Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 162
Filter
1.
Food Funct ; 15(19): 10110-10120, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39291835

ABSTRACT

Lead (Pb) is a highly toxic metal with no physiological function in humans, accumulates in the body through food intake, and causes gut microbiome disorders and other hazards. In the present study, we examined the efficacy of a combination of chondroitin sulfate and Lactiplantibacillus plantarum CCFM8661 (CCFM8661 + CS) on tissue Pb accumulation and pathological damage to the liver and kidneys, gut microbiota, and fecal metabolites in Pb-exposed mice. Oral administration of CCFM8661 + CS to Pb-exposed mice reduced Pb accumulation in the liver, kidney, and bone tissues (from 3.70, 14.11 and 121.20 mg g-1 wet tissue to 2.26, 8.72 and 65.57 mg g-1 wet tissue, respectively) and increased total antioxidant capacity, superoxide dismutase, and glutathione in the liver and kidneys. Additionally, gut microbiome analysis showed that CCFM8661 + CS intervention attenuated Pb-induced perturbation in gut microbiota, altering the abundance of bacteria such as Faecalibaculum, Ruminococcaceae UCG 014, Anaerostipes, and Enterorhabdus. Untargeted metabolomics analyses showed that CCFM8661 + CS significantly increased cinnamoylglycine, hippuric acid, and equol (to 31.24, 28.77 and 20.13 times the baseline, respectively) and decreased guanine and 4-coumaric acid (0.30 and 0.09 times the baseline, respectively) in the feces, affecting pathways such as purine and amino acid metabolism. Further analyses showed that promoting Pb excretion and restoring the Pb-impaired gut microbiome and its metabolism may be important contributors to CCFM8661 + CS alleviation of Pb toxicity.


Subject(s)
Chondroitin Sulfates , Gastrointestinal Microbiome , Lead , Metabolomics , Animals , Chondroitin Sulfates/metabolism , Chondroitin Sulfates/pharmacology , Mice , Gastrointestinal Microbiome/drug effects , Lead/toxicity , Lead/metabolism , Male , Kidney/drug effects , Kidney/metabolism , Lactobacillus plantarum/metabolism , Liver/metabolism , Liver/drug effects , Feces/microbiology , Lead Poisoning/metabolism , Lead Poisoning/drug therapy , Probiotics/pharmacology
2.
Food Chem ; 463(Pt 1): 141019, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39243605

ABSTRACT

The unique flavors of fermented foods significantly influence consumer purchasing choices, prompting widespread scientific interest in the flavor development process. Fermented rice and wheat foods are known for their unique flavors and they occupy an important place in the global diet. Many of these are produced on an industrial scale using starter cultures, whereas others rely on spontaneous fermentation, homemade production, or traditional activities. Microorganisms are key in shaping the sensory properties of fermented products through different metabolic pathways, thus earning the title "the essence of fermentation." Therefore, this study systematically summarizes the key microbial communities and their interactions that contribute positively to iconic fermented rice and wheat foods, such as steamed bread, bread, Mifen, and rice wine. This study revealed the mechanism by which these core microbial communities affect flavor and revealed the strategies of core microorganisms and related enzymes to enhance flavor during fermentation.

3.
Curr Res Food Sci ; 9: 100822, 2024.
Article in English | MEDLINE | ID: mdl-39263204

ABSTRACT

Sporolactobacillus is a genus of lactic acid bacteria, which can be widely found in soil. According to NCBI, only 20 strains of the genus Sporolactobacillus have been identified through phenotypic and genotypic analysis, indicating their relatively low numbers compared to other lactic acid bacteria. Currently, there is a growing interest in isolating and studying Sporolactobacillus, particularly focusing on its physiological characteristics and conducting in vitro experiments. This paper provides a review of the sources and physiological characteristics of Sporolactobacillus, along with genotype analysis, carbohydrate metabolism traits, and potential antibacterial properties. It also delves into basic physiological characteristics, lactic acid production, and applications, offering insights for the future utilization of Sporolactobacillus and laying a foundation for exploring its potential applications.

4.
Int J Biol Macromol ; 278(Pt 4): 135098, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39197612

ABSTRACT

The importance of synergy has been underscored in recent medical research for augmenting the efficacy of therapeutic interventions, targeting multiple biological pathways simultaneously. Our prior research elucidated that Dendrobium officinale polysaccharide (DOP) has the potential to prolong the lifespan of Caenorhabditis elegans (C. elegans) via regulating gut microbiota. Concurrently, spermidine (Spd), as a mimicking caloric restriction, facilitates autophagy and exerts a pronounced anti-aging effect. To enhance the anti-aging capabilities of DOP, we conducted a comprehensive study examining the combined effects of DOP and Spd in C. elegans, incorporating metabolomics analysis to investigate the underlying mechanisms. A combination of 250 mg/L DOP and 29.0 mg/L Spd yielded the most favorable outcomes in lifespan extension, evidencing a synergistic effect with a combination index (CI) of 0.65. In oxidative and heat stress tolerance assays, the observed CIs were 0.50 and 0.33, respectively. Metabolomic analysis highlighted significant alterations in metabolites related to lipid, nucleotide and energy metabolism, notably regulating glycerol 3-phosphate, linoleoyl glycerol, docosapentaenoic acid and ß-nicotinamide mononucleotide, nicotinamide adenine dinucleotide. The effects of DS on lipid metabolism were further validated using Oil Red O staining and triglyceride level in C. elegans. The results indicated that DS may primarily be via modulating lipid metabolism. To further confirm these findings, a high-fat diet-induced mouse model was employed. Consequently, it can be inferred that the synergistic anti-aging impact of DOP and Spd is likely mediated primarily through alterations in lipid metabolic processes.


Subject(s)
Caenorhabditis elegans , Dendrobium , Energy Metabolism , Lipid Metabolism , Metabolomics , Polysaccharides , Spermidine , Animals , Polysaccharides/pharmacology , Polysaccharides/chemistry , Dendrobium/chemistry , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Metabolomics/methods , Lipid Metabolism/drug effects , Energy Metabolism/drug effects , Spermidine/pharmacology , Spermidine/metabolism , Mice , Drug Synergism , Nucleotides/metabolism , Nucleotides/pharmacology , Aging/drug effects , Aging/metabolism , Longevity/drug effects , Oxidative Stress/drug effects
5.
Foods ; 13(14)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39063297

ABSTRACT

The mechanism of metabolites produced by lactic acid bacteria in mediating microbial interactions has been difficult to ascertain. This study comparatively evaluated the antimicrobial effect of the novel bacterium Pediococcus acidilactici CCFM18 and explored the global chemical view of its interactions with indicator bacteria. P. acidilactici CCFM18 had sufficiently strong antimicrobial activity to effectively inhibit the growth of the indicator bacteria and enhance their intracellular reactive oxygen species (ROS) level. The emerging technique of matrix-assisted laser desorption ionization-time-of-flight (MALDI-TOF) imaging mass spectrometry indicated that P. acidilactici CCFM18 increased the production of pediocin PA-1 and the penocin A profile during its interaction with the indicator bacteria, thus differing from P. acidilactici CCFM28 (a commonly used laboratory strain). Strikingly, the production of coagulin A was triggered only by signaling molecules made by the competing strain L. thermophilus, suggesting an idiosyncratic response from P. acidilactici CCFM18. Bioinformatic mining of the P. acidilactici CCFM18 draft genome sequence revealed gene loci that code for the complex secondary metabolites analyzed via MSI. Taken together, these results illustrate that chemical interactions between P. acidilactici CCFM18 and indicator bacteria exhibit high complexity and specificity and can drive P. acidilactici CCFM18 to produce different secondary metabolites.

