401
|
Labinskyy V, Bellomo M, Chandler MP, Young ME, Lionetti V, Qanud K, Bigazzi F, Sampietro T, Stanley WC, Recchia FA. Chronic activation of peroxisome proliferator-activated receptor-alpha with fenofibrate prevents alterations in cardiac metabolic phenotype without changing the onset of decompensation in pacing-induced heart failure. J Pharmacol Exp Ther 2007; 321:165-71. [PMID: 17215446 DOI: 10.1124/jpet.106.116871] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Severe heart failure (HF) is characterized by profound alterations in cardiac metabolic phenotype, with down-regulation of the free fatty acid (FFA) oxidative pathway and marked increase in glucose oxidation. We tested whether fenofibrate, a pharmacological agonist of peroxisome proliferator-activated receptor-alpha, the nuclear receptor that activates the expression of enzymes involved in FFA oxidation, can prevent metabolic alterations and modify the progression of HF. We administered 6.5 mg/kg/day p.o. fenofibrate to eight chronically instrumented dogs over the entire period of high-frequency left ventricular pacing (HF + Feno). Eight additional HF dogs were not treated, and eight normal dogs were used as a control. [3H]Oleate and [14C]Glucose were infused intravenously to measure the rate of substrate oxidation. At 21 days of pacing, left ventricular end-diastolic pressure was significantly lower in HF + Feno (14.1 +/- 1.6 mm Hg) compared with HF (18.7 +/- 1.3 mm Hg), but it increased up to 25 +/- 2 mm Hg, indicating end-stage failure, in both groups after 29 +/- 2 days of pacing. FFA oxidation was reduced by 40%, and glucose oxidation was increased by 150% in HF compared with control, changes that were prevented by fenofibrate. Consistently, the activity of myocardial medium chain acyl-CoA dehydrogenase, a marker enzyme of the FFA beta-oxidation pathway, was reduced in HF versus control (1.46 +/- 0.25 versus 2.42 +/- 0.24 micromol/min/gram wet weight (gww); p < 0.05) but not in HF + Feno (1.85 +/- 0.18 micromol/min/gww; N.S. versus control). Thus, preventing changes in myocardial substrate metabolism in the failing heart causes a modest improvement of cardiac function during the progression of the disease, with no effects on the onset of decompensation.
Collapse
|
402
|
Grey A, Bolland M, Gamble G, Wattie D, Horne A, Davidson J, Reid IR. The peroxisome proliferator-activated receptor-gamma agonist rosiglitazone decreases bone formation and bone mineral density in healthy postmenopausal women: a randomized, controlled trial. J Clin Endocrinol Metab 2007; 92:1305-10. [PMID: 17264176 DOI: 10.1210/jc.2006-2646] [Citation(s) in RCA: 307] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CONTEXT Thiazolidinediones, which are peroxisome proliferator-activated receptor-gamma agonists, are widely prescribed to patients with disorders characterized by insulin resistance. Preclinical studies suggest that peroxisome proliferator-activated receptor-gamma signaling negatively regulates bone formation and bone density. Human data on the skeletal effects of thiazolidinediones are currently available only from observational studies. OBJECTIVE The objective of the study was to determine whether rosiglitazone, a thiazolidinedione, inhibits bone formation. DESIGN The study was a 14-wk randomized, double-blind, placebo-controlled trial. SETTING The study was conducted in the general community. PATIENTS Fifty healthy, postmenopausal women participated in the study. INTERVENTION Intervention was rosiglitazone 8 mg/d. MAIN OUTCOME MEASURES The primary end point was biochemical markers of bone formation, and secondary end points were a bone resorption marker and bone mineral density. RESULTS The osteoblast markers procollagen type I N-terminal propeptide and osteocalcin declined by 13% (P<0.005 vs. placebo) and 10% (P=0.04 vs. placebo), respectively, in the rosiglitazone group. These changes were evident by 4 wk and persisted for the duration of the study. There was no change in the serum beta-C-terminal telopeptide of type I collagen, a marker of bone resorption (P=0.9 vs. placebo). Total hip bone density fell in the rosiglitazone group (mean change from baseline rosiglitazone -1.9%, placebo -0.2%; between-group difference 1.7%, 95% confidence interval 0.6-2.7, P<0.01); lumbar spine bone density fell significantly from baseline values in the rosiglitazone group (P=0.02 vs. baseline) but was not significantly different between groups (mean change from baseline rosiglitazone -1.2%, placebo -0.2%; between-group difference 1.0%, 95% confidence interval -0.2-2.3, P=0.13). CONCLUSIONS Short-term therapy with rosiglitazone exerts detrimental skeletal effects by inhibiting bone formation. Skeletal end points should be included in future long-term studies of thiazolidinedione use.
Collapse
Affiliation(s)
- Andrew Grey
- Department of Medicine, University of Auckland, and LabPlus, Auckland City Hospital, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
403
|
Abstract
OBJECTIVE This study was undertaken to quantify the expression of muscle GLUT in type 2 diabetes and to determine if treatment with an insulin-enhancing thiazolidenedione drug, pioglitazone, would alter its expression. RESEARCH DESIGN AND METHODS Twelve patients with type 2 diabetes were randomly assigned to treatment with either pioglitazone or placebo in a double-blinded 8-week protocol. Protein and mRNA for GLUT4 and GLUT5 were quantified in muscle homogenates from biopsies of vastus lateralis before and after treatment. The five additional GLUT family isoforms expressed in muscle had mRNA quantified in these samples. RESULTS Baseline and posttreatment repeat measurements of GLUT4 protein were not different from control measurements. Compared with normal subjects, GLUT5 protein increased 2.5-fold, and GLUT5 mRNA was 82% higher in the pretreatment samples from the diabetic subjects. Concentrations of mRNA for the six other GLUTs (GLUT1, GLUT3, GLUT4, GLUT8, GLUT11, and GLUT12) were not different from control subjects before or after treatment. The proportion of type I (red) fibers (46%) in diabetic muscle was not affected by pioglitazone treatment. Pioglitazone treatment decreased muscle GLUT5 mRNA and protein by 52 and 40%, respectively, whereas placebo did not alter GLUT5 expression. Both red and white fibers had higher GLUT5 expression in the baseline diabetic muscle samples, and a pioglitazone-related decrease in GLUT5 protein also occurred in both. CONCLUSIONS GLUT5 was dramatically increased in diabetic muscle, and pioglitazone treatment reversed this overexpression. The role of this fructose transporter expression in the insulin-enhancing effect of pioglitazone in muscle is unclear.
Collapse
Affiliation(s)
- Charles A Stuart
- Department of Internal Medicine, East Tennessee State University, James H. Quillen College of Medicine, Johnson City, Tennessee 37614-0622, USA.
| | | | | |
Collapse
|
404
|
Abstract
There is a progressive deterioration in beta-cell function and mass in type 2 diabetics. It was found that islet function was about 50% of normal at the time of diagnosis, and a reduction in beta-cell mass of about 60% was shown at necropsy. The reduction of beta-cell mass is attributable to accelerated apoptosis. The major factors for progressive loss of beta-cell function and mass are glucotoxicity, lipotoxicity, proinflammatory cytokines, leptin, and islet cell amyloid. Impaired beta-cell function and possibly beta-cell mass appear to be reversible, particularly at early stages of the disease where the limiting threshold for reversibility of decreased beta-cell mass has probably not been passed. Among the interventions to preserve or "rejuvenate" beta-cells, short-term intensive insulin therapy of newly diagnosed type 2 diabetes will improve beta-cell function, usually leading to a temporary remission time. Another intervention is the induction of beta-cell "rest" by selective activation of ATP-sensitive K+ (K(ATP)) channels, using drugs such as diazoxide. A third type of intervention is the use of antiapoptotic drugs, such as the thiazolidinediones (TZDs), and incretin mimetics and enhancers, which have demonstrated significant clinical evidence of effects on human beta-cell function. The TZDs improve insulin secretory capacity, decrease beta-cell apoptosis, and reduce islet cell amyloid with maintenance of neogenesis. The TZDs have indirect effects on beta-cells by being insulin sensitizers. The direct effects are via peroxisome proliferator-activated receptor gamma activation in pancreatic islets, with TZDs consistently improving basal beta-cell function. These beneficial effects are sustained in some individuals with time. There are several trials on prevention of diabetes with TZDs. Incretin hormones, which are released from the gastrointestinal tract in response to nutrient ingestion to enhance glucose-dependent insulin secretion from the pancreas, aid the overall maintenance of glucose homeostasis through slowing of gastric emptying, inhibition of glucagon secretion, and control of body weight. From the two major incretins, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), only the first one or its mimetics or enhancers can be used for treatment because the diabetic beta-cell is resistant to GIP action. Because of the rapid inactivation of GLP-1 by dipeptidyl peptidase (DPP)-IV, several incretin analogs were developed: GLP-1 receptor agonists (incretin mimetics) exenatide (synthetic exendin-4) and liraglutide, by conjugation of GLP-1 to circulating albumin. The acute effect of GLP-1 and GLP-1 receptor agonists on beta-cells is stimulation of glucose-dependent insulin release, followed by enhancement of insulin biosynthesis and stimulation of insulin gene transcription. The chronic action is stimulating beta-cell proliferation, induction of islet neogenesis, and inhibition of beta-cell apoptosis, thus promoting expansion of beta-cell mass, as observed in rodent diabetes and in cultured beta-cells. Exenatide and liraglutide enhanced postprandial beta-cell function. The inhibition of the activity of the DPP-IV enzyme enhances endogenous GLP-1 action in vivo, mediated not only by GLP-1 but also by other mediators. In preclinical studies, oral active DPP-IV inhibitors (sitagliptin and vildagliptin) also promoted beta-cell proliferation, neogenesis, and inhibition of apoptosis in rodents. Meal tolerance tests showed improvement in postprandial beta-cell function. Obviously, it is difficult to estimate the protective effects of incretin mimetics and enhancers on beta-cells in humans, and there is no clinical evidence that these drugs really have protective effects on beta-cells.
Collapse
Affiliation(s)
- Bernardo L Wajchenberg
- Endocrine Service and Diabetes and Heart Center of The Heart Institute, Hospital das Clinicas of The University of São Paulo Medical School, São Paulo, SP 05403-000, Brazil.
| |
Collapse
|
405
|
Stoddart LA, Brown AJ, Milligan G. Uncovering the pharmacology of the G protein-coupled receptor GPR40: high apparent constitutive activity in guanosine 5'-O-(3-[35S]thio)triphosphate binding studies reflects binding of an endogenous agonist. Mol Pharmacol 2007; 71:994-1005. [PMID: 17200419 DOI: 10.1124/mol.106.031534] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In cells lacking expression of Ca(2+)-mobilizing G proteins, coexpression of human GPR40 and Galpha(q) allowed medium- and long-chain fatty acids to elevate intracellular [Ca(2+)]. This was also observed when human embryonic kidney (HEK) 293 cells were transfected with a GPR40-Galpha(q) fusion protein. The kinetic of elevation of intracellular [Ca(2+)] slowed with increasing fatty acid chain length, suggesting different ligand on-rates, whereas the addition of fatty acid-free bovine serum albumin reduced signals, presumably by binding the fatty acids. To allow effective ligand equilibration, GPR40-Galpha(q) was used in guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding assays. After expression of GPR40-Galpha(q) in HEK293 cells and membrane preparation basal binding of [(35)S]GTPgammaSinGalpha(q) immunoprecipitates was high and not elevated substantially by fatty acids. However, treatment of membranes with fatty acid-free bovine serum albumin reduced the basal [(35)S]GTPgammaS binding in a concentration-dependent manner and allowed the responsiveness and pharmacology at GPR40 of each of the fatty acids thiazolidinediones and a novel small-molecule agonist to be uncovered. Membranes of rat INS-1E cells that express GPR40 endogenously provided similar observations. The high apparent constitutive activity of GPR40-Galpha(q) was also reversed by a small-molecule GPR40 antagonist, and basal [(35)S]GTPgammaS binding was prevented by the selective Galpha(q)/Galpha(11) inhibitor YM-254890. The current studies provide novel insights into the pharmacology of GPR40 and indicate that G protein-coupled receptors which respond to fatty acids, and potentially to other lipid ligands, can be occupied by endogenous agonists before assay and that this may mask the pharmacology of the receptor and may be mistaken for high levels of constitutive activity.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Davidson Building University of Glasgow, Glasgow G12 8QQ, Scotland, UK.
| | | | | |
Collapse
|
406
|
Paoletti R, Bolego C, Poli A, Cignarella A. Metabolic syndrome, inflammation and atherosclerosis. Vasc Health Risk Manag 2007; 2:145-52. [PMID: 17319458 PMCID: PMC1993992 DOI: 10.2147/vhrm.2006.2.2.145] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The inflammatory component of atherogenesis has been increasingly recognized over the last decade. Inflammation participates in all stages of atherosclerosis, not only during initiation and during evolution of lesions, but also with precipitation of acute thrombotic complications. The metabolic syndrome is associated with increased risk for development of both cardiovascular disease and type-2 diabetes in humans. Central obesity and insulin resistance are thought to represent common underlying factors of the syndrome, which features a chronic low-grade inflammatory state. Diagnosis of the metabolic syndrome occurs using defined threshold values for waist circumference, blood pressure, fasting glucose and dyslipidemia. The metabolic syndrome appears to affect a significant proportion of the population. Therapeutic approaches that reduce the levels of proinflammatory biomarkers and address traditional risk factors are particularly important in preventing cardiovascular disease and, potentially, diabetes. The primary management of metabolic syndrome involves healthy lifestyle promotion through moderate calorie restriction, moderate increase in physical activity and change in dietary composition. Treatment of individual components aims to control atherogenic dyslipidemia using fibrates and statins, elevated blood pressure, and hyperglycemia. While no single treatment for the metabolic syndrome as a whole yet exists, emerging therapies offer potential as future therapeutic approaches.
Collapse
Affiliation(s)
- Rodolfo Paoletti
- Department of Pharmacological Sciences, University of Milan, Italy.
| | | | | | | |
Collapse
|
407
|
. PB, . MR, . MS. Effect of Fenofibrate in Pressure Overload-induced Experimental Cardiac Hypertrophy. ACTA ACUST UNITED AC 2007. [DOI: 10.3923/ijbc.2007.104.110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
408
|
Di Paola R, Cuzzocrea S. Peroxisome proliferator-activated receptors ligands and ischemia-reperfusion injury. Naunyn Schmiedebergs Arch Pharmacol 2007; 375:157-75. [PMID: 17394034 DOI: 10.1007/s00210-007-0141-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Accepted: 01/28/2007] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to a subfamily of transcription nuclear factors. Three isoforms of PPARs have been identified: alpha, beta/delta and gamma, encoded by different genes and distributed in various tissues. They play important roles in metabolic processes like regulation of glucose and lipid redistribution. They also have anti-atherogenic, anti-inflammatory as well as antihypertensive functions. There is good evidence that ligands of PPARs reduce tissue injury associated with ischemia and reperfusion. The potential utility of PPAR ligands in ischemia and reperfusion will be discussed in this review.
Collapse
Affiliation(s)
- Rosanna Di Paola
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Torre Biologica-Policlinico Universitario, Via C. Valeria-Gazzi, 98100 Messina, Italy
| | | |
Collapse
|
409
|
Abstract
Type 2 diabetes mellitus is a complex disease combining defects in insulin secretion and insulin action. New compounds called thiazolidinediones or glitazones have been developed for reducing insulin resistance. After the withdrawal of troglitazone because of liver toxicity, two compounds are currently used in clinical practice, rosiglitazone and pioglitazone. These compounds are generally used in combination with other pharmacological agents. Because they are metabolised via cytochrome P450 (CYP), glitazones are exposed to numerous pharmacokinetic interactions. CYP2C8 and CYP3A4 are the main isoenzymes catalysing biotransformation of pioglitazone (as with troglitazone), whereas rosiglitazone is metabolised by CYP2C9 and CYP2C8. For both rosiglitazone and pioglitazone, the most relevant interactions have been described in healthy volunteers with rifampicin (rifampin), which results in a significant decrease of area under the plasma concentration-time curve [AUC] (54-65% for rosiglitazone, p<0.001; 54% for pioglitazone, p<0.001), and with gemfibrozil, which results in a significant increase of AUC (130% for rosiglitazone, p<0.001; 220-240% for pioglitazone, p<0.001). The relevance of such drug-drug interactions in patients with type 2 diabetes remains to be evaluated. However, in the absence of clinical data, it is prudent to reduce the dosage of each glitazone by half in patients treated with gemfibrozil. Conversely, rosiglitazone and pioglitazone do not seem to significantly affect the pharmacokinetics of other compounds. Although some food components have also been shown to potentially interfere with drugs metabolised with the CYP system, no published study deals specifically with these possible CYP-mediated food-drug interactions with glitazones.
Collapse
Affiliation(s)
- André J Scheen
- Division of Diabetes, Nutrition and Metabolic Disorders and Division of Clinical Pharmacology, Department of Medicine, CHU Sart Tilman, University of Liège, Liège, Belgium.
| |
Collapse
|
410
|
Goldberg RB, Mendez AJ. Severe acquired (secondary) high-density lipoprotein deficiency. J Clin Lipidol 2007; 1:41-56. [DOI: 10.1016/j.jacl.2007.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Accepted: 02/06/2007] [Indexed: 10/20/2022]
|
411
|
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors involved in the transcriptional regulation of key metabolic pathways such as lipid metabolism, adipogenesis, and insulin sensitivity. More recent work implicates all 3 PPAR isotypes (alpha, gamma, and delta, also known as beta or beta/delta) in inflammatory and atherosclerotic pathways. Because these nuclear receptors are activated by extracellular signals and control multiple gene targets, PPARs can be seen as nodes that control multiple inputs and outputs involved in energy balance, providing insight into how metabolism and the vasculature may be integrated. The ongoing clinical use of fibrates, which activate PPARalpha, and thiazolidinediones, which activate PPARgamma, establishes these receptors as viable drug targets, whereas considerable in vitro animal model and human surrogate marker studies suggest that PPAR activation may limit inflammation and atherosclerosis. Together, these various observations have stimulated intense interest in PPARs as therapeutic targets and led to large-scale cardiovascular end-point trials with PPAR agonists. The first of these studies has generated mixed results that require careful review, especially in anticipation of additional clinical trial data and ongoing attempts to develop novel PPAR modulators. Such analysis of the existing PPAR data, the appropriate use of currently approved PPAR agonists, and continued progress in PPAR therapeutics will be predicated on a better understanding of PPAR biology.
Collapse
Affiliation(s)
- Jonathan D Brown
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Mass, USA
| | | |
Collapse
|
412
|
Abstract
Diabetes mellitus and heart failure (HF), both of which are associated with high rates of adverse cardiovascular outcomes, commonly coexist. Given the marked increases in diabetes prevalence in developed countries, the proportion of the population with both conditions is likely to increase substantially. This article reviews the epidemiology of HF and diabetes, the mechanisms whereby diabetes causes HF, and the pharmacotherapy of both HF and diabetes. Specific challenges in treating patients with both HF and diabetes are also addressed.
Collapse
Affiliation(s)
- Frederick A Masoudi
- Division of Cardiology, Department of Medicine, Denver Health Medical Center, Denver, Colorado 80204, USA.
| | | |
Collapse
|
413
|
Best L, Elliott AC, Brown PD. Curcumin induces electrical activity in rat pancreatic beta-cells by activating the volume-regulated anion channel. Biochem Pharmacol 2007; 73:1768-75. [PMID: 17382910 DOI: 10.1016/j.bcp.2007.02.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 02/09/2007] [Accepted: 02/12/2007] [Indexed: 11/21/2022]
Abstract
Curcumin, the principal active component of turmeric, is reported to exert a number of therapeutic actions, including a hypoglycaemic/antidiabetic action. The underlying mechanisms to this action are essentially unknown. We have investigated the hypothesis that a direct stimulatory action on the pancreatic beta-cell could contribute towards the hypoglycaemic activity of this compound. Electrical and ion channel activity were recorded in rat beta-cells using the patch-clamp technique. beta-Cell volume was measured using a video-imaging technique. Insulin release was measured from intact islets by radioimmunoassay. Curcumin (2-10 microM) activated the volume-regulated anion channel in beta-cells. Single channel studies indicated that activation was the result of increased channel open probability. This effect was accompanied by depolarisation of the cell membrane potential, the generation of electrical activity and enhanced insulin release. Curcumin also decreased beta-cell volume, presumably reflecting loss of Cl(-) (and hence water) as a result of anion channel activation. These findings are consistent with the suggestion that Cl(-) fluxes play an important role in regulating beta-cell function. The stimulation of beta-cell function by curcumin could contribute to the hypoglycaemic actions of this compound, and these findings identify a novel potential therapeutic target for the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Leonard Best
- Department of Medicine, University of Manchester, Oxford Road, Manchester M13 9WL, UK.
| | | | | |
Collapse
|
414
|
Ghisletti S, Huang W, Ogawa S, Pascual G, Lin ME, Willson TM, Rosenfeld MG, Glass CK. Parallel SUMOylation-dependent pathways mediate gene- and signal-specific transrepression by LXRs and PPARgamma. Mol Cell 2007; 25:57-70. [PMID: 17218271 PMCID: PMC1850387 DOI: 10.1016/j.molcel.2006.11.022] [Citation(s) in RCA: 431] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 10/13/2006] [Accepted: 11/21/2006] [Indexed: 01/15/2023]
Abstract
Transrepression is widely utilized to negatively regulate gene expression, but the mechanisms by which different nuclear receptors effect gene- and signal-specific transrepression programs remain poorly understood. Here, we report the identification of alternative SUMOylation-dependent mechanisms that enable PPARgamma and LXRs to negatively regulate overlapping but distinct subsets of proinflammatory genes. Ligand-dependent conjugation of SUMO2/3 to LXRs or SUMO1 to PPARgamma targets them to promoters of TLR target genes, where they prevent the signal-dependent removal of NCoR corepressor complexes required for transcriptional activation. SUMO1-PPARgamma and SUMO2/3-LXRs inhibit distinct NCoR clearance mechanisms, allowing promoter- and TLR-specific patterns of repression. Mutational analysis and studies of naturally occurring oxysterol ligands indicate that the transactivation and SUMOylation-dependent transrepression activities of LXRs can be independently regulated. These studies define parallel but functionally distinct pathways that are utilized by PPARgamma and LXRs to differentially regulate complex programs of gene expression that control immunity and homeostasis.
Collapse
Affiliation(s)
- Serena Ghisletti
- Department of Cellular and Molecular Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
| | - Wendy Huang
- Department of Cellular and Molecular Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
- Biomedical Sciences Graduate Program, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
| | - Sumito Ogawa
- Department of Cellular and Molecular Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
| | - Gabriel Pascual
- Department of Cellular and Molecular Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
| | - Mu-En Lin
- Biomedical Sciences Graduate Program, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
| | - Timothy M. Willson
- GlaxoSmithKline, 5 Moore Drive, PO Box 13398 Research Triangle Park, NC 27709
| | - Michael G. Rosenfeld
- Department of Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
- Howard Hughes Medical Institute
| | - Christopher K. Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
- Department of Medicine, University of California, San Diego 9500 Gilman Drive, La Jolla CA 92093
- Correspondence: Office: 858 534 6011 Fax: 858 822 2127
| |
Collapse
|
415
|
Ghisletti S, Huang W, Ogawa S, Pascual G, Lin ME, Willson TM, Rosenfeld MG, Glass CK. Parallel SUMOylation-dependent pathways mediate gene- and signal-specific transrepression by LXRs and PPARgamma. Mol Cell 2007. [PMID: 17218271 DOI: 10.1016/jmolcel.2006.11.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Transrepression is widely utilized to negatively regulate gene expression, but the mechanisms by which different nuclear receptors effect gene- and signal-specific transrepression programs remain poorly understood. Here, we report the identification of alternative SUMOylation-dependent mechanisms that enable PPARgamma and LXRs to negatively regulate overlapping but distinct subsets of proinflammatory genes. Ligand-dependent conjugation of SUMO2/3 to LXRs or SUMO1 to PPARgamma targets them to promoters of TLR target genes, where they prevent the signal-dependent removal of NCoR corepressor complexes required for transcriptional activation. SUMO1-PPARgamma and SUMO2/3-LXRs inhibit distinct NCoR clearance mechanisms, allowing promoter- and TLR-specific patterns of repression. Mutational analysis and studies of naturally occurring oxysterol ligands indicate that the transactivation and SUMOylation-dependent transrepression activities of LXRs can be independently regulated. These studies define parallel but functionally distinct pathways that are utilized by PPARgamma and LXRs to differentially regulate complex programs of gene expression that control immunity and homeostasis.
Collapse
Affiliation(s)
- Serena Ghisletti
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
416
|
Lanne B, Dahllöf B, Lindahl C, Ebefors K, Kanmert I, von Bahr H, Miliotis T, Nyström AC, Arnerup G, Paulsons I, Kerb S, Oakes N. PPARalpha and PPARgamma regulation of liver and adipose proteins in obese and dyslipidemic rodents. J Proteome Res 2007; 5:1850-9. [PMID: 16889406 DOI: 10.1021/pr060004o] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Zucker fatty rats and ob/ob mice are both frequently used hyperlipidemic and insulin-resistant spontaneous genetic models of obesity. We used them to study the effect of PPAR agonists on the protein-expression level in liver and white adipose tissue. PPARalpha-agonist treatments of the rats resulted in that 27% of the quantified hepatic proteins were altered; implicating pronounced peroxisome proliferation and increase in capacity for beta-oxidation of fatty acids although no correction of plasma triglycerides were obtained. On treatment with PPARgamma agonists, adipose proteins were regulated to a much larger extent in the rats compared to mice, 18% and 2%, respectively.
Collapse
Affiliation(s)
- Boel Lanne
- AstraZeneca R&D Mölndal, S-431 83 Mölndal, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
417
|
Cantin LD, Liang S, Ogutu H, Iwuagwu CI, Boakye K, Bullock WH, Burns M, Clark R, Claus T, delaCruz FE, Daly M, Ehrgott FJ, Johnson JS, Keiper C, Livingston JN, Schoenleber RW, Shapiro J, Town C, Yang L, Tsutsumi M, Ma X. Indanylacetic acid derivatives carrying aryl-pyridyl and aryl-pyrimidinyl tail groups—new classes of PPAR γ/δ and PPAR α/γ/δ agonists. Bioorg Med Chem Lett 2007; 17:1056-61. [PMID: 17157013 DOI: 10.1016/j.bmcl.2006.11.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 11/07/2006] [Accepted: 11/08/2006] [Indexed: 10/23/2022]
Abstract
Modulation of PPAR activities represents an attractive approach for the treatment of diabetes with associated cardiovascular complications. The indanylacetic acid structural motif has proven useful in the generation of potent and tunable PPAR ligands. Modification of the substituents on the linker and the heterocycle tail group allowed for the modulation of the selectivity at the different receptor subtypes. Compound 33 was evaluated in vivo, where it displayed the desired reduction of glucose levels and increase in HDL levels in various animal models.
Collapse
Affiliation(s)
- Louis-David Cantin
- Department of Chemistry Research, Bayer Pharmaceuticals Corporation, West Haven, CT 06516, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
418
|
Scarsi M, Podvinec M, Roth A, Hug H, Kersten S, Albrecht H, Schwede T, Meyer UA, Rücker C. Sulfonylureas and glinides exhibit peroxisome proliferator-activated receptor gamma activity: a combined virtual screening and biological assay approach. Mol Pharmacol 2007; 71:398-406. [PMID: 17082235 DOI: 10.1124/mol.106.024596] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Most drugs currently employed in the treatment of type 2 diabetes either target the sulfonylurea receptor stimulating insulin release (sulfonylureas, glinides), or target the peroxisome proliferator-activated receptor (PPARgamma) improving insulin resistance (thiazolidinediones). Our work shows that sulfonylureas and glinides additionally bind to PPARgamma and exhibit PPARgamma agonistic activity. This activity was predicted in silico by virtual screening and confirmed in vitro in a binding assay, a transactivation assay, and by measuring the expression of PPARgamma target genes. Among the measured compounds, gliquidone and glipizide (two sulfonylureas), as well as nateglinide (a glinide), exhibit PPARgamma agonistic activity at concentrations comparable with those reached under pharmacological treatment. The most active of these compounds, gliquidone, is shown to be as potent as pioglitazone at inducing PPARgamma target gene expression. This dual mode of action of sulfonylureas and glinides may open new perspectives for the molecular pharmacology of antidiabetic drugs, because it provides evidence that drugs can be designed that target both the sulfonylurea receptor and PPARgamma. Targeting both receptors could increase pancreatic insulin secretion and improve insulin resistance. Glinides, sulfonylureas, and other acidified sulfonamides may be promising leads in the development of new PPARgamma agonists. In addition, we provide a unified concept of the PPARgamma binding ability of seemingly disparate compound classes.
