MerTK Drives Proliferation and Metastatic Potential in Triple-Negative Breast Cancer

Int J Mol Sci. 2024 May 8;25(10):5109. doi: 10.3390/ijms25105109.

Abstract

Triple-negative breast cancer (TNBC) is characterized by the absence of the estrogen receptor, progesterone receptor, and receptor tyrosine kinase HER2 expression. Due to the limited number of FDA-approved targeted therapies for TNBC, there is an ongoing need to understand the molecular underpinnings of TNBC for the development of novel combinatorial treatment strategies. This study evaluated the role of the MerTK receptor tyrosine kinase on proliferation and invasion/metastatic potential in TNBC. Immunohistochemical analysis demonstrated MerTK expression in 58% of patient-derived TNBC xenografts. The stable overexpression of MerTK in human TNBC cell lines induced an increase in proliferation rates, robust in vivo tumor growth, heightened migration/invasion potential, and enhanced lung metastases. NanoString nCounter analysis of MerTK-overexpressing SUM102 cells (SUM102-MerTK) revealed upregulation of several signaling pathways, which ultimately drive cell cycle progression, reduce apoptosis, and enhance cell survival. Proteomic profiling indicated increased endoglin (ENG) production in SUM102-MerTK clones, suggesting that MerTK creates a conducive environment for increased proliferative and metastatic activity via elevated ENG expression. To determine ENG's role in increasing proliferation and/or metastatic potential, we knocked out ENG in a SUM102-MerTK clone with CRISPR technology. Although this ENG knockout clone exhibited similar in vivo growth to the parental SUM102-MerTK clone, lung metastasis numbers were significantly decreased ~4-fold, indicating that MerTK enhances invasion and metastasis through ENG. Our data suggest that MerTK regulates a unique proliferative signature in TNBC, promoting robust tumor growth and increased metastatic potential through ENG upregulation. Targeting MerTK and ENG simultaneously may provide a novel therapeutic approach for TNBC patients.

Keywords: MerTK; TNBC; endoglin; lung metastases.

MeSH terms

  • Animals
  • Apoptosis / genetics
  • Cell Line, Tumor
  • Cell Movement / genetics
  • Cell Proliferation*
  • Endoglin / genetics
  • Endoglin / metabolism
  • Female
  • Gene Expression Regulation, Neoplastic
  • Humans
  • Lung Neoplasms / genetics
  • Lung Neoplasms / metabolism
  • Lung Neoplasms / pathology
  • Lung Neoplasms / secondary
  • Mice
  • Neoplasm Metastasis
  • Signal Transduction
  • Triple Negative Breast Neoplasms* / genetics
  • Triple Negative Breast Neoplasms* / metabolism
  • Triple Negative Breast Neoplasms* / pathology
  • c-Mer Tyrosine Kinase* / genetics
  • c-Mer Tyrosine Kinase* / metabolism

Substances

  • c-Mer Tyrosine Kinase
  • MERTK protein, human
  • Endoglin

Grants and funding

The authors would like to thank the University of Wisconsin Carbone Cancer Center (UWCCC) Breast DOT (to D.L.W) for the funds to complete this project. This work was also supported by the NIH/NCI P30 CA014520 (UW Comprehensive Cancer Center Grant, CCSG) and the Office of the Director—NIH (grant no. S10 OD023526) for use of TRIP lab facilities and services. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.