Histone demethylase KDM4C prevents cellular senescence in JAK2-mutated neoplasms

in: Oncology Research and Treatment (2022)
Tannert, Astrid; Ernst, Philipp; Kirchner, Laura; Schnöder, Tina M.; Huber, Nicolas; Perner, Florian; Eifert, Theresa; Hsu, Chen-Jen; Tubio-Santamaria, Nuria; Ungelenk, Martin; Hübner, Christian A.; Clement, Joachim H.; Hochhaus, Andreas; Heidel, Florian H.
Background: JAK2 V617F mutations are associated with myeloproliferative neoplasms (MPN). Current therapeutic options are ineffective to eradicate JAK2-mutated clones. Hence, MPN patients are at risk of progressing to secondary acute myeloid leukemia. We sought to determine genetic vulnerabilities that may prevent persistence and progression of JAK2-mutated clones. Methods: Genome-scale lentiviral CRISPR-Cas9 screen. Proliferation and cell growth competition assay. Transcriptome analysis using RNAsequencing. Senescence screen using SA-sGal assay, laser scanning microscopy and Western blot. In vivo KDM4C CRISPR-Cas9 knockout using NXG Mouse Xenograft. Result: Using genome-scale CRISPR-Cas9 screen KDM4C showed dependence in HEL cells irrespective of co-treatment with Ruxolitinib. Genetic deletion of KDM4C in human and murine JAK2-mutated cells led to significant loss of proliferative advantage compared to non-deleted controls. Transcriptome analyses on HEL cells following KDM4C-deletion showed significant deregulation of senescence associated genes. GSEA revealed also enriched PI3K/AKT/mTOR and NF-κB signaling. A significant increase in SA-sGal and p21 expression was detected. NXG mice with JAK2-mutated leukemia and KDM4C-KO showed significantly improved survival compared to non-targeting controls. Discussion: Upon treatment with Ruxolitinib, JAK2-mutated cells survived depending on KDM4C expression. In vivo models of AML have shown delayed disease development following deletion of KDM4C. The evasion of senescence by KDM4C in JAK2-mutated cells observed here has been described so far in melanocytic cells, and its loss was also associated with loss of H3K9me3. Conclusion: JAK2-mutated cells are dependent on KDM4C activity irrespective to JAK-inhibitor treatment. Inhibition of KDM4C results in a proliferative disadvantage of JAK2-mutant cells, which can be explained by induction of cellular senescence. Sequential therapy combining KDM4C inactivation and senolysis may represent a promising therapeutic approach for JAK2-mutated MPN.

DOI: Array

Third party cookies & scripts

This site uses cookies. For optimal performance, smooth social media and promotional use, it is recommended that you agree to third party cookies and scripts. This may involve sharing information about your use of the third-party social media, advertising and analytics website.
For more information, see privacy policy and imprint.
Which cookies & scripts and the associated processing of your personal data do you agree with?

You can change your preferences anytime by visiting privacy policy.