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










Publication year range
1.
Microorganisms ; 11(7)2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37512945

ABSTRACT

The relationship between the microbiota profile and exposure to stress is not well understood. Therefore, we used a rat model of unpredictable chronic mild stress (UCMS) to investigate this relationship. Depressive-like behaviors were measured in Female Sprague Dawley rats using the sucrose preference test and the Porsolt swim test. Anxiety-like behaviors were measured with the light-dark box test. Fecal corticosterone, cecal microbiota (composition and organic acids), plasma gut permeability (lipopolysaccharide-binding protein, LBP) and plasma inflammation (12 cytokines) markers were measured. Atypical behaviors were observed in female rats following UCMS, but no depressive-like behaviors were observed. Circulating concentrations of cytokines granulocyte-macrophage colony-stimulating factor and cytokine-induced neutrophil chemoattractant 1 were higher in UCMS-exposed female rats; plasma LBP and cecal organic acid levels remained unchanged. Our results reflect a resilient and adaptive phenotype for female SD rats. The relative abundance of taxa from the Clostridiales order and Desulfovibrionaceae family did, however, correlate both positively and negatively with anxiety-like behaviors and plasma cytokine concentrations, regardless of UCMS exposure, supporting the brain-to-gut influence of mild anxiety with a microbiota profile that may involve inflammatory pathways.

2.
Pediatr Res ; 93(7): 2005-2013, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36319696

ABSTRACT

BACKGROUND: Oral microbial therapy has been studied as an intervention for a range of gastrointestinal disorders. Though research suggests that microbial exposure may affect the gastrointestinal system, motility, and host immunity in a pediatric population, data have been inconsistent, with most prior studies being in neither a randomized nor placebo-controlled setting. The aim of this randomized, placebo-controlled study was to evaluate the efficacy of a synbiotic on increasing weekly bowel movements (WBMs) in constipated children. METHODS: Sixty-four children (3-17 years of age) were randomized to receive a synbiotic (n = 33) comprising mixed-chain length oligosaccharides and nine microbial strains, or placebo (n = 31) for 84 days. Stool microbiota was analyzed on samples collected at baseline and completion. The primary outcome was a change from baseline of WBMs in the treatment group compared to placebo. RESULTS: Treatment increased (p < 0.05) the number of WBMs in children with low baseline WBMs, despite broadly distinctive baseline microbiome signatures. Sequencing revealed that low baseline microbial richness in the treatment group significantly anticipated improvements in constipation (p = 0.00074). CONCLUSIONS: These findings suggest the potential for (i) multi-species-synbiotic interventions to improve digestive health in a pediatric population and (ii) bioinformatics-based methods to predict response to microbial interventions in children. IMPACT: Synbiotic microbial treatment improved the number of spontaneous weekly bowel movements in children compared to placebo. Intervention induced an increased abundance of bifidobacteria in children, compared to placebo. All administered probiotic species were enriched in the gut microbiome of the intervention group compared to placebo. Baseline microbial richness demonstrated potential as a predictive biomarker for response to intervention.


Subject(s)
Probiotics , Synbiotics , Child , Humans , Infant , Gastrointestinal Tract/microbiology , Probiotics/therapeutic use , Constipation/therapy , Feces/microbiology , Double-Blind Method
3.
Food Funct ; 13(18): 9355-9371, 2022 Sep 22.
Article in English | MEDLINE | ID: mdl-35972507

ABSTRACT

It is important to understand variability in consumer chewing behavior for designing food products that deliver desired functionalities for target consumer segments. In this study, we selected 29 participants, representing the large range of chewing variation we had observed in 142 healthy young adults, and investigated the influence of chewing behavior on gastrointestinal digestion and colonic fermentation, using in vitro models and brown rice as a model food. Chewing behavior measured by video observations and chewing outcome differed widely between participants, resulting in large differences in the digestibility of carbohydrates. Inter-individual differences in chewing behavior and chewing outcome also significantly affected in vitro patterns of microbial composition and the production of organic acid metabolites, resulting from colonic fermentation, which is increasingly recognized to be important for human health. These digestion/fermentation outcomes were largely related with the chewing time per mouthful, proportion of bolus particles bigger than 2 mm and amount of saliva added to the bolus during chewing. No significant relationships were found with other chewing trajectory and oral physiological measures. These results suggest that modification of chewing may be an effective strategy to control blood glucose levels and to shape gut microbiota and their metabolites, without altering diets, and that further in vivo studies are warranted to confirm these in vitro findings.


