Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
1.
J Biol Chem ; 299(8): 104967, 2023 08.
Article in English | MEDLINE | ID: mdl-37380079

ABSTRACT

Salmonella enterica serovar Typhimurium melibiose permease (MelBSt) is a prototype of the Na+-coupled major facilitator superfamily transporters, which are important for the cellular uptake of molecules including sugars and small drugs. Although the symport mechanisms have been well-studied, mechanisms of substrate binding and translocation remain enigmatic. We have previously determined the sugar-binding site of outward-facing MelBSt by crystallography. To obtain other key kinetic states, here we raised camelid single-domain nanobodies (Nbs) and carried out a screening against the WT MelBSt under 4 ligand conditions. We applied an in vivo cAMP-dependent two-hybrid assay to detect interactions of Nbs with MelBSt and melibiose transport assays to determine the effects on MelBSt functions. We found that all selected Nbs showed partial to complete inhibitions of MelBSt transport activities, confirming their intracellular interactions. A group of Nbs (714, 725, and 733) was purified, and isothermal titration calorimetry measurements showed that their binding affinities were significantly inhibited by the substrate melibiose. When titrating melibiose to the MelBSt/Nb complexes, Nb also inhibited the sugar-binding. However, the Nb733/MelBSt complex retained binding to the coupling cation Na+ and also to the regulatory enzyme EIIAGlc of the glucose-specific phosphoenolpyruvate/sugar phosphotransferase system. Further, EIIAGlc/MelBSt complex also retained binding to Nb733 and formed a stable supercomplex. All data indicated that MelBSt trapped by Nbs retained its physiological functions and the trapped conformation is similar to that bound by the physiological regulator EIIAGlc. Therefore, these conformational Nbs can be useful tools for further structural, functional, and conformational analyses.


Subject(s)
Single-Domain Antibodies , Symporters , Single-Domain Antibodies/metabolism , Melibiose/metabolism , Symporters/metabolism , Ion Transport , Sodium/metabolism
2.
Evolution ; 76(12): 2811-2828, 2022 12.
Article in English | MEDLINE | ID: mdl-36181481

ABSTRACT

Understanding the basis of biological diversity remains a central problem in evolutionary biology. Using microbial systems, adaptive diversification has been studied in (a) spatially heterogeneous environments, (b) temporally segregated resources, and (c) resource specialization in a homogeneous environment. However, it is not well understood how adaptive diversification can take place in a homogeneous environment containing a single resource. Starting from an isogenic population of yeast Saccharomyces cerevisiae, we report rapid adaptive diversification, when propagated in an environment containing melibiose as the carbon source. The diversification is driven due to a public good enzyme α-galactosidase, which hydrolyzes melibiose into glucose and galactose. The diversification is driven by mutations at a single locus, in the GAL3 gene in the S. cerevisiae GAL/MEL regulon. We show that metabolic co-operation involving public resources could be an important mode of generating biological diversity. Our study demonstrates sympatric diversification of yeast starting from an isogenic population and provides detailed mechanistic insights into the factors and conditions responsible for generating and maintaining the population diversity.


Subject(s)
Melibiose , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Melibiose/metabolism , Galactose/metabolism , Genotype , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
3.
Int J Mol Sci ; 23(18)2022 Sep 06.
Article in English | MEDLINE | ID: mdl-36142128

ABSTRACT

Actinidia eriantha is a unique germplasm resource for kiwifruit breeding. Genetic diversity and nutrient content need to be evaluated prior to breeding. In this study, we looked at the metabolites of three elite A. eriantha varieties (MM-11, MM-13 and MM-16) selected from natural individuals by using a UPLC-MS/MS-based metabolomics approach and transcriptome, with a total of 417 metabolites identified. The biosynthesis and metabolism of phenolic acid, flavonoids, sugars, organic acid and AsA in A. eriantha fruit were further analyzed. The phenolic compounds accounted for 32.37% of the total metabolites, including 48 phenolic acids, 60 flavonoids, 7 tannins and 20 lignans and coumarins. Correlation analysis of metabolites and transcripts showed PAL (DTZ79_15g06470), 4CL (DTZ79_26g05660 and DTZ79_29g0271), CAD (DTZ79_06g11810), COMT (DTZ79_14g02670) and FLS (DTZ79_23g14660) correlated with polyphenols. There are twenty-three metabolites belonging to sugars, the majority being sucrose, glucose arabinose and melibiose. The starch biosynthesis-related genes (AeglgC, AeglgA and AeGEB1) were expressed at lower levels compared with metabolism-related genes (AeamyA and AeamyB) in three mature fruits of three varieties, indicating that starch was converted to soluble sugar during fruit maturation, and the expression level of SUS (DTZ79_23g00730) and TPS (DTZ79_18g05470) was correlated with trehalose 6-phosphate. The main organic acids in A. eriantha fruit are citric acid, quinic acid, succinic acid and D-xylonic acid. Correlation analysis of metabolites and transcripts showed ACO (DTZ79_17g07470) was highly correlated with citric acid, CS (DTZ79_17g00890) with oxaloacetic acid, and MDH1 (DTZ79_23g14440) with malic acid. Based on the gene expression, the metabolism of AsA acid was primarily through the L-galactose pathway, and the expression level of GMP (DTZ79_24g08440) and MDHAR (DTZ79_27g01630) highly correlated with L-Ascorbic acid. Our study provides additional evidence for the correlation between the genes and metabolites involved in phenolic acid, flavonoids, sugars, organic acid and AsA synthesis and will help to accelerate the kiwifruit molecular breeding approaches.


Subject(s)
Actinidia , Lignans , Actinidia/genetics , Actinidia/metabolism , Arabinose , Ascorbic Acid/metabolism , Chromatography, Liquid , Citric Acid/metabolism , Coumarins/metabolism , Fruit/genetics , Fruit/metabolism , Galactose/metabolism , Glucose/metabolism , Humans , Hydroxybenzoates , Lignans/metabolism , Melibiose/metabolism , Metabolomics , Oxaloacetates/metabolism , Phosphates/metabolism , Plant Breeding , Polyphenols/metabolism , Quinic Acid/metabolism , Starch/metabolism , Succinates/metabolism , Sucrose/metabolism , Tandem Mass Spectrometry , Tannins/metabolism , Transcriptome , Trehalose/metabolism
4.
Sci Rep ; 12(1): 7520, 2022 05 07.
Article in English | MEDLINE | ID: mdl-35525899

ABSTRACT

Melibiose-derived AGE (MAGE) is an advanced glycation end-product formed in vitro in anhydrous conditions on proteins and protein-free amino acids during glycation with melibiose. Our previous studies revealed the presence of MAGE antigen in the human body and tissues of several other species, including muscles, fat, extracellular matrix, and blood. MAGE is also antigenic and induces generation of anti-MAGE antibody. The aim of this paper was to identify the proteins modified by MAGE present in human body fluids, such as serum, plasma, and peritoneal fluids. The protein-bound MAGE formed in vivo has been isolated from human blood using affinity chromatography on the resin with an immobilized anti-MAGE monoclonal antibody. Using mass spectrometry and immunochemistry it has been established that MAGE epitope is present on several human blood proteins including serum albumin, IgG, and IgA. In serum of diabetic patients, mainly the albumin and IgG were modified by MAGE, while in healthy subjects IgG and IgA carried this modification, suggesting the novel AGE can impact protein structure, contribute to auto-immunogenicity, and affect function of immunoglobulins. Some proteins in peritoneal fluid from cancer patients modified with MAGE were also observed and it indicates a potential role of MAGE in cancer.


Subject(s)
Body Fluids , Melibiose , Body Fluids/metabolism , Glycation End Products, Advanced/metabolism , Humans , Immunoglobulin A , Immunoglobulin G , Melibiose/metabolism , Serum Albumin/analysis
5.
J Mol Biol ; 434(12): 167598, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35461877

ABSTRACT

Cation selectivity and coupling are important attributes of cation-coupled symporters. Salmonella typhimurium melibiose permease (MelBSt) catalyzes the co-transport of galactosides with cations (H+, Li+, or Na+). 3-D crystal structures of MelBSt have revealed the molecular recognition for sugar substrates, but the cation binding and coupling mechanisms have not been defined to atomic levels. In its human homolog MFSD2A, a lethal mutation was mapped at its Na+-binding pocket; however, none of the structures in this subfamily resolved its cation binding. In this study, molecular dynamics simulations reveal the binding interactions of Na+ and Li+ with MelBSt. Interestingly, Thr121, the lethal mutation position in MFSD2A, forms stable interaction with Na+ but is at a distance from Li+. Most mutations among 11 single-site Thr121 mutants of MelBSt exhibited little effects on the galactoside binding, but largely altered the cation selectivity with severe inhibitions on Na+ binding. Few mutants (Pro and Ala) completely lost the Na+ binding and Na+-coupled transport, but their Li+ or H+ modes of activity were largely retained. It can be concluded that Thr121 is necessary for Na+ binding, but not required for the binding of H+ or Li+, so a subset of the Na+-binding pocket is enough for Li+ binding. In addition, the protein stability for some mutants can be only retained in the presence of Li+, but not by Na+ due to the lack of affinity. This finding, together with other identified thermostable mutants, supports that the charge balance of the cation-binding site plays an important role in MelBSt protein stability.


Subject(s)
Bacterial Proteins , Salmonella typhimurium , Symporters , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cations/metabolism , Humans , Lithium/metabolism , Melibiose/metabolism , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Sodium/metabolism , Symporters/chemistry , Symporters/genetics , Symporters/metabolism
6.
J Clin Microbiol ; 60(1): e0153021, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34586892

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) O80:H2, belonging to sequence type ST301, is among the main causes of hemolytic and uremic syndrome in Europe, a major concern in young children. Aside from the usual intimin and Shiga toxin virulence factors (VFs), this emerging serotype possesses a mosaic plasmid combining extra-intestinal VF- and antibiotic resistance-encoding genes. This hybrid pathotype can be involved in invasive infections, a rare occurrence in EHEC infections. Here, we aimed to optimize its detection, improve its clinical diagnosis, and identify its currently unknown reservoir. O80:H2 EHEC strains isolated in France between 2010 and 2018 were phenotypically and genetically analyzed and compared with non-O80 strains. The specificity and sensitivity of a PCR test and a culture medium designed, based on the molecular and phenotypic signatures of O80:H2 EHEC, were assessed on a collection of strains and stool samples. O80:H2 biotype analysis showed that none of the strains (n = 137) fermented melibiose versus 5% of non-O80 EHEC (n = 19/352). This loss of metabolic function is due to deletion of the entire melibiose operon associated with the insertion of a 70-pb sequence (70mel), a genetic scar shared by all ST301 strains. This metabolic hallmark was used to develop a real-time PCR test (100% sensitivity, 98.3% specificity) and a melibiose-based culture medium including antibiotics, characterized by 85% specificity and sensitivity for clinical specimens. These new tools may facilitate the diagnosis of this atypical clone, help the food industry to identify the reservoir and improve our epidemiological knowledge of this threatening and emerging clone.


Subject(s)
Drug Resistance, Bacterial , Enterohemorrhagic Escherichia coli , Hemolytic-Uremic Syndrome , Anti-Bacterial Agents/pharmacology , Child , Child, Preschool , Culture Media , Drug Resistance, Bacterial/genetics , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/isolation & purification , Enterohemorrhagic Escherichia coli/metabolism , Fermentation , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/microbiology , Humans , Melibiose/metabolism
7.
Commun Biol ; 4(1): 931, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34341464

ABSTRACT

Major facilitator superfamily_2 transporters are widely found from bacteria to mammals. The melibiose transporter MelB, which catalyzes melibiose symport with either Na+, Li+, or H+, is a prototype of the Na+-coupled MFS transporters, but its sugar recognition mechanism has been a long-unsolved puzzle. Two high-resolution X-ray crystal structures of a Salmonella typhimurium MelB mutant with a bound ligand, either nitrophenyl-α-D-galactoside or dodecyl-ß-D-melibioside, were refined to a resolution of 3.05 or 3.15 Å, respectively. In the substrate-binding site, the interaction of both galactosyl moieties on the two ligands with MelBSt are virturally same, so the sugar specificity determinant pocket can be recognized, and hence the molecular recognition mechanism for sugar binding in MelB has been deciphered. The conserved cation-binding pocket is also proposed, which directly connects to the sugar specificity pocket. These key structural findings have laid a solid foundation for our understanding of the cooperative binding and symport mechanisms in Na+-coupled MFS transporters, including eukaryotic transporters such as MFSD2A.


Subject(s)
Bacterial Proteins/chemistry , Crystallography, X-Ray , Melibiose/metabolism , Salmonella typhimurium/metabolism , Symporters/chemistry
8.
Biomolecules ; 11(4)2021 04 10.
Article in English | MEDLINE | ID: mdl-33920190

ABSTRACT

The patho-mechanism of changes in the thyroid gland, including carcinogenesis, is a complex process, which involves oxidative stress. The goal of our investigation was to verify the extent of stress in the thyroid gland related to glycation. The study samples were comprised of blood sera, thyroid, and adipose tissue sections probed from 37 patients diagnosed with thyroid cancers and goiter. Using immuno-enzymatic and fluorometric assays we analyzed the content of advanced glycation end-products (AGEs), pentosidine, receptors for advanced glycation end-products (RAGE), scavenger receptor class (SR)-A, SR-B, glutathione, malondialdehyde and nitric oxide synthase. In addition to classic AGEs, a recent study detected the melibiose-derived glycation (MAGE) product. We demonstrated the presence of AGEs, MAGE and their receptors of the RAGE and SR-A. In addition, in the control samples of thyroid glands SR-B groups were detected as well as of pathological groups without noticeable tendency to antigen concentration in the area of carcinogenesis. Fluorescent AGEs correlate positively with glutathione, which supports the assumption that glycation stress leads to augmentation of oxidative stress and increase of the intensity of antioxidant mechanisms.


Subject(s)
Glycation End Products, Advanced/metabolism , Oxidative Stress , Thyroid Diseases/metabolism , Thyroid Gland/metabolism , Adult , Aged , Female , Humans , Male , Melibiose/metabolism , Middle Aged , Nitric Oxide Synthase/metabolism , Receptor for Advanced Glycation End Products/metabolism , Thyroid Diseases/pathology , Thyroid Gland/pathology
9.
Sci Rep ; 11(1): 2940, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536563

ABSTRACT

Non-enzymatic modification of proteins by carbohydrates, known as glycation, leads to generation of advanced glycation end-products (AGEs). In our study we used in vitro generated AGEs to model glycation in vivo. We discovered in vivo analogs of unusual melibiose-adducts designated MAGEs (mel-derived AGEs) synthesized in vitro under anhydrous conditions with bovine serum albumin and myoglobin. Using nuclear magnetic resonance spectroscopy we have identified MAGEs as a set of isomers, with open-chain and cyclic structures, of the fructosamine moiety. We generated a mouse anti-MAGE monoclonal antibody and show for the first time that the native and previously undescribed analogous glycation product exists in living organisms and is naturally present in tissues of both invertebrates and vertebrates, including humans. We also report MAGE cross-reactive auto-antibodies in patients with diabetes. We anticipate our approach for modeling glycation in vivo will be a foundational methodology in cell biology. Further studies relevant to the discovery of MAGE may contribute to clarifying disease mechanisms and to the development of novel therapeutic options for diabetic complications, neuropathology, and cancer.


Subject(s)
Diabetes Mellitus/immunology , Dietary Carbohydrates/immunology , Epitopes/immunology , Glycation End Products, Advanced/immunology , Melibiose/immunology , Animals , Autoantibodies/blood , Autoantibodies/immunology , Cross Reactions , Diabetes Mellitus/blood , Diabetes Mellitus/metabolism , Dietary Carbohydrates/metabolism , Glycation End Products, Advanced/metabolism , Glycosylation , Humans , Melibiose/metabolism , Mice
10.
Yeast ; 38(1): 117-126, 2021 01.
Article in English | MEDLINE | ID: mdl-33141945

ABSTRACT

In many yeast species, the three genes at the centre of the galactose catabolism pathway, GAL1, GAL10 and GAL7, are neighbours in the genome and form a metabolic gene cluster. We report here that some yeast strains in the genus Torulaspora have much larger GAL clusters that include genes for melibiase (MEL1), galactose permease (GAL2), glucose transporter (HGT1), phosphoglucomutase (PGM1) and the transcription factor GAL4, in addition to GAL1, GAL10, and GAL7. Together, these eight genes encode almost all the steps in the pathway for catabolism of extracellular melibiose (a disaccharide of galactose and glucose). We show that a progenitor 5-gene cluster containing GAL 7-1-10-4-2 was likely present in the common ancestor of Torulaspora and Zygotorulaspora. It added PGM1 and MEL1 in the ancestor of most Torulaspora species. It underwent further expansion in the T. pretoriensis clade, involving the fusion of three progenitor clusters in tandem and the gain of HGT1. These giant GAL clusters are highly polymorphic in structure, and subject to horizontal transfers, pseudogenization and gene losses. We identify recent horizontal transfers of complete GAL clusters from T. franciscae into one strain of T. delbrueckii, and from a relative of T. maleeae into one strain of T. globosa. The variability and dynamic evolution of GAL clusters in Torulaspora indicates that there is strong natural selection on the GAL pathway in this genus.


Subject(s)
Galactose/metabolism , Genes, Fungal , Melibiose/metabolism , Metabolic Networks and Pathways/genetics , Multigene Family , Torulaspora/genetics , Torulaspora/metabolism
11.
Methods Mol Biol ; 2132: 257-266, 2020.
Article in English | MEDLINE | ID: mdl-32306333

ABSTRACT

LecA/PA-IL (Pfam PF07828) is a soluble galactose-binding lectin from bacterium Pseudomonas aeruginosa. The lectin is specific for α-galactose present on glycosphingolipids of the globoside family and has therefore been proposed to play a role in cell adhesion and in internalization of bacteria in epithelial cells. The lectin has also direct toxic activity. Search for high-affinity inhibitors can be performed on the recombinant lectin, with use of surface plasmon resonance assays and structural studies.


Subject(s)
Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Melibiose/metabolism , Pseudomonas aeruginosa/metabolism , Bacterial Adhesion , Binding Sites , Crystallography, X-Ray , Galectins/chemistry , Galectins/metabolism , Globosides/metabolism , Models, Molecular , Protein Conformation , Pseudomonas aeruginosa/chemistry , Surface Plasmon Resonance
12.
Int J Biol Macromol ; 152: 727-734, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32092418

ABSTRACT

Bacteroides thetaiotaomicron (B. thetaiotaomicron), which resides in the human intestinal tract, has a number of carbohydrate enzymes, including glycoside hydrolase (GH) family 97. Only a few GH 97 enzymes have been characterized to date. In this study, a novel α-galactosidase (Bt_3294) was cloned from B. thetaiotaomicron, expressed in Escherichia coli, and purified using affinity chromatography. This novel enzyme showed optimal activity at 60 °C and pH 7.0. Enzyme activity was reduced by 94.4% and 95.7% in the presence of 5 mM Ca2+ and Fe2+, respectively. It is interesting that Bt_3294 specifically hydrolyzed shorter α-galactosyl oligosaccharides, such as melibiose and raffinose. The D-values of Bt_3294 at 40 °C and 50 °C were about 107 and 6 min, respectively. After immobilization of Bt_3294, the D-values at 40 °C and 50 °C were about 37.6 and 29.7 times higher than those of the free enzyme, respectively. As a practical application, the immobilized Bt_3294 was used to hydrolyze raffinose family oligosaccharides (RFOs) in soy milk, decreasing the RFOs by 98.9%.


Subject(s)
Bacteroides thetaiotaomicron/metabolism , Enzymes, Immobilized/metabolism , Glycoside Hydrolases/metabolism , alpha-Galactosidase/metabolism , Hydrogen-Ion Concentration , Melibiose/metabolism , Oligosaccharides/metabolism , Raffinose/metabolism , Soy Milk/metabolism
13.
Protein Expr Purif ; 170: 105574, 2020 06.
Article in English | MEDLINE | ID: mdl-31978534

ABSTRACT

BACKGROUND: Lectins are known to possess interesting biological properties such as anti microbial, nematicidal, anti tumor and anti viral activities. Lantana camara from verbenaceae family is a medicinal plant known for possessing anti oxidant and anticancer activities. Since anticancer activity is reported in plant lectins, leaves of Lantana camara was used to check the presence of lectin. METHODS AND RESULTS: Here we report the purification, characterization and biological properties of a lectin from Lantana camara (LCL) leaves. LCL was purified by ion exchange chromatography on CM-cellulose column followed by affinity chromatography on mucin coupled Sepharose 4B column and gel filtration chromatography on Superdex G75 column. LCL is a glycoprotein with 10% of the carbohydrate and is blood group non specific. SDS-PAGE analysis of affinity purified LCL showed two proteins with apparent molecular weight of 14.49 kDa and 17.4 kDa which were subsequently separated by Gel filtration chromatography on Superdex G75 column. Hapten inhibition studies of LCL revealed its highest affinity for Chitin, Milibiose, α-D-Methyl galactopyranoside and glycoproteins like mucin, asialomucin. LCL showed strong binding to human colon adenocarcinoma HT29 cells with MFI of 242 which was effectively blocked by 68.1 and 62.5% by both mucin and milibiose. LCL showed dose and time dependent growth inhibitory effects on HT29 cells with IC50 of 3.75  µg/ml at 48 h. LCL has potent antibacterial and anti fungal activity. CONCLUSION: LCL can be explored for its clinical potential.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Antineoplastic Agents/pharmacology , Lantana/chemistry , Plant Lectins/pharmacology , Anti-Bacterial Agents/isolation & purification , Antifungal Agents/isolation & purification , Antineoplastic Agents/isolation & purification , Chitin/chemistry , Chitin/metabolism , Chromatography, Affinity , Chromatography, Gel , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Fungi/drug effects , Fungi/growth & development , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , HT29 Cells , Humans , Melibiose/chemistry , Melibiose/metabolism , Methylgalactosides/chemistry , Methylgalactosides/metabolism , Microbial Sensitivity Tests , Mucins/chemistry , Mucins/metabolism , Plant Extracts/chemistry , Plant Leaves/chemistry , Plant Lectins/isolation & purification , Plants, Medicinal , Protein Binding
14.
Biosci Biotechnol Biochem ; 84(3): 507-517, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31718466

