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










Database
Language
Publication year range
1.
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.
Diabetes Obes Metab ; 19(12): 1722-1731, 2017 12.
Article in English | MEDLINE | ID: mdl-28497570

ABSTRACT

AIMS: To conduct a comprehensive pre-clinical study of the novel ultra-long acting insulin analogue LAPS Insulin115. METHODS: Pharmacokinetic/pharmacodynamic studies comparing LAPS Insulin115 with other basal insulins were conducted in genetically diabetic (db/db) mice. Insulin signalling in the major target organs was analysed using Western blot after single subcutaneous injection in wild-type male Wistar rats. Using in vitro assays we analysed transendothelial transport, insulin receptor (IR) interaction, and the mitogenic and metabolic properties of LAPS Insulin115. Furthermore, IR downregulation after long-term exposure to high concentrations of LAPS Insulin115 was analysed using an in vitro desensitization/resensitization model. RESULTS: The novel Fc-conjugated insulin derivative LAPS Insulin115 showed an extensively prolonged pharmacokinetic and pharmacodynamic profile in rodents. Despite its size of 59 kDa, LAPS Insulin115 passes the vascular endothelial barrier and induces insulin signalling in all major target tissues in rats. In vitro, LAPS Insulin115 showed a very slow onset of action because of its reduced IR affinity; however, after long-term stimulation it was equipotent in respect to its metabolic potency and showed no increased mitogenic action when compared with regular insulin. Remarkably, under conditions of chronic exposure, LAPS Insulin115 does not induce irreversible desensitization of target cells, which is probably attributable to much less prominent IR downregulation. CONCLUSION: Thus, LAPS Insulin115 exhibits a unique in vivo and in vitro profile and thereby represents an excellent candidate for a once-weekly insulin analogue.


Subject(s)
Drugs, Investigational/pharmacology , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Insulin, Long-Acting/pharmacology , Receptor, Insulin/agonists , Signal Transduction/drug effects , Absorption, Physiological , Animals , Cell Line , Cells, Cultured , Drugs, Investigational/chemistry , Drugs, Investigational/metabolism , Drugs, Investigational/therapeutic use , Half-Life , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fc Fragments/therapeutic use , Insulin, Long-Acting/genetics , Insulin, Long-Acting/metabolism , Insulin, Long-Acting/therapeutic use , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Male , Mice, Mutant Strains , Organ Specificity , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats, Wistar , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Toxicity Tests, Chronic
3.
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
4.
Front Pharmacol ; 7: 497, 2016.
Article in English | MEDLINE | ID: mdl-28101054

ABSTRACT

Background: Obesity is associated with impaired vascular function. In the cardiovascular system, protease-activated receptor 2 (PAR2) exerts multiple functions such as the control of the vascular tone. In pathological conditions, PAR2 is related to vascular inflammation. However, little is known about the impact of obesity on PAR2 in the vasculature. Therefore, we explored the role of PAR2 as a potential link between obesity and cardiovascular diseases. Methods: C57BL/6 mice were fed with either a chow or a 60% high fat diet for 24 weeks prior to isolation of aortas. Furthermore, human coronary artery endothelial cells (HCAEC) and human coronary smooth muscle cells (HCSMC) were treated with conditioned medium obtained from in vitro differentiated primary human adipocytes. To investigate receptor interaction vascular endothelial growth factor receptor 2 (VEGFR2) was blocked by exposure to calcium dobesilate and a VEGFR2 neutralization antibody, before treatment with PAR2 activating peptide. Student's t-test or one-way were used to determine statistical significance. Results: Both, high fat diet and exposure to conditioned medium increased PAR2 expression in aortas and human vascular cells, respectively. In HCSMC, conditioned medium elicited proliferation as well as cyclooxygenase 2 induction, which was suppressed by the PAR2 antagonist GB83. Specific activation of PAR2 by the PAR2 activating peptide induced proliferation and cyclooxygenase 2 expression which were abolished by blocking the VEGFR2. Additionally, treatment of HCSMC with the PAR2 activating peptide triggered VEGFR2 phosphorylation. Conclusion: Under obesogenic conditions, where circulating levels of pro-inflammatory adipokines are elevated, PAR2 arises as an important player linking obesity-related adipose tissue inflammation to atherogenesis. We show for the first time that the underlying mechanisms of these pro-atherogenic effects involve a potential transactivation of the VEGFR2 by PAR2.

5.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...