scholarly journals Saturated Fatty Acid-Induced Cytotoxicity in Liver Cells Does Not Involve Phosphatase and Tensin Homologue Deleted on Chromosome 10

2013 ◽  
Vol 2013 ◽  
pp. 1-8 ◽  
Author(s):  
Dong Wang ◽  
Yuren Wei ◽  
Melinda Frye ◽  
Christopher L. Gentile ◽  
Michael J. Pagliassotti

Liver specific deletion of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN) induces steatosis and hypersensitivity to insulin. Saturated fatty acids, which induce endoplasmic reticulum stress and cell death, appear to increase PTEN, whereas unsaturated fatty acids which do not induce endoplasmic reticulum stress or cell death reduce this protein. In the present study, the role of PTEN in saturated fatty acid-induced cytotoxicity was examined in H4IIE and HepG2 liver cells. Palmitate and stearate increased the expression of PTEN, whereas the unsaturated fatty acids, oleate and linoleate, reduced PTEN expression in both cell types. SiRNA-mediated knockdown of PTEN did not increase liver cell triglyceride stores or reduce palmitate- or stearate-mediated ER stress or apoptosis. These results suggest that PTEN does not play a significant role in saturated fatty acid-induced cytotoxicity in these liver cell models and in the absence of insulin.

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi169-vi169
Author(s):  
Kevin Murnan ◽  
Serena Tommasini-Ghelfi ◽  
Lisa Hurley ◽  
Corey Dussold ◽  
Daniel Wahl ◽  
...  

Abstract Increased de novo synthesis, mobilization and uptake of fatty acids are required to provide sufficient lipids for membrane biogenesis in support of rapid tumor cell division and growth. In addition to their structural roles as components of the plasma membrane, fatty acid-derived lipids regulate ferroptotic cell death, a type of programmed cell death, when oxidized by iron-dependent lipoxygenase enzymes. De novo lipogenesis and the defense against oxidative lipid damage require large amounts of cytosolic NADPH. Our group has recently found that HGG up-regulate wild-type Isocitrate dehydrogenase 1 (referred to hereafter as ‘wt-IDH1high HGG’) to generate large quantities of cytosolic NADPH. RNAi-mediated knockdown of wt-IDH1, alone and in combination with radiation therapy (RT), slows the growth of patient-derived HGG xenografts, while overexpression of wt-IDH1 promotes intracranial HGG growth. Isotope tracer and liquid chromatography-based lipidomic studies indicated that wt-IDH1 supports the de novo biosynthesis of mono-unsaturated fatty acids (MUFAs) and promotes the incorporation of monounsaturated phospholipids into the plasma membrane, while displacing polyunsaturated fatty acid (PUFA) phospholipids. In addition, enhanced NADPH production in wt-IDH1high HGG increases glutathione (GSH) level, reduces reactive oxygen species (ROS), activates the phospholipid peroxidase glutathione peroxidase 4 (GPX4)-driven lipid repair pathway, and dampens the accumulation of PUFA-containing lipid peroxides, known executioners of ferroptosis. To pharmacologically target wt-IDH1,we have used and characterized wt-IDH1i-13, a first-in-class competitive α,β-unsaturated enone (AbbVie). wt-IDH1i-13 potently inhibits wt-IDH1 enzymatic activity, by covalently binding to the NADP+ binding pocket. Our data indicate that wt-IDH1i-13 promotes ferroptosis, which can be rescued by pre-treatment of cells with the peroxyl scavenger and ferroptosis inhibitor ferrostatin. wt-IDH1i-13 is brain-penetrant, and similar to genetic ablation, reduces progression and extends the survival of wt-IDH1high HGG bearing mice, alone and in combination with RT. These studies credential to wt-IDH1i-13 as a novel therapeutic modality for the treatment of wt-IDH1 gliomas.


2020 ◽  
Vol 245 (14) ◽  
pp. 1268-1279
Author(s):  
Jun Wang ◽  
Yingli Chen ◽  
Qing Song ◽  
Alexandra Griffiths ◽  
Zhenyuan Song

Lipotoxicity, defined as the cell death and/or cellular dysfunction induced by ectopic lipid deposition, plays a pathological role in the development of many metabolic diseases. Although endoplasmic reticulum stress is a well-documented mechanism behind, how endoplasmic reticulum stress is initiated during lipotoxicity remains obscure. In this study, using palmitate exposure (a 16-C saturated fatty acid) of AML12 hepatocytes, a non-transformed murine hepatocyte cell line, as an experimental model, we identified mammalian target of rapamycin complex 1 (mTORC1) to be a critical contributor to palmitate-elicited lipotoxicity, manifested by incremental triglycerides secretion and cell death. Unlike oleate (an 18-C monounsaturated fatty acid), palmitate strongly induced mTORC1 activation in hepatocytes. Importantly, mTOR inhibitors, torin-1, and rapamycin prevented hepatocytes from palmitate-induced triglyceride overproduction and cell death. We further showed that the intracellular metabolism of palmitate is required for its stimulatory effect on mTORC1. Whereas the inhibition of long-chain acyl-CoA synthetase, which converts palmitate to palmitoyl-CoA, attenuated mTORC1 activation and protected against cell death, the inhibition of stearoyl-CoA desaturase-1, the enzyme desaturating palmitate to palmitoleate, strengthened mTORC1 activation and aggravated triglyceride overproduction and cell death. Our further investigations revealed that the palmitate-induced mTORC1 activation was required for its endoplasmic reticulum stress-inducing action as mTORC1 inhibition ablated palmitate-induced activation of IRE1α, one of the three canonical pathways activated during unfolded protein response. Finally, our data demonstrated that IRE1α inhibition ameliorated triglyceride overproduction and cell death in response to palmitate exposure. Collectively, our data identify that mTORC1-IRE1α pathway is coordinately implicated in the development of lipotoxicity in hepatocyte. Impact statement Lipotoxicity induced by saturated fatty acids (SFA) plays a pivotal role in the pathogenesis of a variety of obesity-related metabolic disorders; however, the exact mechanism(s) underlying lipotoxicity development remains elusive. The liver plays a central role in regulating intrahepatic and circulatory lipid homeostasis. In the current study, we identified that mammalian target of rapamycin complex 1 (mTORC1) activation plays an important role in regulating the detrimental effects of SFA palmitate in hepatocytes, in specific cell death, and TG overproduction. Furthermore, our data confirmed that palmitate-induced mTORC1 activation is attributable to its stimulatory effect on IRE1α, one of three canonical pathways activated during ER stress. Importantly, IRE1α inhibition prevented palmitate-triggered cell death and TG overproduction, suggesting mTORC1-IRE1α pathway is mechanistically implicated in palmitate lipotoxicity. The data obtained in the current investigation support future study to explore the therapeutic potential of targeting the mTORC1-IRE1α pathway as a novel clinical strategy for the treatment of metabolic disorders involving lipotoxicity.


Sign in / Sign up

Export Citation Format

Share Document