Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
J Physiol Biochem ; 79(2): 367-369, 2023 May.
Article in English | MEDLINE | ID: mdl-37310667

ABSTRACT

This Special Issue of the Journal of Physiology and Biochemistry contains 7 contributions that have been elaborated in the context of the mini-network "Consortium of Trans-Pyrenean Investigations on Obesity and Diabetes" (CTPIOD), which is on its 18th year of existence. This scientific community, mostly involving research groups from France and Spain, but also open to participants coming from all over the world, is focusing its attention on the prevention and the novel treatments of obesity, diabetes, non-alcoholic fatty liver disease, and other noncommunicable diseases. Accordingly, this special issue covers some nutritional, pharmacologic, and genetic aspects of the current knowledge of metabolic diseases. Some of these papers emerge from the lectures of the 18th Conference on Trans-Pyrenean Investigations in Obesity and Diabetes, organized by the University of Clermont-Ferrand and celebrated online in November 30, 2021.


Subject(s)
Diabetes Mellitus , Non-alcoholic Fatty Liver Disease , Humans , Obesity/metabolism , Spain
2.
J. physiol. biochem ; 79(2): 367-369, may. 2023.
Article in English | IBECS | ID: ibc-222548

ABSTRACT

This Special Issue of the Journal of Physiology and Biochemistry contains 7 contributions that have been elaborated in the context of the mini-network “Consortium of Trans-Pyrenean Investigations on Obesity and Diabetes” (CTPIOD), which is on its 18th year of existence. This scientific community, mostly involving research groups from France and Spain, but also open to participants coming from all over the world, is focusing its attention on the prevention and the novel treatments of obesity, diabetes, non-alcoholic fatty liver disease, and other noncommunicable diseases. Accordingly, this special issue covers some nutritional, pharmacologic, and genetic aspects of the current knowledge of metabolic diseases. Some of these papers emerge from the lectures of the 18th Conference on Trans-Pyrenean Investigations in Obesity and Diabetes, organized by the University of Clermont-Ferrand and celebrated online in November 30, 2021. (AU)


Subject(s)
Humans , Non-alcoholic Fatty Liver Disease , Diabetes Mellitus , Obesity/metabolism , Spain
3.
Lipids Health Dis ; 18(1): 178, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31615571

ABSTRACT

BACKGROUND: Chronic illnesses like obesity, type 2 diabetes (T2D) and cardiovascular diseases, are worldwide major causes of morbidity and mortality. These pathological conditions involve interactions between environmental, genetic, and epigenetic factors. Recent advances in nutriepigenomics are contributing to clarify the role of some nutritional factors, including dietary fatty acids in gene expression regulation. This systematic review assesses currently available information concerning the role of the different fatty acids on epigenetic mechanisms that affect the development of chronic diseases or induce protective effects on metabolic alterations. METHODS: A targeted search was conducted in the PubMed/Medline databases using the keywords "fatty acids and epigenetic". The data were analyzed according to the PRISMA-P guidelines. RESULTS: Consumption fatty acids like n-3 PUFA: EPA and DHA, and MUFA: oleic and palmitoleic acid was associated with an improvement of metabolic alterations. On the other hand, fatty acids that have been associated with the presence or development of obesity, T2D, pro-inflammatory profile, atherosclerosis and IR were n-6 PUFA, saturated fatty acids (stearic and palmitic), and trans fatty acids (elaidic), have been also linked with epigenetic changes. CONCLUSIONS: Fatty acids can regulate gene expression by modifying epigenetic mechanisms and consequently result in positive or negative impacts on metabolic outcomes.


Subject(s)
Cardiovascular Diseases/genetics , Diabetes Mellitus, Type 2/genetics , Dietary Fats/administration & dosage , Epigenesis, Genetic , Lipid Metabolism/genetics , Obesity/genetics , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/prevention & control , Chronic Disease , DNA Methylation , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Disease Models, Animal , Fatty Acids/administration & dosage , Fatty Acids/adverse effects , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Gene-Environment Interaction , Humans , Insulin Resistance , Lipid Metabolism/drug effects , Obesity/metabolism , Obesity/pathology , Obesity/prevention & control , Trans Fatty Acids/administration & dosage , Trans Fatty Acids/adverse effects
4.
Food Funct ; 9(8): 4207-4215, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29993072

ABSTRACT

Autophagy eliminates damaged cellular components. In the liver, it has been proposed that it mediates the breakdown of lipid droplets. This study aimed to compare the involvement of autophagy and the oxidative status in the effects of resveratrol and energy restriction as therapeutic tools for managing liver steatosis. In addition, potential additive or synergic effects were studied. Rats were fed a high-fat high-sucrose diet for 6 weeks and then divided into four experimental groups and fed a standard diet: a control group (C), a resveratrol-treated group (RSV, 30 mg kg-1 d-1), an energy restricted group (R, -15%), and an energy restricted group treated with resveratrol (RR). Liver triacylglycerols (TGs) were measured by Folch's method. TBARS, GSH, GSSG, GPx and SOD were assessed using commercial kits. The protein expression of beclin, atg5 and p62, as well as ratios of pSer555 ULK1/total ULK1, pSer757 ULK1/total ULK1 and LC3 II/I were determined by western blotting. Energy restriction increased the protein expression of beclin, atg5 and pSer757 ULK1/total ULK1 and LC3 II/I ratios, and reduced the protein expression of p62, thus indicating that it induced autophagy activation. The effects of resveratrol were similar but less marked than the hypocaloric diet. No differences were observed in oxidative stress determinants except for TBARS, which was decreased by energy restriction. In conclusion, resveratrol can reverse partially dietary-induced hepatic lipid accumulation, although less efficiently than energy restriction. The delipidating effect of energy restriction is mediated in part by the activation of autophagy; however, the involvement of this process in the effects of resveratrol is less clear.


Subject(s)
Autophagy/physiology , Fatty Liver/chemically induced , Fatty Liver/drug therapy , Resveratrol/pharmacology , Animals , Blotting, Western , Caloric Restriction , Diet, High-Fat/adverse effects , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/adverse effects , Male , Non-alcoholic Fatty Liver Disease/prevention & control , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sucrose/administration & dosage , Sucrose/adverse effects
5.
J Physiol Biochem ; 74(4): 579-589, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29790117

ABSTRACT

The main aim of this investigation was to study the regulatory roles of let-7b and miR-155-3p on the expression of inflammation-associated genes in monocytes, macrophages, and lipopolysaccharide (LPS)-activated macrophages (AcM). A second goal was to analyze the potential modulatory roles of different fatty acids, including oleic, palmitic, eicosapentaenoic (EPA), and docosahexaenoic (DHA), on the expression of these miRNAs in the three cell types. This hypothesis was tested in human acute monocytic leukemia cells (THP-1), which were differentiated into macrophages with 2-O-tetradecanoylphorbol-13-acetate (TPA) and further activated with LPS for 24 h. Monocytes, macrophages, and AcM were transfected with a negative control, or mimics for miR-155-3p and miR-let-7b-5p. The expression of both miRNAs and some proinflammatory genes was analyzed by qRT-PCR. Interestingly, let-7b mimic reduced the expression of IL6 and TNF in monocytes, and SERPINE1 expression in LPS-activated macrophages. However, IL6, TNF, and SERPINE1 were upregulated in macrophages by let-7b mimic. IL6 expression was higher in the three types of cells after transfecting with miR-155-3p mimic. Similarly, expression of SERPINE1 was increased by miR-155-3p mimic in monocytes and macrophages. However, TLR4 was downregulated by miR-155-3p in monocytes and macrophages. Regarding the effects of the different fatty acids, oleic acid increased the expression of let-7b in macrophages and AcM and also increased the expression of miR-155 in monocytes when compared with DHA but not when compared with non-treated cells. Overall, these results suggest anti- and proinflammatory roles of let-7b and miR-155-3p in THP-1 cells, respectively, although these outcomes are strongly dependent on the cell type. Noteworthy, oleic acid might exert beneficial anti-inflammatory effects in immune cells (i.e., non-activated and LPS-activated macrophages) by upregulating the expression of let-7b.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation , Macrophages/metabolism , MicroRNAs/metabolism , Monocytes/metabolism , Cell Differentiation/drug effects , Docosahexaenoic Acids/metabolism , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Interleukin-6/agonists , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , MicroRNAs/chemistry , Monocytes/immunology , Oleic Acid/metabolism , Plasminogen Activator Inhibitor 1/agonists , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , RNA/metabolism , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Cell Endocrinol ; 473: 257-267, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29407195

ABSTRACT

Obesity is characterized by hypertrophy and hyperplasia of adipose tissue, which have been related to the development of hypoxia and insulin resistance. On the other hand, caveolin-1 (Cav-1), one of the main proteins of caveolae, promotes insulin receptor (IR) phosphorylation and the subsequent activation of insulin signaling. In this work we investigated the effect of hypoxia on Cav-1 regulation and the status of insulin signaling in 3T3-L1 adipocytes. Our results showed that hypoxia inhibited adipogenesis and insulin signaling in adipocytes. Furthermore, 48 h of hypoxia reduced insulin-induced glucose uptake while increased basal glucose uptake. This result was consistent with the upregulation of glucose transporter GLUT1 and the downregulation of GLUT4, which also showed defective translocation to plasma membrane when adipocytes were stimulated with insulin. In addition, the expression of caveolae-related proteins was reduced by hypoxia and chromatin immunoprecipitation assay demonstrated that Cav-1 transcription was directly regulated by HIF-1. These results strengthen the role of caveolae in insulin signaling and help to explain adipocyte response to hypoxia.


Subject(s)
Adipocytes/metabolism , Adipocytes/pathology , Caveolae/metabolism , Insulin/metabolism , Signal Transduction , 3T3-L1 Cells , Animals , Caveolins/metabolism , Cell Hypoxia/genetics , Cell Membrane/metabolism , Down-Regulation/genetics , Glucose Transporter Type 4/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/metabolism , Mice , Phosphorylation , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Triglycerides/metabolism
7.
Nutr Metab Cardiovasc Dis ; 28(2): 165-172, 2018 02.
Article in English | MEDLINE | ID: mdl-29331538

ABSTRACT

BACKGROUND AND AIMS: A precise nutrigenetic management of hypercholesterolemia involves the understanding of the interactions between the individual's genotype and dietary intake. The aim of this study was to analyze the response to two dietary energy-restricted interventions on cholesterol changes in carriers of two ADRB2 polymorphisms. METHODS AND RESULTS: A 4-month nutritional intervention was conducted involving two different hypo-energetic diets based on low-fat (LF) and moderately high-protein (MHP) dietary patterns. A total of 107 unrelated overweight/obese individuals were genotyped for two ADRB2 non-synonymous polymorphisms: Arg16Gly (rs1042713) and Gln27Glu (rs1042714). Genotyping was performed by next-generation sequencing and haplotypes were phenotypically screened. Anthropometric measurements and the biochemical profile were assessed by conventional methods. Both diets induced cholesterol decreases at the end of both nutritional interventions. Interestingly, phenotypical differences were observed according to the Arg16Gly polymorphism. Within the MHP group, Gly16Gly homozygotes had lower reductions in total cholesterol (-6.5 mg/dL vs. -24.2 mg/dL, p = 0.009), LDL-c levels (-1.4 mg/dL vs. -16.5 mg/dL, p = 0.005), and non-HDL-c (-4.5 mg/dL vs. -21.5 mg/dL, p = 0.008) than Arg16 allele carriers. Conversely, within the LF group, Gly16Gly homozygotes underwent similar falls in total cholesterol (-18.5 mg/dL vs. -18.7 mg/dL, ns), LDL-c levels (-9.7 mg/dL vs. -13.1 mg/dL, ns), and non-HDL-c (-15.3 mg/dL vs. -15.7 mg/dL, ns) than Arg16 allele carriers. The Gln27Glu polymorphism and the Gly16/Glu27 haplotype showed similar, but not greater effects. CONCLUSIONS: An energy-restricted LF diet could be more beneficial than a MHP diet to reduce serum cholesterol, LDL-c, and non-HDL-c among Gly16Gly genotype carriers. CLINICALTRIALS.GOV: Identifier: NCT02737267.


Subject(s)
Caloric Restriction , Cholesterol/blood , Diet, Fat-Restricted , Diet, High-Protein , Obesity/diet therapy , Obesity/genetics , Polymorphism, Genetic , Receptors, Adrenergic, beta-2/genetics , Weight Loss , Adult , Biomarkers/blood , Female , Haplotypes , Heterozygote , Homozygote , Humans , Male , Middle Aged , Obesity/blood , Obesity/physiopathology , Phenotype , Time Factors , Treatment Outcome
8.
Appetite ; 120: 230-239, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28888730

ABSTRACT

Individual differences in taste perception may influence appetite, dietary intakes, and subsequently, disease risk. Correlations of DNA methylation patterns at taste transducing genes with BMI and dietary intakes were studied. A nutriepigenomic analysis within the Methyl Epigenome Network Association (MENA) project was conducted in 474 adults. DNA methylation in peripheral white blood cells was analyzed by a microarray approach. KEGG pathway analyses were performed concerning the characterization and discrimination of genes involved in the taste transduction pathway. Adjusted FDR values (p < 0.0001) were used to select those CpGs that showed best correlation with BMI. A total of 29 CpGs at taste transducing genes met the FDR criteria. However, only 12 CpGs remained statistically significant after linear regression analyses adjusted for age and sex. These included cg15743657 (TAS1R2), cg02743674 (TRPM5), cg01790523 (SCN9A), cg15947487 (CALHM1), cg11658986 (ADCY6), cg04149773 (ADCY6), cg02841941 (P2RY1), cg02315111 (P2RX2), cg08273233 (HTR1E), cg14523238 (GABBR2), cg12315353 (GABBR1) and cg05579652 (CACNA1C). Interestingly, most of them were implicated in the sweet taste signaling pathway, except CACNA1C (sour taste). In addition, TAS1R2 methylation at cg15743657 was strongly correlated with total energy (p < 0.0001) and carbohydrate intakes (p < 0.0001). This study suggests that methylation in genes related to sweet taste could be an epigenetic mechanism associated with obesity.


Subject(s)
Body Mass Index , DNA Methylation/genetics , Dietary Carbohydrates , Eating/genetics , Taste Perception/genetics , Adult , Female , Humans , Linear Models , Male , Middle Aged , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics
9.
Pediatr Obes ; 13(3): 149-158, 2018 03.
Article in English | MEDLINE | ID: mdl-28614626

ABSTRACT

BACKGROUND: The global prevalence of childhood overweight and obesity has increased in the last years. Epigenetic dysregulation affecting gene expression could be a determinant in early-life obesity onset and accompanying complications. OBJECTIVE: The aim of the present investigation was to analyse the putative association between DNA methylation and childhood obesity. METHODS: DNA was isolated from white blood cells of 24 children obtained from the GENOI study and was hybridized in a 450K methylation array. Two CpG sites associated with obesity were validated in 91 children by MassArray® EpiTyper™ technology. RESULTS: Genome-wide analysis identified 734 CpGs (783 genes) differentially methylated between cases (n = 12) and controls (n = 12). Ingenuity Pathway Analysis showed that these genes were involved in oxidative stress and circadian rhythm signalling pathways. Moreover, the DNA methylation levels of VIPR2, GRIN2D, ADCYAP1R1, PER3 and PTPRS regions correlated with the obesity trait. EpiTyper™ validation also identified significant correlations between methylation levels of CpG sites on PTPRS and PER3 with BMI z-score. CONCLUSIONS: This study identified several CpG sites and specifically several CpGs in the PTPRS and PER3 genes differentially methylated between obese and non-obese children, suggesting a role for DNA methylation concerning development of childhood obesity.


Subject(s)
DNA Methylation/genetics , Pediatric Obesity/genetics , Period Circadian Proteins/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Case-Control Studies , Child , CpG Islands , Cross-Sectional Studies , Female , Genome-Wide Association Study , Humans , Male , Phenotype , Reproducibility of Results
10.
Sci Rep ; 7: 41903, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28211912

ABSTRACT

The characterization of the epigenetic changes within the obesity-related adipose tissue will provide new insights to understand this metabolic disorder, but adipose tissue is not easy to sample in population-based studies. We aimed to evaluate the capacity of circulating leukocytes to reflect the adipose tissue-specific DNA methylation status of obesity susceptibility. DNA samples isolated from subcutaneous adipose tissue and circulating leukocytes were hybridized in the Infinium HumanMethylation 450 BeadChip. Data were compared between samples from obese (n = 45) and non-obese (n = 8-10) patients by Wilcoxon-rank test, unadjusted for cell type distributions. A global hypomethylation of the differentially methylated CpG sites (DMCpGs) was observed in the obese subcutaneous adipose tissue and leukocytes. The overlap analysis yielded a number of genes mapped by the common DMCpGs that were identified to reflect the obesity state in the leukocytes. Specifically, the methylation levels of FGFRL1, NCAPH2, PNKD and SMAD3 exhibited excellent and statistically significant efficiencies in the discrimination of obesity from non-obesity status (AUC > 0.80; p < 0.05) and a great correlation between both tissues. Therefore, the current study provided new and valuable DNA methylation biomarkers of obesity-related adipose tissue pathogenesis through peripheral blood analysis, an easily accessible and minimally invasive biological material instead of adipose tissue.


Subject(s)
DNA Methylation , Leukocytes/metabolism , Obesity/genetics , Subcutaneous Fat/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , CpG Islands , Female , Genome, Human , Humans , Male , Middle Aged , Muscle Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Serine Endopeptidases/genetics , Smad3 Protein/genetics
11.
Biofactors ; 43(3): 371-378, 2017 May 06.
Article in English | MEDLINE | ID: mdl-28218490

ABSTRACT

Resveratrol (RSV) has been proposed as an energy restriction mimetic. This study aimed to compare the effects of RSV and energy restriction on insulin resistance induced by an obesogenic diet. Any additive effect of both treatments was also analyzed. Rats were fed a high-fat high-sucrose diet for 6 weeks. They were then distributed in four experimental groups which were either fed a standard control diet (C), or treated with RSV (30 mg/kg/d), or submitted to energy restriction (R, 15%), or treated with RSV and submitted to energy restriction (RR). A glucose tolerance test was performed, and serum glucose, insulin, fructosamine, adiponectin, and leptin concentrations determined. Muscle triacylglycerol content and protein expression of insulin receptor (IRß), protein kinase B (Akt), Akt substrate of 160 kDa (AS160) and glucose transporter 4 (GLUT-4) were measured. In RSV rats, fructosamine concentrations were reduced, HOMA-IR remained unchanged, but glucose tolerance was improved, without changes in phosphorylation of IRß, Akt, and AS160 or in GLUT-4 protein expression. Rats under energy restriction showed an improvement in all the markers related to glycemic control, as well as increased phosphorylation of AS160 and protein expression of GLUT-4. In rats from RR group the results were similar to R group, with the exception of IRß and Akt phosphorylation, which were increased. In conclusion, mild energy restriction is more efficient than intake of RSV within a standard balanced diet, and acts by means of a different mechanism from that of RSV. No additive effects between RSV and energy restriction were observed. © 2017 BioFactors, 43(3):371-378, 2017.


Subject(s)
Antioxidants/pharmacology , Blood Glucose/metabolism , Caloric Restriction , Muscle, Skeletal/drug effects , Obesity/prevention & control , Stilbenes/pharmacology , Adiponectin/blood , Animals , Diet, High-Fat/adverse effects , Fructosamine/blood , Gene Expression/drug effects , Glucose Tolerance Test , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Insulin/blood , Insulin Resistance , Leptin/blood , Male , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Resveratrol , Sucrose/administration & dosage , Triglycerides/blood
12.
Pediatr Obes ; 12(1): 19-27, 2017 02.
Article in English | MEDLINE | ID: mdl-26780939

ABSTRACT

BACKGROUND: Epigenetic mechanisms may be involved in obesity onset and its consequences. The aim of the present study was to evaluate whether DNA methylation status in microRNA (miRNA) coding regions is associated with childhood obesity. MATERIAL AND METHODS: DNA isolated from white blood cells of 24 children (identification sample: 12 obese and 12 non-obese) from the Grupo Navarro de Obesidad Infantil study was hybridized in a 450 K methylation microarray. Several CpGs whose DNA methylation levels were statistically different between obese and non-obese were validated by MassArray® in 95 children (validation sample) from the same study. RESULTS: Microarray analysis identified 16 differentially methylated CpGs between both groups (6 hypermethylated and 10 hypomethylated). DNA methylation levels in miR-1203, miR-412 and miR-216A coding regions significantly correlated with body mass index standard deviation score (BMI-SDS) and explained up to 40% of the variation of BMI-SDS. The network analysis identified 19 well-defined obesity-relevant biological pathways from the KEGG database. MassArray® validation identified three regions located in or near miR-1203, miR-412 and miR-216A coding regions differentially methylated between obese and non-obese children. CONCLUSIONS: The current work identified three CpG sites located in coding regions of three miRNAs (miR-1203, miR-412 and miR-216A) that were differentially methylated between obese and non-obese children, suggesting a role of miRNA epigenetic regulation in childhood obesity.


Subject(s)
DNA Methylation , Pediatric Obesity/genetics , Child , Epigenesis, Genetic , Female , Genetic Predisposition to Disease , Humans , Male , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis/methods
13.
J Physiol Biochem ; 73(3): 445-455, 2016 Aug.
Article in English | MEDLINE | ID: mdl-28181167

ABSTRACT

Epigenetic processes, including DNA methylation, might be modulated by environmental factors such as the diet, which in turn have been associated with the onset of several diseases such as obesity or cardiovascular events. Meanwhile, Mediterranean diet (MedDiet) has demonstrated favourable effects on cardiovascular risk, blood pressure, inflammation and other complications related to excessive adiposity. Some of these effects could be mediated by epigenetic modifications. Therefore, the objective of this study was to investigate whether the adherence to MedDiet is associated with changes in the methylation status from peripheral blood cells. A subset of 36 individuals was selected within the Prevención con Dieta Mediterránea (PREDIMED)-Navarra study, a randomised, controlled, parallel trial with three groups of intervention in high cardiovascular risk volunteers, two with a MedDiet and one low-fat control group. Changes in methylation between baseline and 5 years were studied. DNA methylation arrays were analysed by several robust statistical tests and functional classifications. Eight genes related to inflammation and immunocompetence (EEF2, COL18A1, IL4I1, LEPR, PLAGL1, IFRD1, MAPKAPK2, PPARGC1B) were finally selected as changes in their methylation levels correlated with adherence to MedDiet and because they presented sensitivity related to a high variability in methylation changes. Additionally, EEF2 methylation levels positively correlated with concentrations of TNF-α and CRP. This report is apparently the first showing that adherence to MedDiet is associated with the methylation of the reported genes related to inflammation with a potential regulatory impact.


Subject(s)
Diabetes Mellitus, Type 2/diet therapy , Diet, Mediterranean , Leukocytes/metabolism , Aged , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , DNA Methylation , Diabetes Mellitus, Type 2/metabolism , Epigenesis, Genetic , Female , Humans , Inflammation/genetics , Inflammation/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Male , Middle Aged , Transcriptome , Treatment Adherence and Compliance , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
14.
Cell Mol Biol (Noisy-le-grand) ; 61(8): 9-16, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26667767

ABSTRACT

A DNA microarray analysis was conducted in Caco-2 cells to analyse the protective effects of trans-resveratrol on enterocyte physiology and metabolism in pro-inflammatory conditions. Cells were pre-treated with 50 µΜ of trans-resveratrol and, subsequently, lipopolysaccharide (LPS) was added for 48 h. The microarray analysis revealed 121 genes differentially expressed between resveratrol-treated and non-treated cells (B> 0, is the odd thatthe gene is differentially expressed). Inhibitor of DNA binding 1 (ID1), histidine-rich glycoprotein (HRG), NADPH oxidase (NOX1) and sprouty homolog 1 (SPRY), were upregulated by LPS treatment, but significantly blocked by trans-resveratrol pre-treatment (padj< 0.05, after adjusting for Benjamini-Hocheberg procedure). Moreover, genes implicated in synthesis of lipids (z-score= -1.195) and concentration of cholesterol (z-score= -0.109), were markedly downregulated by trans-resveratrol. Other genes involved in fat turnover, but also in cell death and survival function, such as transcription factors Krüppel-like factor 5 (KLF5) and amphiregulin (AREG), were also significantly inhibited by trans-resveratrol pre-treatment. RT-qPCR-data confirmed the microarray results. Special mention deserves acyl-CoA synthetase long-chain family member 3 (ACSL3) and endothelial lipase (LIPG), which were downregulated by this stilbene and have been previously associated with fatty acid synthesis and obesity in other tissues. This study envisages that trans-resveratrol might exert an important anti-lipogenic effect at intestinal level under pro-inflammatory conditions, which has not been previously described.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol/metabolism , Enterocytes/drug effects , Lipogenesis/drug effects , Stilbenes/pharmacology , Amphiregulin , Caco-2 Cells , Cell Death/drug effects , Cell Death/genetics , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , EGF Family of Proteins/genetics , EGF Family of Proteins/metabolism , Enterocytes/cytology , Enterocytes/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lipase/genetics , Lipase/metabolism , Lipogenesis/genetics , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microarray Analysis , NADPH Oxidase 1 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteins , Resveratrol , Signal Transduction
15.
J Nutr Biochem ; 26(6): 651-60, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25762527

ABSTRACT

Diet-induced obesity is associated to an imbalance in the normal gut microbiota composition. Resveratrol and quercetin, widely known for their health beneficial properties, have low bioavailability, and when they reach the colon, they are targets of the gut microbial ecosystem. Hence, the use of these molecules in obesity might be considered as a potential strategy to modulate intestinal bacterial composition. The purpose of this study was to determine whether trans-resveratrol and quercetin administration could counteract gut microbiota dysbiosis produced by high-fat sucrose diet (HFS) and, in turn, improve gut health. Wistar rats were randomised into four groups fed an HFS diet supplemented or not with trans-resveratrol [15 mg/kg body weight (BW)/day], quercetin (30 mg/kg BW/day) or a combination of both polyphenols at those doses. Administration of both polyphenols together prevented body weight gain and reduced serum insulin levels. Moreover, individual supplementation of trans-resveratrol and quercetin effectively reduced serum insulin levels and insulin resistance. Quercetin supplementation generated a great impact on gut microbiota composition at different taxonomic levels, attenuating Firmicutes/Bacteroidetes ratio and inhibiting the growth of bacterial species previously associated to diet-induced obesity (Erysipelotrichaceae, Bacillus, Eubacterium cylindroides). Overall, the administration of quercetin was found to be effective in lessening HFS-diet-induced gut microbiota dysbiosis. In contrast, trans-resveratrol supplementation alone or in combination with quercetin scarcely modified the profile of gut bacteria but acted at the intestinal level, altering the mRNA expression of tight-junction proteins and inflammation-associated genes.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Quercetin/administration & dosage , Stilbenes/administration & dosage , Animals , Bacillus/drug effects , Bacillus/growth & development , Bacteroidetes/drug effects , Bacteroidetes/growth & development , DNA, Bacterial/genetics , Diet, High-Fat/adverse effects , Dietary Supplements , Fatty Acids, Volatile/metabolism , Firmicutes/drug effects , Firmicutes/growth & development , Gas Chromatography-Mass Spectrometry , Insulin Resistance , Obesity/microbiology , Rats , Rats, Wistar , Resveratrol , Sucrose/administration & dosage , Sucrose/adverse effects , Weight Gain
16.
Br J Pharmacol ; 172(11): 2756-68, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25046997

ABSTRACT

Many nutrients are known for a wide range of activities in prevention and alleviation of various diseases. Recently, their potential role in regulating human health through effects on epigenetics has become evident, although specific mechanisms are still unclear. Thus, nutriepigenetics/nutriepigenomics has emerged as a new and promising field in current epigenetics research in the past few years. In particular, polyphenols, as part of the central dynamic interaction between the genome and the environment with specificity at physiological concentrations, are well known to affect mechanisms underlying human health. This review summarizes the effects of dietary compounds on epigenetic mechanisms in the regulation of gene expression including expression of enzymes and other molecules responsible for drug absorption, distribution, metabolism and excretion in cancer, metabolic syndrome, neurodegenerative disorders and hormonal dysfunction.


Subject(s)
Diet , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Metabolic Syndrome/genetics , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Phytoestrogens/pharmacology , Trace Elements/pharmacology , Vitamins/pharmacology , Antineoplastic Agents/pharmacology , Coffee , Curcumin/pharmacology , Folic Acid/pharmacology , Food , Gene Expression Regulation/drug effects , Humans , Polyphenols/pharmacology , S-Adenosylmethionine/pharmacology , Selenium/pharmacology , Vitamin B 12/pharmacology , Vitamin B Complex/pharmacology
17.
J Anim Physiol Anim Nutr (Berl) ; 99(5): 834-40, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25521806

ABSTRACT

Maternal nutrition during pregnancy and lactation influences offspring development and health. Novel studies have described the effects on next generation obesity-related features depending on maternal macro- and micro-nutrient perinatal feeding. We hypothesized that the maternal obesogenic diet during pregnancy and lactation programs an obese phenotype, while maternal micronutrient supplementation at these stages could partially prevent these features. Thus, the aim was to assess the influence of a perinatal maternal feeding with an obesogenic diet enriched in fat and sucrose and a micronutrient supplementation during pregnancy and lactation on offspring growth and obese phenotypical features during life course. Female Wistar rats were assigned to four dietary groups during pregnancy and lactation: control, control supplemented with micronutrients (choline, betaine, folic acid and vitamin B12 ), high-fat sucrose (HFS) and HFS supplemented. At weaning, the offspring were transferred to a chow diet, and weight and fat mass were measured at weeks 3, 12 and 20. At birth, both male and female offspring from mothers fed the obesogenic diet showed lower body weight (-5 and -6%, respectively), while only female offspring weight decreased by maternal micronutrient supplementation (-5%). During lactation, maternal HFS diet was associated with increased body weight, while micronutrient supplementation protected against body weight gain. Whole body fat mass content increased at weeks 3, 12 and 20 (from 16 to 65%) due to maternal HFS diet. Maternal micronutrient supplementation decreased offspring fat mass content at week 3 (-8%). Male offspring showed higher adiposity than females at weeks 12 and 20. In conclusion, maternal HFS feeding during pregnancy and lactation was associated with a low offspring weight at birth and obese phenotypical features during adult life in a sex- and time-dependent manner. Furthermore, maternal methyl donor supplementation protected against body weight gain in male offspring during lactation and in female offspring also during juvenile period.


Subject(s)
Animal Feed/analysis , Body Fat Distribution , Diet/veterinary , Animal Nutritional Physiological Phenomena , Animals , Female , Male , Maternal Nutritional Physiological Phenomena , Pregnancy , Rats , Rats, Wistar
18.
Benef Microbes ; 6(1): 97-111, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25213025

ABSTRACT

The gastrointestinal tract harbours a 'superorganism' called the gut microbiota, which is known to play a crucial role in the onset and development of diverse diseases. This internal ecosystem, far from being a static environment, can be manipulated by diet and dietary components. Feeding animals with high-fat sucrose (HFS) diets entails diet-induced obesity, a model which is usually used in research to mimic the obese phenotype of Western societies. The aim of the present study was to identify gut microbiota dysbiosis and associated metabolic changes produced in male Wistar rats fed a HFS diet for 6 weeks and compare it with the basal microbial composition. For this purpose, DNA extracted from faeces at baseline and after treatment was analysed by amplification of the V4-V6 region of the 16S ribosomal DNA (rDNA) gene using 454 pyrosequencing. Short-chain fatty acids, i.e. acetate, propionate and butyrate, were also evaluated by gas chromatography-mass spectrometry. At the end of the treatment, gut microbiota composition significantly differed at phylum level (Firmicutes, Bacteroidetes and Proteobacteria) and class level (Erisypelotrichi, Deltaproteobacteria, Bacteroidia and Bacilli). Interestingly, the class Clostridia showed a significant decrease after HFS diet treatment, which correlated with visceral adipose tissue, and is likely mediated by dietary carbohydrates. Of particular interest, Clostridium cluster XIVa species were significantly reduced and changes were identified in the relative abundance of other specific bacterial species (Mitsuokella jalaludinii, Eubacterium ventriosum, Clostridium sp. FCB90-3, Prevotella nanceiensis, Clostridium fusiformis, Clostridium sp. BNL1100 and Eubacterium cylindroides) that, in some cases, showed opposite trends to their relative families. These results highlight the relevance of characterising gut microbial population differences at species level and contribute to understand the plausible link between diet and specific gut bacterial species that are able to influence the inflammatory status, intestinal barrier function and obesity development.


Subject(s)
Biota/drug effects , Diet/methods , Dysbiosis , Fats/administration & dosage , Gastrointestinal Tract/microbiology , Sucrose/administration & dosage , Animals , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Fatty Acids/analysis , Feces/chemistry , Feces/microbiology , Fermentation , Gas Chromatography-Mass Spectrometry , Phylogeny , RNA, Ribosomal, 16S/genetics , Rats, Wistar , Sequence Analysis, DNA
19.
J Dev Orig Health Dis ; 5(5): 385-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25084068

ABSTRACT

Methyl donor supplementation has been reported to prevent obesity-induced liver fat accumulation in adult rats. We hypothesized that this protection could be mediated by perinatal nutrition. For this purpose, we assessed the response to an obesogenic diet (high-fat-sucrose, HFS) during adulthood depending on maternal diet during lactation. Female Wistar rats fed control diet during pregnancy were assigned to four postpartum dietary groups: control, control supplemented with methyl donors (choline, betaine, folic acid, vitamin B12), HFS and HFS supplemented with methyl donors. At weaning, the male offspring was transferred to a chow diet and at week 12th assigned to a control or a HFS diet during 8 weeks. The offspring whose mothers were fed HFS during lactation showed increased adiposity (19%, P<0.001). When fed the HFS diet as adults, offspring whose mothers were HFS supplemented had more body fat (23%, P<0.001) than those from HFS non-supplemented. However, they showed lower liver fat accumulation (-18%, P<0.001). Srebf1, Dnmt1 and Lepr liver mRNA levels increased after adulthood HFS feeding. In those animals HFS fed during adulthood, previous maternal HFS decreased Lepr and Dnmt1 expression levels when compared with c-HFS offspring, while the supplementation of control and HFS-fed dams, respectively, induced higher hepatic Mme and Lepr mRNA levels after adult HFS intake compared with hfs-HFS offspring. In conclusion, maternal HFS diet during lactation influenced the response to an obesogenic diet in the adult progeny. Interestingly, dietary methyl donor supplementation in lactating mothers fed an obesogenic diet reduced liver fat accumulation, but increased adipose tissue storage in adult HFS-fed offspring.


Subject(s)
DNA Methylation/drug effects , Diet , Lactation/physiology , Liver/metabolism , Obesity/complications , Adipose Tissue/metabolism , Animals , Betaine/administration & dosage , Body Composition , Choline/administration & dosage , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Diet, High-Fat , Dietary Sucrose/administration & dosage , Dietary Supplements , Female , Folic Acid/administration & dosage , Gene Expression/drug effects , Lactation/drug effects , Liver/chemistry , Liver/drug effects , Male , Maternal Nutritional Physiological Phenomena , Obesity/etiology , Pregnancy , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Leptin/genetics , Vitamin B 12/administration & dosage
20.
J Physiol Biochem ; 70(2): 603-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24500802

ABSTRACT

Some causal bases of stroke remain unclear, but the nutritional effects on the epigenetic regulation of different genes may be involved. The aim was to assess the impact of epigenetic processes of human tumor necrosis factor (TNF-α) and paraoxonase (PON) promoters in the susceptibility to stroke when considering body composition and dietary intake. Twenty-four patients (12 non-stroke/12 stroke) were matched by sex (12 male/12 female), age (mean 70 ± 12 years old), and BMI (12 normal-weight/12 obese; mean 28.1 ± 6.7 kg/m(2)). Blood cell DNA was isolated and DNA methylation levels of TNF-α (-186 to +349 bp) and PON (-231 to +250 bp) promoters were analyzed by the Sequenom EpiTYPER approach. Histone modifications (H3K9ac and H3K4me3) were analyzed also by chromatin immunoprecipitation in a region of TNF-α (-297 to -185). Total TNF-α promoter methylation was lower in stroke patients (p < 0.001) and showed no interaction with body composition (p = 0.807). TNF-α and PON total methylation levels correlated each other (r = 0.44; p = 0.031), especially in stroke patients (r = 0.72; p = 0.008). The +309 CpG methylation site from TNF-α promoter was related to body weight (p = 0.027) and the region containing three CpGs (from -170 to -162 bp) to the percentage of lipid intake and dietary indexes (p < 0.05) in non-stroke patients. The methylation of PON +15 and +241 CpGs was related to body weight (p = 0.021), waist circumference (p = 0.020), and energy intake (p = 0.018), whereas +214 was associated to the quality of the diet (p < 0.05) in non-stroke patients. When comparing stroke vs non-stroke patients regarding the histone modifications analyzed at TNF-α promoter, no changes were found, although a significant association was identified between circulating TNF-α level and H3K9ac with H3K4me3. TNF-α and PON promoter methylation levels could be involved in the susceptibility to stroke and obesity outcome, respectively. The dietary intake and body composition may influence this epigenetic regulation in non-stroke patients.


Subject(s)
Aryldialkylphosphatase/genetics , Diet , Epigenesis, Genetic , Obesity/genetics , Promoter Regions, Genetic , Stroke/genetics , Tumor Necrosis Factor-alpha/genetics , Aged , Base Sequence , DNA Methylation , DNA Primers , Female , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...