Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Food Res Int ; 191: 114720, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39059916

RESUMO

Mangosteen (Garcinia mangostana L.) is a tasty, polyphenol-rich tropical fruit. The edible part is highly appreciated by its aroma, taste and texture. The non-edible part, rich in polyphenols, has been traditionally used in Thai medicine. In this work, flavonoids and phenolic acid/derivatives were identified in mangosteen extracts (ME) from edible and non-edible portions. We first studied the effects of MEs on the growth, metabolism, antioxidant capacity, biofilm formation and antimicrobial capacity of eight bifidobacteria and lactobacilli strains from intestinal origin and two commercial probiotic strains (BB536 and GG). ME concentrations higher than 10-20 % were inhibitory for all strains. However, ME concentrations of 5 % significantly (P < 0.01) increased all strains antioxidant capacity, reduced biofilm-formation, and enhanced inhibition against Gram-positive pathogens. To apply these knowledge, bifunctional fermented milk products were elaborated with 5 % ME and individual strains, which were selected taking into account their growth with ME, and the widest range of values on antioxidant capacity, biofilm formation and antimicrobial activity (bifidobacteria INIA P2 and INIA P467, lactobacilli INIA P459 and INIA P708, and reference strain GG). Most strains survived well manufacture, refrigerated storage and an in vitro simulation of major conditions encountered in the gastrointestinal tract. As expected, products supplemented with ME showed higher polyphenol content and antioxidant capacity levels than control. After sensory evaluation, products containing strains INIA P2, INIA P708 and GG outstood as best.


Assuntos
Antioxidantes , Biofilmes , Produtos Fermentados do Leite , Garcinia mangostana , Lactobacillus , Extratos Vegetais , Extratos Vegetais/farmacologia , Garcinia mangostana/química , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Antioxidantes/farmacologia , Lactobacillus/efeitos dos fármacos , Lactobacillus/metabolismo , Produtos Fermentados do Leite/microbiologia , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Probióticos , Flavonoides/farmacologia , Flavonoides/análise , Humanos , Frutas/química , Frutas/microbiologia , Fermentação , Hidroxibenzoatos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Polifenóis/farmacologia
2.
Nutrients ; 16(13)2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38999750

RESUMO

(1) Background: Recently, academic studies are demonstrating that the cholesterol-lowering effects of pectin oligosaccharides (POSs) are correlated to intestinal flora. However, the mechanisms of POS on cholesterol metabolisms are limited, and the observations of intestinal flora are lacking integrative analyses. (2) Aim and methods: To reveal the regulatory mechanisms of POS on cholesterol metabolism via an integrative analysis of the gut microbiota, the changes in gut microbiota structure and metabolite composition after POS addition were investigated using Illumina MiSeq sequencing and non-targeted metabolomics through in vitro gut microbiota fermentation. (3) Results: The composition of fecal gut flora was adjusted positively by POS. POS increased the abundances of the cholesterol-related bacterial groups Bacteroidetes, Bifidobacterium and Lactobacillus, while it decreased conditional pathogenic Escherichia coli and Enterococcus, showing good prebiotic activities. POS changed the composition of gut microbiota fermentation metabolites (P24), causing significant changes in 221 species of fermentation metabolites in a non-targeted metabolomics analysis and promoting the production of short-chain fatty acids. The abundances of four types of cholesterol metabolism-related metabolites (adenosine monophosphate, cyclic adenosine monophosphate, guanosine and butyrate) were significantly higher in the P24 group than those in the control group without POS addition. (4) Conclusion: The abovementioned results may explain the hypocholesterolemic effects of POS and promotion effects on cholesterol efflux of P24. These findings indicated that the potential regulatory mechanisms of citrus POS on cholesterol metabolism are modulated by cholesterol-related gut microbiota and specific metabolites.


Assuntos
Colesterol , Fezes , Fermentação , Microbioma Gastrointestinal , Oligossacarídeos , Pectinas , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Pectinas/farmacologia , Pectinas/metabolismo , Colesterol/metabolismo , Oligossacarídeos/farmacologia , Fezes/microbiologia , Humanos , Prebióticos , Masculino , Metabolômica , Ácidos Graxos Voláteis/metabolismo , Bifidobacterium/metabolismo , Bifidobacterium/efeitos dos fármacos , Feminino , Bactérias/metabolismo , Bactérias/efeitos dos fármacos , Bactérias/classificação , Citrus
3.
J Sci Food Agric ; 104(14): 8500-8510, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38923512

RESUMO

BACKGROUND: Kratom (Mitragyna speciosa) has a long history of traditional use. It contains various alkaloids and polyphenols. The properties of kratom's alkaloids have been well-documented. However, the property of kratom's polyphenols in water-soluble phase have been less frequently reported. This study assessed the effects of water-soluble Mitragyna speciosa (kratom) extract (MSE) on gut microbiota and their metabolite production in fecal batch culture. RESULTS: The water-soluble kratom extract (MSE0) and the water-soluble kratom extract after partial sugar removal (MSE50) both contained polyphenols, with total phenolic levels of 2037.91 ± 51.13 and 3997.95 ± 27.90 mg GAE/g extract, respectively and total flavonoids of 81.10 ± 1.00 and 84.60 ± 1.43 mg CEQ/g extract. The gut microbiota in fecal batch culture was identified by 16S rRNA gene sequencing at 0 and 24 h of fermentation. After fermentation, MSE50 stimulated the growth of Bifidobacterium more than MSE0. MSE0 gave the highest total fatty acids level among the treatments. The phenolic metabolites produced by some intestinal microbiota during fecal fermentation at 24 h were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The major metabolite of biotransformation of both water-soluble MSEs by intestinal microbiota was pyrocatechol (9.85-11.53%). CONCLUSION: The water-soluble MSEs and their produced metabolites could potentially be used as ingredients for functional and medicinal food production that supports specific gut microbiota. © 2024 Society of Chemical Industry.


Assuntos
Fezes , Fermentação , Microbioma Gastrointestinal , Mitragyna , Extratos Vegetais , Polifenóis , Microbioma Gastrointestinal/efeitos dos fármacos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Extratos Vegetais/metabolismo , Fezes/microbiologia , Mitragyna/química , Mitragyna/metabolismo , Polifenóis/metabolismo , Polifenóis/farmacologia , Bactérias/classificação , Bactérias/genética , Bactérias/metabolismo , Bactérias/efeitos dos fármacos , Bactérias/isolamento & purificação , Humanos , Masculino , Flavonoides/metabolismo , Flavonoides/farmacologia , Bifidobacterium/metabolismo , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/efeitos dos fármacos
4.
Front Immunol ; 12: 772532, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970262

RESUMO

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


Assuntos
Antibacterianos/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/administração & dosagem , Lacticaseibacillus rhamnosus , Neoplasias/terapia , Probióticos/uso terapêutico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Bacteroides/efeitos dos fármacos , Bacteroides/crescimento & desenvolvimento , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/crescimento & desenvolvimento , Linhagem Celular Tumoral , Fezes/microbiologia , Camundongos Endogâmicos BALB C , Neoplasias/microbiologia
5.
Nutrients ; 13(11)2021 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-34836220

RESUMO

Grapes provide a rich source of polyphenols and fibers. This study aimed to evaluate the effect of the daily consumption of 46 g of whole grape powder, providing the equivalent of two servings of California table grapes, on the gut microbiome and cholesterol/bile acid metabolism in healthy adults. This study included a 4-week standardization to a low-polyphenol diet, followed by 4 weeks of 46 g of grape powder consumption while continuing the low-polyphenol diet. Compared to the baseline, 4 weeks of grape powder consumption significantly increased the alpha diversity index of the gut microbiome. There was a trend of increasing Verrucomicrobia (p = 0.052) at the phylum level, and a significant increase in Akkermansia was noted. In addition, there was an increase in Flavonifractor and Lachnospiraceae_UCG-010, but a decrease in Bifidobacterium and Dialister at the genus level. Grape powder consumption significantly decreased the total cholesterol by 6.1% and HDL cholesterol by 7.6%. There was also a trend of decreasing LDL cholesterol by 5.9%, and decreasing total bile acid by 40.9%. Blood triglyceride levels and body composition were not changed by grape powder consumption. In conclusion, grape powder consumption significantly modified the gut microbiome and cholesterol/bile acid metabolism.


Assuntos
Ácidos e Sais Biliares/metabolismo , Colesterol/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Vitis/química , Adulto , Akkermansia/efeitos dos fármacos , Bifidobacterium/efeitos dos fármacos , Colesterol/sangue , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Polifenóis/metabolismo , Pós , Triglicerídeos/sangue , Verrucomicrobia/efeitos dos fármacos , Adulto Jovem
6.
Cancer Sci ; 112(10): 4050-4063, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34289209

RESUMO

Astragalus polysaccharides (APS), the main effective component of Astragalus membranaceus, can inhibit tumor growth, but the underlying mechanisms remain unclear. Previous studies have suggested that APS can regulate the gut microenvironment, including the gut microbiota and fecal metabolites. In this work, our results showed that APS could control tumor growth in melanoma-bearing mice. It could reduce the number of myeloid-derived suppressor cells (MDSC), as well as the expression of MDSC-related molecule Arg-1 and cytokines IL-10 and TGF-ß, so that CD8+ T cells could kill tumor cells more effectively. However, while APS were administered with an antibiotic cocktail (ABX), MDSC could not be reduced, and the growth rate of tumors was accelerated. Consistent with the changes in MDSC, the serum levels of IL-6 and IL-1ß were lowest in the APS group. Meanwhile, we found that fecal suspension from mice in the APS group could also reduce the number of MDSC in tumor tissues. These results revealed that APS regulated the immune function in tumor-bearing mice through remodeling the gut microbiota. Next, we focused on the results of 16S rRNA, which showed that APS significantly regulated most microorganisms, such as Bifidobacterium pseudolongum, Lactobacillus johnsonii and Lactobacillus. According to the Spearman analysis, the changes in abundance of these microorganisms were related to the increase of metabolites like glutamate and creatine, which could control tumor growth. The present study demonstrates that APS attenuate the immunosuppressive activity of MDSC in melanoma-bearing mice by remodeling the gut microbiota and fecal metabolites. Our findings reveal the therapeutic potential of APS to control tumor growth.


Assuntos
Astrágalo/química , Linfócitos T CD8-Positivos/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Melanoma/tratamento farmacológico , Células Supressoras Mieloides/efeitos dos fármacos , Polissacarídeos/farmacologia , Animais , Antibacterianos/administração & dosagem , Arginase/efeitos dos fármacos , Arginase/metabolismo , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/metabolismo , Combinação de Medicamentos , Transplante de Microbiota Fecal , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Tolerância Imunológica , Interleucina-10/metabolismo , Interleucina-1beta/sangue , Interleucina-6/sangue , Lactobacillus/efeitos dos fármacos , Masculino , Melanoma/imunologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , RNA Ribossômico 16S/análise , Fator de Crescimento Transformador beta/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral/imunologia
7.
Sci Rep ; 11(1): 11264, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34050197

RESUMO

Short-term changes in dietary intake can induce changes in gut microbiome. While various dietary polyphenols have been shown to modulate gut microflora, the acute influence of polyphenol-rich mixed spices has not been explored in a controlled setting. We investigated the effects of a single serving of mixed spices Indian curry consumption, in two separate doses, on the gut microbiome in 15 healthy, Singaporean Chinese males, with age and BMI of 23.5 ± 2.4 years and 22.9 ± 2.2 kg/m2 respectively. We found that a low-polyphenol, no spices Dose 0 Control (D0C) meal led to an increase in Bacteroides and a decrease in Bifidobacterium. In comparison to D0C, there was significant suppression of Bacteroides (p < 0.05) and an increase in Bifidobacterium (p < 0.05) with increasing doses of curry meal Dose 1 Curry (D1C) and Dose 2 Curry (D2C) containing 6 g and 12 g mixed spices respectively. Significant correlations were also found between bacterial changes and plasma phenolic acids. No differences between treatments were observed in the alpha-diversity of the gut microflora. This study has shown that a single serving of mixed spices can significantly modify/restore certain commensal microbes, particularly in people who do not regularly consume these spices.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Polifenóis/farmacologia , Bacteroides/efeitos dos fármacos , Bifidobacterium/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Humanos , Masculino , Refeições , Período Pós-Prandial/efeitos dos fármacos , Singapura , Especiarias/microbiologia , Adulto Jovem
8.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188494, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33346129

RESUMO

Bacteria present in probiotics, particularly the common Lactobacillus and Bifidobacterium microbes, have been found to induce anti-cancer action by enhancing cancer cell apoptosis and protecting against oxidative stress. Probiotics supplements also decrease the cancer-producing microorganism Fusobacterium. Studies have demonstrated that gut microbiota modifies the effect of chemo/radiation therapy. Gut microbes not only enhance the action of chemotherapy drugs but also reduce the side effects of these medications. Additionally, gut microbes reduce immunotherapy toxicity, in particular, the presence of Bacteroidetes or Bifidobacterium decreases the development of colitis by ipilimumab therapy. Probiotics supplements containing Bifidobacterium also reduce chemotherapy-induced mucositis and radiation-induced diarrhea. This review focused on elucidating the mechanism behind the anti-cancer action of Bifidobacterium species. Available studies have revealed Bifidobacterium species decrease cancer cell proliferation via the inhibition of growth factor signaling as well as inducing mitochondrial-mediated apoptosis. Moreover, Bifidobacterium species reduce the adverse effects of chemo/immuno/radiation therapy by inhibiting proinflammatory cytokines. Further clinical studies are needed to identify the powerful and suitable Bifidobacterium strain for the development of adjuvant therapy to support chemo/immuno/radiation therapy.


Assuntos
Antineoplásicos/uso terapêutico , Microbioma Gastrointestinal/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Probióticos/uso terapêutico , Antineoplásicos/efeitos adversos , Bifidobacterium/efeitos dos fármacos , Citocinas/genética , Suplementos Nutricionais/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Lactobacillus/efeitos dos fármacos , Neoplasias/genética , Neoplasias/microbiologia , Neoplasias/patologia
9.
Nutrients ; 12(9)2020 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-32933181

RESUMO

Human milk oligosaccharides (HMOs) shape the gut microbiota in infants by selectively stimulating the growth of bifidobacteria. Here, we investigated the impact of HMOs on adult gut microbiota and gut barrier function using the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), Caco2 cell lines, and human intestinal gut organoid-on-chips. We showed that fermentation of 2'-O-fucosyllactose (2'FL), lacto-N-neotetraose (LNnT), and combinations thereof (MIX) led to an increase of bifidobacteria, accompanied by an increase of short chain fatty acid (SCFA), in particular butyrate with 2'FL. A significant reduction in paracellular permeability of FITC-dextran probe was observed using Caco2 cell monolayers with fermented 2'FL and MIX, which was accompanied by an increase in claudin-8 gene expression as shown by qPCR, and a reduction in IL-6 as determined by multiplex ELISA. Using gut-on-chips generated from human organoids derived from proximal, transverse, and distal colon biopsies (Colon Intestine Chips), we showed that claudin-5 was significantly upregulated across all three gut-on-chips following treatment with fermented 2'FL under microfluidic conditions. Taken together, these data show that, in addition to their bifidogenic activity, HMOs have the capacity to modulate immune function and the gut barrier, supporting the potential of HMOs to provide health benefits in adults.


Assuntos
Bifidobacterium/efeitos dos fármacos , Colo/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Leite Humano/química , Oligossacarídeos/farmacologia , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Ácido Butírico/metabolismo , Células CACO-2 , Claudinas/metabolismo , Colo/metabolismo , Colo/microbiologia , Ensaio de Imunoadsorção Enzimática , Fermentação , Humanos , Imunidade , Lactente , Interleucina-6/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Trissacarídeos/farmacologia , Regulação para Cima
10.
Toxicol Appl Pharmacol ; 391: 114900, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32061593

RESUMO

Methotrexate (MTX) is a widely used therapeutic agent for the treatment of cancer and autoimmune diseases. However, its efficacy is often limited by adverse effects, such as intestinal toxicity. Although treatment with leucovorin (LV) is the most common method to reduce the toxic effects of MTX, it may also compromise the therapeutic effects of MTX. The gut microbiome has been reported to be associated with the intestinal toxicity of MTX. In this study, the intestinal damage of MTX was ameliorated by treatment with LV. Moreover, the population, diversity, and principal components of the gut microbiota in MTX-treated mice were restored by treatment with LV. The only element of the gut microbiota that was significantly changed after treatment with LV was Bifidobacterium, and supplementation with Bifidobacterium longum ameliorated MTX-induced intestinal damage. In conclusion, our results suggest that the balance and the composition of gut microbiota have an important role in the LV-mediated protection against MTX-induced intestinal toxicity. This work provides foundation of data in support of a new potential mechanism for the prevention of MTX-induced intestinal toxicity.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Enteropatias/induzido quimicamente , Enteropatias/tratamento farmacológico , Leucovorina/uso terapêutico , Metotrexato/toxicidade , Animais , Bifidobacterium/efeitos dos fármacos , Colo/patologia , DNA Bacteriano/genética , Enteropatias/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Ribossômico 16S/genética , Redução de Peso/efeitos dos fármacos
11.
Bull Exp Biol Med ; 168(3): 309-312, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31938911

RESUMO

We studied the effect of combined antimicrobial therapy with amoxicillin, metronidazole, and clarithromycin on the severity of ischemia/reperfusion myocardial injury in Wistar rats with alimentary obesity and acute inflammation of the large intestine. General ischemia/reperfusion was reproduced on Langendorff-perfused isolated hearts and infarct size was estimated. Acute inflammation of the large intestine was accompanied by an increase in the blood levels of proinflammatory cytokines. The presence of obesity and acute inflammation of the large intestine did not significantly affect the infarct size in comparison with the control. Administration of antimicrobial drugs to animals with obesity and acute inflammation of the large intestine led to a significant increase in the infarct size, which should be considered when prescribing antimicrobial therapy to patients with comorbidity.


Assuntos
Inflamação/tratamento farmacológico , Inflamação/imunologia , Intestino Grosso/imunologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Anti-Infecciosos/uso terapêutico , Bifidobacterium/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Inflamação/metabolismo , Interleucina-8/metabolismo , Intestino Grosso/microbiologia , Lactobacillus/efeitos dos fármacos , Masculino , Miocárdio/metabolismo , Obesidade/tratamento farmacológico , Obesidade/imunologia , Obesidade/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
12.
Molecules ; 24(20)2019 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-31640295

RESUMO

Two by-products containing phenols and polysaccharides, a "pâté" (OP) from the extra virgin olive oil milling process and a decoction of pomegranate mesocarp (PM), were investigated for their effects on human microbiota using the SHIME® system. The ability of these products to modulate the microbial community was studied simulating a daily intake for nine days. Microbial functionality, investigated in terms of short chain fatty acids (SCFA) and NH4+, was stable during the treatment. A significant increase in Lactobacillaceae and Bifidobacteriaceae at nine days was induced by OP mainly in the proximal tract. Polyphenol metabolism indicated the formation of tyrosol from OP mainly in the distal tract, while urolithins C and A were produced from PM, identifying the human donor as a metabotype A. The results confirm the SHIME® system as a suitable in vitro tool to preliminarily investigate interactions between complex botanicals and human microbiota before undertaking more challenging human studies.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Olea/química , Fenóis/administração & dosagem , Polissacarídeos/administração & dosagem , Punica granatum/química , Compostos de Amônio/metabolismo , Bifidobacterium/classificação , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/isolamento & purificação , DNA Bacteriano/análise , Ácidos Graxos Voláteis/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactobacillaceae/classificação , Lactobacillaceae/efeitos dos fármacos , Lactobacillaceae/isolamento & purificação , Fenóis/química , Fenóis/farmacologia , Filogenia , Polissacarídeos/química , Polissacarídeos/farmacologia
13.
Food Funct ; 10(7): 4134-4142, 2019 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-31241125

RESUMO

This study aimed to investigate the effect of putrescine on the immune function and intestinal bacteria of weaning piglets. Twenty-four male castrated weaning piglets on their 21st day were randomly assigned into four groups: control (basal diet) and treatment groups given basal diets supplemented with 0.05%, 0.1%, and 0.15% putrescine for 11 days. Results were as follows: (1) Dietary putrescine increased the villus height, width, height/crypt depth and surface area, and decreased the diarrhea index (P < 0.05). (2) Dietary putrescine increased the lysozyme and acid phosphatase activities and the amount of immunoglobulin M, antibacterial peptides, and transforming growth factor ß1, but decreased the mRNA levels of tumor necrosis factor α, interleukin-6, interleukin-8 and inducible nitric oxide synthase (P < 0.05). (3) Dietary putrescine increased the mRNA expression of the mammalian target of rapamycin, signal transducer and activator of transcription, and Janus kinase 2 but decreased the mRNA expression of nuclear factor-kappa B P65 (P < 0.05). (4) Dietary putrescine increased the population of total bacteria, Lactobacillus, and Bifidobacterium and decreased that of Escherichia coli in the colon and cecum (P < 0.05). (5) Finally, dietary putrescine increased the concentrations of butyrate and total volatile fatty acids in the colon and those of acetate, propionate, and total volatile fatty acids in the cecum (P < 0.05). Overall, putrescine can enhance intestinal development, improve immune functions, and regulate the population of intestinal bacteria in weaning piglets.


Assuntos
Suplementos Nutricionais , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Intestinos/microbiologia , Putrescina/farmacologia , Desmame , Fosfatase Ácida/metabolismo , Animais , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/crescimento & desenvolvimento , Butiratos/metabolismo , Ceco/metabolismo , Ceco/microbiologia , Colo/metabolismo , Colo/microbiologia , Diarreia/prevenção & controle , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Ácidos Graxos Voláteis/metabolismo , Imunoglobulina M , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Janus Quinase 2/metabolismo , Lactobacillus/efeitos dos fármacos , Lactobacillus/crescimento & desenvolvimento , Masculino , Muramidase/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Suínos
14.
Biomed Res Int ; 2019: 4530203, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31073525

RESUMO

Effects of the microbiome associated with diarrhea-predominant irritable bowel syndrome (IBS-D) on the gut have been reported, but no study has reported the effects of the IBS-D gut microbiome on the liver. We transplanted the fecal microbiota from an IBS-D patient and from a healthy volunteer to GF rats. The hepatic inflammation, serum biochemical parameters and metabolome, fecal microbiota profile, fecal short-chain fatty acids (SCFAs), and correlations among them before and after berberine intervention were assessed. Compared with the healthy control fecal microbiome transplantation (FMT) rats, the fecal microbiota of IBS-D patients induces significant Kupffer cell hyperplasia, hepatic sinusoid hypertrophy, and elevated levels of hepatic tumor necrosis factor-α and interferon-γ and decreases the synthesis of ALB in GF rats. This is possibly related to Faecalibacterium and Bifidobacterium attributable to fecal formate, acetate, and propionate levels, which are associated with the host linoleic acid pathway. Berberine can partially reverse the Kupffer cell hyperplasia, Faecalibacterium, fecal formate, acetate, and propionate by modulating the gut microbiome composition. These results may imply that IBS-D not only is an intestinal functional disorder but can cause liver inflammation, thus providing some implications regarding the clinical cognition and treatment of IBS-D.


Assuntos
Berberina/administração & dosagem , Diarreia/tratamento farmacológico , Inflamação/tratamento farmacológico , Síndrome do Intestino Irritável/tratamento farmacológico , Animais , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/patogenicidade , Diarreia/microbiologia , Diarreia/patologia , Modelos Animais de Doenças , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/patogenicidade , Transplante de Microbiota Fecal , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Inflamação/microbiologia , Inflamação/patologia , Síndrome do Intestino Irritável/microbiologia , Síndrome do Intestino Irritável/patologia , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/patologia , Fígado/microbiologia , Fígado/patologia , Ratos
15.
J Nutr Biochem ; 67: 20-27, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30831460

RESUMO

Iron supplements are widely consumed; however most of the iron is not absorbed and enters the colon where potentially pathogenic bacteria can utilise it for growth. This study investigated the effect of iron availability on human gut microbial composition and function using an in vitro colonic fermentation model inoculated with faecal microbiota from healthy adult donors, as well as examining the effect of iron on the growth of individual gut bacteria. Batch fermenters were seeded with fresh faecal material and supplemented with the iron chelator, bathophenanthroline disulphonic acid (BPDS). Samples were analysed at regular intervals to assess impact on the gut bacterial communities. The growth of Escherichia coli and Salmonella typhimurium was significantly impaired when cultured independently in iron-deficient media. In contrast, depletion of iron did not affect the growth of the beneficial species, Lactobacillus rhamnosus, when cultured independently. Analysis of the microbiome composition via 16S-based metataxonomics indicated that under conditions of iron chelation, the relative abundance decreased for several taxa, including a 10% decrease in Escherichia and a 15% decrease in Bifidobacterium. Metabolomics analysis using 1 H-NMR indicated that the production of SCFAs was reduced under iron-limited conditions. These results support previous studies demonstrating the essentiality of iron for microbial growth and metabolism, but, in addition, they indicate that iron chelation changes the gut microbiota profile and influences human gut microbial homeostasis through both compositional and functional changes.


Assuntos
Colo/microbiologia , Microbioma Gastrointestinal/fisiologia , Ferro/farmacocinética , Técnicas Bacteriológicas , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/crescimento & desenvolvimento , Disponibilidade Biológica , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Fermentação , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Microbiota , RNA Ribossômico 16S , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/patogenicidade
16.
Br J Nutr ; 121(5): 549-559, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30688188

RESUMO

Wholegrain oats are known to modulate the human gut microbiota and have prebiotic properties (increase the growth of some health-promoting bacterial genera within the colon). Research to date mainly attributes these effects to the fibre content; however, oat is also a rich dietary source of polyphenols, which may contribute to the positive modulation of gut microbiota. In vitro anaerobic batch-culture experiments were performed over 24 h to evaluate the impact of two different doses (1 and 3 % (w/v)) of oat bran, matched concentrations of ß-glucan extract or polyphenol mix, on the human faecal microbiota composition using 16S RNA gene sequencing and SCFA analysis. Supplementation with oats increased the abundance of Proteobacteria (P <0·01) at 10 h, Bacteroidetes (P <0·05) at 24 h and concentrations of acetic and propionic acid increased at 10 and 24 h compared with the NC. Fermentation of the 1 % (w/v) oat bran resulted in significant increase in SCFA production at 24 h (86 (sd 27) v. 28 (sd 5) mm; P <0·05) and a bifidogenic effect, increasing the relative abundance of Bifidobacterium unassigned at 10 h and Bifidobacterium adolescentis (P <0·05) at 10 and 24 h compared with NC. Considering the ß-glucan treatment induced an increase in the phylum Bacteroidetes at 24 h, it explains the Bacteriodetes effects of oats as a food matrix. The polyphenol mix induced an increase in Enterobacteriaceae family at 24 h. In conclusion, in this study, we found that oats increased bifidobacteria, acetic acid and propionic acid, and this is mediated by the synergy of all oat compounds within the complex food matrix, rather than its main bioactive ß-glucan or polyphenols. Thus, oats as a whole food led to the greatest impact on the microbiota.


Assuntos
Avena/química , Bacteroidetes/efeitos dos fármacos , Bifidobacterium/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Grãos Integrais , Ácido Acético/metabolismo , Fezes/microbiologia , Fermentação/efeitos dos fármacos , Humanos , Polifenóis/farmacologia , Prebióticos , Propionatos/metabolismo , Proteobactérias/efeitos dos fármacos , beta-Glucanas/farmacologia
17.
J Hum Nutr Diet ; 32(1): 53-62, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29984532

RESUMO

BACKGROUND: Evidence has shown that anthocyanins, a subclass of polyphenol, are metabolised in the gut, modulate bacterial species and exert bioactive effects through this interaction. METHODS: A systematic literature review was undertaken to determine the level of current evidence for the association between anthocyanin intake and changes in gut microbiota populations. The studies included were also assessed for the different techniques used in microbiota determination. Following the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, scientific databases, including Scopus, PubMed, ScienceDirect, Web of Science and MEDLINE, were searched up to June 2017. Details on population/sample, study design, intervention/control, dosage and method of microbiota determination were extracted. RESULTS: Six studies (three in vitro, two animal and one human trials) were included in the review, which showed that anthocyanins induced a significant proliferative effect on Bifidobacterium spp., known for their wide use in probiotics and for the treatment of irritable bowel syndrome. There was also an observed inhibition of Clostridium histolyticum, which was shown to be pathogenic in humans. The depth of analysis is an important consideration for the choice of microbiota determination technique with respect to a comprehensive, high-resolution microbiota analysis or analysis of the main microbiota taxa. CONCLUSIONS: Very limited research has been carried out in the area of anthocyanins and gut microbiota; beneficial effects have generally been observed, and further clinical trials in humans are needed to confirm changes to gut microbes in relation to dietary anthocyanin intake and potential health benefits.


Assuntos
Antocianinas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Probióticos/farmacologia , Animais , Bifidobacterium/efeitos dos fármacos , Clostridium histolyticum/efeitos dos fármacos , Humanos
18.
Sci Rep ; 8(1): 12772, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143740

RESUMO

In the current report, we describe the identification of three genetically distinct groups of prophages integrated into three different chromosomal sites of human gut-associated Bifidobacterium breve and Bifidobacterium longum strains. These bifidobacterial prophages are distantly related to temperate actinobacteriophages of several hosts. Some prophages, integrated within the dnaJ2 gene, are competent for induction, excision, replication, assembly and lysis, suggesting that they are fully functional and can generate infectious particles, even though permissive hosts have not yet been identified. Interestingly, several of these phages harbor a putative phase variation shufflon (the Rin system) that generates variation of the tail-associated receptor binding protein (RBP). Unlike the analogous coliphage-associated shufflon Min, or simpler Cin and Gin inversion systems, Rin is predicted to use a tyrosine recombinase to promote inversion, the first reported phage-encoded tyrosine-family DNA invertase. The identification of bifidobacterial prophages with RBP diversification systems that are competent for assembly and lysis, yet fail to propagate lytically under laboratory conditions, suggests dynamic evolution of bifidobacteria and their phages in the human gut.


Assuntos
Bifidobacterium/virologia , Microbioma Gastrointestinal , Prófagos/fisiologia , Sítios de Ligação Microbiológicos/genética , Sequência de Bases , Bifidobacterium/efeitos dos fármacos , Evolução Biológica , Microbioma Gastrointestinal/efeitos dos fármacos , Genoma Viral , Especificidade de Hospedeiro/efeitos dos fármacos , Especificidade de Hospedeiro/genética , Humanos , Mitomicina/farmacologia , Prófagos/efeitos dos fármacos , Prófagos/genética , Prófagos/ultraestrutura , Vírion/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
19.
J Nutr Biochem ; 59: 160-172, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30055451

RESUMO

Tart cherries have been reported to exert potential health benefits attributed to their specific and abundant polyphenol content. However, there is a need to study the impact and fate of tart cherries polyphenols in the gut microbiota. Here, tart cherries, pure polyphenols (and apricots) were submitted to in vitro bacterial fermentation assays and assessed through 16S rRNA gene sequence sequencing and metabolomics. A short-term (5 days, 8 oz. daily) human dietary intervention study was also conducted for microbiota analyses. Tart cherry concentrate juices were found to contain expected abundances of anthocyanins (cyanidin-glycosylrutinoside) and flavonoids (quercetin-rutinoside) and high amounts of chlorogenic and neochlorogenic acids. Targeted metabolomics confirmed that gut microbes were able to degrade those polyphenols mainly to 4-hydroxyphenylpropionic acids and to lower amounts of epicatechin and 4-hydroxybenzoic acids. Tart cherries were found to induce a large increase of Bacteroides in vitro, likely due to the input of polysaccharides, but prebiotic effect was also suggested by Bifidobacterium increase from chlorogenic acid. In the human study, two distinct and inverse responses to tart cherry consumption were associated with initial levels of Bacteroides. High-Bacteroides individuals responded with a decrease in Bacteroides and Bifidobacterium, and an increase of Lachnospiraceae, Ruminococcus and Collinsella. Low-Bacteroides individuals responded with an increase in Bacteroides or Prevotella and Bifidobacterium, and a decrease of Lachnospiraceae, Ruminococcus and Collinsella. These data confirm that gut microbiota metabolism, in particular the potential existence of different metabotypes, needs to be considered in studies attempting to link tart cherries consumption and health.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Polifenóis/farmacologia , Prunus avium/química , Adulto , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/genética , Feminino , Fermentação , Sucos de Frutas e Vegetais , Microbioma Gastrointestinal/genética , Humanos , Masculino , Fenóis/metabolismo , Polifenóis/análise , Polifenóis/farmacocinética
20.
Nutr Diabetes ; 8(1): 15, 2018 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-29549311

RESUMO

BACKGROUND: Non-digestible carbohydrates present in cereals such as fructans and arabinoxylans represent promising prebiotic nutrients to prevent the development of obesity and related metabolic disorders. OBJECTIVE AND DESIGN: The aim of this study was to determine the corrective effects of wheat bran-derived arabinoxylan oligosaccharides in obese mice fed a western diet (WD). WD was given for 4 weeks before wheat bran extract (WBE) supplementation (5%) for an additional 4 weeks, whereas a control group received the standard diet. RESULTS: Bifidogenic effect of WBE was evidenced by an induction of both Bifidobacterium animalis and Bifidobacterium pseudolongum in the caecal content. WBE supplementation normalised WD-induced fat-mass expansion, steatosis, hypercholesterolemia, hyperleptinemia, hyperglycemia and hyperinsulinemia reaching the values of control mice. The reduced glucose-dependent insulinotropic polypeptide (GIP) release observed in WD + WBE mice may be a protective mechanism in terms of reducing adipose tissue storage, hepatic steatosis and glucose homoeostasis. CONCLUSION: We found that WBE completely abolished WD-induced metabolic disorders. Those results might be useful to take into account nutritional advices to treat obesity and related metabolic disorders such as type 2 diabetes, hypercholesterolaemia and fatty liver diseases when obesity was already established.


Assuntos
Bifidobacterium/efeitos dos fármacos , Dieta Ocidental/efeitos adversos , Doenças Metabólicas/tratamento farmacológico , Obesidade/tratamento farmacológico , Oligossacarídeos/uso terapêutico , Triticum/química , Xilanos/uso terapêutico , Tecido Adiposo/metabolismo , Animais , Bifidobacterium/crescimento & desenvolvimento , Glicemia/metabolismo , Ceco/microbiologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/microbiologia , Fibras na Dieta , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Fígado Gorduroso/microbiologia , Polipeptídeo Inibidor Gástrico/sangue , Hipercolesterolemia/sangue , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/microbiologia , Hiperglicemia/sangue , Hiperglicemia/tratamento farmacológico , Hiperglicemia/microbiologia , Hiperinsulinismo/sangue , Hiperinsulinismo/tratamento farmacológico , Hiperinsulinismo/microbiologia , Leptina/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/complicações , Obesidade/metabolismo , Obesidade/microbiologia , Oligossacarídeos/farmacologia , Prebióticos , Xilanos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA