O-GlcNAcylation-inducing treatments inhibit estrogen receptor α expression and confer resistance to 4-OH-tamoxifen in human breast cancer-derived MCF-7 cells

PLoS One. 2013 Jul 11;8(7):e69150. doi: 10.1371/journal.pone.0069150. Print 2013.

Abstract

O-GlcNAcylation (addition of N-acetyl-glucosamine on serine or threonine residues) is a post-translational modification that regulates stability, activity or localization of cytosolic and nuclear proteins. O-linked N-acetylgluocosmaine transferase (OGT) uses UDP-GlcNAc, produced in the hexosamine biosynthetic pathway to O-GlcNacylate proteins. Removal of O-GlcNAc from proteins is catalyzed by the β-N-Acetylglucosaminidase (OGA). Recent evidences suggest that O-GlcNAcylation may affect the growth of cancer cells. However, the consequences of O-GlcNAcylation on anti-cancer therapy have not been evaluated. In this work, we studied the effects of O-GlcNAcylation on tamoxifen-induced cell death in the breast cancer-derived MCF-7 cells. Treatments that increase O-GlcNAcylation (PUGNAc and/or glucosoamine) protected MCF-7 cells from death induced by tamoxifen. In contrast, inhibition of OGT expression by siRNA potentiated the effect of tamoxifen on cell death. Since the PI-3 kinase/Akt pathway is a major regulator of cell survival, we used BRET to evaluate the effect of PUGNAc+glucosamine on PIP3 production. We observed that these treatments stimulated PIP3 production in MCF-7 cells. This effect was associated with an increase in Akt phosphorylation. However, the PI-3 kinase inhibitor LY294002, which abolished the effect of PUGNAc+glucosamine on Akt phosphorylation, did not impair the protective effects of PUGNAc+glucosamine against tamoxifen-induced cell death. These results suggest that the protective effects of O-GlcNAcylation are independent of the PI-3 kinase/Akt pathway. As tamoxifen sensitivity depends on the estrogen receptor (ERα) expression level, we evaluated the effect of PUGNAc+glucosamine on the expression of this receptor. We observed that O-GlcNAcylation-inducing treatment significantly reduced the expression of ERα mRNA and protein, suggesting a potential mechanism for the decreased tamoxifen sensitivity induced by these treatments. Therefore, our results suggest that inhibition of O-GlcNAcylation may constitute an interesting approach to improve the sensitivity of breast cancer to anti-estrogen therapy.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acetylglucosamine / analogs & derivatives
  • Acetylglucosamine / pharmacology
  • Antineoplastic Agents, Hormonal / pharmacology
  • Biosynthetic Pathways
  • Breast Neoplasms / genetics*
  • Breast Neoplasms / metabolism
  • Cell Death / drug effects
  • Drug Resistance, Neoplasm* / genetics
  • Estrogen Receptor alpha / genetics*
  • Estrogen Receptor alpha / metabolism
  • Female
  • Gene Expression Regulation, Neoplastic* / drug effects
  • Hexosamines / biosynthesis
  • Humans
  • Insulin-Like Growth Factor I / pharmacology
  • MCF-7 Cells
  • Oximes / pharmacology
  • Phenylcarbamates / pharmacology
  • Phosphatidylinositol 3-Kinases / metabolism
  • Promoter Regions, Genetic
  • Protein Processing, Post-Translational* / drug effects
  • Proto-Oncogene Proteins c-akt / metabolism
  • Selective Estrogen Receptor Modulators / pharmacology*
  • Signal Transduction / drug effects
  • Tamoxifen / analogs & derivatives
  • Tamoxifen / pharmacology*

Substances

  • Antineoplastic Agents, Hormonal
  • Estrogen Receptor alpha
  • Hexosamines
  • Oximes
  • Phenylcarbamates
  • Selective Estrogen Receptor Modulators
  • Tamoxifen
  • N-acetylglucosaminono-1,5-lactone O-(phenylcarbamoyl)oxime
  • Insulin-Like Growth Factor I
  • Phosphatidylinositol 3-Kinases
  • Proto-Oncogene Proteins c-akt
  • Acetylglucosamine

Grants and funding

SK was supported by a grant of the Higher Education Commission of Pakistan and by the French Centre National de la Recherche Scientifique, Ressources propres: Grant n° 9ADO1204/1B1BIOCE. YF, EM and CPE were supported by an Agence Nationale de la Recherche grant (ANR Genopath Diab-O-Glyc). This work was supported by the “Association pour la Recherche sur le Cancer” (Grant n° 1069) and the “Ligue contre le Cancer” (Comité de Paris, Grant n° RS10/75-46). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.