Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Cell ; 41(5): 853-870.e13, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37084735

ABSTRACT

We uncover a tumor-suppressive process in urothelium called transcriptional-translational conflict caused by deregulation of the central chromatin remodeling component ARID1A. Loss of Arid1a triggers an increase in a nexus of pro-proliferation transcripts, but a simultaneous inhibition of the eukaryotic elongation factor 2 (eEF2), which results in tumor suppression. Resolution of this conflict through enhancing translation elongation speed enables the efficient and precise synthesis of a network of poised mRNAs resulting in uncontrolled proliferation, clonogenic growth, and bladder cancer progression. We observe a similar phenomenon in patients with ARID1A-low tumors, which also exhibit increased translation elongation activity through eEF2. These findings have important clinical implications because ARID1A-deficient, but not ARID1A-proficient, tumors are sensitive to pharmacologic inhibition of protein synthesis. These discoveries reveal an oncogenic stress created by transcriptional-translational conflict and provide a unified gene expression model that unveils the importance of the crosstalk between transcription and translation in promoting cancer.


Subject(s)
Chromatin , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics
2.
JCI Insight ; 6(11)2021 06 08.
Article in English | MEDLINE | ID: mdl-34032633

ABSTRACT

Using genetically engineered mouse models, this work demonstrates that protein synthesis is essential for efficient urothelial cancer formation and growth but dispensable for bladder homeostasis. Through a candidate gene analysis for translation regulators implicated in this dependency, we discovered that phosphorylation of the translation initiation factor eIF4E at serine 209 is increased in both murine and human bladder cancer, and this phosphorylation corresponds with an increase in de novo protein synthesis. Employing an eIF4E serine 209 to alanine knock-in mutant mouse model, we show that this single posttranslational modification is critical for bladder cancer initiation and progression, despite having no impact on normal bladder tissue maintenance. Using murine and human models of advanced bladder cancer, we demonstrate that only tumors with high levels of eIF4E phosphorylation are therapeutically vulnerable to eFT508, the first clinical-grade inhibitor of MNK1 and MNK2, the upstream kinases of eIF4E. Our results show that phospho-eIF4E plays an important role in bladder cancer pathogenesis, and targeting its upstream kinases could be an effective therapeutic option for bladder cancer patients with high levels of eIF4E phosphorylation.


Subject(s)
Carcinoma, Transitional Cell/genetics , Cell Transformation, Neoplastic/genetics , Eukaryotic Initiation Factor-4E/metabolism , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , Urinary Bladder Neoplasms/genetics , Urothelium/metabolism , Animals , Butylhydroxybutylnitrosamine/toxicity , Carcinoma, Transitional Cell/chemically induced , Carcinoma, Transitional Cell/metabolism , Cell Transformation, Neoplastic/metabolism , Drug Screening Assays, Antitumor , Gene Knock-In Techniques , Homeostasis , Humans , Mice , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Ribosomal Proteins/genetics , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...