Oncogenic role and target properties of the lysine-specific demethylase KDM1A in chronic lymphocytic leukemia

Qu Jiang, Johanna Stachelscheid, Johannes Bloehdorn, Alicja Pacholewska, Christoph Aszyk, Francien Grotenhuijs, Tony Müller, Ozlem Onder, Prerana Wagle, Carmen D. Herling, Maria Kleppe, Zhefang Wang, Kevin R. Coombes, Sandra Robrecht, Priya S. Dalvi, Bianca Plosnita, Petra Mayer, Lynne V. Abruzzo, Janine Altmüller, Birgit GathofThorsten Persigehl, Kirsten Fischer, Billy Jebaraj, Hugh Y. Rienhoff, Rupert Ecker, Yue Zhao, Christiane J. Bruns, Stephan Stilgenbauer, Kojo Elenitoba-Johnson, Michael Hallek, Michal R. Schweiger, Margarete Odenthal, Elena Vasyutina, Marco Herling

Research output: Contribution to journalArticlepeer-review

4 Scopus citations

Abstract

In chronic lymphocytic leukemia (CLL), epigenetic alterations are considered to centrally shape the transcriptional signatures that drive disease evolution and underlie its biological and clinical subsets. Characterizations of epigenetic regulators, particularly histone-modifying enzymes, are very rudimentary in CLL. In efforts to establish effectors of the CLL-associated oncogene T-cell leukemia 1A (TCL1A), we identified here the lysine-specific histone demethylase KDM1A to interact with the TCL1A protein in B cells in conjunction with an increased catalytic activity of KDM1A. We demonstrate that KDM1A is upregulated in malignant B cells. Elevated KDM1A and associated gene expression signatures correlated with aggressive disease features and adverse clinical outcomes in a large prospective CLL trial cohort. Genetic Kdm1a knockdown in Eμ-TCL1A mice reduced leukemic burden and prolonged animal survival, accompanied by upregulated p53 and proapoptotic pathways. Genetic KDM1A depletion also affected milieu components (T, stromal, and monocytic cells), resulting in significant reductions in their capacity to support CLL-cell survival and proliferation. Integrated analyses of differential global transcriptomes (RNA sequencing) and H3K4me3 marks (chromatin immunoprecipitation sequencing) in Eμ-TCL1A vs iKdm1aKD;Eμ-TCL1A mice (confirmed in human CLL) implicate KDM1A as an oncogenic transcriptional repressor in CLL which alters histone methylation patterns with pronounced effects on defined cell death and motility pathways. Finally, pharmacologic KDM1A inhibition altered H3K4/9 target methylation and revealed marked anti–B-cell leukemic synergisms. Overall, we established the pathogenic role and effector networks of KDM1A in CLL via tumor-cell intrinsic mechanisms and its impacts in cells of the microenvironment. Our data also provide rationales to further investigate therapeutic KDM1A targeting in CLL.

Original languageEnglish (US)
Pages (from-to)44-61
Number of pages18
JournalBlood
Volume142
Issue number1
DOIs
StatePublished - Jul 6 2023

ASJC Scopus subject areas

  • Biochemistry
  • Immunology
  • Hematology
  • Cell Biology

Fingerprint

Dive into the research topics of 'Oncogenic role and target properties of the lysine-specific demethylase KDM1A in chronic lymphocytic leukemia'. Together they form a unique fingerprint.

Cite this