosa/genética , Fucosa/metabolismo , Fucosiltransferasas/genética , Glicoforinas/genética , Glicoforinas/metabolismo , Hemoglobinas/genética , Humanos , Células K562 , Ratones , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
18.
J Biol Chem ; 287(41): 33973-82, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22872641

RESUMEN

Fucα1-6 oligosaccharide has a variety of biological functions and serves as a biomarker for hepatocellular carcinoma because of the elevated presence of fucosylated α-fetoprotein (AFP) in this type of cancer. In this study we purified a novel Fucα1-6-specific lectin from the mushroom Pholiota squarrosa by ion-exchange chromatography and affinity chromatography on thyroglobulin-agarose. The purified lectin was designated as PhoSL (P. squarrosa lectin). SDS-PAGE, MALDI-TOF mass spectrometry, and N-terminal amino acid sequencing indicate that PhoSL has a molecular mass of 4.5 kDa and consists of 40 amino acids (NH(2)-APVPVTKLVCDGDTYKCTAYLDFGDGRWVAQWDTNVFHTG-OH). Isoelectric focusing of the lectin showed bands near pI 4.0. The lectin activity was stable between pH 2.0 and 11.0 and at temperatures ranging from 0 to 100 °C for incubation times of 30 min. When PhoSL was investigated with frontal affinity chromatography using 132 pyridylaminated oligosaccharides, it was found that the lectin binds only to core α1-6-fucosylated N-glycans and not to other types of fucosylated oligosaccharides, such as α1-2-, α1-3-, and α1-4-fucosylated glycans. Furthermore, PhoSL bound to α1-6-fucosylated AFP but not to non-fucosylated AFP. In addition, PhoSL was able to demonstrate the differential expression of α1-6 fucosylation between primary and metastatic colon cancer tissues. Thus, PhoSL will be a promising tool for analyzing the biological functions of α1-6 fucosylation and evaluating Fucα1-6 oligosaccharides as cancer biomarkers.


Asunto(s)
Fucosa/química , Proteínas Fúngicas/química , Lectinas/química , Oligosacáridos/química , Pholiota/química , Secuencia de Aminoácidos , Antígenos de Neoplasias/metabolismo , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Fucosa/genética , Fucosa/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/aislamiento & purificación , Proteínas Fúngicas/metabolismo , Humanos , Lectinas/genética , Lectinas/aislamiento & purificación , Lectinas/metabolismo , Neoplasias Hepáticas/metabolismo , Datos de Secuencia Molecular , Oligosacáridos/genética , Oligosacáridos/metabolismo , Pholiota/genética , Pholiota/metabolismo , Unión Proteica , Estabilidad Proteica
19.
J Biol Chem ; 287(34): 28276-90, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22733825

RESUMEN

The modification of α1,6-linked fucose residues attached to the proximal (reducing-terminal) core N-acetylglucosamine residue of N-glycans by ß1,4-linked galactose ("GalFuc" epitope) is a feature of a number of invertebrate species including the model nematode Caenorhabditis elegans. A pre-requisite for both core α1,6-fucosylation and ß1,4-galactosylation is the presence of a nonreducing terminal N-acetylglucosamine; however, this residue is normally absent from the final glycan structure in invertebrates due to the action of specific hexosaminidases. Previously, we have identified two hexosaminidases (HEX-2 and HEX-3) in C. elegans, which process N-glycans. In the present study, we have prepared a hex-2;hex-3 double mutant, which possesses a radically altered N-glycomic profile. Whereas in the double mutant core α1,3-fucosylation of the proximal N-acetylglucosamine was abolished, the degree of galactosylation of core α1,6-fucose increased, and a novel Galα1,2Fucα1,3 moiety attached to the distal core N-acetylglucosamine residue was detected. Both galactosylated fucose moieties were also found in two parasitic nematodes, Ascaris suum and Oesophagostomum dentatum. As core modifications of N-glycans are known targets for fungal nematotoxic lectins, the sensitivity of the C. elegans double hexosaminidase mutant was assessed. Although this mutant displayed hypersensitivity to the GalFuc-binding lectin CGL2 and the N-acetylglucosamine-binding lectin XCL, the mutant was resistant to CCL2, which binds core α1,3-fucose. Thus, the use of C. elegans mutants aids the identification of novel N-glycan modifications and the definition of in vivo specificities of nematotoxic lectins with potential as anthelmintic agents.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Epítopos/metabolismo , Fucosa/metabolismo , Hexosaminidasas/metabolismo , Polisacáridos/metabolismo , Acetilglucosamina/genética , Acetilglucosamina/metabolismo , Animales , Antihelmínticos/farmacología , Ascaris suum/genética , Ascaris suum/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Diseño de Fármacos , Epítopos/genética , Fucosa/genética , Galectina 2/farmacología , Glicosilación , Hexosaminidasas/genética , Mutación , Oesophagostomum/genética , Oesophagostomum/metabolismo , Polisacáridos/genética
20.
J Biol Chem ; 287(4): 2500-8, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22084235

RESUMEN

α1,6-Fucosyltransferase (Fut8) knock-out (Fut8(-/-)) mice showed an abnormality in pre-B cell generation. Membrane assembly of pre-BCR is a crucial checkpoint for pre-B cell differentiation and proliferation in both humans and mice. The assembly of pre-BCR on the cell surface was substantially blocked in the Fut8-knockdown pre-B cell line, 70Z/3-KD cells, and then completely restored by re-introduction of the Fut8 gene to 70Z/3-KD (70Z/3-KD-re) cells. Moreover, loss of α1,6-fucosylation (also called core fucosylation) of µHC was associated with the suppression of the interaction between µHC and λ5. In contrast to Fut8(+/+) CD19(+)CD43(-) cells, the subpopulation expressing the µHC·λ5 complex in the Fut8(-/-) CD19(+)CD43(-) cell fraction was decreased. The pre-BCR-mediated tyrosine phosphorylation of CD79a and activation of Btk were attenuated in Fut8-KD cells, and restored in 70Z/3-KD-re cells. The frequency of CD19(low)CD43(-) cells (pre-B cell enriched fraction) was also reduced in Fut8(-/-) bone marrow cells, and then the levels of IgM, IgG, and IgA of 12-week-old Fut8(-/-) mice sera were significantly lower than those of Fut8(+/+) mice. Our results suggest that the core fucosylation of µHC mediates the assembly of pre-BCR to regulate pre-BCR intracellular signaling and pre-B cell proliferation.


Asunto(s)
Fucosa/metabolismo , Fucosiltransferasas/metabolismo , Cadenas mu de Inmunoglobulina/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/fisiología , Animales , Fucosa/genética , Fucosa/inmunología , Fucosiltransferasas/genética , Fucosiltransferasas/inmunología , Glicosilación , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Inmunoglobulina M/metabolismo , Cadenas lambda de Inmunoglobulina/genética , Cadenas lambda de Inmunoglobulina/inmunología , Cadenas lambda de Inmunoglobulina/metabolismo , Cadenas mu de Inmunoglobulina/genética , Cadenas mu de Inmunoglobulina/inmunología , Ratones , Ratones Noqueados , Células Precursoras de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA