Ldb1 is required for Lmo2 oncogene–induced thymocyte self-renewal and T-cell acute lymphoblastic leukemia

Author:

Li LiQi1,Mitra Apratim2,Cui Kairong3,Zhao Bin1ORCID,Choi Seeyoung1,Lee Jan Y.1,Stamos Daniel B.1,El-Khoury Dalal1,Warzecha Claude1,Pfeifer Karl2ORCID,Hardwick Joyce45,Zhao Keji3,Venters Bryan4,Davé Utpal P.45,Love Paul E.1ORCID

Affiliation:

1. Section on Hematopoiesis and Lymphocyte Biology and

2. Section on Genomic Imprinting, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD;

3. Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD;

4. Vanderbilt University Medical Center, Nashville, TN; and

5. Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, R.L. Roudebush Veterans Affairs Hospital–Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN

Abstract

Abstract Prolonged or enhanced expression of the proto-oncogene Lmo2 is associated with a severe form of T-cell acute lymphoblastic leukemia (T-ALL), designated early T-cell precursor ALL, which is characterized by the aberrant self-renewal and subsequent oncogenic transformation of immature thymocytes. It has been suggested that Lmo2 exerts these effects by functioning as component of a multi-subunit transcription complex that includes the ubiquitous adapter Ldb1 along with b-HLH and/or GATA family transcription factors; however, direct experimental evidence for this mechanism is lacking. In this study, we investigated the importance of Ldb1 for Lmo2-induced T-ALL by conditional deletion of Ldb1 in thymocytes in an Lmo2 transgenic mouse model of T-ALL. Our results identify a critical requirement for Ldb1 in Lmo2-induced thymocyte self-renewal and thymocyte radiation resistance and for the transition of preleukemic thymocytes to overt T-ALL. Moreover, Ldb1 was also required for acquisition of the aberrant preleukemic ETP gene expression signature in immature Lmo2 transgenic thymocytes. Co-binding of Ldb1 and Lmo2 was detected at the promoters of key upregulated T-ALL driver genes (Hhex, Lyl1, and Nfe2) in preleukemic Lmo2 transgenic thymocytes, and binding of both Ldb1 and Lmo2 at these sites was reduced following Cre-mediated deletion of Ldb1. Together, these results identify a key role for Ldb1, a nonproto-oncogene, in T-ALL and support a model in which Lmo2-induced T-ALL results from failure to downregulate Ldb1/Lmo2-nucleated transcription complexes which normally function to enforce self-renewal in bone marrow hematopoietic progenitors.

Publisher

American Society of Hematology

Subject

Cell Biology,Hematology,Immunology,Biochemistry

Reference53 articles.

1. The genetics and mechanisms of T cell acute lymphoblastic leukaemia;Belver;Nat Rev Cancer,2016

2. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia;Coustan-Smith;Lancet Oncol,2009

3. T-cell acute lymphoblastic lymphoma induced in transgenic mice by the RBTN1 and RBTN2 LIM-domain genes;Fisch;Oncogene,1992

4. T cell tumours of disparate phenotype in mice transgenic for Rbtn-2;Larson;Oncogene,1994

5. LIM domain only-2 (LMO2) induces T-cell leukemia by two distinct pathways;Smith;PLoS One,2014

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3