DOI QR코드

DOI QR Code

Regulatory Network of MicroRNAs, Target Genes, Transcription Factors and Host Genes in Endometrial Cancer

  • Xue, Lu-Chen (Department of Software Engineering, Jilin University) ;
  • Xu, Zhi-Wen (Department of Computer Science and Technology, Jilin University) ;
  • Wang, Kun-Hao (Department of Computer Science and Technology, Jilin University) ;
  • Wang, Ning (Department of Computer Science and Technology, Jilin University) ;
  • Zhang, Xiao-Xu (Department of Software Engineering, Jilin University) ;
  • Wang, Shang (Department of Computer Science and Technology, Jilin University)
  • Published : 2015.02.25

Abstract

Genes and microRNAs (miRNAs) have important roles in human oncology. However, most of the biological factors are reported in disperse form which makes it hard to discover the pathology. In this study, genes and miRNAs involved in human endometrial cancer(EC) were collected and formed into regulatory networks following their interactive relations, including miRNAs targeting genes, transcription factors (TFs) regulating miRNAs and miRNAs included in their host genes. Networks are constructed hierarchically at three levels: differentially expressed, related and global. Among the three, the differentially expressed network is the most important and fundamental network that contains the key genes and miRNAs in EC. The target genes, TFs and miRNAs are differentially expressed in EC so that any mutation in them may impact on EC development. Some key pathways in networks were highlighted to analyze how they interactively influence other factors and carcinogenesis. Upstream and downstream pathways of the differentially expressed genes and miRNAs were compared and analyzed. The purpose of this study was to partially reveal the deep regulatory mechanisms in EC using a new method that combines comprehensive genes and miRNAs together with their relationships. It may contribute to cancer prevention and gene therapy of EC.

Keywords

References

  1. Bae J, Won M, Kim DY, et al (2012). Identification of differentially expressed microRNAs in endometrial cancer cells after progesterone treatment. Int J Gynecol Cancer, 22, 561-5. https://doi.org/10.1097/IGC.0b013e31824927db
  2. Banno K, Yanokura M, Kisu I, et al (2013). MicroRNAs in endometrial cancer. Int J Clin Oncol, 18, 186-92. https://doi.org/10.1007/s10147-013-0526-9
  3. Baskerville S, Bartel DP (2005). Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes. RNA, 11, 241-7. https://doi.org/10.1261/rna.7240905
  4. Bell DW (2014). Novel genetic targets in endometrial cancer. Expert Opin Ther Targets, 18, 725-30. https://doi.org/10.1517/14728222.2014.909414
  5. Chekmenev DS, Haid C, Kel AE (2005). P-Match: transcription factor binding site search by combining patterns and weight matrices. Nucleic Acids Res, 33, 432-7. https://doi.org/10.1093/nar/gki441
  6. Cheung LWT (2012). High Frequency of PIK3R1 and PIK3R2 mutations in endometrial cancer elucidates a novel mechanism for regulation of PTENprotein stability. Cancer Discov, 2, 750-1. https://doi.org/10.1158/2159-8290.CD-12-0280
  7. Choi CH, Park YA, Choi JJ, et al (2012). Angiotensin II type I receptor and miR-155 in endometrial cancers: Synergistic antiproliferative effects of anti-miR-155 and losartan on endometrial cancer cells. Gynecol Oncol, 126, 124-31. https://doi.org/10.1016/j.ygyno.2012.04.020
  8. Cohn DE, Fabbri M, Valeri N, et al (2010). Comprehensive miRNA profiling of surgically staged endometrial cancer. Am J Obstet Gynecol, 202, 656.
  9. Dong PX, Kaneuchi M, Watari H, Sudo S, Sakuragi N (2014). MicroRNA-106b modulates epithelial-mesenchymal transition by targeting TWIST1 in invasive endometrial cancer cell lines. Mol Carcinog, 53, 349-59. https://doi.org/10.1002/mc.21983
  10. Fujita PA, Rhead B, Zweig AS, et al (2011). The UCSC Genome Browser database: update 2011. Nucleic Acids Res, 39, 876-82. https://doi.org/10.1093/nar/gkq963
  11. Hobert O (2008). Gene regulation by transcription factors and microRNAs. Science, 319, 1785-6. https://doi.org/10.1126/science.1151651
  12. Hsu SD, Tseng YT, Shrestha S, et al (2014). miRTarBase update 2014: an information resource for experimentally validated miRNA-target interactions. Nucleic Acids Res, 42, 78-85.
  13. Janiec-Jankowska A, Konopka B, Goluda C, Najmola U (2010). TP53 mutations in endometrial cancers relation to PTEN gene defects. Int J Gynecol Cancer, 20, 196-202. https://doi.org/10.1111/IGC.0b013e3181c83675
  14. Jiang QH, Wang YD, Hao YY, et al (2009). miR2Disease: a manually curated database for microRNA deregulation in human disease. Nucleic Acids Res, 37, 98-104.
  15. Kawaguchi M, Banno K, Yanokura M, et al (2009). Analysis of candidate target genes for mononucleotide repeat mutation in microsatellite instability-high (MSI-H) endometrial cancer. Int J Oncol, 35, 977-82.
  16. Kozomara A, Griffiths-Jones S (2011). miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res, 39, 152-7.
  17. Krakstad C, Birkeland E, Seidel D, et al (2012). High-throughput mutation profiling of pri-mary and metastatic endometrial cancers identifies KRAS, FGFR2 and PIK3CA to Be Frequently Mutated. PLoS One, 7, 52795. https://doi.org/10.1371/journal.pone.0052795
  18. Li B, Chen H, Shang LX, Gao W (2014). MicroRNA-543 suppresses endometrial cancer on-cogenicity via targeting FAK and TWIST1 expression. Arch Gynecol Obstet, 290, 533-41. https://doi.org/10.1007/s00404-014-3219-3
  19. Li BL, Lu C, Lu W, et al (2013). miR-130b is an EMTrelated microRNA that targets DIC-ER1 for aggression in endometrial cancer. Med Oncol, 30, 484. https://doi.org/10.1007/s12032-013-0484-0
  20. Li BL, Lu W, Lu C, et al (2013). CpG island hypermethylationassociated silencing of microRNAs promotes human endometrial cancer. Cancer Cell Int, 13, 44. https://doi.org/10.1186/1475-2867-13-44
  21. Li J, Xu ZW, Wang KH, et al (2013). Networks of microRNAs and genes in retinoblastomas. Asian Pac J Cancer Prev, 14, 6631-6. https://doi.org/10.7314/APJCP.2013.14.11.6631
  22. Lv SQ, Kim YH, Giulio F, et al (2012). Genetic Alterations in MicroRNAs in medulloblastomas. Brain Pathol, 22, 230-9. https://doi.org/10.1111/j.1750-3639.2011.00523.x
  23. Mackay HJ, Eisenhauer EA, Kamel-Reid S, et al (2014). Molecular determinants of outcome with mammalian target of rapamycin inhibition in endometrial cancer. Cancer, 120, 603-10. https://doi.org/10.1002/cncr.28414
  24. Myatt SS, Wang J, Monteiro LJ, et al (2010). Definition of microRNAs that repress expres-sion of the tumor suppressor gene FOXO1 in endometrial cancer. Cancer Res, 70, 367-77. https://doi.org/10.1158/0008-5472.CAN-09-1891
  25. Papadopoulos GL, Reczko M, Simossis VA, Sethupathy P, Hatzigeorgiou AG (2009). The database of experimentally supported targets: a functional update of TarBase. Nucleic Acids Res, 37, 155-8. https://doi.org/10.1093/nar/gkn809
  26. Price JC, Pollock LM, Rudd ML, et al (2013). Sequencing of candidate chromosome insta-bility genes in endometrial cancers cancers reveals somatic mutations in ESCO1, CHTF18, and MRE11A. PLoS One, 8, 63313. https://doi.org/10.1371/journal.pone.0063313
  27. Qin XY, Yan L, Zhao XB, Li CY, Fu YB (2012). microRNA-21 overexpression contributes to cell proliferation by targeting PTEN in endometrioid endometrial cancer. Oncol Lett, 4, 1290-6.
  28. Ramon LA, Braza-Boils A, Gilabert J, et al (2012). microRNAs related to angiogenesis are dysregulated in endometrioid endometrial cancer. Hum Reprod, 27, 3036-45. https://doi.org/10.1093/humrep/des292
  29. Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A (2004). Identification of mammalian microRNA host genes and transcription units. Genome Res, 14, 1902-10. https://doi.org/10.1101/gr.2722704
  30. Samulak D, Grosman-Dziewiszek P, Michalska MM, et al (2014). Evaluation of expression of the PTEN gene, oestrogen and progesterone receptors as diagnostic and predictive factors in endometrial cancer. Pathol Oncol Res, 20, 191-6. https://doi.org/10.1007/s12253-013-9684-3
  31. Shang C, Lu YM, Meng LR (2012). MicroRNA-125b downregulation mediates endometrial cancer invasion by targeting ERBB2. Med Sci Monit, 18, 149-55.
  32. Thanapprapasr D, Thanapprapasr K (2013). Molecular therapy as a future strategy in endometrial cancer. Asian Pac J Cancer Prev, 14, 3419-23. https://doi.org/10.7314/APJCP.2013.14.6.3419
  33. Torres A, Torres K, Wdowiak P, Paszkowski T, Maciejewski R (2013). Selection and validation of endogenous controls for microRNA expression studies in endometrioid endometrial cancer tissues. Gynecol Oncol, 130, 588-94. https://doi.org/10.1016/j.ygyno.2013.06.026
  34. Tran DH, Satou K, Ho TB, Pham TH (2010). Computational discovery of miR-TF regulatory modules in human genome. Bioinformation, 4, 371-7. https://doi.org/10.6026/97320630004371
  35. Tsuruta T, Kozaki K, Uesugi A, et al (2011). miR-152 Is a tumor suppressor microRNA That is silenced by DNA hypermethylation in endometrial cancer. Cancer Res, 71, 6450-62. https://doi.org/10.1158/0008-5472.CAN-11-0364
  36. Wang HY, Shen J, Jiang CP, Liu BR (2014). How to explain the contradiction of microRNA 200c expression and survival in solid tumors?: a meta-analysis. Asian Pac J Cancer Prev, 15, 3687-90. https://doi.org/10.7314/APJCP.2014.15.8.3687
  37. Wang J, Lu M, Qiu CX, Cui QH (2010). TransmiR: a transcription factor-microRNA regulation database. Nucleic Acids Res, 38, 119-22.
  38. Wang N, Xu ZW, Wang KH, Zhu MH, Li Y (2014). Construction and analysis of regulatory genetic networks in cervical cancer based on involved microRNAs, target genes, transcription factors and host genes. Oncol Lett, 7, 1279-83.
  39. Wu D, Huang HJ, He CN, Wang KY (2013). MicroRNA-199a-3p regulates endometrial cancer cell proliferation by targeting mammalian target of rapamycin (mTOR). Int J Gynecol Cancer, 23, 1191-7. https://doi.org/10.1097/IGC.0b013e31829ea779
  40. Xiao FF, Zuo ZX, Cai GS, et al (2009). miRecords: an integrated resource for microRNA-target interactions. Nucleic Acids Res, 37, 105-10.
  41. Xu YY, Wu HJ, Ma HD, et al (2013). MicroRNA-503 suppresses proliferation and cell-cycle progression of endometrioid endometrial cancer by negatively regulating cyclin D1. Febs J, 280, 3768-79. https://doi.org/10.1111/febs.12365
  42. Ye WW, Xue JS, Zhang Q, et al (2014). MiR-449a functions as a tumor suppressor in endometrial cancer by targeting CDC25A. Oncol Rep, 32, 1193-9.
  43. Zhang GY, Hou XX, Li Y, Zhao M (2014). MiR-205 inhibits cell apoptosis by targeting phosphatase and tensin homolog deleted on chromosome ten in endometrial cancer ishikawa cells. BMC Cancer, 14, 440. https://doi.org/10.1186/1471-2407-14-440