4.7 Article

Splicing machinery dysregulation drives glioblastoma development/aggressiveness: oncogenic role of SRSF3

期刊

BRAIN
卷 143, 期 -, 页码 3273-3293

出版社

OXFORD UNIV PRESS
DOI: 10.1093/brain/awaa273

关键词

glioblastoma; splicing machinery; SRSF3; PDGFRB pathway; antitumour therapy

资金

  1. Junta de Andalucia [CTS1406, BIO-0139]
  2. Spanish Ministry of Science, Innovation and Universities [FPU16/05059, FPU14/04290, PID2019-105564RB-I00]
  3. Instituto de Salud Carlos III
  4. European Union (ERDF/ESF, 'Investing in your future') [PI16/00264, PI17/02287]
  5. Spanish Ministry of Economy and Competitiveness Projects [BFU2016-80360-R, TIN2017-83445-P, SAF2013-45111-R, SAF2015-72455-EXP]
  6. CIBERobn
  7. Comunidad de Madrid Project [S2017/BMD-3770]
  8. World Cancer Research (WCR) Project [16-1177]
  9. Fundacion Botin (Spain)
  10. Samuel Oschin Comprehensive Cancer Institute (SOCCI)
  11. NIH [R33CA236687, R03NS101529]
  12. American Cancer Society grant [RSG-16-217-01-TBG]
  13. SOCCI Jack Mishkin Discovery, Prevention & Genetics, and Cancer Biology Awards

向作者/读者索取更多资源

Glioblastomas remain the deadliest brain tumour, with a dismal similar to 12-16-month survival from diagnosis. Therefore, identification of new diagnostic, prognostic and therapeutic tools to tackle glioblastomas is urgently needed. Emerging evidence indicates that the cellular machinery controlling the splicing process (spliceosome) is altered in tumours, leading to oncogenic splicing events associated with tumour progression and aggressiveness. Here, we identify for the first time a profound dysregulation in the expression of relevant spliceosome components and splicing factors (at mRNA and protein levels) in well characterized cohorts of human high-grade astrocytomas, mostly glioblastomas, compared to healthy brain control samples, being SRSF3, RBM22, PTBP1 and RBM3 able to perfectly discriminate between tumours and control samples, and between proneural-like or mesenchymal-like tumours versus control samples from different mouse models with gliomas. Results were confirmed in four additional and independent human cohorts. Silencing of SRSF3, RBM22, PTBP1 and RBM3 decreased aggressiveness parameters in vitro (e.g. proliferation, migration, tumorsphere-formation, etc.) and induced apoptosis, especially SRSF3. Remarkably, SRSF3 was correlated with patient survival and relevant tumour markers, and its silencing in vivo drastically decreased tumour development and progression, likely through a molecular/cellular mechanism involving PDGFRB and associated oncogenic signalling pathways (PI3K-AKT/ERK), which may also involve the distinct alteration of alternative splicing events of specific transcription factors controlling PDGFRB (i.e. TP73). Altogether, our results demonstrate a drastic splicing machinery-associated molecular dysregulation in glioblastomas, which could potentially be considered as a source of novel diagnostic and prognostic biomarkers as well as therapeutic targets for glioblastomas. Remarkably, SRSF3 is directly associated with glioblastoma development, progression, aggressiveness and patient survival and represents a novel potential therapeutic target to tackle this devastating pathology.

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