The University of Southampton
University of Southampton Institutional Repository

Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) in human lung microvascular endothelial cells controls oxidative stress, reactive oxygen-mediated cell signaling and inflammatory responses

Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) in human lung microvascular endothelial cells controls oxidative stress, reactive oxygen-mediated cell signaling and inflammatory responses
Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) in human lung microvascular endothelial cells controls oxidative stress, reactive oxygen-mediated cell signaling and inflammatory responses
Background: Perturbation of endothelial function in people with cystic fibrosis (CF) has been reported, which may be associated with endothelial cell expression of the cystic fibrosis transmembrane conductance regulator (CFTR). Previous reports indicate that CFTR activity upregulates endothelial barrier function, endothelial nitric oxide synthase (eNOS) expression and NO release, while limiting interleukin-8 (IL-8) release, in human umbilical vein endothelial cells (HUVECs) in cell culture. In view of reported microvascular dysfunction in people with CF we investigated the role of CFTR expression and activity in the regulation of oxidative stress, cell signaling and inflammation in human lung microvascular endothelial cells (HLMVECs) in cell culture.

Methods: HLMVECs were cultured in the absence and presence of the CFTR inhibitor GlyH-101 and CFTR siRNA. CFTR expression was analyzed using qRT-PCR, immunocytochemistry (IHC) and western blot, and function by membrane potential assay. IL-8 expression was analyzed using qRT-PCR and ELISA. Nrf2 expression, and NF-κB and AP-1 activation were determined using IHC and western blot. The role of the epidermal growth factor receptor (EGFR) in CFTR signaling was investigated using the EGFR tyrosine kinase inhibitor AG1478. Oxidative stress was measured as intracellular ROS and hydrogen peroxide (H2O2) concentration. VEGF and SOD-2 were measured in culture supernatants by ELISA.

Results: HLMVECs express low levels of CFTR that increase following inhibition of CFTR activity. Inhibition of CFTR, significantly increased intracellular ROS and H2O2 levels over 30 min and significantly decreased Nrf2 expression by 70% while increasing SOD-2 expression over 24 h. CFTR siRNA significantly increased constitutive expression of IL-8 by HLMVECs. CFTR inhibition activated the AP-1 pathway and increased IL-8 expression, without effect on NF-κB activity. Conversely, TNF-α activated the NF-κB pathway and increased IL-8 expression. The effects of TNF-α and GlyH-101 on IL-8 expression were additive and inhibited by AG1478. Inhibition of both CFTR and EGFR in HLMVECs significantly increased VEGF expression. The antioxidant N-acetyl cysteine significantly reduced ROS production and the increase in IL-8 and VEGF expression following CFTR inhibition.

Conclusion: Functional endothelial CFTR limits oxidative stress and contributes to the normal anti-inflammatory state of HLMVECs. Therapeutic strategies to restore endothelial CFTR function in CF are warranted.

cell signaling, CFTR, cystic fibrosis, endothelium, inflammation, oxidative stress
1664-042X
Khalaf, Maha
68ac6fc8-8018-4286-acc6-89b22e65d839
Scott-Ward, Toby
2f1bc67d-24c2-4088-95b4-718a230f3ecd
Causer, Adam
17d4182a-d52f-4b92-89fc-99cec813e98d
Saynor, Zoe
a4357c7d-db59-4fa5-b24f-58d2f7e74e39
Shepherd, Anthony
f7073e22-cda6-4816-a4b4-f6246e1aa42e
Górecki, Dariusz
6406abcf-0561-40dc-b41f-32f55795eb04
Lewis, Anthony
2e8de8fb-88fa-45cf-b660-80977fdb4966
Laight, David
a1a931ad-a60a-4016-95c7-13ff8833f91e
Shute, Janis
a5eef853-50ae-4abf-9e34-1c873601437f
Khalaf, Maha
68ac6fc8-8018-4286-acc6-89b22e65d839
Scott-Ward, Toby
2f1bc67d-24c2-4088-95b4-718a230f3ecd
Causer, Adam
17d4182a-d52f-4b92-89fc-99cec813e98d
Saynor, Zoe
a4357c7d-db59-4fa5-b24f-58d2f7e74e39
Shepherd, Anthony
f7073e22-cda6-4816-a4b4-f6246e1aa42e
Górecki, Dariusz
6406abcf-0561-40dc-b41f-32f55795eb04
Lewis, Anthony
2e8de8fb-88fa-45cf-b660-80977fdb4966
Laight, David
a1a931ad-a60a-4016-95c7-13ff8833f91e
Shute, Janis
a5eef853-50ae-4abf-9e34-1c873601437f

Khalaf, Maha, Scott-Ward, Toby, Causer, Adam, Saynor, Zoe, Shepherd, Anthony, Górecki, Dariusz, Lewis, Anthony, Laight, David and Shute, Janis (2020) Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) in human lung microvascular endothelial cells controls oxidative stress, reactive oxygen-mediated cell signaling and inflammatory responses. Frontiers in Physiology, 11, [879]. (doi:10.3389/fphys.2020.00879).

Record type: Article

Abstract

Background: Perturbation of endothelial function in people with cystic fibrosis (CF) has been reported, which may be associated with endothelial cell expression of the cystic fibrosis transmembrane conductance regulator (CFTR). Previous reports indicate that CFTR activity upregulates endothelial barrier function, endothelial nitric oxide synthase (eNOS) expression and NO release, while limiting interleukin-8 (IL-8) release, in human umbilical vein endothelial cells (HUVECs) in cell culture. In view of reported microvascular dysfunction in people with CF we investigated the role of CFTR expression and activity in the regulation of oxidative stress, cell signaling and inflammation in human lung microvascular endothelial cells (HLMVECs) in cell culture.

Methods: HLMVECs were cultured in the absence and presence of the CFTR inhibitor GlyH-101 and CFTR siRNA. CFTR expression was analyzed using qRT-PCR, immunocytochemistry (IHC) and western blot, and function by membrane potential assay. IL-8 expression was analyzed using qRT-PCR and ELISA. Nrf2 expression, and NF-κB and AP-1 activation were determined using IHC and western blot. The role of the epidermal growth factor receptor (EGFR) in CFTR signaling was investigated using the EGFR tyrosine kinase inhibitor AG1478. Oxidative stress was measured as intracellular ROS and hydrogen peroxide (H2O2) concentration. VEGF and SOD-2 were measured in culture supernatants by ELISA.

Results: HLMVECs express low levels of CFTR that increase following inhibition of CFTR activity. Inhibition of CFTR, significantly increased intracellular ROS and H2O2 levels over 30 min and significantly decreased Nrf2 expression by 70% while increasing SOD-2 expression over 24 h. CFTR siRNA significantly increased constitutive expression of IL-8 by HLMVECs. CFTR inhibition activated the AP-1 pathway and increased IL-8 expression, without effect on NF-κB activity. Conversely, TNF-α activated the NF-κB pathway and increased IL-8 expression. The effects of TNF-α and GlyH-101 on IL-8 expression were additive and inhibited by AG1478. Inhibition of both CFTR and EGFR in HLMVECs significantly increased VEGF expression. The antioxidant N-acetyl cysteine significantly reduced ROS production and the increase in IL-8 and VEGF expression following CFTR inhibition.

Conclusion: Functional endothelial CFTR limits oxidative stress and contributes to the normal anti-inflammatory state of HLMVECs. Therapeutic strategies to restore endothelial CFTR function in CF are warranted.

Text
fphys-11-00879 - Version of Record
Available under License Creative Commons Attribution.
Download (5MB)

More information

Accepted/In Press date: 29 June 2020
Published date: 29 July 2020
Keywords: cell signaling, CFTR, cystic fibrosis, endothelium, inflammation, oxidative stress

Identifiers

Local EPrints ID: 494353
URI: http://eprints.soton.ac.uk/id/eprint/494353
ISSN: 1664-042X
PURE UUID: 6e40567b-acbf-42ab-bd51-bc59e85bc4d2
ORCID for Zoe Saynor: ORCID iD orcid.org/0000-0003-0674-8477

Catalogue record

Date deposited: 04 Oct 2024 17:03
Last modified: 05 Oct 2024 02:18

Export record

Altmetrics

Contributors

Author: Maha Khalaf
Author: Toby Scott-Ward
Author: Adam Causer
Author: Zoe Saynor ORCID iD
Author: Anthony Shepherd
Author: Dariusz Górecki
Author: Anthony Lewis
Author: David Laight
Author: Janis Shute

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.

×