Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Gut Microbes ; 16(1): 2363021, 2024.
Article in English | MEDLINE | ID: mdl-38860973

ABSTRACT

Dietary fiber supplements are a strategy to close the 'fiber gap' and induce targeted modulations of the gut microbiota. However, higher doses of fiber supplements cause gastrointestinal (GI) symptoms that differ among individuals. What determines these inter-individual differences is insufficiently understood. Here we analyzed findings from a six-week randomized controlled trial that evaluated GI symptoms to corn bran arabinoxylan (AX; n = 15) relative to non-fermentable microcrystalline cellulose (MCC; n = 16) at efficacious supplement doses of 25 g/day (females) or 35 g/day (males) in adults with excess weight. Self-reported flatulence, bloating, and stomach aches were evaluated weekly. Bacterial taxa involved in AX fermentation were identified by bioorthogonal non-canonical amino acid tagging. Associations between GI symptoms, fecal microbiota features, and diet history were systematically investigated. AX supplementation increased symptoms during the first three weeks relative to MCC (p < 0.05, Mann-Whitney tests), but subjects 'adapted' with symptoms reverting to baseline levels toward the end of treatment. Symptom adaptations were individualized and correlated with the relative abundance of Bifidobacterium longum at baseline (rs = 0.74, p = 0.002), within the bacterial community that utilized AX (rs = 0.69, p = 0.006), and AX-induced shifts in acetate (rs = 0.54, p = 0.039). Lower baseline consumption of animal-based foods and higher whole grains associated with less severity and better adaptation. These findings suggest that humans do 'adapt' to tolerate efficacious fiber doses, and this process is linked to their microbiome and dietary factors known to interact with gut microbes, providing a basis for the development of strategies for improved tolerance of dietary fibers.


Subject(s)
Bifidobacterium longum , Dietary Fiber , Feces , Gastrointestinal Microbiome , Xylans , Xylans/metabolism , Humans , Feces/microbiology , Feces/chemistry , Male , Female , Dietary Fiber/metabolism , Middle Aged , Gastrointestinal Microbiome/drug effects , Bifidobacterium longum/metabolism , Adult , Dietary Supplements/analysis , Fermentation , Aged , Adaptation, Physiological
2.
Carbohydr Polym ; 339: 122248, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823916

ABSTRACT

Arabinoxylan is a major hemicellulose in the sugarcane plant cell wall with arabinose decorations that impose steric restrictions on the activity of xylanases against this substrate. Enzymatic removal of the decorations by arabinofuranosidases can allow a more efficient arabinoxylan degradation by xylanases. Here we produced and characterized a recombinant Bifidobacterium longum arabinofuranosidase from glycoside hydrolase family 43 (BlAbf43) and applied it, together with GH10 and GH11 xylanases, to produce xylooligosaccharides (XOS) from wheat arabinoxylan and alkali pretreated sugarcane bagasse. The enzyme synergistically enhanced XOS production by GH10 and GH11 xylanases, being particularly efficient in combination with the latter family of enzymes, with a degree of synergism of 1.7. We also demonstrated that the enzyme is capable of not only removing arabinose decorations from the arabinoxylan and from the non-reducing end of the oligomeric substrates, but also hydrolyzing the xylan backbone yielding mostly xylobiose and xylose in particular cases. Structural studies of BlAbf43 shed light on the molecular basis of the substrate recognition and allowed hypothesizing on the structural reasons of its multifunctionality.


Subject(s)
Bifidobacterium longum , Cellulose , Endo-1,4-beta Xylanases , Glucuronates , Glycoside Hydrolases , Oligosaccharides , Saccharum , Xylans , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Glycoside Hydrolases/metabolism , Glycoside Hydrolases/chemistry , Glucuronates/metabolism , Glucuronates/chemistry , Endo-1,4-beta Xylanases/metabolism , Endo-1,4-beta Xylanases/chemistry , Xylans/metabolism , Xylans/chemistry , Saccharum/chemistry , Saccharum/metabolism , Cellulose/chemistry , Cellulose/metabolism , Bifidobacterium longum/enzymology , Bifidobacterium longum/metabolism , Hydrolysis , Substrate Specificity , Recombinant Proteins/metabolism , Recombinant Proteins/chemistry , Disaccharides
3.
Gut Microbes ; 16(1): 2353229, 2024.
Article in English | MEDLINE | ID: mdl-38752423

ABSTRACT

Members of the genus Bifidobacterium are commonly found in the human gut and are known to utilize complex carbohydrates that are indigestible by the human host. Members of the Bifidobacterium longum subsp. longum taxon can metabolize various plant-derived carbohydrates common to the human diet. To metabolize such polysaccharides, which include arabinoxylan, bifidobacteria need to encode appropriate carbohydrate-active enzymes in their genome. In the current study, we describe two GH43 family enzymes, denoted here as AxuA and AxuB, which are encoded by B. longum subsp. longum NCIMB 8809 and are shown to be required for cereal-derived arabinoxylan metabolism by this strain. Based on the observed hydrolytic activity of AxuA and AxuB, assessed by employing various synthetic and natural substrates, and based on in silico analyses, it is proposed that both AxuA and AxuB represent extracellular α-L-arabinofuranosidases with distinct substrate preferences. The variable presence of the axuA and axuB genes and other genes previously described to be involved in the metabolism of arabinose-containing glycans can in the majority cases explain the (in)ability of individual B. longum subsp. longum strains to grow on cereal-derived arabinoxylans and arabinan.


Subject(s)
Bifidobacterium longum , Edible Grain , Glycoside Hydrolases , Xylans , Xylans/metabolism , Glycoside Hydrolases/metabolism , Glycoside Hydrolases/genetics , Edible Grain/microbiology , Edible Grain/metabolism , Bifidobacterium longum/enzymology , Bifidobacterium longum/metabolism , Bifidobacterium longum/genetics , Substrate Specificity , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans
4.
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
5.
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
6.
Kaohsiung J Med Sci ; 40(6): 530-541, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38647095

ABSTRACT

We previously found that the relative abundance of Bifidobacterium was increased after chemotherapy; however, the role of Bifidobacterium longum in chemotherapeutic drug resistance in ovarian cancer (OVC) remains unclear. This study aimed to understand the potential effects and mechanism of B. longum extracellular vesicles (B. longum-EVs) on carboplatin (CBP) resistance in OVC. Eight normal and 11 ovarian tissues were collected and the expression of B. longum genomic DNA and its association with acquired CBP resistance in OVC patients was determined. After isolating EVs by ultracentrifugation from B. longum (ATCC 15707), CBP-resistant A2780 cells were treated with PBS, CBP, B. longum-EVs, or CBP + B. longum-EVs, and subsequently analyzed by CCK-8, Edu staining, Annexin V/PI double staining, wound healing, and Transwell assays to detect cell viability, proliferation, apoptosis, migration, and invasion, respectively. MRP1, ATP7A, ATP7B, and p53 expression as well as p53 phosphorylation were measured by western blot analysis. S15A mutation of p53 was assessed to examine the potential role of p53 Ser15 phosphorylation in CBP-resistant OVC. B. longum levels were elevated and positively associated with CBP resistance in OVC patients. Only high concentrations of B. longum-EVs attenuated A2780 cell proliferation, apoptosis, migration, and invasion. B. longum-EVs exposure significantly enhanced the sensitivity of CBP-resistant A2780 cells to CBP and decreased the expression of drug resistance-related proteins. The effect of B. longum-EVs on reversing CBP resistance was completely inhibited by S15A mutation of p53. B. longum-EVs enhanced the sensitivity of OVC cells to CBP through p53 phosphorylation on Ser15.


Subject(s)
Bifidobacterium longum , Carboplatin , Drug Resistance, Neoplasm , Extracellular Vesicles , Ovarian Neoplasms , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Female , Phosphorylation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Extracellular Vesicles/metabolism , Carboplatin/pharmacology , Carboplatin/therapeutic use , Cell Line, Tumor , Bifidobacterium longum/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects
7.
Appl Microbiol Biotechnol ; 108(1): 215, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38363367

