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:

Tissue-resident memory CD8+ T cells possess unique transcriptional, epigenetic and functional adaptations to different tissue environments

Abstract

Tissue-resident memory T cells (TRM cells) provide protective immunity, but the contributions of specific tissue environments to TRM cell differentiation and homeostasis are not well understood. In the present study, the diversity of gene expression and genome accessibility by mouse CD8+ TRM cells from distinct organs that responded to viral infection revealed both shared and tissue-specific transcriptional and epigenetic signatures. TRM cells in the intestine and salivary glands expressed transforming growth factor (TGF)-β-induced genes and were maintained by ongoing TGF-β signaling, whereas those in the fat, kidney and liver were not. Constructing transcriptional–regulatory networks identified the transcriptional repressor Hic1 as a critical regulator of TRM cell differentiation in the small intestine and showed that Hic1 overexpression enhanced TRM cell differentiation and protection from infection. Provision of a framework for understanding how CD8+ TRM cells adapt to distinct tissue environments, and identification of tissue-specific transcriptional regulators mediating these adaptations, inform strategies to boost protective memory responses at sites most vulnerable to infection.

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: TRM cells in distinct tissue microenvironments possess unique transcriptional programs.
Fig. 2: ScRNA-seq identifies distinct tissue and function-specific transcriptional programs in TRM cells.
Fig. 3: Sustained TGF-β signaling is required for the maintenance of TRM cells in IEL and SG.
Fig. 4: TRM cells in distinct tissue microenvironments possess unique epigenetic programs.
Fig. 5: Loss of Hic1 prevents the formation of SI TRM cells.
Fig. 6: Hic1 overexpression enhances the formation of SI TRM. cells.

Similar content being viewed by others

Data availability

All bulk RNA-seq, ATAC-seq and scRNA-seq datasets have been uploaded to the Gene Expression Omnibus repository (accession no. GSE182276). The following published datasets were used in addition: accession nos. GSE125527 (ref. 45), GSE70813 (ref. 10), GSE131847 (ref. 7), PRJNA414132 (ref. 20), GSE117568 (ref. 42), GSE63340 (ref. 17) and GSE128197 (ref. 41). The mouse reference genome mm10 has been used for RNA-seq, ATAC-seq and scRNA-seq analysis.

References

  1. Steinert, E. M. et al. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell 161, 737–749 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Skon, C. N. et al. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14, 1285–1293 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gebhardt, T. et al. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 10, 524–530 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Masopust, D. & Soerens, A. G. Tissue-resident T cells and other resident leukocytes. Annu. Rev. Immunol. 37, 521–546 (2019).

  5. Milner, J. J. et al. Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours. Nature 552, 253–257 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mackay, L. K. et al. The developmental pathway for CD103+CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 14, 1294–1301 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Kurd, N. S. et al. Early precursors and molecular determinants of tissue-resident memory CD8+ T lymphocytes revealed by single-cell RNA sequencing. Sci. Immunol. 5, eaaz6894 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Casey, K. A. et al. Antigen-independent differentiation and maintenance of effector-like resident memory T cells in tissues. J. Immunol. 188, 4866–4875 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Milner, J. J. et al. Heterogenous populations of tissue-resident CD8+ T cells are generated in response to infection and malignancy. Immunity 52, 808–824.e807 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mackay, L. K. et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459–463 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Hombrink, P. et al. Programs for the persistence, vigilance and control of human CD8+ lung-resident memory T cells. Nat. Immunol. 17, 1467–1478 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Du, N. et al. EGR2 is critical for peripheral naive T-cell differentiation and the T-cell response to influenza. Proc. Natl Acad. Sci. USA 111, 16484–16489 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Boddupalli, C. S. et al. ABC transporters and NR4A1 identify a quiescent subset of tissue-resident memory T cells. J. Clin. Invest. 126, 3905–3916 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Mackay, L. K. et al. T-box transcription factors combine with the cytokines TGF-beta and IL-15 to control tissue-resident memory T cellfate. Immunity 43, 1101–1111 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Ma, C., Mishra, S., Demel, E. L., Liu, Y. & Zhang, N. TGF-beta controls the formation of kidney-resident T cells via promoting effector T cell extravasation. J. Immunol. 198, 749–756 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Zhang, N. & Bevan, M. J. Transforming growth factor-beta signaling controls the formation and maintenance of gut-resident memory T cells by regulating migration and retention. Immunity 39, 687–696 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Frizzell, H. et al. Organ-specific isoform selection of fatty acid-binding proteins in tissue-resident lymphocytes. Sci. Immunol. 5, eaay9283 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Krausgruber, T. et al. Structural cells are key regulators of organ-specific immune responses. Nature 583, 296–302 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Han, S. J. et al. White adipose tissue is a reservoir for memory T cells and promotes protective memory responses to infection. Immunity 47, 1154–1168.e1156 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Milner, J. J. et al. Delineation of a molecularly distinct terminally differentiated memory CD8 T cell population. Proc. Natl Acad. Sci. USA 117, 25667–25678 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pan, Y. et al. Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism. Nature 543, 252–256 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc. 9, 209–222 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Schenkel, J. M. et al. IL-15-Independent maintenance of tissue-resident and boosted effector memory CD8 T cells. J. Immunol. 196, 3920–3926 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Svensson, M. et al. CCL25 mediates the localization of recently activated CD8alphabeta+ lymphocytes to the small-intestinal mucosa. J. Clin. Invest. 110, 1113–1121 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zabel, B. A. et al. Human G protein-coupled receptor GPR-9-6/CC chemokine receptor 9 is selectively expressed on intestinal homing T lymphocytes, mucosal lymphocytes, and thymocytes and is required for thymus-expressed chemokine-mediated chemotaxis. J. Exp. Med. 190, 1241–1256 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mazo, I. B. et al. Bone marrow is a major reservoir and site of recruitment for central memory CD8+ T cells. Immunity 22, 259–270 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Kim, D. et al. CXCL12 secreted from adipose tissue recruits macrophages and induces insulin resistance in mice. Diabetologia 57, 1456–1465 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. 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 

  30. O’Flanagan, C. H. et al. Dissociation of solid tumor tissues with cold active protease for single-cell RNA-seq minimizes conserved collagenase-associated stress responses. Genome Biol. 20, 210 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Wu, J. et al. T cell factor 1 suppresses CD103+ lung tissue-resident memory T cell development. Cell Rep. 31, 107484 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Liao, W. et al. The downregulation of IL-18R defines bona fide kidney-resident CD8+ T cells. iScience 24, 101975 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Thom, J. T., Weber, T. C., Walton, S. M., Torti, N. & Oxenius, A. The salivary gland acts as a sink for tissue-resident memory CD8+ T cells, facilitating protection from local cytomegalovirus infection. Cell Rep. 13, 1125–1136 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Nath, A. P. et al. Comparative analysis reveals a role for TGF-beta in shaping the residency-related transcriptional signature in tissue-resident memory CD8+ T cells. PLoS ONE 14, e0210495 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yu, B. et al. Epigenetic landscapes reveal transcription factors that regulate CD8+ T cell differentiation. Nat. Immunol. 18, 573–582 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhang, K., Wang, M., Zhao, Y. & Wang, W. Taiji: system-level identification of key transcription factors reveals transcriptional waves in mouse embryonic development. Sci. Adv. 5, eaav3262 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wang, D. et al. The transcription factor Runx3 establishes chromatin accessibility of cis-regulatory landscapes that drive memory cytotoxic T lymphocyte formation. Immunity 48, 659–674.e656 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zaid, A. et al. Persistence of skin-resident memory T cells within an epidermal niche. Proc. Natl Acad. Sci. USA 111, 5307–5312 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Khan, T. N., Mooster, J. L., Kilgore, A. M., Osborn, J. F. & Nolz, J. C. Local antigen in nonlymphoid tissue promotes resident memory CD8+ T cell formation during viral infection. J. Exp. Med. 213, 951–966 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Burrows, K. et al. The transcriptional repressor HIC1 regulates intestinal immune homeostasis. Mucosal Immunol. 10, 1518–1528 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Beura, L. K. et al. CD4+ resident memory T cells dominate immunosurveillance and orchestrate local recall responses. J. Exp. Med. 216, 1214–1229 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ricardo-Gonzalez, R. R. et al. Tissue signals imprint ILC2 identity with anticipatory function. Nat. Immunol. 19, 1093–1099 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Borges da Silva, H. et al. The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8+ T cells. Nature 559, 264–268 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Borges da Silva, H. et al. Sensing of ATP via the purinergic receptor P2RX7 promotes CD8+ TRM cell generation by enhancing their sensitivity to the cytokine TGF-beta. Immunity 53, 158–171 e156 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. 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 

  46. Milner, J. J. & Goldrath, A. W. Transcriptional programming of tissue-resident memory CD8+ T cells. Curr. Opin. Immunol. 51, 162–169 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mani, V. et al. Migratory DCs activate TGF-beta to precondition naive CD8+ T cells for tissue-resident memory fate. Science 366, eaav5728 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Stark, R. et al. TRM maintenance is regulated by tissue damage via P2RX7. Sci. Immunol. 3, eaau1022 (2018).

    Article  PubMed  Google Scholar 

  49. Hashimoto-Hill, S., Friesen, L., Kim, M. & Kim, C. H. Contraction of intestinal effector T cells by retinoic acid-induced purinergic receptor P2X7. Mucosal Immunol. 10, 912–923 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Mucida, D., Park, Y. & Cheroutre, H. From the diet to the nucleus: vitamin A and TGF-beta join efforts at the mucosal interface of the intestine. Semin. Immunol. 21, 14–21 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Heiss, K. et al. High sensitivity of intestinal CD8+ T cells to nucleotides indicates P2X7 as a regulator for intestinal T cell responses. J. Immunol. 181, 3861–3869 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. McFarland, A. P. et al. Multi-tissue single-cell analysis deconstructs the complex programs of mouse natural killer and type 1 innate lymphoid cells in tissues and circulation. Immunity 54, 1320–1337.e1324 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Chen, R. et al. In vivo RNA interference screens identify regulators of antiviral CD4+ and CD8+ T cell differentiation. Immunity 41, 325–338 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  56. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  57. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinf. 14, 7 (2013).

    Article  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e1821 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e727 2018).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  Google Scholar 

  65. Davis, C. A. et al. The encyclopedia of DNA elements (ENCODE): data portal update. Nucleic Acids Res. 46, D794–D801 (2018).

    Article  CAS  PubMed  Google Scholar 

  66. Ross-Innes, C. S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was funded by the National Institutes of Health (grant no. AI067545 to A.W.G. and no. AI132122 to A.W.G. and J.T.Chang) and the American Cancer Society Postdoctoral Fellowship (grant no. PF-20-048-01-LIB to J.T.Crowl). ATAC-seq and scRNA-seq using the 10× Genomics platform was conducted at the IGM Genomics Center, UCSD and supported by grant nos. P30KC063491 and P30CA023100. A.W.G. is a UCSD Tata Chancellor’s Professor. M.H. was supported by the German Research Foundation fellowship (no. HE 8656/1-1). We thank H. Nguyen for assistance with measuring LCMV titers, the Goldrath laboratory members for technical advice, helpful discussion and critical reading of the manuscript and the Immunological Genome Project for reagents and sample/data processing.

Author information

Authors and Affiliations

Authors

Contributions

J.T. Crowl, M.H., J.T. Chang and A.W.G. conceived the project and performed the methodology. J.T. Crowl, M.H., A.F., C.T., K.D.O., J.J.M. and Z.E. did the investigations. J.T. Crowl, M.H., A.F. and J.J.M. carried out the formal analysis. J.T. Crowl, M.H. and A.W.G. wrote the original draft of the paper. J.T. Crowl, M.H., J.T. Chang, K.D.O. and A.W.G. wrote the paper. J.T. Chang. and A.W.G. supervised the project. A.W.G. and J.T. Chang acquired the funds.

Corresponding author

Correspondence to Ananda W. Goldrath.

Ethics declarations

Competing interests

A.W.G. is a member of the ArsenalBio scientific advisory board. J.T. Crowl is a current employee of Outpace Bio. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Ioana Visan in collaboration with the Nature Immunology 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 Gating strategy.

a, Gating strategy used to identify indicated IV TRM populations.

Extended Data Fig. 2 Phenotypic characterization of TRM after LM-gp33 infection and expression of select genes in TRM from other published datasets.

a-b, CD69 and CD103 expression by CD8+ TRM isolated from tissues 30-40 days after infection with LM-GP33. Representative flow cytometry plots (a) and quantification (b). c-d, Percent of GZMA+ (c) and GZMB+ (d) P14 cells isolated from the indicated tissues 30-40 days after infection with LM-GP33 as assessed by flow cytometry. Datasets are from (e) Mackay et al, Science 2016. (f) Han et al, Immunity 2017. (g) ex vivo PD1 and Lag3 expression in P14 cells isolated from the indicated tissues. Quantification of flow cytometry data in b, c and d displays the mean ± SD for 10 mice from 3 experimental replicates. Data in g shows a representative experiment with 3 mice from a total of 3 experiments with 10 mice. Significance was calculated using a one-way ANOVA and corrected for multiple comparisons using Tukey’s test. ****p < 0.0001.

Extended Data Fig. 3 Collagenase digestion induces upregulation of a subset of genes also associated with tissue residency.

a-d, P14 cells were adoptively transferred into CD45 congenic hosts one day prior to infection with LCMV. 30-40 days after initial infection, P14 cells were isolated from tissues using no additional treatment (NoTx), collagenase (Coll), or a cold active protease (CAP). a, Quantification of the number of P14 cells recovered from each tissue using the indicated digestion methods. b, Percent of P14 cells expressing CD69 (top left), CD103 (top center left), IL-18R1 (top center right), CD8a (top right), KLRG1 (bottom left), CD127 (bottom center), or CD62L (bottom right) assessed by flow cytometry. c-e, RNA-sequencing of P14 cells isolated from the spleen or kidney using NoTx, Coll, dithioerythritol (DTE), or CAP. c, Differentially expressed genes (348) were clustered with k-means = 3. Select genes in each cluster displayed on the right. Genes that were upregulated in CAP-treated tissues compared to CAP-treated spleens indicated with an asterisk. d, Principal Component Analysis. e, Cd69 expression by P14 cells isolated from the spleen or kidney with CAP. f,g, Genes included in the TRM signatures from this paper (left), Milner et al, Nature 2017 (center) and Mackay et al, Science 2016 (right) were selected. f, Corresponding expression values for collagenase-digested kidney, CAP-digested kidney, and CAP-digested spleen samples were plotted. Each gene in the corresponding TRM signature is represented by a single point and colored by influence of digestion on expression. g, Venn diagram of the preceding data. h, Principal component analysis of RNA-sequencing data from Fig. 1 with all digestion-associated genes removed. Genes were considered digestion-associated if they were expressed above a minimum threshold and at >1.5 fold in collagenase-digested kidney compared to CAP-digested kidney samples. Graphs in a and b display the mean ± SD for 10 mice from 3 experimental replicates. RNA-seq data displayed in c-f contains 2-3 experimental replicates for each sample, and tissues from multiple mice were pooled. Graph in e displays the mean ± SD. Significance calculated using a two-way ANOVA and correcting for multiple comparisons using Dunnett’s test. *p < 0.05, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 4 Top enriched genes identified in bulk RNA-sequencing of TRM are also found in scRNA-sequencing.

a, The top 5 genes enriched in bulk RNA-sequening samples for TRM isolated from the blood, IEL, SG, fat, and liver are shown on a UMAP dimensional reduction plot.

Extended Data Fig. 5 Removal of digestion-associated gene signature from the TRM gene signature does not alter the enrichment of tissue signature.

a,b, scRNA-sequencing data described in Fig. 2. Each cell was scored based on the enrichment of genes included in the indicated signatures. Cells were colored by score on a UMAP dimensional reduction (a) and separated by cluster and ordered based on score (b).

Extended Data Fig. 6 TRM differentiation programs are a source of intra-tissue heterogeneity.

a, UMAP dimensional reduction of scRNA-sequencing of TRM separated by tissue. Cells were colored by the expression of the indicated genes. Scales are consistent across tissues to allow for comparison within and among tissues. b-c, Expression of CD69, Ly6C, IL18R1 on P14 cells harvested 30-40 days after initial infection with LCMV. Representative flow cytometry plots (b) and quantification (c). d, Quantification of IL18R1 expression on P14 cells harvested from the indicated tissues 30-40 days after initial infection with LM-GP33. Quantification of flow cytometry data in c and d displays the mean ± SD for 6 (c) 10 (d) mice from 2 experimental replicates.

Extended Data Fig. 7 TRM in distinct tissue microenvironments possess unique epigenetic programs.

a-d, ATAC-seq of P14 CD8+ T cells in the spleen and IV P14 CD8+ T cells isolated from the IEL, kidney, SG, fat, and liver. a, Pearson correlation for all peaks across all samples. b, Annotation of the genomic region type for all identified accessible regions (left) and DAR (right). c,d, Shared and unique upregulated DAR (c) and downregulated DAR (d) in each tissue compared to the spleen for all DAR with a p-value <0.05 and a fold change >4 using a Wald statistics.

Extended Data Fig. 8 Blimp1 deletion impairs TRM formation in the IEL and SG more than the kidney.

a-c, Gzmb-Cre/Prdm1fl/fl (WT) and Gzmb-Cre+/Prdm1fl/fl (KO) were transferred at a 1:1 ratio into congenically distinct recipients one day prior to infection with LCMV. Tissues were harvested 60 days after initial infection. a, Ratio of KO to WT P14 cells in the indicated tissues. b-c, % of CD69+ (b) and CD103+ (c) P14 cells for WT and KO populations. Graphs display mean ± SD for a combined 2 experimental replicates, each with m = 4 mice. Significance in (a) calculated with a one-way ANOVA using Tukey’s multiple comparison test. Significance in (b-c) calculated with a two-way ANOVA using with Sidak’s multiple comparison test. ****p < 0.0001.

Extended Data Fig. 9 Hic1 is critical for the differentiation of small intestine TRM.

a, Hic1 expression by resident immune cell populations isolated from the indicated tissues. b-g, 1:1 mixed transfer of P14 cells transduced with a control shRNA or a Hic1-targeting shRNA. b-c, Percentage of P14 cells that are CD69+CD103 (left) or CD69+CD103+ (right) on day 7-8 (b) or day 20-21 post-infection with LCMV (c). d, Percentage of P14 cells that are CD69+CD103 (left) or CD69+CD103+ (right) on day 20 post-infection with LM-GP33. e, Percentage of P14 cells that were terminal effectors (TE, KLRG1+CD127) or memory precursors (MP, KLRG1CD127+) on day 7-8 post-infection with LCMV. f, Percentage of P14 cells that are terminal effector memory (tTEM, CD127-CD62L-), effector memory (TEM, CD127+CD62L-), or central memory (TCM, CD127+CD62L+) on day 20-21 post-infection with LCMV. g-h, Percentage of P14 cells that were TE or MP on day 7 (g) or day 20 (h) after infection with LM-GP33. i-l, 1:1 mixed transfer of P14 cells transduced with a control vector or a Hic1-overexpression vector. i-j, Percentage of P14 cells that are CD69+CD103 (left) or CD69+CD103+ (right) on day 7-8 (i) or day 20-21 (j) post-infection with LCMV. k, Percentage of P14 cells that were TE or MP on day 7-8 post-infection with LCMV l, Percentage of P14 cells that were tTEM, TEM, or TCM on day 20-21 post-infection with LCMV. m, P2xr7 expression by resident immune cell populations isolated from the indicated tissues. Graphs in a and m display mean ± SD for the expression values from RNA-Seq samples (22 samples for CD8+, 17 samples for CD4+, 18 samples for Macrophages, 26 samples for ILC2). Graphs in b, c, e, f, il display mean ± SD for 11 mice from 3 experimental replicates. Graphs in d, g, and h display mean ± SD for 8 mice from 2 individual experiments. Significance calculated with a two-way ANOVA using with Sidak’s multiple comparison test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 10 Human TRM recapitulate phenotypes observed in murine TRM.

a-c, Single-cell RNA-sequencing of healthy human tissue in Boland et al, Science Immunology 2020. a, Hic1 expression after MAGIC imputation. b,c, Individual cells are scored based on enrichment for genes included in the TGFβ signature (b) and TRM signature (c). Single cell data was pooled from 13 different healthy donors for PBMC and rectum biopsies and 10 healthy donors for intestinal samples. Boxplot shows median. The lower and upper hinges correspond to the first and third quartiles. The upper whisker extends from the hinge to the largest value no further than 1.5 * IQR from the hinge. Statistics were calculated by aggregating the scRNA data to pseudo-bulk samples for each patient and cell type. A T statistics test as implemented in the R package limma was then used to calculate the P values.

Supplementary information

Reporting summary

Supplementary Table 1

RNA-seq analysis of memory CD8+ T cells isolated from distinct tissues.

Supplementary Table 2

RNA-seq analysis of memory CD8+ T cells isolated using distinct methods.

Supplementary Table 3

ScRNA-seq analysis of memory CD8+ T cells isolated from individual tissues.

Supplementary Table 4

ATAC-seq analysis of memory CD8+ T cells isolated from the indicated tissues.

Supplementary Table 5

PageRank analysis of memory CD8+ T cells isolated from distinct tissues.

Supplementary Table 6

RNA-seq analysis of control and Hic1-overexpressing CD8+ T cells from the spleen.

Supplementary Table 7

Antibodies used in the present paper.

Supplementary Table 8

Gene signatures used in the present paper.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Crowl, J.T., Heeg, M., Ferry, A. et al. Tissue-resident memory CD8+ T cells possess unique transcriptional, epigenetic and functional adaptations to different tissue environments. Nat Immunol 23, 1121–1131 (2022). https://doi.org/10.1038/s41590-022-01229-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01229-8

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