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:

Transcriptional analysis of cystic fibrosis airways at single-cell resolution reveals altered epithelial cell states and composition

Abstract

Cystic fibrosis (CF) is a lethal autosomal recessive disorder that afflicts more than 70,000 people. People with CF experience multi-organ dysfunction resulting from aberrant electrolyte transport across polarized epithelia due to mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CF-related lung disease is by far the most important determinant of morbidity and mortality. Here we report results from a multi-institute consortium in which single-cell transcriptomics were applied to define disease-related changes by comparing the proximal airway of CF donors (n = 19) undergoing transplantation for end-stage lung disease with that of previously healthy lung donors (n = 19). Disease-dependent differences observed include an overabundance of epithelial cells transitioning to specialized ciliated and secretory cell subsets coupled with an unexpected decrease in cycling basal cells. Our study yields a molecular atlas of the proximal airway epithelium that will provide insights for the development of new targeted therapies for CF airway disease.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Single-cell transcriptome atlas of the epithelium lining proximal airways of control donors and donors with end-stage CF lung disease.
Fig. 2: Expansion of secretory function, including mucus secretion and anti-microbial activity, in CF secretory cells.
Fig. 3: Cilia-related gene expression is vastly expanded outside of the main cilia subgroups in CF.
Fig. 4: Depletion of metabolic stability, basal epithelial function and cellular division is widespread in CF lung basal cells.

Similar content being viewed by others

Data availability

Sequence data that support the findings of this study have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus ‘GenBank’ with accession code GSE150674.

References

  1. Carraro, G. et al. Single-cell reconstruction of human basal cell diversity in normal and idiopathic pulmonary fibrosis lungs. Am. J. Respir. Crit. Care Med. 202, 1540–1550 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  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. Pan, S., Iannotti, M. J. & Sifers, R. N. Analysis of serpin secretion, misfolding, and surveillance in the endoplasmic reticulum. Methods Enzymol. 499, 1–16 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Rokicki, W., Rokicki, M., Wojtacha, J. & Dzeljijli, A. The role and importance of club cells (Clara cells) in the pathogenesis of some respiratory diseases. Kardiochir. Torakochirurgia Pol. 13, 26–30 (2016).

    PubMed  PubMed Central  Google Scholar 

  6. Chen, G. et al. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J. Clin. Invest. 119, 2914–2924 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Widdicombe, J. H. & Wine, J. J. Airway gland structure and function. Physiol. Rev. 95, 1241–1319 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Yu, X., Ng, C. P., Habacher, H. & Roy, S. Foxj1 transcription factors are master regulators of the motile ciliogenic program. Nat. Genet. 40, 1445–1453 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Horani, A. et al. Establishment of the early cilia preassembly protein complex during motile ciliogenesis. Proc. Natl Acad. Sci. USA 115, E1221–E1228 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ather, J. L. et al. Serum amyloid A activates the NLRP3 inflammasome and promotes Th17 allergic asthma in mice. J. Immunol. 187, 64–73 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Rock, J. R., Randell, S. H. & Hogan, B. L. Airway basal stem cells: a perspective on their roles in epithelial homeostasis and remodeling. Dis. Model Mech. 3, 545–556 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Okuda, K. et al. Secretory cells dominate airway CFTR expression and function in human airway superficial epithelia. Am. J. Respir. Crit. Care Med. https://doi.org/10.1164/rccm.202008-3198OC (2020).

  13. Xia, C., Braunstein, Z., Toomey, A. C., Zhong, J. & Rao, X. S100 proteins as an important regulator of macrophage inflammation. Front. Immunol. 8, 1908 (2017).

    Article  PubMed  CAS  Google Scholar 

  14. Akram, K. M. et al. An innate defense peptide BPIFA1/SPLUNC1 restricts influenza A virus infection. Mucosal Immunol. 11, 1008 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Thomas, J. et al. Transcriptional control of genes involved in ciliogenesis: a first step in making cilia. Biol. Cell 102, 499–513 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Mihalj, M. et al. Differential expression of TFF1 and TFF3 in patients suffering from chronic rhinosinusitis with nasal polyposis. Int. J. Mol. Sci. 20, 5461 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  17. Eckmann, L. Defence molecules in intestinal innate immunity against bacterial infections. Curr. Opin. Gastroenterol. 21, 147–151 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Bals, R., Weiner, D. J. & Wilson, J. M. The innate immune system in cystic fibrosis lung disease. J. Clin. Invest. 103, 303–307 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tang, A. C. et al. Endoplasmic reticulum stress and chemokine production in cystic fibrosis airway cells: regulation by STAT3 modulation. J. Infect. Dis. 215, 293–302 (2017).

    CAS  PubMed  Google Scholar 

  20. Petraki, C. D., Papanastasiou, P. A., Karavana, V. N. & Diamandis, E. P. Cellular distribution of human tissue kallikreins: immunohistochemical localization. Biol. Chem. 387, 653–663 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Brooks, E. R. & Wallingford, J. B. Multiciliated cells. Curr. Biol. 24, R973–982 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hoh, R. A., Stowe, T. R., Turk, E. & Stearns, T. Transcriptional program of ciliated epithelial cells reveals new cilium and centrosome components and links to human disease. PLoS ONE 7, e52166 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bonser, L. R. et al. The endoplasmic reticulum resident protein AGR3. required for regulation of ciliary beat frequency in the airway. Am. J. Respir. Cell Mol. Biol. 53, 536–543 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Didon, L. et al. RFX3 modulation of FOXJ1 regulation of cilia genes in the human airway epithelium. Respir. Res 14, 70 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Goldfarbmuren, K. C. et al. Dissecting the cellular specificity of smoking effects and reconstructing lineages in the human airway epithelium. Nat. Commun. 11, 2485 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wells, J. M. & Watt, F. M. Diverse mechanisms for endogenous regeneration and repair in mammalian organs. Nature 557, 322–328 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Teixeira, V. H. et al. Stochastic homeostasis in human airway epithelium is achieved by neutral competition of basal cell progenitors. eLife 2, e00966 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Tezuka, T., Takahashi, M. & Katsunuma, N. Cystatin alpha is one of the component proteins of keratohyalin granules. J. Dermatol 19, 756–760 (1992).

    Article  CAS  PubMed  Google Scholar 

  29. O’Shaughnessy, R. F. et al. AKT-dependent HspB1 (Hsp27) activity in epidermal differentiation. J. Biol. Chem. 282, 17297–17305 (2007).

    Article  PubMed  Google Scholar 

  30. Randell, S. H., Walstad, L., Schwab, U. E., Grubb, B. R. & Yankaskas, J. R. Isolation and culture of airway epithelial cells from chronically infected human lungs. Vitr. Cell Dev. Biol. Anim. 37, 480–489 (2001).

    Article  CAS  Google Scholar 

  31. Voynow, J. A., Fischer, B. M., Roberts, B. C. & Proia, A. D. Basal-like cells constitute the proliferating cell population in cystic fibrosis airways. Am. J. Respir. Crit. Care Med. 172, 1013–1018 (2005).

    Article  PubMed  Google Scholar 

  32. Leigh, M. W., Kylander, J. E., Yankaskas, J. R. & Boucher, R. C. Cell proliferation in bronchial epithelium and submucosal glands of cystic fibrosis patients. Am. J. Respir. Cell Mol. Biol. 12, 605–612 (1995).

    Article  CAS  PubMed  Google Scholar 

  33. King, N. E. et al. Correction of airway stem cells: genome editing approaches for the treatment of cystic fibrosis. Hum. Gene Ther. 31, 956–972 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Xu, Y. et al. Single-cell RNA sequencing identifies diverse roles of epithelial cells in idiopathic pulmonary fibrosis. JCI Insight 1, e90558 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Neuberger, T., Burton, B., Clark, H. & Van Goor, F. Use of primary cultures of human bronchial epithelial cells isolated from cystic fibrosis patients for the pre-clinical testing of CFTR modulators. Methods Mol. Biol. 741, 39–54 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Aros, C. J. et al. High-throughput drug screening identifies a potent Wnt inhibitor that promotes airway basal stem cell homeostasis. Cell Rep. 30, 2055–2064 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hegab, A. E. et al. Isolation and in vitro characterization of basal and submucosal gland duct stem/progenitor cells from human proximal airways. Stem Cells Transl. Med. 1, 719–724 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hegab, A. E. et al. Aldehyde dehydrogenase activity enriches for proximal airway basal stem cells and promotes their proliferation. Stem Cells Dev. 23, 664–675 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Hegab, A. E., Ha, V. L., Attiga, Y. S., Nickerson, D. W. & Gomperts, B. N. Isolation of basal cells and submucosal gland duct cells from mouse trachea. J. Vis. Exp. 14, e3731 (2012).

    Google Scholar 

  41. Paul, M. K. et al. Dynamic changes in intracellular ROS levels regulate airway basal stem cell homeostasis through Nrf2-dependent Notch signaling. Cell Stem Cell 15, 199–214 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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 

  43. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. 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 

  45. Young, M. D. & Behjati, S. SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data. GigaScience 9, giaa151 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. https://doi.org/10.1038/nbt.4314 (2018).

  48. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Tirosh, I. et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539, 309–313 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank S. Reynolds for helpful input in reviewing this manuscript. This work was supported by the Cystic Fibrosis Foundation (GOMPER17XX0 (B.N.G.), STRIPP17XX0 (B.R.S.), CARRAR19G0 (G.C.) and BOUCHE15R0 (S.H.R.)), the Tobacco-Related Disease Research Program (HIPRA 29IP-0597) (B.N.G.), National Institutes of Health (NIH) grant R01CA208303 (B.N.G.), the National Heart, Lung, and Blood Institute (PO1 HL108793) (B.R.S.), NIH grant DK065988 (S.H.R.), a grant from the W. M. Keck Foundation (B.N.G.) and a grant from Celgene/BMS (B.R.S.). J.L. was supported by the UCLA Tumor Cell Biology Training Program (USHHS Ruth L. Kirschstein Institutional National Research Service Award T32 CA009056). S.S. was supported by the UCLA Broad Stem Cell Research Center–Rose Hills Foundation Training Award and, currently, the UCLA Dissertation Year Fellowship. K.P. and B.N.G. were supported by the UCLA Broad Stem Cell Research Center, the David Geffen School of Medicine and the Jonsson Comprehensive Cancer Center, and K.P. was supported by the NIH (P01 GM099134). The research of K.P. was also supported, in part, by a Faculty Scholar grant from the Howard Hughes Medical Institute. J.E. was funded by the NIH (DP1DA044371), and J.E. and B.N.G. were supported by the Jonsson Comprehensive Cancer Center and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Ablon Scholars Award. C.J.A. and D.W.S. were supported by a UCLA Medical Scientist Training Program grant (NIH NIGMS GM008042), the NIH/NCI NRSA Predoctoral F31 Diversity Fellowship F31CA239655 (C.J.A.), the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Training Grant (C.J.A.), the T32 National Research Service Award in Tumor Cell Biology CA009056 (C.J.A.), the Parker B. Francis Foundation Fellowship (C.Y.) and UCLA CTSI KL2- NCATS KL2TR001882 (C.Y.).

Author information

Authors and Affiliations

Authors

Contributions

A.L.R. was previously known as Amy L. Firth. G.C., J.L. and J.M. designed and performed experiments, analyzed the data and prepared the manuscript. S.S., Z.L., A.P., G.Z., B.K., C.J.A., B.A.C., P.V., C.Y., D.W.S., E.I., T.M.R., E.W., A.S., M.M., A.L. and J.L. assisted in tissue handling, sampling, processing and sorting for scRNA-seq cell culture. J.E. supervised J.L. and S.S. S.H.R., E.K.V., A.L.R. and M.M. provided expertise and/or tissue analysis. K.P., J.M., B.R.S. and B.N.G. supervised the study and prepared the manuscript. All authors reviewed and edited the final manuscript.

Corresponding authors

Correspondence to Kathrin Plath, John E. Mahoney, Barry R. Stripp or Brigitte N. Gomperts.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Medicine thanks Tuomas Tammela, Jeffrey Whitsett, and the other, anonymous reviewers for their contribution to the peer review of this work. Editor recognition statement: Jerome Staal was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team

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

Extended data

Extended Data Fig. 1 Cell subsets identified across institutions.

a, Visualization of the distribution of cells from the three institutions in the integrated embedding, showed by institution and (b) by samples of origin, visualized by UMAP. cf, Network distributions with differences between institutions, visualized by UMAP. g, Major cell types identified using previously described markers, visualized by UMAP. h, Ionocyte and NE cell subsets analyzed independently of other cell types, visualized by UMAP. i, CO and CF sample contribution to cell populations and subsets, visualized by a stacked column chart. The ‘s’ indicates submucosal gland samples derived from matching ‘*’ CO and CF lungs. j, Signatures of major cell types in 10706 ALI cells, created using previously published ALI gene lists, shown by violin plots. Overlaid are boxplots showing the quartiles, whiskers showing 1.5 times interquartile range, and dots showing outliers. k, Distribution of major cell type proportions in freshly isolated and ALI datasets, for 38 and 5 independent biological samples respectively. Error bars show the standard error of the mean. l, CFTR expression level per subtype, scaled over all cells.

Extended Data Fig. 2. Secretory cell networks.

a, Heatmap showing the percent of normalized expression of the seven secretory networks across the secretory subset groups, divided by CO and CF. Each cell shows the average expression of all cells in that category, normalized by row. b, Heatmap showing the percent of normalized expression within the secretory subset groups for the top five genes selected from each secretory network based on their pan-institutional identity as either the most Up or Down in CF within the given network. Up/Down and Network classification is shown by annotation to left of heatmap and in key at right. Note for Net S7, only three genes qualified as pan-institutional. c, Bar plots showing the average expression of all genes in the remaining individual secretory networks per secretory subset group, in CO or CF cells.

Extended Data Fig. 3 Ciliated cell networks.

a, Heatmap showing the percent of normalized expression of all ten ciliated networks across the ciliated subset groups, divided by CO and CF. Each cell shows the average expression of all cells in that category, normalized by row. b, Heatmap showing the percent of normalized expression within the ciliated subset groups for the top five genes selected from each ciliated network based on their pan-institutional identity as either the most Up or Down in CF within the given network. Up/Down and Network classification is shown by annotation to left of heatmap and in key at right. c, Bar plots showing the average expression of all genes in the remaining individual ciliated networks per ciliated subset group, in CO or CF cells.

Extended Data Fig. 4 Changes in CO and CF cilia biogenesis.

aj, For distinct categories of genes related to cilia biogenesis, the expansion of cilia gene expression is shown by violin plots and UMAP, indicating the changes in CO and CF for each cell subset. Overlaid are boxplots showing the quartiles, whiskers showing 1.5 times interquartile range, and dots showing outliers. Each Pair of CO and CF show the associated P value (Wilcox test).

Extended Data Fig. 5 Surface markers of basal cell subsets.

a, Scaled expression of the top differentially expressed CD marker genes that inform specific basal cell subsets, visualized by heatmap. b, FACS plots showing segregation of total basal cells (CD326+, CD271+, CD45-, CD31−) into basal subsets based on their preferential expression of CD66 and CD266, in freshly isolated CO (upper panel) and primary hBE culture (lower panel).

Extended Data Fig. 6 Basal cell networks.

a, Heatmap showing the percent of normalized expression of the ten basal networks across the basal subset groups, divided by CO and CF. Each cell shows the average expression of all cells in that category, normalized by row. b, Heatmap showing the percent of normalized expression within the basal subset groups for the top five genes selected from each basal network based on their pan-institutional identity as either the most Up or Down in CF within the given network. Up/Down and Network classification is shown by annotation to left of heatmap and in key at right c, Bar plots showing the average expression of all genes in the remaining individual basal networks per basal subset group, in CO or CF cells.

Extended Data Fig. 7. Proliferative basal cells in CO and CF.

a, Scoring of the proliferative state (generated using a gene signature from Basal2 subset, Supplementary Table 2), of primary hBE from CO and CF, visualized by UMAP. b, Same scoring showed as violin plots with pairwise t-test comparison of CO and CF, *: p < 2.22e-16 (Wilcox test). Overlaid are boxplots showing the quartiles, whiskers showing 1.5 times interquartile range, and dots showing outliers. 3 clones were sampled for each condition.

Extended Data Fig. 8 Counting proliferative basal cell in CO and CF.

a, Representative IF images of airways showing KRT5 (green) and PCNA (cyan), all nuclei are counterstained with DAPI (blue) in the merged image. Scale bar shows 75 μm. b, Representative examples of watershed segmentation for isolated KRT5 and PCNA staining. c, Representative images indicating counting of KRT5 (green) and PCNA (cyan) expressing cells in the segmented images. Scale bar shows 75 μm. Red and yellow boxes highlight areas that provide 4x zoomed images. d, Segmentation data assumes a normal distribution. Each data point represents a possible cell and its corresponding area. Red line represents the mean area of the data and black line represents two standard deviations above the mean area. Representative tiles scan regions taken at 20x magnification for non-CF (e) and CF (f) subjects stained for KRT5 (green), PCNA (cyan) and nuclei are counterstained with DAPI (blue). Dimensions of the airways are indicated by the white lines. In all cases, images are representative of 14 CF and 17 CO fields of view.

Extended Data Fig. 9 FACs isolation of airway epithelial cells.

Representative FACS plots for isolation of epithelial cells to use in scRNAseq with 10X Genomics. Cell debris were excluded on the basis of FSC-A versus SSC-A, then doublets were removed using Trigger Pulse Width versus FSC-A (Influx). Dead cells were identified and excluded on the base of staining with DAPI. Negative gating for CD45, CD31, and CD235a, combined with positive gating for EPCAM (CD326) were used to identify epithelial cells.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Carraro, G., Langerman, J., Sabri, S. et al. Transcriptional analysis of cystic fibrosis airways at single-cell resolution reveals altered epithelial cell states and composition. Nat Med 27, 806–814 (2021). https://doi.org/10.1038/s41591-021-01332-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01332-7

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