Skip to main content

Transcriptomic characterisation of acute myeloid leukemia cell lines bearing the same t(9;11) driver mutation reveals different molecular signatures

Abstract

Background

Acute myeloid leukemia (AML) is the most common type of acute leukemia, accounting for 20% of cases in children and adolescents. Genome-wide studies have identified genes that are commonly mutated in AML, including many epigenetic regulators involved in either DNA methylation (DNMT3A, TET2, IDH1/2) or histone post-translational modifications (ASXL1, EZH2, MLL1). Several cell lines derived from AML patients are widely used in cancer research. Whether important differences in these cell lines exist remains poorly characterised.

Results

Here, we used RNA sequencing (RNA-Seq) to contrast the transcriptome of four commonly used AML-derived cell lines: THP-1, NOMO-1, MOLM-13 bearing the common initiating t(9;11) translocation, and MV4.11 bearing the t(4;11) translocation. Gene set enrichment analyses and comparison of key transcription and epigenetic regulator genes revealed important differences in the transcriptome, distinguishing these AML models. Among these, we found striking differences in the expression of clusters of genes located on chromosome 19 encoding Zinc Finger (ZNF) transcriptional repressors. Low expression of many ZNF genes within these clusters is associated with poor survival in AML patients.

Conclusion

The present study offers a valuable resource by providing a detailed comparative characterisation of the transcriptome of cell lines within the same AML subtype used as models for leukemia research.

Peer Review reports

Introduction

Leukemia results from the accumulation of mutations in oncogenes (gain-of-function) and/or tumour suppressor genes (TSG) (loss-of-function), giving rise to an imbalance between proliferation/self-renewal and differentiation in the hematopoietic stem and progenitor cell (HSPC) pool [1]. In acute myeloid leukemia (AML), initiating mutations (such as the commonly occurring AF9-MLL translocation) dysregulate stem cell fate decisions in HSPCs, generating pre-leukemic stem cells (pre-LSCs) [2]. Under permissive conditions, pre-LSCs acquire cooperating mutations which generate leukemic stem cells (LSCs) that drive leukemogenesis and are difficult to eradicate using currently available treatments [3, 4].

The primary and secondary genetic targets that drive the emergence of LSC and eventually leukemogenesis can be broadly classified as either conferring proliferative and survival advantages (e.g. FLT3, KIT, RASs), altering differentiation and apoptosis (e.g. PML-RARA, RUNX1, MLL1), and a third important category of mutations which leads to an alteration of cellular epigenetic state [5, 6]. Many of the mutations in this last group (including DNMT3, TET2 and IDH1/2) affect the DNA methylation machinery, disrupting normal DNA methylation in LSC. Other mutations (including ASXL1, EZH2 and MLL1) affect proteins involved in post-translational modifications of histones, which are important for transcriptional regulation, DNA repair, regulation of the cell cycle and differentiation [7]. Epigenetic pathways are of particular interest from a therapeutic perspective, due to the inherent plasticity of epigenetic modification patterns and because many epigenetic regulators can be targeted by drugs [8,9,10,11], some of which are currently approved for clinical applications [12].

The choice of cell line to be used as an experimental model to study leukemia in the laboratory is a complex multi-factorial decision, which has important ramifications for the results observed and their interpretation. The most important considerations include the disease subtype represented by a cell line (most frequently indicated by the French-American-British (FAB) classification system) as well as the different driver genetic events (chromosomal translocations or mutations) [13]. However, immortalised cells also bear multiple secondary genetic mutations that exert widespread quantitative and qualitative alterations on the molecular characteristics of the cell, including the transcriptome. The influence of this complexity on cellular phenotype is frequently overlooked in the interpretation of experimental findings.

Fundamental research on AML is often performed in one of several well-established cell line models. However, many of these commonly used cell lines remain incompletely characterised, and, in particular, direct molecular comparisons of these different models are lacking. Here, we have used RNA sequencing (RNA-Seq) to compare the transcriptome of three commonly used AML cell lines bearing a common driver event, the t(9;11) translocation (THP-1, NOMO-1, MOLM-13) together with another AML cell line (MV4.11) bearing a different translocation, t(4:11). Pairwise comparisons between all cell lines revealed that pathways related to the regulation of gene transcription were among the most significantly different between the cell lines. More specifically, we found alteration of the p53 pathway, the HOX gene clusters, key hematopoietic transcription factors and epigenetic regulators, all relevant to AML. We also observed striking differences in the expression of genes encoding C2H2 Zinc Finger (ZNF) transcriptional repressors clustered on chromosome 19 (Chr19) which have prognostic significance in AML patients. This study provides a transcriptomic resource together with additional insights that will improve the choice of AML cell lines as models for pre-clinical research such as drug screens.

Results and discussion

Transcriptomic characterisation of AML cell line models

All AML cell lines used in this study belong to the same subtype (M5) classification of the FAB system [13]. THP-1, MOLM-13 and NOMO-1 are characterised by a t(9;11) translocation (p22;q23 involving MLLT3-KMT2A fusion genes) [14, 15] common in AML, whereas MV4.11 has a t(4;11) translocation (q21;q23 involving AFF1-KMT2A fusion genes) [16], which is more common in acute lymphoblastic leukemia (ALL) [17], but with myeloid features [18, 19]. Induction of t(9;11) or t(4;11) is sufficient to promote myeloid [20] and lymphoblastic [21] leukemia respectively. Of the three t(9;11) cell lines, THP-1 was derived from a 1-year old male [22]; NOMO-1 from a 31-year old female [23]; and MOLM-13 from a 20-year old male [15]. MV4.11 was derived from a 10-year old male [16].

To compare these cell lines, we performed RNA-Seq using deep sequencing (> 120 M paired-end reads per sample). Surprisingly, clustering and principal component analysis (PCA) revealed the closest concordance in transcriptomic profile between MV4.11 (t(4;11)) and MOLM-13 (t(9;11)), with the two other t(9;11) cell lines being more distally clustered (Fig. 1A and B) (discussed further below). Comparative analysis of cell-type characteristics using CIBERSORTX [24] revealed that NOMO-1 exhibited a predominantly monocytic transcriptome, with the other three cell lines exhibiting stronger macrophage characteristics (Fig. 1C). As reported in previous studies [25, 26], our RNA-Seq datasets revealed the presence of multiple distinct fusion transcripts involving the primary breakpoint in MOLM-13, MV4.11 and THP-1 cell lines (Fig. 1D, E). Specifically, in MOLM-13 two different break points were observed within the KMT2A locus, leading to 2 different chimeric transcripts of KMT2A-MLLT3. In the case of MV4.11, reciprocal fusion KMT2A-AFF1 transcripts were detected (Fig. 1D, E). In THP-1, fusion transcripts were detected between KMT2A and two different partners on chromosome 9, MLLT3 and SNAPC3. A full analysis of all fusion transcripts detected in each cell line is also provided in Supplementary Tables 1, 2, 3, and 4.

Fig. 1
figure 1

Characterisation of AML cell lines’ identity. (A) Sample clustering based on expression distances after a variance stabilizing transformation of read counts. (B) Principal component analysis (PCA) of the AML cell line dataset. (C) Characterisation of AML cell line identity. Inferred phenotypic profiles of the four AML cell lines were generated with CIBERSORTX. Published RNA-Seq data from THP-1 obtained from ATCC and DSMZ cell line repositories were used as a comparison with our own THP-1 dataset (Clone 5). The phenotypic characteristic of THP-1 is completely altered when chemically induced to differentiate into macrophages in culture (THP-1 control vs. THP-1 + PMA). Published RNA-Seq data from SUPT1 and EBV cell lines is included as comparisons for T-cell and B-cell profiles, respectively. (D) Genomic coordinates of fusion transcripts spanning the primary breakpoints detected in the cell lines. (E) Quantification of junction reads for the MLL fusion transcripts. The counts shown indicate the number of RNA-Seq fragments containing a split read at the site of the fusion junction

To further explore differences between the transcriptomes, we carried out pairwise comparisons between all cell lines (Fig. 2, Supplementary Table 5). These analyses revealed widescale changes in gene expression, with between 3,777 and 5,786 differentially expressed genes (DEG) (cut-offs of fold change > 2.5 and padj < 0.01) observed between any cell line pair. For each cell line, we identified the genes specifically up- or down- regulated relative to all other three lines (Fig. 2A, Supplementary Table 5). Gene ontology (GO) enrichment tests revealed alteration of distinct biological processes and pathways (Fig. 2B). Specifically, NOMO-1 showed relative enrichment of signatures associated with cytokine and innate immune signalling pathways, MV4.11 of pathways associated with immune (Natural Killer) cell activation, signalling and protein kinase regulation, and THP-1 of pathways associated with regulation of the actin cytoskeleton (Fig. 2B). In contrast, MOLM-13 showed few enriched ontologies, but relative depletion of signalling pathways (particularly Wnt-signalling), and signatures of cell migration and adhesion. Notably, pathways related to transcription factor binding, RNA polymerase II activity and gene expression were among the most significantly altered pathways specific to each cell line, suggesting that alterations in transcriptional regulation are a primary determinant of the different characteristics of these cell lines, regardless of the translocation status.

Fig. 2
figure 2

Differential expression analysis of AML cell line models. (A) Venn diagrams representing genes down- or up- regulated (cutoffs fold change > 2.5 and padj < 0.01) in the indicated cell line, relative to each of the three other cell lines. (B) The overlap of the gene sets (dark blue) in (A) was used in the GO enrichment analyses, where bar charts indicate GO terms significantly enriched (red) or depleted (blue) in each cell line (FDR-corrected p < 0.05)

Differential expression of transcription factors and epigenetic regulators

Following the above observation, we focused further on differences in key transcription factors (TF) and epigenetic regulators which have been strongly implicated in the aetiology of AML. The members of the homeobox (HOX) family of TF are key regulators of normal hematopoiesis [27,28,29] and frequently dysregulated (mostly upregulated) in AML, where they have been associated as prognostic factors [30]. Focused analysis revealed striking differences between these AML cell lines in expression of the HOX genes (Supplementary Fig. 1). While most genes of the HOXB cluster are more prominently expressed in MV4.11 (t(4;11)) (Supplementary Fig. 1A, B), the HOXA cluster appears to be separated into two distinct regulatory subdomains, with the 5’ subdomain (HOXA10, 11 and 13) also strongest in MV4.11, while the 3’ subdomain (HOXA1-9) is most prominently activated in NOMO-1 (t(9;11)) (Supplementary Fig. 1A, C). Interestingly, the 3’ and 5’ subdomains of the HOXA cluster have been shown to be subject to distinct epigenetic regulation and display different expression profiles during monocytic differentiation [31]. HOXA9 and its cofactor MEIS1, have previously been identified as key target genes of leukemogenic MLL fusion proteins [2, 32,33,34] and enforced coexpression of both genes leads to rapid development of AML [35]. Of interest, we observed lowest expression of HOXA9 and no expression of MEIS1 in THP-1 (Supplementary Fig. 1D). The depletion of these key mediators of MLL activity in THP-1 is consistent with previous results indicating that THP-1 was less sensitive to depletion of MLL than MOLM-13, NOMO-1 and MV4.11 [33]. Interestingly, it has been previously shown that expression of MEIS1 varies between THP-1 cells accessed from different biorepository [36].

Our analysis also indicated differences between the cell lines in the expression of several epigenetic regulators known to be implicated in myeloid malignancies, including regulators of DNA methylation (Fig. 3A), components of the polycomb repressive complexes (PRC1, Fig. 3B and PRC2, Fig. 3C), and other histone modifying enzymes (lysine acetyl transferase KAT2B, Fig. 3D, lysine demethylases KDM4A, KDM5B and KDM5D, Fig. 3E). Other epigenetic regulator genes which can harbour driver mutations in AML (DNMT3A, ASXL1) [37,38,39] were expressed at similar levels in all four cell lines (Supplementary Table 6). Dysregulation of the Polycomb-mediated histone modification H3K27me3, including loss-of-function of the H3K27 methyltransferase EZH2 [40,41,42,43] or demethylase KDM6A/UTX [44,45,46,47,48,49,50] are common features of myeloid malignancies. As reported previously [51,52,53], western blot and whole genome sequencing (WGS) analysis confirmed that THP-1 lacks expression of UTX due to partial deletion of the single KDM6A allele (X-linked) in this cell line (Fig. 3C, Supplementary Fig. 2). Western blot analysis also revealed a striking depletion of the total levels of H3K27me3 in NOMO-1 (Fig. 3F), which is associated with both lower expression of EZH2 (Fig. 3C) as well as high expression of KDM6A gene (Fig. 3C) and its protein product UTX (Fig. 3F). Consistent with this observation, NOMO-1 also showed elevated expression of key genes which have been shown to be repressed by EZH2 in AML (Fig. 3H), including target genes implicated in chemotherapy resistance, relapse, and poor prognosis [42]. Altogether, these findings suggest important differences in epigenetic regulation between the compared AML models.

Fig. 3
figure 3

Differential expression of epigenetic regulators in AML cell lines. Bar charts showing expression (transcripts per million, TPM) in RNA-Seq datasets of the AML cell lines for the following genes: (A) Regulators of DNA methylation are represented by DNA methyltransferase DNMT3B and demethylases TET1 and TET2. (B) BMI1 and (C) EZH2 (H3K27 methyltransferase) and JARID2 (DNA binding protein) are core components of polycomb repressive complexes 1 (PRC1) and 2 (PRC2) respectively, and KDM6A, KDM6B, UTY are H3K27 demethylases. (D) Lysine acetyl transferase KAT2B (E) Lysine demethylases (KDM4A, 5B, and 5D). (F) Western blot analysis of UTX and H3K27me3 levels in AML cell lines. Both β-actin and Histone H3 were used as loading control. Recurrent protein degradation is notable in MOLM-13 cell line. (G) Bar charts showing expression of PRTN3 (H) Clustered heatmap representation of genes previously shown to be repressed by PcG in AML (scaling applied to rows)

Of note, the western blot analyses also suggested higher levels of protein degradation in MOLM-13 (Fig. 3F), which also displayed very high expression of PRTN3 encoding Proteinase 3 (PR3, myeoblastin, Fig. 3G), a serine protease localized on the cellular membrane of polymorphonuclear neutrophils and identified as a leukemia associated antigen [54]. These observations suggest that MOLM-13 may not be the AML model of choice for carrying out proteomic studies.

We also identified significant differences in the expression of key transcriptional regulators of hematopoiesis such as GATA2, TAL2 and NF-E2 (Supplementary Fig. 3), all hallmarks of hematological malignancies [28, 55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79]. Other key regulators of hematopoiesis known to be involved in AML such as JUND, ZEB1, IRF, C/EBP or Forkhead Box (FOX) families [68, 72,73,74, 76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96] also present notable differences in expression between the four cell lines (Supplementary Fig. 3). Striking differences in expression were also observed for other important regulators of hematopoiesis, including CITED4 and NFYC [97,98,99] (both strongly expressed in THP-1) (Supplementary Fig. 3). Of particular note, TP53 was expressed only in MV4.11 (t(4;11)) and MOLM-13 (t(9;11)), the two cell lines whose transcriptomes clustered most closely, despite bearing different primary translocations (Fig. 1A, B). Interestingly, ontology analysis of genes which are upregulated in MOLM-13 or MV4.11 relative to THP-1 and NOMO-1, revealed a striking enrichment of p53 related signatures (Supplementary Fig. 4A, B, C & D), suggesting that common activation of the p53 signalling pathway may be one important factor contributing to the transcriptional similarity of these two cell lines. In addition to the deletion of one allele of TP53 which has previously been described in THP-1 [100], WGS of this cell line also confirmed a 26 bp frameshift deletion in exon 5 of the other TP53 allele [101] (Supplementary Fig. 4E), likely contributing to nonsense-mediated mRNA decay [102]. A frameshift mutation (p.C242fs*5) has also been previously reported in one TP53 allele in NOMO-1 [100].

