Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.417
Filter
1.
World J Microbiol Biotechnol ; 40(9): 280, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39060821

ABSTRACT

Synbiotics are complex preparations of prebiotics that can be selectively utilized by live microorganisms to improve host health. Synbiotics are divided into complementary synbiotics, which consist of probiotics and prebiotics with independent functions, and synergistic synbiotics, which consist of prebiotics that are selectively used by gut microorganisms. Complementary synbiotics used in human clinical trials include Lactobacillus spp. and Bifidobacterium spp. as probiotics, and fructooligosaccharides, galactooligosaccharides, and inulin as prebiotics. Over the past five years, synbiotics have been most commonly used in patients with metabolic disorders, including obesity, and immune and gastrointestinal disorders. Several studies have observed alterations in the microbial community; however, these changes did not lead to significant improvements in disease outcomes or biochemical and hematological markers. The same synbiotics have been applied to individuals with different gut environments. As a result, even with the same synbiotics, there are non-responders who do not respond to the applied synbiotics due to the different intestinal environment for each individual. Therefore, to obtain meaningful results, applying different synbiotics depending on the individual is necessary. Synergistic synbiotics are one solution to circumvent this problem, as they combine elements that can effectively improve health, even in non-responders. This review aims to explain the concept of synbiotics, highlight recent human clinical trials, and explore the current state of research on synergistic synbiotics.


Subject(s)
Gastrointestinal Microbiome , Health Promotion , Prebiotics , Probiotics , Synbiotics , Humans , Health Promotion/methods , Disease Management , Oligosaccharides/metabolism , Bifidobacterium/metabolism , Lactobacillus/metabolism , Obesity/therapy , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/therapy
2.
Food Res Int ; 191: 114720, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39059916

ABSTRACT

Mangosteen (Garcinia mangostana L.) is a tasty, polyphenol-rich tropical fruit. The edible part is highly appreciated by its aroma, taste and texture. The non-edible part, rich in polyphenols, has been traditionally used in Thai medicine. In this work, flavonoids and phenolic acid/derivatives were identified in mangosteen extracts (ME) from edible and non-edible portions. We first studied the effects of MEs on the growth, metabolism, antioxidant capacity, biofilm formation and antimicrobial capacity of eight bifidobacteria and lactobacilli strains from intestinal origin and two commercial probiotic strains (BB536 and GG). ME concentrations higher than 10-20 % were inhibitory for all strains. However, ME concentrations of 5 % significantly (P < 0.01) increased all strains antioxidant capacity, reduced biofilm-formation, and enhanced inhibition against Gram-positive pathogens. To apply these knowledge, bifunctional fermented milk products were elaborated with 5 % ME and individual strains, which were selected taking into account their growth with ME, and the widest range of values on antioxidant capacity, biofilm formation and antimicrobial activity (bifidobacteria INIA P2 and INIA P467, lactobacilli INIA P459 and INIA P708, and reference strain GG). Most strains survived well manufacture, refrigerated storage and an in vitro simulation of major conditions encountered in the gastrointestinal tract. As expected, products supplemented with ME showed higher polyphenol content and antioxidant capacity levels than control. After sensory evaluation, products containing strains INIA P2, INIA P708 and GG outstood as best.


Subject(s)
Antioxidants , Biofilms , Cultured Milk Products , Garcinia mangostana , Lactobacillus , Plant Extracts , Plant Extracts/pharmacology , Garcinia mangostana/chemistry , Biofilms/drug effects , Biofilms/growth & development , Antioxidants/pharmacology , Lactobacillus/drug effects , Lactobacillus/metabolism , Cultured Milk Products/microbiology , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Probiotics , Flavonoids/pharmacology , Flavonoids/analysis , Humans , Fruit/chemistry , Fruit/microbiology , Fermentation , Hydroxybenzoates/pharmacology , Gastrointestinal Microbiome/drug effects , Polyphenols/pharmacology
3.
Nutrients ; 16(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38999750

