J Korean Med Sci.  2022 Sep;37(37):e276. 10.3346/jkms.2022.37.e276.

Role of Autophagy in the Pathogenesis of Diabetes and Therapeutic Potential of Autophagy Modulators in the Treatment of Diabetes and Metabolic Syndrome

Affiliations
  • 1Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea

Abstract

Autophagy is critically involved in the maintenance of intracellular nutrient homeostasis and organelle function. Dysregulated autophagy is likely to play a role in the development of metabolic disorders and diabetes because autophagy is critical in the rejuvenation of dysfunctional or stressed endoplasmic reticulum and mitochondria that play a crucial role in the development of diabetes. Indeed, systemic autophagy insufficiency led to the increased tissue lipid content, aggravated metabolic and finally more severe diabetes when metabolic stress was imposed, suggesting that autophagy insufficiency of dysfunction of lysosome, an effector organelle of autophagy, due to aging, genetic predisposition or environmental factors could be an underlying cause of diabetes. Conversely, autophagy enhancer could improve metabolic profile of obese mice by reducing tissue lipid content and ameliorating metabolic inflammation. Furthermore, clearance of human islet amyloid polypeptide (hIAPP) oligomer and amyloid that accumulate in pancreatic islets of > 90% of diabetes patients was also dependent on autophagy. Consistently, autophagy enhancer could improve glucose profile and β-cell function of transgenic mice expressing amyloidogenic hIAPP in pancreatic β-cells, which was accompanied by reduced accumulation of hIAPP oligomer or amyloid, ameliorated β-cell apoptosis and increased β-cell mass. These results suggest that autophagy enhancer could be a novel therapeutic modality against diabetes associated with lipid overload and human diabetes characterized by islet amyloid accumulation.

Keyword

Autophagy; ER; Mitochondria; Metabolic Inflammation; Islet Amyloid; IAPP

Figure

  • Fig. 1 Aggravated metabolic profile and inflammasome activation in obese mice with autophagy haploinsufficiency. (A) Monitoring of non-fasting blood glucose level showed more severe and persistent diabetes in autophagy-haploinsufficient Atg7+/--ob/ob mice compared to autophagy-competent Atg7+/+-ob/ob mice. (B) Intraperitoneal glucose tolerance test showed more severe glucose intolerance in Atg7+/--ob/ob mice compared to Atg7+/+-ob/ob mice. (C) Lipid content in the liver tissue assessed by ORO staining was higher in Atg7+/--ob/ob mice compared to Atg7+/+-ob/ob mice, showing aggravated fatty liver. (D) Immunoblot showed increased maturation of pro-IL-1β to IL-1β in stromal vascular fraction of adipose tissue from Atg7+/--ob/ob mice compared to that from Atg7+/+-ob/ob mice.IL = interleukin.Reproduced from Lim et al. Systemic autophagy insufficiency compromises adaptation to metabolic stress and facilitates progression from obesity to diabetes. Nat Commun 2014;5:4934.

  • Fig. 2 Improved glucose profile and fatty liver of obese mice by autophagy enhancer. (A) Monitoring of non-fasting blood glucose levels showed improved glucose profile after administration of MSL-7, an autophagy enhancer, to ob/ob (upper) and HFD-fed mice (lower). (B) Intraperitoneal glucose tolerance test showed improved glucose tolerance after administration of MSL-7 to ob/ob (upper) and HFD-fed mice (lower). (C) Hematoxyling and eosin staining showed ameliorated fatty liver after administration of MSL-7 to ob/ob mice.HFD = high-fat diet.Reproduced from Lim et al. A novel autophagy enhancer as therapeutic agent against metabolic syndrome and diabetes. Nat Commun 2018;9:1483.

  • Fig. 3 Reduced metabolic inflammation and inflammasome activation by autophagy enhancer. (A) Immunohistochemistry of adipose tissue sections from ob/ob mice using F4/80 antibody showed decreased number of CLS after administration of MSL for 8 weeks. (B) Real-time reverse transcription polymerase chain reaction using mRNA from adipose tissue showed reduced expression of inflammatory cytokines after administration of MSL for 8 weeks. (C) Content of IL-1β in the culture supernatant of primary peritoneal macrophages treated with 200 mM PA and/or 500 ng/mL LPS was reduced by MSL or MSL-7, as determined by enzyme-linked immunosorbent assay. (D) Mitochondrial ROS in primary peritoneal macrophages treated with 200 mM PA and/or 500 ng/mL LPS was reduced by MSL, as determined by MitoSox Red staining followed by FACS analysis.CLS = crown-like structure, TNF = tumor necrosis factor, IL = interleukin, PA = palmitic acid, LPS = lipopolysaccharide.Reproduced from Lim H et al. A novel autophagy enhancer as therapeutic agent against metabolic syndrome and diabetes. Nat Commun 9:1483, 2018

  • Fig. 4 Proposed model for improved metabolic profile in obesity-induced diabetes and human-type diabetes by an autophagy enhancer (MSL-7). Autophagy enhancer can reduce tissue lipid accumulation through activation of lipophagy in the presence of metabolic stress or lipid overload (left axis). Autophagy enhancer can also expedite rejuvenation of stressed or dysfunctional mitochondria which plays an important role in inflammasome activation, and, thereby, reduce metabolic inflammation (middle axis). Lipids such as palmitic acid can induce mitochondrial stress and activate inflammasome (gray arrow). Autophagy enhancer can accelerate clearance of hIAPP oligomer in pancreatic β-cells and protect β-cells from hIAPP oligomer-mediated apoptosis since hIAPP clearance is predominantly cleared by autophagy or lysosomal degradation rather than proteasomal degradation (right axis). Combined effects of these three axes and their interaction finally lead to the improvement of metabolic profile of obesity-induced diabetes and human-type diabetes characterized by islet amyloid accumulation through amelioration of insulin resistance and improvement of β-cell function.hIAPP = human islet amyloid polypeptide.

  • Fig. 5 Metabolic effect of autophagy enhancer on human-type diabetes of hIAPP+ mice. (A) Monitoring of non-fasting blood glucose in hIAPP+ mice during HFD feeding showed amelioration of diabetes in HFD-fed hIAPP+ mice by MSL-7, an autophagy enhancer. (B) Fasting blood glucose of HFD-fed hIAPP+ mice was also reduced by MSL-7 treatment for 8 weeks. (C) Intraperitoneal glucose tolerance test showed improved glucose tolerance by MSL-7 administration to HFD-fed hIAPP+ mice for 8 weeks. (D) Insulinogenic index representing β-cell function was also improved by MSL-7 administration to HFD-fed hIAPP+ mice for 8 weeks.hIAPP = human islet amyloid polypeptide, HFD = high-fat diet.Reproduced from Kim et al. An autophagy enhancer ameliorates diabetes of human IAPP-transgenic mice through clearance of amyloidogenic oligomer. Nat Commun 2021;12:183..

  • Fig. 6 Pancreatic islets of human-type diabetes of hIAPP+ mice treated with autophagy enhancer. (A) Immunostaining with A11 antibody showed that accumulation of hIAPP oligomer in pancreatic islets of HFD-fed hIAPP+ mice was significantly reduced by treatment with MSL-7, an autophagy enhancer for 8 weeks. (B) FSB staining showed that accumulation of islet amyloid in pancreatic islets of HFD-fed hIAPP+ mice was significantly reduced by treatment with MSL-7 for 8 weeks. (C) TUNEL staining combined with insulin immunohistochemistry showed that the number of apoptotic β-cells in pancreatic islets of HFD-fed hIAPP+ mice was significantly reduced by treatment with MSL-7 for 8 weeks. (D) Relative β-cell mass estimated by insulin immunohistochemistry and subsequent point counting was reduced in HFD-fed hIAPP+ mice, which was significantly improved by MSL-7 treatment for 8 weeks.hIAPP = human islet amyloid polypeptide, HFD = high-fat diet.Reproduced from Kim et al. An autophagy enhancer ameliorates diabetes of human IAPP-transgenic mice through clearance of amyloidogenic oligomer. Nat Commun 2021;12:183.


Reference

1. Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science. 2000; 290(5497):1717–1721. PMID: 11099404.
Article
2. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011; 147(4):728–741. PMID: 22078875.
Article
3. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature. 2006; 441(7095):885–889. PMID: 16625204.
Article
4. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006; 441(7095):880–884. PMID: 16625205.
Article
5. Ashford TP, Porter KR. Cytoplasmic components in hepatic cell lysosomes. J Cell Biol. 1962; 12(1):198–202. PMID: 13862833.
Article
6. Deter RL, De Duve C. Influence of glucagon, an inducer of cellular autophagy, on some physical properties of rat liver lysosomes. J Cell Biol. 1967; 33(2):437–449. PMID: 4292315.
Article
7. Pfeifer U. Inhibition by insulin of the formation of autophagic vacuoles in rat liver. A morphometric approach to the kinetics of intracellular degradation by autophagy. J Cell Biol. 1978; 78(1):152–167. PMID: 670291.
Article
8. Tsukada M, Ohsumi Y. Isolation and characterization of autophagy-defective mutants of Saccharomyces cerevisiae. FEBS Lett. 1993; 333(1-2):169–174. PMID: 8224160.
Article
9. Klionsky DJ, Abdel-Aziz AK, Abdelfatah S, Abdellatif M, Abdoli A, Abel S, et al. Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition). Autophagy. 2021; 17(1):1–382. PMID: 33634751.
10. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019; 176(1-2):11–42. PMID: 30633901.
Article
11. Park K, Lee MS. Current status of autophagy enhancers in metabolic disorders and other diseases. Front Cell Dev Biol. 2022; 10:811701. PMID: 35237600.
Article
12. Rubinsztein DC, Codogno P, Levine B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat Rev Drug Discov. 2012; 11(9):709–730. PMID: 22935804.
Article
13. Shoji-Kawata S, Sumpter R, Leveno M, Campbell GR, Zou Z, Kinch L, et al. Identification of a candidate therapeutic autophagy-inducing peptide. Nature. 2013; 494(7436):201–206. PMID: 23364696.
Article
14. Yu L, McPhee CK, Zheng L, Mardones GA, Rong Y, Peng J, et al. Termination of autophagy and reformation of lysosomes regulated by mTOR. Nature. 2010; 465(7300):942–946. PMID: 20526321.
Article
15. Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009; 9(1):35–51. PMID: 19117545.
Article
16. Özcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004; 306(5695):457–461. PMID: 15486293.
Article
17. Pescovitz MD, Greenbaum CJ, Krause-Steinrauf H, Becker DJ, Gitelman SE, Goland R, et al. Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N Engl J Med. 2009; 361(22):2143–2152. PMID: 19940299.
Article
18. Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Rothman DL, et al. Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science. 2003; 300(5622):1140–1142. PMID: 12750520.
Article
19. Gubas A, Dikic I. A guide to the regulation of selective autophagy receptors. FEBS J. 2022; 289(1):75–89. PMID: 33730405.
Article
20. Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol. 2008; 183(5):795–803. PMID: 19029340.
Article
21. Kim J, Lim YM, Lee MS. The role of autophagy in systemic metabolism and human-type diabetes. Mol Cells. 2018; 41(1):11–17. PMID: 29370692.
22. Yasukochi Y, Sakuma J, Takeuchi I, Kato K, Oguri M, Fujimaki T, et al. Two novel susceptibility loci for type 2 diabetes mellitus identified by longitudinal exome-wide association studies in a Japanese population. Genomics. 2019; 111(1):34–42. PMID: 29273463.
Article
23. Kim KH, Lee MS. Autophagy--a key player in cellular and body metabolism. Nat Rev Endocrinol. 2014; 10(6):322–337. PMID: 24663220.
Article
24. Lim YM, Lim H, Hur KY, Quan W, Lee HY, Cheon H, et al. Systemic autophagy insufficiency compromises adaptation to metabolic stress and facilitates progression from obesity to diabetes. Nat Commun. 2014; 5(1):4934. PMID: 25255859.
Article
25. Misawa T, Takahama M, Kozaki T, Lee H, Zou J, Saitoh T, et al. Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat Immunol. 2013; 14(5):454–460. PMID: 23502856.
Article
26. Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011; 17(2):179–188. PMID: 21217695.
Article
27. Wen H, Gris D, Lei Y, Jha S, Zhang L, Huang MT, et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol. 2011; 12(5):408–415. PMID: 21478880.
Article
28. Kim J, Kim SH, Kang H, Lee S, Park SY, Cho Y, et al. TFEB-GDF15 axis protects against obesity and insulin resistance as a lysosomal stress response. Nat Metab. 2021; 3(3):410–427. PMID: 33758420.
Article
29. Schönfeld P, Wojtczak L. Fatty acids as modulators of the cellular production of reactive oxygen species. Free Radic Biol Med. 2008; 45(3):231–241. PMID: 18482593.
Article
30. Fu S, Yang L, Li P, Hofmann O, Dicker L, Hide W, et al. Aberrant lipid metabolism disrupts calcium homeostasis causing liver endoplasmic reticulum stress in obesity. Nature. 2011; 473(7348):528–531. PMID: 21532591.
Article
31. Fernández AF, Bárcena C, Martínez-García GG, Tamargo-Gómez I, Suárez MF, Pietrocola F, et al. Autophagy couteracts weight gain, lipotoxicity and pancreatic β-cell death upon hypercaloric pro-diabetic regimens. Cell Death Dis. 2017; 8(8):e2970. PMID: 28771229.
Article
32. He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature. 2012; 481(7382):511–515. PMID: 22258505.
Article
33. Pyo JO, Yoo SM, Ahn HH, Nah J, Hong SH, Kam TI, et al. Overexpression of Atg5 in mice activates autophagy and extends lifespan. Nat Commun. 2013; 4(1):2300. PMID: 23939249.
Article
34. Castillo K, Nassif M, Valenzuela V, Rojas F, Matus S, Mercado G, et al. Trehalose delays the progression of amyotrophic lateral sclerosis by enhancing autophagy in motoneurons. Autophagy. 2013; 9(9):1308–1320. PMID: 23851366.
Article
35. Dehay B, Bové J, Rodríguez-Muela N, Perier C, Recasens A, Boya P, et al. Pathogenic lysosomal depletion in Parkin’s disease. J Neurosci. 2010; 30(37):12534–12544.
36. Lin F, Ghislat G, Luo S, Renna M, Siddiqi F, Rubinsztein DC. XIAP and cIAP1 amplifications induce Beclin 1-dependent autophagy through NFκB activation. Hum Mol Genet. 2015; 24(10):2899–2913. PMID: 25669656.
Article
37. Xie Q, Lin Q, Li D, Chen J. Imatinib induces autophagy via upregulating XIAP in GIST882 cells. Biochem Biophys Res Commun. 2017; 488(4):584–589. PMID: 28528977.
Article
38. Zummo FP, Cullen KS, Honkanen-Scott M, Shaw JA, Lovat PE, Arden C. Glucagon-Like Peptide 1 protects pancreatic β-cells from death by increasing autophagic flux and restoring lysosomal function. Diabetes. 2017; 66(5):1272–1285. PMID: 28232493.
Article
39. Zummo FP, Krishnanda SI, Georgiou M, O’Harte FP, Parthsarathy V, Cullen KS, et al. Exendin-4 stimulates autophagy in pancreatic β-cells via the RAPGEF/EPAC-Ca 2+-PPP3/calcineurin-TFEB axis. Autophagy. Forthcoming. 2021.
40. Farkas T, Høyer-Hansen M, Jäättelä M. Identification of novel autophagy regulators by a luciferase-based assay for the kinetics of autophagic flux. Autophagy. 2009; 5(7):1018–1025. PMID: 19652534.
Article
41. Lim H, Lim YM, Kim KH, Jeon YE, Park K, Kim J, et al. A novel autophagy enhancer as a therapeutic agent against metabolic syndrome and diabetes. Nat Commun. 2018; 9(1):1438. PMID: 29650965.
Article
42. Barlow AD, Nicholson ML, Herbert TP. Evidence for rapamycin toxicity in pancreatic β-cells and a review of the underlying molecular mechanisms. Diabetes. 2013; 62(8):2674–2682. PMID: 23881200.
Article
43. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley AF, et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell. 2006; 22(2):159–168. PMID: 16603397.
Article
44. Settembre C, Di Malta C, Polito VA, Garcia Arencibia M, Vetrini F, Erdin S, et al. TFEB links autophagy to lysosomal biogenesis. Science. 2011; 332(6036):1429–1433. PMID: 21617040.
45. Kahn SE, Andrikopoulos S, Verchere CB. Islet amyloid: a long-recognized but underappreciated pathological feature of type 2 diabetes. Diabetes. 1999; 48(2):241–253. PMID: 10334297.
Article
46. Westermark P, Andersson A, Westermark GT. Islet amyloid polypeptide, islet amyloid, and diabetes mellitus. Physiol Rev. 2011; 91(3):795–826. PMID: 21742788.
Article
47. Gebre-Medhin S, Olofsson C, Mulder H. Islet amyloid polypeptide in the islets of Langerhans: friend or foe? Diabetologia. 2000; 43(6):687–695. PMID: 10907112.
Article
48. Venkatanarayan A, Raulji P, Norton W, Chakravarti D, Coarfa C, Su X, et al. IAPP-driven metabolic reprogramming induces regression of p53-deficient tumours in vivo. Nature. 2015; 517(7536):626–630. PMID: 25409149.
Article
49. Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature. 2006; 443(7113):780–786. PMID: 17051204.
Article
50. Janson J, Soeller WC, Roche PC, Nelson RT, Torchia AJ, Kreutter DK, et al. Spontaneous diabetes mellitus in transgenic mice expressing human islet amyloid polypeptide. Proc Natl Acad Sci U S A. 1996; 93(14):7283–7288. PMID: 8692984.
Article
51. Verchere CB, D’Alessio DA, Palmiter RD, Weir GC, Bonner-Weir S, Baskin DG, et al. Islet amyloid formation associated with hyperglycemia in transgenic mice with pancreatic beta cell expression of human islet amyloid polypeptide. Proc Natl Acad Sci U S A. 1996; 93(8):3492–3496. PMID: 8622964.
Article
52. Kim J, Cheon H, Jeong YT, Quan W, Kim KH, Cho JM, et al. Amyloidogenic peptide oligomer accumulation in autophagy-deficient β cells induces diabetes. J Clin Invest. 2014; 124(8):3311–3324. PMID: 25036705.
Article
53. Rivera JF, Costes S, Gurlo T, Glabe CG, Butler PC. Autophagy defends pancreatic β cells from human islet amyloid polypeptide-induced toxicity. J Clin Invest. 2014; 124(8):3489–3500. PMID: 25036708.
Article
54. Shigihara N, Fukunaka A, Hara A, Komiya K, Honda A, Uchida T, et al. Human IAPP-induced pancreatic β cell toxicity and its regulation by autophagy. J Clin Invest. 2014; 124(8):3634–3644. PMID: 25036706.
Article
55. Haataja L, Gurlo T, Huang CJ, Butler PC. Islet amyloid in type 2 diabetes, and the toxic oligomer hypothesis. Endocr Rev. 2008; 29(3):303–316. PMID: 18314421.
Article
56. Paulsson JF, Andersson A, Westermark P, Westermark GT. Intracellular amyloid-like deposits contain unprocessed pro-islet amyloid polypeptide (proIAPP) in beta cells of transgenic mice overexpressing the gene for human IAPP and transplanted human islets. Diabetologia. 2006; 49(6):1237–1246. PMID: 16570161.
Article
57. Jayasinghe SA, Langen R. Lipid membranes modulate the structure of islet amyloid polypeptide. Biochemistry. 2005; 44(36):12113–12119. PMID: 16142909.
Article
58. Park K, Verchere CB. Identification of a heparin binding domain in the N-terminal cleavage site of pro-islet amyloid polypeptide. J Biol Chem. 2001; 276(20):16611–16616. PMID: 11145957.
Article
59. Kim J, Park K, Kim MJ, Lim H, Kim KH, Kim SW, et al. An autophagy enhancer ameliorates diabetes of human IAPP-transgenic mice through clearance of amyloidogenic oligomer. Nat Commun. 2021; 12(1):183. PMID: 33420039.
Article
60. Kayed R, Head E, Sarsoza F, Saing T, Cotman CW, Necula M, et al. Fibril specific, conformation dependent antibodies recognize a generic epitope common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar oligomers. Mol Neurodegener. 2007; 2(1):18–28. PMID: 17897471.
61. Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW, et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003; 300(5618):486–489. PMID: 12702875.
Article
62. Cunningham CN, Williams JM, Knupp J, Arunagiri A, Arvan P, Tsai B. Cells deploy a two-pronged strategy to rectify misfolded proinsulin aggregates. Mol Cell. 2019; 75(3):442–456.e4. PMID: 31176671.
Article
63. Arunagiri A, Haataja L, Pottekat A, Pamenan F, Kim S, Zeltser LM, et al. Proinsulin misfolding is an early event in the progression to type 2 diabetes. eLife. 2019; 8:e44532. PMID: 31184302.
Article
64. Bachar-Wikstrom E, Wikstrom JD, Ariav Y, Tirosh B, Kaiser N, Cerasi E, et al. Stimulation of autophagy improves endoplasmic reticulum stress-induced diabetes. Diabetes. 2013; 62(4):1227–1237. PMID: 23274896.
Article
65. Hresko RC, Mueckler M. mTOR.RICTOR is the Ser473 kinase for Akt/protein kinase B in 3T3-L1 adipocytes. J Biol Chem. 2005; 280(49):40406–40416. PMID: 16221682.
Article
66. Lamming DW, Ye L, Katajisto P, Goncalves MD, Saitoh M, Stevens DM, et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science. 2012; 335(6076):1638–1643. PMID: 22461615.
Article
67. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005; 307(5712):1098–1101. PMID: 15718470.
Article
68. Park K, Lim H, Kim J, Hwang H, Lee YS, Bae SH, et al. Essential role of lysosomal Ca2+-mediated TFEB activation in mitophagy and functional adaptation of pancreatic b-cells to metabolic stress. Nat Commun. 2022; 13:1300. PMID: 35288580.
69. Soleimanpour SA, Gupta A, Bakay M, Ferrari AM, Groff DN, Fadista J, et al. The diabetes susceptibility gene Clec16a regulates mitophagy. Cell. 2014; 157(7):1577–1590. PMID: 24949970.
Article
70. Ko MS, Yun JY, Baek IJ, Jang JE, Hwang JJ, Lee SE, et al. Mitophagy deficiency increases NLRP3 to induce brown fat dysfunction in mice. Autophagy. 2021; 17(5):1205–1221. PMID: 32400277.
Article
71. Wu H, Wang Y, Li W, Chen H, Du L, Liu D, et al. Deficiency of mitophagy receptor FUNDC1 impairs mitochondrial quality and aggravates dietary-induced obesity and metabolic syndrome. Autophagy. 2019; 15(11):1882–1898. PMID: 30898010.
Article
72. Bordi M, Berg MJ, Mohan PS, Peterhoff CM, Alldred MJ, Che S, et al. Autophagy flux in CA1 neurons of Alzheimer hippocampus: increased induction overburdens failing lysosomes to propel neuritic dystrophy. Autophagy. 2016; 12(12):2467–2483. PMID: 27813694.
Article
73. Fernando R, Castro JP, Flore T, Deubel S, Grune T, Ott C. Age-related maintenance of the autophagy-lysosomal system Is dependent on skeletal muscle type. Oxid Med Cell Longev. 2020; 2020:4908162. PMID: 32774673.
Article
74. Hütter E, Skovbro M, Lener B, Prats C, Rabøl R, Dela F, et al. Oxidative stress and mitochondrial impairment can be separated from lipofuscin accumulation in aged human skeletal muscle. Aging Cell. 2007; 6(2):245–256. PMID: 17376148.
Article
75. Liu WJ, Shen TT, Chen RH, Wu HL, Wang YJ, Deng JK, et al. Autophagy-lysosome pathway in renal tubular epithelial cells Is disrupted by advanced glycation end products in diabetic nephropathy. J Biol Chem. 2015; 290(33):20499–20510. PMID: 26100632.
Article
76. Zhao Y, Zhang W, Jia Q, Feng Z, Guo J, Han X, et al. High dose vitamin E attenuates diabetic nephropathy via alleviation of autophagic stress. Front Physiol. 2019; 9:1939. PMID: 30719008.
Article
Full Text Links
  • JKMS
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