Oncotarget

Priority Research Papers:

Mitochondrial mass, a new metabolic biomarker for stem-like cancer cells: Understanding WNT/FGF-driven anabolic signaling

Rebecca Lamb, Gloria Bonuccelli, Béla Ozsvári, Maria Peiris-Pagès, Marco Fiorillo, Duncan L. Smith, Generoso Bevilacqua, Chiara Maria Mazzanti, Liam A. McDonnell, Antonio Giuseppe Naccarato, Maybo Chiu, Luke Wynne, Ubaldo E. Martinez-Outschoorn, Federica Sotgia and Michael P. Lisanti _

PDF  |  HTML  |  How to cite

Oncotarget. 2015; 6:30453-30471. https://doi.org/10.18632/oncotarget.5852

Metrics: PDF 4419 views  |   HTML 5716 views  |   ?  


Abstract

Rebecca Lamb1,2,*, Gloria Bonuccelli1,2,*, Béla Ozsvári1,2, Maria Peiris-Pagès1,2, Marco Fiorillo1,2,3, Duncan L. Smith4, Generoso Bevilacqua5,6, Chiara Maria Mazzanti5, Liam A. McDonnell5, Antonio Giuseppe Naccarato6, Maybo Chiu1,2, Luke Wynne1,2, Ubaldo E. Martinez-Outschoorn7, Federica Sotgia1,2 and Michael P. Lisanti1,2

1 The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester, UK

2 The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, Manchester, UK

3 The Department of Pharmacy, Health and Nutritional Sciences, The University of Calabria, Cosenza, Italy

4 The Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK

5 FPS – The Pisa Science Foundation, Pisa, Italy

6 Department of Pathology, Pisa University Hospital, Pisa, Italy

7 The Sidney Kimmel Cancer Center, Philadelphia, PA, USA

* These authors should be considered as co-first authors

Correspondence to:

Michael P. Lisanti, email:

Federica Sotgia, email:

Keywords: mitochondria, MitoTracker, MMTV, WNT, FGF

Received: July 21, 2015 Accepted: August 22, 2015 Published: September 28, 2015

Abstract

Here, we developed an isogenic cell model of “stemness” to facilitate protein biomarker discovery in breast cancer. For this purpose, we used knowledge gained previously from the study of the mouse mammary tumor virus (MMTV). MMTV initiates mammary tumorigenesis in mice by promoter insertion adjacent to two main integration sites, namely Int-1 (Wnt1) and Int-2 (Fgf3), which ultimately activates Wnt/β-catenin signaling, driving the propagation of mammary cancer stem cells (CSCs). Thus, to develop a humanized model of MMTV signaling, we over-expressed WNT1 and FGF3 in MCF7 cells, an ER(+) human breast cancer cell line. We then validated that MCF7 cells over-expressing both WNT1 and FGF3 show a 3.5-fold increase in mammosphere formation, and that conditioned media from these cells is also sufficient to promote stem cell activity in untransfected parental MCF7 and T47D cells, as WNT1 and FGF3 are secreted factors. Proteomic analysis of this model system revealed the induction of i) EMT markers, ii) mitochondrial proteins, iii) glycolytic enzymes and iv) protein synthesis machinery, consistent with an anabolic CSC phenotype. MitoTracker staining validated the expected WNT1/FGF3-induced increase in mitochondrial mass and activity, which presumably reflects increased mitochondrial biogenesis. Importantly, many of the proteins that were up-regulated by WNT/FGF-signaling in MCF7 cells, were also transcriptionally over-expressed in human breast cancer cells in vivo, based on the bioinformatic analysis of public gene expression datasets of laser-captured patient samples. As such, this isogenic cell model should accelerate the discovery of new biomarkers to predict clinical outcome in breast cancer, facilitating the development of personalized medicine.

Finally, we used mitochondrial mass as a surrogate marker for increased mitochondrial biogenesis in untransfected MCF7 cells. As predicted, metabolic fractionation of parental MCF7 cells, via MitoTracker staining, indicated that high mitochondrial mass is a new metabolic biomarker for the enrichment of anabolic CSCs, as functionally assessed by mammosphere-forming activity. This observation has broad implications for understanding the role of mitochondrial biogenesis in the propagation of stem-like cancer cells. Technically, this general metabolic approach could be applied to any cancer type, to identify and target the mitochondrial-rich CSC population.

The implications of our work for understanding the role of mitochondrial metabolism in viral oncogenesis driven by random promoter insertions are also discussed, in the context of MMTV and ALV infections.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 5852