Oncotarget

Research Papers:

Oncogenic driver FGFR3-TACC3 is dependent on membrane trafficking and ERK signaling

Katelyn N. Nelson, April N. Meyer, Clark G. Wang and Daniel J. Donoghue _

PDF  |  HTML  |  How to cite

Oncotarget. 2018; 9:34306-34319. https://doi.org/10.18632/oncotarget.26142

Metrics: PDF 1910 views  |   HTML 2809 views  |   ?  


Abstract

Katelyn N. Nelson1, April N. Meyer1, Clark G. Wang1 and Daniel J. Donoghue1,2

1Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, USA

2UCSD Moores Cancer Center and University of California San Diego, La Jolla, California, USA

Correspondence to:

Daniel J. Donoghue, email: [email protected]

Keywords: fibroblast growth factor receptor; fusion protein; glioblastoma; FGFR3-TACC3; chromosomal translocation

Abbreviations: FGFR3: Fibroblast Growth Factor Receptor 3; TACC3: Transforming Acidic Coiled-Coil Containing Protein 3

Received: August 29, 2018     Accepted: September 08, 2018     Published: September 28, 2018

ABSTRACT

Fusion proteins resulting from chromosomal translocations have been identified as oncogenic drivers in many cancers, allowing them to serve as potential drug targets in clinical practice. The genes encoding FGFRs, Fibroblast Growth Factor Receptors, are commonly involved in such translocations, with the FGFR3-TACC3 fusion protein frequently identified in many cancers, including glioblastoma, cervical cancer, bladder cancer, nasopharyngeal carcinoma, and lung adenocarcinoma among others. FGFR3-TACC3 retains the entire extracellular domain and most of the kinase domain of FGFR3, with its C-terminal domain fused to TACC3. We examine here the effects of targeting FGFR3-TACC3 to different subcellular localizations by appending either a nuclear localization signal (NLS) or a myristylation signal, or by deletion of the normal signal sequence. We demonstrate that the oncogenic effects of FGFR3-TACC3 require either entrance to the secretory pathway or plasma membrane localization, leading to overactivation of canonical MAPK/ERK pathways. We also examined the effects of different translocation breakpoints in FGFR3-TACC3, comparing fusion at TACC3 exon 11 with fusion at exon 8. Transformation resulting from FGFR3-TACC3 was not affected by association with the canonical TACC3-interacting proteins Aurora-A, clathrin, and ch-TOG. We have shown that kinase inhibitors for MEK (Trametinib) and FGFR (BGJ398) are effective in blocking cell transformation and MAPK pathway upregulation. The development of personalized medicines will be essential in treating patients who harbor oncogenic drivers such as FGFR3-TACC3.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 26142