Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Nutr Biochem ; 94: 108751, 2021 08.
Article in English | MEDLINE | ID: mdl-33915261

ABSTRACT

Diets rich in mono or polyunsaturated fats have been associated with a healthy phenotype, but there is controversial evidence about coconut oil (CO), which is rich in saturated medium-chain fatty acids. Therefore, the purpose of the present work was to study whether different types of oils rich in polyunsaturated (soybean oil, SO), monounsaturated (olive oil, OO), or saturated fatty acids (coconut oil, CO) can regulate the gut microbiota, insulin sensitivity, inflammation, mitochondrial function in wild type and PPARα KO mice. The group that received SO showed the highest microbial diversity, increase in Akkermansia muciniphila, high insulin sensitivity and low grade inflammation, The OO group showed similar insulin sensitivity and insulin signaling than SO, increase in Bifidobacterium, increase in fatty acid oxidation and low grade inflammation. The CO consumption led to the lowest bacterial diversity, a 9-fold increase in the LPS concentration leading to metabolic endotoxemia, hepatic steatosis, increased lipogenesis, highest LDL-cholesterol concentration and the lowest respiratory capacity and fatty acid oxidation in the mitochondria. The absence of PPARα decreased alpha diversity and increased LPS concentration particularly in the CO group, and increased insulin sensitivity in the groups fed SO or OO. These results indicate that consuming mono or polyunsaturated fatty acids produced health benefits at the recommended intake but a high concentration of oils (three times the recommended oil intake in rodents) significantly decreased the microbial alpha-diversity independent of the type of oil.


Subject(s)
Coconut Oil/pharmacology , Gastrointestinal Microbiome/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Olive Oil/pharmacology , PPAR alpha/metabolism , Soybean Oil/pharmacology , Animals , Bacteria/classification , Bacteria/genetics , Cells, Cultured , Computational Biology , DNA, Bacterial/genetics , Feces/chemistry , Gene Expression Regulation/drug effects , Genotype , Glucose Intolerance , Hepatocytes/drug effects , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Oxygen Consumption/drug effects , PPAR alpha/genetics , RNA, Bacterial/genetics , RNA, Ribosomal, 16S , Random Allocation , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
2.
Nutrients ; 12(9)2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32971775

ABSTRACT

BACKGROUND: Magnesium is a mineral that modulates several physiological processes. However, its relationship with intestinal microbiota has been scarcely studied. Therefore, this study aimed to assess the role of dietary magnesium content to modulate the intestinal microbiota of Wistar male rats. METHODS: Rats were randomly assigned one of three diets: a control diet (C-Mg; 1000 mg/kg), a low magnesium content diet (L-Mg; 60 mg/kg), and a high magnesium content diet (H-Mg; 6000 mg/kg), for two weeks. After treatment, fecal samples were collected. Microbiota composition was assessed by sequencing the V3-V4 hypervariable region. RESULTS: The C-Mg and L-Mg groups had more diversity than H-Mg group. CF231, SMB53, Dorea, Lactobacillus and Turibacter were enriched in the L-Mg group. In contrast, the phyla Proteobacteria, Parabacteroides, Butyricimonas, and Victivallis were overrepresented in the H-Mg group. PICRUSt analysis indicated that fecal microbiota of the L-Mg group were encoded with an increased abundance of metabolic pathways involving carbohydrate metabolism and butanoate metabolism. CONCLUSION: Dietary magnesium supplementation can result in intestinal dysbiosis development in a situation where there is no magnesium deficiency. Conversely, low dietary magnesium consumption is associated with microbiota with a higher capacity to harvest energy from the diet.


Subject(s)
Diet , Gastrointestinal Microbiome/drug effects , Magnesium/administration & dosage , Animals , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Bacterial Load , Bacteroidetes/isolation & purification , Butyric Acid/metabolism , Carbohydrate Metabolism , Dietary Supplements/adverse effects , Dysbiosis/chemically induced , Feces/microbiology , Firmicutes/isolation & purification , Magnesium/adverse effects , Magnesium Deficiency/microbiology , Male , Proteobacteria/isolation & purification , Rats , Rats, Wistar
3.
Clin Exp Rheumatol ; 38 Suppl 126(4): 34-39, 2020.
Article in English | MEDLINE | ID: mdl-32896265

ABSTRACT

OBJECTIVES: Lipid mediators derived from polyunsaturated fatty acids (FA), have been related to inflammation and immune response regulation. Herein we evaluated the intake and serum levels of ω-3 and ω-6 FA among patients with primary Sjögren's syndrome (pSS), and correlated with ocular/oral sicca symptoms, disease activity and a panel of chemokines/cytokines. METHODS: We included 108 patients and 100 controls. Dietary information was obtained from a food questionnaire of one-day reminder and processed using a nutritional software. Among the SS group, we measured serum ω-3 (α-linolenic acid [α-LN], eicosapentaenoic acid [EPA], docosahexaenoic acid [DHA]) and ω-6 (linoleic acid [LA], arachidonic acid [AA]) by gas chromatography flame ionization. We scored the ESSDAI, ESPRI, Schirmer-I test and NSWSF. In a subsample, we assessed the OSDI, ophthalmologic staining scores and measured CXCL8, CXCL10, CCL2, IL-22 and IL-21 in saliva, and CXCL8, CXCL10, CCL2 and CXCL9 in tears by Luminometry. RESULTS: ω-3 and ω-6 intake was lower in SS patients than controls, and did not correlate with serum levels. We found a negative correlation between α-LN and the OSDI and ESSDAI, as well as DHA and ESSDAI. In tears, AA positively correlated with CXCL9, whereas in saliva, α-LN, DHA and the ω3 sum negatively correlated with CCL2. We observed a negative correlation between the ω6 sum and IL-21. CONCLUSIONS: pSS patients had deficient omega intake. Lower ocular symptoms, ESSDAI scores and salivary CCL2 correlated with higher ω-3 levels, possible suggesting a role in chronic inflammation. Further studies are warranted to deepen in the knowledge of this association.


Subject(s)
Fatty Acids, Omega-3 , Sjogren's Syndrome , Docosahexaenoic Acids , Fatty Acids, Omega-6 , Humans , Inflammation , Sjogren's Syndrome/diagnosis
4.
Int J Mol Sci ; 21(15)2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32752280

ABSTRACT

Goat's milk is a rich source of bioactive compounds (peptides, conjugated linoleic acid, short chain fatty acids, monounsaturated and polyunsaturated fatty acids, polyphenols such as phytoestrogens and minerals among others) that exert important health benefits. However, goat's milk composition depends on the type of food provided to the animal and thus, the abundance of bioactive compounds in milk depends on the dietary sources of the goat feed. The metabolic impact of goat milk rich in bioactive compounds during metabolic challenges such as a high-fat (HF) diet has not been explored. Thus, we evaluated the effect of milk from goats fed a conventional diet, a conventional diet supplemented with 30% Acacia farnesiana (AF) pods or grazing on metabolic alterations in mice fed a HF diet. Interestingly, the incorporation of goat's milk in the diet decreased body weight and body fat mass, improved glucose tolerance, prevented adipose tissue hypertrophy and hepatic steatosis in mice fed a HF diet. These effects were associated with an increase in energy expenditure, augmented oxidative fibers in skeletal muscle, and reduced inflammatory markers. Consequently, goat's milk can be considered a non-pharmacologic strategy to improve the metabolic alterations induced by a HF diet. Using the body surface area normalization method gave a conversion equivalent daily human intake dose of 1.4 to 2.8 glasses (250 mL per glass/day) of fresh goat milk for an adult of 60 kg, which can be used as reference for future clinical studies.


Subject(s)
Energy Metabolism/drug effects , Fatty Acids/administration & dosage , Fatty Liver/prevention & control , Milk/chemistry , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Obesity/prevention & control , Animals , Biomarkers/analysis , Diet, High-Fat/adverse effects , Dietary Supplements , Fatty Liver/etiology , Gene Expression/drug effects , Goats , Insulin Resistance , Linoleic Acids, Conjugated/administration & dosage , Male , Mice, Inbred C57BL , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Obesity/etiology
5.
Mol Nutr Food Res ; 64(17): e2000532, 2020 09.
Article in English | MEDLINE | ID: mdl-32729948

ABSTRACT

SCOPE: Soy protein is a high-quality protein and its consumption has been associated with a reduction of serum cholesterol and triglycerides and an improvement in insulin resistance. However, it is not known whether the effects of soy protein are mediated by the gut microbiota. Thus, the aim of this study is to assess whether using antibiotics to partially eradicate the gut microbiota can prevent the beneficial effects of soy protein in rats. METHODS AND RESULTS: Thus, rats are fed one of the following diets for 16 weeks: casein control, soy protein control, high-fat casein, and high-fat soy protein. The rats are then treated for 4 weeks with antibiotics. Body weight and composition, energy expenditure, glucose tolerance test, metabolic endotoxemia, and gut microbiota are measured before and after treatment with antibiotic. The results show that soy protein consumption decreases weight gain, body fat, metabolic endotoxemia, and increases energy expenditure and glucose tolerance. Antibiotic treatment suppresses all these metabolic effects. These changes are accompanied by modifying the diversity and taxonomy of the gut microbiota. CONCLUSION: In conclusion, the evidence suggests that the health benefits of soy protein are partly dependent of the gut microbiota.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Soybean Proteins/pharmacology , Adipose Tissue/drug effects , Ampicillin/adverse effects , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/adverse effects , Biomarkers/metabolism , Body Composition/drug effects , Caseins/pharmacology , Diet, High-Fat/adverse effects , Endotoxemia/chemically induced , Energy Metabolism/drug effects , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Inflammation/genetics , Inflammation/metabolism , Male , Neomycin/adverse effects , Neomycin/pharmacology , Rats, Wistar , Weight Gain/drug effects
7.
Nutrients ; 12(4)2020 Apr 23.
Article in English | MEDLINE | ID: mdl-32340138

ABSTRACT

There is limited information on the effect of black beans (BB) as a source of protein and resistant starch on the intestinal microbiota. The purpose of the present work was to study the effect of cooked black beans with and without high fat and sugar (HF + S) in the diet on body composition, energy expenditure, gut microbiota, short-chain fatty acids, NF-κB, occluding and insulin signaling in a rat model and the area under the curve for glucose, insulin and incretins in healthy subjects. The consumption of BB reduced the percentage of body fat, the area under the curve of glucose, serum leptin, LPS, glucose and insulin concentrations and increased energy expenditure even in the presence of HF + S. These results could be mediated in part by modification of the gut microbiota, by increasing a cluster of bacteria in the Clostridia class, mainly R. bromii, C. eutactus, R. callidus, R. flavefaciens and B. pullicaecorum and by an increase in the concentration of fecal butyrate. In conclusion, the consumption of BB can be recommended to prevent insulin resistance and metabolic endotoxemia by modifying the gut microbiota. Finally, the groups fed BB showed lower abundance of hepatic FMO-3, even with a high-fat diet protecting against the production of TMAO and obesity.


Subject(s)
Clostridiales , Dietary Supplements , Fabaceae , Gastrointestinal Microbiome , Insulin Resistance , Animals , Body Fat Distribution , Butyrates/metabolism , Endotoxemia/prevention & control , Energy Metabolism , Glucose/metabolism , Healthy Volunteers , Leptin/metabolism , Liver/metabolism , Male , Models, Animal , Oxygenases/metabolism , Rats, Wistar , Spondylitis, Ankylosing/microbiology
8.
PLoS One ; 11(7): e0159631, 2016.
Article in English | MEDLINE | ID: mdl-27438015

ABSTRACT

Edible and medicinal mushrooms contain bioactive compounds with promising effects on several cardiovascular risk biomarkers. However, strains of Ganoderma lucidum of Mexican origin have not yet been studied. Standardized extracts of G. lucidum (Gl) were given to C57BL/6 mice fed a high-cholesterol diet compared with the drug simvastatin. The effects of the extracts on serum biochemical parameters, liver lipid content, cholesterol metabolism, and the composition of gut microbiota were assessed. Acetylsalicylic acid (10 mM) added to the cultivation substrate modulated properties of Gl extracts obtained from mature basidiomata. Compared to the high-cholesterol diet group, the consumption of Gl extracts significantly reduced total serum cholesterol (by 19.2% to 27.1%), LDL-C (by 4.5% to 35.1%), triglyceride concentration (by 16.3% to 46.6%), hepatic cholesterol (by 28.7% to 52%) and hepatic triglycerides (by 43.8% to 56.6%). These effects were associated with a significant reduction in the expression of lipogenic genes (Hmgcr, Srebp1c, Fasn, and Acaca) and genes involved in reverse cholesterol transport (Abcg5 and Abcg8), as well as an increase in Ldlr gene expression in the liver. No significant changes were observed in the gene expression of Srebp2, Abca1 or Cyp7a1. In several cases, Gl-1 or Gl-2 extracts showed better effects on lipid metabolism than the drug simvastatin. A proposed mechanism of action for the reduction in cholesterol levels is mediated by α-glucans and ß-glucans from Gl, which promoted decreased absorption of cholesterol in the gut, as well as greater excretion of fecal bile acids and cholesterol. The prebiotic effects of Gl-1 and Gl-2 extracts modulated the composition of gut microbiota and produced an increase in the Lactobacillaceae family and Lactobacillus genus level compared to the control group, high-cholesterol diet group and group supplemented with simvastatin. Mexican genetic resources of Gl represent a new source of bioactive compounds showing hypocholesterolemic properties and prebiotic effects.


Subject(s)
Cardiovascular Diseases/drug therapy , Cholesterol/blood , Hypercholesterolemia/drug therapy , Plant Extracts/administration & dosage , Prebiotics/administration & dosage , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/chemistry , Cardiovascular Diseases/pathology , Dietary Supplements , Humans , Hypercholesterolemia/blood , Hypercholesterolemia/pathology , Lactobacillus/chemistry , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Plant Extracts/chemistry , Reishi/chemistry , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL