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:

Dynamic CD4+ T cell heterogeneity defines subset-specific suppression and PD-L1-blockade-driven functional restoration in chronic infection

Abstract

Inhibiting PD-1:PD-L1 signaling has transformed therapeutic immune restoration. CD4+ T cells sustain immunity in chronic infections and cancer, yet little is known about how PD-1 signaling modulates CD4+ helper T (TH) cell responses or the ability to restore CD4+ TH-mediated immunity by checkpoint blockade. We demonstrate that PD-1:PD-L1 specifically suppressed CD4+ TH1 cell amplification, prevents CD4+ TH1 cytokine production and abolishes CD4+ cytotoxic killing capacity during chronic infection in mice. Inhibiting PD-L1 rapidly restored these functions, while simultaneously amplifying and activating TH1-like T regulatory cells, demonstrating a system-wide CD4–TH1 recalibration. This effect coincided with decreased T cell antigen receptor signaling, and re-directed type I interferon (IFN) signaling networks towards dominant IFN-γ-mediated responses. Mechanistically, PD-L1 blockade specifically targeted defined populations with pre-established, but actively suppressed proliferative potential, with limited impact on minimally cycling TCF-1+ follicular helper T cells, despite high PD-1 expression. Thus, CD4+ T cells require unique differentiation and functional states to be targets of PD-L1-directed suppression and therapeutic restoration.

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: PD-L1 blockade specifically amplifies and functionally enhances CD4+ TH1 cells.
Fig. 2: Pretherapy cycling CD4+ SMARTA T cells are expanded upon PD-L1 blockade.
Fig. 3: PD-L1 blockade specifically expands and activates TH1-like Treg cells.
Fig. 4: PD-L1 blockade expands and induces tissue infiltration of Treg cells in nonlymphoid organs.
Fig. 5: PD-L1 blockade enhances TH1 gene programs and terminal differentiation of TH1 cells.
Fig. 6: PD-L1 blockade reorients intracellular interferon and TCR signaling.
Fig. 7: PD-L1 blockade restores virus-specific CD4+ CTL function.

Similar content being viewed by others

Data availability

The RNA-seq data generated in this paper have been deposited in the Gene Expression Omnibus (GEO) database under accession number GSE163345. Source data are provided with this paper.

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Fahey, L. M. et al. Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells. J. Exp. Med. 208, 987–999 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Petrovas, C. et al. CD4 T follicular helper cell dynamics during SIV infection. J. Clin. Invest. 122, 3281–3294 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lindqvist, M. et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J. Clin. Invest. 122, 3271–3280 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Feng, J. et al. Patients with chronic hepatitis C express a high percentage of CD4(+)CXCR5(+) T follicular helper cells. J. Gastroenterol. 47, 1048–1056 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Snell, L. M. et al. Overcoming CD4 Th1 cell fate restrictions to sustain antiviral CD8 T cells and control persistent virus infection. Cell Rep. 16, 3286–3296 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. He, R. et al. Follicular CXCR5-expressing CD8(+) T cells curtail chronic viral infection. Nature 537, 421–428 (2016).

    Article  Google Scholar 

  9. 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 

  10. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Siddiqui, I. et al. Intratumoral Tcf1(+)PD-1(+)CD8(+) T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211.e10 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1(-)CD8(+) tumor-infiltrating T cells. Immunity 50, 181–194.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Miller, B. C. et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Roemer, M. G. M. et al. Major histocompatibility complex class II and programmed death ligand 1 expression predict outcome after programmed death 1 blockade in classic Hodgkin lymphoma. J. Clin. Oncol. 36, 942–950 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cader, F. Z. et al. A peripheral immune signature of responsiveness to PD-1 blockade in patients with classical Hodgkin lymphoma. Nat. Med. 26, 1468–1479 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Wei, S. C. et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 170, 1120–1133.e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Huang, A. C. et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545, 60–65 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Brooks, D. G., McGavern, D. B. & Oldstone, M. B. Reprogramming of antiviral T cells prevents inactivation and restores T cell activity during persistent viral infection. J. Clin. Invest. 116, 1675–1685 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Elsaesser, H., Sauer, K. & Brooks, D. G. IL-21 is required to control chronic viral infection. Science 324, 1569–1572 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Brooks, D. G., Teyton, L., Oldstone, M. B. & McGavern, D. B. Intrinsic functional dysregulation of CD4 T cells occurs rapidly following persistent viral infection. J. Virol. 79, 10514–10527 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xu, L. et al. The transcription factor TCF-1 initiates the differentiation of T(FH) cells during acute viral infection. Nat. Immunol. 16, 991–999 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Choi, Y. S. et al. LEF-1 and TCF-1 orchestrate T(FH) differentiation by regulating differentiation circuits upstream of the transcriptional repressor Bcl6. Nat. Immunol. 16, 980–990 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Parish, I. A. et al. Chronic viral infection promotes sustained Th1-derived immunoregulatory IL-10 via BLIMP-1. J. Clin. Invest. 124, 3455–3468 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. Macleod, B. L. et al. A network of immune and microbial modifications underlies viral persistence in the gastrointestinal tract. J. Exp. Med. 217, e20191473 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. Frohlich, A. et al. IL-21R on T cells is critical for sustained functionality and control of chronic viral infection. Science 324, 1576–1580 (2009).

    Article  PubMed  Google Scholar 

  28. Yi, J. S., Du, M. & Zajac, A. J. A vital role for interleukin-21 in the control of a chronic viral infection. Science 324, 1572–1576 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  32. Paley, M. A. et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gabrysova, L. et al. c-Maf controls immune responses by regulating disease-specific gene networks and repressing IL-2 in CD4(+) T cells. Nat. Immunol. 19, 497–507 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wilson, E. B. et al. Blockade of chronic type I interferon signaling to control persistent LCMV infection. Science 340, 202–207 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Teijaro, J. R. et al. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340, 207–211 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Au-Yeung, B. B. et al. A sharp T-cell antigen receptor signaling threshold for T-cell proliferation. Proc. Natl Acad. Sci. USA 111, E3679–E3688 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fazilleau, N., McHeyzer-Williams, L. J., Rosen, H. & McHeyzer-Williams, M. G. The function of follicular helper T cells is regulated by the strength of T cell antigen receptor binding. Nat. Immunol. 10, 375–384 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ray, J. P. et al. Transcription factor STAT3 and type I interferons are corepressive insulators for differentiation of follicular helper and T helper 1 cells. Immunity 40, 367–377 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Levine, A. G. et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature 546, 421–425 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Penaloza-MacMaster, P. et al. Interplay between regulatory T cells and PD-1 in modulating T cell exhaustion and viral control during chronic LCMV infection. J. Exp. Med. 211, 1905–1918 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hollern, D. P. et al. B cells and T follicular helper cells mediate response to checkpoint inhibitors in high mutation burden mouse models of breast cancer. Cell 179, 1191–1206.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wang, C. J. et al. CTLA-4 controls follicular helper T-cell differentiation by regulating the strength of CD28 engagement. Proc. Natl Acad. Sci. USA 112, 524–529 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Wing, J. B., Ise, W., Kurosaki, T. & Sakaguchi, S. Regulatory T cells control antigen-specific expansion of Tfh cell number and humoral immune responses via the coreceptor CTLA-4. Immunity 41, 1013–1025 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Sage, P. T., Paterson, A. M., Lovitch, S. B. & Sharpe, A. H. The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity 41, 1026–1039 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Barber, D. L. et al. Tuberculosis following PD-1 blockade for cancer immunotherapy. Sci. Transl. Med. 11, eaat2702 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Yokosuka, T. et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 209, 1201–1217 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sledzinska, A. et al. Regulatory T cells restrain interleukin-2- and Blimp-1-dependent acquisition of cytotoxic function by CD4(+) T cells. Immunity 52, 151–166 e156 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Oh, D. Y. et al. Intratumoral CD4(+) T cells mediate anti-tumor cytotoxicity in human bladder cancer. Cell 181, 1612–1625.e13 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Snell, L. M. et al. CD8+ T cell priming in established chronic viral infection preferentially directs differentiation of memory-like cells for sustained immunity. Immunity 49, 678–694.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  52. Levine, J. H. et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162, 184–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Weber, L. M., Nowicka, M., Soneson, C. & Robinson, M. D. diffcyt: differential discovery in high-dimensional cytometry via high-resolution clustering. Commun. Biol. 2, 183 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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 

  56. Mootha, V. K. et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G. D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS ONE 5, e13984 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Godec, J. et al. Compendium of immune signatures identifies conserved and species-specific biology in response to inflammation. Immunity 44, 194–206 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kinsella, R. J. et al. Ensembl BioMarts: a hub for data retrieval across taxonomic space. Database (Oxford) 2011, bar030 (2011).

    Article  Google Scholar 

  60. Yates, A. et al. Ensembl 2016. Nucleic Acids Res. 44, D710–D716 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. 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 

  62. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Anders, S., Pyl, P. T. & Huber, W. HTSeq – a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Griffith, M., Walker, J. R., Spies, N. C., Ainscough, B. J. & Griffith, O. L. Informatics for RNA sequencing: a web resource for analysis on the cloud. PLoS Comput. Biol. 11, e1004393 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer, 2016).

Download references

Acknowledgements

We thank past and present members of the Brooks laboratory for technical help and discussion. This work was supported by the Canadian Institutes of Health Research (CIHR) Foundation Grant FDN148386 (D.G.B.), the National Institutes of Health (NIH) grant AI085043 (D.G.B.), the Scotiabank Research Chair to D.G.B, and a training grant from the Fonds de la Recherche en Santé du Québec (L.M.S.).

Author information

Authors and Affiliations

Authors

Contributions

L.M.S. and D.G.B. designed the research. L.M.S., W.X., M.G., B.L.M., H.J.E., S.L., K.H. and N.A. performed the experiments. L.M.S., D.A.-R., G.B., S.N., and R.P. analyzed the data. S.E. and T.L.M. contributed technical expertise and discussion. L.M.S. and D.G.B. wrote the paper.

Corresponding authors

Correspondence to Laura M. Snell or David G. Brooks.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Robert Thimme, Santosha Vardhana and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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.

Extended data

Extended Data Fig. 1 CyTOF gating scheme.

Plots show the successive gating scheme for (a) SMARTA CD4 + T cells and (b) Treg cells.

Extended Data Fig. 2 Enhancement of virus-specific CD4+ TH1 cells by PD-L1 blockade occurs prior to decreased virus titers.

Virus-specific CD4+ SMARTA T cells were transferred into naïve mice which were subsequently infected with LCMV-Cl13 a day later. Twenty-five days after infection mice were treated with either isotype or anti-PD-L1 antibody and subsequently every 3 days for a total of 3 treatments. Mice were sacrificed 60 h following the first treatment or two days after the third treatment and splenic virus-specific CD4+ SMARTA T cells were analyzed. (a) Longitudinal plasma virus titers pre-treatment and then following one (n = 5) or three (n = 5) isotype or anti-PD-L1 treatments. (b) Log fold change of abundance of CD4+ SMARTA T cell PhenoGraph clusters at 60 h after the first antibody treatment. P-values of cluster abundances are calculated by the edge R test in the diffcyt R package. *, p < 0.05. (c) Total virus-specific CD4+ SMARTA T cell numbers at 60 h after the first isotype (n = 5) or anti-PD-L1 (n = 5) treatment. (d) Proportions and total number of endogenous (that is, non-SMARTA) TH1 cells (CD39 hi) vs TCF-1+ cells after the first isotype (n = 5) or anti-PD-L1 (n = 5) treatment. Foxp3+ Treg cells and SMARTA cells were gated out and plots are gated on the remaining CD4+ PD-1+ (activated) cells. (e) Proportion of virus-specific CD4+ T cells producing IFN-γ, TNF, and IL-10 following ex vivo LCMV-GP61–80 peptide stimulation. Data are from 60 h after the first isotype (n = 5) or anti-PD-L1 (n = 5) treatment. (f) Kinetics of CD4+ SMARTA T cells in each PhenoGraph cluster between 1 and 3 isotype or anti-PD-L1 treatments. (g) Total number of CD4+ SMARTA TH1 (TCF-1-negative) and TCF-1+ virus-specific T cells in liver and lung following 3 anti-PD-L1 treatments. (ac) n = 5 mice per group examined over four experiments totalling n = 19 isotype and n = 20 anti-PDL1 mice. (df) n = 5 mice per group examined over 2 experiments totaling 10 mice per group. Data in (G) is cumulative of 2 experiments with n = 11 for isotype mice and n = 12 for anti-PDL1 treatment groups. *: p < 0.05. unpaired, two-tailed Student’s t-test (a, cg). Box plots indicate the median, upper and lower quartile, and the whiskers show the high and low value. Line graphs show the average and error bars indicate the standard deviation (SD).

Source data

Extended Data Fig. 3 Pre-therapy cycling CD4+ SMARTA T cell populations are targets of anti-PD-L1 blockade.

(a) Diffusion pseudotime map of CD4+ SMARTA T cells colored by pseudotime starting at Tip (T)3. T1 – T3 designate the algorithm-derived cellular tips and the lines and arrows the pathways derived from T3 as a starting point. (b) Representative flow plots of Ki67+ SMARTA cells following 1 treatment in spleen. (c) UMAPs depict Ki67 expression in CD4+ SMARTA T cells and the graph the proportion of Ki67+ of SMARTA cells in each cluster following the third isotype or anti-PD-L1 treatment. (d) CD4+ SMARTA T cells in the spleen 10 days after LCMV-Cl13 infection mice. (Top) UMAP and heatmap of CyTOF data of CD4 + SMARTA T cells clustered with PhenoGraph. (Bottom) UMAPs illustrating the single cell expression of the indicated protein. The graph shows proportion of SMARTA cells that are Ki67+ in each PhenoGraph cluster. (a) n = 5 mice per group and are representative of total n = 9 mice examined over two experiments. (b, c) n = 5 mice per group and are representative of total n = 10 mice per group over 2 experiments. (d) n = 4 mice per group and representative of n = 9 mice total over 2 experiments. *: p < 0.05. unpaired, two-tailed Student’s t-test between isoytpe and aPD-L1 of each cluster (c), or between clusters 1 (Tfh) and Th1 clusters 4 and 6 (d). Box plots indicate the median, upper and lower quartile, and the whiskers show the high and low value.

Source data

Extended Data Fig. 4 PD-L1 blockade targets cycling, TH1-phenotype Treg cells.

(a) Total number of Foxp3+ Treg cells in each cluster at 60 h after the first isotype or anti-PD-L1 treatment. (b) Representative 2D CyTOF plots of T-bet, Bcl-6 and TCF-1 expression in total Treg cells. (c) To deplete Treg cells, LCMV-Cl13 infected Foxp3DTR mice were treated with DT or as a control PBS on days 23, day 24, day 27 and day 30 after infection. Mice were sacrificed at day 33 and splenocytes isolated for flow cytometric analysis. Representative FACS plots depict SLAM vs GzmB expression in splenic Foxp3-negative effector CD4+ T cells. Box plots depict total numbers of TH1, TFH and GzmB+ Foxp3-negative CD4+ T cells. (d) Total number of Foxp3+ Treg cells in each cluster following 3 treatments. (e) The colors in the heatmap designate the arcsinh ratio of the MSI change for the indicated protein within each PhenoGraph cluster of Treg cells. The graph is comparing protein expression in Foxp3+ Treg cells following the third anti-PD-L1 versus isotype treatment, with (red) increased with anti-PD-L1 treated mice or (blue) increased in isotype-treated mice. P-values are calculated by the limma test in the diffcyt R package. *, p-value<0.05. (f) UMAPs and bar graph depict the expression and proportion of Ki67+ cells in each Treg PhenoGraph cluster following 3 anti-PD-L1 or isotype treatments. (g) Representative protein expression plots gated on Ki67+ Treg cells in spleens of mice following 3 anti-PD-L1 or isotype treatments. (a) n = 5 mice in the isotype group and n = 4 in the anti-PDL1 group. The experiment is representative of n = 18 isotype and n = 16 anti-PDL1 mice examined over three experiments. (c) n = 5 mice in the PBS group and n = 7 mice in the DT group. The experiment is representative of n = 8 mice in the isotype and n = 11 mice in the DT group examined over two experiments. (dg) n = 5 mice per group.The experiment is representative of n = 15 mice per group examined over three experiments. *: p < 0.05; (unpaired, two-tailed Student’s t-test (a, c, d, f). Box plots indicate the median, upper and lower quartile, and the whiskers show the high and low value.

Source data

Extended Data Fig. 5 Single cell transcriptomic analyses of virus-specific CD4+ T cells following PD-L1 blockade.

(a) Serum Viral Titers in mice prior and post 3 anti-PD-L1 treatments. Line graph indicates average +/-SD of four mice per group. Data are representative of 1 single cell experiment with a total of n = 4 mice per group. Unpaired t-test was used to compare isotype pre and post therapy, and between PDL1 pre and post therapy. (b, c) Data are from one scRNA-seq experiment comprised of pooled cells from four individual mice per group. SMARTA cells were FACS-sorted following the third antibody treatment and analyzed by scRNA-seq. (b) Bar graph depicts differential expression of genes in sorted SLAMF1+ TH1 versus CXCR5+ TFH SMARTA cells from bulk RNA-seq at day 7 after LCMV-Armstrong infection. (c) UMAPs show IFNγ and IL10 gene expression.

Source data

Extended Data Fig. 6 Pathway analysis of CD4+ SMARTA T cells following PD-L1 blockade.

(a) Histograms depict Log10 transformed RNA expression of the ISGs Irf7 and Oas1a, and TH1-related genes Ifngr1 and Ccl5 in total virus-specific SMARTA CD4+ T cells, in the TH1 c2, c3, and in the TFH c5 from the scRNA-seq data from Fig. 5. Numbers in each histogram represent the percent of cells expressing the RNA. Blue histograms are anti-PD-L1 treated and those outlined in black are isotype treated. (b) Box plots indicate the GMFI of Nur77 protein expression in SMARTA cells 60 h after the first isotype (n = 5) or anti-PD-L1 (n = 6) antibody treatment. Data are representative of two experiments totaling 11 isotype and 12 anti-PD-L1 treated mice. *: p < 0.05, unpaired, two-tailed Student’s t-test. (c) The graph shows gene set enrichment analysis (GSEA) of differentially expressed pathways upregulated in CD4+ SMARTA T cells in anti-PD-L1 treatment vs isotype treatment. Pathways are colored by normalized enrichment score (NES). Data are representative of one scRNA-seq experiment comprised of pooled cells from four individual mice per group (Isotype or anti-PD-L1).

Source data

Extended Data Fig. 7 Cytotoxic gene expression in all SMARTA clusters from single cell analysis.

Violin plots depict expression of genes associated with cytotoxic function in all clusters following isotype and anti-PD-L1 treatment. Data are representative of one scRNA-seq experiment comprised of pooled cells from four individual mice per group.

Extended Data Fig. 8 Flow gating scheme for sorting.

Plots show the successive gating scheme for the sorting of SMARTA CD4 + T cells.

Supplementary information

Source data

Source Data Fig. 1

Numerical source data.

Source Data Fig. 2

Numerical source data.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 6

Numerical source data.

Source Data Fig. 7

Numerical source data.

Source Data Extended Data Fig. 2

Numerical source data.

Source Data Extended Data Fig. 3

Numerical source data.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 5

Numerical source data.

Source Data Extended Data Fig. 6

Numerical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Snell, L.M., Xu, W., Abd-Rabbo, D. et al. Dynamic CD4+ T cell heterogeneity defines subset-specific suppression and PD-L1-blockade-driven functional restoration in chronic infection. Nat Immunol 22, 1524–1537 (2021). https://doi.org/10.1038/s41590-021-01060-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01060-7

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