Online first
Original paper
Published online: 2024-06-19

open access

Page views 123
Article views/downloads 84
Get Citation

Connect on Social Media

Connect on Social Media

Mitofusin 2 inhibits high glucose-induced apoptosis of human lens epithelial cells via modulating mitochondrial function and autophagy

Yuan-Yi Guo1, Jiang-Yue Zhao2, Han-Rong Li2, Zhuo Guo2, Hao-Yue Shen2

Abstract

Introduction. Diabetic cataract (DC) is a common ocular complication of diabetes. Mitofusin 2 (MFN2), a mitochondrial fusion protein, is involved in the pathogenesis of cataract and diabetic complications. However, its role and molecular mechanisms in DC remain unclear.

Materials and methods. DC models in rats were induced by intraperitoneal injection of streptozocin (STZ) for 12 weeks. We measured the body weight of rats, blood glucose concentrations, sorbitol dehydrogenase (SDH) activity and advanced glycation end products (AGE) content in the lenses of rats. MFN2 mRNA and protein expression levels in the lenses were detected by RT-qPCR and western blot assays. In vitro, human lens epithelial (HLE) B3 cells were treated for 48 h with 25 mM glucose (high glucose, HG) to induce cell damage. To determine the role of MFN2 in HG-induced cell damage, HLE-B3 cells were transfected with lentivirus loaded with MFN2 overexpression plasmid or short hairpin RNA (shRNA) to overexpress or knock down MFN2 expression, followed by HG exposure. Cell viability was assessed by CCK-8 assay. Flow cytometry was used to detect cell apoptosis and reactive oxygen species (ROS) level. JC-1 staining showed the changes in mitochondrial membrane potential (Δψm). The mediators related to apoptosis, mitochondrial damage, and autophagy were determined.

Results. STZ-administrated rats showed reduced body weight, increased blood glucose levels, elevated SDH activity and AGE content, suggesting successful establishment of the DC rat model. Interestingly, MFN2 expression was significantly downregulated in DC rat lens and HG-induced HLE-B3 cells. Further analysis showed that under HG conditions, MFN2 overexpression enhanced cell viability and inhibited apoptosis accompanied by decreased Bax, cleaved caspase-9 and increased Bcl-2 expression in HLE-B3 cells. MFN2 overexpression also suppressed the mitochondrial damage elicited by HG as manifested by reduced ROS production, recovered Δψm and increased mitochondrial cytochrome c (Cyto c) level. Moreover, MFN2 overexpression increased LC3BⅡ/LC3BⅠ ratio and Beclin-1 expression, but decreased p62 level, and blocked the phosphorylation of mTOR in HG-treated HLE-B3 cells. In contrast, MFN2 silencing exerted opposite effects.

Conclusions. Our findings indicate that MFN2 expression may be essential for preventing lens epithelial cell apoptosis during development of diabetic cataract.

Article available in PDF format

View PDF Download PDF file

References

  1. Greenberg MJ, Bamba S. Diabetic cataracts. Dis Mon. 2021; 67(5): 101134.
  2. Obrosova IG, Chung SSM, Kador PF. Diabetic cataracts: mechanisms and management. Diabetes Metab Res Rev. 2010; 26(3): 172–180.
  3. Peterson SR, Silva PA, Murtha TJ, et al. Cataract Surgery in Patients with Diabetes: Management Strategies. Semin Ophthalmol. 2018; 33(1): 75–82.
  4. Haddad NM, Sun JK, Abujaber S, et al. Cataract surgery and its complications in diabetic patients. Semin Ophthalmol. 2014; 29(5-6): 329–337.
  5. Murtha T, Cavallerano J. The management of diabetic eye disease in the setting of cataract surgery. Curr Opin Ophthalmol. 2007; 18(1): 13–18.
  6. Long AC, Agler A, Colitz CMH, et al. Isolation and characterization of primary canine lens epithelial cells. Vet Ophthalmol. 2008; 11(1): 38–42.
  7. Su D, Hu S, Guan L, et al. Down-regulation of GJA3 is associated with lens epithelial cell apoptosis and age-related cataract. Biochem Biophys Res Commun. 2017; 484(1): 159–164.
  8. Li WC, Kuszak JR, Dunn K, et al. Lens epithelial cell apoptosis appears to be a common cellular basis for non-congenital cataract development in humans and animals. J Cell Biol. 1995; 130(1): 169–181.
  9. Li WC, Spector A. Lens epithelial cell apoptosis is an early event in the development of UVB-induced cataract. Free Radic Biol Med. 1996; 20(3): 301–311.
  10. Rovira-Llopis S, Bañuls C, Diaz-Morales N, et al. Mitochondrial dynamics in type 2 diabetes: Pathophysiological implications. Redox Biol. 2017; 11: 637–645.
  11. Delmotte P, Sieck GC. Endoplasmic reticulum stress and mitochondrial function in airway smooth muscle. Front Cell Dev Biol. 2019; 7: 374.
  12. Zorzano A, Hernández-Alvarez MI, Sebastián D, et al. Mitofusin 2 as a driver that controls energy metabolism and insulin signaling. Antioxid Redox Signal. 2015; 22(12): 1020–1031.
  13. Muñoz JP, Ivanova S, Sánchez-Wandelmer J, et al. Mfn2 modulates the UPR and mitochondrial function via repression of PERK. EMBO J. 2013; 32(17): 2348–2361.
  14. Tang WX, Wu WH, Zeng XXi, et al. Early protective effect of mitofusion 2 overexpression in STZ-induced diabetic rat kidney. Endocrine. 2012; 41(2): 236–247.
  15. Mi X, Tang W, Chen X, et al. Mitofusin 2 attenuates the histone acetylation at collagen IV promoter in diabetic nephropathy. J Mol Endocrinol. 2016; 57(4): 233–249.
  16. Zhang R, Garrett Q, Zhou H, et al. Upregulation of miR-195 accelerates oxidative stress-induced retinal endothelial cell injury by targeting mitofusin 2 in diabetic rats. Mol Cell Endocrinol. 2017; 452: 33–43.
  17. Zhao J, Wu X, Wu D, et al. Embryonic surface ectoderm-specific mitofusin 2 conditional knockout induces congenital cataracts in mice. Sci Rep. 2018; 8(1): 1522.
  18. Yang J, Wang T, Zhang Y, et al. Altered expression of mitofusin 2 in penile tissues of diabetic rats. Andrologia. 2014; 46(5): 522–528.
  19. Bahmani F, Bathaie SZ, Aldavood SJ, et al. Prevention of α-crystallin glycation and aggregation using l-lysine results in the inhibition of in vitro catalase heat-induced-aggregation and suppression of cataract formation in the diabetic rat. Int J Biol Macromol. 2019; 132: 1200–1207.
  20. Zhao F, Maren NA, Kosentka PZ, et al. An optimized protocol for stepwise optimization of real-time RT-PCR analysis. Hortic Res. 2021; 8(1): 179–332.
  21. He L, Zhang N, Wang L, et al. Quercetin inhibits AQP1 translocation in high-glucose-cultured SRA01/04 cells through PI3K/AKT/mTOR pathway. Curr Mol Pharmacol. 2021; 14(4): 587–596.
  22. Li J, Sun Q, Qiu X, et al. Downregulation of AMPK dependent FOXO3 and TFEB involves in the inhibition of autophagy in diabetic cataract. Curr Eye Res. 2022; 47(4): 555–564.
  23. Kim J, Kim CS, Sohn E, et al. Lens epithelial cell apoptosis initiates diabetic cataractogenesis in the Zucker diabetic fatty rat. Graefes Arch Clin Exp Ophthalmol. 2010; 248(6): 811–818.
  24. Babizhayev MA, Yegorov YE. Reactive oxygen species and the aging eye: specific role of metabolically active mitochondria in maintaining lens function and in the initiation of the oxidation-induced maturity onset cataract--a novel platform of mitochondria-targeted antioxidants with broad therapeutic potential for redox regulation and detoxification of oxidants in eye diseases. Am J Ther. 2016; 23(1): e98–117.
  25. Yao Ke, Ye P, Zhang Li, et al. Epigallocatechin gallate protects against oxidative stress-induced mitochondria-dependent apoptosis in human lens epithelial cells. Mol Vis. 2008; 14: 217–223.
  26. Li Yi, Jia Yi, Zhou J, et al. Effect of methionine sulfoxide reductase B1 silencing on high-glucose-induced apoptosis of human lens epithelial cells. Life Sci. 2013; 92(3): 193–201.
  27. Yu H, Hong X, Liu L, et al. Cordycepin decreases ischemia/reperfusion injury in diabetic hearts via upregulating ampk/mfn2-dependent mitochondrial fusion. Front Pharmacol. 2021; 12: 754005.
  28. Hu L, Ding M, Tang D, et al. Targeting mitochondrial dynamics by regulating Mfn2 for therapeutic intervention in diabetic cardiomyopathy. Theranostics. 2019; 9(13): 3687–3706.
  29. Hu Y, Chen H, Zhang L, et al. The AMPK-MFN2 axis regulates MAM dynamics and autophagy induced by energy stresses. Autophagy. 2021; 17(5): 1142–1156.
  30. Tur J, Pereira-Lopes S, Vico T, et al. Mitofusin 2 in macrophages links mitochondrial ROS production, cytokine release, phagocytosis, autophagy, and bactericidal activity. Cell Rep. 2020; 32(8): 108079.
  31. Arruda AP, Hotamisligil GS, Fu S, et al. Phenotypic assays identify azoramide as a small-molecule modulator of the unfolded protein response with antidiabetic activity. Sci Transl Med. 2015; 7(292): 292ra98–397.
  32. Naón D, Hernández-Alvarez MI, Shinjo S, et al. Genes involved in mitochondrial biogenesis/function are induced in response to bilio-pancreatic diversion in morbidly obese individuals with normal glucose tolerance but not in type 2 diabetic patients. Diabetologia. 2009; 52(8): 1618–1627.
  33. Ding Y, Liu N, Zhang D, et al. Mitochondria-associated endoplasmic reticulum membranes as a therapeutic target for cardiovascular diseases. Front Pharmacol. 2024; 15: 1398381.
  34. de Brito O, Scorrano L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature. 2008; 456: 605–610.
  35. Cao Y, Chen Z, Hu J, et al. Mfn2 regulates high glucose-induced mams dysfunction and apoptosis in podocytes via PERK pathway. Front Cell Dev Biol. 2021; 9: 769213.
  36. Barlow AD, Thomas DC. Autophagy in diabetes: β-cell dysfunction, insulin resistance, and complications. DNA Cell Biol. 2015; 34(4): 252–260.
  37. Tao T, Xu H. Autophagy and obesity and diabetes. Adv Exp Med Biol. 2020; 1207: 445–461.
  38. Clarke PGH, Puyal J. Autophagic cell death exists. Autophagy. 2012; 8(6): 867–869.
  39. Fitzwalter BE, Thorburn A. Recent insights into cell death and autophagy. FEBS J. 2015; 282(22): 4279–4288.
  40. Li Ji, Ye W, Xu W, et al. Activation of autophagy inhibits epithelial to mesenchymal transition process of human lens epithelial cells induced by high glucose conditions. Cell Signal. 2020; 75: 109768.
  41. Xue R, Meng Q, Lu Di, et al. Mitofusin2 induces cell autophagy of pancreatic cancer through inhibiting the PI3K/AKT/mTOR signaling pathway. Oxid Med Cell Longev. 2018; 2018: 2798070.



Folia Histochemica et Cytobiologica