Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Jul;25(7):1245-1256.
doi: 10.1038/s41590-024-01875-0. Epub 2024 Jun 17.

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

Affiliations

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

Zachary Strongin et al. Nat Immunol. 2024 Jul.

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 TCF1-CD39+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.

PubMed Disclaimer

Conflict of interest statement

Competing interests

ZS is employed by and/or has financial interests in Merck & Co., Inc.

Figures

Extended Data Fig. 1
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
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
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
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
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
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
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
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
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
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.
Figure 1.
Figure 1.. TOX is upregulated on CD8+ T cells after SIV infection.
a) Representative flow cytometry staining of TOX in total CD8+ T cells in lymph nodes (LN) isolated from macaques infected with SIV at day 42 and late chronic infection (mean 17 months p.i.) or left uninfected. b) Frequency of TOX+ cells in total CD8+ T cells in the LN of uninfected macaques (n=10), or in SIV-infected macaques at day 42 p.i. (n=28.) and late chronic infection(mean 17 months p.i.) (n=10). c) Representative flow cytometry staining of TOX in CD95CD28+ naïve CD8+ T cells (CD8+ TN cells), CD95+ memory CD8+ T cells (CD8+ TM cells) and SIV-specific CM9+CD8+ T cells at day 42 p.i. d) Frequency of TOX+ cell with CD8+ TN cells, CD8+ TM cells and CM9+CD8+ T cells in the LN of A*01+ macaques (n=7) at day 42 p.i. e) UMAP plot of flow cytometry data showing expression patterns TOX, TCF1, CD39, PD-1, TIGIT, CD101, Ki-67, GzmB and EOMES in CD8+ TM cells at day 42p.i. f) Heatmap showing the expression of markers of stemness, differentiation and effector capacity within a subset of clusters that represent CD95+GzmB+CD8+ TEFF cells and TCF1hiCD39hi CD8+ T cells, as identified through Phenograph analysis of flow cytometry data. P values were determined using Kruskal-Wallis one-way ANOVA with Dunn’s multiple comparisons (b) or Friedman’s one-way ANOVA with Dunn’s multiple comparisons (d). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 2.
Figure 2.. TCF1+CD39+CD8+ T cells expand after SIV infection and are a unique phenotypic population.
a,b) Representative staining of TCF1 and CD39 (a) and frequency of TCF1+CD39, TCF1+CD39+ and TCF1CD39+ cells within CD8+ TM cells (b) in the LN of SIV-infected macaques at day 42 p.i. (n=28) and late chronic infection (mean 17 months p.i.) (n=10) or uninfected (n=10). c) Expression of TOX, PD-1, TIGIT, CD101 and Ki-67 in TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of CD8+ TM cells at day 42 p.i. in the LN of SIV infected macaques (n=28 macaques). d-f) Representative staining of GzmB and GzmK (d) and frequency of GzmB+ (e), GzmBGzmK+, GzmB+GzmK+ and GzmB+GzmK cells in TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets (f) within LN CD8+ TM cells as in c ((e) n=28, (f) n=27). h) Frequency of CD107a+ and CD107a+IFNγ+ cells within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of CD8+ TM cells as in c in response to SIV Gag peptide stimulation (n=26 macaques). Background subtracted values from only animals exhibiting significant responses (>0.01% of cells responding) are shown. P values were determined using two-way ANOVA with Tukey’s multiple comparisons test (b,f), Friedman test one way-ANOVA with Dunn’s multiple comparisons (c,e) or mixed effects two-way ANOVA with Holm-Šídák multiple comparison. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 3.
Figure 3.. LN SIV-specific TCF1+CD39+CD8+ T cells maintain a dual effector and stem-like transcriptional profile.
a) Representative flow cytometry staining of SIV-specific CM9+CD8+ TM cells is shown. b) UMAP cluster analyses of LN SIV-specific CM9+CD8+ TM cells from 5 macaques at day 42 p.i. c) UMAP plots showing expression of GZMA, GZMB, GZMK, PRF1, IFNG, PDCD1, TIGIT, LAG3, CTLA4, TCF7, SELL, JUN, FOS, IL7R, MKI67, BRCA1, IRF7, MX1, ENTPD1 and TOX in LN SIV-specific CM9+CD8+ TM cells as in b. d) UMAP projection of TCF7+ENTPD1+ CM9+CD8+ TM cells as in b. E) Distribution of all of LN SIV-specific CM9+CD8+ TM cells and CM9+TCF7+ENTPD1+ CD8+ TM cells among the 5 identified clusters. f-g) Volcano plot displaying differentially expressed genes between cluster 2 (stem) and TCF7+ENTPD1+ CD8+ TM cells (f) and cluster 1 (effector) and TCF7+ENTPD1+ CD8+ TM cells as in b. Selected genes with significantly different expression are listed. H) Top 3 clonotypes within TCF7+ENTPD1+ CD8+ TM cells in one representative macaque. Each color represents a unique TCR clonotype. I) Top 5 clonotypes of TCF7+ENTPD1+ CD8+ TM cells from one representative macaque and the cluster distribution frequency of clones across all SIV-specific CM9+CD8+ TM cells. Number of total cells expressing each TCR clonotype is denoted below each bar.
Figure 4.
Figure 4.. TOX+ and TCF1+CD39+CD8+ TM cell frequencies are associated with reduced viral burden.
a) Association of frequency of TOX+ cells within LN CD8+ TM cells and plasma viral load at day 42 p.i.(n=28). b) Association of frequency of TOX+ cells within LN SIV-specific CM9+CD8+ TM cells and plasma viral load as in a (n=8). c) Association of frequency of TCF1+CD39, TCF1+CD39+ and TCF1CD39+ cells within LN CD8+ TM cells and plasma viral load as in a (n=28). d-e) Association of frequency of TOX+ (d) or TCF1+CD39+ (e) cells within LN CD8+ TM cells and total SIV DNA in LN CD4+ TM cells as in a (n=26). f-g) Association of frequency of TOX+ (f) or TCF1+CD39+ (g) cells within LN CD8+ TM cells and memory CD4+ T cell counts in peripheral blood (n=28). All correlations were evaluated using two-tailed Spearman analysis.
Figure 5.
Figure 5.. TCF1+CD39+CD8+ TM cells preferentially penetrate the follicular environment.
a-b) Representative flow cytometry staining (a) and Frequency of CXCR5+ cells within TCF1+CD39, TCF1+CD39+, TCF1CD39+, TOX+ and TOX subsets of CD8+ TM cells in LN at day 42 p.i. (b) (n=27 macaques). c) Association of CXCR5 expression frequency within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of CD8+ TM cellsand plasma viral load as in b (n=27). d) Association of CXCR5 expression frequency within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of CD8+ TM cells and SIV DNA levels in LN CD4+ TM cells as in b (n=25). e) Quantification of the proportion of total CD8+ T cells expressing TOX and TCF1 within B cell follicles or T cell zones as in b (n=8). f) RNAscope with PhenoCycler-Fusion immunostaining-based proximity scoring analysis of TOX+TCF1+CD8+ T cells with SIV-infected CD4+ cells in the T cell zone and B cell follicle as in b (n=11–21 animals). P values were determined using Friedman test one-way ANOVA with Dunn’s multiple comparisons (b, within TCF1/CD39 subsets), two-tailed Wilcoxon (b, within TOX+/TOX) or two-way ANOVA with Holm-Šídák multiple comparison. Correlations were deteremined using two-tailed Spearman analysis. Differences in proximity scores were assessed on mean values of proximity scores for infected cells versus noninfected ones within the different tissue compartments. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 6.
Figure 6.. TCF1+CD39+CD8+ TM cells contract during long-term ART but remain associated with viral burden.
a-b) Representative flow cytometry staining (a) and frequency of TOX+ cells within within LN CD8+ TM cells from longitudinally matched macaques at day 42 p.i. and week 64 post-ART initiation (b) (n=28 day 42 p.i., n=25 ART). c-d) Representative flow cytometry staining (c) and frequency of TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells as in b (n=25). e) Expression of TOX, PD-1, TIGIT, CD101, Ki-67 and GzmB within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells as in b (n=25). F) Association between frequency of LN TCF1+CD39+ CD8+ TM cells and levels of intact SIV DNA in total LN CD4+ T cells at week 64 post-ART initiation (n=24). P values were determined using two-tailed Wilcoxon (b), multiple two-tailed Wilcoxon (d) or Friedman test one way-ANOVA with Dunn’s multiple comparisons. Correlation evaluated using two-tailed Spearman analysis. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Figure 7.
Figure 7.. HIV infection results in expansion of LN HIV-specific TCF1+CD39+CD8+ TM cells.
a-b) Representative flow cytometry staining (a) and (b) proportion of LN TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells in uninfected individuals (n=4), ART-naïve PLWH (n=18) and ART-suppressed PLWH (n=10). c-d) Expression of TOX, PD-1, TIGIT and Ki-67 (c), GzmB (d) within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells ART-naïve PLWH (n=18 participants). e) Proportion of cells within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells expressing one or both of Gzmb and Gzmk as in c (n=18 participants). f) Proportion of cells within TCF1+CD39, TCF1+CD39+ and TCF1CD39+ subsets of LN CD8+ TM cells expressing CD107a and CD107a in combination with production of IFNy in response to HIV Gag peptide stimulation as in c (n=8 participants). Background substracted values and only samples exhibiting significant responses (>0.01% of cells responding) are shown. P values were determined using two-way ANOVA with Holm-Šídák multiple comparison (b), Friedman test one-wayANOVA with Dunn’s multiple comparisons (c,d,f) or two-way ANOVA with Tukey’s multiple comparison. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

References

    1. Blank CU et al. Defining ‘T cell exhaustion’. Nat Rev Immunol 19, 665–674 (2019). - PMC - PubMed
    1. Alfei F et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019). - PubMed
    1. Scott AC et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019). - PMC - PubMed
    1. Khan O et al. TOX transcriptionally and epigenetically programs CD8(+) T cell exhaustion. Nature 571, 211–218 (2019). - PMC - PubMed
    1. 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). - PMC - PubMed

Methods only references

    1. McGary CS 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 e775 (2017). - PMC - PubMed
    1. 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 U S A 113, E3413–3422 (2016). - PMC - PubMed
    1. Langner CA & Brenchley JM FRugally Optimized DNA Octomer (FRODO) qPCR Measurement of HIV and SIV in Human and Nonhuman Primate Samples. Curr Protoc 1, e93 (2021). - PMC - PubMed
    1. Strongin Z et al. The role of CD101-expressing CD4 T cells in HIV/SIV pathogenesis and persistence. PLoS Pathog 18, e1010723 (2022). - PMC - PubMed
    1. Hoang TN et al. Baricitinib treatment resolves lower-airway macrophage inflammation and neutrophil recruitment in SARS-CoV-2-infected rhesus macaques. Cell 184, 460–475.e421 (2021). - PMC - PubMed

MeSH terms