Skip to main content
Top
Published in: Critical Care 1/2020

01-12-2020 | Antibiotic | Research

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

Authors: Rodrigue Dessein, Marvin Bauduin, Teddy Grandjean, Rémi Le Guern, Martin Figeac, Delphine Beury, Karine Faure, Christelle Faveeuw, Benoit Guery, Philippe Gosset, Eric Kipnis

Published in: Critical Care | Issue 1/2020

Login to get access

Abstract

Background

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.

Methods

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.

Results

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.

Conclusions

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.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sekirov I, Russell SL, Antunes LCM, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90:859–904.CrossRef Sekirov I, Russell SL, Antunes LCM, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90:859–904.CrossRef
2.
go back to reference Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17:219–32.CrossRef Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17:219–32.CrossRef
3.
go back to reference Willing BP, Russell SL, Finlay BB. Shifting the balance: antibiotic effects on host-microbiota mutualism. Nat Rev Microbiol. 2011;9:233–43.CrossRef Willing BP, Russell SL, Finlay BB. Shifting the balance: antibiotic effects on host-microbiota mutualism. Nat Rev Microbiol. 2011;9:233–43.CrossRef
4.
go back to reference Dang AT, Marsland BJ. Microbes, metabolites, and the gut-lung axis. Mucosal Immunol. 2019;352:539. Dang AT, Marsland BJ. Microbes, metabolites, and the gut-lung axis. Mucosal Immunol. 2019;352:539.
5.
go back to reference Robak OH, Heimesaat MM, Kruglov AA, Prepens S, Ninnemann J, Gutbier B, et al. Antibiotic treatment-induced secondary IgA deficiency enhances susceptibility to Pseudomonas aeruginosa pneumonia. J Clin Invest. 2018;128:3535–45.CrossRef Robak OH, Heimesaat MM, Kruglov AA, Prepens S, Ninnemann J, Gutbier B, et al. Antibiotic treatment-induced secondary IgA deficiency enhances susceptibility to Pseudomonas aeruginosa pneumonia. J Clin Invest. 2018;128:3535–45.CrossRef
6.
go back to reference Brown RL, Sequeira RP, Clarke TB. The microbiota protects against respiratory infection via GM-CSF signaling. Nat Commun. 2017;8:1512.CrossRef Brown RL, Sequeira RP, Clarke TB. The microbiota protects against respiratory infection via GM-CSF signaling. Nat Commun. 2017;8:1512.CrossRef
7.
go back to reference Boutoille D, Marechal X, Pichenot M, Chemani C, Guery B, Faure K. FITC-albumin as a marker for assessment of endothelial permeability in mice: comparison with 125I-albumin. Exp Lung Res. 2009;35:263–71.CrossRef Boutoille D, Marechal X, Pichenot M, Chemani C, Guery B, Faure K. FITC-albumin as a marker for assessment of endothelial permeability in mice: comparison with 125I-albumin. Exp Lung Res. 2009;35:263–71.CrossRef
8.
go back to reference Fagundes CT, Amaral FA, Vieira AT, Soares AC, Pinho V, Nicoli JR, et al. Transient TLR activation restores inflammatory response and ability to control pulmonary bacterial infection in germfree mice. J Immunol. 2012;188:1411–20.CrossRef Fagundes CT, Amaral FA, Vieira AT, Soares AC, Pinho V, Nicoli JR, et al. Transient TLR activation restores inflammatory response and ability to control pulmonary bacterial infection in germfree mice. J Immunol. 2012;188:1411–20.CrossRef
9.
go back to reference Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJTH, de Boer JD, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016;65:575–583. Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJTH, de Boer JD, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016;65:575–583.
10.
go back to reference Russell SL, Gold MJ, Willing BP, Thorson L, McNagny KM, Finlay BB. Perinatal antibiotic treatment affects murine microbiota, immune responses and allergic asthma. Gut Microbes. 2013;4:158–64.CrossRef Russell SL, Gold MJ, Willing BP, Thorson L, McNagny KM, Finlay BB. Perinatal antibiotic treatment affects murine microbiota, immune responses and allergic asthma. Gut Microbes. 2013;4:158–64.CrossRef
11.
go back to reference Josefsdottir KS, Baldridge MT, Kadmon CS, King KY. Antibiotics impair murine hematopoiesis by depleting the intestinal microbiota. Blood. 2017;129:729–39.CrossRef Josefsdottir KS, Baldridge MT, Kadmon CS, King KY. Antibiotics impair murine hematopoiesis by depleting the intestinal microbiota. Blood. 2017;129:729–39.CrossRef
12.
go back to reference Thackray LB, Handley SA, Gorman MJ, Poddar S, Bagadia P, Briseño CG, et al. Oral antibiotic treatment of mice exacerbates the disease severity of multiple flavivirus infections. Cell Rep. 2018;22:3440–6.CrossRef Thackray LB, Handley SA, Gorman MJ, Poddar S, Bagadia P, Briseño CG, et al. Oral antibiotic treatment of mice exacerbates the disease severity of multiple flavivirus infections. Cell Rep. 2018;22:3440–6.CrossRef
13.
go back to reference Tsapogas P, Mooney CJ, Brown G, Rolink A. The cytokine Flt3-ligand in normal and malignant hematopoiesis. Int J Mol Sci. 2017;18:1115.CrossRef Tsapogas P, Mooney CJ, Brown G, Rolink A. The cytokine Flt3-ligand in normal and malignant hematopoiesis. Int J Mol Sci. 2017;18:1115.CrossRef
14.
go back to reference Tsapogas P, Swee LK, Nusser A, Nuber N, Kreuzaler M, Capoferri G, et al. In vivo evidence for an instructive role of fms-like tyrosine kinase-3 (FLT3) ligand in hematopoietic development. Haematologica. 2014;99:638–46.CrossRef Tsapogas P, Swee LK, Nusser A, Nuber N, Kreuzaler M, Capoferri G, et al. In vivo evidence for an instructive role of fms-like tyrosine kinase-3 (FLT3) ligand in hematopoietic development. Haematologica. 2014;99:638–46.CrossRef
15.
go back to reference Maraskovsky E, Brasel K, Teepe M, Roux ER, Lyman SD, Shortman K, et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J Exp Med. 1996;184:1953–62.CrossRef Maraskovsky E, Brasel K, Teepe M, Roux ER, Lyman SD, Shortman K, et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J Exp Med. 1996;184:1953–62.CrossRef
16.
go back to reference Beshara R, Sencio V, Soulard D, Barthélémy A, Fontaine J, Pinteau T, et al. Alteration of Flt3-ligand-dependent de novo generation of conventional dendritic cells during influenza infection contributes to respiratory bacterial superinfection. Sant AJ, editor. PLoS Pathog. 2018;14:e1007360.CrossRef Beshara R, Sencio V, Soulard D, Barthélémy A, Fontaine J, Pinteau T, et al. Alteration of Flt3-ligand-dependent de novo generation of conventional dendritic cells during influenza infection contributes to respiratory bacterial superinfection. Sant AJ, editor. PLoS Pathog. 2018;14:e1007360.CrossRef
17.
go back to reference DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381:2043–50.CrossRef DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381:2043–50.CrossRef
18.
go back to reference Roquilly A, Torres A, Villadangos JA, Netea MG, Dickson R, Becher B, et al. Pathophysiological role of respiratory dysbiosis in hospital-acquired pneumonia. Lancet Respir Med. 2019;7:710–20.CrossRef Roquilly A, Torres A, Villadangos JA, Netea MG, Dickson R, Becher B, et al. Pathophysiological role of respiratory dysbiosis in hospital-acquired pneumonia. Lancet Respir Med. 2019;7:710–20.CrossRef
Metadata
Title
Antibiotic-related gut dysbiosis induces lung immunodepression and worsens lung infection in mice
Authors
Rodrigue Dessein
Marvin Bauduin
Teddy Grandjean
Rémi Le Guern
Martin Figeac
Delphine Beury
Karine Faure
Christelle Faveeuw
Benoit Guery
Philippe Gosset
Eric Kipnis
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Critical Care / Issue 1/2020
Electronic ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-020-03320-8

Other articles of this Issue 1/2020

Critical Care 1/2020 Go to the issue