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:

Fibroblastic reticular cells enhance T cell metabolism and survival via epigenetic remodeling

Abstract

Lymph node fibroblastic reticular cells (FRCs) respond to signals from activated T cells by releasing nitric oxide, which inhibits T cell proliferation and restricts the size of the expanding T cell pool. Whether interactions with FRCs also support the function or differentiation of activated CD8+ T cells is not known. Here we report that encounters with FRCs enhanced cytokine production and remodeled chromatin accessibility in newly activated CD8+ T cells via interleukin-6. These epigenetic changes facilitated metabolic reprogramming and amplified the activity of pro-survival pathways through differential transcription factor activity. Accordingly, FRC conditioning significantly enhanced the persistence of virus-specific CD8+ T cells in vivo and augmented their differentiation into tissue-resident memory T cells. Our study demonstrates that FRCs play a role beyond restricting T cell expansion—they can also shape the fate and function of CD8+ T cells.

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: FRCs upregulate both immunosuppressive and stimulatory molecules in response to activated T cells.
Fig. 2: FRCs enhance CD8+ T cell cytokine production independently of Nos2 expression.
Fig. 3: FRC-derived IL-6 is necessary and sufficient for the enhanced production of CD8+ T cell cytokines.
Fig. 4: FRCs or IL-6 alter enhancers in activated CD8+ T cells that regulate distinct patterns of gene expression.
Fig. 5: FRCs or IL-6 amplify metabolic and survival pathways in activated CD8+ T cells via epigenetic and transcriptional alterations.
Fig. 6: FRCs enhance the bioenergetic state of activated CD8+ T cells.
Fig. 7: FRC-derived signals extend the longevity of virus-specific CD8+ T cells during influenza infection.

Similar content being viewed by others

Data availability

All data that relate to or support the observations described in this study are available from the corresponding authors upon reasonable request. The ATAC-seq and RNA-seq data have been deposited in the Gene Expression Omnibus (GEO) database under accession no. GSE136905. The Microarray data are accessible through GEO accession no. GSE136958.

References

  1. Mueller, S. N. & Germain, R. N. Stromal cell contributions to the homeostasis and functionality of the immune system. Nat. Rev. Immunol. 9, 618–629 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Turley, S. J., Fletcher, A. L. & Elpek, K. G. The stromal and haematopoietic antigen-presenting cells that reside in secondary lymphoid organs. Nat. Rev. Immunol. 10, 813–825 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Roozendaal, R. & Mebius, R. E. Stromal cell–immune cell interactions. Annu. Rev. Immunol. 29, 23–43 (2011).

  4. Malhotra, D. et al. Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks. Nat. Immunol. 13, 499–510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Malhotra, D., Fletcher, A. L. & Turley, S. J. Stromal and hematopoietic cells in secondary lymphoid organs: partners in immunity. Immunol. Rev. 251, 160–176 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Luther, S. A., Vogt, T. K. & Siegert, S. Guiding blind T cells and dendritic cells: a closer look at fibroblastic reticular cells found within lymph node T zones. Immunol. Lett. 138, 9–11 (2011).

    Article  CAS  PubMed  Google Scholar 

  7. Perez-Shibayama, C., Gil-Cruz, C. & Ludewig, B. Fibroblastic reticular cells at the nexus of innate and adaptive immune responses. Immunol. Rev. 289, 31–41 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Alexandre, Y. O. & Mueller, S. N. Stromal cell networks coordinate immune response generation and maintenance. Immunol. Rev. 283, 77–85 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Brown, F. D. & Turley, S. J. Fibroblastic reticular cells: organization and regulation of the T lymphocyte life cycle. J. Immunol. 194, 1389–1394 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Fletcher, A. L., Acton, S. E. & Knoblich, K. Lymph node fibroblastic reticular cells in health and disease. Nat. Rev. Immunol. 15, 350–361 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chang, J. E. & Turley, S. J. Stromal infrastructure of the lymph node and coordination of immunity. Trends Immunol. 36, 30–39 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Luther, S. A., Tang, H. L., Hyman, P. L., Farr, A. G. & Cyster, J. G. Coexpression of the chemokines ELC and SLC by T zone stromal cells and deletion of the ELC gene in the plt/plt mouse. Proc. Natl Acad. Sci. USA 97, 12694–12699 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bajenoff, M. et al. Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes. Immunity 25, 989–1001 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Asperti-Boursin, F., Real, E., Bismuth, G., Trautmann, A. & Donnadieu, E. CCR7 ligands control basal T cell motility within lymph node slices in a phosphoinositide 3-kinase independent manner. J. Exp. Med. 204, 1167–1179 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Worbs, T., Mempel, T. R., Bolter, J., von Andrian, U. H. & Forster, R. CCR7 ligands stimulate the intranodal motility of T lymphocytes in vivo. J. Exp. Med. 204, 489–495 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Denton, A. E., Roberts, E. W., Linterman, M. A. & Fearon, D. T. Fibroblastic reticular cells of the lymph node are required for retention of resting but not activated CD8(+) T cells. Proc. Natl Acad. Sci. USA 111, 12139–12144 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Schumann, K. et al. Immobilized chemokine fields and soluble chemokine gradients cooperatively shape migration patterns of dendritic cells. Immunity 32, 703–713 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Marsland, B. J. et al. CCL19 and CCL21 induce a potent proinflammatory differentiation program in licensed dendritic cells. Immunity 22, 493–505 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Link, A. et al. Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells. Nat. Immunol. 8, 1255–1265 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Acton, S. E. et al. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Nature 514, 498–502 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Astarita, J. L. et al. The CLEC-2-podoplanin axis controls the contractility of fibroblastic reticular cells and lymph node microarchitecture. Nat. Immunol. 16, 75–84 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Lukacs-Kornek, V. et al. Regulated release of nitric oxide by nonhematopoietic stroma controls expansion of the activated T cell pool in lymph nodes. Nat. Immunol. 12, 1096–1104 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Siegert, S. et al. Fibroblastic reticular cells from lymph nodes attenuate T cell expansion by producing nitric oxide. PloS ONE 6, e27618 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Khan, O. et al. Regulation of T cell priming by lymphoid stroma. PloS ONE 6, e26138 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cremasco, V. et al. B cell homeostasis and follicle confines are governed by fibroblastic reticular cells. Nat. Immunol. 15, 973–981 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang, Y. et al. Plasma cell output from germinal centers is regulated by signals from Tfh and stromal cells. J. Exp. Med. 215, 1227–1243 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Huang, H. Y. et al. Identification of a new subset of lymph node stromal cells involved in regulating plasma cell homeostasis. Proc. Natl Acad. Sci. USA 115, E6826–E6835 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Brown, G. C. Regulation of mitochondrial respiration by nitric oxide inhibition of cytochrome c oxidase. Biochim. Biophys. Acta Bioenerg. 1504, 46–57 (2001).

    Article  CAS  Google Scholar 

  29. Wink, D. A. et al. DNA deaminating ability and genotoxicity of nitric-oxide and its progenitors. Science 254, 1001–1003 (1991).

    Article  CAS  PubMed  Google Scholar 

  30. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  31. Buck, M. D., O’Sullivan, D. & Pearce, E. L. T cell metabolism drives immunity. J. Exp. Med. 212, 1345–1360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. MacIver, N. J., Michalek, R. D. & Rathmell, J. C. Metabolic regulation of T lymphocytes. Annu. Rev. Immunol. 31, 259–283 (2013).

  33. Chang, C.-H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wang, R. N. et al. The transcription factor myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35, 871–882 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tyrakis, P. A. et al. S-2-hydroxyglutarate regulates CD8(+) T-lymphocyte fate. Nature 540, 236–241 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Finlay, D. K. et al. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8(+) T cells. J. Exp. Med. 209, 2441–2453 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Roychoudhuri, R. et al. BACH2 regulates CD8(+) T cell differentiation by controlling access of AP-1 factors to enhancers. Nat. Immunol. 17, 851–860 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Li, Q. S., Eppolito, C., Odunsi, K. & Shrikant, P. A. Antigen-induced Erk1/2 activation regulates Ets-1-mediated sensitization of CD8+ T cells for IL-12 responses. J. Leukoc. Biol. 87, 257–263 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Kurachi, M. et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8(+) T cells. Nat. Immunol. 15, 373–383 (2014).

    Article  CAS  PubMed Central  Google Scholar 

  40. Xin, G. et al. A critical role of IL-21-induced BATF in sustaining CD8-T-cell-mediated chronic viral control. Cell Rep. 13, 1118–1124 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gray, S. M., Amezquita, R. A., Guan, T. X., Kleinstein, S. H. & Kaech, S. M. Polycomb repressive complex 2-mediated chromatin repression guides effector CD8(+) T cell terminal differentiation and loss of multipotency. Immunity 46, 596–608 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Portt, L., Norman, G., Clapp, C., Greenwood, M. & Greenwood, M. T. Anti-apoptosis and cell survival: a review. Biochim. Biophys. Acta Mol. Cell Res. 1813, 238–259 (2011).

    Article  CAS  Google Scholar 

  44. Scott-Browne, J. P. et al. Dynamic changes in chromatin accessibility occur in CD8(+) T cells responding to viral infection. Immunity 45, 1327–1340 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yu, B. F. 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 

  46. Scharer, C. D., Bally, A. P. R., Gandham, B. & Boss, J. M. Cutting edge: chromatin accessibility programs CD8 T cell memory. J. Immunol. 198, 2238–2243 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. van der Windt, G. J. W. et al. Mitochondrial respiratory capacity is a critical regulator of CD8(+) T cell memory development. Immunity 36, 68–78 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Pan, Y. D. 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 

  49. Hunter, C. A. & Jones, S. A. IL-6 as a keystone cytokine in health and disease. Nat. Immunol. 16, 448–457 (2015).

    Article  CAS  PubMed  Google Scholar 

  50. Kopf, M. et al. Impaired immune and acute-phase responses in interleukin-6-deficient mice. Nature 368, 339–342 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Ladel, C. H. et al. Lethal tuberculosis in interleukin-6-deficient mutant mice. Infect. Immun. 65, 4843–4849 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lauder, S. N. et al. Interleukin-6 limits influenza-induced inflammation and protects against fatal lung pathology. Eur. J. Immunol. 43, 2613–2625 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Pasztoi, M. et al. Mesenteric lymph node stromal cell-derived extracellular vesicles contribute to peripheral de novo induction of Foxp3+ regulatory T cells. Eur. J. Immunol. 47, 2142–2152 (2017).

  54. Fletcher, A. et al. Reproducible isolation of lymph node stromal cells reveals site-dependent differences in fibroblastic reticular cells. Front. Immunol. 2, 35 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Yu, M. et al. Fibroblastic reticular cells of the lymphoid tissues modulate T cell activation threshold during homeostasis via hyperactive cyclooxygenase-2/prostaglandin E-2 axis. Sci. Rep. 7, 3350 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Knoblich, K. et al. The human lymph node microenvironment unilaterally regulates T-cell activation and differentiation. PloS Biol. 16, 24 (2018).

    Article  CAS  Google Scholar 

  57. Rodda, L. B. et al. Single-cell RNA sequencing of lymph node stromal cells reveals niche-associated heterogeneity. Immunity 48, 1014–1028 (2018).

    Article  CAS  Google Scholar 

  58. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Liberzon, A. et al. The molecular signatures database hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Pircher, H., Burki, K., Lang, R., Hengartner, H. & Zinkernagel, R. M. Tolerance induction in double specific T-cell receptor transgenic mice varies with antigen. Nature 342, 559–561 (1989).

    Article  CAS  PubMed  Google Scholar 

  61. Laidlaw, B. J. et al. Cooperativity between CD8+ T cells, non-neutralizing antibodies, and alveolar macrophages is important for heterosubtypic influenza virus immunity. PloS Pathog. 9, e1003207 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mueller, S. N. et al. Qualitatively different memory CD8+ T cells are generated after lymphocytic choriomeningitis virus and influenza virus infections. J. Immunol. 185, 2182–2190 (2010).

    Article  CAS  PubMed  Google Scholar 

  63. Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 1193–1203 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Dunham, I. et al. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the US National Institutes of Health (grant no. RO1 5R01DK074500 to S.J.T. and W.N.H.; no. P01AI108545 to A.H.S; no. T32 CA207021 to M.W.L) and by a Howard Hughes Medical Institute Gilliam Fellowship for Advanced Study (to F.D.B). We thank members of the Turley, Haining and Sharpe laboratories for scientific insights and discussions. We also thank A. E. Mayfield for critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

F.D.B. conceived and conducted most of the experiments, analyzed and interpreted data and wrote the manuscript. D.R.S. conducted experiments and analyzed and interpreted data. J.G., M.W.L., V.L.-K., F.A.S., H-J.K., K.B.Y., S.J.H.R., K.B. and V.N.K. conducted experiments and interpreted data. J.D.T. and I.A.S. discussed data and provided technical assistance. V.C., N.N.D. and B.D.M. discussed and interpreted data. A.H.S., W.N.H. and S.J.T. directed the study, analyzed and interpreted results and wrote the manuscript.

Corresponding authors

Correspondence to Arlene H. Sharpe, W. Nicholas Haining or Shannon J. Turley.

Ethics declarations

Competing interests

V.N.K. and S.J.T. are employees of Genentech. W.N.H. is an employee of Merck. V.C. is an employee of Novartis. F.D.B. is an employee of Neon Therapeutics. The authors declare no competing interests.

Additional information

Peer review information L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Integrated supplementary information

Supplementary Figure 1 FRCs dampen IFN-γ production yet enhance the production of IL-2 and TNF in newly activated CD8+ T cells.

(a) Gating strategy to highlight CD8+ T cells within whole splenocyte mixtures (24 h post anti-CD3/CD28 activation). (b) ICS of IFN-γ in CD8+ T cells activated in splenocyte mixtures via anti-CD3/CD28 for 24 h or activated via anti-CD3/CD28 for 24 h in the presence of wild-type FRCs. (c) Summary of results in b (n = 4 [Activated T cells] and n = 5 [Activated T cells + WT FRCs] biologically independent cell cultures per group). ****P < 0.0001 (two-tailed Student’s t-test). Data in c are composite of two biologically independent experiments (mean ± s.d). (d) ICS for IL-2 (top) and TNF (bottom) in purified CD8+ T cells activated via plate-bound anti-CD3 and soluble anti-CD28 for 24 h or activated via plate-bound anti-CD3 and soluble anti-CD28 in the presence of WT FRCs. (e) Summary of results in d as percentage of cytokine positive cells (n = 4 biologically independent cell cultures per group) **P = 0.0053 (IL-2), **P = 0.0018 (TNF) (two-tailed Student’s t-test) Data in e are a composite of two biologically independent experiments (mean ± s.d).

Supplementary Figure 2 CD8+ T cells activated alone or near Nos2−/− FRCs experience the same degree of initial activation and proliferation.

(a) CD25 expression on resting CD8+ T cells (resting), those activated in splenocyte mixtures via soluble anti-CD3/CD28 for 24 h (activated) or activated via anti-CD3/CD28 for 24 h in the presence of Nos2−/− FRCs. Top panel, representative plots; bottom panel, summary (n = 2 biologically independent cell cultures per group). (b) CFSE dilution in same CD8+ T cell conditions as a at 48 h post activation. Top panel, representative plots; bottom panel, summary (n = 3 biologically independent cell cultures per group). ns P = 0.2829 (two-tailed Student’s t-test). Data are representative of two biologically independent experiments (mean ± s.d).

Supplementary Figure 3 Characteristics of chromatin accessible regions, RNA transcripts and epigenetic signatures in CD8+ T cells.

(a) Scatterplot displaying correlation in peak intensity between biological replicates for each CD8+ T cell condition. The conditions were CD8+ T cells sorted following activation in whole splenocyte mixture via soluble anti-CD3/CD28 for 48 h (Act), activated with anti-CD3/CD28 in presence of Nos2−/− FRCs (FRC) or activated with anti-CD3/CD28 plus 100ng/ml recombinant IL-6 (IL-6). (b) Principal component analysis of biological replicates for CD8+ T cells under each condition across all 81, 821 ChARs. (c) ATAC-seq fragment sizes for all three conditions (n = 3 biologically independent cell cultures per group). (d) Fold enrichment of regions annotated for histone marks and enhancer states for all three conditions. Data represent one experiment with three biologically independent cell cultures per group. (e) Scatterplot displaying correlation in gene expression between biological replicates for each CD8+ T cell condition. The conditions were CD8+ T cells sorted following activation in whole splenocyte mixtures via soluble anti-CD3/CD28 for 48 h (Act), activated with anti-CD3/CD28 in presence of Nos2−/− FRCs (FRC) or activated with anti-CD3/CD28 plus 100ng/ml recombinant IL-6 (IL-6). (f) Principal component analysis of biological replicates for CD8+ T cells under each condition. (n = 3 biologically independent cell cultures [FRC] and 2 biologically independent cell cultures [Act and IL-6]). (g) Display of gene expression using hierarchical clustering of replicates by condition with Pearson correlation. Data represent one experiment (n = 3 biologically independent cell cultures [FRC] and 2 biologically independent cell cultures [Act only and Act + IL-6]). (h) Overlap of H3K4me3 peaks identified in naive, terminal effector and memory precursor cells (Gray et al. 2017) with the FRC, IL-6 and Act conditions. (i) Overlap of ATAC-seq profiles corresponding to naive, effector and memory T cells (Sen et al. 2016) with the FRC, IL-6 and Act conditions.

Supplementary Figure 4 IL-6 enhances phospho S6 levels and neutral lipid accumulation in activated T cells.

(a) Phospho flow cytometry of p-S6 levels in resting CD8+ T cells or those activated in whole splenocyte mixtures via anti-CD3/CD28 for 48 h or activated via anti-CD3/CD28 for 48 h in the presence of recombinant IL-6 (100ng/ml). Left panel, representative plot; right panel, summary (n = 6 biologically independent cell cultures per group) ****P < 0.0001 (two-tailed Student’s t-test) (b) Bodipy staining in CD8+ T cells either solely activated for 48 h with anti-CD3/CD28 or activated via anti-CD3/CD28 in the presence of recombinant IL-6 (100ng/ml). Left panel, representative plot; right panel, summary (n = 4 [Activated T cells] and 5 [Activated T cells + IL-6] biologically independent cell cultures) ns P = 0.0720 (two-tailed Student’s t-test) Data are a composite of two biologically independent experiments (mean ± s.d).

Supplementary Figure 5 FRCs extend the longevity of activated CD8+ T cells in vitro and during acute LCMV infection.

(a) Frequency of viable CD8+ T cells activated in splenocyte mixtures via soluble anti-CD3/CD28 for 48 h versus CD8+ T cells activated via anti-CD3/CD28 for 48 h in the presence of Nos2−/− FRCs. (b) Summary of results in a (n = 2 biologically independent cell cultures per group). (c) Frequency of viable T cells from a after being sorted and allowed to rest in alpha-mem media (supplemented with 10% fetal bovine serum and 1% penicillin streptomycin) for three days. (d) Summary of results in c (n = 2 biologically independent cell cultures per group). Data in b and d are representative of two biologically independent experiments. (e) Competitive frequencies of pre-activated P14 T cells before transfer (Day 0) and 7 days following transfer into wild type recipients infected with LCMV Armstrong. Transferred T cells were recovered from the lungs of infected animals. Top panel, representative plots; bottom panel, summary (n = 3 mice per group) *P = 0.0185 (two-tailed ratio paired t-test). (f) Frequency of competitive mixes at Day 0 and 7 days post LCMV infection. Transferred T cells were recovered from the spleen of infected animals. Top panel, representative plots; bottom panel, summary (n = 10 mice per group).****P < 0.0001 (two-tailed ratio paired t-test). Data in f are representative of three biologically independent experiments. (g) BrdU incorporation in transferred P14 T cells 8 days after the onset of influenza infection. Transferred T cells were recovered from the lungs of infected animals. (h) Summary of results in g (n = 5 mice per group). ns P = 0.4011 (two-tailed paired t-test) (mean ± s.d).

Supplementary Figure 6 Neutralizing IL-6 prior to adoptive transfer decreases the absolute number of recovered IL-2+ and TNF+ P14 T cells.

(a) Competitive frequencies of P14 T cells activated in whole splenocyte mixture with anti-CD3/CD28 in the presence of Nos2−/− FRCs and 10 μg/ml anti-IL-6 blocking antibody versus P14 T cells activated under the same conditions with 10 μg/ml of isotype control antibody. Il6−/− recipients were used for the transfers in a. Top panel, representative plots; bottom panel, summary. (n = 7 mice per group) ***P = 0.0004 (two-tailed ratio paired t-test) (b) Representative plots of ICS for IL-2 (left) and TNF (right) from CD8+ T cells recovered from the lungs of infected mice 9 days post influenza infection (n = 7 mice per group) (c) Summary of results in b (n = 7 mice per group) ns P = 0.0562 (IL-2), ns P = 0.6148 (TNF) (two-tailed paired t-test). (d) Absolute number of recovered cytokine positive P14 T cells. (n = 7 mice per group) *P = 0.0375 (IL-2), *P = 0.0265 (TNF) (two-tailed paired t-test), data in c shown as mean ± s.d. For b-d, the transferred cells were re-challenged in vitro with gp33 peptide (KAVYNFATC) for 6 h in the presence of Brefeldin A, followed by ICS staining to measure cytokine production.

Supplementary Figure 7 Generation and infection of bone marrow chimeras.

(a) Percent chimerism (following tail vein bleed) in C57BL/6J (WT) or Il6−/− host 6-12 weeks after transfer of donor cells. Representative plots shown in a. (b) Left panel, summary of data in a; right panel, hematopoietic cell lineage reconstitution in irradiated recipients. n = 10 mice per group (mean ± s.d) (c) Frequency of CD45 lymph node stromal cells. Top panel, representative plots; bottom panel, graphic display (n = pooled popliteal lymph nodes from 3 mice). (d) ICS for IL-6 in stromal populations from popliteal lymph nodes. The lymph nodes were harvested on day 8 following CFA/OVA immunization and OT-1 T cell injection. 100 μg Brefeldin A was injected intravenously (tail vein) to block protein transport 5 h prior to lymph node harvest and digest. Single cell suspensions were placed in Brefeldin A for an additional 2 h prior to ICS staining. Top panel, representative plots; bottom panel, graphic display. Data are representative of two biologically independent experiments with pooled popliteal lymph nodes from 3 wild-type C57BL/6J mice. (e) Frequency of CD44+, CD8+ T cells within the lungs of WT and Il6−/− bone marrow chimeras. Analysis conducted in animals that successfully cleared influenza infection from Fig. 7h (n = 6 of 10 mice for WT recipients and n = 1 of 10 mice for Il6−/− recipients) (mean ± SEM).

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brown, F.D., Sen, D.R., LaFleur, M.W. et al. Fibroblastic reticular cells enhance T cell metabolism and survival via epigenetic remodeling. Nat Immunol 20, 1668–1680 (2019). https://doi.org/10.1038/s41590-019-0515-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0515-x

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