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:

BATF3 programs CD8+ T cell memory

Abstract

Antiviral CD8+ T cell responses are characterized by an initial activation/priming of T lymphocytes followed by a massive proliferation, subset differentiation, population contraction and the development of a stable memory pool. The transcription factor BATF3 has been shown to play a central role in the development of conventional dendritic cells, which in turn are critical for optimal priming of CD8+ T cells. Here we show that BATF3 was expressed transiently within the first days after T cell priming and had long-lasting T cell–intrinsic effects. T cells that lacked Batf3 showed normal expansion and differentiation, yet succumbed to an aggravated contraction and had a diminished memory response. Vice versa, BATF3 overexpression in CD8+ T cells promoted their survival and transition to memory. Mechanistically, BATF3 regulated T cell apoptosis and longevity via the proapoptotic factor BIM. By programing CD8+ T cell survival and memory, BATF3 is a promising molecule to optimize adoptive T cell therapy in patients.

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: BATF3 has a CD8+ T cell–intrinsic function.
Fig. 2: BATF3 regulates memory transition and survival of CD8+ T cells.
Fig. 3: The lack of BATF3 impacts CD8+ T cell fitness.
Fig. 4: BATF3 regulates CD8+ T cell survival via the BIM.
Fig. 5: BATF3 gain-of-function promotes CD8+ T cell survival in vitro and in vivo.
Fig. 6: BATF3 gain-of-function promotes CD8+ T cell fitness.
Fig. 7: BATF3 is a potential target for immunotherapy in humans.

Similar content being viewed by others

Data availability

The sequencing data are available at NCBI Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo) under accession numbers GSE143504 and GSE153341. The data that support the findings of this study are available from the corresponding authors upon request.

References

  1. Kaech, S. M. & Cui, W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bedoui, S., Heath, W. R. & Mueller, S. N. CD4+ T-cell help amplifies innate signals for primary CD8+ T-cell immunity. Immunol. Rev. 272, 52–64 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Eickhoff, S. et al. Robust anti-viral immunity requires multiple distinct T cell–dendritic cell interactions. Cell 162, 1322–1337 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hor, J. L. et al. Spatiotemporally distinct interactions with dendritic cell subsets facilitates CD4+ and CD8+ T cell activation to localized viral infection. Immunity 43, 554–565 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Shaulian, E. & Karin, M. AP-1 as a regulator of cell life and death. Nat. Cell Biol. 4, E131–E136 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Karin, M., Liu, Z. & Zandi, E. AP-1 function and regulation. Curr. Opin. Cell Biol. 9, 240–246 (1997).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. 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 

  9. Iacobelli, M., Wachsman, W. & McGuire, K. L. Repression of IL-2 promoter activity by the novel basic leucine zipper p21SNFT protein. J. Immunol. 165, 860–868 (2000).

    Article  CAS  PubMed  Google Scholar 

  10. Grajales-Reyes, G. E. et al. Batf3 maintains autoactivation of Irf8 for commitment of a CD8α+ conventional DC clonogenic progenitor. Nat. Immunol. 16, 708–717 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Schlitzer, A. et al. Identification of cDC1- and cDC2-committed DC progenitors reveals early lineage priming at the common DC progenitor stage in the bone marrow. Nat. Immunol. 16, 718–728 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Edelson, B. T. et al. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8α+ conventional dendritic cells. J. Exp. Med. 207, 823–836 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. Karwacz, K. et al. Critical role of IRF1 and BATF in forming chromatin landscape during type 1 regulatory cell differentiation. Nat. Immunol. 18, 412–421 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Punkenburg, E. et al. Batf-dependent Th17 cells critically regulate IL-23 driven colitis-associated colon cancer. Gut 65, 1139–1150 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Schraml, B. U. et al. The AP-1 transcription factor Batf controls TH17 differentiation. Nature 460, 405–409 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kuroda, S. et al. Basic leucine zipper transcription factor, ATF-like (BATF) regulates epigenetically and energetically effector CD8 T-cell differentiation via Sirt1 expression. Proc. Natl Acad. Sci. USA 108, 14885–14889 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Quigley, M. et al. Transcriptional analysis of HIV-specific CD8+ T cells shows that PD-1 inhibits T cell function by upregulating BATF. Nat. Med. 16, 1147–1151 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Logan, M. R., Jordan-Williams, K. L., Poston, S., Liao, J. & Taparowsky, E. J. Overexpression of Batf induces an apoptotic defect and an associated lymphoproliferative disorder in mice. Cell Death Dis. 3, e310 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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.e1125 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Tscharke, D. C. et al. Identification of poxvirus CD8+ T cell determinants to enable rational design and characterization of smallpox vaccines. J. Exp. Med. 201, 95–104 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gerlach, C. et al. The chemokine receptor CX3CR1 defines three antigen-experienced CD8+ T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity 45, 1270–1284 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bottcher, J. P. et al. Functional classification of memory CD8+ T cells by CX3CR1 expression. Nat. Commun. 6, 8306 (2015).

