Oncotarget

Research Papers:

Mansouramycin C kills cancer cells through reactive oxygen species production mediated by opening of mitochondrial permeability transition pore

Shan Kuang, Ge Liu, Ruobing Cao, Linlin Zhang, Qiang Yu and Chaomin Sun _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:104057-104071. https://doi.org/10.18632/oncotarget.22004

Metrics: PDF 1394 views  |   HTML 2728 views  |   ?  


Abstract

Shan Kuang1,2, Ge Liu1,2,3, Ruobing Cao1,2,3, Linlin Zhang1,2,3, Qiang Yu4 and Chaomin Sun1,2

1Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China

2Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China

3College of Earth Science, University of Chinese Academy of Sciences, Beijing, China

4Division of Tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China

Correspondence to:

Chaomin Sun, email: [email protected]

Keywords: mansouramycin C, marine-derived isoquinolinequinone, reactive oxygen species, mitochondrial permeability transition pore, anticancer drug

Received: May 17, 2017     Accepted: October 02, 2017     Published: October 24, 2017

ABSTRACT

Cancer is one of the deadliest diseases in the world and the search for novel anticancer agents is urgently required. Marine-derived isoquinolinequinones have exhibited promising anticancer activities. However, the exact mechanisms of cytotoxic activities of these isoquinolinequinones are poorly characterized. In this study, we investigated the anticancer effects and molecular mechanisms of mansouramycin C (Mm C), a cytotoxic isoquinolinequinone isolated from a marine streptomycete. We demonstrated that Mm C preferentially killed cancer cells and the cytotoxic effects were mediated by reactive oxygen species (ROS) generation. Mass spectrometry based proteomic analysis of Mm C-treated A549 cells revealed that many ROS-related proteins were differentially expressed. Proteomic-profiling after Mm C treatment identified oxidative phosphorylation as the most significant changes in pathways. Analysis also revealed extensive defects in mitochondrial structure and function. Furthermore, we disclosed that Mm C-induced ROS generation was caused by opening of mitochondrial permeability transition pore. Notably, Mm C synergized with sorafenib to induce cell death in A549 cells. Hence, we propose that the marine-derived natural compound Mm C is a potent inducer of the mitochondrial permeability transition and a promising anticancer drug candidate. Moreover, molecular mechanisms of Mm C shed new light on the understanding of the cytotoxic mechanisms of marine-derived isoquinolinequiones.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 22004