Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

ERCC6L that is up-regulated in high grade of renal cell carcinoma enhances cell viability in vitro and promotes tumor growth in vivo potentially through modulating MAPK signalling pathway

A Correction to this article was published on 28 January 2022

This article has been updated

Abstract

Renal cell carcinoma (RCC), which is one of the most diagnosed urological malignancies worldwide, is usually associated with abnormality in both genetic and cellular processes. In the present study, through analyzing The Cancer Genome Atlas (TCGA) dataset, we screened out ERCC6L as a candidate gene that is potentially related to the development of RCC based on its increased expression in ccRCC tissues compared with normal kidney tissues as well as its possible relevance to cancer prognosis. Evidence indicates that ERCC6L is an indispensable component of mammalian cell mitosis, while it fails to disclose the role of ERCC6L in tumorigenesis. By using RT-PCR, it was confirmed that the mRNA expression of ERCC6L was upregulated in RCC tissues as compared to normal controls in 28 pared samples. In addition, the immunohistochemistry study in a tissue microarray (TMA) containing 150 ccRCC samples showed that the staining score of ERCC6L was positively correlated with the Fuhrman grade of cancers. Next, when the expression of ERCC6L was lowered by specific shRNA, the cell viability was significantly inhibited in 786-O and Caki-1 cells, while the apoptosis was induced accordingly. At the same time, RCC cells those were transfected with shRNA targeting to ERCC6L grew significantly slower than parental cells in immunodeficient mice. These results consistently suggest that ERCC6L may play a role in regulating the cell viability of RCC both in vitro and in vivo. Further, gene expression microarray analysis followed by the validating western blot after knocking down ERCC6L expression in 786-O cells highlighted the involvement of MAPK signaling pathway in regulation of ERCC6L on cellular process of RCC. In conclusion, the present study suggests a likely promoting role of ERCC6L on the development of RCC. Thus, further study to explore the potential utility of ERCC6L as a novel therapeutic target of RCC is clearly needed.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Change history

References

  1. Ferlay JSI, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, et al. GLOBOCAN 2012 v1.0, cancer incidence and mortality worldwide [Internet]. Lyon: International Agency for Research on Cancer; 2013. http://globocan.iarc.fr.

    Google Scholar 

  2. Xu Y, Chen X, Li Y. Ercc6l, a gene of SNF2 family, may play a role in the teratogenic action of alcohol. Toxicol Lett. 2005;157:233–9.

    Article  CAS  Google Scholar 

  3. Baumann C, Korner R, Hofmann K, Nigg EA. PICH, a centromere-associated SNF2 family ATPase, is regulated by Plk1 and required for the spindle checkpoint. Cell. 2007;128:101–14.

    Article  CAS  Google Scholar 

  4. Toyoshima-Morimoto F, Taniguchi E, Shinya N, Iwamatsu A, Nishida E. Polo-like kinase 1 phosphorylates cyclin B1 and targets it to the nucleus during prophase. Nature. 2001;410:215–20.

    Article  CAS  Google Scholar 

  5. Lane HA, Nigg EA. Antibody microinjection reveals an essential role for human polo-like kinase 1 (Plk1) in the functional maturation of mitotic centrosomes. J Cell Biol. 1996;135:1701–13.

    Article  CAS  Google Scholar 

  6. Lee K, Rhee K. PLK1 phosphorylation of pericentrin initiates centrosome maturation at the onset of mitosis. J Cell Biol. 2011;195:1093–101.

    Article  CAS  Google Scholar 

  7. Glover DM. Polo kinase and progression through M phase in Drosophila: a perspective from the spindle poles. Oncogene. 2005;24:230–7.

    Article  CAS  Google Scholar 

  8. Holtrich U, Wolf G, Brauninger A, Karn T, Bohme B, Rubsamen-Waigmann H, et al. Induction and down-regulation of PLK, a human serine/threonine kinase expressed in proliferating cells and tumors. Proc Natl Acad Sci USA. 1994;91:1736–40.

    Article  CAS  Google Scholar 

  9. Wolf G, Elez R, Doermer A, Holtrich U, Ackermann H, Stutte HJ, et al. Prognostic significance of polo-like kinase (PLK) expression in non-small cell lung cancer. Oncogene. 1997;14:543–9.

    Article  CAS  Google Scholar 

  10. Knecht R, Elez R, Oechler M, Solbach C, von Ilberg C, Strebhardt K. Prognostic significance of polo-like kinase (PLK) expression in squamous cell carcinomas of the head and neck. Cancer Res. 1999;59:2794–7.

    CAS  PubMed  Google Scholar 

  11. Takai N, Hamanaka R, Yoshimatsu J, Miyakawa I. Polo-like kinases (Plks) and cancer. Oncogene. 2005;24:287–91.

    Article  CAS  Google Scholar 

  12. Biebricher A, Hirano S, Enzlin JH, Wiechens N, Streicher WW, Huttner D, et al. PICH: a DNA translocase specially adapted for processing anaphase bridge DNA. Mol Cell. 2013;51:691–701.

    Article  CAS  Google Scholar 

  13. Leng M, Besusso D, Jung SY, Wang Y, Qin J. Targeting Plk1 to chromosome arms and regulating chromosome compaction by the PICH ATPase. Cold Sh Q B. 2008;7:1480–9.

    CAS  Google Scholar 

  14. Kurasawa Y, Yu-Lee LY. PICH and cotargeted Plk1 coordinately maintain prometaphase chromosome arm architecture. Mol Biol Cell. 2010;21:1188–99.

    Article  CAS  Google Scholar 

  15. Nie J, Liu L, Xing G, Zhang M, Wei R, Guo M, et al. CKIP-1 acts as a colonic tumor suppressor by repressing oncogenic Smurf1 synthesis and promoting Smurf1 autodegradation. Oncogene. 2014;33:3677–87.

    Article  CAS  Google Scholar 

  16. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell . 2011;144:646–74.

    Article  CAS  Google Scholar 

  17. Burotto M, Chiou VL, Lee JM, Kohn EC. The MAPK pathway across different malignancies: a new perspective. Ann NY Acad Sci. 2014;120:3446–56.

    CAS  Google Scholar 

  18. Duff JL, Monia BP, Berk BC. Mitogen-activated protein (MAP) kinase is regulated by the MAP kinase phosphatase (MKP-1) in vascular smooth muscle cells. Effect of actinomycin D and antisense oligonucleotides. J Biol Chem. 1995;270:7161–6.

    Article  CAS  Google Scholar 

  19. Chu Y, Solski PA, Khosravi-Far R, Der CJ, Kelly K. The mitogen-activated protein kinase phosphatases PAC1, MKP-1, and MKP-2 have unique substrate specificities and reduced activity in vivo toward the ERK2 sevenmaker mutation. J Biol Chem. 1996;271:6497–501.

    Article  CAS  Google Scholar 

  20. Camps M, Nichols A, Arkinstall S. Dual specificity phosphatases: a gene family for control of MAP kinase function. FASEB J. 2000;14:6–16.

    Article  CAS  Google Scholar 

  21. Slack DN, Seternes OM, Gabrielsen M, Keyse SM. Distinct binding determinants for ERK2/p38alpha and JNK map kinases mediate catalytic activation and substrate selectivity of map kinase phosphatase-1. J Biol Chem. 2001;276:16491–500.

    Article  CAS  Google Scholar 

  22. O’Donnell A, Yang S-H, Sharrocks AD. MAP kinase-mediated c-fos regulation relies on a histone acetylation relay switch. Mol Cell. 2008;29:780–5.

    Article  Google Scholar 

  23. Murphy LO, Smith S, Chen RH, Fingar DC, Blenis J. Molecular interpretation of ERK signal duration by immediate early gene products. Nat Cell Biol. 2002;4:556–64.

    Article  CAS  Google Scholar 

  24. Chalmers CJ, Gilley R, March HN, Balmanno K, Cook SJ. The duration of ERK 1/2 activity determines the activation of c-Fos and Fra-1 and the composition and quantitative transcriptional output of AP-1. Cell Signal. 2007;19:695–704.

    Article  CAS  Google Scholar 

  25. Pu SY, Yu Q, Wu H, Jiang JJ, Chen XQ, He YH, et al. ERCC6L, a DNA helicase, is involved in cell proliferation and associated with survival and progress in breast and kidney cancers. Oncotarget. 2017;8:42116–24.

    Article  Google Scholar 

  26. Chen Z, Gibson TB, Robinson F, Silvestro L, Pearson G, Xu B, et al. MAP kinases. Chem Rev. 2001;101:2449–76.

    Article  CAS  Google Scholar 

  27. Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001;410:37–40.

    Article  CAS  Google Scholar 

  28. Wada T, Penninger JM. Mitogen-activated protein kinases in apoptosis regulation. Oncogene. 2004;23:2838–49.

    Article  CAS  Google Scholar 

  29. Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K, et al. Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 2001;22:153–83.

    CAS  PubMed  Google Scholar 

  30. Treisman R. Regulation of transcription by MAP kinase cascades. Curr Opin Cell Biol. 1996;8:205–15.

    Article  CAS  Google Scholar 

  31. Hynes NE, MacDonald G. ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol. 2009;21:177–84.

    Article  CAS  Google Scholar 

  32. Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003;3:459–65.

    Article  CAS  Google Scholar 

  33. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr., Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546–58.

    Article  CAS  Google Scholar 

  34. Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116:855–67.

    Article  CAS  Google Scholar 

  35. Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3:11–22.

    Article  CAS  Google Scholar 

  36. Owens DM, Keyse SM. Differential regulation of MAP kinase signalling by dual-specificity protein phosphatases. Oncogene. 2007;26:3203–13.

    Article  CAS  Google Scholar 

  37. Keyse SM. Protein phosphatases and the regulation of mitogen-activated protein kinase signalling. Curr Opin Cell Biol. 2000;12:186–92.

    Article  CAS  Google Scholar 

  38. Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, et al. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov. 2006;5:835–44.

    Article  CAS  Google Scholar 

  39. Menzies AM, Long GV. Dabrafenib and trametinib, alone and in combination for BRAF-mutant metastatic melanoma. Clin Cancer Res. 2014;20:2035–43.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Funding

This study is partially supported by the National Natural Science Foundation of China (grant nos. 81372766 and 81572532), and the Liaoning “Climbing” scholarship.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yu Zeng.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

The original online version of this article was revised: The authors wish to notify the readers of the following change: In “Depletion of ERCC6L induces apoptosis” section, in the sentence “The percentages of apoptotic cells in the sh-ERCC6L and sh-Ctrl groups were 8.33% and 3.80% respectively (P < 0.01) and 9.29% and 3.94% respectively (P < 0.01) in the 786-O cells and Caki-1 cells (Fig. 2G)”; the percentages of apoptotic cell in the sh-ERCC6L and sh-Ctrl groups in the 786-0 cells should be “8.46% and 5.02%” and the P value should be “P < 0.05”. Figure 2G, left panel, the above two graphs of flow cytometry of 786-0 cells should be replaced by the correct graphs, and in the right panel, the left two columns representing apoptotic cells of 786-0 cells should be replaced by those representing the correct number of apoptotic cells of 786-0 cells. Finally, at the end of “Fig. 2” legend, “*P < 0.05” should be added before the last sentence “P < 0.01”.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, G., Yu, Z., Fu, S. et al. ERCC6L that is up-regulated in high grade of renal cell carcinoma enhances cell viability in vitro and promotes tumor growth in vivo potentially through modulating MAPK signalling pathway. Cancer Gene Ther 26, 323–333 (2019). https://doi.org/10.1038/s41417-018-0064-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41417-018-0064-8

This article is cited by

Search

Quick links