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










Publication year range
1.
Chem Commun (Camb) ; 56(73): 10746-10749, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32789356

ABSTRACT

Here we propose a general strategy to label carbohydrates with N-methyl-anthranilic acid at the anomeric position. Through two examples, we demonstrate that the generated glycoprobes are suitable for fluorescence-based binding/competition assays. Our approach is expected to readily generate series of glycoprobes dedicated to screening assays for the discovery of drugs targeting carbohydrate-protein interactions.


Subject(s)
Fluorescent Dyes/chemistry , Glycosides/chemistry , ortho-Aminobenzoates/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Glycosides/chemical synthesis , Glycosides/metabolism , Maltose-Binding Proteins/metabolism , Protein Binding , Spectrometry, Fluorescence , ortho-Aminobenzoates/chemical synthesis , ortho-Aminobenzoates/metabolism
2.
Org Biomol Chem ; 18(25): 4831-4842, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32608461

ABSTRACT

Proteoglycans (PGs) are complex macromolecules that are composed of glycosaminoglycan (GAG) chains covalently attached to a core protein through a tetrasaccharide linker. Biosynthesis of PGs is complex and involves a large number of glycosyltranferases. We report herein for the first time the synthesis of a collection of various sulfoforms of the disaccharide GlcA-1,3-ß-d-Gal and trisaccharides GlcNAc-1,4-α-d-GlcA-1,3-ß-d-Gal and GalNAc-1,4-ß-d-GlcA-1,3-ß-d-Gal using a regioselective glycosylation. Preliminary results on the impact of sulfation of these disaccharides upon recombinant chondroitin sulfate N-acetylgalactosaminyltransferase-1 (CSGalNAcT-1) involved in chondroitin sulfate chain initiation is also reported.


Subject(s)
Oligosaccharides/chemical synthesis , Proteoglycans/chemistry , Glycosylation , Molecular Conformation , Oligosaccharides/chemistry , Stereoisomerism
3.
Semin Cancer Biol ; 62: 68-85, 2020 05.
Article in English | MEDLINE | ID: mdl-31711992

ABSTRACT

Heparansulfate (HS) modifications are master regulators of the cross-talk between cell and matrix and modulate the biological activity of an array of HS binding proteins, including growth factors and chemokines, morphogens and immunity cell receptors. This review will highlight the importance of HS maturation mediated by N-deactetylase/sulfotransferases, 2O- and 6O-sulfotransferases in cancer biology, and will focus on the 3O-sulfotransferases and on the terminal, rare 3O-sulfation, and their important but still enigmatic impact in cancer progression. The review will also discuss the molecular mechanisms of action of these HS modifications with regards to ligand interactions and signaling in the cancer process and their clinical significance.


Subject(s)
Heparitin Sulfate/metabolism , Neoplasms/metabolism , Sulfotransferases/metabolism , Animals , Biosynthetic Pathways , Carrier Proteins , Cell Proliferation , Disease Susceptibility , Fibroblast Growth Factors/metabolism , Humans , Ligands , Neoplasms/etiology , Protein Binding , Transforming Growth Factor beta/metabolism
4.
Hum Mol Genet ; 27(20): 3475-3487, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29931299

ABSTRACT

Proteoglycans are among the most abundant and structurally complex biomacromolecules and play critical roles in connective tissues. They are composed of a core protein onto which glycosaminoglycan (GAG) side chains are attached via a linker region. Biallelic mutations in B3GALT6, encoding one of the linker region glycosyltransferases, are known to cause either spondyloepimetaphyseal dysplasia (SEMD) or a severe pleiotropic form of Ehlers-Danlos syndromes (EDS). This study provides clinical, molecular and biochemical data on 12 patients with biallelic B3GALT6 mutations. Notably, all patients have features of both EDS and SEMD. In addition, some patients have severe and potential life-threatening complications such as aortic dilatation and aneurysm, cervical spine instability and respiratory insufficiency. Whole-exome sequencing, next generation panel sequencing and direct sequencing identified biallelic B3GALT6 mutations in all patients. We show that these mutations reduce the amount of ß3GalT6 protein and lead to a complete loss of galactosyltransferase activity. In turn, this leads to deficient GAG synthesis, and ultrastructural abnormalities in collagen fibril organization. In conclusion, this study redefines the phenotype associated with B3GALT6 mutations on the basis of clinical, molecular and biochemical data in 12 patients, and provides an in-depth assessment of ß3GalT6 activity and GAG synthesis to better understand this rare condition.


