Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Food Sci ; 89(4): 2465-2481, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38380680

RESUMO

Camellia seed oil (CO) has high nutritional value and multiple bioactivities. However, the specific anti-fatigue characteristics and the implied mechanism of CO have not yet been fully elucidated. Throughout this investigation, male C57BL/6J mice, aged 8 weeks, underwent exhaustive exercise with or without CO pretreatment (2, 4, and 6 mL/kg BW) for 28 days. CO could extend the rota-rod and running time, reduce blood urea nitrogen levels and serum lactic acid, and increase muscle and hepatic glycogen, adenosine triphosphate, and anti-oxidative indicators. Additionally, CO could upregulate the mRNA and Nrf2 protein expression levels, as well as enhance the levels of its downstream antioxidant enzymes and induce the myofiber-type transformation from fast to slow and attenuate the gut mechanical barrier. Moreover, CO could ameliorate gut dysbiosis by reducing Firmicutes to Bacteroidetes ratio at the phylum level, increasing the percentage of Alistipes, Alloprevotella, Lactobacillus, and Muribaculaceae, and decreasing the proportion of Dubosiella at the genus level. In addition, specific bacterial taxa, which were altered by CO, showed a significant correlation with partial fatigue-related parameters. These findings suggest that CO may alleviate fatigue by regulating antioxidant capacity, muscle fiber transformation, gut mechanical barrier, and gut microbial composition in mice. PRACTICAL APPLICATION: Our study revealed that camellia seed oil (CO) could ameliorate exercise-induced fatigue in mice by modulating antioxidant capacity, muscle fiber, and gut microbial composition in mice. Our results promote the application of CO as an anti-fatigue functional food that targets oxidative stress, myofiber-type transformation, and microbial community.


Assuntos
Camellia , Microbioma Gastrointestinal , Camundongos , Masculino , Animais , Antioxidantes/farmacologia , Microbioma Gastrointestinal/genética , Camundongos Endogâmicos C57BL , Fadiga/tratamento farmacológico , Fadiga/metabolismo , Óleos de Plantas/farmacologia , Bacteroidetes , Firmicutes , Fibras Musculares Esqueléticas
2.
Plant Foods Hum Nutr ; 79(1): 98-105, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38085440

RESUMO

A water-soluble polysaccharide (EP) was purified from edible algae Enteromorpha prolifera. Gel permeation chromatography (GPC), ion chromatography (IC), and fourier transform infrared (FT-IR) were performed to characterize its structure. EP was defined as a low molecular weight (6625 Da) composed of rhamnose, glucose, glucuronic acid, xylose, galactose, arabinose, and mannose. Moreover, it was a sulfated polysaccharide with a degree of substitution (DS) of 1.48. Then, the high-fat diet/streptozotocin (HFD/STZ) induced diabetic mouse model was established to support evidence for a novel hypoglycemic mechanism. Results showed that blood glucose (47.32%), liver index (7.65%), epididymal fat index (16.86%), serum total cholesterol (26.78%) and triglyceride (37.61%) in the high-dose EP (HEP) group were significantly lower than those in the HFD group. Noticeably, the content of liver glycogen in the HEP group was significantly higher (62.62%) than that in the HFD group, indicating the promotion of glycogen synthesis. These beneficial effects were attributed to significantly increased protein kinase B (AKT) phosphorylation and its downstream signaling response. Further studies showed that diabetic mice exhibited excessive O-GlcNAcylation level and high expression of O-linked ß-D-N-acetylglucosamine transferase (OGT), which were decreased by 62.21 and 30.43% in the HEP group. This result suggested that EP had a similar effect to OGT inhibitors, which restored AKT phosphorylation and prevented pathoglycemia. This work reveals a novel hypoglycemic mechanism of EP, providing a theoretical basis for further studies on its pharmacological properties in improvement of T2DM.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Algas Comestíveis , Ulva , Animais , Camundongos , Diabetes Mellitus Tipo 2/prevenção & controle , Proteínas Proto-Oncogênicas c-akt , Sulfatos , Diabetes Mellitus Experimental/tratamento farmacológico , Espectroscopia de Infravermelho com Transformada de Fourier , Hipoglicemiantes/farmacologia , Polissacarídeos/farmacologia
3.
Food Funct ; 14(3): 1685-1698, 2023 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-36692407

RESUMO

Although conjugated linoleic acid (CLA) has been shown to have anti-obesity properties, the effect and mechanism of CLA in alleviating glycolipid metabolism disorders remains unclear. In this work, it was observed that rats fed a high-fat diet (HFD) had lower body weight and body fat levels after 9 weeks of low-dose and high-dose CLA interventions. The results of blood biochemical indices showed that CLA significantly reduced the levels of total cholesterol, triglycerides, fasting blood glucose and insulin. Additionally, high-dose CLA could restore the intestinal microbiota composition, including increasing the relative abundances of short-chain fatty acid (SCFA)-producing microbiota, such as Dubosiella, Faecalibaculum and Bifidobacterium; decreasing the relative abundances of Enterococcus and Ruminococcus_2; and increasing the content of SCFAs in feces and serum. Further analysis showed that high-dose CLA could increase the expression levels of Insr, Irs-2, Akt and Glut4 in the liver tissue of HFD-induced obese rats. Consistently, high dose of CLA could reversibly improve the downregulation of INSR, AKT, PI3K and GLUT4 protein expression caused by HFD and reverse the decline in AKT phosphorylation levels. Correlation clustering analysis with a heatmap showed that the changes in specific microbiota induced by high-dose CLA were correlated with changes in obesity-related indices and gene expression. The molecular docking analysis showed that the molecular docking of SCFAs with the IRS-2, AKT and GLUT4 proteins had high linking activity. The results supported that CLA can alleviate glycolipid metabolic imbalances associated with obesity by altering the intestinal microbiota to induce the production of SCFAs and thereby activate the INSR/IRS-2/AKT/GLUT4 pathway. This study supports CLA may be preferentially used by the intestinal microbiota of the host to promote its health.


Assuntos
Microbioma Gastrointestinal , Ácidos Linoleicos Conjugados , Doenças Metabólicas , Ratos , Animais , Ácidos Linoleicos Conjugados/química , Glicolipídeos , Proteínas Proto-Oncogênicas c-akt , Simulação de Acoplamento Molecular , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Ácidos Graxos Voláteis
4.
Food Funct ; 11(5): 4485-4498, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32378684

RESUMO

To investigate whether polysaccharides from Enteromorpha prolifera (EPP) could protect against acute hepatic injury induced by CCl4, ICR mice were pretreated with EPP (150, 300, and 450 mg kg-1) and silymarin (100 mg kg-1) for 28 days before CCl4 induction. Pretreatment with EPP attenuated CCl4-induced elevated serum transaminase activities and histopathological alterations in the liver. In addition, EPP prevented CCl4-induced reduction of protein levels of phosphorylated nuclear factor E2-related factor 2 (p-Nrf2)/Nrf2, heme oxygenase-1 (HO-1), and mRNA levels of NADPH quinineoxidoreductase-1 (NQO-1), which, in turn, reduced hepatic oxidative stress injury. Furthermore, the hepatic protein levels of inflammatory mediators and the phosphorylation of nuclear factor-kappaB p65 (NF-κB p65) and I kappaB alpha (IκBα), and the mRNA levels of Toll-like receptor 2 (TLR2), TLR4, and prolyl-isomerase-1 (Pin-1) in the inflammatory signaling pathway were recovered in the EPP pretreated groups. Moreover, EPP prevented the hepatocellular apoptotic changes with inhibition of B-cell lymphoma 2 (Bcl-2), and the induction of Bcl-2-associated X (Bax) and Cleaved caspase-3 caused by CCl4. Taken together, these results indicated that EPP protected against hepatic injury induced by CCl4-derived reactive intermediates through the activation of Nrf2/HO-1 signaling, and suppression of oxidative stress, inflammation and apoptosis.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Clorófitas , Polissacarídeos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Tetracloreto de Carbono , Modelos Animais de Doenças , Heme Oxigenase-1/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Polissacarídeos/uso terapêutico , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos
5.
Mol Nutr Food Res ; 59(8): 1491-503, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25944715

RESUMO

SCOPE: Ursolic acid (UA) is a triterpenoid compound with multifold biological functions. Our previous studies have reported that UA protects against high-fat diet-induced obesity and improves insulin resistance (IR). However, the potential mechanisms are still undefined. Free fatty acid (FFA) metabolism in skeletal muscle plays a central role in obesity and IR. Therefore, in this study, we investigated the effect and the potential mechanisms of UA on skeletal muscle FFA metabolism. METHODS AND RESULTS: In diet-induced obese rats, 0.5% UA supplementation for 6 weeks markedly reduced body weight, increased energy expenditure, decreased FFA level in serum and skeletal muscle and triglyceride content in skeletal muscle. In vitro, the data provided directly evidence that UA significantly increased fluorescently labeled FFA uptake and (3) H-labeled palmitic acid ß-oxidation. UA-activated AMP-activated protein kinase (AMPK) and downstream targets were involved in the increase of FFA catabolism. Moreover, upregulated uncoupling protein 3 (UCP3) by UA contributed to AMPK activation via elevating adenosine monophosphate/adenosine triphosphate ratio. CONCLUSION: UA increases FFA burning through enhancing skeletal muscle FFA uptake and ß-oxidation via an UCP3/AMPK-dependent pathway, which provides a novel perspective on the biological function of UA against obesity and IR.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Suplementos Nutricionais , Metabolismo Energético , Ácidos Graxos não Esterificados/metabolismo , Canais Iônicos/agonistas , Proteínas Mitocondriais/agonistas , Músculo Esquelético/metabolismo , Triterpenos/uso terapêutico , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Absorção Fisiológica , Animais , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos não Esterificados/sangue , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Camundongos , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Músculo Esquelético/enzimologia , Obesidade/sangue , Obesidade/dietoterapia , Obesidade/etiologia , Obesidade/metabolismo , Interferência de RNA , Distribuição Aleatória , Ratos Sprague-Dawley , Sistemas do Segundo Mensageiro , Organismos Livres de Patógenos Específicos , Proteína Desacopladora 3 , Ácido Ursólico
6.
World J Gastroenterol ; 20(47): 17932-40, 2014 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-25548491

RESUMO

AIM: To study the prevalence and clinical biochemical, blood cell and metabolic features of lean-non-alcoholic fatty liver disease (lean-NAFLD) and its association with other diseases. METHODS: Demographic, biochemical and blood examinations were conducted in all the subjects in this study. We classified the subjects into four groups according to their weight and NAFLD status: lean-control, lean-NAFLD [body mass index (BMI) < 24 kg/m(2)], overweight-obese control and overweight-obese NAFLD. One-way analysis of variance (ANOVA) was used to compare the means of continuous variables (age, BMI, blood pressure, glucose, lipid, insulin, liver enzymes and blood cell counts) and the χ (2) test was used to compare the differences in frequency of categorical variables (sex, education, physical activity, smoking, alcohol consumption and prevalence of hypertension, hyperlipidemia, diabetes, metabolic syndrome central obesity and obesity). Both univariate and multivariate logistic regression models were adopted to calculate odds ratios (ORs) and predict hyperlipidemia, hypertension, diabetes and metabolic syndrome when we respectively set all controls, lean-control and overweight-obese-control as references. In multivariate logistic regression models, we adjusted potential confounding factors, including age, sex, smoking, alcohol consumption and physical activity. RESULTS: The prevalence of NAFLD was very high in China. NAFLD patients were older, had a higher BMI, waist circumference, blood pressure, fasting blood glucose, insulin, blood lipid, liver enzymes and uric acid than the controls. Although lean-NAFLD patients had lower BMI and waist circumstance, they had significantly higher visceral adiposity index than overweight-obese controls. Lean-NAFLD patients had comparable triglyceride, cholesterin and low-density lipoprotein cholesterin to overweight-obese NAFLD patients. In blood cell examination, both lean and overweight-obese NAFLD was companied by higher white blood cell count, red blood cell count, hemoglobin and hematocrit value. All NAFLD patients were at risk of hyperlipidemia, hypertension, diabetes and metabolic syndrome (MetS). Lean-NAFLD was more strongly associated with diabetes (OR = 2.47, 95%CI: 1.14-5.35), hypertension (OR = 1.72, 95%CI: 1.00-2.96) and MetS (OR = 3.19, 95%CI: 1.17-4.05) than overweight-obese-NAFLD (only OR for MetS was meaningful: OR = 1.89, 95%CI: 1.29-2.77). NAFLD patients were more likely to have central obesity (OR = 1.97, 95%CI: 1.38-2.80), especially in lean groups (OR = 2.17, 95%CI: 1.17-4.05). CONCLUSION: Lean-NAFLD has unique results in demographic, biochemical and blood examinations, and adds significant risk for diabetes, hypertension and MetS in lean individuals.


Assuntos
Povo Asiático , Peso Corporal Ideal/etnologia , Síndrome Metabólica/etnologia , Hepatopatia Gordurosa não Alcoólica/etnologia , Magreza/etnologia , Adulto , Idoso , Biomarcadores/sangue , Índice de Massa Corporal , Distribuição de Qui-Quadrado , China/epidemiologia , Diabetes Mellitus/etnologia , Dislipidemias/etnologia , Feminino , Humanos , Hipertensão/etnologia , Modelos Logísticos , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/diagnóstico , Pessoa de Meia-Idade , Análise Multivariada , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Obesidade/etnologia , Razão de Chances , Prevalência , Fatores de Risco , Magreza/sangue , Magreza/diagnóstico , Adulto Jovem
7.
PLoS One ; 8(5): e63770, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691094

RESUMO

BACKGROUND: Nuciferine is a major active aporphine alkaloid from the leaves of N. nucifera Gaertn that possesses anti-hyperlipidemia, anti-hypotensive, anti-arrhythmic, and insulin secretagogue activities. However, it is currently unknown whether nuciferine can benefit hepatic lipid metabolism. METHODOLOGY/PRINCIPAL FINDINGS: In the current study, male golden hamsters were randomly divided into four groups fed a normal diet, a high-fat diet (HFD), or a HFD supplemented with nuciferine (10 and 15 mg/kg·BW/day). After 8 weeks of intervention, HFD-induced increases in liver and visceral adipose tissue weight, dyslipidemia, liver steatosis, and mild necroinflammation in hamsters were analyzed. Nuciferine supplementation protected against HFD-induced changes, alleviated necroinflammation, and reversed serum markers of metabolic syndrome in hamsters fed a HFD. RT-PCR and western blot analyses revealed that hamsters fed a HFD had up-regulated levels of genes related to lipogenesis, increased free fatty acid infiltration, and down-regulated genes involved in lipolysis and very low density lipoprotein secretion. In addition, gene expression of cytochrome P4502E1 and tumor necrosis factor-α were also increased in the HFD group. Nuciferine supplementation clearly suppressed HFD-induced alterations in the expression of genes involved in lipid metabolism. CONCLUSIONS/SIGNIFICANCE: Nuciferine supplementation ameliorated HFD-induced dyslipidemia as well as liver steatosis and injury. The beneficial effects of nuciferine were associated with altered expression of hepatic genes involved in lipid metabolism.


Assuntos
Aporfinas/farmacologia , Gorduras na Dieta/efeitos adversos , Fígado Gorduroso/prevenção & controle , Fígado/lesões , Animais , Peso Corporal/efeitos dos fármacos , Cricetinae , Gorduras na Dieta/administração & dosagem , Fígado Gorduroso/etiologia , Comportamento Alimentar/efeitos dos fármacos , Regulação da Expressão Gênica , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Mesocricetus , Tamanho do Órgão/efeitos dos fármacos , Estresse Oxidativo , Reação em Cadeia da Polimerase em Tempo Real , Ferimentos e Lesões/prevenção & controle
8.
Int J Cardiol ; 168(3): 2548-60, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23602294

RESUMO

BACKGROUND: Limited studies have addressed the effects of calcium supplementation (CaS) on serum total cholesterol (TC) in postmenopausal women and the results are inconclusive. Moreover, the potential mechanisms through which CaS regulates cholesterol metabolism in the absence of estrogen are still sealed for the limitation of human being study. METHODS: Cross-sectional survey, animal and in vitro experiments were conducted to investigate the effect of CaS on endogenous cholesterol metabolism in estrogen deficiency and identify its potential mechanisms. Ovariectomized rats were used to mimic estrogen deficiency. In vitro, HepG2 cell line was exposed to estradiol and/or calcium treatment. RESULTS: We demonstrated that CaS significantly increased serum TC and the risk of hypercholesterolemia and myocardial infarction in postmenopausal women. Increased serum TC in estrogen deficiency was caused mainly by decreased cholesterol catabolism rather than increased synthesis. This was mediated by reduced 7α-hydroxylase resulting from increased liver intracellular Ca(2+) concentrations, reduced intracellular basal cAMP and subsequent up-regulation of SREBP-1c and SHP expression. Estrogen had a protective role in preventing CaS-induced TC increase by activating the G-protein coupled estrogen receptor, which mediated the estrogen effect through the transient receptor potential canonical 1 cation channel. CONCLUSIONS: CaS increases endogenous serum TC via decreasing hepatic cholesterol catabolism in estrogen deficiency. G-protein coupled estrogen receptor is shown to be a key target in mediating CaS-induced TC increase. CaS should be monitored for the prevention of serum TC increase during menopause.


Assuntos
Cálcio/administração & dosagem , Colesterol/sangue , Estrogênios/deficiência , Receptores Acoplados a Proteínas G/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Colesterol/metabolismo , Estudos Transversais , Suplementos Nutricionais , Feminino , Ratos , Ratos Wistar
9.
Mol Nutr Food Res ; 55(12): 1809-18, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22038976

RESUMO

SCOPE: Mangiferin, a natural polyphenol, has been shown to have hypolipidemic effect in rat and mouse. However, the mechanism of action is not well understood. This study was conducted to determine the effect and mechanism of action of mangiferin on hyperlipidemia induced in hamsters by a high-fat diet. METHODS AND RESULTS: Forty male hamsters were randomly assigned to normal control, high-fat control, and high fat with mangiferin (50 and 150 mg/kg BW) groups. Mangiferin treatment significantly decreased final body weight, liver weight and visceral fat-pad weight, serum triglyceride (TG) and total free fatty acid (FFA) concentrations, hepatic TG levels and hepatic and muscle total FFA contents. Mangiferin upregulated mRNA expression of peroxisome proliferator-activated receptor-α (PPAR-α), fatty acid translocase (CD36) and carnitine palmitoyltransferase 1 (CPT-1), but downregulated mRNA expression of sterol regulatory element-binding protein 1c (SREBP-1c), acetyl CoA carboxylase (ACC), acyl-CoA:diacylglycerol acyltransferase 2 (DGAT-2) and microsomal triglyceride transfer protein (MTP) in liver. Mangiferin also stimulated mRNA expression of PPAR-α, CD36, CPT-1 and lipoprotein lipase (LPL) in muscle. CONCLUSIONS: The results suggest that mangiferin may ameliorate hypertriglyceridemia partly by modulating the expression levels of genes involved in lipid oxidation and lipogenesis.


Assuntos
Dieta Hiperlipídica , Gorduras na Dieta/administração & dosagem , Hiperlipidemias/tratamento farmacológico , Hipolipemiantes/farmacologia , Xantonas/farmacologia , Acetil-CoA Carboxilase/efeitos dos fármacos , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Antígenos CD36/efeitos dos fármacos , Antígenos CD36/genética , Antígenos CD36/metabolismo , Carnitina O-Palmitoiltransferase/efeitos dos fármacos , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cricetinae , Diacilglicerol O-Aciltransferase/efeitos dos fármacos , Diacilglicerol O-Aciltransferase/genética , Diacilglicerol O-Aciltransferase/metabolismo , Regulação para Baixo , Ácidos Graxos não Esterificados/sangue , Hipertrigliceridemia/tratamento farmacológico , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Lipase Lipoproteica/efeitos dos fármacos , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Tamanho do Órgão/efeitos dos fármacos , PPAR alfa/efeitos dos fármacos , PPAR alfa/genética , PPAR alfa/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/efeitos dos fármacos , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/sangue , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA