Cardiovasc Prev Pharmacother.  2022 Apr;4(2):57-62. 10.36011/cpp.2022.4.e9.

Targets for rescue from fatty acid-induced lipotoxicity in pancreatic beta 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

A persistent intake of excess calories increases plasma levels of free fatty acids, particularly the saturated form that has been shown to exert toxic effects on pancreatic beta cells by inducing dysfunction and apoptosis (i.e., lipotoxicity). An insufficient supply of insulin due to beta cell failure is a major factor in the onset and progression of type 2 diabetes; therefore, it is crucial to understand the cellular mechanisms of lipotoxicity to prevent beta cell failure. Many studies on the effects of lipotoxicity have demonstrated the various factors responsible for beta cell impairment, but the mechanisms of dysfunction and apoptosis resulting from lipotoxicity have not been fully described. This review discusses lipotoxicity-induced alterations of cellular mechanisms, and assesses drugs such as incretin mimetics, thiazolidinedione, and clusterin. Understanding the molecular mechanisms of lipotoxicity-induced beta cell failure is useful in guiding the development of new therapeutic targets for diabetes treatment.

Keyword

Pancreatic beta cell; Lipotoxicity; Inflammation; Endoplasmic recticulum stress; Autophagy

Figure

  • Fig. 1. Summary of lipotoxicity by fatty acids (FAs) and autophagy activation by clusterin. (A) Chronic exposure to FAs induces beta cell dysfunction and apoptosis through cellular lipid synthesis, inflammation, and endoplasmic reticulum (ER) stress. Activated sterol regulatory element-binding protein-1c (SREBP1c) by FAs is translocated to the nucleus and upregulates the expression of fatty acid synthase (Fas) and acetyl-CoA carboxylase-1 (Acc1) for synthesis of cellular lipid. Expression of inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and IL-1β, is upregulated by c-Jun N-terminal kinase (JNK)/nuclear factor-kappa B (NF-κB) activation, and severe ER stress by FAs ultimately causes the expression of C/EBP homologous protein (CHOP), an apoptotic protein. This alteration of cellular mechanisms is a strong inducer of the apoptosis of pancreatic beta cells. Exendin-4 (Ex-4) or pioglitazone (PIO) showed a strong effect in the rescue of pancreatic beta cells from lipotoxicity. Treatment with Ex-4 maintains lipid metabolism by repressing the activation of SREBP1c. Pioglitazone blocks inflammatory responses repressing JNK/NF-κB activation and decreases CHOP expression mediated by the attenuation of ER stress. Protein kinase RNA-like endoplasmic reticulum kinase (PERK), Activating transcription factor-4 (ATF4), phosphorylated eukaryotic translation initiation factor 2 alpha subunit (pe-IF2α). (B) Autophagy activation protects pancreatic beta cells from lipotoxicity. Clusterin (CLU) facilitates the binding of microtubule-associated protein light chain 3 (LC3) with autophagy-related gene-3 (Atg3) and p62, activating autophagy.


Reference

1. DeFronzo RA. Lilly lecture 1987. The triumvirate: beta-cell, muscle, liver. A collusion responsible for NIDDM. Diabetes. 1988; 37:667–87.
2. Cohrs CM, Panzer JK, Drotar DM, Enos SJ, Kipke N, Chen C, et al. Dysfunction of persisting β cells is a key feature of early type 2 diabetes pathogenesis. Cell Rep. 2020; 31:107469.
Article
3. Carlsson M, Wessman Y, Almgren P, Groop L. High levels of nonesterified fatty acids are associated with increased familial risk of cardiovascular disease. Arterioscler Thromb Vasc Biol. 2000; 20:1588–94.
Article
4. Salgin B, Ong KK, Thankamony A, Emmett P, Wareham NJ, Dunger DB. Higher fasting plasma free fatty acid levels are associated with lower insulin secretion in children and adults and a higher incidence of type 2 diabetes. J Clin Endocrinol Metab. 2012; 97:3302–9.
Article
5. Mahendran Y, Cederberg H, Vangipurapu J, Kangas AJ, Soininen P, Kuusisto J, et al. Glycerol and fatty acids in serum predict the development of hyperglycemia and type 2 diabetes in Finnish men. Diabetes Care. 2013; 36:3732–8.
Article
6. Riserus U, Willett WC, Hu FB. Dietary fats and prevention of type 2 diabetes. Prog Lipid Res. 2009; 48:44–51.
Article
7. Hodge AM, English DR, O'Dea K, Sinclair AJ, Makrides M, Gibson RA, et al. Plasma phospholipid and dietary fatty acids as predictors of type 2 diabetes: interpreting the role of linoleic acid. Am J Clin Nutr. 2007; 86:189–97.
Article
8. Sobczak AI, A Blindauer C, J Stewart A. Changes in plasma free fatty acids associated with type-2 diabetes. Nutrients. 2019; 11:2022.
Article
9. Poitout V, Hagman D, Stein R, Artner I, Robertson RP, Harmon JS. Regulation of the insulin gene by glucose and fatty acids. J Nutr. 2006; 136:873–6.
Article
10. Ritz-Laser B, Meda P, Constant I, Klages N, Charollais A, Morales A, et al. Glucose-induced preproinsulin gene expression is inhibited by the free fatty acid palmitate. Endocrinology. 1999; 140:4005–14.
Article
11. Hagman DK, Hays LB, Parazzoli SD, Poitout V. Palmitate inhibits insulin gene expression by altering PDX-1 nuclear localization and reducing MafA expression in isolated rat islets of Langerhans. J Biol Chem. 2005; 280:32413–8.
Article
12. Wang Y, Wang PY, Takashi K. Chronic effects of different non-esterified fatty acids on pancreatic islets of rats. Endocrine. 2006; 29:169–73.
Article
13. Maris M, Robert S, Waelkens E, Derua R, Hernangomez MH, D'Hertog W, et al. Role of the saturated nonesterified fatty acid palmitate in beta cell dysfunction. J Proteome Res. 2013; 12:347–62.
Article
14. Dixon G, Nolan J, McClenaghan NH, Flatt PR, Newsholme P. Arachidonic acid, palmitic acid and glucose are important for the modulation of clonal pancreatic beta-cell insulin secretion, growth and functional integrity. Clin Sci (Lond). 2004; 106:191–9.
15. El-Assaad W, Buteau J, Peyot ML, Nolan C, Roduit R, Hardy S, et al. Saturated fatty acids synergize with elevated glucose to cause pancreatic beta-cell death. Endocrinology. 2003; 144:4154–63.
16. Acosta-Montano P, Garcia-Gonzalez V. Effects of dietary fatty acids in pancreatic beta cell metabolism, implications in homeostasis. Nutrients. 2018; 10:393.
Article
17. Unger RH, Zhou YT. Lipotoxicity of beta-cells in obesity and in other causes of fatty acid spillover. Diabetes. 2001; 50 Suppl 1:S118–21.
Article
18. Maestre I, Jordan J, Calvo S, Reig JA, Cena V, Soria B, et al. Mitochondrial dysfunction is involved in apoptosis induced by serum withdrawal and fatty acids in the beta-cell line INS-1. Endocrinology. 2003; 144:335–45.
19. Piro S, Anello M, Di Pietro C, Lizzio MN, Patane G, Rabuazzo AM, et al. Chronic exposure to free fatty acids or high glucose induces apoptosis in rat pancreatic islets: possible role of oxidative stress. Metabolism. 2002; 51:1340–7.
Article
20. Weir GC, Bonner-Weir S. Five stages of evolving beta-cell dysfunction during progression to diabetes. Diabetes. 2004; 53 Suppl 3:S16–21.
Article
21. Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A. Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic beta-cell apoptosis. Endocrinology. 2006; 147:3398–407.
Article
22. Shimabukuro M, Zhou YT, Levi M, Unger RH. Fatty acid-induced beta cell apoptosis: a link between obesity and diabetes. Proc Natl Acad Sci U S A. 1998; 95:2498–502.
23. Unger RH, Orci L. Lipoapoptosis: its mechanism and its diseases. Biochim Biophys Acta. 2002; 1585:202–12.
Article
24. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest. 2002; 109:1125–31.
Article
25. Brown MS, Goldstein JL. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell. 1997; 89:331–40.
Article
26. Amemiya-Kudo M, Shimano H, Hasty AH, Yahagi N, Yoshikawa T, Matsuzaka T, et al. Transcriptional activities of nuclear SREBP-1a, -1c, and -2 to different target promoters of lipogenic and cholesterogenic genes. J Lipid Res. 2002; 43:1220–35.
Article
27. Pai JT, Guryev O, Brown MS, Goldstein JL. Differential stimulation of cholesterol and unsaturated fatty acid biosynthesis in cells expressing individual nuclear sterol regulatory element-binding proteins. J Biol Chem. 1998; 273:26138–48.
Article
28. Tovar AR, Torre-Villalvazo I, Ochoa M, Elias AL, Ortiz V, Aguilar-Salinas CA, et al. Soy protein reduces hepatic lipotoxicity in hyperinsulinemic obese Zucker fa/fa rats. J Lipid Res. 2005; 46:1823–32.
Article
29. Kakuma T, Lee Y, Higa M, Wang Zw, Pan W, Shimomura I, et al. Leptin, troglitazone, and the expression of sterol regulatory element binding proteins in liver and pancreatic islets. Proc Natl Acad Sci U S A. 2000; 97:8536–41.
Article
30. 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
31. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007; 356:1517–26.
Article
32. Cai K, Qi D, Wang O, Chen J, Liu X, Deng B, et al. TNF-α acutely upregulates amylin expression in murine pancreatic beta cells. Diabetologia. 2011; 54:617–26.
Article
33. Montane J, Klimek-Abercrombie A, Potter KJ, Westwell-Roper C, Bruce Verchere C. Metabolic stress, IAPP and islet amyloid. Diabetes Obes Metab. 2012; 14 Suppl 3:68–77.
Article
34. Masters SL, Dunne A, Subramanian SL, Hull RL, Tannahill GM, Sharp FA, et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1β in type 2 diabetes. Nat Immunol. 2010; 11:897–904.
Article
35. van Raalte DH, Diamant M. Glucolipotoxicity and beta cells in type 2 diabetes mellitus: target for durable therapy? Diabetes Res Clin Pract. 2011; 93 Suppl 1:S37–46.
Article
36. Yung J, Giacca A. Role of c-Jun N-terminal Kinase (JNK) in obesity and type 2 diabetes. Cells. 2020; 9:706.
Article
37. Igoillo-Esteve M, Marselli L, Cunha DA, Ladriere L, Ortis F, Grieco FA, et al. Palmitate induces a pro-inflammatory response in human pancreatic islets that mimics CCL2 expression by beta cells in type 2 diabetes. Diabetologia. 2010; 53:1395–405.
Article
38. Nakatani Y, Kaneto H, Kawamori D, Yoshiuchi K, Hatazaki M, Matsuoka TA, et al. Involvement of endoplasmic reticulum stress in insulin resistance and diabetes. J Biol Chem. 2005; 280:847–51.
Article
39. Marchetti P, Bugliani M, Lupi R, Marselli L, Masini M, Boggi U, et al. The endoplasmic reticulum in pancreatic beta cells of type 2 diabetes patients. Diabetologia. 2007; 50:2486–94.
Article
40. Laybutt DR, Preston AM, Akerfeldt MC, Kench JG, Busch AK, Biankin AV, et al. Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia. 2007; 50:752–63.
Article
41. Johnson JD. Proteomic identification of carboxypeptidase E connects lipid-induced beta-cell apoptosis and dysfunction in type 2 diabetes. Cell Cycle. 2009; 8:38–42.
Article
42. Li G, Mongillo M, Chin KT, Harding H, Ron D, Marks AR, et al. Role of ERO1-alpha-mediated stimulation of inositol 1,4,5-triphosphate receptor activity in endoplasmic reticulum stress-induced apoptosis. J Cell Biol. 2009; 186:783–92.
43. Hong SW, Lee J, Cho JH, Kwon H, Park SE, Rhee EJ, et al. Pioglitazone attenuates palmitate-induced inflammation and endoplasmic reticulum stress in pancreatic β-cells. Endocrinol Metab (Seoul). 2018; 33:105–13.
Article
44. Pistrosch F, Passauer J, Fischer S, Fuecker K, Hanefeld M, Gross P. In type 2 diabetes, rosiglitazone therapy for insulin resistance ameliorates endothelial dysfunction independent of glucose control. Diabetes Care. 2004; 27:484–90.
Article
45. Gong L, Jin H, Li Y, Quan Y, Yang J, Tang Q, et al. Rosiglitazone ameliorates skeletal muscle insulin resistance by decreasing free fatty acids release from adipocytes. Biochem Biophys Res Commun. 2020; 533:1122–8.
Article
46. Ishida H, Takizawa M, Ozawa S, Nakamichi Y, Yamaguchi S, Katsuta H, et al. Pioglitazone improves insulin secretory capacity and prevents the loss of beta-cell mass in obese diabetic db/db mice: possible protection of beta cells from oxidative stress. Metabolism. 2004; 53:488–94.
Article
47. Lin CY, Gurlo T, Haataja L, Hsueh WA, Butler PC. Activation of peroxisome proliferator-activated receptor-gamma by rosiglitazone protects human islet cells against human islet amyloid polypeptide toxicity by a phosphatidylinositol 3'-kinase-dependent pathway. J Clin Endocrinol Metab. 2005; 90:6678–86.
48. 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
49. 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.
50. 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
51. 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.
52. 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.
53. Hong SW, Lee J, Kim MJ, Moon SJ, Kwon H, Park SE, et al. Clusterin protects lipotoxicity-induced apoptosis via upregulation of autophagy in insulin-secreting cells. Endocrinol Metab (Seoul). 2020; 35:943–53.
Article
Full Text Links
  • CPP
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