Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Non-Canonical EZH2 Transcriptionally Activates RelB in Triple Negative Breast Cancer

  • Cortney L. Lawrence,

    Affiliation Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America

  • Albert S. Baldwin

    abaldwin@med.unc.edu

    Affiliation Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America

Abstract

Enhancer of zeste homology 2 (EZH2) is the methyltransferase component of the polycomb repressive complex (PRC2) which represses gene transcription via histone H3 trimethylation at lysine 23 (H3K27me3). EZH2 activity has been linked with oncogenesis where it is thought to block expression of certain tumor suppressors. Relative to a role in cancer, EZH2 functions to promote self-renewal and has been shown to be important for the tumor-initiating cell (TIC) phenotype in breast cancer. Recently a non-canonical role for EZH2 has been identified where it promotes transcriptional activation of certain genes. Here we show that EZH2, through a methyltransferase-independent mechanism, promotes the transcriptional activation of the non-canonical NF-κB subunit RelB to drive self-renewal and the TIC phenotype of triple-negative breast cancer cells.

Introduction

Studies have shown that a subpopulation of cells within a tumor exhibit the ability to self-renew and to initiate tumor formation, while the majority of the cells within the tumor are unable to self-renew and are poorly tumorigenic. These cancer stem cells (CSCs) or tumor initiating cells (TICs) are long-lived and have been shown to be resistant to chemotherapy and to promote tumor reoccurrence and metastasis [1,2]. Importantly, studies have shown that TICs are enriched in high grade tumors and are a predictor of poor prognosis [3]. TICs are observed in multiple subtypes of breast cancer [4,5], including Her2+ cancers and triple negative breast cancer (TNBC), an aggressive subtype of breast cancer characterized by the loss of estrogen receptor (ER), progesterone receptor (PR) and HER-2/neu [6]. Determining signaling pathways critical to TICs offers the potential for new treatments of cancer. In this regard, activities of the NF-κB transcription factor family as well as activities of the histone methyltransferase EZH2 have been shown to be important in self-renewal of triple-negative breast cancer (TNBC).

The NF-κB family consists of 5 proteins, p65 (RelA), RelB, c-Rel, p50/p105 and p52/p100, all sharing a common Rel homology domain [7]. Upon stimulation of cells with cytokines or other ligands, NF-κB becomes activated by one of two pathways. The canonical pathway consists of the activation of the canonical IκB kinase (IKK) complex, causing IKKβ to phosphorylate IκBα resulting in its degradation and the subsequent release of p65-p50 dimers. In the non-canonical pathway activated IKKα phosphorylates p100 resulting in proteolytic processing to p52 and release of p52-RelB dimers [7]. In the nucleus, NF-κB dimers activate the transcription of genes involved in cell cycle regulation, inflammation and suppression of apoptosis. Constitutively activated NF-κB can lead to oncogenic transformation through inflammation, cell proliferation, invasion and inhibition of apoptosis [811]. Furthermore, NF-κB has been shown to be important for the maintenance of breast TICs isolated from different subtypes of breast cancer including HER2+ [12,13] and TNBC [14]. It was shown that both canonical and non-canonical NF-κB promoted the TIC phenotype in triple-negative breast cancer cells [14]. Consistent with this, the NF-κB family of transcription factors has been shown to be activated in HER2+ and TNBC cells [14,15].

EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2) which represses gene transcription through the trimethylation of lysine 27 on histone H3 (H3K27me3) [16]. Upregulation of EZH2 levels has been associated with several cancers [16,17] promoting tumorigenesis partly through the silencing of critical target genes [18]. Additionally, EZH2 has been identified as a transcriptional activator of several genes in various cancers, but the mechanisms are less well characterized as compared to the role of EZH2 within the PCR2 complex. EZH2 was found to activate Notch1 and RAF1-β-catenin signaling in breast cancer to promote self-renewal [19,20]. In prostate cancer cells, the histone methyltransferase (HMT) activity of EZH2 is required in order to interact with androgen receptor and activate transcription, yet this is polycomb independent [21]. EZH2 promotes transcriptional activation of c-myc and cyclinD1 in conjunction with estrogen and WNT signaling and does not require its HMT activity [22]. EZH2 was shown to be important in SWI/SNF mutant cancers requiring both catalytic and non-catalytic functions [23]. Furthermore, EZH2 was reported to interact with the RelA/p65 and RelB subunits of NF-κB in TNBC to activate the transcription of several cytokines, and this was found to be independent of HMT activity [24]. Phosphorylation of EZH2 by CDK2 was shown to promote the oncogenic phenotype in TNBC [25]. Interestingly, TNBC tumors were shown to have elevated levels of EZH2 but surprisingly low levels of H3K27me3 [26] suggesting an HMT-independent role of EZH2 in this tumor subtype. Similarly, in ER- breast cancer cells RelB has been shown to be constitutively active since ER is known to inhibit RelB expression [27,28]. Recently, both EZH2 and RelB have been shown to play a role in the self-renewal of breast TICs [14,19,20] however the interdependence of these two pathways in the maintenance of TNBC TICs has not been explored. Our data identifies an HMT-independent role of EZH2 as a transcriptional activator of RelB in TNBC contributing to the maintenance of triple-negative breast cancer TICs.

