Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Am J Physiol Endocrinol Metab ; 306(10): E1176-87, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24714397

ABSTRACT

Glycerol-3-phosphate acyltransferases (GPATs) catalyze the first step in the synthesis of glycerolipids and glycerophospholipids. Microsomal GPAT, the major GPAT activity, is encoded by at least two closely related genes, GPAT3 and GPAT4. To investigate the in vivo functions of GPAT3, we generated Gpat3-deficient (Gpat3(-/-)) mice. Total GPAT activity in white adipose tissue of Gpat3(-/-) mice was reduced by 80%, suggesting that GPAT3 is the predominant GPAT in this tissue. In liver, GPAT3 deletion had no impact on total GPAT activity but resulted in a 30% reduction in N-ethylmaleimide-sensitive GPAT activity. The Gpat3(-/-) mice were viable and fertile and exhibited no obvious metabolic abnormalities on standard laboratory chow. However, when fed a high-fat diet, female Gpat3(-/-) mice showed decreased body weight gain and adiposity and increased energy expenditure. Increased energy expenditure was also observed in male Gpat3(-/-) mice, although it was not accompanied by a significant change in body weight. GPAT3 deficiency lowered fed, but not fasted, glucose levels and tended to improve glucose tolerance in diet-induced obese male and female mice. On a high-fat diet, Gpat3(-/-) mice had enlarged livers and displayed a dysregulation in cholesterol metabolism. These data establish GPAT3 as the primary GPAT in white adipose tissue and reveal an important role of the enzyme in regulating energy, glucose, and lipid homeostasis.


Subject(s)
Adipose Tissue, White/enzymology , Cholesterol/metabolism , Energy Metabolism/genetics , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Obesity/enzymology , Animals , Diet/adverse effects , Female , Glycerol-3-Phosphate O-Acyltransferase/genetics , Homeostasis/genetics , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics
2.
ACS Med Chem Lett ; 4(1): 118-23, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-24900572

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) catalyzes the conversion of inactive glucocorticoid cortisone to its active form, cortisol. The glucocorticoid receptor (GR) signaling pathway has been linked to the pathophysiology of diabetes and metabolic syndrome. Herein, the structure-activity relationship of a series of piperazine sulfonamide-based 11ß-HSD1 inhibitors is described. (R)-3,3,3-Trifluoro-2-(5-(((R)-4-(4-fluoro-2-(trifluoromethyl)phenyl)-2-methylpiperazin-1-yl)sulfonyl)thiophen-2-yl)-2-hydroxypropanamide 18a (HSD-621) was identified as a potent and selective 11ß-HSD1 inhibitor and was ultimately selected as a clinical development candidate. HSD-621 has an attractive overall pharmaceutical profile and demonstrates good oral bioavailability in mouse, rat, and dog. When orally dosed in C57/BL6 diet-induced obesity (DIO) mice, HSD-621 was efficacious and showed a significant reduction in both fed and fasting glucose and insulin levels. Furthermore, HSD-621 was well tolerated in drug safety assessment studies.

3.
Nat Biotechnol ; 29(11): 1028-33, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22020385

ABSTRACT

Effective therapies are needed to control excessive bleeding in a range of clinical conditions. We improve hemostasis in vivo using a conformationally pliant variant of coagulation factor Xa (FXa(I16L)) rendered partially inactive by a defect in the transition from zymogen to active protease. Using mouse models of hemophilia, we show that FXa(I16L) has a longer half-life than wild-type FXa and does not cause excessive activation of coagulation. Once clotting mechanisms are activated to produce its cofactor FVa, FXa(I16L) is driven to the protease state and restores hemostasis in hemophilic animals upon vascular injury. Moreover, using human or murine analogs, we show that FXa(I16L) is more efficacious than FVIIa, which is used to treat bleeding in hemophilia inhibitor patients. FXa(I16L) may provide an effective strategy to enhance blood clot formation and act as a rapid pan-hemostatic agent for the treatment of bleeding conditions.


Subject(s)
Enzyme Precursors/therapeutic use , Factor Xa/therapeutic use , Hemophilia A/drug therapy , Hemostatics/therapeutic use , Animals , Blood Coagulation/genetics , Disease Models, Animal , Enzyme Precursors/pharmacokinetics , Factor VIIa/genetics , Factor VIIa/metabolism , Factor Xa/pharmacokinetics , Gene Expression , HEK293 Cells , Hemorrhage/drug therapy , Hemostasis/genetics , Hemostatics/pharmacokinetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Thrombelastography , Thrombin/metabolism
4.
J Biol Chem ; 286(48): 41838-41851, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21990351

ABSTRACT

Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is one of two known DGAT enzymes that catalyze the final step in triglyceride synthesis. Findings from genetically modified mice as well as pharmacological studies suggest that inhibition of DGAT1 is a promising strategy for the treatment of obesity and type 2 diabetes. Here we characterize a tool DGAT1 inhibitor compound, T863. We found that T863 is a potent inhibitor for both human and mouse DGAT1 in vitro, which acts on the acyl-CoA binding site of DGAT1 and inhibits DGAT1-mediated triacylglycerol formation in cells. In an acute lipid challenge model, oral administration of T863 significantly delayed fat absorption and resulted in lipid accumulation in the distal small intestine of mice, mimicking the effects of genetic ablation of DGAT1. In diet-induced obese mice, oral administration of T863 for 2 weeks caused weight loss, reduction in serum and liver triglycerides, and improved insulin sensitivity. In addition to the expected triglyceride-lowering activity, T863 also lowered serum cholesterol. Hepatic IRS2 protein was dramatically up-regulated in mice treated with T863, possibly contributing to improved insulin sensitivity. In differentiated 3T3-L1 adipocytes, T863 enhanced insulin-stimulated glucose uptake, suggesting a possible role for adipocytes to improve insulin sensitivity upon DGAT1 inhibition. These results reveal novel mechanistic insights into the insulin-sensitizing effects of DGAT1 inhibition in mouse models. Taken together, our study provides a comprehensive evaluation of a small molecule inhibitor for DGAT1 and suggests that pharmacological inhibition of DGAT1 holds promise in treating diverse metabolic disorders.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Insulin Resistance , Liver/enzymology , Weight Loss/drug effects , 3T3-L1 Cells , Administration, Oral , Animals , Binding Sites , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Mice, Obese , Triglycerides/blood
5.
J Lipid Res ; 51(7): 1971-81, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20181984

ABSTRACT

Acyl-CoA:glycerol-3-phosphate acyltransferase (GPAT) catalyzes the first step during de novo synthesis of glycerolipids. Mammals have at least four GPAT isoforms. Here we report the further characterization of the two recently identified microsomal GPAT3 and GPAT4. Both enzymes are highly expressed in adipose tissues. However, while GPAT3 is highly (approximately 60-fold) induced during adipocyte differentiation, GPAT4 induction is only modest (approximately 5-fold), leading to a lower abundance of GPAT4 mRNA in adipocytes. While overexpression of GPAT3 and GPAT4 in either insect or mammalian cells results in a comparable increase of GPAT activity, shRNA-mediated knockdown of GPAT3, but not GPAT4, in 3T3-L1 adipocytes led to a significant decrease in GPAT activity, a profound inhibition of lipid accumulation, and a lack of expression of several adipogenic markers during adipocyte differentiation. These data suggest that GPAT3 may encode the major GPAT isoform in adipocytes and play an important role in adipogenesis. Furthermore, we have shown that both GPAT3 and GPAT4 are phosphorylated by insulin at Ser and Thr residues, leading to increased GPAT activity that is sensitive to wortmannin. Our results reveal a link between the lipogenic effects of insulin and microsomal GPAT3 and GPAT4, implying their importance in glycerolipid biosynthesis.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adipogenesis/physiology , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Insulin/metabolism , Isoenzymes/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/classification , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 3T3-L1 Cells , Amino Acid Sequence , Animals , Glycerol-3-Phosphate O-Acyltransferase/classification , Glycerol-3-Phosphate O-Acyltransferase/genetics , Hep G2 Cells , Humans , Isoenzymes/classification , Isoenzymes/genetics , Mice , Molecular Sequence Data , Phosphorylation , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Alignment , Tissue Distribution
6.
J Med Chem ; 52(17): 5449-61, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19673466

ABSTRACT

Cortisol and the glucocorticoid receptor signaling pathway have been implicated in the development of diabetes and obesity. The reduction of cortisone to cortisol is catalyzed by 11beta-hydroxysteroid dehydrogenase type I (11beta-HSD1). 2,4-Disubsituted benzenesulfonamides were identified as potent inhibitors of both the human and mouse enzymes. The lead compounds displayed good pharmacokinetics and ex vivo inhibition of the target in mice. Cocrystal structures of compounds 1 and 20 bound to human 11beta-HSD1 were obtained. Compound 20 was found to achieve high concentrations in target tissues, resulting in 95% inhibition in the ex vivo assay when dosed with a food mix (0.5 mg of drug per g of food) after 4 days. Compound 20 was efficacious in a mouse diet-induced obesity model and significantly reduced fed glucose and fasted insulin levels. Our findings suggest that 11beta-HSD1 inhibition may be a valid target for the treatment of diabetes.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Diet/adverse effects , Enzyme Inhibitors/pharmacology , Obesity/enzymology , Obesity/etiology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Disease Models, Animal , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Conformation , Obesity/drug therapy , Structure-Activity Relationship
7.
Proc Natl Acad Sci U S A ; 106(1): 262-7, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19116277

ABSTRACT

In a search for more effective anti-diabetic treatment, we used a process coupling low-affinity biochemical screening with high-throughput co-crystallography in the design of a series of compounds that selectively modulate the activities of all three peroxisome proliferator-activated receptors (PPARs), PPARalpha, PPARgamma, and PPARdelta. Transcriptional transactivation assays were used to select compounds from this chemical series with a bias toward partial agonism toward PPARgamma, to circumvent the clinically observed side effects of full PPARgamma agonists. Co-crystallographic characterization of the lead molecule, indeglitazar, in complex with each of the 3 PPARs revealed the structural basis for its PPAR pan-activity and its partial agonistic response toward PPARgamma. Compared with full PPARgamma-agonists, indeglitazar is less potent in promoting adipocyte differentiation and only partially effective in stimulating adiponectin gene expression. Evaluation of the compound in vivo confirmed the reduced adiponectin response in animal models of obesity and diabetes while revealing strong beneficial effects on glucose, triglycerides, cholesterol, body weight, and other metabolic parameters. Indeglitazar has now progressed to Phase II clinical evaluations for Type 2 diabetes mellitus (T2DM).


Subject(s)
Drug Discovery/methods , Hypoglycemic Agents/therapeutic use , PPAR gamma/agonists , Peroxisome Proliferator-Activated Receptors/agonists , Adipocytes/cytology , Adiponectin/genetics , Animals , Cell Differentiation/drug effects , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Humans , Hypoglycemic Agents/pharmacology , Mice , Obesity/drug therapy , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Rats , Transcriptional Activation/drug effects
8.
Arch Biochem Biophys ; 483(1): 45-54, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19121282

ABSTRACT

Tissue specific amplification of glucocorticoid action through NADPH-dependent reduction of inactive glucocorticoid precursors by 11beta-hydroxysteroid dehydrogenase (11beta-HSD1) contributes to the development of visceral obesity, insulin resistance and Type 2 Diabetes. Hexose-6-phosphate dehydrogenase (H6PDH) is believed to supply NADPH for the reductase activity of 11beta-HSD1 in the lumen of the endoplasmic reticulum (ER), where the two enzymes are co-localized. We report here expression and purification of full-length and truncated N-terminal domain (NTD) of H6PDH in a mammalian expression system. Interestingly, both full-length H6PDH and the truncated NTD are secreted into the culture medium in the absence of 11beta-HSD1. Purified full-length H6PDH is a bi-functional enzyme with glucose-6-phosphate dehydrogenase (G6PDH) activity as well as 6-phosphogluconolactonase (6PGL) activity. Using co-immunoprecipitation experiments with purified H6PDH and 11beta-HSD1, and with cell lysates expressing H6PDH and 11beta-HSD1, we observe direct physical interaction between the two enzymes. We also show the modulation of 11beta-HSD1 directionality by H6PDH using overexpression and siRNA knockdown systems. The NTD retains the ability to interact with 11beta-HSD1 physically as well as modulate 11beta-HSD1 directionality indicating that the NTD of H6PDH is sufficient for the regulation of the 11beta-HSD1 activity.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Carbohydrate Dehydrogenases/metabolism , Glucocorticoids/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Base Sequence , Carbohydrate Dehydrogenases/genetics , Catalysis , Cell Line , DNA Primers/genetics , Gluconates/metabolism , Humans , In Vitro Techniques , Kinetics , Mutagenesis, Site-Directed , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Transfection
9.
J Biol Chem ; 284(12): 8136-47, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19141617

ABSTRACT

Adipocyte-secreted proteins play important roles in metabolic regulation through autocrine, paracrine, and endocrine mechanisms. Using transcriptional profiling, we identified coiled-coil domain containing 80 (Ccdc80; also known as DRO1 and URB) as a novel secreted protein highly expressed in white adipose tissue. In 3T3-L1 cells Ccdc80 is expressed and secreted in a biphasic manner with high levels in postconfluent preadipocytes and terminally differentiated adipocytes. To determine whether Ccdc80 regulates adipocyte differentiation, Ccdc80 expression was manipulated using both knockdown and overexpression approaches. Small hairpin RNA-mediated silencing of Ccdc80 in 3T3-L1 cells inhibits adipocyte differentiation. This phenotype was partially reversed by treating the knockdown cells with Ccdc80-containing conditioned medium from differentiated 3T3-L1 cells. Molecular studies indicate that Ccdc80 is required for the full inhibition of T-cell factor-mediated transcriptional activity, down-regulation of Wnt/beta-catenin target genes during clonal expansion, and the subsequent induction of C/EBPalpha and peroxisome proliferator-activated receptor gamma. Surprisingly, overexpression of Ccdc80 in 3T3-L1 cells also inhibits adipocyte differentiation without affecting the repression of the Wnt/beta-catenin signaling pathway. Taken together, these data suggest that Ccdc80 plays dual roles in adipogenesis by mechanisms that involve at least in part down-regulation of Wnt/beta-catenin signaling and induction of C/EBPalpha and peroxisome proliferator-activated receptor gamma.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Adipose Tissue/metabolism , Glycoproteins/metabolism , Signal Transduction/physiology , 3T3-L1 Cells , Adipocytes/cytology , Adipose Tissue/cytology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/physiology , Extracellular Matrix Proteins , Gene Silencing , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
PPAR Res ; 2008: 125387, 2008.
Article in English | MEDLINE | ID: mdl-18989368

ABSTRACT

The prevalence of obesity in the USA and worldwide has reached epidemic proportions during the last two decades. Drugs currently available for the treatment of obesity provide no more than 5% placebo-adjusted weight loss and are associated with undesirable side effects. Peroxisome proliferator-activated receptor (PPAR) modulators offer potential benefits for the treatment of obesity and its associated complications but their development has been complicated by biological, technical, and regulatory challenges. Despite significant challenges, PPAR modulators are attractive targets for the treatment of obesity and could offer a viable alternative to the millions of patients who fail to lose weight following rigorous dieting and exercise protocols. In addition, PPAR modulators have the potential-added benefit of ameliorating the associated comorbidities.

12.
J Biol Chem ; 283(27): 19049-57, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18458083

ABSTRACT

Acyl-CoA-dependent lysophospholipid acyltransferases play an important role in attaining the appropriate molecular species of phospholipids. A number of genes encoding these activities were recently identified. It has become clear that multiple genes can encode one enzymatic activity and that a given gene may encode multiple activities. Here we report the identification of a gene encoding a mammalian acyl-CoA-dependent lysophospholipid acyltransferase with prominent activity toward ethanolamine-containing lysophospholipids, which we termed acyl-CoA:lysophosphatidylethanolamine acyltransferase 2, LPEAT2 (previously annotated as AYTL3 or AGPAT7). LPEAT2 is predominantly expressed in brain, coinciding with an enrichment of phosphatidylethanolamine in this tissue. Ectopic expression of LPEAT2 in mammalian HEK293T cells led to a dramatic increase (up to 9-fold) in LPEAT activity when compared with cells transfected with empty vector or an unrelated acyltransferase. LPEAT2 also exhibited significant acyl-CoA-dependent acyltransferase activity toward 1-O-alkenyl-lysophosphatidylethanolamine, lysophosphatidylglycerol, 1-O-alkyl-lysophosphatidylcholine, lysophosphatidylserine, and lysophosphatidylcholine but lacked appreciable acylating activity toward glycerol 3-phosphate, lysophosphatidic acid, lysophosphatidylinositol, and diacylglycerol, demonstrating multiple but selective functions of LPEAT2 as an enzyme involved in phospholipid remodeling. LPEAT2 recognizes a broad range of medium and long chain fatty acyl-CoA, and its activity was not affected by Ca(2+). When overexpressed in mammalian cells, LPEAT2 is localized to the endoplasmic reticulum. siRNA-mediated knockdown of LPEAT2 in HEK293T cells significantly decreased LPEAT and 1-alkenyl-LPEAT activities but did not affect other lysophospholipid acylating activities. These findings identify LPEAT2 as an important enzyme in the biosynthesis of ethanolamine-containing phospholipids, especially in brain.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Brain/enzymology , Endoplasmic Reticulum/enzymology , Lysophospholipids/biosynthesis , Nerve Tissue Proteins/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase , Cell Line , Endoplasmic Reticulum/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lysophospholipids/genetics , Nerve Tissue Proteins/genetics , Organ Specificity/physiology , Substrate Specificity/physiology
13.
Anal Biochem ; 365(2): 174-84, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17481567

ABSTRACT

Considerable effort exists within drug discovery to develop novel compounds to improve the underlying metabolic defects in type 2 diabetes. One approach is focused on inhibition of the tyrosine phosphatase, PTP1B, an important negative regulator of both insulin and leptin signaling. Historically, tyrosine phosphatase assays have used either small organic phosphates or, alternatively, phosphorylated peptides from the target proteins themselves. In characterizing inhibitors of PTP1B, measuring turnover of small organic phosphates is limited to evaluation of compounds that bind the active site itself. Peptide substrates allow identification of additional subsets of inhibitors (e.g., those that bind the second aryl-phosphate site), but assays of peptide turnover often involve detection steps that then limit full kinetic evaluation of inhibitors. Here we use a polyclonal antibody specific for the phosphorylated insulin receptor to allow much more sensitive detection of peptide phosphorylation. This kinetically robust enzyme-linked immunosorbent assay (ELISA) gives k(cat) and K(m) values for a phosphorylated insulin receptor peptide consistent with values determined by a continuous fluorescence-based assay. Furthermore, IC50 values determined for well-behaved active site inhibitors agree well with values determined for p-nitrophenyl phosphate cleavage. This assay permits full characterization of a larger subset of inhibitors as drug candidates for this promising target.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Protein Tyrosine Phosphatases/metabolism , Receptor, Insulin/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Molecular Structure , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/antagonists & inhibitors , Sensitivity and Specificity , Time Factors
14.
Endocrinology ; 148(2): 501-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17095592

ABSTRACT

It has been recently proposed that obestatin, a peptide encoded by the ghrelin gene, reduces food intake by activating the orphan G protein-coupled receptor GPR39. To gain further insights into the role of GPR39 in body weight homeostasis, we characterized the phenotype of mice with targeted disruption of the GPR39 gene. Body weight, adiposity, and food intake were found to be similar between GPR39(+/+) and GPR39(-/-) mice. Furthermore, fasting glucose and insulin levels were similar between both genotypes. Injection of obestatin peptide (1 micromol/kg, ip) obtained from multiple sources did not consistently inhibit food intake in wild-type mice after an overnight fast, and no difference in food intake was observed between wild-type and GPR39 knockout mice after injection of the peptide. Finally, ectopic expression of GPR39 in HEK293T cells revealed a constitutive activation of the receptor that was unaffected by stimulation with obestatin. Our phenotypic characterization suggests that GPR39 is not a major modulator of food intake in mice, although a more subtle role cannot be excluded. The role of GPR39 in normal physiology requires further study and should be conducted independently of the function of obestatin.


Subject(s)
Body Weight/physiology , Eating/physiology , Homeostasis/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Cell Line , Eating/drug effects , Ghrelin , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Hormones/genetics , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Phenotype , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/metabolism , Transfection
15.
Proc Natl Acad Sci U S A ; 103(52): 19695-700, 2006 Dec 26.
Article in English | MEDLINE | ID: mdl-17170135

ABSTRACT

Acyl-CoA:glycerol-3-phosphate acyltransferase (GPAT) catalyzes the first step during de novo synthesis of triacylglycerol. It has been well recognized that mammals possess multiple enzymatically distinct proteins with GPAT activity. Although the mitochondrial-associated GPAT has been cloned and extensively characterized, the molecular identity of the endoplasmic reticulum (ER)-associated GPAT, which accounts for the majority of total GPAT activity in most tissues, has remained elusive. Here we report the identification of genes encoding human and mouse ER-associated GPAT (termed GPAT3). GPAT3 is a member of the acyltransferase family predominantly expressed in tissues characterized by active lipid metabolism, such as adipose tissue, small intestine, kidney, and heart. Ectopic expression of GPAT3 leads to a significant increase in N-ethylmaleimide-sensitive GPAT activity, whereas acyltransferase activity toward a variety of other lysophospholipids, as well as neutral lipid substrates, is not altered. Overexpression of GPAT3 in mammalian cells results in increased triacylglycerol, but not phospholipid, formation. GPAT3 is localized to the ER when overexpressed in COS-7 cells. GPAT3 mRNA is dramatically up-regulated during adipocyte differentiation, is reciprocally regulated in adipose tissue and liver of ob/ob mice, and is up-regulated in mice treated with a peroxisome proliferator-activated receptor gamma (PPARgamma) agonist. A substantial loss of GPAT activity in 3T3-L1 adipocytes was achieved by reducing GPAT3 mRNA levels through GPAT3-specific siRNA knockdown. These findings identify GPAT3 as a previously uncharacterized triacylglycerol biosynthetic enzyme. Similar to other lipogenic enzymes, GPAT3 may be useful as a target for the treatment of obesity.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Glycerol-3-Phosphate O-Acyltransferase/chemistry , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Microsomes/enzymology , Triglycerides/biosynthesis , 1-Acylglycerol-3-Phosphate O-Acyltransferase/chemistry , Adipocytes/enzymology , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Computational Biology , Endoplasmic Reticulum/enzymology , Gene Expression Regulation, Enzymologic , Glycerol-3-Phosphate O-Acyltransferase/genetics , Humans , Male , Mice , Molecular Sequence Data , Obesity/enzymology , Obesity/genetics , Organ Specificity , PPAR gamma/metabolism , Phospholipids/biosynthesis , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid
16.
Trends Endocrinol Metab ; 17(7): 284-90, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16870465

ABSTRACT

Nuclear receptors represent novel targets for the development of therapeutic agents for the treatment of numerous diseases, including type 2 diabetes, obesity dyslipidemia, atherosclerosis and the metabolic syndrome. There have been many recent advances in the development of new therapeutic agents for a subset of these receptors, including the peroxisome proliferator-activated receptors, the liver X receptors and the farnesoid X receptor. To date, the synthesis of selective modulators that regulate the activity of these receptors has been empirical. However, a detailed understanding of the molecular basis for selective modulation, as well as new insights into the biology of these receptors, might open the door to the rational design of a new generation of therapeutic agents with improved safety and efficacy.


Subject(s)
Drug Delivery Systems , Metabolic Diseases/drug therapy , Receptors, Cytoplasmic and Nuclear/physiology , Animals , DNA-Binding Proteins/physiology , Humans , Liver X Receptors , Metabolic Diseases/physiopathology , Orphan Nuclear Receptors , Peroxisome Proliferator-Activated Receptors/physiology , RNA-Binding Proteins/physiology
17.
Vascul Pharmacol ; 45(3): 154-62, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16765099

ABSTRACT

OBJECTIVE AND DESIGN: Elevated blood pressure and insulin resistance are strongly associated in patients. We explored the potential for the anti-hypertensive angiotensin II type 1-receptor (ATR(1)) antagonists to improve insulin sensitivity through modulation of the nuclear receptor PPARgamma, in vitro and in vivo compared to the potent insulin sensitizer, rosiglitazone. METHODS: PPARgamma modulation by ATR(1) antagonists was measured first by direct recruitment of PGC-1, followed by trans-activation reporter assays in cells, and promotion of adipogenesis in fibroblast and pre-adipocyte cell lines. Improvement of insulin sensitivity was measured as changes in levels of glucose, insulin, and adiponectin in ob/ob mice. RESULTS: Telmisartan, candesartan, irbesartan, and losartan (but not valsartan or olmesartan) each served as bona fide PPARgamma ligands in vitro, with EC(50) values between 3 and 5 micro mol/l. However, only telmisartan, and to a lesser extent candesartan, resulted in significant PPARgamma agonism in cells. In vivo, although rosiglitazone significantly lowered both glucose (33%, p<0.01) and insulin (61%, p<0.01) levels and increased expression of adiponectin (74%, p<0.001), sartan treatment had no effect. CONCLUSIONS: Many members of the sartan family of ATR(1) antagonists are PPARgamma ligands in cell-free assays but their modulation of PPARgamma in cells is relatively weak. Furthermore, none appear to improve insulin sensitivity in a rodent model under conditions where other insulin sensitizers, including rosiglitazone, do. These results question whether reported effects of sartans on insulin sensitivity may be through other means, and should guide further efforts to develop dual agents to treat hypertension and insulin resistance.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Hypoglycemic Agents/pharmacology , Insulin Resistance , PPAR gamma/agonists , 3T3-L1 Cells , Adipogenesis/drug effects , Adiponectin/blood , Angiotensin II Type 1 Receptor Blockers/chemistry , Animals , Blood Glucose/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , Hypoglycemic Agents/chemistry , Insulin/blood , Male , Mice , Mice, Obese , Obesity/blood , Obesity/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Promoter Regions, Genetic/drug effects , Recombinant Proteins/agonists , Rosiglitazone , Structure-Activity Relationship , Thiazolidinediones/pharmacology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transfection
18.
Drug Discov Today ; 11(9-10): 405-11, 2006 May.
Article in English | MEDLINE | ID: mdl-16635802

ABSTRACT

Bone morphogenetic proteins (BMPs) and growth differentiation factors (GDFs) control the development and homeostasis of multiple tissue types in many organisms, from humans to invertebrates. These morphogens are expressed in a tissue-specific manner and they signal by binding to serine-threonine kinase receptors, resulting in coordinated changes in gene expression that regulate the differentiation and development of multiple tissue types. In addition, these proteins are regulated post-transcriptionally through binding to several soluble proteins. In this review we focus on a subset of BMPs and GDFs that have been implicated in the pathophysiology of type 2 diabetes and cardiovascular disease.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , Cardiovascular Diseases/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Metabolic Diseases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Atherosclerosis/metabolism , Bone Morphogenetic Protein 7 , Bone Morphogenetic Protein Receptors/drug effects , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Proteins/antagonists & inhibitors , Cardiovascular Agents/pharmacology , Cardiovascular Diseases/genetics , Diabetes Mellitus, Type 2/metabolism , Growth Differentiation Factor 3 , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypoglycemic Agents/pharmacology , Kidney Diseases/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors
19.
Bioorg Med Chem Lett ; 15(11): 2865-9, 2005 Jun 02.
Article in English | MEDLINE | ID: mdl-15911270

ABSTRACT

The design, synthesis, and biological evaluation of arylsulfonamidooxazoles as 11beta-HSD1 inhibitors and the serendipitous discovery of beta-keto sulfones as potent 11beta-HSD1 inhibitors are described here. These two classes of compounds are not active against 11beta-HSD2 and therefore may have significant therapeutic potential for metabolic syndrome, type 2 diabetes and related metabolic dysfunctions.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Oxazoles/pharmacology , Sulfones/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Oxazoles/chemical synthesis , Oxazoles/chemistry , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/chemistry
20.
Curr Opin Pharmacol ; 5(3): 328-32, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15907921

ABSTRACT

Myostatin is a secreted protein that negatively regulates skeletal muscle mass determining both muscle fiber number and size. The myostatin pathway is conserved and regulates muscle mass in a number of animal species ranging from fish to humans. Inhibition of myostatin using a variety of therapeutic approaches can increase muscle mass in a number of animal models of human disease, including muscular dystrophy.


Subject(s)
Muscular Diseases/therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Humans , Muscular Diseases/physiopathology , Myostatin , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta/adverse effects , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...