Targeting GABARAPL1/HIF-2a axis to induce tumor cell apoptosis in nasopharyngeal carcinoma

Document Type : Original Article

Authors

Department of Radiation Oncology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province 570311, People’s Republic of China

Abstract

Objective(s): The primary gene mutations associated with nasopharyngeal carcinoma (NPC) are located within the phosphoinositide 3-kinase-mammalian target of rapamycin signaling pathways, which have inhibitory effects on autophagy. Compounds that target autophagy could potentially be used to treat NPC. However, autophagy-related molecular targets in NPC remain to be elucidated. We aimed to examine levels of autophagy-related genes, including autophagy-related 4B cysteine peptidase (ATG4B) and gamma-aminobutyric acid (GABA) type A receptor-associated protein-like 1 (GABARAPL1), in NPC cells and explored their potential role as novel targets for the treatment of NPC.
Materials and Methods: The mRNA and protein expression of autophagy-related genes were detected in several NPC cells. Levels of GABARAPL1 were modified by either overexpression or knockdown, followed by examining downstream targets using RT-qPCR and western blotting. The role of GABARAPL1 in NPC proliferation and apoptosis was examined by flow cytometry. Furthermore, the role of GABARAPL1 was assessed in vivo using a nude mouse xenograft tumor model. The underlying mechanism by which GABARAPL1 regulated nasopharyngeal tumor growth was investigated.
Results: Autophagy-related 4B cysteine peptidase (ATG4B), GABARAPL1, and Unc-51-like kinase 1 (ULK1) were significantly down-regulated in multiple NPC cell lines. Overexpression of GABARAPL1 up-regulated the expression of autophagy-related proteins, decreased the level of hypoxia-inducible factor (HIF)-2α, and induced apoptosis in NPC cells. Importantly, overexpression of GABARAPL1 slowed tumor growth. Western blotting showed that autophagy was activated, and HIF-2α was down-regulated in tumor tissues.
Conclusion: HIF-2α, as a substrate for autophagic degradation, may play an interesting role during NPC progression. 

Keywords

Main Subjects


1. Renaud S, Lefebvre A, Mordon S, Moralès O, Delhem N. Novel therapies boosting t cell immunity in epstein barr virus-associated nasopharyngeal carcinoma. Int J Mol Sci 2020; 21:4292-4311.
2. Xia C, Yu XQ, Zheng R, Zhang S, Zeng H, Wang J, et al. Spatial and temporal patterns of nasopharyngeal carcinoma mortality in China, 1973-2005. Cancer Lett 2017; 401:33-38.
3. Siak PY, Khoo AS, Leong CO, Hoh BP, Cheah SC. Current status and future perspectives about molecular biomarkers of nasopharyngeal carcinoma. Cancers (Basel) 2021; 13:3490-3516.
4. Peng H, Chen L, Guo R, Zhang Y, Li WF, Mao YP, et al. Clinical treatment considerations in the intensity-modulated radiotherapy era for patients with N0-category nasopharyngeal carcinoma and enlarged neck lymph nodes. Chin J Cancer 2017; 36:32-40.
5. Jiromaru R, Nakagawa T, Yasumatsu R. Advanced nasopharyngeal carcinoma: Current and emerging treatment options. Cancer Manag Res 2022; 14:2681-2689.
6. Peng Z, Wang Y, Fan R, Gao K, Xie S, Wang F, et al. Treatment of recurrent nasopharyngeal carcinoma: A sequential challenge. Cancers (Basel) 2022; 14:4111-4128.
7. Xi H, Wang S, Wang B, Hong X, Liu X, Li M, et al. The role of interaction between autophagy and apoptosis in tumorigenesis (Review). Oncol Rep 2022; 48:208-223.
8. Khan SU, Fatima K, Aisha S, Hamza B, Malik F. Redox balance and autophagy regulation in cancer progression and their therapeutic perspective. Med Oncol 2022; 40:12-32.
9. Liu PF, Tsai KL, Hsu CJ, Tsai WL, Cheng JS, Chang HW, et al. Drug repurposing screening identifies tioconazole as an atg4 inhibitor that suppresses autophagy and sensitizes cancer cells to chemotherapy. Theranostics 2018; 8:830-845.
10. Tong Y, Wang S, Wang Y, Wang F, Xie K, Song F. [Changes in the expression and phosphorylation state of autophagy-related protein ATG4 in nervous tissues of hens treated with tri-ortho-cresyl phosphate]. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi 2015; 33:7-10.
11. Maruyama T, Noda NN. Autophagy-regulating protease Atg4: structure, function, regulation and inhibition. J Antibiot (Tokyo) 2017; 71:72-78.
12. Rothe K, Lin H, Lin KB, Leung A, Wang HM, Malekesmaeili M, et al. The core autophagy protein ATG4B is a potential biomarker and therapeutic target in CML stem/progenitor cells. Blood 2014; 123:3622-3634.
13. Bortnik S, Choutka C, Horlings HM, Leung S, Baker JH, Lebovitz C, et al. Identification of breast cancer cell subtypes sensitive to ATG4B inhibition. Oncotarget 2016; 7:66970-66988.
14. Liu PF, Chen HC, Cheng JS, Tsai WL, Lee HP, Wang SC, et al. Association of atg4b and phosphorylated atg4b proteins with tumorigenesis and prognosis in oral squamous cell carcinoma. Cancers (Basel) 2019; 11:1854-1876.
15. Du X, Qi Z, Xu J, Guo M, Zhang X, Yu Z, et al. Loss of GABARAPL1 confers ferroptosis resistance to cancer stem-like cells in hepatocellular carcinoma. Mol Oncol 2022; 16:3703-3719.
16. Hirose J, Yamazoe A, Hosoyama A, Kimura N, Suenaga H, Watanabe T, et al. Draft Genome Sequence of the Polychlorinated Biphenyl-Degrading Bacterium Comamonas testosteroni KF712 (NBRC 110673). Genome Announc 2015; 3:e01214-15.
17. Liu L, Yan L, Liao N, Wu WQ, Shi JL. A review of ulk1-mediated autophagy in drug resistance of cancer. Cancers (Basel) 2020; 12:352-374.
18. He JH, Liao XL, Wang W, Li DD, Chen WD, Deng R, et al. Apogossypolone, a small-molecule inhibitor of Bcl-2, induces radiosensitization of nasopharyngeal carcinoma cells by stimulating autophagy. Int J Oncol 2014; 45:1099-1108.
19. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001; 25:402-408.
20. Zhu L, Li L, Zhang Q, Yang X, Zou Z, Hao B, et al. NOS1 S-nitrosylates PTEN and inhibits autophagy in nasopharyngeal carcinoma cells. Cell Death Discov 2017; 3:17011-17020.
21. Song L, Ma L, Chen G, Huang Y, Sun X, Jiang C, et al. [Autophagy inhibitor 3-methyladenine enhances the sensitivity of nasopharyngeal carcinoma cells to chemotherapy and radiotherapy]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2016; 41:9-18.
22. Song L, Liu H, Ma L, Zhang X, Jiang Z, Jiang C. Inhibition of autophagy by 3-MA enhances endoplasmic reticulum stress-induced apoptosis in human nasopharyngeal carcinoma cells. Oncol Lett 2013; 6:1031-1038.
23. Chow SE, Chen YW, Liang CA, Huang YK, Wang JS. Wogonin induces cross-regulation between autophagy and apoptosis via a variety of Akt pathway in human nasopharyngeal carcinoma cells. J Cell Biochem 2012; 113:3476-3485.
24. Huang J, Li HB, Yu S, Yuan F, Lou ZP. Autophagy related 4B, upregulated by HIF-1α, attenuates the sensitivity to cisplatin in nasopharyngeal carcinoma cells. Eur Rev Med Pharmacol Sci 2020; 24:4793-4802.
25. Chan EY. Regulation and function of uncoordinated-51 like kinase proteins. Antioxid Redox Signal 2012; 17:775-785.
26. Ding Y, Choi ME. Regulation of autophagy by TGF-β: Emerging role in kidney fibrosis. Semin Nephrol 2014; 34:62-71.
27. Cheng JZh, Chen JJ, Wang ZG, Yu D. MicroRNA-185 inhibits cell proliferation while promoting apoptosis and autophagy through negative regulation of TGF-β1/mTOR axis and HOXC6 in nasopharyngeal carcinoma. Cancer Biomark 2018;23:107-123. 
28. Masoud GN, Li W. HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta Pharm Sin B 2015; 5:378-389.
29. Daskalaki I, Gkikas I, Tavernarakis N. Hypoxia and selective autophagy in cancer development and therapy. Front Cell Dev Biol 2018; 6:104-125.
30. Liu XD, Yao J, Tripathi DN, Ding Z, Xu Y, Sun M, et al. Autophagy mediates HIF2α degradation and suppresses renal tumorigenesis. Oncogene 2015; 34:2450-2460.
31. Sui J, Wu J, Li X, Ma J, Cao X, Gao W, et al. [The expression and significance of hypoxia inducible factor-1alpha and microvessel density in human nasopharyngeal carcinoma]. Lin Chuang Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2008; 22:269-272.
32. Xueguan L, Xiaoshen W, Yongsheng Z, Chaosu H, Chunying S, Yan F. Hypoxia inducible factor-1 alpha and vascular endothelial growth factor expression are associated with a poor prognosis in patients with nasopharyngeal carcinoma receiving radiotherapy with carbogen and nicotinamide. Clin Oncol (R Coll Radiol) 2008; 20:606-612.
33. Benders AA, Tang W, Middeldorp JM, Greijer AE, Thorne LB, Funkhouser WK, et al. Epstein-Barr virus latent membrane protein 1 is not associated with vessel density nor with hypoxia inducible factor 1 alpha expression in nasopharyngeal carcinoma tissue. Head Neck Pathol 2009; 3:276-282.
34. Wan XB, Fan XJ, Huang PY, Dong D, Zhang Y, Chen MY, et al. Aurora-A activation, correlated with hypoxia-inducible factor-1α, promotes radiochemoresistance and predicts poor outcome for nasopharyngeal carcinoma. Cancer Sci 2012; 103:1586-1594.
35. Jonasch E, Futreal PA, Davis IJ, Bailey ST, Kim WY, Brugarolas J, et al. State of the science: An update on renal cell carcinoma. Mol Cancer Res 2012; 10:859-880.
36. Vanharanta S, Shu W, Brenet F, Hakimi AA, Heguy A, Viale A, et al. Epigenetic expansion of VHL-HIF signal output drives multiorgan metastasis in renal cancer. Nat Med 2013; 19:50-56.
37. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013; 499:43-49.
38. Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 2013; 45:860-867.
39. Starker LF, Akerström T, Long WD, Delgado-Verdugo A, Donovan P, Udelsman R, et al. Frequent germ-line mutations of the MEN1, CASR, and HRPT2/CDC73 genes in young patients with clinically non-familial primary hyperparathyroidism. Horm Cancer 2012; 3:44-51.
40. Kitagawa N, Kondo S, Wakisaka N, Zen Y, Nakanishi Y, Tsuji A, et al. Expression of seven-in-absentia homologue 1 and hypoxia-inducible factor 1 alpha: Novel prognostic factors of nasopharyngeal carcinoma. Cancer Lett 2013; 331:52-57.
41. Zhang C, Yang X, Zhang Q, Yang B, Xu L, Qin Q, et al. Berberine radiosensitizes human nasopharyngeal carcinoma by suppressing hypoxia-inducible factor-1α expression. Acta Otolaryngol 2014; 134:185-192.