Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PeerJ ; 12: e16771, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38406279

RESUMO

Background: Gastric cancer is a very common gastrointestinal tumor with a high mortality rate. Nintedanib has been shown to significantly reduce tumor cell proliferation and increase apoptosis in gastric cancer cells in vitro. However, its systemic action mechanism on gastric cancer cells remains unclear. A high-throughput proteomic approach should help identify the potential mechanisms and targets of nintedanib on gastric cancer cells. Methods: The effects of nintedanib on the biological behavior of gastric cancer cells were evaluated. A cytotoxic proliferation assay was performed to estimate the half maximal inhibitory concentration (IC50). AGS cells were divided into control, and nintedanib-treated groups (5 µM, 48 h), and differential protein expression was investigated using tandem mass tags (TMT) proteomics. The molecular mechanisms of these differentially expressed proteins and their network interactions were then analyzed using bioinformatics, and potential nintedanib targets were identified. Results: This study identified 845 differentially expressed proteins in the nintedanib-treated group (compared to the control group), comprising 526 up-regulated and 319 down-regulated proteins. Bioinformatics analysis revealed that the differentially expressed proteins were primarily enriched in biological pathways for branched-chain amino acid metabolism, steroid biosynthesis, propionate metabolism, fatty acid metabolism, lysosome, peroxisome, and ferroptosis. Key driver analysis revealed that proteins, such as enoyl-CoA hydratase and 3-hydroxyacyl CoA dehydrogenase (EHHADH), isocitrate dehydrogenase 1 (IDH1), acyl-CoA oxidase 1 (ACOX1), acyl-CoA oxidase 2 (ACOX2), acyl-CoA oxidase 3 (ACOX3), and acetyl-CoA acyltransferase 1 (ACAA1) could be linked with nintedanib action. Conclusion: Nintedanib inhibits the proliferation, invasion, and metastasis of gastric cancer cells. The crossover pathways and protein networks predicted by proteomics should provide more detailed molecular information enabling the use of nintedanib against gastric cancer.


Assuntos
Indóis , Neoplasias Gástricas , Humanos , Acil-CoA Oxidase/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Proteômica , Fígado/metabolismo , Enzima Bifuncional do Peroxissomo/metabolismo
2.
J Lipid Res ; 64(3): 100326, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36592657

RESUMO

Compared with other species, freshwater fish are more capable of synthesizing DHA via same biosynthetic pathways. Freshwater fish have a "Sprecher" pathway to biosynthesize DHA in a peroxisome-dependent manner. Enoyl-CoA hydratase/3-hydroxyacyl CoA dehydrogenase (Ehhadh) is involved in the hydration and dehydrogenation reactions of fatty acid ß-oxidation in peroxisomes. However, the role of Ehhadh in the synthesis of DHA in freshwater fish remains largely unclear. In this study, the knockout of Ehhadh significantly inhibited DHA synthesis in zebrafish. Liver transcriptome analysis showed that Ehhadh deletion significantly inhibited SREBF and PPAR signaling pathways and decreased the expression of PUFA synthesis-related genes. Our results from the analysis of transgenic zebrafish (Tg:Ehhadh) showed that Ehhadh overexpression significantly increased the DHA content in the liver and significantly upregulated the expression of genes related to PUFA synthesis. In addition, the DHA content in the liver of Tg:Ehhadh fed with linseed oil was significantly higher than that of wildtype, but the expression of PUFA synthesis-related genes fads2 and elovl2 were significantly lower, indicating that Ehhadh had a direct effect on DHA synthesis. In conclusion, our results showed that Ehhadh was essential for DHA synthesis in the "Sprecher" pathway, and Ehhadh overexpression could promote DHA synthesis. This study provides insight into the role of Ehhadh in freshwater fish.


Assuntos
Enoil-CoA Hidratase , Peixe-Zebra , Animais , Enzima Bifuncional do Peroxissomo/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , Enoil-CoA Hidratase/farmacologia , Peroxissomos/metabolismo , Fígado/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/genética , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/farmacologia , Acetiltransferases/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Biomed Res Int ; 2021: 9917060, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33997049

RESUMO

Enoyl-CoA hydratase and 3-hydroxyacyl CoA dehydrogenase (EHHADH), a member of the 3-hydroxyacyl-CoA dehydrogenase family, were previously demonstrated to be involved in the tumorigenesis of various cancer types. This study is aimed at determining of the diagnostic and prognostic value of EHHADH in osteosarcoma (OS). The overexpression of EHHADH was found both in OS and also other sarcoma types, and according to the retrospective cohort study, the EHHADH level was related to the overall survival and disease-free survival of the OS patients. Furthermore, knockdown of EHHADH under the influence of EHHADH small interfering RNA significantly suppressed the proliferation ability of the tumor cells. Moreover, EHHADH overexpressed was found in human OS tissues. In summary, the progression of OS could be enhanced by EHHADH, which may be a potential diagnostic and prognostic biomarker for OS patients.


Assuntos
Osteossarcoma , Enzima Bifuncional do Peroxissomo , Linhagem Celular Tumoral , Humanos , Osteossarcoma/diagnóstico , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/mortalidade , Enzima Bifuncional do Peroxissomo/genética , Enzima Bifuncional do Peroxissomo/metabolismo , Prognóstico , Mapas de Interação de Proteínas/genética , RNA Interferente Pequeno/genética , Estudos Retrospectivos
4.
BMC Cancer ; 21(1): 48, 2021 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-33430801

RESUMO

BACKGROUND: Cisplatin-based chemotherapy is recommended as the primary treatment for advanced bladder cancer (BC) with unresectable or metastatic disease. However, the benefits are limited due to the acquisition of drug resistance. The mechanisms of resistance remain unclear. Although there are some reports that some molecules are associated with cisplatin resistance in advanced BC, those reports have not been fully investigated. Therefore, we undertook a new search for cisplatin resistance-related genes targeted by tumor suppressive microRNAs as well as genes that were downregulated in cisplatin-resistant BC cells and clinical BC tissues. METHODS: First, we established cisplatin-resistant BOY and T24 BC cell lines (CDDP-R-BOY, CDDP-R-T24). Then, Next Generation Sequence analysis was performed with parental and cisplatin-resistant cell lines to search for the microRNAs responsible for cisplatin resistance. We conducted gain-of-function analysis of microRNAs and their effects on cisplatin resistance, and we searched target genes comprehensively using Next Generation mRNA sequences. RESULTS: A total of 28 microRNAs were significantly downregulated in both CDDP-R-BOY and CDDP-R-T24. Among them, miR-486-5p, a tumor suppressor miRNA, was negatively correlated with the TNM classification of clinical BC samples in The Cancer Genome Atlas (TCGA) database. Transfection of miRNA-486-5p significantly inhibited cancer cell proliferation, migration, and invasion, and also improved the cells' resistance to cisplatin. Among the genes targeted by miRNA-486-5p, we focused on enoyl-CoA, hydratase/3-hydroxyacyl CoA dehydrogenase (EHHADH), which is involved in the degradation of fatty acids. EHHADH was directly regulated by miRNA-486-5p as determined by a dual-luciferase reporter assay. Loss-of-function study using EHHADH si-RNA showed significant inhibitions of cell proliferation, migration, invasion and the recovery of cisplatin sensitivity. CONCLUSION: Identification of EHHADH as a target of miRNA-486-5p provides novel insights into the potential mechanisms of cisplatin resistance in BC.


Assuntos
Biomarcadores Tumorais/metabolismo , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Enzima Bifuncional do Peroxissomo/metabolismo , Neoplasias da Bexiga Urinária/patologia , Animais , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Enzima Bifuncional do Peroxissomo/genética , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Molecules ; 22(7)2017 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-28684700

RESUMO

A patented organotin di-n-butyl-di-(4-chlorobenzohydroxamato)tin (DBDCT) with high a antitumor activity was designed, however, its antitumor and toxic mechanisms have not yet been clearly illustrated. Hepatic proteins of DBDCT-treated rats were identified and analyzed using LC-MS/MS with label-free quantitative technology. In total, 149 differentially expressed proteins were successfully identified. Five protein and mRNA expressions were involved in the peroxisome proliferator-activated receptor (PPAR) signaling pathway, including a scavenger receptor (CD36), adipocyte fatty acid binding protein 4 (FABP4), enoyl-CoA hydratase (EHHADH), acetyl-CoA acyltransferase 1 (ACAA1), and phosphoenolpyruvate carboxykinase (PEPCK) in DBDCT-treated Rat Liver (BRL) cells. PPAR-α and PPAR-λ were also significantly decreased at both protein and mRNA levels. Furthermore, compared with the DBDCT treatment group, a special blocking agent of PPAR-λ T0070907 was used to evaluate the relationship between PPAR-λ and its downstream genes. Our studies indicated that DBDCT may serve as a modulator of PPAR-λ, further up-regulating CD36, FABP4 and EHHADH on the PPAR signal pathway.


