Combined GLUT1 and OXPHOS inhibition eliminates acute myeloid leukemia cells by restraining their metabolic plasticity

Author:

Rodriguez-Zabala Maria12,Ramakrishnan Ramprasad12,Reinbach Katrin12,Ghosh Somadri12,Oburoglu Leal23ORCID,Falqués-Costa Antoni1ORCID,Bellamkonda Kishan1,Ehinger Mats4,Peña-Martínez Pablo1ORCID,Puente-Moncada Noelia1ORCID,Lilljebjörn Henrik1,Cammenga Jörg25,Pronk Cornelis Jan26ORCID,Lazarevic Vladimir5,Fioretos Thoas1,Hagström-Andersson Anna K.1ORCID,Woods Niels-Bjarne23ORCID,Järås Marcus12ORCID

Affiliation:

1. 1Division of Clinical Genetics, Lund University, Lund, Sweden

2. 2Lund Stem Cell Center, Lund University, Lund, Sweden

3. 3Division of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden

4. 4Division of Pathology, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden

5. 5Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden

6. 6Childhood Cancer Center, Skåne University Hospital, Lund, Sweden

Abstract

Abstract Acute myeloid leukemia (AML) is initiated and propagated by leukemia stem cells (LSCs), a self-renewing population of leukemia cells responsible for therapy resistance. Hence, there is an urgent need to identify new therapeutic opportunities targeting LSCs. Here, we performed an in vivo CRISPR knockout screen to identify potential therapeutic targets by interrogating cell surface dependencies of LSCs. The facilitated glucose transporter type 1 (GLUT1) emerged as a critical in vivo metabolic dependency for LSCs in a murine MLL::AF9–driven model of AML. GLUT1 disruption by genetic ablation or pharmacological inhibition led to suppression of leukemia progression and improved survival of mice that received transplantation with LSCs. Metabolic profiling revealed that Glut1 inhibition suppressed glycolysis, decreased levels of tricarboxylic acid cycle intermediates and increased the levels of amino acids. This metabolic reprogramming was accompanied by an increase in autophagic activity and apoptosis. Moreover, Glut1 disruption caused transcriptional, morphological, and immunophenotypic changes, consistent with differentiation of AML cells. Notably, dual inhibition of GLUT1 and oxidative phosphorylation (OXPHOS) exhibited synergistic antileukemic effects in the majority of tested primary AML patient samples through restraining of their metabolic plasticity. In particular, RUNX1-mutated primary leukemia cells displayed striking sensitivity to the combination treatment compared with normal CD34+ bone marrow and cord blood cells. Collectively, our study reveals a GLUT1 dependency of murine LSCs in the bone marrow microenvironment and demonstrates that dual inhibition of GLUT1 and OXPHOS is a promising therapeutic approach for AML.

Publisher

American Society of Hematology

Subject

Hematology

Cited by 1 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

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