Abstract

Epithelial ovarian cancer (EOC) generates multicellular aggregates called spheroids that detach from the primary tumor and disseminate through ascites. Spheroids possess a number of characteristics of tumor dormancy including withdrawal from the cell cycle and resistance to chemotherapeutics. This report systematically analyzes the effects of RNAi depletion of 21 genes that are known to contribute to negative regulation of the cell cycle in 10 ovarian cancer cell lines. Interestingly, spheroid cell viability was compromised by loss of some cyclin-dependent kinase inhibitors such as p57Kip2, as well as Dyrk1A, Lin52, and E2F5 in most cell lines tested. Many genes essential for EOC spheroid viability are pertinent to the mammalian DREAM repressor complex. Mechanistically, the data demonstrate that DREAM is assembled upon the induction of spheroid formation, which is dependent upon Dyrk1A. Loss of Dyrk1A results in retention of the b-Myb-MuvB complex, elevated expression of DREAM target genes, and increased DNA synthesis that is coincident with cell death. Inhibition of Dyrk1A activity using pharmacologic agents Harmine and INDY compromises viability of spheroids and blocks DREAM assembly. In addition, INDY treatment improves the response to carboplatin, suggesting this is a therapeutic target for EOC treatment.Implications: Loss of negative growth control mechanisms in cancer dormancy lead to cell death and not proliferation, suggesting they are an attractive therapeutic approach. Mol Cancer Res; 15(4); 371-81. ©2016 AACR.

Highlights

  • Metastatic dissemination of cancer cells is the major source of cancer-related mortality [1]

  • We show that Dyrk1A is essential for DREAM assembly in spheroids, cell-cycle exit, and that DREAM target genes are upregulated as a result

  • Because of the heterogenous mutational landscape in epithelial ovarian cancer (EOC) [23], knockdowns were performed in 10 cell lines from gynecologic malignancies including seven cell lines derived from the ascites obtained from patients presenting with EOC and three established EOC cell lines (OVCAR5, OVCAR8, and HEY; ref. 24)

Read more

Summary

Introduction

Metastatic dissemination of cancer cells is the major source of cancer-related mortality [1]. These cells may enter dormancy and as a consequence become impervious to standard chemotherapy paradigms dependent on DNA replication and a highly active metabolic state [1, 2]. Tumor cell dormancy is relevant to malignancies that generate ascites such as epithelial ovarian cancer (EOC). These tumors shed cells into the ascites which aggregate to form multicellular clusters, or spheroids. The persistence of drug-resistant EOC cells remains a major hurdle to successful cancer treatment and underscores the necessity to understand biochemical mechanisms critical to the

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