RESUMEN
Background: Current treatments for inflammatory bowel disease (IBD) are relatively futile and the extended use of drugs may reduce effectiveness. Several probiotic strains have shown promise in relieving/treating IBD symptoms. Objectives: The current study investigated the impact of fermented soymilk with a mixture of probiotic starter cultures containing Lactobacillus rhamnosus, L. casei, L. plantarum, L. acidophilus, Bifidobacterium longum, and B. animalis subsp. lactis in rats with dextran sulfate sodium (DSS)-induced colitis compared to control. Methods: Rats were randomly assigned to five groups (5 rats/group; n = 25): G1: negative normal control; G2: positive control (DSS); G3: DSS with sulfasalazine (DSS-Z); G4: DSS with soymilk (DSS-SM), and G5: DSS with fermented soymilk (DSS-FSM). Parameters monitored included the following: the disease activity index (DAI), macroscopic and histological assessments of colitis, and a fecal microbial analysis performed to assess the severity of inflammation and ulceration. Results: The DSS-FSM rats group exhibited lower DAI scores (p < 0.05) than other treated groups during the induction period. A macroscopical examination revealed no ulceration or swelling in the intestinal mucosa of rats in the DSS-FSM-treated group, resembling the findings in the negative control group. In the positive control (DSS group), the colon tissue showed increased inflammation (p < 0.05), whereas those in the DSS-SM- and DSS-FSM-treated rats groups did not show significant macroscopic scores of colitis. The positive DSS control and DSS-Z groups had crypt erosion and ulceration areas, severe crypt damage, and epithelial surface erosion, which were absent in the negative control and DSS-FSM groups. The counts of Lactobacillus spp. and Bifidobacterium spp. remained stable in both G1 and G5 over 4 weeks. The consumption of fermented soymilk with a mixture of probiotics could minimize the severity of DSS-induced colitis in rats. Conclusion, it was found that fermented soymilk containing Lactobacilli and Bifidobacterium might be an effective vehicle for reducing the severity of DSS-induced colitis in rats.
Asunto(s)
Bifidobacterium , Colitis , Sulfato de Dextran , Fermentación , Lactobacillus , Probióticos , Leche de Soja , Animales , Colitis/inducido químicamente , Colitis/terapia , Colitis/microbiología , Ratas , Masculino , Lactobacillus/metabolismo , Bifidobacterium/metabolismo , Heces/microbiología , Colon/microbiología , Colon/patología , Colon/metabolismo , Modelos Animales de Enfermedad , Ratas Sprague-DawleyRESUMEN
Obesity is a major global health concern. Studies suggest that the gut microflora may play a role in protecting against obesity. Probiotics, including lactic acid bacteria and Bifidobacterium, have garnered attention for their potential in obesity prevention. However, the effects of Bifidobacterium-fermented products on obesity have not been thoroughly elucidated. Bifidobacterium, which exists in the gut of animals, is known to enhance lipid metabolism. During fermentation, it produces acetic acid, which has been reported to improve glucose tolerance and insulin resistance, and exhibit anti-obesity and anti-diabetic effects. Functional foods have been very popular around the world, and fermented milk is a good candidate for enrichment with probiotics. In this study, we aim to evaluate the beneficial effects of milks fermented with Bifidobacterium strains on energy metabolism and obesity prevention. Three Bifidobacterium strains (Bif-15, Bif-30, and Bif-39), isolated from newborn human feces, were assessed for their acetic acid production and viability in milk. These strains were used to ferment milk. Otsuka-Long-Evans Tokushima Fatty (OLETF) rats administered Bif-15-fermented milk showed significantly lower weight gain compared to those in the water group. The phosphorylation of AMPK was increased and the expression of lipogenic genes was suppressed in the liver of rats given Bif-15-fermented milk. Additionally, gene expression related to respiratory metabolism was significantly increased in the soleus muscle of rats given Bif-15-fermented milk. These findings suggest that milk fermented with the Bifidobacterium strain Bif-15 can improve lipid metabolism and suppress obesity.