Collapse
Affiliation(s)
- Marco Scarsi
- Biozentrum, University of Basel, Klingelbergstr. 50-70, CH-4056 Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
419
|
Gervois P, Fruchart JC, Staels B. Drug Insight: mechanisms of action and therapeutic applications for agonists of peroxisome proliferator-activated receptors. ACTA ACUST UNITED AC 2007; 3:145-56. [PMID: 17237841 DOI: 10.1038/ncpendmet0397] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Accepted: 09/08/2006] [Indexed: 12/19/2022]
Abstract
Intensive preclinical investigations have delineated a role for peroxisome proliferator-activated receptors (PPARs) in energy metabolism and inflammation. PPARs are activated by natural lipophilic ligands such as fatty acids and their derivatives. Normalization of lipid and glucose metabolism is achieved via pharmacological modulation of PPAR activity. PPARs may also alter atherosclerosis progression through direct effects on the vascular wall. PPARs regulate genes involved in the recruitment of leukocytes to endothelial cells, in vascular inflammation, in macrophage lipid homeostasis, and in thrombosis. PPARs therefore modulate metabolic and inflammatory perturbations that predispose to cardiovascular diseases and type 2 diabetes. The hypolipidemic fibrates and the antidiabetic thiazolidinediones are drugs that act via PPARalpha and PPARgamma, respectively, and are used in clinical practice. PPARbeta/delta ligands are currently in clinical evaluation. The pleiotropic actions of PPARs and the fact that chemically diverse PPAR agonists may induce distinct pharmacological responses have led to the emergence of new concepts for drug design. A more precise understanding of the molecular pathways implicated in the response to chemically distinct PPAR agonists should provide new opportunities for targeted therapeutic applications in the management of the metabolic syndrome, type 2 diabetes, and cardiovascular diseases.
Collapse
Affiliation(s)
- Philippe Gervois
- INSERM U 545, Institut Pasteur de Lille, Université de Lille 2, Lille, France.
| | | | | |
Collapse
|
420
|
Chintharlapalli S, Papineni S, Safe S. 1,1-bis(3'-indolyl)-1-(p-substitutedphenyl)methanes inhibit growth, induce apoptosis, and decrease the androgen receptor in LNCaP prostate cancer cells through peroxisome proliferator-activated receptor gamma-independent pathways. Mol Pharmacol 2007; 71:558-69. [PMID: 17093136 DOI: 10.1124/mol.106.028696] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
1,1-bis(3'-indolyl)-1-(p-substitutedphenyl)methanes (C-DIMs) containing para-trifluoromethyl, t-butyl, and phenyl groups are a novel class of peroxisome proliferator-activated receptor (PPAR)gamma agonists. In LNCaP prostate cancer cells, these compounds induce PPARgamma-dependent transactivation, inhibit cell proliferation, and induce apoptosis. In addition, these PPARgamma agonists modulate a number of antiproliferative and proapoptotic responses, including induction of p27, activating transcription factor 3, and nonsteroidal anti-inflammatory drug-activated gene-1 and down-regulation of cyclin D1 and caveolin-1. Moreover, the PPARgamma antagonist 2-chloro-5-nitrobenzanilide (GW9662) does not inhibit these effects. The C-DIM compounds also abrogate androgen receptor (AR)-mediated signaling and decrease prostate-specific antigen (PSA) and AR protein expression, and these responses were PPARgamma-independent. The effects of C-DIMs on AR and PSA were due to decreased AR and PSA mRNA expression in LNCaP cells. Thus, this series of methylene-substituted diindolylmethane derivatives simultaneously activate multiple pathways in LNCaP cells, including ablation of androgen-responsiveness and down-regulation of caveolin-1. Both of these responses are associated with activation of proapoptotic pathways in this cell line.
Collapse
Affiliation(s)
- Sudhakar Chintharlapalli
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, Vet. Res. Bldg. 409, College Station, TX 77843-4466, USA
| | | | | |
Collapse
|
421
|
|
422
|
Oon Han H, Kim SH, Kim KH, Hur GC, Joo Yim H, Chung HK, Ho Woo S, Dong Koo K, Lee CS, Sung Koh J, Kim GT. Design and synthesis of oxime ethers of α-acyl-β-phenylpropanoic acids as PPAR dual agonists. Bioorg Med Chem Lett 2007; 17:937-41. [PMID: 17157019 DOI: 10.1016/j.bmcl.2006.11.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 11/09/2006] [Accepted: 11/15/2006] [Indexed: 01/24/2023]
Abstract
Oxime ethers of alpha-acyl-beta-phenylpropanoic acids were prepared to apply as PPARalpha and gamma dual agonists. Among them, compound 11l proved to exhibit potent in vitro activities with EC(50) of 19 and 13nM in PPARalpha and gamma, respectively. It showed better glucose lowering effects than rosiglitazone 1 and ameliorated the lipid profile like plasma triglyceride in db/db mice model.
Collapse
Affiliation(s)
- Hee Oon Han
- Research and Development, LG Life Sciences, 104-1 Munji-dong, Yuseong-gu, Daejon 305-380, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
423
|
Hondares E, Pineda-Torra I, Iglesias R, Staels B, Villarroya F, Giralt M. PPARdelta, but not PPARalpha, activates PGC-1alpha gene transcription in muscle. Biochem Biophys Res Commun 2007; 354:1021-7. [PMID: 17275789 DOI: 10.1016/j.bbrc.2007.01.092] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Accepted: 01/18/2007] [Indexed: 11/23/2022]
Abstract
PGC-1alpha induces mitochondrial biogenesis in muscle and its activity has been related to insulin sensitization. Here, we report that fibrates induce PGC-1alpha gene expression in muscle both in vivo and in vitro. However, only activation via PPARdelta but not PPARalpha underlies this effect. PPARdelta induces PGC-1alpha gene transcription through a PPAR-response element in the PGC-1alpha promoter. Moreover, PGC-1alpha coactivates the PPARdelta-responsiveness of its own gene. A further positive autoregulatory loop of control relies on the induction of PPARdelta expression by PGC-1alpha. These data point to a distinct value of PPARdelta rather than PPARalpha agonists in the improvement of oxidative metabolism in muscle.
Collapse
Affiliation(s)
- Elayne Hondares
- Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
424
|
Phulwani NK, Feinstein DL, Gavrilyuk V, Akar C, Kielian T. 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) and ciglitazone modulate Staphylococcus aureus-dependent astrocyte activation primarily through a PPAR-gamma-independent pathway. J Neurochem 2007; 99:1389-1402. [PMID: 17074064 PMCID: PMC2423669 DOI: 10.1111/j.1471-4159.2006.04183.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain abscesses arise from a focal parenchymal infection by various pathogens, particularly Staphylococcus aureus. We have shown that astrocytes are activated upon exposure to S. aureus and may contribute to the excessive tissue damage characteristic of brain abscess. Therefore, modulating astrocyte activation may facilitate a reduction in brain abscess severity. Peroxisome proliferator activated receptor-gamma (PPAR-gamma) agonists are potent inhibitors of microglial activation; however, the effects of these compounds on S. aureus-dependent astrocyte activation have not yet been examined. Here, we demonstrate that two chemically distinct PPAR-gamma agonists, 15-deoxy-delta12,14-prostaglandin J2 (15d-PGJ2) and ciglitazone, suppress the production of several pro-inflammatory molecules in S. aureus-stimulated astrocytes including interleukin-1beta and nitric oxide (NO). Interestingly, 15d-PGJ2 attenuated Toll-like receptor 2 (TLR2) and inducible nitric oxide synthase expression, but failed to modulate macrophage inflammatory protein-2 (MIP-2/CXCL2) production, suggesting that 15d-PGJ2 is not a global inhibitor of astrocyte activation. Another novel finding of this study was the fact that both 15d-PGJ2 and ciglitazone were capable of attenuating pre-existing astrocyte activation, indicating their potential benefit in a therapeutic setting. Importantly, 15d-PGJ2 and ciglitazone were still capable of inhibiting S. aureus-induced pro-inflammatory mediator release in PPAR-gamma-deficient astrocytes, supporting PPAR-gamma-independent effects of these compounds. Collectively, these results suggest that 15d-PGJ2 and ciglitazone exert their anti-inflammatory actions on astrocytes primarily independent of the PPAR-gamma pathway.
Collapse
Affiliation(s)
- Nirmal K. Phulwani
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Douglas L. Feinstein
- Department of Anesthesiology, University of Illinois and Jesse Brown Veterans Affairs, Chicago, Illinois, USA
| | - Vitaliy Gavrilyuk
- Department of Anesthesiology, University of Illinois and Jesse Brown Veterans Affairs, Chicago, Illinois, USA
| | - Candan Akar
- Department of Anesthesiology, University of Illinois and Jesse Brown Veterans Affairs, Chicago, Illinois, USA
| | - Tammy Kielian
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
425
|
Mittra S, Sangle G, Tandon R, Sharma S, Roy S, Khanna V, Gupta A, Sattigeri J, Sharma L, Priyadarsiny P, Khattar SK, Bora RS, Saini KS, Bansal VS. Increase in weight induced by muraglitazar, a dual PPARalpha/gamma agonist, in db/db mice: adipogenesis/or oedema? Br J Pharmacol 2007; 150:480-7. [PMID: 17211457 PMCID: PMC2189717 DOI: 10.1038/sj.bjp.0707000] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Muraglitazar, a dual PPARalpha/gamma agonist, caused a robust increase in body weight in db/db mice. The purpose of the study was to see if this increase in weight was due to oedema and/or adipogenesis. EXPERIMENTAL APPROACH The affinity of muraglitazar at PPARalpha/gamma receptors was characterized using transactivation assays. Pre-adipocyte differentiation, expression of genes for adipogenesis (aP2), fatty acid oxidation (ACO) and sodium reabsorption (ENaCgamma and Na+, K+-ATPase); haemodilution parameters and serum electrolytes were measured to delineate the role of muraglitazar in causing weight gain vis a vis rosiglitazone. KEY RESULTS Treatment with muraglitazar (10 mg kg(-1)) for 14 days significantly reduced plasma glucose and triglycerides. Reduction in plasma glucose was significantly greater than after similar treatment with rosiglitazone (10 mg kg(-1)). A marked increase in weight was also observed with muraglitazar that was significantly greater than with rosiglitazone. Muraglitazar increased aP2 mRNA and caused adipocyte differentiation in 3T3-L1 cells similar to rosiglitazone. It also caused a marked increase in ACO mRNA in the liver of the treated mice. Expression of mRNA for ENaCgamma and Na+, K+-ATPase in kidneys was up-regulated after either treatment. Increased serum electrolytes and decreased RBC count, haemoglobin and haematocrit were observed with both muraglitazar and rosiglitazone. CONCLUSIONS AND IMPLICATIONS Although muraglitazar has a better glucose lowering profile, it also has a greater potential for weight gain than rosiglitazone. In conclusion, muraglitazar causes both robust adipogenesis and oedema in a 14-day treatment of db/db mice as observed in humans.
Collapse
Affiliation(s)
- S Mittra
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - G Sangle
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - R Tandon
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - S Sharma
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - S Roy
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - V Khanna
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - A Gupta
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - J Sattigeri
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - L Sharma
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - P Priyadarsiny
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - S K Khattar
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - R S Bora
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - K S Saini
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
| | - V S Bansal
- Ranbaxy Research Laboratories, New Drug Discovery Research Gurgaon, Haryana, India
- Author for correspondence:
| |
Collapse
|
426
|
Sunder‐Plassmann R. Cytochrome P450: Another Player in the Myocardial Infarction Game? Adv Clin Chem 2007. [DOI: 10.1016/s0065-2423(06)43008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
427
|
Kho JS, Park SJ, Im SI, Choi BR, Kwak CH, Hwang JY. Peroxisome Proliferator-Activated Receptor Gamma(PPAR-γ) Agonist Improves Endothelial Function in Diabetic Patients with Metabolic Syndrome: Pivotal Role of NOx and Inflammation. Korean Circ J 2007. [DOI: 10.4070/kcj.2007.37.5.221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Jin-Sin Kho
- Department of Internal Medicine, College of Medicine, Gyeong-Sang National University, Jinju, Korea
| | - Sung-Ji Park
- Department of Internal Medicine, College of Medicine, Gyeong-Sang National University, Jinju, Korea
| | - Sung-Il Im
- Department of Internal Medicine, College of Medicine, Gyeong-Sang National University, Jinju, Korea
| | - Bong-Ryong Choi
- Department of Internal Medicine, College of Medicine, Gyeong-Sang National University, Jinju, Korea
| | - Choong-Hwan Kwak
- Department of Internal Medicine, College of Medicine, Gyeong-Sang National University, Jinju, Korea
| | - Jin-Yong Hwang
- Department of Internal Medicine, College of Medicine, Gyeong-Sang National University, Jinju, Korea
| |
Collapse
|
428
|
Mahindroo N, Peng YH, Lin CH, Tan UK, Prakash E, Lien TW, Lu IL, Lee HJ, Hsu JTA, Chen X, Liao CC, Lyu PC, Chao YS, Wu SY, Hsieh HP. Structural basis for the structure-activity relationships of peroxisome proliferator-activated receptor agonists. J Med Chem 2006; 49:6421-4. [PMID: 17034149 DOI: 10.1021/jm060663c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Type 2 diabetes has rapidly reached an epidemic proportion becoming a major threat to global public health. PPAR agonists have emerged as a leading class of oral antidiabetic drugs. We report a structure biology analysis of novel indole-based PPAR agonists to explain the structure-activity relationships and present a critical analysis of reasons for change in selectivity with change in the orientation of the same scaffolds. The results would be helpful in designing novel PPAR agonists.
Collapse
Affiliation(s)
- Neeraj Mahindroo
- Division of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
429
|
Bhaskaran S, Santanam N, Penumetcha M, Parthasarathy S. Inhibition of Atherosclerosis in Low-Density Lipoprotein Receptor-Negative Mice by Sesame Oil. J Med Food 2006; 9:487-90. [PMID: 17201634 DOI: 10.1089/jmf.2006.9.487] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Diet has profound effects on the development of atherosclerosis. Fatty acid composition, antioxidants, and other components such as lignans have major effects on the atherosclerotic process. Sesame oil has both mono- and polyunsaturated fatty acid constituents in equal proportions. In addition, it also has high levels of numerous antioxidants and inducers of peroxisome proliferator-activated receptor. The objective of this study was to determine the anti-atherosclerotic effects of sesame oil. In this study, male low-density lipoprotein (LDL) receptor (LDLR) -/- mice were fed atherogenic diet or atherogenic diet reformulated with the same level of sesame oil (sesame oil diet). Plasma lipids and atherosclerotic lesions were quantified after 3 months of feeding. Sesame oil-containing diet significantly reduced the atherosclerotic lesion formation and plasma cholesterol, triglyceride, and LDL cholesterol levels in LDLR -/- mice. These findings suggest that sesame oil could inhibit atherosclerosis lesion formation effectively, perhaps because of the synergistic actions of fatty acid and nonsaponifiable components.
Collapse
Affiliation(s)
- Shylesh Bhaskaran
- Department of Pathology, Louisiana State University, Health Sciences Center, New Orleans, LA, USA
| | | | | | | |
Collapse
|
430
|
Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O'Rahilly S, Palmer CNA, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev 2006; 58:726-41. [PMID: 17132851 DOI: 10.1124/pr.58.4.5] [Citation(s) in RCA: 734] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The three peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of the nuclear hormone receptor superfamily. They share a high degree of structural homology with all members of the superfamily, particularly in the DNA-binding domain and ligand- and cofactor-binding domain. Many cellular and systemic roles have been attributed to these receptors, reaching far beyond the stimulation of peroxisome proliferation in rodents after which they were initially named. PPARs exhibit broad, isotype-specific tissue expression patterns. PPARalpha is expressed at high levels in organs with significant catabolism of fatty acids. PPARbeta/delta has the broadest expression pattern, and the levels of expression in certain tissues depend on the extent of cell proliferation and differentiation. PPARgamma is expressed as two isoforms, of which PPARgamma2 is found at high levels in the adipose tissues, whereas PPARgamma1 has a broader expression pattern. Transcriptional regulation by PPARs requires heterodimerization with the retinoid X receptor (RXR). When activated by a ligand, the dimer modulates transcription via binding to a specific DNA sequence element called a peroxisome proliferator response element (PPRE) in the promoter region of target genes. A wide variety of natural or synthetic compounds was identified as PPAR ligands. Among the synthetic ligands, the lipid-lowering drugs, fibrates, and the insulin sensitizers, thiazolidinediones, are PPARalpha and PPARgamma agonists, respectively, which underscores the important role of PPARs as therapeutic targets. Transcriptional control by PPAR/RXR heterodimers also requires interaction with coregulator complexes. Thus, selective action of PPARs in vivo results from the interplay at a given time point between expression levels of each of the three PPAR and RXR isotypes, affinity for a specific promoter PPRE, and ligand and cofactor availabilities.
Collapse
Affiliation(s)
- Liliane Michalik
- Center for Integrative Genomics, National Research Centre "Frontiers in Genetics," University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
431
|
Warshawsky AM, Alt CA, Brozinick JT, Harkness AR, Hawkins ED, Henry JR, Matthews DP, Miller AR, Misener EA, Montrose-Rafizadeh C, Rhodes GA, Shen Q, Vance JA, Udodong UE, Wang M, Zhang TY, Zink RW. Synthesis and evaluation of aminomethyl dihydrocinnamates as a new class of PPAR ligands. Bioorg Med Chem Lett 2006; 16:6328-33. [PMID: 17005394 DOI: 10.1016/j.bmcl.2006.09.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 09/01/2006] [Accepted: 09/06/2006] [Indexed: 01/19/2023]
Abstract
PPAR ligands with varied subtype selectivity have been synthesized using an achiral aminomethyl dihydrocinnamate template. Several compounds in this series have demonstrated potent plasma glucose and triglyceride lowering capability in rodent models of type 2 diabetes.
Collapse
|
432
|
Zhang F, Sjöholm A, Zhang Q. Pioglitazone acutely influences glucose-sensitive insulin secretion in normal and diabetic human islets. Biochem Biophys Res Commun 2006; 351:750-5. [PMID: 17084385 DOI: 10.1016/j.bbrc.2006.10.103] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Accepted: 10/21/2006] [Indexed: 10/24/2022]
Abstract
We have studied acute effects of the PPARgamma agonist pioglitazone in vitro on human islets from both non-diabetic and type 2 diabetic subjects. In 5 mM glucose, pioglitazone caused a transient increase in insulin secretion in non-diabetic, but not diabetic, islets. Continuous presence of the drug suppressed insulin release in both non-diabetic and diabetic islets. In islets from non-diabetic subjects, both high glucose and tolbutamide-stimulated insulin secretion was inhibited by pioglitazone. When islets were continuously perifused with 5 mM glucose, short-term pretreatment with pioglitazone caused approximately 2-fold increase in insulin secretion after drug withdrawal. Pioglitazone pretreatment of diabetic islets restored their glucose sensitivity. Examination of cytosolic free Ca(2+) concentration ([Ca(2+)](i)) in non-diabetic islets revealed slight Ca(2+) transient by pioglitazone at 3 mM glucose with no significant changes at high glucose. Our data suggest that short-term pretreatment with pioglitazone primes both healthy and diabetic human islets for enhanced glucose-sensitive insulin secretion.
Collapse
Affiliation(s)
- Fan Zhang
- Karolinska Institutet, Department of Internal Medicine, Stockholm South Hospital, SE-11883 Stockholm, Sweden
| | | | | |
Collapse
|
433
|
Subramanian S, DeRosa MA, Bernal-Mizrachi C, Laffely N, Cade WT, Yarasheski KE, Cryer PE, Semenkovich CF. PPARalpha activation elevates blood pressure and does not correct glucocorticoid-induced insulin resistance in humans. Am J Physiol Endocrinol Metab 2006; 291:E1365-71. [PMID: 16868225 PMCID: PMC4303180 DOI: 10.1152/ajpendo.00230.2006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Fibrates, activators of the nuclear receptor PPARalpha, improve dyslipidemia, but their effects on insulin resistance and vascular disease are unresolved. To test the hypothesis that PPARalpha activation improves insulin resistance and vascular function, we determined the effects of fenofibrate in healthy adults with insulin resistance induced by short-term glucocorticoid administration. Eighteen normal-weight subjects were studied in four stages: at baseline, after 21 days of fenofibrate (160 mg/day) alone, after 3 days of dexamethasone (8 mg/day) added to fenofibrate, and after 3 days of dexamethasone added to placebo (dexamethasone alone). Dexamethasone alone caused hyperinsulinemia, increased glucose, decreased glucose disposal, and reduced insulin-induced suppression of hepatic glucose production as determined by hyperinsulinemic euglycemic clamp and increased systolic blood pressure as determined by ambulatory monitoring, features associated with an insulin-resistant state. Fenofibrate improved fasting LDL and total cholesterol in the setting of dexamethasone treatment but had no significant effect on levels of insulin or glucose, insulin-stimulated glucose disposal, or insulin suppression of glucose production during clamps, or ambulatory monitored blood pressure. In the absence of dexamethasone, fenofibrate lowered fasting triglycerides and cholesterol but unexpectedly increased systolic blood pressure by ambulatory monitoring. These data suggest that PPARalpha activation in humans does not correct insulin resistance induced by glucocorticoids and may adversely affect blood pressure.
Collapse
Affiliation(s)
- Savitha Subramanian
- Endocrinology, Metabolism, and Lipid Research, Dept. of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
434
|
Goebel M, Clemenz M, Unger T. Effective treatment of hypertension by AT(1) receptor antagonism: the past and future of telmisartan. Expert Rev Cardiovasc Ther 2006; 4:615-29. [PMID: 17081084 DOI: 10.1586/14779072.4.5.615] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Lowering blood pressure is the most effective treatment method to ensure a reduction in the total risk for cardiovascular morbidity and mortality. The renin-angiotensin system plays an important role in volume homeostasis and blood pressure regulation and is a target for several groups of pharmaceutical agents. Angiotensin II receptor blockers represent the newest class of antihypertensive compounds. They prevent the binding of angiotensin II to the subtype 1 receptor (AT(1)), which is believed to mediate most of the physiological actions relevant to the regulation of blood pressure. Telmisartan, a widely used AT(1) receptor antagonist, is a highly selective compound with high potency, a long duration of action and a tolerability profile similar to placebo. Numerous randomized clinical trials and community-based studies have demonstrated that oral telmisartan and combinations of telmisartan with hydrochlorothiazide are at least as effective in lowering blood pressure as all other hypertensive medications. This has been demonstrated in different populations of adult patients with mild-to-moderate essential hypertension, including patients with coexisting Type 2 diabetes, metabolic syndrome or renal impairment. Several large-scale, long-term, clinical endpoint studies are in progress to assess the beneficial effects of telmisartan on hypertension-related end-organ damage in patients at high risk of renal, cardiac and vascular damage whose blood pressure is well controlled. The most recent data from clinical trials and latest research regarding telmisartan will be reviewed in this article.
Collapse
Affiliation(s)
- Matthias Goebel
- Universitätsmedizin Berlin, Center for Cardiovascular Research (CCR)/Institut für Pharmakologie und Toxikologie, Charité Campus Mitte, Charité--Hessische Str. 3-4 10115 Berlin, Germany.
| | | | | |
Collapse
|
435
|
Lally S, Owens D, Tomkin GH. The different effect of pioglitazone as compared to insulin on expression of hepatic and intestinal genes regulating post-prandial lipoproteins in diabetes. Atherosclerosis 2006; 193:343-51. [PMID: 17109865 DOI: 10.1016/j.atherosclerosis.2006.09.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Revised: 08/30/2006] [Accepted: 09/27/2006] [Indexed: 10/23/2022]
Abstract
This study investigates lipoprotein composition in diabetes before and after treatment with insulin or pioglitazone and its relationship to gene expression of five genes found in liver and intestine which are involved in cholesterol homeostasis. Thirty zucker diabetic fatty fa/fa and 10 lean rats were examined. mRNA for 3-hydroxy3-methylglutaryl coenzyme A reductase (HMGCoA), microsomal triglyceride transfer protein (MTTP), Niemann Pick C1-like 1 (NPC1L1) and ATP binding cassette transporters (ABC) G5 and G8 was determined using real-time, reverse transcriptase (RT-PCR). Cholesterol, triglyceride, apo B48 and apo B100 were elevated in chylomicrons and very low density lipoproteins (VLDL) of untreated diabetic animals (p<0.02). For similar blood glucose pioglitazone was more effective than insulin in normalising the lipoproteins. In diabetic animals, HMGCoA reductase, MTTP and NPC1L1 mRNA were significantly elevated (p<0.02) and ABCG5 and ABCG8 were significantly reduced (p<0.02) in the liver. Pioglitazone significantly reduced hepatic MTTP and NPC1L1 mRNA (p<0.0001) and significantly increased ABCG5 and G8 mRNA (p<0.0001) as compared to insulin. In conclusion diabetes was associated with major changes in mRNA levels of proteins involved in the regulation of post-prandial lipoproteins. Pioglitazone and insulin have different effects on post-prandial lipoprotein metabolism in part due their effect on genes regulating cholesterol synthesis and lipoprotein assembly.
Collapse
Affiliation(s)
- S Lally
- Department of Diabetes and Endocrinology, Trinity College, Dublin 2, Ireland
| | | | | |
Collapse
|
436
|
Nayagam VM, Wang X, Tan YC, Poulsen A, Goh KC, Ng T, Wang H, Song HY, Ni B, Entzeroth M, Stünkel W. SIRT1 modulating compounds from high-throughput screening as anti-inflammatory and insulin-sensitizing agents. ACTA ACUST UNITED AC 2006; 11:959-67. [PMID: 17099246 DOI: 10.1177/1087057106294710] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The nicotinamide adenine dinucleotide (NAD(+))-dependent protein deacetylase SIRT1 has been linked to fatty acid metabolism via suppression of peroxysome proliferator-activated receptor gamma (PPAR-gamma) and to inflammatory processes by deacetylating the transcription factor NF-kappaB. First, modulation of SIRT1 activity affects lipid accumulation in adipocytes, which has an impact on the etiology of a variety of human metabolic diseases such as obesity and insulin-resistant diabetes. Second, activation of SIRT1 suppresses inflammation via regulation of cytokine expression. Using high-throughput screening, the authors identified compounds with SIRT1 activating and inhibiting potential. The biological activity of these SIRT1-modulating compounds was confirmed in cell-based assays using mouse adipocytes, as well as human THP-1 monocytes. SIRT1 activators were found to be potent lipolytic agents, reducing the overall lipid content of fully differentiated NIH L1 adipocytes. In addition, the same compounds have anti-inflammatory properties, as became evident by the reduction of the proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha). In contrast, a SIRT1 inhibitory compound showed a stimulatory activity on the differentiation of adipocytes, a feature often linked to insulin sensitization.
Collapse
|
437
|
Abstract
Type 2 diabetes is a global epidemic contributing to significant cardiovascular morbidity and mortality. The high prevalence of cardiovascular disease can largely be attributed to the metabolic syndrome with its multiple cardiovascular risk factors, including central obesity, hypertension, glucose intolerance, chronic inflammation, and dyslipidemia. The peroxisome proliferator-activated receptor-gamma agonists, the thiazolidinediones, may potentially correct the inflammatory disarray, endothelial dysfunction, dyslipidemia, and plaque vulnerability associated with diabetic cardiovascular disease through their effects on insulin resistance and fat metabolism, yet they can also exacerbate congestive heart failure. This review summarizes basic science, animal, and human data on the effects of thiazolidinediones on cardiovascular disease.