Materials and Methods

Cell Culture and Reagents

SUM-149 cells were cultured in HuMEC supplemented with 5% fetal bovine serum (FBS) (Sigma), HuMEC supplement and bovine pituitary extract (Gibco). MDA-MB-231, MCF7 and 293T cells were cultured in Dulbecco’s modified Eagles medium (DMEM) (Gibco) with 10% FBS. HCC-1428 and T47D cells were cultured in RPMI (Gibco) supplemented with 10% FBS. All cell lines were obtained from ATCC. 3-Deazaneplanocin A (DZNep) was obtained from Sigma Aldrich. UNC1999 was a generous gift from the Jian Jin lab.

siRNA Transfection

Small interfering RNAs (siRNA; siGenome SMART pool) against p65 (M-003533-02-0005), RelB (M-004767-02-0005), EZH2 (M-004218-03-0005), SUZ12 (M-006957-00-0005) and Non-Targeting #3 (D-001210-03-20) were purchased from GE Dharmacon. For tumorsphere assays cells were transfected with 50 nM siRNA with Dharmafect 1 reagent according to the manufacturer’s instructions. For all other studies, 25 nM siRNA was used. Samples were collected 72 hours post transfection for analysis.

Tumorsphere Formation Assay

24 hours post siRNA transfection cells were plated for tumorsphere assays. Cells treated with UNC1999 were plated for the tumorsphere assay and treated with UNC1999 or vehicle for the first 3 days in culture. Cells were cultured under previously described conditions [14] and visually counted after a total of 7 days in culture.

Immunoblotting

Cells were lysed in buffer containing 62.5 mmol/L Tris (pH 6.5), 5% glycerol, 2% SDS, 5% 2-mercaptoethanol, 1× protease inhibitor complete mixture (Roche Applied Science) and 1x phosphatase inhibitor cocktail 3 (Sigma-Aldrich). Protein concentrations were determined using Bradford assay against a standard of bovine serum albumin. Lysate protein (25 μg) was resolved by electrophoresis on SDS-polyacrylamide gels (Mini-PROTEAN TGX Precast Gels, Bio-Rad) and transferred to Immobilon-P membranes (Millipore). Membranes were blocked in 5% milk. Primary antibodies were p65 (8242), EZH2 (5246), SUZ12 (3737) and Actin (3700) from Cell Signaling Technology, RelB (sc-226) from Santa Cruz Biotechnology, H3K27me3 (07–449) from Millipore. Membranes were incubated with secondary antibodies conjugated to horseradish peroxidase (Promega) and complexes were detected using the Supersignal West Pico Chemiluminescent Substrate Kit (Pierce).

Quantitative RT-PCR

Total RNA was isolated using TRIzol (Life Technologies). cDNA was synthesized from 1 μg of RNA using the iScript cDNA Synthesis Kit according to manufacturer’s protocol (Bio-Rad). Quantitative PCR was performed with diluted cDNA using Taqman Gene Expression Assay primer-probe sets from Applied Biosystems. Relative expression was normalized to glucuronidase beta using the ΔΔCt method.

Dual-Luciferase Reporter Assay

The RelB promoter containing 898 base pairs of sequence directly upstream of the start site was cloned into the PGL3-Basic vector (Promega) using SacI and XhoI restriction enzymes (New England Biolabs). 293 cells were transfected with 500 ng of the RelB reporter construct, 50 ng pRL-TK Renilla luciferase construct (Promega) and 500 ng of either pFLAG-CMV-p65 or pCMVHA-hEZH2 (Addgene, #24230) using polyethylenimine linear (Sigma-Aldrich). The cells were harvested 24 hours later in passive lysis buffer and analyzed according to Dual-Luciferase Assay System protocol (Promega). Activity was measured using an Lmax Luminometer (Molecular Devices) and relative light units were normalized to pRL-TK Renilla luciferase light units.

Chromatin Immunoprecipitation Assay

The ChIP assay was adapted from [29] 2x106 cells per immunoprecipitation were crosslinked with formaldehyde and the reaction was quenched by the addition of glycine. Cells were washed as previously described, lysed first in 5 mM PIPES pH 8, 85 mM KCl, 0.5% NP40, nuclei were collected and lysed in 50mM Tris pH 8, 10 mM EDTA pH 8.0, 1% SDS, then sonicated to yield DNA fragments less than 1000 base pairs using a Bioruptor from Diagenode. Aliquots corresponding to 2x106 cells were diluted one fold in 2% Triton X-100, 300 mM NaCl, 1% Sodium Deoxycholate and precleared with a slurry of protein G-magnetic beads (Cell Signaling Technology), then incubated with 3 μg p65 (sc-109), RelB (sc-226) from Santa Cruz Biotechnology, EZH2 (39875) from Active Motif, SUZ12 (3737) from Cell Signaling Technology, H3K27me3 (07–449) or IgG (12–371) from Millipore. All solutions contained 1× protease inhibitor complete mixture (Roche Applied Science) and 1x phosphatase inhibitor cocktail 3 (Sigma-Aldrich). Antibody-protein-DNA complexes were captured with a slurry of protein G-magnetic beads. Beads were pelleted, and the supernatant from the IgG immunoprecipitation was used as input DNA. Beads were washed as previously described [29], the protein-DNA complexes were eluted in 2% SDS, 10 mM DTT, 0.1 M NaHCO3 and the cross-links were reversed by the addition of 0.2 M NaCl and incubation at 65° overnight. DNA was RNase and Proteinase K treated, phenol-chloroform extracted, ethanol-precipitated, and dissolved in TE.

Quantitative PCR

Quantitative PCR (QPCR) was performed with 1μL of input DNA or ChIP DNA per reaction with Maxima SYBR Green/Rox (Fermentas). Absolute quantities were calculated using standards of input DNA. Primer sequences are RelB -631 5’-GGGTTACAACAACGCACAA-3’, RelB-529 5’CCTCCAAGGTCTCGCTAC-3’, MYT 5’-GCTGTGGGGAAAGGTAAGTC-3’, MYT 5’-ATGTCTCCTCTGTCAGACGC-3’.

Results

RelB and EZH2 Are Required for the Maintenance of TNBC TICs

Previously our group showed that both canonical and non-canonical NF-κB signaling is required for the maintenance of TNBC TICs [14]. This data in combination with recent reports of EZH2 having a role in TNBC TIC maintenance [19,20] led us to examine the importance of both EZH2 and the non-canonical NF-κB subunit RelB in the self-renewal capacity of TNBC TICs. A hallmark of breast cancer TICs is the ability to form tumorspheres when plated at a low density in media containing no serum on low adhesion plates [30]. Knocking down RelB or EZH2 in both SUM-149 and MDA-MB-231 TNBC cell lines significantly reduced the number of tumorspheres formed (Fig 1A), as expected. Additionally, inhibiting the HMT activity of EZH2 with the small molecule, UNC1999 [31, 32], also reduced tumorsphere formation suggesting an HMT-dependent role of EZH2 in the maintenance of TNBC TICs (Fig 1B). Previous reports demonstrated that the HMT activity of EZH2 is not required in the expansion of stem cells in non-tumorigenic MCF10A cells [19] implicating two distinct roles of EZH2 in stem cell maintenance that potentially differs in tumorigenic versus non-tumorigenic cell lines.

thumbnail
Fig 1. EZH2 and RelB are Required for the Maintenance of TNBC TIC.

(A) Top panel, Quantification of tumorspheres formed by 100 SUM-149 or MDA-MB-231 cells expressing control siRNA or siRNA targeting EZH2 or RelB. Values given as means ± standard deviation from three biological repeats. *P<0.05, **P< 0.005 versus controls; two tailed unpaired t-test. Bottom panel, Immunoblots of indicated proteins in adherent SUM-149 or MDA-MB-231 cells expressing the indicated siRNA construct. β-actin serves as a loading control. (B) Quantification of tumorspheres formed by either 100 SUM-149 or MDA-MB-231 cells treated daily for 3 days with 1μM UNC1999. Values given as means ± standard deviation from three biological repeats. **P< 0.005 versus controls; two tailed unpaired t-test.

https://doi.org/10.1371/journal.pone.0165005.g001

In order to determine if these effects are unique to the triple negative subtype of breast cancer, the contribution of RelB and EZH2 to the self-renewal capacity of several ER+ cell lines was tested. Knockdown of EZH2 only weakly reduced self-renewal of these cells (Fig 2A), while RelB knockdown had a more pronounced response. Additionally, treatment of these ER+ cell lines with EZH2 inhibitor UNC1999 had no effect on tumorsphere formation (Fig 2B), thereby demonstrating that the HMT activity of EZH2 is not required for the maintenance of tumorspheres in some ER+ breast cancer cells.

thumbnail
Fig 2. Self-renewal of TICs in ER+ Breast Cancer Cells is not Dependent on EZH2 and RelB.

(A) Top Panel, Quantification of tumorspheres formed by 100 MCF7, HCC-1428 or T47D cells expressing control siRNA or siRNA targeting EZH2 or RelB. Values given as means ± standard deviation from three biological repeats. *P<0.05 versus controls; two tailed unpaired t-test. Bottom Panel, Immunoblots of indicated proteins in adherent MCF7, HCC-1428 or T47D cells expressing indicted siRNA construct. β-actin serves as a loading control. (B) Left panel, quantification of tumorpsheres formed by 100 MCF7, HCC-1428 or T47D cells treated daily for 3 days with 1μM UNC1999. Values given as means ± standard deviation from three biological repeats. Right panel, immunoblots of the indicated proteins in adherent MCF7, HCC-1428 or T47D cells treated daily for 3 days with 1μM UNC1999 to validate EZH2 inhibition. β-actin serves as a loading control.

https://doi.org/10.1371/journal.pone.0165005.g002

EZH2 Is a Transcriptional Activator of RelB

The role of EZH2 as a transcriptional activator is less well characterized than its role as a transcriptional repressor in the PRC2 complex. It has been previously reported that EZH2 forms a complex with p65 and RelB in TNBC to regulate the transcription of a group of cytokines [3]. To potentially link EZH2 with RelB, we found that, in three different TNBC cell lines, the protein levels of RelB strongly decreases when EZH2 is depleted with either siRNA or shRNA (Fig 3A, and see S1 Fig). RelB levels also decrease after knockdown of p65, which is consistent with the ability of p65 to regulate RelB expression [33]. Importantly, RelB RNA levels decreased with EZH2 knockdown (Fig 3B) indicating transcriptional control of RelB by EZH2. To further address the potential for EZH2 to transcriptionally activate RelB, cells were transfected with p65 or EZH2 and with a reporter containing the RelB promoter. Luciferase activity increased with p65 or EZH2 to a statistically significant level (Fig 3C), suggesting that both p65 and EZH2 can activate the transcription of RelB. Consistent with this, ChIP analysis of the RelB promoter in TNBC cells demonstrated an enrichment of both EZH2 and p65 (Fig 3D) thereby providing further evidence that EZH2 is a transcriptional activator of RelB in TNBC.

thumbnail
Fig 3. EZH2 is a transcriptional activator of RelB.

(A) Immunoblots of indicated proteins in SUM-149, MDA-MB-231 or BT-549 cells expressing control siRNA or siRNA targeting p65, RelB or EZH2. β-actin serves as a loading control. (B) Real time PCR showing expression of RelB in SUM-149, MDA-MB-231 and BT-549 cells. Expression was normalized to glucuronidase beta. Values given as means ± standard deviation from three biological repeats. (C) Dual luciferase assay from HEK293 cells co-expressing the RelB Reporter with either Flag-p65 or HA-EZH2 and Renilla luciferase. Relative light units (RLU) was normalized to Renilla luciferase light units and expressed as fold over the RelB promoter alone. Values given as means ± standard deviation of four biological repeats. *P = 0.05, **P< 0.05 versus controls; two tailed unpaired t-test. (D) ChIP analysis of p65 and EZH2 binding at the RelB promoter in SUM-149 and MDA-MB-231 cells. Non-specific binding was estimated from IgG immunoprecipitates. Values given as means ± standard deviation from three technical repeats of one representative experiment from the three biological repeats.

https://doi.org/10.1371/journal.pone.0165005.g003

The Histone Methyltransferase Activity of EZH2 Is Dispensable for the Regulation of RelB Transcription

It was important to determine if the methyltransferase activity of EZH2 is required for RelB transcription in TNBC. EZH2 was inhibited with small molecule inhibitors UNC1999 or DZNep, which blocked histone methyltransferase activity of EZH2 as detected by a decrease in H3K27me3 levels. Interestingly, this did not lead to a decrease in RelB levels (Fig 4A). To further validate that the regulation of RelB by EZH2 is PCR2-independent, we knocked down SUZ12, the subunit of the PCR2 complex which bridges the interaction of EZH2 to nucleosomes and is required for EZH2 catalytic activity [16]. As expected SUZ12 reduced EZH2 levels. Interestingly, RelB levels were elevated over the RelB levels observed upon knocking down EZH2 suggesting that the EZH2 pool remaining after knocking down SUZ12 is able to contribute to RelB transcription (Fig 4B). The histone methylation mark that EZH2 lays down, H3K27me3, was inhibited upon SUZ12 siRNA further indicating that the HMT activity of EZH2 is not required in regulation of RelB. To further validate that EZH2 is acting independent of the PRC2 complex, enrichment of SUZ12 at the RelB promoter was determined. Although EZH2 was again found to bind at the RelB promoter in TNBC cells, there was no enrichment of SUZ12 at this promoter (Fig 4C). The MYT1 promoter was shown to be regulated by the PRC2 complex [34] and was therefore used as a positive control. Indeed there was significant enrichment of EZH2 and SUZ12 in both TNBC cell lines (Fig 4C) further indicating that EZH2 is binding the RelB promoter and activating its transcription independent of the PRC2 complex.

thumbnail
Fig 4. The HMT activity of EZH2 is not Required for RelB Transcription.

(A) Immunoblots of indicated proteins in SUM-149 or MDA-MB-231 cells treated with the indicated concentrations of UNC1999 (left panel) or DZNep (right panel) for 72 hours. β-actin serves as a loading control. (B) Immunoblots of indicated proteins in SUM-149 or MDA-MB-231 cells expressing control siRNA or siRNA targeting EZH2, SUZ12 or both EZH2 and SUZ12. β-actin serves as a loading control. (C) ChIP analysis of EZH2 and SUZ12 binding at the RelB promoter or the MYT1 Promoter as a positive control in SUM-149 and MDA-MB-231 cells. Non-specific binding was estimated from IgG immunoprecipitates. Values given as means ± standard deviation from three technical repeats of one representative experiment from the three biological repeats.

https://doi.org/10.1371/journal.pone.0165005.g004

Discussion

Our studies demonstrate that the non-canonical NF-κB subunit RelB is transcriptionally regulated by EZH2 in TNBC partly contributing to the maintenance of CSCs. Previously it was shown that RelB [14] and EZH2 [19,20] are important for the self-renewal of breast cancer TICs. Here we demonstrate a mechanism linking these previous observations. EZH2 has been identified as a transcriptional activator in TNBC regulating the expression of Notch [19], as well as IL-6 and TNF [24]. In the latter study, regulation of IL6 and TNF was proposed to involve EZH2 in complex with RelB and p65. We showed that IL6 promotes the formation of TNBC TICs and that exogenous IL6 rescues tumorsphere formation when NF-κB is inhibited [14]. With our finding that EZH2 is a transcriptional activator of RelB it is likely that the upregulation of RelB by EZH2 feeds into the previously described pathway targeting IL-6 (and potentially other cytokines), to drive self-renewal. Since knockdown of p65 was shown to promote TNBC self-renewal and since p65 promotes RelB expression with EZH2, it is possible that a primary role for p65 in self-renewal is through the cooperation with EZH2, to promote RelB expression.

Our studies determined that the histone methyltransferase activity of EZH2 is not required for the regulation of RelB transcription, yet tumorsphere number decreases when treated with inhibitors of EZH2 catalytic activity suggesting a role for the histone methyltransferase activity of EZH2 in tumorsphere maintenance. We speculate that the HMT-dependent activity of EZH2 might involve STAT3 signaling. It has been shown that EZH2 methylates STAT3 in glioblastoma stem-like cells which is required for the activation of STAT3-dependent activation of stem cell-associated transcriptional factors and efficient tumorsphere formation [35]. Additionally, the JAK2/STAT3/IL6 pathway has been shown to be required for the growth of CD44-CD24+ breast cancer cells [36], cell surface markers believed to be associated with breast TICs [30]. This suggests a possible role for EZH2 in promoting breast cancer stem cells through the methylation and activation of STAT3. Interestingly, this pathway could be preferential for the triple negative subtype since UNC1999 treatment does not appear to affect tumorsphere formation in ER+ breast cancer cell lines possibly identifying a pathway specific to the aggressive TNBC subtype which could be therapeutically targeted through inhibition of EZH2 activity.

RelB was originally identified to function primarily in the regulation of adaptive immune response [37] and the involvement of RelB in tumorigenesis was less well characterized. RelB was shown to promote tumorigenicity of prostate cancer cells partly due to regulating IL8 levels [38]. Importantly, in prostate cancer tissue nuclear RelB was observed in more samples than p65 and there was a significant correlation between nuclear RelB and patient Gleason score [39]. Similarly, high nuclear levels of active RelB were found in carcinogen-induced murine mammary tumors [40] and nuclear RelB was found to be significantly elevated in nuclei from ER- inflammatory breast cancer tissue samples [41]. Additionally, inhibition of RelB with vitamin D3 sensitized breast cancers to ionizing radiation [42]. A signature gene set has been identified to be regulated by EZH2 and NF-κB specifically in TNBC [24]. Patients with high expression of this gene set were shown to have lower brain and lung metastasis-free survival emphasizing the importance of this signaling pathway in TNBC. The data reported here provide data linking the non-canonical functions of EZH2 with control of RelB, to drive self-renewal of TNBC cells. This mechanism may underlie key aspects of the oncogenic functions for EZH2 and RelB.

Supporting Information

S1 Fig. RelB Levels Decrease after Knocking Down EZH2 with shRNA.

Immunoblots of the indicated proteins in SUM-149 and MDA-MB-231 cells after transduction with a lentivirus carrying pLKO.1-shScramble or pLKO.1-shEZH2 construct. β-actin serves as a loading control.

https://doi.org/10.1371/journal.pone.0165005.s001

(TIF)

Acknowledgments

We thank members of the Baldwin laboratory for helpful suggestions.

Author Contributions

  1. Conceptualization: AB CL.
  2. Formal analysis: CL.
  3. Funding acquisition: AB.
  4. Investigation: CL.
  5. Methodology: AB CL.
  6. Project administration: AB.
  7. Resources: CL.
  8. Supervision: AB.
  9. Validation: CL.
  10. Visualization: CL.
  11. Writing – original draft: CL.
  12. Writing – review & editing: AB CL.

References

  1. 1. Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell 2014 Mar 6;14(3):275–291. pmid:24607403
  2. 2. Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 2008 May 7;100(9):672–679. pmid:18445819
  3. 3. Lee HE, Kim JH, Kim YJ, Choi SY, Kim SW, Kang E, et al. An increase in cancer stem cell population after primary systemic therapy is a poor prognostic factor in breast cancer. Br J Cancer 2011 May 24;104(11):1730–1738. pmid:21559013
  4. 4. Nakshatri H, Srour EF, Badve S. Breast cancer stem cells and intrinsic subtypes: controversies rage on. Curr Stem Cell Res Ther 2009 Jan;4(1):50–60. pmid:19149630
  5. 5. Luo M, Clouthier SG, Deol Y, Liu S, Nagrath S, Azizi E, et al. Breast cancer stem cells: current advances and clinical implications. Methods Mol Biol 2015;1293:1–49. pmid:26040679
  6. 6. Prat A, Adamo B, Cheang MC, Anders CK, Carey LA, Perou CM. Molecular characterization of basal-like and non-basal-like triple-negative breast cancer. Oncologist 2013;18(2):123–133. pmid:23404817
  7. 7. Hayden MS, Ghosh S. NF-kappaB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev 2012 Feb 1;26(3):203–234. pmid:22302935
  8. 8. Bradford JW, Baldwin AS. IKK/nuclear factor-kappaB and oncogenesis: roles in tumor-initiating cells and in the tumor microenvironment. Adv Cancer Res 2014;121:125–145. pmid:24889530
  9. 9. Staudt LM. Oncogenic activation of NF-kappaB. Cold Spring Harb Perspect Biol 2010 Jun;2(6):a000109. pmid:20516126
  10. 10. DiDonato JA, Mercurio F, Karin M. NF-kappaB and the link between inflammation and cancer. Immunol Rev 2012 Mar;246(1):379–400. pmid:22435567
  11. 11. Perkins ND. The diverse and complex roles of NF-kappaB subunits in cancer. Nat Rev Cancer 2012 Jan 19;12(2):121–132. pmid:22257950
  12. 12. Cao Y, Luo JL, Karin M. IkappaB kinase alpha kinase activity is required for self-renewal of ErbB2/Her2-transformed mammary tumor-initiating cells. Proc Natl Acad Sci U S A 2007 Oct 2;104(40):15852–15857. pmid:17890319
  13. 13. Liu M, Sakamaki T, Casimiro MC, Willmarth NE, Quong AA, Ju X, et al. The canonical NF-kappaB pathway governs mammary tumorigenesis in transgenic mice and tumor stem cell expansion. Cancer Res 2010 Dec 15;70(24):10464–10473. pmid:21159656
  14. 14. Kendellen MF, Bradford JW, Lawrence CL, Clark KS, Baldwin AS. Canonical and non-canonical NF-kappaB signaling promotes breast cancer tumor-initiating cells. Oncogene 2014 Mar 6;33(10):1297–1305. pmid:23474754
  15. 15. Yamaguchi N, Ito T, Azuma S, Ito E, Honma R, Yanagisawa Y, et al. Constitutive activation of nuclear factor-kappaB is preferentially involved in the proliferation of basal-like subtype breast cancer cell lines. Cancer Sci 2009 Sep;100(9):1668–1674. pmid:19538528
  16. 16. Kim KH, Roberts CW. Targeting EZH2 in cancer. Nat Med 2016 Feb 4;22(2):128–134. pmid:26845405
  17. 17. Yamaguchi H, Hung MC. Regulation and Role of EZH2 in Cancer. Cancer Res Treat 2014 Jul;46(3):209–222. pmid:25038756
  18. 18. Min J, Zaslavsky A, Fedele G, McLaughlin SK, Reczek EE, De Raedt T, et al. An oncogene-tumor suppressor cascade drives metastatic prostate cancer by coordinately activating Ras and nuclear factor-kappaB. Nat Med 2010 Mar;16(3):286–294. pmid:20154697
  19. 19. Gonzalez ME, Moore HM, Li X, Toy KA, Huang W, Sabel MS, et al. EZH2 expands breast stem cells through activation of NOTCH1 signaling. Proc Natl Acad Sci U S A 2014 Feb 25;111(8):3098–3103. pmid:24516139
  20. 20. Chang CJ, Yang JY, Xia W, Chen CT, Xie X, Chao CH, et al. EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-beta-catenin signaling. Cancer Cell 2011 Jan 18;19(1):86–100. pmid:21215703
  21. 21. Xu K, Wu ZJ, Groner AC, He HH, Cai C, Lis RT, et al. EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent. Science 2012 Dec 14;338(6113):1465–1469. pmid:23239736
  22. 22. Shi B, Liang J, Yang X, Wang Y, Zhao Y, Wu H, et al. Integration of estrogen and Wnt signaling circuits by the polycomb group protein EZH2 in breast cancer cells. Mol Cell Biol 2007 Jul;27(14):5105–5119. pmid:17502350
  23. 23. Kim KH, Kim W, Howard TP, Vazquez F, Tsherniak A, Wu JN, et al. SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat Med 2015 Dec;21(12):1491–1496. pmid:26552009
  24. 24. Lee ST, Li Z, Wu Z, Aau M, Guan P, Karuturi RK, et al. Context-specific regulation of NF-kappaB target gene expression by EZH2 in breast cancers. Mol Cell 2011 Sep 2;43(5):798–810. pmid:21884980
  25. 25. Yang CC, LaBaff A, Wei Y, Nie L, Xia W, Huo L, et al. Phosphorylation of EZH2 at T416 by CDK2 contributes to the malignancy of triple negative breast cancers. Am J Transl Res 2015 Jun 15;7(6):1009–1020. pmid:26279746
  26. 26. Holm K, Grabau D, Lovgren K, Aradottir S, Gruvberger-Saal S, Howlin J, et al. Global H3K27 trimethylation and EZH2 abundance in breast tumor subtypes. Mol Oncol 2012 Oct;6(5):494–506. pmid:22766277
  27. 27. Wang X, Belguise K, Kersual N, Kirsch KH, Mineva ND, Galtier F, et al. Oestrogen signalling inhibits invasive phenotype by repressing RelB and its target BCL2. Nat Cell Biol 2007 Apr;9(4):470–478. pmid:17369819
  28. 28. Naderi A, Meyer M, Dowhan DH. Cross-regulation between FOXA1 and ErbB2 signaling in estrogen receptor-negative breast cancer. Neoplasia 2012 Apr;14(4):283–296. pmid:22577344
  29. 29. Bronder JL, Moran RG. A defect in the p53 response pathway induced by de novo purine synthesis inhibition. J Biol Chem 2003 Dec 5;278(49):48861–48871. pmid:14517211
  30. 30. Fillmore CM, Kuperwasser C. Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res 2008;10(2):R25. pmid:18366788
  31. 31. Konze KD, Ma A, Li F, Barsyte-Lovejoy D, Parton T, Macnevin CJ, et al. An orally bioavailable chemical probe of the Lysine Methyltransferases EZH2 and EZH1. ACS Chem Biol 2013;8(6):1324–1334. pmid:23614352
  32. 32. Xu B, On DM, Ma A, Parton T, Konze KD, Pattenden SG, et al. Selective inhibition of EZH2 and EZH1 enzymatic activity by a small molecule suppresses MLL-rearranged leukemia. Blood 2015 Jan 8;125(2):346–357. pmid:25395428
  33. 33. Bren GD, Solan NJ, Miyoshi H, Pennington KN, Pobst LJ, Paya CV. Transcription of the RelB gene is regulated by NF-kappaB. Oncogene 2001 Nov 22;20(53):7722–7733. pmid:11753650
  34. 34. Kirmizis A, Bartley SM, Kuzmichev A, Margueron R, Reinberg D, Green R, et al. Silencing of human polycomb target genes is associated with methylation of histone H3 Lys 27. Genes Dev 2004 Jul 1;18(13):1592–1605. pmid:15231737
  35. 35. Kim E, Kim M, Woo DH, Shin Y, Shin J, Chang N, et al. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell 2013 Jun 10;23(6):839–852. pmid:23684459
  36. 36. Marotta LL, Almendro V, Marusyk A, Shipitsin M, Schemme J, Walker SR, et al. The JAK2/STAT3 signaling pathway is required for growth of CD44(+)CD24(-) stem cell-like breast cancer cells in human tumors. J Clin Invest 2011 Jul;121(7):2723–2735. pmid:21633165
  37. 37. Burkly L, Hession C, Ogata L, Reilly C, Marconi LA, Olson D, et al. Expression of relB is required for the development of thymic medulla and dendritic cells. Nature 1995 Feb 9;373(6514):531–536. pmid:7845467
  38. 38. Xu Y, Josson S, Fang F, Oberley TD, St Clair DK, Wan XS, et al. RelB enhances prostate cancer growth: implications for the role of the nuclear factor-kappaB alternative pathway in tumorigenicity. Cancer Res 2009 Apr 15;69(8):3267–3271. pmid:19351823
  39. 39. Lessard L, Begin LR, Gleave ME, Mes-Masson AM, Saad F. Nuclear localisation of nuclear factor-kappaB transcription factors in prostate cancer: an immunohistochemical study. Br J Cancer 2005 Oct 31;93(9):1019–1023. pmid:16205698
  40. 40. Demicco EG, Kavanagh KT, Romieu-Mourez R, Wang X, Shin SR, Landesman-Bollag E, et al. RelB/p52 NF-kappaB complexes rescue an early delay in mammary gland development in transgenic mice with targeted superrepressor IkappaB-alpha expression and promote carcinogenesis of the mammary gland. Mol Cell Biol 2005 Nov;25(22):10136–10147. pmid:16260626
  41. 41. Van Laere SJ, Van der Auwera I, Van den Eynden GG, Elst HJ, Weyler J, Harris AL, et al. Nuclear factor-kappaB signature of inflammatory breast cancer by cDNA microarray validated by quantitative real-time reverse transcription-PCR, immunohistochemistry, and nuclear factor-kappaB DNA-binding. Clin Cancer Res 2006 Jun 1;12(11 Pt 1):3249–3256.
  42. 42. Mineva ND, Wang X, Yang S, Ying H, Xiao ZX, Holick MF, et al. Inhibition of RelB by 1,25-dihydroxyvitamin D3 promotes sensitivity of breast cancer cells to radiation. J Cell Physiol 2009 Sep;220(3):593–599. pmid:19373868