Diabetes Metab J.  2017 Feb;41(1):10-19. 10.4093/dmj.2017.41.1.10.

Nuclear Receptors Resolve Endoplasmic Reticulum Stress to Improve Hepatic Insulin Resistance

Affiliations
  • 1Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Korea. jaemanlee@knu.ac.kr
  • 2BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University School of Medicine, Daegu, Korea.

Abstract

Chronic endoplasmic reticulum (ER) stress culminating in proteotoxicity contributes to the development of insulin resistance and progression to type 2 diabetes mellitus. Pharmacologic interventions targeting several different nuclear receptors have emerged as potential treatments for insulin resistance. The mechanistic basis for these antidiabetic effects has primarily been attributed to multiple metabolic and inflammatory functions. Here we review recent advances in our understanding of the association of ER stress with insulin resistance and the role of nuclear receptors in promoting ER stress resolution and improving insulin resistance in the liver.

Keyword

Diabetes mellitus, type 2; Endoplasmic reticulum stress; Hepatic steatosis; Insulin resistance; Receptors, cytoplasmic and nuclear; Unfolded protein response

MeSH Terms

Diabetes Mellitus, Type 2
Endoplasmic Reticulum Stress*
Endoplasmic Reticulum*
Insulin Resistance*
Insulin*
Liver
Receptors, Cytoplasmic and Nuclear*
Unfolded Protein Response
Insulin
Receptors, Cytoplasmic and Nuclear

Figure

  • Fig. 1 Canonical endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) pathways in mammals. Homeostatic perturbations leads to the accumulation of unfolded or misfolded proteins in the ER, which causes the dissociation of binding protein/glucose-regulated protein 78 (BiP/GRP78) from the luminal domain of three ER stress sensors PKR-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1), and activating transcription factor 6 (ATF6) to exert its chaperone function. BiP/GRP78 dissociation renders PERK to undergo dimerization and transphosphorylation, and activation of its kinase activity, which then phosphorylates Ser51 on eukaryotic initiation factor 2α (eIF2α) to decrease global protein translation but selectively increases translation of ATF4 mRNA. ATF4 acting as a transcription factor induces expression of UPR target genes involved in amino acid (AA) biosynthesis, anti-oxidant response, and apoptosis. C/EBP homologous protein (CHOP) as one of direct ATF4 target genes subsequently activates expression of growth arrest and DNA damage-inducible protein (GADD34), a regulatory subunit of protein phosphatase 1 (PP1), which contributes to dephosphorylation of eIF2α to resume protein translation. ER oxidoreductin 1 (ERO1), another CHOP target gene, is an ER oxidoreductase. IRE1, the most conserved branch of UPR pathways throughout eukaryotes also undergoes dimerization (or oligomerization), transphosphorylation, and activation of endoRNase activity. This RNase function removes a 26-base intron from X-box binding protein 1 (Xbp1) mRNA to generate Xbp1s, a shorter spliced form of Xbp1 mRNA (XBP1s). XBP1s, a basic leucine zipper (bZIP) transcription factor controls a diverse range of UPR target genes including protein fold, ER-associated degradation (ERAD), trafficking, lipogenesis, inflammation, etc. This RNase activity also leads to a regulated IRE1-dependent decay (RIDD) of mRNA to reduce protein loading in the ER. Activation of ATF6 translocates from ER to Golgi apparatus with unknown processes where it is proteolytically cleaved by the Ser protease site 1 and site 2 proteases (S1P/S2P), generating ATF6(n), n-terminal truncated form of bZIP transcription factor. ATF6(n) activates the expression of its target genes involved in protein folding, ERAD, and ER biogenesis. Overall, activation of each sensor generates bZIP transcription factor ATF4, XBP1s, and ATF6(n), respectively that induces the expression of their relevant target genes associated with protein-folding fidelity, ERAD, ER biogenesis, lipogenesis, inflammation, amino acid biosynthesis, anti-oxidant response, etc. Both PERK and IRE1 contribute to reducing protein loading in the ER by suppressing global protein translation via phosphorylation of eIF2α and triggering RIDD of mRNA, respectively. Therefore, ER stress is alleviated by various feedback mechanisms from three branches of UPR pathways at the level of transcription, post-transcription or translation. However, prolonged and unmitigated ER stress induces apoptosis by ATF4-CHOP pathway.

  • Fig. 2 The role of nuclear receptor liver receptor homolog-1 (LRH-1) in endoplasmic reticulum (ER) stress resolution. In addition to the activation of three known canonical branches of unfolded protein response (UPR) pathways, LRH-1 is recruited to the promoter of polo-like kinase 3 (Plk3) gene upon ER stress. Moreover, ER stress increases transcription of Lrh-1 gene as well as its transcriptional activity probably via inositol-requiring enzyme 1 (IRE1) and/or activating transcription factor 6 (ATF6) dependent manner. PLK3, an atypical kinase phosphorylates ATF2, which induces the expression of its target genes including beta-1,3 glucuronyltransferase 3 (B3gat3), cysteine rich with EFG like domains 1 (Creld1), and multiple coagulation factor deficiency 2 (Mcfd2). Intriguingly, like liver-specific Lrh-1 knockout (KO) (Lrh-1LKO) mice, Plk3 KO mice or wild-type (WT) mice with hepatic overexpression of dominant negative ATF2 are also defective to resolve ER stress upon tunicamycin challenge. Therefore, identifying other potential ATF2 targeting genes as well as PLK3 substrates in both cytoplasm and nucleus could enrich our understanding of this unexpected but essential nuclear receptor-driven ER stress resolution pathway. BiP/GRP78, binding protein/glucose-regulated protein 78; PERK, PKR-like ER kinase; eIF2, eukaryotic initiation factor 2; XBP1s, shorter spliced form of X-box binding protein 1 (XBP1) mRNA; S1P/S2P, site 1 and site 2 proteases; ERAD, ER-associated degradation.


Reference

1. Flamment M, Hajduch E, Ferre P, Foufelle F. New insights into ER stress-induced insulin resistance. Trends Endocrinol Metab. 2012; 23:381–390.
2. Lippincott-Schwartz J, Bonifacino JS, Yuan LC, Klausner RD. Degradation from the endoplasmic reticulum: disposing of newly synthesized proteins. Cell. 1988; 54:209–220.
3. Schroder M, Kaufman RJ. The mammalian unfolded protein response. Annu Rev Biochem. 2005; 74:739–789.
4. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007; 8:519–529.
5. Ota T, Gayet C, Ginsberg HN. Inhibition of apolipoprotein B100 secretion by lipid-induced hepatic endoplasmic reticulum stress in rodents. J Clin Invest. 2008; 118:316–332.
6. Gregor MF, Hotamisligil GS. Thematic review series: adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res. 2007; 48:1905–1914.
7. Back SH, Kaufman RJ. Endoplasmic reticulum stress and type 2 diabetes. Annu Rev Biochem. 2012; 81:767–793.
8. Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011; 334:1081–1086.
9. Sha H, He Y, Yang L, Qi L. Stressed out about obesity: IRE1alpha-XBP1 in metabolic disorders. Trends Endocrinol Metab. 2011; 22:374–381.
10. Shamu CE, Walter P. Oligomerization and phosphorylation of the Ire1p kinase during intracellular signaling from the endoplasmic reticulum to the nucleus. EMBO J. 1996; 15:3028–3039.
11. Sidrauski C, Walter P. The transmembrane kinase Ire1p is a site-specific endonuclease that initiates mRNA splicing in the unfolded protein response. Cell. 1997; 90:1031–1039.
12. Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, Nagata K, Harding HP, Ron D. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004; 18:3066–3077.
13. Lee S, Kim SM, Dotimas J, Li L, Feener EP, Baldus S, Myers RB, Chutkow WA, Patwari P, Yoshioka J, Lee RT. Thioredoxin-interacting protein regulates protein disulfide isomerases and endoplasmic reticulum stress. EMBO Mol Med. 2014; 6:732–743.
14. Hollien J, Weissman JS. Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science. 2006; 313:104–107.
15. Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, Ron D. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000; 287:664–666.
16. Nishitoh H, Saitoh M, Mochida Y, Takeda K, Nakano H, Rothe M, Miyazono K, Ichijo H. ASK1 is essential for JNK/SAPK activation by TRAF2. Mol Cell. 1998; 2:389–395.
17. Marciniak SJ, Ron D. Endoplasmic reticulum stress signaling in disease. Physiol Rev. 2006; 86:1133–1149.
18. Kim JK, Kim YJ, Fillmore JJ, Chen Y, Moore I, Lee J, Yuan M, Li ZW, Karin M, Perret P, Shoelson SE, Shulman GI. Prevention of fat-induced insulin resistance by salicylate. J Clin Invest. 2001; 108:437–446.
19. Bertolotti A, Wang X, Novoa I, Jungreis R, Schlessinger K, Cho JH, West AB, Ron D. Increased sensitivity to dextran sodium sulfate colitis in IRE1beta-deficient mice. J Clin Invest. 2001; 107:585–593.
20. Tsuru A, Fujimoto N, Takahashi S, Saito M, Nakamura D, Iwano M, Iwawaki T, Kadokura H, Ron D, Kohno K. Negative feedback by IRE1beta optimizes mucin production in goblet cells. Proc Natl Acad Sci U S A. 2013; 110:2864–2869.
21. Namba T, Tanaka K, Ito Y, Ishihara T, Hoshino T, Gotoh T, Endo M, Sato K, Mizushima T. Positive role of CCAAT/enhancer-binding protein homologous protein, a transcription factor involved in the endoplasmic reticulum stress response in the development of colitis. Am J Pathol. 2009; 174:1786–1798.
22. Yoshida H, Haze K, Yanagi H, Yura T, Mori K. Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. J Biol Chem. 1998; 273:33741–33749.
23. Zhang K, Shen X, Wu J, Sakaki K, Saunders T, Rutkowski DT, Back SH, Kaufman RJ. Endoplasmic reticulum stress activates cleavage of CREBH to induce a systemic inflammatory response. Cell. 2006; 124:587–599.
24. Stirling J, O'Hare P. CREB4, a transmembrane bZip transcription factor and potential new substrate for regulation and cleavage by S1P. Mol Biol Cell. 2006; 17:413–426.
25. Murakami T, Kondo S, Ogata M, Kanemoto S, Saito A, Wanaka A, Imaizumi K. Cleavage of the membrane-bound transcription factor OASIS in response to endoplasmic reticulum stress. J Neurochem. 2006; 96:1090–1100.
26. Ma Z, Que H, Ni Y, Huang H, Liu Y, Liu T, Li X, Sun Q, Liu S. Cloning and characterization of SCIRR69: a novel transcriptional factor belonging to the CREB/ATF family. Mol Biol Rep. 2012; 39:7665–7672.
27. Ohoka N, Yoshii S, Hattori T, Onozaki K, Hayashi H. TRB3, a novel ER stress-inducible gene, is induced via ATF4-CHOP pathway and is involved in cell death. EMBO J. 2005; 24:1243–1255.
28. Gotoh T, Oyadomari S, Mori K, Mori M. Nitric oxide-induced apoptosis in RAW 264.7 macrophages is mediated by endoplasmic reticulum stress pathway involving ATF6 and CHOP. J Biol Chem. 2002; 277:12343–12350.
29. Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, Sadri N, Yun C, Popko B, Paules R, Stojdl DF, Bell JC, Hettmann T, Leiden JM, Ron D. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 2003; 11:619–633.
30. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Gorgun C, Glimcher LH, Hotamisligil GS. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004; 306:457–461.
31. Lee MW, Chanda D, Yang J, Oh H, Kim SS, Yoon YS, Hong S, Park KG, Lee IK, Choi CS, Hanson RW, Choi HS, Koo SH. Regulation of hepatic gluconeogenesis by an ER-bound transcription factor, CREBH. Cell Metab. 2010; 11:331–339.
32. Zhou Y, Lee J, Reno CM, Sun C, Park SW, Chung J, Lee J, Fisher SJ, White MF, Biddinger SB, Ozcan U. Regulation of glucose homeostasis through a XBP-1-FoxO1 interaction. Nat Med. 2011; 17:356–365.
33. Cao J, Dai DL, Yao L, Yu HH, Ning B, Zhang Q, Chen J, Cheng WH, Shen W, Yang ZX. Saturated fatty acid induction of endoplasmic reticulum stress and apoptosis in human liver cells via the PERK/ATF4/CHOP signaling pathway. Mol Cell Biochem. 2012; 364:115–129.
34. Jo H, Choe SS, Shin KC, Jang H, Lee JH, Seong JK, Back SH, Kim JB. Endoplasmic reticulum stress induces hepatic steatosis via increased expression of the hepatic very low-density lipoprotein receptor. Hepatology. 2013; 57:1366–1377.
35. Ai D, Baez JM, Jiang H, Conlon DM, Hernandez-Ono A, Frank-Kamenetsky M, Milstein S, Fitzgerald K, Murphy AJ, Woo CW, Strong A, Ginsberg HN, Tabas I, Rader DJ, Tall AR. Activation of ER stress and mTORC1 suppresses hepatic sortilin-1 levels in obese mice. J Clin Invest. 2012; 122:1677–1687.
36. Lee JN, Ye J. Proteolytic activation of sterol regulatory element-binding protein induced by cellular stress through depletion of Insig-1. J Biol Chem. 2004; 279:45257–45265.
37. Ariyama Y, Shimizu H, Satoh T, Tsuchiya T, Okada S, Oyadomari S, Mori M, Mori M. Chop-deficient mice showed increased adiposity but no glucose intolerance. Obesity (Silver Spring). 2007; 15:1647–1656.
38. Maris M, Overbergh L, Gysemans C, Waget A, Cardozo AK, Verdrengh E, Cunha JP, Gotoh T, Cnop M, Eizirik DL, Burcelin R, Mathieu C. Deletion of C/EBP homologous protein (Chop) in C57Bl/6 mice dissociates obesity from insulin resistance. Diabetologia. 2012; 55:1167–1178.
39. Zhang W, Hietakangas V, Wee S, Lim SC, Gunaratne J, Cohen SM. ER stress potentiates insulin resistance through PERK-mediated FOXO phosphorylation. Genes Dev. 2013; 27:441–449.
40. Jiang S, Yan C, Fang QC, Shao ML, Zhang YL, Liu Y, Deng YP, Shan B, Liu JQ, Li HT, Yang L, Zhou J, Dai Z, Liu Y, Jia WP. Fibroblast growth factor 21 is regulated by the IRE1alpha-XBP1 branch of the unfolded protein response and counteracts endoplasmic reticulum stress-induced hepatic steatosis. J Biol Chem. 2014; 289:29751–29765.
41. Kim KH, Jeong YT, Oh H, Kim SH, Cho JM, Kim YN, Kim SS, Kim DH, Hur KY, Kim HK, Ko T, Han J, Kim HL, Kim J, Back SH, Komatsu M, Chen H, Chan DC, Konishi M, Itoh N, Choi CS, Lee MS. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat Med. 2013; 19:83–92.
42. Fayard E, Auwerx J, Schoonjans K. LRH-1: an orphan nuclear receptor involved in development, metabolism and steroidogenesis. Trends Cell Biol. 2004; 14:250–260.
43. Fernandez-Marcos PJ, Auwerx J, Schoonjans K. Emerging actions of the nuclear receptor LRH-1 in the gut. Biochim Biophys Acta. 2011; 1812:947–955.
44. Lee JM, Lee YK, Mamrosh JL, Busby SA, Griffin PR, Pathak MC, Ortlund EA, Moore DD. A nuclear-receptor-dependent phosphatidylcholine pathway with antidiabetic effects. Nature. 2011; 474:506–510.
45. Mamrosh JL, Lee JM, Wagner M, Stambrook PJ, Whitby RJ, Sifers RN, Wu SP, Tsai MJ, Demayo FJ, Moore DD. Nuclear receptor LRH-1/NR5A2 is required and targetable for liver endoplasmic reticulum stress resolution. Elife. 2014; 3:e01694.
46. Pilkis SJ, Granner DK. Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis. Annu Rev Physiol. 1992; 54:885–909.
47. Barthel A, Schmoll D. Novel concepts in insulin regulation of hepatic gluconeogenesis. Am J Physiol Endocrinol Metab. 2003; 285:E685–E692.
48. Kim DK, Ryu D, Koh M, Lee MW, Lim D, Kim MJ, Kim YH, Cho WJ, Lee CH, Park SB, Koo SH, Choi HS. Orphan nuclear receptor estrogen-related receptor gamma (ERRgamma) is key regulator of hepatic gluconeogenesis. J Biol Chem. 2012; 287:21628–21639.
49. Misra J, Kim DK, Choi W, Koo SH, Lee CH, Back SH, Kaufman RJ, Choi HS. Transcriptional cross talk between orphan nuclear receptor ERRgamma and transmembrane transcription factor ATF6alpha coordinates endoplasmic reticulum stress response. Nucleic Acids Res. 2013; 41:6960–6974.
50. Misra J, Chanda D, Kim DK, Cho SR, Koo SH, Lee CH, Back SH, Choi HS. Orphan nuclear receptor Errgamma induces C-reactive protein gene expression through induction of ER-bound Bzip transmembrane transcription factor CREBH. PLoS One. 2014; 9:e86342.
51. Kim DK, Gang GT, Ryu D, Koh M, Kim YN, Kim SS, Park J, Kim YH, Sim T, Lee IK, Choi CS, Park SB, Lee CH, Koo SH, Choi HS. Inverse agonist of nuclear receptor ERRgamma mediates antidiabetic effect through inhibition of hepatic gluconeogenesis. Diabetes. 2013; 62:3093–3102.
52. Xiong X, Wang X, Lu Y, Wang E, Zhang Z, Yang J, Zhang H, Li X. Hepatic steatosis exacerbated by endoplasmic reticulum stress-mediated downregulation of FXR in aging mice. J Hepatol. 2014; 60:847–854.
53. Su Q, Baker C, Christian P, Naples M, Tong X, Zhang K, Santha M, Adeli K. Hepatic mitochondrial and ER stress induced by defective PPARalpha signaling in the pathogenesis of hepatic steatosis. Am J Physiol Endocrinol Metab. 2014; 306:E1264–E1273.
54. Chan SM, Sun RQ, Zeng XY, Choong ZH, Wang H, Watt MJ, Ye JM. Activation of PPARalpha ameliorates hepatic insulin resistance and steatosis in high fructose-fed mice despite increased endoplasmic reticulum stress. Diabetes. 2013; 62:2095–2105.
55. Klein J, Ott V, Schutt M, Klein HH. Recurrent hypoglycaemic episodes in a patient with type 2 diabetes under fibrate therapy. J Diabetes Complications. 2002; 16:246–248.
56. Flory JH, Ellenberg S, Szapary PO, Strom BL, Hennessy S. Antidiabetic action of bezafibrate in a large observational database. Diabetes Care. 2009; 32:547–551.
57. Yoshiuchi K, Kaneto H, Matsuoka TA, Kasami R, Kohno K, Iwawaki T, Nakatani Y, Yamasaki Y, Shimomura I, Matsuhisa M. Pioglitazone reduces ER stress in the liver: direct monitoring of in vivo ER stress using ER stress-activated indicator transgenic mice. Endocr J. 2009; 56:1103–1111.
58. Park MH, Park JY, Lee HJ, Kim DH, Park D, Jeong HO, Park CH, Chun P, Moon HR, Chung HY. Potent anti-diabetic effects of MHY908, a newly synthesized PPAR alpha/gamma dual agonist in db/db mice. PLoS One. 2013; 8:e78815.
59. Han KL, Choi JS, Lee JY, Song J, Joe MK, Jung MH, Hwang JK. Therapeutic potential of peroxisome proliferators: activated receptor-alpha/gamma dual agonist with alleviation of endoplasmic reticulum stress for the treatment of diabetes. Diabetes. 2008; 57:737–745.
60. Rong X, Albert CJ, Hong C, Duerr MA, Chamberlain BT, Tarling EJ, Ito A, Gao J, Wang B, Edwards PA, Jung ME, Ford DA, Tontonoz P. LXRs regulate ER stress and inflammation through dynamic modulation of membrane phospholipid composition. Cell Metab. 2013; 18:685–697.
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr