Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Adv Nutr ; 15(1): 100128, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37827490

RESUMEN

Recent evidence suggests that the timing of introduction, types, and amounts of complementary foods/allergenic foods may influence the risk of allergic disease. However, the evidence has not been updated and comprehensively synthesized. The Cochrane Library, EMBASE, Web of Science, and PubMed databases were searched from the inception of each database up to 31 May 2023 (articles prior to 2000 were excluded manually). Statistical analyses were performed using RevMan 5. The GRADE approach was followed to rate the certainty of evidence. Compared with >6 mo, early introduction of eggs (≤6 mo of age) might reduce the risk of food allergies in preschoolers aged <6 y (odds ratio [OR], 0.65; 95% confidence interval [CI], 0.53, 0.81), but had no effect on asthma or atopic dermatitis (AD). Consumption of fish at 6-12 mo might reduce the risk of asthma in children (aged 5-17 y) compared with late introduction after 12 mo (OR, 0.61; 95% CI: 0.52, 0.72). Introduction of allergenic foods for ≤6 mo of age, compared with >6 mos, was a protective factor for the future risk (children aged ≤10 y) of AD (OR, 0.93; 95% CI: 0.89, 0.97). Probiotic intervention for infants at high risk of allergic disease significantly reduced the risk of food allergy at ages 0-3 y (OR, 0.72; 95% CI: 0.56, 0.94), asthma at 6-12 y (OR, 0.61; 95% CI: 0.41, 0.90), and AD at aged <6 y (3-6 y: OR, 0.70; 95% CI: 0.52, 0.94; 0-3 y: OR, 0.73; 95% CI: 0.59, 0.91). Early introduction of complementary foods or the high-dose vitamin D supplementation in infancy was not associated with the risk of developing food allergies, asthma, or AD during childhood. Early introduction to potential allergen foods for normal infants or probiotics for infants at high risk of allergies may protect against development of allergic disease. This study was registered at PROSPERO as CRD42022379264.


Asunto(s)
Asma , Dermatitis Atópica , Hipersensibilidad a los Alimentos , Lactante , Niño , Animales , Humanos , Prevalencia , Dieta , Hipersensibilidad a los Alimentos/epidemiología , Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/prevención & control , Dermatitis Atópica/epidemiología , Dermatitis Atópica/etiología , Dermatitis Atópica/prevención & control , Asma/epidemiología , Asma/etiología , Asma/prevención & control , Huevos
2.
Food Funct ; 14(15): 6896-6913, 2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37427574

RESUMEN

Clinical antibiotics used worldwide could diminish the intestinal barrier, enhance contact with microbiota and intestinal immune cells, and induce inflammation. We found that ciprofloxacin treatment of Salmonella enterica serovar Typhimurium infection resulted in the destruction of the intestinal barrier, with decreased concentrations of MUC2, ZO-1, and occludin in the jejunum and colon. Ganoderma lucidum ethanol extracts (GLE), as a prebiotic food extract, significantly decreased inflammation-related enzymes, including COX-2, MPO, and iNOS, and pro-inflammatory cytokines (IL-6, IL-1ß, IL-17, and TNF-α), and protected the intestinal barrier by increasing the concentration of MUC2, ZO-1, and occludin. Meanwhile it significantly increased the abundances of Salmonella, Parabacteroides, Acinetobacter, Enterococcus, and Escherichia-Shigella, which increased the risk of pathogenic bacterial infections. Prebiotic G. lucidum polysaccharide (GLP) provided a significant intestinal barrier, improving the concentration of ZO-1, occludin, and MUC2 in the colon and jejunum. The synergistic effects of GLP and ciprofloxacin were hypothesized to reverse the negative effects resulting from ciprofloxacin alone, as the concentrations of ZO-1, occludin, and MUC2 were significantly increased in the jejunum and colon, especially in the colon. Also, the synergistic effect increased the abundances of probiotic bacteria Lachnospiraceae NK4A136, Ruminococcaceae UGG-014, Lactobacillus, and Parabacteroides. In conclusion, combined GLP and ciprofloxacin therapy against Salmonella infection alleviated the side effects resulting from the clinical application of the antibiotic alone, and increased the probiotic bacterial population.


Asunto(s)
Microbioma Gastrointestinal , Reishi , Infecciones por Salmonella , Humanos , Ciprofloxacina/farmacología , Ocludina/genética , Salmonella typhimurium/fisiología , Inflamación/tratamiento farmacológico , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Prebióticos , Bacterias/genética
3.
Pharmacol Res ; 194: 106867, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37499703

RESUMEN

Most diets and medications enhance host health via microbiota-dependent ways, but it is in the present situation of untargeted regulation. Non-targeted regulation may lead to the ineffectiveness of dietary supplements or drug treatment. Microbiota-directed food, aiming to improve diseases by targeting specific microbes without affecting other bacteria, have been proposed to deal with this problem. However, there is currently no universally applicable method to explore such foods or drugs. In this review, thirty studies on recent efforts in microbiota directed diets and medications are summarized from various databases. The methods used to find new foods and medications are primarily divided into four groups depending on the experimental models: in vivo and in vitro, as well as predictions based on bioinformatics. We also discuss their implementation, interpretation, and respective limitations, and describe the present situation. We further put forward a framework for microbiota-directed foods and medicine according to above methods and other microbiome manipulation, which will spur precision medicine.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Dieta , Suplementos Dietéticos , Diseño de Fármacos
4.
Biomedicines ; 11(4)2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-37189666

RESUMEN

Taurine, an abundant free amino acid, plays multiple roles in the body, including bile acid conjugation, osmoregulation, oxidative stress, and inflammation prevention. Although the relationship between taurine and the gut has been briefly described, the effects of taurine on the reconstitution of intestinal flora homeostasis under conditions of gut dysbiosis and underlying mechanisms remain unclear. This study examined the effects of taurine on the intestinal flora and homeostasis of healthy mice and mice with dysbiosis caused by antibiotic treatment and pathogenic bacterial infections. The results showed that taurine supplementation could significantly regulate intestinal microflora, alter fecal bile acid composition, reverse the decrease in Lactobacillus abundance, boost intestinal immunity in response to antibiotic exposure, resist colonization by Citrobacter rodentium, and enhance the diversity of flora during infection. Our results indicate that taurine has the potential to shape the gut microbiota of mice and positively affect the restoration of intestinal homeostasis. Thus, taurine can be utilized as a targeted regulator to re-establish a normal microenvironment and to treat or prevent gut dysbiosis.

5.
Nutrients ; 15(7)2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37049578

RESUMEN

Recent studies have demonstrated that disturbances in the gut microbiota and microbiota -derived metabolites contribute to the pathogenesis of Parkinson's disease (PD), suggesting that probiotic treatments that restore them may delay disease progression. This study aimed to examine the attenuating efficacy of L. plantarum CCFM405 and the potential mechanisms in mice with rotenone-induced PD. Our results indicate that L. plantarum CCFM405 ameliorated rotenone-induced motor deficits and constipation, decreased dopaminergic neuronal death, reduced intestinal inflammation and neuroinflammation, and raised dopamine levels, 5-HT, and associated metabolites in the striatal region of the brain in mice with PD. Sequencing of 16S rRNA from fecal microbiota revealed that L. plantarum CCFM405 normalized the gut bacterial composition in mice with PD, as evidenced by the increased relative abundance of the following genus, Bifidobacterium, Turicibacter, and Faecalibaculum, and decreased relative abundance of Alistipes, Bilophila, Akkermansia, and Escherichia-Shigella. The PICRUSt-predicted gut microbiota function revealed that L. plantarum CCFM405 enhanced the biosynthesis of amino acid pathways, particularly valine, leucine, and isoleucine (branched-chain amino acids, BCAAs). A non-metabolomic analysis of the serum and feces showed that L. plantarum CCFM405 markedly increased the levels of BCAAs. Pathway enrichment analysis based on the KEGG database further suggested that L. plantarum CCFM405 supplementation can promote BCAAs biosynthesis. Collectively, L. plantarum CCFM405 can help to prevent rotenone-induced PD by modulating the gut microbiota-metabolite axis. BCAAs may play a dominant role in L. plantarum CCFM405-associated neuroprotection in PD mice. This probiotic could be utilized as a potential food supplement in the management of PD.


Asunto(s)
Microbioma Gastrointestinal , Lactobacillus plantarum , Enfermedad de Parkinson , Animales , Ratones , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/prevención & control , Microbioma Gastrointestinal/fisiología , Lactobacillus plantarum/fisiología , Rotenona/toxicidad , Aminoácidos de Cadena Ramificada , ARN Ribosómico 16S/genética , Dopamina
6.
Crit Rev Food Sci Nutr ; : 1-17, 2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36168909

RESUMEN

The application of probiotics is limited by the loss of survival due to food processing, storage, and gastrointestinal tract. Encapsulation is a key technology for overcoming these challenges. The review focuses on the latest progress in probiotic encapsulation since 2020, especially precision engineering on microbial surfaces and microbial-mediated role. Currently, the encapsulation materials include polysaccharides and proteins, followed by lipids, which is a traditional mainstream trend, while novel plant extracts and polyphenols are on the rise. Other natural materials and processing by-products are also involved. The encapsulation types are divided into rough multicellular encapsulation, precise single-cell encapsulation, and microbial-mediated encapsulation. Recent emerging techniques include cryomilling, 3D printing, spray-drying with a three-fluid coaxial nozzle, and microfluidic. Encapsulated probiotics applied in food is an upward trend in which "classic probiotic foods" (yogurt, cheese, butter, chocolate, etc.) are dominated, supplemented by "novel probiotic foods" (tea, peanut butter, and various dry-based foods). Future efforts mainly include the effect of novel encapsulation materials on probiotics in the gut, encapsulation strategy oriented by microbial enthusiasm and precise encapsulation, development of novel techniques that consider both cost and efficiency, and co-encapsulation of multiple strains. In conclusion, encapsulation provides a strong impetus for the food application of probiotics.

7.
Nutrients ; 14(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35057570

RESUMEN

During pregnancy and lactation, considerable factors that affect the maternal microbiome are associated with the advancement of numerous diseases, which can potentially affect offspring health. Probiotics have shown potential for the maintenance of microbiota homeostasis of mothers in this period. The specific objective of this study was to investigate whether the application of Akkermansia muciniphila (A. muciniphila) during pregnancy and lactation impacts maternal and offspring health. Here we show that dams fed with A. muciniphila is safe, enhances the intestinal barrier and alters gut microbiota composition and diversity at the end of lactation, including the significant enrichment of A. muciniphila and Ruminococcus_1 in offspring from probiotic-fed dams. However, compared with the control group, the fecal metabolites of the A. muciniphila group only changed slightly. Additionally, A. muciniphila supplementation did not significantly increase the abundance of A. muciniphila in the fecal microbiota of offspring mice. Compared with the control group, the fecal metabolic profile of three-week-old offspring of mice fed with A. muciniphila were significantly changed, containing the D-glutamine and D-glutamate metabolism pathways. These results provided evidence that A. muciniphila supplementation in mice during pregnancy and lactation is safe and seemed to have a more beneficial effect on dams. In the future, using probiotics to regulate maternal microbiomes during pregnancy and lactation could be shown to have a more lasting and beneficial effect.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Lactancia/fisiología , Probióticos/administración & dosificación , Akkermansia , Animales , Animales Recién Nacidos/microbiología , Animales Recién Nacidos/fisiología , Suplementos Dietéticos , Heces/química , Femenino , Promoción de la Salud , Estado de Salud , Ratones , Ratones Endogámicos C57BL , Embarazo
8.
Crit Rev Food Sci Nutr ; 62(6): 1427-1452, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33198506

RESUMEN

The oral antibiotic therapies administered widely to people and animals can cause gut dysbiosis and barrier disruption inevitably. Increasing attention has been directed toward antibiotic-induced gut dysbiosis, which involves a loss of diversity, changes in the abundances of certain taxa and consequent effects on their metabolic capacity, and the spread of antibiotic-resistant bacterial strains. Treatment with beta-lactam, glycopeptide, and macrolide antibiotics is associated with the depletion of beneficial commensal bacteria in the genera Bifidobacterium and Lactobacillus. The gut microbiota is a reservoir for antibiotic resistance genes, the prevalence of which increases sharply after antibiotic ingestion. The intestinal barrier, which comprises secretory, physical, and immunological barriers, is also a target of antibiotics. Antibiotic induced changes in the gut microbiota composition could induce weakening of the gut barrier through changes in mucin, cytokine, and antimicrobial peptide production by intestinal epithelial cells. Reports have indicated that dietary interventions involving prebiotics, probiotics, omega-3 fatty acids, and butyrate supplementation, as well as fecal microbiota transplantation, can alleviate antibiotic-induced gut dysbiosis and barrier injuries. This review summarizes the characteristics of antibiotic-associated gut dysbiosis and barrier disruption, as well as the strategies for alleviating this condition. This information is intended to provide a foundation for the exploration of safer, more efficient, and affordable strategies to prevent or relieve antibiotic-induced gut injuries.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Animales , Antibacterianos/toxicidad , Disbiosis/inducido químicamente , Disbiosis/prevención & control , Humanos , Lactobacillus
9.
Food Res Int ; 144: 110331, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34053534

RESUMEN

The link between the gut microbiome and bone health has begun to attract widespread interest in recent years. The gut microbiome are vital in many diseases involving bone loss. Probiotics, prebiotics, and dietary supplements have been suggested to protect bone health by altering the composition of the gut microbiota. Notably, studying the relationship between the gut microbiome and bone health can provide a basis for the prevention and treatment of bone diseases. This review focuses on the link between the gut microbiome and bone diseases, exploring current knowledge of the mechanisms by which gut bacteria affect bone health. In addition, the influences of dietary supplements on the interactions between the gut microbiome and bone health are discussed. This knowledge will promote new ideas for gut microbiota-mediated dietary interventions in patients with bone diseases.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Densidad Ósea , Dieta , Humanos , Prebióticos
10.
Environ Microbiol ; 23(6): 3164-3181, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33876566

RESUMEN

Sulfate-reducing bacteria (SRB) are widespread in human guts, yet their expansion has been linked to colonic diseases. We report the isolation, sequencing and physiological characterization of strain QI0027T , a novel SRB species belonging to the class Desulfovibrionia. Metagenomic sequencing of stool samples from 45 Chinese individuals, and comparison with 1690 Desulfovibrionaceae metagenome-assembled genomes recovered from humans of diverse geographic locations, revealed the presence of QI0027T in 22 further individuals. QI0027T encoded nitrogen fixation genes and based on the acetylene reduction assay, actively fixed nitrogen. Transcriptomics revealed that QI0027T overexpressed 42 genes in nitrogen-limiting conditions compared to cultures supplemented with ammonia, including genes encoding nitrogenases, a urea uptake system and the urease complex. Reanalyses of 835 public stool metatranscriptomes showed that nitrogenase genes from Desulfovibrio bacteria were expressed in six samples suggesting that nitrogen fixation might be active in the gut environment. Although frequently thought of as a nutrient-rich environment, nitrogen fixation can occur in the human gut. Animals are often nitrogen limited and have evolved diverse strategies to capture biologically active nitrogen, ranging from amino acid transporters to stable associations with beneficial microbes that provide fixed nitrogen. QI0027T is the first Desulfovibrio human isolate for which nitrogen fixation has been demonstrated, suggesting that some sulfate-reducing bacteria could also play a role in the availability of nitrogen in the gut.


Asunto(s)
Desulfovibrio , Fijación del Nitrógeno , Animales , Bacterias/metabolismo , Desulfovibrio/genética , Desulfovibrio/metabolismo , Humanos , Nitrogenasa/metabolismo , Oxidación-Reducción , Filogenia , Sulfatos
11.
J Med Food ; 24(3): 319-330, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33739885

RESUMEN

Diabetes mellitus is a global health problem, and its prevalence continues to increase. Dietary supplements, including probiotics, prebiotics, and plant extracts, have been shown to alleviate diabetes. In this study, the synergistic effects of two types of dietary supplements were investigated in a mouse model of type 2 diabetes mellitus (T2DM). Sixty mice were divided into the following six groups: control, model (induced by a high-fat diet and intraperitoneal injection of streptozotocin), drug (metformin), probiotic (Lactobacillus spp.), formula A (probiotics, plant extracts, and soybean peptide), and formula B (probiotics, prebiotics, and soybean peptide). All three dietary interventions (probiotic, formula A, and formula B groups) significantly reduced the blood glucose level and oral glucose tolerance level and effectively improved some biochemical parameters (e.g., chronic inflammation, oxidative stress, and blood lipid level) and regulated gut microbiota. Notably, formula B exhibited a better ability on reducing the blood glucose level, regulating the gut microbiota, and increasing the short-chain fatty acid levels compared with the probiotics alone and formula A. Thus, formula B may exert synergistic protective effects against T2DM through a mechanism involving probiotics and prebiotics of gut microbiota regulation. This study provides a theoretical basis for the application of probiotic dietary supplements to the treatment of T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Probióticos , Animales , Suplementos Dietéticos , Ratones , Prebióticos
12.
Food Funct ; 12(3): 1121-1134, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33427835

RESUMEN

Irritable bowel syndrome (IBS) is a chronic intestinal disorder accompanied by low-grade inflammation, visceral hypersensitivity, and gut microbiota dysbiosis. Several studies have indicated that Lactobacillus supplementation can help to alleviate IBS symptoms and that these effects are strain-specific. Therefore, this study aimed to investigate the key physiological characteristics and functional genes contributing to the IBS-alleviating effects of Lactobacillus. An IBS model was established by subjecting C57BL/6 mice to Citrobacter rodentium ingestion and water avoidance stress. Lactobacillus strains with different physiological characteristics were administered to mice intragastrically for 4 weeks (5 × 109 CFU/0.2 mL per mouse per day). Indicators of colonic inflammation, visceral hypersensitivity, and gut microbiota were also evaluated. Finally, differences in functional genes between Lactobacillus strains were analyzed by a comparative genomic analysis, and the relationships between the physiological characteristics, functional genes, and IBS-alleviating effects of the strains were quantified using correlation analysis. Among the eight tested Lactobacillus strains, only Lactobacillus plantarum CCFM8610 significantly inhibited the expression of IL-1ß, IL-6, PAR-2, and mast cell tryptase. L. plantarum CCFM8610 also significantly increased the intestinal barrier function, inhibited visceral hypersensitivity symptoms, and modulated the gut microbiota diversity and composition. The correlation analysis of factors associated with the IBS-alleviating effects of Lactobacillus revealed the ability to synthesize conjugated linoleic acid as the most strongly associated physiological characteristic and COG1028-related genes as the most strongly associated functional genes. In conclusion, these findings can facilitate the rapid screening of Lactobacillus strains with IBS-alleviating effects and lay a foundation for studies of the related mechanisms.


Asunto(s)
Síndrome del Colon Irritable/microbiología , Lactobacillus/genética , Probióticos/farmacología , Animales , Citrobacter rodentium , Colon/microbiología , Colon/patología , Corticosterona/sangre , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Humanos , Inflamación/patología , Inflamación/prevención & control , Lactobacillus/fisiología , Ácidos Linoleicos Conjugados , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Triptasas/metabolismo
13.
Nutrients ; 12(9)2020 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-32825154

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is a disorder characterized by the excess accumulation of fat in the hepatocytes. It is commonly associated with severe obesity and inflammation. Free fatty acids (FFAs) are the key to regulate lipid metabolism and immune response in hepatocyte cells. This study examined the effects of AEN (alcohol extract of nutmeg, the seed of Myristica fragrans Houtt.) on the inhibition of lipid synthesis and inflammation in vitro and in vivo and on high-fat diet-induced obesity in NAFLD mice. Our results showed that AEN treatment could downregulate the expression of lipid synthesis-related genes fatty acid synthase (FASN) and sterol regulatory element-binding protein 1c (SREBP-1c) and lower the lipid content of cells. AEN also inhibited FFAs-mediated inflammation-related cytokines interleukin-6 (IL-6) and tumor necrosis factor α (TNFα) expression in cells. In a mouse model, AEN reduced the bodyweight of obese mice and improved NAFLD without affecting food intake. Further analysis revealed that AEN significantly reduced inflammation level, cholesterol and lipid accumulation, blood glucose, and other liver function indexes in mice fed with a high-fat diet. In conclusion, AEN inhibited the aggravation of obesity and inflammation by downregulating lipid-gene expression in the liver to ameliorate NAFLD.


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Inflamación/tratamiento farmacológico , Myristica/química , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Animales , Línea Celular , Modelos Animales de Enfermedad , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Inflamación/prevención & control , Interleucina-6/metabolismo , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/prevención & control , Células RAW 264.7 , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
Microb Pathog ; 147: 104403, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32707316

RESUMEN

Traditional therapies for Helicobacter pylori (H. pylori) infection remain hindered by the antibiotic resistance of the pathogen and the poor therapeutic compliance of patients. To address these issues, probiotics have been added as an adjunctive therapy. This meta-analysis aimed to evaluate the efficacy of probiotic supplementation during standard therapy on the eradication rate of H. pylori infection and incidence of therapy-related side effects. Four online databases were searched for eligible studies without language restriction. Review Manager (REVMAN, Version 5.3) was used to perform all data analyses. Forty articles including 5792 patients met our criteria and were included in the analysis. Notably, probiotic supplementation improved the eradication rate by approximately 10% relative to the control group [odds ratio (OR), 1.94, 95% confidence interval (CI): 1.70-2.22, P < 0.00001]. The incidence of total side effects (OR, 0.56, 95% CI: 0.45-0.70, P < 0.00001) and individual symptoms (e.g., diarrhea, vomiting and nausea, constipation, epigastric pain, taste disturbance) also decreased significantly with probiotic supplementation. No other differences in side effects were observed between the experimental and control groups. Moreover, a longer duration (≥10 days) of probiotic treatment had positive effects on both eradication rate of H. pylori and incidence of overall side effects.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Probióticos , Antibacterianos/efectos adversos , Suplementos Dietéticos , Quimioterapia Combinada , Infecciones por Helicobacter/tratamiento farmacológico , Humanos , Incidencia , Resultado del Tratamiento
15.
Microbiol Res ; 233: 126409, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31927503

RESUMEN

Insomnia is a common health problem in modern societies. GABA, an inhibitory neurotransmitter, can promote relaxation and reduce anxiety. In this study, milk was fermented with Lactobacillus brevis DL1-11, a strain with high GABA-producing capacity. The potential beneficial effects of this fermented milk on anxiety and sleep quality were evaluated in animal experiments. Sixty mice were divided into control, non-GABA fermented milk (NGFM), low-dose GABA fermented milk (LGFM, 8.83 mg/kg.bw), medium-dose GABA fermented milk (MGFM, 16.67 mg/kg.bw), high-dose GABA fermented milk (HGFM, 33.33 mg/kg.bw) and diazepam groups. The results of open field test and elevated plus-maze test indicated decreases in anxiety behavior after oral HGFM administration. Moreover, mice in the HGFM group exhibited a significantly prolonged sleep time after an intraperitoneal injection of sodium pentobarbital and a shortened sleep latency after an intraperitoneal injection of sodium barbital. These results indicate a beneficial effect of HGFM on sleep. Additionally, significant increases in the relative abundances of Ruminococcus, Adlercreutzia and Allobaculum and the levels of some short-chain fatty acids (SCFAs), such as butyric acid, were observed in the HGFM group. The results suggest that GABA-fermented milk may improve sleep and the protective pathways may involve in regulation of gut microbiota and increase of SCFAs level.


Asunto(s)
Productos Lácteos Cultivados , Microbioma Gastrointestinal , Levilactobacillus brevis , Trastornos del Inicio y del Mantenimiento del Sueño/terapia , Ácido gamma-Aminobutírico/química , Animales , Ácidos Grasos Volátiles/análisis , Masculino , Ratones , Ratones Endogámicos ICR
16.
Food Funct ; 10(9): 6147-6156, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31498347

RESUMEN

The beneficial effects of the essential metal zinc (Zn) and probiotics on gut health have been well documented, but how they synergistically affect intestinal physiology is not thoroughly understood. In this study, the Zn-enriching ability of 33 probiotics in a medium or an aqueous solution was evaluated. A Lactobacillus plantarum strain, CCFM242, with a superior Zn-enriching ability was screened. Among the cellular components, the cell wall played the most important role in the Zn binding of L. plantarum CCFM242. The carboxyl and amino groups on the surface of the strain were also vital for Zn enrichment. Upon optimization of the Zn-enriching procedure, the Zn-binding ability of this strain reached 24.89 ± 0.50 mg g-1 dry biomass. Compared to the treatment of ZnSO4 or L. plantarum CCFM242, oral supplementation with Zn-enriched L. plantarum CCFM242 resulted in a higher serum Zn level, enhanced levels of mRNA expression of colonic tight junctions, increased levels of short-chain fatty acids (SCFAs) in colonic contents, and stronger modulatory effects on the anti-oxidant and immune defense systems in the gut of normal mice. Zn-Enriched L. plantarum CCFM242 treatment also offered more significant protective effects against dextran sodium sulfate (DSS)-induced colitis in mice compared to the treatment of ZnSO4 or L. plantarum CCFM242 alone. The synergistic effect of Zn-enriched L. plantarum CCFM242 may be due to the increased tolerance of the strain to the gastrointestinal tract conditions and the higher bioavailability of Zn after the metal-enrichment process.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Lactobacillus plantarum/fisiología , Probióticos/administración & dosificación , Zinc/administración & dosificación , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/fisiopatología , Colon/efectos de los fármacos , Colon/fisiopatología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Homeostasis/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
17.
Food Funct ; 10(9): 5398-5412, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31397465

RESUMEN

In this study, the effects of three doses (diets containing <0.01, 0.15, 0.40 mg kg-1 Se) and two forms (sodium selenite and selenomethionine) of dietary Se supplementation on the intestinal physiology of untreated, dextran sodium sulfate-treated, and Salmonella typhimurium-infected mice were evaluated. The underlying modes of action of the varied doses and forms of Se supplementation were analyzed using fecal metabolomic and jejunal proteomic approaches. Compared with adequate Se (0.15 mg kg-1 Se) supplementation, Se-deficiency supplementation adversely affected the gut barrier and intestinal immune responses of the untreated mice and increased their susceptibility to experimental colitis and pathogen infection. In contrast, supranutritional Se (0.40 mg kg-1 Se) supplementation improved mouse intestinal physiology compared with adequate Se supplementation. Varied doses of Se supplementation differentially perturbed the fecal metabolic profiles of and jejunal protein expression in mice. Further, both forms of dietary Se supplementation, i.e., sodium selenite and selenomethionine, showed similar effects on the gut barrier and intestinal immune homeostasis but differentially affected fecal metabolites, such as neurosubstances and immunomodulators, and induced significant proteomic variations in various pathways, including the xenobiotic detoxification pathway and glutathione metabolism. Our results indicate that the doses and chemical forms of Se should be considered when developing dietary nutritional supplements for gut health.


Asunto(s)
Intestinos/inmunología , Infecciones por Salmonella/inmunología , Selenometionina/administración & dosificación , Selenito de Sodio/administración & dosificación , Animales , Suplementos Dietéticos/análisis , Humanos , Intestinos/efectos de los fármacos , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteómica , Infecciones por Salmonella/tratamiento farmacológico , Infecciones por Salmonella/microbiología , Salmonella typhimurium/fisiología
18.
Food Funct ; 10(3): 1330-1339, 2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30741300

RESUMEN

Patulin (PAT) is a mycotoxin widely found in fruits and vegetables. Several reviews and studies have hypothesized that in vivo PAT toxicity is related to gut barrier dysfunction, but evidence for this is not substantial. The goal of the present study was to further demonstrate the role of the gut barrier in food-borne PAT toxicity. In vitro assays showed that PAT exposure induced significant cell death, inhibited the mRNA expressions of tight junction proteins and increased gut permeability in Caco-2 cell monolayers. An acute PAT exposure animal trial reported for the first time an association between PAT-induced disruption of the gut barrier and endotoxemia in mice. Sub-chronic PAT exposure also inhibited the expression of ZO-1 in the gut and induced both intestinal and systematic inflammation in mice. Dietary supplements with previously reported protective effects on the gut barrier, such as docosahexaenoic acid and Lactobacillus plantarum CCFM8610, were able to recover the PAT-induced gut barrier dysfunction and significantly alleviate PAT toxicity in vivo. Another L. plantarum strain, CCFM11, with poor gut barrier modulation ability, failed to exhibit identical protective effects against PAT toxicity to L. plantarum CCFM8610. Our results indicated that PAT-induced disruption of the gut barrier and bacterial translocation may be another toxic mechanism of PAT besides its inherent cytotoxicity. Gut barrier protection may be considered an important target for the prevention of PAT toxicity.


Asunto(s)
Suplementos Dietéticos , Ácidos Docosahexaenoicos/farmacología , Contaminación de Alimentos , Patulina/toxicidad , Probióticos/administración & dosificación , Animales , Células CACO-2 , Ácidos Docosahexaenoicos/administración & dosificación , Humanos , Lactobacillus plantarum , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
19.
Front Microbiol ; 9: 2134, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30254621

RESUMEN

Lead (Pb) intoxication is a serious food safety issue, and the development of relevant dietary strategies is an area of ongoing research. In this study, two different dietary supplements were designed and evaluated for their effects against Pb toxicity in mice. Dietary supplement A contained grape seed extract, tea polyphenols and Lactobacillus plantarum CCFM8661, and dietary supplement B contained vitamin C, calcium carbonate, zinc acetate, and L. plantarum CCFM8661. The results showed that both dietary supplements could effectively decrease Pb levels, protect aminolevulinic acid dehydratase, superoxide dismutase and catalase activities and recover glutathione, zinc protoporphyrin and malondialdehyde levels in tissues and blood of mice. A step-through passive avoidance task confirmed that the dietary supplements could recover the learning and memory capacities of Pb-exposed mice. The protective effects of both dietary supplements to alleviate oxidative stress and cognitive impairments were superior to the chelator treatment. Administration of the dietary supplements during Pb exposure offered more significant protection than administration after Pb exposure. Animal safety evaluation also indicated that these dietary supplements barely induced side effects in the mice. This study provides evidence that dietary supplements containing probiotics, micronutrients, and plant extracts can be considered a new dietary strategy against Pb toxicity.

20.
Ecotoxicol Environ Saf ; 143: 307-314, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28570951

RESUMEN

We investigated the protection offered by the probiotic Lactobacillus plantarum CCFM639 against waterborne Al exposure in tilapia. Fish were allocated to control, CCFM639-only, Al-only or Al plus CCFM639 groups. The fish were exposed to 2.73mg/L Al ions for 4 weeks. The probiotic was incorporated into the fish diet at 108 CFU/g and provided twice daily. Our results showed that L. plantarum CCFM639 significantly enhanced feed utilization, growth performance and antioxidant ability in the absence of waterborne Al exposure. When fish were exposed to Al, dietary supplementation with the strain effectively decreased the death rate and accumulation of Al in tissues, and enhanced growth performance. Moreover, Al-induced changes in hematobiochemical parameters and hepatic oxidative stress and histopathology were also alleviated. Therefore, L. plantarum CCFM639 may be a novel dietary supplement for fish to enhance growth performance and prevent aquaculture and food safety problems induced by Al pollution.


Asunto(s)
Aluminio/toxicidad , Lactobacillus plantarum , Estrés Oxidativo/efectos de los fármacos , Tilapia/crecimiento & desarrollo , Contaminantes Químicos del Agua/toxicidad , Aluminio/metabolismo , Alimentación Animal , Animales , Suplementos Dietéticos , Explotaciones Pesqueras , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Probióticos , Tilapia/metabolismo , Contaminantes Químicos del Agua/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA