Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
ACS Synth Biol ; 13(3): 862-875, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38357862

ABSTRACT

Enzymes are indispensable biocatalysts for numerous industrial applications, yet stability, selectivity, and restricted substrate recognition present limitations for their use. Despite the importance of enzyme engineering in overcoming these limitations, success is often challenged by the intricate architecture of enzymes derived from natural sources. Recent advances in computational methods have enabled the de novo design of simplified scaffolds with specific functional sites. Such scaffolds may be advantageous as platforms for enzyme engineering. Here, we present a strategy for the de novo design of a simplified scaffold of an endo-α-N-acetylgalactosaminidase active site, a glycoside hydrolase from the GH101 enzyme family. Using a combination of trRosetta hallucination, iterative cycles of deep-learning-based structure prediction, and ProteinMPNN sequence design, we designed proteins with 290 amino acids incorporating the active site while reducing the molecular weight by over 100 kDa compared to the initial endo-α-N-acetylgalactosaminidase. Of 11 tested designs, six were expressed as soluble monomers, displaying similar or increased thermostabilities compared to the natural enzyme. Despite lacking detectable enzymatic activity, the experimentally determined crystal structures of a representative design closely matched the design with a root-mean-square deviation of 1.0 Å, with most catalytically important side chains within 2.0 Å. The results highlight the potential of scaffold hallucination in designing proteins that may serve as a foundation for subsequent enzyme engineering.


Subject(s)
Bacterial Proteins , Glycoside Hydrolases , Catalytic Domain , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/metabolism , Bacterial Proteins/metabolism , Substrate Specificity
2.
Chemistry ; 29(44): e202300982, 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37217457

ABSTRACT

Glycoside hydrolases (GHs) are a class of enzymes with emerging roles in a range of disease. Selective GH inhibitors are sought to better understand their functions and assess the therapeutic potential of modulating their activities. Iminosugars are a promising class of GH inhibitors but typically lack the selectivity required to accurately perturb biological systems. Here, we describe a concise synthesis of iminosugar inhibitors of N-acetyl-α-galactosaminidase (α-NAGAL), the GH responsible for cleaving terminal α-N-acetylgalactosamine residues from glycoproteins and other glycoconjugates. Starting from non-carbohydrate precursors, this modular synthesis supported the identification of a potent (490 nM) and α-NAGAL selective (∼200-fold) guanidino-containing derivative DGJNGuan. To illustrate the cellular activity of this new inhibitor, we developed a quantitative fluorescence image-based method to measure levels of the Tn-antigen, a cellular glycoprotein substrate of α-NAGAL. Using this assay, we show that DGJNGuan exhibits excellent inhibition of α-NAGAL within cells using patient derived fibroblasts (EC50 =150 nM). Moreover, in vitro and in cell assays to assess levels of lysosomal ß-hexosaminidase substrate ganglioside GM2 show that DGJNGuan is selective whereas DGJNAc exhibits off-target inhibition both in vitro and within cells. DGJNGuan is a readily produced and selective tool compound that should prove useful for investigating the physiological roles of α-NAGAL.


Subject(s)
Hexosaminidases , beta-N-Acetylhexosaminidases , Humans , alpha-N-Acetylgalactosaminidase/chemistry , Lysosomes , Glycoconjugates , Glycoproteins
3.
Arch Biochem Biophys ; 725: 109280, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35605676

ABSTRACT

Endo-α-N-acetylgalactosaminidase from Bifidobacterium longum (EngBF) belongs to the glycoside hydrolase family GH101 and has a strict preference towards the mucin type glycan, Galß1-3GalNAc, which is O-linked to serine or threonine residues on glycopeptides and -proteins. While other enzymes of the GH101 family exhibit a wider substrate spectrum, no GH101 member has until recently been reported to process the α2-3 sialidated mucin glycan, Neu5Acα2-3Galß1-3GalNAc. However, work published by others (ACS Chem Biol 2021, 16, 2004-2015) during the preparation of the present manuscript demonstrated that the enzymes from several bacteria are able to hydrolyze this glycan from the fluorophore, methylumbelliferyl. Based on molecular docking using the EngBF homolog, EngSP from Streptococcus pneumoniae, substitution of active site amino acid residues with the potential to allow for accommodation of Neu5Acα2-3Galß1-3GalNAc were identified. Based on this analysis, the mutant EngBF variants W750A, Q894A, K1199A, E1294A and D1295A were prepared and tested, for activity towards the Neu5Acα2-3Galß1-3GalNAc O-linked glycan present on bovine fetuin. Among the mutant EngBF variants listed above, only E1294A was shown to release Neu5Acα2-3Galß1-3GalNAc from fetuin, which subsequently was also demonstrated for the substitutions: E1294 M, E1294H and E1294K. In addition, the kcat/KM of the EngBF variants for cleavage of the Neu5Acα2-3Galß1-3GalNAc glycan increased between 5 and 70 times from pH 4.5 to pH 6.0.


Subject(s)
Bifidobacterium longum , Animals , Bifidobacterium longum/metabolism , Cattle , Fetuins , Molecular Docking Simulation , Mucins/metabolism , Polysaccharides/chemistry , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/genetics
4.
Biochimie ; 195: 90-99, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34826537

ABSTRACT

Glycoside hydrolase family 31 (GH31) is a diversified family of anomer-retaining α-glycoside hydrolases, such as α-glucosidase and α-xylosidase, among others. Recently, GH31 α-N-acetylgalactosaminidases (Nag31s) have been identified to hydrolyze the core of mucin-type O-glycans and the crystal structure of a gut bacterium Enterococcus faecalis Nag31 has been reported. However, the mechanisms of substrate specificity and hydrolysis of Nag31s are not well investigated. Herein, we show that E. faecalis Nag31 has the ability to release N-acetylgalactosamine (GalNAc) from O-glycoproteins, such as fetuin and mucin, but has low activity against Tn antigen. Mutational analysis and crystal structures of the Michaelis complexes reveal that residues of the active site work in concert with their conformational changes to act on only α-N-acetylgalactosaminides. Docking simulations using GalNAc-attached peptides suggest that the enzyme mainly recognizes GalNAc and side chains of Ser/Thr, but not strictly other peptide residues. Moreover, quantum mechanics calculations indicate that the enzyme preferred p-nitrophenyl α-N-acetylgalactosaminide to Tn antigen and that the hydrolysis progresses through a conformational itinerary, 4C1 → 1S3 → 4C1, in GalNAc of substrates. Our results provide novel insights into the diversification of the sugar recognition and hydrolytic mechanisms of GH31 enzymes.


Subject(s)
Glycoside Hydrolases , Catalytic Domain , Glycoside Hydrolases/chemistry , Hydrolysis , Substrate Specificity , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/metabolism
5.
Insect Mol Biol ; 30(4): 367-378, 2021 08.
Article in English | MEDLINE | ID: mdl-33742736

ABSTRACT

Horizontal gene transfer is an important evolutionary mechanism not only for bacteria but also for eukaryotes. In the domestic silkworm Bombyx mori, a model species of lepidopteran insects, some enzymes are known to have been acquired by horizontal transfer; however, the enzymatic features of protein BmNag31, belonging to glycoside hydrolase family 31 (GH31) and whose gene was predicted to be transferred from Enterococcus sp. are unknown. In this study, we reveal that the transcription of BmNag31 increases significantly during the prepupal to pupal stage, and decreases in the adult stage. The full-length BmNag31 and its truncated mutants were heterologously expressed in Escherichia coli and characterized. Its catalytic domain exhibits α-N-acetylgalactosaminidase activity and the carbohydrate-binding module family 32 domain shows binding activity towards N-acetylgalactosamine, similar to the Enterococcus faecalis homolog, EfNag31A. Gel filtration chromatography and blue native polyacrylamide gel electrophoresis analyses indicate that BmNag31 forms a hexamer whereas EfNag31A is monomeric. These results provide insights into the function of lepidopteran GH31 α-N-acetylgalactosaminidase.


Subject(s)
Bombyx , alpha-N-Acetylgalactosaminidase/chemistry , Animals , Biological Evolution , Bombyx/genetics , Bombyx/metabolism , Enterococcus/genetics , Escherichia coli , Gene Transfer, Horizontal , Genes, Bacterial , Glycoside Hydrolases/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , alpha-N-Acetylgalactosaminidase/genetics , alpha-N-Acetylgalactosaminidase/metabolism
6.
Org Biomol Chem ; 19(4): 789-793, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33411870

ABSTRACT

As part of a search for a substrate for droplet-based microfluidic screening assay of α-N-acetylgalactosaminidases, spectral and physical characteristics of a series of coumarin derivatives were measured. From among these a new coumarin-based fluorophore, Jericho Blue, was selected as having optimal characteristics for our screen. A reliable method for the challenging synthesis of coumarin glycosides of α-GalNAc was then developed and demonstrated with nine examples. The α-GalNAc glycoside of Jericho Blue prepared in this way was shown to function well under screening conditions.


Subject(s)
Coumarins/chemistry , Coumarins/chemical synthesis , Enzyme Assays/instrumentation , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Lab-On-A-Chip Devices , alpha-N-Acetylgalactosaminidase/metabolism , Chemistry Techniques, Synthetic , Substrate Specificity , alpha-N-Acetylgalactosaminidase/chemistry
7.
FEBS Lett ; 594(14): 2282-2293, 2020 07.
Article in English | MEDLINE | ID: mdl-32367553

ABSTRACT

Glycoside hydrolases catalyze the hydrolysis of glycosidic linkages in carbohydrates. The glycoside hydrolase family 31 (GH31) contains α-glucosidase, α-xylosidase, α-galactosidase, and α-transglycosylase. Recent work has expanded the diversity of substrate specificity of GH31 enzymes, and α-N-acetylgalactosaminidases (αGalNAcases) belonging to GH31 have been identified in human gut bacteria. Here, we determined the first crystal structure of a truncated form of GH31 αGalNAcase from the human gut bacterium Enterococcus faecalis. The enzyme has a similar fold to other reported GH31 enzymes and an additional fibronectin type 3-like domain. Additionally, the structure in complex with N-acetylgalactosamine reveals that conformations of the active site residues, including its catalytic nucleophile, change to recognize the ligand. Our structural analysis provides insight into the substrate recognition and catalytic mechanism of GH31 αGalNAcases.


Subject(s)
Enterococcus faecalis/enzymology , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/classification , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Enterococcus faecalis/genetics , Hydrolysis , Kinetics , Models, Molecular , Structure-Activity Relationship , alpha-N-Acetylgalactosaminidase/genetics
8.
J Mol Neurosci ; 70(1): 45-55, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31468281

ABSTRACT

Schindler disease is a rare autosomal recessive lysosomal storage disorder caused by a deficiency in alpha-N-acetylgalactosaminidase (α-NAGA) activity due to defects in the NAGA gene. Accumulation of the enzyme's substrates results in clinically heterogeneous symptoms ranging from asymptomatic individuals to individuals with severe neurological manifestations. Here, a 5-year-old Emirati male born to consanguineous parents presented with congenital microcephaly and severe neurological manifestations. Whole genome sequencing revealed a homozygous missense variant (c.838C>A; p.L280I) in the NAGA gene. The allele is a reported SNP in the ExAC database with a 0.0007497 allele frequency. The proband's asymptomatic sister and cousin carry the same genotype in a homozygous state as revealed from the family screening. Due to the extreme intrafamilial heterogeneity of the disease as seen in previously reported cases, we performed further analyses to establish the pathogenicity of this variant. Both the proband and his sister showed abnormal urine oligosaccharide patterns, which is consistent with the diagnosis of Schindler disease. The α-NAGA activity was significantly reduced in the proband and his sister with 5.9% and 12.1% of the mean normal activity, respectively. Despite the activity loss, p.L280I α-NAGA processing and trafficking were not affected. However, protein molecular dynamic simulation analysis revealed that this amino acid substitution is likely to affect the enzyme's natural dynamics and hinders its ability to bind to the active site. Functional analysis confirmed the pathogenicity of the identified missense variant and the diagnosis of Schindler disease. Extreme intrafamilial clinical heterogeneity of the disease necessitates further studies for proper genetic counseling and management.


Subject(s)
Lysosomal Storage Diseases/genetics , Mutation, Missense , Neuroaxonal Dystrophies/genetics , Phenotype , alpha-N-Acetylgalactosaminidase/deficiency , Adult , Catalytic Domain , Cells, Cultured , Child , Female , Humans , Lysosomal Storage Diseases/pathology , Male , Neuroaxonal Dystrophies/pathology , Pedigree , Protein Binding , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/genetics , alpha-N-Acetylgalactosaminidase/metabolism
9.
Sci Rep ; 9(1): 15199, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31645583

ABSTRACT

To overcome the laborious identification of crystallisation conditions for protein X-ray crystallography, we developed a method where the examined protein is immobilised as a guest molecule in a universal host lattice. We applied crystal engineering to create a generic crystalline host lattice under reproducible, predefined conditions and analysed the structures of target guest molecules of different size, namely two 15-mer peptides and green fluorescent protein (sfGFP). A fusion protein with an N-terminal endo-α-N-acetylgalactosaminidase (EngBF) domain and a C-terminal designed ankyrin repeat protein (DARPin) domain establishes the crystal lattice. The target is recruited into the host lattice, always in the same crystal form, through binding to the DARPin. The target structures can be determined rapidly from difference Fourier maps, whose quality depends on the size of the target and the orientation of the DARPin.


Subject(s)
Crystallization/methods , Crystallography, X-Ray/methods , Proteins/chemistry , Animals , Ankyrin Repeat , Bifidobacterium longum/enzymology , Databases, Protein , Green Fluorescent Proteins/chemistry , Humans , Models, Molecular , Peptides/chemistry , Protein Conformation , Protein Domains , Recombinant Fusion Proteins/chemistry , alpha-N-Acetylgalactosaminidase/chemistry
10.
FEBS Lett ; 593(1): 52-66, 2019 01.
Article in English | MEDLINE | ID: mdl-30411345

ABSTRACT

N-Acetylglucosamine-6-phosphate deacetylase (NagA) and glucosamine-6-phosphate deaminase (NagB) are branch point enzymes that direct amino sugars into different pathways. For Staphylococcus aureus NagA, analytical ultracentrifugation and small-angle X-ray scattering data demonstrate that it is an asymmetric dimer in solution. Initial rate experiments show hysteresis, which may be related to pathway regulation, and kinetic parameters similar to other bacterial isozymes. The enzyme binds two Zn2+ ions and is not substrate inhibited, unlike the Escherichia coli isozyme. S. aureus NagB adopts a novel dimeric structure in solution and shows kinetic parameters comparable to other Gram-positive isozymes. In summary, these functional data and solution structures are of use for understanding amino sugar metabolism in S. aureus, and will inform the design of inhibitory molecules.


Subject(s)
Aldose-Ketose Isomerases/chemistry , Aldose-Ketose Isomerases/metabolism , Staphylococcus aureus/enzymology , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Kinetics , Models, Molecular , Protein Multimerization , Scattering, Small Angle , Staphylococcus aureus/chemistry , Ultracentrifugation , X-Ray Diffraction , Zinc/metabolism
11.
Interdiscip Sci ; 10(1): 81-92, 2018 Mar.
Article in English | MEDLINE | ID: mdl-27138754

ABSTRACT

Schindler disease is a lysosomal storage disorder caused due to deficiency or defective activity of alpha-N-acetylgalactosaminidase (α-NAGA). Mutations in gene encoding α-NAGA cause wide range of diseases, characterized with mild to severe clinical features. Molecular effects of these mutations are yet to be explored in detail. Therefore, this study was focused on four missense mutations of α-NAGA namely, S160C, E325K, R329Q and R329W. Native and mutant structures of α-NAGA were analysed to determine geometrical deviations such as the contours of root mean square deviation, root mean square fluctuation, percentage of residues in allowed regions of Ramachandran plot and solvent accessible surface area, using conformational sampling technique. Additionally, global energy-minimized structures of native and mutants were further analysed to compute their intra-molecular interactions, hydrogen bond dilution and distribution of secondary structure. In addition, docking studies were also performed to determine variations in binding energies between native and mutants. The deleterious effects of mutants were evident due to variations in their active site residues pertaining to spatial conformation and flexibility, comparatively. Hence, variations exhibited by mutants, namely S160C, E325K, R329Q and R329W to that of native, consequently, lead to the detrimental effects causing Schindler disease. This study computationally explains the underlying reasons for the pathogenesis of the disease, thereby aiding future researchers in drug development and disease management.


Subject(s)
Point Mutation/genetics , alpha-N-Acetylgalactosaminidase/genetics , Amino Acid Sequence , Biocatalysis , Humans , Hydrogen Bonding , Mutant Proteins/chemistry , Protein Denaturation , Protein Structure, Secondary , Substrate Specificity , Temperature , alpha-N-Acetylgalactosaminidase/chemistry
12.
J Biol Chem ; 292(29): 12126-12138, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28546425

ABSTRACT

The α-N-acetylgalactosaminidase from the probiotic bacterium Bifidobacterium bifidum (NagBb) belongs to the glycoside hydrolase family 129 and hydrolyzes the glycosidic bond of Tn-antigen (GalNAcα1-Ser/Thr). NagBb is involved in assimilation of O-glycans on mucin glycoproteins by B. bifidum in the human gastrointestinal tract, but its catalytic mechanism has remained elusive because of a lack of sequence homology around putative catalytic residues and of other structural information. Here we report the X-ray crystal structure of NagBb, representing the first GH129 family structure, solved by the single-wavelength anomalous dispersion method based on sulfur atoms of the native protein. We determined ligand-free, GalNAc, and inhibitor complex forms of NagBb and found that Asp-435 and Glu-478 are located in the catalytic domain at appropriate positions for direct nucleophilic attack at the anomeric carbon and proton donation for the glycosidic bond oxygen, respectively. A highly conserved Asp-330 forms a hydrogen bond with the O4 hydroxyl of GalNAc in the -1 subsite, and Trp-398 provides a stacking platform for the GalNAc pyranose ring. Interestingly, a metal ion, presumably Ca2+, is involved in the recognition of the GalNAc N-acetyl group. Mutations at Asp-435, Glu-478, Asp-330, and Trp-398 and residues involved in metal coordination (including an all-Ala quadruple mutant) significantly reduced the activity, indicating that these residues and the metal ion play important roles in substrate recognition and catalysis. Interestingly, NagBb exhibited some structural similarities to the GH101 endo-α-N-acetylgalactosaminidases, but several critical differences in substrate recognition and reaction mechanism account for the different activities of these two enzymes.


Subject(s)
Acetylgalactosamine/metabolism , Bacterial Proteins/metabolism , Bifidobacterium bifidum/enzymology , Coenzymes/metabolism , Glycoside Hydrolases/metabolism , Metals/metabolism , alpha-N-Acetylgalactosaminidase/metabolism , Acetylgalactosamine/chemistry , Amino Acid Sequence , Amino Acid Substitution , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Catalytic Domain , Coenzymes/chemistry , Conserved Sequence , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Ligands , Metals/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Probiotics , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Structural Homology, Protein , alpha-N-Acetylgalactosaminidase/antagonists & inhibitors , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/genetics
13.
BMC Biochem ; 18(1): 9, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28545388

ABSTRACT

BACKGROUND: The marine invertebrate starfish was found to contain a novel α-N-acetylgalactosaminidase, α-GalNAcase II, which catalyzes removal of terminal α-N-acetylgalactosamine (α-GalNAc), in addition to a typical α-N-acetylgalactosaminidase, α-GalNAcase I, which catalyzes removal of terminal α-N-acetylgalactosamine (α-GalNAc) and, to a lesser extent, galactose. The interrelationship between α-GalNAcase I and α-GalNAcase II and the molecular basis of their differences in substrate specificity remain unknown. RESULTS: Chemical and structural comparisons between α-GalNAcase I and II using immunostaining, N-terminal amino acid sequencing and peptide analysis showed high homology to each other and also to other glycoside hydrolase family (GHF) 27 members. The amino acid sequence of peptides showed conserved residues at the active site as seen in typical α-GalNAcase. Some substitutions of conserved amino acid residues were found in α-GalNAcase II that were located near catalytic site. Among them G171 and A173, in place of C171 and W173, respectively in α-GalNAcase were identified to be responsible for lacking intrinsic α-galactosidase activity of α-GalNAcase II. Chemical modifications supported the presence of serine, aspartate and tryptophan as active site residues. Two tryptophan residues (W16 and W173) were involved in α-galactosidase activity, and one (W16) of them was involved in α-GalNAcase activity. CONCLUSIONS: The results suggested that α-GalNAcase I and II are closely related with respect to primary and higher order structure and that their structural differences are responsible for difference in substrate specificities.


Subject(s)
Asterina/enzymology , alpha-N-Acetylgalactosaminidase/chemistry , Animals , Catalytic Domain , Molecular Sequence Data , Sequence Homology, Amino Acid , Substrate Specificity , alpha-Galactosidase/metabolism , alpha-N-Acetylgalactosaminidase/metabolism
14.
Arch Biochem Biophys ; 606: 167-79, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27503803

ABSTRACT

Disagreement exists regarding the O-glycan structure attached to human vitamin D binding protein (DBP). Previously reported evidence indicated that the O-glycan of the Gc1S allele product is the linear core 1 NeuNAc-Gal-GalNAc-Thr trisaccharide. Here, glycan structural evidence is provided from glycan linkage analysis and over 30 serial glycosidase-digestion experiments which were followed by analysis of the intact protein by electrospray ionization mass spectrometry (ESI-MS). Results demonstrate that the O-glycan from the Gc1F protein is the same linear trisaccharide found on the Gc1S protein and that the hexose residue is galactose. In addition, the putative anti-cancer derivative of DBP known as Gc Protein-derived Macrophage Activating Factor (GcMAF, which is formed by the combined action of ß-galactosidase and neuraminidase upon DBP) was analyzed intact by ESI-MS, revealing that the activating E. coli ß-galactosidase cleaves nothing from the protein-leaving the glycan structure of active GcMAF as a Gal-GalNAc-Thr disaccharide, regardless of the order in which ß-galactosidase and neuraminidase are applied. Moreover, glycosidase digestion results show that α-N-Acetylgalactosamindase (nagalase) lacks endoglycosidic function and only cleaves the DBP O-glycan once it has been trimmed down to a GalNAc-Thr monosaccharide-precluding the possibility of this enzyme removing the O-glycan trisaccharide from cancer-patient DBP in vivo.


Subject(s)
Macrophage-Activating Factors/chemistry , Polysaccharides/chemistry , Alleles , Disaccharides/chemistry , Escherichia coli/enzymology , Glycoside Hydrolases/chemistry , Glycosides/chemistry , Glycosylation , Humans , Macrophage Activation , Mannose/chemistry , Neuraminidase/chemistry , Spectrometry, Mass, Electrospray Ionization , Trifluoroacetic Acid/chemistry , Trisaccharides/chemistry , Vitamin D-Binding Protein/chemistry , alpha-N-Acetylgalactosaminidase/chemistry , beta-Galactosidase/chemistry
15.
Glycobiology ; 25(8): 806-11, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25964111

ABSTRACT

A facile enzymatic synthesis of the methylumbelliferyl ß-glycoside of the type 2 A blood group tetrasaccharide in good yields is reported. Using this compound, we developed highly sensitive fluorescence-based high-throughput assays for both endo-ß-galactosidase and α-N-acetylgalactosaminidase activity specific for the oligosaccharide structure of the blood group A antigen. We further demonstrate the potential to use this assay to screen the expressed gene products of metagenomic libraries in the search for efficient blood group antigen-cleaving enzymes.


Subject(s)
ABO Blood-Group System/chemistry , Glycosides/chemical synthesis , Hymecromone/chemical synthesis , Oligosaccharides/chemical synthesis , alpha-N-Acetylgalactosaminidase/chemistry , beta-Galactosidase/chemistry , ABO Blood-Group System/metabolism , Biological Assay , Escherichia coli/enzymology , Escherichia coli/genetics , Fluorescence , Gene Expression , Gene Library , Glycosides/biosynthesis , High-Throughput Screening Assays , Humans , Hymecromone/metabolism , Metagenome , Oligosaccharides/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , alpha-N-Acetylgalactosaminidase/genetics , alpha-N-Acetylgalactosaminidase/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
16.
Org Lett ; 16(21): 5512-5, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25330411

ABSTRACT

The synthesis of 1,2-cis-homoiminosugars bearing an NHAc group at the C-2 position is described. The key step to prepare these α-D-GlcNAc and α-D-GalNAc mimics utilizes a ß-amino alcohol skeletal rearrangement applied to an azepane precursor. This strategy also allows access to naturally occurring α-HGJ and α-HNJ. The α-D-GlcNAc-configured iminosugar was coupled to a glucoside acceptor to yield a novel pseudodisaccharide. Preliminary glycosidase inhibition evaluation indicates that the α-D-GalNAc-configured homoiminosugar is a potent and selective α-N-acetylgalactosaminidase inhibitor.


Subject(s)
Amino Alcohols/chemistry , Amino Sugars/chemistry , Enzyme Inhibitors/chemistry , Galactosamine/chemistry , Glucosamine/chemistry , alpha-N-Acetylgalactosaminidase/antagonists & inhibitors , alpha-N-Acetylgalactosaminidase/chemistry , Galactosamine/analogs & derivatives , Glucosamine/analogs & derivatives , Molecular Structure
17.
Blood Transfus ; 12(1): 61-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24333060

ABSTRACT

BACKGROUND: It is well known that the buffer plays a key role in the enzymatic reaction involved in blood group conversion. In previous study, we showed that a glycine buffer is suitable for A to O or B to O blood group conversion. In this study, we investigated the use of 5% glucose and other buffers for A to O or B to O blood group conversion by α-N-acetylgalactosaminidase or α-galactosidase. MATERIALS AND METHODS: We compared the binding ability of α-N-acetylgalactosaminidase/α-galactosidase with red blood cells (RBC) in different reaction buffers, such as normal saline, phosphate-buffered saline (PBS), a disodium hydrogen phosphate-based buffer (PCS), and 5% commercial glucose solution. The doses of enzymes necessary for the A/B to O conversion in different reaction buffers were determined and compared. The enzymes' ability to bind to RBC was evaluated by western blotting, and routine blood typing and fluorescence activated cell sorting was used to evaluate B/A to O conversion efficiency. RESULTS: The A to O conversion efficiency in glucose buffer was similar to that in glycine buffer with the same dose (>0.06 mg/mL pRBC). B to O conversion efficiency in glucose buffer was also similar to that in glycine buffer with the same dose (>0.005 mg/mL pRBC). Most enzymes could bind with RBC in glycine or glucose buffer, but few enzymes could bind with RBC in PBS, PCS, or normal saline. CONCLUSION: These results indicate that 5% glucose solution provides a suitable condition for enzymolysis, especially for enzymes combining with RBC. Meanwhile, the conversion efficiency of A/B to O was similar in glucose buffer and glycine buffer. Moreover, 5% glucose solution has been used for years in venous transfusion, it is safe for humans and its cost is lower. Our results do, therefore, suggest that 5% glucose solution could become a novel suitable buffer for A/B to O blood group conversion.


Subject(s)
Bacterial Proteins/chemistry , Blood Group Antigens/chemistry , Erythrocytes/chemistry , Glucose/chemistry , alpha-Galactosidase/chemistry , alpha-N-Acetylgalactosaminidase/chemistry , Bacteroides fragilis/enzymology , Buffers , Humans
18.
Artif Cells Nanomed Biotechnol ; 41(1): 32-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23030311

ABSTRACT

BACKGROUND: Enzymatical conversion of A or B RBCs into group O RBCs (ECORBCs) was achieved by using α-N-acetylgalactosaminidase and α-galactosidase, respectively. Now, we initiated AB to O-RBC conversion by using these two enzymes together. But α-N-acetylgalactosaminidase and α-galactosidase's preserving and their reaction buffer were quite different. The aim of this study is to confirm an available system for converting AB to O RBCs, especially to study the maximal permission amount of PCS which was brought to the system-accompanied enzyme addition. METHOD: Enzyme activity was detected by using GalNAc-pNp or Gal-pNp as substrates. The efficiency of the conversion of A or B antigen was evaluated by routine method and measured by fluorescence-activated cell sorting analysis. The optimal buffer component and the doses of α-N-acetylgalactosaminidase and α-galactosidase were confirmed according to A and B antigen epitope removal efficiency. RESULTS: The activity of α-N-acetylgalactosaminidase and α-galactosidase was not decreased drastically when they were kept in PCS Buffer in 4°C. The optimal reaction buffer composed of glycine 250 mM and NaCl 3 mM, pH 6.8 and PCS less than 10%(v/v). For converting A(1)B to O RBCs completely, the doses of α-N-acetylgalactosaminidase and α-galactosidase were confirmed as 0.015 mg/ml packed RBCs(pRBCs) for A(1) antigen epitopes and 0.005 mg/ml pRBCs for B epitopes. Approximately 0.004 mg α-N-acetylgalactosaminidase and 0.005 mg α-galactosidase were required to convert 1 ml pRBCs. CONCLUSION: Our studies indicated that α-N-acetylgalactosaminidase and α-galactosidase were stable in PCS buffer and a modified protocol which was propitious to converting AB to O RBCs was provided.


