Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Biol Pharm Bull ; 40(5): 675-680, 2017.
Article in English | MEDLINE | ID: mdl-28458353

ABSTRACT

Sodium glucose cotransporter 2 (SGLT2) inhibitors improve hyperglycemia in patients with type 2 diabetes mellitus (T2DM) by increasing urinary glucose excretion. In addition to their antihyperglycemic effect, SGLT2 inhibitors also reduce body weight and fat mass in obese and overweight patients with T2DM. However, whether or not SGLT2 inhibitors similarly affect body composition of non-obese patients with T2DM remains unclear. In this study, we investigated the effect of the SGLT2 inhibitor ipragliflozin on body composition in a Goto-Kakizaki (GK) rat model of non-obese T2DM. GK rats were treated with ipragliflozin once daily for 9 weeks, starting at 23 weeks of age. Body composition was then analyzed using dual-energy X-ray absorptiometry. Treatment with ipragliflozin increased urinary glucose excretion, reduced hemoglobin A1c (HbA1c) levels and suppressed body weight gain as the dose increased. Body composition analysis revealed that body fat mass was lower in the ipragliflozin-treated groups than in the control group, while lean body mass and bone mineral contents were comparable between groups. Thus, an SGLT2 inhibitor ipragliflozin was found to promote preferential loss of fat mass in a rat model of non-obese T2DM. Ipragliflozin might also promote preferential loss of fat in non-obese patients with T2DM.


Subject(s)
Adipose Tissue/drug effects , Diabetes Mellitus, Type 2/drug therapy , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Sodium-Glucose Transporter 2 Inhibitors , Thiophenes/pharmacology , Absorptiometry, Photon , Adipose Tissue/pathology , Animals , Body Composition/drug effects , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Eating/drug effects , Glycosuria/metabolism , Male , Rats , Rats, Wistar , Sodium-Glucose Transporter 2 , Weight Loss/drug effects
3.
Proc Natl Acad Sci U S A ; 103(28): 10741-6, 2006 Jul 11.
Article in English | MEDLINE | ID: mdl-16818881

ABSTRACT

The c-Jun N-terminal kinases (JNKs) are key regulators of inflammation and interfere with insulin action in cultured cells and whole animals. Obesity increases total JNK activity, and JNK1, but not JNK2, deficiency results in reduced adiposity and improved insulin sensitivity. Interestingly, a higher-than-normal level of JNK activation is observed in Jnk2(-/-) mice, particularly in the liver, indicating an interaction between the isoforms that might have masked the metabolic activity of JNK2 in isolated mutant mice. To address the role of the JNK2 isoform in metabolic homeostasis, we intercrossed Jnk1(-/-) and Jnk2(-/-) mice and examined body weight and glucose metabolism in the resulting mutant allele combinations. Among all of the viable genotypes examined, we observed only reduced body weight and increased insulin sensitivity in Jnk1(-/-) and Jnk1(+/-)Jnk2(-/-) mice. These two groups of mice also exhibited reduced total JNK activity and cytokine expression in liver tissue compared with all other genotypes examined. These data indicate that the JNK2 isoform is also involved in metabolic regulation, but its function is not obvious when JNK1 is fully expressed because of regulatory crosstalk between the two isoforms.


Subject(s)
Insulin Resistance , Mitogen-Activated Protein Kinase 8/physiology , Mitogen-Activated Protein Kinase 9/physiology , Obesity/enzymology , Animals , Cell Line , Genetic Markers , Glucose/metabolism , Humans , Insulin Resistance/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Lipid Metabolism/genetics , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Obesity/genetics
4.
Eur J Pharmacol ; 519(1-2): 182-90, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16039648

ABSTRACT

Effect of 3-(2,4-dichlorobenzyl)-2-methyl-N-(pentylsulfonyl)-3H-benzimidazole-5-carboxamide (FK614), a novel nonthiazolidinedione peroxisome proliferator-activated receptor (PPAR) gamma agonist, on glucose tolerance and insulin resistance in peripheral tissues and in liver using Zucker fatty rats (genetically obese and insulin-resistant) was evaluated and compared to other insulin sensitizers. FK614 (0.32, 1 and 3.2 mg/kg), two thiazolidinedione PPAR gamma agonists, rosiglitazone (0.1, 0.32, 1 and 3.2 mg/kg) and pioglitazone (1, 3.2 and 10 mg/kg), and a biguanide, metformin (320 and 1000 mg/kg), were orally administered to Zucker fatty rats once a day for 14 days. Zucker fatty rats treated with FK614 and rosiglitazone were subjected to evaluation by oral glucose tolerance test. Ameliorating effect of each compound on peripheral and hepatic insulin resistance was evaluated using a euglycemic-hyperinsulineamic clamp procedure. FK614 and rosiglitazone dose-dependently improved impaired glucose tolerance in Zucker fatty rats. In addition, FK614 dose-dependently ameliorated peripheral and hepatic insulin resistance in Zucker fatty rats, with the degree of its effect in peripheral tissues almost equivalent to that in liver when compared at each dose tested. Similar data indicating ameliorating effects on insulin resistance was obtained for rosiglitazone and pioglitazone. Metformin showed less potent effects than other insulin sensitizers and its effect in liver tended to be greater than that in peripheral tissues. These findings suggest clinical potential for FK614 as a treatment of type 2 diabetes, acting by ameliorating insulin resistance both in peripheral tissues and liver.


Subject(s)
Benzimidazoles/pharmacology , Insulin Resistance , PPAR gamma/agonists , Administration, Oral , Animals , Benzimidazoles/administration & dosage , Blood Glucose/metabolism , Dose-Response Relationship, Drug , Glucose/metabolism , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Intolerance/prevention & control , Glucose Tolerance Test , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Insulin/blood , Insulin/pharmacology , Liver/drug effects , Liver/metabolism , Male , Metformin/administration & dosage , Metformin/pharmacology , Pioglitazone , Rats , Rats, Zucker , Rosiglitazone , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology , Time Factors
5.
J Antibiot (Tokyo) ; 58(10): 640-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16392680

ABSTRACT

The pharmacological effect of FR177391, isolated from Serratia liquefaciens No. 1821, was studied in normal animals and various types of animal models of hypertriglyceridemia. Treatment of normal mice with FR177391 resulted in an increase in heparin-releasable lipoprotein lipase (LPL) activity in the blood and epididymal fat tissue. FR177391 treatment decreased triglyceride (TG) and increased high-density lipoprotein cholesterol in the blood in normal rats following 7 days treatment, suggesting potent LPL activating properties of FR177391. Both Triton WR1339-induced severe and fructose-induced mild hypertriglyceridemia in rats were attenuated by FR177391 treatment. Severely elevated levels of TG in db/db mice, an insulin resistant diabetic animal model, also significantly decreased from 14 days of treatment with FR177391. FR177391 treatment for 9 days caused a decrease in the elevated levels of TG in mice induced by intraperitoneal inoculation of murine lymphoma EL-4. Overall, this study demonstrated that FR177391 can be possibly a LPL activating agent and that FR177391 treatment improved hypertriglyceridemia in various rat and mouse animal models. These results suggest that FR177391 is a promising candidate compound for the management of hypertriglyceridemia.


Subject(s)
Acetates/pharmacology , Heterocyclic Compounds/pharmacology , Hypertriglyceridemia/physiopathology , Hypolipidemic Agents/pharmacology , Serratia/chemistry , Triglycerides/blood , Animals , Hypertriglyceridemia/genetics , Hypertriglyceridemia/pathology , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/isolation & purification , Mice , Mice, Inbred BALB C , Rats
6.
Eur J Pharmacol ; 494(2-3): 273-81, 2004 Jun 28.
Article in English | MEDLINE | ID: mdl-15212984

ABSTRACT

We evaluated antidiabetic effects of 3-(2,4-dichlorobenzyl)-2-methyl-N-(pentylsulfonyl)-3 H-benzimidazole-5-carboxamide (FK614), a benzimidazole derivative without a thiazolidinedione structure, which was obtained using C57BL/KsJ-db/db mice (db/db mice). In db/db mice, the potency of FK614 for hypoglycemic effect was comparable to that of rosiglitazone and approximately 15-fold greater than that of pioglitazone. FK614 also showed a potent attenuating effect on hypertriglyceridemia in db/db mice, as well as rosiglitazone and pioglitazone. In C57BL/6J-ob/ob mice (ob/ob mice), ED(50) values of FK614 and pioglitazone for hypoinsulinemic effect were 1.3 and 11.8 mg/kg, respectively. FK614 also improved the impaired glucose tolerance in ob/ob mice. In normal rats, FK614 did not influence plasma glucose and insulin levels but significantly decreased both plasma triglyceride and nonesterified fatty acid levels. FK614 was found to activate peroxisome proliferator-activated receptor (PPAR)gamma-mediated transcriptional activity in the reporter gene assay as well as thiazolidinedione derivatives, although its maximum effect was less than that of thiazolidinedione derivatives. In rat toxicity studies, hemodilution effects for FK614 were less than that for rosiglitazone. Overall, these studies suggest that FK614 improves insulin resistance in such animal models through activation of PPARgamma-mediated transcriptional activity and that it would be a new therapeutic candidate with potential for the treatment of type 2 diabetic patients.


Subject(s)
Benzimidazoles/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Animals , Benzimidazoles/toxicity , Blood Glucose/metabolism , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Female , Hypoglycemic Agents/toxicity , Insulin/blood , Insulin Resistance/physiology , Lipids/blood , Male , Mice , Mice, Inbred C57BL , PPAR gamma/metabolism , Pioglitazone , Plasmids/genetics , Rats , Rats, Sprague-Dawley , Thiazolidinediones/therapeutic use , Transcription, Genetic/drug effects , Transfection
7.
Eur J Pharmacol ; 477(2): 171-8, 2003 Sep 12.
Article in English | MEDLINE | ID: mdl-14519421

ABSTRACT

Our recent study suggests that there is a reciprocal mechanism to maintain cGMP content, via both a decrease in cGMP degradation (decrease in cGMP-phosphodiesterase activity) and an increase in synthesis of cGMP (increase in guanylate cyclase activity) in the kidney of cyclosporin A-treated rats. We undertook this study to clarify the role of cGMP-phosphodiesterase in cyclosporin A nephrotoxicity by evaluating N-(3,4-dimethoxybenzyl)-2-[[(1R)-2-hydroxy-1-methylethyl]amino]-5-nitrobenzamide (FR226807), a phosphodiesterase type 5 inhibitor, in an animal model. Male spontaneous hypertensive rats (SHR) were treated with cyclosporin A (50 mg/kg) for 2 weeks or with cyclosporin A and FR226807 (3.2 mg/kg or 10 mg/kg) for 2 weeks. Cyclosporin A-treated rats showed renal dysfunction and histological change compared with vehicle-treated rats. Administration of FR226807 improved the renal dysfunction (increase in serum creatinine and fractional excretion of sodium, and decrease in creatinine clearance) as well as the pathological changes (tubular vacuolization) induced by cyclosporin A in SHR. At the molecular level, administration of FR226807 resulted in a further increase in cGMP content in the kidney, aorta and platelets from cyclosporin A-treated rats. Our present study demonstrates that cGMP-phosphodiesterase plays an important role in the cyclosporin A nephrotoxicity and also suggests that further inhibition of cGMP-phosphodiesterase is a potential pharmacological target for preventing cyclosporin A nephrotoxicity.


Subject(s)
Benzamides/pharmacology , Cyclosporine/antagonists & inhibitors , Immunosuppressive Agents/antagonists & inhibitors , Kidney Diseases/prevention & control , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases , Animals , Aorta/metabolism , Blood Platelets/metabolism , Body Weight/drug effects , Cyclic GMP/blood , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5 , Cyclosporine/pharmacology , Guanylate Cyclase/metabolism , Immunosuppressive Agents/pharmacology , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/chemically induced , Kidney Diseases/physiopathology , Male , Nitric Oxide Synthase/metabolism , Potassium/blood , Rats , Rats, Inbred SHR , Sodium/blood
8.
Dig Dis Sci ; 48(7): 1398-405, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12870804