Collapse
|
438
|
Yumuk VD. Targeting Components of the Stress System as Potential Therapies for the Metabolic Syndrome: The Peroxisome-Proliferator-Activated Receptors. Ann N Y Acad Sci 2006; 1083:306-18. [PMID: 17148746 DOI: 10.1196/annals.1367.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The three peroxisome-proliferator-activated receptor (PPAR) subtypes PPAR-alpha, PPAR-gamma, and PPAR-delta are ligand-activated transcription factors of the nuclear receptor family. PPARs form obligate heterodimers with the retinoid X receptor, which bind to peroxisome-proliferator-response elements (PPREs). PPAR-alpha is expressed mainly in liver, brown fat, kidney, heart, and skeletal muscle; PPAR-gamma in intestine and adipose tissue; PPAR-alpha and PPAR-gamma are both expressed in vascular endothelium, smooth muscle cells, macrophages, and foam cells; PPAR-delta in skeletal muscle, human embryonic kidney, intestine, heart, adipose tissue, developing brain, and keratinocytes. Intense interest in the development of drugs with new mechanisms of action for the metabolic syndrome has focused attention on nuclear receptors, such as PPARs that function as regulators of energy homeostasis. Agonists of PPAR-alpha and PPAR-gamma are currently used to treat diabetic dyslipidemia and type 2 diabetes. Dual PPAR-alpha/gamma agonists and PPAR-alpha/gamma/delta pan-agonists are under investigation for treatment of cardiovascular disease and the metabolic syndrome. Selective PPAR modulators (SPPARMs) are PPAR ligands that possess desirable efficacy and improved tolerance. Efforts are being made to identify novel partial agonists or antagonists for PPAR-gamma in order to combine their antidiabetic and antiobesity effects. Glucocorticoids are major mediators of the stress response and could be the link between stress and PPAR activator signaling and thus may affect the downstream metabolic pathways involved in fuel homeostasis.
Collapse
Affiliation(s)
- Volkan D Yumuk
- Istanbul University Cerrahpasa Medical Faculty, Department of Medicine, Division of Endocrinology and Metabolism, Istanbul 34303, Turkey.
| |
Collapse
|
439
|
Haider DG, Mittermayer F, Schaller G, Artwohl M, Baumgartner-Parzer SM, Prager G, Roden M, Wolzt M. Free fatty acids normalize a rosiglitazone-induced visfatin release. Am J Physiol Endocrinol Metab 2006; 291:E885-90. [PMID: 16735449 DOI: 10.1152/ajpendo.00109.2006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The detrimental effect of elevated free fatty acids (FFAs) on insulin sensitivity can be improved by thiazolidinediones (TZDs) in patients with type 2 diabetes mellitus. It is unknown whether this salutary action of TZD is associated with altered release of the insulin-mimetic adipocytokine visfatin. In this study, we investigated whether visfatin concentrations are altered by FFA and TZD treatment. In a randomized, double-blind, placebo-controlled, parallel-group study 16 healthy volunteers received an infusion of triglycerides/heparin to increase plasma FFA after 3 wk of treatment with rosiglitazone (8 mg/day, n = 8) or placebo (n = 8), and circulating plasma visfatin was measured. As a corollary, human adipocytes were incubated with synthetic fatty acids and rosiglitazone to assess visfatin release in vitro. The results were that rosiglitazone treatment increased systemic plasma visfatin concentrations from 0.6 +/- 0.1 to 1.7 +/- 0.2 ng/ml (P < 0.01). Lipid infusion caused a marked elevation of plasma FFA but had no effect on circulating visfatin in controls. In contrast, elevated visfatin concentrations in subjects receiving rosiglitazone were normalized by lipid infusion. In isolated adipocytes, visfatin was released into supernatant medium by acute addition and long-term treatment of rosiglitazone. This secretion was blocked by synthetic fatty acids and by inhibition of phosphatidylinositol 3-kinase or Akt. In conclusion, release of the insulin-mimetic visfatin may represent a major mechanism of metabolic TZD action. The presence of FFA antagonizes this action, which may have implications for visfatin bioactivity.
Collapse
Affiliation(s)
- Dominik G Haider
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
440
|
Epple R, Azimioara M, Russo R, Xie Y, Wang X, Cow C, Wityak J, Karanewsky D, Bursulaya B, Kreusch A, Tuntland T, Gerken A, Iskandar M, Saez E, Martin Seidel H, Tian SS. 3,4,5-Trisubstituted isoxazoles as novel PPARδ agonists. Part 2. Bioorg Med Chem Lett 2006; 16:5488-92. [PMID: 16931011 DOI: 10.1016/j.bmcl.2006.08.052] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 08/08/2006] [Accepted: 08/09/2006] [Indexed: 01/20/2023]
Abstract
A series of PPARdelta-selective agonists was investigated and optimized for a favorable in vivo pharmacokinetic profile. Isoxazole LCI765 (17d) was found to be a potent and selective PPARdelta agonist with good in vivo PK properties in mouse (C(max)=5.1 microM, t(1/2)=3.1 h). LCI765 regulated expression of genes involved in energy homeostasis in relevant tissues when dosed orally in C57BL6 mice. A co-crystal structure of compound LCI765 and the LBD of PPARdelta is discussed.
Collapse
Affiliation(s)
- Robert Epple
- Department of Medicinal Chemistry, The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
441
|
Peroxisome Proliferator-Activated Receptors at the Crossroads of Obesity, Diabetes, and Cardiovascular Disease. J Am Coll Cardiol 2006. [DOI: 10.1016/j.jacc.2006.04.097] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
442
|
Wang Y, Lam KSL, Kraegen EW, Sweeney G, Zhang J, Tso AWK, Chow WS, Wat NMS, Xu JY, Hoo RLC, Xu A. Lipocalin-2 is an inflammatory marker closely associated with obesity, insulin resistance, and hyperglycemia in humans. Clin Chem 2006; 53:34-41. [PMID: 17040956 DOI: 10.1373/clinchem.2006.075614] [Citation(s) in RCA: 429] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Lipocalin-2, a 25-kDa secreted glycoprotein, is a useful biomarker for early detection of various renal injuries. Because lipocalin-2 is abundantly expressed in adipose tissue and liver, we investigated its relevance to obesity-related pathologies. METHODS We used real-time PCR and in-house immunoassays to quantify the mRNA and serum concentrations of lipocalin-2 in C57BL/KsJ db/db obese mice and their age- and sex-matched lean littermates. We analyzed the association between serum lipocalin-2 concentrations and various metabolic and inflammatory variables in 229 persons (121 men and 108 women) recruited from a previous cross-sectional study, and we evaluated the effect of the insulin-sensitizing drug rosiglitazone on serum lipocalin-2 concentrations in 32 diabetic patients (21 men and 11 women). RESULTS Compared with the lean littermates, lipocalin-2 mRNA expression in adipose tissue and liver and its circulating concentrations were significantly increased in db/db diabetic/obese mice (P <0.001). These changes were normalized after rosiglitazone treatment. In humans, circulating lipocalin-2 concentrations were positively correlated (P <0.005) with adiposity, hypertriglyceridemia, hyperglycemia, and the insulin resistance index, but negatively correlated (P = 0.002) with HDL cholesterol. There was also a strong positive association between lipocalin-2 concentrations and high sensitivity C-reactive protein (hs-CRP), independent of age, sex, and adiposity (P = 0.007). Furthermore, rosiglitazone-mediated decreases in lipocalin-2 concentrations correlated significantly with increases in insulin sensitivity (r = 0.527; P = 0.002) and decreases in hs-CRP concentrations (r = 0.509; P = 0.003). CONCLUSIONS Lipocalin-2 is an inflammatory marker closely related to obesity and its metabolic complications. Measurement of serum lipocalin-2 might be useful for evaluating the outcomes of various clinical interventions for obesity-related metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Yu Wang
- Genome Research Center and Department of Biochemistry, University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
443
|
Inoue N, Nagao K, Wang YM, Noguchi H, Shirouchi B, Yanagita T. Dietary conjugated linoleic acid lowered tumor necrosis factor-alpha content and altered expression of genes related to lipid metabolism and insulin sensitivity in the skeletal muscle of Zucker rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2006; 54:7935-9. [PMID: 17002473 DOI: 10.1021/jf061284v] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Type-2 diabetes is characterized by obesity-related insulin resistance. Insulin resistance and accompanying hyperinsulinemia have been reported to play an important role in pathogenesis of the metabolic syndrome. Conjugated linoleic acid (CLA), a mixture of positional and geometric isomers of linoleic acid, has attracted considerable attention because of its potentially beneficial biological effects. Previous studies showed that dietary CLA alleviates diabetes through improvement of glucose tolerance and insulin-stimulated glucose transport activity in skeletal muscle of diabetic rats. Skeletal muscle plays an important role both in insulin-mediated glucose metabolism and in lipid metabolism. In the present study, we evaluated comprehensively the effect of dietary CLA on the expression of genes related to lipid metabolism and insulin sensitivity in the skeletal muscle of obese, diabetic Zucker rats. After 8 weeks of feeding, expression of lipogenic genes was decreased in tendency, while expression of lipolytic genes was markedly increased by dietary CLA. Additionally, expression of genes-related insulin sensitivity, such as adiponectin receptor 1, was significantly enhanced, and mRNA level of peroxisome proliferator activated receptor-alpha, known as a transcriptional factor related lipid metabolism and insulin signaling in skeletal muscle, was markedly increased in CLA-fed rats. We also showed that dietary CLA significantly decreased the level of tumor necrosis factor-alpha (TNF-alpha), associated with the development of insulin resistance, in the skeletal muscle of Zucker rats. We suppose that the attenuated TNF-alpha accumulation in skeletal muscle may contribute to the alteration of expression of several genes and the alleviation of insulin resistance in CLA-fed Zucker rats.
Collapse
Affiliation(s)
- Nao Inoue
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga 840-8502, Japan
| | | | | | | | | | | |
Collapse
|
444
|
Arulmozhi DK, Dwyer DS, Bodhankar SL. Antipsychotic induced metabolic abnormalities: An interaction study with various PPAR modulators in mice. Life Sci 2006; 79:1865-72. [PMID: 16828808 DOI: 10.1016/j.lfs.2006.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Revised: 05/31/2006] [Accepted: 06/13/2006] [Indexed: 10/24/2022]
Abstract
Abnormalities in glucose and lipid regulation have been reported in schizophrenia during antipsychotic medications. The objectives of the present study were to evaluate the effect of various peroxisome proliferator-activated receptor modulators viz. glimepiride, rosiglitazone and fenofibrate on chlorpromazine, clozapine and ziprasidone induced hyperglycemia and hyperlipidemia in mice. Male Swiss albino mice were orally treated with chlorpromazine, clozapine and ziprasidone concurrently with the antidiabetic medications for 7 days. Plasma glucose, insulin and triglyceride levels were determined at the end of the study. Chlorpromazine and clozapine elevated the glucose and triglyceride levels in normal mice, with no effect on insulin but ziprasidone increased the basal triglyceride and insulin levels and did not have any effect on glucose. Glimepiride and rosiglitazone showed beneficial glucose and triglyceride lowering effects in chlorpromazine and clozapine animals and no effect on insulin levels. Fenofibrate significantly reduced the glucose levels only in animals treated with clozapine, and exhibited significant reduction of triglyceride levels in chlorpromazine, clozapine and ziprasidone treated animals. All three antidiabetic/hypolipidemic agents lowered triglyceride and insulin levels in ziprasidone treated animals. The results of the present studies suggest that hyperglycemia, hyperinsulinemia and hypertriglyceridemia induced by various antipsychotics may involve diverse mechanisms.
Collapse
Affiliation(s)
- D K Arulmozhi
- Department of Pharmacology, New Chemical Entity Research, Lupin Research Park, Village Nande, Taluk Mulshi, Pune 411 042, Maharashtra, India.
| | | | | |
Collapse
|
445
|
Liu J, Kalantarinia K, Rosner MH. Endocrinology and Dialysis: Management of Lipid Abnormalities Associated with End-Stage Renal Disease. Semin Dial 2006; 19:391-401. [PMID: 16970739 DOI: 10.1111/j.1525-139x.2006.00193.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The management of lipid abnormalities in patients with end-stage renal disease (ESRD) remains controversial. Large, well-designed studies investigating the effects of dyslipidemia on cardiovascular (CV) morbidity and mortality and the role of cholesterol lowering drugs in reducing mortality in ESRD patients are lacking. While it seems reasonable to suspect that dyslipidemia and its treatment in ESRD patients will affect CV morbidity and mortality similar to that in the general population, recent studies have suggested that this may not be the case. Furthermore, the pharmacokinetics of lipid lowering drugs are altered in patients with ESRD and must be considered when treating this group of patients. This article reviews the major classes of drugs used to treat dyslipidemia, emphasizing their role in patients with ESRD.
Collapse
Affiliation(s)
- Jia Liu
- Department of Internal Medicine, Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia 22908, USA
| | | | | |
Collapse
|
446
|
Abstract
PURPOSE OF REVIEW The prevalence of obesity is increasing at an alarming rate, and the obesity epidemic is driving the epidemic in type 2 diabetes. High-risk obesity is characterized by abdominal obesity with evidence of abnormal glucose and lipid metabolism, and a state of heightened inflammation. RECENT FINDINGS With increasing body weight, lipid accumulation occurs not only in adipose tissue, but in other organs as well. This 'lipotoxicity' in liver, muscle, islets, and elsewhere may account for many of the features of the metabolic syndrome. Adipose tissue produces many proteins, some of which are inflammatory cytokines, and others of which are antiinflammatory or which improve insulin sensitivity. SUMMARY The treatment of obesity requires the identification of the high-risk patient, and the institution of lifestyle measures with a long-term outlook, and an avoidance of heavily marketed fads. Current research will likely lead to improved medications in the future.
Collapse
Affiliation(s)
- Behzad Molavi
- The Central Arkansas Veterans Healthcare System, Arkansas 72205, USA.
| | | | | |
Collapse
|
447
|
Huang C, Zhang Y, Gong Z, Sheng X, Li Z, Zhang W, Qin Y. Berberine inhibits 3T3-L1 adipocyte differentiation through the PPARγ pathway. Biochem Biophys Res Commun 2006; 348:571-8. [PMID: 16890192 DOI: 10.1016/j.bbrc.2006.07.095] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2006] [Accepted: 07/18/2006] [Indexed: 02/05/2023]
Abstract
Berberine (BBR), a compound purified from Cortidis rhizoma, reduces serum cholesterol, triglycerides, and LDL-cholesterol of hypercholesterolemic patients and high fat diet fed animals, and increases hepatic LDLR mRNA and protein levels through a post-transcriptional mechanism. BBR also enhances the hypoglycemic action of insulin in diabetic animal models. Here, we show that BBR inhibits the differentiation of 3T3-L1 preadipocytes induced by DM and suppresses the mitotic clonal expansion of 3T3-L1 preadipocytes in a time- and dose-dependent manner. Gene expression analysis and Western blot analysis reveal that the BBR inhibits the mRNA and protein levels of adipogenesis related transcription factors PPARgamma and C/EBPalpha and their upstream regulator, C/EBPbeta. Reporter gene assays demonstrate that the full-length PPARgamma and alpha transcription activities are inhibited by BBR. Using real-time PCR, we have also found that the PPAR target genes that are involved in adipocyte differentiation, such as aP2, CD36, ACO, LPL, and other adipocyte markers, are suppressed by BBR. These studies suggest that BBR works on multiple molecular targets as an inhibitor of PPARgamma and alpha, and is a potential weight reducing, hypolipidemic, and hypoglycemic drug.
Collapse
Affiliation(s)
- Cheng Huang
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of CAS, Chinese Academy of Sciences, 294 Tai Yuan Road, Shanghai 200031, China
| | | | | | | | | | | | | |
Collapse
|
448
|
Denke M, Pearson T, McBride P, Gazzara RA, Brady WE, Tershakovec AM. Ezetimibe added to ongoing statin therapy improves LDL-C goal attainment and lipid profile in patients with diabetes or metabolic syndrome. Diab Vasc Dis Res 2006; 3:93-102. [PMID: 17058629 DOI: 10.3132/dvdr.2006.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
This analysis of the Ezetimibe Add-on to Statin for Effectiveness (EASE) trial examined the effectiveness and safety of ezetimibe 10 mg added to ongoing statin therapy in patients with diabetes, metabolic syndrome without diabetes, or neither disorder who had low-density lipoprotein cholesterol (LDL-C) levels exceeding National Cholesterol Education Program Adult Treatment Panel III (NCEP-ATP III) goals. After six weeks of treatment, ezetimibe added to statin reduced LDL-C in patients with diabetes by 28%, metabolic syndrome by 24%, or neither by 26%, compared with a 3% reduction for placebo for each group. In each group, more patients receiving ezetimibe plus statin reached LDL-C goal (67-74%) compared with those receiving placebo plus statin (19-22%). Other parameters demonstrating greater improvement with ezetimibe included triglycerides, apolipoprotein (Apo)B/Apo A-I ratio, high-density lipoprotein cholesterol (HDL-C), and C-reactive protein. Ezetimibe plus statin was well tolerated in each group. Ezetimibe added to ongoing statin therapy offers a new treatment option that is consistently effective in improvement of lipid profiles and attainment of LDL-C goals in patients with without diabetes or metabolic syndrome.
Collapse
Affiliation(s)
- Margo Denke
- University of Texas Health Science Center in San Antonio, Kerrville, TX, USA
| | | | | | | | | | | |
Collapse
|
449
|
Pourcet B, Fruchart JC, Staels B, Glineur C. Selective PPAR modulators, dual and pan PPAR agonists: multimodal drugs for the treatment of Type 2 diabetes and atherosclerosis. Expert Opin Emerg Drugs 2006; 11:379-401. [PMID: 16939380 DOI: 10.1517/14728214.11.3.379] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
More than 70% of patients with Type 2 diabetes mellitus (T2DM) die because of cardiovascular diseases. Current therapeutic strategies are based on separate treatment of insulin resistance and dyslipidaemia. Development of drugs with multimodal activities should improve management of the global cardiovascular risk of T2DM patients and result in better patient compliance. New therapeutic strategies are aimed at targeting the entire spectrum of dysfunctioning organs, cells and regulatory pathways implicated in the pathogenesis of T2DM, dyslipidaemia and atherosclerosis. PPAR family members play major roles in the regulation of lipid metabolism, glucose homeostasis and inflammatory processes, making these transcription factors ideal targets for therapeutic strategies against these diseases. This review discusses why PPARs and development of novel selective PPAR modulators, dual and pan PPAR agonists constitute promising approaches for the treatment of diabetes, dyslipidaemia and atherosclerosis.
Collapse
Affiliation(s)
- Benoit Pourcet
- Institut Pasteur de Lille, Département d'Athérosclérose, 01 rue du Professeur Calmette, BP 245, Lille 59019, France
| | | | | | | |
Collapse
|
450
|
Colca J. 66th Annual Meeting of the American Diabetes Association: 9 - 13 June 2006, Washington DC, USA. Expert Opin Investig Drugs 2006; 15:1119-23. [PMID: 16916278 DOI: 10.1517/13543784.15.9.1119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The 66th annual meeting of the American Diabetes Association was held in Washington, DC and attracted the largest attendance ever for this meeting. This meeting continues to be grow in size and quality as the most useful and successful confluence of scientific, medical, and commercial advances relating to diabetes.
Collapse
Affiliation(s)
- Jerry Colca
- Kalamazoo Metabolic Research, 125 South Kalamazoo Mall 640, Kalamazoo, MI 49007, USA.
| |
Collapse
|