02.10.2015 Views

studies

2015SupplementFULLTEXT

2015SupplementFULLTEXT

SHOW MORE
SHOW LESS
  • No tags were found...

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

646A AASLD ABSTRACTS HEPATOLOGY, October, 2015<br />

883<br />

Lipid-induced Endoplasmic Reticulum Stress But Not<br />

Hepatic Steatosis Contributes to Blockage of Autophagosome-Lysosome<br />

Fusion<br />

Koichiro Miyagawa, Shinji Oe, Yuichi Honma, Masaru Harada;<br />

University of Occupational and Environmental Health, Kitakyushu,<br />

Japan<br />

Purpose: Autophagy is crucial for intracellular quality control of<br />

proteins and intracellular organelles in various tissues. Several<br />

<strong>studies</strong> showed that blockage of hepatic autophagic degradation<br />

system occurred in obese, and that had an important role<br />

in the development of nonalcoholic fatty liver disease (NAFLD).<br />

However, the mechanism of this blockage has not been fully<br />

elucidated. In this study, we evaluated how lipid overload is<br />

linked to impairment of autophagy in hepatocytes. Methods:<br />

We investigated the effect of lipid overload on hepatic autophagy<br />

in cultured hepatocytes-derived cells treated with monounsaturated<br />

fatty acids (MUFAs) or saturated fatty acids (SFAs).<br />

Autophagic flux was analyzed by immunoblotting and fluorescence<br />

microscopy. We also evaluated the relationship between<br />

hepatic steatosis, endoplasmic reticulum (ER) stress, and autophagy<br />

in this model. Results: SFAs but not MUFAs induced ER<br />

stress and accumulation of autophagosomes despite of lower<br />

accumulation of lipid droplets compared with MUFAs. Microtubule-associated<br />

protein 1 light chain 3 (LC3) turnover assay<br />

demonstrated that SFAs induced the reduction of the rate of<br />

lysosomal degradation of autophagosomes without change<br />

in autophagosome synthesis. Moreover, we monitored autophagic<br />

flux in cells transfected with mRFP-GFP tandem fluorescence-tagged<br />

LC3 (tf-LC3). GFP fluorescence is quenched in<br />

the lysosome but mRFP is not, indicating that GFP and mRFP<br />

are co-localized before the fusion with lysosomes. SFAs but not<br />

MUFAs induced GFP and mRFP overlapping, indicating that<br />

SFAs induced accumulation of autophagosomes. SFAs also<br />

suppressed co-localization of mRFP and lysosome-associated<br />

membrane protein 1 (Lamp1), suggesting that SFAs-induced<br />

impairment of autophagic flux was due to blockage of autophagosome-lysosome<br />

fusion. Furthermore, we assessed lysosomal<br />

acidification. Cells treated with SFA were labeled for<br />

Lysotracker Red and then stained with Lamp1. We could not<br />

identify Lysotracker-negative Lamp1 in SFAs-treated cells as<br />

well as vehicle, suggesting that lysosomal acidification was not<br />

impaired by SFAs treatment. We also revealed that SFAs did<br />

not affect cathepsin activity. These results suggested that SFAs<br />

impaired autophagosome-lysosome fusion without change in<br />

lysosomal function. Moreover, we found that this impairment<br />

occurred in an ER stress-dependent manner. Conclusions: The<br />

disturbance of autophagic flux in NAFLD is due to the blockage<br />

of autophagosome-lysosome fusion, and that involves in the<br />

degree of ER stress but not the intracellular accumulation of<br />

lipid droplets.<br />

Disclosures:<br />

The following authors have nothing to disclose: Koichiro Miyagawa, Shinji Oe,<br />

Yuichi Honma, Masaru Harada<br />

884<br />

CHOP and the Unfolded Protein Response Regulate<br />

NF-κB Activity through IRAK2 in the Pathogenesis of<br />

Non-Alcoholic Steatohepatitis<br />

Jeffrey A. Willy 1 , James L. Stevens 1 , Howard C. Masuoka 2 , Ronald<br />

C. Wek 1 ; 1 Biochemistry and Molecular Biology, Indiana University<br />

School of Medicine, Indianapolis, IN; 2 Department of Medicine,<br />

Indiana University School of Medicine, Indianapolis, IN<br />

Free fatty acid (FFA) induction of cell death is an important feature<br />

of NASH and has been associated with disruption of the<br />

endoplasmic reticulum and activation of the Unfolded Protein<br />

Response (UPR). The mechanisms by which this stress pathway<br />

results in deleterious effects at the organ and cellular levels<br />

are not well understood. In this study, we examined the role of<br />

the UPR in NASH. The UPR features transcriptional and translational<br />

control of gene expression involved in stress remediation,<br />

including the transcription factor CHOP, which can trigger<br />

death processes during chronic endoplasmic reticulum stress.<br />

Primary hepatocytes and HepG2 cells were exposed to physiologic<br />

levels of either saturated or unsaturated FFA. We found<br />

that the saturated FFA palmitate, but not the unsaturated FFA<br />

oleate, induced hepatocyte cell death as a result of inhibition<br />

of autophagic flux in a CHOP-dependent manner. Additionally,<br />

saturated FFA exposure resulted in activation of NF-κB through<br />

expression of IRAK2, resulting in secretion of many cytokines,<br />

including TNFα and IL-8, which contribute to both hepatic cell<br />

death and inflammation. Depletion of CHOP or the RelA subunit<br />

of NF-κB in hepatocytes by shRNA alleviated both autophagy<br />

and cytokine secretion resulting in enhanced cell viability<br />

and lowered inflammatory responses during exposure to saturated<br />

FFA. We next investigated the relevance of these findings<br />

in human liver disease. We showed that the CHOP-IRAK2-<br />

NF-κB pathway is upregulated in liver biopsies from patients<br />

with NASH, in combination with inhibition of autophagic flux,<br />

as measured by accumulation of LC3b and P62. Secretion of<br />

CHOP-dependent cytokines such as TNFα and IL-8 are also<br />

increased in the sera of NASH patients. Interestingly, while<br />

UPR activation was conserved in primary rodent hepatocytes,<br />

there was no induction of IRAK2 and NF-κB in primary rodent<br />

hepatocytes following saturated FFA exposure, providing a<br />

potential explanation for the shortcomings in regards to inflammation<br />

and fibrosis in current rodent models of NASH relative<br />

to the human disease. In conclusion, we have identified a novel<br />

pathway that is upregulated in NASH patients that plays an<br />

active role in both the hepatotoxicity and inflammation at the<br />

level of the hepatocyte following excess saturated FFA. A better<br />

understanding of the mechanism by which the UPR contributes<br />

to hepatocyte toxicity during FFA exposure may provide new<br />

approaches for the diagnosis and treatment of liver disease.<br />

Disclosures:<br />

The following authors have nothing to disclose: Jeffrey A. Willy, James L. Stevens,<br />

Howard C. Masuoka, Ronald C. Wek<br />

885<br />

Deletion of G-protein-coupled bile acid receptor (Gpbar-<br />

1, Tgr5) alleviates fasting induced hepatic steatosis by<br />

altering hepatic lipid metabolism<br />

Ajay C. Donepudi, Shannon M. Boehme, John Chiang; Northeast<br />

Ohio Medical University, Rootstown, OH<br />

Introduction: Bile acids and its receptors such as farnesoid X<br />

receptor (Fxr) and G-protein-coupled bile acid receptor (Gpbar-<br />

1, Tgr5) play a major role in nutrient homeostasis. Tgr5 has<br />

been shown to regulate glucose homeostasis by regulating insulin<br />

secretion. Tgr5 is also involved in regulating basal metabolic

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!