scholarly journals Enhancement of radiosensitivity in human glioblastoma cells by the DNA N-mustard alkylating agent BO-1051 through augmented and sustained DNA damage response

2011 ◽  
Vol 6 (1) ◽  
pp. 7 ◽  
Author(s):  
Pei-Ming Chu ◽  
Shih-Hwa Chiou ◽  
Tsann-Long Su ◽  
Yi-Jang Lee ◽  
Li-Hsin Chen ◽  
...  
2019 ◽  
Vol 133 ◽  
pp. 77-86 ◽  
Author(s):  
Ramon Lopez Perez ◽  
Nils H. Nicolay ◽  
Jörg-Christian Wolf ◽  
Moritz Frister ◽  
Peter Schmezer ◽  
...  

2021 ◽  
Author(s):  
Rodolfo Bortolozo Serafim ◽  
Cibele Cardoso ◽  
Vanessa Arfelli ◽  
Valeria Valente ◽  
Leticia Fröhlich Archangelo

Abstract PIMREG expression strongly correlates with cellular proliferation in both malignant and normal cells. Throughout embryo development, PIMREG expression is prominent at the central nervous system. Recent studies have described high levels of PIMREG transcripts in different types of tumors and correlated with patient survival and tumor aggressiveness. Given the emerging significance of PIMREG in carcinogenesis and its putative role in the context of the nervous system, we investigated the expression and function of PIMREG in gliomas, the most common primary brain tumors. We performed an extensive analysis of PIMREG expression in tumors samples of glioma patients, assessed the effects of PIMREG silencing and overexpression on the sensitivity of glioblastoma cell lines treated with genotoxic agents commonly used for treating patients and assessed for treatment response, proliferation and migration. We show that glioblastoma exhibits the highest levels of PIMREG expression among all cancers analyzed and that elevated PIMREG expression is a biomarker for glioma progression and patient outcome. Moreover, PIMREG is induced by genotoxic agents and its silencing renders glioblastoma cells sensitive to temozolomide treatment and affects ATR- and ATM-dependent signaling. Our data demonstrate that PIMREG plays a role in DNA damage response and temozolomide resistance of glioblastoma cells and further support the PIMREG role in tumorigenesis.


Tumor Biology ◽  
2015 ◽  
Vol 37 (6) ◽  
pp. 7719-7727 ◽  
Author(s):  
Andrej Besse ◽  
Jiri Sana ◽  
Radek Lakomy ◽  
Leos Kren ◽  
Pavel Fadrus ◽  
...  

PLoS ONE ◽  
2010 ◽  
Vol 5 (8) ◽  
pp. e12124 ◽  
Author(s):  
Resham Lal Gurung ◽  
Shi Ni Lim ◽  
Aik Kia Khaw ◽  
Jasmine Fen Fen Soon ◽  
Kirthan Shenoy ◽  
...  

2015 ◽  
Vol 356 (2) ◽  
pp. 525-535 ◽  
Author(s):  
Lotte M.E. Berghauser Pont ◽  
Kishan Naipal ◽  
Jenneke J. Kloezeman ◽  
Subramanian Venkatesan ◽  
Martin van den Bent ◽  
...  

2010 ◽  
Vol 24 (6) ◽  
pp. 424-435 ◽  
Author(s):  
Bo Cui ◽  
Stewart P. Johnson ◽  
Nancy Bullock ◽  
Francis Ali-Osman ◽  
Darell D. Bigner ◽  
...  

Author(s):  
Elena Navarro-Carrasco ◽  
Pedro A. Lazo

BackgroundGlioblastomas treated with temozolomide frequently develop resistance to pharmacological treatments. Therefore, there is a need to find alternative drug targets to reduce treatment resistance based on tumor dependencies. A possibility is to target simultaneously two proteins from different DNA-damage repair pathways to facilitate tumor cell death. Therefore, we tested whether targeting the human chromatin kinase VRK1 by RNA interference can identify this protein as a novel molecular target to reduce the dependence on temozolomide in combination with olaparib, based on synthetic lethality.Materials and MethodsDepletion of VRK1, an enzyme that regulates chromatin dynamic reorganization and facilitates resistance to DNA damage, was performed in glioblastoma cells treated with temozolomide, an alkylating agent used for GBM treatment; and olaparib, an inhibitor of PARP-1, used as sensitizer. Two genetically different human glioblastoma cell lines, LN-18 and LN-229, were used for these experiments. The effect on the DNA-damage response was followed by determination of sequential steps in this process: H4K16ac, γH2AX, H4K20me2, and 53BP1.ResultsThe combination of temozolomide and olaparib increased DNA damage detected by labeling free DNA ends, and chromatin relaxation detected by H4K16ac. The combination of both drugs, at lower doses, resulted in an increase in the DNA damage response detected by the formation of γH2AX and 53BP1 foci. VRK1 depletion did not prevent the generation of DNA damage in TUNEL assays, but significantly impaired the DNA damage response induced by temozolomide and olaparib, and mediated by γH2AX, H4K20me2, and 53BP1. The combination of these drugs in VRK1 depleted cells resulted in an increase of glioblastoma cell death detected by annexin V and the processing of PARP-1 and caspase-3.ConclusionDepletion of the chromatin kinase VRK1 promotes tumor cell death at lower doses of a combination of temozolomide and olaparib treatments, and can be a novel alternative target for therapies based on synthetic lethality.


1993 ◽  
Vol 33 (5) ◽  
pp. 275-279 ◽  
Author(s):  
Wei ZHANG ◽  
Akira HARA ◽  
Takatsugu MURAKAWA ◽  
Shuji NIIKAWA ◽  
Takashi ANDOH ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document