Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 300
Filter
1.
Nat Chem Biol ; 20(6): 732-741, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38321209

ABSTRACT

Glycosylation is a critical post-translational protein modification that affects folding, half-life and functionality. Glycosylation is a non-templated and heterogeneous process because of the promiscuity of the enzymes involved. We describe a platform for sequential glycosylation reactions for tailored sugar structures (SUGAR-TARGET) that allows bespoke, controlled N-linked glycosylation in vitro enabled by immobilized enzymes produced with a one-step immobilization/purification method. We reconstruct a reaction cascade mimicking a glycosylation pathway where promiscuity naturally exists to humanize a range of proteins derived from different cellular systems, yielding near-homogeneous glycoforms. Immobilized ß-1,4-galactosyltransferase is used to enhance the galactosylation profile of three IgGs, yielding 80.2-96.3% terminal galactosylation. Enzyme recycling is demonstrated for a reaction time greater than 80 h. The platform is easy to implement, modular and reusable and can therefore produce homogeneous glycan structures derived from various hosts for functional and clinical evaluation.


Subject(s)
Enzymes, Immobilized , Galactosyltransferases , Glycosylation , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Humans , Galactosyltransferases/metabolism , Galactosyltransferases/chemistry , Polysaccharides/metabolism , Polysaccharides/chemistry , Protein Processing, Post-Translational
2.
Mol Cell Proteomics ; 22(8): 100595, 2023 08.
Article in English | MEDLINE | ID: mdl-37328064

ABSTRACT

B4GALT1 encodes ß-1,4-galactosyltransferase 1, an enzyme that plays a major role in glycan synthesis in the Golgi apparatus by catalyzing the addition of terminal galactose. Studies increasingly suggest that B4GALT1 may be involved in the regulation of lipid metabolism pathways. Recently, we discovered a single-site missense variant Asn352Ser (N352S) in the functional domain of B4GALT1 in an Amish population, which decreases the level of LDL-cholesterol (LDL-c) as well as the protein levels of ApoB, fibrinogen, and IgG in the blood. To systematically evaluate the effects of this missense variant on protein glycosylation, expression, and secretion, we developed a nano-LC-MS/MS-based platform combined with TMT-labeling for in-depth quantitative proteomic and glycoproteomic analyses in the plasma of individuals homozygous for the B4GALT1 missense variant N352S versus non-carriers (n = 5 per genotype). A total of 488 secreted proteins in the plasma were identified and quantified, 34 of which showed significant fold changes in protein levels between N352S homozygotes and non-carriers. We determined N-glycosylation profiles from 370 glycosylation sites in 151 glycoproteins and identified ten proteins most significantly associated with decreased galactosylation and sialyation in B4GALT1 N352S homozygotes. These results further support that B4GALT1 N352S alters the glycosylation profiles of a variety of critical target proteins, thus governing the functions of these proteins in multiple pathways, such as those involved in lipid metabolism, coagulation, and the immune response.


Subject(s)
Galactosyltransferases , Proteomics , Humans , Amish/genetics , Galactosyltransferases/genetics , Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Glycosylation , Tandem Mass Spectrometry
3.
Molecules ; 28(2)2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36677618

ABSTRACT

UDP-Gal: glycoprotein-N-acetylgalactosamine ß-1,3-galactosyltransferase (T-synthase, EC 2.4.1.122) catalyses the transfer of the monosaccharide galactose from UDP-Gal to GalNAc-Ser/Thr, synthesizing the core 1 mucin type O-glycan. Such glycans play important biological roles in a number of recognition processes. The crucial role of these glycans is acknowledged for mammals, but a lot remains unknown regarding invertebrate and especially mollusc O-glycosylation. Although core O-glycans have been found in snails, no core 1 ß-1,3-galactosyltransferase has been described so far. Here, the sequence of the enzyme was identified by a BlastP search of the NCBI Biomphalaria glabrata database using the human T-synthase sequence (NP_064541.1) as a template. The obtained gene codes for a 388 amino acids long transmembrane protein with two putative N-glycosylation sites. The coding sequence was synthesised and expressed in Sf9 cells. The expression product of the putative enzyme displayed core 1 ß-1,3-galactosyltransferase activity using pNP-α-GalNAc as the substrate. The enzyme showed some sequence homology (49.40% with Homo sapiens, 53.69% with Drosophila melanogaster and 49.14% with Caenorhabditis elegans) and similar biochemical parameters with previously characterized T-synthases from other phyla. In this study we present the identification, expression and characterisation of the UDP-Gal: glycoprotein-N-acetylgalactosamine ß-1,3-galactosyltransferase from the fresh-water snail Biomphalaria glabrata, which is the first cloned T-synthase from mollusc origin.


Subject(s)
Biomphalaria , Galactosyltransferases , Animals , Humans , Acetylgalactosamine , Amino Acid Sequence , Biomphalaria/enzymology , Biomphalaria/genetics , Caenorhabditis elegans , Drosophila melanogaster , Galactosyltransferases/genetics , Galactosyltransferases/chemistry , Mucins , Polysaccharides/chemistry , Uridine Diphosphate
4.
Biochem J ; 478(21): 3939-3955, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34693969

ABSTRACT

Galactinol synthase (GolS) catalyzes the key regulatory step in the biosynthesis of Raffinose Family Oligosaccharides (RFOs). Even though the physiological role and regulation of this enzyme has been well studied, little is known about active site amino acids and the structure-function relationship with substrates of this enzyme. In the present study, we investigate the active site amino acid and structure-function relationship for this enzyme. Using a combination of three-dimensional homology modeling, molecular docking along with a series of deletion, site-directed mutagenesis followed by in vitro biochemical and in vivo functional analysis; we have studied active site amino acids and their interaction with the substrate of chickpea and Arabidopsis GolS enzyme. Our study reveals that the GolS protein possesses GT8 family-specific several conserved motifs in which NAG motif plays a crucial role in substrate binding and catalytic activity of this enzyme. Deletion of entire NAG motif or deletion or the substitution (with alanine) of any residues of this motif results in complete loss of catalytic activity in in vitro condition. Furthermore, disruption of NAG motif of CaGolS1 enzyme disrupts it's in vivo cellular function in yeast as well as in planta. Together, our study offers a new insight into the active site amino acids and their substrate interaction for the catalytic activity of GolS enzyme. We demonstrate that NAG motif plays a vital role in substrate binding for the catalytic activity of galactinol synthase that affects overall RFO synthesis.


Subject(s)
Arabidopsis Proteins , Arabidopsis/metabolism , Galactosyltransferases , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Catalytic Domain , Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Protein Conformation , Protein Domains
5.
J Biol Chem ; 297(1): 100843, 2021 07.
Article in English | MEDLINE | ID: mdl-34058199

ABSTRACT

Peters Plus Syndrome (PTRPLS OMIM #261540) is a severe congenital disorder of glycosylation where patients have multiple structural anomalies, including Peters anomaly of the eye (anterior segment dysgenesis), disproportionate short stature, brachydactyly, dysmorphic facial features, developmental delay, and variable additional abnormalities. PTRPLS patients and some Peters Plus-like (PTRPLS-like) patients (who only have a subset of PTRPLS phenotypes) have mutations in the gene encoding ß1,3-glucosyltransferase (B3GLCT). B3GLCT catalyzes the transfer of glucose to O-linked fucose on thrombospondin type-1 repeats. Most B3GLCT substrate proteins belong to the ADAMTS superfamily and play critical roles in extracellular matrix. We sought to determine whether the PTRPLS or PTRPLS-like mutations abrogated B3GLCT activity. B3GLCT has two putative active sites, one in the N-terminal region and the other in the C-terminal glycosyltransferase domain. Using sequence analysis and in vitro activity assays, we demonstrated that the C-terminal domain catalyzes transfer of glucose to O-linked fucose. We also generated a homology model of B3GLCT and identified D421 as the catalytic base. PTRPLS and PTRPLS-like mutations were individually introduced into B3GLCT, and the mutated enzymes were evaluated using in vitro enzyme assays and cell-based functional assays. Our results demonstrated that PTRPLS mutations caused loss of B3GLCT enzymatic activity and/or significantly reduced protein stability. In contrast, B3GLCT with PTRPLS-like mutations retained enzymatic activity, although some showed a minor destabilizing effect. Overall, our data supports the hypothesis that loss of glucose from B3GLCT substrate proteins is responsible for the defects observed in PTRPLS patients, but not for those observed in PTRPLS-like patients.


Subject(s)
Cleft Lip/enzymology , Cleft Lip/genetics , Cornea/abnormalities , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Growth Disorders/enzymology , Growth Disorders/genetics , Limb Deformities, Congenital/enzymology , Limb Deformities, Congenital/genetics , Mutation/genetics , ADAMTS Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Biocatalysis , Cornea/enzymology , Enzyme Stability , Fucose/metabolism , Galactosyltransferases/chemistry , Glucose/metabolism , Glucosyltransferases/chemistry , HEK293 Cells , Humans , Kinetics , Models, Molecular , Protein Domains , Repetitive Sequences, Amino Acid , Structural Homology, Protein
6.
Org Lett ; 23(5): 1738-1741, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33576634

ABSTRACT

Proteoglycans have important biological activities. To improve the overall synthetic efficiency, a new chemoenzymatic route has been established for the proteoglycan linkage region bearing a galactose-xylose disaccharide. The xylosylated glycopeptides were synthesized via solid phase synthesis, which was followed by the addition of the galactose unit by the galactosyl transferase ß4GalT7. This work leads to a better understanding of the acceptor preference of ß4GalT7 and opens the door for expeditious synthesis of the proteoglycan linkage region.


Subject(s)
Galactose/chemistry , Galactosyltransferases/metabolism , Glycopeptides/chemical synthesis , Proteoglycans/chemistry , Xylose/chemistry , Disaccharides/chemistry , Galactosyltransferases/chemistry , Glycopeptides/chemistry , Molecular Structure
7.
J Biol Chem ; 296: 100299, 2021.
Article in English | MEDLINE | ID: mdl-33460651

ABSTRACT

The human Gb3/CD77 synthase, encoded by the A4GALT gene, is an unusually promiscuous glycosyltransferase. It synthesizes the Galα1→4Gal linkage on two different glycosphingolipids (GSLs), producing globotriaosylceramide (Gb3, CD77, Pk) and the P1 antigen. Gb3 is the major receptor for Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli. A single amino acid substitution (p.Q211E) ramps up the enzyme's promiscuity, rendering it able to attach Gal both to another Gal residue and to GalNAc, giving rise to NOR1 and NOR2 GSLs. Human Gb3/CD77 synthase was long believed to transfer Gal only to GSL acceptors, therefore its GSL products were, by default, considered the only human Stx receptors. Here, using soluble, recombinant human Gb3/CD77 synthase and p.Q211E mutein, we demonstrate that both enzymes can synthesize the P1 glycotope (terminal Galα1→4Galß1→4GlcNAc-R) on a complex type N-glycan and a synthetic N-glycoprotein (saposin D). Moreover, by transfection of CHO-Lec2 cells with vectors encoding human Gb3/CD77 synthase and its p.Q211E mutein, we demonstrate that both enzymes produce P1 glycotopes on N-glycoproteins, with the mutein exhibiting elevated activity. These P1-terminated N-glycoproteins are recognized by Stx1 but not Stx2 B subunits. Finally, cytotoxicity assays show that Stx1 can use P1 N-glycoproteins produced in CHO-Lec2 cells as functional receptors. We conclude that Stx1 can recognize and use P1 N-glycoproteins in addition to its canonical GSL receptors to enter and kill the cells, while Stx2 can use GSLs only. Collectively, these results may have important implications for our understanding of the Shiga toxin pathology.


Subject(s)
Galactosyltransferases/chemistry , Globosides/chemistry , Shiga Toxin 1/chemistry , Trihexosylceramides/chemistry , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Animals , Binding Sites , CHO Cells , Carbohydrate Sequence , Cricetulus , Enterohemorrhagic Escherichia coli/chemistry , Enterohemorrhagic Escherichia coli/pathogenicity , Galactose/chemistry , Galactose/metabolism , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Gene Expression , Globosides/biosynthesis , Globosides/metabolism , Glucose/chemistry , Glucose/metabolism , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Shiga Toxin 1/metabolism , Shiga Toxin 2/chemistry , Shiga Toxin 2/metabolism , Trihexosylceramides/biosynthesis
8.
FEBS Lett ; 595(1): 110-122, 2021 01.
Article in English | MEDLINE | ID: mdl-33098688

ABSTRACT

O-GlcNAcylation is a post-translational modification catalysed by O-GlcNAc transferase (OGT). Missense mutations in OGT have been associated with developmental disorders, OGT-linked congenital disorder of glycosylation (OGT-CDG), which are characterized by intellectual disability. OGT relies on the hexosamine biosynthetic pathway (HBP) for provision of its UDP-GlcNAc donor. We considered whether mutations in UDP-N-acetylhexosamine pyrophosphorylase (UAP1), which catalyses the final step in the HBP, would phenocopy OGT-CDG mutations. A de novo mutation in UAP1 (NM_001324114:c.G685A:p.A229T) was reported in a patient with intellectual disability. We show that this mutation is pathogenic and decreases the stability and activity of the UAP1 isoform AGX1 in vitro. X-ray crystallography reveals a structural shift proximal to the mutation, leading to a conformational change of the N-terminal domain. These data suggest that the UAP1A229T missense mutation could be a contributory factor to the patient phenotype.


Subject(s)
Developmental Disabilities/genetics , Galactosyltransferases/genetics , Hexosamines/biosynthesis , Mutation, Missense , Amino Acid Sequence , Animals , Crystallography, X-Ray , Enzyme Stability , Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Humans , Protein Processing, Post-Translational , Sequence Homology, Amino Acid
9.
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
10.
Sci Rep ; 10(1): 13514, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32782311

ABSTRACT

Chloroplast membranes have a high content of the uncharged galactolipids monogalactosyldiacylglycerol (MGDG) and digalactosyldiacylglycerol (DGDG). These galactolipids are essential for the biogenesis of plastids and functioning of the photosynthetic machinery. A monotopic glycosyltransferase, monogalactosyldiacylglycerol synthase synthesizes the bulk of MGDG. It is embedded in the outer leaflet of the inner envelope membrane of chloroplasts. The protein transfers a galactose residue from UDP-galactose to diacylglycerol (DAG); it needs anionic lipids such as phosphatidylglycerol (PG) to be active. The intricacy of the organization and the process of active complex assembly and synthesis have been investigated at the Coarse-Grained and All-Atom of computer simulation levels to cover large spatial and temporal scales. The following self-assembly process and catalytic events can be drawn; (1) in the membrane, in the absence of protein, there is a spontaneous formation of PG clusters to which DAG molecules associate, (2) a reorganization of the clusters occurs in the vicinity of the protein once inserted in the membrane, (3) an accompanying motion of the catalytic domain of the protein brings DAG in the proper position for the formation of the active complex MGD1/UDP-Gal/DAG/PG for which an atomistic model of interaction is proposed.


Subject(s)
Chloroplasts/metabolism , Galactolipids/metabolism , Galactosyltransferases/metabolism , Intracellular Membranes/metabolism , Models, Molecular , Galactosyltransferases/chemistry , Intracellular Membranes/enzymology , Protein Conformation
11.
Blood Transfus ; 18(6): 471-477, 2020 11.
Article in English | MEDLINE | ID: mdl-32281923

ABSTRACT

BACKGROUND: Mutations of the ABO gene may cause the dysfunction of ABO glycosyltransferase (GT) that can result in weak ABO phenotypes. Here, we identified two novel weak ABO subgroup alleles and explored their mechanisms that caused Ael and Bel phenotypes. MATERIALS AND METHODS: The ABO phenotyping and genotyping were performed by serological studies and direct DNA sequencing of the ABO gene. The role of the novel mutations were evaluated by a three-dimensional model, predicting protein structure changes, and in vitro expression assay. The total glycosyltransferase transfer capacity in supernatant of transfected cells was examined. RESULTS: We identified a mutation c. 955C>G (p. L319V) of A allele in an Ael subject and a mutation c. 272T>C (p. L91P) of B allele in a Bel subject. In silico analysis showed that the mutation p. L319V of the A allele and p. L91P of the B allele may change the local conformation of GT and impair the catalysis of H to A or B antigen conversion. In vitro expression study showed that mutation p. L319V impaired H to A antigen conversion, although it did not affect the expression of glycosyltransferase A. CONCLUSIONS: Two novel "el"-type ABO subgroup alleles were identified. Both of the two novel mutations can change the local conformation of GTs and reduce protein stability. GTA mutation p. L319V can impair the conversion from H to A antigen and causes the Ael phenotype.


Subject(s)
ABO Blood-Group System/genetics , Galactosyltransferases/genetics , Mutation, Missense , Point Mutation , ABO Blood-Group System/chemistry , Alleles , Amino Acid Substitution , Blood Grouping and Crossmatching , Computer Simulation , Galactosyltransferases/chemistry , Genetic Association Studies , HeLa Cells , Humans , Models, Molecular , Phenotype , Protein Conformation , Protein Stability , Recombinant Proteins/metabolism
12.
Enzyme Microb Technol ; 135: 109489, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32146932

ABSTRACT

The successful enzymatic synthesis of various ganglioside-related oligosaccharides requires many available glycan-processing enzymes. However, the number of available glycan-processing enzymes remains limited. In this study, the full-length CgtA43456 (ß-(1→4)-N-acetylgalactosaminyltransferase) and CgtB11168 (ß-(1→3)-galactosyltransferase) were successfully produced from Escherichia coli through the optimization of E. coli-preferable codon usage, selection of E. coli strain, and use of the molecular chaperone GroEL-GroES (GroEL/ES). The CgtA43456 enzyme was produced as a soluble form in E. coli C41(DE3) co-expressed with codon-optimized CgtA43456 and GroEL/ES. However, soluble CgtB11168 was well expressed in E. coli C41(DE3) with only the codon-optimized CgtB11168. Rather, when co-expressed with GroEL/ES, total production of CgtB11168 was reduced. Using immobilized-metal affinity chromatography, the CgtA43456 and CgtB11168 proteins were obtained with approximately 75-78 % purity. The purified CgtA43456 showed a specific activity of 21 mU/mg using UDP-N-acetylgalactosamine and GM3 trisaccharide as donor and acceptor, respectively. The purified CgtB11168 catalyzed the transfer of galactose from UDP-Gal to GM2 tetrasaccharide with a specific activity of 16 mU/mg. We propose that they could be used as catalysts for enzymatic synthesis of GM1 ganglioside-related oligosaccharides.


Subject(s)
Bacterial Proteins/genetics , Campylobacter jejuni/enzymology , Galactosyltransferases/genetics , Galactosyltransferases/isolation & purification , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Campylobacter jejuni/chemistry , Campylobacter jejuni/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Gene Expression , N-Acetylgalactosaminyltransferases/chemistry , N-Acetylgalactosaminyltransferases/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Substrate Specificity
13.
Glycobiology ; 30(6): 396-406, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32100029

ABSTRACT

Mono- and digalactosyldiacylglycerol are essential galactolipids for the biogenesis of plastids and functioning of the photosynthetic machinery. In Arabidopsis, the first step of galactolipid synthesis is catalyzed by monogalactosyldiacylglycerol synthase 1 (MGD1), a monotopic protein located in the inner envelope membrane of chloroplasts, which transfers a galactose residue from UDP-galactose to diacylglycerol (DAG). MGD1 needs anionic lipids such as phosphatidylglycerol (PG) to be active, but the mechanism by which PG activates MGD1 is still unknown. Recent studies shed light on the catalytic mechanism of MGD1 and on the possible PG binding site. Particularly, Pro189 was identified as a potential residue implicated in PG binding and His155 as the putative catalytic residue. In the present study, using a multifaceted approach (Langmuir membrane models, atomic force microscopy, molecular dynamics; MD), we investigated the membrane binding properties of native MGD1 and mutants (P189A and H115A). We demonstrated that both residues are involved in PG binding, thus suggesting the existence of a PG-His catalytic dyad that should facilitate deprotonation of the nucleophile hydroxyl group of DAG acceptor. Interestingly, MD simulations showed that MGD1 induces a reorganization of lipids by attracting DAG molecules to create an optimal platform for binding.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Galactosyltransferases/metabolism , Phosphatidylglycerols/metabolism , Adsorption , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Lipids/chemistry , Mutation
14.
Biochemistry ; 59(8): 999-1009, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32067450

ABSTRACT

Galactolipids are characteristic lipids of the photosynthetic membranes. They are highly enriched in the chloroplast and are present in photosystem structures. There are two major types of galactolipids, i.e., monogalactosyldiacylglycerol and digalactosyldiacylglycerol (DGDG) in chloroplastic membranes, which amount to ∼50 and ∼20 mol % of the total chloroplast lipids, respectively. Under phosphate-limiting conditions, the amount of DGDG increases dramatically for rescuing phosphate from phospholipids. In Arabidopsis thaliana, the gene digalactosyldiacylglycerol synthase 2 (DGD2) encodes a membrane-associated glycosyltransferase. The gene expression is highly responsive to phosphate starvation and is significantly upregulated in this case. To understand the molecular mechanism of DGD2, we established a protocol for DGD2 expression and purification in an Escherichia coli-based system. The work involved optimization of the expression condition and the purification protocol and a careful selection of buffer additives. It was found that a removal of around 70 C-terminal residues was necessary to produce a homogeneous monomeric protein sample with high purity, which was highly active. The purified sample was characterized by an activity assay for enzyme kinetics in which a range of membrane mimetics with different lipid compositions were used. The results demonstrate that DGD2 activity is stimulated by the presence of negatively charged lipids, which highlight the importance of the membrane environment in modulating the enzyme's activity. The study also paves way for future biophysical and structural studies of the enzyme.


Subject(s)
Chloroplast Proteins/chemistry , Galactolipids/chemical synthesis , Membrane Proteins/chemistry , Amino Acid Sequence , Arabidopsis/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Arabidopsis Proteins/isolation & purification , Chloroplast Proteins/genetics , Chloroplast Proteins/isolation & purification , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Galactosyltransferases/isolation & purification , Kinetics , Lipid Bilayers/chemistry , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Sequence Alignment , Sequence Deletion , Unilamellar Liposomes/chemistry
15.
J Biol Chem ; 294(39): 14383-14393, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31395657

ABSTRACT

ß-1,4-Galactosyltransferase 1 (B4GALT1) and ST6 ß-galactoside α-2,6-sialyltransferase 1 (ST6GAL1) catalyze the successive addition of terminal ß-1,4-linked galactose and α-2,6-linked sialic acid to N-glycans. Their exclusive interaction in the Golgi compartment is a prerequisite for their full catalytic activity, whereas a lack of this interaction is associated with cancers and hypoxia. To date, no structural information exists that shows how glycosyltransferases functionally assemble with each other. Using molecular docking simulations to predict interaction surfaces, along with mutagenesis screens and high-throughput FRET analyses in live cells to validate these predictions, we show here that B4GALT1 and ST6GAL1 interact via highly charged noncatalytic surfaces, leaving the active sites exposed and accessible for donor and acceptor substrate binding. Moreover, we found that the assembly of ST6GAL1 homomers in the endoplasmic reticulum before ST6GAL1 activation in the Golgi utilizes the same noncatalytic surface, whereas B4GALT1 uses its active-site surface for assembly, which silences its catalytic activity. Last, we show that the homomeric and heteromeric B4GALT1/ST6GAL1 complexes can assemble laterally in the Golgi membranes without forming cross-cisternal contacts between enzyme molecules residing in the opposite membranes of each Golgi cisterna. Our results provide detailed mechanistic insights into the regulation of glycosyltransferase interactions, the transitions between B4GALT1 and ST6GAL1 homo- and heteromers in the Golgi, and cooperative B4GALT1/ST6GAL1 function in N-glycan synthesis.


Subject(s)
Antigens, CD/chemistry , Galactosyltransferases/chemistry , Molecular Docking Simulation , Protein Multimerization , Sialyltransferases/chemistry , Animals , Antigens, CD/metabolism , Binding Sites , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Fluorescence Resonance Energy Transfer , Galactosyltransferases/metabolism , Golgi Apparatus/metabolism , Sialyltransferases/metabolism , Static Electricity
16.
Org Biomol Chem ; 17(24): 5920-5924, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31165848

ABSTRACT

Utilising a fast and sensitive screening method based on imidazolium-tagged probes, we report unprecedented reversible activity of bacterial ß1,4-galactosyltransferases to catalyse the transgalactosylation from lactose to N-acetylglucosamine to form N-acetyllactosamine in the presence of UDP. The process is demonstrated by the preparative scale synthesis of pNP-ß-LacNAc from lactose using ß1,4-galactosyltransferase NmLgtB-B as the only biocatalyst.


Subject(s)
Amino Sugars/biosynthesis , Galactosyltransferases/metabolism , Lactose/metabolism , Amino Sugars/chemistry , Biocatalysis , Galactosyltransferases/chemistry , Lactose/chemistry , Molecular Structure , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
18.
Redox Biol ; 24: 101182, 2019 06.
Article in English | MEDLINE | ID: mdl-30959459

ABSTRACT

Glycosylation, a common modification of cellular proteins and lipids, is often altered in diseases and pathophysiological states such as hypoxia, yet the underlying molecular causes remain poorly understood. By utilizing lectin microarray glycan profiling, Golgi pH and redox screens, we show here that hypoxia inhibits terminal sialylation of N- and O-linked glycans in a HIF- independent manner by lowering Golgi oxidative potential. This redox state change was accompanied by loss of two surface-exposed disulfide bonds in the catalytic domain of the α-2,6-sialyltransferase (ST6Gal-I) and its ability to functionally interact with B4GalT-I, an enzyme adding the preceding galactose to complex N-glycans. Mutagenesis of selected cysteine residues in ST6Gal-I mimicked these effects, and also rendered the enzyme inactive. Cells expressing the inactive mutant, but not those expressing the wild type ST6Gal-I, were able to proliferate and migrate normally, supporting the view that inactivation of the ST6Gal-I help cells to adapt to hypoxic environment. Structure comparisons revealed similar disulfide bonds also in ST3Gal-I, suggesting that this O-glycan and glycolipid modifying sialyltransferase is also sensitive to hypoxia and thereby contribute to attenuated sialylation of O-linked glycans in hypoxic cells. Collectively, these findings unveil a previously unknown redox switch in the Golgi apparatus that is responsible for the catalytic activation and cooperative functioning of ST6Gal-I with B4GalT-I.


Subject(s)
Galactosyltransferases/metabolism , Golgi Apparatus/metabolism , Oxidation-Reduction , Sialyltransferases/metabolism , Animals , Catalysis , Cell Line , Cell Movement , Cell Proliferation , Disulfides/metabolism , Galactosyltransferases/chemistry , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Models, Molecular , Molecular Conformation , Polysaccharides/metabolism , Sialyltransferases/chemistry , beta-D-Galactoside alpha 2-6-Sialyltransferase
19.
Sci Rep ; 8(1): 17566, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30514885

ABSTRACT

Galactofuranosyltransferases are poorly described enzymes despite their crucial role in the virulence and the pathogenicity of numerous microorganisms. These enzymes are considered as potential targets for therapeutic action. In addition to the only well-characterised prokaryotic GlfT2 from Mycobacterium tuberculosis, four putative genes in Leishmania major were previously described as potential galactofuranosyltransferases. In this study, we have cloned, over-expressed, purified and fully determined the kinetic parameters of these four eukaryotic enzymes, thus demonstrating their unique potency in catalysing the transfer of the galactofuranosyl moiety into acceptors. Their individual promiscuity revealed to be different, as some of them could efficiently use NDP-pyranoses as donor substrates in addition to the natural UDP-galactofuranose. Such results pave the way for the development of chemoenzymatic synthesis of furanosyl-containing glycoconjugates as well as the design of improved drugs against leishmaniasis.


Subject(s)
Galactose/analogs & derivatives , Galactosyltransferases/biosynthesis , Galactosyltransferases/chemistry , Leishmania major/enzymology , Protozoan Proteins/biosynthesis , Protozoan Proteins/chemistry , Uridine Diphosphate/analogs & derivatives , Biocatalysis , Escherichia coli/genetics , Galactose/metabolism , Kinetics , Substrate Specificity , Uridine Diphosphate/metabolism
20.
PLoS One ; 13(10): e0205571, 2018.
Article in English | MEDLINE | ID: mdl-30352055

ABSTRACT

Most glycosyltransferases, including B4GalT1 (EC 2.4.1.38), are known to assemble into enzyme homomers and functionally relevant heteromers in vivo. However, it remains unclear why and how these enzymes interact at the molecular/atomic level. Here, we solved the crystal structure of the wild-type human B4GalT1 homodimer. We also show that B4GalT1 exists in a dynamic equilibrium between monomer and dimer, since a purified monomer reappears as a mixture of both and as we obtained crystal forms of the monomer and dimer assemblies in the same crystallization conditions. These two crystal forms revealed the unliganded B4GalT1 in both the open and the closed conformation of the Trp loop and the lid regions, responsible for donor and acceptor substrate binding, respectively. The present structures also show the lid region in full in an open conformation, as well as a new conformation for the GlcNAc acceptor loop (residues 272-288). The physiological relevance of the homodimer in the crystal was validated by targeted mutagenesis studies coupled with FRET assays. These showed that changing key catalytic amino acids impaired homomer formation in vivo. The wild-type human B4GalT1 structure also explains why the variant proteins used for crystallization in earlier studies failed to reveal the homodimers described in this study.


Subject(s)
Galactosyltransferases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Escherichia coli , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Galactosyltransferases/isolation & purification , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Multimerization
SELECTION OF CITATIONS
SEARCH DETAIL
...