Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Pharmacol Exp Ther ; 368(3): 514-523, 2019 03.
Article in English | MEDLINE | ID: mdl-30606762

ABSTRACT

Thiazolidinediones (TZDs) are peroxisome proliferator-activated receptor γ (PPARγ) agonists that represent an effective class of insulin-sensitizing agents; however, clinical use is associated with weight gain and peripheral edema. To elucidate the role of PPARγ expression in endothelial cells (ECs) in these side effects, EC-targeted PPARγ knockout (Pparg ΔEC) mice were placed on a high-fat diet to promote PPARγ agonist-induced plasma volume expansion, and then treated with the TZD rosiglitazone. Compared with Pparg-floxed wild-type control (Pparg f/f) mice, Pparg ΔEC treated with rosiglitazone are resistant to an increase in extracellular fluid, water content in epididymal and inguinal white adipose tissue, and plasma volume expansion. Interestingly, histologic assessment confirmed significant rosiglitazone-mediated capillary dilation within white adipose tissue of Pparg f/f mice, but not Pparg ΔEC mice. Analysis of ECs isolated from untreated mice in both strains suggested the involvement of changes in endothelial junction formation. Specifically, compared with cells from Pparg f/f mice, Pparg ΔEC cells had a 15-fold increase in focal adhesion kinase, critically important in EC focal adhesions, and >3-fold significant increase in vascular endothelial cadherin, the main component of focal adhesions. Together, these results indicate that rosiglitazone has direct effects on the endothelium via PPARγ activation and point toward a critical role for PPARγ in ECs during rosiglitazone-mediated plasma volume expansion.


Subject(s)
Adipose Tissue/metabolism , Endothelial Cells/metabolism , Hypoglycemic Agents/pharmacology , PPAR gamma/deficiency , Rosiglitazone/pharmacology , Vascular Remodeling/physiology , Adipose Tissue/blood supply , Adipose Tissue/drug effects , Animals , Endothelial Cells/drug effects , Gene Deletion , Male , Mice , Mice, Transgenic , PPAR gamma/genetics , Plasma Volume/drug effects , Plasma Volume/physiology , Vascular Remodeling/drug effects
2.
Am J Physiol Endocrinol Metab ; 315(3): E386-E393, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29870677

ABSTRACT

Fructose consumption in humans and animals has been linked to enhanced de novo lipogenesis, dyslipidemia, and insulin resistance. Hereditary deficiency of ketohexokinase (KHK), the first enzymatic step in fructose metabolism, leads to essential fructosuria in humans, characterized by elevated levels of blood and urinary fructose following fructose ingestion but is otherwise clinically benign. To address whether KHK deficiency is associated with altered glucose and lipid metabolism, a Khk knockout (KO) mouse line was generated and characterized. NMR spectroscopic analysis of plasma following ingestion of [6-13C] fructose revealed striking differences in biomarkers of fructose metabolism. Significantly elevated urine and plasma 13C-fructose levels were observed in Khk KO vs. wild-type (WT) control mice, as was reduced conversion of 13C-fructose into plasma 13C-glucose and 13C-lactate. In addition, the observation of significant levels of fructose-6-phosphate in skeletal muscle tissue of Khk KO, but not WT, mice suggests a potential mechanism, whereby fructose is metabolized via muscle hexokinase in the absence of KHK. Khk KO mice on a standard chow diet displayed no metabolic abnormalities with respect to ambient glucose, glucose tolerance, body weight, food intake, and circulating trigylcerides, ß-hydroxybutyrate, and lactate. When placed on a high-fat and high-fructose (HF/HFruc) diet, Khk KO mice had markedly reduced liver weight, triglyceride levels, and insulin levels. Together, these results suggest that Khk KO mice may serve as a good model for essential fructosuria in humans and that inhibition of KHK offers the potential to protect from diet-induced hepatic steatosis and insulin resistance.


Subject(s)
Diet , Fructokinases/deficiency , Fructose Metabolism, Inborn Errors/genetics , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Animals , Blood Glucose/metabolism , Body Weight/genetics , Diet, High-Fat , Eating/genetics , Fructokinases/genetics , Fructokinases/metabolism , Fructose Metabolism, Inborn Errors/metabolism , Fructosephosphates/blood , Glucose Intolerance/genetics , Insulin Resistance , Lipid Metabolism/genetics , Mice, Knockout
3.
Radiology ; 287(2): 581-589, 2018 05.
Article in English | MEDLINE | ID: mdl-29156148

ABSTRACT

Purpose To evaluate the biodistribution, metabolism, and pharmacokinetics of a new type I collagen-targeted magnetic resonance (MR) probe, CM-101, and to assess its ability to help quantify liver fibrosis in animal models. Materials and Methods Biodistribution, pharmacokinetics, and stability of CM-101 in rats were measured with mass spectrometry. Bile duct-ligated (BDL) and sham-treated rats were imaged 19 days after the procedure by using a 1.5-T clinical MR imaging unit. Mice were treated with carbon tetrachloride (CCl4) or with vehicle two times a week for 10 weeks and were imaged with a 7.0-T preclinical MR imaging unit at baseline and 1 week after the last CCl4 treatment. Animals were imaged before and after injection of 10 µmol/kg CM-101. Change in contrast-to-noise ratio (ΔCNR) between liver and muscle tissue after CM-101 injection was used to quantify liver fibrosis. Liver tissue was analyzed for Sirius Red staining and hydroxyproline content. The institutional subcommittee for research animal care approved all in vivo procedures. Results CM-101 demonstrated rapid blood clearance (half-life = 6.8 minutes ± 2.4) and predominately renal elimination in rats. Biodistribution showed low tissue gadolinium levels at 24 hours (<3.9% injected dose [ID]/g ± 0.6) and 10-fold lower levels at 14 days (<0.33% ID/g ± 12) after CM-101 injection with negligible accumulation in bone (0.07% ID/g ± 0.02 and 0.010% ID/g ± 0.004 at 1 and 14 days, respectively). ΔCNR was significantly (P < .001) higher in BDL rats (13.6 ± 3.2) than in sham-treated rats (5.7 ± 4.2) and in the CCl4-treated mice (18.3 ± 6.5) compared with baseline values (5.2 ± 1.0). Conclusion CM-101 demonstrated fast blood clearance and whole-body elimination, negligible accumulation of gadolinium in bone or tissue, and robust detection of fibrosis in rat BDL and mouse CCl4 models of liver fibrosis. © RSNA, 2017 Online supplemental material is available for this article.


Subject(s)
Fibrosis/pathology , Gadolinium/pharmacokinetics , Liver Cirrhosis/diagnostic imaging , Liver/pathology , Magnetic Resonance Imaging , Polysaccharides, Bacterial/pharmacokinetics , Animals , Carbon Tetrachloride/pharmacokinetics , Disease Models, Animal , Fibrosis/diagnostic imaging , Half-Life , Liver/diagnostic imaging , Mass Spectrometry , Mice , Rats , Tissue Distribution
4.
Bioorg Med Chem Lett ; 26(23): 5724-5728, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27815121

ABSTRACT

The transformation of an aryloxybutanoic acid ultra high-throughput screening (uHTS) hit into a potent and selective series of G-protein coupled receptor 120 (GPR120) agonists is reported. uHTS hit 1 demonstrated an excellent rodent pharmacokinetic profile and selectivity over the related fatty acid receptor GPR40, but only modest GPR120 potency. Optimization of the "left-hand" aryl group led to compound 6, which demonstrated a GPR120 mechanism-based pharmacodynamic effect in a mouse oral glucose tolerance test (oGTT). Further optimization gave rise to the benzofuran propanoic acid series (exemplified by compound 37), which demonstrated acute mechanism-based pharmacodynamic effects. The combination of in vivo efficacy and attractive rodent pharmacodynamic profiles suggests compounds generated from this series may afford attractive candidates for the treatment of Type 2 diabetes.


Subject(s)
Benzofurans/chemistry , Benzofurans/pharmacology , Propionates/chemistry , Propionates/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Benzofurans/blood , Blood Glucose/analysis , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Hypoglycemic Agents/blood , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Mice , Propionates/blood , Receptors, G-Protein-Coupled/metabolism
5.
Nat Med ; 13(6): 695-702, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17529981

ABSTRACT

The metabolism of vitamin A and the diverse effects of its metabolites are tightly controlled by distinct retinoid-generating enzymes, retinoid-binding proteins and retinoid-activated nuclear receptors. Retinoic acid regulates differentiation and metabolism by activating the retinoic acid receptor and retinoid X receptor (RXR), indirectly influencing RXR heterodimeric partners. Retinoic acid is formed solely from retinaldehyde (Rald), which in turn is derived from vitamin A. Rald currently has no defined biologic role outside the eye. Here we show that Rald is present in rodent fat, binds retinol-binding proteins (CRBP1, RBP4), inhibits adipogenesis and suppresses peroxisome proliferator-activated receptor-gamma and RXR responses. In vivo, mice lacking the Rald-catabolizing enzyme retinaldehyde dehydrogenase 1 (Raldh1) resisted diet-induced obesity and insulin resistance and showed increased energy dissipation. In ob/ob mice, administrating Rald or a Raldh inhibitor reduced fat and increased insulin sensitivity. These results identify Rald as a distinct transcriptional regulator of the metabolic responses to a high-fat diet.


Subject(s)
Adipogenesis/physiology , Diet/adverse effects , Growth Inhibitors/physiology , Obesity/metabolism , Obesity/prevention & control , Retinaldehyde/physiology , 3T3-L1 Cells , Adipogenesis/genetics , Animals , Female , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , NIH 3T3 Cells , Obesity/physiopathology , Rabbits , Retinaldehyde/biosynthesis , Retinaldehyde/genetics
6.
J Lipid Res ; 54(10): 2615-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23828778

ABSTRACT

Hepatic glucose overproduction is a major characteristic of type 2 diabetes. Because glucagon is a key regulator for glucose homeostasis, antagonizing the glucagon receptor (GCGR) is a possible therapeutic strategy for the treatment of diabetes mellitus. To study the effect of hepatic GCGR inhibition on the regulation of lipid metabolism, we generated siRNA-mediated GCGR knockdown (si-GCGR) in the db/db mouse. The hepatic knockdown of GCGR markedly reduced plasma glucose levels; however, total plasma cholesterol was increased. The detailed lipid analysis showed an increase in the LDL fraction, and no change in VLDL HDL fractions. Further studies showed that the increase in LDL was the result of over-expression of hepatic lipogenic genes and elevated de novo lipid synthesis. Inhibition of hepatic glucagon signaling via siRNA-mediated GCGR knockdown had an effect on both glucose and lipid metabolism in db/db mice.


Subject(s)
Diabetes Mellitus, Type 2/blood , Lipogenesis , Liver/metabolism , Receptors, Glucagon/genetics , Animals , Blood Glucose , Cholesterol/blood , Diabetes Mellitus, Type 2/therapy , Gene Expression , Gene Knockdown Techniques , Lipoproteins, LDL/blood , Male , Mice , Mice, Obese , RNA Interference , RNA, Small Interfering/genetics , Receptors, Glucagon/metabolism , Triglycerides/blood , Triglycerides/metabolism
7.
J Lipid Res ; 53(1): 51-65, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22021650

ABSTRACT

In an attempt to understand the applicability of various animal models to dyslipidemia in humans and to identify improved preclinical models for target discovery and validation for dyslipidemia, we measured comprehensive plasma lipid profiles in 24 models. These included five mouse strains, six other nonprimate species, and four nonhuman primate (NHP) species, and both healthy animals and animals with metabolic disorders. Dyslipidemic humans were assessed by the same measures. Plasma lipoprotein profiles, eight major plasma lipid fractions, and FA compositions within these lipid fractions were compared both qualitatively and quantitatively across the species. Given the importance of statins in decreasing plasma low-density lipoprotein cholesterol for treatment of dyslipidemia in humans, the responses of these measures to simvastatin treatment were also assessed for each species and compared with dyslipidemic humans. NHPs, followed by dog, were the models that demonstrated closest overall match to dyslipidemic humans. For the subset of the dyslipidemic population with high plasma triglyceride levels, the data also pointed to hamster and db/db mouse as representative models for practical use in target validation. Most traditional models, including rabbit, Zucker diabetic fatty rat, and the majority of mouse models, did not demonstrate overall similarity to dyslipidemic humans in this study.


Subject(s)
Disease Models, Animal , Dyslipidemias/blood , Lipids/blood , Animals , Cricetinae , Dogs , Dyslipidemias/drug therapy , Fatty Acids/blood , Humans , Mice , Primates , Simvastatin/therapeutic use , Triglycerides/blood
9.
Bioorg Med Chem Lett ; 20(20): 6088-92, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20832306
10.
Bioorg Med Chem Lett ; 20(3): 1298-301, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20064714

ABSTRACT

Systematic structure-activity relationship (SAR) studies of a screening lead led to the discovery of a series of thiazolidinediones (TZDs) as potent GPR40 agonists. Among them, compound C demonstrated an acute mechanism-based glucose-lowering in an intraperitoneal glucose tolerance test (IPGTT) in lean mice, while no effects were observed in GPR40 knock-out mice.


Subject(s)
Drug Discovery/methods , Receptors, G-Protein-Coupled/agonists , Thiazolidinediones/chemistry , Animals , Mice , Mice, Knockout , Protein Binding/physiology , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship , Thiazolidinediones/agonists , Thiazolidinediones/pharmacology
11.
Biochem J ; 418(2): 413-20, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19035854

ABSTRACT

GLUT4 (glucose transporter 4) plays important roles in glucose homoeostasis in vivo. GLUT4 expression and function are diminished in diabetic human and animal subjects. The goal of the present study is to develop a cell-based assay for identifying negative regulators of GLUT4 translocation as potential targets for the treatment of Type 2 diabetes. Traditional GLUT4 translocation assays performed in differentiated myocytes or adipocytes are difficult to perform, particularly in HTS (high-throughput screening) mode. In the present study, we stably co-expressed c-Myc and eGFP [enhanced GFP (green fluorescent protein)] dual-tagged recombinant GLUT4 with recombinant IRS1 (insulin receptor substrate 1) in HEK-293 cells (human embryonic kidney cells) (HEK-293.IRS1.GLUT4 cells). Insulin treatment stimulated both glucose uptake and GLUT4 translocation in these cells. GLUT4 translocation is quantified by a TRF (time-resolved fluorescence) assay in a 96-well HTS format. TRF assays confirmed insulin-stimulated GLUT4 translocation, which can be inhibited by PI3K (phosphoinositide 3-kinase) or Akt [also called PKB (protein kinase B)] inhibitors. Treatment with palmitate increased IRS1 serine phosphorylation and reduced insulin-stimulated Akt phosphorylation and GLUT4 translocation, indicating insulin resistance. Knockdown of PTEN (phosphatase and tensin homologue deleted on chromosome 10) and PTP1B (protein tyrosine phosphatase 1B) gene expression by siRNA (small interfering RNA) treatment significantly increased GLUT4 translocation only in cells treated with palmitate but not in untreated cells. Similar results were obtained on treatment with siRNA of JNK1 (c-Jun N-terminal kinase 1), S6K1 (ribosomal protein S6 kinase, 70 kDa, polypeptide 1) and PKC(theta) (protein kinase C theta). In summary, we have established and validated a novel GLUT4 translocation assay that is optimal for identifying negative regulators of GLUT4 translocation. In combination with more physiologically relevant secondary assays in myotubes and adipocytes, this assay system can be used to identify potential novel therapeutic targets for the treatment of Type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Evaluation, Preclinical/methods , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Cells, Cultured , Gene Knockdown Techniques , Glucose/metabolism , Glucose Transporter Type 4/genetics , Humans , Hypoglycemic Agents/isolation & purification , Insulin/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Isoenzymes/genetics , Mitogen-Activated Protein Kinase 8/genetics , PTEN Phosphohydrolase/genetics , Palmitic Acid/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/genetics , Protein Kinase C-theta , Protein Transport , Ribosomal Protein S6 Kinases/genetics , Transfection
12.
Cell Rep Med ; 1(4): 100056, 2020 07 21.
Article in English | MEDLINE | ID: mdl-33205063

ABSTRACT

Fibrosis, or the accumulation of extracellular matrix, is a common feature of many chronic diseases. To interrogate core molecular pathways underlying fibrosis, we cross-examine human primary cells from various tissues treated with TGF-ß, as well as kidney and liver fibrosis models. Transcriptome analyses reveal that genes involved in fatty acid oxidation are significantly perturbed. Furthermore, mitochondrial dysfunction and acylcarnitine accumulation are found in fibrotic tissues. Substantial downregulation of the PGC1α gene is evident in both in vitro and in vivo fibrosis models, suggesting a common node of metabolic signature for tissue fibrosis. In order to identify suppressors of fibrosis, we carry out a compound library phenotypic screen and identify AMPK and PPAR as highly enriched targets. We further show that pharmacological treatment of MK-8722 (AMPK activator) and MK-4074 (ACC inhibitor) reduce fibrosis in vivo. Altogether, our work demonstrate that metabolic defect is integral to TGF-ß signaling and fibrosis.


Subject(s)
Fibrosis/genetics , Fibrosis/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Adenylate Kinase/metabolism , Animals , Benzimidazoles/pharmacology , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression/genetics , Gene Expression Profiling/methods , Humans , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Specificity/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Transcriptome/genetics , Transforming Growth Factor beta/metabolism
13.
Biochemistry ; 48(2): 492-8, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19105608

ABSTRACT

Previous studies demonstrated that the naturally occurring electrophile and PPARgamma ligand, nitrolinoleic acid (NO(2)-LA), exists as a mixture of four regioisomers [Alexander, R. L., et al. (2006) Biochemistry 45, 7889-7896]. We hypothesized that these alternative isomers have distinct bioactivities; therefore, to determine if the regioisomers are quantitatively or qualitatively different with respect to PPARgamma activation, NO(2)-LA was separated into three fractions which were identified by NMR (13-NO(2)-LA, 12-NO(2)-LA, and a mixture of 9- and 10-NO(2)-LA) and characterized for PPARgamma interactions. A competition radioligand binding assay showed that all three NO(2)-LA fractions had similar binding affinities for PPARgamma (IC(50) = 0.41-0.60 microM) that were comparable to that of the pharmaceutical ligand, rosiglitazone (IC(50) = 0.25 microM). However, when PPARgamma-dependent transcription activation was examined, there were significant differences observed among the NO(2)-LA fractions. Each isomer behaved as a partial agonist in this reporter gene assay; however, the 12-NO(2) derivative was the most potent with respect to maximum activation of PPARgamma and an EC(50) of 0.045 microM (compare with the rosiglitazone EC(50) of 0.067 microM), while the 13-NO(2) and 9- and 10-NO(2) derivatives were considerably less effective with EC(50) values of 0.41-0.62 microM. We conclude that the regioisomers of NO(2)-LA are not functionally equivalent. The 12-NO(2) derivative appears to be the most potent in PPARgamma-dependent transcription activation, whereas the weaker PPARgamma agonists, 13-NO(2) and 9- and 10-NO(2), may be relatively more important in signaling via other, PPARgamma-independent pathways in which this family of nitrolipid electrophiles is implicated.


Subject(s)
Linoleic Acids/pharmacology , Nitro Compounds/pharmacology , PPAR gamma/metabolism , Binding, Competitive , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Clone Cells , Dose-Response Relationship, Drug , Female , Genes, Reporter , Humans , Inhibitory Concentration 50 , Linoleic Acids/chemistry , Luciferases/metabolism , Nitro Compounds/chemistry , Nuclear Magnetic Resonance, Biomolecular , PPAR gamma/agonists , PPAR gamma/genetics , Radioligand Assay , Rosiglitazone , Stereoisomerism , Thiazolidinediones/metabolism , Transcriptional Activation/drug effects , Transduction, Genetic
14.
Mol Pharmacol ; 73(1): 62-74, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17940191

ABSTRACT

Despite their proven antidiabetic efficacy, widespread use of peroxisome proliferator-activated receptor (PPAR)gamma agonists has been limited by adverse cardiovascular effects. To overcome this shortcoming, selective PPARgamma modulators (SPPARgammaMs) have been identified that have antidiabetic efficacy comparable with full agonists with improved tolerability in preclinical species. The results of structural studies support the proposition that SPPARgammaMs interact with PPARgamma differently from full agonists, thereby providing a physical basis for their novel activities. Herein, we describe a novel PPARgamma ligand, SPPARgammaM2. This compound was a partial agonist in a cell-based transcriptional activity assay, with diminished adipogenic activity and an attenuated gene signature in cultured human adipocytes. X-ray cocrystallography studies demonstrated that, unlike rosiglitazone, SPPARgammaM2 did not interact with the Tyr473 residue located within helix 12 of the ligand binding domain (LBD). Instead, SPPARgammaM2 was found to bind to and activate human PPARgamma in which the Tyr473 residue had been mutated to alanine (hPPARgammaY473A), with potencies similar to those observed with the wild-type receptor (hPPARgammaWT). In additional studies, we found that the intrinsic binding and functional potencies of structurally distinct SPPARgammaMs were not diminished by the Y473A mutation, whereas those of various thiazolidinedione (TZD) and non-TZD PPARgamma full agonists were reduced in a correlative manner. These results directly demonstrate the important role of Tyr473 in mediating the interaction of full agonists but not SPPARgammaMs with the PPARgamma LBD, thereby providing a precise molecular determinant for their differing pharmacologies.


Subject(s)
PPAR gamma/metabolism , Tyrosine/metabolism , Humans , Ligands
15.
Bioorg Med Chem Lett ; 18(17): 4798-801, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18701276

ABSTRACT

A series of highly functionalized 3-aroyl and 3-phenoxy-2-methyl-7-azaindoles have been identified, which are potent selective PPARgamma modulators (SPPARgammaMs). Addition of substituents at the 6-position of the 7-azaindoles improves in vitro potency and pharmacokinetics. 7-Azaindoles have significantly improved off-target profiles compared to the parent indole series.


Subject(s)
Indoles/chemistry , Indoles/pharmacology , PPAR gamma/metabolism , Animals , Humans , Mice , PPAR gamma/agonists , Rats , Rats, Sprague-Dawley
16.
ACS Med Chem Lett ; 9(11): 1082-1087, 2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30429949

ABSTRACT

We report new SSTR5 antagonists with enhanced potency, subtype selectivity, and minimal off-target activities as compared to previously reported compounds. Starting from the reported SSTR5 antagonist 1, we systematically surveyed changes in the central core and head piece while maintaining the diphenyl tail group constant. From this study the azaspirodecanone 10 emerged as a new highly potent and selective SSTR5 antagonist. Compound 10 lowered glucose excursion by 94% in an oral glucose tolerance test (OGTT) in mice following a 3 mg/kg oral dose. The compound increased both total and active circulating incretin hormone GLP-1 levels in mice at a dose of 10 mg/kg. A synergistic effect was also demonstrated when compound 10 was coadministered with a DPP-4 inhibitor, substantially increasing circulating active GLP-1[7-36] amide and insulin in response to a glucose challenge.

17.
Biochim Biophys Acta ; 1761(12): 1401-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17110163

ABSTRACT

Farnesoid X receptor (FXR), a bile-acid-activated member of the nuclear receptor superfamily, is essential in regulating bile-acid, cholesterol, and triglyceride homeostasis. Disruption of the FXR gene in mice results in a proatherosclerotic lipid profile with increased serum cholesterols and triglycerides. However, the role of FXR in foam-cell formation and atherosclerosis development remains unclear. The current study showed that the peritoneal macrophages isolated from FXR-null mice took up less oxidized LDL-cholesterol (oxLDL-C), which was accompanied by a marked reduction in CD36 expression in these cells. This result appears to be FXR-independent, as FXR was not detected in the peritoneal macrophages. To assess to what extent FXR modulates atherosclerosis development, FXR/ApoE double-null mice were generated. Female mice were used for atherosclerosis analysis. Compared to ApoE-null mice, the FXR/ApoE double-null mice were found to have less atherosclerotic lesion area in the aorta, despite a further increase in the serum cholesterols and triglycerides. Our results indicate that disruption of the FXR gene could attenuate atherosclerosis development, most likely resulting from reduced oxLDL-C uptake by macrophages. Our study cautions the use of serum lipid levels as a surrogate marker to determine the efficiency of FXR modulators in treating hyperlipidemia.


Subject(s)
Atherosclerosis/etiology , DNA-Binding Proteins/metabolism , Foam Cells/metabolism , Foam Cells/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biological Transport, Active , Cholesterol, LDL/metabolism , Cytokines/biosynthesis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Gene Expression , Homeostasis , In Vitro Techniques , Lipids/blood , Lipoproteins, LDL/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
18.
Mol Cell Biol ; 22(8): 2607-19, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11909955

ABSTRACT

Disruption of the peroxisome proliferator-activated receptor gamma (PPAR gamma) gene causes embryonic lethality due to placental dysfunction. To circumvent this, a PPAR gamma conditional gene knockout mouse was produced by using the Cre-loxP system. The targeted allele, containing loxP sites flanking exon 2 of the PPAR gamma gene, was crossed into a transgenic mouse line expressing Cre recombinase under the control of the alpha/beta interferon-inducible (MX) promoter. Induction of the MX promoter by pIpC resulted in nearly complete deletion of the targeted exon, a corresponding loss of full-length PPAR gamma mRNA transcript and protein, and marked reductions in basal and troglitazone-stimulated expression of the genes encoding lipoprotein lipase, CD36, LXR alpha, and ABCG1 in thioglycolate-elicited peritoneal macrophages. Reductions in the basal levels of apolipoprotein E (apoE) mRNA in macrophages and apoE protein in total plasma and high-density lipoprotein (HDL) were also observed in pIpC-treated PPAR gamma-MXCre(+) mice. Basal cholesterol efflux from cholesterol-loaded macrophages to HDL was significantly reduced after disruption of the PPAR gamma gene. Troglitazone selectively inhibited ABCA1 expression (while rosiglitazone, ciglitazone, and pioglitazone had little effect) and cholesterol efflux in both PPAR gamma-deficient and control macrophages, indicating that this drug can exert paradoxical effects on cholesterol homeostasis that are independent of PPAR gamma. Together, these data indicate that PPAR gamma plays a critical role in the regulation of cholesterol homeostasis by controlling the expression of a network of genes that mediate cholesterol efflux from cells and its transport in plasma.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Apolipoproteins E/genetics , Cholesterol/metabolism , Macrophages/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Alleles , Animals , Biological Transport, Active , Exons , Female , Gene Expression/drug effects , Gene Targeting , Integrases/genetics , Lipoprotein Lipase/genetics , Macrophages/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Poly I-C/pharmacology , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Deletion , Transcription Factors/metabolism , Viral Proteins/genetics
19.
ACS Med Chem Lett ; 8(1): 96-101, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28105282

ABSTRACT

GPR120 (FFAR4) is a fatty acid sensing G protein coupled receptor (GPCR) that has been identified as a target for possible treatment of type 2 diabetes. A selective activator of GPR120 containing a chromane scaffold has been designed, synthesized, and evaluated in vivo. Results of these efforts suggest that chromane propionic acid 18 is a suitable tool molecule for further animal studies. Compound 18 is selective over the closely related target GPR40 (FFAR1), has a clean off-target profile, demonstrates suitable pharmacokinetic properties, and has been evaluated in wild-type/knockout GPR120 mouse oGTT studies.

20.
ACS Med Chem Lett ; 8(1): 49-54, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28105274

ABSTRACT

Type 2 diabetes mellitus (T2DM) is an ever increasing worldwide epidemic, and the identification of safe and effective insulin sensitizers, absent of weight gain, has been a long-standing goal of diabetes research. G-protein coupled receptor 120 (GPR120) has recently emerged as a potential therapeutic target for treating T2DM. Natural occurring, and more recently, synthetic agonists have been associated with insulin sensitizing, anti-inflammatory, and fat metabolism effects. Herein we describe the design, synthesis, and evaluation of a novel spirocyclic GPR120 agonist series, which culminated in the discovery of potent and selective agonist 14. Furthermore, compound 14 was evaluated in vivo and demonstrated acute glucose lowering in an oral glucose tolerance test (oGTT), as well as improvements in homeostatic measurement assessment of insulin resistance (HOMA-IR; a surrogate marker for insulin sensitization) and an increase in glucose infusion rate (GIR) during a hyperinsulinemic euglycemic clamp in diet-induced obese (DIO) mice.

SELECTION OF CITATIONS
SEARCH DETAIL