Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
FASEB J ; 30(3): 1207-17, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26644352

ABSTRACT

Although mutations in the Wnt/ß-catenin signaling pathway are linked with the metabolic syndrome and type 2 diabetes in humans, the mechanism is unclear. High-fat-fed male C57BL/6 mice were treated for 4 wk with a 2'-O-methoxyethyl chimeric antisense oligonucleotide (ASO) to decrease hepatic and adipose expression of ß-catenin. ß-Catenin mRNA decreased by ≈80% in the liver and by 70% in white adipose tissue relative to control ASO-treated mice. ß-Catenin ASO improved hepatic insulin sensitivity and increased insulin-stimulated whole body glucose metabolism, as assessed during hyperinsulinemic-euglycemic clamp in awake mice. ß-Catenin ASO altered hepatic lipid composition in high-fat-fed mice. There were reductions in hepatic triglyceride (44%, P < 0.05) and diacylglycerol content (60%, P < 0.01) but a 30% increase in ceramide content (P < 0.001). The altered lipid content was attributed to decreased expression of sn-1,2 diacylglycerol acyltransferase and mitochondrial acyl-CoA:glycerol-sn-3-phosphate acyltransferase and an increase in serine palmitoyl transferase. The decrease in cellular diacyglycerol was associated with a 33% decrease in PKCε activation (P < 0.05) and 64% increase in Akt2 phosphorylation (P < 0.05). In summary, Reducing ß-catenin expression decreases expression of enzymes involved in hepatic fatty acid esterification, ameliorates hepatic steatosis and lipid-induced insulin resistance.


Subject(s)
Fatty Liver/prevention & control , Insulin Resistance/physiology , Oligonucleotides, Antisense/pharmacology , beta Catenin/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Dietary Fats/metabolism , Diglycerides/metabolism , Fatty Acids/metabolism , Fatty Liver/drug therapy , Fatty Liver/genetics , Fatty Liver/metabolism , Glucose/metabolism , Insulin/metabolism , Lipids/physiology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/genetics , Protective Agents/pharmacology , Triglycerides/metabolism
2.
Am J Physiol Endocrinol Metab ; 307(9): E773-83, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25159329

ABSTRACT

The steroid receptor coactivator 1 (SRC1) regulates key metabolic pathways, including glucose homeostasis. SRC1(-/-) mice have decreased hepatic expression of gluconeogenic enzymes and a reduction in the rate of endogenous glucose production (EGP). We sought to determine whether decreasing hepatic and adipose SRC1 expression in normal adult rats would alter glucose homeostasis and insulin action. Regular chow-fed and high-fat-fed male Sprage-Dawley rats were treated with an antisense oligonucleotide (ASO) against SRC1 or a control ASO for 4 wk, followed by metabolic assessments. SRC1 ASO did not alter basal EGP or expression of gluconeogenic enzymes. Instead, SRC1 ASO increased insulin-stimulated whole body glucose disposal by ~30%, which was attributable largely to an increase in insulin-stimulated muscle glucose uptake. This was associated with an approximately sevenfold increase in adipose expression of lipocalin-type prostaglandin D2 synthase, a previously reported regulator of insulin sensitivity, and an approximately 70% increase in plasma PGD2 concentration. Muscle insulin signaling, AMPK activation, and tissue perfusion were unchanged. Although GLUT4 content was unchanged, SRC1 ASO increased the cleavage of tether-containing UBX domain for GLUT4, a regulator of GLUT4 translocation. These studies point to a novel role of adipose SRC1 as a regulator of insulin-stimulated muscle glucose uptake.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glucose Intolerance/drug therapy , Insulin Resistance , Muscle, Skeletal/drug effects , Nuclear Receptor Coactivator 1/antagonists & inhibitors , Oligodeoxyribonucleotides, Antisense/therapeutic use , Adipose Tissue/drug effects , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Biological Transport/drug effects , Diet, High-Fat/adverse effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Glucose Transporter Type 4/agonists , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/metabolism , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/agonists , Lipocalins/genetics , Lipocalins/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Muscle, Skeletal/metabolism , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Prostaglandin D2/blood , Prostaglandin D2/metabolism , Protein Interaction Domains and Motifs , Proteolysis/drug effects , Rats, Sprague-Dawley
3.
PLoS One ; 8(7): e66923, 2013.
Article in English | MEDLINE | ID: mdl-23922646

ABSTRACT

Obesity is a primary risk factor for multiple metabolic disorders. Many drugs for the treatment of obesity, which mainly act through CNS as appetite suppressants, have failed during development or been removed from the market due to unacceptable adverse effects. Thus, there are very few efficacious drugs available and remains a great unmet medical need for anti-obesity drugs that increase energy expenditure by acting on peripheral tissues without severe side effects. Here, we report a novel approach involving antisense inhibition of fibroblast growth factor receptor 4 (FGFR4) in peripheral tissues. Treatment of diet-induce obese (DIO) mice with FGFR4 antisense oligonucleotides (ASO) specifically reduced liver FGFR4 expression that not only resulted in decrease in body weight (BW) and adiposity in free-feeding conditions, but also lowered BW and adiposity under caloric restriction. In addition, combination treatment with FGFR4 ASO and rimonabant showed additive reduction in BW and adiposity. FGFR4 ASO treatment increased basal metabolic rate during free-feeding conditions and, more importantly, prevented adaptive decreases of metabolic rate induced by caloric restriction. The treatment increased fatty acid oxidation while decreased lipogenesis in both liver and fat. Mechanistic studies indicated that anti-obesity effect of FGFR4 ASO was mediated at least in part through an induction of plasma FGF15 level resulted from reduction of hepatic FGFR4 expression. The anti-obesity effect was accompanied by improvement in plasma glycemia, whole body insulin sensitivity, plasma lipid levels and liver steatosis. Therefore, FGFR4 could be a potential novel target and antisense reduction of hepatic FGFR4 expression could be an efficacious therapy as an adjunct to diet restriction or to an appetite suppressant for the treatment of obesity and related metabolic disorders.


Subject(s)
Adiposity/drug effects , Basal Metabolism/drug effects , Obesity/metabolism , Oligonucleotides, Antisense/pharmacology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Animals , Bile Acids and Salts/metabolism , Body Weight/drug effects , Caloric Restriction , Diet , Drug Therapy, Combination , Fatty Acids/metabolism , Fatty Liver/blood , Fatty Liver/genetics , Fatty Liver/pathology , Feeding Behavior/drug effects , Fibroblast Growth Factors/blood , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin/pharmacology , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/blood , Obesity/drug therapy , Obesity/genetics , Oligonucleotides, Antisense/therapeutic use , Oxidation-Reduction/drug effects , Piperidines/pharmacology , Piperidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , Rimonabant
4.
Diabetes ; 62(7): 2183-94, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23423574

ABSTRACT

We measured the mRNA and protein expression of the key gluconeogenic enzymes in human liver biopsy specimens and found that only hepatic pyruvate carboxylase protein levels related strongly with glycemia. We assessed the role of pyruvate carboxylase in regulating glucose and lipid metabolism in rats through a loss-of-function approach using a specific antisense oligonucleotide (ASO) to decrease expression predominantly in liver and adipose tissue. Pyruvate carboxylase ASO reduced plasma glucose concentrations and the rate of endogenous glucose production in vivo. Interestingly, pyruvate carboxylase ASO also reduced adiposity, plasma lipid concentrations, and hepatic steatosis in high fat-fed rats and improved hepatic insulin sensitivity. Pyruvate carboxylase ASO had similar effects in Zucker Diabetic Fatty rats. Pyruvate carboxylase ASO did not alter de novo fatty acid synthesis, lipolysis, or hepatocyte fatty acid oxidation. In contrast, the lipid phenotype was attributed to a decrease in hepatic and adipose glycerol synthesis, which is important for fatty acid esterification when dietary fat is in excess. Tissue-specific inhibition of pyruvate carboxylase is a potential therapeutic approach for nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes.


Subject(s)
Adiposity/physiology , Gluconeogenesis/physiology , Insulin Resistance/physiology , Liver/enzymology , Pyruvate Carboxylase/metabolism , Adipose Tissue/enzymology , Adult , Animals , Fatty Liver/enzymology , Female , Glycerol/metabolism , Humans , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Rats, Zucker
5.
Proc Natl Acad Sci U S A ; 110(5): 1869-74, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23302688

ABSTRACT

Comparative gene identification 58 (CGI-58) is a lipid droplet-associated protein that promotes the hydrolysis of triglyceride by activating adipose triglyceride lipase. Loss-of-function mutations in CGI-58 in humans lead to Chanarin-Dorfman syndrome, a condition in which triglyceride accumulates in various tissues, including the skin, liver, muscle, and intestines. Therefore, without adequate CGI-58 expression, lipids are stored rather than used for fuel, signaling intermediates, and membrane biosynthesis. CGI-58 knockdown in mice using antisense oligonucleotide (ASO) treatment also leads to severe hepatic steatosis as well as increased hepatocellular diacylglycerol (DAG) content, a well-documented trigger of insulin resistance. Surprisingly, CGI-58 knockdown mice remain insulin-sensitive, seemingly dissociating DAG from the development of insulin resistance. Therefore, we sought to determine the mechanism responsible for this paradox. Hyperinsulinemic-euglycemic clamp studies reveal that the maintenance of insulin sensitivity with CGI-58 ASO treatment could entirely be attributed to protection from lipid-induced hepatic insulin resistance, despite the apparent lipotoxic conditions. Analysis of the cellular compartmentation of DAG revealed that DAG increased in the membrane fraction of high fat-fed mice, leading to PKCε activation and hepatic insulin resistance. However, DAG increased in lipid droplets or lipid-associated endoplasmic reticulum rather than the membrane of CGI-58 ASO-treated mice, and thus prevented PKCε translocation to the plasma membrane and induction of insulin resistance. Taken together, these results explain the disassociation of hepatic steatosis and DAG accumulation from hepatic insulin resistance in CGI-58 ASO-treated mice, and highlight the importance of intracellular compartmentation of DAG in causing lipotoxicity and hepatic insulin resistance.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Diglycerides/metabolism , Endoplasmic Reticulum/metabolism , Insulin Resistance , Lipids/chemistry , Liver/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Diet, High-Fat , Endoplasmic Reticulum/drug effects , Gene Expression/drug effects , Gene Knockdown Techniques , Humans , Immunoblotting , Injections, Intraperitoneal , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Protein Kinase C-epsilon/metabolism , Protein Transport/drug effects , Reverse Transcriptase Polymerase Chain Reaction
6.
Endocrinology ; 154(1): 36-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23161873

ABSTRACT

By 2030, nearly half of Americans will have nonalcoholic fatty liver disease. In part, this epidemic is fueled by the increasing consumption of caloric sweeteners coupled with an innate capacity to convert sugar into fat via hepatic de novo lipogenesis. In addition to serving as substrates, monosaccharides also increase the expression of key enzymes involved in de novo lipogenesis via the carbohydrate response element-binding protein (ChREBP). To determine whether ChREBP is a potential therapeutic target, we decreased hepatic expression of ChREBP with a specific antisense oligonucleotide (ASO) in male Sprague-Dawley rats fed either a high-fructose or high-fat diet. ChREBP ASO treatment decreased plasma triglyceride concentrations compared with control ASO treatment in both diet groups. The reduction was more pronounced in the fructose-fed group and attributed to decreased hepatic expression of ACC2, FAS, SCD1, and MTTP and a decrease in the rate of hepatic triglyceride secretion. This was associated with an increase in insulin-stimulated peripheral glucose uptake, as assessed by the hyperinsulinemic-euglycemic clamp. In contrast, ChREBP ASO did not alter hepatic lipid content or hepatic insulin sensitivity. Interestingly, fructose-fed rats treated with ChREBP ASO had increased plasma uric acid, alanine transaminase, and aspartate aminotransferase concentrations. This was associated with decreased expression of fructose aldolase and fructokinase, reminiscent of inherited disorders of fructose metabolism. In summary, these studies suggest that targeting ChREBP may prevent fructose-induced hypertriglyceridemia but without the improvements in hepatic steatosis and hepatic insulin responsiveness.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fructose/pharmacology , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Liver/drug effects , Liver/metabolism , Male , Oligonucleotides, Antisense/genetics , Rats , Rats, Sprague-Dawley , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Triglycerides/metabolism , fas Receptor/genetics , fas Receptor/metabolism
7.
Hepatology ; 57(5): 1763-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23175050

ABSTRACT

UNLABELLED: Genome-wide array studies have associated the patatin-like phospholipase domain-containing 3 (PNPLA3) gene polymorphisms with hepatic steatosis. However, it is unclear whether PNPLA3 functions as a lipase or a lipogenic enzyme and whether PNPLA3 is involved in the pathogenesis of hepatic insulin resistance. To address these questions we treated high-fat-fed rats with specific antisense oligonucleotides to decrease hepatic and adipose pnpla3 expression. Reducing pnpla3 expression prevented hepatic steatosis, which could be attributed to decreased fatty acid esterification measured by the incorporation of [U-(13) C]-palmitate into hepatic triglyceride. While the precursors for phosphatidic acid (PA) (long-chain fatty acyl-CoAs and lysophosphatidic acid [LPA]) were not decreased, we did observe an ∼20% reduction in the hepatic PA content, ∼35% reduction in the PA/LPA ratio, and ∼60%-70% reduction in transacylation activity at the level of acyl-CoA:1-acylglycerol-sn-3-phosphate acyltransferase. These changes were associated with an ∼50% reduction in hepatic diacylglycerol (DAG) content, an ∼80% reduction in hepatic protein kinase Cε activation, and increased hepatic insulin sensitivity, as reflected by a 2-fold greater suppression of endogenous glucose production during the hyperinsulinemic-euglycemic clamp. Finally, in humans, hepatic PNPLA3 messenger RNA (mRNA) expression was strongly correlated with hepatic triglyceride and DAG content, supporting a potential lipogenic role of PNPLA3 in humans. CONCLUSION: PNPLA3 may function primarily in a lipogenic capacity and inhibition of PNPLA3 may be a novel therapeutic approach for treatment of nonalcoholic fatty liver disease-associated hepatic insulin resistance.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/chemically induced , Fatty Liver/physiopathology , Insulin Resistance/physiology , Lipids/adverse effects , Membrane Proteins/physiology , Phospholipases A2/physiology , Animals , Biopsy , Diglycerides/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Membrane Proteins/drug effects , Membrane Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Phospholipases A2/drug effects , Phospholipases A2/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Triglycerides/metabolism
8.
J Med Chem ; 51(19): 6173-87, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18788731

ABSTRACT

A novel series of symmetrical ureas of [(7-amino(2-naphthyl))sulfonyl]phenylamines were designed, synthesized, and tested for their ability to increase glucose transport in mouse 3T3-L1 adipocytes, a surrogate readout for activation of the insulin receptor (IR) tyrosine kinase (IRTK). A structure-activity relationship was established that indicated glucose transport activity was dependent on the presence of two acidic functionalities, two sulfonamide linkages, and a central urea or 2-imidazolidinone core. Compound 30 was identified as a potent and selective IRTK activator. At low concentrations, 30 was able to increase the tyrosine phosphorylation of the IR stimulated by submaximal insulin. At higher concentrations, 30 was able to increase tyrosine the phosphorylation levels of the IR in the absence of insulin. When administered intraperitoneally (ip) and orally (po), 30 improved glucose tolerance in hypoinsulinemic, streptozotocin-treated rats. These data provide pharmacological validation that small molecule IRTK activators represent a potential new class of antidiabetic agents.


Subject(s)
Aniline Compounds/pharmacology , Drug Design , Receptor, Insulin/drug effects , Sulfonamides/pharmacology , Urea/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Administration, Oral , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Animals , Binding Sites , Blood Glucose/analysis , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Fibroblasts/drug effects , Glucose Tolerance Test , Injections, Intraperitoneal , Male , Mice , Molecular Structure , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Stereoisomerism , Streptozocin/administration & dosage , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Urea/analogs & derivatives , Urea/chemistry
10.
J Biol Chem ; 282(19): 14291-9, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17353188

ABSTRACT

To investigate the role of low molecular weight protein-tyrosine phosphatase (LMW-PTP) in glucose metabolism and insulin action, a specific antisense oligonucleotide (ASO) was used to reduce its expression both in vitro and in vivo. Reduction of LMW-PTP expression with the ASO in cultured mouse hepatocytes and in liver and fat tissues of diet-induced obese (DIO) mice and ob/ob mice led to increased phosphorylation and activity of key insulin signaling intermediates, including insulin receptor-beta subunit, phosphatidylinositol 3-kinase, and Akt in response to insulin stimulation. The ASO-treated DIO and ob/ob animals showed improved insulin sensitivity, which was reflected by a lowering of both plasma insulin and glucose levels and improved glucose and insulin tolerance in DIO mice. The treatment did not decrease body weight or increase metabolic rate. These data demonstrate that LMW-PTP is a key negative regulator of insulin action and a potential novel target for the treatment of insulin resistance and type 2 diabetes.


