Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.349
Filtrar
1.
NPJ Biofilms Microbiomes ; 10(1): 59, 2024 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-39034349

RESUMEN

The dominant bacteria in the hindgut of calves play an important role in their growth and health, which could even lead to lifelong consequences. However, the identification of core probiotics in the hindgut and its mechanism regulating host growth remain unclear. Here, a total of 1045 fecal samples were analyzed by 16S rRNA gene sequencing from the 408 Holstein dairy calves at the age of 0, 14, 28, 42, 56, and 70 days to characterize the dynamic changes of core taxa. Moreover, the mechanisms of nutrient metabolism of calf growth regulated by core bacteria were investigated using multi-omics analyses. Finally, fecal microbiota transplantation (FMT) in mice were conducted to illustrate the potential beneficial effects of core bacteria. Four calf enterotypes were identified and enterotypes dominated by Bifidobacterium and Oscillospiraceae_UCG-005 were representative. The frequency of enterotype conversion shifted from variable to stable. The close relationship observed between phenotype and enterotype, revealing a potential pro-growth effect of Bifidobacterium, might be implemented by promoting the use of carbohydrate, activating the synthesis of volatile fatty acids, amino acids and vitamin B6, and inhibiting methane production in the hindgut. The FMT results indicated the beneficial effect of Bifidobacterium on host growth and hindgut development. These results support the notion that the Bifidobacterium-dominated fecal microbiome would be an important driving force for promoting the host growth in the early life. Our findings provide new insights into the potential probiotic mining and application strategies to promote the growth of young animals or improve their growth retardation.


Asunto(s)
Bifidobacterium , Trasplante de Microbiota Fecal , Heces , Microbioma Gastrointestinal , ARN Ribosómico 16S , Animales , Heces/microbiología , Bovinos , ARN Ribosómico 16S/genética , Bifidobacterium/genética , Bifidobacterium/crecimiento & desarrollo , Ratones , Trasplante de Microbiota Fecal/métodos , Fenotipo , Probióticos/administración & dosificación , Filogenia , ADN Bacteriano/genética
2.
Food Res Int ; 191: 114720, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39059916

RESUMEN

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/farmacología
3.
Molecules ; 29(12)2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38930817

RESUMEN

With the aim to obtain controlled-release systems and to preserve the antioxidant, immunomodulatory, and prebiotic activity of the bioactive compounds, microencapsulation of both honeydew honey and royal jelly into biopolymeric microparticles based on rye bran heteropolysaccharides (HPS) was successfully performed. Honeydew honey and royal jelly microcapsules were prepared by spray-drying method and were characterized in terms of morphology and biological properties. Due to the resistance of the obtained encapsulates to the acidic pH in the stomach and digestive enzymes, the microcapsules showed prebiotic properties positively influencing both the growth, retardation of the dying phase, and the pro-adhesive properties of probiotic bacteria, i.e., Bifidobacterium spp. and lactic acid bacteria. Moreover, as a result of fermentation of the microcapsules of bee products in the lumen of the large intestine, an increased synthesis of short-chain fatty acids, i.e., butyric acid, was found on average by 39.2% in relation to the SCFA concentrations obtained as a result of fermentation of native bee products, thus opening new perspectives for the exploitation of honeydew honey and royal jelly loaded microcapsules for nutraceutical applications.


Asunto(s)
Cápsulas , Microbioma Gastrointestinal , Miel , Prebióticos , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Abejas , Ácidos Grasos Volátiles/metabolismo , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Fermentación , Probióticos , Ácidos Grasos
4.
Appl Environ Microbiol ; 90(7): e0024724, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-38888338

RESUMEN

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 & desarrollo
5.
Microbiol Res ; 285: 127741, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38761487

RESUMEN

Fructooligosaccharides (FOS) are a common prebiotic widely used in functional foods. Meanwhile, Saccharomyces boulardii is a fungal probiotic frequenly used in the clinical treatment of diarrhea. Compared with single use, the combination of prebiotics and probiotics as symbiotics may be more effective in regulating gut microbiota as recently reported in the literature. The present study aimed to investigate the effects of FOS, S. boulardii and their combination on the structure and metabolism of the gut microbiota in healthy primary and secondary school students using an in vitro fermentation model. The results indicated that S. boulardii alone could not effectively regulate the community structure and metabolism of the microbiota. However, both FOS and the combination of FOS and S. boulardii could effectively regulate the microbiota, significantly inhibiting the growth of Escherichia-Shigella and Bacteroides, and controlling the production of the gases including H2S and NH3. In addition, both FOS and the combination could significantly promote the growth of Bifidobacteria and Lactobacillus, lower environmental pH, and enhance several physiological functions related to synthesis and metabolism. Nevertheless, the combination had more unique benefits as it promoted the growth of Lactobacillus, significantly increased CO2 production and enhanced the functional pathways of carbon metabolism and pyruvic acid metabolism. These findings provide guidance for clinical application and a theoretical basis for the development of synbiotic preparations.


Asunto(s)
Fermentación , Microbioma Gastrointestinal , Oligosacáridos , Prebióticos , Probióticos , Saccharomyces boulardii , Estudiantes , Oligosacáridos/metabolismo , Oligosacáridos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Saccharomyces boulardii/metabolismo , Humanos , Probióticos/metabolismo , Niño , Masculino , Adolescente , Femenino , Lactobacillus/metabolismo , Lactobacillus/crecimiento & desarrollo , Bacterias/metabolismo , Bacterias/clasificación , Heces/microbiología , Bifidobacterium/metabolismo , Bifidobacterium/crecimiento & desarrollo
6.
Food Chem ; 453: 139644, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-38761735

RESUMEN

This work developed and characterized the physicochemical properties of a type A gelatin and amidated low-methoxyl pectin complex coacervate (GA-LMAP-CC) hydrogel and evaluated its suitability for preserving the viability of probiotics under in vitro gastrointestinal conditions. The formation of GA-LMAP-CC was achieved via height electrostatic attraction at pH 3 and a mixing ratio of 1, exhibiting thermoreversible gel behavior. The hydrogel had a porosity of 44% and a water absorption capacity of up to 12 times. Water absorption profiles were obtained at different pH values (2, 5, and 7). The influence of GA-LMAP-CC depended on the medium, which controlled the hydration and water absorption rate. GA-LMAP-CC promoted the viability of B. longum BB536 and L. acidophilus strains under simulated gastrointestinal conditions, thereby enhancing their potential for intestinal colonization. The hydrogel has suitable properties for potential application in food and pharmaceutical areas to encapsulate and preserve probiotics.


Asunto(s)
Gelatina , Hidrogeles , Pectinas , Probióticos , Pectinas/química , Gelatina/química , Probióticos/química , Hidrogeles/química , Viabilidad Microbiana/efectos de los fármacos , Lactobacillus acidophilus/química , Lactobacillus acidophilus/crecimiento & desarrollo , Lactobacillus acidophilus/metabolismo , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Concentración de Iones de Hidrógeno , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología
7.
Food Res Int ; 187: 114417, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38763667

RESUMEN

Resistant starch serves as a prebiotic in the large intestine, aiding in the maintenance of a healthy intestinal environment and mitigating associated chronic illnesses. This study aimed to investigate the impact of resistant starch-enriched brown rice (RBR) on intestinal health and functionality. We assessed changes in resistant starch concentration, structural alterations, and branch chain length distribution throughout the digestion process using an in vitro model. The efficacy of RBR in the intestinal environment was evaluated through analyses of its prebiotic potential, effects on intestinal microbiota, and intestinal function-related proteins in obese animals fed a high-fat diet. RBR exhibited a higher yield of insoluble fraction in both the small and large intestines compared to white and brown rice. The total digestible starch content decreased, while the resistant starch content significantly increased during in vitro digestion. Furthermore, RBR notably enhanced the growth of four probiotic strains compared to white and brown rice, displaying higher proliferation activity than the positive control, FOS. Notably, consumption of RBR by high-fat diet-induced obese mice suppressed colon shortening, increased Bifidobacteria growth, and improved intestinal permeability. These findings underscore the potential prebiotic and gut health-promoting attributes of RBR, offering insights for the development of functional foods aimed at preventing gastrointestinal diseases.


Asunto(s)
Dieta Alta en Grasa , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Obesidad , Oryza , Prebióticos , Almidón , Animales , Oryza/química , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Almidón/metabolismo , Masculino , Obesidad/metabolismo , Ratones Obesos , Almidón Resistente , Probióticos , Digestión , Bifidobacterium/crecimiento & desarrollo
8.
J Food Sci ; 89(5): 2546-2556, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38578148

RESUMEN

2'-Fucosyllactose (2'-FL) is postulated to provide health benefits and promote the growth of probiotics. This work was undertaken to study the effects of 2'-FL on the viability of starter cultures and Bifidobacterium strains of human origin in yogurt during refrigerated storage. Yogurts were produced containing 2'-FL (0 or 2 g/L) and Bifidobacterium strains of human origin (Bifidobacterium longum subsp. longum BB536 or Bifidobacterium longum subsp. infantis ATCC 15697) at a concentration of at least 109 CFU/mL. All yogurts were stored at 4°C for 5 weeks. Results showed that 2'-FL was stable in yogurts for at least 5 weeks of cold storage, and the addition of 2'-FL did not significantly alter yogurt fermentation parameters, associated metabolites, and the viability of mixed yogurt starter cultures and Bifidobacterium strains (p > 0.05). The addition of bifidobacteria had a negative impact (p < 0.05) on the survival rate of starter cultures, Streptococcus thermophilus and Lactobacillus delbureckii subsp. bulgaricus. Meanwhile, it is difficult to maintain a high survival rate of bifidobacteria in final yogurt products, and the addition of 2'-FL could not enhance the viability of bifidobacteria. B. longum BB536 survived at a level higher than 106 CFU/g for 28 days, while B. infantis ATCC15697 maintained this level for only 7 days. In summary, this study has shown the impact of 2'-FL and bifidobacterial species on yogurt properties, and results suggest that it is promising to use 2'-FL in yogurt products as a prebiotic. PRACTICAL APPLICATION: Yogurt is known for its beneficial effects on human health and nutrition. This study reported the production of symbiotic yogurt containing bifidobacteria and 2'-fucosyllactose (2'-FL) as a functional food for specified health uses. The viability of yogurt starter cultures and probiotic bifidobacterial strains was analyzed in this study. Moreover, this research demonstrated that 2'-FL could be added to yogurt without affecting the characteristics of yogurt significantly.


Asunto(s)
Bifidobacterium , Fermentación , Almacenamiento de Alimentos , Probióticos , Trisacáridos , Yogur , Yogur/microbiología , Trisacáridos/farmacología , Bifidobacterium/crecimiento & desarrollo , Humanos , Almacenamiento de Alimentos/métodos , Refrigeración , Streptococcus thermophilus/crecimiento & desarrollo , Viabilidad Microbiana , Microbiología de Alimentos , Recuento de Colonia Microbiana
9.
Sci Rep ; 12(1): 1432, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35082322

RESUMEN

Faecal (FM) and colon mucosal associated microbiota (MAM) were studied in a model of colorectal cancer (CRC), the Apc-mutated Pirc rats, and in age-paired wt F344 rats. Principal Coordinates Analysis indicated that samples' distribution was driven by age, with samples of young rats (1 month old; without tumours) separated from older ones (11-month-old; bearing tumours). Diversity analysis showed significant differences between FM and MAM in older Pirc rats, and between MAM of both Pirc and wt rats and the tumour microbiota, enriched in Enterococcus, Escherichia/Shigella, Proteus and Bifidobacteriaceae. In young animals, Pirc FM was enriched in the genus Delftia, while wt FM was enriched in Lactobacillus and Streptococcus. Some CRC biomarkers and faecal short chain fatty acids (SCFAs) were also measured. Colon proliferation and DClK1 expression, a pro-survival mucosal marker, were higher in Pirc than in wt rats, while the mucin MUC2, was lower in Pirc rats. Branched SCFAs were higher in Pirc than in wt animals. By Spearman analysis CRC biomarkers correlated with FM (in both young and old rats) and with MAM (in young rats), suggesting a specific relationship between the gut microbiota profile and these functional mucosal parameters deserving further investigation.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinogénesis/genética , Colon/microbiología , Neoplasias del Colon/genética , Quinasas Similares a Doblecortina/genética , Mucina 2/genética , Factores de Edad , Animales , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/aislamiento & purificación , Biomarcadores de Tumor/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Colon/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/microbiología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Quinasas Similares a Doblecortina/metabolismo , Enterococcus/crecimiento & desarrollo , Enterococcus/aislamiento & purificación , Escherichia/crecimiento & desarrollo , Escherichia/aislamiento & purificación , Ácidos Grasos Volátiles/metabolismo , Heces/microbiología , Regulación de la Expresión Génica , Lactobacillus/crecimiento & desarrollo , Lactobacillus/aislamiento & purificación , Masculino , Mucina 2/metabolismo , Análisis de Componente Principal , Proteus/crecimiento & desarrollo , Proteus/aislamiento & purificación , Ratas , Ratas Endogámicas F344 , Shigella/crecimiento & desarrollo , Shigella/aislamiento & purificación , Streptococcus/crecimiento & desarrollo , Streptococcus/aislamiento & purificación
10.
Front Immunol ; 12: 772532, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34970262

RESUMEN

Emerging evidence supports that the efficacy of immune checkpoint blockade (ICB) therapy is associated with the host's gut microbiota, as prior antibiotic intake often leads to poor outcome and low responsiveness toward ICB treatment. Therefore, we hypothesized that the efficacy of ICB therapy like anti-programmed cell death protein-1 (PD-1) treatment required an intact host gut microbiota, and it was established that probiotics could enhance the recovery of gut microbiota disruption by external stimuli. Thus, the present study aimed to evaluate the effect of the probiotics, Lactobacillus rhamnosus Probio-M9, on recovering antibiotic-disrupted gut microbiota and its impact on the outcome of ICB therapy in tumor-bearing mice. We first disrupted the mouse microbiota by antibiotics and then remediated the gut microbiota by probiotics or naturally. Tumor transplantation was then performed, followed by anti-PD-1-based antitumor therapy. Changes in the fecal metagenomes and the tumor suppression effect were monitored during different stages of the experiment. Our results showed that Probio-M9 synergized with ICB therapy, significantly improving tumor inhibition compared with groups not receiving the probiotic treatment (P < 0.05 at most time points). The synergistic effect was accompanied by effective restoration of antibiotic-disrupted fecal microbiome that was characterized by a drastically reduced Shannon diversity value and shifted composition of dominating taxa. Moreover, probiotic administration significantly increased the relative abundance of beneficial bacteria (e.g., Bifidobacterium pseudolongum, Parabacteroides distasonis, and some Bacteroides species; 0.0001 < P < 0.05). The gut microbiome changes were accompanied by mild reshaping of the functional metagenomes characterized by enrichment in sugar degradation and vitamin and amino acid synthesis pathways. Collectively, this study supported that probiotic administration could enhance the efficacy and responsiveness of anti-PD-1-based immunotherapy, and Probio-M9 could be a potential candidate of microbe-based synergistic tumor therapeutics. The preclinical data obtained here would support the design of future human clinical trials for further consolidating the current findings and for safety assessment of probiotic adjunctive treatment in ICB therapy.


Asunto(s)
Antibacterianos/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Lacticaseibacillus rhamnosus , Neoplasias/terapia , Probióticos/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Bacteroides/efectos de los fármacos , Bacteroides/crecimiento & desarrollo , Bifidobacterium/efectos de los fármacos , Bifidobacterium/crecimiento & desarrollo , Línea Celular Tumoral , Heces/microbiología , Ratones Endogámicos BALB C , Neoplasias/microbiología
11.
Microb Cell Fact ; 20(1): 213, 2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34794462

