The University of Southampton
University of Southampton Institutional Repository

Investigation into the Roles of Epithelial-Mesenchymal Transition (EMT) in Idiopathic Pulmonary Fibrosis (IPF)

Investigation into the Roles of Epithelial-Mesenchymal Transition (EMT) in Idiopathic Pulmonary Fibrosis (IPF)
Investigation into the Roles of Epithelial-Mesenchymal Transition (EMT) in Idiopathic Pulmonary Fibrosis (IPF)
Idiopathic pulmonary fibrosis (IPF) is a type of chronic, interstitial lung disease, characterised by aberrant extracellular matrix (ECM) deposition. It is an ageing-associated disease, with a poor prognosis and limited treatments are available. Epithelial-mesenchymal transition (EMT) is a biological process where epithelial cells lose cell polarity and cell junctions, gaining a mesenchymal phenotype. The role of EMT, in the generation of fibroblasts in IPF, is controversial, with studies now suggesting that EMT contributes indirectly.
Here, we utilised bioinformatic analysis to evaluate IPF microarray datasets. We showed that the epidermal growth factor receptor-mitogen-activated protein kinase (EGFR-MAPK) pathway and EMT could both be involved in the pathogenesis of IPF. These findings were validated with proteomic analysis of RAS-activated alveolar type II epithelial (ATIIER:KRASV 12) cells, where EMT was identified as a top process. In vitro studies of ATIIER:KRASV 12 cells were used to dissect the mechanisms underlying these processes. We found that RAS-activated EMT was induced via the EGFR-RAS-ERK-ZEB1 axis, but it was insufficient to induce the expression of ECM components in IPF. Autophagy was shown to be reduced in IPF. As autophagy and EMT have a complicated relationship, we explored this in IPF. Using ATII cells, we showed that autophagy inhibition induced EMT via the p62/NF-κB/Snail2 axis. In a similar manner to RAS-induced EMT, autophagy inhibition-induced EMT was insufficient to produce ECM components, however it did mediate secreted factors which were able to augment fibrosis via Snail2.
Together, these findings demonstrate the importance of the EGFR-RAS signalling pathway and autophagy in the pathogenesis of IPF. EMT was induced in ATII cells, but was not sufficient to augment fibrosis directly. We demonstrate that secreted factors, induced by EMT, promote fibrosis. This thesis advances the understanding of the mechanisms underlying the activation of EMT in the fibrotic lung, but also, how this can indirectly result in fibrosis by paracrine signalling mechanisms.
University of Southampton
Hill, Charlotte
6d1cfed3-11b1-48af-b171-b8726ab673eb
Hill, Charlotte
6d1cfed3-11b1-48af-b171-b8726ab673eb
Wang, Yihua
f5044a95-60a7-42d2-87d6-5f1f789e3a7e

Hill, Charlotte (2021) Investigation into the Roles of Epithelial-Mesenchymal Transition (EMT) in Idiopathic Pulmonary Fibrosis (IPF). University of Southampton, Doctoral Thesis, 379pp.

Record type: Thesis (Doctoral)

Abstract

Idiopathic pulmonary fibrosis (IPF) is a type of chronic, interstitial lung disease, characterised by aberrant extracellular matrix (ECM) deposition. It is an ageing-associated disease, with a poor prognosis and limited treatments are available. Epithelial-mesenchymal transition (EMT) is a biological process where epithelial cells lose cell polarity and cell junctions, gaining a mesenchymal phenotype. The role of EMT, in the generation of fibroblasts in IPF, is controversial, with studies now suggesting that EMT contributes indirectly.
Here, we utilised bioinformatic analysis to evaluate IPF microarray datasets. We showed that the epidermal growth factor receptor-mitogen-activated protein kinase (EGFR-MAPK) pathway and EMT could both be involved in the pathogenesis of IPF. These findings were validated with proteomic analysis of RAS-activated alveolar type II epithelial (ATIIER:KRASV 12) cells, where EMT was identified as a top process. In vitro studies of ATIIER:KRASV 12 cells were used to dissect the mechanisms underlying these processes. We found that RAS-activated EMT was induced via the EGFR-RAS-ERK-ZEB1 axis, but it was insufficient to induce the expression of ECM components in IPF. Autophagy was shown to be reduced in IPF. As autophagy and EMT have a complicated relationship, we explored this in IPF. Using ATII cells, we showed that autophagy inhibition induced EMT via the p62/NF-κB/Snail2 axis. In a similar manner to RAS-induced EMT, autophagy inhibition-induced EMT was insufficient to produce ECM components, however it did mediate secreted factors which were able to augment fibrosis via Snail2.
Together, these findings demonstrate the importance of the EGFR-RAS signalling pathway and autophagy in the pathogenesis of IPF. EMT was induced in ATII cells, but was not sufficient to augment fibrosis directly. We demonstrate that secreted factors, induced by EMT, promote fibrosis. This thesis advances the understanding of the mechanisms underlying the activation of EMT in the fibrotic lung, but also, how this can indirectly result in fibrosis by paracrine signalling mechanisms.

Text
Charlotte Hill FINAL Thesis - Version of Record
Available under License University of Southampton Thesis Licence.
Download (96MB)
Text
Permission to deposit thesis - form_CH YW HW - Version of Record
Restricted to Repository staff only
Available under License University of Southampton Thesis Licence.

More information

Published date: 2021

Identifiers

Local EPrints ID: 452407
URI: http://eprints.soton.ac.uk/id/eprint/452407
PURE UUID: e0ff8e11-9375-4d38-a0f6-7a921adcc629
ORCID for Yihua Wang: ORCID iD orcid.org/0000-0001-5561-0648

Catalogue record

Date deposited: 09 Dec 2021 18:09
Last modified: 17 Mar 2024 06:53

Export record

Contributors

Author: Charlotte Hill
Thesis advisor: Yihua Wang ORCID iD

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.

×