Assuntos
Fígado/metabolismo , Fígado/patologia , Compostos Orgânicos de Estanho/toxicidade , PPAR alfa/metabolismo , Proteômica/métodos , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Antígenos CD36/metabolismo , Caspases/metabolismo , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Proteínas de Ligação a Ácido Graxo/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Compostos Orgânicos de Estanho/química , PPAR alfa/genética , Enzima Bifuncional do Peroxissomo/genética , Enzima Bifuncional do Peroxissomo/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar
6.
Cell Rep ; 15(7): 1423-1429, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27160910

RESUMO

We recently reported an autosomal dominant form of renal Fanconi syndrome caused by a missense mutation in the third codon of the peroxisomal protein EHHADH. The mutation mistargets EHHADH to mitochondria, thereby impairing mitochondrial energy production and, consequently, reabsorption of electrolytes and low-molecular-weight nutrients in the proximal tubule. Here, we further elucidate the molecular mechanism underlying this pathology. We find that mutated EHHADH is incorporated into mitochondrial trifunctional protein (MTP), thereby disturbing ß-oxidation of long-chain fatty acids. The resulting MTP deficiency leads to a characteristic accumulation of hydroxyacyl- and acylcarnitines. Mutated EHHADH also limits respiratory complex I and corresponding supercomplex formation, leading to decreases in oxidative phosphorylation capacity, mitochondrial membrane potential maintenance, and ATP generation. Activity of the Na(+)/K(+)-ATPase is thereby diminished, ultimately decreasing the transport activity of the proximal tubule cells.


Assuntos
Síndrome de Fanconi/metabolismo , Rim/metabolismo , Rim/patologia , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Animais , Transporte Biológico , Extratos Celulares , Metabolismo Energético , Síndrome de Fanconi/complicações , Síndrome de Fanconi/patologia , Ácidos Graxos/metabolismo , Células LLC-PK1 , Microscopia Confocal , Doenças Mitocondriais/complicações , Doenças Mitocondriais/patologia , Mutação/genética , Oxirredução , Enzima Bifuncional do Peroxissomo/metabolismo , Proteômica , ATPase Trocadora de Sódio-Potássio/metabolismo , Frações Subcelulares/metabolismo , Suínos
7.
Reprod Sci ; 23(10): 1422-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27076444

RESUMO

Preeclampsia (PE) is a pregnancy disorder characterized by high blood pressure, placental oxidative stress, and proteinuria. In a GeneFishing experiment using human preeclamptic placenta, expression of acyl-coenzyme A dehydrogenase very long chain (ACADVL), which is involved in fatty acid ß-oxidation (FAO), was detected. To investigate the correlation between PE and FAO, this study subjected in vitro BeWo cells and in vivo pregnant mice to oxidative stress induced by hypoxia. Hypoxic condition, which oxygen supply is insufficient in cells and placenta, created a similar state to placental oxidative stress in PE, as evidenced by increased hypoxic (oxoguanine DNA glycosylase 1, hypoxia inducible factor 1 alpha subunit) and preeclamptic markers (soluble fms-like tyrosine kinase 1) both in vitro and in vivo. Increased expression of FAO-related genes (ACADVL, enoyl-coenzyme A hydratase/3-hydroxyacyl coenzyme A dehydrogenase) was observed in these models as well as in cases of preeclamptic preterm labor. In the in vivo liver model, messenger RNA expression of gluconeogenesis-related genes increased. Consequently, these results suggest that expression of FAO-related genes is regulated by hypoxic conditions and onset time of PE and affects maternal gluconeogenesis during pregnancy in patients with PE.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Ácidos Graxos/metabolismo , Estresse Oxidativo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Animais , Gasometria , Hipóxia Celular , Linhagem Celular Tumoral , DNA Glicosilases/metabolismo , Feminino , Gluconeogênese , Glicogenólise , Humanos , Fígado/metabolismo , Camundongos , Enzima Bifuncional do Peroxissomo/metabolismo , Gravidez , RNA Mensageiro/metabolismo
8.
Cell Rep ; 14(10): 2273-80, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26947074

RESUMO

The cJun NH2-terminal kinase (JNK)-signaling pathway is implicated in metabolic syndrome, including dysregulated blood glucose concentration and insulin resistance. Fibroblast growth factor 21 (FGF21) is a target of the hepatic JNK-signaling pathway and may contribute to the regulation of glycemia. To test the role of FGF21, we established mice with selective ablation of the Fgf21 gene in hepatocytes. FGF21 deficiency in the liver caused marked loss of FGF21 protein circulating in the blood. Moreover, the protective effects of hepatic JNK deficiency to suppress metabolic syndrome in high-fat diet-fed mice were not observed in mice with hepatocyte-specific FGF21 deficiency, including reduced blood glucose concentration and reduced intolerance to glucose and insulin. Furthermore, we show that JNK contributes to the regulation of hepatic FGF21 expression during fasting/feeding cycles. These data demonstrate that the hepatokine FGF21 is a key mediator of JNK-regulated metabolic syndrome.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Doenças Metabólicas/etiologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Glicemia/análise , Células Cultivadas , Dieta Hiperlipídica , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/citologia , Hepatócitos/metabolismo , Insulina/sangue , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Leptina/sangue , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Masculino , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Enzima Bifuncional do Peroxissomo/genética , Enzima Bifuncional do Peroxissomo/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Resistina/sangue , Transdução de Sinais , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
9.
Circulation ; 131(23): 2061-2069, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-25862742

RESUMO

BACKGROUND: Epidemiological findings suggest a relationship between Alzheimer disease (AD), inflammation, and dyslipidemia, although the nature of this relationship is not well understood. We investigated whether this phenotypic association arises from a shared genetic basis. METHODS AND RESULTS: Using summary statistics (P values and odds ratios) from genome-wide association studies of >200 000 individuals, we investigated overlap in single-nucleotide polymorphisms associated with clinically diagnosed AD and C-reactive protein (CRP), triglycerides, and high- and low-density lipoprotein levels. We found up to 50-fold enrichment of AD single-nucleotide polymorphisms for different levels of association with C-reactive protein, low-density lipoprotein, high-density lipoprotein, and triglyceride single-nucleotide polymorphisms using a false discovery rate threshold <0.05. By conditioning on polymorphisms associated with the 4 phenotypes, we identified 55 loci associated with increased AD risk. We then conducted a meta-analysis of these 55 variants across 4 independent AD cohorts (total: n=29 054 AD cases and 114 824 healthy controls) and discovered 2 genome-wide significant variants on chromosome 4 (rs13113697; closest gene, HS3ST1; odds ratio=1.07; 95% confidence interval=1.05-1.11; P=2.86×10(-8)) and chromosome 10 (rs7920721; closest gene, ECHDC3; odds ratio=1.07; 95% confidence interval=1.04-1.11; P=3.38×10(-8)). We also found that gene expression of HS3ST1 and ECHDC3 was altered in AD brains compared with control brains. CONCLUSIONS: We demonstrate genetic overlap between AD, C-reactive protein, and plasma lipids. By conditioning on the genetic association with the cardiovascular phenotypes, we identify novel AD susceptibility loci, including 2 genome-wide significant variants conferring increased risk for AD.


Assuntos
Doença de Alzheimer/genética , Proteína C-Reativa/metabolismo , Dislipidemias/genética , Estudo de Associação Genômica Ampla , Inflamação/genética , Lipídeos/sangue , Herança Multifatorial/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/epidemiologia , Biomarcadores/metabolismo , Encéfalo/metabolismo , Proteína C-Reativa/genética , Dislipidemias/complicações , Feminino , Humanos , Inflamação/complicações , Lipídeos/genética , Masculino , Enzima Bifuncional do Peroxissomo/genética , Enzima Bifuncional do Peroxissomo/metabolismo , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Sulfotransferases/genética , Sulfotransferases/metabolismo
10.
PLoS One ; 9(1): e87179, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475249

RESUMO

Uridine, a pyrimidine nucleoside, can modulate liver lipid metabolism although its specific acting targets have not been identified. Using mice with fenofibrate-induced fatty liver as a model system, the effects of uridine on liver lipid metabolism are examined. At a daily dosage of 400 mg/kg, fenofibrate treatment causes reduction of liver NAD(+)/NADH ratio, induces hyper-acetylation of peroxisomal bifunctional enzyme (ECHD) and acyl-CoA oxidase 1 (ACOX1), and induces excessive accumulation of long chain fatty acids (LCFA) and very long chain fatty acids (VLCFA). Uridine co-administration at a daily dosage of 400 mg/kg raises NAD(+)/NADH ratio, inhibits fenofibrate-induced hyper-acetylation of ECHD, ACOX1, and reduces accumulation of LCFA and VLCFA. Our data indicates a therapeutic potential for uridine co-administration to prevent fenofibrate-induced fatty liver.


