Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Eur Rev Med Pharmacol Sci ; 24(24): 12938-12947, 2020 12.
Article in English | MEDLINE | ID: mdl-33378044

ABSTRACT

OBJECTIVE: Abnormal lipid metabolism plays a role that cannot be ignored in articular cartilage bone marrow lesions, synovial inflammation, and the destruction of chondrocytes (CHs). Ceramide is one of the key constructions of membrane lipid bilayers, which is an intracellular lipid mediator regulating varieties of cellular behaviors. The purpose of this study was to explore the role of ceramide and its inhibitor in the development of the CHs degeneration. PATIENTS AND METHODS: CHs were isolated from the cartilage collecting from the osteoarthritis (OA) patients, and oleic acid/palmitic (O/P) acid was used to induce CHs lipid disordered. Then, myriocin was used to inhibit the accumulation of ceramide. After that, the apoptosis, cell viability, glucose uptake, oxidative stress, and the chondrogenic gene expression were tested to evaluate the degenerated degree of CHs. RESULTS: Results revealed that O/P induced CH apoptosis, ceramide accumulation, a higher level of oxidative stress, IL-1ß and MMP-13, but it also decreased the collagen-Ⅱ and SOX-9 expressions and affected the glucose uptake of CHs. After the stimulation of myriocin, the side effects induced by O/P was partly reversed. CONCLUSIONS: O/P induces the accumulation of ceramide and the degeneration of CHs, and myriocin can reject the harmful effect caused by O/P via the suppression of ceramide.


Subject(s)
Ceramides/antagonists & inhibitors , Chondrocytes/drug effects , Fatty Acids, Monounsaturated/pharmacology , Oleic Acid/antagonists & inhibitors , Palmitates/antagonists & inhibitors , Adult , Aged , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Ceramides/metabolism , Chondrocytes/metabolism , Female , Humans , Male , Middle Aged , Oleic Acid/pharmacology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Palmitates/pharmacology
2.
Alcohol ; 83: 57-65, 2020 03.
Article in English | MEDLINE | ID: mdl-31499142

ABSTRACT

Several conditions are marked by increased susceptibility to, and enhanced severity of, bacterial infections. Alcohol use disorder, one of these conditions, is known to predispose to bacterial pneumonia by suppressing the lung's innate immune system, and more specifically by disrupting critical alveolar macrophage (AM) functions. Recently, we established that chronic ethanol consumption also perturbs surfactant lipid homeostasis in the lung and that elevated concentrations of free fatty acids contribute to blocking essential AM functions, such as agonist-induced cytokine expression. In this study, we extend these observations by showing that elevated free fatty acid levels impair metabolic responses to lipopolysaccharide (LPS) in AMs. In particular, we show that the glycolytic reprogramming characteristic of LPS-stimulated AMs is blunted by the saturated fatty acid palmitate, whereas oleate, an unsaturated fatty acid, or ethanol alone, had no effect on this adaptive metabolic response. Additionally, we found that elevated concentrations of palmitate induced mitochondrial oxidative stress and that glycolytic reprogramming and cytokine production to LPS could be partially restored in AMs by either pharmacologically blocking palmitate entry into mitochondria or administering a mitochondrial-specific antioxidant. Taken together, these findings suggest that alcohol and elevated levels of saturated fatty acids conspire to impair pulmonary innate immunity by altering metabolic responses in AMs. Additionally, our findings suggest that targeting the mechanisms involved in fatty acid metabolism can restore pulmonary immunity and possibly limit bacterial pneumonia in individuals with alcohol use disorder.


Subject(s)
Ethanol/toxicity , Glycolysis/drug effects , Lipid Metabolism/drug effects , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/physiology , Animals , Cell Line , Cytokines/metabolism , Fatty Acids/metabolism , Immunity/drug effects , Immunity/physiology , Macrophages, Alveolar/ultrastructure , Mitochondria/metabolism , Oxidative Stress/drug effects , Palmitates/antagonists & inhibitors , Palmitates/metabolism , Palmitates/pharmacology , Rats
3.
Life Sci ; 206: 84-92, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-29802940

ABSTRACT

AIMS: High level of saturated fatty acids leads to mitochondrial dysfunction and inflammation in the development of insulin resistance in skeletal muscle. We recently found that puerarin improved impaired insulin signaling in skeletal muscle in diabetic animals and in myotubes in vitro. However, whether puerarin can act directly on muscle cells to alleviate lipid-induced mitochondrial dysfunction and inflammation remains obscure. This study was conducted to analyze the attributive properties of puerarin against mitochondrial dysfunction and inflammation in skeletal muscle cells with insulin resistance. MAIN METHODS: The effects of puerarin on mitochondrial biogenesis, oxidative phosphorylation, dynamics of fusion, fission and mitophagy, oxidative stress, as well as inflammatory response and insulin sensitivity in L6 myotubes treated with palmitate were examined. KEY FINDINGS: Puerarin pretreatment improve insulin sensitivity and prevented muscle cells from palmitate-induced mitochondrial dysfunction manifested by the increases of complex I activity, mitochondrial membrane potential and ATP generation, and the decrease of reactive oxygen species (ROS) production. Augmented expression of genes involved in mitochondrial biogenesis, oxidative phosphorylation, and the detoxification of ROS were also observed upon puerarin supplementation. Moreover, puerarin modulated mitochondrial fusion and fission, and rescued palmitate-impaired mitophagy via phosphatase and tensin homolog-induced putative kinase 1(PINK1)/Parkin pathway. In addition, puerarin attenuated palmitate-induced inflammation through the inhibition of toll-like receptor 4/nuclear factor-κB signaling pathway. SIGNIFICANCE: Our findings indicated that puerarin could act directly on muscle cells to attenuate palmitate-induced mitochondrial dysfunction, impaired mitophagy and inflammatory response, thereby contributing to the improvement of insulin sensitivity.


Subject(s)
Inflammation/prevention & control , Isoflavones/pharmacology , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/prevention & control , Mitophagy/drug effects , Muscle Fibers, Skeletal/drug effects , Palmitates/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Cell Fusion , Cell Line , Insulin Resistance , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Muscle/drug effects , Muscle Fibers, Skeletal/pathology , Oxidative Phosphorylation/drug effects , Palmitates/toxicity , Rats
4.
Mar Drugs ; 15(6)2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28632169

ABSTRACT

Astaxanthin, an antioxidant agent, can protect pancreatic ß-cells of db/db mice from glucotoxicity and resolve chronic inflammation in adipose tissue. Nonetheless, the effects of astaxanthin on free-fatty-acid-induced inflammation and cellular stress in ß-cells remain to be demonstrated. Meanwhile, palmitate enhances the secretion of pro-inflammatory adipokines monocyte chemoattractant protein-1 (MCP-1) and VEGF120 (vascular endothelial growth factor). We therefore investigated the influence of astaxanthin on palmitate-stimulated MCP-1 and VEGF120 secretion in mouse insulinoma (MIN6) pancreatic ß-cells. Furthermore, whether astaxanthin prevents cellular stress in MIN6 cells was also assessed. Pre-treatment with astaxanthin or with N-acetyl-cysteine (NAC) which is an antioxidant drug, significantly attenuated the palmitate-induced MCP-1 release through downregulation of phosphorylated c-Jun NH2-terminal protein kinase (JNK) pathways, and suppressed VEGF120 through the PI3K/Akt pathways relative to the cells stimulated with palmitate alone. In addition, palmitate significantly upregulated homologous protein (CHOP) and anti-glucose-regulated protein (GRP78), which are endoplasmic reticulum (ER) stress markers, in MIN6 cells. On the other hand, astaxanthin attenuated the increased CHOP content, but further up-regulated palmitate-stimulated GRP78 protein expression. By contrast, NAC had no effects on either CHOP or GRP78 enhancement induced by palmitate in MIN6 cells. In conclusion, astaxanthin diminishes the palmitate-stimulated increase in MCP-1 secretion via the downregulation of JNK pathways in MIN6 cells, and affects VEGF120 secretion through PI3K/Akt pathways. Moreover, astaxanthin can prevent not only oxidative stress caused endogenously by palmitate but also ER stress, which NAC fails to attenuate, via upregulation of GRP78, an ER chaperon.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects , Palmitates/pharmacology , Acetylcysteine/pharmacology , Animals , Cell Line, Tumor , Chemokine CCL2/metabolism , Endoplasmic Reticulum Chaperone BiP , MAP Kinase Signaling System/drug effects , Mice , Palmitates/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xanthophylls/pharmacology
5.
Apoptosis ; 21(5): 546-57, 2016 May.
Article in English | MEDLINE | ID: mdl-26921179

ABSTRACT

Elevated circulatory free fatty acids (FFAs) especially saturated FFAs, such as palmitate (PA), are detrimental to the heart. However, mechanisms responsible for this phenomenon remain unknown. Here, the role of JAK2/STAT3 in PA-induced cytotoxicity was investigated in cardiomyocytes. We demonstrate that PA suppressed the JAK2/STAT3 pathway by dephosphorylation of JAK2 (Y1007/1008) and STAT3 (Y705), and thus blocked the translocation of STAT3 into the nucleus. Conversely, phosphorylation of S727, another phosphorylated site of STAT3, was increased in response to PA treatment. Pretreatment of JNK inhibitor, but not p38 MAPK inhibitor, inhibited STAT3 (S727) activation induced by PA and rescued the phosphorylation of STAT3 (Y705). The data suggested that JNK may be another upstream factor regulating STAT3, and verified the important function of P-STAT3 (Y705) in PA-induced cardiomyocyte apoptosis. Sodium orthovanadate (SOV), a protein tyrosine phosphatase inhibitor, obviously inhibited PA-induced apoptosis by restoring JAK2/STAT3 pathways. This effect was diminished by STAT3 inhibitor Stattic. Collectively, our data suggested a novel mechanism that the inhibition of JAK2/STAT3 activation was responsible for palmitic lipotoxicity and SOV may act as a potential therapeutic agent by targeting JAK2/STAT3 in lipotoxic cardiomyopathy treatment.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Janus Kinase 2/metabolism , Myocytes, Cardiac/drug effects , Palmitates/toxicity , STAT3 Transcription Factor/metabolism , Vanadates/pharmacology , Animals , Cell Line , Cells, Cultured , Janus Kinase 2/antagonists & inhibitors , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Palmitates/antagonists & inhibitors , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Rats, Sprague-Dawley , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects
6.
Pharmazie ; 71(12): 733-737, 2016 12 01.
Article in English | MEDLINE | ID: mdl-29442004

ABSTRACT

Rapamycin (Rap), a specific inhibitor of the mTOR signaling, has been shown to affect lipid metabolism in vitro and in vivo. Sirt-1, an NAD+ dependent deacetylase, regulates a variety of cellular processes, including aging, lifespan extension and glucose and lipid metabolism. Herein, we applied a cellular steatosis model to investigate whether rapamycin's role in lipid metabolism is sirt 1-associated. Cells were exposed to palmitate stimulation for 48 h with or without rapamycin treatment. Lipid droplets in AML12 cells were observed by oil red O staining, and the intracellular lipid content was measured. We found that rapamycin treatment, at a relatively low concentration, significantly attenuated lipid aggregation, whereas knockdown of sirt-1 by siRNA abrogated rapamycin's effect on ameliorating lipid accumulation. Moreover, rapamycin exposure increased the expression levels of sirt-1 and AMPK, and enhanced sirt-1 deacetylase activity in steatotic AML12 hepatocytes. This is the first report demonstrating that rapamycin ameliorates lipid accumulation through upregulating sirt-1 signaling supporting the hypothesis that rapamycin may positively influence sirt-1 signaling in maintaining metabolic homeostasis.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Palmitates/antagonists & inhibitors , Sirolimus/pharmacology , Sirtuin 1/drug effects , Animals , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Mice , Mitogen-Activated Protein Kinases/biosynthesis , Palmitates/pharmacology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects
7.
Biochem Biophys Res Commun ; 441(4): 770-6, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24189472

ABSTRACT

BACKGROUND: Saturated free fatty acids (FFAs), such as palmitate, cause ß-cell apoptosis whereas unsaturated FFAs, e.g. oleate, are not harmful. The toxicity of palmitate could be mediated through endoplasmic reticulum (ER) stress which triggers the activation of a signal responding cascade also called unfolded protein response (UPR). We investigated whether or not palmitate induced ß-cell apoptosis through UPR activation and whether or not oleate as a monounsaturated fatty acid could counteract these effects. METHODS: INS-1E ß-cells were incubated with palmitate [0.5mM], oleate [1mM] or the combination [0.5/1mM] for 1, 6 and 24h. Viability and induction of apoptosis were measured by WST-1 assay and FITC-Annexin/PI-staining, respectively. Western blot analyses were performed for UPR specific proteins and mRNA expression of target molecules was determined by qPCR. RESULTS: Palmitate significantly decreased viability (29±8.8%) of INS-1E ß-cells compared to controls after 24h. Stimulation with oleate showed no effect on viability but the combination of oleate and palmitate improved viability compared to palmitate treated cells (55±9.3%) or controls (26±5.3%). The number of apoptotic cells was increased 2-fold after 24h incubation with palmitate compared to controls. Again, oleate showed no effect but in combination ameliorated the effect of palmitate to control level. Phosphorylation of eIF2α was increased after 6 and 24h incubation with palmitate. In contrast, oleate had no effect and in combination prevented phosphorylation of eIF2α. Increased Xbp1 splicing was visible already 6h after palmitate treatment and remained elevated at 24h. The combination with oleate abolished Xbp1 splicing. Interestingly, mRNA expression of the chaperones Bip, Pdi, Calnexin and Grp94 was not altered by FFA treatment. Only the proapoptotic transcription factor Chop was significantly enhanced by palmitate incubation. In accordance with sustained cell survival the combination as well as oleate alone, did not result in increased Chop levels compared to controls. In summary, we showed that oleate protects INS-1E ß-cells from palmitate-induced apoptosis by the suppression of ER stress which was independent of chaperone activation.


Subject(s)
Apoptosis/drug effects , Cytoprotection , Insulin-Secreting Cells/drug effects , Oleic Acid/pharmacology , Palmitates/antagonists & inhibitors , Unfolded Protein Response/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Insulin-Secreting Cells/pathology , Molecular Chaperones/metabolism , Palmitates/pharmacology , Rats , Transcription Factor CHOP/antagonists & inhibitors , Up-Regulation/drug effects
8.
Pharmazie ; 68(4): 270-3, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23700793

ABSTRACT

This study aimed to investigate the effect of pioglitazone (PIO) on insulin secretion and mitochondrial ultrastructure and function in 3 cells. HIT-T15 cells were treated with control or palmitate (free fat acids, FFA) or/and PIO and divided into 7 groups: Control group; 0.5 mmol/l FFA (LF); 0.5 mmol/l FFA plus 10-7 mol/I PIO (LFLP); 0.5 mmol/l FFA plus 10-5mol/I PIO (LFHP); 1.0 mmol/l FFA (HF); 1.0 mmol/l FFA plus 10-7mol/I PIO (HFLP); 1.0 mmol/l FFA plus 10-5 mol/I PIO (HFHP). Apoptotic peaks, mitochondrial ultrastructure, ATP/ADP, mRNA levels of peroxisome proliferater activated receptor gamma coactivator-1 (PGC-1) and nucleus respiratory factor-1 (NRF-1) as well as insulin secretion were measured. The results showed that palmitate impaired mitochondrion structure, which could be alleviated by PIO. Palmitate could increase apoptotic peaks, decrease ATP/ADP ratio, enhance the expression of PGC-1 mRNA and NRF-1 mRNA, and decrease glucose stimulated insulin secretion (GSIS). In contrast, PIO could decrease apoptotic peaks, restore partly ATP/ADP ratio, decrease the expression of PGC-1 mRNA and NRF-1 mRNA, and increase GSIS level. These results demonstrate that PIO could ameliorate palmitate induced damage to mitochondrion ultrastructure and function and restore GSIS, accompanied by the modulation of PGC-1 and NRF-1 expression. These findings provide new insight into the hypoglycemic effects of PIO and help develop new agents for diabetes therapy.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Mitochondria/ultrastructure , Palmitates/antagonists & inhibitors , Palmitates/toxicity , Thiazolidinediones/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Cell Line , Chromatography, High Pressure Liquid , Flow Cytometry , Heat-Shock Proteins/biosynthesis , Humans , In Situ Nick-End Labeling , Indicators and Reagents , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/ultrastructure , Microscopy, Electron , Mitochondria/drug effects , Nuclear Respiratory Factor 1/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pioglitazone , Real-Time Polymerase Chain Reaction , Transcription Factors/biosynthesis
9.
Life Sci ; 88(17-18): 803-9, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21396380

ABSTRACT

AIMS: Tetramethylpyrazine (TMP), one of the active ingredients isolated from a Chinese herbal prescription, possesses protective effects against oxidative stress caused by high glucose in endothelial cells. In this study, the role of TMP in preventing muscle cells from palmitate-induced oxidative damage was investigated and the possible mechanisms of action elucidated. MAIN METHODS: Mitochondrial reactive oxygen species (ROS) were measured in C2C12 myotubes, a palmitate-induced oxidative stress cell model, with or without TMP. Both mitochondrial membrane potential (MMP) and oxygen consumption were assessed in conjunction with quantification of mitochondrial DNA and mitochondrial biogenesis-related factors, such as peroxisome proliferator-activated receptor-γ coactivator 1 α (PGC1α), nuclear respiratory factor 1 (NRF1) and mitochondrial transcription factor A (Tfam), by real-time polymerase chain reaction. Expression of mitochondrial respiratory chain complex III as an index of mitochondrial function was evaluated by immunoblotting, and glucose transport into the C2C12 myotube examined by analyzing 2-deoxy-[(3)H]glucose uptake. KEY FINDINGS: TMP significantly alleviated palmitate-induced mitochondrial ROS production, mitigated mitochondrial dysfunction and increased D-loop mRNA expression as compared with the control. This was accompanied by a marked reversal of palmitate-induced down-regulation in the expression of mitochondrial biogenesis-related factors (PGC1α, NRF1 and Tfam) and decreased glucose uptake in C2C12 myotubes. As a result, cell respiration, as reflected by the elevated expression of mitochondrial respiratory chain complex III and oxygen consumption, was enhanced. SIGNIFICANCE: TMP is capable of protecting C2C12 myotubes against palmitate-induced oxidative damage and mitochondrial dysfunction, and improving glucose uptake in muscle cells partially through the up-regulation of mitochondrial biogenesis.


Subject(s)
Antioxidants/pharmacology , Mitochondria, Muscle/drug effects , Muscle Fibers, Skeletal/drug effects , Oxidative Stress/drug effects , Palmitates/pharmacology , Pyrazines/pharmacology , Animals , Blotting, Western , Cell Line , Glucose/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Oxygen Consumption/drug effects , Palmitates/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Clin Sci (Lond) ; 120(5): 195-206, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20840078

ABSTRACT

Chronic exposure of pancreatic ß-cells to saturated non-esterified fatty acids can lead to inhibition of insulin secretion and apoptosis. Several previous studies have demonstrated that saturated fatty acids such as PA (palmitic acid) are detrimental to ß-cell function compared with unsaturated fatty acids. In the present study, we describe the effect of the polyunsaturated AA (arachidonic acid) on the function of the clonal pancreatic ß-cell line BRIN-BD11 and demonstrate AA-dependent attenuation of PA effects. When added to ß-cell incubations at 100 µM, AA can stimulate cell proliferation and chronic (24 h) basal insulin secretion. Microarray analysis and/or real-time PCR indicated significant AA-dependent up-regulation of genes involved in proliferation and fatty acid metabolism [e.g. Angptl (angiopoietin-like protein 4), Ech1 (peroxisomal Δ3,5,Δ2,4-dienoyl-CoA isomerase), Cox-1 (cyclo-oxygenase-1) and Cox-2, P<0.05]. Experiments using specific COX and LOX (lipoxygenase) inhibitors demonstrated the importance of COX-1 activity for acute (20 min) stimulation of insulin secretion, suggesting that AA metabolites may be responsible for the insulinotropic effects. Moreover, concomitant incubation of AA with PA dose-dependently attenuated the detrimental effects of the saturated fatty acid, so reducing apoptosis and decreasing parameters of oxidative stress [ROS (reactive oxygen species) and NO levels] while improving the GSH/GSSG ratio. AA decreased the protein expression of iNOS (inducible NO synthase), the p65 subunit of NF-κB (nuclear factor κB) and the p47 subunit of NADPH oxidase in PA-treated cells. These findings indicate that AA has an important regulatory and protective ß-cell action, which may be beneficial to function and survival in the 'lipotoxic' environment commonly associated with Type 2 diabetes mellitus.


Subject(s)
Arachidonic Acid/pharmacology , Insulin-Secreting Cells/drug effects , Palmitates/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cyclooxygenase Inhibitors/pharmacology , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Gene Expression Regulation/drug effects , Glutathione/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Lipoxygenase Inhibitors/pharmacology , Nitrites/metabolism , Oxidative Stress/drug effects , Palmitates/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
11.
Clin Sci (Lond) ; 120(5): 179-81, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21044045

ABSTRACT

Saturated fatty acids are toxic to pancreatic ß-cells. By inducing apoptosis, they contribute to a decrease in ß-cell mass, a hallmark of Type 2 diabetes. In the present issue of Clinical Science, Keane and co-workers show that the polyunsaturated fatty acid arachidonic acid protects the ß-cell against the toxic effects of palmitate. As Type 2 diabetes is characterized by subclinical inflammation, and arachidonic acid and metabolites thereof are produced during states of inflammation, it is possible that pancreatic ß-cells use arachidonic acid as a compound for self-protection.


Subject(s)
Arachidonic Acid/pharmacology , Insulin-Secreting Cells/drug effects , Palmitates/antagonists & inhibitors , Animals , Arachidonic Acid/metabolism , Arachidonic Acid/physiology , Diabetes Mellitus, Type 2/physiopathology , Humans , Insulin-Secreting Cells/metabolism , Palmitates/pharmacology
12.
Biochem Biophys Res Commun ; 402(2): 265-71, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20937260

ABSTRACT

Free fatty acid (FFA)-bound albumin, which is filtrated through the glomeruli and reabsorbed into proximal tubular cells, is one of the crucial mediators of tubular damage in proteinuric kidney disease. In this study, we examined the role of each kind of FFA on renal tubular damage in vitro and tried to identify its molecular mechanism. In cultured proximal tubular cells, a saturated fatty acid, palmiate, increased the expression of monocyte chemoattractant protein-1 (MCP-1), but this effect was abrogated by co-incubation of monounsaturated fatty acid, oleate, or ω-3 polyunsaturated fatty acid, eicosapentaenoic acid (EPA). Palmitate led to intracellular accumulation of diacylglycerol (DAG) and subsequent activation of protein kinase C protein family. Among the several PKC inhibitors, rottlerin, a PKCθ inhibitor, prevented palmitate-induced MCP-1 expression via inactivation of NFB pathway. Overexpression of dominant-negative PKCθ also inhibited palmitate-induced activation of MCP-1 promoter. Furthermore, palmitate enhanced PKCθ-dependent mitochondrial apoptosis, which was also prevented by co-incubation with oleate or EPA through restoration of pro-survival Akt pathway. Moreover, oleate and EPA inhibited palmitate-induced PKCθ activation through the conversion of intracellular DAG to triglyceride with the restoration of diacylglycerol acyltransferase 2 expression. These results suggest that oleate and EPA have protective effects against the palmitate-induced renal tubular cell damage by inhibiting PKCθ activation.


Subject(s)
Apoptosis/drug effects , Cytoprotection , Eicosapentaenoic Acid/pharmacology , Inflammation/chemically induced , Kidney Tubules, Proximal/drug effects , Oleic Acid/pharmacology , Palmitates/antagonists & inhibitors , Animals , Cells, Cultured , Chemokine CCL2/genetics , Diacylglycerol O-Acyltransferase/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice , NF-kappa B/metabolism , Palmitates/toxicity , Promoter Regions, Genetic/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C-theta , Proto-Oncogene Proteins c-akt/metabolism , Transcriptional Activation , Triglycerides/metabolism
13.
Br J Nutr ; 102(11): 1557-63, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19622194

ABSTRACT

Oleate has been shown to protect against palmitate-induced insulin resistance. The present study investigates mechanisms involved in the interaction between oleate and palmitate on insulin-stimulated glucose uptake by L6 skeletal muscle cells. L6 myotubes were cultured for 6 h with palmitate or oleate alone, and combinations of palmitate with oleate, with and without phosphatidylinositol 3-kinase (PI3-kinase) inhibition. Insulin-stimulated glucose uptake, measured by uptake of 2-deoxy-d-[3H]glucose, was almost completely prevented by 300 microm-palmitate. Cells incubated with oleate up to 750 micromol/l maintained a significant increase in insulin-stimulated glucose uptake. Co-incubation of 50-300 microm-oleate with 300 microm-palmitate partially prevented the decrease in insulin-stimulated glucose uptake associated with palmitate. Adding the PI3-kinase inhibitors wortmannin (10- 7 mol/l) or LY294002 (25 micromol/l) to 50 microm-oleate plus 300 microm-palmitate significantly reduced the beneficial effect of oleate against palmitate-induced insulin resistance, indicating that activation of PI3-kinase is involved in the protective effect of oleate. Thus, the prevention of palmitate-induced insulin resistance by oleate in L6 muscle cells is associated with the ability of oleate to maintain insulin signalling through PI3-kinase.


Subject(s)
Insulin Resistance/physiology , Muscle Fibers, Skeletal/drug effects , Oleic Acid/pharmacology , Palmitates/antagonists & inhibitors , Androstadienes/pharmacology , Animals , Cell Membrane/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chromones/pharmacology , Deoxyglucose/pharmacokinetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/physiology , Palmitates/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Rats , Wortmannin
14.
Biochemistry (Mosc) ; 73(7): 783-90, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18707586

ABSTRACT

Palmitate-induced uncoupling, which involves ADP/ATP and aspartate/glutamate antiporters, has been studied in liver mitochondria of old rats (22-26 months) under conditions of lipid peroxidation and inhibition of oxidative stress by antioxidants--thiourea, Trolox, and ionol. It has been shown that in liver mitochondria of old rats in the absence of antioxidants and under conditions of overproduction of conjugated dienes, the protonophoric uncoupling activity of palmitate is not suppressed by either carboxyatractylate or aspartate used separately. However, the combination of carboxyatractylate and aspartate decreased uncoupling activity of palmitate by 81%. In this case, palmitate-induced uncoupling is limited by a stage insensitive to both carboxyatractylate and aspartate. In the presence of antioxidants, the palmitate-induced protonophoric uncoupling activity is suppressed by either carboxyatractylate or aspartate used separately. Under these conditions, palmitate-induced uncoupling is limited by a stage sensitive to carboxyatractylate (ADP/ATP antiporter) or aspartate (aspartate/glutamate antiporter). In the absence of antioxidants, the uncoupling activity of palmitate is not suppressed by ADP either in the absence or in the presence of aspartate. However, in the presence of thiourea, Trolox, or ionol ADP decreased the uncoupling activity of palmitate by 38%. It is concluded that in liver mitochondria of old rats the development of oxidative stress in the presence of physiological substrates of ADP/ATP and aspartate/glutamate antiporters (ADP and aspartate) results in an increase of the protonophoric uncoupling activity of palmitate.


Subject(s)
Aging/physiology , Amino Acid Transport System X-AG/metabolism , Mitochondria, Liver/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Oxidative Stress , Palmitates/pharmacology , Uncoupling Agents/pharmacology , Animals , Antioxidants/pharmacology , Antiporters/metabolism , Aspartic Acid/pharmacology , Atractyloside/analogs & derivatives , Atractyloside/pharmacology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Respiration/drug effects , Lipid Peroxidation , Male , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Palmitates/antagonists & inhibitors , Rats , Uncoupling Agents/antagonists & inhibitors
15.
FEBS J ; 274(23): 6094-105, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17970749

ABSTRACT

Fatty acids can stimulate the secretory activity of insulin-producing beta-cells. At elevated concentrations, they can also be toxic to isolated beta-cells. This toxicity varies inversely with the cellular ability to accumulate neutral lipids in the cytoplasm. To further examine whether cytoprotection can be achieved by decreasing cytoplasmic levels of free acyl moieties, we investigated whether palmitate toxicity is also lowered by stimulating its beta-oxidation. Lower rates of palmitate-induced beta-cell death were measured in the presence of L-carnitine as well as after addition of peroxisome proliferator-activated receptor alpha (PPARalpha) agonists, conditions leading to increased palmitate oxidation. In contrast, inhibition of mitochondrial beta-oxidation by etomoxir increased palmitate toxicity. A combination of PPARalpha and retinoid X receptor (RXR) agonists acted synergistically and led to complete protection; this was associated with enhanced expression levels of genes involved in mitochondrial and peroxisomal beta-oxidation, lipid metabolism, and peroxisome proliferation. PPARalpha-RXR protection was abolished by the carnitine palmitoyl transferase 1 inhibitor etomoxir. These observations indicate that PPARalpha and RXR regulate beta-cell susceptibility to long-chain fatty acid toxicity by increasing the rates of beta-oxidation and by involving peroxisomes in fatty acid metabolism.


Subject(s)
Insulin-Secreting Cells/metabolism , PPAR alpha/agonists , Palmitates/toxicity , Protective Agents/metabolism , Retinoid X Receptors/agonists , Alitretinoin , Animals , Carbon Radioisotopes/metabolism , Carnitine/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Clofibrate/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Gene Expression/drug effects , Glucose/metabolism , Kinetics , Male , Mitochondria/genetics , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Palmitates/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tretinoin/metabolism
16.
Br J Pharmacol ; 151(5): 610-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17471172

ABSTRACT

BACKGROUND AND PURPOSE: A number of studies have implicated adipocyte-derived factors in the development of insulin resistance. Intracellular redox status has been reported to play a significant role in the modulation of insulin action. This study was designed to investigate the potential of crocetin, a potent antioxidant, to protect adipocytes against the induction of insulin insensitivity and disordered expression of tumor necrosis factor (TNF)-alpha and adiponectin in vitro. EXPERIMENTAL APPROACH: We used palmitate to induce insulin resistance in freshly isolated rat adipocytes, and observed the effect of crocetin, N-acetylcysteine, diphenyleneiodonium, rotenone and oxypurinol. Insulin sensitivity was measured using 2-deoxy-D-[1-(3)H]-glucose uptake assay. Levels of glucose transporter 4, TNF-alpha and adiponectin were evaluated by immunoblot analysis, and levels of mRNA for TNF-alpha and adiponectin by reverse transcription-polymerase chain reaction analysis. Intracellular production of reactive oxygen species (ROS) was determined spectrofluorometrically using 2',7'-dichlorofluorescin diacetate. KEY RESULTS: Palmitate induced a 45% decrease in insulin-stimulated glucose uptake in adipocytes. The mRNA and protein expression of TNF-alpha were enhanced by 64% and 59% respectively whereas the mRNA and protein expression of adiponectin were reduced by 43% and 36% respectively by palmitate treatment. These changes were accompanied by a 54% increase in intracellular ROS levels. Crocetin, N-acetylcysteine and diphenyleneiodonium were found to attenuate these abnormalities. CONCLUSIONS AND IMPLICATIONS: Crocetin blocked the impaired insulin-stimulated glucose uptake and disordered TNF-alpha and adiponectin expression induced by palmitate in rat adipocytes. Inactivation of NADPH oxidase may account for these observations.


Subject(s)
Adipocytes/metabolism , Adiponectin/biosynthesis , Antioxidants/pharmacology , Carotenoids/pharmacology , Insulin Resistance , Palmitates/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Acetylcysteine/pharmacology , Adipocytes/drug effects , Animals , Antimetabolites , Blotting, Western , Deoxyglucose , Glucose/metabolism , Glucose Transporter Type 4/biosynthesis , Male , Onium Compounds/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction , Vitamin A/analogs & derivatives
17.
Biochem Biophys Res Commun ; 324(4): 1204-9, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15504342

ABSTRACT

The fuel sensing enzyme AMP-activated protein kinase (AMPK) enhances processes that generate ATP when stresses such as exercise or glucose deprivation make cells energy deficient. We report here a novel role of AMPK, to prevent the activation of NF-kappaB in endothelial cells exposed to the fatty acid palmitate or the cytokine TNF-alpha. Incubation of cultured human umbilical vein endothelial cells (HUVEC) with elevated levels of palmitate (0.4mM) increased NF-kappaB reporter gene expression by 2- to 4-fold within 8h and caused a 7-fold increase in VCAM-1 mRNA expression at 24h. In contrast, no increase in reporter gene expression was detected for AP-1, glucocorticoid-, cyclic AMP-, or serum response elements. Similar increases in NF-kappaB activation and VCAM-1 expression were not observed in cells incubated with an elevated concentration of glucose (25mM). The increases in NF-kappaB activation and VCAM-1 expression caused by palmitate were markedly inhibited by co-incubation with the AMPK activator AICAR and, where studied, by expression of a constitutively active AMPK. Likewise, AMPK activation inhibited the increase in NF-kappaB reporter gene expression observed in HUVEC incubated with TNF-alpha. The results suggest that AMPK inhibits the activation of NF-kappaB caused by both palmitate and TNF-alpha. The mechanism responsible for this action, as well as its relevance to the reported anti-atherogenic actions of exercise, metformin, thiazolidinediones, and adiponectin, all of which have been shown to activate AMPK, remains to be determined.


Subject(s)
Endothelium, Vascular/metabolism , Multienzyme Complexes/physiology , NF-kappa B/antagonists & inhibitors , Palmitates/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Transcriptional Activation , AMP-Activated Protein Kinases , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium, Vascular/enzymology , Humans , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Umbilical Veins/cytology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
18.
Insect Biochem Mol Biol ; 33(12): 1299-306, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14599501

ABSTRACT

The corpus allatum (CA) of adult female Ceratitis capitata produces methyl palmitate (MP) in vitro, in addition to JHB(3) and JH III. Biosynthesized MP migrates on TLC and co-elutes from RP-18 HPLC with synthetic MP. Its identity is verified herein by GCMS. MP production is up-regulated twofold by mevastatin, an inhibitor of mevalonic acid-dependent isoprene biosynthesis. Fosmidomycin, an inhibitor of mevalonic acid-independent isoprene synthesis in graminaceous plants, up-regulates MP synthesis by about fourfold. However, it does not depress JHB(3) biosynthesis concurrently. This suggests that the initial enzyme(s) in the conversion of 1-deoxy-xylulose 5-phosphate to isoprene is presumably present in C. capitata, but is inhibited by fosmidomycin, and this inhibition diverts precursors to MP synthesis. Phytol, an acyclic diterpene, might be suppressing isoprene biosynthesis by CA, thereby resulting in a fourfold increase in the MP biosynthesis. Linolenic acid is an end-product and its presence in incubation media up-regulates MP biosynthesis by twofold, presumably due to the feedback diversion to biosynthesis of C(16:0) and its methyl ester. Biosynthesis of MP is markedly depressed after mating, while otherwise maintained at significantly higher levels in virgin females. MP biosynthesis is significantly reduced in virgin females by direct axonal control but is less consistent after mating.


Subject(s)
Ceratitis capitata/metabolism , Corpora Allata/metabolism , Fosfomycin/analogs & derivatives , Lovastatin/analogs & derivatives , Palmitates/metabolism , Animals , Chromatography, Thin Layer , Fatty Acids, Monounsaturated/antagonists & inhibitors , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Female , Fosfomycin/pharmacology , Gas Chromatography-Mass Spectrometry , Gene Expression Regulation/drug effects , Linoleic Acid/pharmacology , Lovastatin/pharmacology , Palmitates/antagonists & inhibitors , Palmitates/chemistry , Palmitates/isolation & purification , Phytol/pharmacology , Sesquiterpenes/antagonists & inhibitors , Sesquiterpenes/metabolism , Sexual Behavior, Animal/physiology , Tritium
19.
Am J Physiol Endocrinol Metab ; 284(3): E611-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12453824

ABSTRACT

Triglyceride hydrolysis by the perfused rat hindlimb is enhanced with serotonin-induced nonnutritive flow (NNF) and may be due to the presence of nonnutritive route-associated connective tissue fat cells. Here, we assess whether NNF influences muscle uptake of 0.55 mM palmitate in the perfused hindlimb. Comparisons were made with insulin-mediated glucose uptake. NNF induced during 60 nM insulin infusion inhibited hindlimb oxygen uptake from 22.0 +/- 0.5 to 9.7 +/- 0.8 micromol x g(-1) x h(-1) (P < 0.001), 1-methylxanthine metabolism (indicator of nutritive flow) from 5.8 +/- 0.4 to 3.8 +/- 0.4 nmol x min(-1) x g(-1) (P = 0.004), glucose uptake from 29.2 +/- 1.7 to 23.1 +/- 1.8 micromol x g(-1) x h(-1) (P = 0.005) and muscle 2-deoxyglucose uptake from 82.1 +/- 4.6 to 41.6 +/- 6.7 micromol x g(-1) x h(-1) (P < 0.001). Palmitate uptake, unaffected by insulin alone, was inhibited by NNF in extensor digitorum longus, white gastrocnemius, and tibialis anterior muscles; average inhibition was from 13.9 +/- 1.2 to 6.9 +/- 1.4 micromol x g(-1) x h(-1) (P = 0.02). Thus NNF impairs both fatty acid and glucose uptake by muscle by restricting flow to myocytes but, as shown previously, favors triglyceride hydrolysis and uptake into nearby connective tissue fat cells. The findings have implications for lipid partitioning in limb muscles between myocytes and attendant adipocytes.


Subject(s)
Animal Nutritional Physiological Phenomena , Hindlimb , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Palmitates/antagonists & inhibitors , Animals , Glucose/pharmacokinetics , Insulin/pharmacology , Male , Perfusion , Rats , Rats, Wistar , Regional Blood Flow/physiology
20.
Biochem Biophys Res Commun ; 244(1): 11-4, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9514876

ABSTRACT

Inhibitors of fatty acid oxidation, 2-bromopalmitic acid (Br-C16) and 4-bromocrotonic acid (Br-C4) were examined for their effect on glucose transport in 3T3-L1 adipocytes. Whereas Br-C16 was without effect, Br-C4 augmented basal but inhibited insulin-stimulated 2-deoxyglucose uptake in a dose- and time-dependent manner. Immunoblot analysis indicated that following Br-C4 pretreatment, the content of GLUT1 in plasma membranes was increased whereas insulin-induced translocation of GLUT4 was greatly eliminated. The total cellular amount of GLUT1 or GLUT4, on the other hand, was not altered. Thus these results seem to suggest that Br-C4 has opposite effect on basal and insulin-stimulated glucose transport by a mechanism other than its inhibition of fatty acid oxidation. The translocation processes for both GLUT1 and GLUT4 transporters appears to be altered.


Subject(s)
Adipocytes/metabolism , Crotonates/pharmacology , Glucose/metabolism , Insulin/pharmacology , Muscle Proteins , 3T3 Cells , Adipocytes/drug effects , Animals , Biological Transport/drug effects , Deoxyglucose/metabolism , Drug Synergism , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Mice , Monosaccharide Transport Proteins/physiology , Oxidation-Reduction/drug effects , Palmitates/antagonists & inhibitors , Palmitates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...