Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
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.
Biochem Biophys Res Commun ; 706: 149746, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38461646

ABSTRACT

Polyglycine hydrolases are fungal effectors composed of an N-domain with unique sequence and structure and a C-domain that resembles ß-lactamases, with serine protease activity. These secreted fungal proteins cleave Gly-Gly bonds within a polyglycine sequence in corn ChitA chitinase. The polyglycine hydrolase N-domain (PND) function is unknown. In this manuscript we provide evidence that the PND does not directly participate in ChitA cleavage. In vitro analysis of site-directed mutants in conserved residues of the PND of polyglycine hydrolase Es-cmp did not specifically impair protease activity. Furthermore, in silico structural models of three ChitA-bound polyglycine hydrolases created by High Ambiguity Driven protein-protein DOCKing (HADDOCK) did not predict significant interactions between the PND and ChitA. Together these results suggest that the PND has another function. To determine what types of PND-containing proteins exist in nature we performed a computational analysis of Foldseek-identified PND-containing proteins. The analysis showed that proteins with PNDs are present throughout biology as either single domain proteins or fused to accessory domains that are diverse but are usually proteases or kinases.


Subject(s)
Peptide Hydrolases , Peptides , Peptides/chemistry , Peptide Hydrolases/metabolism , Endopeptidases/metabolism , Proteolysis
3.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S11-S13, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29762369

ABSTRACT

OBJECTIVES: Maltase-glucoamylase and sucrase-isomaltase are enzymes in the brush-border membrane of the small intestinal lumen responsible for the breakdown of postamylase starch polysaccharides to release monomeric glucose. As such, they are critical players in healthy nutrition and their malfunction can lead to severe disorders. METHODS: This review covers investigations of the structures and functions of these enzymes. RESULTS: Each consists of 2 enzyme domains of the glycoside hydrolase family GH31 classification, yet with somewhat differing enzymatic properties. CONCLUSIONS: Crystallographic structures of 3 of the domains have been published. Insights into substrate binding and specificity will be discussed, along with future lines of inquiry related to the enzymes' roles in disease and potential avenues for therapeutics.


Subject(s)
Intestine, Small/physiology , Sucrase-Isomaltase Complex/chemistry , alpha-Glucosidases/chemistry , Crystallography , Humans , Starch/metabolism , Sucrase-Isomaltase Complex/physiology , alpha-Glucosidases/physiology
4.
Antonie Van Leeuwenhoek ; 111(9): 1645-1660, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29492769

ABSTRACT

The plant growth-promoting rhizobacterium Pseudomonas sp. UW4 was transformed to increase the biosynthesis of the auxin, indole-3-acetic acid (IAA). Four native IAA biosynthesis genes from strain UW4 were individually cloned into an expression vector and introduced back into the wild-type strain. Quantitative real-time polymerase chain reaction analysis revealed that the introduced genes ami, nit, nthAB and phe were all overexpressed in these transformants. A significant increase in the production of IAA was observed for all modified strains. Canola plants inoculated with the modified strains showed enhanced root elongation under gnotobiotic conditions. The growth rate and 1-aminocyclopropane-1-carboxylate deaminase activity of transformant strains was lower compared to the wild-type. The indoleacetic acid biosynthesis pathways and the role of this phytohormone in the mechanism of plant growth stimulation by Pseudomonas sp. UW4 is discussed.


Subject(s)
Biosynthetic Pathways/genetics , Indoleacetic Acids/metabolism , Pseudomonas/genetics , Bacterial Load , Bacterial Proteins/genetics , Carbon-Carbon Lyases/metabolism , Cloning, Molecular , Gene Expression , Genetic Vectors , Germ-Free Life , Indoleacetic Acids/chemistry , Molecular Structure , Plant Growth Regulators/chemistry , Plant Growth Regulators/metabolism , Plant Roots/growth & development , Pseudomonas/growth & development , Pseudomonas/metabolism , Real-Time Polymerase Chain Reaction
5.
Biochem Cell Biol ; 94(3): 241-6, 2016 06.
Article in English | MEDLINE | ID: mdl-27093479

ABSTRACT

The human digestive system is host to a highly populated ecosystem of bacterial species that significantly contributes to our assimilation of dietary carbohydrates. Bacteroides thetaiotaomicron is a member of this ecosystem, and participates largely in the role of the gut microbiome by breaking down dietary complex carbohydrates. This process of acquiring glycans from the colon lumen is predicted to rely on the mechanisms of proteins that are part of a classified system known as polysaccharide utilization loci (PUL). These loci are responsible for binding substrates at the cell outer membrane, internalizing them, and then hydrolyzing them within the periplasm into simple sugars. Here we report our investigation into specific components of a PUL, and suggest an alternative starch utilization system in B. thetaiotaomicron. Our analysis of an outer membrane binding protein, a SusD homolog, highlights its contribution to this PUL by acquiring starch-based sugars from the colon lumen. Through our structural characterization of two Family GH31 α-glucosidases, we reveal the flexibility of this bacterium with respect to utilizing a range of starch-derived glycans with an emphasis on branched substrates. With these results we demonstrate the predicted function of a gene locus that is capable of contributing to starch hydrolysis in the human colon.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacteroides thetaiotaomicron/metabolism , Colon/microbiology , Polysaccharides/metabolism , Starch/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacteroides thetaiotaomicron/chemistry , Crystallography, X-Ray , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Structural Homology, Protein , alpha-Glucosidases/chemistry
6.
J Biol Chem ; 288(19): 13563-74, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23536181

ABSTRACT

BACKGROUND: The enzyme "GluI" is key to the synthesis of critical glycoproteins in the cell. RESULTS: We have determined the structure of GluI, and modeled binding with its unique sugar substrate. CONCLUSION: The specificity of this interaction derives from a unique conformation of the substrate. SIGNIFICANCE: Understanding the mechanism of the enzyme is of basic importance and relevant to potential development of antiviral inhibitors. Processing α-glucosidase I (GluI) is a key member of the eukaryotic N-glycosylation processing pathway, selectively catalyzing the first glycoprotein trimming step in the endoplasmic reticulum. Inhibition of GluI activity impacts the infectivity of enveloped viruses; however, despite interest in this protein from a structural, enzymatic, and therapeutic standpoint, little is known about its structure and enzymatic mechanism in catalysis of the unique glycan substrate Glc3Man9GlcNAc2. The first structural model of eukaryotic GluI is here presented at 2-Å resolution. Two catalytic residues are proposed, mutations of which result in catalytically inactive, properly folded protein. Using Autodocking methods with the known substrate and inhibitors as ligands, including a novel inhibitor characterized in this work, the active site of GluI was mapped. From these results, a model of substrate binding has been formulated, which is most likely conserved in mammalian GluI.


Subject(s)
Membrane Glycoproteins/chemistry , Molecular Docking Simulation , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , alpha-Glucosidases/chemistry , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Catalytic Domain , Crystallography, X-Ray , Glucose/chemistry , Glycoside Hydrolase Inhibitors , Hydrogen Bonding , Kinetics , Membrane Glycoproteins/antagonists & inhibitors , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Structural Homology, Protein , Substrate Specificity , Trisaccharides/chemistry
7.
Appl Environ Microbiol ; 80(15): 4640-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24837382

ABSTRACT

Indole-3-acetic acid (IAA) is a fundamental phytohormone with the ability to control many aspects of plant growth and development. Pseudomonas sp. strain UW4 is a rhizospheric plant growth-promoting bacterium that produces and secretes IAA. While several putative IAA biosynthetic genes have been reported in this bacterium, the pathways leading to the production of IAA in strain UW4 are unclear. Here, the presence of the indole-3-acetamide (IAM) and indole-3-acetaldoxime/indole-3-acetonitrile (IAOx/IAN) pathways of IAA biosynthesis is described, and the specific role of two of the enzymes (nitrilase and nitrile hydratase) that mediate these pathways is assessed. The genes encoding these two enzymes were expressed in Escherichia coli, and the enzymes were isolated and characterized. Substrate-feeding assays indicate that the nitrilase produces both IAM and IAA from the IAN substrate, while the nitrile hydratase only produces IAM. The two nitrile-hydrolyzing enzymes have very different temperature and pH optimums. Nitrilase prefers a temperature of 50°C and a pH of 6, while nitrile hydratase prefers 4°C and a pH of 7.5. Based on multiple sequence alignments and motif analyses, physicochemical properties and enzyme assays, it is concluded that the UW4 nitrilase has an aromatic substrate specificity. The nitrile hydratase is identified as an iron-type metalloenzyme that does not require the help of a P47K activator protein to be active. These data are interpreted in terms of a preliminary model for the biosynthesis of IAA in this bacterium.


Subject(s)
Aminohydrolases/metabolism , Bacterial Proteins/metabolism , Hydro-Lyases/metabolism , Indoleacetic Acids/metabolism , Indoles/metabolism , Pseudomonas/enzymology , Amino Acid Sequence , Aminohydrolases/chemistry , Aminohydrolases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biotransformation , Enzyme Stability , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Hydrogen-Ion Concentration , Molecular Sequence Data , Pseudomonas/chemistry , Pseudomonas/genetics , Pseudomonas/metabolism , Sequence Alignment , Substrate Specificity , Temperature
8.
J Biol Chem ; 287(44): 36917-21, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22988246

ABSTRACT

The quality of starch digestion, related to the rate and extent of release of dietary glucose, is associated with glycemia-related problems such as diabetes and other metabolic syndrome conditions. Here, we found that the rate of glucose generation from starch is unexpectedly associated with mucosal α-glucosidases and not just α-amylase. This understanding could lead to a new approach to regulate the glycemic response and glucose-related physiologic responses in the human body. There are six digestive enzymes for starch: salivary and pancreatic α-amylases and four mucosal α-glucosidases, including N- and C-terminal subunits of both maltase-glucoamylase and sucrase-isomaltase. Only the mucosal α-glucosidases provide the final hydrolytic activities to produce substantial free glucose. We report here the unique and shared roles of the individual α-glucosidases for α-glucans persisting after starch is extensively hydrolyzed by α-amylase (to produce α-limit dextrins (α-LDx)). All four α-glucosidases share digestion of linear regions of α-LDx, and three can hydrolyze branched fractions. The α-LDx, which were derived from different maize cultivars, were not all equally digested, revealing that the starch source influences glucose generation at the mucosal α-glucosidase level. We further discovered a fraction of α-LDx that was resistant to the extensive digestion by the mucosal α-glucosidases. Our study further challenges the conventional view that α-amylase is the only rate-determining enzyme involved in starch digestion and better defines the roles of individual and collective mucosal α-glucosidases. Strategies to control the rate of glucogenesis at the mucosal level could lead to regulation of the glycemic response and improved glucose management in the human body.


Subject(s)
Carbohydrate Metabolism , Dextrins/chemistry , Glucose/chemistry , Mucous Membrane/enzymology , alpha-Glucosidases/chemistry , Animals , Humans , Hydrolysis , Kinetics , Mice , Molecular Weight , Protein Subunits/chemistry , Starch/chemistry , Zea mays/chemistry , alpha-Amylases/chemistry
9.
J Biol Chem ; 287(38): 31929-38, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22851177

ABSTRACT

Starch digestion involves the breakdown by α-amylase to small linear and branched malto-oligosaccharides, which are in turn hydrolyzed to glucose by the mucosal α-glucosidases, maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI). MGAM and SI are anchored to the small intestinal brush-border epithelial cells, and each contains a catalytic N- and C-terminal subunit. All four subunits have α-1,4-exohydrolytic glucosidase activity, and the SI N-terminal subunit has an additional exo-debranching activity on the α-1,6-linkage. Inhibition of α-amylase and/or α-glucosidases is a strategy for treatment of type 2 diabetes. We illustrate here the concept of "toggling": differential inhibition of subunits to examine more refined control of glucogenesis of the α-amylolyzed starch malto-oligosaccharides with the aim of slow glucose delivery. Recombinant MGAM and SI subunits were individually assayed with α-amylolyzed waxy corn starch, consisting mainly of maltose, maltotriose, and branched α-limit dextrins, as substrate in the presence of four different inhibitors: acarbose and three sulfonium ion compounds. The IC(50) values show that the four α-glucosidase subunits could be differentially inhibited. The results support the prospect of controlling starch digestion rates to induce slow glucose release through the toggling of activities of the mucosal α-glucosidases by selective enzyme inhibition. This approach could also be used to probe associated metabolic diseases.


Subject(s)
Glucose/metabolism , Mucous Membrane/enzymology , alpha-Glucosidases/metabolism , Animals , Diabetes Mellitus/metabolism , Drosophila melanogaster , Glycoside Hydrolases/chemistry , Glycosylation , Humans , Hydrolysis , Inhibitory Concentration 50 , Intestinal Mucosa/metabolism , Kinetics , Mice , Models, Chemical , Obesity/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry
10.
Acta Crystallogr D Struct Biol ; 79(Pt 2): 168-176, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36762862

ABSTRACT

Polyglycine hydrolases (PGHs) are secreted fungal proteases that cleave the polyglycine linker of Zea mays ChitA, a defensive chitinase, thus overcoming one mechanism of plant resistance to infection. Despite their importance in agriculture, there has been no previous structural characterization of this family of proteases. The objective of this research was to investigate the proteolytic mechanism and other characteristics by structural and biochemical means. Here, the first atomic structure of a polyglycine hydrolase was identified. It was solved by X-ray crystallography using a RoseTTAFold model, taking advantage of recent technical advances in structure prediction. PGHs are composed of two domains: the N- and C-domains. The N-domain is a novel tertiary fold with an as-yet unknown function that is found across all kingdoms of life. The C-domain shares structural similarities with class C ß-lactamases, including a common catalytic nucleophilic serine. In addition to insights into the PGH family and its relationship to ß-lactamases, the results demonstrate the power of complementing experimental structure determination with new computational techniques.


Subject(s)
Chitinases , Peptides , Peptide Hydrolases , beta-Lactamases/chemistry , Chitinases/chemistry , Endopeptidases , Crystallography, X-Ray
11.
Protein Expr Purif ; 86(2): 135-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23036359

ABSTRACT

Microorganisms in the human gut outnumber human cells by a factor of 10. These microbes have been shown to have relevance to the human immune, nutrition and metabolic systems. A dominant symbiont of this environment is Bacteroides thetaiotaomicron which is characterized as being involved in degrading non-digestible plant polysaccharides. This organism's genome is highly enriched in genes predicted to be involved in the hydrolysis of various glycans. Presented here is a comparative functional analysis of two α-glucosidases (designated BT_0339 and BT_3299), Family 31 Glycoside Hydrolases from B. thetaiotaomicron. The purpose of this research is to explore the contributions these enzymes may have to human nutrition and specifically starch digestion. Expression of both α-glucosidases in pET-29a expression vector resulted in high levels of expressed protein in the soluble fraction. Two-step purification allowed for the isolation of the enzymes of interest in significant yield and fractions were observed to be homogenous. Both enzymes demonstrated activity on maltose, isomaltose and malto-oligosaccharide substrates and low level of activity on lactose and sucrose. Enzymatic kinetics revealed these enzymes both preferentially cleave the α1-6 linkage in comparison to the expected α1-4 and specifically favor maltose-derived substrates of longer length. The flexible hydrolytic capabilities of BT_0339 and BT_3299 reveal the ability of this bacterium to maintain its dominant position in its environment by utilizing an array of substrates. Specifically, these enzymes demonstrate an important aspect of this organism's contribution to starch digestion in the distal gut and the overall energy intake of humans.


Subject(s)
Bacterial Proteins/biosynthesis , Bacterial Proteins/isolation & purification , Bacteroides/enzymology , alpha-Glucosidases/biosynthesis , alpha-Glucosidases/isolation & purification , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacteroides/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Maltose/chemistry , Maltose/metabolism , Models, Molecular , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sequence Alignment , alpha-Glucosidases/chemistry , alpha-Glucosidases/genetics
12.
Food Chem ; 383: 132456, 2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35182873

ABSTRACT

Isomaltooligosaccharides (IMOs) have been characterized as dietary fibers that resist digestion in the small intestine; however, previous studies suggested that various α-glycosidic linkages in IMOs were hydrolyzed by mammalian α-glucosidases. This study investigated the hydrolysis of IMOs by small intestinal α-glucosidases from rat and human recombinant sucrase-isomaltase complex compared to commonly used fungal amyloglucosidase (AMG) in vitro. Interestingly, mammalian α-glucosidases fully hydrolyzed various IMOs to glucose at a slow rate compared with linear maltooligosaccharides, whereas AMG could not fully hydrolyze IMOs because of its very low hydrolytic activity on α-1,6 linkages. This suggests that IMOs have been misjudged as prebiotic ingredients that bypass the small intestine due to the nature of the assay used. Instead, IMOs can be applied in the food industry as slowly digestible materials to regulate the glycemic response and energy delivery in the mammalian digestive system, rather than as dietary fibers.


Subject(s)
Dietary Fiber , alpha-Glucosidases , Animals , Blood Glucose , Dietary Carbohydrates , Glucose , Hydrolysis , Mammals , Rats
13.
J Biol Chem ; 285(23): 17763-70, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20356844

ABSTRACT

Human maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI) are small intestinal enzymes that work concurrently to hydrolyze the mixture of linear alpha-1,4- and branched alpha-1,6-oligosaccharide substrates that typically make up terminal starch digestion products. MGAM and SI are each composed of duplicated catalytic domains, N- and C-terminal, which display overlapping substrate specificities. The N-terminal catalytic domain of human MGAM (ntMGAM) has a preference for short linear alpha-1,4-oligosaccharides, whereas N-terminal SI (ntSI) has a broader specificity for both alpha-1,4- and alpha-1,6-oligosaccharides. Here we present the crystal structure of the human ntSI, in apo form to 3.2 A and in complex with the inhibitor kotalanol to 2.15 A resolution. Structural comparison with the previously solved structure of ntMGAM reveals key active site differences in ntSI, including a narrow hydrophobic +1 subsite, which may account for its additional substrate specificity for alpha-1,6 substrates.


Subject(s)
Oligo-1,6-Glucosidase/chemistry , Sucrase/chemistry , alpha-Glucosidases/chemistry , Animals , Carbohydrates/chemistry , Crystallography, X-Ray/methods , Diabetes Mellitus/metabolism , Drosophila/metabolism , Humans , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Kinetics , Polysaccharides/chemistry , Protein Structure, Tertiary , Substrate Specificity
14.
Chemistry ; 17(52): 14817-25, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22127878

ABSTRACT

The synthesis and glucosidase inhibitory activities of two C-3'- and C-5'-ß-maltose-extended analogues of the naturally occurring sulfonium-ion inhibitor, de-O-sulfonated ponkoranol, are described. The compounds are designed to test the specificity towards four intestinal glycoside hydrolase family 31 (GH31) enzyme activities, responsible for the hydrolysis of terminal starch products and sugars into glucose, in humans. The target sulfonium-ion compounds were synthesized by means of nucleophilic attack of benzyl protected 1,4-anhydro-4-thio-D-arabinitol at the C-6 position of 6-O-trifluoromethanesulfonyl trisaccharides as alkylating agents. The alkylating agents were synthesized from D-glucose by glycosylation at C-4 or C-2 with maltosyl trichloroacetimidate. Deprotection of the coupled products by using a two-step sequence, followed by reduction afforded the final compounds. Evaluation of the target compounds for inhibition of the four glucosidase activities indicated that selective inhibition of one enzyme over the others is possible.


Subject(s)
Intestinal Mucosa/metabolism , Intestines/enzymology , Maltose/antagonists & inhibitors , Maltose/chemistry , Maltose/chemical synthesis , Starch/chemistry , Sucrase-Isomaltase Complex/antagonists & inhibitors , Sucrase-Isomaltase Complex/metabolism , Sulfonium Compounds/antagonists & inhibitors , Sulfonium Compounds/chemistry , Thiophenes/antagonists & inhibitors , Thiophenes/chemical synthesis , alpha-Glucosidases/chemistry , alpha-Glucosidases/chemical synthesis , Catalysis , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Stereoisomerism , Thiophenes/chemistry
15.
Protein Expr Purif ; 79(1): 96-101, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21640829

