scholarly journals A nitric oxide-releasing prodrug promotes apoptosis in human renal carcinoma cells: Involvement of reactive oxygen species

2021 ◽  
Vol 19 (1) ◽  
pp. 635-645
Author(s):  
Jindong Xie ◽  
Lieqian Chen ◽  
Dongqiang Huang ◽  
Weiwei Yue ◽  
Jingyu Chen ◽  
...  

Abstract Background JS-K is a nitric oxide (NO)-releasing prodrug of the O2-arylated diazeniumdiolate group that shows pronounced cytotoxicity and antitumor properties in numerous cancer models, including in vitro as well as in vivo. Reactive oxygen species (ROS) induce carcinogenesis by altering the redox status, causing increment in vulnerability to oxidative stress. Material and methods To determine the effect of JS-K, a glutathione S-transferase (GST)-activated NO-donor prodrug, on the induction of ROS accumulation, proliferation, and apoptosis in human renal carcinoma cells, we measured the changes of cell proliferation, apoptosis, ROS growth, and initiation of the mitochondrial signaling pathway before and after JS-K treatment. Results In vitro, dose- and time-dependent development of renal carcinoma cells were controlled for JS-K, and JS-K also triggered ROS aggregation and cell apoptosis. Treatment with JS-K induces the levels of pro-apoptotic proteins (Bak and Bax) and decrease the number of anti-apoptotic protein (Bcl-2). In fact, JS-K-induced apoptosis was reversed by the antioxidant N-acetylcysteine, and oxidized glutathione, a pro-oxidant, improved JS-K-induced apoptosis. Finally, we demonstrated that in renal carcinoma cells, JS-K improved the chemosensitivity of doxorubicin. Conclusion Our data indicate that JS-K-released NO induce apoptosis of renal carcinoma cells by increasing ROS levels.

PLoS ONE ◽  
2012 ◽  
Vol 7 (7) ◽  
pp. e40727 ◽  
Author(s):  
Szu-Ying Wu ◽  
Yann-Lii Leu ◽  
Ya-Ling Chang ◽  
Tian-Shung Wu ◽  
Ping-Chung Kuo ◽  
...  

2020 ◽  
Vol 16 (3) ◽  
pp. e1008379 ◽  
Author(s):  
Artur Santos-Miranda ◽  
Julliane Vasconcelos Joviano-Santos ◽  
Grazielle Alves Ribeiro ◽  
Ana Flávia M. Botelho ◽  
Peter Rocha ◽  
...  

2011 ◽  
Vol 25 (3) ◽  
pp. 692-698 ◽  
Author(s):  
Min Jung Choi ◽  
Eun Jung Park ◽  
Kyoung Jin Min ◽  
Jong-Wook Park ◽  
Taeg Kyu Kwon

Life Sciences ◽  
2005 ◽  
Vol 76 (16) ◽  
pp. 1873-1881 ◽  
Author(s):  
Jun-Nian Zheng ◽  
Ya-Feng Sun ◽  
Dong-Sheng Pei ◽  
Jun-Jie Liu ◽  
Xiao-Qing Sun ◽  
...  

2020 ◽  
Vol 16 (10) ◽  
pp. e1009049
Author(s):  
Artur Santos-Miranda ◽  
Julliane Vasconcelos Joviano-Santos ◽  
Grazielle Alves Ribeiro ◽  
Ana Flávia M. Botelho ◽  
Peter Rocha ◽  
...  

Antioxidants ◽  
2020 ◽  
Vol 9 (6) ◽  
pp. 523
Author(s):  
Yiru Wang ◽  
Chengmin Li ◽  
Julang Li ◽  
Genlin Wang ◽  
Lian Li

Negative energy balance (NEB) during the perinatal period can affect dairy cow follicular development and reduce the fecundity. Non-esterified fatty acid (NEFA) concentration is elevated during NEB, and is known to be toxic for multiple cell types. In the ovary, NEB increased NEFA, and may influences follicular growth and development. However, the effect and mechanism of NEFA on granulosa cells (GCs) in vitro remains unknown. In this study, we found that NEFA dose-dependently induced apoptosis in primary cultured granulosa cells. Mechanistically, our data showed that NEFA significantly increased reactive oxygen species (ROS) levels, resulting in the activation of endoplasmic reticulum stress (ERS) and eventually cell apoptosis in GCs. Moreover, NEFA also increased the phosphorylation levels of ERK1/2 and p38MAPK pathways, upregulated the expression of p53 and potentially promoted its translocation to the nuclear, thus transcriptionally activated Bax, a downstream gene of this pathway. NEFA also promoted nuclear factor E2 (Nrf2) expression and its level in the nuclear. To elucidate the mechanism of NEFA action, N-acetyl-l-cysteine (NAC), a ROS scavenger was used to verify the role of ROS in NEFA induced apoptosis of GCs. NAC pretreatment reversed the NEFA-induced ERS-related protein and apoptosis-related protein levels. Meanwhile, NAC pretreatment also blocked the phosphorylation of ERK1/2 and p38 induced by NEFA, and the nucleation of Nrf2 and p53, suggesting that ROS plays a crucial role in regulating the NEFA-induced apoptosis of GCs. Together, these findings provide an improved understanding of the mechanisms underlying GCs apoptosis, which could potentially be useful for improving ovarian function.


Sign in / Sign up

Export Citation Format

Share Document