Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Biochem Pharmacol ; 197: 114933, 2022 03.
Article in English | MEDLINE | ID: mdl-35093393

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is becoming an increasingly serious disease worldwide. Unfortunately, no specific drug has been approved to treat NAFLD. Accumulating evidence suggests that lipotoxicity, which is induced by an excess of intracellular triacylglycerols (TAGs), is a potential mechanism underlying the ill-defined progression of NAFLD. Under physiological conditions, a balance is maintained between TAGs and free fatty acids (FFAs) in the liver. TAGs are catabolized to FFAs through neutral lipolysis and/or lipophagy, while FFAs can be anabolized to TAGs through an esterification reaction. However, in the livers of patients with NAFLD, lipophagy appears to fail. Reversing this abnormal state through several lipophagic molecules (mTORC1, AMPK, PLIN, etc.) facilitates NAFLD amelioration; therefore, restoring failed lipophagy may be a highly efficient therapeutic strategy for NAFLD. Here, we outline the lipophagy phases with the relevant important proteins and discuss the roles of lipophagy in the progression of NAFLD. Additionally, the potential candidate drugs with therapeutic value targeting these proteins are discussed to show novel strategies for future treatment of NAFLD.


Subject(s)
Autophagy/drug effects , Drug Delivery Systems/methods , Lipid Metabolism/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/physiology , Berberine/administration & dosage , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Fibroblast Growth Factors/administration & dosage , Humans , Lipid Metabolism/physiology , Lipolysis/drug effects , Lipolysis/physiology , Liver/drug effects , Mechanistic Target of Rapamycin Complex 1/administration & dosage , Transient Receptor Potential Channels/administration & dosage , Triglycerides/antagonists & inhibitors , Triglycerides/metabolism
2.
Neurotox Res ; 39(6): 1937-1945, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34792763

ABSTRACT

Acute ischemic stroke is a challenging disease that threatens the life of older people. Dysfunction of brain endothelial cells is reported to be involved in the pathogenesis of acute ischemic stroke. Ramelteon is a novel agonist of melatonin receptor developed for the treatment of insomnia. Recently, the promising protective effect of Ramelteon on brain injury has been widely reported. The present study aims to investigate the protective effect of Ramelteon against free fatty acid (FFA)-induced damages in brain vascular endothelial cells and the underlying mechanism. Firstly, we discovered that Ramelteon administration remarkably reversed the decreased cell viability, increased LDH release, activated oxidative stress, and excessive released inflammatory factors caused by FFAs. Secondly, Ramelteon extensively suppressed the attachment of U937 monocytes to bEnd.3 brain endothelial cells induced by FFAs. In addition, the elevated expression of E-selectin and the reduced expression of KLF2 induced by FFAs were pronouncedly alleviated by Ramelteon. Lastly, silencing of KLF2 abolished the protective effects of Ramelteon against FFA-induced expression of E-selectin and the attachment of U937 monocytes to bEnd.3 brain endothelial cells. In conclusion, Ramelteon mitigated FFA-induced attachment of monocytes to brain vascular endothelial cells by increasing the expression of KLF2 and reducing the expression of E-selectin.


Subject(s)
Brain/drug effects , Endothelial Cells/drug effects , Fatty Acids, Nonesterified/metabolism , Indenes/pharmacology , Monocytes/drug effects , Blotting, Western , Brain/pathology , Cell Death/drug effects , Endothelial Cells/pathology , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Nonesterified/antagonists & inhibitors , Humans , L-Lactate Dehydrogenase/metabolism , Monocytes/pathology , Real-Time Polymerase Chain Reaction , U937 Cells/drug effects
3.
Biochem Pharmacol ; 178: 114100, 2020 08.
Article in English | MEDLINE | ID: mdl-32540485

ABSTRACT

Short-chain acyl-CoA dehydrogenase (SCAD), the rate-limiting enzyme for fatty acid ß-oxidation, has a negative regulatory effect on pathological cardiac hypertrophy and fibrosis. Furthermore, flavin adenine dinucleotide (FAD) can enhance the expression and enzyme activity of SCAD. However, whether FAD can inhibit pathological cardiac hypertrophy and fibrosis remains unclear. Therefore, we observed the effect of FAD on pathological cardiac hypertrophy and fibrosis. FAD significantly inhibited PE-induced cardiomyocyte hypertrophy and AngII-induced cardiac fibroblast proliferation. In addition, FAD ameliorated pathological cardiac hypertrophy and fibrosis in SHR. FAD significantly increased the expression and enzyme activity of SCAD. Meanwhile, ATP content was increased, the content of free fatty acids and reactive oxygen species were decreased by FAD in vivo and in vitro. In addition, molecular dynamics simulations were also used to provide insights into the structural stability and dynamic behavior of SCAD. The results demonstrated that FAD may play an important structural role on the SCAD dimer stability and maintenance of substrate catalytic pocket to increase the expression and enzyme activity of SCAD. In conclusion, FAD can inhibit pathological cardiac hypertrophy and fibrosis through activating SCAD, which may be a novel effective treatment for pathological cardiac hypertrophy and fibrosis, thus prevent them from developing into heart failure.


Subject(s)
Butyryl-CoA Dehydrogenase/genetics , Cardiomegaly/prevention & control , Cardiotonic Agents/pharmacology , Fibroblasts/drug effects , Flavin-Adenine Dinucleotide/pharmacology , Gene Expression Regulation/drug effects , Adenosine Triphosphate/biosynthesis , Animals , Binding Sites , Butyryl-CoA Dehydrogenase/metabolism , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Proliferation/drug effects , Energy Metabolism/drug effects , Energy Metabolism/genetics , Enzyme Stability , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Heart Failure/prevention & control , Male , Molecular Dynamics Simulation , Myocardium/enzymology , Myocardium/pathology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Rats , Rats, Inbred SHR , Rats, Wistar , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
4.
Psychopharmacology (Berl) ; 235(8): 2335-2347, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29931581

ABSTRACT

RATIONALE: Depression and anxiety can cause the development of chronic pain. However, the mechanism of chronic pain induced by emotional dysfunction is still unknown. Previously, we demonstrated that the G protein-coupled receptor 40/free fatty acid receptor 1 (GPR40/FFAR1) signaling in the brain is related to regulation of both pain and emotion. In the present study, we proved that the role of GPR40/FFAR1 signaling in the development of chronic pain is induced by emotional dysfunction. RESULTS: Repeated social defeat (SD)-stressed mice showed the impairment of social interaction and anxiety behavior. These mice also caused pain prolongation after paw-incision comparison with non-SD mice. This pain prolongation was markedly continued by infusion of the GPR40/FFAR1 antagonist, GW1100 during SD stress but not non-SD stress. Although, infusion of the GW1100 during SD stress did not cause deterioration of the emotional behavior. Furthermore, GW1100-treated SD-mice showed strong tendency of emotional dysfunction after paw incision. CONCLUSION: Our findings indicate that the dysfunction of fatty acids-GPR40/FFAR1 signaling in the brain underlying stress condition might be related to the development of chronic pain.


Subject(s)
Chronic Pain/metabolism , Fatty Acids, Nonesterified/metabolism , Interpersonal Relations , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Stress, Psychological/metabolism , Animals , Benzoates/administration & dosage , Brain/drug effects , Brain/metabolism , Chronic Pain/psychology , Fatty Acids, Nonesterified/antagonists & inhibitors , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pyrimidines/administration & dosage , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/psychology
5.
Cell Physiol Biochem ; 42(4): 1635-1644, 2017.
Article in English | MEDLINE | ID: mdl-28738323

ABSTRACT

BACKGROUND: This study aimed to investigate whether exogenous hydrogen sulfide (H2S) can protect the RAW264.7 macrophages against the inflammation induced by free fatty acids (FFA) by blunting NLRP3 inflammasome activation via a specific TLR4/NF-κB pathway. METHODS: RAW264.7 macrophages were exposed to increasing concentrations of FFA for up to 3 days to induce FFA-induced inflammation. The cells were pretreated with NaHS (a donor of H2S) before exposure to FFA. Cell viability, cell apoptosis, TLR4, NF-κB, NLRP3 inflammasome, IL-1ß, IL-18 and cleaved caspase-3 expression were measured by a combination of MTT assay, ELISA, and immunoblotting. RESULTS: H2S attenuated FFA-induced cell apoptosis, and reduced the expression of NLRP3, ASC, pro-caspase-1, caspase-1, IL- 1ß, IL-18 and caspase-3. In addition, H2S inhibited the FFA-induced activation of TLR4 and NF-κB. Furthermore, NLRP3 inflammasome activation was regulated by the TLR4 and NF-κB pathway. CONCLUSION: The present study demonstrated for the first time that H2S appears to suppress FFA-induced macrophage inflammation and apoptosis by inhibiting the TLR4/ NF-κB pathway and its downstream NLRP3 inflammasome activation. Thus H2S might possess potential in the treatment of diseases resulting from FFA overload like insulin resistance and type diabetes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Fatty Acids, Nonesterified/antagonists & inhibitors , Hydrogen Sulfide/pharmacology , Macrophages/drug effects , Sulfides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/immunology , Cell Line , Cell Survival/drug effects , Fatty Acids, Nonesterified/pharmacology , Gene Expression Regulation , Hydrogen Sulfide/chemistry , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/cytology , Macrophages/immunology , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Sulfides/chemistry , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
6.
Biochem Pharmacol ; 138: 140-149, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28522407

ABSTRACT

Pancreatic ß-cell lipotoxicity is a central feature of the pathogenesis of type 2 diabetes. To study the mechanism by which fatty acids cause ß-cell death and develop novel approaches to prevent it, a high-throughput screen on the ß-cell line INS1 was carried out. The cells were exposed to palmitate to induce cell death and compounds that reversed palmitate-induced cytotoxicity were ascertained. Hits from the screen were analyzed by an increasingly more stringent testing funnel, ending with studies on primary human islets treated with palmitate. MAP4K4 inhibitors, which were not part of the screening libraries but were ascertained by a bioinformatics analysis, and the endocannabinoid anandamide were effective at inhibiting palmitate-induced apoptosis in INS1 cells as well as primary rat and human islets. These targets could serve as the starting point for the development of therapeutics for type 2 diabetes.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Computational Biology , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Female , High-Throughput Screening Assays , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Small Molecule Libraries , Tissue Culture Techniques
7.
Diabetes Obes Metab ; 19(9): 1306-1311, 2017 09.
Article in English | MEDLINE | ID: mdl-28304146

ABSTRACT

In view of the occurrence of diabetic ketoacidosis associated with the use of sodium-glucose transport protein-2 inhibitors in patients with type 1 diabetes (T1DM) and the relative absence of this complication in patients treated with liraglutide in spite of reductions in insulin doses, we investigated the effect of liraglutide on ketogenesis. Twenty-six patients with inadequately controlled T1DM were randomly divided into 2 groups of 13 patients each. After an overnight fast, patients were injected, subcutaneously, with either liraglutide 1.8 mg or with placebo. They were maintained on their basal insulin infusion and were followed up in our clinical research unit for 5 hours. The patients injected with placebo maintained their glucose and glucagon concentrations without an increase, but there was a significant increase in free fatty acids (FFA), acetoacetate and ß-hydoxybutyrate concentrations. In contrast, liraglutide significantly reduced the increase in FFA, and totally prevented the increase in acetoacetate and ß-hydroxybutyrate concentrations while suppressing glucagon and ghrelin concentrations. Thus, a single dose of liraglutide is acutely inhibitory to ketogenesis.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon/antagonists & inhibitors , Hypoglycemic Agents/therapeutic use , Ketone Bodies/antagonists & inhibitors , Lipolysis/drug effects , Liraglutide/therapeutic use , Adult , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Double-Blind Method , Drug Resistance , Drug Therapy, Combination , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/blood , Female , Ghrelin/antagonists & inhibitors , Ghrelin/blood , Glucagon/blood , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Injections, Subcutaneous , Insulin/administration & dosage , Insulin/therapeutic use , Insulin Infusion Systems , Ketone Bodies/biosynthesis , Ketone Bodies/blood , Liraglutide/administration & dosage , Male , Middle Aged
8.
J Cardiovasc Pharmacol ; 68(6): 465-472, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27606935

ABSTRACT

Puerarin, a type of isoflavone, was shown to have multiple protective effects on myocardial injury. The objective of this study was to investigate the role of puerarin in the progression of lipotoxic cardiomyopathy. Primary cardiomyocytes were isolated from FATP1 transgenic (Tg) mice with lipotoxic cardiomyopathy, and various concentrations of puerarin were used to incubate with the cardiomyocytes. Our results showed low-dose puerarin (≤20 µM) treatment increased the cell viability and decreased the accumulation of free fatty acid (FFA). The data on enzyme-linked immunosorbent assay indicated that 15 µM puerarin treatment greatly increased Na-K-ATPase activity and decreased C-reactive protein secretion, thus suppressing the expression of CD36, a key contributor to the FFA accumulation. Additionally, low-dose puerarin (≤100 mg/kg body weight) administration improved Na-K-ATPase activity. Our data on serum analysis and histological detection in vivo indicated that systemic inflammation, CD36-induced lipid infiltration, and cardiomyocyte apoptosis were markedly alleviated in Tg mice injected with 90 mg/kg dose of puerarin. Finally, the uptake rates of H-palmitate and C-glucose were monitored on ex vivo working hearts that were obtained from wild-type (WT), Tg-control, and Tg-puerarin mice. Compared with WT hearts, Tg hearts displayed a significant decrease in Na/K-ATPase activity and glucose consumption rate and an increase in palmitate uptake rate and FFA accumulation. In Tg-puerarin hearts, Na/K-ATPase activity and glucose consumption rate were significantly rescued, and palmitate uptake and FFA accumulation were sharply suppressed. In conclusion, low-dose puerarin suppressed Na-K-ATPase-mediated CD36 expression and systemic inflammation and alleviated cardiac lipotoxicity in vitro and in vivo.


Subject(s)
CD36 Antigens/antagonists & inhibitors , Fatty Acids, Nonesterified/antagonists & inhibitors , Isoflavones/pharmacology , Myocytes, Cardiac/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Vasodilator Agents/pharmacology , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Fatty Acids, Nonesterified/metabolism , Gene Expression , Inflammation/drug therapy , Inflammation/metabolism , Isoflavones/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Vasodilator Agents/therapeutic use
9.
J Cardiovasc Pharmacol ; 67(1): 39-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26322921

ABSTRACT

CD36 is a key transporter involved in fatty acid (FA) uptake and contributes to the accumulation of FA in cardiomyocytes. The objective of this study was to investigate the role of ouabain, a glycoside regulator of Na(+)/K(+)-ATPase, in the regulation of CD36 expression and FA accumulation. FATP1 transgenic (Tg) mice with lipotoxic cardiomyopathy displayed significantly increased cardiac CD36 expression and free fatty acid accumulation. The data on enzyme-linked immunosorbent assay showed that endogenous ouabain was decreased in the serum of Tg mice versus wild-type mice. CD36 expression and free fatty acid accumulation in their primary cardiomyocytes were abated by treatment with 0.15-0.30 µM ouabain. CD36 expression was suppressed by 0.2 µM ouabain treatment, and the suppression was rescued by C-reactive protein. CD36 expression and free fatty acid accumulation in the heart were markedly reduced in Tg mice injected with 30 or 40 ng of ouabain (P < 0.01). Obvious fatty infiltration was found in noninjected Tg mice but not in the mice injected with 40 ng of ouabain. In conclusion, low-dose exogenous ouabain increased Na(+)/K(+)-ATPase activity, suppressed C-reactive protein-mediated CD36 expression, and alleviated murine cardiac lipotoxicity in vitro and in vivo.


Subject(s)
CD36 Antigens/biosynthesis , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/toxicity , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Ouabain/administration & dosage , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Fatty Acids, Nonesterified/antagonists & inhibitors , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Biochem Cell Biol ; 93(6): 566-73, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26335364

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) describes a spectrum of lesions ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). The excess influx of fatty acids (FAs) into the liver is recognized as a main cause of simple steatosis formation and progression to NASH. Recently, administration of lactoferrin (LF), a glycoprotein present in milk, was suggested to prevent NAFLD development. However, the effect of LF on the contribution of FA to NAFLD development remains unclear. In this study, the effects of LF on FA mixture (FAm)-induced lipotoxicity using human hepatocarcinoma G2 cells were assessed. FAm significantly decreased cell viability and increased intracellular lipid accumulation, whereas LF significantly recovered cell viability without affecting lipid accumulation. FAm-induced lactic dehydrogenase (LDH) and caspase-3/7 activities were significantly decreased by LF and SP600125, a c-Jun N-terminal kinase (JNK) specific inhibitor. We also found that LF added to FAm-treated cells induced Akt phosphorylation, which contributed to inhibition of JNK signaling pathway-dependent apoptosis. Akt inhibitor VIII, an allosteric Akt inhibitor, significantly attenuated the effect of LF on LDH activity and abrogated the ones on cell viability and caspase-3/7 activity. In summary, the present study has revealed that LF has a protective effect on FAm-induced lipotoxicity in a HepG2 model of NAFLD and identified the activation of the Akt signaling pathway as a possibly major mechanism.


Subject(s)
Lactoferrin/pharmacology , Lipid Metabolism/drug effects , Lipotropic Agents/pharmacology , Liver/drug effects , MAP Kinase Signaling System/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Proto-Oncogene Proteins c-akt/agonists , Animals , Anthracenes/pharmacology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Cattle , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Lactoferrin/antagonists & inhibitors , Lactoferrin/chemistry , Lactoferrin/metabolism , Lipotropic Agents/chemistry , Lipotropic Agents/metabolism , Liver/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/pharmacology
11.
Mol Nutr Food Res ; 59(8): 1443-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25943029

ABSTRACT

SCOPE: Resveratrol (RSV), a natural polyphenol, has been reported to attenuate nonalcoholic fatty liver disease (NAFLD); however, its underlying mechanism is unclear. Autophagy was recently identified as a critical protective mechanism during NAFLD development. Therefore, we investigated the role of autophagy in the beneficial effects of RSV on hepatic steatosis. METHODS AND RESULTS: Via Oil red O staining, triglyceride, and ß-hydroxybutyrate detection, we found that RSV decreased palmitate-induced lipid accumulation and stimulated fatty acid ß-oxidation in hepatocytes. Based on Western blot assay, confocal microscopy and transmission electron microscopy, we found that RSV induced autophagy in hepatocytes, whereas autophagy inhibition markedly abolished RSV-mediated hepatic steatosis improvement. Moreover, RSV increased cAMP levels and the levels of SIRT1 (sirtuin 1), pPRKA (phosphorylated protein kinase A), and pAMPK (phosphorylated AMP-activated protein kinase), as well as SIRT1 activity in HepG2 cells. Incubation with inhibitors of AC (adenylyl cyclase), PRKA, AMPK, SIRT1, or with AC, PRKA, AMPK, or SIRT1 siRNA abolished RSV-mediated autophagy. Similar results were obtained in mice with hepatic steatosis. CONCLUSION: RSV improved hepatic steatosis partially by inducing autophagy via the cAMP-PRKA-AMPK-SIRT1 signaling pathway, which provides new evidence regarding RSV's effects on NAFLD treatment.


Subject(s)
Antioxidants/therapeutic use , Autophagy , Cyclic AMP/agonists , Dietary Supplements , Liver/metabolism , Non-alcoholic Fatty Liver Disease/diet therapy , Second Messenger Systems , Stilbenes/therapeutic use , Adenylyl Cyclases/chemistry , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Antioxidants/metabolism , Autophagy/drug effects , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Mice, 129 Strain , Microscopy, Electron, Transmission , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , RNA Interference , Resveratrol , Second Messenger Systems/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/chemistry , Sirtuin 1/genetics , Sirtuin 1/metabolism , Stilbenes/metabolism
12.
Endocr J ; 61(12): 1213-20, 2014.
Article in English | MEDLINE | ID: mdl-25231694

ABSTRACT

We evaluated the effect of sitagliptin on glycemic control, endogenous insulin secretion, and beta cell function in Japanese patients with type 2 diabetes mellitus (T2DM) receiving a combination of oral antidiabetics and basal insulin analog glargine (basal-supported oral therapy [BOT]). Twenty-one patients showing inadequate glycemic control with BOT were given dipeptidylpeptidase-4 inhibitor (DPP-4I) sitagliptin at 50 mg/day for 12 weeks. Clinical markers of glycemic control, HbA1c, glycated albumin (GA), and 1,5-anhydroglucitol (1,5-AG), were measured before and 4 and 12 weeks after the start of sitagliptin. A 2-hour morning meal test was performed upon enrollment and at 12 weeks, and plasma glucose (PG), serum C-peptide, and plasma intact proinsulin (PI) were measured. HbA1c, GA, and 1,5-AG at 4 and 12 weeks were significantly improved over enrollment levels. The area under the PG concentration curve (AUC-PG) during the meal test at 12 weeks was significantly reduced (from 350 ± 17 mg ï½¥ hr/dL before sitagliptin treatment to 338 ± 21 mg ï½¥ hr/dL [mean ± SE], P < 0.05,); the AUC-C-peptide was unchanged (from 3.4 ± 0.4 ng ï½¥ hr/mL to 3.6 ± 0.5 ng ï½¥ hr/mL). However, both fasting and 2-hour PI/C-peptide ratios at 12 weeks were significantly decreased (from 13.3 ± 2.3 to 11.1 ± 2.0 [P < 0 .05] and from 9.5 ± 1.6 to 5.3 ± 0.9 [P < 0.01], respectively). Adding sitagliptin to BOT in Japanese T2DM patients appears to improve glycemic control without increasing endogenous insulin secretion and to reduce fasting and 2-hour postprandial PI/C-peptide ratios.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Drug Resistance , Hyperglycemia/prevention & control , Insulin-Secreting Cells/drug effects , Proinsulin/metabolism , Pyrazines/therapeutic use , Triazoles/therapeutic use , Administration, Oral , Aged , Algorithms , Biomarkers/blood , C-Peptide/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/ethnology , Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Drug Monitoring , Drug Therapy, Combination , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/blood , Female , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Insulin Glargine , Insulin, Long-Acting/administration & dosage , Insulin, Long-Acting/therapeutic use , Insulin-Secreting Cells/metabolism , Japan , Male , Proinsulin/blood , Pyrazines/administration & dosage , Sitagliptin Phosphate , Triazoles/administration & dosage
13.
Mol Nutr Food Res ; 58(10): 2053-65, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25044948

ABSTRACT

SCOPE: To investigate whether docosahexaenoic acid (DHA) could inhibit linoleic acid (LA) induced monocyte chemoattractant protein (MCP)-1 expression in human retinal pigment epithelial (RPE) cells. METHODS AND RESULTS: ARPE-19 cells were pretreated with DHA and then exposed to LA. The expression of MCP-1 and PPARγ was examined using RT-PCR and Western blot analysis. LA at 10, 25, or 50 µM induced expression of MCP ARPE-19 cells in a dose-dependent manner (p < 0.05). DHA at 50 and 100 µM effectively inhibited LA-induced MCP-1 expression and production (p < 0.05) and NF-κB activation. In addition, the culture medium from LA-stimulated ARPE-19 cells could induce tube formation in choroidal endothelial cells (RF6A), whereas 100 µM DHA inhibited tube formation. DHA at 100 µM increased the expression and activity of PPARγ (p < 0.05). Pretreatment with PPARγ inhibitor (GW9662) abolished the inhibitory effect of DHA (100 µM) on LA-induced IκB degradation, p65 translocation, and MCP-1 expression in ARPE-19 cells (p < 0.05), as well as tube formation in RF6A. CONCLUSION: DHA reduced LA-induced MCP-1 expression via a PPARγ- and NF-κB-dependent pathway in ARPE-19 cells. These results suggest the molecular mechanisms underlying the beneficial effects of increased consumption of DHA and reduced consumption of LA on age-related macular degeneration.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Docosahexaenoic Acids/metabolism , NF-kappa B/metabolism , PPAR gamma/metabolism , Retinal Pigment Epithelium/metabolism , Signal Transduction , Up-Regulation , Anilides/pharmacology , Arachidonic Acid/adverse effects , Arachidonic Acid/antagonists & inhibitors , Cell Line , Chemokine CCL2/agonists , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Choroid/drug effects , Choroid/immunology , Choroid/metabolism , Choroidal Neovascularization/etiology , Choroidal Neovascularization/immunology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/prevention & control , Culture Media, Conditioned/metabolism , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/therapeutic use , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Humans , Linoleic Acid/adverse effects , Linoleic Acid/antagonists & inhibitors , Macular Degeneration/etiology , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/prevention & control , NF-kappa B/genetics , Osmolar Concentration , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Promoter Regions, Genetic/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/immunology , Signal Transduction/drug effects , Up-Regulation/drug effects
14.
J Diabetes Res ; 2014: 391476, 2014.
Article in English | MEDLINE | ID: mdl-24804268

ABSTRACT

This study aimed to explore the effect of angiotensin (1-7) (Ang (1-7)) on palmitate-induced apoptosis in islet endothelial cells and the mechanism of action. MS-1 cells were treated with palmitate in the presence or absence of Ang (1-7). The percentage of apoptotic cells was determined by DNA fragmentation and flow cytometry. Reactive oxygen species (ROS) production was measured using a Reactive Oxygen Species Assay Kit. Expression of AKT, eNOS, C-Jun N-terminal kinase (JNK), and p38 was detected by western blotting. Compared with palmitate treated group, palmitate-induced apoptosis was decreased in MS-1 cells which were preincubated with Ang (1-7) (P < 0.05). Palmitate decreased the phosphorylation of AKT and eNOS, and Ang (1-7) increased the phosphorylation of these kinases (P < 0.05), with a concomitant reduction in MS-1 cells apoptosis. Ang (1-7) also inhibited the palmitate-induced ROS production and attenuated the apoptosis-related signaling molecule JNK and p38 activation (all P < 0.05). PI3K/AKT, eNOS, p38 MAPK, and JNK inhibitors blocked the antilipoapoptosis of Ang (1-7) (all P < 0.05). Our findings suggest that Ang (1-7) reduces palmitate-induced islet endothelial cells apoptosis. AKT/eNOS/NO signaling and JNK and p38 pathway are involved in the Ang (1-7)-mediated modulation of islet endothelial cells lipoapoptosis.


Subject(s)
Angiotensin I/pharmacology , Apoptosis/drug effects , Endothelium, Vascular/drug effects , Islets of Langerhans/drug effects , MAP Kinase Signaling System/drug effects , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-akt/agonists , Angiotensin I/antagonists & inhibitors , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Antihypertensive Agents/antagonists & inhibitors , Antihypertensive Agents/pharmacology , Cell Line, Transformed , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/chemistry , MAP Kinase Kinase 4/metabolism , Mice , Microvessels/drug effects , Microvessels/enzymology , Microvessels/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/chemistry , Palmitic Acid/antagonists & inhibitors , Palmitic Acid/metabolism , Peptide Fragments/antagonists & inhibitors , Protective Agents/chemistry , Protective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Arch Pharm Res ; 37(9): 1169-76, 2014.
Article in English | MEDLINE | ID: mdl-24633463

ABSTRACT

Non-alcoholic fatty liver disease is associated with inhibited AMP-activated kinase (AMPK) and activation of sterol regulatory element binding protein 1 (SREBP-1). AMPK phosphorylation inhibits SREBP-1, a major transcription factor of de novo lipogenesis, by inhibiting the liver X receptor (LXR) or by direct phosphorylation. Resveratrol, a polyphenol, has regulatory effects on hepatic lipid metabolism as a potent AMPK activator. In this study, we evaluated the anti-steatogenic effects of resveratrol and its derivatives and identified the molecular mechanism in vitro and in vivo. Resveratrol and its derivatives decreased lipid accumulation by free fatty acids (FFA mixture; 0.5 mM, oleic acid:palmitic acid = 2: 1) in H4IIEC3 cells. Synthesized derivatives of resveratrol had lower cytotoxicity than the parental molecule with similar potency. SY-102 suppressed SREBP-1 maturation by T0901317, an LXR agonist, and decreased SRE luciferase activity and the mRNA levels of lipogenic genes. Inhibition of AMPK by pre-treatment with compound C completely blocked the effects of SY-102. To evaluate their efficacy in vivo, mice were fed a high-fat diet for 5 days, and resveratrol or SY-102 was administered orally for the last 2 days. Oral administration of the SY-102 increased AMPK phosphorylation, followed by reduced hepatic triglyceride accumulation to a similar extent as resveratrol. These data demonstrate that SY-102, a synthesized derivative of resveratrol, might provide a promising therapeutic effect against fatty liver disease.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hepatocytes/drug effects , Lipotropic Agents/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Stilbenes/therapeutic use , AMP-Activated Protein Kinases/chemistry , Animals , Cell Line , Cell Survival/drug effects , Enzyme Activation/drug effects , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Gene Expression Regulation/drug effects , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Lipotropic Agents/adverse effects , Lipotropic Agents/pharmacology , Male , Methylation , Mice, Inbred ICR , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Random Allocation , Rats , Resveratrol , Specific Pathogen-Free Organisms , Stilbenes/adverse effects , Stilbenes/chemistry , Stilbenes/pharmacology
16.
Plant Signal Behav ; 9(7): e28982, 2014.
Article in English | MEDLINE | ID: mdl-25763484

ABSTRACT

The fungal pathogen Fusarium graminearum is the causal agent of Fusarium head blight (FHB); a devastating crop disease resulting in heavy yield losses and grain contamination with mycotoxins. We recently showed that the secreted lipase FGL1, a virulence factor of F. graminearum, targets plant defense-related callose biosynthesis during wheat head infection. This effector-like function is based on a FGL1-mediated release of polyunsaturated free fatty acids (FFA) that can inhibit callose synthase activity. The importance of FGL1 in successful wheat head colonization was demonstrated in FGL1 disruption mutants (Δfgl1), where infection was restricted to directly inoculated spikelets and accompanied by strong callose deposition in the spikelet's phloem. The application of polyunsaturated FFA to Δfgl1-infected spikelets prevented callose deposition in the phloem and partially restored wheat head colonization.   The comparative analysis of 3 wheat cultivars revealed that the level of resistance to FHB correlated with resistance to FFA-dependent inhibition of callose biosynthesis. Therefore, resistance of callose biosynthesis to FFA inhibition might be used as marker and/or direct target in the breeding of FHB-resistant wheat cultivars.


Subject(s)
Disease Resistance/genetics , Fatty Acids, Nonesterified/metabolism , Fusarium/pathogenicity , Glucans/biosynthesis , Glucosyltransferases/antagonists & inhibitors , Phenotype , Triticum , Breeding , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Unsaturated/metabolism , Fusarium/metabolism , Inflorescence , Lipase/metabolism , Mycotoxins/metabolism , Phloem/microbiology , Plant Diseases/microbiology , Plant Proteins/metabolism , Species Specificity , Triticum/genetics , Triticum/metabolism , Triticum/microbiology , Virulence Factors/metabolism
17.
CNS Neurosci Ther ; 19(4): 252-61, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23521913

ABSTRACT

AIMS: The damage of human brain vascular endothelial cells (HBVECs) is the key pathogenesis of diabetes-associated cerebral vascular complications. The aim of this study was to elucidate the effects of glutathione (GSH) on free fatty acids (FFAs)-induced HBVECs apoptosis, oxidative stress, and the involved possible signaling pathway. METHODS: After culturing HBVECs for 72 h with GSH and FFAs, we determined cell proliferation by CCK8, detected apoptosis by caspase-3 and Annexin V-FITC/PI staining, and judged oxygen stress by determining the reactive oxygen species (ROS) and the mitochondrial membrane potential (MMP). We investigated whether the Akt pathway was involved in FFAs-induced signaling pathway alteration and whether GSH influenced the above effects. RESULTS: After being cultured in 200 µM FFAs for 72 h, the HBVECs proliferation significantly decreased; HBVECs apoptosis increased; the ROS levels increased; and the HBVECs MMP subsequently decreased. FFAs induced a significant decrease in phosphorylated active Akt. These alterations were obviously prevented when 1 mM GSH was added to culture medium containing FFAs, and the above effects of GSH were blocked by Akt inhibitor. CONCLUSION: GSH may prevent FFAs-induced HBVECs damage, oxidative stress, and apoptosis through activating the Akt pathway.


Subject(s)
Apoptosis/physiology , Endothelial Cells/physiology , Fatty Acids, Nonesterified/toxicity , Glutathione/physiology , Oxidative Stress , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Brain/cytology , Brain/metabolism , Cell Survival/physiology , Cells, Cultured , Endothelial Cells/metabolism , Fatty Acids, Nonesterified/antagonists & inhibitors , Humans , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
18.
J Physiol ; 591(11): 2897-909, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23529132

ABSTRACT

Free fatty acids (FFAs) have been implicated in the pathogenesis of insulin resistance. Reducing plasma FFA concentration in obese and type 2 diabetic (T2DM) subjects improves insulin sensitivity. However, the molecular mechanism by which FFA reduction improves insulin sensitivity in human subjects is not fully understood. In the present study, we tested the hypothesis that pharmacological FFA reduction enhances insulin action by reducing local (muscle) inflammation, leading to improved insulin signalling. Insulin-stimulated total glucose disposal (TGD), plasma FFA species, muscle insulin signalling, IBα protein, c-Jun phosphorylation, inflammatory gene (toll-like receptor 4 and monocyte chemotactic protein 1) expression, and ceramide and diacylglycerol (DAG) content were measured in muscle from a group of obese and T2DM subjects before and after administration of the antilipolytic drug acipimox for 7 days, and the results were compared to lean individuals. We found that obese and T2DM subjects had elevated saturated and unsaturated FFAs in plasma, and acipimox reduced all FFA species. Acipimox-induced reductions in plasma FFAs improved TGD and insulin signalling in obese and T2DM subjects. Acipimox increased IBα protein (an indication of decreased IB kinase-nuclear factor B signalling) in both obese and T2DM subjects, but did not affect c-Jun phosphorylation in any group. Acipimox also decreased inflammatory gene expression, although this reduction only occurred in T2DM subjects. Ceramide and DAG content did not change. To summarize, pharmacological FFA reduction improves insulin signalling in muscle from insulin-resistant subjects. This beneficial effect on insulin action could be related to a decrease in local inflammation. Notably, the improvements in insulin action were more pronounced in T2DM, indicating that these subjects are more susceptible to the toxic effect of FFAs.


Subject(s)
Chemokine CCL2/metabolism , Fatty Acids, Nonesterified/blood , Hypolipidemic Agents/pharmacology , Insulin/metabolism , Muscle, Skeletal/metabolism , Pyrazines/pharmacology , Administration, Oral , Adult , Case-Control Studies , Ceramides/metabolism , Chemokine CCL2/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diglycerides/metabolism , Fatty Acids, Nonesterified/antagonists & inhibitors , Female , Glucose/metabolism , Humans , Hypolipidemic Agents/administration & dosage , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Insulin/genetics , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Middle Aged , Muscle, Skeletal/drug effects , Obesity/blood , Obesity/metabolism , Pyrazines/administration & dosage , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
19.
J Clin Endocrinol Metab ; 97(9): 3277-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22761459

ABSTRACT

BACKGROUND/AIM: We tested the hypothesis that a persistent reduction in free fatty acid (FFA) levels improves cardiac function and systemic insulin sensitivity via a reduction in the myocardial and skeletal muscle adiposities and a modulation in adipokine release. METHODS: Study subjects (body mass index 22-30 kg/m(2), 57 ± 3 yr old) underwent magnetic resonance imaging and spectroscopy to measure the cardiac function and the amounts of fat inside and around the myocardium and skeletal muscle, before (n = 10) and after acute (n = 8) and 1 wk (n = 7, one excluded from analysis) lowering of circulating FFA by acipimox. Circulating adipokines (leptin, adiponectin, resistin, TNFα, IL-6, IL-8, plasminogen activator inhibitor-I, macrophage chemoattractant protein-1) were measured. RESULTS: The ejection fraction (62 ± 2 vs. 56 ± 1%, P = 0.0035), cardiac output (6.6 ± 0.3 vs. 5.5 ± 0.2 liters/min, P = 0.0018), and forward work (708 ± 49 vs. 539 ± 44 mm Hg × liters/min, P = 0.018) were significantly lower after 1 wk of FFA lowering. In the six subjects undergoing all sessions, the stroke and end-diastolic volumes were also reduced, insulin sensitivity was increased by 33%, and adiponectinemia was decreased (-26%, P = 0.03). No change in intracellular cardiac and skeletal muscle triglyceride levels was observed. Metabolic changes correlated with the lowering of FFA. The reduction in cardiac function was related with changes in glycemia and insulin sensitivity, whereas the deflection in left ventricular work was correlated with the decline in FFA, lipid, and blood pressure levels. CONCLUSIONS: A 1-wk FFA depletion suppressed cardiac function and improved insulin sensitivity. Intracellular triglyceride deposits in the heart and skeletal muscle played no role in the observed changes. Our data show that FFA participate in the physiological regulation of adipokine levels.


Subject(s)
Adiponectin/metabolism , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/blood , Heart/drug effects , Hypolipidemic Agents/pharmacology , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Muscle, Skeletal/metabolism , Myocardium/metabolism , Pyrazines/pharmacology , Adipokines/metabolism , Adult , Aged , Female , Heart Function Tests , Hemodynamics/drug effects , Hemodynamics/physiology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/chemistry , Muscle, Skeletal/drug effects , Myocardium/chemistry , Stroke Volume/drug effects , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
20.
J Huazhong Univ Sci Technolog Med Sci ; 31(2): 185-189, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21505982

ABSTRACT

This study investigated the effects and molecular mechanisms of genistein in improving insulin resistance induced by free fatty acids (FFAs) in HepG2 hepatocytes. A model of insulin resistance in HepG2 cells was established by adding palmitic acid (0.5 mmol/L) to the culture medium and the cells were treated by genistein. Glucose consumption of HepG2 cells was determined by glucose oxidase method. The levels of c-jun N-terminal kinase (JNK) phosphorylation, insulin receptor substrate-1 (IRS-1) Ser307 phosphorylation, JNK, IRS-1, phosphatidylinositol-3-kinase p85 (PI-3K p85) and glucose transporter 1 (GLUT1) proteins were detected by Western blotting. The results showed that after the treatment with palmitic acid for 24 h, the insulin-stimulated glucose transport in HepG2 cells was inhibited, and the glucose consumption was substantially reduced. Meanwhile, the expressions of IRS-1, PI-3K p85 protein and GLUT1 were obviously reduced, while the levels of JNK phosphorylation and IRS-1 Ser307 phosphorylation and the expression of JNK protein were significantly increased, as compared with cells of normal control. However, the aforementioned indices, which indicated the existence of insulin resistance, were reversed by genistein at 1-4 µmol/L in a dose-dependent manner. It was concluded that insulin resistance induced by FFAs in HepG2 hepatocytes could be improved by genistein. Genistein might reverse FFAs-induced insulin resistance in HepG2 cells by targeting JNK.


Subject(s)
Fatty Acids, Nonesterified/antagonists & inhibitors , Genistein/pharmacology , Hepatocytes/drug effects , Insulin Resistance , MAP Kinase Kinase 4/metabolism , Fatty Acids, Nonesterified/pharmacology , Hep G2 Cells , Hepatocytes/metabolism , Humans , Phytoestrogens/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...