Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biochimie ; 98: 86-101, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24287293

ABSTRACT

The peroxisomal 3-ketoacyl-CoA thiolase B (ThB) catalyzes the thiolytic cleavage of straight chain 3-ketoacyl-CoAs. Up to now, the ability of ThB to interfere with lipid metabolism was studied in mice fed a laboratory chow enriched or not with the synthetic agonist Wy14,643, a pharmacological activator of the nuclear hormone receptor PPARα. The aim of the present study was therefore to determine whether ThB could play a role in obesity and lipid metabolism when mice are chronically fed a synthetic High Fat Diet (HFD) or a Low Fat Diet (LFD) as a control diet. To investigate this possibility, wild-type (WT) mice and mice deficient for Thb (Thb(-/-)) were subjected to either a synthetic LFD or a HFD for 25 weeks, and their responses were compared. First, when fed a normal regulatory laboratory chow, Thb(-/-) mice displayed growth retardation as well as a severe reduction in the plasma level of Growth Hormone (GH) and Insulin Growth Factor-I (IGF-I), suggesting alterations in the GH/IGF-1 pathway. When fed the synthetic diets, the corrected energy intake to body mass was significantly higher in Thb(-/-) mice, yet those mice were protected from HFD-induced adiposity. Importantly, Thb(-/-) mice also suffered from hypoglycemia, exhibited reduction in liver glycogen stores and circulating insulin levels under the LFD and the HFD. Thb deficiency was also associated with higher levels of plasma HDL (High Density Lipoproteins) cholesterol and increased liver content of cholesterol under both the LFD and the HFD. As shown by the plasma lathosterol to cholesterol ratio, a surrogate marker for cholesterol biosynthesis, whole body cholesterol de novo synthesis was increased in Thb(-/-) mice. By comparing liver RNA from WT mice and Thb(-/-) mice using oligonucleotide microarray and RT-qPCR, a coordinated decrease in the expression of critical cholesterol synthesizing genes and an increased expression of genes involved in bile acid synthesis (Cyp7a1, Cyp17a1, Akr1d1) were observed in Thb(-/-) mice. In parallel, the elevation of the lathosterol to cholesterol ratio as well as the increased expression of cholesterol synthesizing genes were observed in the kidney of Thb(-/-) mice fed the LFD and the HFD. Overall, the data indicate that ThB is not fully interchangeable with the thiolase A isoform. The present study also reveals that modulating the expression of the peroxisomal ThB enzyme can largely reverberate not only throughout fatty acid metabolism but also cholesterol, bile acid and glucose metabolism.


Subject(s)
Acetyl-CoA C-Acyltransferase/deficiency , Animals , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Cholesterol, HDL/blood , Diet, High-Fat , Dietary Fats/administration & dosage , Glucose/metabolism , Growth Hormone/blood , Insulin-Like Growth Factor I/metabolism , Intestine, Small/metabolism , Liver Glycogen/metabolism , Mice
2.
Biochimie ; 93(5): 876-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21352884

ABSTRACT

Peroxisomal 3-ketoacyl-CoA thiolase B (Thb) catalyzes the final step in the peroxisomal ß-oxidation of straight-chain acyl-CoAs and is under the transcription control of the nuclear hormone receptor PPARα. PPARα binds to and is activated by the synthetic compound Wy14,643 (Wy). Here, we show that the magnitude of Wy-mediated induction of peroxisomal ß-oxidation of radiolabeled (1-(14)C) palmitate was significantly reduced in mice deficient for Thb. In contrast, mitochondrial ß-oxidation was unaltered in Thb(-/-) mice. Given that Wy-treatment induced Acox1 and MFP-1/-2 activity at a similar level in both genotypes, we concluded that the thiolase step alone was responsible for the reduced peroxisomal ß-oxidation of fatty acids. Electron microscopic analysis and cytochemical localization of catalase indicated that peroxisome proliferation in the liver after Wy-treatment was normal in Thb(-/-) mice. Intriguingly, micro-array analysis revealed that mRNA levels of genes encoding cholesterol biosynthesis enzymes were upregulated by Wy in Wild-Type (WT) mice but not in Thb(-/-) mice, which was confirmed at the protein level for the selected genes. The non-induction of genes encoding cholesterol biosynthesis enzymes by Wy in Thb(-/-) mice appeared to be unrelated to defective SREBP-2 or PPARα signaling. No difference was observed in the plasma lathosterol/cholesterol ratio (a marker for de novo cholesterol biosynthesis) between Wy-treated WT and Thb(-/-) mice, suggesting functional compensation. Overall, we conclude that ThA and SCPx/SCP2 thiolases cannot fully compensate for the absence of ThB. In addition, our data indicate that ThB is involved in the regulation of genes encoding cholesterol biosynthesis enzymes in the liver, suggesting that the peroxisome could be a promising candidate for the correction of cholesterol imbalance in dyslipidemia.


Subject(s)
Acetyl-CoA C-Acyltransferase/metabolism , Liver/enzymology , PPAR alpha/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Up-Regulation , Acetyl-CoA C-Acyltransferase/genetics , Animals , Cholesterol/genetics , Cholesterol/metabolism , Gene Deletion , Gene Expression Regulation , Hepatomegaly/genetics , Hepatomegaly/pathology , Humans , Lipid Metabolism/genetics , Liver/pathology , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Palmitates/metabolism , Peroxisome Proliferators/pharmacology , Peroxisomes/metabolism , Pyrimidines/pharmacology , Signal Transduction
3.
Biochimie ; 91(11-12): 1376-86, 2009.
Article in English | MEDLINE | ID: mdl-19772884

ABSTRACT

The peroxisomal 3-ketoacyl-CoA thiolase B (Thb) gene was previously identified as a direct target gene of PPARalpha, a nuclear hormone receptor activated by hypolipidemic fibrate drugs. To better understand the role of ThB in hepatic lipid metabolism in mice, Sv129 wild-type and Thb null mice were fed or not the selective PPARalpha agonist Wy14,643 (Wy). Here, it is shown that in contrast to some other mouse models deficient for peroxisomal enzymes, the hepatic PPARalpha signaling cascade in Thb null mice was normal under regular conditions. It is of interest that the hypotriglyceridemic action of Wy was reduced in Thb null mice underlining the conclusion that neither thiolase A nor SCPx/SCP2 thiolase can fully substitute for ThB in vivo. Moreover, a significant increased in the expression of lipogenic genes such as Stearoyl CoA Desaturase-1 (SCD1) was observed in Thb null mice fed Wy. Elevation of Scd1 mRNA and protein levels led to higher SCD1 activity, through a molecular mechanism that is probably SREBP1 independent. In agreement with higher SCD1, enrichment of liver mono-unsaturated fatty acids of the n-7 and n-9 series was found in Thb null mice fed Wy. Overall, we show that the reduced peroxisomal beta-oxidation of fat observed in Thb null mice fed Wy is associated with enhanced hepatic lipogenesis, through the combined elevation of microsomal SCD1 protein and activity. Ultimately, not only the amount but also the quality of the hepatic fatty acid pool is modulated upon the deletion of Thb.


Subject(s)
Lipid Metabolism/drug effects , PPAR alpha/antagonists & inhibitors , Peroxisomes/drug effects , Pyrimidines/pharmacology , Stearoyl-CoA Desaturase/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Fatty Acids/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lipid Metabolism/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Microsomes, Liver/pathology , Peroxisomes/metabolism , RNA, Messenger/drug effects
4.
Cancer Lett ; 222(2): 217-26, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15863271

ABSTRACT

Humans appear to be refractory to some effects of peroxisome proliferators including alterations in cell proliferation, whereas rodents are susceptible. In this study, differences between the human and rat response to peroxisome proliferators were evaluated using rat and human tumour liver cell lines. Rat 7777 cells were more responsive than human HepG2 cells to ciprofibrate as they exhibited a higher decrease in cell number than HepG2, and underwent apoptosis. Results from these studies reveal a surprising response in tumour cell lines as the typical in vivo response of increased cell proliferation and reduced apoptosis was not observed in rat tumour cell lines at concentrations greater than those used to elicit the former response.


Subject(s)
Cell Proliferation/drug effects , Clofibric Acid/analogs & derivatives , Clofibric Acid/pharmacology , Hypolipidemic Agents/pharmacology , Liver/cytology , Animals , Apoptosis , Cell Line , Dose-Response Relationship, Drug , Fibric Acids , Rats
5.
Biochimie ; 86(11): 763-70, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15589684

ABSTRACT

Brown (BAT) and white (WAT) adipose tissues play a key role in the body energy balance orchestrated by the central nervous system. Hibernators have developed a seasonal obesity to respond to inhospitable environment. Jerboa is one of the deep hibernator originated from sub-desert highlands. Thus, this animal represents an excellent model to study cold adaptation mechanism. We report that the adipogenic factor PPARgamma exhibits a differential expression between BAT and WAT at mRNA level. A specific induction was only seen in WAT of pre-hibernating jerboa. Interestingly, PPAR beta/delta is specifically induced in BAT and brain of pre-hibernating jerboa, highlighting for the first time the possible key role of this ubiquitous isoform in the cold adaptation of this true hibernator. Inductions of PPARgamma(2) in WAT and PPAR beta/delta in BAT are blunted by a hypolipemic drug, the ciprofibrate. These changes may be correlated with hibernation arrest and death of treated jerboa. Mitochondrial acyl-CoA dehydrogenase and peroxisomal acyl-CoA oxidase activities in brown and white adipose tissues are decreased up to 85% during cold acclimatization (without food privation). These enzyme activities are subject to a strong induction in BAT and in WAT (3.4-7.5 fold) during the hibernation period. The BAT thermogenesis marker is also largely induced (approximately 4 fold of UCP1 mRNA level) during pre-hibernation period. Unexpectedly, treatment with ciprofibrate deeply affects lipolysis in BAT by increasing acyl-CoA dehydrogenase activity (3.4 fold) and acyl-CoA oxidase at both activity and mRNA levels (2.8 and 3.8 fold, respectively) and enhances strongly UCP1 mRNA level (9.5 fold) during pre-hibernation.


Subject(s)
Acclimatization/physiology , Adipose Tissue/metabolism , Clofibric Acid/analogs & derivatives , Gene Expression Regulation/physiology , Hibernation/physiology , Lipid Metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Acyl-CoA Dehydrogenase/metabolism , Acyl-CoA Oxidase , Animals , Carrier Proteins/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Clofibric Acid/pharmacology , Cold Temperature , Energy Metabolism , Fibric Acids , Gene Expression Regulation/genetics , Hibernation/drug effects , Ion Channels , Lipids/genetics , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins , Oxidoreductases/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Phospholipases/drug effects , Phospholipases/genetics , Phospholipases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rodentia , Uncoupling Protein 1
6.
Biochimie ; 86(11): 849-56, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15589695

ABSTRACT

The peroxisomal beta-oxidation system consists of four steps catalysed by three enzymes: acyl-CoA oxidase, 3-hydroxyacyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase (multifunctional enzyme) and thiolase. In humans, thiolase activity is encoded by one gene, whereas in rodents, three enzymes encoded by three distinct genes (i.e. thiolase A, thiolase B and SCP2/thiolase) catalyse the thiolase activity. So far, acyl-CoA oxidase- and multifunctional enzyme-deficient patients have been identified and knock-out mice for these genes have been produced. Conversely, no isolated thiolase-deficient patient has been found, and no thiolase (A or B)-deficient mice have been generated. Hence, to better understand the cause of isolated human thiolase deficiency, we disrupted the catalytic site of the mouse thiolase B by homologous recombination in order to analyse the phenotype of these thiolase B-deficient mice. Mice, made homozygous for the mutation, lack expression of thiolase B mRNA and are viable, fertile and healthy at birth. They exhibit no detectable phenotype defects and no compensation, rather a slight decrease in other peroxisomal thiolase (thiolase A and SCPx) mRNAs, was found.


Subject(s)
Acetyl-CoA C-Acyltransferase/genetics , Disease Models, Animal , Lipid Metabolism , Peroxisomal Disorders/metabolism , Peroxisomes/genetics , Peroxisomes/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Embryo, Mammalian/cytology , Gene Expression Regulation, Enzymologic , Humans , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Peroxisomal Disorders/genetics , Peroxisomes/enzymology , Phenotype , RNA, Messenger/genetics , Stem Cells/cytology , Structure-Activity Relationship
7.
BMC Biochem ; 5: 3, 2004 Mar 25.
Article in English | MEDLINE | ID: mdl-15043762

ABSTRACT

BACKGROUND: In rats, two peroxisomal 3-ketoacyl-CoA thiolase genes (A and B) have been cloned, whereas only one thiolase gene is found in humans. The aim of this study was thus to clone the different mouse thiolase genes in order to study both their tissue expression and their associated enzymatic activity. RESULTS: In this study, we cloned and characterized two mouse peroxisomal 3-ketoacyl-CoA thiolase genes (termed thiolase A and B). Both thiolase A and B genes contain 12 exons and 11 introns. Using RNA extracted from mouse liver, we cloned the two corresponding cDNAs. Thiolase A and B cDNAs possess an open reading frame of 1272 nucleotides encoding a protein of 424 amino acids. In the coding sequence, the two thiolase genes exhibited approximately equal to 97% nucleotide sequence identity and approximately equal to 96% identity at the amino acid level. The tissue-specific expression of the two peroxisomal 3-ketoacyl-CoA thiolase genes was studied in mice. Thiolase A mRNA was mainly expressed in liver and intestine, while thiolase B mRNA essentially exhibited hepatic expression and weaker levels in kidney, intestine and white adipose tissue. Thiolase A and B expressions in the other tissues such as brain or muscle were very low though these tissues were chiefly involved in peroxisomal disorders. At the enzymatic level, thiolase activity was detected in liver, kidney, intestine and white adipose tissue but no significant difference was observed between these four tissues. Moreover, thiolase A and B genes were differently induced in liver of mice treated with fenofibrate. CONCLUSION: Two mouse thiolase genes and cDNAs were cloned. Their corresponding transcripts are mostly expressed in the liver of mice and are differently induced by fenofibrate.


Subject(s)
Acetyl-CoA C-Acyltransferase/genetics , Peroxisomes/enzymology , Acetyl-CoA C-Acyltransferase/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Fenofibrate/pharmacology , Gene Components , Gene Expression/drug effects , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Tissue Distribution
8.
Lipids Health Dis ; 3: 4, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-15046640

ABSTRACT

BACKGROUND: Beta-oxidation of long and very long chain fatty acyl-CoA derivatives occurs in peroxisomes, which are ubiquitous subcellular organelles of eukaryotic cells. This pathway releases acetyl-CoA as precursor for several key molecules such as cholesterol. Numerous enzymes participating to cholesterol and fatty acids biosynthesis pathways are co-localized in peroxisomes and some of their encoding genes are known as targets of the NFY transcriptional regulator. However, until now no interaction between NFY transcription factor and genes encoding peroxisomal beta-oxidation has been reported. RESULTS: This work studied the interactions between NFY factor with the rat gene promoters of two enzymes of the fatty acid beta-oxidation, MFP-1 (multifunctional protein type 1) and ThB (thiolase B) and their involvement in the cholesterol dependent-gene regulation. Binding of this nuclear factor to the ATTGG motif of the MFP-1 and of the ThB promoters was demonstrated by EMSA (Electrophoretic Mobility Shift Assay) and super shift assay. In contrast, in spite of the presence of putative Sp1 binding sites in these promoters, competitive EMSA did not reveal any binding. The promoter-dependent luciferase gene expression was downregulated by cholesterol in MFP-1 and ThB promoters harbouring constructs. CONCLUSIONS: This work describes for the first time a NFY interaction with promoter sequences of the peroxisomal beta-oxidation encoding genes. It suggests that cholesterol would negatively regulate the expression of genes involved in beta-oxidation, which generates the initial precursor for its own biosynthesis, via at least the NFY transcription factor.

9.
Biochem Biophys Res Commun ; 311(1): 149-55, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-14575706

ABSTRACT

Expression of the rat peroxisomal 3-ketoacyl-CoA thiolase gene B is induced by peroxisome proliferators. Although a sequence element like a peroxisome proliferator-activated receptor (PPAR)-binding site is located in the promoter region of this gene, we previously found that this element is competent for the activation by hepatocyte nuclear factor-4, but not functional with PPARalpha. We describe here a new peroxisome proliferator-response element located in the intron 3 (+1422/+1434) that binds in vitro the PPARalpha/retinoid X receptor alpha heterodimer and confers the induction by PPARalpha in transfection assays. We propose a model of regulation of the rat thiolase B gene involving those elements in the promoter and intron 3.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , DNA-Binding Proteins , Gene Expression Regulation, Enzymologic/physiology , Liver/enzymology , Peroxisomes/enzymology , Peroxisomes/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Hepatocyte Nuclear Factor 4 , Introns , Peroxisome Proliferators/metabolism , Phosphoproteins/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Structure-Activity Relationship , Transcription Factors/genetics
10.
Biochimie ; 85(7): 707-14, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14505827

ABSTRACT

Jerboa (Jaculus orientalis) is a deep hibernator originating from sub-desert highlands and represents an excellent model to help to understand the incidence of seasonal variations of food intake and of body as well as environmental temperatures on lipid metabolism. In jerboa, hibernation processes are characterized by changes in the size of mitochondria, the number of peroxisomes in liver and in the expression of enzymes linked to fatty acid metabolism. In liver and kidney, cold acclimatization shows an opposite effect on the activities of the mitochondrial acyl-CoA dehydrogenase (-50%) and the peroxisomal acyl-CoA oxidase (AOX) (+50%), while in brown and white adipose tissues, both activities are decreased down to 85%. These enzymes activities are subject to a strong induction in brown and in white adipose tissue (3.4- to 7.5-fold, respectively) during the hibernation period which is characterized by a low body temperature (around 10 degrees C) and by starvation. Expression level of AOX mRNA and protein are increased during both pre-hibernation and hibernation periods. Unexpectedly, treatment with ciprofibrate, a hypolipemic agent, deeply affects lipolysis in brown adipose tissue by increasing acyl-CoA dehydrogenase activity (3.4-fold), both AOX activity and mRNA levels (2.8- and 3.8-fold, respectively) during pre-hibernation. Therefore, during pre-hibernation acclimatization, there is a negative regulation of fatty acid degradation allowing to accumulate a lipid stock which is later degraded during the hibernation period (starvation) due to a positive regulation of enzymes providing the required energy for animal survival.


Subject(s)
Acclimatization/physiology , Cold Temperature , Fatty Acids/metabolism , Hibernation , Peroxisomes/metabolism , Rodentia/physiology , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Body Temperature Regulation/physiology , Enzyme Activation , Liver/cytology , Liver/enzymology , Liver/ultrastructure , Male , Mitochondria/enzymology , Peroxisomes/ultrastructure , RNA, Messenger/genetics , Rodentia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...