Abstract

Recent findings demonstrate that aberrant downregulation of the iron-exporter protein, ferroportin (FPN1), is associated with poor prognosis and osteoclast differentiation in multiple myeloma (MM). Here, we show that FPN1 was downregulated in MM and that clustered regularly interspaced short palindromic repeat (CRISPR)-mediated FPN1 knockout promoted MM cell growth and survival. Using a microRNA target-scan algorithm, we identified miR-17-5p as an FPN1 regulator that promoted cell proliferation and cell cycle progression, and inhibited apoptosis—both in vitro and in vivo. miR-17-5p inhibited retarded tumor growth in a MM xenograft model. Moreover, restoring FPN1 expression at least partially abrogated the biological effects of miR-17-5p in MM cells. The cellular iron concentration regulated the expression of the iron-regulatory protein (IRP) via the 5′-untranslated region of IRP messenger RNA and modulated the post-transcriptional stability of FPN1. Bioinformatics analysis with subsequent chromatin immunoprecipitation-polymerase chain reaction and luciferase activity experiments revealed that the transcription factor Nrf2 drove FPN1 transcription through promoter binding and suppressed miR-17-5p (which also increased FPN1 expression). Nrf2-mediated FPN1 downregulation promoted intracellular iron accumulation and reactive oxygen species. Our study links FPN1 transcriptional and post-transcriptional regulation with MM cell growth and survival, and validates the prognostic value of FPN1 and its utility as a novel therapeutic target in MM.

Highlights

  • Iron is an essential nutrient for various cellular processes, including DNA replication, cell-cycle progression, heme synthesis, and enzyme-mediated functions

  • We demonstrated that clustered regularly interspaced short palindromic repeat (CRISPR)-mediated FPN1 knockout in MM cells promoted liable iron pool (LIP) and reactive oxygen species (ROS) accumulation, which are important for malignant cell growth and survival

  • To investigate whether miRNAs are associated with aberrant FPN1 expression and MM progression, online miRNA target-prediction tools were used to generate FPN1-targeting miRNA candidates

Read more

Summary

Introduction

Iron is an essential nutrient for various cellular processes, including DNA replication, cell-cycle progression, heme synthesis, and enzyme-mediated functions. Cellular iron homeostasis is regulated by a sophisticated system that responds to intracellular iron levels and a sophisticated gene network that maintains the intracellular storage, utilization, and export of iron[1]. Ferroportin (FPN1; known as SLC40A1) is a cell-. Many cancers exhibit an increased need for intracellular iron and persistent iron stimulation can increase the risk for tumorigenesis[8]. Given the important iron-regulatory role of FPN1, its dysregulation may contribute to persistent iron stimulation[9] and cancer development[10]. FPN1 expression is regulated at the messenger RNA (mRNA). Official journal of the Cell Death Differentiation Association. Kong et al Cell Death and Disease (2019)10:624. Iron released from hemoglobin regulates iron exporter FPN1 translation, involving the IRE within the FPN1 5′-

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