Abstract

The p53-mediated nucleolar stress response associated with inhibition of ribosomal RNA transcription was previously shown to potentiate killing of tumor cells. Here, we asked whether targeting of ribosome biogenesis can be used as the basis for selective p53-dependent cytoprotection of nonmalignant cells. Temporary functional inactivation of the 60S ribosome assembly factor Bop1 in a 3T3 cell model markedly increased cell recovery after exposure to camptothecin or methotrexate. This was due, at least in part, to reversible pausing of the cell cycle preventing S phase associated DNA damage. Similar cytoprotective effects were observed after transient shRNA-mediated silencing of Rps19, but not several other tested ribosomal proteins, indicating distinct cellular responses to the inhibition of different steps in ribosome biogenesis. By temporarily inactivating Bop1 function, we further demonstrate selective killing of p53-deficient cells with camptothecin while sparing isogenic p53-positive cells. Thus, combining cytotoxic treatments with inhibition of select post-transcriptional steps of ribosome biogenesis holds potential for therapeutic targeting of cells that have lost p53.

Highlights

  • The curative potential of chemotherapeutic agents is limited by their toxicity toward normal cells

  • To investigate whether short-term targeting of specific steps in ribosome biogenesis could fulfill these criteria, we performed cell survival assays in which we split cells into two treatment groups, temporarily disrupted ribosome biogenesis in one of the groups, and treated all cells with a cytotoxic chemical (Fig. 1a)

  • We began to examine the effects of targeting post-transcriptional ribosome biogenesis steps by reversibly inhibiting function of the conserved 60S subunit ribosome assembly factor Bop[152, 53]

Read more

Summary

Introduction

The curative potential of chemotherapeutic agents is limited by their toxicity toward normal cells. Compared with Pol I transcription, ribosome assembly is significantly more complex[27, 28], offering a wide diversity of targets that could be potentially exploited for activating p53 in a nongenotoxic manner. It remains to be established whether transiently inhibiting post-transcriptional ribosome biogenesis steps can elicit prosurvival responses, suitable for modulating cell sensitivity to chemotherapeutic agents. Selective killing of p53-deficient cells while protecting p53-positive cells can be achieved These findings suggest that targeting specific post-transcriptional ribosome assembly steps can be used in combination with existing cytotoxic drugs to design improved therapeutic interventions in cancer

Methods
Results
Conclusion
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