Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Pharmacol Sin ; 42(2): 264-271, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32737468

ABSTRACT

Previous studies demonstrated that prolonged exposure to elevated levels of free fatty acids (FFA), especially saturated fatty acids, could lead to pancreatic ß-cell apoptosis, which plays an important role in the progression of type 2 diabetes (T2D). Diacylglycerol acyltransferase 1 (DGAT1), an enzyme that catalyzes the final step of triglyceride (TG) synthesis, has been reported as a novel target for the treatment of multiple metabolic diseases. In this study we evaluated the potential beneficial effects of DGAT1 inhibitors on pancreatic ß-cells, and further verified their antidiabetic effects in db/db mice. We showed that DGAT1 inhibitors (4a and LCQ908) at the concentration of 1 µM significantly ameliorated palmitic acid (PA)-induced apoptosis in MIN6 pancreatic ß-cells and primary cultured mouse islets; oral administration of a DGAT1 inhibitor (4a) (100 mg/kg) for 4 weeks significantly reduced the apoptosis of pancreatic islets in db/db mice. Meanwhile, 4a administration significantly decreased fasting blood glucose and TG levels, and improved glucose tolerance and insulin tolerance in db/db mice. Furthermore, we revealed that pretreatment with 4a (1 µM) significantly alleviated PA-induced intracellular lipid accumulation, endoplasmic reticulum (ER) stress, and proinflammatory responses in MIN6 cells, which might contribute to the protective effects of DGAT1 inhibitors on pancreatic ß-cells. These findings provided a better understanding of the antidiabetic effects of DGAT1 inhibitors.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Animals , Apoptosis/drug effects , Blood Glucose/drug effects , Cell Line , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Endoplasmic Reticulum Stress/drug effects , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Palmitic Acid/toxicity
2.
Acta Pharmacol Sin ; 40(9): 1193-1204, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30833709

ABSTRACT

Gluconeogenesis is a major source of hyperglycemia in patients with type 2 diabetes mellitus (T2DM), thus targeting gluconeogenesis to suppress glucose production is a promising strategy for anti-T2DM drug discovery. In our preliminary in vitro studies, we found that a small-molecule (E)-3-(2-(quinoline-4-yl)vinyl)-1H-indol-6-ol (QVO) inhibited the hepatic glucose production (HGP) in primary hepatocytes. We further revealed that QVO suppressed hepatic gluconeogenesis involving calmodulin-dependent protein kinase kinase ß- and liver kinase B1-adenosine monophosphate-activated protein kinase (AMPK) pathways as well as AMPK-independent mitochondrial function-related signaling pathway. To evaluate QVO's anti-T2DM activity in vivo, which was impeded by the complicated synthesis route of QVO with a low yield, we designed and synthesized 4-[2-(1H-indol-3-yl)vinyl]quinoline (IVQ) as a prodrug with easier synthesis route and higher yield. IVQ did not inhibit the HGP in primary hepatocytes in vitro. Pharmacokinetic studies demonstrated that IVQ was quickly converted to QVO in mice and rats following administration. In both db/db and ob/ob mice, oral administration of IVQ hydrochloride (IVQ-HCl) (23 and 46 mg/kg every day, for 5 weeks) ameliorated hyperglycemia, and suppressed hepatic gluconeogenesis and activated AMPK signaling pathway in the liver tissues. Furthermore, IVQ caused neither cardiovascular system dysfunction nor genotoxicity. The good druggability of IVQ has highlighted its potential in the treatment of T2DM and the prodrug design for anti-T2DM drug development.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gluconeogenesis/drug effects , Hypoglycemic Agents/therapeutic use , Indoles/therapeutic use , Prodrugs/therapeutic use , Quinolines/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Experimental/drug therapy , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex III/antagonists & inhibitors , Enzyme Activators/therapeutic use , Enzyme Activators/toxicity , Enzyme Inhibitors/therapeutic use , Enzyme Inhibitors/toxicity , Glucose-6-Phosphatase/antagonists & inhibitors , Hepatocytes/drug effects , Hypoglycemic Agents/toxicity , Indoles/toxicity , Liver/drug effects , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Phosphoenolpyruvate Carboxykinase (GTP)/antagonists & inhibitors , Prodrugs/toxicity , Quinolines/toxicity , Signal Transduction/drug effects
3.
J Mol Endocrinol ; 59(2): 151-169, 2017 08.
Article in English | MEDLINE | ID: mdl-28637808

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease with complicated pathogenesis and targeting gluconeogenesis inhibition is a promising strategy for anti-diabetic drug discovery. G protein-coupled receptors (GPCRs) are classified as distinct families by heterotrimeric G proteins, primarily including Gαs, Gαi and Gαq. Gαs-coupled GPCRs function potently in the regulation of hepatic gluconeogenesis by activating cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway and Gαi-coupled GPCRs exhibit inhibitory effect on adenylyl cyclase and reduce intracellular cAMP level. However, little is known about the regulation of Gαq-coupled GPCRs in hepatic gluconeogenesis. Here, small-molecule 2-(2,4-dimethoxy-3-methylphenyl)-7-(thiophen-2-yl)-9-(trifluoromethyl)-2,3-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-one (DMT) was determined to suppress hepatic glucose production and reduce mRNA levels of gluconeogenic genes. Treatment of DMT in db/db mice decreased fasting blood glucose and hemoglobin A1C (HbA1c) levels, while improved glucose tolerance and pyruvate tolerance. Mechanism study demonstrated that DMT-inhibited gluconeogenesis by regulating the Gαq/phospholipase C (PLC)/inositol-1,4,5-triphosphate receptor (IP3R)-mediated calcium (Ca2+)/calmodulin (CaM)/phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/forkhead box protein O1 (FOXO1) signaling pathway. To our knowledge, DMT might be the first reported small molecule able to suppress hepatic gluconeogenesis by regulating Gαq signaling, and our current work has also highlighted the potential of DMT in the treatment of T2DM.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gluconeogenesis/drug effects , Liver/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Animals , Calcium/metabolism , Calmodulin/metabolism , Forkhead Box Protein O1/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/pathology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Insulin/pharmacology , Liver/drug effects , Male , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Thiophenes/blood , Thiophenes/chemistry , Thiophenes/pharmacokinetics , Type C Phospholipases/metabolism
4.
Phytother Res ; 29(7): 1040-5, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25826437

ABSTRACT

(2'R)-2',3'-Dihydro-2'-(1-hydroxy-1-methylethyl)-2,6'-bibenzofuran-6,4'-diol (DHMB) is a natural compound extracted from Morus notabilis. It was found that DHMB acts as a competitive inhibitor against mushroom tyrosinase with a Ki value of 14.77 µM. Docking results further indicated that it could form strong interactions with one copper ion with a distance of 2.7 Å, suggesting the mechanism of inhibition might be due to chelating copper ions in the active site. Furthermore, melanin production in B16-F10 murine melanoma cells was significantly inhibited by DHMB in a concentration-dependent manner without cytotoxicity. The results of western blotting also showed that DHMB decreased 3-isobuty-1-methxlzanthine-induced mature tyrosinase expression. Taken together, these findings indicated that DHMB may be a new promising pigmentation-altering agent for agriculture, cosmetic, and therapeutic applications.


Subject(s)
Agaricales/enzymology , Benzofurans/chemistry , Melanins/biosynthesis , Melanoma, Experimental/metabolism , Monophenol Monooxygenase/antagonists & inhibitors , Animals , Cell Line, Tumor , Cyclic AMP/metabolism , Enzyme Inhibitors/chemistry , Mice , Molecular Docking Simulation , Morus/chemistry
5.
J Endocrinol ; 224(3): 327-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25572265

ABSTRACT

Impaired glucose-stimulated insulin secretion (GSIS) and increasing ß-cell death are two typical dysfunctions of pancreatic ß-cells in individuals that are destined to develop type 2 diabetes, and improvement of ß-cell function through GSIS enhancement and/or inhibition of ß-cell death is a promising strategy for anti-diabetic therapy. In this study, we discovered that the small molecule, N-(2-benzoylphenyl)-5-bromo-2-thiophenecarboxamide (BBT), was effective in both potentiating GSIS and protecting ß-cells from cytokine- or streptozotocin (STZ)-induced cell death. Results of further studies revealed that cAMP/PKA and long-lasting (L-type) voltage-dependent Ca(2) (+) channel/CaMK2 pathways were involved in the action of BBT against GSIS, and that the cAMP/PKA pathway was essential for the protective action of BBT on ß-cells. An assay using the model of type 2 diabetic mice induced by high-fat diet combined with STZ (STZ/HFD) demonstrated that BBT administration efficiently restored ß-cell functions as indicated by the increased plasma insulin level and decrease in the ß-cell loss induced by STZ/HFD. Moreover, the results indicated that BBT treatment decreased fasting blood glucose and HbA1c and improved oral glucose tolerance further highlighting the potential of BBT in anti-hyperglycemia research.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Homeostasis/drug effects , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Thiophenes/pharmacology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Hypoglycemic Agents/therapeutic use , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Streptozocin , Thiophenes/therapeutic use
6.
J Org Chem ; 79(2): 599-607, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24344740

ABSTRACT

Aphadilactones A-D (1-4), four diastereoisomers possessing an unprecedented carbon skeleton, were isolated from the Meliaceae plant Aphanamixis grandifolia. Their challenging structures and absolute configurations were determined by a combination of spectroscopic data, chemical degradation, fragment synthesis, experimental CD spectra, and ECD calculations. Aphadilactone C (3) with the 5S,11S,5'S,11'S configuration showed potent and selective inhibition against the diacylglycerol O-acyltransferase-1 (DGAT-1) enzyme (IC50 = 0.46 ± 0.09 µM, selectivity index > 217) and is the strongest natural DGAT-1 inhibitor discovered to date. In addition, compounds 1-4 showed significant antimalarial activities with IC50 values of 190 ± 60, 1350 ± 150, 170 ± 10, and 120 ± 50 nM, respectively.


Subject(s)
Antimalarials/pharmacology , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diterpenes/pharmacology , Enzyme Inhibitors/pharmacology , Meliaceae/chemistry , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Antimalarials/isolation & purification , Diacylglycerol O-Acyltransferase/metabolism , Dimerization , Diterpenes/chemistry , Diterpenes/isolation & purification , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Humans , Molecular Conformation , Parasitic Sensitivity Tests , Recombinant Proteins/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...