Abstract
Over 60% of supratentorial (ST) ependymomas harbor a ZFTA-RELA (ZRfus) gene fusion (formerly C11orf95-RELA). To study the biology of ZRfus, we developed an autochthonous mouse tumor model using in utero electroporation (IUE) of the embryonic mouse brain. Integrative epigenomic and transcriptomic mapping was performed on IUE driven ZRfus tumors by CUT&RUN, ChIP, ATAC, and RNA sequencing and compared to human ZRfus driven ependymoma. In addition to direct canonical NF-κB pathway activation, ZRfus dictates a neoplastic transcriptional program and binds to thousands of unique sites across the genome that are enriched with Plagl family transcription factor (TF) motifs. ZRfus activates gene expression programs through recruitment of transcriptional co-activators (Brd4, Ep300, Cbp, Pol2) that are amenable to pharmacologic inhibition. Downstream ZRfus target genes converge on developmental programs marked by Plagl transcription factor proteins, and activate neoplastic programs enriched in Mapk, focal adhesion, and gene imprinting networks.
INTRODUCTION
Ependymoma is an aggressive and chemo-resistant pediatric brain tumor, with treatment limited to surgical resection and radiation (1). Although histologically similar, ependymomas are divided into at least nine molecular subtypes associated with distinct genetic and epigenetic alterations (2–6). Over 60% of supratentorial (ST) ependymomas are characterized by an oncogenic fusion between zinc finger translocation associated (ZFTA, formerly C11orf95) and v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA) (7). Gene fusions involving YAP1 or ZFTA and other gene partners are less frequent (3). ZFTA-RELA fusion (denoted ZRfus) protein is typically the sole genetic driver detected in ST ependymoma (7). When expressed in the developing mouse brain, or in transplanted neural stem cells (NSCs), ZRfus is capable of cellular transformation and tumor initiation (7,8). These findings suggest that the ZRfus protein and its direct target genes may represent candidates for therapy.
RELA (also known as p65 protein) is a transcription factor that mediates NF-κB pathway activation in diverse processes such as inflammation, cellular metabolism, and chemotaxis (9). In a normal cellular context, the majority of RELA is sequestered in the cytosol by IkBα protein. Tumor necrosis factor alpha (TNF-α) or interleukin-1 (IL-1) cytokine induction results in phosphorylation of IkBα, proteasomal degradation, and translocation of RELA to the nucleus. Within the nuclear compartment, RELA undergoes post-translational modifications to recruit transcriptional co-activators, such as BRD4, to activate inflammatory gene expression programs (10). This occurs via binding of RELA to both enhancers and stretch-super enhancers and accumulation of active chromatin modifications, demarcated by H3K27 acetylation (H3K27ac) (10,11). RELA protein interacts as both homo-dimers and hetero-dimers with nuclear factor kappa B subunit 1 (NFKB1/P50) to activate target gene expression (9).
While the normal molecular function of ZFTA protein is currently unknown, its gene fusion with RELA results in constitutive nuclear translocation (7). The fragment of ZFTA, that undergoes gene fusion, contains at least one to four zinc finger domains suggesting that this fragment may contribute to novel DNA binding and transcriptional regulation (7). Additionally, ZFTA fuses to other genes, often transcription factors (TF) or transcriptional co-activators, such as YAP1 and MAML2 in ependymoma, or Myocardin-like 2 (MKL2) in chondroid lipomas (7,12). Our study reveals that ZRfus modulates the tumor epigenome through de novo binding of DNA at specific motifs. Accompanying ZRfus binding are transcriptional complexes associated with wide-spread patterns of active chromatin, through recruitment of Brd4, Ep300, and Crebbp(Cbp), for which specific small molecule inhibitors are currently available. Elucidation of transcriptional co-regulators and mapping of ZRfus downstream genes reveals molecular targets and pathways that may contribute to ependymoma development, and may represent leads for future therapy.
RESULTS
An autochthonous model of mouse ZFTA-Rela ependymoma
To investigate the molecular function of the ZRfus oncogene in ependymoma, we utilized PiggyBAC (pB) transposon based in utero electroporation (IUE) technology to model the most common Type 1 ZRfus fusion variant (denoted ZRfus1, Fig.1A–D). As a control, we introduced green fluorescence protein (GFP) pB vectors by IUE in age-matched mice which did not generate tumors or affect mouse survival. IUE technology provided several advantages: 1) Over-expression of ZRfus1 in a native context during embryonic brain development, 2) Simultaneous introduction of HA-tagged ZRfus1 along with DNA constructs: GFP, luciferase, and CRISPR/Cas9 deletion of Trp53, and 3) Study of ZRfus1 tumor biology all within an immune-competent environment (13). The resulting mouse tumors recapitulated histologic features of ependymoma (Fig.1B) and showed sustained ZRfus1 nuclear localization (Fig.1C). Consequently, the mice succumbed to brain tumors with a median age of about 60 days (Fig.1D). ZRfus1 tumors were micro-dissected by GFP positive signal and compared against contralateral normal brain (non-GFP positive) tissue by RNA-seq. Differential gene expression analysis and comparison against an independent RCAS-TVA ZRfus1 driven ependymoma mouse model demonstrated a significant correlation of neoplastic transcriptional programs compared to matched normal brain (Fig.1E, R=0.79, p < 2.2e–16, Supplementary Table S1, Supplemental Data Fig.S1) (8). The IUE ZRfus1 model positively associated with human ZRfus1 transcriptional programs as opposed to other ependymoma subtypes, including gene expression of Dlk1, Lmx1b, Ephb2, Igf2, Gli2, and Akt1 (Fig.1F–H). To further support the consistency between our mouse model and its reflection of the human disease, we directly compared the transcriptome of our IUE model to different ZRFUS1 mouse models and human ZRfus1 datasets described in Kupp et al., (Co-Submitted Cancer Discovery Manuscript)(14) and Zheng et al., (Co-Submitted Cancer Discovery Manuscript)(15). We observed a significant overlap between the datasets and derived a ZRfus1 93 gene signature consistent across all datasets, including our IUE model (Fig.1H, Supplementary Table S2). These findings demonstrate that the IUE ZRfus1 driven mouse model of ependymoma recapitulates several key aspects of human ZFTA-RELA ependymoma.
Figure 1. Establishment of a native ZFTA-Rela driven mouse model by embryonic in utero electroporation (IUE).
(A) Schematic of in utero electroporation technique used for introduction of piggyBAC DNA plasmids at embryonic day 16.5 (E16.5) (B) Luciferase, GFP imaging, and histology of IUE ZRfus1 murine ependymoma, and (C) confirmation of fusion protein nuclear localization. (D) Survival of mice reaching tumor endpoint as a result of ZRfus1expression. (E) Pearson correlation of IUE vs RCAS driven ZFTA-RELA mouse ependymoma(8) and (F) association with human ZFTA-RELA human tumor expression (18). (G) Comparison between genes upregulated in mouse IUE- ZRfus1 tumors vs matched normal brain and upregulated in ZRfus human ependymoma vs other ependymoma subtypes. Shared genes were restricted to those with a log2 fold change greater than 1. Agreement score represents the product of the fold change in mouse and human comparisons. (H) Waterfall plot of 93 ZFTA-Rela signature genes ordered by agreement score representing the product of Log2 fold change (ZRfus1/Normal Brain) and Log2 fold change (ZFTA-Rela/Non-Rela ependymoma).
ZRfus1 engages open and active chromatin at de novo promoter and enhancer loci
Using brain tumors from the IUE ZRfus1 mouse model, we acutely dissociated neoplastic GFP positive cells to perform two orthogonal methods of chromatin profiling, ChIP followed by sequencing (ChIP-seq) and CUT&RUN (Fig.2A–C, Supplemental Data Fig.S2A). CUT&RUN permitted native cross-linking-free genomic mapping of proteins and histone modifications and reduction of non-specific cross-linking artifacts often present in ChIP-seq experiments (16). ChIP-seq and CUT&RUN directed against the HA tag was performed on HA-tagged ZRfus1 protein, as well as CUT&RUN against Rela, H3K27ac, and IgG (negative control), and ATAC-seq (to measure accessible chromatin). Combining two independent experiments, we identified 6845 shared ZRfus1 binding sites. This set of peaks was further filtered to exclude non-specific signal using an IgG control. The resulting 5608 peaks significantly overlapped with regions also identified by ChIP-seq (Fig.2B,D, Supplemental Data Fig.S2). CUT&RUN of HA tagged ZRfus1 and Rela revealed mostly sub-nucleosomal binding of ZRfus1 with small fragment patterns (< 120 bp) as expected of transcription factor proteins (Fig.2A,C, Supplemental Data Fig.S2B,C) (16). ATAC-seq and H3K27ac CUT&RUN demonstrated that HA-ZRfus1 binding was localized to ‘open and accessible’ and active chromatin, respectively (Fig.2D–E) and enriched in both active enhancer and promoter loci (Fig.2F). ZRfus1 was bound to promoters and proximal enhancers of known ependymoma oncogenes such as Ephb2, Ccnd1, Akt1, and Notch1 (Fig.2G–H) (17,18). Interestingly, expression of the ZRfus1 protein led to a global elevation of H3K27ac, but also H3K27me3, a mark of poised and repressive chromatin (Supplemental Data Fig.S2D–F). These findings support that ZRfus1 protein directly engages DNA as a transcription factor oncoprotein, and is associated with open and active chromatin surrounding many genes, including known ependymoma oncogenes.
Figure 2. ZFTA-Rela binds open and active chromatin of promoter and enhancer loci.
(A) Schematic of proteins and epigenetic marks profiled using CUT&RUN, ChIP-seq, or ATAC-seq in IUE ZRfus1 ependymoma. (B) Venn diagram of the intersection between ZRfus1 peaks detected using CUT&RUN and ChIP-seq data. (C) CUT&RUN fragment size profiles of HA and Rela (p65). (D-E) Localization of ZRfus1 by HA-CUT&RUN and Rela-CUT&RUN in open chromatin (ATAC-seq) and actively transcribed chromatin (H3K27ac CUT&RUN). (F) Distribution of ZRfus1 binding sites at different genomic regions. (G-H) Binding profiles of ZRfus1 at the promoters and enhancers of ependymoma oncogenes.
Transcriptional co-activators and RNA polymerase are recruited to ZRfus1 bound genomic loci
To assess whether ZRfus1 activates gene expression programs, we performed RNA-seq. The ZRfus1 bound genes showed higher expression levels as compared to mouse contralateral normal brain (non-GFP+) tissue (Fig.3A). 920 ZRfus1 bound genes that were at least 2-fold upregulated were identified including common ZRfus1 signature genes such as: Ccnd1, Ephb2, and Gli2 (Fig.3B, p < 0.05, Supplementary Table S3). We reasoned that localization of ZRfus1 to enhancers and promoters might indicate aberrant recruitment of transcriptional activation complexes, particularly proteins involved in transcriptional initiation and elongation such as Ep300, Cbp, Brd4, and phosphorylated forms of RNA polymerase II (Serine 2 and 5 phosphorylation of the carboxy terminal domain). Supporting this, in the ZRfus1 IUE model, CUT&RUN signals demonstrated co-recruitment of Brd4, Ep300, Cbp, and Ser2/5 phosphorylated RNA polymerase II at most ZRfus1 binding sites. (Fig.3C, Supplemental Data Fig.S3). These findings are consistent with a manuscript co-submitted to Cancer Discovery by Kupp et al.,(14) in which ZRfus1 was shown to physically bind both Brd4 and Ep300 to promote gene transcription, and ZFTA was reported to facilitate Ep300 mediated gene activation.
Figure 3. Recruitment of transcriptional activation proteins to ZFTA-Rela binding sites.
(A-B) Gene expression of ZRfus1 targets in mouse tumors versus contralateral normal brain (C) Recruitment of transcriptional co-regulators Brd4, Ep300, Crebbp, Ser2-Pol2, and Ser5-Pol2 to ZRfus1 binding sites as compared to IgG control. (D) CRISPR-CAS9 KO of ZRfus1 and Rela (E) Pie chart depicting the number of genes that were down-regulated, up-regulated, or exhibited no change in expression between non-targeting and Rela KO (F) Heatmap of selected genes in the NF-kB, ZRfus1 targets, and neuronal differentiation pathway that were down-regulated or up-regulated (G) Differential gene expression between non-targeting and Rela KO experiments across three cell passages. Represented in the waterfall plot is the Log2 fold-change between KO versus non targeting across n = 24,428 genes/transcripts. (H) Pathway analysis of the top down-regulated genes following Rela-KO as compared to non-targeting controls.
To determine whether the effects on gene transcription were direct, we generated knockout (KO) models of cell lines derived from IUE:ZRfus1 using CRISPR/Cas9 deletion of Rela and ZRfus1 with varying efficiencies (Fig.3D). Using sgRNA:Rela targeting constructs, ZRfus1 KO resulted in decreased expression of 872 (23%) of ZRfus1 target genes while 173 (4%) were up-regulated, and 2780 (73%) unchanged (Fig.3E). Loss of ZRfus1 and Rela did not result in a global change in transcriptional output as measured by RNA ERCC spike-in controls (Supplemental Data Fig.S3A,B). Down-regulated genes included several ZRfus1 specific genes such as Dlk1, Lmx1b, and Akt1, and also members of the NF-kB pathway such as Cxcl2, Icam1, and Stat1 (Fig.3F–G, Supplementary Table S4, Supplemental Data Fig.S3C). Most notably, the predominant pathway down-regulated upon ZRfus1 and Rela KO included genes involved in nervous system development and neural cell differentiation (Fig.3H). The top genes down-regulated were markers of oligodendrocyte (Olig1/2) and radial glial cell identity (Fabp7), while the genes marking neuronal cells (Dlx5/6 and Gchfr) were up-regulated (19,20) (Fig 3F). Similarly, chemical inhibition of Ep300/Cbp using inhibitors A485 versus A486 (inactive isomeric control) significantly impaired expression of ZRfus1 target genes and was associated with decreased cellular viability at 72 hours of treatment (Supplemental Data Fig.S3 D–E) (21). Our data demonstrates that ZRfus1 recruits transcriptional activation complexes, including Brd4, Ep300, Cbp, and RNA Pol2 to drive neoplastic gene transcription. Genetic ablation of ZRfus1 or chemical inhibition of transcriptional co-regulators impairs ZRfus1 target gene expression and cell identity programs.
ZRfus1 binds Plagl TF family DNA motifs to promote neoplastic gene expression
We hypothesized that ZRfus1, which harbors a C2H2 zinc finger domain, would promote aberrant DNA binding in addition to its function to drive nuclear localization (7). To mark and remove canonical NF-κB binding sites from tumor-specific sites, we compared ZRfus1 binding against mouse embryonic fibroblasts treated with TNF-α, a method of stimulating Rela nuclear localization and studying inflammatory gene expression (22). The majority of Rela-bound sites in the context of inflammation were absent or weakly bound by ZRfus1 (Fig.4A). Tumor-specific DNA binding of ZRfus1 was enriched for the Plagl1 or Plagl2 transcription factor family motifs consisting of a core ‘GGGCC’ DNA sequence (Fig.4B, Enrichment: p < 1e−1488, Supplementary Table S5). Also, shared programs between tumor and inflammatory programs were enriched for both Rela and Plagl2 motifs (Fig.4B, Rela: 1e−387 and Plagl2: 1e−118, Supplementary Table S6). An enrichment of Plagl TF family motifs was also observed in the ZRfus1 HA ChIP-seq data thus further validating our findings using an orthogonal technique (Supplemental Data Fig. S4A–C). Interestingly, in a small number of cases, we identified Rela binding sites directly flanked by adjacent Plagl family motifs suggesting possible co-recruitment and utilization of endogenous Rela proteins (Supplemental Data Fig.S4D–G). Further, Tead family motifs were found to be enriched at a subset of tumor-specific and shared inflammatory loci, and also bound by H3K27ac, Brd4, Ep300 and Pol2 (Supplemental Data Fig.S4C). This is potentially relevant as other subtypes of supratentorial ependymoma are driven by YAP1 gene fusions, suggesting potentially shared oncogenic mechanisms(3,23).
Figure 4. ZFTA-Rela engages NF-κB and Plagl (‘GGGCC’) motifs.
(A) Comparison of HA- and Rela- CUT&RUN binding in ZRfus1 ependymoma against mouse embryonic fibroblasts cells treated with TNF-α. Motif enrichment analysis (B) and Distribution of peaks at different genomic regions (C) for tumor-specific, shared, and inflammation-specific binding sites. (D) Example of ZRfus1 binding at the Lmx1b locus, recruitment of Brd4 and Ep300, and presence of multiple Plagl motifs. (E) Lmx1b gene expression in IUE ZRfus1 tumor versus normal brain. (F) Comparison of DNA motifs identified in human ZFTA-RELA and non-ZFTA-RELA ependymoma using HOMER (see methods).
ZRfus1 binding profiles in a tumor-specific or shared context was comparable between enhancers and promoters, unlike enhancer and super enhancer centric profiles observed in canonical TNF-α driven Rela activation (Fig.4C) (10). As an example, we observed ZRfus1 binding at the proximal enhancer of Lmx1b, along with Brd4 and Ep300 recruitment and presence of three central ‘GGGCC’ motifs (Fig.4D). Lmx1b expression was elevated in ZRfus1-driven mouse tumors and human ependymoma, both associated with the presence of a proximal ZRfus1-specific super enhancer (Fig.4E). To confirm our findings in human tumor tissue, we mapped nucleosome-free regions of H3K27ac ChIP data in ZFTA-RELA vs non-ZFTA-RELA primary ependymoma samples (18). We found that PLAGL family TF motifs were the top-ranking DNA motif observed in a ZFTA-RELA specific context (Fig.4F, Supplemental Data Fig. S4H, S5A–E). Therefore, our findings demonstrate that ZRfus1 directly binds a tumor-specific program distinct from the canonical NF-κB pathway, and converges on activation of PLAGL TF target genes in both mouse and human ependymoma.
ZRfus1 regulates transcriptional regulatory circuits to drive super enhancer gene expression
We have previously shown that enhancers and super enhancers (SE) regulate subtype-specific gene expression programs in human ependymoma, including ZFTA-RELA tumors (18). To elucidate the mechanisms of oncogene activation in ZRfus1 ependymoma, we mapped super enhancers by Ranking of Super Enhancer (ROSE) analysis and identified several genes such as Dlk1, Lmx1b, and Ephb2 which also harbor SEs in human ZFTA-RELA ependymoma (Fig.5A, Supplementary Table S7) (18). Of note, 73% (511 of 703) of SEs identified harbored at least one overlapping ZRfus1 binding site, suggesting a significant contribution of ZRfus1 binding to SE driven gene expression (Fig.5B–C). As an example, we observed a 5’ SE predicted to regulate Dlk1, which contained ZRfus1 binding as well as Brd4 recruitment to sites of open chromatin (Fig.5D). Dlk1 was over-expressed in IUE:ZRfus1 tumors compared to normal brain, and had significantly higher expression in human ZFTA-RELA fusion driven ependymoma in comparison to other ependymoma subtypes. (Fig.5E). These findings are in line with single-cell RNA sequencing characterization of human ependymoma that pinpoints DLK1 as a top signature gene in a unique and tumor cell specific transcriptional program in ZRfus driven human tumors (24).
Figure 5. ZFTA-RELA disrupts core regulatory circuitry programs to drive SE gene expression.
(A) Hockey-stick plot depicting super enhancers detected in IUE: ZRfus1 murine ependymoma (B) Venn diagram showing overlap of HA: ZRfus1 peaks and SEs (C) Box-violin plot depicting up-regulation of SE associated genes in tumor vs. normal brain (D) HA-ZRfus1 CUT&RUN, ATAC-seq, Brd4 CUT&RUN, and H3K27ac CUT&RUN shown at the Dlk1 locus (E) Dlk1 expression in mouse and human ZRfus ependymoma compared to normal brain and other ependymoma subtypes, respectively (F) Schematic of identification of core regulatory circuit (CRC) transcription factors (G) Top CRC module enriched in IUE-ZRfus1 ependymoma including predicted binding sites and engagement of ZRfus1 at Plagl1/2 and Rela motifs (H) CUT&RUN localization of Plagl2, HA-ZRfus1 and Rela at super enhancers that harbor the Plagl1/2 or Rela motif.
To dissect the transcription factors that regulate ZRfus1 SEs, we conducted core regulatory circuitry analysis (CRC, Fig.5F). This method searches regions of open chromatin (demarcated by ATAC-seq) within H3K27ac-defined SEs (i.e. nucleosome-free regions). Core transcriptional circuits are defined by TFs that are regulated by SEs, which are transcribed and translated, and subsequently bind other SEs (25,26). CRC analysis identified the top-ranking set of eleven TFs that form a putative regulatory circuit including: Plagl2, Rela, Sox7, Sp2, Sox3, Foxo1, Srf, Smad1, Stat3, Zfp3, and Hic1 (Fig.5G, Rank = 1, Score = 284.091, Supplementary Table S8). To distinguish between ZRfus1 and Plagl2 binding at SEs and regulation of CRC transcriptional circuits, we performed CUT&RUN of Plagl2 (Fig.5H, Supplemental Data Fig. S5F). While ZRfus1 shared some binding sites with Plagl2, the occupancy at SEs, containing a Plagl2 motif or Rela motif, was largely a result of ZRfus1 binding. These findings support that ZRfus1 may act as an oncogenic TF by invading a CRC network, binding directly to Plagl1/2 and Rela motifs, and engaging SEs that regulate downstream ependymoma gene expression programs.
ZRfus1 tumor transcriptomes match radial glial cells that may give rise to ependymoma
Ependymomas are thought to arise from a heterogeneous population of radial glial cells during embryonic brain development (17,27). To investigate the developmental origins in our IUE:ZRfus1 model, we compared tumor transcriptomes against a single-cell RNA sequencing atlas of the mouse developing cortex (Supplemental Data Fig.S5G–I) (28). As compared to normal brain, we found that cells with the closest and most specific enrichment were embryonic day E15 dorsal and ventral radial glial cells (Supplemental Data Fig.S5G). Astrocytes of early post-natal stages showed varied degrees of enrichment. Plagl1 and Plagl2 gene expression was also found to be present and modestly elevated during this embryonic stage (Supplemental Data Fig.S5G). We observed more restricted patterns in the human developing brain cortex by querying the CoDEx database, and found Plagl1 expression to be enriched in outer radial glial cells (Supplemental Data Fig.S5J) (29). These findings support a possible hypothesis that ZRfus1 may bind, maintain, and/or activate Plagl target gene expression in developing radial glial cells that subsequently give rise to ependymoma.
ZRfus1 cistrome uncovers oncogenic pathways and molecular targets
The Plagl1 and Plagl2 proteins have diverse roles in both development (30) and tumorigenesis and have been shown across several cell types and tissues to regulate and express an imprinted gene network (including Axl, Dlk1, Igf2), Wnt pathway (Wnt and Fzd family genes) and ECM target genes (Integrin family) (30–33). Within ZRfus1 tumors, we detected direct binding enrichment of genes associated with NF-κB pathway activation such as TNF-α signaling, Epstein-Barr Virus Infection, Toxoplasmosis, and Osteoclast differentiation (Supplemental Data Fig. S6A–E). In the tumor-specific program, we identified genes elevated and associated with Wnt signaling pathway activity, Cushing syndrome, Hepatocellular carcinoma, focal adhesion, and members of the MAPK signaling pathway (Fig.6A). Many of the identified target genes identified contained previously unreported therapeutic leads in ZFTA-RELA ependymoma such as Cdk4/6, Gsk3B, Akt1, Mapkapk2/3, Fgfr4, Ngfr, Brd7, and Ep300. Further integration of ZRfus1 tumor-specific programs with the Washington University Drug Gene Interaction Database (34) revealed downstream candidate genes that are possibly clinically actionable (Akt1, Cdk4/6, Fgfr1/4), mapped in the ‘druggable’ genome space (Axl, Csf1r, Dlk1, Ephb2/4, Notch1), protein kinases (Cdk1/4/6, Igf2r, Ccnd1/3), proteases (Adamts enzymes, Stat3, Smad3), methyltransferases (Prdm1/16), and cell surface proteins (Acvrl1, Cd40/44/63, Pdgfrb) (Fig.6B). Our findings demonstrate that genes regulated by ZRfus1 represent a novel set of candidates relevant to ependymoma development which may represent possible therapeutic leads for future characterization.
Figure 6. Pathway enrichment of ZFTA-RELA specific and inflammatory-shared target genes.
(A) Tumor-specific programs enriched with Plagl TF motifs. Color of nodes highlight differential gene expression between mouse ZRfus1 and contralateral brain. (B) Integration of ZRfus1 bound and over-expressed genes with the Washington University Drug-Gene Interaction database to identify candidate drugs and small molecule inhibitors as potential therapeutic candidates against ependymoma.
DISCUSSION
Supratentorial ependymomas are often initiated by a single gene fusion (7,8,23,35). ZFTA-RELA is the most common alteration, however YAP1 and other gene fusions have been reported (See Zheng et al., co-submitted Cancer Discovery manuscript)(15). Together, ependymoma gene fusions have been shown to promote nuclear localization, and are thought to lead to promiscuous transcription factor activity, aberrant chromatin regulation, and upregulation of oncogenes. This activity may be tied to developmental programs that are essential for ependymoma cell growth (18). Our study demonstrates, in a natively forming brain tumor mouse model, that ZRfus1 directly engages accessible DNA and active chromatin. ZRfus1 binding is associated with expression of known ependymoma oncogenes, such as Ephb2, Notch1 and Ccnd1 and expression patterns are highly consistent with different mouse and human datasets of ZRfus driven tumors (See Kupp et al., and Zheng et al., Cancer Discovery Manuscript)(14,15). Our collective analysis across these different studies led to synthesis of a core set of 93 signature genes elevated in ZRfus driven mouse and human tumors.
Previous reports have suggested aberrant NF-κB activity as a central driver and mechanism of transformation in ZFTA-RELA ependymoma (7). This hypothesis has prompted clinical trial concepts to inhibit NF-κB signaling using known small molecules against the canonical NF-κB pathway. While our data reports a shared inflammatory program directly activated by ZRfus1, a major component of ZRfus1 binding is seen in a tumor-specific context and not observed in canonical TNF-α driven gene expression. This suggests that the ZFTA fragment of the ZRfus1 fusion protein confers oncogenic properties and is supported by several points: 1) ZFTA is fused with other genes in ependymoma (Reference to Zheng et al., co-submitted Cancer Discovery manuscript)(15), and 2) Hyperactive Rela is unable to drive tumorigenesis (8,12). On the other hand, the ZFTA fragment alone is unable to initiate tumors, therefore tying its function to its fusion partner, often a transcription factor and/or co-activator (7). Indeed, Kupp et al.,(14) have demonstrated a high similarity between ZFTA-Rela and ZFTA-Ep300 transcriptional programs, consistent with our data that reveals co-recruitment of transcriptional machinery including Brd4, Ep300, Cbp, and Pol2. These findings suggest that therapeutic strategies that disrupt Rela transcriptional activity may be more effective than drugs that target cytosolic regulation of Rela (i.e. IKK inhibitors). An alternate approach could be the disruption of transcriptional co-factors such as BRD4 and EP300/CBP that are recruited and associated with transcription of ZRfus1 bound genes. Indeed, ZFTA-RELA driven patient-derived xenograft (PDX) models show sensitivity to BET bromodomain inhibitors, such as JQ1, and IUE:ZRfus1 tumor cells to Ep300/Cbp inhibitors (18). It is not yet clear whether ZFTA-RELA tumors, in vivo, are more sensitive to perturbation of oncogene transcription (i.e. Transcriptional Addiction) to an extent where a therapeutic window can be achieved that spares effects on normal cells.
Our data points to tumor-specific DNA binding of ZRfus1 and convergence on the core binding sequence of Plagl family TFs. This group of TFs has been shown to be expressed in a variety of cancer types with diverse tumor suppressive and oncogenic roles and regulate an imprinted gene network along with Wnt pathway activation (30,36–41). Plagl2, specifically, has been reported to be amplified and over-expressed in glioblastoma and positively contributes to glioma-genesis through inhibition of cell differentiation (33). While our data does not exclude the functional role of Plagl1/2, it does suggest that ZRfus1 may be utilizing shared DNA binding sites for target gene activation. ZRfus1 directed transcriptional programs converge on upregulation of Plagl-associated Mapk, focal adhesion, imprinted and Wnt pathway genes. These Plagl genesets are enriched for putative molecular targets that are potentially clinically actionable, and hold promise for future lead development, such as druggable genome candidates, kinases, and cell surface proteins.
We hypothesize that Plagl1/2 programs may be active during embryonic brain development. Supporting this, we show that ZRfus1 tumor expression matches Plagl1/2 positive radial glial cells (RGCs) of the developing cortex, and consistent with data that RGCs contain cells of origin of ependymoma (27,42). In line with other pediatric brain tumors, we hypothesize that our data supports a model of stalled development where stem- or progenitor- cell gene expression programs are aberrantly sustained (28). Related to this, unclear is the role and expression patterns of ZFTA in normal development and whether its cis-regulatory elements facilitate cell and temporal specific expression of ZRfus1 expression during tumorigenesis. Indeed, several examples in other pediatric brain tumors exemplify how structural alterations not only activate oncogenes but also ‘hijack’ developmental enhancers to promote gene expression(43–45).
Our findings shed light on the transcriptional programs de-regulated by ZFTA-RELA and reveal potential downstream molecular targets that may hold important cellular vulnerabilities. As a single genetic driver, ‘drugging’ the ZFTA-RELA fusion specifically, as opposed to co-regulators and downstream targets would likely be the most effective. Solving the structure of ZFTA-RELA fusion with emerging protein crystallography approaches and identification of binding surfaces of the ZRfus protein could facilitate the design of proteolysis targeting chimeras for inhibition and/or protein degradation. However, a crucial question as to whether ZRfus is still required for tumor maintenance in vivo following transformation and establishment of epigenetic marks, remains unanswered. Lessons learned from studying ZFTA-RELA are likely to apply to many other fusion-driven pediatric and adult tumors in terms of developmental modeling, epigenetic characterization, and pre-clinical therapeutic testing.
METHODS
Western blot and Immunohistochemistry
Western blot analysis was performed using standard techniques. Antibodies used included Anti-HA tag antibody (Abcam catalogue no. ab9110, RRID:AB_307019), Lamin B1 (Abcam catalogue no. ab16048), β-Tubulin (CST 86298), β-Actin (Millipore catalogue no. A1978), BRD4 (Bethyl, Cat# A301–985100A), Crebbp (Cbp) (Cell Signaling Technologies, Cat#7389s) and EP300(Abcam, Cat# ab10485). Immunohistochemistry was performed on deparaffinized FFPE sections with antigen retrieval in sodium citrate (10mM pH6) at 95C for 15 min. Sections were blocked with 5% normal horse serum in PBS-T for 1h and primary antibodies added for an overnight incubation at 4C. IHC for the antibodies H3K27me3 (CST9733 1:250–1:500 dilution), H3K27ac (Abcam 1:250–1:500 dilution), and HA (Sigma – 11867431001 at 1:1000 dilution).
In Utero Electroporation (IUE) and tumor cell dissociation
All animal procedures in this study were performed with IACUC approval. In utero electroporation was performed as described previously, and plasmids provided as a gift from Dr. Joseph LoTurco PhD (13). After anesthesia with 5% isoflurane, pregnant mice, at E16.5, were subjected to abdominal incision to expose the uterus. DNA plasmid cocktail (pBCAG-HA-ZRFUS1, pbCAG-eGFP, pX330-sgTp53, GLAST-PBase, pBCAG-Luc) was injected into the lateral ventricles with a glass pipette. Electric pulses were then delivered to the embryos by gently clasping their heads with forceps-shaped electrodes. Six 33V pulses of 55 ms were applied at 100 ms intervals. The uterus was then repositioned into the abdominal cavity and the abdominal wall and the skin were then sutured. Following birth, pups were traced on a weekly basis by bioluminescent imaging to monitor brain tumor formation. Mouse tumors were collected based on isolation of GFP+ tumors and dissociation into single-cells using a Brain Tumor Dissociation kit (MACS, #130-095-942) following manufacturer’s instructions.
Cell Culture
Cells were grown in neural basal medium (Invitrogen) supplemented with sodium pyruvate, glutamine, B27, N2, bFGF (10 ng/mL) and rhEGF (20ng/mL). Cells were grown on treated cell culture dishes coated with Matrigel (Corning). All lines tested negative for mycoplasma contamination, as assessed using a PCR based approach. Tumor-derived cell lines were confirmed to be authentic and unique by STR fingerprinting.
Fractionation of nuclear and cytoplasmic extracts.
Mouse tumor cells were grown to full confluency and washed with ice-cold PBS and then collected with a cell scraper in ice-cold E1 buffer (50mM HEPES-KOH, 140 mM NaCl, 1 mM EDTA, 10% glycerol, 0.5% NP-40, 0.25% Triton X-100, 1 mM DTT supplemented with 1X protease inhibitor cocktail) for each volume of cell pellet. Resuspended cells were spun down at 1,100g at 4°C for 2 minutes and supernatant (cytoplasm fraction) was collected in a fresh tube. Cell pellet was resuspended in the same volume of the E1 buffer and spun down at 1,100 xg for 2 minutes. Supernatant was discarded and pellet was resuspended in the same volume of the E1 buffer and incubated on ice for 10 minutes before spinning down at 1,100 xg for 2 minutes at 4°C. Supernatant was discarded and pellet was gently resuspended in 2 volumes of ice-cold E2 buffer (10 mM Tris-HCl, 200 mM NaCl, 1mM EDTA, 0.5 mM EGTA, supplemented with 1x protease inhibitor cocktail) and centrifuged at 1,100 xg for 2 min at 4°C. The supernatant (nuclear fraction) was collected in a fresh tube.
CRISPR Knock-out (KO) line generation
sgRNA sequences targeting Rela and non-targeting control (D98) were designed using CRISPOR tool and the constructs were generated by plasmid cloning following GECKO protocol (RRID:SCR_015935). sgRNA sequences are provided in Supplementary Table S9. To produce lentiviral particles, HEK293T cells were seeded 24 hours prior to transfection and were transfected with 5 ug pMD2.G, 5 ug psPAX2 and 5 ug Lenti-CRISPR-v2 using calcium phosphate transfection method (RRID:Addgene_12260). Medium was refreshed 24 hours post-transfection. The media containing the viral particles were collected at 48 and 72 hours time-points and filtered with 0.45-μm filters. Mouse tumor cells were seeded into 10-cm plates 24hr prior to infection and were infected with freshly collected lenti-viruses with 8 μg/mL Polybrene for 24 hours. Infected cells were selected with 2 μg/mL puromycin for 3 days. Knock-out efficiencies were tested with western blot analysis.
RNA preparation for qRT-PCR and RNA-seq
RNA was extracted using TRIzol or Qiagen miRNA easy kit according to the manufacturer’s instructions and cDNA was prepared from ~1ug total RNA using iScript™ gDNA clear cDNA Synthesis Kit (Bio-Rad, #1725034). Quantitative PCR was performed using iTaq Universal SYBR® Green Supermix (Bio-Rad, #172–5120) using the manufacturer’s recommended protocol. Primers are listed in Supplementary Table S7. In the case of RNA-seq, ERCC spike-ins (Thermofisher) were added according to manufactures recommendations and protocols before proceeding with library preparation. RNA-seq library preps were performed using Kapa RNA Hyperprep kit with RiboErase (HMR) according to manufacturer’s recommendations.
CUT&RUN
CUT&RUN assay was performed as described in Skene and Henikoff. (2018)(16). Briefly, 0.5–1 million cells were captured with BioMagPlus Concanavalin A beads and incubated with primary antibody for 10–20 mins at room temperature. After washing away the EDTA in the buffer and unbound antibody with dig-wash buffer (20mM HEPES pH 7.5, 150mM NaCl, 0.5mM Spermidine, 1x Complete Protease Inhibitor EDTA-Free and 0.05% Digitonin), protein A-MNase was added and incubated for 10–20mins. The cells were washed again and placed in an ice-water pre-chilled metal block at least 5mins. CaCl2 was added to the final concentration of 2 mM to activate protein A-MNase for 30mins on the ice-water chilled metal block. The reaction was stopped by addition of equal volume of 2XSTOP buffer (340 mM NaCl, 20mM EDTA, 4mM EGTA, 0.02% Digitonin, 5μL/ml RNase A, 50μg/ml glycogen and 2 pg/ml heterologous spike-in DNA). The protein-DNA complex was released and DNA was extracted with Gel and PCR Clean-up kit (Macherey-Nagel NucleoSpin®, cat. no.740609.250) or Phenol-chloroform-isoamyl alcohol precipitation (for small fragment DNA), followed by Qubit fluorometer and Agilent 4200 Tapestation quality and size distribution control.
Library Preparation and Sequencing for CUT&RUN
KAPA Hyper prep kit (Cat# KK8504, KAPA Biosystems)and KAPA Dual-indexed adapter kit (Cat# KK8722 KAPA Biosystems) were used to construct the CUT&RUN DNA library for sequencing on an Illumina platform following the manufacturer’s instructions. After adaptor ligation, 2x volume of AMPure XP beads was used to recovery the small fragments. After 12–14 cycles of PCR amplification, the product was cleaned up with AMPure XP beads. 3% gel purification was performed if the PCR dimers were too much. Perform paired-end Illumina sequencing on the barcoded libraries following the manufacturer’s instructions. Briefly, the mixed libraries were denatured according to the standard protocol from Illumina. 1.3 mL of 1.8 pM diluted library pool was loaded to a NextSeq 500/550 Mid Output Kit v2 (150 cycles), and sequenced in the NextSeq 500 platform. Paired-end sequencing was then performed (2×75 bp, 8 bp index).
Cut & Run and ChIP data processing
Paired-end reads were adapter and quality trimmed using Trimgalore (v0.6.5, default parameters, http://www.bioinformatics.babraham.ac.uk/projects/trim_galore/) (RRID:SCR_011847) and aligned to mouse genome mm10 using Bowtie2 (v2.3.5.1, parameters: --local -D 20 -R 3 -N 0 -L 20 -i S,1,0.50 --no-unal --no-mixed --no-discordant --phred33 -I 10 -X 700) (RRID:SCR_016368). Duplicated reads were then marked and removed using picard MarkDuplicates (v2.21.1) (http://broadinstitute.github.io/picard/), and Samtools (v1.9) (RRID:SCR_002105), respectively with default parameters. For Rela and HA samples, only fragments of size less than 120bp were retained. Deeptools (v3.4.3) (RRID:SCR_016366) was used to convert all the resulting BAM files to Bigwig format for visualization. MACS2 (v2.2.7.1) was used to call peaks, on the resulting BAM files, with a p-value threshold of 1e-3. A set of 6845 peaks were inferred in the HA CUT&RUN by overlapping the called peaks from the two independent mice using bedtools2. This set was further filtered to remove any overlaps with non-specific IgG peak signals (from both mice), resulting in 5608 peaks. The peaks were then annotated to nearest genomic features using annotatePeaks.pl from Homer (v4.11.1)(46).
RNA sequencing and ATAC sequencing analysis
RNA-seq and ATAC-seq analysis was performed using Genialis Expression software (https://www.genialis.com) deployed locally on BCM computational infrastructure. Briefly, the RNA-seq pipeline run on the Genialis platform comprised the following steps. The raw reads were filtered to remove adapters and poor-quality reads using BBDuk (v37.9, https://sourceforge.net/projects/bbmap/). The resulting reads were mapped to the reference genomes (ENSEMBL 92) using STAR (v2.7.0) (RRID:SCR_015899). FeatureCounts (v1.6.3) (RRID:SCR_012919) was used for gene expression level quantification followed by DEseq2 (RRID:SCR_000154) for differential gene expression analysis (47). Lowly expression genes with expression count summed over all samples below 10 were filtered out from the input matrix to DESeq2. The paired-end reads from ATAC-seq were trimmed using BBDuk (v37.9) and mapped to the reference genome mm10 using Bowtie2 (v2.3.4.1). MACS2 (v2.1.1.20160309) was then used to call peaks on the aligned reads using p-value cutoff of 0.01 (parameters --shift -75 --extsize 150 --nomodel --call-summits --nolambda --keep-dup all -p 0.01)
DNA Motif analysis
To find motifs enriched around the binding sites, Homer findMotifsGenome.pl and MEME suite (v5.1.0) (48) were used. Further, we utilized the Rela ChIP -seq data in the GEO accession GSM3895240 (48) to differentiate the tumor-specific binding profile of ZRfus1 from the canonical inflammatory context. For this, the overlaps between the peaks in GSM3895240 and the 5608 HA CUT&RUN peaks were inferred using bedtools2. Homer motif analysis was performed on each of the peaks sets; unique to HA:ZRfus1, shared with and unique to the GSM3895240 Rela peaks. Enrichment of specific pathways in each of these groups was then computed using the R package Clusterprofiler (RRID:SCR_016884). To identify DNA motifs in human H3K27ac datasets, first peaks were identified in H3K27ac peaks then valleys (nucleosome-free regions) mapped which contain TF binding sites. Human ZFTA-RELA specific H3K27ac valley regions were compared against non-RELA ependymoma (as background) using HOMER (RRID:SCR_010881).
Super Enhancers and Core Regulatory Circuitry Analysis
Super enhancers were identified based on H3K27ac and IgG control CUT&RUN data using ROSE (49) with a stitching distance of 12.5 kb and exclusion of peaks within 2.5 kb of a promoter. Regions of H3K27ac overlapping ATAC peaks, along with the identified super enhancers, were provided as an input to determine core regulatory circuitry (https://github.com/linlabcode/CRC) (25).
Developmental Mapping Analysis
To project ZRfus1 tumor and normal brain transcriptomes to the scRNAseq atlas of the developing mouse forebrain(28), we evaluated enrichment of the gene signature of each atlas cell type in each sample, using single-sample sample gene set enrichment analysis (ssGSEA)(50). The most enriched cell type signatures in ZRfus1 tumors were selected for visualization by computing the median rank of each cell type signature across ZRfus1 tumor samples based on ssGSEA scores, and taking the top 15.
Supplementary Material
STATEMENT OF SIGNIFICANCE.
Ependymomas are aggressive brain tumors. While drivers of supratentorial ependymoma (ZFTA- and YAP1- associated gene fusions) have been discovered, their functions remains unclear. Our study investigates the biology of ZFTA-RELA driven ependymoma, specifically, mechanisms of transcriptional de-regulation and direct downstream gene networks that may be leveraged for potential therapeutic testing.
Funding Support and Acknowledgements:
SCM is supported by Cancer Prevention Research Institute of Texas (CPRIT) scholar award (RR170023), Alex’s Lemonade Stand Foundation (ALSF) A award, THINC Seed Grant, Helis Foundation Award, and NIH-National Institute of Neurological Disease and Stroke (R01NS116361). KCB is supported by grant funding from Hyundai Hope on Wheels, Rally Foundation, and St. Baldrick’s Foundation Early Career Development Award. JY is supported by a THINC Seed Grant, K12 award (5K12CA090433-17), and an Alex’s Lemonade Stand Foundation Center of Excellent Developmental Therapeutic Scholar Award. CK is supported by Canadian Institutes of Health Research (CIHR) #PJT-156086, and NSERC RGPIN-2016-04911. BD is supported by National Cancer Institute-Cancer Therapeutic Discovery (U01-CA217842 to BD) and Cancer Prevention Research Institute of Texas (RP150334 to BD). HCH is supported by grants from CPRIT (RR170036), NCI (R00CA187565), and NIGMS (R35GM137996).
Footnotes
Conflicts of Interest:
The authors declare no potential conflicts of interest
Data Accession
All raw and processed sequencing data from our study may be found at GEO: GSE161679.
REFERENCES
- 1.Merchant TE, Bendel AE, Sabin ND, Burger PC, Shaw DW, Chang E, et al. Conformal Radiation Therapy for Pediatric Ependymoma, Chemotherapy for Incompletely Resected Ependymoma, and Observation for Completely Resected, Supratentorial Ependymoma. J Clin Oncol 2019;37(12):974–83 doi 10.1200/jco.18.01765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pajtler KW, Wen J, Sill M, Lin T, Orisme W, Tang B, et al. Molecular heterogeneity and CXorf67 alterations in posterior fossa group A (PFA) ependymomas. Acta Neuropathol 2018;136(2):211–26 doi 10.1007/s00401-018-1877-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Pajtler KW, Witt H, Sill M, Jones DT, Hovestadt V, Kratochwil F, et al. Molecular Classification of Ependymal Tumors across All CNS Compartments, Histopathological Grades, and Age Groups. Cancer cell 2015;27(5):728–43 doi 10.1016/j.ccell.2015.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hoffman LM, Donson AM, Nakachi I, Griesinger AM, Birks DK, Amani V, et al. Molecular subgroup-specific immunophenotypic changes are associated with outcome in recurrent posterior fossa ependymoma. Acta Neuropathol 2014;127(5):731–45 doi 10.1007/s00401-013-1212-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bayliss J, Mukherjee P, Lu C, Jain SU, Chung C, Martinez D, et al. Lowered H3K27me3 and DNA hypomethylation define poorly prognostic pediatric posterior fossa ependymomas. Science translational medicine 2016;8(366):366ra161 doi 10.1126/scitranslmed.aah6904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wani K, Armstrong TS, Vera-Bolanos E, Raghunathan A, Ellison D, Gilbertson R, et al. A prognostic gene expression signature in infratentorial ependymoma. Acta Neuropathol 2012;123(5):727–38 doi 10.1007/s00401-012-0941-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Parker M, Mohankumar KM, Punchihewa C, Weinlich R, Dalton JD, Li Y, et al. C11orf95-RELA fusions drive oncogenic NF-kappaB signalling in ependymoma. Nature 2014;506(7489):451–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ozawa T, Arora S, Szulzewsky F, Juric-Sekhar G, Miyajima Y, Bolouri H, et al. A De Novo Mouse Model of C11orf95-RELA Fusion-Driven Ependymoma Identifies Driver Functions in Addition to NF-kappaB. Cell Rep 2018;23(13):3787–97 doi 10.1016/j.celrep.2018.04.099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hoesel B, Schmid JA. The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer 2013;12:86 doi 10.1186/1476-4598-12-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Brown JD, Lin CY, Duan Q, Griffin G, Federation A, Paranal RM, et al. NF-κB directs dynamic super enhancer formation in inflammation and atherogenesis. Mol Cell 2014;56(2):219–31 doi 10.1016/j.molcel.2014.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Parker SC, Stitzel ML, Taylor DL, Orozco JM, Erdos MR, Akiyama JA, et al. Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants. Proceedings of the National Academy of Sciences of the United States of America 2013;110(44):17921–6 doi 10.1073/pnas.1317023110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Huang D, Sumegi J, Dal Cin P, Reith JD, Yasuda T, Nelson M, et al. C11orf95-MKL2 is the resulting fusion oncogene of t(11;16)(q13;p13) in chondroid lipoma. Genes Chromosomes Cancer 2010;49(9):810–8 doi 10.1002/gcc.20788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yu K, Lin CJ, Hatcher A, Lozzi B, Kong K, Huang-Hobbs E, et al. PIK3CA variants selectively initiate brain hyperactivity during gliomagenesis. Nature 2020;578(7793):166–71 doi 10.1038/s41586-020-1952-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Robert Kupp LR, Terranova Sabrina, Nathan Erica, Ballereau Stephane, Stark Rory, Chilamakuri Chandra Sekhar Reddy, Hoffmann Nadin, Wickham-Rahrmann Katherine, Widdess Marcus, Arabzade Amir, Zhao Yanhua, Varadharajan Srinidhi, Zheng Tuyu, Murugesan Mohankumar K., Pfister Stefan, Kawauchi Daisuke, Pajtler Kristian W., Deneen Benjamin, Mack Stephen, Masih Katherine, Gryder Berkley, Khan Javed, and Gilbertson Richard J.,. C11orf95 translocations constitute ependymoma chromatin remodeling and transcription factors. Cancer Discovery 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tuyu Zheng DRG, Okonechnikov Konstantin, Korshunov Andrey, Sill Martin, Maass Kendra K., Benites Patricia, da Silva Goncalves, Ryzhova Marina, Gojo Johannes, Stichel Damian, Arabzade Amir, Kupp Robert, Benzel Julia, Taya Shinichiro, Adachi Toma, Shiraishi Ryo, Gerber Nicolas U., Sturm Dominik, Ecker Jonas, Sievers Philipp, Selt Florian, Chapman Rebecca, Haberler Christine, Figarella-Branger Dominique, Reifenberger Guido, Fleischhack Gudrun, Rutkowski Stefan, Donson Andrew M., Ramaswamy Vijay, Capper David, Ellison David W., Herold-Mende Christel, Schüller Ulrich, Brandner Sebastian, Driever Pablo Hernáiz, Kros Johan M., Snuderl Matija, Milde Till, Grundy Richard G., Hoshino Mikio, Mack Stephen C., Gilbertson Richard J., Jones David T.W., Kool Marcel, von Deimling Andreas, Pfister Stefan M., Sahm Felix, Kawauchi Daisuke, Pajtler Kristian W.,. Cross-species genomics reveals oncogenic dependencies in ZFTA/C11orf95 fusionpositive supratentorial ependymomas. Cancer Discovery 2021. [DOI] [PubMed] [Google Scholar]
- 16.Skene PJ, Henikoff JG, Henikoff S. Targeted in situ genome-wide profiling with high efficiency for low cell numbers. Nat Protoc 2018;13(5):1006–19 doi 10.1038/nprot.2018.015. [DOI] [PubMed] [Google Scholar]
- 17.Johnson RA, Wright KD, Poppleton H, Mohankumar KM, Finkelstein D, Pounds SB, et al. Cross-species genomics matches driver mutations and cell compartments to model ependymoma. Nature 2010;466(7306):632–6 doi 10.1038/nature09173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mack SC, Pajtler KW, Chavez L, Okonechnikov K, Bertrand KC, Wang X, et al. Therapeutic targeting of ependymoma as informed by oncogenic enhancer profiling. Nature 2018;553(7686):101–5 doi 10.1038/nature25169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kapatos G, Hirayama K, Shimoji M, Milstien S. GTP cyclohydrolase I feedback regulatory protein is expressed in serotonin neurons and regulates tetrahydrobiopterin biosynthesis. Journal of neurochemistry 1999;72(2):669–75 doi 10.1046/j.1471-4159.1999.0720669.x. [DOI] [PubMed] [Google Scholar]
- 20.de Lombares C, Heude E, Alfama G, Fontaine A, Hassouna R, Vernochet C, et al. Dlx5 and Dlx6 expression in GABAergic neurons controls behavior, metabolism, healthy aging and lifespan. Aging 2019;11(17):6638–56 doi 10.18632/aging.102141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lasko LM, Jakob CG, Edalji RP, Qiu W, Montgomery D, Digiammarino EL, et al. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature 2017;550(7674):128–32 doi 10.1038/nature24028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ngo KA, Kishimoto K, Davis-Turak J, Pimplaskar A, Cheng Z, Spreafico R, et al. Dissecting the Regulatory Strategies of NF-κB RelA Target Genes in the Inflammatory Response Reveals Differential Transactivation Logics. Cell Rep 2020;30(8):2758–75.e6 doi 10.1016/j.celrep.2020.01.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Pajtler KW, Wei Y, Okonechnikov K, Silva PBG, Vouri M, Zhang L, et al. YAP1 subgroup supratentorial ependymoma requires TEAD and nuclear factor I-mediated transcriptional programmes for tumorigenesis. Nat Commun 2019;10(1):3914 doi 10.1038/s41467-019-11884-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gojo J, Englinger B, Jiang L, Hübner JM, Shaw ML, Hack OA, et al. Single-Cell RNA-Seq Reveals Cellular Hierarchies and Impaired Developmental Trajectories in Pediatric Ependymoma. Cancer cell 2020;38(1):44–59.e9 doi 10.1016/j.ccell.2020.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Saint-Andre V, Federation AJ, Lin CY, Abraham BJ, Reddy J, Lee TI, et al. Models of human core transcriptional regulatory circuitries. Genome Res 2016;26(3):385–96 doi 10.1101/gr.197590.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mack SC, Singh I, Wang X, Hirsch R, Wu Q, Villagomez R, et al. Chromatin landscapes reveal developmentally encoded transcriptional states that define human glioblastoma. J Exp Med 2019;216(5):1071–90 doi 10.1084/jem.20190196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Taylor MD, Poppleton H, Fuller C, Su X, Liu Y, Jensen P, et al. Radial glia cells are candidate stem cells of ependymoma. Cancer cell 2005;8(4):323–35 doi 10.1016/j.ccr.2005.09.001. [DOI] [PubMed] [Google Scholar]
- 28.Jessa S, Blanchet-Cohen A, Krug B, Vladoiu M, Coutelier M, Faury D, et al. Stalled developmental programs at the root of pediatric brain tumors. Nat Genet 2019;51(12):1702–13 doi 10.1038/s41588-019-0531-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Polioudakis D, de la Torre-Ubieta L, Langerman J, Elkins AG, Shi X, Stein JL, et al. A Single-Cell Transcriptomic Atlas of Human Neocortical Development during Mid-gestation. Neuron 2019;103(5):785–801.e8 doi 10.1016/j.neuron.2019.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Adnani L, Dixit R, Chen X, Balakrishnan A, Modi H, Touahri Y, et al. Plag1 and Plagl2 have overlapping and distinct functions in telencephalic development. Biol Open 2018;7(11) doi 10.1242/bio.038661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Varrault A, Dantec C, Le Digarcher A, Chotard L, Bilanges B, Parrinello H, et al. Identification of Plagl1/Zac1 binding sites and target genes establishes its role in the regulation of extracellular matrix genes and the imprinted gene network. Nucleic Acids Res 2017;45(18):10466–80 doi 10.1093/nar/gkx672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Varrault A, Gueydan C, Delalbre A, Bellmann A, Houssami S, Aknin C, et al. Zac1 Regulates an Imprinted Gene Network Critically Involved in the Control of Embryonic Growth. Developmental Cell 2006;11(5):711–22 doi 10.1016/j.devcel.2006.09.003. [DOI] [PubMed] [Google Scholar]
- 33.Zheng H, Ying H, Wiedemeyer R, Yan H, Quayle SN, Ivanova EV, et al. PLAGL2 regulates Wnt signaling to impede differentiation in neural stem cells and gliomas. Cancer cell 2010;17(5):497–509 doi 10.1016/j.ccr.2010.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cotto KC, Wagner AH, Feng YY, Kiwala S, Coffman AC, Spies G, et al. DGIdb 3.0: a redesign and expansion of the drug-gene interaction database. Nucleic Acids Res 2018;46(D1):D1068–d73 doi 10.1093/nar/gkx1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Eder N, Roncaroli F, Dolmart MC, Horswell S, Andreiuolo F, Flynn HR, et al. YAP1/TAZ drives ependymoma-like tumour formation in mice. Nat Commun 2020;11(1):2380 doi 10.1038/s41467-020-16167-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li N, Li D, Du Y, Su C, Yang C, Lin C, et al. Overexpressed PLAGL2 transcriptionally activates Wnt6 and promotes cancer development in colorectal cancer. Oncol Rep 2019;41(2):875–84 doi 10.3892/or.2018.6914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Majem B, Parrilla A, Jiménez C, Suárez-Cabrera L, Barber M, Marín A, et al. MicroRNA-654–5p suppresses ovarian cancer development impacting on MYC, WNT and AKT pathways. Oncogene 2019;38(32):6035–50 doi 10.1038/s41388-019-0860-0. [DOI] [PubMed] [Google Scholar]
- 38.Sekiya R, Maeda M, Yuan H, Asano E, Hyodo T, Hasegawa H, et al. PLAGL2 regulates actin cytoskeletal architecture and cell migration. Carcinogenesis 2014;35(9):1993–2001 doi 10.1093/carcin/bgu081. [DOI] [PubMed] [Google Scholar]
- 39.Wu L, Yuan W, Chen J, Zhou Z, Shu Y, Ji J, et al. Increased miR-214 expression suppresses cell migration and proliferation in Hirschsprung disease by interacting with PLAGL2. Pediatr Res 2019;86(4):460–70 doi 10.1038/s41390-019-0324-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wu L, Zhou Z, Han S, Chen J, Liu Z, Zhang X, et al. PLAGL2 promotes epithelial-mesenchymal transition and mediates colorectal cancer metastasis via β-catenin-dependent regulation of ZEB1. Br J Cancer 2020;122(4):578–89 doi 10.1038/s41416-019-0679-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhou J, Liu H, Zhang L, Liu X, Zhang C, Wang Y, et al. DJ-1 promotes colorectal cancer progression through activating PLAGL2/Wnt/BMP4 axis. Cell Death Dis 2018;9(9):865 doi 10.1038/s41419-018-0883-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Vladoiu MC, El-Hamamy I, Donovan LK, Farooq H, Holgado BL, Sundaravadanam Y, et al. Childhood cerebellar tumours mirror conserved fetal transcriptional programs. Nature 2019;572(7767):67–73 doi 10.1038/s41586-019-1158-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Morton AR, Dogan-Artun N, Faber ZJ, MacLeod G, Bartels CF, Piazza MS, et al. Functional Enhancers Shape Extrachromosomal Oncogene Amplifications. Cell 2019;179(6):1330–41.e13 doi 10.1016/j.cell.2019.10.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sin-Chan P, Mumal I, Suwal T, Ho B, Fan X, Singh I, et al. A C19MC-LIN28A-MYCN Oncogenic Circuit Driven by Hijacked Super-enhancers Is a Distinct Therapeutic Vulnerability in ETMRs: A Lethal Brain Tumor. Cancer cell 2019;36(1):51–67.e7 doi 10.1016/j.ccell.2019.06.002. [DOI] [PubMed] [Google Scholar]
- 45.Northcott PA, Lee C, Zichner T, Stütz AM, Erkek S, Kawauchi D, et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 2014;511(7510):428–34 doi 10.1038/nature13379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 2010;38(4):576–89 doi 10.1016/j.molcel.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15(12):550 doi 10.1186/s13059-014-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bailey TL, Boden M, Buske FA, Frith M, Grant CE, Clementi L, et al. MEME SUITE: tools for motif discovery and searching. Nucleic Acids Res 2009;37(Web Server issue):W202–8 doi 10.1093/nar/gkp335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 2013;153(2):320–34 doi 10.1016/j.cell.2013.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Barbie DA, Tamayo P, Boehm JS, Kim SY, Moody SE, Dunn IF, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 2009;462(7269):108–12 doi 10.1038/nature08460. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.