Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.059
Filter
1.
Food Chem ; 462: 141010, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39217745

ABSTRACT

In this study, we investigated how different proportions blends of Rhamnogalacturonan-I pectic polysaccharides and hesperidin impact the gut microbiota and metabolites using an in vitro simulated digestion and fermentation model. The results indicated that both of them could modulate the gut microbiota and produce beneficial metabolites. However, their blends in particular proportions (such as 1:1) exhibited remarkable synergistic effects on modulating the intestinal microenvironment, surpassing the effects observed with individual components. Specifically, these blends could benefit the host by increasing short-chain fatty acids production (such as acetate), improving hesperidin bioavailability, producing more metabolites (such as hesperetin, phenolic acids), and promoting the growth of beneficial bacteria. This synergistic and additive effect was inseparable from the role of gut microbiota. Certain beneficial bacteria, such as Blautia, Faecalibacterium, and Prevotella, exhibited strong preferences for those blends, thereby contributing to host health through participating in carbohydrate and flavonoid metabolism.


Subject(s)
Bacteria , Gastrointestinal Microbiome , Hesperidin , Pectins , Hesperidin/pharmacology , Hesperidin/metabolism , Gastrointestinal Microbiome/drug effects , Bacteria/metabolism , Bacteria/genetics , Bacteria/drug effects , Bacteria/classification , Bacteria/isolation & purification , Humans , Pectins/metabolism , Pectins/chemistry , Pectins/pharmacology , Fermentation , Polysaccharides/pharmacology , Polysaccharides/metabolism , Polysaccharides/chemistry , Fatty Acids, Volatile/metabolism , Digestion , Models, Biological
2.
J Environ Sci (China) ; 147: 414-423, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39003059

ABSTRACT

The anaerobic acid production experiments were conducted with the pretreated kitchen waste under pH adjustment. The results showed that pH 8 was considered to be the most suitable condition for acid production, especially for the formation of acetic acid and propionic acid. The average value of total volatile fatty acid at pH 8 was 8814 mg COD/L, 1.5 times of that under blank condition. The average yield of acetic acid and propionic acid was 3302 mg COD/L and 2891 mg COD/L, respectively. The activities of key functional enzymes such as phosphotransacetylase, acetokinase, oxaloacetate transcarboxylase and succinyl-coA transferase were all enhanced. To further explore the regulatory mechanisms within the system, the distribution of microorganisms at different levels in the fermentation system was obtained by microbial sequencing, results indicating that the relative abundances of Clostridiales, Bacteroidales, Chloroflexi, Clostridium, Bacteroidetes and Propionibacteriales, which were great contributors for the hydrolysis and acidification, increased rapidly at pH 8 compared with the blank group. Besides, the proportion of genes encoding key enzymes was generally increased, which further verified the mechanism of hydrolytic acidification and acetic acid production of organic matter under pH regulation.


Subject(s)
Fatty Acids, Volatile , Hydrogen-Ion Concentration , Fatty Acids, Volatile/metabolism , Fermentation , Acetic Acid/metabolism , Bioreactors
3.
Front Cell Infect Microbiol ; 14: 1434687, 2024.
Article in English | MEDLINE | ID: mdl-39372498

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a chronic lung disease that affects premature infants and leads to long-term pulmonary complications. The pathogenesis of BPD has not been fully elucidated yet. In recent years, the microbiome and its metabolites, especially short-chain fatty acids (SCFAs), in the gut and lungs have been demonstrated to be involved in the development and progression of the disease. This review aims to summarize the current knowledge on the potential involvement of the microbiome and SCFAs, especially the latter, in the development and progression of BPD. First, we introduce the gut-lung axis, the production and functions of SCFAs, and the role of SCFAs in lung health and diseases. We then discuss the evidence supporting the involvement of the microbiome and SCFAs in BPD. Finally, we elaborate on the potential mechanisms of the microbiome and SCFAs in BPD, including immune modulation, epigenetic regulation, enhancement of barrier function, and modulation of surfactant production and the gut microbiome. This review could advance our understanding of the microbiome and SCFAs in the pathogenesis of BPD, which also helps identify new therapeutic targets and facilitate new drug development.


Subject(s)
Bronchopulmonary Dysplasia , Fatty Acids, Volatile , Gastrointestinal Microbiome , Lung , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/microbiology , Humans , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Lung/microbiology , Lung/metabolism , Lung/pathology , Microbiota , Infant, Premature , Infant, Newborn , Animals , Epigenesis, Genetic
4.
Anim Sci J ; 95(1): e13997, 2024.
Article in English | MEDLINE | ID: mdl-39362838

ABSTRACT

This study investigated the impact of different levels of coated cysteamine hydrochloride (CSH) supplementation on ruminal fermentation, nutrient digestibility, and blood metabolites in Charolais cross bulls. Twelve bulls were allotted to three feeding treatments in a replicated 3 × 3 Latin square design: 0% CSH (control), 0.5% CSH, and 1.0% CSH in concentrate. Animals were fed concentrate at 1.5% of body weight. Dry matter intake (DMI) and DMI as a percentage of body weight showed no significant differences among treatments (p > 0.10). Nutrient digestibility was consistent across treatments, except for a slight decrease in NDF digestibility with 1% CSH (p = 0.07). Ruminal pH, ammonia nitrogen, volatile fatty acid (VFA) proportions, and total VFA concentration were similar among treatments (p > 0.05). Total bacteria, fungal zoospores, and protozoa populations in the rumen did not vary significantly (p > 0.05). Blood glucose and triglyceride concentrations remained stable (p > 0.05), while blood urea nitrogen (BUN) levels were higher in CSH-supplemented groups (p < 0.05). In conclusion, incorporating CSH levels ranging from 0.5% to 1.0% into the diet did not adversely affect feed intake, ruminal fermentation, or microbial populations. Additionally, 1.0% CSH improved BUN concentration in growing Charolais cross bulls.


Subject(s)
Animal Feed , Blood Urea Nitrogen , Cysteamine , Diet , Dietary Supplements , Digestion , Eating , Fermentation , Rumen , Animals , Cattle/metabolism , Cattle/growth & development , Rumen/metabolism , Rumen/microbiology , Male , Digestion/drug effects , Cysteamine/administration & dosage , Diet/veterinary , Animal Nutritional Physiological Phenomena , Blood Glucose/metabolism , Fatty Acids, Volatile/metabolism , Triglycerides/blood
5.
Gut Microbes ; 16(1): 2409207, 2024.
Article in English | MEDLINE | ID: mdl-39360560

ABSTRACT

BACKGROUND: Despite achieving endoscopic remission, over 20% of inflammatory bowel disease (IBD) patients experience chronic abdominal pain. Visceral pain and the microbiome exhibit sex-dependent interactions, while visceral pain in IBD shows a sex bias. Our aim was to evaluate whether post-inflammatory microbial perturbations contribute to visceral hypersensitivity in a sex-dependent manner. METHODS: Males, cycling females, ovariectomized, and sham-operated females were given dextran sodium sulfate to induce colitis and allowed to recover. Germ-free recipients received sex-appropriate and cross-sex fecal microbial transplants (FMT) from post-inflammatory donor mice. Visceral sensitivity was assessed by recording visceromotor responses to colorectal distention. The composition of the microbiota was evaluated via 16S rRNA gene V4 amplicon sequencing, while the metabolome was assessed using targeted (short chain fatty acids - SCFA) and semi-targeted mass spectrometry. RESULTS: Post-inflammatory cycling females developed visceral hyperalgesia when compared to males. This effect was reversed by ovariectomy. Both post-inflammatory males and females exhibited increased SCFA-producing species, but only males had elevated fecal SCFA content. FMT from post-inflammatory females transferred visceral hyperalgesia to both males and females, while FMT from post-inflammatory males could only transfer visceral hyperalgesia to males. CONCLUSIONS: Female sex, hormonal status as well as the gut microbiota play a role in pain modulation. Our data highlight the importance of considering biological sex in the evaluation of visceral pain.


Subject(s)
Colitis , Dysbiosis , Gastrointestinal Microbiome , Visceral Pain , Male , Female , Animals , Dysbiosis/microbiology , Visceral Pain/microbiology , Visceral Pain/physiopathology , Visceral Pain/metabolism , Colitis/microbiology , Mice , Mice, Inbred C57BL , Fecal Microbiota Transplantation , Sex Factors , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/metabolism , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Dextran Sulfate , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Chronic Pain/microbiology , Chronic Pain/physiopathology , Inflammation/microbiology , Hyperalgesia/microbiology
6.
Front Endocrinol (Lausanne) ; 15: 1392418, 2024.
Article in English | MEDLINE | ID: mdl-39363899

ABSTRACT

Bone is a dynamic tissue that is constantly remodeled throughout adult life. Recently, it has been shown that bone turnover decreases shortly after food consumption. This process has been linked to the fermentation of non-digestible food ingredients such as inulin by gut microbes, which results in the production of the short-chain fatty acids (SCFAs) acetate, propionate and butyrate. SCFAs exert various metabolic functions, which in part can be explained by activation of G protein-coupled receptors (Gpr) 41 and 43. However, the potential relevance of a SCFA-Gpr41/43 signaling axis for bone metabolism has not been established. The aim of our study is to investigate the role of Gpr41/43 in bone metabolism and osteogenic differentiation of mesenchymal stem cells. For this purpose, we analyzed the skeletal phenotype of wild type controls (WT) and Gpr41/43 double knockout (Gpr41/43 dKO) mice fed either a chow or an inulin-enriched diet. In addition, we isolated bone marrow derived mesenchymal stem cells from WT and Gpr41/43 dKO mice and differentiated them into osteoblasts in the absence or presence of acetate. MicroCT scanning of femoral bones of Gpr41/43 dKO mice revealed a significant increase of trabecular bone volume and trabecular compared to WT controls. Treatment of WT bone marrow-derived osteoblasts with acetate resulted in decreased mineralization and substantial downregulation of bone formation markers such as Phex, Ptgs2 and Col1a1. Notably, this effect was strongly attenuated in differentiated osteoblasts lacking Gpr41/43. Inversely, acetate supplementation resulted in higher levels of adipocyte marker genes including Pparg, Lpl and Adipoq in bone marrow-derived cells from WT mice, an effect blunted in differentiated cells isolated from Gpr41/43 dKO mice. Overall, these data indicate that acetate regulates bone architecture via SCFA-Gpr41/43 signaling by modulating the osteogenic versus adipogenic differentiation of mesenchymal stem cells.


Subject(s)
Adipogenesis , Cell Differentiation , Mesenchymal Stem Cells , Mice, Knockout , Osteogenesis , Receptors, G-Protein-Coupled , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Adipogenesis/physiology , Osteogenesis/physiology , Fatty Acids, Volatile/metabolism , Mice, Inbred C57BL , Bone Density , Male , Osteoblasts/metabolism , Osteoblasts/cytology , Cells, Cultured
7.
Front Cell Infect Microbiol ; 14: 1436477, 2024.
Article in English | MEDLINE | ID: mdl-39355267

ABSTRACT

Aims: Hyperglycemia is one of the adverse effects of tacrolimus (TAC), but the underlying mechanism is not fully identified. We used multi-omics analysis to evaluate the changes in the gut microbiota and metabolic profile of rats with TAC-induced diabetes. Methods: To establish a diabetic animal model, Sprague Dawley rats were divided randomly into two groups. Those in the TAC group received intraperitoneal injections of TAC (3 mg/kg) for 8 weeks, and those in the CON group served as the control. 16S rRNA sequencing was used to analyze fecal microbiota. The metabolites of the two groups were detected and analyzed by nontargeted and targeted metabolomics, including amino acids (AAs), bile acids (BAs), and short-chain fatty acids (SCFAs). Results: The rats treated with TAC exhibited hyperglycemia as well as changes in the gut microbiota and metabolites. Specifically, their gut microbiota had significantly higher abundances of Escherichia-Shigella, Enterococcus, and Allobaculum, and significantly lower abundances of Ruminococcus, Akkermansia, and Roseburia. In addition, they had significantly reduced serum levels of AAs including asparagine, aspartic acid, glutamic acid, and methionine. With respect to BAs, they had significantly higher serum levels of taurocholic acid (TCA), and glycochenodeoxycholic acid (GCDCA), but significantly lower levels of taurodeoxycholic acid (TDCA) and tauroursodeoxycholic acid (TUDCA). There were no differences in the levels of SCFAs between the two groups. Correlations existed among glucose metabolism indexes (fasting blood glucose and fasting insulin), gut microbiota (Ruminococcus and Akkermansia), and metabolites (glutamic acid, hydroxyproline, GCDCA, TDCA, and TUDCA). Conclusions: Both AAs and BAs may play crucial roles as signaling molecules in the regulation of TAC-induced diabetes.


Subject(s)
Amino Acids , Feces , Gastrointestinal Microbiome , Metabolomics , RNA, Ribosomal, 16S , Rats, Sprague-Dawley , Tacrolimus , Animals , Gastrointestinal Microbiome/drug effects , Tacrolimus/pharmacology , Rats , Male , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Amino Acids/metabolism , Amino Acids/blood , Diabetes Mellitus, Experimental/metabolism , Bile Acids and Salts/metabolism , Fatty Acids, Volatile/metabolism , Metabolome/drug effects , Disease Models, Animal , Hyperglycemia/metabolism , Bacteria/classification , Bacteria/drug effects , Bacteria/metabolism , Bacteria/genetics , Blood Glucose/metabolism , Immunosuppressive Agents
8.
Front Immunol ; 15: 1456030, 2024.
Article in English | MEDLINE | ID: mdl-39351241

ABSTRACT

The human microbiome has recently emerged as a focal point in cancer research, specifically in anti-tumor immunity, immunotherapy, and chemotherapy. This review explores microbial-derived metabolites, emphasizing their crucial roles in shaping fundamental aspects of cancer treatment. Metabolites such as short-chain fatty acids (SCFAs), Trimethylamine N-Oxide (TMAO), and Tryptophan Metabolites take the spotlight, underscoring their diverse origins and functions and their profound impact on the host immune system. The focus is on SCFAs' remarkable ability to modulate immune responses, reduce inflammation, and enhance anti-tumor immunity within the intricate tumor microenvironment (TME). The review critically evaluates TMAO, intricately tied to dietary choices and gut microbiota composition, assessing its implications for cancer susceptibility, progression, and immunosuppression. Additionally, the involvement of tryptophan and other amino acid metabolites in shaping immune responses is discussed, highlighting their influence on immune checkpoints, immunosuppression, and immunotherapy effectiveness. The examination extends to their dynamic interaction with chemotherapy, emphasizing the potential of microbial-derived metabolites to alter treatment protocols and optimize outcomes for cancer patients. A comprehensive understanding of their role in cancer therapy is attained by exploring their impacts on drug metabolism, therapeutic responses, and resistance development. In conclusion, this review underscores the pivotal contributions of microbial-derived metabolites in regulating anti-tumor immunity, immunotherapy responses, and chemotherapy outcomes. By illuminating the intricate interactions between these metabolites and cancer therapy, the article enhances our understanding of cancer biology, paving the way for the development of more effective treatment options in the ongoing battle against cancer.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Immunotherapy , Neoplasms , Tryptophan , Tumor Microenvironment , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Neoplasms/drug therapy , Immunotherapy/methods , Gastrointestinal Microbiome/immunology , Tumor Microenvironment/immunology , Animals , Fatty Acids, Volatile/metabolism , Tryptophan/metabolism , Methylamines/metabolism , Methylamines/immunology , Antineoplastic Agents/therapeutic use
9.
Nutrients ; 16(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39275152

ABSTRACT

Wild rice (WLD) attenuated hyperglycemia, hyperlipidemia and chronic inflammation in mice receiving a high-fat diet (HFD) versus white rice (WHR), but the underlying mechanism is not well understood. We examined the influence of HFD + WLD on gut microbiota, short chain fatty acids (SCFAs) and the correlation with metabolic or inflammatory markers in mice versus HFD + WHR. C57BL/6J mice received HFD + 26 g weight (wt) % WHR or WLD or 13 g wt% WHR + 13 g wt% WLD (WTWD) for 12 weeks. Plasma levels of glucose, cholesterol and triglycerides, insulin resistance and inflammatory markers after overnight fasting were lower, and the abundances of fecal Lactobacillus gasseri and propionic acid were higher in HFD + WLD-fed mice than in HFD + WHR-fed mice. The anti-inflammatory effects of HFD + WTWD were weaker than HFD + WLD but were greater than those in HFD + WHR-fed mice. Abundances of fecal Lactobacillus gasseri and propionic acid in mice receiving HFD + WLD were higher than those in mice fed with HFD + WHR. The abundances of fecal L. gasseri and propionic acid negatively correlated with metabolic and inflammatory markers. The findings of the present study suggest that WLD attenuated metabolic and inflammatory disorders in mice on HFD. Interactions between WLD components and gut microbiota may upregulate fecal SCFAs, and the latter may be attributed to the benefits of WLD on metabolism and inflammation in mice on HFD.


Subject(s)
Biomarkers , Diet, High-Fat , Dysbiosis , Fatty Acids, Volatile , Feces , Gastrointestinal Microbiome , Mice, Inbred C57BL , Oryza , Animals , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Fatty Acids, Volatile/metabolism , Male , Mice , Feces/microbiology , Feces/chemistry , Biomarkers/blood , Inflammation , Blood Glucose/metabolism , Insulin Resistance , Triglycerides/blood , Propionates
10.
Nutrients ; 16(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39275251

ABSTRACT

Inulin is a plant polysaccharide which, due to its chemical structure, is not digestible by human gut enzymes but by some bacteria of the human microbiota, acting as a prebiotic. Consequently, inulin consumption has been associated with changes in the composition of the intestinal microbiota related to an improvement of the metabolic state, counteracting different obesity-related disturbances. However, the specific mechanisms of action, including bacterial changes, are not exactly known. Here, a bibliographic review was carried out to study the main effects of inulin on human metabolic health, with a special focus on the mechanisms of action of this prebiotic. Inulin supplementation contributes to body weight and BMI control, reduces blood glucose levels, improves insulin sensitivity, and reduces inflammation markers, mainly through the selective favoring of short-chain fatty acid (SCFA)-producer species from the genera Bifidobacterium and Anaerostipes. These SCFAs have been shown to ameliorate glucose metabolism and decrease hepatic lipogenesis, reduce inflammation, modulate immune activity, and improve anthropometric parameters such as body weight or BMI. In conclusion, the studies collected suggest that inulin intake produces positive metabolic effects through the improvement of the intestinal microbiota and through the metabolites produced by its fermentation.


Subject(s)
Gastrointestinal Microbiome , Inulin , Prebiotics , Humans , Inulin/pharmacology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Fatty Acids, Volatile/metabolism , Obesity/metabolism , Obesity/microbiology , Body Mass Index , Blood Glucose/metabolism , Body Weight/drug effects , Insulin Resistance
11.
Nutrients ; 16(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39275259

ABSTRACT

Traditional fermented foods are known to offer cardiovascular health benefits. However, the potential of fermented Chinese chives (FCC) in reducing coronary heart disease (CHD) remains unclear. This study employed anaerobic fermentation to investigate Lactiplantibacillus plantarum (L. plantarum) P470 from FCC. The results indicated that L. plantarum P470 enhanced hydroxyl radical scavenging and exhibited anti-inflammatory effects on RAW264.7 macrophages in the fecal fermentation supernatant of CHD patients. These effects were attributed to the modulation of gut microbiota and metabolites, including short-chain fatty acids (SCFAs). Specifically, L. plantarum P470 increased the abundance of Bacteroides and Lactobacillus while decreasing Escherichia-Shigella, Enterobacter, Veillonella, Eggerthella, and Helicobacter in CHD patient fecal samples. Furthermore, L. plantarum P470 regulated the biosynthesis of unsaturated fatty acids and linoleic acid metabolism. These findings suggest that L. plantarum P470 from FCC can improve the fecal physiological status in patients with CHD by modulating intestinal microbiota, promoting SCFA production, and regulating lipid metabolism.


Subject(s)
Coronary Disease , Fatty Acids, Volatile , Feces , Fermented Foods , Gastrointestinal Microbiome , Lactobacillus plantarum , Humans , Feces/microbiology , Coronary Disease/microbiology , Mice , Animals , Fermented Foods/microbiology , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Male , Fermentation , Female , Middle Aged , RAW 264.7 Cells , Aged , Probiotics/pharmacology
12.
Nutrients ; 16(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39275295

ABSTRACT

Rice bran, which is abundant in dietary fiber and phytochemicals, provides multiple health benefits. Nonetheless, its effects on neuroinflammation and gut microbiota in postmenopausal conditions are still not well understood. This study investigated the effects of rice bran and/or tea seed oil supplementation in d-galactose-injected ovariectomized (OVX) old mice fed a fructose drink. The combination of d-galactose injection, ovariectomy, and fructose drink administration creates a comprehensive model that simulates aging in females under multiple metabolic stressors, including oxidative stress, estrogen deficiency, and high-sugar diets, and allows the study of their combined impact on metabolic disorders and related diseases. Eight-week-old and 6-8-month-old female C57BL/6 mice were used. The mice were divided into six groups: a sham + young mice, a sham + old mice, an OVX + soybean oil, an OVX + soybean oil with rice bran, an OVX + tea seed oil (TO), and an OVX + TO with rice bran diet group. The OVX groups were subcutaneously injected with d-galactose (100 mg/kg/day) and received a 15% (v/v) fructose drink. The rice bran and tea seed oil supplementation formed 10% of the diet (w/w). The results showed that the rice bran with TO diet increased the number of short-chain fatty acid (SCFA)-producing Clostridia and reduced the number of endotoxin-producing Tannerellaceae, which mitigated imbalances in the gut-liver-brain axis. Rice bran supplementation reduced the relative weight of the liver, levels of hepatic triglycerides and total cholesterol; aspartate transaminase and alanine aminotransferase activity; brain levels of proinflammatory cytokines, including interleukin-1ß and tumor necrosis factor-α; and plasma 8-hydroxy-2-deoxyguanosine. This study concludes that rice bran inhibits hepatic fat accumulation, which mitigates peripheral metaflammation and oxidative damage and reduces neuroinflammation in the brain.


Subject(s)
Fructose , Gastrointestinal Microbiome , Mice, Inbred C57BL , Oryza , Ovariectomy , Animals , Gastrointestinal Microbiome/drug effects , Female , Mice , Neuroinflammatory Diseases , Dietary Fiber/pharmacology , Dietary Fiber/administration & dosage , Fatty Acids, Volatile/metabolism , Liver/metabolism , Liver/drug effects , Galactose , Brain/metabolism , Brain/drug effects
13.
J Food Sci ; 89(10): 6465-6480, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39289810

ABSTRACT

Goji berry (Lycium barbarum L.) is a nutrient-rich fruit and has received enormous interest for its health benefits. The beneficial effects of goji berry are linked to the absorption of bioactive compounds within the gastrointestinal digestion process and colon fermentation. Nonetheless, how certain bioactive compounds were released, and metabolism changed of the consumption of whole goji berries were still unclear. Therefore, the present study aimed to evaluate the digestion characteristics of key bioactive compounds in whole goji berries with an in vitro digestion model, and the effects of whole goji berries on the structure of gut microbiota were also investigated. Results showed that a significant release of carbohydrates during the digestion process, peaking within the first 15 min of the intestinal phase (421.4 ± 5.82 mg GE/g, dry weight, respectively), was observed, and the phenolic release reached the highest in the first 15 min of the gastric phase. Meanwhile, the bioaccessibilities of phenolic compounds and carbohydrates were determined to be 63.87% and 80.40%, respectively, after intestinal digestion. In addition, the undigested fractions of goji berries could be further fermented to produce short-chain fatty acids, which decreased the colon pH value (from 7.38 to 6.71) as well as the Firmicutes/Bacteroidetes ratio. Moreover, the goji berries regulated the composition of gut microbiota by promoting beneficial bacteria such as Bacteroides, Parabacteroides, and Paraclostridium, whereas inhibiting the proliferation of harmful bacteria (e.g., Fusobacterium). Our results indicated that the goji berry exhibited significant bioactivity during the digestion and fermentation stage and might provide some new insights into the utilization of goji berries in healthy food processing.


Subject(s)
Digestion , Fatty Acids, Volatile , Fermentation , Fruit , Gastrointestinal Microbiome , Lycium , Gastrointestinal Microbiome/physiology , Fruit/chemistry , Lycium/chemistry , Humans , Fatty Acids, Volatile/metabolism , Bacteria/classification , Bacteria/metabolism , Phenols/analysis , Phenols/metabolism
14.
Cell Biochem Funct ; 42(7): e4108, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39228159

ABSTRACT

Short-chain fatty acids (SCFAs) are essential molecules produced by gut bacteria that fuel intestinal cells and may also influence overall health. An imbalance of SCFAs can result in various acute and chronic diseases, including diabetes, obesity and colorectal cancer (CRC). This review delves into the multifaceted roles of SCFAs, including a brief discussion on their source and various gut-residing bacteria. Primary techniques used for detection of SCFAs, including gas chromatography, high-performance gas chromatography, nuclear magnetic resonance and capillary electrophoresis are also discussed through this article. This review study also compiles various synthesis pathways of SCFAs from diverse substrates such as sugar, acetone, ethanol and amino acids. The different pathways through which SCFAs enter cells for immune response regulation are also highlighted. A major emphasis is the discussion on diseases associated with SCFA dysregulation, such as anaemia, brain development, CRC, depression, obesity and diabetes. This includes exploring the relationship between SCFA levels across ethnicities and their connection with blood pressure and CRC. In conclusion, this review highlights the critical role of SCFAs in maintaining gut health and their implications in various diseases, emphasizing the need for further research on SCFA detection, synthesis and their potential as diagnostic biomarkers. Future studies of SCFAs will pave the way for the development of novel diagnostic tools and therapeutic strategies for optimizing gut health and preventing diseases associated with SCFA dysregulation.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Humans , Fatty Acids, Volatile/metabolism , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Obesity/metabolism
15.
Carbohydr Polym ; 344: 122527, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39218534

ABSTRACT

The root of Millettia pulchra (YLS) has been traditionally used as a folk medicine for the treatment of depression and insomnia in the Zhuang nationality of China, and its polysaccharides have potential antidepressant effect. In this study, a novel homogeneous polysaccharide (YLP-1) was purified from the crude polysaccharides of YLS, and it is mainly composed of glucose, arabinose and mannose with molar ratio of 87.25%, 10.77%, and 1.98%, respectively. YLP-1 is a novel α-glucan with the backbone of 1,4-Glcp and branched at C6 of 1,4,6-Glcp to combine 1,4-Manp and 1,5-Araf. The microstructure of YLP-1 displayed a uniform ellipsoidal-like chain morphology and dispersed uniformly in solution. YLP-1 effectively ameliorated depression-like ethological behaviors and restored the decreased catecholamine levels in chronic variable stress (CVS)-induced depression rats. Additionally, it significantly improved the disturbance of gut microbiota induced by CVS stimuli, particularly affecting bacteria that produce short-chain fatty acids (SCFAs), such as bacteria species Lactobacillus spp.. In vitro fermentation study further confirmed that YLP-1 intake could promote SCFAs production by Lactobacillus spp. YLP-1 also mitigated the disruption of tryptophan metabolites in urine and serum. These findings provide evidences for the further development of YLP-1 as a macromolecular antidepressant drug.


Subject(s)
Antidepressive Agents , Fatty Acids, Volatile , Gastrointestinal Microbiome , Millettia , Polysaccharides , Tryptophan , Animals , Gastrointestinal Microbiome/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/chemistry , Male , Rats , Polysaccharides/pharmacology , Polysaccharides/chemistry , Millettia/chemistry , Tryptophan/metabolism , Fatty Acids, Volatile/metabolism , Depression/drug therapy , Depression/metabolism , Rats, Sprague-Dawley
16.
Carbohydr Polym ; 344: 122535, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39218555

ABSTRACT

Oat ß-(1 â†’ 3, 1 â†’ 4)-d-glucan (OBG), a linear polysaccharide primarily found in oat bran, has been demonstrated to possess immunomodulatory properties and regulate gut microbiota. This study aimed to investigate the impact of low molecular weight (Mw) OBG (155.2 kDa) on colonic injury and allergic symptoms induced by food allergy (FA), and to explore its potential mechanism. In Experiment 1, results indicated that oral OBG improved colonic inflammation and epithelial barrier, and significantly relieved allergy symptoms. Importantly, the OBG supplement altered the gut microbiota composition, particularly increasing the abundance of Lachnospiraceae and its genera, and promoted the production of short-chain fatty acids, especially butyrate. However, in Experiment 2, the gut microbial depletion eliminated these protective effects of OBG on the colon in allergic mice. Further, in Experiment 3, fecal microbiota transplantation and sterile fecal filtrate transfer directly validated the role of OBG-mediated gut microbiota and its metabolites in relieving FA and its induced colonic injury. Our findings suggest that low Mw OBG can alleviate FA-induced colonic damage by increasing Lachnospiraceae abundance and butyrate production, and provide novel insights into the health benefits and mechanisms of dietary polysaccharide intervention for FA.


Subject(s)
Avena , Butyrates , Colon , Food Hypersensitivity , Gastrointestinal Microbiome , Animals , Gastrointestinal Microbiome/drug effects , Mice , Colon/pathology , Colon/drug effects , Colon/metabolism , Butyrates/metabolism , Avena/chemistry , Clostridiales , beta-Glucans/pharmacology , beta-Glucans/chemistry , Mice, Inbred BALB C , Male , Glucans/pharmacology , Glucans/chemistry , Fatty Acids, Volatile/metabolism , Fecal Microbiota Transplantation
17.
J Nutr Sci Vitaminol (Tokyo) ; 70(4): 311-317, 2024.
Article in English | MEDLINE | ID: mdl-39218692

ABSTRACT

Chronic inflammation in adipose tissue is thought to contribute to insulin resistance, which involves the gut microbiota. Our previous studies have demonstrated that ingestion of 1-kestose can alter the gut microbiota composition, increase cecal butyrate levels, and improve insulin resistance in Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Additionally, we found that 1-kestose supplementation ameliorated insulin resistance in obese rat models fed a high-fat diet (HFD), although the effects of 1-kestose on the abundance of inflammation-related gene in adipose tissue and gut microbiota composition in these rats were not explored. This study aimed to investigate the impact of 1-kestose on these parameters in HFD-fed rats, compared to OLETF rats. Male Sprague-Dawley rats were divided into two dietary groups, control or HFD, for 19 wk. Each group was further subdivided to receive either tap water or tap water supplemented with 2% (w/v) 1-kestose throughout the study. We evaluated gene expression in adipose tissue, as well as short-chain fatty acids (SCFAs) levels and microbial composition in the cecum contents. 1-Kestose intake restored the increased relative abundance of tumor necrosis factor (Tnf) mRNA in adipose tissue and the reduced level of butyrate in the cecum contents of HFD-fed rats to those observed in control diet-fed rats. Additionally, 1-kestose consumption changed the composition of the gut microbiota, increasing Butyricicoccus spp., decreasing UGC-005 and Streptococcus spp., in the cecum contents of HFD-fed rats. Our findings suggest that 1-kestose supplementation reduces adipose tissue inflammation and increases butyrate levels in the gut of HFD-fed rats, associated with changes in the gut microbiota composition, distinct from those seen in OLETF rats.


Subject(s)
Adipose Tissue , Cecum , Diet, High-Fat , Fatty Acids, Volatile , Gastrointestinal Microbiome , Inflammation , RNA, Messenger , Rats, Sprague-Dawley , Animals , Gastrointestinal Microbiome/drug effects , Male , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Inflammation/metabolism , RNA, Messenger/metabolism , Rats , Fatty Acids, Volatile/metabolism , Cecum/microbiology , Cecum/metabolism , Insulin Resistance , Rats, Inbred OLETF , Obesity/metabolism , Obesity/microbiology , Dietary Supplements , Butyrates/metabolism
18.
Chin J Dent Res ; 27(3): 193-202, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39221980

ABSTRACT

Short-chain fatty acids (SCFAs) are the metabolites identified in both the oral cavity and the gut. They play an important role in the triggering, development and progression of systemic diseases. SCFAs can alter the gut microbial components, intestinal epithelium and host immune system, and are also associated with cancer incidence. Salivary SCFAs, produced by the oral microbiome, are correlated with some oral diseases. The occurrence of systemic diseases associated with gut SCFAs is more clearly defined than oral SCFAs. Salivary SCFAs can enter the bloodstream directly via inflamed gingiva to cause continuous low-grade systemic inflammation. Hence, salivary SCFAs could be an indicator for the early diagnosis of systemic diseases. Furthermore, they provide a basis for understanding the oral-systemic axis driven through salivary SCFAs in the pathogenesis of several diseases.


Subject(s)
Fatty Acids, Volatile , Saliva , Humans , Fatty Acids, Volatile/metabolism , Saliva/chemistry , Saliva/metabolism , Gastrointestinal Microbiome/physiology
19.
World J Microbiol Biotechnol ; 40(10): 314, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249571

ABSTRACT

This research propounds an innovative technology focused on sustainability to increase the biomass yield of Akkermansia muciniphila, the next-generation probiotic, using prebiotic sources to replace or reduce animal mucin levels. A series of experimental design approaches were developed aiming to optimize the growth of Akkermansiamuciniphila by incorporating extracts of green leafy vegetables and edible mushroom into the cultivation media. Experiments using kale extract (KE), Brassica oleracea L., associated with lyophilized mushroom extract (LME) of Pleurotus ostreatus were the most promising, highlighting the assays with 0.376% KE and 0.423% LME or 1.05% KE and 0.5% LME, in which 3.5 × 1010 CFU (Colony Forming Units) mL- 1 was achieved - higher than in experiments in optimized synthetic media. Such results enhance the potential of using KE and LME not only as mucin substitutes, but also as a source to increase Akkermansia muciniphila biomass yields and release short-chain fatty acids. The work is relevant to the food and pharmaceutical industries in the preparation of the probiotic ingredient.


Subject(s)
Akkermansia , Biomass , Culture Media , Prebiotics , Probiotics , Verrucomicrobia , Akkermansia/growth & development , Culture Media/chemistry , Verrucomicrobia/growth & development , Verrucomicrobia/metabolism , Pleurotus/growth & development , Pleurotus/metabolism , Fatty Acids, Volatile/metabolism , Plant Extracts/chemistry , Brassica/growth & development , Brassica/microbiology
20.
Front Cell Infect Microbiol ; 14: 1449030, 2024.
Article in English | MEDLINE | ID: mdl-39286812

ABSTRACT

Short-chain fatty acids (SCFAs), a subset of organic fatty acids with carbon chains ranging from one to six atoms in length, encompass acetate, propionate, and butyrate. These compounds are the endproducts of dietary fiber fermentation, primarily catalyzed by the glycolysis and pentose phosphate pathways within the gut microbiota. SCFAs act as pivotal energy substrates and signaling molecules in the realm of animal nutrition, exerting a profound influence on the intestinal, immune system, and intestinal barrier functions. Specifically, they contibute to 60-70% of the total energy requirements in ruminants and 10-25% in monogastric animals. SCFAs have demonstrated the capability to effectively modulate intestinal pH, optimize the absorption of mineral elements, and impede pathogen invasion. Moreover, they enhance the expression of proteins associated with intestinal tight junctions and stimulate mucus production, thereby refining intestinal tissue morphology and preserving the integrity of the intestinal structure. Notably, SCFAs also exert anti-inflammatory properties, mitigating inflammation within the intestinal epithelium and strengthening the intestinal barrier's defensive capabilities. The present review endeavors to synthesize recent findings regarding the role of SCFAs as crucial signaling intermediaries between the metabolic activities of gut microbiota and the status of porcine cells. It also provides a comprehensive overview of the current literature on SCFAs' impact on immune responses within the porcine intestinal mucosa.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Immunity, Mucosal , Intestinal Mucosa , Animals , Fatty Acids, Volatile/metabolism , Swine , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Animal Nutritional Physiological Phenomena
SELECTION OF CITATIONS
SEARCH DETAIL