4.7 Article

Prrx1 isoform switching regulates pancreatic cancer invasion and metastatic colonization

Journal

GENES & DEVELOPMENT
Volume 30, Issue 2, Pages 233-247

Publisher

COLD SPRING HARBOR LAB PRESS, PUBLICATIONS DEPT
DOI: 10.1101/gad.263327.115

Keywords

Prrx1a; Prrx1b; pancreatic cancer; metastasis; EMT; MET

Funding

  1. National Institutes of Health (NIH) [R01 DK060694]
  2. NIH/National Cancer Institute (NCI) [F30 CA180601-10]
  3. NIH/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) [T32-DK007066]
  4. National Pancreas Foundation
  5. German Cancer Aid Foundation (Max Eder Program, Deutsche Krebshilfe) [111273]
  6. AGA-Actavis Research Award in Pancreatic Disorders
  7. Honjo International Scholarship Foundation
  8. NIH/NIDDK Center for Molecular Studies in Digestive and Liver Diseases (Molecular Pathology and Imaging [P30-DK050306]
  9. Molecular Biology/Gene Expression
  10. Cell Culture, and Transgenic and Chimeric Mouse Cores
  11. American Cancer Society [RP-10-033-01-CCE]
  12. NCI [R01CA113669, 15H04925]
  13. Grants-in-Aid for Scientific Research [15H04925] Funding Source: KAKEN

Ask authors/readers for more resources

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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available