Abstract

Simple SummaryNeddylation is a process in which the small ubiquitin-like molecule NEDD8 is covalently conjugated to target proteins by sequential enzymatic reactions. Because neddylation plays critical roles in regulating cancer growth and migration, it is emerging as an effective therapeutic target. The major tumor suppressor protein p53 reduces cancer cell migration and is inhibited by neddylation. As p53 is lost or mutated in 50% of various cancer types, this study attempted to investigate how neddylation affects cancer cell migration according to p53 status. Neddylation blockade reduced or caused no change in migration of wild type or mutant p53 cancer cell lines. In contrast, neddylation blockade induced migration of p53-null cancer cell lines. These results were mediated by the differential effect of neddylation blockade on the epithelial–mesenchymal transition activator Slug according to p53 status. Thus, the p53 status of cancer cells should be considered when developing neddylation-targeted anticancer drugs.The tumor suppressor protein p53 is frequently inactivated in human malignancies, in which it is associated with cancer aggressiveness and metastasis. Because p53 is heavily involved in epithelial–mesenchymal transition (EMT), a primary step in cell migration, p53 regulation is important for preventing cancer metastasis. p53 function can be modulated by diverse post-translational modifications including neddylation, a reversible process that conjugates NEDD8 to target proteins and inhibits the transcriptional activity of p53. However, the role of p53 in cancer migration by neddylation has not been fully elucidated. In this study, we reported that neddylation blockade induces cell migration depending on p53 status, specifically via the EMT-promoting transcription factor Slug. In cancer cell lines expressing wild type p53, neddylation blockade increased the transcriptional activity of p53 and expression of its downstream genes p21 and MDM2, eventually promoting proteasomal degradation of Slug. In the absence of p53, neddylation blockade increased cell migration by activating the PI3K/Akt/mTOR/Slug signaling axis. Because mutant p53 was transcriptionally inactivated but maintained the ability to bind to Slug, neddylation blockade did not affect the migration of cells expressing mutant p53. Our findings highlight how the p53 expression status influences neddylation-mediated cell migration in multiple cancer cell lines via Slug.

Highlights

  • Cancer metastasis is a sequential process by which cancer cells spread from their original site to distant organs

  • Neddylation Blockade Induces Cancer Cell Migration according to p53 Status

  • Given that neddylation blockade increases the migration of p53-null cancer cell lines [20,25,28], we investigated the relationship between p53 and neddylation blockade in cancer cell migration using different cancer cell lines with wild type p53 or p53-null characteristics

Read more

Summary

Introduction

Cancer metastasis is a sequential process by which cancer cells spread from their original site to distant organs. Metastatic cancer is the leading cause of cancer-related mortality [1] and elucidating the dynamics of cancer metastasis is regarded as a powerful strategy for effective cancer treatment. The EMT process involves EMT-related transcription factors such as Snail, Zeb, Twist, and Slug [8]. These factors promote EMT by binding to E-box domain of E-cadherin promoter, a major epithelial gene, and repressing its expression [9]. Loss of p53 function promotes cell migration and tumor metastasis [14]. More than 50% of human cancers involve p53 deletion or a functional mutation in p53 [15]; a better understanding of the relationship between p53 disruption and EMT will help identify the cause of metastasis in affected cancers

Methods
Results
Discussion
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