[PDF][PDF] Sema3C promotes the survival and tumorigenicity of glioma stem cells through Rac1 activation

J Man, J Shoemake, W Zhou, X Fang, Q Wu, A Rizzo… - Cell reports, 2014 - cell.com
J Man, J Shoemake, W Zhou, X Fang, Q Wu, A Rizzo, R Prayson, S Bao, JN Rich
Cell reports, 2014cell.com
Different cancer cell compartments often communicate through soluble factors to facilitate
tumor growth. Glioma stem cells (GSCs) are a subset of tumor cells that resist standard
therapy to contribute to disease progression. How GSCs employ a distinct secretory program
to communicate with and nurture each other over the nonstem tumor cell (NSTC) population
is not well defined. Here, we show that GSCs preferentially secrete Sema3C and
coordinately express PlexinA2/D1 receptors to activate Rac1/nuclear factor (NF)-κB …
Summary
Different cancer cell compartments often communicate through soluble factors to facilitate tumor growth. Glioma stem cells (GSCs) are a subset of tumor cells that resist standard therapy to contribute to disease progression. How GSCs employ a distinct secretory program to communicate with and nurture each other over the nonstem tumor cell (NSTC) population is not well defined. Here, we show that GSCs preferentially secrete Sema3C and coordinately express PlexinA2/D1 receptors to activate Rac1/nuclear factor (NF)-κB signaling in an autocrine/paracrine loop to promote their own survival. Importantly, Sema3C is not expressed in neural progenitor cells (NPCs) or NSTCs. Disruption of Sema3C induced apoptosis of GSCs, but not NPCs or NSTCs, and suppressed tumor growth in orthotopic models of glioblastoma. Introduction of activated Rac1 rescued the Sema3C knockdown phenotype in vivo. Our study supports the targeting of Sema3C to break this GSC-specific autocrine/paracrine loop in order to improve glioblastoma treatment, potentially with a high therapeutic index.
cell.com