Subject(s)
Ehlers-Danlos Syndrome/genetics , Exome Sequencing , Galactosyltransferases/genetics , Mutation , Phenotype , Adult , Child , Child, Preschool , Ehlers-Danlos Syndrome/enzymology , Ehlers-Danlos Syndrome/pathology , Enzyme Assays , Female , Galactosyltransferases/metabolism , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Infant , Male
5.
Org Biomol Chem ; 15(45): 9653-9669, 2017 Nov 22.
Article in English | MEDLINE | ID: mdl-29116283

ABSTRACT

Proteoglycans (PGs) are complex macromolecules that are composed of glycosaminoglycan (GAG) chains covalently attached to a core protein through a tetrasaccharide linker. The biosynthesis of PGs is complex and involves a large number of glycosyltranferases. Here we present a structure-activity study of human ß4GalT7, which transfers the first Gal residue onto a xyloside moiety of the linkage region. An efficient and regiocontrolled synthesis of a library of modified analogs of 4-methylumbelliferyl xyloside (XylMU) is reported herein. Hydroxyl groups at the position C-2, C-3 or C-4 have been epimerized and/or replaced by a hydrogen or a fluorine, while the anomeric oxygen was replaced by either a sulfur or a sulfone. The effect of these compounds on human ß4GalT7 activity in vitro and on GAG biosynthesis in cellulo was then evaluated.


Subject(s)
Galactosyltransferases/metabolism , Glycosides/biosynthesis , Small Molecule Libraries/metabolism , Carbohydrate Conformation , Glycosides/chemistry , Humans , Small Molecule Libraries/chemistry , Structure-Activity Relationship
6.
Chem Biol Drug Des ; 89(3): 319-326, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27618481

ABSTRACT

Different mono-xylosides and their corresponding xylobiosides obtained by a chemo-enzymatic approach featuring various substituents attached to a triazole ring were probed as priming agents for glycosaminoglycan (GAG) biosynthesis in the xylosyltransferase-deficient pgsA-745 Chinese hamster ovary cell line. Xylosides containing a hydrophobic aglycone moiety were the most efficient priming agents. Mono-xylosides induced higher GAG biosynthesis in comparison with their corresponding xylobiosides. The influence of the degree of polymerization of the carbohydrate part on the priming activity was investigated through different experiments. We demonstrated that in case of mono-xylosides, the cellular uptake as well as the affinity and the catalytic efficiency of ß-1,4-galactosyltransferase 7 were higher than for xylobiosides. Altogether, these results indicate that hydrophobicity of the aglycone and degree of polymerization of glycone moiety were critical factors for an optimal priming activity for GAG biosynthesis.


Subject(s)
Glycosaminoglycans/biosynthesis , Glycosides/chemistry , Glycosides/metabolism , Animals , CHO Cells , Cricetulus , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Humans , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Structure-Activity Relationship , UDP Xylose-Protein Xylosyltransferase
7.
J Biol Chem ; 290(12): 7658-70, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25568325

ABSTRACT

Among glycosaminoglycan (GAG) biosynthetic enzymes, the human ß1,4-galactosyltransferase 7 (hß4GalT7) is characterized by its unique capacity to take over xyloside derivatives linked to a hydrophobic aglycone as substrates and/or inhibitors. This glycosyltransferase is thus a prime target for the development of regulators of GAG synthesis in therapeutics. Here, we report the structure-guided design of hß4GalT7 inhibitors. By combining molecular modeling, in vitro mutagenesis, and kinetic measurements, and in cellulo analysis of GAG anabolism and decorin glycosylation, we mapped the organization of the acceptor binding pocket, in complex with 4-methylumbelliferone-xylopyranoside as prototype substrate. We show that its organization is governed, on one side, by three tyrosine residues, Tyr(194), Tyr(196), and Tyr(199), which create a hydrophobic environment and provide stacking interactions with both xylopyranoside and aglycone rings. On the opposite side, a hydrogen-bond network is established between the charged amino acids Asp(228), Asp(229), and Arg(226), and the hydroxyl groups of xylose. We identified two key structural features, i.e. the strategic position of Tyr(194) forming stacking interactions with the aglycone, and the hydrogen bond between the His(195) nitrogen backbone and the carbonyl group of the coumarinyl molecule to develop a tight binder of hß4GalT7. This led to the synthesis of 4-deoxy-4-fluoroxylose linked to 4-methylumbelliferone that inhibited hß4GalT7 activity in vitro with a Ki 10 times lower than the Km value and efficiently impaired GAG synthesis in a cell assay. This study provides a valuable probe for the investigation of GAG biology and opens avenues toward the development of bioactive compounds to correct GAG synthesis disorders implicated in different types of malignancies.


