Abstract
A hallmark of pulmonary fibrosis is the aberrant activation of lung fibroblasts into pathological fibroblasts that produce excessive extracellular matrix1,2,3. Thus, the identification of key regulators that promote the generation of pathological fibroblasts can inform the development of effective countermeasures against disease progression. Here we use two mouse models of pulmonary fibrosis to show that LEPR+ fibroblasts that arise during alveologenesis include SCUBE2+ alveolar fibroblasts as a major constituent. These alveolar fibroblasts in turn contribute substantially to CTHRC1+POSTN+ pathological fibroblasts. Genetic ablation of POSTN+ pathological fibroblasts attenuates fibrosis. Comprehensive analyses of scRNA-seq and scATAC-seq data reveal that RUNX2 is a key regulator of the expression of fibrotic genes. Consistently, conditional deletion of Runx2 with LeprcreERT2 or Scube2creERT2 reduces the generation of pathological fibroblasts, extracellular matrix deposition and pulmonary fibrosis. Therefore, LEPR+ cells that include SCUBE2+ alveolar fibroblasts are a key source of pathological fibroblasts, and targeting Runx2 provides a potential treatment option for pulmonary fibrosis.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 51 print issues and online access
$199.00 per year
only $3.90 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout





Similar content being viewed by others
Data availability
The scRNA-seq and scATAC-seq data generated in this study have been deposited into the GEO (accession numbers GSE229523, GSE276546 and GSE278419). The publicly available scRNA-seq data for mouse lung development (accession numbers GSE160876 and GSE165063), normal adult mouse lungs (accession numbers GSE132771, GSE201698 and GSE211713), bleomycin-challenged mouse lungs (accession numbers GSE131800, GSE132771, GSE183545 and GSE201698), silica-exposed mouse lungs (accession GSE184854), CCl4-treated mouse liver (accession number GSE171904), human patients with IPF (accession number GSE136831) and bulk RNA-seq data for human patients with IPF (accession numbers GSE124685 and GSE134692) were used for analyses. Source data are provided with this paper.
Code availability
The codes used in this study for scRNA-seq and scATAC-seq analyses are available from the corresponding authors upon request. No custom code was generated.
Change history
17 June 2025
A Correction to this paper has been published: https://doi.org/10.1038/s41586-025-09254-x
References
Noble, P. W., Barkauskas, C. E. & Jiang, D. Pulmonary fibrosis: patterns and perpetrators. J. Clin. Invest. 122, 2756–2762 (2012).
Lagares, D. et al. ADAM10-mediated ephrin-B2 shedding promotes myofibroblast activation and organ fibrosis. Nat. Med. 23, 1405–1415 (2017).
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).
Mayr, C. H. et al. Integrative analysis of cell state changes in lung fibrosis with peripheral protein biomarkers. EMBO Mol. Med. 13, e12871 (2021).
Kramann, R. et al. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 16, 51–66 (2015).
Xie, T. et al. Transcription factor TBX4 regulates myofibroblast accumulation and lung fibrosis. J. Clin. Invest. 126, 3063–3079 (2016).
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 (2017).
Zepp, J. A. et al. Distinct mesenchymal lineages and niches promote epithelial self-renewal and myofibrogenesis in the lung. Cell 170, 1134–1148 (2017).
Tsukui, T. et al. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat. Commun. 11, 1920 (2020).
Lee, J. H. et al. Anatomically and functionally distinct lung mesenchymal populations marked by Lgr5 and Lgr6. Cell 170, 1149–1163 (2017).
Nabhan, A. N., 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).
Abdollahi, A. et al. Inhibition of platelet-derived growth factor signaling attenuates pulmonary fibrosis. J. Exp. Med. 201, 925–935 (2005).
Frangogiannis, N. Transforming growth factor-β in tissue fibrosis. J. Exp. Med. 217, e20190103 (2020).
Boyd, D. F. et al. Exuberant fibroblast activity compromises lung function via ADAMTS4. Nature 587, 466–471 (2020).
Zhou, B. O., Yue, R., Murphy, M. M., Peyer, J. G. & Morrison, S. J. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell 15, 154–168 (2014).
Decker, M. et al. Leptin-receptor-expressing bone marrow stromal cells are myofibroblasts in primary myelofibrosis. Nat. Cell Biol. 19, 677–688 (2017).
Negretti, N. M. et al. A single-cell atlas of mouse lung development. Development 148, dev199512 (2021).
Curras-Alonso, S. et al. An interactive murine single-cell atlas of the lung responses to radiation injury. Nat. Commun. 14, 2445 (2023).
Saito, S. et al. Angiopoietin-like 4 is a critical regulator of fibroblasts during pulmonary fibrosis development. Am. J. Respir. Cell Mol. Biol. 69, 328–339 (2023).
Knudsen, L. et al. Alveolar micromechanics in bleomycin-induced lung injury. Am. J. Respir. Cell Mol. Biol. 59, 757–769 (2018).
Niethamer, T. K. et al. Defining the role of pulmonary endothelial cell heterogeneity in the response to acute lung injury. eLife 9, e53072 (2020).
Upagupta, C., Shimbori, C., Alsilmi, R. & Kolb, M. Matrix abnormalities in pulmonary fibrosis. Eur. Respir. Rev. 27, 180033 (2018).
Melms, J. C. et al. A molecular single-cell lung atlas of lethal COVID-19. Nature 595, 114–119 (2021).
Travaglini, K. J. et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature 587, 619–625 (2020).
Dahlgren, M. W. et al. Adventitial stromal cells define group 2 innate lymphoid cell tissue niches. Immunity 50, 707–722 (2019).
Tsukui, T., Wolters, P. J. & Sheppard, D. Alveolar fibroblast lineage orchestrates lung inflammation and fibrosis. Nature 631, 627–634 (2024).
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).
Hung, C. et al. Role of lung pericytes and resident fibroblasts in the pathogenesis of pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 188, 820–830 (2013).
Baek, S. H. et al. Single cell transcriptomic analysis reveals organ specific pericyte markers and identities. Front. Cardiovasc. Med. 9, 876591 (2022).
Kanisicak, O. et al. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat. Commun. 7, 12260 (2016).
Ogawa, T., Shichino, S., Ueha, S., Bando, K. & Matsushima, K. Profibrotic properties of C1q+ interstitial macrophages in silica-induced pulmonary fibrosis in mice. Biochem. Biophys. Res. Commun. 599, 113–119 (2022).
Nguyen, H. C. B., Adlanmerini, M., Hauck, A. K. & Lazar, M. A. Dichotomous engagement of HDAC3 activity governs inflammatory responses. Nature 584, 286–290 (2020).
Xie, Z. et al. Gene set knowledge discovery with Enrichr. Curr. Protoc. 1, e90 (2021).
Vierstra, J. et al. Global reference mapping of human transcription factor footprints. Nature 583, 729–736 (2020).
Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).
Geng, Y. et al. PEAR1 regulates expansion of activated fibroblasts and deposition of extracellular matrix in pulmonary fibrosis. Nat. Commun. 13, 7114 (2022).
Parimon, T. et al. Syndecan-1 promotes lung fibrosis by regulating epithelial reprogramming through extracellular vesicles. JCI Insight 5, e129359 (2019).
Yang, W. et al. Single-cell transcriptomic analysis reveals a hepatic stellate cell-activation roadmap and myofibroblast origin during liver fibrosis in mice. Hepatology 74, 2774–2790 (2021).
Misharin, A. V. et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J. Exp. Med. 214, 2387–2404 (2017).
Leslie, J. et al. FPR-1 is an important regulator of neutrophil recruitment and a tissue-specific driver of pulmonary fibrosis. JCI Insight 5, e125937 (2020).
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).
McDonough, J. E. et al. Transcriptional regulatory model of fibrosis progression in the human lung. JCI Insight 4, e131597 (2019).
Sivakumar, P. et al. RNA sequencing of transplant-stage idiopathic pulmonary fibrosis lung reveals unique pathway regulation. ERJ Open Res. 5, 00117–02019 (2019).
Naik, P. K. et al. Periostin promotes fibrosis and predicts progression in patients with idiopathic pulmonary fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 303, L1046–L1056 (2012).
Uchida, M. et al. Periostin, a matricellular protein, plays a role in the induction of chemokines in pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 46, 677–686 (2012).
Seo, H. S. & Serra, R. Tgfbr2 is required for development of the skull vault. Dev. Biol. 334, 481–490 (2009).
Wu, M., Chen, G. & Li, Y. P. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res. 4, 16009 (2016).
Raaz, U. et al. Transcription factor Runx2 promotes aortic fibrosis and stiffness in type 2 diabetes mellitus. Circ. Res. 117, 513–524 (2015).
Mummler, C. et al. Cell-specific expression of runt-related transcription factor 2 contributes to pulmonary fibrosis. FASEB J. 32, 703–716 (2018).
Wohlfahrt, T. et al. PU.1 controls fibroblast polarization and tissue fibrosis. Nature 566, 344–349 (2019).
DeFalco, J. et al. Virus-assisted mapping of neural inputs to a feeding center in the hypothalamus. Science 291, 2608–2613 (2001).
Yata, Y. et al. DNase I-hypersensitive sites enhance α1(I) collagen gene expression in hepatic stellate cells. Hepatology 37, 267–276 (2003).
Takarada, T. et al. An analysis of skeletal development in osteoblast-specific and chondrocyte-specific runt-related transcription factor-2 (Runx2) knockout mice. J. Bone Miner. Res. 28, 2064–2069 (2013).
Fang, Y. et al. Epithelial Wntless regulates postnatal alveologenesis. Development 149, dev199505 (2022).
Fang, Y. et al. Follistatin like-1 aggravates silica-induced mouse lung injury. Sci. Rep. 7, 399 (2017).
Xu, C. et al. Hepatic neddylation deficiency triggers fatal liver injury via inducing NF-κB-inducing kinase in mice. Nat. Commun. 13, 7782 (2022).
Jiang, M. et al. Transitional basal cells at the squamous–columnar junction generate Barrett’s oesophagus. Nature 550, 529–533 (2017).
Jiang, M. et al. VEGF receptor 2 (KDR) protects airways from mucus metaplasia through a Sox9-dependent pathway. Dev. Cell 56, 1646–1660 (2021).
Janssen, H. et al. Monocytes re-enter the bone marrow during fasting and alter the host response to infection. Immunity 56, 783–796 (2023).
Shao, Z. et al. DNA-PKcs has KU-dependent function in rRNA processing and haematopoiesis. Nature 579, 291–296 (2020).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).
Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).
Qiu, X. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).
Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).
Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).
Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).
Wang, A. et al. Single-cell multiomic profiling of human lungs reveals cell-type-specific and age-dynamic control of SARS-CoV2 host genes. eLife 9, e62522 (2020).
Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).
Liu, X. et al. HER2 drives lung fibrosis by activating a metastatic cancer signature in invasive lung fibroblasts. J. Exp. Med. 219, e20220126 (2022).
Konkimalla, A. et al. Transitional cell states sculpt tissue topology during lung regeneration. Cell Stem Cell 30, 1486–1502 (2023).
Acknowledgements
We thank the colleagues in the Que Laboratory for their critical input to the study. Runx2flox/flox mouse strain (acc.no. CDB0832K) was kindly obtained from RIKEN BioResource Research Center (www2.clst.riken.jp/arg/mutant%20mice%20list.html). This work is partly supported by NIH grants R01HL152293, R01HL159675 and Department of Defense grant W81XWH2110196 (to J.Q.), NIH grants R01HL172990 and P01HL108793 (to D.J.), American Heart Association award 24CDA1268568 and the Pulmonary Fibrosis Foundation Scholars Program 1272558 (to X.L.). Flow cytometry was performed at the Columbia Center for Translational Immunology (CCTI) Flow Cytometry Core at Columbia University Medical Center, supported in part by the Office of the Director, National Institutes of Health under the awards S10RR027050 and S10OD020056. The CCHD microscopy core is supported by S10 OD032447 from the NIH. This research was also funded in part through the NIH/NCI Cancer Center Support Grant P30CA013696 and used the Genetically Modified Mouse Models/GMMMSR Core and the Genomics and High Throughput Screening Shared Resource.
Author information
Authors and Affiliations
Contributions
Y.F. and J.Q. designed experiments, analysed data and wrote the manuscript. Y.F., C.-S.L. and J.Q. generated the LeprcreERT2 and Higd1bcreERT2 knock-in mouse lines. Y.F. performed immunostaining, imaging, flow cytometry and mouse genetics. S.S.W.C. performed immunostaining, imaging and mouse genetics. Y.F. and C.X. performed scRNA-seq analyses. L.X., R.L. and Y.K. performed scATAC-seq analyses. K.Y., Y.H., X.L., D.J., T. Takarada, T. Tsukui and D.S. provided materials. A.S. and H.H. oversaw and performed tissue collection. X.S. and J.Q. supervised this work. All authors reviewed and approved the final manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature thanks Christopher Buckley 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 Lepr+ mesenchymal cells in the developing and adult mouse lung.
a-b, UMAP plot showing the different cell populations (a) in the lungs collected at E18.5, P0, P3, P7 and P14 (b). Re-analysis of the datasets GSE160876 and GSE165063. c, UMAP plot showing expression of Lepr in the developing lungs. d, UMAP plot showing the integration of lung mesenchymal cells at E18.5, P0, P3, P7 and P14. e, Dot plot showing the representative markers for each mesenchymal cell population. f, tdTomato (tdT) expression in the lungs of Lepr-Cre;R26tdT mice at different postnatal stages. P: Postnatal. g, tdT and α-SMA expression in the lungs of Lepr-Cre;R26tdT mice at different postnatal stages. The arrows indicate rare tdT+ smooth muscle cells. aw: airway, bv: blood vessel. h, Schematic depicting the generation of Lepr-CreERT2 mouse strain. i-j, Schematic diagram for tamoxifen injection and the representative immunostaining image of tdT in the lungs of Lepr-CreERT2;R26tdT mice at P14 (i) and P20 (j). k, Schematic depicting the use of tamoxifen-containing chow to feed Lepr-CreERT2;R26tdT mice and representative immunostaining images of surfactant protein C (SPC) and tdT in lung tissues. l, UMAP plot showing different mesenchymal populations from the adult control lungs that integrate the datasets GSE132771, GSE201698 and GSE211713. m, Dot plot showing the representative markers for each mesenchymal population. n-o, UMAP plot showing expression level of Lepr (n) and Scube2 (o). p, Blended feature plots showing co-expression of Lepr and Scube2. q, Representative images of tdT immunostaining and Scube2 RNA in situ hybridization. r-s, Quantification analysis showing the percentage of Scube2+ tdT+ cells in total tdT+ cells (r, n = 5) and in Scube2+ alveolar fibroblasts (s, n = 5). Data are representative of at least three independent experiments. Data are mean ± SEM. Scale bars: 100 µm (20 µm in magnified views).
Extended Data Fig. 2 Lepr-Cre labeled cells contribute to pathological fibroblasts.
a, Representative images of migrated tdT+EGFP+ and tdT-EGFP+ lung mesenchymal cells triggered by PDGF-BB. b, Quantification of the migrated cells (DAPI+) in a (tdT+EGFP+: n = 3, tdT-EGFP+: n = 3). c, Relative expression of matrix genes in tdT+EGFP+ and tdT-EGFP+ lung mesenchyme (RT-qPCR) (tdT+EGFP+: n = 3, tdT-EGFP+: n = 3). d, Expression of Acta2, Col1a1 and Ccn2 in tdT+EGFP+ and tdT-EGFP+ lung mesenchymal cells treated with/without TGF-β1 (RT-qPCR) (n = 3 for each group). e, tdT and EGFP expression in the lungs of Lepr-Cre;R26tdT;Col1a1-EGFP mice challenged with saline or bleomycin. f, Quantification of tdT+ cells (Saline: n = 6, Bleomycin: n = 6). g-h, Quantification of the proportion of α-SMA+tdT+ cells among α-SMA+ cells (g) and among tdT+ cells (h) (Saline: n = 6, Bleomycin: n = 6). i, Gating strategy to identify α-SMA+ tdT+ cells in live mesenchymal cells. j, FACS analysis of tdT+ and α-SMA+ cells isolated from the lung of Lepr-Cre;R26tdT mice treated with saline or bleomycin. k, Flow cytometric quantification for the percentage of α-SMA+ tdT+ cells in the lungs of Lepr-Cre;R26tdT mice treated with saline or bleomycin (Saline: n = 3, Bleomycin: n = 4). l, Experimental schematic and representative H&E images. m, α-SMA+ tdT+ cells in the lungs of Lepr-Cre;R26tdT mice exposed to saline or silica. n, Quantification of α-SMA+ tdT+ cells (Saline: n = 6, Silica: n = 6). o, Experimental schematic and representative H&E images. p, α-SMA and tdT expression in lung tissues. q, Quantification of α-SMA+ tdT+ cells in the lungs of Lepr-CreERT2;R26tdT mice following silica exposure (Saline: n = 6, Silica: n = 6). Data are representative of at least three independent experiments. Data are mean ± SEM. Statistical analysis was performed using unpaired two-tailed t-test with Welch’s correction (b, c, f, g, h, k, n, q) and two-way analysis of variance with multiple comparisons test with Sidak’s correction (d). Scale bars: l, o: 200 µm, a, e, m, p: 100 µm (e, m, p: 20 µm in magnified views).
Extended Data Fig. 3 scRNA-seq reveals the contribution of Lepr-Cre labeled alveolar fibroblasts to pathological fibroblasts.
a, Schematic diagram for scRNA-seq experimental design. b, UMAP plot showing the transcript levels of tdTomato. c, UMAP plot showing different cell populations. d, Red dots showing the cells from saline-treated lungs and blue dots showing the cells from bleomycin-treated lungs. e, Dot plot showing the representative markers for each population. f, UMAP plot showing four different fibroblast populations. g, Red and blue dots showing the cells from saline- and bleomycin-treated lungs, respectively. h, Dot plot showing the representative markers for each fibroblast population. i, Frequency of each fibroblast population in saline and bleomycin-treated lungs. j, UMAP plot showing the score of alveolar fibroblast signature. k-l, UMAP plot showing the expression level of the alveolar fibroblast markers Npnt (k) and Scube2 (l). m, UMAP plot showing the adventitial fibroblast signature score. n, UMAP plot showing the expression level of the adventitial fibroblast marker Pi16. o, Representative immunostaining image of Pi16 and tdT in the adventitial cuffs. aw: airway. p, UMAP plot showing the pathological fibroblast signature score. q, UMAP plot showing the expression level of the pathological fibroblast marker Cthrc1. r, Representative immunostaining image of Postn and tdT in the lungs of Lepr-Cre;R26tdT mice treated with bleomycin. s, UMAP plot showing pseudotime analysis with Monocle3. t-u, Trajectory analysis with Monocle2 showing cells ordered by pseudotime (t) and cell type (u). v, UMAP plot showing the expression of Lepr in the mesenchymal cells of the lungs isolated from Lepr-Cre;R26tdT mice challenged with saline or bleomycin. Data are representative of at least three independent experiments. Scale bars: o and r: 100 µm (o: 20 µm in magnified views).
Extended Data Fig. 4 scRNA-seq analysis reveals the heterogeneous lung fibroblasts labeled by Lepr-CreERT2.
a, Gating strategy to sort tdT+ lung mesenchymal cells (CD45-EpCAM-CD31-) from the lungs of Lepr-CreERT2;R26tdT mice treated with bleomycin or saline for scRNA-seq. b, tdTomato transcript levels are shown by UMAP plot. c, UMAP plot showing multiple Lepr-CreERT2 labeled cell populations. d, Red and blue dots showing the cells from saline- and bleomycin-treated lungs, respectively. e, Dot plot showing the representative markers for each population. f, Violin plot showing the significantly increased fibrosis-associated genes upon bleomycin challenge. g, 3D UMAP plot showing four different Lepr-CreERT2 labeled fibroblast populations. h-i, UMAP plot showing the expression level of the alveolar fibroblast markers Npnt (h) and Inmt (i). j, UMAP plot showing the adventitial fibroblast signature score. k-l, UMAP plot showing the expression level of the adventitial fibroblast markers Dcn (k) and Pi16 (l). m, Schematic depicting for the use of tamoxifen (Tmx)-containing chow to feed Lepr-CreERT2;R26tdT mice and representative immunostaining image of Pi16 and tdT in the adventitial cuffs. aw: airway. bv: blood vessel. n, Schematic depicting for the use of Tmx-containing chow and bleomycin challenge of Lepr-CreERT2;R26tdT mice, and representative images of Postn+ tdT+ pathological fibroblasts in lung tissues. Data are representative of at least three independent experiments. Statistical analysis was performed using two-sided Wilcoxon Rank Sum test (f). Scale bars: m and n: 100 µm (20 µm in magnified views).
Extended Data Fig. 5 Lepr-CreERT2 labeled alveolar fibroblasts generate pathological fibroblasts.
a, The cell distribution of Lepr-CreERT2 labeled fibroblast clusters along with the color-coded pseudotime (upper panel). Heatmap showing dynamic gene expression in fibroblast clusters (lower panel). Representative genes (left) and enriched pathways (right) in alveolar fibroblasts and pathological fibroblasts are shown, respectively. b, The expression levels of the alveolar fibroblast makers Scube2, Npnt, Inmt and the pathological fibroblast markers Cthrc1, Postn along pseudotime analysis. c-d, The top ten GO terms (c) and KEGG pathways (d) associated with genes enriched in pathological fibroblasts. e, UMAP plot showing the enrichment of Tgfβ1 transcript in pathological fibroblasts. f, UMAP plot showing the expression of Lepr transcript in lung mesenchymal cells of Lepr-CreERT2;R26tdT mice challenged with saline or bleomycin.
Extended Data Fig. 6 A novel Higd1b-CreERT2 mouse strain specifically labels pericytes which rarely contribute to pathological fibroblasts during lung fibrosis.
a, UMAP plot showing different mesenchymal populations from the adult control lungs that integrate the datasets GSE132771, GSE201698 and GSE211713. b-c, UMAP plot showing the expression levels of Higd1b (b) and Cox4i2 (c) in pericytes. Of note, 88% pericytes expressed Higd1b. d, Schematic depicting the generation of Higd1b-CreERT2 mouse strain. e, Schematic diagram for Tmx injection of adult Higd1b-CreERT2;R26-tdT mice and representative images showing immunostaining of pericyte marker NG2 and tdT. f, Schematic diagram for Tmx injection and bleomycin challenge of adult Higd1b-CreERT2;R26-tdT mice and representative images showing immunostaining of α-SMA, ERG and tdT. g, Representative images of two examples showing immunostaining of Cthrc1 and tdT in the lungs of Higd1b-CreERT2;R26-tdT mice challenged with bleomycin. h, Quantification analysis showing the rare contribution of pericytes to Cthrc1+ pathological fibroblasts in the lungs of Higd1b-CreERT2;R26-tdT mice (n = 6). Data are representative of at least three independent experiments. Data are mean ± SEM. i-j, Representative images showing immunostaining of tdT, Collagen I (i) and Ki67 (j) in the lungs of Higd1b-CreERT2;R26-tdT mice challenged with bleomycin. Scale bars: 100 µm (20 µm in magnified views).
Extended Data Fig. 7 Postn+ pathological fibroblasts are identified in silica-challenged lungs.
a, UMAP plot showing four different lung fibroblast populations in silica-treated lungs. The results were generated by re-analysis of the dataset GSE184854. b, Dot plot showing the representative markers for each fibroblast population. c-d, Postn expression shown by UMAP plot (c) and Violin Plot (d). e, Schematic diagram for Tmx injection and silica exposure of Postn-CreER;R26tdT mice and representative images showing immunostaining of α-SMA and tdT in lung tissues. f, Quantification analysis showing increased tdT+ cells upon silica exposure (Saline: n = 6, Silica: n = 6). g, Representative images showing immunostaining of α-SMA, ERG and Endomucin in lung tissues. Data are representative of at least three independent experiments. Data are mean ± SEM. Statistical analysis was performed using unpaired two-tailed t-test with Welch’s correction. Scale bars: 100 µm (20 µm in magnified views).
Extended Data Fig. 8 The TF-binding motif enrichment identified by scATAC-seq.
a, UMAP plot showing the expression of Runx2 transcripts in lung mesenchymal cells from Lepr-Cre;R26tdT mice challenged with saline or bleomycin. b, Violin plot showing the increased expression of Runx2 in the lungs of Lepr-CreERT2;R26tdT mice challenged with bleomycin. c, Violin plot showing the enrichment of Runx2 in pathological fibroblasts and proliferating fibroblasts of bleomycin-treated Lepr-CreERT2;R26tdT mouse lungs. d, Schematic depicting the experimental design for scATAC-seq. e, UMAP plot of scATAC-seq showing various lung mesenchymal populations. f, Blue and red dots showing the cells from saline- and bleomycin-treated lungs, respectively. g, Heatmap showing the representative markers for each lung mesenchymal population. h-i, UMAP plot showing various lung mesenchymal populations (h) from bleomycin-treated (blue dots) and control (red dots) lungs (i) that integrate the published scRNA-seq datasets GSE131800, GSE132771, GSE183545 and GSE201698. j, Dot plot showing the representative markers for each fibroblast population. k-l, UMAP plot (k) and Violin Plot (l) showing the enrichment of Runx2 transcripts in pathological fibroblasts. m, RT-qPCR analysis of Runx2 transcripts in FACs-sorted tdT+ cells isolated from the lungs of Lepr-Cre;R26tdT mice treated with bleomycin or saline (Saline: n = 3, Bleomycin: n = 3). n-o, UMAP plot showing the quiescent and active hepatic stellate cells (n) of control (red dots) and CCl4-treated (blue dots) mice (o). The dataset GSE171904 was used for re-analysis. HSC: Hepatic stellate cell. p-q. UMAP plot showing the expression of Cthrc1 (p) and Runx2 (q) in hepatic stellate cells. Data are representative of at least three independent experiments. Data are mean ± SEM. Statistical analysis was performed using two-sided Wilcoxon Rank Sum test (b) and unpaired two-tailed t-test with Welch’s correction (m).
Extended Data Fig. 9 Conditional deletion of Runx2 attenuates bleomycin-induced lung fibrosis.
a,b, FACS analysis to quantify bone marrow monocytes among CD45+ cells (Lepr-Cre;Runx2+/+: n = 3, Lepr-Cre;Runx2f/f: n = 3). c,d, FACS analysis to quantify bone marrow neutrophils among CD45+ cells (Lepr-Cre;Runx2+/+: n = 3, Lepr-Cre;Runx2f/f: n = 3). e, Representative H&E staining images of untreated lungs from Lepr-Cre;Runx2+/+ and Lepr-Cre;Runx2f/f mice. f, Experimental schematic and representative H&E staining images of the lungs from Lepr-Cre;Runx2+/+ and Lepr-Cre;Runx2f/f mice after bleomycin challenge. g, Quantification of fibrotic areas in the lungs of Lepr-Cre;Runx2+/+ (n = 9) and Lepr-Cre;Runx2f/f mice (n = 9). h, Hydroxyproline content in the lungs of Lepr-Cre;Runx2+/+ (n = 7) and Lepr-Cre;Runx2f/f mice (n = 7) treated with bleomycin. i, α-SMA and SPC expression in the lungs of Lepr-Cre;Runx2+/+ and Lepr-Cre;Runx2f/f mice after bleomycin challenge. j, Quantification of α-SMA+ cells in the lungs of Lepr-Cre;Runx2+/+ (n = 6) and Lepr-Cre;Runx2f/f mice (n = 6). k, Experimental schematic and representative Picro-Sirius Red staining images. l, α-SMA and tdT expression in the lungs of Lepr-CreERT2;Runx2+/+;R26tdT and Lepr-CreERT2;Runx2f/f;R26tdT mice. m, Quantification of α-SMA+ cells in the lungs of Lepr-CreERT2;Runx2+/+;R26tdT (n = 6) and Lepr-CreERT2;Runx2f/f;R26tdT mice (n = 6). n, Experimental schematic and representative Picro-Sirius Red staining images. o, α-SMA and tdT expression in the lungs of Scube2-CreERT2;Runx2+/+;R26tdT and Scube2-CreERT2;Runx2f/f;R26tdT mice. p, Quantification of α-SMA+ cells in the lungs of Scube2-CreERT2;Runx2+/+;R26tdT (n = 6) and Scube2-CreERT2;Runx2f/f;R26tdT mice (n = 6). Data are representative of at least three independent experiments. Data are mean ± SEM. Statistical analysis was performed using unpaired two-tailed t-test with Welch’s correction. Scale bars: e, f, k and n:1 mm (e and f: 200 µm in magnified views, k and n: 100 µm in magnified views), i, l and o: 100 µm (20 µm in magnified views).
Extended Data Fig. 10 Increased expression of RUNX2 in IPF lungs.
a, Separated UMAP plots showing fibroblasts from normal human lungs and IPF lungs. These results were generated through the re-analysis of the dataset GSE136831. b, Dot plot of scRNA-seq showing the representative markers for each fibroblast population. c-f, UMAP plot showing the expression levels of LEPR (c), SCUBE2 (d), COL1A1 (e) and ACTA2 (f). g, Violin plot of scRNA-seq showing the expression levels of ECM associated genes. h, Venn diagram showing the common and differential genes expressed in pathological fibroblasts of human IPF and bleomycin-treated mouse lungs. i, Increased transcript levels of RUNX2 in IPF lungs revealed by re-analyzing the bulk RNA seq dataset GSE134692 (Normal: n = 26, IPF: n = 46). Data are mean ± SEM. Statistical analysis was performed using unpaired two-tailed t-test with Welch’s correction.
Supplementary information
Supplementary Fig. 1
Gating strategy used to identify CD45+Ly6G–CD11b+Ly6C+ monocytes and CD45+Ly6G–CD11b+Ly6C+ neutrophils in mouse bone marrow.
Supplementary Table 1
Top 100 differentially expressed genes in Leprcre-labelled lung fibroblast populations.
Supplementary Table 2
Top 100 differentially expressed genes in LeprcreERT2-labelled lung fibroblast populations.
Supplementary Table 3
Marker genes of pathological fibroblast in human IPF and mouse bleomycin-treated lungs.
Supplementary Table 4
Primers used for RT–qPCR.
Supplementary Table 5
Oligonucleotide sequences of siRNA.
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.
About this article
Cite this article
Fang, Y., Chung, S.S.W., Xu, L. et al. RUNX2 promotes fibrosis via an alveolar-to-pathological fibroblast transition. Nature 640, 221–230 (2025). https://doi.org/10.1038/s41586-024-08542-2
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41586-024-08542-2
This article is cited by
-
COL10A1+ fibroblasts promote colorectal cancer metastasis and M2 macrophage polarization with pan-cancer relevance
Journal of Experimental & Clinical Cancer Research (2025)
-
HBV activates hepatic stellate cells through RUNX2/ITGBL1 axis
Virology Journal (2025)
-
Pulmonary fibrosis: from mechanisms to therapies
Journal of Translational Medicine (2025)
-
Fibroblast activation and heterogeneity in fibrotic disease
Nature Reviews Nephrology (2025)