ABSTRACT

The metabolite urolithin A, a metabolite of the dietary polyphenol ellagic acid (EA), has significant health benefits for humans. However, studies on the gut microbiota involved in ellagic acid metabolism are limited. In this study, we conducted in vitro fermentation of EA using human intestinal microbiome combined with antibiotics (vancomycin, polymyxin B sulfate, and amphotericin B). Liquid chromatography-mass spectrometry (LC-MS/MS) analysis demonstrated that the production capacity of urolithin A by gut microbiota co-treated with polymyxin B sulfate and amphotericin B (22.39 µM) was similar to that of untreated gut microbiota (24.26 µM). Macrogenomics (high-throughput sequencing) was used to analyze the composition and structure of the gut microbiota. The results showed that the abundance of Bifidobacterium longum, Bifidobacterium adolescentis, and Bifidobacterium bifidum in the gut microbiota without antibiotic treatment or co-treated with polymyxin B sulfate and amphotericin B during EA fermentation was higher than that in other antibiotic treatment gut microbiota. Therefore, B. longum, B. adolescentis, and B. bifidum may be new genera involved in the conversion of EA to urolithin A. In conclusion, the study revealed unique interactions between polyphenols and gut microbiota, deepening our understanding of the relationship between phenolic compounds like EA and the gut microbiota. These findings may contribute to the development of gut bacteria as potential probiotics for further development. KEY POINTS: • Intestinal microbiome involved in ellagic acid metabolism. • Gram-positive bacteria in the intestinal microbiome are crucial for ellagic acid metabolism. • Bifidobacterium longum, Bifidobacterium adolescentis, and Bifidobacterium bifidum participate in ellagic acid metabolism.


Subject(s)
Bifidobacterium longum , Coumarins , Gastrointestinal Microbiome , Humans , Ellagic Acid/metabolism , Chromatography, Liquid , Polymyxin B , Amphotericin B , Tandem Mass Spectrometry , Bifidobacterium longum/metabolism , Anti-Bacterial Agents
8.
NPJ Biofilms Microbiomes ; 10(1): 13, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38396001

ABSTRACT

Both gut microbiome and microRNAs (miRNAs) play a role in the development of hepatic encephalopathy (HE). However, the functional link between the microbiome and host-derived miRNAs in faeces remains poorly understood. In the present study, patients with HE had an altered gut microbiome and faecal miRNAs compared with patients with chronic hepatitis B. Transferring faeces and faecal miRNAs from patients with HE to the recipient mice aggravated thioacetamide-induced HE. Oral gavage of hsa-miR-7704, a host-derived miRNA highly enriched in faeces from patients with HE, aggravated HE in mice in a microbiome-dependent manner. Mechanistically, hsa-miR-7704 inhibited the growth and adhesion of Bifidobacterium longum by suppressing proB. B. longum and its metabolite acetate alleviated HE by inhibiting microglial activation and ammonia production. Our findings reveal the role of miRNA-microbiome axis in HE and suggest that faecal hsa-miR-7704 are potential regulators of HE progression.


Subject(s)
Bifidobacterium longum , Hepatic Encephalopathy , MicroRNAs , Animals , Humans , Mice , Bifidobacterium longum/genetics , Bifidobacterium longum/metabolism , Feces/microbiology , Hepatic Encephalopathy/genetics , Hepatic Encephalopathy/microbiology , MicroRNAs/genetics , MicroRNAs/metabolism
9.
J Agric Food Chem ; 71(46): 17819-17832, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37906736

ABSTRACT

The immunomodulatory potential of certain bacterial strains suggests that they could be beneficial in the treatment of rheumatoid arthritis (RA). In this study, we investigated the effects of Bifidobacterium longum subsp. infantis B6MNI on the progression of collagen-induced arthritis (CIA) in rats as well as its influence on the gut microbiota and fecal metabolites. Forty-eight female Wistar rats were divided into six groups that included a B6MNI group with CIA and intragastrically administered B. longum subsp. infantis B6MNI (109 CFU/day/rat), a control group (CON), and a CIA group, both of which were intracardiacally administered the same volume of saline. Rats were sacrificed after short-term (ST, 4 weeks) or long-term (LT, 6 weeks) administration. The results indicate that B. longum subsp. infantis B6MNI can modulate the gut microbiota and fecal metabolites, including 5-hydroxyindole-3-acetic acid (5-HIAA), which in turn impacts the expression of Pim-1 and immune cell differentiation, then through the JAK-STAT3 pathway affects joint inflammation, regulates osteoclast differentiation factors, and delays the progression of RA. Our results also suggest that B. longum subsp. infantis B6MNI is most efficacious for the early or middle stages of RA.