ABSTRACT

(1) Background: Recently, academic studies are demonstrating that the cholesterol-lowering effects of pectin oligosaccharides (POSs) are correlated to intestinal flora. However, the mechanisms of POS on cholesterol metabolisms are limited, and the observations of intestinal flora are lacking integrative analyses. (2) Aim and methods: To reveal the regulatory mechanisms of POS on cholesterol metabolism via an integrative analysis of the gut microbiota, the changes in gut microbiota structure and metabolite composition after POS addition were investigated using Illumina MiSeq sequencing and non-targeted metabolomics through in vitro gut microbiota fermentation. (3) Results: The composition of fecal gut flora was adjusted positively by POS. POS increased the abundances of the cholesterol-related bacterial groups Bacteroidetes, Bifidobacterium and Lactobacillus, while it decreased conditional pathogenic Escherichia coli and Enterococcus, showing good prebiotic activities. POS changed the composition of gut microbiota fermentation metabolites (P24), causing significant changes in 221 species of fermentation metabolites in a non-targeted metabolomics analysis and promoting the production of short-chain fatty acids. The abundances of four types of cholesterol metabolism-related metabolites (adenosine monophosphate, cyclic adenosine monophosphate, guanosine and butyrate) were significantly higher in the P24 group than those in the control group without POS addition. (4) Conclusion: The abovementioned results may explain the hypocholesterolemic effects of POS and promotion effects on cholesterol efflux of P24. These findings indicated that the potential regulatory mechanisms of citrus POS on cholesterol metabolism are modulated by cholesterol-related gut microbiota and specific metabolites.


Subject(s)
Cholesterol , Feces , Fermentation , Gastrointestinal Microbiome , Oligosaccharides , Pectins , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Pectins/pharmacology , Pectins/metabolism , Cholesterol/metabolism , Oligosaccharides/pharmacology , Feces/microbiology , Humans , Prebiotics , Male , Metabolomics , Fatty Acids, Volatile/metabolism , Bifidobacterium/metabolism , Bifidobacterium/drug effects , Female , Bacteria/metabolism , Bacteria/drug effects , Bacteria/classification , Citrus
4.
Nutrients ; 16(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892631

ABSTRACT

This study investigated the effect of astragalus polysaccharide (APS, an ingredient with hypoglycemic function in a traditional Chinese herbal medicine) on gut microbiota and metabolites of type 2 diabetes mellitus (T2DM) patients using a simulated fermentation model in vitro. The main components of APS were isolated, purified, and structure characterized. APS fermentation was found to increase the abundance of Lactobacillus and Bifidobacterium and decrease the Escherichia-Shigella level in the fecal microbiota of T2DM patients. Apart from increasing propionic acid, APS also caused an increase in all-trans-retinoic acid and thiamine (both have antioxidant properties), with their enrichment in the KEGG pathway associated with thiamine metabolism, etc. Notably, APS could also enhance fecal antioxidant properties. Correlation analysis confirmed a significant positive correlation of Lactobacillus with thiamine and DPPH-clearance rate, suggesting the antioxidant activity of APS was related to its ability to enrich some specific bacteria and upregulate their metabolites.


Subject(s)
Antioxidants , Astragalus Plant , Diabetes Mellitus, Type 2 , Feces , Fermentation , Gastrointestinal Microbiome , Polysaccharides , Gastrointestinal Microbiome/drug effects , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Polysaccharides/pharmacology , Astragalus Plant/chemistry , Feces/microbiology , Antioxidants/pharmacology , Male , Female , Middle Aged , Thiamine/pharmacology , Thiamine/metabolism , Bifidobacterium/metabolism , Bifidobacterium/drug effects , Lactobacillus/metabolism , Lactobacillus/drug effects , Hypoglycemic Agents/pharmacology
5.
Molecules ; 29(12)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38930817

ABSTRACT

With the aim to obtain controlled-release systems and to preserve the antioxidant, immunomodulatory, and prebiotic activity of the bioactive compounds, microencapsulation of both honeydew honey and royal jelly into biopolymeric microparticles based on rye bran heteropolysaccharides (HPS) was successfully performed. Honeydew honey and royal jelly microcapsules were prepared by spray-drying method and were characterized in terms of morphology and biological properties. Due to the resistance of the obtained encapsulates to the acidic pH in the stomach and digestive enzymes, the microcapsules showed prebiotic properties positively influencing both the growth, retardation of the dying phase, and the pro-adhesive properties of probiotic bacteria, i.e., Bifidobacterium spp. and lactic acid bacteria. Moreover, as a result of fermentation of the microcapsules of bee products in the lumen of the large intestine, an increased synthesis of short-chain fatty acids, i.e., butyric acid, was found on average by 39.2% in relation to the SCFA concentrations obtained as a result of fermentation of native bee products, thus opening new perspectives for the exploitation of honeydew honey and royal jelly loaded microcapsules for nutraceutical applications.


Subject(s)
Capsules , Gastrointestinal Microbiome , Honey , Prebiotics , Animals , Gastrointestinal Microbiome/drug effects , Bees , Fatty Acids, Volatile/metabolism , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Fermentation , Probiotics , Fatty Acids
6.
Nutrients ; 16(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38931211

ABSTRACT

Iron deficiency remains a public health challenge globally. Prebiotics have the potential to improve iron bioavailability by modulating intestinal bacterial population, increasing SCFA production, and stimulating expression of brush border membrane (BBM) iron transport proteins among iron-deficient populations. This study intended to investigate the potential effects of soluble extracts from the cotyledon and seed coat of three pea (Pisum sativum) varieties (CDC Striker, CDC Dakota, and CDC Meadow) on the expression of BBM iron-related proteins (DCYTB and DMT1) and populations of beneficial intestinal bacteria in vivo using the Gallus gallus model by oral gavage (one day old chicks) with 1 mL of 50 mg/mL pea soluble extract solutions. The seed coat treatment groups increased the relative abundance of Bifidobacterium compared to the cotyledon treatment groups, with CDC Dakota seed coat (dark brown pigmented) recording the highest relative abundance of Bifidobacterium. In contrast, CDC Striker Cotyledon (dark-green-pigmented) significantly increased the relative abundance of Lactobacillus (p < 0.05). Subsequently, the two dark-pigmented treatment groups (CDC Striker Cotyledon and CDC Dakota seed coats) recorded the highest expression of DCYTB. Our study suggests that soluble extracts from the pea seed coat and dark-pigmented pea cotyledon may improve iron bioavailability by affecting intestinal bacterial populations.


Subject(s)
Chickens , Gastrointestinal Microbiome , Iron , Pisum sativum , Prebiotics , Animals , Gastrointestinal Microbiome/drug effects , Iron/metabolism , Plant Extracts/pharmacology , Intestines/microbiology , Seeds , Bifidobacterium/metabolism , Cotyledon , Lactobacillus/metabolism , Cation Transport Proteins
7.
Nutrients ; 16(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38931248

ABSTRACT

Human milk provides essential nutrients for infants but also consists of human milk oligosaccharides (HMOs), which are resistant to digestion by the infant. Bifidobacteria are among the first colonizers, providing various health benefits for the host. This is largely facilitated by their ability to efficiently metabolize HMOs in a species-specific way. Nevertheless, these abilities can vary significantly by strain, and our understanding of the mechanisms applied by different strains from the same species remains incomplete. Therefore, we assessed the effects of strain-level genomic variation in HMO utilization genes on growth on HMOs in 130 strains from 10 species of human associated bifidobacteria. Our findings highlight the extent of genetic diversity between strains of the same species and demonstrate the effects on species-specific HMO utilization, which in most species is largely retained through the conservation of a core set of genes or the presence of redundant pathways. These data will help to refine our understanding of the genetic factors that contribute to the persistence of individual strains and will provide a better mechanistic rationale for the development and optimization of new early-life microbiota-modulating products to improve infant health.


Subject(s)
Bifidobacterium , Milk, Human , Oligosaccharides , Species Specificity , Bifidobacterium/genetics , Bifidobacterium/metabolism , Humans , Oligosaccharides/metabolism , Genetic Variation , Infant , Genes, Bacterial
8.
Int J Biol Macromol ; 272(Pt 1): 132906, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38851991

ABSTRACT

Sourdough bread enriched with soluble fiber (by in-situ exopolysaccharides production) and insoluble fiber (by gazpacho by-products addition) showed prebiotic effects an in vitro dynamic colonic fermentation performance with obese volunteer's microbiota. Bifidobacterium population was maintained whereas Lactobacillus increased throughout the colonic sections. Conversely, Enterobacteriaceae and Clostridium groups clearly decreased. Specific bacteria associated with beneficial effects increased in the ascending colon (Lactobacillus fermentum, Lactobacillus paracasei, Bifidobacterium longum and Bifidobacterium adolescentis) whereas Eubacterium eligens, Alistipes senegalensis, Prevotella copri and Eubacterium desmolans increased in the transversal and descending colon. Additionally, Blautia faecis and Ruminococcus albus increased in the transversal colon, and Bifidobacterium longum, Roseburia faecis and Victivallis vadensis in the descending colon. Bifidobacterium and Lactobacillus fermented the in-situ exopolysaccharides and released pectins from gazpacho by-products, as well as cellulosic degraded bacteria. This increased the short and medium chain fatty acids. Acetic acid, as well as butyric acid, increased throughout the colonic tract, which showed greater increases only in the transversal and descending colonic segments. Conversely, propionic acid was slightly affected by the colonic fermentation. These results show that sourdough bread is a useful food matrix for the enrichment of vegetable by-products (or other fibers) in order to formulate products with microbiota modulatory capacities.


Subject(s)
Bread , Dysbiosis , Fermentation , Bread/microbiology , Humans , Dysbiosis/microbiology , Gastrointestinal Microbiome/drug effects , Dietary Fiber/metabolism , Polysaccharides, Bacterial/pharmacology , Colon/microbiology , Colon/metabolism , Bifidobacterium/metabolism , Male , Lactobacillus/metabolism
9.
NPJ Biofilms Microbiomes ; 10(1): 47, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898089

ABSTRACT

Throughout the life span of a host, bifidobacteria have shown superior colonization and glycan abilities. Complex glycans, such as human milk oligosaccharides and plant glycans, that reach the colon are directly internalized by the transport system of bifidobacteria, cleaved into simple structures by extracellular glycosyl hydrolase, and transported to cells for fermentation. The glycan utilization of bifidobacteria introduces cross-feeding activities between bifidobacterial strains and other microbiota, which are influenced by host nutrition and regulate gut homeostasis. This review discusses bifidobacterial glycan utilization strategies, focusing on the cross-feeding involved in bifidobacteria and its potential health benefits. Furthermore, the impact of cross-feeding on the gut trophic niche of bifidobacteria and host health is also highlighted. This review provides novel insights into the interactions between microbe-microbe and host-microbe.


Subject(s)
Bifidobacterium , Gastrointestinal Microbiome , Homeostasis , Polysaccharides , Humans , Bifidobacterium/metabolism , Bifidobacterium/physiology , Polysaccharides/metabolism , Host Microbial Interactions , Animals , Fermentation
10.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38896583

ABSTRACT

Probiotics have gained significant attention as a potential strategy to improve health by modulating host-microbe interactions, particularly in situations where the normal microbiota has been disrupted. However, evidence regarding their efficacy has been inconsistent, with considerable interindividual variability in response. We aimed to explore whether a common genetic variant that affects the production of mucosal α(1,2)-fucosylated glycans, present in around 20% of the population, could explain the observed interpersonal differences in the persistence of commonly used probiotics. Using a mouse model with varying α(1,2)-fucosylated glycans secretion (Fut2WT or Fut2KO), we examined the abundance and persistence of Bifidobacterium strains (infantis, breve, and bifidum). We observed significant differences in baseline gut microbiota characteristics between Fut2WT and Fut2KO littermates, with Fut2WT mice exhibiting enrichment of species able to utilize α(1,2)-fucosylated glycans. Following antibiotic exposure, only Fut2WT animals showed persistent engraftment of Bifidobacterium infantis, a strain able to internalize α(1,2)-fucosylated glycans, whereas B. breve and B. bifidum, which cannot internalize α(1,2)-fucosylated glycans, did not exhibit this difference. In mice with an intact commensal microbiota, the relationship between secretor status and B. infantis persistence was reversed, with Fut2KO animals showing greater persistence compared to Fut2WT. Our findings suggest that the interplay between a common genetic variation and antibiotic exposure plays a crucial role in determining the dynamics of B. infantis in the recipient gut, which could potentially contribute to the observed variation in response to this commonly used probiotic species.


Subject(s)
Anti-Bacterial Agents , Fucosyltransferases , Galactoside 2-alpha-L-fucosyltransferase , Gastrointestinal Microbiome , Probiotics , Animals , Mice , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Probiotics/administration & dosage , Anti-Bacterial Agents/pharmacology , Bifidobacterium longum subspecies infantis/genetics , Bifidobacterium longum subspecies infantis/metabolism , Polysaccharides/metabolism , Host Microbial Interactions , Mice, Inbred C57BL , Mice, Knockout , Bifidobacterium/genetics , Bifidobacterium/metabolism
11.
Int J Biol Macromol ; 273(Pt 1): 133053, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38857723

ABSTRACT

Trehalose (α-d-glucopyranosyl-(1-1)-α-D-glucopyranoside) has found applications in diverse food products as a sweetener, stabilizer, and humectant. Recent attention has focused on trehalose due to its contradictory effects on the virulence of Clostridium difficile. In this study, we investigate the impact of novel trehalose-derived galactooligosaccharides (Treh-GOS) on the human gut microbiota using in vitro fecal fermentation models. Distinct Treh-GOS structures elicit varying taxonomic responses. For instance, ß-Gal-(1-4)-trehalose [DP3(1-4)] leads to an increase of Bifidobacterium, comparable to results observed with commercial GOS. Conversely, ß-Gal-(1-6)-trehalose [DP3(1-6)] prompts an increase in Lactobacillus. Notably, both of these trisaccharides yield the highest concentrations of butyric acid across all samples. On the other hand, Treh-GOS tetrasaccharide mixture (DP4), featuring a novel trehalose galactosylation in both glucose units, fosters the growth of Parabacteroides. Our findings underscore the capacity of novel Treh-GOS to modulate the human gut microbiota. Consequently, these innovative galactooligosaccharides emerge as promising candidates for novel prebiotic applications.


Subject(s)
Fermentation , Gastrointestinal Microbiome , Oligosaccharides , Trehalose , Trehalose/pharmacology , Trehalose/chemistry , Gastrointestinal Microbiome/drug effects , Humans , Oligosaccharides/pharmacology , Oligosaccharides/chemistry , Fermentation/drug effects , Feces/microbiology , Prebiotics , Bifidobacterium/drug effects , Bifidobacterium/metabolism
12.
Appl Environ Microbiol ; 90(7): e0024724, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38888338

ABSTRACT

The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE: By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.


Subject(s)
Bifidobacterium , Extracellular Vesicles , Limosilactobacillus reuteri , Probiotics , Limosilactobacillus reuteri/metabolism , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/growth & development , Extracellular Vesicles/metabolism , Humans , Bifidobacterium/metabolism , Bifidobacterium/genetics , Bifidobacterium/growth & development
13.
Carbohydr Polym ; 339: 122292, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823937

ABSTRACT

Through adaptive laboratory evolution (ALE) of Sphingomonas sp. ATCC 31555, fermentation for production of low-molecular-weight welan gum (LMW-WG) was performed using glycerol as sole carbon source. During ALE, GPC-MALS analysis revealed a gradual decrease in WG molecular weight with the increase of adaptation cycles, accompanied by changes in solution conformation. LMW-WG was purified and structurally analyzed using GPC-MALS, monosaccharide composition analysis, infrared spectroscopy, NMR analysis, atomic force microscopy, and scanning electron microscopy. Subsequently, LMW-WG obtains hydration, transparency, antioxidant activity, and rheological properties. Finally, an in vitro simulation colon reactor was used to evaluate potential prebiotic properties of LMW-WG as dietary fiber. Compared with WG produced using sucrose as substrate, LMW-WG exhibited a fourfold reduction in molecular weight while maintaining moderate viscosity. Structurally, L-Rha nearly completely replaced L-Man. Furthermore, LMW-WG demonstrated excellent hydration, antioxidant activity, and high transparency. It also exhibited resistance to saliva and gastrointestinal digestion, showcasing a favorable colonization effect on Bifidobacterium, making it a promising symbiotic agent.


Subject(s)
Antioxidants , Fermentation , Glycerol , Molecular Weight , Sphingomonas , Glycerol/chemistry , Glycerol/metabolism , Antioxidants/chemistry , Antioxidants/pharmacology , Sphingomonas/metabolism , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/pharmacology , Viscosity , Prebiotics , Bifidobacterium/metabolism
14.
Int J Biol Macromol ; 268(Pt 2): 131836, 2024 May.
Article in English | MEDLINE | ID: mdl-38692553

ABSTRACT

Multiple species of Bifidobacterium exhibit the ability to bioconvert conjugated fatty acids (CFAs), which is considered an important pathway for these strains to promote host health. However, there has been limited progress in understanding the enzymatic mechanism of CFA bioconversion by bifidobacteria, despite the increasing number of studies identifying CFA-producing strains. The protein responsible for polyunsaturated fatty acid (PUFA) isomerization in B. breve CCFM683 has recently been discovered and named BBI, providing a starting point for exploring Bifidobacterium isomerases (BIs). This study presents the sequence classification of membrane-bound isomerases from four common Bifidobacterium species that produce CFA. Heterologous expression, purification, and enzymatic studies of the typical sequences revealed that all possess a single c9, t11 isomer as the product and share common features in terms of enzymatic properties and catalytic kinetics. Using molecular docking and alanine scanning, Lys84, Tyr198, Asn202, and Leu245 located in the binding pocket were identified as critical to the catalytic activity, a finding further confirmed by site-directed mutagenesis-based screening assays. Overall, these findings provide insightful knowledge concerning the molecular mechanisms of BIs. This will open up additional opportunities for the use of bifidobacteria and CFAs in probiotic foods and precision nutrition.


Subject(s)
Bifidobacterium , Fatty Acids, Unsaturated , Bifidobacterium/enzymology , Bifidobacterium/genetics , Bifidobacterium/metabolism , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Molecular Docking Simulation , Isomerism , Kinetics , Amino Acid Sequence , Mutagenesis, Site-Directed , Probiotics/metabolism
15.
Gut Microbes ; 16(1): 2347728, 2024.
Article in English | MEDLINE | ID: mdl-38706226

ABSTRACT

Indole in the gut is formed from dietary tryptophan by a bacterial tryptophan-indole lyase. Indole not only triggers biofilm formation and antibiotic resistance in gut microbes but also contributes to the progression of kidney dysfunction after absorption by the intestine and sulfation in the liver. As tryptophan is an essential amino acid for humans, these events seem inevitable. Despite this, we show in a proof-of-concept study that exogenous indole can be converted to an immunomodulatory tryptophan metabolite, indole-3-lactic acid (ILA), by a previously unknown microbial metabolic pathway that involves tryptophan synthase ß subunit and aromatic lactate dehydrogenase. Selected bifidobacterial strains converted exogenous indole to ILA via tryptophan (Trp), which was demonstrated by incubating the bacterial cells in the presence of (2-13C)-labeled indole and l-serine. Disruption of the responsible genes variedly affected the efficiency of indole bioconversion to Trp and ILA, depending on the strains. Database searches against 11,943 bacterial genomes representing 960 human-associated species revealed that the co-occurrence of tryptophan synthase ß subunit and aromatic lactate dehydrogenase is a specific feature of human gut-associated Bifidobacterium species, thus unveiling a new facet of bifidobacteria as probiotics. Indole, which has been assumed to be an end-product of tryptophan metabolism, may thus act as a precursor for the synthesis of a host-interacting metabolite with possible beneficial activities in the complex gut microbial ecosystem.


Subject(s)
Bifidobacterium , Gastrointestinal Microbiome , Indoles , Tryptophan , Tryptophan/metabolism , Humans , Indoles/metabolism , Bifidobacterium/metabolism , Bifidobacterium/genetics , Tryptophan Synthase/metabolism , Tryptophan Synthase/genetics , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism
16.
Microb Biotechnol ; 17(5): e14443, 2024 May.
Article in English | MEDLINE | ID: mdl-38722820

ABSTRACT

Pectin structures have received increasing attention as emergent prebiotics due to their capacity to promote beneficial intestinal bacteria. Yet the collective activity of gut bacterial communities to cooperatively metabolize structural variants of this substrate remains largely unknown. Herein, the characterization of a pectin methylesterase, BpeM, from Bifidobacterium longum subsp. longum, is reported. The purified enzyme was able to remove methyl groups from highly methoxylated apple pectin, and the mathematical modelling of its activity enabled to tightly control the reaction conditions to achieve predefined final degrees of methyl-esterification in the resultant pectin. Demethylated pectin, generated by BpeM, exhibited differential fermentation patterns by gut microbial communities in in vitro mixed faecal cultures, promoting a stronger increase of bacterial genera associated with beneficial effects including Lactobacillus, Bifidobacterium and Collinsella. Our findings demonstrate that controlled pectin demethylation by the action of a B. longum esterase selectively modifies its prebiotic fermentation pattern, producing substrates that promote targeted bacterial groups more efficiently. This opens new possibilities to exploit biotechnological applications of enzymes from gut commensals to programme prebiotic properties.


Subject(s)
Carboxylic Ester Hydrolases , Feces , Malus , Pectins , Prebiotics , Malus/microbiology , Pectins/metabolism , Feces/microbiology , Carboxylic Ester Hydrolases/metabolism , Carboxylic Ester Hydrolases/genetics , Fermentation , Humans , Bifidobacterium longum/metabolism , Bifidobacterium longum/enzymology , Gastrointestinal Microbiome , Bifidobacterium/enzymology , Bifidobacterium/metabolism
17.
Food Chem ; 453: 139644, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-38761735

ABSTRACT

This work developed and characterized the physicochemical properties of a type A gelatin and amidated low-methoxyl pectin complex coacervate (GA-LMAP-CC) hydrogel and evaluated its suitability for preserving the viability of probiotics under in vitro gastrointestinal conditions. The formation of GA-LMAP-CC was achieved via height electrostatic attraction at pH 3 and a mixing ratio of 1, exhibiting thermoreversible gel behavior. The hydrogel had a porosity of 44% and a water absorption capacity of up to 12 times. Water absorption profiles were obtained at different pH values (2, 5, and 7). The influence of GA-LMAP-CC depended on the medium, which controlled the hydration and water absorption rate. GA-LMAP-CC promoted the viability of B. longum BB536 and L. acidophilus strains under simulated gastrointestinal conditions, thereby enhancing their potential for intestinal colonization. The hydrogel has suitable properties for potential application in food and pharmaceutical areas to encapsulate and preserve probiotics.


Subject(s)
Gelatin , Hydrogels , Pectins , Probiotics , Pectins/chemistry , Gelatin/chemistry , Probiotics/chemistry , Hydrogels/chemistry , Microbial Viability/drug effects , Lactobacillus acidophilus/chemistry , Lactobacillus acidophilus/growth & development , Lactobacillus acidophilus/metabolism , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Hydrogen-Ion Concentration , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology
18.
Microbiol Res ; 285: 127741, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38761487

ABSTRACT

Fructooligosaccharides (FOS) are a common prebiotic widely used in functional foods. Meanwhile, Saccharomyces boulardii is a fungal probiotic frequenly used in the clinical treatment of diarrhea. Compared with single use, the combination of prebiotics and probiotics as symbiotics may be more effective in regulating gut microbiota as recently reported in the literature. The present study aimed to investigate the effects of FOS, S. boulardii and their combination on the structure and metabolism of the gut microbiota in healthy primary and secondary school students using an in vitro fermentation model. The results indicated that S. boulardii alone could not effectively regulate the community structure and metabolism of the microbiota. However, both FOS and the combination of FOS and S. boulardii could effectively regulate the microbiota, significantly inhibiting the growth of Escherichia-Shigella and Bacteroides, and controlling the production of the gases including H2S and NH3. In addition, both FOS and the combination could significantly promote the growth of Bifidobacteria and Lactobacillus, lower environmental pH, and enhance several physiological functions related to synthesis and metabolism. Nevertheless, the combination had more unique benefits as it promoted the growth of Lactobacillus, significantly increased CO2 production and enhanced the functional pathways of carbon metabolism and pyruvic acid metabolism. These findings provide guidance for clinical application and a theoretical basis for the development of synbiotic preparations.


Subject(s)
Fermentation , Gastrointestinal Microbiome , Oligosaccharides , Prebiotics , Probiotics , Saccharomyces boulardii , Students , Oligosaccharides/metabolism , Oligosaccharides/pharmacology , Gastrointestinal Microbiome/drug effects , Saccharomyces boulardii/metabolism , Humans , Probiotics/metabolism , Child , Male , Adolescent , Female , Lactobacillus/metabolism , Lactobacillus/growth & development , Bacteria/metabolism , Bacteria/classification , Feces/microbiology , Bifidobacterium/metabolism , Bifidobacterium/growth & development
19.
Microbiol Res ; 283: 127709, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593579

ABSTRACT

Bifidobacterium longum subsp. infantis commonly colonizes the human gut and is capable of metabolizing L-fucose, which is abundant in the gut. Multiple studies have focused on the mechanisms of L-fucose utilization by B. longum subsp. infantis, but the regulatory pathways governing the expression of these catabolic processes are still unclear. In this study, we have conducted a structural and functional analysis of L-fucose metabolism transcription factor FucR derived from B. longum subsp. infantis Bi-26. Our results indicated that FucR is a L-fucose-sensitive repressor with more α-helices, fewer ß-sheets, and ß-turns. Transcriptional analysis revealed that FucR displays weak negative self-regulation, which is counteracted in the presence of L-fucose. Isothermal titration calorimetry indicated that FucR has a 2:1 stoichiometry with L-fucose. The key amino acid residues for FucR binding L-fucose are Asp280 and Arg331, with mutation of Asp280 to Ala resulting in a decrease in the affinity between FucR and L-fucose with the Kd value from 2.58 to 11.68 µM, and mutation of Arg331 to Ala abolishes the binding ability of FucR towards L-fucose. FucR specifically recognized and bound to a 20-bp incomplete palindrome sequence (5'-ACCCCAATTACGAAAATTTTT-3'), and the affinity of the L-fucose-loaded FucR for the DNA fragment was lower than apo-FucR. The results provided new insights into the regulating L-fucose metabolism by B. longum subsp. infantis.


Subject(s)
Bifidobacterium longum , Bifidobacterium , Humans , Bifidobacterium/genetics , Bifidobacterium/metabolism , Fucose/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Carbohydrate Metabolism , Bifidobacterium longum/genetics , Bifidobacterium longum/metabolism
20.
Nutrients ; 16(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38674850

ABSTRACT

Polyphenols and fermentable fibers have shown favorable effects on gut microbiota composition and metabolic function. However, few studies have investigated whether combining multiple fermentable fibers or polyphenols may have additive beneficial effects on gut microbial states. Here, an in vitro fermentation model, seeded with human stool combined from 30 healthy volunteers, was supplemented with blends of polyphenols (PP), dietary fibers (FB), or their combination (PPFB) to determine influence on gut bacteria growth dynamics and select metabolite changes. PP and FB blends independently led to significant increases in the absolute abundance of select beneficial taxa, namely Ruminococcus bromii, Bifidobacterium spp., Lactobacillus spp., and Dorea spp. Total short-chain fatty acid concentrations, relative to non-supplemented control (F), increased significantly with PPFB and FB supplementation but not PP. Indole and ammonia concentrations decreased with FB and PPFB supplementation but not PP alone while increased antioxidant capacity was only evident with both PP and PPFB supplementation. These findings demonstrated that, while the independent blends displayed selective positive impacts on gut states, the combination of both blends provided an additive effect. The work outlines the potential of mixed substrate blends to elicit a broader positive influence on gut microbial composition and function to build resiliency toward dysbiosis.


Subject(s)
Dietary Fiber , Fatty Acids, Volatile , Feces , Fermentation , Gastrointestinal Microbiome , Indoles , Polyphenols , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Polyphenols/pharmacology , Humans , Dietary Fiber/pharmacology , Dietary Fiber/administration & dosage , Feces/microbiology , Fatty Acids, Volatile/metabolism , Adult , Male , Ammonia/metabolism , Female , Bacteria/metabolism , Bacteria/growth & development , Bacteria/drug effects , Antioxidants/pharmacology , Bifidobacterium/metabolism , Lactobacillus/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL