Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 190
Filtrar
1.
Food Chem ; 463(Pt 1): 141083, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39241427

RESUMEN

Chickpea milk is a nutrient-rich plant-based milk, but its pronounced beany flavour limits consumer acceptance. To address this issue, chickpea milk was fermented using two strains of Lactiplantibacillus plantarum, FMBL L23251 and L23252, which efficiently utilize chickpea milk. L. plantarum FMBL L23251 demonstrated superior fermentation characteristics. Fermentation with L. plantarum FMBL L23251 resulted in a 1.90-fold increase in vitamin B3 (271.66 ng/ml to 516.15 ng/ml) and a 1.58-fold increase in vitamin B6 (91.24 ng/ml to 144.16 ng/ml) through the L-aspartic acid pathway and the 1-deoxy-D-xylulose-5-phosphate (DXP)-independent pathway, respectively. Furthermore, L. plantarum FMBL L23251 effectively removed beany flavours due to its enhanced pathway for pyruvate metabolism. The main aldehydes are converted into corresponding alcohols or acids, resulting in 87.74 % and 96.99 % reductions in hexanal and 2-pentyl-furan, respectively. In summary, the fermentation of L. plantarum FMBL L23251 generated fermented chickpea milk that is rich in B vitamins and provides a better flavour.

2.
Food Chem ; 463(Pt 1): 141019, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39243605

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-39263204

RESUMEN

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.
Food Funct ; 15(19): 10110-10120, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39291835

RESUMEN

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.


Asunto(s)
Sulfatos de Condroitina , Microbioma Gastrointestinal , Plomo , Metabolómica , Animales , Sulfatos de Condroitina/metabolismo , Sulfatos de Condroitina/farmacología , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Plomo/toxicidad , Plomo/metabolismo , Masculino , Riñón/efectos de los fármacos , Riñón/metabolismo , Lactobacillus plantarum/metabolismo , Hígado/metabolismo , Hígado/efectos de los fármacos , Heces/microbiología , Intoxicación por Plomo/metabolismo , Intoxicación por Plomo/tratamiento farmacológico , Probióticos/farmacología
5.
Food Funct ; 15(17): 8797-8809, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39114922

RESUMEN

Probiotics can alleviate alcoholic liver disease. However, whether inactive counterparts can produce similar outcomes requires further investigation. We investigated the effects of viable (V) and dead (D) Lactobacillus paracasei CCFM1120 on alcohol-induced ALD mice. The results showed that CCFM1120V and D ameliorated the disease symptoms and intestinal injury. Specifically, these interventions strengthened the intestinal barrier, as evidenced by the increased expression of ZO-1 (zonula occludens 1), occludin, and claudin-1 in the colon and the restored ileal microstructure, including the villi and crypts. In addition, they enhanced the antioxidant capacity of the liver by reducing the production of malondialdehyde and increasing the levels of glutathione and superoxide dismutase. The activation of Nrf2 and HO-1 may be responsible for recovering the antioxidant capacity. Interventions can decrease mouse TNF-α, IL-6 and IL-1ß content in serum, probably through the TLR4/MyD88/NF-κB pathway. Furthermore, they possess the ability to restore the quantities of bacteria responsible for producing butyric acid, such as Lactobacillus, Blautia, Bifidobacterium, Ruminococcaceae, Faecalibaculum and Lachnospiraceae. Taken together, CCFM1120V and D apparently can modify the composition of the gut microbiota, foster the gastrointestinal equilibrium, fortify the intestinal barrier, augment the antioxidant capacity of the liver, and effectively shield it from ethanol-induced injury.


Asunto(s)
Microbioma Gastrointestinal , Lacticaseibacillus paracasei , Hepatopatías Alcohólicas , Factor 88 de Diferenciación Mieloide , Factor 2 Relacionado con NF-E2 , FN-kappa B , Probióticos , Receptor Toll-Like 4 , Animales , Hepatopatías Alcohólicas/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Receptor Toll-Like 4/metabolismo , FN-kappa B/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Probióticos/farmacología , Masculino , Lacticaseibacillus paracasei/metabolismo , Transducción de Señal , Ratones Endogámicos C57BL , Hígado/metabolismo , Antioxidantes/metabolismo , Proteínas de la Membrana , Hemo-Oxigenasa 1
6.
Foods ; 13(14)2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39063297

RESUMEN

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.

7.
Front Microbiol ; 15: 1399743, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39021621

RESUMEN

Little has been known about symbiotic relationships and host specificity for symbionts in the human gut microbiome so far. Bifidobacteria are a paragon of the symbiotic bacteria biota in the human gut. In this study, we characterized the population genetic structure of three bifidobacteria species from 58 healthy mother-infant pairs of three ethnic groups in China, geographically isolated, by Rep-PCR, multi-locus sequence analysis (MLSA), and in vitro carbohydrate utilization. One hundred strains tested were incorporated into 50 sequence types (STs), of which 29 STs, 17 STs, and 4 STs belong to B. longum subsp. longum, B. breve, and B. animalis subsp. lactis, respectively. The conspecific strains from the same mother-child pair were genetically very similar, supporting the vertical transmission of Bifidobacterium phylotypes from mother to offspring. In particular, results based on allele profiles and phylogeny showed that B. longum subsp. longum and B. breve exhibited considerable intraspecies genetic heterogeneity across three ethnic groups, and strains were clustered into ethnicity-specific lineages. Yet almost all strains of B. animalis subsp. lactis were incorporated into the same phylogenetic clade, regardless of ethnic origin. Our findings support the hypothesis of co-evolution between human gut symbionts and their respective populations, which is closely linked to the lifestyle of specific bacterial lineages. Hence, the natural and evolutionary history of Bifidobacterium species would be an additional consideration when selecting bifidobacterial strains for industrial and therapeutic applications.

8.
NPJ Biofilms Microbiomes ; 10(1): 47, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898089

RESUMEN

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.


Asunto(s)
Bifidobacterium , Microbioma Gastrointestinal , Homeostasis , Polisacáridos , Humanos , Bifidobacterium/metabolismo , Bifidobacterium/fisiología , Polisacáridos/metabolismo , Interacciones Microbiota-Huesped , Animales , Fermentación
9.
Food Funct ; 15(14): 7441-7451, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38904342

RESUMEN

Liver injury is a life-threatening condition, and the hepatoprotective potential of cyanidin-3-glucoside (C3G) has been previously demonstrated. However, due to the low bioavailability, it has been doubtful that relatively low concentrations of intact C3G in vivo could account for these bioactivities. In this study, the hepatoprotective effects of intragastric and intravenous administration of C3G were investigated in a CCl4 induced liver injury model. Intragastric C3G administration was more effective than intravenous C3G injection in reducing serum damage biomarkers, oxidative stress, and inflammatory responses, indicating that absorption of C3G into the bloodstream does not fully account for its observed benefits in vivo. Furthermore, intragastric C3G administration modulated the gut microbiota structure and increased the contents of five metabolites in the feces and serum with high inter-individual variation, indicating the key role of the interaction between C3G and the gut microbiota. At equivalent doses, the metabolites cyanidin and protocatechuic acid exhibited greater efficacy than C3G in reducing apoptosis and ROS production by activating the Nrf2 pathway in an AAPH-induced oxidative stress model. To achieve the desired health effects via C3G-rich food intake, more attention should be paid to microbially derived catabolites. Screening of specific metabolite-producing strains will help overcome individual differences and enhance the health-promoting effects of C3G.


Asunto(s)
Antocianinas , Microbioma Gastrointestinal , Glucósidos , Estrés Oxidativo , Microbioma Gastrointestinal/efectos de los fármacos , Antocianinas/farmacología , Antocianinas/administración & dosificación , Animales , Glucósidos/farmacología , Glucósidos/administración & dosificación , Masculino , Estrés Oxidativo/efectos de los fármacos , Hígado/metabolismo , Hígado/efectos de los fármacos , Ratas , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Ratones , Sustancias Protectoras/farmacología , Sustancias Protectoras/administración & dosificación , Ratas Sprague-Dawley , Administración Intravenosa
10.
Adv Nutr ; 15(6): 100233, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38908894

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Recien Nacido Prematuro , Probióticos , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos/administración & dosificación , Recién Nacido , Bifidobacterium , Heces/microbiología , Bacterias/clasificación , Lactobacillus , Femenino
11.
Food Res Int ; 188: 114309, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38823823

RESUMEN

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.


Asunto(s)
Productos Lácteos Cultivados , Fermentación , Ligilactobacillus salivarius , Metabolómica , Metabolómica/métodos , Ligilactobacillus salivarius/metabolismo , Productos Lácteos Cultivados/microbiología , Niacina/metabolismo , Microbiología de Alimentos , Productos Lácteos/microbiología , Gusto , Compuestos Orgánicos Volátiles/análisis , Compuestos Orgánicos Volátiles/metabolismo , Animales
12.
Cell Genom ; 4(6): 100559, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38740021

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Microbioma Gastrointestinal/genética , China , Animales , Humanos , Ratones , Masculino , Femenino , Genoma Bacteriano/genética , Genoma Microbiano , Heces/microbiología , Obesidad/microbiología , Adulto , Ratones Endogámicos C57BL
13.
Food Funct ; 15(12): 6629-6641, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38812427

RESUMEN

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.


Asunto(s)
Etanol , Mucosa Intestinal , Limosilactobacillus reuteri , Probióticos , Limosilactobacillus reuteri/fisiología , Animales , Ratones , Mucosa Intestinal/metabolismo , Probióticos/farmacología , Masculino , Ratones Endogámicos C57BL , Humanos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Intestinos/microbiología
14.
Food Res Int ; 186: 114287, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729740

RESUMEN

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.


Asunto(s)
Bacterias , Fermentación , Microbioma Gastrointestinal , beta-Glucanos , beta-Glucanos/metabolismo , Microbioma Gastrointestinal/fisiología , Humanos , Bacterias/metabolismo , Bacterias/clasificación , Fibras de la Dieta/metabolismo , Metabolómica
16.
Food Chem ; 450: 139309, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-38631200

RESUMEN

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.


Asunto(s)
Bacteroides , Heces , Fermentación , Flammulina , Microbioma Gastrointestinal , Humanos , Flammulina/metabolismo , Flammulina/química , Heces/microbiología , Bacteroides/metabolismo , Polisacáridos/metabolismo , Polisacáridos/química , Ácidos Grasos Volátiles/metabolismo , Bacterias/metabolismo , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Masculino , Adulto
17.
Food Funct ; 15(9): 4763-4772, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38590256

RESUMEN

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.


Asunto(s)
Akkermansia , Microbioma Gastrointestinal , Inulina , Oligosacáridos , Polifenoles , Microbioma Gastrointestinal/efectos de los fármacos , Polifenoles/farmacología , Inulina/farmacología , Humanos , Oligosacáridos/farmacología , Oligosacáridos/metabolismo , Heces/microbiología , Verrucomicrobia , Prebióticos , Galactosa
18.
Food Funct ; 15(7): 3327-3339, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38465411

RESUMEN

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.


Asunto(s)
Colitis Ulcerosa , Colitis , Humanos , Ratones , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/genética , Colitis Ulcerosa/microbiología , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/genética , Bacteroides/genética , Intestinos , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Colon
19.
Food Funct ; 15(7): 3709-3721, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38488198

RESUMEN

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.


Asunto(s)
Bifidobacterium adolescentis , Microbioma Gastrointestinal , beta-Glucanos , Ratones , Animales , Saccharomyces cerevisiae , beta-Glucanos/farmacología , Diarrea/inducido químicamente , Diarrea/tratamiento farmacológico , Antibacterianos/efectos adversos
20.
Appl Environ Microbiol ; 90(3): e0207423, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38319094

RESUMEN

Bifidobacterium breve, one of the main bifidobacterial species colonizing the human gastrointestinal tract in early life, has received extensive attention for its purported beneficial effects on human health. However, exploration of the mode of action of such beneficial effects exerted by B. breve is cumbersome due to the lack of effective genetic tools, which limits its synthetic biology application. The widespread presence of CRISPR-Cas systems in the B. breve genome makes endogenous CRISPR-based gene editing toolkits a promising tool. This study revealed that Type I-C CRISPR-Cas systems in B. breve can be divided into two groups based on the amino acid sequences encoded by cas gene clusters. Deletion of the gene coding uracil phosphoribosyl-transferase (upp) was achieved in five B. breve strains from both groups using this system. In addition, translational termination of uracil phosphoribosyl-transferase was successfully achieved in B. breve FJSWX38M7 by single-base substitution of the upp gene and insertion of three stop codons. The gene encoding linoleic acid isomerase (bbi) in B. breve, being a characteristic trait, was deleted after plasmid curing, which rendered it unable to convert linoleic acid into conjugated linoleic acid, demonstrating the feasibility of successive editing. This study expands the toolkit for gene manipulation in B. breve and provides a new approach toward functional genome editing and analysis of B. breve strains.IMPORTANCEThe lack of effective genetic tools for Bifidobacterium breve is an obstacle to studying the molecular mechanisms of its health-promoting effects, hindering the development of next-generation probiotics. Here, we introduce a gene editing method based on the endogenous CRISPR-Cas system, which can achieve gene deletion, single-base substitution, gene insertion, and successive gene editing in B. breve. This study will facilitate discovery of functional genes and elucidation of molecular mechanisms of B. breve pertaining to health-associated benefits.


Asunto(s)
Bifidobacterium breve , Sistemas CRISPR-Cas , Humanos , Edición Génica/métodos , Bifidobacterium breve/genética , Ácido Linoleico , Transferasas/genética , Uracilo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA