Insulin-like Growth Factor/Insulin-like Growth Factor 1 Receptor (IGF/IGF1R) signaling plays critical roles in regulating cell growth, proliferation and apoptosis. Both IGFs (IGF1 and IGF2) and IGF1R are commonly involved in human cancers, including leukemia. However, little is known about the role of this pathway in normal hematopoiesis and how its activation contributes to leukemogenesis. Cancer development is highly dependent on cellular context, which is determined by intrinsic genetic programs in the affected cells and by their responses to microenvironment stimuli. The long-term objective of this project is to understand the cell type-specific interplay of signaling pathways and oncogenes, which forms the basis for developing novel and more effective targeted cancer therapies and for cancer prevention. Down syndrome acute megakaryoblastic leukemia (DS-AMKL) and a related preleukemic condition called transient leukemia (DS- TL) are used as a model to study this. DS-AMKL is a unique pediatric leukemia characterized by the triad of fetal origin, trisomy 21, and somatic mutations in the hematopoietic transcription factor, GATA1 (leading to production of a shorter variant of GATA1 called GATA1s). By analyzing Gata1s knockin mice and human DS-TL/DS-AMKL cells, it was found that GATA1s leads to hyperproliferation of fetal megakaryocytic progenitors (MPs), but not their adult counterparts. Fetal MPs are highly dependent on IGF/IGF1R signaling for their proliferation and survival, whereas adult MPs are not. GATA1s mutant fetal MPs and leukemic cells are both hypersensitive to inhibition and activation of IGF/IGF1R signaling. Based on these preliminary data, it is hypothesized that GATA1 may serve as a brake to restrict the IGF/IGF1R signaling (engine)-stimulated proliferation of fetal MPs, likely through repression of E2F target genes, whereas GATA1s is defective in this function. Additional hypotheses include: overactive IGF/IGF1R signaling may cooperate with GATA1s in vivo to initiate DS-TL/DS-AMKL; and fetal and adult hematopoiesis may have different requirements for IGF/IGF1R signaling, a feature capitalized upon by certain types of leukemia to drive their malignant transformation.
Three specific aims are proposed to test these: (1) To determine the molecular mechanism by which GATA1 controls target genes of IGF/IGF1R signaling (e.g., E2F targets, including Myc) in fetal megakaryocytes; (2) To test whether constitutively active IGF/IGF1R signaling cooperates with GATA1s in vitro and in vivo to enhance the self-renewal proliferation of fetal MPs. This will be done using transgenic approaches to ectopically express IGF2 in mouse fetal liver cells; (3) To establish and to compare the in vivo role of IGF/IGF1R signaling in fetal versus adult stage hematopoiesis, by conditionally knocking out Igf1r at different stages of hematopoiesis. It is expected that these studies will significantly enhance our understanding of the role of IGF/IGF1R signaling in blood development, and of how its activation contributes to leukemia in a cellular context-dependent manner.

Public Health Relevance

This project studies a major growth-promoting pathway, the IGF/IGF1R signaling pathway, in different stages of blood development, and determines whether and how overactivity of this pathway during fetal development cooperates with a defective nuclear regulatory protein called GATA1 to initiate a unique type of leukemia found in Down syndrome children. The long-term goal is to develop novel and more effective targeted cancer therapies and cancer preventive strategies based on a better understanding of pathogenic programs leading to cancer development in a cellular context-dependent manner.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL107663-05
Application #
8823652
Study Section
Molecular and Cellular Hematology (MCH)
Program Officer
Thomas, John
Project Start
2011-06-01
Project End
2016-03-31
Budget Start
2015-04-01
Budget End
2016-03-31
Support Year
5
Fiscal Year
2015
Total Cost
$363,125
Indirect Cost
$141,500
Name
Brigham and Women's Hospital
Department
Type
DUNS #
030811269
City
Boston
State
MA
Country
United States
Zip Code
02115
Xie, Ying; Koch, Mia Lee; Zhang, Xin et al. (2017) Reduced Erg Dosage Impairs Survival of Hematopoietic Stem and Progenitor Cells. Stem Cells 35:1773-1785
Chimge, Nyam-Osor; Little, Gillian H; Baniwal, Sanjeev K et al. (2016) RUNX1 prevents oestrogen-mediated AXIN1 suppression and ?-catenin activation in ER-positive breast cancer. Nat Commun 7:10751
Tao, Luwei; van Bragt, Maaike P A; Laudadio, Elizabeth et al. (2014) Lineage tracing of mammary epithelial cells using cell-type-specific cre-expressing adenoviruses. Stem Cell Reports 2:770-9
Stankov, M V; El Khatib, M; Kumar Thakur, B et al. (2014) Histone deacetylase inhibitors induce apoptosis in myeloid leukemia by suppressing autophagy. Leukemia 28:577-88
Maroz, A; Stachorski, L; Emmrich, S et al. (2014) GATA1s induces hyperproliferation of eosinophil precursors in Down syndrome transient leukemia. Leukemia 28:1259-70
van Bragt, Maaike P A; Hu, Xin; Xie, Ying et al. (2014) RUNX1, a transcription factor mutated in breast cancer, controls the fate of ER-positive mammary luminal cells. Elife 3:e03881
Emmrich, Stephan; Rasche, Mareike; Schöning, Jennifer et al. (2014) miR-99a/100~125b tricistrons regulate hematopoietic stem and progenitor cell homeostasis by shifting the balance between TGF? and Wnt signaling. Genes Dev 28:858-74