Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.512
Filter
1.
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
2.
Curr Microbiol ; 81(7): 168, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38733376

ABSTRACT

In 2018, Nouioui et al. proposed that Bifidobacterium coryneforme was a later synonym of Bifidobacterium indicum on the basis of the digital DNA-DNA hybridization (dDDH) value (85.0%) between B. coryneforme LMG 18911T and B. indicum LMG 11587T. However, in the study of Scardovi et al. (1970), the type strains of B. indicum and B. coryneforme only exhibited 60% DNA-DNA hybridization value. In the present study, the genomes of B. coryneforme CGMCC 1.2279T, B. coryneforme JCM 5819T, B. indicum JCM 1302T, B. indicum CGMCC 1.2275T, B. indicum DSM 20214T, B. indicum LMG 27437T, B. indicum ATCC 25912T, B. indicum KCTC 3230T, B. indicum CCUG 34985T, were sequenced, and the taxonomic relationship between B. coryneforme and B. indicum was re-evaluated. On the basis of the results presented here, (i) ATCC 25912 and DSM 20214 deposited by Vittorio Scardovi are two different strains; (ii) the type strain of B. indicum is ATCC 25912T (= JCM 1302T = LMG 27437T = CGMCC 1.2275T = KCTC 3230T), and not DSM 20214 (= BCRC 14674 = CCUG 34985 = LMG 11587); (iii) B. coryneforme and B. indicum represent two different species of the genus Bifidobacterium; (iv) strain DSM 20214 (= BCRC 14674 = CCUG 34985 = LMG 11587) belongs to B. coryneforme.


Subject(s)
Bifidobacterium , DNA, Bacterial , Genome, Bacterial , Phylogeny , Bifidobacterium/genetics , Bifidobacterium/classification , Bifidobacterium/isolation & purification , DNA, Bacterial/genetics , Nucleic Acid Hybridization , Bacterial Typing Techniques , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
3.
Nutrients ; 16(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732599

ABSTRACT

In this study, a systematic review of randomized clinical trials conducted from January 2000 to December 2023 was performed to examine the efficacy of psychobiotics-probiotics beneficial to mental health via the gut-brain axis-in adults with psychiatric and cognitive disorders. Out of the 51 studies involving 3353 patients where half received psychobiotics, there was a notably high measurement of effectiveness specifically in the treatment of depression symptoms. Most participants were older and female, with treatments commonly utilizing strains of Lactobacillus and Bifidobacteria over periods ranging from 4 to 24 weeks. Although there was a general agreement on the effectiveness of psychobiotics, the variability in treatment approaches and clinical presentations limits the comparability and generalization of the findings. This underscores the need for more personalized treatment optimization and a deeper investigation into the mechanisms through which psychobiotics act. The research corroborates the therapeutic potential of psychobiotics and represents progress in the management of psychiatric and cognitive disorders.


Subject(s)
Mental Disorders , Probiotics , Randomized Controlled Trials as Topic , Humans , Probiotics/therapeutic use , Female , Mental Disorders/drug therapy , Mental Disorders/therapy , Cognition Disorders/drug therapy , Male , Treatment Outcome , Adult , Brain-Gut Axis/drug effects , Middle Aged , Gastrointestinal Microbiome/drug effects , Lactobacillus , Aged , Bifidobacterium
4.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791112

ABSTRACT

Probiotic feed additives have attracted considerable research interest in recent years because the effectiveness of probiotics can differ across microbial strains and the supplemented macroorganisms. The present study was conducted on 16 lambs divided equally into two groups (C-control and E-experimental). The examined lambs were aged 11 days at the beginning of the experiment and 40 days at the end of the experiment. The diet of group E lambs was supplemented with a multi-strain probiotic formulation (Lactobacillus plantarum AMT14, Lactobacillus plantarum AMT4, Lactobacillus rhamnosus AMT15, and Bifidobacterium animalis AMT30), whereas group C lambs did not receive the probiotic additive. At the beginning of the experiment (day 0) and on experimental days 15 and 30, blood was sampled from the jugular vein to determine and compare: phagocytic activity (Phagotest) and oxidative metabolism (Phagoburst) of peripheral blood granulocytes and monocytes by flow cytometry. An analysis of the phagocytic activity of granulocytes and monocytes revealed significantly higher levels of phagocytic activity (expressed as the percentage of phagocytic cells and mean fluorescence intensity) in lambs that were administered the multi-strain probiotic formulation compared with lambs in the control group. The probiotic feed additive also exerted a positive effect on the oxidative metabolism of both granulocytes and monocytes (expressed as the percentage of oxidative metabolism and mean fluorescence intensity) after stimulation with Escherichia coli bacteria and with PMA (4-phorbol-12-ß-myristate-13-acetate). These findings suggest that the tested probiotic formulation may have a positive effect on the immune status of lambs.


