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:

Hobit- and Blimp-1-driven CD4+ tissue-resident memory T cells control chronic intestinal inflammation

An Author Correction to this article was published on 07 March 2019

This article has been updated

Abstract

Although tissue-resident memory T cells (TRM cells) have been shown to regulate host protection in infectious disorders, their function in inflammatory bowel disease (IBD) remains to be investigated. Here we characterized TRM cells in human IBD and in experimental models of intestinal inflammation. Pro-inflammatory TRM cells accumulated in the mucosa of patients with IBD, and the presence of CD4+CD69+CD103+ TRM cells was predictive of the development of flares. In vivo, functional impairment of TRM cells in mice with double knockout of the TRM-cell-associated transcription factors Hobit and Blimp-1 attenuated disease in several models of colitis, due to impaired cross-talk between the adaptive and innate immune system. Finally, depletion of TRM cells led to a suppression of colitis activity. Together, our data demonstrate a central role for TRM cells in the pathogenesis of chronic intestinal inflammation and suggest that these cells could be targets for future therapeutic approaches in IBD.

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 are increased in the gut of IBD patients and have a pro-inflammatory phenotype.
Fig. 2: CD4+, but not CD8+, intestinal TRM cells are associated with clinical flares of IBD.
Fig. 3: Hobit–Blimp-1 double deficiency protects from T cell transfer colitis.
Fig. 4: Hobit–Blimp-1 double deficiency leads to reduced leukocyte recruitment with reduced expression of pro-inflammatory cytokines.
Fig. 5: HobitBlimp-1 DKO mice are protected in various experimental colitis models.
Fig. 6: Protection of HobitBlimp-1 DKO mice in acute DSS colitis results from impaired adaptive-innate cross-talk.
Fig. 7: Depletion of TRM cells protects from experimental colitis.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon request. RNA-sequencing data (Fig. 4a,c) are accessible in the Gene Expression Omnibus database with the accession code GSE122880.

Change history

  • 07 March 2019

    In the version of this article initially published, a portion of the Acknowledgements section (“the Clinical Research Group CEDER of the German Research Council (DFG)”) was incorrect. The correct statement is as follows: “...the Collaborative Research Center TRR241 of the German Research Council (DFG)...”. The error has been corrected in the HTML and PDF version of the article.

References

  1. Baumgart, D. C. & Sandborn, W. J. Crohn’s disease. Lancet 380, 1590–1605 (2012).

    Article  Google Scholar 

  2. Strober, W., Fuss, I. & Mannon, P. The fundamental basis of inflammatory bowel disease. J. Clin. Invest. 117, 514–521 (2007).

    Article  CAS  Google Scholar 

  3. Macdonald, T. T. & Monteleone, G. Immunity, inflammation, and allergy in the gut. Science 307, 1920–1925 (2005).

    Article  CAS  Google Scholar 

  4. Kaser, A., Zeissig, S. & Blumberg, R. S. Inflammatory bowel disease. Annu. Rev. Immunol. 28, 573–621 (2010).

    Article  CAS  Google Scholar 

  5. Neurath, M. F. Cytokines in inflammatory bowel disease. Nat. Rev. Immunol. 14, 329–342 (2014).

    Article  CAS  Google Scholar 

  6. Lee, J. C. et al. Genome-wide association study identifies distinct genetic contributions to prognosis and susceptibility in Crohn’s disease. Nat. Genet. 49, 262–268 (2017).

    Article  CAS  Google Scholar 

  7. Ananthakrishnan, A. N. et al. Environmental triggers in IBD: a review of progress and evidence. Nat. Rev. Gastroenterol. Hepatol. 15, 39–49 (2018).

    Article  Google Scholar 

  8. Kaser, A. & Tilg, H. Novel therapeutic targets in the treatment of IBD. Expert. Opin. Ther. Targets 12, 553–563 (2008).

    Article  CAS  Google Scholar 

  9. Powrie, F. et al. Inhibition of Th1 responses prevents inflammatory bowel disease in scid mice reconstituted with CD45RBhi CD4+ T cells. Immunity 1, 553–562 (1994).

    Article  CAS  Google Scholar 

  10. Zundler, S. & Neurath, M. F. Pathogenic T cell subsets in allergic and chronic inflammatory bowel disorders. Immunol. Rev. 278, 263–276 (2017).

