Elsevier

Experimental Cell Research

Volume 359, Issue 2, 15 October 2017, Pages 405-414
Experimental Cell Research

GSKJ4, an H3K27me3 demethylase inhibitor, effectively suppresses the breast cancer stem cells

https://doi.org/10.1016/j.yexcr.2017.08.024Get rights and content

Highlights

  • H3K27me3 is closely associated with BCSCs.

  • GSKJ4 could effectively inhibit the self-renewal and expansion of BCSCs.

  • GSKJ4 could serve as a promising target agent for BCSCs.

Abstract

Recently, studies have been suggested that H3K27me3 is implicated with maintenance of cancer stem cells (CSCs), however, the roles of H3K27me3 in Breast cancer stem cells (BCSCs) remain poorly investigated. Here we explore the functionallities of H3K27me3 on BCSCs, we identify H3K27me3 as a negative modulator of BCSCs and suggest GSKJ4 is a promising drug targeting BCSCs. We show that the H3K27me3 level is decreased in mammosphere-derived BCSCs. In breast cancer cells, we demonstrate that GSKJ4 could markedly inhibit the proliferation. Strikingly, we show that GSKJ4 could effectively suppress BCSCs including expansion, self-renewal capacity, and the expression of stemness-related markers. Additionally, our xenograft model confirms that GSKJ4 is able to effectively inhibit the tumorigenicity of MDA-MB-231. Mechanistically, the inhibition effects of GSKJ4 on BCSCs are via inhibiting demethylases JMJD3 and UTX with methyltransferase EZH2 unchanged, which enhances H3K27me3 level. H3K27me3 activating leads to reduction of BCSCs expansion, self-renewal and global level of stemness factors. Collectively, our results provide strong supports that H3K27me3 exerts a suppressive influence on BCSCs and reveal that GSKJ4 is capable to be a prospective agent targeting BCSCs.

Introduction

In the past twenty years, a significant trend towards a worldwide reduction in mortality from breast cancer has been observed. This trend has been largely attributed to improved early detection methods and the development of improved treatment strategies. However, a substantial fraction of breast cancer patients still relapse and even die after five years. Current opinion proposes that this failure is largely due to the existence of breast cancer stem cells (BCSCs), which have unlimited proliferative potential and are responsible for drug resistance and disease relapse [1], [2]. Therefore, therapeutic strategies specifically targeting BCSCs are warranted.

Cancer stem cells (CSCs), which are found in most solid tumours, share with normal stem cells several properties, such as self-renewal, a better ability to repair DNA and resistance to apoptosis and hypoxia. It is conceivable that several key regulatory pathways involved in the regulation of self-renewal and differentiation of stem cells, such as WNT [3], Notch [4] and Sonic Hedgehog [5], are also frequently deregulated in CSCs. In addition, more and more studies have revealed that epigenetic mechanisms play a vital role in the maintenance of stemness in normal stem cells [6], [7], and the influence of epigenetic modifications on self-renewal and the pluripotency of CSCs is gaining a great deal of attention as well [8], [9], [10].

As a critical factor in epigenetic modification, histone methylation is mediated by methyltransferases that catalyse the mono-, di-, or tri-methylation of specific lysine residues [11] and is involved in many biological processes. Of note, four lysine residues (K4, K9, K27, K36) in the conserved N-terminal tail of the histone are primary targets of specific histone methyltransferases and demethylases. Furthermore, methylation (mono-, di-, and tri-methylation) on trimethylated lysine 27 of histone H3 (H3K27me3) induces transcriptional repression, and thereby is associated with controlling gene expression patterns [12]. By virtue of its influence on chromatin configuration, which could modulate the accessibility of transcription factors and the transcriptional activity of nearby genes, H3K27me3 is found to be involved in the normal development and disease [13], [14], [15], especially in several cancer types, such as breast [16], ovarian [17], and prostate cancer [18]. Notably, for breast cancer, Wei et al. found that the loss of H3K27me3 tended to be associated with a poor prognosis [11]. Another study also confirmed the positive correlation between H3K27me3 and luminal A breast cancer after adjusting for reproductive and lifestyle breast cancer risk factors [19]. These results suggest that H3K27me3 participates in breast cancer carcinogenesis, however, the underlying mechanisms for its involvement remain unknown.

Interestingly, a rapidly growing body of research has revealed that the stemness of both normal stem cells and CSCs is also maintained by the epigenetic marker H3K27me3. For example, Hawkins et al. evaluated the H3K27me3 levels in human embryonic stem (ES) cells with several types of lineage-committed cells [20]. As a result, high H3K27me3 levels in all lineage-committed cells were observed when compared with ES cells. Moreover, a recent study also demonstrated that increased H3K27me3 levels were associated with the differentiation of CSCs in ovarian cancer [21]. As CSCs have much in common with normal stem cells and as the loss of H3K27me3 is closely related with breast cancer and CSCs, we hypothesize that global changes in the H3K27me3 levels might play a key role in stemness maintenance of BCSCs. Hence, drugs targeting the abundance of H3K27me3 might help to eliminate BCSCs.

GSKJ4 is a novel, selective inhibitor of the jumonji family of histone demethylases JMJD3 and UTX, which are the H3K27me2/3-specific demethylases that catalyse the demethylation of H3K27me2/3 [22], [23]. To test our hypothesis, GSKJ4 was employed to evaluate the effects of H3K27me3 demethylation inhibition on the phenotypes and biological functions of BCSCs. The tumourigenicity of the breast cancer cell line after GSKJ4 treatment was also analysed in vivo. The aim of the present study was to evaluate whether the pharmacologic inhibition of H3K27me3 demethylation could specifically target CSCs of breast cancer and to explore the possible molecular mechanisms for this process.

Section snippets

Cell culture

Human breast cancer cell lines MCF7 and MDA-MB-231 were purchased from the American Type Culture Collection (ATCC) and were maintained in medium as indicated by ATCC's instructions. The cell lines were cultured in a humidified atmosphere with 5% carbon dioxide at 37 °C and were subcultured until 90% confluency was reached.

Reagents

Rabbit monoclonal anti-H3K27me3 (CAT: 14034S), anti-UTX (CAT: 33510S), anti-NANOG (CAT: 4903S), anti-SOX2 (CAT: 3579S), anti-OCT4 (CAT: 2750S) and anti-GAPDH (CAT: 5174S) were

A decreased global H3K27me3 level is observed in BCSCs

As a reliable method to enrich CSCs from breast cancer cells in vitro [26], the mammosphere culture platform was successfully established previously [24], [27]. In the present study, an elevated expression of stemness markers including NANOG, SOX2 and OCT4 was found in the mammospheres (Fig. 1A-B). Of note, we found a decreased global H3K27me3 level in the mammosphere-derived BCSCs when compared with adherent breast cancer cells (Fig. 1A). Taken together, these results support the possibility

Discussion

For decades, mounting evidence has suggested that BCSCs are responsible for cancer progression, metastasis and recurrence [30], [31], [32], [33]. Conventional therapies, though effective at killing the majority of cancer cells, often eventually fail due to not eliminating the subpopulation of CSCs, leading in turn to the development and recurrence of cancers [34]. Therefore, it is critical to find ways to eliminate CSCs thoroughly.

To this end, accumulating preclinical studies and clinical

Conclusions

Collectively, our results suggest that H3K27me3 plays a negative role in modulating the behaviours of BCSCs and provide strong supports for the hypothesis that GSKJ4 is a prospective drug target. Mechanistically, this effect results from JMJD3 and UTX inactivation. Thus, our findings suggest that epigenetic modifications, including histone methylation, hold the potential to eliminate BCSCs. The specificity and off-target effects remains to be evaluated in the future.

Conflict of interest

All authors declare no conflict of interest and are in agreement with the publication of this manuscript.

Acknowledgements

This work was supported by Key R&D Project of Jiangsu Province (BE2016677), the Youth Science Foundation of Nanchang Province (20171BAB215045) and Suzhou Science and Technology Project (SS201508).

References (55)

  • A. Pavlopoulou et al.

    Determinants of resistance to chemotherapy and ionizing radiation in breast cancer stem cells

    Cancer Lett.

    (2016)
  • J. Zhu et al.

    Genome-wide chromatin state transitions associated with developmental and environmental cues

    Cell

    (2013)
  • C. Ginestier et al.

    ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome

    Cell Stem Cell

    (2007)
  • A. Wang et al.

    Heterogeneity in cancer stem cells

    Cancer Lett.

    (2015)
  • F. De Santa et al.

    The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing

    Cell

    (2007)
  • M. Kakarala et al.

    Implications of the cancer stem-cell hypothesis for breast cancer prevention and therapy

    J. Clin. Oncol.

    (2008)
  • M. Dean et al.

    Tumour stem cells and drug resistance

    Nat. Rev. Cancer

    (2005)
  • B.J. Morrison et al.

    Breast cancer stem cells: implications for therapy of breast cancer

    Breast Cancer Res.

    (2008)
  • H. Harrison et al.

    Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor

    Cancer Res.

    (2010)
  • S. Liu et al.

    Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells

    Cancer Res

    (2006)
  • T.S. Mikkelsen et al.

    Genome-wide maps of chromatin state in pluripotent and lineage-committed cells

    Nature

    (2007)
  • M. Widschwendter et al.

    Epigenetic stem cell signature in cancer

    Nat. Genet.

    (2007)
  • Y. Huang et al.

    Targeting tumourigenicity of breast cancer stem-like cells using combination epigenetic therapy: something old and something new

    J. Thorac. Dis.

    (2016)
  • Y. Wei et al.

    Loss of trimethylation at lysine 27 of histone H3 is a predictor of poor outcome in breast, ovarian, and pancreatic cancers

    Mol. Carcinog.

    (2008)
  • K.H. Yoo et al.

    Histone Demethylase KDM6A Controls the Mammary Luminal Lineage through Enzyme-Independent Mechanisms

    Mol. Cell Biol.

    (2016)
  • A.H. Wang et al.

    The histone chaperone Spt6 coordinates histone H3K27 demethylation and myogenesis

    EMBO J.

    (2013)
  • W. Jiang et al.

    Histone H3K27me3 demethylases KDM6A and KDM6B modulate definitive endoderm differentiation from human ESCs by regulating WNT signaling pathway

    Cell Res.

    (2013)
  • Cited by (73)

    • Modulation of epigenetic methylation enzymes by synthetic and natural agents

      2023, Transcription and Translation in Health and Disease
    • Targeting histone methylation to reprogram the transcriptional state that drives survival of drug-tolerant myeloid leukemia persisters

      2022, iScience
      Citation Excerpt :

      Also cisplatin-selected melanoma cells showed to have reduced H3K4me3 levels and high KDM5A (Roesch et al., 2010), underlining involvement of epigenetic modifiers in development and maintenance of drug-resistant cell populations. Several KDMs, including KDM5 and KDM6, have been implicated in both therapy resistance (Dalvi et al., 2017; Hinohara et al., 2018; Liau et al., 2017; Pisco et al., 2013; Sharma et al., 2010) and persistence of cancer stem cells within solid tumors (Taube et al., 2017; Yan et al., 2017). KDMs, like methyl transferases, influence histone methylation, affecting gene expression.

    • The inhibitors of KDM4 and KDM6 histone lysine demethylases enhance the anti-growth effects of erlotinib and HS-173 in head and neck cancer cells

      2021, European Journal of Pharmaceutical Sciences
      Citation Excerpt :

      Such pro-apoptotic activity was also observed after inhibition of KDM4 by A1 and B3 in prostate cancer cells (Duan et al., 2015). The inhibition of KDM6 isoforms by GSK-J4 has been shown to lead to anti-cancer effects by reducing cell proliferation and inducing apoptosis in glioma, acute myeloid leukemia, lung cancer, breast cancer, ovarian cancer and thyroid cancer cells (Chu et al., 2020; Li et al., 2018; Lin et al., 2020; Sakaki et al., 2015; Sui et al., 2017; Watarai et al., 2016; Yan et al., 2017). In this study, GSK-J4 did not significantly affect the cell cycle in CAL27 cells, however it increased the population of cells in the S phase in FaDu cells at the higher concentration.

    View all citing articles on Scopus
    1

    Ningning Yan and Liang Xu contributed equally to this work.

    View full text