Your browser doesn't support javascript.
loading
Glucose deprivation elicits phenotypic plasticity via ZEB1-mediated expression of NNMT.
Kanska, Justyna; Aspuria, Paul-Joseph P; Taylor-Harding, Barbie; Spurka, Lindsay; Funari, Vincent; Orsulic, Sandra; Karlan, Beth Y; Wiedemeyer, W Ruprecht.
Affiliation
  • Kanska J; Women's Cancer Program at the Samuel Oschin Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
  • Aspuria PP; Women's Cancer Program at the Samuel Oschin Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
  • Taylor-Harding B; Women's Cancer Program at the Samuel Oschin Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
  • Spurka L; Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
  • Funari V; Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
  • Orsulic S; Women's Cancer Program at the Samuel Oschin Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
  • Karlan BY; Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA 90048, USA.
  • Wiedemeyer WR; Women's Cancer Program at the Samuel Oschin Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
Oncotarget ; 8(16): 26200-26220, 2017 Apr 18.
Article in En | MEDLINE | ID: mdl-28412735
ABSTRACT
Glucose is considered the primary energy source for all cells, and some cancers are addicted to glucose. Here, we investigated the functional consequences of chronic glucose deprivation in serous ovarian cancer cells. We found that cells resistant to glucose starvation (glucose-restricted cells) demonstrated increased metabolic plasticity that was dependent on NNMT (Nicotinamide N-methyltransferase) expression. We further show that ZEB1 induced NNMT, rendered cells resistant to glucose deprivation and recapitulated metabolic adaptations and mesenchymal gene expression observed in glucose-restricted cells. NNMT depletion reversed metabolic plasticity in glucose-restricted cells and prevented de novo formation of glucose-restricted colonies. In addition to its role in glucose independence, we found that NNMT was required for other ZEB1-induced phenotypes, such as increased migration. NNMT protein levels were also elevated in metastatic and recurrent tumors compared to matched primary carcinomas, while normal ovary and fallopian tube tissue had no detectable NNMT expression. Our studies define a novel ZEB1/NNMT signaling axis, which elicits mesenchymal gene expression, as well as phenotypic and metabolic plasticity in ovarian cancer cells upon chronic glucose starvation. Understanding the causes of cancer cell plasticity is crucial for the development of therapeutic strategies to counter intratumoral heterogeneity, acquired drug resistance and recurrence in high-grade serous ovarian cancer (HGSC).
Subject(s)
Key words

Full text: 1 Database: MEDLINE Main subject: Phenotype / Gene Expression Regulation, Neoplastic / Nicotinamide N-Methyltransferase / Zinc Finger E-box-Binding Homeobox 1 / Glucose Type of study: Prognostic_studies Limits: Female / Humans Language: En Year: 2017 Type: Article

Full text: 1 Database: MEDLINE Main subject: Phenotype / Gene Expression Regulation, Neoplastic / Nicotinamide N-Methyltransferase / Zinc Finger E-box-Binding Homeobox 1 / Glucose Type of study: Prognostic_studies Limits: Female / Humans Language: En Year: 2017 Type: Article