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:

An early precursor CD8+ T cell that adapts to acute or chronic viral infection

Abstract

This study examines the origin and differentiation of stem-like CD8+ T cells that are essential for sustained T cell immunity in chronic viral infections and cancer and also have a key role in PD-1 directed immunotherapy1,2,3,4,5,6,7,8,9,10. These PD-1+TCF-1+TOX+ stem-like CD8+ T cells (also known as precursors of exhausted T cells8,9) have a distinct program that enables them to adapt to chronic antigen stimulation. Here, using the mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection, we find that virus-specific stem-like CD8+ T cells are generated early (day 5) during chronic infection, suggesting that this crucial fate commitment occurs irrespective of the infection outcome. Indeed, we find that nearly identical populations of stem-like CD8+ T cells were generated early during acute or chronic LCMV infection, and that antigen was essential for maintaining the stem-like phenotype. We performed reciprocal adoptive transfer experiments to determine the fate of these early stem-like CD8+ T cells after viral clearance versus persistence. After transfer of day 5 stem-like CD8+ T cells from chronically infected mice into acutely infected mice, these cells downregulated canonical markers of the chronic stem-like CD8+ T cells and expressed markers (CD127 and CD62L) associated with central memory CD8+ T cells. Reciprocally, when day 5 stem-like cells from acutely infected mice were transferred into chronically infected mice, these CD8+ T cells functioned like chronic resource cells and responded effectively to PD-1 therapy. These findings highlight the ability of these early PD-1+TCF-1+TOX+ stem-like CD8+ T cells to adapt their differentiation trajectory to either an acute or a chronic viral infection. Importantly, our study shows that the host is prepared a priori to deal with a potential chronic infection.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Early fate decision of stem-like CD8+ T cells during chronic infection.
Fig. 2: Differences between early and late stem-like CD8+ T cells as chronic infection progresses.
Fig. 3: Identical populations of stem-like CD8+ T cells are generated early after acute and chronic infection.
Fig. 4: Antigen is required for maintenance of the stem-like CD8+ T cell phenotype.
Fig. 5: Early stem-like CD8+ T cells from chronic viral infection express memory CD8+ T cell markers after transfer into acutely infected mice.
Fig. 6: Early stem-like CD8+ T cells from acute infection function as chronic resource cells after transfer into chronically infected mice and respond to PD-1 blockade.

Similar content being viewed by others

Data availability

Raw RNA-seq data have been uploaded to the NCBI Gene Expression Omnibus under identifier GSE284012. Raw scRNA-seq data have been uploaded to the NCBI Gene Expression Omnibus under identifier GSE284013. Raw data for ATAC–seq analysis have been uploaded to the NCBI Gene Expression Omnibus under identifier GSE269785. Previously published datasets GSE84105 (ref. 1), GSEA datasets HALLMARK_MITOTIC_SPINDLE and HALLMARK_G2M_CHECKPOINT41 were also used for analysis in this study. Source data are provided with this paper.

References

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Wu, T. et al. The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci. Immunol. 1, eaai8593 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Leong, Y. A. et al. CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat. Immunol. 17, 1187–1196 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. He, R. et al. Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412–416 (2016).

    Article  ADS  CAS  PubMed  Google Scholar 

  6. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. McLane, L. M., Abdel-Hakeem, M. S. & Wherry, E. J. CD8 T cell exhaustion during chronic viral infection and cancer. Ann. Rev. Immunol. 37, 457–495 (2019).

    Article  CAS  Google Scholar 

  9. Hashimoto, M. et al. CD8 T cell exhaustion in chronic infection and cancer: opportunities for interventions. Ann. Rev. Med. 69, 301–318 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Gill, A. L. et al. PD-1 blockade increases the self-renewal of stem-like CD8 T cells to compensate for their accelerated differentiation into effectors. Sci. Immunol. 8, eadg0539 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Im, S. J., Konieczny, B. T., Hudson, W. H., Masopust, D. & Ahmed, R. PD-1+ stemlike CD8 T cells are resident in lymphoid tissues during persistent LCMV infection. Proc. Natl Acad. Sci. USA 117, 4292–4299 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Utzschneider, D. T. et al. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat. Immunol. 21, 1256–1266 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Yao, C. et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 20, 890–901 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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).

    Article  CAS  PubMed  Google Scholar 

  15. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Crotty, S. Follicular Helper CD4 T Cells (TFH). Ann. Rev. Immunol. 29, 621–663 (2011).

    Article  CAS  Google Scholar 

  19. Hale, J. S. et al. Distinct memory CD4+ T cells with commitment to T follicular helper- and T helper 1-cell lineages are generated after acute viral infection. Immunity 38, 805–817 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jadhav, R. R. et al. Epigenetic signature of PD-1+ TCF1+ CD8 T cells that act as resource cells during chronic viral infection and respond to PD-1 blockade. Proc. Natl Acad. Sci. USA 116, 14113–14118 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Abdel-Hakeem, M. S. et al. Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat. Immunol. 22, 1008–1019 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hu, Y. et al. TGF-β regulates the stem-like state of PD-1+ TCF-1+ virus-specific CD8 T cells during chronic infection. J. Exp. Med. 219, e20211574 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Au-Yeung, B. B. et al. IL-2 modulates the TCR signaling threshold for CD8 but not CD4 T cell proliferation on a single-cell level. The J. Immunol. 198, 2445–2456 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Kaech, S. M. et al. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat. Immunol. 4, 1191–1198 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Joshi, N. S. et al. Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sarkar, S. et al. Functional and genomic profiling of effector CD8 T cell subsets with distinct memory fates. J. Exp. Med. 205, 625–640 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Youngblood, B. et al. Effector CD8 T cells dedifferentiate into long-lived memory cells. Nature 552, 404–409 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Akondy, R. S. et al. Origin and differentiation of human memory CD8 T cells after vaccination. Nature 552, 362–367 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Abadie, K. et al. Reversible, tunable epigenetic silencing of TCF1 generates flexibility in the T cell memory decision. Immunity 57, 271–286 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Eberhardt, C. S. et al. Functional HPV-specific PD-1+ stem-like CD8 T cells in head and neck cancer. Nature 597, 279–284 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Prokhnevska, N. et al. CD8+ T cell activation in cancer comprises an initial activation phase in lymph nodes followed by effector differentiation within the tumor. Immunity 56, 107–124 (2023).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Ahmed, R. et al. Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice. Role in suppression of cytotoxic T lymphocyte response and viral persistence. J. Exp. Med. 160, 521–540 (1984).

  35. Wherry, E. J., Blattman, J. N., Murali-krishna, K., Van Der Most, R. & Ahmed, R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional Impairment. J. Virol. 77, 4911–3927 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. West, E. E. et al. PD-L1 blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells. J. Clin. Invest. 123, 2604–2615 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Galkina, E. et al. Preferential migration of effector CD8+ T cells into the interstitium of the normal lung. J. Clin. Invest. 115, 3473–3483 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. DeTomaso, D. & Yosef, N. FastProject: a tool for low-dimensional analysis of single-cell RNA-seq data. BMC Bioinform. 17, 315 (2016).

    Article  Google Scholar 

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

    Article  Google Scholar 

  42. Zerbino, D. R. et al. Ensembl 2018. Nucleic Acids Res. 46, D754–D761 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  46. Hadley, W. ggplot2: elegant graphics for data analysis (Springer, 2016).

  47. Guo, M. et al. EZH2 represses the B cell transcriptional program and regulates antibody-secreting cell metabolism and antibody production. J. Immunol. 200, 1039–1052 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institute of Health grants R01AI030048 (R.A.) and R01AI165706 (B.B.A.-Y.); HHMI Hanna Gray Fellowship GT16001 (C.B.M.); Burroughs Wellcome Fund Postdoctoral Enrichment Program 1022361 (C.B.M.); Achievement Rewards for College Scientists Standard Scholarship (A.L.G.); NIH/NIBIB Trailblazer Award R21EB027327 (H.Y.K.); NSF Graduate Research Fellowships (K.A. and E.C.C.). We thank staff at the Emory University School of Medicine’s Flow Cytometry Core (K. Fife and R. Karaffa), the Emory Integrated Cellular Imaging Core (L. Fox-Goharioon), the Emory Integrated Genomics Core (C. Scharer) and the Yerkes Nonhuman Primate Genomics Core (K. Pellegrini and S. Bosinger; NIH P51OD011132). We acknowledge the NIH/NIAID BioArt Repository for use of their publicly available graphics in our paper (https://bioart.niaid.nih.gov/bioart/279; https://bioart.niaid.nih.gov/discover?q=T%20cell).

Author information

Authors and Affiliations

Authors

Contributions

D.T.M., R.M.V., Y.H., M.H., D.Y.C. and R.A. designed experiments. D.T.M., R.M.V., Y.H., A.W., T.H.N., J.L., J.L.R., D.Y.C., N.P. and M.A.C. performed experiments. D.T.M., R.M.V., Y.H. and R.A. analysed experiments. E.S., D.J.M. and H.T.K. analysed scRNA-seq data. D.T.M., C.B.M., C.D.S. and A.L.G. analysed RNA-seq data. C.D.S. analysed ATAC–seq data. B.B.A.-Y., E.C.C., K.A., A.K., H.Y.K. and J.K. contributed materials. D.T.M., R.M.V. and R.A. wrote the manuscript. All of the authors contributed to writing the manuscript and providing feedback.

Corresponding author

Correspondence to Rafi Ahmed.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Stephen Jameson and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Early anatomical commitment of stem-like CD8 T cells during chronic infection.

a, On days 5 and >45 after chronic infection, mice were injected with fluorescently-labelled anti-CD8α (3 μg/mouse) and euthanized 3 min later for flow cytometry analysis. b, Representative flow plots/histograms showing frequencies of GP33+TCF-1+Tim-3 (stem) and TCF-1Tim-3+ (effect./TD) CD8+ T cells in the white pulp (CD8α i.v. antibody negative) or the red pulp (CD8α i.v. antibody positive). Numbers on flow plots are frequencies. c, Frequencies of stem and effect./TD CD8+ T cells in the white pulp (left) and red pulp. Mean ± SEM is shown. Results pooled from 2 of 3 independent experiments with n = 10 mice (day 5) and n = 10 mice (day >45). P values shown were calculated using Wilcoxon matched-pairs signed rank test. d, Representative immunofluorescent histology showing the location of stem-like (green) and effector (orange) CD8+ T cells in the spleen on days 5 and >45 after chronic infection. Dashed lines denote border between the white pulp (WP) and red pulp (RP). Stem-like cells were identified as CD8β+PD-1+TCF-1+DAPI+ cells. Effector cells were identified CD8β+PD-1+TCF-1DAPI+ cells. e, Frequency of indicated subsets in white pulp (left) and red pulp (right) based on image analysis. (n = 3-4 mice per experiment). Summary graphs show mean ± SEM and are pooled from 2 independent experiments with n = 6 mice (day 5), and n = 7 mice (day >45). P values shown were calculated using Wilcoxon matched-pairs signed rank test.

Source data

Extended Data Fig. 2 scRNA-seq of LCMV GP33-specific CD8+ T cells on days 5 and >45 after chronic infection.

a, Gating strategy for sorting of splenic DbGP33+CD8+ T cells on days 5 (60,000 cells) and >45 (40,000 cells) for scRNA-seq. b,c, Gene set enrichment analysis enumerating enrichment of day 5 and >45 cell clusters for stem-like and effect/TD gene signatures: GSE84105, Im et al.1. Max = maximum. d,e,UMAP showing day 5 cell clusters (d) and expression of selected genes by each cluster (e). f,g, UMAP showing day >45 cell clusters (f) and expression of selected genes by each cluster (g).

Source data

Extended Data Fig. 3 RNA-seq and ATAC-seq analyses of sorted CD8 T cell subsets on days 5 and >45 after chronic infection.

a, Gating strategy for sorting of day 5 and >45 PD-1+ stem-like (CD73+ Tim-3−) and effector/terminally differentiated (CD73Tim-3+) CD8+ T cells for RNA-seq. b, 2,500 P14 cells were transferred into congenically distinct naïve mice prior to intravenous infection with 2 × 106 LCMV clone 13. Gating strategy for sorting of day 5 and >45 LCMV GP33-specific P14 stem-like (CD73+ Tim-3−) and effector/terminally differentiated (CD73Tim-3+) CD8+ T cells for ATAC-seq. Naïve CD8+ T cells (CD44PD-1) from uninfected mice were used as a control in each analysis. c,d, Genome plots showing chromatin accessibility (ATAC-seq, left) and normalized counts (RNA-seq, right) for Slamf6 (c) and Xcl1 (d) in naïve CD8 T cells from uninfected mice and day 5 vs. >day 45 stem-like and effector CD8 T cells from chronic infection. The location of the transcription start site is indicated by an arrow and a bar depicts the location of the gene body. Bar graphs for normalized counts are based on technical replicates with n = 3 per group. e, Density scatter plot comparing the log2 fold-change in ATAC-seq (x-axis) to the corresponding RNA-seq (y-axis) data for the indicated comparison. Each dot represents accessibility of a single peak and the matching gene expression of the nearest gene. Blue lines indicate data density. Linear correlation trend lines are plotted, and Pearson correlation coefficient and significance of trend are indicated. Coloured dots correspond to the data for Pdcd1, Xcl1, Tcf7, and Gzmb as indicated. f, Heatmap of the Pearson correlation coefficient between ATAC-seq and RNA-seq data for each pairwise comparison between the sample groups. RNA- and ATAC-seq results are each from 1 independent experiment.

Source data

Extended Data Fig. 4 Changes in stem-like CD8 T cells as chronic infection progresses from days 5 to >45.

a, Enrichment of day 5 and >45 GP33+CD8+ T cell clusters for cell cycle gene signature based on gene set enrichment analysis based on scRNA seq analysis. Cell cycle signature combines genes from two GSEA datasets: HALLMARK_MITOTIC_SPINDLE and HALLMARK_G2M_CHECKPOINT (ref. 41). b, Expression of Mki67 by day 5 and day >45 GP33+ cells based on scRNA-seq analysis. c, FACs analysis of splenic GP33+TCF-1+Tim-3 stem-like CD8+ T cells from days 5 and >45. Histogram (left) and summary graph (right) showing cell size based on forward scatter (FSC-A). Mean ± SEM is shown. Results pooled from 3 independent experiments with n = 10 mice (D5 stem) and n = 10 mice (>D45 stem). P values shown were calculated using two-tailed, unpaired Mann-Whitney U test. d, Enrichment for indicated gene sets by PD-1+ stem-like CD8+ T cells from day 5 versus >45 (RNA-seq). e, Bar graph showing relative expression of top 50 most significant differentially expressed genes (DEGs) from mTORC1 signalling pathway between PD-1+ day 5 and >45 stem-like CD8+ T cells (RNA-seq). f, Expression of indicated genes by naïve (CD44PD-1), and PD-1+ day 5 and >45 stem-like CD8+ T cells based on normalized RNA counts. Circles correspond to technical replicates with n = 3 per group. RNA- and ATAC-seq results are each from 1 independent experiment.

Source data

Extended Data Fig. 5 Identical populations of virus-specific stem-like CD8+ T cells are generated early after acute and chronic LCMV infection.

a,b, Phenotypic analysis of GP33+CD8+ T cells in the spleen on day 5 after LCMV Armstrong (acute, 2 × 106 PFU i.v.) or clone 13 (chronic, 2 × 106 PFU i.v.) infection. Flow plots are representative of 3 independent experiments (n = 4-5 recipient mice per donor per experiment). c,d, Frequency (c) and number (d) of GP33+PD-1+TCF-1+CD8+ T cells in the spleen on day 5 after acute versus chronic infection. For c and d, mean ± SEM are shown. Results are pooled from 3 independent experiments with n = 15 mice (acute) and n = 15 mice (chronic). P values shown were calculated using two-tailed, unpaired Mann-Whitney U test. e, Gating strategy used for sorting LCMV-specific GP33+ P14 stem-like (PD-1+CD73+Tim-3) and effector (PD-1+CD73Tim-3+) CD8+ T cells from Armstrong or clone 13-infected spleens on day 5 for RNA- and ATAC-seq. f, Heatmaps showing relative expression (left) and accessibility (right) of selected genes by indicated subsets. g, Accessibility tracks (ATAC-seq, left) and normalized counts (RNA-seq, right) for selected genes. The location of the transcription start site is indicated by an arrow and a bar depicts the location of the gene body. Bar graphs for normalized counts are based on technical replicates with n = 3 per group. RNA- and ATAC-seq results are each from 1 independent experiment. h, On day 5 after acute or chronic infection, mice were injected with fluorescently-labelled anti-CD8α (3 μg/mouse) and euthanized 3 min later for flow cytometry analysis. i, Flow plots showing frequencies of GP33+TCF-1+Tim-3 (stem-like) and TCF-1Tim-3+ (effect./TD) CD8+ T cells in the white pulp (CD8α i.v. negative) or the red pulp (CD8α i.v. positive). Numbers on flow plots are frequencies. Flow plots are representative of 1 of 2 independent experiments (n = 3-5 mice per infection group). j, Frequencies of stem-like and effect./TD CD8+ T cells in the white pulp (left) and red pulp (right). Graph shows 1 of 2 independent experiments with n = 3 mice (Arm) and n = 3 mice (Cl13).

Source data

Extended Data Fig. 6 Stem-like CD8+ T cells from acute and chronic infection are equally able to expand, self-renew, and differentiate into effector cells after clone 13 challenge.

a, LCMV-specific GP33+ P14 stem and effect. CD8+ T cells (CD45.1+CD45.2) were isolated from the spleens of LCMV Armstrong or clone 13-infected mice on day 5 and transferred into naïve mice (CD45.1CD45.2+). Recipients were then challenged i.v. with 2 × 106 PFU of LCMV Cl13. b, Gating strategy used to isolate stem-like (Slamf6+ Tim-3−) and effector (Slamf6Tim-3+) P14 CD8 T+ cells from acute versus chronic LCMV infection on day 5 to evaluate their response to clone 13 challenge. c, FACs plots showing frequency of donor cells recovered from indicated recipient livers on day 7 after clone 13 challenge. Data are representative of 1 of 3 independent experiments. d, Numbers of donor cells recovered from indicated recipient livers on day 7 after clone 13 challenge. Mean ± SEM is shown. Results pooled from 3 independent experiments with n = 9 mice (Arm/Cl13 stem donor) and n = 9 mice (Arm/Cl13 effect. donor). P values shown were calculated using two-tailed, unpaired Mann-Whitney U test. e, FACs analysis of stem-like donor cells from acute (top) or chronic (bottom) infection pre- and on day 7 post-clone 13 challenge. Data are representative of 1 of 3 independent experiments. f, FACs analysis of effector donor cells from acute (top) or chronic (bottom) infection pre- and on day 7 post-clone 13 challenge. Data are representative of 1 of 3 independent experiments. g, Numbers of splenic stem-like (TCF-1+ GzmB−) and effector (TCF-1GzmB+) CD8+ T cells derived from indicated effector donor cells. Results pooled from 3 independent experiments with n = 9 mice (Arm effector donor) and n = 9 mice (Cl13 effect. donor). P values shown were calculated using two-tailed, unpaired Mann-Whitney U test.

Source data

Extended Data Fig. 7 Antigen is required for the maintenance of the stem-like CD8+ T cell phenotype during acute viral infection.

Mice were infected intravenously with 2 × 106 PFU of LCMV Armstrong (acute). a, Viral titre kinetics (blue curve) and numbers of LCMV-specific GP33+CD8+ T cells in the spleen over time. Viral titre data shown are from one independent experiment with n = 5 mice per time point and are consistent with historical data from our lab. Black line shows number of GP33+CD8+ T cells at indicated time points. Data pooled from 3 independent experiments with n = 15 mice (day 5), n = 12 mice (day 8), and n = 12 mice (day 14). Mean ± SEM are shown. b, Frequency of GP33+PD-1+TOX+TCF-1+ stem-like CD8+ T cells in the spleen at indicated times post- acute infection. Results pooled from 3 independent experiments with n = 12 mice (D5), n = 9 mice (D8), n = 9 mice (D14). Mean ± SEM is shown. P values shown were calculated using two-tailed, unpaired Mann-Whitney U test. c, Expression of indicated markers on GP33+TCF-1+CD8+ T cells at indicated times after acute LCMV infection. Numbers in flow plots indicate frequency. Flow plots are representative of 1 of 3 independent experiments (n = 3-5 mice per time point per experiment). d, On day 5 after acute infection, mice were injected i.v. with 200 μg of GP33 peptide on days 5, 7, and 10. After clearance of infection (day 14), the phenotype of GP33+CD8+ T cells was examined in acutely infected mice that were untreated or injected with peptide. e, Flow plots showing frequencies of GP33+CD44+CD8+ T cells in the spleen in the two groups of mice. Data are representative of 1 of 3 independent experiments (n = 3-5 mice per group). f, Frequency of GP33+CD8+ T cells in the spleen. Results pooled from 4 independent experiments with n = 13 mice (day 5), n = 18 mice (day 14, no pep.), n = 12 mice (day 14, GP33 pep.). Mean ± SEM is shown. P values shown were calculated using two-tailed, unpaired Mann-Whitney U test. g, Expression of indicated markers on GP33+TCF-1+CD8+ T cells. Data are representative of 1 of 4 independent experiments (n = 3-5 mice per group). h, Frequency of GP33+PD-1+TCF-1+CD8+ T cells in the spleen for indicated groups of mice. Results pooled from 4 independent experiments with n = 13 mice (day 5), n = 18 mice (day 14, no pep.), n = 12 mice (day 14, GP33 pep.). Mean ± SEM is shown. P values shown were calculated using two-tailed, unpaired Mann-Whitney U test.

Source data

Extended Data Fig. 8 Antigen is required for the maintenance of the stem-like CD8+ T cell phenotype during chronic viral infection.

a, 2,000 P14 CD8+ T cells were transferred into congenically distinct naïve mice. Recipients were then infected with either wild-type LCMV clone 13 alone (WT Cl13, black) or mixed 1:2 with a mutant GP33-deficient clone 13 strain (WT/Mut, red). b, FACs analysis of P14s or endogenous LCMV-specific DbGP276+CD8+ T cells. Flow plots are representative of 1 of 2 independent experiments (WT Cl13 n = 5-10 mice, WT/Mut Cl13 n = 4-5 mice). c, Nur77-GFP reporter mice were infected with either acute or chronic LCMV infection. d, Frequencies of Nur77-GFP+ cells among indicated GP33+ subsets. Results pooled from 2 independent experiments with n = 8 mice (chronic LCMV, >D45) and n = 4 mice (Acute LCMV, >D60) with mean ± SEM is shown.

Source data

Extended Data Fig. 9 Adoptive transfer of stem-like and effector CD8+ T cells from day 5 LCMV clone 13 (chronic) mice into day 5 LCMV Armstrong (acute) infected mice.

Same experimental setup as shown in Fig. 5a. a, Frequency of stem-like and effector donor cells recovered from the spleens of LCMV Armstrong-infected recipients on days 8/15 post-transfer. Data representative of 1 of 3 independent experiments. b, Expression of CD127, KLRG1, CD62L and TCF-1 on stem-like donor cells in the spleen pre- (day 5 p.i., chronic) and post-transfer (days 8/15 post-transfer, acute). Data representative of 1 of 3 independent experiments. c, Frequencies of KLRG1+CD127 (left) and KLRG1CD127+ (right) P14 CD8 T cells derived from stem donor cells post-transfer on days 8/15 post-transfer. Results pooled from 3 independent experiments with n = 10 mice (stem donor recipients) with mean ± SEM is shown. P values shown were calculated using two-tailed, unpaired Mann-Whitney U test. d, Expression of CD127, KLRG1, CD62L and TCF-1 on effector donor cells in the spleen pre- (day 5 p.i., chronic) and post-transfer (days 8/15 post-transfer, acute). e, Frequencies of KLRG1+CD127 (left) and KLRG1CD127+ (right) P14 CD8 T cells derived from effector donor cells post-transfer on days 8/15 post-transfer. Results pooled from 3 independent experiments with n = 16 mice (effect. donor recipients). Mean ± SEM is shown. P values shown were calculated using two-tailed, unpaired Mann-Whitney U test.

Source data

Extended Data Fig. 10 Early stem-like CD8+ T cells from acute viral infection become chronic resource CD8+ T cells after transfer into chronically infected mice.

a, 2 × 103TCF7-YFP reporter P14 CD8+ T cells (CD45.1CD45.2+) were transferred into congenically distinct naïve mice (CD45.1+CD45.2) prior to intravenous infection with 2 × 106 PFU of LCMV Armstrong (acute). On day 5, TCF7-YFP+Tim-3 (stem) andTCF7-YFPTim-3+ (effect.) P14 CD8+ T cells (CD45.1CD45.2+) were FACS sorted from the spleen and 1 × 106 cells of each sorted subset were transferred intravenously into separate groups of congenically distinct, day 5 LCMV clone 13 (chronic) infected mice (CD45.1+CD45.2). The recovery and phenotype of donor cells were analysed 8 days after transfer. b, Flow plots showing stem and effector subsets pre- and post-sort with purity indicated. c, FACs analysis showing frequencies of stem-like and effector P14 donor cells recovered from indicated tissues. Flow plots represent 1 independent experiment. d, Numbers of stem-like and effector P14 donor cells recovered from indicated tissues. Data are from 1 independent experiment with n = 3 mice (stem donor recipients) and n = 4 mice (effector donor recipients). e, Expression of indicated markers on stem-like donor P14 cells pre- (spleen, day 5 p.i., acute) and day 8 post-transfer (spleen, liver and lung, chronic). Arrow indicates differentiation trajectory of donor Tim-3 stem-like cells into Tim-3+ effector cells. f, Expression of indicated markers on effector donor P14 cells pre-transfer (spleen, day 5 p.i., acute, top panel) and day 8 post-transfer (spleen, chronic, bottom panel). Flow plots represent 1 independent experiment.

Source data

Extended Data Fig. 11 Longitudinal tracking of day 5 stem-like and effector CD8+ T cells from acute viral infection following transfer into chronically infected mice.

a, 2 × 103 TCF7-YFP reporter P14 CD8+ T cells (CD45.1CD45.2+) were transferred into congenically distinct naïve mice (CD45.1+CD45.2) before i.v. infection with 2 × 106 PFU of LCMV Armstrong (acute). On day 5 p.i. stem-like (TCF7-YFP+ Tim-3−) and effector (TCF7-YFPTim-3+ (effect.) P14 T cells were isolated from the spleen via FACS and transferred into separate groups of day 5 LCMV clone 13-infected recipients (CD45.1+CD45.2), which had been transiently depleted of CD4+ T cells before infection. b, Flow plots showing stem and effector subsets pre- and post-sort with purity indicated. c, Frequencies of stem-like and effector P14 donor cells recovered from the spleens, livers, and lungs, of chronic LCMV recipient mice at indicated times post-transfer. Data represent one independent experiment. d, Expression of TCF-1 and Tim-3 on stem-like P14 donor cells pre-transfer (spleen, day 5 p.i., acute) and at indicated times post-transfer in the livers and lungs of chronically infected recipients. Data represent one independent experiment. e, UMAP plot showing scRNA-seq analysis of stem donor P14 cells on day 15 post-transfer (right) and endogenous GP33+CD8+ T cells from stem donor P14 recipient mice (left). UMAPs represent 1 independent experiment.

Supplementary information

Reporting Summary

Supplementary Table 1

Detailed information for the antibodies used in flow cytometry and immunohistochemistry.

Supplementary Table 2

Specific Gene Ontology pathways and identifiers used for analysis in Fig. 2d,e.

Source data

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

McManus, D.T., Valanparambil, R.M., Medina, C.B. et al. An early precursor CD8+ T cell that adapts to acute or chronic viral infection. Nature 640, 772–781 (2025). https://doi.org/10.1038/s41586-024-08562-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41586-024-08562-y

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