Dexamethasone alleviates tumor-associated brain damage and angiogenesis

Children and adults with the most aggressive form of brain cancer, malignant gliomas or glioblastoma, often develop cerebral edema as a life-threatening complication. This complication is routinely treated with dexamethasone (DEXA), a steroidal anti-inflammatory drug with pleiotropic action profile....

Full description

Saved in:
Bibliographic Details
Published inPloS one Vol. 9; no. 4; p. e93264
Main Authors Fan, Zheng, Sehm, Tina, Rauh, Manfred, Buchfelder, Michael, Eyupoglu, Ilker Y, Savaskan, Nicolai E
Format Journal Article
LanguageEnglish
Published United States Public Library of Science 01.04.2014
Public Library of Science (PLoS)
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Children and adults with the most aggressive form of brain cancer, malignant gliomas or glioblastoma, often develop cerebral edema as a life-threatening complication. This complication is routinely treated with dexamethasone (DEXA), a steroidal anti-inflammatory drug with pleiotropic action profile. Here we show that dexamethasone reduces murine and rodent glioma tumor growth in a concentration-dependent manner. Low concentrations of DEXA are already capable of inhibiting glioma cell proliferation and at higher levels induce cell death. Further, the expression of the glutamate antiporter xCT (system Xc-; SLC7a11) and VEGFA is up-regulated after DEXA treatment indicating early cellular stress responses. However, in human gliomas DEXA exerts differential cytotoxic effects, with some human glioma cells (U251, T98G) resistant to DEXA, a finding corroborated by clinical data of dexamethasone non-responders. Moreover, DEXA-resistant gliomas did not show any xCT alterations, indicating that these gene expressions are associated with DEXA-induced cellular stress. Hence, siRNA-mediated xCT knockdown in glioma cells increased the susceptibility to DEXA. Interestingly, cell viability of primary human astrocytes and primary rodent neurons is not affected by DEXA. We further tested the pharmacological effects of DEXA on brain tissue and showed that DEXA reduces tumor-induced disturbances of the microenvironment such as neuronal cell death and tumor-induced angiogenesis. In conclusion, we demonstrate that DEXA inhibits glioma cell growth in a concentration and species-dependent manner. Further, DEXA executes neuroprotective effects in brains and reduces tumor-induced angiogenesis. Thus, our investigations reveal that DEXA acts pleiotropically and impacts tumor growth, tumor vasculature and tumor-associated brain damage.
Bibliography:Competing Interests: The authors have declared that no competing interests exist.
Conceived and designed the experiments: NES ZF IYE. Performed the experiments: ZF TS. Analyzed the data: ZF ST IYE NES. Contributed reagents/materials/analysis tools: NES ZF TS MB MR IYE. Wrote the paper: NES ZF TS MB MR IYE. Performed all cell culture experiments and proliferation assays: ZF TS. Conducted the brain slice and VOGIM experiments: ZF. Performed analytical procedures: MR ZF. Contributed to the discussion of the content and editing of the final manuscript before submission: NES ZF TS MB MR IYE.
ISSN:1932-6203
1932-6203
DOI:10.1371/journal.pone.0093264