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:

Single-cell transcriptomic analyses reveal distinct immune cell contributions to epithelial barrier dysfunction in checkpoint inhibitor colitis

Abstract

Immune checkpoint inhibitor (ICI) therapy has revolutionized oncology, but treatments are limited by immune-related adverse events, including checkpoint inhibitor colitis (irColitis). Little is understood about the pathogenic mechanisms driving irColitis, which does not readily occur in model organisms, such as mice. To define molecular drivers of irColitis, we used single-cell multi-omics to profile approximately 300,000 cells from the colon mucosa and blood of 13 patients with cancer who developed irColitis (nine on anti-PD-1 or anti-CTLA-4 monotherapy and four on dual ICI therapy; most patients had skin or lung cancer), eight controls on ICI therapy and eight healthy controls. Patients with irColitis showed expanded mucosal Tregs, ITGAEHi CD8 tissue-resident memory T cells expressing CXCL13 and Th17 gene programs and recirculating ITGB2Hi CD8 T cells. Cytotoxic GNLYHi CD4 T cells, recirculating ITGB2Hi CD8 T cells and endothelial cells expressing hypoxia gene programs were further expanded in colitis associated with anti-PD-1/CTLA-4 therapy compared to anti-PD-1 therapy. Luminal epithelial cells in patients with irColitis expressed PCSK9, PD-L1 and interferon-induced signatures associated with apoptosis, increased cell turnover and malabsorption. Together, these data suggest roles for circulating T cells and epithelial–immune crosstalk critical to PD-1/CTLA-4-dependent tolerance and barrier function and identify potential therapeutic targets for irColitis.

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: Overview of checkpoint colitis cohort.
Fig. 2: Expanded ITGAEHi tissue-resident memory and ITGB2Hi CD8 T cell subsets in irColitis.
Fig. 3: IL26, IL17A and CXCL13 are upregulated in colon mucosal ITGAEHi CD8 T cells in irColitis.
Fig. 4: Expanded IL17A and CXCL13-expressing CD4 T cell effectors and TNFRSF4-expressing Tregs in patients with irColitis.
Fig. 5: Epithelial and mesenchymal remodeling in irColitis.
Fig. 6: Identification of putative therapeutic targets to treat irColitis and comparison of irColitis to diverse tumor immune microenvironments.

Similar content being viewed by others

Data availability

The scRNA-seq and snRNA-seq data and results are available for browsing at https://villani.mgh.harvard.edu/ircolitis. The same processed data files are available on the Gene Expression Omnibus (GSE206301) and Zenodo (https://zenodo.org/doi/10.5281/zenodo.8088435). Raw human sequencing data are available at the database of Genotypes and Phenotypes (dbGaP) (phs003418.v1.p1). The dbGaP provides authorized access to sequencing data that requires a formal request be made to the appropriate National Institutes of Health Data Access Committee.

Further information and requests for resources and reagents should be directed to, and will be fulfilled by, the lead contact, A.-C.V. (avillani@mgh.harvard.edu).

Code availability

Source code for data analysis and the website is available on GitHub (https://github.com/villani-lab/ircolitis) and has been archived at Zenodo (https://zenodo.org/doi/10.5281/zenodo.8088435).

References

  1. Wolchok, J. D. et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 377, 1345–1356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Arnaud-Coffin, P. et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int. J. Cancer 145, 639–648 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Chen, J. H., Pezhouh, M. K., Lauwers, G. Y. & Masia, R. Histopathologic features of colitis due to immunotherapy with anti-PD-1 antibodies. Am. J. Surg. Pathol. 41, 643–654 (2017).

    Article  PubMed  Google Scholar 

  4. Verschuren, E. C. et al. Clinical, endoscopic, and histologic characteristics of ipilimumab-associated colitis. Clin. Gastroenterol. Hepatol. 14, 836–842 (2016).

    Article  PubMed  Google Scholar 

  5. Curran, M. A., Montalvo, W., Yagita, H. & Allison, J. P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl Acad. Sci. USA 107, 4275–4280 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Heul, A. V. & Stappenbeck, T. Establishing a mouse model of checkpoint inhibitor-induced colitis: pilot experiments and future directions. J. Allergy Clin. Immunol. 141, AB119 (2018).

    Article  Google Scholar 

  7. Perez-Ruiz, E. et al. Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy. Nature 569, 428–432 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Callahan, M. K. et al. Evaluation of serum IL-17 levels during ipilimumab therapy: correlation with colitis. J. Clin. Oncol. 29, 2505 (2011).

    Article  Google Scholar 

  9. Shahabi, V. et al. Gene expression profiling of whole blood in ipilimumab-treated patients for identification of potential biomarkers of immune-related gastrointestinal adverse events. J. Transl. Med. 11, 75 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Das, R. et al. Early B cell changes predict autoimmunity following combination immune checkpoint blockade. J. Clin. Invest. 128, 715–720 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Chaput, N. et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 28, 1368–1379 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Oh, D. Y. et al. Immune toxicities elicited by CTLA-4 blockade in cancer patients are associated with early diversification of the T-cell repertoire. Cancer Res. 77, 1322–1330 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Sasson, S. C. et al. Mucosal-associated invariant T (MAIT) cells are activated in the gastrointestinal tissue of patients with combination ipilimumab and nivolumab therapy-related colitis in a pathology distinct from ulcerative colitis. Clin. Exp. Immunol. 202, 335–352 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sasson, S. C. et al. Interferon-gamma–producing CD8+ tissue resident memory T cells are a targetable hallmark of immune checkpoint inhibitor–colitis. Gastroenterology 161, 1229–1244 (2021).

    Article  CAS  PubMed  Google Scholar 

  15. Luoma, A. M. et al. Molecular pathways of colon inflammation induced by cancer immunotherapy. Cell 182, 655–671 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Brahmer, J. R. et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. J. Immunother. Cancer 9, e002435 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Thompson, J. A. et al. NCCN Guidelines Insights: management of immunotherapy-related toxicities, version 1.2020. J. Natl Compr. Cancer Netw. 18, 230–241 (2020).

    Article  CAS  Google Scholar 

  18. Faje, A. T. et al. High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma. Cancer 124, 3706–3714 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Bai, X. et al. Early use of high-dose glucocorticoid for the management of irAE is associated with poorer survival in patients with advanced melanoma treated with anti-PD-1 monotherapy. Clin. Cancer Res. 27, 5993–6000 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. FitzPatrick, M. E. B. et al. Human intestinal tissue-resident memory T cells comprise transcriptionally and functionally distinct subsets. Cell Rep. 34, 108661 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Walling, B. L. & Kim, M. LFA-1 in T cell migration and differentiation. Front. Immunol. 9, 952 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Singer, M. et al. A distinct gene module for dysfunction uncoupled from activation in tumor-infiltrating T cells. Cell 166, 1500–1511 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Buggert, M. et al. The identity of human tissue-emigrant CD8+ T cells. Cell 183, 1946–1961 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hu, W., Wang, Y., Fang, Z., He, W. & Li, S. Integrated characterization of lncRNA-immune interactions in prostate cancer. Front. Cell Dev. Biol. 9, 641891 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Yost, K. E. et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 25, 1251–1259 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Giles, J. R. et al. Human epigenetic and transcriptional T cell differentiation atlas for identifying functional T cell-specific enhancers. Immunity 55, 557–574 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gerlach, C. et al. The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity 45, 1270–1284 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Abu-Sbeih, H. et al. Outcomes of vedolizumab therapy in patients with immune checkpoint inhibitor–induced colitis: a multi-center study. J. Immunother. Cancer 6, 142 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Kim, Y. U., Kee, P., Danila, D. & Teng, B.-B. A critical role of PCSK9 in mediating IL-17-producing T cell responses in hyperlipidemia. Immune Netw. 19, e41 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Deng, Z., Wang, S., Wu, C. & Wang, C. IL-17 inhibitor-associated inflammatory bowel disease: A study based on literature and database analysis. Front. Pharmacol. 14, 1124628 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rao, D. A. et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 542, 110–114 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Shimizu, J., Yamazaki, S., Takahashi, T., Ishida, Y. & Sakaguchi, S. Stimulation of CD25+CD4+ regulatory T cells through GITR breaks immunological self-tolerance. Nat. Immunol. 3, 135–142 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Vu, M. D. et al. OX40 costimulation turns off Foxp3+ Tregs. Blood 110, 2501–2510 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Oh, D. Y. & Fong, L. Cytotoxic CD4+ T cells in cancer: expanding the immune effector toolbox. Immunity 54, 2701–2711 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Menzel, K. et al. Cathepsins B, L and D in inflammatory bowel disease macrophages and potential therapeutic effects of cathepsin inhibition in vivo. Clin. Exp. Immunol. 146, 169–180 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Baklien, K. & Brandtzaeg, P. Comparative mapping of the local distribution of immunoglobulin-containing cells in ulcerative colitis and Crohn’s disease of the colon. Clin. Exp. Immunol. 22, 197–209 (1975).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Li, J. et al. KIR+CD8+ T cells suppress pathogenic T cells and are active in autoimmune diseases and COVID-19. Science 376, eabi9591 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chung, H. & Rice, C. M. T time for ADAR: ADAR1 is required for T cell self‐tolerance. EMBO Rep. 19, e47237 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Yi, W. et al. Targeted regulation of self-peptide presentation prevents type I diabetes in mice without disrupting general immunocompetence. J. Clin. Invest. 120, 1324–1336 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  43. Leung, E. et al. Polymorphisms in the organic cation transporter genes SLC22A4 and SLC22A5 and Crohn’s disease in a New Zealand Caucasian cohort. Immunol. Cell Biol. 84, 233–236 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Serrano León, A. et al. Single-nucleotide polymorphisms in SLC22A23 are associated with ulcerative colitis in a Canadian white cohort. Am. J. Clin. Nutr. 100, 289–294 (2014).

    Article  PubMed  Google Scholar 

  45. Dicay, M. S. et al. Interferon-γ suppresses intestinal epithelial aquaporin-1 expression via Janus kinase and STAT3 activation. PLoS ONE 10, e0118713 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Kim, J. et al. Constitutive and inducible expression of B7 family of ligands by human airway epithelial cells. Am. J. Respir. Cell Mol. Biol. 33, 280–289 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Xie, J. et al. Slit2/Robo1 mitigates DSS-induced ulcerative colitis by activating autophagy in intestinal stem cell. Int. J. Biol. Sci. 16, 1876–1887 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sharpe, A. H. & Pauken, K. E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 18, 153–167 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Türei, D. et al. Integrated intra- and intercellular signaling knowledge for multicellular omics analysis. Mol. Syst. Biol. 17, e9923 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kim, J., Kim, M.-G., Jeong, S. H., Kim, H. J. & Son, S. W. STAT3 maintains skin barrier integrity by modulating SPINK5 and KLK5 expression in keratinocytes. Exp. Dermatol. 31, 223–232 (2022).

    Article  CAS  PubMed  Google Scholar 

  51. Matsuoka, K. et al. DOP056 Efficacy and safety of anti-fractalkine monoclonal antibody, E6011, in patients with Crohn’s disease who had lost response to anti-TNFα agents: a multicentre, open-label, phase 1/2 study. J. Crohns Colitis 12, S070 (2018).

    Article  Google Scholar 

  52. Xia, X. et al. Regulation of PCSK9 expression and function: mechanisms and therapeutic implications. Front. Cardiovasc. Med. 8, 764038 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yang, W. et al. Potentiating the antitumour response of CD8+ T cells by modulating cholesterol metabolism. Nature 531, 651–655 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lei, L. et al. Inhibition of proprotein convertase subtilisin/kexin type 9 attenuates 2,4,6-trinitrobenzenesulfonic acid-induced colitis via repressing toll-like receptor 4/nuclear factor-kappa B. Kaohsiung J. Med. Sci. 36, 705–711 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Zheng, L. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021).

    Article  PubMed  Google Scholar 

  56. Hueber, W. et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 61, 1693–1700 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  58. Buchan, S. L. et al. Antibodies to costimulatory receptor 4-1BB enhance anti-tumor immunity via T regulatory cell depletion and promotion of CD8 T cell effector function. Immunity 49, 958–970 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Riudavets, M. et al. Correlation between immune-related adverse events (irAEs) and efficacy in patients with solid tumors treated with immune-checkpoints inhibitors (ICIs). J. Clin. Oncol. 36, 3064 (2018).

    Article  Google Scholar 

  60. Crotty, S. Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 29, 621–663 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Veatch, J. R. et al. Neoantigen-specific CD4+ T cells in human melanoma have diverse differentiation states and correlate with CD8+ T cell, macrophage, and B cell function. Cancer Cell 40, 393–409 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Yang, M. et al. CXCL13 shapes immunoactive tumor microenvironment and enhances the efficacy of PD-1 checkpoint blockade in high-grade serous ovarian cancer. J. Immunother. Cancer 9, e001136 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Song, X. et al. Growth factor FGF2 cooperates with interleukin-17 to repair intestinal epithelial damage. Immunity 43, 488–501 (2015).

    Article  CAS  PubMed  Google Scholar 

  65. Lee, J. S. et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity 43, 727–738 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Song, X., He, X., Li, X. & Qian, Y. The roles and functional mechanisms of interleukin-17 family cytokines in mucosal immunity. Cell. Mol. Immunol. 13, 418–431 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Dubin, K. et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat. Commun. 7, 10391 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Baruch, E. N. et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 371, 602–609 (2021).

    Article  CAS  PubMed  Google Scholar 

  69. 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  CAS  PubMed  PubMed Central  Google Scholar 

  70. Böttcher, J. P. et al. Functional classification of memory CD8+ T cells by CX3CR1 expression. Nat. Commun. 6, 8306 (2015).

    Article  PubMed  Google Scholar 

  71. Yamauchi, T. et al. T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors. Nat. Commun. 12, 1402 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Ricanek, P. et al. Reduced expression of aquaporins in human intestinal mucosa in early stage inflammatory bowel disease. Clin. Exp. Gastroenterol. 8, 49–67 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Leber, A. et al. Activation of NLRX1 by NX-13 alleviates inflammatory bowel disease through immunometabolic mechanisms in CD4+ T cells. J. Immunol. 203, 3407–3415 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Lee, G. et al. P120 BBT-401 is a selective Pellino-1 protein–protein interaction inhibitor in clinical development targeting a first-in-class drug for UC treatment. Inflamm. Bowel Dis. 25, S58 (2019).

    Article  Google Scholar 

  75. Kim, Y.-I. et al. Local stabilization of hypoxia-inducible factor-1α controls intestinal inflammation via enhanced gut barrier function and immune regulation. Front. Immunol. 11, 609689 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Geukes Foppen, M. H. et al. Immune checkpoint inhibition-related colitis: symptoms, endoscopic features, histology and response to management. ESMO Open 3, e000278 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Wright, A. P., Piper, M. S., Bishu, S. & Stidham, R. W. Systematic review and case series: flexible sigmoidoscopy identifies most cases of checkpoint inhibitor-induced colitis. Aliment. Pharmacol. Ther. 49, 1474–1483 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Adam, M., Potter, A. S. & Potter, S. S. Psychrophilic proteases dramatically reduce single-cell RNA-seq artifacts: a molecular atlas of kidney development. Development 144, 3625–3632 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Drokhlyansky, E. et al. The human and mouse enteric nervous system at single-cell resolution. Cell 182, 1606–1622 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Methodol. 57, 289–300 (1995).

    Article  Google Scholar 

  82. Li, B. et al. Cumulus provides cloud-based data analysis for large-scale single-cell and single-nucleus RNA-seq. Nat. Methods 17, 793–798 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Gaublomme, J. T. et al. Nuclei multiplexing with barcoded antibodies for single-nucleus genomics. Nat. Commun. 10, 2907 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Batson, J., Royer, L. & Webber, J. Molecular cross-validation for single-cell RNA-seq. Preprint at bioRxiv https://doi.org/10.1101/786269 (2019).

  85. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Traag, V., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at arXiv https://doi.org/10.48550/arXiv.1802.03426 (2020).

  88. Lun, A. T., Bach, K. & Marioni, J. C. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75 (2016).

    Article  PubMed  Google Scholar 

  89. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  90. James, K. R. et al. Distinct microbial and immune niches of the human colon. Nat. Immunol. 21, 343–353 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. 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  PubMed  PubMed Central  Google Scholar 

  92. Gupta, N. T. et al. Change-O: a toolkit for analyzing large-scale B cell immunoglobulin repertoire sequencing data. Bioinformatics 31, 3356–3358 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Bates, D., Mächler, M., Bolker, B. & Walker, S. Fitting linear mixed-effects models using lme4. J. Stat. Softw. 67, 1–48 (2015).

Download references

Acknowledgements

We are deeply grateful to all donors and their families as well as to the Severe Immunotherapy Complications service at Massachusetts General Hospital. We acknowledge the contributions of E. Drokhlyansky, R. Xavier, M. Biton and Hacohen laboratory members for their feedback on experimental design and data interpretation. We thank C. Flayer and K. Gallagher for help with generating Luminex data. This work was supported by several training grants, including National Institute of Allergy and Infectious Diseases grant T32AR007258 (to K.S.); National Heart, Lung, and Blood Institute grant K08HL157725; an American Heart Association Career Development Award (to P.S.); two National Institute of Diabetes and Digestive and Kidney Diseases training grants (1K08DK127246-01A1 and T32DK007191 (to M.F.T)); a Spanish Society of Medical Oncology grant (to L.Z.); 1T32CA207021 (to J.H.C.); 1K08CA273547-01A1 (to J.H.C.); Massachusetts General Hospital Fund for Medical Discovery (to J.H.C. and M.F.T.); Massachusetts General Hospital Krantz Stewardship (to J.H.C.); Society for Immunotherapy of Cancer/AstraZeneca Forward Fund (to J.H.C.); and National Cancer Institute R00CA259511 (to K.P.). This work was made possible by generous support from the National Institutes of Health Director’s New Innovator Award (DP2CA247831 to A.-C.V.); the Massachusetts General Hospital Transformative Scholar in Medicine Award (to A.-C.V.); the Damon Runyon-Rachleff Innovation Award (to A.-C.V.); the Melanoma Research Alliance Young Investigator Award (https://doi.org/10.48050/pc.gr.143739 to A.-C.V.); a Kraft Foundation award (to K.L.R. and A.-C.V.); the Arthur, Sandra, and Sarah Irving Fund for Gastrointestinal Immuno-Oncology (to J.H.C., N.H. and A.-C.V.); Gordon Pugh (to K.L.R.); the Adelson Foundation (to G.M.B.); R01AI169188-01 (to M.D.); the Fariborz Maseeh Award for Innovative Medical Education (to M.D.); the Peter and Ann Lambertus Family Foundation (to M.D. and R.J.S.); Merck (to R.J.S.); and the American College of Gastroenterology Clinical Research Award and R01AG068390 (to H.K.). This work was also made possible by the generous support of an anonymous donor (to K.L.R. and A.-C.V.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

M.F.T. and A.-C.V. conceived of and led the study. M.F.T. and A.-C.V. led experimental design. M.F.T. carried out experiments, with assistance from K.M., J.T., P.S., M.N., A.T. and B.Y.A. K.S. designed and performed computational analysis, with input and assistance from B.L., M.N., N.S. and S.R. M.F.T., L.T.N. and J.H.C. designed and performed microscopy experiments, with input and assistance from K.H.X., Y.S. and V.J. T.E. and K.P. provided input for scRNA-seq/snRNA-seq experiments, protocols and data interpretation. M.F.T., L.Z., C.J.P., T.S., R.G., P.Y.C., R.J.S., D.J., G.M.B., H.K., K.L.R. and M.D. provided clinical expertise and coordinated and performed sample acquisition and/or administrative coordination. M.F.T. and M.D. performed endoscopic examinations. M.D. provided additional expertise on study design. N.H. contributed to biological expertise and study design and provided advice. B.L. contributed to computational expertise and provided advice. A.-C.V. managed and supervised the study. A.-C.V. and K.L.R. raised funding for this work. M.F.T., K.S. and A.-C.V. wrote the paper, with input from all authors.

Corresponding authors

Correspondence to Molly Fisher Thomas, Kamil Slowikowski or Alexandra-Chloé Villani.

Ethics declarations

Competing interests

From 9 August 2021, B.L. is an employee of Genentech. M.D. has received consulting fees from Genentech, Partner Therapeutics, SQZ Biotech, AzurRx, Eli Lilly, Mallinckrodt Pharmaceuticals, Aditum, Foghorn Therapeutics, Palleon, Sorriso Pharmaceuticals, Generate Biomedicines, Asher Bio, Neoleukin Therapeutics, Moderna, Alloy Therapeutics, Third Rock Ventures, DE Shaw Research, Agenus and Curie Bio. M.D. is also a member of the scientific advisory board for Veravas, Monod Bio, Axxis Bio and Cerberus Therapeutics. R.J.S. is a consultant for Bristol Myers Squibb, Marengo, Merck, Novartis, Pfizer and Replimune. H.K. received research funding from Pfizer and Takeda. H.K. received consulting fees from Aditium Bio, AbbVie and Takeda. H.K. serves on the scientific advisory board of Vivante Health. D.J. reports grants and personal fees from Novartis, Genentech, Syros and Eisai. D.J. reports personal fees from Vibliome, PIC Therapeutics, Mapkure and Relay Therapeutics. D.J. reports grants from Pfizer, Amgen, InventisBio, Arvinas, Takeda, Blueprint Medicines, AstraZeneca, Ribon Therapeutics and Infinity that are outside the submitted work. G.M.B. has sponsored research agreements through her institution with Olink Proteomics, Teiko Bio, InterVenn Biosciences and Palleon Pharmaceuticals. G.M.B. served on advisory boards for Iovance, Merck, Nektar Therapeutics, Novartis and Ankyra Therapeutics. G.M.B. consults for Merck, InterVenn Biosciences, Iovance and Ankyra Therapeutics. She holds equity in Ankyra Therapeutics. K.P. is a consultant for Santa Ana Bio. N.H. holds equity in and advises Danger Bio/Related Sciences, is on the scientific advisory board of Repertoire Immune Medicines and CytoReason, owns equity in and has licensed patents to BioNTech and receives research funding from Bristol Myers Squibb and Calico Life Sciences. K.L.R. has received advisory board fees from SAGA Diagnostics and institutional research support from Bristol Myers Squibb. A.-C.V. received consulting fees from Merck and Bristol Myers Squibb. A.-C.V. has a financial interest in 10x Genomics. The company designs and manufactures gene sequencing technology for use in research, and such technology is being used in this research. A-.C.V.’s interests were reviewed by Massachusetts General Hospital and Mass General Brigham in accordance with their institutional policies. M.F.T., K.S., K.M., P.S., N.S., J.T., M.N., L.Z., N.P.S., A.T., S.R., B.Y.A., L.T.N., J.H.C., T.E., Y.S., K.H.X., V.J., C.J.P., T.S., R.G. and P.Y.C. do not have competing interests to declare.

Peer review

Peer review information

Nature Medicine thanks Douglas Johnson, Barbara Maier and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Anna Maria Ranzoni, in collaboration with the Nature Medicine 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 Detailed cell type composition of each donor for immune cells from colon tissue.

(a) Estimated number of cells per biopsy in each CD8 T/GDT/NK cell cluster for cases (n = 5) and controls (n = 7). (b) Detailed composition of each patient across cell clusters for CD8 T/GDT/NK cells. (c) Estimated number of cells per biopsy in each CD4 T cell cluster for cases (n = 5) and controls (n = 7). (d) Detailed composition of each patient across cell clusters for CD4 T cells. Cell clusters depicted with different colors in UMAP embeddings of the (e) eight mononuclear phagocyte subsets and (g) 13 B cell subsets from colon tissue. Detailed composition of each patient across cell clusters for (f) mononuclear phagocytes and (h) B cells from colon tissue. For panels B, D, F, and H, every column represents one individual donor referenced, labeled at the bottom of the heatmap. Boxplots show median and interquartile range. Heatmap color indicates percent of a patient’s cells assigned to each cell cluster. NK natural killer. GDT gamma delta T cells. Related to Figs. 24.

Extended Data Fig. 2 Tissue cell type abundance associated with control patients on anti-PD-1 versus no therapy.

(a) Abundance analysis of tissue cell clusters in control patients on anti-PD-1 therapy (n = 4) versus healthy controls not on ICI therapy (n = 8) (that is, those undergoing screening colonoscopy). Each dot represents a patient. Composition of each donor is reported as a percent of cells from each patient in each cluster, and box plots show median and interquartile range. Error-bars indicate logistic regression OR 95% CI for differential abundance of cells from controls on anti-PD-1 versus healthy controls not on ICI therapy for each cell cluster. Unadjusted likelihood ratio test p-values are shown. MNP cell cluster 8 was detected in only one patient in the control anti-PD-1 group, so the logistic regression model was not fit for this cluster. (b) Volcano plots show log fold-change (x-axis) and negative log10 P-value (two-sided) (y-axis) for each gene. Number of differentially expressed genes (FC > 1.5 and FDR < 10%) is shown, and some of the top genes are labeled. ADAR expression is shown for cluster 9 CD4 Tregs. Dots represent individual patients. Feature plots for HLA-DOB and ATP11A expression in MP cells is shown. (c) Number of differentially expressed genes in each cell cluster (FC > 1.5 and FDR < 10%). Related to Fig. 1.

Extended Data Fig. 3 TCR and BCR detection and diversity analysis for immune cells from colon tissue and paired blood specimens.

UMAP embedding with color depicting cells with TCR (A, B, E, F) or BCR (C, D) are shown (left most column) for (a) CD8 T cells from colon tissue, (b) CD4 T cells from colon tissue (c) B cells from colon tissue (d) B cells from blood, (e) CD4 T cells from blood, and (f) CD8 T cells from blood. TCR (A, B, E, F) or BCR (C, D) diversity plots are shown in the remaining three columns including the number of distinct clones per patient (second column from the left), cumulative percent of cells with the top N unique TCR or BCR clones (second column from the right), and Hill diversity index (right most column). Related to Figs. 2 and 4.

Extended Data Fig. 4 Detailed analysis of CD8 T/Gamma Delta T/NK cells and CD4 T cells from blood.

(a) UMAP embedding, (b) normalized gene expression, and (c) cell subset abundance difference between irColitis cases and controls for CD8 T/GDT/NK cells. (d) Percent of alpha beta CD8 T cell TCR clones shared between paired blood and tissue for each individual patient. Dots represent patients. irColitis case versus control two-sided t-test p-value is shown. (e) Volcano plot of pseudobulk differential gene expression with all cells for the contrast of irColitis cases versus controls for CD8 T/GDT/NK cells. The x-axis indicates the fold-change and y-axis indicates negative log10 p-value (two-sided) reported by limma. (f) Spearman correlation between each cluster and each of the reported CD8 T cell subsets from Giles et al, Immunity27. (g) UMAP embedding, (h) normalized gene expression, and (i) cell subset abundance difference between irColitis cases and controls for CD4 T cells. (j) Volcano plot of pseudobulk differential gene expression with all cells for the contrast of irColitis cases versus controls for CD4 T cells. (k) Spearman correlation between each cluster and each of the reported CD4 T cell subsets from Giles et al, Immunity27. Panels B, H show normalized gene expression (mean zero, unit variance) for selected genes, showing relative expression across cell clusters. Panels C, I show cell subset abundance differences between irColitis cases in orange and controls in gray, unadjusted likelihood ratio test p-values. Boxplots show median and interquartile range of patient cell type compositions where each dot represents a patient. The cellular composition of each patient is reported as the percentage of cells from a patient in each cell cluster. Error-bars indicate logistic regression OR 95% CI for differential abundance of cells from irColitis cases for each cell cluster. GDT gamma delta T cell. NK natural killer. Related to Figs. 2 and 4.

Extended Data Fig. 5 Detailed analysis of MP cells from colon tissue.

(a) UMAP embedding of 2,242 MP cells. Colors indicate cell cluster identities, which are listed on the right. (b) Normalized expression (mean zero, unit variance) of selected genes showing relative expression across cell clusters. Heatmap rows are aligned to every cluster defined in the UMAP. (c) Cell subset abundance differences between cases in orange (n = 13) and controls in gray (n = 14) across all MP cell subsets. Boxplots show patient cell type compositions where each dot represents a patient. Mononuclear phagocyte composition of each patient is reported as the percent of cells from a patient in each cell cluster, and box plots show median and interquartile range. Error bars indicate logistic regression 95% CI of OR for differential abundance of case cells for each cell cluster, and unadjusted likelihood ratio test p-values are shown. (d) Volcano of pseudobulk differential gene expression with all cells for the contrast of irColitis cases versus controls. The x-axis indicates the fold-change and y-axis indicates negative log10 p-value reported by limma (two-sided). (e) Bar plots showing differentially expressed (DE) genes per MP cluster (fold-change > 1.5 and FDR < 5%). (f) MP cell gene expression fold-change and log2CPM for cases (orange) and controls (black) is reported for selected genes across T cells (left and middle columns). Heatmap color indicates FC differences between irColitis cases and controls (right column). White dot indicates FDR < 5%. Panels show representative genes across multiple biological themes. (g) Gene expression for PD-1 ligands PD-L1 (CD274) and PD-L2 (PDCD1LG2) in each respective UMAP embedding (left panels). Estimated fold-changes between case and control for each myeloid cell cluster (middle). Gene expression values for the cells from each patient in each myeloid cell cluster where dots represent individual patients (right). Error bars show 95% CI and box plots show median and interquartile range. Related to Fig. 1.

Extended Data Fig. 6 Detailed analysis of B cells from colon tissue.

(a) UMAP embedding of 40,352 B cells. Colors indicate cell cluster identities, which are listed on the right. (b) Normalized expression (mean zero, unit variance) of selected genes showing relative expression across cell clusters. Heatmap rows are aligned to every cluster defined in the UMAP. (c) Cell subset abundance differences between cases in orange (n = 13) and controls in gray (n = 14) across all B cell subsets. Boxplots show patient cell type compositions where each dot represents a patient. B cell composition of each patient is reported as the percent of cells from a patient in each cell cluster. Error-bars indicate 95% CI of logistic regression OR for differential abundance of case cells for each cell cluster, and unadjusted likelihood ratio test p-values are shown. (d) Volcano of pseudobulk differential gene expression with all B cells for the contrast of irColitis cases versus controls. The x-axis indicates the fold-change and y-axis indicates negative log10 p-value reported by limma. (e) Bar plots showing differentially expressed (DE) genes per B cluster (fold-change greater than 1.5 and FDR less than 5%). (f) Ratio of IgG to IgA plasma cells across individual irColitis cases and controls. Each dot represents an individual patient. P-value for Wilcoxon rank sum test is shown. (g–h) irColitis in a B cell-depleted patient receiving ICI therapy for lymphoma. (G) Multispectral immunofluorescence staining of fixed colon mucosal tissue from patient C14* with a 7-color panel: DAPI (blue), panCK (gray), CD8A (aqua), PD-1 (orange), FOXP3 (yellow), CD68 (pink), and PD-L1 (green). (H) tSNE-embedding of 3,295 CD45+-sorted cells from a patient depleted of B cells. Cell cluster identity and top three AUC genes in parentheses are shown. MP: mononuclear phagocyte. Box plots show median and interquartile range. Related to Fig. 1.

Extended Data Fig. 7 Detailed analysis of epithelial and mesenchymal cells from tissue.

(a) Detailed composition of each patient across cell clusters for epithelial and mesenchymal nuclei from colon tissue. Heatmap color indicates percent of a patient’s cells assigned to each cell cluster. Every column represents one individual donor, referenced at the bottom of the heatmap. Upper right panel shows unique cell clusters depicted with different colors in UMAP embedding, which match the color scheme of the cell subset identities listed on the right side of the heatmap. (b) Schematic representing computationally-predicted cell-cell interactions between T cells expressing PD-1 (PDCD1) and cells expressing the PD-1 receptors PD-L1 (CD274) and PD-L2 (PDCD1LG2) (top cartoon). Three middle panels show differential expression analysis of means of gene pairs in indicated tissue cell types with cases (orange) and controls (black). Dots represent individual patients. Box plots show median and interquartile range. Limma fold-changes (FC) and two-sided p-values are shown. Bottom panels show feature plots of gene expression (Log2CPM) level in the UMAP embedding for MP cells from Extended Data Fig. 5a (left two panels) and CD8/GDT/NK cells from Fig. 2a (right two panels) presented separately for cells from cases and controls. Number and percentage of cells (from cases or from controls) with detected expression are reported at the bottom of each feature plot. (c) Feature plots use color to indicate gene expression (Log2CPM) level for selected epithelial and mesenchymal genes in the UMAP embedding in panel A. Number and percentage of nuclei with detected expression of each candidate gene are reported at the bottom of each feature plot. (d) FC, 95% CI, and gene expression (Log2CPM) (two left columns) for cases (orange) (n = 12) and controls (black) (n = 14) is reported for a set of genes organized across 7 biological themes. Heatmap (five right columns) indicates FC differences between cases and controls. White dot indicates FDR < 5%. (e) Feature plots show gene expression in nuclei from cases (left) and controls (right) using UMAP embedding in panel A. All feature plots shown in panels B, C, and E use color to indicate gene expression (Log2CPM). Number and percentage of cells with detected expression of each candidate gene are reported at the bottom of each feature plot. Related to Fig. 5.

Extended Data Fig. 8 Receptor-ligand interactions predict altered cellular communication in irColitis.

(a) Schematic of cell-cell communication inference. Predicted communication between two cell types is defined as proportional to the transcript levels of ligand and receptor genes in the two cell types. (b) Left: PDCD1 (log2CPM) (encoding PD-1) in tissue CD8 T cells (x-axis) and CD274 (encoding PD-L1) in epithelial and mesenchymal nuclei (y-axis). Right: Gene expression of PDCD1 and its ligands CD274 and PDCD1LG2 (encoding PD-L2) across blood immune cell types (top panels) and tissue immune, epithelial, and mesenchymal cells/nuclei (epi./mes.) (lower panels). Error-bars show 95% CI of fold-changes (black indicates FDR < 5%), cases (orange) and controls (black). Box plots show median and interquartile range. (c) Gene expression for ligand-receptor gene pairs, dots represent patients. X-axis indicates expression of HAVCR2, ITGB2, ITGAL, IL17A, IL26, CXCL13 in CD8 T cluster 3 cells (top row) or cluster 5 cells (bottom row). Y-axis indicates expression in epithelial cluster 2 cells (CEACAM1, ICAM1, IL17RA, IL17RC, IL20RA, IL10RB) or CD4 T cluster 6 cells (CXCR5). Unadjusted p-value and FDR (q-value) from differential expression (t-test) of the mean of each gene pair between cases and controls. (d) Differential expression analysis of means of gene pairs, focusing on putative communication between CD8 T cell clusters from Fig. 2a, b and other immune, epithelial, and mesenchymal cell lineages (Methods). Top: Tissue ITGB2HI circulatory (CD8-11, 3). Bottom: Activated effector CD8 TRM (ITGAEHI GZMBHI) (CD8-7, 2, 5). Heatmap color indicates FC between irColitis cases and controls. White dot indicates FDR < 5%. (e) DE analysis with five major tissue cell lineages (columns). Gene pairs are in five biological themes. White dots indicate FDR < 5%. (f) Left: correlations of cell cluster abundances within cases (x-axis) and within controls (y-axis), signed Spearman p-values. Right: abundance of tissue CD8-5 cells (x-axis) vs E-18 cells (y-axis). (g) Left: correlations of cell cluster abundances (cases and controls), x-axis Spearman correlation, y-axis -log10 p-value. Right: abundance of tissue CD8-11 (x-axis) vs E-13 (y-axis). E/M: epithelial and mesenchymal nuclei, B: B cells, MP: mononuclear phagocytes, CD8: CD8 T/gamma delta T/NK cells, CD4: CD4 T cells. Related to Figs. 25.

Extended Data Fig. 9 Spearman analysis of ligand-receptor gene pairs across multiple colon mucosal cell types.

(a) Volcano plot of Spearman correlations of percent of cells expressing pairs of genes, for all 10,101 gene-and-cell-type pairs (1,441 total unique gene pairs, tested for each pair of cell lineages). X-axis indicates correlation coefficient and y-axis p-value. (b) Number of gene pairs with FDR < 5% is depicted as an edge connecting each pair of cell lineages. Edge thickness and color is proportional to the number of gene pairs. (c) Spearman correlation of percent of cells with gene expression for selected pairs of genes for each pair of major cell lineages. (d) Heatmap color depicts the signed Spearman p-value for each pair of genes, for each pair of cell lineages. White dots indicate FDR < 5%. Columns indicate pairs of different cell lineages. E: epithelial and mesenchymal nuclei, B: B cells, MP: mononuclear phagocytes, CD8: CD8 T/GDT/NK cells, CD4: CD4 T cells. Pairs of genes in boldface are shown in panel (C). Related to Figs. 25.

Extended Data Fig. 10 Illustration of epithelial-immune interactions associated with irColitis.

Cartoon illustrating the major findings of our study showing that irColitis is defined by the colon mucosal expansion of ITGAEHI CD8 TRM T cells expressing CXCL13 and Th17 gene programs, ITGB2Hi CD8 T cells that may recirculate, Tregs, CD4 T cells expressing CXCL13 and IL17A, and ISGHi MP cells. Putative ligand/receptor pathways that recruit circulating cells to the endothelium (CX3CR1-CX3CL1, ITGAL/ITGB2-ICAM-1/2, CXCR3-CXCL9/10/11) and retain expanded CD8 T cells in tissue (ITGAL/ITGB2-ICAM-1, CXCR3-CXCL9/10/11) are shown. Epithelial defects in irColitis include decreased stem cells, increased transit amplifying cells, and top crypt epithelial cells with marked upregulation of interferon-stimulated genes (ISGs), CASP1, ZBP1, ICAM1, CD274/PD-L1, and CXCL10/11 and downregulation of aquaporin (AQP) water channel genes. Mesenchymal alterations in irColitis are notable for increased endothelial cells. The white oval at the bottom right shows the part of the crypt depicted in greater detail in the upper part of the cartoon. Related to Figs. 16.

Supplementary information

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

Thomas, M.F., Slowikowski, K., Manakongtreecheep, K. et al. Single-cell transcriptomic analyses reveal distinct immune cell contributions to epithelial barrier dysfunction in checkpoint inhibitor colitis. Nat Med (2024). https://doi.org/10.1038/s41591-024-02895-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41591-024-02895-x

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research