-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

3667 Overexpression and Knockout of Mir-126 Both Promote Leukemogenesis through Targeting Distinct Gene Signaling

Oncogenes and Tumor Suppressors
Program: Oral and Poster Abstracts
Session: 603. Oncogenes and Tumor Suppressors: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Rui Su, PhD1, Zejuan Li, MD, PhD2,3, Ping Chen2, Yuanyuan Li4*, Hu Chao4*, Yungui Wang, PhD1,2*, Stephen Arnovitz2*, Miao He4*, Sandeep Gurbuxani, MBBS, PhD5, Zhixiang Zuo6*, Abdel G. Elkahloun7*, Shenglai Li4*, Hengyou Weng, PhD1,2, Hao Huang4*, Mary Beth Neilly2*, Shusheng Wang8*, Richard A. Larson, MD4, Jiwang Zhang9*, Xi Jiang, PhD1,2, Minjie Wei10*, Jie Jin, MD, PhD11, Paul P. Liu12* and Jianjun Chen, PhD1,2

1Department of Cancer Biology, University of Cincinnati, Cincinnati, OH
2Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
3University of Chicago Medical Center, Chicago, IL
4Department of Medicine, University of Chicago, Chicago
5Department of Pathology, University of Chicago, Chicago, IL
6University of Cincinnati, Cincinnati, OH
7Genetics and Molecular Biology Branch, National Human Genome Research Institute, Bethesda
8Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas
9Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood
10Department of Pharmacology, China Medical University, Shenyang, China
11Department of Hematology & Institute of Hematology, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
12Translational and Functional Genomics Branch and Microarray Core, National Human Genome Research Institute, NIH, Bethesda

MicroRNAs (miRNAs) have been implicated in the pathogenesis of various types of cancers. We reported previously that miR-126 likely functioned as an oncogene in acute myeloid leukemia (AML) (Li Z, et al. PNAS. 2008 Oct 7; 105:15535-40), which was confirmed by others (de Leeuw DC, et al. Cancer Research. 2014 Apr 1,74:2094-105; Dorrance AM, et al. Leukemia. 2015). However, it was also reported that miR-126 knockdown in normal hematopoietic stem/progenitor cells (HSPCs) resulted in expansion of long-term HSCs (Lechman ER, et al. Cell Stem Cell. 2012 Dec 7,11:799-811). Thus, the role of miR-126 in normal and malignant hematopoiesis warrants further investigation.

Here we show that miR-126 is particularly overexpressed in t(8;21) AML and increased expression of miR-126 is associated with poor prognosis in patients with t(8;21) AML. To determine the function of miR-126 in leukemogenesis, we conducted both gain- and loss-of-function in vivo studies of miR-126 in t(8;21) AML models via mouse bone marrow transplantation (BMT). Surprisingly, we found that both overexpression and knockout of miR-126 accelerated leukemogenesis by the t(8;21) fusion genes, AML1-ETO (AE) or AML1-ETO9a (AE9a; a potent oncogenic isoform of AE). For example, forced expression of miR-126 (by retroviral transduction?) significantly accelerated AE9a-mediated leukemogenesis (median survival of AE9a+miR-126 vs. AE9a: 234 days vs. 317 days, p<0.01), and AE9a-transduced miR-126-/- HSPCs (miR-126KO+AE9a) caused leukemia significantly faster than did AE9a-transduced wild-type HSPCs (AE9a) in recipients (median survival: 130 days vs. 317 days, p<0.0001).

To assess the impact of miR-126 overexpression and depletion on long-term self-renewal of t(8;21) leukemia stem/initiating cells (LSCs/LICs), we performed serial mouse BMT assays with leukemic BM cells collected from the prior generation of BMT recipients as donor cells. In the secondary mouse BMT assay, we found that the AE9a+miR-126 group showed similar leukemia development to the miR-126KO+AE9a group (median survival: 79 days vs. 81 days, p=0.22), and both groups had significantly shorter survival (p<0.01) than did the AE9a group (median survival: 116 days). Strikingly, the AE9a+miR-126 group developed leukemia significantly faster than did the miR-126KO+AE9a group in both tertiary and quaternary BMT (median survival: 28 days vs. 43 days, p=0.0001 in tertiary; 35 days vs. 49 days, p<0.0001 in quaternary), and both groups developed leukemia significantly faster (p<0.0001) than did the AE9a group. Our limiting dilution assays with mouse BM leukemic cells collected from secondary BMT recipients as donor cells showed that the estimated LSCs/LICs frequency of the AE9a+miR-126 group is significantly greater (p<0.001) than that of the miR-126KO+AE9a group (1/2,476 vs. 1/27,399; >10 fold), and both were significantly higher (p<0.01) than that (1/166,619) of the AE9a group. Furthermore, consistent with the association of increased expression of miR-126 with poor survival of t(8;21) AML patients, our in vivo and in vitro studies demonstrated that depletion or inhibition of miR-126 significantly sensitized t(8;21)-fusion-induced mouse leukemia or primary human leukemia cells to standard chemotherapy.

Our mechanistic studies indicate that miR-126 overexpression activates genes that are highly expressed in LSCs/LICs and/or primitive HSPCs through directly targeting ERRFI1 and SPRED1, which in turn activate the MAPK signaling pathway. On the other hand, miR-126 knockout activates genes that are highly expressed in committed, more differentiated hematopoietic cells and triggers the WNT/β-catenin signaling pathway by inducing FZD7 expression. We show that ERRFI1, SPRED1 and FZD7 are bona fide targets of miR-126, and their increased expression is associated favorable survival of t(8;21) AML patients. Overall, our data show that both gain- and loss-of-function of a single miRNA (e.g., miR-126) can promote tumorigenesis and enhance long-term self-renewal/progression of LSCs/LICs, through targeting distinct gene signaling and thus being associated with different biological consequences. As miR-126 depletion can sensitize AML cells to standard chemotherapy, our data also suggest that miR-126 represents a promising therapeutic target.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH