DOI QR코드

DOI QR Code

The Role of Kif4A in Doxorubicin-Induced Apoptosis in Breast Cancer Cells

  • Wang, Hui (Department of Pathology, The Third Affiliated Hospital of Soochow University) ;
  • Lu, Changqing (Department of Pathology, The Third Affiliated Hospital of Soochow University) ;
  • Li, Qing (Department of Pathology, The Third Affiliated Hospital of Soochow University) ;
  • Xie, Jun (Department of Pathology, The Third Affiliated Hospital of Soochow University) ;
  • Chen, Tongbing (Department of Pathology, The Third Affiliated Hospital of Soochow University) ;
  • Tan, Yan (Department of Pathology, The Third Affiliated Hospital of Soochow University) ;
  • Wu, Changping (Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University) ;
  • Jiang, Jingting (Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University)
  • Received : 2014.07.28
  • Accepted : 2014.09.05
  • Published : 2014.11.30

Abstract

This study was to investigate the mechanism and role of Kif4A in doxorubicin-induced apoptosis in breast cancer. Using two human breast cancer cell lines MCF-7 (with wild-type p53) and MDA-MB-231 (with mutant p53), we quantitated the expression levels of kinesin super-family protein 4A (Kif4A) and poly (ADP-ribose) Polymerase-1 (PARP-1) by Western blot after doxorubicin treatment and examined the apoptosis by flow cytometry after treatment with doxorubicin and PARP-1 inhibitor, 3-Aminobenzamide (3-ABA). Our results showed that doxorubicin treatment could induce the apoptosis of MCF-7 and MDA-MB-231 cells, the down-regulation of Kif4A and upregulation of poly(ADP-ribose) (PAR). The activity of PARP-1 or PARP-1 activation was significantly elevated by doxorubicin treatment in dose- and time-dependent manners (P < 0.05), while doxorubicin treatment only slightly elevated the level of cleaved fragments of PARP-1 (P > 0.05). We further demonstrated that overexpression of Kif4A could reduce the level of PAR and significantly increase apoptosis. The effect of doxorubicin on apoptosis was more profound in MCF-7 cells compared with MDA-MB-231 cells (P < 0.05). Taken together, our results suggest that the novel role of Kif4A in doxorubicin-induced apoptosis in breast cancer cells is achieved by inhibiting the activity of PARP-1.

Keywords

References

  1. Alli, E., Sharma, V.B., Hartman, A.R., Lin, P.S., McPherson, L., and Ford, J.M. (2011). Enhanced sensitivity to cisplatin and gemcitabine in Brca1-deficient murine mammary epithelial cells. BMC Pharmacol. 11, 7.
  2. Annunziata, C.M., and Bates, S.E. (2010). PARP inhibitors in BRCA1/BRCA2 germline mutation carriers with ovarian and breast cancer. F1000 Biol. Rep. 2.
  3. Borysov, S.I., Granic, A., Padmanabhan, J., Walczak, C.E., and Potter, H. (2011). Alzheimer Abeta disrupts the mitotic spindle and directly inhibits mitotic microtubule motors. Cell Cycle 10, 1397-1410. https://doi.org/10.4161/cc.10.9.15478
  4. Campalans, A., Kortulewski, T., Amouroux, R., Menoni, H., Vermeulen, W., and Radicella, J.P. (2013). Distinct spatiotemporal patterns and PARP dependence of XRCC1 recruitment to single-strand break and base excision repair. Nucleic Acids Res. 41, 3115-3129. https://doi.org/10.1093/nar/gkt025
  5. Farmer, H., McCabe, N., Lord, C.J., Tutt, A.N., Johnson, D.A., Richardson, T.B., Santarosa, M., Dillon, K.J., Hickson, I., Knights, C., et al. (2005). Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917-921. https://doi.org/10.1038/nature03445
  6. Hu, C.K., Coughlin, M., Field, C.M., and Mitchison, T.J. (2011). KIF4 regulates midzone length during cytokinesis. Curr. Biol. 21, 815-824. https://doi.org/10.1016/j.cub.2011.04.019
  7. Huambachano, O., Herrera, F., Rancourt, A., and Satoh, M.S. (2011). Double-stranded DNA binding domain of poly(ADP-ribose) polymerase-1 and molecular insight into the regulation of its activity. J. Biol. Chem. 286, 7149-7160. https://doi.org/10.1074/jbc.M110.175190
  8. Hydock, D.S., Lien, C.Y., Jensen, B.T., Parry, T.L., Schneider, C.M., and Hayward, R. (2012). Rehabilitative exercise in a rat model of doxorubicin cardiotoxicity. Exp. Biol. Med. 237, 1483-1492. https://doi.org/10.1258/ebm.2012.012137
  9. Ibragimova, I., and Cairns, P. (2011). Assays for hypermethylation of the BRCA1 gene promoter in tumor cells to predict sensitivity to PARP-inhibitor therapy. Methods Mol. Biol. 780, 277-291. https://doi.org/10.1007/978-1-61779-270-0_17
  10. Jacot, W., Thezenas, S., Senal, R., Viglianti, C., Laberenne, A.C., Lopez-Crapez, E., Bibeau, F., Bleuse, J.P., Romieu, G., and Lamy, P.J. (2013). BRCA1 promoter hypermethylation, 53BP1 protein expression and PARP-1 activity as biomarkers of DNA repair deficit in breast cancer. BMC Cancer 13, 523. https://doi.org/10.1186/1471-2407-13-523
  11. Javle, M., and Curtin, N.J. (2011). The role of PARP in DNA repair and its therapeutic exploitation. Br. J. Cancer 105, 1114-1122. https://doi.org/10.1038/bjc.2011.382
  12. Langelier, M.F., Planck, J.L., Roy, S., and Pascal, J.M. (2011). Crystal structures of poly(ADP-ribose) polymerase-1 (PARP-1) zinc fingers bound to DNA: structural and functional insights into DNA-dependent PARP-1 activity. J. Biol. Chem. 286, 10690-10701. https://doi.org/10.1074/jbc.M110.202507
  13. Lee Kraus, W., and Hottiger, M.O. (2013). PARP-1 and gene regulation: progress and puzzles. Mol. Aspects Med. 34, 1109-1123. https://doi.org/10.1016/j.mam.2013.01.005
  14. Martinez-Romero, R., Canuelo, A., Siles, E., Oliver, F.J., and Martinez-Lara, E. (2012). Nitric oxide modulates hypoxiainducible factor-1 and poly(ADP-ribose) polymerase-1 cross talk in response to hypobaric hypoxia. J. Appl. Physiol. 112, 816-823. https://doi.org/10.1152/japplphysiol.00898.2011
  15. Midorikawa, R., Takei, Y., and Hirokawa, N. (2006). KIF4 motor regulates activity-dependent neuronal survival by suppressing PARP-1 enzymatic activity. Cell 125, 371-383. https://doi.org/10.1016/j.cell.2006.02.039
  16. Munoz-Gamez, J.A., Martin-Oliva, D., Aguilar-Quesada, R., Canuelo, A., Nunez, M.I., Valenzuela, M.T., Ruiz de Almodovar, J.M., De Murcia, G., and Oliver, F.J. (2005). PARP inhibition sensitizes p53-deficient breast cancer cells to doxorubicin-induced apoptosis. Biochem. J. 386, 119-125. https://doi.org/10.1042/BJ20040776
  17. Oonk, A.M., van Rijn, C., Smits, M.M., Mulder, L., Laddach, N., Savola, S.P., Wesseling, J., Rodenhuis, S., Imholz, A.L., and Lips, E.H. (2012). Clinical correlates of 'BRCAness' in triple-negative breast cancer of patients receiving adjuvant chemotherapy. Ann. Oncol. 23, 2301-2305. https://doi.org/10.1093/annonc/mdr621
  18. Shrestha, S., Wilmeth, L.J., Eyer, J., and Shuster, C.B. (2012). PRC1 controls spindle polarization and recruitment of cytokinetic factors during monopolar cytokinesis. Mol. Biol. Cell 23, 1196-1207. https://doi.org/10.1091/mbc.E11-12-1008
  19. Stanisavljevic, J., Porta-de-la-Riva, M., Batlle, R., de Herreros, A.G., and Baulida, J. (2011). The p65 subunit of NF-kappaB and PARP1 assist Snail1 in activating fibronectin transcription. J. Cell Sci. 124, 4161-4171. https://doi.org/10.1242/jcs.078824
  20. Sukowati, C.H., Rosso, N., Pascut, D., Anfuso, B., Torre, G., Francalanci, P., Croce, L.S., and Tiribelli, C. (2012). Gene and functional up-regulation of the BCRP/ABCG2 transporter in hepatocellular carcinoma. BMC Gastroenterol. 12, 160. https://doi.org/10.1186/1471-230X-12-160
  21. Sun, S., Chen, Z., Li, L., Sun, D., Tian, Y., Pan, H., Bi, H., Huang, M., Zeng, S., and Jiang, H. (2012). The two enantiomers of tetrahydropalmatine are inhibitors of P-gp, but not inhibitors of MRP1 or BCRP. Xenobiotica 42, 1197-1205. https://doi.org/10.3109/00498254.2012.702247
  22. Villar, V.H., Vogler, O., Martinez-Serra, J., Ramos, R., Calabuig-Farinas, S., Gutierrez, A., Barcelo, F., Martin-Broto, J., and Alemany, R. (2012). Nilotinib counteracts P-glycoproteinmediated multidrug resistance and synergizes the antitumoral effect of doxorubicin in soft tissue sarcomas. PLoS One 7, e37735. https://doi.org/10.1371/journal.pone.0037735
  23. Wandke, C., Barisic, M., Sigl, R., Rauch, V., Wolf, F., Amaro, A.C., Tan, C.H., Pereira, A.J., Kutay, U., Maiato, H., et al. (2012). Human chromokinesins promote chromosome congression and spindle microtubule dynamics during mitosis. J. Cell Biol. 198, 847-863. https://doi.org/10.1083/jcb.201110060
  24. Wu, G., Zhou, L., Khidr, L., Guo, X.E., Kim, W., Lee, Y.M., Krasieva, T., and Chen, P.L. (2008). A novel role of the chromokinesin Kif4A in DNA damage response. Cell Cycle 7, 2013-2020. https://doi.org/10.4161/cc.7.13.6130
  25. Xu, Y., Diao, Y., Qi, S., Pan, X., Wang, Q., Xin, Y., Cao, X., Ruan, J., Zhao, Z., Luo, L., et al. (2013). Phosphorylated Hsp27 activates ATM-dependent p53 signaling and mediates the resistance of MCF-7 cells to doxorubicin-induced apoptosis. Cell. Signal. 25, 1176-1185. https://doi.org/10.1016/j.cellsig.2013.01.017
  26. Zaremba, T., Thomas, H., Cole, M., Plummer, E.R., and Curtin, N.J. (2010). Doxorubicin-induced suppression of poly(ADP-ribose) polymerase-1 (PARP-1) activity and expression and its implication for PARP inhibitors in clinical trials. Cancer Chemother. Pharmacol. 66, 807-812. https://doi.org/10.1007/s00280-010-1359-0
  27. Zheng, Z., Aojula, H., and Clarke, D. (2010). Reduction of doxorubicin resistance in P-glycoprotein overexpressing cells by hybrid cell-penetrating and drug-binding peptide. J. Drug Target. 18, 477-487. https://doi.org/10.3109/10611860903548347

Cited by

  1. Increased anti-tumour activity by exosomes derived from doxorubicin-treated tumour cells via heat stress vol.31, pp.5, 2015, https://doi.org/10.3109/02656736.2015.1036384
  2. Mitochondria-Derived Reactive Oxygen Species Play an Important Role in Doxorubicin-Induced Platelet Apoptosis vol.16, pp.5, 2015, https://doi.org/10.3390/ijms160511087
  3. Systematic Identification and Assessment of Therapeutic Targets for Breast Cancer Based on Genome-Wide RNA Interference Transcriptomes vol.8, pp.3, 2017, https://doi.org/10.3390/genes8030086
  4. Upregulate KIF4A Enhances Proliferation, Invasion of Hepatocellular Carcinoma and Indicates poor prognosis Across Human Cancer Types vol.7, pp.1, 2017, https://doi.org/10.1038/s41598-017-04176-9
  5. Enhanced cytotoxic activity of doxorubicin through the inhibition of autophagy in triple negative breast cancer cell line vol.1861, pp.2, 2017, https://doi.org/10.1016/j.bbagen.2016.11.013
  6. RNA-Seq based genome-wide analysis reveals loss of inter-chromosomal regulation in breast cancer vol.7, pp.1, 2017, https://doi.org/10.1038/s41598-017-01314-1
  7. The kinesin Eg5 inhibitor K858 induces apoptosis but also survivin-related chemoresistance in breast cancer cells vol.34, pp.4, 2016, https://doi.org/10.1007/s10637-016-0345-8
  8. HOTAIR upregulates an 18-gene cell cycle-related mRNA network in glioma vol.50, pp.4, 2017, https://doi.org/10.3892/ijo.2017.3901
  9. Anthocyanin Attenuates Doxorubicin-Induced Cardiomyotoxicity via Estrogen Receptor-α/β and Stabilizes HSF1 to Inhibit the IGF-IIR Apoptotic Pathway vol.17, pp.9, 2016, https://doi.org/10.3390/ijms17091588
  10. Prognostic genes of breast cancer revealed by gene co-expression network analysis vol.14, pp.4, 2017, https://doi.org/10.3892/ol.2017.6779
  11. Kinesin superfamily: roles in breast cancer, patient prognosis and therapeutics vol.37, pp.7, 2018, https://doi.org/10.1038/onc.2017.406
  12. A novel PHD-finger protein 14/KIF4A complex overexpressed in lung cancer is involved in cell mitosis regulation and tumorigenesis vol.8, pp.12, 2014, https://doi.org/10.18632/oncotarget.14962
  13. Gene co-expression network reveals shared modules predictive of stage and grade in serous ovarian cancers vol.8, pp.26, 2014, https://doi.org/10.18632/oncotarget.17785
  14. Pre-clinical pharmacology of AZD3965, a selective inhibitor of MCT1: DLBCL, NHL and Burkitt’s lymphoma anti-tumor activity vol.8, pp.41, 2017, https://doi.org/10.18632/oncotarget.18215
  15. Distinct Diagnostic and Prognostic Values of Kinesin Family Member Genes Expression in Patients with Breast Cancer vol.24, pp.None, 2014, https://doi.org/10.12659/msm.913401
  16. An integrated bioinformatical analysis to evaluate the role of KIF4A as a prognostic biomarker for breast cancer vol.11, pp.None, 2014, https://doi.org/10.2147/ott.s164730
  17. Integrated analysis reveals down-regulation of SPARCL1 is correlated with cervical cancer development and progression vol.21, pp.2, 2014, https://doi.org/10.3233/cbm-170501
  18. Enhanced expression of KIF4A in colorectal cancer is associated with lymph node metastasis vol.15, pp.2, 2014, https://doi.org/10.3892/ol.2017.7555
  19. Identification of KIF11 As a Novel Target in Meningioma vol.11, pp.4, 2014, https://doi.org/10.3390/cancers11040545
  20. Overexpression of CDCA5 , KIF4A , TPX2 , and FOXM1 Coregulated Cell Cycle and Promoted Hepatocellular Carcinoma Development vol.27, pp.6, 2014, https://doi.org/10.1089/cmb.2019.0254
  21. Integrated bioinformatics analysis for the screening of hub genes and therapeutic drugs in ovarian cancer vol.13, pp.None, 2014, https://doi.org/10.1186/s13048-020-0613-2
  22. Identification of KIF4A as a pan-cancer diagnostic and prognostic biomarker via bioinformatics analysis and validation in osteosarcoma cell lines vol.9, pp.None, 2014, https://doi.org/10.7717/peerj.11455
  23. Noncovalent Functionalized Graphene Nanocarriers from Graphite for Treating Thyroid Cancer Cells vol.7, pp.6, 2014, https://doi.org/10.1021/acsbiomaterials.1c00067
  24. Potential Prognostic Biomarkers of NIMA (Never in Mitosis, Gene A)-Related Kinase (NEK) Family Members in Breast Cancer vol.11, pp.11, 2014, https://doi.org/10.3390/jpm11111089