Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 4138, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36914879

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant that induces the progression of steatosis to steatohepatitis with fibrosis in mice. Furthermore, TCDD reprograms hepatic metabolism by redirecting glycolytic intermediates while inhibiting lipid metabolism. Here, we examined the effect of TCDD on hepatic acetyl-coenzyme A (acetyl-CoA) and ß-hydroxybutyrate levels as well as protein acetylation and ß-hydroxybutyrylation. Acetyl-CoA is not only a central metabolite in multiple anabolic and catabolic pathways, but also a substrate used for posttranslational modification of proteins and a surrogate indicator of cellular energy status. Targeted metabolomic analysis revealed a dose-dependent decrease in hepatic acetyl-CoA levels coincident with the phosphorylation of pyruvate dehydrogenase (E1), and the induction of pyruvate dehydrogenase kinase 4 and pyruvate dehydrogenase phosphatase, while repressing ATP citrate lyase and short-chain acyl-CoA synthetase gene expression. In addition, TCDD dose-dependently reduced the levels of hepatic ß-hydroxybutyrate and repressed ketone body biosynthesis gene expression. Moreover, levels of total hepatic protein acetylation and ß-hydroxybutyrylation were reduced. AMPK phosphorylation was induced consistent with acetyl-CoA serving as a cellular energy status surrogate, yet subsequent targets associated with re-establishing energy homeostasis were not activated. Collectively, TCDD reduced hepatic acetyl-CoA and ß-hydroxybutyrate levels eliciting starvation-like conditions despite normal levels of food intake.


Subject(s)
Fatty Liver , Polychlorinated Dibenzodioxins , Mice , Animals , Acetyl Coenzyme A/metabolism , Polychlorinated Dibenzodioxins/toxicity , 3-Hydroxybutyric Acid/metabolism , Liver/metabolism , Fatty Liver/metabolism
2.
Sci Rep ; 11(1): 15689, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344994

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a persistent environmental contaminant, induces steatosis by increasing hepatic uptake of dietary and mobilized peripheral fats, inhibiting lipoprotein export, and repressing ß-oxidation. In this study, the mechanism of ß-oxidation inhibition was investigated by testing the hypothesis that TCDD dose-dependently repressed straight-chain fatty acid oxidation gene expression in mice following oral gavage every 4 days for 28 days. Untargeted metabolomic analysis revealed a dose-dependent decrease in hepatic acyl-CoA levels, while octenoyl-CoA and dicarboxylic acid levels increased. TCDD also dose-dependently repressed the hepatic gene expression associated with triacylglycerol and cholesterol ester hydrolysis, fatty acid binding proteins, fatty acid activation, and 3-ketoacyl-CoA thiolysis while inducing acyl-CoA hydrolysis. Moreover, octenoyl-CoA blocked the hydration of crotonyl-CoA suggesting short chain enoyl-CoA hydratase (ECHS1) activity was inhibited. Collectively, the integration of metabolomics and RNA-seq data suggested TCDD induced a futile cycle of fatty acid activation and acyl-CoA hydrolysis resulting in incomplete ß-oxidation, and the accumulation octenoyl-CoA levels that inhibited the activity of short chain enoyl-CoA hydratase (ECHS1).


Subject(s)
Environmental Pollutants/pharmacology , Liver/drug effects , Liver/metabolism , Oxidation-Reduction/drug effects , Polychlorinated Dibenzodioxins/pharmacology , Thiolester Hydrolases/antagonists & inhibitors , Acyl Coenzyme A/metabolism , Fatty Acids/metabolism , Lipid Metabolism/drug effects , Mitochondria/metabolism , Peroxisomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...