Oncotarget

Research Papers:

Integrin αVβ3 silencing sensitizes malignant glioma cells to temozolomide by suppression of homologous recombination repair

Markus Christmann, Kathrin Diesler, Dragomira Majhen, Christian Steigerwald, Nancy Berte, Halima Freund, Nikolina Stojanović, Bernd Kaina, Maja Osmak, Andreja Ambriović-Ristov and Maja T. Tomicic _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:27754-27771. https://doi.org/10.18632/oncotarget.10897

Metrics: PDF 2814 views  |   HTML 5311 views  |   ?  


Abstract

Markus Christmann1,*, Kathrin Diesler1, Dragomira Majhen2, Christian Steigerwald1, Nancy Berte1 Halima Freund1, Nikolina Stojanović2, Bernd Kaina1, Maja Osmak2, Andreja Ambriović-Ristov2, Maja T. Tomicic1,*

1Department of Toxicology, University Medical Center Mainz, D-55131 Mainz, Germany

2Laboratory for Cell Biology and Signaling, Division of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia

*These authors contributed equally to this work

Correspondence to:

Maja T. Tomicic, email: [email protected]

Andreja Ambriović-Ristov, email: [email protected]

Keywords: malignant gliomas, integrin αVβ3 silencing, temozolomide, homologous recombination repair, Rad51

Received: March 30, 2016     Accepted: July 19, 2016     Published: July 28, 2016

ABSTRACT

Integrins have been suggested as possible targets in anticancer therapy. Here we show that knockdown of integrins αVβ3, αVβ5, α3β1 and α4β1 and pharmacological inhibition using a cyclo-RGD integrin αVβ3/αVβ5 antagonist sensitized multiple high-grade glioma cell lines to temozolomide (TMZ)-induced cytotoxicity. The greatest effect was observed in LN229 cells upon integrin β3 silencing, which led to inhibition of the FAK/Src/Akt/NFκB signaling pathway and increased formation of γH2AX foci. The integrin β3 knockdown led to the proteasomal degradation of Rad51, reduction of Rad51 foci and reduced repair of TMZ-induced DNA double-strand breaks by impairing homologous recombination efficiency. The down-regulation of β3 in Rad51 knockdown (LN229-Rad51kd) cells neither further sensitized them to TMZ nor increased the number of γH2AX foci, confirming causality between β3 silencing and Rad51 reduction. RIP1 was found cleaved and IκBα significantly less degraded in β3-silenced/TMZ-exposed cells, indicating inactivation of NFκB signaling. The anti-apoptotic proteins Bcl-xL, survivin and XIAP were proteasomally degraded and caspase-3/-2 cleaved. Increased H2AX phosphorylation, caspase-3 cleavage, reduced Rad51 and RIP1 expression, as well as sustained IκBα expression were also observed in mouse glioma xenografts treated with the cyclo-RGD inhibitor and TMZ, confirming the molecular mechanism in vivo. Our data indicates that β3 silencing in glioma cells represents a promising strategy to sensitize high-grade gliomas to TMZ therapy.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 10897