One striking observation from our data was that among genes that are differentially expressed between cell lines, there was a strong enrichment for members of the C2H2 Zinc finger (ZNF) family (HGNC Group ID 28) (Fig. 4A, Supplementary Table 6). Compared to their overall abundance in the human genome (1.3% of annotated genes), the C2H2 ZNF family was particularly enriched among the set of genes that were specifically depleted in MOLM-13 (73 out of 1162 genes, 6.3%) and NOMO-1 (19 out of 438 genes, 4.3%) and among the set of genes that were specifically increased in THP-1 (60 out of 1378 genes; 4.4%). Approximately half of the C2H2 ZNF family contain a KRAB (Krüppel-associated box) transcriptional repressor domain [103, 104]. Members of the C2H2 ZNF family are predominantly localised on chromosome 19 (Fig. 4B), where they are organised within 6 main clusters [105] (Supplementary Fig. 5). Interestingly, we observed striking differences between the cell lines in expression of these individual clusters, including strong expression of cluster 2 in THP-1 and generally poor expression of cluster 5 in MOLM-13 (Fig. 4C and Supplementary Fig. 6). We also observed several clear examples of physically proximal subclusters of ZNF genes within the primary clusters which showed correlating expression profiles, suggesting co-ordinated local regulation of proximal genes, consistent with observations in other cell types [92]. For example, a sub-cluster of genes within cluster 2 (ZNF257 to ZNF730) showed highly specific activation in THP-1 (Fig. 4C, D), while an adjacent subcluster (ZNF724 to ZNF254) was elevated in both THP-1 and MV4.11 (Fig. 4C, E). Within cluster 5, two subclusters (ZNF577 to ZNF432 and ZNF808 to ZNF816) with striking depletion in MOLM-13 (Fig. 4C, F) are separated by a distinct subcluster of four genes (ZNF616 to ZNF480) which are highly expressed in this cell line (Fig. 4C). To explore further, we analysed the prognostic significance of expression of these chr19 ZNF genes in patients of The Cancer Genome Atlas (TCGA) Acute Myeloid Leukemia cohort [44]. Interestingly, we observed correlations between expression of many of these ZNF proteins and prognostic outcome, with low expression most frequently associated with poor overall survival (Fig. 5, Supplementary Table 7).

Fig. 4
figure 4

Differential expression of C2H2 Zinc Finger family transcription factors in AML cell lines. (A) Bar charts indicating the percentage of genes specifically up- or down- regulated in each cell line which are members of the C2H2 ZNF family. The dashed line indicates the overall percentage of this family across the genome (B) Bar charts indicating the number of total and differentially expressed C2H2 ZNF genes on each chromosome. Differentially expressed ZNFs are those that showed differential expression (in the same direction) between one cell line and all other three cell lines in pairwise comparisons as shown in Fig. 2. (C) Heatmap representation of the expression of ZNF genes within cluster 2 (left) and cluster 5 (right) on chromosome 19 (scaled within rows). Hierarchical clustering was applied only to columns (samples) and ZNF genes are shown according to their physical position within the gene cluster. Brackets indicate sub-clusters of physically-proximal genes showing co-ordinated expression profiles across the cell lines as described in the text. (D-F) Bar charts showing expression (TPM) of the indicated ZNF genes from different subclusters as described in the text

Fig. 5
figure 5

Survival analysis of ZNF genes on chromosome 19. (A) Map of chromosome 19 indicating the location of the 6 main clusters of ZNF genes. (B) Kaplan-Meier analysis of overall survival in patients of the LAML cohort (n = 132) with low (< median, blue) or high (> median, red) expression of selected ZNF genes within each cluster. The gene highlighted in orange (ZNF296) has the opposite behaviour to that observed for all the other genes

One important role of these KRAB-containing ZNF proteins is the maintenance of heterochromatin and transcriptional repression at genomic retroelements [105, 106]. Expression of these genes has been associated with pro- [106] or anti- [107] tumorigenic influences in different malignancies, suggesting disease-specific effects. As well as their DNA binding-dependent function, a recent study showed that many members of the C2H2 ZNF family also bind directly to RNA, and can regulate post-transcriptional processes of splicing, polyadenylation and m6A modification, significantly expanding our understanding of the regulatory importance of this protein family [104]. Although these ZNF clusters have not previously been implicated in AML, our observations suggest that differential expression of these ZNF proteins on chr19 could exert important functional effects in patients and in the cell line models studied here, and that the role of these ZNF in AML is worthy of further study and functional characterisation.

Conclusion

While studying the complex biological processes involved in leukemia, it is of great importance to define the most suitable cell line model required for a specific study. The detailed transcriptomic profiling of four commonly used AML cell lines reported here has revealed important differences in the expression of genes encoding TFs and epigenetic regulators, several of which have previously been implicated in the aetiology of the disease. Importantly, most differences in expression of these factors were not overtly correlated with the primary cytogenetic translocation present in each cell line demonstrating the importance of thorough molecular characterisation of these cellular models.

Materials and methods

Cell lines

Cells lines were kindly donated by David Hume (THP-1, clone 5) and Kamil Kranc (NOMO-1, MOLM-13, MV4.11). The THP-1 subclone (clone 5) used in this study was previously selected for its greater ability to differentiate into macrophages [108]. All were cultured in RPMI 1640 with the same batch of endotoxin free FBS (10%) (Gibco Lot #08F7582K). All cells were tested for mycoplasma using the Venor™ GeM Mycoplasma Detection Kit (MP0025– Sigma-Aldrich).

RNA extraction

Total RNA was extracted with Tri Reagent per the manufacturer’s instructions (AM9738– Thermo Fisher), followed by TURBO DNase treatment (AM2238– Thermo Fisher). RNA quality was assessed by Agilent Tapestation 2200 with all samples achieving a RIN > 9.5.

RNA-Seq library Preparation and sequencing

The Illumina TruSeq Stranded mRNA kit was used for RNA-Seq library preparation, with one round of RiboZero Gold for ribo-depletion. Preparation of libraries was carried out in a single batch. Pooled libraries (one full lane per cell line) were sequenced together on a Illumina HiSeq 4000 instrument at Edinburgh Genomics (http://genomics.ed.ac.uk/) generating 2 × 150 bp paired-end reads. Read counts are shown in Supplementary Table 8.

Bioinformatics analysis

RNA-Seq data was aligned to the reference human genome (GRCh38) using STAR (v 2.7.1a) [109] allowing a maximum of 20 multimappers per read. Mapping rates were very high in all cases, above 90% (Supplementary Table 8). Aligned RNA-Seq data was analysed using the immune cell deconvolution software CIBERSORTX [24] running on relative mode with 1,000 permutations, using the LM22 signature matrix (microarray profile of 22 defined human immune cell types) and subset to only display monocyte, M0, M1 and M2 macrophages. Quantification of fusion transcripts was carried out using STARFusion (v1.10.0). For differential expression analysis, gene level read counts were obtained using featureCounts (v. 1.6.3) [110]. Following removal of genes poorly expressed in all samples (maximum mapped read count < 20), and genes on the Y chromosome, genes which were differentially expressed between pairs of cell lines (fold change > 2.5 and padj < 0.01) were determined using the DESeq2 package with Median Ratio (MR) normalisation [111] in R [112]. Overlaps and Venn diagrams of up- or down- regulated genes in a single cell line compared to each of the other three cell lines were carried out using Biovenn [113]. Ontology analysis was performed using the Gene Ontology Resource tool (release 17.6.24) [114] with the GO Biological Process_Slim and GO Molecular Function_Slim collections [115] after applying a cutoff for enrichment or depletion of FDR q < 0.05. GO terms within the same hierarchy were reduced to retain only the most specialised “child” term. Ontology analysis in Supplementary Fig. 4 was performed using the “Compute Overlaps” tool of MSigDB (https://www.gsea-msigdb.org/gsea/msigdb/index.jsp), with the Ontology Gene Sets (C5), applying a cutoff for enrichment of FDR q < 0.05. Heatmaps were established from MRN-normalised expression values using Heatmaper [116] with scaling applied to rows (genes). Where shown, hierarchical dendograms indicate the outcome of complete linkage clustering using the Euclidean measure of distance. Gene expression bar charts display the mean ± SD of each distribution and were analysed by ordinary one-way ANOVA with Bonferroni’s correction for multiple testing using GraphPad Prism (v10.3.1). Only significant pairwise comparisons are indicated (* p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001).

Kaplan-meier survival analysis

The patient survival and expression data (FPKM-UQ) were extracted from TCGA Acute Myeloid Leukemia (LAML) [44] via Xena browser [117]. For each gene, patients were segregated into groups of high (> median, n66) and low (< median, n66) expressors and Kaplan-Meier survival analysis was carried out using GraphPad Prism 10.3.1. The Log-rank test p value is indicated.

Whole genome sequencing

The genomic DNA of all AML cell lines was obtained using phenol/chloroform extraction. The genomic DNA was randomly sheared into short fragments. The obtained fragments were end repaired, A-tailed, and further ligated with Illumina adapters. The fragments with adapters were size-selected, PCR amplified and purified. The library was checked with Qubit and real-time PCR for quantification and Bioanalyzer to determine size distribution. Next Generation sequencing libraries were prepared using Illumina SeqLab specific TruSeq PCR-Free High Throughput library preparation kits. Sequencing was performed using NovaSeq X Plus (PE150) instrument by NOVOGENE at 25x coverage.

Western blot

Cells were resuspended in 1x PBS and lysed with 1x pre-heated PBS with SDS 2% (1% final). The cells were then boiled at 95 °C for 5 min, with agitation every minute. Lysates were pipetted up and down or sonicated to reduce viscosity, if necessary, then diluted 1:1 in 2x loading (SB) buffer (130mM Tris pH 6.8, 5% SDS, 20% Glycerol, 10% β-Mercaptoethanol, 0.01% Bromophenol Blue). Ten µg of cell lysates were separated on SDS-PAGE gradient (NuPAGE Bis-Tris 4–12%). Antibodies used were Histone H3 (Abcam 1791), UTX (Cell Signaling Technology #33510), H3K27me3 by Millipore (07-449) and β-actin-HRP (Sigma, A3854).

Data availability

THP-1 RNA-Seq data sets from ATCC and DSMZ cell repositories were obtained through the Gene Expression Omnibus (GEO) GSE130985 [36]. RNA-Seq data set for control and PMA treated THP-1 were obtained from BioProject PRJNA449980 [118], SUPT1 from BioProject PRJNA449980 [118] and EBV from Bioproject PRJNA518137 [119]. WGS generated during this study are available at the Sequence Read Archive (SRA) repository under accession number PRJNA1152337 and RNA-Seq datasets of this study have been deposited with the GEO data bank under accession number GSE274887.

References

  1. Brazel AJ, Vernimmen D. The complexity of epigenetic diseases. J Pathol. 2016;238(2):333–44.

    Article  PubMed  Google Scholar 

  2. Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, Levine JE, Wang J, Hahn WC, Gilliland DG, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006;442(7104):818–22.

    Article  CAS  PubMed  Google Scholar 

  3. Horton SJ, Huntly BJ. Recent advances in acute myeloid leukemia stem cell biology. Haematologica. 2012;97(7):966–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shlush LI, Zandi S, Mitchell A, Chen WC, Brandwein JM, Gupta V, Kennedy JA, Schimmer AD, Schuh AC, Yee KW, et al. Identification of pre-leukaemic Haematopoietic stem cells in acute leukaemia. Nature. 2014;506(7488):328–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lagunas-Rangel FA, Chavez-Valencia V, Gomez-Guijosa MA, Cortes-Penagos C. Acute myeloid Leukemia-Genetic alterations and their clinical prognosis. Int J Hematol Oncol Stem Cell Res. 2017;11(4):328–39.

    PubMed  PubMed Central  Google Scholar 

  6. Pedersen-Bjergaard J, Andersen MK, Andersen MT, Christiansen DH. Genetics of therapy-related myelodysplasia and acute myeloid leukemia. Leukemia. 2008;22(2):240–8.

    Article  CAS  PubMed  Google Scholar 

  7. Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell. 2012;48(4):491–507.

    Article  CAS  PubMed  Google Scholar 

  8. Daigle SR, Olhava EJ, Therkelsen CA, Majer CR, Sneeringer CJ, Song J, Johnston LD, Scott MP, Smith JJ, Xiao Y, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20(1):53–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150(1):12–27.

    Article  CAS  PubMed  Google Scholar 

  10. Borkin D, He S, Miao H, Kempinska K, Pollock J, Chase J, Purohit T, Malik B, Zhao T, Wang J, et al. Pharmacologic Inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell. 2015;27(4):589–602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen CW, Koche RP, Sinha AU, Deshpande AJ, Zhu N, Eng R, Doench JG, Xu H, Chu SH, Qi J, et al. DOT1L inhibits SIRT1-mediated epigenetic Silencing to maintain leukemic gene expression in MLL-rearranged leukemia. Nat Med. 2015;21(4):335–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cai SF, Chen CW, Armstrong SA. Drugging chromatin in cancer: recent advances and novel approaches. Mol Cell. 2015;60(4):561–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Skopek R, Palusinska M, Kaczor-Keller K, Pingwara R, Papierniak-Wygladala A, Schenk T, Lewicki S, Zelent A, Szymanski L. Choosing the right cell line for acute myeloid leukemia (AML) research. Int J Mol Sci. 2023; 24(6).

  14. Drexler HG, Quentmeier H, MacLeod RA. Malignant hematopoietic cell lines: in vitro models for the study of MLL gene alterations. Leukemia. 2004;18(2):227–32.

    Article  CAS  PubMed  Google Scholar 

  15. Matsuo Y, MacLeod RA, Uphoff CC, Drexler HG, Nishizaki C, Katayama Y, Kimura G, Fujii N, Omoto E, Harada M, et al. Two acute monocytic leukemia (AML-M5a) cell lines (MOLM-13 and MOLM-14) with interclonal phenotypic heterogeneity showing MLL-AF9 fusion resulting from an occult chromosome insertion, ins(11;9)(q23;p22p23). Leukemia. 1997;11(9):1469–77.

    Article  CAS  PubMed  Google Scholar 

  16. Lange B, Valtieri M, Santoli D, Caracciolo D, Mavilio F, Gemperlein I, Griffin C, Emanuel B, Finan J, Nowell P, et al. Growth factor requirements of childhood acute leukemia: establishment of GM-CSF-dependent cell lines. Blood. 1987;70(1):192–9.

    Article  CAS  PubMed  Google Scholar 

  17. Meyer C, Burmeister T, Groger D, Tsaur G, Fechina L, Renneville A, Sutton R, Venn NC, Emerenciano M, Pombo-de-Oliveira MS, et al. The MLL recombinome of acute leukemias in 2017. Leukemia. 2018;32(2):273–84.

    Article  CAS  PubMed  Google Scholar 

  18. Parkin JL, Arthur DC, Abramson CS, McKenna RW, Kersey JH, Heideman RL, Brunning RD. Acute leukemia associated with the t(4;11) chromosome rearrangement: ultrastructural and Immunologic characteristics. Blood. 1982;60(6):1321–31.

    Article  CAS  PubMed  Google Scholar 

  19. Stong RC, Kersey JH. In vitro culture of leukemic cells in t(4;11) acute leukemia. Blood. 1985;66(2):439–43.

    Article  CAS  PubMed  Google Scholar 

  20. Schneidawind C, Jeong J, Schneidawind D, Kim IS, Duque-Afonso J, Wong SHK, Iwasaki M, Breese EH, Zehnder JL, Porteus M, et al. MLL leukemia induction by t(9;11) chromosomal translocation in human hematopoietic stem cells using genome editing. Blood Adv. 2018;2(8):832–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rice S, Jackson T, Crump NT, Fordham N, Elliott N, O’Byrne S, Fanego M, Addy D, Crabb T, Dryden C, et al. A human fetal liver-derived infant MLL-AF4 acute lymphoblastic leukemia model reveals a distinct fetal gene expression program. Nat Commun. 2021;12(1):6905.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tsuchiya S, Yamabe M, Yamaguchi Y, Kobayashi Y, Konno T, Tada K. Establishment and characterization of a human acute monocytic leukemia cell line (THP-1). Int J Cancer. 1980;26(2):171–6.

    Article  CAS  PubMed  Google Scholar 

  23. Kato Y, Ogura M, Okumura M, Morishima Y, Horita T, Ohno R, Saito H, Hirabayashi N. Establishment of peroxidase positive, human monocytic leukemia cell line (NOMO-1) and its characteristics. Acta Haematol Japan. 1986;49(277).

  24. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri AA, Scherer F, Khodadoust MS, Esfahani MS, Luca BA, Steiner D, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol. 2019;37(7):773–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hilden JM, Chen CS, Moore R, Frestedt J, Kersey JH. Heterogeneity in MLL/AF-4 fusion messenger RNA detected by the polymerase chain reaction in t(4;11) acute leukemia. Cancer Res. 1993;53(17):3853–6.

    CAS  PubMed  Google Scholar 

  26. Montemurro L, Tonelli R, Fazzina R, Martino V, Marino F, Pession A. Identification of two MLL-MLLT3 (alias MLL-AF9) chimeric transcripts in the MOLM-13 cell line. Cancer Genet Cytogenet. 2004;154(1):96–7.

    Article  CAS  PubMed  Google Scholar 

  27. Giampaolo A, Sterpetti P, Bulgarini D, Samoggia P, Pelosi E, Valtieri M, Peschle C. Key functional role and lineage-specific expression of selected HOXB genes in purified hematopoietic progenitor differentiation. Blood. 1994;84(11):3637–47.

    Article  CAS  PubMed  Google Scholar 

  28. Kawagoe H, Humphries RK, Blair A, Sutherland HJ, Hogge DE. Expression of HOX genes, HOX cofactors, and MLL in phenotypically and functionally defined subpopulations of leukemic and normal human hematopoietic cells. Leukemia. 1999;13(5):687–98.

    Article  CAS  PubMed  Google Scholar 

  29. Pineault N, Helgason CD, Lawrence HJ, Humphries RK. Differential expression of Hox, Meis1, and Pbx1 genes in primitive cells throughout murine hematopoietic ontogeny. Exp Hematol. 2002;30(1):49–57.

    Article  CAS  PubMed  Google Scholar 

  30. Alharbi RA, Pettengell R, Pandha HS, Morgan R. The role of HOX genes in normal hematopoiesis and acute leukemia. Leukemia. 2013;27(5):1000–8.

    Article  CAS  PubMed  Google Scholar 

  31. Rousseau M, Crutchley JL, Miura H, Suderman M, Blanchette M, Dostie J. Hox in motion: tracking HoxA cluster conformation during differentiation. Nucleic Acids Res. 2014;42(3):1524–40.

    Article  CAS  PubMed  Google Scholar 

  32. Armstrong SA, Staunton JE, Silverman LB, Pieters R, den Boer ML, Minden MD, Sallan SE, Lander ES, Golub TR, Korsmeyer SJ. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat Genet. 2002;30(1):41–7.

    Article  CAS  PubMed  Google Scholar 

  33. Lu B, Klingbeil O, Tarumoto Y, Somerville TDD, Huang YH, Wei Y, Wai DC, Low JKK, Milazzo JP, Wu XS, et al. A transcription factor addiction in leukemia imposed by the MLL promoter sequence. Cancer Cell. 2018;34(6):970–e981978.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Collins CT, Hess JL. Deregulation of the HOXA9/MEIS1 axis in acute leukemia. Curr Opin Hematol. 2016;23(4):354–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kroon E, Krosl J, Thorsteinsdottir U, Baban S, Buchberg AM, Sauvageau G. Hoxa9 transforms primary bone marrow cells through specific collaboration with Meis1a but not Pbx1b. EMBO J. 1998;17(13):3714–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Noronha N, Ehx G, Meunier MC, Laverdure JP, Theriault C, Perreault C. Major multilevel molecular divergence between THP-1 cells from different biorepositories. Int J Cancer. 2020;147(7):2000–6.

    Article  CAS  PubMed  Google Scholar 

  37. Gallipoli P, Giotopoulos G, Huntly BJ. Epigenetic regulators as promising therapeutic targets in acute myeloid leukemia. Ther Adv Hematol. 2015;6(3):103–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Pastore F, Levine RL. Epigenetic regulators and their impact on therapy in acute myeloid leukemia. Haematologica. 2016;101(3):269–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, Han J, Wei X. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther. 2019;4:62.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Mechaal A, Menif S, Abbes S, Safra I. EZH2, new diagnosis and prognosis marker in acute myeloid leukemia patients. Adv Med Sci. 2019;64(2):395–401.

    Article  PubMed  Google Scholar 

  41. Chu MQ, Zhang TJ, Xu ZJ, Gu Y, Ma JC, Zhang W, Wen XM, Lin J, Qian J, Zhou JD. EZH2 dysregulation: potential biomarkers predicting prognosis and guiding treatment choice in acute myeloid leukaemia. J Cell Mol Med. 2020;24(2):1640–9.

    Article  CAS  PubMed  Google Scholar 

  42. Kempf JM, Weser S, Bartoschek MD, Metzeler KH, Vick B, Herold T, Volse K, Mattes R, Scholz M, Wange LE, et al. Loss-of-function mutations in the histone methyltransferase EZH2 promote chemotherapy resistance in AML. Sci Rep. 2021;11(1):5838.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Stomper J, Meier R, Ma T, Pfeifer D, Ihorst G, Blagitko-Dorfs N, Greve G, Zimmer D, Platzbecker U, Hagemeijer A, et al. Integrative study of EZH2 mutational status, copy number, protein expression and H3K27 trimethylation in AML/MDS patients. Clin Epigenetics. 2021;13(1):77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cancer Genome Atlas Research N, Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson A, Hoadley K, Triche TJ Jr., Laird PW, et al. Genomic and epigenomic landscapes of adult de Novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74.

