Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Biol Chem ; 299(2): 102848, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36587768

RESUMO

In eukaryotes, carnitine is best known for its ability to shuttle esterified fatty acids across mitochondrial membranes for ß-oxidation. It also returns to the cytoplasm, in the form of acetyl-L-carnitine (LAC), some of the resulting acetyl groups for posttranslational protein modification and lipid biosynthesis. While dietary LAC supplementation has been clinically investigated, its effects on cellular metabolism are not well understood. To explain how exogenous LAC influences mammalian cell metabolism, we synthesized isotope-labeled forms of LAC and its analogs. In cultures of glucose-limited U87MG glioma cells, exogenous LAC contributed more robustly to intracellular acetyl-CoA pools than did ß-hydroxybutyrate, the predominant circulating ketone body in mammals. The fact that most LAC-derived acetyl-CoA is cytosolic is evident from strong labeling of fatty acids in U87MG cells by exogenous 13C2-acetyl-L-carnitine. We found that the addition of d3-acetyl-L-carnitine increases the supply of acetyl-CoA for cytosolic posttranslational modifications due to its strong kinetic isotope effect on acetyl-CoA carboxylase, the first committed step in fatty acid biosynthesis. Surprisingly, whereas cytosolic carnitine acetyltransferase is believed to catalyze acetyl group transfer from LAC to coenzyme A, CRAT-/- U87MG cells were unimpaired in their ability to assimilate exogenous LAC into acetyl-CoA. We identified carnitine octanoyltransferase as the key enzyme in this process, implicating a role for peroxisomes in efficient LAC utilization. Our work has opened the door to further biochemical investigations of a new pathway for supplying acetyl-CoA to certain glucose-starved cells.


Assuntos
Acetilcoenzima A , Acetilcarnitina , Carnitina Aciltransferases , Carnitina , Acetilcoenzima A/metabolismo , Acetilcarnitina/farmacologia , Carnitina/metabolismo , Carnitina Aciltransferases/metabolismo , Carnitina O-Acetiltransferase/genética , Carnitina O-Acetiltransferase/metabolismo , Ácidos Graxos/metabolismo , Glucose/metabolismo , Oxirredução , Humanos , Linhagem Celular Tumoral
2.
J Invest Dermatol ; 143(2): 305-316.e5, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36058299

RESUMO

Circulating tumor cells are the key link between a primary tumor and distant metastases, but once in the bloodstream, loss of adhesion induces cell death. To identify the mechanisms relevant for melanoma circulating tumor cell survival, we performed RNA sequencing and discovered that detached melanoma cells and isolated melanoma circulating tumor cells rewire lipid metabolism by upregulating fatty acid (FA) transport and FA beta-oxidation‒related genes. In patients with melanoma, high expression of FA transporters and FA beta-oxidation enzymes significantly correlates with reduced progression-free and overall survival. Among the highest expressed regulators in melanoma circulating tumor cells were the carnitine transferases carnitine O-octanoyltransferase and carnitine acetyltransferase, which control the shuttle of peroxisome-derived medium-chain FAs toward mitochondria to fuel mitochondrial FA beta-oxidation. Knockdown of carnitine O-octanoyltransferase or carnitine acetyltransferase and short-term treatment with peroxisomal or mitochondrial FA beta-oxidation inhibitors thioridazine or ranolazine suppressed melanoma metastasis in mice. Carnitine O-octanoyltransferase and carnitine acetyltransferase depletion could be rescued by medium-chain FA supplementation, indicating that the peroxisomal supply of FAs is crucial for the survival of nonadherent melanoma cells. Our study identifies targeting the FA-based cross-talk between peroxisomes and mitochondria as a potential therapeutic opportunity to challenge melanoma progression. Moreover, the discovery of the antimetastatic activity of the Food and Drug Administration‒approved drug ranolazine carries translational potential.


Assuntos
Melanoma , Células Neoplásicas Circulantes , Camundongos , Animais , Carnitina O-Acetiltransferase/genética , Carnitina O-Acetiltransferase/metabolismo , Carnitina Aciltransferases/genética , Carnitina Aciltransferases/metabolismo , Ranolazina , Oxirredução , Ácidos Graxos/metabolismo , Melanoma/tratamento farmacológico , Carnitina/metabolismo
3.
Appl Biochem Biotechnol ; 193(5): 1469-1481, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33484445

RESUMO

Inhibition of lipid accumulation is the key step to prevent nonalcoholic fatty liver (NAFL) progressing to nonalcoholic steatohepatitis. We aimed to study the effect of low-molecular-weight citrus pectin (LCP) against lipid accumulation and the underlying mechanism. Oleic acid (OA)-induced lipid deposition in HepG2 cells was applied to mimic in vitro model of lipid accumulation. Oil Red O (ORO) stain result showed lipid accumulation was significantly reduced, and levels of adipose triglyceride lipase (ATGL) and carnitine palmitoyltransferase-1 (CPT-1), involved in triacylglycerol catabolism and fatty acid ß-oxidation, detected by RT-qPCR were increased after OA-stimulated HepG2 cells treated with LCP. RNA sequencing analysis identified 740 differentially expressed genes (DEGs) in OA-stimulated HepG2 cells treated with the LCP group (OA+LCP group), and bioinformatics analysis indicated that some DEGs were enriched in lipid metabolism-related processes and pathways. The expression of the top 8 known DEGs in the OA+LCP group was then verified by RT-qPCR, which showed that fold change (abs) of METTL7B was the highest among the 8 candidates. In addition, overexpression of METTL7B in HepG2 cells significantly inhibited the lipid accumulation and enhanced levels of ATGL and CPT-1. In conclusion, LCP inhibited lipid accumulation through the upregulation of METTL7B, and further enhancement of ATGL and CPT-1 levels. LCP is expected to develop as a promising agent to ameliorate fat accumulation in NAFL.


Assuntos
Proteínas de Transporte/metabolismo , Pectinas/farmacologia , Carnitina Aciltransferases/metabolismo , Proteínas de Transporte/genética , Biologia Computacional , Células Hep G2 , Humanos , Lipase/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos
4.
Ann Nutr Metab ; 68 Suppl 3: 15-20, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27931032

RESUMO

Alterations in muscle fatty acid metabolism have been implicated in mediating the severity of insulin resistance. In the insulin resistant heart fatty acids are favored as an energy source over glucose, which is thus associated with increased fatty acid oxidation, and an overall decrease in glycolysis and glucose oxidation. In addition, excessive uptake and beta-oxidation of fatty acids in obesity and diabetes can compromise cardiac function. In animal studies, mice fed a high fat diet (HFD) show cardiac insulin resistance in which the accumulation of intra-myocardial diacylglycerol has been implicated, likely involving parallel signaling pathways. A HFD also results in accumulation of fatty acid oxidation byproducts in muscle, further contributing to insulin resistance. Carnitine acetyltransferase (CrAT) has an essential role in the cardiomyocyte because of its need for large amounts of carnitine. In the cardiomyocyte, carnitine switches energy substrate preference in the heart from fatty acid oxidation to glucose oxidation. This carnitine-induced switch in fatty acid oxidation to glucose oxidation is due to the presence of cytosolic CrAT and reverse CrAT activity. Accordingly, inhibition of fatty acid oxidation, or stimulation of CrAT, may be a novel approach to treatment of insulin resistance.


Assuntos
Carnitina Aciltransferases/metabolismo , Carnitina/metabolismo , Diabetes Mellitus/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Resistência à Insulina , Miocárdio/metabolismo , Obesidade/metabolismo , Animais , Carnitina/deficiência , Carnitina/uso terapêutico , Carnitina Aciltransferases/química , Deficiências Nutricionais/dietoterapia , Deficiências Nutricionais/metabolismo , Deficiências Nutricionais/fisiopatologia , Deficiências Nutricionais/prevenção & controle , Diabetes Mellitus/dietoterapia , Diabetes Mellitus/etiologia , Diabetes Mellitus/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Diglicerídeos/metabolismo , Coração/fisiopatologia , Humanos , Músculos/enzimologia , Músculos/metabolismo , Miocárdio/enzimologia , Obesidade/dietoterapia , Obesidade/etiologia , Obesidade/fisiopatologia , Oxirredução , Disfunção Ventricular/etiologia , Disfunção Ventricular/prevenção & controle
5.
Br J Nutr ; 112(6): 1034-40, 2014 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-25201308

RESUMO

Obesity has become a public health concern due to its positive association with the incidence of many diseases, and coffee components including chlorogenic acid (CGA) and caffeine have been demonstrated to play roles in the suppression of fat accumulation. To investigate the mechanism by which CGA and caffeine regulate lipid metabolism, in the present study, forty mice were randomly assigned to four groups and fed diets containing no CGA or caffeine, CGA, caffeine, or CGA+caffeine for 24 weeks. Body weight, intraperitoneal adipose tissue (IPAT) weight, and serum biochemical parameters were measured, and the activities and mRNA and protein expression of lipid metabolism-related enzymes were analysed. There was a decrease in the body weight and IPAT weight of mice fed the CGA+caffeine diet. There was a significant decrease in the serum and hepatic concentrations of total cholesterol, TAG and leptin of mice fed the CGA+caffeine diet. The activities of carnitine acyltransferase (CAT) and acyl-CoA oxidase (ACO) were increased in mice fed the caffeine and CGA+caffeine diets, while the activity of fatty acid synthase (FAS) was suppressed in those fed the CGA+caffeine diet. The mRNA expression levels of AMP-activated protein kinase (AMPK), CAT and ACO were considerably up-regulated in mice fed the CGA+caffeine diet, while those of PPARγ2 were down-regulated. The protein expression levels of AMPK were increased and those of FAS were decreased in mice fed the CGA+caffeine diet. These results indicate that CGA+caffeine suppresses fat accumulation and body weight gain by regulating the activities and mRNA and protein expression levels of hepatic lipid metabolism-related enzymes and that these effects are stronger than those exerted by CGA and caffeine individually.


Assuntos
Cafeína/uso terapêutico , Ácido Clorogênico/uso terapêutico , Suplementos Nutricionais , Fígado Gorduroso/prevenção & controle , Regulação Enzimológica da Expressão Gênica , Fígado/metabolismo , Acil-CoA Oxidase/química , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Adiposidade , Animais , Carnitina Aciltransferases/química , Carnitina Aciltransferases/genética , Carnitina Aciltransferases/metabolismo , Indução Enzimática , Repressão Enzimática , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Hiperlipidemias/prevenção & controle , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Leptina/sangue , Leptina/metabolismo , Metabolismo dos Lipídeos , Fígado/enzimologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos ICR , Tamanho do Órgão , Distribuição Aleatória
6.
Biochim Biophys Acta ; 1821(10): 1341-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22819991

RESUMO

The carnitine/acylcarnitine translocase (CACT), an integral protein of the mitochondrial inner membrane, belongs to the carnitine-dependent system of fatty acid transport into mitochondria, where beta-oxidation occurs. CACT exchanges cytosolic acylcarnitine or free carnitine for carnitine in the mitochondrial matrix. The object of this study was to investigate in rat liver the effect, if any, of diets enriched with saturated fatty acids (beef tallow, BT, the control), n-3 polyunsaturated fatty acids (PUFA) (fish oil, FO), n-6 PUFA (safflower oil, SO), and mono-unsaturated fatty acids (MUFA) (olive oil, OO) on the activity and expression of CACT. Translocase exchange rates increased, in parallel with CACT mRNA abundance, upon FO-feeding, whereas OO-dietary treatment induced a decrease in both CACT activity and expression. No changes were observed upon SO-feeding. Nuclear run-on assay revealed that FO-treatment increased the transcriptional rate of CACT mRNA. On the other hand, only in the nuclei of hepatocytes from OO-fed rats splicing of the last intron of CACT pre-mRNA and the rate of formation of the 3'-end were affected. Overall, these findings suggest that compared to the BT-enriched diet, the SO-enriched diet did not influence CACT activity and expression, whereas FO- and OO-feeding alters CACT activity in an opposite fashion, i.e. modulating its expression at transcriptional and post-transcriptional levels, respectively.


Assuntos
Carnitina Aciltransferases/metabolismo , Gorduras na Dieta/farmacologia , Mitocôndrias Hepáticas/enzimologia , Animais , Carnitina Aciltransferases/análise , Carnitina Aciltransferases/genética , Masculino , Lipídeos de Membrana/análise , Estabilidade de RNA , RNA Mensageiro/análise , Ratos , Ratos Wistar
7.
Curr Med Chem ; 17(32): 3842-54, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20858216

RESUMO

Riboflavin, commonly known as vitamin B2, is the precursor of flavin cofactors. It is present in our typical diet, and inside the cells it is metabolized to FMN and FAD. As a result of their rather unique and flexible chemical properties these flavins are among the most important redox cofactors present in a large series of different enzymes. A problem in riboflavin metabolism or a low intake of this vitamin will have consequences on the level of FAD and FMN in the cell, resulting in disorders associated with riboflavin deficiency. In a few number of cases, riboflavin deficiency is associated with impaired oxidative folding, cell damage and impaired heme biosynthesis. More relevant are several studies referring reduced activity of enzymes such as dehydrogenases involved in oxidative reactions, respiratory complexes and enzymes from the fatty acid ß-oxidation pathway. The role of this vitamin in mitochondrial metabolism, and in particular in fatty acid oxidation, will be discussed in this review. The basic aspects concerning riboflavin and flavin metabolism and deficiency will be addressed, as well as an overview of the role of the different flavoenzymes and flavin chemistry in fatty acid ß-oxidation, merging clinical, cellular and biochemical perspectives. A number of recent studies shedding new light on the cellular processes and biological effects of riboflavin supplementation in metabolic disease will also be overviewed. Overall, a deeper understanding of these emerging roles of riboflavin intake is essential to design better therapies.


Assuntos
Mitocôndrias/metabolismo , Riboflavina/fisiologia , Carnitina Aciltransferases/metabolismo , Humanos , Deficiência Múltipla de Acil Coenzima A Desidrogenase/metabolismo , Oxirredução , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Riboflavina/metabolismo
8.
Mol Membr Biol ; 25(2): 152-63, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18307102

RESUMO

The mitochondrial carnitine/acylcarnitine carrier (CAC) is characterized by the presence of a distinct motif, RXXPANAAXF, within its sixth transmembrane alpha-helix. In this study, we analysed the role of the amino acids of this motif in the structure-function relationships of the human CAC by using two complementary approaches. First, we performed functional analysis in the model fungus Aspergillus nidulans of selected mutations with structural and functional relevance. Second, similar mutant human CACs were biochemically characterized after their reconstitution into liposomes. Both analyses have provided relevant information on the importance and role of the CAC motif residues in the activity and metabolic function of CAC. Only the two adjacent alanines, Ala281 and Ala282 in the human CAC, have been found not to be crucial for transport activity and in vivo function. Results obtained from amino acid substitutions of residues Arg275, Asn280 and Phe284 of human CAC together with structural analysis using molecular modelling of the carrier suggest that R275, N280 and F284 are involved in substrate binding during acylcarnitine/carnitine translocation. Furthermore, functional analysis of mutations of residues Pro278 and Ala279 in A. nidulans, together with kinetic data in reconstituted liposomes, suggest a predominant structural role for these amino acids.


Assuntos
Aminoácidos/metabolismo , Carnitina Aciltransferases/química , Carnitina Aciltransferases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Aspergillus nidulans/enzimologia , Aspergillus nidulans/crescimento & desenvolvimento , Transporte Biológico , Calorimetria , Carnitina/metabolismo , Humanos , Cinética , Lipossomos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Fatores de Tempo , Transformação Genética , Trítio/metabolismo
9.
Cell Mol Life Sci ; 65(6): 982-90, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18264800

RESUMO

Peroxisomes metabolize a variety of lipids, acting as a chain-shortening system that produces acyl-CoAs of varying chain lengths, including acetyl-CoA and propionyl-CoA. It is, however, still largely unknown how beta-oxidation products exit peroxisomes and where they are further metabolized. Peroxisomes contain carnitine acetyltransferase (CRAT) and carnitine octanoyltransferase (CROT) that produce carnitine esters for transport out of peroxisomes, together with recently characterized acyl-CoA thioesterases (ACOTs) that produce free fatty acids. Here we have performed tissue expression profiling of the short- and medium-chain carnitine acyltransferases Crat, Crot and the short- and medium-chain thioesterases (Acot12) and (Acot5), and show that they are largely expressed in different tissues, suggesting that they do not compete for the same substrates but rather provide complementary systems for transport of metabolites across the peroxisomal membrane. These data also explain earlier observed tissue differences in peroxisomal production of acetyl-CoA/acetyl-carnitine/acetate and underscores the differences in peroxisome function in various organs.


Assuntos
Carnitina Aciltransferases/metabolismo , Peroxissomos/metabolismo , Tioléster Hidrolases/metabolismo , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Carnitina Aciltransferases/química , Carnitina Aciltransferases/genética , Catalase/metabolismo , Regulação Enzimológica da Expressão Gênica , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Mitocôndrias/enzimologia , Dados de Sequência Molecular , Especificidade de Órgãos , Oxirredução , Alinhamento de Sequência
10.
Mol Aspects Med ; 25(5-6): 521-32, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15363639

RESUMO

The carnitine-acylcarnitine translocase (CACT) is one of the components of the carnitine cycle. The carnitine cycle is necessary to shuttle long-chain fatty acids from the cytosol into the intramitochondrial space where mitochondrial beta-oxidation of fatty acids takes place. The oxidation of fatty acids yields acetyl-coenzyme A (CoA) units, which may either be degraded to CO(2) and H(2)O in the citric acid cycle to produce ATP or converted into ketone bodies which occurs in liver and kidneys. Metabolic consequences of a defective CACT are hypoketotic hypoglycaemia under fasting conditions, hyperammonemia, elevated creatine kinase and transaminases, dicarboxylic aciduria, very low free carnitine and an abnormal acylcarnitine profile with marked elevation of the long-chain acylcarnitines. Clinical signs and symptoms in CACT deficient patients, are a combination of energy depletion and endogenous toxicity. The predominantly affected organs are brain, heart and skeletal muscle, and liver, leading to neurological abnormalities, cardiomyopathy and arrythmias, skeletal muscle damage and liver dysfunction. Most patients become symptomatic in the neonatal period with a rapidly progressive deterioration and a high mortality rate. However, presentations at a later age with a milder phenotype have also been reported. The therapeutic approach is the same as in other long-chain fatty acid disorders and includes intravenous glucose (+/- insulin) administration to maximally inhibit lipolysis and subsequent fatty acid oxidation during the acute deterioration, along with other measures such as ammonia detoxification, depending on the clinical features. Long-term strategy consists of avoidance of fasting with frequent meals and a special diet with restriction of long-chain fatty acids. Due to the extremely low free carnitine concentrations, carnitine supplementation is often needed. Acylcarnitine profiling in plasma is the assay of choice for the diagnosis at a metabolite level. However, since the acylcarnitine profile observed in CACT-deficient patients is identical to that in CPT2-deficient patients, definitive identification of CACT-deficiency in a certain patient requires determination of the activity of CACT. Subsequently, mutational analysis of the CACT gene can be performed. So far, 9 different mutations have been identified in the CACT gene.


Assuntos
Carnitina Aciltransferases/deficiência , Carnitina Aciltransferases/metabolismo , Animais , Carnitina/metabolismo , Carnitina Aciltransferases/genética , Homeostase , Humanos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Mutação/genética
11.
Mol Aspects Med ; 25(5-6): 475-93, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15363637

RESUMO

Cells contain limited and sequestered pools of Coenzyme A (CoA) that are essential for activating carboxylate metabolites. Some acyl-CoA esters have high metabolic and signalling impact, so control of CoA ester concentrations is important. This and transfer of the activated acyl moieties between cell compartments without wasting energy on futile cycles of hydrolysis and resynthesis is achieved through the carnitine system. The location, properties of and deficiencies in the carnitine acyltransferases are described in relation to their influence on the CoA pools in the cell and, hence, on metabolism. The protection of free CoA pools in disease states is achieved by excretion of acyl-carnitine so that carnitine supplementation is required where unwanted acyl groups build up, such as in some inherited disorders of fatty acid oxidation. Acetyl-carnitine improves cognition in the brain and propionyl-carnitine improves cardiac performance in heart disease and diabetes. The therapeutic effects of carnitine and its esters are discussed in relation to the integrative influence of the carnitine system across CoA pools. Recent evidence for sequestered pools of activated acetate for synthesis of malonyl-CoA, for the synthesis of polyunsaturated fatty acids and for the inhibition of carnitine palmitoyltransferase 1 to regulate fatty acid oxidation is reviewed.


Assuntos
Carnitina Aciltransferases/metabolismo , Coenzima A/metabolismo , Doença , Saúde , Animais , Carnitina/química , Carnitina/metabolismo , Carnitina/uso terapêutico , Humanos , Mitocôndrias/metabolismo
12.
J Biol Chem ; 278(40): 38796-802, 2003 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-12882971

RESUMO

The carnitine-dependent transport of long-chain fatty acids is essential for fatty acid catabolism. In this system, the fatty acid moiety of acyl-CoA is transferred enzymatically to carnitine, and the resultant product, acylcarnitine, is imported into the mitochondrial matrix through a transporter named carnitine-acylcarnitine translocase (CACT). Here we report a novel mammalian protein homologous to CACT. The protein, designated as CACL (CACT-like), is localized to the mitochondria and has palmitoylcarnitine transporting activity. The tissue distribution of CACL is similar to that of CACT; both are expressed at a higher level in tissues using fatty acids as fuels, except in the brain, where only CACL is expressed. In addition, CACL is induced by partial hepatectomy or fasting. Thus, CACL may play an important role cooperatively with its homologue CACT in a stress-induced change of lipid metabolism, and may be specialized for the metabolism of a distinct class of fatty acids involved in brain function.


Assuntos
Carnitina Aciltransferases/química , Carnitina Aciltransferases/metabolismo , Carnitina Aciltransferases/farmacologia , Carnitina/análogos & derivados , Fígado/fisiologia , Mitocôndrias/enzimologia , Proteínas Mitocondriais/farmacologia , Células 3T3 , Sequência de Aminoácidos , Animais , Transporte Biológico , Northern Blotting , Western Blotting , Encéfalo/fisiologia , Carnitina/química , DNA Complementar/metabolismo , Escherichia coli/metabolismo , Ácidos Graxos/metabolismo , Privação de Alimentos , Teste de Complementação Genética , Humanos , Imuno-Histoquímica , Metabolismo dos Lipídeos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Proteínas Mitocondriais/química , Dados de Sequência Molecular , Palmitoilcarnitina/química , Plasmídeos/metabolismo , Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Distribuição Tecidual
13.
J Nutr Sci Vitaminol (Tokyo) ; 49(5): 320-6, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14703306

RESUMO

In this study, we examined the effects of sesamin and vegetable oil on the concentrations of polyunsaturated fatty acid (PUFA) and lipids (triacylglycerol, free cholesterol, and phospholipid), and beta-oxidation enzyme activities in the rat liver. Rats were fed a diet containing 5% (low-fat diet) or 20% (high-fat diet) salad oil (rapeseed oil: soybean oil, 7:3) with or without sesamin (0.5% w/w) for 4 wk. As a result, the concentrations of linoleic acid (LA, n-6), alpha-linolenic acid (ALA, n-3), and total PUFA in the liver increased significantly as the result of the high-fat diet. In the high-fat diet groups, sesamin administration decreased the concentrations of LA, ALA, and total PUFA to almost the same level as the low-fat diet group, while it increased the concentrations of dihomo-gamma-linolenic acid (DGLA, n-6) and arachidonic acid (AA, n-6). The activities of carnitine acyltransferase and acyl-CoA dehydrogenase in liver mitochondria were enhanced by the intake of the high-fat diet, and were further enhanced by the administration of sesamin. Peroxisomal acyl-CoA oxidase activity was also enhanced by sesamin, while it was not affected by the dietary fat level. These results suggest that sesamin suppressed the increase of hepatic PUFA concentration caused by feeding the high-fat diet through enhancing the enzyme activities of fatty acid beta-oxidation and PUFA metabolism from LA and ALA.


Assuntos
Gorduras na Dieta/administração & dosagem , Dioxóis/administração & dosagem , Ácidos Graxos Insaturados/metabolismo , Lignanas/administração & dosagem , Ácido 8,11,14-Eicosatrienoico/análise , Acil-CoA Desidrogenase/metabolismo , Animais , Ácido Araquidônico/análise , Carnitina Aciltransferases/metabolismo , Ácidos Graxos Monoinsaturados , Ácidos Graxos Insaturados/análise , Ácido Linoleico/análise , Fígado/química , Fígado/efeitos dos fármacos , Masculino , Mitocôndrias Hepáticas/enzimologia , Óleos de Plantas/administração & dosagem , Óleo de Brassica napus , Ratos , Ratos Wistar , Óleo de Soja/administração & dosagem , Ácido alfa-Linolênico/análise
14.
Biochem Biophys Res Commun ; 263(1): 213-8, 1999 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-10486279

RESUMO

To study the putative role of human carnitine octanoyltransferase (COT) in the beta-oxidation of branched-chain fatty acids, we identified and cloned the cDNA encoding human COT and expressed it in the yeast Saccharomyces cerevisiae. Enzyme activity measurements showed that COT efficiently converts one of the end products of the peroxisomal beta-oxidation of pristanic acid, 4, 8-dimethylnonanoyl-CoA, to its corresponding carnitine ester. Production of the carnitine ester of this branched/medium-chain acyl-CoA within the peroxisome is required for its transport to the mitochondrion where further beta-oxidation occurs. In contrast, 4, 8-dimethylnonanoyl-CoA is not a substrate for carnitine acetyltransferase, another acyltransferase localized in peroxisomes, which catalyzes the formation of carnitine esters of the other products of pristanic acid beta-oxidation, namely acetyl-CoA and propionyl-CoA. Our results shed new light on the function of COT in fatty acid metabolism and point to a crucial role of COT in the beta-oxidation of branched-chain fatty acids.


Assuntos
Carnitina Aciltransferases/genética , Carnitina Aciltransferases/metabolismo , Ácidos Graxos/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Clonagem Molecular , Primers do DNA/genética , DNA Complementar/genética , Expressão Gênica , Humanos , Técnicas In Vitro , Microcorpos/metabolismo , Dados de Sequência Molecular , Oxirredução , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
15.
Lipids ; 33(5): 539-43, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9625603

RESUMO

We report on hepatic effects obtained in vivo by treating mice with different doses of fenoprofen, an arylpropionic acid previously shown to inhibit in vitro peroxisomal very long chain fatty acid oxidation. A strong and dose-related induction of peroxisomal palmitoyl-CoA oxidase, and of carnitine acyltransferase and acyl-CoA hydrolase activities was recorded in liver homogenates of mice fed diets supplemented with different contents [0.01, 0.05, 0.1, or 1% (w/w)] of fenoprofen for 6 d. Peroxisomal glycolate oxidase and mitochondrial butyryl-CoA, octanoyl-CoA, and palmitoyl-CoA dehydrogenases were unaffected or increased. Hepatic catalase activity was significantly increased in mice fed the diet with 0.05 and 0.1% fenoprofen but, surprisingly, was not stimulated in mice fed the 1% fenoprofen-containing diet. A time-related but unequal induction of acyl-CoA oxidases and catalase was observed with the 0.1% fenoprofen diet: at 21 d of treatment, the induction of lignoceroyl-CoA and palmitoyl-CoA oxidase activities were five-fold stronger than that of catalase activity. In mice treated with 1% fenoprofen for up to 6 d, only acyl-CoA oxidase activities were found to be significantly increased. Morphometric analysis of the liver peroxisomes in mice treated with 0.1% fenoprofen evidenced an increase in size, volume density, and surface density along with a reduced ratio between perimeter and area of the peroxisomal profiles. No morphological marker for very long chain fatty acid deposition could be detected in livers from fenoprofen-treated animals. Our findings clearly demonstrate that fenoprofen acts as a peroxisome proliferator in the liver of mice and do not support the occurrence of in vivo reduction of very long chain fatty acid oxidation in liver from treated animals.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Fenoprofeno/farmacologia , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Carnitina Aciltransferases/metabolismo , Fígado/enzimologia , Fígado/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos , Microcorpos/enzimologia , Microcorpos/ultraestrutura , Tamanho do Órgão/efeitos dos fármacos , Oxirredutases/metabolismo , Palmitoil-CoA Hidrolase/metabolismo , Fatores de Tempo
16.
Eur J Biochem ; 247(3): 1029-37, 1997 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9288928

RESUMO

The cDNA for bovine liver carnitine octanoyltransferase (COT) has been cloned by a combination of lambda gt11 library screening and 3' rapid amplification of cDNA ends (3'-RACE). The cDNA comprises 338 bases of 5' non-coding sequence, a reading frame of 1839 bases including the stop codon, and 820 bases of 3' non-coding DNA. The deduced amino acid sequence of 612 residues predicts a protein with a calculated mass of 70263 Da and pI 6.28. The enzyme was expressed in recombinant soluble form in Escherichia coli and was purified by a two-step procedure to near-homogeneity with a yield of purified protein of 2-3 mg/l culture. Recombinant COT had similar kinetic properties to those of the enzyme isolated directly from beef liver. Arg505 in COT, conserved in all reported carnitine acyltransferase sequences but replaced by asparagine or isoleucine in the choline acetyltransferases, was converted to asparagine by site-directed mutagenesis. This single mutation resulted in a greater than 1650-fold increase in the Km value for COT towards carnitine, but had little effect on the value of k(cat) or the Km value for the acyl-CoA substrate. In addition, although choline was an extremely poor substrate for COT, the k(cat)/Km ratio towards this substrate was increased fourfold as a result of the mutation. These data support the notion that Arg505 in COT, and other carnitine acyltransferases, contributes to substrate binding by forming a salt bridge with the carboxylate moiety of carnitine.


Assuntos
Arginina/metabolismo , Ácidos Carboxílicos/metabolismo , Carnitina Aciltransferases/genética , Carnitina/metabolismo , Fígado/enzimologia , Sequência de Aminoácidos , Animais , Arginina/genética , Sequência de Bases , Carnitina/química , Carnitina Aciltransferases/metabolismo , Bovinos , Clonagem Molecular , DNA Complementar , Escherichia coli/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
17.
Pathol Biol (Paris) ; 40(9): 910-9, 1992 Nov.
Artigo em Francês | MEDLINE | ID: mdl-1296165

RESUMO

Although L-carnitine is not considered as an essential nutrient, endogenous synthesis may fail to ensure adequate L-carnitine levels in neonates, especially those born prematurely. Free L-carnitine is found in many foods, mainly those from animal sources. Absorption of free L-carnitine is virtually complete. Lysine and methionine are necessary ingredients for the biosynthesis of L-carnitine. All tissues in the body can produce deoxy-carnitine but, in humans, the enzyme that enables hydroxylation of deoxy-carnitine to carnitine is found only in the liver, brain and kidneys. Complex exchanges of carnitine and its precursors occur between tissues. Muscles take up carnitine from the bloodstream and contain most of the body carnitine stores. L-carnitine and L-carnitine esters are eliminated mainly through the kidneys, which may play a central role in the homeostasis of this compound. Thyroid hormones adrenocorticotrophin (ACTH), and diet all influence urinary excretion of L-carnitine. Free L-carnitine can be assayed in plasma and urine and is occasionally measured in muscle biopsy specimens. Plasma L-carnitine levels may not accurately reflect L-carnitine body stores. L-carnitine ensures transfer of fatty acids to the mitochondria where they undergo oxidation. This process is associated with production of short-chain acylcarnitine which exit from the mitochondria or peroxisomes. L-carnitine ensures regeneration of coenzyme A and is thus involved in energy metabolism. L-carnitine also ensures elimination of xenobiotic substances. Carnitine deficiencies are common. Currently, these deficiencies are classified into two groups. In deficiencies with myopathy, only the muscles are deficient in L-carnitine, perhaps as a result of a primary anomaly of the L-carnitine transport system in muscles. In systemic deficiencies, L-carnitine levels are low in the plasma and in all body tissues. Systemic L-carnitine deficiencies are usually the result of a variety of disease states including deficient intake in premature infants or long-term parenteral nutrition; renal failure; organic acidemias; and Reye's syndrome. Modifications in L-carnitine metabolism have also been reported in patients with diabetes mellitus, malignancies, myocardial ischemia, and alcohol abuse. A large number of supplementation trials have been carried out.


Assuntos
Carnitina/metabolismo , Adolescente , Adulto , Transporte Biológico Ativo/fisiologia , Carnitina/biossíntese , Carnitina/deficiência , Carnitina Aciltransferases/metabolismo , Criança , Pré-Escolar , Ácidos Graxos/metabolismo , Humanos , Lactente , Recém-Nascido , Doenças Musculares/metabolismo , Insuficiência Renal/metabolismo , Xenobióticos/metabolismo
18.
Prostaglandins ; 37(3): 335-44, 1989 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2748915

RESUMO

The investigations previously carried out by Grataroli and colleagues (1) to elucidate the relationships between dietary fatty acids, lipid composition, prostaglandin E2 production and phospholipase A2 activity in the rat gastric mucosa are, here, extended. In the present investigations, fatty acid and prostaglandin E2 catabolizing enzymes were assayed in gastric mucosa from rats fed either a low fat diet (corn oil: 4.4% w/w) (referred as control group), a corn oil-enriched diet (17%) or a salmon oil-enriched diet (12.5%) supplemented with corn oil (4.5%) (referred as groups of treated animals) for eight weeks. Peroxisomal fatty acyl-CoA beta-oxidation was induced in the treated animals whereas the activities of catalase and mitochondrial tyramine oxidase were increased and normal, respectively. Mitochondrial acyl-CoA dehydrogenations occurred at higher rates and carnitine acyltransferase activities were enhanced. In addition, the induction of peroxisomal but not mitochondrial prostaglandoyl-E2-CoA beta-oxidation could be demonstrated. Induction of peroxisomal oxidation of fatty acids and prostaglandins is suggested to contribute to the decrease of prostaglandin E2 production in the treated animals, especially those receiving the salmon oil diet, that the above mentioned authors originally reported.


Assuntos
Óleo de Milho/farmacologia , Dinoprostona/metabolismo , Ácidos Graxos/metabolismo , Óleos de Peixe/farmacologia , Mucosa Gástrica/metabolismo , Óleos de Plantas/farmacologia , Animais , Carnitina Aciltransferases/metabolismo , Dieta , Ácidos Graxos Dessaturases/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/enzimologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Oxirredução , Ratos , Ratos Endogâmicos , Salmão
19.
Biochemistry ; 27(25): 9000-6, 1988 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-3233218

RESUMO

Several complementary DNAs for the peroxisomal enzyme carnitine octanoyltransferase (COT), cloned in the expression vector lambda gt11, have been isolated. Together, these clones cover 2143 bp of the COT cDNA sequence with an open reading frame for 523 amino acids. Northern analysis showed the mRNA size for this enzyme to be 3.5 kilobases. The 523 residue long amino acid sequence amounts to a molecular mass of 60,269 daltons, indicating that the cloned cDNAs contain most or all of the coding sequence for COT (Mr approximately 62,000). Hybridization studies showed that the increased COT activity in the liver of rats, fed the potent peroxisome-proliferating drug Wy-14,643, is associated with a more than 40-fold rise in the steady-state level of the COT mRNA.


Assuntos
Aciltransferases/metabolismo , Carnitina Aciltransferases/metabolismo , Clonagem Molecular , Fígado/enzimologia , Microcorpos/enzimologia , Transcrição Gênica , Sequência de Aminoácidos , Animais , Anticolesterolemiantes , Sequência de Bases , Northern Blotting , Carnitina Aciltransferases/biossíntese , Carnitina Aciltransferases/genética , DNA/genética , Eletroforese em Gel de Poliacrilamida , Fígado/ultraestrutura , Microcorpos/efeitos dos fármacos , Dados de Sequência Molecular , Peso Molecular , Hibridização de Ácido Nucleico , Pirimidinas/farmacologia , RNA Mensageiro/genética , Ratos
20.
Pediatr Res ; 22(6): 748-54, 1987 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-3431962

RESUMO

Chlorpromazine and related drugs including trifluoperazine, clopenthixol, and fluphenazine are in vitro inhibitors of mitochondrial carnitine palmitoyltransferase and cytochrome c oxidase and of peroxisomal carnitine octanoyltransferase from mouse heart and liver. By contrast with 0.1% ethyl 2(5(4-chlorophenyl)pentyl) oxiran-2-carboxylic acid or 0.1% clofibrate-containing diets, the treatment of mice with 0.1% chlorpromazine-containing diet fails to induce peroxisomal proliferation in liver and heart. An 0.5% chlorpromazine-containing diet did induce peroxisomal proliferation. Inhibition of peroxisomal beta-oxidation presumably via the reduction of carnitine octanoyltransferase by chlorpromazine elicits the appearance in liver of lamellar structures resembling those seen in human peroxisomal disorders and induces accumulation of very long-chain fatty acids in plasma. The peroxisomal proliferation induced by administration of high dose chlorpromazine is ascribed to its ability to depress mitochondrial fatty acid oxidation by impairing cytochrome c oxidase and carnitine palmitoyltransferase activities.


Assuntos
Clorpromazina/farmacologia , Gorduras na Dieta/administração & dosagem , Compostos de Epóxi/farmacologia , Éteres Cíclicos/farmacologia , Microcorpos/efeitos dos fármacos , Animais , Carnitina Aciltransferases/metabolismo , Catalase/metabolismo , Coração/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/ultraestrutura , Masculino , Camundongos , Microcorpos/metabolismo , Microcorpos/ultraestrutura , Microscopia Eletrônica , Miocárdio/metabolismo , Miocárdio/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA