Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 706: 149746, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38461646

RESUMEN

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.


Asunto(s)
Péptido Hidrolasas , Péptidos , Péptidos/química , Péptido Hidrolasas/metabolismo , Endopeptidasas/metabolismo , Proteolisis
2.
Nat Chem Biol ; 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321209

RESUMEN

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.

3.
Acta Crystallogr D Struct Biol ; 79(Pt 2): 168-176, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36762862

RESUMEN

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.


Asunto(s)
Quitinasas , Péptidos , Péptido Hidrolasas , beta-Lactamasas/química , Quitinasas/química , Endopeptidasas , Cristalografía por Rayos X
4.
Food Chem ; 383: 132456, 2022 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-35182873

RESUMEN

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.


Asunto(s)
Fibras de la Dieta , alfa-Glucosidasas , Animales , Glucemia , Carbohidratos de la Dieta , Glucosa , Hidrólisis , Mamíferos , Ratas
5.
Biochim Biophys Acta Gen Subj ; 1863(9): 1410-1416, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31254546

RESUMEN

BACKGROUND: Starch constitutes one of the main sources of nutrition in the human diet and is broken down through a number of stages of digestion. Small intestinal breakdown of starch-derived substrates occurs through the mechanisms of small intestinal brush border enzymes, maltase-glucoamylase and sucrase-isomaltase. These enzymes each contain two functional enzymatic domains, and though they share sequence and structural similarities due to their evolutionary conservation, they demonstrate distinct substrate preferences and catalytic efficiency. The N-terminal isomaltase domain of sucrase-isomaltase has a unique ability to actively hydrolyze isomaltose substrates in contrast to the sucrase, maltase and glucoamylase enzymes. METHODS: Through phylogenetic analysis, structural comparisons and mutagenesis, we were able to identify specific residues that play a role in the distinct substrate preference. Mutational analysis and comparison with wild-type activity provide evidence that this role is mediated in part by affecting interactions between the sucrase and isomaltase domains in the intact molecule. RESULTS: The sequence analysis revealed three residues proposed to play key roles in isomaltase specificity. Mutational analysis provided evidence that these residues in isomaltase can also affect activity in the partner sucrase domain, suggesting a close interaction between the domains. MAJOR CONCLUSIONS: The sucrase and isomaltase domains are closely interacting in the mature protein. The activity of each is affected by the presence of the other. GENERAL SIGNIFICANCE: There has been little experimental evidence previously of the effects on activity of interactions between the sucrase-isomaltase enzyme domains. By extension, similar interactions might be expected in the other intestinal α-glucosidase, maltase-glucoamylase.


Asunto(s)
Filogenia , Almidón/metabolismo , Complejo Sacarasa-Isomaltasa/metabolismo , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Hidrólisis , Cinética , Especificidad por Sustrato
6.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S11-S13, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29762369

RESUMEN

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.


Asunto(s)
Intestino Delgado/fisiología , Complejo Sacarasa-Isomaltasa/química , alfa-Glucosidasas/química , Cristalografía , Humanos , Almidón/metabolismo , Complejo Sacarasa-Isomaltasa/fisiología , alfa-Glucosidasas/fisiología
7.
Antonie Van Leeuwenhoek ; 111(9): 1645-1660, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29492769

RESUMEN

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.


Asunto(s)
Vías Biosintéticas/genética , Ácidos Indolacéticos/metabolismo , Pseudomonas/genética , Carga Bacteriana , Proteínas Bacterianas/genética , Liasas de Carbono-Carbono/metabolismo , Clonación Molecular , Expresión Génica , Vectores Genéticos , Vida Libre de Gérmenes , Ácidos Indolacéticos/química , Estructura Molecular , Reguladores del Crecimiento de las Plantas/química , Reguladores del Crecimiento de las Plantas/metabolismo , Raíces de Plantas/crecimiento & desarrollo , Pseudomonas/crecimiento & desarrollo , Pseudomonas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
PLoS One ; 12(3): e0172545, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28273103

RESUMEN

The techniques of metagenomics have allowed researchers to access the genomic potential of uncultivated microbes, but there remain significant barriers to determination of gene function based on DNA sequence alone. Functional metagenomics, in which DNA is cloned and expressed in surrogate hosts, can overcome these barriers, and make important contributions to the discovery of novel enzymes. In this study, a soil metagenomic library carried in an IncP cosmid was used for functional complementation for ß-galactosidase activity in both Sinorhizobium meliloti (α-Proteobacteria) and Escherichia coli (γ-Proteobacteria) backgrounds. One ß-galactosidase, encoded by six overlapping clones that were selected in both hosts, was identified as a member of glycoside hydrolase family 2. We could not identify ORFs obviously encoding possible ß-galactosidases in 19 other sequenced clones that were only able to complement S. meliloti. Based on low sequence identity to other known glycoside hydrolases, yet not ß-galactosidases, three of these ORFs were examined further. Biochemical analysis confirmed that all three encoded ß-galactosidase activity. Lac36W_ORF11 and Lac161_ORF7 had conserved domains, but lacked similarities to known glycoside hydrolases. Lac161_ORF10 had neither conserved domains nor similarity to known glycoside hydrolases. Bioinformatic and structural modeling implied that Lac161_ORF10 protein represented a novel enzyme family with a five-bladed propeller glycoside hydrolase domain. By discovering founding members of three novel ß-galactosidase families, we have reinforced the value of functional metagenomics for isolating novel genes that could not have been predicted from DNA sequence analysis alone.


Asunto(s)
Metagenómica , beta-Galactosidasa/genética , Bacterias/clasificación , Bacterias/enzimología , Bacterias/genética , Clonación Molecular , Biología Computacional/métodos , Expresión Génica , Metagenoma , Metagenómica/métodos , Modelos Moleculares , Anotación de Secuencia Molecular , Sistemas de Lectura Abierta , Filogenia , Conformación Proteica , Análisis de Secuencia de ADN , Microbiología del Suelo , beta-Galactosidasa/química , beta-Galactosidasa/metabolismo
9.
J Agric Food Chem ; 64(33): 6487-94, 2016 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-27480812

RESUMEN

The mammalian mucosal α-glucosidase complexes, maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI), have two catalytic subunits (N- and C-termini). Concurrent with the desire to modulate glycemic response, there has been a focus on di-/oligosaccharides with unusual α-linkages that are digested to glucose slowly by these enzymes. Here, we look at disaccharides with various possible α-linkages and their hydrolysis. Hydrolytic properties of the maltose and sucrose isomers were determined using rat intestinal and individual recombinant α-glucosidases. The individual α-glucosidases had moderate to low hydrolytic activities on all α-linked disaccharides, except trehalose. Maltase (N-terminal MGAM) showed a higher ability to digest α-1,2 and α-1,3 disaccharides, as well as α-1,4, making it the most versatile in α-hydrolytic activity. These findings apply to the development of new glycemic oligosaccharides based on unusual α-linkages for extended glycemic response. It also emphasizes that mammalian mucosal α-glucosidases must be used in in vitro assessment of digestion of such carbohydrates.


Asunto(s)
Digestión , Disacáridos/química , Intestino Delgado/enzimología , Complejo Sacarasa-Isomaltasa/química , alfa-Glucosidasas/química , Animales , Glucosa/química , Concentración de Iones de Hidrógeno , Hidrólisis , Maltosa/química , Oligosacáridos/química , Ratas , Proteínas Recombinantes/química , Almidón/química
10.
Biochem Cell Biol ; 94(3): 241-6, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27093479

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Bacteroides thetaiotaomicron/metabolismo , Colon/microbiología , Polisacáridos/metabolismo , Almidón/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Bacteroides thetaiotaomicron/química , Cristalografía por Rayos X , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Homología Estructural de Proteína , alfa-Glucosidasas/química
11.
Appl Environ Microbiol ; 80(15): 4640-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24837382

RESUMEN

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.


Asunto(s)
Aminohidrolasas/metabolismo , Proteínas Bacterianas/metabolismo , Hidroliasas/metabolismo , Ácidos Indolacéticos/metabolismo , Indoles/metabolismo , Pseudomonas/enzimología , Secuencia de Aminoácidos , Aminohidrolasas/química , Aminohidrolasas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biotransformación , Estabilidad de Enzimas , Hidroliasas/química , Hidroliasas/genética , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Pseudomonas/química , Pseudomonas/genética , Pseudomonas/metabolismo , Alineación de Secuencia , Especificidad por Sustrato , Temperatura
12.
Scientifica (Cairo) ; 2014: 976895, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24579057

RESUMEN

Across the world, many ice active bacteria utilize ice crystal controlling proteins for aid in freezing tolerance at subzero temperatures. Ice crystal controlling proteins include both antifreeze and ice nucleation proteins. Antifreeze proteins minimize freezing damage by inhibiting growth of large ice crystals, while ice nucleation proteins induce formation of embryonic ice crystals. Although both protein classes have differing functions, these proteins use the same ice binding mechanisms. Rather than direct binding, it is probable that these protein classes create an ice surface prior to ice crystal surface adsorption. Function is differentiated by molecular size of the protein. This paper reviews the similar and different aspects of bacterial antifreeze and ice nucleation proteins, the role of these proteins in freezing tolerance, prevalence of these proteins in psychrophiles, and current mechanisms of protein-ice interactions.

13.
Protein Sci ; 23(5): 586-93, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24616181

RESUMEN

Maize ChitA chitinase is composed of a small, hevein-like domain attached to a carboxy-terminal chitinase domain. During fungal ear rot, the hevein-like domain is cleaved by secreted fungal proteases to produce truncated forms of ChitA. Here, we report a structural and biochemical characterization of truncated ChitA (ChitA ΔN), which lacks the hevein-like domain. ChitA ΔN and a mutant form (ChitA ΔN-EQ) were expressed and purified; enzyme assays showed that ChitA ΔN activity was comparable to the full-length enzyme. Mutation of Glu62 to Gln (ChitA ΔN-EQ) abolished chitinase activity without disrupting substrate binding, demonstrating that Glu62 is directly involved in catalysis. A crystal structure of ChitA ΔN-EQ provided strong support for key roles for Glu62, Arg177, and Glu165 in hydrolysis, and for Ser103 and Tyr106 in substrate binding. These findings demonstrate that the hevein-like domain is not needed for enzyme activity. Moreover, comparison of the crystal structure of this plant class IV chitinase with structures from larger class I and II enzymes suggest that class IV chitinases have evolved to accommodate shorter substrates.


Asunto(s)
Quitinasas/química , Quitinasas/metabolismo , Zea mays/enzimología , Quitinasas/genética , Cristalografía por Rayos X , Hidrólisis , Modelos Moleculares , Mutación Puntual , Unión Proteica , Conformación Proteica , Zea mays/química , Zea mays/genética
14.
Mol Nutr Food Res ; 58(5): 1111-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24442968

RESUMEN

SCOPE: The four mucosal α-glucosidases, which differ in their digestive roles, generate glucose from glycemic carbohydrates and accordingly can be viewed as a control point for rate of glucose delivery to the body. In this study, individual recombinant enzymes were used to understand how α-glucan oligomers are digested by each enzyme, and how intermediate α-amylolyzed starches are hydrolyzed, to elucidate a strategy for moderating the glycemic spike of rapidly digestible starchy foods. METHODS AND RESULTS: The C-terminal maltase-glucoamylase (ctMGAM, commonly termed "glucoamylase") was able to rapidly hydrolyze longer maltooligosaccharides, such as maltotetraose and maltopentaose, to glucose. Moreover, it was found to convert larger size maltodextrins, as would be produced early in α-amylase digestion of starch, efficiently to glucose. It is postulated that ctMGAM has the additional capacity to hydrolyze large α-amylase products that are produced immediately on starch digestion in the duodenum and contribute to the rapid generation of glucose from starch-based meals. CONCLUSION: The findings suggest that partial inhibition of ctMGAM, such as by natural inhibitors found in foods, might be used to moderate the early stage of high glycemic response, as well as to extend digestion distally; thereby having relevance in regulating glucose delivery to the body.


Asunto(s)
Glucosa/metabolismo , Almidón/química , alfa-Glucosidasas/metabolismo , Glucemia/metabolismo , Digestión , Duodeno/metabolismo , Humanos , Hidrólisis , Maltosa/análogos & derivados , Maltosa/metabolismo , Membrana Mucosa/metabolismo , Oligosacáridos/metabolismo , Polisacáridos/metabolismo , Periodo Posprandial , Proteínas Recombinantes/metabolismo , alfa-Amilasas/metabolismo
15.
Stem Cell Investig ; 1: 6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-27358853

RESUMEN

Cutaneous metastasis as an initial presentation occurs in 0.8% of patients with internal malignancies, and is poorly understood in its molecular pathogenesis. We reported a case in which a 61-year-old male patient initially presented with rapidly growing skin nodule on his left chest wall, then developed dyspnea and loss of weight. Echocardiogram showed a large pericardial effusion with right ventricular collapse. PET/CT revealed moderate pleural effusion and multiple lymphadenopathies with hypermetabolic concentration of radiotracer in the lymph nodes as well as in the chest wall skin mass. Biopsy of the skin mass and pericardial/pleural fluids revealed metastatic adenocarcinoma consistent with lung primary with KRAS mutation. Palliative chemotherapy was administered without resulting in any improvement. This is the first case report to show that KRAS-mutant lung adenocarcinoma can be associated with cutaneous metastasis.

16.
Stand Genomic Sci ; 8(1): 106-11, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23961315

RESUMEN

This report summarizes the events of the 1(st) International Functional Metagenomics Workshop. The workshop was held on May 7 and 8, 2012, in St. Jacobs, Ontario, Canada and was focused on building an international functional metagenomics community, exploring strategic research areas, and identifying opportunities for future collaboration and funding. The workshop was initiated by researchers at the University of Waterloo with support from the Ontario Genomics Institute (OGI), Natural Sciences and Engineering Research Council of Canada (NSERC) and the University of Waterloo.

17.
PLoS One ; 8(4): e59745, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23565164

RESUMEN

For digestion of starch in humans, α-amylase first hydrolyzes starch molecules to produce α-limit dextrins, followed by complete hydrolysis to glucose by the mucosal α-glucosidases in the small intestine. It is known that α-1,6 linkages in starch are hydrolyzed at a lower rate than are α-1,4 linkages. Here, to create designed slowly digestible carbohydrates, the structure of waxy corn starch (WCS) was modified using a known branching enzyme alone (BE) and an in combination with ß-amylase (BA) to increase further the α-1,6 branching ratio. The digestibility of the enzymatically synthesized products was investigated using α-amylase and four recombinant mammalian mucosal α-glucosidases. Enzyme-modified products (BE-WCS and BEBA-WCS) had increased percentage of α-1,6 linkages (WCS: 5.3%, BE-WCS: 7.1%, and BEBA-WCS: 12.9%), decreased weight-average molecular weight (WCS: 1.73×10(8) Da, BE-WCS: 2.76×10(5) Da, and BEBA-WCS 1.62×10(5) Da), and changes in linear chain distributions (WCS: 21.6, BE-WCS: 16.9, BEBA-WCS: 12.2 DPw). Hydrolysis by human pancreatic α-amylase resulted in an increase in the amount of branched α-limit dextrin from 26.8% (WCS) to 56.8% (BEBA-WCS). The α-amylolyzed samples were hydrolyzed by the individual α-glucosidases (100 U) and glucogenesis decreased with all as the branching ratio increased. This is the first report showing that hydrolysis rate of the mammalian mucosal α-glucosidases is limited by the amount of branched α-limit dextrin. When enzyme-treated materials were gavaged to rats, the level of postprandial blood glucose at 60 min from BEBA-WCS was significantly higher than for WCS or BE-WCS. Thus, highly branched glucan structures modified by BE and BA had a comparably slow digesting property both in vitro and in vivo. Such highly branched α-glucans show promise as a food ingredient to control postprandial glucose levels and to attain extended glucose release.


Asunto(s)
Glucosa/biosíntesis , Glucosa/química , Polisacáridos/química , Polisacáridos/metabolismo , alfa-Glucosidasas/metabolismo , Animales , Glucemia , Humanos , Hidrólisis , Masculino , Peso Molecular , Membrana Mucosa/enzimología , Resonancia Magnética Nuclear Biomolecular , Ratas , Proteínas Recombinantes/metabolismo , Almidón/química , Almidón/metabolismo
18.
J Biol Chem ; 288(19): 13563-74, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23536181

RESUMEN

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.


Asunto(s)
Glicoproteínas de Membrana/química , Simulación del Acoplamiento Molecular , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , alfa-Glucosidasas/química , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/química , Conformación de Carbohidratos , Secuencia de Carbohidratos , Dominio Catalítico , Cristalografía por Rayos X , Glucosa/química , Inhibidores de Glicósido Hidrolasas , Enlace de Hidrógeno , Cinética , Glicoproteínas de Membrana/antagonistas & inhibidores , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Homología Estructural de Proteína , Especificidad por Sustrato , Trisacáridos/química
19.
Cytotechnology ; 65(4): 513-21, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23076800

RESUMEN

The Atlantic salmon (Salmo salar) serum lectin (SSL) is a soluble C-type lectin that binds bacteria, including salmon pathogens. This lectin is a cysteine-rich oligomeric protein. Consequently, a Drosophila melanogaster expression system was evaluated for use in expressing SSL. A cDNA encoding SSL was cloned into a vector designed to express it as a fusion protein with a hexahistidine tag, under the control of the Drosophila methallothionein promoter. The resulting construct was stably transfected into Drosophila S2 cells. After CdCl2 induction, transfected S2 cells secreted recombinant SSL into the cell culture medium. A cell line derived from stably transformed polyclonal cell populations expressing SSL was used for large-scale expression of SSL. Recombinant SSL was purified from the culture medium using a two-step purification scheme involving affinity binding to yeast cells and metal-affinity chromatography. Although yields of SSL were very low, correct folding and functionality of the recombinant SSL purified in this manner was demonstrated by its ability to bind to Aeromonas salmonicida. Therefore, Drosophila S2 cells may be an ideal system for the production of SSL if yields can be increased.

20.
Protein Expr Purif ; 86(2): 135-41, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23036359

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/aislamiento & purificación , Bacteroides/enzimología , alfa-Glucosidasas/biosíntesis , alfa-Glucosidasas/aislamiento & purificación , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bacteroides/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Maltosa/química , Maltosa/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia , alfa-Glucosidasas/química , alfa-Glucosidasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...