DOI QR코드

DOI QR Code

Methylation of O6-Methyl Guanine Methyltransferase Gene Promoter in Meningiomas - Comparison between Tumor Grades I, II, and III

  • Larijani, Leila (Department of Biology, School of Basic Sciences, Science and Research Branch Islamic Azad University (IAU)) ;
  • Madjd, Zahra (Oncopathology Research Centre, Iran University of Medical Sciences) ;
  • Samadikuchaksaraei, Ali (Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences) ;
  • Younespour, Shima (Department of Epidemiology and biostatistics, School of Public Health, Tehran University of Medical Sciences) ;
  • Zham, Hanieh (Department of Pathology, Cancer Research Centre) ;
  • Rakhshan, Azadeh (Department of Pathology, Shahid Beheshti University of Medical Science) ;
  • Mohammadi, Foruzan (Department of Pathology, Masih Daneshvari Hospital, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Science) ;
  • Rahbari, Ali (Department of Pathology, Shahid Beheshti University of Medical Science) ;
  • Moradi, Afshin (Department of Pathology, Cancer Research Centre)
  • Published : 2014.01.15

Abstract

Background: Meningiomas are the second most common primary intracranial tumors after gliomas. Epigenetic biomarkers such as DNA methylation, which is found in many tumors and is thus important in tumorigenesis can help diagnose meningiomas and predict response to adjuvant chemotherapy. We investigated aberrant O6-methyl guanine methyltransferase (MGMT) methylation in meningiomas. Materials and Methods: Sixty-one patients were classified according to the WHO grading, and MGMT promoter methylation status was examined via the methylation-Specific PCR(MSP) method. Results: MGMT promoter methylation was found in 22.2% of grade I, 35% of grade I with atypical features, 36% of grade II, and 42.9% of grade III tumors. Conclusions: There was an increase, albeit not statistically significant, in MGMT methylation with a rise in the tumor grade. Higher methylation levels were also observed in the male gender.

Keywords

References

  1. Asioli S, Senetta R, Maldi E,et al (2007). "Benign" metastatic meningioma: clinico-pathological analysis of one case metastasising to the lung and overview on the concepts of either primitive or metastatic meningiomas of the lung. Virchows Arch, 450, 591-4. https://doi.org/10.1007/s00428-007-0392-9
  2. Baylin SB, Herman JG (2000). DNA hypermethylation in tumorigenesis: epigenetics joins genetics. Trends Genet 16, 168-74. https://doi.org/10.1016/S0168-9525(99)01971-X
  3. Bello MJ, Aminoso C, Lopez-Marin I, et al (2004). DNA methylation of multiple promoter-associated CpG islands in meningiomas: relationship with the allelic status at 1p and 22q. Acta Neuropathol, 108, 413-21. https://doi.org/10.1007/s00401-004-0911-6
  4. Bird A (2002). DNA methylation patterns and epigenetic memory. Genes Dev, 16, 6-21. https://doi.org/10.1101/gad.947102
  5. Bird AP (1986). CpG-rich islands and the function of DNA methylation. Nature, 321, 209-13. https://doi.org/10.1038/321209a0
  6. Brait M, Ford JG, Papaiahgari S, et al (2009). Association between lifestyle factors and CpG island methylation in a cancer-free population. Cancer Epidemiol Biomarkers Prev, 18, 2984-91. https://doi.org/10.1158/1055-9965.EPI-08-1245
  7. Brandes AA, Franceschi E, Tosoni A, et al (2008). MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol, 26, 2192-7. https://doi.org/10.1200/JCO.2007.14.8163
  8. Costa BM, Caeiro C, Guimaraes I, et al (2010). Prognostic value of MGMT promoter methylation in glioblastoma patients treated with temozolomide-based chemoradiation: a Portuguese multicentre study. Oncol Rep, 23, 1655-62.
  9. de Robles P, McIntyre J, Kalra S, et al (2008). Methylation status of MGMT gene promoter in meningiomas. Cancer Genet Cytogenet, 187, 25-7. https://doi.org/10.1016/j.cancergencyto.2008.07.006
  10. Dunn J, Baborie A, Alam F, et al (2009). Extent of MGMT promoter methylation correlates with outcome in glioblastomas given temozolomide and radiotherapy. Br J Cancer, 101, 124-31. https://doi.org/10.1038/sj.bjc.6605127
  11. Enokida H, Shiina H, Urakami S, et al (2005). Ethnic grouprelated differences in CpG hypermethylation of the GSTP1 gene promoter among African-American, Caucasian and Asian patients with prostate cancer. Int J Cancer, 116, 174-81. https://doi.org/10.1002/ijc.21017
  12. Esteller M, Garcia-Foncillas J, Andion E, et al (2000). Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med, 343, 1350-4. https://doi.org/10.1056/NEJM200011093431901
  13. Feinberg AP, Vogelstein B (1983). Hypomethylation of ras oncogenes in primary human cancers. Biochem Biophys Res Commun, 111, 47-54. https://doi.org/10.1016/S0006-291X(83)80115-6
  14. Figueroa BE, Quint DJ, McKeever PE, et al (1999). Extracranial metastatic meningioma. Br J Radiol, 72, 513-6. https://doi.org/10.1259/bjr.72.857.10505022
  15. Havik AB, Brandal P, Honne H, et al (2012). MGMT promoter methylation in gliomas-assessment by pyrosequencing and quantitative methylation-specific PCR. J Transl Med, 10, 36. https://doi.org/10.1186/1479-5876-10-36
  16. Hegi ME, Diserens AC, Godard S, et al (2004). Clinical trial substantiates the predictive value of O-6-methylguanine-DNA methyltransferase promoter methylation in glioblastoma patients treated with temozolomide. Clin Cancer Res, 10, 1871-4. https://doi.org/10.1158/1078-0432.CCR-03-0384
  17. Hegi ME, Diserens AC, Gorlia T, et al (2005). MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med, 352, 997-1003. https://doi.org/10.1056/NEJMoa043331
  18. Herfarth KK, Brent TP, Danam RP, et al (1999). A specific CpG methylation pattern of the MGMT promoter region associated with reduced MGMT expression in primary colorectal cancers. Mol Carcinog, 24, 90-8. https://doi.org/10.1002/(SICI)1098-2744(199902)24:2<90::AID-MC3>3.0.CO;2-B
  19. Hibi K, Sakata M, Yokomizo K, et al (2009). Methylation of the MGMT gene is frequently detected in advanced gastric carcinoma. Anticancer Res, 29, 5053-5.
  20. Jenuwein T, Allis CD (2001). Translating the histone code. Science, 293, 1074-80. https://doi.org/10.1126/science.1063127
  21. Kang GH, Lee HJ, Hwang KS, et al (2003). Aberrant CpG island hypermethylation of chronic gastritis, in relation to aging, gender, intestinal metaplasia, and chronic inflammation. Am J Pathol, 163, 1551-6. https://doi.org/10.1016/S0002-9440(10)63511-0
  22. Lai JC, Wu JY, Cheng YW, et al (2009). O6-Methylguanine-DNA methyltransferase hypermethylation modulated by 17betaestradiol in lung cancer cells. Anticancer Res, 29, 2535-40.
  23. Leng S, Bernauer AM, Hong C, et al (2011). The A/G allele of rs16906252 predicts for MGMT methylation and is selectively silenced in premalignant lesions from smokers and in lung adenocarcinomas. Clin Cancer Res, 17, 2014-23. https://doi.org/10.1158/1078-0432.CCR-10-3026
  24. Li E (2002). Chromatin modification and epigenetic reprogramming in mammalian development. Nat Rev Genet, 3, 662-73. https://doi.org/10.1038/nrg887
  25. Liu J, Morgan M, Hutchison K, et al (2010). A study of the influence of sex on genome wide methylation. PLoS One, 5, 10028. https://doi.org/10.1371/journal.pone.0010028
  26. Liu Y, Pang JC, Dong S, et al (2005). Aberrant CpG island hypermethylation profile is associated with atypical and anaplastic meningiomas. Hum Pathol, 36, 416-25. https://doi.org/10.1016/j.humpath.2005.02.006
  27. Louis DN, Ohgaki H, Wiestler OD, et al (2007). The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol, 114, 97-109. https://doi.org/10.1007/s00401-007-0243-4
  28. Ludlum DB (1990). DNA alkylation by the haloethylnitrosoureas: nature of modifications produced and their enzymatic repair or removal. Mutat Res, 233, 117-26. https://doi.org/10.1016/0027-5107(90)90156-X
  29. Majumdar S, Buckles E, Estrada J, et al (2011). Aberrant DNA methylation and prostate cancer. Curr Genomics, 12, 486-505. https://doi.org/10.2174/138920211797904061
  30. Moradi A, Semnani V, Djam H, et al (2008). Pathodiagnostic parameters for meningioma grading. J Clin Neurosci, 15, 1370-5. https://doi.org/10.1016/j.jocn.2007.12.005
  31. Nakano M, Tanaka T, Nakamura A, et al (2012). Multiple pulmonary metastases following total removal of a bilateral parasagittal meningioma with complete occlusion of the superior sagittal sinus: report of a Case. Case Report Neurol Med, 2012, 121470.
  32. Pegg AE, Dolan ME, Moschel RC (1995). Structure, function, and inhibition of O6-alkylguanine-DNA alkyltransferase. Prog Nucleic Acid Res Mol Biol, 51, 167-223. https://doi.org/10.1016/S0079-6603(08)60879-X
  33. Perry A, Gutmann DH, Reifenberger G (2004). Molecular pathogenesis of meningiomas. J Neurooncol, 70, 183-202. https://doi.org/10.1007/s11060-004-2749-0
  34. Ramakrishnamurthy TV, Murty AV, Purohit AK, et al (2002). Benign meningioma metastasizing through CSF pathways: a case report and review of literature. Neurol India, 50, 326-9.
  35. Rivera AL, Pelloski CE, Gilbert MR, et al (2010). MGMT promoter methylation is predictive of response to radiotherapy and prognostic in the absence of adjuvant alkylating chemotherapy for glioblastoma. Neuro Oncol 12, 116-21. https://doi.org/10.1093/neuonc/nop020
  36. Russo VEA, Martienssen RA, Riggs AD (1996). Epigenetic mechanisms of gene regulation. Cold Spring Harbor Laboratory Press, Plainview, N.Y.
  37. Sarter B, Long TI, Tsong WH, et al (2005). Sex differential in methylation patterns of selected genes in Singapore Chinese. Hum Genet, 117, 402-3. https://doi.org/10.1007/s00439-005-1317-9
  38. Sato F, Tsuchiya S, Meltzer SJ, et al (2011). MicroRNAs and epigenetics. FEBS J, 278, 1598-609. https://doi.org/10.1111/j.1742-4658.2011.08089.x
  39. Sciuscio D, Diserens AC, van Dommelen K,et al (2011). Extent and patterns of MGMT promoter methylation in glioblastoma-and respective glioblastoma-derived spheres. Clin Cancer Res, 17, 255-66. https://doi.org/10.1158/1078-0432.CCR-10-1931
  40. Sharma S, Salehi F, Scheithauer BW, et al (2009). Role of MGMT in tumor development, progression, diagnosis, treatment and prognosis. Anticancer Res, 29, 3759-68.
  41. Tang K, Jin Q, Yan W, et al (2012). Clinical correlation of MGMT protein expression and promoter methylation in Chinese glioblastoma patients. Med Oncol, 29, 1292-6. https://doi.org/10.1007/s12032-011-9901-4
  42. Wallace K, Grau MV, Levine AJ, et al (2010). Association between folate levels and CpG Island hypermethylation in normal colorectal mucosa. Cancer Prev Res (Phila), 3, 1552-64. https://doi.org/10.1158/1940-6207.CAPR-10-0047
  43. Wu JY, Wang J, Lai JC, et al (2008). Association of O6-methylguanine-DNA methyltransferase (MGMT). promoter methylation with p53 mutation occurrence in non-small cell lung cancer with different histology, gender, and smoking status. Ann Surg Oncol, 15, 3272-7. https://doi.org/10.1245/s10434-008-0078-9

Cited by

  1. Meningeal Hemangiopericytomas and Meningomas: a Comparative Immunohistochemical and Genetic Study vol.16, pp.16, 2015, https://doi.org/10.7314/APJCP.2015.16.16.6871
  2. Temozolomide for recurrent meningiomas: a case-report with unexpected clinical and radiological response vol.127, pp.1, 2016, https://doi.org/10.1007/s11060-015-2016-6
  3. Global epigenetic profiling identifies methylation subgroups associated with recurrence-free survival in meningioma vol.133, pp.3, 2017, https://doi.org/10.1007/s00401-017-1678-x
  4. Comprehensive analysis of Iranian reports of pediatric central nervous system tumors vol.33, pp.9, 2017, https://doi.org/10.1007/s00381-017-3468-0
  5. DNA hypermethylation as a predictor of extramural vascular invasion (EMVI) in rectal cancer vol.19, pp.3, 2018, https://doi.org/10.1080/15384047.2017.1416933