Our long-term goal is to delineate the underlying molecular mechanisms and physiological significance of the maintenance of endoplasmic reticulum (ER) homeostasis by three key quality-control systems, ERassociated degradation (ERAD), autophagy and unfolded protein response (UPR). Although recent studies have implied a possible role of UPR in the pathogenesis of obesity and type-2 diabetes, the maintenance and physiological significance of ER homeostasis in adipocytes remains enigma. Our recent data demonstrate that Sel1L, a key adaptor protein for the E3 ligase Hrd1 in mammalian ERAD, plays a critical role in adipocytes and metabolic regulation. Adipocyte-specific Sel1L-deficient mice (Sel1L?adipo) are protected against diet-induced obesity with elevated UPR and autophagy, but uncoupled from inflammation and cell death in WAT. Moreover, our data demonstrate a critical requirement of Sel1L for the secretion of lipoprotein lipase (LPL), which may account for postprandial hypertriglyceridemia of Sel1L?adipo mice. Thus, our data point to an indispensable role of Sel1L in adipocyte function in the pathogenesis of obesity. However, underlying molecular mechanism(s) by which Sel1L affects adipocyte function and obesity remain largely unclear. We hypothesize that Sel1L regulates adipocyte function and metabolism via both Hrd1/ERAD-dependent and -independent mechanisms, and via the crosstalk among three ER qualitycontrol systems (Sel1L-Hrd1 ERAD, UPR and autophagy) in adipocytes. Using an array of adipocytespecific knockout mouse models coupled with in vitro mechanistic studies, we will determine mechanistically how Se1L regulates adipocyte function and metabolism with a particular emphasis on Hrd1/ERAD dependency in Aim 1 and on the crosstalk among three ER quality-control systems in adipocytes in Aim 2. The generation and characterization of several adipocyte-specific double knockout mouse models with various levels of ER stress will elucidate not only the functional crosstalk among key ER quality-control machineries, but also the cellular and pathological consequences of perturbed ER homeostasis in the pathogenesis of obesity and type-2 diabetes. Finally, this study may identify a novel endogenous substrate of the Sel1L-Hrd1 ERAD complex and establish a novel mechanism underlying a feedback regulatory loop between Sel1L-Hrd1 ERAD and IRE1??signaling.

Public Health Relevance

TO HUMAN HEALTH: Protein misfolding is detrimental to the cell and has been linked to the pathogenesis of several human diseases. Despite nearly a decade of effort, the role of ER homeostasis in adipocytes in the pathogenesis of obesity remains vague. Our preliminary data point to adipocyte Sel1L as a key regulator in ER homeostasis and metabolism. A successful completion of this study will have a powerful impact on our understanding of the physiological significance of ER quality-control systems and the maintenance of ER homeostasis in adipocytes in the context of diet-induced obesity.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK105393-02
Application #
9053304
Study Section
Cellular Aspects of Diabetes and Obesity Study Section (CADO)
Program Officer
Haft, Carol R
Project Start
2015-04-11
Project End
2016-07-31
Budget Start
2016-02-01
Budget End
2016-07-31
Support Year
2
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Cornell University
Department
Nutrition
Type
Sch of Home Econ/Human Ecology
DUNS #
872612445
City
Ithaca
State
NY
Country
United States
Zip Code
14850
Kim, Geun Hyang; Shi, Guojun; Somlo, Diane Rm et al. (2018) Hypothalamic ER-associated degradation regulates POMC maturation, feeding, and age-associated obesity. J Clin Invest 128:1125-1140
Hwang, Jiwon; Qi, Ling (2018) Quality Control in the Endoplasmic Reticulum: Crosstalk between ERAD and UPR pathways. Trends Biochem Sci 43:593-605
Bhattacharya, Asmita; Sun, Shengyi; Wang, Heting et al. (2018) Hepatic Sel1L-Hrd1 ER-associated degradation (ERAD) manages FGF21 levels and systemic metabolism via CREBH. EMBO J 37:
Qi, Ling; Tsai, Billy; Arvan, Peter (2017) New Insights into the Physiological Role of Endoplasmic Reticulum-Associated Degradation. Trends Cell Biol 27:430-440
Shi, Guojun; Somlo, Diane RM; Kim, Geun Hyang et al. (2017) ER-associated degradation is required for vasopressin prohormone processing and systemic water homeostasis. J Clin Invest 127:3897-3912
Dijk, Wieneke; Mattijssen, Frits; de la Rosa Rodriguez, Montserrat et al. (2017) Hypoxia-Inducible Lipid Droplet-Associated Is Not a Direct Physiological Regulator of Lipolysis in Adipose Tissue. Endocrinology 158:1231-1251
Oteng, Antwi-Boasiako; Bhattacharya, Asmita; Brodesser, Susanne et al. (2017) Feeding Angptl4-/- mice trans fat promotes foam cell formation in mesenteric lymph nodes without leading to ascites. J Lipid Res 58:1100-1113
Sun, Shengyi; Lourie, Rohan; Cohen, Sara B et al. (2016) Epithelial Sel1L is required for the maintenance of intestinal homeostasis. Mol Biol Cell 27:483-90
Ji, Yewei; Kim, Hana; Yang, Liu et al. (2016) The Sel1L-Hrd1 Endoplasmic Reticulum-Associated Degradation Complex Manages a Key Checkpoint in B Cell Development. Cell Rep 16:2630-2640
Sun, Shengyi; Shi, Guojun; Sha, Haibo et al. (2015) IRE1? is an endogenous substrate of endoplasmic-reticulum-associated degradation. Nat Cell Biol 17:1546-55

Showing the most recent 10 out of 14 publications