scholarly journals Sp1 Contributes to Radioresistance of Cervical Cancer Through Targeting G2/M Cell Cycle Checkpoint CDK1 [Corrigendum]

2021 ◽  
Vol Volume 13 ◽  
pp. 8525-8526
Author(s):  
Yuan-Run Deng ◽  
Xiao-Jing Chen ◽  
Wei Chen ◽  
Lan-Fang Wu ◽  
Hui-Ping Jiang ◽  
...  
2019 ◽  
Vol Volume 11 ◽  
pp. 5835-5844 ◽  
Author(s):  
Yuan-Run Deng ◽  
Xiao-Jing Chen ◽  
Wei Chen ◽  
Lan-Fang Wu ◽  
Hui-Ping Jiang ◽  
...  

2005 ◽  
Vol 62 (5) ◽  
pp. 1390-1398 ◽  
Author(s):  
Ferdinando Cerciello ◽  
Barbara Hofstetter ◽  
Sherweif Abdel Fatah ◽  
Mohamed Zaghloul ◽  
Bernhard Odermatt ◽  
...  

2007 ◽  
Vol 67 (13) ◽  
pp. 6286-6292 ◽  
Author(s):  
Kazuhiko Yamane ◽  
Jane E. Schupp ◽  
Timothy J. Kinsella

Oncogene ◽  
2007 ◽  
Vol 26 (40) ◽  
pp. 5851-5865 ◽  
Author(s):  
E Schmitt ◽  
M Beauchemin ◽  
R Bertrand

2007 ◽  
Vol 81 (12) ◽  
pp. 6718-6730 ◽  
Author(s):  
Tathagata Choudhuri ◽  
Subhash C. Verma ◽  
Ke Lan ◽  
Masanao Murakami ◽  
Erle S. Robertson

ABSTRACT Epstein-Barr virus (EBV) infects most of the human population and persists in B lymphocytes for the lifetime of the host. The establishment of latent infection by EBV requires the expression of a unique repertoire of genes. The product of one of these viral genes, the EBV nuclear antigen 3C (EBNA3C), is essential for the growth transformation of primary B lymphocytes in vitro and can regulate the transcription of a number of viral and cellular genes important for the immortalization process. This study demonstrates an associated function of EBNA3C which involves the disruption of the G2/M cell cycle checkpoint. We show that EBNA3C-expressing lymphoblastoid cell lines treated with the drug nocodazole, which is known to block cells at the G2/M transition, did not show a G2/M-specific checkpoint arrest. Analyses of the cell cycles of cells expressing EBNA3C demonstrated that the expression of this essential EBV nuclear antigen is capable of releasing the G2/M checkpoint arrest induced by nocodazole. This G2/M arrest in response to nocodazole was also abolished by caffeine, suggesting an involvement of the ATM/ATR signaling pathway in the regulation of this cell cycle checkpoint. Importantly, we show that the direct interaction of EBNA3C with Chk2, the ATM/ATR signaling effector, is responsible for the release of this nocodazole-induced G2/M arrest and that this interaction leads to the serine 216 phosphorylation of Cdc25c, which is sequestered in the cytoplasm by 14-3-3. Overall, our data suggest that EBNA3C can directly regulate the G2/M component of the host cell cycle machinery, allowing for the release of the checkpoint block.


2000 ◽  
Vol 276 (2) ◽  
pp. 1127-1132 ◽  
Author(s):  
Jaleh Doostzadeh-Cizeron ◽  
Nicholas H. A. Terry ◽  
David W. Goodrich

2006 ◽  
Vol 25 (4) ◽  
pp. 763-773 ◽  
Author(s):  
Gustavo Pedraza-Alva ◽  
Miroslav Koulnis ◽  
Colette Charland ◽  
Tina Thornton ◽  
James L Clements ◽  
...  

2002 ◽  
Vol 22 (22) ◽  
pp. 7831-7841 ◽  
Author(s):  
Eugene S. Kandel ◽  
Jennifer Skeen ◽  
Nathan Majewski ◽  
Antonio Di Cristofano ◽  
Pier Paolo Pandolfi ◽  
...  

ABSTRACT Activation of Akt, or protein kinase B, is frequently observed in human cancers. Here we report that Akt activation via overexpression of a constitutively active form or via the loss of PTEN can overcome a G2/M cell cycle checkpoint that is induced by DNA damage. Activated Akt also alleviates the reduction in CDC2 activity and mitotic index upon exposure to DNA damage. In addition, we found that PTEN null embryonic stem (ES) cells transit faster from the G2/M to the G1 phase of the cell cycle when compared to wild-type ES cells and that inhibition of phosphoinositol-3-kinase (PI3K) in HEK293 cells elicits G2 arrest that is alleviated by activated Akt. Furthermore, the transition from the G2/M to the G1 phase of the cell cycle in Akt1 null mouse embryo fibroblasts (MEFs) is attenuated when compared to that of wild-type MEFs. These results indicate that the PI3K/PTEN/Akt pathway plays a role in the regulation of G2/M transition. Thus, cells expressing activated Akt continue to divide, without being eliminated by apoptosis, in the presence of continuous exposure to mutagen and accumulate mutations, as measured by inactivation of an exogenously expressed herpes simplex virus thymidine kinase (HSV-tk) gene. This phenotype is independent of p53 status and cannot be reproduced by overexpression of Bcl-2 or Myc and Bcl-2 but seems to counteract a cell cycle checkpoint mediated by DNA mismatch repair (MMR). Accordingly, restoration of the G2/M cell cycle checkpoint and apoptosis in MMR-deficient cells, through reintroduction of the missing component of MMR, is alleviated by activated Akt. We suggest that this new activity of Akt in conjunction with its antiapoptotic activity may contribute to genetic instability and could explain its frequent activation in human cancers.


2007 ◽  
Vol 27 (2) ◽  
pp. 214-227 ◽  
Author(s):  
Feng Zhu ◽  
Xiaojun Xia ◽  
Bigang Liu ◽  
Jianjun Shen ◽  
Yuhui Hu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document