Abstract

Camptothecin (CPT), a topoisomerase (Top) I-targeting drug that stabilizes Top1-DNA covalent adducts, can induce S-phase-specific cytotoxicity due to the arrest of progressing replication forks. However, CPT-induced non-S-phase cytotoxicity is less well characterized. In this study, we have identified topoisomerase IIβ (Top2β) as a specific determinant for CPT sensitivity, but not for many other cytotoxic agents, in non-S-phase cells. First, quiescent mouse embryonic fibroblasts (MEFs) lacking Top2β were shown to be hypersensitive to CPT with prominent induction of apoptosis. Second, ICRF-187, a Top2 catalytic inhibitor known to deplete Top2β, specifically sensitized MEFs to CPT. To explore the molecular basis for CPT hypersensitivity in Top2β-deficient cells, we found that upon CPT exposure, the RNA polymerase II large subunit (RNAP LS) became progressively depleted, followed by recovery to nearly the original level in wild-type MEFs, whereas RNAP LS remained depleted without recovery in Top2β-deficient cells. Concomitant with the reduction of the RNAP LS level, the p53 protein level was greatly induced. Interestingly, RNAP LS depletion has been well documented to lead to p53-dependent apoptosis. Altogether, our findings support a model in which Top2β deficiency promotes CPT-induced apoptosis in quiescent non-S-phase cells, possibly due to RNAP LS depletion and p53 accumulation.

Highlights

  • The sensitivity of non-S-phase cells to Top1-targeting drugs has been under-explored

  • Top2␤ Deficiency Sensitizes Quiescent mouse embryonic fibroblasts (MEFs) to Camptothecin-induced Cell Death—Top1-targeting drugs (e.g. CPT) are known to induce S-phase-specific cytotoxicity; this is largely attributed to the arrest of the advancing replication forks by the drug-induced Top1 cleavage complexes [22]

  • Using serum-starved primary MEFs as a model, we have previously shown that Top2␤ knock-out MEFs (Top2␤Ϫ/Ϫ) were highly resistant to Top2-targeting drugs (e.g. VP-16) as compared with wild-type MEFs (Top2␤ϩ/ϩ) [36, 37]

Read more

Summary

Background

The sensitivity of non-S-phase cells to Top1-targeting drugs has been under-explored. Our findings support a model in which Top2␤ deficiency promotes CPT-induced apoptosis in quiescent nonS-phase cells, possibly due to RNAP LS depletion and p53 accumulation. Camptothecin (CPT), a Top1-targeting drug, can interfere with the catalytic cycle of Top by inhibiting the religation reaction and increases the half-life of the transient Top1DNA covalent adducts ( known as Top cleavage complexes) [1, 20]. It has been demonstrated that the collision of Top cleavage complexes with DNA replication machineries and the subsequently generated DNA double strand breaks are responsible for the S-phase-specific cytotoxicity of CPT [22]. CPT treatment, possibly due to the reduced replication-mediated Top cleavage complex processing [25] This S-phase-selective cytotoxicity enables Top1-based chemotherapy to be efficacious in treating several types of cancers, including ovarian, lung, and colon cancers [20, 24, 26]. The increased sensitivity is associated with the depletion of RNA polymerase II large subunit (RNAP LS), p53 accumulation, general transcription inhibition, and apoptosis induction

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