Skip to main content

Advertisement

Log in

CDC6 is up-regulated and a poor prognostic signature in glioblastoma multiforme

  • Research Article
  • Published:
Clinical and Translational Oncology Aims and scope Submit manuscript

Abstract

Purpose

Glioblastoma multiforme (GBM) represents the most common and the most malignant type of brain tumor. Cell division cycle 6 (CDC6), a gene associated with DNA replication initiation, has been proven to be associated with the prognosis of multiple tumors. In this study, we aim to explore the association between CDC6 expression and GBM carcinogenesis and prognosis.

Methods

CDC6 expression in normal cells and GBM cells was explored by analyzing TCGA dataset, as well as by RT-PCR and western blot methods. Survival analysis was performed by the Kaplan–Meier method. Multivariate Cox-regression analysis was adopted to estimate the independence of CDC6 as a GBM prognostic factor.

Results and conclusions

Elevated CDC6 levels in GBM tumor tissues compared with those in normal brain tissues were illustrated by analyzing the gene expression profiles from TCGA dataset, and confirmed by RT-PCR and western blot assays in GBM tumor and normal human astrocyte cell lines. Kaplan–Meier analysis indicated the negative influence of high CDC6 expression on GBM overall survival (OS) probability and days to progression (D2P) after initial treatment, but not on days to recurrence (D2R) after initial treatment. Multivariate Cox regression analysis showed CDC6 as an independent signature marker gene for GBM prognosis. In addition, the combination of CDC6 mRNA expression and CpG island methylator phenotype (CIMP) could sensitively predict 3-year OS and D2P. In conclusion, our study uncovered the role of CDC6 in GBM carcinogenesis and prognosis for the first time, which could shed new light on GBM diagnosis and treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Tyler B, DiMeco F, Grossman R, Pradilla G. Current and future novel treatments for glioblastoma multiforme. Biomed Res Int. 2014;2014:432195. https://doi.org/10.1155/2014/432195.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Kalkan R. Glioblastoma multiforme: the genetic perspective of the treatment planning. Crit Rev Eukaryot Gene Expr. 2015;25(4):281–5. https://doi.org/10.1615/critreveukaryotgeneexpr.2015014259.

    Article  PubMed  Google Scholar 

  3. Williams DS. Glioblastoma multiforme. J Insur Med. 2014;44(1):62–4.

    PubMed  Google Scholar 

  4. Kotliarova S, Fine HA. SnapShot: glioblastoma multiforme. Cancer Cell. 2012;21(5):710-e1. https://doi.org/10.1016/j.ccr.2012.04.031.

    Article  CAS  Google Scholar 

  5. Maiese K. “Micromanaging” glioblastoma multiforme: the potential of MicroRNAs, circular RNAs, and the hippo pathway as novel treatment strategies. Curr Neurovasc Res. 2018;15(3):173–4. https://doi.org/10.2174/1567202615999180711123907.

    Article  PubMed  Google Scholar 

  6. Khan IS, Ehtesham M. Targeting glioblastoma cancer stem cells: the next great hope? Neurosurg Focus. 2014;37(6):E7. https://doi.org/10.3171/2014.9.focus14509.

    Article  PubMed  Google Scholar 

  7. Kim GS, Lee I, Kim JH, Hwang DS. The replication protein Cdc6 suppresses centrosome over-duplication in a manner independent of its ATPase activity. Mol Cells. 2017;40(12):925–34. https://doi.org/10.14348/molcells.2017.0191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Borlado LR, Mendez J. CDC6: from DNA replication to cell cycle checkpoints and oncogenesis. Carcinogenesis. 2008;29(2):237–43. https://doi.org/10.1093/carcin/bgm268.

    Article  CAS  PubMed  Google Scholar 

  9. Parker MW, Botchan MR, Berger JM. Mechanisms and regulation of DNA replication initiation in eukaryotes. Crit Rev Biochem Mol Biol. 2017;52(2):107–44. https://doi.org/10.1080/10409238.2016.1274717.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Teixeira LK, Reed SI. Cdc6: Skin in the carcinogenesis game. Cell Cycle. 2016;15(3):313. https://doi.org/10.1080/15384101.2015.1131528.

    Article  CAS  PubMed  Google Scholar 

  11. Kim YH, Byun YJ, Kim WT, Jeong P, Yan C, Kang HW, et al. CDC6 mRNA expression is associated with the aggressiveness of prostate cancer. J Korean Med Sci. 2018;33(47):e303. https://doi.org/10.3346/jkms.2018.33.e303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Deng Y, Jiang L, Wang Y, Xi Q, Zhong J, Liu J, et al. High expression of CDC6 is associated with accelerated cell proliferation and poor prognosis of epithelial ovarian cancer. Pathol Res Pract. 2016;212(4):239–46. https://doi.org/10.1016/j.prp.2015.09.014.

    Article  CAS  PubMed  Google Scholar 

  13. Rossi E, Klersy C, Manca R, Zuffardi O, Solcia E. Correlation between genomic alterations assessed by array comparative genomic hybridization, prognostically informative histologic subtype, stage, and patient survival in gastric cancer. Hum Pathol. 2011;42(12):1937–45. https://doi.org/10.1016/j.humpath.2011.02.016.

    Article  CAS  PubMed  Google Scholar 

  14. Jiang W, Yu Y, Liu J, Zhao Q, Wang J, Zhang J, et al. Downregulation of Cdc6 inhibits tumorigenesis of osteosarcoma in vivo and in vitro. Biomed Pharmacother = Biomedecine Pharmacotherapie. 2019;115:108949. https://doi.org/10.1016/j.biopha.2019.108949.

    Article  CAS  PubMed  Google Scholar 

  15. Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, et al. LncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215. J Cell Physiol. 2019;234(6):9105–17. https://doi.org/10.1002/jcp.27587.

    Article  CAS  PubMed  Google Scholar 

  16. Mahadevappa R, Neves H, Yuen SM, Bai Y, McCrudden CM, Yuen HF, et al. The prognostic significance of Cdc6 and Cdt1 in breast cancer. Sci Rep. 2017;7(1):985. https://doi.org/10.1038/s41598-017-00998-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hu Y, Wang L, Li Z, Wan Z, Shao M, Wu S, et al. Potential prognostic and diagnostic values of CDC6, CDC45, ORC6 and SNHG7 in colorectal cancer. OncoTargets Ther. 2019;12:11609–21. https://doi.org/10.2147/OTT.S231941.

    Article  CAS  Google Scholar 

  18. Yan X, Wan H, Hao X, Lan T, Li W, Xu L, et al. Importance of gene expression signatures in pancreatic cancer prognosis and the establishment of a prediction model. Cancer Manag Res. 2019;11:273–83. https://doi.org/10.2147/CMAR.S185205.

    Article  CAS  PubMed  Google Scholar 

  19. Sugimoto N, Fujita M. Molecular mechanism for chromatin regulation during MCM loading in mammalian cells. Adv Exp Med Biol. 2017;1042:61–78. https://doi.org/10.1007/978-981-10-6955-0_3.

    Article  CAS  PubMed  Google Scholar 

  20. Ohta S, Koide M, Tokuyama T, Yokota N, Nishizawa S, Namba H. Cdc6 expression as a marker of proliferative activity in brain tumors. Oncol Rep. 2001;8(5):1063–6. https://doi.org/10.3892/or.8.5.1063.

    Article  CAS  PubMed  Google Scholar 

  21. Stangeland B, Mughal AA, Grieg Z, Sandberg CJ, Joel M, Nygard S, et al. Combined expressional analysis, bioinformatics and targeted proteomics identify new potential therapeutic targets in glioblastoma stem cells. Oncotarget. 2015;6(28):26192–215. https://doi.org/10.18632/oncotarget.4613.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Young A, Berry R, Holloway AF, Blackburn NB, Dickinson JL, Skala M, et al. RNA-seq profiling of a radiation resistant and radiation sensitive prostate cancer cell line highlights opposing regulation of DNA repair and targets for radiosensitization. BMC Cancer. 2014;14:808. https://doi.org/10.1186/1471-2407-14-808.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Yu X, Liu Y, Yin L, Peng Y, Peng Y, Gao Y, et al. Radiation-promoted CDC6 protein stability contributes to radioresistance by regulating senescence and epithelial to mesenchymal transition. Oncogene. 2019;38(4):549–63. https://doi.org/10.1038/s41388-018-0460-4.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to X. Li.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interest.

Research involving human participants and/or animals

Not applicable.

Informed consent

Not applicable.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

12094_2020_2449_MOESM1_ESM.pdf

Figure S1 CDC6 protein level is elevated in high-grade GBM tumor samples compared with that in low-grade GBM tumor samples. (PDF 11102 kb)

12094_2020_2449_MOESM2_ESM.pdf

Figure S2 Time-dependent ROC curves for 1-year OS and D2P (A) and 3-year OS and D2P (B) of GBM samples for estimating the performance of the combination of age, radiation, temozolomide chemotherapy, CDC6, and CIMP status in prognosis predicting. AUC values were provided. TP, True positive. FP, False positive. Rad, Radiation. TMZ, temozolomide chemotherapy. (PDF 432 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, H., Zhou, X., Yuan, G. et al. CDC6 is up-regulated and a poor prognostic signature in glioblastoma multiforme. Clin Transl Oncol 23, 565–571 (2021). https://doi.org/10.1007/s12094-020-02449-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12094-020-02449-w

Keywords

Navigation