ABSTRACT

A recent study by our group demonstrated that a 1-week infusion of insulin-like growth factor-I increases bile flow volume and bile acid secretion in rats, suggesting it is important in in vivo choleresis. In the present study, the effect in rats of a single administration of insulin-like growth factor-I on bile flow volume was investigated and compared with the choleretic drugs prednisolone and ursodeoxycholic acid. A significant and long-lasting increase in bile flow volume was observed in rats treated with insulin-like growth factor-I or prednisolone. Ursodeoxycholic acid significantly, but transiently increased. Combined treatment using insulin-like growth factor-I with prednisolone or ursodeoxycholic acid additively increased bile flow volume. Overall, this study demonstrated that the stimulatory effect of insulin-like growth factor-I on bile flow volume is almost equally potent to that of prednisolone and ursodeoxycholic acid, indicating the possible therapeutic potential of insulin-like growth factor-I in cholestatic liver diseases.


Subject(s)
Bile/drug effects , Cholagogues and Choleretics/pharmacology , Digestive System Physiological Phenomena/drug effects , Insulin-Like Growth Factor I/pharmacology , Prednisolone/pharmacology , Ursodeoxycholic Acid/pharmacology , Animals , Bile/physiology , Male , Rats , Rats, Sprague-Dawley
9.
Nature ; 420(6913): 333-6, 2002 Nov 21.
Article in English | MEDLINE | ID: mdl-12447443

ABSTRACT

Obesity is closely associated with insulin resistance and establishes the leading risk factor for type 2 diabetes mellitus, yet the molecular mechanisms of this association are poorly understood. The c-Jun amino-terminal kinases (JNKs) can interfere with insulin action in cultured cells and are activated by inflammatory cytokines and free fatty acids, molecules that have been implicated in the development of type 2 diabetes. Here we show that JNK activity is abnormally elevated in obesity. Furthermore, an absence of JNK1 results in decreased adiposity, significantly improved insulin sensitivity and enhanced insulin receptor signalling capacity in two different models of mouse obesity. Thus, JNK is a crucial mediator of obesity and insulin resistance and a potential target for therapeutics.


Subject(s)
Insulin Resistance/physiology , Mitogen-Activated Protein Kinases/metabolism , Obesity/enzymology , Obesity/physiopathology , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Adipose Tissue/physiopathology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diet , Disease Models, Animal , Gene Deletion , Homeostasis , Hyperinsulinism/complications , Hyperinsulinism/enzymology , Hyperinsulinism/genetics , Hyperinsulinism/physiopathology , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Insulin Resistance/genetics , Lipid Metabolism , Mice , Mice, Knockout , Mice, Obese , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/deficiency , Mitogen-Activated Protein Kinases/genetics , Obesity/complications , Obesity/genetics , Phosphoproteins/metabolism , Phosphorylation , Receptor, Insulin/metabolism , Signal Transduction/drug effects
10.
Endocrinology ; 143(4): 1502-11, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11897709

ABSTRACT

Aberrant TNF alpha expression in adipocytes is a molecular mechanism by which insulin action is modulated in adipose tissue. While this might be a compensatory response to limit adipose expansion, neither the mechanisms underlying this local effect nor its systemic biological consequences have been studied. It is also not clear whether TNF alpha-induced insulin resistance in adipocyte alone is responsible for systemic insulin resistance in the absence of obesity. In a transgenic mouse model deficient in endogenous TNF alpha, we demonstrate that specific expression of the transmembrane TNF alpha (mTNF alpha) in adipocytes leads to decreased whole body adipose mass, and local, but not systemic insulin resistance. These data demonstrate that exclusive action of TNF alpha in adipose tissue strongly inhibits insulin action at this site and leads to reduced adiposity in mice. However, this isolated adipocyte insulin resistance in the context of reduced fat mass and/or the absence of obesity is insufficient to alter systemic glucose homeostasis.


Subject(s)
Adipose Tissue/physiology , Insulin Resistance/physiology , Tumor Necrosis Factor-alpha/physiology , Adipocytes/metabolism , Animals , Blotting, Northern , Blotting, Southern , Cell Membrane/metabolism , Eating , Feces/chemistry , Glucose/metabolism , Glucose Tolerance Test , Immunoblotting , Indicators and Reagents , Insulin/blood , Lipid Metabolism , Lipids/analysis , Male , Mice , Mice, Transgenic , Organ Size/physiology , Phenotype , Precipitin Tests , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...