Oncotarget

Research Papers:

Cathepsin L secretion by host and neoplastic cells potentiates invasion

Samantha S. Dykes, Henrietta O. Fasanya and Dietmar W. Siemann

PDF  |  Full Text  |  Supplementary Files  |  How to cite

Oncotarget. 2019; 10:5560-5568. https://doi.org/10.18632/oncotarget.27182

Metrics: PDF 1169 views  |   Full Text 2145 views  |   ?  


Abstract

Samantha S. Dykes1, Henrietta O. Fasanya1 and Dietmar W. Siemann1

1 Department of Radiation Oncology, University of Florida, Gainesville, FL, USA

Correspondence to:

Samantha S. Dykes,email: [email protected]

Keywords: breast cancer; macrophage; invasion; cathepsin L

Received: May 23, 2019     Accepted: July 21, 2019     Published: September 17, 2019

ABSTRACT

The presence of macrophages within breast tumors correlates with metastatic potential. These tumor-associated macrophages often take on a pro-tumorigenic (M2-like) phenotype resulting in the secretion of growth factors and proteases, including the lysosomal protease cathepsin L. Since cathepsin L also is frequently secreted by breast cancer cells and contributes to tumor invasion, metastasis, and angiogenesis, we hypothesized that secretion of cathepsin L by both tumor-associated macrophages and neoplastic cells would facilitate the metastatic phenotype. Our results showed that the novel cathepsin L/K inhibitors KGP94 and KGP207 could inhibit in vitro M2 macrophage invasion and reduce the macrophage-stimulated invasion of 4T1 murine breast cancer cells. KGP94 and KGP207 treatment also reduced the expression of several M2-associated markers, suggesting that cathepsin L activity may be important for IL-4-driven M0 to M2 differentiation. In addition, cathepsin L shRNA knockdown studies revealed that cathepsin L from both the tumor cell and the macrophage population is important for tumor cell invasion. Thus our data suggest that tumor cells and macrophages may both contribute to the cathepsin L-driven metastatic phenotype of breast cancer. Taken together, these studies highlight the importance of cathepsin L in macrophage functions and suggest that cathepsin inhibition strategies may be therapeutically beneficial by impairing the progression of tumors with high infiltration of M2 macrophages.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 27182