RESUMEN

The intestinal microecological environment is critical to an infant's growth. For those infants consuming milk power, it is very important to improve the intestinal microecological environment to promote the healthy growth of infants. In this paper, Milk protein hydrolysate (MPH), consisting of different proportions of proteins and small molecule peptides (5:5, 4:6, 3:7, 2:8, 1:9) were added to infant formula powder (IFP). The effects of MFP-enriched IFP addition on proliferation and metabolism of Bifidobacterium L80 were studied. Compared with MPH-free IFP, MFP-enriched IFP with 1:9 of proteins to small molecule peptides significantly enhanced the proliferation of Bifidobacterium L80, resulting in higher cell density, greater viable counts and higher titratable acidity. MFP-enriched IFP increased the content of seven organic acids and H2O2 in the system, and improved the antibacterial activity to E. coli BL21. This study suggested that MPH could be an effective addition to infant formula powder to promote the growth of Bifidobacterium, so to improve the intestinal health of infants.


Asunto(s)
Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Caseínas/metabolismo , Intestinos/microbiología , Proteínas de la Leche/metabolismo , Hidrolisados de Proteína/metabolismo , Proteína de Suero de Leche/metabolismo , Animales , Caseínas/química , Humanos , Fórmulas Infantiles/química , Proteínas de la Leche/química , Hidrolisados de Proteína/química , Proteína de Suero de Leche/química
12.
Gut Microbes ; 13(1): 1986666, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34705611

RESUMEN

Understanding how exogenous microbes stably colonize the animal gut is essential to reveal mechanisms of action and tailor effective probiotic treatments. Bifidobacterium species are naturally enriched in the gastrointestinal tract of breast-fed infants. Human milk oligosaccharides (HMOs) are associated with this enrichment. However, direct mechanistic proof of the importance of HMOs in this colonization is lacking given milk contains additional factors that impact the gut microbiota. This study examined mice supplemented with the HMO 2'fucosyllactose (2'FL) together with a 2'FL-consuming strain, Bifidobacterium pseudocatenulatum MP80. 2'FL supplementation creates a niche for high levels of B.p. MP80 persistence, similar to Bifidobacterium levels seen in breast-fed infants. This synergism impacted gut microbiota composition, activated anti-inflammatory pathways and protected against chemically-induced colitis. These results demonstrate that bacterial-milk glycan interactions alone drive enrichment of beneficial Bifidobacterium and provide a model for tunable colonization thus facilitating insight into mechanisms of health promotion by bifidobacteriain neonates.


Asunto(s)
Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Colitis/prevención & control , Leche Humana/metabolismo , Oligosacáridos/metabolismo , Animales , Lactancia Materna , Colitis/metabolismo , Colitis/microbiología , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Gut Microbes ; 13(1): 1973835, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34553672

RESUMEN

Certain existing prebiotics meant to facilitate the growth of beneficial bacteria in the intestine also promote the growth of other prominent bacteria. Therefore, the growth-promoting effects of ß-galactosides on intestinal bacteria were analyzed. Galactosyl-ß1,4-l-rhamnose (Gal-ß1,4-Rha) selectively promoted the growth of Bifidobacterium. Bifidobacterium longum subsp. longum 105-A (JCM 31944) has multiple solute-binding proteins belonging to ATP-binding cassette transporters for sugars. Each strain in the library of 11 B. longum subsp. longum mutants, in which each gene of the solute-binding protein was disrupted, was cultured in a medium containing Gal-ß1,4-Rha as the sole carbon source, and only the BL105A_0502 gene-disruption mutant showed delayed and reduced growth compared to the wild-type strain. BL105A_0502 homolog is highly conserved in bifidobacteria. In a Gal-ß1,4-Rha-containing medium, Bifidobacterium longum subsp. infantis JCM 1222T, which possesses BLIJ_2090, a homologous protein to BL105A_0502, suppressed the growth of enteric pathogen Clostridioides difficile, whereas the BLIJ_2090 gene-disrupted mutant did not. In vivo, administration of B. infantis and Gal-ß1,4-Rha alleviated C. difficile infection-related weight loss in mice. We have successfully screened Gal-ß1,4-Rha as a next-generation prebiotic candidate that specifically promotes the growth of beneficial bacteria without promoting the growth of prominent bacteria and pathogens.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Bifidobacterium/crecimiento & desarrollo , Clostridioides difficile/crecimiento & desarrollo , Disacáridos/farmacología , Prebióticos/análisis , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Bifidobacterium/genética , Bifidobacterium longum subspecies infantis/genética , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL
14.
Int J Biol Macromol ; 189: 151-159, 2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34400230

RESUMEN

Gut microbial fermentation of soluble dietary fibers promotes general and substrate-specific health benefits. In this study, the fermentation characteristics of two soluble branched-dietary fibers, namely, agavin (a type of agave fructans) and digestion-resistant maltodextrin (RD) were investigated against cellulose, using a simulated colonic fermenter apparatus employing a mixed culture of swine fecal bacteria. After 48 h of complete fermentation period, the microbial composition was different among all groups, where Bifidobacterium spp. and Lactobacillus spp. dominated the agavin treatment, while the members of the families Lachnospiraceae and Prevotellaceae dominated the RD treatment. Agavin treatment exhibited a clearly segregated two-phased prolonged fermentation trend compared to RD treatment as manifested by the fermentation rates. Further, the highest short-chain fatty acids production even at the end of the fermentation cycle, acidic pH, and the negligible concentration of ammonia accumulation demonstrated favorable fermentation attributes of agavin compared to RD. Therefore, agavin might be an effective and desirable substrate for the colonic microbiota than RD with reference to the expressed microbial taxa and fermentation attributes. This study revealed a notable significance of the structural differences of fermentable fibers on the subsequent fermentation characteristics.


Asunto(s)
Técnicas de Cultivo Celular por Lotes , Colon/fisiología , Fermentación , Fructanos/metabolismo , Polisacáridos/metabolismo , Amoníaco/análisis , Animales , Bifidobacterium/crecimiento & desarrollo , Biodiversidad , Recuento de Colonia Microbiana , Digestión , Ácidos Grasos Volátiles/metabolismo , Concentración de Iones de Hidrógeno , Lactobacillus/crecimiento & desarrollo , Microbiota , Nitrógeno/análisis , Filogenia , Porcinos , Factores de Tiempo
15.
Microb Genom ; 7(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34100697

RESUMEN

Although the beneficial effects of probiotics are likely to be associated with their ability to colonize the gut, little is known about the characteristics of good colonizers. In a systematic analysis of the comparative genomics, we tried to elucidate the genomic contents that account for the distinct host adaptability patterns of Lactobacillus and Bifidobacterium species. The Bifidobacterium species, with species-level phylogenetic structures affected by recombination among strains, broad mucin-foraging activity, and dietary-fibre-degrading ability, represented niche conservatism and tended to be host-adapted. The Lactobacillus species stretched across three lifestyles, namely free-living, nomadic and host-adapted, as characterized by the variations of bacterial occurrence time, guanine-cytosine (GC) content and genome size, evolution event frequency, and the presence of human-adapted bacterial genes. The numbers and activity of host-adapted factors, such as bile salt hydrolase and intestinal tissue-anchored elements, were distinctly distributed among the three lifestyles. The strains of the three lifestyles could be separated with such a collection of colonization-related genomic content (genes, genome size and GC content). Thus, our work provided valuable information for rational selection and gut engraftment prediction of probiotics. Here, we have found many interesting predictive results for bacterial gut fitness, which will be validated in vitro and in vivo.


Asunto(s)
Bifidobacterium/genética , Microbioma Gastrointestinal/genética , Genoma Bacteriano , Genómica , Lactobacillus/genética , Amidohidrolasas , Animales , Bifidobacterium/clasificación , Bifidobacterium/crecimiento & desarrollo , Tracto Gastrointestinal/microbiología , Humanos , Lactobacillus/clasificación , Lactobacillus/crecimiento & desarrollo , Estilo de Vida , Ratones , Filogenia , Probióticos , ARN Ribosómico 16S/genética
16.
Nutr Metab Cardiovasc Dis ; 31(8): 2458-2470, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34090773

RESUMEN

BACKGROUND AND AIMS: Insufficient dietary fiber (DF) intake is associated with increased blood pressure (BP) and the mode of action is unclear. The intake of DF supplements by participants in previous interventional studies was still far below the amount recommended by the World Health Organization. Therefore, this study aims to explore the effect of supplementing relatively sufficient DF on BP and gut microbiota in patients with essential hypertension (HTN). METHODS AND RESULTS: Fifty participants who met the inclusion criteria were randomly divided into the DF group (n = 25) and control group (n = 25). All the participants received education on regular dietary guidance for HTN. In addition to dietary guidance, one bag of oat bran (30 g/d) supplement (containing DF 8.9 g) was delivered to the DF group. The office BP (oBP), 24 h ambulatory blood pressure, and gut microbiota were measured at baseline and third month. After intervention, the office systolic blood pressure (oSBP; P < 0.001) and office diastolic blood pressure (oDBP; P < 0.028) in the DF group were lower than those in the control group. Similarly, the changes in 24hmaxSBP (P = 0.002), 24hmaxDBP (P = 0.001), 24haveSBP (P < 0.007), and 24haveDBP (P = 0.008) were greater in the DF group than in the control group. The use of antihypertensive drugs in the DF group was significantly reduced (P = 0.021). The ß diversity, including Jaccard (P = 0.008) and Bray-Curtis distance (P = 0.004), showed significant differences (P < 0.05) between two groups by the third month. The changes of Bifidobacterium (P = 0.019) and Spirillum (P = 0.006) in the DF group were significant. CONCLUSIONS: Increased DF (oat bran) supplement improved BP, reduced the amount of antihypertensive drugs, and modulated the gut microbiota. TRIAL REGISTRATION NUMBER: ChiCTR1900024055.


Asunto(s)
Avena , Bifidobacterium/crecimiento & desarrollo , Presión Sanguínea , Fibras de la Dieta/administración & dosificación , Grano Comestible , Hipertensión Esencial/dietoterapia , Microbioma Gastrointestinal , Spirillum/crecimiento & desarrollo , Adulto , Monitoreo Ambulatorio de la Presión Arterial , China , Disbiosis , Hipertensión Esencial/diagnóstico , Hipertensión Esencial/microbiología , Hipertensión Esencial/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Valor Nutritivo , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento
17.
BMC Microbiol ; 21(1): 154, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-34030655

RESUMEN

BACKGROUND: Bifidobacteria are commensal microbes of the mammalian gastrointestinal tract. In this study, we aimed to identify the intestinal colonization mechanisms and key metabolic pathways implemented by Bifidobacterium dentium. RESULTS: B. dentium displayed acid resistance, with high viability over a pH range from 4 to 7; findings that correlated to the expression of Na+/H+ antiporters within the B. dentium genome. B. dentium was found to adhere to human MUC2+ mucus and harbor mucin-binding proteins. Using microbial phenotyping microarrays and fully-defined media, we demonstrated that in the absence of glucose, B. dentium could metabolize a variety of nutrient sources. Many of these nutrient sources were plant-based, suggesting that B. dentium can consume dietary substances. In contrast to other bifidobacteria, B. dentium was largely unable to grow on compounds found in human mucus; a finding that was supported by its glycosyl hydrolase (GH) profile. Of the proteins identified in B. dentium by proteomic analysis, a large cohort of proteins were associated with diverse metabolic pathways, indicating metabolic plasticity which supports colonization of the dynamic gastrointestinal environment. CONCLUSIONS: Taken together, we conclude that B. dentium is well adapted for commensalism in the gastrointestinal tract.


Asunto(s)
Bifidobacterium/metabolismo , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Ácidos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bifidobacterium/genética , Bifidobacterium/crecimiento & desarrollo , Tracto Gastrointestinal/fisiología , Genoma Bacteriano , Glucosa/metabolismo , Humanos , Simbiosis
18.
Commun Biol ; 4(1): 541, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972677

RESUMEN

This study aims to understand the mechanistic basis underlying the response of Bifidobacterium to lactulose ingestion in guts of healthy Japanese subjects, with specific focus on a lactulose transporter. An in vitro assay using mutant strains of Bifidobacterium longum subsp. longum 105-A shows that a solute-binding protein with locus tag number BL105A_0502 (termed LT-SBP) is primarily involved in lactulose uptake. By quantifying faecal abundance of LT-SBP orthologues, which is defined by phylogenetic analysis, we find that subjects with 107 to 109 copies of the genes per gram of faeces before lactulose ingestion show a marked increase in Bifidobacterium after ingestion, suggesting the presence of thresholds between responders and non-responders to lactulose. These results help predict the prebiotics-responder and non-responder status and provide an insight into clinical interventions that test the efficacy of prebiotics.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/metabolismo , Bifidobacterium/crecimiento & desarrollo , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Lactulosa/administración & dosificación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Bifidobacterium/efectos de los fármacos , Bifidobacterium/metabolismo , Estudios Transversales , Femenino , Fármacos Gastrointestinales/administración & dosificación , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Persona de Mediana Edad , Adulto Joven
19.
Food Chem ; 355: 129608, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-33799260

RESUMEN

Effects of enzymatic hydrolysis on the structural, rheological, and functional properties of mulberry leaf polysaccharide (MLP) were characterized in this study. The enzymatic hydrolysis of MLP raised the carbonyl, carboxyl, and hydroxyl groups from 7.21 ± 0.86 to 10.08 ± 0.28 CO/100 Glu, 9.40 ± 0.13 to 17.55 ± 0.34 COOH/100 Glu, and 5.71 ± 0.33 to 8.14 ± 0.24 OH/100 Glu, respectively. Meanwhile, an increase in thixotropic performance and structure-recovery capacities were observed in hydrolyzed MLP, while the molecular weight, surface tension, apparent viscosity, and thermal stability were decreased. An improved antioxidant activity of MLP was also achieved after the enzymatic degradation. Moreover, the hydrolyzed MLP showed greater ability to promote the growths of Bifidobacterium bifidum, Bifidobacterium adolescentis, Lactobacillus rhamnosus, and Lactobacillus acidophilus and the production of acetic acid, butyric acid, and lactic acid. The results demonstrate that enzymatic modification is a useful approach for polysaccharide processing.


Asunto(s)
Glicósido Hidrolasas/metabolismo , Morus/química , Morus/metabolismo , Polisacáridos/química , Antioxidantes/química , Bifidobacterium/efectos de los fármacos , Bifidobacterium/crecimiento & desarrollo , Hidrólisis , Lactobacillus/efectos de los fármacos , Lactobacillus/crecimiento & desarrollo , Hojas de la Planta/química , Hojas de la Planta/metabolismo , Polisacáridos/metabolismo , Polisacáridos/farmacología , Prebióticos , Reología , Viscosidad
20.
Methods Mol Biol ; 2278: 1-12, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33649943

RESUMEN

Since their discovery, bifidobacteria have been considered to represent cornerstone commensal microorganisms in the host-microbiome interface at the intestinal level. Bifidobacteria have therefore enjoyed increasing scientific and commercial interest as a source of microorganisms with probiotic potential. However, since functional and probiotic traits are strictly strain-dependent, there is a constant need to isolate, cultivate, and characterize novel strains, activities that require the utilization of appropriate media, as well as robust isolation, cultivation, and preservation techniques. Besides, effective isolation of bifidobacteria from natural environments might require different manipulation and cultivation media and conditions depending on the specific characteristics of the sample material, the presence of competitive microbiota, the metabolic state in which bifidobacteria might be encountered within the sample and the particular metabolic traits of the bifidobacterial species adapted to such inhabitation.A wide array of culture media recipes have been described in the literature to routinely isolate and grow bifidobacteria under laboratory conditions. However, there is not a single and universally applicable medium for effective isolation, recovery, and cultivation of bifidobacteria, as each growth medium has its own particular advantages and limitations. Besides, the vast majority of these media formulations was not specifically formulated for these microorganisms, and thus information on bifidobacterial cultivation options is scarce while being scattered throughout literature. This chapter intends to serve as a resource summarizing the options to cultivate bifidobacteria that have been described to date, highlighting the main advantages and limitations of each of them.


Asunto(s)
Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/aislamiento & purificación , Medios de Cultivo/química , Probióticos/aislamiento & purificación , Bifidobacterium/metabolismo , Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/metabolismo , Microbioma Gastrointestinal , Humanos , Probióticos/metabolismo , Simbiosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA