Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Sci Rep ; 9(1): 18338, 2019 12 04.
Article En | MEDLINE | ID: mdl-31797889

Radiolabeled exendin derivatives are promising for non-invasive quantification of pancreatic beta cell mass (BCM); longitudinal observation of BCM for evaluation of therapeutic effects has not been achieved. The aim of this study is to demonstrate the usefulness of our developing method using [Lys12(111In-BnDTPA-Ahx)]exendin-4 to detect longitudinal changes in BCM. We performed a longitudinal study with obese type 2 diabetes model (db/db) mice administered canagliflozin, which is reported to preserve BCM. Six-week-old mice were assigned to a canagliflozin-administered group or a control group. Blood glucose levels of the canagliflozin group were significantly lower than those of the control group. Plasma insulin levels, insulin secretion during OGTT and insulin content in the pancreas were preserved in the canagliflozin group in comparison with those in the control group. According to SPECT/CT imaging analysis using [Lys12(111In-BnDTPA-Ahx)]exendin-4, pancreatic uptake was significantly decreased in the control group, whereas there was no significant change in the canagliflozin group. After nine weeks, both pancreatic uptake and BCM of the canagliflozin group were significantly higher than those of the control group, and a correlation between them was observed. In conclusion, our imaging method confirmed the BCM-preservation effect of canagliflozin, and demonstrated its potential for longitudinal evaluation of BCM.


Canagliflozin/pharmacology , Diabetes Mellitus, Type 2/diagnostic imaging , Exenatide/isolation & purification , Insulin-Secreting Cells/drug effects , Animals , Blood Glucose , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Exenatide/chemistry , Exenatide/genetics , Humans , Indium Radioisotopes/chemistry , Indium Radioisotopes/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/pathology , Mice, Inbred NOD , Pancreas/diagnostic imaging , Pancreas/drug effects , Pancreas/pathology , Single Photon Emission Computed Tomography Computed Tomography
2.
J Pharmacol Sci ; 132(4): 255-261, 2016 Dec.
Article En | MEDLINE | ID: mdl-27889414

We investigated whether structurally different sodium-glucose cotransporter (SGLT) 2 inhibitors, when co-administered with dipeptidyl peptidase-4 (DPP4) inhibitors, could enhance glucagon-like peptide-1 (GLP-1) secretion during oral glucose tolerance tests (OGTTs) in rodents. Three different SGLT inhibitors-1-(ß-d-Glucopyranosyl)-4-chloro-3-[5-(6-fluoro-2-pyridyl)-2-thienylmethyl]benzene (GTB), TA-1887, and canagliflozin-were examined to assess the effect of chemical structure. Oral treatment with GTB plus a DPP4 inhibitor enhanced glucose-induced plasma active GLP-1 (aGLP-1) elevation and suppressed glucose excursions in both normal and diabetic rodents. In DPP4-deficient rats, GTB enhanced glucose-induced aGLP-1 elevation without affecting the basal level, whereas metformin, previously reported to enhance GLP-1 secretion, increased both the basal level and glucose-induced elevation. Oral treatment with canagliflozin and TA-1887 also enhanced glucose-induced aGLP-1 elevation when co-administered with either teneligliptin or sitagliptin. These data suggest that structurally different SGLT2 inhibitors enhance plasma aGLP-1 elevation and suppress glucose excursions during OGTT when co-administered with DPP4 inhibitors, regardless of the difference in chemical structure. Combination treatment with DPP4 inhibitors and SGLT2 inhibitors having moderate SGLT1 inhibitory activity may be a promising therapeutic option for improving glycemic control in patients with type 2 diabetes mellitus.


Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1/blood , Hypoglycemic Agents/pharmacology , Sodium-Glucose Transporter 2 Inhibitors , Animals , Canagliflozin/pharmacology , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Monosaccharides/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Inbred F344 , Sitagliptin Phosphate/pharmacology , Sodium-Glucose Transporter 1/antagonists & inhibitors , Thiazolidines/pharmacology
3.
J Pharmacol Exp Ther ; 358(1): 94-102, 2016 07.
Article En | MEDLINE | ID: mdl-27189972

Canagliflozin, a selective sodium/glucose cotransporter (SGLT) 2 inhibitor, suppresses the renal reabsorption of glucose and decreases blood glucose level in patients with type 2 diabetes. A characteristic of canagliflozin is its modest SGLT1 inhibitory action in the intestine at clinical dosage. To reveal its mechanism of action, we investigated the interaction of canagliflozin with SGLT1 and SGLT2. Inhibition kinetics and transporter-mediated uptake were examined in human SGLT1- or SGLT2-expressing cells. Whole-cell patch-clamp recording was conducted to examine the sidedness of drug action. Canagliflozin competitively inhibited SGLT1 and SGLT2, with high potency and selectivity for SGLT2. Inhibition constant (Ki) values for SGLT1 and SGLT2 were 770.5 and 4.0 nM, respectively. (14)C-canagliflozin was suggested to be transported by SGLT2; however, the transport rate was less than that of α-methyl-d-glucopyranoside. Canagliflozin inhibited α-methyl-d-glucopyranoside-induced SGLT1- and SGLT2-mediated inward currents preferentially from the extracellular side and not from the intracellular side. Based on the Ki value, canagliflozin is estimated to sufficiently inhibit SGLT2 from the urinary side in renal proximal tubules. The Ki value for SGLT1 suggests that canagliflozin suppresses SGLT1 in the small intestine from the luminal side, whereas it does not affect SGLT1 in the heart and skeletal muscle, considering the maximal concentration of plasma-unbound canagliflozin. Similarly, SGLT1 in the kidney would not be inhibited, thereby aiding in the prevention of hypoglycemia. After binding to SGLT2, canagliflozin may be reabsorbed by SGLT2, which leads to the low urinary excretion and prolonged drug action of canagliflozin.


Canagliflozin/pharmacology , Hypoglycemic Agents/pharmacology , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors , Action Potentials/drug effects , Animals , Binding, Competitive , CHO Cells , Canagliflozin/metabolism , Cricetulus , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hypoglycemic Agents/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kinetics , Patch-Clamp Techniques , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 2/genetics , Transfection
4.
J Pharmacol Exp Ther ; 354(3): 279-89, 2015 Sep.
Article En | MEDLINE | ID: mdl-26105952

The sodium glucose cotransporter (SGLT) 1 plays a major role in glucose absorption and incretin hormone release in the gastrointestinal tract; however, the impact of SGLT1 inhibition on plasma glucagon-like peptide-1 (GLP-1) levels in vivo is controversial. We analyzed the effects of SGLT1 inhibitors on GLP-1 secretion in normoglycemic and hyperglycemic rodents using phloridzin, CGMI [3-(4-cyclopropylphenylmethyl)-1-(ß-d-glucopyranosyl)-4-methylindole], and canagliflozin. These compounds are SGLT2 inhibitors with moderate SGLT1 inhibitory activity, and their IC50 values against rat SGLT1 and mouse SGLT1 were 609 and 760 nM for phloridzin, 39.4 and 41.5 nM for CGMI, and 555 and 613 nM for canagliflozin, respectively. Oral administration of these inhibitors markedly enhanced and prolonged the glucose-induced plasma active GLP-1 (aGLP-1) increase in combination treatment with sitagliptin, a dipeptidyl peptidase-4 (DPP4) inhibitor, in normoglycemic mice and rats. CGMI, the most potent SGLT1 inhibitor among them, enhanced glucose-induced, but not fat-induced, plasma aGLP-1 increase at a lower dose compared with canagliflozin. Both CGMI and canagliflozin delayed intestinal glucose absorption after oral administration in normoglycemic rats. The combined treatment of canagliflozin and a DPP4 inhibitor increased plasma aGLP-1 levels and improved glucose tolerance compared with single treatment in both 8- and 13-week-old Zucker diabetic fatty rats. These results suggest that transient inhibition of intestinal SGLT1 promotes GLP-1 secretion by delaying glucose absorption and that concomitant inhibition of intestinal SGLT1 and DPP4 is a novel therapeutic option for glycemic control in type 2 diabetes mellitus.


Diabetes Mellitus, Experimental/blood , Glucagon-Like Peptide 1/blood , Intestinal Mucosa/metabolism , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 1/metabolism , Animals , CHO Cells , Cricetulus , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Dipeptidyl Peptidase 4/metabolism , Glucose/metabolism , Humans , Hyperglycemia/blood , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Intestinal Absorption/drug effects , Intestines/drug effects , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Rats, Zucker , Sodium-Glucose Transporter 2/metabolism
5.
J Pharmacol Sci ; 127(4): 456-61, 2015 Apr.
Article En | MEDLINE | ID: mdl-25892328

To assess the impact of concomitant inhibition of sodium-glucose cotransporter (SGLT) 2 and dipeptidyl peptidase IV (DPP4) for the treatment of type 2 diabetes mellitus (T2DM), the effect of combined treatment with canagliflozin, a novel SGLT2 inhibitor, and teneligliptin, a DPP4 inhibitor, on glucose intolerance was investigated in Zucker diabetic fatty (ZDF) rats. Canagliflozin potently inhibited human and rat SGLT2 and moderately inhibited human and rat SGLT1 activities but did not affect DPP4 activity. In contrast, teneligliptin inhibited human and rat DPP4 activities but not SGLT activities. A single oral treatment of canagliflozin and teneligliptin suppressed plasma glucose elevation in an oral glucose tolerance test in 13 week-old ZDF rats. This combination of agents elevated plasma active GLP-1 levels in a synergistic manner, probably mediated by intestinal SGLT1 inhibition, and further improved glucose intolerance. In the combination-treated animals, there was no pharmacokinetic interaction of the drugs and no further inhibition of plasma DPP4 activity compared with that in the teneligliptin-treated animals. These results suggest that the inhibition of SGLT2 and DPP4 improves glucose intolerance and that combined treatment with canagliflozin and teneligliptin is a novel therapeutic option for glycemic control in T2DM.


Canagliflozin/pharmacology , Canagliflozin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucose Intolerance/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Thiazolidines/pharmacology , Thiazolidines/therapeutic use , Administration, Oral , Animals , Canagliflozin/administration & dosage , Cells, Cultured , Cricetinae , Cricetulus , Diabetes Mellitus, Type 2/blood , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Therapy, Combination , Glucagon-Like Peptide 1/blood , Humans , Hypoglycemic Agents/administration & dosage , Male , Pyrazoles/administration & dosage , Rats, Zucker , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 , Sodium-Glucose Transporter 2 Inhibitors , Thiazolidines/administration & dosage
6.
PLoS One ; 10(1): e0116851, 2015.
Article En | MEDLINE | ID: mdl-25615826

BACKGROUND: Despite its insulin sensitizing effects, pioglitazone may induce weight gain leading to an increased risk of development of insulin resistance. A novel sodium glucose co-transporter 2 (SGLT2) inhibitor, canagliflozin, provides not only glycemic control but also body weight reduction through an insulin-independent mechanism. The aim of this study was to investigate the combined effects of these agents on body weight control and insulin sensitivity. METHODS: Effects of combination therapy with canagliflozin and pioglitazone were evaluated in established diabetic KK-Ay mice and prediabetic Zucker diabetic fatty (ZDF) rats. RESULTS: In the KK-Ay mice, the combination therapy further improved glycemic control compared with canagliflozin or pioglitazone monotherapy. Furthermore, the combination significantly attenuated body weight and fat gain induced by pioglitazone and improved hyperinsulinemia. In the ZDF rats, early intervention with pioglitazone monotherapy almost completely prevented the progressive development of hyperglycemia, and no further improvement was observed by add-on treatment with canagliflozin. However, the combination significantly reduced pioglitazone-induced weight gain and adiposity and improved the Matsuda index, suggesting improved whole-body insulin sensitivity. CONCLUSIONS: Our study indicates that combination therapy with canagliflozin and pioglitazone improves insulin sensitivity partly by preventing glucotoxicity and, at least partly, by attenuating pioglitazone-induced body weight gain in two different obese diabetic animal models. This combination therapy may prove to be a valuable option for the treatment and prevention of obese type 2 diabetes.


Canagliflozin/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Hyperinsulinism/drug therapy , Hypoglycemic Agents/administration & dosage , Obesity/drug therapy , Thiazolidinediones/administration & dosage , Adipose Tissue/drug effects , Animals , Body Weight/drug effects , Canagliflozin/therapeutic use , Cells, Cultured , Drug Therapy, Combination , Hyperinsulinism/chemically induced , Hypoglycemic Agents/therapeutic use , Mice , Obesity/chemically induced , Pioglitazone , Rats , Rats, Zucker , Thiazolidinediones/therapeutic use
7.
J Pharmacol Exp Ther ; 351(2): 423-31, 2014 Nov.
Article En | MEDLINE | ID: mdl-25216746

Sodium-glucose cotransporter 2 (SGLT2) plays a major role in renal glucose reabsorption. To analyze the potential of insulin-independent blood glucose control, the effects of the novel SGLT2 inhibitor canagliflozin on renal glucose reabsorption and the progression of hyperglycemia were analyzed in Zucker diabetic fatty (ZDF) rats. The transporter activity of recombinant human and rat SGLT2 was inhibited by canagliflozin, with 150- to 12,000-fold selectivity over other glucose transporters. Moreover, in vivo treatment with canagliflozin induced glucosuria in mice, rats, and dogs in a dose-dependent manner. It inhibited apparent glucose reabsorption by 55% in normoglycemic rats and by 94% in hyperglycemic rats. The inhibition of glucose reabsorption markedly reduced hyperglycemia in ZDF rats but did not induce hypoglycemia in normoglycemic animals. The change in urinary glucose excretion should not be used as a marker to predict the glycemic effects of this SGLT2 inhibitor. In ZDF rats, plasma glucose and HbA1c levels progressively increased with age, and pancreatic ß-cell failure developed at 13 weeks of age. Treatment with canagliflozin for 8 weeks from the prediabetic stage suppressed the progression of hyperglycemia, prevented the decrease in plasma insulin levels, increased pancreatic insulin contents, and minimized the deterioration of islet structure. These results indicate that selective inhibition of SGLT2 with canagliflozin controls the progression of hyperglycemia by inhibiting renal glucose reabsorption in ZDF rats. In addition, the preservation of ß-cell function suggests that canagliflozin treatment reduces glucose toxicity via an insulin-independent mechanism.


Glucose/metabolism , Glucosides/pharmacology , Hyperglycemia/drug therapy , Hyperglycemia/pathology , Kidney/drug effects , Thiophenes/pharmacology , Animals , Blood Glucose/drug effects , Canagliflozin , Diabetes Mellitus, Experimental , Dogs , Glycated Hemoglobin/metabolism , Humans , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Kidney/metabolism , Kidney/pathology , Mice , Rats , Rats, Zucker , Sodium-Glucose Transporter 2/metabolism
8.
Lipids Health Dis ; 11: 75, 2012 Jun 14.
Article En | MEDLINE | ID: mdl-22698140

BACKGROUND: Resynthesis of triglycerides in enterocytes of the small intestine plays a critical role in the absorption of dietary fat. Acyl-CoA:monoacylglycerol acyltransferase-2 (MGAT2) is highly expressed in the small intestine and catalyzes the synthesis of diacylglycerol from monoacylglycerol and acyl-CoA. To determine the physiological importance of MGAT2 in metabolic disorders and lipid metabolism in the small intestine, we constructed and analyzed Mgat2-deficient mice. RESULTS: In oral fat tolerance test (OFTT), Mgat2-deficient mice absorbed less fat into the circulation. When maintained on a high-fat diet (HFD), Mgat2-deficient mice were protected from HFD-induced obesity and insulin resistance. Heterozygote (Mgat2+/-) mice had an intermediate phenotype between Mgat2+/+ and Mgat2-/- and were partially protected from metabolic disorders. Despite of a decrease in fat absorption in the Mgat2-deficient mice, lipid levels in the feces and small intestine were comparable among the genotypes. Oxygen consumption was increased in the Mgat2-deficient mice when maintained on an HFD. A prominent upregulation of the genes involved in fatty acid oxidation was observed in the duodenum but not in the liver of the Mgat2-deficient mice. CONCLUSION: These results suggest that MGAT2 has a pivotal role in lipid metabolism in the small intestine, and the inhibition of MGAT2 activity may be a promising strategy for the treatment of obesity-related metabolic disorders.


Diet, High-Fat/adverse effects , Fats/metabolism , Insulin Resistance/physiology , Intestinal Absorption/physiology , Obesity/metabolism , Animals , Body Composition/genetics , Body Composition/physiology , Glucose Tolerance Test , Insulin Resistance/genetics , Intestinal Absorption/genetics , Mice , Mice, Knockout , N-Acetylglucosaminyltransferases , Obesity/genetics , Polymerase Chain Reaction
9.
Biochem Biophys Res Commun ; 328(2): 369-74, 2005 Mar 11.
Article En | MEDLINE | ID: mdl-15694357

Hyperglycemia-induced activation of hexosamine biosynthesis pathway (HBP) has been implicated in the development of insulin resistance in skeletal muscles. In the present study, the content of uridine-5'-diphospho-N-acetylglucosamine, the end product of the HBP, was elevated in skeletal muscle of obese diabetic KKA(y) mice, compared with control mice. To elucidate the effect of elevated HBP in the skeletal muscle, we treated C2C12 myoblasts with glucosamine, an intermediate metabolite of the HBP. Glucosamine induced lipid accumulation and significantly increased the mRNA expression levels of peroxisome proliferator-activated receptor gamma, adiponectin, and aP2 in C2C12 myoblasts. Similar mRNA changes were observed in skeletal muscles of Sprague-Dawley rats treated with glucosamine infusion. Our results provide a possible explanation of hyperglycemia-induced insulin resistance in skeletal muscle.


Adipocytes/metabolism , Adipocytes/pathology , Lipid Metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/metabolism , Myoblasts/pathology , Adipocytes/drug effects , Animals , Cell Differentiation/drug effects , Cell Line , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Hexosamines/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Myoblasts/drug effects , Rats , Rats, Sprague-Dawley , Uridine Diphosphate N-Acetylglucosamine/metabolism
...