Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.471
Filter
1.
Nutrients ; 16(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125327

ABSTRACT

A fiber-rich diet is considered beneficial for gut health. An inflamed gut with a dysbiotic bacterial community can result in altered fiber metabolism depending on the fiber's physicochemical properties. This study examined the effect of fiber's physicochemical properties on fiber fermentation in the presence of healthy and colitis-associated bacteria. Sixteen fibers with different levels of solubility, complexity, and fermentation rate were used in in vitro fermentation with healthy human gut bacteria. Resistant maltodextrins (RMD), pectin (HMP), inulin (ChIn), and wheat bran (WB) were selected for fermentation using ulcerative colitis (UC)-associated bacteria to assess bacterial dysbiosis effect. UC-associated gut microbiota showed a significant reduction in α-and ß-diversity indices compared to healthy-associated microbiota. The differences in the gut microbiota composition and diversity between the donors resulted in decreased fermentation rates with UC-associated bacteria. Fiber fermentation metabolites, short-chain fatty acids (SCFA) and gas production were significantly lower in the presence of UC-associated bacteria for all four fibers tested. Overall, we conclude that dietary fiber properties and microbial dysbiosis are influential in fiber fermentation and metabolite production in the gut.


Subject(s)
Bacteria , Dietary Fiber , Dysbiosis , Fatty Acids, Volatile , Fermentation , Gastrointestinal Microbiome , Dietary Fiber/pharmacology , Gastrointestinal Microbiome/physiology , Humans , Dysbiosis/microbiology , Bacteria/metabolism , Bacteria/classification , Fatty Acids, Volatile/metabolism , Pectins/metabolism , Colitis, Ulcerative/microbiology , Inulin/metabolism , Male , Adult , Female , Polysaccharides
2.
Nutrients ; 16(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125398

ABSTRACT

Persimmon fruit processing-derived waste and by-products, such as peels and pomace, are important sources of dietary fiber and phytochemicals. Revalorizing these by-products could help promote circular nutrition and agricultural sustainability while tackling dietary deficiencies and chronic diseases. In this study, fiber-rich fractions were prepared from the by-products of Sharoni and Brilliant Red persimmon varieties. These fractions were quantified for their phenolic composition and assessed for their ability to promote the growth of beneficial human colonic Firmicutes species and for their in vitro anti-inflammatory potential. Gallic and protocatechuic acids, delphinidin, and cyanidin were the main phenolics identified. Faecalibacterium prausnitzii strains showed significantly higher growth rates in the presence of the Brilliant Red fraction, generating more than double butyrate as a proportion of the total short-chain fatty acids (39.5% vs. 17.8%) when compared to glucose. The fiber-rich fractions significantly decreased the inflammatory effect of interleukin-1ß in Caco-2 cells, and the fermented fractions (both from Sharoni and Brilliant Red) significantly decreased the inflammatory effect of interleukin-6 and tumor necrosis factor-α in the RAW 264.7 cells. Therefore, fiber-rich fractions from persimmon by-products could be part of nutritional therapies as they reduce systemic inflammation, promote the growth of beneficial human gut bacteria, and increase the production of beneficial microbial metabolites such as butyrate.


Subject(s)
Anti-Inflammatory Agents , Colon , Dietary Fiber , Diospyros , Humans , Dietary Fiber/pharmacology , Dietary Fiber/analysis , Diospyros/chemistry , Mice , Anti-Inflammatory Agents/pharmacology , Colon/microbiology , Colon/drug effects , Colon/metabolism , Animals , RAW 264.7 Cells , Caco-2 Cells , Gastrointestinal Microbiome/drug effects , Firmicutes , Faecalibacterium prausnitzii , Fruit/chemistry , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Hydroxybenzoates/pharmacology , Hydroxybenzoates/analysis , Phenols/pharmacology , Phenols/analysis , Fermentation , Gallic Acid/pharmacology , Anthocyanins/pharmacology , Anthocyanins/analysis
3.
Nutrients ; 16(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39125432

ABSTRACT

Phallus atrovolvatus, a wild edible mushroom, has attracted increasing interest for consumption due to its unique taste and beneficial health benefits. This study determined the chemical components in the so-called fruiting body during the egg and mature stages and investigated its gut microbiota-modulating activities. The egg stage contained higher total carbohydrates, dietary fiber, glucans, ash, and fat, while the total protein content was lower than in the mature stage. Two consumption forms, including cooked mushrooms and a mushroom aqueous extract from both stages, were used in this study. An in vitro gut fermentation was performed for 24 h to assess gut microbiota regulation. All mushroom-supplemented fermentations increased short-chain fatty acid (SCFA) production compared to the blank control. Furthermore, all mushroom supplementations promoted the growth of Bifidobacterium and Streptococcus. Samples from the mature stage increased the relative abundance of Clostridium sensu stricto 1, while those from the egg stage increased the Bacteroides group. The inhibition of harmful bacteria, including Escherichia-Shigella, Klebsiella, and Veillonella, was only observed for the mature body. Our findings demonstrate that P. atrovolvatus exhibits potential benefits on gut health by promoting SCFA production and the growth of beneficial bacteria, with the mature stage demonstrating superior effects compared to the egg stage.


Subject(s)
Fermentation , Fruiting Bodies, Fungal , Gastrointestinal Microbiome , Gastrointestinal Microbiome/drug effects , Fatty Acids, Volatile/metabolism , Agaricales/chemistry , Bacteria/growth & development , Bacteria/metabolism , Dietary Fiber/pharmacology , Dietary Fiber/analysis , Humans
4.
Nutrients ; 16(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39203714

ABSTRACT

Scientific evidence shows that dietary patterns are a key environmental determinant of mental health. Dietary constituents can modify epigenetic patterns and thus the gene expression of relevant genetic variants in various mental health conditions. In the present work, we describe some nutrigenomic effects of dietary fiber, phenolic compounds (plant secondary metabolites), and fatty acids on mental health outcomes, with emphasis on their possible interactions with genetic and epigenetic aspects. Prebiotics, through their effects on the gut microbiota, have been associated with modulation in the neuroendocrine response to stress and the facilitation of the processing of positive emotions. Some of the genetic and epigenetic mechanisms include the serotonin neurotransmitter system (TPH1 gene) and the brain-derived neurotrophic factor (inhibition of histone deacetylases). The consumption of phenolic compounds exerts a positive role in neurocognitive domains. The evidence showing the involvement of genetic and epigenetic factors comes mainly from animal models, highlighting the role of epigenetic mechanisms through miRNAs and methyltransferases as well as the effect on the expression of apoptotic-related genes. Long-chain n-3 fatty acids (EPA and DHA) have been mainly related to psychotic and mood disorders, but the genetic and epigenetic evidence is scarce. Studies on the genetic and epigenetic basis of these interactions need to be promoted to move towards a precision and personalized approach to medicine.


Subject(s)
Dietary Fiber , Epigenesis, Genetic , Fatty Acids , Mental Health , Humans , Dietary Fiber/pharmacology , Animals , Gastrointestinal Microbiome/drug effects , Phenols/pharmacology , Nutrigenomics , Mental Disorders/genetics
5.
Nutrients ; 16(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39203797

ABSTRACT

The gut microbiota are mainly composed of Bacteroidetes and Firmicutes and are crucial for metabolism and immunity. Muribaculaceae are a family of bacteria within the order Bacteroidetes. Muribaculaceae produce short-chain fatty acids via endogenous (mucin glycans) and exogenous polysaccharides (dietary fibres). The family exhibits a cross-feeding relationship with probiotics, such as Bifidobacterium and Lactobacillus. The alleviating effects of a plant-based diet on inflammatory bowel disease, obesity, and type 2 diabetes are associated with an increased abundance of Muribaculaceae, a potential probiotic bacterial family. This study reviews the current findings related to Muribaculaceae and systematically introduces their diversity, metabolism, and function. Additionally, the mechanisms of Muribaculaceae in the alleviation of chronic diseases and the limitations in this field of research are introduced.


Subject(s)
Bacteroidetes , Gastrointestinal Microbiome , Probiotics , Gastrointestinal Microbiome/physiology , Humans , Fatty Acids, Volatile/metabolism , Animals , Diabetes Mellitus, Type 2/microbiology , Dietary Fiber/pharmacology , Obesity/microbiology
6.
Int J Biol Macromol ; 277(Pt 3): 134401, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39097049

ABSTRACT

An imbalance between energy intake and energy expenditure predisposes obesity and its related metabolic diseases. Soluble dietary fiber has been shown to improve metabolic homeostasis mainly via microbiota reshaping. However, the application and metabolic effects of insoluble fiber are less understood. Herein, we employed nanotechnology to design citric acid-crosslinked carboxymethyl cellulose nanofibers (CL-CNF) with a robust capacity of expansion upon swelling. Supplementation with CL-CNF reduced food intake and delayed digestion rate in mice by occupying stomach. Besides, CL-CNF treatment mitigated diet-induced obesity and insulin resistance in mice with enhanced energy expenditure, as well as ameliorated inflammation in adipose tissue, intestine and liver and reduced hepatic steatosis, without any discernible signs of toxicity. Additionally, CL-CNF supplementation resulted in enrichment of probiotics such as Bifidobacterium and decreased in the relative abundances of deleterious microbiota expressing bile salt hydrolase, which led to increased levels of conjugated bile acids and inhibited intestinal FXR signaling to stimulate the release of GLP-1. Taken together, our findings demonstrate that CL-CNF administration protects mice from diet-induced obesity and metabolic dysfunction by reducing food intake, enhancing energy expenditure and remodeling gut microbiota, making it a potential therapeutic strategy against metabolic diseases.


Subject(s)
Energy Metabolism , Gastrointestinal Microbiome , Nanofibers , Obesity , Animals , Nanofibers/chemistry , Obesity/metabolism , Obesity/prevention & control , Mice , Gastrointestinal Microbiome/drug effects , Energy Metabolism/drug effects , Cellulose/pharmacology , Cellulose/chemistry , Male , Insulin Resistance , Mice, Inbred C57BL , Diet, High-Fat/adverse effects , Solubility , Carboxymethylcellulose Sodium/chemistry , Carboxymethylcellulose Sodium/pharmacology , Dietary Fiber/pharmacology
7.
J Agric Food Chem ; 72(29): 16237-16249, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38984620

ABSTRACT

Dietary fiber and polyphenols have been shown to possess antiobesity properties. However, their combined effects need further investigation. This study investigated the individual and combined effects of arabinoxylan oligosaccharides (AXOS) from rice bran and green tea polyphenols (GTP) in high-fat diet-induced obese mice. We found that the combination of AXOS and GTP (A + G) significantly reduced overall fat mass and improved lipid profiles, although the effects were not synergistic. AXOS and GTP regulated lipid metabolism in different tissues and exhibited counteractive effects on gut microbiota. AXOS decreased α diversity and promoted Bifidobacterium, with GTP counteracting these effects. In vitro fermentation confirmed that GTP counteracted AXOS-induced microbiota changes in a dose-dependent manner. This study highlights the potential of tailored combinations of dietary fiber and polyphenols to treat obesity while considering their complex microbial interplay.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Mice, Inbred C57BL , Obesity , Oligosaccharides , Polyphenols , Tea , Xylans , Animals , Xylans/administration & dosage , Xylans/pharmacology , Xylans/metabolism , Polyphenols/pharmacology , Polyphenols/administration & dosage , Polyphenols/chemistry , Gastrointestinal Microbiome/drug effects , Diet, High-Fat/adverse effects , Obesity/metabolism , Obesity/drug therapy , Obesity/microbiology , Obesity/diet therapy , Mice , Oligosaccharides/administration & dosage , Oligosaccharides/pharmacology , Male , Tea/chemistry , Humans , Bacteria/classification , Bacteria/drug effects , Bacteria/isolation & purification , Bacteria/metabolism , Bacteria/genetics , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Plant Extracts/chemistry , Camellia sinensis/chemistry , Dietary Fiber/metabolism , Dietary Fiber/pharmacology , Oryza/chemistry
8.
Mediators Inflamm ; 2024: 6263447, 2024.
Article in English | MEDLINE | ID: mdl-39015676

ABSTRACT

Group 2 innate lymphoid cells (ILC2) strongly modulate COPD pathogenesis. However, the significance of microbiota in ILC2s remains unelucidated. Herein, we investigated the immunomodulatory role of short-chain fatty acids (SCFAs) in regulating ILC2-associated airway inflammation and explores its associated mechanism in COPD. In particular, we assessed the SCFA-mediated regulation of survival, proliferation, and cytokine production in lung sorted ILC2s. To elucidate butyrate action in ILC2-driven inflammatory response in COPD models, we administered butyrate to BALB/c mice via drinking water. We revealed that SCFAs, especially butyrate, derived from dietary fiber fermentation by gut microbiota inhibited pulmonary ILC2 functions and suppressed both IL-13 and IL-5 synthesis by murine ILC2s. Using in vivo and in vitro experimentation, we validated that butyrate significantly ameliorated ILC2-induced inflammation. We further demonstrated that butyrate suppressed ILC2 proliferation and GATA3 expression. Additionally, butyrate potentially utilized histone deacetylase (HDAC) inhibition to enhance NFIL3 promoter acetylation, thereby augmenting its expression, which eventually inhibited cytokine production in ILC2s. Taken together, the aforementioned evidences demonstrated a previously unrecognized role of microbial-derived SCFAs on pulmonary ILC2s in COPD. Moreover, our evidences suggest that metabolomics and gut microbiota modulation may prevent lung inflammation of COPD.


Subject(s)
Butyrates , Dietary Fiber , Lymphocytes , Mice, Inbred BALB C , Pulmonary Disease, Chronic Obstructive , Animals , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Mice , Butyrates/pharmacology , Lymphocytes/metabolism , Dietary Fiber/pharmacology , Dietary Fiber/therapeutic use , Fatty Acids, Volatile/metabolism , Inflammation/metabolism , Gastrointestinal Microbiome , Male , Cytokines/metabolism , Humans , GATA3 Transcription Factor/metabolism
9.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000076

ABSTRACT

The gut microbiota is a diverse bacterial community consisting of approximately 2000 species, predominantly from five phyla: Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. The microbiota's bacterial species create distinct compounds that impact the host's health, including well-known short-chain fatty acids. These are produced through the breakdown of dietary fibers and fermentation of undigested carbohydrates by the intestinal microbiota. The main short-chain fatty acids consist of acetate, propionate, and butyrate. The concentration of butyrate in mammalian intestines varies depending on the diet. Its main functions are use as an energy source, cell differentiation, reduction in the inflammatory process in the intestine, and defense against oxidative stress. It also plays an epigenetic role in histone deacetylases, thus helping to reduce the risk of colon cancer. Finally, butyrate affects the gut-brain axis by crossing the brain-blood barrier, making it crucial to determine the right concentrations for both local and peripheral effects. In recent years, there has been a significant amount of attention given to the role of dietary polyphenols and fibers in promoting human health. Polyphenols and dietary fibers both play crucial roles in protecting human health and can produce butyrate through gut microbiota fermentation. This paper aims to summarize information on the key summits related to the negative correlation between intestinal microbiota diversity and chronic diseases to guide future research on determining the specific activity of butyrate from polyphenols and dietary fibers that can carry out these vital functions.


Subject(s)
Butyrates , Dietary Fiber , Gastrointestinal Microbiome , Polyphenols , Gastrointestinal Microbiome/drug effects , Dietary Fiber/metabolism , Dietary Fiber/pharmacology , Humans , Polyphenols/pharmacology , Butyrates/metabolism , Animals , Fatty Acids, Volatile/metabolism , Fermentation
10.
Nutrients ; 16(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39064648

ABSTRACT

The complex interactions between intestinal microbiota and metabolic disorders are well-documented, with implications for glucose metabolism, energy expenditure, and intestinal permeability. Prebiotics induce beneficial changes in gut microbiota composition in prediabetes, while postbiotics can enhance gut barrier function, complementing each other to improve glucose metabolism and insulin sensitivity. This study investigated the effects of a 12-week dietary fibre (DF) supplement on gut health, metabolic function, and diet. The supplement contained konjac glucomannan (KGM), galacto-oligosaccharides (GOSs), and exopolysaccharides (EPSs) from Bifidobacterium breve. In a randomised, double-blind, placebo-controlled, parallel-group clinical trial, 53 prediabetic volunteers were randomly assigned to either a daily DF supplement (YMETA) or a placebo (cellulose microcrystalline) for 12 weeks, followed by a 4-week follow-up. Measurements included gut microbiota composition, glycated haemoglobin (HbA1c), fasting plasma glucose (FPG), plasma lipids, anthropometry, body composition, blood pressure, and dietary intake. The intervention group showed a significant increase in alpha diversity and butyrate-producing bacteria, with reductions in HbA1c and FPG levels below prediabetes thresholds. No significant changes were observed in the placebo group. This study suggests that manipulating the human gut microbiome through dietary interventions could be a promising therapeutic approach to managing prediabetes and preventing or delaying diabetes.


Subject(s)
Bifidobacterium breve , Dietary Fiber , Gastrointestinal Microbiome , Glycated Hemoglobin , Mannans , Oligosaccharides , Prebiotics , Prediabetic State , Humans , Gastrointestinal Microbiome/drug effects , Double-Blind Method , Prediabetic State/therapy , Prediabetic State/diet therapy , Prebiotics/administration & dosage , Glycated Hemoglobin/metabolism , Male , Female , Oligosaccharides/administration & dosage , Middle Aged , Dietary Fiber/pharmacology , Dietary Fiber/administration & dosage , Adult , Mannans/pharmacology , Blood Glucose/metabolism , Dietary Supplements , Galactose
11.
Nutrients ; 16(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39064683

ABSTRACT

ß-glucans found in cereal grains have been previously demonstrated to improve blood glucose control; however, current understanding points to their high viscosity as the primary mechanism of action. In this work, we present a novel, highly soluble, low-viscosity ß-glucan fiber (HS-BG fiber) and a preclinical dataset that demonstrates its impact on two mechanisms related to the prevention of hyperglycemia. Our results show that HS-BG inhibits the activity of two key proteins involved in glucose metabolism, the α-glucosidase enzyme and the SGLT1 transporter, thereby having the potential to slow starch digestion and subsequent glucose uptake. Furthermore, we demonstrate in a multi-donor fecal fermentation model that HS-BG is metabolized by several different members of the gut microbiome, producing high amounts of short-chain fatty acids (SCFAs), known agonists of GPR43 receptors in the gut related to GLP-1 secretion. The production of SCFAs was verified in the translational gut model, SHIME®. Moreover, HS-BG fiber fermentation produces compounds that restored permeability in disrupted epithelial cells, decreased inflammatory chemokines (CXCL10, MCP-1, and IL-8), and increased anti-inflammatory marker (IL-10), which could improve insulin resistance. Together, these data suggest that the novel HS-BG fiber is a promising new functional ingredient that can be used to modulate postprandial glycemic responses while the high solubility and low viscosity enable easy formulation in both beverage and solid food matrices.


Subject(s)
Blood Glucose , Dietary Fiber , Fatty Acids, Volatile , Gastrointestinal Microbiome , Permeability , Sodium-Glucose Transporter 1 , beta-Glucans , Dietary Fiber/pharmacology , beta-Glucans/pharmacology , Blood Glucose/metabolism , Humans , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Fatty Acids, Volatile/metabolism , Sodium-Glucose Transporter 1/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Fermentation , Solubility , Feces/chemistry , Feces/microbiology , Viscosity , Glycoside Hydrolase Inhibitors/pharmacology , Hyperglycemia/prevention & control , Intestinal Barrier Function
12.
Int J Food Sci Nutr ; 75(6): 571-581, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38982571

ABSTRACT

Fructans are commonly used as dietary fibre supplements for their ability to promote the growth of beneficial gut microbes. However, fructan consumption has been associated with various dosage-dependent side effects. We characterised side effects in an exploratory analysis of a randomised trial in healthy adults (n = 40) who consumed 18 g/day inulin or placebo. We found that individuals weighing more or habitually consuming higher fibre exhibited the best tolerance. Furthermore, we identified associations between gut microbiome composition and host tolerance. Specifically, higher levels of Christensenellaceae R-7 group were associated with gastrointestinal discomfort, and a machine-learning-based approach successfully predicted high levels of flatulence, with [Ruminococcus] torques group and (Oscillospiraceae) UCG-002 sp. identified as key predictive taxa. These data reveal trends that can help guide personalised recommendations for initial inulin dosage. Our results support prior ecological findings indicating that fibre supplementation has the greatest impact on individuals whose baseline fibre intake is lowest.


Subject(s)
Dietary Fiber , Dietary Supplements , Fructans , Gastrointestinal Microbiome , Inulin , Humans , Dietary Fiber/pharmacology , Male , Adult , Female , Gastrointestinal Microbiome/drug effects , Fructans/pharmacology , Inulin/pharmacology , Young Adult , Body Weight , Middle Aged , Flatulence
13.
Int J Biol Macromol ; 275(Pt 1): 133620, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960238

ABSTRACT

Dietary fibers have attracted much attention due to their multiple benefits on gut health. In this work, the protective mechanism of dietary fiber from sweetpotato residues (SRDF) on the high-fat diet (HFD)-induced intestinal barrier injury was investigated using microbiome-metabolomics-based approach. The physicochemical property analysis demonstrated a thermal stability below 200 °C and porous pectin-polysaccharide structure of SRDF with high in vitro functional activities. The biochemical analysis indicated that SRDF significantly ameliorated intestinal barrier function by improving intestinal morphology and permeability and inhibiting inflammatory response. Microbiome analysis demonstrated that SRDF significantly reversed the HFD-induced dysbacteriosis, decreased the ratio of Firmicutes/Bacteroides and enhanced the relative abundance of probiotics, such as Muribaculaceae and Bifidobacteriaceae. Metabolomics analysis showed that SRDF also significantly altered the metabolic profile in the colon, wherein the differential metabolites were mainly involved in amino acid metabolism (especially tryptophan). Pearson correlation coefficient identified the beneficial relationship between intestinal microbiome and metabolome induced by SRDF. The limitation of this study was that the mouse model may not fully replicate the human intestinal responses due to the difference between the standard environmental conditions and natural world. Generally, our results implied the great potential of SRDF as a functional food ingredient.


Subject(s)
Diet, High-Fat , Dietary Fiber , Gastrointestinal Microbiome , Ipomoea batatas , Metabolomics , Animals , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Ipomoea batatas/chemistry , Dietary Fiber/pharmacology , Dietary Fiber/metabolism , Mice , Metabolomics/methods , Male , Metabolome/drug effects , Intestines/pathology , Intestines/microbiology , Intestines/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice, Inbred C57BL
14.
Food Chem ; 459: 140345, 2024 Nov 30.
Article in English | MEDLINE | ID: mdl-38986204

ABSTRACT

Considering the high abundance of bound polyphenols (BP) in whole grain dietary fiber (DF), this study utilized multi-omics approach to evaluate the impact of BP of defatted rice bran insoluble DF (RIDF) in modulating obesity. Mice on high-fat diet were gavage-administered RIDF, BP-removed or formulated RIDF. The results indicated that DF significantly reduced serum total cholesterol, triglycerides, high-density and low-density lipoprotein cholesterol levels. Moreover, hepatic lipid accumulation and damage induced by high-fat diet were significantly ameliorated with DF intervention. The presence of BP increased the abundance of beneficial bacteria g_Akkermansia and g_Butyricicocus, as well as the expression of butyric acid/propionic acid. Furthermore, the expression of hepatic lipids and lipid-like molecules was significantly decreased under the combined intervention of BP and DF, and this was accompanied by alterations in genes related to lipid, sterol, and cholesterol metabolic biological processes. These findings suggest that BP contribute to the anti-obesity effects of DF.


Subject(s)
Anti-Obesity Agents , Diet, High-Fat , Dietary Fiber , Obesity , Oryza , Polyphenols , Animals , Humans , Male , Mice , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Bacteria/genetics , Bacteria/metabolism , Bacteria/classification , Bacteria/isolation & purification , Bacteria/drug effects , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Dietary Fiber/metabolism , Dietary Fiber/analysis , Dietary Fiber/pharmacology , Gastrointestinal Microbiome/drug effects , Lipid Metabolism/drug effects , Liver/metabolism , Liver/drug effects , Mice, Inbred C57BL , Multiomics , Obesity/metabolism , Obesity/drug therapy , Obesity/diet therapy , Obesity/genetics , Oryza/chemistry , Oryza/metabolism , Polyphenols/pharmacology , Polyphenols/chemistry , Polyphenols/administration & dosage , Polyphenols/metabolism , Triglycerides/metabolism
15.
J Nutr Biochem ; 133: 109699, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38972609

ABSTRACT

Dietary strategies rich in fiber have been demonstrated to offer benefits to individuals afflicted with rheumatoid arthritis (RA). However, the specific mechanisms through which a high-fiber diet (HFD) mitigates RA's autoimmunity remain elusive. Herein, we investigate the influence of pectin- and inulin-rich HFD on collagen-induced arthritis (CIA). We establish that HFD significantly alleviates arthritis in CIA mice by regulating the Th17/Treg balance. The rectification of aberrant T cell differentiation by the HFD is linked to the modulation of gut microbiota, augmenting the abundance of butyrate in feces. Concurrently, adding butyrate to the drinking water mirrors the HFD's impact on ameliorating CIA, encompassing arthritis mitigation, regulating intestinal barrier integrity, and restoring the Th17/Treg equilibrium. Butyrate reshapes the metabolic profile of CD4+ T cells in an AMPK-dependent manner. Our research underscores the importance of dietary interventions in rectifying gut microbiota for RA management and offers an explanation of how diet-derived microbial metabolites influence RA's immune-inflammatory-reaction.


Subject(s)
Arthritis, Experimental , CD4-Positive T-Lymphocytes , Gastrointestinal Microbiome , Inulin , Pectins , Animals , Gastrointestinal Microbiome/drug effects , Pectins/pharmacology , Inulin/pharmacology , Inulin/administration & dosage , Arthritis, Experimental/diet therapy , Arthritis, Experimental/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Male , Mice , Dietary Supplements , Th17 Cells/immunology , Th17 Cells/metabolism , Mice, Inbred DBA , Dietary Fiber/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Arthritis, Rheumatoid/diet therapy , Arthritis, Rheumatoid/immunology , Butyrates/metabolism
16.
Eur J Clin Nutr ; 78(9): 757-764, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39003347

ABSTRACT

BACKGROUND: Mycoprotein is a high-fibre food previously shown to reduce postprandial glucose concentrations when ingested within a mixed-meal. We applied a dual stable isotope tracer approach to determine whether this is due to a reduced rate of appearance of glucose, in participants of ranging BMI. METHODS: Twenty-four adults (F = 8, BMI 30 ± 6 kg·m-2) attended 2 trials in a double-blind, randomised, cross-over design. Participants ingested two energy and macronutrient matched milk-based drinks (enriched with 1000 mg [U-13C6] glucose in a subset of 12 participants), containing 50 g glucose and either 0 (CON) or 20 g (MYC) mycoprotein. A primed continuous intravenous infusion of D-[6,6-2H2] glucose determined plasma glucose kinetics over 6 h. Postprandial time-course, and AUC, of glucose and insulin concentration, rate of disappearance (RdT) and appearance of exogenous (RaEx), endogenous (EGP), and total (RaT) plasma glucose were assessed using two- and one-way ANOVA. RESULTS: Drink ingestion increased blood glucose and serum insulin concentrations (P < 0.05) and were comparable between conditions (P > 0.05). Both RaT and RdT were higher with MYC compared with CON over 6 h (mean 6 h glucose appearance and disappearance increased by 5 and 9%, respectively, P < 0.05). RaEx was not affected by MYC ingestion over 6 h (P > 0.05). The mean contribution of EGP to total glucose appearance was 15% greater with MYC, with a trend towards significance (P = 0.05). There was no relationship between BMI and the response to MYC ingestion for any of the variables (P < 0.05). CONCLUSION: The ingestion of mycoprotein within a mixed-meal impacted postprandial glucose kinetics, but not blood glucose or serum insulin concentrations, in individuals of ranging BMI. CLINICAL TRIAL REGISTRY NUMBER AND WEBSITE: This trial was registered at clinicaltrials.gov as NCT04084639 and can be accessed at https://clinicaltrials.gov/ct2/show/NCT04084639 .


Subject(s)
Blood Glucose , Cross-Over Studies , Postprandial Period , Humans , Blood Glucose/metabolism , Male , Female , Adult , Double-Blind Method , Meals , Insulin/blood , Middle Aged , Dietary Fiber/administration & dosage , Dietary Fiber/pharmacology , Young Adult , Kinetics
17.
Nutrients ; 16(14)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39064788

ABSTRACT

Bifidobacterium animalis subsp. lactis GCL2505 in combination with inulin has been shown to have several health benefits, including an improvement in the intestinal microbiota and a reduction in human visceral fat. Previous studies have suggested that the visceral fat reduction of GCL2505 and inulin may be achieved by improving daily energy expenditure. This parallel, placebo-controlled, randomized, double-blind study was conducted to evaluate the effects of GCL2505 and inulin on resting energy expenditure (REE) in overweight or mildly obese Japanese adults (n = 44). Participants ingested 1 × 1010 colony forming units of GCL2505 and 5.0 g of inulin daily for 4 weeks. REE score at week 4 was set as the primary endpoint. At week 4, the REE score of the GCL2505 and inulin group was significantly higher than that of the placebo group, with a difference of 84.4 kcal/day. In addition, fecal bifidobacteria counts were significantly increased in the GCL2505 and inulin group. Our results indicated that the intake of GCL2505 and inulin improves energy balance, which is known to be a major factor of obesity, by modulating the microbiota in the gut. This is the first report to demonstrate the effects of probiotics and dietary fiber on REE in humans.


Subject(s)
Dietary Fiber , Feces , Gastrointestinal Microbiome , Inulin , Obesity , Probiotics , Humans , Double-Blind Method , Male , Female , Probiotics/administration & dosage , Dietary Fiber/administration & dosage , Dietary Fiber/pharmacology , Middle Aged , Adult , Inulin/administration & dosage , Inulin/pharmacology , Feces/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Microbiome/drug effects , Obesity/microbiology , Obesity/diet therapy , Energy Metabolism , Bifidobacterium , Overweight/microbiology , Overweight/diet therapy , Bifidobacterium animalis , Japan , Basal Metabolism/drug effects
18.
Nat Commun ; 15(1): 5413, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926424

ABSTRACT

Diet composition impacts metabolic health and is now recognized to shape the immune system, especially in the intestinal tract. Nutritional imbalance and increased caloric intake are induced by high-fat diet (HFD) in which lipids are enriched at the expense of dietary fibers. Such nutritional challenge alters glucose homeostasis as well as intestinal immunity. Here, we observed that short-term HFD induced dysbiosis, glucose intolerance and decreased intestinal RORγt+ CD4 T cells, including peripherally-induced Tregs and IL17-producing (Th17) T cells. However, supplementation of HFD-fed male mice with the fermentable dietary fiber fructooligosaccharides (FOS) was sufficient to maintain RORγt+ CD4 T cell subsets and microbial species known to induce them, alongside having a beneficial impact on glucose tolerance. FOS-mediated normalization of Th17 cells and amelioration of glucose handling required the cDC2 dendritic cell subset in HFD-fed animals, while IL-17 neutralization limited FOS impact on glucose tolerance. Overall, we uncover a pivotal role of cDC2 in the control of the immune and metabolic effects of FOS in the context of HFD feeding.


Subject(s)
Dendritic Cells , Diet, High-Fat , Homeostasis , Mice, Inbred C57BL , Oligosaccharides , Animals , Oligosaccharides/pharmacology , Diet, High-Fat/adverse effects , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Male , Mice , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/drug effects , Glucose/metabolism , Interleukin-17/metabolism , Dietary Fiber/pharmacology , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Dysbiosis/immunology , Gastrointestinal Microbiome/drug effects
19.
Physiol Rep ; 12(11): e16047, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38837588

ABSTRACT

Acetate is a short-chain fatty acid (SCFA) that is produced by microbiota in the intestinal tract. It is an important nutrient for the intestinal epithelium, but also has a high plasma concentration and is used in the various tissues. Acetate is involved in endurance exercise, but its role in resistance exercise remains unclear. To investigate this, mice were administered either multiple antibiotics with and without oral acetate supplementation or fed a low-fiber diet. Antibiotic treatment for 2 weeks significantly reduced grip strength and the cross-sectional area (CSA) of muscle fiber compared with the control group. Intestinal concentrations of SCFAs were reduced in the antibiotic-treated group. Oral administration of acetate with antibiotics prevented antibiotic-induced weakness of skeletal muscle and reduced CSA of muscle fiber. Similarly, a low-fiber diet for 1 year significantly reduced the CSA of muscle fiber and fecal and plasma acetate concentrations. To investigate the role of acetate as an energy source, acetyl-CoA synthase 2 knockout mice were used. These mice had a shorter lifespan, reduced skeletal muscle mass and smaller CSA of muscle fiber than their wild type littermates. In conclusion, acetate derived from the intestinal microbiome can contribute to maintaining skeletal muscle performance.


Subject(s)
Acetates , Gastrointestinal Microbiome , Mice, Inbred C57BL , Muscle Strength , Muscle, Skeletal , Animals , Acetates/pharmacology , Acetates/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Mice , Male , Muscle Strength/drug effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Mice, Knockout , Anti-Bacterial Agents/pharmacology , Fatty Acids, Volatile/metabolism , Dietary Fiber/pharmacology , Dietary Fiber/metabolism
20.
Nutrients ; 16(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38892504

ABSTRACT

Arabic gum, a high molecular weight heteropolysaccharide, is a promising prebiotic candidate as its fermentation occurs more distally in the colon, which is the region where most chronic colonic diseases originate. Baobab fiber could be complementary due to its relatively simple structure, facilitating breakdown in the proximal colon. Therefore, the current study aimed to gain insight into how the human gut microbiota was affected in response to long-term baobab fiber and Arabic gum supplementation when tested individually or as a combination of both, allowing the identification of potential complementary and/or synergetic effects. The validated Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), an in vitro gut model simulating the entire human gastrointestinal tract, was used. The microbial metabolic activity was examined, and quantitative 16S-targeted Illumina sequencing was used to monitor the gut microbial composition. Moreover, the effect on the gut microbial metabolome was quantitatively analyzed. Repeated administration of baobab fiber, Arabic gum, and their combination had a significant effect on the metabolic activity, diversity index, and community composition of the microbiome present in the simulated proximal and distal colon with specific impacts on Bifidobacteriaceae and Faecalibacterium prausnitzii. Despite the lower dosage strategy (2.5 g/day), co-supplementation of both compounds resulted in some specific synergistic prebiotic effects, including a biological activity throughout the entire colon, SCFA synthesis including a synergy on propionate, specifically increasing abundance of Akkermansiaceae and Christensenellaceae in the distal colon region, and enhancing levels of spermidine and other metabolites of interest (such as serotonin and ProBetaine).


Subject(s)
Dietary Fiber , Gastrointestinal Microbiome , Gum Arabic , Prebiotics , Humans , Gastrointestinal Microbiome/drug effects , Gum Arabic/pharmacology , Dietary Fiber/pharmacology , Dietary Supplements , Colon/microbiology , Colon/metabolism , Colon/drug effects , Fermentation , Bacteria/drug effects , Bacteria/classification
SELECTION OF CITATIONS
SEARCH DETAIL