Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Eur J Nutr ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662018

ABSTRACT

PURPOSE: Impaired gut barrier function is associated with systemic inflammation and many chronic diseases. Undigested dietary proteins are fermented in the colon by the gut microbiota which produces nitrogenous metabolites shown to reduce barrier function in vitro. With growing evidence of sex-based differences in gut microbiotas, we determined whether there were sex by dietary protein interactions which could differentially impact barrier function via microbiota modification. METHODS: Fermentation systems were inoculated with faeces from healthy males (n = 5) and females (n = 5) and supplemented with 0.9 g of non-hydrolysed proteins sourced from whey, fish, milk, soya, egg, pea, or mycoprotein. Microbial populations were quantified using fluorescence in situ hybridisation with flow cytometry. Metabolite concentrations were analysed using gas chromatography, solid phase microextraction coupled with gas chromatography-mass spectrometry and ELISA. RESULTS: Increased protein availability resulted in increased proteolytic Bacteroides spp (p < 0.01) and Clostridium coccoides (p < 0.01), along with increased phenol (p < 0.01), p-cresol (p < 0.01), indole (p = 0.018) and ammonia (p < 0.01), varying by protein type. Counts of Clostridium cluster IX (p = 0.03) and concentration of p-cresol (p = 0.025) increased in males, while females produced more ammonia (p = 0.02), irrespective of protein type. Further, we observed significant sex-protein interactions affecting bacterial populations and metabolites (p < 0.005). CONCLUSIONS: Our findings suggest that protein fermentation by the gut microbiota in vitro is influenced by both protein source and the donor's sex. Should these results be confirmed through human studies, they could have major implications for developing dietary recommendations tailored by sex to prevent chronic illnesses.

2.
J Appl Microbiol ; 135(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38337173

ABSTRACT

AIMS: This study explored the effect of three different prebiotics, the human milk oligosaccharide 2'-fucosyllactose (2'-FL), an oligofructose-enriched inulin (fructo-oligosaccharide, or FOS), and a galacto-oligosaccaride (GOS) mixture, on the faecal microbiota from patients with ulcerative colitis (UC) using in vitro batch culture fermentation models. Changes in bacterial groups and short-chain fatty acid (SCFA) production were compared. METHODS AND RESULTS: In vitro pH controlled batch culture fermentation was carried out over 48 h on samples from three healthy controls and three patients with active UC. Four vessels were run, one negative control and one for each of the prebiotic substrates. Bacterial enumeration was carried out using fluorescence in situ hybridization with flow cytometry. SCFA quantification was performed using gas chromatography mass spectrometry. All substrates had a positive effect on the gut microbiota and led to significant increases in total SCFA and propionate concentrations at 48 h. 2'-FL was the only substrate to significantly increase acetate and led to the greatest increase in total SCFA concentration at 48 h. 2'-FL best suppressed Desulfovibrio spp., a pathogen associated with UC. CONCLUSIONS: 2'FL, FOS, and GOS all significantly improved the gut microbiota in this in vitro study and also led to increased SCFA.


Subject(s)
Colitis, Ulcerative , Prebiotics , Humans , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Fermentation , In Situ Hybridization, Fluorescence , Feces/microbiology , Fatty Acids, Volatile , Oligosaccharides/pharmacology , Bacteria/genetics
3.
Nutr Res Rev ; : 1-9, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38586996

ABSTRACT

Iron is essential for many physiological functions of the body, and it is required for normal growth and development. Iron deficiency (ID) is the most common form of micronutrient malnutrition and is particularly prevalent in infants and young children in developing countries. Iron supplementation is considered the most effective strategy to combat the risk of ID and ID anaemia (IDA) in infants, although iron supplements cause a range of deleterious gut-related problems in malnourished children. The purpose of this review is to assess the available evidence on the effect of iron supplementation on the gut microbiota during childhood ID and to further assess whether prebiotics offer any benefits for iron supplementation. Prebiotics are well known to improve gut-microbial health in children, and recent reports indicate that prebiotics can mitigate the adverse gut-related effects of iron supplementation in children with ID and IDA. Thus, provision of prebiotics alongside iron supplements has the potential for an enhanced strategy for combatting ID and IDA among children in the developing world. However, further understanding is required before the benefit of such combined treatments of ID in nutritionally deprived children across populations can be fully confirmed. Such enhanced understanding is of high relevance in resource-poor countries where ID, poor sanitation and hygiene, alongside inadequate access to good drinking water and poor health systems, are serious public health concerns.

4.
Microbiology (Reading) ; 169(3)2023 03.
Article in English | MEDLINE | ID: mdl-36947574

ABSTRACT

Staphylococcus aureus is a common colonizer of the human gut and in doing so it must be able to resist the actions of the host's innate defences. Bile salts are a class of molecules that possess potent antibacterial activity that control growth. Bacteria that colonize and survive in that niche must be able to resist the action of bile salts, but the mechanisms by which S. aureus does so are poorly understood. Here we show that FadB is a bile-induced oxidoreductase which mediates bile salt resistance and when heterologously expressed in Escherichia coli renders them resistant. Deletion of fadB attenuated survival of S. aureus in a model of the human distal colon.


Subject(s)
Cholates , Staphylococcal Infections , Humans , Staphylococcus aureus/genetics , Bile Acids and Salts/pharmacology , Oxidoreductases
5.
Eur J Nutr ; 62(5): 2205-2215, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37046122

ABSTRACT

PURPOSE: Prebiotic foods can be used to increase production of short-chain fatty acids (SCFA) in the gut. Of the SCFA, propionate is credited with the strongest anorectic activity. In previous work, a 50/50 blend of inulin and arabinoxylan was produced (I + AX) that significantly increased propionate production in an in vitro gut model. This study sought to establish whether chronic consumption of a prebiotic blend of I + AX decreases appetite and energy intake and increases intestinal propionate production in human participants. METHODS: MIXSAT (clinicaltrials.gov id: NCT02846454, August 2016) was a double-blind randomised acute-within-chronic crossover feeding trial in healthy adult men (n = 20). Treatments were 8 g per day I + AX for 21 days or weight-matched maltodextrin control. The primary outcome measure was perceived satiety and appetite during an acute study visit. Secondary outcomes were energy intake in an ad libitum meal, faecal SCFA concentration, and faecal microbiota composition. RESULTS: Perceived satiety and appetite were not affected by the intervention. I + AX was associated with a reduction in energy intake in an ad libitum meal, increased faecal SCFA concentration, and an increase in cell counts of Bifidobacteria, Lactobacilli, and other microbial genera associated with health. IMPLICATIONS: Chronic consumption of this blend of prebiotics decreased energy intake in a single sitting. Further studies are needed to confirm mechanism of action and to determine whether this might be useful in weight control.


Subject(s)
Appetite , Inulin , Adult , Male , Humans , Inulin/pharmacology , Propionates , Cross-Over Studies , Energy Intake , Fatty Acids, Volatile , Prebiotics
6.
Br J Nutr ; 126(2): 219-227, 2021 07 28.
Article in English | MEDLINE | ID: mdl-33032673

ABSTRACT

The recent COVID-19 pandemic has altered the face of biology, social interaction and public health worldwide. It has had a destructive effect upon millions of people and is approaching a devastating one million fatalities. Emerging evidence has suggested a link between the infection and gut microbiome status. This is one of the several factors that may contribute towards severity of infection. Given the fact that the gut is heavily linked to immunity, inflammatory status and the ability to challenge pathogens, it is worthwhile to consider dietary intervention of the gut microbiota as means of potentially challenging the viral outcome. In this context, probiotics and prebiotics have been used to mitigate similar respiratory infections. Here, we summarise links between the gut microbiome and COVID-19 infection, as well as propose mechanisms whereby probiotic and prebiotic interventions may act.


Subject(s)
COVID-19/microbiology , Gastrointestinal Microbiome , Humans , Prebiotics , Probiotics , SARS-CoV-2 , Synbiotics
7.
Molecules ; 25(16)2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32824081

ABSTRACT

In this study, we tested the growth inhibition effect of 22 individual ellagitannins and of pentagalloylglucose on four bacterial species, i.e., Clostridiales perfringens, Escherichia coli, Lactobacillus plantarum and Staphylococcus aureus. All tested compounds showed antimicrobial effects against S. aureus, and almost all against E. coli and C. perfringens. For L. plantarum, no or very weak growth inhibition was detected. The level of inhibition was the greatest for S. aureus and the weakest for C. perfringens. For S. aureus, the molecular size or flexibility of ellagitannins did not show a clear relationship with their antimicrobial activity, even though rugosins E and D and pentagalloylglucose with four or five free galloyl groups had a stronger growth inhibition effect than the other ellagitannins with glucopyranose cores but with less free galloyl groups. Additionally, our results with S. aureus showed that the oligomeric linkage of ellagitannin might have an effect on its antimicrobial activity. For E. coli, the molecular size, but not the molecular flexibility, of ellagitannins seemed to be an important factor. For C. perfringens, both the molecular size and the flexibility of ellagitannin were important factors. In previous studies, corilagin was used as a model for ellagitannins, but our results showed that other ellagitannins are much more efficacious; therefore, the antimicrobial effects of ellagitannins could be more significant than previously thought.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridiales/growth & development , Escherichia coli/growth & development , Hydrolyzable Tannins/pharmacology , Lactobacillus plantarum/growth & development , Staphylococcus aureus/growth & development , Clostridiales/drug effects , Escherichia coli/drug effects , Lactobacillus plantarum/drug effects , Staphylococcus aureus/drug effects
8.
Br J Nutr ; 121(5): 549-559, 2019 03.
Article in English | MEDLINE | ID: mdl-30688188

ABSTRACT

Wholegrain oats are known to modulate the human gut microbiota and have prebiotic properties (increase the growth of some health-promoting bacterial genera within the colon). Research to date mainly attributes these effects to the fibre content; however, oat is also a rich dietary source of polyphenols, which may contribute to the positive modulation of gut microbiota. In vitro anaerobic batch-culture experiments were performed over 24 h to evaluate the impact of two different doses (1 and 3 % (w/v)) of oat bran, matched concentrations of ß-glucan extract or polyphenol mix, on the human faecal microbiota composition using 16S RNA gene sequencing and SCFA analysis. Supplementation with oats increased the abundance of Proteobacteria (P <0·01) at 10 h, Bacteroidetes (P <0·05) at 24 h and concentrations of acetic and propionic acid increased at 10 and 24 h compared with the NC. Fermentation of the 1 % (w/v) oat bran resulted in significant increase in SCFA production at 24 h (86 (sd 27) v. 28 (sd 5) mm; P <0·05) and a bifidogenic effect, increasing the relative abundance of Bifidobacterium unassigned at 10 h and Bifidobacterium adolescentis (P <0·05) at 10 and 24 h compared with NC. Considering the ß-glucan treatment induced an increase in the phylum Bacteroidetes at 24 h, it explains the Bacteriodetes effects of oats as a food matrix. The polyphenol mix induced an increase in Enterobacteriaceae family at 24 h. In conclusion, in this study, we found that oats increased bifidobacteria, acetic acid and propionic acid, and this is mediated by the synergy of all oat compounds within the complex food matrix, rather than its main bioactive ß-glucan or polyphenols. Thus, oats as a whole food led to the greatest impact on the microbiota.


Subject(s)
Avena/chemistry , Bacteroidetes/drug effects , Bifidobacterium/drug effects , Gastrointestinal Microbiome/drug effects , Whole Grains , Acetic Acid/metabolism , Feces/microbiology , Fermentation/drug effects , Humans , Polyphenols/pharmacology , Prebiotics , Propionates/metabolism , Proteobacteria/drug effects , beta-Glucans/pharmacology
9.
Int J Mol Sci ; 20(21)2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31653078

ABSTRACT

Here, we reviewed emerging evidence on the role of the microbial community in colorectal carcinogenesis. A healthy gut microbiota promotes intestinal homeostasis and can exert anti-cancer effects; however, this microbiota also produces a variety of metabolites that are genotoxic and which can negatively influence epithelial cell behaviour. Disturbances in the normal microbial balance, known as dysbiosis, are frequently observed in colorectal cancer (CRC) patients. Microbial species linked to CRC include certain strains of Bacteroides fragilis, Escherichia coli, Streptococcus gallolyticus, Enterococcus faecalis and Fusobacterium nucleatum, amongst others. Whether these microbes are merely passive dwellers exploiting the tumour environment, or rather, active protagonists in the carcinogenic process is the subject of much research. The incidence of chemically-induced tumours in mice models varies, depending upon the presence or absence of these microorganisms, thus strongly suggesting influences on disease causation. Putative mechanistic explanations differentially link these strains to DNA damage, inflammation, aberrant cell behaviour and immune suppression. In the future, modulating the composition and metabolic activity of this microbial community may have a role in prevention and therapy.


Subject(s)
Colorectal Neoplasms/pathology , Gastrointestinal Microbiome , Animals , Bacteroides/isolation & purification , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/microbiology , DNA Damage , Fusobacterium/isolation & purification , Humans , Inflammation , Streptococcus/isolation & purification , Tumor Microenvironment
10.
Int J Food Sci Nutr ; 69(6): 696-704, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29334803

ABSTRACT

Interest in the consumption of gum acacia (GA) has been associated with beneficial health effects, which may be mediated in part by prebiotic activity. Two doses of GA and fructooligosaccharide (FOS) (1 and 2%) were tested for their efficacy over 48 h in pH- and temperature-controlled anaerobic batch cultures inoculated with human faeces. Samples were taken after 0, 5, 10, 24 and 48 h of fermentation. The selective effects of GA (increases in Bifidobacterium spp. and Lactobacillus spp.) were similar to those of the known prebiotic FOS. The 1% dose of substrates showed more enhanced selectivity compared to the 2% dose. The fermentation of GA also led to SCFA production, specifically increased acetate after 10, 24 and 48 h of fermentation, propionate after 48 h and butyrate after 24 and 48 h. In addition, FOS led to significant increase in the main SCFAs. These results suggest that GA displays potential prebiotic properties.


Subject(s)
Feces/microbiology , Gum Arabic/pharmacology , Microbiota/drug effects , Bifidobacterium/drug effects , Fermentation , Humans , Lactobacillus/drug effects , Prebiotics
11.
Br J Nutr ; 114(8): 1226-36, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26428278

ABSTRACT

The reported inverse association between the intake of plant-based foods and a reduction in the prevalence of colorectal cancer may be partly mediated by interactions between insoluble fibre and (poly)phenols and the intestinal microbiota. In the present study, we assessed the impact of palm date consumption, rich in both polyphenols and fibre, on the growth of colonic microbiota and markers of colon cancer risk in a randomised, controlled, cross-over human intervention study. A total of twenty-two healthy human volunteers were randomly assigned to either a control group (maltodextrin-dextrose, 37·1 g) or an intervention group (seven dates, approximately 50 g). Each arm was of 21 d duration and was separated by a 14-d washout period in a cross-over manner. Changes in the growth of microbiota were assessed by fluorescence in situ hybridisation analysis, whereas SCFA levels were assessed using HPLC. Further, ammonia concentrations, faecal water genotoxicity and anti-proliferation ability were also assessed using different assays, which included cell work and the Comet assay. Accordingly, dietary intakes, anthropometric measurements and bowel movement assessment were also carried out. Although the consumption of dates did not induce significant changes in the growth of select bacterial groups or SCFA, there were significant increases in bowel movements and stool frequency (P<0·01; n 21) and significant reductions in stool ammonia concentration (P<0·05; n 21) after consumption of dates, relative to baseline. Furthermore, date fruit intake significantly reduced genotoxicity in human faecal water relative to control (P<0·01; n 21). Our data indicate that consumption of date fruit may reduce colon cancer risk without inducing changes in the microbiota.


Subject(s)
Diet , Fruit , Intestine, Large/microbiology , Microbiota , Phoeniceae , Adolescent , Adult , Ammonia/analysis , Bacteroides/isolation & purification , Bifidobacterium/isolation & purification , Body Mass Index , Cell Proliferation , Cholesterol/blood , Clostridium/isolation & purification , Colon/microbiology , Colonic Neoplasms/prevention & control , Colony Count, Microbial , Cross-Over Studies , DNA Damage , Dietary Fats/administration & dosage , Dietary Fiber/administration & dosage , Dietary Proteins/administration & dosage , Energy Intake , Enterococcus/isolation & purification , Eubacterium/isolation & purification , Feces/chemistry , Feces/microbiology , Female , HT29 Cells , Humans , In Situ Hybridization, Fluorescence , Lactobacillus/isolation & purification , Male , Middle Aged , Ruminococcus/isolation & purification , Triglycerides/blood , Young Adult
12.
Br J Nutr ; 114(4): 586-95, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26218845

ABSTRACT

It is recognised that ageing induces various changes to the human colonic microbiota. Most relevant is a reduction in bifidobacteria, which is a health-positive genus. Prebiotics, such as galacto-oligosaccharides (GOS), are dietary ingredients that selectively fortify beneficial gut microbial groups. Therefore, they have the potential to reverse the age-related decline in bifidobacteria and modulate associated health parameters. We assessed the effect of GOS mixture (Bimuno (B-GOS)) on gut microbiota, markers of immune function and metabolites in forty elderly (age 65-80 years) volunteers in a randomised, double-blind, placebo (maltodextrin)-controlled, cross-over study. The intervention periods consisted of 10 weeks with daily doses of 5·5 g/d with a 4-week washout period in between. Blood and faecal samples were collected for the analyses of faecal bacterial populations and immune and metabolic biomarkers. B-GOS consumption led to significant increases in bacteroides and bifidobacteria, the latter correlating with increased lactic acid in faecal waters. Higher IL-10, IL-8, natural killer cell activity and C-reactive protein and lower IL-1ß were also observed. Administration of B-GOS to elderly volunteers may be useful in positively affecting the microbiota and some markers of immune function associated with ageing.


Subject(s)
Aging/immunology , Bacteria/growth & development , Gastrointestinal Tract/drug effects , Immune System/drug effects , Microbiota/drug effects , Oligosaccharides/pharmacology , Prebiotics , Aged , Aged, 80 and over , Aging/metabolism , Bacteroides/growth & development , Bifidobacterium/growth & development , Biomarkers/blood , C-Reactive Protein/metabolism , Cross-Over Studies , Double-Blind Method , Feces/microbiology , Female , Galactose/pharmacology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Humans , Immune System/metabolism , Immune System/microbiology , Interleukins/blood , Killer Cells, Natural/metabolism , Lactic Acid/metabolism , Male , Metabolomics
13.
Int J Food Sci Nutr ; 65(1): 79-88, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23941288

ABSTRACT

Imbalances in gut microbiota composition during ulcerative colitis (UC) indicate a role for the microbiota in propagating the disorder. Such effects were investigated using in vitro batch cultures (with/without mucin, peptone or starch) inoculated with faecal slurries from healthy or UC patients; the growth of five bacterial groups was monitored along with short-chain fatty acid (SCFA) production. Healthy cultures gave two-fold higher growth and SCFA levels with up to ten-fold higher butyrate production. Starch gave the highest growth and SCFA production (particularly butyrate), indicating starch-enhanced saccharolytic activity. Sulphate-reducing bacteria (SRB) were the predominant bacterial group (of five examined) for UC inocula whereas they were the minority group for the healthy inocula. Furthermore, SRB growth was stimulated by peptone presumably due to the presence of sulphur-rich amino acids. The results suggest raised SRB levels in UC, which could contribute to the condition through release of toxic sulphide.


Subject(s)
Amino Acids, Sulfur/metabolism , Colitis, Ulcerative/microbiology , Colon/microbiology , Dietary Proteins/administration & dosage , Fatty Acids, Volatile/metabolism , Intestinal Mucosa/microbiology , Sulfur-Reducing Bacteria/growth & development , Amino Acids, Sulfur/adverse effects , Butyric Acid/metabolism , Colitis, Ulcerative/diet therapy , Colitis, Ulcerative/metabolism , Colon/metabolism , Diet, Protein-Restricted , Dietary Proteins/adverse effects , Dietary Proteins/metabolism , Feces/microbiology , Female , Fermentation , Gram-Negative Bacteria/classification , Gram-Negative Bacteria/growth & development , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/classification , Gram-Positive Bacteria/growth & development , Gram-Positive Bacteria/isolation & purification , Gram-Positive Bacteria/metabolism , Humans , Intestinal Mucosa/metabolism , Male , Microbial Viability , Middle Aged , Molecular Typing , Mucins/metabolism , Peptones/metabolism , Starch/metabolism , Sulfur-Reducing Bacteria/classification , Sulfur-Reducing Bacteria/isolation & purification , Sulfur-Reducing Bacteria/metabolism
14.
Food Chem ; 409: 135286, 2023 May 30.
Article in English | MEDLINE | ID: mdl-36599291

ABSTRACT

Culinary herbs and spices have previously been recognised for their potential impact on health through antioxidant and antimicrobial properties. They may also be promotors of positive microbial modulation by stimulating beneficial gut bacteria during fermentation, increasing the production of short chain fatty acids and thereby exhibiting a prebiotic effect. In the present paper, current literature around herb and spice consumption, gut microbiota modulation and prospective health benefits were reviewed. Herb and spice consumption can positively modulate gut microbes and possibly play an important role in inflammation related afflictions such as obesity. Current literature indicates that few human studies have been conducted to confirm the impact of herb and spice consumption on gut microbiota in connection with prospective health outcomes and inconsistencies in conclusions therefore remain.


Subject(s)
Gastrointestinal Microbiome , Spices , Humans , Spices/analysis , Antioxidants , Prebiotics , Bacteria/genetics
15.
Metabolites ; 13(3)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36984760

ABSTRACT

Escherichia coli and Staphylococcus aureus are globally among the most prominent bacterial strains associated with antibacterial resistance-caused deaths. Naturally occurring polyphenols, such as hydrolyzable tannins, have been shown to potently inhibit E. coli and S. aureus. The current study investigated the metabolome changes of E. coli and S. aureus cultures after treatments with different hydrolyzable tannins using an NMR metabolomics approach. Additionally, the effect of these tannin treatments influencing a more complex bacterial system was studied in a biomimetic setting with fecal samples inoculated into the growth medium. Metabolite concentration changes were observed in all three scenarios: E. coli, S. aureus, and fecal batch culture. The metabolome of E. coli was more altered by the tannin treatments than S. aureus when compared to control cultures. A dimeric hydrolyzable tannin, rugosin D, was found to be the most effective of the studied compounds in influencing bacterial metabolome changes and in inhibiting E. coli and S. aureus growth. It was also observed that the tannin structure should have both hydrophobic and hydrophilic regions to efficiently influence E. coli and S. aureus growth.

16.
Trends Microbiol ; 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38065786

ABSTRACT

The gut microbiome in the inflammatory bowel disease, ulcerative colitis (UC), is different to that of healthy controls. Patients with UC have relative reductions in abundance of Firmicutes and Bifidobacterium in the colon, and an increase in sulfate-reducing bacteria. Prebiotics are dietary substrates which are selectively metabolised by the human colonic microbiota to confer health benefits to the host. This review explores our current understanding of the potential benefits of prebiotics on various clinical, biochemical, and microbiological endpoints in UC, including new perspectives gained from recent studies in the field. This review looks to the future and highlights the need for appropriately designed trials to explore this potentially exciting new avenue for the treatment of UC.

17.
Nutrients ; 15(11)2023 May 30.
Article in English | MEDLINE | ID: mdl-37299530

ABSTRACT

Probiotic supplements are increasingly being used to target the gut microbiome with a view to improving cognitive and psychological function via the gut-brain axis. One possible mechanism behind the effect of probiotics is through alterations to microbially-derived metabolites including short-chain fatty acids (SCFA) and neurotransmitters. However, research to date has largely been conducted in animal models or under conditions irrelevant to the human gastrointestinal tract (GIT). The aim of the current work was therefore to use anaerobic, pH controlled in vitro batch cultures to (a) assess the production of neuroactive metabolites in human faecal microbiota under conditions relevant to the human GIT, and (b) to explore how several pre-selected probiotic strains may affect bacterial composition and metabolite production. Enumeration of bacteria was assessed using fluorescence in situ hybridisation with flow cytometry, and concentrations of SCFAs and neurotransmitters were measured using gas chromatography and liquid chromatography mass spectroscopy, respectively. GABA, serotonin, tryptophan, and dopamine were successfully detected, suggesting some level of microbial derivation. The addition of Lactococcus lactis W58 and Lactobacillus rhamnosus W198 resulted in a significant increase in lactate after 8 h of fermentation, while no significant effect of probiotics on bacterial composition or neurotransmitter production was found.


Subject(s)
Microbiota , Probiotics , Humans , Animals , Batch Cell Culture Techniques , Dietary Supplements , Bacteria/genetics , Bacteria/metabolism , Feces/microbiology
18.
Front Microbiol ; 14: 1074637, 2023.
Article in English | MEDLINE | ID: mdl-36910170

ABSTRACT

Although iron is an essential nutrient for humans, as well as for almost all other organisms, it is poorly absorbed (~15%) from the diet such that most passes through the upper gut into the large intestine. The colonic microbiota is thus exposed to, and potentially influenced by, such residual iron which could have an impact on human health. The aim of the research described here is to determine how the major forms of dietary iron (inorganic iron and haem) influence metabolic activity and composition of the human gut microbiota by utilizing an in vitro parallel, pH-controlled anaerobic batch culture approach. Controlled iron provision was enabled by the design of a 'modified' low-iron gut-model medium whereby background iron content was reduced from 28 to 5 µM. Thus, the impact of both low and high levels of inorganic and haem iron (18-180 µM and 7.7-77 µM, respectively) could be explored. Gut-microbiota composition was determined using next generation sequencing (NGS) based community profiling (16S rRNA gene sequencing) and flow-fluorescent in situ hybridization (FISH). Metabolic-end products (organic acids) were quantified using gas chromatography (GC) and iron incorporation was estimated by inductively coupled plasma optical emission spectroscopy (ICP-OES). Results showed that differences in iron regime induced significant changes in microbiota composition when low (0.1% w/v) fecal inoculation levels were employed. An increase in haem levels from 7.7 to 77 µM (standard levels employed in gut culture studies) resulted in reduced microbial diversity, a significant increase in Enterobacteriaceae and lower short chain fatty acid (SCFA) production. These effects were countered when 18 µM inorganic iron was also included into the growth medium. The results therefore suggest that high-dietary haem may have a detrimental effect on health since the resulting changes in microbiota composition and SCFA production are indicators of an unhealthy gut. The results also demonstrate that employing a low inoculum together with a low-iron gut-model medium facilitated in vitro investigation of the relationship between iron and the gut microbiota.

19.
Br J Nutr ; 107(10): 1466-75, 2012 May.
Article in English | MEDLINE | ID: mdl-21910949

ABSTRACT

Faecal microbial changes associated with ageing include reduced bifidobacteria numbers. These changes coincide with an increased risk of disease development. Prebiotics have been observed to increase bifidobacteria numbers within humans. The present study aimed to determine if prebiotic galacto-oligosaccharides (GOS) could benefit a population of men and women of 50 years and above, through modulation of faecal microbiota, fermentation characteristics and faecal water genotoxicity. A total of thirty-seven volunteers completed this randomised, double-blind, placebo-controlled crossover trial. The treatments - juice containing 4 g GOS and placebo - were consumed twice daily for 3 weeks, preceded by 3-week washout periods. To study the effect of GOS on different large bowel regions, three-stage continuous culture systems were conducted in parallel using faecal inocula from three volunteers. Faecal samples were microbially enumerated by quantitative PCR. In vivo, following GOS intervention, bifidobacteria were significantly more compared to post-placebo (P = 0·02). Accordingly, GOS supplementation had a bifidogenic effect in all in vitro system vessels. Furthermore, in vessel 1 (similar to the proximal colon), GOS fermentation led to more lactobacilli and increased butyrate. No changes in faecal water genotoxicity were observed. To conclude, GOS supplementation significantly increased bifidobacteria numbers in vivo and in vitro. Increased butyrate production and elevated bifidobacteria numbers may constitute beneficial modulation of the gut microbiota in a maturing population.


Subject(s)
Butyric Acid/metabolism , Colon/microbiology , Feces/microbiology , Galactose/pharmacology , Lactobacillus , Oligosaccharides/pharmacology , Prebiotics , Aged , Aged, 80 and over , Colon/metabolism , Cross-Over Studies , Dietary Supplements , Double-Blind Method , Female , Fermentation , Humans , Male , Metagenome , Middle Aged , Polymerase Chain Reaction
20.
Br J Nutr ; 108(3): 471-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22099384

ABSTRACT

In this placebo-controlled, double-blind, crossover human feeding study, the effects of polydextrose (PDX; 8 g/d) on the colonic microbial composition, immune parameters, bowel habits and quality of life were investigated. PDX is a complex glucose oligomer used as a sugar replacer. The main goal of the present study was to identify the microbial groups affected by PDX fermentation in the colon. PDX was shown to significantly increase the known butyrate producer Ruminococcus intestinalis and bacteria of the Clostridium clusters I, II and IV. Of the other microbial groups investigated, decreases in the faecal Lactobacillus-Enterococcus group were demonstrated. Denaturing gel gradient electrophoresis analysis showed that bacterial profiles between PDX and placebo treatments were significantly different. PDX was shown to be slowly degraded in the colon, and the fermentation significantly reduced the genotoxicity of the faecal water. PDX also affected bowel habits of the subjects, as less abdominal discomfort was recorded and there was a trend for less hard and more formed stools during PDX consumption. Furthermore, reduced snacking was observed upon PDX consumption. This study demonstrated the impact of PDX on the colonic microbiota and showed some potential for reducing the risk factors that may be associated with colon cancer initiation.


Subject(s)
Colon/microbiology , Feces/microbiology , Glucans/pharmacology , Adult , Clostridium/drug effects , Clostridium/growth & development , Cluster Analysis , Colon/drug effects , Cross-Over Studies , Denaturing Gradient Gel Electrophoresis/methods , Double-Blind Method , Eating/drug effects , Enterococcus/drug effects , Enterococcus/growth & development , Feces/chemistry , Female , Fermentation , Humans , Lactobacillus/drug effects , Lactobacillus/growth & development , Male , Middle Aged , Polymerase Chain Reaction/methods , Prebiotics , Risk Factors , Ruminococcus/drug effects , Ruminococcus/growth & development , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL