Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Hypertens ; 34(5): 869-76, 2016 May.
Article in English | MEDLINE | ID: mdl-26895560

ABSTRACT

BACKGROUND: Dipeptidyl peptidase-4 (DPP4) is a key protein in glucose homeostasis and a pharmacological target in type 2 diabetes mellitus. This study explored whether the novel adipokine soluble DPP4 (sDPP4) can cause endothelial dysfunction, an early marker of impaired vascular reactivity. METHOD: Reactivity was studied in mesenteric arteries from 3-month-old female mice, using a small vessel myograph. Thromboxane A2 (TXA2) release was explored in cultured human coronary artery endothelial cells by enzyme immunoassay. RESULTS: Neither the contractility to noradrenaline nor the endothelium-independent relaxations induced by sodium nitroprusside were modified by sDPP4. However, sDPP4 impaired in a concentration-dependent manner the endothelium-dependent relaxation elicited by acetylcholine. The DPP4 inhibitors K579 and linagliptin prevented the defective relaxation induced by sDPP4, as did the protease-activated receptor 2 (PAR2) inhibitor GB83. Downstream of PAR2, the cyclooxygenase (COX) inhibitor indomethacin, the COX2 inhibitor celecoxib or the thromboxane receptors blocker SQ29548 prevented the deleterious effects of sDPP4. Accordingly, sDPP4 triggered the release of TXA2 by endothelial cells, whereas TXA2 release was prevented by inhibiting DPP4, PAR2 or COX. CONCLUSION: In summary, these findings reveal sDPP4 as a direct mediator of endothelial dysfunction, acting through PAR2 activation and the release of vasoconstrictor prostanoids. By interfering with these actions, DPP4 inhibitors might help preserving endothelial function in the context of cardiometabolic diseases.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Dipeptidyl Peptidase 4/metabolism , Endothelium, Vascular/metabolism , Receptor, PAR-2/metabolism , Thromboxane A2/metabolism , Animals , Dipeptidyl Peptidase 4/adverse effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL
8.
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
9.
Arch Physiol Biochem ; 121(5): 194-205, 2015.
Article in English | MEDLINE | ID: mdl-26599229

ABSTRACT

Adipose tissue and skeletal muscle are organs that respond strongly to obesity and physical activity exhibiting high secretory activity. To identify novel putative adipomyokines, comparative expression studies of skeletal muscle and adipose tissue of lean (C57BL/6J) and obese (C57BL/6J on a high-fat diet and NZO) mice, of sedentary and endurance trained C57BL/6J mice and of cattle characterized by different amounts of intramuscular fat were combined with human secretome data and scored. In highly regulated transcripts, we identified 119 myokines, 79 adipokines and 22 adipomyokines. Network analysis of these candidates revealed remodelling of extracellular matrix and tissue fibrosis as relevant functions of several of these candidates. Given the pathophysiogical relevance of fibrosis for adipose-muscle-cross-talk in obesity and type 2 diabetes and its physiological role in exercise adaptation and meat quality of farm animals, they represent interesting candidates for further investigations in different research areas and species.


Subject(s)
Adipokines/metabolism , Adipose Tissue/metabolism , Cytokines/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Proteome , Transcriptome , Adipokines/genetics , Adipose Tissue/cytology , Animals , Cattle , Cells, Cultured , Cytokines/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Regulatory Networks , Humans , Ion Channels/physiology , Male , Metabolic Syndrome/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins/physiology , Muscle, Skeletal/cytology , Obesity/etiology , Physical Conditioning, Animal , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Uncoupling Protein 1
10.
Curr Obes Rep ; 4(4): 411-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26349436

ABSTRACT

The crosstalk between adipose tissue and skeletal muscle has gained considerable interest, since this process, specifically in obesity, substantially drives the pathogenesis of muscle insulin resistance. In this review, we discuss novel concepts and targets of this bidirectional organ communication system. This includes adipo-myokines like apelin and FGF21, inflammasomes, autophagy, and microRNAs (miRNAs). Literature analysis shows that the crosstalk between fat and muscle involves both extracellular molecules and intracellular organelles. We conclude that integration of these multiple crosstalk elements into one network will be required to better understand this process.


Subject(s)
Adipose Tissue/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Receptor Cross-Talk/physiology , Apelin , Exercise , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Humans , Inflammasomes/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism
11.
Arch Physiol Biochem ; 121(3): 81-7, 2015.
Article in English | MEDLINE | ID: mdl-26135380

ABSTRACT

Adipose tissue is not only releasing lipids but also various adipokines that are both dysregulated in the obese state and may contribute to obesity-associated vascular dysfunction and cardiovascular risk. We have previously shown that the combination of adipocyte-conditioned medium (CM) and oleic acid (OA) increases proliferation of human vascular smooth muscle cells (VSMC) in a synergistic way. We identified vascular endothelial growth factor (VEGF) as a component within CM that is responsible for most of the observed effects. In this study, we investigate novel mechanisms that underlie the combined effects of adipokine and oleic acid-induced proliferation of VSMC. Oleic acid leads to significant lipid accumulation in VSMC that is further enhanced by the combined treatment with CM. Accordingly CM stimulates CD36 expression in VSMC while OA is not affecting CD36. Silencing of CD36 was established and prevents lipid accumulation in all tested conditions. CD36 silencing also abrogates CM- and OA-induced proliferation and considerably reduces proliferation induced by the combination of CM and OA. At the same time, VEGF secretion and VEGF-receptor 1 (VEGF-R1) by VSMC was not affected by CD36 silencing. However, VEGF was not able to induce any proliferation in VSMC after CD36 silencing that also blunted VEGF-induced extracellular signal-regulated kinase (ERK) activation. Finally, combined silencing of CD36 together with a blocking antibody against VEGF prevented most of CMOA-induced proliferation. In conclusion, our results demonstrate that CD36 is mediating CM-induced proliferation of VSMC. Induction of CD36 by adipokines enhances the response of VSMC towards VEGF and OA.


Subject(s)
Adipokines/pharmacology , CD36 Antigens/genetics , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Myocytes, Smooth Muscle/drug effects , Oleic Acid/pharmacology , Adipocytes/cytology , Adipocytes/metabolism , Adipokines/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adult , Antibodies, Neutralizing/pharmacology , CD36 Antigens/antagonists & inhibitors , CD36 Antigens/metabolism , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Culture Media, Conditioned/chemistry , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
12.
Horm Mol Biol Clin Investig ; 21(2): 117-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25781556

ABSTRACT

Adipose tissue expansion is associated with adipocyte dysfunction and increased inflammatory processes. In the obese state, adipose tissue is characterized by an impaired intracellular stress defense system and dysbalanced heat shock response. Several members of the heat shock protein (HSP) family have been identified as novel adipokines released upon cellular stress, which might be a molecular link from adipose tissue inflammation to the cardiovascular system. Therefore, this review aims at summarizing and discussing our recent knowledge on HSPs in relation to obesity and their potential links to cardiovascular disease. Of particular importance/interest are two members of the HSP family, HSP60 and heme oxygenase 1 (HO-1), which have been well described as adipokines, and studied in the context of obesity and cardiovascular disease. HSP60 is regarded as a novel molecular link between adipose tissue inflammation and obesity-associated insulin resistance. The role of HO-1 induction in the obese state is well-documented, but a causal relationship between increased HO-1 levels and obesity-associated metabolic diseases is still controversial. Both HSP60 and HO-1 are also forthcoming targets for the treatment of cardiovascular disease, and the current knowledge will also be discussed in this review.


Subject(s)
Cardiovascular Diseases/metabolism , Heat-Shock Proteins/metabolism , Obesity/metabolism , Animals , Chaperonin 60/metabolism , Disease Models, Animal , Heme Oxygenase-1/metabolism , Humans , Inflammation/metabolism , Insulin Resistance , Metabolic Diseases/metabolism , Mitochondrial Proteins/metabolism
13.
FEBS Lett ; 588(21): 3870-7, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25217834

ABSTRACT

Dipeptidyl peptidase 4 is an important drug target for diabetes and a novel adipokine. However, it is unknown how soluble DPP4 (sDPP4) is cleaved from the cell membrane and released into the circulation. We show here that MMP1, MMP2 and MMP14 are involved in DPP4 shedding from human vascular smooth muscle cells (SMC) and MMP9 from adipocytes. Hypoxia increased DPP4 shedding from SMC which is associated with increased mRNA expression of MMP1. Our data suggest that constitutive as well as hypoxia-induced DPP4 shedding occurs due to a complex interplay between different MMPs in cell type-specific manner.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Matrix Metalloproteinases/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Adipocytes/drug effects , Brefeldin A/pharmacology , Cell Hypoxia , Dipeptidyl Peptidase 4/chemistry , Female , Humans , Male , Myocytes, Smooth Muscle/drug effects , Protease Inhibitors/pharmacology , Solubility , Up-Regulation/drug effects
14.
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
15.
Front Horm Res ; 43: 79-92, 2014.
Article in English | MEDLINE | ID: mdl-24943300

ABSTRACT

Adipose tissue (AT) was long perceived as a passive lipid storage depot but it is now considered as an endocrine organ that produces a large number of mediators that affect metabolism, inflammation and coagulation. In obesity, the increased size of adipocytes and chronic low-grade inflammation within AT alter its normal physiological function. AT dysfunction results in altered production and secretion of adipokines, which in turn affect several tissues, e.g. the liver, skeletal muscles and vasculature, in a para- or endocrine manner. Numerous circulating proinflammatory mediators involved in the development of cardiovascular disease (CVD) are directly released from adipocytes, thereby linking obesity to an increased cardiovascular risk. In the current chapter, we focus, on the one hand, on a small selection of novel adipokines with a potentially strong link to CVD: soluble dipeptidyl peptidase-4, visfatin and lipocalin-2. On the other hand, we summarize the most recent findings on the novel cardioprotective adipokines omentin and apelin.


Subject(s)
Adipose Tissue/physiopathology , Cardiovascular Diseases/etiology , Inflammation/physiopathology , Obesity/physiopathology , Acute-Phase Proteins/metabolism , Adipocytes/metabolism , Adipokines/biosynthesis , Apelin , Cytokines/physiology , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , GPI-Linked Proteins/physiology , Glucagon-Like Peptide 1/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Lectins/physiology , Lipocalin-2 , Lipocalins/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Obesity/complications , Proto-Oncogene Proteins/metabolism , Risk Factors
16.
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
17.
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
18.
Diabetes Care ; 36(12): 4083-90, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24130353

ABSTRACT

OBJECTIVE: To study expression of the recently identified adipokine dipeptidyl peptidase-4 (DPP4) in subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) of patients with various BMIs and insulin sensitivities, as well as to assess circulating DPP4 in relation to obesity and insulin sensitivity. RESEARCH DESIGN AND METHODS: DPP4 expression was measured in SAT and VAT from 196 subjects with a wide range of BMIs and insulin sensitivities. DPP4 release was measured ex vivo in paired biopsies from SAT and VAT as well as in vivo from SAT of lean and obese patients. Circulating DPP4 was measured in insulin-sensitive and insulin-resistant BMI-matched obese patients. RESULTS: DPP4 expression was positively correlated with BMI in both SAT and VAT, with VAT consistently displaying higher expression than SAT. Ex vivo release of DPP4 from adipose tissue explants was higher in VAT than in SAT in both lean and obese patients, with obese patients displaying higher DPP4 release than lean controls. Net release of DPP4 from adipose tissue was also demonstrated in vivo with greater release in obese subjects than in lean subjects and in women than in men. Insulin-sensitive obese patients had significantly lower circulating DPP4 than did obesity-matched insulin-resistant patients. In this experiment, DPP4 positively correlated with the amount of VAT, adipocyte size, and adipose tissue inflammation. CONCLUSIONS: DPP4, a novel adipokine, has a higher release from VAT that is particularly pronounced in obese and insulin-resistant patients. Our data suggest that DPP4 may be a marker for visceral obesity, insulin resistance, and the metabolic syndrome.


Subject(s)
Dipeptidyl Peptidase 4/genetics , Gene Expression Regulation , Insulin Resistance/genetics , Intra-Abdominal Fat/enzymology , Obesity/genetics , RNA, Messenger/genetics , Subcutaneous Fat/enzymology , Adipocytes/enzymology , Adipocytes/pathology , Adult , Aged , Aged, 80 and over , Biopsy , Body Mass Index , Cells, Cultured , Dipeptidyl Peptidase 4/biosynthesis , Female , Humans , Insulin/metabolism , Intra-Abdominal Fat/pathology , Male , Middle Aged , Obesity/metabolism , Obesity/pathology , Real-Time Polymerase Chain Reaction , Subcutaneous Fat/pathology , Young Adult
19.
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
20.
Arch Physiol Biochem ; 119(2): 39-51, 2013 May.
Article in English | MEDLINE | ID: mdl-23398498

ABSTRACT

CONTEXT: CD73 converts extracellular AMP to adenosine which is well known to inhibit lipolysis. It is unknown, however, whether adenosine formed directly by CD73 is functionally relevant in this process. OBJECTIVE: We therefore explored the effect of CD73-derived adenosine on body fat of aged mice. RESULTS: In lean mice, extracellular adenosine formation by adipocytes is dependent on CD73. High fat diet down-regulates the expression of CD73 in wildtype mice similar to ob/ob mice. Transgenic mice chronically lacking CD73 (CD73(-/-)) gain significantly less body weight and show decreased superficial white fat content as well as increased serum free fatty acids and triglycerides. In addition, intramyocellular lipid levels are significantly increased. This phenotype is accompanied by an increase in blood glucose and serum insulin levels although insulin secretion and the level of insulin degrading enzyme are unaltered. Additionally, insulin-induced Akt phosphorylation is reduced in skeletal muscle of CD73(-/-) mice. CONCLUSION: CD73-derived adenosine is functionally involved in body fat homeostasis.


Subject(s)
5'-Nucleotidase/deficiency , 5'-Nucleotidase/physiology , Dyslipidemias/etiology , Lipid Metabolism/physiology , Muscles/metabolism , 5'-Nucleotidase/genetics , Adenosine/metabolism , Adenosine/physiology , Adenosine Monophosphate/metabolism , Adipocytes/metabolism , Adipose Tissue/physiology , Animals , Blood Glucose/analysis , Diet, High-Fat , Down-Regulation/physiology , Dyslipidemias/metabolism , Gene Deletion , Homeostasis/physiology , Insulin/blood , Lipolysis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Obesity/metabolism , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...