Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Appl Microbiol Biotechnol ; 107(20): 6205-6217, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37642718

RESUMO

Naringin found in citrus fruits is a flavanone glycoside with numerous biological activities. However, the bitterness, low water-solubility, and low bioavailability of naringin are the main issues limiting its use in the pharmaceutical and nutraceutical industries. Herein, a glucansucrase from isolated Leuconostoc citreum NY87 was used for trans-α-glucosylattion of naringin by using sucrose as substrate. Two naringin glucosides (O-α-D-glucosyl-(1'''' → 6″) naringin (compound 1) and 4'-O-α-D-glucosyl naringin (compound 2)) were purified and determined their structures by nuclear magnetic resonance. The optimization condition for the synthesis of compound 1 was obtained at 10 mM naringin, 200 mM sucrose, and 337.5 mU/mL at 28 °C for 24 h by response surface methodology method. Compound 1 and compound 2 showed 1896- and 3272 times higher water solubility than naringin. Furthermore, the bitterness via the human bitter taste receptor TAS2R39 displayed that compound 1 was reduced 2.9 times bitterness compared with naringin, while compound 2 did not express bitterness at 1 mM. Both compounds expressed higher neuroprotective effects than naringin on human neuroblastoma SH-SY5Y cells treated with 5 mM scopolamine based on cell viability and cortisol content. Compound 1 reduced acetylcholinesterase activity more than naringin and compound 2. These results indicate that naringin glucosides could be utilized as functional material in the nutraceutical and pharmaceutical industries. KEY POINTS: • A novel O-α-D-glucosyl-(1 → 6) naringin was synthesized using glucansucrase from L. citreum NY87. • Naringin glucosides improved water-solubility and neuroprotective effects on SH-SY5Y cells. • Naringin glucosides showed a decrease in bitterness on bitter taste receptor 39.


Assuntos
Flavanonas , Neuroblastoma , Fármacos Neuroprotetores , Humanos , Fármacos Neuroprotetores/farmacologia , Solubilidade , Acetilcolinesterase , Flavanonas/farmacologia , Sacarose/química , Glucosídeos/farmacologia , Glucosídeos/química , Água , Receptores de Superfície Celular
2.
Biosci Biotechnol Biochem ; 87(7): 707-716, 2023 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-37055368

RESUMO

Glycoside hydrolase family 3 (GH3) ß-glucosidase exists in many filamentous fungi. In phytopathogenic fungi, it is involved in fungal growth and pathogenicity. Microdochium nivale is a severe phytopathogenic fungus of grasses and cereals and is the causal agent of pink snow mold, but its ß-glucosidase has not been identified. In this study, a GH3 ß-glucosidase of M. nivale (MnBG3A) was identified and characterized. Among various p-nitrophenyl ß-glycosides, MnBG3A showed activity on d-glucoside (pNP-Glc) and slight activity on d-xyloside. In the pNP-Glc hydrolysis, substrate inhibition occurred (Kis = 1.6 m m), and d-glucose caused competitive inhibition (Ki = 0.5 m m). MnBG3A acted on ß-glucobioses with ß1-3, -6, -4, and -2 linkages, in descending order of kcat/Km. In contrast, the regioselectivity for newly formed products was limited to ß1-6 linkage. MnBG3A has similar features to those of ß-glucosidases from Aspergillus spp., but higher sensitivity to inhibitory effects.


Assuntos
Glicosídeo Hidrolases , beta-Glucosidase , beta-Glucosidase/genética , beta-Glucosidase/metabolismo , Glicosídeos/química , Fungos/metabolismo , Especificidade por Substrato , Cinética
3.
J Struct Biol ; 214(3): 107874, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35688347

RESUMO

An α-glucosidase from Aspergillus sojae, AsojAgdL, exhibits strong transglucosylation activity to produce α-1,6-glucosidic linkages. The most remarkable structural feature of AsojAgdL is that residues 457-560 of AsojAgdL (designated the NC sequence) is not conserved in other glycoside hydrolase family 31 enzymes, and part of this NC sequence is proteolytically cleaved during its maturation. In this study, the enzyme was expressed in Pichia pastoris, and electrophoretic analysis indicated that the recombinant enzyme, rAsojAgdL, consisted of two polypeptide chains, as observed in the case of the enzyme produced in an Aspergillus strain. The crystal structure of rAsojAgdL was determined in complex with the substrate analog trehalose. Electron density corresponding to residues 496-515 of the NC sequence was not seen, and there were no α-helices or ß-strands except for a short α-helix in the structures of residues 457-495 and residues 516-560, both of which belong to the NC sequence. The residues 457-495 and the residues 516-560 both formed extra components of the catalytic domain. The residues 457-495 constituted the entrance of the catalytic pocket of rAsojAgdL, and Gly467, Asp468, Pro469, and Pro470 in the NC sequence were located within 4 Å of Trp400, a key residue involved in binding of the substrate. The results suggest that the proteolytic processing of the NC sequence is related to the formation of the catalytic pocket of AsojAgdL.


Assuntos
Aspergillus , alfa-Glucosidases , Aspergillus/genética , Aspergillus/metabolismo , Domínio Catalítico , Especificidade por Substrato , alfa-Glucosidases/química , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo
4.
Biosci Biotechnol Biochem ; 85(7): 1706-1710, 2021 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-34014266

RESUMO

We constructed enzyme variants of the α-glucosidases from Aspergillus oryzae (AoryAgdS) and Aspergillus sojae (AsojAgdL) by mutating the amino acid residue at position 450. AoryAgdS_H450R acquired the ability to produce considerable amounts of α-1,6-transglucosylation products, whereas AsojAgdL_R450H changed to produce more α-1,3- and α-1,4-transglucosylation products than α-1,6-products. The 450th amino acid residue is critical for the transglucosylation of these α-glucosidases.


Assuntos
Substituição de Aminoácidos , Aspergillus oryzae/enzimologia , Aspergillus/enzimologia , alfa-Glucosidases/metabolismo , Sequência de Aminoácidos , Glicosilação , Homologia de Sequência de Aminoácidos , alfa-Glucosidases/química
5.
Microb Cell Fact ; 19(1): 140, 2020 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-32652991

RESUMO

BACKGROUND: α-Glucosidases are widely distributed enzymes with a varied substrate specificity that are traditionally used in biotechnological industries based on oligo- and polysaccharides as starting materials. According to amino acid sequence homology, α-glucosidases are included into two major families, GH13 and GH31. The members of family GH13 contain several α-glucosidases with confirmed hydrolytic activity on sucrose. Previously, a sucrose splitting activity from the nectar colonizing yeast Metschnikowia reukaufii which produced rare sugars with α-(1→1), α-(1→3) and α-(1→6) glycosidic linkages from sucrose was described. RESULTS: In this study, genes codifying for α-glucosidases from the nectaries yeast M. gruessii and M. reukaufii were characterised and heterologously expressed in Escherichia coli for the first time. Recombinant proteins (Mg-αGlu and Mr-αGlu) were purified and biochemically analysed. Both enzymes mainly displayed hydrolytic activity towards sucrose, maltose and p-nitrophenyl-α-D-glucopyranoside. Structural analysis of these proteins allowed the identification of common features from the α-amylase family, in particular from glycoside hydrolases that belong to family GH13. The three acidic residues comprising the catalytic triad were identified and their relevance for the protein hydrolytic mechanism confirmed by site-directed mutagenesis. Recombinant enzymes produced oligosaccharides naturally present in honey employing sucrose as initial substrate and gave rise to mixtures with the same products profile (isomelezitose, trehalulose, erlose, melezitose, theanderose and esculose) previously obtained with M. reukaufii cell extracts. Furthermore, the same enzymatic activity was detected with its orthologous Mg-αGlu from M. gruessii. Interestingly, the isomelezitose amounts obtained in reactions mediated by the recombinant proteins, ~ 170 g/L, were the highest reported so far. CONCLUSIONS: Mg/Mr-αGlu were heterologously overproduced and their biochemical and structural characteristics analysed. The recombinant α-glucosidases displayed excellent properties in terms of mild reaction conditions, in addition to pH and thermal stability. Besides, the enzymes produced a rare mixture of hetero-gluco-oligosaccharides by transglucosylation, mainly isomelezitose and trehalulose. These compounds are natural constituents of honey which purification from this natural source is quite unviable, what make these enzymes very interesting for the biotechnological industry. Finally, it should be remarked that these sugars have potential applications as food additives due to their suitable sweetness, viscosity and humectant capacity.


Assuntos
Proteínas Fúngicas , Metschnikowia/enzimologia , Proteínas Recombinantes , alfa-Glucosidases , Clonagem Molecular , Escherichia coli/metabolismo , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/química , Cinética , Metschnikowia/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Especificidade por Substrato , Açúcares/metabolismo , alfa-Glucosidases/biossíntese , alfa-Glucosidases/química
6.
Molecules ; 25(3)2020 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-31979165

RESUMO

Steroidal glycosides are important sources of innovative drugs. The increased diversification of steroidal glycosides will expand the probability of discovering active molecules. It is an efficient approach to diversify steroidal glycosides by using steroidal glycosyltransferases. OcUGT1, a uridine diphosphate-d-glucose (UDP-Glc)-dependent glycosyltransferase from Ornithogalum caudatum, is a multifunctional enzyme, and its glycodiversification potential towards steroids has never been fully explored. Herein, the glycodiversification capability of OcUGT1 towards 25 steroids through glucosylation and transglucosylation reactions were explored. Firstly, each of 25 compounds was glucosylated with UDP-Glc. Under the action of OcUGT1, five steroids (testosterone, deoxycorticosterone, hydrocortisone, estradiol, and 4-androstenediol) were glucosylated to form corresponding mono-glucosides and biosides. Next, OcUGT1-mediated transglucosylation activity of these compounds with another sugar donor ortho-nitrophenyl-ß-d-glucopyranoside (oNPGlc) was investigated. Results revealed that the same five steroids could be glucosylated to generate mono-glucosides and biosides by OcUGT1 through transglucosylation reactions. These data indicated that OcUGT1-assisted glycodiversification of steroids could be achieved through glucosylation and transglucosylation reactions. These results provide a way to diversify steroidal glycosides, which lays the foundation for the increase of the probability of obtaining active lead compounds.


Assuntos
Glucosídeos/metabolismo , Glicosídeos/metabolismo , Glicosiltransferases/metabolismo , Esteroides/metabolismo , Glicosilação , Ornithogalum/química
7.
Biochem Biophys Res Commun ; 504(4): 647-653, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30205957

RESUMO

Dalcochinase from Dalbergia cochinchinensis Pierre and linamarase from Manihot esculenta Crantz are ß-glucosidases which share 47% sequence identity, but show distinct substrate specificities in hydrolysis and transglucosylation. Previously, three amino acid residues of dalcochinase, namely I185, N189 and V255, were identified as being important for determining substrate specificity. In this study, kinetic analysis of the ensuing double and triple mutants of dalcochinase showed that only those containing the 185A mutation could appreciably hydrolyze linamarin as well as transfer glucose to 2-methyl-2-propanol. So, the space provided by the I185A mutation appeared to be a prerequisite for accommodation of the aglycone moiety containing three substituents at the carbinol carbon. However, quantitative analysis of the energy parameters revealed mostly antagonistic interactions between these mutations. In addition, the N189F mutant showed a potential for use in enzymatic synthesis of alkyl glucosides via transglucosylation and reverse hydrolysis reactions. Thus, substitution of only 2-3 key residues in the aglycone binding pocket of dalcochinase could convert its specificities to that of linamarase, as well as to be suitable for any chosen hydrolytic or synthetic applications.


Assuntos
Glicosídeos/metabolismo , Mutação , Proteínas de Plantas/genética , beta-Glucosidase/genética , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Sítios de Ligação/genética , Dalbergia/enzimologia , Dalbergia/genética , Glicosídeos/química , Hidrólise , Cinética , Manihot/enzimologia , Manihot/genética , Modelos Moleculares , Estrutura Molecular , Nitrilas/química , Nitrilas/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Domínios Proteicos , Especificidade por Substrato , beta-Glucosidase/química , beta-Glucosidase/metabolismo
8.
Arch Biochem Biophys ; 652: 3-8, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29885290

RESUMO

Amylomaltase (AM) catalyzes inter- and intra-molecular transglycosylation reactions of glucan to yield linear and cyclic oligosaccharide products. The functional roles of the conserved histidine at position 461 in the active site of AM from Corynebacterium glutamicum (CgAM) was investigated. H461 A/S/D/R/W were constructed, their catalytic properties were compared to the wild-type (WT). A significant decrease in transglucosylation activities was observed, especially in H461A mutant, while hydrolysis activity was barely affected. The transglucosylation factor of the H461A-CgAM was decreased by 8.6 folds. WT preferred maltotriose (G3) as substrate for disproportionation reaction, but all H461 mutants showed higher preference for maltose (G2). Using G3 substrate, kcat/Km values of H461 mutated CgAMs were 40-64 folds lower, while the Km values were twice higher than those of WT. All mutants could not produce large-ring cyclodextrin (LR-CD) product. The heat capacity profile indicated that WT had higher thermal stability than H461A. The X-ray structure of WT showed two H-bonds between H461 and heptasaccharide analog at subsite +1, while no such bonding was observed from the model structure of H461A. The importance of H461 on substrate binding with CgAM was evidenced. We are the first to mutate an active site histidine in AM to explore its function.


Assuntos
Proteínas de Bactérias/metabolismo , Corynebacterium glutamicum/enzimologia , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Varredura Diferencial de Calorimetria , Catálise , Domínio Catalítico , Dicroísmo Circular , Cristalografia por Raios X , Estabilidade Enzimática , Sistema da Enzima Desramificadora do Glicogênio/química , Sistema da Enzima Desramificadora do Glicogênio/genética , Glicosilação , Ligação de Hidrogênio , Cinética , Mutação , Estrutura Secundária de Proteína , Especificidade por Substrato
9.
Appl Microbiol Biotechnol ; 102(19): 8145-8152, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30032433

RESUMO

Arbutin, a glucoside of hydroquinone, is used as a powerful skin lightening agent in the cosmeceutical industry because of its strong inhibitory effect on the human tyrosinase activity. It is a natural compound occurring in a number of plants, with a ß-anomeric form of the glycoside bond between glucose and hydroquinone. α-Arbutin, which glycoside bond is generated with α-anomeric form, is the isomer of natural arbutin. α-Arbutin is generally produced by transglucosylation of hydroquinone by microbial glycosyltransferases. It is interesting that α-arbutin is found to be over 10 times more effective than arbutin, and thus biological production of α-arbutin attracts increasing attention. Seven different microbial enzymes have been identified to be able to produce α-arbutin, including α-amylase, sucrose phosphorlase, cyclodextrin glycosyltransferase, α-glucosidase, dextransucrase, amylosucrase, and sucrose isomerase. In this work, enzymatic and microbial production of α-arbutin is reviewed in detail.


Assuntos
Arbutina/biossíntese , Arbutina/metabolismo , Produtos Biológicos/metabolismo , Animais , Bactérias/metabolismo , Glucosídeos/biossíntese , Glucosídeos/metabolismo , Glicosiltransferases/metabolismo , Humanos , Hidroquinonas/metabolismo
10.
Biochem Biophys Res Commun ; 488(3): 516-521, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28522291

RESUMO

Amylomaltase catalyzes α-1,4 glucosyl transfer reaction to yield linear or cyclic oligosaccharide products. The aim of this work is to investigate functional roles of 410s loop unique to amylomaltase from Corynebacterium glutamicum (CgAM). Site-directed mutagenesis of Y418, the residue at the loop tip, was performed. Y418A/S/D/R/W/F - CgAMs were characterized and compared to the wild-type (WT). A significant decrease in starch transglucosylation, disproportionation and cyclization activities was observed. Specificity for G3 substrate in disproportionation reaction was not changed; however, Y418F showed an increase in preference for longer oligosaccharides G5 to G7. The catalytic efficiency of Y418 mutated CgAMs, except for Y418F, was significantly lower (up to 8- and 12- fold for the W and R mutants, respectively) than that of WT. The change was in the kcat, not the Km values which were around 16-20 mM. The profile of large-ring cyclodextrin (LR-CD) product was different; the principal product of Y418A/D/S was shifted to the larger size (CD36-CD40) while that of the WT and Y418F peaked at CD29-CD33. The product yield was reduced especially in W and R mutants. Hence Y418 in 410s loop of CgAM not only contributes to transglucosylation activities but also controls the amount and size of LR-CD products through the proposed hydrophobic stacking interaction and the suitable distance of loop channel for substrate entering. This is the first report to show the effect of the loop tip residue on LR-CD product formation.


Assuntos
Corynebacterium glutamicum/enzimologia , Ciclodextrinas/biossíntese , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Sistema da Enzima Desramificadora do Glicogênio/genética , Glicosilação , Mutagênese Sítio-Dirigida
11.
Appl Microbiol Biotechnol ; 101(16): 6399-6408, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28688044

RESUMO

Aspergillus niger α-glucosidase (ANG), a member of glycoside hydrolase family 31, catalyzes hydrolysis of α-glucosidic linkages at the non-reducing end. In the presence of high concentrations of maltose, the enzyme also catalyzes the formation of α-(1→6)-glucosyl products by transglucosylation and it is used for production of the industrially useful panose and isomaltooligosaccharides. The initial transglucosylation by wild-type ANG in the presence of 100 mM maltose [Glc(α1-4)Glc] yields both α-(1→6)- and α-(1→4)-glucosidic linkages, the latter constituting ~25% of the total transfer reaction product. The maltotriose [Glc(α1-4)Glc(α1-4)Glc], α-(1→4)-glucosyl product disappears quickly, whereas the α-(1→6)-glucosyl products panose [Glc(α1-6)Glc(α1-4)Glc], isomaltose [Glc(α1-6)Glc], and isomaltotriose [Glc(α1-6)Glc(α1-6)Glc] accumulate. To modify the transglucosylation properties of ANG, residue Asn694, which was predicted to be involved in formation of the plus subsites of ANG, was replaced with Ala, Leu, Phe, and Trp. Except for N694A, the mutations enhanced the initial velocity of the α-(1→4)-transfer reaction to produce maltotriose, which was then degraded at a rate similar to that by wild-type ANG. With increasing reaction time, N694F and N694W mutations led to the accumulation of larger amounts of isomaltose and isomaltotriose than achieved with the wild-type enzyme. In the final stage of the reaction, the major product was panose (N694A and N694L) or isomaltose (N694F and N694W).


Assuntos
Aspergillus niger/genética , Aspergillus niger/metabolismo , Mutação , alfa-Glucosidases/química , alfa-Glucosidases/genética , Aspergillus niger/efeitos dos fármacos , Aspergillus niger/enzimologia , Glucanos/metabolismo , Glucanos/farmacologia , Concentração de Íons de Hidrogênio , Hidrólise , Isomaltose/metabolismo , Cinética , Maltose/metabolismo , Maltose/farmacologia , Mutagênese Sítio-Dirigida , Especificidade por Substrato , Trissacarídeos/metabolismo , alfa-Glucosidases/metabolismo
12.
Cell Mol Life Sci ; 73(14): 2727-51, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27137181

RESUMO

α-Glucosidases (AGases) and α-1,4-glucan lyases (GLases) catalyze the degradation of α-glucosidic linkages at the non-reducing ends of substrates to release α-glucose and anhydrofructose, respectively. The AGases belong to glycoside hydrolase (GH) families 13 and 31, and the GLases belong to GH31 and share the same structural fold with GH31 AGases. GH13 and GH31 AGases show diverse functions upon the hydrolysis of substrates, having linkage specificities and size preferences, as well as upon transglucosylation, forming specific α-glucosidic linkages. The crystal structures of both enzymes were determined using free and ligand-bound forms, which enabled us to understand the important structural elements responsible for the diverse functions. A series of mutational approaches revealed features of the structural elements. In particular, amino-acid residues in plus subsites are of significance, because they regulate transglucosylation, which is used in the production of industrially valuable oligosaccharides. The recently solved three-dimensional structure of GLase from red seaweed revealed the amino-acid residues essential for lyase activity and the strict recognition of the α-(1 â†’ 4)-glucosidic substrate linkage. The former was introduced to the GH31 AGase, and the resultant mutant displayed GLase activity. GH13 and GH31 AGases hydrate anhydrofructose to produce glucose, suggesting that AGases are involved in the catabolic pathway used to salvage unutilized anhydrofructose.


Assuntos
Polissacarídeo-Liases/química , Polissacarídeo-Liases/metabolismo , alfa-Glucosidases/química , alfa-Glucosidases/metabolismo , Sequência de Aminoácidos , Glicosilação , Modelos Moleculares , Especificidade por Substrato , Sacarose/metabolismo
13.
Cell Mol Life Sci ; 73(14): 2661-79, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27141938

RESUMO

Amylosucrases and branching sucrases are α-retaining transglucosylases found in the glycoside-hydrolase families 13 and 70, respectively, of the clan GH-H. These enzymes display unique activities in their respective families. Using sucrose as substrate and without mediation of nucleotide-activated sugars, amylosucrase catalyzes the formation of an α-(1 â†’ 4) linked glucan that resembles amylose. In contrast, the recently discovered branching sucrases are unable to catalyze polymerization of glucosyl units as they are rather specific for dextran branching through α-(1 â†’ 2) or α-(1 â†’ 3) branching linkages depending on the enzyme regiospecificity. In addition, GH13 amylosucrases and GH70 branching sucrases are naturally promiscuous and can glucosylate different types of acceptor molecules including sugars, polyols, or flavonoids. Amylosucrases have been the most investigated glucansucrases, in particular to control product profiles or to successfully develop tailored α-transglucosylases able to glucosylate various molecules of interest, for example, chemically protected carbohydrates that are planned to enter in chemoenzymatic pathways. The structural traits of these atypical enzymes will be described and compared, and an overview of the potential of natural or engineered enzymes for glycodiversification and chemoenzymatic synthesis will be highlighted.


Assuntos
Glucosiltransferases/metabolismo , Glicosiltransferases/metabolismo , Família Multigênica , Sequência de Aminoácidos , Glucosiltransferases/química , Glicosiltransferases/química , Cinética , Modelos Moleculares , Engenharia de Proteínas
14.
Biochem Biophys Res Commun ; 478(2): 759-64, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27507216

RESUMO

Amylomaltase catalyzes intermolecular and intramolecular transglucosylation reactions to form linear and cyclic oligosaccharides, respectively. The aim of this work is to investigate the structure-function relationship of amylomaltase from a mesophilic Corynebacterium glutamicum (CgAM). Site-directed mutagenesis was performed to substitute Tyr for Asn287 (N287Y) to determine its role in controlling amylomaltase activity and product formation. Expression of the wild-type (WT) and N287Y was achieved by cultivating recombinant cells in the medium containing lactose at 16 °C for 14 h. The purified mutated enzyme showed a significant decrease in all transglucosylation activities while hydrolysis activity was not changed. Optimum temperature and pH for disproportionation reaction were slightly changed upon mutation while those for cyclization reaction were not changed. Interestingly, N287Y showed a change in large-ring cyclodextrin (LR-CD) product profile in which the larger size was observed together with an increase in thermostability and substrate preference for G5 in addition to G3. The secondary structure of the mutated enzyme was slightly changed in related to the WT as evidenced from circular dichroism analysis. This work thus demonstrates that N287 is required for transglucosylation activities of CgAM. Having an aromatic residue in this position increased thermostability, changed product profile and substrate preference but demolished most enzyme activities.


Assuntos
Substituição de Aminoácidos , Asparagina/química , Proteínas de Bactérias/química , Corynebacterium glutamicum/química , Sistema da Enzima Desramificadora do Glicogênio/química , Oligossacarídeos/química , Asparagina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biocatálise , Domínio Catalítico , Clonagem Molecular , Corynebacterium glutamicum/enzimologia , Ciclização , Ciclodextrinas/química , Ciclodextrinas/metabolismo , Estabilidade Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Sistema da Enzima Desramificadora do Glicogênio/genética , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Hidrólise , Cinética , Lactose/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Oligossacarídeos/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
15.
Biochem Biophys Res Commun ; 441(4): 838-43, 2013 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-24211208

RESUMO

Cholesteryl glucoside (ß-ChlGlc), a monoglucosylated derivative of cholesterol, is involved in the regulation of heat shock responses. ß-ChlGlc, which is rapidly induced in response to heat shock, activates heat shock transcription factor 1 (HSF1) leading to the expression of heat shock protein 70 (HSP70) in human fibroblasts. Identification and biochemical characterization of the enzyme responsible for ß-ChlGlc formation is important for a complete understanding of the molecular mechanisms leading to HSP70-induction following heat shock. Recently, we demonstrated that ß-ChlGlc synthesis is not dependent on UDP-Glucose but glucosylceramide (GlcCer) in animal tissue and human fibroblasts. In this study, we examined the possibility of glucocerebrosidase, a GlcCer-degrading glycosidase, acting as ß-ChlGlc-synthesizing enzyme. Overexpression of ß-glucosidase 1 (GBA1, lysosomal acid ß-glucocerebrosidase) led to an increase in cholesterol glucosylation activity in human fibroblasts. Using a cell line generated from type 2 Gaucher disease patients with severe defects in GBA1 activity, we found that cholesterol glucosylation activity was very low in the cells and the overexpression of GBA1 rescued the activity. In addition, purified recombinant GBA1 exhibits conduritol B-epoxide-sensitive cholesterol glucosylation activity. The optimum pH and temperature for cholesterol glucosylation by GBA1 were at about 5.3 and 43 °C, respectively. Short chain C8:0-GlcCer was the most effective donor for cholesterol glucosylation activity among GlcCer containing saturated fatty acid (C8:0 to C18:0) tested. GlcCer containing mono-unsaturated fatty acid was more preferred substrate for cholesterol glucosylation when compared with GlcCer containing same chain length of saturated fatty acid. These results demonstrate, for the first time, a novel function of GBA1 as a ß-ChlGlc-synthesizing enzyme. Therefore, our results also reveal a new pathway for glycolipid metabolism in mammals.


Assuntos
Colesterol/análogos & derivados , Glucosilceramidase/metabolismo , beta-Glucosidase/metabolismo , Animais , Catálise , Linhagem Celular , Colesterol/biossíntese , Glucosilceramidase/genética , Glicosilação , Humanos , Camundongos , beta-Glucosidase/genética
16.
J Sci Food Agric ; 93(1): 180-6, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22689251

RESUMO

BACKGROUND: Banana is one of the important crops native to tropical Southeast Asia. Since overproduction frequently leads to excessive waste of produce, alternative uses are continuously sought in order to utilise fruits at all stages of maturity. The aim of this study was to investigate the production of isomaltooligosaccharides (IMOs) from banana flour. RESULTS: Banana slurries liquefied by Termamyl SC and saccharified by either Fungamyl 800 L or barley ß-amylase were used for IMO synthesis by Transglucosidase L. After 12 h of transglucosylation, maximum IMO yields of 76.67 ± 2.71 and 70.74 ± 4.09 g L(-1) respectively were achieved. Although the yields were comparable, the IMO profiles obtained through the use of the two saccharification enzymes were different. Glucose and maltose were removed by 10 g L(-1) bakers' yeast fermentation for 12 h. Regarding total sugars, the final IMO mixture was composed of 53% isomaltotriose, 21% isomaltotetraose and 26% maltooligoheptaose and larger oligomers. CONCLUSION: Banana flour could be used as a potential raw material for IMO synthesis.


Assuntos
Frutas , Glucosidases/metabolismo , Isomaltose/biossíntese , Musa , Oligossacarídeos/biossíntese , Saccharomyces cerevisiae , Fermentação , Farinha , Glucose/metabolismo , Hordeum/enzimologia , Maltose/metabolismo , Trissacarídeos/biossíntese , alfa-Amilases/metabolismo , beta-Amilase/metabolismo
17.
Enzyme Microb Technol ; 159: 110065, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35567857

RESUMO

This study aimed to enhance the water solubility and antioxidant properties of mangiferin by transglucosylation using cyclodextrin glycosyltransferase (CGTase) from Thermoanaerobacter sp. The highest mangiferin to mangiferin glucoside conversion yield achieved was 88.9% using 60 mU/mL CGTase, 25 mM mangiferin, and 10% starch (w/v), with incubation at 60 °C for 10 h. The product of transglucosylation was purified and its chemical structure was determined to be glucosyl-α-(1→4)-mangiferin (MGF-g1) using matrix-assisted laser desorption ionization time-of-flight mass spectrometry and nuclear magnetic resonance spectroscopy. The water solubility of MGF-g1 was 5,093 times higher than that of mangiferin. MGF-g1 exhibited 1.6-fold higher 2,2-diphenyl-1-picrylhydrazyl radical scavenging activity, 1.24-fold higher oxygen radical antioxidant capacity, and 1.19-fold higher ferric reducing ability power, compared to mangiferin. Moreover, the cyclooxygenase-2 inhibitory activity (IC50) of mangiferin and MGF-g1 were 76.44 ± 11.7 µM and 59.74 ± 2.8 µM, respectively. Our results suggest that the novel MGF-g1 has potential applications as a functional material in the food and pharmaceutical industries.


Assuntos
Antioxidantes , Água , Antioxidantes/farmacologia , Glucosiltransferases , Solubilidade , Xantonas
18.
Enzyme Microb Technol ; 161: 110111, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35961060

RESUMO

Fisetin (7,3',4'-flavon-3-ol) is a flavonol found in plants, fruits, and vegetables. It exhibits diverse biological activities, including antioxidant, anti-inflammatory, and anti-cancer effects. However, the low water solubility and bioavailability of fisetin restrict its pharmaceutical applications. In this work, we synthesized a novel fisetin-4'-O-α-D-glucopyranoside (FST-G1) using transglucosylation with sucrose, fisetin, and dextransucrase from Leuconostoc mesenteroides NRRL B-1299CB4. The water solubility of FST-G1 (109.8 ± 6.3 mg/L) was enhanced compared to fisetin (13.6 ± 1.3 mg/L). The antioxidant activities of FST-G1 in non-cellular assays, including ORAC, ABTS•+, and FRAP assays, were lower compared to fisetin. However, FST-G1 exhibited higher nitric oxide inhibition (62.5 µM; 92.3 %) in lipopolysaccharide-stimulated RAW 264.7 murine macrophage cells compared to fisetin (81.4 %). Anti-lipid accumulation in mouse 3T3-L1 cells treated with FST-G1 was similar to that in cells treated with fisetin. Taken together, the novel synthesized FST-G1 is expected to become a promising functional material for using in the pharmaceutical industry.


Assuntos
Leuconostoc mesenteroides , Animais , Antioxidantes/farmacologia , Flavonóis , Glucosídeos/química , Glucosiltransferases/química , Leuconostoc , Camundongos , Solubilidade , Água
19.
3 Biotech ; 11(8): 362, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34295607

RESUMO

Arbutin is a naturally occurring glycosylated product of hydroquinone. With the ability to interrupt melanin biosynthesis in epidermal cells, it is a promising cosmetic ingredient. In this study, a novel amylosucrase, Asmet, identified from a thermal spring metagenome, has been characterized for arbutin biosynthesis. Asmet was able to catalyze transglucosylation of hydroquinone to arbutin, taking sucrose as glycosyl donor, in the temperature range of 20 °C to 40 °C and pH 5.0 to 6.0, with the relative activity of 80% or more. The presence of chloride salts of Li, K, and Na at 1 mM concentration did not exhibit any notable effect on the enzyme's activity, unlike Cu, Ni, and Mn, which were observed to be detrimental. The hydroquinone (20 mM) to sucrose ratio of 1:1 to 1:10 was appropriate for the catalytic biosynthesis of arbutin. The maximum hydroquinone to arbutin conversion of 70% was obtained in 24 h of Asmet led catalysis, at 30 °C and pH 6.0. Arbutin production was also demonstrated using low-cost feedstock, table sugar, muscovado, and sweet sorghum stalk extract, as a replacement for sucrose. Whole-cell catalysis of hydroquinone to arbutin transglucosylation was also established.

20.
AMB Express ; 11(1): 108, 2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34287737

RESUMO

Ellagic acid glucoside was synthesized via transglucosylation using sucrose and glucansucrase derived from Leuconostoc mesenteroides B-512 FMCM. After such enzymatic synthesis, the product was purified by 50% ethyl acetate fraction and C18 column chromatography. Modification of ellagic acid glucoside was verified by LC-MS/MS at m/z 485.1 (M + Na)- and m/z 531.1 (M + 3Na)-. The yield of ellagic acid glucoside was 69% (3.47 mM) by response surface methodology using 150 mM sucrose, 300 mU/mL glucansucrase, and 5 mM ellagic acid. The synthesized ellagic acid glucoside showed improved water solubility, up to 58% higher brain nerve cell (SH-SY5Y) protective effect, threefold higher cortisol reducing effect, and fourfold stronger inhibitory effect on acetylcholinesterase (AChE) than ellagic acid. These results indicate that ellagic acid glucoside could be used as a neuroprotective agent.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa