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.

  • Article
  • Published:

Early-life viral infections are associated with disadvantageous immune and microbiota profiles and recurrent respiratory infections

Abstract

The respiratory tract is populated by a specialized microbial ecosystem, which is seeded during and directly following birth. Perturbed development of the respiratory microbial community in early-life has been associated with higher susceptibility to respiratory tract infections (RTIs). Given a consistent gap in time between first signs of aberrant microbial maturation and the observation of the first RTIs, we hypothesized that early-life host–microbe cross-talk plays a role in this process. We therefore investigated viral presence, gene expression profiles and nasopharyngeal microbiota from birth until 12 months of age in 114 healthy infants. We show that the strongest dynamics in gene expression profiles occurred within the first days of life, mostly involving Toll-like receptor (TLR) and inflammasome signalling. These gene expression dynamics coincided with rapid microbial niche differentiation. Early asymptomatic viral infection co-occurred with stronger interferon activity, which was related to specific microbiota dynamics following, including early enrichment of Moraxella and Haemophilus spp. These microbial trajectories were in turn related to a higher number of subsequent (viral) RTIs over the first year of life. Using a multi-omic approach, we found evidence for species-specific host–microbe interactions related to consecutive susceptibility to RTIs. Although further work will be needed to confirm causality of our findings, together these data indicate that early-life viral encounters could impact subsequent host–microbe cross-talk, which is linked to later-life infections.

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

Fig. 1: Differentially expressed genes (DEGs) and gene module activity in the respiratory mucosa over time.
Fig. 2: Module activity driven by first viral infection.
Fig. 3: Gene expression and module activity in relation to RTI susceptibility.
Fig. 4: Early-life nasopharyngeal microbiota development.
Fig. 5: Microbiota development and associations with cumulative module M3 activity.
Fig. 6: HAllA results.

Similar content being viewed by others

Data availability

Both microbiota and gene expression data, including minimal patient metadata, have been deposited at the National Centre for Biotechnology Information GenBank database (accession no. PRJNA740120 and GSE152951, respectively). Full patient metadata are available upon request. Source data are provided for each Figure and Extended Data Figure.

Taxonomic annotations were based on the Silva database (v138.2). For gene set enrichment analyses, gene sets from the Reactome Pathways Database (https://reactome.org/download-data; downloaded 23 March 2020)61 and the Gene Ontology (GO) database (GO.db R package; release 10 July 2019) were used62.

To deconvolute microarray data, we used the ‘single-cell atlas of the airway epithelium’ dataset

(https://www.genomique.eu/cellbrowser/HCA/; hg19 genes annotation; downloaded 17 September 2021). Source data are provided with this paper.

Code availability

Code used to process and analyse the data is available at https://gitlab.com/wsteenhu/MUIS_trx/. A release version of the code has been archived in a Zenodo repository (https://doi.org/10.5281/zenodo.5736115).

References

  1. Levels and Trends in Child Mortality 2019. Estimates developed by the UN Inter-agency Group for Child Mortality Estimation (UNICEF, WHO, World Bank & UN-DESA Population Division, 2019).

  2. Gutiérrez, F. et al. The influence of age and gender on the population-based incidence of community-acquired pneumonia caused by different microbial pathogens. J. Infect. 53, 166–174 (2006).

    Article  PubMed  Google Scholar 

  3. Jensen-fangel, S. et al. Gender differences in hospitalization rates for respiratory tract infections in Danish youth. Scand. J. Infect. Dis. 36, 31–36 (2004).

    Article  PubMed  Google Scholar 

  4. Patarčić, I. et al. The role of host genetic factors in respiratory tract infectious diseases: systematic review, meta-analyses and field synopsis. Sci. Rep. 5, 16119 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Kristensen, K. et al. Caesarean section and hospitalization for respiratory syncytial virus infection: a population-based study. Pediatr. Infect. Dis. J. 34, 145–148 (2015).

    Article  PubMed  Google Scholar 

  6. Moore, H. C., de Klerk, N., Holt, P., Richmond, P. C. & Lehmann, D. Hospitalisation 877 for bronchiolitis in infants is more common after elective caesarean delivery. Arch. Dis. Child 97, 410–414 (2012).

    Article  PubMed  Google Scholar 

  7. Duijts, L., Ramadhani, M. K. & Moll, H. A. Breastfeeding protects against infectious diseases during infancy in industrialized countries. A systematic review. Matern. Child Nutr. 5, 199–210 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Schuez-Havupalo, L., Toivonen, L., Karppinen, S., Kaljonen, A. & Peltola, V. Daycare attendance and respiratory tract infections: a prospective birth cohort study. BMJ Open 7, e014635 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Vanker, A., Gie, R. P. & Zar, H. J. The association between environmental tobacco 886 smoke exposure and childhood respiratory disease: a review. Expert Rev. Respir. Med. 11, 661–673 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bosch, A. A. T. M. et al. Maturation of the infant respiratory microbiota, environmental drivers, and health consequences. A prospective cohort study. Am. J. Respir. Crit. Care Med. 196, 1582–1590 (2017).

    Article  PubMed  Google Scholar 

  11. Man, W. H. et al. Loss of microbial topography between oral and nasopharyngeal microbiota and development of respiratory infections early in life. Am. J. Respir. Crit. Care Med. 200, 760–770 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Man, W. H. et al. Bacterial and viral respiratory tract microbiota and host characteristics in children with lower respiratory tract infections: a matched case-control study. Lancet Resp. Med. 7, 417–426 (2019).

    Article  Google Scholar 

  13. de Steenhuijsen Piters, W. A. A. et al. Nasopharyngeal microbiota, host transcriptome, and disease severity in children with respiratory syncytial virus infection. Am. J. Respir. Crit. Care Med. 194, 1104–1115 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  14. de Steenhuijsen Piters, W. A. A., Binkowska, J. & Bogaert, D. Early life microbiota and respiratory tract infections. Cell Host Microbe 28, 223–232 (2020).

    Article  PubMed  CAS  Google Scholar 

  15. Teo, S. M. et al. The infant nasopharyngeal microbiome impacts severity of lower respiratory infection and risk of asthma development. Cell Host Microbe 17, 704–715 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. van den Bergh, M. R. et al. Associations between pathogens in the upper respiratory tract of young children: interplay between viruses and bacteria. PLoS ONE 7, e47711 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Gollwitzer, E. S. et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat. Med. 20, 642–647 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. 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  PubMed  PubMed Central  Google Scholar 

  19. Torow, N. & Hornef, M. W. The neonatal window of opportunity: setting the stage for life-long host-microbial interaction and immune homeostasis. J. Immunol. 198, 557–563 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Gollwitzer, E. S. & Marsland, B. J. Impact of early-life exposures on immune maturation and susceptibility to disease. Trends Immunol. 36, 684–696 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Hornef, M. W. & Torow, N. ‘Layered immunity’ and the ‘neonatal window of opportunity’ – timed succession of non-redundant phases to establish mucosal host–microbial homeostasis after birth. Immunology 159, 15–25 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Knoop, K. A. et al. Microbial antigen encounter during a preweaning interval is critical for tolerance to gut bacteria. Sci. Immunol. 2, eaao1314 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Constantinides, M. G. et al. MAIT cells are imprinted by the microbiota in early life and promote tissue repair. Science 366, eaax6624 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Scharschmidt, T. C. et al. A wave of regulatory T cells into neonatal skin mediates tolerance to commensal microbes. Immunity 43, 1011–1021 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Davis, N. M., Proctor, D. M., Holmes, S. P., Relman, D. A. & Callahan, B. J. Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. Microbiome 6, 226 (2018).

    PubMed  PubMed Central  Google Scholar 

  27. Newman, A. M. et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 37, 773–782 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. de Steenhuijsen Piters, W. A. A. et al. Interaction between the nasal microbiota and S. pneumoniae in the context of live-attenuated influenza vaccine. Nat. Commun. 10, 2981 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Kollmann, T. R., Levy, O., Montgomery, R. R. & Goriely, S. Innate immune function by toll-like receptors: distinct responses in newborns and the elderly. Immunity 37, 771–783 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rakoff-Nahoum, S. et al. Analysis of gene–environment interactions in postnatal development of the mammalian intestine. Proc. Natl Acad. Sci. USA 112, 1929–1936 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wei, H.-X., Wang, B. & Li, B. IL-10 and IL-22 in mucosal immunity: driving protection and pathology. Front. Immunol. 11, 1315 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ivashkiv, L. B. & Donlin, L. T. Regulation of type I interferon responses. Nat. Rev. Immunol. 14, 36–49 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Trinchieri, G. Type I interferon: friend or foe? J. Exp. Med. 207, 2053–2063 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wilson, R. P. et al. STAT2 dependent Type I interferon response promotes dysbiosis and luminal expansion of the enteric pathogen Salmonella typhimurium. PLoS Pathog. 15, e1007745 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Perkins, D. J. et al. Salmonella typhimurium co-opts the host Type I IFN system to restrict macrophage innate immune transcriptional responses selectively. J. Immunol. 195, 2461–2471 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Sun, K. & Metzger, D. W. Inhibition of pulmonary antibacterial defense by interferon-gamma during recovery from influenza infection. Nat. Med. 14, 558–564 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Tarabichi, Y. et al. The administration of intranasal live attenuated influenza vaccine induces changes in the nasal microbiota and nasal epithelium gene expression profiles. Microbiome 3, 74 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Planet, P. J. et al. Lambda interferon restructures the nasal microbiome and increases susceptibility to Staphylococcus aureus superinfection. mBio 7, e01939-15 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Følsgaard, N. V. et al. Pathogenic bacteria colonizing the airways in asymptomatic neonates stimulates topical inflammatory mediator release. Am. J. Respir. Crit. Care Med. 187, 589–595 (2013).

    Article  PubMed  CAS  Google Scholar 

  40. Teo, S. M. et al. Airway microbiota dynamics uncover a critical window for interplay of pathogenic bacteria and allergy in childhood respiratory disease. Cell Host Microbe 24, 341–352.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Dickson, R. P., Erb-Downward, J. R., Martinez, F. J. & Huffnagle, G. B. The microbiome and the respiratory tract. Annu. Rev. Physiol. 78, 481–504 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Gulraiz, F., Bellinghausen, C., Bruggeman, C. A. & Stassen, F. R. Haemophilus influenzae increases the susceptibility and inflammatory response of airway epithelial cells to viral infections. FASEB J. 29, 849–858 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Kanmani, P. et al. Respiratory commensal bacteria Corynebacterium pseudodiphtheriticum improves resistance of infant mice to respiratory syncytial virus and Streptococcus pneumoniae superinfection. Front. Microbiol. 8, 1613 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Bosch, A. A. T. M. et al. Development of upper respiratory tract microbiota in infancy is affected by mode of delivery. EBioMedicine 9, 336–345 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Haynes:TRIzol RNeasy (OpenWetWare, 2015).

  46. Kauffmann, A. & Huber, W. Microarray data quality control improves the detection of differentially expressed genes. Genomics 95, 138–142 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Irizarry, R. A. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003).

    Article  PubMed  Google Scholar 

  48. Nygaard, V., Rødland, E. A. & Hovig, E. Methods that remove batch effects while retaining group differences may lead to exaggerated confidence in downstream analyses. Biostatistics 17, 29–39 (2016).

    Article  PubMed  Google Scholar 

  49. Caporaso, J. G. et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc. Natl Acad. Sci. USA 108, 4516–4522 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Edgar, R. C. Updating the 97% identity threshold for 16S ribosomal RNA OTUs. Bioinformatics 34, 2371–2375 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Wyllie, A. L. et al. Streptococcus pneumoniae in saliva of Dutch primary school children. PLoS ONE 9, e102045 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Paulson, J. N., Stine, O. C., Bravo, H. C. & Pop, M. Differential abundance analysis for microbial marker-gene surveys. Nat. Methods 10, 1200–1202 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mallick, H. et al. Multivariable association discovery in population-scale meta-omics studies. PLoS Comput. Biol. 17, e1009442 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bakdash, J. Z. & Marusich, L. R. Repeated measures correlation. Front. Psychol. 8, 456 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Jaskowiak, P. A., Campello, R. J. G. B. & Costa, I. G. Proximity measures for clustering gene expression microarray data: a validation methodology and a comparative analysis. IEEE/ACM Trans. Comput. Biol. Bioinf. 10, 845–857 (2013).

    Article  Google Scholar 

  57. de Souto, M. C., Costa, I. G., de Araujo, D. S., Ludermir, T. B. & Schliep, A. Clustering cancer gene expression data: a comparative study. BMC Bioinformatics 9, 497 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Russo, P. S. T. et al. CEMiTool: a bioconductor package for performing comprehensive modular co-expression analyses. BMC Bioinformatics 19, 56 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Oksanen, J. et al. vegan: Community Ecology Package. R package version 2.5-7. https://CRAN.R-project.org/package=vegan (2015).

  60. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Croft, D. et al. Reactome: a database of reactions, pathways and biological processes. Nucleic Acids Res. 39, D691–D697 (2011).

    Article  CAS  PubMed  Google Scholar 

  62. Ashburner, M. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Reimand, J. et al. Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap. Nat. Protoc. 14, 482–517 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Stoney, R. A., Schwartz, J.-M., Robertson, D. L. & Nenadic, G. Using set theory to reduce redundancy in pathway sets. BMC Bioinformatics 19, 386 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Korotkevich, G. et al. Fast gene set enrichment analysis. Preprint at bioRxiv https://doi.org/10.1101/060012 (2016).

  67. Deprez, M. et al. A single-cell atlas of the human healthy airways. Am. J. Respir. Crit. Care Med. 202, 1636–1645 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all volunteers who participated in this study; A. A. T. M. Bosch and all members of the Spaarne Gasthuis Academy research team for their dedication and practical support with participant enrolment and sample collection; and M. Clerc for her support with microarray sample preparation. This work was supported in part by the Netherlands Organisation for Scientific research (NWO-VIDI; grant 91715359) and CSO/NRS through a Scottish Senior Clinical Fellowship award (SCAF/16/03).

Author information

Authors and Affiliations

Authors

Contributions

D.B., M.A.v.H. and E.A.M.S. designed the experiments and wrote the study protocols. D.B., M.A.v.H., P.C.M.d.G. and E.A.M.S. were responsible for (supervision of) participant enrolment, sample and data collection. R.H., K.A. and M.L.J.N.C. were responsible for laboratory processing of samples. W.A.A.d.S.P., D.B. and R.L.W. performed bioinformatic processing and W.A.A.d.S.P., E.M.d.K. and D.B. ran statistical analyses. W.A.A.d.S.P., E.M.d.K. and D.B. wrote the paper. All authors contributed to interpretation of the results, critically revised the manuscript for important intellectual content, and approved the final manuscript.

Corresponding author

Correspondence to Debby Bogaert.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Leopoldo Segal and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Between-dataset correlations and shared host and environmental determinants.

a, Mantel tests quantifying the variance explained (square of Mantel r statistic) between each pair of datasets (percentages/blue shades). The Mantel test is a test of correlation between pairs of dissimilarity matrices derived from the original microbiota, gene expression or viral data. Mantel statistics were calculated between subjects (interindividual) or within subjects over time (intraindividual; see Methods). b, PERMANOVA results showing the association between host and environmental variables and each dataset (that is microbiota, gene expression and viral data). Asterisks denote Benjamini-Hochberg (BH)-corrected statistical significance (correction per dataset; *, q ≤ 0.05; **, q ≤ 0.01; ***, q ≤ 0.001). The variance explained is estimated for each variable independently (percentages/blue shades). ‘All’ refers to a test including all metadata. For each column, the total n for each dataset is shown in square brackets. ‘full’ indicates the maximum samples available for that dataset. ‘matched’ refers to the set of paired samples between microbiota and gene-expression data. For each variable tested, the number of degrees of freedom is given in square brackets. Significance of both Mantel and PERMANOVA-tests was based on 1,000 permutations.

Source data

Extended Data Fig. 2 Viral detection rates of a panel of respiratory viruses.

Viruses could be detected from directly after birth and on.

Source data

Extended Data Fig. 3 Timing of first viral detection and age at which parents first report respiratory symptoms.

Time lines of all subjects showing the temporal relationship between first viral detection (red points) and the time frame within which parents first reported respiratory symptoms (black horizontal lines). At each regular visit, parents were asked whether their child had been suffering from RTI symptoms since the last regular visit. We therefore defined the preceding regular visit as ‘start’ and the current visit as ‘end’ of the time frame within which RTI complaints had first occurred. Other sampling moments for each individual are shown in grey. We stratified the individual time lines by timing of first viral presence vs first report of RTI symptoms, with first viral detection preceding first report of symptoms in n=59 infants, viral detection co-occurring with the end of the first RTI episode in n=26 infants and the first viral infection reported after the first RTI episode in n=25 infants. 4/114 infants are not included in this overview as we did not detect a viral infection and/or parents did not report any RTI complaints.

Source data

Extended Data Fig. 4 Dendrogram visualizing an average linkage hierarchical clustering of samples based on the Bray–Curtis dissimilarity matrix.

The length of the branches of the tree structure corresponds with the similarities between samples (n=1,156). Adjacent to the branch ends information on 1) initial clustering, 2) subclustering of a large cluster characterized by Corynebacterium/Dolosigranulum/Moraxella (n=587 samples) and 3) (supervised) stratification of the resulting CDG5 cluster into Moraxella (2)-enriched (CDG5/MOR2) and -depleted (CDG5) subclusters is depicted. Combined, these steps result in the 11 (final) clusters as shown by colour-coded horizontal panels. Clusters are named after the most discriminative Amplicon Sequence Variants (ASVs) within those clusters. Gray panels indicate samples not grouped into clusters consisting of 10 or more samples. A heatmap shows the relative abundance of the 20 highest-ranked ASVs based on mean relative abundance across all samples. Repeated samples from individuals were included in this clustering analysis to optimize cluster identification and increase comparability across time points.

Source data

Extended Data Fig. 5 Microbiota transitions related to cumulative module M1 activity.

Kaplan-Meier curves depicting cumulative module M1 activity in relation to age at which a given infant first transitioned into a Corynebacterium/Dolosigranulum (CDG5), Corynebacterium (5)/Dolosigranulum/Moraxella (2) (CDG5/MOR2), Haemophilus (HAE) or Moraxella (2) (MOR2) cluster. Cumulative events are shown on the y-axis. P values shown are based on logrank tests. AUC-values were classified as ‘low’ (below median) or ‘high’ (above median) compared to all other subjects with M1 AUC-values over that interval (see Fig. 5b).

Source data

Extended Data Fig. 6 Relationship between presence and abundance of S. pneumoniae (lytA) and members of the Streptococcus genus.

a, Boxplots depicting the relationship between presence/absence of S. pneumoniae based on lytA-qPCR results (CT <40 cycles) and centre logratio (clr) transformed relative abundance of several members of the Streptococcus genus. Only streptococci present in at least 100 samples were shown. P values were based on mixed linear effects models with pneumococcal presence/absence based on lytA-qPCR as predictor and subject as random intercept. These results indicate that pneumococcal presence is strongly associated with Streptococcus (13) abundance. Box plots represent the 25th and 75th percentiles (lower and upper boundaries of boxes, respectively), the median (middle horizontal line), and measurements that fall within 1.5 times the interquartile range (IQR; distance between 25th and 75th percentiles; whiskers). b, Correlation plot showing the relationship between S. pneumoniae abundance based on lytA-qPCR results (CT-values) and centre logratio (clr) transformed relative abundance of Streptococcus (13). P values and repeated measures correlation coefficients (r) are based on the ‘rmcorr’-R package. P values were calculated including all data, as well as only data from samples in which S. pneumoniae and Streptococcus (13) were detected (presence defined as CT <40 cycles and clr transformed relative abundance> 4, respectively; dotted lines). The shaded area surrounding the correlation line represents the 95% confidence interval.

Source data

Extended Data Fig. 7 Relative abundance Z-score of microbiota members over time.

Z-scores were calculated by subtracting the mean and subdividing by the standard deviation across all samples for each given Amplicon Sequence Variant (ASV). Similar maturation patterns of Corynebacterium (5)/Dolosigranulum pigrum (7), and Moraxella (2)/Haemophilus (12)/Streptococcus (13) were observed, suggesting that differences in genes associated with these ASVs are not explained by a residual effect of age.

Source data

Extended Data Fig. 8 HAllA-associated microbiota in relation to RTI susceptibility.

Association between Corynebacterium (5), Dolosigranulum pigrum (7), Moraxella (2), Haemophilus (12) and Streptococcus (13) abundance and the number of mild RTIs over the first year of life. Colored bars indicate the time window within which a significant difference between groups (3–4 and 5–7 vs 0–2 RTIs) was detected. Bar height correlates with effect size (‘Area’). Values depicted in/on top of bars are q-values. Associations with q-value ≤0.1 are depicted (see Supplementary Table 8).

Source data

Extended Data Fig. 9 Flowchart.

Overview of all statistical analyses used. Both analyses and the figures/tables where results of these analyses can be found are shown. Links between analyses/nested analyses are depicted using arrows. Asterisks denote those analyses that were data-driven. ORA, overrepresentation analysis.

Extended Data Fig. 10 Sequencing depth and rarefaction curves.

a, Raincloud plot indicating the distribution of the number of reads per sample. Only samples with a read count of ≥3,000 reads (after decontamination/before filtering rare taxa) were included. The distribution of read counts was approximately log-normal (n=1,156 samples; median 23,938 reads, range 3,184–190,874 reads). b, Rarefaction curves for samples with <15,000 reads (n=175 samples). For this subset of samples with lower numbers of reads, we find that rarefaction curves generally saturate ~3,000 reads (dotted line), indicating samples were sequenced sufficiently deep to capture the microbial diversity.

Source data

Supplementary information

Reporting Summary

Peer Review File

41564_2021_1043_MOESM3_ESM.xlsx

Supplementary Tables 1–8 Supplementary Tables 18.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Steenhuijsen Piters, W.A.A., Watson, R.L., de Koff, E.M. et al. Early-life viral infections are associated with disadvantageous immune and microbiota profiles and recurrent respiratory infections. Nat Microbiol 7, 224–237 (2022). https://doi.org/10.1038/s41564-021-01043-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-021-01043-2

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