Asunto(s)
Bifidobacterium , Metabolismo de los Lípidos , Lipogénesis , Obesidad , Animales , Obesidad/metabolismo , Obesidad/microbiología , Bifidobacterium/metabolismo , Ratas , Humanos , Masculino , Probióticos , Músculo Esquelético/metabolismo , Productos Lácteos Cultivados/microbiología , Leche/metabolismo , Leche/microbiología , Fermentación , Ratas Endogámicas OLETF , Hígado/metabolismo , Metabolismo EnergéticoRESUMEN
The role of the gut microbiota in the gut-brain axis has attracted attention in recent years. Some gut microbiota produces γ-aminobutyric acid (GABA), a major inhibitory neurotransmitter in mammals, in vitro, but the correlation between gut microbiota composition and intestinal GABA concentration, as well as the action of intestinal GABA in vivo, are poorly understood. Herein, we found that the intestinal GABA concentration was increased in mice by the intervention of the gut microbiota with neomycin or Bifidobacterium bifidum TMC3115 (TMC3115). Administration of TMC3115 reduced anxiety without affecting serum levels of serotonin, corticosterone, or GABA. We further found that intestinal epithelial cells expressed GABA receptor subunits and mediated mitogen-activated protein kinase signaling upon GABA stimulation. In addition, administration of TMC3115 induced mitogen-activated protein kinase signaling in colonic epithelial cells but not in small intestinal epithelial cells in mice. These results indicate that GABA produced by the gut microbiota, mainly in the colon, may affect host behavioral characteristics via GABA receptors expressed in intestinal epithelial cells without being transferred to the blood. This study suggests a novel mechanism by which intestinal GABA exerts physiological effects, even in the presence of the blood-brain barrier.
Asunto(s)
Ansiedad , Células Epiteliales , Microbioma Gastrointestinal , Ácido gamma-Aminobutírico , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Ácido gamma-Aminobutírico/metabolismo , Ansiedad/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Neomicina/farmacología , Ratones Endogámicos C57BL , Receptores de GABA/metabolismo , Bifidobacterium/metabolismo , Probióticos/farmacología , HumanosRESUMEN
Human milk oligosaccharides (HMOs) are essentially unaffected by the digestive enzymes of the nursling and are known for their ability to enrich certain microbial species in the infant gut microbiota, in particular bifidobacteria. HMO metabolism has been studied in various bifidobacterial species such as B. breve, B. bifidum, and B. longum subsp. infantis. In the current study, we describe differential growth abilities elicited by twenty-three newly isolated Bifidobacterium pseudocatenulatum strains on particular HMOs, such as 2'-fucosyllactose (2'FL), 3-fucosyllactose (3FL), lacto-N-tetraose (LNT), and lacto-N-neotetraose (LNnT). Through gene-trait matching and comparative genome analysis, we identified genes involved in the degradation of fucosylated HMOs in this strain set, while we employed a transcriptomic approach to facilitate the identification and characterization of genes and associated enzymes involved in LNT metabolism by strain B. pseudocatenulatum MM0196. A total of 252 publicly available genomes of the B. pseudocatenulatum taxon were screened for homologs of the glycosyl hydrolases (GHs) identified here as being required for selected HMO metabolism. From this analysis, it is clear that all members of this species possess homologs of the genes involved in LNT degradation, while genes required for degradation of fucosylated HMOs are variably present.IMPORTANCEOur findings allow a better understanding of the complex interaction between Bifidobacterium and its host and provide a roadmap toward future applications of B. pseudocatenulatum as a probiotic with a focus on infant health. Furthermore, our investigations have generated information on the role of HMOs in shaping the infant gut microbiota, thus also facilitating applications of HMOs in infant nutrition, with potential extension into the mature or adult gut microbiota. Supplementation of HMOs is known to result in the modulation of bacterial communities toward a higher relative abundance of bifidobacteria, which in turn enforces their ability to modulate particular immune functions and strengthen the intestinal barrier. This work may therefore inspire future studies to improve the formulation of neonatal nutritional products, aimed at facilitating the development of a healthy digestive and immune system and reducing the differences in gut microbiota composition observed between breastfed and formula-fed babies or full-term and preterm infants.
Asunto(s)
Bifidobacterium pseudocatenulatum , Leche Humana , Oligosacáridos , Leche Humana/química , Oligosacáridos/metabolismo , Humanos , Bifidobacterium pseudocatenulatum/genética , Bifidobacterium pseudocatenulatum/metabolismo , Genoma Bacteriano , Microbioma Gastrointestinal , Trisacáridos/metabolismo , Bifidobacterium/genética , Bifidobacterium/metabolismoRESUMEN
Global concerns over folate deficiency, the risks of excessive synthetic folic acid consumption, and food loss implications for environmental sustainability and food security drive needs of innovative approaches that align food by-product valorisation with folate bio-enrichment. This study explored the use of three fruit by-products extracts (grape, passion fruit, and pitaya) and whey to develop a folate bio-enriched fermented whey-based beverage. Three strains (Lacticaseibacillus rhamnosus LGG, Bifidobacterium infantis BB-02, and Streptococcus thermophilus TH-4) were tested for folate production in different fermentation conditions in modified MRS medium and in a whey-based matrix prepared with water extracts of these fruit by-products. B. infantis BB-02 and S. thermophilus TH-4, alone and in co-culture, were the best folate producers. The selection of cultivation conditions, including the presence of different substrates and pH, with grape by-product water extract demonstrating the most substantial effect on folate production among the tested extracts, was crucial for successfully producing a biofortified fermented whey-based beverage (FWBB). The resulting FWBB provided 40.7 µg of folate per 100 mL after 24 h of fermentation at 37 °C, effectively leveraging food by-products. Moreover, the beverage showed no cytotoxicity in mouse fibroblast cells tests. This study highlights the potential for valorising fruit by-products and whey for the design of novel bioenriched foods, promoting health benefits and contributing to reduced environmental impact from improper disposal.
Asunto(s)
Fermentación , Ácido Fólico , Frutas , Suero Lácteo , Animales , Frutas/química , Ratones , Humanos , Suero Lácteo/química , Bebidas/microbiología , Streptococcus thermophilus/metabolismo , Streptococcus thermophilus/crecimiento & desarrollo , Lacticaseibacillus rhamnosus/metabolismo , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Bifidobacterium/metabolismo , Bifidobacterium/crecimiento & desarrollo , Vitis/químicaRESUMEN
The microbial ecology of raw milk cheeses is determined by bacteria originating from milk and milk-producing animals. Recently, it has been shown that members of the Bifidobacterium mongoliense species may become transmitted along the Parmigiano Reggiano cheese production chain and ultimately may colonize the consumer intestine. However, there is a lack of knowledge regarding the molecular mechanisms that mediate the interaction between B. mongoliense and the human gut. Based on 128 raw milk cheeses collected from different Italian regions, we isolated and characterized 10 B. mongoliense strains. Comparative genomics allowed us to unveil the presence of enzymes required for the degradation of sialylated host-glycans in B. mongoliense, corroborating the appreciable growth on de Man-Rogosa-Sharpe (MRS) medium supplemented with 3'-sialyllactose (3'-SL) or 6'-sialyllactose (6'-SL). The B. mongoliense BMONG18 was chosen, due to its superior ability to utilize 3'-SL and mucin as representative strain, to investigate its behavior when co-inoculated with other bifidobacterial species. Conversely, members of other bifidobacterial species did not appear to benefit from the presence of BMONG18, highlighting a competitive scenario for nutrient acquisition. Transcriptomic data of BMONG18 reveal no significant differences in gene expression when cultivated in a gut simulating medium (GSM), regardless of whether cheese was included or not. Furthermore, BMONG18 was shown to exhibit high adhesion capabilities to HT29-MTX human cells, in line with its colonization ability of a human host.IMPORTANCEFermented foods are nourishments produced through controlled microbial growth that play an essential role in worldwide human nutrition. Research interest in fermented foods has increased since the 80s, driven by growing awareness of their potential health benefits beyond mere nutritional content. Bifidobacterium mongoliense, previously identified throughout the production process of Parmigiano Reggiano cheese, was found to be capable of establishing itself in the intestines of its consumers. Our study underscores molecular mechanisms through which this bifidobacterial species, derived from food, interacts with the host and other gut microbiota members.
Asunto(s)
Bifidobacterium , Queso , Microbioma Gastrointestinal , Leche , Queso/microbiología , Bifidobacterium/genética , Bifidobacterium/metabolismo , Bifidobacterium/crecimiento & desarrollo , Humanos , Leche/microbiología , Animales , ItaliaRESUMEN
Prenatal stress (PNS) profoundly impacts maternal and offspring health, with enduring effects including microbiome alterations, neuroinflammation, and behavioral disturbances such as reductions in social behavior. Converging lines of evidence from preclinical and clinical studies suggest that PNS disrupts tryptophan (Trp) metabolic pathways and reduces gut Bifidobacteria, a known beneficial bacterial genus that metabolizes Trp. Specifically, previous work from our lab demonstrated that human prenatal mood disorders in mothers are associated with reduced Bifidobacterium dentium in infants at 13 months. Given that Bifidobacterium has been positively associated with neurodevelopmental and other health benefits and is depleted by PNS, we hypothesized that supplementing PNS-exposed pregnant dams with B. dentium would ameliorate PNS-induced health deficits. We measured inflammatory outputs, Trp metabolite levels and enzymatic gene expression in dams and fetal offspring, and social behavior in adult offspring. We determined that B. dentium reduced maternal systemic inflammation and fetal offspring neuroinflammation, while modulating tryptophan metabolism and increasing kynurenic acid and indole-3-propionic acid intergenerationally. Additional health benefits were demonstrated by the abrogation of PNS-induced reductions in litter weight. Finally, offspring of the B. dentium cohort demonstrated increased sociability in males primarily and increased social novelty primarily in females. Together these data illustrate that B. dentium can orchestrate interrelated host immune, metabolic and behavioral outcomes during and after gestation for both dam and offspring and may be a candidate for prevention of the negative sequelae of stress.
Asunto(s)
Inflamación , Efectos Tardíos de la Exposición Prenatal , Conducta Social , Triptófano , Femenino , Embarazo , Animales , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/microbiología , Triptófano/metabolismo , Inflamación/metabolismo , Masculino , Bifidobacterium/metabolismo , Estrés Psicológico/metabolismo , Microbioma Gastrointestinal/fisiología , Conducta Animal/fisiología , Probióticos/farmacología , RatasRESUMEN
This study investigated the structural changes of resistant starch (RS) derived from autoclaved lentil starch (ALRS) and untreated lentil starch (ULRS) during in vitro colonic fermentation, as well as their regulatory effects on the composition of the intestinal microbiota. Following in vitro fermentation, both RS samples exhibited a progressive decrease in molecular weight and a gradual increase in double helix/order. Bifidobacterium was more abundant in ULRS during the initial period of fermentation, while ALRS showed higher abundance in the later stage. ALRS demonstrated greater production of short-chain fatty acids (SCFAs) compared to ULRS, likely attributed to its higher structural order and faster fermentation pattern. The distinct surface morphologies of ULRS and ALRS played a crucial role in determining the accessibility of RS substrates for microbial fermentation. These different structural patterns also influenced the shifts in microbial composition in fecal cultures, leading to variations in SCFAs production through anaerobic fermentation.
Asunto(s)
Ácidos Grasos Volátiles , Fermentación , Microbioma Gastrointestinal , Lens (Planta) , Almidón , Almidón/metabolismo , Almidón/química , Ácidos Grasos Volátiles/metabolismo , Lens (Planta)/metabolismo , Lens (Planta)/química , Lens (Planta)/microbiología , Bacterias/metabolismo , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Humanos , Heces/microbiología , Bifidobacterium/metabolismo , Bifidobacterium/crecimiento & desarrolloRESUMEN
Bacteriocins are broad or narrow-spectrum antimicrobial compounds that have received significant scientific attention due to their potential to treat infections caused by antibiotic-resistant pathogenic bacteria. The genome of Bifidobacterium pseudocatenulatum MM0196, an antimicrobial-producing, fecal isolate from a healthy pregnant woman, was shown to contain a gene cluster predicted to encode Pseudocin 196, a novel lantibiotic, in addition to proteins involved in its processing, transport and immunity. Following antimicrobial assessment against various indicator strains, protease-sensitive Pseudocin 196 was purified to homogeneity from cell-free supernatant. MALDI TOF mass spectrometry confirmed that the purified antimicrobial compound corresponds to a molecular mass of 2679 Da, which is consistent with that deduced from its genetic origin. Pseudocin 196 is classified as a lantibiotic based on its similarity to lacticin 481, a lanthionine ring-containing lantibiotic produced by Lactococcus lactis. Pseudocin 196, the first reported bacteriocin produced by a B. pseudocatenulatum species of human origin, was shown to inhibit clinically relevant pathogens, such as Clostridium spp. and Streptococcus spp. thereby highlighting the potential application of this strain as a probiotic to treat and prevent bacterial infections.
Asunto(s)
Antibacterianos , Bacteriocinas , Bifidobacterium , Bacteriocinas/farmacología , Bacteriocinas/genética , Bacteriocinas/metabolismo , Bacteriocinas/química , Humanos , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/metabolismo , Bifidobacterium/genética , Bifidobacterium/efectos de los fármacos , Bifidobacterium/metabolismo , Femenino , Clostridium/genética , Clostridium/efectos de los fármacos , Clostridium/metabolismo , Heces/microbiología , Streptococcus/efectos de los fármacos , Streptococcus/genética , Streptococcus/metabolismo , Embarazo , Familia de Multigenes , Pruebas de Sensibilidad Microbiana , Genoma Bacteriano , Probióticos/farmacologíaRESUMEN
BACKGROUND AND PURPOSE: Colorectal cancer (CRC) is a widespread malignancy with a complex and not entirely elucidated pathogenesis. This study aims to explore the role of Bifidobacterium in the urea cycle (UC) and its influence on the progression of CRC, a topic not extensively studied previously. EXPERIMENTAL APPROACH: Utilizing both bioinformatics and experimental methodologies, this research involved analyzing bacterial abundance in CRC patients in comparison to healthy individuals. The study particularly focused on the abundance of BA. Additionally, transcriptomic data analysis and cellular experiments were conducted to investigate the impact of Bifidobacterium on ammonia metabolism and mitochondrial function, specifically examining its regulation of the key UC gene, ALB. KEY RESULTS: The analysis revealed a significant decrease in Bifidobacterium abundance in CRC patients. Furthermore, Bifidobacterium was found to suppress ammonia metabolism and induce mitochondrial dysfunction through the regulation of the ALB gene, which is essential in the context of UC. These impacts contributed to the suppression of CRC cell proliferation, a finding corroborated by animal experimental results. CONCLUSIONS AND IMPLICATIONS: This study elucidates the molecular mechanism by which Bifidobacterium impacts CRC progression, highlighting its role in regulating key metabolic pathways. These findings provide potential targets for novel therapeutic strategies in CRC treatment, emphasizing the importance of microbiota in cancer progression.
Asunto(s)
Bifidobacterium , Neoplasias Colorrectales , Urea , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Bifidobacterium/metabolismo , Humanos , Urea/metabolismo , Animales , Proliferación Celular , Amoníaco/metabolismo , Ratones , Mitocondrias/metabolismo , Línea Celular Tumoral , Masculino , Microbioma Gastrointestinal/fisiología , FemeninoRESUMEN
To enhance health benefits, a probiotic can be co-administered with a metabolizable prebiotic forming a synergistic synbiotic. We assessed the synergies resulting from combining Bifidobacterium longum subsp. infantis LMG 11588 and an age-adapted blend of six human milk oligosaccharides (HMOs) in ex vivo colonic incubation bioreactors seeded with fecal background microbiota from infant and toddler donors. When HMOs were combined with B. infantis LMG 11588, they were rapidly and completely consumed. This resulted in increased short chain fatty acid (SCFA) production compared to the summed SCFA production from individual ingredients (synergy). Remarkably, HMOs were partially consumed for specific infant donors in the absence of B. infantis LMG 11588, yet all donors showed increased SCFA production upon B. infantis LMG 11588 supplementation. We found specific bacterial taxa associated with the differential response pattern to HMOs. Our study shows the importance of carefully selecting pre- and probiotic into a synergistic synbiotic that could benefit infants.
Asunto(s)
Bifidobacterium longum subspecies infantis , Ácidos Grasos Volátiles , Leche Humana , Oligosacáridos , Humanos , Leche Humana/metabolismo , Leche Humana/química , Oligosacáridos/metabolismo , Ácidos Grasos Volátiles/metabolismo , Lactante , Bifidobacterium longum subspecies infantis/metabolismo , Bifidobacterium/metabolismo , Probióticos/administración & dosificación , Microbioma Gastrointestinal , Heces/microbiología , Femenino , Simbióticos/administración & dosificación , PreescolarRESUMEN
The honeybee gut microbiome is crucial for degrading diverse pollen glycans. Yet it is unclear how this process shapes the interactions among bacteria. Here, we demonstrate a conditional mutualistic interaction between strains of two honeybee gut bacteria Bifidobacterium asteroides and Gilliamella apicola. When co-occurring in vitro and in vivo, Bifidobacterium provides complementary demethylation service to promote Gilliamella growth on methylated homogalacturonan, an enriched polysaccharide of pectin. In exchange, Gilliamella shares digestive products with Bifidobacterium, through which a positive interaction is established. This positive interaction vanishes when Bifidobacterium is not required on a non-methylated diet. Results from biochemical and gene expression analyses combined with model simulation further suggest that the ratio change of the two major homogalacturonan breakdown products, galacturonic acid (GalA) and di-GalA, determines the bacterial interaction. This study unravels how glycan metabolism may shape the interactions between honeybee gut bacteria.
Asunto(s)
Bifidobacterium , Microbioma Gastrointestinal , Pectinas , Simbiosis , Abejas/microbiología , Animales , Pectinas/metabolismo , Microbioma Gastrointestinal/fisiología , Simbiosis/fisiología , Bifidobacterium/metabolismo , Bifidobacterium/genética , Polisacáridos/metabolismo , Ácidos HexurónicosRESUMEN
(1) Background: Recently, academic studies are demonstrating that the cholesterol-lowering effects of pectin oligosaccharides (POSs) are correlated to intestinal flora. However, the mechanisms of POS on cholesterol metabolisms are limited, and the observations of intestinal flora are lacking integrative analyses. (2) Aim and methods: To reveal the regulatory mechanisms of POS on cholesterol metabolism via an integrative analysis of the gut microbiota, the changes in gut microbiota structure and metabolite composition after POS addition were investigated using Illumina MiSeq sequencing and non-targeted metabolomics through in vitro gut microbiota fermentation. (3) Results: The composition of fecal gut flora was adjusted positively by POS. POS increased the abundances of the cholesterol-related bacterial groups Bacteroidetes, Bifidobacterium and Lactobacillus, while it decreased conditional pathogenic Escherichia coli and Enterococcus, showing good prebiotic activities. POS changed the composition of gut microbiota fermentation metabolites (P24), causing significant changes in 221 species of fermentation metabolites in a non-targeted metabolomics analysis and promoting the production of short-chain fatty acids. The abundances of four types of cholesterol metabolism-related metabolites (adenosine monophosphate, cyclic adenosine monophosphate, guanosine and butyrate) were significantly higher in the P24 group than those in the control group without POS addition. (4) Conclusion: The abovementioned results may explain the hypocholesterolemic effects of POS and promotion effects on cholesterol efflux of P24. These findings indicated that the potential regulatory mechanisms of citrus POS on cholesterol metabolism are modulated by cholesterol-related gut microbiota and specific metabolites.
Asunto(s)
Colesterol , Heces , Fermentación , Microbioma Gastrointestinal , Oligosacáridos , Pectinas , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Pectinas/farmacología , Pectinas/metabolismo , Colesterol/metabolismo , Oligosacáridos/farmacología , Heces/microbiología , Humanos , Prebióticos , Masculino , Metabolómica , Ácidos Grasos Volátiles/metabolismo , Bifidobacterium/metabolismo , Bifidobacterium/efectos de los fármacos , Femenino , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Bacterias/clasificación , CitrusRESUMEN
Mangosteen (Garcinia mangostana L.) is a tasty, polyphenol-rich tropical fruit. The edible part is highly appreciated by its aroma, taste and texture. The non-edible part, rich in polyphenols, has been traditionally used in Thai medicine. In this work, flavonoids and phenolic acid/derivatives were identified in mangosteen extracts (ME) from edible and non-edible portions. We first studied the effects of MEs on the growth, metabolism, antioxidant capacity, biofilm formation and antimicrobial capacity of eight bifidobacteria and lactobacilli strains from intestinal origin and two commercial probiotic strains (BB536 and GG). ME concentrations higher than 10-20 % were inhibitory for all strains. However, ME concentrations of 5 % significantly (P < 0.01) increased all strains antioxidant capacity, reduced biofilm-formation, and enhanced inhibition against Gram-positive pathogens. To apply these knowledge, bifunctional fermented milk products were elaborated with 5 % ME and individual strains, which were selected taking into account their growth with ME, and the widest range of values on antioxidant capacity, biofilm formation and antimicrobial activity (bifidobacteria INIA P2 and INIA P467, lactobacilli INIA P459 and INIA P708, and reference strain GG). Most strains survived well manufacture, refrigerated storage and an in vitro simulation of major conditions encountered in the gastrointestinal tract. As expected, products supplemented with ME showed higher polyphenol content and antioxidant capacity levels than control. After sensory evaluation, products containing strains INIA P2, INIA P708 and GG outstood as best.
Asunto(s)
Antioxidantes , Biopelículas , Productos Lácteos Cultivados , Garcinia mangostana , Lactobacillus , Extractos Vegetales , Extractos Vegetales/farmacología , Garcinia mangostana/química , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Antioxidantes/farmacología , Lactobacillus/efectos de los fármacos , Lactobacillus/metabolismo , Productos Lácteos Cultivados/microbiología , Bifidobacterium/efectos de los fármacos , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Probióticos , Flavonoides/farmacología , Flavonoides/análisis , Humanos , Frutas/química , Frutas/microbiología , Fermentación , Hidroxibenzoatos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Polifenoles/farmacologíaRESUMEN
Synbiotics are complex preparations of prebiotics that can be selectively utilized by live microorganisms to improve host health. Synbiotics are divided into complementary synbiotics, which consist of probiotics and prebiotics with independent functions, and synergistic synbiotics, which consist of prebiotics that are selectively used by gut microorganisms. Complementary synbiotics used in human clinical trials include Lactobacillus spp. and Bifidobacterium spp. as probiotics, and fructooligosaccharides, galactooligosaccharides, and inulin as prebiotics. Over the past five years, synbiotics have been most commonly used in patients with metabolic disorders, including obesity, and immune and gastrointestinal disorders. Several studies have observed alterations in the microbial community; however, these changes did not lead to significant improvements in disease outcomes or biochemical and hematological markers. The same synbiotics have been applied to individuals with different gut environments. As a result, even with the same synbiotics, there are non-responders who do not respond to the applied synbiotics due to the different intestinal environment for each individual. Therefore, to obtain meaningful results, applying different synbiotics depending on the individual is necessary. Synergistic synbiotics are one solution to circumvent this problem, as they combine elements that can effectively improve health, even in non-responders. This review aims to explain the concept of synbiotics, highlight recent human clinical trials, and explore the current state of research on synergistic synbiotics.
Asunto(s)
Microbioma Gastrointestinal , Promoción de la Salud , Prebióticos , Probióticos , Simbióticos , Humanos , Promoción de la Salud/métodos , Manejo de la Enfermedad , Oligosacáridos/metabolismo , Bifidobacterium/metabolismo , Lactobacillus/metabolismo , Obesidad/terapia , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/terapiaRESUMEN
Extracellular vesicles (EVs) are nanoparticles secreted by various organisms. Methods for modifying EVs functionally have garnered attention for developing EV-based therapeutic systems. However, most technologies used to integrate these functions are limited to mammalian-derived EVs and a promising modification method for bacteria-derived EVs has not yet been developed. In this study, we propose a novel method for the versatile functionalization of immunostimulatory probiotic Bifidobacteria-derived EVs (B-EVs) using amino acid metabolic labeling and azide-alkyne click reaction. Azide D-alanine (ADA), a similar molecule to D-alanine in bacteria cell-wall peptidoglycan, was selected as an azide group-functionalized amino acid. Azide-modified B-EVs were isolated from Bifidobacteria incubated with ADA. The physicochemical and compositional characteristics, as well as adjuvanticity of B-EVs against immune cells were not affected by azide loading, demonstrating that this functionalization approach can retain the endogenous usefulness of B-EVs. By using the fluorescent B-EVs obtained by this method, the intracellular trafficking of B-EVs after uptake by immune cells was successfully observed. Furthermore, this method enabled the formulation of B-EVs for hydrogelation and enhanced adjuvanticity in the host. Our findings will be helpful for further development of EV-based immunotherapy.
Asunto(s)
Azidas , Bifidobacterium , Química Clic , Vesículas Extracelulares , Inmunoterapia , Vesículas Extracelulares/metabolismo , Bifidobacterium/metabolismo , Azidas/química , Animales , Inmunoterapia/métodos , Alanina/química , Probióticos/administración & dosificación , Ratones , Aminoácidos/química , Aminoácidos/metabolismo , Humanos , Células RAW 264.7RESUMEN
The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE: By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.
Asunto(s)
Bifidobacterium , Vesículas Extracelulares , Limosilactobacillus reuteri , Probióticos , Limosilactobacillus reuteri/metabolismo , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/crecimiento & desarrollo , Vesículas Extracelulares/metabolismo , Humanos , Bifidobacterium/metabolismo , Bifidobacterium/genética , Bifidobacterium/crecimiento & desarrolloRESUMEN
BACKGROUND: Kratom (Mitragyna speciosa) has a long history of traditional use. It contains various alkaloids and polyphenols. The properties of kratom's alkaloids have been well-documented. However, the property of kratom's polyphenols in water-soluble phase have been less frequently reported. This study assessed the effects of water-soluble Mitragyna speciosa (kratom) extract (MSE) on gut microbiota and their metabolite production in fecal batch culture. RESULTS: The water-soluble kratom extract (MSE0) and the water-soluble kratom extract after partial sugar removal (MSE50) both contained polyphenols, with total phenolic levels of 2037.91 ± 51.13 and 3997.95 ± 27.90 mg GAE/g extract, respectively and total flavonoids of 81.10 ± 1.00 and 84.60 ± 1.43 mg CEQ/g extract. The gut microbiota in fecal batch culture was identified by 16S rRNA gene sequencing at 0 and 24 h of fermentation. After fermentation, MSE50 stimulated the growth of Bifidobacterium more than MSE0. MSE0 gave the highest total fatty acids level among the treatments. The phenolic metabolites produced by some intestinal microbiota during fecal fermentation at 24 h were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The major metabolite of biotransformation of both water-soluble MSEs by intestinal microbiota was pyrocatechol (9.85-11.53%). CONCLUSION: The water-soluble MSEs and their produced metabolites could potentially be used as ingredients for functional and medicinal food production that supports specific gut microbiota. © 2024 Society of Chemical Industry.
Asunto(s)
Heces , Fermentación , Microbioma Gastrointestinal , Mitragyna , Extractos Vegetales , Polifenoles , Microbioma Gastrointestinal/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/química , Extractos Vegetales/metabolismo , Heces/microbiología , Mitragyna/química , Mitragyna/metabolismo , Polifenoles/metabolismo , Polifenoles/farmacología , Bacterias/clasificación , Bacterias/genética , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Bacterias/aislamiento & purificación , Humanos , Masculino , Flavonoides/metabolismo , Flavonoides/farmacología , Bifidobacterium/metabolismo , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/efectos de los fármacosRESUMEN
Through adaptive laboratory evolution (ALE) of Sphingomonas sp. ATCC 31555, fermentation for production of low-molecular-weight welan gum (LMW-WG) was performed using glycerol as sole carbon source. During ALE, GPC-MALS analysis revealed a gradual decrease in WG molecular weight with the increase of adaptation cycles, accompanied by changes in solution conformation. LMW-WG was purified and structurally analyzed using GPC-MALS, monosaccharide composition analysis, infrared spectroscopy, NMR analysis, atomic force microscopy, and scanning electron microscopy. Subsequently, LMW-WG obtains hydration, transparency, antioxidant activity, and rheological properties. Finally, an in vitro simulation colon reactor was used to evaluate potential prebiotic properties of LMW-WG as dietary fiber. Compared with WG produced using sucrose as substrate, LMW-WG exhibited a fourfold reduction in molecular weight while maintaining moderate viscosity. Structurally, L-Rha nearly completely replaced L-Man. Furthermore, LMW-WG demonstrated excellent hydration, antioxidant activity, and high transparency. It also exhibited resistance to saliva and gastrointestinal digestion, showcasing a favorable colonization effect on Bifidobacterium, making it a promising symbiotic agent.
Asunto(s)
Antioxidantes , Fermentación , Glicerol , Peso Molecular , Sphingomonas , Glicerol/química , Glicerol/metabolismo , Antioxidantes/química , Antioxidantes/farmacología , Sphingomonas/metabolismo , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/farmacología , Viscosidad , Prebióticos , Bifidobacterium/metabolismoRESUMEN
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.