Skip to main content

Ca2+-dependent demethylation of phosphatase PP2Ac promotes glucose deprivation-induced cell death independently of inhibiting glycolysis.

Publication ,  Journal Article
Lee, HY; Itahana, Y; Schuechner, S; Fukuda, M; Je, HS; Ogris, E; Virshup, DM; Itahana, K
Published in: Sci Signal
January 9, 2018

Cancer cells increase glucose metabolism to support aerobic glycolysis. However, only some cancer cells are acutely sensitive to glucose withdrawal, and the underlying mechanism of this selective sensitivity is unclear. We showed that glucose deprivation initiates a cell death pathway in cancer cells that is dependent on the kinase RIPK1. Glucose withdrawal triggered rapid plasma membrane depolarization and an influx of extracellular calcium into the cell through the L-type calcium channel Cav1.3 (CACNA1D), followed by activation of the kinase CAMK1. CAMK1 and the demethylase PPME1 were required for the subsequent demethylation and inactivation of the catalytic subunit of the phosphatase PP2A (PP2Ac) and the phosphorylation of RIPK1. Plasma membrane depolarization, PP2Ac demethylation, and cell death were prevented by glucose and, unexpectedly, by its nonmetabolizable analog 2-deoxy-d-glucose (2-DG), a glycolytic inhibitor. These findings reveal a previously unknown function of glucose as a signaling molecule that protects cells from death induced by plasma membrane depolarization, independently of its role in glycolysis. Components of this cancer cell death pathway represent potential therapeutic targets against cancer.

Duke Scholars

Altmetric Attention Stats
Dimensions Citation Stats

Published In

Sci Signal

DOI

EISSN

1937-9145

Publication Date

January 9, 2018

Volume

11

Issue

512

Location

United States

Related Subject Headings

  • Tumor Cells, Cultured
  • Signal Transduction
  • Receptor-Interacting Protein Serine-Threonine Kinases
  • Protein Phosphatase 2
  • Phosphorylation
  • Neoplasms
  • Humans
  • Glycolysis
  • Glucose
  • Demethylation
 

Citation

APA
Chicago
ICMJE
MLA
NLM
Lee, H. Y., Itahana, Y., Schuechner, S., Fukuda, M., Je, H. S., Ogris, E., … Itahana, K. (2018). Ca2+-dependent demethylation of phosphatase PP2Ac promotes glucose deprivation-induced cell death independently of inhibiting glycolysis. Sci Signal, 11(512). https://doi.org/10.1126/scisignal.aam7893
Lee, Ha Yin, Yoko Itahana, Stefan Schuechner, Masahiro Fukuda, H Shawn Je, Egon Ogris, David M. Virshup, and Koji Itahana. “Ca2+-dependent demethylation of phosphatase PP2Ac promotes glucose deprivation-induced cell death independently of inhibiting glycolysis.Sci Signal 11, no. 512 (January 9, 2018). https://doi.org/10.1126/scisignal.aam7893.
Lee HY, Itahana Y, Schuechner S, Fukuda M, Je HS, Ogris E, et al. Ca2+-dependent demethylation of phosphatase PP2Ac promotes glucose deprivation-induced cell death independently of inhibiting glycolysis. Sci Signal. 2018 Jan 9;11(512).
Lee, Ha Yin, et al. “Ca2+-dependent demethylation of phosphatase PP2Ac promotes glucose deprivation-induced cell death independently of inhibiting glycolysis.Sci Signal, vol. 11, no. 512, Jan. 2018. Pubmed, doi:10.1126/scisignal.aam7893.
Lee HY, Itahana Y, Schuechner S, Fukuda M, Je HS, Ogris E, Virshup DM, Itahana K. Ca2+-dependent demethylation of phosphatase PP2Ac promotes glucose deprivation-induced cell death independently of inhibiting glycolysis. Sci Signal. 2018 Jan 9;11(512).

Published In

Sci Signal

DOI

EISSN

1937-9145

Publication Date

January 9, 2018

Volume

11

Issue

512

Location

United States

Related Subject Headings

  • Tumor Cells, Cultured
  • Signal Transduction
  • Receptor-Interacting Protein Serine-Threonine Kinases
  • Protein Phosphatase 2
  • Phosphorylation
  • Neoplasms
  • Humans
  • Glycolysis
  • Glucose
  • Demethylation