Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Clin Pharmacol ; 45(5): 504-13, 2005 May.
Article in English | MEDLINE | ID: mdl-15831773

ABSTRACT

This open-label, randomized, placebo-controlled, incomplete-block, 3-period crossover pilot study investigated the effects of peroxisome proliferator-activated receptor alpha- and gamma-agonists on biomarkers of lipid and glucose metabolism in 12 nondiabetic subjects. Plasma samples were collected before and after each 14-day treatment with placebo, fenofibrate (201 mg/d), rosiglitazone (4 mg twice daily), and combined fenofibrate (201 mg/d) plus rosiglitazone (4 mg twice daily). Except for triglycerides (P < .042) and free fatty acids (P < .074), no significant interaction was demonstrated between fenofibrate and rosiglitazone; thus, the effect due to each drug alone was evaluated (presence/absence of drug). Fenofibrate significantly (P < .050) increased lipoprotein lipase activity (35%) and decreased apolipoproteins B (13%) and C-III (20%). Rosiglitazone significantly (P < .050) decreased fasting glucose (7.3%) and increased apolipoprotein C-III (19%) and adiponectin (137%). Fenofibrate and rosiglitazone also produced effects on triglycerides and free fatty acids, but it was not possible to determine if these effects were synergistic in nature.


Subject(s)
Blood Glucose/metabolism , Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , Lipids/blood , PPAR alpha/agonists , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Adolescent , Adult , Biomarkers/blood , Cross-Over Studies , Drug Interactions , Humans , Male , Pilot Projects , Rosiglitazone
2.
J Clin Invest ; 108(8): 1167-74, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11602624

ABSTRACT

Metformin is a widely used drug for treatment of type 2 diabetes with no defined cellular mechanism of action. Its glucose-lowering effect results from decreased hepatic glucose production and increased glucose utilization. Metformin's beneficial effects on circulating lipids have been linked to reduced fatty liver. AMP-activated protein kinase (AMPK) is a major cellular regulator of lipid and glucose metabolism. Here we report that metformin activates AMPK in hepatocytes; as a result, acetyl-CoA carboxylase (ACC) activity is reduced, fatty acid oxidation is induced, and expression of lipogenic enzymes is suppressed. Activation of AMPK by metformin or an adenosine analogue suppresses expression of SREBP-1, a key lipogenic transcription factor. In metformin-treated rats, hepatic expression of SREBP-1 (and other lipogenic) mRNAs and protein is reduced; activity of the AMPK target, ACC, is also reduced. Using a novel AMPK inhibitor, we find that AMPK activation is required for metformin's inhibitory effect on glucose production by hepatocytes. In isolated rat skeletal muscles, metformin stimulates glucose uptake coincident with AMPK activation. Activation of AMPK provides a unified explanation for the pleiotropic beneficial effects of this drug; these results also suggest that alternative means of modulating AMPK should be useful for the treatment of metabolic disorders.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Multienzyme Complexes/metabolism , Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/pharmacology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Enzyme Activation/drug effects , Fatty Acids/metabolism , Gene Expression/drug effects , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , In Vitro Techniques , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Protein Kinase Inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Sterol Regulatory Element Binding Protein 1
3.
Eur J Pharmacol ; 427(3): 285-93, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11567659

ABSTRACT

Dyslipidemia, a major risk factor for cardiovascular disease, may be directly linked to diabetic hyperglycemia and insulin resistance. An appropriate dyslipidemic animal model that has diabetes would provide an important tool for research on the treatment of diabetic dyslipidemia. Ten days of high fat feeding in golden Syrian hamsters resulted in a significant increase in insulin resistance and baseline serum lipid levels accompanied by a pronounced dyslipidemia. Thirteen days of treatment with fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARalpha) selective agonist, produced a dose-dependent decrease in serum lipid levels. The pattern observed was characterized by lowered very-low-density lipoprotein (VLDL) and low-density lipoprotein (LDL) and raised high-density lipoprotein (HDL) cholesterol in a fashion similar to that seen in man. Diabetic conditions were also significantly improved by fenofibrate with a normalization of impaired glucose tolerance and an improvement of insulin sensitivity during an oral glucose tolerance test. These data suggest that fenofibrate may correct not only the dyslipidemia but also the insulin resistance caused by a high fat diet, and the high fat fed hamster may be a good animal model for research on the treatment of diabetic dyslipidemia with PPARalpha selective agonists.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Dietary Fats/administration & dosage , Fenofibrate/pharmacology , Hyperlipidemias/prevention & control , Hypolipidemic Agents/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Cholesterol, VLDL/blood , Cholesterol, VLDL/drug effects , Cricetinae , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Dose-Response Relationship, Drug , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Hydroxymethylglutaryl CoA Reductases/genetics , Hyperlipidemias/blood , Hyperlipidemias/chemically induced , Insulin/blood , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Mesocricetus , Oxidation-Reduction/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists
4.
Biochem Biophys Res Commun ; 279(2): 330-6, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11118287

ABSTRACT

11beta-hydroxysteroid dehydrogenase type 1 (11betaHSD1) is an enzyme that converts cortisone to the active glucocorticoid, cortisol. Cortisol-cortisone interconversion plays a key role in the regulation of glucose metabolism, since mice deficient in 11betaHSD1 are resistant to diet-induced hyperglycemia. Peroxisome proliferator activator receptors (PPAR) are key regulators of glucose and lipid homeostasis. We observed a striking downregulation of murine hepatic 11betaHSD1 expression and activity after chronic treatment of wild-type mice with PPARalpha agonists, while 11betaHSD1 in the livers of PPARalpha knockout mice, or in mice treated for only 7 h with PPARalpha agonists, was unaltered. Our results are the first to show PPARalpha agonists can affect glucocorticoid metabolism in the liver by altering 11betaHSD1 expression after chronic treatment. Regulation of active glucocorticoid levels in the liver by PPARalpha agonists may in turn affect glucose metabolism, consistent with reports of their antidiabetic effects.


Subject(s)
Fenofibrate/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Hepatocytes/enzymology , Hydroxysteroid Dehydrogenases/genetics , Liver/enzymology , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Cells, Cultured , Cricetinae , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Feedback , Gene Expression Regulation, Enzymologic/drug effects , Mice , Mice, Knockout , Models, Biological , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/agonists , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic/drug effects
5.
Mol Pharmacol ; 58(3): 470-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10953038

ABSTRACT

Peroxisome proliferators are a diverse group of compounds that cause hepatic hypertrophy and hyperplasia, increase peroxisome number, and on chronic high-dose administration, lead to rodent liver tumorigenesis. Various lines of evidence have led to the conclusion that these agents induce their pleiotropic effects exclusively via agonism of peroxisome proliferator-activated receptor (PPAR)alpha, a member of the steroid receptor superfamily involved in the regulation of fatty acid metabolism. Recently, agonists of two other members of this receptor family have been identified. PPARgamma is predominantly expressed in adipocytes where it mediates differentiation; PPARdelta is a widely expressed orphan receptor with yet unresolved physiologic functions. In the course of characterizing newer PPAR ligands, we noted that highly selective PPARgamma agonists or dual PPARgamma/PPARdelta agonists, lacking apparent murine PPARalpha agonist activity, cause peroxisome proliferation in CD-1 mice. We therefore made use of PPARalpha knockout mice to investigate whether these effects resulted from agonism of PPARalpha by these agents at very high dose levels or whether PPARgamma (or PPARdelta) agonism alone can result in peroxisome proliferation. We report here that several parameters linked to the hepatic peroxisome proliferation response in mice that were seen with these agents resulted from PPARalpha-independent effects.


Subject(s)
Peroxisome Proliferators/pharmacology , Peroxisomes/drug effects , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/metabolism , Animals , Female , Gene Expression Regulation , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred ICR , Organ Size/drug effects , Peroxisomes/physiology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/agonists , Transcription Factors/deficiency , Transcription Factors/genetics
6.
J Biol Chem ; 274(10): 6718-25, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10037770

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) include three receptor subtypes encoded by separate genes: PPARalpha, PPARdelta, and PPARgamma. PPARgamma has been implicated as a mediator of adipocyte differentiation and the mechanism by which thiazolidinedione drugs exert in vivo insulin sensitization. Here we characterized novel, non-thiazolidinedione agonists for PPARgamma and PPARdelta that were identified by radioligand binding assays. In transient transactivation assays these ligands were agonists of the receptors to which they bind. Protease protection studies showed that ligand binding produced specific alterations in receptor conformation. Both PPARgamma and PPARdelta directly interacted with a nuclear receptor co-activator (CREB-binding protein) in an agonist-dependent manner. Only the PPARgamma agonists were able to promote differentiation of 3T3-L1 preadipocytes. In diabetic db/db mice all PPARgamma agonists were orally active insulin-sensitizing agents producing reductions of elevated plasma glucose and triglyceride concentrations. In contrast, selective in vivo activation of PPARdelta did not significantly affect these parameters. In vivo PPARalpha activation with WY-14653 resulted in reductions in elevated triglyceride levels with minimal effect on hyperglycemia. We conclude that: 1) synthetic non-thiazolidinediones can serve as ligands of PPARgamma and PPARdelta; 2) ligand-dependent activation of PPARdelta involves an apparent conformational change and association of the receptor ligand binding domain with CREB-binding protein; 3) PPARgamma activation (but not PPARdelta or PPARalpha activation) is sufficient to potentiate preadipocyte differentiation; 4) non-thiazolidinedione PPARgamma agonists improve hyperglycemia and hypertriglyceridemia in vivo; 5) although PPARalpha activation is sufficient to affect triglyceride metabolism, PPARdelta activation does not appear to modulate glucose or triglyceride levels.


Subject(s)
Adipocytes/cytology , Diabetes Mellitus, Experimental/drug therapy , Ligands , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/agonists , Transcription Factors/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Blood Glucose/drug effects , Cell Differentiation/drug effects , Cell Line , Diabetes Mellitus, Experimental/blood , Humans , Mice , Protein Conformation , Receptors, Cytoplasmic and Nuclear/chemistry , Transcription Factors/chemistry
7.
Endocrinology ; 139(12): 4920-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9832429

ABSTRACT

A role for peroxisome proliferator-activated receptors, PPAR gamma and PPAR alpha, as regulators of energy homeostasis and lipid metabolism, has been suggested. Recently, three distinct uncoupling protein isoforms, UCP-1, UCP-2, and UCP-3, have also been identified and implicated as mediators of thermogenesis. Here, we examined whether in vivo PPAR gamma or PPAR alpha activation regulates the expression of all three UCP isoforms. Rats or lean and db/db mice were treated with PPAR gamma [thiazolidinedione (TZD)] or PPAR alpha (WY-14643) agonists, followed by measurement of messenger RNAs (mRNAs) for UCP-1, UCP-2, and UCP-3 in selected tissues where they are expressed. TZD treatment (AD 5075 at 5 mg/kg x day) of rats (14 days) increased brown adipose tissue (BAT) depot size and induced the expression of each UCP mRNA (3x control levels for UCP-1 and UCP-2, 2.5x control for UCP-3). In contrast, UCP-2 and UCP-3 mRNA levels were not affected in white adipose tissue or skeletal muscle. Chronic (30 days) low-dose (0.3 mg/kg x day) TZD treatment induced UCP-1 mRNA and protein in BAT (2.5x control). In contrast, chronic TZD treatment (30 mg/kg x day) suppressed UCP-1 mRNA (>80%) and protein (50%) expression in BAT. This was associated with further induction of UCP-2 expression (>10-fold) and an increase in the size of lipid vacuoles, a decrease in the number of lipid vacuoles in each adipocyte, and an increase in the size of the adipocytes. TZD treatment of db/db mice (BRL 49653 at 10 mg/kg x day for 10 days) also induced UCP-1 and UCP-3 (but not UCP-2) expression in BAT. PPAR alpha is present in BAT, as well as liver. Treatment of rats or db/db mice with WY-14643 did not affect expression of UCP-1, -2, or -3 in BAT. Hepatic UCP-2 mRNA was increased (4x control level) in db/db and lean mice, although this effect was not observed in rats. Thus, in vivo PPAR gamma activation can induce expression of UCP-1, -2, and -3 in BAT; whereas chronic-intense PPAR gamma activation may cause BAT to assume white adipose tissue-like phenotype with increased UCP-2 levels. PPAR alpha activation in mice is sufficient to induce liver UCP-2 expression.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation/physiology , Membrane Proteins/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Thiazolidinediones , Transcription Factors/physiology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Carrier Proteins/metabolism , Dose-Response Relationship, Drug , Ion Channels , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Thiazoles/pharmacology , Time Factors , Uncoupling Protein 1
8.
Endocrinology ; 139(12): 5034-41, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9832442

ABSTRACT

Thiazolidinedione (TZD) insulin sensitizers are specific agonists of peroxisome proliferator activated receptor (PPAR)gamma. However, their mechanism of action and the in vivo target tissue(s) that mediate insulin sensitization remain poorly defined. Although PPARgamma messenger RNA expression has been reported in skeletal muscle, the expression of PPARgamma within myocytes in intact muscle tissue has not been examined. An antipeptide PPARgamma antibody was generated; immunohistochemistry was then used to demonstrate that PPARgamma is present within nuclei of myocytes [in both skeletal (white and red fibers) and cardiac tissue (rodent and human)]. The effect of insulin sensitizer treatment on muscle insulin action was studied using ob/ob mice after 4 days dosing with a potent (6 nM PPARgamma Kd) TZD (10 mg/kg x day). 2-deoxyglucose (2-DOG) uptake was then assessed in freshly isolated soleus muscles from lean vs. ob/ob vs. TZD-treated ob/ob mice. In lean mouse muscles, 2-DOG uptake was stimulated by 82%, 95%, 165% (with 25, 100, 2000 microU/ml insulin); muscles from ob/ob were severely insulin resistant (<80% stimulation with 2000 microU/ml insulin). Muscles from TZD-treated ob/ob displayed a normal insulin response with 100 (71%) or 2000 (158%) microU/ml insulin. Additional studies were performed using ZDF rats treated with/without TZD for 7 days. In vivo 2-DOG glucose uptake into soleus, gastrocnemius, and diaphragm muscles was measured during euglycemic-hyperinsulinemic clamp. Compared with lean rats, muscle 2-DOG uptake in ZDF was reduced by 52% (soleus) or 71% (diaphragm). Partial (40-60%) normalization of the reduced 2-DOG uptake was evident in TZD-treated ZDF rats. In contrast to the effect of in vivo treatment on muscle insulin action, preincubation of isolated soleus muscles from naive lean or ob/ob mice for 5 h with 100 nM TZD did not affect insulin-stimulated 2-DOG uptake. We conclude: 1) PPARgamma is expressed in myocytes within skeletal and cardiac muscle. 2) In vivo activation of PPARgamma by treatment of insulin-resistant mice/rats with a potent TZD corrects impaired muscle insulin action. 3) The lack of a direct effect on muscle after 5 h in vitro TZD incubation suggests that changes in insulin action may require a longer duration of PPARgamma activation or that improved muscle insulin sensitivity may result from an indirect in vivo effect of PPARgamma activation (e.g. changes in systemic lipid metabolism).


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Muscle, Skeletal/physiology , Receptors, Cytoplasmic and Nuclear/agonists , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/agonists , Animals , Deoxyglucose/pharmacokinetics , Humans , In Vitro Techniques , Insulin/pharmacology , Male , Mice/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myocardium/metabolism , Obesity/genetics , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism
9.
Diabetes ; 46(9): 1526-31, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9287059

ABSTRACT

To address the hypothesis that tumor necrosis factor (TNF)-alpha has a role in obesity-associated insulin resistance or the regulation of in vivo lipid metabolism, mice with targeted disruption of the TNF-alpha gene were generated and studied. The absence of TNF-alpha protein in TNF-null (-/-) mice was confirmed. Lean or obese (gold-thioglucose [GTG]-injected) homozygous (-/-) mice were compared with lean or obese age- and sex-matched wild-type (+/+) mice derived from the same line at 13, 19, and 28 weeks of age. The following parameters were significantly affected in lean -/- versus +/+ mice: Body weight was not affected until week 28 (decreased by 14%); epididymal fat pad weight also decreased (25%) at this time, as did percentage body fat (16%), while percentage body protein was increased 13%. Fed plasma insulin levels decreased 47% (28 weeks), triglyceride levels decreased (all three ages; maximum 35% at 19 weeks), and fed plasma leptin decreased 33% (28 weeks). Fasting glucose was slightly (10%) reduced, but the glucose response to an oral glucose tolerance test (OGTT) was not affected. There was a trend (NS) toward increased total adipose tissue lipoprotein lipase in -/- versus +/+ mice. GTG-treatment resulted in obese -/- and +/+ mice with equal mean body weights (42 and 58% increased weight versus lean mice). The following parameters were significantly different in obese -/- mice: fasting plasma glucose decreased 13% (28 weeks), fed plasma insulin decreased 67% (28 weeks), and insulin response to OGTT was decreased by 50%. For both groups of obese mice, glucose levels during the OGTT were substantially increased compared with those in lean mice; however, mean stimulated glucose levels were 20% lower in obese -/- versus +/+ mice. We conclude 1) that TNF-alpha functions to regulate plasma triglycerides and body adiposity and 2) that although TNF-alpha contributes to reduced insulin sensitivity in older or obese mice, the absence of TNF-alpha is not sufficient to substantially protect against insulin resistance in the GTG hyperphagic model of rodent obesity.


Subject(s)
Obesity/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Aurothioglucose/pharmacology , Hypothalamus/physiopathology , Insulin/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional , Obesity/metabolism , Triglycerides/blood
10.
Endocrinology ; 137(10): 4189-95, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8828476

ABSTRACT

The thiazolidinediones are novel insulin sensitizers that serve as orally active antidiabetic agents, in rodents, nonhuman primates, and man. We have examined the effects of 4-week oral administration of three thiazolidinediones (AD-5075, BRL 49653, and CS-045) on plasma glucose and triglyceride concentrations in obese hyperglycemic db/db mice. All three agents lower plasma glucose and triglyceride concentrations. Normal levels of glucose are achieved after treatment with AD-5075 (> 1.7 mg/kg) or BRL 49653 (> or = 30 mg/kg), whereas CS-045 (100 or 300 mg/kg) produces only modest reductions in either parameter. Although the thiazolidinediones have demonstrated insulin-sensitizing activities both in vivo and in vitro, their primary molecular target has been unclear. We have compared the in vivo antidiabetic actions described above with the in vitro activities on peroxisomal proliferator-activated receptor-gamma (PPAR gamma). Hamster PPAR gamma 1 was transiently expressed in COS-1 cells to study the binding of [3H]AD-5075. The concentrations of compounds needed to displace radiolabeled AD-5075 from PPAR gamma correlate with their in vivo potency; the Ki values for displacement by cold AD-5075, BRL 49653, and CS-045 are 22, 68, and 1600 nM, respectively. To examine activation of the receptor, it was transiently cotransfected into COS-1 cells with a reporter plasmid containing two copies of a peroxisome proliferator response element. The EC50 values for activation are 2, 6, and 140 nM for AD-5075, BRL 49653, and CS-045, respectively. We have also analyzed limited proteolytic digests of in vitro translated hamster PPAR gamma. The thiazolidinediones produce a conformational change in PPAR gamma analogous to those produced by agonists of other nuclear hormone receptors. In the presence of saturating concentrations of either AD-5075 or BRL 49653, a receptor fragment of 27 kDa is protected from proteolysis by trypsin. These data support the conclusion that the antidiabetic actions of the thiazolidinediones are directly mediated through binding to PPAR gamma and the resulting active conformation of the receptor. Therefore, binding and transactivation assays using PPAR gamma should serve to identify other novel therapeutic agents with potential antidiabetic activities.


Subject(s)
Diabetes Mellitus/drug therapy , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/drug effects , Thiazoles/therapeutic use , Transcription Factors/chemistry , Transcription Factors/drug effects , Animals , Blood Glucose/analysis , COS Cells , Cricetinae , Diabetes Mellitus/blood , Diabetes Mellitus/genetics , Hyperglycemia/genetics , Hypertriglyceridemia/genetics , Insulin Resistance/genetics , Male , Mice/genetics , Molecular Conformation , Peptide Hydrolases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Triglycerides/blood
11.
Endocrinology ; 137(5): 1984-90, 1996 May.
Article in English | MEDLINE | ID: mdl-8612539

ABSTRACT

Thiazolidinedione derivatives are a novel class of insulin-sensitizing agents that have demonstrated effective antidiabetic activity in vivo. Here, the effects of the potent thiazolidinedione derivative, AD-5075, on the rate-limiting enzyme of glycogen synthesis, glycogen synthase, were investigated in cultured rat adipose tissue. Short term preincubation of adipose tissue with AD-5075 potentiated acute insulin stimulation of I-form glycogen synthase activity in a concentration-dependent (EC50 approximately, 61nM) and time-dependent (t1/2 approximately, 2.3 h) manner. The thiazolidinedione derivative increased the responsiveness of I-form glycogen synthase activity to insulin stimulation at both maximal and submaximal insulin concentrations. In contrast, it had no effect on total glycogen synthase activity. Isoproterenol inhibited acute insulin activation of I-form glycogen synthase activity in a dose-dependent manner; maximal inhibition was attained at a concentration of 3 nM. AD-5075 antagonized isoproterenol inhibition of insulin's action. The concentration of glycogenolytic agent required to attain maximal inhibition was increased an order of magnitude in tissue treated with the antidiabetic agent. Short term preincubation of adipose tissue under hyperglycemic conditions (15 or 25 mM glucose) decreased insulin-stimulated I-form glycogen synthase activity. Concurrent treatment of the tissue with AD-5075 abrogated this glucose toxicity-induced inhibition of insulin action. Wortmannin, an inhibitor of phosphatidylinositol 3-kinase, blocked insulin activation of glycogen synthase in a dose-dependent manner. Half-maximal inhibition was observed at approximately 0.3 microM, and maximal inhibition occurred at 1.0 microM. AD-5075 did not antagonize wortmannin's inhibitory action. These results indicate that thiazolidinediones can act directly on adipose tissues to augment an important metabolic effect of insulin and counteract the inhibitory effects of catecholamines or hyperglycemia. As insulin stimulation of glycogen synthase remains wortmannin inhibitable in the presence of AD-5075, the effects of thiazolidinediones on insulin signal transduction may be phosphatidylinositol 3-kinase-dependent.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/enzymology , Glycogen Synthase/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Thiazoles/pharmacology , Thiazolidinediones , Animals , Catecholamines/pharmacology , Drug Synergism , Glucose/pharmacology , Insulin Resistance , Kinetics , Male , Rats , Rats, Wistar
12.
J Biol Chem ; 271(16): 9455-9, 1996 Apr 19.
Article in English | MEDLINE | ID: mdl-8621615

ABSTRACT

Obese (ob) is a recently identified gene involved in the regulation of energy balance in the mouse. We report here that AD-5075, a potent thiazolidinedione which lowered plasma glucose and triglyceride in Zucker diabetic fatty (ZDF) rats and db/db mice, decreased the expression of the ob gene in these animal models of obesity and non-insulin-dependent diabetes mellitus. The level of adipose ob mRNA in ZDF rats was 3-fold greater than that detected in the Zucker lean littermates. Chronic treatment with AD-5075 elicited a 67 and 70% reduction of ob mRNA in ZDF and control lean rats, respectively. Furthermore, the amount of adipose ob mRNA in db/db mice was 7 times higher than that detected in lean littermates. Treatment of db/db mice with AD-5075 resulted in a 78% reduction of the level of ob mRNA with parallel changes in circulating level of the ob gene product, leptin. The reduction of the ob mRNA in the Zucker lean rats was accompanied by significantly greater food intake and weight gain. However, in ZDF rats and db/db mice, there was profound increase in body weight without hyperphagia. The results demonstrate that the expression of the ob gene is up-regulated in these two rodent models of diabetes compared to their lean counterparts and that such overexpression is attenuated by treatment with an agent that improves insulin sensitivity and glucose homeostasis in vivo.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus/genetics , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Obesity , Protein Biosynthesis , Proteins/genetics , Thiazoles/pharmacology , Thiazolidinediones , Adipose Tissue/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blood Glucose/metabolism , DNA Primers , DNA Probes , DNA, Complementary , Diabetes Mellitus/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Homeostasis/drug effects , Leptin , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Zucker , Reference Values , Triglycerides/blood
13.
J Clin Invest ; 89(6): 1885-91, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1601995

ABSTRACT

The oxidative modification of low density lipoprotein (LDL) may play an important role in atherosclerosis. We found that the antioxidant N,N'-diphenyl-1,4-phenylenediamine (DPPD) inhibits in vitro LDL oxidation at concentrations much lower than other reported antioxidants. To test whether DPPD could prevent atherosclerosis, New Zealand White rabbits were fed either a diet containing 0.5% cholesterol and 10% corn oil (control group) or the same diet also containing 1% DPPD (DPPD-fed group) for 10 wk. Plasma total cholesterol levels were not different between the two groups, but DPPD feeding increased the levels of triglyceride (73%, P = 0.007) and HDL cholesterol (26%, P = 0.045). Lipoproteins from DPPD-fed rabbits contained DPPD and were much more resistant to oxidation than control lipoproteins. After 10 wk, the DPPD-fed animals had less severe atherosclerosis than did the control animals: thoracic aorta lesion area was decreased by 71% (P = 0.0007), and aortic cholesterol content was decreased by 51% (P = 0.007). Although DPPD cannot be given to humans because it is a mutagen, our results indicate that orally active antioxidants can have antiatherosclerotic activity. This strongly supports the theory that oxidized LDL plays an important role in the pathogenesis of atherosclerosis.


Subject(s)
Antioxidants/pharmacology , Arteriosclerosis/prevention & control , Cholesterol/metabolism , Lipoproteins, LDL/metabolism , Phenylenediamines/pharmacology , Administration, Oral , Animals , Antioxidants/chemistry , Arteriosclerosis/metabolism , Cholesterol/administration & dosage , Cholesterol/blood , Humans , Kinetics , Male , Oxidation-Reduction , Phenylenediamines/chemistry , Rabbits , Structure-Activity Relationship
14.
Lipids ; 26(12): 997-1003, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1819756

ABSTRACT

Addition of platelet-activating factor (PAF; 1-O-alkyl-2-acetyl-sn-glycero-3-phosphocholine) to leukocyte-rich plasma from several species resulted in the rapid and pronounced activation of the PAF biosynthetic enzyme acetyl-CoA:1-O-alkyl-sn-glycero-3-phosphocholine acetyltransferase (EC 2.3.1.67). Activation of acetyltransferase by PAF occurred in leukocyte-rich plasma from human, chimpanzee, rhesus monkey, and dog. The neutrophil was indicated to be the major cellular source of the activatable acetyltransferase in leukocyte-rich plasma. The induction of acetyltransferase was substantial with 10 nM PAF, and maximal at 10-30 seconds. Measurable acetyltransferase activation was significantly greater when the PAF-activated cells were separated from the plasma by centrifugation before the acetyltransferase assay. This may be due in part to the removal of the PAF-specific acetylhydrolase present in plasma which can cleave the acetyl group from PAF. Measuring PAF activation of acetyltransferase in leukocyte-rich plasma can be useful to determine the potency of PAF antagonists with neutrophils in plasma compared to isolated neutrophils in aqueous buffer, and as an ex vivo assay to determine the efficacy and plasma concentration equivalents of antagonists administered to whole animals. The PAF antagonist L-659,989 was shown to be 3-5 times more potent in inhibiting PAF induction of acetyltransferase in isolated human neutrophils than in human leukocyte-rich plasma, with IC50 values of 10 nM and 40 nM, respectively. In the ex vivo assay, oral administration of the PAF antagonist L-667,131 to dogs resulted in very substantial inhibition of PAF induction of acetyltransferase in the leukocyte-rich plasma.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Acetyltransferases/blood , Leukocytes/enzymology , Neutrophils/enzymology , Platelet Activating Factor/pharmacology , Acetyltransferases/antagonists & inhibitors , Animals , Enzyme Activation , Furans/pharmacology , Humans , Kinetics , Macaca mulatta , Pan troglodytes , Platelet Activating Factor/antagonists & inhibitors , Rats , Reference Values
15.
Biochem Biophys Res Commun ; 150(3): 1213-20, 1988 Feb 15.
Article in English | MEDLINE | ID: mdl-2829894

ABSTRACT

The title compound, L-659,989, is a highly potent, competitive, and selective antagonist of the binding of [3H]PAF to its receptors in platelet membranes from rabbits and humans. It exhibits equilibrium inhibition constants for PAF binding of 1.1 nM (rabbit) to 9.0 nM (human), values that are at least 1-2 orders of magnitude lower than those of other PAF antagonists tested. L-659,989 potently inhibits PAF-induced aggregation of rabbit platelets and degranulation of rat (ED50 4.5 nM) and human (ED50 10 nM) neutrophils. L-659,989 inhibits PAF-induced extravasation and lysosomal enzyme release in rats, and is active orally in female rats (ED50 0.2 mg/kg) with an extraordinary oral duration of action of 12 to 16 hours at 1.0 mg/kg p.o.


Subject(s)
Furans/pharmacology , Platelet Activating Factor/antagonists & inhibitors , Platelet Membrane Glycoproteins , Receptors, Cell Surface/drug effects , Receptors, G-Protein-Coupled , Acetylglucosaminidase/blood , Animals , Binding, Competitive , Blood Platelets/drug effects , Blood Platelets/metabolism , Cattle , Cell Membrane/metabolism , Cytoplasmic Granules/drug effects , Female , Guinea Pigs , Hematocrit , Humans , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/ultrastructure , Platelet Aggregation , Platelet Aggregation Inhibitors , Rabbits , Rats , Receptors, Cell Surface/metabolism
16.
Proc Natl Acad Sci U S A ; 84(21): 7557-61, 1987 Nov.
Article in English | MEDLINE | ID: mdl-3478712

ABSTRACT

Platelet activating factor (1-alkyl-2-acetyl-sn-glycero-3-phosphocholine; PAF) induced in isolated rat peritoneal and human peripheral neutrophils a rapid and potent activation of the PAF biosynthetic enzyme acetyl-CoA:1-alkyl-sn-glycero-3-phosphocholine O2-acetyltransferase (EC 2.3.1.67). The PAF-induced activation of the neutrophil acetyltransferase (8-10 times basal neutrophil activity) was maximal within 30 sec after PAF addition, as was the PAF-stimulated degranulation. After 1 min of PAF stimulation, the elevated acetyltransferase activity steadily decreased. Within 2 min of stimulation of neutrophils with 10(-6) M PAF, the 7-fold increase in acetyltransferase activity was coincident with substantial PAF synthesis (as measured by [3H]acetate incorporation into PAF), which was 14% of the PAF synthesis induced by the Ca2+ ionophore A23187 at 10(-5) M. PAF activation of the acetyltransferase and PAF synthesis required intact neutrophils as they did not occur in cells broken by sonication. The neutrophil acetyltransferase was 10-30 times more sensitive to activation by PAF than was degranulation as the acetyltransferase activation was evident with 10(-9) M PAF and was about maximal with 3 x 10(-8) M PAF. The unstimulated and PAF-induced acetyltransferase exhibited the same Km for acetyl-CoA (67 microM), but the Vmax for the PAF-induced enzyme (1667 pmol/min per 10(7) cells) was 10 times that of the unstimulated enzyme (175 pmol/min per 10(7) cells). The PAF induction of the acetyltransferase was less sensitive to inhibition by the specific PAF receptor antagonist L-652,731 than was PAF-induced degranulation. This, along with the differing sensitivities to PAF, suggests that acetyltransferase activation and degranulation induced by PAF either involve two different PAF receptors or involve one receptor type with different receptor occupancy requirements. Escherichia coli alkaline phosphatase, which greatly decreased the activity of the acetyltransferase in spleen microsomes, had little or no effect on the basal or PAF-induced neutrophil acetyltransferase. Thus, by stimulating the activity of acetyltransferase, PAF induces in neutrophils the synthesis of more PAF, thereby probably augmenting the neutrophil response to the initial PAF.


Subject(s)
Acetyltransferases/blood , Neutrophils/enzymology , Platelet Activating Factor/biosynthesis , Animals , Enzyme Activation , In Vitro Techniques , Kinetics , Microsomes/enzymology , Platelet Activating Factor/pharmacology , Rats , Spleen/enzymology
17.
J Pharmacol Exp Ther ; 239(3): 841-5, 1986 Dec.
Article in English | MEDLINE | ID: mdl-3795043

ABSTRACT

Trans-2,5-bis-(3,4,5-trimethoxyphenyl)tetrahydrofuran (L652, 731), a potent and specific receptor antagonist of platelet activating factor (PAF) (Hwang et al., J. Biol. Chem. 260: 15639-15645, 1985), potently inhibits several PAF-induced in vivo responses in rats. Intravenously and p.o. administered L-652,731 gave a dose-response inhibition of PAF-induced lysosomal hydrolase secretion and extravasation with ED50 values of 1 and 3 mg/kg, respectively. Inhibitions of 87% were achieved with 50 mg/kg p.o. After a single 5-mg/kg p.o. dose, L-652,731 achieved 50 to 60% inhibition of PAF-induced lysosomal hydrolase secretion and extravasation by 0.5 to 1.5 hr with near maximum inhibition lasting through 6 hr. A 20-mg/kg p.o. dose of L-652,731 inhibited PAF-induced leukopenia and neutropenia by 96 and 73%, respectively. The most substantial inhibitions of the extravasation and lysosomal hydrolase secretion induced by PAF or soluble immune complexes were achieved by p.o. L-652,731 (20 mg/kg) with moderate inhibition by dexamethasone and little or no inhibition by antagonists/inhibitors of histamine H1 or H2 or serotonin receptors or cyclooxygenase. Intravenous infusion of a 0.4 mg of L-652,731 bolus inhibited the hypotensive responses from subsequent PAF infusions by a maximum of 72% and with a half-life duration of action of 2.5 hr. Intravenous infusion of L-652,731 results in an immediate 87% reversal of the extreme hypotension induced by a previous endotoxin injection. Thus, with its good p.o. activity, long duration of action and specificity in inhibiting PAF-induced responses in vivo, L-652,731 is a very useful tool in determining the role of PAF in mediating different pathophysiological processes.


Subject(s)
Furans/pharmacology , Platelet Activating Factor/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Animals , Blood Pressure/drug effects , Endotoxins/antagonists & inhibitors , Female , Hydrolases/metabolism , Leukopenia/chemically induced , Leukopenia/prevention & control , Lysosomes/enzymology , Neutropenia/chemically induced , Neutropenia/prevention & control , Rats , Rats, Inbred Strains
18.
J Med Chem ; 29(10): 1917-21, 1986 Oct.
Article in English | MEDLINE | ID: mdl-3020247

ABSTRACT

The six (racemic or meso) isomers of 3,4-dimethyl-2,5-bis(3,4-dimethoxyphenyl)tetrahydrofuran and four corresponding desmethyl analogues were prepared and assayed as inhibitors of platelet activating factor (PAF) receptor binding to rabbit platelet plasma membranes. The inhibition by these isomers is stereodependent and varies with the gross shape of the molecules as determined by the molecular mechanics program MM2. The most potent PAF antagonist in this group of compounds is trans-2,5-bis(3,4,5-trimethoxyphenyl)tetrahydrofuran (L-652,731, 14) with an IC50 of 0.02 microM.


Subject(s)
Furans/pharmacology , Plant Extracts/pharmacology , Platelet Activating Factor/antagonists & inhibitors , Animals , Furans/chemical synthesis , Lignans , Molecular Conformation , Plant Extracts/chemical synthesis , Rabbits , Structure-Activity Relationship
19.
J Immunol ; 136(12): 4659-68, 1986 Jun 15.
Article in English | MEDLINE | ID: mdl-3011900

ABSTRACT

A new synthetic compound, L-652,731 (trans-2,5-(3,4,5-trimethoxyphenyl) tetrahydrofuran), which has been demonstrated by Hwang et al. to be a potent and specific platelet-activating factor (PAF) receptor antagonist causes 100% inhibition of 1 microM PAF-induced neutrophil degranulation at 50 microM, but has no effect on neutrophil degranulation induced by precipitating immune complexes (323 micrograms/ml), fMet-Leu-Phe (10(-7) M), or the calcium ionophore A23187 (10(-5) M). Intravenous infusion of 1 mumol L-652,731 results in almost 100% inhibition of hypotension induced by PAF but not that induced by isoproterenol, histamine, bradykinin, or acetylcholine. With the use of this novel PAF receptor antagonist, the in vivo mediator role of PAF in the soluble immune complex-induced hypotension, extravasation, vascular lysosomal hydrolase secretion, and neutropenia in rats was determined. The hypotension, extravasation, and lysosomal hydrolase release induced by immune complex infusion take 2 to 10 min longer to occur than the same responses elicited by PAF infusion. The neutropenia response is immediate with both stimuli. L-652,731 when orally administered to rats (20 mg/kg, 1.5 hr before PAF infusion) inhibited PAF-induced hypotension (69%), extravasation (76%), vascular lysosomal hydrolase release (79%), and neutropenia (73%). The same L-652,731-dosing regimen inhibited immune complex-stimulated hypotension (87%), extravasation (77%), and vascular lysosomal hydrolase release (31%). The initial and complete neutropenia induced by immune complex infusion was not inhibited in L-652,731-pretreated rats, but the rate of return of neutrophils to the blood was faster in the latter rats. Rats with blocked circulation to the liver still exhibited extensive extravasation and vascular lysosomal hydrolase release in response to PAF, but there was no extravasation and greatly reduced hydrolase release in response to immune complexes. Thus PAF is indicated to be a major mediator of soluble immune complex-induced hypotension and vascular permeability and a minor mediator of immune complex-induced lysosomal hydrolase release in rats. PAF probably does not mediate the initial and complete neutropenia stimulated by immune complexes. The liver is probably the major site for PAF production in response to circulating immune complexes.


Subject(s)
Anaphylaxis/immunology , Antigen-Antibody Complex/physiology , Furans/pharmacology , Platelet Activating Factor/physiology , Platelet Membrane Glycoproteins , Receptors, Cell Surface/drug effects , Receptors, G-Protein-Coupled , Anaphylaxis/enzymology , Anaphylaxis/physiopathology , Animals , Antigen-Antibody Complex/analysis , Capillary Permeability/drug effects , Female , Hydrolases/metabolism , Hypotension/physiopathology , Kinetics , Liver/physiopathology , Lysosomes/enzymology , Neutropenia/physiopathology , Platelet Activating Factor/metabolism , Rats , Rats, Inbred Strains
20.
Biochem Med ; 34(2): 241-6, 1985 Oct.
Article in English | MEDLINE | ID: mdl-4084247

ABSTRACT

Human placental beta-glucocerebrosidase modified by covalent attachment of N2-(N2, N6-bis [3-(alpha-D-mannopyranosylthio)propionyl]-L- lysyl)-N6-[3-(alpha-D-mannopyranosylthio)propionyl]-L-lysine was administered to rats by intravenous injection. Comparison of enzyme distribution in isolated liver cell populations indicates an increase in enzyme-specific activity of 18-fold in nonparenchymal cells and only 1.5-fold to hepatocytes compared to uninjected control animals. This macrophage-specific delivery of an active lysosomal enzyme has potential for application in enzyme replacement trials.


Subject(s)
Glucosidases/administration & dosage , Glucosylceramidase/administration & dosage , Kupffer Cells/metabolism , Liver/metabolism , Placenta/enzymology , Animals , Glucosylceramidase/metabolism , Glycopeptides/administration & dosage , Humans , Injections, Intravenous , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...