Cancers | 2021

Quinacrine Induces Nucleolar Stress in Treatment-Refractory Ovarian Cancer Cell Lines

 
 
 
 
 
 
 
 
 
 
 
 

Abstract


Simple Summary A high mortality rate in ovarian cancer imposes the need for improved therapy; most patients initially respond to systemic chemotherapy but later relapse with fatal treatment-refractory tumors. We previously evaluated repurposing the antimalarial agent quinacrine as an anticancer agent in chemoresistant ovarian cancer. Quinacrine has a long history of use in humans and we have demonstrated selectivity characteristics for reducing drug resistant tumors. In this study, we analyze different responses to quinacrine between drug resistant and sensitive cell lines to identify major pathways related to this selectivity. We confirm our results in notoriously drug resistant high-grade serous ovarian cancer cells and describe nucleostemin as a new quinacrine target related to ribosomal biogenesis and nucleolar stress. This study provides preclinical evidence that quinacrine may be effective against relapsed/refractory ovarian cancer. Abstract A considerable subset of gynecologic cancer patients experience disease recurrence or acquired resistance, which contributes to high mortality rates in ovarian cancer (OC). Our prior studies showed that quinacrine (QC), an antimalarial drug, enhanced chemotherapy sensitivity in treatment-refractory OC cells, including artificially generated chemoresistant and high-grade serous OC cells. In this study, we investigated QC-induced transcriptomic changes to uncover its cytotoxic mechanisms of action. Isogenic pairs of OC cells generated to be chemoresistant and their chemosensitive counterparts were treated with QC followed by RNA-seq analysis. Validation of selected expression results and database comparison analyses indicated the ribosomal biogenesis (RBG) pathway is inhibited by QC. RBG is commonly upregulated in cancer cells and is emerging as a drug target. We found that QC attenuates the in vitro and in vivo expression of nucleostemin (NS/GNL3), a nucleolar RBG and DNA repair protein, and the RPA194 catalytic subunit of Pol I that results in RBG inhibition and nucleolar stress. QC promotes the redistribution of fibrillarin in the form of extranuclear foci and nucleolar caps, an indicator of nucleolar stress conditions. In addition, we found that QC-induced downregulation of NS disrupted homologous recombination repair both by reducing NS protein levels and PARylation resulting in reduced RAD51 recruitment to DNA damage. Our data suggest that QC inhibits RBG and this inhibition promotes DNA damage by directly downregulating the NS–RAD51 interaction. Additionally, QC showed strong synergy with PARP inhibitors in OC cells. Overall, we found that QC downregulates the RBG pathway, induces nucleolar stress, supports the increase of DNA damage, and sensitizes cells to PARP inhibition, which supports new therapeutic stratagems for treatment-refractory OC. Our work offers support for targeting RBG in OC and determines NS to be a novel target for QC.

Volume 13
Pages None
DOI 10.3390/cancers13184645
Language English
Journal Cancers

Full Text