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:

Lung regeneration by multipotent stem cells residing at the bronchioalveolar-duct junction

An Author Correction to this article was published on 07 March 2019

This article has been updated

Abstract

Characterizing the stem cells responsible for lung repair and regeneration is important for the treatment of pulmonary diseases. Recently, a unique cell population located at the bronchioalveolar-duct junctions has been proposed to comprise endogenous stem cells for lung regeneration. However, the role of bronchioalveolar stem cells (BASCs) in vivo remains debated, and the contribution of such cells to lung regeneration is not known. Here we generated a genetic lineage-tracing system that uses dual recombinases (Cre and Dre) to specifically track BASCs in vivo. Fate-mapping and clonal analysis showed that BASCs became activated and responded distinctly to different lung injuries, and differentiated into multiple cell lineages including club cells, ciliated cells, and alveolar type 1 and type 2 cells for lung regeneration. This study provides in vivo genetic evidence that BASCs are bona fide lung epithelial stem cells with deployment of multipotency and self-renewal during lung repair and regeneration.

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: Genetic lineage tracing of CC10+SPC+ BASCs with dual recombinases.
Fig. 2: Maintenance of BASCs during lung homeostasis.
Fig. 3: BASCs contribute to club cells and ciliated cells after bronchiolar injury.
Fig. 4: BASCs expand and differentiate into AT1 and AT2 cells after bleomycin-induced lung injury.
Fig. 5: Clonal analysis of BASCs in naphthalene- or bleomycin-induced lung injuries.
Fig. 6: Single-cell RNA sequencing of BASCs.
Fig. 7: BASCs are multipotent resident epithelial stem cells for lung repair and regeneration.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

Data availability

The generated sequencing data have been deposited in the GEO database under accession code GSE118891.

Change history

  • 07 March 2019

    In the version of this article initially published, the following grant numbers and recipients were missing from the Acknowledgements: XDB19000000 to H.J. and B.Z.; 81430066 and 31621003 to H.J.; 2017YFA0505500 to H.J.; and 15XD1504000 to H.J. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Kuo, C. S. & Krasnow, M. A. Formation of a neurosensory organ by epithelial cell slithering. Cell 163, 394–405 (2015).

    Article  CAS  Google Scholar 

  2. Morrisey, E. E. & Hogan, B. L. Preparing for the first breath: genetic and cellular mechanisms in lung development. Dev. Cell 18, 8–23 (2010).

    Article  CAS  Google Scholar 

  3. Rock, J. R. & Hogan, B. L. Epithelial progenitor cells in lung development, maintenance, repair, and disease. Annu. Rev. Cell Dev. Biol. 27, 493–512 (2011).

    Article  CAS  Google Scholar 

  4. Hogan, B. L. et al. Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 15, 123–138 (2014).

    Article  CAS  Google Scholar 

  5. Desai, T. J., Brownfield, D. G. & Krasnow, M. A. Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature 507, 190–194 (2014).

    Article  CAS  Google Scholar 

  6. Kumar, P. A. et al. Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell 147, 525–538 (2011).

    Article  CAS  Google Scholar 

  7. Chen, F. & Krasnow, M. A. Progenitor outgrowth from the niche in Drosophila trachea is guided by FGF from decaying branches. Science 343, 186–189 (2014).

    Article  CAS  Google Scholar 

  8. Zuo, W. et al. p63+Krt5+ distal airway stem cells are essential for lung regeneration. Nature 517, 616–620 (2014).

    Article  Google Scholar 

  9. Peng, T. et al. Hedgehog actively maintains adult lung quiescence and regulates repair and regeneration. Nature 526, 578–582 (2015).

    Article  CAS  Google Scholar 

  10. Hong, K. U., Reynolds, S. D., Watkins, S., Fuchs, E. & Stripp, B. R. Basal cells are a multipotent progenitor capable of renewing the bronchial epithelium. Am. J. Pathol. 164, 577–588 (2004).

    Article  CAS  Google Scholar 

  11. Hong, K. U., Reynolds, S. D., Watkins, S., Fuchs, E. & Stripp, B. R. In vivo differentiation potential of tracheal basal cells: evidence for multipotent and unipotent subpopulations. Am. J. Physiol. Lung Cell. Mol. Physiol. 286, L643–L649 (2004).

    Article  CAS  Google Scholar 

  12. Rock, J. R. et al. Basal cells as stem cells of the mouse trachea and human airway epithelium. Proc. Natl Acad. Sci. USA 106, 12771–12775 (2009).

    Article  CAS  Google Scholar 

  13. Rawlins, E. L. et al. The role of Scgb1a1+ Clara cells in the long-term maintenance and repair of lung airway, but not alveolar, epithelium. Cell Stem Cell 4, 525–534 (2009).

    Article  CAS  Google Scholar 

  14. Hong, K. U., Reynolds, S. D., Giangreco, A., Hurley, C. M. & Stripp, B. R. Clara cell secretory protein-expressing cells of the airway neuroepithelial body microenvironment include a label-retaining subset and are critical for epithelial renewal after progenitor cell depletion. Am. J. Respir. Cell Mol. Biol. 24, 671–681 (2001).

    Article  CAS  Google Scholar 

  15. Barkauskas, C. E. et al. Type 2 alveolar cells are stem cells in adult lung. J. Clin. Invest. 123, 3025–3036 (2013).

    Article  CAS  Google Scholar 

  16. Zacharias, W. J. et al. Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 555, 251–255 (2018).

    Article  CAS  Google Scholar 

  17. Nabhan, A., Brownfield, D. G., Harbury, P. B., Krasnow, M. A. & Desai, T. J. Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science 359, 1118–1123 (2018).

    Article  CAS  Google Scholar 

  18. Vaughan, A. E. et al. Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury. Nature 517, 621–625 (2014).

    Article  Google Scholar 

  19. Tata, P. R. et al. Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature 503, 218–223 (2013).

    Article  CAS  Google Scholar 

  20. Giangreco, A., Reynolds, S. D. & Stripp, B. R. Terminal bronchioles harbor a unique airway stem cell population that localizes to the bronchoalveolar duct junction. Am. J. Pathol. 161, 173–182 (2002).

    Article  Google Scholar 

  21. Kim, C. F. et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 121, 823–835 (2005).

    Article  CAS  Google Scholar 

  22. Nolen-Walston, R. D. et al. Cellular kinetics and modeling of bronchioalveolar stem cell response during lung regeneration. Am. J. Physiol. Lung Cell. Mol. Physiol. 294, L1158–L1165 (2008).

    Article  CAS  Google Scholar 

  23. Dovey, J. S., Zacharek, S. J., Kim, C. F. & Lees, J. A. Bmi1 is critical for lung tumorigenesis and bronchioalveolar stem cell expansion. Proc. Natl Acad. Sci. USA 105, 11857–11862 (2008).

    Article  CAS  Google Scholar 

  24. Zacharek, S. J. et al. Lung stem cell self-renewal relies on BMI1-dependent control of expression at imprinted loci. Cell Stem Cell 9, 272–281 (2011).

    Article  CAS  Google Scholar 

  25. Lee, J. H. et al. Lung stem cell differentiation in mice directed by endothelial cells via a bmp4-nfatc1-thrombospondin-1 axis. Cell 156, 440–455 (2014).

    Article  CAS  Google Scholar 

  26. He, L. et al. Enhancing the precision of genetic lineage tracing using dual recombinases. Nat. Med. 23, 1488–1498 (2017).

    Article  CAS  Google Scholar 

  27. Zhang, H. et al. Genetic lineage tracing identifies endocardial origin of liver vasculature. Nat. Genet. 48, 537–543 (2016).

    Article  Google Scholar 

  28. Madisen, L. et al. Transgenic mice for intersectional targeting of neural sensors and effectors with high specificity and performance. Neuron 85, 942–958 (2015).

    Article  CAS  Google Scholar 

  29. Madisen, L. et al. A robust and high-throughput cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  Google Scholar 

  30. Zhang, H. et al. Endocardium contributes to cardiac fat. Circ. Res. 118, 254–265 (2016).

    Article  CAS  Google Scholar 

  31. Reinert, R. B. et al. Tamoxifen-induced Cre-loxP recombination is prolonged in pancreatic islets of adult mice. PLoS One 7, e33529 (2012).

    Article  CAS  Google Scholar 

  32. Rock, J. R. et al. Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc. Natl Acad. Sci. USA 108, E1475–E1483 (2011).

    Article  CAS  Google Scholar 

  33. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    Article  CAS  Google Scholar 

  34. Montoro, D. T. et al. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 12771, 319–324 (2018).

    Article  Google Scholar 

  35. Plasschaert, L. W. et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 545, 377–381 (2018).

    Article  Google Scholar 

  36. Honda, H. et al. Leucine-rich α-2 glycoprotein promotes lung fibrosis by modulating TGF-β signaling in fibroblasts. Physiol. Rep. 5, e13556 (2017).

    Article  Google Scholar 

  37. Wang, X. et al. LRG1 promotes angiogenesis by modulating endothelial TGFβ signalling. Nature 499, 306–311 (2013).

    Article  CAS  Google Scholar 

  38. Schultz, C. J., Torres, E., Londos, C. & Torday, J. S. Role of adipocyte differentiation-related protein in surfactant phospholipid synthesis by type II cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 283, L288–L296 (2002).

    Article  CAS  Google Scholar 

  39. El Agha, E. et al. Two-way conversion between lipogenic and myogenic fibroblastic phenotypes marks the progression and resolution of lung fibrosis. Cell Stem Cell 20, 261–273.e3 (2016).

    Article  Google Scholar 

  40. Zhang, X. et al. Comparative membrane proteomic analysis between lung adenocarcinoma and normal tissue by iTRAQ labeling mass spectrometry. Am. J. Transl. Res. 6, 267–280 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, X. D. et al. Identification of adipophilin as a potential diagnostic tumor marker for lung adenocarcinoma. Int. J. Clin. Exp. Med. 7, 1190–1196 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Xu, X. et al. Evidence for type II cells as cells of origin of K-Ras-induced distal lung adenocarcinoma. Proc. Natl Acad. Sci. USA 109, 4910–4915 (2012).

    Article  CAS  Google Scholar 

  43. Noh, M. S. et al. Magnetic surface-enhanced Raman spectroscopic (M-SERS) dots for the identification of bronchioalveolar stem cells in normal and lung cancer mice. Biomaterials 30, 3915–3925 (2009).

    Article  CAS  Google Scholar 

  44. Kotton, D. N. & Morrisey, E. E. Lung regeneration: mechanisms, applications and emerging stem cell populations. Nat. Med. 20, 822–832 (2014).

    Article  CAS  Google Scholar 

  45. Pardo-Saganta, A. et al. Parent stem cells can serve as niches for their daughter cells. Nature 523, 597–601 (2015).

    Article  CAS  Google Scholar 

  46. Jain, R. et al. Plasticity of Hopx+ type I alveolar cells to regenerate type II cells in the lung. Nat. Commun. 6, 6727 (2015).

    Article  CAS  Google Scholar 

  47. Giangreco, A. et al. Stem cells are dispensable for lung homeostasis but restore airways after injury. Proc. Natl Acad. Sci. USA 106, 9286–9291 (2009).

    Article  CAS  Google Scholar 

  48. Tian, X. et al. Subepicardial endothelial cells invade the embryonic ventricle wall to form coronary arteries. Cell Res. 23, 1075–1090 (2013).

    Article  CAS  Google Scholar 

  49. Liu, Q. et al. Genetic targeting of sprouting angiogenesis using Apln-CreER. Nat. Commun. 6, 6020 (2015).

    Article  CAS  Google Scholar 

  50. Degryse, A. L. et al. Repetitive intratracheal bleomycin models several features of idiopathic pulmonary fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 299, L442–L452 (2010).

    Article  CAS  Google Scholar 

  51. He, L. et al. Preexisting endothelial cells mediate cardiac neovascularization after injury. J. Clin. Invest. 127, 2968–2981 (2017).

    Article  Google Scholar 

  52. Liu, Q. et al. Genetic lineage tracing identifies in situ Kit-expressing cardiomyocytes. Cell Res. 26, 119–130 (2016).

    Article  CAS  Google Scholar 

  53. Chen, J. et al. Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nat. Protoc. 12, 566–580 (2017).

    Article  CAS  Google Scholar 

  54. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    Article  CAS  Google Scholar 

  55. Pertea, M., Kim, D., Pertea, G. M., Leek, J. T. & Salzberg, S. L. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protoc. 11, 1650–1667 (2016).

    Article  CAS  Google Scholar 

  56. Anders, S., Pyl, P. T. & Huber, W. HTSeq: a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  Google Scholar 

  57. Kiselev, V. Y. et al. SC3: consensus clustering of single-cell RNA-seq data. Nat. Methods 14, 483–486 (2017).

    Article  CAS  Google Scholar 

  58. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the Strategic Priority Research Program of the Chinese Academy of Sciences (CAS, XDB19000000 to H.J. and B.Z.; XDA16020204 to B.Z.; XDA16020404 to G.P.; and XDA16020501 to N.J.), the National Science Foundation of China (31730112, 91639302, 31625019, 91849202 and 81761138040 to B.Z.; 31601168 to Q.L.; 31701292 and 81872241 to L.H.; and 31571503, 91749122 and 81872132 to X.T.; 81430066 and 31621003 to H.J.), the National Key Research and Development Program of China (2018YFA0107900 and 2016YFC1300600 to X.T.; 2018YFA0108100 and 2017YFC1001303 to L.H.; 2017YFA0505500 to H.J.), the Key Project of Frontier Sciences of CAS (QYZDB-SSW-SMC003), the Shanghai Science and Technology Commission (17ZR1449600 to B.Z., 17ZR1449800 to X.T., 15XD1504000 to H.J. and 15XD1504000 to B.Z.), the Shanghai Yangfan Project (16YF1413400 to L.H.), the China Postdoctoral Innovative Talent Support Program (BX20180338 to Y.L.), China Young Talents Lift Engineering (YESS20160050 to Q.L. and 2017QNRC001 to L.H.), the collaboration fund of Research Beyond Borders at Boehringer Ingelheim Pharma GmbH (B.Z.), Astrazeneca (B.Z.) and a Royal Society-Newton Advanced Fellowship (B.Z., NA170109) and the Program for Guangdong Introducing Innovative and Entrepreneurial Teams (2017ZT07S347 to B.Z.). We thank the Shanghai Model Organisms Center, Inc. (SMOC) and Nanjing Biomedical Research Institute of Nanjing University for mouse generation. We also acknowledge technical help from L. Qiu, W. Bian, T. Zhang and members of National Center for Protein Science Shanghai for assistance in flow cytometry and microscopy and Y. Xing for antibody sharing. We thank P. Nicklin, M. Franti and W. Zhang for valuable suggestions and comments on this study. We also thank the Genome Tagging Project (GTP) Center for support.

Author information

Authors and Affiliations

Authors

Contributions

Q.L., K.L. and B.Z. designed the study and wrote the manuscript. Q.L., K.L. and G.C. performed experiments and analyzed the data. W.G., G.C., G.P. and N.J. performed scRNA-seq and analyzed data. X.H., S.Y., Z.Q., Y.L., R.Y., W.P., L.Z., L.H., H.Z., W.Y., M.T., X.T., D.C., Y.N., S.H., T.R., Z.Q., H.H. and Y.A.Z. bred the mice, performed experiments or provided material, important suggestions and valuable comments. H.J. designed the study and provided valuable comments. B.Z. supervised the study and analyzed the data.

Corresponding authors

Correspondence to Guangdun Peng, Hongbin Ji or Bin Zhou.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Scgb1a1-CreER targets mainly CC10+ club cells.

(a) Strategy for labeling of CC10+ cells by Cre-loxP system. (b) Whole-mount bright-field and fluorescence view of lung from adult Scgb1a1-CreER;R26-tdTomato mice 1 week post Tamoxifen induction. (c-f) Immunostaining for tdTomato and lineage markers CC10, CK5, SPC, aSMA or VE-cad on lung sections. White arrowheads in c indicate weak expression of CC10 in some tdTomato+ cells at BADJ regions. Boxed region are magnified on the right. Scale bars, yellow, 1 mm; white 100 µm. Each image is representative of 5 individual samples.

Supplementary Figure 2 Cell labeling by Scgb1a1-CreER;R26-tdTomato.

(a) Immunostaining for tdTomato and SPC on lung sections collected from Scgb1a1-CreER;R26-tdTomato mice treated with 0.2 mg/g tamoxifen. In addition to club cell labeling, Scgb1a1-CreER also labels a subset of AT2 cells (arrowheads). (b) Immunostaining for CC10, SPC and tdTomato on lung tissue sections from Scgb1a1-CreER;R26-tdTomato mice treated with 0.025 mg/g tamoxifen. A significant fewer tdTomato+ AT2 cells (arrowhead) were detected in 0.025 mg/g tamoxifen treated mice (b) compared with 0.2 mg/g tamoxifen treated mice (a). In 0.025 mg/g tamoxifen treated samples, tdTomato+ BASCs (arrows) could also be detected. Scale bars, 100 µm. Each figure is representative of 5 individual biological samples.

Supplementary Figure 3 Sftpc-DreER targets mainly SPC+ AT2 cells.

(a) Schematic figure showing knock-in strategy for Sftpc-DreER allele by homologous recombination. (b) Strategy for labeling of SPC+ cells by Dre-rox recombination. (c) Whole-mount fluorescence image showing tdTomato labeling of lung from Sftpc-DreER;R26-RSR-tdTomato mouse at 1 week after tamoxifen treatment. Dotted lines mark trachea. (d-h) Immunostaining for tdTomato and cell lineage markers SPC, CC10, CK5, T1a, aSMA and VE-cad on Sftpc-DreER;R26-RSR-tdTomato lung. Boxed regions are magnified on the right. XZ and YZ indicate signals from dotted lines on Z-stack image. Scale bars, yellow, 1 mm; white, 100 µm. Each image is representative of 5 individual samples.

Supplementary Figure 4 Sftpc-DreER labels most AT2 cells (CC10–SPC+) and BASCs (CC10+SPC+), and very few club cells (CC10+SPC–).

(a-d) Immunostaining for tdTomato, CC10 and SPC on lung tissue sections collected from Sftpc-DreER;R26-RSR-tdTomato mice at 1week after tamoxifen induction (0.2 mg/g). The labeled BASCs (arrowheads) were located at BADJs. Boxed regions in a are magnified in b,c,d. Scale bars, 100 µm. Each figure is representative of 5 individual biological samples.

Supplementary Figure 5 BASCs-Tracer does not label AT1 cells, ciliated cells or neuroendocrine cells.

(a-c) Immunostaining for tdTomato and T1a (AT1 cell marker), β-tubulin (ciliated cell marker) or CGRP (neuroendocrine cell marker) on BASCs-Tracer mouse lung sections. Tamoxifen was induced one week before analysis. Scale bars, 100 µm. Each image is representative of 5 individual samples.

Supplementary Figure 6 BASCs-Tracer does not label K5+ or P63+ cells.

(a-d) Immunostaining for tdTomato and K5 (a) or P63 (c) on lung tissue sections. Tamoxifen was induced at 1 week before tissue collection. K5 and P63 could be detected in the lung trachea (b) or stomach (d) tissue sections of mouse. Scale bars, 100 µm. Each image is a representative of 4 biologically independent sample.

Supplementary Figure 7 Scgb1a1-CreER or Sftpc-DreER rarely labels cells by R26-RSR-LSL-tdTomato reporter.

(a,b) Schematic figure showing crossing of Scgb1a1-CreER or Sftpc-DreER with R26-RSR-LSL-tdTomato reporter respectively. Tamoxifen was injected one week before tissue analysis. (c,d) Whole-mount fluorescence and bright-field view of lungs from Scgb1a1-CreER;R26-RSR-LSL-tdTomato mouse (c) or Sftpc-DreER;R26-RSR-LSL-tdTomato mouse (d). (e-l) Immunostaining for tdTomato and cell lineage markers T1a, SPC, CC10 and CK5. Boxed region are magnified on the right. Scale bars, yellow, 1 mm; white, 100 µm. Each image is representative of 5 individual samples.

Supplementary Figure 8 Negligible leakiness of BASCs-Tracer mouse without tamoxifen induction.

(a,b) Immunostaining for tdTomato, SPC and CC10 on lung sections of BASCs-Tracer mouse. Boxed region is magnified in the below images. Most fields in the lung sections are negative for tdTomato (a), with only very few fields that contain sparse tdTomato+ cells (b). The extremely rare tdTomato+ cell is located in BADJ region, and is expressing SPC and CC10 (arrowhead). (c) The sparse tdTomato+ cell (arrowhead) does not express AT1 cell marker T1a. Scale bars, 100 µm. Each image is representative of 5 individual samples.

Supplementary Figure 9 BASCs differentiate mainly into club cells after naphthalene-induced lung injury.

Immunostaining for CC10, tdTomato and SPC on lung tissue sections after naphthalene treatment. Yellow arrowheads indicate tdTomato+CC10+SPC+ BASCs at BADJ; white arrowheads indicate tdTomato+CC10+SPC club cells. Scale bars, 100 µm. Each image is representative of 5 individual samples.

Supplementary Figure 10 BASCs contribute to club cells and ciliated cells after bronchiolar injury.

(a) Schematic figure showing timeline for tamoxifen (Tam), Naphthalene (Naph.) treatment, and lung tissue analysis. Mice were treated with naphthalene at 3 weeks after tamoxifen induction. (b) Diagram showing BSACs regenerate terminal bronchiole after Naph. injury. (c) Immunostaining for CC10, SPC and tdTomato on lung section shows a subset of tdTomato+ cells residing in BADJ continue to express CC10 and SPC (arrowheads), while the majority of tdTomato+ cells are CC10+SPC and detected in the terminal bronchiole. (d) Immunostaining for tdTomato, CC10 and β-Tubulin or Acetylated-Tubulin on lung section shows tdTomato+ ciliated cells (arrowheads). (e) Immunostaining for CGRP, tdTomato and CC10 on tissue sections shows tdTomato+ cells (arrowheads) do not express CGRP. (f) Immunostaining for T1a and tdTomato on lung section shows tdTomato+ cells (arrowheads) do not express T1a. Scale bars, 100 µm. Each image is a representative of five individual samples.

Supplementary Figure 11 Expansion of BASC-derived AT1 and AT2 cells after bleomycin-induced lung injury.

(a) Schematic figure showing experimental design. EdU was injected at 24 hours before analysis. (b) Immunostaining for EdU on lung sections after bleomycin or Vehicle treatment. (c) Quantification of the percentage of EdU+ cells in bleomycin (Bleom.) or vehicle-treated lung tissues. P is calculated by two-tailed t-test; n = 5 biologically independent mice; data are showing by box and whiskers plot; box spans the interquartile range with band inside the box represents median and whiskers represent maximum and minimum values. (d-f) Immunostaining for tdTomato, EdU and SPC, CC10 or T1a on sections. Dotted line demarcates bronchioles. Boxed regions are magnified on the right. Arrowheads indicate EdU+tdTomato+ cells. YZ indicate signals from dotted lines on Z-stack images in f (Vehicle). Scale bars, 100 µm. (g) Immunostaining for CC10, tdTomato and SPC on lung tissue sections shows that the majority of tdTomato+ cells are SPC+CC10 close to the BADJs. Each image is a representative of 5 individual samples.

Supplementary Figure 12 BASCs do not contribute to fibroblasts, pericytes, smooth muscle cells or endothelial cells in bleomycin-treated lung.

(a) Sirius Red staining on bleomycin or vehicle treated lung sections. (b-g) Immunostaining for tdTomato and different cell lineage markers PDGFRa, PECAM, PDGFRb, T1a, aSMA and VE-cad. XZ and YZ indicates signals from dotted lines on Z-stack images. Boxed regions are magnified on the right. Scale bars, 100 µm. Each image is representative of 5 individual samples.

Supplementary Figure 13 BASCs contribute to AT1 and AT2 cells after alveolar injury.

(a) Schematic figure showing timeline for tamoxifen (Tam), bleomycin treatment, and lung tissue analysis. Bleomycin is treated at 3 weeks after tamoxifen induction. (b) Diagram showing BASCs regenerate alveoli after bleomycin injury. (c) Immunostaining for T1a and tdTomato on lung section shows a subset of tdTomato+ cells residing in BADJ region express AT1 (arrowheads). XZ and YZ indicate signals from dotted lines on Z-stack images. (d) Immunostaining for SPC and tdTomato on lung section shows tdTomato+SPC+ AT2 cells (arrowheads). (e) Immunostaining for CC10, SPC and tdTomato on tissue sections shows most tdTomato+ cells (arrowheads) differentiate into SPC+ AT2 cells and do not express CC10. Scale bars, 100 µm. Each image is a representative of five individual samples.

Supplementary Figure 14 Clonal analysis of BASCs after lung injuries.

(a) Immunostaining for GFP, YFP, RFP, Acetylated-tubulin on Sftpc-DreER;Scgb1a1-CreER;R26-Confetti2 mouse lung sections after naphthalene treatment. Arrowheads indicate YFP+Acetylated-tubulin+ cell or RFP+Acetylated-tubulin+ cells in naphthalene-induced lung. (b-e) Immunostaining for GFP, YFP, RFP, CC10, SPC or T1a on Sftpc-DreER;Scgb1a1-CreER;R26-Confetti2 mouse lung sections after naphthalene, vehicle or bleomycin treatment. Arrowheads indicates GFP/YFP+SPC+ cell in naphthalene-induced lung (b), GFP+SPC+ cell in vehicle-induced lung (c), RFP+CC10+ cell in bleomycin-induced lung (d) and RFP+T1a cell in vehicle-treated lung (e). Scale bars, 100 µm. Each image is a representative of 4 individual samples.

Supplementary Figure 15 Comparison of cell clusters from scRNA-seq.

(a) t-SNE of 480 scRNA-seq profiles (points), colored by expression of selected AT2, Club cell and Ciliated cell markers. (b) Pearson correlation coefficients (r) between 4 cell clusters. (c,d) Distribution of expression levels of Plin2 and Lrg1 in each cell cluster, Violin plots show the Gaussian kernel probability densities of the data. (e-h) t-SNE plot of 455 scRNA-seq profiles (points) including AT2, BASCs-1, BASCs-2 and Club cell clusters, showing genes enriched in BASCs-1 (e,f) and BASCs-2 (g,h) subpopulation.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Q., Liu, K., Cui, G. et al. Lung regeneration by multipotent stem cells residing at the bronchioalveolar-duct junction. Nat Genet 51, 728–738 (2019). https://doi.org/10.1038/s41588-019-0346-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0346-6

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