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.
Cell Physiol Biochem ; 39(4): 1648-62, 2016.
Article in English | MEDLINE | ID: mdl-27626926

ABSTRACT

BACKGROUND/AIMS: Nonalcoholic fatty liver disease (NAFLD) progression to fibrosis, cirrhosis and hepatocellular carcinoma, alters the cellular composition of this organ. During late-stage NAFLD, fibrotic and possibly cancerous cells can proliferate and, like normal hepatocytes, are exposed to high concentrations of fatty acids from both surrounding tissue and circulating lipid sources. We hypothesized that primary human activated hepatic stellate cells and epithelial hepatoma (HepG2) cells respond differently to lipotoxic conditions, and investigated the mechanisms involved. METHODS: Primary activated hepatic stellate cells and HepG2 cells were exposed to pathophysiological concentrations of fatty acids and comparative studies of lipid metabolic and stress response pathways were performed. RESULTS: Both cell types remained proliferative during exposure to a combination of palmitate plus oleate reflective of the general saturated versus unsaturated fatty acid composition of western diets. However, exposure to either high palmitate or high oleate alone induced cytotoxicity in activated stellate cells, while only palmitate caused cytotoxicity in HepG2 cells. mRNA microarray and biochemical comparisons revealed that stellate cells stored markedly less fatty acids as neutral lipids, and had reduced capacity for beta-oxidation. Similar to previous observations in HepG2 cells, palmitate, but not oleate, induced ER stress and actin stress fiber formation in activated stellate cells. In contrast, oleate, but not palmitate, induced the inflammatory signal TXNIP, decreased cytoskeleton proteins, and decreased cell polarity preceding cell death in activated stellate cells. CONCLUSIONS: Palmitate-induced lipotoxicity was associated with ER stress pathways in both primary activated hepatic stellate cells and epithelial hepatoma cells, whereas high oleate caused lipotoxicity only in activated stellate cells, possibly through a distinct mechanism involving disruption of cytoskeleton components. This may have implications for optimal dietary fatty acid compositions during various stages of NAFLD.


Subject(s)
Hepatic Stellate Cells/drug effects , Lipid Metabolism/drug effects , Oleic Acid/toxicity , Palmitic Acid/toxicity , Stress, Physiological/drug effects , Transcriptome , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Death/drug effects , Cell Polarity/drug effects , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Hep G2 Cells , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Lipid Metabolism/genetics , Oligonucleotide Array Sequence Analysis , Organ Specificity , Oxidation-Reduction , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Physiological/genetics
2.
PLoS One ; 10(6): e0131269, 2015.
Article in English | MEDLINE | ID: mdl-26102086

ABSTRACT

Elongation factor 1A-1 (eEF1A-1) has non-canonical functions in regulation of the actin cytoskeleton and apoptosis. It was previously identified through a promoter-trap screen as a mediator of fatty acid-induced cell death (lipotoxicity), and was found to participate in this process downstream of ER stress. Since ER stress is implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD), we investigated the mechanism of action of eEF1A-1 in hepatocyte lipotoxicity. HepG2 cells were exposed to excess fatty acids, followed by assessments of ER stress, subcellular localization of eEF1A-1, and cell death. A specific inhibitor of eEF1A-1 elongation activity, didemnin B, was used to determine whether its function in protein synthesis is involved in lipotoxicity. Within 6 h, eEF1A-1 protein was modestly induced by high palmitate, and partially re-localized from its predominant location at the ER to polymerized actin at the cell periphery. This early induction and subcellular redistribution of eEF1A-1 coincided with the onset of ER stress, and was later followed by cell death. Didemnin B did not prevent the initiation of ER stress by high palmitate, as indicated by eIF2α phosphorylation. However, consistent with sustained inhibition of eEF1A-1-dependent elongation activity, didemnin B prevented the recovery of protein synthesis and increase in GRP78 protein that are normally associated with later phases of the response to ongoing ER stress. This resulted in decreased palmitate-induced cell death. Our data implicate eEF1A-1, and its function in protein synthesis, in hepatocyte lipotoxicity.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Hepatocytes/drug effects , Palmitates/toxicity , Peptide Chain Elongation, Translational , Peptide Elongation Factor 1/physiology , Animals , Apoptosis/drug effects , Depsipeptides/pharmacology , Dietary Fats/toxicity , Dietary Sucrose/toxicity , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation/drug effects , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Hep G2 Cells , Hepatocytes/metabolism , Humans , Leptin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Animal , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...