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.

  • Brief Communication
  • Published:

Single-cell meta-analysis of inflammatory bowel disease with scIBD

Abstract

Understanding the heterogeneous intestinal microenvironment is critical to uncover the pathogenesis of inflammatory bowel disease (IBD). Recent advances in single-cell RNA sequencing (scRNA-seq) have identified certain cell types and genes that could contribute to IBD; however, a comprehensively integrated analysis of these scRNA-seq datasets is not yet available. Here we introduce scIBD, a platform for single-cell meta-analysis of IBD with interactive and visualization features, which combines highly curated single-cell datasets in a uniform workflow, enabling identifying rare or less-characterized cell types in IBD and dissecting the commonalities, as well as the differences between ulcerative colitis and Crohn’s disease. scIBD also incorporates multifunctional information—including regulon activity, GWAS-implicated risk genes and genes targeted by therapeutics—to infer clinically relevant cell-type specificity. Collectively, scIBD is a user-friendly web-based platform for the community to analyze the transcriptome features and gene regulatory networks associated with the pathogenesis and treatment of IBD at single-cell resolution.

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: Single-cell transcriptional atlas and disease-associated cell subtypes in IBD.
Fig. 2: Linking GWAS-risk genes and therapeutic targets to cell subtypes in scIBD.

Similar content being viewed by others

Data availability

All scRNA-seq datasets used in this study were obtained from published research articles. Previously published scRNA-seq are available under accession codes GSE114374 (ref. 3), GSE116222 (ref. 4), GSE148837 (ref. 5), GSE134809 (ref. 17), GSE125527 (ref. 22), GSE157477 (ref. 24), GSE121380 (validation cohort by B. Huang et al.)28, GSE158702 (ref. 58) (from the GEO database; https://www.ncbi.nlm.nih.gov/geo), SCP259 (ref. 16) (from the Single Cell Portal database; https://singlecell.broadinstitute.org/), E-MTAB-8901 (ref. 25), E-MTAB-9543 (adult tissues)27, E-MTAB-9536 (fetal tissues)27 (from the EBI array-express database; https://www.ebi.ac.uk/arrayexpress), HRA000072 (discovery cohort by B. Huang et al.28, from the GSA database; https://ngdc.cncb.ac.cn/gsa-human) and SDY176520 (from the Import database; https://www.immport.org). See Supplementary Table 1 for details. The datasets underlying scIBD can be accessed at the scIBD website or through Figshare59. The source data underlying Figs. 1 and 2, Extended Data Fig. 1e,f and Extended Data Figs. 28 and 10 are available in Figshare60. Source Data are provided with this paper.

Code availability

The code used to construct the scIBD website is stored in GitHub at https://github.com/niehu-szbl/scIBD_website_code and Figshare61 and running at http://scibd.cn. All other codes are deposited in GitHub at https://github.com/niehu-szbl/scIBD_SourceCode and Figshare62.

References

  1. Chang, J. T. Pathophysiology of inflammatory bowel diseases. N. Engl. J. Med. 383, 2652–2664 (2020).

    Article  Google Scholar 

  2. Graham, D. B. & Xavier, R. J. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 578, 527–539 (2020).

    Article  Google Scholar 

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

    Article  Google Scholar 

  4. Parikh, K. et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature 567, 49–55 (2019).

    Article  Google Scholar 

  5. Corridoni, D. et al. Single-cell atlas of colonic CD8+ T cells in ulcerative colitis. Nat. Med. 26, 1480–1490 (2020).

    Article  Google Scholar 

  6. de Lange, K. M. et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat. Genet. 49, 256–261 (2017).

    Article  Google Scholar 

  7. Huang, H. et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 547, 173–178 (2017).

    Article  Google Scholar 

  8. Jostins, L. et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    Article  Google Scholar 

  9. Liu, J. Z. et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 47, 979–986 (2015).

    Article  Google Scholar 

  10. Duerr, R. H. et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314, 1461–1463 (2006).

    Article  Google Scholar 

  11. Baumgart, D. C. & Le Berre, C. Newer biologic and small-molecule therapies for inflammatory bowel disease. N. Engl. J. Med. 385, 1302–1315 (2021).

    Article  Google Scholar 

  12. Mao, R. & Chen, M. Precision medicine in IBD: genes, drugs, bugs and omics. Nat. Rev. Gastroenterol. Hepatol. 19, 81–82 (2022).

    Article  Google Scholar 

  13. Massimino, L. et al. The inflammatory bowel disease transcriptome and metatranscriptome meta-analysis (IBD TaMMA) framework. Nat. Comput. Sci. 1, 511–515 (2021).

    Article  Google Scholar 

  14. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  Google Scholar 

  15. Papalexi, E. & Satija, R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat. Rev. Immunol. 18, 35–45 (2018).

    Article  Google Scholar 

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

    Article  Google Scholar 

  17. Martin, J. C. et al. Single-cell analysis of Crohn’s disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. Cell 178, 1493–1508 (2019).

    Article  Google Scholar 

  18. Uniken Venema, W. T. et al. Single-cell RNA sequencing of blood and ileal T cells from patients with Crohn’s disease reveals tissue-specific characteristics and drug targets. Gastroenterology 156, 812–815 (2019).

    Article  Google Scholar 

  19. Uzzan, M. et al. Ulcerative colitis is characterized by a plasmablast-skewed humoral response associated with disease activity. Nat. Med. 28, 766–779 (2022).

    Article  Google Scholar 

  20. 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  Google Scholar 

  21. Huang, L. J. et al. Multiomics analyses reveal a critical role of selenium in controlling T cell differentiation in Crohn’s disease. Immunity 54, 1728–1744 (2021).

    Article  Google Scholar 

  22. Boland, B. S. et al. Heterogeneity and clonal relationships of adaptive immune cells in ulcerative colitis revealed by single-cell analyses. Sci. Immunol. 5, eabb4432 (2020).

    Article  Google Scholar 

  23. Mitsialis, V. et al. Single-cell analyses of colon and blood reveal distinct immune cell signatures of ulcerative colitis and Crohn’s disease. Gastroenterology 159, 591–608 (2020).

    Article  Google Scholar 

  24. Jaeger, N. et al. Single-cell analyses of Crohn’s disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions. Nat. Commun. 12, 1921 (2021).

    Article  Google Scholar 

  25. Elmentaite, R. et al. Single-cell sequencing of developing human gut reveals transcriptional links to childhood Crohn’s disease. Dev. Cell 55, 771–783 (2020).

    Article  Google Scholar 

  26. Devlin, J. C. et al. Single-cell transcriptional survey of ileal-anal pouch immune cells from ulcerative colitis patients. Gastroenterology 160, 1679–1693 (2021).

    Article  Google Scholar 

  27. Elmentaite, R. et al. Cells of the human intestinal tract mapped across space and time. Nature 597, 250–255 (2021).

    Article  Google Scholar 

  28. Huang, B. et al. Mucosal profiling of pediatric-onset colitis and IBD reveals common pathogenics and therapeutic pathways. Cell 179, 1160–1176 (2019).

    Article  Google Scholar 

  29. Nie, H. et al. Single-Cell Meta-analysis of Inflammatory Bowel Disease with scIBD (Institute of Cancer Research, Shenzhen Bay Laboratory, 2022); http://scibd.cn

  30. Polański, K. et al. BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics 36, 964–965 (2020).

    Article  Google Scholar 

  31. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  Google Scholar 

  32. Laffitte, B. A. et al. LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proc. Natl. Acad. Sci. USA 98, 507–512 (2001).

    Article  Google Scholar 

  33. Liu, M. et al. Transcription factor c-Maf is a checkpoint that programs macrophages in lung cancer. J. Clin. Invest. 130, 2081–2096 (2020).

    Article  Google Scholar 

  34. Sweet, D. R., Fan, L. & Jain, M. K. Taking KLF9 to “Cort” for crimes against metabolism. J. Clin. Invest. 129, 2178–2180 (2019).

    Article  Google Scholar 

  35. MacFie, T. S. et al. DUOX2 and DUOXA2 form the predominant enzyme system capable of producing the reactive oxygen species H2O2 in active ulcerative colitis and are modulated by 5-aminosalicylic acid. Inflamm. Bowel Dis. 20, 514–524 (2014).

    Article  Google Scholar 

  36. de Bruyn, M. & Ferrante, M. Failure of MMP-9 antagonists in IBD: Demonstrating the importance of molecular biology and well-controlled early phase studies. J Crohns Colitis 12, 1011–1013 (2018).

    Article  Google Scholar 

  37. Li, Y. et al. COX-2-PGE(2) signaling impairs intestinal epithelial regeneration and associates with TNF inhibitor responsiveness in ulcerative colitis. EBioMedicine 36, 497–507 (2018).

    Article  Google Scholar 

  38. Koyama, M. et al. MHC class II antigen presentation by the intestinal epithelium initiates graft-versus-host disease and is influenced by the microbiota. Immunity 51, 885–898 (2019).

    Article  Google Scholar 

  39. Ntunzwenimana, J. C. et al. Functional screen of inflammatory bowel disease genes reveals key epithelial functions. Genome Med. 13, 181 (2021).

    Article  Google Scholar 

  40. Lotfollahi, M. et al. Mapping single-cell data to reference atlases by transfer learning. Nat. Biotechnol. 40, 121–130 (2021).

  41. Barrett, T. et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 41, D991–D995 (2013).

    Article  Google Scholar 

  42. CNCB-NGDC Members and Partners. Database resources of the national genomics data center, China national center for bioinformation in 2022. Nucleic Acids Res. 50, D27–D38 (2022).

  43. Papatheodorou, I. et al. Expression atlas update: from tissues to single cells. Nucleic Acids Res. 48, D77–D83 (2020).

    Google Scholar 

  44. Ghoussaini, M. et al. Open Targets Genetics: systematic identification of trait-associated genes using large-scale genetics and functional genomics. Nucleic Acids Res. 49, D1311–D1320 (2021).

    Article  Google Scholar 

  45. Mountjoy, E. et al. An open approach to systematically prioritize causal variants and genes at all published human GWAS trait-associated loci. Nat. Genet. 53, 1527–1533 (2021).

    Article  Google Scholar 

  46. 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 

  47. Zhang, C. et al. Single-cell RNA sequencing reveals intrahepatic and peripheral immune characteristics related to disease phases in HBV-infected patients. Gut 72, 153–167 (2022).

    Article  Google Scholar 

  48. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  Google Scholar 

  49. Wu, T. et al. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).

    Google Scholar 

  50. Shen, L. GeneOverlap: Test and Visualize Gene Overlaps (R package version 1.26.0, 2020); http://shenlab-sinai.github.io/shenlab-sinai/

  51. Van de Sande, B. et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nat. Protoc. 15, 2247–2276 (2020).

    Article  Google Scholar 

  52. Van Der Maaten, L. Accelerating t-SNE using tree-based algorithms. J. Mach. Learn. Res. 15, 3221–3245 (2014).

    MathSciNet  MATH  Google Scholar 

  53. Suo, S. et al. Revealing the critical regulators of cell identity in the mouse cell atlas. Cell Rep. 25, 1436–1445 (2018).

    Article  Google Scholar 

  54. Chang, W. et al. shiny Web Application Framework for R (CRAN, 2021); https://cran.r-project.org/web/packages/shiny/index.html

  55. Chang, W. et al. shinydashboard: Create Dashboards with ‘shiny’ (CRAN, 2018); https://cran.r-project.org/web/packages/shinydashboard/index.html

  56. Angerer, P. et al. anndata: ‘anndata’ for R (CRAN, 2023); https://cloud.r-project.org/web/packages/anndata/index.html

  57. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer-Verlag, 2009).

  58. Fawkner-Corbett, D. et al. Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell 184, 810–826 (2021).

    Article  Google Scholar 

  59. Nie, H. et al. scIBD Website Data (Figshare, 2023); https://doi.org/10.6084/m9.figshare.22316641

  60. Nie, H. et al. scIBD Source Data (Figshare, 2023); https://doi.org/10.6084/m9.figshare.22730543

  61. Nie, H. et al. scIBD Website Code (Figshare, 2023); https://doi.org/10.6084/m9.figshare.22731374

  62. Nie, H. et al. scIBD Source Code (Figshare, 2023); https://doi.org/10.6084/m9.figshare.22731620

Download references

Acknowledgements

We thank the National Natural Science Foundation of China (grant nos. 82122049, 82272803 and 82341011 to L.Z.; grant no. 32100579 to C.Y.), the National Key Research and Development Program of China (grant no. 2022YFA0912600 to L.Z.) and the Guangdong Basic and Applied Basic Research Foundation (2020A1515110857 to C.Y.) for their support. We thank the Shenzhen Bay Laboratory supercomputing center.

Author information

Authors and Affiliations

Authors

Contributions

H.N. and L.Z. provided conceptualization and wrote the original draft. H.N. performed the data search and acquisition. H.N., P.L. and J.L. contributed to bioinformatics analysis and platform development, and wrote the guide. H.N., P.L., Y.Z., Y.W., J.L., C.Y. and L.Z. reviewed and edited the manuscript. C.Y. and L.Z. provided supervision and resources and performed funding acquisition.

Corresponding author

Correspondence to Lei Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Computational Science thanks Tamas Korcsmaros and Ali Rahnavard for their contribution to the peer review of this work. Primary Handling Editor: Fernando Chirigati, in collaboration with the Nature Computational Science team. Peer reviewer reports are available.

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 Overview of resources, methodologies, and utilities of scIBD.

a, Summary of scRNA-seq datasets used in this study. All single-cell transcriptomic datasets were retrieved from public databases, including SCP, Immport, SRA, GSA, and EBI. Clinical trials, therapeutic drugs, therapeutic targets, and GWAS-risk genes for IBD were also curated in scIBD. b, In-house workflow to pre-process, integrate, and analyze these scRNA-seq datasets uniformly. c, Utilities of scIBD. scIBD provides various interactive visualization and data analysis interface. d, Method to classify T/NK compartment into three sub-compartments: CD4+ T cell, CD8+ T cell, and ILC compartment. e, Expression levels of IGHA and IGHG genes in non-circulating plasma cells. f, Percentage of IgA+, IgG+, IgA-IgG-, and IgA+IgG+ plasma cells among non-circulating plasma cells. An expression cutoff corresponding to log2(CP10K + 1) = 6 accurately discriminated between IgA+ and IgG+ plasma cells.

Source data

Extended Data Fig. 2 Cellular composition and distribution in scIBD across different developmental stages and datasets.

a, Percentage of cells in each major cluster in scIBD. b, Percentage of cells in each major cluster across fetal, pediatric, and adult stages. c,d, The cellular distribution colored by developmental stage (c) and dataset (d). e, Cell composition of CD4+ T cells, CD8+ T cells, myeloid cells, ILCs, B/plasma cells, epithelial cells, and mesenchymal cells in pediatric and adult individuals.

Source data

Extended Data Fig. 3 Batch correction of scRNA-seq data with BBKNN improved the integration of CD8+ T cells and epithelial cells.

a,b UMAP plots showing the distribution of datasets and cell subtypes before and after batch correction by BBKNN in CD8+ T cells (n = 184,220) (a) and epithelial cells (n = 195,735) (b). c, Boxplot showing the Local Inverse Simpson’s Index (LISI) distribution of data presented in a and b. In the boxplot, center lines denote median values; whiskers denote 1.5 x the interquartile range; the upper and lower limits of the box plot indicate the maximum and minimum values, respectively. The P-values were calculated by two-sided Wilcoxon rank-sum test. P-values < 2.2 × 10−16 for all comparisons. **** indicates P-value < 0.0001. d, UMAP plots showing the cell subtypes in each major cluster.

Extended Data Fig. 4 Examination of rare or less characterized cell subtypes in multiple datasets in scIBD.

ac, Number of LAMP3+ DCs (a), CD8+ Tc17 cells (b) and DUOX2+ epithelial cells (c) in individual datasets. d, Gene expression of LAMP3 and CCR7 in each cell subtype of myeloid cells in the dataset C. S. Smillie et al., Cell, 2019. e, Gene expression of IL17A, IL22, IL26 and RORC in each cell subtype of CD8+ T cells in the dataset C. S. Smillie et al., Cell, 2019. f, Gene expression levels of DUOX2 and DUOXA2 across all cell subtypes of epithelial cells in the dataset C. S. Smillie et al., Cell, 2019. g, Gene expression levels of DUOX2 and DUOXA2 in enterocytes in healthy (n = 3,994), non-inflamed (n = 2,928), and inflamed (n = 2,112) tissues in the dataset C. S. Smillie et al., Cell, 2019. P-values < 2.2 × 10−16 for all comparisons. **** indicates P-value < 0.0001. Two-sided Wilcoxon rank-sum test.

Source data

Extended Data Fig. 5 Preference of disease status for immune cell subtypes.

a,b, Tissue preference (a) and disease status preference (b) of each immune cluster in adult individuals estimated by the Ro/e. ++++, Ro/e > 2; ++, 1 < Ro/e ≤ 2; +, 0.5 ≤ Ro/e ≤ 1; +/−, 0 < Ro/e < 0.5; −, Ro/e = 0; in which Ro/e denotes the ratio of observed to expected cell number. c, Volcano plot showing differentially expressed genes between CD8+ Tc17 cells (n = 5,258) and CD4+ Th17 cells (n = 5,937). Two-sided Wilcoxon rank-sum test; fold change ≥ 1.25. d, Dot plot showing gene expression of transcriptional factors including MAF, NR1H3, ETV5, KLF9 in myeloid cells.

Source data

Extended Data Fig. 6 Preference of disease status for non-immune cell subtypes.

ac, Disease status preference of each cell subtype in epithelial (a), mesenchymal (b) and endothelial (c) compartments in adult individuals estimated by the Ro/e. ++++, Ro/e > 2; ++, 1 < Ro/e ≤ 2; +, 0.5 ≤ Ro/e ≤ 1; +/−, 0 < Ro/e < 0.5; −, Ro/e = 0; in which Ro/e denotes the ratio of observed to expected cell number. d, Frequencies of disease-associated cell subsets in each major cluster for each sample. Each dot represents a sample. There were n1, n2, and n3 samples present in each major cluster with a cell count greater than 100 in the CD, HC, and UC groups, respectively. For epithelial cells, n2 = 69, n3 = 34; For mesenchymal cells, n1 = 2, n2 = 39, n3 = 29; For endothelial cells, n1 = 2, n2 = 16, n3 = 17. In the box plots, center lines denote median values; whiskers denote 1.5 x the interquartile range; the upper and lower limits of the box plot indicate the maximum and minimum values, respectively. P-values for HC versus UC were 3.76 × 10−6, 0.028 in DUOX2+ epithelial cell and M-like cell, respectively. P-values for HC versus CD, HC versus UC, and CD versus UC were 0.250, 5.07 × 10−3, 0.340 in inflammatory fibroblast; 0.277, 0.041, 0.027 in reticular fibroblast; 0.227, 1.51 × 10−3, 0.461 in immature pericyte; 0.341, 1.97 × 10−5, 0.083 in adult venous EC (SELE + ), 0.280, 2.89 × 10−3, 0.307 in cycling EC, respectively. ns, not significant; * < 0.05, ** < 0.01, *** < 0.001, **** < 0.0001; One-sided t-test. e, Dot plots showing the signature genes of the above disease-associated cell subsets.

Source data

Extended Data Fig. 7 Major cluster-specific risk genes identified in scIBD.

a, Heatmap of gene expression levels of major cluster-specific risk genes of IBD. The expression value across each cell was scaled to a range from 0 to 1 with the function “rescale” in package “scales” in R. b, Violin plot of minor cluster-specific risk genes which were up-regulated in CD or UC comparing with health. The P-values were calculated by two-sided Wilcox rank-sum test. The number of cells in each group was displayed below the violin plot. P-values for HC versus UC were 1.71 × 10-4, 9.88 × 10−4, 0.016 in the first three comparisons, respectively. P-values for HC versus CD, HC versus UC, and CD versus UC were 4.43 × 10−16, 1.93 × 10201, 6.33 × 10−4; 1.07 × 10−3, 0.629, 0.074; 8.30 × 10−22, 5.04 × 10−5, 4.14 × 10−11; 7.70 × 10−8, 1.10 × 10−29, 2.49 × 10−10; 8.16 × 10−7, 4.29 × 10−5, 0.164, respectively. ns, not significant; * < 0.05, ** < 0.01, *** < 0.001, **** < 0.0001; Two-sided Wilcox rank-sum test.

Source data

Extended Data Fig. 8 Cell subtype-specific therapeutic target genes identified in scIBD.

a, Heatmap of gene expression of cell subtype-specific therapeutic target genes for IBD. The expression value across each cell subtype was scaled to a range from 0 to 1 with function “rescale” in package “scales” in R. b, Violin plot of cell subtype-specific therapeutic target genes which were up-regulated in UC or CD comparing with health. The number of cells in each group was displayed below the violin plot. P-values for HC versus CD, HC versus UC, and CD versus UC were 0.016, 0.418, 9.86 × 10−4; 4.32 × 10−38, 5.16 × 10−61, 9.20 × 10−5; 1.92 × 10−12, 7.09 × 10−11, 8.23 × 10−7; 0.010, 0.506, 5.37 × 10−3, respectively. P-values for HC versus UC were 0.038, 4.23 × 10−13 for the last two comparisons, respectively. ns, not significant; * < 0.05, ** < 0.01, *** < 0.001, **** < 0.0001; Two-sided Wilcox rank-sum test.

Source data

Extended Data Fig. 9 Characterize the features of scIBD.

scIBD scRNA-seq data were gathered from 12 studies. In addition to traditional analysis, regulon inference was also conducted. To facilitate the visualization and exploration of the meta-analysis results, a user-friendly web platform for scIBD was created, allowing users to browse, search, analyze online, and download all relevant metadata and analytical results.

Extended Data Fig. 10 Two examples to demonstrate the generality of this web resource.

a, Dot plot showing the gene expression levels of MHC II molecules across the cell subtypes of epithelial cells. b, Global UMAP visualization of gene expression of MHC II molecules in scIBD. The expression level of MHC II molecules was calculated by the average expression of HLA-DRA and HLA-DRB1. c, Comparison of the gene expression of MHC II molecules across cell subtypes in the epithelial compartment. d, Comparison of gene expression of MHC II molecules in DUOX2+ epithelial cells (left) and enterocytes (right) across disease statuses. e,f, Differential regulons between healthy (n = 63,411 cells) and UC (n = 19,339 cells) (e), and between healthy and CD (n = 1,123 cells) (f) in epithelial compartment. The horizontal dashed line represents the adjusted-P value threshold of 0.01. The vertical dashed lines represent fold change thresholds of +1.5 or –1.5. The P-values were calculated by two-sided Student’s t-test, then adjusted by the ‘Benjamini & Hochberg’ method. g, Comparison of the activity of PITX1 regulon among cell subtypes in epithelial compartment. h, Comparison of the activity of PITX1 regulon across disease statuses in DUOX2+ epithelial cells from adult individuals. i, Comparison of the activity of PITX1 regulon between colon and rectum in DUOX2+ epithelial cells in inflamed tissues from adult UC patients.

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

Summary of scRNA-seq datasets used in this study.

Supplementary Table 2

Differential gene expression analysis of Tc17 cells and Th17 cells.

Supplementary Table 3

Differential gene expression analysis of Tc17 cells in UC and CD.

Supplementary Table 4

Differential gene expression analysis of AREG+ RTMs and APOE+ RTMs.

Supplementary Table 5

The specificity scores of regulons of APOE+ RTMs and AREG+ RTMs.

Supplementary Table 6

Differential regulon activity analysis of epithelial cells in different disease statuses.

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

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. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

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

Nie, H., Lin, P., Zhang, Y. et al. Single-cell meta-analysis of inflammatory bowel disease with scIBD. Nat Comput Sci 3, 522–531 (2023). https://doi.org/10.1038/s43588-023-00464-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43588-023-00464-9

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