Induction of G1 Cell Cycle Arrest in Human Glioma Cells by Salinomycin Through Triggering ROS-Mediated DNA Damage In Vitro and In Vivo

2016 ◽  
Vol 42 (4) ◽  
pp. 997-1005 ◽  
Author(s):  
Shi-Jun Zhao ◽  
Xian-Jun Wang ◽  
Qing-Jian Wu ◽  
Chao Liu ◽  
Da-Wei Li ◽  
...  
2016 ◽  
Vol 13 (2) ◽  
pp. 1007-1013 ◽  
Author(s):  
Aisha Maimaitili ◽  
Zunhua Shu ◽  
Xiaojiang Cheng ◽  
Kadeer Kaheerman ◽  
Alifu Sikandeer ◽  
...  

2015 ◽  
Vol 35 (7) ◽  
pp. 953-959 ◽  
Author(s):  
Xiao-yan Fu ◽  
Shuai Zhang ◽  
Kun Wang ◽  
Ming-feng Yang ◽  
Cun-dong Fan ◽  
...  

Molecules ◽  
2018 ◽  
Vol 23 (5) ◽  
pp. 1072 ◽  
Author(s):  
Li Li ◽  
Yi Yang ◽  
Mingxia Wu ◽  
Zanyang Yu ◽  
Chengqiang Wang ◽  
...  

2012 ◽  
Vol 28 (6) ◽  
pp. 2278-2284 ◽  
Author(s):  
RUIJIAN ZHANG ◽  
RUIJUN WANG ◽  
HONG CHANG ◽  
FEI WU ◽  
CHUNTAO LIU ◽  
...  

2019 ◽  
Author(s):  
Hardeep Kaur ◽  
GN Krishnaprasad ◽  
Michael Lichten

AbstractIn Saccharomyces cerevisiae, the conserved Sgs1-Top3-Rmi1 helicase-decatenase regulates homologous recombination by limiting accumulation of recombination intermediates that are precursors of crossovers. In vitro studies have suggested that the dissolution of double-Holliday junction joint molecules by Sgs1-driven convergent junction migration and Top3-Rmi1 mediated strand decatenation could be responsible for this. To ask if dissolution occurs in vivo, we conditionally depleted Sgs1 and/or Rmi1 during return to growth, a procedure where recombination intermediates formed during meiosis are resolved when cells resume the mitotic cell cycle. Sgs1 depletion during return to growth delayed joint molecule resolution, but ultimately most were resolved and cells divided normally. In contrast, Rmi1 depletion resulted in delayed and incomplete joint molecule resolution, and most cells did not divide. rad9Δ mutation restored cell division in Rmi1-depleted cells, indicating that the DNA damage checkpoint caused this cell cycle arrest. Restored cell division in rad9Δ, Rmi1-depleted cells frequently produced anucleate cells, consistent with the suggestion that persistent recombination intermediates prevented chromosome segregation. Our findings indicate that Sgs1-Top3-Rmi1 acts in vivo, as it does in vitro, to promote recombination intermediate resolution by dissolution. They also indicate that, in the absence of Top3-Rmi1 activity, unresolved recombination intermediates persist and activate the DNA damage response, which is usually thought to be activated by much earlier DNA damage-associated lesions.


Oncotarget ◽  
2019 ◽  
Vol 10 (29) ◽  
pp. 2824-2834 ◽  
Author(s):  
Atif J. Khan ◽  
Stephanie LaCava ◽  
Monal Mehta ◽  
Devora Schiff ◽  
Aditya Thandoni ◽  
...  

2018 ◽  
Vol 120 (1) ◽  
pp. 622-633 ◽  
Author(s):  
Dandan Song ◽  
Hongsheng Liang ◽  
Bo Qu ◽  
Yijing Li ◽  
Jingjing Liu ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (13) ◽  
pp. 4136-4145 ◽  
Author(s):  
Mingli Yang ◽  
Song Wu ◽  
Xuekun Su ◽  
W. Stratford May

Abstract We previously identified JAZ as a novel zinc finger (ZF) protein by screening a murine interleukin-3 (IL-3)–dependent NFS/N1.H7 myeloid cell cDNA library. JAZ is a member of a new class of ZFPs that is evolutionarily conserved and preferentially binds to dsRNA, but its function was unknown. Now, we report that the stress of IL-3 growth factor withdrawal up-regulates JAZ expression in hematopoietic cells in association with p53 activation and induction of cell death. Biochemical analysis reveals that JAZ associates with p53 to stimulate its transcriptional activity in p53-expressing cells, but not in p53-null cells unless complemented with p53. JAZ functions to mediate G1 cell-cycle arrest followed by apoptosis in a p53-dependent mechanism that is associated with up-regulation of p21 and BAX, dephosphorylation of Rb, and repression of cyclin A. Of importance, siRNA “knockdown” of endogenous JAZ inhibits p53 transcriptional activity, decreases the G1/G0 population, and attenuates stress-induced cell death. While JAZ directly binds p53 in vitro in a mechanism requiring p53's C-terminal regulatory domain but independent of dsRNA, the dsRNA-binding ZF domains are required for JAZ's stimulatory role of p53 in vivo by dictating its nuclear localization. Thus, JAZ is a novel negative regulator of cell growth by positively regulating p53.


Sign in / Sign up

Export Citation Format

Share Document