4.7 Article

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

期刊

BLOOD
卷 137, 期 24, 页码 3351-3364

出版社

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2020007452

关键词

-

资金

  1. National Institutes of Health, National Cancer Institute [R01-CA158768]
  2. Marie Curie Action Career Integration Grant from European Commission [PCIG10-GA-2011-304160]
  3. Asociacion Espanola Contra el Cancer [GCB14-2035]
  4. Instituto de Salud Carlos III (ISCIII)-Red Tematica de Investigacion Cooperativa en Cancer (RTICC) [RD12/0036/0049]
  5. Agencia de Gestio d'Ajuts Universitaris i de Recerca (AGAUR) [SGR 870, SGR 01743]
  6. Spanish Ministry of Science and Innovation ISCIII [PIE13/00022]
  7. Association pour la Recherche sur le Cancer [SAE20140601346]
  8. National Health and Medical Research Council of Australia [1053792, 1102609]
  9. Cancer Council of Victoria [1184873]
  10. Victorian Cancer Agency [MCRF 17028]
  11. Dyson Bequest Fellowship
  12. Formacion de Personal Investigador (FPI) grant from the Spanish Ministry of Science and Innovation [BES-2015-075840]
  13. European Regional Development Fund (FEDER)
  14. Juan de la Cierva [FJCI-2014-20422]
  15. Spanish Ministry of Science and Innovation [SAF2011-24967, SAF2014-52162-P, SAF2017-84301-P]
  16. National Health and Medical Research Council of Australia [1102609] Funding Source: NHMRC

向作者/读者索取更多资源

The study found that inhibition of ribosome biogenesis suppresses MYC-driven B-cell lymphomas, with a critical role played by the impaired RiBi checkpoint (IRBC). Additionally, the use of Actinomycin D (ActD) can significantly prolong the survival of lymphoma mice with normal p53 status.
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 E mu-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 Administrationapproved anticancer RNA polymerase I inhibitor Actinomycin D (ActD) dramatically prolonged the survival of mice harboring Trp53(+/+);E mu-Myc but not Trp53(-/-);E mu-Myc lymphomas, which provides a rationale for treatingMYC-driven B-cell lymphomaswith ActD. Importantly, the molecular effects of ActD on E mu-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.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据