The University of Southampton
University of Southampton Institutional Repository

Cancer associated fibroblasts predict for poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma

Cancer associated fibroblasts predict for poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma
Cancer associated fibroblasts predict for poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma
Interactions between cancer cells and cancer-associated fibroblasts (CAF) play an important role in tumour development and progression. In this study we investigated the functional role of CAF in oesophageal adenocarcinoma (EAC). We used immunochemistry to analyse a cohort of EAC patients (183 patients) for CAF markers related to disease mortality. We characterized CAF and normal oesophageal fibroblasts (NOF) using western blotting, immunofluorescence and gel contraction. Transwell assays, 3-D organotypic culture and xenograft models were used to examine effects on EAC cell function, and dissect molecular mechanisms regulating invasion. Most EAC (93%) contained CAF with a myofibroblastic (?-SMA-positive) phenotype, which correlated significantly with poor survival (p?=?0.016; HR 7. 1 (1.7-29.4). Primary CAF, isolated from EAC, have a contractile, myofibroblastic phenotype, and promote EAC cell invasion in vitro (Transwell assays, p?=?<0.05; organotypic culture, p?<?0.001) and in vivo (p?=?<0.05). In vitro, this pro-invasive effect is modulated through the matricellular protein periostin. Periostin is secreted by CAF, and acts as a ligand for EAC cell integrins ?v?3 and ?v?5, promoting activation of the PI3kinase/Akt pathway. In patient samples, periostin expression at the tumour cell/stromal interface correlates with poor overall and disease-free survival. Our study highlights the importance of the tumour stroma in EAC progression. Paracrine interaction between CAF-secreted periostin and EAC-expressed integrins results in PI3 kinase/Akt activation and increased tumour cell invasion. Most EAC contain a myofibroblastic CAF-rich stroma; this may explain the aggressive, highly infiltrative nature of the disease, and suggests that stromal targeting may produce therapeutic benefit in EAC patients
466-477
Underwood, Timothy J.
8e81bf60-edd2-4b0e-8324-3068c95ea1c6
Hayden, Annette L.
80301564-d83f-404c-abd0-0f8acf6c2e60
Derouet, Mathieu
03c558fa-9373-492e-99e1-d35e1cfbbd86
Garcia, Edwin
d25d8ad5-2763-4051-8f43-dca12fa1b545
Noble, Fergus
4f14574c-28f2-4e04-bd95-f53c7649e1fa
White, Michael J.
fcbaf8a0-d2ec-4dd2-9aa5-4a252ab95d58
Thirdborough, Steve
161784fb-c8e3-4beb-86b1-cd8bc8ddf8de
Mead, Abbie
93c092b9-d3d3-4763-b020-04d8ac48f503
Clemons, Nicholas
372bb72d-0cdd-45d8-99b0-32e8f35c93e0
Mellone, Massimiliano
b0301b32-14f8-4203-9026-b7f90885cab9
Uzoho, Chudy
a214c0bf-7c7a-4aee-86b6-2ee0cb9ff343
Primrose, John N
d85f3b28-24c6-475f-955b-ec457a3f9185
Blaydes, Jeremy P
e957f999-fd91-4f77-ad62-5b4ef069b15b
Thomas, Gareth J
2ff54aa9-a766-416b-91ee-cf1c5be74106
Underwood, Timothy J.
8e81bf60-edd2-4b0e-8324-3068c95ea1c6
Hayden, Annette L.
80301564-d83f-404c-abd0-0f8acf6c2e60
Derouet, Mathieu
03c558fa-9373-492e-99e1-d35e1cfbbd86
Garcia, Edwin
d25d8ad5-2763-4051-8f43-dca12fa1b545
Noble, Fergus
4f14574c-28f2-4e04-bd95-f53c7649e1fa
White, Michael J.
fcbaf8a0-d2ec-4dd2-9aa5-4a252ab95d58
Thirdborough, Steve
161784fb-c8e3-4beb-86b1-cd8bc8ddf8de
Mead, Abbie
93c092b9-d3d3-4763-b020-04d8ac48f503
Clemons, Nicholas
372bb72d-0cdd-45d8-99b0-32e8f35c93e0
Mellone, Massimiliano
b0301b32-14f8-4203-9026-b7f90885cab9
Uzoho, Chudy
a214c0bf-7c7a-4aee-86b6-2ee0cb9ff343
Primrose, John N
d85f3b28-24c6-475f-955b-ec457a3f9185
Blaydes, Jeremy P
e957f999-fd91-4f77-ad62-5b4ef069b15b
Thomas, Gareth J
2ff54aa9-a766-416b-91ee-cf1c5be74106

Underwood, Timothy J., Hayden, Annette L., Derouet, Mathieu, Garcia, Edwin, Noble, Fergus, White, Michael J., Thirdborough, Steve, Mead, Abbie, Clemons, Nicholas, Mellone, Massimiliano, Uzoho, Chudy, Primrose, John N, Blaydes, Jeremy P and Thomas, Gareth J (2015) Cancer associated fibroblasts predict for poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma. The Journal of Pathology, 235, 466-477. (doi:10.1002/path.4467). (PMID:25345775)

Record type: Article

Abstract

Interactions between cancer cells and cancer-associated fibroblasts (CAF) play an important role in tumour development and progression. In this study we investigated the functional role of CAF in oesophageal adenocarcinoma (EAC). We used immunochemistry to analyse a cohort of EAC patients (183 patients) for CAF markers related to disease mortality. We characterized CAF and normal oesophageal fibroblasts (NOF) using western blotting, immunofluorescence and gel contraction. Transwell assays, 3-D organotypic culture and xenograft models were used to examine effects on EAC cell function, and dissect molecular mechanisms regulating invasion. Most EAC (93%) contained CAF with a myofibroblastic (?-SMA-positive) phenotype, which correlated significantly with poor survival (p?=?0.016; HR 7. 1 (1.7-29.4). Primary CAF, isolated from EAC, have a contractile, myofibroblastic phenotype, and promote EAC cell invasion in vitro (Transwell assays, p?=?<0.05; organotypic culture, p?<?0.001) and in vivo (p?=?<0.05). In vitro, this pro-invasive effect is modulated through the matricellular protein periostin. Periostin is secreted by CAF, and acts as a ligand for EAC cell integrins ?v?3 and ?v?5, promoting activation of the PI3kinase/Akt pathway. In patient samples, periostin expression at the tumour cell/stromal interface correlates with poor overall and disease-free survival. Our study highlights the importance of the tumour stroma in EAC progression. Paracrine interaction between CAF-secreted periostin and EAC-expressed integrins results in PI3 kinase/Akt activation and increased tumour cell invasion. Most EAC contain a myofibroblastic CAF-rich stroma; this may explain the aggressive, highly infiltrative nature of the disease, and suggests that stromal targeting may produce therapeutic benefit in EAC patients

Text
Underwood et al., 2015.pdf - Version of Record
Available under License Creative Commons Attribution.
Download (6MB)

More information

e-pub ahead of print date: 8 January 2015
Published date: January 2015
Organisations: Cancer Sciences

Identifiers

Local EPrints ID: 370608
URI: http://eprints.soton.ac.uk/id/eprint/370608
PURE UUID: 8be8ff3f-99b1-42c0-8480-188e4844bfa9
ORCID for Timothy J. Underwood: ORCID iD orcid.org/0000-0001-9455-2188
ORCID for Massimiliano Mellone: ORCID iD orcid.org/0000-0002-4964-9340
ORCID for John N Primrose: ORCID iD orcid.org/0000-0002-2069-7605
ORCID for Jeremy P Blaydes: ORCID iD orcid.org/0000-0001-8525-0209

Catalogue record

Date deposited: 31 Oct 2014 16:30
Last modified: 15 Mar 2024 03:17

Export record

Altmetrics

Contributors

Author: Annette L. Hayden
Author: Mathieu Derouet
Author: Edwin Garcia
Author: Fergus Noble
Author: Michael J. White
Author: Abbie Mead
Author: Nicholas Clemons
Author: Massimiliano Mellone ORCID iD
Author: Chudy Uzoho
Author: John N Primrose ORCID iD
Author: Gareth J Thomas

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×