Skip to main content
Top
Published in: Breast Cancer Research 1/2020

01-12-2020 | Breast Cancer | Research article

FAK activates AKT-mTOR signaling to promote the growth and progression of MMTV-Wnt1-driven basal-like mammary tumors

Authors: Ritama Paul, Ming Luo, Xueying Mo, Jason Lu, Syn Kok Yeo, Jun-Lin Guan

Published in: Breast Cancer Research | Issue 1/2020

Login to get access

Abstract

Background

Breast cancer is a heterogeneous disease. Hence, stratification of patients based on the subtype of breast cancer is key to its successful treatment. Among all the breast cancer subtypes, basal-like breast cancer is the most aggressive subtype with limited treatment options. Interestingly, we found focal adhesion kinase (FAK), a cytoplasmic tyrosine kinase, is highly overexpressed and activated in basal-like breast cancer.

Methods

To understand the role of FAK in this subtype, we generated mice with conditional deletion of FAK and a knock-in mutation in its kinase domain in MMTV-Wnt1-driven basal-like mammary tumors. Tumor initiation, growth, and metastasis were characterized for these mice cohorts. Immunohistochemical and transcriptomic analysis of Wnt1-driven tumors were also performed to elucidate the mechanisms underlying FAK-dependent phenotypes. Pharmacological inhibition of FAK and mTOR in human basal-like breast cancer cell lines was also tested.

Results

We found that in the absence of FAK or its kinase function, growth and metastasis of the tumors were significantly suppressed. Furthermore, immunohistochemical analyses of cleaved caspase 3 revealed that loss of FAK results in increased tumor cell apoptosis. To further investigate the mechanism by which FAK regulates survival of the Wnt1-driven tumor cells, we prepared an isogenic pair of mammary tumor cells with and without FAK and found that FAK ablation increased their sensitivity to ER stress-induced cell death, as well as reduced tumor cell migration and tumor sphere formation. Comparative transcriptomic profiling of the pair of tumor cells and gene set enrichment analysis suggested mTOR pathway to be downregulated upon loss of FAK. Immunoblot analyses further confirmed that absence of FAK results in reduction of AKT and downstream mTOR pathways. We also found that inhibition of FAK and mTOR pathways both induces apoptosis, indicating the importance of these pathways in regulating cell survival.

Conclusions

In summary, our studies show that in a basal-like tumor model, FAK is required for survival of the tumor cells and can serve as a potential therapeutic target.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci. 2001;98(19):10869-74. Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci. 2001;98(19):10869-74.
5.
6.
go back to reference Lim E, Wu D, Pal B, Bouras T, Asselin-Labat ML, Vaillant F, et al. Transcriptome analyses of mouse and human mammary cell subpopulations reveal multiple conserved genes and pathways. Breast Cancer Res. 2010;12(2):R21. https://doi.org/10.1186/bcr2560. Lim E, Wu D, Pal B, Bouras T, Asselin-Labat ML, Vaillant F, et al. Transcriptome analyses of mouse and human mammary cell subpopulations reveal multiple conserved genes and pathways. Breast Cancer Res. 2010;12(2):R21. https://​doi.​org/​10.​1186/​bcr2560.
9.
go back to reference Nagy T, Luo M, Guan J-L, Liu S, Fan H, Wei H, et al. Mammary epithelial-specific ablation of the focal adhesion kinase suppresses mammary tumorigenesis by affecting mammary cancer stem/progenitor cells. Cancer Res. 2009;69(2):466–74.CrossRef Nagy T, Luo M, Guan J-L, Liu S, Fan H, Wei H, et al. Mammary epithelial-specific ablation of the focal adhesion kinase suppresses mammary tumorigenesis by affecting mammary cancer stem/progenitor cells. Cancer Res. 2009;69(2):466–74.CrossRef
11.
go back to reference Provenzano PP, Inman DR, Eliceiri KW, Beggs HE, Keely PJ. Mammary epithelial-specific disruption of focal adhesion kinase retards tumor formation and metastasis in a transgenic mouse model of human breast cancer. Am J Pathol. 2008;173(5):1551–65. [cited 2019 Mar 30] Available from: http://www.ncbi.nlm.nih.gov/pubmed/18845837. Provenzano PP, Inman DR, Eliceiri KW, Beggs HE, Keely PJ. Mammary epithelial-specific disruption of focal adhesion kinase retards tumor formation and metastasis in a transgenic mouse model of human breast cancer. Am J Pathol. 2008;173(5):1551–65. [cited 2019 Mar 30] Available from: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​18845837.
12.
go back to reference Lazaro G, Smith C, Goddard L, Jordan N, McClelland R, Barrett-Lee P, et al. Targeting focal adhesion kinase in ER+/HER2+ breast cancer improves trastuzumab response. Endocr Relat Cancer. 2013;20(5):691–704.CrossRef Lazaro G, Smith C, Goddard L, Jordan N, McClelland R, Barrett-Lee P, et al. Targeting focal adhesion kinase in ER+/HER2+ breast cancer improves trastuzumab response. Endocr Relat Cancer. 2013;20(5):691–704.CrossRef
13.
go back to reference Lahlou H, Sanguin-Gendreau V, Frame MC, Muller WJ. Focal adhesion kinase contributes to proliferative potential of ErbB2 mammary tumour cells but is dispensable for ErbB2 mammary tumour induction in vivo. Breast Cancer Res. 2012;14(1):R36. Published 2012 Feb 28. https://doi.org/10.1186/bcr3131. Lahlou H, Sanguin-Gendreau V, Frame MC, Muller WJ. Focal adhesion kinase contributes to proliferative potential of ErbB2 mammary tumour cells but is dispensable for ErbB2 mammary tumour induction in vivo. Breast Cancer Res. 2012;14(1):R36. Published 2012 Feb 28. https://​doi.​org/​10.​1186/​bcr3131.
15.
18.
go back to reference Provenzano PP, Inman DR, Eliceiri KW, Beggs HE, Keely PJ. Mammary epithelial-specific disruption of focal adhesion kinase retards tumor formation and metastasis in a transgenic mouse model of human breast cancer. Am J Pathol. 2008;173(5):1551–65. https://doi.org/10.2353/ajpath.2008.080308. Provenzano PP, Inman DR, Eliceiri KW, Beggs HE, Keely PJ. Mammary epithelial-specific disruption of focal adhesion kinase retards tumor formation and metastasis in a transgenic mouse model of human breast cancer. Am J Pathol. 2008;173(5):1551–65. https://​doi.​org/​10.​2353/​ajpath.​2008.​080308.
20.
go back to reference Chen Y, Wei H, Liu F, Guan JL. Hyperactivation of mammalian target of rapamycin complex 1 (mTORC1) promotes breast cancer progression through enhancing glucose starvation-induced autophagy and Akt signaling. J Biol Chem. 2014. 289(2):1164–73. https://doi.org/10.1074/jbc.M113.526335. Chen Y, Wei H, Liu F, Guan JL. Hyperactivation of mammalian target of rapamycin complex 1 (mTORC1) promotes breast cancer progression through enhancing glucose starvation-induced autophagy and Akt signaling. J Biol Chem. 2014. 289(2):1164–73. https://​doi.​org/​10.​1074/​jbc.​M113.​526335.
25.
go back to reference Shen TL, Park AYJ, Alcaraz A, Peng X, Jang I, Koni P, et al. Conditional knockout of focal adhesion kinase in endothelial cells reveals its role in angiogenesis and vascular development in late embryogenesis. J Cell Biol. 2005;169(6):941–52.CrossRef Shen TL, Park AYJ, Alcaraz A, Peng X, Jang I, Koni P, et al. Conditional knockout of focal adhesion kinase in endothelial cells reveals its role in angiogenesis and vascular development in late embryogenesis. J Cell Biol. 2005;169(6):941–52.CrossRef
26.
go back to reference Zhao X, Peng X, Sun S, Park AYJ, Guan J-L. Role of kinase-independent and -dependent functions of FAK in endothelial cell survival and barrier function during embryonic development. J Cell Biol. 2010;189(6):955–65. Available from: https://doi.org/10.1083/jcb.200912094. Zhao X, Peng X, Sun S, Park AYJ, Guan J-L. Role of kinase-independent and -dependent functions of FAK in endothelial cell survival and barrier function during embryonic development. J Cell Biol. 2010;189(6):955–65. Available from: https://​doi.​org/​10.​1083/​jcb.​200912094.
27.
go back to reference Luo M, Zhao X, Chen S, Liu S, Wicha MS, Guan JL. Distinct FAK activities determine progenitor and mammary stem cell characteristics. Cancer Res. 2013;73(734):5591–602.CrossRef Luo M, Zhao X, Chen S, Liu S, Wicha MS, Guan JL. Distinct FAK activities determine progenitor and mammary stem cell characteristics. Cancer Res. 2013;73(734):5591–602.CrossRef
30.
go back to reference Nagy T, Wei H, Shen TL, Peng X, Liang CC, Gan B, et al. Mammary epithelial-specific deletion of the focal adhesion kinase gene leads to severe lobulo-alveolar hypoplasia and secretory immaturity of the murine mammary gland. J Biol Chem. 2007;282(43):31766–76. https://doi.org/10.1074/jbc.M705403200. Nagy T, Wei H, Shen TL, Peng X, Liang CC, Gan B, et al. Mammary epithelial-specific deletion of the focal adhesion kinase gene leads to severe lobulo-alveolar hypoplasia and secretory immaturity of the murine mammary gland. J Biol Chem. 2007;282(43):31766–76. https://​doi.​org/​10.​1074/​jbc.​M705403200.
32.
go back to reference Pereira B, Chin SF, Rueda OM, Vollan HKM, Provenzano E, Bardwell HA, et al. The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat Commun. 2016;7:11479. Published 2016 May 10. https://doi.org/10.1038/ncomms11479. Pereira B, Chin SF, Rueda OM, Vollan HKM, Provenzano E, Bardwell HA, et al. The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat Commun. 2016;7:11479. Published 2016 May 10. https://​doi.​org/​10.​1038/​ncomms11479.
38.
42.
Metadata
Title
FAK activates AKT-mTOR signaling to promote the growth and progression of MMTV-Wnt1-driven basal-like mammary tumors
Authors
Ritama Paul
Ming Luo
Xueying Mo
Jason Lu
Syn Kok Yeo
Jun-Lin Guan
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2020
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-020-01298-3

Other articles of this Issue 1/2020

Breast Cancer Research 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine