Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.

The development of an effective vaccine against tuberculosis (Tb) represents one of the major medical challenges of this century. Mycobacterium bovis Bacille Calmette-Guerin (BCG), the only vaccine available at present, is mostly effective at preventing disseminated Tb in children, but shows variabl...

Full description

Bibliographic Details
Main Authors: Luciana Berod, Philipp Stüve, Filipa Varela, Jochen Behrends, Maxine Swallow, Friederike Kruse, Freyja Krull, Peyman Ghorbani, Christian T Mayer, Christoph Hölscher, Tim Sparwasser
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2014-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC4106855?pdf=render
id doaj-9c867284bf9842eca0a4ad860381e6e4
record_format Article
spelling doaj-9c867284bf9842eca0a4ad860381e6e42020-11-24T21:38:55ZengPublic Library of Science (PLoS)PLoS ONE1932-62032014-01-0197e10280410.1371/journal.pone.0102804Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.Luciana BerodPhilipp StüveFilipa VarelaJochen BehrendsMaxine SwallowFriederike KruseFreyja KrullPeyman GhorbaniChristian T MayerChristoph HölscherTim SparwasserThe development of an effective vaccine against tuberculosis (Tb) represents one of the major medical challenges of this century. Mycobacterium bovis Bacille Calmette-Guerin (BCG), the only vaccine available at present, is mostly effective at preventing disseminated Tb in children, but shows variable protection against pulmonary Tb, the most common form in adults. The reasons for this poor efficacy are not completely understood, but there is evidence that T regulatory cells (Tregs) might be involved. Similarly, Tregs have been associated with the immunosuppression observed in patients infected with Tb and are therefore believed to play a role in pathogen persistence. Thus, Treg depletion has been postulated as a novel strategy to potentiate M. bovis BCG vaccination on one side, while on the other, employed as a therapeutic approach during chronic Tb infection. Yet since Tregs are critically involved in controlling autoimmune inflammation, elimination of Tregs may therefore also incur the danger of an excessive inflammatory immune response. Thus, understanding the dynamics and function of Tregs during mycobacterial infection is crucial to evaluate the potential of Treg depletion as a medical option. To address this, we depleted Tregs after infection with M. bovis BCG or Mycobacterium tuberculosis (Mtb) using DEREG mice, which express the diphtheria toxin (DT) receptor under the control of the FoxP3 locus, thereby allowing the selective depletion of FoxP3+ Tregs. Our results show that after depletion, the Treg niche is rapidly refilled by a population of DT-insensitive Tregs (diTregs) and bacterial load remains unchanged. On the contrary, impaired rebound of Tregs in DEREG × FoxP3GFP mice improves pathogen burden, but is accompanied by detrimental autoimmune inflammation. Therefore, our study provides the proof-of-principle that, although a high degree of Treg depletion may contribute to the control of mycobacterial infection, it carries the risk of autoimmunity.http://europepmc.org/articles/PMC4106855?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Luciana Berod
Philipp Stüve
Filipa Varela
Jochen Behrends
Maxine Swallow
Friederike Kruse
Freyja Krull
Peyman Ghorbani
Christian T Mayer
Christoph Hölscher
Tim Sparwasser
spellingShingle Luciana Berod
Philipp Stüve
Filipa Varela
Jochen Behrends
Maxine Swallow
Friederike Kruse
Freyja Krull
Peyman Ghorbani
Christian T Mayer
Christoph Hölscher
Tim Sparwasser
Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.
PLoS ONE
author_facet Luciana Berod
Philipp Stüve
Filipa Varela
Jochen Behrends
Maxine Swallow
Friederike Kruse
Freyja Krull
Peyman Ghorbani
Christian T Mayer
Christoph Hölscher
Tim Sparwasser
author_sort Luciana Berod
title Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.
title_short Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.
title_full Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.
title_fullStr Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.
title_full_unstemmed Rapid rebound of the Treg compartment in DEREG mice limits the impact of Treg depletion on mycobacterial burden, but prevents autoimmunity.
title_sort rapid rebound of the treg compartment in dereg mice limits the impact of treg depletion on mycobacterial burden, but prevents autoimmunity.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2014-01-01
description The development of an effective vaccine against tuberculosis (Tb) represents one of the major medical challenges of this century. Mycobacterium bovis Bacille Calmette-Guerin (BCG), the only vaccine available at present, is mostly effective at preventing disseminated Tb in children, but shows variable protection against pulmonary Tb, the most common form in adults. The reasons for this poor efficacy are not completely understood, but there is evidence that T regulatory cells (Tregs) might be involved. Similarly, Tregs have been associated with the immunosuppression observed in patients infected with Tb and are therefore believed to play a role in pathogen persistence. Thus, Treg depletion has been postulated as a novel strategy to potentiate M. bovis BCG vaccination on one side, while on the other, employed as a therapeutic approach during chronic Tb infection. Yet since Tregs are critically involved in controlling autoimmune inflammation, elimination of Tregs may therefore also incur the danger of an excessive inflammatory immune response. Thus, understanding the dynamics and function of Tregs during mycobacterial infection is crucial to evaluate the potential of Treg depletion as a medical option. To address this, we depleted Tregs after infection with M. bovis BCG or Mycobacterium tuberculosis (Mtb) using DEREG mice, which express the diphtheria toxin (DT) receptor under the control of the FoxP3 locus, thereby allowing the selective depletion of FoxP3+ Tregs. Our results show that after depletion, the Treg niche is rapidly refilled by a population of DT-insensitive Tregs (diTregs) and bacterial load remains unchanged. On the contrary, impaired rebound of Tregs in DEREG × FoxP3GFP mice improves pathogen burden, but is accompanied by detrimental autoimmune inflammation. Therefore, our study provides the proof-of-principle that, although a high degree of Treg depletion may contribute to the control of mycobacterial infection, it carries the risk of autoimmunity.
url http://europepmc.org/articles/PMC4106855?pdf=render
work_keys_str_mv AT lucianaberod rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT philippstuve rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT filipavarela rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT jochenbehrends rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT maxineswallow rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT friederikekruse rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT freyjakrull rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT peymanghorbani rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT christiantmayer rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT christophholscher rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
AT timsparwasser rapidreboundofthetregcompartmentinderegmicelimitstheimpactoftregdepletiononmycobacterialburdenbutpreventsautoimmunity
_version_ 1725933783465590784