Authors

Rebecca Lamb, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of ManchesterFollow
Gloria Bonuccelli, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of ManchesterFollow
Béla Ozsvári, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester
Maria Peiris-Pagès, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of ManchesterFollow
Marco Fiorillo, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester; The Department of Pharmacy, Health and Nutritional Sciences, The University of CalabriaFollow
Duncan L Smith, The Cancer Research UK Manchester Institute, University of ManchesterFollow
Generoso Bevilacqua, FPS - The Pisa Science Foundation; Department of Pathology, Pisa University Hospital
Chiara Maria Mazzanti, FPS - The Pisa Science Foundation
Liam A McDonnell, FPS - The Pisa Science Foundation
Antonio Giuseppe Naccarato, Department of Pathology, Pisa University Hospital
Maybo Chiu, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester
Luke Wynne, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester
Ubaldo E Martinez-Outschoorn, The Sidney Kimmel Cancer CenterFollow
Federica Sotgia, Manchester Breast Centre and Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, UK; The Jefferson Stem Cell Biology and Regenerative Medicine Center, Philadelphia, PA; Departments of Stem Cell Biology & Regenerative Medicine, and Cancer Biology, Thomas Jefferson UniversityFollow
Michael P. Lisanti, The Breast Cancer Now Research Unit, Institute of Cancer Sciences, University of Manchester; The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of ManchesterFollow

Document Type

Article

Publication Date

10-13-2015

Comments

This article has been peer reviewed. It was published in: Oncotarget.

Volume 6, Issue 31, 2015, Pages 30453-30471.

The published version is available at DOI: 10.18632/oncotarget.5852

Copyright © 2015 The Authors

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

Creative Commons License
This work is licensed under a Creative Commons Attribution 3.0 License.

PubMed ID

26421711

Included in

Oncology Commons

Share

COinS