Subject(s)
Granulocytes , Monocytes , Phagocytosis , Probiotics , Animals , Probiotics/administration & dosage , Probiotics/pharmacology , Phagocytosis/drug effects , Monocytes/metabolism , Monocytes/drug effects , Sheep , Granulocytes/metabolism , Granulocytes/drug effects , Administration, Oral , Oxidation-Reduction/drug effects , Lactobacillus , Animal Feed , Bifidobacterium
5.
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
6.
Food Res Int ; 187: 114417, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763667

ABSTRACT

Resistant starch serves as a prebiotic in the large intestine, aiding in the maintenance of a healthy intestinal environment and mitigating associated chronic illnesses. This study aimed to investigate the impact of resistant starch-enriched brown rice (RBR) on intestinal health and functionality. We assessed changes in resistant starch concentration, structural alterations, and branch chain length distribution throughout the digestion process using an in vitro model. The efficacy of RBR in the intestinal environment was evaluated through analyses of its prebiotic potential, effects on intestinal microbiota, and intestinal function-related proteins in obese animals fed a high-fat diet. RBR exhibited a higher yield of insoluble fraction in both the small and large intestines compared to white and brown rice. The total digestible starch content decreased, while the resistant starch content significantly increased during in vitro digestion. Furthermore, RBR notably enhanced the growth of four probiotic strains compared to white and brown rice, displaying higher proliferation activity than the positive control, FOS. Notably, consumption of RBR by high-fat diet-induced obese mice suppressed colon shortening, increased Bifidobacteria growth, and improved intestinal permeability. These findings underscore the potential prebiotic and gut health-promoting attributes of RBR, offering insights for the development of functional foods aimed at preventing gastrointestinal diseases.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Mice, Inbred C57BL , Obesity , Oryza , Prebiotics , Starch , Animals , Oryza/chemistry , Gastrointestinal Microbiome/drug effects , Mice , Starch/metabolism , Male , Obesity/metabolism , Mice, Obese , Resistant Starch , Probiotics , Digestion , Bifidobacterium/growth & development
7.
Sci Rep ; 14(1): 11181, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755201

ABSTRACT

Gut microbiota manipulation may reverse metabolic abnormalities in obesity. Our previous studies demonstrated that inulin supplementation significantly promoted Bifidobacterium and fat-free mass in obese children. We aimed to study gut-muscle axis from inulin supplementation in these children. In clinical phase, the plasma samples from 46 participants aged 7-15 years, were analyzed for muscle biomarkers before and after 6-month inulin supplementation. In parallel, the plausible mechanism of muscle production via gut-muscle axis was examined using macrophage cell line. Bifidobacterium was cultured in semi-refined medium with inulin used in the clinical phase. Cell-free supernatant was collected and used in lipopolysaccharide (LPS)-induced macrophage cell line to determine inflammatory and anti-inflammatory gene expression. In clinical phase, IL-15 and creatinine/cystatin C ratio significantly increased from baseline to the 6th month. In vitro study showed that metabolites derived from Bifidobacterium capable of utilizing inulin contained the abundance of SCFAs. In the presence of LPS, treatment from Bifidobacterium + inulin downregulated TNF-α, IL-6, IL-1ß, and iNOS, but upregulated FIZZ-1 and TGF-ß expression. Inulin supplementation promoted the muscle biomarkers in agreement with fat-free mass gain, elucidating by Bifidobacterium metabolites derived from inulin digestion showed in vitro anti-inflammatory activity and decreased systemic pro-inflammation, thus promoting muscle production via gut-muscle axis response.Clinical Trial Registry number: NCT03968003.


Subject(s)
Bifidobacterium , Dietary Supplements , Gastrointestinal Microbiome , Inulin , Inulin/pharmacology , Inulin/administration & dosage , Humans , Child , Adolescent , Male , Gastrointestinal Microbiome/drug effects , Female , Biomarkers , Pediatric Obesity/metabolism , Macrophages/metabolism , Macrophages/drug effects , Lipopolysaccharides , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects
8.
Nutrients ; 16(9)2024 May 06.
Article in English | MEDLINE | ID: mdl-38732641

ABSTRACT

Numerous studies have investigated the immunomodulatory effects of yogurt, but the underlying mechanism remained elusive. This study aimed to elucidate the alleviating properties of yogurt on immunosuppression and proposed the underlying mechanism was related to the metabolite D-lactate. In the healthy mice, we validated the safety of daily yogurt consumption (600 µL) or D-lactate (300 mg/kg). In immunosuppressed mice induced by cyclophosphamide (CTX), we evaluated the immune regulation of yogurt and D-lactate. The result showed that yogurt restored body weight, boosted immune organ index, repaired splenic tissue, recovered the severity of delayed-type hypersensitivity reactions and increased serum cytokines (IgA, IgG, IL-6, IFN-γ). Additionally, yogurt enhanced intestinal immune function by restoring the intestinal barrier and upregulating the abundance of Bifidobacterium and Lactobacillus. Further studies showed that D-lactate alleviated immunosuppression in mice mainly by promoting cellular immunity. D-lactate recovered body weight and organ development, elevated serum cytokines (IgA, IgG, IL-6, IFN-γ), enhanced splenic lymphocyte proliferation and increased the mRNA level of T-bet in splenic lymphocyte to bolster Th1 differentiation. Finally, CTX is a chemotherapeutic drug, thus, the application of yogurt and D-lactate in the tumor-bearing mouse model was initially explored. The results showed that both yogurt (600 µL) and D-lactate (300 mg/kg) reduced cyclophosphamide-induced immunosuppression without promoting tumor growth. Overall, this study evaluated the safety, immune efficacy and applicability of yogurt and D-lactate in regulating immunosuppression. It emphasized the potential of yogurt as a functional food for immune regulation, with D-lactate playing a crucial role in its immunomodulatory effects.


Subject(s)
Cyclophosphamide , Cytokines , Lactic Acid , Yogurt , Animals , Mice , Lactic Acid/blood , Cytokines/metabolism , Male , Immunosuppression Therapy , Spleen/drug effects , Spleen/metabolism , Spleen/immunology , Mice, Inbred BALB C , Hypersensitivity, Delayed/immunology , Gastrointestinal Microbiome/drug effects , Lactobacillus , Bifidobacterium
9.
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
10.
Gut Microbes ; 16(1): 2357176, 2024.
Article in English | MEDLINE | ID: mdl-38798019

ABSTRACT

Resistance to antibiotics in newborns is a huge concern as their immune system is still developing, and infections and resistance acquisition in early life have short- and long-term consequences for their health. Bifidobacterium species are important commensals capable of dominating the infant gut microbiome and are known to be less prone to possess antimicrobial resistance genes than other taxa that may colonize infants. We aimed to study the association between Bifidobacterium-dominated infant gut microbiota and the antibiotic resistant gene load in neonates, and to ascertain the perinatal factors that may contribute to the antibiotic resistance acquisition. Two hundred infant fecal samples at 7 days and 1 month of age from the MAMI birth cohort were included in the study and for whom maternal-neonatal clinical records were available. Microbiota profiling was carried out by 16S rRNA amplicon sequencing, and targeted antibiotic resistance genes (ARGs) including tetM, tetW, tetO, blaTEM, blaSHV and ermB were quantified by qPCR. Infant microbiota clustered into two distinct groups according to their Bifidobacterium genus abundance: high and low. The main separation of groups or clusters at each time point was performed with an unsupervised non-linear algorithm of k-means partitioning to cluster data by time points based on Bifidobacterium genus relative abundance. Microbiota composition differed significantly between both groups, and specific bifidobacterial species were enriched in each cluster. Lower abundance of Bifidobacterium in the infant gut was associated with a higher load of antibiotic resistance genes. Our results highlight the relevance of Bifidobacterium genus in the early acquisition and establishment of antibiotic resistance in the gut. Further studies are needed to develop strategies to promote a healthy early colonization and fight against the spread of antibiotic resistances.


Subject(s)
Anti-Bacterial Agents , Bifidobacterium , Drug Resistance, Bacterial , Feces , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Bifidobacterium/genetics , Bifidobacterium/drug effects , Bifidobacterium/isolation & purification , Infant, Newborn , Gastrointestinal Microbiome/drug effects , Feces/microbiology , Anti-Bacterial Agents/pharmacology , Female , RNA, Ribosomal, 16S/genetics , Drug Resistance, Bacterial/genetics , Male , Infant
11.
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
12.
Sci Rep ; 14(1): 11560, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773299

ABSTRACT

IBD is a disorder which could be caused by oxidative stress. This investigation aims to determine if probiotics and postbiotics can control oxidative stress and inflammation and compare the effectiveness of these two probiotic and postbiotic mixtures of substances. 88 strains of Lactobacillus and Bifidobacterium were tested for antioxidant activity. Male wild-type C57BL/6 mice were divided into four experimental groups, namely high fat diet (HFD) + PBS, HFD + DSS, HFD + DSS + 109 cfu/ml of probiotics, and HFD + DSS + 109 cfu/ml of postbiotics. The phenotypical indices and pathological scores were assessed. The expression of genes related to NF-kB and Nrf2 signaling pathways and enzymes associated with oxidant/anti-oxidant activities, and proinflammatory/inflammatory cytokines were assessed. In contrast to the groups exposed to DSS, mice treated with probiotics mixture and postbiotics mixture alongside DSS displayed alleviation of DSS-induced adverse effects on phenotypical characteristics, as well as molecular indices such as the Nrf2 and NF-kB related genes, with a greater emphasis on the postbiotics component. In accordance with the findings of the present investigation, it can be inferred that even in using a high-fat dietary regimen as an inducer of oxidative stress, the emergence of inflammation can be effectively addressed through the utilization of probiotics and, more specifically, postbiotics.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Colitis , Dextran Sulfate , Disease Models, Animal , Mice, Inbred C57BL , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Probiotics , Signal Transduction , Animals , Probiotics/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Male , Mice , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Anti-Inflammatory Agents/pharmacology , Signal Transduction/drug effects , Oxidative Stress/drug effects , Lactobacillus , Bifidobacterium , Diet, High-Fat/adverse effects
13.
Cancer Immunol Immunother ; 73(5): 94, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38564002

ABSTRACT

The advent of tumor immunotherapy in patients has revolutionized the treatment of tumors and significantly improved survival rates for a wide range of tumors. However, the full therapeutic potential of immune checkpoint inhibitors (ICIs) has yet to be realized, as not all patients have a lasting survival benefit from them, and a significant proportion of patients show primary or acquired resistance to immunotherapy. Bifidobacterium is one of the most common probiotics, and its antitumor and immunomodulatory effects have been demonstrated in recent years, but its immunomodulatory effects in tumors, especially on ICIs and in combination, have not been extensively studied in clinical practice, and its effects on the immune system and the mechanisms that modulate immunotherapy are largely unknown. Therefore, this review will focus on the immunomodulatory effects of Bifidobacteria in malignancies and the possible mechanisms of action of Bifidobacteria on immunotherapy in the hope of providing a basis for further research and better application of Bifidobacteria in clinical practice.


Subject(s)
Immunomodulation , Immunotherapy , Humans , Bifidobacterium , Immune Checkpoint Inhibitors
14.
Article in English | MEDLINE | ID: mdl-38661726

ABSTRACT

A novel bifidobacterium (designated F753-1T) was isolated from the gut of honeybee (Apis mellifera). Strain F753-1T was characterized using a polyphasic taxonomic approach. Strain F753-1T was phylogenetically related to the type strains of Bifidobacterium mizhiensis, Bifidobacterium asteroides, Bifidobacterium choladohabitans, Bifidobacterium mellis, Bifidobacterium apousia and Bifidobacterium polysaccharolyticum, having 98.4-99.8 % 16S rRNA gene sequence similarities. The phylogenomic tree indicated that strain F753-1T was most closely related to the type strains of B. mellis and B. choladohabitans. Strain F753-1T had the highest average nucleotide identity (94.1-94.5 %) and digital DNA-DNA hybridization (56.3 %) values with B. mellis Bin7NT. Acid production from amygdalin, d-fructose, gentiobiose, d-mannose, maltose, sucrose and d-xylose, activity of α-galactosidase, pyruvate utilization and hydrolysis of hippurate could differentiate strain F753-1T from B. mellis CCUG 66113T and B. choladohabitans JCM 34586T. Based upon the data obtained in the present study, a novel species, Bifidobacterium apis sp. nov., is proposed, and the type strain is F753-1T (=CCTCC AB 2023227T=JCM 36562T=LMG 33388T).


Subject(s)
Bacterial Typing Techniques , Bifidobacterium , DNA, Bacterial , Nucleic Acid Hybridization , Phylogeny , RNA, Ribosomal, 16S , Sequence Analysis, DNA , Bees/microbiology , Animals , RNA, Ribosomal, 16S/genetics , Bifidobacterium/isolation & purification , Bifidobacterium/classification , Bifidobacterium/genetics , DNA, Bacterial/genetics , Fatty Acids , Base Composition , Gastrointestinal Microbiome
15.
Sci Rep ; 14(1): 7983, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38575668

ABSTRACT

Dimension reduction has been used to visualise the distribution of multidimensional microbiome data, but the composite variables calculated by the dimension reduction methods have not been widely used to investigate the relationship of the human gut microbiome with lifestyle and disease. In the present study, we applied several dimension reduction methods, including principal component analysis, principal coordinate analysis (PCoA), non-metric multidimensional scaling (NMDS), and non-negative matrix factorization, to a microbiome dataset from 186 subjects with symptoms of  allergic rhinitis (AR) and 106 controls. All the dimension reduction methods supported that the distribution of microbial data points appeared to be continuous rather than discrete. Comparison of the composite variables calculated from the different dimension reduction methods showed that the characteristics of the composite variables differed depending on the distance matrices and the dimension reduction methods. The first composite variables calculated from PCoA and NMDS with the UniFrac distance were strongly associated with AR (FDR adjusted P = 2.4 × 10-4 for PCoA and P = 2.8 × 10-4 for NMDS), and also with the relative abundance of Bifidobacterium and Prevotella. The abundance of Bifidobacterium was also linked to intake of several nutrients, including carbohydrate, saturated fat, and alcohol via composite variables. Notably, the association between the composite variables and AR was much stronger than the association between the relative abundance of individual genera and AR. Our results highlight the usefulness of the dimension reduction methods for investigating the association of microbial composition with lifestyle and disease in clinical research.


Subject(s)
Gastrointestinal Microbiome , Rhinitis, Allergic , Humans , Bifidobacterium , Prevotella , Multidimensional Scaling Analysis
16.
Food Res Int ; 184: 114246, 2024 May.
Article in English | MEDLINE | ID: mdl-38609225

ABSTRACT

Food-derived mucins are glycoproteins rich in sialic acid, but their digestive properties and potential health benefits for humans have been scarcely investigated. In this work, ovomucin (OVM, rich in N-acetylneuraminic acid, about 3 %), porcine small intestinal mucin (PSIM, rich in N-glycolylneuraminic acid, about 1 %), the desialylated OVM (AOVM) and the desialylated PSIM (APSIM) were selected to examine their digestion and their impact on the gut microbiota of elderly individuals. The results shown that, the proportion of low-molecular-weight proteins increased after simulated digestion of these four mucins, with concomitant comparable antioxidant activity observed. Desialylation markedly increased the degradation and digestion rate of mucins. In vitro fecal fermentation was conducted with these mucins using fecal samples from individuals of different age groups: young, low-age and high-age elderly. Fecal fermentation with mucin digestive solution stimulated the production of organic acids in the group with fecal sample of the elderly individuals. Among them, the OVM group demonstrated the most favorable outcomes. The OVM and APSIM groups elevated the relative abundance of beneficial bacteria such as Lactobacillus and Bifidobacterium, while diminishing the presence of pathogenic bacteria such as Klebsiella. Conversely, the probiotic effects of AOVM and PSIM were attenuated or even exhibited adverse effects. Hence, mucins originating from different sources and possessing distinct glycosylation patterns exhibit diverse biological functions. Our findings can offer valuable insights for developing a well-balanced and nutritious diet tailored to the elderly population.


Subject(s)
Gastrointestinal Microbiome , Mucins , Humans , Aged , Animals , Swine , Diet , Food , Bifidobacterium
17.
Cell Host Microbe ; 32(4): 573-587.e5, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38569545

ABSTRACT

Microbiota assembly in the infant gut is influenced by diet. Breastfeeding and human breastmilk oligosaccharides promote the colonization of beneficial bifidobacteria. Infant formulas are supplemented with bifidobacteria or complex oligosaccharides, notably galacto-oligosaccharides (GOS), to mimic breast milk. To compare microbiota development across feeding modes, this randomized controlled intervention study (German Clinical Trial DRKS00012313) longitudinally sampled infant stool during the first year of life, revealing similar fecal bacterial communities between formula- and breast-fed infants (N = 210) but differences across age. Infant formula containing GOS sustained high levels of bifidobacteria compared with formula containing B. longum and B. breve or placebo. Metabolite and bacterial profiling revealed 24-h oscillations and circadian networks. Rhythmicity in bacterial diversity, specific taxa, and functional pathways increased with age and was strongest following breastfeeding and GOS supplementation. Circadian rhythms in dominant taxa were further maintained ex vivo in a chemostat model. Hence, microbiota rhythmicity develops early in life and is impacted by diet.


Subject(s)
Infant Formula , Microbiota , Infant , Female , Humans , Infant Formula/microbiology , Breast Feeding , Milk, Human , Bifidobacterium , Feces/microbiology , Oligosaccharides/metabolism , Circadian Rhythm
18.
Nutrients ; 16(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38674840

ABSTRACT

Throughout infancy, IgA is crucial for maintaining gut mucosal immunity. This study aims to determine whether supplementing newborn mice with eight different strains of Bifidobacterium longum subsp. infantis might regulate their IgA levels. The strains were gavaged to BALB/C female (n = 8) and male (n = 8) dams at 1-3 weeks old. Eight strains of B. longum subsp. infantis had strain-specific effects in the regulation of intestinal mucosal barriers. B6MNI, I4MI, and I10TI can increase the colonic IgA level in females and males. I8TI can increase the colonic IgA level in males. B6MNI was also able to significantly increase the colonic sIgA level in females. B6MNI, I4MI, I8TI, and I10TI regulated colonic and Peyer's patch IgA synthesis genes but had no significant effect on IgA synthesis pathway genes in the jejunum and ileum. Moreover, the variety of sIgA-coated bacteria in male mice was changed by I4MI, I5TI, I8TI, and B6MNI. These strains also can decrease the relative abundance of Escherichia coli. These results indicate that B. longum subsp. infantis can promote IgA levels but show strain specificity. Different dietary habits with different strains of Bifidobacterium may have varying effects on IgA levels when supplemented in early infancy.


Subject(s)
Bifidobacterium longum subspecies infantis , Bifidobacterium , Immunoglobulin A , Intestinal Mucosa , Mice, Inbred BALB C , Probiotics , Animals , Female , Male , Immunoglobulin A/metabolism , Mice , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Probiotics/administration & dosage , Gastrointestinal Microbiome , Animals, Newborn , Intestines/microbiology , Intestines/immunology , Immunity, Mucosal , Species Specificity , Colon/microbiology , Colon/immunology , Colon/metabolism , Immunoglobulin A, Secretory/metabolism
19.
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
20.
Genes (Basel) ; 15(4)2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38674400

ABSTRACT

Bifidobacterium longum subsp. infantis YLGB-1496 (YLGB-1496) is a probiotic strain isolated from human breast milk. The application of YLGB-1496 is influenced by carbohydrate utilization and genetic stability. This study used genome sequencing and morphology during continuous subculture to determine the carbohydrate utilization characteristics and genetic stability of YLGB-1496. The complete genome sequence of YLGB-1496 consists of 2,758,242 base pairs, 2442 coding sequences, and a GC content of 59.87%. A comparison of carbohydrate transport and metabolism genes of Bifidobacterium longum subsp. infantis (B. infantis) showed that YLGB-1496 was rich in glycosyl hydrolase 13, 20, 25, and 109 gene families. During continuous subculture, the growth characteristics and fermentation activity of the strain were highly stable. The bacterial cell surface and edges of the 1000th-generation strains were progressively smoother and well-defined, with no perforations or breaks in the cell wall. There were 20 SNP loci at the 1000th generation, fulfilling the requirement of belonging to the same strain. The presence of genes associated with cell adhesion and the absence of resistance genes supported the probiotic characteristics of the strain. The data obtained in this study provide insights into broad-spectrum carbohydrate utilization, genomic stability, and probiotic properties of YLGB-1496, which provide theoretical support to promote the use of YLGB-1496.


Subject(s)
Bifidobacterium , Carbohydrate Metabolism , Genome, Bacterial , Bifidobacterium/genetics , Bifidobacterium/metabolism , Carbohydrate Metabolism/genetics , Humans , Probiotics , Genomic Instability , Bifidobacterium longum subspecies infantis/genetics , Bifidobacterium longum subspecies infantis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...