Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 597(18): 2345-2357, 2023 09.
Article in English | MEDLINE | ID: mdl-37552213

ABSTRACT

SLC35A2 and SLC35A3 are homologous proteins with postulated nucleotide sugar transporting activities. Unlike SLC35A2, whose specificity for UDP-Gal is well-established, the UDP-GlcNAc transporting activity initially attributed to SLC35A3 has recently been put into question. In this study, we constructed two hybrid proteins (SLC35A2-SLC35A3 and SLC35A3-SLC35A2) and expressed them in a previously generated SLC35A2/SLC35A3 double knockout HEK293T cell line. Our idea was to force equivalent stoichiometry of the two proteins in the cells in order to reproduce the behavior of the SLC35A2/SLC35A3 complexes in the Golgi membrane. The hybrid proteins were able to fully restore glycosylation in the double knockout. In contrast, the expression of SLC35A3 alone in these cells improved galactosylation only to a limited extent. Our study shows that the proper glycosylation requires a balanced cooperation between SLC35A2 and SLC35A3.


Subject(s)
Monosaccharide Transport Proteins , Uridine Diphosphate , Humans , Glycosylation , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , HEK293 Cells , Biological Transport , Uridine Diphosphate/metabolism
2.
Biochim Biophys Acta Mol Cell Res ; 1870(5): 119462, 2023 06.
Article in English | MEDLINE | ID: mdl-36933771

ABSTRACT

Nucleotide sugar transporters (NSTs) are multitransmembrane proteins, localized in the Golgi apparatus and/or endoplasmic reticulum, which provide glycosylation enzymes with their substrates. It has been demonstrated that NSTs may form complexes with functionally related glycosyltransferases, especially in the N-glycosylation pathway. However, potential interactions of NSTs with enzymes mediating the biosynthesis of mucin-type O-glycans have not been addressed to date. Here we report that UDP-galactose transporter (UGT; SLC35A2) associates with core 1 ß-1,3-galactosyltransferase 1 (C1GalT1; T-synthase). This provides the first example of an interaction between an enzyme that acts exclusively in the O-glycosylation pathway and an NST. We also found that SLC35A2 associated with the C1GalT1-specific chaperone Cosmc, and that the endogenous Cosmc was localized in both the endoplasmic reticulum and Golgi apparatus of wild-type HEK293T cells. Furthermore, in SLC35A2-deficient cells protein levels of C1GalT1 and Cosmc were decreased and their Golgi localization was less pronounced. Finally, we identified SLC35A2 as a novel molecular target for the antifungal agent itraconazole. Based on our findings we propose that NSTs may contribute to the stabilization of their interaction partners and help them to achieve target localization in the cell, most likely by facilitating their assembly into larger functional units.


Subject(s)
Galactosyltransferases , Molecular Chaperones , Humans , HEK293 Cells , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Glycosylation , Golgi Apparatus/genetics , Golgi Apparatus/metabolism
3.
Biochem Biophys Res Commun ; 635: 46-51, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36257191

ABSTRACT

The sialylation of glycoconjugates is performed by a variety of sialyltransferases using CMP-sialic acid (CMP-Sia) as a substrate. Sialylation requires the translocation of CMP-Sia across the Golgi membranes. This function has been assigned to SLC35A1, the only CMP-Sia transporter identified to date. Mutations in the SLC35A1 gene cause a subtype of congenital disorder of glycosylation (CDG). Over the past several years, heterologous complexes formed in the Golgi membrane by some SLC35A subfamily members and functionally related glycosyltransferases have been reported. However, to date no interaction between SLC35A1 and a sialyltransferase has been identified. In this study we attempted to clarify the role of SLC35A1 in α2,3 sialylation of N-glycans. We showed that SLC35A1 associates with ST3Gal4, the main α2,3-sialyltransferase acting on N-glycans. This phenomenon is compromised by the E196K (but not T156R) mutation in the SLC35A1 gene. We also demonstrated that the E196K mutant is less efficient in restoring N-glycan sialylation upon expression in the SLC35A1 knockout cells. On the basis of our findings, we propose that the interaction between SLC35A1 and ST3Gal4 may be important for proper sialylation.


