Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
2.
Cytometry A ; 95(8): 869-884, 2019 08.
Article in English | MEDLINE | ID: mdl-30994973

ABSTRACT

Ezetimibe (EZE) and glucuronidated EZE (EZE-Glu) differentially target Niemann-Pick C1-like 1 (NPC1L1) and CD13 (aminopeptidase-N) to inhibit intestinal cholesterol absorption and cholesterol processing in other cells, although the precise molecular mechanisms are not fully elucidated. Cellular effects of EZE, EZE-Glu, and the low-absorbable EZE-analogue S6130 were investigated on human monocyte-derived macrophages upon loading with atherogenic lipoproteins. EZE and S6130, but not EZE-Glu disturbed the colocalization of CD13 and its coreceptor CD64 (Fcγ receptor I) in membrane microdomains, and decreased the presence of both receptors in detergent-resistant membrane fractions. Biotinylated cholesterol absorption inhibitor C-5 (i.e., derivative of EZE) was rapidly internalized to perinuclear tubular structures of cells, resembling endoplasmic reticulum (ER), but CD13 was detected on extracellular sites of the plasma membrane and endolysosomal vesicles. Administration of EZE, but not of EZE-Glu or S6130, was associated with decreased cellular cholesteryl ester content, indicating the sterol-O acyltransferase 1 (SOAT1)-inhibition by EZE. Furthermore, EZE decreased the expression of molecules involved in cholesterol uptake and synthesis, in parallel with increased apolipoprotein A-I-mediated cholesterol efflux and upregulation of efflux-effectors. However, NPC1L1 the other claimed molecular target of EZE, was not detected in macrophages, thereby excluding this protein as target for EZE in macrophages. Thus, EZE is very likely a CD13-linked microdomain-disruptor and SOAT1-inhibitor in macrophages leading to in vitro anti-atherosclerotic effects through a decrease of net cellular cholesterol content. © 2019 International Society for Advancement of Cytometry.


Subject(s)
CD13 Antigens/ultrastructure , Cholesterol/isolation & purification , Flow Cytometry , Membrane Transport Proteins/genetics , Receptors, IgG/ultrastructure , Atherosclerosis/genetics , Biological Transport/drug effects , CD13 Antigens/antagonists & inhibitors , Cholesterol/metabolism , Ezetimibe/pharmacology , Glucuronates/genetics , Humans , Macrophages/metabolism , Macrophages/ultrastructure , Membrane Microdomains/drug effects , Membrane Microdomains/ultrastructure , Membrane Transport Proteins/metabolism , Monocytes/metabolism , Monocytes/ultrastructure , Receptors, IgG/antagonists & inhibitors
3.
BMC Med ; 16(1): 232, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30541568

ABSTRACT

Lifestyle factors conferring increased diabetes risk are associated with elevated basal insulin levels (hyperinsulinaemia). The latter predicts later obesity in children and adolescents.A causal role of hyperinsulinaemia for adipose tissue growth is probable because pharmacological reduction of insulin secretion lowers body weight in people who are obese. Genetic inactivation of insulin gene alleles in mice also lowers their systemic insulin levels and prevents or ameliorates high-fat diet-induced obesity. Hyperinsulinaemia causes weight gain because of a physiological property of insulin. Insulin levels that are on the high side of normal, or which are slightly elevated, are sufficient to suppress lipolysis and promote lipogenesis in adipocytes. The effect of insulin on glucose transport or hepatic glucose production requires six or two times higher hormone levels, respectively.It seems justified to suggest a lifestyle that avoids high insulin levels in order to limit anabolic fat tissue activity.


Subject(s)
Insulin/metabolism , Life Style , Obesity/etiology , Adipose Tissue/growth & development , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Glucose/metabolism , Humans , Hyperinsulinism/complications , Hyperinsulinism/metabolism , Insulin Resistance , Lipolysis , Obesity/genetics , Obesity/metabolism , Weight Gain
4.
Cell Rep ; 18(9): 2269-2279, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28249170

ABSTRACT

Plasma metabolite concentrations reflect the activity of tissue metabolic pathways and their quantitative determination may be informative about pathogenic conditions. We searched for plasma lipid species whose concentrations correlate with various parameters of glucose homeostasis and susceptibility to type 2 diabetes (T2D). Shotgun lipidomic analysis of the plasma of mice from different genetic backgrounds, which develop a pre-diabetic state at different rates when metabolically stressed, led to the identification of a group of sphingolipids correlated with glucose tolerance and insulin secretion. Quantitative analysis of these and closely related lipids in the plasma of individuals from two population-based prospective cohorts revealed that specific long-chain fatty-acid-containing dihydroceramides were significantly elevated in the plasma of individuals who will progress to diabetes up to 9 years before disease onset. These lipids may serve as early biomarkers of, and help identify, metabolic deregulation in the pathogenesis of T2D.


Subject(s)
Biomarkers/blood , Ceramides/blood , Diabetes Mellitus, Type 2/blood , Disease Susceptibility/blood , Adult , Aged , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Tolerance Test/methods , Humans , Insulin/blood , Insulin Resistance/physiology , Lipids/blood , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Middle Aged , Prospective Studies , Sphingolipids/blood
5.
Mol Cell Endocrinol ; 428: 133-41, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27033324

ABSTRACT

The sodium-dependent organic anion transporter SOAT specifically transports sulfated steroid hormones and is supposed to play a role in testicular steroid regulation and male fertility. The present study aimed to identify novel specific SOAT inhibitors for further in vitro and in vivo studies on SOAT function. More than 100 compounds of different molecular structures were screened for inhibition of the SOAT-mediated transport of dehydroepiandrosterone sulfate in stably transfected SOAT-HEK293 cells. Twenty-five of these with IC50 values covering four orders of magnitude were selected as training set for 3D pharmacophore modelling. The SOAT pharmacophore features were calculated by CATALYST and consist of three hydrophobic sites and two hydrogen bond acceptors. By substrate database screening, compound T 0511-1698 was predicted as a novel SOAT inhibitor with an IC50 of 15 µM. This value was confirmed by cell-based transport assays. Therefore, the developed SOAT pharmacophore model demonstrated its suitability in predicting novel SOAT inhibitors.


Subject(s)
Dehydroepiandrosterone Sulfate/metabolism , Drug Evaluation, Preclinical , Models, Molecular , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/chemistry , Quantitative Structure-Activity Relationship , Bile Acids and Salts/chemistry , Bile Acids and Salts/pharmacology , HEK293 Cells , Humans , Inhibitory Concentration 50 , Reproducibility of Results
6.
Handb Exp Pharmacol ; 233: 373-435, 2016.
Article in English | MEDLINE | ID: mdl-26330256

ABSTRACT

The armamentarium for the treatment of dyslipidemia today comprises six different modes of action with overall around 24 different drugs. The treatment of lipid disorders was revolutionized with the introduction of statins which have become the most important therapeutic option available today to reduce and prevent atherosclerosis and its detrimental consequences like cardiovascular diseases and stroke. With and optimized reduction of elevated LDL levels with statins, the risk for cardiovascular diseases (CVD) can be reduced by 30%, indicating a residual remaining risk of 70% for the development and progression of CVD notifying still a high medical need for more effective antilipidemic drugs. Consequently, the search for novel lipid-modifying drugs is still one of the most active areas in research and development in the pharmaceutical industry. Major focus lies on approaches to LDL-lowering drugs superior to statins with regard to efficacy, safety, and patient compliance and on approaches modifying plasma levels and functionality of HDL particles based on the clinically validated inverse relationship between high-plasma HDL levels and the risk for CVD. The available drugs today for the treatment of dyslipidemia are small organic molecules or nonabsorbable polymers for binding of bile acids to be applied orally. Besides small molecules for novel targets, biological drugs such as monoclonal antibodies, antisense or gene-silencing oligonucleotides, peptidomimetics, reconstituted synthetic HDL particles and therapeutic proteins are novel approaches in clinical development are which have to be applied by injection or infusion. The promising clinical results of several novel drug candidates, particularly for LDL cholesterol lowering with monoclonal antibodies raised against PCSK9, may indicate more than a decade after the statins, the entrance of new breakthrough therapies to treat lipid disorders.


Subject(s)
Dyslipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Ezetimibe/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Niacin/therapeutic use , Triglycerides/blood
7.
Biol Chem ; 392(1-2): 77-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21194371

ABSTRACT

Membrane transporters are major determinants for the pharmacokinetic, safety and efficacy behavior of drugs. Available technologies to study function and structure of transport proteins has strongly stimulated research in transporter biology and uncovered their importance for the drug discovery and development process, especially for drug absorption and disposition. Physiological transport systems are investigated as potential ferries to improve drug absorption and membrane permeation and to achieve organ-specific drug action. In particular, the bile acid transport systems in the liver and the small intestine and the oligopeptide transporters are of significant importance for molecular drug delivery.


Subject(s)
Bile Acids and Salts , Drug Delivery Systems , Membrane Transport Proteins/metabolism , Bile Acids and Salts/metabolism , Membrane Transport Proteins/chemistry , Molecular Structure
8.
Regul Pept ; 164(2-3): 58-64, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20570597

ABSTRACT

The glucagon-like peptide-1 (GLP-1) receptor represents an established therapeutic target in type 2 diabetes mellitus (T2DM). Agents that activate this receptor improve glucose tolerance alongside a low risk of hypoglycaemia, and have the potential to modify disease progression. Lixisenatide is a new potent and selective GLP-1 receptor agonist currently in development. The preclinical pharmacological profile of Lixisenatide suggests actions that are highly relevant to the long-term maintenance of glucose homeostasis. Lixisenatide protected Ins-1 cells (a rat-derived beta-cell line) from both lipid- and cytokine-induced apoptosis. More importantly, Lixisenatide also prevented lipotoxicity-induced insulin depletion in human islets and preserved insulin production, storage and pancreatic beta-cell function in vitro. Enhancement of insulin biosynthesis and pancreatic beta-cell volume could also be demonstrated in animal models of type 2 diabetes. The improvement of glucose-stimulated insulin secretion provided by Lixisenatide occurred in a strictly glucose-dependent manner. In animal models of diabetes, Lixisenatide improved basal blood glucose and HbA(1c) with a rapid onset and sustained duration of action, and prevented the deterioration of pancreatic responsiveness and glucose homeostasis. Lixisenatide also delayed gastric emptying and reduced food intake. The efficacy/safety profile of Lixisenatide is currently being studied further in an extensive ongoing Phase III clinical study programme. This article reviews the preclinical pharmacological profile of Lixisenatide.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Peptides/therapeutic use , Receptors, Glucagon/antagonists & inhibitors , Animals , Glucagon-Like Peptide-1 Receptor , Humans
9.
Obesity (Silver Spring) ; 18(10): 1952-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20168311

ABSTRACT

This is the first study to examine the effect of subchronic olanzapine (OLZ) on energy homeostasis in rats, covering all aspects of energy balance, including energy intake as metabolizable energy, storage, and expenditure. We further analyzed whether, and by which mechanism, the CB1-antagonist AVE1625 might attenuate OLZ-induced body weight gain. For this purpose, we selected juvenile female Hanover Wistar rats that robustly and reproducibly demonstrated weight gain on OLZ treatment, accepting limitations to model the aberrations on lipid and carbohydrate metabolism. Rats received 2 mg/kg OLZ orally twice daily for 12 days. Body weight and body composition were analyzed. Moreover daily food intake, energy expenditure, and substrate oxidation were determined in parallel to motility and body core temperature. OLZ treatment resulted in substantial body weight gain, in which lean and fat mass increased significantly. OLZ-treated rats showed hyperphagia that manifested in increased carbohydrate oxidation and lowered fat oxidation (FO). Energy expenditure was increased, motility decreased, but there was no indication for hypothermia in OLZ-treated rats. Coadministration of OLZ and AVE1625 (10 mg/kg orally once daily) attenuated body weight gain, diminishing the enhanced food intake while maintaining increased energy expenditure and decreased motility. Our data reveal that energy expenditure was enhanced in OLZ-treated rats, an effect not critically influenced by motility. Energy uptake, however, exceeded energy expenditure and led to a positive energy balance, confirming hyperphagia as the major driving factor for OLZ-induced weight gain. Combination of OLZ treatment with the CB1-antagonist AVE1625 attenuated body weight gain in rats.


Subject(s)
Anti-Obesity Agents/therapeutic use , Antipsychotic Agents/adverse effects , Benzodiazepines/adverse effects , Energy Metabolism/drug effects , Hydrocarbons, Halogenated/therapeutic use , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Sulfonamides/therapeutic use , Weight Gain/drug effects , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Antipsychotic Agents/administration & dosage , Benzodiazepines/administration & dosage , Dietary Carbohydrates/metabolism , Dietary Fats/metabolism , Energy Intake/drug effects , Female , Hydrocarbons, Halogenated/administration & dosage , Hydrocarbons, Halogenated/pharmacology , Hyperphagia/drug therapy , Hyperphagia/etiology , Hyperphagia/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/prevention & control , Olanzapine , Oxidation-Reduction , Rats , Rats, Wistar , Sulfonamides/administration & dosage , Sulfonamides/pharmacology
10.
Acta Pharmacol Sin ; 30(7): 935-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19503102

