Abstract

Cancer cell stemness is essential for enabling malignant progression and clonal evolution. Cancer cell fate is likely determined by complex mechanisms involving both cell-intrinsic pathways and stress signals from tumor microenvironment. In this study, we examined the role of the tumor suppressor BRCA1 and hypoxia in the regulation of cancer cell stemness using genetically matched breast cancer cell lines. We have found that BRCA1, a multifunctional protein involved in DNA repair and epigenetic regulation, plays a critical role in the regulation of cancer stem cell (CSC)-like characteristics. Reconstitution of BRCA1 resulted in significant decrease of the CSC-like populations in breast cancer cells whereas down-regulation of BRCA1 resulted in significant increase of the CSC-like populations. Furthermore, the BRCA1-reconstituted tumor cells are more sensitive to the histone deacetylase (HDAC) inhibitor-induced loss of stemness than the BRCA1-deficient cells are. Surprisingly, hypoxia preferentially blocks HDAC inhibitor-induced differentiation of the BRCA1-reconstituted breast cancer cells. In light of the increasing numbers of clinical trials involving HDAC inhibitors in human cancers, our observations strongly suggest that the BRCA1 status and tumor hypoxia should be considered as potentially important clinical parameters that may affect the therapeutic efficacy of HDAC inhibitors.

Highlights

  • Tumor cells, even cell lines in vitro, consist of mixed populations some of which are capable of tumor initiation and possess stem cell-like characteristics

  • We found that ALDH1A1 was significantly (p = 0.0012) down-regulated in BRCA1-reconstituted cells

  • The cell fate of cancer cells is likely to be determined by both cell-intrinsic pathways and by stress signals from tumor microenvironment

Read more

Summary

Introduction

Even cell lines in vitro, consist of mixed populations some of which are capable of tumor initiation and possess stem cell-like characteristics. BRCA1 has been shown to be required for differentiation of mammary stem/progenitor cells to luminal epithelial cells[10,11], suggesting that BRCA1 constitutes an important intrinsic pathway involved in cell fate determination. Increasing evidence shows that hypoxia, a condition of oxygen deficiency and a hallmark of tumor microenvironment (TME), up-regulates CSC-related genes, promotes self-renewal and suppresses cell differentiation[15,16]. A number of in vitro studies have shown that hypoxia or hypoxia-sensing pathways play a significant role in the maintenance of the CSC phenotype in breast cancer cells[17,18,19,20,21,22,23].

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