Advanced Search

Submit Manuscript

Volume 18, No 4, Apr 2008

ISSN: 1001-0602 
EISSN: 1748-7838 2018 
impact factor 17.848* 
(Clarivate Analytics, 2019)

Volume 18 Issue 4, April 2008: 458-471

ORIGINAL ARTICLES

Redox status of thioredoxin-1 (TRX1) determines the sensitivity of human liver carcinoma cells (HepG2) to arsenic trioxide-induced cell death

Changhai Tian1, Ping Gao1, Yanhua Zheng3, Wen Yue1, Xiaohui Wang1, Haijing Jin1 and Quan Chen1,2

1The Laboratory of Apoptosis and Cancer Biology, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, The Graduate School of Chinese Academy of Sciences, Chinese Academy of Sciences, Datun Rd, Chao Yang District, Beijing 100101, China

2College of Life Sciences, Nan Kai University, Tianjin 300071, China

3Department of Neuro-Oncology, MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
Correspondence: Quan Chen(chenq@ioz.ac.cn)

Intracellular redox homeostasis plays a critical role in determining tumor cells' sensitivity to drug-induced apoptosis. Here we investigated the role of thioredoxin-1 (TRX1), a key component of redox regulation, in arsenic trioxide (As2O3)-induced apoptosis. Over-expression of wild-type TRX1 in HepG2 cells led to the inhibition of As2O3-induced cytochrome c (cyto c) release, caspase activation and apoptosis, and down-regulation of TRX1 expression by RNAi sensitized HepG2 cells to As2O3-induced apoptosis. Interestingly, mutation of the active site of TRX1 from Cys32/35 to Ser32/35 converted this molecule from an apoptotic protector to an apoptotic promoter. In an effort to understand the mechanisms of this conversion, we used isolated mitochondria from mouse liver and found that recombinant wild-type TRX1 could protect mitochondria from the apoptotic changes. In contrast, the mutant form of TRX1 alone elicited mitochondria-related apoptotic changes, including the mitochondrial permeability transition pore (mPTP) opening, loss of mitochondrial membrane potential, and cyto c release from mitochondria. These apoptotic effects were inhibited by cyclosporine A (CsA), indicating that mutant TRX1 targeted to mPTP. Alteration of TRX1 from its reduced form to oxidized form in vivo by 2,4-dinitrochlorobenzene (DNCB), a specific inhibitor of TRX reductase, also sensitized HepG2 cells to As2O3-induced apoptosis. These data suggest that TRX1 plays a central role in regulating apoptosis by blocking cyto c release, and inactivation of TRX1 by either mutation or oxidization of the active site cysteines may sensitize tumor cells to As2O3-induced apoptosis.


Cell Research (2008) 18:458-471. doi: 10.1038/cr.2007.112; published online 24 December 2007

FULL TEXT | PDF

Browse 1526