Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Nutrients ; 15(16)2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37630731

ABSTRACT

Bifidobacterium animalis subsp. lactis HN019 is a probiotic with several documented human health benefits. Interest in probiotics has led to the development of new formats that probiotics, including HN019, can be supplemented into. In this study, we looked at common HN019 formats such as frozen culture and freeze-dried powder as well as supplementing it into the following food matrices: yogurts (dairy, soy, and oat based), xanthan gum-based tablets, pulpless orange juice, whey sports drink, and dark chocolate (70% cocoa). In this work, our aim was to investigate whether the food matrix that carried HN019 via simulated human digestion (a dual model system mimicking both upper and lower gastrointestinal digestion) influenced probiotic delivery. To that end, we validated and used a real-time qPCR assay to detect HN019 after simulated digestion. In addition, we also measured the effect on a panel of metabolites. After simulated digestion, we were able to detect HN019 from all the matrices tested, and the observed changes to the metabolite profile were consistent with those expected from the food matrix used. In conclusion, this work suggests that the food matrix supplemented with HN019 did not interfere with delivery to the colon via simulated human digestion.


Subject(s)
Bifidobacterium , Digestion , Humans , Bifidobacterium/genetics , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Lactic Acid/metabolism , Fatty Acids/metabolism , Colon/metabolism , Colon/microbiology
2.
Probiotics Antimicrob Proteins ; 15(3): 491-501, 2023 06.
Article in English | MEDLINE | ID: mdl-34671923

ABSTRACT

Breast milk was long considered a sterile environment, but now it is known to harbor many bacteria that will shape the newborn microbiota. The benefits of breastfeeding to newborn health are, on some level, related to the presence of beneficial bacteria in human milk. Therefore, this study aims to investigate and isolate potential probiotics present in human milk that might be associated with improved health in infants, being potential candidates to be used in simulated human milk formula. Milk samples of 24 healthy mothers were collected at three time points: 30 min (colostrum), 5-9 days (transitional milk), and 25-30 days (mature milk) postpartum. Samples were evaluated by culturing, and the isolated bacteria were identified by MALDI-TOF MS and 16S DNA sequencing. In vitro screening for probiotics properties was performed, and the potential probiotics were mono-associated with germ-free mice to evaluate their ability to colonize the gastrointestinal tract. The microorganisms were submitted to the spray-drying process to check their viability for a potential simulated milk formula production. Seventy-seven bacteria were isolated from breast milk pertaining to four bacterial genera (Staphylococcus, Streptococcus, Leuconostoc, and Lacticaseibacillus). Four potential probiotics were selected: Lacticaseibacillus rhamnosus (n = 2) and Leuconostoc mesenteroides (n = 2). Isolates were able to colonize the gastrointestinal tract of germ-free mice and remained viable after the spray-drying process. In conclusion, breast milk harbors a unique microbiota with beneficial microorganisms that will impact the newborn gut colonization, being an essential source of probiotic candidates to be used in a formula of simulated maternal milk.


Subject(s)
Milk, Human , Probiotics , Infant , Female , Pregnancy , Humans , Animals , Mice , Milk, Human/microbiology , Bifidobacterium/genetics , Bacteria/genetics , Colostrum/microbiology
3.
Microb Cell Fact ; 21(1): 33, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35255900

ABSTRACT

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


Subject(s)
Bifidobacterium adolescentis , Bifidobacterium/genetics , Bifidobacterium/metabolism , Bifidobacterium adolescentis/genetics , Bifidobacterium adolescentis/metabolism , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Humans , gamma-Aminobutyric Acid
4.
Cell Rep Med ; 1(5): 100077, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32904427

ABSTRACT

Supplementation with members of the early-life microbiota as "probiotics" is increasingly used in attempts to beneficially manipulate the preterm infant gut microbiota. We performed a large observational longitudinal study comprising two preterm groups: 101 infants orally supplemented with Bifidobacterium and Lactobacillus (Bif/Lacto) and 133 infants non-supplemented (control) matched by age, sex, and delivery method. 16S rRNA gene profiling on fecal samples (n = 592) showed a predominance of Bifidobacterium and a lower abundance of pathobionts in the Bif/Lacto group. Metabolomic analysis showed higher fecal acetate and lactate and a lower fecal pH in the Bif/Lacto group compared to the control group. Fecal acetate positively correlated with relative abundance of Bifidobacterium, consistent with the ability of the supplemented Bifidobacterium strain to metabolize human milk oligosaccharides into acetate. This study demonstrates that microbiota supplementation is associated with a Bifidobacterium-dominated preterm microbiota and gastrointestinal environment more closely resembling that of full-term infants.


Subject(s)
Bifidobacterium/physiology , Gastrointestinal Microbiome/physiology , Infant, Premature/metabolism , Infant, Premature/physiology , Lactobacillus/physiology , Metabolome/physiology , Bifidobacterium/genetics , Breast Feeding/methods , Dietary Supplements/microbiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Humans , Infant , Infant, Newborn , Lactobacillus/genetics , Longitudinal Studies , Milk, Human/microbiology , Probiotics/administration & dosage , RNA, Ribosomal, 16S/genetics
5.
Biomolecules ; 10(8)2020 08 13.
Article in English | MEDLINE | ID: mdl-32823755

ABSTRACT

The proper development of the early gastrointestinal tract (GIT) microbiota is critical for newborn ruminants. This microbiota is susceptible to modification by diverse external factors (such as diet) that can lead to long-lasting results when occurring in young ruminants. Dietary supplementation with prebiotics, ingredients nondigestible and nonabsorbable by the host that stimulate the growth of beneficial GIT bacteria, has been applied worldwide as a potential approach in order to improve ruminant health and production yields. However, how prebiotics affect the GIT microbiota during ruminants' early life is still poorly understood. We investigated the effect of milk supplementation with a combination of two well-known prebiotics, fructooligosaccharides (FOS) from sugar beet and garlic residues (all together named as "additive"), exerted on preweaned lamb growth and the composition of their fecal microbiota, by using 16S rRNA gene amplicon high-throughput sequencing. The results showed a significant increase in the mean daily weight gain of lambs fed with the additive. Lamb fecal microbiota was also influenced by the additive intake, as additive-diet lambs showed lower bacterial diversity and were significantly more abundant in Bifidobacterium, Enterococcus, Lactobacillus and Veillonella. These bacteria have been previously reported to confer beneficial properties to the ruminant, including promotion of growth and health status, and our results showed that they were strongly linked to the additive intake and the increased weight gain of lambs. This study points out the combination of FOS from sugar beet and garlic residues as a potential prebiotic to be used in young ruminants' nutrition in order to improve production yields.


Subject(s)
Bacteria/classification , Beta vulgaris/chemistry , Body Weight/drug effects , Garlic/chemistry , Oligosaccharides/administration & dosage , Plant Extracts/administration & dosage , Animals , Animals, Newborn , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Bifidobacterium/classification , Bifidobacterium/genetics , Bifidobacterium/growth & development , Bifidobacterium/isolation & purification , Dietary Supplements , Gastrointestinal Microbiome , High-Throughput Nucleotide Sequencing , Lactobacillus/classification , Lactobacillus/genetics , Lactobacillus/growth & development , Lactobacillus/isolation & purification , Milk/chemistry , Oligosaccharides/pharmacology , Plant Extracts/pharmacology , Prebiotics/administration & dosage , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Sheep , Veillonella/classification , Veillonella/genetics , Veillonella/growth & development , Veillonella/isolation & purification
6.
Proc Natl Acad Sci U S A ; 116(51): 25909-25916, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31776248

ABSTRACT

Bees acquire carbohydrates from nectar and lipids; and amino acids from pollen, which also contains polysaccharides including cellulose, hemicellulose, and pectin. These potential energy sources could be degraded and fermented through microbial enzymatic activity, resulting in short chain fatty acids available to hosts. However, the contributions of individual microbiota members to polysaccharide digestion have remained unclear. Through analysis of bacterial isolate genomes and a metagenome of the honey bee gut microbiota, we identify that Bifidobacterium and Gilliamella are the principal degraders of hemicellulose and pectin. Both Bifidobacterium and Gilliamella show extensive strain-level diversity in gene repertoires linked to polysaccharide digestion. Strains from honey bees possess more such genes than strains from bumble bees. In Bifidobacterium, genes encoding carbohydrate-active enzymes are colocated within loci devoted to polysaccharide utilization, as in Bacteroides from the human gut. Carbohydrate-active enzyme-encoding gene expressions are up-regulated in response to particular hemicelluloses both in vitro and in vivo. Metabolomic analyses document that bees experimentally colonized by different strains generate distinctive gut metabolomic profiles, with enrichment for specific monosaccharides, corresponding to predictions from genomic data. The other 3 core gut species clusters (Snodgrassella and 2 Lactobacillus clusters) possess few or no genes for polysaccharide digestion. Together, these findings indicate that strain composition within individual hosts determines the metabolic capabilities and potentially affects host nutrition. Furthermore, the niche specialization revealed by our study may promote overall community stability in the gut microbiomes of bees.


Subject(s)
Bees/microbiology , Bees/physiology , Digestion , Gastrointestinal Microbiome/physiology , Plants/chemistry , Polysaccharides/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Bifidobacterium/genetics , Bifidobacterium/metabolism , Gammaproteobacteria/genetics , Gammaproteobacteria/metabolism , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Gene Expression Regulation , Genome, Bacterial , Lactobacillus/genetics , Metagenome , Microbiota , Neisseriaceae/genetics , Pollen/chemistry
7.
Biomolecules ; 9(9)2019 08 21.
Article in English | MEDLINE | ID: mdl-31438572

ABSTRACT

Hop cones (Humulus lupulus L.) have been used throughout history as an additive in beer brewing and as herbal supplements with medicinal and culinary properties. The objective of this study was to ascertain the effect of a range of concentrations of a supercritical CO2 extract of hops on the composition and metabolism of human gut bacterial communities using in vitro batch culture systems. Fermentations were conducted over 24 h using a mixed human fecal inoculum. Microbial metabolism was assessed by measuring organic acid production and microbial community alterations were determined by 16S rRNA gene sequencing. Butyrate, an important short chain fatty acid in maintaining colonic well-being, decreased at elevated concentrations of hops, which may partly be accounted for by the concomitant reduction of Eubacterium and Coprococcus, known butyrate-producing genera, and also the inhibition of Bifidobacterium, a beneficial organism that has a butyrogenic effect through metabolic cross-feeding with intestinal commensals. The hops compounds also caused dose-dependent increases in the potentially pathogenic Enterobacteriaceae and potentially beneficial Akkermansia. Thus, hops compounds had a significant impact on the structure of the bacterial consortium, which warrants further study including human clinical trials.


Subject(s)
Butyrates/metabolism , Chromatography, Supercritical Fluid , Humulus/chemistry , Microbiota/drug effects , Plant Extracts/chemistry , Bifidobacterium/drug effects , Bifidobacterium/genetics , Bifidobacterium/metabolism , Carbon Dioxide/chemistry , Eubacterium/drug effects , Eubacterium/genetics , Eubacterium/metabolism , Humans , Humulus/metabolism , Plant Extracts/pharmacology , Principal Component Analysis , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/metabolism
8.
Int Immunopharmacol ; 74: 105681, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31220694

ABSTRACT

Salmonella typhimurium (S. t.) is one of the main pathogens that causes acute gastroenteritis. To evaluate the anti-inflammatory mechanism of Astragalus polysaccharide (APS) in vivo and its influence on the intestinal flora, BALB/c mice were infected with S. t. to establish a model of diarrhea. The disease activity index (DAI) scores showed that APS attenuated S. t.-induced weight loss and diarrhea in mice. APS significantly reduced the index of the liver and spleen as well as the ALT and AST levels in serum (P < 0.05). Hematoxylin and eosin (H&E) results indicated that APS significantly increased jejunum villus height and crypt depth and reduced the infiltration of inflammatory cells (P < 0.05). Additionally, APS increased the tight junction (TJ) proteins expression levels of ZO-1, Occludin and Claudin-1 in the jejunum. The results of 16S rDNA showed that APS significantly increased the number of Lactobacillus and Bifidobacterium spp. to normal levels (compared with the control group). In addition, APS significantly decreased the mRNA expression levels of the proinflammatory cytokines TNF-α, IL-1ß, IL-6 and IL-17 in the jejunum (P < 0.01) as well as the proteins expression levels of COX-2 and iNOS (P < 0.05). Western blot confirmed that prefeeding with APS inhibited S. t.-induced expression of TLR4 and MyD88 in the jejunum and further inhibited nuclear factor-κB (NF-κB) activation, including the nuclear translocation of the p65 NF-κB subunit and the phosphorylation and degradation of IκB-α. This was the key to APS inhibition of the production of inflammatory factors and inflammatory mediators in the jejunum.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Bifidobacterium/genetics , Diarrhea/therapy , Gastrointestinal Microbiome/genetics , Lactobacillus/genetics , Liver/metabolism , Polysaccharides/therapeutic use , Salmonella Infections/therapy , Salmonella typhimurium/physiology , Tight Junctions/metabolism , Animals , Astragalus Plant/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Signal Transduction , Tight Junctions/pathology
9.
Anaerobe ; 56: 40-45, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30738138

ABSTRACT

In the present study, we investigated the potential of Bifidobacterium spp., isolated from ruminal fluid samples from buffaloes (Bubalus bubalis) for conjugated linoleic acid (CLA) production. A total of 294 isolates were obtained from 86 ruminal fluid samples using Bifidus Selective Medium (BSM) medium, and based on phospoketolase assay, 24 isolates were presumptively confirmed to be Bifidobacterium species. Further, the isolates were confirmed morphologically, biochemically and by PCR assays for genus specific (16s rDNA) and transaldolase genes. All 24 strains were positive for conversion of linoleic acid (LA) to CLA by spectrophotometric screening. Gas chromatographic analysis showed that the strains produced cis9, trans11 and tran10, cis12 CLA isomers in LA-supplemented deMan-Rogosa-Sharpe (MRS) broth. The strains were identified as B. thermophilum (n = 21) and B. pseudolongum (n = 3) based on 16 rDNA sequence analysis. The study shows that Bifidobacterium spp., present in the rumens of buffaloes produce CLA from LA and the strains may have the potential to be used as probiotics to enhance the nutraceutical value of ruminant food products.


Subject(s)
Bifidobacterium/isolation & purification , Bifidobacterium/metabolism , Buffaloes , Linoleic Acids, Conjugated/metabolism , Rumen/microbiology , Animals , Bacteriological Techniques , Bifidobacterium/classification , Bifidobacterium/genetics , Body Fluids/microbiology , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Phylogeny , Polymerase Chain Reaction , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Transaldolase/genetics
10.
J Nutr Biochem ; 63: 54-61, 2019 01.
Article in English | MEDLINE | ID: mdl-30342317

ABSTRACT

BACKGROUND AND AIMS: Breast milk jaundice (BMJ) is common and benign, but neonatal cholestasis (NC) is rare and not benign, so early differentiation between NC and non-NC jaundice is important and may facilitate diagnosis and treatment. Gut microbiota plays an important role in enterohepatic circulation, which in turn plays an important role in the secretion of bilirubin. We aimed to determine the composition of gut microbiota in patients with NC and BMJ, and to identify the gut microbiota composition associated with NC and BMJ. METHODS: Data on age, gender, delivery, feeding mode, serum total bilirubin, direct bilirubin, and liver function were collected for NC patients, BMJ patients and healthy controls, respectively. Shotgun metagenomic sequencing and metagenome-wide association were performed. RESULTS: Forty NC patients, 16 patients affected by BMJ, and 14 healthy controls (CON) without jaundice were enrolled. A significant increase in species richness, especially Bacteroides, was found in NC patients. The abundances of potentially pathogenic species and KEGG orthologies (KOs) of virulence factor genes were positively correlated with serum bilirubin level. The abundances of nine species of Bifidobacterium and three KOs of galactose metabolism were significantly decreased in the jaundice group (NC and BMJ) and were negatively correlated with serum bilirubin level. CONCLUSIONS: The gut microbiota in NC patients is characterized by a significant increase in species richness, possibly due to the proliferation of potentially pathogenic species. Additionally, the gut microbiota in jaundice patients is characterized by a decreased abundance of Bifidobacterium. Decreased Bifidobacterium has been associated with elevated bilirubin and abnormal gut microbiota galactose metabolic pathway. Further, ten bacteria species were identified as potential biomarker of jaundice. KEY POINTS: Question Is there any alteration of gut microbiotain neonatal cholestasis patients? Does gut microbiota have any involvement in the occurrence of neonatal cholestasis or breast milk jaundice? Findings The alteration of gut microbiota in neonatal cholestasis patients mainly manifested as a significant increase in species richness and an increased abundance of potentially pathogenic species, while the main manifestation in jaundice patients was a significant decrease in Bifidobacterium which may be involved in the metabolism of bilirubin through the galactose metabolic pathway. Meaning The results suggest that an imbalance of gut microbiota exist in neonatal cholestasis and breast milk jaundice patients, primarily in the form of a substantial reduction in the abundance of Bifidobacterium, suggesting the possibility of intervention treatment for neonatal cholestasis and breast milk jaundice by supplementing probiotics.


Subject(s)
Bilirubin/blood , Dysbiosis/blood , Gastrointestinal Microbiome , Jaundice, Neonatal/blood , Bifidobacterium/genetics , Breast Feeding , Case-Control Studies , Child, Preschool , Cholestasis/blood , Cholestasis/microbiology , Dysbiosis/microbiology , Female , Gastrointestinal Microbiome/genetics , Humans , Infant , Infant, Newborn , Jaundice, Neonatal/microbiology , Male
11.
Nutr Hosp ; 35(6): 1394-1400, 2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30525855

ABSTRACT

BACKGROUND: patients with cervical cancer (CC) receiving chemotherapy and radiotherapy have several gastrointestinal adverse effects. OBJECTIVE: to evaluate the effect of dietary symbiotic supplementation on fecal calprotectin (FCP), bacterial DNA levels, and gastrointestinal adverse effects in patients with CC. METHODS: clinical, controlled, randomized, double-blind trial. Patients consumed symbiotics or placebo three times a day for seven weeks. FCP was assessed by Elisa method. DNA from probiotic and pathogenic bacteria were determined by quantitative real-time polymerase chain reaction. Diarrheal evacuations were evaluated with the Bristol stool form scale and nausea and vomiting were measured using the scale of the National Institute of Cancerology of the United States. RESULTS: after a seven-week treatment, FCP concentration was lower in the symbiotic group compared to the control group (p < 0.001). Stool consistency in the placebo and symbiotic groups was similar at baseline. A significant improvement in stool consistency was obtained in both groups at the end of the intervention (p < 0.001). The concentrations and total proportions of the probiotic and pathogenic bacteria were similar in both groups. Nausea significantly diminished in both groups (p < 0.001) at the end of the trial. Furthermore, the symbiotic group had a statistically significant decrease in the frequency and intensity of vomiting when compared to the control group (p < 0.001). CONCLUSIONS: the symbiotic treatment decreases significantly the FCP levels and the frequency and intensity of vomiting in patients with CC.


Subject(s)
Feces/chemistry , Feces/microbiology , Leukocyte L1 Antigen Complex/analysis , Prebiotics/administration & dosage , Probiotics/administration & dosage , Uterine Cervical Neoplasms/therapy , Adult , Antineoplastic Agents/adverse effects , Bifidobacterium/genetics , DNA, Bacterial/analysis , Dietary Supplements , Double-Blind Method , Escherichia coli/genetics , Female , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/prevention & control , Humans , Inflammation/etiology , Inflammation/prevention & control , Lactobacillales/genetics , Middle Aged , Placebos , Radiotherapy/adverse effects , Salmonella/genetics , Uterine Cervical Neoplasms/complications
12.
BMC Microbiol ; 18(1): 184, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30424728

ABSTRACT

BACKGROUND: The ProPrems trial, a multi-center, double-blind, placebo-controlled randomized trial, previously reported a 54% reduction in necrotizing enterocolitis (NEC) of Bell stage 2 or more from 4.4 to 2.0% in 1099 infants born before 32 completed weeks' gestation and weighing < 1500 g, receiving probiotic supplementation (with Bifidobacterium longum subsp. infantis BB-02, Streptococcus thermophilus TH-4 and Bifidobacterium animalis subsp. lactis BB-12). This sub-study investigated the effect of probiotic supplementation on the gut microbiota in a cohort of very preterm infants in ProPrems. RESULTS: Bifidobacterium was found in higher abundance in infants who received the probiotics (AOR 17.22; 95% CI, 3.49-84.99, p < 0.001) as compared to the placebo group, and Enterococcus was reduced in infants receiving the probiotic during the supplementation period (AOR 0.27; 95% CI, 0.09-0.82, p = 0.02). CONCLUSION: Probiotic supplementation with BB-02, TH-4 and BB-12 from soon after birth increased the abundance of Bifidobacterium in the gut microbiota of very preterm infants. Increased abundance of Bifidobacterium soon after birth may be associated with reducing the risk of NEC in very preterm infants.


Subject(s)
Dietary Supplements/analysis , Enterocolitis, Necrotizing/prevention & control , Gastrointestinal Microbiome , Infant, Extremely Premature/growth & development , Probiotics/administration & dosage , Bifidobacterium/genetics , Bifidobacterium/isolation & purification , Bifidobacterium/radiation effects , Cohort Studies , Double-Blind Method , Enterocolitis, Necrotizing/microbiology , Female , Humans , Infant , Infant, Newborn , Male , Streptococcus thermophilus/genetics , Streptococcus thermophilus/isolation & purification , Streptococcus thermophilus/physiology
13.
J Immunol ; 201(11): 3229-3243, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30341184

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) in common marmosets is a translationally relevant model of the chronic neurologic disease multiple sclerosis. Following the introduction of a new dietary supplement in our purpose-bred marmoset colony, the percentage of marmosets in which clinically evident EAE could be induced by sensitization against recombinant human myelin oligodendrocyte glycoprotein in IFA decreased from 100 to 65%. The reduced EAE susceptibility after the dietary change coincided with reduced Callitrichine herpesvirus 3 expression in the colony, an EBV-related γ1-herpesvirus associated with EAE. We then investigated, in a controlled study in marmoset twins, which disease-relevant parameters were affected by the dietary change. The selected twins had been raised on the new diet for at least 12 mo prior to the study. In twin siblings reverted to the original diet 8 wk prior to EAE induction, 100% disease prevalence (eight out of eight) was restored, whereas in siblings remaining on the new diet the EAE prevalence was 75% (six out of eight). Spinal cord demyelination, a classical hallmark of the disease, was significantly lower in new-diet monkeys than in monkeys reverted to the original diet. In new-diet monkeys, the proinflammatory T cell response to recombinant human myelin oligodendrocyte glycoprotein was significantly reduced, and RNA-sequencing revealed reduced apoptosis and enhanced myelination in the brain. Systematic typing of the marmoset gut microbiota using 16S rRNA sequencing demonstrated a unique, Bifidobacteria-dominated composition, which changed after disease induction. In conclusion, targeted dietary intervention exerts positive effects on EAE-related parameters in multiple compartments of the marmoset's gut-immune-CNS axis.


Subject(s)
Bifidobacterium/genetics , Brain/physiology , Cells/immunology , Dietary Supplements , Encephalomyelitis, Autoimmune, Experimental/diet therapy , Multiple Sclerosis/diet therapy , Spinal Cord/pathology , Animals , Apoptosis , Callithrix , Cells, Cultured , Demyelinating Diseases , Diet Therapy , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Herpesvirus 3, Human , Humans , Myelin-Oligodendrocyte Glycoprotein/immunology , RNA, Ribosomal, 16S/genetics , Sequence Analysis, RNA
14.
Sci Rep ; 8(1): 13958, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228375

ABSTRACT

Gut microbiota of breast-fed infants are generally rich in bifidobacteria. Recent studies show that infant gut-associated bifidobacteria can assimilate human milk oligosaccharides (HMOs) specifically among the gut microbes. Nonetheless, little is known about how bifidobacterial-rich communities are shaped in the gut. Interestingly, HMOs assimilation ability is not related to the dominance of each species. Bifidobacterium longum susbp. longum and Bifidobacterium breve are commonly found as the dominant species in infant stools; however, they show limited HMOs assimilation ability in vitro. In contrast, avid in vitro HMOs consumers, Bifidobacterium bifidum and Bifidobacterium longum subsp. infantis, are less abundant in infant stools. In this study, we observed altruistic behaviour by B. bifidum when incubated in HMOs-containing faecal cultures. Four B. bifidum strains, all of which contained complete sets of HMO-degrading genes, commonly left HMOs degradants unconsumed during in vitro growth. These strains stimulated the growth of other Bifidobacterium species when added to faecal cultures supplemented with HMOs, thereby increasing the prevalence of bifidobacteria in faecal communities. Enhanced HMOs consumption by B. bifidum-supplemented cultures was also observed. We also determined the complete genome sequences of B. bifidum strains JCM7004 and TMC3115. Our results suggest B. bifidum-mediated cross-feeding of HMOs degradants within bifidobacterial communities.


Subject(s)
Bacterial Proteins/metabolism , Bifidobacteriales Infections/metabolism , Bifidobacterium/metabolism , Feces/microbiology , Milk, Human/metabolism , Oligosaccharides/metabolism , Adult , Bacterial Proteins/genetics , Bifidobacteriales Infections/microbiology , Bifidobacterium/classification , Bifidobacterium/genetics , Cells, Cultured , Child, Preschool , Dietary Supplements , Female , Gastrointestinal Microbiome , Genome, Bacterial , Humans , Infant , Male
15.
Food Funct ; 9(8): 4480-4492, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-30080211

ABSTRACT

This study assessed the effects of supplementation with honey from Mimosa quadrivalvis L. (malícia) (MH) produced by jandaíra stingless bees (Melipona subnitida D.) on the lipid metabolism, antioxidant status and some intestinal health parameters of rats with diet-induced dyslipidaemia. Rats were randomly divided into four groups: healthy control (HC), dyslipidaemic control (DC), healthy experimental (EHH) and dyslipidaemic experimental (EDH). Malícia honey (MH) was administered (1000 mL kg-1) via orogastric feeding for 35 days. Dyslipidaemic rats supplemented with MH showed lower food consumption, increased glucose tolerance and superoxide dismutase (SOD) activity, and decreased total cholesterol, LDL and AST blood levels. Counts of Bifidobacterium spp. and Lactobacillus spp. and excretion of organic acids in faeces increased in dyslipidaemic rats supplemented with MH. MH supplementation protected the colon and liver from tissue damage induced by the dyslipidaemic diet. These results show the beneficial effects of MH on the lipid metabolism, antioxidant status and intestinal health of rats with diet-induced dyslipidaemia.


Subject(s)
Dyslipidemias/diet therapy , Honey/analysis , Mimosa/chemistry , Animals , Antioxidants/metabolism , Bees , Bifidobacterium/genetics , Bifidobacterium/growth & development , Bifidobacterium/isolation & purification , Cholesterol/metabolism , Dietary Supplements/analysis , Dyslipidemias/metabolism , Dyslipidemias/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Glucose/metabolism , Humans , Lactobacillus/genetics , Lactobacillus/growth & development , Lactobacillus/isolation & purification , Male , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
16.
Drug Discov Ther ; 12(3): 161-169, 2018.
Article in English | MEDLINE | ID: mdl-29998997

ABSTRACT

Alterations in gut microbiota have been known to play a critical role in metabolic syndrome. However, the microbial features in elderly patients with metabolic syndrome remain unclear. A traditional Chinese Herbal Formula, Yangyin Tiluo Decoction (YTD), can alleviate metabolic syndrome and cardiovascular disease. To characterize gut microbiota in elder patients and effects of YTD on gut microbiota during treatment of metabolic syndrome, 11 healthy elderly persons and 12 elderly persons (aged 60-90 years) with metabolic syndrome were enrolled. The patients were randomly assigned to receive YTD for 4 weeks (200 mL of the decoction two times daily). The microbial composition in healthy control, pre- and post- YTD treatment group were analyzed by 16S rRNA sequencing of fecal DNAs. Biochemical measurements were conducted for elderly patients. The results showed a high inter-individual variation of gut microbiota in elderly persons. The gut microbiota was dominated by phylum Firmicutes and Actinobacteria, which was distinct from the previously defined microbiota in Irish elderly persons. The elderly patients with metabolic syndrome had higher proportions of Lactobacillus and Bifidobacterium, and lower proportions of Anaerostipes, Coprococcus, Ruminococcus than healthy controls. YTD treatment reduced the abundance of genus Bacteroidales Incertae Sedis and species Enterobacteriaceae Incertae Sedis. The concentration of plasma lipoprotein (a) was also reduced, which was negatively correlated with the abundance of an Acinetobacter species. These results reveal a remarkable dominance of Firmicutes and Actinobacteria, and highlight the distinct gut microbiota in elderly patients with metabolic syndrome, which may be involved in pathogenesis. Furthermore, the benefits of YTD treatment were observed, providing an approach to improve metabolic syndrome in elderly patients.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Metabolic Syndrome/microbiology , Actinobacteria/genetics , Aged , Aged, 80 and over , Animals , Bifidobacterium/genetics , Case-Control Studies , Drugs, Chinese Herbal/therapeutic use , Female , Firmicutes/genetics , Gastrointestinal Microbiome/genetics , Humans , Lactobacillus/genetics , Ligusticum , Lycium , Male , Metabolic Syndrome/drug therapy , Middle Aged , Oligochaeta , Panax notoginseng , Polygonatum , RNA, Ribosomal, 16S/genetics , Rehmannia , Rhodiola , Ruminococcus/genetics
17.
Mol Nutr Food Res ; 62(12): e1800178, 2018 06.
Article in English | MEDLINE | ID: mdl-29750437

ABSTRACT

SCOPE: GTPs (green tea polyphenols) exert anti-CRC (colorectal cancer) activity. The intestinal microbiota and intestinal colonization by bacteria of oral origin has been implicated in colorectal carcinogenesis. GT modulates the composition of mouse gut microbiota harmonious with anticancer activity. Therefore, the effect of green tea liquid (GTL) consumption on the gut and oral microbiome is investigated in healthy volunteers (n = 12). METHODS AND RESULTS: 16S sequencing and phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) analysis of both fecal and saliva samples (collected before intervention, after 2 weeks of GTL (400 mL per day) and after a washout period of one week) in healthy volunteers show changes in microbial diversity and core microbiota and difference in clear classification (partial least squares-discriminant analysis [PLS-DA]). An irreversible, increased FIR:BAC (Firmicutes to Bacteroidetes ratio), elevated SCFA producing genera, and reduction of bacterial LPS synthesis in feces are discovered in response to GTL. GTL alters the salivary microbiota and reduces the functional pathways abundance relevance to carcinogenesis. Similar bacterial networks in fecal and salivary microbiota datasets comprising putative oral bacteria are found and GTL reduces the fecal levels of Fusobacterium. Interestingly, both Lachnospiraceae and B/E (Bifidobacterium to Enterobacteriacea ratio-markers of colonization resistance [CR]) are negatively associated with the presence of oral-like bacterial networks in the feces. CONCLUSION: These results suggest that GTL consumption causes both oral and gut microbiome alterations.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Saliva/microbiology , Tea , Adult , Bifidobacterium/genetics , Bifidobacterium/isolation & purification , Feces/microbiology , Female , Fusobacterium/genetics , Fusobacterium/isolation & purification , Healthy Volunteers , Humans , Male , Middle Aged , RNA, Ribosomal, 16S
18.
Biomed Res Int ; 2018: 1879168, 2018.
Article in English | MEDLINE | ID: mdl-29682522

ABSTRACT

Antibiotic growth promoters have been used for decades in poultry farming as a tool to maintain bird health and improve growth performance. Global concern about the recurrent emergence and spreading of antimicrobial resistance is challenging the livestock producers to search for alternatives to feed added antibiotics. The use of phytogenic compounds appears as a feasible option due to their ability to emulate the bioactive properties of antibiotics. However, detailed description about the effects of in-feed antibiotics and alternative natural products on chicken intestinal microbiota is lacking. High-throughput sequencing of 16S rRNA gene was used to study composition of cecal microbiota in broiler chickens supplemented with either bacitracin or a blend of chestnut and quebracho tannins over a 30-day grow-out period. Both tannins and bacitracin had a significant impact on diversity of cecal microbiota. Bacitracin consistently decreased Bifidobacterium while other bacterial groups were affected only at certain times. Tannins-fed chickens showed a drastic decrease in genus Bacteroides while certain members of order Clostridiales mainly belonging to the families Ruminococcaceae and Lachnospiraceae were increased. Different members of these groups have been associated with an improvement of intestinal health and feed efficiency in poultry, suggesting that these bacteria could be associated with productive performance of birds.


Subject(s)
Bacitracin/pharmacology , Chickens/microbiology , Gastrointestinal Microbiome/drug effects , Microbiota/drug effects , Tannins/pharmacology , Animal Feed , Animals , Anti-Bacterial Agents/pharmacology , Bacteroides/drug effects , Bacteroides/genetics , Bifidobacterium/drug effects , Bifidobacterium/genetics , Clostridiales/drug effects , Clostridiales/genetics , Intestines/microbiology , Microbiota/genetics , RNA, Ribosomal, 16S/genetics
19.
Benef Microbes ; 9(2): 269-278, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29380644

ABSTRACT

Honey bees are important pollinators of several crops and ecosystems, having a great ecological and economic value. In Europe, the restricted use of chemicals and therapeutic agents in the beekeeping sector has stimulated the search for natural alternatives with a special focus on gut symbionts. The modulation of the gut microbiota has been recognised as a practical and successful approach in the entomological field for the management of insect-related problems. To date, only a few studies have investigated the effect of bacterial supplementation on the health status of colonies, colony productivity and gut symbionts. To this purpose, a preparation of sugar syrup containing bifidobacteria and lactobacilli isolated from bee gut was sprayed on the frames of an apiary located in open field once a week for four weeks. Treated and control hives were monitored for two months for brood extension, honey and pollen harvest. The presence of beneficial gut microorganisms within bee gut was investigated with denaturing gradient gel electrophoresis and next generation sequencing. The administered bacteria led to a significant increase of brood population (46.2%), pollen (53.4%) and harvestable honey in honey supers (59.21%). Analysis of the gut microbiota on the new generation of bees in treated hives showed an increase in relative abundance of Acetobacteraceae and Bifidobacterium spp., which are known to be involved in bee nutrition and protection.


Subject(s)
Bees/microbiology , Bifidobacterium/physiology , Dietary Supplements , Lactobacillus/physiology , Probiotics , Animal Feed , Animals , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Bifidobacterium/genetics , DNA, Bacterial/genetics , Gastrointestinal Microbiome/physiology , Genome, Bacterial/genetics , Lactobacillus/genetics , RNA, Ribosomal, 16S/genetics
20.
Biomed Res Int ; 2017: 7585989, 2017.
Article in English | MEDLINE | ID: mdl-29214176

ABSTRACT

OBJECTIVE: To investigate the gut microbiota differences of obese children compared with the control healthy cohort to result in further understanding of the mechanism of obesity development. METHODS: We evaluated the 16S rRNA gene, the enterotypes, and quantity of the gut microbiota among obese children and the control cohort and learned the differences of the gut microbiota during the process of weight reduction in obese children. RESULTS: In the present study, we learned that the gut microbiota composition was significantly different between obese children and the healthy cohort. Next we found that functional changes, including the phosphotransferase system, ATP-binding cassette transporters, flagellar assembly, and bacterial chemotaxis were overrepresented, while glycan biosynthesis and metabolism were underrepresented in case samples. Moreover, we learned that the amount of Bifidobacterium and Lactobacillus increased among the obese children during the process of weight reduction. CONCLUSION: Our results might enrich the research between gut microbiota and obesity and further provide a clinical basis for therapy for obesity. We recommend that Bifidobacterium and Lactobacillus might be used as indicators of healthy conditions among obese children, as well as a kind of prebiotic and probiotic supplement in the diet to be an auxiliary treatment for obesity.


Subject(s)
Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Microbiota/genetics , Obesity/microbiology , Adolescent , Asian People , Bifidobacterium/genetics , Bifidobacterium/physiology , Child , Child, Preschool , Cohort Studies , Diet , Dietary Supplements , Humans , Lactobacillus/genetics , Lactobacillus/physiology , Prebiotics/administration & dosage , Probiotics/administration & dosage , RNA, Ribosomal, 16S/genetics , Weight Loss/physiology
SELECTION OF CITATIONS
SEARCH DETAIL