6.
NPJ Biofilms Microbiomes ; 10(1): 47, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898089

ABSTRACT

Throughout the life span of a host, bifidobacteria have shown superior colonization and glycan abilities. Complex glycans, such as human milk oligosaccharides and plant glycans, that reach the colon are directly internalized by the transport system of bifidobacteria, cleaved into simple structures by extracellular glycosyl hydrolase, and transported to cells for fermentation. The glycan utilization of bifidobacteria introduces cross-feeding activities between bifidobacterial strains and other microbiota, which are influenced by host nutrition and regulate gut homeostasis. This review discusses bifidobacterial glycan utilization strategies, focusing on the cross-feeding involved in bifidobacteria and its potential health benefits. Furthermore, the impact of cross-feeding on the gut trophic niche of bifidobacteria and host health is also highlighted. This review provides novel insights into the interactions between microbe-microbe and host-microbe.


Subject(s)
Bifidobacterium , Gastrointestinal Microbiome , Homeostasis , Polysaccharides , Humans , Bifidobacterium/metabolism , Bifidobacterium/physiology , Polysaccharides/metabolism , Host Microbial Interactions , Animals , Fermentation
7.
Food Res Int ; 188: 114309, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823823

ABSTRACT

Previous studies have demonstrated that Ligilactobacillus salivarius CCFM 1266 exhibits anti-inflammatory properties and the capability to synthesize niacin. This study aimed to investigate the fermentative abilities of L. salivarius CCFM 1266 in fermented milk. Metabonomic analysis revealed that fermentation by L. salivarius CCFM 1266 altered volatile flavor compounds and metabolite profiles, including heptanal, nonanal, and increased niacin production. Genomic investigations confirmed that L. salivarius CCFM 1266 possess essential genes for the metabolism of fructose and mannose, affirming its proficiency in utilizing fructooligosaccharides and mannan oligosaccharides. The addition of fructooligosaccharides and mannan oligosaccharides during the fermentation process significantly facilitated the proliferation of L. salivarius CCFM 1266 in fermented milk, with growth exceeding 107 colony-forming units (CFU)/mL. This intervention not only augmented the microbial density but also modified the metabolite composition of fermented milk, resulting in an elevated presence of advantageous flavor compounds such as nonanal, 2,3-pentanedione, and 3-methyl-2-butanone. However, its influence on improving the texture of fermented milk was observed to be minimal. Co-fermentation of L. salivarius CCFM 1266 with commercial fermentation starters indicated that L. salivarius CCFM 1266 was compatible, similarly altering metabolite composition and increasing niacin content in fermented milk. In summary, the findings suggest that L. salivarius CCFM 1266 holds substantial promise as an adjunctive fermentation starter, capable of enhancing the nutritional diversity of fermented milk products.


Subject(s)
Cultured Milk Products , Fermentation , Ligilactobacillus salivarius , Metabolomics , Metabolomics/methods , Ligilactobacillus salivarius/metabolism , Cultured Milk Products/microbiology , Niacin/metabolism , Food Microbiology , Dairy Products/microbiology , Taste , Volatile Organic Compounds/analysis , Volatile Organic Compounds/metabolism , Animals
8.
Adv Nutr ; 15(6): 100233, 2024 06.
Article in English | MEDLINE | ID: mdl-38908894

ABSTRACT

Microbiota in early life is closely associated with the health of infants, especially premature ones. Probiotics are important drivers of gut microbiota development in preterm infants; however, there is no consensus regarding the characteristics of specific microbiota in preterm infants receiving probiotics. In this study, we performed a meta-analysis of 5 microbiome data sets (1816 stool samples from 706 preterm infants) to compare the gut microbiota of preterm infants exposed to probiotics with that of preterm infants not exposed to probiotics across populations. Despite study-specific variations, we found consistent differences in gut microbial composition and predicted functional pathways between the control and probiotic groups across different cohorts of preterm infants. The enrichment of Acinetobacter, Bifidobacterium, and Lactobacillus spp and the depletion of the potentially pathogenic bacteria Finegoldia, Veillonella, and Klebsiella spp. were the most consistent changes in the gut microbiota of preterm infants supplemented with probiotics. Probiotics drove microbiome transition into multiple preterm gut community types, and notably, preterm gut community type 3 had the highest α-diversity, with enrichment of Bifidobacterium and Bacteroides spp. At the functional level, the major predicted microbial pathways involved in peptidoglycan biosynthesis consistently increased in preterm infants supplemented with probiotics; in contrast, the crucial pathways associated with heme biosynthesis consistently decreased. Interestingly, Bifidobacterium sp. rather than Lactobacillus sp. gradually became dominant in gut microbiota of preterm infants using mixed probiotics, although both probiotic strains were administered at the same dosage. Taken together, our meta-analysis suggests that probiotics contribute to reshaping the microbial ecosystem of preterm infants at both the taxonomic and functional levels of the bacterial community. More standardized and relevant studies may contribute to better understanding the crosstalk among probiotics, the gut microbiota, and subsequent disease risk, which could help to give timely nutritional feeding guidance to preterm infants. This systematic review and meta-analysis was registered at PROSPERO (https://www.crd.york.ac.uk/PROSPERO/) as CRD42023447901.


Subject(s)
Gastrointestinal Microbiome , Infant, Premature , Probiotics , Humans , Gastrointestinal Microbiome/drug effects , Probiotics/administration & dosage , Infant, Newborn , Bifidobacterium , Feces/microbiology , Bacteria/classification , Lactobacillus , Female
9.
Food Funct ; 15(12): 6629-6641, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38812427

ABSTRACT

Gastrointestinal inflammation and intestinal barrier function have important effects on human health. Alcohol, an important foodborne hazard factor, damages the intestinal barrier, increasing the risk of disease. Lactobacillus reuteri strains have been reported to reduce gastrointestinal inflammation and strengthen the intestinal barrier. In this study, we selected three anti-inflammatory L. reuteri strains to evaluate their role in the protection of the intestinal barrier and their immunomodulatory activity in a mouse model of gradient alcohol intake. Among the three strains tested (FSCDJY33M3, FGSZY33L6, and FCQHCL8L6), L. reuteri FSCDJY33M3 was found to protect the intestinal barrier most effectively, possibly due to its ability to reduce the expression of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-alpha (TNF-α) and increase the expression of tight junction proteins (occludin, claudin-3). Genomic analysis suggested that the protective effects of L. reuteri FSCDJY33M3 may be related to functional genes and glycoside hydrolases associated with energy production and conversion, amino acid transport and metabolism, carbohydrate transport and metabolism, and DNA replication, recombination, and repair. These genes include COG2856, COG1804, COG2071, and COG1061, which encode adenine deaminase, acyl-CoA transferases, glutamine amidotransferase, RNA helicase, and glycoside hydrolases, including GH13_20, GH53, and GH70. Our results identified functional genes that may be related to protection against alcohol-induced intestinal barrier damage, which might be useful for screening lactic acid bacterial strains that can protect the intestinal barrier.


Subject(s)
Ethanol , Intestinal Mucosa , Limosilactobacillus reuteri , Probiotics , Limosilactobacillus reuteri/physiology , Animals , Mice , Intestinal Mucosa/metabolism , Probiotics/pharmacology , Male , Mice, Inbred C57BL , Humans , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Intestines/microbiology
10.
Environ Microbiol ; 26(5): e16622, 2024 May.
Article in English | MEDLINE | ID: mdl-38757466

ABSTRACT

Microbial communities that reduce nitrous oxide (N2O) are divided into two clades, nosZI and nosZII. These clades significantly differ in their ecological niches and their implications for N2O emissions in terrestrial environments. However, our understanding of N2O reducers in aquatic systems is currently limited. This study investigated the relative abundance and diversity of nosZI- and nosZII-type N2O reducers in rivers and their impact on N2O emissions. Our findings revealed that stream sediments possess a high capacity for N2O reduction, surpassing N2O production under high N2O/NO3- ratio conditions. This study, along with others in freshwater systems, demonstrated that nosZI marginally dominates more often in rivers. While microbes containing either nosZI and nosZII were crucial in reducing N2O emissions, the net contribution of nosZII-containing microbes was more significant. This can be attributed to the nir gene co-occurring more frequently with the nosZI gene than with the nosZII gene. The diversity within each clade also played a role, with nosZII species being more likely to function as N2O sinks in streams with higher N2O concentrations. Overall, our findings provide a foundation for a better understanding of the biogeography of stream N2O reducers and their effects on N2O emissions.


Subject(s)
Bacteria , Nitrous Oxide , Rivers , Nitrous Oxide/metabolism , Rivers/microbiology , Rivers/chemistry , Bacteria/genetics , Bacteria/classification , Bacteria/metabolism , Geologic Sediments/microbiology , Oxidation-Reduction , Phylogeography , Phylogeny , Microbiota
11.
Cell Genom ; 4(6): 100559, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38740021

ABSTRACT

The gut microbiome displays genetic differences among populations, and characterization of the genomic landscape of the gut microbiome in China remains limited. Here, we present the Chinese Gut Microbial Reference (CGMR) set, comprising 101,060 high-quality metagenomic assembled genomes (MAGs) of 3,707 nonredundant species from 3,234 fecal samples across primarily rural Chinese locations, 1,376 live isolates mainly from lactic acid bacteria, and 987 novel species relative to worldwide databases. We observed region-specific coexisting MAGs and MAGs with probiotic and cardiometabolic functionalities. Preliminary mouse experiments suggest a probiotic effect of two Faecalibacillus intestinalis isolates in alleviating constipation, cardiometabolic influences of three Bacteroides fragilis_A isolates in obesity, and isolates from the genera Parabacteroides and Lactobacillus in host lipid metabolism. Our study expands the current microbial genomes with paired isolates and demonstrates potential host effects, contributing to the mechanistic understanding of host-microbe interactions.


Subject(s)
Gastrointestinal Microbiome , Probiotics , Gastrointestinal Microbiome/genetics , China , Animals , Humans , Mice , Male , Female , Genome, Bacterial/genetics , Genome, Microbial , Feces/microbiology , Obesity/microbiology , Adult , Mice, Inbred C57BL
12.
Food Res Int ; 186: 114287, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729740

ABSTRACT

The gut microbiota is widely acknowledged as a crucial factor in regulating host health. The structure of dietary fibers determines changes in the gut microbiota and metabolic differences resulting from their fermentation, which in turn affect gut microbe-related health effects. ß-Glucan (BG) is a widely accessible dietary fiber to humans, and its structural characteristics vary depending on the source. However, the interactions between different structural BGs and gut microbiota remain unclear. This study used an in vitro fermentation model to investigate the effects of BG on gut microbiota, and microbiomics and metabolomics techniques to explore the relationship between the structure of BG, bacterial communities, and metabolic profiles. The four sources of BG (barley, yeast, algae, and microbial fermentation) contained different types and proportions of glycosidic bonds, which differentially altered the bacterial community. The BG from algal sources, which contained only ß(1 â†’ 4) glycosidic bonds, was the least metabolized by the gut microbiota and caused limited metabolic changes. The other three BGs contain more diverse glycosidic bonds and can be degraded by bacteria from multiple genera, causing a wider range of metabolic changes. This work also suggested potential synergistic degradation relationships between gut bacteria based on BG. Overall, this study deepens the structural characterization-microbial-functional understanding of BGs and provides theoretical support for the development of gut microbiota-targeted foods.


Subject(s)
Bacteria , Fermentation , Gastrointestinal Microbiome , beta-Glucans , beta-Glucans/metabolism , Gastrointestinal Microbiome/physiology , Humans , Bacteria/metabolism , Bacteria/classification , Dietary Fiber/metabolism , Metabolomics
13.
Food Chem ; 450: 139309, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38631200

ABSTRACT

Flammulina velutipes, a widely cultivated species of edible fungus, exhibits diverse functional activities attributed to its polysaccharides. In this study, we employed an in vitro model to investigate the impact of F. velutipes polysaccharides (FVP) fermentation on gut microbiota, with a particular focus on Bacteroides. FVP fermentation resulted in the proliferation of microbiota associated with short-chain fatty acid (SCFA) metabolism and suppression of Escherichia-Shigella. Bacteroides emerged as potential primary degraders of FVP, with species-level analysis identifying the preference of B. thetaiotaomicron and B. intestinalis in FVP degradation. Metabolomics analysis revealed significant increases in hypoxanthine and 7-methyladenine contents, with histidine metabolism emerging as the most enriched pathway. B. nordii and B. xylanisolvens exhibited the most influence on amino acid and SCFA metabolism. Understanding the mechanisms by which gut microbiota metabolize FVP can provide valuable insights into the potential of FVP to promote intestinal health and disease prevention.


Subject(s)
Bacteroides , Feces , Fermentation , Flammulina , Gastrointestinal Microbiome , Humans , Flammulina/metabolism , Flammulina/chemistry , Feces/microbiology , Bacteroides/metabolism , Polysaccharides/metabolism , Polysaccharides/chemistry , Fatty Acids, Volatile/metabolism , Bacteria/metabolism , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Male , Adult
14.
Food Funct ; 15(9): 4763-4772, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38590256

ABSTRACT

Inulins, galacto-oligosaccharides (GOS) and polyphenols are considered to stimulate the growth of Akkermansia muciniphila (A. muciniphila) in the gut. We performed a meta-analysis of six microbiome studies (821 stool samples from 451 participants) to assess the effects of inulin, GOS, and polyphenols on the abundance of A. muciniphila in the gut. The intervention of GOS increased the relative abundance of A. muciniphila in healthy participants. Additionally, metabolic pathways associated with carbohydrate metabolism and short-chain fatty acid release were enriched following the GOS intervention. Furthermore, after the GOS intervention, the coexisting microbial communities of A. muciniphila, such as Eubacterium hallii and Bacteroides, exhibited an enhanced correlation with A. muciniphila. In conclusion, our findings suggest that GOS may promote the growth of A. muciniphila in the gut by modulating the gut microbiota composition.


Subject(s)
Akkermansia , Gastrointestinal Microbiome , Inulin , Oligosaccharides , Polyphenols , Gastrointestinal Microbiome/drug effects , Polyphenols/pharmacology , Inulin/pharmacology , Humans , Oligosaccharides/pharmacology , Oligosaccharides/metabolism , Feces/microbiology , Verrucomicrobia , Prebiotics , Galactose
16.
Int J Occup Med Environ Health ; 37(2): 165-175, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38529760

ABSTRACT

OBJECTIVES: Occupational stress is a common complaint in nurses, who perceived more sense of effort-reward imbalance (ERI). Suboptimal health status (SHS) is a state between health and disease. However, the correlation between ERI and SHS is unclear. Therefore, the aim of this study was to examine the prevalence of SHS and ERI and evaluate the relationship between ERI and SHS in clinical nurses by a cross-sectional study. MATERIAL AND METHODS: The current cross-sectional study was conducted through an online survey at Dongping People's Hospital in China. A total of 633 completed surveys were received. Effort-reward imbalance was measured by subscales of the ERI questionnaire. SHS was measured by the Suboptimal Health Status Questionnaire - 25 (SHSQ-25). The relationship between ERI and SHS in nurses was subsequently assessed by Spearman's correlation coefficient and logistic regression model. RESULTS: The mean age of the optimal health status (OHS) group (M±SD 26.3±7.3 years) was younger than the SHS group (M±SD 30.3±6.9 years). The prevalence of SHS was 54.5% (345/633). Female nurses aged ≥30 years, a junior college or university graduate educational level, smokers, and nurses without regular exercise were at a higher risk of SHS. In Spearman's correlation analysis, ERI reflected by the effort-reward ratio was correlated with SHSQ-25 score (r = 0.662, p < 0.001). In logistic regression, ERI was strongly associated with SHS after potential confounding factors adjusting (OR 27.924, 95% CI 22.845-34.132). CONCLUSIONS: The prevalence of SHS was significantly high in clinical nurses. Administrators should pay more attention to health status of female nurses aged ≥30 years, with a junior college or bachelor's degree, smoking, and without regular exercise to reduce the SHS and ERI. Int J Occup Med Environ Health. 2024;37(2):166-75.


Subject(s)
Health Status , Nursing Staff, Hospital , Occupational Stress , Reward , Humans , Cross-Sectional Studies , Female , Adult , Occupational Stress/epidemiology , Occupational Stress/psychology , China/epidemiology , Male , Nursing Staff, Hospital/psychology , Nursing Staff, Hospital/statistics & numerical data , Surveys and Questionnaires , Prevalence , Middle Aged
17.
Food Funct ; 15(7): 3327-3339, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38465411

ABSTRACT

Bacteroides is a common intestinal bacterium closely associated with host colitis. However, relevant studies have been focused on the genus level, which could not identify the major Bacteroides species associated with intestinal disease. Thus, we have evaluated the Bacteroides species structure in healthy people and mouse intestinal tracts and explored the change in major Bacteroides species during colitis development. The results demonstrated that B. uniformis with a high abundance in the intestinal tract of healthy people and mice may be a core species that contributes to colitis remission. The results of animal experiments reported that B. uniformis FNMHLBE1K1 (1K1) could alleviate the severity of colitis and enhance the expression of the tight junction protein occludin by regulating gut microbiota. Notably, the protective roles of 1K1 may be attributed to some specific genes. This study revealed that B. uniformis is a key microbe influencing the occurrence and development of colitis and it provides a scientific basis for screening the next generation of probiotics.


Subject(s)
Colitis, Ulcerative , Colitis , Humans , Mice , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis/chemically induced , Colitis/drug therapy , Colitis/genetics , Bacteroides/genetics , Intestines , Dextran Sulfate/adverse effects , Disease Models, Animal , Mice, Inbred C57BL , Colon
18.
Food Funct ; 15(7): 3709-3721, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38488198

ABSTRACT

Antibiotic-associated diarrhea (AAD) is a self-limiting condition that can occur during antibiotic therapy. Our previous studies have found that a combination of Bacteroides uniformis and Bifidobacterium adolescentis can effectively alleviate AAD. However, the use of B. uniformis is still strictly limited. Therefore, this study attempted to use yeast ß-glucan to enrich the abundance of B. uniformis in the intestine and supplement Bifidobacterium adolescentis to exert a synergistic effect. The lincomycin hydrochloride-induced AAD model was administered yeast ß-glucan or a mixture of B. adolescentis CCFM1285 by gavage for one week. Subsequently, changes in the colonic histopathological structure, inflammatory factors, intestinal epithelial permeability and integrity, metabolites, and gut microbiota diversity were assessed. We found that yeast ß-glucan, alone or in combination with B. adolescentis CCFM1285, can help attenuate systemic inflammation, increase the rate of tissue structural recovery, regulate metabolism, and restore the gut microbiota. Specifically, the combination of yeast ß-glucan and B. adolescentis CCFM1285 was more effective in decreasing interleukin-6 levels, improving pathological changes in the colon, and upregulating occludin expression. Therefore, our study showed that the combination of yeast ß-glucan and B. adolescentis CCFM1285 is an efficacious treatment for AAD.


Subject(s)
Bifidobacterium adolescentis , Gastrointestinal Microbiome , beta-Glucans , Mice , Animals , Saccharomyces cerevisiae , beta-Glucans/pharmacology , Diarrhea/chemically induced , Diarrhea/drug therapy , Anti-Bacterial Agents/adverse effects
19.
Food Funct ; 15(8): 4338-4353, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38533674

ABSTRACT

Vitamin D deficiency (VDD) during early life emerges as a potential risk factor for autism spectrum disorder (ASD). Individuals with autism commonly exhibit lower vitamin D (VD) levels compared to the general population, and VD deficiency is prevalent during pregnancy and lactation. Moreover, gastrointestinal comorbidity, prevalent in ASD patients, correlates closely with disruptions in the gut microbiota and altered intestinal permeability. Therefore, it is fascinating and significant to explore the effects of maternal VD deficiency during pregnancy and lactation on the maturation of the gut microbiota of the offspring and its relevance to autism spectrum disorders. In this study, we established maternal pregnancy and lactation VD-deficient mouse models, employed shotgun macrogenomic sequencing to unveil alterations in the gut microbiome of offspring mice, and observed autism-related behaviours. Furthermore, fecal microbial transplantation (FMT) reversed repetitive and anxious behaviours and alleviated social deficits in offspring mice by modulating the gut microbiota and increasing short-chain fatty acid levels in the cecum, along with influencing the concentrations of claudin-1 and occludin in the colon. Our findings confirm that VDD during pregnancy and lactation is a risk factor for autism in the offspring, with disturbances in the structure and function of the offspring's gut microbiota contributing at least part of the effect. The study emphasises the importance of nutrition and gut health early in life. Simultaneously, this study further demonstrates the effect of VDD on ASD and provides potential ideas for early prevention and intervention of ASD.


Subject(s)
Autism Spectrum Disorder , Gastrointestinal Microbiome , Vitamin D Deficiency , Animals , Vitamin D Deficiency/complications , Mice , Female , Male , Pregnancy , Autism Spectrum Disorder/microbiology , Autism Spectrum Disorder/metabolism , Homeostasis , Mice, Inbred C57BL , Disease Models, Animal , Autistic Disorder/metabolism , Autistic Disorder/microbiology , Fecal Microbiota Transplantation , Behavior, Animal , Lactation , Vitamin D/metabolism , Prenatal Exposure Delayed Effects
20.
Int J Biol Macromol ; 261(Pt 2): 129815, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38296122

ABSTRACT

In this study, we investigated the impact of Lactiplantibacillus plantarum (L. plantarum) CCFM8661 on the gut microbiota, and the serum and fecal metabolomes in lead (Pb)-exposed individuals. The volunteers recruited for this study were divided into two treatment groups, (i) the placebo (control) and (ii) the L. plantarum CCFM8661 treatment groups. The analysis revealed that probiotic intervention reversed some of the changes in Pb exposure-induced intestinal bacterial abundance, including the abundance of Parabacteroides, Bacteroides, Clostridiaceae, and Erysipelotrichaceae. An analysis of the fecal metabolome identified 26 differential metabolites involved in purine metabolism, unsaturated fatty acid metabolism, and other pathways. Serum metabolite analysis showed that L. plantarum CCFM8661 treatment altered the serum metabolite levels of various metabolic pathways, such as the glycerophospholipid, amino acid, and glutathione metabolism pathways. These results suggest that L. plantarum CCFM8661 may have beneficial effects on Pb-exposed populations by modulating the gut microbiota, host serum metabolism, and the metabolism of the gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Lactobacillus plantarum , Probiotics , Humans , Amino Acids , Bacteroides , Bacteroidetes , Lead/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL