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:

Runx3 drives a CD8+ T cell tissue residency program that is absent in CD4+ T cells

Abstract

Tissue-resident memory T cells (TRM cells) provide rapid and superior control of localized infections. While the transcription factor Runx3 is a critical regulator of CD8+ T cell tissue residency, its expression is repressed in CD4+ T cells. Here, we show that, as a direct consequence of this Runx3-deficiency, CD4+ TRM cells lacked the transforming growth factor (TGF)-β-responsive transcriptional network that underpins the tissue residency of epithelial CD8+ TRM cells. While CD4+ TRM cell formation required Runx1, this, along with the modest expression of Runx3 in CD4+ TRM cells, was insufficient to engage the TGF-β-driven residency program. Ectopic expression of Runx3 in CD4+ T cells incited this TGF-β-transcriptional network to promote prolonged survival, decreased tissue egress, a microanatomical redistribution towards epithelial layers and enhanced effector functionality. Thus, our results reveal distinct programming of tissue residency in CD8+ and CD4+ TRM cell subsets that is attributable to divergent Runx3 activity.

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: Runx1 and Runx3 differentially program CD4+ and CD8+ TRM cell development.
Fig. 2: Enforced Runx3 expression increases epithelial residency in CD4+ T cells.
Fig. 3: Runx3 induces a CD8-like TRM cell transcriptional signature in CD4+ T cells.
Fig. 4: Runx3 promotes chromatin accessibility at TGF-β regulated genes.
Fig. 5: Runx3 programs TGF-β-responsiveness in CD4+ T cells to establish a CD8+ TRM cell-like transcriptome in skin.
Fig. 6: Runx3-induced tissue residency depends on TGF-β signaling.
Fig. 7: Runx3 enforces protective CD8+ TRM cell-like immune surveillance by CD4+ T cells in skin.

Similar content being viewed by others

Data availability

All original data is available from the corresponding author upon reasonable request. RNA-seq and ATAC-seq data is available in the Gene Expression Omnibus database under accession codes GSE182511 and GSE198611, respectively. Source data are provided with this paper.

Code availability

The code generated and used for the analysis of sequencing data are available from the corresponding author on reasonable request.

References

  1. Jameson, S. C. & Masopust, D. Understanding subset diversity in T cell memory. Immunity 48, 214–226 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 

  4. Kok, L. et al. A committed tissue-resident memory T cell precursor within the circulating CD8+ effector T cell pool. J. Exp. Med. 217, e20191711 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kurd, 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 

  6. Kok, L., Masopust, D. & Schumacher, T. N. The precursors of CD8+ tissue resident memory T cells: from lymphoid organs to infected tissues. Nat. Rev. Immunol. 22, 283–293 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Hirai, T. et al. Keratinocyte-mediated activation of the cytokine TGF-β maintains skin recirculating memory CD8+ T cells. Immunity 50, 1249–1261.e1245 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang, N. & Bevan, J. M. Transforming growth factor-β 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  CAS  Google Scholar 

  9. Christo, S. N. et al. Discrete tissue microenvironments instruct diversity in resident memory T cell function and plasticity. Nat. Immunol. 22, 1140–1151 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. 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 

  12. 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 

  13. 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 

  14. Nguyen, Q. P., Deng, T. Z., Witherden, D. A. & Goldrath, A. W. Origins of CD4+ circulating and tissue‐resident memory T‐cells. Immunology 157, 3–12 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Schreiner, D. & King, C. G. CD4+ memory T cells at home in the tissue: mechanisms for health and disease. Front Immunol. 9, 2394 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Hondowicz, B. D., Kim, K. S., Ruterbusch, M. J., Keitany, G. J. & Pepper, M. IL-2 is required for the generation of viral-specific CD4+ Th1 tissue-resident memory cells and B cells are essential for maintenance in the lung. Eur. J. Immunol. 48, 80–86 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Turner, D. L. & Farber, D. L. Mucosal resident memory CD4 T cells in protection and immunopathology. Front Immunol. 5, 331 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Kumar, B. V. et al. Human tissue-resident memory T cells are defined by core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep. 20, 2921–2934 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Collins, N. et al. Skin CD4+ memory T cells exhibit combined cluster-mediated retention and equilibration with the circulation. Nat. Commun. 7, 11514 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Iijima, N. & Iwasaki, A. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 346, 93–98 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gebhardt, T. et al. Different patterns of peripheral migration by memory CD4+ and CD8+ T cells. Nature 477, 216–219 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Ariotti, S. et al. Tissue-resident memory CD8+ T cells continuously patrol skin epithelia to quickly recognize local antigen. Proc. Natl Acad. Sci. USA 109, 19739–19744 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Shin, B. et al. Runx1 and Runx3 drive progenitor to T-lineage transcriptome conversion in mouse T cell commitment via dynamic genomic site switching. Proc. Natl Acad. Sci. USA 118, e2019655118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Setoguchi, R. et al. Repression of the transcription factor Th-POK by Runx complexes in cytotoxic T cell development. Science 319, 822–825 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Luckey, M. A. et al. The transcription factor ThPOK suppresses Runx3 and imposes CD4+ lineage fate by inducing the SOCS suppressors of cytokine signaling. Nat. Immunol. 15, 638–645 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ciucci, T. et al. The emergence and functional fitness of memory CD4+ T cells require the transcription factor Thpok. Immunity 50, 91–105.e104 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Djuretic, I. M. et al. Transcription factors T-bet and Runx3 cooperate to activate Ifng and silence Il4 in T helper type 1 cells. Nat. Immunol. 8, 145–153 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Komine, O. et al. The Runx1 transcription factor inhibits the differentiation of naive CD4+ T cells into the Th2 lineage by repressing GATA3 expression. J. Exp. Med. 198, 51–61 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Reis, B. S., Rogoz, A., Costa-Pinto, F. A., Taniuchi, I. & Mucida, D. Mutual expression of the transcription factors Runx3 and ThPOK regulates intestinal CD4+ T cell immunity. Nat. Immunol. 14, 271–280 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  32. 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 

  33. Schenkel, J. M. et al. T cell memory. Resident memory CD8 T cells trigger protective innate and adaptive immune responses. Science 346, 98–101 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ariotti, S. et al. T cell memory. Skin-resident memory CD8+ T cells trigger a state of tissue-wide pathogen alert. Science 346, 101–105 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Taniuchi, I. et al. Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development. Cell 111, 621–633 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Moreau, J. M., Velegraki, M., Bolyard, C., Rosenblum, M. D. & Li, Z. Transforming growth factor-β1 in regulatory T cell biology. Sci. Immunol. 7, eabi4613 (2022).

    Article  CAS  PubMed  Google Scholar 

  37. Korn, T., Bettelli, E., Oukka, M. & Kuchroo, V. K. IL-17 and Th17 cells. Annu. Rev. Immunol. 27, 485–517 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Mucida, D. et al. Transcriptional reprogramming of mature CD4+ helper T cells generates distinct MHC class II–restricted cytotoxic T lymphocytes. Nat. Immunol. 14, 281–289 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Keller, H. R. et al. The molecular basis and cellular effects of distinct CD103 expression on CD4 and CD8 T cells. Cell. Mol. Life Sci. 78, 5789–5805 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Fonseca, R. et al. Developmental plasticity allows outside-in immune responses by resident memory T cells. Nat. Immunol. 21, 412–421 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Stolley, J. M. et al. Retrograde migration supplies resident memory T cells to lung-draining LN after influenza infection. J. Exp. Med. 217, e20192197 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Klicznik, M. M. et al. Human CD4+CD103+ cutaneous resident memory T cells are found in the circulation of healthy individuals. Sci. Immunol. 4, eaav8995 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Masopust, D. et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 207, 553–564 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. 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 

  46. Cheroutre, H. & Husain, M. M. CD4 CTL: living up to the challenge. Semin. Immunol. 25, 273–281 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Delacher, M. et al. Single-cell chromatin accessibility landscape identifies tissue repair program in human regulatory T cells. Immunity 54, 702–720.e17 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Durand, A. et al. Profiling the lymphoid-resident T cell pool reveals modulation by age and microbiota. Nat. Commun. 9, 68 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. 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 

Download references

Acknowledgements

We thank the Flow Cytometry Unit and Bioresources Facility at Peter Doherty Institute (University of Melbourne) for technical assistance. This work was supported by a Howard Hughes Medical Institute and Bill and Melinda Gates International Research Scholarship OPP1175796, National Health and Medical Research Council (NHMRC) AP1113293 to F.RC. and L.K.M. S.L.P. was supported by a NHMRC EL1 Investigator Grant GNT1175626. N.G.Z. was supported by FAPESP (BEPE 2019/12431-2). A.T.S. was supported by the National Institutes of Health (U01CA260852 and UM1HG012076), the Parker Institute for Cancer Immunotherapy and a Pew-Stewart Scholars for Cancer Research Award. L.K.M is a Senior Medical Research Fellow supported by the Sylvia and Charles Viertel Charitable Foundation.

Author information

Authors and Affiliations

Authors

Contributions

R.F., T.N.B., L.C.G., S.D., A.O., S.L.P., M.E., S.K.S., S.N.C., N.M.Z., N.G.Z., F.A.B., C.A.L., K.M. and A.Z. performed experiments and analyzed data; S.N.M., T.P.S., A.T.S. and L.K.M. provided supervision; R.F., T.N.B., S.L.P. and L.K.M. contributed to experimental design; R.F., T.N.B., F.R.C. and L.K.M. prepared the manuscript; A.T.S., F.R.C. and L.K.M. provided funding; F.R.C. and L.K.M. led the research program.

Corresponding author

Correspondence to Laura K. Mackay.

Ethics declarations

Competing interests

The authors declare no competing interests. A.T.S. is a founder of Immunai and Cartography Biosciences and receives unrelated research funding from Merck Research Laboratories, Allogene Therapeutics, and Arsenal Biosciences, L.K.M. receives unrelated research funding from Pfizer.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. 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 Distribution and phenotype of memory T cells in the epithelia.

(a-d) Microscopy showing CD4+ SMARTA and CD8+ P14 cells in the small intestine (SI) 7 and 30 d.p.i. with LCMV (IEL cells highlighted by teal (CD4+) and yellow (CD8+) circles) (a), kinetics of CD4+ SMARTA and CD8+ P14 cells (**P=0.0079, 0.0043, 0.0079 respectively, two-tailed Mann-Whitney’s test) (b), CD69 and CD103 expression in SI-IEL at 7, 14 or 30 d.p.i. (***P=0.007, <0.0001 respectively, multiple paired t-tests) (c) and representative flow plots 30 d.p.i. (d). (e-i) CD69 and CD103 expression in CD4+ gDT-II and CD8+ gBT-I cells from skin at 7, 14 or 30d post-HSV infection (***P=0.0028, <0.0001 respectively, multiple paired t-tests) (e) and representative flow plots in skin 30 d.p.i. (f), representative histograms of Runx3 expression in spleen or skin 30 d.p.i. (g), representative histograms of ThPOK and Runx3 expression (h) and Runx3 expression (gMFI) in spleen 7 d.p.i. normalized to naïve (i). (j) Correlation plots of Runx1 or Runx3 and CD103 gMFI on endogenous Foxp3CD69+CD4+ and CD69+CD8+ isolated from skin 14 d.p.i. with HSV (linear regression line with 95% confidence interval, P showing slope is non-zero). (k, l) Runx1 and Runx3 expression in CD8+ gBT-I cells (k) and CD4+ gDT-II cells (l) in vitro prior to transfer into recipient mice following CRISPR-Cas9 mediated editing of CD19 (Ctrl), Runx1 (sgRunx1), or Runx3 (sgRunx3). (m) Log2-fold change of sgCtrl and sgRunx1 CD4+ SMARTA cells (as in k, **P=0.002, paired t-test) in the spleen and SI-IEL >20d post-LCMV infection. Data are representative of 2 independent experiments with (a) n=3, (b-i) n=5, (j) 1 experiment, n=5, (k) 2 independent biological replicates, (m) pooled from 2 independent experiments, with n=5 mice. Symbols represent (c, e, j, m) mice, (b, i) mean; error bars indicate SEM. Box plots represent the median, interquartile range, and minimum/maximum whiskers.

Source data

Extended Data Fig. 2 Ectopic Runx3 enhances CD8+ TRM cell formation in the epithelia.

(a, b) Schematic and frequency of CD8+ P14 cells transduced with control (CD8-Ctrl) or Runx3 (CD8-Runx3) retroviruses in the spleen, skin and SI-IEL 14d post-LCMV infection and DNFB skin treatment (a) and enumeration of total or CD69+CD103+ cells in the spleen, skin (**P=0.0022, two-tailed paired t-test) and SI-IEL (***P=0.0002 and 0.0007 respectively) (b). Data pooled from (a, b) 2 independent experiments, n=4 mice. Symbols represent individual mice; bars represent mean; error bars indicate SEM.

Source data

Extended Data Fig. 3 Runx3 increases CD4+ TRM cell formation in the epithelia.

(a-c) Schematic and frequency of CD4+ gDT-II cells transduced with control (CD4-Ctrl) or Runx3 (CD4-Runx3) retroviruses in the spleen and skin (a), epidermis/dermis proportion (**P=0.0048, two-tailed paired t-test) (b) and CD69 and CD103 expression in the epidermis (**P=0.0021, two-tailed paired t-test) and dermis (***P=0.0001, two-tailed paired t-test) (c) 14d post-HSV infection. (d-f) Schematic (d), frequency of CD4+ SMARTA cells transduced with control (CD4-Ctrl) or Runx3 (CD4-Runx3) retroviruses (e) and number of total and CD69+CD103+ cells in spleen, skin (*P=0.043, two-tailed paired t-test) and SI-IEL (*P=0.0432 and 0.0332) (f) 14d post-LCMV infection in mice treated on the skin with DNFB. (g, h) Representative histograms of Runx3 (g) and expression (MFI) by endogenous CD4+, CD8+ and CD4+CD8αα+ T cells (h) in the spleen and SI-IEL >90d post-LCMV infection. (i) Runx3 expression by endogenous CD8+ T cells (CD8-Endog) and transduced Ctrl- (CD4-Ctrl) or Runx3-transduced (CD4-Runx3) CD4+ gDT-II (spleen and skin) or CD4+ SMARTA (SI-IEL) cells as in (a-f) 14 d.p.i. with HSV or LCMV respectively. Data pooled from (a-c) 2 independent experiments with n=10 (d-f) 2 independent experiments, n=5 or representative of (g, h) 2 independent experiments, n=5 and (i) 2 independent experiments, n=3 mice. Symbols represent individual mice; bars indicate mean; error bars indicate SEM.

Source data

Extended Data Fig. 4 Runx3 induces a CD8+ TRM cell-like signature in CD4+ T cells.

(a) Heatmap showing expression of the top TRM cell DEGs (derived from the comparison between skin CD8+ TRM cells vs spleen CD8+ TCIRC cells from GSE70813) in CD4-Runx3 and CD8-Ctrl samples relative to CD4-Ctrl cells, by CD8+ gBT-I cells transduced with control (CD8-Ctrl) and CD4+ gDT-II cells transduced with control (CD4-Ctrl) or Runx3-expressing (CD4-Runx3) retroviruses isolated from the skin 14 d.p.i. (columns represents independent samples, color scale is based on Z-score). (b) GSEA in CD8-Ctrl vs CD4-Ctrl (top) or CD4-Runx3 vs CD8-Ctrl cells (bottom) for the top 100 DEGs in skin CD8+ TRM vs splenic CD8+ TCIRC cells. (c) Scatter plot showing DEGs between skin CD8+ TRM vs splenic CD8+ TCIRC cells and CD8-Ctrl vs CD4-Ctrl or CD8-Ctrl vs CD4-Runx3 (dots represent up- (red) or down-regulated (blue) genes in skin CD8+ TRM vs splenic CD8+ TCIRC cells, quadrants represent up- (red) or down-regulated (blue) genes in CD8-Ctrl vs CD4-Ctrl or CD8-Ctrl vs CD4-Runx3 comparisons). (d) Scatter plots of standardized log fold-change of CD8-Ctrl vs CD4-Ctrl (x-axis) and CD4-Runx3 vs CD4-Ctrl (y-axis; orange dots highlight top skin TRM cell genes (left panel), blue and red dots highlight top 200 down- (middle panel) and up-regulated (right panel) genes in skin TRM cells) (d). Data pooled from 2 independent experiments, n=2 biological replicates pooled from 10 mice. Green line represents least-squares regression line fitted to orange points.

Extended Data Fig. 5 Runx3 promotes epigenetic remodeling in CD4+ T cells.

(a) Scatter plots showing peak accessibility changes in CD8-Ctrl cells treated with TGF-β vs Ctrl and CD4-Ctrl (top) or CD4-Runx3 (bottom) treated with TGF-β vs Ctrl. (b) Scatter plots showing peak accessibility changes in CD8-Runx3 vs CD8-Ctrl cells and CD4-Runx3 vs CD4-Ctrl treated with TGF-β (bottom) or untreated (top; orange dots highlight TGF-β regulated genes). (c) Itga1, Cd244, S1pr1 and Ly6c genome tracks (height normalized) in CD4+ gDT-II and CD8+ gBT-I cells transduced with control (CD4-Ctrl and CD8-Ctrl) or Runx3-expressing (CD4-Runx3 and CD8-Runx3) retroviruses cultured ± TGF-β for 48 hours. Data representative of 2 independent experiments, with n=2 technical replicates.

Extended Data Fig. 6 Runx3 induces expression of TGF-β-regulated genes in CD4+ T cells.

(a) Heatmap showing the top TGF-β regulated genes (derived from the comparison between skin wild-type (WT) CD8+ T cells vs Tgfbr2/ CD8+ T cells 14d post-HSV infection from GSE178769) in CD4-Runx3 and CD8-Ctrl samples relative to CD4-Ctrl expression, by CD8+ gBT-I cells transduced with control (CD8-Ctrl) and CD4+ gDT-II cells transduced with either control (CD4-Ctrl) or Runx3-expressing (CD4-Runx3) retroviruses isolated from the skin 14d post-HSV infection (columns represents independent samples, color scale is based on Z-score). (b) GSEA in CD8-Ctrl vs CD4-Ctrl (top) or CD4-Runx3 vs CD8-Ctrl cells (bottom) for the top 100 DEGs in skin WT vs Tgfbr2/ cells. (c) Scatter plot showing DEGs between WT vs Tgfbr2/ cells and CD8-Ctrl vs CD4-Ctrl (left) or CD8-Ctrl vs CD4-Runx3 (right, dots represent up- (red) and down-regulated (blue) genes in WT vs Tgfbr2/ cells comparison, quadrants represents up- (red) and down-regulated (blue) genes in CD8-Ctrl vs CD4-Ctrl or CD8-Ctrl vs CD4-Runx3 comparisons). (d) Scatter plots of standardized log-fold changes of CD8-Ctrl vs CD4-Ctrl (x-axis) and CD4-Runx3 vs CD4-Ctrl (y-axis) for all genes (orange dots highlight top TGF-β regulated genes (left panel), blue and red dots highlight top 200 down-regulated (middle panel) and up-regulated (right panel) genes in WT cells). Data pooled from 2 independent experiments, n=2 biological replicates pooled from 10 mice. Green line represents least-squares regression line fitted to orange dots.

Extended Data Fig. 7 Runx3 promotes enhanced local immune protection by skin CD4+ T cells.

(a) Intracellular cytokine production of IFNγ and TNFα by CD4+ gDT-II cells transduced with either control (CD4-Ctrl) or Runx3-expressing (CD4-Runx3) retroviruses 4 hours after PMA+ionomycin stimulation. (b, c) Viral titer (*P=0.0111, two-tailed unpaired t-test) 6d post-HSV infection (b), and enumeration of transduced cells, inflammatory monocytes, NK cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, B cells and dendritic cells 3 d.p.i (*P=0.0455, 0.0301 and 0.021 respectively, two-tailed unpaired t-test) (c) in the skin of C57BL/6 mice transferred i.d. with CD4+ gDT-II cells transduced with control (CD4-Ctrl) or Runx3-expressing (CD4-Runx3) retroviruses 14d prior to HSV infection. Data pooled from 2 independent experiments with (a) n=2 biological replicates, (b, c) n=5 mice. Symbols represent (a) biological replicates or (b, c) mice. Bars represent mean; error bars indicate SEM. Box plots show the median, interquartile range and minimum/maximum whiskers.

Source data

Supplementary information

Supplementary Information

Supplementary methods, references and gating strategy.

Reporting Summary

Peer Review File

Supplementary Video 1

CD4+ T cells from uGFP-reporter mice were transduced with an empty-Ametrine-encoding retrovirus (CD4-Ctrl) and CD4+ T cells from ubiTomato-reporter mice were transduced with a Runx3-GFP-encoding retrovirus (CD4-Runx3). Transduced cells were cotransferred i.d. into recipient mice and intravital two-photon microscopy performed at day 14 post-i.d. transfer. SHG..

Supplementary Table 1

List of antibodies.

Source data

Source Data Fig. 1

Source Data Fig. 1.

Source Data Fig. 2

Source Data Fig. 2.

Source Data Fig. 5

Source Data Fig. 5.

Source Data Fig. 6

Source Data Fig. 6.

Source Data Fig. 7

Source Data Fig. 7.

Source Data Extended Data Fig. 1

Source Data Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Source Data Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Source Data Extended Data Fig. 3.

Source Data Extended Data Fig. 7

Source Data Extended Data Fig. 7.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fonseca, R., Burn, T.N., Gandolfo, L.C. et al. Runx3 drives a CD8+ T cell tissue residency program that is absent in CD4+ T cells. Nat Immunol 23, 1236–1245 (2022). https://doi.org/10.1038/s41590-022-01273-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01273-4

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