Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmacogenomics J ; 5(5): 298-304, 2005.
Article in English | MEDLINE | ID: mdl-16027736

ABSTRACT

Several studies have reported on structural abnormalities, decreased myelination and oligodendrocyte dysfunction in post-mortem brains from schizophrenic patients. Glia-derived cholesterol is essential for both myelination and synaptogenesis in the CNS. Lipogenesis and myelin synthesis are thus interesting etiological candidate targets in schizophrenia. Using a microarray approach, we here demonstrate that the antipsychotic drugs clozapine and haloperidol upregulate several genes involved in cholesterol and fatty acid biosynthesis in cultured human glioma cells, including HMGCR (3-hydroxy-3-methylglutaryl-coenzyme A reductase), HMGCS1 (3-hydroxy-3-methylglutaryl-coenzyme A synthase-1), FASN (fatty acid synthase) and SCD (stearoyl-CoA desaturase). The changes in gene expression were followed by enhanced HMGCR-enzyme activity and elevated cellular levels of cholesterol and triglycerides. The upregulated genes are all known to be controlled by the sterol regulatory element-binding protein (SREBP) transcription factors. We show that clozapine and haloperidol both activate the SREBP system. The antipsychotic-induced SREBP-mediated increase in glial cell lipogenesis could represent a novel mechanism of action, and may also be relevant for the metabolic side effects of antipsychotics.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Fatty Acid Synthases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Haloperidol/pharmacology , Hydroxymethylglutaryl-CoA Synthase/genetics , Cell Line, Tumor , Cholesterol/biosynthesis , Cholesterol/genetics , Fatty Acid Synthases/metabolism , Fatty Acids/biosynthesis , Fatty Acids/genetics , Gene Expression Profiling , Glioma , Humans , Hydroxymethylglutaryl-CoA Synthase/metabolism , Oligonucleotide Array Sequence Analysis , RNA/metabolism , Schizophrenia/drug therapy , Schizophrenia/genetics , Time Factors , Up-Regulation
2.
Biochimie ; 87(1): 15-20, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15733731

ABSTRACT

Much data indicates that lowering of plasma triglyceride levels by hypolipidemic agents is caused by a shift in the liver metabolism towards activation of peroxisome proliferator activated receptor (PPAR)alpha-regulated fatty acid catabolism in mitochondria. Feeding rats with lipid lowering agents leads to hypolipidemia, possibly by increased channeling of fatty acids to mitochondrial fatty acid oxidation at the expense of triglyceride synthesis. Our hypothesis is that increased hepatic fatty acid oxidation and ketogenesis drain fatty acids from blood and extrahepatic tissues and that this contributes significantly to the beneficial effects on fat mass accumulation and improved peripheral insulin sensitivity. To investigate this theory we employ modified fatty acids that change the plasma profile from atherogenic to cardioprotective. One of these novel agents, tetradecylthioacetic acid (TTA), is of particular interest due to its beneficial effects on lipid transport and utilization. These hypolipidemic effects are associated with increased fatty acid oxidation and altered energy state parameters of the liver. Experiments in PPAR alpha-null mice have demonstrated that the effects hypolipidemic of TTA cannot be explained by altered PPAR alpha regulation alone. TTA also activates the other PPARs (e.g., PPAR delta) and this might compensate for deficiency of PPAR alpha. Altogether, TTA-mediated clearance of blood triglycerides may result from a lowered level of apo C-III, with a subsequently induction of hepatic lipoprotein lipase activity and (re)uptake of fatty acids from very low density lipoprotein (VLDL). This is associated with an increased hepatic capacity for fatty acid oxidation, causing drainage of fatty acids from the blood stream. This can ultimately be linked to hypolipidemia, anti-adiposity, and improved insulin sensitivity.


Subject(s)
Fatty Acids/metabolism , Liver/physiopathology , Metabolic Syndrome/physiopathology , Animals , Fatty Acids, Nonesterified/blood , Humans , Metabolic Syndrome/prevention & control , Mitochondria, Liver/metabolism , Obesity/physiopathology , Oxidation-Reduction , PPAR alpha/physiology , Signal Transduction , Sulfides/pharmacology , Triglycerides/blood
3.
Leukemia ; 16(11): 2292-301, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12399975

ABSTRACT

The lipid metabolism is important in the regulation of cell proliferation. We have examined effects of a fatty acid analogue, tetradecylthioacetic acid (TTA), on the functional phenotype of native, human AML cells. TTA inhibited AML blast proliferation in the presence of single cytokines (GM-CSF and SCF: P > 0.05, 35 patients with detectable proliferation) and a combination of cytokines (P < 0.005, n = 21). This antiproliferative effect was generally stronger than for the normal fatty acid palmitic acid (PA). Both TTA and PA increased the secretion of tumor necrosis factor alpha (TNFalpha) (P < 0.05, 27 patients with detectable cytokine release), but only PA increased interleukein 1beta (IL-1beta) release (P < 0.005, n = 34). AML blast populations varied significantly in their levels and activities of metabolites and enzymes characterizing oxidative status and fatty acid metabolism, and there was no significant correlation between the intrinsic oxidative status and the effects of PA and TTA on blast proliferation. Although TTA reduced the proliferation of mitogen-stimulated normal T cells derived from healthy individuals (P < 0.05, n = 8), no adverse effects were seen on peripheral blood cell counts (reticulocytes, platelets, total white blood cells, differential leukocyte counts) for healthy volunteers receiving TTA (oral administration of 1000 mg/day for 7 consecutive days). Our results suggest that TTA can inhibit AML blast proliferation through pathways that are unrelated to autocrine cytokine secretion and intrinsic oxidative status.


Subject(s)
Antioxidants/pharmacology , Cell Division/drug effects , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid/pathology , Palmitic Acid/pharmacology , Sulfides/pharmacology , Acute Disease , Adult , Aged , Aged, 80 and over , Blast Crisis/pathology , Blood Cell Count , Carnitine O-Palmitoyltransferase/metabolism , Female , Glutathione/metabolism , Humans , Interleukin-1/metabolism , Interleukin-6/metabolism , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/metabolism , Lymphocyte Activation/drug effects , Male , Middle Aged , Oxidation-Reduction , Oxidoreductases/metabolism , Superoxide Dismutase/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
4.
Carcinogenesis ; 22(11): 1747-55, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11698335

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) are transcription factors involved in fatty acid metabolism and energy homeostasis. The PPARs also play crucial roles in the control of cellular growth and differentiation. Especially, the recently emerged concept of ligand-dependent PPARgamma-mediated inhibition of cancer cell proliferation through induction of G(1)-phase arrest and differentiation is of clinical interest to cancer therapy. Tetradecylthioacetic acid (TTA) is a sulphur-substituted saturated fatty acid analog with unique biochemical properties. In this study, we investigated the effects of TTA-administration on cell proliferation in glioma cancer models. The rat glioma cell line BT4Cn, whether grown in culture or implanted in rats, expressed significant levels of PPARgamma and PPARdelta, with PPARgamma being the predominant PPAR subtype. In BT4Cn cells, TTA activated all PPAR subtypes in a dose-dependent manner. In cell culture experiments, the PPARgamma-selective ligand BRL49653 moderately inhibited growth of BT4Cn cells, whereas administration of TTA resulted in a marked growth inhibition. Administration of the PPARgamma-selective antagonist GW9662 abolished BRL49653-induced growth inhibition, but only marginally reduced the effect of TTA. TTA reduced tumor growth and increased the survival time of rats with implanted BT4Cn tumor. TTA-induced apoptosis in BT4Cn cells, and the administration of TTA led to cytochrome c release from mitochondria and increased the glutathione content in glioma cells. In conclusion, our results indicate that TTA inhibits proliferation of glioma cancer cells through both PPARgamma-dependent and PPARgamma-independent pathways, of which the latter appears to predominate.


Subject(s)
Antioxidants/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfides/pharmacology , Transcription Factors/metabolism , Tumor Cells, Cultured/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Division/drug effects , Cytochrome c Group/metabolism , DNA Primers/chemistry , Gene Expression/drug effects , Glioma/metabolism , Glioma/pathology , Glutathione/metabolism , In Vitro Techniques , Ligands , Male , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Plasmids , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured/physiology
5.
Lipids ; 36(3): 305-13, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11337987

ABSTRACT

Antiproliferative properties of molecular regulators of lipid metabolism have been increasingly studied during recent years. Discussion is ongoing concerning optimal treatment conditions and assays used for monitoring proliferation and cytotoxicity. The objective of the present work was to optimize methods and treatment conditions used for studying antiproliferative effects of fatty acids and analogs, represented by palmitic acid (PA) and the beta-oxidation-restricted fatty acid analog tetradecylthioacetic acid (TTA), in rat (BT4Cn) and human (D54Mg and GaMg) glioma cell lines. Changes in [3H]thymidine incorporation preceded changes in cell number in TTA-treated glioma cell cultures, and the growth inhibition was more significantly expressed by [3H]thymidine incorporation than cell number. Addition of bovine serum albumin decreased cellular fatty acid uptake and reduced the effects of TTA and PA on [3H]thymidine incorporation. Determination of the antiproliferative effect of TTA in BT4Cn cells by MTT conversion and [3H]thymidine incorporation yielded concordant results. TTA-mediated reduction in cell number corresponded to reduction in cellular protein and total DNA content in BT4Cn cells. Reduced growth potential in TTA-treated multicellular D54Mg and GaMg spheroids supported the findings from monolayer cultures. In conclusion, cell density, treatment period, fatty acid administration, and methods for growth determination may profoundly influence the outcome of cell growth experiments. Thus, experimental conditions should be carefully controlled when performing cell growth experiments, and effects on cell growth should preferably be confirmed by different methods.


Subject(s)
Cell Division/drug effects , Fatty Acids/pharmacology , Animals , Cell Count , DNA/biosynthesis , Fatty Acids/administration & dosage , Glioma/pathology , Humans , Kinetics , Palmitic Acid/pharmacology , Proteins/analysis , Rats , Serum Albumin, Bovine/metabolism , Serum Albumin, Bovine/pharmacology , Sodium Hydroxide/pharmacology , Sulfides/pharmacology , Tumor Cells, Cultured
6.
Biochem Pharmacol ; 61(6): 639-49, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11266648

ABSTRACT

During aerobic metabolism, a small amount of partially reduced oxygen is produced, yielding reactive oxygen species (ROS). Peroxisomes and mitochondria are major contributors to cellular ROS production, which is normally balanced by consumption by antioxidants. The fatty acid analogue tetradecylthioacetic acid (TTA) promotes mitochondrial and peroxisomal proliferation, and may induce oxidative stress and change the growth potential of cancer cells. In the present study, we found that TTA reduced [(3)H]thymidine incorporation in the glioma cell lines BT4Cn (rat), D54Mg (human), and GaMg (human) in a dose- and time-dependent manner. The 50% inhibitory TTA doses were approximately 125 microM for BT4Cn and D54Mg cells and 40 microM for GaMg cells after 4 days. alpha-Tochopherol counteracted this inhibition in GaMg cells. TTA enhanced the oxidation of [1-(14)C]palmitic acid, which could be explained by stimulation of enzymes involved in peroxisomal (fatty acyl-CoA oxidase) and/or mitochondrial (carnitine palmitoyltransferase) fatty acid oxidation. The glutathione content and the activities of glutathione peroxidase, glutathione reductase, and glutathione S-transferase were differentially affected. Increased malondialdehyde (MDA) production was seen in TTA-treated GaMg and D54Mg cells, but not in BT4Cn cells, in vitro. In BT4Cn tumor tissue from TTA-treated rats, MDA was increased while the alpha-tocopherol content tended to decrease. TTA increased the level of cytosolic cytochrome c in BT4Cn cells, which suggests induction of apoptotic cascades. Although several mechanisms are likely to be involved in the TTA-mediated effects on growth, we propose that modulation of cellular redox conditions caused by changes in fatty acid metabolism may be of vital importance.


Subject(s)
Antineoplastic Agents/pharmacology , Fatty Acids/metabolism , Glioma/metabolism , Sulfides/pharmacology , Animals , Antioxidants/pharmacology , Carnitine O-Palmitoyltransferase/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Cell Division/drug effects , Cytochrome c Group/metabolism , Cytosol/enzymology , Glioma/pathology , Glutathione/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Peroxisomes/drug effects , Peroxisomes/metabolism , Rats , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
7.
Biochem J ; 343 Pt 1: 191-7, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10493929

ABSTRACT

Hypolipidaemic fatty acid derivatives and polyunsaturated fatty acids decrease concentrations of plasma triacylglycerol by mechanisms that are not fully understood. Because poor susceptibility to beta- and/or omega-oxidation is apparently a determinant of the peroxisome proliferating and hypolipidaemic capacity of fatty acids and derivatives, the relative importance of activation of the peroxisome-proliferator-activated receptor alpha (PPARalpha), fatty acid oxidation and triacylglycerol synthesis were examined. We have compared the effects of differentially beta-oxidizable fatty acids on these parameters in primary cultures of rat hepatocytes. Tetradecylthioacetic acid (TTA), 2-methyleicosapentaenoic acid and 3-thia-octadecatetraenoic acid, which are non-beta-oxidizable fatty acid derivatives, were potent activators of a glucocorticoid receptor (GR)-PPARalpha chimaera. This activation was paradoxically reflected in an substantially increased oxidation of [1-(14)C]palmitic acid and/or oleic acid. The incorporation of [1-(14)C]palmitic acid and/or oleic acid into cell-associated and secreted triacylglycerol was decreased by 15-20% and 30% respectively with these non-beta-oxidizable fatty acid derivatives. The CoA ester of TTA inhibited the esterification of 1, 2-diacylglycerol in rat liver microsomes. Both eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) activated GR-PPARalpha. EPA increased the oxidation of [1-(14)C]palmitic acid but DHA had no effect. The CoA ester of EPA inhibited the esterification of 1, 2-diacylglycerol, whereas DHA-CoA had no effect. The ratio between synthesized triacylglycerol and diacylglycerol was lower in hepatocytes cultured with EPA in the medium compared with DHA or oleic acid, indicating a decreased conversion of diacylglycerol to triacylglycerol. Indeed, the incorporation of [1-(14)C]oleic acid into secreted triacylglycerol was decreased by 20% in the presence of EPA. In conclusion, a decreased availability of fatty acids for triacylglycerol synthesis by increased mitochondrial beta-oxidation and decreased triacylglycerol formation caused by inhibition of diacylglycerol acyltransferase might explain the hypolipidaemic effect of TTA and EPA.


Subject(s)
Acyltransferases/antagonists & inhibitors , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Fatty Acids/metabolism , Liver/drug effects , Triglycerides/biosynthesis , Animals , Diacylglycerol O-Acyltransferase , Docosahexaenoic Acids/chemistry , Eicosapentaenoic Acid/chemistry , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/pharmacology , Liver/enzymology , Liver/metabolism , Male , Oxidation-Reduction , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/agonists , Recombinant Fusion Proteins/agonists , Transcription Factors/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...