Subject(s)
Arthritis, Experimental , Bifidobacterium longum , Female , Rats , Animals , Bifidobacterium/metabolism , Hydroxyindoleacetic Acid/metabolism , Arthritis, Experimental/drug therapy , Rats, Wistar , Inflammation/drug therapy , Bifidobacterium longum/metabolism
10.
Cell Stress Chaperones ; 28(6): 909-920, 2023 11.
Article in English | MEDLINE | ID: mdl-37828395

ABSTRACT

Oxidative stress is implicated in numerous diseases, with benzo(α)pyrene (BaP) known for causing substantial oxidative damage. Bifidobacterium longum (B. longum) is recognized as an antioxidant bacterium for certain hosts, yet its influence on oxidative damages instigated by BaP remains undetermined. In our study, we introduced various strains of Caenorhabditis elegans (C. elegans) to BaP to trigger oxidative stress, subsequently treating them with different forms of B. longum to evaluate its protective effects. Additionally, we explored the role of daf-16 in this context. Our findings indicated that in wild-type N2 C. elegans, B. longum-even in the form of inactivated bacteria or bacterial ultrasonic lysates (BULs)-significantly extended lifespan. BaP exposure notably decreased lifespan, superoxide dismutase (SOD) activity, and motility, while simultaneously down-regulating the expression of reactive oxygen species (ROS)-associated genes (sod-3, sek-1, cat-1) and daf-16 downstream genes (sod-3, ctl-2). However, it significantly increased the ROS level, malondialdehyde (MDA) content, and lipofuscin accumulation and up-regulated another daf-16 downstream gene (clk-1) (P <0.05). Interestingly, when further treated with B. longum peptide-1 (BLP-1), opposite effects were observed, and all the aforementioned indices changed significantly. In the case of RNAi (daf-16) C. elegans, BaP exposure significantly shortened the lifespan (P <0.05), which was only slightly prolonged upon further treatment with BLP-1. Furthermore, the expression of daf-16 downstream genes showed minor alterations in RNAi C. elegans upon treatment with either BaP or BLP-1. In conclusion, our findings suggest that B. longum acts as a probiotic for C. elegans. BLP-1 was shown to safeguard C. elegans from numerous oxidative damages induced by BaP, but these protective effects were contingent upon the daf-16 gene.


Subject(s)
Bifidobacterium longum , Caenorhabditis elegans Proteins , Animals , Caenorhabditis elegans/metabolism , Reactive Oxygen Species/metabolism , Benzo(a)pyrene/toxicity , Benzo(a)pyrene/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Bifidobacterium longum/metabolism , Oxidative Stress , Peptides/metabolism , Superoxide Dismutase/metabolism , Longevity , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/pharmacology
11.
Int J Mol Sci ; 24(16)2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37628988

ABSTRACT

Melanin produced by melanocytes protects our skin against ultraviolet (UV) radiation-induced cell damage and oxidative stress. Melanin overproduction by hyperactivated melanocytes is the direct cause of skin hyperpigmentary disorders, such as freckles and melasma. Exploring natural whitening agents without the concern of toxicity has been highly desired. In this study, we focused on a Bifidobacterium longum strain, ZJ1, isolated from a Chinese centenarian, and we evaluated the anti-melanogenic activity of the distinctive extracts of ZJ1. Our results demonstrated that whole lysate (WL) and bacterial lysate (BL) of ZJ1 ferments efficiently reduce α-melanocyte-stimulating hormone (α-MSH)-induced melanin production in B16-F10 cells as well as the melanin content in zebrafish embryos. BL and WL downregulate melanogenesis-related gene expression and indirectly inhibit intracellular tyrosinase activity. Furthermore, they both showed antioxidant activity in a menadione-induced zebrafish embryo model. Our results suggest that ZJ1 fermentation lysates have application potential as therapeutic reagents for hyperpigmentary disorders and whitening agents for cosmetics.


Subject(s)
Antioxidants , Bifidobacterium longum , Bleaching Agents , Hyperpigmentation , Melanins , Animals , Humans , Antioxidants/pharmacology , Bifidobacterium longum/isolation & purification , Bifidobacterium longum/metabolism , Centenarians , East Asian People , Hyperpigmentation/drug therapy , Hyperpigmentation/metabolism , Melanins/metabolism , Zebrafish , Aged, 80 and over
12.
Nutrients ; 15(15)2023 Jul 29.
Article in English | MEDLINE | ID: mdl-37571319

ABSTRACT

Neuropsychiatric disorders including Alzheimer's disease (AD) may cause gut inflammation and dysbiosis. Gut inflammation-suppressing probiotics alleviate neuropsychiatric disorders. Herein, to understand whether anti-inflammatory probiotics Lactobacillus mucosae NK41 and Bifidobacterium longum NK46, which suppressed tumor necrosis factor (TNF)-α expression in lipopolysaccharide (LPS)-stimulated macrophages, could alleviate cognitive impairment, we first examined their effects on cognitive function, gut inflammation, and gut microbiota composition in 5xFAD-transgenic mice. Oral administration of NK41 or NK46 decreased cognitive impairment-like behaviors, hippocampal amyloid-ß (Aß), TNF-α and interleukin (IL)-1ß expression, hippocampal NF-κB+Iba1+ cell population, and Aß accumulation, while hippocampal brain-derived neurotropic factor (BDNF) and IL-10 expression and BDNF+NeuN+ cell population increased. They also decreased TNF-α and IL-1ß expression and NF-κB+CD11c+ cell population in the colon. They also reduced fecal and blood LPS levels and gut Proteobacteria and Verrucomicrobia populations (including Akkkermansiaceae), which are positively associated with hippocampal TNF-α and fecal LPS levels and negatively correlated with hippocampal BDNF level. However, they increased Odoribactericeae, which positively correlated with BDNF expression level and TNF-α to IL-10 expression ratio. The combination of NK41 and NK46 (4:1, NKc), which potently inhibited TNF-α expression in LPS-stimulated macrophages, additively alleviated cognitive impairment-like behaviors in 5xFAD-transgenic or aged mice. NKc increased hippocampal BDNF+NeuN+ cell population and BDNF expression in 5xFAD-transgenic or aged mice, while hippocampal TNF-α and IL-1ß expression decreased. NKc also decreased TNF-α and IL-1ß expression in the colon and LPS levels in the blood and feces. These findings suggest that gut bacteria and its product LPS may be closely connected with occurrence of cognitive impairment and neuroinflammation and the combination of NK41 and NK46 can additively alleviate cognitive impairment and neuroinflammation by inducing NF-κB-suppressed BDNF expression and suppressing LPS-producing gut bacteria.


Subject(s)
Bifidobacterium longum , Cognitive Dysfunction , Colitis , Animals , Mice , Bifidobacterium longum/metabolism , Interleukin-10 , Tumor Necrosis Factor-alpha/metabolism , Neuroinflammatory Diseases , NF-kappa B/metabolism , Dysbiosis/complications , Lipopolysaccharides/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Colitis/microbiology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/complications , Mice, Transgenic , Inflammation/complications , Mice, Inbred C57BL
13.
Sci Rep ; 13(1): 11076, 2023 07 08.
Article in English | MEDLINE | ID: mdl-37422465

ABSTRACT

Polyphosphate (poly-P) biosynthesis in bacteria has been linked to many physiological processes and has been characterized as an interesting functional molecule involved in intestinal homeostasis. We determined the capacity for poly-P production of 18 probiotic strains mainly belonging to Bifidobacterium and former Lactobacillus genera, showing that poly-P synthesis varied widely between strains and is dependent on the availability of phosphate and the growth phase. Bifidobacteria were especially capable of poly-P synthesis and poly-P kinase (ppk) genes were identified in their genomes together with a repertoire of genes involved in phosphate transport and metabolism. In Bifidobacterium longum KABP042, the strain we found with highest poly-P production, variations in ppk expression were linked to growth conditions and presence of phosphate in the medium. Moreover, the strain produced poly-P in presence of breast milk and lacto-N-tetraose increased the amount of poly-P synthesized. Compared to KABP042 supernatants low in poly-P, exposure of Caco-2 cells to KABP042 supernatants rich in poly-P resulted in decreased epithelial permeability and increased barrier resistance, induction of epithelial protecting factors such as HSP27 and enhanced expression of tight junction protein genes. These results highlight the role of bifidobacteria-derived poly-P as a strain-dependent functional factor acting on epithelial integrity.


Subject(s)
Bifidobacterium longum , Probiotics , Female , Humans , Bifidobacterium longum/metabolism , Polyphosphates/metabolism , Caco-2 Cells , Intestines/microbiology , Bifidobacterium
14.
Food Funct ; 14(11): 5355-5374, 2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37212199

ABSTRACT

Irritable bowel syndrome (IBS) is a functional intestinal disorder without clear pathological mechanisms. Classical treatments for IBS are not always effective and are usually accompanied by side effects. Selenium-enriched Bifidobacterium longum DD98 (Se-B. longum DD98) is a selenized probiotic strain which has shown many beneficial effects on the gastrointestinal tract, but its effects on IBS and the underlying mechanism are unclear. This study aims to investigate the relieving effects of Se-B. longum DD98 on chronic unpredictable mild stress (CUMS)-induced IBS in mice. The model mice were treated with saline, B. longum DD98, or Se-B. longum DD98 while receiving CUMS. The results suggest that Se-B. longum DD98 significantly relieved the intestinal symptoms of IBS mice and reduced intestinal permeability and inflammation. The depression and anxiety-like behaviors of IBS mice were also improved by Se-B. longum DD98. In addition, the expression of serotonin (5-HT), γ-aminobutyric acid (GABA), neuropeptide Y (NPY), and brain-derived neurotrophic factor (BDNF), which are indicators closely related to mood and brain-gut axis, were up-regulated in mice treated with Se-B. longum DD98. Furthermore, the 16S rRNA sequencing study showed that Se-B. longum DD98 effectively restored the relative abundance of intestinal microbes (e.g., Lactobacillus, Desulfovibrio, Akkermansia) and regulated the impaired diversity of gut microbiota in IBS mice. These results suggest that Se-B. longum DD98 positively acts on the brain-gut axis by improving intestinal functions and regulating mood-associated behaviors and indicators of IBS mice. Therefore, this Se-enriched probiotic strain could be considered a promising candidate for the alleviation of CUMS-induced IBS.


Subject(s)
Bifidobacterium longum , Irritable Bowel Syndrome , Probiotics , Selenium , Mice , Animals , Irritable Bowel Syndrome/microbiology , Bifidobacterium longum/metabolism , Selenium/metabolism , RNA, Ribosomal, 16S/metabolism , Intestines , Probiotics/pharmacology
15.
J Agric Food Chem ; 71(23): 8915-8930, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37255290

ABSTRACT

Changes in the functions of the intestinal barrier occur in parallel with the development of sepsis. The protection by Bifidobacterium strains (BB, BL, BB12, and BLBB) was evaluated in mice injected with lipopolysaccharide (LPS). The results revealed an increase in the ratio of ileal villus length to crypt depth in the BLBB group compared with that in the LPS group, as were the number of IgA+ plasma, CD4+/CD8+ T, and dendritic cells. The levels of diamine oxidase (DAO) and d-lactic acid in the serum were lessened in the BLBB group after LPS injection compared with that in the LPS group. In addition, the BLBB group exhibited an increased expression level of tight junction proteins (zonula occludens-1, occludin, and claudin-1), mucin (MUC2) mRNA, reduced NFκB/MAPK signaling pathways, and decreased expression levels of inflammatory cytokines (IL-1ß, IL-6, and TNF-α). The BLBB group enriched the relative abundance of Muribaculaceae, Lachnospiraceae_NK4A136_group, Clostridia_Ucg-014, and Alistipes, resulting in an increase in strains producing short-chain fatty acids. Furthermore, the BLBB group leads to higher levels of deoxycholic acid and biosynthesized linoleate. This study suggested that the BLBB group could enhance the capacity of the intestinal barrier and intestinal mucosal immunity, reduce intestinal inflammation, and improve the composition of gut microbiota. Bifidobacterium bifidum E3 combined with Bifidobacterium longum subsp. infantis E4 may thus serve as a probiotic against the intestinal injury caused by LPS.


Subject(s)
Bifidobacterium bifidum , Bifidobacterium longum , Intestinal Diseases , Mice , Animals , Lipopolysaccharides/adverse effects , Lipopolysaccharides/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , MAP Kinase Signaling System , Bifidobacterium longum/genetics , Bifidobacterium longum/metabolism
16.
Food Res Int ; 167: 112711, 2023 05.
Article in English | MEDLINE | ID: mdl-37087214

ABSTRACT

Arabinoxylan (AX) and arabinoxylo-oligosaccharides (AXOS) derived therefrom are emergent prebiotics with promising health promoting properties, likely linked to its capacity to foster beneficial species in the human gut. Bifidobacteria appear to be one taxa that is frequently promoted following AX or AXOS consumption, and that is known to establish metabolic cross-feeding networks with other beneficial commensal species. Therefore, probiotic bifidobacteria with the capability to metabolize AX-derived prebiotics represent interesting candidates to develop novel probiotic and synbiotic combinations with AX-based prebiotics. In this work we have deepen into the metabolic capabilities of bifidobacteria related to AX and AXOS metabolization through a combination of in silico an in vitro tools. Both approaches revealed that Bifidobacterium longum and, particularly, B. longum subsp. longum, appears as the better equipped to metabolize complex AX substrates, although other related subspecies such as B. longum subsp. infantis, also hold some machinery related to AXOS metabolization. This correlates to the growth profiles exhibited by representative strains of both subspecies in AX or AXOS enriched media. Based on these results, we formulated a differential carbohydrate free medium (CFM) supplemented with a combination of AX and AXOS that enabled to recover a wide diversity of Bifidobacterium species from complex fecal samples, while allowing easy discrimination of AX metabolising strains by the appearance of a precipitation halo. This new media represent an appealing alternative to isolate novel probiotic bifidobacteria, rapidly discriminating their capacity to metabolize structurally complex AX-derived prebiotics. This can be convenient to assist formulation of novel functional foods and supplements, including bifidobacterial species with capacity to metabolize AX-derived prebiotic ingredients.


Subject(s)
Bifidobacterium longum , Synbiotics , Humans , Bifidobacterium longum/metabolism , Bifidobacterium/metabolism , Xylans , Oligosaccharides/metabolism , Prebiotics
17.
Acta Crystallogr D Struct Biol ; 79(Pt 4): 290-303, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36974963

ABSTRACT

Phosphoketolase and transketolase are thiamine diphosphate-dependent enzymes and play a central role in the primary metabolism of bifidobacteria: the bifid shunt. The enzymes both catalyze phosphorolytic cleavage of xylulose 5-phosphate or fructose 6-phosphate in the first reaction step, but possess different substrate specificity in the second reaction step, where phosphoketolase and transketolase utilize inorganic phosphate (Pi) and D-ribose 5-phosphate, respectively, as the acceptor substrate. Structures of Bifidobacterium longum phosphoketolase holoenzyme and its complex with a putative inhibitor, phosphoenolpyruvate, were determined at 2.5 Šresolution by serial femtosecond crystallography using an X-ray free-electron laser. In the complex structure, phosphoenolpyruvate was present at the entrance to the active-site pocket and plugged the channel to thiamine diphosphate. The phosphate-group position of phosphoenolpyruvate coincided well with those of xylulose 5-phosphate and fructose 6-phosphate in the structures of their complexes with transketolase. The most striking structural change was observed in a loop consisting of Gln546-Asp547-His548-Asn549 (the QN-loop) at the entrance to the active-site pocket. Contrary to the conformation of the QN-loop that partially covers the entrance to the active-site pocket (`closed form') in the known crystal structures, including the phosphoketolase holoenzyme and its complexes with reaction intermediates, the QN-loop in the current ambient structures showed a more compact conformation with a widened entrance to the active-site pocket (`open form'). In the phosphoketolase reaction, the `open form' QN-loop may play a role in providing the binding site for xylulose 5-phosphate or fructose 6-phosphate in the first step, and the `closed form' QN-loop may help confer specificity for Pi in the second step.


Subject(s)
Bifidobacterium longum , Thiamine Pyrophosphate , Thiamine Pyrophosphate/chemistry , Thiamine Pyrophosphate/metabolism , Bifidobacterium longum/metabolism , Crystallography, X-Ray , Transketolase/chemistry , Transketolase/metabolism , Phosphoenolpyruvate , Temperature , Xylulose , Catalytic Domain , Fructose
18.
J Agric Food Chem ; 71(2): 1100-1112, 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36604158

ABSTRACT

Glucoraphanin, rich in broccoli seed extract (BSE), is generally inert but can be biotransformed into active sulforaphane by gut bacteria. This study aimed to screen probiotics with glucoraphanin-metabolizing ability and explore the effect of a combination of strain and BSE on colitis induced by dextran sulfate sodium (DSS) in mice. Bifidobacterium longum CCFM1206 was isolated from healthy adult feces. Ultra-high-performance liquid chromatography Q Exactive mass spectrometry analysis revealed the presence of sulforaphane, sulforaphane-l-cysteine, and erucin in the BSE supernatant fermented by B. longum CCFM1206 in vitro. Combined and individual interventions of BSE and B. longum CCFM1206 were applied to explore the effects on DSS-induced colitis. The results suggested that the combination of B. longum CCFM1206 and BSE could ameliorate colitis symptoms, relieve colonic inflammatory reactions and oxidative stress, and protect the intestinal barrier in DSS-induced mice. In comparison to the BSE intervention alone, the combined intervention of B. longum CCFM1206 and BSE promoted the generation of sulforaphane and sulforaphane-N-acetylcysteine in mice colon from 220.88 ± 19.81 to 333.99 ± 36.46 nmol/g and from 232.04 ± 26.48 to 297.50 ± 40.08 nmol/g dry weight feces, respectively. According to quantitative reverse transcription polymerase chain reaction and immunohistochemical analysis, B. longum CCFM1206 and BSE effectively activated the transcription and expression of genes related to the Nrf2 signaling pathway. These results were intended to elucidate that probiotics could elevate the bioactivity of dietary phytochemicals in vivo, and the combination had potential for therapeutic treatment of colitis.


Subject(s)
Bifidobacterium longum , Colitis , Mice , Animals , Bifidobacterium longum/metabolism , Dextrans/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Colon/metabolism , Biotransformation , Sulfates/metabolism , Sodium/metabolism , Dextran Sulfate/metabolism , Disease Models, Animal , Mice, Inbred C57BL
19.
Nat Chem Biol ; 19(2): 218-229, 2023 02.
Article in English | MEDLINE | ID: mdl-36443572

ABSTRACT

Bifidobacteria are early colonizers of the human gut and play central roles in human health and metabolism. To thrive in this competitive niche, these bacteria evolved the capacity to use complex carbohydrates, including mammalian N-glycans. Herein, we elucidated pivotal biochemical steps involved in high-mannose N-glycan utilization by Bifidobacterium longum. After N-glycan release by an endo-ß-N-acetylglucosaminidase, the mannosyl arms are trimmed by the cooperative action of three functionally distinct glycoside hydrolase 38 (GH38) α-mannosidases and a specific GH125 α-1,6-mannosidase. High-resolution cryo-electron microscopy structures revealed that bifidobacterial GH38 α-mannosidases form homotetramers, with the N-terminal jelly roll domain contributing to substrate selectivity. Additionally, an α-glucosidase enables the processing of monoglucosylated N-glycans. Notably, the main degradation product, mannose, is isomerized into fructose before phosphorylation, an unconventional metabolic route connecting it to the bifid shunt pathway. These findings shed light on key molecular mechanisms used by bifidobacteria to use high-mannose N-glycans, a perennial carbon and energy source in the intestinal lumen.


Subject(s)
Bifidobacterium longum , Mannose , Animals , Humans , Mannose/metabolism , Bifidobacterium longum/metabolism , Cryoelectron Microscopy , Polysaccharides/chemistry , Mannosidases/metabolism , Glycoside Hydrolases/chemistry , Bifidobacterium/metabolism , Mammals
20.
Metallomics ; 15(1)2023 01 10.
Article in English | MEDLINE | ID: mdl-36583695

ABSTRACT

The influence of the fermentation process on selenite metabolism by a probiotic Bifidobacterium longum DD98 and its consequent enrichment in selenium (Se) were studied. The effects of sodium selenite (Na2SeO3) concentration (18-400 µg/ml), feeding time (12, 16, and 24 h), and fermentation stage (secondary and tertiary fermentation) were evaluated by measuring (i) the total Se content and its distribution between the water-soluble metabolome fraction and the water-insoluble fraction; (ii) the total concentrations of the two principal Se compounds produced: selenomethionine (SeMet) and γ-glutamyl-selenomethionine (γ-Glu-SeMet), and (iii) the speciation of Se in the metabolite fraction. The results revealed that the fermentation process notably changed the Se incorporation into metabolites (γ-Glu-SeMet and free SeMet) and proteins (bound-SeMet) in B. longum DD98. In particular, the production of SeMet was negatively correlated to that of γ-Glu-SeMet when no red precipitate was seen in the bacteria. The study offers a tool for the control of the optimization of the fermentation process towards the desired molecular speciation of the incorporated Se and hence contributes to the production of Se-enriched probiotics with good qualities and bioactivities.


Subject(s)
Bifidobacterium longum , Probiotics , Selenium , Selenium/metabolism , Selenomethionine/metabolism , Selenious Acid , Fermentation , Bifidobacterium longum/metabolism , Sodium Selenite/metabolism , Sodium Selenite/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...