Cargando…

A PRC2-Kdm5b axis sustains tumorigenicity of acute myeloid leukemia

Acute myeloid leukemias (AMLs) with the NUP98-NSD1 or mixed lineage leukemia (MLL) rearrangement (MLL-r) share transcriptomic profiles associated with stemness-related gene signatures and display poor prognosis. The molecular underpinnings of AML aggressiveness and stemness remain far from clear. St...

Descripción completa

Detalles Bibliográficos
Autores principales: Ren, Zhihong, Kim, Arum, Huang, Yu-Ting, Pi, Wen-Chieh, Gong, Weida, Yu, Xufen, Qi, Jun, Jin, Jian, Cai, Ling, Roeder, Robert G., Chen, Wei-Yi, Wang, Gang Greg
Formato: Online Artículo Texto
Lenguaje:English
Publicado: National Academy of Sciences 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8892512/
https://www.ncbi.nlm.nih.gov/pubmed/35217626
http://dx.doi.org/10.1073/pnas.2122940119
Descripción
Sumario:Acute myeloid leukemias (AMLs) with the NUP98-NSD1 or mixed lineage leukemia (MLL) rearrangement (MLL-r) share transcriptomic profiles associated with stemness-related gene signatures and display poor prognosis. The molecular underpinnings of AML aggressiveness and stemness remain far from clear. Studies with EZH2 enzymatic inhibitors show that polycomb repressive complex 2 (PRC2) is crucial for tumorigenicity in NUP98-NSD1(+) AML, whereas transcriptomic analysis reveal that Kdm5b, a lysine demethylase gene carrying “bivalent” chromatin domains, is directly repressed by PRC2. While ectopic expression of Kdm5b suppressed AML growth, its depletion not only promoted tumorigenicity but also attenuated anti-AML effects of PRC2 inhibitors, demonstrating a PRC2–|Kdm5b axis for AML oncogenesis. Integrated RNA sequencing (RNA-seq), chromatin immunoprecipitation followed by sequencing (ChIP-seq), and Cleavage Under Targets & Release Using Nuclease (CUT&RUN) profiling also showed that Kdm5b directly binds and represses AML stemness genes. The anti-AML effect of Kdm5b relies on its chromatin association and/or scaffold functions rather than its demethylase activity. Collectively, this study describes a molecular axis that involves histone modifiers (PRC2–|Kdm5b) for sustaining AML oncogenesis.