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:

BACH2 enforces the transcriptional and epigenetic programs of stem-like CD8+ T cells

An Author Correction to this article was published on 03 March 2021

This article has been updated

Abstract

During chronic infection and cancer, a self-renewing CD8+ T cell subset maintains long-term immunity and is critical to the effectiveness of immunotherapy. These stem-like CD8+ T cells diverge from other CD8+ subsets early after chronic viral infection. However, pathways guarding stem-like CD8+ T cells against terminal exhaustion remain unclear. Here, we show that the gene encoding transcriptional repressor BACH2 is transcriptionally and epigenetically active in stem-like CD8+ T cells but not terminally exhausted cells early after infection. BACH2 overexpression enforced stem-like cell fate, whereas BACH2 deficiency impaired stem-like CD8+ T cell differentiation. Single-cell transcriptomic and epigenomic approaches revealed that BACH2 established the transcriptional and epigenetic programs of stem-like CD8+ T cells. In addition, BACH2 suppressed the molecular program driving terminal exhaustion through transcriptional repression and epigenetic silencing. Thus, our study reveals a new pathway that enforces commitment to stem-like CD8+ lineage and prevents an alternative terminally exhausted cell fate.

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: Bach2 locus is epigenetically and transcriptionally active in stem-like CD8+ T cells.
Fig. 2: BACH2 overexpression promotes stem-like CD8+ differentiation and inhibits exhaustion.
Fig. 3: BACH2 establishes a stem-like transcriptional program at the single-cell level.
Fig. 4: BACH2 overexpression alters the expression of genes and pathways in stem-like CD8+ T cells.
Fig. 5: BACH2 deficiency impairs the differentiation of stem-like CD8+ T cells.
Fig. 6: BACH2 is required for the transcriptional program of stem-like CD8+ T cells.
Fig. 7: BACH2 regulates the epigenetic program of antiviral CD8+ T cells.

Similar content being viewed by others

Data availability

All data generated to support this study are available within the paper. The ATAC-seq (Figs. 1 and 7 and Extended Data Figs. 1 and 7), scRNA-seq (Figs. 3 and 6 and Extended Data Figs. 3 and 6) and RNA-seq (Figs. 4 and 6 and Extended Data Figs. 4 and 6) data have been deposited at the Gene Expression Omnibus (GEO accession no. GSE152379). H3K27ac ChIP–seq and RNA-seq data of stem-like and terminally exhausted CD8 T cells were previously published (GEO accession no. GSE119943).

Code availability

Custom code used for ATAC-seq, RNA-seq and scRNA-seq analyses is available from the corresponding author upon request.

Change history

References

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

    CAS  PubMed  Google Scholar 

  2. 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).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  4. 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 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  7. 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 

  8. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  10. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Chu, H. H. et al. Continuous effector CD8+ T cell production in a controlled persistent infection is sustained by a proliferative intermediate population. Immunity 45, 159–171 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Jansen, C. S. et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. 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 (2019).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Blackburn, S. D., Shin, H., Freeman, G. J. & Wherry, E. J. Selective expansion of a subset of exhausted CD8+ T cells by αPD-L1 blockade. Proc. Natl Acad. Sci. USA 105, 15016–15021 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. 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).

    CAS  PubMed  Google Scholar 

  20. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. 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 

  24. 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 

  25. Gautam, S. et al. The transcription factor c-Myb regulates CD8+ T cell stemness and antitumor immunity. Nat. Immunol. 20, 337–349 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Huang, Z. et al. IL-27 promotes the expansion of self-renewing CD8+ T cells in persistent viral infection. J. Exp. Med. 216, 1791–1808 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Igarashi, K., Kurosaki, T. & Roychoudhuri, R. BACH transcription factors in innate and adaptive immunity. Nat. Rev. Immunol. 17, 437–450 (2017).

    CAS  PubMed  Google Scholar 

  30. Kim, E. H. et al. Bach2 regulates homeostasis of Foxp3+ regulatory T cells and protects against fatal lung disease in mice. J. Immunol. 192, 985–995 (2014).

    CAS  PubMed  Google Scholar 

  31. Roychoudhuri, R. et al. BACH2 regulates CD8+ T cell differentiation by controlling access of AP-1 factors to enhancers. Nat. Immunol. 17, 851–860 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Roychoudhuri, R. et al. BACH2 represses effector programs to stabilize Treg-mediated immune homeostasis. Nature 498, 506–510 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Herndler-Brandstetter, D. et al. KLRG1+ effector CD8+ T cells lose KLRG1, differentiate into all memory T cell lineages, and convey enhanced protective. Immunity 48, 716–729 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Richer, M. J., Lang, M. L. & Butler, N. S. T cell fates zipped up: how the Bach2 basic leucine zipper transcriptional repressor directs T cell differentiation and function. J. Immunol. 197, 1009–1015 (2016).

    CAS  PubMed  Google Scholar 

  35. Singer, M. et al. A distinct gene module for dysfunction uncoupled from activation in tumor-infiltrating T cells. Cell 166, 1500–1511 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. 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 

  39. Tsukumo, S. et al. Bach2 maintains T cells in a naive state by suppressing effector memory-related genes. Proc. Natl Acad. Sci. USA 110, 10735–10740 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. 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).

    PubMed  PubMed Central  Google Scholar 

  42. Shan, Q. et al. The transcription factor Runx3 guards cytotoxic CD8+ effector T cells against deviation towards follicular helper T cell lineage. Nat. Immunol. 18, 931–939 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Vahedi, G. et al. Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520, 558–562 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Afzali, B. et al. BACH2 immunodeficiency illustrates an association between super-enhancers and haploinsufficiency. Nat. Immunol. 18, 813–823 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, D. et al. The transcription factor Runx3 establishes chromatin accessibility of cis-regulatory landscapes that drive memory cytotoxic T lymphocyte formation. Immunity 48, 659–674 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  47. Pircher, H., Burki, K., Lang, R., Hengartner, H. & Zinkernagel, R. M. Tolerance induction in double specific T-cell receptor transgenic mice varies with antigen. Nature 342, 559–561 (1989).

    CAS  PubMed  Google Scholar 

  48. Kometani, K. et al. Repression of the transcription factor Bach2 contributes to predisposition of IgG1 memory B cells toward plasma cell differentiation. Immunity 39, 136–147 (2013).

    CAS  PubMed  Google Scholar 

  49. Wu, T. et al. TCF1 is required for the T follicular helper cell response to viral infection. Cell Rep. 12, 2099–2110 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Ji, Y. et al. miR-155 harnesses Phf19 to potentiate cancer immunotherapy through epigenetic reprogramming of CD8+ T cell fate. Nat. Commun. 10, 2157 (2019).

    PubMed  PubMed Central  Google Scholar 

  51. Leisegang, M. et al. Enhanced functionality of T cell receptor-redirected T cells is defined by the transgene cassette. J. Mol. Med. 86, 573–583 (2008).

    CAS  PubMed  Google Scholar 

  52. Shih, H. Y. et al. Developmental acquisition of regulomes underlies innate lymphoid cell functionality. Cell 165, 1120–1133 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 1193–1203 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  56. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. 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).

    CAS  PubMed  Google Scholar 

  58. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Trapnell, C. & Salzberg, S. L. How to map billions of short reads onto genomes. Nat. Biotechnol. 27, 455–457 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Kupfer (CU), G. Hedlund (CU), K. Helm (CU), Y. Zhang (CU), M. Kirby (NHGRI), S. Anderson (NHGRI), J. Reilley (NIAID), R. Handon (NHGRI), L. Garrett (NHGRI), I. Ginty (NHGRI), G. Gutierrez-Cruz (NIAMS) and S. Dell’Orso (NIAMS) for their excellent technical support. This research was supported in part by an NIH grant (AG056524) to T.W.; the intramural programs of the NIAMS (to J.J.O.), NIAID (to P.L.S.), NHGRI (to P.L.S.), NINDS (to D.B.M.), National Cancer Institute (to L.G.) and NIDDK (to B.A.); an NIH Office of Dietary Supplements Research Scholar scholarship to D.C.; grants from the NIH (AI105343, AI117950, AI082630, AI112521, AI115712, AI108545 and CA210944) and Stand Up To Cancer to E.J.W.; and NIH grant (CA234842) to Z.C. E.J.W. is also supported by the Parker Institute for Cancer Immunotherapy, which supports the Cancer Immunology program at Penn Institute for Immunology.

Author information

Authors and Affiliations

Authors

Contributions

T.W. and C.Y. conceived the study. T.W. and C.Y. designed the experiments. C.Y., G.L., T.W., Z.Z., Y.S., E.A.M., D.C., M.A.D., W.S. and J.C. performed the experiments. C.Y., T.W., G.L., H.-W.S., Z.C., P.L.S., J.J.O., D.B.M., L.G., B.A., E.J.W. and Y.Z. analyzed and interpreted the results. T.W. and C.Y. drafted the manuscript. J.S. designed and generated retroviral gRNA constructs targeting Prdm1 and Runx3. K.K. and T.K. provided mouse models critical to this study.

Corresponding authors

Correspondence to Chen Yao or Tuoqi Wu.

Ethics declarations

Competing interests

E.J.W. has consulting agreements with and/or is on the scientific advisory board for Merck, Roche, Pieris, Elstar, Related Sciences and Surface Oncology. E.J.W. is a founder of Surface Oncology and Arsenal Biosciences. E.J.W. has a patent licensing agreement on the PD-1 pathway with Roche/Genentech. L.G. is inventor on a patent describing methods for the generation and isolation of stem-like memory T cells. L.G. has consulting agreements with Lyell Immunopharma and Advaxis Immunotherapies.

Additional information

Peer review information Nature Immunology thanks Stephen Jameson 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 Stem-like and terminally exhausted CD8+ T cells exhibited distinct open chromatin landscapes.

a, Experimental setup has been described in Fig. 1a. Gating strategy for sorting stem-like (Ly108hiTIM-3lo) and terminally exhausted (Ly108loTIM-3hi) P14 cells on day 7 p.i.. b, Venn diagram illustrating the numbers of ATAC-Seq peaks in stem-like and terminally exhausted CD8+ T cells. c, Percentages of common and variable ATAC-Seq peaks, as defined in Extended Data Fig. 1b, annotated to Intergenic, Intronic, and Promoter-TSS regions. Filled bars represent common peaks; unfilled bars represent variable peaks. P values were determined by the Chi-squared test. d, Volcano plots of differentially accessible chromatin regions between stem-like and terminally exhausted CD8+ T cells are shown. The x axis represents log2 fold change of chromatin accessibility; the y axis represents -log10FDR. The horizontal dash line indicates FDR = 0.1; vertical dash lines indicate fold change of +/− 1.2. Dots represent peaks associated with significantly upregulated genes (fold change >1.5, FDR < 0.05) in stem-like (red) or terminally exhausted (green) CD8+ T cells, and non-significantly regulated genes (grey). FDR is calculated by edgeR. ATAC-Seq data include n = 2 independent experiments per group. e, The transcript abundance of Bach2 and Prdm1 in stem-like and terminally exhausted P14 CD8+ T cells harvested on day 7 (left, n = 2 independent samples) and day 20 (right, n = 3 independent samples) after LCMV clone 13 infection are shown in bar graphs. FDR was determined using edgeR. f, Cas9; P14 CD8+ T cells transduced with control or Prdm1 gRNA constructs were adoptively transferred into C57BL/6 mice that were subsequently infected with LCMV clone 13. Splenic P14 cells were analyzed on day 7 p.i.. The protein expression of BACH2 in control or Prdm1 gRNA transduced P14 CD8+ T cells are shown in representative FACS plot (left) and bar graph (right). g. A Venn diagram shows transcription factors (TFs) upregulated in stem-like compared to terminally exhausted CD8+ T cells (red) and TFs with motif enriched in the differentially accessible chromatin regions between stem-like and terminally exhausted CD8+ T cells (blue). Statistical significance in f was calculated with a two-sided Student’s t-test. ****P < 0.0001.

Extended Data Fig. 2 BACH2 overexpression enhances stem-like CD8+ T cell differentiation during chronic LCMV infection.

a, Representative FACS plots of TCF-1 and TIM-3 expression in pMIG and BACH2 OE P14 CD8+ T cells on day 28 p.i. b, Left panel: representative FACS plots of pMIG and BACH2 OE P14 cells in CD8+ T cells on day 14 and day 28 p.i.. Right panel: fold changes in the numbers of BACH2 OE P14 cells relative to the numbers of pMIG P14 cells on day 14 and day 28 p.i.. n = 5 mice/group. c-e, FACS analyses of PD-1 (c, n = 5 mice/group), TIM-3 (d, n = 5 mice/group), TIGIT (e, n = 5 mice/group) expression in pMIG and BACH2 OE P14 CD8+ T cells on day 14 p.i.. f-h, FACS analyses of PD-1 (f, n = 5 mice/group), TIM-3 (g, n = 5 mice/group), TIGIT (h, n = 5 mice/group) expression in pMIG and BACH2 OE P14 CD8+ T cells on day 28 p.i.. i,j, FACS analyses of KLRG1 expression in pMIG and BACH2 OE P14 CD8+ T cells on day 14 (i, n = 5 mice/group) and day 28 (j, n = 5 mice/group) p.i.. k,l, FACS analyses of EOMES expression in pMIG and BACH2 OE P14 CD8+ T cells on day 14 (k, n = 5 mice/group) and day 28 (l, n = 5 mice/group) p.i.. m-o, FACS analyses of granzyme B (m, n = 5 mice/group), IFNγ (n, n = 5 mice/group), TNFα (o, n = 5 mice/group) expression in pMIG and BACH2 OE P14 CD8+ T cells on day 7 p.i.. p, Numbers of pMIG (n = 4 mice) and BACH2 OE (n = 5 mice) P14 CD8+ T cells in the liver (left) and the ratio of P14 cells in the liver versus P14 cells in the spleen in pMIG (n = 4 mice) and BACH2 OE (n = 5 mice) groups (right) on day 7 p.i.. q, Numbers of pMIG (n = 4 mice) and BACH2 OE (n = 5 mice) P14 CD8+ T cells in the lung (left) and the ratio of P14 cells in the lung versus P14 cells in spleen in pMIG (n = 4 mice) and BACH2 OE (n = 5 mice) groups (right) on day 7 p.i.. r,s, Representative FACS plots (left) and bar graph (right) showing the percentage of stem-like (TCF-1hiTIM-3lo) pMIG (n = 4 mice) and BACH2 OE (n = 5 mice) P14 CD8+ T cells in the liver (r) and lung (s). Data are representative of at least two independent experiments. Circles represent individual mice. Bar graphs represent mean ± s.d.. Statistical significance was calculated with a two-sided Student’s t-test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 3 ScRNA-Seq analyses of pMIG and BACH2 OE P14 cells.

a, Violin plots illustrating the mRNA amounts of differentially expressed genes between pMIG (iris blue) and BACH2 OE (red) P14 cells on day 7 p.i.. b, Left panel: UMAP projection of P14 cells colored by cell-cycle phases (G1: red; S: blue; G2/M: green). Middle panel: percentages of cells in different cell-cycle phases from each cluster defined in Fig. 3c. Right panel: percentages of cells in cluster 0 pMIG P14 cells or cluster 0 BACH2 OE P14 cells that are in different cell-cycle phases. c, Volcano plots showing differentially expressed genes between cluster 0 pMIG P14 cells and cluster 0 BACH2 OE P14 cells.

Extended Data Fig. 4 RNA-Seq analyses of pMIG and BACH2 OE stem-like P14 CD8+ T cells.

a, Correlation heatmap of RNA-Seq data from independent samples (n = 3/group) of pMIG and BACH2 OE stem-like (Ly108hiTIM-3lo) P14 cells in Fig. 4a. b, Significantly enriched pathways in Fig. 4c determined by GSEA. c,d, FACS analyses of phospho-ribosomal protein S6 (c) and phospho-AKT (d) in pMIG (n = 5 mice) and BACH2 OE (n = 4 mice) P14 cells on day 7 p.i.. e, Percentages of dead (Annexin V+ Aqua Live/Dead+) cells among pMIG (n = 4 mice) and BACH2 OE (n = 5 mice) P14 cells on day 7 p.i.. Data in c-e are representative of at least two independent experiments. Circles represent individual mice. Bar graphs represent mean ± s.d.. Statistical significance in c-e was calculated with a two-sided Student’s t-test. **P < 0.01, ***P < 0.001.

Extended Data Fig. 5 BACH2 deficiency impairs the differentiation of stem-like CD8+ T cells and persistence of antiviral CD8+ T cells during chronic LCMV infection.

Bach2loxP/loxP; Cre/ERT2 CD8+ T cells were transduced with P14 TCR and adoptively transferred to B6 CD45.1 mice. Recipients were treated with vehicle (control) or tamoxifen (Bach2 iKO) and infected with LCMV clone 13. a-f, Splenocytes were analyzed on day 7 post-infection. a, Representative FACS plots (left) and bar graph (right) showing the percentage of stem-like (TCF-1hiTIM-3lo) control (n = 5 mice) and Bach2 iKO (n = 4 mice) P14 cells. b,c, Numbers of stem-like (TCF-1hi) (b) and terminally exhausted (TCF-1lo) (c) control (n = 5 mice) and Bach2 iKO (n = 4 mice) P14 cells. d-f, FACS analysis of granzyme B (d), IFNγ (e), TNFα (f) in control (n = 5 mice) and Bach2 iKO (n = 4 mice) P14 cells. g-i, Splenocytes were analyzed two-week post-infection. g, Representative FACS plots (left) and bar graph (right) showing the percentage of stem-like (TCF-1hiTIM-3lo) control (n = 5 mice) and Bach2 iKO (n = 4 mice) P14 cells. h,i, Numbers of stem-like (TCF-1hi) (h) and terminally exhausted (TCF-1lo) (i) control (n = 5 mice) and Bach2 iKO (n = 4 mice) P14 cells. j-l, Splenocytes were analyzed one-month post-infection. j, Bar graph showing the percentage of stem-like control (n = 5 mice) and Bach2 iKO (n = 5 mice) P14 cells. k,l, Numbers of stem-like (k) and terminally exhausted (l) control (n = 5 mice) and Bach2 iKO (n = 5 mice) P14 cells. Data are representative of at least two independent experiments. Circles represent individual mice. Bar graphs represent mean ± s.d.. Statistical significance was calculated with a two-sided Student’s t-test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Extended Data Fig. 6 Transcriptome analysis of control and Bach2 gRNA transduced P14 cells.

a, Violin plots illustrating the mRNA amounts of differentially expressed genes between control (iris blue) and Bach2 gRNA (purple) transduced P14 cells on day 7 p.i.. Each dot represents one cell. b, Correlation between WT GP33 tetramer+ stem-like CD8+ T cells and control stem-like P14 CD8+ T cells (left) or between Bach2 KO GP33 tetramer+ stem-like CD8+ T cells and Bach2 gRNA stem-like P14 CD8+ T cells (right). 3 independent samples per condition. c, GSEA of RNA-Seq data from WT and Bach2 KO GP33 tetramer+ stem-like CD8+ T cells shows the enrichment of gene sets containing genes upregulated (left) or downregulated (right) in Bach2 gRNA transduced stem-like P14 cells relative to control stem-like P14 cells. d, Significantly enriched pathways in Fig. 6h determined by GSEA.

Extended Data Fig. 7 The molecular program downstream of BACH2.

a, Experimental setup has been described in Fig. 7a. A pie chart illustrates the genomic distribution of differentially accessible (DA) regions between pMIG and BACH2 OE stem-like P14 cells. b, Experimental setup has been described in Fig. 7f. A pie chart illustrates the genomic distribution of DA regions between control and Bach2 gRNA transduced stem-like P14 cells. c,d, Cas9; P14 CD8+ T cells co-transduced with pMKO GFP vector expressing Bach2 gRNA and SL21 VEX vector expressing Runx3 gRNA were adoptively transferred into C57BL/6 mice that were subsequently infected with LCMV clone 13 (n = 4 mice). Splenic P14 cells were analyzed on day 7 p.i.. c, Representative FACS plots of Ly108 and TIM-3 expression on GFP-VEX-, GFP+VEX-, GFP-VEX+, and GFP+VEX+ P14 CD8+ T cells. d, Percentage of stem-like (Ly108hiTIM-3lo) cells within GFP-VEX-, GFP+VEX-, GFP-VEX+, and GFP+VEX+ P14 CD8+ T cells. e-g, Cas9; P14 CD8+ T cells co-transduced with pMKO GFP vector expressing Bach2 gRNA and SL21 VEX vector expressing Prdm1 gRNA were adoptively transferred into C57BL/6 mice that were then infected with LCMV clone 13 (n = 5 mice). Splenic P14 cells were analyzed on day 7 p.i.. Representative FACS plots of Ly108 and TIM-3 expression on GFP-VEX-, GFP+VEX-, GFP-VEX+, and GFP+VEX+ P14 CD8+ T cells (e) and percentage of stem-like (Ly108hiTIM-3lo) cells within GFP-VEX-, GFP+VEX-, GFP-VEX+, and GFP+VEX+ P14 cells (f) are shown. g, CD62L expression on GFP-VEX-, GFP+VEX-, GFP-VEX+, and GFP+VEX+ P14 cells. Data in c-g are representative of at least two independent experiments. Circles represent individual mice. Lines in d,f,g connect data points from the same individual mice. Statistical significance in d,f,g was calculated with a two-sided Student’s paired t-test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yao, C., Lou, G., Sun, HW. et al. BACH2 enforces the transcriptional and epigenetic programs of stem-like CD8+ T cells. Nat Immunol 22, 370–380 (2021). https://doi.org/10.1038/s41590-021-00868-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00868-7

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