Acute myeloid leukemia (AML) is a heterogeneous disease with diverse gene mutations and chromosomal abnormalities. Core binding factor (CBF) leukemias, those with translocations or inversions that affect transcription factor genes RUNX1 or CBFB, account for approximately 24% of adult acute myeloid leukemia (AML) and 25% of pediatric acute lymphocytic leukemia. The encoded proteins, RUNX1 and CBFbeta, form a heterodimer to regulate gene expression, and they are both required for hematopoiesis in vertebrate animals from zebrafish to man. Extensive clinical studies have demonstrated that CBFB-MYH11 and RUNX1-ETO, the two common fusion genes in CBF leukemia, are the best biomarkers for diagnosis, prognosis, and residual disease monitoring of CBF leukemia patients. Even though CBF leukemias have better initial remission rate and better prognosis than most AML cases, current chemotherapy is associated with significant morbidity and mortality, and the long-term survival (>5 year) is only around 50-60%. Over the years we have used mouse models and a variety of research tools to characterize the CBFB-MYH11 fusion gene, determine the effect of the encoded protein, CBFbeta-SMMHC, on normal hematopoiesis, and understand the leukemogenesis process associated with the fusion gene. We have generated both conventional and conditional knock-in mouse models to study CBFB-MYH11. Using these mouse models we have demonstrated that Cbfb-MYH11 dominantly inhibits Runx1 and Cbfb function during definitive hematopoiesis, resulting in complete lack of definitive hematopoiesis in the heterozygous Cbfb-MYH11 knockin embryos. We also showed that Cbfb-MYH11 is necessary but not sufficient for leukemia, and we were able to identify cooperating genetic events in the mouse models. We have generated knock-in mouse models expressing truncated Cbfb-MYH11 to determine the importance of functional domains of CBFbeta-SMMHC. Overall our lab has been recognized in the field as the major contributor to the understanding of CBFB-MYH11 leukemia. In the last fiscal year we focused on using mouse models to study the mechanisms of leukemogenesis by CBFB-MYH11. In the first specific aim we determined if RUNX1 is important for leukemogenesis by CBFB-MYH11. Previously dominant negative inhibition of normal RUNX1 and CBFβ functions has been considered as a potential mechanism for CBFβ-SMMHC. However, recently we showed that Cbfb-MYH11 knockin embryos have primitive hematopoiesis defects that do not seem to result from RUNX1 repression (Hyde et al., Blood, 2010). Moreover, knockin mice expressing a modified CBFβ-SMMHC protein with decreased RUNX1-binding ability developed leukemia faster than those that express the full-length CBFβ-SMMHC (Kamikubo et al., Cancer Cell, 2010). These findings suggested that RUNX1-repression may not be important for leukemogenesis, and raised the possibility that CBFβ-SMMHC may induce leukemia independent of RUNX1. To test this hypothesis, we have used three Runx1 deficient models to determine if RUNX1 is required for leukemogenesis by CBFβ-SMMHC. In Cbfb+/MYH11 embryos that are also Runx1-/-, or with a semi-dominant-negative Runx1 allele, Runx1+/lz, the primitive hematopoietic defect induced by Cbfb-MYH11 was rescued, even though Runx1 deficient embryos did not have primitive hematopoietic defects. During definitive hematopoiesis in adults, CBFβ-SMMHC increased proliferation of progenitor cells and induced an abnormal pre-leukemic progenitor population. These defects were also rescued by the semi-dominant-negative allele, Runx1+/lz, or a conditional Runx1 null. Finally, leukemia development was significantly delayed in Cbfb+/MYH11, Runx1+/lz or Cbfb+/MYH11, conditional Runx1 null mice. Overall, our findings suggest that RUNX1 activity is required for Cbfb-MYH11-induced hematopoietic defects and leukemogenesis. In the second specific aim we studied the potential cooperation between CHD7 and CBFB-MYH11 for leukemogenesis. The chromodomain-helicase-DNA binding protein 7 (CHD7) interacts with RUNX1 and suppresses RUNX1 function during hematopoiesis. We hypothesized that CHD7 also plays a role in leukemogenesis by CBFB-MYH11, since CBFB-MYH11 requires RUNX1 for leukemia. To test this hypothesis, we crossed conditional Chd7 knockout mice (Chd7f/f) with Cbfb-MYH11 knockin mice to generate transgenic mice expressing Cbfbeta-SMMHC but deficient for CHD7. We found that the hematopoietic progenitor cell populations were significantly lower in these transgenic mice than control mice, which was likely due to reduced cellular proliferation. Importantly, it took much longer time for these transgenic mice to develop leukemia than the mice only expressing CBFbeta-SMMHC. We also showed that CHD7 is a partner of the RUNX1-CBFbeta-SMMHC transcription complex and that CHD7 could enhance transcription of RUNX1 and CBFbeta-SMMHCs target genes. These data indicate that CHD7 deficiency inhibits Cbfb-MYH11 induced leukemogenesis through inhibiting RUNX1 activity in regulating transcription and cellular proliferation.

Project Start
Project End
Budget Start
Budget End
Support Year
21
Fiscal Year
2015
Total Cost
Indirect Cost
Name
Human Genome Research
Department
Type
DUNS #
City
State
Country
Zip Code
Mitsuda, Yoshihide; Morita, Ken; Kashiwazaki, Gengo et al. (2018) RUNX1 positively regulates the ErbB2/HER2 signaling pathway through modulating SOS1 expression in gastric cancer cells. Sci Rep 8:6423
Zhao, L; Alkadi, H; Kwon, E M et al. (2017) The C-terminal multimerization domain is essential for leukemia development by CBF?-SMMHC in a mouse knockin model. Leukemia 31:2841-2844
Hyde, R Katherine; Liu, Paul; Friedman, Alan D (2017) RUNX1 and CBF? Mutations and Activities of Their Wild-Type Alleles in AML. Adv Exp Med Biol 962:265-282
Zhen, Tao; Kwon, Erika M; Zhao, Ling et al. (2017) Chd7 deficiency delays leukemogenesis in mice induced by Cbfb-MYH11. Blood 130:2431-2442
Jiang, Xi; Hu, Chao; Ferchen, Kyle et al. (2017) Targeted inhibition of STAT/TET1 axis as a therapeutic strategy for acute myeloid leukemia. Nat Commun 8:2099
Morita, Ken; Maeda, Shintaro; Suzuki, Kensho et al. (2017) Paradoxical enhancement of leukemogenesis in acute myeloid leukemia with moderately attenuated RUNX1 expressions. Blood Adv 1:1440-1451
Morita, Ken; Noura, Mina; Tokushige, Chieko et al. (2017) Autonomous feedback loop of RUNX1-p53-CBFB in acute myeloid leukemia cells. Sci Rep 7:16604
Sood, Raman; Kamikubo, Yasuhiko; Liu, Paul (2017) Role of RUNX1 in hematological malignancies. Blood 129:2070-2082
Morita, Ken; Suzuki, Kensho; Maeda, Shintaro et al. (2017) Genetic regulation of the RUNX transcription factor family has antitumor effects. J Clin Invest 127:2815-2828
Jiang, Xi; Hu, Chao; Arnovitz, Stephen et al. (2016) miR-22 has a potent anti-tumour role with therapeutic potential in acute myeloid leukaemia. Nat Commun 7:11452

Showing the most recent 10 out of 40 publications