Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Diabetes Obes Metab ; 17(1): 98-101, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25238025

ABSTRACT

The sodium-dependent glucose transporter 2 (SGLT2) inhibitor remogliflozin etabonate (RE) was evaluated in a 12-week, double-blind, randomized, placebo- and active-controlled, parallel-group study. A total of 252 newly diagnosed and drug-naïve people with type 2 diabetes and glycated haemoglobin (HbA1c) concentrations of 7.0-≤9.5% (53-80 mmol/mol) were recruited. Participants were randomized to RE (100, 250, 500 or 1000 mg once daily or 250 mg twice daily), placebo or 30 mg pioglitazone once daily. The primary endpoint was change in HbA1c concentration from baseline. Secondary endpoints included changes in fasting plasma glucose, body weight and lipid profiles, safety and tolerability. We observed a statistically significant trend in the RE dose-response relationship for change from baseline in HbA1c at week 12 (p < 0.047). RE was generally well tolerated and no effects on LDL cholesterol were observed.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucosides/administration & dosage , Hyperglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Membrane Transport Modulators/administration & dosage , Prodrugs/administration & dosage , Pyrazoles/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors , Cohort Studies , Diabetes Mellitus, Type 2/blood , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Follow-Up Studies , Glucosides/adverse effects , Glucosides/therapeutic use , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Intention to Treat Analysis , Membrane Transport Modulators/adverse effects , Membrane Transport Modulators/therapeutic use , Middle Aged , Patient Dropouts , Pioglitazone , Prodrugs/adverse effects , Prodrugs/therapeutic use , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Thiazolidinediones/administration & dosage , Thiazolidinediones/adverse effects , Thiazolidinediones/therapeutic use , Weight Loss/drug effects
2.
Diabetes Obes Metab ; 17(1): 94-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25223369

ABSTRACT

We compared the efficacy of twice-daily doses of remogliflozin etabonate (RE) and once-daily pioglitazone with placebo for reduction in glycated haemoglobin (HbA1c) concentration. In this 12-week, double-blind, randomized, active- and placebo-controlled trial, 336 treatment-naïve subjects with type 2 diabetes and an HbA1c of 7.0-9.5% (53-80 mmol/mol) were randomized to RE (50, 100, 250, 500 or 1000 mg twice daily), matching placebo or 30 mg pioglitazone once daily. The primary endpoint was change in HbA1c from baseline. Other endpoints included changes in body weight, lipid levels, safety and tolerability. RE produced a decreasing dose response in HbA1c at week 12 (p < 0.001), with reductions in HbA1c versus placebo ranging from 0.64 to 1.07% (p < 0.001). Statistically significant reductions in body weight for RE compared with placebo were also observed. Twice-daily RE resulted in a dose-ordered improvement in glycaemic control and was generally well tolerated.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucosides/administration & dosage , Hyperglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Membrane Transport Modulators/administration & dosage , Prodrugs/administration & dosage , Pyrazoles/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Follow-Up Studies , Glucosides/adverse effects , Glucosides/therapeutic use , Glycated Hemoglobin/analysis , Humans , Hypertriglyceridemia/complications , Hypertriglyceridemia/prevention & control , Hypoglycemia/prevention & control , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Intention to Treat Analysis , Membrane Transport Modulators/adverse effects , Membrane Transport Modulators/therapeutic use , Middle Aged , Patient Dropouts , Pioglitazone , Prodrugs/adverse effects , Prodrugs/therapeutic use , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Thiazolidinediones/administration & dosage , Thiazolidinediones/adverse effects , Thiazolidinediones/therapeutic use , Weight Loss/drug effects
3.
Tissue Eng ; 7(6): 729-41, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11749730

ABSTRACT

Human adipose tissue represents an abundant reservoir of stromal cells with potential utility for tissue engineering. The current study demonstrates the ability of human adipose tissue-derived stromal cells to display some of the hallmarks of osteoblast differentiation in vitro. Following treatment with ascorbate, beta-glycerophosphate, dexamethasone, and 1,25 dihydroxy vitamin D(3), adipose tissue-derived stromal cells mineralize their extracellular matrix based on detection of calcium phosphate deposits using Alizarin Red and von Kossa histochemical stains. Fourier transform infrared analysis demonstrates the apatitic nature of these crystals. Mineralization is accompanied by increased expression or activity of the osteoblast-associated proteins osteocalcin and alkaline phosphatase. These and other osteoblast-associated gene markers are detected based on polymerase chain reaction. In contrast, the adipocyte gene markers--leptin, lipoprotein lipase, and peroxisome proliferator activated receptor gamma2--are reduced under mineralization conditions, consistent with the reciprocal relationship postulated to exist between adipocytes and osteoblasts. The current work supports the presence of a multipotent stromal cell population within human extramedullary adipose tissue. These findings have potential implications for human bone tissue bioengineering.


