Copper enriches efficacy of Dp44mT in breast cancer cells

Copper enriches efficacy of Dp44mT in breast cancer cells

Neoplastic cells need essential metals, such as iron and copper, for cellular functions and rapid growth. Metal chelation and changes in their redox cycle in favor of oxidative stress may be critical for making these cells vulnerable to cell death. The HER2-overexpressing breast cancer cell line, MCF7-HER2, and its vehicle control, MCF7-vec cells, were treated with CuCl2, FeSO4, and Dp44mT. Reactive oxygen species (ROS) production in response to these chemicals was detected with flow cytometry, and cell viability was measured by MTT assay in the cells. ROS levels were relatively increased by Dp44mT in the cells, and this was reversed by a combination of iron, while a copper combination further induced ROS. Parallel changes were observed in the inhibition of cell growth by Dp44mT, and NAC partially rescued the inhibition. Additionally, copper decreased the CSC population by half relative to the control in MCF7-HER2 cells. Copper supplement enriches ROS production mediated by Dp44mT in MCF7 cells. However, iron addition recovers Dp44mT-depleted iron levels, yet has no effect on ROS generation. Copper treatment also decreases the proportion of CSCs. In this manner, Dp44mT depletes iron and binds copper to form a redox active complex, which leads to oxidative stress. This dual cytotoxic case is significant for the survival of cancer cells.

___

  • Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003). Prospective identification of tumorigenic breast cancer cells. P Natl Acad Sci USA 100: 3983-3988.
  • Buss JL, Greene BT, Turner J, Torti FM, Torti SV (2004). Iron chelators in cancer chemotherapy. Curr Top Med Chem 4: 1623-1635.
  • Cai L, Li XK, Song Y, Cherian MG (2005). Essentiality, toxicology and chelation therapy of zinc and copper. Curr Med Chem 12: 2753-2763.
  • De Domenico I, McVey Ward D, Kaplan J (2008). Regulation of iron acquisition and storage: consequences for iron-linked disorders. Nat Rev Mol Cell Biol 9: 72-81.
  • Denoyer D, Masaldan S, La Fontaine S, Cater MA (2015). Targeting copper in cancer therapy: 'Copper That Cancer'. Metallomics 7: 1459-1476.
  • Finney L, Mandava S, Ursos L, Zhang W, Rodi D, Vogt S, Legnini D, Maser J, Ikpatt F, Olopade OI et al. (2007). X-ray fluorescence microscopy reveals large-scale relocalization and extracellular translocation of cellular copper during angiogenesis. P Natl Acad Sci USA 104: 2247-2252.
  • Fox SB, Gasparini G, Harris AL (2001). Angiogenesis: pathological, prognostic, and growth-factor pathways and their link to trial design and anticancer drugs. Lancet Oncol 2: 278-289.
  • Gourley M, Williamson JS (2000). Angiogenesis: new targets for the development of anticancer chemotherapies. Curr Pharm Des 6: 417-439.
  • Grubman A, White AR (2014). Copper as a key regulator of cell signalling pathways. Expert Rev Mol Med 16: e11.
  • Gu Y, Fu J, Lo P, Wang S, Wang Q, Chen H (2011). The effect of B27 supplement on promoting in vitro propagation of Her2/neu-transformed mammary tumorspheres. J Biotech Res 3: 7-18.
  • Gupte A, Mumper RJ (2009). Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat Rev 35: 32-46.
  • Huang YL, Sheu JY, Lin TH (1999). Association between oxidative stress and changes of trace elements in patients with breast cancer. Clin Biochem 32: 131-136.
  • Jansson PJ, Hawkins CL, Lovejoy DB, Richardson DR (2010a). The iron complex of Dp44mT is redox-active and induces hydroxyl radical formation: an EPR study. J Inorg Biochem 104: 1224-1228.
  • Jansson PJ, Sharpe PC, Bernhardt PV, Richardson DR (2010b). Novel thiosemicarbazones of the ApT and DpT series and their copper complexes: identification of pronounced redox activity and characterization of their antitumor activity. J Med Chem 53: 5759-5769.
  • Jomova K, Valko M (2011). Advances in metal-induced oxidative stress and human disease. Toxicology 283: 65-87.
  • Kalinowski DS, Richardson DR (2005). The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev 57: 547-583.
  • Korkaya H, Paulson A, Iovino F, Wicha MS (2008). HER2 regulates the mammary stem/progenitor cell population driving tumorigenesis and invasion. Oncogene 27: 6120-6130.
  • Lane DJ, Merlot AM, Huang ML, Bae DH, Jansson PJ, Sahni S, Kalinowski DS, Richardson DR (2015). Cellular iron uptake, trafficking and metabolism: key molecules and mechanisms and their roles in disease. Biochim Biophys Acta 1853: 1130-1144.
  • Lo PK, Kanojia D, Liu X, Singh UP, Berger FG, Wang Q, Chen H (2012). CD49f and CD61 identify Her2/neu-induced mammary tumor initiating cells that are potentially derived from luminal progenitors and maintained by the integrin-TGF beta signaling. Oncogene 24: 2614-2626.
  • Magnifico A, Albano L, Campaner S, Delia D, Castiglioni F, Gasparini P, Sozzi G, Fontanella E, Menard S, Tagliabue E (2009). Tumor-initiating cells of HER2-positive carcinoma cell lines express the highest oncoprotein levels and are sensitive to trastuzumab. Clin Cancer Res 15: 2010-2021.
  • Merlot AM, Kalinowski DS, Richardson DR (2013). Novel chelators for cancer treatment: where are we now? Antioxid Redox Signal 18: 973-1006.
  • Pahl PM, Horwitz LD (2005). Cell permeable iron chelators as potential cancer chemotherapeutic agents. Cancer Invest 23: 683-691.
  • Rao VA, Klein SR, Agama KK, Toyoda E, Adachi N, Pommier Y, Shacter EB (2009). The iron chelator Dp44mT causes DNA damage and selective inhibition of topoisomerase IIα in breast cancer cells. Cancer Res 69: 948-957.
  • Richardson DR, Sharpe PC, Lovejoy DB, Senaratne D, Kalinowski DS, Islam M, Bernhardt PV (2006). Dipyridyl thiosemicarbazone chelators with potent and selective antitumor activity form iron complexes with redox activity. J Med Chem 49: 6510-6521.
  • Tian J, Peehl DM, Zheng W, Knox SJ (2010). Anti-tumor and radiosensitization activities of the iron chelator HDp44mT are mediated by effects on intracellular redox status. Cancer Lett 298: 231-237.
  • Torti SV, Torti FM (2013). Iron and cancer: more ore to be mined. Nat Rev Cancer 13: 342-355.
  • Toyokuni S (2009). Role of iron in carcinogenesis: cancer as a ferrotoxic disease. Cancer Sci 100: 9-16.
  • Trinder D, Zak O, Aisen P (1996). Transferrin receptor-independent uptake of differic transferrin by human hepatoma cells with antisense inhibition of receptor expression. Hepatology 23:1512-1520.
  • Turski ML, Thiele DJ (2009). New roles for copper metabolism in cell proliferation, signaling, and disease. J Biol Chem 284: 717-721.
  • Whitnall M, Howard J, Ponka P, Richardson DR (2006). A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. P Natl Acad Sci USA 103: 14901-14906.
  • Yu Y, Kalinowski DS, Kovacevic Z, Siafakas AR, Jansson PJ, Stefani C, Lovejoy DB, Sharpe PC, Bernhardt PV, Richardson DR (2009). Thiosemicarbazones from the old to new: iron chelators that are more than just ribonucleotide reductase inhibitors. J Med Chem 2: 5271-5294.
  • Yu Y, Wong J, Lovejoy DB, Kalinowski DS, Richardson DR (2006). Chelators at the cancer coalface: desferrioxamine to Triapine and beyond. Clin Cancer Res 12: 6876-6883.
  • Yuan J, Lovejoy DB, Richardson DR (2004). Novel di-2-pyridyl-derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment. Blood 104: 1450-1458.
Turkish Journal of Biology-Cover
  • ISSN: 1300-0152
  • Yayın Aralığı: Yılda 6 Sayı
  • Yayıncı: TÜBİTAK