scispace - formally typeset
Search or ask a question

Showing papers on "Melibiose published in 2019"


Journal ArticleDOI
04 Jul 2019
TL;DR: According to principal component analysis (PCA), galactose, arabinose, and melibiose were the most important sugars in separating the fruits of strawberries from blueberries, while panose, ribose, stachyose, galactOSE, maltose, rhamnoses, and raffinose wereThe most important sugar component in leaves recognition.
Abstract: The objective of this study was to determine and compare the sugar profile, distribution in fruits and leaves and sink-source relationship in three strawberry (‘Favette’, ‘Alba’ and ‘Clery’) and three blueberry cultivars (‘Bluecrop’, ‘Duke’ and ‘Nui’) grown in organic (OP) and integrated production systems (IP). Sugar analysis was done using high-performance anion-exchange chromatography (HPAEC) with pulsed amperometric detection (PAD). The results showed that monosaccharide glucose and fructose and disaccharide sucrose were the most important sugars in strawberry, while monosaccharide glucose, fructose, and galactose were the most important in blueberry. Source-sink relationship was different in strawberry compared to blueberry, having a much higher quantity of sugars in its fruits in relation to leaves. According to principal component analysis (PCA), galactose, arabinose, and melibiose were the most important sugars in separating the fruits of strawberries from blueberries, while panose, ribose, stachyose, galactose, maltose, rhamnose, and raffinose were the most important sugar component in leaves recognition. Galactitol, melibiose, and gentiobiose were the key sugars that split out strawberry fruits and leaves, while galactose, maltotriose, raffinose, fructose, and glucose divided blueberry fruits and leaves in two groups. PCA was difficult to distinguish between OP and IP, because the stress-specific responses of the studied plants were highly variable due to the different sensitivity levels and defense strategies of each cultivar, which directly affected the sugar distribution. Due to its high content of sugars, especially fructose, the strawberry cultivar ‘Clery’ and the blueberry cultivars ‘Bluecrop’ and ‘Nui’ could be singled out in this study as being the most suitable cultivars for OP.

60 citations


Journal ArticleDOI
30 Apr 2019-Forests
TL;DR: In this article, the authors showed that heat stress increased the cellular concentration of H2O2 and the activities of antioxidant enzymes in Populus tomentosa Carr, and the expression levels of most genes related to the light-harvesting complexes and photosynthetic electron transport system were downregulated by heat stress.
Abstract: Plants have evolved mechanisms of stress tolerance responses to heat stress. However, little is known about metabolic responses to heat stress in trees. In this study, we exposed Populus tomentosa Carr. to control (25 °C) and heat stress (45 °C) treatments and analyzed the metabolic and transcriptomic effects. Heat stress increased the cellular concentration of H2O2 and the activities of antioxidant enzymes. The levels of proline, raffinose, and melibiose were increased by heat stress, whereas those of pyruvate, fumarate, and myo-inositol were decreased. The expression levels of most genes (PSB27, PSB28, LHCA5, PETB, and PETC) related to the light-harvesting complexes and photosynthetic electron transport system were downregulated by heat stress. Association analysis between key genes and altered metabolites indicated that glycolysis was enhanced, whereas the tricarboxylic acid (TCA) cycle was suppressed. The inositol phosphate; galactose; valine, leucine, and isoleucine; and arginine and proline metabolic pathways were significantly affected by heat stress. In addition, several transcription factors, including HSFA2, HSFA3, HSFA9, HSF4, MYB27, MYB4R1, and bZIP60 were upregulated, whereas WRKY13 and WRKY50 were downregulated by heat stress. Interestingly, under heat stress, the expression of DREB1, DREB2, DREB2E, and DREB5 was dramatically upregulated at 12 h. Our results suggest that proline, raffinose, melibiose, and several genes (e.g., PSB27, LHCA5, and PETB) and transcription factors (e.g., HSFAs and DREBs) are involved in the response to heat stress in P. tomentosa.

45 citations


Journal ArticleDOI
TL;DR: Raffinose family oligosaccharides (RFOs) negatively affect nutritional value of legume-derived food and feed and a high performance α-galactosidase excelled on catalytic efficiency, thermostability, pH stability and protease-resistance that could efficiently hydrolyze RFOs was developed.

24 citations


Journal ArticleDOI
TL;DR: A mutant in the SUC2 gene (encoding invertase) was constructed and this mutant yielded a very high rate of molasses bioconversion to alpha-galactosidase, and a procedure to recycle the yeast biomass, by using it as nitrogen source to supplement molasses, was developed.
Abstract: Molasses are sub-products of the sugar industry, rich in sucrose and containing other sugars like raffinose, glucose, and fructose. Alpha-galactosidases (EC. 3.2.1.22) catalyze the hydrolysis of alpha-(1,6) bonds of galactose residues in galacto-oligosaccharides (melibiose, raffinose, and stachyose) and complex galactomannans. Alpha-galactosidases have important applications, mainly in the food industry but also in the pharmaceutical and bioenergy sectors. However, the cost of the enzyme limits the profitability of most of these applications. The use of cheap sub-products, such as molasses, as substrates for production of alpha-galactosidases, reduces the cost of the enzymes and contributes to the circular economy. Alpha-galactosidase is a specially indicated bioproduct since, at the same time, it allows to use the raffinose present in molasses. This work describes the development of a two-step system for the valuation of beet molasses, based on their use as substrate for alpha-galactosidase and bioethanol production by Saccharomyces cerevisiae. Since this yeast secretes high amounts of invertase, to avoid congest the secretory route and to facilitate alpha-galactosidase purification from the culture medium, a mutant in the SUC2 gene (encoding invertase) was constructed. After a statistical optimization of culture conditions, this mutant yielded a very high rate of molasses bioconversion to alpha-galactosidase. In the second step, the SUC2 wild type yeast strain fermented the remaining sucrose to ethanol. A procedure to recycle the yeast biomass, by using it as nitrogen source to supplement molasses, was also developed.

20 citations


Journal ArticleDOI
TL;DR: Characteristics and improvement in productivity that have been achieved contribute in making ScAGal a good candidate for application in the elimination of raffinose family oligosaccharides found in many products of the food industry.
Abstract: α-Galactosidases are enzymes that act on galactosides present in many vegetables, mainly legumes and cereals, have growing importance with respect to our diet. For this reason, the use of their catalytic activity is of great interest in numerous biotechnological applications, especially those in the food industry directed to the degradation of oligosaccharides derived from raffinose. The aim of this work has been to optimize the recombinant production and further characterization of α-galactosidase of Saccharomyces cerevisiae. The MEL1 gene coding for the α-galactosidase of S. cerevisiae (ScAGal) was cloned and expressed in the S. cerevisiae strain BJ3505. Different constructions were designed to obtain the degree of purification necessary for enzymatic characterization and to improve the productive process of the enzyme. ScAGal has greater specificity for the synthetic substrate p-nitrophenyl-α-d-galactopyranoside than for natural substrates, followed by the natural glycosides, melibiose, raffinose and stachyose; it only acts on locust bean gum after prior treatment with β-mannosidase. Furthermore, this enzyme strongly resists proteases, and shows remarkable activation in their presence. Hydrolysis of galactose bonds linked to terminal non-reducing mannose residues of synthetic galactomannan-oligosaccharides confirms that ScAGal belongs to the first group of α-galactosidases, according to substrate specificity. Optimization of culture conditions by the statistical model of Response Surface helped to improve the productivity by up to tenfold when the concentration of the carbon source and the aeration of the culture medium was increased, and up to 20 times to extend the cultivation time to 216 h. ScAGal characteristics and improvement in productivity that have been achieved contribute in making ScAGal a good candidate for application in the elimination of raffinose family oligosaccharides found in many products of the food industry.

18 citations


Journal ArticleDOI
TL;DR: Results suggest that LEGI demonstrates potential for elimination of indigestible oligosaccharides in Lentinula edodes.

11 citations


Journal ArticleDOI
TL;DR: The α-galactosidase CLEAs, with their satisfactory enzymatic characteristics, have much potential for use in the food and feed industry.
Abstract: Background Cross-linked enzyme aggregates (CLEAs) of α-galactosidase, partially purified from maize (Zea mays) flour, were prepared. The impact of various parameters on enzyme activity was examined to optimize the immobilization procedure. Biochemical characterization of the free and immobilized enzyme was carried out. Stability (thermal, pH, storage and operational stability) and reusability tests were performed. The potential use of the free enzyme and the CLEAs in hydrolysis processes of raffinose-type oligosaccharides present in soymilk was investigated. Results α-galactosidase CLEAs were prepared with 47% activity recovery under optimum conditions [1:5 (v/v) enzyme solution:saturated ammonium sulfate solution ratio; 7.5 mg protein and 0.1% (v/v) glutaraldehyde, 6 h, 4 °C, 150 rpm]. α-galactosidase CLEAs exhibited increased stability in comparison to the free enzyme. The CLEAs and the free enzyme showed a maximum activity at 40°C and their optimal pH values were5.5 and 6.0, respectively. Kinetic constants (KM , Vmax and kcat ) were calculated for the free enzyme and the CLEAs in the presence of p-nitrophenyl-α-d-galactopyranoside, stachyose, melibiose and raffinose. The effect of various chemicals and sugars on enzyme activity showed that both enzyme forms were significantly inhibited by HgCl2 and galactose. The CLEAs hydrolyzed 85% of raffinose and 96% of stachyose. Conclusion The α-galactosidase CLEAs, with their satisfactory enzymatic characteristics, have much potential for use in the food and feed industry. © 2019 Society of Chemical Industry.

10 citations


Journal ArticleDOI
TL;DR: The results revealed modus operandi of a new carbohydrate utilization system in B. subtilis responsible for utilization of α-galactosides of the raffinose family of oligosaccharides (RFOs) and the operon was renamed to melREDCA.
Abstract: Bacillus subtilis is a heterotrophic soil bacterium that hydrolyzes different polysaccharides mainly found in the decomposed plants. These carbohydrates are mainly cellulose, hemicellulose, and the raffinose family of oligosaccharides (RFOs). RFOs are soluble α-galactosides, such as raffinose, stachyose, and verbascose, that rank second only after sucrose in abundance. Genome sequencing and transcriptome analysis of B. subtilis indicated the presence of a putative α-galactosidase-encoding gene (melA) located in the msmRE-amyDC-melA operon. Characterization of the MelA protein showed that it is a strictly Mn2+- and NAD+-dependent α-galactosidase able to hydrolyze melibiose, raffinose, and stachyose. Transcription of the msmER-amyDC-melA operon is under control of a σA-type promoter located upstream of msmR (PmsmR), which is negatively regulated by MsmR. The activity of PmsmR was induced in the presence of melibiose and raffinose. MsmR is a transcriptional repressor that binds to two binding sites at PmsmR located upstream of the −35 box and downstream of the transcriptional start site. MsmEX-AmyCD forms an ATP-binding cassette (ABC) transporter that probably transports melibiose into the cell. Since msmRE-amyDC-melA is a melibiose utilization system, we renamed the operon melREDCA. IMPORTANCEBacillus subtilis utilizes different polysaccharides produced by plants. These carbohydrates are primarily degraded by extracellular hydrolases, and the resulting oligo-, di-, and monosaccharides are transported into the cytosol via phosphoenolpyruvate-dependent phosphotransferase systems (PTS), major facilitator superfamily, and ATP-binding cassette (ABC) transporters. In this study, a new carbohydrate utilization system of B. subtilis responsible for the utilization of α-galactosides of the raffinose family of oligosaccharides (RFOs) was investigated. RFOs are synthesized from sucrose in plants and are mainly found in the storage organs of plant leaves. Our results revealed the modus operandi of a new carbohydrate utilization system in B. subtilis.

9 citations


Journal ArticleDOI
TL;DR: Overall, a change in anomeric configuration at the reducing end and/or anomersic substitution (methylation) changed the conformational space of the terminal CH2OH group significantly.

4 citations


Journal ArticleDOI
TL;DR: The effect of different carbon sources on the growth as well as on the production of cellulase of the new strain of Microbulbifer hydrolyticus IRE-31 was studied, which laid a good foundation for the further research in the productionof cellulase by marine bacteria.
Abstract: Cellulase is an important enzyme that can be used to breakdown lignocellulose into glucose. Microbulbifer hydrolyticus IRE-31(ATCC 700072) is a kind of marine bacterium, which could grow in high salinity medium and has fast-strong growth ability. In this study, a novel strain was screened from Microbulbifer hydrolyticus IRE-31 through mutations to produce cellulase. The effect of different carbon sources on the growth as well as on the production of cellulase of the new strain was studied. Carboxymethyl-cellulase (CMCase) activity selected to represent cellulase was proven to be effectively promoted while xylose, galactose, and melibiose as well as glucose were used as carbon sources. When xylose and glucose were chosen to be further investigated, 472.57 U/L and 266.01 U/L CMCase activity were obtained from 30 g/L glucose and 10 g/L xylose, respectively. These results clarified the effect of different carbon sources on the production of cellulase, which laid a good foundation for the further research in the production of cellulase by marine bacteria.

3 citations


Journal ArticleDOI
TL;DR: The indium-mediated allylation reaction has been applied to melibiose and yielded the unprotected elongated disaccharides in the equilibrium of the pyranoid as well as furanoid isomers in both anomeric forms, respectively.
Abstract: The indium-mediated allylation reaction has been applied to melibiose, a disaccharidic substrate. This elongation methodology allows for a short, efficient and diastereoselective approach towards complex glycosylated carbohydrate structures. The stereochemical outcome of the key intermediates, allylated disaccharides, has been determined by X-ray analysis. Ozonolysis of the introduced double bond yielded the unprotected elongated disaccharides in the equilibrium of the pyranoid as well as furanoid isomers in both anomeric forms, respectively. Per-O-acetylation has been performed to facilitate separation of the isomeric mixture for structural identification. The main product revealed to adopt a β-pyranoid form of the elongated unit at the reducing end of the disaccharide.

Journal ArticleDOI
TL;DR: The results indicate that IOSs induce browning and generation of the majority of the monitored odorants, in particular 4-hydroxy-2,5-dimethyl-3(2 H)-furanone, 2,3-butanedione, 2-acetyl-1-pyrroline, and 2- acetyl-2-thiazoline, far more than the other oligosaccharides and to a higher or similar degree to that of the monos
Abstract: The generation of selected Maillard-derived odorants from iso-oligosaccharides (IOSs), namely, from isomaltose, isomaltotriose, isomaltulose, and melibiose, was studied and compared with that from other oligosaccharides (maltose, lactose, and panose) and monosaccharides (glucose, galactose, and fructose). The study was carried out in binary mixtures of sugar and amino acids (glycine, proline, and cysteine) and upon wafer baking. The results indicate that IOSs induce browning and generation of the majority of the monitored odorants, in particular 4-hydroxy-2,5-dimethyl-3(2 H)-furanone, 2,3-butanedione, 2-acetyl-1-pyrroline, 2-propionyl-1-pyrroline, 2-acetylthiazole, and 2-acetyl-2-thiazoline, far more than the other oligosaccharides and to a higher or similar degree to that of the monosaccharides. Plausible mechanisms, consistent with the yields obtained from individual sugars, were proposed for the formation of the studied compounds. This newly obtained data brought for the first time evidence about the extraordinary potential of IOSs in the formation of several potent food odorants.

Journal ArticleDOI
01 Jan 2019
TL;DR: In this article, a recombinant α-galactosidase produced by Aspergillus sojae Ta1 was purified using anion exchange and hydrophobic interaction chromatography with an overall yield of 56% and 64.7 U/mg protein specific activity.
Abstract: Galactooligosaccharides are well-known functional food ingredients with prebiotic properties. Recent trend for the use of galactooligosaccharides in the food industry leads the search for new enzymes for their production. α-Galactosidase from Aspergillus fumigatus IMI 385708, possessing a highly efficient debranching ability on polymeric substrates, is also able to perform transgalactosylation. In this study, recombinant α-galactosidase produced by Aspergillus sojae Ta1 was purified 18.7-fold using anion exchange and hydrophobic interaction chromatography with an overall yield of 56% and 64.7 U/mg protein specific activity. The V max and K m values for the hydrolysis of p NPGal were 78 U/mg protein and 0.45 mM, respectively. Optimum pH (pH 4.5) and temperatures (50-60°C) for recombinant α-galactosidase activity were determined. For the synthesis of oligosaccharides, purified and characterized recombinant α-galactosidase was used in the transgalactosylation of various mono- and disaccharides using p NPGal ( p -nitrophenyl-α- D- galactopyranoside) as galactose donor. Di- and trisaccharides obtained by transgalactosylation were analysed by TLC, ESI-MS, and HPLC analysis. Among 12 acceptor candidates, α-galactosidase transgalactosylated galactose, glucose, mannose, cellobiose, lactose, maltose, and sucrose efficiently, however, did not transgalactosylate xylose, arabinose, fucose, fructose, and melibiose.