    Article  PubMed  CAS  Google Scholar 

  26. Yajima, T. et al. IL-15 regulates CD8+ T cell contraction during primary infection. J. Immunol. 176, 507–515 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Rubinstein, M. P. et al. IL-7 and IL-15 differentially regulate CD8+ T-cell subsets during contraction of the immune response. Blood 112, 3704–3712 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. van der Windt, G. J. et al. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36, 68–78 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Pearce, E. L. et al. Enhancing CD8+ T-cell memory by modulating fatty acid metabolism. Nature 460, 103–107 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. O’Neill, L. A., Kishton, R. J. & Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 16, 553–565 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Yamazaki, C. et al. Critical roles of a dendritic cell subset expressing a chemokine receptor, XCR1. J. Immunol. 190, 6071–6082 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Alexandre, Y. O. et al. XCR1+ dendritic cells promote memory CD8+ T cell recall upon secondary infections with Listeria monocytogenes or certain viruses. J. Exp. Med. 213, 75–92 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mattiuz, R. et al. Novel Cre-expressing mouse strains permitting to selectively track and edit type 1 conventional dendritic cells facilitate disentangling their complexity in vivo. Front Immunol. 9, 2805 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kitano, M. et al. Imaging of the cross-presenting dendritic cell subsets in the skin-draining lymph node. Proc. Natl Acad. Sci. USA 113, 1044–1049 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. D’Cruz, L. M., Rubinstein, M. P. & Goldrath, A. W. Surviving the crash: transitioning from effector to memory CD8+ T cell. Semin. Immunol. 21, 92–98 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Pellegrini, M., Belz, G., Bouillet, P. & Strasser, A. Shutdown of an acute T cell immune response to viral infection is mediated by the proapoptotic Bcl-2 homology 3-only protein BIM. PNAS 100, 14175–14180 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hildeman, D. A. et al. Activated T cell death in vivo mediated by proapoptotic Bcl-2 family member BIM. Immunity 16, 759–767 (2002).

    Article  CAS  PubMed  Google Scholar 

  38. Tussiwand, R. et al. Compensatory dendritic cell development mediated by BATF–IRF interactions. Nature 490, 502–507 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sionov, R. V., Vlahopoulos, S. A. & Granot, Z. Regulation of BIM in health and disease. Oncotarget 6, 23058–23134 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Hajji, N. & Joseph, B. Epigenetic regulation of cell life and death decisions and deregulation in cancer. Essays Biochem. 48, 121–146 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Shaulian, E. & Karin, M. AP-1 in cell proliferation and survival. Oncogene 20, 2390–2400 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lopez-Bergami, P., Lau, E. & Ronai, Z. Emerging roles of ATF2 and the dynamic AP1 network in cancer. Nat. Rev. Cancer 10, 65–76 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Weiser, C. et al. Ectopic expression of transcription factor BATF3 induces B-cell lymphomas in a murine B-cell transplantation model. Oncotarget 9, 15942–15951 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Jiang, H., Lei, R., Ding, S. W. & Zhu, S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinf. 15, 182 (2014).

    Article  Google Scholar 

  47. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Liao, Y., Smyth, G. K. & Shi, W. The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads. Nucleic Acids Res. 47, e47 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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  Google Scholar 

  50. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinf. 14, 128 (2013).

    Article  Google Scholar 

  52. Huang, R. et al. TheNCATS BioPlanet: an integrated platform for exploring the universe of cellular signaling pathways for toxicology, systems biology, and chemical genomics. Front. Pharmacol. 10, 445 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Core Unit for FACS and the Core Unit SysMed of the IZKF Würzburg for supporting this study, H.C. Probst (Institute for Immunology, Mainz University) and T. Kaisho (Immunology Frontier Research Center, Osaka University) for providing the P14 and XCR1-DTR-Venus mice31, respectively. The B8R tetramer was obtained through the National Institutes of Health Tetramer Core Facility. This work was supported by grants through the German Research Foundation, SFB TR 221 (project A03 to M. Hudecek and H.E.), Germany’s Excellence Strategy – EXC2151 – (390873048 to M. Hölzel) and German Research Foundation Emmy Noether programme (GA2129/2-1 to G.G.) and by grants of the European Research Council to W.K. (819329- STEP2) and G.G. (759176-TissueLymphoContexts). W.K., G.G. and M.V. are supported by the by the Max Planck Society (Max Planck Research Groups).

Author information

Authors and Affiliations

Authors

Contributions

M.A.A., K. Komander, M.V. and W.K. conceptualized the study and analyzed data. M.A.A., K. Komander, K. Knöpper, A.E.P., H.W., S.E., T.G, J.W. and A.G. planned and performed experiments and analyzed the data. K. Knöpper and M. Hölzel analyzed transcriptome data. A.E.P. generated critical reagents. M. Hudecek provided critical reagents. A.K., N.G., H.E., M. Hudecek, G.G., M. Hölzel provided intellectual input and gave conceptual advice. M.A.A. and W.K. wrote the manuscript with input from all authors. W.K. and G.G. provided research funds.

Corresponding authors

Correspondence to Marco A. Ataide or Wolfgang Kastenmüller.

Ethics declarations

Competing interests

M.A.A., M. Hudecek and W.K. are currently applying for patents relating to the content of this manuscript.

Additional information

Peer review information 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 Batf3–/– and WT leukocyte chimerism.

Relative abundance of leukocytes after prime (d8; VV) and recall response (d6; VV) of bone marrow chimeric mice. Data are representative of two (n = 9 at prime and n = 7 at recall) independent experiments. Error bars indicate the mean ± SEM. Comparison between groups was calculated using two-tailed unpaired Student’s t-test.

Extended Data Fig. 2 OT-I CD8+ T cell immune response during L.m. OVA infection.

Kinetic analysis of co-transferred WT (GFP) and Batf3−/− (CD45.1) OT-I cells in the blood of WT recipients after L.m.-OVA i.p. Data are representative of three independent experiments (n = 4). Error bars indicate the mean ± SEM. Comparison between groups was calculated using the One sample t-test. ***, p = 0.0003.

Extended Data Fig. 3 Measurement of mitochondrial potential during the contraction phase of CD8+ T cell immune response.

Representative dotplots, histograms and statistical analysis of mitochondrial membrane potential of the co-transferred WT (GFP) and Batf3−/− (CD45.1) OT-I cells. Cells were analyzed at d8 and d12 post- VV-OVA infection. Data are representative of two independent experiments (n = 4). Error bars indicate the mean ± SEM. Comparison between groups was calculated using the paired Student’s t-test. * = p ≤ 0.05.

Extended Data Fig. 4 BATF3 programs T cell survival via the proapoptotic factor BIM.

(a) Kinetic analysis of co-transferred WT (CD45.1 / CD45.2) and Batf3−/− (CD45.1) OT-I cells in the blood of IL15 deficient (IL15-/-) recipients after or VV-OVA i.p. infection. (b) qRT-PCR of Batf in ex vivo isolated WT and Batf3−/− CD8 OT-I cells after VV-OVA infection, or 48 h hours post anti-CD3/CD28 in vitro stimulation. (c) Heatmap showing the 166 differentially expressed genes by comparing WT versus Batf3−/− CD8 T cells in vitro stimulated with anti-CD3/CD28 for 48 h or 72 h. Columns represent technical replicates. (d) Sequence and schematic of the retroviral plasmid carrying the Bcl2l11 shRNA. pLMPd-shRNA-Ametrine were provided by Transomic®. The shRNA position is marked in red and reported by Ametrine (yellow). The cassette carrying the shRNA is flanked by LTRs (dark grey) and MESV Psi (light grey). Ametrine (yellow) is active by a PGK promoter (light grey). LTR: long terminal repeat; MESV: murine embryonic stem cell virus; Psi: RNA target site for packaging; PGK: PGK promoter. Data are representative of one experiment (n = 4). Error bars indicate the mean ± SEM. Comparison between groups (n = 4) was calculated using the One sample t-test. **, p = 0.007.

Extended Data Fig. 5 Schematics of the retroviral vectors.

For both retroviral over-expression plasmids pMIG was used as a backbone. In both cases the inserts, Batf3-IRES-GFP (dark blue and green, respectively) and Batf-IRES-GFP (light blue and green, respectively), were inserted between the LTRs (dark grey) and downstream of MESV Psi (light grey) and gag (grey-blue). As mock control pMIGR1-Ametrine was used. LTR: long terminal repeat; gag: structural precursor protein for packaging; MESV: murine embryonic stem cell virus; Psi: RNA target site for packaging.

Extended Data Fig. 6 BATF3-, but not BATF- overexpression promotes CD8+ T cell survival in vitro.

(a, b) Naïve CD8 T cells were isolated, activated and retrovirally transduced with pMIG- Ametrine (empty vector) or carrying the murine Batf3, pMIG-BATF3-GFP (BATF3) or the murine Batf, pMIG-BATF-GFP (BATF). Cells were resting for 48 h in culture after transduction and for additional 48 h after sorting before in vitro activation with anti-CD3/CD28. (a) Representative dotplots and statistical analysis of the in vitro kinetics of WT CD8 T cells coculture (50:50 – empty vector: BATF3) and (b) 50:50 – empty vector: BATF) in two different concentrations of IL-15. (c) Representative dotplots and statistical analysis of the in vitro kinetics of WT versus Batf3-/- CD8 T cells coculture (50:50 – empty vector: empty vector or empty vector: BATF3). Data are representative of one (a; n = 4) or two (b, c; n = 4) independent experiments. Error bars indicate the mean ± SEM. Comparison between groups was calculated using the One sample t-test. **, p = 0.007.

Extended Data Fig. 7 BATF3 overexpression programs CD8+ T cell stemness.

(a-d) Representative dotplots and gating strategy for (a) IL-7R, (b) ICOS, (c) intracellular CTLA-4 and (d) Tim-3 expression analysis in resting and after anti-CD3/CD28 stimulation of 50:50 (empty vector: BATF3) CD8+ T cell coculture. (e) Statistical analysis of Glycolitic Proton Eflux Rate (GlycoPER) readouts and (f) Oxygen Consuption Rate (OCR) parameters measured in resting state and 24 h after anti-CD3/CD28 stimulation. (a-f) Data are representative of two independent experiments (n = 3). Error bars indicate the mean ± SEM. Comparison between groups was calculated using two-tailed unpaired Student’s t-test.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ataide, M.A., Komander, K., Knöpper, K. et al. BATF3 programs CD8+ T cell memory. Nat Immunol 21, 1397–1407 (2020). https://doi.org/10.1038/s41590-020-0786-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0786-2

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