Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Jun;39(6):1526-1531.
doi: 10.1038/s41375-025-02593-8. Epub 2025 Apr 9.

DEK::NUP214 acts as an XPO1-dependent transcriptional activator of essential leukemia genes

Affiliations

DEK::NUP214 acts as an XPO1-dependent transcriptional activator of essential leukemia genes

Fadimana Kaya et al. Leukemia. 2025 Jun.

Abstract

The t(6;9)(p22.3;q34.1) translocation/DEK::NUP214 fusion protein defines a distinct subgroup of younger AML patients classified as a separate disease entity by the World Health Organization. DEK is a nuclear factor with multifunctional roles, including gene regulation, while its fusion partner, NUP214, plays a pivotal role in nuclear export by interacting with transport receptors such as XPO1. However, the precise mechanism by which DEK::NUP214 drives leukemia remains unclear. A comprehensive multi-omics comparison of 57 AML primary samples (including whole genome sequencing, targeted sequencing, transcriptomics, and drug screening with >500 compounds) revealed that t(6;9) cases display a selective response to XPO1 inhibitors (Selinexor & Eltanexor) and a distinct transcriptomic signature characterized by the overexpression of FOXC1 and HOX genes that are key leukemia mediators. CUT&RUN experiments demonstrated the direct binding of DEK::NUP214 to the promoters of FOXC1 and HOXA/B clusters. Strikingly, the expression of these genes and the binding of DEK::NUP214 to their regulatory regions were selectively reduced upon XPO1 inhibition in t(6;9) cells. Altogether, these results identified a novel function of DEK::NUP214 as an XPO1-dependent transcriptional activator of key leukemia drivers and provide a rationale to explore the use of XPO1 inhibitors in this patient population.

PubMed Disclaimer

Conflict of interest statement

Competing interests: The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. AML primary samples with t(6;9)/DEK::NUP214 display a unique transcriptional signature.
A RNA-seq unsupervised clustering of 57 cytogenetically poor-risk AML samples [10], including four t(6;9) cases. Different colors represent distinct cytogenetic AML subtypes. B Volcano plot representing differentially expressed genes in t(6;9) primary AML samples vs other cytogenetic AML subtypes (RNA-seq analysis integrating our cohort of 57 poor-risk cases and 691 cases from the Leucegene cohort (Leucegene project, https://leucegene.ca/) (significance determined by Kernel MCC > 0.105 and log2FoldChange (l2fc) > 0.58). Kernel MCC is a shorthand for Matthews Correlation Coefficient of a kernel density estimation (KDE)-based classifier. C mRNA expression levels HOXA and HOXB genes in t(6;9) samples compared to KMT2A-rearranged and other poor-risk AMLs (n = 57). D mRNA expression level of FOXC1 in t(6;9) patients compared to other poor-risk AMLs (n = 57). E FOXC1 protein expression across the AML cell line panel (OCI-AML3, KASUMI-1, THP-1, FKH-1, P31-FUJ, MV4;11 and K562). GAPDH was used as an endogenous control. F Bar chart showing FOXC1 silencing efficiency by RT-qPCR after shRNA/scramble transduction of FKH-1 cells. Relative expression levels were calculated using the ΔCT method, normalized to the average of GAPDH and 18S rRNA. G Representative flow cytometry plot for the apoptosis assay, with FOXC1 knockdown (KD)/scramble (scr) FKH-1 cells stained with Annexin V (X-axis) and DAPI (Y-axis). The bar graph illustrates the percentage of live cells (DAPI Annexin V), early apoptosis (DAPI Annexin V+), late apoptosis (DAPI+ Annexin V+), and dead cells (DAPI+ Annexin V). H Bar graph representing cell cycle analysis by flow cytometry based on DNA content stained with DAPI of FOXC1 KD/scr FKH-1 cells. Each group of bars indicates the percentage distribution of cells in G0/G1, S, and G2/M phases. I Bar graph showing the number of colonies of FOXC1 KD/scr FKH-1 cells after the colony forming unit (CFU) assay. J Representative images of colonies corresponding to the CFU assay. Asterisks indicate statistical significance (*: p < 0.05, **: p < 0.01, ***: p < 0.001, ****: p < 0.0001 and ‘ns’ denotes non-significant differences).
Fig. 2
Fig. 2. DEK::NUP214 specifically binds to the regulatory regions of key leukemia genes in an XPO1-dependent manner.
A In vitro drug screening using a 527-drug panel in 4 t(6;9) patients and 145 primary AML samples from other cytogenetic groups [10, 14]. The table shows the top-ranked compounds most specific and efficient for t(6;9) patients, with the XPO1 inhibitors (Selinexor and Eltanexor) highlighted in gray. B Volcano plot representing differentially expressed genes in two t(6;9) primary AML samples treated with Selinexor vs DMSO treatment control (RNA-seq results). Genes highlighted in the plot were significantly downregulated (p < 0.05) after XPO1 inhibition. C IGV genome browser plots representing the binding peaks on the FOXC1, HOXA and HOXB promoters corresponding to the CUT&RUN experiments performed in DN-HA-293T and EV-293T cells using anti-HA antibody (and anti-IgG as a control). D CUT&RUN-qPCR results show fold enrichment at the promoter regions of the FOXC1, HOXA and HOXB genes clusters in the t(6;9) AML FKH-1 and non-t(6;9) AML KASUMI-1 cells, compared to their respective IgG controls. FKH-1_NUP214 and KASUMI-1_NUP214 refer to CUT&RUN performed with the NUP214 antibody, while FKH-1_IgG and KASUMI-1_IgG indicate the corresponding controls performed with the IgG antibody. Fold enrichment was calculated relative to the FKH-1_IgG sample. Statistical significance is indicated as ***: p < 0.001, ***: p < 0.0001, and ns non-significant differences. (n = 3). E IGV genome browser plots representing the binding peaks on the FOXC1, HOXA and HOXB promoters corresponding to the CUT&RUN experiments performed in FKH-1 and KASUMI-1 cells after treatment with 200 nM Selinexor for 48 h and using anti-NUP214 antibody (and anti-IgG as a control) (Selix: treated with Selinexor; DMSO: treated with DMSO).

References

    1. Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114:937–51. - PubMed
    1. Slovak ML, Gundacker H, Bloomfield CD, Dewald G, Appelbaum FR, Larson RA, et al. A retrospective study of 69 patients with t(6;9)(p23;q34) AML emphasizes the need for a prospective, multicenter initiative for rare ‘poor prognosis’ myeloid malignancies. Leukemia. 2006;20:1295–7. - PubMed
    1. Mendes A, Fahrenkrog B. NUP214 in Leukemia: it’s more than transport. Cells. 2019;8:1–20. - PMC - PubMed
    1. Saito S, Cigdem S, Okuwaki M, Nagata K. Leukemia-associated Nup214 fusion proteins disturb the XPO1-mediated nuclear-cytoplasmic transport pathway and thereby the NF-κB signaling pathway. Mol Cell Biol. 2016;36:1820–35. - PMC - PubMed
    1. Riveiro-Falkenbach E, Soengas MS. Control of tumorigenesis and chemoresistance by the DEK oncogene. Clin Cancer Res. 2010;16:2932–8. - PMC - PubMed

MeSH terms