Assuntos
Fígado Gorduroso/prevenção & controle , Fenofibrato/efeitos adversos , Hipolipemiantes/efeitos adversos , Substâncias Protetoras/farmacologia , Uridina/farmacologia , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Ácidos Graxos/metabolismo , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/enzimologia , Fígado Gorduroso/patologia , Regulação da Expressão Gênica , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NAD/metabolismo , Oxirredução/efeitos dos fármacos , Enzima Bifuncional do Peroxissomo/genética , Enzima Bifuncional do Peroxissomo/metabolismo
11.
N Engl J Med ; 370(2): 129-38, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24401050

RESUMO

BACKGROUND: In renal Fanconi's syndrome, dysfunction in proximal tubular cells leads to renal losses of water, electrolytes, and low-molecular-weight nutrients. For most types of isolated Fanconi's syndrome, the genetic cause and underlying defect remain unknown. METHODS: We clinically and genetically characterized members of a five-generation black family with isolated autosomal dominant Fanconi's syndrome. We performed genomewide linkage analysis, gene sequencing, biochemical and cell-biologic investigations of renal proximal tubular cells, studies in knockout mice, and functional evaluations of mitochondria. Urine was studied with the use of proton nuclear magnetic resonance ((1)H-NMR) spectroscopy. RESULTS: We linked the phenotype of this family's Fanconi's syndrome to a single locus on chromosome 3q27, where a heterozygous missense mutation in EHHADH segregated with the disease. The p.E3K mutation created a new mitochondrial targeting motif in the N-terminal portion of EHHADH, an enzyme that is involved in peroxisomal oxidation of fatty acids and is expressed in the proximal tubule. Immunocytofluorescence studies showed mistargeting of the mutant EHHADH to mitochondria. Studies of proximal tubular cells revealed impaired mitochondrial oxidative phosphorylation and defects in the transport of fluids and a glucose analogue across the epithelium. (1)H-NMR spectroscopy showed elevated levels of mitochondrial metabolites in urine from affected family members. Ehhadh knockout mice showed no abnormalities in renal tubular cells, a finding that indicates a dominant negative nature of the mutation rather than haploinsufficiency. CONCLUSIONS: Mistargeting of peroxisomal EHHADH disrupts mitochondrial metabolism and leads to renal Fanconi's syndrome; this indicates a central role of mitochondria in proximal tubular function. The dominant negative effect of the mistargeted protein adds to the spectrum of monogenic mechanisms of Fanconi's syndrome. (Funded by the European Commission Seventh Framework Programme and others.).


Assuntos
Síndrome de Fanconi/genética , Túbulos Renais Proximais/metabolismo , Mitocôndrias/metabolismo , Mutação de Sentido Incorreto , Enzima Bifuncional do Peroxissomo/genética , Sequência de Aminoácidos , Animais , População Negra , Cromossomos Humanos Par 3 , Modelos Animais de Doenças , Síndrome de Fanconi/etnologia , Feminino , Ligação Genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Linhagem , Enzima Bifuncional do Peroxissomo/química , Enzima Bifuncional do Peroxissomo/metabolismo , Fenótipo , Análise de Sequência de DNA
12.
Obesity (Silver Spring) ; 21(6): 1200-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23666909

RESUMO

OBJECTIVE: The effects of different amounts of omega 3-polyunsaturated fatty acids in diets with normal or high content of fat on lipid and carbohydrate metabolism were investigated. DESIGN AND METHODS: Mice were fed for 8 weeks on diets enriched with fish oil or lard at 10% or 60% of energy. Energy balance and energy expenditure were analyzed. Fatty acid (FA) oxidative capacity of the liver and the activity of enzymes involved in this pathway were assessed. RESULTS: Fish oil-fed mice had lower body weight and adiposity compared with lard-fed animals, despite having lower rates of oxygen consumption. Mice fed diets containing fish oil also displayed lower glycemia, reduced fat content in the liver, and improved glucose tolerance compared with lard-fed animals. The fish oil-containing diets increased markers of hepatic peroxisomal content and increased the generation of metabolites derived from FA ß-oxidation in liver homogenates. In contrast, no changes were observed in the content of mitochondrial electron transport chain proteins or carnitine palmitoyl transferase-1 in the liver, indicating little direct effect of fish oil on mitochondrial metabolism. CONCLUSION: Collectively, our findings suggest that the energy inefficient oxidation of FAs in peroxisomes may be an important mechanism underlying the protection against obesity and glucose intolerance of fish oil administration.


Assuntos
Dieta , Óleos de Peixe/administração & dosagem , Intolerância à Glucose/prevenção & controle , Obesidade/prevenção & controle , Enzima Bifuncional do Peroxissomo/metabolismo , Adiposidade/efeitos dos fármacos , Animais , Metabolismo dos Carboidratos/efeitos dos fármacos , Carnitina O-Palmitoiltransferase/metabolismo , Gorduras na Dieta/administração & dosagem , Metabolismo Energético/efeitos dos fármacos , Ácidos Graxos Ômega-3/administração & dosagem , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Camundongos , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...