Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Nutr ; 152(6): 1426-1437, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35102419

ABSTRACT

BACKGROUND: Resistant starch (RS) confers many health benefits, mostly through the microbial production of SCFAs, but foods containing appreciable RS are limited. High-amylose wheat (HAW) is high in RS and lowers the glycemic response of foods, but whether it can improve gastrointestinal health measures is unknown. OBJECTIVES: The objective of this study was to determine whether daily consumption of HAW food products improved markers of gastrointestinal health in healthy men and women compared with similar foods made from conventional wheat. METHODS: Eighty healthy adults (47 women and 33 men) were enrolled in a 4-arm parallel, randomized-controlled, double-blind trial. After a 2-wk low-dietary fiber run-in period, they were randomly allocated to 1 of 4 treatment groups: low-amylose wheat (LAW)-refined (LAW-R), LAW-wholemeal (LAW-W), HAW-refined (HAW-R), and HAW-wholemeal (HAW-W) and consumed the assigned test bread (160 g/d) and biscuits (75 g/d) for 4 wk. Fecal biochemical markers were measured at baseline and 4 wk. Microbial abundance and diversity were quantified using 16S ribosomal RNA sequencing and perceived gut comfort by a semiquantitative questionnaire completed at baseline, 2 wk, and 4 wk. RESULTS: HAW showed similar effects on fecal output and excretion of total SCFA compared with LAW, but changes were observed in secondary measures for the refined treatment groups. At 4 wk, the HAW-R group had 38% higher fecal butyrate excretion than the LAW-R group (P < 0.05), and higher fecal SCFA-producing bacteria, Roseburia inulinivorans (P < 0.001), than at baseline. In comparison with baseline, LAW-R increased fecal p-cresol concentration, and fecal abundance of a p-cresol-producing bacterium, Clostridium from the Peptostreptococcaceae family, but both were reduced by HAW-R. Amylose level did not affect measures of fecal consistency or adversely affecting digestive comfort. CONCLUSIONS: Increasing RS intake of healthy adults by substituting refined conventional wheat with refined HAW modulates fecal metabolites and microbes associated with gastrointestinal health.This trial was registered at anzctr.org.au as ACTRN12618001060235.


Subject(s)
Gastrointestinal Microbiome , Adult , Amylose , Bacteria , Biomarkers , Feces/microbiology , Female , Flour , Humans , Male , Resistant Starch , Triticum
2.
Nutrients ; 12(8)2020 Aug 09.
Article in English | MEDLINE | ID: mdl-32784883

ABSTRACT

A probiotic and prebiotic food ingredient combination was tested for synergistic functioning in modulation of the colonic microbiome and remediation of the gastrointestinal immune and inflammatory responses in a spontaneous colitic mouse model. Bacillus coagulans MTCC5856 spores with capability to metabolise complex plant polysaccharides were supplemented with complex whole-plant prebiotic sugarcane fibre (PSCF). The combined and individual efficacies were tested for their influence on the outcomes of chronic inflammation in Muc2 mutant colitic Winnie mice. The mice were fed normal chow diet supplemented with either ingredient or a combination for 21 days. Synbiotic combined supplementation ameliorated clinical symptoms and histological colonic damage scores more effectively than either B. coagulans or PSCF alone. PSCF and B. coagulans alone also induced considerable immunomodulatory effects. Synbiotic supplementation however was the most efficacious in modulating the overall immune profile compared to the unsupplemented Winnie-control. The augmented synbiotic effect could potentially be due to a combination of increased levels of fermentation products, direct immune-modulating abilities of the components, their capability to reduce colonic epithelial damage and/or modulation of the microbiota. The beneficial effects of the supplementation with a complex plant fibre and a fibre-degrading probiotic parallel the effects seen in human microbiota with high plant fibre diets.


Subject(s)
Colitis/immunology , Colitis/microbiology , Dietary Fiber/administration & dosage , Prebiotics/administration & dosage , Probiotics/administration & dosage , Synbiotics/administration & dosage , Animals , Bacillus coagulans , Colon/immunology , Colon/microbiology , Disease Models, Animal , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Inflammation , Mice
3.
Metabolites ; 10(3)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155792

ABSTRACT

Our understanding of the human gut microbiome has grown exponentially. Advances in genome sequencing technologies and metagenomics analysis have enabled researchers to study microbial communities and their potential function within the context of a range of human gut related diseases and disorders. However, up until recently, much of this research has focused on characterizing the gut microbiological community structure and understanding its potential through system wide (meta) genomic and transcriptomic-based studies. Thus far, the functional output of these microbiomes, in terms of protein and metabolite expression, and within the broader context of host-gut microbiome interactions, has been limited. Furthermore, these studies highlight our need to address the issues of individual variation, and of samples as proxies. Here we provide a perspective review of the recent literature that focuses on the challenges of exploring the human gut microbiome, with a strong focus on an integrated perspective applied to these themes. In doing so, we contextualize the experimental and technical challenges of undertaking such studies and provide a framework for capitalizing on the breadth of insight such approaches afford. An integrated perspective of the human gut microbiome and the linkages to human health will pave the way forward for delivering against the objectives of precision medicine, which is targeted to specific individuals and addresses the issues and mechanisms in situ.

4.
Eur J Nutr ; 59(8): 3669-3689, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32067099

ABSTRACT

PURPOSE: The research goal is to develop dietary strategies to help address the growing incidence of inflammatory bowel diseases (IBD). This study has investigated the effectiveness of green banana resistant starch (GBRS) and probiotic Bacillus coagulans MTCC5856 spores for the amelioration of dextran-sulfate sodium (DSS)-induced colitis in mice. METHODS: Eight-week-old C57BL/6 mice were fed standard rodent chow diet supplemented with either B. coagulans, GBRS or its synbiotic combination. After 7 days supplementation, colitis was induced by adding 2% DSS in drinking water for 7 days while continuing the supplemented diets. Animal health was monitored and after 14 days all animals were sacrificed to measure the biochemical and histochemical changes associated with each supplement type. RESULTS: The disease activity index and histological damage score for DSS-control mice (6.1, 17.1, respectively) were significantly higher (p < 0.0001) than the healthy mice. Synbiotic supplementation alleviated these markers (- 67%, - 94% respectively) more adequately than B. coagulans (- 52%, - 58% respectively) or GBRS (- 57%, - 26%, respectively) alone. Compared to DSS-control synbiotic supplementation significantly (p < 0.0001) maintained expressions of tight junction proteins. Moreover, synbiotic effects accounted for ~ 40% suppression of IL-1ß and ~ 29% increase in IL-10 levels in serum while also reducing C-reactive protein (- 37%) compared to that of the DSS-control. While, B. coagulans alone could not induce additional levels of short-chain fatty acid (SCFA) production beyond the caecum, the synbiotic combination with GBRS resulted in substantial increased SCFA levels across the whole length of the colon. CONCLUSION: The synbiotic supplementation with B. coagulans and GBRS ameliorated the overall inflammatory status of the experimental IBD model via synergistic functioning. This supports researching its application in mitigating inflammation in human IBD.


Subject(s)
Bacillus coagulans , Colitis , Inflammatory Bowel Diseases , Musa , Probiotics , Synbiotics , Animals , Colon , Dextran Sulfate/toxicity , Disease Models, Animal , Inflammation , Inflammatory Bowel Diseases/therapy , Mice , Mice, Inbred C57BL , Prebiotics , Resistant Starch , Spores, Bacterial
5.
Nutrients ; 11(6)2019 Jun 07.
Article in English | MEDLINE | ID: mdl-31181695

ABSTRACT

Distribution of the microbiota varies according to the location in the gastrointestinal (GI) tract. Thus, dysbiosis during aging may not be limited to faecal microbiota and extend to the other parts of the GI tract, especially the cecum and colon. Lactobacillus acidophilus DDS-1, a probiotic strain, has been shown to modulate faecal microbiota and its associated metabolic phenotype in aging mice. In the present study, we investigated the effect of L. acidophilus DDS-1 supplementation on caecal- and mucosal-associated microbiota, short-chain fatty acids (SCFAs) and immunological profiles in young and aging C57BL/6J mice. Besides differences in the young and aging control groups, we observed microbial shifts in caecal and mucosal samples, leading to an alteration in SCFA levels and immune response. DDS-1 treatment increased the abundances of beneficial bacteria such as Akkermansia spp. and Lactobacillus spp. more effectively in caecal samples than in mucosal samples. DDS-1 also enhanced the levels of butyrate, while downregulating the production of inflammatory cytokines (IL-6, IL-1ß, IL-1α, MCP-1, MIP-1α, MIP-1ß, IL-12 and IFN-γ) in serum and colonic explants. Our findings suggest distinct patterns of intestinal microbiota, improvements in SCFA and immunological profiles with DDS-1 supplementation in aging mice.


Subject(s)
Aging , Butyric Acid/metabolism , Dysbiosis/prevention & control , Gastrointestinal Microbiome , Inflammation/prevention & control , Lactobacillus acidophilus/growth & development , Probiotics/therapeutic use , Aging/immunology , Aging/metabolism , Animals , Bacteria/growth & development , Cecum/microbiology , Colon/metabolism , Colon/microbiology , Cytokines/blood , Cytokines/metabolism , Down-Regulation , Dysbiosis/microbiology , Fatty Acids, Volatile/metabolism , Feces/microbiology , Inflammation/microbiology , Intestinal Mucosa/microbiology , Mice, Inbred C57BL , Models, Animal
6.
Nutrients ; 11(4)2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30979002

ABSTRACT

Inflammatory bowel diseases (IBD) are a chronic inflammatory disorders with increasing global incidence. Synbiotic, which is a two-point approach carrying probiotic and prebiotic components in mitigating inflammation in IBD, is thought to be a pragmatic approach owing to the synergistic outcomes. In this study, the impacts of dietary supplementation with probiotic Bacillus coagulans MTCC5856 spores (B. coagulans) and prebiotic whole plant sugar cane fibre (PSCF) was assessed using a murine model of IBD. Eight-week-old C57BL/6 mice were fed a normal chow diet supplemented with either B. coagulans, PSCF or its synbiotic combination. After seven days of supplementation, colitis was induced with dextran sulfate sodium (DSS) in drinking water for seven days during the continuation of the supplemented diets. Synbiotic supplementation ameliorated disease activity index and histological score (-72%, 7.38, respectively), more effectively than either B. coagulans (-47%, 10.1) and PSCF (-53%, 13.0) alone. Synbiotic supplementation also significantly (p < 0.0001) prevented the expression of tight junction proteins and modulated the altered serum IL-1ß (-40%), IL-10 (+26%), and C-reactive protein (CRP) (-39%) levels. Synbiotic supplementations also raised the short-chain fatty acids (SCFA) profile more extensively compared to the unsupplemented DSS-control. The synbiotic health outcome effect of the probiotic and prebiotic combinations may be associated with a synergistic direct immune-regulating efficacy of the components, their ability to protect epithelial integrity, stimulation of probiotic spores by the prebiotic fibre, and/or with stimulation of greater levels of fermentation of fibres releasing SCFAs that mediate the reduction in colonic inflammation. Our model findings suggest synbiotic supplementation should be tested in clinical trials.


Subject(s)
Dietary Fiber/administration & dosage , Inflammatory Bowel Diseases/therapy , Probiotics/administration & dosage , Saccharum , Spores, Bacterial , Synbiotics/administration & dosage , Animals , Bacillus coagulans , C-Reactive Protein/analysis , Colon/ultrastructure , Diet , Dietary Supplements , Disease Models, Animal , Female , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-1beta/blood , Male , Mice , Mice, Inbred C57BL , Tight Junctions/pathology
7.
Nutrients ; 10(9)2018 Sep 06.
Article in English | MEDLINE | ID: mdl-30200669

ABSTRACT

Recent evidence suggests that gut microbiota shifts can alter host metabolism even during healthy aging. Lactobacillus acidophilus DDS-1, a probiotic strain, has shown promising probiotic character in vitro, as well as in clinical studies. The present study was carried out to investigate whether DDS-1 can modulate the host metabolic phenotype under the condition of age-affected gut microbial shifts in young and aging C57BL/6J mice. Collected fecal samples were analyzed using 16S rRNA gene sequencing for identifying gut microbiota and untargeted gas chromatography-mass spectrometry (GC-MS) metabolomics analysis. Gut microbial shifts were observed in the control groups (young and aging), leading to an alteration in metabolism. Principal coordinate analysis (PCoA) of microbiota indicated distinct separation in both the DDS-1-treated groups. L. acidophilus DDS-1 increased the relative abundances of beneficial bacteria, such as Akkermansia muciniphila and Lactobacillus spp., and reduced the relative levels of opportunistic bacteria such as Proteobacteria spp. Metabolic pathway analysis identified 10 key pathways involving amino acid metabolism, protein synthesis and metabolism, carbohydrate metabolism, and butanoate metabolism. These findings suggest that modulation of gut microbiota by DDS-1 results in improvement of metabolic phenotype in the aging mice.


Subject(s)
Aging/metabolism , Energy Metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Lactobacillus acidophilus/physiology , Probiotics/administration & dosage , Age Factors , Animals , Feces/chemistry , Feces/microbiology , Female , Gas Chromatography-Mass Spectrometry , Male , Metabolomics/methods , Mice, Inbred C57BL , Phenotype , Ribotyping
8.
Inflamm Bowel Dis ; 22(12): 2767-2787, 2016 12.
Article in English | MEDLINE | ID: mdl-27824648

ABSTRACT

BACKGROUND: Dysbiosis of the gut microbiota may be involved in the pathogenesis of inflammatory bowel disease (IBD). However, the mechanisms underlying the role of the intestinal microbiome and metabolome in IBD onset and its alteration during active treatment and recovery remain unknown. Animal models of chronic intestinal inflammation with similar microbial and metabolomic profiles would enable investigation of these mechanisms and development of more effective treatments. Recently, the Winnie mouse model of colitis closely representing the clinical symptoms and characteristics of human IBD has been developed. In this study, we have analyzed fecal microbial and metabolomic profiles in Winnie mice and discussed their relevance to human IBD. METHODS: The 16S rRNA gene was sequenced from fecal DNA of Winnie and C57BL/6 mice to define operational taxonomic units at ≥97% similarity threshold. Metabolomic profiling of the same fecal samples was performed by gas chromatography-mass spectrometry. RESULTS: Composition of the dominant microbiota was disturbed, and prominent differences were evident at all levels of the intestinal microbiome in fecal samples from Winnie mice, similar to observations in patients with IBD. Metabolomic profiling revealed that chronic colitis in Winnie mice upregulated production of metabolites and altered several metabolic pathways, mostly affecting amino acid synthesis and breakdown of monosaccharides to short chain fatty acids. CONCLUSIONS: Significant dysbiosis in the Winnie mouse gut replicates many changes observed in patients with IBD. These results provide justification for the suitability of this model to investigate mechanisms underlying the role of intestinal microbiota and metabolome in the pathophysiology of IBD.


Subject(s)
Colitis/microbiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Inflammatory Bowel Diseases/microbiology , Metabolome/genetics , Animals , Disease Models, Animal , Dysbiosis/microbiology , Gas Chromatography-Mass Spectrometry , Humans , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/analysis
9.
Trials ; 14: 404, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24279904

ABSTRACT

BACKGROUND: Age-related cognitive decline (ARCD) is of major societal concern in an ageing population, with the development of dietary supplements providing a promising avenue for amelioration of associated deficits. Despite initial interest in the use of phospholipids (PLs) for ARCD, in recent years there has been a hiatus in such research. Because of safety concerns regarding PLs derived from bovine cortex, and the equivocal efficacy of soybean-derived PLs, there is an important need for the development of new PL alternatives. Phospholipids derived from milk proteins represent one potential candidate treatment. METHODS: In order to reduce the effects of age-associated memory impairment (AAMI) the Phospholipid Intervention for Cognitive Ageing Reversal (PLICAR) was developed to test the efficacy of a milk protein concentrate rich in natural, non-synthetic milk phospholipids (Lacprodan® PL-20). PLICAR is a randomized, double-blind, placebo-controlled parallel-groups study where 150 (N = 50/group) AAMI participants aged > 55 years will be randomized to receive a daily supplement of Lacprodan® PL-20 or one of two placebos (phospholipid-free milk protein concentrate or inert rice starch) over a 6-month (180-day) period. Participants will undergo testing at baseline, 90 days and 180 days. The primary outcome is a composite memory score from the Rey Auditory Verbal Learning Test. Secondary outcomes include cognitive (verbal learning, working memory, prospective and retrospective memory, processing speed and attention), mood (depression, anxiety, stress and visual analogue scales), cardiovascular (blood pressure, blood velocity and pulse wave pressure), gastrointestinal microbiota and biochemical measures (oxidative stress, inflammation, B vitamins and Homocysteine, glucoregulation and serum choline). Allelic differences in the Apolipoprotein E and (APOE) and Methylenetetrahydrofolate reductase (MTHFR) gene will be included for subgroup analysis. A subset (N = 60; 20/group)) will undergo neuroimaging using functional magnetic resonance imaging (fMRI) and magnetoencephalography (MEG) in order to further explore in vivo central mechanisms of action of Lacprodan® PL-20. This study will enable evaluation of the efficacy of milk-derived phospholipids for AAMI, and their mechanisms of action. TRIAL REGISTRATION: The trial is jointly funded by Arla Foods and Swinburne University of Technology, currently recruiting and is registered on the Australian New Zealand Clinical Trials Registry as ACTRN12613000347763.


Subject(s)
Clinical Protocols , Memory Disorders/drug therapy , Milk Proteins/therapeutic use , Phospholipids/therapeutic use , Affect/drug effects , Aged , Aging , Cognition/drug effects , Double-Blind Method , Humans , Middle Aged , Milk Proteins/pharmacology , Outcome Assessment, Health Care , Phospholipids/pharmacology
10.
Autism Res ; 5(6): 419-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22997101

ABSTRACT

Many children with autism spectrum disorders (ASDs) suffer from gastrointestinal problems such as diarrhoea, constipation and abdominal pain. This has stimulated investigations into possible abnormalities of intestinal microbiota in autistic patients. Therefore, we designed this study to identify differences (and/or similarities) in the microbiota of children with autism (without gastrointestinal dysfunction: n = 23; with gastrointestinal dysfunction: n = 28) and their neurotypical siblings (n = 53) who share a similar environment using bacterial tag-encoded FLX amplicon pyrosequencing. Regardless of the diagnosis and sociodemographic characteristics, overall, Firmicutes (70%), Bacteroidetes (20%) and Proteobacteria (4%) were the most dominant phyla in samples. Results did not indicate clinically meaningful differences between groups. The data do not support the hypothesis that the gastrointestinal microbiota of children with ASD plays a role in the symptomatology of ASD. Other explanations for the gastrointestinal dysfunction in this population should be considered including elevated anxiety and self-restricted diets.


Subject(s)
Autistic Disorder/microbiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Tract/microbiology , Metagenome , Siblings , Autistic Disorder/complications , Child , Child, Preschool , Feces/microbiology , Female , Gastrointestinal Diseases/complications , Humans , Male , Principal Component Analysis
SELECTION OF CITATIONS
SEARCH DETAIL