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:

Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage

Abstract

Tissue fibrosis is a major cause of mortality that results from the deposition of matrix proteins by an activated mesenchyme. Macrophages accumulate in fibrosis, but the role of specific subgroups in supporting fibrogenesis has not been investigated in vivo. Here, we used single-cell RNA sequencing (scRNA-seq) to characterize the heterogeneity of macrophages in bleomycin-induced lung fibrosis in mice. A novel computational framework for the annotation of scRNA-seq by reference to bulk transcriptomes (SingleR) enabled the subclustering of macrophages and revealed a disease-associated subgroup with a transitional gene expression profile intermediate between monocyte-derived and alveolar macrophages. These CX3CR1+SiglecF+ transitional macrophages localized to the fibrotic niche and had a profibrotic effect in vivo. Human orthologs of genes expressed by the transitional macrophages were upregulated in samples from patients with idiopathic pulmonary fibrosis. Thus, we have identified a pathological subgroup of transitional macrophages that are required for the fibrotic response to injury.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Reference-based annotation of scRNA-seq.
Fig. 2: Reference data sets reveal heterogeneity of macrophage subtypes in lung fibrosis.
Fig. 3: Hierarchical clustering by reference annotation reveals a transitional macrophage in fibrosis.
Fig. 4: Markers of macrophages identified by scRNA-seq are found in multiple fibrosis models.
Fig. 5: Transitional macrophages expressing CX3CR1, MHCII, and SiglecF localize to sites of fibroblast accumulation after lung injury.
Fig. 6: Pdgf-aa is a macrophage-derived trophic factor for fibroblasts.
Fig. 7: Cx3cr1-derived macrophages drive fibroblast accumulation and fibrosis after lung injury.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. RNA-seq data have been deposited in the GEO repository under accession numbers GSE111664, GSE111690, and GSE114005 and at ArrayExpress under accession number E-MTAB-7142.

References

  1. Rockey, D. C., Bell, P. D. & Hill, J. A. Fibrosis—a common pathway to organ injury and failure. N. Engl. J. Med. 373, 96 (2015).

    Article  Google Scholar 

  2. Blackwell, T. S. et al. Future directions in idiopathic pulmonary fibrosis research. An NHLBI workshop report. Am. J. Respir. Crit. Care Med. 189, 214–222 (2014).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. 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.e263 (2017).

    Article  CAS  Google Scholar 

  5. 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).

    Article  CAS  Google Scholar 

  6. 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  Google Scholar 

  7. Schneider, C. et al. Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat. Immunol. 15, 1026–1037 (2014).

    Article  CAS  Google Scholar 

  8. Tan, S. Y. & Krasnow, M. A. Developmental origin of lung macrophage diversity. Development 143, 1318–1327 (2016).

    Article  CAS  Google Scholar 

  9. Gibbons, M. A. et al. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am. J. Respir. Crit. Care Med. 184, 569–581 (2011).

    Article  CAS  Google Scholar 

  10. 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).

    Article  CAS  Google Scholar 

  11. Satoh, T. et al. Identification of an atypical monocyte and committed progenitor involved in fibrosis. Nature 541, 96–101 (2017).

    Article  CAS  Google Scholar 

  12. Heng, T. S. & Painter, M. W. The Immunological Genome Project Consortium. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  Google Scholar 

  13. The ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  Google Scholar 

  14. Stunnenberg, H. G. International Human Epigenome Consortium & Hirst, M. The International Human Epigenome Consortium: a blueprint for scientific collaboration and discovery. Cell 167, 1145–1149 (2016).

  15. SingleR GitHub repository, https://github.com/dviraran/SingleR.

  16. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17, 77 (2016).

    Article  Google Scholar 

  17. Amit, I. et al. Unbiased reconstruction of a mammalian transcriptional network mediating pathogen responses. Science 326, 257–263 (2009).

    Article  CAS  Google Scholar 

  18. Shalek, A. K. et al. Single-cell RNA-seq reveals dynamic paracrine control of cellular variation. Nature 510, 363–369 (2014).

    Article  CAS  Google Scholar 

  19. Helft, J. et al. GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c(+)MHCII(+) macrophages and dendritic cells. Immunity 42, 1197–1211 (2015).

    Article  CAS  Google Scholar 

  20. Guilliams, M. & Malissen, B. A death notice for in-vitro-generated GM-CSF dendritic cells? Immunity 42, 988–990 (2015).

    Article  CAS  Google Scholar 

  21. Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  Google Scholar 

  22. Kang, H. M. et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat. Biotechnol. 36, 89–94 (2018).

    Article  CAS  Google Scholar 

  23. Li, H. et al. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat. Genet. 49, 708–718 (2017).

    Article  CAS  Google Scholar 

  24. SingleR web tool, http://comphealth.ucsf.edu/SingleR/.

  25. Gautier, E. L. et al. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 13, 1118–1128 (2012).

    Article  CAS  Google Scholar 

  26. Hume, D. A., Mabbott, N., Raza, S. & Freeman, T. C. Can DCs be distinguished from macrophages by molecular signatures? Nat. Immunol. 14, 187–189 (2013).

    Article  CAS  Google Scholar 

  27. Randolph, G. & Merad, M. Reply to: “Can DCs be distinguished from macrophages by molecular signatures?”. Nat. Immunol. 14, 189–190 (2013).

    Article  CAS  Google Scholar 

  28. Altboum, Z. et al. Digital cell quantification identifies global immune cell dynamics during influenza infection. Mol. Syst. Biol. 10, 720 (2014).

    Article  Google Scholar 

  29. Gibbings, S. L. et al. Three unique interstitial macrophages in the murine lung at steady state. Am. J. Respir. Cell Mol. Biol. 57, 66–76 (2017).

    Article  CAS  Google Scholar 

  30. Gong, T. & Szustakowski, J. D. DeconRNASeq: a statistical framework for deconvolution of heterogeneous tissue samples based on mRNA-Seq data. Bioinformatics 29, 1083–1085 (2013).

    Article  CAS  Google Scholar 

  31. Huang, L., Nazarova, E. V., Tan, S., Liu, Y. & Russell, D. G. Growth of Mycobacterium tuberculosis in vivo segregates with host macrophage metabolism and ontogeny. J. Exp. Med. 215, 1135–1152 (2018).

    Article  CAS  Google Scholar 

  32. Naikawadi, R. P. et al. Telomere dysfunction in alveolar epithelial cells causes lung remodeling and fibrosis. JCI Insight 1, e86704 (2016).

    Article  Google Scholar 

  33. Verhaak, R. G. et al. Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J. Clin. Invest. 123, 517–525 (2013).

    CAS  PubMed  Google Scholar 

  34. Yang, I. V. et al. Expression of cilium-associated genes defines novel molecular subtypes of idiopathic pulmonary fibrosis. Thorax 68, 1114–1121 (2013).

    Article  Google Scholar 

  35. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  Google Scholar 

  36. Zhou, X. et al. Circuit design features of a stable two-cell system. Cell 172, 744–757 e717 (2018).

    Article  CAS  Google Scholar 

  37. Xie, T. et al. Single-cell deconvolution of fibroblast heterogeneity in mouse pulmonary fibrosis. Cell Rep. 22, 3625–3640 (2018).

    Article  CAS  Google Scholar 

  38. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  Google Scholar 

  39. Richeldi, L., Collard, H. R. & Jones, M. G. Idiopathic pulmonary fibrosis. Lancet 389, 1941–1952 (2017).

    Article  Google Scholar 

  40. Noble, P. W., Barkauskas, C. E. & Jiang, D. Pulmonary fibrosis: patterns and perpetrators. J. Clin. Invest. 122, 2756–2762 (2012).

    Article  CAS  Google Scholar 

  41. Wollin, L. et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur. Respir. J. 45, 1434–1445 (2015).

    Article  CAS  Google Scholar 

  42. Olson, L. E. & Soriano, P. Increased PDGFRα activation disrupts connective tissue development and drives systemic fibrosis. Dev. Cell 16, 303–313 (2009).

    Article  CAS  Google Scholar 

  43. Iwayama, T. et al. PDGFRα signaling drives adipose tissue fibrosis by targeting progenitor cell plasticity. Genes Dev. 29, 1106–1119 (2015).

    Article  CAS  Google Scholar 

  44. Horikawa, S. et al. PDGFRα plays a crucial role in connective tissue remodeling. Sci. Rep. 5, 17948 (2015).

    Article  CAS  Google Scholar 

  45. Hayes, B. J. et al. Activation of platelet-derived growth factor receptor alpha contributes to liver fibrosis. PLoS ONE 9, e92925 (2014).

    Article  Google Scholar 

  46. Demaria, M. et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31, 722–733 (2014).

    Article  CAS  Google Scholar 

  47. Chen, Y. T. et al. Platelet-derived growth factor receptor signaling activates pericyte–myofibroblast transition in obstructive and post-ischemic kidney fibrosis. Kidney Int. 80, 1170–1181 (2011).

    Article  CAS  Google Scholar 

  48. Kitagawa, M. et al. Phf14, a novel regulator of mesenchyme growth via platelet-derived growth factor (PDGF) receptor-alpha. J. Biol. Chem. 287, 27983–27996 (2012).

    Article  CAS  Google Scholar 

  49. Qin, D., Xia, Y. & Whitesides, G. M. Soft lithography for micro- and nanoscale patterning. Nat. Protoc. 5, 491–502 (2010).

    Article  CAS  Google Scholar 

  50. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  52. Dai, M. et al. Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data. Nucleic Acids Res. 33, e175 (2005).

    Article  Google Scholar 

  53. Aran, D., Hu, Z. & Butte, A. J. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 18, 220 (2017).

    Article  Google Scholar 

  54. Lachmann, A. et al. Massive mining of publicly available RNA-seq data from human and mouse. Nat. Commun. 9, 1366 (2018).

    Article  Google Scholar 

  55. 10X single-cell datasets, https://support.10xgenomics.com/single-cell-gene-expression/datasets.

  56. Raghu, G. et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am. J. Respir. Crit. Care Med. 183, 788–824 (2011).

    Article  Google Scholar 

  57. Ware, L. B. et al. Assessment of lungs rejected for transplantation and implications for donor selection. Lancet 360, 619–620 (2002).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by a UCSF Marcus Award to M.B. and A.R.A., a National Institutes of Health grant (HL131560) to M.B., a Gruss Lipper Postdoctoral Fellowship to D.A., a UCSF Nina Ireland Program award to R.P.N. and P.J.W., a National Institutes of Health grant (HL139897) to P.J.W., and a National Institutes of Health award (National Institute of Allergy and Infectious Diseases Bioinformatics Support Contract HHSN272201200028C) to A.J.B. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. We thank D. Erle, A. Barczak, W. Eckalbar, and M. Adkisson of the UCSF Functional Genomics Core Facility, the UCSF Center for Advanced Technology, the Gladstone Institutes’ Histology & Light Microscopy Core, and D. Sheppard for his insightful comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

D.A. developed SingleR and performed computational analysis of single-cell data under the guidance of A.J.B. A.P.L. performed in vivo and in vitro experiments under the guidance of M.B. and with the assistance of E.W. and S.C. L.L. performed microfluidic capture of single-cell transcriptomes, library preparation, and sequencing under the guidance of A.R.A. V.F., A.H., and E.W. prepared breeding and experimental stocks of genetically modified mice and performed lung injury models under the guidance of M.B. P.J.W. contributed acquisition, storage, and processing of human samples and, with R.P.N., acquired lung microarray data from mice with telomere dysfunction. D.A. prepared the figures. M.B. conceived of the work, supervised experimental planning and execution, and wrote the manuscript with input from D.A., A.P.L., and L.L.

Corresponding author

Correspondence to Mallar Bhattacharya.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Comparison of annotations of PBMC data with reference-based methods for cell type annotation.

This figure accompanies Fig. 1c. t-SNE plots of the PBMC-4K scRNA-seq data21. The plots are colored by the top annotation using the methods presented by Kang et al.22 (top left), Li et al.23 (top right) and SingleR (bottom left). Kang et al. correlated 173 differentially expressed genes learned by comparison of scRNA-seq clusters of PBMCs. Li et al. (RCA) used a bulk microarray reference dataset for correlation. SingleR introduced a fine-tuning step to refine correlation with bulk datasets. While all methods agreed on the general annotations of monocytes, T cells, B cells and NK cells, annotations differed for cellular subtypes: RCA annotated the left-most cells in cluster 4 as NK cells, while the method of Kang et al. and SingleR annotated them as CD8+ T cells, which was supported by the expression of individual genes CD3E (general T cell marker) and CD8A. It is of note that NKG7 and GNLY are commonly used as NK cell markers but are also expressed in activated CD8+ T cells. The method of Kang et al. and RCA annotated the cells in cluster 3 as CD4+ T cells, while SingleR annotated 47.7% of this cluster as naive CD8+ T cells and 30.4% as central memory CD8+ T cells (TCM). The CD8A marker, along with CCR7 and SELL (markers for naïve cells), supported this annotation. The expression of IL7R (marker for memory T cells) in some of those cells suggested that this cluster also contained memory cells, as annotated by SingleR. FOXP3 (a marker of regulatory T cells) was found only in a small proportion of Treg cells (in the sorted Treg cells, only 5.6% of cells expressed FOXP3); however, SingleR suggested that many of the cells in cluster 1 were in fact Treg cells. In this comparison, only SingleR was able to distinguish highly similar cell states, and these differential annotations were supported by gene markers viewed individually.

Supplementary Figure 2 Quality control of the single-cell experiment and analysis.

a, t-SNE projection showing non-zero, unique molecular identifiers (UMIs), mitochondria fraction, and cell cycle score38 (n = 8,366 cells from n = 3 biologically independent mice for bleomycin, n = 6 biologically independent mice for control). No observed clusters associated with mitochondrial gene expression; n = 30 cells can be suggested as outliers based on mitochondrial percentage. Clusters of cycling cells represent ~ 10% of macrophages and T cells and were not used in the macrophage analysis. The number of genes per cell and mitochondrial fraction is shown as a function of UMI per cell. b, The t-SNE plot colored by batches. c, The t-SNE plot is colored by the maximal SingleR score. d, The t-SNE plot is colored by –log10 P value for the SingleR annotation using the chi-squared outliers test (gray used for –log10 P value >5). All cells in this analysis received a P value <0.05. The alveolar macrophages and fibroblasts (as defined by SingleR) received higher confidence than other cells.

Supplementary Figure 3 SingleR detailed annotations of mouse lungs.

This plot accompanies Fig. 2b. Here we present all SingleR annotations (not just the main types). To reduce the number of possible cell types in the data, annotations with fewer than 15 cells were marked as ‘Other’ and are colored in black.

Supplementary Figure 4 Heat map of SingleR scores using lung reference datasets.

This heat map accompanies Fig. 2c. We used RNA-seq datasets as reference—lung macrophages from Gibbings et al.29 (downloaded from GEO accession number GSE94135) and lung dendritic cells from Altboum et al.28 (downloaded from GEO accession number GSE49932). The two datasets were combined for a lung-specific reference. The heat maps show the SingleR scores after one round, without fine-tuning, of the top-scored cell types. Scores were normalized to [0,1]. a, The majority of cells in the macrophage cluster (see Fig. 2b) were annotated to alveolar macrophages, whereas the remaining cells were most correlated with interstitial macrophage type 3, which includes CD11c+ macrophages and not dendritic cells. Interestingly, the macrophage cluster was split into two subclusters, as we show in Fig. 2c. b, Cells from the dendritic cell cluster (see Fig. 2b) are annotated as dendritic cells, showing that the annotations are not an artifact of the reference datasets.

Supplementary Figure 5 C2 expresses both alveolar and interstitial macrophage genes.

In all scatterplots, individual genes are plotted and the vertical axis is log fold change between alveolar macrophages (AM) and interstitial macrophages (IM) in independent bulk reference datasets (top, GSE94135; bottom, GSE108844). Positive levels represent genes upregulated in AM. The horizontal axis in the left plots is the log fold change between cluster C1 and cluster C2 in the lung scRNA-seq dataset. Positive levels represent genes upregulated in C1. The horizontal axis in the right plots is the log fold change between C2 (the postulated transitional cluster) and C3 (the interstitial cluster) in the single-cell data. Positive levels represent genes upregulated in C2. Genes are colored red if significant in bulk (log(FC) > 1, adjusted P value < 0.05), green if significant in single cells (adjusted P value < 0.05), blue if significant in both, and purple if non-significant. Percentage is the percent of significant genes in single cells (SC) that are in the upper-right or lower-left quadrants. R is Pearson’s coefficient of significant genes in SC. P value in correlations < 0.00001. Fold ratios were computed with the lung scRNA-seq dataset (n = 3 biologically independent mice for bleomycin, n = 6 biologically independent mice for control).

Supplementary Figure 6 Flow cytometric gating.

a, Cells were dissociated from lungs of WT mice 14 d after bleomycin injury and stained with SiglecF, CD11c, and MHC II antibodies. SiglecF+CD11c+ cells were sorted into MHCIIhi and MHClo populations, with the threshold defined by MHC II staining in an uninjured mouse. Representative data are shown. b, Lung cells were dissociated from CX3CR1CreERT2 / Rosa26-loxp-STOP-loxp-TdTomato mice induced with tamoxifen 1 d prior to and during bleomycin injury and stained with SiglecF and MHC II antibodies. Representative data are shown.

Supplementary Figure 7 Depletion of the macrophage subpopulation during bleomycin fibrosis.

a, Fluorescence microscopy demonstrating ablation of macrophages detected by immunofluorescence for SiglecF and macrophage marker MerTK26 in areas of scar by second-harmonic (SH) imaging 21 d after bleomycin injury in wild-type mice (left) and tamoxifen-induced Cx3cr1CreERT2/Rosa26-loxp-STOP-loxp-Diptheria Toxin A mice (right), with quantification in five fields of view per mouse (n = 3 biologically independent mice per group). The Wilcoxon rank-sum test two-sided P value is presented. Scale bar, 50 μm. The box plot center line is the median, box limits are the upper and lower quartiles, and whiskers denote the largest and smallest values no more than 1.5 times the interquartile range from the limits. b, H&E staining 21 d after injury of wild-type mice (left) and tamoxifen-induced Cx3cr1CreERT2 / Rosa26-loxp-STOP-loxP-Diptheria Toxin A mice (right). Scale bar, 0.5 mm. Images are representative of three independent replicates with similar results.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Notes 1 and 2

Reporting Summary

Supplementary Table 1

Drop-seq batches

Supplementary Table 2

Differentially expressed genes between cluster C1 and cluster C

Supplementary Table 3

Mouse and human orthologs of cluster C1 genes

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aran, D., Looney, A.P., Liu, L. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol 20, 163–172 (2019). https://doi.org/10.1038/s41590-018-0276-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0276-y

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