Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Toxicol Appl Pharmacol ; 252(3): 237-49, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21338618

ABSTRACT

Development of a preventive strategy against tubular damage associated with proteinuria is of great importance. Recently, free fatty acid (FFA) toxicities accompanying proteinuria were found to be a main cause of tubular damage, which was aggravated by insufficiency of peroxisome proliferator-activated receptor alpha (PPARα), suggesting the benefit of PPARα activation. However, an earlier study using a murine acute tubular injury model, FFA-overload nephropathy, demonstrated that high-dose treatment of PPARα agonist (0.5% clofibrate diet) aggravated the tubular damage as a consequence of excess serum accumulation of clofibrate metabolites due to decreased kidney elimination. To induce the renoprotective effects of PPARα agonists without drug accumulation, we tried a pretreatment study using low-dose clofibrate (0.1% clofibrate diet) using the same murine model. Low-dose clofibrate pretreatment prevented acute tubular injuries without accumulation of its metabolites. The tubular protective effects appeared to be associated with the counteraction of PPARα deterioration, resulting in the decrease of FFAs influx to the kidney, maintenance of fatty acid oxidation, diminution of intracellular accumulation of undigested FFAs, and attenuation of disease developmental factors including oxidative stress, apoptosis, and NFκB activation. These effects are common to other fibrates and dependent on PPARα function. Interestingly, however, clofibrate pretreatment also exerted PPARα-independent tubular toxicities in PPARα-null mice with FFA-overload nephropathy. The favorable properties of fibrates are evident when PPARα-dependent tubular protective effects outweigh their PPARα-independent tubular toxicities. This delicate balance seems to be easily affected by the drug dose. It will be important to establish the appropriate dosage of fibrates for treatment against kidney disease and to develop a novel PPARα activator that has a steady serum concentration regardless of kidney dysfunction.


Subject(s)
Clofibrate/pharmacology , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/toxicity , Hypolipidemic Agents/pharmacology , Kidney Tubules/drug effects , PPAR alpha/metabolism , Proteinuria/metabolism , Animals , Female , Histocytochemistry , Kidney Tubules/metabolism , Mice , PPAR alpha/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms
10.
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
11.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(1): 48-51, 2011 Jan.
Article in Zh | MEDLINE | ID: mdl-21355300

ABSTRACT

OBJECTIVE: To investigate whether Astragalus membranaceus (AM) can protect endothelium-dependent vasodilatation (EDV) function of aorta from the damage induced by high level of free fatty acid (FFA). METHODS: Ten male SD rats, 8 weeks old and 250-300 g in weight, were sacrificed and thoracic aorta were harvested. Aorta rings incubated in organ baths were divided into three groups, Control group, FFA group and FFA+ AM group. The control group was incubated in 20 mL Krebs-Henseleit solution; the FFA group was incubated in 20 mL KH solution mixed with FFA(800 micromol/L) the FFA + AM group was incubated in 20 mL KH solution mixed with FFA (800 micromol/L) and AM (4 g/L). The relaxation levels of aorta rings response to acetylcholine and sodium nitroprusside were measured, the expression of NF-kappaB and the level of NOx in the organ bath were analyzed by immunohistochemistry. RESULTS: Severe endothelial dysfunction were induced in FFA group (maximal vasorelaxation in response to Ach: 61.1% +/- l6.9% vs. 93.1% +/- 2.7% in control, P < 0.05), while EDV in FFA+AM group was significantly improved by the incubation with AM (P < 0.05). Compared with the control group (104.1 +/- 14.2) micromol/g, NOx levels of FFA group was (83.1 +/- 8.4) micromol/g (P < 0.05), and the treatment of AM increased the levels of NOx (98.8 +/- 10.7) micromol/g (P < 0.05). The control vascular ring had a little NF-kappaB expression in endothelial nucleus, FFA increased the activation of NF-kappaB, while the treatment of AM lower the elevated NF-kappaB level. CONCLUSION: FFA can directly injure EDV, while AM may ameliorate it, with the possible mechanism related to the signal pathway of NF-kappaB and NO.


Subject(s)
Antioxidants/pharmacology , Astragalus propinquus/chemistry , Endothelial Cells/physiology , Fatty Acids, Nonesterified/antagonists & inhibitors , Oxidative Stress/drug effects , Animals , Aorta/cytology , Cells, Cultured , Drugs, Chinese Herbal/pharmacology , Endothelial Cells/cytology , Male , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley
12.
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
13.
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
14.
Biochem Biophys Res Commun ; 390(3): 557-63, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19818732

ABSTRACT

The free fatty acid receptor, GPR40, is implicated in the pathophysiology of type 2 diabetes, and is a new potential drug target for the treatment of type 2 diabetes. Its antagonist is thought to be not only a useful chemical probe for further exploring the function of GPR40 but also a lead structure for drug development. With virtual screening based on a homology model followed by a cell-based calcium mobilization assay, we found that sulfonamides are a new class of small organic antagonists for GPR40. One of the compounds, DC260126, dose-dependently inhibited GPR40-mediated Ca(2+) elevations stimulated by linoleic acid, oleic acid, palmitoleic acid and lauric acid (IC(50): 6.28+/-1.14, 5.96+/-1.12, 7.07+/-1.42, 4.58+/-1.14 microM, respectively), reduced GTP-loading and ERK1/2 phosphorylation stimulated by linoleic acid in GPR40-CHO cells, suppressed palmitic acid potentiated glucose-stimulated insulin secretion, and negatively regulated GPR40 mRNA expression induced by oleic acid in Min6 cells.


Subject(s)
Fatty Acids, Nonesterified/antagonists & inhibitors , Hypoglycemic Agents/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Sulfonamides/chemistry , Animals , CHO Cells , Cattle , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/pharmacology , Models, Molecular , Protein Conformation , Receptors, G-Protein-Coupled/genetics , Sulfonamides/isolation & purification , Sulfonamides/pharmacology
15.
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
16.
Circulation ; 114(20): 2130-7, 2006 Nov 14.
Article in English | MEDLINE | ID: mdl-17088453

ABSTRACT

BACKGROUND: Metabolic modulators that enhance myocardial glucose metabolism by inhibiting free fatty acid (FFA) metabolism may improve cardiac function in heart failure patients. We studied the effect of acute FFA withdrawal on cardiac function in patients with heart failure caused by idiopathic dilated cardiomyopathy (IDCM). METHODS AND RESULTS: Eighteen fasting nondiabetic patients with IDCM (14 men, 4 women, aged 58.8+/-8.0 years, ejection fraction 33+/-8.8%) and 8 matched healthy controls underwent examination of myocardial perfusion and oxidative and FFA metabolism, before and after acute reduction of serum FFA concentrations by acipimox, an inhibitor of lipolysis. Metabolism was monitored by positron emission tomography and [15O]H2O, [11C]acetate, and [11C]palmitate. Left ventricular function and myocardial work were echocardiographically measured, and efficiency of forward work was calculated. Acipimox decreased myocardial FFA uptake by >80% in both groups. Rate-pressure product and myocardial perfusion remained unchanged, whereas stroke volume decreased similarly in both groups. In the healthy controls, reduced cardiac work was accompanied by decreased oxidative metabolism (from 0.071+/-0.019 to 0.055+/-0.016 min(-1), P<0.01). In IDCM patients, cardiac work fell, whereas oxidative metabolism remained unchanged and efficiency fell (from 35.4+/-12.6 to 31.6+/-13.3 mm Hg x L x g(-1), P<0.05). CONCLUSIONS: Acutely decreased serum FFA depresses cardiac work. In healthy hearts, this is accompanied by parallel decrease in oxidative metabolism, and myocardial efficiency is preserved. In failing hearts, FFA depletion did not downregulate oxidative metabolism, and myocardial efficiency deteriorated. Thus, failing hearts are unexpectedly more dependent than healthy hearts on FFA availability. We propose that both glucose and fatty acid oxidation are required for optimal function of the failing heart.


Subject(s)
Cardiac Output, Low/etiology , Cardiac Output, Low/physiopathology , Cardiomyopathy, Dilated/complications , Fatty Acids, Nonesterified/antagonists & inhibitors , Heart/physiopathology , Hypolipidemic Agents/pharmacology , Myocardium/metabolism , Aged , Cardiac Output, Low/diagnostic imaging , Cardiac Output, Low/metabolism , Coronary Circulation , Echocardiography , Fatty Acids, Nonesterified/metabolism , Female , Heart Rate/drug effects , Humans , Male , Middle Aged , Oxidation-Reduction , Positron-Emission Tomography , Pyrazines/pharmacology , Ventricular Function, Left
17.
Clin Pharmacol Ther ; 81(2): 205-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17259945

ABSTRACT

We explored the mechanisms by which a 4-month, placebo-controlled pioglitazone treatment (45 mg/day) improves glycemic control in type II diabetic patients (T2D, n=27) using physiological testing (6-h mixed meal) and a triple tracer technique ([6,6-(2)H(2)]glucose infusion, (2)H(2)O and [6-(3)H]glucose ingestion) to measure endogenous glucose production (EGP), gluconeogenesis (GNG), insulin-mediated glucose clearance and beta-cell glucose sensitivity (by c-peptide modeling). Compared to sex/age/weight-matched non-diabetic controls, T2D patients showed inappropriately (for prevailing insulinemia) raised glucose production (1.05[0.53] vs 0.71[0.36]mmol min(-1) kg(ffm)(-1) pM, P=0.03) because of enhanced GNG (73.1+/-2.4 vs 59.5+/-3.6%, P<0.01) persisting throughout the meal, reduced insulin-mediated glucose clearance (6[5] vs 12[13]ml min(-1) kg(ffm)(-1) nM(-1), P<0.005), and impaired beta-cell glucose-sensitivity (27[38] vs 71[37]pmol min(-1) m(-2) mM(-1), P=0.002). Compared to placebo, pioglitazone improved glucose overproduction (P=0.0001), GNG and glucose underutilization (P=0.05) despite lower insulinemia. GNG improvement was quantitatively related to raised adiponectin. beta-cell glucose sensitivity was unchanged. In mild-to-moderate T2D, pioglitazone monotherapy decreased fasting and post-prandial glycemia, principally via inhibition of gluconeogenesis, improved hepatic and peripheral insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Thiazolidinediones/therapeutic use , Adiponectin/blood , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Double-Blind Method , Drug Administration Schedule , Fasting/blood , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Female , Glucagon/blood , Gluconeogenesis/drug effects , Glucose Tolerance Test , Glycated Hemoglobin/metabolism , Glycolysis/drug effects , Humans , Hyperglycemia/blood , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/blood , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Lactates/blood , Male , Middle Aged , Pioglitazone , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology
18.
J Mol Endocrinol ; 38(1-2): 147-58, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17242177

ABSTRACT

Type 2 diabetes is characterized by peripheral insulin resistance, pancreatic beta-cells dysfunction, and decreased beta-cell mass with increased rate of apoptosis. Chronic exposure to high levels of free fatty acids (FFAs) has detrimental effects on beta-cell function and survival. FFAs have adverse effects on mitochondrial function, with a consequent increase in the production of reactive oxygen species. Hepatocyte growth factor (HGF) plays a critical role in promoting beta-cell survival. In the present study, we investigated whether HGF was capable of protecting beta-cells from death induced by prolonged exposure to FFAs. RINm5F cell line was cultured in the presence of FFAs (oleate:palmitate 2:1) for 72 h in order to induce apoptosis. Simultaneous administration of HGF and FFAs significantly suppressed the impaired insulin secretion and FFA-induced apoptosis. Specifically, HGF exerted its protective effect by counteracting: (i) the overproduction of either hydrogen peroxide and superoxide anion, (ii) the reduction of intracellular gamma-glutamylcysteinylglycine level, and (iii) the depolarization of mitochondrial membrane, induced by prolonged FFAs exposure. These effects appear to be mediated by bcl-2 and phosphatidylinositol 3 kinase (PI3K)/Akt pathways. Indeed, HGF increased mRNA and protein expression of bcl-2 downregulated by FFAs-treatment; moreover, pre-treatment with the specific PI3-kinase inhibitor LY294002, significantly abolished the protective effect of HGF. In conclusion, in rat insulin-producing RINm5F cells, HGF exerts its prosurvival effect by counteracting the increased intracellular oxidative stress and, consequently, by inhibiting apoptosis induced by chronic exposure to FFAs.


Subject(s)
Apoptosis/physiology , Fatty Acids, Nonesterified/antagonists & inhibitors , Hepatocyte Growth Factor/physiology , Oxidative Stress/physiology , Animals , Cell Line , Fatty Acids, Nonesterified/physiology , Insulin-Secreting Cells/metabolism , Rats
19.
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
20.
J Med Chem ; 49(11): 3251-6, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16722643

ABSTRACT

There is an urgent medical need for orally effective drugs to replace insulin injections for the treatment of diabetes mellitus. Vanadium complexes with insulin-mimetic activities have recently been proposed as candidates as new antidiabetic drugs. Following in vitro and in vivo studies on a group of bis(3-hydroxy-4-pyronato)oxovanadium(IV) (1) complexes with VO(O4) coordination mode, bis(allixinato)oxovanadium(IV) (3) which contains allixin, a garlic component, was found to be the most potent antidiabetic agent among them. Complex 3 with a high in vitro insulin-mimetic activity in terms of both free fatty acid (FFA)-release inhibitory and glucose-uptake enhancing activities in isolated rat adipocytes exhibited a high hypoglycemic effect in type 1 diabetic model mice by both intraperitoneal injections and oral administrations. Complex 3 is thus proposed to be one of the most effective candidates for antidiabetic therapy.


Subject(s)
Hypoglycemic Agents/chemical synthesis , Organometallic Compounds/chemical synthesis , Pyrones/chemistry , Vanadium , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Fatty Acids, Nonesterified/antagonists & inhibitors , Garlic , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Mice , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Rats , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL