The recurrent chromosome translocation t(12;22)(p12;q11) is associated with myeloproliferative disorders and acute myeloid leukemia (AML), and creates an MN1-TEL fusion gene in which the sequence encoding the N-terminal region of the transcription factor TEL is replaced by almost the entire coding sequence of the transcriptional coactivator MN1. We have shown that MN1-TEL is an oncogene that stimulates growth of hematopoietic cells and increases their self-renewal potential. MN1-TEL alone is insufficient to cause hematopoietic disease, but it causes AML in mice in cooperation with overexpression of the homeobox protein HOXA9, a combination also found in t(12;22) patient samples. Surprisingly, overexpression of MN1 alone in bone marrow of mice rapidly induced myeloproliferative disease (MPD). In addition, MN1 gene expression is also upregulated in samples of AML patients characterized by inversion chromosome 16 (inv16), which encodes the CBFP-MYH11 fusion protein, and in AML samples overexpressing the immortalizing gene EVIL Moreover, coexpression of CBFP-MYH11 and MN1 in mouse BM led to rapid development of AML, strongly suggesting a functional cooperation between two proteins. This suggests that the role of MN1 in myeloid malignancy in more extensive than previously known and goes well beyond its involvement in the t(12;22). Although we determined that MN1 can interact with the transcription coactivators p300 and RAC and stimulates transcription of the retinoic acid receptor in HeLa cells, we know little about the mechanistic pathways that involve MN1 and MN1-TEL in myeloid cells. Therefore, extensive analysis will be performed of MN1 and MN1-TEL transcription complexes in transformed mouse myeloid cells and direct transcriptional target genes of these transcription factors will be identified (Specific Aim 1). Mn1 knockout mice we will be used to determine the role of Mn1 in normal myelopoiesis. By using retroviral mutagenesis and introduction of directed mutations we will determine which genetic lesions cooperate with MN1-TEL and MN1 in leukemogenesis. Together these studies should provide extensive insights into the molecular mechanism and genetic pathways involved in MN1-TEL and MN1 leukemia. These studies should also provide leads for the development of novel therapies for AML involving these transcription factors. ? ? ?

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA072996-11
Application #
7278274
Study Section
Cancer Molecular Pathobiology Study Section (CAMP)
Program Officer
Mufson, R Allan
Project Start
1997-07-01
Project End
2011-06-30
Budget Start
2007-08-01
Budget End
2008-06-30
Support Year
11
Fiscal Year
2007
Total Cost
$327,240
Indirect Cost
Name
St. Jude Children's Research Hospital
Department
Type
DUNS #
067717892
City
Memphis
State
TN
Country
United States
Zip Code
38105
Numata, Masashi; Yener, Mehmet Deniz; Ekmekçi, Sema S?rma et al. (2015) High MN1 expression increases the in vitro clonogenic activity of primary mouse B-cells. Leuk Res 39:906-12
Kandilci, Ayten; Grosveld, Gerard C (2009) Reintroduction of CEBPA in MN1-overexpressing hematopoietic cells prevents their hyperproliferation and restores myeloid differentiation. Blood 114:1596-606
Grosveld, Gerard C (2009) Gamma-secretase inhibitors: Notch so bad. Nat Med 15:20-1
Grosveld, Gerard C (2007) MN1, a novel player in human AML. Blood Cells Mol Dis 39:336-9
van Wely, Karel H M; Molijn, Anco C; Buijs, Arjan et al. (2003) The MN1 oncoprotein synergizes with coactivators RAC3 and p300 in RAR-RXR-mediated transcription. Oncogene 22:699-709
Potter, M D; Buijs, A; Kreider, B et al. (2000) Identification and characterization of a new human ETS-family transcription factor, TEL2, that is expressed in hematopoietic tissues and can associate with TEL1/ETV6. Blood 95:3341-8
Van Rompaey, L; Dou, W; Buijs, A et al. (1999) Tel, a frequent target of leukemic translocations, induces cellular aggregation and influences expression of extracellular matrix components. Neoplasia 1:526-36