Login / Signup

O-GlcNAc transferase regulates glioblastoma acetate metabolism via regulation of CDK5-dependent ACSS2 phosphorylation.

Lorela CirakuZachary A BacigalupaJing JuRebecca A MoellerGiang Le MinhRusia H LeeMichael D SmithChristina M FerrerSophie TrefelyLuke T IzzoMary T DoanWiktoria A GocalLuca D'AgostinoWenyin ShiJoshua G JacksonChristos D KatsetosKathryn E WellenNathaniel W SnyderMauricio J Reginato
Published in: Oncogene (2022)
Glioblastomas (GBMs) preferentially generate acetyl-CoA from acetate as a fuel source to promote tumor growth. O-GlcNAcylation has been shown to be elevated by increasing O-GlcNAc transferase (OGT) in many cancers and reduced O-GlcNAcylation can block cancer growth. Here, we identify a novel mechanism whereby OGT regulates acetate-dependent acetyl-CoA and lipid production by regulating phosphorylation of acetyl-CoA synthetase 2 (ACSS2) by cyclin-dependent kinase 5 (CDK5). OGT is required and sufficient for GBM cell growth and regulates acetate conversion to acetyl-CoA and lipids. Elevating O-GlcNAcylation in GBM cells increases phosphorylation of ACSS2 on Ser-267 in a CDK5-dependent manner. Importantly, we show that ACSS2 Ser-267 phosphorylation regulates its stability by reducing polyubiquitination and degradation. ACSS2 Ser-267 is critical for OGT-mediated GBM growth as overexpression of ACSS2 Ser-267 phospho-mimetic rescues growth in vitro and in vivo. Importantly, we show that pharmacologically targeting OGT and CDK5 reduces GBM growth ex vivo. Thus, the OGT/CDK5/ACSS2 pathway may be a way to target altered metabolic dependencies in brain tumors.
Keyphrases
  • cell cycle
  • fatty acid
  • protein kinase
  • cell proliferation
  • induced apoptosis
  • squamous cell carcinoma
  • cell cycle arrest
  • cancer therapy
  • young adults
  • tyrosine kinase
  • endoplasmic reticulum stress
  • pi k akt