A cellular census of human lungs identifies novel cell states in health and in asthma (original) (raw)

Data availability

Data requests for raw and analyzed data and materials will fall under two categories. Datasets from healthy live volunteers and live volunteers with asthma will be promptly reviewed by the University of Groningen. Any data and materials that can be shared will be released via a Material Transfer Agreement. These datasets can be found on European Genome–phenome Archive (https://www.ebi.ac.uk/ega/home) EGAS00001001755. Datasets generated from lung resection samples using Drop-seq can be accessed in GSE130148. Datasets generated from deceased donors fall under Open Access Policies of the Human Cell Atlas (https://www.humancellatlas.org for details). This data can be accessed at European Genome–phenome Archive (https://www.ebi.ac.uk/ega/home) EGAS00001002649. Interactive exploration tool: www.lungcellatlas.org.

References

  1. Tata, P. R. & Rajagopal, J. Plasticity in the lung: making and breaking cell identity. Development 144, 755–766 (2017).
    Article CAS PubMed PubMed Central Google Scholar
  2. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).
    Article CAS PubMed PubMed Central Google Scholar
  3. Montoro, D. T. et al. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 560, 319–324 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  4. Plasschaert, L. W. et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 560, 377–381 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  5. Bisset, L. R. & Schmid-Grendelmeier, P. Chemokines and their receptors in the pathogenesis of allergic asthma: progress and perspective. Curr. Opin. Pulm. Med. 11, 35–42 (2005).
    Article CAS PubMed Google Scholar
  6. Colvin, R. A. et al. Synaptotagmin-mediated vesicle fusion regulates cell migration. Nat. Immunol. 11, 495–502 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  7. Urawa, M. et al. Protein S is protective in pulmonary fibrosis. J. Thromb. Haemost. 14, 1588–1599 (2016).
    Article CAS PubMed Google Scholar
  8. Wujak, A. et al. FXYD1 negatively regulates Na+/K+-ATPase activity in lung alveolar epithelial cells. Respir. Physiol. Neurobiol. 220, 54–61 (2016).
    Article CAS PubMed Google Scholar
  9. Krotova, K. et al. Alpha-1 antitrypsin-deficient macrophages have increased matriptase-mediated proteolytic activity. Am. J. Respir. Cell Mol. Biol. 57, 238–247 (2017).
    Article CAS PubMed PubMed Central Google Scholar
  10. Vogl, T. et al. S100A12 is expressed exclusively by granulocytes and acts independently from MRP8 and MRP14. J. Biol. Chem. 274, 25291–25296 (1999).
    Article CAS PubMed Google Scholar
  11. Mitchell, A. et al. LILRA5 is expressed by synovial tissue macrophages in rheumatoid arthritis, selectively induces pro-inflammatory cytokines and IL-10 and is regulated by TNF-α, IL-10 and IFN-γ. Eur. J. Immunol. 38, 3459–3473 (2008).
    Article CAS PubMed Google Scholar
  12. Condon, T. V., Sawyer, R. T., Fenton, M. J. & Riches, D. W. H. Lung dendritic cells at the innate-adaptive immune interface. J. Leukoc. Biol. 90, 883–895 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  13. Baumann, U., Routes, J. M., Soler-Palacín, P. & Jolles, S. The lung in primary immunodeficiencies: new concepts in infection and inflammation. Front. Immunol. 9, 1837 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  14. Holgate, S. T. et al. Asthma. Nat. Rev. Dis. Primers 1, 15025 (2015).
    Article PubMed PubMed Central Google Scholar
  15. Lopez-Guisa, J. M. et al. Airway epithelial cells from asthmatic children differentially express proremodeling factors. J. Allergy Clin. Immunol. 129, 990–997.e6 (2012).
    Article CAS PubMed PubMed Central Google Scholar
  16. Alcala, S. E. et al. Mitotic asynchrony induces transforming growth factor-β1 secretion from airway epithelium. Am. J. Respir. Cell Mol. Biol. 51, 363–369 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  17. Harkness, L. M., Ashton, A. W. & Burgess, J. K. Asthma is not only an airway disease, but also a vascular disease. Pharmacol. Ther. 148, 17–33 (2015).
    Article CAS PubMed Google Scholar
  18. Balzar, S. et al. Mast cell phenotype, location, and activation in severe asthma. Data from the Severe Asthma Research Program. Am. J. Respir. Crit. Care Med. 183, 299–309 (2011).
    Article PubMed Google Scholar
  19. Truyen, E. et al. Evaluation of airway inflammation by quantitative Th1/Th2 cytokine mRNA measurement in sputum of asthma patients. Thorax 61, 202–208 (2006).
    Article CAS PubMed PubMed Central Google Scholar
  20. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).
    CAS PubMed PubMed Central Google Scholar
  21. Erle, D. J. & Sheppard, D. The cell biology of asthma. J. Cell Biol. 205, 621–631 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  22. Danahay, H. et al. Notch2 is required for inflammatory cytokine-driven goblet cell metaplasia in the lung. Cell Rep. 10, 239–252 (2015).
    Article CAS PubMed Google Scholar
  23. Gomi, K., Arbelaez, V., Crystal, R. G. & Walters, M. S. Activation of NOTCH1 or NOTCH3 signaling skews human airway basal cell differentiation toward a secretory pathway. PLoS ONE 10, e0116507 (2015).
    Article CAS PubMed PubMed Central Google Scholar
  24. Ordovas-Montanes, J. et al. Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature 560, 649–654 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  25. Luo, W. et al. Airway epithelial expression quantitative trait loci reveal genes underlying asthma and other airway diseases. Am. J. Respir. Cell Mol. Biol. 54, 177–187 (2016).
    Article CAS PubMed PubMed Central Google Scholar
  26. Wu, C. A. et al. Bronchial epithelial cells produce IL-5: implications for local immune responses in the airways. Cell. Immunol. 264, 32–41 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  27. Laitinen, L. A., Laitinen, A. & Haahtela, T. Airway mucosal inflammation even in patients with newly diagnosed asthma. Am. Rev. Respir. Dis. 147, 697–704 (1993).
    Article CAS PubMed Google Scholar
  28. Arima, M. & Fukuda, T. Prostaglandin D2 and TH2 inflammation in the pathogenesis of bronchial asthma. Korean J. Intern. Med. 26, 8–18 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  29. Xue, L. et al. Prostaglandin D2 activates group 2 innate lymphoid cells through chemoattractant receptor-homologous molecule expressed on TH2 cells. J. Allergy Clin. Immunol. 133, 1184–1194 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  30. Dougherty, R. H. et al. Accumulation of intraepithelial mast cells with a unique protease phenotype in TH2-high asthma. J. Allergy Clin. Immunol. 125, 1046–1053.e8 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  31. Hol, B. E., van de Graaf, E. A., Out, T. A., Hische, E. A. & Jansen, H. M. IgM in the airways of asthma patients. Int. Arch. Allergy Appl. Immunol. 96, 12–18 (1991).
    Article CAS PubMed Google Scholar
  32. Muehling, L. M., Lawrence, M. G. & Woodfolk, J. A. Pathogenic CD4+ T cells in patients with asthma. J. Allergy Clin. Immunol. 140, 1523–1540 (2017).
    Article CAS PubMed PubMed Central Google Scholar
  33. Oja, A. E. et al. Trigger-happy resident memory CD4+ T cells inhabit the human lungs. Mucosal Immunol. 11, 654–667 (2018).
    Article CAS PubMed Google Scholar
  34. Mitson-Salazar, A. et al. Hematopoietic prostaglandin D synthase defines a proeosinophilic pathogenic effector human TH2 cell subpopulation with enhanced function. J. Allergy Clin. Immunol. 137, 907–918.e9 (2016).
    Article CAS PubMed Google Scholar
  35. Wambre, E. et al. A phenotypically and functionally distinct human T H2 cell subpopulation is associated with allergic disorders. Sci. Transl. Med. 9, eaam9171 (2017).
    Article CAS PubMed PubMed Central Google Scholar
  36. Lam, E. P. S. et al. IL-25/IL-33-responsive TH2 cells characterize nasal polyps with a default TH17 signature in nasal mucosa. J. Allergy Clin. Immunol. 137, 1514–1524 (2016).
    Article CAS PubMed PubMed Central Google Scholar
  37. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal–fetal interface in humans. Nature 563, 347–353 (2018).
    Article CAS PubMed Google Scholar
  38. Weckmann, M., Kopp, M. V., Heinzmann, A. & Mattes, J. Haplotypes covering the TNFSF10 gene are associated with bronchial asthma. Pediatr. Allergy Immunol. 22, 25–30 (2011).
    Article PubMed Google Scholar
  39. Harada, M. et al. Thymic stromal lymphopoietin gene promoter polymorphisms are associated with susceptibility to bronchial asthma. Am. J. Respir. Cell Mol. Biol. 44, 787–793 (2011).
    Article CAS PubMed Google Scholar
  40. Grotenboer, N. S., Ketelaar, M. E., Koppelman, G. H. & Nawijn, M. C. Decoding asthma: translating genetic variation in IL33 and IL1RL1 into disease pathophysiology. J. Allergy Clin. Immunol. 131, 856–865 (2013).
    Article CAS PubMed Google Scholar
  41. Holgate, S. T. et al. Epithelial-mesenchymal communication in the pathogenesis of chronic asthma. Proc. Am. Thorac. Soc. 1, 93–98 (2004).
    Article CAS PubMed Google Scholar
  42. Heijink, I. H. et al. Down-regulation of E-cadherin in human bronchial epithelial cells leads to epidermal growth factor receptor-dependent Th2 cell-promoting activity. J. Immunol. 178, 7678–7685 (2007).
    Article CAS PubMed Google Scholar
  43. Song, J. et al. Aberrant DNA methylation and expression of SPDEF and FOXA2 in airway epithelium of patients with COPD. Clin. Epigenetics 9, 42 (2017).
    Article CAS PubMed PubMed Central Google Scholar
  44. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).
    Article CAS PubMed Google Scholar
  45. Wu, T. D. & Nacu, S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26, 873–881 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  46. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
    Article CAS PubMed Google Scholar
  47. van den Brink, S. C. et al. Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat. Methods 14, 935–936 (2017).
    Article CAS PubMed Google Scholar
  48. Young, M. D. & Behjati, S. SoupX removes ambient RNA contamination from droplet based single cell RNA sequencing data. Preprint at https://doi.org/10.1101/303727 (2018).
  49. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, P281–291.E9 (2019).
    Article CAS Google Scholar
  50. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).
    Article CAS PubMed PubMed Central Google Scholar
  51. Mereu, E. et al. matchSCore: matching single-cell phenotypes across tools and experiments. Preprint at https://doi.org/10.1101/314831 (2018).

Download references

Acknowledgements

We thank J. Eliasova (scientific illustrator) for support with design of figures, the Sanger Single Cell Genomics Core Facility for support with the SmartSeq2 protocol, E. Rawlins for feedback and critical reading of the manuscript, L. Mamanova for the technical support, as well as all the members of the Teichmann lab for scientific input. We are grateful to the Cambridge Biorepository for Translational Medicine (CBTM) for the provision of tissue from deceased organ donors, to all tissue donors, and to K. Sjollema and the UMCG Imaging and Microscopy Center. We gratefully acknowledge the provision of human biomaterial and clinical data from the CPC-M bioArchive and its partners at the Asklepios Biobank Gauting, the Klinikum der Universität München, and the Ludwig-Maximilians-Universität München. This project was funded by and part of the Open Targets collaboration (https://www.opentargets.org), a GlaxoSmithKline collaborative agreement with University Medical Center Groningen, Wellcome (WT206194), the Lung Foundation Netherlands (projects no. 5.1.14.020 and 4.1.18.226), Health-Holland, Top Sector Life Sciences and Health, and Human Cell Atlas Wellcome Stragetic Science Support (211276/Z/18/Z). R.V.-T. was supported by EMBO and HFSP Long Term fellowships. T.G. by the Marie Curie ENLIGHT-TEN training network. L.M.S. acknowledges funding from the European Union’s Horizon 2020 research and innovation program under the Marie Sklodowska-Curie grant agreement no. 753039. H.B.S. acknowledges funding by the Helmholtz Association and the German Center for Lung Research (DZL). F.J.T. acknowledges financial support by the German Research Foundation (DFG) within the Collaborative Research Centre 1243. F.J.T acknowledges subproject A17, by the Helmholtz Association (Incubator grant sparse2big, grant number ZT-I-0007) and by the Chan Zuckerberg Initiative D.A.F. (advised fund of Silicon Valley Community Foundation), grant number 182835. A.C. and P.M.S. acknowledge European Research Council (project 677501 – ZF_Blood). This publication is part of the Human Cell Atlas (www.humancellatlas.org/publications).

Author information

Author notes

  1. These authors contributed equally: F. A. Vieira Braga, G. Kar, M. Berg, O. A. Carpaij, K. Polanski.
  2. These authors jointly supervised this work: M. C. Nawijn, S. A. Teichmann.

Authors and Affiliations

  1. Wellcome Sanger Institute, Cambridge, UK
    Felipe A. Vieira Braga, Gozde Kar, Krzysztof Polanski, Tomás Gomes, Eirini S. Fasouli, Mirjana Efremova, Roser Vento-Tormo, Carlos Talavera-López, Paulina M. Strzelecka, Helen V. Firth, Ana Cvejic, Kerstin B. Meyer & Sarah A. Teichmann
  2. Open Targets, Cambridge, UK
    Felipe A. Vieira Braga, Gozde Kar, Eirini S. Fasouli & Sarah A. Teichmann
  3. Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
    Marijn Berg, Sharon Brouwer, Laura Hesse, Jian Jiang, Marnix R. Jonker, Marjan Luinge, Corry-Anke Brandsma, Wim Timens & Martijn C. Nawijn
  4. Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
    Marijn Berg, Orestes A. Carpaij, Sharon Brouwer, Laura Hesse, Jian Jiang, Marnix R. Jonker, Marjan Luinge, Corry-Anke Brandsma, Wim Timens, Gerard H. Koppelman, Maarten van den Berge & Martijn C. Nawijn
  5. Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
    Orestes A. Carpaij & Maarten van den Berge
  6. Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
    Lukas M. Simon, Subarna Palit & Fabian J. Theis
  7. Allergic Inflammation Discovery Performance Unit, Respiratory Therapy Area, GlaxoSmithKline, Stevenage, UK
    Karen Affleck & Antoon J. van Oosterhout
  8. TranslaTUM, Technische Universität München, Munich, Germany
    Subarna Palit
  9. Institute of Virology, Technische Universität München, Munich, Germany
    Subarna Palit
  10. German Center for Infection Research, Partner Site Munich, Munich, Germany
    Subarna Palit
  11. Department of Haematology, University of Cambridge, Cambridge, UK
    Paulina M. Strzelecka & Ana Cvejic
  12. Cambridge Stem Cell Institute, Cambridge, UK
    Paulina M. Strzelecka & Ana Cvejic
  13. Department of Surgery, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
    Krishnaa T. Mahbubani & Kourosh Saeb-Parsy
  14. Helmholtz Zentrum München, Institute of Lung Biology and Disease, Member of the German Center for Lung Research (DZL), Munich, Germany
    Ilias Angelidis, Maximilian Strunz & Herbert B. Schiller
  15. Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children’s Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
    Gerard H. Koppelman
  16. Department of Mathematics, Technische Universität München, Garching, Germany
    Fabian J. Theis
  17. Theory of Condensed Matter Group, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
    Sarah A. Teichmann

Authors

  1. Felipe A. Vieira Braga
    You can also search for this author inPubMed Google Scholar
  2. Gozde Kar
    You can also search for this author inPubMed Google Scholar
  3. Marijn Berg
    You can also search for this author inPubMed Google Scholar
  4. Orestes A. Carpaij
    You can also search for this author inPubMed Google Scholar
  5. Krzysztof Polanski
    You can also search for this author inPubMed Google Scholar
  6. Lukas M. Simon
    You can also search for this author inPubMed Google Scholar
  7. Sharon Brouwer
    You can also search for this author inPubMed Google Scholar
  8. Tomás Gomes
    You can also search for this author inPubMed Google Scholar
  9. Laura Hesse
    You can also search for this author inPubMed Google Scholar
  10. Jian Jiang
    You can also search for this author inPubMed Google Scholar
  11. Eirini S. Fasouli
    You can also search for this author inPubMed Google Scholar
  12. Mirjana Efremova
    You can also search for this author inPubMed Google Scholar
  13. Roser Vento-Tormo
    You can also search for this author inPubMed Google Scholar
  14. Carlos Talavera-López
    You can also search for this author inPubMed Google Scholar
  15. Marnix R. Jonker
    You can also search for this author inPubMed Google Scholar
  16. Karen Affleck
    You can also search for this author inPubMed Google Scholar
  17. Subarna Palit
    You can also search for this author inPubMed Google Scholar
  18. Paulina M. Strzelecka
    You can also search for this author inPubMed Google Scholar
  19. Helen V. Firth
    You can also search for this author inPubMed Google Scholar
  20. Krishnaa T. Mahbubani
    You can also search for this author inPubMed Google Scholar
  21. Ana Cvejic
    You can also search for this author inPubMed Google Scholar
  22. Kerstin B. Meyer
    You can also search for this author inPubMed Google Scholar
  23. Kourosh Saeb-Parsy
    You can also search for this author inPubMed Google Scholar
  24. Marjan Luinge
    You can also search for this author inPubMed Google Scholar
  25. Corry-Anke Brandsma
    You can also search for this author inPubMed Google Scholar
  26. Wim Timens
    You can also search for this author inPubMed Google Scholar
  27. Ilias Angelidis
    You can also search for this author inPubMed Google Scholar
  28. Maximilian Strunz
    You can also search for this author inPubMed Google Scholar
  29. Gerard H. Koppelman
    You can also search for this author inPubMed Google Scholar
  30. Antoon J. van Oosterhout
    You can also search for this author inPubMed Google Scholar
  31. Herbert B. Schiller
    You can also search for this author inPubMed Google Scholar
  32. Fabian J. Theis
    You can also search for this author inPubMed Google Scholar
  33. Maarten van den Berge
    You can also search for this author inPubMed Google Scholar
  34. Martijn C. Nawijn
    You can also search for this author inPubMed Google Scholar
  35. Sarah A. Teichmann
    You can also search for this author inPubMed Google Scholar

Contributions

S.A.T., M.C.N., M.v.d.B., K.A., A.J.v.O. and H.B.S. designed the project. F.A.V.B., M.C.N. and S.A.T. wrote the paper. F.A.V.B., O.A.C., S.B., L.H., J.J., E.S.F., P.M.S., K.T.M., I.A., M.R.J. and M.S. generated the data. F.A.V.B., G.K., M.B., L.M.S., T.G., J.J., M.E., S.P., K.P., H.V.F. and F.J.T. analyzed the data. F.A.V.B., G.K., O.A.C., L.M.S., T.G., J.J., R.V.-T., K.A., S.P., A.C., K.S.-P., W.T., G.H.K., A.J.v.O., C.T.-L., H.B.S., M.v.d.B, F.J.T., M.B. and K.B.M. interpreted the data. All authors read the manuscript, offered feedback, and approved it before submission.

Corresponding authors

Correspondence toMartijn C. Nawijn or Sarah A. Teichmann.

Ethics declarations

Competing interests

K.A. and A.J.v.O. are employees of GlaxoSmithKline. M.B., O.A.C., S.B., M.v.d.B. and M.C.N. received project funding from GlaxoSmithKline.

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 Unbiased clustering of upper airway, lower airway, and parenchymal lung tissue cells and strategy to divide the dataset used in Figs. 1 and 2.

a, t-SNE plot depicting unbiased cluster assignment of combined dataset of parenchyma and upper and lower airways, highlighting EPCAM-high cell clusters (epithelial cells) in blue and other cell types in green. b, t-SNE as in a, highlighting expression of cell lineage markers. c, t-SNE plot of only epithelial cells, as defined in a. d, t-SNE as in c, showing expression of individual genes used for cell-type assignment of different epithelial subsets. e, Table depicting cell-type assignment of clusters identified in c. Alveolar type 1 cells were not identified in an individual cluster in this analysis and they were selected by the high expression of the type 1 marker AGER. Panels ae, n = 12 individuals. Complete sample distribution in Fig. 1b.

Extended Data Fig. 2 Resection lung material analysis via Drop-seq.

a, t-SNE showing single cells obtained from lung resection samples analyzed using Drop-seq. b, Heat map depicting top differentially expressed genes by log fold change among the clusters present in a. c, matchSCore analysis comparing the clusters present in the Drop-seq analysis of lung resection material to the clusters identified in Figs. 1 and 2. Panels a and b, n = 4 individuals. Panel c, n = 16 individuals.

Extended Data Fig. 3 Ionocyte cell identity and molecular profile.

a, Immunohistochemistry staining for FOXI1, using hematoxylin as counterstain. b, Fluorescent staining for ionocyte markers: mouse anti-FOXI1 and rabbit anti-CFTR. cf, Fluorescent staining for FOXI1 (ionocyte-specific) and other epithelial markers: neuroendocrine cell marker (rabbit anti-synaptophysin) (c), ciliated cell marker (mouse anti-α-tubulin) (d), basal cell marker (mouse anti-α-KRT5) (e), and secretory cell marker (mouse anti-MUC5AC) (f). DAPI (blue) stains nuclei. Representative picture out of ten individuals analyzed.

Extended Data Fig. 4 Marker gene analysis of specific goblet, ciliated, and neuroendocrine cells in human lungs.

a, Heat map depicting the expression of marker genes identified by differential expression analysis comparing goblet 1 versus goblet 2 cell types. b, Heat map depicting the expression of marker genes identified by differential expression analysis. Ciliated 1 genes and shared nasal signature genes generated by comparing ciliated 1 versus ciliated 2. Ciliated 2 genes generated by comparing ciliated 2 versus the combination of ciliated 1, goblet 1, and goblet 2, to subtract the nasal signature. c, t-SNE plot depicting the expression of the neuroendocrine marker CHGA. d, Heat map depicting the expression of neuroendocrine markers in neuroendocrine cells as well as in the other clusters identified in our analysis. Neuroendocrine cells identified based on CHGA expression. Neuroendocrine markers obtained from ref. 24. Panels ad, n = 12 individuals. Complete sample distribution in Fig. 1b.

Extended Data Fig. 5 Cell-type assignment strategy for the assignment of non-epithelial cells discussed in Fig. 2.

a, t-SNE colored by unbiased clustering of the non-epithelial dataset described in Fig. 2. b, t-SNE depicting single cells colored by their respective sample origin. c, t-SNE as in a, showing the expression of lineage markers used for cell-type assignment. d, Table with the strategy used for cluster cell assignment of the cell types present in Fig. 2. e, Cluster distribution in the two donors from which we collected paired nasal brushes, airway brushes, and airway biopsies. f, Violin plots depicting the expression of immunoglobulin genes in the B cell cluster divided by tissue of origin. Representative genes of IgG, IgA, IgE, and IgM producing B cells depicted. Panels af, n = 12 individuals. Complete sample distribution in Fig. 2a.

Extended Data Fig. 6 Unbiased clustering of lower airway biopsy samples of healthy volunteers and volunteers with asthma and the strategy used to divide the dataset depicted in Fig. 3.

a, t-SNE plot depicting unbiased cluster assignment of combined dataset of healthy control and asthma lower airway biopsies, highlighting EPCAM-high cell clusters (epithelial cells) in blue and other cell types in green. b, t-SNE as in a, highlighting expression of cell lineage markers. c, t-SNE plot of only epithelial cells, as defined in a. d, t-SNE as in c, showing expression of individual genes used for cell-type assignment of different epithelial subsets. e, Table depicting cell-type assignment of clusters identified in c. f, Fluorescence in situ hybridization image of lung airway biopsy of one asthma patient. Green marks FOXJ1 (ciliated marker), red marks MUC5AC (secretory marker), and blue marks DAPI (nuclei). White arrow points to areas in which both MUC5AC and FOXJ1 are co-expressed in the exact same region (yellow color) or are contained in the vicinities of one nucleus, suggesting co-expression of the transcripts in the same cell. n = 6 healthy volunteers and 6 volunteers with asthma. Full sample distribution in Fig. 3b. Panels ae, n = 12 individuals. Panel f depicts the analysis performed in n = 1 asthma donor.

Extended Data Fig. 7 IL-4/IL-13- and Notch-driven gene transcription signatures in goblet cell metaplasia in asthma.

a, Violin plot depicting IL-4/IL-13 genes signature expression in each cluster of biopsy epithelial cells (signature obtained from ref. 24). b, Violin plot depicting the expression of the IL-4/IL-13 signature (as in a) in healthy control and asthma cells of selected clusters. c, Violin plot depicting Notch signature genes (HES4, HES5, HEY1, HEY2, HEYL, and NRARP) expression in each cluster of biopsy epithelial cells. d, Violin plot depicting the expression of Notch signature genes (as in c) in healthy control and asthma cells of selected clusters. eg, Expression of secretory specific (HES4 and SPDEF) and ciliated specific (FOXJ1) transcription factors in club (e), goblet (f), ciliated (g), and mucous ciliated (h) cells. Panels ah, n = 12 individuals. Complete sample distribution in Fig. 3b.

Extended Data Fig. 8 Clustering and cell-type assignment of non-epithelial cells in the airways of healthy control patients and patients with asthma and their expression of prostaglandin enzymes.

a, t-SNE depicting unbiased clustering of the non-epithelial dataset described in Fig. 4. b, t-SNE as in a, showing the expression of lineage markers used for cell-type assignment. c, Table with the strategy used for cluster cell assignment of the cell types present in Fig. 4. d, Cartoon illustrating the eicosanoids pathway, its receptors and in which cells each gene is expressed in the lungs, with special notation for asthma alterations. Panels ac, n = 12 individuals. Complete sample distribution in Fig. 4a. Panel d was generated based on n = 15 individuals. Complete sample distribution in Figs. 3b, 4a, and 5b.

Extended Data Fig. 9 Comparative analysis of bulk transcriptomes versus single-cell RNA-seq data on matched airway biopsies.

a, UMAP displaying bulk transcriptomes of biopsies before digestion (blue), of the single-cell suspension obtained from digestion and used to load the 10x microfluidics device (orange) and pseudo-bulk samples generated by collapsing all the data obtained from single-cell transcriptomes (green). b, Heat map depicting expression of several epithelial and non-epithelial cell markers, as shown in Extended Data Figs. 6 and 8. Gene markers of rare cells (neuroendocrine, eosinophils, and tuft) not identified in our single-cell clusters depicted in specific colors. Panels ac, n = 8 individuals.

Extended Data Fig. 10 Canonical T cell markers expression in TH CD4 clusters.

a, Heat map depicting genes differentially expressed in the nine clusters of CD4 T cells using an initial unbiased clustering method, followed by manual selection of TH1, TH2, and TH17 clusters based on canonical cytokines. af, Violin plots depicting canonical markers of tissue-resident memory cells (b) and TH1 (c), TH2 (d), TH17 (e), and Treg (f) CD4 T cells. Depicted markers based on literature search of well-established T cell subsets. Panels af, n = 15 individuals. Complete sample distribution in Fig. 5b.

Supplementary information

Source data

Rights and permissions

About this article

Cite this article

Vieira Braga, F.A., Kar, G., Berg, M. et al. A cellular census of human lungs identifies novel cell states in health and in asthma.Nat Med 25, 1153–1163 (2019). https://doi.org/10.1038/s41591-019-0468-5

Download citation