Antibiotic-related gut dysbiosis induces lung immunodepression and worsens lung infection in mice.

Détails

Ressource 1Télécharger: 33076936_BIB_B0E4D6B6DD9C.pdf (2317.05 [Ko])
Etat: Public
Version: Final published version
Licence: CC BY 4.0
ID Serval
serval:BIB_B0E4D6B6DD9C
Type
Article: article d'un périodique ou d'un magazine.
Collection
Publications
Institution
Titre
Antibiotic-related gut dysbiosis induces lung immunodepression and worsens lung infection in mice.
Périodique
Critical care
Auteur⸱e⸱s
Dessein R., Bauduin M., Grandjean T., Le Guern R., Figeac M., Beury D., Faure K., Faveeuw C., Guery B., Gosset P., Kipnis E.
ISSN
1466-609X (Electronic)
ISSN-L
1364-8535
Statut éditorial
Publié
Date de publication
15/10/2020
Peer-reviewed
Oui
Volume
24
Numéro
1
Pages
611
Langue
anglais
Notes
Publication types: Journal Article
Publication Status: epublish
Résumé
Gut dysbiosis due to the adverse effects of antibiotics affects outcomes of lung infection. Previous murine models relied on significant depletion of both gut and lung microbiota, rendering the analysis of immune gut-lung cross-talk difficult. Here, we study the effects of antibiotic-induced gut dysbiosis without lung dysbiosis on lung immunity and the consequences on acute P. aeruginosa lung infection.
C57BL6 mice received 7 days oral vancomycin-colistin, followed by normal regimen or fecal microbial transplant or Fms-related tyrosine kinase 3 ligand (Flt3-Ligand) over 2 days, and then intra-nasal P. aeruginosa strain PAO1. Gut and lung microbiota were studied by next-generation sequencing, and lung infection outcomes were studied at 24 h. Effects of vancomycin-colistin on underlying immunity and bone marrow progenitors were studied in uninfected mice by flow cytometry in the lung, spleen, and bone marrow.
Vancomycin-colistin administration induces widespread cellular immunosuppression in both the lung and spleen, decreases circulating hematopoietic cytokine Flt3-Ligand, and depresses dendritic cell bone marrow progenitors leading to worsening of P. aeruginosa lung infection outcomes (bacterial loads, lung injury, and survival). Reversal of these effects by fecal microbial transplant shows that these alterations are related to gut dysbiosis. Recombinant Flt3-Ligand reverses the effects of antibiotics on subsequent lung infection.
These results show that gut dysbiosis strongly impairs monocyte/dendritic progenitors and lung immunity, worsening outcomes of P. aeruginosa lung infection. Treatment with a fecal microbial transplant or immune stimulation by Flt3-Ligand both restore lung cellular responses to and outcomes of P. aeruginosa following antibiotic-induced gut dysbiosis.
Mots-clé
Antibiotics, Dysbiosis, Fecal microbial transplant, Flt3-ligand, Murine model, Pseudomonas aeruginosa
Pubmed
Web of science
Open Access
Oui
Création de la notice
26/10/2020 15:01
Dernière modification de la notice
30/04/2021 7:14
Données d'usage