Abstract

Primary cilia are lost during cancer development, but the mechanism regulating cilia degeneration is not determined. While transcription factor nuclear factor-erythroid 2-like 2 (NRF2) protects cells from oxidative, proteotoxic, and metabolic stress in normal cells, hyperactivation of NRF2 is oncogenic, although the detailed molecular mechanisms by which uncontrolled NRF2 activation promotes cancer progression remain unclear. Here, we report that NRF2 suppresses hedgehog (Hh) signaling through Patched 1 (PTCH1) and primary ciliogenesis via p62/sequestosome 1 (SQSTM1). PTCH1, a negative regulator of Hh signaling, is an NRF2 target gene, and as such, hyperactivation of NRF2 impairs Hh signaling. NRF2 also suppresses primary cilia formation through p62-dependent inclusion body formation and blockage of Bardet–Biedl syndrome 4 (BBS4) entrance into cilia. Simultaneous ablation of PTCH1 and p62 completely abolishes NRF2-mediated inhibition of both primary ciliogenesis and Hh signaling. Our findings reveal a previously unidentified role of NRF2 in controlling a cellular organelle, the primary cilium, and its associated Hh signaling pathway and also uncover a mechanism by which NRF2 hyperactivation promotes tumor progression via primary cilia degeneration and aberrant Hh signaling. A better understanding of the crosstalk between NRF2 and primary cilia/Hh signaling could not only open new avenues for cancer therapeutic discovery but could also have significant implications regarding pathologies other than cancer, including developmental disorders, in which improper primary ciliogenesis and Hh signaling play a major role.

Highlights

  • Nuclear factor-erythroid 2-like 2 (NRF2) is a transcription factor that mediates cellular redox, metabolic and protein homeostasis [1,2]

  • Immunofluorescence (IF) staining for acetylated tubulin (Ac-Tub) and ADP-ribosylation factor-like protein 13B (ARL13B) clearly showed more ciliated cells in the Nrf2−/− mouse embryonic fibroblast (MEF) cells compared to the Nrf2+/+ MEF cells (Fig 1A), as well as higher protein levels of Ac-Tub and ARL13B (Fig 1B, S1A Fig)

  • Consistent with the MEF data, the percentage of ciliated cells and the level of Ac-Tub or ARL13B were significantly higher in NRF2−/− BEAS-2B and NRF2−/− H838 cell lines compared to their respective NRF2+/+ controls (Fig 1A and 1B, S1A Fig)

Read more

Summary

Introduction

Nuclear factor-erythroid 2-like 2 (NRF2) is a transcription factor that mediates cellular redox, metabolic and protein homeostasis [1,2]. NRF2 is negatively regulated by Kelch-like ECH-associated protein 1 (KEAP1), a substrate adaptor protein of the Cullin (Cul3)-Ring-Box 1 (Rbx1) E3-ligase complex that targets NRF2 for ubiquitylation and degradation by the 26S proteasome [3]. KEAP1 functions as a molecular sensor through its cysteines, especially C151, controlling activation of the NRF2 pathway [4]. NRF2 negatively regulates primary cilia and Hh signaling

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