Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
PeerJ ; 12: e16771, 2024.
Article in English | MEDLINE | ID: mdl-38406279

ABSTRACT

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.


Subject(s)
Indoles , Stomach Neoplasms , Humans , Acyl-CoA Oxidase/metabolism , Stomach Neoplasms/drug therapy , Proteomics , Liver/metabolism , Peroxisomal Bifunctional Enzyme/metabolism
2.
J Lipid Res ; 64(3): 100326, 2023 03.
Article in English | MEDLINE | ID: mdl-36592657

ABSTRACT

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.


Subject(s)
Enoyl-CoA Hydratase , Zebrafish , Animals , Peroxisomal Bifunctional Enzyme/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , Enoyl-CoA Hydratase/pharmacology , Peroxisomes/metabolism , Liver/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/pharmacology , Acetyltransferases/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
Biomed Res Int ; 2021: 9917060, 2021.
Article in English | MEDLINE | ID: mdl-33997049

ABSTRACT

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.


Subject(s)
Osteosarcoma , Peroxisomal Bifunctional Enzyme , Cell Line, Tumor , Humans , Osteosarcoma/diagnosis , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/mortality , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism , Prognosis , Protein Interaction Maps/genetics , RNA, Small Interfering/genetics , Retrospective Studies
4.
BMC Cancer ; 21(1): 48, 2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33430801

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Peroxisomal Bifunctional Enzyme/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Peroxisomal Bifunctional Enzyme/genetics , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor Assays
5.
Molecules ; 22(7)2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28684700

ABSTRACT

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.


Subject(s)
Liver/metabolism , Liver/pathology , Organotin Compounds/toxicity , PPAR alpha/metabolism , Proteomics/methods , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , CD36 Antigens/metabolism , Caspases/metabolism , Cell Line , Enzyme Activation/drug effects , Fatty Acid-Binding Proteins/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Organotin Compounds/chemistry , PPAR alpha/genetics , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar
6.
Cell Rep ; 15(7): 1423-1429, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160910

ABSTRACT

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.


Subject(s)
Fanconi Syndrome/metabolism , Kidney/metabolism , Kidney/pathology , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Animals , Biological Transport , Cell Extracts , Energy Metabolism , Fanconi Syndrome/complications , Fanconi Syndrome/pathology , Fatty Acids/metabolism , LLC-PK1 Cells , Microscopy, Confocal , Mitochondrial Diseases/complications , Mitochondrial Diseases/pathology , Mutation/genetics , Oxidation-Reduction , Peroxisomal Bifunctional Enzyme/metabolism , Proteomics , Sodium-Potassium-Exchanging ATPase/metabolism , Subcellular Fractions/metabolism , Swine
7.
Reprod Sci ; 23(10): 1422-33, 2016 10.
Article in English | MEDLINE | ID: mdl-27076444

ABSTRACT

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.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Fatty Acids/metabolism , Oxidative Stress , Placenta/metabolism , Pre-Eclampsia/metabolism , Animals , Blood Gas Analysis , Cell Hypoxia , Cell Line, Tumor , DNA Glycosylases/metabolism , Female , Gluconeogenesis , Glycogenolysis , Humans , Liver/metabolism , Mice , Peroxisomal Bifunctional Enzyme/metabolism , Pregnancy , RNA, Messenger/metabolism
8.
Cell Rep ; 14(10): 2273-80, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947074

ABSTRACT

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.


Subject(s)
Fibroblast Growth Factors/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Metabolic Diseases/etiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Blood Glucose/analysis , Cells, Cultured , Diet, High-Fat , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/genetics , Gene Expression Regulation/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Insulin/blood , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Leptin/blood , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism , Protein Kinase Inhibitors/pharmacology , Real-Time Polymerase Chain Reaction , Resistin/blood , Signal Transduction , Mitogen-Activated Protein Kinase Kinase Kinase 11
9.
Circulation ; 131(23): 2061-2069, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25862742

ABSTRACT

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.


Subject(s)
Alzheimer Disease/genetics , C-Reactive Protein/metabolism , Dyslipidemias/genetics , Genome-Wide Association Study , Inflammation/genetics , Lipids/blood , Multifactorial Inheritance/genetics , Aged , Aged, 80 and over , Alzheimer Disease/epidemiology , Biomarkers/metabolism , Brain/metabolism , C-Reactive Protein/genetics , Dyslipidemias/complications , Female , Humans , Inflammation/complications , Lipids/genetics , Male , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism , Phenotype , Polymorphism, Single Nucleotide/genetics , Risk Factors , Sulfotransferases/genetics , Sulfotransferases/metabolism
10.
PLoS One ; 9(1): e87179, 2014.
Article in English | MEDLINE | ID: mdl-24475249

ABSTRACT

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.


Subject(s)
Fatty Liver/prevention & control , Fenofibrate/adverse effects , Hypolipidemic Agents/adverse effects , Protective Agents/pharmacology , Uridine/pharmacology , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Biological Transport/drug effects , Fatty Acids/metabolism , Fatty Liver/chemically induced , Fatty Liver/enzymology , Fatty Liver/pathology , Gene Expression Regulation , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD/metabolism , Oxidation-Reduction/drug effects , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism
11.
N Engl J Med ; 370(2): 129-38, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24401050

ABSTRACT

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.).


Subject(s)
Fanconi Syndrome/genetics , Kidney Tubules, Proximal/metabolism , Mitochondria/metabolism , Mutation, Missense , Peroxisomal Bifunctional Enzyme/genetics , Amino Acid Sequence , Animals , Black People , Chromosomes, Human, Pair 3 , Disease Models, Animal , Fanconi Syndrome/ethnology , Female , Genetic Linkage , Humans , Male , Mice , Mice, Knockout , Molecular Sequence Data , Pedigree , Peroxisomal Bifunctional Enzyme/chemistry , Peroxisomal Bifunctional Enzyme/metabolism , Phenotype , Sequence Analysis, DNA
12.
Obesity (Silver Spring) ; 21(6): 1200-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23666909

ABSTRACT

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.


Subject(s)
Diet , Fish Oils/administration & dosage , Glucose Intolerance/prevention & control , Obesity/prevention & control , Peroxisomal Bifunctional Enzyme/metabolism , Adiposity/drug effects , Animals , Carbohydrate Metabolism/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Dietary Fats/administration & dosage , Energy Metabolism/drug effects , Fatty Acids, Omega-3/administration & dosage , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Male , Mice , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...