Skip Navigation

Publication Detail

Title: Radiation-induced glioblastoma signaling cascade regulates viability, apoptosis and differentiation of neural stem cells (NSC).

Authors: Ivanov, Vladimir N; Hei, Tom K

Published In Apoptosis, (2014 Dec)

Abstract: Ionizing radiation alone or in combination with chemotherapy is the main treatment modality for brain tumors including glioblastoma. Adult neurons and astrocytes demonstrate substantial radioresistance; in contrast, human neural stem cells (NSC) are highly sensitive to radiation via induction of apoptosis. Irradiation of tumor cells has the potential risk of affecting the viability and function of NSC. In this study, we have evaluated the effects of irradiated glioblastoma cells on viability, proliferation and differentiation potential of non-irradiated (bystander) NSC through radiation-induced signaling cascades. Using media transfer experiments, we demonstrated significant effects of the U87MG glioblastoma secretome after gamma-irradiation on apoptosis in non-irradiated NSC. Addition of anti-TRAIL antibody to the transferred media partially suppressed apoptosis in NSC. Furthermore, we observed a dramatic increase in the production and secretion of IL8, TGFβ1 and IL6 by irradiated glioblastoma cells, which could promote glioblastoma cell survival and modify the effects of death factors in bystander NSC. While differentiation of NSC into neurons and astrocytes occurred efficiently with the corresponding differentiation media, pretreatment of NSC for 8 h with medium from irradiated glioblastoma cells selectively suppressed the differentiation of NSC into neurons, but not into astrocytes. Exogenous IL8 and TGFβ1 increased NSC/NPC survival, but also suppressed neuronal differentiation. On the other hand, IL6 was known to positively affect survival and differentiation of astrocyte progenitors. We established a U87MG neurosphere culture that was substantially enriched by SOX2(+) and CD133(+) glioma stem-like cells (GSC). Gamma-irradiation up-regulated apoptotic death in GSC via the FasL/Fas pathway. Media transfer experiments from irradiated GSC to non-targeted NSC again demonstrated induction of apoptosis and suppression of neuronal differentiation of NSC. In summary, intercellular communication between glioblastoma cells and bystander NSC/NPC could be involved in the amplification of cancer pathology in the brain.

PubMed ID: 25273222 Exiting the NIEHS site

MeSH Terms: Apoptosis/radiation effects*; Cell Communication/radiation effects*; Cell Differentiation/radiation effects*; Cell Survival/radiation effects; Cytokines/metabolism; Embryonic Stem Cells/metabolism; Embryonic Stem Cells/pathology; Embryonic Stem Cells/radiation effects*; Fas Ligand Protein/metabolism; Gamma Rays; Glioblastoma/metabolism*; Glioblastoma/pathology; Humans; Ligands; Neural Stem Cells/metabolism; Neural Stem Cells/pathology; Neural Stem Cells/radiation effects*; Receptors, Death Domain/metabolism; Signal Transduction/radiation effects*; TNF-Related Apoptosis-Inducing Ligand/metabolism

Back
to Top