Chromosome 16 inversion, inv(16)(p13q22), is one of the most frequent chromosome abnormalities in human acute myeloid leukemia (AML), comprising almost 100% of subtype M4Eo and up to 15% of all AML. This inv(16) leads to an in-frame fusion of CBFB and MYH11 genes. CBFB-MYH11 encodes a fusion protein between CBFβ, which is an obligate partner of RUNX1 or AML1, and smooth muscle myosin heavy chain (SMMHC). Using knock-in mouse models we have previously demonstrated that Cbfb-MYH11 dominantly blocks Runx1/Cbfb function in hematopoiesis and predisposes mice to AML (requiring a second hit from ENU or retroviral mutagenesis). However, the molecular mechanism underlying these findings remains unclear. Current hypotheses, which are based on previous in vitro studies, focus on the ability of CBFβ-SMMHC to dominantly inhibit RUNX1/CBFβ and include (1) CBFβ-SMMHC binding RUNX1 with higher affinity than CBFβ; (2) CBFβ-SMMHC sequestration of RUNX1 in the cytoplasm; (3) CBFβ-SMMHC stabilization of RUNX1 by decreasing RUNX1 ubiquitination; and (4) CBFβ-SMMHC repression of RUNX1 transactivation, which is dependent on SMMHC multimerization and repressor recruitment. To test these hypotheses in vivo, we generated knock in chimeric and F1 heterozygous mice expressing CBFβ-SMMHC with C-terminal and internal deletions. One of the deletions removes only the domain responsible for high affinity binding of RUNX1 (HABD). The CBFβ-SMMHC protein with HABD deletion did not bind RUNX1 with higher affinity than CBFβ and was hypothesized to be unable to dominantly repress RUNX1. Consistent with this hypothesis, the HABD-deleted protein was less efficient in sequestering RUNX1 and caused less severe hematopoietic defects in F1 embryos than full length CBFβ-SMMHC. In contrast to mice expressing full length CBFβ-SMMHC, which develop AML only after ENU or retroviral mutagenesis, most HABD-deleted chimeric and all HABD-deleted F1 mice developed AML spontaneously shortly after birth. A larger deletion removed both the HABD and the RUNX1 stabilization domain (RSD). CBFβ-SMMHC with the HABD-RSD double deletion did not bind RUNX1 with high affinity and could not sequester RUNX1 in the cytoplasm. Mice expressing HABD-RSD double deleted CBFβ-SMMHC had normal hematopoiesis, did not develop leukemia spontaneously and developed T cell (not myeloid) malignancies after ENU treatment. These data suggest that HABD and RSD are important for CBFβ-SMMHC to block RUNX1 function and to dominantly impair hematopoiesis. The accelerated leukemogenesis associated with HABD deletion and induction of T cell malignancies in mice expressing HABD-RSD double deleted CBFβ-SMMHC strongly support the hypothesis that CBFβ-SMMHC can induce leukemia through RUNX1-inhibition-independent pathways.? ? Using the Cbfb-MYH11 knockin mouse model we have further characterized hematopoietic defects induced by Cbfb-MYH11 and discovered novel mechanisms for such defects. In the peripheral blood of Cbfb+/MYH11 embryos before embryonic day 12.5 (E12.5), we have observed the increase of morphologically immature primitive hematopoietic cells, which has not been described in the Cbfb-/- embryos. This finding raises the possibility that the Cbfb-MYH11 gene product has activities not related to the inhibition of normal Cbfb function. To address this hypothesis, we further characterized differentiation of primitive hematopoietic cells in Cbfb+/+, Cbfb+/MYH11, and Cbfb-/- embryos. By FACS, we found that over 90% of the primitive blood cells of Cbfb+/+ and Cbfb+/- E10.5 embryos showed high expression of the differentiation marker TER-119, and were negative for the progenitor marker, c-kit. In contrast, in the blood of Cbfb+/MYH11 embryos, we found increased expression of c-kit, and decreased expression of TER-119 indicating an increase of immature, progenitor cells. We also saw this delay in differentiation in the blood of E11.5 and E12.5 Cbfb+/MYH11 embryos, although the severity decreased with age. In addition, we found increased BrdU incorporation and annexin V staining in the peripheral blood of E12.5 Cbfb+/MYH11 embryos as compared to blood from Cbfb+/+ embryos. The rates of BrdU incorporation and annexin V staining in the blood of Cbfb-/- embryos were indistinguishable from wildtype. Together, these results indicate that the Cbfb-MYH11 gene product causes defects in differentiation, proliferation, and apoptosis in primitive blood that is independent of its ability to inhibit normal Cbfb activity. In order to address potential targets of this Cbfb-independent activity, we performed microarray analysis with mRNA from the peripheral blood of Cbfb+/+ and Cbfb+/MYH11 E12.5 embryos. One gene that showed significant upregulation in the blood from Cbfb+/MYH11 embryos was Csfrβ, the common β dimerization partner of the IL-3, Il-5, and GM-CSFα receptors. By FACS, we confirmed that Csfrβ (also known as CD131) is also upregulated at the protein level. In contrast, expression of Csfrβ in the blood of Cbfb-/- embryos is indistinguishable from wildtype indicating that signaling through Csfrβ and its dimerization partners could play a role in the Cbfb-independent activities of the Cbfb-MYH11 gene product. This effect is not unique to embryonic blood as adult mice that conditionally express Cbfb-MYH11 develop a population of pre-monocytic, Csfrβ+ cells in their peripheral blood as early as one week after induction of the fusion protein. In addition, leukemic cells induced in the Cbfb-MYH11 knockin mice express high levels of Csfrβ. These results indicate that expression of Csfrβ is an almost immediate consequence of Cbfb-MYH11 expression and that Csfrβ may be a direct target of the fusion protein. In conclusion we demonstrated that Cbfb-MYH11 is able to impair hematopoiesis through Cbfb-independent pathways and can affect cell proliferation and survival in addition to blockage of differentiation. We also showed evidence that Cbfb-MYH11 may directly affect signaling pathways through upregulation of Csfrβ.

Agency
National Institute of Health (NIH)
Institute
National Human Genome Research Institute (NHGRI)
Type
Intramural Research (Z01)
Project #
1Z01HG000030-13
Application #
7594296
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
13
Fiscal Year
2007
Total Cost
$1,515,419
Indirect Cost
Name
National Human Genome Research Institute
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Han, Wonshik; Nicolau, Monica; Noh, Dong-Young et al. (2010) Characterization of molecular subtypes of Korean breast cancer: an ethnically and clinically distinct population. Int J Oncol 37:51-9
Belele, Christiane L; English, Milton A; Chahal, Jagman et al. (2009) Differential requirement for Gata1 DNA binding and transactivation between primitive and definitive stages of hematopoiesis in zebrafish. Blood 114:5162-72
van de Sluis, Bart; Muller, Patricia; Duran, Karen et al. (2007) Increased activity of hypoxia-inducible factor 1 is associated with early embryonic lethality in Commd1 null mice. Mol Cell Biol 27:4142-56
Zhao, Ling; Cannons, Jennifer L; Anderson, Stacie et al. (2007) CBFB-MYH11 hinders early T-cell development and induces massive cell death in the thymus. Blood 109:3432-40
Markus, Jan; Garin, Matthew T; Bies, Juraj et al. (2007) Methylation-independent silencing of the tumor suppressor INK4b (p15) by CBFbeta-SMMHC in acute myelogenous leukemia with inv(16). Cancer Res 67:992-1000
Carella, C; Bonten, J; Sirma, S et al. (2007) MN1 overexpression is an important step in the development of inv(16) AML. Leukemia 21:1679-90
Lee, Sanggyu; Chen, Jianjun; Zhou, Guolin et al. (2006) Gene expression profiles in acute myeloid leukemia with common translocations using SAGE. Proc Natl Acad Sci U S A 103:1030-5
Kuo, Ya-Huei; Landrette, Sean F; Heilman, Susan A et al. (2006) Cbf beta-SMMHC induces distinct abnormal myeloid progenitors able to develop acute myeloid leukemia. Cancer Cell 9:57-68
Kundu, Mondira; Compton, Sheila; Garrett-Beal, Lisa et al. (2005) Runx1 deficiency predisposes mice to T-lymphoblastic lymphoma. Blood 106:3621-4
van Wijnen, Andre J; Stein, Gary S; Gergen, J Peter et al. (2004) Nomenclature for Runt-related (RUNX) proteins. Oncogene 23:4209-10

Showing the most recent 10 out of 17 publications