Biopolym. Cell. 2021; 37(1):23-32.
Molecular Biomedicine
Methylation pattern of tumor-suppressor gene promoters as putative noninvasive diagnostic markers for prostate cancer
1Mankovska O. S., 2Korsakova A. S., 2Cherniavskyi K. R., 3Kononenko O. A., 3Stakhovskyy E. O., 4Bondarenko Yu. M., 1, 5Kashuba V. I., 1Gerashchenko G. V.
  1. Institute of Molecular Biology and Genetics, NAS of Ukraine
    150, Akademika Zabolotnoho Str., Kyiv, Ukraine, 03143
  2. Educational and Scientific Center "Institute of Biology and Medicine",
    Taras Shevchenko National University of Kyiv
    64/13, Volodymyrska Str., Kyiv, Ukraine, 01601
  3. National Cancer Institute
    33/43, Lomonosova Str., Kyiv, Ukraine, 03022
  4. State Institution «Institute of Urology of NAMS of Ukraine»
    9-a, Yu. Kotsubyns'koho Str., Kyiv, Ukraine, 04053
  5. Karolinska Institutet
    Stockholm SE-171 77, Sweden

Abstract

Aim. To assess the rate of promoter methylation of putative TSGs for PCa in tumor tissue and in urine of PCa patients for better understanding of regulation of gene expression upon the PCa development and to evaluate the possibility to use the data on TSGs’ methylation for the development of noninvasive PCa markers. Methods. A quantitative methyl-specific PCR (qMSP) was used for the analysis of a methylation rate in prostate tissues and cell lines, and an ordinary MSP was performed for the study of urine samples. Results. We found that the RASSF1A promoter demonstrated a higher methylation rate in the TMPRSS2:ERG fusion positive PCa. The methylation of NKX3.1, PTEN and RASSF1A in DNA from urine was more common for cancer patients than for healthy donors. The promoters of CDH1 and GDF15 were methylated more frequently in PCa patients, than in patients with inflammatory disease. Conclusions. The abovementioned five genes can form a panel for early non-invasive detection of PCa. This set can be combined with the detection of the TMPRSS2:ERG fusion transcript. More work should be done to understand the molecular mechanisms explaining the functional role of promoter methylation of the selected genes.
Keywords: gene promoter methylation, tumor suppressor genes, noninvasive diagnostics, prostate cancer

References

[1] Angeles AK, Bauer S, Ratz L, Klauck SM, Sültmann H. Genome-Based Classification and Therapy of Prostate Cancer. Diagnostics (Basel). 2018;8(3):62.
[2] Pentyala S, Whyard T, Pentyala S, Muller J, Pfail J, Parmar S, Helguero CG, Khan S. Prostate cancer markers: An update. Biomed Rep. 2016;4(3):263-268.
[3] Tolkach Y, Kristiansen G. The Heterogeneity of Prostate Cancer: A Practical Approach. Pathobiology. 2018;85(1-2):108-116.
[4] Wallis CJ, Nam RK. Prostate Cancer Genetics: A Review. EJIFCC. 2015;26(2):79-91.
[5] Chen YC, Tsao CM, Kuo CC, Yu MH, Lin YW, Yang CY, Li HJ, Yan MD, Wang TJ, Chou YC, Su HY. Quantitative DNA methylation analysis of selected genes in endometrial carcinogenesis. Taiwan J Obstet Gynecol. 2015;54(5):572-9.
[6] Olkhov-Mitsel E, Van der Kwast T, Kron KJ, Ozcelik H, Briollais L, Massey C, Recker F, Kwiatkowski M, Fleshner NE, Diamandis EP, Zlotta AR, Bapat B. Quantitative DNA methylation analysis of genes coding for kallikrein-related peptidases 6 and 10 as biomarkers for prostate cancer. Epigenetics. 2012;7(9):1037-45.
[7] Bakavicius A, Daniunaite K, Zukauskaite K, Barisiene M, Jarmalaite S, Jankevicius F. Urinary DNA methylation biomarkers for prediction of prostate cancer upgrading and upstaging. Clin Epigenetics. 2019;11(1):115.
[8] Larsen LK, Lind GE, Guldberg P, Dahl C. DNA-Methylation-Based Detection of Urological Cancer in Urine: Overview of Biomarkers and Considerations on Biomarker Design, Source of DNA, and Detection Technologies. Int J Mol Sci. 2019;20(11):2657.
[9] Gerashchenko GV, Mankovska OS, Dmitriev AA, Mevs LV, Rosenberg EE, Pikul MV, Marynychenko MV, Gryzodub OP, Stakhovsky EO, Kashuba VI. Expression of epithelial-mesenchymal transition-related genes in pros-tate tumours. Biopolym Cell. 2017; 33(5):335–35.
[10] Zhang J, Hu S, Li Y. KRT18 is correlated with the malignant status and acts as an oncogene in colorectal cancer. Biosci Rep. 2019;39(8):BSR20190884.
[11] Imtiaz H, Afroz S, Hossain MA, Bellah SF, Rahman MM, Kadir MS, Sultana R, Mazid MA, Rahman MM. Genetic polymorphisms in CDH1 and Exo1 genes elevate the prostate cancer risk in Bangladeshi population. Tumour Biol. 2019;41(3):1010428319830837.
[12] Satelli A, Li S. Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell Mol Life Sci. 2011;68(18):3033-46.
[13] Geybels MS, Fang M, Wright JL, Qu X, Bibikova M, Klotzle B, Fan JB, Feng Z, Ostrander EA, Nelson PS, Stanford JL. PTEN loss is associated with prostate cancer recurrence and alterations in tumor DNA methylation profiles. Oncotarget. 2017;8(48):84338-84348.
[14] Bhatia-Gaur R, Donjacour AA, Sciavolino PJ, Kim M, Desai N, Young P, Norton CR, Gridley T, Cardiff RD, Cunha GR, Abate-Shen C, Shen MM. Roles for Nkx3.1 in prostate development and cancer. Genes Dev. 1999;13(8):966-77.
[15] Dubois F, Bergot E, Zalcman G, Levallet G. RASSF1A, puppeteer of cellular homeostasis, fights tumorigenesis, and metastasis-an updated review. Cell Death Dis. 2019;10(12):928.
[16] Wischhusen J, Melero I, Fridman WH. Growth/Differentiation Factor-15 (GDF-15): From Biomarker to Novel Targetable Immune Checkpoint. Front Immunol. 2020;11:951.
[17] Mankovska O, Skrypnikova O, Panasenko G, Kononenko O, Vikarchuk M, Stakhovskyy E, Kashuba V. Detection of methylation of VIM, TMEFF2 and GDF15 in the urine of patient with bladder cancer in Ukrainian population. NaUKMA Res Pape Biol Ecol. 2016; 184: 23–9.
[18] Sambrook J, Fritsch EE, Maniatis T. Molecular Cloning: A Laboratory Manual 2nd Edition. Cold Spring Harbor Laboratory Press 1989. 625 p