ABSTRACT

Membrane-bound quinoprotein glucose dehydrogenase from acetic acid bacteria produces lactobionic acid by the oxidation of lactose. Its enzymatic activity on lactose and maltose is much lower than that on D-glucose. For that reason, the activity of the enzyme on disaccharides has been considered low. In this study, we show that the isomaltose-oxidizing activity of acetic acid bacteria is much higher than their lactose-oxidizing activity. In addition to isomaltose, the enzyme oxidized gentiobiose and melibiose to the same extent. According to the characteristics of the isomaltose-oxidizing activity and investigations using dehydrogenase-deficient mutant bacteria, we identified the responsible enzyme as membrane-bound quinoprotein glucose dehydrogenase.Abbreviations: AAB: acetic acid bacteria; m-GDH: membrane-bound quinoprotein glucose dehydrogenase; DCIP: 2,6-dichlorophenolindophenol; DP: degree of polymerization; HPAEC-PAD: high-performance anion-exchange chromatography with pulsed amperometric detection; NMR: nuclear magnetic resonance; TLC: thin layer chromatography; COSY: correlation spectroscopy.


Subject(s)
Acetic Acid/metabolism , Bacteria/metabolism , Disaccharides/metabolism , Glucose 1-Dehydrogenase/metabolism , Isomaltose/metabolism , Melibiose/metabolism , Oxidation-Reduction
15.
Mol Biotechnol ; 61(9): 633-649, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31177409

ABSTRACT

Galactose oxidase catalyzes a two-electron oxidation, mainly from the C6 hydroxyl group of D-galactose, with the concomitant reduction of water to hydrogen peroxide. This enzyme is secreted by Fusarium species and has several biotechnological applications. In this study, a screening of galactose oxidase production among species of the Fusarium fujikuroi species complex demonstrated Fusarium subglutinans to be the main producer. The truncated F. subglutinans gaoA gene coding for the mature galactose oxidase was expressed from the prokaryotic vector pTrcHis2B in the E. coli Rosetta™ (DE3) strain. The purified recombinant enzyme presented temperature and pH optima of 30 °C and 7.0, respectively, KM of 132.6 ± 18.18 mM, Vmax of 3.2 ± 0.18 µmol of H2O2/min, kcat of 12,243 s-1, and a catalytic efficiency (kcat/KM) of 9.2 × 104 M-1 s-1. In the presence of 50% glycerol, the enzyme showed a T50 of 59.77 °C and was stable for several hours at pH 8.0 and 4 °C. Besides D-(+)-galactose, the purified enzyme also acted against D-(+)-raffinose, α-D-(+)-melibiose, and methyl-α-D-galactopyranoside, and was strongly inhibited by SDS. Although the F. subglutinans gaoA gene was successfully expressed in E. coli, its endogenous transcription was not confirmed by RT-PCR.


Subject(s)
Fusarium/enzymology , Galactose Oxidase/metabolism , Galactose/chemistry , Recombinant Proteins/metabolism , Amino Acid Sequence , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Fusarium/chemistry , Galactose/metabolism , Galactose Oxidase/chemistry , Galactose Oxidase/genetics , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hydrogen-Ion Concentration , Melibiose/chemistry , Melibiose/metabolism , Methylgalactosides/chemistry , Methylgalactosides/metabolism , Models, Molecular , Oxidation-Reduction , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Raffinose/chemistry , Raffinose/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Temperature
16.
J Bacteriol ; 201(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31138628

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 (P msmR ), which is negatively regulated by MsmR. The activity of P msmR was induced in the presence of melibiose and raffinose. MsmR is a transcriptional repressor that binds to two binding sites at P msmR 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 melREDCAIMPORTANCEBacillus 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.


Subject(s)
Bacillus subtilis/genetics , Bacterial Proteins/genetics , Oligosaccharides/metabolism , Operon , Raffinose/metabolism , Bacillus subtilis/metabolism , Galactosides/metabolism , Melibiose/metabolism , Sucrose/metabolism , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
17.
BMC Biol ; 16(1): 85, 2018 08 03.
Article in English | MEDLINE | ID: mdl-30075778

ABSTRACT

BACKGROUND: Membrane lipids play critical roles in the structure and function of membrane-embedded transporters. Salmonella typhimurium MelB (MelBSt) is a symporter coupling melibiose translocation with a cation (Na+, Li+, or H+). We present an extensive study on the effects of specific phospholipids on the structure of MelBSt and the melibiose transport catalyzed by this protein. RESULTS: Lipidomic analysis and thin-layer chromatography (TLC) experiments reveal that at least one phosphatidylethanolamine (PE) and one phosphatidylglycerol (PG) molecule associate with MelBSt at high affinities. Solid-state nuclear magnetic resonance (ssNMR) spectroscopy experiments confirmed the presence of lipid tails and glycerol backbones that co-purified with MelBSt; headgroups of PG were also observed. Studies with lipid-engineered strains, including PE-deficient, cardiolipin (CL)- and PG-deficient, or CL-deficient strains, show that lack of PE or PG, however not CL, largely inhibits both H+- and Na+-coupled melibiose active transport to different extents. Interestingly, neither the co-substrate binding (melibiose or Na+) nor MelBSt folding and stability are affected by changing lipid compositions. Remarkably, the delipidated MelBSt with only 2-3 bound lipids, regardless of the headgroup species, also exhibits unchanged melting temperature values as shown by circular dichroism spectroscopy. CONCLUSIONS: (1) Lipid tails and glycerol backbones of interacting PE and PG may contribute to the stability of the structure of MelBSt. (2) The headgroups of PE and PG, but not of CL, play important roles in melibiose transport; however, lipid headgroups do not modulate the folding and stability of MelBSt.


Subject(s)
Bacterial Proteins/genetics , Melibiose/metabolism , Salmonella typhimurium/genetics , Symporters/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cardiolipins/chemistry , Cardiolipins/metabolism , Melibiose/chemistry , Phosphatidylethanolamines/chemistry , Phosphatidylglycerols/chemistry , Salmonella typhimurium/metabolism , Symporters/chemistry , Symporters/metabolism
18.
Anaerobe ; 52: 100-110, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29935270

ABSTRACT

The aim of the study was to investigate the metabolism of non-digestible oligo- and polysaccharides by fecal microbiota, using isothermal microcalorimetry. The five tested substrates were raffinose, melibiose, a mixture of oligo- and polysaccharides produced from raffinose by levansucrase, levan synthesized from raffinose, and levan from timothy grass. Two inocula were comprised of pooled fecal samples from overweight or normal-weight children, from healthy adult volunteers and a pure culture of Bacteroides thetaiotaomicron as a reference bacterium for colon microbiota. The growth was analyzed based on the heat evolution curves, and the production of organic acids and gases. Taxonomic profiles of the microbiota were assessed by 16S rDNA sequencing. Raffinose and melibiose promoted the growth of bifidobacteria in all fecal pools. Several pool-specific substrate-related responses to raffinose and melibiose were revealed. Lactate-producing bacteria (Streptococcus and Enterococcus) became enriched in the pool of overweight children resulting in lactic acid as the major fermentation product on short saccharides. Acetic and butyric acids were prevalent at fermentation in the normal-weight pool coinciding with the enrichment of Catenibacterium. In the adult pool, the specific promotion of Bacteroides and Lachnospiraceae by levans was disclosed. In the fecal pool of normal-weight children, levans stimulated the growth of Senegalimassilia and Lachnoclostridium and this particular pool also showed the highest maximum heat production rate at levan fermentation. Levans and raffinose-derived oligosaccharides, but not raffinose and melibiose were completely fermented by a pure culture of Bacteroides thetaiotaomicron. The main conclusion from the study is that fecal microbiota of normal and overweight children have different compositions and they respond in specific manners to non-digestible oligo- and polysaccharides: raffinose, melibiose, raffinose-derived oligosaccharides and levans. The potential of the tested saccharides to support a healthy balance of colon microbiota requires further studies.


Subject(s)
Bacteria/metabolism , Feces/microbiology , Fructans/metabolism , Gastrointestinal Microbiome , Melibiose/metabolism , Overweight/microbiology , Raffinose/metabolism , Adolescent , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Child , Child, Preschool , Female , Fermentation , Humans , Male
19.
J Gen Physiol ; 149(11): 1029-1039, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-29054867

ABSTRACT

The Na+-coupled melibiose symporter MelB, which can also be coupled to H+ or Li+ transport, is a prototype for the glycoside-pentoside-hexuronide:cation symporter family. Although the 3-D x-ray crystal structure of Salmonella typhimurium MelB (MelBSt) has been determined, the symport mechanisms for the obligatory coupled transport are not well understood. Here, we apply isothermal titration calorimetry to determine the energetics of Na+ and melibiose binding to MelBSt, as well as protonation of this transporter. Studies of the thermodynamic cycle for the formation of the Na+-MelBSt-melibiose ternary complex at pH 7.45 reveal that the binding of Na+ and melibiose is cooperative. The binding affinity for one substrate (Na+ or melibiose) is increased by the presence of the other by about eightfold. The coupling free energies (ΔΔG) of either substrate binding are ∼5 kJ/mol, and binding of both substrates releases a free energy of ∼35 kJ/mol. Measurements of the Na+-binding enthalpy at three different pH values, including the pKa value of MelB, indicate that the binding of one Na+ displaces one H+ per MelBSt molecule. In addition, the absolute dissociation constants for Na+ and H+, determined by competitive binding, show that MelBSt is selective for H+ over Na+ by ∼1,000-fold at a pKa of 6.25. Thus, the Na+ coupling in MelBSt is based not on ion selectivity but on ion concentrations and competitive binding because of a much higher Na+ concentration under physiological conditions. Such a selectivity feature seems to be common for membrane transport proteins that can bind both H+ and Na+ at a common site.


Subject(s)
Symporters/metabolism , Hydrogen/metabolism , Melibiose/metabolism , Molecular Dynamics Simulation , Protein Binding , Salmonella typhimurium/metabolism , Sodium/metabolism , Symporters/chemistry
20.
J Biosci Bioeng ; 124(3): 289-295, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28479043

ABSTRACT

The putative gene cluster involved in the degradation of the raffinose family oligosaccharides (RFO) was identified in Caldicellulosiruptor bescii. Within the cluster, the gene encoding a putative α-galactosidase (CbAga36) was cloned and expressed in Escherichia coli. Size exclusion chromatography of the purified rCbAga36 indicated that the native form was a tetramer. Its primary sequence was similar to the family of glycoside hydrolase 36. The purified recombinant CbAga36 (rCbAga36) was optimally active at pH 5.0 and 70°C and had a half-life of 15 h and 10 h at 70°C and 80°C, respectively. rCbAga36 showed high activity with the artificial substrate (p-nitrophenyl α-d-galactopyranoside, pNPαGal) exhibiting lower Km and higher kcat than natural substrates such as melibiose and raffinose. Although rCbAga36 demonstrated preferential activity toward the hydrolysis of RFO such as raffinose and stachyose, it did not degrade the polymeric galactomannans. Our results imply that CbAga36 may play a role in the degradation of RFO, transported into the cytoplasm via a transporter into galactose, which is further utilized as an energy source in C. bescii. Furthermore, its ability to synthesize novel oligosaccharides by transglycosylation renders this enzyme potentially useful for the production of dietary oligosaccharides with novel function.


Subject(s)
Gram-Positive Bacteria/enzymology , alpha-Galactosidase/metabolism , Enzyme Stability , Escherichia coli/genetics , Galactose/metabolism , Glycolysis , Glycosides/metabolism , Gram-Positive Bacteria/genetics , Half-Life , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Melibiose/metabolism , Oligosaccharides/metabolism , Raffinose/metabolism , Substrate Specificity , alpha-Galactosidase/genetics , alpha-Galactosidase/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...