DOI QR코드

DOI QR Code

ATAD2 is Highly Expressed in Ovarian Carcinomas and Indicates Poor Prognosis

  • Wan, Wei-Na (Department of Ultrasound, First Affiliated Hospital of China Medical University) ;
  • Zhang, Yi-Xia (Department of Ultrasound, First Affiliated Hospital of China Medical University) ;
  • Wang, Xue-Mei (Department of Ultrasound, First Affiliated Hospital of China Medical University) ;
  • Liu, Yan-Jun (Department of Ultrasound, First Affiliated Hospital of China Medical University) ;
  • Zhang, Yu-Qin (Department of Ultrasound, First Affiliated Hospital of China Medical University) ;
  • Que, Yan-Hong (Department of Ultrasound, First Affiliated Hospital of China Medical University) ;
  • Zhao, Wen-Jing (Department of Ultrasound, First Affiliated Hospital of China Medical University)
  • 발행 : 2014.03.30

초록

The purpose of this study was to explore the expression of ATAD2 in ovarian tumor tissue as well as its relationship with degree of malignancy. Tumor tissue from 110 cases of ovarian cancer was collected in accordance with the Declaration of Helsinki for evaluation of ATAD2 expression iimmunohistochemistry, quantitative PCR (qPCR) and Western blotting. The correlation between the ATAD2 expression and and the prognosis of ovarian cancer was evaluated by Cox regression model. In addition, HO-8910 and OVCAR-3 cells were transfected with two siRNAs targeting ATAD2. Cell viability was evaluated with MTT assay, and cell migration by transwell migration assay. ATAD2 was shown to be highly expressed in 65.5% (72/110) of ovarian cancer cases, both at transcriptional and protein levels. Moreover, highly expression was positively correlated with degree of malignancy. Knock-down of ATAD2 in HO-8910 and OVCAR-3 cells was found to reduce cell migration. In addition, follow-up visits of the patients demonstrated that the 5-year survival rate was lower in patients with high expression of ATAD2. Our study suggested that ovarian tumor tissue may have highly expressed ATAD2, which is associated with tumor stage, omentum-metastasis, ascites and CA-125. Increased ATAD2 may play important roles in tumor proliferation and migration. ATAD2 could serve in particular as a prognostic marker and a therapeutic target for ovarian cancer.

키워드

참고문헌

  1. Bodoor K, Haddad Y, Alkhateeb A, et al (2014). DNA hypermethylation of cell cycle (p15 and p16) and apoptotic (p14, p53, DAPK and TMS1) genes in peripheral blood of leukemia patients. Asian Pac J Cancer Prev, 15, 75-84 https://doi.org/10.7314/APJCP.2014.15.1.75
  2. Caron C, Lestrat C, Marsal S, et al (2010). Functional characterization of ATAD2 as a new cancer/testis factor and a predictor of poor prognosis in breast and lung cancers. Oncogene, 29, 5171-81. https://doi.org/10.1038/onc.2010.259
  3. Chan KK, Wei N, Liu SS, et al (2008). Estrogen receptor subtypes in ovarian cancer: a clinical correlation. Obstet Gynecol, 111, 144-51. https://doi.org/10.1097/01.AOG.0000296715.07705.e9
  4. Ciro M, Prosperini E, Quarto M, et al (2009). ATAD2 is a novel cofactor for MYC, overexpressed and amplified in aggressive tumors. Cancer Res, 69, 8491-8. https://doi.org/10.1158/0008-5472.CAN-09-2131
  5. Coser KR, Chesnes J, Hur J, et al (2003). Global analysis of ligand sensitivity of estrogen inducible and suppressible genes in MCF7/BUS breast cancer cells by DNA microarray. Proc Natl Acad Sci USA, 100, 13994-9. https://doi.org/10.1073/pnas.2235866100
  6. Desmedt C, Piette F, Loi S, et al (2007). Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res, 13, 3207-14. https://doi.org/10.1158/1078-0432.CCR-06-2765
  7. Eeckhoute J, Carroll JS, Geistlinger TR, Torres-Arzayus MI, Brown M (2006). A cell-type-specific transcriptional network required for estrogen regulation of cyclin D1 and cell cycle progression in breast cancer. Genes Dev, 20, 2513-26. https://doi.org/10.1101/gad.1446006
  8. Foekens JA, Atkins D, Zhang Y, et al (2006). Multicenter validation of a gene expression-based prognostic signature in lymph node-negative primary breast cancer. J Clin Oncol, 24, 1665-71. https://doi.org/10.1200/JCO.2005.03.9115
  9. Gadducci A, Cosio S, Carpi A, Nicolini A, Genazzani AR (2004). Serum tumor markers in the management of ovarian, endometrial and cervical cancer. Biomed Pharmacother, 58, 24-38. https://doi.org/10.1016/j.biopha.2003.11.003
  10. Ghisletti S, Meda C, Maggi A, Vegeto E (2005). 17$\beta$-estradiol inhibits inflammatory gene expression by controlling NF- ${\kappa}B$ intracellular localization. Mol Cell Biol, 25, 2957-68. https://doi.org/10.1128/MCB.25.8.2957-2968.2005
  11. Haibe-Kains B, Desmedt C, Piette F, et al (2008). Comparison of prognostic gene expression signatures for breast cancer. BMC Genomics, 9, 394. https://doi.org/10.1186/1471-2164-9-394
  12. Horn LC, Beckmann MW, Beller A, et al (2010). Changes in the TNM classification of gynecological tumors. Pathologe, 31, 367-73. https://doi.org/10.1007/s00292-010-1308-z
  13. Hsia EY, Kalashnikova EV, Revenko AS, et al (2010). Deregulated E2F and the AAA+ coregulator ANCCA drive proto-oncogene ACTR/AIB1 overexpression in breast cancer. Mol Cancer Res, 8, 183-93. https://doi.org/10.1158/1541-7786.MCR-09-0095
  14. Hsia EY, Zou JX, Chen HW (2009). The roles and action mechanisms of p160/SRC coactivators and the ANCCA coregulator in cancer. Prog Mol Biol Transl Sci, 87, 261-98. https://doi.org/10.1016/S1877-1173(09)87008-7
  15. Ivshina AV, George J, Senko O, et al (2006). Genetic reclassification of histologic grade delineates new clinical subtypes of breast cancer. Cancer Res, 66, 10292-301. https://doi.org/10.1158/0008-5472.CAN-05-4414
  16. Jemal A, Murray T, Ward E, et al (2005). Cancer statistics. CA Cancer J Clin, 55, 10-30. https://doi.org/10.3322/canjclin.55.1.10
  17. Kalashnikova EV, Revenko AS, Gemo AT, et al (2010). ANCCA/ ATAD2 overexpression identifies breast cancer patients with poor prognosis, acting to drive proliferation and survival of triple-negative cells through control of B-Myb and EZH2. Cancer Res, 70, 9402-12. https://doi.org/10.1158/0008-5472.CAN-10-1199
  18. Kobayashi E, Ueda Y, Matsuzaki S, et al (2012). Biomarkers for screening, diagnosis, and monitoring of ovarian cancer. Cancer Epidemiol Biomarkers Prev, 21, 1902-12. https://doi.org/10.1158/1055-9965.EPI-12-0646
  19. Li J, Dowdy S, Tipton T, et al (2009). HE4 as a biomarker for ovarian and endometrial cancer management. Expert Rev Mol Diagn, 9, 555-66. https://doi.org/10.1586/erm.09.39
  20. Lutz AM, Willmann JK, Drescher CW, et al (2011). Early diagnosis of ovarian carcinoma: is a solution in sight? Radiology, 259, 329-45. https://doi.org/10.1148/radiol.11090563
  21. Ma XJ, Salunga R, Tuggle JT, et al (2003). Gene expression profiles of human breast cancer progression. Proc Natl Acad Sci U S A, 100, 5974-9. https://doi.org/10.1073/pnas.0931261100
  22. Ngwenya S, Safe S (2003). Cell context-dependent differences in the induction of E2F-1 gene expression by 17 beta-estradiol in MCF-7 and ZR-75 cells. Endocrinology, 144, 1675-85. https://doi.org/10.1210/en.2002-0009
  23. Nossov V, Amneus M, Su F, et al (2008). The early detection of ovarian cancer: from traditional methods to proteomics. Can we really do better than serum CA-125? Am J Obstet Gynecol, 199, 215-23. https://doi.org/10.1016/j.ajog.2008.04.009
  24. O'Lone R, Frith MC, Karlsson EK, Hansen U (2004). Genomic targets of nuclear estrogen receptors. Mol Endocrinol, 18, 1859-75. https://doi.org/10.1210/me.2003-0044
  25. Prajoko YW, Aryandono T (2014). Expression of nuclear factor kappa B (NF-${\kappa}B$) as a predictor of poor pathologic response to chemotherapy in patients with locally advanced breast cancer. Asian Pac J Cancer Prev, 15, 595-98 https://doi.org/10.7314/APJCP.2014.15.2.595
  26. Raeder MB, Birkeland E, Trovik J, et al (2013). Integrated genomic analysis of the 8q24 amplification in endometrial cancers identifies ATAD2 as essential to MYC-dependent cancers. PLoS One, 8, e54873. https://doi.org/10.1371/journal.pone.0054873
  27. Rao BR, Slotman BJ ( 1991). Endocrine factors in common epithelial ovarian cancer. Endocr Rev, 12, 14-26. https://doi.org/10.1210/edrv-12-1-14
  28. Revenko AS, Kalashnikova EV, Gemo AT, Zou JX, Chen HW (2010). Chromatin loading of E2F-MLL complex by cancerassociated coregulator ANCCA via reading a specific histone mark. Mol Cell Biol, 30, 5260-72. https://doi.org/10.1128/MCB.00484-10
  29. Sorensen SS, Mosgaard BJ (2011). Combination of cancer antigen 125 and carcinoembryonic antigen can improve ovarian cancer diagnosis. Dan Med Bull, 58, A4331.
  30. Teschendorff AE, Naderi A, Barbosa-Morais NL, et al (2006). A consensus prognostic gene expression classifier for ER positive breast cancer. Genome Biol, 7, R101. https://doi.org/10.1186/gb-2006-7-10-r101
  31. van 't Veer LJ, Dai H, van de Vijver MJ, et al (2002). Gene expression profiling predicts clinical outcome of breast cancer. Nature, 415, 530-6. https://doi.org/10.1038/415530a
  32. Visintin I, Feng Z, Longton G, et al (2008). Diagnostic markers for early detection of ovarian cancer. Clin Cancer Res, 14, 1065-72. https://doi.org/10.1158/1078-0432.CCR-07-1569
  33. Wang Y, Klijn JG, Zhang Y, et al (2005). Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet, 365, 671-9. https://doi.org/10.1016/S0140-6736(05)17947-1
  34. Wrzeszczynski KO, Varadan V, Byrnes J, et al (2011). Identification of tumor suppressors and oncogenes from genomic and epigenetic features in ovarian cancer. PLoS One, 6, e28503. https://doi.org/10.1371/journal.pone.0028503
  35. Zou JX, Guo L, Revenko AS, et al (2009). Androgen-induced coactivator ANCCA mediates specific androgen receptor signaling in prostate cancer. Cancer Res, 69, 3339-46. https://doi.org/10.1158/0008-5472.CAN-08-3440
  36. Zou JX, Revenko AS, Li LB, Gemo AT, Chen HW (2007). ANCCA, an estrogen-regulated AAA+ ATPase coactivator for ERalpha, is required for coregulator occupancy and chromatin modification. Proc Natl Acad Sci USA, 104, 18067-72. https://doi.org/10.1073/pnas.0705814104

피인용 문헌

  1. Impact of Prognostic Factors on Survival Rates in Patients with Ovarian Carcinoma vol.15, pp.15, 2014, https://doi.org/10.7314/APJCP.2014.15.15.6087
  2. Prognostic Value of Peritoneal Washing Cytology in Gynecologic Malignancies: a Controversial Issue vol.15, pp.21, 2014, https://doi.org/10.7314/APJCP.2014.15.21.9405
  3. Oncogene ATAD2 promotes cell proliferation, invasion and migration in cervical cancer vol.33, pp.5, 2015, https://doi.org/10.3892/or.2015.3867
  4. Overexpression of ANCCA/ATAD2 in endometrial carcinoma and its correlation with tumor progression and poor prognosis vol.36, pp.6, 2015, https://doi.org/10.1007/s13277-015-3089-8
  5. Atad2 is a generalist facilitator of chromatin dynamics in embryonic stem cells vol.8, pp.4, 2015, https://doi.org/10.1093/jmcb/mjv060
  6. Bromodomain inhibitors and cancer therapy: From structures to applications vol.12, pp.5, 2017, https://doi.org/10.1080/15592294.2016.1265710
  7. Biological function and histone recognition of family IV bromodomain-containing proteins vol.233, pp.3, 2017, https://doi.org/10.1002/jcp.26010
  8. HIF-1α promoted vasculogenic mimicry formation in hepatocellular carcinoma through LOXL2 up-regulation in hypoxic tumor microenvironment vol.36, pp.1, 2017, https://doi.org/10.1186/s13046-017-0533-1
  9. ATAD2 in cancer: a pharmacologically challenging but tractable target vol.22, pp.1, 2018, https://doi.org/10.1080/14728222.2018.1406921
  10. Identification of a novel ligand for the ATAD2 bromodomain with selectivity over BRD4 through a fragment growing approach vol.16, pp.11, 2018, https://doi.org/10.1039/C8OB00099A
  11. ATAD2 silencing decreases VEGFA secretion through targeting has-miR-520a to inhibit angiogenesis in colorectal cancer pp.1208-6002, 2018, https://doi.org/10.1139/bcb-2018-0081
  12. Disulfide bridge formation influences ligand recognition by the ATAD2 bromodomain vol.87, pp.2, 2018, https://doi.org/10.1002/prot.25636