Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cells ; 10(8)2021 08 13.
Article in English | MEDLINE | ID: mdl-34440850

ABSTRACT

A highly complex network of organ communication plays a key role in regulating metabolic homeostasis, specifically due to the modulation of the insulin signaling machinery. As a paradigm, the role of adipose tissue in organ crosstalk has been extensively investigated, but tissues such as muscles and the liver are equally important players in this scenario. Perturbation of organ crosstalk is a hallmark of insulin resistance, emphasizing the importance of crosstalk molecules in the modulation of insulin signaling, potentially leading to defects in insulin action. Classically secreted proteins are major crosstalk molecules and are able to affect insulin signaling in both directions. In this review, we aim to focus on some crosstalk mediators with an impact on the early steps of insulin signaling. In addition, we also summarize the current knowledge on the role of extracellular vesicles in relation to insulin signaling, a more recently discovered additional component of organ crosstalk. Finally, an attempt will be made to identify inter-connections between these two pathways of organ crosstalk and the potential impact on the insulin signaling network.


Subject(s)
Insulin/metabolism , Signal Transduction , Adipose Tissue/metabolism , Exosomes/genetics , Exosomes/metabolism , Extracellular Vesicles/metabolism , Humans , Liver/metabolism , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , MicroRNAs/metabolism , Receptor, Insulin/metabolism
3.
Physiology (Bethesda) ; 36(2): 102-113, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33595385

ABSTRACT

There is an urgent need for developing effective drugs to combat the obesity and Type 2 diabetes mellitus epidemics. The endocannabinoid system plays a major role in energy homeostasis. It comprises the cannabinoid receptors 1 and 2 (CB1 and CB2), endogenous ligands called endocannabinoids and their metabolizing enzymes. Because the CB1 receptor is overactivated in metabolic alterations, pharmacological blockade of the CB1 receptor arose as a promising candidate to treat obesity. However, because of the wide distribution of CB1 receptors in the central nervous system, their negative central effects halted further therapeutic use. Although the CB2 receptor is mostly peripherally expressed, its role in metabolic homeostasis remains unclear. This review discusses the potential of CB1 and CB2 receptors at the peripheral level to be therapeutic targets in metabolic diseases. We focus on the impact of pharmacological intervention and/or silencing on peripheral cannabinoid receptors in organs/tissues relevant for energy homeostasis. Moreover, we provide a perspective on novel therapeutic strategies modulating these receptors. Targeting CB1 with peripherally restricted antagonists, neutral antagonists, inverse agonists, or monoclonal antibodies could represent successful strategies. CB2 agonism has shown promising results at preclinical level. Beyond classic antagonism and agonism targeting orthosteric sites, the recently described crystal structures of CB1 and CB2 open new possibilities for therapeutic interventions with negative and positive allosteric modulators. The challenge of simultaneously targeting CB1 and CB2 might be possible by developing dual-steric ligands. The future will tell whether these promising strategies result in a renaissance of the cannabinoid receptors as therapeutic targets in metabolic diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Diseases , Diabetes Mellitus, Type 2/drug therapy , Endocannabinoids , Humans , Metabolic Diseases/drug therapy , Obesity , Receptors, Cannabinoid
5.
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
6.
Front Immunol ; 6: 386, 2015.
Article in English | MEDLINE | ID: mdl-26284071

ABSTRACT

Dipeptidyl-peptidase 4 (DPP4) is a glycoprotein of 110 kDa, which is ubiquitously expressed on the surface of a variety of cells. This exopeptidase selectively cleaves N-terminal dipeptides from a variety of substrates, including cytokines, growth factors, neuropeptides, and the incretin hormones. Expression of DPP4 is substantially dysregulated in a variety of disease states including inflammation, cancer, obesity, and diabetes. Since the incretin hormones, glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide (GIP), are major regulators of post-prandial insulin secretion, inhibition of DPP4 by the gliptin family of drugs has gained considerable interest for the therapy of type 2 diabetic patients. In this review, we summarize the current knowledge on the DPP4-incretin axis and evaluate most recent findings on DPP4 inhibitors. Furthermore, DPP4 as a type II transmembrane protein is also known to be cleaved from the cell membrane involving different metalloproteases in a cell-type-specific manner. Circulating, soluble DPP4 has been identified as a new adipokine, which exerts both para- and endocrine effects. Recently, a novel receptor for soluble DPP4 has been identified, and data are accumulating that the adipokine-related effects of DPP4 may play an important role in the pathogenesis of cardiovascular disease. Importantly, circulating DPP4 is augmented in obese and type 2 diabetic subjects, and it may represent a molecular link between obesity and vascular dysfunction. A critical evaluation of the impact of circulating DPP4 is presented, and the potential role of DPP4 inhibition at this level is also discussed.

7.
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
8.
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
9.
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
10.
PLoS One ; 8(9): e73680, 2013.
Article in English | MEDLINE | ID: mdl-24040023

ABSTRACT

Brown adipose tissue has gained interest as a potential target to treat obesity and metabolic diseases. Irisin is a newly identified hormone secreted from skeletal muscle enhancing browning of white fat cells, which improves systemic metabolism by increasing energy expenditure in mice. The discovery of irisin raised expectations of its therapeutic potential to treat metabolic diseases. However, the effect of irisin in humans is unclear. Analyses of genomic DNA, mRNA and expressed sequence tags revealed that FNDC5, the gene encoding the precursor of irisin, is present in rodents and most primates, but shows in humans a mutation in the conserved start codon ATG to ATA. HEK293 cells transfected with a human FNDC5 construct with ATA as start codon resulted in only 1% full-length protein compared to human FNDC5 with ATG. Additionally, in vitro contraction of primary human myotubes by electrical pulse stimulation induced a significant increase in PGC1α mRNA expression. However, FNDC5 mRNA level was not altered. FNDC5 mRNA expression in muscle biopsies from two different human exercise studies was not changed by endurance or strength training. Preadipocytes isolated from human subcutaneous adipose tissue exhibited differentiation to brite human adipocytes when incubated with bone morphogenetic protein (BMP) 7, but neither recombinant FNDC5 nor irisin were effective. In conclusion, our findings suggest that it is rather unlikely that the beneficial effect of irisin observed in mice can be translated to humans.


Subject(s)
Codon, Initiator/genetics , Fibronectins/genetics , Gene Expression Profiling , Mutation , Adipocytes/cytology , Adipocytes/metabolism , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Female , Fibronectins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Microscopy, Fluorescence , Middle Aged , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Young Adult
11.
Arch Physiol Biochem ; 119(4): 151-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23721302

ABSTRACT

Obesity is considered a worldwide health concern. Most of obesity therapies are aimed at decreasing energy intake. However, recent data suggest that increasing cellular energy expenditure could be a useful approach to reduce adiposity. Adaptive thermogenesis, a biological process within the brown fat by which energy is dissipated in mitochondria, is a great tool to increase energy expenditure. Several studies have confirmed the presence of brown adipose tissue in adult humans, whose activity may make it a target for the treatment of obesity. Differentiation of brown adipocytes could be a potent tool to promote weight loss by increasing energy expenditure. Here we review the mechanisms potentially associated with expansion and activation of brown adipose tissue, and modulation of adaptive thermogenesis. Controlling one or more of these pathways could induce a positive regulation of brown adipogenesis. A better understanding of these molecular pathways could potentially result in novel anti-obesity therapies.


Subject(s)
Adipocytes, White/metabolism , Adipose Tissue, Brown/metabolism , Energy Metabolism , Obesity/therapy , Thermogenesis , Adipocytes, White/cytology , Adipocytes, White/pathology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/pathology , Animals , Humans , Obesity/metabolism , Obesity/pathology
12.
PLoS One ; 8(3): e59697, 2013.
Article in English | MEDLINE | ID: mdl-23555749

ABSTRACT

CONTEXT: Adipokines are linked to the development of cardiovascular dysfunction in type 2 diabetes (DM2). In DM2-patients, circulating levels of omentin-1, an adipokine preferentially expressed in epicardial adipose tissue, are decreased. This study investigated whether omentin-1 has a cardioprotective function. METHODS: Omentin-1 levels in plasma and cardiac fat depots were determined in DM2-patients versus controls. Moreover, the relation between omentin-1 levels and cardiac function was examined in men with uncomplicated DM2. Finally, we determined whether omentin-1 could reverse the induction of cardiomyocyte dysfunction by conditioned media derived from epicardial adipose tissue from patients with DM2. RESULTS: Omentin-1 was highly expressed and secreted by epicardial adipose tissue, and reduced in DM2. Circulating omentin-1 levels were lower in DM2 versus controls, and positively correlated with the diastolic parameters early peak filling rate, early deceleration peak and early deceleration mean (all P<0.05). The improved diastolic function following pioglitazone treatment associated with increases in omentin-1 levels (P<0.05). In vitro, exposure of cardiomyocytes to conditioned media derived from epicardial adipose tissue from patients with DM2 induced contractile dysfunction and insulin resistance, which was prevented by the addition of recombinant omentin. CONCLUSION: These data identify omentin-1 as a cardioprotective adipokine, and indicate that decreases in omentin-1 levels could contribute to the induction of cardiovascular dysfunction in DM2.


Subject(s)
Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Lectins/metabolism , Myocytes, Cardiac/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Aged , Animals , Case-Control Studies , Cytokines/blood , Cytokines/pharmacology , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , GPI-Linked Proteins/blood , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/pharmacology , Gene Expression Regulation/drug effects , Humans , Insulin/metabolism , Lectins/blood , Lectins/pharmacology , Male , Middle Aged , Muscle Contraction/drug effects , Myocytes, Cardiac/drug effects , Pioglitazone , Rats , Signal Transduction/drug effects , Thiazolidinediones/pharmacology
13.
Circulation ; 126(19): 2324-34, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23065384

ABSTRACT

BACKGROUND: Secreted factors from epicardial adipose tissue (EAT) have been implicated in the development of cardiomyocyte dysfunction. This study aimed to assess whether alterations in the secretory profile of EAT in patients with type 2 diabetes mellitus (DM2) affect contractile function and insulin action in cardiomyocytes. METHODS AND RESULTS: Contractile function and insulin action were analyzed in primary adult rat cardiomyocytes incubated with conditioned media (CM) generated from explants of EAT biopsies obtained from patients without and with DM2. CM from subcutaneous and pericardial adipose tissue biopsies from the same patients served as the control. Cardiomyocytes treated with CM (EAT) from DM2 patients showed reductions in sarcomere shortening, cytosolic Ca(2+) fluxes, expression of sarcoplasmic endoplasmic reticulum ATPase 2a, and decreased insulin-mediated Akt-Ser473-phosphorylation as compared with CM from the other groups. Profiling of the CM showed that activin A, angiopoietin-2, and CD14 selectively accumulated in CM-EAT-DM2 versus CM-EAT in patients without DM2 and CM from the other fat depots. Accordingly, EAT biopsies from DM2 patients were characterized by clusters of CD14-positive monocytes. Furthermore, SMAD2-phosphorylation, a downstream target of activin A signaling, was elevated in cardiomyocytes treated with CM (EAT) from DM2 patients, and the detrimental effects of CM (EAT) from DM2 patients were partially abolished in cardiomyocytes pretreated with a neutralizing antibody against activin A. Finally, both recombinant activin A and angiopoietin-2 reduced cardiomyocyte contractile function, but only activin A reduced the expression of sarcoplasmic endoplasmic reticulum ATPase 2a. CONCLUSIONS: Collectively, our data implicate DM2-related alterations in the secretory profile of EAT in the pathogenesis of diabetes mellitus-related heart disease.


Subject(s)
Activins/pharmacology , Adipose Tissue/metabolism , Angiopoietin-2/pharmacology , Diabetes Mellitus, Type 2/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Pericardium/metabolism , Activins/metabolism , Adipose Tissue/pathology , Aged , Angiopoietin-2/metabolism , Animals , Biopsy , Calcium/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Humans , Insulin/metabolism , Male , Models, Animal , Myocytes, Cardiac/pathology , Pericardium/pathology , Rats , Rats, Inbred Lew , Sarcomeres/drug effects , Sarcomeres/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/drug effects
14.
Methods Mol Biol ; 933: 255-63, 2012.
Article in English | MEDLINE | ID: mdl-22893412

ABSTRACT

Glucose disposal in skeletal muscle is a major target for insulin action and assessment of insulin-regulated glucose uptake under in vitro conditions allows the direct determination of insulin sensitivity in this organ. For this purpose, a variety of muscle preparations from different parts of the body can be used. We describe here a detailed protocol for using epitrochlearis muscle strips and additionally for using primary skeletal muscle cells.


Subject(s)
Glucose/metabolism , Insulin Resistance , Insulin/metabolism , Muscle, Skeletal/metabolism , Animals , Biological Transport , Cells, Cultured , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Rats
15.
Nat Rev Endocrinol ; 8(12): 709-16, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22847239

ABSTRACT

Obesity is the hallmark of the metabolic syndrome and predisposes patients to the development of major chronic metabolic diseases including type 2 diabetes mellitus. Adipose tissue expansion in obesity is characterized by increasing infiltration of proinflammatory immune cells into adipose tissue causing chronic, low-grade inflammation. Phenotypic switching of macrophages is an important mechanism of adipose tissue inflammation, and there is involvement of cells from the adaptive immune system in this process. T-cell phenotype changes and recruitment of B cells and T cells precedes macrophage infiltration. Cytokines and chemokines produced by immune cells influence localized and systemic inflammation, which is a pathogenic link between obesity and insulin resistance. Antigens absorbed from the gut might contribute to T-cell activation and recruitment into visceral adipose tissue in obesity. This Review summarizes, in the context of obesity, the evidence for infiltration of adipose tissue by cells of the adaptive immune system, how adaptive system cells affect innate cell populations and the influence of adaptive immune cells on the development of insulin resistance.


Subject(s)
Adaptive Immunity , Diabetes Mellitus, Type 2/immunology , Insulin Resistance/immunology , Obesity/immunology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Inflammation Mediators/physiology , Obesity/metabolism
16.
Mol Cell Endocrinol ; 362(1-2): 194-201, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22750100

ABSTRACT

It is widely accepted that obesity is a major risk factor for the development of atherosclerosis. In this context, adipose tissue produces a variety of adipokines and releases free fatty acids, contributing to a chronic-low grade inflammation state implicated in vascular complications. In this study, we investigated the role of adipokines, oleic acid (OA), palmitic acid (PA), and the combinations on activation of NF-κB target genes in human vascular smooth muscle cells (SMC) to assess the hypothesis of synergistic interactions between these molecules. Adipocyte-conditioned medium (CM), generated from human adipocytes, in combination with low concentrations of OA, but not PA, induces SMC proliferation and activation of the transcription factor NF-κB in a synergistic way. Combined treatment of CM and OA further regulates a set of downstream NF-κB target genes including angiopoietin-1, activin A, and MMP-1, all critically involved in SMC dysfunction. This suggests that the lipotoxic potential of fatty acids is substantially enhanced by the presence of adipocyte-derived factors.


Subject(s)
Adipokines/physiology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Oleic Acid/physiology , Palmitic Acid/pharmacology , Activins/genetics , Activins/metabolism , Adipocytes/metabolism , Adipokines/pharmacology , Angiopoietins/genetics , Angiopoietins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Culture Media, Conditioned , Female , Gene Expression , Gene Expression Regulation , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Obesity/metabolism , Obesity/pathology , Oleic Acid/pharmacology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology , Vasculitis/metabolism , Vasculitis/pathology
18.
Arch Physiol Biochem ; 118(3): 92-101, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22691105

ABSTRACT

Studies have shown the implication of specific adipokines or fatty acids (FA) in the pathogenesis of insulin resistance. However, the interplay of adipokines with FA remains poorly understood. This study aimed to investigate the combined effects of adipokines and low concentrations of palmitic acid (PA, 100 µmol/l) on skeletal muscle metabolism. Human skeletal muscle cells were incubated with adipocyte-conditioned medium (CM), PA or PA+CM, and FA transporter and FA metabolism were analysed. CM-incubation increased CD36 level (1.8 fold) and PA-uptake (1.4 fold). However, only co-application of PA+CM resulted in profound lipid accumulation (5.3 fold), 60% reduction of PA-oxidation and 3.5 fold increased diacylglycerol content. Our results support a novel role for adipokines in the pathogenesis of T2D by increasing the lipotoxic potential of PA, notably of low concentrations. This implies an increased lipotoxic risk already at an early stage of weight gain, when lipolysis has not yet contributed to increased plasma free FA levels.


Subject(s)
Adipokines/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipokines/pharmacology , Biological Transport , CD36 Antigens/biosynthesis , Cells, Cultured , Culture Media, Conditioned , Diglycerides/metabolism , Fatty Acid Transport Proteins/biosynthesis , Humans , Lipid Peroxidation , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Palmitic Acid/metabolism , Palmitic Acid/pharmacology
19.
Arch Physiol Biochem ; 118(3): 84-91, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22616691

ABSTRACT

CONTEXT: Recent secretome analyses suggest that human fat cells secrete hundreds of proteins (adipokines). OBJECTIVE: We made an overall analysis of their potential functional importance. MATERIALS AND METHODS: A secretome of 347 adipokines was evaluated by in silico analysis of their expression during adipocyte differentiation, regulation by obesity and adipose region. The gene expression in human adipose tissue was investigated in microarray studies using samples from different adipose depots from lean or obese patients. RESULTS: 60% of the adipokines were regulated by obesity and 50% between visceral and subcutaneous adipose region. Eight adipokines, all novel, scored particularly high in the in silico analysis. Among those, four were both regulated by obesity and adipose region, namely WNT1-inducible-signaling pathway protein 2, transmembrane glycoprotein NMB, inter-alpha-trypsin inhibitor heavy chain H5, and complement C4-A. Furthermore, many adipokines were extracellular matrix proteins. CONCLUSION: Several novel adipokines have potential important functional features warranting in depth analysis.


Subject(s)
Adipocytes/metabolism , Gene Expression Regulation , Intra-Abdominal Fat/metabolism , Obesity/genetics , Subcutaneous Fat/metabolism , Adipocytes/cytology , Adipokines , CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Complement C4a/genetics , Complement C4a/metabolism , Gene Expression , Gene Expression Profiling , Humans , Intra-Abdominal Fat/cytology , Metabolome , Metabolomics , Obesity/metabolism , Obesity/pathology , Oligonucleotide Array Sequence Analysis , Proteinase Inhibitory Proteins, Secretory/genetics , Proteinase Inhibitory Proteins, Secretory/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Subcutaneous Fat/cytology
20.
J Clin Endocrinol Metab ; 97(7): E1187-91, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22523336

ABSTRACT

CONTEXT: Low testosterone accompanied by elevated estradiol associates with the development of metabolic dysfunction in men. OBJECTIVE: The aim of the study was to explore the hypothesis that alterations in sex steroid levels induce metabolic dysfunction through adipokines. DESIGN: Circulating levels of sex steroids and 28 adipokines were determined in a cross-sectional study of morbidly obese men and aged-matched controls, as well as in a randomized clinical trial with healthy young men in which obesity-related alterations in sex steroid levels were mimicked by treatment with an aromatase inhibitor plus estradiol patches. RESULTS: Morbidly obese men had lower testosterone levels than normal-weight controls. Estradiol levels were increased in morbidly obese men (without DM2) as compared to normal-weight controls. Circulating levels of multiple proinflammatory cytokines, including IL-1Ra, IL-5, IL-6, IL-10, leptin, monocyte chemoattractant protein 1 (MCP1), and macrophage inflammatory protein 1α, positively associated with estradiol and negatively with testosterone. The associations with estradiol, but not with testosterone, remained significant after adjusting for adipocyte cell size. In a separate clinical trial, the direct adverse effects of lowering testosterone and raising estradiol on MCP1 were substantiated in vivo. CONCLUSIONS: Initial alterations in sex steroid levels may contribute to metabolic dysfunction through adverse effects on adipokine levels in obese men. The direct adverse effects on MCP1, a chemokine highly linked to the development of metabolic dysfunction, were substantiated in a trial mimicking obesity-related alterations of sex steroid levels in healthy young males.


Subject(s)
Chemokine CCL2/blood , Gonadal Steroid Hormones/blood , Gonadal Steroid Hormones/pharmacology , Metabolic Diseases/etiology , Obesity/complications , Adipokines/blood , Adipokines/metabolism , Adult , Case-Control Studies , Chemokine CCL2/physiology , Comorbidity , Cross-Sectional Studies , Gonadal Steroid Hormones/physiology , Humans , Male , Metabolic Diseases/blood , Metabolic Diseases/epidemiology , Middle Aged , Obesity/blood , Obesity/epidemiology , Randomized Controlled Trials as Topic , Risk Factors , Sex Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...