Impaired ribosome biogenesis checkpoint activation induces p53-dependent MCL-1 degradation and MYC-driven lymphoma death

Author:

Domostegui Ana1ORCID,Peddigari Suresh2,Mercer Carol A.2ORCID,Iannizzotto Flavia1ORCID,Rodriguez Marta L.3,Garcia-Cajide Marta1,Amador Virginia3,Diepstraten Sarah T.45,Kelly Gemma L.45,Salazar Ramón6,Kozma Sara C.1,Kusnadi Eric P.78ORCID,Kang Jian78ORCID,Gentilella Antonio19,Pearson Richard B.781011,Thomas George112,Pelletier Joffrey1ORCID

Affiliation:

1. Laboratory of Cancer Metabolism, Molecular Mechanisms and Experimental Therapy in Oncology Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain;

2. Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH;

3. August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain;

4. The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia;

5. Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia;

6. Catalan Institute of Oncology, Molecular Mechanisms and Experimental Therapy in Oncology Program, IDIBELL, Barcelona, Spain;

7. Peter MacCallum Cancer Centre, Melbourne, VIC, Australia;

8. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia;

9. Department of Biochemistry and Physiology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain;

10. Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia;

11. Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia; and

12. Department of Physiological Sciences, Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain

Abstract

Abstract MYC-driven B-cell lymphomas are addicted to increased levels of ribosome biogenesis (RiBi), offering the potential for therapeutic intervention. However, it is unclear whether inhibition of RiBi suppresses lymphomagenesis by decreasing translational capacity and/or by p53 activation mediated by the impaired RiBi checkpoint (IRBC). Here we generated Eμ-Myc lymphoma cells expressing inducible short hairpin RNAs to either ribosomal protein L7a (RPL7a) or RPL11, the latter an essential component of the IRBC. The loss of either protein reduced RiBi, protein synthesis, and cell proliferation to similar extents. However, only RPL7a depletion induced p53-mediated apoptosis through the selective proteasomal degradation of antiapoptotic MCL-1, indicating the critical role of the IRBC in this mechanism. Strikingly, low concentrations of the US Food and Drug Administration–approved anticancer RNA polymerase I inhibitor Actinomycin D (ActD) dramatically prolonged the survival of mice harboring Trp53+/+;Eμ-Myc but not Trp53–/–;Eμ-Myc lymphomas, which provides a rationale for treating MYC-driven B-cell lymphomas with ActD. Importantly, the molecular effects of ActD on Eμ-Myc cells were recapitulated in human B-cell lymphoma cell lines, highlighting the potential for ActD as a therapeutic avenue for p53 wild-type lymphoma.

Publisher

American Society of Hematology

Subject

Cell Biology,Hematology,Immunology,Biochemistry

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