Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Metabolism ; 59(6): 914-20, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20197197

ABSTRACT

Pioglitazone is prescribed to improve insulin sensitivity in type 2 diabetes mellitus patients and has been discussed as a therapy for metabolic syndrome. Pioglitazone and other thiazolidinediones are associated with fluid retention and edema that may exacerbate existing or developing congestive heart failure, which is often present in these patients. Using a nonhuman primate model, our aims were to evaluate (1) whether fluid shifts were detectable in normoglycemic monkeys, (2) which fluid compartment changed, and (3) whether fluid retention was dose dependent. Seventeen adult male cynomolgus macaques (Macaca fascicularis) were studied in a Latin square design such that all animals received 0, 1, 2, and 5 mg/kg pioglitazone for 6 weeks with 2 weeks of washout between dosing intervals. Doses approximated human exposures achieved with 30, 45, and 60 mg. At the end of each period, animals were weighed and underwent dual-absorption x-ray absorption scanning for body composition measurements. Fluid volumes were quantitated by Evans blue dilution for plasma volume, equilibration of sodium bromide for extracellular water, and deuterated water for total body water. Significant (P < .05) effects were seen with expansion of PV at both the 2- and 5-mg/kg doses, along with reduced plasma sodium at 5 mg/kg; however, surrogate end points used to indicate fluid retention (body weight, hematocrit, total protein, and albumin) did not change significantly. Significant trends toward increases in interstitial fluid and extracellular water with increasing dose were apparent. Pioglitazone effectively improved metabolic status by significantly decreasing fasting glucose and triglycerides and increasing adiponectin. We conclude that thiazolidinedione-related plasma volume expansion occurs in nondiabetic primates and that fluid retention is detectable when compartments are directly measured.


Subject(s)
Fluid Shifts/physiology , Hypoglycemic Agents/therapeutic use , Obesity/drug therapy , Obesity/physiopathology , PPAR gamma/agonists , Prediabetic State/drug therapy , Prediabetic State/physiopathology , Thiazolidinediones/therapeutic use , Absorptiometry, Photon , Animals , Blood Glucose/metabolism , Body Composition/drug effects , Body Composition/physiology , Body Water/physiology , Dose-Response Relationship, Drug , Endpoint Determination , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Hypoglycemic Agents/blood , Macaca fascicularis , Male , Pioglitazone , Thiazolidinediones/blood
2.
Am J Physiol Renal Physiol ; 297(1): F55-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19403648

ABSTRACT

PPARgamma agonists are synthetic ligands for the peroxisome proliferator-activated receptor-gamma (PPARgamma). These agents have insulin-sensitizing properties but can cause fluid retention, thereby limiting their usefulness in patients at risk for cardiovascular disease. The side effect etiology is unknown, but the nature of presentation suggests modulation of renal salt and water homeostasis. In a well-characterized cell culture model of the principal cell type [Madin-Darby canine kidney (MDCK)-C7], PPARgamma agonists inhibit vasopressin-stimulated Cl(-) secretion with agonist dose-response relationships that mirror receptor transactivation profiles. Analyses of the components of the vasopressin-stimulated intracellular signaling pathway indicated no PPARgamma agonist-induced changes in basolateral membrane conductances, intracellular cAMP, protein kinase A, or total cellular adenine nucleotides. The PPARgamma agonist-induced decrease in anion secretion is the result of decreased mRNA of the final effector in the pathway, the apically located cystic fibrosis transmembrane regulator (CFTR). These data showing that CFTR is a target for PPARgamma agonists may provide new insights into the physiology of PPARgamma agonist-induced fluid retention.


Subject(s)
Chlorides/metabolism , Kidney/drug effects , Kidney/metabolism , PPAR gamma/agonists , Vasopressins/pharmacology , Animals , Biological Transport/drug effects , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dogs , Dose-Response Relationship, Drug , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/metabolism , Kidney/cytology , Ligands , Models, Animal , Oxazoles/pharmacology , PPAR gamma/metabolism , Pioglitazone , RNA, Messenger/metabolism , Signal Transduction/drug effects , Thiazolidinediones/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
3.
Clin Cancer Res ; 15(1): 217-25, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19118049

ABSTRACT

PURPOSE: Insulin-like growth factor-I receptor and phosphoinositide 3-kinase/AKT/mammalian target of rapamycin pathways are among the most active areas of drug discovery in cancer research. However, due to their integral roles in insulin signaling, inhibitors targeting these pathways often lead to hyperglycemia and hyperinsulinemia. We investigated the mechanism of hyperglycemia induced by GSK690693, a pan-AKT kinase inhibitor in clinical development, as well as methods to ameliorate these side effects. EXPERIMENTAL DESIGN: The effect of GSK690693 on blood glucose, insulin, and glucagon levels was characterized in mice. We then evaluated the effects of commonly prescribed antidiabetic agents on GSK690693-induced hyperglycemia. The mechanism of blood glucose increase was evaluated using fasting and tracer uptake studies and by measuring liver glycogen levels. Finally, approaches to manage AKT inhibitor-induced hyperglycemia were designed using fasting and low carbohydrate diet. RESULTS: We report that treatment with antidiabetic agents does not significantly affect GSK690693-induced hyperglycemia in rodents. However, administration of GSK690693 in mice significantly reduces liver glycogen (approximately 90%), suggesting that GSK690693 may inhibit glycogen synthesis and/or activate glycogenolysis. Consistent with this observation, fasting before drug administration reduces baseline liver glycogen levels and attenuates hyperglycemia. Further, GSK690693 also inhibits peripheral glucose uptake and introduction of a low-carbohydrate (7%) or 0% carbohydrate diet after GSK690693 administration effectively reduces diet-induced hyperglycemia in mice. CONCLUSIONS: The mechanism of GSK690693-induced hyperglycemia is related to peripheral insulin resistance, increased gluconeogenesis, and/or hepatic glycogenolysis. A combination of fasting and low carbohydrate diet can reduce the magnitude of hyperglycemia induced by an AKT inhibitor.


Subject(s)
Hyperglycemia/chemically induced , Oxadiazoles/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Diet, Carbohydrate-Restricted , Fasting , Female , Hyperglycemia/prevention & control , Liver Glycogen/metabolism , Male , Mice , Mice, SCID , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
4.
Environ Health Perspect ; 116(11): 1456-62, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19057696

ABSTRACT

BACKGROUND: Developmental exposures to organophosphate pesticides are virtually ubiquitous. These agents are neurotoxicants, but recent evidence also points to lasting effects on metabolism. OBJECTIVES: We administered parathion to neonatal rats. In adulthood, we assessed the impact on weight gain, food consumption, and glucose and lipid homeostasis, as well as the interaction with the effects of a high-fat diet. METHODS: Neonatal rats were given parathion on postnatal days 1-4 using doses (0.1 or 0.2 mg/kg/day) that straddle the threshold for barely detectable cholinesterase inhibition and the first signs of systemic toxicity. In adulthood, animals were either maintained on standard lab chow or switched to a high-fat diet for 7 weeks. RESULTS: In male rats on a normal diet, the low-dose parathion exposure caused increased weight gain but also evoked signs of a prediabetic state, with elevated fasting serum glucose and impaired fat metabolism. The higher dose of parathion reversed the weight gain and caused further metabolic defects. Females showed greater sensitivity to metabolic disruption, with weight loss at either parathion dose, and greater imbalances in glucose and lipid metabolism. At 0.1 mg/kg/day parathion, females showed enhanced weight gain on the high-fat diet; This effect was reversed in the 0.2-mg/kg/day parathion group, and was accompanied by even greater deficits in glucose and fat metabolism. CONCLUSIONS: Neonatal low-dose parathion exposure disrupts glucose and fat homeostasis in a persistent and sex-selective manner. Early-life toxicant exposure to organophosphates or other environmental chemicals may play a role in the increased incidence of obesity and diabetes.


Subject(s)
Dietary Fats/administration & dosage , Insecticides/pharmacology , Parathion/pharmacology , Sex Factors , Animals , Animals, Newborn , Body Weight/drug effects , Feeding Behavior/drug effects , Female , Male , Rats
5.
PLoS Genet ; 4(7): e1000137, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18654634

ABSTRACT

In 404 Lep(ob/ob) F2 progeny of a C57BL/6J (B6) x DBA/2J (DBA) intercross, we mapped a DBA-related quantitative trait locus (QTL) to distal Chr1 at 169.6 Mb, centered about D1Mit110, for diabetes-related phenotypes that included blood glucose, HbA1c, and pancreatic islet histology. The interval was refined to 1.8 Mb in a series of B6.DBA congenic/subcongenic lines also segregating for Lep(ob). The phenotypes of B6.DBA congenic mice include reduced beta-cell replication rates accompanied by reduced beta-cell mass, reduced insulin/glucose ratio in blood, reduced glucose tolerance, and persistent mild hypoinsulinemic hyperglycemia. Nucleotide sequence and expression analysis of 14 genes in this interval identified a predicted gene that we have designated "Lisch-like" (Ll) as the most likely candidate. The gene spans 62.7 kb on Chr1qH2.3, encoding a 10-exon, 646-amino acid polypeptide, homologous to Lsr on Chr7qB1 and to Ildr1 on Chr16qB3. The largest isoform of Ll is predicted to be a transmembrane molecule with an immunoglobulin-like extracellular domain and a serine/threonine-rich intracellular domain that contains a 14-3-3 binding domain. Morpholino knockdown of the zebrafish paralog of Ll resulted in a generalized delay in endodermal development in the gut region and dispersion of insulin-positive cells. Mice segregating for an ENU-induced null allele of Ll have phenotypes comparable to the B.D congenic lines. The human ortholog, C1orf32, is in the middle of a 30-Mb region of Chr1q23-25 that has been repeatedly associated with type 2 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Receptors, Cell Surface/genetics , Amino Acid Sequence , Animals , Base Sequence , Blood Glucose/genetics , Chromosomes, Mammalian , Cloning, Molecular , Crosses, Genetic , Glucose Tolerance Test/methods , Haplotypes , Homozygote , Insulin/blood , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Obese , Molecular Sequence Data , Mutation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Quantitative Trait Loci
6.
J Med Chem ; 49(9): 2758-71, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16640337

ABSTRACT

The synthesis of a series of phenethanolamine aniline agonists that contain an aniline ring on the right-hand side of the molecule substituted at the meta position with a benzoic acid or a pyridyl carboxylate is described. Several of the analogues (e.g., 34, 36-38, 40, and 44) have high beta(3) adrenergic receptor (AR) potency and selectivity against beta(1) and beta(2) ARs in Chinese hamster ovary (CHO) cells expressing beta ARs. The dog pharmacokinetic profile of some of these analogues showed >25% oral bioavailability and po half-lives of at least 1.5 h. Among the compounds described herein, the 3,3'-biarylaniline carboxylate derivatives 36, 38 and the phenylpyridyl derivative 44 demonstrated outstanding in vitro properties and reasonable dog pharmacokinetic profiles. These three analogues also showed dose dependent beta(3) AR mediated responses in mice. The ease of synthesis and superior dog pharmacokinetics of compound 38 relative to that of 44 in combination with its in vitro profile led us to choose this compound as a development candidate for the treatment of type 2 diabetes.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Aniline Compounds/chemistry , Ethanolamine/chemistry , Ethanolamine/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Animals , Blood Glucose/metabolism , Cell Line , Cricetinae , Cyclic AMP/metabolism , Dogs , Ethanolamine/chemical synthesis , Glycosylation/drug effects , Hemoglobins/metabolism , Humans , Male , Mice , Molecular Structure , Structure-Activity Relationship
7.
Biochem Biophys Res Commun ; 341(1): 209-17, 2006 Mar 03.
Article in English | MEDLINE | ID: mdl-16414018

ABSTRACT

The current study examined the relationship between skeletal muscle levels of adiponectin and parameters of insulin sensitivity. A high fat/sucrose diet (HFD) for 20 weeks resulted in significant increases in body weight, serum insulin, triglycerides (TG), and free fatty acids (FFA) (all p < 0.01). Interestingly, this diet leads to a slight increase in serum adiponectin, but significant decreases in gastrocnemius muscle and white adipose adiponectin (all p < 0.05). HFD for 4 weeks also resulted in a significant decrease in muscle adiponectin, which correlated with serum insulin, TG, and FFA (all p < 0.05). Treatment of the 4-week HFD rats with a PPARgamma agonist GI262570 ameliorated the diet-induced hyperinsulinemia and dyslipidemia, and effectively restored muscle adiponectin (all p < 0.05). This study demonstrated that HFD-induced hyperinsulinemia and dyslipidemia appeared without changes in serum adiponectin, but were associated with decreased tissue adiponectin. This provides the first evidence for a connection between tissue adiponectin and diet-induced hyperinsulinemia and dyslipidemia.


Subject(s)
Adiponectin/metabolism , Dietary Fats/metabolism , Dietary Sucrose/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Animals , Dose-Response Relationship, Drug , Male , Oxazoles/administration & dosage , Rats , Rats, Sprague-Dawley , Tyrosine/administration & dosage , Tyrosine/analogs & derivatives
8.
Environ Health Perspect ; 113(10): 1291-4, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16203236

ABSTRACT

Developmental exposure to chlorpyrifos alters cell signaling both in the brain and in peripheral tissues, affecting the responses to a variety of neurotransmitters and hormones. We administered 1 mg/kg/day chlorpyrifos to rats on postnatal days 1-4, a regimen below the threshold for systemic toxicity. When tested in adulthood, chlorpyrifos-exposed animals displayed elevations in plasma cholesterol and triglycerides, without underlying alterations in nonesterified free fatty acids and glycerol. This effect was restricted to males. Similarly, in the postprandial state, male rats showed hyperinsulinemia in the face of normal circulating glucose levels but demonstrated appropriate reduction of circulating insulin concentrations after fasting. These outcomes and sex selectivity resemble earlier findings at the cellular level, which identified hepatic hyperresponsiveness to gluconeogenic inputs from beta-adrenoceptors or glucagon receptors. Our results thus indicate that apparently subtoxic neonatal chlorpyrifos exposure, devoid of effects on viability or growth but within the parameters of human fetal or neonatal exposures, produce a metabolic pattern for plasma lipids and insulin that resembles the major adult risk factors for atherosclerosis and type 2 diabetes mellitus.


Subject(s)
Chlorpyrifos/toxicity , Hyperinsulinism/chemically induced , Hyperlipidemias/chemically induced , Insecticides/toxicity , Sex Factors , Animals , Animals, Newborn , Body Weight , Female , Growth , Male , Pregnancy , Rats , Rats, Sprague-Dawley
9.
Pflugers Arch ; 451(3): 445-53, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16170524

ABSTRACT

Selective agonists of peroxisome proliferator-activated receptor gamma (PPARgamma) are anti-diabetic drugs that enhance cellular responsiveness to insulin. However, in some patients, fluid retention, plasma volume expansion, and edema have been observed. It is well established that insulin regulates Na(+) reabsorption via the epithelial sodium channel (ENaC) located in the distal tubule. Therefore, we hypothesized that these agonists may positively modulate insulin-stimulated ENaC activity leading to increased Na(+) reabsorption and fluid retention. Using electrophysiological techniques, dose-response curves for insulin-mediated Na(+) transport in the A6, M-1, and mpkCCD(cl4) cell lines were performed. Each line demonstrated hormone efficacy within physiological concentration ranges and, therefore, can be used to monitor clinically relevant effects of pharmacological agents which may affect electrolyte transport. Immunodetection and quantitative PCR analyses showed that each cell line expresses viable and functional PPARgamma receptors. Despite this finding, two PPARgamma agonists, pioglitazone and GW7845 did not directly enhance basal or insulin-stimulated Na(+) flux via ENaC, as shown by electrophysiological methodologies. These studies provide important results, which eliminate insulin-mediated ENaC activation as a candidate mechanism underlying the fluid retention observed with PPARgamma agonist use.


Subject(s)
Insulin/physiology , PPAR gamma/agonists , Sodium Channels/physiology , Sodium/metabolism , Animals , Cell Line , Edema/chemically induced , Epithelial Sodium Channels , Hypoglycemic Agents/adverse effects , Kidney/metabolism , Mice , Xenopus laevis
10.
Am J Physiol Endocrinol Metab ; 289(2): E328-36, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15797988

ABSTRACT

It is suggested that insulin resistance and metabolic maladaptation of the heart are causes of contractile dysfunction. We tested the hypothesis whether systemic PPARgamma activation, by changing the metabolic profile in a model of insulin resistance and type 2 diabetes (the ZDF rat) in vivo, improves contractile function of the heart in vitro. Male Zucker diabetic fatty (ZDF) and Zucker lean (ZL) rats, at 53-56 days of age, were treated with either GI-262570 (a nonthiazolidinedione PPARgamma agonist; A) or vehicle (V) for 1 wk. Agonist treatment resulted in correction of hyperglycemia and dyslipidemia, as well as in reduced hyperinsulinemia. The accumulation of triacylglycerols in the myocardium, characteristic of the ZDF rat, disappeared with treatment. Cardiac power and rates of glucose oxidation in the isolated working heart were significantly reduced in ZDF-V rats, but both parameters increased to nondiabetic levels with agonist treatment. In ZDF-V hearts, transcript levels of PPARalpha-regulated genes and of myosin heavy chain-beta were upregulated, whereas GLUT4 was downregulated compared with ZL. Agonist treatment of ZDF rats reduced PPARalpha-regulated genes and increased transcripts of GLUT4 and GLUT1. In conclusion, by changing the metabolic profile, reducing myocardial lipid accumulation, and promoting the downregulation of PPARalpha-regulated genes, PPARgamma activation leads to an increased capacity of the myocardium to oxidize glucose and to a tighter coupling of oxidative metabolism and contraction in the setting of insulin resistance and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , PPAR gamma/metabolism , Adaptation, Physiological , Animals , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , In Vitro Techniques , Insulin Resistance/physiology , Male , Myocardial Contraction/drug effects , Oxazoles/pharmacology , PPAR gamma/agonists , Rats , Rats, Inbred Strains , Rats, Zucker , Signal Transduction/drug effects , Signal Transduction/physiology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Triglycerides/metabolism , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
11.
J Pharmacol Exp Ther ; 312(2): 718-25, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15475592

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARgamma) agonists have been shown to have significant therapeutic benefits such as desirable glycemic control in type 2 diabetic patients; however, these agents may cause fluid retention in susceptible individuals. Since PPARgamma is expressed selectively in distal nephron epithelium, we studied the mechanism of PPARgamma agonist-induced fluid retention using male Sprague-Dawley rats treated with either vehicle or GI262570 (farglitazar), a potent PPARgamma agonist. GI262570 (20 mg/kg/day) induced a plasma volume expansion. The plasma volume expansion was accompanied by a small but significant decrease in plasma potassium concentration. Small but significant increases in plasma sodium and chloride concentrations were also observed. These changes in serum electrolytes suggested an activation of the renal mineralocorticoid response system; however, GI262570-treated rats had lower plasma levels of aldosterone compared with vehicle-treated controls. mRNA levels for a group of genes involved in distal nephron sodium and water absorption are changed in the kidney medulla with GI262570 treatment. In addition, due to a possible rebound effect on epithelial sodium channel (ENaC) activity, a low dose of amiloride did not prevent GI262570-induced fluid retention. On the contrary, the rebound effect after amiloride treatment potentiated GI262570-induced plasma volume expansion. This is at least partially due to a synergistic effect of GI262570 and the rebound from amiloride treatment on ENaCalpha expression. In summary, our current data suggest that GI262570 can increase water and sodium reabsorption in distal nephron by stimulating the ENaC and Na,K-ATPase system. This may be an important mechanism for PPARgamma agonist-induced fluid retention.


Subject(s)
Electrolytes/metabolism , Kidney Tubules, Distal/metabolism , Nephrons/metabolism , Oxazoles/pharmacology , PPAR gamma/agonists , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , Water/metabolism , Actins/biosynthesis , Aldosterone/blood , Amiloride/pharmacology , Animals , Blood Volume/drug effects , Diuretics/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Sodium Channels , Gene Expression/drug effects , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/drug effects , Male , Nephrons/cytology , Nephrons/drug effects , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism , Sodium Channels/biosynthesis , Sodium Channels/genetics
12.
J Clin Invest ; 114(11): 1564-76, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15578089

ABSTRACT

PPARalpha, beta/delta, and gamma regulate genes involved in the control of lipid metabolism and inflammation and are expressed in all major cell types of atherosclerotic lesions. In vitro studies have suggested that PPARs exert antiatherogenic effects by inhibiting the expression of proinflammatory genes and enhancing cholesterol efflux via activation of the liver X receptor-ABCA1 (LXR-ABCA1) pathway. To investigate the potential importance of these activities in vivo, we performed a systematic analysis of the effects of PPARalpha, beta, and gamma agonists on foam-cell formation and atherosclerosis in male LDL receptor-deficient (LDLR(-/-)) mice. Like the PPARgamma agonist, a PPARalpha-specific agonist strongly inhibited atherosclerosis, whereas a PPARbeta-specific agonist failed to inhibit lesion formation. In concert with their effects on atherosclerosis, PPARalpha and PPARgamma agonists, but not the PPARbeta agonist, inhibited the formation of macrophage foam cells in the peritoneal cavity. Unexpectedly, PPARalpha and PPARgamma agonists inhibited foam-cell formation in vivo through distinct ABCA1-independent pathways. While inhibition of foam-cell formation by PPARalpha required LXRs, activation of PPARgamma reduced cholesterol esterification, induced expression of ABCG1, and stimulated HDL-dependent cholesterol efflux in an LXR-independent manner. In concert, these findings reveal receptor-specific mechanisms by which PPARs influence macrophage cholesterol homeostasis. In the future, these mechanisms may be exploited pharmacologically to inhibit the development of atherosclerosis.


Subject(s)
Arteriosclerosis/metabolism , Foam Cells/physiology , PPAR alpha/metabolism , PPAR delta/metabolism , PPAR gamma/metabolism , PPAR-beta/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/pathology , Cholesterol/metabolism , Cholesterol, Dietary , DNA-Binding Proteins , Gene Expression Regulation , Humans , Liver X Receptors , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Orphan Nuclear Receptors , PPAR alpha/agonists , PPAR alpha/genetics , PPAR delta/agonists , PPAR delta/genetics , PPAR gamma/agonists , PPAR gamma/genetics , PPAR-beta/agonists , PPAR-beta/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Triglycerides/metabolism
13.
BMC Pharmacol ; 4: 23, 2004 Oct 18.
Article in English | MEDLINE | ID: mdl-15491498

ABSTRACT

BACKGROUND: PPARgamma agonists ameliorate insulin resistance and dyslipidemia in type 2 diabetic patients. Adiponectin possesses insulin sensitizing properties, and predicts insulin sensitivity of both glucose and lipid metabolism. In diet-induced insulin resistant rats and ZDF rats, the current studies determined the correlation between PPARgamma agonist-upregulated fatty acid binding protein(FABP3) mRNA in adipose tissue and PPARgamma agonist-elevated serum adiponectin, and the correlation between PPARgamma agonist-elevated serum adiponectin and PPARgamma agonist-mediated efficacy in insulin sensitization and lipid lowering. RESULTS: Parallel groups of SD rats were fed a high fat/sucrose (HF) diet for 4 weeks. These rats were orally treated for the later 2 weeks with vehicle, either PPARgamma agonist GI262570 (0.2-100 mg/kg, Q.D.), or GW347845 (3 mg/kg, B.I.D). Rats on HF diet showed significant increases in postprandial serum triglycerides, free fatty acids (FFA), insulin, and area under curve (AUC) of serum insulin during an oral glucose tolerance test, but showed no change in serum glucose, adiponectin, and glucose AUC. Treatment with GI262570 dose-dependently upregulated adipose FABP3 mRNA, and increased serum adiponectin. There was a position correlation between adipose FABP3 mRNA and serum adiponectin (r = 0.7350, p < 0.01). GI262570 dose-dependently decreased the diet-induced elevations in triglycerides, FFA, insulin, and insulin AUC. Treatment with GW347845 had similar effects on serum adiponectin and the diet-induced elevations. There were negative correlations for adiponectin versus triglycerides, FFA, insulin, and insulin AUC (For GI262570, r = -0.7486, -0.4581, -0.4379, and -0.3258 respectively, all p < 0.05. For GW347845, r = -0.6370, -0.6877, -0.5512, and -0.3812 respectively, all p < 0.05). In ZDF rats treated with PPARgamma agonists pioglitazone (3-30 mg/kg, B.I.D.) or GW347845 (3 mg/kg, B.I.D.), there were also negative correlations for serum adiponectin versus glucose, triglycerides, FFA (for pioglitazone, r = -0.7005, -0.8603, and -0.9288 respectively; for GW347845, r = -0.9721, -0.8483, and -0.9453 respectively, all p < 0.01). CONCLUSIONS: This study demonstrated that (a) PPARgamma agonists improved insulin sensitivity and ameliorated dyslipidemia in HF fed rats and ZDF rats, which were correlated with serum adiponectin; (b) Serum adiponectin was positively correlated with adipose FABP3 mRNA in GI262570-treated rats. These data suggest that serum adiponectin can serve as a biomarker for both in vivo PPARgamma activation and PPARgamma agonist-induced efficacy on insulin resistance and dyslipidemia in rats.


Subject(s)
Insulin/physiology , Intercellular Signaling Peptides and Proteins/blood , Lipids/blood , PPAR gamma/agonists , PPAR gamma/metabolism , Adiponectin , Animals , Biomarkers/blood , Carrier Proteins/metabolism , Diabetes Mellitus/metabolism , Fatty Acid-Binding Proteins , Insulin/blood , Insulin Resistance/physiology , Male , Obesity/metabolism , Oxazoles/pharmacology , Pioglitazone , Rats , Rats, Sprague-Dawley , Rats, Zucker , Thiazolidinediones/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
14.
J Pharmacol Exp Ther ; 310(3): 1226-33, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15126643

ABSTRACT

This study provides novel data on the regional hemodynamic effects of the peroxisome proliferator-activated receptor-gamma activator, GI 262570 [(S)-2-(2-benzoylphenylamino)-3-[4-[2-(5-methyl-2-phenyl-2-oxazol-4-yl)ethoxy]phenyl]propionic acid], in conscious, male Sprague-Dawley rats. Administration of GI 262570 twice daily for 4 days caused a slowly developing, modest fall in mean arterial blood pressure, associated with a progressive, hyperemic hindquarters vasodilatation, but with no consistent changes in renal or mesenteric hemodynamics. The hindquarters vasodilator effect of GI 262570 was not inhibited by the beta2-adrenoceptor antagonist, ICI 118551 ((+/-)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl) amino]-2-butanol hydrochloride), and was still apparent in the presence of the alpha-adrenoceptor antagonist, phentolamine. Neither the latter, nor antagonism of angiotensin (AT1) and endothelin (ETA and ETB) receptors unmasked vasodilator responses to GI 262570 in the renal or mesenteric vascular beds. In the presence of GI 262570, vasodilator responses to acetylcholine and vasoconstrictor responses to methoxamine were normal. Furthermore, the cardiovascular responses to nonselective nitric-oxide synthase inhibition were not influenced by GI 262570. Collectively, these results indicate that the vasodilator action of GI 262570 is specific to the hindquarters vascular bed (of those studied), does not involve alpha- or beta2-adrenoceptors, and is not associated with a change in basal or stimulated nitric oxide release.


Subject(s)
Hemodynamics/drug effects , Oxazoles/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Tyrosine/pharmacology , Acetylcholine/pharmacology , Animals , Indans/pharmacology , Male , Methoxamine/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Naphthyridines/pharmacology , Nitric Oxide/metabolism , Phentolamine/pharmacology , Propanolamines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Tyrosine/analogs & derivatives
15.
J Cardiovasc Pharmacol ; 42(3): 436-41, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12960690

ABSTRACT

PPARgamma agonists ameliorate insulin resistance and lower blood pressure. Volume expansion/edema has been observed in susceptible patients treated with these agents. Alterations of renal hemodynamics affect renal tubular reabsorption, and thus may contribute to volume expansion. This study seeks to determine whether volume expansion caused by a PPARgamma agonist, GI262570, is related to changes in glomerular filtration rate, effective renal plasma flow, or renal filtration fraction. Chronically catheter-implanted conscious rats were studied to determine the effects on glomerular filtration rate, effective renal plasma flow, and renal filtration fraction after 1, 4, and 10 days of GI262570 treatment (8 mg/kg, p.o., B.I.D.). Elevated adipose mRNA of PPARgamma target genes confirmed PPARgamma activation in GI262570-treated rats. GI262570 treatment for 10 days decreased hematocrit, hemoglobin, and serum albumin (all P < 0.05), indicating volume expansion, but did not alter glomerular filtration rate, effective renal plasma flow, or renal filtration fraction. However, nitrate + nitrite was significantly higher in plasma and hind limb muscle of GI262570-treated rats (both P < 0.05). This study demonstrated that treatment with PPARgamma agonist GI262570 resulted in volume expansion and increased nitric oxide, but did not affect glomerular filtration rate, effective renal plasma flow, or renal filtration fraction, indicating PPARgamma agonist-induced volume expansion is not related to changes in renal filtration fraction, and increased nitric oxide may contribute to the PPARgamma agonist-induced blood-pressure lowering.


Subject(s)
Glomerular Filtration Rate/drug effects , Hypoglycemic Agents/pharmacology , Nitric Oxide/metabolism , Oxazoles , Receptors, Cytoplasmic and Nuclear/agonists , Renal Plasma Flow, Effective/drug effects , Transcription Factors/agonists , Animals , Male , Nitrates/blood , Nitric Oxide/blood , Nitrites/blood , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Tyrosine/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...