Abstract
Enhancer of Zeste Homologue 2 (EZH2) is the catalytic subunit of the polycomb repressive complex 2 (PRC2) that is critical for determining cell identity. An epigenetic writer, EZH2 has a well-defined role in transcriptional repression by depositing trimethyl marks on lysine 27 of histone H3. However, there is mounting evidence that histone methyltransferases like EZH2 exert histone methyltransferase–independent functions. The relevance of these functions to breast cancer progression and their regulatory mechanisms are only beginning to become understood. Here, we review the current understanding of EZH2 H3K27me3-independent, noncanonical, functions and their regulation in breast cancer.
Polycomb Structure and Function
Epigenetic modifications, such as post-translational modifications that occur on histone tails, correlate with whether chromatin conforms to an open state (euchromatin) associated with active gene transcription, or a closed state that prevents gene transcription (heterochromatin). These modifications are broadly associated with three classes of proteins: those that place the modifications, those that read the modifications, and those that remove the modifications.1
Polycomb group (PcG) and trithorax group (TrxG) are two of the most well-studied protein complexes responsible for post-translational modifications on histone tails, and consequently, epigenetic regulation. Genome-wide chromatin immunoprecipitation studies have revealed that PRC2 represses transcription of hundreds of genes to maintain pluripotency of embryonic stem cells.2 The canonical catalytic subunit of PRC2 is EZH2, a 746–amino acid protein containing a C-terminal SET domain common to many lysine methyltransferases3 catalyzes the exchange of one to three methyl groups from donor S-adenosylmethionine (SAM) onto H3K27. The H3K27me3 form of H3K27 is associated with transcriptional repressive functions and, in most contexts, is associated with genome-wide distribution of PRC2. Additional domains of EZH2 include its WD-40 binding domain (WDB) responsible for interaction with EED, and domains 1 and 2 thought to mediate protein–protein interactions.4 Two SANT (Swi3, Ada2, N-CoR, TFIIIB) domains are less well characterized but may be important for interaction with histone tails.5 Interestingly, structural studies have found that SANT1 is a histone binding domain with specificity for the histone H4 N-terminal tail6 and sits adjected to a stimulation response motif (SRM) that stabilizes SANT1. Additionally, a cysteine-rich domain (CXC) sits immediately N-terminal to the SET domain, which is important for methyltransferase activity.5 More recently, a transactivating domain (TAD) was observed comprising the SANT1 and SRM (stimulation response motif) regions.7
In addition to the well-described transcriptional and post-transcriptional mechanisms involving c-MYC,8 miRNAs, and other regulators,9,10 the function of EZH2 is regulated at a post-translational level in normal and neoplastic cells (Table 1).4,11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41
Table 1.
Modification | Site | Modifier | Function | Reference |
---|---|---|---|---|
Phosphorylation | S21 | AKT | Suppresses EZH2 HMT activity by impeding affinity of EZH2 for nucleosome substrate Promotes the noncanonical methylation of STAT3 and STAT3 activity in GBM cells Appears to be required for androgen-independent growth of castration-resistant prostate cancer cells Appears to be required for the transcriptional activation activity of EZH2 in CRPC |
4,11,12 |
Phosphorylation | Y244 | JAK3 | Promotes dissociation of PRC2 members; Increases noncanonicalEZH2 interaction with RNA PolII, promotes proliferation of NK/T-cell lymphoma cells | 13 |
Phosphorylation | T261 | CDK5-related protein | Promotes degradation by F-box and WD repeat domain-containing 7 | 14 |
Phosphorylation | T311 | AMP kinase | Disrupts interaction between EZH2 and SUZ12, resulting in reduced canonical H3K27 trimethylation Associated with better survival in breast and ovarian cancer patients |
15 |
Phosphorylation | T345 | CDK1, CDK2, PKCg | Promotes degradation; cell-cycle dependent, increases binding to HOTAIR ncRNA and 5′ end of Xist Promotes recruitment of EZH2 to target gene loci and maintenance of H3K27me3 levels at these target loci in prostate cells |
16, 17, 18 |
Phosphorylation | S363 | GSK3β | Not yet known | 19 |
Phosphorylation | T367 | p38α GSK3β |
Promotes satellite cell differentiation in response to TNFα through PJA1-mediated degradation of EZH2 Induces cytoplasmic localization of EZH2 and binding to vinculin and other cytoskeletal regulators of cell migration and invasion; critical for TNBC metastasis Contributes to histologic diversity and behavior of metaplastic breast tumors Reduces H3K27me3 activity and reduces migratory/invasive properties in MCF12A and MDA-MB-231 overexpressing cells |
19, 20, 21, 22 |
Phosphorylation | T416 | CDK2 | Enhances migration, invasion, stemness in TNBC cells Serves as a docking site for the forkhead-associated domain of NIPP1, which prevents dephosphorylation and is required for EZH2 association with proliferation loci |
23,24 |
Phosphorylation | T487 | CDK1 PKCg |
Promotes ubiquitination and degradation by the proteasome; negatively regulates proliferation in PC3 prostate cancer cells Disrupts binding with SUZ12 and EED, thereby suppressing H3K27 methyltransferase activity Promotes differentiation of mesenchymal stem cells |
25, 26, 27 |
Phosphorylation | Y641 | JAK2 | Promotes B-TrCP (FBXW1)-mediated degradation | 28 |
Phosphorylation | S652 | ATM | May negatively regulate interaction with PRC2 members SUZ12 and EED and negatively regulate stability | 29 |
Phosphorylation | S690 | PKCg | Not yet known | 27 |
Phosphorylation | S734 | ATM | May negatively regulate interaction with PRC2 members SUZ12 and EED and negatively regulate stability | 29 |
O-GlcNacylation | S75 | OGT | Likely positively regulates EZH2 protein stability | 30 |
Acetylation | K348 | PCAF and SIRT1 (deacetylation) | Decreases phosphorylation at T345 and T487, increases EZH2 stability, may affect H3K27me3-mediated repression of EZH2 target genes May enhance transcriptional silencing of HOXA10 in lung carcinoma cells; may enhance lung cancer cell migration and invasion |
31 |
Methylation | K307 | SMYD2 | Dimethylation at K307 critical for EZH2 protein stability, protecting from ubiquitination and degradation and thereby contributing to breast cancer cell invasion | 32 |
Methylation | R432 | PRMT1 | Asymmetric demethylation which inhibits CDK-1 mediated phosphorylation of EZH2 at T345 and T487, thereby preventing ubiquitination by TRAF6 Contributes to breast cancer cell invasion and metastasis |
33 |
Methylation | K510 | PRC2 (auto) | Increases activity, promotes accessibility to H3 tail | 34,35 |
Methylation | K514 | PRC2 (auto) | Increases activity, promotes accessibility to H3 tail Significantly reduced in diffuse intrinsic pontine glioma cells carrying H3K27M mutation |
34,35 |
Methylation | K515 | PRC2 (auto) | Not known | 34,35 |
Ubiquitination | K421 | SMURF2 | Proteasomal degradation | 36,37 |
Ubiquitination | Praja1, FBXW, FBXW7, TRAF6 | Protein instability and degradation | 14,20,33,38, 39, 40 | |
PARylation | D233, E239 | PARP1 | Loss of PARylation results in PRC2 dissociation, EZH2 down-regulation, and decreased H3K27me3 | 41 |
Shown are the demonstrated post-translational modifications of EZH2 occurring across multiple systems.
GBM, glioblastoma multiforme.
EZH2 Function in Cancer
Deregulation of epigenetic proteins is commonly seen in cancer. Alterations in EZH2 have a complex role, exerting both tumor-suppressive and tumor-promoting functions depending on the context. Deregulation of EZH2 typically results in its overexpression, which often correlates to poor prognosis. Indeed, overexpression of EZH2 has been observed in multiple tumors, including prostate,42, 43, 44 breast,44, 45, 46, 47, 48, 49 and others. Beyond its role as a biomarker, EZH2 has a well-established role as an oncogene; ectopic expression of EZH2 in nontumorigenic cells promotes their proliferation,42,45 and genetic knockdown of EZH2 reduces the proliferation, migration, invasion, and stem properties in multiple cancer cell lines and xenograft models.23,48, 49, 50, 51
How overexpression of EZH2 promotes cancer in individual tumor types is not well understood. Several putative tumor suppressors repressed by EZH2 have been described in prostate cancer and include repression of CDH1,43 RUNX3,52 and others (reviewed by Kim and Roberts53). However, for other tumor types, fewer targets have been identified, and their regulation by EZH2 may be context-specific, as in the case of tobacco-mediated induction of PRC2 repressing the tumor suppressor DKK1 in lung cancer.54
Activating somatic heterozygous mutations of EZH2 have been observed in up to 30% of non-Hodgkin lymphomas of follicular and germinal center diffuse large B-cell (GCB-DLBCL) subtypes.55,56 These predominantly occur within the SET domain (hotspot mutations at Y641, A677G, and A687) and alter EZH2 substrate specificity toward di- and trimethylated H3K27 over the monomethylated state.57 The resulting altered specificity, such as in the case of Y641, affects both a global increase in H3K27me3 and a total redistribution of the repressive mark at PRC2-regulated loci that are sufficient for tumorigenesis58 and is a rationale for clinical use of EZH2 inhibitors. Indeed, to date, three SAM-competitive EZH2 methyltransferase inhibitors (tazemetostat/EPZ-6348, GSK2816126, and CPI-1205) have reached phase I/phase II clinical trials for patients with follicular lymphomas, DLBCL harboring these gain-of-function mutations.59 These inhibitors are also being examined in a genetically defined set of solid tumors harboring inactivating mutations in members of the ATP-dependent chromatin remodeling SWI/SNF complex that results in oncogenic dependency on EZH2.60,61 A fourth compound, MAK683, is the only EZH2 inhibitor currently in clinical trials that does not target the methyltransferase activity directly, but rather disrupts EZH2–EED interaction, the proof of principle for which is described in Kim et al.62
In breast cancer, EZH2 was first observed to be overexpressed in approximately 55% of invasive breast carcinomas. Its expression is inversely correlated with poor breast cancer survival,45 suggesting its role as a potential biomarker. Multiple studies have corroborated this finding.44,47,63, 64, 65, 66 High expression of EZH2 is significantly associated with estrogen receptor- (ER) and progesterone receptor- (PR) negative status, higher histologic grade, and triple-negative breast cancer (TNBC) status45,64 of breast tumors. Additionally, EZH2 may have a role that precedes frank tumor dissemination, because higher levels of EZH2 expression in normal breast epithelium correlate with increased risk of breast cancer.67 Together, the data have demonstrated a role of EZH2 as a potential biomarker of breast cancer development and aggressive breast cancers.
Gain- and loss-of-function studies have demonstrated that EZH2 exerts oncogenic functions in breast cells. Overexpression of EZH2 in benign mammary epithelial cells is sufficient to increase anchorage-independent growth and invasion, and this phenotype is dependent on an intact SET domain.45 Conversely, genetic inhibition of EZH2 using shRNA is sufficient to reduce the proliferation of ER-negative breast cancer cells in vitro and in vivo by delaying G2/M cell cycle transition.50 Independent of proliferation, genetic inhibition with shRNA or pharmacologic depletion of EZH2 with 3-deazaneplanocin A (DZNep) is sufficient to reduce migration, invasion, and random motility of TNBC cells and is accompanied by the acquisition of a MET phenotype and restoration of E-cadherin.48 EZH2 may also contribute to PARP inhibitor resistance, and pharmacologic inhibition of EZH2 with a specific inhibitor can restore PARP inhibitor sensitivity.41 EZH2 is capable of conferring tamoxifen resistance to ER-positive breast cancer cells by repressing the estrogen receptor alpha cofactor GREB1, which causes a redistribution of other ERα cofactors p300 and CBP. This may explain why tamoxifen can have a paradoxical growth-stimulating effect in tamoxifen-resistant ER-positive breast cancer cell lines.66 These studies suggest that in breast cancer alone, EZH2 has a diverse set of oncogenic functions.
The role of EZH2 in breast cancer initiation is starting to be unraveled. Overexpression of EZH2 in the breast acinar epithelium is associated with an increased risk of invasive carcinoma.67,68 Transgenic mouse models of conditional EZH2 overexpression in mammary epithelial cells (MMTV LTR-driven EZH2) show disrupted terminal end bud development, a hyperbranching phenotype, and precocious intraductal epithelial hyperplasia, a precursor to carcinoma in situ.69 Data suggest that EZH2 overexpression accelerates breast cancer initiation in MMTV-neu mice and increases the numbers of tumor initiating cells in this model through transcriptional activation of Notch1 via EZH2 interaction with RelA and RelB NF-κB components.51 Further investigations are necessary to define the role of EZH2 in breast cancer initiation.
Several putative mechanisms have been posited to explain how overexpression of EZH2 confers proliferative, migratory, and invasive potential to breast cancer cells. These include transcriptional repression of tumor suppressors such as RUNX3,52 RAD51-paralog proteins,70 and RKIP.71 EZH2 has been shown to promote epithelial–mesenchymal transition by promoting signaling through protumorigenic kinases, such as by activating the p38 MAPK.48 Several recent studies show that a subset of TNBCs display a decoupling of EZH2 and H3K27me3. TNBCs display higher levels of EZH2 and lower levels of H3K27me3, as indicated by immunohistochemistry.72 Stratification of patients by EZH2 and H3K27me3 shows that the combination of high EZH2/low H3K27me3 portends the worst overall survival in breast cancer patients, irrespective of ER status.72 These observations suggest the existence of H3K27me3-independent functions of EZH2, such as transactivating functions and nonhistone protein methylation, which are the focus of intense investigation and are summarized below.
Noncanonical Functions of EZH2 in Breast Cancer
The reported noncanonical mechanisms of EZH2 in breast tumorigenesis are summarized in Figure 1. Independent of its H3K27me3 activity, EZH2 can transcriptionally activate genes. Underscoring the importance of cellular context are the different roles of EZH2 in studies of ER-positive versus ER-negative breast cancer. In ER-negative basal-like breast cancers, EZH2 activates transcription of NF-κB targets independent of other PRC2 members.73 In this setting, EZH2 forms a complex with RelA and RelB on NF-κB target genes and is associated with activating H3K4 marks rather than H3K27me3 marks.73 Interestingly, EZH2 may itself transcriptionally activate RelB in TNBC, adding an additional layer of complexity.74 By contrast, in ER-positive breast cancer cell lines, EZH2 interacts with ERα and β-catenin upon E2 treatment. In response to estradiol treatment, ERα, β-catenin, and EZH2 form a complex on the MYC (alias c-myc) promoter and activate its transcription, independent of the histone methyltransferase (HMT) activity of EZH2.75 Furthermore, EZH2 interacts with the transcription factor TRIM28 and positively regulates transcription of approximately 100 genes in ER-positive breast cancer cell lines. Through chromatin immunoprecipitation quantitative PCR, EZH2 and TRIM28 were found to co-occupy regions within 10 kb of the transcription start site.76
EZH2 transcriptionally activates Notch1 in TNBC to increase the number of tumor-initiating cells in cell lines and in animal models.51 In this instance, EZH2 binds to the proximal Notch1 promoter with cofactors RelA and RelB to activate transcription independent of H3K27me3, and associated with H3K4 activating marks. Studies involving generation of myc-tagged EZH2 deletion mutants involving the amino-terminal homology domains I and II (ΔHI and ΔHII), the carboxyl-terminal SET domain (ΔSET), and the nuclear localization signal (ΔNLS), as well the EZH2-H689A mutant, which has reduced HMT activity, in MCF10A cells,51 indicate that EZH2 HMT activity is dispensable for Notch1 transcriptional activation and stem cell expansion in nontumorigenic breast cells. The translational relevance of these findings was validated in human tissue samples where high levels of EZH2 are significantly associated with activated NOTCH1 protein and increased tumor-initiating cells in triple-negative invasive carcinomas.51
A role for EZH2 in transcriptional activation was reported in other systems, including intestinal stem cells,77 and in castration-resistant prostate cancer. In the latter, chromatin immunoprecipitation sequencing of EZH2 and H3K27me3 has shown sites where EZH2 does not colocalize with the repressive mark. These solo peaks are instead associated with H3K4me2 and -3 and RNA pol II, and EZH2 knockdown decreases levels of active mark at these sites. These data are interesting because they not only support a transactivating role of EZH2, but also suggest that EZH2 might function as both a transcriptional repressor and activator within the same type of cell.4 Taken together, these studies reveal the complexity of EZH2 functions in tumorigenesis where the protein is capable of exerting repressive and activating functions depending on cellular context.
How EZH2 switches from a transcriptional repression role to a transcriptional activation role is not fully understood. A disordered transactivation domain (TAD) contains the SRM and SANT1 regions and exists in a structurally locked state in an alpha-helix bundle structure bound by the SBD of EZH2. Phosphorylation at Y244 by JAK3 or at S21 by AKT reduces SBD binding, thereby allowing the transactivation domain to interact with transcriptional coactivators such as the histone acetyltransferase p300.7
In recent years, there has been growing evidence that EZH2 may undergo phosphorylation, which plays an important role in regulating EZH2 noncanonical functions. EZH2 phosphorylation events have been linked to decreased EZH2 transcriptional repressor function through reduction of H3K27me3.
One of the first indications that phosphorylation modifies EZH2 functions in cancer progression was published in 2005, when Cha et al11 characterized the phosphorylation of S21 by Akt in MDA-MB-453 breast cancer cells. Modification of this site results in decreased affinity of EZH2 for H3, and therefore less methyltransferase activity compared with the unphosphorylated enzyme.78 The mechanism appears to rely on reduced affinity of EZH2 to chromatin but does not affect the composition of the PRC2. Interestingly, several novel functions of EZH2 in malignancies other than breast have been demonstrated as a result of S21 phosphorylation: in multiple myeloma, where S21 phosphorylation is implicated in cell-adhesion–mediated drug resistance78; in glioblastoma, where S21 phosphorylation promotes cancer progression via nonhistone methylation of STAT312; and in prostate cancer, where S21 phosphorylation switches EZH2 to a transcriptional coactivator of AR.4 However, these same mechanisms have not been definitively shown in breast cancers.
There are two phosphorylation sites that do not affect EZH2 methyltransferase activity directly, but instead lead to decreased activity due to the poor binding of phosphorylated forms to the scaffolding proteins of the polycomb repressive complex. Wan et al15 showed that AMPK, which is activated in conditions of energy stress, phosphorylates EZH2 at T311, leading to dissociation from the PRC2 and less H3K27 methylation at EZH2 target sites in breast cancer cells. Increased active AMPK and pT311-EZH2 are both found in breast and ovarian cancers, but only pT311-EZH2 correlates with higher survival.
Cyclin-dependent kinase 1 and 2 (CDK1 and CDK2) phosphorylation of EZH2 plays an important role in regulating EZH2 in breast cancer, with CDK1 promoting noncanonical functions. CDK1 phosphorylation of EZH2 at T487, which lies within the SUZ12 binding region of EZH2, disrupts the interaction between EZH2 and SUZ12/EED, and regulates EZH2 protein stability.25 Functionally, CDK1-dependent phosphorylation of EZH2 at T487 suppresses methylation of H3K27 and promotes osteogenic differentiation of human mesenchymal stem cells.83 In breast cancer MCF7 cells, loss of phosphorylation at T487 does not affect cell proliferation but significantly increases cancer cell migration and invasion, suggesting that phosphorylation at this site is associated with EZH2 metastatic functions.17,26 In line with these findings, the methyltransferase PRMT1 promotes metastasis of MDA-MB-231 and MCF7 breast cancer cells by methylating EZH2 at R342, which prevents phosphorylation of nearby T487. Interestingly, phosphoproteomic analyses have demonstrated T487 phosphorylation is significantly elevated in basal-like breast cancers compared with other subtypes,79 suggesting a context-specific role for this event.
p38 Mitogen activated kinase phosphorylates EZH2 at T367. First described in myoblasts and satellite cells, it was recently shown to play an important role in TNBC progression. In muscle cells, activation of the p38 signaling pathway via tumor necrosis factor α (TNFα), as in case of muscle injury, results in p38α-mediated phosphorylation of EZH2 at threonine 372 (which corresponds to T367; the five amino acid difference resulting from using different EZH2 isoforms as reference), and recruitment of EZH2 to the Pax7 promoter.21 In skeletal muscle progenitors, MyoD induces an E3 ubiquitin ligase Praja1 to ubiquitinate and subsequently degrade EZH2 in response to p38-mediated phosphorylation at T372.20,21
In breast cancer, p38 phosphorylation of EZH2 at T367 has a novel function. EZH2 and p38 are concordantly expressed in invasive breast carcinomas, and interact in breast cancer cells.48 Recent studies further characterized this interaction by showing that p38 phosphorylates EZH2 at T367 and developed and validated a phosphorylation-specific antibody for this site.80 Interestingly, in normal lobules, if present, pEZH2-T367 is exclusively nuclear, but in invasive breast cancer cells is expressed in the cytoplasm as well as in the nucleus. There is a significant increase in cytoplasmic pEZH2-T367 with cancer progression, and high cytoplasmic pEZH2-T367 in invasive carcinomas are significantly associated with higher histologic grade, ER-, PR-, and triple-negative status. Mechanistically, phosphorylation results in reduced global H3K27me3 levels by controlling EZH2 subcellular localization and promoting movement to the cytoplasm. In the cytoplasm, EZH2 complexes with cytoskeletal proteins such as vinculin to promote breast cancer cell adhesion, migration, and invasion. pEZH2-T367 retains the ability to bind to PRC2 members EED and SUZ12 in TNBC cells, suggesting the intriguing hypothesis that phosphorylation of EZH2 at this specific site may promote methylation of cytoplasmic proteins, which warrants further investigation.80 Supporting this hypothesis are preliminary studies suggesting that EZH2 catalyzes p38α methylation, as well as data showing that EZH2 methylates nonhistone proteins in normal and in neoplastic cells (Table 2).12,48,81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91
Table 2.
Substrate | Site | Peptide | Function and notes | Reference |
---|---|---|---|---|
β-Catenin | K49me3 | LSGGNP | Promotes repression of Sox1 and Sox3 in ESCs, competes with acetylation at the same site | 81 |
EloA | K754me | TVKIAP | Modulates expression of low-expression PRC2 target genes in mouse ESCs | 82 |
Fra-2 | K104me, K104me2 | GVITIG | May prevent Fra-2 from transcriptionally regulating epidermal differentiation genes | 83 |
GATA4 | K299me | LYMLHG | Reduces its transcriptional activity by preventing p300 acetylation in HL-1 cardiac muscle cells | 84 |
Histone H2B | K120me | AVTYTS | May compete with H2BK120-ub in vitro and in cancer cell lines | 85 |
Jarid2 | K116me3, K116me2 | AQRFAQ | Mimics trimethylated histone tail and allosterically activates PRC2 in ESCs | 86,87 |
p38α | K139me3, K165me3 | RGLYIH CELILD |
May regulate p38 activity | 48 and unpublished data |
PLZF | K430me | SGMTYG | Based on predictive software∗ | 88 |
RORα | K38me | SARSEP | Promotes ubiquitination and degradation in HEK293 cells | 89 |
Stat3 | K49me2 | AASESH | Promotes transcription of STAT3 target genes in response to IL-6 in colon cancer cells | 90 |
Stat3 | K180me3 | KTLSQG | Promotes activation of STAT3 signaling in glioblastoma stem-like cells | 12 |
Talin | K2454me3 | EAMRLQ | Disrupts binding of Talin to F-actin in neutrophils and dendritic cells | 91 |
Shown are the putative nonhistone targets of EZH2, along with the sequences of the three amino acids.
ESC, embryonic stem cell.
Prediction of Methylation Sites Based on Enhanced Feature Encoding Scheme (PMeS).
In vivo, using knockdown-rescue orthotopic xenograft mouse models of breast cancer, no difference was noted in primary tumor growth between tumors whose EZH2 had been replaced with EZH2 or with a phospho-deficient T367A mutant. However, luciferase analysis demonstrated that loss of T367 phosphorylation of EZH2 resulted in significantly less metastatic burden, supporting a role for T367 in mediating the migratory and invasive properties of EZH2 in breast cancer. Interestingly, the existence of cytoplasmic EZH2 has been described previously.47 In metaplastic breast carcinomas, the most aggressive form of TNBC, there is differential expression and subcellular localization of pEZH2-T367 among different histologic subtypes, suggesting that pEZH2-T367 may play a role in the histologic diversity of these tumors.22
Translational Implications of Noncanonical EZH2 Functions in Breast Cancer
There are still limited data on how inhibition of EZH2 functions affects tumorigenesis in preclinical models of breast cancer. Inhibition of canonical EZH2 function, assessed by global levels of H3K27me3, using the SAM-competitive small molecule GSK126 at low micromolar concentrations appears to have no effect on cell proliferation of three TNBC cell lines, despite derepression of at least two canonical EZH2 targets, CDKN1A and CDKN1C, and may even promote invasion of breast cancer cells through derepression of MMPs.92 These data have been replicated recently in the MMTV-PyVmT model of breast cancer, in which mice develop tumors resembling luminal B breast cancer subtype. Interestingly, treatment of mice harboring luminal B tumors with the EZH2 inhibitor GSK126 has minimal effects on primary tumor size but significantly reduces lung metastases.93 Recently, Yomtubian et al94 found similar results using orthotopic LM2 as well as human TNBC patient-derived xenograft mouse models of breast cancer; GSK-126 treatment had no effect on primary tumor size, but significantly reduced breast cancer lung metastases, which was attributed to an effect on a small subpopulation of metastatic cells.
The oncogenic alterations in pathways directly involving EZH2 are frequent across multiple types of cancer, which make targeting EZH2 a tantalizing option for cancer treatment. However, the body of work of EZH2 function in breast cancer supports that it may have multiple, discrete oncogenic functions that are not fully explained by its histone methyltransferase activity and that have yet to be fully delineated. This consideration is important, because several potent SAM-competitive EZH2 inhibitors have been developed over the past 5 years and have reached phase I to III clinical trials. The majority of these are with the inhibitor tazemetostat, which was approved by the Food and Drug Administration in 2020 for metastatic or locally advanced epithelioid sarcoma. However, given the increasing evidence of noncanonical functions of EZH2 in promoting cancer, the use of SAM-competitive inhibitors may not be as effective as other small molecules that disrupt EZH2 protein–protein interactions. These include MAK683, which binds to EED and disrupts the EZH2 interaction with histone tail, and is being actively investigated in clinical trials (reviewed by Eich et al95). More recently, proteolysis-targeting chimeras (PROTACs), as well as EZH2 degraders, have offered novel mechanisms of degrading EZH2 and are exciting new modalities added to the armament of PRC2 inhibition.96, 97, 98 These data highlight the need for a greater understanding of the noncanonical functions of EZH2 in order to develop effective management options for breast cancers.
Footnotes
Supported by NIH grants R01CA125577 (C.G.K.), R01CA107469 (C.G.K.), and F30CA19084 (T.A.); and Department of Defense grant W81XWH-19-1-1-0093 (C.G.K.).
Disclosures: None declared.
C.G.K. is the 2020 recipient of the American Society for Investigative Pathology Outstanding Investigator Award, which is presented annually to a midcareer investigator with demonstrated excellence in research in experimental pathology. Portions of this work were presented at the 2020 Pathobiology for Investigators, Students, and Academicians scientific meeting, held virtually from November 9 to 13, 2020, where C.G.K. delivered the Outstanding Investigator Award lecture.
References
- 1.Arrowsmith C.H., Bountra C., Fish P.V., Lee K., Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11:384–400. doi: 10.1038/nrd3674. [DOI] [PubMed] [Google Scholar]
- 2.Lee T.I., Jenner R.G., Boyer L.A., Guenther M.G., Levine S.S., Kumar R.M., Chevalier B., Johnstone S.E., Cole M.F., Isono K.i., Koseki H., Fuchikami T., Abe K., Murray H.L., Zucker J.P., Yuan B., Bell G.W., Herbolsheimer E., Hannett N.M., Sun K., Odom D.T., Otte A.P., Volkert T.L., Bartel D.P., Melton D.A., Gifford D.K., Jaenisch R., Young R.A. Control of developmental regulators by polycomb in human embryonic stem cells. Cell. 2006;125:301–313. doi: 10.1016/j.cell.2006.02.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rea S., Eisenhaber F., O'Carroll D., Strahl B.D., Sun Z.-W., Schmid M., Opravil S., Mechtler K., Ponting C.P., Allis C.D., Jenuwein T. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature. 2000;406:593–599. doi: 10.1038/35020506. [DOI] [PubMed] [Google Scholar]
- 4.Xu K., Wu Z.J., Groner A.C., He H.H., Cai C., Lis R.T., Wu X., Stack E.C., Loda M., Liu T., Xu H., Cato L., Thornton J.E., Gregory R.I., Morrissey C., Vessella R.L., Montironi R., Magi-Galluzzi C., Kantoff P.W., Balk S.P., Liu X.S., Brown M. EZH2 oncogenic activity in castration-resistant prostate cancer cells is polycomb-independent. Science. 2012;338:1465–1469. doi: 10.1126/science.1227604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tan J.z., Yan Y., Wang X.x., Jiang Y., Xu H.E. EZH2: biology, disease, and structure-based drug discovery. Acta Pharmacol Sin. 2014;35:161–174. doi: 10.1038/aps.2013.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Weaver T.M., Liu J., Connelly K.E., Coble C., Varzavand K., Dykhuizen E.C., Musselman C.A. The EZH2 SANT1 domain is a histone reader providing sensitivity to the modification state of the H4 tail. Sci Rep. 2019;9:987. doi: 10.1038/s41598-018-37699-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Jiao L., Shubbar M., Yang X., Zhang Q., Chen S., Wu Q., Chen Z., Rizo J., Liu X. A partially disordered region connects gene repression and activation functions of EZH2. Proc Natl Acad Sci U S A. 2020;117:16992–17002. doi: 10.1073/pnas.1914866117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sander S., Bullinger L., Klapproth K., Fiedler K., Kestler H.A., Barth T.F.E., Möller P., Stilgenbauer S., Pollack J.R., Wirth T. MYC stimulates EZH2 expression by repression of its negative regulator miR-26a. Blood. 2008;112:4202–4212. doi: 10.1182/blood-2008-03-147645. [DOI] [PubMed] [Google Scholar]
- 9.Cao P., Deng Z., Wan M., Huang W., Cramer S.D., Xu J., Lei M., Sui G. MicroRNA-101 negatively regulates Ezh2 and its expression is modulated by androgen receptor and HIF-1α/HIF-1β. Mol Cancer. 2010;9:108. doi: 10.1186/1476-4598-9-108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chen K., Xiao H., Zeng J., Yu G., Zhou H., Huang C., Yao W., Xiao W., Hu J., Guan W., Wu L., Huang J., Huang Q., Xu H., Ye Z. Alternative splicing of EZH2 pre-mRNA by SF3B3 contributes to the tumorigenic potential of renal cancer. Clin Cancer Res. 2017;23:3428–3441. doi: 10.1158/1078-0432.CCR-16-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cha T.-L., Zhou B.P., Xia W., Wu Y., Yang C.-C., Chen C.-T., Ping B., Otte A.P., Hung M.-C. Akt-mediated phosphorylation of EZH2 suppresses methylation of lysine 27 in histone H3. Science. 2005;310:306–310. doi: 10.1126/science.1118947. [DOI] [PubMed] [Google Scholar]
- 12.Kim E., Kim M., Woo D.-H., Shin Y., Shin J., Chang N., Oh Y T., Kim H., Rheey J., Nakano I., Lee C., Joo K M., Rich J N., Nam D.-H., Lee J. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell. 2013;23:839–852. doi: 10.1016/j.ccr.2013.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yan J., Li B., Lin B., Lee P.T., Chung T.-H., Tan J., Bi C., Lee X.T., Selvarajan V., Ng S.-B., Yang H., Yu Q., Chng W.-J. EZH2 phosphorylation by JAK3 mediates a switch to noncanonical function in natural killer/T-cell lymphoma. Blood. 2016;128:948–958. doi: 10.1182/blood-2016-01-690701. [DOI] [PubMed] [Google Scholar]
- 14.Jin X., Yang C., Fan P., Xiao J., Zhang W., Zhan S., Liu T., Wang D., Wu H. CDK5/FBW7-dependent ubiquitination and degradation of EZH2 inhibits pancreatic cancer cell migration and invasion. J Biol Chem. 2017;292:6269–6280. doi: 10.1074/jbc.M116.764407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wan L., Xu K., Wei Y., Zhang J., Han T., Fry C., Zhang Z., Wang Y.V., Huang L., Yuan M., Xia W., Chang W.-C., Huang W.-C., Liu C.-L., Chang Y.-C., Liu J., Wu Y., Jin V.X., Dai X., Guo J., Liu J., Jiang S., Li J., Asara J.M., Brown M., Hung M.-C., Wei W. Phosphorylation of EZH2 by AMPK suppresses PRC2 methyltransferase activity and oncogenic function. Mol Cell. 2018;69:279–291.e5. doi: 10.1016/j.molcel.2017.12.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chen S., Bohrer L.R., Rai A.N., Pan Y., Gan L., Zhou X., Bagchi A., Simon J.A., Huang H. Cyclin-dependent kinases regulate epigenetic gene silencing through phosphorylation of EZH2. Nat Cell Biol. 2010;12:1108. doi: 10.1038/ncb2116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Li Z., Hou P., Fan D., Dong M., Ma M., Li H., Yao R., Li Y., Wang G., Geng P., Mihretab A., Liu D., Zhang Y., Huang B., Lu J. The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer. Cell Death Differ. 2017;24:59–71. doi: 10.1038/cdd.2016.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kaneko S., Li G., Son J., Xu C.-F., Margueron R., Neubert T.A., Reinberg D. Phosphorylation of the PRC2 component Ezh2 is cell cycle-regulated and up-regulates its binding to ncRNA. Genes Dev. 2010;24:2615–2620. doi: 10.1101/gad.1983810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ko H.-W., Lee H.-H., Huo L., Xia W., Yang C.-C., Hsu J.L., Li L.-Y., Lai C.-C., Chan L.-C., Cheng C.-C., Labaff A.M., Liao H.-W., Lim S.-O., Li C.-W., Wei Y., Nie L., Yamaguchi H., Hung M.-C. GSK3β inactivation promotes the oncogenic functions of EZH2 and enhances methylation of H3K27 in human breast cancers. Oncotarget. 2016;7:57131–57144. doi: 10.18632/oncotarget.11008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Consalvi S., Brancaccio A., Dall'Agnese A., Puri P.L., Palacios D. Praja1 E3 ubiquitin ligase promotes skeletal myogenesis through degradation of EZH2 upon p38α activation. Nat Commun. 2017;8:13956. doi: 10.1038/ncomms13956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Palacios D., Mozzetta C., Consalvi S., Caretti G., Saccone V., Proserpio V., Marquez V.E., Valente S., Mai A., Forcales S.V., Sartorelli V., Puri P.L. TNF/p38 alpha/polycomb signalling to Pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration. Cell Stem Cell. 2010;7:455–469. doi: 10.1016/j.stem.2010.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.McMullen E.R., Skala S.L., Gonzalez M.E., Djomehri S., Chandrashekar D.S., Varambally S., Kleer C.G. Subcellular localization of EZH2 phosphorylated at T367 stratifies metaplastic breast carcinoma subtypes. Breast Cancer. 2020;28:496–505. doi: 10.1007/s12282-020-01189-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Yang C.-C., LaBaff A., Wei Y., Nie L., Xia W., Huo L., Yamaguchi H., Hsu Y.-H., Hsu J.L., Liu D., Lang J., Du Y., Lien H.-C., Li L.-Y., Deng R., Chan L.-C., Yao J., Kleer C.G., Hortobagyi G.N., Hung M.-C. Phosphorylation of EZH2 at T416 by CDK2 contributes to the malignancy of triple negative breast cancers. Am J Transl Res. 2015;7:1009–1020. [PMC free article] [PubMed] [Google Scholar]
- 24.Minnebo N., Görnemann J., O'Connell N., Van Dessel N., Derua R., Vermunt M.W., Page R., Beullens M., Peti W., Van Eynde A., Bollen M. NIPP1 maintains EZH2 phosphorylation and promoter occupancy at proliferation-related target genes. Nucleic Acids Res. 2013;41:842–854. doi: 10.1093/nar/gks1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wu S.C., Zhang Y. Cyclin-dependent kinase 1 (CDK1)-mediated phosphorylation of enhancer of zeste 2 (Ezh2) regulates its stability. J Biol Chem. 2011;286:28511–28519. doi: 10.1074/jbc.M111.240515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wei Y., Chen Y.-H., Li L.-Y., Lang J., Yeh S.-P., Shi B., Yang C.-C., Yang J.-Y., Lin C.-Y., Lai C.-C., Hung M.-C. CDK1-dependent phosphorylation of EZH2 suppresses methylation of H3K27 and promotes osteogenic differentiation of human mesenchymal stem cells. Nat Cell Biol. 2011;13:87–94. doi: 10.1038/ncb2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Nakanishi Y., Reina-Campos M., Nakanishi N., Llado V., Elmen L., Peterson S., Campos A., De S.K., Leitges M., Ikeuchi H., Pellecchia M., Blumberg R.S., Diaz-Meco M.T., Moscat J. Control of Paneth cell fate, intestinal inflammation, and tumorigenesis by PKCλ/ι. Cell Rep. 2016;16:3297–3310. doi: 10.1016/j.celrep.2016.08.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sahasrabuddhe A.A., Chen X., Chung F., Velusamy T., Lim M.S., Elenitoba-Johnson K.S.J. Oncogenic Y641 mutations in EZH2 prevent Jak2/β-TrCP-mediated degradation. Oncogene. 2014;34:445–454. doi: 10.1038/onc.2013.571. [DOI] [PubMed] [Google Scholar]
- 29.Li J., Hart R.P., Mallimo E.M., Swerdel M.R., Kusnecov A.W., Herrup K. EZH2-mediated H3K27 trimethylation mediates neurodegeneration in ataxia-telangiectasia. Nat Neurosci. 2013;16:1745. doi: 10.1038/nn.3564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chu C.-S., Lo P.-W., Yeh Y.-H., Hsu P.-H., Peng S.-H., Teng Y.-C., Kang M.-L., Wong C.-H., Juan L.-J. O-GlcNAcylation regulates EZH2 protein stability and function. Proc Natl Acad Sci U S A. 2014;111:1355–1360. doi: 10.1073/pnas.1323226111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wan J., Zhan J., Li S., Ma J., Xu W., Liu C., Xue X., Xie Y., Fang W., Chin Y.E., Zhang H. PCAF-primed EZH2 acetylation regulates its stability and promotes lung adenocarcinoma progression. Nucleic Acids Res. 2015;43:3591–3604. doi: 10.1093/nar/gkv238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zeng Y., Qiu R., Yang Y., Gao T., Zheng Y., Huang W., Gao J., Zhang K., Liu R., Wang S., Hou Y., Yu W., Leng S., Feng D., Liu W., Zhang X., Wang Y. Regulation of EZH2 by SMYD2-mediated lysine methylation is implicated in tumorigenesis. Cell Rep. 2019;29:1482–1498.e4. doi: 10.1016/j.celrep.2019.10.004. [DOI] [PubMed] [Google Scholar]
- 33.Li Z., Wang D., Lu J., Huang B., Wang Y., Dong M., Fan D., Li H., Gao Y., Hou P., Li M., Liu H., Pan Z.-Q., Zheng J., Bai J. Methylation of EZH2 by PRMT1 regulates its stability and promotes breast cancer metastasis. Cell Death Differ. 2020;27:3226–3242. doi: 10.1038/s41418-020-00615-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lee C.-H., Yu J.-R., Granat J., Saldaña-Meyer R., Andrade J., LeRoy G., Jin Y., Lund P., Stafford J.M., Garcia B.A., Ueberheide B., Reinberg D. Automethylation of PRC2 promotes H3K27 methylation and is impaired in H3K27M pediatric glioma. Genes Dev. 2019;33:1428–1440. doi: 10.1101/gad.328773.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wang X., Long Y., Paucek R.D., Gooding A.R., Lee T., Burdorf R.M., Cech T.R. Regulation of histone methylation by automethylation of PRC2. Genes Dev. 2019;33:1416–1427. doi: 10.1101/gad.328849.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yu Y.-L., Chou R.-H., Shyu W.-C., Hsieh S.-C., Wu C.-S., Chiang S.-Y., Chang W.-J., Chen J.-N., Tseng Y.-J., Lin Y.-H., Lee W., Yeh S.-P., Hsu J.L., Yang C.-C., Hung S.-C., Hung M.-C. Smurf2-mediated degradation of EZH2 enhances neuron differentiation and improves functional recovery after ischaemic stroke. EMBO Mol Med. 2013;5:531–547. doi: 10.1002/emmm.201201783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hu P., Nebreda A.R., Hanenberg H., Kinnebrew G.H., Ivan M., Yoder M.C., Filippi M.-D., Broxmeyer H.E., Kapur R. P38α/JNK signaling restrains erythropoiesis by suppressing Ezh2-mediated epigenetic silencing of Bim. Nat Commun. 2018;9:3518. doi: 10.1038/s41467-018-05955-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Shen Z., Chen L., Yang X., Zhao Y., Pier E., Zhang X., Yang X., Xiong Y. Downregulation of Ezh2 methyltransferase by FOXP3: new insight of FOXP3 into chromatin remodeling? Biochim Biophys Acta. 2013;1833:2190–2200. doi: 10.1016/j.bbamcr.2013.05.014. [DOI] [PubMed] [Google Scholar]
- 39.Zoabi M., Sadeh R., de Bie P., Marquez V.E., Ciechanover A. PRAJA1 is a ubiquitin ligase for the polycomb repressive complex 2 proteins. Biochem Biophys Res Commun. 2011;408:393–398. doi: 10.1016/j.bbrc.2011.04.025. [DOI] [PubMed] [Google Scholar]
- 40.Lu W., Liu S., Li B., Xie Y., Izban M.G., Ballard B.R., Sathyanarayana S.A., Adunyah S.E., Matusik R.J., Chen Z. SKP2 loss destabilizes EZH2 by promoting TRAF6-mediated ubiquitination to suppress prostate cancer. Oncogene. 2017;36:1364–1373. doi: 10.1038/onc.2016.300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yamaguchi H., Du Y., Nakai K., Ding M., Chang S.S., Hsu J.L., Yao J., Wei Y., Nie L., Jiao S., Chang W.C., Chen C.H., Yu Y., Hortobagyi G.N., Hung M.C. EZH2 contributes to the response to PARP inhibitors through its PARP-mediated poly-ADP ribosylation in breast cancer. Oncogene. 2018;37:208–217. doi: 10.1038/onc.2017.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Varambally S., Dhanasekaran S.M., Zhou M., Barrette T.R., Kumar-Sinha C., Sanda M.G., Ghosh D., Pienta K.J., Sewalt R.G.A.B., Otte A.P., Rubin M.A., Chinnaiyan A.M. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419:624–629. doi: 10.1038/nature01075. [DOI] [PubMed] [Google Scholar]
- 43.Cao Q., Yu J., Dhanasekaran S.M., Kim J.H., Mani R.S., Tomlins S.A., Mehra R., Laxman B., Cao X., Yu J., Kleer C.G., Varambally S., Chinnaiyan A.M. Repression of E-cadherin by the polycomb group protein EZH2 in cancer. Oncogene. 2008;27:7274–7284. doi: 10.1038/onc.2008.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bachmann I.M., Halvorsen O.J., Collett K., Stefansson I.M., Straume O., Haukaas S.A., Salvesen H.B., Otte A.P., Akslen L.A. EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast. J Clin Oncol. 2006;24:268–273. doi: 10.1200/JCO.2005.01.5180. [DOI] [PubMed] [Google Scholar]
- 45.Kleer C.G., Cao Q., Varambally S., Shen R., Ota I., Tomlins S.A., Ghosh D., Sewalt R.G.A.B., Otte A.P., Hayes D.F., Sabel M.S., Livant D., Weiss S.J., Rubin M.A., Chinnaiyan A.M. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A. 2003;100:11606–11611. doi: 10.1073/pnas.1933744100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gonzalez M.E., DuPrie M.L., Krueger H., Merajver S.D., Ventura A.C., Toy K.A., Kleer C.G. Histone methyltransferase EZH2 induces Akt-dependent genomic instability and BRCA1 inhibition in breast cancer. Cancer Res. 2011;71:2360–2370. doi: 10.1158/0008-5472.CAN-10-1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pang J., Toy K.A., Griffith K.A., Awuah B., Quayson S., Newman L.A., Kleer C.G. Invasive breast carcinomas in Ghana: high frequency of high grade, basal-like histology and high EZH2 expression. Breast Cancer Res Treat. 2012;135:59–66. doi: 10.1007/s10549-012-2055-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Moore H.M., Gonzalez M.E., Toy K.A., Cimino-Mathews A., Argani P., Kleer C.G. EZH2 inhibition decreases p38 signaling and suppresses breast cancer motility and metastasis. Breast Cancer Res Treat. 2013;138:741–752. doi: 10.1007/s10549-013-2498-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chang C.-J., Yang J.-Y., Xia W., Chen C.-T., Xie X., Chao C.-H., Woodward W.A., Hsu J.-M., Hortobagyi G.N., Hung M.-C. EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-β-catenin signaling. Cancer Cell. 2011;19:86–100. doi: 10.1016/j.ccr.2010.10.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gonzalez M.E., Li X., Toy K., DuPrie M., Ventura A.C., Banerjee M., Ljungman M., Merajver S.D., Kleer C.G. Downregulation of EZH2 decreases growth of estrogen receptor-negative invasive breast carcinoma and requires BRCA1. Oncogene. 2009;28:843–853. doi: 10.1038/onc.2008.433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gonzalez M.E., Moore H.M., Li X., Toy K.A., Huang W., Sabel M.S., Kidwell K.M., Kleer C.G. EZH2 expands breast stem cells through activation of NOTCH1 signaling. Proc Natl Acad Sci U S A. 2014;111:3098–3103. doi: 10.1073/pnas.1308953111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Fujii S., Ito K., Ito Y., Ochiai A. Enhancer of zeste homologue 2 (EZH2) down-regulates RUNX3 by increasing histone H3 methylation. J Biol Chem. 2008;283:17324–17332. doi: 10.1074/jbc.M800224200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kim K.H., Roberts C.W.M. Targeting EZH2 in cancer. Nat Med. 2016;22:128–134. doi: 10.1038/nm.4036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hussain M., Rao M., Humphries A.E., Hong J.A., Liu F., Yang M., Caragacianu D., Schrump D.S. Tobacco smoke induces polycomb-mediated repression of Dickkopf-1 in lung cancer cells. Cancer Res. 2009;69:3570–3578. doi: 10.1158/0008-5472.CAN-08-2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Morin R.D., Johnson N.A., Severson T.M., Mungall A.J., An J., Goya R. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet. 2010;42:181–185. doi: 10.1038/ng.518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Morin R.D., Mendez-Lago M., Mungall A.J., Goya R., Mungall K.L., Corbett R.D. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476:298–303. doi: 10.1038/nature10351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.McCabe M.T., Graves A.P., Ganji G., Diaz E., Halsey W.S., Jiang Y., Smitheman K.N., Ott H.M., Pappalardi M.B., Allen K.E., Chen S.B., Della Pietra A., 3rd, Dul E., Hughes A.M., Gilbert S.A., Thrall S.H., Tummino P.J., Kruger R.G., Brandt M., Schwartz B., Creasy C.L. Mutation of A677 in histone methyltransferase EZH2 in human B-cell lymphoma promotes hypertrimethylation of histone H3 on lysine 27 (H3K27) Proc Natl Acad Sci U S A. 2012;109:2989–2994. doi: 10.1073/pnas.1116418109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Souroullas G.P., Jeck W.R., Parker J.S., Simon J.M., Liu J.-Y., Paulk J., Xiong J., Clark K.S., Fedoriw Y., Qi J., Burd C.E., Bradner J.E., Sharpless N.E. An oncogenic Ezh2 mutation induces tumors through global redistribution of histone 3 lysine 27 trimethylation. Nat Med. 2016;22:632–640. doi: 10.1038/nm.4092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gulati N., Béguelin W., Giulino-Roth L. Enhancer of zeste homolog 2 (EZH2) inhibitors. Leuk Lymphoma. 2018;59:1574–1585. doi: 10.1080/10428194.2018.1430795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Knutson S.K., Warholic N.M., Wigle T.J., Klaus C.R., Allain C.J., Raimondi A., Porter Scott M., Chesworth R., Moyer M.P., Copeland R.A., Richon V.M., Pollock R.M., Kuntz K.W., Keilhack H. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci U S A. 2013;110:7922–7927. doi: 10.1073/pnas.1303800110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Januario T., Ye X., Bainer R., Alicke B., Smith T., Haley B., Modrusan Z., Gould S., Yauch R.L. PRC2-mediated repression of SMARCA2 predicts EZH2 inhibitor activity in SWI/SNF mutant tumors. Proc Natl Acad Sci U S A. 2017;114:12249–12254. doi: 10.1073/pnas.1703966114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kim W., Bird G.H., Neff T., Guo G., Kerenyi M.A., Walensky L.D., Orkin S.H. Targeted disruption of the EZH2-EED complex inhibits EZH2-dependent cancer. Nat Chem Biol. 2013;9:643–650. doi: 10.1038/nchembio.1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hussein Y.R., Sood A.K., Bandyopadhyay S., Albashiti B., Semaan A., Nahleh Z., Roh J., Han H.D., Lopez-Berestein G., Ali-Fehmi R. Clinical and biological relevance of enhancer of zeste homolog 2 in triple-negative breast cancer. Hum Pathol. 2012;43:1638–1644. doi: 10.1016/j.humpath.2011.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang X., Hu B., Shen H., Zhou H., Xue X., Chen Y., Chen S., Han Y., Yuan B., Zhao H., Zhi Q., Kuang Y. Clinical and prognostic relevance of EZH2 in breast cancer: a meta-analysis. Biomed Pharmacother. 2015;75:218–225. doi: 10.1016/j.biopha.2015.07.038. [DOI] [PubMed] [Google Scholar]
- 65.Holm K., Grabau D., Lövgren K., Aradottir S., Gruvberger-Saal S., Howlin J., Saal L.H., Ethier S.P., Bendahl P.-O., Stål O., Malmström P., Fernö M., Rydén L., Hegardt C., Borg Å., Ringnér M. Global H3K27 trimethylation and EZH2 abundance in breast tumor subtypes. Mol Oncol. 2012;6:494–506. doi: 10.1016/j.molonc.2012.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wu Y., Zhang Z., Cenciarini M.E., Proietti C.J., Amasino M., Hong T., Yang M., Liao Y., Chiang H.-C., Kaklamani V.G., Jeselsohn R., Vadlamudi R.K., Huang T.H.-M., Li R., De Angelis C., Fu X., Elizalde P.V., Schiff R., Brown M., Xu K. Tamoxifen resistance in breast cancer is regulated by the EZH2-ERα-GREB1 transcriptional axis. Cancer Res. 2018;78:671–684. doi: 10.1158/0008-5472.CAN-17-1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Beca F., Kensler K., Glass B., Schnitt S.J., Tamimi R.M., Beck A.H. EZH2 protein expression in normal breast epithelium and risk of breast cancer: results from the Nurses' Health Studies. Breast Cancer Res. 2017;19:21. doi: 10.1186/s13058-017-0817-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ding L., Erdmann C., Chinnaiyan A.M., Merajver S.D., Kleer C.G. Identification of EZH2 as a molecular marker for a precancerous state in morphologically normal breast tissues. Cancer Res. 2006;66:4095–4099. doi: 10.1158/0008-5472.CAN-05-4300. [DOI] [PubMed] [Google Scholar]
- 69.Li X., Gonzalez M.E., Toy K., Filzen T., Merajver S.D., Kleer C.G. Targeted overexpression of EZH2 in the mammary gland disrupts ductal morphogenesis and causes epithelial hyperplasia. Am J Pathol. 2009;175:1246–1254. doi: 10.2353/ajpath.2009.090042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Zeidler M., Varambally S., Cao Q., Chinnaiyan A.M., Ferguson D.O., Merajver S.D., Kleer C.G. The polycomb group protein EZH2 impairs DNA repair in breast epithelial cells. Neoplasia. 2005;7:1011–1019. doi: 10.1593/neo.05472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ren G., Baritaki S., Marathe H., Feng J., Park S., Beach S., Bazeley P.S., Beshir A.B., Fenteany G., Mehra R., Daignault S., Al-Mulla F., Keller E., Bonavida B., de la Serna I., Yeung K.C. Polycomb protein EZH2 regulates tumor invasion via the transcriptional repression of the metastasis suppressor RKIP in breast and prostate cancer. Cancer Res. 2012;72:3091–3104. doi: 10.1158/0008-5472.CAN-11-3546. [DOI] [PubMed] [Google Scholar]
- 72.Bae W.K., Yoo K.H., Lee J.S., Kim Y., Chung I.-J., Park M.H., Yoon J.H., Furth P.A., Hennighausen L. The methyltransferase EZH2 is not required for mammary cancer development, although high EZH2 and low H3K27me3 correlate with poor prognosis of ER-positive breast cancers. Mol Carcinog. 2015;54:1172–1180. doi: 10.1002/mc.22188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lee S T., Li Z., Wu Z., Aau M., Guan P., Karuturi R.K.M., Liou Y C., Yu Q. Context-specific regulation of NF-κB target gene expression by EZH2 in breast cancers. Mol Cell. 2011;43:798–810. doi: 10.1016/j.molcel.2011.08.011. [DOI] [PubMed] [Google Scholar]
- 74.Lawrence C.L., Baldwin A.S. Non-canonical EZH2 transcriptionally activates RelB in triple negative breast cancer. PLoS One. 2016;11:e0165005-e. [Google Scholar]
- 75.Shi B., Liang J., Yang X., Wang Y., Zhao Y., Wu H., Sun L., Zhang Y., Chen Y., Li R., Zhang Y., Hong M., Shang Y. Integration of estrogen and Wnt signaling circuits by the polycomb group protein EZH2 in breast cancer cells. Mol Cell Biol. 2007;27:5105–5119. doi: 10.1128/MCB.00162-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Li J., Xi Y., Li W., McCarthy R.L., Stratton S.A., Zou W., Li W., Dent S.Y., Jain A.K., Barton M.C. TRIM28 interacts with EZH2 and SWI/SNF to activate genes that promote mammosphere formation. Oncogene. 2017;36:2991–3001. doi: 10.1038/onc.2016.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Jung H.-Y., Jun S., Lee M., Kim H.-C., Wang X., Ji H., McCrea P.D., Park J.-I. PAF and EZH2 induce Wnt/β-catenin signaling hyperactivation. Mol Cell. 2013;52:193–205. doi: 10.1016/j.molcel.2013.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kikuchi J., Koyama D., Wada T., Izumi T., Hofgaard P.O., Bogen B., Furukawa Y. Phosphorylation-mediated EZH2 inactivation promotes drug resistance in multiple myeloma. J Clin Invest. 2015;125:4375–4390. doi: 10.1172/JCI80325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sviderskiy V.O., Blumenberg L., Gorodetsky E., Karakousi T.R., Hirsh N., Alvarez S.W., Terzi E.M., Kaparos E., Whiten G.C., Ssebyala S., Tonzi P., Mir H., Neel B.G., Huang T.T., Adams S., Ruggles K.V., Possemato R. Hyperactive CDK2 activity in basal-like breast cancer imposes a genome integrity liability that can be exploited by targeting DNA polymerase ε. Mol Cell. 2020;80:682–698.e7. doi: 10.1016/j.molcel.2020.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Anwar T., Arellano-Garcia C., Ropa J., Chen Y.-C., Kim H.S., Yoon E., Grigsby S., Basrur V., Nesvizhskii A.I., Muntean A., Gonzalez M.E., Kidwell K.M., Nikolovska-Coleska Z., Kleer C.G. p38-mediated phosphorylation at T367 induces EZH2 cytoplasmic localization to promote breast cancer metastasis. Nat Commun. 2018;9:2801. doi: 10.1038/s41467-018-05078-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hoffmeyer K., Junghans D., Kanzler B., Kemler R. Trimethylation and acetylation of β-catenin at lysine 49 represent key elements in ESC pluripotency. Cell Rep. 2017;18:2815–2824. doi: 10.1016/j.celrep.2017.02.076. [DOI] [PubMed] [Google Scholar]
- 82.Ardehali M.B., Anselmo A., Cochrane J.C., Kundu S., Sadreyev R.I., Kingston R.E. Polycomb repressive complex 2 methylates elongin A to regulate transcription. Mol Cell. 2017;68:872–884.e6. doi: 10.1016/j.molcel.2017.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wurm S., Zhang J., Guinea-Viniegra J., García F., Muñoz J., Bakiri L., Ezhkova E., Wagner E.F. Terminal epidermal differentiation is regulated by the interaction of Fra-2/AP-1 with Ezh2 and ERK1/2. Genes Dev. 2015;29:144–156. doi: 10.1101/gad.249748.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.He A., Shen X., Ma Q., Cao J., von Gise A., Zhou P., Wang G., Marquez V.E., Orkin S.H., Pu W.T. PRC2 directly methylates GATA4 and represses its transcriptional activity. Genes Dev. 2012;26:37–42. doi: 10.1101/gad.173930.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kogure M., Takawa M., Saloura V., Sone K., Piao L., Ueda K., Ibrahim R., Tsunoda T., Sugiyama M., Atomi Y., Nakamura Y., Hamamoto R. The oncogenic polycomb histone methyltransferase EZH2 methylates lysine 120 on histone H2B and competes ubiquitination. Neoplasia. 2013;15:1251–1261. doi: 10.1593/neo.131436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Sanulli S., Justin N., Teissandier A., Ancelin K., Portoso M., Caron M., Michaud A., Lombard B., da Rocha S T., Offer J., Loew D., Servant N., Wassef M., Burlina F., Gamblin S J., Heard E., Margueron R. Jarid2 methylation via the PRC2 complex regulates H3K27me3 deposition during cell differentiation. Mol Cell. 2015;57:769–783. doi: 10.1016/j.molcel.2014.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kasinath V., Faini M., Poepsel S., Reif D., Feng X.A., Stjepanovic G., Aebersold R., Nogales E. Structures of human PRC2 with its cofactors AEBP2 and JARID2. Science. 2018;359:940–944. doi: 10.1126/science.aar5700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Vasanthakumar A., Xu D., Lun A.T., Kueh A.J., van Gisbergen K.P., Iannarella N., Li X., Yu L., Wang D., Williams B.R., Lee S.C., Majewski I.J., Godfrey D.I., Smyth G.K., Alexander W.S., Herold M.J., Kallies A., Nutt S.L., Allan R.S. A non-canonical function of Ezh2 preserves immune homeostasis. EMBO Rep. 2017;18:619–631. doi: 10.15252/embr.201643237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lee J M., Lee J S., Kim H., Kim K., Park H., Kim J.-Y., Lee S H., Kim I.S., Kim J., Lee M., Chung C H., Seo S.-B., Yoon J.-B., Ko E., Noh D.-Y., Kim K I., Kim K K., Baek S H. EZH2 generates a methyl degron that is recognized by THE DCAF1/DDB1/CUL4 E3 ubiquitin ligase complex. Mol Cell. 2012;48:572–586. doi: 10.1016/j.molcel.2012.09.004. [DOI] [PubMed] [Google Scholar]
- 90.Dasgupta M., Dermawan J.K.T., Willard B., Stark G.R. STAT3-driven transcription depends upon the dimethylation of K49 by EZH2. Proc Natl Acad Sci U S A. 2015;112:3985–3990. doi: 10.1073/pnas.1503152112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gunawan M., Venkatesan N., Loh J.T., Wong J.F., Berger H., Neo W.H., Li L.Y.J., La Win M.K., Yau Y.H., Guo T., See P.C.E., Yamazaki S., Chin K.C., Gingras A.R., Shochat S.G., Ng L.G., Sze S.K., Ginhoux F., Su I.H. The methyltransferase Ezh2 controls cell adhesion and migration through direct methylation of the extranuclear regulatory protein talin. Nat Immunol. 2015;16:505–516. doi: 10.1038/ni.3125. [DOI] [PubMed] [Google Scholar]
- 92.Mahara S., Lee P.L., Feng M., Tergaonkar V., Chng W.J., Yu Q. HIFI-α activation underlies a functional switch in the paradoxical role of Ezh2/PRC2 in breast cancer. Proc Natl Acad Sci U S A. 2016;113:E3735–E3744. doi: 10.1073/pnas.1602079113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hirukawa A., Smith H.W., Zuo D., Dufour C.R., Savage P., Bertos N., Johnson R.M., Bui T., Bourque G., Basik M., Giguère V., Park M., Muller W.J. Targeting EZH2 reactivates a breast cancer subtype-specific anti-metastatic transcriptional program. Nat Commun. 2018;9:2547. doi: 10.1038/s41467-018-04864-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yomtoubian S., Lee S.B., Verma A., Izzo F., Markowitz G., Choi H., Cerchietti L., Vahdat L., Brown K.A., Andreopoulou E., Elemento O., Chang J., Inghirami G., Gao D., Ryu S., Mittal V. Inhibition of EZH2 catalytic activity selectively targets a metastatic subpopulation in triple-negative breast cancer. Cell Rep. 2020;30:755–770.e6. doi: 10.1016/j.celrep.2019.12.056. [DOI] [PubMed] [Google Scholar]
- 95.Eich M.L., Athar M., Ferguson J.E., 3rd, Varambally S. EZH2-targeted therapies in cancer: hype or a reality. Cancer Res. 2020;80:5449–5458. doi: 10.1158/0008-5472.CAN-20-2147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hsu J.H.-R., Rasmusson T., Robinson J., Pachl F., Read J., Kawatkar S., O’ Donovan D.H., Bagal S., Code E., Rawlins P., Argyrou A., Tomlinson R., Gao N., Zhu X., Chiarparin E., Jacques K., Shen M., Woods H., Bednarski E., Wilson D.M., Drew L., Castaldi M.P., Fawell S., Bloecher A. EED-targeted PROTACs degrade EED, EZH2, and SUZ12 in the PRC2 complex. Cell Chem Biol. 2020;27:41–46.e17. doi: 10.1016/j.chembiol.2019.11.004. [DOI] [PubMed] [Google Scholar]
- 97.Potjewyd F., Turner A.W., Beri J., Rectenwald J.M., Norris-Drouin J.L., Cholensky S.H., Margolis D.M., Pearce K.H., Herring L.E., James L.I. Degradation of polycomb repressive complex 2 with an EED-targeted bivalent chemical degrader. Cell Chem Biol. 2020;27:47–56.e15. doi: 10.1016/j.chembiol.2019.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Ma A., Stratikopoulos E., Park K.-S., Wei J., Martin T.C., Yang X., Schwarz M., Leshchenko V., Rialdi A., Dale B., Lagana A., Guccione E., Parekh S., Parsons R., Jin J. Discovery of a first-in-class EZH2 selective degrader. Nat Chem Biol. 2020;16:214–222. doi: 10.1038/s41589-019-0421-4. [DOI] [PMC free article] [PubMed] [Google Scholar]