    Article  Google Scholar 

  45. Mar BG, Bullinger L, Basu E, Schlis K, Silverman LB, Dohner K, Armstrong SA. Sequencing histone-modifying enzymes identifies UTX mutations in acute lymphoblastic leukemia. Leukemia. 2012;26(8):1881–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jankowska AM, Makishima H, Tiu RV, Szpurka H, Huang Y, Traina F, Visconte V, Sugimoto Y, Prince C, O’Keefe C, et al. Mutational spectrum analysis of chronic myelomonocytic leukemia includes genes associated with epigenetic regulation: UTX, EZH2, and DNMT3A. Blood. 2011;118(14):3932–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kar SA, Jankowska A, Makishima H, Visconte V, Jerez A, Sugimoto Y, Muramatsu H, Traina F, Afable M, Guinta K, et al. Spliceosomal gene mutations are frequent events in the diverse mutational spectrum of chronic myelomonocytic leukemia but largely absent in juvenile myelomonocytic leukemia. Haematologica. 2012;98(1):107–13.

    Article  PubMed  Google Scholar 

  48. Gozdecka M, Meduri E, Mazan M, Tzelepis K, Dudek M, Knights AJ, Pardo M, Yu L, Choudhary JS, Metzakopian E, et al. UTX-mediated enhancer and chromatin remodeling suppresses myeloid leukemogenesis through noncatalytic inverse regulation of ETS and GATA programs. Nat Genet. 2018;50(6):883–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Garg M, Nagata Y, Kanojia D, Mayakonda A, Yoshida K, Haridas Keloth S, Zang ZJ, Okuno Y, Shiraishi Y, Chiba K, et al. Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood. 2015;126(22):2491–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Adati N, Huang MC, Suzuki T, Suzuki H, Kojima T. High-resolution analysis of aberrant regions in autosomal chromosomes in human leukemia THP-1 cell line. BMC Res Notes. 2009;2:153.

    Article  PubMed  PubMed Central  Google Scholar 

  52. van Haaften G, Dalgliesh GL, Davies H, Chen L, Bignell G, Greenman C, Edkins S, Hardy C, O’Meara S, Teague J, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet. 2009;41(5):521–3.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Liu J, Mercher T, Scholl C, Brumme K, Gilliland DG, Zhu N. A functional role for the histone demethylase UTX in normal and malignant hematopoietic cells. Exp Hematol. 2012;40(6):487–e498483.

    Article  CAS  PubMed  Google Scholar 

  54. Greiner J, Bullinger L, Guinn BA, Dohner H, Schmitt M. Leukemia-associated antigens are critical for the proliferation of acute myeloid leukemia cells. Clin Cancer Res. 2008;14(22):7161–6.

    Article  CAS  PubMed  Google Scholar 

  55. Li Z, Chen P, Su R, Hu C, Li Y, Elkahloun AG, Zuo Z, Gurbuxani S, Arnovitz S, Weng H, et al. PBX3 and MEIS1 cooperate in hematopoietic cells to drive acute myeloid leukemias characterized by a core transcriptome of the MLL-Rearranged disease. Cancer Res. 2016;76(3):619–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hahn CN, Chong CE, Carmichael CL, Wilkins EJ, Brautigan PJ, Li XC, Babic M, Lin M, Carmagnac A, Lee YK, et al. Heritable GATA2 mutations associated with Familial myelodysplastic syndrome and acute myeloid leukemia. Nat Genet. 2011;43(10):1012–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Katerndahl CDS, Rogers ORS, Day RB, Cai MA, Rooney TP, Helton NM, Hoock M, Ramakrishnan SM, Nonavinkere Srivatsan S, Wartman LD, et al. Tumor suppressor function of Gata2 in acute promyelocytic leukemia. Blood. 2021;138(13):1148–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nanaa A, Viswanatha D, Xie Z, Jevremovic D, Nguyen P, Salama ME, Greipp P, Bessonen K, Gangat N, Patnaik M, et al. Clinical and biological characteristics and prognostic impact of somatic GATA2 mutations in myeloid malignancies: a single institution experience. Blood Cancer J. 2021;11(6):122.

    Article  PubMed  PubMed Central  Google Scholar 

  59. O’Neil J, Shank J, Cusson N, Murre C, Kelliher M. TAL1/SCL induces leukemia by inhibiting the transcriptional activity of E47/HEB. Cancer Cell. 2004;5(6):587–96.

    Article  PubMed  Google Scholar 

  60. Cardoso BA, de Almeida SF, Laranjeira AB, Carmo-Fonseca M, Yunes JA, Coffer PJ, Barata JT. TAL1/SCL is downregulated upon histone deacetylase Inhibition in T-cell acute lymphoblastic leukemia cells. Leukemia. 2011;25(10):1578–86.

    Article  CAS  PubMed  Google Scholar 

  61. Sanda T, Leong WZ. TAL1 as a master oncogenic transcription factor in T-cell acute lymphoblastic leukemia. Exp Hematol. 2017;53:7–15.

    Article  CAS  PubMed  Google Scholar 

  62. Yang L, Chen F, Zhu H, Chen Y, Dong B, Shi M, Wang W, Jiang Q, Zhang L, Huang X, et al. 3D genome alterations associated with dysregulated HOXA13 expression in high-risk T-lineage acute lymphoblastic leukemia. Nat Commun. 2021;12(1):3708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Peeken JC, Jutzi JS, Wehrle J, Koellerer C, Staehle HF, Becker H, Schoenwandt E, Seeger TS, Schanne DH, Gothwal M, et al. Epigenetic regulation of NFE2 overexpression in myeloproliferative neoplasms. Blood. 2018;131(18):2065–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Jutzi JS, Basu T, Pellmann M, Kaiser S, Steinemann D, Sanders MA, Hinai ASA, Zeilemaker A, Bojtine Kovacs S, Koellerer C, et al. Altered NFE2 activity predisposes to leukemic transformation and myelosarcoma with AML-specific aberrations. Blood. 2019;133(16):1766–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Marcault C, Zhao LP, Maslah N, Verger E, Daltro de Oliveira R, Soret-Dulphy J, Cazaux M, Gauthier N, Roux B, Clappier E, et al. Impact of NFE2 mutations on AML transformation and overall survival in patients with myeloproliferative neoplasms. Blood. 2021;138(21):2142–8.

    Article  CAS  PubMed  Google Scholar 

  66. Lehmann W, Mossmann D, Kleemann J, Mock K, Meisinger C, Brummer T, Herr R, Brabletz S, Stemmler MP, Brabletz T. ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat Commun. 2016;7:10498.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Scott CL, Omilusik KD. ZEBs: novel players in immune cell development and function. Trends Immunol. 2019;40(5):431–46.

    Article  CAS  PubMed  Google Scholar 

  68. Almotiri A, Alzahrani H, Menendez-Gonzalez JB, Abdelfattah A, Alotaibi B, Saleh L, Greene A, Georgiou M, Gibbs A, Alsayari A et al. Zeb1 modulates hematopoietic stem cell fates required for suppressing acute myeloid leukemia. J Clin Invest. 2021;131(1).

  69. Goossens S, Radaelli E, Blanchet O, Durinck K, Van der Meulen J, Peirs S, Taghon T, Tremblay CS, Costa M, Farhang Ghahremani M, et al. ZEB2 drives immature T-cell lymphoblastic leukaemia development via enhanced tumour-initiating potential and IL-7 receptor signalling. Nat Commun. 2015;6:5794.

    Article  CAS  PubMed  Google Scholar 

  70. Goossens S, Wang J, Tremblay CS, De Medts J, T’Sas S, Nguyen T, Saw J, Haigh K, Curtis DJ, Van Vlierberghe P, et al. ZEB2 and LMO2 drive immature T-cell lymphoblastic leukemia via distinct oncogenic mechanisms. Haematologica. 2019;104(8):1608–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Postigo AA. Opposing functions of ZEB proteins in the regulation of the TGFbeta/BMP signaling pathway. EMBO J. 2003;22(10):2443–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu B, Sun Y, Jiang F, Zhang S, Wu Y, Lan Y, Yang X, Mao N. Disruption of Smad5 gene leads to enhanced proliferation of high-proliferative potential precursors during embryonic hematopoiesis. Blood. 2003;101(1):124–33.

    Article  CAS  PubMed  Google Scholar 

  73. Wang J, Clay-Gilmour AI, Karaesmen E, Rizvi A, Zhu Q, Yan L, Preus L, Liu S, Wang Y, Griffiths E, et al. Genome-Wide association analyses identify variants in IRF4 associated with acute myeloid leukemia and myelodysplastic syndrome susceptibility. Front Genet. 2021;12:554948.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Heintel D, Zojer N, Schreder M, Strasser-Weippl K, Kainz B, Vesely M, Gisslinger H, Drach J, Gaiger A, Jager U, et al. Expression of MUM1/IRF4 mRNA as a prognostic marker in patients with multiple myeloma. Leukemia. 2008;22(2):441–5.

    Article  CAS  PubMed  Google Scholar 

  75. Nerlov C. The C/EBP family of transcription factors: a paradigm for interaction between gene expression and proliferation control. Trends Cell Biol. 2007;17(7):318–24.

    Article  CAS  PubMed  Google Scholar 

  76. Truong BT, Lee YJ, Lodie TA, Park DJ, Perrotti D, Watanabe N, Koeffler HP, Nakajima H, Tenen DG, Kogan SC. CCAAT/Enhancer binding proteins repress the leukemic phenotype of acute myeloid leukemia. Blood. 2003;101(3):1141–8.

    Article  CAS  PubMed  Google Scholar 

  77. Koschmieder S, Rosenbauer F, Steidl U, Owens BM, Tenen DG. Role of transcription factors C/EBPalpha and PU.1 in normal hematopoiesis and leukemia. Int J Hematol. 2005;81(5):368–77.

    Article  CAS  PubMed  Google Scholar 

  78. Akasaka T, Balasas T, Russell LJ, Sugimoto KJ, Majid A, Walewska R, Karran EL, Brown DG, Cain K, Harder L, et al. Five members of the CEBP transcription factor family are targeted by recurrent IGH translocations in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Blood. 2007;109(8):3451–61.

    Article  CAS  PubMed  Google Scholar 

  79. Matsushita H, Nakajima H, Nakamura Y, Tsukamoto H, Tanaka Y, Jin G, Yabe M, Asai S, Ono R, Nosaka T, et al. C/EBPalpha and C/EBPvarepsilon induce the monocytic differentiation of myelomonocytic cells with the MLL-chimeric fusion gene. Oncogene. 2008;27(53):6749–60.

    Article  CAS  PubMed  Google Scholar 

  80. Wang J, Xiong Y. HSH2D contributes to methotrexate resistance in human Tcell acute lymphoblastic leukaemia. Oncol Rep. 2020;44(5):2121–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Ekberg J, Holm C, Jalili S, Richter J, Anagnostaki L, Landberg G, Persson JL. Expression of Cyclin A1 and cell cycle proteins in hematopoietic cells and acute myeloid leukemia and links to patient outcome. Eur J Haematol. 2005;75(2):106–15.

    Article  CAS  PubMed  Google Scholar 

  82. Ochsenreither S, Majeti R, Schmitt T, Stirewalt D, Keilholz U, Loeb KR, Wood B, Choi YE, Bleakley M, Warren EH, et al. Cyclin-A1 represents a new Immunogenic targetable antigen expressed in acute myeloid leukemia stem cells with characteristics of a cancer-testis antigen. Blood. 2012;119(23):5492–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Yang R, Morosetti R, Koeffler HP. Characterization of a second human Cyclin A that is highly expressed in testis and in several leukemic cell lines. Cancer Res. 1997;57(5):913–20.

    CAS  PubMed  Google Scholar 

  84. Stirewalt DL, Meshinchi S, Kopecky KJ, Fan W, Pogosova-Agadjanyan EL, Engel JH, Cronk MR, Dorcy KS, McQuary AR, Hockenbery D, et al. Identification of genes with abnormal expression changes in acute myeloid leukemia. Genes Chromosomes Cancer. 2008;47(1):8–20.

    Article  CAS  PubMed  Google Scholar 

  85. Liao C, Wang XY, Wei HQ, Li SQ, Merghoub T, Pandolfi PP, Wolgemuth DJ. Altered myelopoiesis and the development of acute myeloid leukemia in Transgenic mice overexpressing Cyclin A1. Proc Natl Acad Sci U S A. 2001;98(12):6853–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Krug U, Yasmeen A, Beger C, Baumer N, Dugas M, Berdel WE, Muller-Tidow C. Cyclin A1 regulates WT1 expression in acute myeloid leukemia cells. Int J Oncol. 2009;34(1):129–36.

    CAS  PubMed  Google Scholar 

  87. Daniele G, Simonetti G, Fusilli C, Iacobucci I, Lonoce A, Palazzo A, Lomiento M, Mammoli F, Marsano RM, Marasco E, et al. Epigenetically induced ectopic expression of UNCX impairs the proliferation and differentiation of myeloid cells. Haematologica. 2017;102(7):1204–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Li HJ, Yu PN, Huang KY, Su HY, Hsiao TH, Chang CP, Yu MH, Lin YW. NKX6.1 functions as a metastatic suppressor through epigenetic regulation of the epithelial-mesenchymal transition. Oncogene. 2016;35(17):2266–78.

    Article  CAS  PubMed  Google Scholar 

  89. Taylor KH, Pena-Hernandez KE, Davis JW, Arthur GL, Duff DJ, Shi H, Rahmatpanah FB, Sjahputera O, Caldwell CW. Large-scale CpG methylation analysis identifies novel candidate genes and reveals methylation hotspots in acute lymphoblastic leukemia. Cancer Res. 2007;67(6):2617–25.

    Article  CAS  PubMed  Google Scholar 

  90. Murao S, Stevens FJ, Ito A, Huberman E. Myeloperoxidase: a myeloid cell nuclear antigen with DNA-binding properties. Proc Natl Acad Sci U S A. 1988;85(4):1232–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR, Sultan C. Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British cooperative group. Ann Intern Med. 1985;103(4):620–5.

    Article  CAS  PubMed  Google Scholar 

  92. Weinberg OK, Dennis J, Zia H, Chen P, Chu A, Koduru P, Luu HS, Fuda F, Chen W. Adult mixed phenotype acute leukemia (MPAL): b/myeloid MPAL(isoMPO) is distinct from other MPAL subtypes. Int J Lab Hematol. 2023;45(2):170–8.

    Article  PubMed  Google Scholar 

  93. Wang WJ, Gehris BT, Rivera D, Ak S, Feng D, Wang W, Hu Z. Bone marrow-restricted aberrant myeloperoxidase expression in B-acute lymphoblastic leukemia: A diagnostic dilemma and mimicry of mixed phenotype acute leukemia. EJHaem. 2024;5(2):403–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Somerville TD, Wiseman DH, Spencer GJ, Huang X, Lynch JT, Leong HS, Williams EL, Cheesman E, Somervaille TC. Frequent derepression of the mesenchymal transcription factor gene FOXC1 in acute myeloid leukemia. Cancer Cell. 2015;28(3):329–42.

    Article  CAS  PubMed  Google Scholar 

  95. Simeoni F, Romero-Camarero I, Camera F, Amaral FMR, Sinclair OJ, Papachristou EK, Spencer GJ, Lie ALM, Lacaud G, Wiseman DH, et al. Enhancer recruitment of transcription repressors RUNX1 and TLE3 by mis-expressed FOXC1 blocks differentiation in acute myeloid leukemia. Cell Rep. 2021;36(12):109725.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Wang Y, Ma X, Huang J, Yang X, Kang M, Sun X, Li H, Wu Y, Zhang H, Zhu Y, et al. Somatic FOXC1 insertion mutation remodels the immune microenvironment and promotes the progression of childhood acute lymphoblastic leukemia. Cell Death Dis. 2022;13(5):431.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Bungartz G, Land H, Scadden DT, Emerson SG. NF-Y is necessary for hematopoietic stem cell proliferation and survival. Blood. 2012;119(6):1380–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Lawson H, van de Lagemaat LN, Barile M, Tavosanis A, Durko J, Villacreces A, Bellani A, Mapperley C, Georges E, Martins-Costa C, et al. CITED2 coordinates key hematopoietic regulatory pathways to maintain the HSC pool in both steady-state hematopoiesis and transplantation. Stem Cell Rep. 2021;16(11):2784–97.

    Article  CAS  Google Scholar 

  99. Landrette SF, Kuo YH, Hensen K, van Barjesteh S, Perrat PN, Van de Ven WJ, Delwel R, Castilla LH. Plag1 and Plagl2 are oncogenes that induce acute myeloid leukemia in Cooperation with Cbfb-MYH11. Blood. 2005;105(7):2900–7.

    Article  CAS  PubMed  Google Scholar 

  100. van der Meer D, Barthorpe S, Yang W, Lightfoot H, Hall C, Gilbert J, Francies HE, Garnett MJ. Cell model Passports-a hub for clinical, genetic and functional datasets of preclinical cancer models. Nucleic Acids Res. 2019;47(D1):D923–9.

    Article  PubMed  Google Scholar 

  101. Sugimoto K, Toyoshima H, Sakai R, Miyagawa K, Hagiwara K, Ishikawa F, Takaku F, Yazaki Y, Hirai H. Frequent mutations in the p53 gene in human myeloid leukemia cell lines. Blood. 1992;79(9):2378–83.

    Article  CAS  PubMed  Google Scholar 

  102. Donehower LA, Soussi T, Korkut A, Liu Y, Schultz A, Cardenas M, Li X, Babur O, Hsu TK, Lichtarge O, et al. Integrated analysis of TP53 gene and pathway alterations in the Cancer genome atlas. Cell Rep. 2019;28(11):3010.

    Article  CAS  PubMed  Google Scholar 

  103. Huntley S, Baggott DM, Hamilton AT, Tran-Gyamfi M, Yang S, Kim J, Gordon L, Branscomb E, Stubbs L. A comprehensive catalog of human KRAB-associated zinc finger genes: insights into the evolutionary history of a large family of transcriptional repressors. Genome Res. 2006;16(5):669–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Nabeel-Shah S, Pu S, Burns JD, Braunschweig U, Ahmed N, Burke GL, Lee H, Radovani E, Zhong G, Tang H, et al. C2H2-zinc-finger transcription factors bind RNA and function in diverse post-transcriptional regulatory processes. Mol Cell. 2024;84(19):3810–e38253810.

    Article  CAS  PubMed  Google Scholar 

  105. Lukic S, Nicolas JC, Levine AJ. The diversity of zinc-finger genes on human chromosome 19 provides an evolutionary mechanism for defense against inherited endogenous retroviruses. Cell Death Differ. 2014;21(3):381–7.

    Article  CAS  PubMed  Google Scholar 

  106. Martins F, Rosspopoff O, Carlevaro-Fita J, Forey R, Offner S, Planet E, Pulver C, Pak H, Huber F, Michaux J, et al. A cluster of evolutionarily recent KRAB zinc finger proteins protects Cancer cells from replicative Stress-Induced inflammation. Cancer Res. 2024;84(6):808–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Heyliger SO, Soliman KFA, Saulsbury MD, Reams RR. Prognostic relevance of ZNF844 and Chr 19p13.2 KRAB-Zinc finger proteins in clear cell renal carcinoma. Cancer Genomics Proteom. 2022;19(3):305–27.

    Article  CAS  Google Scholar 

  108. Kawaji H, Severin J, Lizio M, Waterhouse A, Katayama S, Irvine KM, Hume DA, Forrest AR, Suzuki H, Carninci P, et al. The FANTOM web resource: from mammalian transcriptional landscape to its dynamic regulation. Genome Biol. 2009;10(4):R40.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.

    Article  CAS  PubMed  Google Scholar 

  110. Liao Y, Smyth GK, Shi W. FeatureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30.

    Article  CAS  PubMed  Google Scholar 

  111. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Team RC. R: A language and environment for statistical computing. Vienna, Austria 2013.

  113. Hulsen T, de Vlieg J, Alkema W. BioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics. 2008;9:488.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Thomas PD, Ebert D, Muruganujan A, Mushayahama T, Albou LP, Mi H. PANTHER: making genome-scale phylogenetics accessible to all. Protein Sci. 2022;31(1):8–22.

    Article  CAS  PubMed  Google Scholar 

  115. Gene Ontology C, Aleksander SA, Balhoff J, Carbon S, Cherry JM, Drabkin HJ, Ebert D, Feuermann M, Gaudet P, Harris NL et al. The gene ontology knowledgebase in 2023. Genetics. 2023;224(1).

  116. Babicki S, Arndt D, Marcu A, Liang Y, Grant JR, Maciejewski A, Wishart DS. Heatmapper: web-enabled heat mapping for all. Nucleic Acids Res. 2016;44(W1):W147–153.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Goldman MJ, Craft B, Hastie M, Repecka K, McDade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 2020;38(6):675–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Liu H, Lorenzini PA, Zhang F, Xu S, Wong MSM, Zheng J, Roca X. Alternative splicing analysis in human monocytes and macrophages reveals MBNL1 as major regulator. Nucleic Acids Res. 2018;46(12):6069–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Wang C, Li D, Zhang L, Jiang S, Liang J, Narita Y, Hou I, Zhong Q, Zheng Z, Xiao H et al. RNA sequencing analyses of gene expression during Epstein-Barr virus infection of primary B lymphocytes. J Virol. 2019; 93(13).

Download references

Acknowledgements

We thank our colleagues Emily Clark and Philipp Voigt for critically reading the manuscript, Alison Meynert for bioinformatic advice, and Louie van de Lagemaat for curating the RNA-Seq datasets obtained by Edinburgh Genomics. We are also very grateful to Donald Dunbar and Sebastien Guizard for their bioinformatic advice during the revision of this manuscript.

Funding

Elise Georges was supported by a Grant from The Genetics Society. Douglas Vernimmen was supported by a University of Edinburgh Chancellor’s Fellowship and by Institute Strategic Grant funding to the Roslin Institute from the BBSRC [BBS/E/D/10002071] and [BBS/E/RL/230001B]. William Ho was supported by a research grant from The Kay Kendall Leukemia Fund and Kamila Malysz by the Melville Trust for care and cure cancer. Barbara Shih was partially funded by a BBSRC Core Capability Grant BB/CCG1780/1 awarded to The Roslin Institute.

Author information

Authors and Affiliations

Authors

Contributions

EG wrote original draft, WH performed data analysis, MUI isolated samples, LE performed data analysis and curation, KM performed data analysis, US made data available, ALA co-wrote manuscript, DJM wrote, reviewed and edited draft, DG performed data analysis, wrote, reviewed and edited draft, BS performed data analysis, DV supervised the work and co-wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Douglas Vernimmen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Georges, E., Ho, W., Iturritza, M.U. et al. Transcriptomic characterisation of acute myeloid leukemia cell lines bearing the same t(9;11) driver mutation reveals different molecular signatures. BMC Genomics 26, 300 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-025-11415-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-025-11415-1

Keywords