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.

  • Resource
  • Published:

Single-cell profiling identifies mechanisms of inflammatory heterogeneity in chronic rhinosinusitis

Abstract

The heterogeneous cellular microenvironment of human airway chronic inflammatory diseases, including chronic rhinosinusitis (CRS) and asthma, is still poorly understood. Here, we performed single-cell RNA sequencing (scRNA-seq) on the nasal mucosa of healthy individuals and patients with three subtypes of CRS and identified disease-specific cell subsets and molecules that specifically contribute to the pathogenesis of CRS subtypes. As such, ALOX15+ macrophages contributed to the type 2 immunity-driven pathogenesis of one subtype of CRS, eosinophilic CRS with nasal polyps (eCRSwNP), by secreting chemokines that recruited eosinophils, monocytes and T helper 2 (TH2) cells. An inhibitor of ALOX15 reduced the release of proinflammatory chemokines in human macrophages and inhibited the overactivation of type 2 immunity in a mouse model of eosinophilic rhinosinusitis. Our findings advance the understanding of the heterogeneous immune microenvironment and the pathogenesis of CRS subtypes and identify potential therapeutic approaches for the treatment of CRS and potentially other type 2 immunity-mediated diseases.

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: Identification of cell types in human nasal mucosal microenvironment by 10x Genomics scRNA-seq.
Fig. 2: Compositions and functions of epithelial subsets in control and CRS.
Fig. 3: Overactivated type 2 immune response of lymphocytes contributes to the pathogenesis of eCRSwNP.
Fig. 4: ALOX15+ cDC2s and LAMP3+ DCs primed type 2 immune response in eCRSwNP.
Fig. 5: Enriched ALOX15+ macrophages play key roles in type 2 immune response of eCRSwNP.
Fig. 6: ALOX15 blockade treatment alleviates the type 2 immune response in vitro and in vivo.

Similar content being viewed by others

Data availability

The accession number for the raw sequencing data from human samples in this paper is GSA (Genome Sequence Archive) HRA000772. GRCh38 human genome was used as the reference genome (https://cf.10xgenomics.com/supp/cell-exp/refdata-gex-GRCh38-2020-A.tar.gz). Source data are provided with this paper.

References

  1. Yii, A. C. A. et al. Precision medicine in united airways disease: a “treatable traits” approach. Allergy 73, 1964–1978 (2018).

    Article  CAS  Google Scholar 

  2. Samitas, K., Carter, A., Kariyawasam, H. H. & Xanthou, G. Upper and lower airway remodelling mechanisms in asthma, allergic rhinitis and chronic rhinosinusitis: the one airway concept revisited. Allergy 73, 993–1002 (2018).

    Article  CAS  Google Scholar 

  3. Fokkens, W. J. et al. European Position Paper on Rhinosinusitis and Nasal Polyps 2020. Rhinology 58, 1–464 (2020).

    Article  Google Scholar 

  4. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention, 2021 http://www.ginasthma.org (2021).

  5. Bachert, C. et al. Adult chronic rhinosinusitis. Nat. Rev. Dis. Prim. 6, 86 (2020).

    Article  Google Scholar 

  6. Schleimer, R. P. Immunopathogenesis of chronic rhinosinusitis and nasal polyposis. Annu Rev. Pathol. 12, 331–357 (2017).

    Article  CAS  Google Scholar 

  7. Delemarre, T., Bochner, B. S., Simon, H. U. & Bachert, C. Rethinking neutrophils and eosinophils in chronic rhinosinusitis. J. Allergy Clin. Immunol. 148, 327–335 (2021).

    Article  CAS  Google Scholar 

  8. Lou, H., Wang, C. & Zhang, L. Endotype-driven precision medicine in chronic rhinosinusitis. Expert Rev. Clin. Immunol. 15, 1171–1183 (2019).

    Article  CAS  Google Scholar 

  9. Cao, P. P., Wang, Z. C., Schleimer, R. P. & Liu, Z. Pathophysiologic mechanisms of chronic rhinosinusitis and their roles in emerging disease endotypes. Ann. Allergy Asthma Immunol. 122, 33–40 (2019).

    Article  CAS  Google Scholar 

  10. Ordovas-Montanes, J. et al. Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature 560, 649–654 (2018).

    Article  CAS  Google Scholar 

  11. Vieira Braga, F. A. et al. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat. Med. 25, 1153–1163 (2019).

    Article  CAS  Google Scholar 

  12. Xu, X., Ong, Y. K. & Wang, Y. Novel findings in immunopathophysiology of chronic rhinosinusitis and their role in a model of precision medicine. Allergy 75, 769–780 (2020).

    Article  Google Scholar 

  13. Whitsett, J. A. & Alenghat, T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat. Immunol. 16, 27–35 (2015).

    Article  CAS  Google Scholar 

  14. Montoro, D. T. et al. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 560, 319–324 (2018).

    Article  CAS  Google Scholar 

  15. Zuo, W. L. et al. Ontogeny and biology of human small airway epithelial club cells. Am. J. Respir. Crit. Care Med. 198, 1375–1388 (2018).

    Article  CAS  Google Scholar 

  16. Hawkins, F. J. et al. Derivation of airway basal stem cells from human pluripotent stem cells. Cell. Stem Cell. 28, 79–95 e78 (2021).

    Article  CAS  Google Scholar 

  17. Pavlidis, S. et al. "T2-high” in severe asthma related to blood eosinophil, exhaled nitric oxide and serum periostin.Eur. Respir. J. 53, 1800938 (2019).

    Article  CAS  Google Scholar 

  18. Li, Z. et al. 15-Lipoxygenase 1 in nasal polyps promotes CCL26/eotaxin 3 expression through extracellular signal-regulated kinase activation. J. Allergy Clin. Immunol. 144, 1228–1241 (2019).

    Article  CAS  Google Scholar 

  19. Yan, B. et al. Epithelium-derived cystatin SN enhances eosinophil activation and infiltration through IL-5 in patients with chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 144, 455–469 (2019).

    Article  CAS  Google Scholar 

  20. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    Article  CAS  Google Scholar 

  21. Teufelberger, A. R. et al. The IL-33/ST2 axis is crucial in type 2 airway responses induced by Staphylococcus aureus-derived serine protease-like protein D. J. Allergy Clin. Immunol. 141, 549–559 (2018).

    Article  CAS  Google Scholar 

  22. Bottcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018).

    Article  CAS  Google Scholar 

  23. Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).

    Article  CAS  Google Scholar 

  24. Zhang, Q. et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829–845 (2019).

    Article  CAS  Google Scholar 

  25. Buchheit, K. M. et al. IL-5Rα marks nasal polyp IgG4- and IgE-expressing cells in aspirin-exacerbated respiratory disease. J. Allergy Clin. Immunol. 145, 1574–1584 (2020).

    Article  CAS  Google Scholar 

  26. Stevens, W. W. et al. Associations between inflammatory endotypes and clinical presentations in chronic rhinosinusitis. J. Allergy Clin. Immunol. Pract. 7, 2812–2820 (2019).

    Article  Google Scholar 

  27. Collin, M. & Bigley, V. Human dendritic cell subsets: an update. Immunology 154, 3–20 (2018).

    Article  CAS  Google Scholar 

  28. Böttcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018).

    Article  Google Scholar 

  29. Weizman, O. E. et al. ILC1 confer early host protection at initial sites of viral infection. Cell 171, 795–808 (2017).

    Article  CAS  Google Scholar 

  30. Lugrin, J. & Martinon, F. The AIM2 inflammasome: sensor of pathogens and cellular perturbations. Immunological Rev. 281, 99–114 (2018).

    Article  CAS  Google Scholar 

  31. Rothe, T. et al. 12/15-Lipoxygenase-mediated enzymatic lipid oxidation regulates DC maturation and function. J. Clin. Invest. 125, 1944–1954 (2015).

    Article  Google Scholar 

  32. Ryan, E. J. et al. Dendritic cell-associated lectin-1: a novel dendritic cell-associated, C-type lectin-like molecule enhances T cell secretion of IL-4. J. Immunol. 169, 5638–5648 (2002).

    Article  CAS  Google Scholar 

  33. Eguíluz-Gracia, I. et al. Rapid recruitment of CD14+ monocytes in experimentally induced allergic rhinitis in human subjects. J. Allergy Clin. Immunol. 137, 1872–1881 (2016).

    Article  Google Scholar 

  34. 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 

  35. Subrata, L. S. et al. Interactions between innate antiviral and atopic immunoinflammatory pathways precipitate and sustain asthma exacerbations in children. J. Immunol. 183, 2793–2800 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  37. Minhas, P. S. et al. Macrophage de novo NAD+ synthesis specifies immune function in aging and inflammation. Nat. Immunol. 20, 50–63 (2019).

    Article  CAS  Google Scholar 

  38. Zhang, J., Pang, Y., Xie, T. & Zhu, L. CXCR4 antagonism in combination with IDO1 inhibition weakens immune suppression and inhibits tumor growth in mouse breast cancer bone metastases. Onco Targets Ther. 12, 4985–4992 (2019).

    Article  CAS  Google Scholar 

  39. Karin, N., Wildbaum, G. & Thelen, M. Biased signaling pathways via CXCR3 control the development and function of CD4+ T cell subsets. J. Leukoc. Biol. 99, 857–862 (2016).

    Article  CAS  Google Scholar 

  40. He, Y., Hara, H. & Núñez, G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem. Sci. 41, 1012–1021 (2016).

    Article  CAS  Google Scholar 

  41. Das, U. N. Arachidonic acid and other unsaturated fatty acids and some of their metabolites function as endogenous antimicrobial molecules: a review. J. Adv. Res. 11, 57–66 (2018).

    Article  CAS  Google Scholar 

  42. David, B. A. & Kubes, P. Exploring the complex role of chemokines and chemoattractants in vivo on leukocyte dynamics. Immunol. Rev. 289, 9–30 (2019).

    Article  CAS  Google Scholar 

  43. Tharakan, A. et al. Characterization of a novel, papain-inducible murine model of eosinophilic rhinosinusitis. Int. Forum Allergy Rhinol. 8, 513–521 (2018).

    Article  Google Scholar 

  44. Snodgrass, R. G. & Brune, B. Regulation and functions of 15-lipoxygenases in human macrophages. Front. Pharmacol. 10, 719 (2019).

    Article  CAS  Google Scholar 

  45. Shi, L. L. et al. Disease-specific T-helper cell polarizing function of lesional dendritic cells in different types of chronic rhinosinusitis with nasal polyps. Am. J. Respir. Crit. Care Med. 190, 628–638 (2014).

    Article  Google Scholar 

  46. Kristjansson, R. P. et al. A loss-of-function variant in ALOX15 protects against nasal polyps and chronic rhinosinusitis. Nat. Genet. 51, 267–276 (2019).

    Article  CAS  Google Scholar 

  47. Sachs-Olsen, C. et al. Eoxins: a new inflammatory pathway in childhood asthma. J. Allergy Clin. Immunol. 126, 859–867 (2010).

    Article  CAS  Google Scholar 

  48. Feltenmark, S. et al. Eoxins are proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells. PNAS 105, 680–685 (2008).

    Article  CAS  Google Scholar 

  49. Lambrechts, D. et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 24, 1277–1289 (2018).

    Article  CAS  Google Scholar 

  50. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  Google Scholar 

  51. Fonseka, C. Y. et al. Mixed-effects association of single cells identifies an expanded effector CD4+ T cell subset in rheumatoid arthritis.Sci. Transl. Med. 10, eaaq0305 (2018).

    Article  Google Scholar 

  52. Zimmerman, K. D., Espeland, M. A. & Langefeld, C. D. A practical solution to pseudoreplication bias in single-cell studies. Nat. Commun. 12, 738 (2021).

    Article  CAS  Google Scholar 

  53. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  Google Scholar 

  54. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. 20, 59 (2019).

    Article  Google Scholar 

  55. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  Google Scholar 

  56. Haghverdi, L., Büttner, M., Wolf, F. A., Buettner, F. & Theis, F. J. Diffusion pseudotime robustly reconstructs lineage branching. Nat. Methods 13, 845–848 (2016).

    Article  CAS  Google Scholar 

  57. Whitfield, M. L., George, L. K., Grant, G. D. & Perou, C. M. Common markers of proliferation. Nat. Rev. Cancer 6, 99–106 (2006).

    Article  CAS  Google Scholar 

  58. Zhang, L. et al. Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell 181, 442–459 e429 (2020).

    Article  CAS  Google Scholar 

  59. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308 e1236 (2018).

    Article  CAS  Google Scholar 

  60. Miller, J. C. et al. Deciphering the transcriptional network of the dendritic cell lineage. Nat. Immunol. 13, 888–899 (2012).

    Article  CAS  Google Scholar 

  61. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 e1310 (2019).

    Article  CAS  Google Scholar 

  62. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  Google Scholar 

  63. Travaglini, K. J. et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature 587, 619–625 (2020).

    Article  CAS  Google Scholar 

  64. Mei, H. E., Leipold, M. D. & Maecker, H. T. Platinum-conjugated antibodies for application in mass cytometry. Cytom. A 89, 292–300 (2016).

    Article  CAS  Google Scholar 

  65. Samusik, N., Good, Z., Spitzer, M. H., Davis, K. L. & Nolan, G. P. Automated mapping of phenotype space with single-cell data. Nat. Methods 13, 493–496 (2016).

    Article  CAS  Google Scholar 

  66. van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

Download references

Acknowledgements

We thank C. Liu and X. Yang for technical support with the development of murine rhinosinusitis models and Q. Yang and X. Chen from Guidon Pharmaceutics for technical support with the MACS and Luminex assays. We also thank H. Weng for her help refining the wording of this manuscript. Extended Data Fig. 10 was created using BioRender.com. This project was supported by grants from the National Natural Science Foundation of China (82071027 to W.L.; 82101200 to W.W.; 82071791 and 81602503 to J.Z.; and 91542117, 81673010, U20A20374 and 81471574 to W.H.), the Natural Science Foundation of Beijing (7202162 to W.L.), the CAMS Innovation Fund for Medical Sciences (2021-I2M-1-005 to J.Z.; 2021-I2M-1-035 to H.C.; and 2021-I2M-1-053 to Y.X.), the Non-profit Central Research Institute Fund of Chinese Academy of Medical Science (2019XK320034 to W.L.) and CAMS Central Public Welfare Scientific Research Institute Basal Research Expenses (2017PT31014 and 2018PT32004 to H.C. and 2018PT31052 to J.Z.).

Author information

Authors and Affiliations

Authors

Contributions

W.W., J.Z., W.L. and W.H. designed the study, W.L., X.W. and Y.Z. performed surgeries, W.W., Y.X., Lun Wang, Z.Z., S.A., J.H. and Lei Wang collected patient samples and performed single-cell experiments, Y.X., M.C., H.C. and W.W performed in vitro and in vivo experiments. L.Z., W.W. and M.C. performed histologic analyses, W.W., Lun Wang, Y.X., Z.H., W.J. and Y.L. analyzed data; W.W., Lun Wang, Y.X., J.Z., W.L., W.H., H.C., Y.H., W.J. and Z.G. interpreted data; and W.W., Lun Wang and J.Z. wrote the manuscript. All authors read the manuscript, offered feedback and approved it before submission.

Corresponding authors

Correspondence to Wei He, Wei Lv or Jianmin Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Katherine Kedzierska and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial 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 Functional characterization of the epithelial cells in control and CRS patients.

a, tSNE plots showing expression of marker genes for all 8 major cell types defined in Fig. 1b. b, tSNE plots showing expression of marker genes for epithelial subsets defined in Fig. 2a. c, Heatmaps of PAGA graph displaying the evolving expression pattern of key genes in four developing pathways of epithelial cells in Fig. 2d. d, Heatmap of the enriched GO biological processes terms among the different epithelial clusters (full list of P value adjusted by Benjamini–Hochberg procedure given in Supplementary Table 5). e, Violin plots of the proliferation score of epithelial subsets with plot center and box corresponding to median and IQR respectively. f-h, Violin plots of the expression levels of selected genes in goblet (f), ciliated (g) and glandular (h) subsets among control and CRS subtypes (MAST with RE adjusted by Benjamini–Hochberg procedure; full list of P value given in Supplementary Table 4). This figure supports Figs. 1, 2.

Extended Data Fig. 2 Pathologic fibroblasts contribute to the abnormal tissue remodeling and nasal polyp formation.

a, tSNE plots displaying 7,558 fibroblasts from 21 individuals (5 healthy controls, 5 CRSsNP, 5 neCRSwNP, and 6 eCRSwNP patients) separated into 8 subsets (left) and their corresponding patient group of origin (right). b, tSNE plots showing expression of marker genes for fibroblast subsets defined in panel a. c, Box and whisker plots showing the fraction of fibroblasts originating from 4 groups in each cluster, with plot center, box and whiskers corresponding to median, IQR and 1.5 × IQR, respectively (MASC adjusted by Benjamini–Hochberg procedure; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; Supplementary Table 2). d, Bubble heatmap showing marker genes across 8 fibroblast clusters from panel a (nonparametric two-sided Wilcoxon rank sum test adjusted by Bonferroni method; full list of P value given in Supplementary Table 3). e, Heatmap of the enriched GO biological processes terms among the different fibroblast clusters (full list of P value adjusted by Benjamini–Hochberg procedure given in Supplementary Table 5). All panels depict the same number of cells and individuals described in panel a.

Extended Data Fig. 3 Compositions and pathologic vascular functions of smooth muscle cells and endothelial cells in control and CRS patients.

a, tSNE plots displaying 1,953 smooth muscle cells from 21 individuals (5 healthy controls, 5 CRSsNP, 5 neCRSwNP, and 6 eCRSwNP patients) separated into 4 subsets (left) and their corresponding patient group of origin (right). b, tSNE plots showing expression of marker genes for SMC subsets defined in panel a. c, Box and whisker plots showing the fraction of SMC originating from 4 groups in each cluster, with plot center, box and whiskers corresponding to median, IQR and 1.5 × IQR, respectively (MASC adjusted by Benjamini–Hochberg procedure; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; Supplementary Table 2). d, Violin plots of the expression levels of selected genes in SMCs among control and CRS subtypes (MAST with RE adjusted by Benjamini–Hochberg procedure; full list of P value given in Supplementary Table 4). e, tSNE plots displaying 8,715 endothelial cells from 21 individuals (5 healthy controls, 5 CRSsNP, 5 neCRSwNP, and 6 eCRSwNP patients) separated into 5 subsets (left) and their corresponding patient group of origin (right). f, tSNE plots showing expression of marker genes for endothelial subsets defined in panel e. g, Box and whisker plots showing the fraction of endothelial cells originating from 4 groups in each cluster, with plot center, box and whiskers corresponding to median, IQR and 1.5 × IQR, respectively (MASC adjusted by Benjamini–Hochberg procedure; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; Supplementary Table 2). h, Violin plots of the expression levels of selected genes in endothelial cells among control and CRS subtypes (MAST with RE adjusted by Benjamini–Hochberg procedure; full list of P value given in Supplementary Table 4). i, Violin plot showing the expression levels of PLAT in 8 major cell types of nasal mucosa. All panels depict the same number of cells and individuals described in panel a or e.

Extended Data Fig. 4 Compositions and functions of T and B lymphocytes contributes to the pathogenesis of eCRSwNP.

a, tSNE plots showing expression of marker genes for generalized T cell subsets defined in Fig. 3a. b, Violin plots of the expression levels of selected genes in CD8+ TRM among control and CRS subtypes (MAST with RE adjusted by Benjamini–Hochberg procedure; full list of P value given in Supplementary Table 4; full list of P value given in Supplementary Table 4). c, tSNE plots showing expression of marker genes for B cell subsets defined in Fig. 3e. d, Box and whisker plots showing the fraction of B lymphocytes originating from 4 groups (5 healthy controls, 5 CRSsNP, 5 neCRSwNP, and 6 eCRSwNP patients) in each cluster, with plot center, box and whiskers corresponding to median, IQR and 1.5 × IQR, respectively (MASC adjusted by Benjamini–Hochberg procedure; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; Supplementary Table 2). This figure supports Fig. 3.

Extended Data Fig. 5 Chemokine signaling interactions in nasal immune microenvironment.

Dot plots showing expression of chemokine ligands (left) and receptors (right) in all nasal cells; only significant interactions between chemokine ligands and receptors were connected by lines. Colored lines highlight the previously unknown ligand sources (target cells) with migrating immune cell types expressing cognate receptor.

Extended Data Fig. 6 Distinct compositions and functions of myeloid cells in control and CRS patients.

a, tSNE plots showing expression of marker genes for myeloid subsets defined in Fig. 4a. b, Violin plots of the expression levels of selected genes in mast cell subsets (nonparametric two-sided Wilcoxon rank sum test adjusted by Bonferroni method; full list of P value given in Supplementary Table 3). c, Correlation analysis of the gene expression similarity of DC subsets in CRS and HCC. d, Violin plots of the M1 (top) and M2 (bottom) phenotype score across 10 macrophage clusters with plot center and box corresponding to median and IQR respectively, calculated by the mean expression of corresponding signature genes. e, tSNE plot separately showing the disease-specific distribution of 11 macrophages clusters in four patient groups. The serial number of each cell cluster is the same with Fig. 5a. f, Violin plots of selected gene expression in tissue-resident macrophages with plot center and box corresponding to median and IQR respectively (nonparametric two-sided Wilcoxon rank sum test adjusted by Bonferroni method). g, Violin plots of selected gene expression in all macrophages with plot center and box corresponding to median and IQR respectively (nonparametric two-sided Wilcoxon rank sum test adjusted by Bonferroni method). h, Graphic summary illustrating the cell origins of arachidonic acid metabolic pathway including enzymes, receptors and their alterations in CRS subtypes. EnC, Endothelial cell; EpC, Epithelial cell; Fb, Fibroblast; MC, Mast cell; Mφ, Macrophage; NE, Neutrophil; NK, NK-CX3CR1; PC, Plasma cell; SMC, Smooth Muscle cell. This figure supports Figs. 4, 5.

Extended Data Fig. 7 Mass cytometry analysis of cell compositions in control and CRS patients.

a, tSNE plot displaying clustering of immune and nonimmune cells pooled from 50,000 cellular events in each group and equally divided among each individual (3 healthy controls, 3 CRSsNP patients, 3 neCRSwNP patients, and 5 eCRSwNP patients). b, tSNE plots showing expression of marker genes for cell subsets defined in the panel a. c, tSNE plot displaying clustering of myeloid cells from panel a. d, tSNE plots showing expression of marker genes for myeloid subsets defined in the panel c. e, tSNE plot displaying clustering of T cells and NK cells from panel a. f, tSNE plots showing expression of marker genes for T cell and NK cell subsets defined in the panel e. g, Box and whisker plots showing the proportion of the CX3CR1+ and CD209+ tissue-resident macrophages in all macrophages and the proportion of the CD8+ TRM in all T cells with plot center, box and whiskers corresponding to median, IQR and 1.5 × IQR, respectively. Unpaired two-sided t-tests. *P < 0.05. h, Violin plots showing the expression levels of CD1b in cDC2s among control and CRS subtypes. Unpaired two-sided t-tests. *P < 0.05, **P < 0.01, ****P < 0.0001. All panels depict the same number of individuals described in panel a.

Extended Data Fig. 8 Expression of ALOX15 in human nasal samples.

a, Representative immunofluorescent assay was performed with anti-ALOX15 (green) and anti-CD3 (red) for T cells, anti-CD20 (red) for B cells, anti-CD138 (red) for plasma cells, anti-EMBP (red) for eosinophils, anti-CD11c (red) for DCs, and anti-Tryptase (red) for mast cells (n = 4 for each group). Scale bars, 10 µm. Results are presented as Mean ± SEM in bar plots. Unpaired two-sided t-tests. *P < 0.05, **P < 0.01. b, The Pearson correlation analysis (two-sided) between ALOX15 expression and four clinical parameters evaluating CRS severity, including tissue eosinophil count/HPF, serum total IgE level, SNOT-22 score, and CT Lund-MacKay score. The expression levels of ALOX15 measured by ELISA in surgical nasal samples of an independent cohort of patients with CRSsNP (n = 27), neCRSwNP (n = 28), and eCRSwNP (n = 32). This figure supports Fig. 6.

Extended Data Fig. 9 ALOX15 blockade treatment alleviates the type 2 immune response in vitro and in vivo.

a, Chemokines with insignificant modulation by ALOX15 in IL-4/IL-13-stimulating macrophages co-incubated with or without PD146176 (n = 3 for each group). Unpaired two-sided t-tests. *P < 0.05. b, Representative western blot confirming the knockdown of ALOX15 expression in IL-4/IL-13-stimulating macrophages transfected with ALOX15 siRNA or scramble siRNA (n = 4 for each group). Unpaired two-sided t-tests. **P < 0.01, ****P < 0.0001. c, Chemokines with insignificant modulation by ALOX15 in IL-4/IL-13-stimulating macrophages transfected with ALOX15 siRNA or scramble siRNA (n = 5 for each group). Unpaired two-sided t-tests. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. d, Schematic drawing showing treatment strategy of a papain-induced eosinophilic rhinosinusitis murine model. e, Flow cytometry gating strategy for total immune cells (CD45+), epithelial cells (CD45EpCAM+), CD4+ T cells (CD45+CD3+CD4+), eosinophils (CD45+Siglec-F+), B cells (CD45+B220+), macrophages (CD45+F4/80+) in murine sinonasal mucosa. f, Representative plots of flow cytometry showing CD45, EPCAM, CD3, CD4, Siglec-F, B220, F4/80 expression in murine sinonasal mucosa under the indicated conditions at Day 16 (n = 4 for each group), related to Fig. 6g. This figure supports Fig. 6.

Source data

Extended Data Fig. 10 Graphic summary of novel insights on the immune microenvironment in eCRSwNP from this study.

The graphic summary illustrates novel insights on different nasal mucosa immune microenvironments between eCRSwNP and healthy controls, and how the type 2 immune response promotes the pathogenesis of nasal polyps in our scRNA-seq study. In eCRSwNP, the hallmarks of pathogenesis included: (1) hyperplasia of basal cells and deficiency of protective cells and molecules, (2) extracellular matrix remodeling dysfunction by fibroblasts, macrophages, vascular endothelial cells, and smooth muscle cells (SMCs), (3) elevated levels of TH2 cells, ILC2s, IL5RA+ plasma cells, and cytotoxic CX3CR1+ CD8+ TEFF cells and NK cells, and deficiency of CD8+ TRM cells, (4) defects in the NK-cDC1 immunologic surveillance axis and overactivation of the ALOX15+ cDC2-TH2 axis, and very importantly, (5) enrichment of ALOX15+ macrophages with a prominent chemotactic effect as a driver of the type 2 immunity by promoting the infiltration of various inflammatory cells.

Supplementary information

Supplementary Information

Supplementary Text 1–8 and associated references.

Reporting Summary

Supplementary Table 1

Supplementary Tables 1–7.

Source data

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, W., Xu, Y., Wang, L. et al. Single-cell profiling identifies mechanisms of inflammatory heterogeneity in chronic rhinosinusitis. Nat Immunol 23, 1484–1494 (2022). https://doi.org/10.1038/s41590-022-01312-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01312-0

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