Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters










Publication year range
2.
Bioorg Med Chem Lett ; 40: 127979, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33766763

ABSTRACT

α-Glucosidase inhibition is a valid approach for controlling hyperglycemia in diabetes. In the current study, new molecules as a hybrid of isoxazole and dibenzazepine scaffolds were designed, based on their literature as antidiabetic agents. For this, a series of dibenzazepine-linked isoxazoles (33-54) was prepared using Nitrile oxide-Alkyne cycloaddition (NOAC) reaction, and evaluated for their α-glucosidase inhibitory activities to explore new hits for treatment of diabetes. Most of the compounds showed potent inhibitory potency against α-glucosidase (EC 3.2.1.20) enzyme (IC50 = 35.62 ± 1.48 to 333.30 ± 1.67 µM) using acarbose as a reference drug (IC50 = 875.75 ± 2.08 µM). Structure-activity relationship, kinetics and molecular docking studies of active isoxazoles were also determined to study enzyme-inhibitor interactions. Compounds 33, 40, 41, 46, 48-50, and 54 showed binding interactions with critical amino acid residues of α-glucosidase enzyme, such as Lys156, Ser157, Asp242, and Gln353.


Subject(s)
Dibenzazepines/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Hypoglycemic Agents/chemistry , Isoxazoles/chemistry , 3T3 Cells , Animals , Cycloaddition Reaction , Dibenzazepines/chemical synthesis , Dibenzazepines/toxicity , Enzyme Assays , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/toxicity , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/toxicity , Isoxazoles/chemical synthesis , Isoxazoles/toxicity , Kinetics , Mice , Molecular Docking Simulation , Molecular Structure , Oligo-1,6-Glucosidase/metabolism , Protein Binding , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
3.
Nutrients ; 13(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33375084

ABSTRACT

Congenital sucrase-isomaltase deficiency (CSID) is a rare metabolic intestinal disorder with reduced or absent activity levels of sucrase-isomaltase (SI). Interestingly, the main symptoms of CSID overlap with those in irritable bowel syndrome (IBS), a common functional gastrointestinal disorder with unknown etiology. Recent advances in genetic screening of IBS patients have revealed rare SI gene variants that are associated with IBS. Here, we investigated the biochemical, cellular and functional phenotypes of several of these variants. The data demonstrate that the SI mutants can be categorized into three groups including immature, mature but slowly transported, and finally mature and properly transported but with reduced enzymatic activity. We also identified SI mutant phenotypes that are deficient but generally not as severe as those characterized in CSID patients. The variable effects on the trafficking and function of the mutations analyzed in this study support the view that both CSID and IBS are heterogeneous disorders, the severity of which is likely related to the biochemical phenotypes of the SI mutants as well as the environment and diet of patients. Our study underlines the necessity to screen for SI mutations in IBS patients and to consider enzyme replacement therapy as an appropriate therapy as in CSID.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/metabolism , Irritable Bowel Syndrome/genetics , Irritable Bowel Syndrome/metabolism , Mutation , Protein Transport , Sucrase-Isomaltase Complex/deficiency , Animals , COS Cells , Chlorocebus aethiops , Oligo-1,6-Glucosidase/genetics , Oligo-1,6-Glucosidase/metabolism , Phenotype , Sucrase-Isomaltase Complex/genetics , Sucrase-Isomaltase Complex/metabolism
4.
Sci Rep ; 10(1): 15178, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938976

ABSTRACT

We have previously discovered that heated honey but not unheated honey could induce the secretion of granulocyte-colony stimulating factor (G-CSF) in the MCE301 intestinal epithelial cells. The objective of this study was to identify compounds in honey that could contribute to this activity. We bought several kinds of commercial honey samples derived from different flowers, as well as corn syrup samples, in the markets of China and Japan, and heated them at 180 °C for 30 min. MCE301 cells were treated with the medium containing the samples, and G-CSF levels in the medium were measured by ELISA. By comparing their activities and sugar contents, we discovered that isomaltose was primarily implicated. The optimum heating conditions for isomaltose were at 180 °C for 60 min or at 200 °C for 15-30 min, and these time- and temperature-dependencies were similar to those of honey in our previous study. When heated isomaltose was partitioned by dialysis, the active ingredients were transferred into a high-molecular-weight fraction. By size-exclusion HPLC analysis, the average molecular weight of heated isomaltose was 790 kDa. When heated isomaltose was hydrolyzed by acids, glucose was subsequently produced. Maltose, sucrose, turanose, and trehalose did not exhibited any activity when heated at 180 °C for 60 min, indicating that the glucose groups with α(1 → 6)-binding in the isomaltose molecule play important roles in its activity when oxidatively polymerized by heat. The stimulating activity of heated isomaltose was inhibited by toll-like receptor 4 (TLR4) inhibitor, suggesting that heated isomaltose activates TLR4 to induce G-CSF. Since G-CSF is clinically used for cancer patients to accelerate their recovery from neutropenia following chemotherapy or accompanied with aplastic anemia, these findings indicate that honey which contains high level of isomaltose could improve immunosuppressive conditions when honey is heated, and that heated isomaltose might be of potential therapeutic use in patients with compromised immunity caused by chemotherapeutic agents.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , Honey , Intestinal Mucosa/metabolism , Neutropenia/therapy , Neutrophils/metabolism , Oligo-1,6-Glucosidase/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Granulocyte Colony-Stimulating Factor/therapeutic use , Heating , Mice , Neutrophils/pathology , Polymerization
5.
Int J Food Sci Nutr ; 71(5): 572-580, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31976784

ABSTRACT

Resistant starch (RS) consumption has beneficial effects on health, such as reduced postprandial blood glucose levels. In this study, we evaluated the effect of a 14-day diet containing RS on α-glucosidase activity and the expression of genes related to carbohydrate digestion/absorption in rats. We examined whether the effects of RS persist when the rats were shifted to a control diet. The results suggest that RS consumption reduces α-glucosidase activity and Mgam, Si and Sglt1 mRNA levels in the proximal jejunum. In addition, RS consumption appeared to influence the serum GIP level, up to 2 days after the animals were shifted to a control diet. To our knowledge, this is the first report that RS has a sustained effect on gut hormone expression and the expression of genes related to carbohydrate digestion/absorption in the proximal jejunum.


Subject(s)
Carbohydrate Metabolism/drug effects , Digestion , Gastric Inhibitory Polypeptide/blood , Intestinal Absorption , Intestine, Small/drug effects , Resistant Starch/pharmacology , alpha-Glucosidases/metabolism , Animals , Carbohydrate Metabolism/genetics , Diet , Feeding Behavior , Gastric Inhibitory Polypeptide/genetics , Intestine, Small/metabolism , Jejunum/drug effects , Jejunum/metabolism , Male , Oligo-1,6-Glucosidase/genetics , Oligo-1,6-Glucosidase/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Sucrase/genetics , Sucrase/metabolism , alpha-Glucosidases/genetics
6.
Sci Rep ; 9(1): 13908, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31558734

ABSTRACT

Recently, wild strains of Saccharomyces cerevisiae isolated from a variety of natural resources have been used to make bread, beer, wine, and sake. In the current study, we isolated wild S. cerevisiae MC strain from the carnation (Dianthus caryophyllus L) flower and produced sake using its cerulenin-resistant mutant strain MC87-46. Then, we characterized the components, including ethanol, amino acids, organic acids, and sugars, in the fermented sake. Sake brewed with MC87-46 is sweet owing to the high content of isomaltose, which was at a concentration of 44.3 mM. The low sake meter value of -19.6 is most likely due to this high isomaltose concentration. The genomic DNA of MC87-46 encodes for isomaltases IMA1, IMA2, IMA3, IMA4 and IMA5, as well as the isomaltose transporter gene, AGT1. However, these genes were not induced in MC87-46 by isomaltose, and the strain did not possess isomaltase activity. These results show that MC87-46 cannot utilize isomaltose, resulting in its accumulation in the fermented sake. Isomaltose concentrations in sake brewed with MC87-46 were 24.6-fold more than in commercial sake. These findings suggest that MC87-46 may be useful for commercial application in Japanese sake production because of its unique flavour and nutrient profile.


Subject(s)
Alcoholic Beverages/standards , Fermentation , Isomaltose/metabolism , Saccharomyces cerevisiae/metabolism , Dianthus/microbiology , Industrial Microbiology/methods , Oligo-1,6-Glucosidase/genetics , Oligo-1,6-Glucosidase/metabolism , Saccharomyces cerevisiae/pathogenicity , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
7.
Biochem Biophys Res Commun ; 483(1): 115-121, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28042032

ABSTRACT

Among members of the glycoside hydrolase (GH) family, sucrose isomerase (SIase) and oligo-1,6-glucosidase (O16G) are evolutionarily closely related even though their activities show different specificities. A gene (Avin_08330) encoding a putative SIase (AZOG: Azotobacterglucocosidase) from the nitrogen-fixing bacterium Azotobacter vinelandii is a type of pseudo-SIase harboring the "RLDRD" motif, a SIase-specific region in 329-333. However, neither sucrose isomerization nor hydrolysis activities were observed in recombinant AZOG (rAZOG). The rAZOG showed similar substrate specificity to Bacillus O16G as it catalyzes the hydrolysis of isomaltulose and isomaltose, which contain α-1,6-glycosidic linkages. Interestingly, rAZOG could generate isomaltose from the small substrate methyl-α-glucoside (MαG) via intermolecular transglycosylation. In addition, sucrose isomers isomaltulose and trehalulose were produced when 250 mM fructose was added to the MαG reaction mixture. The conserved regions I and II of AZOG are shared with many O16Gs, while regions III and IV are very similar to those of SIases. Strikingly, a shuffled AZOG, in which the N-terminal region of SIase containing conserved regions I and II was exchanged with the original enzyme, exhibited a production of sucrose isomers. This study demonstrates an evolutionary relationship between SIase and O16G and suggests some of the main regions that determine the specificity of SIase and O16G.


Subject(s)
Azotobacter vinelandii/enzymology , Bacterial Proteins/metabolism , Glucosyltransferases/metabolism , Amino Acid Motifs , Azotobacter vinelandii/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biotechnology , Catalytic Domain , Conserved Sequence , Disaccharides/metabolism , Evolution, Molecular , Genes, Bacterial , Genetic Variation , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Isomaltose/analogs & derivatives , Isomaltose/metabolism , Models, Molecular , Oligo-1,6-Glucosidase/chemistry , Oligo-1,6-Glucosidase/genetics , Oligo-1,6-Glucosidase/metabolism , Phylogeny , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Sucrose/metabolism
8.
J Nutr ; 145(6): 1147-55, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25878206

ABSTRACT

BACKGROUND: The proportion of starch disappearing from the small intestinal lumen is generally lower in ruminants than in monogastric animals, and there are indications that the starch digestion capacity in ruminants is limited. OBJECTIVES: Milk-fed calves were used to study the rate-limiting enzyme in starch hydrolysis and to quantify starch fermentation in ruminants. METHODS: Forty male Holstein-Friesian calves were fed milk replacer containing either lactose (control) or 1 of 4 corn starch products. The following starch products differed in the enzyme ratios required for their complete hydrolysis to glucose: gelatinized starch [α-amylase and (iso)maltase], maltodextrin [(iso)maltase and α-amylase], maltodextrin with α-1,6-branching (isomaltase, maltase, and α-amylase), and maltose (maltase). In the adaptation period, calves were stepwise exposed to an increasing dose of the starch product for 14 wk to allow maximal adaptation of all enzyme systems involved. In the experimental period, apparent total tract and ileal starch product disappearance, total tract starch product fermentation, and α-amylase, maltase, and isomaltase activities were determined at 18% inclusion of the starch product. RESULTS: Maltase and isomaltase activities in the brush border did not increase for any of the starch product treatments. Luminal α-amylase activity was lower in the proximal (3.9 ± 3.2 and 2.7 ± 1.7 U/mg Co for control and starch product calves, respectively) but greater in the distal small intestine of starch-fed calves than in control calves (0.0 ± 0.0 and 6.4 ± 1.5 U/mg Co for control and starch product calves, respectively; means ± SEs for control and means ± pooled SEMs for starch product treatments). Apparent ileal (61.6% ± 6.3%) and total tract (99.1% ± 0.4%) starch product disappearance did not differ between starch product treatments, suggesting that maltase activity limits starch digestion in ruminants. Total tract starch product fermentation averaged 414 ± 43 g/d, corresponding to 89% of intake, of which half was fermented before the terminal ileum, regardless of starch product treatment. CONCLUSION: Fermentation, rather than enzymatic digestion, is the main reason for small intestinal starch disappearance in milk-fed calves.


Subject(s)
Fermentation , Intestine, Small/enzymology , Intestine, Small/metabolism , Lactose/metabolism , Starch/metabolism , Animal Feed , Animals , Blood Glucose/metabolism , Cattle , Digestion , Glucose/metabolism , Male , Oligo-1,6-Glucosidase/metabolism , Polysaccharides/metabolism , Zea mays/chemistry , alpha-Amylases/metabolism , alpha-Glucosidases/metabolism
9.
J Agric Food Chem ; 63(15): 3873-9, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25816913

ABSTRACT

In this study, it was hypothesized that dietary phenolic compounds selectively inhibit the individual C- and N-terminal (Ct, Nt) subunits of the two small intestinal α-glucosidases, maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI), for a modulated glycemic carbohydrate digestion. The inhibition by chlorogenic acid, caffeic acid, gallic acid, (+)-catechin, and (-)-epigallocatechin gallate (EGCG) on individual recombinant human Nt-MGAM and Nt-SI and on mouse Ct-MGAM and Ct-SI was assayed using maltose as the substrate. Inhibition constants, inhibition mechanisms, and IC50 values for each combination of phenolic compound and enzymatic subunit were determined. EGCG and chlorogenic acid were found to be more potent inhibitors for selectively inhibiting the two subunits with highest activity, Ct-MGAM and Ct-SI. All compounds displayed noncompetitive type inhibition. Inhibition of fast-digesting Ct-MGAM and Ct-SI by EGCG and chlorogenic acid could lead to a slow, but complete, digestion of starch for improved glycemic response of starchy foods with potential health benefit.


Subject(s)
Enzyme Inhibitors/chemistry , Glucan 1,4-alpha-Glucosidase/chemistry , Glucose/chemistry , Oligo-1,6-Glucosidase/chemistry , Phenol/chemistry , Sucrase/chemistry , alpha-Glucosidases/chemistry , Animals , Digestion , Enzyme Inhibitors/metabolism , Glucan 1,4-alpha-Glucosidase/metabolism , Humans , Kinetics , Mice , Oligo-1,6-Glucosidase/metabolism , Phenol/metabolism , Sucrase/metabolism , alpha-Glucosidases/metabolism
10.
Drug Metab Pharmacokinet ; 29(1): 44-51, 2014.
Article in English | MEDLINE | ID: mdl-23822979

ABSTRACT

  Human induced pluripotent stem (iPS) cells were differentiated into the endoderm using activin A and were then treated with fibroblast growth factor 2 (FGF2) for differentiation into intestinal stem cell-like cells. These immature cells were then differentiated into enterocyte-like cells using epidermal growth factor (EGF) in 2% fetal bovine serum (FBS). At the early stage of differentiation, mRNA expression of caudal type homeobox 2 (CDX2), a major transcription factor related to intestinal development and differentiation, and leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5), an intestinal stem cell marker, was markedly increased by treatment with FGF2. When cells were cultured in medium containing EGF and a low concentration of FBS, mRNAs of specific markers of intestinal epithelial cells, including sucrase-isomaltase, the intestinal oligopeptide transporter SLC15A1/peptide transporter 1 (PEPT1), and the major metabolizing enzyme CYP3A4, were expressed. In addition, sucrase-isomaltase protein expression and uptake of ß-Ala-Lys-N-7-amino-4-methylcoumarin-3-acetic acid (ß-Ala-Lys-AMCA), a fluorescence-labeled substrate of the oligopeptide transporter, were detected. These results demonstrate a simple and direct method for differentiating human iPS cells into functional enterocyte-like cells.


Subject(s)
Erythrocytes/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Biomarkers/metabolism , Cattle , Cell Differentiation , Cell Line , Coumarins/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Dipeptides/metabolism , Erythrocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Oligo-1,6-Glucosidase/genetics , Oligo-1,6-Glucosidase/metabolism , Peptide Transporter 1 , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Sucrase/genetics , Sucrase/metabolism , Symporters/genetics , Symporters/metabolism
11.
J Biol Chem ; 287(27): 23104-18, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22584580

ABSTRACT

Porcine pancreatic α-amylase (PPA) binds to N-linked glycans of glycoproteins (Matsushita, H., Takenaka, M., and Ogawa, H. (2002) J. Biol Chem., 277, 4680-4686). Immunostaining revealed that PPA is located at the brush-border membrane (BBM) of enterocytes in the duodenum and that the binding is inhibited by mannan but not galactan, indicating that PPA binds carbohydrate-specifically to BBM. The ligands for PPA in BBM were identified as glycoprotein N-glycans that are significantly involved in the assimilation of glucose, including sucrase-isomaltase (SI) and Na(+)/Glc cotransporter 1 (SGLT1). Binding of SI and SGLT1 in BBM to PPA was dose-dependent and inhibited by mannan. Using BBM vesicles, we found functional changes in PPA and its ligands in BBM due to the N-glycan-specific interaction. The starch-degrading activity of PPA and maltose-degrading activity of SI were enhanced to 240 and 175%, respectively, while Glc uptake by SGLT1 was markedly inhibited by PPA at high but physiologically possible concentrations, and the binding was attenuated by the addition of mannose-specific lectins, especially from Galanthus nivalis. Additionally, recombinant human pancreatic α-amylases expressed in yeast and purified by single-step affinity chromatography exhibited the same carbohydrate binding specificity as PPA in binding assays with sugar-biotinyl polymer probes. The results indicate that mammalian pancreatic α-amylases share a common carbohydrate binding activity and specifically bind to the intestinal BBM. Interaction with N-glycans in the BBM activated PPA and SI to produce much Glc on the one hand and to inhibit Glc absorption by enterocytes via SGLT1 in order to prevent a rapid increase in blood sugar on the other.


Subject(s)
Duodenum/metabolism , Glycoproteins/metabolism , Microvilli/metabolism , Pancreatic alpha-Amylases/metabolism , Polysaccharides/metabolism , Animals , Blood Glucose/metabolism , Duodenum/cytology , Enterocytes/enzymology , Galactans/metabolism , Glycomics/methods , Glycoproteins/isolation & purification , Glycoside Hydrolases/metabolism , Glycosylation , Homeostasis/physiology , Humans , Lectins/metabolism , Ligands , Mannans/metabolism , Oligo-1,6-Glucosidase/metabolism , Pancreatic alpha-Amylases/pharmacology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sodium-Glucose Transporter 1/metabolism , Starch/metabolism , Sucrase/metabolism , Swine
12.
J Biosci Bioeng ; 112(6): 545-50, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21925939

ABSTRACT

The structures of the E277A isomaltase mutant from Saccharomyces cerevisiae in complex with isomaltose or maltose were determined at resolutions of 1.80 and 1.40Å, respectively. The root mean square deviations between the corresponding main-chain atoms of free isomaltase and the E277Α-isomaltose complex structures and those of free isomaltase and the E277A-maltose complex structures were found to be 0.131Å and 0.083Å, respectively. Thus, the amino acid substitution and ligand binding do not affect the overall structure of isomaltase. In the E277A-isomaltose structure, the bound isomaltose was readily identified by electron densities in the active site pocket; however, the reducing end of maltose was not observed in the E277A-maltose structure. The superposition of maltose onto the E277A-maltose structure revealed that the reducing end of maltose cannot bind to the subsite +1 due to the steric hindrance from Val216 and Gln279. The amino acid sequence comparisons with α-glucosidases showed that a bulky hydrophobic amino acid residue is conserved at the position of Val216 in α-1,6-glucosidic linkage hydrolyzing enzymes. Similarly, a bulky amino acid residue is conserved at the position of Gln279 in α-1,6-glucosidic linkage-only hydrolyzing α-glucosidases. Ala, Gly, or Asn residues were located at the position of α-1,4-glucosidic linkage hydrolyzing α-glucosidases. Two isomaltase mutant enzymes - V216T and Q279A - hydrolyzed maltose. Thus, the amino acid residues at these positions may be largely responsible for determining the substrate specificity of α-glucosidases.


Subject(s)
Maltose/chemistry , Oligo-1,6-Glucosidase/chemistry , Saccharomyces cerevisiae/enzymology , Amino Acid Substitution , Catalytic Domain , Crystallography, X-Ray , Kinetics , Maltose/metabolism , Models, Chemical , Oligo-1,6-Glucosidase/genetics , Oligo-1,6-Glucosidase/metabolism , Substrate Specificity , alpha-Glucosidases/chemistry , alpha-Glucosidases/metabolism
13.
J Pediatr Gastroenterol Nutr ; 45(1): 32-43, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17592362

ABSTRACT

BACKGROUND: Starches are the major source of dietary glucose in weaned children and adults. However, small intestine alpha-glucogenesis by starch digestion is poorly understood due to substrate structural and chemical complexity, as well as the multiplicity of participating enzymes. Our objective was dissection of luminal and mucosal alpha-glucosidase activities participating in digestion of the soluble starch product maltodextrin (MDx). PATIENTS AND METHODS: Immunoprecipitated assays were performed on biopsy specimens and isolated enterocytes with MDx substrate. RESULTS: Mucosal sucrase-isomaltase (SI) and maltase-glucoamylase (MGAM) contributed 85% of total in vitro alpha-glucogenesis. Recombinant human pancreatic alpha-amylase alone contributed <15% of in vitro alpha-glucogenesis; however, alpha-amylase strongly amplified the mucosal alpha-glucogenic activities by preprocessing of starch to short glucose oligomer substrates. At low glucose oligomer concentrations, MGAM was 10 times more active than SI, but at higher concentrations it experienced substrate inhibition whereas SI was not affected. The in vitro results indicated that MGAM activity is inhibited by alpha-amylase digested starch product "brake" and contributes only 20% of mucosal alpha-glucogenic activity. SI contributes most of the alpha-glucogenic activity at higher oligomer substrate concentrations. CONCLUSIONS: MGAM primes and SI activity sustains and constrains prandial alpha-glucogenesis from starch oligomers at approximately 5% of the uninhibited rate. This coupled mucosal mechanism may contribute to highly efficient glucogenesis from low-starch diets and play a role in meeting the high requirement for glucose during children's brain maturation. The brake could play a constraining role on rates of glucose production from higher-starch diets consumed by an older population at risk for degenerative metabolic disorders.


Subject(s)
Enterocytes/metabolism , Glucan 1,4-alpha-Glucosidase/metabolism , Glucose/metabolism , Polysaccharides/metabolism , alpha-Glucosidases/metabolism , Animals , Biopsy , Child , Digestion , Duodenum/enzymology , Enterocytes/enzymology , Humans , Immunoprecipitation , Intestinal Mucosa/enzymology , Mice , Oligo-1,6-Glucosidase/metabolism , Starch/metabolism
14.
Biochem Biophys Res Commun ; 342(1): 66-72, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16480684

ABSTRACT

The homeobox gene Cdx2 is involved in the regulation of the expression of intestine specific markers such as sucrase-isomaltase and lactase-phlorizin hydrolase. Previous studies performed with immortalized or transformed intestinal cell lines have provided evidence that Cdx2 can promote morphological and functional differentiation in these experimental models. However, no data exist concerning the implication of this factor in normal human intestinal cell physiology. In the present work, we have investigated the role of Cdx2 in normal human intestinal epithelial crypt (HIEC) cells that lack this transcription factor. The establishment of HIEC cells expressing Cdx2 in an inducible manner shows that forced expression of Cdx2 significantly alters the proliferation of intestinal crypt cells and stimulates dipeptidylpeptidase IV expression but is not sufficient to trigger intestinal terminal differentiation. These observations suggest that Cdx2 requires additional factors to activate the enterocyte differentiation program in normal undifferentiated cells.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Homeodomain Proteins/metabolism , Intestinal Mucosa/metabolism , Intestines/cytology , Biomarkers , CDX2 Transcription Factor , Cell Differentiation , Cell Line , Cell Proliferation , Dipeptidyl Peptidase 4/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Oligo-1,6-Glucosidase/metabolism , Sucrose/metabolism
15.
J Biochem ; 134(4): 543-50, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14607981

ABSTRACT

The alpha-glucosidase of Bacillus sp. strain SAM1606 is a member of glycosyl hydrolase family 13, and shows an extraordinarily broad substrate specificity and is one of very few alpha-glucosidases that can efficiently hydrolyze the alpha-1,1-glucosidic linkage of alpha,alpha'-trehalose (trehalose). Phylogenetic analysis of family-13 enzymes suggests that SAM1606 alpha-glucosidase may be evolutionally derived from an alpha-1,6-specific ancestor, oligo-1,6-glucosidase (O16G). Indeed, replacement of Pro(273*) and Thr(342*) of B. cereus O16G by glycine and asparagine (the corresponding residues in the SAM1606 enzyme), respectively, was found to cause 192-fold enhancement of the relative catalytic efficiency for trehalose, suggesting that O16G may easily "evolved" into an enzyme with an extended substrate specificity by substitution of a limited number of amino acids, including that at position 273* (an asterisk indicates the amino-acid numbering of the SAM1606 sequence). To probe the role of the amino acid at position 273* of alpha-glucosidase in determination of the substrate specificity, the amino acid at position 273 of SAM1606 alpha-glucosidase was replaced by all other naturally occurring amino acids, and the resultant mutants were kinetically characterized. The results showed that substitution of bulky residues (e.g., isoleucine and methionine) for glycine at this position resulted in large increases in the K(m) values for trehalose and maltose, whereas the affinity to isomaltose was only minimally affected by such an amino-acid substitution at this position. Three-dimensional structural models of the enzyme-substrate complexes of the wild-type and mutant SAM1606 alpha-glucosidases were built to explore the mechanism responsible for these observations. It is proposed that substitution by glycine at position 273* could eliminate steric hindrance around subsite +1 that originally occurred in parental O16G and is, at least in part, responsible for the acquired broad substrate specificity of SAM1606 alpha-glucosidase.


Subject(s)
Bacillus/enzymology , alpha-Glucosidases/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Asparagine/chemistry , Bacillus/metabolism , Binding Sites , Catalysis , Evolution, Molecular , Glycine/chemistry , Isomaltose/chemistry , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , Oligo-1,6-Glucosidase/metabolism , Phylogeny , Plasmids/metabolism , Proline/chemistry , Serine/chemistry , Substrate Specificity , Threonine/chemistry , Trehalose/chemistry , alpha-Glucosidases/chemistry
16.
J Cell Biochem ; 86(3): 440-50, 2002.
Article in English | MEDLINE | ID: mdl-12210751

ABSTRACT

Rat small intestinal epithelial cells and human colon adenocarcinoma cells cultured on Matrigel expressed the differentiation specific enzyme, sucrase-isomaltase, as determined by indirect immunofluorescence. Rat small intestinal epithelial cells, rat colonocytes, and human colon adenocarcinoma cells developed an altered morphology when cultured on Matrigel and became apoptotic within 24-48 h. Benzo[a]pyrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin caused a 2- and 5-fold induction, respectively, of ethoxyresorufin-o-deethylase activity in rat small intestinal epithelial cells cultured on Matrigel. Benzo[a]pyrene- or 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced ethoxyresorufin-o-deethylase activity in rat small intestinal epithelial cells cultured on plastic was not detected. 2,3,7,8-tetrachlorodibenzo-p-dioxin treatment caused a 14-fold induction of transfected, rat CYP1A1-promoter-luciferase activity in rat small intestinal epithelial cells cultured on Matrigel. Benzo[a]pyrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin treatment induced ethoxyresorufin-o-deethylase activity by 6- and 1.6-fold, respectively in rat colonocytes cultured on Matrigel. Induction of ethoxyresorufin-o-deethylase activity was not observed in rat colonocytes cultured on plastic. CYP1A1-promoter-luciferase activity was induced 3-fold by 2,3,7,8-tetrachlorodibenzo-p-dioxin in rat colonocytes cultured on Matrigel. Induction of CYP1A1-promoter-luciferase activity in rat small intestinal epithelial cells or rat colonocytes cultured on plastic was not observed. Ethoxyresorufin-o-deethylase activity in human colon adenocarcinoma cells, cultured on either plastic or Matrigel, was induced 7-fold by benzo[a]pyrene. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced ethoxyresorufin-o-deethylase activity was 2-fold greater in human colon adenocarcinoma cells cultured on Matrigel compared to cells cultured on plastic. Extracellular matrix-mediated differentiation and apoptosis of intestinal cells provide in vitro systems for study of the regulation of CYP1A1 expression, carcinogen activation in the gut and mechanism(s) of apoptosis of colon cancer cells.


Subject(s)
Apoptosis , Basement Membrane/enzymology , Colon/enzymology , Colonic Neoplasms/enzymology , Cytochrome P-450 CYP1A1/metabolism , Intestinal Mucosa/enzymology , Intestine, Small/enzymology , Animals , Basement Membrane/cytology , Benzo(a)pyrene/pharmacology , Caco-2 Cells , Cell Culture Techniques , Cell Differentiation , Cell Size , Collagen , Colon/cytology , Colon/drug effects , Colon/pathology , Colonic Neoplasms/pathology , Cytochrome P-450 CYP1A1/chemistry , Cytochrome P-450 CYP1A1/genetics , Drug Combinations , Enzyme Induction/drug effects , Humans , Intestinal Mucosa/cytology , Intestine, Small/cytology , Laminin , Molecular Weight , Oligo-1,6-Glucosidase/metabolism , Plastics , Promoter Regions, Genetic/genetics , Proteoglycans , Rats , Sucrase/metabolism , Time Factors , Tumor Cells, Cultured
17.
J Mol Biol ; 321(1): 149-62, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12139940

ABSTRACT

4-alpha-Glucanotransferase (GTase) is an essential enzyme in alpha-1,4-glucan metabolism in bacteria and plants. It catalyses the transfer of maltooligosaccharides from an 1,4-alpha-D-glucan molecule to the 4-hydroxyl group of an acceptor sugar molecule. The crystal structures of Thermotoga maritima GTase and its complex with the inhibitor acarbose have been determined at 2.6A and 2.5A resolution, respectively. The GTase structure consists of three domains, an N-terminal domain with the (beta/alpha)(8) barrel topology (domain A), a 65 residue domain, domain B, inserted between strand beta3 and helix alpha6 of the barrel, and a C-terminal domain, domain C, which forms an antiparallel beta-structure. Analysis of the complex of GTase with acarbose has revealed the locations of five sugar-binding subsites (-2 to +3) in the active-site cleft lying between domain B and the C-terminal end of the (beta/alpha)(8) barrel. The structure of GTase closely resembles the family 13 glycoside hydrolases and conservation of key catalytic residues previously identified for this family is consistent with a double-displacement catalytic mechanism for this enzyme. A distinguishing feature of GTase is a pair of tryptophan residues, W131 and W218, which, upon the carbohydrate inhibitor binding, form a remarkable aromatic "clamp" that captures the sugar rings at the acceptor-binding sites +1 and +2. Analysis of the structure of the complex shows that sugar residues occupying subsites from -2 to +2 engage in extensive interactions with the protein, whereas the +3 glucosyl residue makes relatively few contacts with the enzyme. Thus, the structure suggests that four subsites, from -2 to +2, play the dominant role in enzyme-substrate recognition, consistent with the observation that the smallest donor for T.maritima GTase is maltotetraose, the smallest chain transferred is a maltosyl unit and that the smallest residual fragment after transfer is maltose. A close similarity between the structures of GTase and oligo-1,6-glucosidase has allowed the structural features that determine differences in substrate specificity of these two enzymes to be analysed.


Subject(s)
Acarbose/metabolism , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/metabolism , Thermotoga maritima/enzymology , Acarbose/chemistry , Acarbose/pharmacology , Binding Sites , Calcium/metabolism , Carbohydrate Sequence , Catalysis , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Glycogen Debranching Enzyme System/antagonists & inhibitors , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Oligo-1,6-Glucosidase/chemistry , Oligo-1,6-Glucosidase/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits , Substrate Specificity
18.
Biotechnol Appl Biochem ; 35(1): 35-43, 2002 02.
Article in English | MEDLINE | ID: mdl-11834128

ABSTRACT

To improve the production of oligo-1,6-glucosidase from the obligately thermophilic Bacillus thermoglucosidasius KP1006 in Escherichia coli, the combined expression of oligo-1,6-glucosidase with various chaperone proteins of Hsp (heat-shock protein) 60 team proteins (GroES and GroEL) or Hsp70 team proteins (GrpE, DnaK and DnaJ) from the same thermophile was examined. This attempt was based on the facts that, (i) among glycosyl hydrolases of Family 13, bacillary oligo-1,6-glucosidases share highest homology with yeast alpha-glucosidase, and (ii) this yeast enzyme interacts with GroEL. In B. thermoglucosidasius Hsp60 team proteins, in particular, GroEL brought about a remarkable rise in expression of B. thermoglucosidasius oligo-1,6-glucosidase, while Hsp70 team proteins had no significant effect. The effect of B. thermoglucosidasius GroEL on oligo-1,6-glucosidase expression was supported by the finding that thermally inactivated B. thermoglucosidasius oligo-1,6-glucosidase was revived by B. thermoglucosidasius GroEL. Although the molecular mass of B. thermoglucosidasius oligo-1,6-glucosidase (66 kDa) exceeds the major range of substrates for GroEL proteins, the GroEL molecules probably recognized the alpha/beta motifs contained in the N-terminal domain and the subdomain of the oligo-1,6-glucosidase. Here we show that (i) the production of B. thermoglucosidasius oligo-1,6-glucosidase in E. coli was improved 3.8-fold by Hsp60 team proteins, (ii) the system can function for the expression of other glycosyl hydrolases of Family 13 that have defects in expression and (iii) the combinatorial expression of thermostable proteins with GroEL from the same thermophile in E. coli can increase the production of thermostable enzymes, preventing problems derived from differences in protein biogenesis.


Subject(s)
Bacillus/enzymology , Chaperonin 60/metabolism , Escherichia coli/genetics , Oligo-1,6-Glucosidase/genetics , Amino Acid Sequence , Bacillus/genetics , Base Sequence , Chaperonin 60/genetics , Chaperonin 60/isolation & purification , Cloning, Molecular , Enzyme Activation , Genes, Bacterial , Hot Temperature , Kinetics , Molecular Sequence Data , Oligo-1,6-Glucosidase/chemistry , Oligo-1,6-Glucosidase/metabolism , Plasmids , Promoter Regions, Genetic , Sequence Homology, Amino Acid
19.
Biosci Biotechnol Biochem ; 65(9): 2058-64, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11676021

ABSTRACT

Three active site residues (Asp199, Glu255, Asp329) and two substrate-binding site residues (His103, His328) of oligo-1,6-glucosidase (EC 3.2.1.10) from Bacillus cereus ATCC7064 were identified by site-directed mutagenesis. These residues were deduced from the X-ray crystallographic analysis and the comparison of the primary structure of the oligo-1,6-glucosidase with those of Saccharomyces carlsbergensis alpha-glucosidase, Aspergillus oryzae alpha-amylase and pig pancreatic alpha-amylase which act on alpha-1,4-glucosidic linkages. The distances between these putative residues of B. cereus oligo-1,6-glucosidase calculated from the X-ray analysis data closely resemble those of A. oryzae alpha-amylase and pig pancreatic alpha-amylase. A single mutation of Asp199-->Asn, Glu255-->Gln, or Asp329-->Asn resulted in drastic reduction in activity, confirming that three residues are crucial for the reaction process of alpha-1,6-glucosidic bond cleavage. Thus, it is identified that the basic mechanism of oligo-1,6-glucosidase for the hydrolysis of alpha-1,6-glucosidic linkage is essentially the same as those of other amylolytic enzymes belonging to Family 13 (alpha-amylase family). On the other hand, mutations of histidine residues His103 and His328 resulted in pronounced dissimilarity in catalytic function. The mutation His328-->Asn caused the essential loss in activity, while the mutation His103-->Asn yielded a mutant enzyme that retained 59% of the k0/Km of that for the wild-type enzyme. Since mutants of other alpha-amylases acting on alpha-1,4-glucosidic bond linkage lost most of their activity by the site-directed mutagenesis at their equivalent residues to His103 and His328, the retaining of activity by His103-->Asn mutation in B. cereus oligo-1,6-glucosidase revealed the distinguished role of His103 for the hydrolysis of alpha-1,6-glucosidic bond linkage.


Subject(s)
Bacillus cereus/enzymology , Oligo-1,6-Glucosidase/genetics , Oligo-1,6-Glucosidase/metabolism , Binding Sites , Catalytic Domain , Electrophoresis, Gel, Two-Dimensional , Kinetics , Mutagenesis, Site-Directed , Oligo-1,6-Glucosidase/chemistry , Point Mutation
20.
Biosci Biotechnol Biochem ; 65(7): 1458-63, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11515526

ABSTRACT

Alpha-D-glucosylglycerol (GG) is a mixture of 2-O-alpha-D-glucosylglycerol (GG-II), (2R)-1-O-alpha-D-glucosylglycerol (R-GG-I) and (2S)-1-O-alpha-D-glucosylglycerol (S-GG-I). GG has been found to be slightly hydrolyzed in vitro only by rat intestinal enzymes, but hardly at all by other digestive juices. GG suppressed the hydrolysis of maltose, sucrose and isomaltose by rat intestinal enzymes because the amount of glucose in the digestion of a mixture of GG and disaccharide was less than the sum of that in each individual digestion. The consumption of GG was suppressed by isomaltose, but promoted by maltose, with the hydrolysis of GG being suppressed. Sucrose appeared to suppress only the consumption of S-GG-I, suggesting that S-GG-I was hydrolyzed by the active site of sucrase in a sucrase-isomaltase complex. Transglucosylation seems to have occurred more frequently in the individual digestion of maltose and isomaltose than in that of GG and sucrose. GG seemed to promote transglucosylation in the presence of maltose, to suppress it with sucrose, and to delay it with isomaltose.


Subject(s)
Disaccharides/metabolism , Glucosides/pharmacology , Intestine, Small/enzymology , Animals , Digestion/drug effects , Glucosides/chemistry , Glucosides/metabolism , Glycosylation , Hydrolysis , In Vitro Techniques , Isomaltose/metabolism , Kinetics , Maltose/metabolism , Oligo-1,6-Glucosidase/metabolism , Rats , Substrate Specificity , Sucrase/metabolism , Sucrose/metabolism , alpha-Glucosidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...