Endocrinol Metab.  2020 Dec;35(4):943-953. 10.3803/EnM.2020.768.

Clusterin Protects Lipotoxicity-Induced Apoptosis via Upregulation of Autophagy in Insulin-Secreting Cells

Affiliations
  • 1Institute of Medical Research, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
  • 2Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea

Abstract

Background
There is a great need to discover factors that could protect pancreatic β-cells from apoptosis and thus prevent diabetes mellitus. Clusterin (CLU), a chaperone protein, plays an important role in cell protection in numerous cells and is involved in various cellular mechanisms, including autophagy. In the present study, we investigated the protective role of CLU through autophagy regulation in pancreatic β-cells.
Methods
To identify the protective role of CLU, mouse insulinoma 6 (MIN6) cells were incubated with CLU and/or free fatty acid (FFA) palmitate, and cellular apoptosis and autophagy were examined.
Results
Treatment with CLU remarkably upregulated microtubule-associated protein 1-light chain 3 (LC3)-II conversion in a doseand time-dependent manner with a significant increase in the autophagy-related 3 (Atg3) gene expression level, which is a mediator of LC3-II conversion. Moreover, co-immunoprecipitation and fluorescence microscopy experiments showed that the molecular interaction of LC3 with Atg3 and p62 was markedly increased by CLU. Stimulation of LC3-II conversion by CLU persisted in lipotoxic conditions, and FFA-induced apoptosis and dysfunction were simultaneously improved by CLU treatment. Finally, inhibition of LC3-II conversion by Atg3 gene knockdown markedly attenuated the cytoprotective effect of CLU.
Conclusion
Taken together, these findings suggest that CLU protects pancreatic β-cells against lipotoxicity-induced apoptosis via autophagy stimulation mediated by facilitating LC3-II conversion. Thus, CLU has therapeutic effects on FFA-induced pancreatic β-cell dysfunction.

Keyword

Clusterin; Autophagy; LC3-II conversion; Autophagy related protein 3; Insulin-secreting cells

Figure

  • Fig. 1 Clusterin (CLU) facilitates light chain 3 (LC3)-II conversion in mouse insulinoma 6 (MIN6) cells in a dose- and time-dependent manner. (A) MIN6 cells in one group were exposed to CLU for 24 hours at concentrations of 0, 1, 10, and 100 μg/L. MIN6 cells in another group were incubated with 10 μg/L CLU for 0, 6, 24, and 48 hours. LC3-I and -II were detected by Western blotting, and the level of LC3-II was normalized to LC3-I and β-actin. All values are expressed as mean±standard error of the mean (n=4–6). (B) LC3 in MIN6 cells was detected by immunolabeling after exposure to 10 μg/L CLU or vehicle (VEH) for 24 hours. The nuclei were visualized by 4′,6-diamidino-2-phenylindole (DAPI). Scale bars=20 μm. aP<0.01 and bP<0.001 compared to 0 μg/L treated cells; cP<0.001 and dP<0.05 compared to CLU-exposed cells for 0 hour.

  • Fig. 2 Clusterin (CLU) remarkably increases light chain 3 (LC3) binding with autophagy-related 3 (Atg3) and p62. Mouse insulinoma 6 (MIN6) cells were treated with 10 μg/L CLU for 24 hours, and protein levels were analyzed from whole lysates. (A) Levels of Atg3, Atg5, and Atg7 were detected by Western blotting and normalized to β-actin. (B, C) Morphological association of LC3 with Atg3 or p62 was analyzed by double immunolabeling using fluorescein isothiocyanate (FITC) and tetramethylrhodamine (TRITC), and co-localized areas are indicated by arrowheads. The nuclei were visualized by 4′,6-diamidino-2-phenylindole (DAPI). Scale bars=20 μm. (D) The number of double-positive cells was counted, and the results are expressed as mean±standard error of the mean per 100 DAPI-positive cells (n=3 in each treatment group). (E) Protein lysates were prepared for co-immunoprecipitation (IP) using LC3. Immunoblotting showed the co-precipitation of Atg3 and p62 with LC3. Rabbit immunoglobulin G (IgG) was used as a negative control. aP<0.001 and bP<0.05 compared to vehicle (VEH).

  • Fig. 3 Clusterin (CLU) protects pancreatic β-cells against lipotoxicity-induced apoptosis. Mouse insulinoma 6 (MIN6) cells were incubated with 0.5 mM palmitate (PA) in the presence or absence of 10 μg/L CLU for 24 hours. (A, B) Autophagy-inducing or -related proteins, including light chain 3 (LC3), autophagy-related 3 (Atg3), p62, beclin-1, Unc-51 like autophagy activating kinase (ULK)1, phosphor-mammalian target of rapamycin (mTOR), and β-actin were measured by Western blotting, and the respective ratios of LC3-II, Atg3, p62, and beclin-1 to LC3-I and β-actin were described (n=3–4 in each treatment group). (C, D) Terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL) staining was performed to detect apoptosis in MIN6 cells, and TUNEL-positive signals are indicated by red spots. Apoptotic cells are represented by arrowheads that indicate red spots in the nuclei. The nuclei were visualized by 4′,6-diamidino-2-phenylindole (DAPI). Scale bars=20 μm. The number of apoptotic cells was counted and described by a ratio to 100 cells (n=5–6). (E, F) The protein level of cleaved caspase-3 (c-casp3), a marker of apoptosis, was examined by Western blotting, and the level of c-casp3 was normalized to total caspase-3 (t-casp3). All values are expressed as mean±standard error of the mean (n=3–4). aP<0.05 and bP<0.01 compared to vehicle (VEH); cP<0.05 compared to PA.

  • Fig. 4 Knockdown of the autophagy-related 3 (Atg3) gene attenuates clusterin (CLU)-induced cell protection in pancreatic β-cells. Mouse insulinoma 6 (MIN6) cells were transfected with 10 nM small interfering RNA (siRNA) for the Atg3 gene or si-scramble (Scr), then incubated in 0 or 0.5 mM palmitate (PA) media with or without 10 μg/L CLU for 24 hours. (A–C) The levels of light chain 3 (LC3)-I, II, and Atg3 were analyzed by Western blotting and normalized to the LC3-I and β-actin levels of each sample, respectively. (D, E) Terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL) staining was performed to detect apoptosis after treatment, and apoptotic cells are indicated by arrowheads. The nuclei were visualized by 4′,6-diamidino-2-phenylindole (DAPI). Scale bars=20 μm. The number of apoptotic cells was counted and described by a ratio to 100 cells (n=4). (F, G) Cleaved caspase-3 (c-casp3) and total caspase-3 (t-casp3) were measured by Western blotting, and the level of c-casp3 was normalized to t-casp3. All values are expressed as mean±standard error of the mean (n=4). NS, non-significant; NT, non-transfection. aP<0.05 and bP<0.01 compared to vehicle (VEH) of the si-Scr group; cP<0.05, dP<0.01, and eP<0.001 compared to PA of the si-Scr group.


Cited by  1 articles

Targets for rescue from fatty acid-induced lipotoxicity in pancreatic beta cells
Seok-Woo Hong, Won-Young Lee
Cardiovasc Prev Pharmacother. 2022;4(2):57-62.    doi: 10.36011/cpp.2022.4.e9.


Reference

1. Poitout V, Amyot J, Semache M, Zarrouki B, Hagman D, Fontes G. Glucolipotoxicity of the pancreatic beta cell. Biochim Biophys Acta. 2010; 1801:289–98.
Article
2. Ardestani A, Li S, Annamalai K, Lupse B, Geravandi S, Dobrowolski A, et al. Neratinib protects pancreatic beta cells in diabetes. Nat Commun. 2019; 10:5015.
Article
3. Ardestani A, Maedler K. MST1: a promising therapeutic target to restore functional beta cell mass in diabetes. Diabetologia. 2016; 59:1843–9.
Article
4. Boya P, Gonzalez-Polo RA, Casares N, Perfettini JL, Dessen P, Larochette N, et al. Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol. 2005; 25:1025–40.
Article
5. Eskelinen EL, Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta. 2009; 1793:664–73.
Article
6. Mandrup-Poulsen T, Egeberg J, Nerup J, Bendtzen K, Nielsen JH, Dinarello CA. Ultrastructural studies of time-course and cellular specificity of interleukin-1 mediated islet cytotoxicity. Acta Pathol Microbiol Immunol Scand C. 1987; 95:55–63.
Article
7. Kaniuk NA, Kiraly M, Bates H, Vranic M, Volchuk A, Brumell JH. Ubiquitinated-protein aggregates form in pancreatic beta-cells during diabetes-induced oxidative stress and are regulated by autophagy. Diabetes. 2007; 56:930–9.
8. Ebato C, Uchida T, Arakawa M, Komatsu M, Ueno T, Komiya K, et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab. 2008; 8:325–32.
Article
9. Jung HS, Chung KW, Won Kim J, Kim J, Komatsu M, Tanaka K, et al. Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab. 2008; 8:318–24.
10. Zoubeidi A, Gleave M. Small heat shock proteins in cancer therapy and prognosis. Int J Biochem Cell Biol. 2012; 44:1646–56.
Article
11. Lau SH, Sham JS, Xie D, Tzang CH, Tang D, Ma N, et al. Clusterin plays an important role in hepatocellular carcinoma metastasis. Oncogene. 2006; 25:1242–50.
Article
12. July LV, Akbari M, Zellweger T, Jones EC, Goldenberg SL, Gleave ME. Clusterin expression is significantly enhanced in prostate cancer cells following androgen withdrawal therapy. Prostate. 2002; 50:179–88.
Article
13. Oh SB, Kim MS, Park S, Son H, Kim SY, Kim MS, et al. Clusterin contributes to early stage of Alzheimer’s disease pathogenesis. Brain Pathol. 2019; 29:217–31.
Article
14. Park S, Mathis KW, Lee IK. The physiological roles of apolipoprotein J/clusterin in metabolic and cardiovascular diseases. Rev Endocr Metab Disord. 2014; 15:45–53.
Article
15. Daimon M, Oizumi T, Karasawa S, Kaino W, Takase K, Tada K, et al. Association of the clusterin gene polymorphisms with type 2 diabetes mellitus. Metabolism. 2011; 60:815–22.
Article
16. Zoubeidi A, Chi K, Gleave M. Targeting the cytoprotective chaperone, clusterin, for treatment of advanced cancer. Clin Cancer Res. 2010; 16:1088–93.
Article
17. Lee J, Hong SW, Kwon H, Park SE, Rhee EJ, Park CY, et al. Resveratrol, an activator of SIRT1, improves ER stress by increasing clusterin expression in HepG2 cells. Cell Stress Chaperones. 2019; 24:825–33.
Article
18. Gregory JM, Whiten DR, Brown RA, Barros TP, Kumita JR, Yerbury JJ, et al. Clusterin protects neurons against intracellular proteotoxicity. Acta Neuropathol Commun. 2017; 5:81.
Article
19. Pereira RM, Mekary RA, da Cruz Rodrigues KC, Anaruma CP, Ropelle ER, da Silva ASR, et al. Protective molecular mechanisms of clusterin against apoptosis in cardiomyocytes. Heart Fail Rev. 2018; 23:123–9.
Article
20. Zhang H, Kim JK, Edwards CA, Xu Z, Taichman R, Wang CY. Clusterin inhibits apoptosis by interacting with activated Bax. Nat Cell Biol. 2005; 7:909–15.
Article
21. Ammar H, Closset JL. Clusterin activates survival through the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem. 2008; 283:12851–61.
Article
22. Hong SW, Lee J, Park SE, Rhee EJ, Park CY, Oh KW, et al. Repression of sterol regulatory element-binding protein 1-c is involved in the protective effects of exendin-4 in pancreatic β-cell line. Mol Cell Endocrinol. 2012; 362:242–52.
Article
23. Alnasser HA, Guan Q, Zhang F, Gleave ME, Nguan CY, Du C. Requirement of clusterin expression for prosurvival autophagy in hypoxic kidney tubular epithelial cells. Am J Physiol Renal Physiol. 2016; 310:F160–73.
Article
24. Zhang F, Kumano M, Beraldi E, Fazli L, Du C, Moore S, et al. Clusterin facilitates stress-induced lipidation of LC3 and autophagosome biogenesis to enhance cancer cell survival. Nat Commun. 2014; 5:5775.
Article
25. Choi SE, Lee SM, Lee YJ, Li LJ, Lee SJ, Lee JH, et al. Protective role of autophagy in palmitate-induced INS-1 beta-cell death. Endocrinology. 2009; 150:126–34.
26. Riahi Y, Wikstrom JD, Bachar-Wikstrom E, Polin N, Zucker H, Lee MS, et al. Autophagy is a major regulator of beta cell insulin homeostasis. Diabetologia. 2016; 59:1480–91.
Article
27. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2012; 8:445–544.
28. Randall-Demllo S, Chieppa M, Eri R. Intestinal epithelium and autophagy: partners in gut homeostasis. Front Immunol. 2013; 4:301.
Article
29. Zoubeidi A, Ettinger S, Beraldi E, Hadaschik B, Zardan A, Klomp LW, et al. Clusterin facilitates COMMD1 and I-kappaB degradation to enhance NF-kappaB activity in prostate cancer cells. Mol Cancer Res. 2010; 8:119–30.
30. Kim YS, Choi MY, Ryu JH, Lee DH, Jeon BT, Roh GS, et al. Clusterin interaction with Bcl-xL is associated with seizure-induced neuronal death. Epilepsy Res. 2012; 99:240–51.
Article
31. Yamada Y, Suzuki NN, Hanada T, Ichimura Y, Kumeta H, Fujioka Y, et al. The crystal structure of Atg3, an autophagy-related ubiquitin carrier protein (E2) enzyme that mediates Atg8 lipidation. J Biol Chem. 2007; 282:8036–43.
Article
32. Wang C, Jiang K, Gao D, Kang X, Sun C, Zhang Q, et al. Clusterin protects hepatocellular carcinoma cells from endoplasmic reticulum stress induced apoptosis through GRP78. PLoS One. 2013; 8:e55981.
Article
33. Liu T, Liu PY, Tee AE, Haber M, Norris MD, Gleave ME, et al. Over-expression of clusterin is a resistance factor to the anti-cancer effect of histone deacetylase inhibitors. Eur J Cancer. 2009; 45:1846–54.
Article
34. Sallman DA, Chen X, Zhong B, Gilvary DL, Zhou J, Wei S, et al. Clusterin mediates TRAIL resistance in prostate tumor cells. Mol Cancer Ther. 2007; 6:2938–47.
Article
35. Park IS, Che YZ, Bendayan M, Kang SW, Min BH. Up-regulation of clusterin (sulfated glycoprotein-2) in pancreatic islet cells upon streptozotocin injection to rats. J Endocrinol. 1999; 162:57–65.
Article
36. Xie MJ, Motoo Y, Su SB, Sawabu N. Expression of clusterin in pancreatic acinar cell injuries in vivo and in vitro. Pancreas. 2001; 22:126–34.
Article
37. Lee S, Hong SW, Min BH, Shim YJ, Lee KU, Lee IK, et al. Essential role of clusterin in pancreas regeneration. Dev Dyn. 2011; 240:605–15.
Article
38. Savkovic V, Gantzer H, Reiser U, Selig L, Gaiser S, Sack U, et al. Clusterin is protective in pancreatitis through anti-apoptotic and anti-inflammatory properties. Biochem Biophys Res Commun. 2007; 356:431–7.
Article
Full Text Links
  • ENM
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