Discoidin domain receptor 1 (DDR1) ablation promotes tissue fibrosis and hypoxia to induce aggressive basal-like breast cancers

  1. Zena Werb1
  1. 1Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA;
  2. 2Division of Breast Oncology, Saitama Cancer Center, Saitama 362-0806, Japan;
  3. 3Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, San Francisco, California 94143, USA;
  4. 4Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, California 94143, USA;
  5. 5Department of Radiation Oncology, University of California at San Francisco, San Francisco, California 94143, USA
  1. Corresponding author: zena.werb{at}ucsf.edu
  • Present addresses: 6Department of Physiology and Biophysics, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA; 7Harper Cancer Research Institute, Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN 46617, USA; 8Faculty of Medicine, Bar-Ilan University, Henrietta Sold 8, Safed, Israel; 9Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA.

Abstract

The discoidin domain receptor 1 (DDR1) is overexpressed in breast carcinoma cells. Low DDR1 expression is associated with worse relapse-free survival, reflecting its controversial role in cancer progression. We detected DDR1 on luminal cells but not on myoepithelial cells of DDR1+/+ mice. We found that DDR1 loss compromises cell adhesion, consistent with data that older DDR1−/− mammary glands had more basal/myoepithelial cells. Basal cells isolated from older mice exerted higher traction forces than the luminal cells, in agreement with increased mammary branches observed in older DDR1−/− mice and higher branching by their isolated organoids. When we crossed DDR1−/− mice with MMTV-PyMT mice, the PyMT/DDR1−/− mammary tumors grew faster and had increased epithelial tension and matricellular fibrosis with a more basal phenotype and increased lung metastases. DDR1 deletion induced basal differentiation of CD90+CD24+ cancer cells, and the increase in basal cells correlated with tumor cell mitoses. K14+ basal cells, including K8+K14+ cells, were increased adjacent to necrotic fields. These data suggest that the absence of DDR1 provides a growth and adhesion advantage that favors the expansion of basal cells, potentiates fibrosis, and enhances necrosis/hypoxia and basal differentiation of transformed cells to increase their aggression and metastatic potential.

Keywords

Footnotes

  • Received May 2, 2017.
  • Accepted January 24, 2018.

This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first six months after the full-issue publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml). After six months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.

| Table of Contents

Life Science Alliance