Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 953
Filtrar
1.
Mol Neurobiol ; 61(1): 498-509, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37639065

RESUMO

Despite the anatomical separation, strong evidence suggested a bidirectional association between gut microbiota and central nervous system. Cross-talk between gut microbiota and brain has an important role in the pathophysiology of neurodegenerative disorders and regenerative processes. However, choosing the appropriate probiotics and combination therapy of probiotics to provide a synergistic effect is very crucial. In the present study, we investigated the effect of Lactobacillus casei (L. casei) and Bifidobacterium breve (B. breve) on alternation performance, oxidant/antioxidant biomarkers, the extent of demyelination, and the expression level of HO-1, Nrf-2, Olig2, MBP, PDGFRα, and BDNF in cuprizone (CPZ)-induced demyelination model of rat corpus callosum. In order to induce this model, rats received oral administration of CPZ 0.6% w/w in corn oil for 28 days. Then, L. casei, B. breve, or their combinations were orally administrated for 28 days. Y maze test was performed to investigate the alternation performance. Oxidant/antioxidant biomarkers were determined by colorimetric methods. Extent of demyelination was investigated using FluoroMyelin staining. The genes' expression levels of antioxidant and myelin lineage cells were assessed by quantitative real time PCR. The results showed the probiotics supplementation significantly improve the alternation performance and antioxidant capacity in demyelinated corpus callosum. Interestingly, B. breve supplementation alleviated demyelination and oxidative stress levels more than the administration of L. casei alone or the combination of two probiotics. These observations suggest that B. breve could provide a supplementary strategy for the treatment of multiple sclerosis by increasing antioxidant capacity and remyelination.


Assuntos
Bifidobacterium breve , Doenças Desmielinizantes , Lacticaseibacillus casei , Probióticos , Ratos , Animais , Cuprizona/toxicidade , Antioxidantes , Bifidobacterium/fisiologia , Probióticos/farmacologia , Probióticos/uso terapêutico , Estresse Oxidativo , Doenças Desmielinizantes/induzido quimicamente , Biomarcadores , Oxidantes
2.
Sci Total Environ ; 912: 169556, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38135070

RESUMO

The widespread use of cyproconazole (CPZ) enhances food security but may pose potential risks to non-target organisms. Therefore, we applied Multi-omics techniques to reveal the response of the intestinal barrier to CPZ exposure and explore whether the Bifidobacterium intervention experiment can repair the damage. First, we found that exposure to CPZ at environmentally relevant concentrations led to intestinal injury phenotype, significantly down-regulated intestinal protein gene expression, and up-regulated pro-inflammatory gene expression, further causing intestinal dysbacteriosis and metabolic disorders. In particular, by combining analysis of gut microbiota and metabolites, we noticed acetate, a key metabolite, which decreased sharply after exposure to high concentration of CPZ. Expectedly, after supplementing with Bifidobacterium (a core bacterium that produces acetate), we noticed that the acetate content was quickly restored. Further, we also verified that the increase in acetate content after Bifidobacterium supplementation at least partially promoted IL-22 secretion, which in turn stimulated the secretion of ß-defensins (zfbd-1, zfbd-2, zfbd-3), thereby repairing the intestinal damage. In conclusion, our work confirms the potential of Bifidobacterium to improve intestinal damage and metabolic dysbiosis caused by CPZ exposure. It provides directional recommendations for the application of probiotics to repair the toxicological risk of pesticide exposure.


Assuntos
Microbioma Gastrointestinal , Triazóis , Peixe-Zebra , Animais , Bifidobacterium/fisiologia , Microbioma Gastrointestinal/genética , Acetatos
3.
Braz J Microbiol ; 54(3): 2197-2204, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37261620

RESUMO

The aim of the work was to evaluate antagonistic activity of Lactobacillus spp. and Bifidobacterium spp. in vitro against cariogenic Streptococcus mutans UA 159 and viability in chewing gum, during storage. Antagonistic activity was evaluated in vitro by the "spot on the lawn" test. Two bacteria were chosen and subjected to lyophilization and microencapsulation using the atomization method, containing polyvinylpyrrolidone polymer and lactose as encapsulating agents. For application in food matrices, four treatments were elaborated: chewing gum containing lyophilized B. lactis B94 (BLL), microencapsulated B. lactis B94 (BLE), lyophilized L. brevis (LBL), and microencapsulated L. brevis (LBE). Both microorganisms demonstrated a high capacity for inhibition against S. mutans, when compared to oral antiseptic chlorhexidine 0.2% in vitro, and according to the test of sensitivity profile to proteolytic enzymes, all the bacteria tested are producers of antimicrobial peptides, resulting in the inhibitory activity of the cariogenic bacterium. Furthermore, the viability of B. lactis B94 and L. brevis was maintained after microencapsulation, indicating that the process was efficient, with no significant difference (p < 0.05) between the results. And, in the chewing gum containing the bacteria during the storage period (33 days), it was found that cell immobilization did not significantly influence (p < 0.05) the counts of L. brevis but benefited the viability of B. lactis B94. Therefore, both probiotic bacteria are producers of antimicrobial substances with the ability to inhibit S. mutans, in vitro. The microencapsulation was considered efficient since it influenced the viability of B. lactis B94 (> 8 log CFU/g); however, the microencapsulation did not influence the viability of L. brevis since in both lyophilized and encapsulated form; the concentration of the bacteria remained above 8 log CFU/g during the storage period of the chewing gum.


Assuntos
Probióticos , Streptococcus mutans , Lactobacillus/fisiologia , Goma de Mascar , Bifidobacterium/fisiologia , Probióticos/farmacologia
4.
Cells ; 12(9)2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37174650

RESUMO

Gut microbiota dysbiosis with increased pathogenic bacteria and decreased beneficial bacteria is associated with colorectal cancer (CRC) development. This study examined the effect of a newly developed probiotic formula in modulating CRC-related bacteria. We developed a probiotic formula containing three bifidobacteria (B. adolescentis, B. longum, and B. bifidum) based on the identification of bacterial species that showed significant correlations with CRC-related bacteria including Fusobacterium nucleatum (Fn), Lachnoclostridium sp. m3, Clostridium hathewayi (Ch), and Bacteroides clarus (Bc). We co-cultured Fn with each bifidobacterium or the combined formula and examined the growth of Fn by qPCR. The three individual bifidobacteria significantly inhibited the growth of Fn compared to the control treatment (24~65% inhibition; all p < 0.001). The combination of the three bifidobacteria showed a greater inhibitory effect on Fn growth (70% inhibition) than the individual bifidobacteria (all p < 0.05). We further examined the effect of the probiotic formula in a pilot study of 72 subjects (40 on probiotics; 32 with no intervention) for 4 weeks and followed them up for 12 weeks. The relative fecal abundances of the bifidobacteria in the formula and the CRC-related markers (Fn, m3, Ch, and Bc) were quantitated by qPCR before and after the intervention, and the combined CRC risk score (4Bac; Fn, m3, Ch, and Bc) was evaluated. Subjects with probiotics intervention showed significantly increased abundances of the bifidobacteria from week 2 to week 5 compared to baseline (p < 0.05), and the abundances dropped to baseline levels after the cessation of the intervention. There were significant decreases in the levels of CRC-related markers (Fn and m3) and the CRC risk score (4Bac) from week 2 to week 12 compared to baseline levels (p < 0.05) in the intervention group but not in the control group. A novel probiotic formula containing B. adolescentis, B. longum, and B. bifidum was effective in inhibiting the growth of F. nucleatum in vitro and improving the gut microbial environment against CRC development.


Assuntos
Neoplasias Colorretais , Probióticos , Humanos , Projetos Piloto , Probióticos/uso terapêutico , Fezes/microbiologia , Bifidobacterium/fisiologia
5.
J Gastroenterol Hepatol ; 38(7): 1089-1098, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37119032

RESUMO

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.


Assuntos
Doenças do Sistema Digestório , Probióticos , Humanos , Bifidobacterium/fisiologia , Consenso , População do Leste Asiático , Enterococcus , Lactobacillus/fisiologia , Pós , Probióticos/uso terapêutico , Cápsulas , Microbioma Gastrointestinal
6.
Food Funct ; 14(1): 354-368, 2023 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-36511157

RESUMO

B. longum subsp. infantis is a subspecies of Bifidobacterium longum, and very few strains are shown to have immunomodulatory effects. In the present study, the improvement of dextran sulphate sodium (DSS)-induced colitis by four B. longum subsp. infantis strains was compared. The results showed that B. longum subsp. infantis FJSYZ1M3 could significantly decrease disease activity index (DAI), inhibit weight loss and colon shortening, and attenuate colon tissue damage in DSS-induced colitis mice. And B. longum subsp. infantis FJSYZ1M3 intervention improved the integrity of intestinal tight junctions, relieved mucus layer damage and inhibited epithelial cell apoptosis, thereby maintaining the intestinal barrier. Additionally, B. longum subsp. infantis FJSYZ1M3 significantly affected the levels of inflammatory cytokines IL-6, IL-1ß, and IL-10 in the colon, thus relieving inflammation in colitis mice. Furthermore, B. longum subsp. infantis FJSYZ1M3 could ameliorate gut microbiota disturbance caused by DSS exposure and increase the level of butyric acid in cecal contents. In general, these findings suggested that B. longum subsp. infantis FJSYZ1M3 alleviated DSS-induced colitis by maintaining the intestinal barrier, regulating inflammatory cytokines, and modifying the gut microbiota.


Assuntos
Bifidobacterium longum , Colite , Microbioma Gastrointestinal , Camundongos , Animais , Citocinas , Bifidobacterium/fisiologia , Colite/induzido quimicamente , Colite/microbiologia , Bifidobacterium longum subspecies infantis , Colo , Sulfato de Dextrana/efeitos adversos , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
7.
Appl Neuropsychol Adult ; 30(5): 552-560, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-34493130

RESUMO

A growing body of research studies the relationship between probiotic bacteria in the gut and the host organism, including the impact on cognitive functioning. Data from human studies are scarce; however, recent studies point toward the beneficial role of probiotics for cognitive functioning. One of the mechanisms involved in this relationship is the probiotic's ability to influence inflammation and immune response. The aim of this initial study was to investigate the effects of probiotic supplementation with Bifidobacterium Lactis BS01 and Lactobacillus Acidophilus LA02 on cognitive functioning in healthy, young adult females. A total of 53 participants aged 19-31 were enrolled, and 38 completed the trial. A 6-week probiotic or placebo treatment was conducted. Five measures of cognitive functioning were applied pre- and post-treatment. Both groups showed general improvement at the second assessment. Contrary to our hypothesis, the placebo group improved slightly, but significantly, in four out of five measures of cognitive functioning, with the exception of the Wisconsin Card Sorting Test (WCST). The supplementation group improved significantly in two measures of the WCST, compared to the placebo group. Similar results have been previously reported. Probiotic supplementation, while not harmful, might not be beneficial for cognition in the healthy population, or at least not universally.


Assuntos
Bifidobacterium animalis , Probióticos , Feminino , Humanos , Adulto Jovem , Bifidobacterium/fisiologia , Cognição , Método Duplo-Cego , Lactobacillus acidophilus/fisiologia , Probióticos/farmacologia , Probióticos/uso terapêutico
8.
Int J Mol Sci ; 23(18)2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36142723

RESUMO

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.


Assuntos
Infecções por Escherichia coli , Probióticos , Animais , Bifidobacterium/fisiologia , Bifidobacterium longum subspecies infantis/metabolismo , Células CACO-2 , Citocinas/metabolismo , Escherichia coli/metabolismo , Humanos , Lactente , Camundongos , Probióticos/farmacologia , Suínos
9.
J Dairy Sci ; 105(9): 7308-7321, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35931487

RESUMO

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.


Assuntos
Bifidobacterium animalis , Probióticos , Bifidobacterium/fisiologia , Liofilização/veterinária , Probióticos/metabolismo , Hidrolisados de Proteína/farmacologia , Soro do Leite
10.
Front Cell Infect Microbiol ; 12: 914684, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35846768

RESUMO

Objective: To investigate the effect of Bifidobacterium animalis B94 on the prevention and treatment of liver injury in rats and to elucidate the underlying mechanism of this relationship. Methods: Specific pathogen-free (SPF) rats were selected as the healthy control group, liver injury group and B94 treatment group, with 6 rats in each group. After the model was established, the experimental animals were tested for serum liver function indicators, gut microbiota composition, metabolite composition, and histopathology. Results: The albumin/globulin ratio and serum TBA, alanine aminotransferase, aspartate aminotransferase, and indirect bilirubin levels in the B94 treatment group were significantly lower than those in the liver injury group. 16S rRNA analysis showed that the gut microbiota of the three groups of rats were significantly different. Metabolic profile analysis showed that there were significant differences in the gut metabolomes of the three groups. Haematoxylin-eosin staining of the intestinal mucosa and liver tissues showed that the degree of liver and intestinal tissue damage in the B94 treatment group was significantly lower than that in the liver injury group. Conclusion: Bifidobacterium animalis B94 can affect the process of liver injury in rats by improving liver function, reducing intestinal damage, and regulating gut microbiota and metabolite production.


Assuntos
Bifidobacterium animalis , Probióticos , Animais , Bifidobacterium/fisiologia , Bifidobacterium animalis/genética , Fígado , RNA Ribossômico 16S/genética , Ratos
11.
Food Funct ; 13(14): 7520-7528, 2022 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-35763064

RESUMO

Early life stress can considerably interfere in gut microbiome formation and nervous system development. Specific probiotic strains have been proved to exert anti-stress effects by modulating the gut-brain axis. However, little is known about whether probiotic treatment during pregnancy can protect the offspring from early life stress. In this study, Bifidobacterium breve CCFM1025, previously proven to exert microbial and neurobiological regulation effects, was given to pregnant mice. The offspring's gut and brain functions were evaluated when challenged with maternal separation. Intriguingly, treatment with probiotics during pregnancy protected the offspring from maternal separation-induced neurobiological and gastrointestinal disorders such as depression-like behaviour and delayed defecation. Quantification of CCFM1025 was performed, and perinatal transmission of CCFM1025 was further validated, which also explained the reason for increased levels of colonic 5-hydroxytryptamine and caecal short-chain fatty acids in the offspring. Our findings indicated that the effects of probiotics can be perinatally transmitted through gut microbes and that probiotic treatment during pregnancy may have great potential in managing health risks in early life.


Assuntos
Gastroenteropatias , Probióticos , Estresse Psicológico , Animais , Feminino , Camundongos , Gravidez , Bifidobacterium/fisiologia , Motilidade Gastrointestinal , Transmissão Vertical de Doenças Infecciosas , Privação Materna , Probióticos/farmacologia
12.
Microbiol Spectr ; 10(4): e0065722, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35730951

RESUMO

Inflammatory bowel disease (IBD) has become a global public health problem. Although the pathogenesis of the disease is unknown, a potential association between the gut microbiota and inflammatory signatures has been established. Probiotics, especially Lactobacillus or Bifidobacterium, are orally taken as food supplements or microbial drugs by patients with IBD or gastrointestinal disorders due to their safety, efficacy, and power to restore the gut microenvironment. In the current study, we investigated the comprehensive effects of probiotic bacterial consortia consisting of Lactobacillus reuteri, Lactobacillus gasseri, Lactobacillus acidophilus (Lactobacillus spp.), and Bifidobacterium lactis (Bifidobacterium spp.) or their metabolites in a dextran sodium sulfate (DSS)-induced colitis mouse model. Our data demonstrate that probiotic consortia not only ameliorate the disease phenotype but also restore the composition and structure of the gut microbiota. Moreover, the effect of probiotic consortia is better than that of any single probiotic strain. The results also demonstrate that mixed fermentation metabolites are capable of ameliorating the symptoms of gut inflammation. However, the administration of metabolites is not as effective as probiotic consortia with respect to phenotypic characteristics, such as body weight, disease activity index (DAI), and histological score. In addition, mixed metabolites led only to changes in intestinal flora composition. In summary, probiotic consortia and metabolites could exert protective roles in the DSS-induced colitis mouse model by reducing inflammation and regulating microbial dysbiosis. These findings from the current study provide support for the development of probiotic-based microbial products as an alternative therapeutic strategy for IBD. IMPORTANCE IBD is a chronic nonspecific inflammatory disease. IBD is characterized by a wide range of lesions, often involving the entire colon, and is characterized mainly by ulcers and erosions of the colonic mucosa. In the present study, we investigated the efficacy of probiotics on the recovery of gut inflammation and the restoration of gut microecology. We demonstrate that probiotic consortia have a superior effect in inhibiting inflammation and accelerating recovery compared with the effects observed in the control group or groups administered with a single strain. These results support the utilization of probiotic consortia as an alternative therapeutic approach to treat IBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Probióticos , Animais , Bifidobacterium/fisiologia , Colite/tratamento farmacológico , Colite/terapia , Colo/microbiologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Inflamação/patologia , Doenças Inflamatórias Intestinais/terapia , Lactobacillus/fisiologia , Camundongos , Probióticos/farmacologia , Probióticos/uso terapêutico
13.
BMC Gastroenterol ; 22(1): 100, 2022 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-35247974

RESUMO

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 ).


Assuntos
Microbioma Gastrointestinal , Probióticos , Bifidobacterium/fisiologia , Citocinas , Humanos , Mucosa Intestinal , Leucócitos , Probióticos/uso terapêutico
14.
PLoS One ; 17(2): e0262942, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35171916

RESUMO

This study was designed to explore the therapeutics and the mechanisms of a patented and marked gastric acid and intestine juice-resistant probiotics Bifidobacterium lactis BL-99 (B. lactis BL-99) on the intestinal inflammation and functions in the zebrafish models. After feeding for 6 hours, B. lactis BL-99 was fully retained in the larval zebrafish intestinal tract and stayed for over 24 hours. B. lactis BL-99 promoted the intestinal motility and effectively alleviated aluminum sulfate-induced larval zebrafish constipation (p < 0.01). Irregular high glucose diet induced adult zebrafish intestinal functional and metabolic disorders. After fed with B. lactis BL-99, IL-1ß gene expression was significantly down-regulated, and IL-10 and IL-12 gene levels were markedly up-regulated in this model (p < 0.05). The intestinal lipase activity was elevated in the adult zebrafish intestinal functional disorder model after B. lactis BL-99 treatment (p < 0.05), but tryptase content had no statistical changes (p > 0.05). B. lactis BL-99 improved the histopathology of the adult zebrafish intestinal inflammation, increased the goblet cell numbers, and up-and-down metabolites were markedly recovered after treatment of B. lactis BL-99 (p < 0.05). These results suggest that B. lactis BL-99 could relieve intestinal inflammation and promote intestinal functions, at least in part, through modulating intestinal and microbial metabolism to maintain intestinal health.


Assuntos
Bifidobacterium/fisiologia , Inflamação/terapia , Intestinos/metabolismo , Probióticos/uso terapêutico , Compostos de Alúmen/toxicidade , Animais , Constipação Intestinal/induzido quimicamente , Constipação Intestinal/patologia , Constipação Intestinal/terapia , Análise Discriminante , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Glucose/farmacologia , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Intestinos/microbiologia , Intestinos/patologia , Larva/efeitos dos fármacos , Larva/metabolismo , Probióticos/farmacologia , Regulação para Cima/efeitos dos fármacos , Peixe-Zebra/crescimento & desenvolvimento
15.
Nat Genet ; 54(2): 134-142, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35115689

RESUMO

Human genetic variation affects the gut microbiota through a complex combination of environmental and host factors. Here we characterize genetic variations associated with microbial abundances in a single large-scale population-based cohort of 5,959 genotyped individuals with matched gut microbial metagenomes, and dietary and health records (prevalent and follow-up). We identified 567 independent SNP-taxon associations. Variants at the LCT locus associated with Bifidobacterium and other taxa, but they differed according to dairy intake. Furthermore, levels of Faecalicatena lactaris associated with ABO, and suggested preferential utilization of secreted blood antigens as energy source in the gut. Enterococcus faecalis levels associated with variants in the MED13L locus, which has been linked to colorectal cancer. Mendelian randomization analysis indicated a potential causal effect of Morganella on major depressive disorder, consistent with observational incident disease analysis. Overall, we identify and characterize the intricate nature of host-microbiota interactions and their association with disease.


Assuntos
Dieta , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Variação Genética , Interações entre Hospedeiro e Microrganismos , Polimorfismo de Nucleotídeo Único , Sistema ABO de Grupos Sanguíneos/genética , Bifidobacterium/fisiologia , Clostridiales/fisiologia , Estudos de Coortes , Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia , Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/microbiologia , Fibras na Dieta , Enterococcus faecalis/fisiologia , Microbioma Gastrointestinal/genética , Estudo de Associação Genômica Ampla , Humanos , Lactase/genética , Complexo Mediador/genética , Análise da Randomização Mendeliana , Metagenoma , Morganella/fisiologia
16.
Nat Genet ; 54(2): 143-151, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35115690

RESUMO

Host genetics are known to influence the gut microbiome, yet their role remains poorly understood. To robustly characterize these effects, we performed a genome-wide association study of 207 taxa and 205 pathways representing microbial composition and function in 7,738 participants of the Dutch Microbiome Project. Two robust, study-wide significant (P < 1.89 × 10-10) signals near the LCT and ABO genes were found to be associated with multiple microbial taxa and pathways and were replicated in two independent cohorts. The LCT locus associations seemed modulated by lactose intake, whereas those at ABO could be explained by participant secretor status determined by their FUT2 genotype. Twenty-two other loci showed suggestive evidence (P < 5 × 10-8) of association with microbial taxa and pathways. At a more lenient threshold, the number of loci we identified strongly correlated with trait heritability, suggesting that much larger sample sizes are needed to elucidate the remaining effects of host genetics on the gut microbiome.


Assuntos
Sistema ABO de Grupos Sanguíneos/genética , Fenômenos Fisiológicos Bacterianos , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Variação Genética , Interações entre Hospedeiro e Microrganismos , Lactase/genética , Bifidobacterium/fisiologia , Dieta , Fucosiltransferases/genética , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Redes e Vias Metabólicas , Metagenoma , Herança Multifatorial , Países Baixos , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Cloreto de Sódio na Dieta , Triglicerídeos/sangue , Galactosídeo 2-alfa-L-Fucosiltransferase
17.
Oral Dis ; 28(6): 1723-1732, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33749084

RESUMO

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.


Assuntos
Placa Dentária , Probióticos , Bifidobacterium/fisiologia , Placa Dentária/microbiologia , Humanos , Imunidade , Lactobacillus/fisiologia , Probióticos/uso terapêutico
18.
Bull Exp Biol Med ; 172(2): 164-168, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34855091

RESUMO

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.


Assuntos
Antibiose/fisiologia , Parede Celular/fisiologia , Peptidoglicano/metabolismo , Bacillus subtilis/fisiologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Bifidobacterium/fisiologia , Candida/fisiologia , Parede Celular/química , Parede Celular/metabolismo , Enterobacter/fisiologia , Enterococcus faecalis/fisiologia , Escherichia coli/fisiologia , Feminino , Humanos , Lacticaseibacillus casei/fisiologia , Técnicas Microbiológicas , Peptidoglicano/análise , Staphylococcus aureus/fisiologia
19.
Pol J Microbiol ; 70(4): 511-520, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34970318

RESUMO

This study aimed to investigate in vitro effects of the selected prebiotics alone, and in combination with two potential probiotic Lactobacillus strains on the microbial composition of Apis cerana gut microbiota and acid production. Four prebiotics, inulin, fructo-oligosaccharides, xylo-oligosaccharides, and isomalto-oligosaccharides were chosen, and glucose served as the carbon source. Supplementation of this four prebiotics increased numbers of Bifidobacterium and lactic acid bacteria while decreasing the pH value of in vitro fermentation broth inoculated with A. cerana gut microbiota compared to glucose. Then, two potential probiotics derived from A. cerana gut at different dosages, Lactobacillus helveticus KM7 and Limosilactobacillus reuteri LP4 were added with isomalto-oligosaccharides in fermentation broth inoculated with A. cerana gut microbiota, respectively. The most pronounced impact was observed with isomalto-oligosaccharides. Compared to isomalto-oligosaccharides alone, the combination of isomalto-oligosaccharides with both lactobacilli strains induced the growth of Bifidobacterium, LAB, and total bacteria and reduced the proliferation of Enterococcus and fungi. Consistent with these results, the altered metabolic activity was observed as lowered pH in in vitro culture of gut microbiota supplemented with isomalto-oligosaccharides and lactobacilli strains. The symbiotic impact varied with the types and concentration of Lactobacillus strains and fermentation time. The more effective ability was observed with IMO combined with L. helveticus KM7. These results suggested that isomalto-oligosaccharides could be a potential prebiotic and symbiotic with certain lactobacilli strains on A. cerana gut microbiota.


Assuntos
Abelhas , Microbioma Gastrointestinal , Prebióticos , Probióticos , Simbióticos , Animais , Abelhas/microbiologia , Bifidobacterium/fisiologia , Fermentação , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Glucose/farmacologia , Lactobacillus/fisiologia , Oligossacarídeos/farmacologia , Probióticos/farmacologia , Simbióticos/análise
20.
Gut Microbes ; 13(1): 2006586, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34856861

RESUMO

Research on the gut-brain axis has accelerated substantially over the course of the last years. Many reviews have outlined the important implications of understanding the relation of the gut microbiota with human brain function and behavior. One substantial drawback in integrating gut microbiome and brain data is the lack of integrative multivariate approaches that enable capturing variance in both modalities simultaneously. To address this issue, we applied a linked independent component analysis (LICA) to microbiota and brain connectivity data.We analyzed data from 58 healthy females (mean age = â€¯21.5 years). Magnetic Resonance Imaging data were acquired using resting state functional imaging data. The assessment of gut microbial composition from feces was based on sequencing of the V4 16S rRNA gene region. We used the LICA model to simultaneously factorize the subjects' large-scale brain networks and microbiome relative abundance data into 10 independent components of spatial and abundance variation.LICA decomposition resulted in four components with non-marginal contribution of the microbiota data. The default mode network featured strongly in three components, whereas the two-lateralized fronto-parietal attention networks contributed to one component. The executive-control (with the default mode) network was associated to another component. We found that the abundance of Prevotella genus was associated with the strength of expression of all networks, whereas Bifidobacterium was associated with the default mode and frontoparietal-attention networks.We provide the first exploratory evidence for multivariate associative patterns between the gut microbiota and brain network connectivity in healthy humans considering the complexity of both systems.


Assuntos
Encéfalo/fisiologia , Microbioma Gastrointestinal/fisiologia , Rede Nervosa/fisiologia , Bifidobacterium/isolamento & purificação , Bifidobacterium/fisiologia , Encéfalo/diagnóstico por imagem , Eixo Encéfalo-Intestino/fisiologia , Feminino , Microbioma Gastrointestinal/genética , Voluntários Saudáveis , Humanos , Imageamento por Ressonância Magnética , Rede Nervosa/diagnóstico por imagem , Prevotella/isolamento & purificação , Prevotella/fisiologia , Descanso/fisiologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA