Oncotarget

Research Papers:

Targeting stromal-induced pyruvate kinase M2 nuclear translocation impairs oxphos and prostate cancer metastatic spread

Elisa Giannoni, Maria Letizia Taddei, Andrea Morandi, Giuseppina Comito, Maura Calvani, Francesca Bianchini, Barbara Richichi, Giovanni Raugei, Nicholas Wong, Damu Tang and Paola Chiarugi _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:24061-24074. https://doi.org/10.18632/oncotarget.4448

Metrics: PDF 3071 views  |   HTML 4643 views  |   ?  


Abstract

Elisa Giannoni1, Maria Letizia Taddei1, Andrea Morandi1, Giuseppina Comito1, Maura Calvani1, Francesca Bianchini1, Barbara Richichi2, Giovanni Raugei1, Nicholas Wong3, Damu Tang3, Paola Chiarugi1

1Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy

2Department of Chemistry, University of Florence, 50019, Sesto Fiorentino, Italy

3Division of Nephrology, Department of Medicine, McMaster University, L8N4A6, Hamilton, Ontario, Canada

Correspondence to:

Paola Chiarugi, e-mail: [email protected]

Keywords: pyruvate kinase M2, cancer associated fibroblasts, epithelial-mesenchymal transition, hypoxia inducible factor-1α, prostate cancer

Received: March 05, 2015     Accepted: June 17, 2015     Published: June 27, 2015

ABSTRACT

Cancer associated fibroblasts (CAFs) are key determinants of cancer progression. In prostate carcinoma (PCa), CAFs induce epithelial-mesenchymal transition (EMT) and metabolic reprogramming of PCa cells towards oxidative phosphorylation (OXPHOS), promoting tumor growth and metastatic dissemination. We herein establish a novel role for pyruvate kinase M2 (PKM2), an established effector of Warburg-like glycolytic behavior, in OXPHOS metabolism induced by CAFs. Indeed, CAFs promote PKM2 post-translational modifications, such as cysteine oxidation and Src-dependent tyrosine phosphorylation, allowing nuclear migration of PKM2 and the formation of a trimeric complex with hypoxia inducible factor-1α (HIF-1α) and the transcriptional repressor Differentially Expressed in Chondrocytes-1 (DEC1). DEC1 recruitment is mandatory for downregulating miR205 expression, thereby fostering EMT execution and metabolic switch toward OXPHOS. Furthermore, the analysis of a cohort of PCa patients reveals a significant positive correlation between PKM2 nuclear localization and cancer aggressiveness, thereby validating our in vitro observations. Crucially, in vitro and in vivo pharmacological targeting of PKM2 nuclear translocation using DASA-58, as well as metformin, impairs metastatic dissemination of PCa cells in SCID mice. Our study indicates that impairing the metabolic tumor:stroma interplay by targeting the PKM2/OXPHOS axis, may be a valuable novel therapeutic approach in aggressive prostate carcinoma.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 4448