Abstract

Temozolomide (TMZ) is a DNA methylating agent used to treat brain cancer. TMZ-induced O6-methylguanine adducts, in the absence of repair by O6-methylguanine DNA methyltransferase (MGMT), mispair during DNA replication and trigger cycles of futile mismatch repair (MMR). Futile MMR in turn leads to the formation of DNA single and double strand breaks, Chk1 and Chk2 phosphorylation/activation, cell cycle arrest, and ultimately cell death. Although both pChk1 and pChk2 are considered to be biomarkers of TMZ-induced DNA damage, cell-cycle arrest, and TMZ induced cytotoxicity, we found that levels of pChk1 (ser345), its downstream target pCdc25C (ser216), and the activity of its upstream activator ATR, were elevated within 3 hours of TMZ exposure, long before the onset of TMZ-induced DNA double strand breaks, Chk2 phosphorylation/activation, and cell cycle arrest. Furthermore, TMZ-induced early phosphorylation of Chk1 was noted in glioma cells regardless of whether they were MGMT-proficient or MGMT-deficient, and regardless of their MMR status. Early Chk1 phosphorylation was not associated with TMZ-induced reactive oxygen species, but was temporally associated with TMZ-induced alkalai-labile DNA damage produced by the non-O6-methylguanine DNA adducts and which, like Chk1 phosphorylation, was transient in MGMT-proficient cells but persistent in MGMT-deficient cells. These results re-define the TMZ-induced DNA damage response, and show that Chk1 phosphorylation is driven by TMZ-induced mismatch repair-independent DNA damage independently of DNA double strand breaks, Chk2 activation, and cell cycle arrest, and as such is a suboptimal biomarker of TMZ-induced drug action.

Highlights

  • Temozolomide (TMZ) is a chemotherapeutic DNA methylating agent used in the treatment of a variety of malignancies

  • By following the formation of this damage after TMZ exposure and comparing its appearance with activation of the DNA damage response, we here show that the Chk1 phosphorylation previously linked only to TMZ-induced mismatch repair (MMR)-dependent DNA damage in TMZ-sensitive cells occurs in all GBM cells examined long before the activation of Chk2, creation of MMR-dependent TMZ-induced DNA damage, and activation of the G2 checkpoint

  • In studies in which cells were exposed to physiologic, near IC50 concentrations of temozolomide or the related SN1 methylating agent MNNG, and in which effects were monitored 48 hrs after drug exposure, Chk1 phosphorylation was associated with events linked to MMR-dependent processing of drug-induced O6-methyl guanine (O6MG) lesions and drug-induced G2 arrest [26,27]

Read more

Summary

Introduction

Temozolomide (TMZ) is a chemotherapeutic DNA methylating agent used in the treatment of a variety of malignancies. Because published studies have only examined Chk phosphorylation/activation at time points following the resolution of TMZ-induced non-O6MG lesions (.12 hrs post TMZ exposure), or after super-physiologic concentrations of methylating agents [30], we considered the possibility that Chk activation following TMZ exposure may be a consequence of non-cytotoxic, non O6MG, TMZinduced DNA damage, and as such may be a sub-optimal biomarker of TMZ action To address this possibility we examined Chk and Chk phosphorylation at both early and later time points following exposure to clinically achievable concentration of TMZ in isogenic GBM cells in which TMZ sensitivity was genetically or pharmacologically manipulated. These results show that Chk phosphorylation/activation, contrary to previous assumptions, is driven by TMZ-induced mismatch repair-independent, single-stranded DNA damage and is likely a suboptimal biomarker of TMZ-induced drug action

Experimental Procedures
Results
Discussion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call