Your browser doesn't support javascript.
loading
Developing a Tanshinone IIA Memetic by Targeting MIOS to Regulate mTORC1 and Autophagy in Glioblastoma.
Shinhmar, Sonia; Schaf, Judith; Lloyd Jones, Katie; Pardo, Olivier E; Beesley, Philip; Williams, Robin S B.
Affiliation
  • Shinhmar S; Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK.
  • Schaf J; Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK.
  • Lloyd Jones K; Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK.
  • Pardo OE; Division of Cancer, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK.
  • Beesley P; Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK.
  • Williams RSB; Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in En | MEDLINE | ID: mdl-38928292
ABSTRACT
Tanshinone IIA (T2A) is a bioactive compound that provides promise in the treatment of glioblastoma multiforme (GBM), with a range of molecular mechanisms including the inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) and the induction of autophagy. Recently, T2A has been demonstrated to function through sestrin 2 (SESN) to inhibit mTORC1 activity, but its possible impact on autophagy through this pathway has not been investigated. Here, the model system Dictyostelium discoideum and GBM cell lines were employed to investigate the cellular role of T2A in regulating SESN to inhibit mTORC1 and activate autophagy through a GATOR2 component MIOS. In D. discoideum, T2A treatment induced autophagy and inhibited mTORC1 activity, with both effects lost upon the ablation of SESN (sesn-) or MIOS (mios-). We further investigated the targeting of MIOS to reproduce this effect of T2A, where computational analysis identified 25 novel compounds predicted to strongly bind the human MIOS protein, with one compound (MIOS inhibitor 3; Mi3) reducing cell proliferation in two GBM cells. Furthermore, Mi3 specificity was demonstrated through the loss of potency in the D. discoideum mios- cells regarding cell proliferation and the induction of autophagy. In GBM cells, Mi3 treatment also reduced mTORC1 activity and induced autophagy. Thus, a potential T2A mimetic showing the inhibition of mTORC1 and induction of autophagy in GBM cells was identified.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Autophagy / Glioblastoma / Abietanes / Dictyostelium / Mechanistic Target of Rapamycin Complex 1 Limits: Humans Language: En Journal: Int J Mol Sci Year: 2024 Document type: Article Affiliation country: Reino Unido

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Autophagy / Glioblastoma / Abietanes / Dictyostelium / Mechanistic Target of Rapamycin Complex 1 Limits: Humans Language: En Journal: Int J Mol Sci Year: 2024 Document type: Article Affiliation country: Reino Unido