ABSTRACT

Eukaryotic N-glycoprotein processing in the endoplasmic reticulum begins with the catalytic action of processing α-glucosidase I (αGlu). αGlu trims the terminal glucose from nascent glycoproteins in an inverting-mechanism glycoside hydrolysis reaction. αGlu has been studied in terms of kinetic parameters and potential key residues; however, the active site is unknown. A structural model would yield important insights into the reaction mechanism. A model would also be useful in developing specific therapeutics, as αGlu is a viable drug target against viruses with glycosylated envelope proteins. However, due to lack of a high-yielding overexpression and purification scheme, no eukaryotic structural model of αGlu has been determined. To address this issue, we overexpressed the Saccharomyces cerevisiae soluble αGlu, Cwht1p, in the host Pichia pastoris. It was purified in a simple two-step protocol, with a final yield of 4.2mg Cwht1p per liter of growth culture. To test catalytic activity, we developed a modified synthesis of a tetrasaccharide substrate, Glc(3)ManOMe. Cwht1p with Glc(3)ManOMe shows a K(m) of 1.26 mM. Cwht1p crystals were grown and subjected to X-ray irradiation, giving a complete diffraction dataset to 2.04 Å resolution. Work is ongoing to obtain phases so that we may further understand this fundamental member of the N-glycosylation pathway through the discovery of its molecular structure.


Subject(s)
Pichia/genetics , Saccharomyces cerevisiae/enzymology , alpha-Glucosidases/chemistry , alpha-Glucosidases/genetics , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Models, Molecular , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , alpha-Glucosidases/isolation & purification , alpha-Glucosidases/metabolism
16.
Bioorg Med Chem Lett ; 21(21): 6491-4, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21924903

ABSTRACT

Human maltase glucoamylase (MGAM) and sucrase isomaltase (SI) are two human intestinal glucosidases responsible for the final step of starch hydrolysis. MGAM and SI are anchored to the small intestinal brush border epithelial cells and contain two catalytic N-terminal and C-terminal subunits. In this study, we report the inhibition profile of 3'-O-methylponkoranol for the individual recombinant N and C terminal enzymes and compare the inhibitory activities of this compound with de-O-sulfonated ponkoranol. We show that 3'-O-methylponkoranol inhibits the different subunits to different extents, with extraordinary selectivity for C-terminal SI (K(i)=7±2nM). The enzymes themselves could serve as therapeutic targets for the treatment of digestive disorders or their sequelae.


Subject(s)
Enzyme Inhibitors/pharmacology , Gastrointestinal Diseases/drug therapy , Glycoside Hydrolase Inhibitors , Sucrase-Isomaltase Complex/antagonists & inhibitors , Thiophenes/pharmacology , Amylopectin/chemistry , Amylose/chemistry , Carbohydrate Sequence , Enzyme Inhibitors/therapeutic use , Humans , Molecular Sequence Data , Thiophenes/therapeutic use
17.
Bioorg Med Chem ; 19(13): 3929-34, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21669536

ABSTRACT

Inhibition of intestinal α-glucosidases and pancreatic α-amylases is an approach to controlling blood glucose and serum insulin levels in individuals with Type II diabetes. The two human intestinal glucosidases are maltase-glucoamylase and sucrase-isomaltase. Each incorporates two family 31 glycoside hydrolases responsible for the final step of starch hydrolysis. Here we compare the inhibition profiles of the individual N- and C-terminal catalytic subunits of both glucosidases by clinical glucosidase inhibitors, acarbose and miglitol, and newly discovered glucosidase inhibitors from an Ayurvedic remedy used for the treatment of Type II diabetes. We show that features of the compounds introduce selectivity towards the subunits. Together with structural data, the results enhance the understanding of the role of each catalytic subunit in starch digestion, helping to guide the development of new compounds with subunit specific antidiabetic activity. The results may also have relevance to other metabolic diseases such as obesity and cardiovascular disease.


Subject(s)
Starch/metabolism , Sucrase-Isomaltase Complex/metabolism , alpha-Glucosidases/metabolism , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/pharmacology , Acarbose/chemistry , Acarbose/pharmacology , Catalytic Domain , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glycoside Hydrolase Inhibitors , Kinetics , Monosaccharides/chemistry , Selenium Compounds/chemistry , Selenium Compounds/pharmacology , Sucrase-Isomaltase Complex/antagonists & inhibitors , Sugar Alcohols/chemistry , Sugar Alcohols/pharmacology , Sulfates/chemistry , Sulfates/pharmacology
18.
Proc Natl Acad Sci U S A ; 105(28): 9570-5, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18599462

ABSTRACT

Golgi alpha-mannosidase II (GMII) is a key glycosyl hydrolase in the N-linked glycosylation pathway. It catalyzes the removal of two different mannosyl linkages of GlcNAcMan(5)GlcNAc(2), which is the committed step in complex N-glycan synthesis. Inhibition of this enzyme has shown promise in certain cancers in both laboratory and clinical settings. Here we present the high-resolution crystal structure of a nucleophile mutant of Drosophila melanogaster GMII (dGMII) bound to its natural oligosaccharide substrate and an oligosaccharide precursor as well as the structure of the unliganded mutant. These structures allow us to identify three sugar-binding subsites within the larger active site cleft. Our results allow for the formulation of the complete catalytic process of dGMII, which involves a specific order of bond cleavage, and a major substrate rearrangement in the active site. This process is likely conserved for all GMII enzymes-but not in the structurally related lysosomal mannosidase-and will form the basis for the design of specific inhibitors against GMII.


Subject(s)
Drosophila Proteins/chemistry , Mannosidases/chemistry , Animals , Carbohydrates/chemistry , Catalytic Domain , Crystallography, X-Ray , Drosophila Proteins/metabolism , Drosophila melanogaster , Mannosidases/genetics , Mannosidases/metabolism , Mutation , Protein Binding
19.
Biochemistry ; 49(3): 443-51, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20039683

ABSTRACT

An approach to controlling blood glucose levels in individuals with type 2 diabetes is to target alpha-amylases and intestinal glucosidases using alpha-glucosidase inhibitors acarbose and miglitol. One of the intestinal glucosidases targeted is the N-terminal catalytic domain of maltase-glucoamylase (ntMGAM), one of the four intestinal glycoside hydrolase 31 enzyme activities responsible for the hydrolysis of terminal starch products into glucose. Here we present the X-ray crystallographic studies of ntMGAM in complex with a new class of alpha-glucosidase inhibitors derived from natural extracts of Salacia reticulata, a plant used traditionally in Ayuverdic medicine for the treatment of type 2 diabetes. Included in these extracts are the active compounds salacinol, kotalanol, and de-O-sulfonated kotalanol. This study reveals that de-O-sulfonated kotalanol is the most potent ntMGAM inhibitor reported to date (K(i) = 0.03 microM), some 2000-fold better than the compounds currently used in the clinic, and highlights the potential of the salacinol class of inhibitors as future drug candidates.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Enzyme Inhibitors/chemistry , Glycoside Hydrolase Inhibitors , Hypoglycemic Agents/chemistry , Salacia/chemistry , alpha-Glucosidases/chemistry , Acarbose/chemistry , Binding Sites , Crystallography, X-Ray , Diabetes Mellitus, Type 2/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Kinetics , Medicine, Ayurvedic , Plant Extracts/chemistry , Structure-Activity Relationship , Sugar Alcohols/chemistry , Sulfates/chemistry , alpha-Glucosidases/metabolism
20.
Chembiochem ; 11(5): 673-80, 2010 Mar 22.
Article in English | MEDLINE | ID: mdl-20209559

ABSTRACT

Golgi alpha-mannosidase II (GMII) is a key enzyme in the N-glycosylation pathway and is a potential target for cancer chemotherapy. The natural product swainsonine is a potent inhibitor of GMII. In this paper we characterize the binding of 5alpha-substituted swainsonine analogues to the soluble catalytic domain of Drosophila GMII by X-ray crystallography. These inhibitors enjoy an advantage over previously reported GMII inhibitors in that they did not significantly decrease the inhibitory potential of the swainsonine head-group. The phenyl groups of these analogues occupy a portion of the binding site not previously seen to be populated with either substrate analogues or other inhibitors and they form novel hydrophobic interactions. They displace a well-organized water cluster, but the presence of a C(10) carbonyl allows the reestablishment of important hydrogen bonds. Already approximately tenfold more active against the Golgi enzyme than the lysosomal enzyme, these inhibitors offer the potential of being extended into the N-acetylglucosamine binding site of GMII for the creation of even more potent and selective GMII inhibitors.


Subject(s)
Antineoplastic Agents/chemistry , Golgi Apparatus/enzymology , Mannosidases/antagonists & inhibitors , Swainsonine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Binding Sites , Catalytic Domain , Drosophila melanogaster/enzymology , Mannosidases/metabolism , Protein Binding , Swainsonine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL