Tumor-associated microglia/macrophages enhance the invasion of glioma stem-like cells via TGF-β1 signaling pathway

X Ye, S Xu, Y Xin, S Yu, Y Ping, L Chen… - The Journal of …, 2012 - journals.aai.org
X Ye, S Xu, Y Xin, S Yu, Y Ping, L Chen, H Xiao, B Wang, L Yi, Q Wang, X Jiang, L Yang…
The Journal of Immunology, 2012journals.aai.org
The invasion of malignant glioma cells into the surrounding normal brain tissues is crucial
for causing the poor outcome of this tumor type. Recent studies suggest that glioma stem-
like cells (GSLCs) mediate tumor invasion. However, it is not clear whether
microenvironment factors, such as tumor-associated microglia/macrophages (TAM/Ms), also
play important roles in promoting GSLC invasion. In this study, we found that in primary
human gliomas and orthotopical transplanted syngeneic glioma, the number of TAM/Ms at …
Abstract
The invasion of malignant glioma cells into the surrounding normal brain tissues is crucial for causing the poor outcome of this tumor type. Recent studies suggest that glioma stem-like cells (GSLCs) mediate tumor invasion. However, it is not clear whether microenvironment factors, such as tumor-associated microglia/macrophages (TAM/Ms), also play important roles in promoting GSLC invasion. In this study, we found that in primary human gliomas and orthotopical transplanted syngeneic glioma, the number of TAM/Ms at the invasive front was correlated with the presence of CD133+ GSLCs, and these TAM/Ms produced high levels of TGF-β1. CD133+ GSLCs isolated from murine transplanted gliomas exhibited higher invasive potential after being cocultured with TAM/Ms, and the invasiveness was inhibited by neutralization of TGF-β1. We also found that human glioma-derived CD133+ GSLCs became more invasive upon treatment with TGF-β1. In addition, compared with CD133− committed tumor cells, CD133+ GSLCs expressed higher levels of type II TGF-β receptor (TGFBR2) mRNA and protein, and downregulation of TGFBR2 with short hairpin RNA inhibited the invasiveness of GSLCs. Mechanism studies revealed that TGF-β1 released by TAM/Ms promoted the expression of MMP-9 by GSLCs, and TGFBR2 knockdown reduced the invasiveness of these cells in vivo. These results demonstrate that TAM/Ms enhance the invasiveness of CD133+ GSLCs via the release of TGF-β1, which increases the production of MMP-9 by GSLCs. Therefore, the TGF-β1 signaling pathway is a potential therapeutic target for limiting the invasiveness of GSLCs.
journals.aai.org