Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.390
Filtrar
1.
Curr Microbiol ; 81(7): 168, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38733376

RESUMEN

In 2018, Nouioui et al. proposed that Bifidobacterium coryneforme was a later synonym of Bifidobacterium indicum on the basis of the digital DNA-DNA hybridization (dDDH) value (85.0%) between B. coryneforme LMG 18911T and B. indicum LMG 11587T. However, in the study of Scardovi et al. (1970), the type strains of B. indicum and B. coryneforme only exhibited 60% DNA-DNA hybridization value. In the present study, the genomes of B. coryneforme CGMCC 1.2279T, B. coryneforme JCM 5819T, B. indicum JCM 1302T, B. indicum CGMCC 1.2275T, B. indicum DSM 20214T, B. indicum LMG 27437T, B. indicum ATCC 25912T, B. indicum KCTC 3230T, B. indicum CCUG 34985T, were sequenced, and the taxonomic relationship between B. coryneforme and B. indicum was re-evaluated. On the basis of the results presented here, (i) ATCC 25912 and DSM 20214 deposited by Vittorio Scardovi are two different strains; (ii) the type strain of B. indicum is ATCC 25912T (= JCM 1302T = LMG 27437T = CGMCC 1.2275T = KCTC 3230T), and not DSM 20214 (= BCRC 14674 = CCUG 34985 = LMG 11587); (iii) B. coryneforme and B. indicum represent two different species of the genus Bifidobacterium; (iv) strain DSM 20214 (= BCRC 14674 = CCUG 34985 = LMG 11587) belongs to B. coryneforme.


Asunto(s)
Bifidobacterium , ADN Bacteriano , Genoma Bacteriano , Filogenia , Bifidobacterium/genética , Bifidobacterium/clasificación , Bifidobacterium/aislamiento & purificación , ADN Bacteriano/genética , Hibridación de Ácido Nucleico , Técnicas de Tipificación Bacteriana , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN
2.
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
3.
Nutrients ; 16(9)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38732641

RESUMEN

Numerous studies have investigated the immunomodulatory effects of yogurt, but the underlying mechanism remained elusive. This study aimed to elucidate the alleviating properties of yogurt on immunosuppression and proposed the underlying mechanism was related to the metabolite D-lactate. In the healthy mice, we validated the safety of daily yogurt consumption (600 µL) or D-lactate (300 mg/kg). In immunosuppressed mice induced by cyclophosphamide (CTX), we evaluated the immune regulation of yogurt and D-lactate. The result showed that yogurt restored body weight, boosted immune organ index, repaired splenic tissue, recovered the severity of delayed-type hypersensitivity reactions and increased serum cytokines (IgA, IgG, IL-6, IFN-γ). Additionally, yogurt enhanced intestinal immune function by restoring the intestinal barrier and upregulating the abundance of Bifidobacterium and Lactobacillus. Further studies showed that D-lactate alleviated immunosuppression in mice mainly by promoting cellular immunity. D-lactate recovered body weight and organ development, elevated serum cytokines (IgA, IgG, IL-6, IFN-γ), enhanced splenic lymphocyte proliferation and increased the mRNA level of T-bet in splenic lymphocyte to bolster Th1 differentiation. Finally, CTX is a chemotherapeutic drug, thus, the application of yogurt and D-lactate in the tumor-bearing mouse model was initially explored. The results showed that both yogurt (600 µL) and D-lactate (300 mg/kg) reduced cyclophosphamide-induced immunosuppression without promoting tumor growth. Overall, this study evaluated the safety, immune efficacy and applicability of yogurt and D-lactate in regulating immunosuppression. It emphasized the potential of yogurt as a functional food for immune regulation, with D-lactate playing a crucial role in its immunomodulatory effects.


Asunto(s)
Ciclofosfamida , Citocinas , Ácido Láctico , Yogur , Animales , Ratones , Ácido Láctico/sangre , Citocinas/metabolismo , Masculino , Terapia de Inmunosupresión , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/inmunología , Ratones Endogámicos BALB C , Hipersensibilidad Tardía/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Lactobacillus , Bifidobacterium
4.
Gut Microbes ; 16(1): 2347728, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38706226

RESUMEN

Indole in the gut is formed from dietary tryptophan by a bacterial tryptophan-indole lyase. Indole not only triggers biofilm formation and antibiotic resistance in gut microbes but also contributes to the progression of kidney dysfunction after absorption by the intestine and sulfation in the liver. As tryptophan is an essential amino acid for humans, these events seem inevitable. Despite this, we show in a proof-of-concept study that exogenous indole can be converted to an immunomodulatory tryptophan metabolite, indole-3-lactic acid (ILA), by a previously unknown microbial metabolic pathway that involves tryptophan synthase ß subunit and aromatic lactate dehydrogenase. Selected bifidobacterial strains converted exogenous indole to ILA via tryptophan (Trp), which was demonstrated by incubating the bacterial cells in the presence of (2-13C)-labeled indole and l-serine. Disruption of the responsible genes variedly affected the efficiency of indole bioconversion to Trp and ILA, depending on the strains. Database searches against 11,943 bacterial genomes representing 960 human-associated species revealed that the co-occurrence of tryptophan synthase ß subunit and aromatic lactate dehydrogenase is a specific feature of human gut-associated Bifidobacterium species, thus unveiling a new facet of bifidobacteria as probiotics. Indole, which has been assumed to be an end-product of tryptophan metabolism, may thus act as a precursor for the synthesis of a host-interacting metabolite with possible beneficial activities in the complex gut microbial ecosystem.


Asunto(s)
Bifidobacterium , Microbioma Gastrointestinal , Indoles , Triptófano , Triptófano/metabolismo , Humanos , Indoles/metabolismo , Bifidobacterium/metabolismo , Bifidobacterium/genética , Triptófano Sintasa/metabolismo , Triptófano Sintasa/genética , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo
5.
Microb Biotechnol ; 17(5): e14443, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38722820

RESUMEN

Pectin structures have received increasing attention as emergent prebiotics due to their capacity to promote beneficial intestinal bacteria. Yet the collective activity of gut bacterial communities to cooperatively metabolize structural variants of this substrate remains largely unknown. Herein, the characterization of a pectin methylesterase, BpeM, from Bifidobacterium longum subsp. longum, is reported. The purified enzyme was able to remove methyl groups from highly methoxylated apple pectin, and the mathematical modelling of its activity enabled to tightly control the reaction conditions to achieve predefined final degrees of methyl-esterification in the resultant pectin. Demethylated pectin, generated by BpeM, exhibited differential fermentation patterns by gut microbial communities in in vitro mixed faecal cultures, promoting a stronger increase of bacterial genera associated with beneficial effects including Lactobacillus, Bifidobacterium and Collinsella. Our findings demonstrate that controlled pectin demethylation by the action of a B. longum esterase selectively modifies its prebiotic fermentation pattern, producing substrates that promote targeted bacterial groups more efficiently. This opens new possibilities to exploit biotechnological applications of enzymes from gut commensals to programme prebiotic properties.


Asunto(s)
Hidrolasas de Éster Carboxílico , Heces , Malus , Pectinas , Prebióticos , Malus/microbiología , Pectinas/metabolismo , Heces/microbiología , Hidrolasas de Éster Carboxílico/metabolismo , Hidrolasas de Éster Carboxílico/genética , Fermentación , Humanos , Bifidobacterium longum/metabolismo , Bifidobacterium longum/enzimología , Microbioma Gastrointestinal , Bifidobacterium/enzimología , Bifidobacterium/metabolismo
6.
Sci Rep ; 14(1): 11560, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773299

RESUMEN

IBD is a disorder which could be caused by oxidative stress. This investigation aims to determine if probiotics and postbiotics can control oxidative stress and inflammation and compare the effectiveness of these two probiotic and postbiotic mixtures of substances. 88 strains of Lactobacillus and Bifidobacterium were tested for antioxidant activity. Male wild-type C57BL/6 mice were divided into four experimental groups, namely high fat diet (HFD) + PBS, HFD + DSS, HFD + DSS + 109 cfu/ml of probiotics, and HFD + DSS + 109 cfu/ml of postbiotics. The phenotypical indices and pathological scores were assessed. The expression of genes related to NF-kB and Nrf2 signaling pathways and enzymes associated with oxidant/anti-oxidant activities, and proinflammatory/inflammatory cytokines were assessed. In contrast to the groups exposed to DSS, mice treated with probiotics mixture and postbiotics mixture alongside DSS displayed alleviation of DSS-induced adverse effects on phenotypical characteristics, as well as molecular indices such as the Nrf2 and NF-kB related genes, with a greater emphasis on the postbiotics component. In accordance with the findings of the present investigation, it can be inferred that even in using a high-fat dietary regimen as an inducer of oxidative stress, the emergence of inflammation can be effectively addressed through the utilization of probiotics and, more specifically, postbiotics.


Asunto(s)
Antiinflamatorios , Antioxidantes , Colitis , Sulfato de Dextran , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2 , FN-kappa B , Estrés Oxidativo , Probióticos , Transducción de Señal , Animales , Probióticos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Antioxidantes/metabolismo , Antioxidantes/farmacología , Masculino , Ratones , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Antiinflamatorios/farmacología , Transducción de Señal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Lactobacillus , Bifidobacterium , Dieta Alta en Grasa/efectos adversos
7.
Nutrients ; 16(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732599

RESUMEN

In this study, a systematic review of randomized clinical trials conducted from January 2000 to December 2023 was performed to examine the efficacy of psychobiotics-probiotics beneficial to mental health via the gut-brain axis-in adults with psychiatric and cognitive disorders. Out of the 51 studies involving 3353 patients where half received psychobiotics, there was a notably high measurement of effectiveness specifically in the treatment of depression symptoms. Most participants were older and female, with treatments commonly utilizing strains of Lactobacillus and Bifidobacteria over periods ranging from 4 to 24 weeks. Although there was a general agreement on the effectiveness of psychobiotics, the variability in treatment approaches and clinical presentations limits the comparability and generalization of the findings. This underscores the need for more personalized treatment optimization and a deeper investigation into the mechanisms through which psychobiotics act. The research corroborates the therapeutic potential of psychobiotics and represents progress in the management of psychiatric and cognitive disorders.


Asunto(s)
Trastornos Mentales , Probióticos , Ensayos Clínicos Controlados Aleatorios como Asunto , Humanos , Probióticos/uso terapéutico , Femenino , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/terapia , Trastornos del Conocimiento/tratamiento farmacológico , Masculino , Resultado del Tratamiento , Adulto , Eje Cerebro-Intestino/efectos de los fármacos , Persona de Mediana Edad , Microbioma Gastrointestinal/efectos de los fármacos , Lactobacillus , Anciano , Bifidobacterium
8.
Sci Rep ; 14(1): 11181, 2024 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755201

RESUMEN

Gut microbiota manipulation may reverse metabolic abnormalities in obesity. Our previous studies demonstrated that inulin supplementation significantly promoted Bifidobacterium and fat-free mass in obese children. We aimed to study gut-muscle axis from inulin supplementation in these children. In clinical phase, the plasma samples from 46 participants aged 7-15 years, were analyzed for muscle biomarkers before and after 6-month inulin supplementation. In parallel, the plausible mechanism of muscle production via gut-muscle axis was examined using macrophage cell line. Bifidobacterium was cultured in semi-refined medium with inulin used in the clinical phase. Cell-free supernatant was collected and used in lipopolysaccharide (LPS)-induced macrophage cell line to determine inflammatory and anti-inflammatory gene expression. In clinical phase, IL-15 and creatinine/cystatin C ratio significantly increased from baseline to the 6th month. In vitro study showed that metabolites derived from Bifidobacterium capable of utilizing inulin contained the abundance of SCFAs. In the presence of LPS, treatment from Bifidobacterium + inulin downregulated TNF-α, IL-6, IL-1ß, and iNOS, but upregulated FIZZ-1 and TGF-ß expression. Inulin supplementation promoted the muscle biomarkers in agreement with fat-free mass gain, elucidating by Bifidobacterium metabolites derived from inulin digestion showed in vitro anti-inflammatory activity and decreased systemic pro-inflammation, thus promoting muscle production via gut-muscle axis response.Clinical Trial Registry number: NCT03968003.


Asunto(s)
Bifidobacterium , Suplementos Dietéticos , Microbioma Gastrointestinal , Inulina , Inulina/farmacología , Inulina/administración & dosificación , Humanos , Niño , Adolescente , Masculino , Microbioma Gastrointestinal/efectos de los fármacos , Femenino , Biomarcadores , Obesidad Infantil/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Lipopolisacáridos , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos
9.
Cell Host Microbe ; 32(4): 573-587.e5, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38569545

RESUMEN

Microbiota assembly in the infant gut is influenced by diet. Breastfeeding and human breastmilk oligosaccharides promote the colonization of beneficial bifidobacteria. Infant formulas are supplemented with bifidobacteria or complex oligosaccharides, notably galacto-oligosaccharides (GOS), to mimic breast milk. To compare microbiota development across feeding modes, this randomized controlled intervention study (German Clinical Trial DRKS00012313) longitudinally sampled infant stool during the first year of life, revealing similar fecal bacterial communities between formula- and breast-fed infants (N = 210) but differences across age. Infant formula containing GOS sustained high levels of bifidobacteria compared with formula containing B. longum and B. breve or placebo. Metabolite and bacterial profiling revealed 24-h oscillations and circadian networks. Rhythmicity in bacterial diversity, specific taxa, and functional pathways increased with age and was strongest following breastfeeding and GOS supplementation. Circadian rhythms in dominant taxa were further maintained ex vivo in a chemostat model. Hence, microbiota rhythmicity develops early in life and is impacted by diet.


Asunto(s)
Fórmulas Infantiles , Microbiota , Lactante , Femenino , Humanos , Fórmulas Infantiles/microbiología , Lactancia Materna , Leche Humana , Bifidobacterium , Heces/microbiología , Oligosacáridos/metabolismo , Ritmo Circadiano
10.
Cancer Immunol Immunother ; 73(5): 94, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38564002

RESUMEN

The advent of tumor immunotherapy in patients has revolutionized the treatment of tumors and significantly improved survival rates for a wide range of tumors. However, the full therapeutic potential of immune checkpoint inhibitors (ICIs) has yet to be realized, as not all patients have a lasting survival benefit from them, and a significant proportion of patients show primary or acquired resistance to immunotherapy. Bifidobacterium is one of the most common probiotics, and its antitumor and immunomodulatory effects have been demonstrated in recent years, but its immunomodulatory effects in tumors, especially on ICIs and in combination, have not been extensively studied in clinical practice, and its effects on the immune system and the mechanisms that modulate immunotherapy are largely unknown. Therefore, this review will focus on the immunomodulatory effects of Bifidobacteria in malignancies and the possible mechanisms of action of Bifidobacteria on immunotherapy in the hope of providing a basis for further research and better application of Bifidobacteria in clinical practice.


Asunto(s)
Inmunomodulación , Inmunoterapia , Humanos , Bifidobacterium , Inhibidores de Puntos de Control Inmunológico
11.
Artículo en Inglés | MEDLINE | ID: mdl-38661726

RESUMEN

A novel bifidobacterium (designated F753-1T) was isolated from the gut of honeybee (Apis mellifera). Strain F753-1T was characterized using a polyphasic taxonomic approach. Strain F753-1T was phylogenetically related to the type strains of Bifidobacterium mizhiensis, Bifidobacterium asteroides, Bifidobacterium choladohabitans, Bifidobacterium mellis, Bifidobacterium apousia and Bifidobacterium polysaccharolyticum, having 98.4-99.8 % 16S rRNA gene sequence similarities. The phylogenomic tree indicated that strain F753-1T was most closely related to the type strains of B. mellis and B. choladohabitans. Strain F753-1T had the highest average nucleotide identity (94.1-94.5 %) and digital DNA-DNA hybridization (56.3 %) values with B. mellis Bin7NT. Acid production from amygdalin, d-fructose, gentiobiose, d-mannose, maltose, sucrose and d-xylose, activity of α-galactosidase, pyruvate utilization and hydrolysis of hippurate could differentiate strain F753-1T from B. mellis CCUG 66113T and B. choladohabitans JCM 34586T. Based upon the data obtained in the present study, a novel species, Bifidobacterium apis sp. nov., is proposed, and the type strain is F753-1T (=CCTCC AB 2023227T=JCM 36562T=LMG 33388T).


Asunto(s)
Técnicas de Tipificación Bacteriana , Bifidobacterium , ADN Bacteriano , Hibridación de Ácido Nucleico , Filogenia , ARN Ribosómico 16S , Análisis de Secuencia de ADN , Abejas/microbiología , Animales , ARN Ribosómico 16S/genética , Bifidobacterium/aislamiento & purificación , Bifidobacterium/clasificación , Bifidobacterium/genética , ADN Bacteriano/genética , Ácidos Grasos , Composición de Base , Microbioma Gastrointestinal
12.
Microbiol Res ; 283: 127709, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38593579

RESUMEN

Bifidobacterium longum subsp. infantis commonly colonizes the human gut and is capable of metabolizing L-fucose, which is abundant in the gut. Multiple studies have focused on the mechanisms of L-fucose utilization by B. longum subsp. infantis, but the regulatory pathways governing the expression of these catabolic processes are still unclear. In this study, we have conducted a structural and functional analysis of L-fucose metabolism transcription factor FucR derived from B. longum subsp. infantis Bi-26. Our results indicated that FucR is a L-fucose-sensitive repressor with more α-helices, fewer ß-sheets, and ß-turns. Transcriptional analysis revealed that FucR displays weak negative self-regulation, which is counteracted in the presence of L-fucose. Isothermal titration calorimetry indicated that FucR has a 2:1 stoichiometry with L-fucose. The key amino acid residues for FucR binding L-fucose are Asp280 and Arg331, with mutation of Asp280 to Ala resulting in a decrease in the affinity between FucR and L-fucose with the Kd value from 2.58 to 11.68 µM, and mutation of Arg331 to Ala abolishes the binding ability of FucR towards L-fucose. FucR specifically recognized and bound to a 20-bp incomplete palindrome sequence (5'-ACCCCAATTACGAAAATTTTT-3'), and the affinity of the L-fucose-loaded FucR for the DNA fragment was lower than apo-FucR. The results provided new insights into the regulating L-fucose metabolism by B. longum subsp. infantis.


Asunto(s)
Bifidobacterium longum , Bifidobacterium , Humanos , Bifidobacterium/genética , Bifidobacterium/metabolismo , Fucosa/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Metabolismo de los Hidratos de Carbono , Bifidobacterium longum/genética , Bifidobacterium longum/metabolismo
13.
Sci Rep ; 14(1): 7983, 2024 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-38575668

RESUMEN

Dimension reduction has been used to visualise the distribution of multidimensional microbiome data, but the composite variables calculated by the dimension reduction methods have not been widely used to investigate the relationship of the human gut microbiome with lifestyle and disease. In the present study, we applied several dimension reduction methods, including principal component analysis, principal coordinate analysis (PCoA), non-metric multidimensional scaling (NMDS), and non-negative matrix factorization, to a microbiome dataset from 186 subjects with symptoms of  allergic rhinitis (AR) and 106 controls. All the dimension reduction methods supported that the distribution of microbial data points appeared to be continuous rather than discrete. Comparison of the composite variables calculated from the different dimension reduction methods showed that the characteristics of the composite variables differed depending on the distance matrices and the dimension reduction methods. The first composite variables calculated from PCoA and NMDS with the UniFrac distance were strongly associated with AR (FDR adjusted P = 2.4 × 10-4 for PCoA and P = 2.8 × 10-4 for NMDS), and also with the relative abundance of Bifidobacterium and Prevotella. The abundance of Bifidobacterium was also linked to intake of several nutrients, including carbohydrate, saturated fat, and alcohol via composite variables. Notably, the association between the composite variables and AR was much stronger than the association between the relative abundance of individual genera and AR. Our results highlight the usefulness of the dimension reduction methods for investigating the association of microbial composition with lifestyle and disease in clinical research.


Asunto(s)
Microbioma Gastrointestinal , Rinitis Alérgica , Humanos , Bifidobacterium , Prevotella , Análisis de Escalamiento Multidimensional
14.
BMC Res Notes ; 17(1): 123, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689371

RESUMEN

OBJECTIVE: Study of the human infant gut microbiome requires the use of surrogate mammalian species such as mice. We sought to investigate the usefulness of the greater wax moth larva, Galleria mellonella, as an alternative. RESULTS: We have analysed the native gut microbiome of Galleria and developed methods for clearing the native microbiome and introducing species from human infant faecal samples. We find that some species, e.g. enterococci, are more successful at recolonisation, but that others, e.g. Bifidobacterium, are less so. The work paves the way for using Galleria rather than mice in this and similar work.


Asunto(s)
Heces , Microbioma Gastrointestinal , Larva , Mariposas Nocturnas , Animales , Microbioma Gastrointestinal/fisiología , Humanos , Mariposas Nocturnas/microbiología , Larva/microbiología , Lactante , Heces/microbiología , Bifidobacterium/aislamiento & purificación , Enterococcus/aislamiento & purificación
15.
Nutrients ; 16(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38674840

RESUMEN

Throughout infancy, IgA is crucial for maintaining gut mucosal immunity. This study aims to determine whether supplementing newborn mice with eight different strains of Bifidobacterium longum subsp. infantis might regulate their IgA levels. The strains were gavaged to BALB/C female (n = 8) and male (n = 8) dams at 1-3 weeks old. Eight strains of B. longum subsp. infantis had strain-specific effects in the regulation of intestinal mucosal barriers. B6MNI, I4MI, and I10TI can increase the colonic IgA level in females and males. I8TI can increase the colonic IgA level in males. B6MNI was also able to significantly increase the colonic sIgA level in females. B6MNI, I4MI, I8TI, and I10TI regulated colonic and Peyer's patch IgA synthesis genes but had no significant effect on IgA synthesis pathway genes in the jejunum and ileum. Moreover, the variety of sIgA-coated bacteria in male mice was changed by I4MI, I5TI, I8TI, and B6MNI. These strains also can decrease the relative abundance of Escherichia coli. These results indicate that B. longum subsp. infantis can promote IgA levels but show strain specificity. Different dietary habits with different strains of Bifidobacterium may have varying effects on IgA levels when supplemented in early infancy.


Asunto(s)
Bifidobacterium longum subspecies infantis , Bifidobacterium , Inmunoglobulina A , Mucosa Intestinal , Ratones Endogámicos BALB C , Probióticos , Animales , Femenino , Masculino , Inmunoglobulina A/metabolismo , Ratones , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Probióticos/administración & dosificación , Microbioma Gastrointestinal , Animales Recién Nacidos , Intestinos/microbiología , Intestinos/inmunología , Inmunidad Mucosa , Especificidad de la Especie , Colon/microbiología , Colon/inmunología , Colon/metabolismo , Inmunoglobulina A Secretora/metabolismo
16.
Nutrients ; 16(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38674850

RESUMEN

Polyphenols and fermentable fibers have shown favorable effects on gut microbiota composition and metabolic function. However, few studies have investigated whether combining multiple fermentable fibers or polyphenols may have additive beneficial effects on gut microbial states. Here, an in vitro fermentation model, seeded with human stool combined from 30 healthy volunteers, was supplemented with blends of polyphenols (PP), dietary fibers (FB), or their combination (PPFB) to determine influence on gut bacteria growth dynamics and select metabolite changes. PP and FB blends independently led to significant increases in the absolute abundance of select beneficial taxa, namely Ruminococcus bromii, Bifidobacterium spp., Lactobacillus spp., and Dorea spp. Total short-chain fatty acid concentrations, relative to non-supplemented control (F), increased significantly with PPFB and FB supplementation but not PP. Indole and ammonia concentrations decreased with FB and PPFB supplementation but not PP alone while increased antioxidant capacity was only evident with both PP and PPFB supplementation. These findings demonstrated that, while the independent blends displayed selective positive impacts on gut states, the combination of both blends provided an additive effect. The work outlines the potential of mixed substrate blends to elicit a broader positive influence on gut microbial composition and function to build resiliency toward dysbiosis.


Asunto(s)
Fibras de la Dieta , Ácidos Grasos Volátiles , Heces , Fermentación , Microbioma Gastrointestinal , Indoles , Polifenoles , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Polifenoles/farmacología , Humanos , Fibras de la Dieta/farmacología , Fibras de la Dieta/administración & dosificación , Heces/microbiología , Ácidos Grasos Volátiles/metabolismo , Adulto , Masculino , Amoníaco/metabolismo , Femenino , Bacterias/metabolismo , Bacterias/crecimiento & desarrollo , Bacterias/efectos de los fármacos , Antioxidantes/farmacología , Bifidobacterium/metabolismo , Lactobacillus/metabolismo , Adulto Joven
17.
Biomed Pharmacother ; 174: 116561, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38593705

RESUMEN

Pectin and its derivatives have been shown to modulate immune signaling as well as gut microbiota in preclinical studies, which may constitute the mechanisms by which supplementation of specific pectic polysaccharides confers protection against viral respiratory infections. In a double-blind, placebo-controlled rhinovirus (RV16) challenge study, healthy volunteers were randomized to consume placebo (0.0 g/day) (N = 46), low-dose (0.3 g/day) (N = 49) or high-dose (1.5 g/day) (N = 51) of carrot derived rhamnogalacturonan-I (cRG-I) for eight weeks and they were subsequently challenged with RV-16. Here, the effect of 8-week cRG-I supplementation on the gut microbiota was studied. While the overall gut microbiota composition in the population was generally unaltered by this very low dose of fibre, the relative abundance of Bifidobacterium spp. (mainly B. adolescentis and B. longum) was significantly increased by both doses of cRG-1. Moreover, daily supplementation of cRG-I led to a dose-dependent reduction in inter- and intra-individual microbiota heterogeneity, suggesting a stabilizing effect on the gut microbiota. The severity of respiratory symptoms did not directly correlate with the cRG-I-induced microbial changes, but several dominant groups of the Ruminococcaceae family and microbiota richness were positively associated with a reduced and hence desired post-infection response. Thus, the present results on the modulation of the gut microbiota composition support the previously demonstrated immunomodulatory and protective effect of cRG-I during a common cold infection.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Voluntarios Sanos , Pectinas , Humanos , Pectinas/administración & dosificación , Pectinas/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Adulto , Método Doble Ciego , Femenino , Adulto Joven , Rhinovirus/efectos de los fármacos , Persona de Mediana Edad , Heces/microbiología , Bifidobacterium/efectos de los fármacos
18.
Food Funct ; 15(8): 4095-4108, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38563760

RESUMEN

Aging is a degenerative disease in which organisms and neurological functions decline. Emerging research has underscored the vital role of the gut microbiota in age-related processes. However, the identification of aging-associated core microbiota remains limited. In this investigation, we isolated a strain of B. pseudocatenulatum NCU-08 from the feces of centenarians and assessed its impact on aging using a mouse model induced by D-gal. Our study revealed the exceptional probiotic attributes of B. pseudocatenulatum NCU-08. Administration of B. pseudocatenulatum NCU-08 significantly ameliorated age-related memory impairment, motor dysfunction, and anxiety-like behaviors in aging mice (p < 0.01). Moreover, tissue staining analysis demonstrated that B. pseudocatenulatum NCU-08 reduced the intensity of SA-ß-gal-positive in the hippocampus of aging mice. It also reversed pathological damage and structural abnormalities in brain and intestinal tissue. B. pseudocatenulatum NCU-08 inhibited neuroinflammation induced by TLR4/NF-κB (p < 0.01) and preserved the blood-brain barrier integrity by activating the AMPK/Sirt1 pathway (p < 0.05). Furthermore, it mitigated neuronal apoptosis and oxidative stress by upregulating the PI3K/AKT signaling pathway (p < 0.01) and enhancing the activities of antioxidant enzymes, including GSH-Px (p < 0.01), SOD (p < 0.01), and CAT (p < 0.01). Besides, analysis of 16S rRNA sequencing data demonstrated that treatment with B. pseudocatenulatum NCU-08 restored intestinal microbiota homeostasis after senescence. It enhanced the abundance of beneficial bacteria while suppressing the growth of pathogenic microorganisms. In summary, our study unveiled that this novel strain of B. pseudocatenulatum NCU-08 exerts anti-aging effects through regulating the AMPK/Sirt1 pathway and intestinal microbiota. It holds promise as a functional food for promoting anti-aging effects and offers a novel approach to address aging and associated metabolic disorders.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Envejecimiento , Bifidobacterium , Microbioma Gastrointestinal , Probióticos , Transducción de Señal , Sirtuina 1 , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Sirtuina 1/metabolismo , Sirtuina 1/genética , Ratones , Probióticos/farmacología , Transducción de Señal/efectos de los fármacos , Masculino , Proteínas Quinasas Activadas por AMP/metabolismo , Humanos , Ratones Endogámicos C57BL , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos
19.
Food Res Int ; 184: 114246, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38609225

RESUMEN

Food-derived mucins are glycoproteins rich in sialic acid, but their digestive properties and potential health benefits for humans have been scarcely investigated. In this work, ovomucin (OVM, rich in N-acetylneuraminic acid, about 3 %), porcine small intestinal mucin (PSIM, rich in N-glycolylneuraminic acid, about 1 %), the desialylated OVM (AOVM) and the desialylated PSIM (APSIM) were selected to examine their digestion and their impact on the gut microbiota of elderly individuals. The results shown that, the proportion of low-molecular-weight proteins increased after simulated digestion of these four mucins, with concomitant comparable antioxidant activity observed. Desialylation markedly increased the degradation and digestion rate of mucins. In vitro fecal fermentation was conducted with these mucins using fecal samples from individuals of different age groups: young, low-age and high-age elderly. Fecal fermentation with mucin digestive solution stimulated the production of organic acids in the group with fecal sample of the elderly individuals. Among them, the OVM group demonstrated the most favorable outcomes. The OVM and APSIM groups elevated the relative abundance of beneficial bacteria such as Lactobacillus and Bifidobacterium, while diminishing the presence of pathogenic bacteria such as Klebsiella. Conversely, the probiotic effects of AOVM and PSIM were attenuated or even exhibited adverse effects. Hence, mucins originating from different sources and possessing distinct glycosylation patterns exhibit diverse biological functions. Our findings can offer valuable insights for developing a well-balanced and nutritious diet tailored to the elderly population.


Asunto(s)
Microbioma Gastrointestinal , Mucinas , Humanos , Anciano , Animales , Porcinos , Dieta , Alimentos , Bifidobacterium
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA