Oncotarget

Research Papers:

Secretomes from metastatic breast cancer cells, enriched for a prognostically unfavorable LCN2 axis, induce anti-inflammatory MSC actions and a tumor-supportive premetastatic lung

Kayla J. Meade, Francesca Sanchez, Analine Aguayo, Nathalie Nadales, Sarkis G. Hamalian, Toni L. Uhlendorf, Lisa R. Banner and Jonathan A. Kelber

PDF  |  Full Text  |  Supplementary Files  |  How to cite  |  Press Release  |  Video Interview

Oncotarget. 2019; 10:3027-3039. https://doi.org/10.18632/oncotarget.26903

Metrics: PDF 2172 views  |   Full Text 6309 views  |   ?  


Abstract

Kayla J. Meade1,*, Francesca Sanchez1,*, Analine Aguayo1,*, Nathalie Nadales1, Sarkis G. Hamalian1, Toni L. Uhlendorf1, Lisa R. Banner1 and Jonathan A. Kelber1

1 Department of Biology, California State University Northridge, Northridge CA 91330, USA

* These authors contributed equally to this work

Correspondence to:

Jonathan A. Kelber,email: [email protected]

Keywords: premetastatic niche; tumor microenvironment; metastasis; breast cancer secretomes; LCN2

Received: May 29, 2018     Accepted: April 14, 2019     Published: April 30, 2019

ABSTRACT

Cancer metastasis is responsible for the clear majority of cancer-related deaths. Survival and expansion of cancer cells at secondary sites requires that these premetastatic microenvironments be primed by primary tumor cells and their secreted factors. Efforts to date have been limited by immune-deficient in vivo models and/or the need for finely-tuned analysis time points that reduce contributions from early-disseminating cancer cells. In this regard, we developed a tumor cell-free syngeneic breast cancer model for characterizing tumor cell secretome-mediated reprogramming of premetastatic tissues. We demonstrate that secretomes from metastatic breast cancer cells differentially regulate the lung and brain, promoting a tumor-supportive lung microenvironment with both elevated CD73 expression and decreased TNFα expression. Using in vitro models of CD73-positive mesenchymal stem cells (MSCs) and macrophages/monocytes, we tested whether MSCs can mediate anti-inflammatory effects of metastatic breast cancer cells. Notably, conditioned media from metastatic Py230 cells reprogrammed the secretomes of MSCs toward an anti-inflammatory state. Mining transcriptome data from Py8119 and Py230 cells revealed a lipocalin 2 (LCN2) axis that is selectively expressed in the metastatic Py230 cells, predicts poor breast cancer patient survival and is elevated in circulating serum of mice chronically treated with conditioned media from Py230 cells. Taken together, these results establish the utility of an immune-competent tumor cell-free model for characterizing the mechanisms of breast cancer cell priming of the premetastatic niche, demonstrate that MSCs can mediate the anti-inflammatory effects of metastatic breast cancer cells and substantiate LCN2 as a promising therapeutic target for blocking breast cancer progression.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 26903