Prrx1 isoform switching regulates pancreatic cancer invasion and metastatic colonization

  1. Anil K. Rustgi1,2,3,9
  1. 1Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
  2. 2Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
  3. 3Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
  4. 4Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
  5. 5II. Medizinische Klinik, Technical University of Munich, Munich 81675, Germany;
  6. 6Department of Medicine II, Medical Center, University of Freiburg, 79106 Freiburg, Germany;
  7. 7Department of Pathology, Sheikh Ahmad bin Zayed Al Nahyan Pancreatic Cancer Research Center, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA;
  8. 8Department of Translational Molecular Pathology, Sheikh Ahmad bin Zayed Al Nahyan Pancreatic Cancer Research Center, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA;
  9. 9Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
  1. Corresponding authors: anil2{at}mail.med.upenn.edu, rmax{at}mail.med.upenn.edu or maximilian.reichert{at}tum.de

Abstract

The two major isoforms of the paired-related homeodomain transcription factor 1 (Prrx1), Prrx1a and Prrx1b, are involved in pancreatic development, pancreatitis, and carcinogenesis, although the biological role that these isoforms serve in the systemic dissemination of pancreatic ductal adenocarcinoma (PDAC) has not been investigated. An epithelial–mesenchymal transition (EMT) is believed to be important for primary tumor progression and dissemination, whereas a mesenchymal–epithelial transition (MET) appears crucial for metastatic colonization. Here, we describe novel roles for both isoforms in the metastatic cascade using complementary in vitro and in vivo models. Prrx1b promotes invasion, tumor dedifferentiation, and EMT. In contrast, Prrx1a stimulates metastatic outgrowth in the liver, tumor differentiation, and MET. We further demonstrate that the switch from Prrx1b to Prrx1a governs EMT plasticity in both mouse models of PDAC and human PDAC. Last, we identify hepatocyte growth factor ( HGF) as a novel transcriptional target of Prrx1b. Targeted therapy of HGF in combination with gemcitabine in a preclinical model of PDAC reduces primary tumor volume and eliminates metastatic disease. Overall, we provide new insights into the isoform-specific roles of Prrx1a and Prrx1b in primary PDAC formation, dissemination, and metastatic colonization, allowing for novel therapeutic strategies targeting EMT plasticity.

Keywords

Footnotes

  • Received October 5, 2015.
  • Accepted December 11, 2015.

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