Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter December 11, 2013

Methylation of OPCML promoter in ovarian cancer tissues predicts poor patient survival

  • Feng Zhou , Guohua Tao , Xiang Chen , Wei Xie , Manhua Liu and Xingjian Cao EMAIL logo

Abstract

Background: Ovarian cancer is a lethal gynecological malignancy largely due to the lack of biomarkers for early detection and treatment options. Opioid binding protein/cell adhesion molecule-like gene (OPCML) has a tumor-suppressor function in ovarian cancer, and epigenetic inactivation of OPCML induces oncogenic transformation of human ovarian surface epithelial cells.

Methods: This study investigated OPCML promoter methylation levels in ovarian cancer tissues. A total of 30 normal ovarian, 85 benign ovarian tumor, and 102 ovarian cancer tissues were subjected to quantitative methylation-specific PCR analysis of OPCML methylation. Four ovarian cancer cell lines were cultured and treated with the DNA demethylation agent 5-aza-2′-deoxycytidine (5-AZA) for restoring OPCML expression.

Results: The data showed that 80 of 102 (78.4%) ovarian cancer tissues and 28 of 85 (32.9%) benign ovarian tumors had a methylated OPCML gene promoter. In contrast, there was no OPCML gene promoter methylation in any of the 30 normal ovarian samples. OPCML promoter methylation was significantly associated with an older age of the patients (p=0.022), an advanced pathological stage of ovarian cancer (p=0.023), and poor overall survival of ovarian cancer patients (p<0.001). Multivariate analysis data showed that pathological stage, age, and OPCML promoter methylation were all independent factors to predict overall survival of patients. Furthermore, 5-AZA was able to restore expression of OPCML mRNA and protein in ovarian cancer cell lines.

Conclusions: These data indicate that detection of OPCML gene promoter methylation could be a useful biomarker for predicting the prognosis of ovarian cancer patients and disease progression.


Corresponding author: Xingjian Cao, Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China, Phone: +86 513 85061082, E-mail:

References

1. Shigetomi H, Higashiura Y, Kajihara H, Kobayashi H. Targeted molecular therapies for ovarian cancer: an update and future perspectives (Review). Oncol Rep 2012;28:395–408.10.3892/or.2012.1833Search in Google Scholar

2. Mould T. An overview of current diagnosis and treatment in ovarian cancer. Int J Gynecol Cancer 2012;22:S2–4.10.1097/IGC.0b013e318251c8e3Search in Google Scholar

3. Bast RC Jr. Molecular approaches to personalizing management of ovarian cancer. Ann Oncol 2011;22:viii5–15.10.1093/annonc/mdr516Search in Google Scholar

4. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin 2012;62:10–29.10.3322/caac.20138Search in Google Scholar

5. Barnholtz-Sloan JS, Schwartz AG, Qureshi F, Jacques S, Malone J, Munkarah AR. Ovarian cancer: changes in patterns at diagnosis and relative survival over the last three decades. Am J Obstet Gynecol 2003;189:1120–7.10.1067/S0002-9378(03)00579-9Search in Google Scholar

6. Poumpouridou N, Kroupis C. Hereditary breast cancer: beyond BRCA genetic analysis; PALB2 emerges. Clin Chem Lab Med 2011;23:423–34.Search in Google Scholar

7. Tischkowitz M, Hamel N, Carvalho MA, Birrane G, Soni A, van Beers EH, et al. Pathogenicity of the BRCA1 missense variant M1775K is determined by the disruption of the BRCT phosphopeptide-binding pocket: a multi-modal approach. Eur J Hum Genet 2008;16:820–32.10.1038/ejhg.2008.13Search in Google Scholar PubMed PubMed Central

8. Bird A. Perceptions of epigenetics. Nature 2007;447:396–8.10.1038/nature05913Search in Google Scholar PubMed

9. Novak K. Epigenetics changes in cancer cells. Med Gen Med 2004;6:17.Search in Google Scholar

10. Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 2007;8:286–98.10.1038/nrg2005Search in Google Scholar PubMed

11. Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med 2003;349:2042–54.10.1056/NEJMra023075Search in Google Scholar PubMed

12. Bell J. Predicting disease using genomics. Nature 2004;429:453–6.10.1038/nature02624Search in Google Scholar PubMed

13. Press JZ, De Luca A, Boyd N, Young S, Troussard A, Ridge Y, et al. Ovarian carcinomas with genetic and epigenetic BRCA1 loss have distinct molecular abnormalities. BMC Cancer 2008;8:17.10.1186/1471-2407-8-17Search in Google Scholar PubMed PubMed Central

14. Balch C, Huang TH, Brown R, Nephew KP. The epigenetics of ovarian cancer drug resistance and resensitization. Am J Obstet Gynecol 2004;191:1552–72.10.1016/j.ajog.2004.05.025Search in Google Scholar PubMed

15. Staub J, Chien J, Pan Y, Qian X, Narita K, Aletti G, et al. Epigenetic silencing of HSulf-1 in ovarian cancer: implications in chemoresistance. Oncogene 2007;26:4969–78.10.1038/sj.onc.1210300Search in Google Scholar PubMed

16. Sellar GC, Watt KP, Rabiasz GJ, Stronach EA, Li L, Miller EP, et al. OPCML at 11q25 is epigenetically inactivated and has tumor-suppressor function in epithelial ovarian cancer. Nat Genet 2003;34:337–43.10.1038/ng1183Search in Google Scholar PubMed

17. Mei FC, Young TW, Liu J, Cheng X. RAS-mediated epigenetic inactivation of OPCML in oncogenic transformation of human ovarian surface epithelial cells. FASEB J 2006;20:497–9.10.1096/fj.05-4586fjeSearch in Google Scholar PubMed

18. Ntougkos E, Rush R, Scott D, Frankenberg T, Gabra H, Smyth JF, et al. The IgLON family in epithelial ovarian cancer: expression profiles and clinicopathologic correlates. Clin Cancer Res 2005;11:5764–8.10.1158/1078-0432.CCR-04-2388Search in Google Scholar PubMed

19. Teodoridis JM, Hall J, Marsh S, Kannall HD, Smyth C, Curto J, et al. CpG island methylation of DNA damage response genes in advanced ovarian cancer. Cancer Res 2005;65:8961–7.10.1158/0008-5472.CAN-05-1187Search in Google Scholar PubMed

20. Czekierdowski A, Czekierdowska S, Szymanski M, Wielgos M, Kaminski P, Kotarski J. Opioid-binding protein/cell adhesion molecule-like (OPCML) gene and promoter methylation status in women with ovarian cancer. Neuro Endocrinol Lett 2006;27:609–13.Search in Google Scholar

21. Chen H, Ye F, Zhang J, Lu W, Cheng Q, Xie X. Loss of OPCML expression and the correlation with CpG island methylation and LOH in ovarian serous carcinoma. Eur J Gynaecol Oncol 2007;28:464–7.Search in Google Scholar

22. Zhou F, Cao X, Liu M, Wang Y, Tao G. A study of the methylation status of opioid binding protein/cell adhesion molecule-like gene in ovarian cancer using nested methylation-specific polymerase chain reaction detection. Clin Lab 2011;57:421–4.Search in Google Scholar

23. Duarte-Pereira S, Paiva F, Costa VL, Ramalho-Carvalho J, Savva-Bordalo J, Rodrigues A, et al. Prognostic value of opioid binding protein/cell adhesion molecule-like promoter methylation in bladder carcinoma. Eur J Cancer 2011;47:1106–14.10.1016/j.ejca.2010.12.025Search in Google Scholar PubMed

24. Burger H, Foekens JA, Look MP, Meijer-van Gelder ME, Klijn JG, Wiemer EA, et al. RNA expression of breast cancer resistance protein, lung resistance-related protein, multidrug resistance-associated proteins 1 and 2, and multidrug resistance gene 1 in breast cancer: correlation with chemotherapeutic response. Clin Cancer Res 2003;9:827–36.Search in Google Scholar

25. Melichar B, Kroupis C. Cancer epigenomics: moving slowly, but at a steady pace from laboratory bench to clinical practice. Clin Chem Lab Med 2012;50:1699–701.10.1515/cclm-2012-0495Search in Google Scholar PubMed

26. Heichman KA, Warren JD. DNA methylation biomarkers and their utility for solid cancer diagnostics. Clin Chem Lab Med 2012;50:1707–21.Search in Google Scholar

27. Toraño EG, Petrus S, Fernandez AF, Fraga MF. Global DNA hypomethylation in cancer: review of validated methods and clinical significance. Clin Chem Lab Med 2012;50:1733–42.10.1515/cclm-2011-0902Search in Google Scholar PubMed

28. Borley J, Wilhelm-Benartzi C, Brown R, Ghaem-Maghami S. Does tumour biology determine surgical success in the treatment of epithelial ovarian cancer? A systematic literature review. Br J Cancer 2012;107:1069–74.10.1038/bjc.2012.376Search in Google Scholar PubMed PubMed Central

29. Gui T, Shen K. The epidermal growth factor receptor as a therapeutic target in epithelial ovarian cancer. Cancer Epidemiol 2012;36:490–6.10.1016/j.canep.2012.06.005Search in Google Scholar PubMed

30. Kim A, Ueda Y, Naka T, Enomoto T. Therapeutic strategies in epithelial ovarian cancer. J Exp Clin Cancer Res 2012; 31:14.10.1186/1756-9966-31-14Search in Google Scholar PubMed PubMed Central

31. Ng JS, Low JJ, Ilancheran A. Epithelial ovarian cancer. Best Pract Res Clin Obstet Gynaecol 2012;26:337–45.10.1016/j.bpobgyn.2011.12.005Search in Google Scholar PubMed

32. Diaz-Padilla I, Razak AR, Minig L, Bernardini MQ, Maria Del Campo J. Prognostic and predictive value of CA-125 in the primary treatment of epithelial ovarian cancer: potentials and pitfalls. Clin Transl Oncol 2012;14:15–20.10.1007/s12094-012-0756-8Search in Google Scholar PubMed

33. Montagnana M, Danese E, Giudici S, Franchi M, Guidi GC, Plebani M, et al. HE4 in ovarian cancer: from discovery to clinical application. Adv Clin Chem 2011;55:1–20.10.1016/B978-0-12-387042-1.00001-0Search in Google Scholar

34. Lenhard M, Stieber P, Hertlein L, Kirschenhofer A, Fürst S, Mayr D, et al. The diagnostic accuracy of two human epididymis protein 4 (HE4) testing systems in combination with CA125 in the differential diagnosis of ovarian masses. Clin Chem Lab Med 2011;49:2081–8.10.1515/CCLM.2011.709Search in Google Scholar PubMed

35. Reed JE, Dunn JR, du Plessis DG, Shaw EJ, Reeves P, Gee AL, et al. Expression of cellular adhesion molecule ‘OPCML’ is down-regulated in gliomas and other brain tumours. Neuropathol Appl Neurobiol 2007;33:77–85.10.1111/j.1365-2990.2006.00786.xSearch in Google Scholar PubMed

36. Tsou JA, Galler JS, Siegmund KD, Laird PW, Turla S, Cozen W, et al. Identification of a panel of sensitive and specific DNA methylation markers for lung adenocarcinoma. Mol Cancer 2007;6:70.10.1186/1476-4598-6-70Search in Google Scholar PubMed PubMed Central

37. Sriraksa R, Zeller C, El-Bahrawy MA, Dai W, Daduang J, Jearanaikoon P, et al. CpG-island methylation study of liver fluke-related cholangiocarcinoma. Br J Cancer 2011;104:1313–8.10.1038/bjc.2011.102Search in Google Scholar PubMed PubMed Central

38. Cui Y, Ying Y, van Hasselt A, Ng KM, Yu J, Zhang Q, et al. OPCML is a broad tumor suppressor for multiple carcinomas and lymphomas with frequently epigenetic inactivation. PLoS One 2008;3:e2990.10.1371/journal.pone.0002990Search in Google Scholar PubMed PubMed Central

39. Xu XC. Tumor-suppressive activity of retinoic acid receptor-beta in cancer. Cancer Lett 2007;253:14–24.10.1016/j.canlet.2006.11.019Search in Google Scholar PubMed PubMed Central

40. McKie AB, Vaughan S, Zanini E, Okon IS, Louis L, de Sousa C, et al. The OPCML tumor suppressor functions as a cell surface repressor-adaptor, negatively regulating receptor tyrosine kinases in epithelial ovarian cancer. Cancer Discov 2012;2:156–71.10.1158/2159-8290.CD-11-0256Search in Google Scholar PubMed PubMed Central

41. Wu SY, Sood AK. New roles opined for OPCML. Cancer Discov 2012;2:115–6.10.1158/2159-8290.CD-11-0356Search in Google Scholar PubMed PubMed Central

Received: 2013-9-5
Accepted: 2013-11-11
Published Online: 2013-12-11
Published in Print: 2014-5-1

©2014 by Walter de Gruyter Berlin/Boston

Downloaded on 30.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2013-0736/html
Scroll to top button