Subject(s)
ABO Blood-Group System/metabolism , Blood Transfusion/methods , Erythrocytes/metabolism , alpha-Galactosidase/metabolism , alpha-N-Acetylgalactosaminidase/metabolism , ABO Blood-Group System/chemistry , Buffers , Cell Separation , Flow Cytometry , Humans , Protein Stability , alpha-Galactosidase/chemistry , alpha-N-Acetylgalactosaminidase/chemistry
19.
Parasitol Res ; 111(5): 2149-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926676

ABSTRACT

The α-N-acetylgalactosaminidase (α-NAGAL) is an exoglycosidase that selectively cleaves terminal α-linked N-acetylgalactosamines from a variety of sugar chains. A complementary DNA (cDNA) clone encoding a novel Clonorchis sinensis α-NAGAL (Cs-α-NAGAL) was identified in the expressed sequence tags database of the adult C. sinensis liver fluke. The complete coding sequence was 1,308 bp long and encoded a 436-residue protein. The selected glycosidase was manually curated as α-NAGAL (EC 3.2.1.49) based on a composite bioinformatics analysis including a search for orthologues, comparative structure modeling, and the generation of a phylogenetic tree. One orthologue of Cs-α-NAGAL was the Rattus norvegicus α-NAGAL (accession number: NP_001012120) that does not exist in C. sinensis. Cs-α-NAGAL belongs to the GH27 family and the GH-D clan. A phylogenetic analysis revealed that the GH27 family of Cs-α-NAGAL was distinct from GH31 and GH36 within the GH-D clan. The putative 3D structure of Cs-α-NAGAL was built using SWISS-MODEL with a Gallus gallus α-NAGAL template (PDB code 1ktb chain A); this model demonstrated the superimposition of a TIM barrel fold (α/ß) structure and substrate binding pocket. Cs-α-NAGAL transcripts were detected in the adult worm and egg cDNA libraries of C. sinensis but not in the metacercaria. Recombinant Cs-α-NAGAL (rCs-α-NAGAL) was expressed in Escherichia coli, and the purified rCs-α-NAGAL was recognized specifically by the C. sinensis-infected human sera. This is the first report of an α-NAGAL protein in the Trematode class, suggesting that it is a potential diagnostic or vaccine candidate with strong antigenicity.


Subject(s)
Clonorchis sinensis/enzymology , alpha-N-Acetylgalactosaminidase/genetics , Animals , Antibodies, Helminth/blood , Antigens, Helminth/chemistry , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Cloning, Molecular , Clonorchiasis/immunology , Clonorchis sinensis/chemistry , Clonorchis sinensis/genetics , Clonorchis sinensis/immunology , Cluster Analysis , Escherichia coli/genetics , Gene Expression Profiling , Humans , Models, Molecular , Phylogeny , Protein Conformation , Sequence Analysis, DNA , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/immunology
20.
Protein Expr Purif ; 81(1): 106-114, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21982820

ABSTRACT

α-N-Acetylgalactosaminidase (α-GalNAc-ase; EC.3.2.1.49) is an exoglycosidase specific for the hydrolysis of terminal α-linked N-acetylgalactosamine in various sugar chains. The cDNA corresponding to the α-GalNAc-ase gene was cloned from Aspergillus niger, sequenced, and expressed in the yeast Saccharomyces cerevisiae. The α-GalNAc-ase gene contains an open reading frame which encodes a protein of 487 amino acid residues. The molecular mass of the mature protein deduced from the amino acid sequence of this reading frame is 54 kDa. The recombinant protein was purified to apparent homogeneity and biochemically characterized (pI4.4, K(M) 0.56 mmol/l for 2-nitrophenyl 2-acetamido-2-deoxy-α-d-galactopyranoside, and optimum enzyme activity was achieved at pH2.0-2.4 and 50-55°C). Its molecular weight was determined by analytical ultracentrifuge measurement and dynamic light scattering. Our experiments confirmed that the recombinant α-GalNAc-ase exists as two distinct species (70 and 130 kDa) compared to its native form, which is purely monomeric. N-Glycosylation was confirmed at six of the eight potential N-glycosylation sites in both wild type and recombinant α-GalNAc-ase.


Subject(s)
Aspergillus niger/enzymology , Recombinant Proteins/biosynthesis , Saccharomyces cerevisiae/genetics , alpha-N-Acetylgalactosaminidase/biosynthesis , Amino Acid Sequence , Aspergillus niger/genetics , Cell Culture Techniques , Chromatography, Gel , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Glycosylation , Hydrogen-Ion Concentration , Microscopy, Electron , Molecular Sequence Data , Molecular Weight , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Saccharomyces cerevisiae/enzymology , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...