Subject(s)
Adipose Tissue/cytology , Calcification, Physiologic/physiology , Gene Expression Regulation/physiology , Osteoblasts/physiology , Stromal Cells/physiology , Cell Differentiation/physiology , Extracellular Matrix/metabolism , Humans , Osteoblasts/cytology , Stromal Cells/cytology , Tissue Engineering
4.
J Lipid Res ; 42(10): 1543-51, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11590209

ABSTRACT

Fenofibrate is a member of the fibrate class of hypolipidemic agents used clinically to treat hypertriglyceridemia and mixed hyperlipidemia. The fibrates were developed primarily on the basis of their cholesterol and triglyceride lowering in rodents. Fibrates have historically been ineffective at lowering triglycerides in experimentally-induced dyslipidemia in nonhuman primate models. The spontaneously obese rhesus monkey is a well-recognized animal model for the study of human obesity and type 2 diabetes, and many of these monkeys exhibit naturally occurring lipid abnormalities, including elevated triglycerides and low HDL cholesterol (HDL-C), similar to patients with type 2 diabetes. To explore whether the obese rhesus model was predictive of the lipid lowering effects of fibrates, we evaluated fenofibrate in six hypertriglyceridemic, hyperinsulinemic, nondiabetic animals in a 20-week, dose-escalating study. The study consisted of a 4-week baseline period, two treatment periods of 10 mg/kg twice daily (b.i.d) for 4 weeks and 30 mg/kg b.i.d. for 8 weeks, and a 4-week washout period. Fenofibrate (30 mg/kg b.i.d) decreased serum triglycerides 55% and LDL-C 27%, whereas HDL-C increased 35%. Apolipoproteins B-100 and C-III levels were also reduced 70% and 29%, respectively. Food intake, body weight, and plasma glucose were not affected throughout the study. Interestingly, plasma insulin levels decreased 40% during the 30 mg/kg treatment period, suggesting improvement in insulin sensitivity. These results support the use of obese rhesus monkey as an excellent animal model for studying the effects of novel hypolipidemic agents, particularly agents that impact serum triglycerides and HDL-C.


Subject(s)
Fenofibrate/pharmacology , Lipid Metabolism , Macaca mulatta/metabolism , Obesity/metabolism , Amino Acid Sequence , Animals , Apolipoproteins/blood , Base Sequence , Blood Glucose/metabolism , Blotting, Western , Body Weight , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Cloning, Molecular , Disease Models, Animal , Dose-Response Relationship, Drug , Fenofibrate/administration & dosage , Fenofibrate/therapeutic use , Gene Expression Profiling , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Insulin/blood , Lipids/blood , Macaca mulatta/blood , Male , Molecular Sequence Data , Obesity/blood , Obesity/drug therapy , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Triglycerides/blood , Triglycerides/metabolism
5.
Bioorg Med Chem Lett ; 11(9): 1225-7, 2001 May 07.
Article in English | MEDLINE | ID: mdl-11354382

ABSTRACT

Using solid-phase, parallel-array synthesis, a series of urea-substituted thioisobutyric acids was synthesized and assayed for activity on the human PPAR subtypes. GW7647 (3) was identified as a potent human PPARalpha agonist with approximately 200-fold selectivity over PPARgamma and PPARdelta, and potent lipid-lowering activity in animal models of dyslipidemia. GW7647 (3) will be a valuable chemical tool for studying the biology of PPARalpha in human cells and animal models of disease.


Subject(s)
Hypolipidemic Agents/chemical synthesis , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Animals , Butyrates/pharmacology , Cricetinae , Dietary Fats/pharmacology , Drug Design , Hyperlipidemias/blood , Hyperlipidemias/drug therapy , Hypolipidemic Agents/pharmacology , Phenylurea Compounds/pharmacology
6.
Metabolism ; 50(4): 407-13, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11288034

ABSTRACT

While adipocyte differentiation has been studied extensively in murine cultures, the lack of a readily available preadipocyte model has hindered equivalent studies in man. We describe methods for the isolation and culture of primary human stromal cells from surgical adipose tissue specimens. In vitro, the stromal cells rapidly differentiate in response to a combination of adipogenic agents. Among these, glucocorticoids and thiazolidinediones act together to induce the formation of lipid vacuoles within the cells. These morphologic changes accompany the increased expression of 2 characteristic adipocyte proteins, the cytoplasmic enzyme glycerol phosphate dehydrogenase (GPDH) and the secreted cytokine leptin. Likewise, stromal cell differentiation results in elevated mRNA levels for the fatty acid binding protein aP2 and the adipogenic regulatory transcription factors CCAAT/enhancer binding protein alpha (C/EBPalpha) and peroxisome proliferator-activated receptor gamma (PPARgamma) in addition to leptin. The in vitro differentiated stromal cells exhibit a lipolytic response to beta-adrenergic agonists, comparable to that reported with primary human adipocytes. These studies demonstrate the validity of human adipose tissue-derived stromal cells as a reliable in vitro model for investigations of adipocyte metabolism in humans.


Subject(s)
Adipose Tissue/cytology , Glucocorticoids/pharmacology , Stromal Cells/drug effects , Thiazoles/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Cell Differentiation/drug effects , Coloring Agents , Glycerolphosphate Dehydrogenase/metabolism , Humans , Immunoblotting , In Vitro Techniques , Leptin/biosynthesis , Lipectomy , Lipolysis/drug effects , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism
7.
J Cell Biochem ; 81(2): 312-9, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11241671

ABSTRACT

The current study was done to assess if heterogeneity existed in the degree of adipogenesis in stromal cells (preadipocytes) from multiple donors. In addition to conventional lipid-based methods, we have employed a novel signal amplification technology, known as branched DNA, to monitor expression of an adipocyte specific gene product aP2. The fatty acid binding protein aP2 increases during adipocyte differentiation and is induced by thiazolidinediones and other peroxisome proliferator activated receptor gamma ligands. The current work examined the adipogenic induction of aP2 mRNA levels in human adipose tissue stromal cells derived from 12 patients (mean age +/- SEM, 38.9 +/- 3.1) with mild to moderate obesity (mean body mass index +/- SEM, 27.8 +/- 2.4). Based on branched DNA technology, a rapid and sensitive measure of specific RNAs, the relative aP2 level in adipocytes increased by 679 +/- 93-fold (mean +/- SEM, n=12) compared to preadipocytes. Normalization of the aP2 mRNA levels to the housekeeping gene, glyceraldehyde phosphate dehydrogenase, did not significantly alter the fold induction in a subset of 4 patients (803.6 +/- 197.5 vs 1118.5 +/- 308.1). Independent adipocyte differentiation markers were compared between adipocytes and preadipocytes in parallel studies. Leptin secretion increased by up to three-orders of magnitude while measurements of neutral lipid accumulation by Oil Red O and Nile Red staining increased by 8.5-fold and 8.3-fold, respectively. These results indicate that preadipocytes isolated from multiple donors displayed varying degrees of differentiation in response to an optimal adipogenic stimulus in vitro. This work also demonstrates that branched DNA measurement of aP2 is a rapid and sensitive measure of adipogenesis in human stromal cells. The linear range of this assay extends up to three-orders of magnitude and correlates directly with independent measures of cellular differentiation.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Neoplasm Proteins , Tumor Suppressor Proteins , Adult , Azo Compounds/pharmacology , Carrier Proteins/metabolism , Cell Differentiation , Cell Division , DNA/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Fluorescent Dyes/pharmacology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Leptin/metabolism , Obesity/metabolism , Oxazines/pharmacology , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
Physiol Genomics ; 5(1): 11-9, 2001 Feb 07.
Article in English | MEDLINE | ID: mdl-11161002

ABSTRACT

Mouse melanocortin receptors, MC1-R, MC3-R, MC4-R, and MC5-R, when expressed in HEK293 cells and stimulated with either alpha-melanocyte-stimulating hormone (alpha-MSH) or desacetyl-alpha-MSH, mediate increases in intracellular free calcium concentration ([Ca(2+)](i)) with EC(50) values between 0.3 and 4.3 nM. The increase in [Ca(2+)](i) is cholera toxin sensitive and pertussis toxin insensitive. The mechanism involves calcium mobilization from intracellular stores without a transient rise in inositol trisphosphate. Mouse agouti protein (55 nM) is a competitive antagonist of alpha-MSH (6-fold) and desacetyl-alpha-MSH (8-fold), coupling the mMC1-R to increased [Ca(2+)](i). Agouti protein (55 nM) significantly increased the EC(50) for alpha-MSH (3-fold), and 550 nM agouti protein significantly increased the EC(50) for desacetyl-alpha-MSH (4-fold), coupling the mMC4-R to a rise in [Ca(2+)](i). However, agouti protein antagonism of the MC4-R may not be competitive since there was a trend for the maximum response to also increase. There was no significant antagonism of the MC3-R and MC5-R by agouti protein (55 nM). Understanding the physiological relevance of the transduction of a calcium signal by melanocortin peptides may be important for future development of therapeutic targeting of the melanocortin receptors.


Subject(s)
Calcium/metabolism , Intercellular Signaling Peptides and Proteins , Receptor, Melanocortin, Type 3 , Receptors, Corticotropin/physiology , alpha-MSH/analogs & derivatives , Agouti Signaling Protein , Animals , Calcium Channel Blockers/pharmacology , Carbachol/pharmacology , Cell Line , Cholera Toxin/pharmacology , Dose-Response Relationship, Drug , Humans , Inositol Phosphates/metabolism , Ionomycin/pharmacology , Manganese/pharmacology , Mice , Peptide Fragments/pharmacology , Proteins/pharmacology , Receptors, Melanocortin , Signal Transduction/drug effects , Thapsigargin/pharmacology , alpha-MSH/pharmacology
9.
Int J Obes Relat Metab Disord ; 24 Suppl 4: S41-4, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11126240

ABSTRACT

Many organs contain connective tissue or stromal cells and these cells play important roles in growth, development and tissue repair. Subcutaneous adipose tissue represents an accessible reservoir for the isolation of human stromal cells. Ex vivo, the adipose tissue-derived human stromal cells can be expanded more than 100-fold. These primary cultures respond to adipogenic agonists by accumulating lipid and expressing adipocyte specific proteins, including leptin and the peroxisome proliferator-activated receptor gamma (PPARgamma). In contrast, when the adipose tissue-derived stromal cells are exposed to osteogenic factors, they display osteoblastic gene markers and mineralize their extracellular matrix. This work demonstrates that subcutaneous adipose tissue is a readily available source of multipotential stromal cells. It is possible that these cells will be used clinically to treat a broad range of orthopedic, rheumatologic and periodontal disorders.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Bone Regeneration , Cell Differentiation , Bone Marrow Cells/cytology , Cell Differentiation/genetics , Gene Expression Regulation , Humans , Stromal Cells/cytology
10.
Physiol Genomics ; 3(2): 75-82, 2000 Aug 09.
Article in English | MEDLINE | ID: mdl-11015602

ABSTRACT

Intracellular calcium ([Ca(2+)](i)) modulates adipocyte lipid metabolism and inhibits the early stages of murine adipogenesis. Consequently, we evaluated effects of increasing [Ca(2+)](i) in early and late stages of human adipocyte differentiation. Increasing [Ca(2+)](i) with either thapsigargin or A23187 at 0-1 h of differentiation markedly suppressed differentiation, with a 40-70% decrease in triglyceride accumulation and glycerol-3 phosphate dehydrogenase (GPDH) activity (P < 0.005). However, a 1-h pulse of either agent at 47-48 h only modestly inhibited differentiation. Sustained, mild stimulation of Ca(2+) influx with either agouti protein or 10 mM KCl-induced depolarization during 0-48 h of differentiation inhibited triglyceride accumulation and GPDH activity by 20-70% (P < 0.05) and markedly suppressed peroxisome proliferator-activated receptor gamma (PPARgamma) expression. These effects were reversed by Ca(2+) channel antagonism. In contrast, Ca(2+) pulses late in differentiation (71-72 h or 48-72 h) markedly increased these markers of differentiation. Thus increasing [Ca(2+)](i) appears to exert a biphasic regulatory role in human adipocyte differentiation, inhibiting the early stages while promoting the late stage of differentiation and lipid filling.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Calcium Signaling , Calcium/metabolism , Cell Differentiation , Intercellular Signaling Peptides and Proteins , Adipocytes/drug effects , Adipocytes/enzymology , Agouti Signaling Protein , Calcimycin/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Glycerolphosphate Dehydrogenase/metabolism , Humans , Ionophores/pharmacology , Nitrendipine/pharmacology , Potassium Chloride/pharmacology , Proteins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/enzymology , Stem Cells/metabolism , Thapsigargin/pharmacology , Time Factors , Transcription Factors/genetics , Triglycerides/metabolism
11.
Physiol Genomics ; 3(3): 157-62, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-11015611

ABSTRACT

Mice carrying dominant mutations at the agouti locus exhibit ectopic expression of agouti gene transcripts, obesity, and type II diabetes through unknown mechanisms. To gain insight into the role of agouti protein in modulating adiposity, we investigated regulation of a key lipogenic gene, fatty acid synthase (FAS) by agouti alone and in combination with insulin. Both agouti and insulin increase FAS activity in 3T3-L1 and in human adipocytes. Agouti and insulin independently and additively increase FAS activity in 3T3-L1 adipocytes. We further investigated the mechanism responsible for the agouti-induced FAS expression in these cells and demonstrated that both insulin (3-fold increase) and agouti (2-fold) increased FAS gene expression at the transcriptional level. Furthermore, insulin and agouti together exerted additive effects (5-fold increase) on FAS gene transcription. Transfection assays of FAS promoter-luciferase fusion gene constructs into 3T3-L1 adipocytes indicated that the agouti response element(s) is (are) located in the -435 to -415 region (-435/-415) of the FAS promoter. Nuclear proteins binding to this novel sequence are adipocyte specific. Thus the agouti response sequences mapped to a region upstream of the insulin-responsive element (which we previously reported to be located at -67/-52), consistent with additive effects of these two factors on FAS gene transcription.


Subject(s)
Adipocytes/enzymology , Fatty Acid Synthases/genetics , Intercellular Signaling Peptides and Proteins , Proteins/pharmacology , 3T3 Cells , Adipocytes/cytology , Agouti Signaling Protein , Animals , DNA/genetics , Drug Interactions , Gene Expression Regulation, Enzymologic/drug effects , Humans , Insulin/pharmacology , Luciferases/genetics , Luciferases/metabolism , Mice , Nuclear Proteins/metabolism , Oligonucleotides/genetics , Oligonucleotides/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Proteins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Transcription, Genetic/drug effects , Tumor Cells, Cultured
13.
FASEB J ; 13(13): 1833-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10506587

ABSTRACT

A regulatory role for intracellular Ca2+ ([Ca2+]i) in adipocyte lipogenesis, lipolysis and triglyceride accumulation has been demonstrated. Compounds acting on the pancreatic sulfonylurea receptor (SUR) to increase (e.g., glibenclamide) or decrease (e.g., diazoxide) [Ca2+]i cause corresponding increases and decreases in weight gain. However, these weight gain and loss effects have been attributed to insulin release rather than to the primary effects of these compounds on the adipocyte SUR and its associated K(ATP) channel. Accordingly, we have evaluated the direct role of the human adipocyte SUR in regulating adipocyte metabolism. We used RT-PCR with primers for a highly conserved region of SUR1 to demonstrate that human adipocytes express SUR1. The PCR product was confirmed by sequence analysis and used as a probe to demonstrate adipocyte SUR1 expression by Northern blot analysis. Adipocytes exhibited glibenclamide dose-responsive (0-20 microM) increases in [Ca2+]i (P<0.05). Similarly, glibenclamide (10 microM) caused a 67% increase in adipocyte fatty acid synthase activity (P<0.001), a 48% increase in glycerol-3-phosphate dehydrogenase activity (P<0.01) and a 68% inhibition in lipolysis (P<0.01), whereas diazoxide (10 microM) completely prevented each of these effects. These data demonstrate that human adipocytes express a SUR that regulates [Ca2+]i and, consequently, exerts coordinate control over lipogenesis and lipolysis. Accordingly, the adipocyte SUR1 may represent an important target for the development of therapeutic interventions in obesity.


Subject(s)
ATP-Binding Cassette Transporters , Adipocytes/metabolism , Calcium/metabolism , Lipid Metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Diazoxide/pharmacology , Dose-Response Relationship, Drug , Fatty Acid Synthases/drug effects , Glyburide/pharmacology , Glycerolphosphate Dehydrogenase/drug effects , Humans , Lipolysis/drug effects , Sulfonylurea Receptors
14.
Proc Natl Acad Sci U S A ; 96(15): 8579-84, 1999 Jul 20.
Article in English | MEDLINE | ID: mdl-10411918

ABSTRACT

Chronic antagonism of melanocortin receptors by the paracrine-acting agouti gene product induces both yellow fur and a maturity-onset obesity syndrome in mice that ubiquitously express wild-type agouti. Functional analysis of agouti mutations in transgenic mice indicate that the cysteine-rich C terminus, signal peptide, and glycosylation site are required for agouti activity in vivo. In contrast, no biological activity has been ascribed to the conserved basic domain. To examine the functional significance of the agouti basic domain, the entire 29-aa region was deleted from the agouti cDNA, and the resulting mutation (agoutiDeltabasic) was expressed in transgenic mice under the control of the beta-actin promoter (BAPaDeltabasic). Three independent lines of BAPaDeltabasic transgenic mice all developed some degree of yellow pigment in the fur, indicating that the agoutiDeltabasic protein was functional in vivo. However, none of the BAPaDeltabasic transgenic mice developed completely yellow fur, obesity, hyperinsulinemia, or hyperglycemia. High levels of agoutiDeltabasic expression in relevant tissues exceeded the level of agouti expression in obese viable yellow mice, suggesting that suboptimal activity or synthesis of the agoutiDeltabasic protein, rather than insufficient RNA synthesis, accounts for the phenotype of the BAPaDeltabasic transgenic mice. These findings implicate a functional role for the agouti basic domain in vivo, possibly influencing the biogenesis of secreted agouti protein or modulating protein-protein interactions that contribute to effective antagonism of melanocortin receptors.


Subject(s)
Intercellular Signaling Peptides and Proteins , Obesity/genetics , Pigmentation/genetics , Proteins/genetics , Agouti Signaling Protein , Animals , Body Weight , Gene Dosage , Gene Expression/genetics , Glycosylation , Mice , Mice, Transgenic , Mutation , Phenotype , Promoter Regions, Genetic , Protein Sorting Signals/genetics , RNA/analysis , Receptors, Corticotropin/antagonists & inhibitors , Receptors, Melanocortin , Sequence Deletion , Skin/metabolism
15.
Endocrinology ; 140(5): 2167-72, 1999 May.
Article in English | MEDLINE | ID: mdl-10218968

ABSTRACT

Desacetyl-alpha-MSH is more abundant than alpha-MSH in the brain, the fetus, human blood, and amniotic fluid, but there is little information on its ability to interact with melanocortin receptors. The aim of this study is to compare and contrast the ability of desacetyl-alpha-MSH and alpha-MSH to couple melanocortin receptors stably expressed in HEK293 cells, to the protein kinase A (PKA) signaling pathway. Desacetyl-alpha-MSH activated mouse MC1, MC3, MC4 and MC5 receptors with EC50s = 0.13, 0.96, 0.53, and 0.84 nM, and alpha-MSH activated these receptors with EC50s = 0.17, 0.88, 1.05, and 1.34 nM, respectively. Mouse agouti protein competitively antagonized alpha-MSH and desacetyl-alpha-MSH coupling to the MC1-R similarly. In contrast, mouse agouti protein antagonized desacetyl-alpha-MSH much more effectively and potently than alpha-MSH coupling the MC4-R to the PKA signaling pathway. Furthermore, mouse agouti protein (10 nM) significantly reduced (1.4-fold) the maximum response of mMC4-R to desacetyl-alpha-MSH and 100 nM mouse agouti significantly increased (4.8-fold) the EC50. Minimal antagonism of alpha-MSH coupling mMC4-R to the PKA signaling pathway was observed with 10 nM mouse agouti, whereas both 50 and 100 nM mouse agouti appeared to reduce the maximum reponse (1.1- and 1.3-fold, respectively) and increase the EC50 (2.5- and 3.4-fold respectively). Mouse agouti protein did not significantly antagonize either alpha-MSH or desacetyl-alpha-MSH coupling mouse MC3 and MC5 receptors. Understanding the similarities and differences in activation of melanocortin receptors by desacetyl-alpha-MSH and alpha-MSH will contribute to delineating the functional roles for these endogenous melanocortin peptides.


Subject(s)
Intercellular Signaling Peptides and Proteins , Peptide Fragments/pharmacology , Proteins/pharmacology , Receptors, Peptide/antagonists & inhibitors , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology , Adenylyl Cyclases/metabolism , Agouti Signaling Protein , Animals , Binding, Competitive , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression , Humans , Mice , Receptor, Melanocortin, Type 4 , Receptors, Corticotropin/physiology , Receptors, Melanocortin , Receptors, Peptide/genetics , Signal Transduction
16.
Physiol Genomics ; 1(1): 11-9, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-11015556

ABSTRACT

Ubiquitous expression of the mouse agouti gene results in obesity and hyperinsulinemia. Human agouti is expressed in adipose tissue, and we found recombinant agouti protein to stimulate lipogenesis in adipocytes in a Ca(2+)-dependent fashion. However, adipocyte-specific agouti transgenic mice only became obese in the presence of hyperinsulinemia. Because intracellular Ca(2+) concentration ([Ca(2+)](i)) is a primary signal for insulin release, and we have shown agouti protein to increase [Ca(2+)](i) in several cell types, we examined the effects of agouti on [Ca(2+)](i) and insulin release. We demonstrated the expression of agouti in human pancreas and generated recombinant agouti to study its effects on Ca(2+) signaling and insulin release. Agouti (100 nM) stimulated Ca(2+) influx, [Ca(2+)](i) increase, and a marked stimulation of insulin release in two beta-cell lines (RIN-5F and HIT-T15; P < 0. 05). Agouti exerted comparable effects in isolated human pancreatic islets and beta-cells, with a 5-fold increase in Ca(2+) influx (P < 0.001) and a 2.2-fold increase in insulin release (P < 0.01). These data suggest a potential role for agouti in the development of hyperinsulinemia in humans.


Subject(s)
Calcium Signaling/physiology , Fura-2/analogs & derivatives , Gene Expression , Hyperinsulinism/physiopathology , Insulin/metabolism , Intercellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Proteins/metabolism , Agouti Signaling Protein , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Cricetinae , Dose-Response Relationship, Drug , Glucose/metabolism , Glucose/pharmacology , Humans , Hyperinsulinism/genetics , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Manganese/pharmacology , Paracrine Communication/drug effects , Potassium Chloride/metabolism , Potassium Chloride/pharmacology , Proteins/genetics , Proteins/pharmacology , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
17.
IDrugs ; 2(8): 802-6, 1999 Aug.
Article in English | MEDLINE | ID: mdl-16127656

ABSTRACT

HP-228 is a cytokine regulating agent from Trega Biosciences (formerly Houghten Pharmaceuticals). It is in phase II trials for the management of post-surgical pain and inflammation [195065]. The drug was indicated for the treatment of diabetes, obesity and chemotherapy-induced toxicity, but Trega has decided to discontinue the development of HP-228 for these indications [259339].

18.
FASEB J ; 12(13): 1391-6, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9761782

ABSTRACT

Overexpression of the murine agouti gene results in obesity. The human homologue of agouti is expressed primarily in human adipocytes, and we have shown recombinant agouti protein to increase adipocyte intracellular Ca2+([Ca2+]i) and thereby stimulate lipogenesis. However, since recent data demonstrate that increasing adipocyte [Ca2+]i may also inhibit lipolysis, we have investigated the role of agouti-induced [Ca2+]i increases in regulating lipolysis in human adipocytes. Short-term (1 h) exposure to recombinant agouti (100 nM) protein had no effect on basal lipolysis, although longer term treatment (24 h) caused a 60% decrease in basal lipolysis (P<0.0001). Short-term agouti treatment totally inhibited ACTH-induced lipolysis (P<0.05). Since melanocortin receptors (MCR) are involved in some actions of agouti, we next determined whether agouti's antilipolytic effect is exerted through competitive antagonism of the ACTH receptor (MCR-2). Forskolin (1 microM), an adenylate cyclase activator, induced a 48% increase in lipolysis in human adipocytes (P<0.05); this effect was reversed by 100 nM agouti (P<005), demonstrating that the antilipolytic effect of agouti is distal to the ACTH receptor. To determine the role of [Ca2+]i in the antilipolytic effect of agouti, human adipocytes were treated with KCl or arginine vasopressin to stimulate voltage- and receptor-stimulated Ca2+ influx, respectively. Both agents caused inhibition of forskolin-induced lipolysis (P<0.005). Furthermore, agouti's antilipolytic effect was also blocked by the Ca2+ channel blocker nitrendipine. These data demonstrate that agouti exerts a potent antilipolytic effect in human adipocytes via a Ca2+-dependent mechanism. This effect, combined with agouti-induced lipogenesis, represents a coordinate control of adipocyte lipid metabolism that may contribute to an agouti-induced obesity syndrome.


Subject(s)
Adipose Tissue/drug effects , Calcium Signaling/drug effects , Calcium/physiology , Intercellular Signaling Peptides and Proteins , Lipolysis/drug effects , Proteins/pharmacology , Adenosine Triphosphate/physiology , Adipose Tissue/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/pharmacology , Agouti Signaling Protein , Animals , Arginine Vasopressin/pharmacology , Calcium Channels/drug effects , Calcium Channels/physiology , Cells, Cultured , Colforsin/pharmacology , Hair Color/genetics , Humans , Ion Transport , Mice , Obesity/genetics , Obesity/physiopathology , Potassium Chloride/pharmacology , Receptors, Corticotropin/drug effects , Receptors, Corticotropin/physiology , Receptors, Melanocortin , Recombinant Proteins/pharmacology , Second Messenger Systems
19.
Int J Obes Relat Metab Disord ; 22(7): 678-83, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9705029

ABSTRACT

OBJECTIVE: To test the hypothesis that a melanocortin agonist can reverse obesity and insulin resistance in mice overexpressing the agouti protein. EXPERIMENTAL MODEL: Mice overexpressing the agouti protein either by transgene introduction (beta-actin promotor) or by mutation (Ay). DESIGN: NDPMSH was tested for pharmacokinetic suitability. NDPMSH at various doses was administered subcutaneously twice a day for 2-3 weeks. MEASUREMENTS: Fur pigmentation, various fatness parameters (core temperature, fat pad weight and body weight), blood glucose and hormones, fatty acid synthase measurement. RESULTS: NDPMSH caused fur pigmentation and core temperature changes, but failed to affect any metabolic parameters in agouti-dependent manner. CONCLUSION: NDPMSH, as a representation melanocortin agonist, does not compete with agouti in reversing agouti-dependent metabolic effects. This suggests that 1) agouti works via a receptor other than a melanocortin receptor to mediate its metabolic effects, 2) agouti-dependent metabolic effects are mediated through melanocortin receptors but not via antagonism of these receptors, or 3) NDPMSH is pharmacodynamically an inappropriate molecule for these types of studies.


Subject(s)
Diabetes Mellitus/drug therapy , Intercellular Signaling Peptides and Proteins , Obesity , alpha-MSH/analogs & derivatives , alpha-MSH/agonists , Agouti Signaling Protein , Animals , Body Temperature/drug effects , Body Weight/drug effects , Hair Color/drug effects , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Phenotype , Proteins/genetics , Proteins/metabolism , Receptors, Pituitary Hormone/metabolism , Weight Gain/drug effects , alpha-MSH/pharmacokinetics , alpha-MSH/therapeutic use
20.
Biochemistry ; 37(4): 991-7, 1998 Jan 27.
Article in English | MEDLINE | ID: mdl-9454589

ABSTRACT

The agouti protein plays an important role in the development of diabetes and obesity in rodents and has been shown to be a potent antagonist of melanocortin receptors. For this reason alanine-scanning mutagenesis was performed on the agouti protein carboxyl terminus to locate residues important for melanocortin receptor binding inhibition. When agouti residues Arg116 and Phe118 are changed to alanine, very large decreases in agouti affinity for melanocortin receptor 1, 3, and 4 result. Mutation of Phe117 to alanine causes a similar increase in agouti KI app at melanocortin receptor 4. Substitution of agouti residue Asp108 with alanine results in large increases in KI app for all three melanocortin receptors examined. All of these residues are conserved in the agouti-related transcript, ART, whose expression is up-regulated in animal models of obesity. The three-dimensional structure of the agouti carboxyl terminus was modeled, and residues which decrease receptor binding by a factor of > or = 15 when mutated to alanine localize to one side of the structure. These agouti variants with altered receptor selectivity may be useful in determining the role of melanocortin receptors in diabetes and obesity.


Subject(s)
Intercellular Signaling Peptides and Proteins , Proteins/metabolism , Receptors, Corticotropin/antagonists & inhibitors , alpha-MSH/antagonists & inhibitors , Agouti Signaling Protein , Amino Acid Sequence , Animals , Binding Sites , Computer Simulation , DNA Mutational Analysis , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis , Paracrine Communication , Protein Binding , Proteins/genetics , Receptors, Melanocortin
SELECTION OF CITATIONS
SEARCH DETAIL
...