Abstract

BackgroundMigration of breast cancer cells out of a duct or lobule is a prerequisite for invasion and metastasis. However, the factors controlling breast cancer cell migration are not fully elucidated. We previously found that expression of the transcription factor interferon regulatory factor 5 (IRF5) is significantly decreased as a breast lesion progresses from a non-malignant stage to ductal carcinoma in situ and is eventually lost in ~80% of invasive ductal carcinomas examined. Human in vitro and murine in vivo models of invasive breast cancer confirmed an important role for IRF5 in regulating cell motility, invasion and/or metastasis; yet, the mechanism(s) by which this occurs is not known. Since IRF5 is primarily expressed in the cytoplasm of human mammary epithelial cells, we hypothesized that IRF5 may function in a transcription-independent manner to control intrinsic cell migration.ResultsA series of IRF5 deletion mutants were tested in cell motility, invasion and migration assays. A novel, conserved 10 amino acid domain was identified that regulates mammary epithelial cell migration. This region (∆115-125) is downstream of IRF5′s DNA binding domain and therefore when absent, retains IRF5 transcription activity but loses cell migration control. An IRF5 construct with a mutated nuclear localization signal further confirmed that IRF5 controls migration in a cytoplasmic and transcription-independent manner. Candidate cytoskeletal molecules were identified in MDA-MB-231 cells to interact with IRF5 by immunoprecipitation and mass spectrometry analysis. α6-tubulin was independently confirmed to interact with endogenous IRF5 in MCF-10A cells. Alterations in F-actin bundling after staining EV- and IRF5-231 cells with phalloidin suggests that IRF5 may control cell migration/motility through its interaction with cytoskeletal molecules that contribute to the formation of F-actin networks. Last and most notably, we found that IRF5′s control of cell migration is not restricted to mammary epithelial cells but functions in other epithelial cell types suggesting a more global role for this newly identified cell migratory function of IRF5.ConclusionsThese findings are significant as they identify a new regulator of epithelial cell migration and provide specific insight into the mechanism(s) by which loss of IRF5 expression in mammary epithelial cells contributes to breast cancer metastasis.

Highlights

  • Interferon regulatory factor 5 (IRF5) is a member of the IRF family of transcription factors that are expressed in vertebrates

  • interferon regulatory factor 5 (IRF5) expression inhibits the migration of breast cancer cells Experimental in vitro and in vivo data support a role for IRF5 in mammary epithelial cell migration and metastasis

  • Given that all three of these cytoskeletal molecules have been implicated in cancer migration, invasion and metastasis formation [33], in part, through their ability to participate in filamentous (F)-actin networking that is integral for cell motility [33,34], we examined F-actin networking in EV-231 and IRF5-231 cells and found distinct and condensed F-actin bundling at the nucleus of IRF5-231 cells as compared with EV-231 cells (Figure 6B)

Read more

Summary

Introduction

Interferon regulatory factor 5 (IRF5) is a member of the IRF family of transcription factors that are expressed in vertebrates. Most well known is the induction of IRF5 activation by MyD88-dependent Toll like receptor (TLR) signaling [7,12,13,14] In this pathway, ligand bound to receptor induces a cascade of events whereby IRF5 binds to MyD88 and TRAF6 [3,5,13,14], undergoes several post-translational modifications including phosphorylation and possibly ubiquitination [10], and forms hetero- or homodimers that translocate to the nucleus [4,6,12,13,15,16]. IRF5 has been shown to positively regulate cytokines such as Type I interferons (IFNs), interleukin (IL)-6, IL-12, IL-1β, and TNF-α while suppressing IL-10 expression [11,14] Most studies of this protein to date have been performed in lymphocytes, IRF5 is classically thought of as a transcriptional immune regulator [4,7,11,15]. Since IRF5 is primarily expressed in the cytoplasm of human mammary epithelial cells, we hypothesized that IRF5 may function in a transcription-independent manner to control intrinsic cell migration

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