Myelodysplastic syndrome (MDS) is a group of stem cell malignancies most frequent among elderly patients and has a high annual incidence among hematological malignancies with approximately 14,000 new cases diagnosed every year. About 30%-40% of MDS progresses to acute myeloid leukemia (AML). However, the majority of patients succumb from infection or bleeding or treatment complications within 3-5 years. With the increase of life expectancy due to improving medical care, there has been a significant increase in the frequency of MDS and at this time the incidence of MDS (about 5 per 100,000) is higher than that of chronic myelogenous leukemia (CML) or AML in the same age group. The genetic events that drive this disease are not known, however a translocation of chromosome band 3 q26 is a recurring abnormality seen in about 10% of MDS. This genomic site contains EVI1, a gene inappropriately activated in MDS by the chromosomal rearrangement. EVI1-positive MDS patients develop fatal hematopoietic defects rapidly evolving to AML and in general their survival is less than one year. In terms of the biological mechanisms which characterize MDS, it is generally thought that the MDS cell has impaired differentiation and increased apoptosis. As the disease evolves, the cells have increased deregulated proliferation and decreased apoptosis, remain immature, and the number of blast cells increases. The lack of an animal model that reproduces MDS has strongly limited our understanding of this disease. Recent work by our group has generated a significant animal model of MDS by forcing the expression of EVI1 in murine bone marrow (BM) cells. The mice show features of MDS including hypercellular BM, BM apoptosis, and severe cytopenia and anemia. In contrast to what observed in patients, the EVI1-ppsitive murine MDS does not evolve to AML and therefore provides a unique system to understand the disease at a stage which is still potentially treatable. Preliminary studies with this model have given clear clues about molecular pathways that affect erythropoiesis, platelet formation, and cell cycling immediately after expression of EVI1 and suggest that in the EVI1-positive mice the disease progresses from a viable early stage in which the erythropoietic lineage is compromised, to an always fatal late stage characterized by anemia, apoptosis, severe cytopenia, BM apoptosis, leading to death.
The specific aims are designed to identify key steps in this progression that could be used to block the advancement of MDS and to follow the response to treatment of MDS patients. They include the identification of the molecular pathways that disrupt hematopoiesis in EVI1-positive BM cells (first aim) and of the genes that are activated or repressed by EVI1 (second aim).
The third aim will be focused on the role of arsenic trioxide in the treatment of EVI1-positive MDS and on the isolation of small molecules that inhibit EVI1.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL082935-04
Application #
7487814
Study Section
Special Emphasis Panel (ZHL1-CSR-I (S1))
Program Officer
Di Fronzo, Nancy L
Project Start
2005-09-30
Project End
2010-08-31
Budget Start
2008-09-01
Budget End
2010-08-31
Support Year
4
Fiscal Year
2008
Total Cost
$367,420
Indirect Cost
Name
University of Illinois at Chicago
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
098987217
City
Chicago
State
IL
Country
United States
Zip Code
60612
Senyuk, Vitalyi; Premanand, Kavitha; Xu, Peng et al. (2011) The oncoprotein EVI1 and the DNA methyltransferase Dnmt3 co-operate in binding and de novo methylation of target DNA. PLoS One 6:e20793
Dickstein, Jerome; Senyuk, Vitalyi; Premanand, Kavitha et al. (2010) Methylation and silencing of miRNA-124 by EVI1 and self-renewal exhaustion of hematopoietic stem cells in murine myelodysplastic syndrome. Proc Natl Acad Sci U S A 107:9783-8
Laricchia-Robbio, Leopoldo; Premanand, Kavitha; Rinaldi, Ciro R et al. (2009) EVI1 Impairs myelopoiesis by deregulation of PU.1 function. Cancer Res 69:1633-42
Senyuk, Vitalyi; Rinaldi, Ciro Roberto; Li, Donglan et al. (2009) Consistent up-regulation of Stat3 Independently of Jak2 mutations in a new murine model of essential thrombocythemia. Cancer Res 69:262-71
Cattaneo, Francesca; Nucifora, Giuseppina (2008) EVI1 recruits the histone methyltransferase SUV39H1 for transcription repression. J Cell Biochem 105:344-52
Laricchia-Robbio, Leopoldo; Nucifora, Giuseppina (2008) Significant increase of self-renewal in hematopoietic cells after forced expression of EVI1. Blood Cells Mol Dis 40:141-7
Senyuk, Vitalyi; Sinha, Kislay K; Li, Donglan et al. (2007) Repression of RUNX1 activity by EVI1: a new role of EVI1 in leukemogenesis. Cancer Res 67:5658-66
Li, Donglan; Sinha, Kislay K; Hay, Maher A et al. (2007) RUNX1-RUNX1 homodimerization modulates RUNX1 activity and function. J Biol Chem 282:13542-51
Saunthararajah, Yogen; Boccuni, Piernicola; Nucifora, Giuseppina (2006) Combinatorial action of RUNX1 and PU.1 in the regulation of hematopoiesis. Crit Rev Eukaryot Gene Expr 16:183-92
Laricchia-Robbio, Leopoldo; Fazzina, Raffaella; Li, Donglan et al. (2006) Point mutations in two EVI1 Zn fingers abolish EVI1-GATA1 interaction and allow erythroid differentiation of murine bone marrow cells. Mol Cell Biol 26:7658-66