Subject(s)
Digestion , Mastication , Blood Glucose , Fermentation , Food , Humans , Mastication/physiology , Young Adult
4.
Microorganisms ; 10(2)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35208928

ABSTRACT

In a very fascinating read, John Goodsir, a Scottish surgeon, describes how he isolated "vegetable organisms" from the "ejected fluid" from the stomach of his 19-year-old patient [...].

5.
Appl Environ Microbiol ; 88(1): e0148721, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34669436

ABSTRACT

Akkermansia muciniphila is a mucin-degrading bacterium found in the human gut and is often associated with positive human health. However, despite being detected by as early as 1 month of age, little is known about the role of Akkermansia in the infant gut. Human milk oligosaccharides (HMOs) are abundant components of human milk and are structurally similar to the oligosaccharides that comprise mucin, the preferred growth substrate of human-associated Akkermansia. A limited subset of intestinal bacteria has been shown to grow well on HMOs and mucin. We therefore examined the ability of genomically diverse strains of Akkermansia to grow on HMOs. First, we screened 85 genomes representing the four known Akkermansia phylogroups to examine their metabolic potential to degrade HMOs. Furthermore, we examined the ability of representative isolates to grow on individual HMOs in a mucin background and analyzed the resulting metabolites. All Akkermansia genomes were equipped with an array of glycoside hydrolases associated with HMO deconstruction. Representative strains were all able to grow on HMOs with various efficiencies and growth yields. Strain CSUN-19, belonging to the AmIV phylogroup, grew to the highest level in the presence of fucosylated and sialylated HMOs. This activity may be partially related to the increased copy numbers and/or the enzyme activities of the α-fucosidases, α-sialidases, and ß-galactosidases. This study examines the utilization of individual purified HMOs by Akkermansia strains representing all known phylogroups. Further studies are required to examine how HMO ingestion influences gut microbial ecology in infants harboring different Akkermansia phylogroups. IMPORTANCE Human milk oligosaccharides (HMOs) are the third most abundant component of breast milk and provide several benefits to developing infants, including the recruitment of beneficial bacteria to the human gut. Akkermansia strains are largely considered beneficial bacteria and have been detected in colostrum, breast milk, and young infants. A. muciniphila MucT, belonging to the AmI phylogroup, contributes to the HMO deconstruction capacity of the infant. Here, using phylogenomics, we examined the genomic capacities of four Akkermansia phylogroups to deconstruct HMOs. Indeed, each phylogroup contained differences in their genomic capacities to deconstruct HMOs, and representative strains of each phylogroup were able to grow using HMOs. These Akkermansia-HMO interactions potentially influence gut microbial ecology in early life, a critical time for the development of the gut microbiome and infant health.


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Akkermansia , Female , Humans , Infant , Oligosaccharides , Verrucomicrobia
6.
Microorganisms ; 9(10)2021 Oct 03.
Article in English | MEDLINE | ID: mdl-34683410

ABSTRACT

We examined the prebiotic potential of 32 food ingredients on the developing infant microbiome using an in vitro gastroileal digestion and colonic fermentation model. There were significant changes in the concentrations of short-chain fatty-acid metabolites, confirming the potential of the tested ingredients to stimulate bacterial metabolism. The 16S rRNA gene sequencing for a subset of the ingredients revealed significant increases in the relative abundances of the lactate- and acetate-producing Bifidobacteriaceae, Enterococcaceae, and Lactobacillaceae, and lactate- and acetate-utilizing Prevotellaceae, Lachnospiraceae, and Veillonellaceae. Selective changes in specific bacterial groups were observed. Infant whole-milk powder and an oat flour enhanced Bifidobacteriaceae and lactic acid bacteria. A New Zealand-origin spinach powder enhanced Prevotellaceae and Lachnospiraceae, while fruit and vegetable powders increased a mixed consortium of beneficial gut microbiota. All food ingredients demonstrated a consistent decrease in Clostridium perfringens, with this organism being increased in the carbohydrate-free water control. While further studies are required, this study demonstrates that the selected food ingredients can modulate the infant gut microbiome composition and metabolism in vitro. This approach provides an opportunity to design nutrient-rich complementary foods that fulfil infants' growth needs and support the maturation of the infant gut microbiome.

7.
Sci Rep ; 11(1): 9292, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33927231

ABSTRACT

Eight plant-based foods: oat flour and pureed apple, blackcurrant, carrot, gold- and green-fleshed kiwifruit, pumpkin, sweetcorn, were pre-digested and fermented with pooled inocula of weaning infants' faecal bacteria in an in vitro hindgut model. Inulin and water were included as controls. The pre-digested foods were analysed for digestion-resistant fibre-derived sugar composition and standardised to the same total fibre concentration prior to fermentation. The food-microbiome interactions were then characterised by measuring microbial acid and gas metabolites, microbial glycosidase activity and determining microbiome structure. At the physiologically relevant time of 10 h of fermentation, the xyloglucan-rich apple and blackcurrant favoured a propiogenic metabolic and microbiome profile with no measurable gas production. Glucose-rich, xyloglucan-poor pumpkin caused the greatest increases in lactate and acetate (indicative of high fermentability) commensurate with increased bifidobacteria. Glucose-rich, xyloglucan-poor oats and sweetcorn, and arabinogalactan-rich carrot also increased lactate and acetate, and were more stimulatory of clostridial families, which are indicative of increased microbial diversity and gut and immune health. Inulin favoured a probiotic-driven consortium, while water supported a proteolytic microbiome. This study shows that the fibre-derived sugar composition of complementary foods may shape infant gut microbiome structure and metabolic activity, at least in vitro.


Subject(s)
Bacteria/metabolism , Dietary Fiber/analysis , Fermentation , Gastrointestinal Microbiome , Sugars/analysis , Avena/chemistry , Bacteria/classification , Bacteria/enzymology , Carboxylic Acids/metabolism , Dietary Fiber/metabolism , Feces/microbiology , Fruit/chemistry , Glycoside Hydrolases/metabolism , Humans , Infant , Sugars/metabolism , Vegetables/chemistry , Weaning
8.
Microorganisms ; 8(10)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066564

ABSTRACT

Whole kiwifruit ('Hayward' and 'Zesy002') were examined for their bioaminergic potential after being subjected to in vitro gastrointestinal digestion and colonic fermentation. Controls included the prebiotic inulin and water, a carbohydrate-free vehicle. The dopamine precursor l-dihydroxyphenylalanine (L-DOPA) and the serotonin precursor 5-hydroxytryptophan were increased in the kiwifruit gastrointestinal digesta ('Hayward' > 'Zesy002') in comparison to the water digesta. Fermentation of the digesta with human fecal bacteria for 18 h modulated the concentrations of bioamine metabolites. The most notable were the significant increases in L-DOPA ('Zesy002' > 'Hayward') and γ-aminobutyric acid (GABA) ('Hayward' > 'Zesy002'). Kiwifruit increased Bifidobacterium spp. and Veillonellaceae (correlating with L-DOPA increase), and Lachnospira spp. (correlating with GABA). The digesta and fermenta were incubated with Caco-2 cells for 3 h followed by gene expression analysis. Effects were seen on genes related to serotonin synthesis/re-uptake/conversion to melatonin, gut tight junction, inflammation and circadian rhythm with different digesta and fermenta from the four treatments. These indicate potential effects of the substrates and the microbially generated organic acid and bioamine metabolites on intestinal functions that have physiological relevance. Further studies are required to confirm the potential bioaminergic effects of gut microbiota-kiwifruit interactions.

9.
Sci Rep ; 10(1): 13055, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32747727

ABSTRACT

Kiwifruit (KF) contains bioactive compounds with potential anti-inflammatory properties. In this study, we investigated the protective effects of KF on gastric and duodenal damage induced by soluble aspirin in healthy rats. Sixty-four male Sprague Dawley rats were allocated to eight experimental treatments (n = 8) and the experimental diets were fed for 14 days ad libitum. The experimental diets were 20% fresh pureed KF (green-fleshed and gold-fleshed) or 10% glucose solution (control diet). A positive anti-inflammatory control treatment (ranitidine) was included. At the end of the 14-day feeding period, the rats were fasted overnight, and the following morning soluble aspirin (400 mg/kg aspirin) or water (control) was administered by oral gavage. Four hours after aspirin administration, the rats were euthanized and samples taken for analysis. We observed no significant ulcer formation or increase in infiltration of the gastric mucosal inflammatory cells in the rats with the aspirin treatment. Despite this, there were significant changes in gene expression, such as in the duodenum of aspirin-treated rats fed green KF where there was increased expression of inflammation-related genes NOS2 and TNF-alpha. We also observed that gold and green KF diets had a number of contrasting effects on genes related to inflammation and gastro-protective effects.


Subject(s)
Actinidia/chemistry , Aspirin/adverse effects , Duodenum/pathology , Fruit/chemistry , Gastric Mucosa/pathology , Gene Expression Regulation , Inflammation/genetics , Stomach/pathology , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Duodenum/drug effects , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gene Expression Regulation/drug effects , Inflammation/pathology , Male , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Principal Component Analysis , Rats, Sprague-Dawley , Stomach/drug effects , Stomach Ulcer/drug therapy , Stomach Ulcer/genetics , Stomach Ulcer/pathology , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tryptophan/metabolism
10.
PLoS One ; 14(12): e0225845, 2019.
Article in English | MEDLINE | ID: mdl-31800608

ABSTRACT

Bacteria within the digestive tract of adult honey bees are likely to play a key role in the digestion of sugar-rich foods. However, the influence of diet on honey bee gut bacteria is not well understood. During periods of low floral abundance, beekeepers often supplement the natural sources of carbohydrate that honey bees collect, such as nectar, with various forms of carbohydrates such as sucrose (a disaccharide) and invert sugar (a mixture of the monosaccharides glucose and fructose). We compared the effect of these sugar supplements on the relative abundance of bacteria in the gut of bees by feeding bees from a single colony, two natural diets: manuka honey, a monofloral honey with known antibacterial properties, and a hive diet; and artificial diets of invert sugar, sucrose solution, and sucrose solutions containing synthesised compounds associated with the antibacterial properties of manuka honey. 16S ribosomal RNA (rRNA)-based sequencing showed that dietary regimes containing manuka honey, sucrose and invert sugar did not alter the relative abundance of dominant core bacteria after 6 days of being fed these diets. However, sucrose-rich diets increased the relative abundances of three sub-dominant core bacteria, Rhizobiaceae, Acetobacteraceae, and Lactobacillus kunkeei, and decreased the relative abundance of Frischella perrara, all which significantly altered the bacterial composition. Acetogenic bacteria from the Rhizobiaceae and Acetobacteraceae families increased two- to five-fold when bees were fed sucrose. These results suggest that sucrose fuels the proliferation of specific low abundance primary sucrose-feeders, which metabolise sugars into monosaccharides, and then to acetate.


Subject(s)
Bacteria/classification , Bees , Carbohydrates/analysis , Gastrointestinal Tract/microbiology , Honey/analysis , Sucrose/analysis , Animals , Bacteria/genetics , Microbiota , New Zealand , Phylogeny , RNA, Ribosomal, 16S/genetics
11.
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
12.
Microorganisms ; 7(2)2019 Jan 31.
Article in English | MEDLINE | ID: mdl-30709031

ABSTRACT

This article reviews the current evidence associating gut microbiota with factors that impact host circadian-metabolic axis, such as light/dark cycles, sleep/wake cycles, diet, and eating patterns. We examine how gut bacteria possess their own daily rhythmicity in terms of composition, their localization to intestinal niches, and functions. We review evidence that gut bacteria modulate host rhythms via microbial metabolites such as butyrate, polyphenolic derivatives, vitamins, and amines. Lifestyle stressors such as altered sleep and eating patterns that may disturb the host circadian system also influence the gut microbiome. The consequent disruptions to microbiota-mediated functions such as decreased conjugation of bile acids or increased production of hydrogen sulfide and the resultant decreased production of butyrate, in turn affect substrate oxidation and energy regulation in the host. Thus, disturbances in microbiome rhythms may at least partially contribute to an increased risk of obesity and metabolic syndrome associated with insufficient sleep and circadian misalignment. Good sleep and a healthy diet appear to be essential for maintaining gut microbial balance. Manipulating daily rhythms of gut microbial abundance and activity may therefore hold promise for a chrononutrition-based approach to consolidate host circadian rhythms and metabolic homeorhesis.

13.
Anaerobe ; 23: 12-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23916722

ABSTRACT

We investigated the biotransformation of four common dietary polyphenols, rutin, quercetin, chlorogenic acid and caffeic acid, in an in vitro mixed culture model of human intestinal microbiota, to determine effects on human gut bacteria. All four compounds were biotransformed rapidly, disappearing from the medium within 0.5 h and later replaced by known phenolic acid breakdown products, at concentrations up to hundreds of micromolar, much higher than in no-polyphenol control experiments. Quantitative PCR was used to measure effects of the polyphenols on the balance between the major groups of intestinal bacteria that are known to influence gut health, i.e., Bifidobacterium spp., Bacteroidetes, and Firmicutes. Fermentation of polyphenols stimulated proliferation of bifidobacteria and decreased the ratio of Firmicutes to Bacteroidetes, relative to controls. Polyphenols also stimulated short chain fatty acid production by the bacteria. Pure bifidobacterial cultures were treated separately with either fermented media isolated from the incubations, the pure test polyphenols, or the biotransformation products detected in the fermentations. Growth stimulation was observed only with fermented polyphenol media and the pure biotransformation products. It appears that dietary polyphenols may have the ability to modify the gut microbial balance, but this effect is indirect, i.e., it is mediated by biotransformation products, rather than the original plant compounds.


Subject(s)
Bacteria/drug effects , Biota , Gastrointestinal Tract/microbiology , Polyphenols/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Biotransformation , Culture Media/chemistry , Feces/microbiology , Healthy Volunteers , Humans , Male , Models, Theoretical , Real-Time Polymerase Chain Reaction
14.
Adv Food Nutr Res ; 68: 205-17, 2013.
Article in English | MEDLINE | ID: mdl-23394989

ABSTRACT

It is becoming clear that the ecology and functionality of the human gut microbiota are extremely diverse and complex. The microbiota have coevolved with us metabolically to live symbiotically and to share the workload of extracting nutrients and energy from the diet. It is also clear that a diet rich in fruit, vegetables, and whole grain cereals is good for general health and gut health and that this is due partly to the phytochemicals and partly to the nondigestible carbohydrates (or dietary fiber) that are present in plants. Kiwifruit contain polyphenolics and nondigestible carbohydrates in the form of pectic, hemicellulosic, and cellulosic polysaccharides, all of which can be degraded by various members of the gut microbiota and result in beneficial effects. This chapter summarizes how kiwifruit act to modify the colonic microbiota and the resultant beneficial effects on human health.


Subject(s)
Actinidia/chemistry , Colon/microbiology , Carbohydrates/pharmacology , Fermentation , Humans , Polyphenols/pharmacology
15.
J Proteome Res ; 11(12): 5863-75, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23072625

ABSTRACT

It is well accepted that our gut bacteria have coevolved with us in relation to our genetics, diet and lifestyle and are integrated metabolically with us to affect our gut health adversely or beneficially. "Who is there" may vary quite widely between individuals, as might "how they do it", but "what they make" may be less variable. Many different individual species of bacteria can perform the same saccharolytic functions and so the availability of substrate (host or diet-derived) along with the degradative enzymes they possess may be key drivers of gut ecology. In this case study, we discuss detailed microbial ecology and metabolism analysis for three individuals following 48 h of in vitro faecal fermentation, using green kiwifruit as the substrate. In parallel, we have analyzed the chemical changes to the kiwifruit carbohydrates present in the fermenta to close the circle on substrate usage/degradative enzymes possessed/microbes present/microbial byproducts produced. In the absence of host carbohydrate, we see that kiwifruit carbohydrates were differentially utilized to drive microbial diversity, yet resulted in similar byproduct production. The starting ecology of each individual influenced the quantitative and qualitative microbial changes; but not necessarily the metabolic byproduct production. Thus, we propose that it is the consistent functional changes that are relevant for assessment of gut health benefits of any food. We recommend that in this era of large scale genotype/-omics studies that hypothesis-driven, bottom-up research is best placed to interpret metagenomic data in parallel with functional, phenotypic data.


Subject(s)
Actinidia/metabolism , Carbohydrate Metabolism , Feces/microbiology , Fruit/metabolism , Metagenome , Bacteria/genetics , Bacteria/growth & development , Bacteria/metabolism , Bacterial Proteins/metabolism , Biota , Cellulose/metabolism , Culture Media/metabolism , Dietary Carbohydrates/metabolism , Enzyme Activation , Enzyme Assays , Fermentation , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Genes, rRNA , Humans , Polysaccharides/metabolism , RNA, Ribosomal, 16S/metabolism , Solubility
16.
Plant Foods Hum Nutr ; 67(3): 208-14, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22872469

ABSTRACT

The intestinal mucosa is constantly exposed to a variety of microbial species including commensals and pathogens, the latter leaving the host susceptible to infection. Antimicrobial peptides (AMP) are an important part of the first line of defense at mucosal surfaces. Human ß-defensins (HBD) are AMP expressed by colonic epithelial cells, which act as broad spectrum antimicrobials. This study explored the direct and indirect effects of green kiwifruit (KF) on human ß-defensin 1 and 2 (HBD-1 and 2) production by epithelial cells. In vitro digestion of KF pulp consisted of a simulated gastric and duodenal digestion, followed by colonic microbial fermentation using nine human faecal donors. Fermenta from individual donors was sterile filtered and independently added to epithelial cells prior to analysis of HBD protein production. KF products obtained from the gastric and duodenal digestion had no effect on the production of HBD-1 or 2 by epithelial cells, demonstrating that KF does not contain substances that directly modulate defensin production. However, when the digested KF products were further subjected to in vitro colonic fermentation, the fermentation products significantly up-regulated HBD-1 and 2 production by the same epithelial cells. We propose that this effect was predominantly mediated by the presence of short-chain fatty acids (SCFA) in the fermenta. Exposure of cells to purified SCFA confirmed this and HBD-1 and 2 production was up-regulated with acetate, propionate and butyrate. In conclusion, in vitro colonic fermentation of green kiwifruit digest appears to prime defense mechanisms in gut cells by enhancing the production of antimicrobial defensins.


Subject(s)
Actinidia , Anti-Infective Agents/metabolism , Colon/drug effects , Fruit , Intestinal Mucosa/drug effects , Plant Preparations/pharmacology , beta-Defensins/biosynthesis , Adult , Colon/metabolism , Colon/microbiology , Duodenum/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fatty Acids, Volatile/metabolism , Feces/microbiology , Female , Fermentation , Gastric Mucosa/metabolism , HT29 Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Middle Aged , Plant Preparations/metabolism , Up-Regulation
17.
Plant Foods Hum Nutr ; 67(3): 200-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22576129

ABSTRACT

We examined the effects of whole kiwifruit on gut microbiota using an in vitro batch model of gastric-ileal digestion and colonic fermentation. Faecal fermentations of gold and green kiwifruit, inulin and water (control) digests were performed for up to 48 h. As compared to the control, gold and green kiwifruit increased Bifidobacterium spp. by 0.9 and 0.8 log(10) cfu/ml, respectively (P < 0.001), and the Bacteroides-Prevotella-Porphyromonas group by 0.4 and 0.5 log(10) cfu/ml, respectively. Inulin only had a bifidogenic effect (+0.4 log(10) cfu/ml). This was accompanied with increases in microbial glycosidases, especially those with substrate specificities relating to the breakdown of kiwifruit oligosaccharides, and with increased generation of short chain fatty acids. The microbial metabolic activity was sustained for up to 48 h, which we attribute to the complexity of the carbohydrate substrate provided by whole kiwifruit. Kiwifruit fermenta supernatant was also separately shown to affect the in vitro proliferation of Bifidobacterium longum, and its adhesion to Caco-2 intestinal epithelial cells. Collectively, these data suggest that whole kiwifruit may modulate human gut microbial composition and metabolism to produce metabolites conducive to increased bifidobacteria-host association.


Subject(s)
Actinidia/chemistry , Bacteria/drug effects , Bifidobacterium/drug effects , Colon/drug effects , Fruit/chemistry , Oligosaccharides/pharmacology , Prebiotics , Adult , Bacteria/growth & development , Bacteria/metabolism , Bacterial Adhesion/drug effects , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Caco-2 Cells , Colon/metabolism , Colon/microbiology , Fatty Acids, Volatile/metabolism , Feces/microbiology , Female , Fermentation , Gastric Mucosa/metabolism , Glycoside Hydrolases/metabolism , Humans , Ileum/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Inulin/pharmacology , Male , Metagenome/drug effects , Middle Aged , Oligosaccharides/metabolism , Substrate Specificity
18.
Int J Food Microbiol ; 124(3): 295-8, 2008 Jun 10.
Article in English | MEDLINE | ID: mdl-18456359

ABSTRACT

The effect of common dietary polyphenols on growth of human gut bacteria and their adhesion to enterocytes was investigated. The influence on the growth of a probiotic (Lactobacillus rhamnosus), a commensal (Escherichia coli) and two pathogenic bacteria (Staphylococcus aureus, Salmonella typhimurium) was determined, together with effects on adhesion of pathogenic and probiotic bacteria to cultured Caco-2 cells. All polyphenols, except rutin, were found to affect the viability of representative gut flora in vitro, at doses likely to be present in the gastrointestinal tract, but to differing degrees. Naringenin and quercetin were the most active with the lowest minimum inhibitory concentrations for all the four bacteria tested. The remaining polyphenols had the most marked effect on the Gram positive enteropathogen S. aureus. Naringenin and phloridzin were the most effective inhibitors of S. typhimurium adherence to Caco-2 enterocytes while phloridzin and rutin enhanced the adherence of the probiotic L. rhamnosus. Polyphenols appear to have potential to alter gut microecology and, by affecting the total number of beneficial microflora in the gut, may confer positive gut health benefits.


Subject(s)
Bacterial Adhesion/drug effects , Bacterial Adhesion/physiology , Colon/microbiology , Enterocytes/microbiology , Flavonoids/pharmacology , Phenols/pharmacology , Caco-2 Cells , Colony Count, Microbial , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli/physiology , Humans , Lacticaseibacillus rhamnosus/drug effects , Lacticaseibacillus rhamnosus/growth & development , Lacticaseibacillus rhamnosus/physiology , Microbial Sensitivity Tests , Polyphenols , Probiotics , Salmonella typhimurium/drug effects , Salmonella typhimurium/growth & development , Salmonella typhimurium/physiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...