Glucose-deprivation increases thyroid cancer cells sensitivity to metformin.

MedStar author(s):
Citation: Endocrine-Related Cancer. 22(6):919-32, 2015 Dec.PMID: 26362676Institution: MedStar Washington Hospital CenterDepartment: Medicine/EndocrinologyForm of publication: Journal ArticleMedline article type(s): Journal Article | Research Support, U.S. Gov't, Non-P.H.S.Subject headings: *Adenocarcinoma, Follicular/pa [Pathology] | *Carcinoma, Papillary/pa [Pathology] | *Glucose/pd [Pharmacology] | *Metformin/pd [Pharmacology] | *Thyroid Neoplasms/pa [Pathology] | Adenocarcinoma, Follicular/me [Metabolism] | AMP-Activated Protein Kinases/me [Metabolism] | Apoptosis/de [Drug Effects] | Carcinoma, Papillary/me [Metabolism] | Carrier Proteins/bi [Biosynthesis] | Carrier Proteins/ge [Genetics] | Caspases/me [Metabolism] | Cell Division/de [Drug Effects] | Cell Line, Tumor | Culture Media/ch [Chemistry] | Culture Media/pd [Pharmacology] | Deoxyglucose/pd [Pharmacology] | Drug Screening Assays, Antitumor | Drug Synergism | Endoplasmic Reticulum/me [Metabolism] | Enzyme Activation/de [Drug Effects] | Gene Expression Profiling | Gene Expression Regulation, Neoplastic/de [Drug Effects] | Glycolysis/de [Drug Effects] | Glycolysis/ge [Genetics] | Heat-Shock Proteins/bi [Biosynthesis] | Heat-Shock Proteins/ge [Genetics] | Humans | Membrane Potential, Mitochondrial/de [Drug Effects] | Membrane Proteins/bi [Biosynthesis] | Membrane Proteins/ge [Genetics] | Molecular Targeted Therapy | Neoplasm Proteins/bi [Biosynthesis] | Neoplasm Proteins/ge [Genetics] | Phosphorylation/de [Drug Effects] | Protein Processing, Post-Translational/de [Drug Effects] | Thyroid Hormones/bi [Biosynthesis] | Thyroid Hormones/ge [Genetics] | Thyroid Neoplasms/me [Metabolism]Year: 2015ISSN:
  • 1351-0088
Name of journal: Endocrine-related cancerAbstract: Metformin inhibits thyroid cancer cell growth. We sought to determine if variable glucose concentrations in medium alter the anti-cancer efficacy of metformin. Thyroid cancer cells (FTC133 and BCPAP) were cultured in high-glucose (20 mM) and low-glucose (5 mM) medium before treatment with metformin. Cell viability and apoptosis assays were performed. Expression of glycolytic genes was examined by real-time PCR, western blot, and immunostaining. Metformin inhibited cellular proliferation in high-glucose medium and induced cell death in low-glucose medium. In low-, but not in high-glucose medium, metformin induced endoplasmic reticulum stress, autophagy, and oncosis. At micromolar concentrations, metformin induced phosphorylation of AMP-activated protein kinase and blocked p-pS6 in low-glucose medium. Metformin increased the rate of glucose consumption from the medium and prompted medium acidification. Medium supplementation with glucose reversed metformin-inducible morphological changes. Treatment with an inhibitor of glycolysis (2-deoxy-d-glucose (2-DG)) increased thyroid cancer cell sensitivity to metformin. The combination of 2-DG with metformin led to cell death. Thyroid cancer cell lines were characterized by over-expression of glycolytic genes, and metformin decreased the protein level of pyruvate kinase muscle 2 (PKM2). PKM2 expression was detected in recurrent thyroid cancer tissue samples. In conclusion, we have demonstrated that the glucose concentration in the cellular milieu is a factor modulating metformin's anti-cancer activity. These data suggest that the combination of metformin with inhibitors of glycolysis could represent a new strategy for the treatment of thyroid cancer. Copyright © 2015 Society for Endocrinology.All authors: Bikas A, Boyle L, Burman KD, Costello J Jr, Hoperia V, Jensen K, Klubo-Gwiezdzinska J, Larin O, McDaniel D, Patel A, Vasko V, Wartofsky LFiscal year: FY2016Digital Object Identifier: Date added to catalog: 2016-07-15
Holdings
Item type Current library Collection Call number Status Date due Barcode
Journal Article MedStar Authors Catalog Article 26362676 Available 26362676

Metformin inhibits thyroid cancer cell growth. We sought to determine if variable glucose concentrations in medium alter the anti-cancer efficacy of metformin. Thyroid cancer cells (FTC133 and BCPAP) were cultured in high-glucose (20 mM) and low-glucose (5 mM) medium before treatment with metformin. Cell viability and apoptosis assays were performed. Expression of glycolytic genes was examined by real-time PCR, western blot, and immunostaining. Metformin inhibited cellular proliferation in high-glucose medium and induced cell death in low-glucose medium. In low-, but not in high-glucose medium, metformin induced endoplasmic reticulum stress, autophagy, and oncosis. At micromolar concentrations, metformin induced phosphorylation of AMP-activated protein kinase and blocked p-pS6 in low-glucose medium. Metformin increased the rate of glucose consumption from the medium and prompted medium acidification. Medium supplementation with glucose reversed metformin-inducible morphological changes. Treatment with an inhibitor of glycolysis (2-deoxy-d-glucose (2-DG)) increased thyroid cancer cell sensitivity to metformin. The combination of 2-DG with metformin led to cell death. Thyroid cancer cell lines were characterized by over-expression of glycolytic genes, and metformin decreased the protein level of pyruvate kinase muscle 2 (PKM2). PKM2 expression was detected in recurrent thyroid cancer tissue samples. In conclusion, we have demonstrated that the glucose concentration in the cellular milieu is a factor modulating metformin's anti-cancer activity. These data suggest that the combination of metformin with inhibitors of glycolysis could represent a new strategy for the treatment of thyroid cancer. Copyright © 2015 Society for Endocrinology.

English

Powered by Koha