Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Agric Food Chem ; 72(18): 10497-10505, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38659290

ABSTRACT

Despite their broad application potential, the widespread use of ß-1,3-glucans has been hampered by the high cost and heterogeneity associated with current production methods. To address this challenge, scalable and economically viable processes are needed for the production of ß-1,3-glucans with tailorable molecular mass distributions. Glycoside phosphorylases have shown to be promising catalysts for the bottom-up synthesis of ß-1,3-(oligo)glucans since they combine strict regioselectivity with a cheap donor substrate (i.e., α-glucose 1-phosphate). However, the need for an expensive priming substrate (e.g., laminaribiose) and the tendency to produce shorter oligosaccharides still form major bottlenecks. Here, we report the discovery and application of a thermostable ß-1,3-oligoglucan phosphorylase originating from Anaerolinea thermophila (AtßOGP). This enzyme combines a superior catalytic efficiency toward glucose as a priming substrate, high thermostability, and the ability to synthesize high molecular mass ß-1,3-glucans up to DP 75. Coupling of AtßOGP with a thermostable variant of Bifidobacterium adolescentis sucrose phosphorylase enabled the efficient production of tailorable ß-1,3-(oligo)glucans from sucrose, with a near-complete conversion of >99 mol %. This cost-efficient process for the conversion of renewable bulk sugar into ß-1,3-(oligo)glucans should facilitate the widespread application of these versatile functional fibers across various industries.


Subject(s)
Bacterial Proteins , Enzyme Stability , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , beta-Glucans/chemistry , beta-Glucans/metabolism , Bifidobacterium adolescentis/enzymology , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/chemistry , Bifidobacterium adolescentis/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/metabolism , Glucosyltransferases/genetics , Substrate Specificity , Phosphorylases/metabolism , Phosphorylases/chemistry , Phosphorylases/genetics , Clostridiales/enzymology , Clostridiales/genetics , Clostridiales/chemistry , Biocatalysis , Hot Temperature
2.
Appl Environ Microbiol ; 90(2): e0201423, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38294252

ABSTRACT

Bifidobacteria are among the first microbial colonizers of the human gut, being frequently associated with human health-promoting activities. In the current study, an in silico methodology based on an ecological and phylogenomic-driven approach allowed the selection of a Bifidobacterium adolescentis prototype strain, i.e., B. adolescentis PRL2023, which best represents the overall genetic content and functional features of the B. adolescentis taxon. Such features were confirmed by in vitro experiments aimed at evaluating the ability of this strain to survive in the gastrointestinal tract of the host and its ability to interact with human intestinal cells and other microbial gut commensals. In this context, co-cultivation of B. adolescentis PRL2023 and several gut commensals revealed various microbe-microbe interactions and indicated co-metabolism of particular plant-derived glycans, such as xylan.IMPORTANCEThe use of appropriate bacterial strains in experimental research becomes imperative in order to investigate bacterial behavior while mimicking the natural environment. In the current study, through in silico and in vitro methodologies, we were able to identify the most representative strain of the Bifidobacterium adolescentis species. The ability of this strain, B. adolescentis PRL2023, to cope with the environmental challenges imposed by the gastrointestinal tract, together with its ability to switch its carbohydrate metabolism to compete with other gut microorganisms, makes it an ideal choice as a B. adolescentis prototype and a member of the healthy microbiota of adults. This strain possesses a genetic blueprint appropriate for its exploitation as a candidate for next-generation probiotics.


Subject(s)
Bifidobacterium adolescentis , Gastrointestinal Microbiome , Probiotics , Adult , Humans , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/metabolism , Gastrointestinal Microbiome/genetics , Bifidobacterium/genetics , Bifidobacterium/metabolism , Phylogeny
3.
Org Biomol Chem ; 21(11): 2307-2311, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36857722

ABSTRACT

Mutation Q345F in sucrose phosphorylase from Bifidobacterium adolescentis (BaSP) has shown to allow efficient (+)-catechin glucosylation yielding a regioisomeric mixture: (+)-catechin-3'-O-α-D-glucopyranoside, (+)-catechin-5-O-α-D-glucopyranoside and (+)-catechin-3',5-O-α-D-diglucopyranoside with a ratio of 51 : 25 : 24. Here, we efficiently increased the control of (+)-catechin glucosylation regioselectivity with a new variant Q345F/P134D. The same products were obtained with a ratio of 82 : 9 : 9. Thanks to bioinformatics models, we successfully explained the glucosylation favoured at the OH-3' position due to the mutation P134D.


Subject(s)
Bifidobacterium adolescentis , Catechin , Bifidobacterium adolescentis/genetics , Glucosyltransferases/genetics , Mutation
4.
Nutrients ; 14(12)2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35745208

ABSTRACT

The prevalence of diabetes mellitus is increasing globally. Probiotics have been shown to be an effective intervention for diabetes. This study focused on the relieving effects and possible mechanisms of 16 strains of two dominant Bifidobacterium species (B. bifidum and B. adolescentis, which exist in the human gut at different life stages) on type 2 diabetes (T2D). The results indicated that more B. adolescentis strains appeared to be superior in alleviating T2D symptoms than B. bifidum strains. This effect was closely related to the ability of B. adolescentis to restore the homeostasis of the gut microbiota, increase the abundance of short-chain fatty acid-producing flora, and alleviate inflammation in mice with T2D. In addition, compared with B. bifidum, B. adolescentis had a higher number of core genes, and these genes were more evolutionarily stable, including unique environmental tolerance, carbon and nitrogen utilization genes, and a blood sugar regulation gene, glgP. This may be one of the reasons why B. adolescentis is more likely to colonize in the adult gut and show a superior ability to relieve T2D. This study provides insights into future studies aimed at investigating probiotics for the treatment of metabolic diseases.


Subject(s)
Bifidobacterium adolescentis , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Probiotics , Animals , Bifidobacterium/metabolism , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/therapy , Feces/microbiology , Gastrointestinal Microbiome/physiology , Mice , Probiotics/therapeutic use
5.
Microb Cell Fact ; 21(1): 33, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35255900

ABSTRACT

BACKGROUND: Bifidobacteria are gram-positive, probiotic, and generally regarded as safe bacteria. Techniques such as transformation, gene knockout, and heterologous gene expression have been established for Bifidobacterium, indicating that this bacterium can be used as a cell factory platform. However, there are limited previous reports in this field, likely because of factors such as the highly anaerobic nature of this bacterium. Bifidobacterium adolescentis is among the most oxygen-sensitive Bifidobacterium species. It shows strain-specific gamma-aminobutyric acid (GABA) production. GABA is a potent bioactive compound with numerous physiological and psychological functions. In this study, we investigated whether B. adolesentis could be used for mass production of GABA. RESULTS: The B. adolescentis 4-2 strain isolated from a healthy adult human produced approximately 14 mM GABA. It carried gadB and gadC, which encode glutamate decarboxylase and glutamate GABA antiporter, respectively. We constructed pKKT427::Pori-gadBC and pKKT427::Pgap-gadBC plasmids carrying gadBC driven by the original gadB (ori) and gap promoters, respectively. Recombinants of Bifidobacterium were then constructed. Two recombinants with high production abilities, monitored by two different promoters, were investigated. GABA production was improved by adjusting the fermentation parameters, including the substrate concentration, initial culture pH, and co-factor supplementation, using response surface methodology. The optimum initial cultivation pH varied when the promoter region was changed. The ori promoter was induced under acidic conditions (pH 5.2:4.4), whereas the constitutive gap promoter showed enhanced GABA production at pH 6.0. Fed-batch fermentation was used to validate the optimum fermentation parameters, in which approximately 415 mM GABA was produced. The conversion ratio of glutamate to GABA was 92-100%. CONCLUSION: We report high GABA production in recombinant B. adolescentis. This study provides a foundation for using Bifidobacterium as a cell factory platform for industrial production of GABA.


Subject(s)
Bifidobacterium adolescentis , Bifidobacterium/genetics , Bifidobacterium/metabolism , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/metabolism , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Humans , gamma-Aminobutyric Acid
6.
Mol Biotechnol ; 64(1): 75-89, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34542815

ABSTRACT

Xylooligosaccharides having various degrees of polymerization such as xylobiose, xylotriose, and xylotetraose positively affect human health by interacting with gut proteins. The present study aimed to identify proteins present in gut microflora, such as xylosidase, xylulokinase, etc., with the help of retrieved whole-genome annotations and find out the mechanistic interactions of those with the above substrates. The 3D structures of proteins, namely Endo-1,4-beta-xylanase B (XynB) from Lactobacillus brevis and beta-D-xylosidase (Xyl3) from Bifidobacterium adolescentis, were computationally predicted and validated with the help of various bioinformatics tools. Molecular docking studies identified the effectual binding of these proteins to the xylooligosaccharides, and the stabilities of the best-docked complexes were analyzed by molecular dynamic simulation. The present study demonstrated that XynB and Xyl3 showed better effectual binding toward Xylobiose with the binding energies of - 5.96 kcal/mol and - 4.2 kcal/mol, respectively. The interactions were stabilized by several hydrogen bonding having desolvation energy (- 6.59 and - 7.91). The conformational stabilities of the docked complexes were observed in the four selected complexes of XynB-xylotriose, XynB-xylotetraose, Xyl3-xylobiose, and Xyn3-xylotriose by MD simulations. This study showed that the interactions of these four complexes are stable, which means they have complex metabolic activities among each other. Extending these studies of understanding, the interaction between specific probiotics enzymes and their ligands can explore the detailed design of synbiotics in the future.


Subject(s)
Bifidobacterium adolescentis/metabolism , Glucuronates/metabolism , Levilactobacillus brevis/metabolism , Oligosaccharides/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bifidobacterium adolescentis/genetics , Computational Biology , Disaccharides/chemistry , Disaccharides/metabolism , Endo-1,4-beta Xylanases/chemistry , Endo-1,4-beta Xylanases/genetics , Genome, Bacterial/genetics , Glucuronates/chemistry , Humans , Levilactobacillus brevis/genetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Oligosaccharides/chemistry , Probiotics/metabolism , Trisaccharides/chemistry , Trisaccharides/metabolism , Xylosidases/chemistry , Xylosidases/genetics
7.
Nutrients ; 13(3)2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33801119

ABSTRACT

The incidence of obesity, which is closely associated with the gut microbiota and chronic inflammation, has rapidly increased in the past 40 years. Therefore, the probiotic-based modification of the intestinal microbiota composition has been developed as a strategy for the treatment of obesity. In this study, we selected four Bifidobacterium adolescentis strains isolated from the feces of newborn and elderly humans to investigate whether supplementation with B. adolescentis of various origins could alleviate obesity in mice. Male C57BL/6J mice fed a high-fat diet (HFD, 60% energy as fat) received one of the following 14-week interventions: (i) B. adolescentis N4_N3, (ii) B. adolescentis Z25, (iii) B. adolescentis 17_3, (iv) B. adolescentis 2016_7_2, and (v) phosphate-buffered saline. The metabolic parameters, thermogenesis, and immunity of all treated mice were measured. Cecal and colonic microbial profiles were determined by 16S rRNA gene sequencing. Intestinal concentrations of short-chain fatty acids (SCFAs) were measured by gas chromatography-mass spectrometry (GC-MS). The B. adolescentis strains isolated from the feces of elderly humans (B. adolescentis Z25, 17_3, and 2016_7_2) decreased the body weight or weight gain of mice, whilst the strain isolated from the newborn (B. adolescentis N4_N3) increased the body weight of mice. The B. adolescentis strains isolated from the elderly also increased serum leptin concentrations and induced the expression of thermogenesis- and lipid metabolism-related genes in brown adipose tissue. All the B. adolescentis strains alleviated inflammations in the spleen and brain and modified the cecal and colonic microbiota. Particularly, all strains reversed the HFD-induced depletion of Bifidobacterium and reduced the development of beta-lactam resistance. In addition, the B. adolescentis strains isolated from the elderly increased the relative abundances of potentially beneficial genera, such as Bacteroides, Parabacteroides, and Faecalibaculum. We speculate that such increased abundance of commensal bacteria may have mediated the alleviation of obesity, as B. adolescentis supplementation decreased the intestinal production of SCFAs, thereby reducing energy delivery to the host mice. Our results revealed that certain strains of B. adolescentis can alleviate obesity and modify the gut microbiota of mice. The tested strains of B. adolescentis showed different effects on lipid metabolism and immunity regulation, with these effects related to whether they had been isolated from the feces of newborn or elderly humans. This indicates that B. adolescentis from different sources may have disparate effects on host health possibly due to the transmission of origin-specific functions to the host.


Subject(s)
Bifidobacterium adolescentis/isolation & purification , Bifidobacterium adolescentis/metabolism , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Adipose Tissue, Brown/metabolism , Animals , Bifidobacterium adolescentis/genetics , Colon/microbiology , Cytokines/metabolism , Fatty Acids, Volatile/metabolism , Feces/microbiology , Gastrointestinal Microbiome/genetics , Immunity , Inflammation/metabolism , Intestines , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Probiotics , RNA, Ribosomal, 16S/metabolism , Weight Gain
8.
Sci Rep ; 10(1): 14112, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32839473

ABSTRACT

Gamma aminobutyric acid (GABA) is the principal inhibitory neurotransmitter playing a key role in anxiety and depression disorders in mammals. Recent studies revealed that members of the gut microbiota are able to produce GABA modulating the gut-brain axis response. Among members of the human gut microbiota, bifidobacteria are well known to establish many metabolic and physiologic interactions with the host. In this study, we performed genome analyses of more than 1,000 bifidobacterial strains publicly available revealing that Bifidobacterium adolescentis taxon might represent a model GABA producer in human gastrointestinal tract. Moreover, the in silico screening of human/animal metagenomic datasets showed an intriguing association/correlation between B. adolescentis load and mental disorders such as depression and anxiety. Interestingly, in vitro screening of 82 B. adolescentis strains allowed identifying two high GABA producers, i.e. B. adolescentis PRL2019 and B. adolescentis HD17T2H, which were employed in an in vivo trial in rats. Feeding Groningen rats with a supplementation of B. adolescentis strains, confirmed the ability of these microorganisms to stimulate the in vivo production of GABA highlighting their potential implication in gut-brain axis interactions.


Subject(s)
Bifidobacterium adolescentis/genetics , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , gamma-Aminobutyric Acid/genetics , Animals , Anxiety/physiopathology , Bacterial Load , Bifidobacterium adolescentis/classification , Bifidobacterium adolescentis/metabolism , Depression/physiopathology , Humans , Male , Models, Animal , Probiotics/administration & dosage , Rats , gamma-Aminobutyric Acid/biosynthesis , gamma-Aminobutyric Acid/metabolism
9.
Int J Biol Macromol ; 161: 389-397, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32479932

ABSTRACT

Resistant starch (RS) is a complex prebiotic carbohydrate beneficial to the human gut. In the present study, four genes encoding for putative amylolytic enzymes, likely to be responsible for RS-degradation, were identified in the genome of Bifidobacterium adolescentis P2P3 by comparative genomic analysis. Our results showed that only three enzymes (RSD1, RSD2, and RSD3) exhibited non-gelatinized high amylose corn starch (HACS)-degrading activity in addition to typical α-amylase activity. These three RS-degrading enzymes (RSD) were composed of multiple domains, including signal peptide, catalytic domain, carbohydrate binding domains, and putative cell wall-anchoring domains. Typical catalytic domains were conserved by exhibiting seven typical conserved regions (I-VII) found mostly in α-amylases. Analysis of enzymatic activity revealed that RSD2 displayed stronger activity toward HACS-granules than RSD1 and RSD3. Comparative genomics in combination with enzymatic experiments confirmed that RSDs might be the key enzymes used by RS-degrading bifidobacteria to degrade RS in a particular ecological niche, such as the human gut.


Subject(s)
Amylases/metabolism , Bifidobacterium adolescentis/enzymology , Gastrointestinal Microbiome , Resistant Starch/metabolism , Amino Acid Sequence , Amylases/chemistry , Bifidobacterium/classification , Bifidobacterium/enzymology , Bifidobacterium/genetics , Bifidobacterium adolescentis/classification , Bifidobacterium adolescentis/genetics , Computational Biology/methods , Genome, Bacterial , Humans , Hydrolysis , Phylogeny
10.
Enzyme Microb Technol ; 134: 109480, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32044027

ABSTRACT

Isofloridoside (D-isofloridoside and L-isofloridoside) is the main photosynthetic product in red algae. Here, given the importance of isofloridoside, a potentially effective method to produce isofloridoside from galactose and glycerol using whole-cell biocatalysts harboring α-galactosidase was developed. α-Galactosidase-encoding genes from Alicyclobacillus hesperidum, Lactobacillus plantarum, and Bifidobacterium adolescentis were cloned and the proteins were overproduced in Escherichia coli. The α-galactosidase from A. hesperidum (AHGLA) was chosen to synthesize isofloridoside. The effects of reaction pH, temperature, and substrate concentration were investigated. In the optimum biotransformation conditions, the final isofloridoside concentration reached 0.45 M (galactose conversion 23 %). The reaction mixtures were purified using activated charcoal and calcined Celite, and the purified product was identified as a mixture of D- and L-isofloridoside by liquid chromatography-mass spectrometry and nuclear magnetic resonance. This study provides a possible feasible method for the biosynthesis of isofloridoside from low-cost glycerol and galactose.


Subject(s)
Alicyclobacillus/enzymology , Galactose/metabolism , Galactosides/biosynthesis , Glycerol/metabolism , alpha-Galactosidase/metabolism , Alicyclobacillus/genetics , Bifidobacterium adolescentis/enzymology , Bifidobacterium adolescentis/genetics , Biocatalysis , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Hydrolysis , Lactobacillus plantarum/enzymology , Lactobacillus plantarum/genetics , Temperature , alpha-Galactosidase/genetics
11.
Med Sci Monit Basic Res ; 25: 238-244, 2019 Nov 26.
Article in English | MEDLINE | ID: mdl-31767826

ABSTRACT

BACKGROUND Intestinal bacterial communities are not homogenous throughout the gastrointestinal tract. Human research on the gut microbiome often neglects intra-intestinal variability by relying on a single measurement from stool samples. One source of complexity is the adherence to undigested, residual fiber. Currently, no procedure exists to extract RNA from distinct bacterial subpopulations in stool samples. MATERIAL AND METHODS A serial centrifugation procedure was developed in which bacterial RNA could be extracted from distinct stool-fractions - fiber-adherent and non-fiber-adherent bacteria. To test whether the separation procedure yielded distinct bacterial subpopulations, a set of RT-qPCR assays were developed for a fiber-adherent bacterial species, Bifidobacterium adolescentis, then a within-subject repeated-measures study was conducted with 3 human subjects undergoing 4 dietary regimens. At each timepoint, between-fraction differences in gene expression were evaluated. RESULTS The RNA isolation procedure was able to isolate intact RNA in 20 of 24 samples in the fiber-adherent fraction. PurB and sdh were identified as suitable reference genes for B. adolescentis RT-qPCR assays. When subjects were provided a high resistant starch diet, bacterial fractions exhibited different expression of the trp operon (p=0.031). CONCLUSIONS Our study provides human gut microbiome researchers a novel tool for evaluating functional characteristics of bacterial subpopulations in human stool. Moreover, these experiments provide modest support for the existence of a functionally unique fiber-adherent subpopulation of B. adolescentis. Until a more thorough evaluation of the adherent and non-adherent fraction can be performed, researchers should be cautious when generalizing functional data derived solely from unfractionated stool samples.


Subject(s)
Dietary Fiber/microbiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Bacteria/genetics , Bacteria/isolation & purification , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/isolation & purification , Diet , Humans , RNA/genetics , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Reproducibility of Results
12.
J Microbiol Biotechnol ; 29(12): 1904-1915, 2019 Dec 28.
Article in English | MEDLINE | ID: mdl-31635446

ABSTRACT

Resistant starch (RS) is metabolized by gut microbiota and involved in the production of short-chain fatty acids, which are related to a variety of physiological and health effects. Therefore, the availability of RS as a prebiotic is a topic of interest, and research on gut bacteria that can decompose RS is also important. The objectives in this study were 1) to isolate a human gut bacterium having strong degradation activity on non-gelatinized RS, 2) to characterize its RS-degrading characteristics, and 3) to investigate its probiotic effects, including a growth stimulation effect on other gut bacteria and an immunomodulatory effect. Bifidobacterium adolescentis P2P3 showing very strong RS granule utilization activity was isolated. It can attach to RS granules and form them into clusters. It also utilizes high-amylose corn starch granules up to 63.3%, and efficiently decomposes other various types of commercial RS without gelatinization. In a coculture experiment, Bacteroides thetaiotaomicron ATCC 29148, isolated from human feces, was able to grow using carbon sources generated from RS granules by B. adolescentis P2P3. In addition, B. adolescentis P2P3 demonstrated the ability to stimulate secretion of Th1 type cytokines from mouse macrophages in vitro that was not shown in other B. adolescentis. These results suggested that B. adolescentis P2P3 is a useful probiotic candidate, having immunomodulatory activity as well as the ability to feed other gut bacteria using RS as a prebiotic.


Subject(s)
Bifidobacterium adolescentis/metabolism , Gastrointestinal Microbiome/physiology , Starch/metabolism , Adult , Animals , Bifidobacterium adolescentis/classification , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/isolation & purification , Coculture Techniques , Cytokines , Feces/microbiology , Gelatin , Humans , Immunologic Factors , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Phylogeny , Prebiotics/microbiology , Probiotics/pharmacology
13.
Appl Microbiol Biotechnol ; 103(20): 8413-8425, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31399771

ABSTRACT

The introduction of the key non-oxidative glycolytic (NOG) pathway enzyme, phosphoketolases (PKTs), into heterologous hosts can improve the yield of a variety of acetyl CoA-derived products of interest. However, the low specific activity of existing PKTs compared with that of 6-phosphofructokinase (PFK), the key EMP pathway enzyme, largely limits their potential applications. To improve PKT activity, previous attempts have focused on increasing intracellular PKT concentration via the use of strong promoters. Herein, we report the establishment of a growth-coupled evolution strategy for the enrichment and selection of PKT mutants with improved specific activity in Corynebacterium glutamicum hosts with defective PFK. Five mutants from 9 Bifidobacterium adolescentis-source PKT (BA-PKT) mutant libraries were obtained. Site-directed mutagenesis analysis revealed 11 mutant sites which contributed to improved BA-PKT specific activity. Further structural analysis revealed that the mutant sites were located far away from the enzyme active site, which makes them almost unpredictable using a rational design approach. Mutant site recombination led to the construction of a novel mutant, PKTT2A/I6T/H260Y, with Vmax 29.77 ± 1.58 U/mg and Kcat/Km 0.32 ± 0.01 s-1/mM, which corresponds to 73.27 ± 3.25% and 80.16 ± 3.38% improvements, respectively, compared with the wildtype (Vmax; 17.17 ± 0.59 U/mg, Kcat/Km; 0.17 ± 0.01 s-1/mM). Expression of PKTT2A/I6T/H260 in C. glutamicum Z188 resulted in 16.67 ± 2.24% and 18.19 ± 0.53% improvement in L-glutamate titer and yield, respectively, compared with the wildtype BA-PKT. Our findings provide an efficient approach for improving the activity of PKTs. Furthermore, the novel mutants could serve as useful tools in improving the yield of L-glutamate and other acetyl CoA-associated products.


Subject(s)
Aldehyde-Lyases/metabolism , Corynebacterium glutamicum/enzymology , Corynebacterium glutamicum/growth & development , Glutamic Acid/metabolism , Mutation , Recombinant Proteins/metabolism , Aldehyde-Lyases/genetics , Bifidobacterium adolescentis/enzymology , Bifidobacterium adolescentis/genetics , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Directed Molecular Evolution , Kinetics , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Selection, Genetic
14.
Cell Mol Gastroenterol Hepatol ; 6(3): 301-319, 2018.
Article in English | MEDLINE | ID: mdl-30123820

ABSTRACT

Background & Aims: The human gut microbiota is becoming increasingly recognized as a key factor in homeostasis and disease. The lack of physiologically relevant in vitro models to investigate host-microbe interactions is considered a substantial bottleneck for microbiota research. Organoids represent an attractive model system because they are derived from primary tissues and embody key properties of the native gut lumen; however, access to the organoid lumen for experimental perturbation is challenging. Here, we report the development and validation of a high-throughput organoid microinjection system for cargo delivery to the organoid lumen and high-content sampling. Methods: A microinjection platform was engineered using off-the-shelf and 3-dimensional printed components. Microinjection needles were modified for vertical trajectories and reproducible injection volumes. Computer vision (CVis) and microfabricated CellRaft Arrays (Cell Microsystems, Research Triangle Park, NC) were used to increase throughput and enable high-content sampling of mock bacterial communities. Modeling preformed using the COMSOL Multiphysics platform predicted a hypoxic luminal environment that was functionally validated by transplantation of fecal-derived microbial communities and monocultures of a nonsporulating anaerobe. Results: CVis identified and logged locations of organoids suitable for injection. Reproducible loads of 0.2 nL could be microinjected into the organoid lumen at approximately 90 organoids/h. CVis analyzed and confirmed retention of injected cargos in approximately 500 organoids over 18 hours and showed the requirement to normalize for organoid growth for accurate assessment of barrier function. CVis analyzed growth dynamics of a mock community of green fluorescent protein- or Discosoma sp. red fluorescent protein-expressing bacteria, which grew within the organoid lumen even in the presence of antibiotics to control media contamination. Complex microbiota communities from fecal samples survived and grew in the colonoid lumen without appreciable changes in complexity. Conclusions: High-throughput microinjection into organoids represents a next-generation in vitro approach to investigate gastrointestinal luminal physiology and the gastrointestinal microbiota.


Subject(s)
Colon/cytology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/physiology , Microinjections/methods , Organoids/cytology , Animals , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/growth & development , Bifidobacterium adolescentis/isolation & purification , Colon/anatomy & histology , Escherichia coli/genetics , Escherichia coli/growth & development , Feces/microbiology , Humans , Male , Mice , Organoids/anatomy & histology , Single-Cell Analysis , Video Recording , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/growth & development
15.
Sci Rep ; 8(1): 11897, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30093677

ABSTRACT

Multiple mutations in the ß subunit of the RNA polymerase (rpoß) of Mycobacterium tuberculosis (Mtb) are the primary cause of resistance to rifamycin (RIF). In the present study, bifidobacterial rpoß sequences were analyzed to characterize the mutations that contribute to the development of intrinsic resistance to RIF, isoniazid, streptomycin and pyrazinamide. Sequence variations, which mapped to cassettes 1 and 2 of the rpoß pocket, are also found in multidrug-resistant Mtb (MDR Mtb). Growth curves in the presence of osmolytes and different concentrations of RIF showed that the bacteria adapted rapidly by shortening the growth curve lag time. Insight into the adapted rpoß DNA sequences revealed that B. adolescentis harbored mutations both in the RIF pocket and in regions outside the pocket. The minimum inhibitory concentrations (MICs) and mutant prevention concentrations (MPCs) indicated that B. longum, B. adolescentis and B. animalis are resistant to antitubercular drugs. 3D-homology modeling and binding interaction studies using computational docking suggested that mutants had reduced binding affinity towards RIF. RIF-exposed/resistant bacteria exhibited variant protein profiles along with morphological differences, such as elongated and branched cells, surface conversion from rough to smooth, and formation of a concentrating ring.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Bifidobacterium adolescentis/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , RNA Polymerase II/metabolism , Antitubercular Agents/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/growth & development , Binding Sites/genetics , Drug Resistance, Multiple, Bacterial/genetics , Isoniazid/metabolism , Isoniazid/pharmacology , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutation , Protein Binding , Protein Domains , Pyrazinamide/metabolism , Pyrazinamide/pharmacology , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , Rifamycins/metabolism , Rifamycins/pharmacology
16.
J Agric Food Chem ; 65(29): 6030-6041, 2017 Jul 26.
Article in English | MEDLINE | ID: mdl-28664731

ABSTRACT

Cost-efficient (bio)chemical production processes are essential to evaluate the commercial and industrial applications of promising carbohydrates and also are essential to ensure economically viable production processes. Here, the synthesis of the naturally occurring disaccharide kojibiose (2-O-α-d-glucopyranosyl-d-glucopyranoside) was evaluated using different Bifidobacterium adolescentis sucrose phosphorylase variants. Variant L341I_Q345S was found to efficiently synthesize kojibiose while remaining fully active after 1 week of incubation at 55 °C. Process optimization allowed kojibiose production at the kilogram scale, and simple but efficient downstream processing, using a yeast treatment and crystallization, resulted in more than 3 kg of highly pure crystalline kojibiose (99.8%). These amounts allowed a deeper characterization of its potential in food applications. It was found to have possible beneficial health effects, including delayed glucose release and potential to trigger SCFA production. Finally, we compared the bulk functionality of highly pure kojibiose to that of sucrose, hereby mapping its potential as a new sweetener in confectionery products.


Subject(s)
Bifidobacterium adolescentis/metabolism , Disaccharides/metabolism , Bifidobacterium adolescentis/genetics , Biocatalysis , Caco-2 Cells , Fatty Acids, Volatile/metabolism , Fermentation , Gastrointestinal Microbiome , Glucose/metabolism , Humans , Industrial Microbiology , Intestinal Mucosa/metabolism , Intestines/microbiology , Sucrose/metabolism
17.
J Biotechnol ; 258: 69-78, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28153765

ABSTRACT

Xylose-negative Corynebacterium glutamicum has been engineered to utilize xylose as the sole carbon source via either the xylose isomerase (XI) pathway or the Weimberg pathway. Heterologous expression of xylose isomerase and overexpression of a gene encoding for xylulose kinase enabled efficient xylose utilization. In this study, we show that two functionally-redundant transcriptional regulators (GntR1 and GntR2) present on xylose repress the pentose phosphate pathway genes. For efficient xylose utilization, pentose phosphate pathway genes and a phosphoketolase gene were overexpressed with the XI pathway in C. glutamicum. Overexpression of the genes encoding for transaldolase (Tal), 6-phosphogluconate dehydrogenase (Gnd), or phosphoketolase (XpkA) enhanced the growth and xylose consumption rates compared to the wild-type with the XI pathway alone. However, co-expression of these genes did not have a synergetic effect on xylose utilization. For the succinate production from xylose, overexpression of the tal gene with the XI pathway in a succinate-producing strain improved xylose utilization and increased the specific succinate production rate by 2.5-fold compared to wild-type with the XI pathway alone. Thus, overexpression of the tal, gnd, or xpkA gene could be helpful for engineering C. glutamicum toward production of value-added chemicals with efficient xylose utilization.


Subject(s)
Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Metabolic Engineering/methods , Succinic Acid/metabolism , Xylose/metabolism , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Aspergillus nidulans/enzymology , Aspergillus nidulans/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bifidobacterium adolescentis/enzymology , Bifidobacterium adolescentis/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Pentose Phosphate Pathway/genetics , Phosphogluconate Dehydrogenase/genetics , Phosphogluconate Dehydrogenase/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Succinic Acid/analysis , Transaldolase/genetics , Transaldolase/metabolism
18.
Sci Rep ; 6: 23971, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27035119

ABSTRACT

Bifidobacteria are members of the human gut microbiota, being numerically dominant in the colon of infants, while also being prevalent in the large intestine of adults. In this study, we determined and analyzed the pan-genome of Bifidobacterium adolescentis, which is one of many bacteria found in the human adult gut microbiota. In silico analysis of the genome sequences of eighteen B. adolescentis strains isolated from various environments, such as human milk, human feces and bovine rumen, revealed a high level of genetic variability, resulting in an open pan-genome. Compared to other bifidobacterial taxa such as Bifidobacterium bifidum and Bifidobacterium breve, the more extensive B. adolescentis pan-genome supports the hypothesis that the genetic arsenal of this taxon expanded so as to become more adaptable to the variable and changing ecological niche of the gut. These increased genetic capabilities are particularly evident for genes required for dietary glycan-breakdown.


Subject(s)
Bifidobacterium adolescentis/genetics , Gastrointestinal Microbiome , Genetic Variation , Intestines/microbiology , Animals , Bifidobacterium adolescentis/isolation & purification , Carbohydrates/chemistry , Cattle , DNA, Bacterial/genetics , Diet , Feces/microbiology , Genome, Bacterial , Genomics , Humans , Milk/microbiology , Open Reading Frames , Polysaccharides/chemistry , Rumen/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...