Triad1 is an E3 ubiquitin ligase that impairs proliferation of bone marrow progenitor cells and increases in expression during granulopoiesis. We found that Triad1 enhances ubiquitin (Ub) mediated degradation of Fgf- R1, Flt3 and av integrin in myeloid cells. Fgf-R1 and Flt3 activate phospho-inositol-3-kinase; resulting in stabilization of catenin and expression of catenin-target-genes involved in proliferation/survival. Syk is activated by av3 integrin, resulting in Pak1-dependent proliferatin and PLC?-dependent survival. Triad1 also inhibits Ub of p53 by Mdm2. We hypothesize that increasing Triad1 expression during granulopoiesis favors degradation of Fgf-R1, Flt3 and av3 integrin, but stabilizes p53; decreasing proliferation and enhancing sensitivity to apoptosis. This identifies a possible leukemia suppressor function for Triad1, since impaired activity would sustain Fgf-R1, Flt3 and av3 signaling and destabilize p53. Consistent with this, Triad1 is specifically decreased in subsets of acute myeloid leukemia (AML) with MLL-translocations (i.e. 11q23-AML) or activating FLT3 mutation. Our studies identified a mechanism for this. 11q23-AML is characterized by increased expression of a set of HOX genes, including HoxA9 and A10. We found that HoxA9 and A10 regulate transcription of ARIH2 (encoding Triad1) in a manner that requires cytokine-induced tyrosine phosphorylation of the Hox proteins. We found that constitutive activation of Shp2-PTP blocks ARIH2 transcription by preventing Hox phosphorylation. Interestingly, FLT3 mutations are frequent in Hox-over- expressing AML and activate Shp2. A myeloproliferative neoplasm (MPN) develops in mice transplanted with bone marrow expressing MLL1 fusion proteins or overexpressing HoxA9 or A10. This MPN evolves to AML over time, suggesting that Hox-overexpression is inadequate for AML in the absence of cooperating mutations. We find constitutive Shp2-activation performs this function. We hypothesize Triad1 is a leukemia suppressor that decreases proliferation/survival of cytokine-stimulated progenitor cells, and that impaired Triad1-activity facilitates disease progression/drug resistance in Hox-overexpressing AML. We will pursue this via 3 aims:
Aim 1 : Identify Triad1-regulated events that have functional implications for leukemia. The influence of Triad1 on Ub/degradation of Fgf-R1, av3, Flt3 and p53 will be investigated in vitro and in vivo.
Aim 2 : Determine if Triad1 is a leukemia suppressor in AML with Hox-overexpression and Shp2 activation. The contributions of Triad1-expression and Shp2-activation to drug resistance/disease progression will be explored in studies with murine AML models and human AML bone marrow samples.
Aim 3 : Define targetable mechanisms of leukemia suppression by Triad1. Contributions of Fgf-R1, Flt3, av integrin and p53 to leukemogenesis and drug resistance will be explored in vivo in murine models. Hox-overexpressing AML has poor prognosis and is treatment refractory. Clarifying cooperating lesions and down-stream events may suggest therapeutic targets for this subset of individuals.

Public Health Relevance

Triad1 is an E3 ubiquitin ligase that is involved in degradation of cytokine receptors during myelopoiesis and encoded by a gene that is regulated by the homeodomain transcription factors HoxA9 and HoxA10. Triad1 expression is decreased in a subset of acute myeloid leukemia (AML) that is characterized by poor prognosis and overexpression of Hox-proteins. The goal of these studies is to determine if Triad1 is a leukemia suppressor for acute myeloid leukemia with Hox-overexpression and if events down-stream from Triad1 are therapeutic targets in this AML subset.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA195642-05
Application #
9659283
Study Section
Tumor Cell Biology Study Section (TCB)
Program Officer
Klauzinska, Malgorzata
Project Start
2015-04-01
Project End
2021-03-31
Budget Start
2019-04-01
Budget End
2021-03-31
Support Year
5
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Northwestern University at Chicago
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
005436803
City
Chicago
State
IL
Country
United States
Zip Code
60611
Hu, Liping; Huang, Weiqi; Bei, Ling et al. (2018) TP53 Haploinsufficiency Rescues Emergency Granulopoiesis in FANCC-/- Mice. J Immunol 200:2129-2139
Shah, Chirag A; Bei, Ling; Wang, Hao et al. (2016) Cooperation between AlphavBeta3 integrin and the fibroblast growth factor receptor enhances proliferation of Hox-overexpressing acute myeloid leukemia cells. Oncotarget 7:54782-54794
Konieczna, Iwona M; DeLuca, Teresa A; Eklund, Elizabeth A et al. (2016) Hoxa10 null animals exhibit reduced platelet biogenesis. Br J Haematol 173:303-13
Wang, Hao; Bei, Ling; Shah, Chirag A et al. (2015) HoxA10 Terminates Emergency Granulopoiesis by Increasing Expression of Triad1. J Immunol 194:5375-87
Bei, L; Shah, C; Wang, H et al. (2014) Regulation of CDX4 gene transcription by HoxA9, HoxA10, the Mll-Ell oncogene and Shp2 during leukemogenesis. Oncogenesis 3:e135
Bei, Ling; Shah, Chirag; Wang, Hao et al. (2012) ?-Catenin activates the HOXA10 and CDX4 genes in myeloid progenitor cells. J Biol Chem 287:39589-601
Eklund, Elizabeth A; Platanias, Leonidas C (2011) Inhibition of histone deacetylase 6 as a therapeutic strategy for acute lymphocytic leukemia. Leuk Lymphoma 52:1421-2
Eklund, Elizabeth A; Raychaudhuri, Pradip; Platanias, Leonidas C (2010) Correlation between IDH2 mutations and disease status in acute myeloid leukemia. Leuk Lymphoma 51:2157-8
Eklund, Elizabeth A; Platanias, Leonidas C (2009) Screening for microRNAs in myelodysplastic syndromes. Leuk Lymphoma 50:1735-6