-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

3662 CHD7 Deficiency Inhibits CBFB-MYH11-Induced Leukemogenesis

Oncogenes and Tumor Suppressors
Program: Oral and Poster Abstracts
Session: 603. Oncogenes and Tumor Suppressors: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Tao Zhen, PhD1*, Ling Zhao, MD, PhD1*, R. Katherine Hyde, PhD2, Lemlem Alemu1* and Pu Paul Liu, MD, PhD1

1Oncogenesis and Development Section/NHGRI, National Institutes of Health, Bethesda, MD
2University of Nebraska Medical Center, Omaha, NE

Inversion of chromosome 16 is a consistent finding in patients with acute myeloid leukemia subtype M4 with eosinophilia (AML M4Eo), which generates a CBFB-MYH11 fusion gene. Mechanistic studies showed that the interaction between CBFbeta-SMMHC (encoded by CBFB-MYH11) and RUNX1 plays a critical role in the leukemogenesis of this kind of leukemia. Recently, it was shown that chromodomain-helicase-DNA binding protein 7 (CHD7) interacts with RUNX1 and suppresses RUNX1-induced expansion of hematopoietic stem and progenitor cells. These results suggest that CHD7 also plays a role in CBFB-MYH11 induced leukemogenesis. To test this hypothesis, we crossed Cre-based conditional Chd7 knockout mice (Chd7f/f) with Cre-based conditional Cbfb-MYH11 knockin mice (Cbfb+/56M) to generate Chd7f/f, Mx1-Cre, Cbfb+/56M mice, which express Cbfbeta-SMMHC but not CHD7 after poly I:C treatment to induce Cre expression. Three weeks after poly I:C treatment, the Lin- and the Lin-/Sca1-/c-Kit+ (LK) progenitor cell populations were significantly lower in Chd7f/f, Mx1-Cre, Cbfb+/56M mice than that in Mx1-Cre, Cbfb+/56M mice. In addition, there were fewer BrdU+ cells in the Lin- and LK populations in the Chd7f/f, Mx1-Cre, Cbfb+/56M mice. Importantly, Chd7f/f, Mx1-Cre, Cbfb+/56M mice developed leukemia with a much longer latency than the Mx1-Cre, Cbfb+/56M mice. Secondary transplantation assay demostrated that spleen cells isolated from Chd7f/f, Mx1-Cre, Cbfb+/56M and Mx1-Cre, Cbfb+/56M leukemic mice were transplantable, but recipients that received spleen cells from Chd7f/f, Mx1-Cre, Cbfb+/56M leukemic mice had a longer latency than those who received cells from Mx1-Cre, Cbfb+/56M mice. The above results suggest that CHD7 deficiency slows leukemia initiation and development by inhibiting the proliferation ability of the Lin- and LK populations. Further mechanistic studies showed that CHD7 interacted with RUNX1 but not CBFbeta-SMMHC by co-Immunoprecipitation and immunofluorescence staining, suggesting that CHD7 is a partner of the RUNX1-CBFbeta-SMMHC transcription complex. In vitro luciferase assay showed that CHD7 enhanced RUNX1 and CBFbeta-SMMHC’s transcriptional activity on M-CSFR, a RUNX1 target gene. These data indicate that CHD7 is important for Cbfb-MYH11 induced leukemogenesis by facilitating RUNX1 regulation of transcription and cellular proliferation.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH