HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

BK K+ channel blockade inhibits radiation-induced migration/brain infiltration of glioblastoma cells.

Abstract
Infiltration of the brain by glioblastoma cells reportedly requires Ca2+ signals and BK K+ channels that program and drive glioblastoma cell migration, respectively. Ionizing radiation (IR) has been shown to induce expression of the chemokine SDF-1, to alter the Ca2+ signaling, and to stimulate cell migration of glioblastoma cells. Here, we quantified fractionated IR-induced migration/brain infiltration of human glioblastoma cells in vitro and in an orthotopic mouse model and analyzed the role of SDF-1/CXCR4 signaling and BK channels. To this end, the radiation-induced migratory phenotypes of human T98G and far-red fluorescent U-87MG-Katushka glioblastoma cells were characterized by mRNA and protein expression, fura-2 Ca2+ imaging, BK patch-clamp recording and transfilter migration assay. In addition, U-87MG-Katushka cells were grown to solid glioblastomas in the right hemispheres of immunocompromised mice, fractionated irradiated (6 MV photons) with 5 × 0 or 5 × 2 Gy, and SDF-1, CXCR4, and BK protein expression by the tumor as well as glioblastoma brain infiltration was analyzed in dependence on BK channel targeting by systemic paxilline application concomitant to IR. As a result, IR stimulated SDF-1 signaling and induced migration of glioblastoma cells in vitro and in vivo. Importantly, paxilline blocked IR-induced migration in vivo. Collectively, our data demonstrate that fractionated IR of glioblastoma stimulates and BK K+ channel targeting mitigates migration and brain infiltration of glioblastoma cells in vivo. This suggests that BK channel targeting might represent a novel approach to overcome radiation-induced spreading of malignant brain tumors during radiotherapy.
AuthorsLena Edalat, Benjamin Stegen, Lukas Klumpp, Erik Haehl, Karin Schilbach, Robert Lukowski, Matthias Kühnle, Günther Bernhardt, Armin Buschauer, Daniel Zips, Peter Ruth, Stephan M Huber
JournalOncotarget (Oncotarget) Vol. 7 Issue 12 Pg. 14259-78 (Mar 22 2016) ISSN: 1949-2553 [Electronic] United States
PMID26893360 (Publication Type: Journal Article)
Chemical References
  • Biomarkers, Tumor
  • CXCL12 protein, human
  • CXCR4 protein, human
  • Chemokine CXCL12
  • Large-Conductance Calcium-Activated Potassium Channels
  • Receptors, CXCR4
Topics
  • Animals
  • Apoptosis (radiation effects)
  • Biomarkers, Tumor (metabolism)
  • Brain Neoplasms (metabolism, pathology, radiotherapy)
  • Cell Movement (radiation effects)
  • Cell Proliferation (radiation effects)
  • Chemokine CXCL12 (metabolism)
  • Female
  • Glioblastoma (metabolism, pathology, radiotherapy)
  • Humans
  • Large-Conductance Calcium-Activated Potassium Channels (antagonists & inhibitors, metabolism)
  • Male
  • Mice
  • Mice, Inbred BALB C
  • Mice, Nude
  • Radiation, Ionizing
  • Receptors, CXCR4 (metabolism)
  • Tumor Cells, Cultured
  • Xenograft Model Antitumor Assays

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: