E2F6 is essential for cell viability in breast cancer cells during replication stress

E2F6 is essential for cell viability in breast cancer cells during replication stress

E2F6 is a member of the E2F family of transcription factors involved in regulation of a wide variety of genes through bothactivation and repression. E2F6 has been reported as overexpressed in breast cancers but whether or not this is important for tumordevelopment is unclear. We first checked E2F6 expression in tumor cDNAs and the protein level in a range of breast cancer cell lines.RNA interference-mediated depletion was then used to assess the importance of E2F6 expression in cell lines with regard to cell cycleprofile using fluorescence-activated cell sorting and a cell survival assay using (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT). The overexpression of E2F6 was confirmed in breast tumor cDNA samples and breast cancer cell lines. Depletion ofE2F6 in the breast cancer cells reduced cell viability in MCF-7, T-47D, and MDA-MB-231 cells. There was little effect in the nontumorbreast cell line MCF-10A. The deleterious effect on cancer cells was greater during replication stress, leading to an increase in theproportion of breast cancer cells with sub-G1 DNA content. These results suggest that E2F6 might be essential for the survival of breastcancer cells experiencing replication stress, and therefore it could be a target for combined therapy

___

  • Attwooll CE, Denchi L, Helin K (2004). The E2F family: specific functions and overlapping interests. EMBO Journal 23 (24): 4709-4716.
  • Attwooll C, Oddi S, Cartwright P, Prosperini E, Agger K et al. (2005). A novel repressive E2F6 complex containing the polycomb group protein, EPC1, that interacts with EZH2 in a proliferation-specific manner. Journal of Biological Chemistry 280 (2): 1199-1208.
  • Bertoli C, Herlihy AE, Pennycook BR, Kriston-Vizi J, de Bruin RAM (2016). Sustained E2F-dependent transcription is a key mechanism to prevent replication-stress-induced DNA damage. Cell Reports 15 (7): 1412-1422.
  • Bertoli C, Klier S, McGowan C, Wittenberg C, de Bruin RA (2013). Chk1 inhibits E2F6 repressor function in response to replication stress to maintain cell-cycle transcription. Current Biology 23 (17): 629-637.
  • Bieda M, Xu X, Singer MA, Green R, Farnham PJ (2006). Unbiased location analysis of E2F1-binding sites suggests a widespread role for E2F1 in the human genome. Genome Research 16 (5): 595-605.
  • Bindra RS, Glazer PM (2007). Repression of RAD51 gene expression by E2F4/p130 complexes in hypoxia. Oncogene 26 (14): 2048- 2057.
  • Buttitta LA, Edgar BA (2007). Mechanisms controlling cell cycle exit upon terminal differentiation. Current Opinion in Cell Biology 19 (6): 697-704.
  • Cam H, Balciunaite E, Blais A, Spektor A, Scarpulla RC et al. (2004). A common set of gene regulatory networks links metabolism and growth inhibition. Molecular Cell 16 (3): 399-411.
  • Cartwright P, Muller H, Wagener C, Holm K, Helin K (1998). E2F6: a novel member of the E2F family is an inhibitor of E2Fdependent transcription. Oncogene 17 (5): 611-623.
  • Chen HZ, Tsai SY, Leone G (2009). Emerging roles of E2Fs in cancer: an exit from cell cycle control. Nature Reviews Cancer 9 (11): 785-797.
  • Cheng FH, Lin HY, Hwang TW, Chen YC, Huang RL et al. (2019). E2F6 functions as a competing endogenous RNA, and transcriptional repressor, to promote ovarian cancer stemness. Cancer Science 110: 1085-1095.
  • Chong JL, Wenzel PL, Saenz-Robles MT, Nair V, Ferrey A et al. (2009). E2F1-3 switch from activators in progenitor cells to repressors in differentiating cells. Nature 462 (7275): 930-934.
  • Danielian PS, Friesenhahn LB, Faust AM, West JC, Caron AM et al. (2008). E2f3a and E2f3b make overlapping but different contributions to total E2f3 activity. Oncogene 27 (51): 6561- 6570.
  • de Bruin A, Maiti B, Jakoi L, Timmers C, Buerki R et al. (2003). Identification and characterization of E2F7, a novel mammalian E2F family member capable of blocking cellular proliferation. Journal of Biological Chemistry 278 (43): 42041-42049.
  • De Meyer T, Bijsmans IT, Vande Vijver KK, Bekaert S, Oosting J et al. (2009). E2Fs mediate a fundamental cell-cycle deregulation in high-grade serous ovarian carcinomas. Journal of Pathology 217: 14-20.
  • Dimova DK, Dyson NJ (2005). The E2F transcriptional network: old acquaintances with new faces. Oncogene 24 (17): 2810-2826.
  • Di Stefano L, Jensen MR, Helin K (2003). E2F7, a novel E2F featuring DP-independent repression of a subset of E2F-regulated genes. EMBO Journal 22 (23): 6289-6298.
  • Endo-Munoz L, Dahler A, Teakle N, Rickwood D, Hazar-Rethinam M et al. (2009). E2F7 can regulate proliferation, differentiation, and apoptotic responses in human keratinocytes: implications for cutaneous squamous cell carcinoma formation. Cancer Research 69 (5): 1800-1808.
  • Fang ZH, Han ZC (2006). The transcription factor E2F: a crucial switch in the control of homeostasis and tumorigenesis. Histology and Histopathology 21 (4): 403-413.
  • Gaubatz S, Lindeman GJ, Ishida S, Jakoi L, Nevins JR et al. (2000). E2F4 and E2F5 play an essential role in pocket proteinmediated G1 control. Molecular Cell 6 (3): 729-735. doi: 10.1016/S1097-2765(00)00071-X
  • Gaubatz S, Wood JG, Livingston DM (1998). Unusual proliferation arrest and transcriptional control properties of a newly discovered E2F family member, E2F-6. Proceedings of the National Academy of Sciences of the United States of America 95 (16): 9190-9195.
  • Giangrande PH, Zhu W, Schlisio S, Sun X, Mori S et al. (2004). A role for E2F6 in distinguishing G1/S- and G2/M-specific transcription. Genes and Development 18 (23): 2941-2951.
  • Holliday DL, Speirs V (2011). Choosing the right cell line for breast cancer research. Breast Cancer Research 13 (7): 215.
  • Johnson J, Thijssen B, McDermott U, Garnett M, Wessels LF et al. (2016). Targeting the RB-E2F pathway in breast cancer. Oncogene 35 (37): 4829-4835.
  • Kao J, Salari K, Bocanegra M, Choi YL, Girard L et al. (2009). Molecular profiling of breast cancer cell lines defines relevant tumor models and provides a resource for cancer gene discovery. PLoS One 4: e6146.
  • Kherrouche Z, De Launoit Y, Monte D (2004). Human E2F6 is alternatively spliced to generate multiple protein isoforms. Biochemical and Biophysical Research Communications 317 (3): 749-760.
  • Lafta IJ (2016). STAG3 gene expression in breast cancer cells. PhD, University of Sheffield, Sheffield, UK.
  • Lee BK, Bhinge AA, Iyer VR (2011). Wide-ranging functions of E2F4 in transcriptional activation and repression revealed by genome-wide analysis. Nucleic Acids Research 39 (9): 3558- 3573.
  • Li Y, Choi PS, Felsher DW (2014). Oncogene addiction: resetting the safety switch? Oncotarget 5 (18): 7986-7987.
  • Li Y, Huang J, Yang D, Xiang S, Sun J et al. (2018). Expression patterns of E2F transcription factors and their potential prognostic roles in breast cancer. Oncology Letters 15: 9216-9230.
  • Livak KJ, Schmittgen TD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2–∆∆C T method. Methods 25 (4): 402-408.
  • Lu KH, Patterson AP, Wang L, Marquez RT, Atkinson EN et al. (2004). Selection of potential markers for epithelial ovarian cancer with gene expression arrays and recursive descent partition analysis. Clinical Cancer Research 10: 3291-3300. Macheret M, Halazonetis TD (2015). DNA replication stress as a hallmark of cancer. Annual Review of Pathology 10: 425-448.
  • McClellan KA, Slack RS (2007). Specific in vivo roles for E2Fs in differentiation and development. Cell Cycle 6 (23): 2917-2927.
  • Morkel M, Wenkel J, Bannister AJ, Kouzarides T, Hagemeir C (1997). An E2F-like repressor of transcription. Nature 390 (6660): 567- 568.
  • Muller H, Bracken AP, Vernell R, Moroni MC, Christians F et al. (2001). E2Fs regulate the expression of genes involved in differentiation, development, proliferation, and apoptosis. Genes and Development 15 (3): 267-285.
  • Oberley MJ, Inman DR, Farnham PJ (2003). E2F6 negatively regulates BRCA1 in human cancer cells without methylation of histone H3 on lysine 9. Journal of Biological Chemistry 278 (43): 42466-42476.
  • Ogawa H, Ishiguro KI, Gaubatz S, Livingston DM, Nakatani Y (2002). A complex with chromatin modifiers that occupies E2F- and Myc-responsive genes in G0 cells. Science 296 (5570): 1132-1136.
  • Palacios J, Honrado E, Osorio A, Cazorla A, Sarrio D et al. (2005). Phenotypic characterization of BRCA1 and BRCA2 tumors based in a tissue microarray study with 37 immunohistochemical markers. Breast Cancer Research and Treatment 90 (1): 5-14.
  • Polanowska J, Le Cam L, Orsetti B, Valles H, Fabbrizio E et al. (2000). Human E2F5 gene is oncogenic in primary rodent cells and is amplified in human breast tumors. Genes, Chromosomes and Cancer 28 (1): 126-130.
  • Reimer D, Sadr S, Wiedemair A, Goebel G, Concin N et al. (2006). Expression of the E2F family of transcription factors and its clinical relevance in ovarian cancer. Annals of the New York Academy of Sciences 1091: 270-281.
  • Tang H, Liu P, Yang L, Xie X, Ye F et al. (2014). miR-185 suppresses tumor proliferation by directly targeting E2F6 and DNMT1 and indirectly upregulating BRCA1 in triple-negative breast cancer. Molecular Cancer Therapeutics 13 (12): 3185-3197.
  • Thangavelu PU, Lin CY, Vaidyanathan S, Nguyen THM, Dray E et al. (2017). Overexpression of the E2F target gene CENPI promotes chromosome instability and predicts poor prognosis in estrogen receptor-positive breast cancer. Oncotarget 8 (37): 62167-62182.
  • Thomassen M, Tan Q, Kruse TA (2008). Gene expression metaanalysis identifies metastatic pathways and transcription factors in breast cancer. BMC Cancer 8: 394.
  • Trimarchi JM, Fairchild B, Verona R, Moberg K, Andon N et al. (1998). E2F-6, a member of the E2F family that can behave as a transcriptional repressor. Proceedings of the National Academy of Sciences of the United States of America 95 (6): 2850-2855.
  • Trimarchi JM, Fairchild B, Wen J, Lees JA (2001). The E2F6 transcription factor is a component of the mammalian Bmi1- containing polycomb complex. Proceedings of the National Academy of Sciences of the United States of America 98 (4): 1519-1524.
  • Trimarchi JM, Lees JA (2002). Sibling rivalry in the E2F family. Nature Reviews Molecular Cell Biology 3 (1): 11-20.
  • Tsai SY, Opavsky R, Sharma N, Wu L, Naidu S et al. (2008). Mouse development with a single E2F activator. Nature 454 (7208): 1137-1141.
  • Umemura S, Shirane M, Takekoshi S, Kusakabe T, Itoh J et al. (2009). Overexpression of E2F-5 correlates with a pathological basal phenotype and a worse clinical outcome. British Journal of Cancer 100 (5): 764-771.
  • Weijts BG, Bakker WJ, Cornelissen PW, Liang KH, Schaftenaar FH et al. (2012). E2F7 and E2F8 promote angiogenesis through transcriptional activation of VEGFA in cooperation with HIF1. EMBO Journal 31 (19): 3871-3884.
  • Willman CL, Ra H (2006). Genomic Alterations and Chromosomal Aberrations in Human Cancer. In: Hong WK (editor). Cancer Medicine, 7. London, UK: BC Decker Inc., pp. 104-154.
  • Xu X, Bieda M, Jin VX, Rabinovich A, Oberley MJ et al. (2007). A comprehensive ChIP-chip analysis of E2F1, E2F4, and E2F6 in normal and tumor cells reveals interchangeable roles of E2F family members. Genome Research 17 (11): 1550-1561.
  • Zalmas LP, Zhao X, Graham AL, Fisher R, Reilly C et al. (2008). DNA-damage response control of E2F7 and E2F8. EMBO Reports 9 (3): 252-259.