Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Obes (Lond) ; 48(7): 964-972, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38459259

ABSTRACT

BACKGROUND/OBJECTIVES: Proinflammatory cytokines are increased in obese adipose tissue, including inflammasome key masters. Conversely, IL-18 protects against obesity and metabolic dysfunction. We focused on the IL-18 effect in controlling adipose tissue remodeling and metabolism. MATERIALS/SUBJECTS AND METHODS: We used C57BL/6 wild-type (WT) and interleukine-18 deficient (IL-18-/-) male mice fed a chow diet and samples from bariatric surgery patients. RESULTS: IL-18-/- mice showed increased adiposity and proinflammatory cytokine levels in adipose tissue, leading to glucose intolerance. IL-18 was widely secreted by stromal vascular fraction but not adipocytes from mice's fatty tissue. Chimeric model experiments indicated that IL-18 controls adipose tissue expansion through its presence in tissues other than bone marrow. However, IL-18 maintains glucose homeostasis when present in bone marrow cells. In humans with obesity, IL-18 expression in omental tissue was not correlated with BMI or body fat mass but negatively correlated with IRS1, GLUT-4, adiponectin, and PPARy expression. Also, the IL-18RAP receptor was negatively correlated with IL-18 expression. CONCLUSIONS: IL-18 signaling may control adipose tissue expansion and glucose metabolism, as its absence leads to spontaneous obesity and glucose intolerance in mice. We suggest that resistance to IL-18 signaling may be linked with worse glucose metabolism in humans with obesity.


Subject(s)
Adipose Tissue , Interleukin-18 , Mice, Inbred C57BL , Obesity , Animals , Interleukin-18/metabolism , Mice , Male , Adipose Tissue/metabolism , Humans , Obesity/metabolism , Glucose Intolerance/metabolism , Disease Models, Animal , Mice, Knockout
2.
Nutrition ; 58: 167-174, 2019 02.
Article in English | MEDLINE | ID: mdl-30502680

ABSTRACT

OBJECTIVE: Chia seed oil is the richest source of plant-based ω-3 fatty acid, α-linolenic acid, but its potential and mechanisms of action to treat obesity are unclear. The aim of the study was to evaluate the effects of chia oil (ChOi) supplementation on body composition and insulin signaling in skeletal muscles of obese mice. METHODS: Male C57 BL/6 mice (n = 8/group) were fed regular control chow or a high-fat diet (HFD) for 135 d. Another HFD group additionally received ChOi from 90 to 135 d. RESULTS: Consumption of ChOi reduced fat mass accumulation and increased lean mass as evidenced by nuclear magnetic resonance. Moreover, obese mice treated with ChOi showed higher tyrosine phosphorylation of insulin receptor substrate 1, greater activation of protein kinase B, and increased translocation of glucose transporter type 4 in skeletal muscle tissue in response to insulin. ChOi supplementation improved glucose levels and insulin tolerance; decreased serum insulin, leptin, and triacylglycerols; and increased blood high-density lipoprotein cholesterol levels. All these effects caused by the use of ChOi seemed to be independent of the resolution of inflammation because the markers of inflammation were not altered in animals fed the HFD. CONCLUSION: The molecular effects observed in muscle tissue together with changes in body composition may have contributed to the increased glucose tolerance and to the healthy phenotype presented by obese animals treated with ChOi.


Subject(s)
Body Composition/drug effects , Dietary Supplements , Insulin/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Plant Oils/pharmacology , Salvia , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/drug effects , Signal Transduction/drug effects
3.
J Control Release ; 281: 11-18, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29753960

ABSTRACT

Nanoparticles have specific features (lipophilicity, surface charge, composition and size). Studies regarding the biological behavior of nanoparticles in diseases such diabetics and obesity are scarce. Here, we evaluated two nanoparticles: magnetic core mesoporous silica (MSN) (58 nm) and polycaprolactone (PCL) nanoparticle (280 nm) in obese mice. Changes in the biodistribution were observed, especially considering the mononuclear phagocyte system (MPS), and the visceral fat tissue. Nonetheless, our data corroborates the influence of size in the biodistribution in obese animals, supporting that smaller nanoparticles, may show a higher tissue deposition at spleen, due the associated splenomegaly and the complications arising from this state. Finally, our study demonstrated that, in obesity, probably due the low-grade inflammatory state associated with metabolic syndrome a difference in accumulation of nanoparticles was found, with profound impact in the tissue deposition of nanoparticles.


Subject(s)
Magnetite Nanoparticles/chemistry , Obesity/metabolism , Polyesters/chemistry , Silicon Dioxide/chemistry , Animals , Intra-Abdominal Fat/metabolism , Magnetic Resonance Imaging/methods , Magnetics , Male , Mice, Inbred C57BL , Mononuclear Phagocyte System/metabolism , Porosity , Tissue Distribution
4.
Nutrients ; 10(2)2018 Feb 16.
Article in English | MEDLINE | ID: mdl-29462922

ABSTRACT

Adolescent pregnancy increases risk of adverse perinatal outcomes. Placental delivery of long-chain polyunsaturated fatty acids (LCPUFA) is essential for fetal growth and development. In this pilot study, we aimed to assess maternal and fetal status of fatty acids (FA) measured at birth and the expression of key genes involved in FA uptake, transport and metabolism in the placenta of fifteen adolescents and fifteen adults. FA were quantified by gas-liquid chromatography. Placental expression of FA transporters was assessed by quantitative real-time polymerase chain reaction (qRT-PCR) and peroxisome proliferator-activated receptor gamma (PPARγ) was quantified by Western Blot. Adolescents had lower docosahexaenoic acid (DHA, 22:6 n-3) and total n-3 FA levels in maternal erythrocytes and placenta, but these were not different in fetal erythrocytes. Arachidonic acid (AA, 20:4 n-6) concentration was increased in placenta but lower in fetal circulation. Plasma membrane fatty acid binding protein (FABPpm) and fatty acid transport protein (FATP) 4 mRNA expressions were not different, however FATP1, fatty acid translocase (FAT/CD36) and fatty acid binding protein 3 (FABP3) mRNA and PPARγ protein levels were decreased in placenta of adolescents. Despite significant downregulation of FATP1, CD36 and FABP3, there was only a modest decrease in LCPUFA (10%) and AA (12%) and no difference in DHA content in cord blood, suggesting that FA transfer to the fetus was partially protected by other factors in adolescents from this cohort.


Subject(s)
Fatty Acids, Unsaturated/blood , Placenta/metabolism , Placental Circulation , Pregnancy in Adolescence , Adolescent , Adult , Age Factors , Biological Transport , Blotting, Western , Cross-Sectional Studies , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Female , Gas Chromatography-Mass Spectrometry , Humans , PPAR gamma/metabolism , Pilot Projects , Pregnancy , Real-Time Polymerase Chain Reaction , Young Adult
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 139-151, 2017 01.
Article in English | MEDLINE | ID: mdl-27677832

ABSTRACT

Macrophage infiltration into adipose tissue (AT) is a hallmark of the chronic inflammatory response in obesity and is supported by an intense monocyte migration towards AT. Although it has been detected an increased proportion of circulating CD16+ monocyte subsets in obese subjects, the mechanisms underlying this effect and the contribution of these cells to the inflamed profile of obese AT are still poorly understood. We investigated whether factors secreted by human obese omental AT could polarize monocytes to CD16+ enriched phenotype, and how these changes could modify their migratory capacity towards adipose tissue itself. We show that explants of human obese omental AT, obtained during bariatric surgery, released higher levels of MIP1-α, TNFα, leptin and also VEGF, together with increasing amounts of microparticles (MP), when compared to explants of lean subcutaneous AT. A higher content of circulating MP derived from preadipocytes and leukocytes was also detected in plasma of obese subjects. Conditioned media or MP released from obese omental AT increased CD16 and CCR5 expression on CD14+CD16- monocytes and augmented their migratory capacity towards the conditioned media from obese omental AT, itself. This effect was inhibited when MIP1-α was neutralized. Additionally, we demonstrate that MP derived from obese omental AT carry and transfer TLR8 to monocytes, thus triggering an increase in CD16 expression in those cells. Our data shows a positive feedback loop between blood monocytes and obese omental AT, which releases chemotactic mediators and TLR8-enriched MP, thus inducing an up-regulation of CD16+ monocytes, favoring leukocyte infiltration in the obese omental AT.


Subject(s)
Adipose Tissue/immunology , Cell-Derived Microparticles/immunology , Monocytes/immunology , Obesity/immunology , Receptors, CCR5/immunology , Receptors, IgG/immunology , Toll-Like Receptor 8/immunology , Adipose Tissue/pathology , Adult , Cell-Derived Microparticles/pathology , Female , GPI-Linked Proteins/analysis , GPI-Linked Proteins/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Male , Middle Aged , Monocytes/pathology , Obesity/pathology , Receptors, CCR5/analysis , Receptors, IgG/analysis , Toll-Like Receptor 8/analysis
6.
Obesity (Silver Spring) ; 24(12): 2522-2532, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27753270

ABSTRACT

OBJECTIVE: To investigate the role of obesity on the bone marrow microenvironment and evaluate its possible impact on the adipogenic potential of mesenchymal stem cells (MSC). METHODS: C57BL/6 male mice were fed with a high-fat diet (HFD) for 10 weeks. Femurs and tibiae were collected, and bone marrow mesenchymal stem cells (BM-MSC) were isolated and analyzed for proliferative potential, immunophenotype, and expression of adipogenesis markers. Their capacity to produce extracellular matrix proteins and proinflammatory cytokines in vitro was also evaluated. RESULTS: HFD mice presented a significant increase in bone marrow cellularity and higher tumor necrosis factor-α production in vitro. BM-MSC from HFD mice had higher proliferative capacity, produced more extracellular matrix proteins associated with adipogenesis, collagen I, and collagen IV, and showed increased constitutive expression of adipogenic markers, peroxisome proliferator-activated receptor-γ, and CCAAT/enhanced binding protein family-α, without changes in preadipocyte factor-1 expression. Incubation with adipocyte-differentiation medium induced further increase in CCAAT/enhanced binding protein family-α and augmented adiponectin expression in obese BM-MSC. These alterations did not result in increased adipogenic differentiation within the bone marrow. Moreover, BM-HSC from HFD mice, co-cultivated with BM-MSCs from lean mice, exerted paracrine effects on these cells, inducing augment of peroxisome proliferator-activated receptor-γ. CONCLUSIONS: The data suggest that obesity promotes an inflammatory microenvironment in bone marrow that commits BM-MSC to adipogenesis.


Subject(s)
Adipogenesis/physiology , Bone Marrow Cells/physiology , Mesenchymal Stem Cells/physiology , Obesity/physiopathology , Animals , Bone Marrow , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Cell Proliferation , Diet, High-Fat , Inflammation , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , PPAR gamma/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
7.
Nutrients ; 7(1): 335-48, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25569627

ABSTRACT

Hepcidin is a key hormone that induces the degradation of ferroportin (FPN), a protein that exports iron from reticuloendothelial macrophages and enterocytes. The aim of the present study was to experimentally evaluate if the obesity induced by a high-fat diet (HFD) modifies the expression of FPN in macrophages and enterocytes, thus altering the iron bioavailability. In order to directly examine changes associated with iron metabolism in vivo, C57BL/6J mice were fed either a control or a HFD. Serum leptin levels were evaluated. The hepcidin, divalent metal transporter-1 (DMT1), FPN and ferritin genes were analyzed by real-time polymerase chain reaction. The amount of iron present in both the liver and spleen was determined by flame atomic absorption spectrometry. Ferroportin localization within reticuloendothelial macrophages was observed by immunofluorescence microscopy. Obese animals were found to exhibit increased hepcidin gene expression, while iron accumulated in the spleen and liver. They also exhibited changes in the sublocation of splenic cellular FPN and a reduction in the FPN expression in the liver and the spleen, while no changes were observed in enterocytes. Possible explanations for the increased hepcidin expression observed in HFD animals may include: increased leptin levels, the liver iron accumulation or endoplasmic reticulum (ER) stress. Together, the results indicated that obesity promotes changes in iron bioavailability, since it altered the iron recycling function.


Subject(s)
Iron/blood , Obesity/blood , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Diet, High-Fat/adverse effects , Endoplasmic Reticulum Stress , Enterocytes/metabolism , Ferritins/genetics , Ferritins/metabolism , Gene Expression , Hepcidins/genetics , Hepcidins/metabolism , Iron/pharmacokinetics , Leptin/blood , Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Spectrophotometry, Atomic , Spleen/metabolism
8.
PLoS One ; 8(5): e64220, 2013.
Article in English | MEDLINE | ID: mdl-23675529

ABSTRACT

We investigated the consequences of mild maternal malnutrition in rat dams, in terms of thymocyte responses and the putative role of leptin. The young progeny of dams submitted to protein deprivation (PD) during lactation showed at 30 days of age lower body and thymus weights, significant alterations in CD4/CD8-defined T cell subsets without modifications in total thymocyte number as well as in proliferative response. Despite, the rats from PD group did not present alterations in leptin circulating levels, the expression of leptin receptor ObRb was enhanced in their thymocytes. This change was accompanied by an increase in leptin signaling response of thymocytes from PD rats, with an increase in JAK2 and STAT3 phosphorylation after leptin stimulation. Thymocytes from PD rats also presented a decreased rate of spontaneous apoptosis when compared to controls. Accordingly, higher expression of anti-apoptotic protein Bcl-2, and lower of pro-apoptotic protein Bax, with no change of pro-apoptotic Bad, and higher pro-caspase 3 content were detected in PD thymocytes. Moreover, thymocytes from PD group exhibited a constitutive higher nuclear content of p65 NF-kB associated to a lower IkB content in the cytoplasm. Finally, although there was no change in ob gene expression in PD thymocytes, a higher mRNA expression for the Ob gene was observed in the thymic microenvironment from PD animals. Taken together, the results show that mild maternal protein deprivation during lactation affects thymic homeostasis, enhancing leptin activity, which in turn protects thymocytes from apoptosis in the young progeny, with possible consequences upon the immune response of these animals in adult life.


Subject(s)
Apoptosis , Lactation , Leptin/metabolism , Protein-Energy Malnutrition , Thymocytes/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Body Weight , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Diet, Protein-Restricted , Female , Gene Expression Regulation , Immunophenotyping , Janus Kinase 2/metabolism , Leptin/blood , Leptin/genetics , Male , NF-kappa B/metabolism , Rats , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocyte Subsets/metabolism , Thymocytes/immunology , Thymus Gland/cytology
9.
Biol Trace Elem Res ; 149(1): 64-70, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22528770

ABSTRACT

Iron bioavailability seems to be regulated by vitamin A (VA) but the molecular events involved in this mechanism are not well understood. It is also known that retinoids mediate most of their function via interaction with retinoid receptors, which act as ligand-activated transcription factors controlling the expression of a number of target genes. Here, we evaluated the VA effects on the modulation of the levels of mRNA encoding proteins involved in the iron bioavailability, whether in the intestinal absorption process or in the liver iron metabolism. The expression of genes involved in iron intestinal absorption (divalent metal transporter 1, duodenal cytochrome B, ferroportin 1 FPN1, and ferritin) were evaluated in vitro by treating Caco-2 cells with retinoic acid or in vivo by observing the effects of vitamin A deficiency (VAD) in BALB/C mice. Liver hepcidin and ferritin mRNA levels were upregulated by VAD; however, this condition did not promote any change on the expression of those genes that participate in the iron absorption. Moreover, data from the in vitro analysis showed that VA induced FPN1 gene expression by a hepcidin-independent manner. Therefore, the in vivo results support the idea that VAD may not affect iron absorption but would rather affect iron mobilization mechanisms. On the other hand, our results using Caco-2 cells raises the possibility that VA addition to intestinal epithelium may improve iron absorption through the induction of FPN1 gene expression.


Subject(s)
Cation Transport Proteins/metabolism , Gene Expression Regulation , Intestinal Mucosa/metabolism , Iron/metabolism , Vitamin A/analogs & derivatives , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Caco-2 Cells , Cation Transport Proteins/genetics , Dietary Supplements , Diterpenes , Drug Evaluation, Preclinical , Duodenum/drug effects , Duodenum/metabolism , Ferritins/genetics , Ferritins/metabolism , Hepcidins , Humans , Intestinal Absorption , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinyl Esters , Tretinoin/pharmacology , Vitamin A/administration & dosage , Vitamin A/pharmacology , Vitamin A Deficiency/genetics , Vitamin A Deficiency/metabolism
10.
Am J Physiol Endocrinol Metab ; 295(3): E626-36, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18559980

ABSTRACT

Epidemiological and experimental studies have demonstrated that early postnatal nutrition has been associated with long-term effects on glucose homeostasis in adulthood. Recently, our group demonstrated that undernutrition during early lactation affects the expression and activation of key proteins of the insulin signaling cascade in rat skeletal muscle during postnatal development. To elucidate the molecular mechanisms by which undernutrition during early life leads to changes in insulin sensitivity in peripheral tissues, we investigated the insulin signaling in adipose tissue. Adipocytes were isolated from epididymal fat pads of adult male rats that were the offspring of dams fed either a normal or a protein-free diet during the first 10 days of lactation. The cells were incubated with 100 nM insulin before the assays for immunoblotting analysis, 2-deoxyglucose uptake, immunocytochemistry for GLUT4, and/or actin filaments. Following insulin stimulation, adipocytes isolated from undernourished rats presented reduced tyrosine phosphorylation of IR and IRS-1 and increased basal phosphorylation of IRS-2, Akt, and mTOR compared with controls. Basal glucose uptake was increased in adipocytes from the undernourished group, and the treatment with LY294002 induced only a partial inhibition both in basal and in insulin-stimulated glucose uptake, suggesting an involvement of phosphoinositide 3-kinase activity. These alterations were accompanied by higher GLUT4 content in the plasma membrane and alterations in the actin cytoskeleton dynamics. These data suggest that early postnatal undernutrition impairs insulin sensitivity in adulthood by promoting changes in critical steps of insulin signaling in adipose tissue, which may contribute to permanent changes in glucose homeostasis.


Subject(s)
Adipocytes/metabolism , Diet, Protein-Restricted , Glucose Transporter Type 4/metabolism , Lactation/physiology , Oncogene Protein v-akt/metabolism , Protein Kinases/metabolism , Adipocytes/physiology , Adipose Tissue/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Separation , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Female , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Immunohistochemistry , Insulin/metabolism , Insulin/pharmacology , Morpholines/pharmacology , Phosphorylation , Protein Transport , Rats , Rats, Wistar , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Thiazolidines/pharmacology , Tyrosine/metabolism
11.
Int J Exp Pathol ; 87(5): 373-81, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16965565

ABSTRACT

Light cigarette (LC) exposure is supposed to be less hazardous with a decreased incidence of cancer and tobacco-associated diseases. C57BL/6 mouse groups were subjected to smoke from 3, 6 or 12 LC for 60 days and compared with mice exposed to ambient air (EAA) in order to study lung injury by morphometrical and biochemical methods. Bronchoalveolar lavage (BAL) analysis and histology and stereology were performed. Tissue from the right lung was used for measuring thiobarbituric acid reactive substances (TBARS) and Western blot analysis. One way anova was performed followed by the Student-Newman Keuls post-test (P < 0.05). The cellular content of BAL was 95% alveolar macrophages in all groups except in mice exposed to 3 LC, where 23% neutrophils were observed. Emphysema was not observed in three and 6 LC, but it was found in 12 LC parallel to increased volume density (Vv) of airspaces from 61.0 +/- 0.6 (EAA) to 80.9 +/- 1.0 (12 LC) and decreased Vv of elastic fibres from 17.8 +/- 0.9 (EAA) to 11.8 +/- 0.6 (12 LC). All exposed groups to LC showed low TBARS levels compared with mice EAA. Lung tissue from animals exposed to 12 LC showed decreased tissue inhibitor of metalloprotease-2 and increased matrix metalloprotease-12 detection, which suggests an imbalance in extracellular matrix (ECM). Increased tumour necrosis factor-alpha and nuclear factor-kappaB detection were observed in exposed groups to LC when compared with mice EAA. The data suggest that LC is so dangerous to lungs as full-flavour cigarettes inducing ECM imbalance and emphysema.


Subject(s)
Lung/metabolism , NF-kappa B/metabolism , Pulmonary Emphysema/etiology , Tobacco Smoke Pollution/adverse effects , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Lipid Peroxidation , Lung/pathology , Macrophages, Alveolar/pathology , Matrix Metalloproteinase 12 , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Oxidative Stress , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Tissue Inhibitor of Metalloproteinase-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...