HOXA9 expression is a common feature of human acute myelocytic leukemia (AML) and high level expression is correlated with poor prognosis. Moreover, HOXA9 overexpression immortalizes murine marrow progenitors which are arrested at a promyelocytic stage of differentiation when cultured, and causes leukemia in recipient mice following transplantation of HOXA9 expressing bone marrow. Our laboratory is now focused on developing treatment strategies based on blocking HOXA9 activity. Our recent data showed that protein kinase C (PKC) mediates phosphorylation of HOXA9 on S 204 and T205 at the N-terminus of the homeodomain. PKC phosphorylation of HOXA9 on S 204 abrogated the protein's DNA binding in vitro and the ability of endogenous HOXA9 to form cooperative DNA binding complexes with PBX. This HOXA9 phosphorylation was correlated with differentiation of myeloid cell lines and HOXA9-immortalized bone marrow cells. We hypothesize that this myeloid differentiation was in part due to PKC-mediated phosphorylation of HOXA9, which decreases the DNA binding of the homeoprotein. We have also recently determined the first set of downstream gene targets for HOXA9. We propose three aims: 1) to show a causal relationship between HOXA9 phosphorylation and myeloid differentiation, by elucidating the relationship between phosphorylation and HOXA9 pathological activity; 2) part I - determine which PKC isoform acts on HOXA9; part 2 - develop strategies to block HOXA9 transforming activity by: A) using RNAi to directly block HOXA9; B) using PKC stimulators or ser/thr phosphatase inhibitors to block HOXA9 activity in immortalized cell and leukemia models; and C) determine the X-ray crystal structure of HOXA9, to understand how phosphorylation blocks activity. This information will form the basis for future rationale drug design of HOXA9 inhibitors; and 3) to elucidate the direct molecular targets of the HOXA9 transcription factor with and without the MEIS1 and PBX cofactor proteins. The long range goal of this aim is to develop methods to block the biological activity of these downstream effector pathways. Taken together these studies should move us closer to achieving drug-based therapies for the treatment of acute myelogenous leukemias.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA080029-10
Application #
7473133
Study Section
Cancer Molecular Pathobiology Study Section (CAMP)
Program Officer
Mufson, R Allan
Project Start
1999-07-01
Project End
2010-06-30
Budget Start
2008-07-01
Budget End
2010-06-30
Support Year
10
Fiscal Year
2008
Total Cost
$316,812
Indirect Cost
Name
Northern California Institute Research & Education
Department
Type
DUNS #
613338789
City
San Francisco
State
CA
Country
United States
Zip Code
94121
Hu, Yu-Long; Fong, Stephen; Largman, Corey et al. (2010) HOXA9 regulates miR-155 in hematopoietic cells. Nucleic Acids Res 38:5472-8
Hu, Yu-Long; Fong, Steve; Ferrell, Christina et al. (2009) HOXA9 modulates its oncogenic partner Meis1 to influence normal hematopoiesis. Mol Cell Biol 29:5181-92
Shen, Wei-Fang; Hu, Yu-Long; Uttarwar, Lalita et al. (2008) MicroRNA-126 regulates HOXA9 by binding to the homeobox. Mol Cell Biol 28:4609-19
Vijapurkar, Ulka; Fischbach, Neal; Shen, Weifang et al. (2004) Protein kinase C-mediated phosphorylation of the leukemia-associated HOXA9 protein impairs its DNA binding ability and induces myeloid differentiation. Mol Cell Biol 24:3827-37
Dorsam, Sheri Tinnell; Ferrell, Christina M; Dorsam, Glenn P et al. (2004) The transcriptome of the leukemogenic homeoprotein HOXA9 in human hematopoietic cells. Blood 103:1676-84
Shen, W F; Krishnan, K; Lawrence, H J et al. (2001) The HOX homeodomain proteins block CBP histone acetyltransferase activity. Mol Cell Biol 21:7509-22