DOI QR코드

DOI QR Code

Down-regulation of the cyclin E1 oncogene expression by microRNA-16-1 induces cell cycle arrest in human cancer cells

  • Wang, Fu (State Key Laboratory of Virology, College of Life Sciences, Wuhan University) ;
  • Fu, Xiang-Dong (Department of Cellular and Molecular Medicine, University of California) ;
  • Zhou, Yu (State Key Laboratory of Virology, College of Life Sciences, Wuhan University) ;
  • Zhang, Yi (State Key Laboratory of Virology, College of Life Sciences, Wuhan University)
  • Published : 2009.11.30

Abstract

Cyclin E1 (CCNE1), a positive regulator of the cell cycle, controls the transition of cells from G1 to S phase. In numerous human tumors, however, CCNE1 expression is frequently dysregulated, while the mechanism leading to its dysregulation remains incompletely defined. Herein, we showed that CCNE1 expression was subject to post-transcriptional regulation by a microRNA miR-16-1. This was evident at protein level of CCNE1 as well as its mRNA level. Further evident by dual luciferase reporter assay revealed that two evolutionary conserved binding sites on 3' UTR of CCNE1 were the direct functional target sites. Moreover, we showed that miR-16-1 induced G0/G1 cell cycle arrest by targeting CCNE1 and siRNA against CCNE1 partially phenocopied miR-16-1-induced cell cycle phenotype whereas substantially rescued anti-miR-16-1- induced phenotype. Together, all these results demonstrate that miR-16-1 plays a vital role in modulating cellular process in human cancers and indicate the therapeutic potential of miR-16-1 in cancer therapy.

Keywords

References

  1. Sauer, K. and Lehner, C. F. (1995) The role of cyclin E in the regulation of entry into S phase. Prog. Cell Cycle Res. 1, 125-139
  2. Arata, Y., Fujita, M., Ohtani, K., Kijima, S. and Kato, J. Y. (2000) Cdk2-dependent and independent pathways in E2Fmediated S phase induction. J. Biol. Chem. 275, 6337-6345 https://doi.org/10.1074/jbc.275.9.6337
  3. Ma, T., Van Tine, B. A., Wei, Y., Garrett, M. D., Nelson, D., Adams, P. D., Wang, J., Qin, J., Chow, L. T. and Harper, J. W. (2000) Cell cycle-regulated phosphorylation of p220 (NPAT) by cyclin E/Cdk2 in Cajal bodies promotes histone gene transcription. Genes & Dev. 14, 2298-2313 https://doi.org/10.1101/gad.829500
  4. Winey, M. (1999) Cell cycle: driving the centrosome cycle. Curr. Biol. 9, R449-452 https://doi.org/10.1016/S0960-9822(99)80279-6
  5. Ekholm, S. V. and Reed, S. I. (2000) Regulation of G(1) cyclin dependent kinases in the mammalian cell cycle. Curr. Opin. Cell Biol. 12, 676-684 https://doi.org/10.1016/S0955-0674(00)00151-4
  6. Donnellan, R. and Chetty, R. (1999) Cyclin E in human cancers. FASEB. J. 13, 773-780
  7. Sandhu, C., and Slingerland, J. (2000) Deregulation of the cell cycle in cancer. Cancer Detect Prev. 24, 107-118
  8. Tarik, M. and Christoph, G. (2004) Cyclin E. Int. J. Biochem. Cell Biol. 36, 1424-1439 https://doi.org/10.1016/j.biocel.2003.12.005
  9. Akama, Y., Yasui, W., Yokozaki, H., Kuniyasu, H., Kitahara, K., Ishikawa, T. and Tahara, E. (1995) Frequent amplification of the cyclin E gene in human gastric carcinomas. Jpn. J. Cancer Res. 86, 617-621 https://doi.org/10.1111/j.1349-7006.1995.tb02442.x
  10. Demetrick, D. J., Matsumoto, S., Hannon, G. J., Okamoto, K., Xiong, Y., Zhang, H. and Beach, D. H. (1995) Chromosomal mapping of the genes for the human cell cycle proteins cyclin C (CCNC), cyclin E (CCNE), p21 (CDKN1) and KAP (CDKN3) Cytogenet. Cell Genetics. 69, 190-192 https://doi.org/10.1159/000133960
  11. Botner, D. M., and Rosenberg, M. P. (1997) Induction of mammary gland hyperplasia and carcinomas in transgenic mice expressing human cyclin E. Mol. Cell Biol. 17, 453-459 https://doi.org/10.1128/MCB.17.1.453
  12. Bartel, D. P. (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281-297 https://doi.org/10.1016/S0092-8674(04)00045-5
  13. Ambros, V. (2004) The functions of animal microRNAs. Nature 431, 350-355 https://doi.org/10.1038/nature02871
  14. Cho, W. C. (2007) OncomiRs: the discovery and progress of microRNAs in cancers. Mol. Cancer 6, 60-67 https://doi.org/10.1186/1476-4598-6-60
  15. Gregory, R. I. and Shiekhattar, R. (2005) MicroRNA biogenesis and cancer. Cancer Re. 65, 3509-3512 https://doi.org/10.1158/0008-5472.CAN-05-0298
  16. Linsley, P. S., Schelter, J., Burchard, J., Kibukawa, M., Martin, M. M., Bartz, S. R., Johnson, J. M., Cummins, J. M., Raymond, C. K., Dai, H., Chau, N., Cleary, M., Jackson, A. L., Carleton, M. and Lim, L. (2007) Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol. Cell Biol. 27, 2240-2252 https://doi.org/10.1128/MCB.02005-06
  17. Liu, Q., Fu, H., Sun, F., Zhang, H., Tie, Y., Zhu, J., Xing, R., Sun, Z. and Zheng, X. (2008) miR-16 family induces cell cycle arrest by regulating multiple cell cycle genes. Nucleic. Acids. Res. 36, 5391-5404 https://doi.org/10.1093/nar/gkn522
  18. Bonci, D., Coppola, V., Musumeci, M., Addario, A., Giuffrida, R., Memeo, L., D'Urso, L., Pagliuca, A., Biffoni, M., Labbaye, C., Bartucci, M., Muto, G., Peschle, C. and De Maria, R. (2008) The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat. Med. 14, 1271-1277 https://doi.org/10.1038/nm.1880
  19. Lewis, B. P., Shih, I. H., Jones-Rhoades, M. W., Bartel, D. P. and Burge, C. B. (2003) Prediction of mammalian micro RNA targets. Cell 115, 787-798 https://doi.org/10.1016/S0092-8674(03)01018-3
  20. Lall, S., Grun, D., Krek, A., Chen, K., Wang, Y. L., Dewey, C. N., Sood, P., Colombo, T., Bray, N., Macmenamin, P., Kao, H. L., Gunsalus, K. C., Pachter, L., Piano, F. and Rajewsky, N. (2006) A genome-wide map of conserved microRNA targets in C. elegans. Curr. Biol., 16, 460-471 https://doi.org/10.1016/j.cub.2006.01.050
  21. Bino, J., Anton, J. E., Alexei, A., Thomas, T., Chris, S. and Debora, S. M. (2004) Human microRNA targets. PLoS Biol. 2, e363 https://doi.org/10.1371/journal.pbio.0020363
  22. Nam, S., Kim, B., Shin, S. and Lee, S. (2008) miRGator: an integrated system for functional annotation of micro RNAs. Nucleic. Acids. Res. 36, 159-164 https://doi.org/10.1093/nar/gkm829
  23. Calin, G. A., Dumitru, C. D., Shimizu, M., Bichi, R., Zupo, S., Noch, E., Aldler, H., Rattan, S., Keating, M., Rai, K., Rassenti, L., Kipps, T., Negrini, M., Bullrich, F. and Croce, C. M. (2002) Frequent deletions and downregulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. U.S.A. 99, 15524-15529 https://doi.org/10.1073/pnas.242606799
  24. Bottoni, A., Piccin, D., Tagliati, F., Luchin, A., Zatelli, M. C. and degli Uberti, E. C. (2005) miR-15a and miR-16-1 down-regulation in pituitary adenomas. J. Cell Physiol. 204, 280-285 https://doi.org/10.1002/jcp.20282
  25. Kr$\ddot{u}$ger, J. and Rehmsmeier, M. (2006) RNAhybrid: micro-RNA target prediction easy, fast and flexible. Nucleic. Acids Res. 34, 451-454 https://doi.org/10.1093/nar/gkl243
  26. Doench, J. G. and Sharp P. A. (2004) Specificity of micro- RNA target selection in translational repression. Genes & Dev. 18, 504-511 https://doi.org/10.1101/gad.1184404
  27. Carleton, M., Cleary, M. A. and Linsley, P. S. (2007) Micro RNAs and cell cycle regulation. Cell Cycle 6, 2127- 2132 https://doi.org/10.4161/cc.6.17.4641
  28. Moosavi M. A, Yazdanparast, R. and Lotfi, A. (2006) GTP induces S-phase cell-cycle arrest and inhibits DNA synthesis in K562 cells but not in normal human peripheral lymphocytes. J. Biochem. Mol. Biol. 39, 492-501 https://doi.org/10.5483/BMBRep.2006.39.5.492
  29. Gong, L., Jiang, C., Zhang, B., Hu, H., Wang, W. and Liu, X. (2006) Adenovirus-mediated expression of both antisense ornithine decarboxylase and s-denosylmethionine decarboxylase induces G1 arrest in HT-29 cells. J. Biochem. Mol. Biol. 39, 730-736 https://doi.org/10.5483/BMBRep.2006.39.6.730

Cited by

  1. Functional validation of microRNA-target RNA interactions vol.58, pp.2, 2012, https://doi.org/10.1016/j.ymeth.2012.08.002
  2. miR-15/16 complex targets p70S6 kinase1 and controls cell proliferation in MDA-MB-231 breast cancer cells vol.552, pp.2, 2014, https://doi.org/10.1016/j.gene.2014.09.052
  3. MicroRNA-16 Modulates HuR Regulation of Cyclin E1 in Breast Cancer Cells vol.16, pp.4, 2015, https://doi.org/10.3390/ijms16047112
  4. Metapristone (RU486 derivative) inhibits cell proliferation and migration as melanoma metastatic chemopreventive agent vol.90, 2017, https://doi.org/10.1016/j.biopha.2017.03.076
  5. Regulation of proliferation and apoptosis in human osteoblastic cells by microRNA-15b vol.79, 2015, https://doi.org/10.1016/j.ijbiomac.2015.05.017
  6. Signaling pathways in HPV-associated cancers and therapeutic implications vol.25, 2015, https://doi.org/10.1002/rmv.1823
  7. miR-29c induces cell cycle arrest in esophageal squamous cell carcinoma by modulating cyclin E expression vol.32, pp.7, 2011, https://doi.org/10.1093/carcin/bgr078
  8. The hunting of targets: challenge in miRNA research vol.27, pp.1, 2013, https://doi.org/10.1038/leu.2012.179
  9. MicroRNA 16 enhances differentiation of human bone marrow mesenchymal stem cells in a cardiac niche toward myogenic phenotypes in vitro vol.90, pp.25-26, 2012, https://doi.org/10.1016/j.lfs.2012.05.011
  10. Role of microRNAs in skeletal muscle development and rhabdomyosarcoma (Review) vol.11, pp.6, 2015, https://doi.org/10.3892/mmr.2015.3275
  11. A multi-targeted approach to suppress tumor-promoting inflammation vol.35, 2015, https://doi.org/10.1016/j.semcancer.2015.03.006
  12. MicroRNAs as new player in rheumatoid arthritis vol.78, pp.1, 2011, https://doi.org/10.1016/j.jbspin.2010.06.003
  13. Anti-Tumor Activity of Eurycoma longifolia Root Extracts against K-562 Cell Line: In Vitro and In Vivo Study vol.9, pp.1, 2014, https://doi.org/10.1371/journal.pone.0083818
  14. Trichostatin A alters the expression of cell cycle controlling genes and microRNAs in donor cells and subsequently improves the yield and quality of cloned bovine embryos in vitro vol.82, pp.7, 2014, https://doi.org/10.1016/j.theriogenology.2014.07.027
  15. Mir-33 regulates cell proliferation and cell cycle progression vol.11, pp.5, 2012, https://doi.org/10.4161/cc.11.5.19421
  16. Imaging Dendrimer-Grafted Graphene Oxide Mediated Anti-miR-21 Delivery With an Activatable Luciferase Reporter vol.8, pp.14, 2016, https://doi.org/10.1021/acsami.6b02662
  17. MicroRNAs and the cell cycle vol.1812, pp.5, 2011, https://doi.org/10.1016/j.bbadis.2011.02.002
  18. Les microARN : de nouveaux acteurs de la polyarthrite rhumatoïde vol.78, pp.2, 2011, https://doi.org/10.1016/j.rhum.2010.06.004
  19. c-MYC–miRNA circuitry vol.13, pp.2, 2014, https://doi.org/10.4161/cc.27646
  20. CPEB and miR-15/16 Co-Regulate Translation of Cyclin E1 mRNA during Xenopus Oocyte Maturation vol.11, pp.2, 2016, https://doi.org/10.1371/journal.pone.0146792
  21. Identification of microRNA-mRNA modules using microarray data vol.12, pp.1, 2011, https://doi.org/10.1186/1471-2164-12-138
  22. Characterization of microRNA expression in serous ovarian carcinoma vol.34, pp.2, 2014, https://doi.org/10.3892/ijmm.2014.1813
  23. Downregulation of the tumor-suppressor miR-16 via progestin-mediated oncogenic signaling contributes to breast cancer development vol.14, pp.3, 2012, https://doi.org/10.1186/bcr3187
  24. The Induction of microRNA-16 in Colon Cancer Cells by Protein Arginine Deiminase Inhibition Causes a p53-Dependent Cell Cycle Arrest vol.8, pp.1, 2013, https://doi.org/10.1371/journal.pone.0053791
  25. miRNAs and related polymorphisms in rheumatoid arthritis susceptibility vol.11, pp.9, 2012, https://doi.org/10.1016/j.autrev.2011.11.004
  26. microRNA and transcription factor mediated regulatory network for ovarian cancer vol.34, pp.5, 2013, https://doi.org/10.1007/s13277-013-0892-y
  27. miRNA response to DNA damage vol.36, pp.9, 2011, https://doi.org/10.1016/j.tibs.2011.06.002
  28. MicroRNA aberrations: An emerging field for gallbladder cancer management vol.22, pp.5, 2016, https://doi.org/10.3748/wjg.v22.i5.1787
  29. Triptolide inhibits the proliferation of cells from lymphocytic leukemic cell lines in association with downregulation of NF-κB activity and miR-16-1* vol.32, pp.4, 2011, https://doi.org/10.1038/aps.2010.237
  30. A Re-Examination of Global Suppression of RNA Interference by HIV-1 vol.6, pp.2, 2011, https://doi.org/10.1371/journal.pone.0017246
  31. Down-regulation of cyclin E1 expression by microrna-195 accounts for interferon-β-induced inhibition of hepatic stellate cell proliferation vol.226, pp.10, 2011, https://doi.org/10.1002/jcp.22598
  32. Global miRNA expression analysis identifies novel key regulators of plasma cell differentiation and malignant plasma cell vol.45, pp.10, 2017, https://doi.org/10.1093/nar/gkx327
  33. FXR1a-associated microRNP: A driver of specialized non-canonical translation in quiescent conditions vol.14, pp.2, 2017, https://doi.org/10.1080/15476286.2016.1265197
  34. Acute kidney injury: a paradigm for miRNA regulation of the cell cycle vol.42, pp.4, 2014, https://doi.org/10.1042/BST20140093
  35. Canine Mammary Carcinomas: A Comparative Analysis of Altered Gene Expression vol.3, pp.1, 2015, https://doi.org/10.3390/vetsci3010001
  36. The miR-15/107 Group of MicroRNA Genes: Evolutionary Biology, Cellular Functions, and Roles in Human Diseases vol.402, pp.3, 2010, https://doi.org/10.1016/j.jmb.2010.07.051
  37. Noncoding RNAs in DNA Repair and Genome Integrity vol.20, pp.4, 2014, https://doi.org/10.1089/ars.2013.5514
  38. /S phase cyclin, CCNE1, is lost in osteosarcomas vol.292, pp.52, 2017, https://doi.org/10.1074/jbc.M117.808287
  39. Omega-3 fatty acid DHA modulates p53, survivin, and microRNA-16-1 expression in KRAS-mutant colorectal cancer stem-like cells vol.13, pp.1, 2018, https://doi.org/10.1186/s12263-018-0596-4