Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Ageing Res Rev ; 68: 101324, 2021 07.
Article in English | MEDLINE | ID: mdl-33774195

ABSTRACT

Age-related diseases for which there are no effective treatments include cardiovascular diseases; neurodegenerative diseases such as Alzheimer's disease; eye disorders such as cataract and age-related macular degeneration; and, more recently, Severe Acute Respiratory Syndrome (SARS-CoV-2). These diseases are associated with plasma and/or tissue increases in cholesterol derivatives mainly formed by auto-oxidation: 7-ketocholesterol, also known as 7-oxo-cholesterol, and 7ß-hydroxycholesterol. The formation of these oxysterols can be considered as a consequence of mitochondrial and peroxisomal dysfunction, leading to increased in oxidative stress, which is accentuated with age. 7-ketocholesterol and 7ß-hydroxycholesterol cause a specific form of cytotoxic activity defined as oxiapoptophagy, including oxidative stress and induction of death by apoptosis associated with autophagic criteria. Oxiaptophagy is associated with organelle dysfunction and in particular with mitochondrial and peroxisomal alterations involved in the induction of cell death and in the rupture of redox balance. As the criteria characterizing 7-ketocholesterol- and 7ß-hydroxycholesterol-induced cytotoxicity are often simultaneously observed in major age-related diseases (cardiovascular diseases, age-related macular degeneration, Alzheimer's disease) the involvement of these oxysterols in the pathophysiology of the latter seems increasingly likely. It is therefore important to better understand the signalling pathways associated with the toxicity of 7-ketocholesterol and 7ß-hydroxycholesterol in order to identify pharmacological targets, nutrients and synthetic molecules attenuating or inhibiting the cytotoxic activities of these oxysterols. Numerous natural cytoprotective compounds have been identified: vitamins, fatty acids, polyphenols, terpenes, vegetal pigments, antioxidants, mixtures of compounds (oils, plant extracts) and bacterial enzymes. However, few synthetic molecules are able to prevent 7-ketocholesterol- and/or 7ß-hydroxycholesterol-induced cytotoxicity: dimethyl fumarate, monomethyl fumarate, the tyrosine kinase inhibitor AG126, memantine, simvastatine, Trolox, dimethylsufoxide, mangafodipir and mitochondrial permeability transition pore (MPTP) inhibitors. The effectiveness of these compounds, several of which are already in use in humans, makes it possible to consider using them for the treatment of certain age-related diseases associated with increased plasma and/or tissue levels of 7-ketocholesterol and/or 7ß-hydroxycholesterol.


Subject(s)
COVID-19 , Aging , Humans , Hydroxycholesterols , Ketocholesterols , Nutrients , Oils , SARS-CoV-2
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(4): 567-576, 2019 04.
Article in English | MEDLINE | ID: mdl-30312667

ABSTRACT

Acyl-CoA oxidase 1 (ACOX1) deficiency is a rare and severe peroxisomal leukodystrophy associated with a very long-chain fatty acid (VLCFA) ß-oxidation defect. This neurodegenerative disease lacks relevant cell models to further decipher the pathomechanisms in order to identify novel therapeutic targets. Since peroxisomal defects in microglia appear to be a key component of peroxisomal leukodystrophies, we targeted the Acox1 gene in the murine microglial BV-2 cell line. Using CRISPR/Cas9 gene editing, we generated an Acox1-deficient cell line and validated the allelic mutations, which lead to the absence of ACOX1 protein and enzymatic activity. The activity of catalase, the enzyme degrading H2O2, was increased, likely in response to the alteration of redox homeostasis. The mutant cell line grew more slowly than control cells without obvious morphological changes. However, ultrastructural analysis revealed an increased number of peroxisomes and mitochondria associated with size reduction of mitochondria. Changes in the distribution of lipid droplets containing neutral lipids have been observed in mutant cells; lipid analysis revealed the accumulation of saturated and monounsaturated VLCFA. Besides, expression levels of genes encoding interleukin-1 beta and 6 (IL-1ß and IL-6), as well as triggering receptor expressed on myeloid cells 2 (Trem2) were found modified in the mutant cells suggesting modification of microglial polarization and phagocytosis ability. In summary, this Acox1-deficient cell line presents the main biochemical characteristics of the human disease and will serve as a promising model to further investigate the consequences of a specific microglial peroxisomal ß-oxidation defect on oxidative stress, inflammation and cellular functions.


Subject(s)
Acyl-CoA Oxidase/deficiency , Microglia/cytology , Models, Biological , Mutation , Neurodegenerative Diseases/genetics , Acyl-CoA Oxidase/genetics , Animals , CRISPR-Cas Systems , Cell Line , Cell Proliferation , Fatty Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Editing , Hydrogen Peroxide/metabolism , Mice , Microglia/metabolism , Oxidative Stress
3.
Environ Toxicol ; 29(11): 1245-61, 2014 Nov.
Article in English | MEDLINE | ID: mdl-23450722

ABSTRACT

Mussels may concentrate pollutants, with possibly significant side effects on human health. Therefore, mussels (Mytilus galloprovincialis) from two sites of the Moroccan Atlantic coast (Jorf Lasfar [JL], an industrial site, and Oualidia [OL], a vegetable-growing area), were subjected to biochemical analyses to quantify the presence of heavy metals (Cd, Cr, and Pb) and to establish the lipid profile: fatty acid, cholesterol, oxysterol, phytosterol and phospholipid content. In addition, mussel lipid extracts known to accumulate numerous toxic components were tested on murine pancreatic ß-cells (MIN6), and their biological activities were measured with various flow cytometric and biochemical methods to determine their impacts on cell death induction, organelle dysfunctions (mitochondria, lysosomes, and peroxisomes), oxidative stress and insulin secretion. The characteristics of JL and OL lipid extracts were compared with those of commercially available mussels from Spain (SP) used for human consumption. OL and JL contained heavy metals, high amounts of phospholipids, and high levels of oxysterols; the [(unsaturated fatty acids)/(saturated fatty acids)] ratio, which can be considered a sign of environmental stress leading to lipid peroxidation, was low. On MIN6 cells, JL and OL lipid extracts were able to trigger cell death. This event was associated with overproduction of H2 O2 , increased catalase activity, a decreased GSH level, lipid peroxidation and stimulation of insulin secretion. These effects were not observed with SP lipid extracts. These data suggest that some components from OL and JL lipid extracts might predispose to pancreatic dysfunctions. Epidemiological studies would be needed to assess the global risk on human health and the metabolic disease incidence in a context of regular seafood consumption from the OL and JL areas.


Subject(s)
Cadmium/toxicity , Chromium/toxicity , Insulin-Secreting Cells/drug effects , Lead/toxicity , Lipid Metabolism , Mytilus/metabolism , Animals , Cadmium/metabolism , Catalase/metabolism , Chromium/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Lead/metabolism , Lipid Peroxidation/drug effects , Mice , Morocco , Oxidative Stress/drug effects , Spain , Tissue Extracts/metabolism
4.
Neuroscience ; 213: 1-18, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22521832

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) and pseudo neonatal adrenoleukodystrophy (P-NALD) are neurodegenerative demyelinating diseases resulting from the functional loss of the peroxisomal ATP-binding cassette transporter D (ABCD1) and from single peroxisomal enzyme deficiency (Acyl-CoA oxidase1: ACOX1), respectively. As these proteins are involved in the catabolism of very long chain fatty acids (VLCFA: C24:0, C26:0), X-ALD and P-NALD patients are characterized by the accumulation of VLCFA in plasma and tissues. Since peroxisomes are involved in the metabolism of reactive oxygen species (ROS) and nitrogen species (RNS), we examined the impact of VLCFA on the oxidative status of 158N murine oligodendrocytes expressing or not Abcd1 or Acox1. VLCFA triggers an oxidative stress characterized by an overproduction of ROS and RNS associated with lipid peroxidation, protein carbonylation, increased superoxide dismutase (SOD) activity, decreased catalase activity and glutathione level. SiRNA knockdown of Abcd1 or Acox1 increased ROS and RNS production even in the absence of VLCFA, and especially potentialized VLCFA-induced ROS overproduction. Moreover, mainly in cells with reduced Acox1 level, the levels of VLCFA and neutral lipids were strongly enhanced both in untreated and VLCFA - treated cells. Our data obtained on 158N murine oligodendrocytes highlight that VLCFA induce an oxidative stress, and demonstrate that Abcd1 or Acox1 knockdown contributes to disrupt RedOx equilibrium supporting a link between oxidative stress and the deficiency of Abcd1 or Acox1 peroxisomal proteins.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Acyl-CoA Oxidase/metabolism , Fatty Acids/metabolism , Oligodendroglia/metabolism , Oxidative Stress/physiology , RNA Interference , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , Acyl-CoA Oxidase/genetics , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/metabolism , Animals , Blotting, Western , Cells, Cultured , Fatty Acids/pharmacology , Flow Cytometry , Gas Chromatography-Mass Spectrometry , Gene Knockdown Techniques , Mice , Oligodendroglia/drug effects , Oxidation-Reduction , Peroxisomes/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
5.
J Cell Mol Med ; 7(3): 238-48, 2003.
Article in English | MEDLINE | ID: mdl-14594548

ABSTRACT

This paper reviews aspects concerning the genetic regulation of the expression of the well studied peroxisomal genes including those of fatty acid beta-oxidation enzymes; acyl-CoA oxidase, multifunctional enzyme and thiolase from different tissues and species. An important statement is PPARalpha, which is now long known to be in rodents the key nuclear receptor orchestrating liver peroxisome proliferation and enhanced peroxisomal beta-oxidation, does not appear to control so strongly in man the expression of genes involved in peroxisomal fatty acid beta-oxidation related enzymes. In this respect, the present review strengthens among others the emerging concept that, in the humans, the main genes whose expression is up-regulated by PPARalpha are mitochondrial and less peroxisomal genes. A special emphasis is also made on the animal cold adaptation and on need for sustained study of peroxisomal enzymes and genes; challenging that some essential roles of peroxisomes in cell function and regulation still remain to be discovered.


Subject(s)
Peroxisomes/genetics , Peroxisomes/metabolism , Adaptation, Biological , Animals , Fatty Acids/metabolism , Humans , Lipid Metabolism , Peroxisomes/enzymology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
6.
Biochem Soc Trans ; 29(Pt 2): 305-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11356172

ABSTRACT

The enzymes required for the beta-oxidation of fatty acyl-CoA are present in peroxisomes and mitochondria. Administration of hypolipidaemic compounds such as clofibrate to rodents leads to an increase in the volume and density of peroxisomes in liver cells. These proliferators also induce simultaneously the expression of genes encoding acyl-CoA oxidase, enoyl-CoA hydratase-hydroxyacyl-CoA dehydrogenase (multifunctional enzyme) and thiolase (3-ketoacyl-CoA thiolase). All these enzymes are responsible for long-chain and very-long-chain fatty acid beta-oxidation in peroxisomes. Similar results were observed when rat hepatocytes, or liver-derived cell lines, were cultured with a peroxisome proliferator. The increased expression of these genes is due to the stimulation of their transcription rate. These results show that the peroxisome proliferators act on the hepatic cells and regulate the transcription through various cellular components and pathways, including peroxisome-proliferator-activated receptor alpha (PPARalpha). After activation by specific ligands, either fibrates or fatty acid derivatives, PPARalpha binds to a DNA response element: peroxisome-proliferator-responsive element (PPRE), which is a direct repeat of the following consensus sequence: TGACCTXTGACCT, found in the promoter region of the target genes. PPARalpha is expressed mainly in liver, intestine and kidney. PPARalpha is a transcriptional factor, which requires other nuclear proteins for function including retinoic acid X receptor (RXRalpha) and other regulatory proteins. From our results and others we suggest the role of PPARalpha in the regulation of the peroxisomal fatty acid beta-oxidation. In this regard, we showed that although PPARalpha binds to thiolase B gene promoter at -681 to -669, a better response is observed with hepatic nuclear factor 4 ("HNf-4"). Moreover, rat liver PPARalpha regulatory activity is dependent on its phosphorylated state. In contrast, a protein-kinase-C-mediated signal transduction pathway seems to be modified by peroxisome proliferators, leading to an increase in the phosphorylation level of specific proteins, some of which have been shown to be involved in the phosphoinositide metabolism.


Subject(s)
Fatty Acids/metabolism , Peroxisomes/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Gene Expression Regulation, Enzymologic , Guinea Pigs , Liver/cytology , Liver/metabolism , Mice , Peroxisomes/enzymology , Peroxisomes/genetics , Phosphorylation , Protein Kinase C/metabolism , Rats , Signal Transduction , Transcriptional Activation
7.
Gene Expr ; 9(6): 291-304, 2001.
Article in English | MEDLINE | ID: mdl-11764000

ABSTRACT

Peroxisome proliferators, which function as peroxisome proliferator-activated receptor-alpha (PPARalpha) agonists, are a group of structurally diverse nongenotoxic hepatocarcinogens including the fibrate class of hypolipidemic drugs that induce peroxisome proliferation in liver parenchymal cells. Sustained activation of PPARalpha by these agents leads to the development of liver tumors in rats and mice. To understand the molecular mechanisms responsible for the pleiotropic effects of these agents, we have utilized the cDNA microarray to generate a molecular portrait of gene expression in the liver of mice treated for 2 weeks with Wy-14,643, a potent peroxisome proliferator. PPARalpha activation resulted in the stimulation of expression (fourfold or greater) of 36 genes and decreased the expression (fourfold or more decrease) of 671 genes. Enhanced expression of several genes involved in lipid and glucose metabolism and many other genes associated with peroxisome biogenesis, cell surface function, transcription, cell cycle, and apoptosis has been observed. These include: CYP2B9, CYP2B10, monoglyceride lipase, pyruvate dehydrogenase-kinase-4, cell death-inducing DNA-fragmentation factor-alpha, peroxisomal biogenesis factor 11beta, as well as several cell recognition surface proteins including annexin A2, CD24, CD39, lymphocyte antigen 6, and retinoic acid early transcript-gamma, among others. Northern blotting of total RNA extracted from the livers of PPARalpha-/- mice and from mice lacking both PPARalpha and peroxisomal fatty acyl-CoA oxidase (AOX), that were fed control and Wy-14,643-containing diets for 2 weeks, as well as time course of induction following a single dose of Wy-14,643, revealed that upregulation of genes identified by microarray procedure is dependent upon peroxisome proliferation vis-à-vis PPARalpha. However, cell death-inducing DNA-fragmentation factor-alpha mRNA, which is increased in the livers of wild-type mice treated with peroxisome proliferators, was not enhanced in AOX-/- mice with spontaneous peroxisome proliferation. These observations indicate that the activation of PPARalpha leads to increased and decreased expression of many genes not associated with peroxisomes, and that delayed onset of enhanced expression of some genes may be the result of metabolic events occurring secondary to PPARalpha activation and alterations in lipid metabolism.


Subject(s)
Liver/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Animals , DNA, Complementary , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrimidines/pharmacology
8.
Biochem Pharmacol ; 60(8): 1027-32, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11007938

ABSTRACT

The first PPAR (peroxisome proliferator-activated receptor) was cloned in 1990 by Issemann and Green (Nature 347:645-650). This nuclear receptor was so named since it is activated by peroxisome proliferators including several drugs of the fibrate family, plasticizers, and herbicides. This receptor belongs to the steroid receptor superfamily. After activation by a specific ligand, it binds to a DNA response element, PPRE (peroxisome proliferator response element), which is a DR-1 direct repeat of the consensus sequence TGACCT x TGACCT. This mechanism leads to the transcriptional activation of target genes (Motojima et al., J Biol Chem 273:16710-16714, 1998). After the first discovery, several isoforms were characterized in most of the vertebrates investigated. PPAR alpha, activated by hypolipidemic agents of the fibrate family or by leukotrienes; regulates lipid metabolism as well as the detoxifying enzyme-encoding genes. PPAR beta/delta, which is not very well known yet, appears to be more specifically activated by fatty acids. PPAR gamma (subisoforms 1, 2, 3) is activated by the prostaglandin PGJ2 or by antidiabetic thiazolidinediones (Vamecq and Latruffe, Lancet 354:411-418, 1999). This latter isoform is involved in adipogenesis. The level of PPAR expression is largely dependent on the tissue type. PPAR alpha is mainly expressed in liver and kidney, while PPAR beta/delta is almost constitutively expressed. In contrast, PPAR gamma is largely expressed in white adipose tissue. PPAR is a transcriptional factor that requires other nuclear proteins in order to function, i.e. RXRalpha (9-cis-retinoic acid receptor alpha) in all cases in addition to other regulatory proteins. Peroxisomes are specific organelles for very long-chain and polyunsaturated fatty acid catabolism. From our results and those of others, the inventory of the role of PPAR alpha in the regulation of peroxisomal fatty acid beta-oxidation is presented. In relation to this, we showed that PPAR alpha activates peroxisomal beta-oxidation-encoding genes such as acyl-CoA oxidase, multifunctional protein, and thiolase (Bardot et al., FEBS Lett 360:183-186, 1995). Moreover, rat liver PPAR alpha regulatory activity is dependent on its phosphorylated state (Passilly et al., Biochem Pharmacol 58:1001-1008, 1999). On the other hand, some signal transduction pathways such as protein kinase C are modified by peroxisome proliferators that increase the phosphorylation level of some specific proteins (Passilly et al. Eur J Biochem 230:316-321, 1995). From all these findings, PPAR alpha and kinases appear to play an important role in lipid homeostasis.


Subject(s)
Peroxisome Proliferators/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Protein Kinase C/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Transcription Factors/genetics , Transcriptional Activation
9.
Oncol Rep ; 7(4): 847-52, 2000.
Article in English | MEDLINE | ID: mdl-10854556

ABSTRACT

Resveratrol is a polyphenolic compound especially produced by grapevine and consequently found in wine. Based on epidemiological studies resveratrol may act as a cancer chemopreventive compound. The ability of resveratrol to inhibit cell proliferation was studied in rat hepatoma Fao cell line and human hepatoblastoma HepG2 cell line. The results show that resveratrol strongly inhibits cell proliferation at the micromolar range in a time- and dose-dependent manner. Concentrations higher than 50 microM become toxic. Fao cells are more sensitive than HepG2 cells. Interestingly, the presence of ethanol lowers the threshold of resveratrol effect. Resveratrol appears to prevent or to delay the entry to mitosis since no inhibition of [3H]thymidine incorporation is observed, while there is an increase of cell number in S and G2/M phases. In conclusion, resveratrol shows a strong inhibition of hepatic cell proliferation where alcohol may act as an enhancing agent.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Cell Cycle/drug effects , Stilbenes/toxicity , Animals , Cell Division/drug effects , Cell Survival/drug effects , Hepatoblastoma , Humans , Liver Neoplasms , Liver Neoplasms, Experimental , Mitosis/drug effects , Rats , Resveratrol , Tumor Cells, Cultured
10.
Lipids ; 35(12): 1397-404, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11202002

ABSTRACT

The first peroxisome proliferator-activated receptor (PPAR) was cloned in 1990 by Issemann and Green. Many studies have reported the importance of this receptor in the control of gene expression of enzymes involved in lipid metabolic pathways including mitochondrial and peroxisomal fatty acid beta-oxidation, lipoprotein structure [apolipoprotein (apo) A2, apo CIII], and fatty acid synthase. By using radiolabeled molecules, it was shown that peroxisome proliferators bind and activate PPAR. As an alternative method, we developed a fluorescent dansyl (1-dimethylaminonaphthalene-5-sulfonyl) derivative peroxisome proliferator from bezafibrate (DNS-X), a hypolipidemic agent that exhibits an in vitro peroxisome proliferative activity on rat Fao-hepatic derived cultured cells. However, until now, the effect of this new compound on the liver of animals and subcellular localization was unknown. In addition to in vivo rat studies, we present a more efficient large-scale technique of DNS-X purification. Treating rats (DNS-X in the diet at 0.3% w/w) for 6 d leads to a hepatomegaly and a marked increase in liver peroxisomal palmitoyl-CoA oxidase activity. We also developed a method to localize and quantify DNS-X in tissues or cell compartment organelles. The primarily cytosolic distribution of DNS-X was confirmed by direct visualization using fluorescence microscopy of cultured Fao cells. Finally, transfection assay demonstrated that DNS-X enhanced the PPAR alpha activity as well as other peroxisome proliferators do.


Subject(s)
Bezafibrate/chemistry , Fatty Acids/metabolism , Fluorescent Dyes/chemistry , Oxygen/metabolism , Animals , Bezafibrate/analogs & derivatives , Bezafibrate/pharmacology , Cell Division , Cells, Cultured , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Cytosol/metabolism , Hepatomegaly , Kinetics , Liver/cytology , Liver/enzymology , Magnetic Resonance Spectroscopy , Male , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Models, Chemical , Oxidoreductases/metabolism , Peroxisome Proliferators/metabolism , Peroxisomes/enzymology , Plasmids , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors , Titrimetry , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
11.
Cell Biochem Biophys ; 32 Spring: 213-20, 2000.
Article in English | MEDLINE | ID: mdl-11330049

ABSTRACT

The goal of this study was to characterize phosphorylated proteins and to evaluate the changes in their phosphorylation level under the influence of a peroxisome proliferator (PP) with hypolipidemic activity of the fibrate family. The incubation of rat hepatic derived Fao cells with ciprofibrate leads to an overphosphorylation of proteins, especially one of 85 kDa, indicating that kinase (or phosphatase) activities are modified. Moreover, immunoprecipitation of 32P-labeled cell lysates shows that the nuclear receptor, PP-activated receptor, alpha isoform, can exist in a phosphorylated form, and its phosphorylation is increased by ciprofibrate. This study shows that PP acts at different steps of cell signaling. These steps can modulate gene expression of enzymes involved in fatty acid metabolism and lipid homeostasis, as well as in detoxication processes.


Subject(s)
Lipid Metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Gene Expression Regulation , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/genetics , Transcription Factors/genetics
12.
Biochem Pharmacol ; 58(6): 1001-8, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10509752

ABSTRACT

The basic mechanism(s) by which peroxisome proliferators activate peroxisome proliferator-activated receptors (PPARs) is (are) not yet fully understood. Given the diversity of peroxisome proliferators, several hypotheses of activation have been proposed. Among them is the notion that peroxisome proliferators could activate PPARs by changing their phosphorylation status. In fact, it is well known that several members of the nuclear hormone receptor superfamily are regulated by phosphorylation. In this report, we show that the rat Fao hepatic-derived cell line, known to respond to peroxisome proliferators, exhibited a high content of PPARalpha. Alkaline phosphatase treatment of Fao cell lysate as well as immunoprecipitation of PPARalpha from cells prelabeled with [32P] orthophosphate clearly showed that PPARalpha is indeed a phosphoprotein in vivo. Moreover, treatment of rat Fao cells with ciprofibrate, a peroxisome proliferator, increased the phosphorylation level of the PPARalpha. In addition, treatment of Fao cells with phosphatase inhibitors (okadaic acid and sodium orthovanadate) decreased the activity of ciprofibrate-induced peroxisomal acyl-coenzyme A oxidase, an enzyme encoded by a PPARalpha target gene. Our results suggest that the gene expression controlled by peroxisome proliferators could be mediated in part by a modulation of the PPARalpha effect via a modification of the phosphorylation level of this receptor.


Subject(s)
Clofibric Acid/analogs & derivatives , Peroxisome Proliferators/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Acyl-CoA Oxidase , Animals , Cell Line , Clofibric Acid/pharmacology , Enzyme Inhibitors/pharmacology , Fibric Acids , Gene Expression Regulation/drug effects , Microbodies/drug effects , Microbodies/enzymology , Oxidoreductases/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphorylation/drug effects , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
13.
Int J Mol Med ; 3(2): 163-8, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9917524

ABSTRACT

It is known that a short-term exposure of rat, mice or incubation of hepatic cells with fibrate molecules leads to increase in peroxisome number and cell hyperplasia. Further, long-term incubation of cells (at least a year) show transformed characteristics with foci and nodules. To explain the hepatocarcinogenic effect of peroxisome proliferators in rodents we studied the effect of peroxisome proliferators on rat liver oncogenes expression. Earlier, we reported an increase in liver and kidney mRNA level of c-myc and N-myc. Since several metabolic genes are activated by PPAR (peroxisome proliferators activated receptor) through a PPRE (peroxisome proliferator response element), we suggest the involvment of PPAR in oncogene activation, because of the presence of PPRE in the N-myc 5'-upstream region. We showed by flow cytometric analysis that ciprofibrate increased the size of rat Fao derived cell line and the activity of palmitoyl CoA oxidase, a peroxisome proliferation enzyme marker for studying peroxisome proliferation was increased. The above effects which can contribute to hepatocarcinogenesis seem to be restricted to rat and mice, which show strong response to peroxisome proliferators. Indeed, no changes are observed in weak responsive species such as humans (using hepatic derived cell lines) and guinea pig. These data provide arguments for the non-carcinogenic effect of this xenobiotic class in human especially when sensitive, or normal individuals are exposed either to hypolipidaemic agents of the fibrate family.


Subject(s)
Carcinogens/toxicity , Peroxisome Proliferators/toxicity , Xenobiotics/toxicity , Animals , Carcinogenicity Tests , Carcinogens/chemistry , Cell Division/drug effects , Cell Transformation, Neoplastic/chemically induced , Guinea Pigs , Hepatomegaly/etiology , Humans , Oncogenes/physiology , Peroxisome Proliferators/chemistry , Rats , Rodentia , Tumor Cells, Cultured , Xenobiotics/chemistry
14.
Biochem J ; 330 ( Pt 3): 1361-8, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9494107

ABSTRACT

After our previous report on the cloning of two cDNA species in guinea pig, both encoding the same hepatic 79 kDa multifunctional protein 1 (MFP-1) [Caira, Cherkaoui-Malki, Hoefler and Latruffe (1996) FEBS Lett. 378, 57-60], here we report the cloning of a cDNA encoding a second multifunctional peroxisomal protein (MFP-2) in guinea-pig liver. This 2356 nt cDNA encodes a protein of 735 residues (79.7 kDa) whose sequence shows 83% identity with rat MFP-2 [Dieuaide-Noubhani, Novikov, Baumgart, Vanhooren, Fransen, Goethals, Vandekerckhove, Van Veldhoven and Mannaerts (1996) Eur. J. Biochem. 240, 660-666]. In parallel, we studied the effect of ciprofibrate, a hypolipaemic agent also known as peroxisome proliferator in rodent, on the expression of MFP-1 and MFP-2 (2.6 kb) in rats and guinea pigs. By Northern blotting analysis we demonstrated that three MFP-1-related mRNA species are expressed in the guinea-pig liver. The expression of two of them (3.5 and 2.6 kb) is slightly increased by ciprofibrate, whereas the 3.0 kb MFP-1 mRNA is, unlike the rat one, strongly down-regulated in guinea pigs treated with ciprofibrate. In a similar way, the hepatic expression of the guinea-pig 2.6 kb MFP-2 mRNA is also down-regulated in guinea pigs treated with ciprofibrate. These results demonstrate (1) that in contrast with the unique 3.0 kb MFP-1 rat mRNA, at least three hepatic MFP-1-related mRNA species are co-expressed in guinea pig; and (2) that, opposed to the accepted idea of non-responsiveness of the guinea pig to ciprofibrate, this drug affects MFP-1 and MFP-2 gene expression in this species. Also, the mRNA species for acyl-CoA oxidase and thiolase, two other enzymes of the peroxisomal beta-oxidation pathway that are induced severalfold in responsive species are down-regulated in guinea pig. This paper is the first, to our knowledge, reporting the down-regulation of the expression of genes encoding enzymes involved in the peroxisomal beta-oxidation of fatty acids (MFP-1) and bile acid synthesis (MFP-2) in mammals.


Subject(s)
Clofibric Acid/analogs & derivatives , Estradiol Dehydrogenases/biosynthesis , Gene Expression Regulation/drug effects , Liver/metabolism , Microbodies/physiology , Amino Acid Sequence , Animals , Base Sequence , Clofibric Acid/pharmacology , Cloning, Molecular , DNA, Complementary , Enoyl-CoA Hydratase/metabolism , Estradiol Dehydrogenases/chemistry , Fibric Acids , Guinea Pigs , Hypolipidemic Agents/pharmacology , Liver/drug effects , Male , Microbodies/drug effects , Microbodies/enzymology , Molecular Sequence Data , Oxidoreductases/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
16.
Eur J Cell Biol ; 70(2): 150-6, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8793387

ABSTRACT

This report presents new data on mammalian peroxisomes by studying an unusual rodent: the jerboa (Jaculus orientalis). This animal exhibits some unique peroxisomal properties compared to the rat, such as higher cyanide-insensitive palmitoyl-CoA oxidase specific activity, pattern differences in SDS-PAGE peroxisomal proteins as well as in acyl-CoA oxidase immunoblotting. There is also a peculiar response to a peroxisome proliferator, ciprofibrate. With 250 ppm of ciprofibrate in the diet for 2 weeks, we observed a limited liver peroxisome proliferation as well as a palmitoyl-CoA oxidase activity, enzyme content and mRNA increase. However, there was no increase in catalase activity, nor hepatomegaly which are prominent features of peroxisome proliferation in rats treated under the same conditions. The palmitoyl-CoA oxidase activity increase was weak in the kidney and not observed in the heart. Other subcellular organelle marker enzyme activities did not significantly change, especially the mitochondrial D-3-hydroxybutyrate and succinate dehydrogenases, lysosomal acid phosphatase, cytosolic lactate dehydrogenase and the endoplasmic reticulum NADPH-cytochrome c reductase. However, the activity of the liver membrane endoplasmic reticulum linked omega-lauryl hydroxylase (cytochrome P450 IV A1) increases after ciprofibrate treatment. Jerboa also behaves differently compared to the guinea pig after ciprofibrate treatment since the guinea pig has a weak response towards peroxisome proliferators. In conclusion, this first peroxisome study utilizing a different type of rodent as a laboratory animal, reveals that the jerboa shows unique peroxisome properties and responds in a moderate manner to a peroxisome proliferator, ciprofibrate, without leading to any increase in liver mass. This supports the fact that fibrate molecules may have different targets depending upon the species.


Subject(s)
Guinea Pigs/physiology , Liver/ultrastructure , Microbodies/physiology , Rats/physiology , Rodentia/physiology , Animals , Clofibric Acid/analogs & derivatives , Clofibric Acid/pharmacology , DNA/biosynthesis , Female , Fibric Acids , Heart/drug effects , Immunoblotting , Kidney/drug effects , Kidney/enzymology , Liver/drug effects , Liver/enzymology , Male , Microbodies/enzymology , Myocardium/enzymology , Oxidoreductases/metabolism , Proteins/analysis , Species Specificity
17.
FEBS Lett ; 378(1): 57-60, 1996 Jan 02.
Article in English | MEDLINE | ID: mdl-8549802

ABSTRACT

The 2-enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase (HD) is the second enzyme of the peroxisomal beta-oxidation pathway. In human and rat, only one HD mRNA has been so far detected in the liver. This paper reports for the first time in a mammal species, the guinea pig, the cloning and sequencing of two cDNAs encoding an HD. The 3,274 nucleotide-cDNA is a strictly identical but longer copy of the 2,494 nucleotide-form. A 2,178 bp-open reading frame encodes a protein of 726 amino acids (M(r) 79.3 kDa) with the peroxisomal-targeting signal (tripeptide SKL) at the carboxyterminus. Northern blot analysis of HD mRNA identified three mRNAs of respective sizes 3.5, 2.6 and 1.6 kb in the guinea pig liver and kidneys.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/genetics , DNA, Complementary/genetics , Enoyl-CoA Hydratase/genetics , Gene Expression , Liver/enzymology , Microbodies/enzymology , 3-Hydroxyacyl CoA Dehydrogenases/chemistry , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cloning, Molecular , DNA, Complementary/chemistry , Enoyl-CoA Hydratase/chemistry , Guinea Pigs , Kidney/enzymology , Liver/ultrastructure , Molecular Sequence Data , RNA, Messenger/analysis , Sequence Analysis, DNA
18.
Biochem Pharmacol ; 50(7): 1001-6, 1995 Sep 28.
Article in English | MEDLINE | ID: mdl-7575654

ABSTRACT

Peroxisome proliferators (PPs) are non-genotoxic carcinogens in rodents. Their reversible effects on rat liver have been studied with ciprofibrate and fenofibrate. We found that with the hypolipemic drug fenofibrate a pause of 28 days is sufficient for a return to normal status, whereas with the highly potent PP ciprofibrate, the stimulation of ACO mRNA levels remains after its withdrawal. We investigated the effects of the renewal of the treatment with PPs on other peroxisomal parameters and proto-oncogene expression using Wistar rats. Interestingly, c-myc expression was enhanced even upon drug withdrawal, and was more stimulated by the second exposure to ciprofibrate, while c-fos expression was unaltered. However, only slight differences in c-Ha-ras expression were observed. Therefore, the effects of PPs in the Wistar rats are not totally reversible within 28 days following withdrawal, depending on the drug used. These delayed effects of ciprofibrate could be a key to our understanding the hepatocarcinogenic effect of PPs in rodents.


Subject(s)
Clofibric Acid/analogs & derivatives , Liver/drug effects , Microbodies/drug effects , Proto-Oncogene Proteins/biosynthesis , Acyl-CoA Oxidase , Animals , Clofibric Acid/pharmacology , Fenofibrate/pharmacology , Fibric Acids , Liver/metabolism , Male , Microsomes, Liver/drug effects , Mitochondria, Liver/drug effects , Oxidoreductases/biosynthesis , Rats , Rats, Wistar , Time Factors
19.
Biochem Pharmacol ; 49(5): 611-9, 1995 Mar 01.
Article in English | MEDLINE | ID: mdl-7887975

ABSTRACT

The treatment of rats with ciprofibrate, a potent peroxisome proliferator, led to increased levels of the peroxisomal acyl-CoA oxidase (ACO) mRNA. How ciprofibrate functions to elevate ACO mRNA is not known. To help determine the mechanism of ciprofibrate action, in vitro transcription assays were performed. It was determined that ciprofibrate was responsible for a 3.5-fold stimulation of the rate of ACO transcription within 24 hr of ingestion. It was also observed that the transcription rate stimulation following a 2-week ciprofibrate treatment of Wistar rats was maintained following 4 weeks of ciprofibrate withdrawal. Re-introduction of the drug after the 4-week pause resulted in greater stimulation than was initially observed. The results demonstrate that the effect of ciprofibrate is rapid and persists at least twice as long as the initial treatment period. In Zucker rats, both lean and obese, ACO mRNA levels were examined following 2 weeks of ciprofibrate treatment (1 or 3 mg/kg body weight/day). The presence of increased blood levels of triglycerides did not increase ciprofibrate action on transcription, although basal levels of transcription of peroxisomal enzymes were higher in obese rats. The increase in the ACO mRNA level was greater than the transcription rate stimulation suggesting a post-transcriptional regulation.


Subject(s)
Clofibric Acid/analogs & derivatives , Liver/metabolism , Microbodies/enzymology , Obesity/genetics , Oxidoreductases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Acyl-CoA Oxidase , Animals , Clofibric Acid/pharmacology , Fibric Acids , Male , Obesity/blood , Oxidoreductases/genetics , RNA, Messenger/analysis , Rats , Rats, Wistar , Rats, Zucker , Triglycerides/blood
20.
Biochem Mol Biol Int ; 35(1): 65-77, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7735141

ABSTRACT

1. D-3-hydroxybutyrate dehydrogenase EC 1.1.1.30 (BDH) activity was measured in mitochondria of rats submitted to an intermittent feeding treatment with ciprofibrate or fenofibrate, i.e. fibrate analogues with hypolipemic activity and peroxisome proliferation properties. Our data shows an inhibition of rat liver mitochondrial BDH activity. This inhibitory effect is abolished when the treatment is stopped and reappears after a second treatment. 2. Incubation of hypolipemic agents (ciprofibrate, clofibrate, clobuzarit, fenofibrate or 2,4 dichlorophenoxyacetic acid) with submitochondrial linked BDH leads to an inhibition in a concentration dependent manner. 3. The protection by NAD(H) (coenzymes) and by methyl-malonate (a substrate analogue and competitive inhibitor) indicates that the inhibition occurs in the active site. On the other hand, there is a strong protection by phospholipid vesicles. This trapping effect may be attributed to lipophilic properties of hypolipemic agents. 4. Comparative effect of hypolipemic agents on mitochondrial BDH activity from rat liver and from Tetrahymena pyriformis indicates the same inhibition and same protection effects. This supports conservation of the enzymatic properties according to the evolution.


Subject(s)
Hydroxybutyrate Dehydrogenase/drug effects , Hypolipidemic Agents/pharmacology , Intracellular Membranes/enzymology , Microbodies/physiology , Mitochondria/enzymology , 2,4-Dichlorophenoxyacetic Acid/pharmacology , Animals , Binding, Competitive , Clofibrate/analogs & derivatives , Clofibrate/pharmacology , Clofibric Acid/analogs & derivatives , Clofibric Acid/pharmacology , Fenofibrate/pharmacology , Fibric Acids , Hydroxybutyrate Dehydrogenase/metabolism , In Vitro Techniques , Male , Mitochondria/ultrastructure , Mitochondria, Liver/enzymology , Mitochondria, Liver/ultrastructure , Phospholipids/pharmacology , Rats , Rats, Wistar , Tetrahymena pyriformis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL