Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Publication year range
1.
Am J Physiol Endocrinol Metab ; 318(5): E590-E599, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31891536

ABSTRACT

Besides a therapeutic target for type 2 diabetes, dipeptidyl peptidase 4 (DPP4) is an adipokine potentially upregulated in human obesity. We aimed to explore the role of adipocyte-derived DPP4 in diet-induced obesity and insulin resistance with an adipose tissue-specific knockout (AT-DPP4-KO) mouse. Wild-type and AT-DPP4-KO mice were fed for 24 wk with a high fat diet (HFD) and characterized for body weight, glucose tolerance, insulin sensitivity by hyperinsulinemic-euglycemic clamp, and body composition and hepatic fat content. Image and molecular biology analysis of inflammation, as well as adipokine secretion, was performed in AT by immunohistochemistry, Western blot, real-time-PCR, and ELISA. Incretin levels were determined by Luminex kits. Under HFD, AT-DPP4-KO displayed markedly reduced circulating DPP4 concentrations, proving AT as a relevant source. Independently of glucose-stimulated incretin hormones, AT-DPP4-KO had improved glucose tolerance and hepatic insulin sensitivity. AT-DPP4-KO displayed smaller adipocytes and increased anti-inflammatory markers. IGF binding protein 3 (IGFBP3) levels were lower in AT and serum, whereas free IGF1 was increased. The absence of adipose DPP4 triggers beneficial AT remodeling with decreased production of IGFBP3 during HFD, likely contributing to the observed, improved hepatic insulin sensitivity.


Subject(s)
Adipose Tissue/metabolism , Dipeptidyl Peptidase 4/metabolism , Insulin Resistance/physiology , Liver/metabolism , Obesity/metabolism , Adipocytes/metabolism , Adipokines/metabolism , Animals , Body Weight , Diet, High-Fat/adverse effects , Dipeptidyl Peptidase 4/genetics , Immunohistochemistry , Insulin/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Male , Mice , Obesity/etiology , Obesity/genetics
2.
Biochem Biophys Res Commun ; 471(3): 348-54, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26872429

ABSTRACT

DPP4 is a ubiquitously expressed cell surface protease which is also released to the circulation as soluble DPP4 (sDPP4). Recently, we identified DPP4 as a novel adipokine oversecreted in obesity and thus potentially linking obesity to the metabolic syndrome. Furthermore, sDPP4 impairs insulin signaling in an autocrine and paracrine fashion in different cell types. However, it is still unknown which functional role DPP4 might play in adipocytes. Therefore, primary human adipocytes were treated with a specific DPP4 siRNA. Adipocyte differentiation was not affected by DPP4 silencing. Interestingly, DPP4 reduction improved insulin responsiveness of adipocytes at the level of insulin receptor, proteinkinase B (Akt) and Akt substrate of 160 kDa. To investigate whether the observed effects could be attributed to the enzymatic activity of DPP4, human adipocytes were treated with the DPP4 inhibitors sitagliptin and saxagliptin. Our data show that insulin-stimulated activation of Akt is augmented by DPP4 inhibitor treatment. Based on our previous observation that sDPP4 induces insulin resistance in adipocytes, and that adipose DPP4 levels are higher in obese insulin-resistant patients, we now suggest that the abundance of DPP4 might be a regulator of adipocyte insulin signaling.


Subject(s)
Adipocytes/cytology , Adipocytes/physiology , Dipeptidyl Peptidase 4/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Signal Transduction/physiology , Adamantane/administration & dosage , Adamantane/analogs & derivatives , Cell Differentiation/physiology , Cells, Cultured , Dipeptides/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Enzyme Activation , Humans , Hypoglycemic Agents/administration & dosage , Sitagliptin Phosphate/administration & dosage
3.
Biochim Biophys Acta ; 1842(9): 1613-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24928308

ABSTRACT

DPP4 is an ubiquitously expressed cell-surface protease that is shedded to the circulation as soluble DPP4 (sDPP4). We recently identified sDPP4 as a novel adipokine potentially linking obesity to the metabolic syndrome. The aim of this study was to investigate direct effects of sDPP4 on human vascular smooth muscle cells (hVSMCs) and to identify responsible signaling pathways. Using physiological concentrations of sDPP4, we could observe a concentration-dependent activation of ERK1/2 (3-fold) after 6h, which remained stable for up to 24h. Additionally, sDPP4 treatment induced a 1.5-fold phosphorylation of the NF-κB subunit p65. In accordance with sDPP4-induced stress and inflammatory signaling, sDPP4 also stimulates hVSMC proliferation. Furthermore we could observe an increased expression and secretion of pro-inflammatory cytokines like interleukin (IL)-6, IL-8 and MCP-1 (2.5-, 2.4- and 1.5-fold, respectively) by the sDPP4 treatment. All direct effects of sDPP4 on signaling, proliferation and inflammation could completely be prevented by DPP4 inhibition. Bioinformatic analysis and signaling signature induced by sDPP4 suggest that sDPP4 might be an agonist for PAR2. After the silencing of PAR2, the sDPP4-induced ERK activation as well as the proliferation was totally abolished. Additionally, the sDPP4-induced upregulation of IL-6 and IL-8 could completely be prevented by the PAR2 silencing. In conclusion, we show for the first time that sDPP4 directly activates the MAPK and NF-κB signaling cascade involving PAR2 and resulting in the induction of inflammation and proliferation of hVSMC. Thus, our in vitro data might extend the current view of sDPP4 action and shed light on cardiovascular effects of DPP4-inhibitors.


Subject(s)
Cell Proliferation , Dipeptidyl Peptidase 4/metabolism , Inflammation/pathology , Muscle, Smooth, Vascular/pathology , Receptor, PAR-2/metabolism , Amino Acid Sequence , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dipeptides/pharmacology , Dipeptidyl Peptidase 4/genetics , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/genetics , Inflammation/metabolism , Isoxazoles/pharmacology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
4.
Biochim Biophys Acta ; 1843(4): 780-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24418043

ABSTRACT

Monocyte chemoattractant protein-induced protein 1 (MCPIP1) encoded by the ZC3H12a gene (also known as Regnase-1) is involved in the regulation of degradation of mRNA of inflammatory modulators and for processing of pre-miRNA. These functions depend on the presence of the PIN domain. Moreover, MCPIP1 was described as a negative regulator of NF-κB and AP-1 signaling pathways although mechanisms underlying such activity remain unknown. We aimed at determining the role of MCPIP1 in adipogenesis. Here, we present evidence that Mcpip1 transcription is transiently activated during 3T3-L1 transition from pre- to adipocytes. However Mcpip1 protein expression is also strongly decreased at day one after induction of adipogenesis. Knockdown of Mcpip1 results in an upregulation of C/EBPß and PPARγ mRNAs, whereas overexpression of MCPIP1 reduces the level of both transcription factors and impairs adipogenesis. MCPIP1-dependend modulation of C/EBPß and PPARγ levels results in a modulation of the expression of downstream controlled genes. In addition, decreased C/EBPß, but not PPARγ, depends on the activity of the MCPIP1 PIN domain, which is responsible for RNase properties of this protein. Together, these data confirm that MCPIP1 is a key regulator of adipogenesis.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Ribonucleases/genetics , Transcription, Genetic , 3T3-L1 Cells , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Cell Differentiation , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Mice , PPAR gamma/biosynthesis , Signal Transduction
5.
Biochim Biophys Acta ; 1842(2): 275-83, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24333576

ABSTRACT

Cardiovascular complications are common in patients with type 2 diabetes. Adipokines have been implicated in the induction of proliferative and pro-atherogenic alterations in human vascular smooth muscle cells (hVSMC). Other reports demonstrated the importance of the miRNA cluster miR-143/145 in the regulation of VSMC homeostasis and insulin sensitivity. Here we investigated whether the detrimental effects of adipokines on hVSMC function could be ascribed to alterations in miR-143/145 expression. The exposure of hVSMC to conditioned media (CM) from primary human subcutaneous adipocytes increased the expression of smooth muscle α-actin (SMA), and the miR-143/145 cluster, but markedly impaired the insulin-mediated phosphorylation of Akt and its substrate endothelial nitric oxide synthase (eNOS). Furthermore, CM promoted the phosphorylation of SMAD2 and p38, which have both been linked to miR-143/145 induction. Accordingly, the induction of miR-143/145 as well as the inhibition of insulin-mediated Akt- and eNOS-phosphorylation was prevented when hVSMC were treated with pharmacological inhibitors for Alk-4/5/7 and p38 before the addition of CM. The transfection of hVSMC with precursor miR-143, but not with precursor miR-145, resulted in impaired insulin-mediated phosphorylation of Akt and eNOS. This inhibition of insulin signaling by CM and miR-143 is associated with a reduction in the expression of the oxysterol-binding protein-related protein 8 (ORP8). Finally, the knock-down of ORP8 resulted in impaired insulin-mediated phosphorylation of Akt in hVSMC. Thus, the detrimental effects of adipocyte-derived conditioned media on insulin action in primary hVSMC can be ascribed to the Alk- and p38-dependent induction of miR-143 and subsequent downregulation of ORP8.


Subject(s)
Adipocytes/metabolism , Culture Media, Conditioned/pharmacology , Insulin/pharmacology , MicroRNAs/genetics , Myocytes, Smooth Muscle/drug effects , Adipocytes/cytology , Adult , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Culture Media, Conditioned/metabolism , Female , HEK293 Cells , Humans , Insulin/metabolism , Middle Aged , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , RNA Interference , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Smad2 Protein/genetics , Smad2 Protein/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Mol Cell Proteomics ; 11(1): M111.010504, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21947364

ABSTRACT

Adipose tissue is a major endocrine organ, releasing signaling and mediator proteins, termed adipokines, via which adipose tissue communicates with other organs. Expansion of adipose tissue in obesity alters adipokine secretion, which may contribute to the development of metabolic diseases. Although recent profiling studies have identified numerous adipokines, the amount of overlap from these studies indicates that the adipokinome is still incompletely characterized. Therefore, we conducted a complementary protein profiling on concentrated conditioned medium derived from primary human adipocytes. SDS-PAGE/liquid chromatography-electrospray ionization tandem MS and two-dimensional SDS-PAGE/matrix-assisted laser desorption ionization/time of flight MS identified 347 proteins, 263 of which were predicted to be secreted. Fourty-four proteins were identified as novel adipokines. Furthermore, we validated the regulation and release of selected adipokines in primary human adipocytes and in serum and adipose tissue biopsies from morbidly obese patients and normal-weight controls. Validation experiments conducted for complement factor H, αB-crystallin, cartilage intermediate-layer protein, and heme oxygenase-1 show that the release and expression of these factors in adipocytes is regulated by differentiation and stimuli, which affect insulin sensitivity, as well as by obesity. Heme oxygenase-1 especially reveals to be a novel adipokine of interest. In vivo, circulating levels and adipose tissue expression of heme oxygenase-1 are significantly increased in obese subjects compared with lean controls. Collectively, our profiling study of the human adipokinome expands the list of adipokines and further highlights the pivotal role of adipokines in the regulation of multiple biological processes within adipose tissue and their potential dysregulation in obesity.


Subject(s)
Adipocytes/metabolism , Adipokines/metabolism , Adipokines/blood , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adult , Cells, Cultured , Chromatography, Liquid , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Profiling , Heme Oxygenase-1/metabolism , Humans , Male , Obesity/metabolism , Proteome , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/metabolism , Young Adult
7.
Mediators Inflamm ; 2013: 982458, 2013.
Article in English | MEDLINE | ID: mdl-23935253

ABSTRACT

Adipose tissue secrets adipokines and fatty acids, which may contribute to obesity-associated vascular dysfunction and cardiovascular risk. This study investigated which factors are responsible for the synergistic effect of adipokine and oleic acid- (OA-) induced proliferation of human vascular smooth muscle cells (VSMC). Adipocyte-conditioned medium (CM) from human adipocytes induces proliferation of VSMC in correlation to its vascular endothelial growth factor (VEGF) content. CM increases VEGF-receptor (VEGF-R) 1 and 2 expression and VEGF secretion of VSMC, while OA only stimulates VEGF secretion. VEGF neutralization abrogates CM- and OA-induced proliferation and considerably reduces proliferation induced by CM and OA in combination. VEGF release is higher from visceral adipose tissue (VAT) of obese subjects compared to subcutaneous adipose tissue (SAT) and VAT from lean controls. Furthermore, VEGF release from VAT correlates with its proliferative effect. Perivascular adipose tissue (PAT) from type 2 diabetic patients releases significantly higher amounts of VEGF and induces stronger proliferation of VSMC as compared to SAT and SAT/PAT of nondiabetics. In conclusion, VEGF is mediating CM-induced proliferation of VSMC. As this adipokine is released in high amounts from VAT of obese patients and PAT of diabetic patients, VEGF might link adipose tissue inflammation to increased VSMC proliferation.


Subject(s)
Adipocytes/cytology , Adipose Tissue/metabolism , Intra-Abdominal Fat/metabolism , Myocytes, Smooth Muscle/cytology , Vascular Endothelial Growth Factor A/metabolism , Adipokines/metabolism , Adult , Biopsy , Cell Proliferation , Cells, Cultured , Coronary Vessels/pathology , Diabetes Mellitus, Type 2/metabolism , Female , Gene Expression Regulation , Humans , Inflammation , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Obesity/metabolism , Oleic Acid/chemistry , Overweight , Young Adult
8.
Am J Physiol Heart Circ Physiol ; 302(11): H2148-65, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22447947

ABSTRACT

Abdominal obesity is a major risk factor for cardiovascular disease, and recent studies highlight a key role of adipose tissue dysfunction, inflammation, and aberrant adipokine release in this process. An increased demand for lipid storage results in both hyperplasia and hypertrophy, finally leading to chronic inflammation, hypoxia, and a phenotypic change of the cellular components of adipose tissue, collectively leading to a substantially altered secretory output of adipose tissue. In this review we have assessed the adipo-vascular axis, and an overview of adipokines associated with cardiovascular disease is provided. This resulted in a first list of more than 30 adipokines. A deeper analysis only considered adipokines that have been reported to impact on inflammation and NF-κB activation in the vasculature. Out of these, the most prominent link to cardiovascular disease was found for leptin, TNF-α, adipocyte fatty acid-binding protein, interleukins, and several novel adipokines such as lipocalin-2 and pigment epithelium-derived factor. Future work will need to address the potential role of these molecules as biomarkers and/or drug targets.


Subject(s)
Adipokines/physiology , Cardiovascular Diseases/physiopathology , Inflammation/physiopathology , Metabolic Diseases/physiopathology , Adipose Tissue/physiopathology , Animals , Humans , Models, Animal , NF-kappa B/physiology , Obesity/physiopathology , Rats
9.
Hepatology ; 53(5): 1504-14, 2011 May.
Article in English | MEDLINE | ID: mdl-21400557

ABSTRACT

UNLABELLED: Obesity-related hepatic steatosis is a major risk factor for metabolic and cardiovascular disease. Fat reduced hypocaloric diets are able to relieve the liver from ectopically stored lipids. We hypothesized that the widely used low carbohydrate hypocaloric diets are similarly effective in this regard. A total of 170 overweight and obese, otherwise healthy subjects were randomized to either reduced carbohydrate (n = 84) or reduced fat (n = 86), total energy restricted diet (-30% of energy intake before diet) for 6 months. Body composition was estimated by bioimpedance analyses and abdominal fat distribution by magnetic resonance tomography. Subjects were also submitted to fat spectroscopy of liver and oral glucose tolerance testing. In all, 102 subjects completed the diet intervention with measurements of intrahepatic lipid content. Both hypocaloric diets decreased body weight, total body fat, visceral fat, and intrahepatic lipid content. Subjects with high baseline intrahepatic lipids (>5.56%) lost ≈7-fold more intrahepatic lipids compared with those with low baseline values (<5.56%) irrespective of diet composition. In contrast, changes in visceral fat mass and insulin sensitivity were similar between subgroups, with low and high baseline intrahepatic lipids. CONCLUSION: A prolonged hypocaloric diet low in carbohydrates and high in fat has the same beneficial effects on intrahepatic lipid accumulation as the traditional low-fat hypocaloric diet. The decrease in intrahepatic lipids appears to be independent of visceral fat loss and is not tightly coupled with changes in whole body insulin sensitivity during 6 months of an energy restricted diet.


Subject(s)
Caloric Restriction , Diet, Fat-Restricted , Fatty Liver/diet therapy , Overweight/diet therapy , Adult , Diet, Carbohydrate-Restricted , Female , Humans , Male , Middle Aged , Obesity/diet therapy , Prospective Studies
10.
J Cell Mol Med ; 15(5): 1177-88, 2011 May.
Article in English | MEDLINE | ID: mdl-20518853

ABSTRACT

In the context of obesity, perivascular fat produces various adipokines and releases free fatty acids, which may induce inflammation and proliferation in the vascular wall. In this study we investigated how adipokines, oleic acid (OA) and the combined treatment regulate human vascular smooth muscle cell (hVSMC) proliferation and migration and the underlying signalling pathways. Adipocyte-conditioned media (CM) generated from human adipocytes induces a prominent proliferation and migration of hVSMC. Autocrine action of adiponectin totally abolishes CM-induced proliferation. Furthermore, OA but not palmitic acid induces proliferation of hVSMC. CM itself does not contain fatty acids, but CM in combination with OA markedly enhances proliferation of hVSMC in a synergistic way. Both the nuclear factor (NF)-κB and the mammalian target of rapamycin (mTOR) pathway were synergistically activated under these conditions and found to be essential for hVSMC proliferation. Expression of iNOS and production of nitric oxide was only enhanced by combined treatment inducing a marked release of VEGF. Combination of OA and VEGF induces an additive increase of hVSMC proliferation. We could show that the combination of CM and OA led to a synergistic proliferation of hVSMC. Expression of iNOS and production of nitric oxide were only enhanced under these conditions and were paralleled by a marked release of VEGF. These results suggest that the combined elevated release of fatty acids and adipokines by adipose tissue in obesity might be critically related to hVSMC dysfunction, vascular inflammation and the development of atherosclerosis.


Subject(s)
Adipokines/pharmacology , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/drug effects , Oleic Acid/pharmacology , Signal Transduction/drug effects , Adiponectin/metabolism , Adult , Cell Movement/drug effects , Cells, Cultured , Female , Humans , Inflammation/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
11.
Mol Metab ; 53: 101287, 2021 11.
Article in English | MEDLINE | ID: mdl-34224918

ABSTRACT

OBJECTIVE: Recent studies suggest that hypoxia exposure may improve glucose homeostasis, but well-controlled human studies are lacking. We hypothesized that mild intermittent hypoxia (MIH) exposure decreases tissue oxygen partial pressure (pO2) and induces metabolic improvements in people who are overweight/obese. METHODS: In a randomized, controlled, single-blind crossover study, 12 men who were overweight/obese were exposed to MIH (15 % O2, 3 × 2 h/day) or normoxia (21 % O2) for 7 consecutive days. Adipose tissue (AT) and skeletal muscle (SM) pO2, fasting/postprandial substrate metabolism, tissue-specific insulin sensitivity, SM oxidative capacity, and AT and SM gene/protein expression were determined. Furthermore, primary human myotubes and adipocytes were exposed to oxygen levels mimicking the hypoxic and normoxic AT and SM microenvironments. RESULTS: MIH decreased systemic oxygen saturation (92.0 ± 0.5 % vs 97.1 ± 0.3, p < 0.001, respectively), AT pO2 (21.0 ± 2.3 vs 36.5 ± 1.5 mmHg, p < 0.001, respectively), and SM pO2 (9.5 ± 2.2 vs 15.4 ± 2.4 mmHg, p = 0.002, respectively) compared to normoxia. In addition, MIH increased glycolytic metabolism compared to normoxia, reflected by enhanced fasting and postprandial carbohydrate oxidation (pAUC = 0.002) and elevated plasma lactate concentrations (pAUC = 0.005). Mechanistically, hypoxia exposure increased insulin-independent glucose uptake compared to standard laboratory conditions (~50 %, p < 0.001) and physiological normoxia (~25 %, p = 0.019) through AMP-activated protein kinase in primary human myotubes but not in primary human adipocytes. MIH upregulated inflammatory/metabolic pathways and downregulated extracellular matrix-related pathways in AT but did not alter systemic inflammatory markers and SM oxidative capacity. MIH exposure did not induce significant alterations in AT (p = 0.120), hepatic (p = 0.132) and SM (p = 0.722) insulin sensitivity. CONCLUSIONS: Our findings demonstrate for the first time that 7-day MIH reduces AT and SM pO2, evokes a shift toward glycolytic metabolism, and induces adaptations in AT and SM but does not induce alterations in tissue-specific insulin sensitivity in men who are overweight/obese. Future studies are needed to investigate further whether oxygen signaling is a promising target to mitigate metabolic complications in obesity. CLINICAL TRIAL REGISTRATION: This study is registered at the Netherlands Trial Register (NL7120/NTR7325).


Subject(s)
Adipose Tissue/metabolism , Hypoxia/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Overweight/metabolism , Adaptation, Physiological , Adult , Aged , Humans , Male , Middle Aged , Oxygen/metabolism
12.
J Cell Mol Med ; 14(9): 2223-34, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20716126

ABSTRACT

Obesity, insulin resistance and the metabolic syndrome, are characterized by expansion and inflammation of adipose tissue, including the depots surrounding the heart and the blood vessels. Epicardial adipose tissue (EAT) is a visceral thoracic fat depot located along the large coronary arteries and on the surface of the ventricles and the apex of the heart, whereas perivascular adipose tissue (PVAT) surrounds the arteries. Both fat depots are not separated by a fascia from the underlying tissue. Therefore, factors secreted from epicardial and PVAT, like free fatty acids and adipokines, can directly affect the function of the heart and blood vessels. In this review, we describe the alterations found in EAT and PVAT in pathological states like obesity, type 2 diabetes, the metabolic syndrome and coronary artery disease. Furthermore, we discuss how changes in adipokine expression and secretion associated with these pathological states could contribute to the pathogenesis of cardiac contractile and vascular dysfunction.


Subject(s)
Adipose Tissue/physiopathology , Arteries/physiopathology , Cardiovascular Diseases/physiopathology , Pericardium/physiopathology , Animals , Biomarkers/metabolism , Fatty Acids/metabolism , Humans
13.
Curr Opin Clin Nutr Metab Care ; 13(4): 366-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20473150

ABSTRACT

PURPOSE OF REVIEW: Obesity is associated with low-grade chronic inflammation in adipose tissue. This review presents an update on human and rodent studies analyzing the nature of fat-infiltrating immune cells, the time course of adipose tissue infiltration and underlying mechanisms. RECENT FINDINGS: Intensive studies in rodents have shown that not only cells of the innate immune system traffic into adipose tissue but also various lymphocytes of the adaptive immunity are involved in inflammatory processes in fat. Several studies also provide insight in the order of appearance of macrophages and lymphocytes during the onset of obesity. Adipocytes and preadipocytes are also active players by their secretion of chemotactic adipokines. SUMMARY: This review summarizes strong evidence for a link between the action of innate and adaptive immune systems in adipose tissue in the context of obesity and metabolism in rodents, but more studies in humans are necessary to relate this topic to human physiology. Targeting different immune cells at different stages of obesity may eventually lead to novel therapeutic approaches for the metabolic syndrome.


Subject(s)
Adipocytes/immunology , Adipogenesis/immunology , Adipose Tissue/immunology , Inflammation/immunology , Lymphocytes/metabolism , Macrophages/metabolism , Obesity/immunology , Adipokines/metabolism , Animals , Humans , Immunity
14.
Biochim Biophys Acta Proteins Proteom ; 1867(12): 140172, 2019 12.
Article in English | MEDLINE | ID: mdl-30502511

ABSTRACT

Adipose tissue is an endocrine organ, secreting various adipokines, either directly or via extracellular vesicles, including exosomes. Exosomes are vesicles of 40-150 nm size that represent a novel concept of biomolecule release. We purified exosomes from isolated primary human preadipocytes differentiated to mature adipocytes. The analyses of these exosomal preparations by LC-MS identified 884 proteins, so called exoadipokines. The comparison of exoadipokines with previously identified human exosome-associated proteins in ExoCarta database show an overlap of 817 proteins, but also revealed 67 proteins not assigned to human exosomes, yet. We further compared all exoadipokines to our previously reported reference secretome of human adipose tissue (http://diabesityprot.org/), finding 212 common proteins, whereas 672 proteins were specific for the exosomal fraction. Bioinformatic analyses revealed that the 212 common proteins can be assigned to all major functions of adipose tissue secreted proteins e.g. molecules involved in fibrotic processes or inflammation. In contrast, the exosome-specific proteins were rather assigned to signaling pathways and membrane-mediated processes. In conclusion, the isolation of exosomes allows to further specify the functionality of adipokines and exoadipokines as part of the adipocyte secretome in signaling and interorgan crosstalk.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Exosomes/metabolism , Proteome/metabolism , Adipokines/metabolism , Cells, Cultured , Female , Humans , Secretory Pathway
15.
Nutr Diabetes ; 8(1): 49, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30190473

ABSTRACT

BACKGROUND: In subcutaneous adipose tissue (SAT), higher stearoyl-CoA desaturase-1 (SCD1) expression has been related to improved insulin sensitivity in thiazolidinedione-treated type 2 diabetes mellitus patients. In animal models, deficiency of the free fatty acid receptor (FFAR) 2 associated with higher and FFAR4-deficiency with lower insulin sensitivity. We hypothesized that increased FFAR2 expression and reductions in FFAR4 and SCD1 expression in SAT of type 2 diabetes mellitus patients associate positively with insulin resistance and impaired beta cell function. METHODS: Twenty-five type 2 diabetes mellitus patients and 25 glucose-tolerant humans (CON) matched for sex, age, and BMI underwent mixed-meal tests to assess insulin sensitivity (OGIS) and beta cell function (ΔAUC(C-peptide)0-180 min/ΔAUC(glucose)0-180 min) in a cross-sectional study. Gene and protein expression of SCD1 and FFAR2/4 were quantified in SAT biopsies. RESULTS: Insulin sensitivity was 14% and beta cell function 71% (both p < 0.001) lower in type 2 diabetes mellitus patients. In type 2 diabetes mellitus, SCD1 mRNA was fivefold (p < 0.001) and protein expression twofold (p < 0.01) lower. While FFAR2/4 mRNA and protein expression did not differ between groups, FFAR2 protein levels correlated negatively with beta cell function only in CON (r = -0.74, p < 0.01). However, neither SCD1 nor FFAR2/4 protein expression correlated with insulin sensitivity in both groups. CONCLUSIONS: Type 2 diabetes patients have lower SCD1, which does not associate with insulin resistance. Only in non-diabetic humans, FFAR2 associated with impaired beta cell function.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Stearoyl-CoA Desaturase/metabolism , Subcutaneous Fat/metabolism , Adult , Cross-Sectional Studies , Diabetes Mellitus, Type 2/diagnosis , Female , Humans , Insulin Resistance/physiology , Male , Middle Aged
16.
J Clin Endocrinol Metab ; 103(10): 3688-3697, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30020463

ABSTRACT

Context and Objectives: Upper and lower body adipose tissue (AT) exhibits opposing associations with obesity-related cardiometabolic diseases. Recent studies have suggested that altered AT oxygen tension (pO2) may contribute to AT dysfunction. Here, we compared in vivo abdominal (ABD) and femoral (FEM) subcutaneous AT pO2 in women who are overweight and have obesity, and investigated the effects of physiological AT pO2 on human adipocyte function. Design: ABD and FEM subcutaneous AT pO2 and AT blood flow (ATBF) were assessed in eight [BMI (body mass index) 34.4 ± 1.6 kg/m2] postmenopausal women who were overweight with obesity and impaired glucose metabolism. ABD and FEM AT biopsy specimens were collected to determine adipocyte morphology and AT gene expression. Moreover, the effects of prolonged exposure (14 days) to physiological AT pO2 on adipokine expression/secretion, mitochondrial respiration, and glucose uptake were investigated in differentiated human multipotent adipose-derived stem cells. Results: AT pO2 was higher in ABD than FEM AT (62.7 ± 6.6 vs 50.0 ± 4.5 mm Hg, P = 0.013), whereas ATBF was comparable between depots. Maximal uncoupled oxygen consumption rates were substantially lower in ABD than FEM adipocytes for all pO2 conditions. Low physiological pO2 (5% O2) decreased proinflammatory gene expression, increased basal glucose uptake, and altered adipokine secretion in ABD and FEM adipocytes. Conclusions: We demonstrated for the first time, to our knowledge, that AT pO2 is higher in ABD than FEM subcutaneous AT in women who are overweight/with obesity, partly due to a lower oxygen consumption rate in ABD adipocytes. Moreover, low physiological pO2 decreased proinflammatory gene expression and improved the metabolic phenotype in differentiated human adipocytes, whereas more heterogeneous effects on adipokine secretion were found.


Subject(s)
Adipose Tissue/physiopathology , Insulin Resistance , Obesity/physiopathology , Overweight/physiopathology , Oxygen Consumption , Oxygen/metabolism , Adipose Tissue/metabolism , Adult , Aged , Biomarkers/analysis , Body Mass Index , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Obesity/metabolism , Overweight/metabolism , Phenotype , Prognosis , Subcutaneous Fat, Abdominal/physiopathology
17.
FEBS Lett ; 581(22): 4303-8, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17716671

ABSTRACT

Adipocyte-derived factors might play a role in the development of hepatic insulin resistance. Resistin was identified as an adipokine linking obesity and insulin resistance. Resistin is secreted from adipocytes in rodents but in humans it was proposed to originate from macrophages and its impact for insulin resistance has remained elusive. To analyze the role of adipokines in general and resistin as a special adipokine, we cultured the human liver cell line HepG2 with adipocyte-conditioned medium (CM) containing various adipokines such as IL-6 and MCP-1, and resistin. CM and resistin both induce insulin resistance with a robust decrease in insulin-stimulated phosphorylation of Akt and GSK3. Insulin resistance could be prevented by co-treatment with troglitazone but not by co-stimulation with adiponectin. As human adipocytes do not secrete resistin, HepG2 cells were also treated with resistin added into CM. CM with resistin addition induced stronger insulin resistance than CM alone pointing to a specific role of resistin in the initiation of hepatic insulin resistance in humans.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Cell Differentiation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin Resistance , Resistin/pharmacology , Adipocytes/enzymology , Adiponectin/pharmacology , Chromans/pharmacology , Culture Media, Conditioned , Female , Glycogen Synthase Kinase 3/metabolism , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Thiazolidinediones/pharmacology , Troglitazone
18.
Trends Endocrinol Metab ; 17(10): 416-22, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17084639

ABSTRACT

Insulin resistance in skeletal muscle is linked to an elevated adipose tissue mass, as is found in obesity, but can also be observed in lipodystrophy, in which adipose tissue is greatly reduced. Adipose tissue releases endocrine and metabolic mediators and is actively involved in crosstalk with skeletal muscle, a process that precedes and underlies the development of insulin resistance in muscles. Adipokines including tumor necrosis factor alpha, interleukin-6, leptin and adiponectin influence insulin signaling in skeletal muscle. Free fatty acids, their metabolites and ectopic fat in muscle also contribute to insulin resistance. Recent research indicates inflammation, endoplasmic reticulum stress and oxidative stress could be underlying mechanisms at the center of the development of insulin resistance. Insights into the role of macrophages in adipose tissue add to the complicated interplay between adipose tissue and skeletal muscle.


Subject(s)
Adipocytes/physiology , Insulin Resistance , Muscle Cells/physiology , AMP-Activated Protein Kinases , Adipose Tissue/cytology , Animals , Fatty Acids, Nonesterified/physiology , Humans , Lipodystrophy/complications , Macrophages/cytology , Macrophages/physiology , Models, Biological , Multienzyme Complexes/physiology , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/physiology
19.
Obesity (Silver Spring) ; 25(12): 2108-2114, 2017 12.
Article in English | MEDLINE | ID: mdl-29024428

ABSTRACT

OBJECTIVE: Heat shock protein 60 (Hsp60) is an adipokine, and its serum concentrations are higher in patients with obesity compared to lean patients. This study aimed to analyze the effect of bariatric surgery on circulating concentrations of Hsp60 in morbid obesity and their correlation with inflammation and metabolic and cardiovascular risk. METHODS: Fifty-three females with morbid obesity undergoing bariatric surgery were enrolled. Serum parameters and anthropometric measures were obtained at baseline and 3 to 12 months post surgery. RESULTS: During the 12-month observation period, Hsp60 decreased significantly from 31.6 ± 4.7 ng/mL at baseline to 22.3 ± 3.0 ng/mL (3 months), 26.5 ± 5.5 (6 months), and 21.1 ± 3.3 ng/mL (12 months). Preoperatively, Hsp60 concentrations correlated positively with total cholesterol, low-density lipoprotein cholesterol, and apolipoprotein B (ApoB) and negatively with adiponectin. At the end of the observation period, serum Hsp60 positively correlated with triglycerides, ApoB, HbA1c , and C-reactive protein (CRP). Patients in the highest quartile of serum Hsp60 were characterized by significantly elevated CRP and interleukin 6 independently of BMI, glycemia, and insulinemia. At baseline and 12 months after surgery, Hsp60 positively correlated with the ApoB/ApoA1 ratio and the cholesterol/high-density lipoprotein cholesterol ratio. CONCLUSIONS: Hsp60 concentrations are elevated in morbid obesity and decreased after surgery-induced weight loss. Their correlation with inflammatory markers and cardiovascular risk might link obesity and cardiovascular disease.


Subject(s)
Bariatric Surgery/adverse effects , Cardiovascular Diseases/etiology , Chaperonin 60/metabolism , Inflammation/blood , Obesity, Morbid/surgery , Adult , Bariatric Surgery/methods , Female , Humans , Male , Middle Aged , Obesity, Morbid/pathology , Risk Factors
20.
Endocrinology ; 147(5): 2458-67, 2006 May.
Article in English | MEDLINE | ID: mdl-16439461

ABSTRACT

Adipose tissue is a major secretory and endocrine active organ producing a variety of bioactive proteins that may regulate energy metabolism and insulin sensitivity. In several studies, we have already shown that adipocyte-secretory products induce skeletal muscle insulin resistance. However, the precise nature of these factors has remained elusive. Human adipocytes were found to secrete various cytokines including IL-6, IL-8, macrophage inflammatory protein-1alpha/beta, and monocyte chemotactic protein-1 (MCP-1). Among these candidates, MCP-1 alone impaired insulin signaling in skeletal muscle cells at doses similar to its physiological plasma concentrations (200 pg/ml), whereas IL-6, IL-8, and macrophage inflammatory protein-1beta were effective at very high concentrations only. In addition, MCP-1 significantly reduced insulin-stimulated glucose uptake in the myocytes. Expression analysis of chemokine receptors in skeletal muscle cells revealed the presence of chemokine CXC motif receptor 1/2 and chemokine CC motif receptor 1/2/4/5/10. The action of MCP-1 on insulin signaling in skeletal muscle cells occurs via ERK1/2 activation but does not involve activation of the nuclear factor kappaB pathway. In conclusion, our data show that adipocytes secrete various adipokines that may be involved in the negative cross-talk between adipose tissue and skeletal muscle. Human skeletal muscle cells are highly sensitive toward MCP-1, which impairs insulin signaling and glucose uptake at concentrations even below that found in the circulation. However, other cytokines that are released by adipocytes impair insulin action only at supraphysiological concentrations. Therefore, MCP-1 may represent a molecular link in the negative cross-talk between adipose tissue and skeletal muscle assigning a completely novel important role to MCP-1 besides inflammation.


Subject(s)
Adipocytes/cytology , Adipose Tissue/pathology , Chemokine CCL2/physiology , Muscle, Skeletal/metabolism , Adipocytes/metabolism , Adipose Tissue/cytology , Adult , Breast/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Chemokines/metabolism , Cytokines/metabolism , DNA Primers/chemistry , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Glucose/pharmacokinetics , Humans , Immunoblotting , Inflammation , Insulin/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Middle Aged , Muscle Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL