The University of Southampton
University of Southampton Institutional Repository

A system immunology approach to understanding human langerhans cell tolerogenic function

A system immunology approach to understanding human langerhans cell tolerogenic function
A system immunology approach to understanding human langerhans cell tolerogenic function
Langerhans cells (LC) maintain skin homeostasis through orchestrating immunogenic and tolerogenic immune responses in steady-state epidermis and at local lymph nodes after migration.While the mechanisms promoting activation of immune responses has been elucidated, little is known about the molecular mechanisms underpinning LC induced tolerance. We hypothesised that heterogeneous human LC populations existed in situ, which specialised in regulation of immunogenic vs tolerogenic regulation in healthy skin. Within the transcriptome of tolerance regulating LC, we sought to identify key molecular mediators of tolerogenic programming.

Previously published transcriptomic data containing a wide selection of DC subpopulations, including LC was analysed. Here, core mechanisms of tolerogenic programming across DCs was investigated. Overall, LC transcriptomic programming was largely distinct, although common pathways of suppression of stimuli responsiveness were identified in steady-state LCs.

The Drop-seq scRNA-seq protocol was optimised and implemented on steady-state and migrated LCs, to explore heterogeneity amongst populations and identify key molecular regulators of tolerogenic programming. This identified tolerogenic programmes (including IDO1) to be upregulated in migrated LCs, alongside enhanced immunocompetent programming compared to steady-state LCs. The latter populations were split into two subpopulations defined by immaturity and immunocompetency. Transcription factors (TFs) that correlated with enhanced migrated LC tolerogenic programmes included IRF4 and RELB.

Using in vitro experimentation, the importance of LC immunocompetency for mediation of Treg induction was revealed in steady-state LC populations (Immunocompetent/CD86High and Immature/CD86Low). Furthermore, immunocompetent migrated LCs displayed enhanced induction of functionally suppressive Tregs. Inhibition of IDO1 reduced migrated LC tolerogenic potential, revealing the criticalness of tolerogenic programming for LC tolerogenicity.

Mathematical modelling using TFs identified from analyses thought to control
immunogenic (IRF1±IRF4) vs tolerogenic (IRF4, MAP3K14, RELB) programming reflected observations from previous studies in which unstimulated migrated LC display both immunogenic and tolerogenic potential whilst, inflammatory stimuli (TNFa) increases favouring of immunogenic responses.

Overall, our analysis identified critical mechanisms which equip LCs with tolerogenic function and revealed potential mechanisms by which immunogenic vs tolerogenic responses are initiated.
University of Southampton
Davies, James David
a93b4fc9-80a2-4620-ada6-c12f05c5ee38
Davies, James David
a93b4fc9-80a2-4620-ada6-c12f05c5ee38
Polak, Marta
e0ac5e1a-7074-4776-ba23-490bd4da612d
Macarthur, Benjamin
2c0476e7-5d3e-4064-81bb-104e8e88bb6b
Davies, Donna
7de8fdc7-3640-4e3a-aa91-d0e03f990c38

Davies, James David (2020) A system immunology approach to understanding human langerhans cell tolerogenic function. Doctoral Thesis, 302pp.

Record type: Thesis (Doctoral)

Abstract

Langerhans cells (LC) maintain skin homeostasis through orchestrating immunogenic and tolerogenic immune responses in steady-state epidermis and at local lymph nodes after migration.While the mechanisms promoting activation of immune responses has been elucidated, little is known about the molecular mechanisms underpinning LC induced tolerance. We hypothesised that heterogeneous human LC populations existed in situ, which specialised in regulation of immunogenic vs tolerogenic regulation in healthy skin. Within the transcriptome of tolerance regulating LC, we sought to identify key molecular mediators of tolerogenic programming.

Previously published transcriptomic data containing a wide selection of DC subpopulations, including LC was analysed. Here, core mechanisms of tolerogenic programming across DCs was investigated. Overall, LC transcriptomic programming was largely distinct, although common pathways of suppression of stimuli responsiveness were identified in steady-state LCs.

The Drop-seq scRNA-seq protocol was optimised and implemented on steady-state and migrated LCs, to explore heterogeneity amongst populations and identify key molecular regulators of tolerogenic programming. This identified tolerogenic programmes (including IDO1) to be upregulated in migrated LCs, alongside enhanced immunocompetent programming compared to steady-state LCs. The latter populations were split into two subpopulations defined by immaturity and immunocompetency. Transcription factors (TFs) that correlated with enhanced migrated LC tolerogenic programmes included IRF4 and RELB.

Using in vitro experimentation, the importance of LC immunocompetency for mediation of Treg induction was revealed in steady-state LC populations (Immunocompetent/CD86High and Immature/CD86Low). Furthermore, immunocompetent migrated LCs displayed enhanced induction of functionally suppressive Tregs. Inhibition of IDO1 reduced migrated LC tolerogenic potential, revealing the criticalness of tolerogenic programming for LC tolerogenicity.

Mathematical modelling using TFs identified from analyses thought to control
immunogenic (IRF1±IRF4) vs tolerogenic (IRF4, MAP3K14, RELB) programming reflected observations from previous studies in which unstimulated migrated LC display both immunogenic and tolerogenic potential whilst, inflammatory stimuli (TNFa) increases favouring of immunogenic responses.

Overall, our analysis identified critical mechanisms which equip LCs with tolerogenic function and revealed potential mechanisms by which immunogenic vs tolerogenic responses are initiated.

Text
A SYSTEMS IMMUNOLOGY APPROACH TO UNDERSTANDING HUMAN LANGERHANS CELL TOLEROGENIC FUNCTION - Version of Record
Available under License University of Southampton Thesis Licence.
Download (48MB)
Text
Declaration of Authorship
Restricted to Repository staff only
Text
Permission to deposit thesis form
Restricted to Repository staff only

More information

Published date: August 2020

Identifiers

Local EPrints ID: 447763
URI: http://eprints.soton.ac.uk/id/eprint/447763
PURE UUID: 9ad285ce-06dd-49f6-87fa-724737b1e105
ORCID for Benjamin Macarthur: ORCID iD orcid.org/0000-0002-5396-9750
ORCID for Donna Davies: ORCID iD orcid.org/0000-0002-5117-2991

Catalogue record

Date deposited: 19 Mar 2021 17:33
Last modified: 17 Mar 2024 02:51

Export record

Contributors

Author: James David Davies
Thesis advisor: Marta Polak
Thesis advisor: Benjamin Macarthur ORCID iD
Thesis advisor: Donna Davies 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.

×