Oncotarget

Research Papers:

Cardiac glycosides suppress the maintenance of stemness and malignancy via inhibiting HIF-1α in human glioma stem cells

Dae-Hee Lee, Sang Cheul Oh, Amber J. Giles, Jinkyu Jung, Mark R. Gilbert and Deric M. Park _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:40233-40245. https://doi.org/10.18632/oncotarget.16714

Metrics: PDF 2143 views  |   HTML 2681 views  |   ?  


Abstract

Dae-Hee Lee1, Sang Cheul Oh1, Amber J. Giles2, Jinkyu Jung2, Mark R. Gilbert2 and Deric M. Park2

1Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea

2Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, MD, USA

Correspondence to:

Deric M. Park, email: [email protected]

Keywords: cancer stem cell, digitoxin, cardiac glycoside, hypoxia, HIF

Received: September 09, 2016     Accepted: March 11, 2017     Published: March 30, 2017

ABSTRACT

Tissue hypoxia contributes to solid tumor pathogenesis by activating a series of adaptive programs. We previously showed that hypoxia promotes the preferential expansion and maintenance of CD133 positive human glioma stem cells (GSC) in a hypoxia inducible factor 1 alpha (HIF-1α)-dependent mechanism. Here, we examined the activity of digitoxin (DT), a cardiac glycoside and a putative inhibitor of HIF-1α, on human GSC in vitro and in vivo. During hypoxic conditions (1% O2), we observed the effect of DT on the intracellular level of HIF-1α and the extracellular level of vascular endothelial growth factor (VEGF) in human GSC. We found that DT at clinically achievable concentrations, suppressed HIF-1α accumulation during hypoxic conditions in human GSC and established glioma cell lines. DT treatment also significantly attenuated hypoxia-induced expression of VEGF, a downstream target of HIF-1α. Exposure to DT also reduced hypoxia-induced activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. Furthermore, DT potently inhibited neurosphere formation, and decreased CD133 expression even at concentrations that were not overtly cytotoxic. Lastly, treatment with DT reduced GSC engraftment in an in vivo xenograft model of glioblastoma. Intraperitoneal injections of DT significantly inhibited the growth of established glioblastoma xenografts, and suppressed expression of HIF-1α and carbonic anhydrase (CA9), a surrogate marker of hypoxia. Taken together, these results suggest that DT at clinically achievable concentration functions as an inhibitor of HIF-1α, worthy of further investigations in the therapy of glioblastoma.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 16714