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










Publication year range
1.
Nat Commun ; 14(1): 4162, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443109

ABSTRACT

The current obesity epidemic and high prevalence of metabolic diseases necessitate efficacious and safe treatments. Brown adipose tissue in this context is a promising target with the potential to increase energy expenditure, however no pharmacological treatments activating brown adipose tissue are currently available. Here, we identify AXL receptor tyrosine kinase as a regulator of adipose function. Pharmacological and genetic inhibition of AXL enhance thermogenic capacity of brown and white adipocytes, in vitro and in vivo. Mechanistically, these effects are mediated through inhibition of PI3K/AKT/PDE signaling pathway, resulting in induction of nuclear FOXO1 localization and increased intracellular cAMP levels via PDE3/4 inhibition and subsequent stimulation of the PKA-ATF2 pathway. In line with this, both constitutive Axl deletion as well as inducible adipocyte-specific Axl deletion protect animals from diet-induced obesity concomitant with increases in energy expenditure. Based on these data, we propose AXL receptor as a target for the treatment of obesity.


Subject(s)
Adipose Tissue, Brown , Axl Receptor Tyrosine Kinase , Mice , Animals , Adipose Tissue, Brown/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Obesity/metabolism , Adipocytes, White/metabolism , Energy Metabolism , Adipose Tissue, White/metabolism , Thermogenesis/genetics , Adipocytes, Brown/metabolism , Mice, Inbred C57BL , Adipose Tissue/metabolism
2.
Peptides ; 167: 171047, 2023 09.
Article in English | MEDLINE | ID: mdl-37328068

ABSTRACT

The number of diabetic patients is rising globally and concomitantly so do the diabetes associated complications. The gut secretes a variety of proteins to control blood glucose levels and/or food intake. As the drug class of GLP-1 agonists is based on a gut secreted peptide and the positive metabolic effects of bariatric surgery are at least partially mediated by gut peptides, we were interested in other gut secreted proteins which have yet to be explored. In this respect we identified the gut secreted protein FAM3D by analyzing sequencing data from L- and epithelial cells of VSG and sham operated as well as chow and HFD fed mice. FAM3D was overexpressed in diet induced obese mice via an adeno-associated virus (AAV), which resulted in a significant improvement of fasting blood glucose levels, glucose tolerance and insulin sensitivity. The liver lipid deposition was reduced, and the steatosis morphology was improved. Hyperinsulinemic clamps indicated that FAM3D is a global insulin sensitizer and increases glucose uptake into various tissues. In conclusion, the current study demonstrated that FAM3D controls blood glucose levels by acting as an insulin sensitizing protein and improves hepatic lipid deposition.


Subject(s)
Fatty Liver , Insulin Resistance , Mice , Animals , Blood Glucose/metabolism , Insulin/metabolism , Liver/metabolism , Fatty Liver/metabolism , Peptides/pharmacology , Lipids , Mice, Inbred C57BL , Glucose/metabolism , Diet, High-Fat , Cytokines/metabolism
3.
Mol Metab ; 74: 101765, 2023 08.
Article in English | MEDLINE | ID: mdl-37390979

ABSTRACT

OBJECTIVE: Obesity is a major global health problem which can be targeted with new mechanistic diverse pharmacological interventions. Here we evaluate a new long-acting secretin receptor agonist as a potential treatment for obesity. METHODS: BI-3434 was designed as a secretin analog with stabilized peptide backbone and attached fatty acid-based half-life extension group. The peptide was evaluated in vitro for its ability to stimulate cAMP accumulation in a cell line stably expressing recombinant secretin receptor. On the functional level, stimulation of lipolysis in primary adipocytes after treatment with BI-3434 was determined. The ability of BI-3434 to activate secretin receptor in vivo was assessed in a cAMP reporter CRE-Luc mouse model. Furthermore, a diet-induced obesity mouse model was used to test the effects of BI-3434 on body weight and food intake following repeated daily subcutaneous administration alone and in combination with a GLP-1R agonist. RESULTS: BI-3434 potently activated human secretin receptor. However, lipolysis was only weakly induced in primary murine adipocytes. BI-3434 had an extended half-life compared to endogenous secretin and activated target tissues like pancreas, adipose tissue, and stomach in vivo. BI-3434 did not lower food intake in lean or diet-induced obese mice, but it increased energy expenditure after daily administration. This led to a loss of fat mass, which did not translate in a significant effect on body weight. However, treatment in combination with a GLP-1R agonist led to a synergistic effect on body weight loss. CONCLUSIONS: BI-3434 is a highly potent and selective agonist of secretin receptor with an extended pharmacokinetic (PK) profile. Increased energy expenditure after daily treatment with BI-3434 suggests that secretin receptor is involved in metabolic regulation and energy homeostasis. Targeting secretin receptor alone may not be an efficient anti-obesity treatment, but could be combined with anorectic principles like GLP-1R agonists.


Subject(s)
Gastrointestinal Hormones , Secretin , Mice , Humans , Animals , Secretin/pharmacology , Secretin/therapeutic use , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Body Weight , Peptides/pharmacology , Peptides/therapeutic use , Diet, High-Fat/adverse effects
4.
Nat Metab ; 4(1): 90-105, 2022 01.
Article in English | MEDLINE | ID: mdl-35027768

ABSTRACT

Healthy adipose tissue remodeling depends on the balance between de novo adipogenesis from adipogenic progenitor cells and the hypertrophy of adipocytes. De novo adipogenesis has been shown to promote healthy adipose tissue expansion, which confers protection from obesity-associated insulin resistance. Here, we define the role and trajectory of different adipogenic precursor subpopulations and further delineate the mechanism and cellular trajectory of adipogenesis, using single-cell RNA-sequencing datasets of murine adipogenic precursors. We identify Rspo2 as a functional regulator of adipogenesis, which is secreted by a subset of CD142+ cells to inhibit maturation of early progenitors through the receptor Lgr4. Increased circulating RSPO2 in mice leads to adipose tissue hypertrophy and insulin resistance and increased RSPO2 levels in male obese individuals correlate with impaired glucose homeostasis. Taken together, these findings identify a complex cellular crosstalk that inhibits adipogenesis and impairs adipose tissue homeostasis.


Subject(s)
Adipogenesis , Adipose Tissue/metabolism , Metabolic Networks and Pathways , Thrombospondins/genetics , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue/cytology , Animals , Computational Biology , Gene Expression Profiling , Gene Expression Regulation , Genetic Heterogeneity , Humans , Immunophenotyping , Insulin Resistance , Mice , Obesity/etiology , Obesity/metabolism , RNA-Seq , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins , Stem Cells/cytology , Stem Cells/metabolism , Thrombospondins/metabolism
5.
Nat Commun ; 12(1): 7144, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34880217

ABSTRACT

Activation of thermogenic brown and beige adipocytes is considered as a strategy to improve metabolic control. Here, we identify GPR180 as a receptor regulating brown and beige adipocyte function and whole-body glucose homeostasis, whose expression in humans is associated with improved metabolic control. We demonstrate that GPR180 is not a GPCR but a component of the TGFß signalling pathway and regulates the activity of the TGFß receptor complex through SMAD3 phosphorylation. In addition, using genetic and pharmacological tools, we provide evidence that GPR180 is required to manifest Collagen triple helix repeat containing 1 (CTHRC1) action to regulate brown and beige adipocyte activity and glucose homeostasis. In this work, we show that CTHRC1/GPR180 signalling integrates into the TGFß signalling as an alternative axis to fine-tune and achieve low-grade activation of the pathway to prevent pathophysiological response while contributing to control of glucose and energy metabolism.


Subject(s)
Extracellular Matrix Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Thermogenesis/physiology , Transforming Growth Factor beta/metabolism , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Animals , Energy Metabolism , Extracellular Matrix Proteins/genetics , Glucose , Homeostasis , Humans , Male , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Thermogenesis/genetics
7.
Commun Biol ; 4(1): 826, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34211098

ABSTRACT

Genome-wide association studies have identified SLC16A13 as a novel susceptibility gene for type 2 diabetes. The SLC16A13 gene encodes SLC16A13/MCT13, a member of the solute carrier 16 family of monocarboxylate transporters. Despite its potential importance to diabetes development, the physiological function of SLC16A13 is unknown. Here, we validate Slc16a13 as a lactate transporter expressed at the plasma membrane and report on the effect of Slc16a13 deletion in a mouse model. We show that Slc16a13 increases mitochondrial respiration in the liver, leading to reduced hepatic lipid accumulation and increased hepatic insulin sensitivity in high-fat diet fed Slc16a13 knockout mice. We propose a mechanism for improved hepatic insulin sensitivity in the context of Slc16a13 deficiency in which reduced intrahepatocellular lactate availability drives increased AMPK activation and increased mitochondrial respiration, while reducing hepatic lipid content. Slc16a13 deficiency thereby attenuates hepatic diacylglycerol-PKCε mediated insulin resistance in obese mice. Together, these data suggest that SLC16A13 is a potential target for the treatment of type 2 diabetes and non-alcoholic fatty liver disease.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease/genetics , Insulin Resistance/genetics , Lipid Metabolism/genetics , Monocarboxylic Acid Transporters/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Gene Expression , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Monocarboxylic Acid Transporters/deficiency , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Oxygen Consumption/genetics
8.
Mol Metab ; 51: 101237, 2021 09.
Article in English | MEDLINE | ID: mdl-33878401

ABSTRACT

BACKGROUND: A chronic imbalance of energy intake and energy expenditure results in excess fat storage. The obesity often caused by this overweight is detrimental to the health of millions of people. Understanding both sides of the energy balance equation and their counter-regulatory mechanisms is critical to the development of effective therapies to treat this epidemic. SCOPE OF REVIEW: Behaviors surrounding ingestion have been reviewed extensively. This review focuses more specifically on energy expenditure regarding bodyweight control, with a particular emphasis on the organs and attractive metabolic processes known to reduce bodyweight. Moreover, previous and current attempts at anti-obesity strategies focusing on energy expenditure are highlighted. Precise measurements of energy expenditure, which consist of cellular, animal, and human models, as well as measurements of their translatability, are required to provide the most effective therapies. MAJOR CONCLUSIONS: A precise understanding of the components surrounding energy expenditure, including tailored approaches based on genetic, biomarker, or physical characteristics, must be integrated into future anti-obesity treatments. Further comprehensive investigations are required to define suitable treatments, especially because the complex nature of the human perspective remains poorly understood.


Subject(s)
Energy Intake , Energy Metabolism/physiology , Obesity/therapy , Animals , Disease Models, Animal , Humans , Obesity/metabolism , Obesity/physiopathology
10.
Adipocyte ; 7(4): 277-284, 2018.
Article in English | MEDLINE | ID: mdl-30161013

ABSTRACT

Despite increased knowledge of nutrient intake regulation and energy homeostasis, treatment options for obesity remain limited. Food reward consists of two branches: gustatory and post-ingestive nutritive information. Drosophila lacking dSLC5A11 (sodium/glucose cotransporter 6-SGLT6) prefer L-glucose over D-glucose independently of their state of satiety. Human SGLT6 is an active transporter of myo-inositol and D-glucose. We investigated expression of SGLT6 in human tissue and found a significant expression in the small intestine and brain. The preference between a metabolizable and a non-metabolizable sugar was tested in 3 mouse models with a selective and potent SGLT6 inhibitor. No influence on sugar preference was seen with SGLT6 inhibition. These studies suggest that SGLT6 does not play a significant role in nutrient sensing in mammals.


Subject(s)
Anti-Obesity Agents/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Obesity/drug therapy , Obesity/metabolism , Symporters/antagonists & inhibitors , Symporters/metabolism , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/therapeutic use , Caco-2 Cells , Food Preferences/drug effects , Glucose/metabolism , HEK293 Cells , Humans , Inositol/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy
11.
Bioorg Med Chem ; 25(14): 3649-3657, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28528082

ABSTRACT

A potent, in vivo efficacious 11ß hydroxysteroid dehydrogenase type 1 (11ß HSD1) inhibitor (11j) has been identified. Compound 11j inhibited 11ß HSD1 activity in human adipocytes with an IC50 of 4.3nM and in primary human adipose tissue with an IC80 of 53nM. Oral administration of 11j to cynomolgus monkey inhibited 11ß HSD1 activity in adipose tissue. Compound 11j exhibited >1000× selectivity over other hydroxysteroid dehydrogenases, displays desirable pharmacodynamic properties and entered human clinical trials in 2011.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Oxazines/chemistry , Pyridones/chemistry , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Administration, Oral , Animals , Binding Sites , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Drug Evaluation, Preclinical , Half-Life , Inhibitory Concentration 50 , Macaca fascicularis , Molecular Docking Simulation , Oxazines/administration & dosage , Oxazines/pharmacokinetics , Protein Structure, Tertiary , Pyridones/administration & dosage , Pyridones/pharmacokinetics , Rats , Structure-Activity Relationship
12.
Eur J Pharmacol ; 789: 68-74, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27393460

ABSTRACT

Inhibition of local cortisol regeneration from circulating cortisone by blocking 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) has been shown to ameliorate the risk factors associated with the metabolic syndrome. Chronic modulation of glucocorticoid homeostasis may result in hypothalamic-pituitary-adrenal (HPA) axis stimulation. HPA axis over-activation leading androgen excess would be undesirable in a therapeutic intervention designed to treat a chronic condition such as the metabolic syndrome. To address whether 11ß-HSD1 inhibition would lead to excess androgens, we treated female cynomolgus monkeys with a selective inhibitor, BI 135558, for 4 weeks. Continual action of the compound over the dosing period was confirmed by constant plasma exposure, and a maintained change in urinary glucocorticoid metabolites consistent with 11ß-HSD1 inhibition. No significant changes in adrenal function, as evidenced by an adrenocorticotropic hormone (ATCH) challenge, were observed. An examination of androgenic hormones revealed a slight increase in dehydroepiandrosterone sulfate (DHEA-S), while other hormones such as testosterone remained within reference values. Overall, treatment with BI 135558 in monkeys did not result in obvious over-activation of the HPA axis.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Oxazines/pharmacology , Pituitary-Adrenal System/drug effects , Pyridones/pharmacology , Animals , Enzyme Inhibitors/pharmacokinetics , Female , Hypothalamo-Hypophyseal System/physiology , Macaca fascicularis , Oxazines/pharmacokinetics , Pituitary-Adrenal System/physiology , Pyridones/pharmacokinetics , Time Factors
13.
Eur J Pharmacol ; 746: 50-5, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25445047

ABSTRACT

To combat the increased morbidity and mortality associated with the developing diabetes epidemic new therapeutic interventions are desirable. Inhibition of intracellular cortisol generation from cortisone by blocking 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) has been shown to ameliorate the risk factors associated with the metabolic syndrome. A challenge in developing 11ß-HSD1 inhibitors has been the species selectivity of small molecules, as many compounds are primate specific. Here we describe our strategy to identify potent selective 11ß-HSD1 inhibitors while ensuring target engagement in key metabolic tissues, liver and fat. This strategy enabled the identification of the clinical candidate, BI 135585.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Diabetes Mellitus, Type 2/drug therapy , Enzyme Inhibitors/pharmacology , Oxazines/pharmacology , Pyridones/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Catalytic Domain , Diabetes Mellitus, Type 2/enzymology , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Macaca fascicularis , Male , Models, Molecular , Oxazines/chemistry , Oxazines/therapeutic use , Pyridones/chemistry , Pyridones/therapeutic use
14.
Mol Cell Biochem ; 369(1-2): 175-81, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22773307

ABSTRACT

Alongside cell lines such as 3T3-L1 cells, primary cell culture models of adipogenesis have helped in developing an understanding of the process of adipocyte recruitment and maintenance, which may lead to therapeutic advances to treat the growing epidemic of obesity. Recently, it has been demonstrated that fat cell progenitors (DFAT) established through ceiling culture of adipocytes retain an enhanced ability to undergo adipocyte differentiation compared to preadipocytes isolated from the stromal vascular fraction of adipose tissue. Clonal expansion of rat DFAT cells identified differentiation capable and incapable cell strains. To understand the mechanisms underlying these differences, comparison of their transcriptomes by next generation sequencing was performed. Two hundred seventy-eight genes with a significant fold change of 1.4 were detected as being consistently deregulated between differentiating and non-differentiating strains. Bioinformatic network analyses identified components of the extra-cellular matrix and PPARγ as important genes in this process, suggesting crosstalk between ECM and transcription factors influences differentiation. Analyses of the transcriptomes of human DFAT cells in early and late passage (non-differentiating) confirmed the importance of these pathways in maintaining an adipogenic potential.


Subject(s)
Adipocytes , Cell Differentiation , Extracellular Matrix/metabolism , Gene Expression Profiling , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis , Animals , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Mice , PPAR gamma/metabolism , Rats , Transcription Factors/metabolism
15.
Bioorg Med Chem Lett ; 21(19): 5924-7, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21873051

ABSTRACT

Based on a high-throughput screen, cyclopentanecarboxanilides were identified as a new chemotype of non-covalent inhibitors of type I fatty acid synthase (FAS). Starting from initial hits we aimed at generating a tool compound suitable for the in vivo validation of FAS as a therapeutic target. Optimisation yielded BI 99179 which is characterised by high potency, remarkably high selectivity and significant exposure (both peripheral and central) upon oral administration in rats.


Subject(s)
Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacology , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Proline/analogs & derivatives , Administration, Oral , Animals , Benzoxazoles/pharmacokinetics , Benzoxazoles/toxicity , Caco-2 Cells , Cell Line , Cytochrome P-450 Enzyme Inhibitors , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , High-Throughput Screening Assays , Humans , Hypothalamus , Inhibitory Concentration 50 , Injections, Intravenous , Mice , Microsomes, Liver/metabolism , Molecular Conformation , Molecular Targeted Therapy , Permeability , Proline/chemical synthesis , Proline/pharmacokinetics , Proline/pharmacology , Proline/toxicity , Rats , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity
16.
Mol Cell Biochem ; 343(1-2): 257-69, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20574786

ABSTRACT

With increasing rates of obesity driving the incidence of type 2 diabetes and cardiovascular diseases to epidemic levels, understanding of the biology of adipose tissue expansion is a focus of current research. Identification and characterization of secreted proteins of the adipose tissue could provide further insights into the function of adipose tissue and might help to therapeutically influence the development of obesity and associated metabolic disorders. In the present study, we identified human epidermal growth factor-like domain multiple-6 (EGFL6) as an adipose tissue-secreted protein. EGFL6 expression in human subcutaneous adipose tissue significantly increased with obesity and decreased after weight loss. Further, expression and secretion of EGFL6 increased with in vitro differentiation of human preadipocytes, suggesting that mature adipocytes are the main source of EGFL6. Containing epidermal growth factor (EGF)-like repeats, an Arg-Gly-Asp (RGD) integrin binding motif and a mephrin, A5 protein and receptor protein-tyrosine phosphatase mu (MAM) domain, EGFL6 was suggested to be an extra-cellular matrix protein. Recombinant human EGFL6 protein mediated cell adhesion of human adipose tissue-derived stromal vascular cells (AD-SVC) in an RGD-dependent manner. FACS analyses revealed specific binding of the protein to the cell surface of AD-SVC with the binding being predominantly mediated by the EGF-like repeats. Recombinant EGFL6 enhanced proliferation of human AD-SVC as measured by MTS assay and [(14)C]-thymidine incorporation. These results indicate that human EGFL6 is a paracrine/autocrine growth factor of adipose tissue up-regulated in obesity and potentially involved in the process of adipose tissue expansion and the development of obesity.


Subject(s)
Adipose Tissue/metabolism , Blood Vessels/metabolism , Cell Proliferation , Membrane Glycoproteins/genetics , Obesity/genetics , Stromal Cells/metabolism , Adipose Tissue/cytology , Blood Vessels/cytology , Calcium-Binding Proteins , Cell Adhesion , Cell Adhesion Molecules , Cell Separation , Chromatography, Liquid , Culture Media, Conditioned , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology , Tandem Mass Spectrometry , Weight Loss
17.
Eur J Pharmacol ; 580(1-2): 55-62, 2008 Feb 02.
Article in English | MEDLINE | ID: mdl-18031735

ABSTRACT

Small molecules interacting with aminergic G-protein coupled receptors represent a number of very successful drugs. G-protein coupled receptors continue to be a significant group of targets for pharmaceutical intervention, and modifying their activity through small molecules is a major focus of drug development. Previously, these small molecules could be easily fit in models, as agonists, partial agonists or antagonists. More recently, however, these lines have been blurred as it is increasingly recognized that ligands can interact with receptors in various ways. Analysis of beta-adrenoceptors has revealed that several sites or states exist for the individual receptors. The putative atypical beta(4)-adrenoceptor identified on heart and adipose tissue is now recognized as a unique beta(1)-adrenoceptor state. Similarly, a unique beta(3)-adrenoceptor state has been identified using the aryloxypropanolamine CGP-12,177 and cloned receptor systems. Here we expand upon these observations, by describing an atypical state of the beta(3)-adrenoceptor that exists endogenously in adipose tissue. Furthermore, we describe novel arylethanolamine ligands that interact with this atypical state of the beta(3)-adrenoceptor with high affinity and provide additional tools to investigate the atypical beta(3)-adrenoceptor state to determine whether it can be influenced for therapeutic purposes.


Subject(s)
Adipocytes, White/metabolism , Lipolysis/physiology , Receptors, Adrenergic, beta-3/metabolism , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Cyclic AMP/metabolism , Ethanolamines/pharmacology , Female , Ligands , Lipolysis/drug effects , Male , Mice , Mice, Knockout , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-3/genetics
18.
Biochem Biophys Res Commun ; 328(4): 1073-82, 2005 Mar 25.
Article in English | MEDLINE | ID: mdl-15707987

ABSTRACT

Inhibition of fatty acid synthase (FAS) reduces food intake in rodents. As adipose tissue expresses FAS, we sought to investigate the effect of reduced FAS activity on adipocyte differentiation. FAS activity was suppressed either pharmacologically or by siRNA during differentiation of 3T3-L1 cells. Cerulenin (10 microM), triclosan (50 microM), and C75 (50 microM) reduced dramatically visible lipid droplet accumulation, while incorporation of [1-(14C)]acetate into lipids was reduced by 75%, 70%, and 90%, respectively. Additionally, the substances reduced FAS, CEBPalpha, and PPARgamma mRNA by up to 85% compared to that of control differentiated cells. Transient transfection with FAS siRNA suppressed FAS mRNA and FAS activity, and this was accompanied by reduction of CEBPalpha and PPARgamma mRNA levels, and complete prevention of lipid accumulation. CD36, a late marker of differentiation, was also reduced. Together, these results suggest that FAS generated signals may be essential to support preadipocyte differentiation.


Subject(s)
4-Butyrolactone/analogs & derivatives , Adipocytes/cytology , Adipocytes/enzymology , Cell Differentiation/drug effects , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , 3T3-L1 Cells , 4-Butyrolactone/pharmacology , Adipocytes/drug effects , Animals , Cell Differentiation/physiology , Cerulenin/pharmacology , Fatty Acid Synthases/genetics , Gene Silencing , Mice , Triclosan/pharmacology
19.
Regul Pept ; 111(1-3): 21-9, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12609745

ABSTRACT

Recently, an orphan G protein coupled receptor (GPCR) termed NPGPR was described. A shorter variant of this receptor lacking exon 1 was shown to have subnanomolar affinity for neuropeptide FF (NPFF), a pain modulatory peptide, and therefore was named NPFF(2) receptor. Here, we characterize the full-length cloned NPGPR and identify a novel short form lacking exon 2 with a differential pattern of mRNA abundance in several tissues and organs. The NPGPR is most similar to the recently cloned neuropeptide FF (NPFF) receptor which lacks exon 1, but also shows high homology to the orexin and neuropeptide Y (NPY) receptor families, two neuropeptides involved in food intake regulation. Therefore, we used binding studies to examine the interaction of NPFF, orexin and NPY with the NPGPR. [125I] NPFF was displaced by NPFF with an IC(50) of 14.7 +/- 8.8 nM, whereas [125I] Orexin B was displaced by Orexin B with an IC(50) of 415 +/- 195 nM. We conclude that orexins interact with the NPGPR and that the affinity of NPFF for NPGPR is approximately 100-fold lower than for the NPFF2 receptor. We postulate that NPGPR is a splice variant of the family of NPFF receptors and displays a binding profile different from the other members of the NPFF receptor family due to the presence of exon 1. In order to evaluate whether NPGPR levels are affected by the feeding status, we examined the mRNA level using real-time PCR in two feeding models, i.e. before and after diet-induced body weight increase as well as after chronic food restriction in rats. However, hypothalamic NPGPR mRNA was unchanged in both models. Therefore, our evidence does not support the hypothesis that NPGPR is involved in feeding regulation.


Subject(s)
Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins , Pregnancy Proteins/genetics , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Amino Acid Sequence , Animals , CHO Cells , Carrier Proteins/metabolism , Cloning, Molecular , Cricetinae , Exons , Humans , Molecular Sequence Data , Neuropeptides/metabolism , Orexin Receptors , Orexins , Pregnancy Proteins/metabolism , Protein Isoforms/genetics , Protein Splicing/physiology , RNA, Messenger/metabolism , Radioligand Assay , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Tissue Distribution , Transfection
20.
Obes Res ; 10(3): 182-7, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11886941

ABSTRACT

OBJECTIVE: To examine the effects of a cafeteria diet and a chronic treatment with melanocortin agonist (MTII) on mature weight-stable female rats. RESEARCH METHODS AND PROCEDURES: Ex-breeder Chbb:Thom rats (350 to 400 g) were divided into two groups: highly palatable food (HPF) and normal rat chow (RC). Both groups had ab libitum access to rat chow. The HPF group had access to chocolate bars, cookies, cheese, and nuts (approximately 20 g/d). After 21 days, the rats in each group were then divided into control and treated groups. Mini-pumps delivering saline or MTII (1 mg/kg per day) for minimally 28 days were implanted. Oxygen consumption was measured for 17 days in a second group of rats implanted with mini-pumps containing MTII (1 mg/kg per day) or saline. RESULTS: HPF rats ate less (<50%) rat chow than RC rats. After 20 days, the HPF group had reached a plateau and weighed significantly more (p < 0.005) than the RC group (411.7 +/- 9.3 g; n = 17 vs. 365.1 +/- 9.4 g; n = 16). HPF rats and RC rats receiving MTII reduced their pellet intake and body weight in the initial 2 weeks of treatment (day 14, RC-saline: -1.6 +/- 1.8 g; RC-MTII, -22.5 +/- 3.7 g; HPF-saline, -7.1 +/- 1.7 g; HPF-MTII, -30.7 +/- 4.8 g). Subsequently, pellet intake returned to pre-implantation values, although body weights remained reduced in both HPF and RC groups. Oxygen consumption was increased in rats treated with MTII. DISCUSSION: This suggests that MTII initially reduced body weight by limiting food intake; however, maintenance of weight is most likely due to increased energy expenditure under conditions of normal and highly palatable diets in mature animals.


Subject(s)
Eating/drug effects , Obesity/drug therapy , Weight Loss/drug effects , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology , Animals , Blood Glucose/analysis , Calorimetry, Indirect , Disease Models, Animal , Female , Insulin/blood , Leptin/blood , Male , Obesity/metabolism , Oxygen Consumption/drug effects , Rats , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...