Z. M. Younossi, A. B. Koenig, D. Abdelatif, Y. Fazel, L. Henry et al., Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes, Hepatology, vol.64, pp.73-84, 2016.

M. Machado, P. Marques-vidal, C. , and H. , Hepatic histology in obese patients undergoing bariatric surgery, J Hepatol, vol.45, pp.600-606, 2006.

T. Hardy, F. Oakley, Q. M. Anstee, and C. P. Day, Nonalcoholic Fatty Liver Disease: Pathogenesis and Disease Spectrum, Annu Rev Pathol, vol.11, pp.451-496, 2016.

M. J. Pagliassotti, Endoplasmic reticulum stress in nonalcoholic fatty liver disease, Annu Rev Nutr, vol.32, pp.17-33, 2012.

V. Tsutsumi, T. Nakamura, T. Ueno, T. Torimura, and J. Aguirre-garcía, Structure and Ultrastructure of the Normal and Diseased Liver, pp.23-44, 2017.

M. Wang and R. J. Kaufman, Protein misfolding in the endoplasmic reticulum as a conduit to human disease, Nature, vol.529, pp.326-335, 2016.

P. Walter and D. Ron, The Unfolded Protein Response: From Stress Pathway to Homeostatic Regulation, Science, vol.334, p.1081, 2011.

J. Groenendyk, Z. Peng, E. Dudek, X. Fan, M. J. Mizianty et al., Interplay between the oxidoreductase PDIA6 and microRNA-322 controls the response to disrupted endoplasmic reticulum calcium homeostasis, Sci Signal, vol.7, p.54, 2014.

D. Eletto, D. Eletto, D. Dersh, T. Gidalevitz, and Y. Argon, Protein disulfide isomerase A6 controls the decay of IRE1alpha signaling via disulfide-dependent association, Mol Cell, vol.53, pp.562-576, 2014.

A. Higa, S. Taouji, S. Lhomond, D. Jensen, M. E. Fernandez-zapico et al.,

, Endoplasmic Reticulum Stress-Activated Transcription Factor ATF6? Requires the Disulfide Isomerase PDIA5 To Modulate Chemoresistance, Molecular and Cellular Biology, vol.34, pp.1839-1849, 2014.

W. Tirasophon, A. A. Welihinda, and R. J. Kaufman, A stress response pathway from the endoplasmic reticulum to the nucleus requires a novel bifunctional protein kinase/endoribonuclease (Ire1p) in mammalian cells, Genes Dev, vol.12, pp.1812-1824, 1998.

K. P. Lee, M. Dey, D. Neculai, C. Cao, T. E. Dever et al., Structure of the dual enzyme Ire1 reveals the basis for catalysis and regulation in nonconventional RNA splicing, Cell, vol.132, pp.89-100, 2008.

D. Sepulveda, D. Rojas-rivera, D. A. Rodriguez, J. Groenendyk, A. Kohler et al.,

, Interactome Screening Identifies the ER Luminal Chaperone Hsp47 as a Regulator of the Unfolded Protein Response Transducer IRE1alpha, Mol Cell, vol.69, pp.238-252, 2018.

G. E. Karagoz, D. Acosta-alvear, H. T. Nguyen, C. P. Lee, F. Chu et al., An unfolded proteininduced conformational switch activates mammalian IRE1, Elife, vol.6, 2017.

J. Jurkin, T. Henkel, A. F. Nielsen, M. Minnich, J. Popow et al., The mammalian tRNA ligase complex mediates splicing of XBP1 mRNA and controls antibody secretion in plasma cells, EMBO J, vol.33, pp.2922-2936, 2014.

C. Hetz, F. Martinon, D. Rodriguez, and L. H. Glimcher, The unfolded protein response: integrating stress signals through the stress sensor IRE1alpha, Physiol Rev, vol.91, pp.1219-1243, 2011.

A. M. Reimold, A. Etkin, I. Clauss, A. Perkins, D. S. Friend et al., An essential role in liver development for transcription factor XBP-1, Genes & Development, vol.14, pp.152-157, 2000.

K. Zhang, H. N. Wong, B. Song, C. N. Miller, D. Scheuner et al., The unfolded protein response sensor IRE1? is required at 2 distinct steps in B cell lymphopoiesis, Journal of Clinical Investigation, vol.115, pp.268-281, 2005.

Y. Ma and L. M. Hendershot, Delineation of a negative feedback regulatory loop that controls protein translation during endoplasmic reticulum stress, J Biol Chem, vol.278, pp.34864-34873, 2003.

S. B. Cullinan and J. A. Diehl, PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress, J Biol Chem, vol.279, pp.20108-20117, 2004.

Y. Adachi, K. Yamamoto, T. Okada, H. Yoshida, A. Harada et al., ATF6 Is a Transcription Factor Specializing in the Regulation of Quality Control Proteins in the Endoplasmic Reticulum, Cell Structure and Function, vol.33, pp.75-89, 2008.

J. Wu, D. T. Rutkowski, M. Dubois, J. Swathirajan, T. Saunders et al., ATF6alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress, Dev Cell, vol.13, pp.351-364, 2007.

K. Haze, H. Yoshida, H. Yanagi, T. Yura, and K. Mori, Mammalian Transcription Factor ATF6 Is Synthesized as a Transmembrane Protein and Activated by Proteolysis in Response to Endoplasmic Reticulum Stress, Molecular Biology of the Cell, vol.10, pp.3787-3799, 1999.

S. Nadanaka, H. Yoshida, F. Kano, M. Murata, and K. Mori, Activation of Mammalian Unfolded Protein Response Is Compatible with the Quality Control System Operating in the Endoplasmic Reticulum, Molecular Biology of the Cell, vol.15, pp.2537-2548, 2004.

J. Shen, X. Chen, L. Hendershot, and R. Prywes, ER Stress Regulation of ATF6 Localization by Dissociation of BiP/GRP78 Binding and Unmasking of Golgi Localization Signals, Developmental Cell, vol.3, pp.99-111, 2002.

H. Yoshida, T. Matsui, A. Yamamoto, T. Okada, and K. Mori, XBP1 mRNA Is Induced by ATF6 and Spliced by IRE1 in Response to ER Stress to Produce a Highly Active Transcription Factor, Cell, vol.107, pp.881-891, 2001.

H. Yoshida, T. Okada, K. Haze, H. Yanagi, T. Yura et al., ATF6 Activated by Proteolysis Binds in the Presence of NF-Y (CBF) Directly to the cis-Acting Element Responsible for the Mammalian Unfolded Protein Response, Molecular and Cellular Biology, vol.20, pp.6755-6767, 2000.

D. T. Rutkowski, J. Wu, S. H. Back, M. U. Callaghan, S. P. Ferris et al., UPR pathways combine to prevent hepatic steatosis caused by ER stress-mediated suppression of transcriptional master regulators, Dev Cell, vol.15, pp.829-840, 2008.

