Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Agric Food Chem ; 72(9): 5014-5025, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38388339

RESUMO

Nε-carboxymethyllysine (CML) is produced by a nonenzymatic reaction between reducing sugar and ε-amino group of lysine in food and exists as free and bound forms with varying digestibility and absorption properties in vivo, causing diverse interactions with gut microbiota. The effects of different forms of dietary CML on the gut microbiota and intestinal barrier of mice were explored. Mice were exposed to free and bound CML for 12 weeks, and colonic morphology, gut microbiota, fecal short-chain fatty acids (SCFAs), intestinal barrier, and receptor for AGE (RAGE) signaling cascades were measured. The results indicated that dietary-free CML increased the relative abundance of SCFA-producing genera including Blautia, Faecalibacterium, Agathobacter, and Roseburia. In contrast, dietary-bound CML mainly increased the relative abundance of Akkermansia. Moreover, dietary-free and -bound CML promoted the gene and protein expression of zonula occludens-1 and claudin-1. Additionally, the intake of free and bound CML caused an upregulation of RAGE expression but did not activate downstream inflammatory pathways due to the upregulation of oligosaccharyl transferase complex protein 48 (AGER1) expression, indicating a delicate balance between protective and proinflammatory effects in vivo. Dietary-free and -bound CML could modulate the gut microbiota community and increase tight-junction expression, and dietary-free CML might exert a higher potential benefit on gut microbiota and SCFAs than dietary-bound CML.


Assuntos
Microbioma Gastrointestinal , Lisina , Lisina/análogos & derivados , Animais , Camundongos , Lisina/metabolismo , Intestinos , Dieta
2.
Food Funct ; 15(3): 1250-1264, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38194248

RESUMO

Non-alcoholic fatty liver disease (NAFLD) has become one of the most common chronic liver diseases worldwide. NAFLD is caused by numerous factors, including the genetic susceptibility, oxidative stress, unhealthy diet, and gut microbiota dysbiosis. Among these, gut microbiota is a key factor and plays an important role in the development of NAFLD. Therefore, modulating the composition and structure of gut microbiota might provide a new intervention strategy for NAFLD. Highland barley ß-glucan (HBG) is a polysaccharide that can interact with gut microbiota after entering the lower gastrointestinal tract and subsequently improves NAFLD. Therefore, a Western diet was used to induce NAFLD in mouse models and the intervention effects and underlying molecular mechanisms of HBG on NAFLD mice based on gut microbiota were explored. The results indicated that HBG could regulate the composition of gut microbiota in NAFLD mice. In particular, HBG increased the abundance of short-chain fatty acids (SCFA)-producing bacteria (Prevotella-9, Bacteroides, and Roseburia) as well as SCFA contents. The increase in SCFA contents might activate the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway, thereby improving the liver lipid metabolism disorder and reducing liver lipid deposition.


Assuntos
Microbioma Gastrointestinal , Hordeum , Hepatopatia Gordurosa não Alcoólica , beta-Glucanas , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , beta-Glucanas/farmacologia , Dieta Ocidental/efeitos adversos , Fígado/metabolismo , Suplementos Nutricionais , Lipídeos/farmacologia , Camundongos Endogâmicos C57BL , Dieta Hiperlipídica
3.
Crit Rev Food Sci Nutr ; 63(19): 3612-3633, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-34698575

RESUMO

Increased intake of Western diets and ultra-processed foods is accompanied by increased intake of advanced glycation end products (AGEs). AGEs can be generated exogenously in the thermal processing of food and endogenously in the human body, which associated with various chronic diseases. In food, AGEs can be divided into free and bound forms, which differ in their bioavailability, digestion, absorption, gut microbial interactions and untargeted metabolites. We summarized the measurements and contents of free and bound AGE in foods. Moreover, the ingestion, digestion, absorption, excretion, gut microbiota interactions, and metabolites and metabolic pathways between free and bound AGEs based on animal and human studies were compared. Bound AGEs were predominant in most of the selected foods, while beer and soy sauce were rich in free AGEs. Only 10%-30% of AGEs were absorbed into the systemic circulation when orally administered. The excretion of ingested free and bound AGEs was approximately 90% and 60%, respectively. Dietary free CML has a detrimental effect on gut microbiota composition, while bound AGEs have both detrimental and beneficial impacts. Free and bound dietary AGEs changed amino acid metabolism, energy metabolism and carbohydrate metabolism. And besides, bound dietary AGEs altered vitamin metabolism, and glycerolipid metabolism.


Assuntos
Produtos Finais da Glicação Avançada em Alimentos , Microbioma Gastrointestinal , Animais , Humanos , Produtos Finais de Glicação Avançada/metabolismo , Dieta , Alimentos
4.
Food Funct ; 14(2): 845-856, 2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36537141

RESUMO

Advanced glycation end products (AGEs) are increasingly recognized as potentially pathogenic components of processed foods, and long-term consumption of dietary AGEs triggers disruption of the intestinal barrier integrity and increases the risk of chronic diseases. Galactooligosaccharides (GOS) as prebiotics can modulate the intestinal microbiota and improve the intestinal barrier integrity. In this study, we aimed to investigate whether GOS could ameliorate the intestinal barrier damage induced by AGEs. The results showed an increased number of goblet cells (AGEs vs. H-GOS, 133.4 vs. 174.7, p < 0.05) and neutral mucin area (PAS positive area, 7.29% vs. 10.05%, p < 0.05). Upregulated expressions of occludin and claudin-1 and improved intestinal barrier integrity were observed in the H-GOS group. Using 16S rRNA sequencing analysis, we found that GOS significantly reduced the high enrichment of Akkermansia (16.95% vs. 1.29%, p < 0.05) induced by dietary AGEs while increasing the content of short-chain fatty acids. Fecal microbiota transplantation (FMT) showed that AGE-induced damage to the intestinal mucus barrier was reversed in the H-GOS transplanted group. Collectively, GOS ameliorated dietary AGE-induced intestinal barrier damage by reversing the dysregulated state of the intestinal microbiota. Our study lays the foundation for further research on dietary guidelines for populations with high AGE diets.


Assuntos
Produtos Finais da Glicação Avançada em Alimentos , Microbioma Gastrointestinal , Animais , Camundongos , RNA Ribossômico 16S , Oligossacarídeos/farmacologia , Camundongos Endogâmicos C57BL
5.
Food Funct ; 13(22): 11664-11675, 2022 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-36278802

RESUMO

Non-alcoholic fatty liver disease (NAFLD) has become a public health burden. Controlling bile acids (BAs) metabolism and energy expenditure are  potential therapies for NAFLD. Because one of the main health effects of cereal ß-glucan (BG) is its ability to lower cholesterol by interacting with BAs, BG may regulate imbalances of the metabolism of BAs during NAFLD. Therefore, by using metabolic tests coupled with the profiling of hepatic BAs, we have assessed the effect of BG from highland barley on western diet (WD) induced NAFLD mice. BG treatment prevented fat accumulation and increased adipose lipolysis. These moderating effects were associated with an increased energy expenditure. Moreover, BG-treated mice enhanced the production of hepatic BAs, which may be connected with the activation of farnesoid X receptor (FXR) signaling in the liver and inhibition of FXR signaling in the ileum. Our results suggest that BG prevents fat accumulation by increasing energy expenditure, a mechanism associated with major changes in the composition of hepatic BAs.


Assuntos
Hordeum , Hepatopatia Gordurosa não Alcoólica , beta-Glucanas , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ácidos e Sais Biliares/metabolismo , beta-Glucanas/farmacologia , beta-Glucanas/metabolismo , Dieta Ocidental/efeitos adversos , Metabolismo dos Lipídeos , Fígado/metabolismo , Metabolismo Energético , Camundongos Endogâmicos C57BL
6.
Front Nutr ; 9: 916271, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35845787

RESUMO

This study aims to explore the molecular mechanisms of Lycium barbarum polysaccharide (LBP) in alleviating type 2 diabetes through intestinal flora modulation. A high-fat diet (HFD) combined with streptozotocin (STZ) was applied to create a diabetic model. The results indicated that LBP effectively alleviated the symptoms of hyperglycemia, hyperlipidemia, and insulin resistance in diabetic mice. A high dosage of LBP exerted better hypoglycemic effects than low and medium dosages. In diabetic mice, LBP significantly boosted the activities of CAT, SOD, and GSH-Px and reduced inflammation. The analysis of 16S rDNA disclosed that LBP notably improved the composition of intestinal flora, increasing the relative abundance of Bacteroides, Ruminococcaceae_UCG-014, Intestinimonas, Mucispirillum, Ruminococcaceae_UCG-009 and decreasing the relative abundance of Allobaculum, Dubosiella, Romboutsia. LBP significantly improved the production of short-chain fatty acids (SCFAs) in diabetic mice, which corresponded to the increase in the beneficial genus. According to Spearman's correlation analysis, Cetobacterium, Streptococcus, Ralstonia. Cetobacterium, Ruminiclostridium, and Bifidobacterium correlated positively with insulin, whereas Cetobacterium, Millionella, Clostridium_sensu_stricto_1, Streptococcus, and Ruminococcaceae_UCG_009 correlated negatively with HOMA-IR, HDL-C, ALT, AST, TC, and lipopolysaccharide (LPS). These findings suggested that the mentioned genus may be beneficial to diabetic mice's hypoglycemia and hypolipidemia. The up-regulation of peptide YY (PYY), glucagon-like peptide-1 (GLP-1), and insulin were remarkably reversed by LBP in diabetic mice. The real-time PCR (RT-PCR) analysis illustrated that LBP distinctly regulated the glucose metabolism of diabetic mice by activating the IRS/PI3K/Akt signal pathway. These results indicated that LBP effectively alleviated the hyperglycemia and hyperlipidemia of diabetic mice by modulating intestinal flora.

8.
J Agric Food Chem ; 69(32): 9287-9298, 2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34347479

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is characterized by marked imbalances in lipid storage and metabolism. Because the beneficial health effects of cereal ß-glucan (BG) include lowering cholesterol and regulating lipid metabolism, BG may alleviate the imbalances in lipid metabolism observed during NAFLD. The aim of our study was to investigate whether BG from highland barley has an effect on western diet-induced NAFLD in mice. Using lipidomics, we investigated the underlying mechanisms of BG intervention, and identified potential lipid biomarkers. The results reveal that BG (300 mg/kg body weight) significantly alleviated liver steatosis. Lipidomics analysis demonstrated that BG also altered lipid metabolic patterns. We were able to identify 13 differentially regulated lipid species that may be useful as lipid biomarkers. Several genes in the hepatic lipid and cholesterol metabolism pathways were also modulated. These findings provide evidence that BG ameliorates NAFLD by altering liver lipid metabolites and regulating lipid metabolism-related genes.


Assuntos
Hordeum , Hepatopatia Gordurosa não Alcoólica , beta-Glucanas , Animais , Dieta Hiperlipídica/efeitos adversos , Dieta Ocidental , Modelos Animais de Doenças , Hordeum/genética , Metabolismo dos Lipídeos , Lipidômica , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , beta-Glucanas/metabolismo
9.
Mol Nutr Food Res ; 65(8): e2000745, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33629483

RESUMO

SCOPE: Gut barrier dysfunction and inflammation originating from a dysbiotic gut microbiota (GM) are strongly associated with a high-fat diet (HFD). Anthocyanins from Lycium ruthenicum (ACs) show antiobesity effects through modulating the GM. However, the mechanism linking the antiobesity effects of ACs and GM modulation remains obscure. METHODS AND RESULTS: To investigate the ameliorative effects of ACs on colonic barrier dysfunction and inflammation, mice are fed an HFD with or without ACs at doses of 50, 100, and 200 mg kg-1 for 12 weeks. AC supplementation reduced weight gain, enriched short-chain fatty acid (SCFA)-producing bacteria (e.g., Ruminococcaceae, Muribaculaceae, Akkermansia, Ruminococcaceae_UCG-014, and Bacteroides) and SCFA content, depleted endotoxin-producing bacteria (e.g., Helicobacter and Desulfovibrionaceae), and decreased endotoxin (i.e., lipopolysaccharide) levels. SCFAs substantially activated G protein-coupled receptors (GPRs), inhibited histone deacetylases (HDAC), increased intestinal tight junction mRNA and protein expression levels, reduced intestinal permeability, and protected intestinal barrier integrity in HFD-induced mice. These effects mitigate intestinal inflammation by inhibiting the LPS/NF-κB/TLR4 pathway. CONCLUSION: These data indicates that ACs can mitigate colonic barrier dysfunction and inflammation, induce SCFA production and inhibit endotoxin production by modulating the GM in HFD-fed mice. This finding provides a clue for understanding the antiobesity effects of ACs.


Assuntos
Antocianinas/farmacologia , Colite/tratamento farmacológico , Dieta Hiperlipídica/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Lycium/química , Animais , Antocianinas/análise , Antocianinas/química , Colite/etiologia , Colite/microbiologia , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Obesidade/etiologia , Obesidade/prevenção & controle , Receptor 4 Toll-Like/metabolismo
10.
Food Funct ; 11(7): 5749-5767, 2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32602874

RESUMO

Resistant starch (RS) is well known to prevent type 2 diabetes mellitus (T2DM) and obesity. Recently, attention has been paid to gut microbiota which mediates the RS's impact on T2DM and obesity, while a mechanistic understanding of how RS prevents T2DM and obesity through gut microbiota is not clear yet. Therefore, this review aims at exploring the underlying mechanisms of it. RS prevents T2DM and obesity through gut microbiota by modifying selective microbial composition to produce starch-degrading enzymes, promoting the production of intestinal metabolites, and improving gut barrier function. Therefore, RS possessing good functional features can be used to increase the fiber content of healthier food. Furthermore, achieving highly selective effects on gut microbiota based on the slight differences of RS's chemical structure and focusing on the effects of RS on strain-levels are essential to manipulate the microbiota for human health.


Assuntos
Diabetes Mellitus Tipo 2/prevenção & controle , Obesidade/prevenção & controle , Amido Resistente/administração & dosagem , Diabetes Mellitus Tipo 2/microbiologia , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal , Humanos , Inflamação/microbiologia , Inflamação/prevenção & controle , Resistência à Insulina , Obesidade/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA