학술논문

Splicing machinery dysregulation drives glioblastoma development/aggressiveness: oncogenic role of SRSF3
Document Type
article
Source
Brain. 143(11)
Subject
Biomedical and Clinical Sciences
Oncology and Carcinogenesis
Neurosciences
Genetics
Brain Cancer
Rare Diseases
Brain Disorders
Cancer
Detection
screening and diagnosis
4.1 Discovery and preclinical testing of markers and technologies
Aetiology
2.1 Biological and endogenous factors
Alternative Splicing
Apoptosis
Biomarkers
Tumor
Brain Neoplasms
Cell Movement
Cell Proliferation
Gene Expression Regulation
Neoplastic
Gene Silencing
Glioblastoma
Humans
Neoplasm Invasiveness
Receptor
Platelet-Derived Growth Factor beta
Serine-Arginine Splicing Factors
Signal Transduction
Survival Analysis
Xenograft Model Antitumor Assays
glioblastoma
splicing machinery
SRSF3
PDGFRB pathway
antitumour therapy
Medical and Health Sciences
Psychology and Cognitive Sciences
Neurology & Neurosurgery
Biomedical and clinical sciences
Health sciences
Psychology
Language
Abstract
Glioblastomas remain the deadliest brain tumour, with a dismal ∼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.