The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis

Tuberculosis (TB), caused by the airborne bacterial pathogen Mycobacterium tuberculosis, remains a major source of morbidity and mortality worldwide. So far, the study of host-pathogen interactions in TB has mostly focused on the physiology and virulence of the pathogen, as well as, on the various i...

Full description

Bibliographic Details
Main Authors: Alexia Dumas, Dan Corral, André Colom, Florence Levillain, Antonio Peixoto, Denis Hudrisier, Yannick Poquet, Olivier Neyrolles
Format: Article
Language:English
Published: Frontiers Media S.A. 2018-11-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fimmu.2018.02656/full
id doaj-6254d4ed8b4c4acbaf916c408f4a345c
record_format Article
spelling doaj-6254d4ed8b4c4acbaf916c408f4a345c2020-11-25T02:15:43ZengFrontiers Media S.A.Frontiers in Immunology1664-32242018-11-01910.3389/fimmu.2018.02656411523The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosisAlexia DumasDan CorralAndré ColomFlorence LevillainAntonio PeixotoDenis HudrisierYannick PoquetOlivier NeyrollesTuberculosis (TB), caused by the airborne bacterial pathogen Mycobacterium tuberculosis, remains a major source of morbidity and mortality worldwide. So far, the study of host-pathogen interactions in TB has mostly focused on the physiology and virulence of the pathogen, as well as, on the various innate and adaptive immune compartments of the host. Microbial organisms endogenous to our body, the so-called microbiota, interact not only with invading pathogens, but also with our immune system. Yet, the impact of the microbiota on host defense against M. tuberculosis remains poorly understood. In order to address this question, we adapted a robust and reproducible mouse model of microbial dysbiosis based on a combination of wide-spectrum antibiotics. We found that microbiota dysbiosis resulted in an increased early colonization of the lungs by M. tuberculosis during the first week of infection, correlating with an altered diversity of the gut microbiota during this time period. At the cellular level, no significant difference in the recruitment of conventional myeloid cells, including macrophages, dendritic cells and neutrophils, to the lungs could be detected during the first week of infection between microbiota-competent and -deficient mice. At the molecular level, microbiota depletion did not impact the global production of pro-inflammatory cytokines, such as interferon (IFN)γ, tumor necrosis factor (TNF)α and interleukin (IL)-1β in the lungs. Strikingly, a reduced number of mucosal-associated invariant T (MAIT) cells, a population of innate-like lymphocytes whose development is known to depend on the host microbiota, was observed in the lungs of the antibiotics-treated animals after 1week of infection. These cells produced less IL-17A in antibiotics-treated mice. Notably, dysbiosis correction through the inoculation of a complex microbiota in antibiotics-treated animals reversed these phenotypes and improved the ability of MAIT cells to proliferate. Altogether, our results demonstrate that the host microbiota contributes to early protection of lung colonization by M. tuberculosis, possibly through sustaining the function(s) of MAIT cells. Our study calls for a better understanding of the impact of the microbiota on host-pathogen interactions in TB. Ultimately, this study may help to develop novel therapeutic approaches based on the use of beneficial microbes, or components thereof, to boost anti-mycobacterial immunity.https://www.frontiersin.org/article/10.3389/fimmu.2018.02656/fullmicrobiotaMAIT cellsmacrophagetuberculosisIL-17
collection DOAJ
language English
format Article
sources DOAJ
author Alexia Dumas
Dan Corral
André Colom
Florence Levillain
Antonio Peixoto
Denis Hudrisier
Yannick Poquet
Olivier Neyrolles
spellingShingle Alexia Dumas
Dan Corral
André Colom
Florence Levillain
Antonio Peixoto
Denis Hudrisier
Yannick Poquet
Olivier Neyrolles
The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis
Frontiers in Immunology
microbiota
MAIT cells
macrophage
tuberculosis
IL-17
author_facet Alexia Dumas
Dan Corral
André Colom
Florence Levillain
Antonio Peixoto
Denis Hudrisier
Yannick Poquet
Olivier Neyrolles
author_sort Alexia Dumas
title The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis
title_short The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis
title_full The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis
title_fullStr The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis
title_full_unstemmed The Host Microbiota Contributes to Early Protection Against Lung Colonization by Mycobacterium tuberculosis
title_sort host microbiota contributes to early protection against lung colonization by mycobacterium tuberculosis
publisher Frontiers Media S.A.
series Frontiers in Immunology
issn 1664-3224
publishDate 2018-11-01
description Tuberculosis (TB), caused by the airborne bacterial pathogen Mycobacterium tuberculosis, remains a major source of morbidity and mortality worldwide. So far, the study of host-pathogen interactions in TB has mostly focused on the physiology and virulence of the pathogen, as well as, on the various innate and adaptive immune compartments of the host. Microbial organisms endogenous to our body, the so-called microbiota, interact not only with invading pathogens, but also with our immune system. Yet, the impact of the microbiota on host defense against M. tuberculosis remains poorly understood. In order to address this question, we adapted a robust and reproducible mouse model of microbial dysbiosis based on a combination of wide-spectrum antibiotics. We found that microbiota dysbiosis resulted in an increased early colonization of the lungs by M. tuberculosis during the first week of infection, correlating with an altered diversity of the gut microbiota during this time period. At the cellular level, no significant difference in the recruitment of conventional myeloid cells, including macrophages, dendritic cells and neutrophils, to the lungs could be detected during the first week of infection between microbiota-competent and -deficient mice. At the molecular level, microbiota depletion did not impact the global production of pro-inflammatory cytokines, such as interferon (IFN)γ, tumor necrosis factor (TNF)α and interleukin (IL)-1β in the lungs. Strikingly, a reduced number of mucosal-associated invariant T (MAIT) cells, a population of innate-like lymphocytes whose development is known to depend on the host microbiota, was observed in the lungs of the antibiotics-treated animals after 1week of infection. These cells produced less IL-17A in antibiotics-treated mice. Notably, dysbiosis correction through the inoculation of a complex microbiota in antibiotics-treated animals reversed these phenotypes and improved the ability of MAIT cells to proliferate. Altogether, our results demonstrate that the host microbiota contributes to early protection of lung colonization by M. tuberculosis, possibly through sustaining the function(s) of MAIT cells. Our study calls for a better understanding of the impact of the microbiota on host-pathogen interactions in TB. Ultimately, this study may help to develop novel therapeutic approaches based on the use of beneficial microbes, or components thereof, to boost anti-mycobacterial immunity.
topic microbiota
MAIT cells
macrophage
tuberculosis
IL-17
url https://www.frontiersin.org/article/10.3389/fimmu.2018.02656/full
work_keys_str_mv AT alexiadumas thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT dancorral thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT andrecolom thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT florencelevillain thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT antoniopeixoto thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT denishudrisier thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT yannickpoquet thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT olivierneyrolles thehostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT alexiadumas hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT dancorral hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT andrecolom hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT florencelevillain hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT antoniopeixoto hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT denishudrisier hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT yannickpoquet hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
AT olivierneyrolles hostmicrobiotacontributestoearlyprotectionagainstlungcolonizationbymycobacteriumtuberculosis
_version_ 1724894220769034240