K. Yamamoto, K. Takahara, S. Oyadomari, T. Okada, T. Sato et al., Induction of Liver Steatosis and Lipid Droplet Formation in ATF6?-Knockout Mice Burdened with Pharmacological Endoplasmic Reticulum Stress, Molecular Biology of the Cell, vol.21, pp.2975-2986, 2010.

C. Hetz, E. Chevet, and S. A. Oakes, Proteostasis control by the unfolded protein response, Nat Cell Biol, vol.17, pp.829-838, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01175531

H. Urra, E. Dufey, F. Lisbona, D. Rojas-rivera, and C. Hetz, When ER stress reaches a dead end

, Biochim Biophys Acta, vol.1833, pp.3507-3517, 2013.

Y. Deng, Z. V. Wang, C. Tao, N. Gao, W. L. Holland et al., The Xbp1s/GalE axis links ER stress to postprandial hepatic metabolism, J Clin Invest, vol.123, pp.455-468, 2013.

K. Y. Hur, J. S. So, V. Ruda, M. Frank-kamenetsky, K. Fitzgerald et al., IRE1alpha activation protects mice against acetaminophen-induced hepatotoxicity, J Exp Med, vol.209, pp.307-318, 2012.

J. Wang, Y. Qiu, Z. Yang, H. Kim, Q. Qian et al., IRE1? prevents hepatic steatosis by processing and promoting the degradation of select microRNAs, Science Signaling, vol.11, 2018.

U. Ozcan, Q. Cao, E. Yilmaz, A. H. Lee, N. N. Iwakoshi et al., Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes, Science, vol.306, pp.457-461, 2004.

M. F. Gregor, L. Yang, E. Fabbrini, B. S. Mohammed, J. C. Eagon et al., Endoplasmic reticulum stress is reduced in tissues of obese subjects after weight loss, Diabetes, vol.58, pp.693-700, 2009.

P. Puri, F. Mirshahi, O. Cheung, R. Natarajan, J. W. Maher et al., Activation and dysregulation of the unfolded protein response in nonalcoholic fatty liver disease, Gastroenterology, vol.134, pp.568-576, 2008.

J. A. Willy, S. K. Young, A. L. Mosley, S. Gawrieh, J. L. Stevens et al., Function of inhibitor of Bruton's tyrosine kinase isoform alpha (IBTKalpha) in nonalcoholic steatohepatitis links autophagy and the unfolded protein response, J Biol Chem, vol.292, pp.14050-14065, 2017.

S. Lee, S. Kim, S. Hwang, N. J. Cherrington, and D. Ryu, Dysregulated expression of proteins associated with ER stress, autophagy and apoptosis in tissues from nonalcoholic fatty liver disease, Oncotarget, vol.8, pp.63370-63381, 2017.

A. D. Lake, P. Novak, R. N. Hardwick, B. Flores-keown, F. Zhao et al., The adaptive endoplasmic reticulum stress response to lipotoxicity in progressive human nonalcoholic fatty liver disease, Toxicol Sci, vol.137, pp.26-35, 2014.

K. Yoshiuchi, H. Kaneto, T. A. Matsuoka, K. Kohno, T. Iwawaki et al., Direct monitoring of in vivo ER stress during the development of insulin resistance with ER stress-activated indicator transgenic mice, Biochem Biophys Res Commun, vol.366, pp.545-550, 2008.

H. L. Kammoun, H. Chabanon, I. Hainault, S. Luquet, C. Magnan et al., GRP78 expression inhibits insulin and ER stress-induced SREBP-1c activation and reduces hepatic steatosis in mice, J Clin Invest, vol.119, pp.1201-1215, 2009.

Y. Nakatani, H. Kaneto, D. Kawamori, K. Yoshiuchi, M. Hatazaki et al., Involvement of endoplasmic reticulum stress in insulin resistance and diabetes, J Biol Chem, vol.280, pp.847-851, 2005.

K. L. Donnelly, C. I. Smith, S. J. Schwarzenberg, J. Jessurun, M. D. Boldt et al., Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease, J Clin Invest, vol.115, pp.1343-1351, 2005.

S. Wang, Z. Chen, V. Lam, J. Han, J. Hassler et al., IRE1alpha-XBP1s induces PDI expression to increase MTP activity for hepatic VLDL assembly and lipid homeostasis, Cell Metab, vol.16, pp.473-486, 2012.

K. Zhang, S. Wang, J. Malhotra, J. R. Hassler, S. H. Back et al., The unfolded protein response transducer IRE1alpha prevents ER stress-induced hepatic steatosis, EMBO J, vol.30, pp.1357-1375, 2011.

A. Lee, E. F. Scapa, D. E. Cohen, and L. H. Glimcher, Regulation of hepatic lipogenesis by the transcription factor XBP1, Science, vol.320, pp.1492-1496, 2008.

J. S. So, K. Y. Hur, M. Tarrio, V. Ruda, M. Frank-kamenetsky et al., Silencing of lipid metabolism genes through IRE1alpha-mediated mRNA decay lowers plasma lipids in mice, Cell Metab, vol.16, pp.487-499, 2012.

S. Oyadomari, H. P. Harding, Y. Zhang, M. Oyadomari, and R. D. , Dephosphorylation of translation initiation factor 2alpha enhances glucose tolerance and attenuates hepatosteatosis in mice, Cell Metab, vol.7, pp.520-532, 2008.

G. Xiao, T. Zhang, S. Yu, S. Lee, V. Calabuig-navarro et al., ATF4 protein deficiency protects against high fructose-induced hypertriglyceridemia in mice, J Biol Chem, vol.288, pp.25350-25361, 2013.

J. Seo, E. S. Fortuno, J. M. Suh, D. Stenesen, W. Tang et al., Atf4 Regulates Obesity, Glucose Homeostasis, and Energy Expenditure, Diabetes, vol.58, pp.2565-2573, 2009.

J. Villeneuve, S. Lepreux, A. Mulot, A. M. Berard, A. Higa-nishiyama et al., A protective role for CD154 in hepatic steatosis in mice, Hepatology, vol.52, pp.1968-1979, 2010.

D. Dezwaan-mccabe, R. D. Sheldon, M. C. Gorecki, D. F. Guo, E. R. Gansemer et al., ER Stress Inhibits Liver Fatty Acid Oxidation while Unmitigated Stress Leads to AnorexiaInduced Lipolysis and Both Liver and Kidney Steatosis, Cell Rep, vol.19, pp.1794-1806, 2017.

X. Chen, F. Zhang, Q. Gong, A. Cui, S. Zhuo et al., Hepatic ATF6 Increases Fatty Acid Oxidation to Attenuate Hepatic Steatosis in Mice Through Peroxisome Proliferator-Activated Receptor ?, Diabetes, vol.65, 1904.

L. Zeng, M. Lu, K. Mori, S. Luo, A. S. Lee et al., ATF6 modulates SREBP2-mediated lipogenesis, The EMBO Journal, vol.23, pp.950-958, 2004.

M. F. Gregor and G. S. Hotamisligil, Inflammatory mechanisms in obesity, Annu Rev Immunol, vol.29, pp.415-445, 2011.

S. Hummasti and G. S. Hotamisligil, Endoplasmic reticulum stress and inflammation in obesity and diabetes, Circ Res, vol.107, pp.579-591, 2010.

U. Özcan, E. Yilmaz, L. Özcan, M. Furuhashi, E. Vaillancourt et al., Chemical Chaperones Reduce ER Stress and Restore Glucose Homeostasis in a Mouse Model of Type 2

, Diabetes. Science, vol.313, pp.1137-1140, 2006.

M. J. Jurczak, A. H. Lee, F. R. Jornayvaz, H. Y. Lee, A. L. Birkenfeld et al., Dissociation of inositol-requiring enzyme (IRE1alpha)-mediated c-Jun N-terminal kinase activation from hepatic insulin resistance in conditional X-box-binding protein-1 (XBP1) knock-out mice, J Biol Chem, vol.287, pp.2558-2567, 2012.

N. Kumashiro, D. M. Erion, D. Zhang, M. Kahn, S. A. Beddow et al., Cellular mechanism of insulin resistance in nonalcoholic fatty liver disease, Proceedings of the National Academy of Sciences, vol.108, pp.16381-16385, 2011.

Y. Wang, L. Vera, W. H. Fischer, and M. Montminy, The CREB coactivator CRTC2 links hepatic ER stress and fasting gluconeogenesis, Nature, vol.460, pp.534-537, 2009.

L. Ozcan, D. S. Ghorpade, Z. Zheng, J. C. De-souza, K. Chen et al., Hepatocyte DACH1 Is Increased in Obesity via Nuclear Exclusion of HDAC4 and Promotes Hepatic Insulin Resistance, Cell Rep, vol.15, pp.2214-2225, 2016.

L. Yang, P. Li, S. Fu, E. S. Calay, and G. S. Hotamisligil, Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance, Cell Metab, vol.11, pp.467-478, 2010.

Y. Adkins, I. W. Schie, D. Fedor, A. Reddy, S. Nguyen et al., A novel mouse model of nonalcoholic steatohepatitis with significant insulin resistance, Laboratory Investigation, vol.93, p.1313, 2013.

R. A. Sinha, S. You, J. Zhou, M. M. Siddique, B. Bay et al., Impaired autophagic flux is associated with increased endoplasmic reticulum stress during the development of NAFLD, The Journal of Clinical Investigation, vol.122, p.1179, 2012.

K. Castillo, D. Rojas-rivera, F. Lisbona, B. Caballero, M. Nassif et al., BAX inhibitor-1 regulates autophagy by controlling the IRE1alpha branch of the unfolded protein response

D. L. Gorden, D. S. Myers, P. T. Ivanova, E. Fahy, M. R. Maurya et al., Biomarkers of NAFLD progression: a lipidomics approach to an epidemic, J Lipid Res, vol.30, pp.722-736, 2011.

M. E. Miquilena-colina, E. Lima-cabello, S. Sanchez-campos, M. V. Garcia-mediavilla, M. Fernandezbermejo et al., Hepatic fatty acid translocase CD36 upregulation is associated with insulin resistance, hyperinsulinaemia and increased steatosis in nonalcoholic steatohepatitis and chronic hepatitis C, Gut, vol.60, pp.1394-1402, 2011.

R. V. Farese, R. Zechner, C. B. Newgard, and T. C. Walther, The problem of establishing relationships between hepatic steatosis and hepatic insulin resistance, Cell Metab, vol.15, pp.570-573, 2012.

C. A. Nagle, E. L. Klett, and R. A. Coleman, Hepatic triacylglycerol accumulation and insulin resistance, J Lipid Res, vol.50, pp.74-79, 2009.

J. Capeau, Insulin resistance and steatosis in humans, Diabetes & Metabolism, vol.34, pp.649-657, 2008.

S. Fu, L. Yang, P. Li, O. Hofmann, L. Dicker et al., Aberrant lipid metabolism disrupts calcium homeostasis causing liver endoplasmic reticulum stress in obesity, Nature, vol.473, pp.528-531, 2011.

A. P. Arruda, B. M. Pers, G. Parlakgul, E. Guney, K. Inouye et al., Chronic enrichment of hepatic endoplasmic reticulum-mitochondria contact leads to mitochondrial dysfunction in obesity, Nat Med, vol.20, pp.1427-1435, 2014.

R. E. Patterson, S. Kalavalapalli, C. M. Williams, M. Nautiyal, J. T. Mathew et al., Lipotoxicity in steatohepatitis occurs despite an increase in tricarboxylic acid cycle activity, Am J Physiol Endocrinol Metab, vol.310, pp.484-494, 2016.

P. Iozzo, M. Bucci, A. Roivainen, K. Nagren, M. J. Jarvisalo et al., Fatty acid metabolism in the liver, measured by positron emission tomography, is increased in obese individuals, Gastroenterology, vol.139, pp.841-846, 2010.

N. E. Sunny, E. J. Parks, J. D. Browning, and S. C. Burgess, Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease, Cell Metab, vol.14, pp.804-810, 2011.

L. Hager, L. Li, H. Pun, L. Liu, M. A. Hossain et al., Lecithin:cholesterol acyltransferase deficiency protects against cholesterol-induced hepatic endoplasmic reticulum stress in mice, J Biol Chem, vol.287, pp.20755-20768, 2012.

M. F. Gregor and G. S. Hotamisligil, Adipocyte stress: the endoplasmic reticulum and metabolic disease, J Lipid Res, vol.48, pp.1905-1914, 2007.

T. Promlek, Y. Ishiwata-kimata, M. Shido, M. Sakuramoto, K. Kohno et al., Membrane aberrancy and unfolded proteins activate the endoplasmic reticulum stress sensor Ire1 in different ways, Molecular Biology of the Cell, vol.22, pp.3520-3532, 2011.

R. Volmer, K. Van-der-ploeg, and R. D. , Membrane lipid saturation activates endoplasmic reticulum unfolded protein response transducers through their transmembrane domains, Proceedings of the National Academy of Sciences, vol.110, pp.4628-4633, 2013.

S. Fu, S. M. Watkins, and G. S. Hotamisligil, The role of endoplasmic reticulum in hepatic lipid homeostasis and stress signaling, Cell Metab, vol.15, pp.623-634, 2012.

K. Halbleib, K. Pesek, R. Covino, H. F. Hofbauer, D. Wunnicke et al., Activation of the Unfolded Protein Response by Lipid Bilayer Stress, Mol Cell, vol.67, pp.673-684, 2017.

S. Patouraux, D. Rousseau, S. Bonnafous, C. Lebeaupin, C. Luci et al., CD44 is a key player in non-alcoholic steatohepatitis, J Hepatol, vol.67, pp.328-338, 2017.

A. E. Feldstein, A. Canbay, P. Angulo, M. Taniai, L. J. Burgart et al., Hepatocyte apoptosis and fas expression are prominent features of human nonalcoholic steatohepatitis, Gastroenterology, vol.125, pp.437-443, 2003.

J. Deng, P. D. Lu, Y. Zhang, D. Scheuner, R. J. Kaufman et al., Translational repression mediates activation of nuclear factor kappa B by phosphorylated translation initiation factor 2, Mol Cell Biol, vol.24, pp.10161-10168, 2004.

H. Y. Jiang, S. A. Wek, B. C. Mcgrath, D. Scheuner, R. J. Kaufman et al., Phosphorylation of the Subunit of Eukaryotic Initiation Factor 2 Is Required for Activation of NF-B in Response to Diverse Cellular Stresses, Molecular and Cellular Biology, vol.23, pp.5651-5663, 2003.

H. Yamazaki, N. Hiramatsu, K. Hayakawa, Y. Tagawa, M. Okamura et al., Activation of the Akt-NF-kappaB pathway by subtilase cytotoxin through the ATF6 branch of the unfolded protein response, J Immunol, vol.183, pp.1480-1487, 2009.

T. Luedde and R. F. Schwabe, NF-kappaB in the liver-linking injury, fibrosis and hepatocellular carcinoma, Nat Rev Gastroenterol Hepatol, vol.8, pp.108-118, 2011.

D. Pena, A. Leclercq, I. Field, J. George, J. Jones et al., NF-kappaB activation, rather than TNF, mediates hepatic inflammation in a murine dietary model of steatohepatitis, Gastroenterology, vol.129, pp.1663-1674, 2005.

J. A. Willy, S. K. Young, J. L. Stevens, H. C. Masuoka, and R. C. Wek, CHOP links endoplasmic reticulum stress to NF-?B activation in the pathogenesis of nonalcoholic steatohepatitis, Molecular Biology of the Cell, vol.26, pp.2190-2204, 2015.

D. Browning-jeffrey, S. Lidia, S. Dobbins, R. Nuremberg, P. et al., Prevalence of hepatic steatosis in an urban population in the United States: Impact of ethnicity, Hepatology, vol.40, pp.1387-1395, 2004.

A. P. Rolo, J. S. Teodoro, and C. M. Palmeira, Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis, Free Radical Biology and Medicine, vol.52, pp.59-69, 2012.

G. S. , F. B. , R. T. , T. R. , D. Ra et al., Alterations of hepatic ATP homeostasis and respiratory chain during development of non-alcoholic steatohepatitis in a rodent model, European Journal of Clinical Investigation, vol.38, pp.245-252, 2008.

G. Serviddio, F. Bellanti, R. Tamborra, T. Rollo, N. Capitanio et al., Uncoupling protein-2 (UCP2) induces mitochondrial proton leak and increases susceptibility of nonalcoholic steatohepatitis (NASH) liver to ischaemia-reperfusion injury, Gut, vol.57, pp.957-965, 2008.

G. P. Mannaerts, L. J. Debeer, J. Thomas, D. Schepper, and P. J. , Mitochondrial and peroxisomal fatty acid oxidation in liver homogenates and isolated hepatocytes from control and clofibrate-treated rats, Journal of Biological Chemistry, vol.254, pp.4585-4595, 1979.

E. G. Hrycay and S. M. Bandiera, Involvement of Cytochrome P450 in Reactive Oxygen Species Formation and Cancer, Adv Pharmacol, vol.74, pp.35-84, 2015.

D. De-craemer and M. Pauwels, Van den Branden C. Alterations of peroxisomes in steatosis of the human liver: a quantitative study, Hepatology, vol.22, pp.744-752, 1995.

N. Sathish, K. , E. Chundamannil, E. , P. Anna et al., Oxidative stress in experimental liver microvesicular steatosis: Role of mitochondria and peroxisomes, Journal of Gastroenterology and Hepatology, vol.21, pp.1240-1249, 2006.

J. W. Zmijewski, S. Banerjee, H. Bae, A. Friggeri, E. R. Lazarowski et al., Exposure to Hydrogen Peroxide Induces Oxidation and Activation of AMP-activated Protein Kinase, Journal of Biological Chemistry, vol.285, pp.33154-33164, 2010.

K. Begriche, A. Igoudjil, D. Pessayre, and B. Fromenty, Mitochondrial dysfunction in NASH: Causes, consequences and possible means to prevent it, Mitochondrion, vol.6, pp.1-28, 2006.

P. Puri, B. Rebecca, A. , W. Michelle, M. Mirshahi et al., A lipidomic analysis of nonalcoholic fatty liver disease, Hepatology, vol.46, pp.1081-1090, 2007.

F. Caballero, A. Fernández, D. Lacy, A. M. Fernández-checa, J. C. Caballería et al., Enhanced free cholesterol, SREBP-2 and StAR expression in human NASH, Journal of Hepatology, vol.50, pp.789-796, 2009.

S. B. Cullinan and J. A. Diehl, Coordination of ER and oxidative stress signaling: the PERK/Nrf2 signaling pathway, Int J Biochem Cell Biol, vol.38, pp.317-332, 2006.

K. Okada, E. Warabi, H. Sugimoto, M. Horie, K. Tokushige et al., Nrf2 inhibits hepatic iron accumulation and counteracts oxidative stress-induced liver injury in nutritional steatohepatitis, J Gastroenterol, vol.47, pp.924-935, 2012.

R. Shimozono, Y. Asaoka, Y. Yoshizawa, T. Aoki, H. Noda et al., Nrf2 activators attenuate the progression of nonalcoholic steatohepatitis-related fibrosis in a dietary rat model, Mol Pharmacol, vol.84, pp.62-70, 2013.

Q. Ma, Role of nrf2 in oxidative stress and toxicity, Annu Rev Pharmacol Toxicol, vol.53, pp.401-426, 2013.

R. Zhou, A. Tardivel, B. Thorens, I. Choi, and J. Tschopp, Thioredoxin-interacting protein links oxidative stress to inflammasome activation, Nat Immunol, vol.11, pp.136-140, 2010.

A. G. Lerner, J. P. Upton, P. V. Praveen, R. Ghosh, Y. Nakagawa et al., IRE1alpha induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress, Cell Metab, vol.16, pp.250-264, 2012.

C. M. Oslowski, T. Hara, B. O'sullivan-murphy, K. Kanekura, S. Lu et al.,

, Thioredoxin-interacting protein mediates ER stress-induced beta cell death through initiation of the inflammasome, Cell Metab, vol.16, pp.265-273, 2012.

K. Zhang, X. Shen, J. Wu, K. Sakaki, T. Saunders et al., Endoplasmic reticulum stress activates cleavage of CREBH to induce a systemic inflammatory response, Cell, vol.124, pp.587-599, 2006.

M. Yoneda, H. Mawatari, K. Fujita, H. Iida, K. Yonemitsu et al., High-sensitivity C-reactive protein is an independent clinical feature of nonalcoholic steatohepatitis (NASH) and also of the severity of fibrosis in NASH, J Gastroenterol, vol.42, pp.573-582, 2007.

R. Anty, S. Bekri, N. Luciani, M. C. Saint-paul, M. Dahman et al., The inflammatory C-reactive protein is increased in both liver and adipose tissue in severely obese patients independently from metabolic syndrome, Type 2 diabetes, and NASH, Am J Gastroenterol, vol.101, pp.1824-1833, 2006.

C. Vecchi, G. Montosi, K. Zhang, I. Lamberti, S. A. Duncan et al., ER Stress Controls Iron Metabolism Through Induction of Hepcidin, Science, vol.325, p.877, 2009.

K. V. Kowdley, P. Belt, L. A. Wilson, M. M. Yeh, B. A. Neuschwander-tetri et al., Serum ferritin is an independent predictor of histologic severity and advanced fibrosis in patients with nonalcoholic fatty liver disease, Hepatology, vol.55, pp.77-85, 2012.

T. C. Tan, D. H. Crawford, L. A. Jaskowski, V. N. Subramaniam, A. D. Clouston et al., Excess iron modulates endoplasmic reticulum stress-associated pathways in a mouse model of alcohol and high-fat diet-induced liver injury, Lab Invest, vol.93, pp.1295-1312, 2013.

T. Csak, M. Ganz, J. Pespisa, K. Kodys, A. Dolganiuc et al., Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells, Hepatology, vol.54, pp.133-144, 2011.

C. Lebeaupin, E. Proics, C. H. De-bieville, D. Rousseau, S. Bonnafous et al., ER stress induces NLRP3 inflammasome activation and hepatocyte death, Cell Death Dis, vol.6, p.1879, 2015.

G. Szabo and T. Csak, Inflammasomes in liver diseases, J Hepatol, vol.57, pp.642-654, 2012.

A. Wree, A. Eguchi, M. D. Mcgeough, C. A. Pena, C. D. Johnson et al., NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice, Hepatology, vol.59, pp.898-910, 2014.

J. Zhang, K. Zhang, Z. Li, and B. Guo, ER Stress-induced Inflammasome Activation Contributes to Hepatic Inflammation and Steatosis, J Clin Cell Immunol, vol.7, 2016.

J. M. Schattenberg, R. Singh, Y. Wang, J. H. Lefkowitch, R. M. Rigoli et al., JNK1 but not JNK2 promotes the development of steatohepatitis in mice, Hepatology, vol.43, pp.163-172, 2006.

I. Tavares-de-almeida, H. Cortez-pinto, G. Fidalgo, D. Rodrigues, and M. E. Camilo, Plasma total and free fatty acids composition in human non-alcoholic steatohepatitis, Clinical Nutrition, vol.21, pp.219-223, 2002.

K. Yamaguchi, L. Yang, S. Mccall, J. Huang, X. X. Yu et al., Inhibiting triglyceride synthesis improves hepatic steatosis but exacerbates liver damage and fibrosis in obese mice with nonalcoholic steatohepatitis, Hepatology, vol.45, pp.1366-1374, 2007.

P. Hirsova, S. H. Ibrabim, G. J. Gores, and H. Malhi, Lipotoxic lethal and sublethal stress signaling in hepatocytes: relevance to NASH pathogenesis, Journal of Lipid Research, vol.57, pp.1758-1770, 2016.

D. Wang, Y. Wei, and M. J. Pagliassotti, Saturated fatty acids promote endoplasmic reticulum stress and liver injury in rats with hepatic steatosis, Endocrinology, vol.147, pp.943-951, 2006.

K. T. Pfaffenbach, C. L. Gentile, A. M. Nivala, D. Wang, Y. Wei et al., Linking endoplasmic reticulum stress to cell death in hepatocytes: roles of C/EBP homologous protein and chemical chaperones in palmitate-mediated cell death, Am J Physiol Endocrinol Metab, vol.298, pp.1027-1035, 2010.

S. C. Cazanave, J. L. Mott, S. F. Bronk, N. W. Werneburg, C. D. Fingas et al., Death receptor 5 signaling promotes hepatocyte lipoapoptosis, J Biol Chem, vol.286, pp.39336-39348, 2011.

Y. Akazawa, S. Cazanave, J. L. Mott, N. Elmi, S. F. Bronk et al., Palmitoleate attenuates palmitate-induced bim and PUMA up-regulation and hepatocyte lipoapoptosis, Journal of hepatology, vol.52, pp.586-593, 2010.

J. E. Chipuk and D. R. Green, How do BCL-2 proteins induce mitochondrial outer membrane permeabilization?, Trends Cell Biol, vol.18, pp.157-164, 2008.

A. Deniaud, O. Sharaf-el-dein, E. Maillier, D. Poncet, G. Kroemer et al., Endoplasmic reticulum stress induces calcium-dependent permeability transition, mitochondrial outer membrane permeabilization and apoptosis, Oncogene, vol.27, pp.285-299, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00321149

G. Campos, W. Schmidt-heck, A. Ghallab, K. Rochlitz, L. Pütter et al., The transcription factor CHOP, a central component of the transcriptional regulatory network induced upon CCl4 intoxication in mouse liver, is not a critical mediator of hepatotoxicity, Archives of Toxicology, vol.88, pp.1267-1280, 2014.

J. Rieusset, Endoplasmic reticulum-mitochondria calcium signaling in hepatic metabolic diseases, Biochim Biophys Acta, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01608518

S. M. Walenbergh, T. Houben, S. S. Rensen, V. Bieghs, T. Hendrikx et al., Plasma cathepsin D correlates with histological classifications of fatty liver disease in adults and responds to intervention, Sci Rep, vol.6, p.38278, 2016.

H. B. El-serag and K. L. Rudolph, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis, Gastroenterology, vol.132, pp.2557-2576, 2007.

S. Mittal, F. Kanwal, J. Ying, R. Chung, Y. H. Sada et al., Effectiveness of surveillance for hepatocellular carcinoma in clinical practice: A United States cohort, J Hepatol, vol.65, pp.1148-1154, 2016.

Y. Vandewynckel, D. Laukens, E. Bogaerts, A. Paridaens, A. Van-den-bussche et al.,

C. Steenkiste, Modulation of the unfolded protein response impedes tumor cell adaptation to proteotoxic stress: a PERK for hepatocellular carcinoma therapy, Hepatology International, vol.9, pp.93-104, 2015.

C. Guichard, G. Amaddeo, S. Imbeaud, Y. Ladeiro, L. Pelletier et al., Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma, Nat Genet, vol.44, pp.694-698, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00719917

C. Greenman, P. Stephens, R. Smith, G. L. Dalgliesh, C. Hunter et al., Patterns of somatic mutation in human cancer genomes, Nature, vol.446, pp.153-158, 2007.

M. Shuda, N. Kondoh, N. Imazeki, K. Tanaka, T. Okada et al., Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis, Journal of Hepatology, vol.38, pp.605-614, 2003.

T. Kishimoto, K. Kokura, N. Ohkawa, Y. Makino, M. Yoshida et al., Enhanced expression of a new class of liver-enriched b-Zip transcription factors, hepatocarcinogenesis-related transcription factor, in hepatocellular carcinomas of rats and humans, Cell Growth Differentiation, vol.9, pp.337-344, 1998.

M. T. Spiotto, A. Banh, I. Papandreou, H. Cao, M. G. Galvez et al., Imaging the unfolded protein response in primary tumors reveals microenvironments with metabolic variations that predict tumor growth, Cancer Res, vol.70, pp.78-88, 2010.

G. Xu, K. Liu, J. Anderson, K. Patrene, S. Lentzsch et al., Expression of XBP1s in bone marrow stromal cells is critical for myeloma cell growth and osteoclast formation, Blood, vol.119, pp.4205-4214, 2012.

M. Maurel, E. Chevet, J. Tavernier, and S. Gerlo, Getting RIDD of RNA: IRE1 in cell fate regulation, Trends in Biochemical Sciences, vol.39, pp.245-254, 2014.

C. Atorrasagasti, M. Malvicini, J. B. Aquino, L. Alaniz, M. Garcia et al., Overexpression of SPARC obliterates the in vivo tumorigenicity of human hepatocellular carcinoma cells, Int J Cancer, vol.126, pp.2726-2740, 2010.

J. D. Blais, C. L. Addison, R. Edge, T. Falls, H. Zhao et al., Perk-dependent translational regulation promotes tumor cell adaptation and angiogenesis in response to hypoxic stress, Mol Cell Biol, vol.26, pp.9517-9532, 2006.

M. Arai, N. Kondoh, N. Imazeki, A. Hada, K. Hatsuse et al., Transformation-associated gene regulation by ATF6alpha during hepatocarcinogenesis, FEBS Lett, vol.580, pp.184-190, 2006.

D. M. Schewe and J. A. Aguirre-ghiso, ATF6alpha-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo, Proc Natl Acad Sci U S A, vol.105, pp.10519-10524, 2008.

N. Chignard, S. Shang, H. Wang, J. Marrero, C. Brechot et al., Cleavage of endoplasmic reticulum proteins in hepatocellular carcinoma: Detection of generated fragments in patient sera, Gastroenterology, vol.130, pp.2010-2022, 2006.

B. Luo and A. S. Lee, The critical roles of endoplasmic reticulum chaperones and unfolded protein response in tumorigenesis and anticancer therapies, Oncogene, vol.32, pp.805-818, 2013.

Q. Shao, P. Ren, Y. Li, B. Peng, L. Dai et al., Autoantibodies against glucose-regulated protein 78 as serological diagnostic biomarkers in hepatocellular carcinoma, Int J Oncol, vol.41, pp.1061-1067, 2012.

D. Dezwaan-mccabe, J. D. Riordan, A. M. Arensdorf, M. S. Icardi, A. J. Dupuy et al., The stress-regulated transcription factor CHOP promotes hepatic inflammatory gene expression, fibrosis, and oncogenesis, PLoS Genet, vol.9, p.1003937, 2013.

V. Scaiewicz, A. Nahmias, R. T. Chung, T. Mueller, B. Tirosh et al., CCAAT/enhancer-binding protein homologous (CHOP) protein promotes carcinogenesis in the DEN-induced hepatocellular carcinoma model, PLoS One, vol.8, p.81065, 2013.

K. Toriguchi, E. Hatano, K. Tanabe, K. Takemoto, K. Nakamura et al., Attenuation of steatohepatitis, fibrosis, and carcinogenesis in mice fed a methionine-choline deficient diet by CCAAT/enhancer-binding protein homologous protein deficiency, J Gastroenterol Hepatol, vol.29, pp.1109-1118, 2014.

H. Nakagawa, A. Umemura, K. Taniguchi, J. Font-burgada, D. Dhar et al., ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development, Cancer Cell, vol.26, pp.331-343, 2014.

S. Zelber-sagi, J. Godos, and F. Salomone, Lifestyle changes for the treatment of nonalcoholic fatty liver disease: a review of observational studies and intervention trials, Ther Adv Gastroenterol, vol.9, pp.392-407, 2016.

D. V. Monaco-ferreira and L. Va, Weight Regain 10 Years After Roux-en-Y Gastric Bypass, Obes Surg, 2016.

M. A. Van-herck, L. Vonghia, and S. M. Francque, Animal Models of Nonalcoholic Fatty Liver Disease-A Starter's Guide, Nutrients, vol.9, 2017.

A. S. Henkel, A. M. Dewey, K. A. Anderson, S. Olivares, and R. M. Green, Reducing endoplasmic reticulum stress does not improve steatohepatitis in mice fed a methionine-and choline-deficient diet

, American Journal of Physiology-Gastrointestinal and Liver Physiology, vol.303, pp.54-59, 2012.

A. Bashiri, D. Nesan, G. Tavallaee, I. Sue-chue-lam, K. Chien et al., Cellular cholesterol accumulation modulates high fat high sucrose (HFHS) diet-induced ER stress and hepatic inflammasome activation in the development of non-alcoholic steatohepatitis, Biochim Biophys Acta, vol.1861, pp.594-605, 2016.

M. Kandel-kfir, T. Almog, A. Shaish, G. Shlomai, L. Anafi et al., Interleukin1alpha deficiency attenuates endoplasmic reticulum stress-induced liver damage and CHOP expression in mice, J Hepatol, vol.63, pp.926-933, 2015.

M. A. Abdelmegeed, Y. Choi, G. Godlewski, S. Ha, A. Banerjee et al., Cytochrome P450-2E1 promotes fast food-mediated hepatic fibrosis, Scientific Reports, vol.7, p.39764, 2017.

A. R. Mridha, A. Wree, A. Robertson, M. M. Yeh, C. D. Johnson et al., NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice, J Hepatol, vol.66, pp.1037-1046, 2017.

V. Legry, D. M. Van-rooyen, B. Lambert, C. Sempoux, L. Poekes et al., Endoplasmic reticulum stress does not contribute to steatohepatitis in obese and insulinresistant high-fat-diet-fed foz/foz mice, Clin Sci (Lond), vol.127, pp.507-518, 2014.

A. Asgharpour, S. C. Cazanave, T. Pacana, M. Seneshaw, R. Vincent et al., A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer, J Hepatol, vol.65, pp.579-588, 2016.

A. Asgharpour, S. C. Cazanave, T. Pacana, M. Seneshaw, R. Vincent et al., A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer, Journal of Hepatology, vol.65, pp.579-588, 2016.

M. Fujii, Y. Shibazaki, K. Wakamatsu, Y. Honda, Y. Kawauchi et al., A murine model for non-alcoholic steatohepatitis showing evidence of association between diabetes and hepatocellular carcinoma, Med Mol Morphol, vol.46, pp.141-152, 2013.

K. Saito, T. Uebanso, K. Maekawa, M. Ishikawa, R. Taguchi et al., Characterization of hepatic lipid profiles in a mouse model with nonalcoholic steatohepatitis and subsequent fibrosis, Scientific Reports, vol.5, p.12466, 2015.

E. J. Carey, A. H. Ali, and K. D. Lindor, Primary biliary cirrhosis, The Lancet, vol.386, pp.1565-1575, 2015.

B. Mosbah, I. , A. Martel, C. Zaouali, M. A. Bintanel-morcillo et al., Endoplasmic reticulum stress inhibition protects steatotic and non-steatotic livers in partial hepatectomy under ischemia-reperfusion, Cell Death Dis, vol.1, p.52, 2010.

C. Hetz, E. Chevet, and H. P. Harding, Targeting the unfolded protein response in disease, Nat Rev Drug Discov, vol.12, pp.703-719, 2013.

O. Tufanli, T. Akillilar, P. Acosta-alvear, D. Kocaturk, B. Onat et al., Targeting IRE1 with small molecules counteracts progression of atherosclerosis, Proc Natl Acad Sci U S A, vol.114, pp.1395-1404, 2017.

S. Kim, Y. Joe, H. J. Kim, Y. S. Kim, S. O. Jeong et al., Endoplasmic reticulum stressinduced IRE1alpha activation mediates cross-talk of GSK-3beta and XBP-1 to regulate inflammatory cytokine production, J Immunol, vol.194, pp.4498-4506, 2015.

N. Mimura, M. Fulciniti, G. Gorgun, Y. T. Tai, D. Cirstea et al., Blockade of XBP1 splicing by inhibition of IRE1alpha is a promising therapeutic option in multiple myeloma, Blood, vol.119, pp.5772-5781, 2012.

L. Wang, B. G. Perera, S. B. Hari, B. Bhhatarai, B. J. Backes et al., Divergent allosteric control of the IRE1alpha endoribonuclease using kinase inhibitors, Nat Chem Biol, vol.8, pp.982-989, 2012.

R. Ghosh, L. Wang, E. S. Wang, B. G. Perera, A. Igbaria et al., Allosteric inhibition of the IRE1alpha RNase preserves cell viability and function during endoplasmic reticulum stress, Cell, vol.158, pp.534-548, 2014.

S. Morita, S. A. Villalta, H. C. Feldman, A. C. Register, W. Rosenthal et al., Targeting ABL-IRE1alpha Signaling Spares ER-Stressed Pancreatic beta Cells to Reverse Autoimmune Diabetes, Cell Metab, vol.25, pp.883-897, 2017.

R. Loomba, V. Seguritan, W. Li, T. Long, N. Klitgord et al., Gut Microbiome-Based Metagenomic Signature for Non-invasive Detection of Advanced Fibrosis in Human Nonalcoholic Fatty Liver Disease, Cell Metab, vol.25, pp.1054-1062, 2017.

P. Tsaytler, H. P. Harding, R. D. Bertolotti, and A. , Selective Inhibition of a Regulatory Subunit of Protein Phosphatase 1 Restores Proteostasis, Science, vol.332, p.91, 2011.

C. Sidrauski, A. M. Mcgeachy, N. T. Ingolia, and P. Walter, The small molecule ISRIB reverses the effects of eIF2alpha phosphorylation on translation and stress granule assembly, Elife, vol.4, 2015.

C. M. Gallagher, C. Garri, E. L. Cain, K. K. Ang, C. G. Wilson et al., Ceapins are a new class of unfolded protein response inhibitors, selectively targeting the ATF6alpha branch, Elife, vol.5, 2016.

E. Dufey, D. Sepulveda, D. Rojas-rivera, and C. Hetz, Cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. 1. An overview, Am J Physiol Cell Physiol, vol.307, pp.582-594, 2014.

V. Valenzuela, K. L. Jackson, S. P. Sardi, and C. Hetz, Gene Therapy Strategies to Restore ER Proteostasis in Disease, Molecular Therapy, 2018.

J. A. Gomez and D. T. Rutkowski, Experimental reconstitution of chronic ER stress in the liver reveals feedback suppression of BiP mRNA expression, Elife, vol.5, 2016.

G. H. Lee, K. J. Oh, H. R. Kim, H. S. Han, H. Y. Lee et al., Effect of BI-1 on insulin resistance through regulation of CYP2E1, Sci Rep, vol.6, p.32229, 2016.

H. Y. Lee, G. H. Lee, K. R. Bhattarai, B. H. Park, S. H. Koo et al., Bax Inhibitor-1 regulates hepatic lipid accumulation via ApoB secretion, Sci Rep, vol.6, p.27799, 2016.

Y. Zhou, J. Lee, C. M. Reno, C. Sun, S. W. Park et al., Regulation of glucose homeostasis through a XBP-1-FoxO1 interaction, Nat Med, vol.17, pp.356-365, 2011.

H. Nakagawa, Y. Hirata, K. Takeda, Y. Hayakawa, T. Sato et al., Apoptosis signal-regulating kinase 1 inhibits hepatocarcinogenesis by controlling the tumor-suppressing function of stress-activated mitogen-activated protein kinase, Hepatology, vol.54, pp.185-195, 2011.

C. Jiang, L. Wen, C. Yin, W. Xu, B. Shi et al., Apoptosis signal-regulating kinase 1 mediates the inhibitory effect of hepatocyte nuclear factor-4? on hepatocellular carcinoma, Oncotarget, vol.7, pp.27408-27421, 2016.

J. M. Axten, S. P. Romeril, A. Shu, J. Ralph, J. R. Medina et al., Discovery of GSK2656157: An Optimized PERK Inhibitor Selected for Preclinical Development, ACS Med Chem Lett, vol.4, pp.964-968, 2013.

B. Bailly-maitre, B. F. Belgardt, S. D. Jordan, B. Coornaert, M. J. Von-freyend et al., Hepatic Bax inhibitor-1 inhibits IRE1alpha and protects from obesity-associated insulin resistance and glucose intolerance, J Biol Chem, vol.285, pp.6198-6207, 2010.

I. Ben-mosbah, I. Alfany-fernandez, C. Martel, M. A. Zaouali, M. Bintanel-morcillo et al., Endoplasmic reticulum stress inhibition protects steatotic and non-steatotic livers in partial hepatectomy under ischemiareperfusion, Cell death & disease, vol.1, p.52, 2010.

K. F. Chen, W. T. Tai, T. H. Liu, H. P. Huang, Y. C. Lin et al., Sorafenib overcomes TRAIL resistance of hepatocellular carcinoma cells through the inhibition of STAT3, Clin Cancer Res, vol.16, pp.5189-5199, 2010.

C. M. Gallagher, C. Garri, E. L. Cain, K. K. Ang, C. G. Wilson et al., Ceapins are a new class of unfolded protein response inhibitors, selectively targeting the ATF6alpha branch, 2016.

R. Ghosh, L. Wang, E. S. Wang, B. G. Perera, A. Igbaria et al., Allosteric inhibition of the IRE1alpha RNase preserves cell viability and function during endoplasmic reticulum stress, Cell, vol.158, pp.534-548, 2014.

J. A. Gomez and D. T. Rutkowski, Experimental reconstitution of chronic ER stress in the liver reveals feedback suppression of BiP mRNA expression, 2016.

H. L. Kammoun, H. Chabanon, I. Hainault, S. Luquet, C. Magnan et al., GRP78 expression inhibits insulin and ER stress-induced SREBP-1c activation and reduces hepatic steatosis in mice, J Clin Invest, vol.119, pp.1201-1215, 2009.

S. Kim, Y. Joe, H. J. Kim, Y. S. Kim, S. O. Jeong et al., Endoplasmic reticulum stress-induced IRE1alpha activation mediates cross-talk of GSK-3beta and XBP-1 to regulate inflammatory cytokine production, J Immunol, vol.194, pp.4498-4506, 2015.

A. V. Korennykh, P. F. Egea, A. A. Korostelev, J. Finer-moore, C. Zhang et al., The unfolded protein response signals through high-order assembly of Ire1, Nature, vol.457, pp.687-693, 2009.

C. Lebeaupin, E. Proics, C. H. De-bieville, D. Rousseau, S. Bonnafous et al., ER stress induces NLRP3 inflammasome activation and hepatocyte death, vol.6, p.1879, 2015.

C. Lebeaupin, D. Vallée, D. Rousseau, S. Patouraux, S. Bonnafous et al., Bax Inhibitor-1 protects from Non-Alcoholic Steatohepatitis by limiting IRE1? signaling, 2018.

H. Y. Lee, G. H. Lee, K. R. Bhattarai, B. H. Park, S. H. Koo et al., Bax Inhibitor-1 regulates hepatic lipid accumulation via ApoB secretion, Sci Rep, vol.6, p.27799, 2016.

A. G. Lerner, J. P. Upton, P. V. Praveen, R. Ghosh, Y. Nakagawa et al., , 2012.

, IRE1alpha induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress, Cell Metab, vol.16, pp.250-264

R. Loomba, E. Lawitz, P. S. Mantry, S. Jayakumar, S. H. Caldwell et al., The ASK1 Inhibitor Selonsertib in Patients with Nonalcoholic Steatohepatitis: A Randomized, Phase 2 Trial. Hepatology, 2017.

N. Mimura, M. Fulciniti, G. Gorgun, Y. T. Tai, D. Cirstea et al., Blockade of XBP1 splicing by inhibition of IRE1alpha is a promising therapeutic option in multiple myeloma, Blood, vol.119, pp.5772-5781, 2012.

A. J. Moreira, R. Ordonez, C. T. Cerski, J. N. Picada, A. Garcia-palomo et al., Melatonin Activates Endoplasmic Reticulum Stress and Apoptosis in Rats with Diethylnitrosamine-Induced Hepatocarcinogenesis, vol.10, 2015.

S. Morita, S. A. Villalta, H. C. Feldman, A. C. Register, W. Rosenthal et al., Targeting ABL-IRE1alpha Signaling Spares ER-Stressed Pancreatic beta Cells to Reverse Autoimmune Diabetes, Cell Metab, vol.25, pp.883-897, 2017.

H. Nakagawa, Y. Hirata, K. Takeda, Y. Hayakawa, T. Sato et al., , 2011.

, Apoptosis signal-regulating kinase 1 inhibits hepatocarcinogenesis by controlling the tumor-suppressing function of stress-activated mitogen-activated protein kinase, Hepatology, vol.54, pp.185-195

H. Nakagawa, A. Umemura, K. Taniguchi, J. Font-burgada, D. Dhar et al., ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development, Cancer Cell, vol.26, pp.331-343, 2014.

U. Özcan, E. Yilmaz, L. Özcan, M. Furuhashi, E. Vaillancourt et al., Chemical Chaperones Reduce ER Stress and Restore Glucose Homeostasis in a Mouse Model of Type 2 Diabetes, Science, vol.313, pp.1137-1140, 2006.

C. Rojas, B. Pan-castillo, C. Valls, G. Pujadas, S. Garcia-vallve et al., Resveratrol enhances palmitate-induced ER stress and apoptosis in cancer cells, PLoS One, vol.9, p.113929, 2014.

C. Sidrauski, A. M. Mcgeachy, N. T. Ingolia, and P. Walter, The small molecule ISRIB reverses the effects of eIF2alpha phosphorylation on translation and stress granule assembly, 2015.

P. Tsaytler, H. P. Harding, D. Ron, and A. Bertolotti, Selective Inhibition of a Regulatory Subunit of Protein Phosphatase 1 Restores Proteostasis, Science, vol.332, p.91, 2011.

O. Tufanli, P. Telkoparan-akillilar, D. Acosta-alvear, B. Kocaturk, U. I. Onat et al., , 2017.

, Targeting IRE1 with small molecules counteracts progression of atherosclerosis, Proc Natl Acad Sci U S A, vol.114, pp.1395-1404

Y. Vandewynckel, D. Laukens, E. Bogaerts, A. Paridaens, A. Van-den-bussche et al., Modulation of the unfolded protein response impedes tumor cell adaptation to proteotoxic stress: a PERK for hepatocellular carcinoma therapy, Hepatology International, vol.9, pp.93-104, 2015.

L. Wang, B. G. Perera, S. B. Hari, B. Bhhatarai, B. J. Backes et al., Divergent allosteric control of the IRE1alpha endoribonuclease using kinase inhibitors, Nat Chem Biol, vol.8, pp.982-989, 2012.

J. K. Won, S. J. Yu, C. Y. Hwang, S. H. Cho, S. M. Park et al., Protein disulfide isomerase inhibition synergistically enhances the efficacy of sorafenib for hepatocellular carcinoma, Hepatology, vol.66, pp.855-868, 2017.

S. J. Yu, J. H. Yoon, J. I. Yang, E. J. Cho, M. S. Kwak et al., Enhancement of hexokinase II inhibitor-induced apoptosis in hepatocellular carcinoma cells via augmenting ER stress and anti-angiogenesis by protein disulfide isomerase inhibition, J Bioenerg Biomembr, vol.44, pp.101-115, 2012.

H. Zhang, K. Li, Y. Lin, F. Xing, X. Xiao et al., Targeting VCP enhances anticancer activity of oncolytic virus M1 in hepatocellular carcinoma, Science Translational Medicine, vol.9, 2017.

Y. Zhou, J. Lee, C. M. Reno, C. Sun, S. W. Park et al., Regulation of glucose homeostasis through a XBP-1-FoxO1 interaction, Nat Med, vol.17, pp.356-365, 2011.