PHF19 is overexpressed and strongly associated with an unfavorable prognosis in t(8;21) AML
We previously conducted a comprehensive analysis of m6A distribution and gene expression profiling in both parental Kasumi-1 cells and HIF1α-manipulated Kasumi-1 cells using m6A-modified RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) (GSE168778, GSE200605) [11]. By comparing differentially expressed genes with transcripts containing m6A sites identified by MeRIP-seq in both cell types, we identified 14 significantly differentially methylated genes that are positively correlated with HIF1α expression [11]. To further refine our candidate gene selection, mRNA expression profiles from two independent datasets underwent bioinformatics analysis (details provided below). Our analysis revealed that PHF19 exhibited the highest expression in the t(8;21) subtype of AML, and high PHF19 expression was found to have prognostic value in t(8;21) AML. Consequently, our subsequent research focused on the PHF19 gene.
We initially conducted bioinformatic analysis using data from the BeatAML database (http://www.vizome.org/aml) to investigate the expression pattern of PHF19 in AML and elucidate the relevance of PHF19 and WTAP in the t(8;21) AML subgroup. Our findings revealed a significant upregulation of PHF19 expression levels in t(8;21) AML patients compared to non-t(8;21) AML patients or healthy donors (Fig. 1A, Supplementary Fig. 1A). To validate these findings, the expression of PHF19 was examined in the GEO dataset (GSE6891) [23], revealing significantly elevated levels of PHF19 in t(8;21) AML (Fig. 1B). The prognostic implications of high PHF19 expression were further confirmed in a cohort of AML patients from the TARGET database. Notably, PHF19 expression showed a significant association with t(8;21) AML (Fig. 1C). Subsequently, t(8;21) AML patients were stratified into two groups based on their PHF19 expression levels (Fig. 1D). There were no significant differences observed in baseline clinical features between the PHF19high and PHF19low groups (Supplementary Table 1). While overall survival (OS) did not exhibit significant differences between the groups (Fig. 1E), it was evident that the high PHF19 expression group displayed markedly poorer event-free survival (EFS) compared to the low-expression group (Fig. 1F; p = 0.034). With a median follow-up period of 53.1 months, the estimated four-year EFS rates were found to be 57.7% (95% CI, 44.6%-68.7%) and 77.1% (95% CI, 60.6%-87.4%), respectively (Supplementary Table 2; p = 0.034). Furthermore, multivariate analysis indicated that higher expression of PHF19 was an independent risk factor for poorer EFS with marginal significance in t(8;21) AML (Supplementary Table 3; Hazard Risk = 2.04, p = 0.066). To validate the clinical significance and relevance of PHF19 and WTAP in t(8;21) AML, a separate cohort of newly diagnosed patients with t(8;21) AML from the GEO database (GSE16432) [24] were categorized into 4 groups based on their levels of WTAP and PHF19 expression (Fig. 1G). Kaplan-Meier analysis demonstrated that patients in the WTAPhigh/PHF19high group exhibited poorer OS outcomes compared to those in the WTAPlow/PHF19low group (Fig. 1H). Furthermore, the expression of PHF19 showed positive correlation with HIF1α and WTAP expression respectively in t(8;21) AML, indicating an association between HIF1α-WTAP axis and PHF19 (Supplementary Fig. 1B,C, Fig. 1I). Overall, high expression of PHF19 was observed in t(8;21) AML, and its co-overexpression with WTAP was associated with poorer clinical outcomes, suggesting a potential oncogenic role for PHF19 in t(8;21) AML.
PHF19 knockdown inhibits leukemia cell proliferation in both in vitro and in vivo settings
We further investigated the functional roles of PHF19 in t(8;21) AML. To assess the impact of PHF19 on leukemia cell behavior, we generated t(8;21) AML cell lines Kasumi-1 and SKNO-1 with stable knockdown of PHF19 expression using lentivirus-mediated short hairpin RNA (shRNA). The knockdown efficiency was observed at both mRNA and protein levels (Fig. 2A, Supplementary Fig. 2A). Downregulation of PHF19 significantly decreased the number and size of colonies compared to control (Fig. 2B), indicating a reduction in proliferation ability of leukemia cells in vitro. Furthermore, we evaluated the effects of PHF19 knockdown on the differentiation of t(8;21) AML cells by monitoring definitive markers of differentiated cells (CD11b and CD15) using flow cytometry. We found that PHF19 knockdown increased the proportion of cells with myeloid differentiation-specific markers (Fig. 2C). Additionally, we investigated the effect of PHF19 knockdown on apoptosis using flow cytometry in SKNO-1 and Kasumi-1 cell lines, which revealed an increase in apoptotic cells following PHF19 knockdown (Fig. 2D). To determine the functional role of PHF19 on t(8;21) AML cell proliferation in vivo, nude mice were injected with control Kasumi-1 cells and PHF19 knockdown cells to construct a subcutaneous tumor model, and tumor growth was monitored. Our results showed that PHF19 knockdown significantly inhibited tumor formation, resulting in lower tumor volume in nude mice (Fig. 2E), consistent with our in vitro findings. Overall, these data provide supporting evidence for the oncogenic role of PHF19 in t(8;21) AML, motivating us to explore the molecular regulatory basis underlying high expression levels of PHF19.
PHF19 is a potential downstream target of the HIF1α-WTAP axis
Based on our previously reported transcriptome-wide m6A distribution feature and gene expression profiling in parental Kasumi-1 cells, as well as HIF1α-manipulated Kasumi-1 cells using MeRIP-seq and RNA-seq (GSE168778, GSE200605) [11], we identified PHF19 as a significantly differentially methylated gene positively associated with HIF1α expression. Our analysis revealed the presence of m6A peaks in the 3'-untranslated region (3'-UTR) of the PHF19 gene. Furthermore, we confirmed that CoCl2-induced accumulation of HIF1α protein led to an increase in the m6A level of the PHF19 gene, while echinomycin-induced HIF1α inhibition reversed this effect (Fig. 3A). Additionally, it has been previously documented that HIF1α promotes the transcription of WTAP by directly binding to hypoxia response elements within the promoter region of WTAP [11]. To elucidate the molecular mechanism by which the HIF1α-WTAP axis regulates PHF19 expression, t(8;21) AML cell lines Kasumi-1 and SKNO-1 were treated with CoCL2 or CoCL2 + echinomycin. The activation of the HIF1α-WTAP axis induced by CoCL2 resulted in a significant increase in PHF19 protein levels, while inhibition of the HIF1α-WTAP axis by echinomycin reversed the effect of CoCL2 in both cell lines (Fig. 3B). This suggests that PHF19 is positively regulated by the HIF1α-WTAP axis in t(8;21) AML. Consistent with this finding, ectopic expression of HIF1α increased both PHF19 mRNA and protein levels in t(8;21) AML cell lines (Fig. 3C, Supplementary Fig. 3A). The specific regulation of PHF19 by the HIF1α-WTAP axis was further confirmed by findings showing that PHF19 mRNA and protein levels decreased upon knockdown of HIF1α using lentivirus-mediated shRNA transfection in Kasumi-1 and SKNO-1 cells (Fig. 3D, Supplementary Fig. 3B). Taken together, these results indicate that PHF19 may be a downstream target of the HIF1α-WTAP axis.
WTAP-mediated m6A modification increases PHF19 mRNA stability
The strong positive correlation between WTAP and PHF19 expression levels prompted us to further investigate whether WTAP enhances the mRNA m6A modification and expression levels of PHF19 transcripts. To this end, we conducted MeRIP-qPCR, Western blot, and real-time PCR assays to elucidate the changes in m6A modification, protein, and mRNA levels of PHF19 in control leukemia cells and WTAP knockdown cells. As anticipated, our MeRIP-qPCR analyses revealed a significant decrease in the m6A level at the 3’-UTR of PHF19 mRNA transcript in WTAP-knockdown Kasumi-1 cells (Fig. 4A). Consistent with these findings, both the expression of PHF19 mRNA and protein levels were markedly reduced upon targeted knockdown of WTAP by shRNA in Kasumi-1 and SKNO-1 cells (Fig. 4B, Supplementary Fig. 4A). Furthermore, to rule out any potential off-target effects of shRNA, we performed a rescue experiment to confirm the role of WTAP in regulating PHF19 expression. We generated stable cell lines overexpressing WTAP using lentiviruses in both Kasumi-1 and SKNO-1 cell lines. Notably, ectopic overexpression of WTAP significantly elevated PHF19 levels in both cell lines; conversely, co-transfection with WTAP shRNA targeting the 3’-UTR of the gene resulted in reduced PHF19 levels (Fig. 4C). In summary, these results strongly suggest that WTAP plays a crucial role in modulating m6A modification and influencing the expression of PHF19 mRNA.
To investigate the impact of m6A modification on the stability of PHF19 mRNA transcripts, we quantified the changes in PHF19 mRNA decay rates following WTAP knockdown. This was achieved by measuring mRNA abundance after transcriptional inhibition with actinomycin D treatment in both control leukemia cells and WTAP knockdown cells. Our findings revealed that PHF19 mRNA displayed increased instability in WTAP knockdown cells compared to control leukemia cells (Fig. 4D). Collectively, these data suggest that the upregulation of PHF19 levels induced by WTAP is at least partially attributed to the enhanced stability of PHF19 mRNA transcripts.
The silencing of PHF19 leads to a decrease in H3K27me3 levels
PHF19 plays a crucial role in catalyzing histone H3K27 trimethylation (H3K27me3), a repressive chromatin marker associated with epigenetic gene silencing [16]. Therefore, we aimed to investigate the impact of PHF19 on H3K27me3 levels in t(8;21) AML. The specific modulation of H3K27me3 by PHF19 was confirmed by our findings that stable knockdown of PHF19 in Kasumi-1 and SKNO-1 cells led to decreased levels of H3K27me3 (Fig. 5A). Additionally, we conducted a rescue experiment to demonstrate the role of PHF19 in regulating H3K27me3. We first established stable overexpression cell lines for PHF19 in Kasumi-1 and SKNO-1 using lentiviruses. Importantly, increased expression levels of PHF19 significantly elevated H3K27me3 levels in both cell lines, while co-transfection of PHF19 shRNA targeting the 3’-UTR of PHF19 resulted in reduced H3K27me3 levels (Fig. 5B). Consistent with these results, transfection of WTAP shRNA into Kasumi-1 and SKNO-1 cells suppressed both PHF19 and H3K27me3 levels compared to their respective controls (Fig. 5C), indicating that WTAP knockdown leads to downregulation of PHF19 and consequently a global loss of H3K27me3.
Given the evidence that the AML1/ETO-HIF1α axis promotes WTAP expression in t(8;21) AML cells [6, 11], we conducted further investigations to determine whether AML1/ETO positively regulates PHF19 expression and, consequently, H3K27me3 levels. As anticipated, overexpression of AML1/ETO led to significant increases in HIF1α, WTAP, and PHF19 levels. This was accompanied by a concurrent rise in global H3K27me3 levels in ZnSO4-treated U937-A/E cells compared to both U937-MT cells and untreated U937-A/E cells (Supplementary Fig. 5A). These findings collectively support the notion that AML1/ETO may impact the overall level of H3K27me3 through the activation of the HIF1α-WTAP-PHF19 axis.
PHF19 regulates cell cycle and DNA damage response by modifying chromatin accessibility
Given the potential impact of altered genomic H3K27me3 levels on chromatin organization following PHF19 knockdown, we conducted chromatin accessibility analysis using assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq) in control Kasumi-1 cells and PHF19 knockdown cells (Fig. 6A, Supplementary Fig. 6A). Surprisingly, our findings indicated that PHF19 knockdown led to an increase in chromatin accessibility compared to the control (Fig. 6B, Supplementary Table 7,8). Gene ontology (GO) term enrichment analysis of the genes associated with the gained peaks revealed a significant enrichment of cell cycle and DNA damage related terms in the PHF19 knockdown cells, including DNA double-strand break repair and mitotic cell cycle phase transition (Fig. 6B,C, Supplementary Table 9). Furthermore, gene set enrichment analysis (GSEA) using reactome pathways confirmed that pathways related to G1/S DNA damage checkpoints, DNA repair, cell cycle checkpoints, nucleotide excision repair, and global-genome nucleotide excision were enriched in PHF19 knockdown cells (Fig. 6D; Supplementary Table 10). Visualization of ATAC-seq tracks also demonstrated increased chromatin opening at key genes associated with DNA damage checkpoints and cell cycle phase transition following PHF19 knockdown (Supplementary Fig. 6B). These results suggest that expression of genes involved in cell cycle regulation and DNA repair may be altered upon PHF19 knockdown.
To further validate this observation, we conducted differential gene expression analysis using RNA-seq in control Kasumi-1 cells and PHF19 knockdown cells. We found that 3,043 genes were upregulated upon PHF19 knockdown (Fig. 6E, Supplementary Table 11). Additionally, the differential expression analysis of RNA-seq data exhibits many similarities with the differential analysis of ATAC-seq data. The GSEA enrichment analysis of differentially expressed genes from RNA-seq data indicated a positive correlation between PHF19 knockdown cells and gene sets related to G1/S DNA damage checkpoints, cell cycle checkpoints, and DNA repair (Fig. 6F, Supplementary Fig. 6C, Supplementary Table 12).
To investigate the role of PHF19 in regulating cell cycle progression in t(8;21) AML cells, we conducted flow cytometry analysis to examine the cell cycle distribution in PHF19 knockdown cells and control cells. Our findings were consistent with the results of cell colony-forming assays, indicating that knockdown of PHF19 led to cell cycle arrest at G1/S phase (Fig. 7A). These observations suggest that PHF19 promotes leukemia cell growth by facilitating the bypassing of G1 phase arrest through the induction of cell cycle and DNA damage checkpoints inactivation.
Targeting histone methyltransferase PRC2 inhibits t(8;21) AML cell growth
Given the pivotal role of PHF19 in the WTAP-m6A-PHF19 cascade, PHF19 may represent a promising new molecular target for t(8;21) AML. However, due to the current lack of suitable inhibitors against PHF19, we conducted an investigation into the cytotoxicity of polycomb repressive complex 2 (PRC2) histone methyltransferase (HMT) inhibitors as a potential therapeutic agent in t(8;21) AML. Specifically, we treated the Kasumi-1 and SKNO-1 cell lines with 9 selective HMT inhibitors respectively and assessed cell viability using the CCK-8 method. Our results demonstrated that these HMT inhibitors induced varying degrees of decrease in leukemia cell counts (Supplementary Fig. 7A), indicating a favorable responsiveness of both cell lines to the majority of tested compounds. Therefore, targeting HMT presents itself as a promising strategy for t(8;21) AML treatment.