Abstract

Glioblastoma multiforme (GBM), the most common and aggressive form of brain tumor, has been associated with the dysregulation of G‐protein coupled receptor signaling. Data from the Cancer Genome Atlas demonstrate remarkable upregulation of mRNA for the Gα12 protein, which is elevated in 26% of GBM patient tumors (the highest rate for all tumor samples surveyed). The Gα12 protein couples GPCRs to guanine nucleotide exchange factors (GEFs) for RhoA and was originally isolated as an oncogene. Furthermore, the subset of GPCRs that couples to Gα12 to activate RhoA includes S1P, LPA, and thrombin, inflammatory mediators that would be generated in the tumor cell environment. GPCR signaling to RhoA leads to activation of the transcriptional co‐activator YAP (Yes‐associated protein 1 gene). The role of YAP in growth of glioblastoma cells and in vivo GBM tumor development was explored in our previous studies (Yu et al. Oncogene 7(41): 5492–5507, 2018) using human glioblastoma cell lines and glioma stem cells (GSCs) derived from patient derived xenografts (PDX). We showed that shRNA mediated knockdown of YAP in GSC‐23 PDX cells significantly attenuated in vitro self‐renewal capability assessed by limiting dilution, oncogene expression, neurosphere formation, and stem cell gene expression. Orthotopic xenografts of the YAP knockdown PDX cells formed significantly smaller tumors and were of lower morbidity than wild‐type cells. Our current work examines the effects of Gα12, which would transduce effects of known and orphan GPCRs upstream that might be activated in the tumor environment to RhoA. We knocked down Gα12 using shRNA in two GBM cancer stem cell lines, GSC‐23 and HK‐281, decreasing Gα12 mRNA levels by 80% without compensatory upregulation in the expression of the homologous Gα13 protein. Loss of Gα12 attenuated GSC‐23 stemness as determined by limiting dilution assay and reduced mRNA levels for canonical stem cells genes (CCND1, NANONG, OCT4, SOX2 and NESTIN) by 35% to 60%. We are currently examining growth of GSC‐23 control and Gα12 knockdown cells following orthotopic injection into mouse brain. Histopathological findings revealed that the deletion of Gα12 reduced tumor‐derived cell migration and invasiveness. Gα12 knockdown tumors also presented with increased areas of necrosis. In vitro transwell assays confirmed that Gα12 knockdown reduced agonist‐stimulated GSC‐23 cell migration. We are exploring the role of YAP‐dependent genes (e.g. Thbs‐1, Dusp‐1, and CCN1) which were selectively decreased in tumors from PDX cells lacking Gα12 to determine their role in tumor cell migration and survival.Support or Funding InformationNIH grant CA218859

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