Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 956
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 184(15): 3884-3898.e11, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34143954

RESUMEN

Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon ß (IFNß) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.


Asunto(s)
Bifidobacterium/fisiología , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/microbiología , Antibacterianos/farmacología , Biomarcadores/metabolismo , Lactancia Materna , Linfocitos T CD4-Positivos/inmunología , Polaridad Celular , Proliferación Celular , Citocinas/metabolismo , Heces/química , Heces/microbiología , Galectina 1/metabolismo , Microbioma Gastrointestinal , Humanos , Indoles/metabolismo , Recién Nacido , Inflamación/sangre , Inflamación/genética , Mucosa Intestinal/inmunología , Metaboloma , Leche Humana/química , Oligosacáridos/metabolismo , Células Th17/inmunología , Células Th2/inmunología , Agua
2.
Compr Rev Food Sci Food Saf ; 23(5): e13431, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39165140

RESUMEN

Human milk oligosaccharides (HMOs) are an evolutionarily significant advantage bestowed by mothers for facilitating the development of the infant's gut microbiota. They can avoid absorption in the stomach and small intestine, reaching the colon successfully, where they engage in close interactions with gut microbes. This process also enables HMOs to exert additional prebiotic effects, including regulating the mucus layer, promoting physical growth and brain development, as well as preventing and mitigating conditions such as NEC, allergies, and diarrhea. Here, we comprehensively review the primary ways by which gut microbiota, including Bifidobacteria and other genera, utilize HMOs, and we classify them into five central pathways. Furthermore, we emphasize the metabolic benefits of bacteria consuming HMOs, particularly the recently identified intrinsic link between HMOs and the metabolic conversion of tryptophan to indole and its derivatives. We also examine the extensive probiotic roles of HMOs and their recent research advancements, specifically concentrating on the unsummarized role of HMOs in regulating the mucus layer, where their interaction with the gut microbiota becomes crucial. Additionally, we delve into the principal tools used for functional mining of new HMOs. In conclusion, our study presents a thorough analysis of the interaction mechanism between HMOs and gut microbiota, emphasizing the cooperative utilization of HMOs by gut microbiota, and provides an overview of the subsequent probiotic effects of this interaction. This review provides new insights into the interaction of HMOs with the gut microbiota, which will inform the mechanisms by which HMOs function.


Asunto(s)
Microbioma Gastrointestinal , Leche Humana , Oligosacáridos , Prebióticos , Humanos , Microbioma Gastrointestinal/fisiología , Leche Humana/química , Leche Humana/microbiología , Oligosacáridos/química , Probióticos , Lactante , Bacterias/metabolismo , Bifidobacterium/fisiología
3.
J Gastroenterol Hepatol ; 38(7): 1089-1098, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37119032

RESUMEN

BACKGROUND AND AIM: Disturbance of gut microbiota is associated with pathological change in multiple diseases. Probiotics can improve symptoms and exert clinical effects via regulation of gastrointestinal microecological environments, and a probiotic product commonly dispensed by Chinese physicians is a combination of live Bifidobacterium, Lactobacillus, and Enterococcus in powder/capsule form. It contains three strains-of Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis-which can act synergistically to balance the microbiome, regulate immunity, and repair the gut barrier. Although this product has been proven safe and effective in clinical practice, uncertainty remains regarding its use to treat digestive system diseases. To date, there have been no reference standards to guide clinical practice and no relevant expert consensus on this product, in China. METHODS: Following a literature search, evidence was graded and classified according to the grading of recommendations assessment, development, and evaluation (GRADE) system and consensus was secured from a panel of 52 experts. RESULTS: An expert consensus has been formed, on the clinical application of live combined Bifidobacterium, Lactobacillus, and Enterococcus in various digestive system diseases, to provide reference for its clinical use. CONCLUSIONS: Bifidobacterium triple viable powder/capsule may offer benefits, by regulating the balance of intestinal microecology. It can be used for the treatment and prevention of various digestive system diseases with good overall safety; further research is needed to confirm its application in other contexts. The recommendations in this consensus will be confirmed or refined in light of future research and clinical practice.


Asunto(s)
Enfermedades del Sistema Digestivo , Probióticos , Humanos , Bifidobacterium/fisiología , Consenso , Pueblos del Este de Asia , Enterococcus , Lactobacillus/fisiología , Polvos , Probióticos/uso terapéutico , Cápsulas , Microbioma Gastrointestinal
4.
BMC Gastroenterol ; 22(1): 100, 2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35247974

RESUMEN

BACKGROUND: Oral administration of health-promoting bacteria is increasingly used in clinical practise. These bacteria have anti-inflammatory characteristics and modulate the immune system without major reported side effects. The mechanisms of action are not yet fully defined. Our aim was to study systemic effects of probiotics by measurements of leukocytes as well as local effects on rectal mucosal biopsies after adding a standardized inflammatory stimulus in vitro. METHODS: Fourteen healthy subjects were randomized to receive 1010 colony forming units/day orally of the probiotic strain Lactiplantibacillus plantarum 299 (Lp299), n = 7, or Bifidobacterium infantis CURE21 (CURE21), n = 7, for six weeks. Rectal biopsies were taken before and after ingestion of either probiotic strain product, for stimulation in vitro with tumour necrosis factor alpha (TNF-α) at 10 and 100 ng/ml respectively up to 8 h. Blood tests were sampled before and after treatment. Lactate dehydrogenase (LDH) confirmed viable tissue. RESULTS: Composition of the intestinal microbiota was not changed. Systemic leukocytes decreased after administration of CURE21 (P<0.05) and Lp299 (P<0.01). Levels of the pro-inflammatory cytokine IL-6 in rectal mucosa after stimulation with TNF-α were attenuated after ingestion of Lp299. No effect was seen with CURE21. CONCLUSIONS: Administration of these probiotic strains to healthy humans show both a systemic and local reduction of inflammatory response by lowering leukocyte counts, and for Lp299 IL-6 levels in rectal mucosa. Probiotics may play an important role in the reduction of inflammatory responses expected after trauma during surgery or after pelvic irradiation. Trial registration Clinical Trials, registration number NCT01534572, retrospectively registered ( http://www.clinicaltrials.gov ).


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Bifidobacterium/fisiología , Citocinas , Humanos , Mucosa Intestinal , Leucocitos , Probióticos/uso terapéutico
5.
Oral Dis ; 28(6): 1723-1732, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33749084

RESUMEN

OBJECTIVE: Probiotics participate in regulating oral microbiota and reducing the prevalence of oral diseases; however, clinical research on probiotics is insufficient. Therefore, in this study, we performed in vitro screening of potential oral protective probiotic strains and then evaluated the clinical efficacy of the selected strains on maintaining oral health. MATERIALS AND METHODS: Fifty healthy individuals were recruited and randomly assigned into the placebo group and probiotics group, which included three strains of probiotics, Lactobacillus salivarius subs. salicinius AP-32, Lactobacillus paracasei ET-66, and Lactobacillus plantarum LPL28. Each group was blindly administered placebo or probiotics for four weeks. RESULTS: Next-generation sequencing results showed that the oral microbiota of Lactobacillus salivarius in the oral cavity were significantly increased in subjects supplemented with mixed probiotic lozenges. The anti-bacterial activities of viable probiotics were observed within two weeks. Both IgA levels and Lactobacillus and Bifidobacterium abundances in the oral cavity were significantly increased in the experimental groups, along with a reduced formation of plaque. Most participants reported that their oral health conditions and intestinal symptoms had improved. CONCLUSIONS: Overall, our clinical study suggests that oral probiotic lozenges may enhance oral immunity, modulate oral microbiota, and improve oral health.


Asunto(s)
Placa Dental , Probióticos , Bifidobacterium/fisiología , Placa Dental/microbiología , Humanos , Inmunidad , Lactobacillus/fisiología , Probióticos/uso terapéutico
6.
J Dairy Sci ; 105(9): 7308-7321, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35931487

RESUMEN

We evaluated the potential of whey protein hydrolysate as a lyoprotectant for maintaining the cell viability of Bifidobacterium animalis ssp. lactis Probio-M8 during freeze-drying and subsequent storage. The moisture content and water activity of the lyophilized samples treated by different concentrations of whey protein hydrolysate were ≤5.23 ± 0.33 g/100 g and ≤0.102 ± 0.003, respectively. During storage at 25°C and 30°C, whey protein hydrolysate had a stronger protective effect on B. lactis Probio-M8 than the same concentration of whey protein. Using the Excel tool GinaFit, we estimated the microbial inactivation kinetics during storage. Whey protein hydrolysate reduced cell damage caused by an increase in temperature. Whey protein hydrolysate could protect cells by increasing the osmotic pressure as a compatible solute. Whey protein hydrolysate improved cell membrane integrity and reduced the amounts of reactive oxygen species and malondialdehyde produced. The findings indicated that whey protein hydrolysate was a novel antioxidant lyoprotectant that could protect probiotics during freeze-drying and storage.


Asunto(s)
Bifidobacterium animalis , Probióticos , Bifidobacterium/fisiología , Liofilización/veterinaria , Probióticos/metabolismo , Hidrolisados de Proteína/farmacología , Suero Lácteo
7.
Int J Mol Sci ; 23(18)2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36142723

RESUMEN

Previous works have described the activity of Bifidobacterium longum subsp. infantis CECT 7210 (also commercially named B. infantis IM-1®) against rotavirus in mice and intestinal pathogens in piglets, as well as its diarrhea-reducing effect on healthy term infants. In the present work, we focused on the intestinal immunomodulatory effects of B. infantis IM-1® and for this purpose we used the epithelial cell line isolated from colorectal adenocarcinoma Caco-2 and a co-culture system of human dendritic cells (DCs) from peripheral blood together with Caco-2 cells. Single Caco-2 cultures and Caco-2: DC co-cultures were incubated with B. infantis IM-1® or its supernatant either in the presence or absence of Escherichia coli CECT 515. The B. infantis IM-1® supernatant exerted a protective effect against the cytotoxicity caused by Escherichia coli CECT 515 on single cultures of Caco-2 cells as viability reached the values of untreated cells. B. infantis IM-1® and its supernatant also decreased the secretion of pro-inflammatory cytokines by Caco-2 cells and the co-cultures incubated in the presence of E. coli CECT 515, with the response being more modest in the latter, which suggests that DCs modulate the activity of Caco-2 cells. Overall, the results obtained point to the immunomodulatory activity of this probiotic strain, which might underlie its previously reported beneficial effects.


Asunto(s)
Infecciones por Escherichia coli , Probióticos , Animales , Bifidobacterium/fisiología , Bifidobacterium longum subspecies infantis/metabolismo , Células CACO-2 , Citocinas/metabolismo , Escherichia coli/metabolismo , Humanos , Lactante , Ratones , Probióticos/farmacología , Porcinos
8.
Mol Biol Rep ; 48(1): 951-960, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33389533

RESUMEN

Today, the increasing rate of cancer-related mortality, has rendered cancer a major global challenge, and the second leading cause of death worldwide. Conventional approaches in the treatment of cancer mainly include chemotherapy, surgery, immunotherapy, and radiotherapy. However, these approaches still come with certain disadvantages, including drug resistance, and different side effects such as gastrointestinal (GI) irritation (e.g., diarrhea, mucositis). This has encouraged scientists to look for alternative therapeutic methods and adjuvant therapies for a more proper treatment of malignancies. Application of probiotics as an adjuvant therapy in the clinical management of cancer appears to be a promising strategy, with several notable advantages, e.g., increased safety, higher tolerance, and negligible GI side effects. Both in vivo and in vitro analyses have indicated the active role of yeast probiotics in mitigating the rate of cancer cell proliferation, and the induction of apoptosis through regulating the expression of cancer-related genes and cellular pathways. Strain-specific anti-cancer activities of yeast probiotics strongly suggest that their administration along with the current cancer therapies may be an efficient method to reduce the side effects of these approaches. The main purpose of this article is to evaluate the efficacy of yeast probiotics in alleviating the adverse effects associated with cancer therapies.


Asunto(s)
Neoplasias Colorrectales/terapia , Terapia Combinada/métodos , Diarrea/terapia , Probióticos/uso terapéutico , Saccharomyces/fisiología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bifidobacterium/fisiología , Quimioradioterapia Adyuvante/métodos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Diarrea/inmunología , Diarrea/microbiología , Diarrea/patología , Manejo de la Enfermedad , Humanos , Inmunoterapia/métodos , Lactobacillaceae/fisiología
9.
J Appl Microbiol ; 130(4): 1357-1367, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32970888

RESUMEN

AIM OF THE STUDY: Given that phagocytic cells are main players of the host immune response, we studied the interaction of bifidobacteria with monocytic THP-1 cells in nonopsonic conditions. METHODS AND RESULTS: Association/internalization, cell response (expression of HLA-DR and TLR2), M1/M2 macrophage polarization and colocalization of micro-organisms with Lysotracker or transferrin were evaluated. Screening with eight Bifidobacterium strains showed two patterns of interactions with THP-1 cells: high and low association and phagocytosis. Two strains with different surface properties were further studied: B. bifidum CIDCA 5310 and B. adolescentis CIDCA 5317. Strain CIDCA 5310 showed higher levels of colocalization in lysosome than strain CIDCA 5317. Both strains stimulated TLR2 expression. Strain CIDCA 5317 significantly increases HLA-DR expression, however, when cells are stimulated with IFN-γ, strain CIDCA 5310 induces the highest value of expression. Noteworthy, strain CIDCA 5310 was able to upregulate both M1 and M2 markers of macrophage polarization. CONCLUSIONS: Our results demonstrate that bifidobacteria from human origin show different patterns of interaction with phagocytic cells thus leading to different cell responses. These findings add further insight on the mechanisms involved in the biologic effects of probiotics. SIGNIFICANCE AND IMPACT OF THE STUDY: Knowledge of the interaction of bifidobacteria with key players of the host immune response is paramount for the understanding of the mechanisms involved in the beneficial effects.


Asunto(s)
Bifidobacterium/fisiología , Macrófagos/fisiología , Probióticos/farmacología , Comunicación Celular , Línea Celular , Humanos , Interferón-alfa/genética , Interferón-alfa/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/fisiología , Fagocitosis/efectos de los fármacos , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología
10.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33806135

RESUMEN

The establishment of the gut microbiota poses implications for short and long-term health. Bifidobacterium is an important taxon in early life, being one of the most abundant genera in the infant intestinal microbiota and carrying out key functions for maintaining host-homeostasis. Recent metagenomic studies have shown that different factors, such as gestational age, delivery mode, or feeding habits, affect the gut microbiota establishment at high phylogenetic levels. However, their impact on the specific bifidobacterial populations is not yet well understood. Here we studied the impact of these factors on the different Bifidobacterium species and subspecies at both the quantitative and qualitative levels. Fecal samples were taken from 85 neonates at 2, 10, 30, 90 days of life, and the relative proportions of the different bifidobacterial populations were assessed by 16S rRNA-23S rRNA internal transcribed spacer (ITS) region sequencing. Absolute levels of the main species were determined by q-PCR. Our results showed that the bifidobacterial population establishment is affected by gestational age, delivery mode, and infant feeding, as it is evidenced by qualitative and quantitative changes. These data underline the need for understanding the impact of perinatal factors on the gut microbiota also at low taxonomic levels, especially in the case of relevant microbial populations such as Bifidobacterium. The data obtained provide indications for the selection of the species best suited for the development of bifidobacteria-based products for different groups of neonates and will help to develop rational strategies for favoring a healthy early microbiota development when this process is challenged.


Asunto(s)
Bifidobacterium/fisiología , Microbioma Gastrointestinal , Ciencias de la Nutrición del Niño , ADN Intergénico/genética , Heces/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Filogenia , ARN Ribosómico 16S/genética , ARN Ribosómico 23S/genética , Temperatura
11.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34502130

RESUMEN

Bifidobacteria are some of the major agents that shaped the immune system of many members of the animal kingdom during their evolution. Over recent years, the question of concrete mechanisms underlying the immunomodulatory properties of bifidobacteria has been addressed in both animal and human studies. A possible candidate for this role has been discovered recently. The PFNA cluster, consisting of five core genes, pkb2, fn3, aaa-atp, duf58, tgm, has been found in all gut-dwelling autochthonous bifidobacterial species of humans. The sensory region of the species-specific serine-threonine protein kinase (PKB2), the transmembrane region of the microbial transglutaminase (TGM), and the type-III fibronectin domain-containing protein (FN3) encoded by the I gene imply that the PFNA cluster might be implicated in the interaction between bacteria and the host immune system. Moreover, the FN3 protein encoded by one of the genes making up the PFNA cluster, contains domains and motifs of cytokine receptors capable of selectively binding TNF-α. The PFNA cluster could play an important role for sensing signals of the immune system. Among the practical implications of this finding is the creation of anti-inflammatory drugs aimed at alleviating cytokine storms, one of the dire consequences resulting from SARS-CoV-2 infection.


Asunto(s)
Proteínas Bacterianas/genética , Bifidobacterium/fisiología , COVID-19/terapia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , COVID-19/inmunología , COVID-19/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/química , Citocinas/metabolismo , Humanos , Sistema Inmunológico , Operón/genética , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , SARS-CoV-2/aislamiento & purificación
12.
Bull Exp Biol Med ; 172(2): 164-168, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34855091

RESUMEN

We studied the effect of bacterial wall peptidoglycan of 7 bacterial species on the competitive properties of human-associated microorganisms. Addition of peptidoglycan to the culture medium did not change the growth characteristics of the test cultures; however, an increase in the antagonism and hydrophobicity of Bifidobacterium sp. and Enterococcus sp. was observed, while the effect on enterobacteria was predominantly indifferent or inhibitory. The effect did not depend much on the source of peptidoglycan and was equally manifested on both indigenous and probiotic strains. The observed new property of peptidoglycan indicates its participation in the formation and functioning of microbiota. The obtained data on the regulation of the properties of microorganisms provide new possibilities for the correction and maintenance of host homeostasis through host-associated microbiota.


Asunto(s)
Antibiosis/fisiología , Pared Celular/fisiología , Peptidoglicano/metabolismo , Bacillus subtilis/fisiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/fisiología , Bifidobacterium/fisiología , Candida/fisiología , Pared Celular/química , Pared Celular/metabolismo , Enterobacter/fisiología , Enterococcus faecalis/fisiología , Escherichia coli/fisiología , Femenino , Humanos , Lacticaseibacillus casei/fisiología , Técnicas Microbiológicas , Peptidoglicano/análisis , Staphylococcus aureus/fisiología
13.
BMC Microbiol ; 20(1): 144, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32503418

RESUMEN

BACKGROUND: Probiotics are effective to rectify the imbalanced gut microbiota in the diseased cohorts. Two Bifidobacterium strains (LI09 and LI10) were found to alleviate D-galactosamine-induced liver damage (LD) in rats in our previous work. A series of bioinformatic and statistical analyses were performed to determine the vital bacteria in the gut microbiotas altered by the LI09 or LI10 in rats. RESULTS: Two groups of representative phylotypes could distinguish the gut microbiotas of LI09 or LI10 groups from the other groups. Among them, OTU170_Porphyromonadaceae acted as a gatekeeper in LI09 group, while OTU12_Bacteroides was determined with multiple correlations in the gut network of LI10 group. Multiple reduced OTUs associated with LC and increased OTUs associated with health were determined in LI09 or LI10 groups, among which, increased OTU51_Barnesiella and reduced OTU99_Barnesiella could be associated with the protective effects of both the two probiotics. The gut microbiotas in LI09, LI10 and positive control groups were clustered into three clusters, i.e., Cluster_1_Microbiota, Cluster_2_Microbiota and Cluster_3_Microbiota, by Partition Around Medoids clustering analysis. Cluster_2_Microbiota was determined at least dysbiotic status due to its greatest LD dysbiosis ratio, lowest levels of liver function variables and plasma cytokines compared with the two other clustered microbiotas, suggesting the treated rats in Cluster_2 were at better health status. CONCLUSION: Our findings suggest that OTU170_Porphyromonadaceae and OTU12_Bacteroides are vital in the gut microbiotas altered by LI09 and LI10. Characteristics of the LD cohorts treated by LI09 or LI10 at different gut microbial colonization states could help monitor the cohorts' health status.


Asunto(s)
Bacterias/clasificación , Bifidobacterium/fisiología , Enfermedad Hepática Inducida por Sustancias y Drogas/dietoterapia , Probióticos/administración & dosificación , Análisis de Secuencia de ADN/métodos , Animales , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación , Bifidobacterium/clasificación , ADN Bacteriano/genética , Galactosamina/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Filogenia , Probióticos/efectos adversos , Ratas
14.
BMC Microbiol ; 20(1): 357, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33225894

RESUMEN

BACKGROUND: Bifidobacterium longum subsp. infantis (B. infantis) is a commensal bacterium that colonizes the gastrointestinal tract of breast-fed infants. B. infantis can efficiently utilize the abundant supply of oligosaccharides found in human milk (HMO) to help establish residence. We hypothesized that metabolites from B. infantis grown on HMO produce a beneficial effect on the host. RESULTS: In a previous study, we demonstrated that B. infantis routinely dominated the fecal microbiota of a breast fed Bangladeshi infant cohort (1). Characterization of the fecal metabolome of binned samples representing high and low B. infantis populations from this cohort revealed higher amounts of the tryptophan metabolite indole-3-lactic acid (ILA) in feces with high levels of B. infantis. Further in vitro analysis confirmed that B. infantis produced significantly greater quantities of the ILA when grown on HMO versus lactose, suggesting a growth substrate relationship to ILA production. The direct effects of ILA were assessed in a macrophage cell line and intestinal epithelial cell lines. ILA (1-10 mM) significantly attenuated lipopolysaccharide (LPS)-induced activation of NF-kB in macrophages. ILA significantly attenuated TNF-α- and LPS-induced increase in the pro-inflammatory cytokine IL-8 in intestinal epithelial cells. ILA increased mRNA expression of the aryl hydrogen receptor (AhR)-target gene CYP1A1 and nuclear factor erythroid 2-related factor 2 (Nrf2)-targeted genes glutathione reductase 2 (GPX2), superoxide dismutase 2 (SOD2), and NAD(P) H dehydrogenase (NQO1). Pretreatment with either the AhR antagonist or Nrf-2 antagonist inhibited the response of ILA on downstream effectors. CONCLUSIONS: These findings suggest that ILA, a predominant metabolite from B. infantis grown on HMO and elevated in infant stool high in B. infantis, and protects gut epithelial cells in culture via activation of the AhR and Nrf2 pathway.


Asunto(s)
Antiinflamatorios/farmacología , Bifidobacterium/fisiología , Indoles/farmacología , Microbiota , Animales , Antiinflamatorios/análisis , Bifidobacterium/metabolismo , Línea Celular , Endotoxinas/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Heces/química , Heces/microbiología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Indoles/análisis , Lactante , Interleucina-8/metabolismo , Lactosa/metabolismo , Activación de Macrófagos/efectos de los fármacos , Ratones , Leche Humana/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Oligosacáridos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos
15.
FASEB J ; 33(10): 11235-11246, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31299175

RESUMEN

Proton pump inhibitors (PPIs) are used by millions of patients for the treatment of stomach acid-reflux diseases. Although PPIs are generally considered safe, about 13% of the users develop hypomagnesemia. Despite rising attention for this issue, the underlying mechanism is still unknown. Here, we examine whether the gut microbiome is involved in the development of PPI-induced hypomagnesemia in wild-type C57BL/6J mice. After 4 wk of treatment under normal or low dietary Mg2+ availability, omeprazole significantly reduced serum Mg2+ levels only in mice on a low-Mg2+ diet without affecting the mRNA expression of colonic or renal Mg2+ transporters. Overall, 16S rRNA gene sequencing revealed a lower gut microbial diversity in omeprazole-treated mice. Omeprazole induced a shift in microbial composition, which was associated with a 3- and 2-fold increase in the abundance of Lactobacillus and Bifidobacterium, respectively. To examine the metabolic consequences of these microbial alterations, the colonic composition of organic acids was evaluated. Low dietary Mg2+ intake, independent of omeprazole treatment, resulted in a 10-fold increase in formate levels. Together, these results imply that both omeprazole treatment and low dietary Mg2+ intake disturb the gut internal milieu and may pose a risk for the malabsorption of Mg2+ in the colon.-Gommers, L. M. M., Ederveen, T. H. A., van der Wijst, J., Overmars-Bos, C., Kortman, G. A. M., Boekhorst, J., Bindels, R. J. M., de Baaij, J. H. F., Hoenderop, J. G. J. Low gut microbiota diversity and dietary magnesium intake are associated with the development of PPI-induced hypomagnesemia.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Magnesio/metabolismo , Inhibidores de la Bomba de Protones/efectos adversos , Animales , Bifidobacterium/fisiología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Dieta , Lactobacillus/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Omeprazol/efectos adversos , ARN Ribosómico 16S/metabolismo
16.
Arch Microbiol ; 202(7): 1627-1637, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32266422

RESUMEN

Water kefir is a fermented beverage employing a natural microbial consortium, which harbours bifidobacteria, namely Bifidobacterium aquikefiri and Bifidobacterium tibiigranuli. However, little information is available on their metabolic properties or role in the consortium. In this study, we combined genomic and physiologic investigations to predict and characterize the properties of these organisms and their possible role in the consortium. When comparing the genomes of these psychrotrophic organisms with that of the three selected mesophilic probiotic Bifidobacterium strains, we could find 143 genes shared by the 3 known isolates of bifidobacteria from water kefir that do not occur in the probiotic strains. These include genes involved in acid and oxygen tolerance. In addition, their genomically predicted carbohydrate usage and transport suggest adaptation to sucrose and other plant-related sugars. Furthermore, they proved prototrophic for all amino acids in vitro, which enables them to cope with the strong amino acid limitation in water kefir.


Asunto(s)
Bifidobacterium/fisiología , Genoma Bacteriano/genética , Kéfir/microbiología , Probióticos , Bifidobacterium/genética , Bifidobacterium/aislamiento & purificación , Genómica , Consorcios Microbianos/genética , Microbiología del Agua
17.
Nutr Cancer ; 72(8): 1355-1365, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31608714

RESUMEN

Lactobacillus spp. and Bifidobacterium spp. was used to protect against gastrointestinal disorders. The present study evaluated the effects of probiotic mixture (PM) containing Lactobacillus spp. and Bifidobacterium spp. on intestinal mucositis induced by 5-fluorouracil (5-FU). Swiss male mice (25-30 g) were treated with 5-FU (450 mg/kg, ip) and were orally administered (PM). Probiotic mixture 1 (PM-1) is a mixture of two probiotics (Lactobacillus acidophilus and Bifidobacterium lactis) and probiotic mixture 2 (PM-2) is a mixture of four probiotics (Lactobacillus acidophilus, Lactobacillus paracasei, Lactobacillus rhamnosus, and Bifidobacterium lactis). PM-1 and PM-2 decreased histopathological scores in the duodenum and jejunum after mucositis. PM-2 attenuated 5-FU-induced weight loss. On the other hand, PM-1 did not exert a significant effect on weight loss. Both probiotics mixture increased the villus/crypt ratio in all intestinal segments, increased GSH levels in the duodenum and jejunum, and reduced the MDA, MPO, TNF-α, and IL-6 levels in the duodenum, jejunum, and ileum. PM-2 attenuated the delay in gastric emptying. PM-1 and PM-2 prevented epithelial injury in intestinal mucositis by 5-FU, demonstrating the potential use of these probiotics as therapeutic agents against intestinal mucositis.


Asunto(s)
Bifidobacterium/fisiología , Fluorouracilo/efectos adversos , Intestinos/efectos de los fármacos , Lactobacillus/fisiología , Mucositis/prevención & control , Neoplasias/tratamiento farmacológico , Probióticos/farmacología , Animales , Antimetabolitos Antineoplásicos/efectos adversos , Citocinas/metabolismo , Glutatión/metabolismo , Intestinos/patología , Masculino , Ratones , Mucositis/inducido químicamente , Neoplasias/patología , Distribución Aleatoria
18.
Biosci Biotechnol Biochem ; 84(12): 2558-2568, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32862788

RESUMEN

In this study, three strains of lactobacilli and bifidobacteria originally isolated from healthy infants, were tested for their abilities to activate RAW264.7 cells. Gene expression and cytokine production of interleukin-10 (IL-10) of RAW264.7 cells were evaluated. The activation of extracellular regulated protein kinases 1/2 (ERK1/2), p38, and nuclear factor-κB (NK-κB) were also assessed. These results suggest lactobacilli and bifidobacteria in infants may promote production of IL-10 in macrophages, conferring a protective effect in hosts suffering from inflammation. Dimerization of TLR2 and MyD88 and subsequent phosphorylation of the key downstream signaling molecules, such as MAPKs and NK-κB, may be one of the key underlying mechanisms of activation of macrophages by these microbes. Bifidobacteria and lactobacilli induced macrophages to secrete IL-10 in a different manner, which may relate to their abilities to activate key signaling pathways mediated by TLR2 and MyD88.


Asunto(s)
Bifidobacterium/fisiología , Interleucina-10/metabolismo , Intestinos/microbiología , Lactobacillus/fisiología , Activación de Macrófagos , Animales , Humanos , Lactante , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Células RAW 264.7 , Transducción de Señal , Receptor Toll-Like 2/metabolismo
19.
J Dairy Sci ; 103(5): 4013-4025, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32113772

RESUMEN

Breast milk is the main source of nutrition for infants; it contains considerable microflora that can be transmitted to the infant endogenously or by breastfeeding, and it plays an important role in the maturation and development of the immune system. In this study, we isolated and identified lactic acid bacteria (LAB) from human colostrum, and screened 2 strains with probiotic potential. The LAB isolated from 40 human colostrum samples belonged to 5 genera: Lactobacillus, Bifidobacterium, Streptococcus, Enterococcus, and Staphylococcus. We also isolated Propionibacterium and Actinomyces. We identified a total of 197 strains of LAB derived from human colostrum based on their morphology and 16S rRNA sequence, among them 8 strains of Bifidobacterium and 10 strains of Lactobacillus, including 3 Bifidobacterium species and 4 Lactobacillus species. The physiological and biochemical characteristics of strains with good probiotic characteristics were evaluated. The tolerances of some of the Bifidobacterium and Lactobacillus strains to gastrointestinal fluid and bile salts were evaluated in vitro, using the probiotic strains Bifidobacterium lactis BB12 and Lactobacillus rhamnosus GG as controls. Among them, B. lactis Probio-M8 and L. rhamnosus Probio-M9 showed survival rates of 97.25 and 78.33% after digestion for 11 h in artificial gastrointestinal juice, and they exhibited growth delays of 0.95 and 1.87 h, respectively, in 0.3% bile salts. These two strains have the potential for application as probiotics and will facilitate functional studies of probiotics in breast milk and the development of human milk-derived probiotics.


Asunto(s)
Bifidobacterium/fisiología , Calostro/microbiología , Lactobacillales/fisiología , Probióticos , Animales , Bifidobacterium/aislamiento & purificación , Bifidobacterium animalis/aislamiento & purificación , Enterococcus/aislamiento & purificación , Femenino , Humanos , Lactobacillales/aislamiento & purificación , Lactobacillus/aislamiento & purificación , Embarazo , Probióticos/aislamiento & purificación , ARN Ribosómico 16S
20.
Ecotoxicol Environ Saf ; 198: 110616, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32334202

RESUMEN

Honeybees are prone to poisoning after collecting jujube nectar during the jujube flowering period ('honeybee's jujube flower disease'). To explore the mechanism of honeybee poisoning, the gut microbiota of honeybees undergoing the disease were characterised based on amplicon sequencing of the 16 S rRNA gene. Our results showed that the composition and diversity of the gut microbiota were significantly altered in diseased honeybees. We observed a decrease in the relative abundance of Proteobacteria and increased abundances of Firmicutes and Actinobacteria in the midgut and hindgut of diseased honeybees. Moreover, linear discriminant analysis (LDA) effect size revealed significantly selected enrichment of Fructobacillus and Snodgrassella in the midguts from diseased honeybees and Lactobacillus, Bifidobacterium, and Snodgrassella in the hindguts from diseased honeybees. Tax4Fun anylasis indicated that the functional potential of the diseased honeybee gut bacterial community was significantly changed relative to the healthy honeybee. Carbohydrate metabolism, nucleotides metabolism, amino acid synthesis metabolism, coenzyme and vitamins metabolism were increased, while energy metabolism and xenobiotic biodegradation and metabolism were decreased in the diseased honeybees. These results provide a new perspective for evaluating the response of honeybees to jujube flower disease based on changes in the intestinal microflora.


Asunto(s)
Bacterias/efectos de los fármacos , Abejas/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Néctar de las Plantas/toxicidad , Ziziphus/química , Animales , Bacterias/genética , Bifidobacterium/fisiología , Biodiversidad , Sistema Digestivo/microbiología , Microbioma Gastrointestinal/genética , Lactobacillus/fisiología , ARN Ribosómico 16S/genética , Ziziphus/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA