Oncotarget

Research Papers:

Ca2+ influx-mediated dilation of the endoplasmic reticulum and c-FLIPL downregulation trigger CDDO-Me-induced apoptosis in breast cancer cells

Soo Ah Jeong _, In Young Kim, A Reum Lee, Mi Jin Yoon, Hyeseong Cho, Jong-Soo Lee and Kyeong Sook Choi

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:21173-21192. https://doi.org/10.18632/oncotarget.4065

Metrics: PDF 1843 views  |   HTML 2288 views  |   ?  


Abstract

Soo Ah Jeong1,2,*, In Young Kim1,2,*, A Reum Lee1,2, Mi Jin Yoon1,2, Hyeseong Cho1,2, Jong-Soo Lee3, Kyeong Sook Choi1,2

1Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea

2Graduate Program of Cancer Biology, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea

3Department of Life Science, Ajou University, Suwon, Korea

*These authors have contributed equally to this work

Correspondence to:

Kyeong Sook Choi, e-mail: [email protected]

Keywords: CDDO-Me, endoplasmic reticulum, Ca2+, c-FLIPL, apoptosis

Received: January 23, 2015     Accepted: May 12, 2015     Published: May 25, 2015

ABSTRACT

The synthetic triterpenoid 2-cyano-3, 12-dioxooleana-1, 9(11)-dien-C28-methyl ester (CDDO-Me) is considered a promising anti-tumorigenic compound. In this study, we show that treatment with CDDO-Me induces progressive endoplasmic reticulum (ER)-derived vacuolation in various breast cancer cells and ultimately kills these cells by inducing apoptosis. We found that CDDO-Me–induced increases in intracellular Ca2+ levels, reflecting influx from the extracellular milieu, make a critical contribution to ER-derived vacuolation and subsequent cell death. In parallel with increasing Ca2+ levels, CDDO-Me markedly increased the generation of reactive oxygen species (ROS). Interestingly, there exists a reciprocal positive-regulatory loop between Ca2+ influx and ROS generation that triggers ER stress and ER dilation in response to CDDO-Me. In addition, CDDO-Me rapidly reduced the protein levels of c-FLIPL (cellular FLICE-inhibitory protein) and overexpression of c-FLIPL blocked CDDO-Me–induced cell death, but not vacuolation. These results suggest that c-FLIPL downregulation is a key contributor to CDDO-Me–induced apoptotic cell death, independent of ER-derived vacuolation. Taken together, our results show that ER-derived vacuolation via Ca2+ influx and ROS generation as well as caspase activation via c-FLIPL downregulation are responsible for the potent anticancer effects of CDDO-Me on breast cancer cells.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 4065