Subject(s)
Cytidine Monophosphate N-Acetylneuraminic Acid , Sialyltransferases , Sialyltransferases/genetics , Sialyltransferases/metabolism , Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Glycosylation , Mutation , Polysaccharides
4.
Int J Mol Sci ; 23(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35955785

ABSTRACT

Nucleotide sugars (NSs) serve as substrates for glycosylation reactions. The majority of these compounds are synthesized in the cytoplasm, whereas glycosylation occurs in the endoplasmic reticulum (ER) and Golgi lumens, where catalytic domains of glycosyltransferases (GTs) are located. Therefore, translocation of NS across the organelle membranes is a prerequisite. This process is thought to be mediated by a group of multi-transmembrane proteins from the SLC35 family, i.e., nucleotide sugar transporters (NSTs). Despite many years of research, some uncertainties/inconsistencies related with the mechanisms of NS transport and the substrate specificities of NSTs remain. Here we present a comprehensive review of the NS import into the mammalian Golgi, which consists of three major parts. In the first part, we provide a historical view of the experimental approaches used to study NS transport and evaluate the most important achievements. The second part summarizes various aspects of knowledge concerning NSTs, ranging from subcellular localization up to the pathologies related with their defective function. In the third part, we present the outcomes of our research performed using mammalian cell-based models and discuss its relevance in relation to the general context.


Subject(s)
Golgi Apparatus , Nucleotides , Sugars , Animals , Biological Transport , Glycosylation , Golgi Apparatus/metabolism , Mammals/metabolism , Nucleotides/metabolism , Sugars/metabolism
5.
Cells ; 11(15)2022 07 23.
Article in English | MEDLINE | ID: mdl-35892570

ABSTRACT

In mammalian cells, SLC35A2 delivers UDP-galactose for galactosylation reactions that take place predominantly in the Golgi lumen. Mutations in the corresponding gene cause a subtype of a congenital disorder of glycosylation (SLC35A2-CDG). Although more and more patients are diagnosed with SLC35A2-CDG, the link between defective galactosylation and disease symptoms is not fully understood. According to a number of reports, impaired glycosylation may trigger the process of epithelial-to-mesenchymal transition (EMT). We therefore examined whether the loss of SLC35A2 activity would promote EMT in a non-malignant epithelial cell line. For this purpose, we knocked out the SLC35A2 gene in Madin-Darby canine kidney (MDCK) cells. The resulting clones adopted an elongated, spindle-shaped morphology and showed impaired cell-cell adhesion. Using qPCR and western blotting, we revealed down-regulation of E-cadherin in the knockouts, while the fibronectin and vimentin levels were elevated. Moreover, the knockout cells displayed reorganization of vimentin intermediate filaments and altered subcellular distribution of a vimentin-binding protein, formiminotransferase cyclodeaminase (FTCD). Furthermore, depletion of SLC35A2 triggered Golgi compaction. Finally, the SLC35A2 knockouts displayed increased motility and invasiveness. In conclusion, SLC35A2-deficient MDCK cells showed several hallmarks of EMT. Our findings point to a novel role for SLC35A2 as a gatekeeper of the epithelial phenotype.


Subject(s)
Epithelial-Mesenchymal Transition , Kidney , Animals , Dogs , Kidney/metabolism , Madin Darby Canine Kidney Cells , Mammals , Phenotype , Vimentin/metabolism
6.
J Biol Chem ; 298(8): 102206, 2022 08.
Article in English | MEDLINE | ID: mdl-35772493

ABSTRACT

Mutations in the SLC35C1 gene encoding the Golgi GDP-fucose transporter are known to cause leukocyte adhesion deficiency II. However, improvement of fucosylation in leukocyte adhesion deficiency II patients treated with exogenous fucose suggests the existence of an SLC35C1-independent route of GDP-fucose transport, which remains a mystery. To investigate this phenomenon, we developed and characterized a human cell-based model deficient in SLC35C1 activity. The resulting cells were cultured in the presence/absence of exogenous fucose and mannose, followed by examination of fucosylation potential and nucleotide sugar levels. We found that cells displayed low but detectable levels of fucosylation in the absence of SLC35C1. Strikingly, we show that defects in fucosylation were almost completely reversed upon treatment with millimolar concentrations of fucose. Furthermore, we show that even if fucose was supplemented at nanomolar concentrations, it was still incorporated into glycans by these knockout cells. We also found that the SLC35C1-independent transport preferentially utilized GDP-fucose from the salvage pathway over the de novo biogenesis pathway as a source of this substrate. Taken together, our results imply that the Golgi systems of GDP-fucose transport discriminate between substrate pools obtained from different metabolic pathways, which suggests a functional connection between nucleotide sugar transporters and nucleotide sugar synthases.


Subject(s)
Fucose , Guanosine Diphosphate Fucose , Leukocyte-Adhesion Deficiency Syndrome/therapy , Fucose/metabolism , Golgi Apparatus/metabolism , Guanosine Diphosphate Fucose/metabolism , Humans , Membrane Transport Proteins/metabolism , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Polysaccharides/metabolism
7.
J Proteomics ; 249: 104321, 2021 10 30.
Article in English | MEDLINE | ID: mdl-34242836

ABSTRACT

Nucleotide sugar transporters (NSTs) are ER and Golgi-resident members of the solute carrier 35 (SLC35) family which supply substrates for glycosylation by exchanging lumenal nucleotide monophosphates for cytosolic nucleotide sugars. Defective NSTs have been associated with congenital disorders of glycosylation (CDG), however, molecular basis of many types of CDG remains poorly characterized. To better understand the biology of NSTs, we identified potential interaction partners of UDP-galactose transporter (SLC35A2), UDP-N-acetylglucosamine transporter (SLC35A3) and an orphan nucleotide sugar transporter SLC35A4 of to date unassigned specificity. For this purpose, each of the SLC35A2-A4 proteins was used as a bait in four independent pull-down experiments and the identity of the immunoprecipitated material was discovered using MS techniques. From the candidate list obtained, we selected a few for which the interaction was confirmed in vitro using the NanoBiT system, a split luciferase-based luminescent technique. NSTs have been shown to interact with two ATPases (ATP2A2, ATP2C1), Golgi pH regulator B (GPR89B) and calcium channel (TMCO1), which may reflect the regulation of glycosylation by ion homeostasis, and with basigin (BSG). Our findings provide a starting point for the NST interaction network discovery in order to better understand how glycosylation is regulated and linked to other cellular processes. SIGNIFICANCE: Despite the facts that nucleotide sugar transporters are a key component of the protein glycosylation machinery, and deficiencies in their activity underlie serious metabolic diseases, biology, function and regulation of these essential proteins remain enigmatic. In this study we have advanced the field by identifying sets of new potential interaction partners for UDP-galactose transporter (SLC35A2), UDP-N-acetylglucosamine transporter (SLC35A3) and an orphan transporter SLC35A4 of yet undefined role. Several of these new interactions were additionally confirmed in vitro using the NanoBiT system, a split luciferase complementation assay. This work is also significant in that it addresses the overall challenge of discovering membrane protein interaction partners by a detailed comparison of 4 different co-immunoprecipitation strategies and by custom sample preparation and data processing workflows.


Subject(s)
Acetylglucosamine , Uridine Diphosphate Galactose , Biological Transport , Galactose/metabolism , Golgi Apparatus/metabolism , Nucleotides/metabolism , Uridine Diphosphate Galactose/metabolism
8.
J Biol Chem ; 295(48): 16445-16463, 2020 11 27.
Article in English | MEDLINE | ID: mdl-32938718

ABSTRACT

Nucleotide sugar transporters, encoded by the SLC35 gene family, deliver nucleotide sugars throughout the cell for various glycosyltransferase-catalyzed glycosylation reactions. GlcNAc, in the form of UDP-GlcNAc, and galactose, as UDP-Gal, are delivered into the Golgi apparatus by SLC35A3 and SLC35A2 transporters, respectively. However, although the UDP-Gal transporting activity of SLC35A2 has been clearly demonstrated, UDP-GlcNAc delivery by SLC35A3 is not fully understood. Therefore, we analyzed a panel of CHO, HEK293T, and HepG2 cell lines including WT cells, SLC35A2 knockouts, SLC35A3 knockouts, and double-knockout cells. Cells lacking SLC35A2 displayed significant changes in N- and O-glycan synthesis. However, in SLC35A3-knockout CHO cells, only limited changes were observed; GlcNAc was still incorporated into N-glycans, but complex type N-glycan branching was impaired, although UDP-GlcNAc transport into Golgi vesicles was not decreased. In SLC35A3-knockout HEK293T cells, UDP-GlcNAc transport was significantly decreased but not completely abolished. However, N-glycan branching was not impaired in these cells. In CHO and HEK293T cells, the effect of SLC35A3 deficiency on N-glycan branching was potentiated in the absence of SLC35A2. Moreover, in SLC35A3-knockout HEK293T and HepG2 cells, GlcNAc was still incorporated into O-glycans. However, in the case of HepG2 cells, no qualitative changes in N-glycans between WT and SLC35A3 knockout cells nor between SLC35A2 knockout and double-knockout cells were observed. These findings suggest that SLC35A3 may not be the primary UDP-GlcNAc transporter and/or different mechanisms of UDP-GlcNAc transport into the Golgi apparatus may exist.


Subject(s)
Glycosyltransferases/metabolism , Golgi Apparatus/metabolism , Nucleotide Transport Proteins/metabolism , Polysaccharides/biosynthesis , Animals , CHO Cells , Cricetulus , Gene Knockdown Techniques , Glycosyltransferases/genetics , Golgi Apparatus/genetics , HEK293 Cells , Hep G2 Cells , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Nucleotide Transport Proteins/genetics , Polysaccharides/genetics
9.
J Vis Exp ; (163)2020 09 10.
Article in English | MEDLINE | ID: mdl-32986032

ABSTRACT

The goal of this protocol is to explore the applicability of the most recent variant of split luciferase complementation for demonstrating heterologous complexes formed by nucleotide sugar transporters (NSTs). These ER- and Golgi-resident multitransmembrane proteins carry the cytoplasmically synthesized nucleotide sugars across organelle membranes to supply enzymes that mediate glycosylation with their substrates. NSTs exist as dimers and/or higher oligomers. Heterologous interactions between different NSTs have also been reported. To verify whether the technique is suitable for studying the phenomenon of NST heteromerization, we tested it against a combination of the two Golgi-resident NSTs that have been previously shown to associate by several other means. The luciferase complementation assay appears to be particularly suitable for studying interactions between Golgi-resident membrane proteins, as it does not require high expression levels, which often trigger protein mislocalization and increase the risk of false positives.


Subject(s)
Golgi Apparatus/metabolism , Luciferases/metabolism , Membrane Proteins/metabolism , Animals , Biological Transport , Glycosylation , Humans
10.
Glycoconj J ; 37(5): 577-588, 2020 10.
Article in English | MEDLINE | ID: mdl-32827291

ABSTRACT

ß1,4-galactosyltransferase 4 (B4GalT4) is one of seven B4GalTs that belong to CAZy glycosyltransferase family 7 and transfer galactose to growing sugar moieties of proteins, glycolipids, glycosaminoglycans as well as single sugar for lactose synthesis. Herein, we identify two asparagine-linked glycosylation sites in B4GalT4. We found that mutation of one site (Asn220) had greater impact on enzymatic activity while another (Asn335) on Golgi localization and presence of N-glycans at both sites is required for production of stable and enzymatically active protein and its secretion. Additionally, we confirm B4GalT4 involvement in synthesis of keratan sulfate (KS) by generating A375 B4GalT4 knock-out cell lines that show drastic decrease in the amount of KS proteoglycans and no significant structural changes in N- and O-glycans. We show that KS decrease in A375 cells deficient in B4GalT4 activity can be rescued by overproduction of either partially or fully glycosylated B4GalT4 but not with N-glycan-depleted B4GalT4 version.


Subject(s)
Galactosyltransferases/genetics , Glycosaminoglycans/genetics , Golgi Apparatus/genetics , Polysaccharides/genetics , Cell Line , Galactose/genetics , Galactosyltransferases/chemistry , Gene Knockout Techniques , Glycosaminoglycans/chemistry , Glycosylation , Humans , Keratan Sulfate/chemistry , Polysaccharides/metabolism
11.
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
12.
Int J Mol Sci ; 22(1)2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33396746

ABSTRACT

Congenital disorders of glycosylation (CDG) are a group of rare genetic and metabolic diseases caused by alterations in glycosylation pathways. Five patients bearing CDG-causing mutations in the SLC35A1 gene encoding the CMP-sialic acid transporter (CST) have been reported to date. In this study we examined how specific mutations in the SLC35A1 gene affect the protein's properties in two previously described SLC35A1-CDG cases: one caused by a substitution (Q101H) and another involving a compound heterozygous mutation (T156R/E196K). The effects of single mutations and the combination of T156R and E196K mutations on the CST's functionality was examined separately in CST-deficient HEK293T cells. As shown by microscopic studies, none of the CDG-causing mutations affected the protein's proper localization in the Golgi apparatus. Cellular glycophenotypes were characterized using lectins, structural assignment of N- and O-glycans and analysis of glycolipids. Single Q101H, T156R and E196K mutants were able to partially restore sialylation in CST-deficient cells, and the deleterious effect of a single T156R or E196K mutation on the CST functionality was strongly enhanced upon their combination. We also revealed differences in the ability of CST variants to form dimers. The results of this study improve our understanding of the molecular background of SLC35A1-CDG cases.


Subject(s)
Mutation , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Symporters/genetics , Symporters/metabolism , CRISPR-Cas Systems , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Cytidine Monophosphate/metabolism , Flow Cytometry , Gene Knockdown Techniques , Genetic Association Studies , Genetic Predisposition to Disease , Glycoconjugates/metabolism , Glycosylation , HEK293 Cells , Humans , Lectins/metabolism
13.
Int J Mol Sci ; 20(2)2019 Jan 11.
Article in English | MEDLINE | ID: mdl-30641943

ABSTRACT

Solute carrier family 35 member A5 (SLC35A5) is a member of the SLC35A protein subfamily comprising nucleotide sugar transporters. However, the function of SLC35A5 is yet to be experimentally determined. In this study, we inactivated the SLC35A5 gene in the HepG2 cell line to study a potential role of this protein in glycosylation. Introduced modification affected neither N- nor O-glycans. There was also no influence of the gene knock-out on glycolipid synthesis. However, inactivation of the SLC35A5 gene caused a slight increase in the level of chondroitin sulfate proteoglycans. Moreover, inactivation of the SLC35A5 gene resulted in the decrease of the uridine diphosphate (UDP)-glucuronic acid, UDP-N-acetylglucosamine, and UDP-N-acetylgalactosamine Golgi uptake, with no influence on the UDP-galactose transport activity. Further studies demonstrated that SLC35A5 localized exclusively to the Golgi apparatus. Careful insight into the protein sequence revealed that the C-terminus of this protein is extremely acidic and contains distinctive motifs, namely DXEE, DXD, and DXXD. Our studies show that the C-terminus is directed toward the cytosol. We also demonstrated that SLC35A5 formed homomers, as well as heteromers with other members of the SLC35A protein subfamily. In conclusion, the SLC35A5 protein might be a Golgi-resident multiprotein complex member engaged in nucleotide sugar transport.