    Article  CAS  Google Scholar 

  11. Neurath, M. F. et al. The transcription factor T-bet regulates mucosal T cell activation in experimental colitis and Crohn’s disease. J. Exp. Med. 195, 1129–1143 (2002).

    Article  CAS  Google Scholar 

  12. Heller, F., Fuss, I. J., Nieuwenhuis, E. E., Blumberg, R. S. & Strober, W. Oxazolone colitis, a Th2 colitis model resembling ulcerative colitis, is mediated by IL-13-producing NK-T cells. Immunity 17, 629–638 (2002).

    Article  CAS  Google Scholar 

  13. Gerlach, K. et al. TH9 cells that express the transcription factor PU.1 drive T cell-mediated colitis via IL-9 receptor signaling in intestinal epithelial cells. Nat. Immunol. 15, 676–686 (2014).

    Article  CAS  Google Scholar 

  14. Ahern, P. P. et al. Interleukin-23 drives intestinal inflammation through direct activity on T cells. Immunity 33, 279–288 (2010).

    Article  CAS  Google Scholar 

  15. Maul, J. et al. Peripheral and intestinal regulatory CD4+CD25high T cells in inflammatory bowel disease. Gastroenterology 128, 1868–1878 (2005).

    Article  CAS  Google Scholar 

  16. Feagan, B. G. et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 369, 699–710 (2013).

    Article  CAS  Google Scholar 

  17. Vermeire, S. et al. Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet 384, 309–318 (2014).

    Article  CAS  Google Scholar 

  18. Zundler, S. & Neurath, M. F. Novel insights into the mechanisms of gut homing and antiadhesion therapies in inflammatory bowel diseases. Inflamm. Bowel Dis. 23, 617–627 (2017).

    Article  Google Scholar 

  19. Schenkel, J. M., Fraser, K. A., Vezys, V. & Masopust, D. Sensing and alarm function of resident memory CD8+ T cells. Nat. Immunol. 14, 509–513 (2013).

    Article  CAS  Google Scholar 

  20. Jiang, X. et al. Skin infection generates non-migratory memory CD8+ TRM cells providing global skin immunity. Nature 483, 227–231 (2012).

    Article  CAS  Google Scholar 

  21. Sheridan, B. S. et al. Oral infection drives a distinct population of intestinal resident memory CD8+ T cells with enhanced protective function. Immunity 40, 747–757 (2014).

    Article  CAS  Google Scholar 

  22. 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  Google Scholar 

  23. Schenkel, J. M. & Masopust, D. Tissue-resident memory T cells. Immunity 41, 886–897 (2014).

    Article  CAS  Google Scholar 

  24. Park, C. O. & Kupper, T. S. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat. Med. 21, 688–697 (2015).

    Article  CAS  Google Scholar 

  25. 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  Google Scholar 

  26. Mueller, S. N. & Mackay, L. K. Tissue-resident memory T cells: local specialists in immune defence. Nat. Rev. Immunol. 16, 79–89 (2016).

    Article  CAS  Google Scholar 

  27. Mackay, L. K. & Kallies, A. Transcriptional regulation of tissue-resident lymphocytes. Trends Immunol. 38, 94–103 (2017).

    Article  CAS  Google Scholar 

  28. 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  Google Scholar 

  29. 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  Google Scholar 

  30. van Gisbergen, K. P. J. M. et al. Mouse Hobit is a homolog of the transcriptional repressor Blimp-1 that regulates NKT cell effector differentiation. Nat. Immunol. 13, 864–871 (2012).

    Article  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  32. 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  Google Scholar 

  33. 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  Google Scholar 

  34. Stelma, F. et al. Human intrahepatic CD69+CD8+ T cells have a tissue resident memory T cell phenotype with reduced cytolytic capacity. Sci. Rep. 7, 6172 (2017).

    Article  Google Scholar 

  35. Ashouri, J. F. & Weiss, A. Endogenous Nur77 is a specific indicator of antigen receptor signaling in human T and B cells. J. Immunol. 198, 657–668 (2017).

    Article  CAS  Google Scholar 

  36. 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  Google Scholar 

  37. Teijaro, J. R. et al. Cutting edge: tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. J. Immunol. 187, 5510–5514 (2011).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  39. Zundler, S. et al. Three-dimensional cross-sectional light-sheet microscopy imaging of the inflamed mouse gut. Gastroenterology 153, 898–900 (2017).

    Article  Google Scholar 

  40. Fischer, A. et al. Differential effects of α4β7 and GPR15 on homing of effector and regulatory T cells from patients with UC to the inflamed gut in vivo. Gut 65, 1642–1664 (2016).

    Article  CAS  Google Scholar 

  41. Wirtz, S. et al. Chemically induced mouse models of acute and chronic intestinal inflammation. Nat. Protoc. 12, 1295–1309 (2017).

    Article  CAS  Google Scholar 

  42. Fernandez-Ruiz, D. et al. Liver-resident memory CD8+ T cells form a front-line defense against malaria liver-stage infection. Immunity 45, 889–902 (2016).

    Article  CAS  Google Scholar 

  43. Hong, S. et al. Differential regulation of P2X7 receptor activation by extracellular nicotinamide adenine dinucleotide and ecto-ADP-ribosyltransferases in murine macrophages and T cells. J. Immunol. 183, 578–592 (2009).

    Article  CAS  Google Scholar 

  44. Carta, S. et al. Cell stress increases ATP release in NLRP3 inflammasome-mediated autoinflammatory diseases, resulting in cytokine imbalance. Proc. Natl Acad. Sci. USA 112, 2835–2840 (2015).

    Article  CAS  Google Scholar 

  45. Turner, D. L. et al. Biased generation and in situ activation of lung tissue-resident memory CD4 T cells in the pathogenesis of allergic asthma. J. Immunol. 200, 1561–1569 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Oja, A. E. et al. The transcription factor Hobit identifies human cytotoxic CD4+ T cells. Front. Immunol. 8, 325 (2017).

    Article  Google Scholar 

  47. Vieira Braga, F. A. et al. Blimp-1 homolog Hobit identifies effector-type lymphocytes in humans. Eur. J. Immunol. 45, 2945–2958 (2015).

    Article  CAS  Google Scholar 

  48. Strober, W. & Fuss, I. J. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology 140, 1756–1767 (2011).

    Article  CAS  Google Scholar 

  49. Zundler, S. et al. Blockade of αEβ7 integrin suppresses accumulation of CD8+ and Th9 lymphocytes from patients with IBD in the inflamed gut in vivo. Gut 66, 1936–1948 (2017).

    Article  CAS  Google Scholar 

  50. Becker, C., Fantini, M. C. & Neurath, M. F. High resolution colonoscopy in live mice. Nat. Protoc. 1, 2900–2904 (2006).

    Article  CAS  Google Scholar 

  51. Erben, U. et al. A guide to histomorphological evaluation of intestinal inflammation in mouse models. Int. J. Clin. Exp. Pathol. 7, 4557–4576 (2014).

    PubMed  PubMed Central  Google Scholar 

  52. Klingberg, A. et al. Fully automated evaluation of total glomerular number and capillary tuft size in nephritic kidneys using lightsheet microscopy. J. Am. Soc. Nephrol. 28, 452–459 (2017).

    Article  CAS  Google Scholar 

  53. Ullrich, E. et al. BATF-dependent IL-7RhiGM-CSF+T cells control intestinal graft-versus-host disease. J. Clin. Invest. 128, 916–930 (2018).

    Article  Google Scholar 

  54. López-Posadas, R. et al. Rho-A prenylation and signaling link epithelial homeostasis to intestinal inflammation. J. Clin. Invest. 126, 611–626 (2016).

    Article  Google Scholar 

Download references

Acknowledgements

This work was funded by a grant of the German Research foundation to S.Z. (ZU 377/1-1). The research of S.Z., R.A., T.R., M.L., K.H., R.L.-P., C.N., I.A., and M.F.N. was supported by the Interdisciplinary Center for Clinical Research (IZKF) and the ELAN program of the University Erlangen-Nuremberg, the Fritz-Bender-Stiftung, the Else Kröner-Fresenius-Stiftung, the German Crohn’s and Colitis Foundation (DCCV), the Collaborative Research Center TRR241 of the German Research Council (DFG), the DFG topic program on Microbiota, the Emerging Field Initiative, the DFG Collaborative Research Centers 643, 796 and 1181 and the Litwin IBD Pioneers program of the Crohn’s and Colitis Foundation of America (CCFA). The authors thank J. Derdau, D. Dziony, J. Schuster, A. Marosan, and K. Enderle for excellent technical assistance. This work was performed in partial fulfillment of the requirements for obtaining the degree “Dr. rer. biol. hum.” for S.Z.

Author information

Authors and Affiliations

Authors

Contributions

S.Z., E.B., M. Spocinska, M. Slawik, and M.W. performed the experiments. S.Z., L.P.-V., R.S., R.A., T.R., M.L., K.H., R.L.-P., C.N., I.A., K.P.J.M.v.G., and M.F.N. provided clinical samples, protocols, or reagents, or designed experiments; A.B.E. and S.L. performed and analyzed RNA sequencing; S.Z., E.B., M. Spocinska, C.N., I.A., K.P.J.M.v.G., and M.F.N. analyzed and interpreted the data; S.Z. drafted the manuscript with the help of K.P.J.M.v.G. and M.F.N.; all of the authors critically revised the manuscript for important intellectual content.

Corresponding author

Correspondence to Markus F. Neurath.

Ethics declarations

Competing interests

M.F.N. has served as an advisor for Pentax, Giuliani, MSD, Abbvie, Janssen, Takeda and Boehringer. M.F.N. and S.Z. received research support from Takeda and Roche. The other authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Characterization of TRM cells in IBD.

(a) Flow cytometry of lamina propria mononuclear cells (LPMCs). Left panels: Representative dot plots indicating the expression of the T cell markers CD4 or CD8 in total LPMCs and CD69+CD103+ LPMCs. Right panels: Quantification of T cells expressing CD4 and/or CD8 among CD69+CD103+ LPMCs in CON (n = 4), UC (n = 3) and CD (n = 8). (b) Correlation of CD69 mRNA with CXCR6 and CD101 mRNA expression as determined by qPCR in intestinal tissue from patients with IBD (n = 35). Spearman’s R and regression lines are indicated, * P < 0.05. (c, e) mRNA expression of CRTAM1 (c, n = 5) and NR4A1 (e, n = 6) in CD3+CD69+ vs. CD3+CD69- T cells isolated from colonic lamina propria as determined by qPCR. Pairwise normalization to the expression in CD3+CD69+ T cells was performed. *** P < 0.001, paired two-tailed student’s t-test. (d) Quantitative flow cytometry of CD44 mean fluorescence intensity (MFI) in LPMC subsets. n = 16, *** P < 0.001, repeated-measures ANOVA with Newman-Keuls post-hoc test. (f) Analysis of TRM-associated molecules on CD69+ TRM cells from the intestinal lamina propria (LP) and T cells from the peripheral blood. Histograms of the expression of CD49a and CXCR6 representative for two independent experiments are shown. (g) Correlation of CD69 mRNA expression with mRNA expression of IFNG (n = 24), IL13 (n = 26), IL17A (n = 31), IL12A (n = 46), IL12B (n = 28) and TNF (n = 36) as determined by qPCR in intestinal tissue from patients with IBD. Spearman’s R and regression lines are indicated; * P < 0.05, ** P < 0.01, *** P < 0.001.

Supplementary Figure 2 CD4+ and CD8+ TRM cells in IBD.

(a) Histograms showing the expression of CD44, S1PR1 (CD363) and CD11a in CD69+ cells from the peripheral blood (PB) and on CD69+CD103- and CD69+CD103+ CD4+ lamina propria mononuclear cells (LPMCs) from endoscopic biopsies as indicated. Data are representative for two independent experiments. (b) Representative gating strategy for human CD4+ and CD8+ CD69+CD103+ TRM cells. From left to right: After exclusion of doublets, lymphocytes were selected in the foreward-/sideward-scatter. CD4+ and CD8+ T cells were identified and CD69 was plotted against CD103 to select CD69+CD103+ TRM cells. (c) The frequency of CD4+ and CD8+ CD69+ TRM cells (left) and total CD4+ and CD8+ T cells (right) in a cohort of 46 IBD patients was determined by flow cytometry and the respective median was calculated. Flare-free survival of patients with frequencies above (high) or below (low) the median was determined. P-values of one-sided log-rank comparison are indicated.

Supplementary Figure 3 Characterization of mouse TRM cells and comparable abundance of CD4+ Hobit–Blimp-1 double-knockout (DKO) and WT T cells and TRM cells in transfer colitis.

(a) Representative gating strategy for mouse CD4+ CD44+CD69+ TRM cells. From left to right: After exclusion of doublets and dead cells, lymphocytes were selected in the foreward-/sideward-scatter. CD3+CD4+ T cells were identified and CD44 was plotted against CD69 to select CD44+CD69+ TRM cells. (b) Flow cytometric quantification of CD44+CD69+CD103+ TRM frequency in LPMCs of Rag1-/- mice after transfer of naïve CD4+ T cells. Left panels: Representative dot plots of CD3+CD4+CD44+ LPMCs one week and six weeks after transfer; the frequency of CD69+CD103+ TRM cells among lymphoid LPMCs is indicated. Right panels: Quantification of CD103+ CD4+ TRM cell frequency over time. n = 5 (1+2 weeks), n = 4 (3+4 weeks), n = 3 (5 weeks). (c) Flow cytometry of the expression of the TRM-associated molecule CD49a on CD44+CD69+ TRM cells from the intestinal lamina propria (LP) and splenic CD44+ memory T cells. Histograms representative for two independent experiments are shown. (d) Immunohistochemistry staining for CD4 in colon tissue of Rag1-/- mice after transfer of naïve CD4+ WT, Hobit KO or DKO cells. Left panels: Representative images. Right panel: Quantitative data. Bars: 50 µm (upper row), 25 µm (lower row). n = 19 from two independent experiments. (e) Left: Competitive in vivo homing assay (n = 8) with CD4+ T cells from DKO (green) and WT (magenta) mice transferred to the ileocolic artery of Rag1-/- mice. Representative intravital confocal microscopy with arrows highlighting extravasated cells and quantification of homed cells per region of interest (ROI). Right: Non-competitive in vivo homing assay (n = 10) with CD4+ T cells from DKO and WT mice transferred to the ileocolic artery of Rag1-/- mice. Representative dot plots of homed CFSE+ cells in the LP and quantification. (f) Flow cytometry assessment of proliferation and apoptosis of anti-CD3/CD28-stimulated CD4+ WT and DKO T cells. Left panels: Representative flow cytometry showing CFSE dilution after culture of CD4+ T cells for 48 hours. Right panels: Quantification of proliferation and apoptosis as indicated. n = 10. (g) Flow cytometry of CD69 and CD103 expression in CD4+ LPMCs from Rag1-/- mice after transfer of WT or DKO cells. Representative histograms and quantitative data from one representative experiment with six mice out of two independent experiments are shown. (b, d) Center values – mean; error bars – s.e.m., (e,f) Boxplots – median with upper and lower quartile, whiskers – minimum and maximum. (g) Floating bars - median with minimum and maximum. (d) One-way ANOVA, (e-g) two-tailed student’s t-test. n.s. – not significant.

Supplementary Figure 4 Pathogenic mechanisms of TRM cells in T cell transfer colitis.

(a) Functional annotation analysis of previously published whole genome transcript analysis in WT and DKO NKT cells (Mackay et al. Science 352, 459–463) with Ingenuity Pathway Analysis (Qiagen). Using the “Diseases and Functions” mode the function “Recruitment” (subordinate to the function “Cell Movement”) was analyzed. Boxes are sized according to the respective -log(p) value and colored according to the z-score (right) assigned to the respective function. Fisher’s Exact test. (b) Expression of Cxcl3 mRNA in LPMCs from Rag1-/- mice transferred with WT, Hobit KO and DKO cells as determined by qPCR. n = 27 from three independent experiments. (c) Correlation of CD69 mRNA expression with mRNA expression of CCL3 (n = 37) and CCL4 (n = 34) as determined by qPCR in intestinal tissue from patients with IBD. Spearman’s R and regression lines are indicated; * P < 0.05. (d) mRNA expression of CCL3 and CCL4 in CD3+CD69+ vs. CD3+CD69- T cells isolated from colon lamina propria as determined by qPCR. Pairwise normalization to the expression in CD3+CD69+ T cells was performed. n = 6. (e, f) Expression of IL-2 in mRNA from LPMCs of RAG1–/– mice after transfer of WT, Hobit KO and DKO cells as determined by qPCR (e) and in supernatants from anti-CD3/CD28-stimulated LPMCs as determined by multiplex assay (n = 20 from two independent experiments). (g) Left panels: mRNA expression of Gzmb and Prf1 in LPMCs from the colon of Rag1-/- mice after transfer of WT, Hobit KO and DKO cells as determined by qPCR (n = 27 from three independent experiments). Right panel: Granzyme B concentration in the supernatants of anti-CD3/CD28-stimulated LPMCs from such mice as determined by ELISA (n = 30 from three independent experiments). (h) Culture of organoids in pure crypt culture medium (CCM) or with 25 % supernatant from WT T cells or from DKO T cells. Left panels: Representative bright-field and fluorescence microscopy images of organoid architecture and dead dye (red)/nuclei dye (blue) intensity, respectively; right panels: quantification of cell death. n = 24. (i) Immunohistochemistry staining for EpCAM (red) and TUNEL (green) in colon tissue from Rag1-/- mice after transfer of WT, Hobit KO or DKO cells. Left: Representative images, white arrows highlight EpCAM+TUNEL+ apoptotic epithelial cells. Right: Quantification of EpCAM+TUNEL+ cells per high power field (HPF). n = 21 from two independent experiments. Bars: 50 µm (upper row), 25 µm (lower row). (d, i) Center values – mean; error bars – s.e.m., (b, e-h) Boxplots – median with upper and lower quartile, whiskers – minimum and maximum. (b, d-i) * P < 0.05, ** P < 0.01, *** P < 0.001, one-way ANOVA with Newman-Keuls post-hoc testing (b, e-g, i) or paired (d) or unpaired (h) two-tailed student’s t-test.

Supplementary Figure 5 Pathogenic mechanisms of TRM cells in acute DSS-induced colitis.

(a) Immunohistochemistry staining for Foxp3 in colon tissue from WT, Hobit KO or DKO mice with acute DSS colitis. Left: Representative images, white arrows highlight positive cells. Right: Quantification of positive cells per high power field (HPF). n = 26 from three independent experiments; Bars: 50 µm (upper row), 25 µm (lower row). (b) Immunhohistochemistry staining for MPO in colon tissue from WT or DKO mice treated with DSS for three days. Left: Representative images, white arrows highlight positive cells. Right: Quantification of positive cells per HPF. n = 10 from two independent experiments. (c-f) Expression of pro-inflammatory cytokines in mRNA from LPMCs from the colon of WT, Hobit KO and DKO mice with acute DSS colitis as determined by qPCR (c, e) and in supernatants from anti-CD3/CD28-stimulated LPMCs as determined by multiplex assay (d, f). n = 17 (c, Il12a, e, Il13), n = 16 (e, Ifng, Il17a), n = 15 (c, Tnf) from two independent experiments, n = 8 from one representative out of two independent experiments (c, Il1b), n = 22 from three independent experiments (d, f). (g) mRNA expression of Gzmb in LPMCs from the colon of WT, Hobit KO and DKO mice with acute DSS colitis as determined by qPCR (left panel, n = 14 from two independent experiments). Granzyme B concentration in the supernatants of anti-CD3/CD28-stimulated LPMCs from such mice as determined by ELISA (right panel, n = 15 from two independent experiments). (h) Immunohistochemistry staining for EpCAM (red) and TUNEL (green) in the colon of WT, Hobit KO and DKO mice with acute DSS colitis Left: Representative images, white arrows highlight EpCAM+TUNEL+ apoptotic epithelial cells. Right: Quantification of EpCAM+TUNEL+ cells per high power field (HPF). n = 19 from two independent experiments. Bars: 50 µm (upper row), 25 µm (lower row). (a, b, h) Center values – mean; error bars – s.e.m.; (c-g) Boxplots – median with upper and lower quartile, whiskers – minimum and maximum; floating bars - median with minimum and maximum. (a-h) * P < 0.05, ** P < 0.01, *** P < 0.001, two-tailed student’s t-test (b, d IL-12, e Il17a, f IL-13), one-way ANOVA with Newman-Keuls post hoc test (all other panels).

Supplementary Figure 6 Depletion of TRM cells via DT.

(a, b) Flow cytometry of CD4+ T cell (a) and CD103+ CD4+ TRM cell frequency (b) in LPMCs of Rag1-/- mice after transfer of naive CD4+ Hobit-DTR cells and treatment without (“no DT”) or with early or late diphtheria toxin (DT). (a) Representative histograms for 10 mice from one experiment are shown. (b) Left: representative flow cytometry, the frequency of CD69+CD103+ TRM cells among lymphoid LPMCs is indicated. Right: Quantification of CD69+CD103+ TRM cells among total lymphoid LPMCs; n = 21 from three independent experiments. (c) Immunohistochemistry staining for CD4 (left, n = 24) and MPO (right, n = 22) in colon tissue of Rag1-/- mice after transfer of naive CD4+ Hobit-DTR cells and treatment without or with early and late DT. Representative images from three independent experiments with white arrows highlighting CD4+ or MPO+ cells and quantification of CD4+ or MPO+ cells per high power field (HPF). Bars: 50µm (upper row), 25µm (lower row). (d) mRNA expression of chemokines and pro-inflammatory cytokines in LPMCs from the colon of Rag1-/- mice after transfer of naiveCD4+ Hobit-DTR cells and treatment without or with early and late DT as determined by qPCR. n = 14 (Ccl3, Ccl4, Ifng), n = 13 (Cxcl2, Il17, Il1b), n = 12 (Il12a) from two independent experiments, n = 21 from three independent experiments (Il13). (e) Concentration of pro-inflammatory cytokines in the supernatants of anti-CD3/CD28-stimulated LPMCs from the colon of Rag1-/- mice after transfer of naiveCD4+ Hobit-DTR cells and treatment without or with early and late DT as determined by multiplex assay. n = 10 (IFN-γ, IL-13), n = 9 (IL-17A, IL-1β). = 9–10. (b-d) Boxplots – median with upper and lower quartile, whiskers – minimum and maximum. (e) Floating bars - median with minimum and maximum. * P < 0.05, ** P < 0.01, *** P < 0.001, two-tailed student’s t-test (d, Il13, Il17a) or one-way ANOVA with Newman-Keuls post-hoc comparison (all other panels).

Supplementary Figure 7 Depletion of TRM cells via NAD.

(a) Flow cytometry of P2X7-Art2.2-coexpression. Representative control stainings for Fig. 7f showing the expression of P2X7 and Art2.2 in spleen CD3- and CD3+ cells. The frequency of double-positive cells is indicated. (b) Frequency of CD44+CD69- effector memory T cells (TEM) in mice treated with or without NAD. n = 22. (c) Representative (left) and quantitative (right) flow cytometry of CD1d tetramer+ NKT cells in the colon of mice treated with or without NAD as well as representative gating control flow cytometry of CD1d tetramer+ NKT cells in the liver (far right). n = 14. (d) Frequency of CD3+ T and CD1d tetramer+ NKT cells among lymphoid LPMCs after treatment with PBS. n = 8. (e) Immunohistochemistry staining for CD4 (n = 16) and MPO (n = 17) in colon tissue of WT mice with TNBS colitis after treatment with or without NAD from three independent experiments. (f) Flow cytometry of Art2.2 expression in T cells versus non-T cells. Left: Representative histograms of Art2.2 expression on CD4+ T cells, CD3- non T cells and fluorescence minus one (FMO) control for Art2.2. Right: Quantitative expression. n = 10 mice from three independent experiments. (b-f) Boxplots – median with upper and lower quartile, whiskers – min and max. n.s. – not significant, * P < 0.05, ** P < 0.01, *** P < 0.001, paired (d, f) or unpaired (b, c, e) two-tailed student’s t-test.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–3

Reporting Summary

Supplementary Video 1: Light-sheet fluorescence microscopy

Representative animation of a colon sample from a Rag1–/– mouse after transfer of naïve CD4+ T cells from a Hobit–Blimp-1 double-knockout mouse. Whole-mount staining for CD4 (red) and autofluorescence signal (grey) were recorded by light-sheet fluorescence microscopy

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zundler, S., Becker, E., Spocinska, M. et al. Hobit- and Blimp-1-driven CD4+ tissue-resident memory T cells control chronic intestinal inflammation. Nat Immunol 20, 288–300 (2019). https://doi.org/10.1038/s41590-018-0298-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0298-5

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