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:

Alveolar fibroblast lineage orchestrates lung inflammation and fibrosis

Abstract

Fibroblasts are present throughout the body and function to maintain tissue homeostasis. Recent studies have identified diverse fibroblast subsets in healthy and injured tissues1,2, but the origins and functional roles of injury-induced fibroblast lineages remain unclear. Here we show that lung-specialized alveolar fibroblasts take on multiple molecular states with distinct roles in facilitating responses to fibrotic lung injury. We generate a genetic tool that uniquely targets alveolar fibroblasts to demonstrate their role in providing niches for alveolar stem cells in homeostasis and show that loss of this niche leads to exaggerated responses to acute lung injury. Lineage tracing identifies alveolar fibroblasts as the dominant origin for multiple emergent fibroblast subsets sequentially driven by inflammatory and pro-fibrotic signals after injury. We identify similar, but not completely identical, fibroblast lineages in human pulmonary fibrosis. TGFβ negatively regulates an inflammatory fibroblast subset that emerges early after injury and stimulates the differentiation into fibrotic fibroblasts to elicit intra-alveolar fibrosis. Blocking the induction of fibrotic fibroblasts in the alveolar fibroblast lineage abrogates fibrosis but exacerbates lung inflammation. These results demonstrate the multifaceted roles of the alveolar fibroblast lineage in maintaining normal alveolar homeostasis and orchestrating sequential responses to lung injury.

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: Scube2-creER specifically labels alveolar fibroblasts and ablation of these cells leads to the loss of alveolar stem cell niches.
Fig. 2: Lineage tracing by scRNA-seq reveals alveolar fibroblasts as the origin of multiple emergent fibroblast subsets.
Fig. 3: Alveolar fibroblasts sequentially differentiate into inflammatory and fibrotic fibroblasts in mouse and human pulmonary fibrosis.
Fig. 4: Cthrc1-creER mouse demonstrates the pro-fibrotic function of CTHRC1+ fibroblasts.
Fig. 5: Tgfbr2 cKO in alveolar fibroblasts abrogates fibrosis but exacerbates inflammation.

Similar content being viewed by others

Data availability

The scRNA-seq data generated in this study are deposited in Gene Expression Omnibus (GEO) under accession GSE210341. Human scRNA-seq data analyses were performed using publicly available data from accessions GSE132771, GSE147066 and GSE135893Source data are provided with this paper.

Code availability

The codes used in the scRNA-seq analysis are available on GitHub (https://github.com/TatsuyaTsukui/AlveolarLineage).

References

  1. Buechler, M. B. et al. Cross-tissue organization of the fibroblast lineage. Nature 593, 575–579 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Tsukui, T. et al. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat. Commun. 11, 1920 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Plikus, M. V. et al. Fibroblasts: origins, definitions, and functions in health and disease. Cell 184, 3852–3872 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hinz, B. & Lagares, D. Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases. Nat. Rev. Rheumatol. 16, 11–31 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Henderson, N. C., Rieder, F. & Wynn, T. A. Fibrosis: from mechanisms to medicines. Nature 587, 555–566 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ruiz-Villalba, A. et al. Single-cell RNA sequencing analysis reveals a crucial role for CTHRC1 (collagen triple helix repeat containing 1) cardiac fibroblasts after myocardial infarction. Circulation 142, 1831–1847 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Melms, J. C. et al. A molecular single-cell lung atlas of lethal COVID-19. Nature 595, 114–119 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Pakshir, P. et al. The myofibroblast at a glance. J. Cell Sci. 133, jcs227900 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Friedman, S. L., Sheppard, D., Duffield, J. S. & Violette, S. Therapy for fibrotic diseases: nearing the starting line. Sci. Transl. Med. 5, 167sr1 (2013).

    Article  PubMed  Google Scholar 

  10. Narvaez del Pilar, O., Gacha Garay, M. J. & Chen, J. Three-axis classification of mouse lung mesenchymal cells reveals two populations of myofibroblasts. Development 149, dev200081 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. McGowan, S. E. & Torday, J. S. The pulmonary lipofibroblast (lipid interstitial cell) and its contributions to alveolar development. Annu. Rev. Physiol. 59, 43–62 (1997).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hasegawa, K. et al. Fraction of MHCII and EpCAM expression characterizes distal lung epithelial cells for alveolar type 2 cell isolation. Respir. Res. 18, 150 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Gurczynski, S. J. & Moore, B. B. IL-17 in the lung: the good, the bad, and the ugly. Am. J. Physiol. 314, L6–L16 (2018).

    Google Scholar 

  15. Croft, A. P. et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 570, 246–251 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Öhlund, D. et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 214, 579–596 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Boyd, D. F. et al. Exuberant fibroblast activity compromises lung function via ADAMTS4. Nature 587, 466–471 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Korsunsky, I. et al. Cross-tissue, single-cell stromal atlas identifies shared pathological fibroblast phenotypes in four chronic inflammatory diseases. Med 3, 481–518.e14 (2022).

    Article  CAS  PubMed  Google Scholar 

  19. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Liu, W. et al. Antiflammin-1 attenuates bleomycin-induced pulmonary fibrosis in mice. Respir. Res. 14, 101 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Strobel, B. et al. Time and phenotype-dependent transcriptome analysis in AAV-TGFβ1 and Bleomycin-induced lung fibrosis models. Sci. Rep. 12, 12190 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Elyada, E. et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9, 1102–1123 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Adams, T. S. et al. Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci. Adv. 6, eaba1983 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. Habermann, A. C. et al. Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci. Adv. 6, eaba1972 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Katzenstein, A.-L. A. & Myers, J. L. Idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 157, 1301–1315 (1998).

    Article  CAS  PubMed  Google Scholar 

  26. Barratt, S. L., Creamer, A., Hayton, C. & Chaudhuri, N. Idiopathic pulmonary fibrosis (IPF): an overview. J. Clin. Med. 7, 201 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kuhn, C. et al. An immunohistochemical study of architectural remodeling and connective tissue synthesis in pulmonary fibrosis. Am. Rev. Respir. Dis. 140, 1693–1703 (1989).

    Article  PubMed  Google Scholar 

  28. Massagué, J. & Sheppard, D. TGF-β signaling in health and disease. Cell 186, 4007–4037 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Valenzi, E. et al. Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann. Rheum. Dis. 78, 1379–1387 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Dominguez, C. X. et al. Single-cell RNA sequencing reveals stromal evolution into LRRC15+ myofibroblasts as a determinant of patient response to cancer immunotherapy. Cancer Discov. 10, 232–253 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Binks, A. P., Beyer, M., Miller, R. & LeClair, R. J. Cthrc1 lowers pulmonary collagen associated with bleomycin‐induced fibrosis and protects lung function. Physiol. Rep. 5, e13115 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Li, J. et al. Autocrine CTHRC1 activates hepatic stellate cells and promotes liver fibrosis by activating TGF-β signaling. EBioMedicine 40, 43–55 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Voehringer, D., Liang, H.-E. & Locksley, R. M. Homeostasis and effector function of lymphopenia-induced ‘memory-like’ T cells in constitutively T cell-depleted mice. J. Immunol. 180, 4742–4753 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. 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  PubMed  Google Scholar 

  35. Yata, Y. et al. DNase I-hypersensitive sites enhance alpha1(I) collagen gene expression in hepatic stellate cells. Hepatology 37, 267–276 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Redente, E. F. et al. Age and sex dimorphisms contribute to the severity of bleomycin-induced lung injury and fibrosis. Am. J. Physiol. 301, L510–L518 (2011).

    CAS  Google Scholar 

  37. Henderson, N. C. et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat. Med. 19, 1617–1624 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 8, 329–337.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Haghverdi, L., Lun, A. T. L., Morgan, M. D. & Marioni, J. C. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat. Biotechnol. 36, 421–427 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Marsh, S., Salmon, M. & Hoffman, P. samuel-marsh/scCustomize: version 1.1.1. Zenodo https://doi.org/10.5281/zenodo.7534950 (2023).

  43. Susaki, E. A. et al. Versatile whole-organ/body staining and imaging based on electrolyte-gel properties of biological tissues. Nat. Commun. 11, 1982 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Meeker, A. K., Heaphy, C. M., Davis, C. M., Roy, S. & Platz, E. A. Photochemical pre-bleaching of formalin-fixed archival prostate tissues significantly reduces autofluorescence to facilitate multiplex immunofluorescence staining. Preprint at bioRxiv https://doi.org/10.1101/2021.11.09.467916 (2021).

  45. Crowley, G. et al. Quantitative lung morphology: semi-automated measurement of mean linear intercept. BMC Pulm. Med. 19, 206 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank J. Zhang for support with the generation of knock-in mice; W. Eckalbar for support with computational analysis; and C. Molina for assistance with experiments. T.T. was supported by the Japan Society for the Promotion of Science (JSPS Overseas Research Fellowship), the Uehara Memorial Foundation, the Mochida Memorial Foundation for Medical and Pharmaceutical Research, and the Frontiers in Medical Research Fellowship from the California Foundation for Molecular Biology. This work was supported by HL155786 (T.T.), HL142568 (D.S.), a sponsored research agreement from AbbVie (D.S.), and the Nina Ireland Program for Lung Health (P.J.W.). We thank UCSF core facilities: Laboratory for Cell Analysis supported by P30CA082103, Center for Advanced Light Microscopy supported by S10 Shared Instrumentation grant (1S10OD017993-01A1), and UCSF PBBR, Gladstone Transgenic Gene Targeting Core, Gladstone Genomics core, and Center for Advanced Technology supported by UCSF PBBR, RRP IMIA, and 1S10OD028511-01.

Author information

Authors and Affiliations

Authors

Contributions

T.T. and D.S. conceived the study, interpreted the data and wrote the manuscript. T.T. performed and analysed the experiments. P.J.W. procured human samples. D.S. supervised the study.

Corresponding author

Correspondence to Dean Sheppard.

Ethics declarations

Competing interests

D.S. is a founder of Pliant Therapeutics and has received research funding from Abbvie, Pfizer and Pliant Therapeutics. D.S. serves on the Scientific Review Board for Genentech and on the Inflammation Scientific Advisory Board for Amgen. P.J.W. received research funding from Boehringer Ingelheim, Pliant Therapeutics, and Genentech.

Peer review

Peer review information

Nature thanks Christopher Buckley, Thomas Wynn 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 figures and tables

Extended Data Fig. 1 Scube2-CreER specifically labels alveolar fibroblasts, which provide a niche to support AT2 cells.

a, UMAP plots of all lung cells from scRNA-seq data of ref. 2, for cell types (left) and Scube2 expression (right). b, UMAP plots of Col1a1+ cells from scRNA-seq data of ref. 2, for cell types (left) and Scube2 expression (right). cf, Gating strategy to evaluate the specificity of tdTomato+ cells for lineage (CD31, CD45, EpCAM, Mcam, Ter119), Sca1, and CD9. g, Gating strategy for alveolar fibroblasts. h, Flow cytometric quantification for percent tdTomato+ of alveolar fibroblasts. n = 3 mice. i, Pi16 staining of a lung section from Scube2-CreER/Rosa26-tdTomato Col-GFP mouse. tdTomato is shown in magenta. Col-GFP is shown in green. Pi16 is shown in grey. DAPI is shown in blue. aw, airway. j, Three representative planes from z-stack images with z-positions are shown below the images. tdTomato is shown in red. proSP-C is shown in cyan. Asterisks indicate the same proSP-C+ cell. Arrows point to projections extending from alveolar fibroblasts. Scale bars, 200 μm (i) or 5 μm (j). Data are mean ± SEM. Data are representative of at least two independent experiments.

Source data

Extended Data Fig. 2 Scube2-CreER-labelled alveolar fibroblasts provide a niche to support AT2 cells.

a,b, Gating strategy for alveolar fibroblast-ablation experiments with Scube2-CreER/Rosa26-DTA mice. a, Gating strategy for EpCAM+ subpopulations. b, Gating strategy for fibroblast subsets. Lineage markers include CD31, CD45, EpCAM, and Ter119. c, Flow cytometric cell count for each population, normalized to means of vehicle groups. n = 4 (tamoxifen) or 5 (vehicle) mice. d, Representative images of H&E staining of lung sections from alveolar fibroblast-ablation experiments. Scale bars, 200 μm. e, Quantification of mean linear intercept of alveolar regions. n = 4 (tamoxifen) or 5 (vehicle) mice. f, qPCR analysis of whole lung cells 6 days after bleomycin treatment. n = 5 mice. g, Gating strategy for CD4 and γδ T cells. h, Gating strategy for ILCs. i, Flow cytometric quantification in bleomycin-treated lungs. n = 4 (Rosa26-DTA/DTA) or 7 (Rosa26-WT/WT) mice. j, Gating strategy for IL-17a+ cells. Data are mean ± SEM. Data are representative of at least two experiments. Statistical analysis was performed using unpaired two-tailed t-test followed by Holm–Sidak’s multiple-comparisons adjustment (f) or two-way ANOVA followed by Sidak’s multiple comparison test (i).

Source data

Extended Data Fig. 3 Longitudinal scRNA-seq reveals multiple fibroblast subsets that emerge after lung injury.

a, Gating strategy for purifying lineage (CD31, CD45, EpCAM, Ter119)− mesenchymal cells for scRNA-seq. b, UMAP plots for cells obtained before (day 0) or at various time points after bleomycin treatment. c, Dot plots showing the top differentially expressed genes for each cluster. d, GO enrichment analysis by DAVID for differentially expressed genes of inflammatory fibroblasts. e, GO enrichment analysis by DAVID for differentially expressed genes of stress-activated fibroblasts. f, GO over-representation analysis with the Fisher test for all clusters.

Source data

Extended Data Fig. 4 Lineage tracing by scRNA-seq reveals alveolar fibroblasts as the origin of multiple pathologic fibroblast subsets.

a, UMAP plot with tdTomato expression. b, Violin plot for tdTomato shows peaks for tdTomatolow or tdTomatohigh cells. The threshold for tdTomato+ cells was defined as an expression level > 3.5. c, UMAP plots with tdTomato expression split by biological replicates. d, Plot showing percent tdTomato+ of alveolar fibroblasts (x-axis) versus percent tdTomato+ of fibrotic fibroblasts (y-axis) for each biological replicate.

Source data

Extended Data Fig. 5 Scube2-CreER-labelled alveolar fibroblasts differentiate into fibrotic or inflammatory fibroblasts after lung injury.

a, Maximum projection of whole lung imaging for untreated or bleomycin day 14 Scube2-CreER/Rosa26-tdTomato mice. b, Representative optical sections from whole lung imaging. tdTomato is shown in magenta. Autofluorescence is shown in grey (a, b). c, Flow cytometry plots showing the increase of CD9+ cells among Scube2-CreER-labeled (tdTomato+) cells on day 21 after bleomycin treatment. d, Flow cytometric quantification of percent CD9+ of tdTomato+ cells. n = 4 mice. Statistical analysis was performed using unpaired two-tailed t-test. e, qPCR analysis of sorted cells from Scube2-CreER/Rosa26-tdTomato mice. All lineage (CD31, CD45, EpCAM, Ter119)− tdTomato+ cells (untreated or bleomycin day 21) or lineage− tdTomato+ CD9+ cells (bleomycin day 21) were sorted. The y-axis is the relative expression level to the housekeeping gene Rps3. n = 3 mice. f, Saa3 staining in sections from Scube2-CreER/Rosa26-tdTomato mice 7 days after bleomycin treatment. Arrows indicate tdTomato and Saa3 double-positive cells. g, Quantification of percent Saa3+ of tdTomato+ cells. n = 3 mice. tdTomato is shown in red. Saa3 is shown in cyan. DAPI is shown in blue. Collagen 4 is shown in grey. Data are mean ± SEM. Data are representative of at least two independent experiments. Scale bars, 1 mm (a, b), 20 μm (f).

Source data

Extended Data Fig. 6 Alveolar fibroblasts up-regulated activation markers and formed silicotic nodules in the silicosis model.

a, Time course of tamoxifen and silica treatment. b, Representative lung sections of saline or silica-treated mice. c,d, Sirius red staining of silica-treated lung section imaged as bright field (c) or polarized light (d). e, Fluorescence imaging of a sequential section of c,d. f, Representative images of silicotic nodules. g, Histological quantification of % tdTomato+ cells of Pdgfra+ cells inside silicotic nodules. n = 5 mice. h, Flow cytometric analysis of Pdgfra and CD9 on tdTomato+ cells from saline or silica-treated lungs. i, Flow cytometric quantification of %CD9+ Pdgfra-low cells of tdTomato+ cells. n = 5 mice. j,k, qPCR analysis of purified populations. The y-axis is the relative expression level to the housekeeping gene Rps3. n = 5 mice. Scale bars, 200 μm (b), 500 μm (c-e), 50 μm (f). Data are mean ± SEM. Data are representative of at least two independent experiments. Statistical analysis was performed using unpaired two-tailed t-test (i,k) or Tukey’s multiple comparisons test after one-way ANOVA (j).

Source data

Extended Data Fig. 7 TGF-β1 antagonizes inflammatory marker expression induced by IL-1β and induces fibrotic markers.

a, Pseudotime analysis of tdTomato+ clusters suggests that both stress-activated fibroblasts and fibrotic fibroblasts can emerge from inflammatory fibroblasts. b, UMAP plots of re-clustered alveolar, inflammatory, and fibrotic lineage split by days after bleomycin treatment. c, Expression of selected markers on UMAP plots. d, Schematic of sequential cytokine stimulations for primary alveolar fibroblasts. e, qPCR analysis after sequential cytokine stimulations. Group names indicate (first stimulation) → (second stimulation). DMEM means medium-only control. The y-axis is the relative expression level to the housekeeping gene Rps3. n = 3 wells. Data are mean ± SEM. Data are representative of two independent experiments.

Source data

Extended Data Fig. 8 Re-analysis of our publicly available scRNA-seq data from human pulmonary fibrosis reveals inflammatory and fibrotic clusters.

a, UMAP plots of re-clustered pathologic and alveolar clusters shown for individual patients or donors (control). b, Dot plot showing top differentially expressed genes for each cluster. c, GO enrichment analysis by DAVID for differentially expressed genes in inflammatory fibroblast 1 cluster. d, GO enrichment analysis by DAVID for differentially expressed genes in inflammatory fibroblast 2 cluster. e, Heat map showing Spearman’s correlation coefficients of average gene expression from mouse and human emergent clusters. f, GO over-representation analysis with the Fisher test for all clusters.

Source data

Extended Data Fig. 9 Combined analysis of publicly available scRNA-seq data sets from human pulmonary fibrosis from 3 groups reveals conserved inflammatory and fibrotic clusters.

a, UMAP plot of mesenchymal cells from ref. 23. b, Expression levels of selected genes on UMAP plot of mesenchymal cells in ref. 23 show alveolar and pathologic fibroblast clusters. c, UMAP plot of mesenchymal cells from ref. 24. d, Expression levels of selected genes on UMAP plot of mesenchymal cells from ref. 24 show that the “Myofibroblasts” cluster contains alveolar and pathologic fibroblasts. e, UMAP plot after combining alveolar and pathologic fibroblasts from refs. 2,23,24. f, UMAP plots of combined data split by original data set. g, UMAP plots shown for each data set and coloured by samples. h, Expression levels of selected markers for fibrotic (COL1A1, CTHRC1), inflammatory1 (SFRP2, CXCL12), inflammatory2 (SFRP4, CXCL14) and alveolar fibroblasts (NPNT, TCF21) on UMAP plots.

Extended Data Fig. 10 Cthrc1-CreER mouse demonstrates the pro-fibrotic function of Cthrc1+ fibroblasts.

a, Gating strategy for cell size, singlet, and live cells. b, Flow cytometry plots show an increase in lineage− tdTomato+ cells on day 14 after bleomycin treatment. c, Flow cytometric cell count of lineage (CD31, CD45, EpCAM, Ter119)− tdTomato+ cells on day 14. n = 3 (Saline-Vehicle) or 5 (Saline-Tamoxifen, Bleo-Tamoxifen) mice. d, Flow cytometry plots show CD9 expression on tdTomato+ lineage− cells increases between day 14 and 21. e, Flow cytometric quantification of percent CD9+ of tdTomato+ lineage− cells. n = 5 mice. f, Mean fluorescence intensity (MFI) of CD9 on tdTomato+ lineage− cells. n = 5 mice. g, Saa3 staining in sections from Cthrc1-CreER/Rosa26-tdTomato mice 14 days after bleomycin treatment with tamoxifen injected on days 8 -12. tdTomato is shown in red. Saa3 is shown in cyan. Collagen 4 is shown in grey. DAPI is shown in blue. h, Schematic for localization of inflammatory and fibrotic fibroblasts. i, Representative images of sequential lung sections from Cthrc1-CreER/Rosa26-tdTomato mice stained for collagen 1 or Pi16 (shown in grey). tdTomato is shown in red. DAPI is shown in blue. j,k, Image quantifications of the mean distance to collagen 1 or Pi16 from tdTomato or DAPI on the sections. n = 4 mice. l, Representative images of lung sections stained for collagen 1 (shown in grey) from ablation experiments using Cthrc1-CreER/Rosa26-DTA mice. DAPI is shown in blue. m, Image quantification of % collagen 1+ area on the sections. n = 3 (saline), 8 (bleomycin, Rosa26-WT/WT), or 10 (bleomycin, Rosa26-DTA/DTA) mice. Data are mean ± SEM. Data are representative of at least two independent experiments. Scale bars, 20 μm (g), 1 mm (i,l). Statistical analysis was performed using unpaired two-tailed t-test (j,k) or two-way ANOVA followed by Sidak’s multiple comparison test (m).

Source data

Extended Data Fig. 11 Tgfbr2 conditional knockout in alveolar fibroblasts abrogates fibrosis but exacerbates inflammation.

a, Representative images of whole sections stained for collagen 1 (magenta) and collagen 4 (green). Arrows indicate regions of intra-alveolar collagen 1. b, Survival after bleomycin treatment. n = 15 mice. c, qPCR of purified tdTomato+ cells from saline-treated mice showed no difference for fibrotic and inflammatory genes between control and Tgfbr2 cKO. d, Flow cytometric counting of myeloid populations in BAL from saline-treated mice showed no difference between control and Tgfbr2 cKO. n = 5 mice (b,c). e, Gating strategy for myeloid populations in BAL. f, Representative images of sections from bleomycin-treated lungs stained for Saa3 (magenta) and CD68 (green). tdTomato is shown in blue. Magnified single-channel images of yellow rectangles are shown on the right. g,h, Image quantification of Saa3 (g), or CD68 (h). n = 5 (control) or 6 (Tgfbr2 fl/fl) mice. Data are representative of at least two independent experiments. Data are mean ± SEM. Statistical analysis was performed using unpaired two-tailed t-test (g,h). Scale bars, 1 mm (a), 100 μm (f, wide), or 20 μm (f, magnified).

Source data

Supplementary information

Supplementary Table 1

Primer sequences used for qPCR.

Reporting Summary

Supplementary Video 1

3D reconstruction of z-stack images for a section from an uninjured Scube2-creER mouse. z-stack images of a thick section from an uninjured Scube2-creER mouse lung stained for proSP-C and nuclei were reconstructed in 3D. tdTomato is shown in red. proSP-C is shown in cyan. DAPI (nuclei) is shown in blue.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsukui, T., Wolters, P.J. & Sheppard, D. Alveolar fibroblast lineage orchestrates lung inflammation and fibrosis. Nature 631, 627–634 (2024). https://doi.org/10.1038/s41586-024-07660-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07660-1

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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