Subject(s)
Golgi Apparatus/metabolism , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Solute Carrier Proteins/genetics , Solute Carrier Proteins/metabolism , Uridine Diphosphate Sugars/metabolism , Amino Acid Motifs , Chondroitin Sulfate Proteoglycans/metabolism , Cytosol/metabolism , Gene Knockout Techniques , Glycosylation , Hep G2 Cells , Humans , Nucleotide Transport Proteins/chemistry , Uridine Diphosphate Glucuronic Acid/metabolism , Uridine Diphosphate N-Acetylglucosamine/metabolism
14.
PLoS One ; 13(11): e0207521, 2018.
Article in English | MEDLINE | ID: mdl-30458018

ABSTRACT

SLC35B4 belongs to the solute carrier 35 (SLC35) family whose best-characterized members display a nucleotide sugar transporting activity. Using an experimental model of HepG2 cells and indirect immunofluorescent staining, we verified that SLC35B4 was localized to the endoplasmic reticulum (ER). We demonstrated that dilysine motif, especially lysine at position 329, is crucial for the ER localization of this protein in human cells and therefore one should use protein C-tagging with caution. To verify the importance of the protein in glycoconjugates synthesis, we generated SLC35B4-deficient HepG2 cell line using CRISPR-Cas9 approach. Our data showed that knock-out of the SLC35B4 gene does not affect major UDP-Xyl- and UDP-GlcNAc-dependent glycosylation pathways.


Subject(s)
Amino Acid Motifs/genetics , Endoplasmic Reticulum/chemistry , Golgi Apparatus/chemistry , Nucleotide Transport Proteins/chemistry , Amino Acid Sequence/genetics , CRISPR-Cas Systems/genetics , Dipeptides/chemistry , Dipeptides/genetics , Endoplasmic Reticulum/genetics , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Glycosylation , Golgi Apparatus/genetics , Hep G2 Cells , Humans , Lysine/chemistry , Lysine/genetics , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Signal Transduction , Uridine Diphosphate Sugars/chemistry
15.
Biochim Biophys Acta Mol Cell Res ; 1864(5): 825-838, 2017 May.
Article in English | MEDLINE | ID: mdl-28167211

ABSTRACT

SLC35A4 has been classified in the SLC35A subfamily based on amino acid sequence homology. Most of the proteins belonging to the SLC35 family act as transporters of nucleotide sugars. In this study, the subcellular localization of endogenous SLC35A4 was determined via immunofluorescence staining, and it was demonstrated that SLC35A4 localizes mainly to the Golgi apparatus. In silico topology prediction suggests that SLC35A4 has an uneven number of transmembrane domains and its N-terminus is directed towards the Golgi lumen. However, an experimental assay refuted this prediction: SLC35A4 has an even number of transmembrane regions with both termini facing the cytosol. In vivo interaction analysis using the FLIM-FRET approach revealed that SLC35A4 neither forms homomers nor associates with other members of the SLC35A subfamily except SLC35A5. Additional assays demonstrated that endogenous SLC35A4 is 10 to 40nm proximal to SLC35A2 and SLC35A3. To determine SLC35A4 function SLC35A4 knock-out cells were generated with the CRISPR-Cas9 approach. Although no significant changes in glycosylation were observed, the introduced mutation influenced the subcellular distribution of the SLC35A2/SLC35A3 complexes. Additional FLIM-FRET experiments revealed that overexpression of SLC35A4-BFP together with SLC35A3 and the SLC35A2-Golgi splice variant negatively affects the interaction between the two latter proteins. The results presented here strongly indicate a modulatory role for SLC35A4 in intracellular trafficking of SLC35A2/SLC35A3 complexes.


Subject(s)
Monosaccharide Transport Proteins/physiology , Nucleotide Transport Proteins/physiology , Amino Acid Sequence , Animals , Biological Transport/genetics , COS Cells , Carbohydrate Metabolism/genetics , Cell Line, Tumor , Chlorocebus aethiops , Dogs , HEK293 Cells , Hep G2 Cells , Humans , Madin Darby Canine Kidney Cells , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Nucleotide Transport Proteins/chemistry , Nucleotide Transport Proteins/genetics , Sequence Homology, Amino Acid
16.
Methods Mol Biol ; 1496: 133-43, 2016.
Article in English | MEDLINE | ID: mdl-27632007

ABSTRACT

In situ proximity ligation assay (PLA) is a novel, revolutionary technique that can be employed to visualize protein complexes in fixed cells and tissues. This approach enables demonstration of close (i.e., up to 40 nm) proximity between any two proteins of interest that can be detected using a pair of specific antibodies that have been raised in distinct species. Primary antibodies bound to the target proteins are subsequently recognized by two PLA probes, i.e., secondary antibodies conjugated with oligonucleotides that anneal when brought into close proximity in the presence of two connector oligonucleotides and a DNA ligase forming a circular DNA molecule. In the next step, the resulting circular DNA is amplified by a rolling circle polymerase. Finally, fluorescent oligonucleotide probes hybridize to complementary fragments of the amplified DNA molecule, forming a typical, spot-like pattern of PLA signal that reflects subcellular localization of protein complexes. Here we describe the use of in situ PLA in adherent cultures of mammalian cells in order to visualize interactions between Golgi-resident, functionally related glycosyltransferases and nucleotide sugar transporters relevant to N-glycan biosynthesis.


Subject(s)
Antibodies/chemistry , Carrier Proteins/chemistry , Fluorescent Dyes/chemistry , Glucosyltransferases/chemistry , Golgi Apparatus/chemistry , Multiprotein Complexes , Oligonucleotides/chemistry , Animals , COS Cells , Carrier Proteins/metabolism , Chlorocebus aethiops , DNA Ligases/chemistry , Glucosyltransferases/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism
17.
J Biol Chem ; 290(25): 15475-15486, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25944901

ABSTRACT

UDP-galactose transporter (UGT; SLC35A2) and UDP-N-acetylglucosamine transporter (NGT; SLC35A3) form heterologous complexes in the Golgi membrane. NGT occurs in close proximity to mannosyl (α-1,6-)-glycoprotein ß-1,6-N-acetylglucosaminyltransferase (Mgat5). In this study we analyzed whether NGT and both splice variants of UGT (UGT1 and UGT2) are able to interact with four different mannoside acetylglucosaminyltransferases (Mgat1, Mgat2, Mgat4B, and Mgat5). Using an in situ proximity ligation assay, we found that all examined glycosyltransferases are in the vicinity of these UDP-sugar transporters both at the endogenous level and upon overexpression. This observation was confirmed via the FLIM-FRET approach for both NGT and UGT1 complexes with Mgats. This study reports for the first time close proximity between endogenous nucleotide sugar transporters and glycosyltransferases. We also observed that among all analyzed Mgats, only Mgat4B occurs in close proximity to UGT2, whereas the other three Mgats are more distant from UGT2, and it was only possible to visualize their vicinity using proximity ligation assay. This strongly suggests that the distance between these protein pairs is longer than 10 nm but at the same time shorter than 40 nm. This study adds to the understanding of glycosylation, one of the most important post-translational modifications, which affects the majority of macromolecules. Our research shows that complex formation between nucleotide sugar transporters and glycosyltransferases might be a more common phenomenon than previously thought.


Subject(s)
Golgi Apparatus/metabolism , Monosaccharide Transport Proteins/metabolism , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational/physiology , Animals , Biological Transport, Active/physiology , Cell Line, Tumor , Dogs , Fluorescence Resonance Energy Transfer , Glycosylation , Golgi Apparatus/chemistry , Golgi Apparatus/genetics , Humans , Madin Darby Canine Kidney Cells , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/genetics
18.
J Biol Chem ; 288(30): 21850-60, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23766508

ABSTRACT

SLC35A3 is considered the main UDP-N-acetylglucosamine transporter (NGT) in mammals. Detailed analysis of NGT is restricted because mammalian mutant cells defective in this activity have not been isolated. Therefore, using the siRNA approach, we developed and characterized several NGT-deficient mammalian cell lines. CHO, CHO-Lec8, and HeLa cells deficient in NGT activity displayed a decrease in the amount of highly branched tri- and tetraantennary N-glycans, whereas monoantennary and diantennary ones remained unchanged or even were accumulated. Silencing the expression of NGT in Madin-Darby canine kidney II cells resulted in a dramatic decrease in the keratan sulfate content, whereas no changes in biosynthesis of heparan sulfate were observed. We also demonstrated for the first time close proximity between NGT and mannosyl (α-1,6-)-glycoprotein ß-1,6-N-acetylglucosaminyltransferase (Mgat5) in the Golgi membrane. We conclude that NGT may be important for the biosynthesis of highly branched, multiantennary complex N-glycans and keratan sulfate. We hypothesize that NGT may specifically supply ß-1,3-N-acetylglucosaminyl-transferase 7 (ß3GnT7), Mgat5, and possibly mannosyl (α-1,3-)-glycoprotein ß-1,4-N-acetylglucosaminyltransferase (Mgat4) with UDP-GlcNAc.


Subject(s)
Keratan Sulfate/biosynthesis , Membrane Transport Proteins/metabolism , Polysaccharides/biosynthesis , RNA Interference , Animals , Base Sequence , Biological Transport , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Dogs , Fluorescence Resonance Energy Transfer , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Humans , Membrane Transport Proteins/genetics , Microscopy, Confocal , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , N-Acetylglucosaminyltransferases/metabolism , Sequence Analysis, DNA , Uridine Diphosphate Sugars/metabolism
19.
Biochem Biophys Res Commun ; 434(3): 473-8, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23583405

ABSTRACT

The role of UDP-galactose transporter (UGT; SLC35A2) and UDP-N-acetylglucosamine transporter (NGT; SLC35A3) in glycosylation of macromolecules may be coupled and either of the transporters may partially replace the function played by its partner. The aim of this study was to construct chimeric transporters composed of the N-terminal portion of human NGT and the C-terminal portion of human UGT1 or UGT2 (NGT-UGT1 or NGT-UGT2, respectively), as well as of the N-terminal portion of UGT and C-terminal portion of NGT (UGT-NGT), overexpress them stably in UGT-deficient CHO-Lec8 and MDCK-RCA(r) cells, and characterize their involvement in glycosylation. Two chimeric proteins, NGT-UGT1 and NGT-UGT2, did not overexpress properly. In contrast, UGT-NGT chimeric protein was successfully overexpressed and localized properly to the Golgi apparatus. UGT-NGT chimeric transporter delivered UDP-Gal to the Golgi vesicles of UGT-deficient cells, which resulted in the increased content of galactosylated structures to such an extent that the wild-type phenotypes were completely restored. Our data further support our hypothesis that UGT and NGT cooperate in the UDP-Gal delivery for glycosyltransferases located in the Golgi apparatus. In a wider context, the results gained in this study add to our understanding of glycosylation, one of the basic posttranslational modifications, which affects the majority of macromolecules.


Subject(s)
Monosaccharide Transport Proteins/metabolism , Mutation , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Glycosylation , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , Mutagenesis, Site-Directed , Sequence Homology, Amino Acid , Uridine Diphosphate N-Acetylgalactosamine/metabolism
20.
FEBS Lett ; 586(23): 4082-7, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23089177

ABSTRACT

UDP-galactose transporter (UGT; SLC35A2) and UDP-N-acetylglucosamine transporter (NGT; SLC35A3) are evolutionarily related. We hypothesize that their role in glycosylation may be coupled through heterologous complex formation. Coimmunoprecipitation analysis and FLIM-FRET measurements performed on living cells showed that NGT and UGT form complexes when overexpressed in MDCK-RCA(r) cells. We also postulate that the interaction of NGT and UGT may explain the dual localization of UGT2. For the first time we demonstrated in vivo homodimerization of the NGT nucleotide sugar transporter. In conclusion, we suggest that NGT and UGT function in glycosylation is combined via their mutual interaction.


Subject(s)
Golgi Apparatus/metabolism , Membrane Transport Proteins/metabolism , Monosaccharide Transport Proteins/metabolism , Animals , Cell Line , Dogs , Immunoprecipitation , Membrane Transport Proteins/genetics , Monosaccharide Transport Proteins/genetics , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...