ABSTRACT

AIM: To investigate the efficacy of the peroxisome proliferator-activated receptor-alpha (PPARalpha) agonist, AVE8134, in cellular and experimental models of cardiac dysfunction and heart failure. METHODS: In Sprague Dawley rats with permanent ligation of the left coronary artery (post-MI), AVE8134 was compared to the PPARgamma agonist rosiglitazone and in a second study to the ACE inhibitor ramipril. In DOCA-salt sensitive rats, efficacy of AVE8134 on cardiac hypertrophy and fibrosis was investigated. Finally, AVE8134 was administered to old spontaneously hypertensive rats (SHR) at a non-blood pressure lowering dose with survival as endpoint. In cellular models, we studied AVE8134 on hypertrophy in rat cardiomyocytes, nitric oxide signaling in human endothelial cells (HUVEC) and LDL-uptake in human MonoMac-6 cells. RESULTS: In post-MI rats, AVE8134 dose-dependently improved cardiac output, myocardial contractility and relaxation and reduced lung and left ventricular weight and fibrosis. In contrast, rosiglitazone exacerbated cardiac dysfunction. Treatment at AVE8134 decreased plasma proBNP and arginine and increased plasma citrulline and urinary NOx/creatinine ratio. In DOCA rats, AVE8134 prevented development of high blood pressure, myocardial hypertrophy and cardiac fibrosis, and ameliorated endothelial dysfunction. Compound treatment increased cardiac protein expression and phosphorylation of eNOS. In old SHR, treatment with a low dose of AVE8134 improved cardiac and vascular function and increased life expectancy without lowering blood pressure. AVE8134 reduced phenylephrine-induced hypertrophy in adult rat cardiomyocytes. In HUVEC, Ser-1177-eNOS phosphorylation but not eNOS expression was increased. In monocytes, AVE8134 increased the expression of CD36 and the macrophage scavenger receptor 1, resulting in enhanced uptake of oxidized LDL. CONCLUSION: The PPARalpha agonist AVE8134 prevents post-MI myocardial hypertrophy, fibrosis and cardiac dysfunction. AVE8134 has beneficial effects against hypertension-induced organ damages, resulting in decreased mortality. The compound exerts its protective properties by a direct effect on cardiomyocyte hypertrophy, but also indirectly via monocyte signaling and increased endothelial NO production.Acta Pharmacologica Sinica (2009) 30: 935-946; doi: 10.1038/aps.2009.58; published online 8 June 2009.


Subject(s)
Benzoates/therapeutic use , Cardiotonic Agents/therapeutic use , Heart Failure , Oxazoles/therapeutic use , PPAR alpha/agonists , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Benzoates/chemistry , Benzoates/metabolism , Biomarkers/metabolism , Cardiotonic Agents/chemistry , Cardiotonic Agents/metabolism , Cell Line , Disease Progression , Heart Failure/drug therapy , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics , Humans , Hypoglycemic Agents/therapeutic use , Male , Molecular Structure , Nitric Oxide Synthase Type III/metabolism , Oxazoles/chemistry , Oxazoles/metabolism , PPAR alpha/metabolism , Ramipril/therapeutic use , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Rosiglitazone , Survival Rate , Thiazolidinediones/therapeutic use
11.
Cell Signal ; 21(2): 324-38, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19010410

ABSTRACT

Synthesis and degradation of lipids in mammalian adipocytes are tightly and coordinatedly regulated by insulin, fatty acids, reactive oxygen species and drugs. Conversely, the lipogenic or lipolytic state of adipocytes is communicated to other tissues by the secretion of soluble adipocytokines. Here we report that insulin, palmitate, H(2)O(2) and the antidiabetic sulfonylurea drug glimepiride induce the release of the typical lipid droplet (LD) protein, perilipin-A, as well as typical plasma membrane microdomain (DIGs) proteins, such as caveolin-1 and the glycosylphosphatidylinositol (GPI)-anchored proteins, Gce1 and CD73 from rat adipocytes. According to biochemical and morphological criteria these LD and GPI-proteins are embedded within two different types of phospholipid-containing membrane vesicles, collectively called adiposomes. Adiposome release was not found to be causally related to cell lysis or apoptosis. The interaction of Gce1 and CD73 with the adiposomes apparently depends on their intact GPI anchor. Pull-down of caveolin-1, perilipin-A and CD73 together with phospholipids (via binding to annexin-V) as well as mutually of caveolin-1 with CD73 or perilipin-A (via coimmunoprecipitation) argues for their colocalization within the same adiposome vesicle. Taken together, certain lipogenic and anti-lipolytic agents induce the specific release of a subset of LD and DIGs proteins, including certain GPI-proteins, in adiposomes from primary rat adipocytes. Given the (c)AMP-degrading activities of Gce1 and CD73 and LD-forming function of perilipin-A and caveolin-1, the physiological relevance of the release of adiposomes from adipocytes may rely on the intercellular transfer of lipogenic and anti-lipolytic information.


Subject(s)
Adipocytes/metabolism , Glycosylphosphatidylinositols/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Secretory Vesicles/metabolism , 5'-Nucleotidase/metabolism , Adipocytes/drug effects , Adipocytes/enzymology , Animals , Annexin A5/metabolism , Carrier Proteins , Caveolin 1/metabolism , Cyclic AMP/metabolism , Exosomes/physiology , Hydrogen Peroxide/pharmacology , Lipid Metabolism/drug effects , Membrane Microdomains/drug effects , Palmitates/pharmacology , Perilipin-1 , Rats , Sulfonylurea Compounds/pharmacology
12.
Endocrinology ; 149(5): 2557-66, 2008 May.
Article in English | MEDLINE | ID: mdl-18276749

ABSTRACT

The CB1 receptor antagonist, rimonabant, affects the endocannabinoid system and causes a sustained reduction in body weight (BW) despite the transient nature of the reduction in food intake. Therefore, in a multiple-dose study, female candy-fed Wistar rats were treated with rimonabant (10 mg/kg) and matched with pair-fed rats to distinguish between hypophagic action and hypothesized effects on energy expenditure. Within the first week of treatment, rimonabant reduced BW nearly to levels of standard rat chow-fed rats. Evaluation of energy balance (energy expenditure measured by indirect calorimetry in relation to metabolizable energy intake calculated by bomb calorimetry) revealed that increased energy expenditure based on increased fat oxidation contributed more to sustained BW reduction than reduced food intake. A mere food reduction through pair feeding did not result in comparable effects because animals reduced their energy expenditure to save energy stores. Because fat oxidation measured by indirect calorimetry increased immediately after dosing in the postprandial state, the acute effect of rimonabant on lipolysis was investigated in postprandial male rats. Rimonabant elevated free fatty acids postprandially, demonstrating an inherent pharmacological activity of rimonabant to induce lipolysis and not secondarily postabsorptively due to reduced food intake. We conclude that the weight-reducing effect of rimonabant was due to continuously elevated energy expenditure based on increased fat oxidation driven by lipolysis from fat tissue as long as fat stores were elevated. When the amount of endogenous fat stores declined, rimonabant-induced increased energy expenditure was maintained by a re-increase in food intake.


Subject(s)
Candy , Eating/drug effects , Energy Metabolism/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Weight Loss/drug effects , Animals , Anti-Obesity Agents/pharmacology , Blood Glucose/analysis , Blood Glucose/drug effects , Body Weight/drug effects , Diet , Energy Metabolism/physiology , Fatty Acids, Nonesterified/blood , Female , Liver Glycogen/analysis , Male , Photoperiod , Rats , Rats, Wistar , Rimonabant , Time Factors , Weight Loss/physiology
13.
Arzneimittelforschung ; 58(11): 574-80, 2008.
Article in English | MEDLINE | ID: mdl-19137908

ABSTRACT

AVE2268, a substituted glycopyranoside, is an orally active and selective inhibitor of sodium-dependent glucose transporter 2 (SGLT2; IC50 = 13 nmol/L). Investigation of the pharmacological profile of AVE2268 on urinary glucose excretion (UGE) and blood glucose after glucose challenge (po or Intraperitoneal) was performed in mice and rats. AVE2268 caused a dose-dependent increase of UGE in mice (ID30 = 79 +/- 8.1 mg/kg p.o.) and rats (ID30 = 39.8 +/- 4.0 mg/kg p.o.). AVE2268 in mice was more potent to decrease blood glucose ascent when glucose was given intraperitoneally (ID50 = 13.2 +/- 3.9 mg/ kg), compared to orally administered glucose (ID50 = 26.1 +/- 3.9 mg/kg), showing that AVE2268 has no effects on SGLT 1 in the gut in vivo, which is in accordance with ist very low affinity to the SGLT 1 in vitro (IC50 >10,000 nmol/L). During an oral glucose tolerance test, AVE2268 dose-dependently increased UGE, with subsequent decreases of AUC and blood glucose. A highly significant inverse correlation between AUC and UGE was found (p < 0.001). The increase in UGE is linked to the inhibition of SGLT2 only. This profile renders AVE2268 as a new antidiabetic drug for the treatment of type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Glucosides/pharmacology , Glycosuria/metabolism , Hypoglycemic Agents/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Glucose/metabolism , Glucose Tolerance Test , Humans , Kidney/drug effects , Kidney/metabolism , Mice , Phlorhizin/pharmacology , Rats , Sodium-Glucose Transporter 2 , Sodium-Glucose Transporter 2 Inhibitors
14.
Am J Physiol Endocrinol Metab ; 294(1): E88-96, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17957038

ABSTRACT

The mitochondrial pyruvate dehydrogenase complex (PDC) is inactivated in many tissues during starvation and diabetes. We investigated carbohydrate oxidation (CHO) and the regulation of the PDC in lean and obese Zucker diabetic fatty (ZDF) rats during fed and starved conditions as well as during an oral glucose load without and with pharmacologically reduced levels of free fatty acids (FFA) to estimate the relative contribution of FFA on glucose tolerance, CHO, and PDC activity. The increase in total PDC activity (20-45%) was paralleled by increased protein levels ( approximately 2-fold) of PDC subunits in liver and muscle of obese ZDF rats. Pyruvate dehydrogenase kinase-4 (PDK4) protein levels were higher in obese rats, and consequently PDC activity was reduced. Although PDK4 protein levels were rapidly downregulated (57-62%) in both lean and obese animals within 2 h after glucose challenge, CHO over 3 h as well as the peak of PDC activity (1 h after glucose load) in liver and muscle were significantly lower in obese rats compared with lean rats. Similar differences were obtained with pharmacologically suppressed FFA by nicotinic acid, but with significantly improved glucose tolerance in obese rats, as well as increased CHO and delta increases in PDC activity (0-60 min) both in muscle and liver. These results demonstrated the suppressive role of FFA acids on the measured parameters. Furthermore, the results clearly demonstrate a rapid reactivation of PDC in liver and muscle of lean and obese rats after a glucose load and show that PDC activity is significantly lower in obese ZDF rats.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Animals , Blood Glucose/metabolism , Fatty Acids, Nonesterified/metabolism , Glucose Tolerance Test , Hyperinsulinism/metabolism , Male , Oxidation-Reduction , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats , Rats, Zucker , Starvation/metabolism
15.
Am J Physiol Endocrinol Metab ; 293(3): E826-32, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17595216

ABSTRACT

The objective of the present study was to investigate in fed Wistar rats whether the cannabinoid-1 (CB1) receptor antagonist AVE1625 causes primary effects on metabolic blood and tissue parameters as well as metabolic rate, which are independent of reduced caloric intake. After single administration to rats postprandially, AVE1625 caused a slight dose-dependent increase in basal lipolysis. Six hours after single administration, liver glycogen content was dose-dependently reduced to approximately 60% of that of untreated controls. These findings demonstrate a primary acute effect of AVE1625 on induction of 1) lipolysis from fat tissue (increased FFA) and 2) glycogenolysis from the liver (reduced hepatic glycogen). Measured by indirect calorimetry, AVE1625 caused an immediate increase in total energy expenditure, a long-lasting increase of fat oxidation, and a transient increase of glucose oxidation, which were consistent with the acute findings on metabolic blood and tissue parameters. We conclude that, in addition to the well-investigated effects of CB1 receptor antagonists to reduce caloric intake and subsequently body weight, this pharmacological approach is additionally linked to inherently increased lipid oxidation. This oxidation is driven by persistently increased lipolysis from fat tissues, independently of reduced caloric intake, and might significantly contribute to the weight-reducing effect.


Subject(s)
Body Weight/physiology , Eating/physiology , Energy Intake/physiology , Energy Metabolism/physiology , Hydrocarbons, Halogenated/administration & dosage , Lipid Peroxidation/physiology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Sulfonamides/administration & dosage , Animals , Body Weight/drug effects , Eating/drug effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Lipid Peroxidation/drug effects , Male , Rats , Rats, Wistar
16.
J Hepatol ; 47(1): 114-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17448567

ABSTRACT

BACKGROUND/AIMS: Mutations in the ATP8B1 gene can cause Progressive Familial Intrahepatic Cholestasis type 1. We have previously reported that Atp8b1(G308V/G308V) mice, a model for PFIC1, have slightly, but significantly, higher baseline serum bile salt (BS) concentrations compared to wt mice. Upon BS feeding, serum BS concentrations strongly increased in Atp8b1-deficient mice. Despite these findings, we observed only mildly impaired canalicular BS transport. In the present report we tested the hypothesis that Atp8b1(G308V/G308V) mice hyperabsorb BS in the intestine during BS feeding. METHODS: Intestinal BS absorption was measured in intestinal perfusion and in intestinal explants. In addition, we measured BS concentrations in portal blood. Ileal expression of the Fxr-targets Asbt, Ilbp and Shp was assessed. RESULTS: In wt and Atp8b1(G308V/G308V) mice, intestinal taurocholate absorption is primarily mediated by the ileal bile salt transporter Asbt. Neither of the experimental systems revealed enhanced absorption of BS in Atp8b1(G308V/G308V) mice compared to wt mice. In line with these observations, we found no difference in the ileal protein expression of Asbt. Induction of Shp expression during BS feeding also demonstrated that Fxr signalling is intact in Atp8b1(G308V/G308V) mice. CONCLUSIONS: The accumulation of BS in plasma of Atp8b1(G308V/G308V) mice during BS feeding is not caused by increased intestinal BS absorption.


Subject(s)
Adenosine Triphosphatases/genetics , Bile Acids and Salts/metabolism , Intestinal Absorption/physiology , Animals , Bile Acids and Salts/administration & dosage , Bile Acids and Salts/blood , Cholates/administration & dosage , Cholestasis, Intrahepatic/genetics , DNA-Binding Proteins/metabolism , Diet , Disease Models, Animal , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression , Intestinal Absorption/genetics , Mice , Mice, Knockout , Organic Anion Transporters, Sodium-Dependent/metabolism , Perfusion , Phospholipid Transfer Proteins , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Symporters/metabolism , Taurocholic Acid/metabolism , Transcription Factors/metabolism
17.
Curr Med Chem ; 13(9): 997-1016, 2006.
Article in English | MEDLINE | ID: mdl-16611081

ABSTRACT

The enterohepatic circulation of bile acids is a major regulator of serum cholesterol homeostasis. After biosynthesis from cholesterol in the liver, bile acids are secreted with bile into the lumen of the small intestine to aid in the digestion and absorption of fat and fat-soluble vitamins. The bile acids are nearly quantitatively reabsorbed in the terminal ileum by a Na+-dependent transport system (IBAT) and are transported with portal blood to the liver and taken up by a second Na+-/bile acid cotransporter (LBAT) to be resecreted into bile. In the liver bile acids inhibit the rate-limiting enzyme for the conversion of cholesterol into bile acid: cholesterol-7alpha-hydroxylase; interruption of the enterohepatic circulation of bile acids withdraws this feedback inhibition and leads to an upregulation of hepatic LDL-receptors with a concomitant decrease of serum LDL-levels. Specific inhibitors of the ileal bile acid transporter belonging to different chemotypes have been developed in recent years for this purpose, some now entering clinical stage. To exert a profound systemic effect these compounds do not need to be available systemically but can act from the luminal side of the small intestine, which offers the advantage to avoid the well-known adverse side effects of other hypolipidemic drugs like statins due to metabolism and drug-drug interactions in the liver. This implies several aspects in compound optimization and drug development quite different from standard procedures, for example the concept of low absorption drugs was established to avoid systemic side effects. The review article covers the mechanistic and therapeutic principles of the approach and presents an overview on the molecular target, the discovery of specific inhibitors and respective optimization strategies.


Subject(s)
Bile Acids and Salts/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/therapy , Hypolipidemic Agents/therapeutic use , Amino Acid Sequence , Humans , Molecular Sequence Data , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism
18.
Biochim Biophys Acta ; 1758(1): 45-54, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16458850

ABSTRACT

Intestinal cholesterol absorption is specifically inhibited by the 2-azetidinone cholesterol absorption inhibitor ezetimibe. Photoreactive ezetimibe analogues specifically label a 145-kDa protein in the brush border membrane of enterocytes from rabbit small intestine identified as aminopeptidase N (CD13). In zebrafish and mouse small intestinal cytosol, a heterocomplex of M(r) 52 kDa between annexin II and caveolin 1 was suggested as a target of ezetimibe. In contrast, in the cytosol and brush border membrane vesicles (BBMV) from rabbit small intestine of control animals or rabbits treated with the nonabsorbable cholesterol absorption inhibitor AVE 5530, both annexin II and caveolin 1 were exclusively present as monomers without any heterocomplex formation. Upon immunoprecipitation with annexin II a 52-kDa band was observed after immunostaining with annexin II antibodies, whereas no staining of a 52-kDa band occurred with anti-caveolin 1 antibodies. Vice versa, a 52-kDa band obtained by immunoprecipitation with caveolin 1 antibodies did not stain with annexin II-antibodies. The intensity of the 52-kDa band was dependent on the amount of antibody and was also observed with anti-actin or anti-APN antibodies suggesting that the 52-kDa band is a biochemical artefact. After incubation of cytosol or BBMV with radioactively labelled ezetimibe analogues, no significant amounts of the ezetimibe analogues could be detected in the immunoprecipitate with caveolin-1 or annexin II antibodies. Photoaffinity labelling of rabbit small intestinal BBMV with ezetimibe analogues did not result in labelling of proteins being immunoreactive with annexin II, caveolin 1 or a 52-kDa heterocomplex. These findings indicate that the rabbit small intestine does not contain an annexin II/caveolin 1 heterocomplex as a target for ezetimibe.


Subject(s)
Annexin A2/metabolism , Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Caveolin 1/metabolism , Intestine, Small/metabolism , Rabbits/metabolism , Absorption , Animals , Antibodies/immunology , Antibodies/metabolism , Anticholesteremic Agents/metabolism , Azetidines/metabolism , Cytoplasm/metabolism , Enterocytes/metabolism , Ezetimibe , Immunoprecipitation , Intestine, Small/drug effects , Male , Microvilli/metabolism
19.
Cytometry A ; 69(3): 206-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16479608

ABSTRACT

Aminopeptidase N (CD13) was recently identified as a molecular target of the cholesterol absorption inhibitor Ezetimib. Regarding that CD13 is expressed in lipid rafts of monocytic cells, we have investigated whether Ezetimib influences raft function in these cells. Expression of raft-associated antigens (CD11b, CD13, CD14, CD16, CD36, and CD64) was followed by flow cytometry and/or immunoblot in human monocyte-derived macrophages in response to in vitro administration of Ezetimib. Cellular redistribution of CD13 was assessed by confocal imaging. Ezetimib significantly decreased the surface expression of CD13, CD16, CD64, and CD36; furthermore, it induced a shift of CD13 from plasma membrane to intracellular vesicles, and thus it quite likely modulated monocytic raft-assembly.


Subject(s)
Antigens, Surface/analysis , Azetidines/pharmacology , Macrophages/drug effects , Membrane Microdomains/drug effects , Anticholesteremic Agents/pharmacology , Antigens, CD/analysis , Antigens, CD/metabolism , Antigens, Surface/metabolism , CD11b Antigen/analysis , CD11b Antigen/metabolism , CD13 Antigens/analysis , CD13 Antigens/metabolism , CD36 Antigens/analysis , CD36 Antigens/metabolism , Cell Differentiation/drug effects , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Ezetimibe , Flow Cytometry , GPI-Linked Proteins , Humans , Lipopolysaccharide Receptors/analysis , Lipopolysaccharide Receptors/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Microscopy, Fluorescence , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Protein Transport/drug effects , Receptors, IgG/analysis , Receptors, IgG/metabolism
20.
Am J Physiol Endocrinol Metab ; 290(5): E989-97, 2006 May.
Article in English | MEDLINE | ID: mdl-16380389

ABSTRACT

Intramyocellular lipid content (IMCL) serves as a good biomarker of skeletal muscle insulin resistance (IR). However, intracellular fatty acid metabolites [malonyl-CoA, long-chain acyl-CoA (LCACoA)] rather than IMCL are considered to be responsible for IR. This study aimed to investigate dynamics of IMCL and fatty acid metabolites during fed-to-starved-to-refed transition in lean and obese (IR) Zucker diabetic fatty rats in the following different muscle types: soleus (oxidative), extensor digitorum longus (EDL, intermediary), and white tibialis anterior (wTA, glycolytic). In the fed state, IMCL was significantly elevated in obese compared with lean rats in all three muscle types (soleus: 304%, EDL: 333%, wTA: 394%) in the presence of elevated serum triglycerides but similar levels of free fatty acids (FFA), malonyl-CoA, and total LCACoAs. During starvation, IMCL in soleus remained relatively constant, whereas in both rat groups IMCL increased significantly in wTA and EDL after comparable dynamics of starvation-induced FFA availability. The decreases of malonyl-CoA in wTA and EDL during starvation were more pronounced in lean than in obese rats, although there were no changes in soleus muscles for both groups. The concomitant increase in IMCL with the fall of malonyl-CoA support the concept that, as a reaction to starvation-induced FFA availability, muscle will activate lipid oxidation more the lower its oxidative capacity and then store the rest as IMCL.


Subject(s)
Fatty Acids/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Animals , Blood Glucose/metabolism , Body Weight/physiology , Fatty Acids/analysis , Fatty Acids, Nonesterified/blood , Fatty Acids, Unsaturated/analysis , Glucose Clamp Technique , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glycogen Phosphorylase/metabolism , Hexokinase/metabolism , Insulin/blood , Ketone Bodies/blood , Lipids/analysis , Male , Malonyl Coenzyme A/metabolism , Muscle Fibers, Fast-Twitch/chemistry , Muscle Fibers, Fast-Twitch/enzymology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/chemistry , Muscle Fibers, Slow-Twitch/enzymology , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/chemistry , Muscle, Skeletal/enzymology , Rats , Rats, Zucker , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...