According to the most recent NIH Cancer Statistics Review, leukemia, a cancer of blood cells, is the ninth most common type of cancer. Acute myeloid leukemia (AML) is an aggressive form of leukemia with high lethality (~75% of patients die 5 years after being diagnosed) characterized by anemia, and excessive proliferation of abnormal myeloid progenitor cells in the bone marrow (BM). Rearrangements of the chromosomal band 3q26 portend further reduction in survival, and lead to the overexpression of the oncogene Ecotropic Viral Integration Site 1 (EVI1). The severity of 3q26 rearranged AML, the lack of in-depth understanding of the role of EVI1 in leukemia, and the inadequate therapeutic strategies interested our lab and others to investigate EVI1 associated leukemogenesis. While previous groups used transplantation of BM virally transduced to overexpress EVI1, we are the first lab to recapitulate the effects of the 3q26 rearrangements in the mouse by establishing an inducible EVI1-overexpression model, which has provided us with new insights into the mechanisms by which EVI1 induces leukemia. We concluded using our in vivo and in vitro models that EVI1 causes myeloid expansion and blocks both erythropoiesis and lymphopoiesis. As an insight to the molecular mechanism, we previously documented that EVI1 binds to GACAAGATA, which overlaps with the binding site of the master regulator of erythropoiesis GATA-1. Additionally, our data indicate that EVI1 upregulates a previously published GATA-1 blocker, PU.1, and we showed that EVI1 binds to an enhancer upstream of PU.1 encoding gene (Spi-1). Thus, we hypothesize that EVI1 blocks erythroid differentiation by two mechanisms: 1) directly competing with GATA- 1 for key genomic binding sites harboring EVI1/GATA-1 overlap motifs and 2)!binding to Spi-1 enhancer and upregulating PU.1, which suppresses GATA1 function. We will investigate both hypothesized mechanisms using cutting edge techniques including ChIP-seq, ATAC-seq, and CRISPR under the training of my sponsor and collaborator. In order to translate the proposed mechanistic insights into clinical settings and therapeutic strategies, we will perform CRISPR library screening using an in vivo model to identify genes that reverse erythropoiesis blockage associated with EVI1-overexpression. In summary, this fellowship will focus on investigating erythropoiesis blockage and resulting anemia that might explain the increased lethality associated with 3q26 rearranged leukemia, and It will unveil new therapeutic strategies that reverse the leukemia-associated anemia.

Public Health Relevance

This proposal will advance our understanding of the molecular mechanism by which the oncogene EVI1 reprograms hematopoietic differentiation to produce erythroid blockage. Additionally, it will uncover new molecular strategies that reverse anemia-associated leukemia. Together, these studies will improve patients? survival rates, and reduce lethality of leukemia, especially 3q26 rearrangements leukemia.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Predoctoral Individual National Research Service Award (F31)
Project #
1F31HL143994-01
Application #
9608260
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Chang, Henry
Project Start
2018-08-01
Project End
2020-07-31
Budget Start
2018-08-01
Budget End
2019-07-31
Support Year
1
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Rochester
Department
Pharmacology
Type
School of Medicine & Dentistry
DUNS #
041294109
City
Rochester
State
NY
Country
United States
Zip Code
14627
Ayoub, Edward; Wilson, Michael P; McGrath, Kathleen E et al. (2018) EVI1 overexpression reprograms hematopoiesis via upregulation of Spi1 transcription. Nat Commun 9:4239