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:

ETS1 governs pathological tissue-remodeling programs in disease-associated fibroblasts

Abstract

Fibroblasts, the most abundant structural cells, exert homeostatic functions but also drive disease pathogenesis. Single-cell technologies have illuminated the shared characteristics of pathogenic fibroblasts in multiple diseases including autoimmune arthritis, cancer and inflammatory colitis. However, the molecular mechanisms underlying the disease-associated fibroblast phenotypes remain largely unclear. Here, we identify ETS1 as the key transcription factor governing the pathological tissue-remodeling programs in fibroblasts. In arthritis, ETS1 drives polarization toward tissue-destructive fibroblasts by orchestrating hitherto undescribed regulatory elements of the osteoclast differentiation factor receptor activator of nuclear factor-κB ligand (RANKL) as well as matrix metalloproteinases. Fibroblast-specific ETS1 deletion resulted in ameliorated bone and cartilage damage under arthritic conditions without affecting the inflammation level. Cross-tissue fibroblast single-cell data analyses and genetic loss-of-function experiments lent support to the notion that ETS1 defines the perturbation-specific fibroblasts shared among various disease settings. These findings provide a mechanistic basis for pathogenic fibroblast polarization and have important therapeutic implications.

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: A distal enhancer element regulates RANKL gene expression in arthritic synovial fibroblasts.
Fig. 2: E3 deletion ameliorates arthritis-induced bone damage.
Fig. 3: Transcriptional regulation of tissue-destructive genes by ETS1.
Fig. 4: Ets1 deletion in synovial fibroblasts attenuates arthritic joint damage.
Fig. 5: Transcriptome analysis of Ets1-deleted fibroblasts.
Fig. 6: Cross-tissue analysis of fibroblast single-cell datasets reveals the ETS1 expression is associated with perturbation-specific fibroblast phenotypes.
Fig. 7: Ets1-expressing fibroblasts contribute to tissue remodeling and healing upon epithelial injury.

Similar content being viewed by others

Data availability

All the data that support the plots within this paper are available in the main text or in the Supplementary Information. The scRNA-seq data and bulk RNA-seq data produced in this study are available in the public GEO database under accession codes GSE192504 and GSE201310. The referenced publicly available datasets were downloaded from the GEO, BioProject NCBI database, ENCODE Project database, TCGA database, National Bioscience Database Center (NBDC) database and MSigDB Collections C3 datasets: regulatory target gene sets (legacy transcription factor targets, https://www.gsea-msigdb.org/gsea/msigdb/index.jsp). scRNA-seq (dbGaP study accession phs001529.v1.p1, ImmPort SDY998, GSE178341, SCP259, GSE176078, GSE136103, GSE135893, GSE140023 and GSE172261), ChIP–seq (GSE128642, GSE148399, GSE54782, GSE123198, GSE145951 and GSE155882; ENCODE: ENCFF614VLL, ENCFF982OBR, ENCFF577JGN, ENCFF169ARY, ENCFF244ZNY, ENCFF569PTY, ENCFF523ZCW, ENCFF213MVM, ENCFF383CFQ and ENCFF314JXH), ATAC–seq (GSE128644 and BioProject PRJNA643827), Hi-C (NBDC hum0207), bulk RNA-seq (GSE129451 and GSE148395; NBDC hum0207; GSE166927 and GSE166925), TCGA clinical data and sequencing data (pancreatic adenocarcinoma (PanCancer Atlas), adult soft tissue sarcomas, mesothelioma (PanCancer Atlas), lung adenocarcinoma (Firehose Legacy) and colon cancer CPTAC-2 prospective and colorectal adenocarcinoma). Source data are provided with this paper.

Code availability

R scripts for data analysis used in this study are available at GitHub (https://github.com/MingluYAN).

References

  1. Virchow, R. Die cellularpathologie in ihrer begründung auf physiologische und pathologische gewebelehre. Zwanzig vorlesungen gehalten während der monate februar, märz und april 1858 im Pathologischen institute zu Berlin (A. Hirschwald, 1858).

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Davidson, S. et al. Fibroblasts as immune regulators in infection, inflammation and cancer. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-021-00540-z (2021).

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

    Article  PubMed  CAS  Google Scholar 

  5. Koliaraki, V., Prados, A., Armaka, M. & Kollias, G. The mesenchymal context in inflammation, immunity and cancer. Nat. Immunol. 21, 974–982 (2020).

    Article  PubMed  CAS  Google Scholar 

  6. Buckley, C. D. Fibroblast cells reveal their ancestry. Nature 593, 511–512 (2021).

    Article  PubMed  CAS  Google Scholar 

  7. Buckley, C. D. et al. Immune-mediated inflammation across disease boundaries: breaking down research silos. Nat. Immunol. 22, 1344–1348 (2021).

    Article  PubMed  CAS  Google Scholar 

  8. Danks, L. et al. RANKL expressed on synovial fibroblasts is primarily responsible for bone erosions during joint inflammation. Ann. Rheum. Dis. 75, 1187–1195 (2016).

    Article  PubMed  CAS  Google Scholar 

  9. Komatsu, N. et al. Plasma cells promote osteoclastogenesis and periarticular bone loss in autoimmune arthritis. J. Clin. Invest. https://doi.org/10.1172/JCI143060 (2021).

  10. Tsukasaki, M. & Takayanagi, H. Osteoimmunology: evolving concepts in bone–immune interactions in health and disease. Nat. Rev. Immunol. 19, 626–642 (2019).

    Article  PubMed  CAS  Google Scholar 

  11. Takayanagi, H. et al. T cell-mediated regulation of osteoclastogenesis by signalling cross-talk between RANKL and IFN-γ. Nature 408, 600–605 (2000).

    Article  PubMed  CAS  Google Scholar 

  12. Takayanagi, H. et al. Involvement of receptor activator of nuclear factor κB ligand/osteoclast differentiation factor in osteoclastogenesis from synoviocytes in rheumatoid arthritis. Arthritis Rheum. 43, 259–269 (2000).

    Article  PubMed  CAS  Google Scholar 

  13. Takayanagi, H. et al. A new mechanism of bone destruction in rheumatoid arthritis: synovial fibroblasts induce osteoclastogenesis. Biochem. Biophys. Res. Commun. 240, 279–286 (1997).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Stephenson, W. et al. Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation. Nat. Commun. 9, 791 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhang, F. et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 20, 928–942 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Bishop, K. A. et al. Transcriptional regulation of the human TNFSF11 gene in T cells via a cell type-selective set of distal enhancers. J. Cell. Biochem. 116, 320–330 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Onal, M. et al. Unique distal enhancers linked to the mouse Tnfsf11 gene direct tissue-specific and inflammation-induced expression of RANKL. Endocrinology 157, 482–496 (2016).

    Article  PubMed  CAS  Google Scholar 

  19. Loh, C. et al. TNF-induced inflammatory genes escape repression in fibroblast-like synoviocytes: transcriptomic and epigenomic analysis. Ann. Rheum. Dis. 78, 1205–1214 (2019).

    Article  PubMed  CAS  Google Scholar 

  20. Krishna, V. et al. Integration of the transcriptome and genome-wide landscape of BRD2 and BRD4 binding motifs identifies key superenhancer genes and reveals the mechanism of Bet inhibitor action in rheumatoid arthritis synovial fibroblasts. J. Immunol. 206, 422–431 (2021).

    Article  PubMed  CAS  Google Scholar 

  21. Andersson, R. & Sandelin, A. Determinants of enhancer and promoter activities of regulatory elements. Nat. Rev. Genet. 21, 71–87 (2020).

    Article  PubMed  CAS  Google Scholar 

  22. Kollias, G. et al. Animal models for arthritis: innovative tools for prevention and treatment. Ann. Rheum. Dis. 70, 1357–1362 (2011).

    Article  PubMed  Google Scholar 

  23. Alves, C. H., Farrell, E., Vis, M., Colin, E. M. & Lubberts, E. Animal models of bone loss in inflammatory arthritis: from cytokines in the bench to novel treatments for bone loss in the bedside—a comprehensive review. Clin. Rev. Allergy Immunol. 51, 27–47 (2016).

    Article  PubMed  CAS  Google Scholar 

  24. Tsuchiya, H. et al. Parsing multiomics landscape of activated synovial fibroblasts highlights drug targets linked to genetic risk of rheumatoid arthritis. Ann. Rheum. Dis. https://doi.org/10.1136/annrheumdis-2020-218189 (2020).

  25. Bartel, F. O., Higuchi, T. & Spyropoulos, D. D. Mouse models in the study of the Ets family of transcription factors. Oncogene 19, 6443–6454 (2000).

    Article  PubMed  CAS  Google Scholar 

  26. Laslo, P. et al. Multilineage transcriptional priming and determination of alternate hematopoietic cell fates. Cell 126, 755–766 (2006).

    Article  PubMed  CAS  Google Scholar 

  27. Okada, Y. et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506, 376–381 (2014).

    Article  PubMed  CAS  Google Scholar 

  28. Nakamura, Y. et al. Ets-1 regulates TNF-α-induced matrix metalloproteinase-9 and tenascin expression in primary bronchial fibroblasts. J. Immunol. 172, 1945–1952 (2004).

    Article  PubMed  CAS  Google Scholar 

  29. Redlich, K. et al. Overexpression of transcription factor Ets-1 in rheumatoid arthritis synovial membrane: regulation of expression and activation by interleukin-1 and tumor necrosis factor alpha. Arthritis Rheum. 44, 266–274 (2001).

    Article  PubMed  CAS  Google Scholar 

  30. Kim, C. J. et al. The transcription factor Ets1 suppresses T follicular helper type 2 cell differentiation to halt the onset of systemic lupus erythematosus. Immunity 50, 272 (2019).

    Article  PubMed  CAS  Google Scholar 

  31. Armaka, M. et al. Mesenchymal cell targeting by TNF as a common pathogenic principle in chronic inflammatory joint and intestinal diseases. J. Exp. Med. 205, 331–337 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Culemann, S. et al. Locally renewing resident synovial macrophages provide a protective barrier for the joint. Nature 572, 670–675 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Wei, K. et al. Notch signalling drives synovial fibroblast identity and arthritis pathology. Nature 582, 259–264 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Smillie, C. S. et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell 178, 714–730 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Pelka, K. et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell 184, 4734–4752 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Brügger, M. D., Valenta, T., Fazilaty, H., Hausmann, G. & Basler, K. Distinct populations of crypt-associated fibroblasts act as signaling hubs to control colon homeostasis. PLoS Biol. 18, e3001032 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Karpus, O. N. et al. Colonic CD90+ crypt fibroblasts secrete semaphorins to support epithelial growth. Cell Rep. 26, 3698–3708 (2019).

    Article  PubMed  CAS  Google Scholar 

  38. Wu, S. Z. et al. A single-cell and spatially resolved atlas of human breast cancers. Nat. Genet. 53, 1334–1347 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. 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 

  40. 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  PubMed  CAS  Google Scholar 

  41. Guinney, J. et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 21, 1350–1356 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Ellinghaus, D. et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat. Genet. 48, 510–518 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Jasso, G. J. et al. Colon stroma mediates an inflammation-driven fibroblastic response controlling matrix remodeling and healing. PLoS Biol. 20, e3001532 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Kinchen, J. et al. Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell 175, 372–386 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Nagashima, K. et al. Targeted deletion of RANKL in M cell inducer cells by the Col6a1-Cre driver. Biochem. Biophys. Res. Commun. 493, 437–443 (2017).

    Article  PubMed  CAS  Google Scholar 

  46. Singhal, R. & Shah, Y. M. Oxygen battle in the gut: hypoxia and hypoxia-inducible factors in metabolic and inflammatory responses in the intestine. J. Biol. Chem. 295, 10493–10505 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Ryu, J. H. et al. Hypoxia-inducible factor-2α is an essential catabolic regulator of inflammatory rheumatoid arthritis. PLoS Biol. 12, e1001881 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Wohlfahrt, T. et al. PU.1 controls fibroblast polarization and tissue fibrosis. Nature 566, 344–349 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Ramachandran, P. et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. 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  PubMed  PubMed Central  CAS  Google Scholar 

  51. Conway, B. R. et al. Kidney single-cell atlas reveals myeloid heterogeneity in progression and regression of kidney disease. J. Am. Soc. Nephrol. 31, 2833–2854 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Liu, X. et al. Identification of lineage-specific transcription factors that prevent activation of hepatic stellate cells and promote fibrosis resolution. Gastroenterology 158, 1728–1744 (2020).

    Article  PubMed  CAS  Google Scholar 

  53. Cabrera, S. et al. Delayed resolution of bleomycin-induced pulmonary fibrosis in absence of MMP13 (collagenase 3). Am. J. Physiol. Lung Cell. Mol. Physiol. 316, L961–L976 (2019).

  54. Cohen, S. B. et al. Denosumab treatment effects on structural damage, bone mineral density, and bone turnover in rheumatoid arthritis: a twelve-month, multicenter, randomized, double-blind, placebo-controlled, phase II clinical trial. Arthritis Rheum. 58, 1299–1309 (2008).

    Article  PubMed  CAS  Google Scholar 

  55. Takeuchi, T. et al. Effect of denosumab on Japanese patients with rheumatoid arthritis: a dose-response study of AMG 162 (denosumab) in patients with rheumatoId arthritis on methotrexate to validate inhibitory effect on bone Erosion (DRIVE)—a 12-month, multicentre, randomised, double-blind, placebo-controlled, phase II clinical trial. Ann. Rheum. Dis. 75, 983–990 (2016).

    Article  PubMed  CAS  Google Scholar 

  56. Krausgruber, T. et al. Structural cells are key regulators of organ-specific immune responses. Nature 583, 296–302 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Kitazawa, R., Mori, K., Yamaguchi, A., Kondo, T. & Kitazawa, S. Modulation of mouse RANKL gene expression by Runx2 and vitamin D3. J. Cell. Biochem. 105, 1289–1297 (2008).

    Article  PubMed  CAS  Google Scholar 

  58. Vasaikar, S. et al. Proteogenomic analysis of human colon cancer reveals new therapeutic opportunities. Cell 177, 1035–1049 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Khafipour, A. et al. Denosumab regulates gut microbiota composition and cytokines in dinitrobenzene sulfonic acid (DNBS)-experimental colitis. Front. Microbiol. 11, 1405 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Fukushima, K. et al. Dysregulated expression of the nuclear exosome targeting complex component Rbm7 in nonhematopoietic cells licenses the development of fibrosis. Immunity 52, 542–556 (2020).

    Article  PubMed  CAS  Google Scholar 

  61. Hayer, S. et al. ‘SMASH’ recommendations for standardised microscopic arthritis scoring of histological sections from inflammatory arthritis animal models. Ann. Rheum. Dis. 80, 714–726 (2021).

    Article  PubMed  CAS  Google Scholar 

  62. Tsukasaki, M. et al. Stepwise cell fate decision pathways during osteoclastogenesis at single-cell resolution. Nat. Metab. 2, 1382–1390 (2020).

    Article  PubMed  CAS  Google Scholar 

  63. DeTomaso, D. et al. Functional interpretation of single cell similarity maps. Nat. Commun. 10, 4376 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Amemiya, H.M., & Boyle, A.P. The ENCODE Blacklist: Identification of Problematic Regions of the Genome. Sci. Rep. 9, 9354 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Ovcharenko, I., Nobrega, M. A., Loots, G. G. & Stubbs, L. ECR Browser: a tool for visualizing and accessing data from comparisons of multiple vertebrate genomes. Nucleic Acids Res. 32, W280–W286 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Friedrich, M. et al. IL-1-driven stromal–neutrophil interactions define a subset of patients with inflammatory bowel disease that does not respond to therapies. Nat. Med. 27, 1970–1981 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Terashima, T. Asano, T. Sugita, H. Urabe, S. Yin, W. Yin, S. Zang, K. Kusubata, A. Suematsu, K. Kubo, Y. Omata, N. Kamiya, M. Hatakeyama, M. Miyoshi, G. Zhao, K. Ohmura, J. Nishio and K. Nakano for thoughtful discussion and valuable technical assistance. We appreciate the great contribution of K. Onimaru for the data analysis. This work was supported in part by the Japan Agency for Medical Research and Development (AMED) under grant number JP22ek0410073, AMED-CREST under grant number JP22gm1210008 and AMED-PRIME under grant number JP22gm6310029h0001; Japan Society for the Promotion of Science (JSPS): Grants-in-Aid for Specially Promoted Research (15H05703), Scientific Research S (21H05046), Scientific Research B (21H03104 and 22H02844) and Challenging Research (20K21515 and 21K18254). M.Y. was supported by a JSPS Research Fellowship for Young Scientists (19J21942) and a JSPS Postdoctoral Fellowships for Overseas Researchers (22F22108).

Author information

Authors and Affiliations

Authors

Contributions

M.Y. conceived the project and performed most of the experiments and data analyses, interpreted the results and wrote the manuscript. N.K., Y.O., H.I.S., H. Takaba, W.P. and T.N. provided advice on project planning and contributed to data interpretation and manuscript preparation. R.M. constructed the enhancer plasmids, performed reporter assay experiment and contributed to data interpretation. N.C.-N.H. and Y.T. performed computational analyses and contributed to data interpretation. R.K. and S.K. constructed and provided RANKL promoter luciferase plasmid and provided advice on project planning and data interpretation. Y.M., T.S. induced fibrosis model, performed the histological analysis and provided advice on data interpretation. T.O., S.-H.I., C.J.K. contributed to the generation of genetically modified mice and provided advice on data interpretation. G.K. generated and provided the Col6a1-Cre mice, advised on epigenomic data analysis and provided a thoughtful discussion on data interpretation. S.T. advised on project planning, provided the human synovium specimens and a thoughtful discussion on data interpretation. K.O. provided advice on project planning, immunostaining and histological analysis, data interpretation and manuscript preparation. M.T. supervised project planning and data interpretation and wrote the manuscript. H. Takayanagi directed the project and wrote the manuscript.

Corresponding author

Correspondence to Hiroshi Takayanagi.

Ethics declarations

Competing interests

The Department of Osteoimmunology is an endowed department supported by unrestricted grants from AYUMI Pharmaceutical, Chugai Pharmaceutical, MIKI HOUSE and Noevir. S.-H.I. is the CEO of the ImmunoBiome but declares no competing interests for this paper. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Christopher Buckley and Caroline Ospelt for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with the Nature Immunology team.

Additional information

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

Extended data

Extended Data Fig. 1 A distal enhancer regulates RANKL gene expression in arthritic SFs.

a, The expression pattern of representative cell-type marker genes in the CIA synovial cells (n = 2,160) in the UMAP visualization. The feature plots of the expression distribution of the major cell-type marker genes corresponding to the assigned cell types shown in (Fig. 1a). b, c, UMAP plots and unsupervised clustering of the synovial cells of individuals with RA (b) dbGaP Study Accession: phs001529.v1.p1, n = 19,599 cells; (c) ImmPort Accession: SDY998, n = 9,553 cells) colored by cluster. The violin plots on the right sides show the expression of the known SF marker genes. d, UMAP plot of the integrated SFs (n = 11,188 cells) from RA. Eight clusters (F1-F8) identified through unsupervised graph-based clustering are shown in color. Violin plots show the expression of selected genes in the SF sub-clusters. e, The sequence similarity of E1-E5 between humans and other vertebrates analyzed by the ECR Browser. The homologous sequence regions of E1-E5 in the mouse genome are indicated by arrows. f, Epigenomic analyses at the Tnfsf11 locus (mm9) in osteoblasts (vitamin D3 stimulation followed by H3K27ac ChIP–seq and vitamin D receptor (VDR) ChIP–seq (GSE54782)), in T cells (anti-CD3/CD28 antibody stimulation followed by H3K27ac ChIP–seq (GSE123198)), in B cells (IL-2/IL-4/IL-5/CD40L stimulation followed by H3K27ac ChIP–seq (GSE145951)), in cardiac fibroblasts (TGFβ stimulation followed by H3K27ac ChIP–seq (GSE155882)), in arthritic SFs (TNF stimulation followed by ATAC–seq, PRJNA643827)). g, h, The expression levels of Tnfsf11 mRNA in arthritic SFs (WT: control, n = 3; stimulation, n = 2; E3-KO: control: n = 2; stimulation, n = 2) and lymphocytes (WT: control, n = 2; stimulation, n = 3; E3-KO: control: n = 2; stimulation, n = 4). Data were expressed as mean ± s.e.m. P values were determined by two-way ANOVA analysis followed by Turkey’s post hoc test.

Source data

Extended Data Fig. 2 E3 deletion ameliorates arthritis-induced bone damage.

a, Flow cytometry gating strategies and a representative FACS plot of synovial cells from WT and E3-KO AIA (day 7 after intra-articular injection) mice, the number shows a frequency of indicated population from more than three independent experiments. b, Flow cytometry gating strategies and a representative FACS plot of synovial cells from WT and E3-KO STIA (day 10) mice (n = 6-10), data were expressed as mean ± s.e.m. c, Synovial cell number per inflamed joint (WT, n = 6; E3-KO, n = 8) and the frequencies of CD45+ cells (WT, n = 6; E3-KO, n = 8), endothelial cells (WT, n = 6; E3-KO, n = 9), mural cells (WT, n = 6; E3-KO, n = 10), SFs (WT, n = 6; E3-KO, n = 10). Data were expressed as mean ± s.e.m. P values were determined by two-tailed t-test.

Source data

Extended Data Fig. 3 ETS1 regulates the expression of tissue-destructive genes in SFs.

a, Motif enrichment analysis in RANKL enhancers by HOMER software (P values: hypergeometric test) b, The expression levels of ETS family members in RA patients-derived SFs under the indicated conditions (NBDC accession code hum0207). Data were expressed as mean ± s.e.m. P values were determined by one-way ANOVA analysis followed by Turkey’s post hoc test. c, d, The expression levels of ETS1 in RA patient-derived SF sub-clusters F1 to F8 (c) and CIA SFs sub-clusters mFib1 to mFib4 (d). e, Regulatory target gene sets analysis in CIA SFs sub-clusters using MSigDB C3 datasets (legacy transcription factor targets) performed with VISION R package (P values: Wilcoxon rank-sum test). f, Alignment of the ETS1 variants associated with RA. The upper panel shows the P-values of the 29 variants within the ETS1 region (Chr. 11: 127,828,656-128,957,453 in hg19) which exceeded the genome-wide significance threshold (P < 5 × 10-8) in a previously performed RA GWAS study27. The dashed line indicates the genome-wide significance threshold (P < 5 × 10-8). The lower panel (from top to bottom) shows the H3K27ac ChIP–seq (GSE128642) peaks detected in RA patient SFs, candidate cis-regulatory element (cCRE) with promoter-like signatures (cCRE_PLS), cCRE with proximal enhancer-like signatures (cCRE_pELS), cCRE with distal enhancer-like signatures (cCRE_dELS), cCRE enriched with DNase and H3K4me3 signals (cCRE_DNase_H3K4me3) and cCRE enriched with CTCF signals (cCRE_CTCF). Overlapping of the RA-associated ETS1 variants with the H3K27ac ChIP–seq (GSE128642) peaks detected in RA patient SFs and the candidate cCRE are indicated. The red-colored dots (ChIP–seq) and blue-colored dots (cCRE) represent overlaps. The shaded area indicates the overlap among the RA-associated ETS1 variants, and the H3K27ac peaks detected in RA patient SFs and cCRE.

Source data

Extended Data Fig. 4 ETS1 deletion in SFs attenuates arthritis-induced joint damage.

a, The expression of Col6a1 in the CIA synovial cells (n = 2,160) in the UMAP visualization. b, Violin plots showing the expression of Thy1 and Col6a1 in the SF sub-clusters. c, The expression levels of Ets1 mRNA in sorted SFs (Ets1flox, n = 8; Ets1ΔFib, n = 6), leukocytes (Ets1flox, n = 4; Ets1ΔFib, n = 3) and endothelial cells (Ets1flox, n = 6; Ets1ΔFib, n = 5) of the inflamed synovium. Data were expressed as mean ± s.e.m. P values were determined by two-tailed t-test. d, e, Parameters of the micro-CT analysis (bone volume per tissue volume, trabecular number, trabecular thickness and trabecular spacing) of the femurs of Ets1flox and Ets1ΔFib mice (female, n = 4 per group) at the age of 8 weeks under physiological conditions. Data were expressed as mean ± s.e.m. P values were determined by two-tailed t-test (d). Representative micro-CT images (female, 8 weeks). Micro-CT scale bars: 1 mm (e). f, g, Representative FACS plot and gating strategies of STIA (day 16) synovial cells from Ets1flox and Ets1ΔFib mice, n = 6-12 (f). Synovial cell number per inflamed joint and the frequencies of CD3+ cells (Ets1flox, n = 9; Ets1ΔFib, n = 6), and the frequencies of the other cell types in synovial tissue of Ets1flox mice (n = 12) and Ets1ΔFib STIA mice (n = 8). Data were expressed as mean ± s.e.m. P values were determined by two-tailed t-test (g).

Source data

Extended Data Fig. 5 ETS1+ SFs increased in number in the destruction phase during CIA.

a, Representative FACS plot of SFs derived from the mice of untreated control and CIA (inflammation phase: day 28; destruction phase: day 42 to day 50). b, Quantification of the cell number of ETS1+ fibroblasts of n = 5 mice in inflammation phase, n = 6 mice in destruction phase and NOTCH3 + fibroblasts of n = 6 mice in inflammation phase, n = 6 mice in destruction phase in the joints. Data were expressed as mean ± s.e.m. P values were determined by two-tailed t-test.

Source data

Extended Data Fig. 6 RNA-seq analysis of Ets1-deleted fibroblasts.

a, Representative FACS plot of SFs (STIA day 16) from Ets1flox and Ets1ΔFib mice, n = 3. b, Three-dimensional PCA analysis for the top 2500 variable genes in SFs derived from Ets1flox and Ets1ΔFib mice, n = 3. c, Heat map showing the DEGs between Ets1flox- and Ets1ΔFib-SFs. (DEGs: log2 FC > 0.5, P < 0.05 determined by DESeq2 (Wald test with Benjamini and Hochberg method correction)).

Extended Data Fig. 7 Cross-tissue analysis of fibroblast scRNA-seq datasets.

a, b, Mouse perturbed-state fibroblasts atlas4 and the expression of Ets1, Tnfsf11, Mmp13 and Mmp3 in the identified clusters (a) colored by Adamdec1 logcounts and in the fibroblast cells across diseases (b) (https://www.fibroxplorer.com/home). c, Dot plot showing the expression of ETS1, TNFSF11, MMP3 and MMP13 in ulcerative colitis (UC)-associated stromal cells and healthy controls (SCP259). d, Dot plot showing the expression of ETS1, TNFSF11, MMP3 and MMP13 in stromal cells of tumor tissues and adjacent normal colon tissues of CRC patients (GSE178341). e, Dot plot showing the expression of ETS1, TNFSF11, MMP3 and MMP13 across CRC-associated fibroblast clusters (GSE178341). f, Expression levels of TNFSF11, MMP13, MMP3 in TCGA datasets of CRC patients. Groups were split by the expression levels of ETS1 (top third ETS1high, n = 84; bottom third ETS1low, n = 84). Box plots (center: median; limits: upper 75th percentile, lower: 25th percentile); whiskers were drawn independently above and below the box and extended to the maximum and minimum values (1.5*interquartile range, points were outliers). P values were calculated by two-tailed Student’s t-test.

Source data

Extended Data Fig. 8 Ets1-expressing fibroblasts contribute to mucosal remodeling in colitis.

a, UMAP embedding of 34,197 mesenchymal cells derived from colon tissues (GSE172261) of DSS-fed mice colored by cluster. SMCs: smooth muscle cells, BECs: blood endothelial cells, LECs: lymphatic endothelial cells, ICCs: interstitial cells of Cajal, MSCs: mesenchymal stem cells. b, Dot plot showing the expression of representative cell-type marker genes for cell type annotation in (a). c, Representative immunofluorescence images of colons derived from Ets1flox and Ets1ΔFib mice after DSS administration (day 10), n = 3. Scale bar: 100 μm.

Extended Data Fig. 9 Hypoxia may contribute to the ETS1 regulation in fibroblasts.

a, Chromatin conformation and epigenomic analyses of SFs from RA patients at the ETS1 locus (hg19) by Hi-C and H3K4me3, H3K27ac and ATAC-seq (NBDC accession code hum0207 and GSE128644). A loop-like structure appears between the ETS1 promoter (open chromatin region with H3K4me3 signals) and enhancer region (open chromatin region with H3K27ac signals) under the condition (TNF stimulation) that could stimulate ETS1 expression in both synovial and intestinal fibroblasts (shown in (b)). The identified binding motif for hypoxia-inducible factor (HIF) (Jaspar, MA0259.1) was shown below. NS: no stimulation. b, The expression levels of ETS1 in SFs (NS, n = 28; TNF, n = 29) and intestinal fibroblasts (n = 6 per group) (NBDC accession code hum0207 and GSE166927). Data were expressed as mean ± s.e.m. P values were calculated by two-tailed t-test. c, The expression levels of EPAS1 (NS, n = 28; TNF, n = 29) and HIF1A in SFs (NS, n = 28; TNF, n = 29) (NBDC accession code hum020724). Data were expressed as mean ± s.e.m. P values were calculated by two-tailed t-test. d, Representative immunofluorescence images of RA synovium and inflamed colons, n = 2-3. Scale bars: 100 μm. e, Correlation analysis (Pearson r and two-tailed P value) between ETS1 expression and HIF2 (encoded by EPAS1) expression in arthritic SFs (n = 27) (GSE129451) and in tissues of colorectal cancer (CRC, n = 42), Crohn’s disease (CD, n = 34) and ulcerative colitis (UC, n = 13) patients (GSE166925).

Source data

Extended Data Fig. 10 ETS1 and PU.1 expression in fibroblasts of fibrosis and arthritis.

a, UMAP embedding of 6,898 cells derived from cirrhotic human livers (GSE136103) colored by cluster. b, Feature plot showing the expression of representative cell-type marker genes for cell type annotation in (a). c, d, The expression of representative genes (MYH11, RGS5, LUM, COL3A1, PDGFRA, COL1A1) (c) and ETS1 (colored by red), SPI1 (colored by blue) (d) in the identified mesenchymal cells (cluster 10 and cluster 13) of liver cirrhosis in the UMAP visualization. e, UMAP embedding of 119,438 cells derived from pulmonary fibrosis (GSE135893) colored by cell type. f, Feature plot showing the expression of representative cell-type marker genes for cell type annotation in (e). g, Unsupervised graph-based clustering of fibroblasts revealed four populations colored by cluster and the expression of selected genes in the UMAP visualization. h, The expression of ETS1 (colored by red) and SPI1 (colored by blue) in the fibroblasts of pulmonary fibrosis. i, j, A correlation between ETS1 and SPI1 expression in fibroblasts of liver cirrhosis (i) and pulmonary fibrosis (j). Each dot represents a single cell of the annotated clusters. k, UMAP embedding of 16,370 cells of unilateral-ureteric-obstruction (UUO)-induced kidney fibrosis (GSE140023) colored by cluster. l, Violin plots showing the expression of representative cell-type marker genes for cell type annotation in (k). m, Unsupervised graph-based clustering of fibroblasts revealed three populations colored by cluster and the expression of selected genes in the UMAP visualization. n, The expression of Ets1 (colored by red) and Spi1 (colored by blue) in the fibroblasts of kidney fibrosis. o, A correlation between Ets1 and Spi1 expression in fibroblasts of kidney fibrosis. Each dot represents a single cell of the annotated clusters. p, q, Representative H&E stained, Azan stained and immunofluorescence images of lung tissues of untreated, bleomycin-induced lung fibrosis model in mice (p) and RA synovium (q), n = 2-3. Scale bars: 100 μm.

Supplementary information

Reporting Summary

Supplementary Table 1

Sequences of oligonucleotides (sgRNAs, ssODNs and primers)

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Springer Nature or its licensor 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

Yan, M., Komatsu, N., Muro, R. et al. ETS1 governs pathological tissue-remodeling programs in disease-associated fibroblasts. Nat Immunol 23, 1330–1341 (2022). https://doi.org/10.1038/s41590-022-01285-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01285-0

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