Subject(s)
Enzyme Inhibitors/chemistry , Galactosyltransferases/metabolism , Xylosidases/antagonists & inhibitors , Catalytic Domain , Drug Design , Enzyme Inhibitors/pharmacology , Galactosyltransferases/chemistry , Humans , Kinetics , Models, Molecular , Molecular Probes
8.
Front Cell Neurosci ; 8: 349, 2014.
Article in English | MEDLINE | ID: mdl-25389387

ABSTRACT

UDP-glucuronosyltransferases (UGTs) form a multigenic family of membrane-bound enzymes expressed in various tissues, including brain. They catalyze the formation of ß-D-glucuronides from structurally unrelated substances (drugs, other xenobiotics, as well as endogenous compounds) by the linkage of glucuronic acid from the high energy donor, UDP-α-D-glucuronic acid. In brain, UGTs actively participate to the overall protection of the tissue against the intrusion of potentially harmful lipophilic substances that are metabolized as hydrophilic glucuronides. These metabolites are generally inactive, except for important pharmacologically glucuronides such as morphine-6-glucuronide. UGTs are mainly expressed in endothelial cells and astrocytes of the blood brain barrier (BBB). They are also associated to brain interfaces devoid of BBB, such as circumventricular organ, pineal gland, pituitary gland and neuro-olfactory tissues. Beside their key-role as a detoxication barrier, UGTs play a role in the steady-state of endogenous compounds, like steroids or dopamine (DA) that participate to the function of the brain. UGT isoforms of family 1A, 2A, 2B and 3A are expressed in brain tissues to various levels and are known to present distinct but overlapping substrate specificity. The importance of these enzyme species with regard to the formation of toxic, pharmacologically or physiologically relevant glucuronides in the brain will be discussed.

9.
Am J Hum Genet ; 92(6): 935-45, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23664118

ABSTRACT

Proteoglycans are important components of cell plasma membranes and extracellular matrices of connective tissues. They consist of glycosaminoglycan chains attached to a core protein via a tetrasaccharide linkage, whereby the addition of the third residue is catalyzed by galactosyltransferase II (ß3GalT6), encoded by B3GALT6. Homozygosity mapping and candidate gene sequence analysis in three independent families, presenting a severe autosomal-recessive connective tissue disorder characterized by skin fragility, delayed wound healing, joint hyperlaxity and contractures, muscle hypotonia, intellectual disability, and a spondyloepimetaphyseal dysplasia with bone fragility and severe kyphoscoliosis, identified biallelic B3GALT6 mutations, including homozygous missense mutations in family 1 (c.619G>C [p.Asp207His]) and family 3 (c.649G>A [p.Gly217Ser]) and compound heterozygous mutations in family 2 (c.323_344del [p.Ala108Glyfs(∗)163], c.619G>C [p.Asp207His]). The phenotype overlaps with several recessive Ehlers-Danlos variants and spondyloepimetaphyseal dysplasia with joint hyperlaxity. Affected individuals' fibroblasts exhibited a large decrease in ability to prime glycosaminoglycan synthesis together with impaired glycanation of the small chondroitin/dermatan sulfate proteoglycan decorin, confirming ß3GalT6 loss of function. Dermal electron microcopy disclosed abnormalities in collagen fibril organization, in line with the important regulatory role of decorin in this process. A strong reduction in heparan sulfate level was also observed, indicating that ß3GalT6 deficiency alters synthesis of both main types of glycosaminoglycans. In vitro wound healing assay revealed a significant delay in fibroblasts from two index individuals, pointing to a role for glycosaminoglycan defect in impaired wound repair in vivo. Our study emphasizes a crucial role for ß3GalT6 in multiple major developmental and pathophysiological processes.


Subject(s)
Abnormalities, Multiple/genetics , Ehlers-Danlos Syndrome/genetics , Galactosyltransferases/genetics , Glycosaminoglycans/biosynthesis , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/metabolism , Adult , Amino Acid Sequence , Base Sequence , Child , Consanguinity , Ehlers-Danlos Syndrome/diagnostic imaging , Ehlers-Danlos Syndrome/metabolism , Female , Genetic Association Studies , Genetic Pleiotropy , Humans , Infant , Male , Molecular Sequence Data , Mutation, Missense , Pedigree , Radiography , Sequence Analysis, DNA , Wound Healing/genetics
10.
Glycobiology ; 22(4): 561-71, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22156920

ABSTRACT

Glycosaminoglycan (GAG) assembly initiates through the formation of a linkage tetrasaccharide region serving as a primer for both chondroitin sulfate (CS) and heparan sulfate (HS) chain polymerization. A possible role for sulfation of the linkage structure and of the constitutive disaccharide unit of CS chains in the regulation of CS-GAG chain synthesis has been suggested. To investigate this, we determined whether sulfate substitution of galactose (Gal) residues of the linkage region or of N-acetylgalactosamine (GalNAc) of the disaccharide unit influences activity and specificity of chondroitin sulfate N-acetylgalactosaminyltransferase-1 (CSGalNAcT-1), a key glycosyltransferase of CS biosynthesis. We synthesized a series of sulfated and unsulfated analogs of the linkage oligosaccharide and of the constitutive unit of CS and tested these molecules as potential acceptor substrates for the recombinant human CSGalNAcT-1. We show here that sulfation at C4 or C6 of the Gal residues markedly influences CSGalNAcT-1 initiation activity and catalytic efficiency. Kinetic analysis indicates that CSGalNAcT-1 exhibited 3.6-, 1.6-, and 2.2-fold higher enzymatic efficiency due to lower K(m) values toward monosulfated trisaccharides substituted at C4 or C6 position of Gal1, and at C6 of Gal2, respectively, compared with the unsulfated oligosaccharide. This highlights the critical influence of Gal substitution on both CSGalNAcT-1 activity and specifity. No GalNAcT activity was detected toward sulfated and unsulfated analogs of the CS constitutive disaccharide (GlcA-ß1,3-GalNAc), indicating that CSGalNAcT-1 was involved in initiation but not in elongation of CS chains. Our results strongly suggest that sulfation of the linkage region acts as a regulatory signal in CS chain initiation.


Subject(s)
Chondroitin Sulfates/chemistry , N-Acetylgalactosaminyltransferases/chemistry , Acetylglucosamine/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Galactans/chemistry , Galactose/chemistry , Glycosylation , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Oligosaccharides/chemistry , Recombinant Proteins/chemistry , Substrate Specificity
11.
J Biol Chem ; 285(48): 37342-58, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20843813

ABSTRACT

Glycosaminoglycans (GAGs) play a central role in many pathophysiological events, and exogenous xyloside substrates of ß1,4-galactosyltransferase 7 (ß4GalT7), a major enzyme of GAG biosynthesis, have interesting biomedical applications. To predict functional peptide regions important for substrate binding and activity of human ß4GalT7, we conducted a phylogenetic analysis of the ß1,4-galactosyltransferase family and generated a molecular model using the x-ray structure of Drosophila ß4GalT7-UDP as template. Two evolutionary conserved motifs, (163)DVD(165) and (221)FWGWGREDDE(230), are central in the organization of the enzyme active site. This model was challenged by systematic engineering of point mutations, combined with in vitro and ex vivo functional assays. Investigation of the kinetic properties of purified recombinant wild-type ß4GalT7 and selected mutants identified Trp(224) as a key residue governing both donor and acceptor substrate binding. Our results also suggested the involvement of the canonical carboxylate residue Asp(228) acting as general base in the reaction catalyzed by human ß4GalT7. Importantly, ex vivo functional tests demonstrated that regulation of GAG synthesis is highly responsive to modification of these key active site amino acids. Interestingly, engineering mutants at position 224 allowed us to modify the affinity and to modulate the specificity of human ß4GalT7 toward UDP-sugars and xyloside acceptors. Furthermore, the W224H mutant was able to sustain decorin GAG chain substitution but not GAG synthesis from exogenously added xyloside. Altogether, this study provides novel insight into human ß4GalT7 active site functional domains, allowing manipulation of this enzyme critical for the regulation of GAG synthesis. A better understanding of the mechanism underlying GAG assembly paves the way toward GAG-based therapeutics.


Subject(s)
Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Glycosaminoglycans/biosynthesis , Amino Acid Motifs , Amino Acid Sequence , Animals , Catalytic Domain , Galactosyltransferases/genetics , Humans , Invertebrates/chemistry , Invertebrates/classification , Invertebrates/enzymology , Invertebrates/genetics , Models, Molecular , Molecular Sequence Data , Phylogeny , Sequence Alignment , Substrate Specificity , Vertebrates/classification , Vertebrates/genetics , Vertebrates/metabolism
12.
Biochem J ; 432(2): 303-11, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20809901

ABSTRACT

Three mutations of the B4GALT7 gene [encoding ß1,4-GalT7 (ß1,4-galactosyltransferase 7)], corresponding to A186D, L206P and R270C, have been identified in patients with the progeroid form of the Ehlers-Danlos syndrome and are described as being associated with the reduction or loss of ß1,4-GalT7 activity. However, the molecular basis of the reduction or loss of activity remained to be determined. In the present study, wild-type, A186D, L206P and R270C ß1,4-GalT7 were expressed in CHO618 cells as membrane proteins and in Escherichia coli as soluble proteins fused to MBP (maltose-binding protein). The ability of the expressed proteins to transfer galactose from donor to acceptor substrates was systematically characterized by kinetic analysis. The physicochemical properties of soluble proteins were explored by isothermal titration calorimetry, which is a method of choice when determining the thermodynamic parameters of the binding of substrates. Together, the results showed that: (i) the L206P mutation abolished the activity when L206P ß1,4GalT7 was either inserted in the membrane or expressed as a soluble MBP-full-length fusion protein; (ii) the A186D mutation weakly impaired the binding of the donor substrate; and (iii) the R270C mutation strongly impaired the binding of the acceptor substrate. Moreover, the ex vivo consequences of the mutations were investigated by evaluating the priming efficiency of xylosides on GAG (glycosaminoglycan) chain initiation. The results demonstrate a quantitative effect on GAG biosynthesis, depending on the mutation; GAG biosynthesis was fully inhibited by the L206P mutation and decreased by the R270C mutation, whereas the A186D mutation did not affect GAG biosynthesis severely.


Subject(s)
Ehlers-Danlos Syndrome/enzymology , Ehlers-Danlos Syndrome/genetics , Galactosyltransferases/genetics , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Humans , Kinetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Thermodynamics
13.
Biochem J ; 418(3): 605-14, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19032152

ABSTRACT

Human beta1,4-GalT (galactosyltransferase)7 is involved in the biosynthesis of the tetrasaccharide linker protein region (GlcAbeta1-->3Galbeta1-->3Galbeta1-->4Xylbeta1) (where GlcA is glucuronic acid and Xyl is xylose) of proteoglycans, by catalysing the transfer of Gal (galactose) from the uridine 5'-diphosphogalactose to a Xyl residue. This reaction is rate-limiting in glycosaminoglycan biosynthesis. In the present study, we established a large-scale production system of beta1,4-GalT7 fused with the maltose-binding protein to study substrate recognition. Calorimetric binding studies showed that the binding of the donor substrate UDP-Gal largely promoted binding of the acceptor substrate. To identify the structural basis governing substrate recognition, we used a fragment-based approach involving the artificial breakdown of the donor substrate into smaller fragments and characterization of their respective binding to the enzyme by isothermal titration calorimetry. The beta-phosphate, and to a lesser extent the alpha-phosphate, largely contributed to the binding energy. However, the uridine moiety was found to be essential for the optimal positioning of the donor substrate within the binding site. Unexpectedly, the contribution of the Gal moiety in substrate recognition was found to be negligible. Indeed, UDP-Gal, but also various UDP-sugars, could bind to beta1,4-GalT7. Surprisingly, in contrast with other GalTs, soluble beta1,4-GalT7 was able to transfer Glc (glucose), Xyl and, to a lesser extent GlcA and GlcNAc (N-acetyl glucosamine), to acceptor sugars, whereas UDP-Man (mannose) and UDP-GalNAc (N-acetyl galactosamine) were not substrates.


Subject(s)
Galactosyltransferases/metabolism , Carrier Proteins/genetics , Escherichia coli/enzymology , Galactosyltransferases/antagonists & inhibitors , Galactosyltransferases/chemistry , Galactosyltransferases/isolation & purification , HeLa Cells , Humans , Kinetics , Maltose-Binding Proteins , Nuclear Magnetic Resonance, Biomolecular , Recombinant Fusion Proteins/isolation & purification , Substrate Specificity , Thermodynamics , Uridine Diphosphate Sugars/metabolism
14.
Glycobiology ; 17(8): 857-67, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17567734

ABSTRACT

The human beta1,3-glucuronosyltransferases galactose-beta1,3-glucuronosyltransferase I (GlcAT-I) and galactose-beta1,3-glucuronosyltransferase P (GlcAT-P) are key enzymes involved in proteoglycan and HNK-1 carbohydrate epitope synthesis, respectively. Analysis of their acceptor specificity revealed that GlcAT-I was selective toward Galbeta1,3Gal (referred to as Gal2-Gal1), whereas GlcAT-P presented a broader profile. To understand the molecular basis of acceptor substrate recognition, we constructed mutants and chimeric enzymes based on multiple sequence alignment and structural information. The drastic effect of mutations of Glu227, Arg247, Asp252, and Glu281 on GlcAT-I activity indicated a key role for the hydrogen bond network formed by these four conserved residues in dictating Gal2 binding. Investigation of GlcAT-I determinants governing Gal1 recognition showed that Trp243 could not be replaced by its counterpart Phe in GlcAT-P. This result combined with molecular modeling provided evidence for the importance of stacking interactions with Trp at position 243 in the selectivity of GlcAT-I toward Galbeta1,3Gal. Mutation of Gln318 predicted to be hydrogen-bonded to 6-hydroxyl of Gal1 had little effect on GlcAT-I activity, reinforcing the role of Trp243 in Gal1 binding. Substitution of Phe245 in GlcAT-P by Ala selectively abolished Galbeta1,3Gal activity, also highlighting the importance of an aromatic residue at this position in defining the specificity of GlcAT-P. Finally, substituting Phe245, Val320, or Asn321 in GlcAT-P predicted to interact with N-acetylglucosamine (GlcNAc), by their counterpart in GlcAT-I, moderately affected the activity toward the reference substrate of GlcAT-P, N-acetyllactosamine, indicating that its active site tolerates amino acid substitutions, an observation that parallels its promiscuous substrate profile. Taken together, the data clearly define key residues governing the specificity of beta1,3-glucuronosyltransferases.


Subject(s)
CD57 Antigens/biosynthesis , Epitopes/biosynthesis , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/metabolism , Glycosaminoglycans/biosynthesis , Amino Acid Sequence , Binding Sites , CD57 Antigens/chemistry , CD57 Antigens/immunology , Glycosaminoglycans/chemistry , Glycosaminoglycans/immunology , Humans , Models, Molecular , Molecular Sequence Data , Substrate Specificity
15.
Protein Sci ; 15(7): 1667-78, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16815917

ABSTRACT

The beta1,3-glucuronosyltransferases are responsible for the completion of the protein-glycosaminoglycan linkage region of proteoglycans and of the HNK1 epitope of glycoproteins and glycolipids by transferring glucuronic acid from UDP-alpha-D-glucuronic acid (UDP-GlcA) onto a terminal galactose residue. Here, we develop phylogenetic and mutational approaches to identify critical residues involved in UDP-GlcA binding and enzyme activity of the human beta1,3-glucuronosyltransferase I (GlcAT-I), which plays a key role in glycosaminoglycan biosynthesis. Phylogeny analysis identified 119 related beta1,3-glucuronosyltransferase sequences in vertebrates, invertebrates, and plants that contain eight conserved peptide motifs with 15 highly conserved amino acids. Sequence homology and structural information suggest that Y84, D113, R156, R161, and R310 residues belong to the UDP-GlcA binding site. The importance of these residues is assessed by site-directed mutagenesis, UDP affinity and kinetic analyses. Our data show that uridine binding is primarily governed by stacking interactions with the phenyl group of Y84 and also involves interactions with aspartate 113. Furthermore, we found that R156 is critical for enzyme activity but not for UDP binding, whereas R310 appears less important with regard to both activity and UDP interactions. These results clearly discriminate the function of these two active site residues that were predicted to interact with the pyrophosphate group of UDP-GlcA. Finally, mutation of R161 severely compromises GlcAT-I activity, emphasizing the major contribution of this invariant residue. Altogether, this phylogenetic approach sustained by biochemical analyses affords new insight into the organization of the beta1,3-glucuronosyltransferase family and distinguishes the respective importance of conserved residues in UDP-GlcA binding and activity of GlcAT-I.


Subject(s)
Amino Acids/metabolism , Glucuronosyltransferase/metabolism , Mutation , Phylogeny , Uridine Diphosphate Glucuronic Acid/metabolism , Animals , Binding Sites/genetics , Conserved Sequence , Glucuronosyltransferase/genetics , Humans , Mutagenesis, Site-Directed
16.
Protein Expr Purif ; 47(1): 137-43, 2006 May.
Article in English | MEDLINE | ID: mdl-16300963

ABSTRACT

The galactose-beta1,3-glucuronosyltransferase I (GlcAT-I) catalyzes the transfer of glucuronic acid from UDP-alpha-D-glucuronic acid onto the terminal galactose of the trisaccharide glycosaminoglycan-protein linker region of proteoglycans. This enzyme plays a key role in the process of proteoglycan assembly since the completion of the linkage region is essential for the conversion of a core protein into a functional proteoglycan. To investigate the enzymatic properties of human GlcAT-I, we established an expression system for producing a soluble form of enzyme in the methylotrophic yeast Pichia pastoris and developed a three-step purification procedure using a combination of anion exchange, cation exchange and heparin chromatographies. This procedure yielded 1.6 mg homogeneous enzyme from 200 ml yeast cell culture, with a specific activity value of 1.5 micromol/min/mg protein. Analysis of the specificity of GlcAT-I towards Galbeta1-3Gal and Galbeta1-4GlcNAc derivatives known as substrates of the beta1,3-glucuronosyltransferases, showed that the enzyme exhibited a strict selectivity towards Galbeta1-3Gal structures. Thus, the large source of purified active enzyme allowed the determination of the kinetic parameters of GlcAT-I towards the donor substrate UDP-GlcA and the acceptor substrate digalactoside Galbeta1-3Gal.


Subject(s)
Glucuronosyltransferase/genetics , Glucuronosyltransferase/isolation & purification , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Cloning, Molecular , Galactosides/chemistry , Galactosides/metabolism , Glucuronosyltransferase/biosynthesis , Glucuronosyltransferase/chemistry , Humans , Kinetics , Pichia/enzymology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Solubility , Substrate Specificity , Uridine Diphosphate Glucuronic Acid/chemistry , Uridine Diphosphate Glucuronic Acid/metabolism
18.
J Biol Chem ; 280(2): 1417-25, 2005 Jan 14.
Article in English | MEDLINE | ID: mdl-15522873

ABSTRACT

We determined whether the two major structural modifications, i.e. phosphorylation and sulfation of the glycosaminoglycan-protein linkage region (GlcAbeta1-3Galbeta1-3Galbeta1-4Xylbeta1), govern the specificity of the glycosyltransferases responsible for the biosynthesis of the tetrasaccharide primer. We analyzed the influence of C-2 phosphorylation of Xyl residue on human beta1,4-galactosyltransferase 7 (GalT-I), which catalyzes the transfer of Gal onto Xyl, and we evaluated the consequences of C-4/C-6 sulfation of Galbeta1-3Gal (Gal2-Gal1) on the activity and specificity of beta1,3-glucuronosyltransferase I (GlcAT-I) responsible for the completion of the glycosaminoglycan primer sequence. For this purpose, a series of phosphorylated xylosides and sulfated C-4 and C-6 analogs of Galbeta1-3Gal was synthesized and tested as potential substrates for the recombinant enzymes. Our results revealed that the phosphorylation of Xyl on the C-2 position prevents GalT-I activity, suggesting that this modification may occur once Gal is attached to the Xyl residue of the nascent oligosaccharide linkage. On the other hand, we showed that sulfation on C-6 position of Gal1 of the Galbeta1-3Gal analog markedly enhanced GlcAT-I catalytic efficiency and we demonstrated the importance of Trp243 and Lys317 residues of Gal1 binding site for enzyme activity. In contrast, we found that GlcAT-I was unable to use digalactosides as acceptor substrates when Gal1 was sulfated on C-4 position or when Gal2 was sulfated on both C-4 and C-6 positions. Altogether, we demonstrated that oligosaccharide modifications of the linkage region control the specificity of the glycosyltransferases, a process that may regulate maturation and processing of glycosaminoglycan chains.


Subject(s)
Galactosyltransferases/metabolism , Glucuronosyltransferase/metabolism , Glycosaminoglycans/metabolism , Oligosaccharides/metabolism , Proteoglycans/biosynthesis , Sulfur/metabolism , Binding Sites , Carbohydrate Sequence , Chromatography, High Pressure Liquid , Galactosyltransferases/chemistry , Glucuronosyltransferase/chemistry , Glycosaminoglycans/chemistry , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Proteoglycans/chemistry , Substrate Specificity
19.
J Biol Chem ; 278(34): 32219-26, 2003 Aug 22.
Article in English | MEDLINE | ID: mdl-12794088

ABSTRACT

The human beta 1,3-glucuronosyltransferase I (GlcAT-I) is the key enzyme responsible for the completion of glycosaminoglycan-protein linkage tetrasaccharide of proteoglycans (GlcA beta 1,3Gal beta 1,3Gal beta 1,4Xyl beta 1-O-serine). We have investigated the role of aspartate residues Asp194-Asp195-Asp196 corresponding to the glycosyltransferase DXD signature motif, in GlcAT-I function by UDP binding experiments, kinetic analyses, and site-directed mutagenesis. We presented the first evidence that Mn2+ is not only essential for GlcAT-I activity but is also required for cosubstrate binding. In agreement, kinetic studies were consistent with a metal-activated enzyme model whereby activation probably occurs via binding of a Mn2+.UDP-GlcA complex to the enzyme. Mutational analysis showed that the Asp194-Asp195-Asp196 motif is a major element of the UDP/Mn2+ binding site. Furthermore, determination of the individual role of each aspartate showed that substitution of Asp195 as well as Asp196 to alanine strongly impaired GlcAT-I activity, whereas Asp194 replacement produced only a moderate alteration of the enzyme activity. These findings along with molecular modeling and three-dimensional structure comparison of the GlcAT-I catalytic center with that of the Bacillus subtilis glycosyltransferase SpsA provided evidence that the interactions of Asp195 with the ribose moiety of UDP and of Asp196 with the metal cation Mn2+ were crucial for GlcAT-I function. Altogether, these results indicated that, similarly to the SpsA enzyme, the nucleotide binding site of GlcAT-I contains a XDD motif rather than a DXD motif.


Subject(s)
Amino Acid Motifs , Glucuronosyltransferase/metabolism , Base Sequence , DNA Primers , Enzyme Activation , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/genetics , Humans , Kinetics , Manganese/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
20.
J Biol Chem ; 277(28): 25439-45, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-11986319

ABSTRACT

The human beta1,3-glucuronosyltransferase I (GlcAT-I) plays a key role in proteoglycan biosynthesis by catalyzing the transfer of glucuronic acid onto the trisaccharide-protein linkage structure Galbeta1,3Galbeta1,4Xylbeta-O-Ser, a prerequisite step for polymerization of glycosaminoglycan chains. In this study, we identified His(308) and Arg(277) residues as essential determinants for the donor substrate (UDP-glucuronic acid) selectivity of the human GlcAT-I. Analysis of the UDP-glucuronic acid-binding site by computational modeling in conjunction with site-directed mutagenesis indicated that both residues interact with glucuronic acid. Substitution of His(308) by arginine induced major changes in the donor substrate specificity of GlcAT-I. Interestingly, the H308R mutant was able to efficiently utilize nucleotide sugars UDP-glucose, UDP-mannose, and UDP-N-acetylglucosamine, which are not naturally accepted by the wild-type enzyme, as co-substrate in the transfer reaction. To gain insight into the role of Arg(277), site-directed mutagenesis in combination with chemical modification was carried out. Substitution of Arg(277) with alanine abrogated the activity of GlcAT-I. Furthermore, the arginine-directed reagent 2,3-butanedione irreversibly inhibited GlcAT-I, which was effectively protected against inactivation by UDP-glucuronic acid but not by UDP-glucose. It is noteworthy that the activity of the H308R mutant toward UDP-glucose was unaffected by the arginine-directed reagent. Our results are consistent with crucial interactions between the His(308) and Arg(277) residues and the glucuronic acid moiety that governs the specificity of GlcAT-I toward the nucleotide sugar donor substrate.


Subject(s)
Arginine/metabolism , Glucuronosyltransferase/metabolism , Histidine/metabolism , Uridine Diphosphate Glucuronic Acid/metabolism , Base Sequence , DNA Primers , Epoxy Compounds/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/genetics , Humans , Models, Molecular , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...