Abstract

The aggressive and rapidly lethal brain tumor glioblastoma (GBM) is associated with profound tissue stiffening and genomic lesions in key members of the epidermal growth factor receptor (EGFR) pathway. Previous studies from our laboratory have shown that increasing microenvironmental stiffness in culture can strongly enhance glioma cell behaviors relevant to tumor progression, including proliferation, yet it has remained unclear whether stiffness and EGFR regulate proliferation through common or independent signaling mechanisms. Here we test the hypothesis that microenvironmental stiffness regulates cell cycle progression and proliferation in GBM tumor cells by altering EGFR-dependent signaling. We began by performing an unbiased reverse phase protein array screen, which revealed that stiffness modulates expression and phosphorylation of a broad range of signals relevant to proliferation, including members of the EGFR pathway. We subsequently found that culturing human GBM tumor cells on progressively stiffer culture substrates both dramatically increases proliferation and facilitates passage through the G1/S checkpoint of the cell cycle, consistent with an EGFR-dependent process. Western Blots showed that increasing microenvironmental stiffness enhances the expression and phosphorylation of EGFR and its downstream effector Akt. Pharmacological loss-of-function studies revealed that the stiffness-sensitivity of proliferation is strongly blunted by inhibition of EGFR, Akt, or PI3 kinase. Finally, we observed that stiffness strongly regulates EGFR clustering, with phosphorylated EGFR condensing into vinculin-positive focal adhesions on stiff substrates and dispersing as microenvironmental stiffness falls to physiological levels. Our findings collectively support a model in which tissue stiffening promotes GBM proliferation by spatially and biochemically amplifying EGFR signaling.

Highlights

  • Glioblastoma (GBM) is the most commonly diagnosed primary astrocytoma in the United States and is the most deadly primary brain tumor, with a median survival time of only 15 months [1]

  • This list includes integrins, which physically engage the extracellular matrix (ECM) and process mechanical inputs [11,12,13]; specific integrin subtypes have been implicated in GBM tumor initiation, with expression directly correlating with tumorigenicity [13,14,15,16]

  • We find that microenvironmental stiffness amplifies proliferation, is associated with enhanced progression through the G1/S cell cycle checkpoint, and is accompanied by increased expression and/or activity of epidermal growth factor receptor (EGFR), Akt, and phosphoinositide 3-kinase (PI3K)

Read more

Summary

Introduction

Glioblastoma (GBM) is the most commonly diagnosed primary astrocytoma in the United States and is the most deadly primary brain tumor, with a median survival time of only 15 months [1]. These findings led us to hypothesize that microenvironmental stiffness cues can regulate GBM proliferation by modulating EGFR-based signaling [33], which we tested using a combination of defined-stiffness culture substrates, proteomic screens, proliferation and cell cycle analysis, and pharmacological loss-of-function studies.

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call