Your browser doesn't support javascript.
loading
Pharmacological Inhibition of Membrane Signaling Mechanisms Reduces the Invasiveness of U87-MG and U251-MG Glioblastoma Cells In Vitro.
Varricchio, Alanah; Khan, Sidra; Price, Zoe K; Davis, Rohan A; Ramesh, Sunita A; Yool, Andrea J.
Afiliação
  • Varricchio A; Discipline of Physiology, School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia.
  • Khan S; Discipline of Physiology, School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia.
  • Price ZK; Obstetrics and Gynaecology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.
  • Davis RA; Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia.
  • Ramesh SA; Biological Sciences, College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia.
  • Yool AJ; Discipline of Physiology, School of Biomedicine and the Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, SA 5005, Australia.
Cancers (Basel) ; 15(4)2023 Feb 06.
Article em En | MEDLINE | ID: mdl-36831372
ABSTRACT
Impairing the motility of glioblastoma multiforme (GBM) cells is a compelling goal for new approaches to manage this highly invasive and rapidly lethal human brain cancer. Work here characterized an array of pharmacological inhibitors of membrane ion and water channels, alone and in combination, as tools for restraining glioblastoma spread in human GBM cell lines U87-MG and U251-MG. Aquaporins, AMPA glutamate receptors, and ion channel classes (shown to be upregulated in human GBM at the transcript level and linked to mechanisms of motility in other cell types) were selected as pharmacological targets for analyses. Effective compounds reduced the transwell invasiveness of U87-MG and U251-MG glioblastoma cells by 20-80% as compared with controls, without cytotoxicity. The compounds and doses used were AqB013 (14 µM); nifedipine (25 µM); amiloride (10 µM); apamin (10 µM); 4-aminopyridine (250 µM); and CNQX (6-cyano-7-nitroquinoxaline-2,3-dione; 30 µM). Invasiveness was quantified in vitro across transwell filter chambers layered with extracellular matrix. Co-application of each of the ion channel agents with the water channel inhibitor AqB013 augmented the inhibition of invasion (20 to 50% greater than either agent alone). The motility impairment achieved by co-application of pharmacological agents differed between the GBM proneural-like subtype U87-MG and classical-like subtype U251-MG, showing patterns consistent with relative levels of target channel expression (Human Protein Atlas database). In addition, two compounds, xanthurenic acid and caelestine C (from the Davis Open Access Natural Product-based Library, Griffith University QLD), were discovered to block invasion at micromolar doses in both GBM lines (IC50 values from 0.03 to 1 µM), without cytotoxicity, as measured by full mitochondrial activity under conditions matching those in transwell assays and by normal growth in spheroid assays. Mechanisms of action of these agents based on published work are likely to involve modulation of glutamatergic receptor signaling. Treating glioblastoma by the concurrent inhibition of multiple channel targets could be a powerful approach for slowing invasive cell spread without cytotoxic side effects, potentially enhancing the effectiveness of clinical interventions focused on eradicating primary tumors.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2023 Tipo de documento: Article