Down-Regulation of CEND1 Expression Contributes to The Progression and Temozolomide Resistance of Glioma

Document Type : Original Article

Authors

Neurosurgery Department 2, The Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China

Abstract

Objective: This study was conducted to clarify the expression characteristics of cell cycle exit and neuronal differentiation
1 (CEND1) in glioma and its effects on the proliferation, migration, invasion, and resistance to temozolomide (TMZ) of
glioma cells.
Materials and Methods: In this experimental study, CEND1 expression in glioma tissues and its relationship with
patients’ survival were analyzed through bioinformatics. Quantitative real-time polymerase chain reaction (qRT-PCR)
and immunohistochemistry were performed to detect CEND1 expression in glioma tissues. The cell counting kit-8
(CCK-8) method was adopted to detect cell viability and the effects of different concentrations of TMZ on the inhibition
rate of glioma cell proliferation, and the median inhibitory concentration of TMZ (IC50 value) was calculated. 5-Bromo-
2'-deoxyuridine (BrdU), wound healing and Transwell assays were performed to evaluate the impacts of CEND1 on
glioma cell proliferation, migration, and invasion. Besides, the Kyoto Encyclopedia of Genes and Genomes (KEGG)
analysis, Gene Ontology (GO) analysis, and Gene Set Enrichment Analysis (GSEA) were applied to predict the
pathways regulated by CEND1. Nuclear factor-kappa B p65 (NF-κB p65) and phospho-p65 (p-p65) expression were
detected by Western blot.
Results: CEND1 expression was reduced in glioma tissues and cells, and its low expression was significantly
associated with the shorter survival of glioma patients. CEND1 knockdown promoted glioma cell growth, migration,
and invasion, and increased the IC50 value of TMZ, whereas up-regulating CEND1 expression worked oppositely.
Genes co-expressed with CEND1 were enriched in the NF-κB pathway, and knocking down CEND1 facilitated p-p65
expression, while CEND1 overexpression suppressed p-p65 expression.
Conclusion: CEND1 inhibits glioma cell proliferation, migration, invasion, and resistance to TMZ by inhibiting the NF-
κB pathway.

Keywords


  1. Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, et al. Glioma. Nat Rev Dis Primers. 2015; 1: 15017.
  2. Delgado-López PD, Corrales-García EM. Survival in glioblastoma: a review on the impact of treatment modalities. Clin Transl Oncol. 2016; 18(11): 1062-1071.
  3. Tang A, Liang J, Mai Y, Sun H. Advances in research on glioma microenvironment and immunotherapeutic targets. Journal of Cancer Discovery. 2022; 1(1): 14-29.
  4. Zhu P, Du XL, Lu G, Zhu JJ. Survival benefit of glioblastoma patients after FDA approval of temozolomide concomitant with radiation and bevacizumab: a population-based study. Oncotarget. 2017; 8(27): 44015-44031.
  5. Zhu Y, Zhang X, Wang L, Ji Z, Xie M, Zhou X, et al. Loss of SH3GL2 promotes the migration and invasion behaviours of glioblastoma cells through activating the STAT3/MMP2 signalling. J Cell Mol Med. 2017; 21(11): 2685-2694.
  6. Yang JK, Yang JP, Tong J, Jing SY, Fan B, Wang F, et al. Exosomal miR-221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma. J Neurooncol. 2017; 131(2): 255-265.
  7. Gaitanou M, Segklia K, Matsas R. Cend1, a story with many tales: from regulation of cell cycle progression/exit of neural stem cells to brain structure and function. Stem Cells Int. 2019; 2019: 2054783.
  8. Patsavoudi E, Merkouri E, Thomaidou D, Sandillon F, Alonso G, Matsas R. Characterization and localization of the BM88 antigen in the developing and adult rat brain. J Neurosci Res. 1995; 40(4): 506-518.
  9. Sergaki MC, Guillemot F, Matsas R. Impaired cerebellar development and deficits in motor coordination in mice lacking the neuronal protein BM88/Cend1. Mol Cell Neurosci. 2010; 44(1): 15-29.
  10. Politis PK, Makri G, Thomaidou D, Geissen M, Rohrer H, Matsas R. BM88/CEND1 coordinates cell cycle exit and differentiationof neuronal precursors. Proc Natl Acad Sci USA. 2007; 104(45): 17861-17866.
  11. Fleischer T, Frigessi A, Johnson KC, Edvardsen H, Touleimat N, Klajic J, et al. Genome-wide DNA methylation profiles in progression to in situ and invasive carcinoma of the breast with impact on gene transcription and prognosis. Genome Biol. 2014; 15(8): 435.
  12. Tsioras K, Papastefanaki F, Politis PK, Matsas R, Gaitanou M. Functional interactions between BM88/Cend1, Ran-binding protein M and Dyrk1B kinase affect cyclin D1 levels and cell cycle progression/ exit in mouse neuroblastoma cells. PLoS One. 2013; 8(11): e82172.
  13. Politis PK, Thomaidou D, Matsas R. Coordination of cell cycle exit and differentiation of neuronal progenitors. Cell Cycle. 2008; 7(6): 691-697.
  14. Sergaki MC, Guillemot F, Matsas R. Impaired cerebellar development and deficits in motor coordination in mice lacking the neuronal protein BM88/Cend1. Mol Cell Neurosci. 2010; 44(1): 15-29.
  15. Zhang P, Chen XB, Ding BQ, Liu HL, He T. Down-regulation of ABCE1 inhibits temozolomide resistance in glioma through the PI3K/Akt/NF-κB signaling pathway. Biosci Rep. 2018; 38(6): BSR20181711.
  16. Zhao Z, Liu M, Long W, Yuan J, Li H, Zhang C, et al. Knockdown lncRNA CRNDE enhances temozolomide chemosensitivity by regulating autophagy in glioblastoma. Cancer Cell Int. 2021; 21(1): 456.
  17. Politis PK, Akrivou S, Hurel C, Papadodima O, Matsas R. BM88/ Cend1 is involved in histone deacetylase inhibition-mediated growth arrest and differentiation of neuroblastoma cells. FEBS Lett. 2008; 582(5): 741-748.
  18. Athanassiou H, Synodinou M, Maragoudakis E, Paraskevaidis M, Verigos C, Misailidou D, et al. Randomized phase II study of temozolomide and radiotherapy compared with radiotherapy alone in newly diagnosed glioblastoma multiforme. J Clin Oncol. 2005; 23(10): 2372-2377.
  19. Zou Y, Chen M, Zhang S, Miao Z, Wang J, Lu X, et al. TRPC5‑induced autophagy promotes the TMZ‑resistance of glioma cells via the CAMMKβ/AMPKα/mTOR pathway. Oncol Rep. 2019; 41(6): 3413-3423.
  20. Xu X, Wang Z, Liu N, Cheng Y, Jin W, Zhang P, et al. Association between SOX9 and CA9 in glioma, and its effects on chemosensitivity to TMZ. Int J Oncol. 2018; 53(1): 189-202.
  21. Zhu Y, Jia J, Zhao G, Huang X, Wang L, Zhang Y, et al. Multi-responsive nanofibers composite gel for local drug delivery to inhibit recurrence of glioma after operation. J Nanobiotechnology. 2021; 19(1): 198.
  22. Thomas A, Tanaka M, Trepel J, Reinhold WC, Rajapakse VN, Pommier Y. Temozolomide in the Era of Precision Medicine. Cancer Res. 2017; 77(4): 823-826.
  23. Stéphanou A, Ballesta A. pH as a potential therapeutic target to improve temozolomide antitumor efficacy: a mechanistic modeling study. Pharmacol Res Perspect. 2019; 7(1): e00454.
  24. Yan Y, Xu Z, Dai S, Qian L, Sun L, Gong Z. Targeting autophagy to sensitive glioma to temozolomide treatment. J Exp Clin Cancer Res. 2016; 35: 23.
  25. Brigliadori G, Foca F, Dall’Agata M, Rengucci C, Melegari E, Cerasoli S, et al. Defining the cutoff value of MGMT gene promoter methylation and its predictive capacity in glioblastoma. J Neurooncol. 2016; 128(2): 333-339.
  26. Luo H, Chen Z, Wang S, Zhang R, Qiu W, Zhao L, et al. c-MycmiR- 29c-REV3L signalling pathway drives the acquisition of temozolomide resistance in glioblastoma. Brain. 2015; 138(Pt 12): 3654-3672.
  27. Christmann M, Diesler K, Majhen D, Steigerwald C, Berte N, Freund H, et al. Integrin αVβ3 silencing sensitizes malignant glioma cells to temozolomide by suppression of homologous recombination repair. Oncotarget. 2017; 8(17): 27754-27771.
  28. van Nifterik KA, van den Berg J, van der Meide WF, Ameziane N, Wedekind LE, Steenbergen RD, et al. Absence of the MGMT protein as well as methylation of the MGMT promoter predict the sensitivity for temozolomide. Br J Cancer. 2010; 103(1): 29-35.
  29. Qiu ZK, Shen D, Chen YS, Yang QY, Guo CC, Feng BH, et al. Enhanced MGMT expression contributes to temozolomide resistance in glioma stem-like cells. Chin J Cancer. 2014; 33(2): 115-122.
  30. Chahal M, Abdulkarim B, Xu Y, Guiot MC, Easaw JC, Stifani N, et al. O6-Methylguanine-DNA methyltransferase is a novel negative effector of invasion in glioblastoma multiforme. Mol Cancer Ther. 2012; 11(11): 2440-2450.
  31. Sen R, Baltimore D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell. 1986; 46(5): 705-716.
  32. Sharif O, Bolshakov VN, Raines S, Newham P, Perkins ND. Transcriptional profiling of the LPS induced NF-kappaB response in macrophages. BMC Immunol. 2007; 8: 1.
  33. Li H, Lin X. Positive and negative signaling components involved in TNFalpha-induced NF-kappaB activation. Cytokine. 2008; 41(1): 1-8.
  34. Wang Q, Jiang H, Li Y, Chen W, Li H, Peng K, et al. Targeting NFkB signaling with polymeric hybrid micelles that co-deliver siRNA and dexamethasone for arthritis therapy. Biomaterials. 2017; 122: 10-22.
  35. Greten FR, Karin M. The IKK/NF-kappaB activation pathway-a target for prevention and treatment of cancer. Cancer Lett. 2004; 206(2): 193-199.
  36. Tang F, Wang H, Chen E, Bian E, Xu Y, Ji X, et al. LncRNA-ATB promotes TGF-β-induced glioma cells invasion through NF-κB and P38/MAPK pathway. J Cell Physiol. 2019; 234(12): 23302-23314.
  37. Zhao P, Wang M, An J, Sun H, Li T, Li D. A positive feedback loop of miR-30a-5p-WWP1-NF-κB in the regulation of glioma development. Int J Biochem Cell Biol. 2019; 112: 39-49.