Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Lung microbiota promotes tolerance to allergens in neonates via PD-L1

Abstract

Epidemiological data point toward a critical period in early life during which environmental cues can set an individual on a trajectory toward respiratory health or disease1,2,3,4,5,6,7,8. The neonatal immune system matures during this period9, although little is known about the signals that lead to its maturation. Here we report that the formation of the lung microbiota is a key parameter in this process. Immediately following birth, neonatal mice were prone to develop exaggerated airway eosinophilia, release type 2 helper T cell cytokines and exhibit airway hyper-responsiveness following exposure to house dust mite allergens, even though their lungs harbored high numbers of natural CD4+Foxp3+CD25+Helios+ regulatory T (Treg) cells. During the first 2 weeks after birth, the bacterial load in the lungs increased, and representation of the bacterial phyla shifts from a predominance of Gammaproteobacteria and Firmicutes towards Bacteroidetes. The changes in the microbiota were associated with decreased aeroallergen responsiveness and the emergence of a Helios Treg cell subset that required interaction with programmed death ligand 1 (PD-L1) for development. Absence of microbial colonization10 or blockade of PD-L1 during the first 2 weeks postpartum maintained exaggerated responsiveness to allergens through to adulthood. Adoptive transfer of Treg cells from adult mice to neonates before aeroallergen exposure ameliorated disease. Thus, formation of the airway microbiota induces regulatory cells early in life, which, when dysregulated, can lead to sustained susceptibility to allergic airway inflammation in adulthood.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Neonatal mice develop exaggerated allergic airway inflammation after exposure to HDM.
Figure 2: PD-L1 is transiently expressed on CD11b+ DCs in the lung.
Figure 3: The microbiota promotes transient PD-L1 expression and induces Treg cells in the lung.
Figure 4: Blockade of PD-L1 activity during the neonatal period maintains the exaggerated responsiveness to HDM through adulthood.

Similar content being viewed by others

References

  1. von Mutius, E. & Radon, K. Living on a farm: impact on asthma induction and clinical course. Immunol. Allergy Clin. North Am. 28, 631–647, ix–x (2008).

    Article  Google Scholar 

  2. von Mutius, E. & Vercelli, D. Farm living: effects on childhood asthma and allergy. Nat. Rev. Immunol. 10, 861–868 (2010).

    Article  CAS  Google Scholar 

  3. Burke, H. et al. Prenatal and passive smoke exposure and incidence of asthma and wheeze: systematic review and meta-analysis. Pediatrics 129, 735–744 (2012).

    Article  Google Scholar 

  4. Lee, S.L. et al. Foetal exposure to maternal passive smoking is associated with childhood asthma, allergic rhinitis, and eczema. ScientificWorldJournal 2012, 542983 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Stensballe, L.G., Simonsen, J., Jensen, S.M., Bonnelykke, K. & Bisgaard, H. Use of antibiotics during pregnancy increases the risk of asthma in early childhood. J. Pediatr. 162, 832–838.e3 (2013).

    Article  CAS  Google Scholar 

  6. Gern, J.E., Rosenthal, L.A., Sorkness, R.L. & Lemanske, R.F. Jr. Effects of viral respiratory infections on lung development and childhood asthma. J. Allergy Clin. Immunol. 115, 668–674, quiz 675 (2005).

    Article  Google Scholar 

  7. Jackson, D.J. The role of rhinovirus infections in the development of early childhood asthma. Curr. Opin. Allergy Clin. Immunol. 10, 133–138 (2010).

    Article  Google Scholar 

  8. Wu, P. & Hartert, T.V. Evidence for a causal relationship between respiratory syncytial virus infection and asthma. Expert Rev. Anti Infect. Ther. 9, 731–745 (2011).

    Article  Google Scholar 

  9. Holt, P.G. & Jones, C.A. The development of the immune system during pregnancy and early life. Allergy 55, 688–697 (2000).

    Article  CAS  Google Scholar 

  10. Herbst, T. et al. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am. J. Respir. Crit. Care Med. 184, 198–205 (2011).

    Article  CAS  Google Scholar 

  11. Hooper, L.V., Littman, D.R. & Macpherson, A.J. Interactions between the microbiota and the immune system. Science 336, 1268–1273 (2012).

    Article  CAS  Google Scholar 

  12. Macpherson, A.J. & Harris, N.L. Interactions between commensal intestinal bacteria and the immune system. Nat. Rev. Immunol. 4, 478–485 (2004).

    Article  CAS  Google Scholar 

  13. Hilty, M. et al. Disordered microbial communities in asthmatic airways. PLoS ONE 5, e8578 (2010).

    Article  Google Scholar 

  14. Huang, Y.J. et al. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J. Allergy Clin Immunol. 127, 372–381.e1–e3 (2011).

    Article  Google Scholar 

  15. Pragman, A.A., Kim, H.B., Reilly, C.S., Wendt, C. & Isaacson, R.E. The lung microbiome in moderate and severe chronic obstructive pulmonary disease. PLoS ONE 7, e47305 (2012).

    Article  CAS  Google Scholar 

  16. Erb-Downward, J.R. et al. Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS ONE 6, e16384 (2011).

    Article  CAS  Google Scholar 

  17. Marsland, B.J., Yadava, K. & Nicod, L.P. The airway microbiome and disease. Chest 144, 632–637 (2013).

    Article  Google Scholar 

  18. Twigg, H.L. III et al. Use of bronchoalveolar lavage to assess the respiratory microbiome: signal in the noise. Lancet Respir. Med. 1, 354–356 (2013).

    Article  Google Scholar 

  19. Trompette, A. et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 20, 159–166 (2014).

    Article  CAS  Google Scholar 

  20. Francisco, L.M. et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J. Exp. Med. 206, 3015–3029 (2009).

    Article  CAS  Google Scholar 

  21. Wang, L. et al. Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells. Proc. Natl. Acad. Sci. USA 105, 9331–9336 (2008).

    Article  CAS  Google Scholar 

  22. Geuking, M.B. et al. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 34, 794–806 (2011).

    Article  CAS  Google Scholar 

  23. Round, J.L. & Mazmanian, S.K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. USA 107, 12204–12209 (2010).

    Article  CAS  Google Scholar 

  24. Curotto de Lafaille, M.A. et al. Adaptive Foxp3+ regulatory T cell-dependent and -independent control of allergic inflammation. Immunity 29, 114–126 (2008).

    Article  CAS  Google Scholar 

  25. Curotto de Lafaille, M.A. & Lafaille, J.J. Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity 30, 626–635 (2009).

    Article  CAS  Google Scholar 

  26. Rangel-Moreno, J. et al. The development of inducible bronchus-associated lymphoid tissue depends on IL-17. Nat. Immunol. 12, 639–646 (2011).

    Article  CAS  Google Scholar 

  27. Olszak, T. et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489–493 (2012).

    Article  CAS  Google Scholar 

  28. Cahenzli, J., Koller, Y., Wyss, M., Geuking, M.B. & McCoy, K.D. Intestinal microbial diversity during early-life colonization shapes long-term IgE levels. Cell Host Microbe 14, 559–570 (2013).

    Article  CAS  Google Scholar 

  29. Nembrini, C. et al. Bacterial-induced protection against allergic inflammation through a multicomponent immunoregulatory mechanism. Thorax 66, 755–763 (2011).

    Article  Google Scholar 

  30. Hagner, S. et al. Farm-derived gram-positive bacterium Staphylococcus sciuri W620 prevents asthma phenotype in HDM- and OVA-exposed mice. Allergy 68, 322–329 (2013).

    Article  CAS  Google Scholar 

  31. Bacchetti De Gregoris, T., Aldred, N., Clare, A.S. & Burgess, J.G. Improvement of phylum- and class-specific primers for real-time PCR quantification of bacterial taxa. J. Microbiol. Methods 86, 351–356 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work is supported by the Leenaards Foundation in Lausanne and the Swiss National Science Foundation grant 310030_146983, awarded to B.J.M. B.J.M. is a Cloetta Medical Research Fellow and, by holding this title, receives financial support. B.J.M. and C.M.L. are part of the European Cooperation in Science and Technology Action BM1201, Developmental Origins of Chronic Lung Disease. S.S. and C.M.L. are supported by the Wellcome Trust grants 087618/Z/08/Z and 083586/Z/07/Z. C.M.L. is a Wellcome Senior Fellow in Basic Biomedical Science and, by holding this title, receives financial support. We thank D. Pinschewer and S. Kallert for valuable discussions and critical feedback.

Author information

Authors and Affiliations

Authors

Contributions

B.J.M. conceived the study. B.J.M. and E.S.G. designed the study. E.S.G., A.T. and K.Y. performed experiments. S.S. and R.S. performed neonatal lung function experiments. K.D.M. provided germ-free mice. E.S.G., A.T., K.Y., S.S., C.M.L., L.P.N. and B.J.M. provided critical analysis and discussion. E.S.G. and B.J.M. wrote the paper.

Corresponding author

Correspondence to Benjamin J Marsland.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 652 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gollwitzer, E., Saglani, S., Trompette, A. et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med 20, 642–647 (2014). https://doi.org/10.1038/nm.3568

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3568

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing