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:

BATF regulates progenitor to cytolytic effector CD8+ T cell transition during chronic viral infection

Abstract

During chronic viral infection, CD8+ T cells develop into three major phenotypically and functionally distinct subsets: Ly108+TCF-1+ progenitors, Ly108CX3CR1 terminally exhausted cells and the recently identified CX3CR1+ cytotoxic effector cells. Nevertheless, how CX3CR1+ effector cell differentiation is transcriptionally and epigenetically regulated remains elusive. Here, we identify distinct gene regulatory networks and epigenetic landscapes underpinning the formation of these subsets. Notably, our data demonstrate that CX3CR1+ effector cells bear a striking similarity to short-lived effector cells during acute infection. Genetic deletion of Tbx21 significantly diminished formation of the CX3CR1+ subset. Importantly, we further identify a previously unappreciated role for the transcription factor BATF in maintaining a permissive chromatin structure that allows the transition from TCF-1+ progenitors to CX3CR1+ effector cells. BATF directly bound to regulatory regions near Tbx21 and Klf2, modulating their enhancer accessibility to facilitate the transition. These mechanistic insights can potentially be harnessed to overcome T cell exhaustion during chronic infection and cancer.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: SCENIC analysis revealed distinct transcriptional regulatory circuits for CD8+ T cell subsets during chronic viral infection.
Fig. 2: Integrated analysis of single-cell transcriptomes reveals analogous CD8+ subsets arising during acute and chronic viral infections.
Fig. 3: T-bet deficiency significantly diminishes TEFF subset formation and function.
Fig. 4: Distinct H3K4me3 and H3K27me3 patterns are associated with gene expression profiles of progenitor, effector and exhausted CD8+ T cell subsets.
Fig. 5: Distinct enhancer repertoires regulate transcriptional programs of three subsets of CD8+ T cells.
Fig. 6: BATF is required for TPRO cell differentiation into TEFF cells.
Fig. 7: BATF modulates enhancer accessibility to facilitate the TPRO-to-TEFF cell transition.

Similar content being viewed by others

Data availability

Both raw data files and processed data files from ATAC-seq and CUT&Tag-seq experiments were deposited in the GEO database with accession codes GSE149752, GSE149796 and GSE149810. Source data are provided with this paper.

References

  1. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. He, R. et al. Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412–428 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. 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 

  4. 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 

  5. 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 

  6. 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).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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).

  9. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Liu, X. et al. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature 567, 525–529 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. Man, K. et al. Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity 47, 1129–1141 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Martinez, G. J. et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Staron, M. M. et al. The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8+ T cells during chronic infection. Immunity 41, 802–814 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shin, H. et al. A role for the transcriptional repressor Blimp-1 in CD8+ T cell exhaustion during chronic viral infection. Immunity 31, 309–320 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

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

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

  19. 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 

  20. Henning, A. N., Roychoudhuri, R. & Restifo, N. P. Epigenetic control of CD8+ T cell differentiation. Nat. Rev. Immunol. 18, 340–356 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pauken, K. E. et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ghoneim, H. E. et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142–157 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Xin, G. et al. A critical role of IL-21-induced BATF in sustaining CD8-T-cell-mediated chronic viral control. Cell Rep. 13, 1118–1124 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cheung, K. L. et al. Distinct roles of Brd2 and Brd4 in potentiating the transcriptional program for TH17 cell differentiation. Mol. Cell 65, 1068–1080 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kurachi, M. et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat. Immunol. 15, 373–383 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ciofani, M. et al. A validated regulatory network for TH17 cell specification. Cell 151, 289–303 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Rao, R. R., Li, Q., Gubbels Bupp, M. R. & Shrikant, P. A. Transcription factor Foxo1 represses T-bet-mediated effector functions and promotes memory CD8+ T cell differentiation. Immunity 36, 374–387 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhou, X. & Xue, H.-H. Cutting edge: generation of memory precursors and functional memory CD8+ T cells depends on T cell factor-1 and lymphoid enhancer-binding factor-1. J. Immunol. 189, 2722–2726 (2012).

    Article  CAS  Google Scholar 

  31. Bancos, S., Cao, Q., Bowers, W. J. & Crispe, I. N. Dysfunctional memory CD8+ T cells after priming in the absence of the cell cycle regulator E2F4. Cell. Immunol. 257, 44–54 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Utzschneider, D. T. et al. Active maintenance of T cell memory in acute and chronic viral infection depends on continuous expression of FOXO1. Cell Rep. 22, 3454–3467 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. Dominguez, C. X. et al. The transcription factors ZEB2 and T-bet cooperate to program cytotoxic T cell terminal differentiation in response to LCMV viral infection. J. Exp. Med. 212, 2041–2056 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Omilusik, K. D. et al. Transcriptional repressor ZEB2 promotes terminal differentiation of CD8+ effector and memory T cell populations during infection. J. Exp. Med. 212, 2027–2039 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. 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 

  37. Intlekofer, A. M. et al. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat. Immunol. 6, 1236–1244 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kao, C. et al. Transcription factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8+ T cell responses during chronic infection. Nat. Immunol. 12, 663–671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Anderson, A. C. et al. T-bet, a TH1 transcription factor regulates the expression of Tim-3. Eur. J. Immunol. 40, 859–866 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Crompton, J. G. et al. Lineage relationship of CD8+ T cell subsets is revealed by progressive changes in the epigenetic landscape. Cell. Mol. Immunol. 13, 502–513 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Russ, B. E. et al. Distinct epigenetic signatures delineate transcriptional programs during virus-specific CD8+ T cell differentiation. Immunity 41, 853–865 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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  CAS  PubMed  PubMed Central  Google Scholar 

  44. Scott-Browne, J. P. et al. Dynamic changes in chromatin accessibility occur in CD8+ T cells responding to viral infection. Immunity 45, 1327–1340 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wei, J. et al. Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature 576, 471–476 (2019).

  46. Murphy, T. L., Tussiwand, R. & Murphy, K. M. Specificity through cooperation: BATF–IRF interactions control immune-regulatory networks. Nat. Rev. Immunol. 13, 499–509 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Grigoryan, G., Reinke, A. W. & Keating, A. E. Design of protein-interaction specificity gives selective bZIP-binding peptides. Nature 458, 859–864 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Glasmacher, E. et al. A genomic regulatory element that directs assembly and function of immune-specific AP-1–IRF complexes. Science 338, 975–980 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li, P. et al. BATF–JUN is critical for IRF4-mediated transcription in T cells. Nature 490, 543–546 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Betz, B. C. et al. Batf coordinates multiple aspects of B and T cell function required for normal antibody responses. J. Exp. Med. 207, 933–942 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schauder, D. M. et al. E2A-regulated epigenetic landscape promotes memory CD8 T cell differentiation. Proc. Natl Acad. Sci. USA 118, e2013452118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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 

  53. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1930 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. 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 

  56. Meers, M. P., Tenenbaum, D. & Henikoff, S. Peak calling by Sparse Enrichment Analysis for CUT&RUN chromatin profiling. Epigenetics Chromatin 12, 42 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Ye, T., Ravens, S., Krebs, A. R. & Tora, L. Interpreting and visualizing ChIP–seq data with the seqMINER software. Methods Mol. Biol. 1150, 141–152 (2014).

    Article  PubMed  Google Scholar 

  59. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. 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  CAS  Google Scholar 

  63. Shao, Z., Zhang, Y., Yuan, G. C., Orkin, S. H. & Waxman, D. J. MAnorm: a robust model for quantitative comparison of ChIP–seq data sets. Genome Biol. 13, R16 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work is supported by NIH grants AI125741 (W.C.), AI148403 (W.C.), AI153537 (R.A.Z.), DK108557 (D.M.S.) and DK127526 (M.Y.K.); by an American Cancer Society Research Scholar grant (W.C.); and by an Advancing a Healthier Wisconsin Endowment grant (W.C.). R.A.Z. is supported by a Cancer Research Institute Irvington fellowship. D.M.S. and M.Y.K. are members of the Medical Scientist Training Program at the MCW, which is partially supported by a training grant from the NIGMS (T32-GM080202). This research was completed in part with computational resources and technical support provided by the Research Computing Center at the MCW. We thank S. Henikoff for providing the 3×Flag-pA-Tn5-Fl plasmid and N. Zhu for providing the protein A–Tn5 fusion protein.

Author information

Authors and Affiliations

Authors

Contributions

Y.C., R.A.Z., X.W., D.M.S., J.S. and W.C. designed and performed experiments and analyzed data. M.Y.K., S.Z. and R.B. helped with sequencing data analysis and visualization. E.J.T. provided helpful insights and contributed a key biological resource (Batffl/fl mice). Y.C. and W.C. wrote the manuscript. W.C. supervised the study.

Corresponding author

Correspondence to Weiguo Cui.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Stephen Turner, Daniel Utzschneider and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. 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 SCENIC analysis revealed distinct transcriptional regulatory circuits for CD8+ T cell subsets during chronic viral infection.

a, Dot plot showing expression of signature genes for the TPRO cells, TEFF cells, and TEXH cells. b,c,d, t-SNE projections showing binary regulon activity of cell-specific regulons for the TPRO, TEXH and TEFF subsets.

Extended Data Fig. 2 Unsupervised clustering analysis identified three major cell populations in the integrated dataset.

a, Heatmap showing the top 15 differentially expressed genes for each cluster as defined in Fig. 2b. Columns and rows correspond to cells and genes, respectively. Cells from the same cluster are grouped together. The color scale representing Z-Score that is generated from log2 read counts. b,c,d,e,h, Showing the comparison of CD8 T cells from day 9 post-acute infection and day 30 post-chronic infection. Cells are gated on CD8+CD44+ GP33-41+ cells. b, left. Summary data showing the expression of surface makers for SLECs and TEFF cells. b, right. Summary data showing the expression of surface makers for MPECs and TPRO cells. c, Summary data showing the expression of GzmB, proportion of CD8+ T cell degranulating (CD107a+) and producing IFN-γ, and proportion of CD8+ T cell producing TNF and IFN-γ upon ex vivo stimulation with GP33-41 peptide. d,h, Summary data showing the expression of transcription factors and inhibitory receptors. e, Summary data showing the relative cytotoxicity of SLECs and TEFF cells against peptide-pulsed target cells. f, Heatmap showing binary regulon activities of CD8 T cells from day 9 post-acute infection and day 30 post-chronic infection. g, t-SNE projection depicting clustering of cells by regulon activity. b, 10 mice for acute infection and 10 mice for chronic infection. c,d,h, 5 mice for acute infection and 5 mice for chronic infection. e, 10 mice for acute infection and 7 mice for chronic infection. b-h, Data pooled from 2 independent experiments. Data are expressed as mean ± s.e.m. ns = not significant, * p < 0.05, ** p < 0.01, *** p < 0.001. b, Unpaired t-test with two-stage step-up method of Benjamini, Krieger, and Yekutieli. c,d,e,h, Ordinary one-way ANOVA.

Source data

Extended Data Fig. 3 Distinct H3K4me3 and H3K27me3 patterns associated with gene expression profiles of the three CD8+ T cell subsets.

a, Sorting strategy and sort purity of GP33-41 tetramer+ or GP276-286 tetramer+ virus-specific CD8+ T cell subsets that were sorted from C57BL/6 mice at 3-5 weeks post LCMV Cl13 infection. b,c, Heatmap showing gene promoter regions that exhibited differential H3K4me3 enrichment between TEFF cells and TEXH cells. Scale bar representing Z-Score that is generated from log2 RPKM. Bar plot showing the top pathways correlated with these genes by gene ontology (GO) analysis. d, Genome track view of representative gene loci showing H3K4me3 (green, above line) or H3K27me3 (red, below line) peaks. CUT & Tag-seq data are from two independent replicates. Each replicate was pooled together from 2-3 mice.

Extended Data Fig. 4 Distinct enhancer repertoires regulate transcriptional programs of the three CD8+ T cell subsets.

Venn diagram showing overlap of all chromatin-accessible regions (ChARs) detected by ATAC-seq. b, Bar plot showing the distance of ChARs to TSS. Left are ChARs with differential accessibility among the three CD8+ T cells subsets. Right are ChARs shared by the three subsets. c, Heatmap showing MSigDB pathway enrichment signatures in six enhancer peak sets as shown in Fig. 5c (Top). d, MA plot showing M value vs A value of the merged set of TEFF cell and TEXH cell enhancer peaks after normalization. Top 5,000 peaks are highlighted for TEFF cells (cyan) and TEXH cells (red). Dot plot showing the top 10 TFs whose motifs were significantly enriched in TEFF cell-specific enhancers compared to TEXH cell-specific enhancers.

Extended Data Fig. 5 Active and suppressive states of enhancer regions regulate gene expression in the three CD8 T cell subsets.

a, Genome track view of the gene loci showing ATAC-seq, H3K27ac, and H3K27me3 peaks in TPRO, TEXH and TEFF subsets, TFs with predictive binding sites at the enhancers (green shadow) were listed.

Extended Data Fig. 6 BATF is required for TPRO progenitor cell differentiation into TEFF cells.

a, Summary data showing viral titers in the serum of experimental mice on day 8 p.i. and day 30 p.i. b, c, Summary data showing the relative expression of KLRG1 and TCF-1 in three virus-specific CD8+ T cell subsets. d, Experimental design. e, g, Representative flow plots and summary data showing the proportion of three antigen-specific CD8+ T cell subsets in WT and BATF deficient cells on day 28 p.i. with LCMV Cl13. f, Summary data showing the expression of BATF in WT and BATF deficient cells. a, Day 8 data was collected from 4 WT and 6 BATF-HET mice. Day 21 data was collected from 4 WT and 5 BATF-HET mice. b,c, Data was collected from 4 WT and 4 BATF-HET mice. f,g, Data was collected from 8 Batf flox/ flox CreERT2 mT/mG mice. c-j, Data pooled from 2 independent experiments. Data are expressed as mean ± s.e.m. ns = not significant, * p < 0.05, ** p < 0.01, *** p < 0.001. a, Two-tailed unpaired t-test. b,c, Unpaired t-test with Holm-Šídák method. f, Two-tailed paired t-test. g, Paired t-test with two-stage step-up method of Benjamini, Krieger, and Yekutieli.

Source data

Extended Data Fig. 7 BATF regulates chromatin accessibility of CD8+ T cells during chronic infection.

Gzmb-Cre+;Batf+/+;RosamT/mG (WT), Gzmb-Cre+;Batffl/+;RosamT/mG (BATF-HET), and Gzmb-Cre+;Batffl/fl;RosamT/mG mice (BATF-KO) were used for this experiment. At 4 weeks post-LCMV Cl13 infection, CD8+CD44+GFP+ cells, which represent polyclonal LCMV-specific CD8+ T cells, were FACS sorted to perform ATAC-seq experiments. a, Venn diagram showing overlapping and unique enhancer regions in WT, BATF-HET, and BATF-KO CD8+ T cells b, Heatmap showing enhancer regions with differential accessibility. Scale bar representing Z-Score that is generated from log2 FPKM. Each replicate was an individual mouse.

Extended Data Fig. 8 Genome track view of Pdcd1, Tcf7, Id3, Cxcr5, Cx3cr1, and Irf8.

a,b, Genome track view of the gene loci showing ATAC-seq, H3K27ac, and BATF CUT&Tag peaks in TPRO, TEXH and TEFF subsets.

Supplementary information

Source data

Source Data Fig. 3

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Zander, R.A., Wu, X. et al. BATF regulates progenitor to cytolytic effector CD8+ T cell transition during chronic viral infection. Nat Immunol 22, 996–1007 (2021). https://doi.org/10.1038/s41590-021-00965-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00965-7

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