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:

JMJD2 promotes acquired cisplatin resistance in non-small cell lung carcinoma cells

Abstract

Platinum-based drugs such as cisplatin (CP) are the first-line chemotherapy for non-small-cell lung carcinoma (NSCLC). Unfortunately, NSCLC has a low response rate to CP and acquired resistance always occurs. Histone methylation regulates chromatin structure and is implicated in DNA repair. We hypothesize histone methylation regulators are involved in CP resistance. We therefore screened gene expression of known histone methyltransferases and demethylases in three NSCLC cell lines with or without acquired resistance to CP. JMJD2s are a family of histone demethylases that remove tri-methyl groups from H3K9 and H3K36. We found expression of several JMJD2 family genes upregulated in CP-resistant cells, with JMJD2B expression being upregulated in all three CP-resistant NSCLC cell lines. Further analysis showed increased JMJD2 protein expression coincided with decreased H3K9me3 and H3K36me3. Chemical inhibitors of JMJD2-family proteins increased H3K9me3 and H3K36me3 levels and sensitized resistant cells to CP. Mechanistic studies showed that JMJD2 inhibition decreased chromatin association of ATR and Chk1 and inhibited the ATR-Chk1 replication checkpoint. Our results reveal that JMJD2 demethylases are potential therapeutic targets to overcome CP resistance in NSCLC.

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
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Aggarwal C, Borghaei H. Treatment paradigms for advanced non-small cell lung cancer at academic medical centers: involvement in clinical trial endpoint design. Oncologist. 2017;22:700–8.

    Article  Google Scholar 

  2. Johnson DH, Schiller JH, Bunn PA Jr. Recent clinical advances in lung cancer management. J Clin Oncol. 2014;32:973–82.

    Article  CAS  Google Scholar 

  3. Steuer CE, Behera M, Ernani V, Higgins KA, Saba NF, Shin DM, et al. Comparison of concurrent use of thoracic radiation with either carboplatin-paclitaxel or cisplatin-etoposide for patients with stage III non-small-cell lung cancer: a systematic review. JAMA Oncol. 2016;3:1120–9.

    Article  Google Scholar 

  4. Chang A. Chemotherapy, chemoresistance and the changing treatment landscape for NSCLC. Lung Cancer. 2011;71:3–10.

    Article  Google Scholar 

  5. Eastman A, Schulte N. Enhanced DNA repair as a mechanism of resistance to cis-diamminedichloroplatinum(II). Biochemistry. 1988;27:4730–4.

    Article  CAS  Google Scholar 

  6. Kelland LR, Barnard CF, Evans IG, Murrer BA, Theobald BR, Wyer SB, et al. Synthesis and in vitro and in vivo antitumor activity of a series of trans platinum antitumor complexes. J Med Chem. 1995;38:3016–24.

    Article  CAS  Google Scholar 

  7. Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, et al. Molecular mechanisms of cisplatin resistance. Oncogene. 2012;31:1869–83.

    Article  CAS  Google Scholar 

  8. Bhagwat N, Olsen AL, Wang AT, Hanada K, Stuckert P, Kanaar R, et al. XPF-ERCC1 participates in the Fanconi anemia pathway of cross-link repair. Mol Cell Biol. 2009;29:6427–37.

    Article  CAS  Google Scholar 

  9. Ferry KV, Hamilton TC, Johnson SW. Increased nucleotide excision repair in cisplatin-resistant ovarian cancer cells: role of ERCC1-XPF. Biochem Pharmacol. 2000;60:1305–13.

    Article  CAS  Google Scholar 

  10. Usanova S, Piee-Staffa A, Sied U, Thomale J, Schneider A, Kaina B, et al. Cisplatin sensitivity of testis tumour cells is due to deficiency in interstrand-crosslink repair and low ERCC1-XPF expression. Mol Cancer. 2010;9:248.

    Article  Google Scholar 

  11. Britten RA, Liu D, Tessier A, Hutchison MJ, Murray D. ERCC1 expression as a molecular marker of cisplatin resistance in human cervical tumor cells. Int J Cancer. 2000;89:453–7.

    Article  CAS  Google Scholar 

  12. Lord RV, Brabender J, Gandara D, Alberola V, Camps C, Domine M, et al. Low ERCC1 expression correlates with prolonged survival after cisplatin plus gemcitabine chemotherapy in non-small cell lung cancer. Clin Cancer Res. 2002;8:2286–91.

    CAS  PubMed  Google Scholar 

  13. Mendoza J, Martinez J, Hernandez C, Perez-Montiel D, Castro C, Fabian-Morales E, et al. Association between ERCC1 and XPA expression and polymorphisms and the response to cisplatin in testicular germ cell tumours. Br J Cancer. 2013;109:68–75.

    Article  CAS  Google Scholar 

  14. Enoiu M, Jiricny J, Scharer OD. Repair of cisplatin-induced DNA interstrand crosslinks by a replication-independent pathway involving transcription-coupled repair and translesion synthesis. Nucleic Acids Res. 2012;40:8953–64.

    Article  CAS  Google Scholar 

  15. Zhou W, Chen YW, Liu X, Chu P, Loria S, Wang Y, et al. Expression of DNA translesion synthesis polymerase eta in head and neck squamous cell cancer predicts resistance to gemcitabine and cisplatin-based chemotherapy. PLoS ONE. 2013;8:e83978.

    Article  Google Scholar 

  16. Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity. Nat Rev Mol Cell Biol. 2008;9:616–27.

    Article  CAS  Google Scholar 

  17. Kumagai A, Dunphy WG. How cells activate ATR. Cell Cycle. 2006;5:1265–8.

    Article  CAS  Google Scholar 

  18. Gadhikar MA, Sciuto MR, Alves MV, Pickering CR, Osman AA, Neskey DM, et al. Chk1/2 inhibition overcomes the cisplatin resistance of head and neck cancer cells secondary to the loss of functional p53. Mol Cancer Ther. 2013;12:1860–73.

    Article  CAS  Google Scholar 

  19. Sangster-Guity N, Conrad BH, Papadopoulos N, Bunz F. ATR mediates cisplatin resistance in a p53 genotype-specific manner. Oncogene. 2011;30:2526–33.

    Article  CAS  Google Scholar 

  20. Vendetti FP, Lau A, Schamus S, Conrads TP, O’Connor MJ, Bakkenist CJ. The orally active and bioavailable ATR kinase inhibitor AZD6738 potentiates the anti-tumor effects of cisplatin to resolve ATM-deficient non-small cell lung cancer in vivo. Oncotarget. 2015;6:44289–305.

    PubMed  PubMed Central  Google Scholar 

  21. Gursoy-Yuzugullu O, House N, Price BD. Patching broken DNA: nucleosome dynamics and the repair of DNA breaks. J Mol Biol. 2016;428:1846–60.

    Article  CAS  Google Scholar 

  22. Kuo LJ, Yang LX. Gamma-H2AX—a novel biomarker for DNA double-strand breaks. In Vivo. 2008;22:305–9.

    CAS  PubMed  Google Scholar 

  23. Turinetto V, Giachino C. Multiple facets of histone variant H2AX: a DNA double-strand-break marker with several biological functions. Nucleic Acids Res. 2015;43:2489–98.

    Article  CAS  Google Scholar 

  24. Hartlerode AJ, Guan Y, Rajendran A, Ura K, Schotta G, Xie A, et al. Impact of histone H4 lysine 20 methylation on 53BP1 responses to chromosomal double strand breaks. PLoS ONE. 2012;7:e49211.

    Article  CAS  Google Scholar 

  25. Hsiao KY, Mizzen CA. Histone H4 deacetylation facilitates 53BP1 DNA damage signaling and double-strand break repair. J Mol Cell Biol. 2013;5:157–65.

    Article  CAS  Google Scholar 

  26. Wakeman TP, Wang Q, Feng J, Wang XF. Bat3 facilitates H3K79 dimethylation by DOT1L and promotes DNA damage-induced 53BP1 foci at G1/G2 cell-cycle phases. EMBO J. 2012;31:2169–81.

    Article  CAS  Google Scholar 

  27. Sun Y, Jiang X, Xu Y, Ayrapetov MK, Moreau LA, Whetstine JR, et al. Histone H3 methylation links DNA damage detection to activation of the tumour suppressor Tip60. Nat Cell Biol. 2009;11:1376–82.

    Article  CAS  Google Scholar 

  28. Gong F, Clouaire T, Aguirrebengoa M, Legube G, Miller KM. Histone demethylase KDM5A regulates the ZMYND8-NuRD chromatin remodeler to promote DNA repair. J Cell Biol. 2017;216:1959–74.

    Article  CAS  Google Scholar 

  29. Hyun K, Jeon J, Park K, Kim J. Writing, erasing and reading histone lysine methylations. Exp Mol Med. 2017;49:e324.

    Article  CAS  Google Scholar 

  30. Kooistra SM, Helin K. Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol. 2012;13:297–311.

    Article  CAS  Google Scholar 

  31. Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011;21:381–95.

    Article  CAS  Google Scholar 

  32. Dinant C, Houtsmuller AB, Vermeulen W. Chromatin structure and DNA damage repair. Epigenetics Chromatin. 2008;1:9.

    Article  Google Scholar 

  33. Price BD, D’Andrea AD. Chromatin remodeling at DNA double-strand breaks. Cell. 2013;152:1344–54.

    Article  CAS  Google Scholar 

  34. Berry WL, Janknecht R. KDM4/JMJD2 histone demethylases: epigenetic regulators in cancer cells. Cancer Res. 2013;73:2936–42.

    Article  CAS  Google Scholar 

  35. D’Oto A, Tian QW, Davidoff AM, Yang J. Histone demethylases and their roles in cancer epigenetics. J Med Oncol Therapeut. 2016;1:34–40.

    Google Scholar 

  36. Agger K, Cloos PA, Christensen J, Pasini D, Rose S, Rappsilber J, et al. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature. 2007;449:731–4.

    Article  CAS  Google Scholar 

  37. Ezponda T, Dupere-Richer D, Will CM, Small EC, Varghese N, Patel T, et al. UTX/KDM6A loss enhances the malignant phenotype of multiple myeloma and sensitizes cells to EZH2 inhibition. Cell Rep. 2017;21:628–40.

    Article  CAS  Google Scholar 

  38. Agger K, Miyagi S, Pedersen MT, Kooistra SM, Johansen JV, Helin K. Jmjd2/Kdm4 demethylases are required for expression of Il3ra and survival of acute myeloid leukemia cells. Genes Dev. 2016;30:1278–88.

    Article  CAS  Google Scholar 

  39. Northcott PA, Nakahara Y, Wu X, Feuk L, Ellison DW, Croul S, et al. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat Genet. 2009;41:465–72.

    Article  CAS  Google Scholar 

  40. Toyokawa G, Taguchi K, Edagawa M, Shimamatsu S, Toyozawa R, Nosaki K, et al. The prognostic impact of Jumonji domain-containing 2B in patients with resected lung adenocarcinoma. Anticancer Res. 2016;36:4841–6.

    Article  CAS  Google Scholar 

  41. Zhang YW, Otterness DM, Chiang GG, Xie W, Liu YC, Mercurio F, et al. Genotoxic stress targets human Chk1 for degradation by the ubiquitin-proteasome pathway. Mol Cell. 2005;19:607–18.

    Article  CAS  Google Scholar 

  42. Hromas R, Williamson EA, Fnu S, Lee YJ, Park SJ, Beck BD, et al. Chk1 phosphorylation of Metnase enhances DNA repair but inhibits replication fork restart. Oncogene. 2012;31:4245–54.

    Article  CAS  Google Scholar 

  43. Petermann E, Woodcock M, Helleday T. Chk1 promotes replication fork progression by controlling replication initiation. Proc Natl Acad Sci USA. 2010;107:16090–5.

    Article  CAS  Google Scholar 

  44. Seiler JA, Conti C, Syed A, Aladjem MI, Pommier Y. The intra-S-phase checkpoint affects both DNA replication initiation and elongation: single-cell and -DNA fiber analyses. Mol Cell Biol. 2007;27:5806–18.

    Article  CAS  Google Scholar 

  45. Toledo LI, Altmeyer M, Rask MB, Lukas C, Larsen DH, Povlsen LK, et al. ATR prohibits replication catastrophe by preventing global exhaustion of RPA. Cell. 2013;155:1088–103.

    Article  CAS  Google Scholar 

  46. Mutreja K, Krietsch J, Hess J, Ursich S, Berti M, Roessler FK, et al. ATR-Mediated global fork slowing and reversal assist fork traverse and prevent chromosomal breakage at DNA interstrand cross-links. Cell Rep. 2018;24:2629–42 e2625.

    Article  CAS  Google Scholar 

  47. Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014;740:364–78.

    Article  CAS  Google Scholar 

  48. Basu A, Krishnamurthy S. Cellular responses to Cisplatin-induced DNA damage. J Nucleic Acids. 2010;2010:201367.

    PubMed  PubMed Central  Google Scholar 

  49. Nam EA, Cortez D. ATR signalling: more than meeting at the fork. Biochem J. 2011;436:527–36.

    Article  CAS  Google Scholar 

  50. Li CC, Yang JC, Lu MC, Lee CL, Peng CY, Hsu WY, et al. ATR-Chk1 signaling inhibition as a therapeutic strategy to enhance cisplatin chemosensitivity in urothelial bladder cancer. Oncotarget. 2016;7:1947–59.

    PubMed  Google Scholar 

  51. Mallette FA, Mattiroli F, Cui G, Young LC, Hendzel MJ, Mer G, et al. RNF8- and RNF168-dependent degradation of KDM4A/JMJD2A triggers 53BP1 recruitment to DNA damage sites. EMBO J. 2012;31:1865–78.

    Article  CAS  Google Scholar 

  52. Saksouk N, Simboeck E, Dejardin J. Constitutive heterochromatin formation and transcription in mammals. Epigenetics Chromatin. 2015;8:3.

    Article  CAS  Google Scholar 

  53. Zheng H, Chen L, Pledger WJ, Fang J, Chen J. p53 promotes repair of heterochromatin DNA by regulating JMJD2b and SUV39H1 expression. Oncogene. 2014;33:734–44.

    Article  CAS  Google Scholar 

  54. Andrade-Lima LC, Andrade LN, Menck CF. ATR suppresses apoptosis after UVB irradiation by controlling both translesion synthesis and alternative tolerance pathways. J Cell Sci. 2015;128:150–9.

    Article  CAS  Google Scholar 

  55. Gagou ME, Zuazua-Villar P, Meuth M. Enhanced H2AX phosphorylation, DNA replication fork arrest, and cell death in the absence of Chk1. Mol Biol Cell. 2010;21:739–52.

    Article  CAS  Google Scholar 

  56. Sanjiv K, Hagenkort A, Calderon-Montano JM, Koolmeister T, Reaper PM, Mortusewicz O, et al. Cancer-specific synthetic lethality between ATR and CHK1 kinase activities. Cell Rep. 2016;14:298–309.

    Article  CAS  Google Scholar 

  57. Chastain PD 2nd, Brylawski BP, Zhou YC, Rao S, Chu H, Ibrahim JG, et al. DNA damage checkpoint responses in the S phase of synchronized diploid human fibroblasts. Photochem Photobiol. 2015;91:109–16.

    Article  CAS  Google Scholar 

  58. Steward O, Popovich PG, Dietrich WD, Kleitman N. Replication and reproducibility in spinal cord injury research. Exp Neurol. 2012;233:597–605.

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported in part by a grant from Swim Across America (to CGM). We would like to thank the flow cytometry core at the University of Illinois at Chicago for their assistance in helping us run the replication start assay.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carl G. Maki.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Duan, L., Perez, R.E., Chastain, P.D. et al. JMJD2 promotes acquired cisplatin resistance in non-small cell lung carcinoma cells. Oncogene 38, 5643–5657 (2019). https://doi.org/10.1038/s41388-019-0814-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-019-0814-6

This article is cited by

Search

Quick links