Subject(s)
Hyperglycemia/metabolism , Insulin Resistance , Insulin/metabolism , Isoenzymes/metabolism , Obesity/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Body Weight , Glucose Tolerance Test , Hepatocytes/metabolism , Immunoprecipitation , Insulin Receptor Substrate Proteins , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Signal Transduction
11.
Diabetes ; 54(6): 1846-53, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15919808

ABSTRACT

Glucocorticoids (GCs) increase hepatic gluconeogenesis and play an important role in the regulation of hepatic glucose output. Whereas systemic GC inhibition can alleviate hyperglycemia in rodents and humans, it results in adrenal insufficiency and stimulation of the hypothalamic-pituitary-adrenal axis. In the present study, we used optimized antisense oligonucleotides (ASOs) to cause selective reduction of the glucocorticoid receptor (GCCR) in liver and white adipose tissue (WAT) and evaluated the resultant changes in glucose and lipid metabolism in several rodent models of diabetes. Treatment of ob/ob mice with GCCR ASOs for 4 weeks resulted in approximately 75 and approximately 40% reduction in GCCR mRNA expression in liver and WAT, respectively. This was accompanied by approximately 65% decrease in fed and approximately 30% decrease in fasted glucose levels, a 60% decrease in plasma insulin concentration, and approximately 20 and 35% decrease in plasma resistin and tumor necrosis factor-alpha levels, respectively. Furthermore, GCCR ASO reduced hepatic glucose production and inhibited hepatic gluconeogenesis in liver slices from basal and dexamethasone-treated animals. In db/db mice, a similar reduction in GCCR expression caused approximately 40% decrease in fed and fasted glucose levels and approximately 50% reduction in plasma triglycerides. In ZDF and high-fat diet-fed streptozotocin-treated (HFD-STZ) rats, GCCR ASO treatment caused approximately 60% reduction in GCCR expression in the liver and WAT, which was accompanied by a 40-70% decrease in fasted glucose levels and a robust reduction in plasma triglyceride, cholesterol, and free fatty acids. No change in circulating corticosterone levels was seen in any model after GCCR ASO treatment. To further demonstrate that GCCR ASO does not cause systemic GC antagonism, normal Sprague-Dawley rats were challenged with dexamethasone after treating with GCCR ASO. Dexamethasone increased the expression of GC-responsive genes such as PEPCK in the liver and decreased circulating lymphocytes. GCCR ASO treatment completely inhibited the increase in dexamethasone-induced PEPCK expression in the liver without causing any change in the dexamethasone-induced lymphopenia. These studies demonstrate that tissue-selective GCCR antagonism with ASOs may be a viable therapeutic strategy for the treatment of the metabolic syndrome.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Experimental/drug therapy , Liver/metabolism , Oligoribonucleotides, Antisense/pharmacology , Receptors, Glucocorticoid/metabolism , Animals , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Gene Expression/drug effects , Glucocorticoids/metabolism , Hyperglycemia/drug therapy , Hyperlipidemias/drug therapy , Lymphopenia/chemically induced , Lymphopenia/physiopathology , Mice , Mice, Obese , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pro-Opiomelanocortin/metabolism , RNA, Messenger/metabolism , Rats
12.
J Cell Biochem ; 92(6): 1234-45, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15258906

ABSTRACT

Protease inhibitor (PI) therapy for the treatment of patients infected with human immunodeficiency virus is frequently associated with insulin resistance and diabetic complications. These adverse effects of PI treatment result to a large extent from their inhibition of insulin-stimulated glucose transport. Insulin receptor (IR) activators that enhance the insulin signaling pathway could be effective in treating this resistance. However, there are no agents reported that reverse inhibition of insulin action by PIs. Herein, we describe the effects of TLK19781. This compound is a non-peptide, small molecule, activator of the IR. We now report in cultured cells, made insulin resistant HIV by PI treatment, that TLK19781 both increased the content of insulin-stimulated GLUT4 at the plasma membrane, and enhanced insulin-stimulated glucose transport. In addition, oral administration of TLK19781 with the PI, indinavir improved glucose tolerance in rats made insulin resistant. These results suggest, therefore, that IR activators such as TLK19781 may be useful in treating the insulin resistance associated with PIs.


Subject(s)
HIV Protease Inhibitors/pharmacology , Indinavir/pharmacology , Insulin Resistance , Naphthalenes/pharmacology , Receptor, Insulin/agonists , Sulfanilic Acids/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Administration, Oral , Animals , Biological Transport , ErbB Receptors/metabolism , Glucose/metabolism , Glucose Tolerance Test , Glucose Transporter Type 4 , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , In Vitro Techniques , Indinavir/administration & dosage , Indinavir/adverse effects , Mice , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/metabolism , Naphthalenes/administration & dosage , Phosphorylation , Rats , Receptor, Insulin/metabolism , Sulfanilic Acids/administration & dosage
13.
Curr Med Chem ; 9(24): 2231-42, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12470244

ABSTRACT

Diabetes mellitus refers to a spectrum of syndromes characterized by abnormally high levels of glucose in blood. These syndromes are associated with an absolute (Type 1 diabetes) or relative (Type 2 diabetes) deficiency of insulin, coupled with varying degrees of peripheral resistance to the actions of insulin. Clinical studies have shown that controlling hyperglycemia significantly reduces the appearance and progression of the vascular complications associated with diabetes. Insulin's regulation of glucose homeostasis is mediated by a cascade of signaling events that take place upon insulin binding to its cell surface receptor. Autophosphorylation of the receptor and activation of its intrinsic tyrosine kinase are critical processes for transmitting these intracellular signals. Type 1 diabetes patients depend on exogenous insulin to achieve these effects, whereas Type 2 diabetes patients can accomplish a similar response through oral medications that increase the production of endogenous insulin or enhance its actions on the target tissues. Current biochemical and clinical evidence suggests that defects within the insulin receptor itself may be a cause of insulin resistance leading to Type 2 diabetes. This review focuses on the insulin receptor as a target for therapeutic intervention, and describes the recent discovery of small molecules that act on the receptor and either enhance or directly emulate the actions of insulin both in vitro and in vivo.


Subject(s)
Diabetes Mellitus/drug therapy , Hypoglycemic Agents/therapeutic use , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Diabetes Mellitus/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Humans , Hypoglycemic Agents/chemistry , Structure-Activity Relationship
14.
J Biol Chem ; 277(46): 43565-71, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12213804

ABSTRACT

In type 2 diabetes mellitus, impaired insulin signaling leads to hyperglycemia and other metabolic abnormalities. TLK19780, a non-peptide small molecule, is a new member of a novel class of anti-diabetic agents that function as activators of the insulin receptor (IR) beta-subunit tyrosine kinase. In HTC-IR cells, 20 microm TLK19780 enhanced maximal insulin-stimulated IR autophosphorylation 2-fold and increased insulin sensitivity 2-3-fold. In contrast, TLK19780 did not potentiate the action of insulin-like growth factor-1, indicating the selectivity of TLK19780 toward the IR. The predominant effect of TLK19780 was to increase the number of IR that underwent autophosphorylation. Kinetic studies indicated that TLK19780 acted very rapidly, with a maximal effect observed 2 min after addition to insulin-stimulated cells. In 3T3-L1 adipocytes, 5 microm TLK19780 enhanced insulin-stimulated glucose transport, increasing both the sensitivity and maximal responsiveness to insulin. These studies indicate that at low micromolar levels small IR activator molecules can enhance insulin action in various cultured cells and suggest that this effect is mediated by increasing the number of IR that are tyrosine-phosphorylated in response to insulin. These studies suggest that these types of molecules could be developed to treat type 2 diabetes and other clinical conditions associated with insulin resistance.


Subject(s)
Receptor, Insulin/metabolism , Receptor, Insulin/physiology , 3T3 Cells , Adipocytes/metabolism , Animals , Biological Transport , Blotting, Western , CHO Cells , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Cricetinae , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Enzyme-Linked Immunosorbent Assay , Glucose/metabolism , Kinetics , Mice , Models, Chemical , Phosphorylation , Rats , Sulfanilic Acids/pharmacology , Time Factors , Tyrosine/metabolism , Urea/analogs & derivatives , Urea/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...