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:

Lymph nodes fuel KLF2-dependent effector CD8+ T cell differentiation during chronic infection and checkpoint blockade

Abstract

Exhausted CD8+ T (TEX) cell responses are maintained by precursors of exhausted T (TPEX) cells that possess high self-renewal and developmental potential. TPEX cells also drive the proliferative burst of effector T cells upon therapeutic immune checkpoint blockade (ICB). However, the spatial context and signals that regulate their differentiation and function are not well defined. Here we identify developmental and functional compartmentalization of TPEX and TEX cells across secondary lymphoid organs during chronic infection. We show that stem-like CD62L+ TPEX and effector-like CX3CR1+ TEX cells constitute a distinct developmental lineage that is promoted by the lymph node (LN) microenvironment and dependent on the transcription factor KLF2. LNs act as a niche in which migratory dendritic cells provide antigen and costimulatory signals to maintain the proliferative fitness of CD62L+ TPEX cells and generation of CX3CR1+ TEX cells. Moreover, LNs exclusively drive the proliferative burst and systemic dissemination of CX3CR1+ TEX cells during ICB. Thus, our findings identify a unique role for LNs in the maintenance of T cell differentiation and function during systemic chronic infection and ICB therapy.

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: LNs preserve the stemness, proliferative fitness and polyfunctionality of T cells.
Fig. 2: LNs support de novo generation of CX3CR1+ T cells in chronic infection.
Fig. 3: A precursor–progeny relationship defines the differentiation of CD62L+ TPEX to CX3CR1+ TEX cells.
Fig. 4: The transcription factor KLF2 instructs the lineage development of CD62L+ TPEX and CX3CR1+ TEX cells in LNs.
Fig. 5: CCR7 and S1PR1 are required for optimal development of CD62L+ TPEX and CX3CR1+ TEX cells.
Fig. 6: Migratory DCs provide a functional niche in LNs to promote T cell fitness.
Fig. 7: LNs drive the generation of TEFF cells in response to ICB.

Similar content being viewed by others

Data availability

The scRNA-seq data have been deposited in the Gene Expression Omnibus under accession no. GSE268824. All other data are available in the article and Supplementary Information.

Code availability

All analyses can be assessed at https://gitlab.dzne.de/ag-beyer/klf2_lcmv.git. Correcting for ambient genes and GOEA was performed on the docker image alefrol94/scrnaseq.analysis:reticulate using the package scrnaseq.analysis v.0.01. Any additional packages installed were tracked with the renv package v.0.14.0. Trajectory analysis with Palantir and CellRank was performed on the docker image wollmilchsau/scanpy_sccoda:latest (from 4 November 2021). The conda environments used for the analysis have been saved as YAML files.

References

  1. Baessler, A. & Vignali, D. A. A. T cell exhaustion. Annu. Rev. Immunol. 42, 179–206 (2024).

    CAS  PubMed  Google Scholar 

  2. McLane, L. M. et al. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 37, 457–495 (2019).

    CAS  PubMed  Google Scholar 

  3. Blank, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Barber, D. L. et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687 (2006).

    CAS  PubMed  Google Scholar 

  5. Frebel, H. et al. Programmed death 1 protects from fatal circulatory failure during systemic virus infection of mice. J. Exp. Med. 209, 2485–2499 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Odorizzi, P. M. et al. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J. Exp. Med. 212, 1125–1137 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Waldman, A. D. et al. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 20, 651–668 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rausch, L. & Kallies, A. Molecular mechanisms governing CD8 T cell differentiation and checkpoint inhibitor response in cancer. Annu. Rev. Immunol. 43, 515–543 (2025).

    CAS  PubMed  Google Scholar 

  9. Hudson, W. H. et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity 51, 1043–1058 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Zander, R. et al. CD4+ T cell help is required for the formation of a cytolytic CD8+ T cell subset that protects against chronic infection and cancer. Immunity 51, 1028–1042 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, Z. et al. TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision. Immunity 51, 840–855 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Giles, J. R. et al. Shared and distinct biological circuits in effector, memory and exhausted CD8+ T cells revealed by temporal single-cell transcriptomics and epigenetics. Nat. Immunol. 23, 1600–1613 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Daniel, B. et al. Divergent clonal differentiation trajectories of T cell exhaustion. Nat. Immunol. 23, 1614–1627 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Kasmani, M. Y. et al. Clonal lineage tracing reveals mechanisms skewing CD8+ T cell fate decisions in chronic infection. J. Exp. Med. 220, e20220679 (2022).

    PubMed  PubMed Central  Google Scholar 

  15. Kanev, K. et al. Proliferation-competent Tcf1+ CD8 T cells in dysfunctional populations are CD4 T cell help independent. Proc. Natl Acad. Sci. USA 116, 20070–20076 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Im, S. J. et al. PD-1+ stemlike CD8 T cells are resident in lymphoid tissues during persistent LCMV infection. Proc. Natl Acad. Sci. USA 117, 4292–4299 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Beltra, J.-C. et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52, 825–841 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Sandu, I. et al. Exhausted CD8+T cells exhibit low and strongly inhibited TCR signaling during chronic LCMV infection. Nat. Commun. 11, 4454 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Gabriel, S. S. et al. Transforming growth factor-β-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection. Immunity 54, 1698–1714 (2021).

    CAS  PubMed  Google Scholar 

  21. Dähling, S. et al. Type 1 conventional dendritic cells maintain and guide the differentiation of precursors of exhausted T cells in distinct cellular niches. Immunity 55, 656–670 (2022).

    PubMed  Google Scholar 

  22. Tsui, C. et al. MYB orchestrates T cell exhaustion and response to checkpoint inhibition. Nature 609, 354–360 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Wijesinghe, S. K. M. et al. Lymph-node-derived stem-like but not tumor-tissue-resident CD8+ T cells fuel anticancer immunity. Nat. Immunol. 26, 1367–1383 (2025).

    CAS  PubMed  Google Scholar 

  24. Sandu, I. et al. Landscape of exhausted virus-specific CD8 T cells in chronic LCMV infection. Cell Rep. 32, 108078 (2020).

    CAS  PubMed  Google Scholar 

  25. Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019).

    CAS  PubMed  Google Scholar 

  26. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Argüello, R. J. et al. SCENITH: a flow cytometry-based method to functionally profile energy metabolism with single-cell resolution. Cell Metab. 32, 1063–1075 (2020).

    PubMed  PubMed Central  Google Scholar 

  29. Mueller, S. N. & Ahmed, R. High antigen levels are the cause of T cell exhaustion during chronic viral infection. Proc. Natl Acad. Sci. USA 106, 8623–8628 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Richter, K. et al. Antigen amount dictates CD8+ T-cell exhaustion during chronic viral infection irrespective of the type of antigen presenting cell. Eur. J. Immunol. 42, 2290–2304 (2012).

    CAS  PubMed  Google Scholar 

  31. Utzschneider, D. T. et al. High antigen levels induce an exhausted phenotype in a chronic infection without impairing T cell expansion and survival. J. Exp. Med. 213, 1819–1834 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Man, K. et al. The transcription factor IRF4 is essential for TCR affinity-mediated metabolic programming and clonal expansion of T cells. Nat. Immunol. 14, 1155–1165 (2013).

    CAS  PubMed  Google Scholar 

  33. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Fagerberg, E. et al. KLF2 maintains lineage fidelity and suppresses CD8 T cell exhaustion during acute LCMV infection. Science 387, eadn2337 (2025).

    CAS  PubMed  Google Scholar 

  35. Carlson, C. M. et al. Kruppel-like factor 2 regulates thymocyte and T-cell migration. Nature 442, 299–302 (2006).

    CAS  PubMed  Google Scholar 

  36. Bai, A. et al. Krüppel-like factor 2 controls T cell trafficking by activating l-selectin (CD62L) and sphingosine-1-phosphate receptor 1 transcription. J. Immunol. 178, 7632–7639 (2007).

    CAS  PubMed  Google Scholar 

  37. Weber, J. P. et al. ICOS maintains the T follicular helper cell phenotype by down-regulating Krüppel-like factor 2. J. Exp. Med. 212, 217–233 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Yeo, J.-C. et al. Klf2 is an essential factor that sustains ground state pluripotency. Cell Stem Cell 14, 864–872 (2014).

    CAS  PubMed  Google Scholar 

  39. Bromley, S. K. et al. Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat. Immunol. 6, 895–901 (2005).

    CAS  PubMed  Google Scholar 

  40. Cyster, J. G. & Schwab, S. R. Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu. Rev. Immunol. 30, 69–94 (2012).

    CAS  PubMed  Google Scholar 

  41. Salmon, H. et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 44, 924–938 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Jansen, C. S. et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Duraiswamy, J. et al. Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. Cancer Cell 39, 1623–1642 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Schenkel, J. M. et al. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity 54, 2338–2353 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Meiser, P. et al. A distinct stimulatory cDC1 subpopulation amplifies CD8+ T cell responses in tumors for protective anti-cancer immunity. Cancer Cell 41, 1498–1515 (2023).

    CAS  PubMed  Google Scholar 

  46. Domenjo-Vila, E. et al. XCR1+ DCs are critical for T cell-mediated immunotherapy of chronic viral infections. Cell Rep. 42, 112123 (2023).

    CAS  PubMed  Google Scholar 

  47. Bosteels, C. et al. Inflammatory type 2 cDCs acquire features of cDC1s and macrophages to orchestrate immunity to respiratory virus infection. Immunity 52, 1039–1056 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Brown, C. C. et al. Transcriptional basis of mouse and human dendritic cell heterogeneity. Cell 179, 846–863 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Ardouin, L. et al. Broad and largely concordant molecular changes characterize tolerogenic and immunogenic dendritic cell maturation in thymus and periphery. Immunity 45, 305–318 (2016).

    CAS  PubMed  Google Scholar 

  50. Cabeza-Cabrerizo, M. et al. Dendritic cells revisited. Annu. Rev. Immunol. 39, 131–166 (2021).

    CAS  PubMed  Google Scholar 

  51. Alvarez, D. et al. Mechanisms and consequences of dendritic cell migration. Immunity 29, 325–342 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Ugur, M. et al. Lymph node medulla regulates the spatiotemporal unfolding of resident dendritic cell networks. Immunity 56, 1778–1793 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Humblin, E. et al. Sustained CD28 costimulation is required for self-renewal and differentiation of TCF-1+ PD-1+ CD8 T cells. Sci. Immunol. 8, eadg0878 (2023).

    PubMed  PubMed Central  Google Scholar 

  54. Yan, Y. et al. CX3CR1 identifies PD-1 therapy-responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy. JCI Insight 3, e97828 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Wu, T. D. et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature 579, 274–278 (2020).

    CAS  PubMed  Google Scholar 

  57. Rahim, M. K. et al. Dynamic CD8+ T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell 186, 1127–1143 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Abdelfatah, E. et al. Predictive and prognostic implications of circulating CX3CR1+ CD8+ T cells in non-small cell lung cancer patients treated with chemo-immunotherapy. Cancer Res. Commun. 3, 510–520 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Hui, E. et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355, 1428–1433 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Kamphorst, A. O. et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355, 1423–1427 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Viramontes, K. M. et al. PD-1 immune checkpoint blockade and PSGL-1 inhibition synergize to reinvigorate exhausted T cells. Front. Immunol. 13, 869768 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Zander, R. & Cui, W. Exhausted CD8+ T cells face a developmental fork in the road. Trends Immunol. 44, 276–286 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Szabo, P. A. et al. Location, location, location: tissue resident memory T cells in mice and humans. Sci. Immunol. 4, eaas9673 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Miron, M. et al. Human lymph nodes maintain TCF-1hi memory T cells with high functional potential and clonal diversity throughout life. J. Immunol. 201, 2132–2140 (2018).

    CAS  PubMed  Google Scholar 

  67. Mayoux, M. et al. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy. Sci. Transl. Med. 12, eaav7431 (2020).

    CAS  PubMed  Google Scholar 

  68. Dammeijer, F. et al. The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell 38, 685–700 (2020).

    CAS  PubMed  Google Scholar 

  69. Connolly, K. A. et al. A reservoir of stem-like CD8+ T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci. Immunol. 6, eabg7836 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Huang, Q. et al. The primordial differentiation of tumor-specific memory CD8+ T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell 185, 4049–4066 (2022).

    CAS  PubMed  Google Scholar 

  71. Devi, S. et al. Adrenergic regulation of the vasculature impairs leukocyte interstitial migration and suppresses immune responses. Immunity 54, 1219–1230 (2021).

    CAS  PubMed  Google Scholar 

  72. Yamazaki, C. et al. Critical roles of a dendritic cell subset expressing a chemokine receptor, XCR1. J. Immunol. 190, 6071–6082 (2013).

    CAS  PubMed  Google Scholar 

  73. Nüssing, S. et al. Efficient CRISPR/Cas9 gene editing in uncultured naive mouse T cells for in vivo studies. J. Immunol. 204, 2308–2315 (2020).

    PubMed  Google Scholar 

  74. Utzschneider, D. T. et al. T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat. Immunol. 14, 603–610 (2013).

    CAS  PubMed  Google Scholar 

  75. Hadian-Jazi, M. et al. Data reduction for serial crystallography using a robust peak finder. J. Appl. Cryst. 54, 1360–1378 (2021).

    CAS  Google Scholar 

  76. Hadian-Jazi, M. & Sadri, A. A Python package based on robust statistical analysis for serial crystallography data processing. Acta Crystallogr. D Struct. Biol. 79, 820–829 (2023).

    CAS  Google Scholar 

  77. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Choudhary, S. & Satija, R. Comparison and evaluation of statistical error models for scRNA-seq. Genome Biol. 23, 27 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Castanza, A. S. et al. Extending support for mouse data in the Molecular Signatures Database (MSigDB). Nat. Methods 20, 1619–1620 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Howe, D. G. et al. Model organism data evolving in support of translational medicine. Lab. Anim. 47, 277–289 (2018).

    Google Scholar 

  82. Kolberg, L. et al. gprofiler2–an R package for gene list functional enrichment analysis and namespace conversion toolset g:Profiler.F1000Res. 9, ELIXIR-709 (2020).

    PubMed  PubMed Central  Google Scholar 

  83. Angerer, P. et al. destiny: diffusion maps for large-scale single-cell data in R. Bioinformatics 32, 1241–1243 (2016).

    CAS  PubMed  Google Scholar 

  84. Buettner, F. et al. f-scLVM: scalable and versatile factor analysis for single-cell RNA-seq. Genome Biol. 18, 212 (2017).

    PubMed  PubMed Central  Google Scholar 

  85. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    PubMed  PubMed Central  Google Scholar 

  86. Setty, M. et al. Characterization of cell fate probabilities in single-cell data with Palantir. Nat. Biotechnol. 37, 451–460 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Weiler, P. et al. CellRank 2: unified fate mapping in multiview single-cell data. Nat. Methods 21, 1196–1205 (2024).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  89. Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Jin, S. et al. CellChat for systematic analysis of cell-cell communication from single-cell transcriptomics. Nat. Protoc. 20, 180–219 (2025).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Melbourne Cytometry Platform and Walter and Eliza Hall Institute of Medical Research (WEHI) Cytometry Facility for providing flow cytometry services and the Doherty Institute Biological Resource Facility for animal husbandry and management. We thank the WEHI Centre for Dynamic Imaging for providing image acquisition and analysis services and the Molecular Genomics Core facility of the Peter MacCallum Cancer Centre for providing sequencing services. We thank members of the Kallies laboratory for critical reading of the manuscript and for providing feedback. This work was supported by the National Health and Medical Research Council of Australia (Investigator grant no. APP1194779 to D.T.U., Ideas grant no. 2028921 to C.T. and L.R.), the Australian Research Council Discovery Early Career Research Award (no. DE240100827 to C.T.). L.R. is supported by a Deutsche Forschungsgemeinschaft (DFG) (German Research Foundation) Postdoctoral Fellowship. L.H., L.W., C.G.d.G., D.M. and S.K.W. are supported by a Melbourne Research Scholarship. L.W. is supported by a China Scholarship Council-University of Melbourne PhD scholarship. M.D.B. is supported by the Helmholtz Association and the DFG (SFB1454 project no. 432325352, IGK2168/2 project no. 272482170). M.D.B. is a member of the excellence cluster ImmunoSensation2 (EXC2151 project no. 390873048). Z.A. was supported by the DFG through IRTG2168 no. 272482170 and EXC2151 no. 390873048.

Author information

Authors and Affiliations

Authors

Contributions

C.T. and A.K. conceived, designed and supervised the study, and wrote the manuscript. C.T. and L.H., supported by L.W., C.G.d.G., V.C.W., D.M., C.-H.S., H.H., S.L., K.R.W., S.K.W., M.L.M., D.S. and L.R., performed all the experimental work. N.P., A.F., L.Q., D.R., M.H.-J., L.D. and J.S. performed the computational analyses. D.T.U., C.H., S.N.M., M.D.B., S.B. and Z.A. provided biological and technical resources and contributed to student supervision.

Corresponding authors

Correspondence to Carlson Tsui or Axel Kallies.

Ethics declarations

Competing interests

A.K. has received research support from Pfizer. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Stephen Jameson and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Nick Bernard in collaboration with the rest of the Nature Immunology editorial team. 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 Lymph nodes preserve stemness, proliferative and metabolic fitness, and polyfunctionality of CD8+ T cells.

(a–e) Naïve congenically marked (CD45.1+) P14 CD8+ T cells were adoptively transferred into naïve wildtype (CD45.2+) mice, which were subsequently infected with LCMV-Docile. On d25 post-infection (p.i.) CD8+ T cells from spleens and pooled inguinal, brachial, axillary, cervical and mandibular lymph nodes (LN) were analysed using flow cytometry. (a) Quantification showing IL-2 production by P14 TPEX cells (n = 7). (b) Flow cytometry plots and quantification showing IFN-γ and TNF production in CD62L+ and CD62L TPEX cells from spleen and LN after incubation with gp33 peptide (n = 11). (c) Flow cytometry plots and quantification of Ki67+ cells among the P14 TEX cells (n = 21). (d) Quantification showing ex vivo puromycin uptake in splenic and LN TEX cells. (e) Flow cytometry plots and quantification showing IFN-γ and TNF production in P14 TEX cells from spleen and LN after incubation with gp33 peptide (n = 10). (f–k) Naïve wildtype mice were infected with LCMV-Docile. On d25 p.i. CD8+ T cells from spleen and pooled LN were analysed using flow cytometry. (f) Flow cytometry plots and quantification showing the expression of PD-1 and TOX in endogenous polyclonal antigen-responsive CD8+ T cells in spleen and LN in comparison to naïve CD8+ T cells (n = 10). (g) Flow cytometry plots and quantification of TPEX cells among endogenous PD-1+CD8+ T cells in spleen and LN (n = 10). (h) Flow cytometry plots and quantification of CD62L+ cells among endogenous TPEX cells in spleen and LN (n = 20). (i) Flow cytometry plots and quantification showing the frequencies of Ki67+ cells among endogenous TPEX and TEX cells (n = 12). (j) Quantification showing ex vivo puromycin uptake of spleen and LN-derived endogenous antigen-responsive CD8+ T cells (n = 8). (k) Flow cytometry plots and quantification showing IFN-γ and TNF production in endogenous PD-1+CD8+ T cells from spleen and LN after stimulation in the presence or absence of gp33 peptide (n = 26). Flow cytometry plots are representative. Dots in graphs (a–k) represent individual mice; bars represent median. Quantification and statistics derive from unpaired two-tailed t tests (a, c, e, g–i, k) and one-way ANOVA (b, d, f) and are based on all data points across at least two independent experiments.

Extended Data Fig. 2 Functional superiority of endogenous antigen-responsive CD8+ T cells residing in lymph nodes.

(a, b) P14 T cells expressing IRF4-tdTomato were analysed using flow cytometry on d25 p.i. with chronic LCMV. The expression of IRF4 in TPEX and TEX subsets in the spleen and LN was quantified. (a) Flow cytometry plots and (b) quantification showing the expression of IRF4 in exhausted T cell subsets (n = 8). (c–h) Naïve wildtype mice were infected with LCMV-Docile. On d26 p.i., CD8+ T cells from spleen and pooled inguinal, brachial, axillary, cervical and mandibular lymph nodes (LN) were analysed using flow cytometry. (c, d) Flow cytometry plots and quantification showing the frequencies of (c) CX3CR1+ (n = 21) and (d) CD101+ cells (n = 15) among endogenous TEX cells (gated as TCF1). (e, f) Flow cytometry plots and quantification showing expression of (e) CXCR6 (n = 9) and (f) Ly108 (n = 9) among endogenous CX3CR1+ TEX cells in the spleen and LN. (g) Flow cytometry plots and corresponding quantification showing the frequencies of Ki67+ among Ly108intCX3CR1+ and Ly108loCX3CR1+ TEX cells in spleen and LN (n = 14). (h) Flow cytometry plots and corresponding quantification showing the frequencies of Ki67+CX3CR1+ cells among endogenous PD-1+CD8+ T cells in spleen and LN (n = 8). (i–n) Naïve congenically marked (CD45.1+) P14 CD8+ T cells were adoptively transferred into naïve wildtype (CD45.2+) mice, which were subsequently infected with LCMV-cl13. On d32 p.i., P14 T cells in the spleen and pooled LN were analysed using flow cytometry. (i, j) Flow cytometry plots and quantification showing the frequencies of (i) TPEX cells among P14 cells (n = 9) and (j) CD62L+ cells among TPEX cells (n = 9). (k) Flow cytometry plots and quantification showing the frequencies of CD101+ cells among P14 TEX cells (n = 12). (l) Flow cytometry plots and quantification showing the frequencies of Ki67+CX3CR1+ cells among P14 cells in spleen and LN (n = 10). (m, n) Flow cytometry plots and quantification showing the frequencies of Ly108intCX3CR1+ cells among (m) P14 cells (n = 23) and (n) endogenous antigen-responsive CD8+PD-1+ cells (n = 9). Flow cytometry plots are representative. Dots in graphs (b–n) represent individual mice; bars represent median. Quantification and statistics derive from unpaired two-tailed t tests (c–f, h–n) and one-way ANOVA (a, g) and based on all data points across at two (f, h, i–l, n) or three (b–e, g, m) independent experiments.

Extended Data Fig. 3 CX3CR1+ TEX cells develop in lymph nodes.

(a–d) Naïve congenically marked (CD45.1+) P14 CD8+ T cells were adoptively transferred into naïve wildtype (CD45.2+) mice, which were subsequently infected with LCMV-Docile. On d26 p.i., mice were subjected to daily FTY720 treatment for 4 days. On d30 p.i., P14 T cells from the spleen and lymph nodes (LN) were analysed using flow cytometry. (a, b) Quantification showing the (a) frequencies and (b) numbers of P14 TPEX and TEX cells in the presence and absence of FTY720 in the spleen and LN (n = 10). (c) Representative flow cytometry plots and corresponding quantification showing the frequencies of Ly108intCX3CR1+ cells among endogenous antigen-responsive PD-1+CD8+ T cells (n = 10). (d) Quantification showing the absolute numbers of endogenous TPEX and TEX cells in the presence and absence of FTY720 in the spleen and LN (n = 10). (e–i) Naïve congenically marked (CD45.1+) P14 CD8+ T cells were adoptively transferred into naïve wildtype (CD45.2+) mice, which were subsequently infected with LCMV-Docile. P14 cells in the spleen and LN were harvested on d30 p.i. and subjected to scRNA-seq. (e) Normalised gene expression of key identity genes of exhausted T cell populations projected onto the UMAPs. (f) Heatmap showing the expression of cluster signature genes for each cluster from across all cell populations as annotated in Fig. 3a. (g) Annotated UMAP plot of single P14 T cells from spleen and LN of LCMV-Docile infected mice showing expression of the stem-like TPEX cell transcriptional signature22. (h) Box plot showing transcriptional diversity scores (CytoTRACE)33 among splenic and LN-derived TPEX (Spleen: n = 2175; LN: n = 995) and TEX cells (Spleen: n = 2266; LN: n = 1346). (i) Annotated volcano plots showing differentially expressed (DE) genes between splenic and LN total Cx3cr1+ TEX cells (left) and between Cx3cr1+S1pr5+ TEX and Cx3cr1+ TEX cells independent of tissue of origin (right). Dots in graphs (b–d) represent individual mice; bars represent median or mean. Box plot in (h) defines range (maxima and minima), interquartile range and median of the dataset. Quantification and statistics derive from two-tailed t tests (h) and one-way ANOVA (a–d) and based on all data points across at least two independent experiments (a–d).

Extended Data Fig. 4 The development of CX3CR1+ TEX cells is defined by KLF2 expression.

(a) Diffusion maps showing all annotated cell clusters (as per Fig. 3a) and cluster distribution according to origin, spleen and LN. (b) UMAP representations (as per Fig. 3a) showing enrichment of transcriptional signatures for Trajectory 1 (Cx3cr1+ TEX cells, top 50 genes) and Trajectory 2 (Cd101+ TEX cells, top 50 genes), with reciprocal enrichments of these signatures in TPEX cells bordered. (c) Box plots showing enrichment scores of Trajectory 1 and Trajectory 2 signatures among Sell+ and Sell TPEX cell clusters. (d) Diffusion maps (as per Fig. 3e) showing the expressions of selected Traj. 2 genes that were also expressed in some TPEX cells. (e–g) UMAP representation after correction using muVI single cell trajectory analysis using Palantir and CellRank of P14 cells scRNA-seq data showing (e) cell differentiation originating from Tcf7+ TPEX cells and (f) progressing into either Cx3cr1+ (Traj. 1; upper) or Cd101+ (Traj. 2; lower) TEX cells. (g) Box plot showing fate probabilities towards Traj. 1 among Sell+ and Sell TPEX cell clusters. (h–o) Naïve congenically marked (CD45.1+) P14 CD8+ T cells were adoptively transferred into naïve wildtype (CD45.2+) mice, which were subsequently infected with LCMV-Docile or LCMV-cl13. On d28 p.i. CD8+ T cells from spleen and pooled inguinal, brachial, axillary, cervical and mandibular lymph nodes (LN) were analysed using flow cytometry. (h, i) Flow cytometry plots showing the expression of (h) KLF2 and CD69, and (i) CD69 and CD62L expression among KLF2+ and KLF2 TPEX cells in the spleen and LN. (j) Quantification showing the frequency of CD69-expressing TPEX cells stratified by KLF2 expression (n = 9). (k, l) Flow cytometry plots showing the expression of (k) KLF2 and CD69, and (l) CD69 and CD101 among KLF2+ and KLF2 TEX cells in the spleen and LN. (m) Quantification showing the frequencies of CD69-expressing TEX cells stratified by KLF2 expression (n = 9). (n, o) Flow cytometry plots and corresponding quantification showing the frequencies KLF2+ cells among (n) TPEX (n = 6) and (o) TEX cell subsets (n = 6) in the spleen and LN. Dots in graphs (j, m–o) represent individual mice; bars represent median. Flow cytometry plots are representative. Box plots in (c, g) define ranges (maxima and minima), interquartile ranges and medians of the datasets. Quantification and statistics derive from unpaired two-tailed t tests (c, g) and one-way ANOVA (j, m–o) and based on all analysed mice across at least two independent experiments.

Extended Data Fig. 5 KLF2 drives the differentiation of CD62L+ TPEX and CX3CR1+ TEX cells in chronic LCMV infection.

(a–h) Naïve congenically marked KLF2-deficient (Klf2Δ) and control P14 CD8+ T cells were adoptively co-transferred at a 1:1 ratio into naïve wildtype mice, which were subsequently infected with chronic LCMV-cl13 and analysed on d28 p.i. (a) Flow cytometry plots and quantification showing the expression of KLF2 protein in control and Klf2Δ P14 cells in spleen and LN (n = 6). (b) Quantification showing the absolute counts of TPEX and TEX cells in the spleen and LN on d28 p.i. (n = 13). (c) Flow cytometry plots and quantification showing the frequencies of control and Klf2Δ CD62L+ and CD62L TPEX cells in the LN (n = 10). (d) Quantification showing the accumulation of control and Klf2Δ P14 cells in different tissues (n = 7). (e) Flow cytometry plots and quantification showing the lack of CD62L+ and CX3CR1+ cells among Klf2Δ P14 cells in different tissues (n = 6). (f) Flow cytometry plots and quantification showing the frequencies of control and Klf2Δ CX3CR1+ and CD101+ TEX cells in LN (n = 10). (g) Flow cytometry plots and quantification showing the expression of CD69 among control and Klf2Δ P14 cells in the spleen (n = 10). (h) Quantification of PD-1 and TOX expression in control and KLF2-deficient P14 cells in the spleen (n = 8). (i–n) Naïve congenically marked KLF2-deficient (Klf2Δ) and control P14 CD8+ T cells were adoptively co-transferred at a 1:1 ratio into naïve wildtype mice, which were subsequently infected with chronic LCMV-Docile and analysed on d24 p.i. (i) Schematic of experimental setup. (j) Flow cytometry plots and quantification showing the expression of KLF2 protein in control and Klf2Δ P14 cells in spleen and LN (n = 9). (k) Quantification showing the relative accumulation of control and Klf2Δ TPEX and TEX cells in the spleen and LN on d24 p.i. (n = 10). (l) Quantification showing the frequencies of control and Klf2Δ CD62L+ and CD62L TPEX cells in the spleen and LN (n = 10). (m) Quantification showing the frequencies of control and Klf2Δ CX3CR1+ TEX cells in the spleen and LN (n = 10). (n) Quantification showing the expression of CD69 among control and Klf2Δ P14 cells in the spleen and LN (n = 10). Flow cytometry plots are representative. Dots in graphs (b–f, j–n) represent individual mice; bars represent median. Quantification and statistics derive from two-tailed t tests (g, k) and one-way ANOVA (a–f, h, j, l–n) and are based on all data points across at least two independent experiments.

Extended Data Fig. 6 The KLF2-instructed differentiation program involves molecules associated with lymph node trafficking, including CCR7 and S1PR1, which contribute to CX3CR1+ TEX cell differentiation.

(a) Enrichment of a transcriptional signature for stem-like Sell+ TPEX cells (ref. 22) in control versus Klf2Δ TPEX cells. (b) Annotated UMAP plot showing single P14 T cells in spleen and LN. (c, d) Enrichment of transcriptional signatures projected onto a UMAP plot among genes (c) upregulated and (d) down-regulated comparing control and Klf2Δ TPEX cells, showing enrichment in Cx3cr1+ TEX cells and Cd101+ TEX cells, respectively. (e) Enrichment of transcriptional signatures among single P14 T cells derived from spleen and LN based on upregulated and down-regulated genes between control and Klf2Δ TPEX cells, showing reciprocal enrichment in splenic and LN P14 cells, respectively (Spleen TPEX: n = 2175; LN TPEX: n = 995; Spleen TEX: n = 2266; LN TEX: n = 1346). (f–h) Naïve congenically marked control and CCR7-deficient (Ccr7Δ) P14 CD8+ T cells were generated using CRISPR-Cas9-mediated editing. Control and Ccr7Δ P14 cells were cultured in the presence of IL7 for 2 days in vitro and surface expression of CCR7 were analysed using flow cytometry. (f) Schematic of in vitro experimental setup. (g) Flow cytometry plot and quantification of CCR7 expression in control and Ccr7Δ P14 cells (n = 10). (h) Quantification showing the absolute numbers of control and Ccr7Δ TEX cell subsets in the spleen and LN on d26 p.i. (n = 10). (i–l) Naïve congenically marked control and S1pr1Δ P14 CD8+ T cells co-transferred at a 1:1 ratio into naïve wildtype mice, which were subsequently infected with chronic LCMV-cl13 and analysed on d7 and d26 p.i. (i) Flow cytometry plots and quantification showing the expression of CD69 among control and S1pr1Δ P14 cells in the blood at the specified time points (n = 9). (j) Flow cytometry plots and quantification showing the expression of CD69 among control and S1pr1Δ P14 cells in the spleen and LN on d26 p.i. (n = 9). (k) Quantification showing the relative accumulation of control and S1pr1Δ TPEX and TEX cells in the spleen and LN on d7 (n = 8) and d26 p.i. (n = 13). (l) Flow cytometry plots and corresponding quantification showing the frequencies of Ki67+CX3CR1+ cells among control and S1pr1Δ P14 cells in spleen and LN (n = 9). Results in (g) represent four independent experiments. Flow cytometry plots are representative. Dots in graphs (h–l) represent individual mice; bars represent median. Box plots in (d, e) define ranges (maxima and minima), interquartile ranges and medians of the datasets. Quantification and statistics derive from two-tailed t tests (e, g) and one-way ANOVA (d, h–l) and based on all data points across at least two independent experiments (h–l).

Extended Data Fig. 7 LCMV induces influx of migratory cDC1 that express high levels immunostimulatory molecules.

(a–c) Naïve wildtype mice were infected with chronic LCMV-Docile. The spleens and pooled inguinal, brachial, axillary, cervical and mandibular lymph nodes (LN) were harvested on d30 p.i. and enriched for the CD11c+MHCII+ cells by flow cytometric sorting. The samples were then subjected to scRNA-seq. (a) Normalised expression of myeloid/DC marker genes projected onto the UMAPs. (b) Bubble plot comparing the transcript expression of genes related to DC activation among the main DC subsets obtained from the spleen and LN. (c) Annotated bubble plot showing predicted cell-cell interaction classes between different myeloid/DC subsets (senders) and TPEX cells (receivers). (d–h) Naïve wildtype mice were infected with chronic LCMV-Docile. The spleens and pooled LN were harvested on d25 p.i., and DC subsets were analysed using flow cytometry. (d) Schematic of the experimental setup. (e) Representative flow cytometry plots and quantification showing the frequencies of CCR7+ cells among CD11c+MHCII+ cells in the spleen and LN on d25 p.i. (n = 13). (f, g) Representative flow cytometry plots and quantification showing the frequencies of migratory DC defined by (f) CD8αneg (Uninfected: n = 10; LCMV: n = 21) and (g) MHCIIhi (Uninfected: n = 10; LCMV: n = 11) among all LN-derived cDC1 (XCR1+) in infected and uninfected mice. (h) Representative flow cytometry plots and (i) quantification showing the expression of MHCII, CD40, CD86, CD80 and IL-12 among splenic and LN cDC1 subsets on d25 p.i. (n = 6). Dots in graphs (e–g, i) represent individual mice; bars represent median. Quantification and statistics derive from two-tailed t tests (e, f) and one-way ANOVA (g, l) and based on all data points across at least two independent experiments.

Extended Data Fig. 8 TPEX cells interact with cDC1 in the paracortical T cell zones of lymph nodes.

(a–f) Naïve P14 CD8+ T cells, which ubiquitously express tdTomato (ref. 71) were adoptively transferred into naïve Xcr1Venus reporter mice (ref. 72), which were subsequently infected with LCMV-Docile. Lymph nodes (LN) were harvested on d25 p.i., fixed and imaged using confocal microscopy. (a) Representative confocal micrograph and quantification showing the distribution of B220+, TCF1+, CD11c+ CD68+ and P14 cells. (b) Analysis and (c) quantification showing the spatial distribution of TPEX (tdTomato+TCF1+) and TEX (tdTomato+TCF1) cells within different anatomical regions of a LN (Follicular areas: n = 10; Paracortical TCZ: n = 9; Medullary regions: n = 7). (d) Representative confocal micrograph and (e) quantification showing the distribution of XCR1+ dendritic cells within different regions of the LN (Follicular areas: n = 10; Paracortical TCZ: n = 9; Medullary regions: n = 7). (f) Representative confocal micrographs and quantification showing the expression of CD11c in XCR1+ dendritic cells found within different anatomical regions of the LN (TCZ: n = 40; non-TCZ: n = 59). (g–j) Mixed bone marrow chimeric mice containing Ccr7−/− and Xcr1DTR (ref. 72) haematopoietic cells were infected with LCMV-Docile before the administering of Diphtheria toxin (DTX) starting on d18 p.i. P14 cells in the spleen and LN were analysed using flow cytometry on d25 p.i. (g) Representative flow cytometry plots and (h) quantification showing the frequencies of XCR1+ cells in spleen and LN in untreated or DTX treated mice (n = 8). (i) Quantification showing absolute numbers of P14 TPEX and TEX cell subsets in the presence and absence of cDC1 depletion in the spleen and LN (n = 9). (j) Representative flow cytometry plots and corresponding quantification showing the frequencies of Ly108intCX3CR1+ cells among P14 cells in the presence and absence of cDC1 depletion in the spleen and LN (n = 9). Dots in graphs represent (b, e) individual manually defined regions of interest, (f) individual cells, or individual mice (h–j), and bars represent median, and bars represent median. Quantification and statistics derive from unpaired two-tailed t tests (f) and one-way ANOVA (c, e, h–j) and based on all data points across at least two independent experiments.

Extended Data Fig. 9 CD28 signalling promotes exhausted T cell fitness in lymph nodes.

(a–e) Naïve congenically marked (CD45.1+) P14 cells were adoptively transferred into wildtype (CD45.2+) mice, which were then infected with LCMV-Docile and subjected to B7 blockade using anti-CD80/86 antibodies treatment starting on d18 p.i. Endogenous antigen-responsive PD-1+ as well as P14 cells were analysed on d25 p.i. using flow cytometry. (a) Flow cytometry plots and quantification showing the frequencies of Ki67+ cells among endogenous TPEX cells in the spleen and lymph nodes (LN) on d25 p.i. (n = 15). (b) Flow cytometry plots and quantification showing IFN-γ and TNF production of from spleen and LN-derived P14 cells after incubation with gp33 peptide ex vivo (Untreated: n = 20; Anti-B7: n = 16). (c) Flow cytometry plots and quantification showing the frequencies of intermediate Ly108intCX3CR1+ cells among P14 T cells in the spleen and LN on d25 p.i. (Untreated: n = 20; Anti-B7: n = 12). (d) Flow cytometry plots and quantification showing the frequencies of Ki67+CX3CR1+ cells among endogenous antigen-responsive CD8+ T cells in the spleen and LN on d25 p.i. (n = 15). (e) Flow cytometry plots and quantification showing the frequencies of intermediate Ly108intCX3CR1+ cells among endogenous antigen-responsive CD8+ T cells in the spleen and LN on d25 p.i. (n = 18). (f) Flow cytometry plots and quantification showing the surface expression of CD28 among P14 TPEX and TEX cells in the spleen and LN on d25 p.i. (n = 8). Flow cytometry plots are representative. Dots in graphs (a–f) represent individual mice; bars represent median. Quantification and statistics derive from one-way ANOVA (a–f) and based on all data points across at least two (f) or three (a–e) independent experiments.

Extended Data Fig. 10 Immune checkpoint blockade induces TPEX and CX3CR1+ TEX cell proliferation and egress from lymph nodes.

(a) Naïve congenically marked (CD45.1+) P14 CD8+ T cells were adoptively transferred to naïve wildtype (CD45.2+) mice, which were subsequently infected with LCMV-Docile. A single dose of anti-PD-L1 was given on d26 p.i. with or without subsequent daily FTY720 treatment. CD8+ T cells were analysed using flow cytometry on d30 p.i. (a) Representative flow cytometry plots and quantification of Ki67+ cells among the P14 TPEX cells in the spleen and lymph nodes (LN) (Ctrl: n = 17; ICB/FTY720: n = 24). (b–e) Naïve wildtype mice were infected with LCMV-Docile. A single dose of anti-PD-L1 was given on d26 p.i. with or without subsequent daily FTY720 treatment. Endogenous CD8+ T cells in the blood, spleen and pooled LN were analysed using flow cytometry on d30 p.i. (b) Schematic of the experimental setup. (c) Quantification showing the numbers of endogenous CX3CR1+ and CX3CR1 TEX cells (Ctrl: n = 10; ICB: n = 14; ICB/FTY720: n = 18) in 100 µL of peripheral blood on d30 p.i. (d) Representative flow cytometry plots and (e) quantification showing the frequencies of endogenous CX3CR1+ (left – Ctrl: n = 24; ICB: n = 14; ICB/FTY720: n = 24) and proliferating CX3CR1+ (right – Ctrl: n = 19; ICB: n = 9; ICB/FTY720: n = 18) cells in the spleen and LN on d30 p.i. (f, g) Naïve congenically marked (CD45.1+) P14 CD8+ T cells were adoptively transferred to naïve wildtype (CD45.2+) mice, which were subsequently infected with LCMV-Docile. A single dose of anti-PD-L1 was given on d26 p.i. with or without subsequent daily FTY720 treatment. CD8+ T cells were analysed using flow cytometry on d30 p.i. (f) Representative flow cytometry plots and (g) quantification showing the numbers of P14 and endogenous TPEX cells in peripheral blood (Ctrl: n = 10; ICB: n = 14; ICB/FTY720: n = 16). Dots in graphs (a, c, e, g) represent individual mice; bars represent median. Quantification and statistics derive from multiple t tests (b) and one-way ANOVA (c, e, g) and based on all data points across at least two (c, g) or three (a, e) independent experiments.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2 and Tables 1–7.

Reporting Summary

Peer Review File

Supplementary Table 1

T cell markers.

Supplementary Table 2

Trajectory 1 versus trajectory 2 markers.

Supplementary Table 3

KLF2 (KO versus WT) T cell markers.

Supplementary Table 4

KLF2 TPEX (KO versus WT) DE genes.

Supplementary Table 5

Overlapping genes: ChIP versus TPEX DE genes.

Supplementary Table 6

DC/myeloid markers.

Supplementary Table 7

Intercellular communication.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsui, C., Heyden, L., Wen, L. et al. Lymph nodes fuel KLF2-dependent effector CD8+ T cell differentiation during chronic infection and checkpoint blockade. Nat Immunol 26, 1752–1765 (2025). https://doi.org/10.1038/s41590-025-02276-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41590-025-02276-7

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer