The University of Southampton
University of Southampton Institutional Repository

Human endothelial cells modulate CD4+ T cell populations and enhance regulatory T cell suppressive capacity

Human endothelial cells modulate CD4+ T cell populations and enhance regulatory T cell suppressive capacity
Human endothelial cells modulate CD4+ T cell populations and enhance regulatory T cell suppressive capacity
Endothelial cells (ECs) line the luminal surface of blood vessels and have an active role in the recruitment of leukocytes, including immune cell activation. Regulatory T cells (Tregs) are immune suppressor cells that maintain peripheral tolerance and must interact with the endothelium as they traffic into tissue. We hypothesized that human ECs could modulate Tregs and their suppressor function. Cocultures of CD4+ T cells with human umbilical vein ECs (HUVECs) or dermal microvascular ECs (HDMECs) were conducted and analyzed for activation and proliferation after 72 and 120 h using flow cytometry. In monocyte-depleted cultures, human ECs were found to support CD4+ T cell proliferation in the presence of external mitogens phytohemagglutinin or anti-CD3/28 antibodies (aCD3/28). Activation was shown by CD25 expression in these cells that also transiently expressed the Treg transcription factor FOXP3. HUVECs supported the specific concurrent proliferation of both effector T cells and Tregs when cocultured with aCD3/28. Purified Tregs were also functionally activated by prior coculture with EC to suppress effector T (Teff) cell proliferation. Both direct coculture and indirect coculture of EC and Treg showed activation of the Treg suppressive phenotype. However, whereas HUVEC showed enhancement of suppression by both mechanisms, HDMEC only supported Treg suppressive activity via the contact-independent mechanism. In the contact-independent cultures, the soluble mediators IL-6, GM-CSF, or G-CSF released from ECs following interferon-γ activation were not responsible for the enhanced Treg suppressor function. Following direct coculture, Treg expression of inhibitory receptors PD-1 and OX40 was elevated while activated EC expressed the counter ligands programmed death ligand (PD-L)1 and PD-L2. Therefore, human ECs have a role in supporting T cell proliferation and increasing Treg suppressor function. This ability of EC to enhance Treg function could offer novel targets to boost Treg activity during inflammatory disorders.
regulatory T cells, endothelial cells, immune regulation, cell–cell interaction, CD4+ T cells
1664-3224
Lim, Wen Chean
60b7e6ed-1fdb-4de3-b312-75c569726fb5
Olding, Michael
2562d0e0-4ac6-428a-9594-e50874010dd6
Healy, Eugene
400fc04d-f81a-474a-ae25-7ff894be0ebd
Millar, Timothy
ec88510c-ad88-49f6-8b2d-4277c84c1958
Lim, Wen Chean
60b7e6ed-1fdb-4de3-b312-75c569726fb5
Olding, Michael
2562d0e0-4ac6-428a-9594-e50874010dd6
Healy, Eugene
400fc04d-f81a-474a-ae25-7ff894be0ebd
Millar, Timothy
ec88510c-ad88-49f6-8b2d-4277c84c1958

Lim, Wen Chean, Olding, Michael, Healy, Eugene and Millar, Timothy (2018) Human endothelial cells modulate CD4+ T cell populations and enhance regulatory T cell suppressive capacity. Frontiers in Immunology, 9 (565), [565]. (doi:10.3389/fimmu.2018.00565).

Record type: Article

Abstract

Endothelial cells (ECs) line the luminal surface of blood vessels and have an active role in the recruitment of leukocytes, including immune cell activation. Regulatory T cells (Tregs) are immune suppressor cells that maintain peripheral tolerance and must interact with the endothelium as they traffic into tissue. We hypothesized that human ECs could modulate Tregs and their suppressor function. Cocultures of CD4+ T cells with human umbilical vein ECs (HUVECs) or dermal microvascular ECs (HDMECs) were conducted and analyzed for activation and proliferation after 72 and 120 h using flow cytometry. In monocyte-depleted cultures, human ECs were found to support CD4+ T cell proliferation in the presence of external mitogens phytohemagglutinin or anti-CD3/28 antibodies (aCD3/28). Activation was shown by CD25 expression in these cells that also transiently expressed the Treg transcription factor FOXP3. HUVECs supported the specific concurrent proliferation of both effector T cells and Tregs when cocultured with aCD3/28. Purified Tregs were also functionally activated by prior coculture with EC to suppress effector T (Teff) cell proliferation. Both direct coculture and indirect coculture of EC and Treg showed activation of the Treg suppressive phenotype. However, whereas HUVEC showed enhancement of suppression by both mechanisms, HDMEC only supported Treg suppressive activity via the contact-independent mechanism. In the contact-independent cultures, the soluble mediators IL-6, GM-CSF, or G-CSF released from ECs following interferon-γ activation were not responsible for the enhanced Treg suppressor function. Following direct coculture, Treg expression of inhibitory receptors PD-1 and OX40 was elevated while activated EC expressed the counter ligands programmed death ligand (PD-L)1 and PD-L2. Therefore, human ECs have a role in supporting T cell proliferation and increasing Treg suppressor function. This ability of EC to enhance Treg function could offer novel targets to boost Treg activity during inflammatory disorders.

Text
fimmu-09-00565 - Version of Record
Available under License Creative Commons Attribution.
Download (5MB)

More information

Accepted/In Press date: 6 March 2018
e-pub ahead of print date: 23 March 2018
Published date: 23 March 2018
Keywords: regulatory T cells, endothelial cells, immune regulation, cell–cell interaction, CD4+ T cells

Identifiers

Local EPrints ID: 419234
URI: http://eprints.soton.ac.uk/id/eprint/419234
ISSN: 1664-3224
PURE UUID: 03099c86-6932-4724-a404-6e94dd1a9ec1
ORCID for Timothy Millar: ORCID iD orcid.org/0000-0002-4539-2445

Catalogue record

Date deposited: 09 Apr 2018 16:30
Last modified: 15 Mar 2024 19:06

Export record

Altmetrics

Contributors

Author: Wen Chean Lim
Author: Michael Olding
Author: Eugene Healy
Author: Timothy Millar 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.

×