Abstract

IntroductionAmplification of the TNK2 gene in primary tumours correlates with poor prognosis. In accordance, TNK2 overexpression was shown to promote invasion of cancer cells – but the mechanism by which TNK2 mediates these effects is unresolved. TNK2 was suggested to regulate Cdc42-driven migration by activation of breast cancer antioestrogen resistance 1 (BCAR1); however, distinct from this effect is evidence for a role of TNK2 in the regulation of epidermal growth factor receptor (EGFR) endocytosis and degradation. In the present study we sought to investigate whether negative targeting of TNK2 by siRNA could be used to inhibit cancer cell invasion, to establish the contribution of its effect on the EGFR and to consequently attempt to resolve the issue of TNK2's mechanism of action.MethodsWe used siRNA to knockdown expression of TNK2 and its proposed effector BCAR1 in order to analyse the effect of this knockdown on cancer cell behaviour in vitro. We examined morphological changes using phase-contrast microscopy and immunohistochemistry. Functional parameters examined included apoptosis, proliferation, migration and invasion. We also performed flow cytometry analysis to examine EGFR cell surface expression and carried out western blot to examine the total EGFR levels.ResultsWe observed that targeting of TNK2 by siRNA in breast cancer cells resulted in distinct morphological changes characterised by a stellate appearance and an absence of protrusions at membrane edges. These changes were not recapitulated upon siRNA targeting of BCAR1. We thus hypothesised that a component of the effects induced by TNK2 may be independent of BCAR1. Consistent with the idea of an alternative mechanism for TNK2, we observed that TNK2 associates with activated EGFR in breast cancer cells in a TNK2-kinase-independent manner. Furthermore, we demonstrated that TNK2 functions to maintain EGFRs on the cell surface. We could demonstrate that the main functional effect of activating these surface EGFRs in breast cancer cells is stimulation of migration. In accordance, TNK2 silencing by siRNA led to a significant reduction in cell surface EGFR and to a concomitant decrease in the migratory and invasive capacity of breast cancer cells.ConclusionOur data suggest that TNK2 can enhance migration and invasion of breast cancer cells via preservation of EGFR expression, notwithstanding its previously reported signalling via BCAR1, explaining its oncogenic behaviour in vitro and correlation with metastatic human breast cancer in vivo.

Highlights

  • Introduction Amplification of theTNK2 gene in primary tumours correlates with poor prognosis

  • Our data suggest that TNK2 can enhance migration and invasion of breast cancer cells via preservation of epidermal growth factor receptor (EGFR) expression, notwithstanding its previously reported signalling via breast cancer antioestrogen resistance 1 (BCAR1), explaining its oncogenic behaviour in vitro and correlation with metastatic human breast cancer in vivo

  • Propoosseeddrrooleleoof fTTNNKK22inininivnavsaisoinoannadnmd emtaesttaasstisasis. (a) We propose that the effects of TNK2 on cell migration and the cytoskeleton are mediated by epidermal growth factor receptor (EGFR)

Read more

Summary

Introduction

Introduction Amplification of theTNK2 gene in primary tumours correlates with poor prognosis. TNK2 was suggested to regulate Cdc42-driven migration by activation of breast cancer antioestrogen resistance 1 (BCAR1); distinct from this effect is evidence for a role of TNK2 in the regulation of epidermal growth factor receptor (EGFR) endocytosis and degradation. The work on molecular profiling of invasive breast cancer has led to the identification of at least five distinct subtypes in which the most invasive and malignant type is entitled basal-like breast cancer [3] This molecular subtype is predominantly oestrogen receptor alpha-negative, progesterone receptor-negative, human epidermal growth factor receptor 2-negative and EGFR-positive. Developing alternative drug targets in the EGFR signalling pathway as means to treat EGFR-dependent invasive and metastatic breast cancer is imperative

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.