Abstract

Peritoneal dissemination is the primary metastatic route of ovarian cancer (OvCa), and is often accompanied by the accumulation of ascitic fluid. The peritoneal cavity is lined by mesothelial cells (MCs), which can be converted into carcinoma‐associated fibroblasts (CAFs) through mesothelial‐to‐mesenchymal transition (MMT). Here, we demonstrate that MCs isolated from ascitic fluid (AFMCs) of OvCa patients with peritoneal implants also undergo MMT and promote subcutaneous tumour growth in mice. RNA sequencing of AFMCs revealed that MMT‐related pathways – including transforming growth factor (TGF)‐β signalling – are differentially regulated, and a gene signature was verified in peritoneal implants from OvCa patients. In a mouse model, pre‐induction of MMT resulted in increased peritoneal tumour growth, whereas interfering with the TGF‐β receptor reduced metastasis. MC‐derived CAFs showed activation of Smad‐dependent TGF‐β signalling, which was disrupted in OvCa cells, despite their elevated TGF‐β production. Accordingly, targeting Smad‐dependent signalling in the peritoneal pre‐metastatic niche in mice reduced tumour colonization, suggesting that Smad‐dependent MMT could be crucial in peritoneal carcinomatosis. Together, these results indicate that bidirectional communication between OvCa cells and MC‐derived CAFs, via TGF‐β‐mediated MMT, seems to be crucial to form a suitable metastatic niche. We suggest MMT as a possible target for therapeutic intervention and a potential source of biomarkers for improving OvCa diagnosis and/or prognosis. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Highlights

  • A common characteristic of cancers that progress with peritoneal metastasis is that they evolve very rapidly, without symptoms, and are diagnosed at advanced stages [1]

  • We have previously shown that a subset of carcinoma-associated fibroblasts (CAFs) in peritoneal metastases are derived from mesothelial cells (MCs) via mesothelial-to-mesenchymal transition (MMT) [6,7], which is an epithelial-to-mesenchymal transition (EMT)-like process [8,9]

  • Positive immunofluorescence staining for calretinin confirmed their MC nature, and α-smooth muscle actin (α-SMA) staining indicated that ascitic fluid-isolated MCs (AFMCs) had been converted to myofibroblasts (Figure 1A)

Read more

Summary

Introduction

A common characteristic of cancers that progress with peritoneal metastasis is that they evolve very rapidly, without symptoms, and are diagnosed at advanced stages [1]. Debulking surgery followed by platinum–taxane chemotherapy is the current standard of treatment, and improves survival rates in selected patients [2]; there is still limited scope for curing peritoneal carcinomatosis. Survival rates of patients with ovarian cancer (OvCa) at advanced stages are 10–30% [3], making it the fifth leading cause of cancer death in women [4]. We have previously shown that a subset of carcinoma-associated fibroblasts (CAFs) in peritoneal metastases are derived from MCs via mesothelial-to-mesenchymal transition (MMT) [6,7], which is an epithelial-to-mesenchymal transition (EMT)-like process [8,9]. During MMT, MCs acquire a fibroblast-like phenotype, with increased capacity to migrate and to invade the submesothelial compact zone. The acquisition of mesenchymal features by MCs results from a profound genetic reprogramming [8,9]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call