Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 223
Filter
1.
Glycobiology ; 33(10): 784-800, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37471650

ABSTRACT

Recent human H3N2 influenza A viruses have evolved to employ elongated glycans terminating in α2,6-linked sialic acid as their receptors. These glycans are displayed in low abundancies by (humanized) Madin-Darby Canine Kidney cells, which are commonly employed to propagate influenza A virus, resulting in low or no viral propagation. Here, we examined whether the overexpression of the glycosyltransferases ß-1,3-N-acetylglucosaminyltransferase and ß-1,4-galactosyltransferase 1, which are responsible for the elongation of poly-N-acetyllactosamines (LacNAcs), would result in improved A/H3N2 propagation. Stable overexpression of ß-1,3-N-acetylglucosaminyltransferase and ß-1,4-galactosyltransferase 1 in Madin-Darby Canine Kidney and "humanized" Madin-Darby Canine Kidney cells was achieved by lentiviral integration and subsequent antibiotic selection and confirmed by qPCR and protein mass spectrometry experiments. Flow cytometry and glycan mass spectrometry experiments using the ß-1,3-N-acetylglucosaminyltransferase and/or ß-1,4-galactosyltransferase 1 knock-in cells demonstrated increased binding of viral hemagglutinins and the presence of a larger number of LacNAc repeating units, especially on "humanized" Madin-Darby Canine Kidney-ß-1,3-N-acetylglucosaminyltransferase cells. An increase in the number of glycan receptors did, however, not result in a greater infection efficiency of recent human H3N2 viruses. Based on these results, we propose that H3N2 influenza A viruses require a low number of suitable glycan receptors to infect cells and that an increase in the glycan receptor display above this threshold does not result in improved infection efficiency.


Subject(s)
Influenza A Virus, H3N2 Subtype , Influenza A virus , Humans , Animals , Dogs , Influenza A Virus, H3N2 Subtype/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , N-Acetyllactosamine Synthase/metabolism , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A virus/metabolism , Madin Darby Canine Kidney Cells , Polysaccharides/chemistry
2.
Anal Biochem ; 593: 113599, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32004544

ABSTRACT

Split luciferase complementation assay is one of the approaches enabling identification and analysis of protein-protein interactions in vivo. The NanoBiT technology is the most recent improvement of this strategy. Nucleotide sugar transporters and glycosyltransferases of the Golgi apparatus are the key players in glycosylation. Here we demonstrate the applicability of the NanoBiT system for studying homooligomerization of these proteins. We also report and discuss a novel heterologous interaction between UDP-galactose transporter and beta-1,4-galactosyltransferase 1.


Subject(s)
Luminescent Measurements/methods , Monosaccharide Transport Proteins/metabolism , N-Acetyllactosamine Synthase/metabolism , Nanotechnology/methods , Amino Acid Sequence , Animals , Biological Transport , CHO Cells , Cricetulus , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Protein Binding
3.
Chemistry ; 23(71): 18057-18065, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29024190

ABSTRACT

Xyloside analogues with substitution of the endocyclic oxygen atom by sulfur or carbon were investigated as substrates for ß-1,4-galactosyltransferase 7 (ß4GalT7), a key enzyme in the biosynthesis of glycosaminoglycan chains. The analogues with an endocyclic sulfur atom proved to be excellent substrates for ß4GalT7, and were galactosylated approximately fifteen times more efficiently than the corresponding xyloside. The 5a-carba-ß-xylopyranoside in the d-configuration proved to be a good substrate for ß4GalT7, whereas the enantiomer in the l-configuration showed no activity. Further investigations by X-ray crystallography, NMR spectroscopy, and molecular modeling provided a rationale for the pronounced activity of the sulfur analogues. Favorable π-π interactions between the 2-naphthyl moiety and a tyrosine side chain of the enzyme were observed for the thio analogues, which open up for the design of efficient GAG primers and inhibitors.


Subject(s)
N-Acetyllactosamine Synthase/metabolism , Sulfhydryl Compounds/chemistry , Xylose/analogs & derivatives , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Molecular Conformation , Molecular Docking Simulation , N-Acetyllactosamine Synthase/chemistry , Nuclear Magnetic Resonance, Biomolecular , Quantum Theory , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Substrate Specificity , Sulfhydryl Compounds/metabolism , Xylose/metabolism
4.
Glycoconj J ; 34(3): 411-420, 2017 06.
Article in English | MEDLINE | ID: mdl-27744520

ABSTRACT

Proteoglycans and glycosaminoglycans modulate numerous cellular processes relevant to tumour progression, including cell proliferation, cell-matrix interactions, cell motility and invasive growth. Among the glycosaminoglycans with a well-documented role in tumour progression are heparan sulphate, chondroitin/dermatan sulphate and hyaluronic acid/hyaluronan. While the mode of biosynthesis differs for sulphated glycosaminoglycans, which are synthesised in the ER and Golgi compartments, and hyaluronan, which is synthesized at the plasma membrane, these polysaccharides partially compete for common substrates. In this study, we employed a siRNA knockdown approach for heparan sulphate (EXT1) and heparan/chondroitin/dermatan sulphate-biosynthetic enzymes (ß4GalT7) in the aggressive human breast cancer cell line MDA-MB-231 to study the impact on cell behaviour and hyaluronan biosynthesis. Knockdown of ß4GalT7 expression resulted in a decrease in cell viability, motility and adhesion to fibronectin, while these parameters were unchanged in EXT1-silenced cells. Importantly, these changes were associated with a decreased expression of syndecan-1, decreased signalling response to HGF and an increase in the synthesis of hyaluronan, due to an upregulation of the hyaluronan synthases HAS2 and HAS3. Interestingly, EXT1-depleted cells showed a downregulation of the UDP-sugar transporter SLC35D1, whereas SLC35D2 was downregulated in ß4GalT7-depleted cells, indicating an intricate regulatory network that connects all glycosaminoglycans synthesis. The results of our in vitro study suggest that a modulation of breast cancer cell behaviour via interference with heparan sulphate biosynthesis may result in a compensatory upregulation of hyaluronan biosynthesis. These findings have important implications for the development of glycosaminoglycan-targeted therapeutic approaches for malignant diseases.


Subject(s)
Chondroitin Sulfates/biosynthesis , Dermatan Sulfate/analogs & derivatives , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Heparitin Sulfate/biosynthesis , Hyaluronic Acid/biosynthesis , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Chondroitin Sulfates/antagonists & inhibitors , Chondroitin Sulfates/genetics , Dermatan Sulfate/antagonists & inhibitors , Dermatan Sulfate/biosynthesis , Dermatan Sulfate/genetics , Epithelial Cells/pathology , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Heparitin Sulfate/antagonists & inhibitors , Heparitin Sulfate/genetics , Humans , Hyaluronan Synthases/antagonists & inhibitors , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Monosaccharide Transport Proteins/antagonists & inhibitors , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , N-Acetyllactosamine Synthase/antagonists & inhibitors , N-Acetyllactosamine Synthase/genetics , N-Acetyllactosamine Synthase/metabolism , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
5.
Transgenic Res ; 26(3): 375-384, 2017 06.
Article in English | MEDLINE | ID: mdl-28332009

ABSTRACT

Nicotiana tabacum BY-2 suspension cells have several advantages that make them suitable for the production of full-size monoclonal antibodies which can be purified directly from the culture medium. Carbohydrate characterization of an antibody (Lo-BM2) expressed in N. tabacum BY-2 cells showed that the purified Lo-BM2 displays N-glycan homogeneity with a high proportion (>70%) of the complex GnGnXF glycoform. The stable co-expression of a human ß-1,4-galactosyltransferase targeted to different Golgi sub-compartments altered Lo-BM2N-glycosylation and resulted in the production of an antibody that exhibited either hybrid structures containing a low abundance of the plant epitopes (α-1,3-fucose and ß-1,2-xylose), or a large amount of galactose-extended N-glycan structures. These results demonstrate the suitability of stable N-glycoengineered N. tabacum BY-2 cell lines for the production of human-like antibodies.


Subject(s)
Immunoglobulin G/metabolism , N-Acetyllactosamine Synthase/genetics , Nicotiana/genetics , Plants, Genetically Modified/genetics , Chromatography, Affinity , Gene Expression Regulation , Glycosylation , Golgi Apparatus/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/isolation & purification , N-Acetyllactosamine Synthase/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Nicotiana/metabolism
6.
Org Biomol Chem ; 15(47): 10064-10072, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29168532

ABSTRACT

Polyglycosylated calixarenes are efficient and selective multivalent ligands for lectins. However, the chemical decoration of these macrocyclic scaffolds with saccharides of increasing complexity is hampered by the highly complex chemistry of carbohydrates. An alternative to the conventional approach is the enzymatic diversification of simple glycocluster-presented glycans. In this work, we present a highly efficient chemo-enzymatic approach to tetra-N-acetyl-lactosaminylcalix[4]arene via glycan extension catalyzed by a human ß-1,4-galactosyltransferase. This demonstrates that calixarenes can be exhaustively processed by enzymatic glycosyl transfer despite the heavy steric crowding, paving the way to the design and achievement of multivalent ligands based on these macrocyclic scaffolds having complex branched glycans.


Subject(s)
Calixarenes/metabolism , N-Acetyllactosamine Synthase/metabolism , Phenols/metabolism , Calixarenes/chemistry , Glycosylation , Humans , Molecular Conformation , Phenols/chemistry
7.
Biochem Biophys Res Commun ; 478(3): 1248-53, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27553271

ABSTRACT

ß1,4 Galactosyltransferase-I (GalT-I) is expressed as two nearly identical polypeptides that differ only in the length of their cytoplasmic domains. The longer isoform has been implicated as a cell surface receptor for extracellular glycoside ligands, such as laminin. To more stringently test the function of the long GalT-I isoform during cell interactions with laminin, we created multiple independent fibroblastic cell lines that fail to express the long isoform, but which express the short GalT-I isoform normally and appear to have normal intracellular galactosylation. Cells devoid of the long GalT-I isoform are unable to adhere and spread on laminin substrates as well as control cells, but retain near normal interactions with fibronectin, which do not rely upon surface GalT-I function. The loss of the long GalT-I isoform also leads to a loss of actin stress fibers, focal adhesions and rac GTPase activation.


Subject(s)
Cell-Matrix Junctions/metabolism , Fibroblasts/metabolism , N-Acetyllactosamine Synthase/metabolism , Animals , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Line , Cell Movement/drug effects , Cell-Matrix Junctions/drug effects , Embryo, Mammalian/cytology , Enzyme Activation/drug effects , Fibronectins/pharmacology , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Laminin/pharmacology , Mice , Protein Isoforms/metabolism , Rats , Stress Fibers/drug effects , Stress Fibers/metabolism , rac GTP-Binding Proteins/metabolism
8.
Anal Bioanal Chem ; 407(30): 8945-58, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26362153

ABSTRACT

Metabolic engineering of glycans present on antibodies and other glycoproteins is becoming an interesting research area for improving our understanding of the glycome. With knowledge of the sialic acid biosynthetic pathways, the experiments described in this report are based on a published procedure involving the addition of a synthesized azido-mannosamine sugar into cell culture media and evaluation of downstream expression as azido-sialic acid. This unique bioorthogonal sugar has the potential for a variety of "click chemistry" reactions through the azide linkage, which allow for it to be isolated and quantified given the choice of label. In this report, mass spectrometry was used to investigate and optimize the cellular absorption of peracetylated N-azidoacetylmannosamine (Ac4ManNAz) to form N-azidoacetylneuraminic acid (SiaNAz) in a Chinese hamster ovary (CHO) cell line transiently expressing a double mutant trastuzumab (TZMm2), human galactosyltransferase 1 (GT), and human α-2,6-sialyltransferase (ST6). This in vivo approach is compared to in vitro enzymatic addition SiaNAz onto TZMm2 using soluble ß-galactosamide α-2,6-sialyltransferase 1 and CMP-SiaNAz as donor. The in vivo results suggest that for this mAb, concentrations above 100 µM of Ac4ManNAz are necessary to allow for observation of terminal SiaNAz on tryptic peptides of TZMm2 by matrix-assisted laser desorption ionization (MALDI) mass spectrometry. This is further confirmed by a parallel study on the production of EG2-hFc monoclonal antibody (Zhang J et al. Prot Expr Purific 65(1); 77-82, 2009) in the presence of increasing concentrations of Ac4ManNAz.


Subject(s)
Polysaccharides/metabolism , Sialic Acids/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Metabolic Engineering , Molecular Structure , N-Acetyllactosamine Synthase/metabolism , Polysaccharides/chemistry , Sialic Acids/metabolism
9.
Mol Cell Biochem ; 365(1-2): 149-58, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22359038

ABSTRACT

ß-1,4-galactosyltransferase-I (ß-1,4-GalT-I) plays a critical role in the initiation and maintenance of peripheral nervous system inflammatory reaction. However, the exact function of ß-1,4-GalT-I in the regulation of SCs proliferation and apoptosis remains unclear. In this study, we found that low concentration of tumor necrosis factor-alpha (TNF-α) induced SCs proliferation, while high concentration of TNF-α induced SCs apoptosis. Meanwhile, the expressions of ß-1,4-GalT-I, TNFR1, and TNFR2 were changed following. When ß-1,4-GalT I overexpression, low concentration of TNF-α-induced SCs proliferation was partially repressed. Concurrently, the activity of ERK1/2 was decreased. While knocking down ß-1,4-GalT I expression, high concentration of TNF-α-induced SCs apoptosis was partially rescued. Consistent with this, the activity of P38 and JNK were decreased. We also found anti-TNFR2 antibody suppressed low concentration of TNF-α-induced SCs proliferation, while anti-TNFR1 antibody inhibited high concentration of TNF-α-induced SCs apoptosis. Thus, present data show that ß-1,4-GalT I may play an important role in SCs proliferation and apoptosis induced by TNF-α via different signal pathways and TNFR.


Subject(s)
Apoptosis , Cell Proliferation , MAP Kinase Signaling System , N-Acetyllactosamine Synthase/metabolism , Schwann Cells/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Caspase 3/metabolism , Cells, Cultured , JNK Mitogen-Activated Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Schwann Cells/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Methods Mol Biol ; 2303: 477-486, 2022.
Article in English | MEDLINE | ID: mdl-34626402

ABSTRACT

ß-1,4-Galactosyltransferase 7 (ß4GalT7) is a key enzyme in the synthesis of two classes of glycosaminoglycans (GAG), i.e., heparan sulfate (HS) and chondroitin/dermatan sulfate (CS/DS). GAG chains are linear polysaccharides of alternating hexuronic acid and N-acetylhexosamine residues, commonly linked to core proteins to form proteoglycans with important roles in the regulation of a range of biological processes. The biosynthesis of GAGs is initiated by xylosylation of a serine residue of the core protein followed by galactosylation, catalyzed by ß4GalT7. The biosynthesis can also be initiated by xylosides carrying hydrophobic aglycons, such as 2-naphthyl ß-D-xylopyranoside. We have cloned and expressed ß4GalT7, and designed a cell-free assay to measure the activity of this enzyme. The assay employs a 96-well plate format for high throughput. In this chapter, we describe the cloning, expression, and purification of ß4GalT7, as well as assays proposed for development of substrates for GAG priming and for investigating inhibitors of ß4GalT7.


Subject(s)
N-Acetyllactosamine Synthase/metabolism , Chondroitin Sulfates , Glycosaminoglycans , N-Acetyllactosamine Synthase/genetics , Proteoglycans
11.
J Proteomics ; 268: 104717, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36084919

ABSTRACT

IgG N-glycans levels change with advancing age, making it a potential biomarker of aging. ß-1,4-galactosyltransferase (B4GALT) gene expression levels also increase with aging. Ultra performance liquid chromatography (UPLC) was used to examine changes inserum IgG N-glycans at six time points during the aging process. Most serum IgG N-glycans changed with aging in WT but not in CD19-cre B4GALT1 floxed mice. The relative abundance of fucosylated biantennary glycans with or without Neu5Gc structures changed with aging in heterozygous B4GALT1 floxed mice but not in homozygous B4GALT1 floxed mice. Additionally, the aging phenotype was more apparent in WT mice than in B4GALT1 floxed mice. These results demonstrate that fucosylated biantennary glycans and fucosylated biantennary glycans containing N-glycolylneuraminic acid (Neu5Gc)-linked N-acetyllactosamine (LacNAc) were highly associated with aging and were affected by the B4GALT1 floxed mouse genotype. The changing levels of fucosylated monoantennary glycans observed with aging in WT mice was reversed in B4GALT1 floxed mice and was not sex specific. In summary, B-cell-specific ablation of B4GALT1 from a glycoproteomic perspective prevented age-related changes in IgG N-glycans in mice. SIGNIFICANCE: In this study, serum IgG glycoproteomic data in wild-type (WT) and B-cell-specific ablation of ß-1,4-galactosyltransferase 1 mice (B4GALT) were analyzed. Results showed that fucosylated biantennary glycans with or without N-glycolylneuraminic acid (Neu5Gc)-linked N-acetyllactosamine (LacNAc) were highly associated with aging and were also affected by the B4GALT1 floxed mouse genotype. In terms of gender-specific information, the trend towards elevated fucosylated monoantennary glycans in WT mice was not seen in CD19-cre B4GALT1 floxed mice in either sex. B-cell-specific ablation of B4GALT1 plays an important role in age-related glycan changes; its specific functions and mechanisms are worthy of in-depth study. Our data suggest that investigating the relationship between galactosylation and aging may help advance the field of glycoproteomics and aging research.


Subject(s)
Aging , Immunoglobulin G , N-Acetyllactosamine Synthase , Polysaccharides , Aging/genetics , Aging/metabolism , Animals , B-Lymphocytes/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Mice , N-Acetyllactosamine Synthase/genetics , N-Acetyllactosamine Synthase/metabolism , Neuraminic Acids , Phenotype , Polysaccharides/chemistry
12.
J Biol Chem ; 285(20): 15619-15626, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20236943

ABSTRACT

The beta1,4-galactosyltransferase-7 (beta4Gal-T7) enzyme, one of seven members of the beta4Gal-T family, transfers in the presence of manganese Gal from UDP-Gal to an acceptor sugar (xylose) that is attached to a side chain hydroxyl group of Ser/Thr residues of proteoglycan proteins. It exhibits the least protein sequence similarity with the other family members, including the well studied family member beta4Gal-T1, which, in the presence of manganese, transfers Gal from UDP-Gal to GlcNAc. We report here the crystal structure of the catalytic domain of beta4Gal-T7 from Drosophila in the presence of manganese and UDP at 1.81 A resolution. In the crystal structure, a new manganese ion-binding motif (HXH) has been observed. Superposition of the crystal structures of beta4Gal-T7 and beta4Gal-T1 shows that the catalytic pocket and the substrate-binding sites in these proteins are similar. Compared with GlcNAc, xylose has a hydroxyl group (instead of an N-acetyl group) at C2 and lacks the CH(2)OH group at C5; thus, these protein structures show significant differences in their acceptor-binding site. Modeling of xylose in the acceptor-binding site of the beta4Gal-T7 crystal structure shows that the aromatic side chain of Tyr(177) interacts strongly with the C5 atom of xylose, causing steric hindrance to any additional group at C5. Because Drosophila Cd7 has a 73% protein sequence similarity to human Cd7, the present crystal structure offers a structure-based explanation for the mutations in human Cd7 that have been linked to Ehlers-Danlos syndrome.


Subject(s)
N-Acetyllactosamine Synthase/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Drosophila melanogaster , Humans , Manganese/metabolism , Models, Molecular , Molecular Sequence Data , N-Acetyllactosamine Synthase/genetics , N-Acetyllactosamine Synthase/metabolism , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Uridine Diphosphate/metabolism
13.
Inflamm Res ; 60(2): 169-74, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20963624

ABSTRACT

OBJECTIVE AND DESIGN: The carbohydrate moieties of glycoprotein are associated with some inflammatory diseases by affecting a wide range of biological functions of cells. This study aimed to investigate the role of ß-1,4-galactosyltransferase-I (ß-1,4-GalT-I) in adhesion of Schwann cells during inflammation. SUBJECTS: A rat Schwann cell line, RSC 96 was used. METHODS: We used western blotting to detect the expression of ß-1,4-GalT-I. Flow cytomety was used to measure the galactosylation of glycoproteins on cell surfaces. Immunofluorescent staining was used to examine the expression of α6 integrin, focal adhesion kinase (FAK) and F-actin. Tyrosine phosphorylation of FAK was detected by immunoprecipitation. An adhesion assay was performed to investigate the adhesion of Schwann cells. One-way ANOVA was used to compare differences between the operated and the control group. RESULTS: Schwann cell adhesion was induced by LPS stimulation and was accompanied by upregulation of ß-1,4-GalT-I expression and galactosylation of glycoproteins. There was a change of localization of FAK and cytoskeleton organization in LPS treated cells compared with control cells. The pretreated cells enhanced tyrosine phosphorylation of FAK compared with control cells in the adhesion process. With the increased cell surface expression of α6 integrin and ß-1,4-GalT-I, the adhesion of Schwann cells on laminin was increased as well. CONCLUSIONS: These results suggested that ß-1,4-GalT-I may play an important role in adhesion of Schwann cells during inflammation.


Subject(s)
Cell Adhesion/drug effects , Lipopolysaccharides/pharmacology , N-Acetyllactosamine Synthase/metabolism , Schwann Cells/drug effects , Schwann Cells/physiology , Animals , Cell Line , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alpha6/metabolism , Laminin/metabolism , Membrane Glycoproteins/metabolism , Rats , Schwann Cells/cytology , Signal Transduction/physiology , Tyrosine/metabolism
14.
Biol Cell ; 102(11): 593-607, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20735355

ABSTRACT

BACKGROUND INFORMATION: The GA (Golgi apparatus) has an essential role in membrane trafficking, determining the assembly and delivery of UPs (uroplakins) to the APM (apical plasma membrane) of superficial UCs (uroepithelial cells) of urinary bladder. UPs are synchronously and uniformly delivered from the GA to the APM by DFVs (discoidal- or fusiform-shaped vesicles); however, the mechanism of UP delivery is not known. We have used the culture model of UCs with the capacity to undergo terminal differentiation to study the process of uniform delivery of DFVs to the APM and to elucidate the mechanisms involved. RESULTS: By three-dimensional localization using confocal microscopy of immunofluorescence-labelled GA-related markers [GM130 (cis-Golgi matrix protein of 130 kDa), GS15 (Golgi Snare 15 kDa), GS28 and giantin], uroepithelial differentiation-related markers (UPs), MTs (microtubules; α-tubulin) and intermediate filaments [CK7 (cytokeratin 7) and CK20], we found that in non-differentiated, UP-negative UCs the GA is mostly organized as a single ribbon-like structure close to the nucleus, whereas in differentiated, UP-positive UCs the GA is fragmented and spread almost through the entire cell. The FRAP (fluorescence recovery after photobleaching) experiments on the UCs transfected with GalT (trans-Golgi/TGN enzyme ß1,4-galactosyltransferase) fused to fluorescent protein showed that Golgi-resident enzyme cycles freely within ribbon-like GA but not within fragmented GA. By CLEM (correlative light-electron microscopy), we examined the GA fragments in cells expressing UPs. We found that GA fragments are fully functional and similar to the GA fragments that are formed after nocodazole treatment. Furthermore, we demonstrated that the reorganization of GA into a fragmented form is associated with the impairment of the MT organization in the basal, central and subapical cytoplasm and the accumulation of intermediate filaments in the apical cytoplasm that could affect the kinetics of MT star leading to the peripheral fragmentation of the GA in the differentiated UCs. CONCLUSIONS: The fragmentation of the GA and the subsequent spreading of GA to the cell periphery represent one of the key events that promote the uniform delivery of UPs over the entire APM of differentiating UCs and thus are of major importance in the final proper formation and maintenance of the blood-urine barrier.


Subject(s)
Cell Differentiation/physiology , Cell Membrane/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Protein Transport , Urothelium/cytology , Animals , Cells, Cultured , Fluorescence Recovery After Photobleaching , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Hypertrophy , Intermediate Filaments/pathology , Keratin-20/metabolism , Keratin-7/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Microtubules/metabolism , N-Acetyllactosamine Synthase/metabolism , Nocodazole/pharmacology , Swine , Uroplakin II
15.
Biochemistry ; 49(11): 2604-14, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20178374

ABSTRACT

Recombinant glycosyltransferases are potential biocatalysts for the construction of a compound library of oligosaccharides, glycosphingolipids, glycopeptides, and various artificial glycoconjugates on the basis of combined chemical and enzymatic synthetic procedures. The structurally defined glycan-related compound library is a key resource both in the basic studies of their functional roles in various biological processes and in the discovery research of new diagnostic biomarkers and therapeutic reagents. Therefore, it is clear that the immobilization of extremely unstable membrane-bound glycosyltransferases on some suitable supporting materials should enhance the operational stability and activity of recombinant enzymes and makes facile separation of products and recycling use of enzymes possible. Until now, however, it seems that no standardized protocol preventing a significant loss of enzyme activity is available due to the lack of a general method of site-selective anchoring between glycosyltransferases and scaffold materials through a stable covalent bond. Here we communicate a versatile and efficient method for the immobilization of recombinant glycosyltransferases onto commercially available solid supports by means of transpeptidase reaction by Staphylococcus aureus sortase A. This protocol allowed for the first time highly specific conjugation at the designated C-terminal signal peptide moiety of recombinant human beta1,4-galactosyltransferase or recombinant Helicobacter pylori alpha1,3-fucosyltransferase with simple aliphatic amino groups displayed on the surface of solid materials. Site-specifically immobilized enzymes exhibited the desired sugar transfer activity, an improved stability, and a practical reusability required for rapid and large-scale synthesis of glycoconjugates. Considering that most mammalian enzymes responsible for the posttranslational modifications, including the protein kinase family, as well as glycosyltransferases are unstable and highly oriented membrane proteins, the merit of our strategy based on "site-specific" transpeptidation is evident because the reaction proceeds only at an engineered C-terminus without any conformational influence around the active sites of both enzymes as well as heptad repeats of rHFucT required to maintain native secondary and quaternary structures during the dimerization on cell surfaces.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Enzymes, Immobilized/metabolism , Glycosyltransferases/metabolism , Membrane Proteins/metabolism , Recombinant Proteins/metabolism , Staphylococcus aureus/enzymology , Amines/chemistry , Amino Acid Sequence , Animals , Binding Sites , Enzymes, Immobilized/chemistry , Fucosyltransferases/chemistry , Fucosyltransferases/metabolism , Glycosyltransferases/chemistry , Helicobacter pylori/enzymology , Humans , Lewis X Antigen/biosynthesis , Lewis X Antigen/chemistry , Membrane Proteins/chemistry , Models, Molecular , N-Acetyllactosamine Synthase/chemistry , N-Acetyllactosamine Synthase/metabolism , Protein Conformation , Protein Stability , Recombinant Proteins/chemistry , Sepharose/chemistry , Sepharose/metabolism , Substrate Specificity
16.
Bioorg Med Chem Lett ; 20(16): 4906-10, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20620057

ABSTRACT

An efficient synthesis of sialyllactosamine (SiaLacNAc) clusters using carbosilanes as core scaffolds has been accomplished by means of chemical and enzymatic approaches. N-Acetyl-D-glucosamine (GlcNAc) clusters having O-glycosidic linkage or S-glycosidic linkage were chemically synthesized from known intermediates in high yields. The GlcNAc clusters were first used as substrates for beta1,4 galactosyl transferase using UDP-galactose (UDP-Gal) as a sugar source to provide corresponding N-acetyllactosamine clusters. Further sugar elongation of the LacNAc clusters was demonstrated using alpha2,3 sialyl transferase and CMP-neuraminic acid (CMP-NANA) to yield the corresponding SiaLacNAc clusters.


Subject(s)
Amino Sugars/chemistry , Silanes/chemistry , Acetylglucosamine/biosynthesis , Acetylglucosamine/chemistry , Amino Sugars/biosynthesis , Amino Sugars/metabolism , Animals , Carbohydrate Sequence , Cattle , N-Acetyllactosamine Synthase/metabolism , Rats , Uridine Diphosphate Galactose/chemistry
17.
Plant Biotechnol J ; 7(5): 442-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19422604

ABSTRACT

Plant-based transient expression is potentially the most rapid and cost-efficient system for the production of recombinant pharmaceutical proteins, but safety concerns associated with plant-specific N-glycosylation have hampered its adoption as a commercial production system. In this article, we describe an approach based on the simultaneous transient co-expression of an antibody, a suppressor of silencing and a chimaeric human beta1,4-galactosyltransferase targeted for optimal activity to the early secretory pathway in agroinfiltrated Nicotiana benthamiana leaves. This strategy allows fast and high-yield production of antibodies with human-like N-glycans and, more generally, provides solutions to many critical problems posed by the large-scale production of therapeutic and vaccinal proteins, specifically yield, volume and quality.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Nicotiana/metabolism , Polysaccharides/metabolism , Protein Engineering/methods , Antibodies, Monoclonal/isolation & purification , Gene Expression Regulation, Plant , Glycosylation , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/isolation & purification , N-Acetyllactosamine Synthase/genetics , N-Acetyllactosamine Synthase/metabolism , Plant Leaves/genetics , Plant Leaves/metabolism , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Nicotiana/genetics
18.
J Cell Biol ; 117(2): 369-82, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1560031

ABSTRACT

Mesenchymal cell migration and neurite outgrowth are mediated in part by binding of cell surface beta 1,4-galactosyltransferase (GalTase) to N-linked oligosaccharides within the E8 domain of laminin. In this study, we determined whether cell surface GalTase functions during neural crest cell migration and neural development in vivo using antibodies raised against affinity-purified chicken serum GalTase. The antibodies specifically recognized two embryonic proteins of 77 and 67 kD, both of which express GalTase activity. The antibodies also immunoprecipitated and inhibited chick embryo GalTase activity, and inhibited neural crest cell migration on laminin matrices in vitro. Anti-GalTase antibodies were microinjected into the head mesenchyme of stage 7-9 chick embryos or cranial to Henson's node of stage 6 embryos. Anti-avian GalTase IgG decreased cranial neural crest cell migration on the injected side but did not cross the embryonic midline and did not affect neural crest cell migration on the uninjected side. Anti-avian GalTase Fab crossed the embryonic midline and perturbed cranial neural crest cell migration throughout the head. Neural fold elevation and neural tube closure were also disrupted by Fab fragments. Cell surface GalTase was localized to migrating neural crest cells and to the basal surfaces of neural epithelia by indirect immunofluorescence, whereas GalTase was undetectable on neural crest cells prior to migration. These results suggest that, during early embryogenesis, cell surface GalTase participates during neural crest cell migration, perhaps by interacting with laminin, a major component of the basal lamina. Cell surface GalTase also appears to play a role in neural tube formation, possibly by mediating neural epithelial adhesion to the underlying basal lamina.


Subject(s)
Central Nervous System/embryology , N-Acetyllactosamine Synthase/metabolism , Neural Crest/cytology , Animals , Blotting, Western , Cell Membrane/enzymology , Cell Movement , Chick Embryo , Culture Techniques , Fluorescent Antibody Technique , Immunoglobulin Fab Fragments , Immunoglobulin G , Laminin , Microinjections , N-Acetyllactosamine Synthase/immunology , Neural Crest/enzymology
19.
J Cell Biol ; 72(3): 617-27, 1977 Mar.
Article in English | MEDLINE | ID: mdl-402369

ABSTRACT

Fat globule membranes have been isolated from bovine colostrum and bovine milk by the dispersion of the fat in sucrose solutions at 4 degrees C and fractionation by centrifugation through discontinuous sucrose gradients. The morphology and enzymic characteristics of the separated fractions were examined. Fractions comprising a large proportion of the total extracted membrane were thus obtained having high levels of the Golgi marker enzymes UDP-galactose N-acetylglucosamine beta-4-galactosyltransferase and thiamine pyrophosphatase. A membrane-derived form of the galactosyltransferase has been solubilized from fat and purified to homogeneity. This enzyme is larger in molecular weight than previously studied soluble galactosyltransferases, but resembles in size the galactosyltransferase of lactating mammary Golgi membranes. In contrast, when fat globule membranes were prepared by traditional procedures, which involved washing the fat at higher temperatures, before extraction, galactosyltransferase was not present in the membranes, having been released into supernatant fractions, When the enzyme released by this procedure was partially purified and examined by gel filtration, it was found to be of a degraded form resembling in size the soluble galactosyltransferase of milk. The release is therefore attributed to the action of proteolytic enzymes. Our observations contrast with previous biochemical studies which suggested that Golgi membranes do not contribute to the milk fat globule membrane. They are, however, consistent with electron microscope studies of the fat secretion process, which indicate that secretory vesicle membranes, derived from the Golgi apparatus, may provide a large proportion of the fat globule membrane.


Subject(s)
Colostrum/enzymology , Fats , Golgi Apparatus/ultrastructure , Lactose Synthase/metabolism , Milk/enzymology , N-Acetyllactosamine Synthase/metabolism , Pyrophosphatases/metabolism , Thiamine Pyrophosphatase/metabolism , Animals , Cattle , Golgi Apparatus/enzymology , Membranes/enzymology
20.
Science ; 273(5276): 797-801, 1996 Aug 09.
Article in English | MEDLINE | ID: mdl-8670420

ABSTRACT

The mechanism by which Golgi membrane proteins are retained within the Golgi complex in the midst of a continuous flow of protein and lipid is not yet understood. The diffusional mobilities of mammalian Golgi membrane proteins fused with green fluorescent protein from Aequorea victoria were measured in living HeLa cells with the fluorescence photobleaching recovery technique. The diffusion coefficients ranged from 3 x 10(-9) square centimeters per second to 5 x 10(-9) square centimeters per second, with greater than 90 percent of the chimeric proteins mobile. Extensive lateral diffusion of the chimeric proteins occurred between Golgi stacks. Thus, the chimeras diffuse rapidly and freely in Golgi membranes, which suggests that Golgi targeting and retention of these molecules does not depend on protein immobilization.


Subject(s)
Golgi Apparatus/metabolism , Mannosidases/metabolism , Membrane Proteins/metabolism , N-Acetyllactosamine Synthase/metabolism , Receptors, Peptide/metabolism , Aluminum Compounds/pharmacology , Diffusion , Endoplasmic Reticulum/metabolism , Fluorides/pharmacology , Green Fluorescent Proteins , HeLa Cells , Humans , Intracellular Membranes/metabolism , Luminescent Proteins , Microscopy, Confocal , Mutation , Receptors, Peptide/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL