Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Food Funct ; 11(10): 9057-9066, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33021612

ABSTRACT

Chemerin is a pro-inflammatory adipokine that is increased in obesity and associated with obesity-related comorbidities. The aim of this study was to investigate the effects of omega-3 polyunsaturated fatty acids, eicosapentaenoic and docosahexaenoic acids (EPA and DHA), on basal and tumor necrosis factor-α (TNF-α)-induced chemerin production in 3T3-L1 and human subcutaneous cultured adipocytes. The potential involvement of G protein-coupled receptor 120 (GPR120), as well as the actions of DHA-derived specialized proresolving lipid mediators (SPMs), resolvin D1 and D2 (RvD1 and RvD2) and maresin 1 (MaR1), were also evaluated. DHA significantly lowered both basal and TNF-α-stimulated chemerin production in 3T3-L1 and human adipocytes. EPA did not modify basal chemerin production, while it attenuated the induction of chemerin by TNF-α. Silencing of GPR120 using siRNA blocked the ability of DHA and EPA to reduce TNF-α-induced chemerin secretion. Interestingly, treatment with the DHA-derived SPMs RvD1, RvD2 and MaR1 also reversed the stimulatory effect of TNF-α on chemerin production in human adipocytes.


Subject(s)
Adipocytes/drug effects , Chemokines/metabolism , Fatty Acids, Omega-3/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cells, Cultured , Chemokines/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Receptors, G-Protein-Coupled/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Int J Obes (Lond) ; 42(3): 572-579, 2018 03.
Article in English | MEDLINE | ID: mdl-28895586

ABSTRACT

BACKGROUND/OBJECTIVES: The aim of this study was to characterize the effects of Maresin 1 (MaR1) in obesity-related liver steatosis and the mechanisms involved. METHODS: MaR1 effects on fatty liver disease were tested in ob/ob (2-10 µg kg-1 i.p., 20 days) and in diet-induced obese (DIO) mice (2 µg kg-1, i.p., or 50 µg kg-1, oral gavage for 10 days), as well as in cultured hepatocytes. RESULTS: In ob/ob mice, MaR1 reduced liver triglycerides (TG) content, fatty acid synthase (FAS) and stearoyl-CoA desaturase-1 protein expression, while increased acetyl-CoA carboxylase (ACC) phosphorylation and LC3II protein expression, in parallel with a drop in p62 levels. Similar effects on hepatic TG, ACC phosphorylation, p62 and LC3II were observed in DIO mice after MaR1 i.p. injection. Interestingly, oral gavage of MaR1 also decreased serum transaminases, reduced liver weight and TG content. MaR1-treated mice exhibited reduced hepatic lipogenic enzymes content (FAS) or activation (by phosphorylation of ACC), accompanied by upregulation of carnitine palmitoyltransferase (Cpt1a), acyl-coenzyme A oxidase (Acox1) and autophagy-related proteins 5 and 7 (Atg5-7) gene expression, along with increased number of autophagic vacuoles and reduced p62 protein levels. MaR1 also induced AMP-activated protein kinase (AMPK) phosphorylation in DIO mice and in primary hepatocytes, and AMPK inhibition completely blocked MaR1 effects on Cpt1a, Acox1, Atg5 and Atg7 expression. CONCLUSIONS: MaR1 ameliorates liver steatosis by decreasing lipogenic enzymes, while inducing fatty acid oxidation genes and autophagy, which could be related to AMPK activation. Thus, MaR1 may be a new therapeutic candidate for reducing fatty liver in obesity.


Subject(s)
Docosahexaenoic Acids/pharmacology , Fatty Liver/metabolism , Liver , Obesity/metabolism , Animals , Body Weight/drug effects , Cells, Cultured , Diet, High-Fat , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese
3.
J Nutr Biochem ; 37: 76-82, 2016 11.
Article in English | MEDLINE | ID: mdl-27637001

ABSTRACT

Eicosapentaenoic acid (EPA), a n-3 long-chain polyunsaturated fatty acid, has been reported to have beneficial effects in obesity-associated metabolic disorders. The objective of the present study was to determine the effects of EPA on the regulation of genes involved in lipid metabolism, and the ability of EPA to induce mitochondrial biogenesis and beiging in subcutaneous adipocytes from overweight subjects. Fully differentiated human subcutaneous adipocytes from overweight females (BMI: 28.1-29.8kg/m2) were treated with EPA (100-200 µM) for 24 h. Changes in mRNA expression levels of genes involved in lipogenesis, fatty acid oxidation and mitochondrial biogenesis were determined by qRT-PCR. Mitochondrial content was evaluated using MitoTracker® Green stain. The effects on peroxisome proliferator-activated receptor gamma, co-activator 1 alpha (PGC-1α) and AMP-activated protein kinase (AMPK) were also characterized. EPA down-regulated lipogenic genes expression while up-regulated genes involved in fatty acid oxidation. Moreover, EPA-treated adipocytes showed increased mitochondrial content, accompanied by an up-regulation of nuclear respiratory factor-1, mitochondrial transcription factor A and cytochrome c oxidase IV mRNA expression. EPA also promoted the activation of master regulators of mitochondrial biogenesis such as sirtuin 1, PGC1-α and AMPK. In parallel, EPA induced the expression of genes that typify beige adipocytes such as fat determination factor PR domain containing 16, uncoupling protein 1 and cell death-inducing DFFA-like effector A, T-Box protein 1 and CD137. Our results suggest that EPA induces a remodeling of adipocyte metabolism preventing fat storage and promoting fatty acid oxidation, mitochondrial biogenesis and beige-like markers in human subcutaneous adipocytes from overweight subjects.


Subject(s)
Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Eicosapentaenoic Acid/metabolism , Gene Expression Regulation, Enzymologic , Mitochondrial Dynamics , Organelle Biogenesis , Subcutaneous Fat, Abdominal/metabolism , Acyl-CoA Oxidase/chemistry , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Adipocytes, Beige/enzymology , Adipocytes, Beige/pathology , Adipocytes, White/enzymology , Adipocytes, White/pathology , Adipogenesis , Biomarkers/metabolism , Body Mass Index , Carnitine O-Palmitoyltransferase/chemistry , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Energy Metabolism , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Female , Humans , Lipid Metabolism , Osmolar Concentration , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Subcutaneous Fat, Abdominal/enzymology , Subcutaneous Fat, Abdominal/pathology
4.
J Physiol Biochem ; 71(3): 547-58, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25820474

ABSTRACT

Irisin is a myokine/adipokine with potential role in obesity and diabetes. The objectives of the present study were to analyse the relationship between irisin and glucose metabolism at baseline and during an oral glucose tolerance test (OGTT) and to determine the effects of eicosapentaenoic acid (EPA) and/or α-lipoic acid treatment on irisin production in cultured human adipocytes and in vivo in healthy overweight/obese women following a weight loss program. Seventy-three overweight/obese women followed a 30% energy-restricted diet supplemented without (control) or with EPA (1.3 g/day), α-lipoic acid (0.3 g/day) or both EPA + α-lipoic acid (1.3 + 0.3 g/day) during 10 weeks. An OGTT was performed at baseline. Moreover, human adipocytes were treated with EPA (100-200 µM) or α-lipoic acid (100-250 µM) during 24 h. At baseline plasma, irisin circulating levels were positively associated with glucose levels; however, serum irisin concentrations were not affected by the increment in blood glucose or insulin during the OGTT. Treatment with α-lipoic acid (250 µM) upregulated Fndc5 messenger RNA (mRNA) and irisin secretion in cultured adipocytes. In overweight/obese women, irisin circulating levels decreased significantly after weight loss in all groups, while no additional differences were induced by EPA or α-lipoic acid supplementation. Moreover, plasma irisin levels were positively associated with higher glucose concentrations at beginning and at endpoint of the study. The data from the OGTT suggest that glucose is not a direct contributing factor of irisin release. The higher irisin levels observed in overweight/obese conditions could be a protective response of organism to early glucose impairments.


Subject(s)
Eicosapentaenoic Acid/administration & dosage , Fibronectins/blood , Glucose/metabolism , Obesity/blood , Thioctic Acid/administration & dosage , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Adult , Blood Glucose , Caloric Restriction , Cells, Cultured , Double-Blind Method , Female , Humans , Middle Aged , Obesity/diet therapy , Subcutaneous Fat/pathology , Treatment Outcome , Weight Loss , Young Adult
5.
J Physiol Biochem ; 65(1): 43-50, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19588730

ABSTRACT

Several studies have suggested that oxidative stress might cause and aggravate the inflammatory state associated with obesity and could be the link between excessive weight gain and its related disorders such as insulin resistance and cardiovascular diseases. Thus, antioxidant treatment has been proposed as a therapy to prevent and manage obesity and associated complications. Therefore, the aim of the present study was to investigate the effects of supplementation of a standard or high fat diet with the antioxidant lipoic acid (LA) during 56 days, on body weight gain, adiposity, feed efficiency and intestinal sugar absorption, in male Wistar rats. LA supplementation induced a lower body weight gain and adipose tissue size in both control or high fat fed rats accompanied by a reduction in food intake. The group fed on a high fat diet and treated with LA (OLIP group) showed a lower body weight gain than its corresponding Pair-Fed (PF) group (P < 0.05), which received the same amount of food than LA-treated animals but with no LA. In fact, LA induced a reduction on feed efficiency and also significantly decreased intestinal alpha-methylglucoside (alpha-MG) absorption both in lean and obese rats. These results suggest that the beneficial effects of dietary supplementation with LA on body weight gain are mediated, at least in part, by the reduction observed in food intake and feed efficiency. Furthemore, the inhibitory action of LA on intestinal sugar transport could explain in part the lower feed efficiency observed in LA-treated animals and therefore, highlighting the beneficial effects of LA on obesity.


Subject(s)
Carbohydrate Metabolism/drug effects , Dietary Fats/pharmacology , Intestinal Absorption/drug effects , Thioctic Acid/pharmacology , Weight Gain/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Eating , Male , Organ Size , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...