Skip to main content

Advertisement

Log in

Distinct SIV-specific CD8+ T cells in the lymph node exhibit simultaneous effector and stem-like profiles and are associated with limited SIV persistence

  • Article
  • Published:

From Nature Immunology

View current issue Submit your manuscript

Abstract

Human immunodeficiency virus (HIV) cure efforts are increasingly focused on harnessing CD8+ T cell functions, which requires a deeper understanding of CD8+ T cells promoting HIV control. Here we identifiy an antigen-responsive TOXhiTCF1+CD39+CD8+ T cell population with high expression of inhibitory receptors and low expression of canonical cytolytic molecules. Transcriptional analysis of simian immunodeficiency virus (SIV)-specific CD8+ T cells and proteomic analysis of purified CD8+ T cell subsets identified TOXhiTCF1+CD39+CD8+ T cells as intermediate effectors that retained stem-like features with a lineage relationship with terminal effector T cells. TOXhiTCF1+CD39+CD8+ T cells were found at higher frequency than TCF1CD39+CD8+ T cells in follicular microenvironments and were preferentially located in proximity of SIV-RNA+ cells. Their frequency was associated with reduced plasma viremia and lower SIV reservoir size. Highly similar TOXhiTCF1+CD39+CD8+ T cells were detected in lymph nodes from antiretroviral therapy-naive and antiretroviral therapy-suppressed people living with HIV, suggesting this population of CD8+ T cells contributes to limiting SIV and HIV persistence.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: TOX is upregulated on CD8+ T cells after SIV infection.
Fig. 2: TCF1+CD39+CD8+ T cells expand after SIV infection and are a unique phenotypic population.
Fig. 3: LN SIV-specific TCF1+CD39+CD8+ T cells maintain a dual-effector and stem-like transcriptional profile.
Fig. 4: Frequency of TOX+CD8+ and TCF1+CD39+CD8+ TM cell is associated with reduced viral burden.
Fig. 5: TCF1+CD39+CD8+ TM cells preferentially enter the follicular environment.
Fig. 6: TCF1+CD39+CD8+ TM cells contract during long-term ART but remain associated with viral burden.
Fig. 7: HIV infection results in expansion of LN HIV-specific TCF1+CD39+CD8+ TM cells.

Similar content being viewed by others

Data availability

Single-cell RNA-seq data have been deposited in the NCBI GEO database under accession number GSE266576. Proteomics data have been deposited in the PRIDE database under accession number PXD050498 and can be accessed at https://www.ebi.ac.uk/pride/archive/. All other data are present in the article as individual data points, Supplementary Information or from the corresponding authors upon reasonable request.

Code availability

The custom code used in this manuscript is available from the corresponding author upon reasonable request.

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  5. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Seo, H. et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl Acad. Sci. USA 116, 12410–12415 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Sekine, T. et al. TOX is expressed by exhausted and polyfunctional human effector memory CD8+ T cells. Sci. Immunol. 5, eaba7918 (2020).

    CAS  PubMed  Google Scholar 

  9. Heim, K. et al. TOX defines the degree of CD8+ T cell dysfunction in distinct phases of chronic HBV infection. Gut 70, 1550–1560 (2020).

    PubMed  Google Scholar 

  10. 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 

  11. 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 

  12. Collins, D. R., Gaiha, G. D. & Walker, B. D. CD8+ T cells in HIV control, cure and prevention. Nat. Rev. Immunol. 20, 471–482 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Day, C. L. et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443, 350–354 (2006).

    CAS  PubMed  Google Scholar 

  14. Trautmann, L. et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat. Med. 12, 1198–1202 (2006).

    CAS  PubMed  Google Scholar 

  15. Petrovas, C. et al. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J. Exp. Med. 203, 2281–2292 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Hoffmann, M. et al. Exhaustion of activated CD8 T cells predicts disease progression in primary HIV-1 infection. PLoS Pathog. 12, e1005661 (2016).

    PubMed  PubMed Central  Google Scholar 

  17. Chew, G. M. et al. TIGIT marks exhausted T cells, correlates with disease progression, and serves as a target for immune restoration in HIV and SIV infection. PLoS Pathog. 12, e1005349 (2016).

    PubMed  PubMed Central  Google Scholar 

  18. Collins, D. R. et al. Functional impairment of HIV-specific CD8+ T cells precedes aborted spontaneous control of viremia. Immunity 54, 2372–2384 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Bengsch, B. et al. Epigenomic-guided mass cytometry profiling reveals disease-specific features of exhausted CD8 T cells. Immunity 48, 1029–1045 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Reuter, M. A. et al. HIV-specific CD8+ T cells exhibit reduced and differentially regulated cytolytic activity in lymphoid tissue. Cell Rep. 21, 3458–3470 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Nguyen, S. et al. Elite control of HIV is associated with distinct functional and transcriptional signatures in lymphoid tissue CD8+ T cells. Sci. Transl. Med. 11, eaax4077 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Buggert, M. et al. Identification and characterization of HIV-specific resident memory CD8+ T cells in human lymphoid tissue. Sci. Immunol. 3, eaar4526 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Collins, D. R. et al. Cytolytic CD8+ T cells infiltrate germinal centers to limit ongoing HIV replication in spontaneous controller lymph nodes. Sci. Immunol. 8, eade5872 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Ward, A. R., Mota, T. M. & Jones, R. B. Immunological approaches to HIV cure. Semin. Immunol. 51, 101412 (2021).

    CAS  PubMed  Google Scholar 

  25. Deeks, S. G. et al. Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2021. Nat. Med. 27, 2085–2098 (2021).

    CAS  PubMed  Google Scholar 

  26. Huang, S. et al. Increased TOX expression concurrent with PD-1, Tim-3, and CD244 in T cells from patients with non-Hodgkin lymphoma. Asia Pac. J. Clin. Oncol. 18, 143–149 (2022).

    PubMed  Google Scholar 

  27. Wang, X. et al. TOX promotes the exhaustion of antitumor CD8+ T cells by preventing PD1 degradation in hepatocellular carcinoma. J. Hepatol. 71, 731–741 (2019).

    CAS  PubMed  Google Scholar 

  28. Gupta, P. K. et al. CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog. 11, e1005177 (2015).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Canale, F. P. et al. CD39 expression defines cell exhaustion in tumor-infiltrating CD8+ T cells. Cancer Res. 78, 115–128 (2018).

    CAS  PubMed  Google Scholar 

  31. Abbas, H. A. et al. Single cell T cell landscape and T cell receptor repertoire profiling of AML in context of PD-1 blockade therapy. Nat. Commun. 12, 6071 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Jonsson, A. H. et al. Granzyme K+ CD8 T cells form a core population in inflamed human tissue. Sci. Transl. Med. 14, eabo0686 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Arbones, M. L. et al. Lymphocyte homing and leukocyte rolling and migration are impaired in l-selectin-deficient mice. Immunity 1, 247–260 (1994).

    CAS  PubMed  Google Scholar 

  34. Takeuchi, A. et al. CRTAM confers late-stage activation of CD8+ T cells to regulate retention within lymph node. J. Immunol. 183, 4220–4228 (2009).

    CAS  PubMed  Google Scholar 

  35. Guma, M. et al. The CD94/NKG2C killer lectin-like receptor constitutes an alternative activation pathway for a subset of CD8+ T cells. Eur. J. Immunol. 35, 2071–2080 (2005).

    CAS  PubMed  Google Scholar 

  36. Trkola, A. et al. Genetic subtype-independent inhibition of human immunodeficiency virus type 1 replication by CC and CXC chemokines. J. Virol. 72, 396–404 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Cocchi, F. et al. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science 270, 1811–1815 (1995).

    CAS  PubMed  Google Scholar 

  38. Ichii, H. et al. Role for Bcl-6 in the generation and maintenance of memory CD8+ T cells. Nat. Immunol. 3, 558–563 (2002).

    CAS  PubMed  Google Scholar 

  39. Sun, Q. et al. BCL6 promotes a stem-like CD8+ T cell program in cancer via antagonizing BLIMP1. Sci. Immunol. 8, eadh1306 (2023).

    CAS  PubMed  Google Scholar 

  40. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Sekaly, R. et al. Dual blockade of IL-10 and PD-1 leads to control of SIV viral rebound following analytical treatment interruption. Preprint at Res. Sq. https://doi.org/10.21203/rs.3.rs-3175716/v1 (2023).

  42. Petrovas, C. et al. Follicular CD8 T cells accumulate in HIV infection and can kill infected cells in vitro via bispecific antibodies. Sci. Transl. Med. 9, eaag2285 (2017).

    PubMed  PubMed Central  Google Scholar 

  43. Yates, K. B. et al. Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans. Nat. Immunol. 22, 1020–1029 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Bruner, K. M. et al. A quantitative approach for measuring the reservoir of latent HIV-1 proviruses. Nature 566, 120–125 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. 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).

    CAS  PubMed  Google Scholar 

  46. Kiniry, B. E. et al. Differential expression of CD8+ T cell cytotoxic effector molecules in blood and gastrointestinal mucosa in HIV-1 infection. J. Immunol. 200, 1876–1888 (2018).

    CAS  PubMed  Google Scholar 

  47. Passaes, C. et al. Optimal maturation of the SIV-specific CD8+ T cell response after primary infection is associated with natural control of SIV: ANRS SIC study. Cell Rep. 32, 108174 (2020).

    CAS  PubMed  Google Scholar 

  48. Rutishauser, R. L. et al. TCF-1 regulates HIV-specific CD8+ T cell expansion capacity. JCI Insight 6, e136648 (2021).

    PubMed  PubMed Central  Google Scholar 

  49. Mvaya, L. et al. Differential localization and limited cytotoxic potential of duodenal CD8+ T cells. JCI Insight 7, e154195 (2022).

    PubMed  PubMed Central  Google Scholar 

  50. Fardoos, R. et al. HIV specific CD8+ TRM-like cells in tonsils express exhaustive signatures in the absence of natural HIV control. Front. Immunol. 13, 912038 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. McGary, C. S. et al. CTLA-4+PD-1− memory CD4+ T cells critically contribute to viral persistence in antiretroviral therapy-suppressed, SIV-infected rhesus macaques. Immunity 47, 776–788 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Li, H. et al. Envelope residue 375 substitutions in simian-human immunodeficiency viruses enhance CD4 binding and replication in rhesus macaques. Proc. Natl Acad. Sci. USA 113, E3413–E3422 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Langner, C. A. & Brenchley, J. M. FRugally optimized DNA cctomer (FRODO) qPCR measurement of HIV and SIV in human and nonhuman primate samples. Curr. Protoc. 1, e93 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Strongin, Z. et al. The role of CD101-expressing CD4 T cells in HIV/SIV pathogenesis and persistence. PLoS Pathog. 18, e1010723 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Hoang, T. N. et al. Baricitinib treatment resolves lower-airway macrophage inflammation and neutrophil recruitment in SARS-CoV-2-infected rhesus macaques. Cell 184, 460–475 (2021).

    CAS  PubMed  Google Scholar 

  56. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Google Scholar 

  58. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).

    CAS  PubMed  Google Scholar 

  59. Black, S. et al. CODEX multiplexed tissue imaging with DNA-conjugated antibodies. Nat. Protoc. 16, 3802–3835 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Deleage, C. et al. Defining HIV and SIV reservoirs in lymphoid tissues. Pathog. Immun. 1, 68–106 (2016).

    PubMed  PubMed Central  Google Scholar 

  61. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    PubMed  PubMed Central  Google Scholar 

  62. Skowronek, P. et al. Rapid and in-depth coverage of the (phospho-)proteome with deep libraries and optimal window design for dia-PASEF. Mol. Cell Proteom. 21, 100279 (2022).

    CAS  Google Scholar 

  63. Bruderer, R. et al. Optimization of experimental parameters in data-independent mass spectrometry significantly increases depth and reproducibility of results. Mol. Cell Proteom. 16, 2296–2309 (2017).

    CAS  Google Scholar 

  64. Lazar, C., Gatto, L., Ferro, M., Bruley, C. & Burger, T. Accounting for the multiple natures of missing values in label-free quantitative proteomics data sets to compare imputation strategies. J. Proteome Res. 15, 1116–1125 (2016).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This manuscript is dedicated in loving memory of our colleague and friend T. Hoang. We thank S. Jean, S. Ehnert, C. Wallace, J. Wood and all veterinary and animal care staff at the Emory National Primate Research Center. We also thank the Emory Flow Cytometry core (K. Gill) for flow cytometry support, the Emory National Primate Research Center Genomics Core for RNA-seq support, the NIH Tetramer Core facility at Emory University for providing tetramer, K. Easley for providing statistical advice and Accelevir Diagnostics for intact proviral DNA measurements. Peptide pools of SIVmac239 Gag were provided by the HIV Reagent Program, which were contributed by the Division of AIDS at NIAID. This work was supported by UM1AI164562 (M.P.), cofunded by NIH/NHLBI/NIDDK/NINDS/NIDA/NIAID and by the NIAID, NIH under award numbers R37AI141258 (R.S.), R01AI116379 (M.P.) and R56AI150401 (M.P.). Additional support was provided by: NIH OD, ORIP, P51 OD011132 and U42 OD011023. This work was supported by the Center for AIDS Research at Emory University (Division of Intramural Research, NIAID, P30AI050409). This project has been funded in whole or in part with federal funds from the National Cancer Institute, National Institutes of Health, under contract no. 75N91019D00024; Divisions of Intramural Research of the National Institute of Allergy and Infectious Diseases, NIH 1ZIAAI001029 (J.M.B.); B03-16 project was supported by funds from the Mexican Government (Programa Presupuestal P016, Anexo 13 del Decreto del Presupuesto de Egresos de la Federación). Sequencing data were acquired on an Illumina NovaSeq6000 funded by NIH S10 OD026799 (S.E.B.). C.M.B. was supported by Swedish Research Council (grant 2023-00510). The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services nor does mention of trade names, commercial products or organizations imply endorsement by the US government. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Z.S. and M.P. conceptualized the study. Z.S., L.R.M., C.D., M.B.P., M.A.C., C.M.B., T.N.H., E.A.U., M.G., K.N., S.L., A.R.R., J.H. and H.T.K. performed the investigations. Z.S., L.R.M., C.D., M.B.P., C.M.B., M.A.C., E.A.U., M.G., G.K.T., A.R.R. and H.T.K. did the formal analysis. P.M.R.E., M.G.-N., Y.A.L., S.A.-R. and G.R.-T. provided resources. Z.S., L.R.M., C.D., M.A.C., C.M.B. and H.T.K. implemented data visualization. C.D., R.S., D.A.K., G.S., J.B., S.E.B., D.E.G., M.R.B. and M.P. acquired funding to support the work. A.S.-C., J.B., S.E.B., D.E.G., M.R.B., H.T.K. and M.P. supervised the work. Z.S., L.R.M. and M.P. wrote the original draft. Z.S., C.D., L.R.M., H.T.K., M.R.B. and M.P. reviewed and edited the final manuscript.

Corresponding author

Correspondence to Mirko Paiardini.

Ethics declarations

Competing interests

Z.S. is employed by and/or has financial interests in Merck and Co.. All other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Ioana Staicu, in collaboration with the Nature Immunology 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 LN TOXhiTCF1+CD39+CD8+ TM cells express high levels of inhibitory receptors after SIV infection.

a) Expression of PD-1, TIGIT, CD101 and Ki-67 on TOX+ and TOX LN CD8+ TM cells at day 42 p.i. (n = 28 macaques). b) UMAP visualization of Phenograph clusters as in a. c) Heatmap of expression of TOX, TCF1, CD39, PD-1, TIGIT, CD101, Ki-67, GzmB and EOMES across all Phenograph clusters as in a. d) Proportion of total CD8+ TM cell population made up by individual Phenograph clusters (n = 28) as in a. e) Frequency of TCF1+CD39+ cells within LN CD8+ TM cells in 5 unique cohorts of macaques: uninfected (n = 10), day 21 p.i. (n = 7), day 35 p.i. (n = 20), day 42 p.i. (n = 28), late chronic infection (mean 17 months p.i. (n = 10). f-g) Expression of TOX, PD-1, TIGIT, CD101, Ki-67 and GzmB in TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of CD8+ TM cells at day 35 p.i. (n = 20) (f) and late chronic infection (n = 10) (g). P values determined by Wilcoxon matched-pairs signed rank test, Kruskal-Wallis one way ANOVA with Dunn’s multiple comparison correction (e) or two-way ANOVA with Tukey’s multiple comparisons test (f-g). Box and whiskers are displayed via Tukey method. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 2 TCF1+CD39+CD8+ TM cells maintain a dual effector and stem-like proteomic profile.

a) Representative gating strategy for sorting of TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of CD8+ TM at day 42 p.i. b-c) Volcano plots displaying significantly differently expressed proteins of interest between sorted LN TCF1+CD39 and TCF1+CD39+ CD8+ TM cells (b) and between sorted LN TCF1+CD39 and TCF1+CD39+ CD8+ TM cells (c) at day 42 p.i. d) Selected proteins with significantly different expression are listed (n = 4 macaques). All statistical comparisons were performed based on two-tailed Student’s t-tests using Spectronaut and custom R scripts.

Extended Data Fig. 3 Sorting of LN SIV-specific CD8+ TM cells.

a) Sorting strategy for LN SIV-specific CM9+CD8+ TM cells for downstream scRNA-seq at day 42 p.i. b) Plots and frequencies of sorted LN SIV-specific CM9+CD8+ TM cells for all 5 macaques as in a.

Extended Data Fig. 4 Transcriptional clustering of LN SIV-specific CD8+ TM cells overlaps with established human CD8 T cell signatures.

a) Heatmap of top 20 differentially expressed genes across the 5 clusters of LN SIV-specific CM9+CD8+ TM cells at day 42 p.i. b) UMAP projection of cells matching established human CD8+ T cell signatures (Supplementary Data File 1) of cell cycling, type 1 interferon, human stem-like CD8 and human terminally differentiated (TD) CD8. c) Distribution of LN SIV-specific CM9+CD8+ TM cells across the 5 clusters for all macaques as in a. d) Scatter plot of expression levels and thresholds (dotted lines) for identifying TCF7+ENTPD1+ CD8+ TM cells as in a.

Extended Data Fig. 5 TCR analysis of expanded TCF7+ENTPD1+CD8+ TM cells demonstrates lineage relationship with terminally differentiated CD8+ TEFF cells.

a-e) UMAP projection of the top 3 clonotypes within TCF7+ENTPD1+ CD8+ TM cells and the top 5 clonotypes of TCF7+ENTPD1+ CD8+ TM cells population from each macaque and the cluster distribution frequency of those clones across all SIV-specific CM9+CD8+ TM cells at day 42 p.i. Number of total cells expressing each TCR clonotype is shown below each bar.

Extended Data Fig. 6 Response of TCF1+CD39+CD8+ TM cells to dual IL-10 + PD-1 mAb blockade.

a) Study schematic demonstrating dosing regimen and timing of lymph node biopsies (week -1 and week 12 post-treatment) for flow cytometry analysis placebo-treated control (n = 6) and treated (n = 9) macaques. b) Fold change of the frequency of TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells, calculated using frequencies at week 12 post-treatment compared to baseline frequencies at week -1 (control (n = 6) and treated (n = 9)). c) Fold change of the frequency of LN Ki-67+ cells within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of CD8+ TM cells (control (n = 6) and treated (n = 9)). P values determined by multiple Mann-Whitney tests with Holm-Sidak correction for multiple comparison. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 7 TCF1+CD39+CD8+ TM cells are associated with reduced viral burden.

a-b) Association of the frequency of TOX+ (a) and TCF1+ CD39+ (b, red) cells within LN CD8+ TM cells with cell-associated SIV RNA levels in LN at day 42 p.i. (n = 26). c-h) Association of the frequency of TCF1+ CD39+ (c-e, red) and TOX+ (f-h) cells within LN CD8+ TM cells with plasma viral load (c,f), total SIV DNA (d,g) and cell associated RNA (e,h) at day 35 p.i. (n = 20). All correlations were determined using two-tailed Spearman analysis.

Extended Data Fig. 8 Expression of CXCR5 by TOX+TCF1+CD39+CD8+ TM cells is associated with viral burden.

a-c) Association of the frequency of CXCR5+ cells within LN CD8+ TM cells with plasma viral load (a), total SIV DNA (b) and cell associated RNA (c) at day 42 p.i.(a n = 27 macaques, b-c n = 25). d-f) Association of frequency of CXCR5+ cells within TCF1+CD39(d, burgundy), TCF1+CD39+ (e, red) and TCF1CD39+ (f, pink) subsets of LN CD8+ TM cells with cell-associated SIV RNA levels in sorted LN memory CD4 T cells as in a (n = 25). g) Frequency of CXCR5+ cells within LN GzmBGzmK+, GzmB+GzmK+ and GzmB+GzmK CD8+ TM cells LN memory CD8 T cells as in a (n = 27). h) Representative image of immunofluorescence staining of CD39 (green) TCF1 (blue) and TOX (red). i) Representative image of immunofluorescence analysis identifying TCF1+ and TOX+ CD8+ T cells in the B cell follicle and T cell zone. Arrows designate examples of TCF1+ TOX+ CD8+ T cells. j-k) Representative image of immunofluorescence analysis identifying SIVinfected CD4+ T cells and TCF1+TOX+CD8+ T cells in the T cell zone (j) and the dark zone of the b cell follicle (k). The perimeter of analysis is defined by the red circle. Blue arrows indicate uninfected CD4+ T cells not surrounded by TCF1+TOX+CD8+ T cells. The dotted line delimits the edge of the b cell follicle. Number of events analyzed per LN area: B cell follicle SIV+ = 175, B cell follicle SIV− = 178, T cell zone SIV+ = 501, T cell zone SIV− = 475, dark zone B cell follicle SIV+ = 217, dark zone bell cell follicle SIV- = 153. All analyses were performed on samples from animals at early chronic infection (D42 p.i.). Correlations represent Spearman analysis. P values were determined using Friedman test one-way ANOVA with Dunn’s multiple comparison test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 9 TCF1+CD39+CD8+ TM cells express high levels of inhibitory receptors and low cytolytic markers in ART-suppressed PLWH.

a-d) Expression frequency of TOX, PD-1, TIGIT (a), Ki-67 (c), GzmB and GzmK (c-d) in TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells in ART-suppressed PLWH (n = 10). P values determined by two-way ANOVA with Tukey’s multiple comparisons test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 10 TCF1+CD39+CD8+ TM cells are not associated with viral burden in ART-naïve PLWH.

a-c) Associations of the frequency of TCF1+CD39 (burgundy), TCF1+CD39+ (red), and TCF1CD39+ (pink) subsets within LN CD8+ TM cells from ART-naïve PLWH with plasma viral load (n = 18) (a), intact reservoir size as measured by IPDA (n = 10) (b) and total HIV DNA (n = 10) (c). d-e) Associations of the frequency of TCF1+CD39 (burgundy), TCF1+CD39+ (red), and TCF1CD39+ (pink) subsets within LN CD8+ TM cells from ART-Suppressed PLWH with intact reservoir size as measured by IPDA (n = 10) (d) and total HIV DNA (n = 10) (e). Correlation evaluated by two-tailed Spearman analysis.

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Tables 1–3.

Reporting Summary

Peer Review File

Supplementary Data 1

scRNA-seq differentially expressed genes and reference human gene signature sets.

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

Strongin, Z., Raymond Marchand, L., Deleage, C. et al. Distinct SIV-specific CD8+ T cells in the lymph node exhibit simultaneous effector and stem-like profiles and are associated with limited SIV persistence. Nat Immunol 25, 1245–1256 (2024). https://doi.org/10.1038/s41590-024-01875-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-024-01875-0

  • Springer Nature America, Inc.

This article is cited by

Navigation