Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Br J Nutr ; 128(1): 43-54, 2022 07 14.
Article in English | MEDLINE | ID: mdl-34340727

ABSTRACT

Branched-chain amino acids (BCAA) are considered markers of insulin resistance (IR) in subjects with obesity. In this study, we evaluated whether the presence of the SNP of the branched-chain aminotransferase 2 (BCAT2) gene can modify the effect of a dietary intervention (DI) on the plasma concentration of BCAA in subjects with obesity and IR. A prospective cohort study of adult subjects with obesity, BMI ≥ 30 kg/m2, homeostatic model assessment-insulin resistance (HOMA-IR ≥ 2·5) no diagnosed chronic disease, underwent a DI with an energy restriction of 3140 kJ/d and nutritional education for 1 month. Anthropometric measurements, body composition, blood pressure, resting energy expenditure, oral glucose tolerance test results, serum biochemical parameters and the plasma amino acid profile were evaluated before and after the DI. SNP were assessed by the TaqMan SNP genotyping assay. A total of eighty-two subjects were included, and fifteen subjects with a BCAT2 SNP had a greater reduction in leucine, isoleucine, valine and the sum of BCAA. Those subjects also had a greater reduction in skeletal muscle mass, fat-free mass, total body water, blood pressure, muscle strength and biochemical parameters after 1 month of the DI and adjusting for age and sex. This study demonstrated that the presence of the BCAT2 SNP promotes a greater reduction in plasma BCAA concentration after adjusting for age and sex, in subjects with obesity and IR after a 1-month energy-restricted DI.


Subject(s)
Insulin Resistance , Pregnancy Proteins , Adult , Humans , Prospective Studies , Blood Glucose/metabolism , Amino Acids, Branched-Chain , Obesity/metabolism , Transaminases/genetics , Pregnancy Proteins/genetics , Minor Histocompatibility Antigens
2.
Ann Hum Biol ; 49(7-8): 291-298, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36350847

ABSTRACT

BACKGROUND: Plasminogen activator inhibitor 1 (PAI-1) and resistin are associated with dysfunctional adipose tissue (AT)-related metabolic complications. The role of dietary eicosapentaenoic (EPA) and docosahexaenoic (DHA) fatty acids in this relationship is unknown. AIM: To investigate the association of EPA and DHA with PAI-1 and resistin, as well as the role of this association on the glucose metabolism of apparently healthy subjects. SUBJECTS AND METHODS: Thirty-six healthy individuals were included. Validated food frequency questionnaires were used to analyse dietary habits. Inflammatory and glucose metabolism markers were quantified. Subcutaneous AT samples were obtained, and adipocyte number, area, and macrophage content were assessed. RESULTS: In 36 subjects aged 56 ± 8 years and with a body mass index of 26 ± 4 kg/m2, logEPA, and logDHA showed significant association with logresistin and a marginal association with PAI-1. Adipocyte number, area, and lognumber of macrophages per adipocyte significantly correlated with PAI-1 but not with logresistin. Although logEPA and logDHA were independently associated with loginsulin, loginsulin resistance, and C-Peptide, the addition of logresistin, but not of PAI-1, into the multivariable model, abolished the associations. CONCLUSIONS: EPA and DHA could modulate glucose metabolism across AT functional states. Our data indicate that this association is independent of other metabolic risk factors.


Subject(s)
Fatty Acids, Omega-3 , Plasminogen Activator Inhibitor 1 , Humans , Plasminogen Activator Inhibitor 1/metabolism , Resistin/metabolism , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Self Report , Healthy Volunteers , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/pharmacology , Adipose Tissue/metabolism , Glucose/metabolism
3.
Mol Med ; 27(1): 108, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34525937

ABSTRACT

BACKGROUND: Elevations of circulating branched-chain amino acids (BCAA) are observed in humans with obesity and metabolic comorbidities, such as insulin resistance. Although it has been described that microbial metabolism contributes to the circulating pool of these amino acids, studies are still scarce, particularly in pediatric populations. Thus, we aimed to explore whether in early adolescents, gut microbiome was associated to circulating BCAA and in this way to insulin resistance. METHODS: Shotgun sequencing was performed in DNA from fecal samples of 23 early adolescents (10-12 years old) and amino acid targeted metabolomics analysis was performed by LC-MS/MS in serum samples. By using the HUMAnN2 algorithm we explored microbiome functional profiles to identify whether bacterial metabolism contributed to serum BCAA levels and insulin resistance markers. RESULTS: We identified that abundance of genes encoding bacterial BCAA inward transporters were negatively correlated with circulating BCAA and HOMA-IR (P < 0.01). Interestingly, Faecalibacterium prausnitzii contributed to approximately ~ 70% of bacterial BCAA transporters gene count. Moreover, Faecalibacterium prausnitzii abundance was also negatively correlated with circulating BCAA (P = 0.001) and with HOMA-IR (P = 0.018), after adjusting for age, sex and body adiposity. Finally, the association between Faecalibacterium genus and BCAA levels was replicated over an extended data set (N = 124). CONCLUSIONS: We provide evidence that gut bacterial BCAA transport genes, mainly encoded by Faecalibacterium prausnitzii, are associated with lower circulating BCAA and lower insulin resistance. Based on the later, we propose that the relationship between Faecalibacterium prausnitzii and insulin resistance, could be through modulation of BCAA.


Subject(s)
Amino Acids, Branched-Chain/blood , Faecalibacterium prausnitzii/physiology , Gastrointestinal Microbiome , Adolescent , Age Factors , Amino Acids, Branched-Chain/metabolism , Biomarkers , Body Weights and Measures , Child , Female , Humans , Insulin Resistance , Male , Metabolomics/methods , Metagenome , Metagenomics/methods , Obesity/metabolism , Public Health Surveillance
4.
Eur J Nutr ; 60(5): 2435-2447, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33145643

ABSTRACT

PURPOSE: We compared the effect of diets with different amounts and sources of dietary protein on insulin sensitivity (IS) in subjects with obesity and insulin resistance (IR). METHODS: Eighty subjects with obesity (BMI ≥ 30 kg/m2) and IR (Matsuda index < 4.3 and HOMA-IR ≥ 2.5) over 18 years old were randomized to four groups for a one-month period: a normal protein diet (< 20%) with a predominance of animal protein (Animal NP) or vegetable protein (Vegetable NP) and a high-protein diet (25-30%) with a predominance of animal protein (Animal HP) or vegetable protein (Vegetable HP). Baseline and final measurements of body weight, body composition, biochemical parameters, blood pressure (BP), resting energy expenditure and plasma amino acid profiles were performed. RESULTS: Body weight, BMI and waist circumference decreased in all groups. Interestingly, the IS improved more in the Animal HP (Matsuda index; 1.39 vs 2.58, P = 0.003) and in the Vegetable HP groups (Matsuda index; 1.44 vs 3.14, P < 0.0001) after one month. The fat mass, triglyceride levels, C-reactive protein levels and the leptin/adiponectin index decreased; while, the skeletal muscle mass increased in the Animal and Vegetable HP groups. The BP decreased in all groups except the Animal NP group. CONCLUSION: Our study demonstrates that a high-protein hypocaloric diets improves IS by 60-90% after one month in subjects with obesity and IR, regardless of weight loss and the source of protein, either animal or vegetable. TRIAL REGISTRATION: The trial is registered at clinicaltrials.gov (NCT03627104), August 13, 2018.


Subject(s)
Insulin Resistance , Adolescent , Body Mass Index , Diet, Reducing , Dietary Proteins , Humans , Obesity , Weight Loss
5.
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
6.
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
7.
PLoS One ; 19(2): e0299543, 2024.
Article in English | MEDLINE | ID: mdl-38422035

ABSTRACT

Circulating concentration of arginine, alanine, aspartate, isoleucine, leucine, phenylalanine, proline, tyrosine, taurine and valine are increased in subjects with insulin resistance, which could in part be attributed to the presence of single nucleotide polymorphisms (SNPs) within genes associated with amino acid metabolism. Thus, the aim of this work was to develop a Genetic Risk Score (GRS) for insulin resistance in young adults based on SNPs present in genes related to amino acid metabolism. We performed a cross-sectional study that included 452 subjects over 18 years of age. Anthropometric, clinical, and biochemical parameters were assessed including measurement of serum amino acids by high performance liquid chromatography. Eighteen SNPs were genotyped by allelic discrimination. Of these, ten were found to be in Hardy-Weinberg equilibrium, and only four were used to construct the GRS through multiple linear regression modeling. The GRS was calculated using the number of risk alleles of the SNPs in HGD, PRODH, DLD and SLC7A9 genes. Subjects with high GRS (≥ 0.836) had higher levels of glucose, insulin, homeostatic model assessment- insulin resistance (HOMA-IR), total cholesterol and triglycerides, and lower levels of arginine than subjects with low GRS (p < 0.05). The application of a GRS based on variants within genes associated to amino acid metabolism may be useful for the early identification of subjects at increased risk of insulin resistance.


Subject(s)
Insulin Resistance , Young Adult , Humans , Adolescent , Adult , Insulin Resistance/genetics , Cross-Sectional Studies , Genetic Risk Score , Alanine , Arginine
8.
Food Res Int ; 151: 110856, 2022 01.
Article in English | MEDLINE | ID: mdl-34980392

ABSTRACT

Several studies have shown that consumption of honey is associated with various health benefits. However, there is scarce evidence on whether honeys modify the intestinal microbiota by preventing the inflammatory response in the host. Therefore, the aim of the present work was to study the effect of Melipona (Mel) and Mantequilla (Mtq) honeys, which contain different bioactive compounds and antioxidant capacity on gut microbiota and metabolic consequences in comparison with other sweeteners, in particular sucrose (S) and high fructose corn syrup (HFCS) in rats. The results of the present work showed that both honeys have polyphenols, flavonoids, antioxidant and bactericidal activities. Rats fed with both honeys gained less weight and body fat by increasing energy expenditure compared to S or HFCS and increased gene expression of antioxidant enzymes mediated by the transcription factor Nrf2. Analysis of the gut microbiota showed that consumption of both honeys modified the beta-diversity compared to those fed S or HFCS resulting in increased abundance of a specific cluster of bacteria of the Clostridium genus particularly Coprococcus eutactus, Defluviitalea saccharophila, Ruminicoccus gnavus and Ruminicoccus flavefaciens. As a result of the changes in the gut microbiota, there was a decrease in LPS- and TLR4-mediated low-grade inflammation and an increase in sIgA. Consumption of both honeys prevented glucose intolerance and increased adipocyte size compared to S or HFCS. In conclusion, consumption of MtqH or MelH can reduce metabolic endotoxemia by modifying the gut microbiota to prevent glucose intolerance.


Subject(s)
Gastrointestinal Microbiome , High Fructose Corn Syrup , Honey , Animals , Bees , Inflammation/prevention & control , Rats , Sucrose
9.
Food Funct ; 12(7): 3206-3218, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33877245

ABSTRACT

Irritable Bowel Syndrome (IBS) is a functional gastrointestinal disorder characterized by abdominal pain and altered bowel habit. IBS patients report that FODMAP (Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols) diet induce or exacerbate their symptoms. It has been reported that low-FODMAP diet (LFD) improves the symptoms in 50%-80% of IBS patients. We aimed to identify IBS responders and non-responders' patients to LFD by determining baseline fecal microbial composition, sequencing the 16S rRNA gene V3-V4 region. Thirty-two participants with IBS were included, 29 women (90.62%) and three men (9.37%), and instructed to follow a four-week LFD, Visual Analogue Scale for IBS was used to assess intervention response. Twenty-two participants were responders (68.75%), and ten were non-responders (31.25%). Differential abundance analysis of Amplicon Sequence Variant (ASVs), before LFD, identified Prevotella 9 and Veillonella genus in responder group, and Barnesiella, Paraprevotella, Bifidobacterium and Ruminococcus 1 genus in non-responder group. After LFD, differentially abundant ASVs were only identified in R, belonging to Veilonella, Butyrivibrio, and 5 ASVs belonging to Ruminiclostridium 6 genus. Linear Discriminant Analysis (LDA), was used to classify patients by responsiveness, considering baseline abundance of 5 bacterial genera, LDA accuracy model was 96.87%, correctly classifying 95.45% of in responder group and 100% and non-responder group. In conclusion, bacterial biomarkers are useful to classify IBS individuals by responsiveness to LFD.


Subject(s)
Diet , Dietary Carbohydrates/administration & dosage , Fermentation , Gastrointestinal Microbiome/physiology , Irritable Bowel Syndrome/microbiology , Polymers/administration & dosage , Adult , Bacteria/classification , Disaccharides , Feces/microbiology , Female , Humans , Irritable Bowel Syndrome/diet therapy , Male , Mexico , Middle Aged , Monosaccharides , Oligosaccharides
10.
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
11.
Nutrients ; 14(1)2021 12 22.
Article in English | MEDLINE | ID: mdl-35010897

ABSTRACT

Obesity is associated with cognitive deficit and liver alterations; however, it remains unclear whether a combination of functional foods could reverse cognitive damage and to what extent it would be associated with changes in gut microbiota and liver. With this aim, male Wistar rats were fed a high-fat-5%sucrose diet (HFS) for 4 mo. And were then fed for 1 mo. with bioactive foods. At the end of this period, liver, serum, feces, intestine, and brain samples were taken. Body composition, energy expenditure, LPS, hormones, intraperitoneal glucose tolerance test, behavioral tests, and gut microbiota were evaluated. We showed that male rats fed high-fat-sucrose diet developed gut microbiota dysbiosis, increased in body fat, decreased antioxidant activity, decreased brain neuropeptide Y, increased the number of astrocytes and activated microglia, along with reduced spine density associated with deficits in working memory. Ingestion of a combination of nopal, soy protein, curcumin, and chia seed oil (bioactive foods) for three months was associated with an increase in a cluster of bacteria with anti-inflammatory capacity, a decrease in serum LPS levels and an increase in serum eicosapentaenoic acid (EPA) with neuroprotective properties. In the liver, ingestion of bioactive food significantly increased antioxidant enzymes, decreased lipogenesis, reduced inflammation mediated by the TLR4-TNFα pathway along with a decrease in body fat, glucose intolerance, and metabolic inflexibility. Finally, neuroinflammation in the brain was reduced and working memory improved. Our study demonstrates that consumption of bioactive foods was associated with reduced liver, brain, and gut microbiota alterations in obese rats.


Subject(s)
Brain/metabolism , Diet, High-Fat/adverse effects , Dietary Sucrose/administration & dosage , Dietary Sucrose/adverse effects , Food/classification , Liver/metabolism , Animals , Antioxidants , Bacteria/drug effects , Bacteria/genetics , Body Composition , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Glucose Intolerance , Insulin Resistance , Male , Occludin/genetics , Occludin/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Clin Nutr ; 40(6): 4209-4215, 2021 06.
Article in English | MEDLINE | ID: mdl-33583659

ABSTRACT

BACKGROUND & AIMS: The amino acid profile of young adults is modified by sex, body mass index (BMI) and insulin resistance (IR). However, we do not know if age or the presence of specific polymorphisms in the genes of BCAT2 and BCKDH contribute to changes in the amino acid profile, especially in subjects with obesity. Therefore, we have evaluated the effect of age, the presence of IR and the polymorphisms of BCAT2 rs11548193 and BCKDH rs45500792 on the concentration of amino acids in subjects with obesity. METHODS: This was a cross-sectional study conducted with 487 subjects with obesity. Participants underwent a physical examination in which their clinical history was obtained and a blood sample was taken for biochemical, hormonal, and DNA analysis. RESULTS: Adults <30 years old with obesity had higher levels of alanine, arginine, aspartate, histidine, leucine, lysine, methionine, phenylalanine, proline, serine and valine than adults ≥30 years old. Interestingly, regardless of age, we found that arginine, aspartate, serine decreased, while proline and tyrosine increased in the presence of IR; tyrosine and sum of branched-chain amino acids (∑BCAA) were the amino acids that increased in the presence of BCAT2 rs11548193 and BCKDH rs45500792 polymorphisms. CONCLUSIONS: We found that the amino acid profiles of subjects with obesity are differentially modified by age, the presence of IR, and the presence of the BCAT2 rs11548193 and BCKDH rs45500792 polymorphisms.


Subject(s)
3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Age Factors , Insulin Resistance/genetics , Minor Histocompatibility Antigens/genetics , Obesity/genetics , Pregnancy Proteins/genetics , Transaminases/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/blood , Adult , Amino Acids/blood , Cross-Sectional Studies , Female , Humans , Male , Minor Histocompatibility Antigens/blood , Obesity/blood , Polymorphism, Single Nucleotide/genetics , Pregnancy Proteins/blood , Transaminases/blood
13.
Nutrients ; 13(11)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34836148

ABSTRACT

Dietary fiber (DF) is a major substrate for the gut microbiota that contributes to metabolic health. Recent studies have shown that diet-metabolic phenotype effect might be related to individual gut microbial profiles or enterotypes. Thus, the aim of this study was to examine whether microbial enterotypes modify the association between DF intake and metabolic traits. This cross-sectional study included 204 children (6-12 years old) and 75 adults (18-60 years old). Habitual DF intake was estimated with a Food Frequency Questionnaire and biochemical, clinical and anthropometric data were obtained. Gut microbiota was assessed through 16S sequencing and participants were stratified by enterotypes. Correlations adjusting for age and sex were performed to test the associations between dietary fiber components intake and metabolic traits. In children and adults from the Prevotella enterotype, a nominal negative correlation of hemicellulose intake with insulin and HOMA-IR levels was observed (p < 0.05), while in individuals of the other enterotypes, these associations were not observed. Interestingly, the latter effect was not related to the fecal short-chain-fatty acids profile. Our results contribute to understanding the enterotype influence on the diet-phenotype interaction, which ultimate could provide evidence for their use as potential biomarkers for future precision nutrition strategies.


Subject(s)
Dietary Fiber/analysis , Eating/physiology , Gastrointestinal Microbiome/physiology , Insulin Resistance/physiology , Adolescent , Adult , Biomarkers/blood , Child , Cross-Sectional Studies , Diet Surveys , Eating/ethnology , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Humans , Insulin Resistance/ethnology , Male , Mexico/ethnology , Middle Aged , Phenotype , RNA, Ribosomal, 16S/analysis , Young Adult
14.
Gut Microbes ; 12(1): 1801301, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32804018

ABSTRACT

Fat and sweeteners contribute to obesity. However, it is unknown whether specific bacteria are selectively modified by different caloric and noncaloric sweeteners with or without a high-fat diet (HFD). Here, we combined extensive host phenotyping and shotgun metagenomics of the gut microbiota to investigate this question. We found that the type of sweetener and its combination with an HFD selectively modified the gut microbiota. Sucralose and steviol glycosides led to the lowest α-diversity of the gut microbiota. Sucralose increased the abundance of B. fragilis in particular, resulting in a decrease in the abundance of occludin and an increase in proinflammatory cytokines, glucose intolerance, fatty acid oxidation and ketone bodies. Sucrose+HFD showed the highest metabolic endotoxemia, weight gain, body fat, total short chain fatty acids (SCFAs), serum TNFα concentration and glucose intolerance. Consumption of sucralose or sucrose resulted in enrichment of the bacterial genes involved in the synthesis of LPS and SCFAs. Notably, brown sugar and honey were associated with the absence of metabolic endotoxemia, increases in bacterial gene diversity and anti-inflammatory markers such as IL-10 and sIgA, the maintenance of glucose tolerance and energy expenditure, similar to the control group, despite the consumption of an HFD. These findings indicate that the type of sweetener and an HFD selectively modify the gut microbiota, bacterial gene enrichment of metabolic pathways involved in LPS and SCFA synthesis, and metabolic endotoxemia associated with different metabolic profiles.


Subject(s)
Endotoxemia/etiology , Fatty Acids/adverse effects , Sweetening Agents/adverse effects , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Diet, High-Fat/adverse effects , Endotoxemia/metabolism , Endotoxemia/microbiology , Fatty Acids/metabolism , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome , Humans , Male , Rats , Rats, Wistar , Sweetening Agents/metabolism
16.
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
17.
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
18.
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
19.
J Nutr Biochem ; 59: 29-36, 2018 09.
Article in English | MEDLINE | ID: mdl-29960114

ABSTRACT

Metabolic profiling studies have highlighted increases in the plasma free fatty acid (FFA) and branched-chain amino acid (BCAA) concentrations, which are hallmarks of the obese and insulin-resistant phenotype. However, little is known about how the increase of the BCAA concentration modifies the metabolic fate of FFA, and vice versa, in adipocytes. Therefore, we incubated differentiated 3T3-L1 adipocytes or primary adipocytes from rats fed a control or a high-fat diet with: (1) 0, 250, 500 and 1000 µM of leucine and determined the oxidation and incorporation of [1-14C]-palmitate into lipids or proteins or (2) 0, 250, 500 or 1000 µM of palmitate and evaluated the oxidation and incorporation of [U-14C]-leucine into lipids or proteins. Leucine decreased palmitate oxidation and increased its incorporation into the lipid fraction in adipocytes; the latter was reduced in adipocytes from obese rats. However, palmitate increased leucine oxidation in adipocytes as well as reduced leucine incorporation into the protein and lipid fractions in adipocytes from obese rats. These results demonstrate that leucine modifies the metabolic fate of palmitate, and vice versa, in adipocytes and that the metabolic interaction between leucine and palmitate catabolism is altered in adipocytes from obese rats.


Subject(s)
Adipocytes/metabolism , Leucine/metabolism , Obesity/metabolism , Palmitates/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Carnitine O-Palmitoyltransferase/genetics , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Leucine/administration & dosage , Leucine/pharmacokinetics , Lipid Metabolism/drug effects , Male , Membrane Transport Proteins/genetics , Mice , Monocarboxylic Acid Transporters , Obesity/pathology , Palmitates/administration & dosage , Palmitates/pharmacokinetics , Peroxisomal Bifunctional Enzyme/genetics , Rats, Inbred Strains , Rats, Sprague-Dawley
20.
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