Active eosinophils regulate host defence and immune responses in colitis
Language English Country Great Britain, England Media print-electronic
Document type Journal Article, Research Support, Non-U.S. Gov't
PubMed
36509106
PubMed Central
PMC9977678
DOI
10.1038/s41586-022-05628-7
PII: 10.1038/s41586-022-05628-7
Knihovny.cz E-resources
- MeSH
- Single-Cell Gene Expression Analysis MeSH
- B7-1 Antigen metabolism MeSH
- Eosinophils * classification cytology immunology metabolism MeSH
- Inflammatory Bowel Diseases immunology MeSH
- Immunity * MeSH
- Interferon-gamma MeSH
- Interleukin-33 MeSH
- Colitis * immunology pathology MeSH
- Humans MeSH
- Mice MeSH
- Proteome MeSH
- Intestines * immunology pathology MeSH
- T-Lymphocytes MeSH
- Transcriptome MeSH
- Animals MeSH
- Check Tag
- Humans MeSH
- Mice MeSH
- Animals MeSH
- Publication type
- Journal Article MeSH
- Research Support, Non-U.S. Gov't MeSH
- Names of Substances
- B7-1 Antigen MeSH
- Cd274 protein, mouse MeSH Browser
- Interferon-gamma MeSH
- Interleukin-33 MeSH
- Proteome MeSH
In the past decade, single-cell transcriptomics has helped to uncover new cell types and states and led to the construction of a cellular compendium of health and disease. Despite this progress, some difficult-to-sequence cells remain absent from tissue atlases. Eosinophils-elusive granulocytes that are implicated in a plethora of human pathologies1-5-are among these uncharted cell types. The heterogeneity of eosinophils and the gene programs that underpin their pleiotropic functions remain poorly understood. Here we provide a comprehensive single-cell transcriptomic profiling of mouse eosinophils. We identify an active and a basal population of intestinal eosinophils, which differ in their transcriptome, surface proteome and spatial localization. By means of a genome-wide CRISPR inhibition screen and functional assays, we reveal a mechanism by which interleukin-33 (IL-33) and interferon-γ (IFNγ) induce the accumulation of active eosinophils in the inflamed colon. Active eosinophils are endowed with bactericidal and T cell regulatory activity, and express the co-stimulatory molecules CD80 and PD-L1. Notably, active eosinophils are enriched in the lamina propria of a small cohort of patients with inflammatory bowel disease, and are closely associated with CD4+ T cells. Our findings provide insights into the biology of eosinophils and highlight the crucial contribution of this cell type to intestinal homeostasis, immune regulation and host defence. Furthermore, we lay a framework for the characterization of eosinophils in human gastrointestinal diseases.
Botnar Research Center for Child Health Basel Switzerland
Department of Biosystems Science and Engineering ETH Zürich Basel Switzerland
Department of Molecular Life Sciences University of Zürich Zürich Switzerland
Institute for Food Nutrition and Health D HEST ETH Zürich Zürich Switzerland
Institute of Experimental Immunology University of Zürich Zürich Switzerland
Institute of Molecular Cancer Research University of Zürich Zürich Switzerland
Institute of Molecular Genetics of the Czech Academy of Sciences Prague Czech Republic
Institute of Pathology University of Bern Bern Switzerland
Institute of Physiology University of Zürich Zürich Switzerland
See more in PubMed
Marichal T, Mesnil C, Bureau F. Homeostatic eosinophils: characteristics and functions. Front. Med. 2017;4:101. doi: 10.3389/fmed.2017.00101. PubMed DOI PMC
Blanchard C, Wang N, Rothenberg ME. Eosinophilic esophagitis: pathogenesis, genetics, and therapy. J. Allergy Clin. Immunol. 2006;118:1054–1059. doi: 10.1016/j.jaci.2006.07.038. PubMed DOI
Humbles AA, et al. A critical role for eosinophils in allergic airways remodeling. Science. 2004;305:1776–1779. doi: 10.1126/science.1100283. PubMed DOI
Jenerowicz D, Czarnecka-Operacz M, Silny W. Peripheral blood eosinophilia in atopic dermatitis. Acta Dermatovenerol. Alp Pannonica Adriat. 2007;16:47–52. PubMed
Raab, Y., Fredens, K., Gerdin, B. & Hällgren, R. Eosinophil activation in ulcerative colitis: studies on mucosal release and localization of eosinophil granule constituents. Dig. Dis. Sci. 43, 1061–1070 (1998). PubMed
Chu, V. T. et al. Eosinophils promote generation and maintenance of immunoglobulin-A-expressing plasma cells and contribute to gut immune homeostasis. Immunity40, 582–593 (2014). PubMed
Jung Y, et al. IL-1β in eosinophil-mediated small intestinal homeostasis and IgA production. Mucosal Immunol. 2015;8:930–942. doi: 10.1038/mi.2014.123. PubMed DOI PMC
Ignacio, A. et al. Small intestinal resident eosinophils maintain gut homeostasis following microbial colonization. Immunity55, 1250–1267 (2022). PubMed
Sugawara R, et al. Small intestinal eosinophils regulate Th17 cells by producing IL-1 receptor antagonist. J. Exp. Med. 2016;213:555–567. doi: 10.1084/jem.20141388. PubMed DOI PMC
Alhmoud T, et al. Outcomes of inflammatory bowel disease in patients with eosinophil-predominant colonic inflammation. BMJ Open Gastroenterol. 2020;7:e000373. doi: 10.1136/bmjgast-2020-000373. PubMed DOI PMC
Smillie CS, et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell. 2019;178:714–730. doi: 10.1016/j.cell.2019.06.029. PubMed DOI PMC
Sikkema, L. et al. An integrated cell atlas of the human lung in health and disease. Preprint at 10.1101/2022.03.10.483747 (2022).
Lee NA, et al. Expression of IL-5 in thymocytes/T cells leads to the development of a massive eosinophilia, extramedullary eosinophilopoiesis, and unique histopathologies. J. Immunol. 1997;158:1332–1344. doi: 10.4049/jimmunol.158.3.1332. PubMed DOI
Mahmudi-Azer S, Downey GP, Moqbel R. Translocation of the tetraspanin CD63 in association with human eosinophil mediator release. Blood. 2002;99:4039–4047. doi: 10.1182/blood.V99.11.4039. PubMed DOI
Khushman M, et al. Exosomal markers (CD63 and CD9) expression and their prognostic significance using immunohistochemistry in patients with pancreatic ductal adenocarcinoma. J. Gastrointest. Oncol. 2019;10:695–702. doi: 10.21037/jgo.2018.07.02. PubMed DOI PMC
Cohnen A, et al. Surface CD107a/LAMP-1 protects natural killer cells from degranulation-associated damage. Blood. 2013;122:1411–1418. doi: 10.1182/blood-2012-07-441832. PubMed DOI
Mesnil C, et al. Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J. Clin. Invest. 2016;126:3279–3295. doi: 10.1172/JCI85664. PubMed DOI PMC
Schwarzfischer M, et al. TiO2 nanoparticles abrogate the protective effect of the Crohn’s disease-associated variation within the PTPN22 gene locus. Gut. 2022 doi: 10.1136/gutjnl-2021-325911. PubMed DOI
Qiu X, et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods. 2017;14:979–982. doi: 10.1038/nmeth.4402. PubMed DOI PMC
Bergen V, Lange M, Peidli S, Wolf FA, Theis FJ. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 2020;38:1408–1414. doi: 10.1038/s41587-020-0591-3. PubMed DOI
Lange M, et al. CellRank for directed single-cell fate mapping. Nat. Methods. 2022;19:159–170. doi: 10.1038/s41592-021-01346-6. PubMed DOI PMC
McFaline-Figueroa JL, et al. A pooled single-cell genetic screen identifies regulatory checkpoints in the continuum of the epithelial-to-mesenchymal transition. Nat. Genet. 2019;51:1389–1398. doi: 10.1038/s41588-019-0489-5. PubMed DOI PMC
Arnold IC, et al. Eosinophils suppress Th1 responses and restrict bacterially induced gastrointestinal inflammation. J. Exp. Med. 2018;215:2055–2072. doi: 10.1084/jem.20172049. PubMed DOI PMC
Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 2020;15:1484–1506. doi: 10.1038/s41596-020-0292-x. PubMed DOI
Masterson JC, et al. Eosinophil-mediated signalling attenuates inflammatory responses in experimental colitis. Gut. 2015;64:1236–1247. doi: 10.1136/gutjnl-2014-306998. PubMed DOI PMC
Arnold, I. C. et al. The GM–CSF–IRF5 signaling axis in eosinophils promotes antitumor immunity through activation of type 1 T cell responses. J. Exp. Med.217, e20190706 (2020). PubMed PMC
Griseri T, et al. Granulocyte macrophage colony-stimulating factor-activated eosinophils promote interleukin-23 driven chronic colitis. Immunity. 2015;43:187–199. doi: 10.1016/j.immuni.2015.07.008. PubMed DOI PMC
Aibar S, et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods. 2017;14:1083–1086. doi: 10.1038/nmeth.4463. PubMed DOI PMC
Griesenauer B, Paczesny S. The ST2/IL-33 axis in immune cells during inflammatory diseases. Front. Immunol. 2017;8:475. doi: 10.3389/fimmu.2017.00475. PubMed DOI PMC
Kang K, et al. Interferon-γ represses M2 gene expression in human macrophages by disassembling enhancers bound by the transcription factor MAF. Immunity. 2017;47:235–250. doi: 10.1016/j.immuni.2017.07.017. PubMed DOI PMC
Menzella, F. et al. Anti-IL5 therapies for severe eosinophilic asthma: literature review and practical insights. J. Asthma Allergy13, 301–313 (2020). PubMed PMC
Adachi O, et al. Targeted disruption of the MyD88 gene results in loss of IL-1-and IL-18-mediated function. Immunity. 1998;9:143–150. doi: 10.1016/S1074-7613(00)80596-8. PubMed DOI
Hoshino K, et al. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J. Immunol. 1999;162:3749–3752. doi: 10.4049/jimmunol.162.7.3749. PubMed DOI
Rawlins EL, Clark CP, Xue Y, Hogan BLM. The Id2+ distal tip lung epithelium contains individual multipotent embryonic progenitor cells. Development. 2009;136:3741–3745. doi: 10.1242/dev.037317. PubMed DOI PMC
Dent LA, Strath M, Mellor AL, Sanderson CJ. Eosinophilia in transgenic mice expressing interleukin 5. J. Exp. Med. 1990;172:1425–1431. doi: 10.1084/jem.172.5.1425. PubMed DOI PMC
Lee H-M, et al. IFNγ signaling endows DCs with the capacity to control type I inflammation during parasitic infection through promoting T-bet+ regulatory T cells. PLoS Pathog. 2015;11:e1004635. doi: 10.1371/journal.ppat.1004635. PubMed DOI PMC
Oboki K, et al. IL-33 is a crucial amplifier of innate rather than acquired immunity. Proc. Natl Acad. Sci. USA. 2010;107:18581–18586. doi: 10.1073/pnas.1003059107. PubMed DOI PMC
Townsend MJ, Fallon PG, Matthews DJ, Jolin HE, McKenzie AN. T1/St2-deficient mice demonstrate the importance of T1/St2 in developing primary T helper cell type 2 responses. J. Exp. Med. 2000;191:1069–1076. doi: 10.1084/jem.191.6.1069. PubMed DOI PMC
Lee JJ, et al. Defining a link with asthma in mice congenitally deficient in eosinophils. Science. 2004;305:1773–1776. doi: 10.1126/science.1099472. PubMed DOI
Doyle AD, et al. Homologous recombination into the eosinophil peroxidase locus generates a strain of mice expressing Cre recombinase exclusively in eosinophils. J. Leukoc. Biol. 2013;94:17–24. doi: 10.1189/jlb.0213089. PubMed DOI PMC
Diehl GE, et al. Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX3CR1hi cells. Nature. 2013;494:116–120. doi: 10.1038/nature11809. PubMed DOI PMC
Wiles S, Pickard KM, Peng K, MacDonald TT, Frankel G. In vivo bioluminescence imaging of the murine pathogen Citrobacter rodentium. Infect. Immun. 2006;74:5391–5396. doi: 10.1128/IAI.00848-06. PubMed DOI PMC
Hao Y, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573–3587. doi: 10.1016/j.cell.2021.04.048. PubMed DOI PMC
Korsunsky I, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods. 2019;16:1289–1296. doi: 10.1038/s41592-019-0619-0. PubMed DOI PMC
Kowalczyk MS, et al. Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res. 2015;25:1860–1872. doi: 10.1101/gr.192237.115. PubMed DOI PMC
Trapnell C, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 2014;32:381–386. doi: 10.1038/nbt.2859. PubMed DOI PMC
Cacchiarelli D, et al. Aligning single-cell developmental and reprogramming trajectories identifies molecular determinants of myogenic reprogramming outcome. Cell Syst. 2018;7:258–268. doi: 10.1016/j.cels.2018.07.006. PubMed DOI
La Manno G, et al. RNA velocity of single cells. Nature. 2018;560:494–498. doi: 10.1038/s41586-018-0414-6. PubMed DOI PMC
Holland CH, et al. Robustness and applicability of transcription factor and pathway analysis tools on single-cell RNA-seq data. Genome Biol. 2020;21:36. doi: 10.1186/s13059-020-1949-z. PubMed DOI PMC
Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32:2847–2849. doi: 10.1093/bioinformatics/btw313. PubMed DOI
Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer, 2016).
Brummelman J, et al. Development, application and computational analysis of high-dimensional fluorescent antibody panels for single-cell flow cytometry. Nat. Protoc. 2019;14:1946–1969. doi: 10.1038/s41596-019-0166-2. PubMed DOI
Dyer, K. D. et al. Functionally competent eosinophils differentiated ex vivo in high purity from normal mouse bone marrow. J. Immunol.181, 4004–4009 (2008). PubMed PMC
Hatakeyama M, et al. SUSHI: an exquisite recipe for fully documented, reproducible and reusable NGS data analysis. BMC Bioinformatics. 2016;17:228. doi: 10.1186/s12859-016-1104-8. PubMed DOI PMC
Robinson MD, McCarthy DJ, Smyth G. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616. PubMed DOI PMC
Kolde, R. pheatmap: Pretty Heatmaps. R version 1.0.12, https://cran.r-project.org/web/packages/pheatmap/index.html (2019).
Horlbeck MA, et al. Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation. eLife. 2016;5:e19760. doi: 10.7554/eLife.19760. PubMed DOI PMC
Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.J. 2011;17:10–12. doi: 10.14806/ej.17.1.200. DOI
Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat. Methods. 2012;9:357–359. doi: 10.1038/nmeth.1923. PubMed DOI PMC
Li W, et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 2014;15:554. doi: 10.1186/s13059-014-0554-4. PubMed DOI PMC
Stringer C, Wang T, Michaelos M, Pachitariu M. Cellpose: a generalist algorithm for cellular segmentation. Nat. Methods. 2021;18:100–106. doi: 10.1038/s41592-020-01018-x. PubMed DOI
Phipson B, Smyth GK. Permutation P-values should never be zero: calculating exact P-values when permutations are randomly drawn. Stat. Appl. Genet. Mol. Biol. 2010;9:Article39. doi: 10.2202/1544-6115.1585. PubMed DOI
Lohoff T, et al. Integration of spatial and single-cell transcriptomic data elucidates mouse organogenesis. Nat. Biotechnol. 2022;40:74–85. doi: 10.1038/s41587-021-01006-2. PubMed DOI PMC