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:

Tcf1 preprograms the mobilization of glycolysis in central memory CD8+ T cells during recall responses

A Publisher Correction to this article was published on 12 April 2022

This article has been updated

Abstract

The mechanisms underlying the heightened protection mediated by central memory CD8+ T (TCM) cells remain unclear. Here we show that the transcription factor Tcf1 was required in resting TCM cells to generate secondary effector CD8+ T cells and to clear pathogens during recall responses. Recall stimulation of CD8+ TCM cells caused extensive reprogramming of the transcriptome and chromatin accessibility, leading to rapid induction of glycolytic enzymes, cell cycle regulators and transcriptional regulators, including Id3. This cluster of genes did not require Tcf1 in resting CD8+ TCM cells, but depended on Tcf1 for optimal induction and chromatin opening in recall-stimulated CD8+ TCM cells. Tcf1 bound extensively to these recall-induced gene loci in resting CD8+ TCM cells and mediated chromatin interactions that positioned these genes in architectural proximity with poised enhancers. Thus, Tcf1 preprogramed a transcriptional program that supported the bioenergetic and proliferative needs of CD8+ TCM cells in case of a secondary challenge.

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: Tcf1 and Lef1 are essential for generating CD8+ memory precursors and TCM cells.
Fig. 2: Tcf1 is critical for secondary expansion of CD8+ TCM cells in recall responses.
Fig. 3: Tcf1 is required for recall-induced transcriptomic changes in CD8+ TCM cells.
Fig. 4: Tcf1 is necessary for recall-induced open chromatin sites in CD8+ TCM cells.
Fig. 5: Integrative analysis of recall-induced genes by Tcf1 occupancy and chromatin accessibility.
Fig. 6: Tcf1 mediates chromatin interactions to control CD8+ TCM gene expression at resting and recall-stimulated states.
Fig. 7: Tcf1 preprograms recall-induced activation of glycolysis in CD8+ TCM cells.
Fig. 8: Tcf1 acts upstream of Id3 induction to promote CD8+ TCM recall response.

Similar content being viewed by others

Data availability

ChIP-seq, RNA-seq, ATAC-seq and Hi-C data in CD8+ TCM cells are deposited at GEO under accession no. GSE177064.

Change history

References

  1. Martin, M. D. & Badovinac, V. P. Defining memory CD8+ T cell. Front. Immunol. 9, 2692 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Williams, M. A. & Bevan, M. J. Effector and memory CTL differentiation. Annu. Rev. Immunol. 25, 171–192 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Jameson, S. C. & Masopust, D. Understanding subset diversity in T cell memory. Immunity 48, 214–226 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Surh, C. D. & Sprent, J. Homeostasis of naive and memory T cells. Immunity 29, 848–862 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Banerjee, A. et al. Cutting edge: the transcription factor eomesodermin enables CD8+ T cells to compete for the memory cell niche. J. Immunol. 185, 4988–4992 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Zhou, X. et al. Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity 33, 229–240 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kim, M. V., Ouyang, W., Liao, W., Zhang, M. Q. & Li, M. O. The transcription factor Foxo1 controls central-memory CD8+ T cell responses to infection. Immunity 39, 286–297 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. 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 

  9. Laidlaw, B. J., Craft, J. E. & Kaech, S. M. The multifaceted role of CD4(+) T cells in CD8(+) T cell memory. Nat. Rev. Immunol. 16, 102–111 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jeannet, G. et al. Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8+ T cell memory. Proc. Natl Acad. Sci. USA 107, 9777–9782 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hess Michelini, R., Doedens, A. L., Goldrath, A. W. & Hedrick, S. M. Differentiation of CD8+ memory T cells depends on Foxo1. J. Exp. Med. 210, 1189–1200 (2013).

    Article  PubMed  Google Scholar 

  12. Cui, W., Liu, Y., Weinstein, J. S., Craft, J. & Kaech, S. M. An interleukin-21–interleukin-10–STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity 35, 792–805 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Guan, T. et al. ZEB1, ZEB2, and the miR-200 family form a counterregulatory network to regulate CD8(+) T cell fates. J. Exp. Med. 215, 1153–1168 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ichii, H., Sakamoto, A., Kuroda, Y. & Tokuhisa, T. Bcl6 acts as an amplifier for the generation and proliferative capacity of central memory CD8+ T cells. J. Immunol. 173, 883–891 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Ji, Y. et al. Repression of the DNA-binding inhibitor Id3 by Blimp-1 limits the formation of memory CD8+ T cells. Nat. Immunol. 12, 1230–1237 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhao, X., Shan, Q. & Xue, H. H. TCF1 in T cell immunity: a broadened frontier. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-021-00563-6 (2021).

  18. Raghu, D., Xue, H. H. & Mielke, L. A. Control of lymphocyte fate, infection, and tumor immunity by TCF-1. Trends Immunol. 40, 1149–1162 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Gullicksrud, J. A., Shan, Q. & Xue, H. H. Tcf1 at the crossroads of CD4+ and CD8+ T cell identity. Front. Biol. 12, 83–93 (2017).

    Article  CAS  Google Scholar 

  20. Zhao, D. M. et al. Constitutive activation of Wnt signaling favors generation of memory CD8 T cells. J. Immunol. 184, 1191–1199 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. He, B. et al. CD8(+) T cells utilize highly dynamic enhancer repertoires and regulatory circuitry in response to infections. Immunity 45, 1341–1354 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gullicksrud, J. A. et al. Differential requirements for Tcf1 long isoforms in CD8(+) and CD4(+) T cell responses to acute viral infection. J. Immunol. 199, 911–919 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Jameson, S. C. & Masopust, D. Diversity in T cell memory: an embarrassment of riches. Immunity 31, 859–871 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Martin, M. D. et al. Phenotypic and functional alterations in circulating memory CD8+ T cells with time after primary infection. PLoS Pathog. 11, e1005219 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Miyagawa, F. et al. Interferon regulatory factor 8 integrates T-cell receptor and cytokine-signaling pathways and drives effector differentiation of CD8+ T cells. Proc. Natl Acad. Sci. USA 109, 12123–12128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yang, C. Y. et al. The transcriptional regulators Id2 and Id3 control the formation of distinct memory CD8+ T cell subsets. Nat. Immunol. 12, 1221–1229 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. He, S. et al. Ezh2 phosphorylation state determines its capacity to maintain CD8(+) T memory precursors for antitumor immunity. Nat. Commun. 8, 2125 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Shan, Q. et al. Tcf1 and Lef1 provide constant supervision to mature CD8(+) T cell identity and function by organizing genomic architecture. Nat. Commun. 12, 5863 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim, B. H. et al. Benzoxathiole derivative blocks lipopolysaccharide-induced nuclear factor-κB activation and nuclear factor-κB-regulated gene transcription through inactivating inhibitory κB kinase β. Mol. Pharmacol. 73, 1309–1318 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Verma, V. et al. MEK inhibition reprograms CD8(+) T lymphocytes into memory stem cells with potent antitumor effects. Nat. Immunol. 22, 53–66 (2021).

    Article  CAS  PubMed  Google Scholar 

  31. 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 

  32. Grosschedl, R., Giese, K. & Pagel, J. HMG domain proteins: architectural elements in the assembly of nucleoprotein structures. Trends Genet. 10, 94–100 (1994).

    Article  CAS  PubMed  Google Scholar 

  33. Love, J. J. et al. Structural basis for DNA bending by the architectural transcription factor LEF-1. Nature 376, 791–795 (1995).

    Article  CAS  PubMed  Google Scholar 

  34. Lun, A. T. & Smyth, G. K. diffHic: a Bioconductor package to detect differential genomic interactions in Hi-C data. BMC Bioinformatics 16, 258 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Pearce, E. L., Poffenberger, M. C., Chang, C. H. & Jones, R. G. Fueling immunity: insights into metabolism and lymphocyte function. Science 342, 1242454 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  36. 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  Google Scholar 

  37. Pais Ferreira, D. et al. Central memory CD8(+) T cells derive from stem-like Tcf7(hi) effector cells in the absence of cytotoxic differentiation. Immunity 53, 985–1000 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. 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  PubMed  Google Scholar 

  39. Danilo, M., Chennupati, V., Silva, J. G., Siegert, S. & Held, W. Suppression of Tcf1 by inflammatory cytokines facilitates effector CD8 T cell differentiation. Cell Rep. 22, 2107–2117 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Yu, S. et al. The TCF-1 and LEF-1 transcription factors have cooperative and opposing roles in T cell development and malignancy. Immunity 37, 813–826 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Steinke, F. C. et al. TCF-1 and LEF-1 act upstream of Th-POK to promote the CD4(+) T cell fate and interact with Runx3 to silence Cd4 in CD8(+) T cells. Nat. Immunol. 15, 646–656 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Shan, Q. et al. Ectopic Tcf1 expression instills a stem-like program in exhausted CD8(+) T cells to enhance viral and tumor immunity. Cell. Mol. Immunol. 18, 1262–1277 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pertea, M., Kim, D., Pertea, G. M., Leek, J. T. & Salzberg, S. L. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protoc. 11, 1650–1667 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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 

  47. Zhu, A., Ibrahim, J. G. & Love, M. I. Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences. Bioinformatics 35, 2084–2092 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. 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 

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

    Article  PubMed  PubMed Central  Google Scholar 

  50. 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 

  51. Kulakovskiy, I. V. et al. HOCOMOCO: towards a complete collection of transcription factor binding models for human and mouse via large-scale ChIP-Sseq analysis. Nucleic Acids Res. 46, D252–D259 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. 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 

  53. Nora, E. P. et al. Molecular basis of CTCF binding polarity in genome folding. Nat. Commun. 11, 5612 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Alavattam, K. G. et al. Attenuated chromatin compartmentalization in meiosis and its maturation in sperm development. Nat. Struct. Mol. Biol. 26, 175–184 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Durand, N. C. et al. Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments. Cell Syst. 3, 95–98 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wang., Z., Zhang, Y. & Zang, C. BART3D: inferring transcriptional regulators associated with differential chromatin interactions from Hi-C data. Bioinformatics 37, 3075–3078 (2021).

    Article  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the University of Iowa Flow Cytometry Core facility (J. Fishbaugh, H. Vignes and G. Rasmussen) and the HMH-CDI Flow Cytometry Core facility (M. Poulus and W. Tsao) for cell sorting, B. Wagner (University of Iowa Free Radical and Radiation Biology Core) and P. Ramalingam (HMH-CDI) for assistance with the use of Seahorse Extracellular Flux Analyzer, and K. Zhao (NHLBI, NIH) for sharing the Hi-C protocol. This study is supported, in part, by grants from the National Institutes of Health (AI121080 and AI139874 to H.-H.X. and W.P.; AI112579 to H.-H.X. and C.Z.; and GM134880 and AI114543 to V.P.B.).

Author information

Authors and Affiliations

Authors

Contributions

Q.S. and X.C. performed the experiments and analyzed the data. S.S.H. analyzed the ChIP-seq, RNA-seq and ATAC-seq data under supervision of C.Z. S.Z. analyzed the Hi-C data under supervision of W.P. V.P.B. provided key reagents and scientific input. H.H.X. conceived the project and supervised the overall study. H.H.X. and C.Z. wrote the paper and all authors edited the paper.

Corresponding authors

Correspondence to Chongzhi Zang or Hai-Hui Xue.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Double deletion of Tcf1 and Lef1 diminished expression of cytotoxic effector molecules during primary CD8+ T cell responses.

a. Gating stratey to identify CD45.2+ donor-derived P14 CD8+ T cells in CD45.1+ wild-type recipients. b. Validating early deletion of Tcf1 protein by Gzmb-Cre by intracellular stating at 36 hrs after willd-type (WT) or Tcf7ΔGzmb P14 CD8+ T cells were labeled with cell-trace violet (CTV) and adoptively transferred into CD45.1+ recipients followed by i.v. infection with LCMV-Arm the next day. Dot plots are representative from 2 independent experiments, where values denote the numbers of cell division and redline marks signal levels with isotype control staining. c,d. Detection of cytokine production by effector CD8+ T cells in recipient spleens during primary response on day 8 after WT, Tcf7ΔGzmb or Tcf7ΔGzmbLef1ΔGzmb P14 CD8+ T cells were adoptively transferred into CD45.1+ wild-type recipients, followed by LCMV-Arm infection the next day. GP33-induced production of IFN-γ (c), TNF and IL-2 (d) was detected in CD45.2+CD8+ T cells. Representative contour plots (left) are from ≥3 experiments, with values denoting percentages of the gated population. Cumulative data (right) of the percentage of IFN-γ+ population in P14 CD8+ T cells, IFN-γ relative geometric mean fluorescent intensity (gMFI) (c), and the percentage of TNF+ or IL-2+ populations in IFN-γ+ P14 CD8+ T cells (d) are means ± s.d. e. Detection of granzyme B expression in effector CD8+ T cells by intracellular staining on day 8 p.i. Representative histographs (left) are from ≥3 experiments with the values denoting percentage of granzyme B+ cells and those in parentheses denoting gMFI of granzyme B. Cumulative data (left) are means ± s.d. For data in c–e, statistical significance was first determined with one-way ANOVA for multi-group comparisons, and as post hoc correction, Tukey’s test was used for indicated pairwise comparison. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ns, not statistically significant.

Extended Data Fig. 2 Tcf1 is largely dispensable for induction of cytotoxic effector molecules during CD8+ TCM recall response.

a. Detecting cell viability of early effector CD8+ T cells in secondary recipients at 60 hrs after CD45.2+ wild-type or Tcf7ΔGzmb CD62L+CD8+ TCM cells (sorted from the primary recipients on day ≥30–35 p.i., as generated in Fig. 1g) were transferred into CD45.1+ wild-type recipients, which were infected with LCMV-Arm the next day. AnnexinV7-AAD viable cells were detected in CTV+CD45.2+CD8+ T cells, with representative contour plots (left) from 2 independent experiments, and cumulative data (right) as means ± s.d. b, c. Detection of cytokine production in secondary effector CD8+ T cells on day 8 after CD45.2+ wild-type or Tcf7ΔGzmb CD62L+CD8+ TCM cells (sorted from the primary recipients on day ≥30–35 p.i., as generated in Fig. 1g) were transferred into CD45.1+ wild-type recipients, which were infected with LCMV-Arm the next day. GP33-induced production of IFN-γ (b), TNF and IL-2 (c) was detected in CD45.2+CD8+ T cells. Representative contour plots (left) are from 2 independent experiments, with values denoting percentages of the gated population. Cumulative data (right) of the percentage of IFN-γ+ population in P14 CD8+ T cells, IFN-γ relative gMFI (b), and the percentage of TNF+ or IL-2+ populations in IFN-γ+ P14 CD8+ T cells (c) are means ± s.d. d. Detection of granzyme B expression in secondary effector CD8+ T cells by intracellular staining on day 8 p.i. Representative histographs (left) are from 2 independent experiments, with the values denoting percentage of granzyme B+ cells and those in parentheses denoting gMFI of granzyme B. Cumulative data (left) are means ± s.d. ns, not statistically significant; **, p < 0.01 as determined with two-tailed Student’s t-test.

Extended Data Fig. 3 Tcf1 critically regulates transcriptomic changes in CD8+ TCM cells at resting state and after recall stimulation.

a. Principal component analysis (PCA) of the transcriptomes of wild-type or Tcf7ΔGzmb CD8+ TCM cells at resting state and after 24-hr GP33 stimulation, where log2(RPKM + 1) for each gene in different samples were used as the input data. b,d,f. Heatmaps of select genes involved in ‘mitochondria’ and ‘oxidative phosphorylation’ from Cluster 3 + 4a (b), genes in Cluster 1 (d) and Cluster 2 (f), with the color represented row-normalized RPM for the genes across different samples. c,e. Functional annotation of Cluster 1 (c) and Cluster 2 (e) genes, as defined in Fig. 3e, using DAVID Bioinformatic Resources, with select top gene ontology (GO) terms shown in dot plots and statistical significance denoted with a color scale.

Extended Data Fig. 4 Tcf1 critically regulates chromatin accessibility changes in CD8+ TCM cells at resting state and after recall stimulation.

a. Principal component analysis (PCA) of the ChrAcc profiles of wild-type or Tcf7ΔGzmb CD8+ TCM cells at resting state and after 24-hr GP33 stimulation, where normalized ATAC-seq signal on merged peaks were used as input data. b. Functional annotation of recall-‘closed’ chromatin sites in wild-type CD8+ TCM cells (the 1,484 sites in Fig. 4d) using GREAT analysis, with select top GO terms shown in dot plots and statistical significance denoted with a color scale. c,d. Motif analysis of recall-‘opened’ 17,763 (c) and –‘closed’ 1,484 ChrAcc sites (d) in GP33-stimulated WT CD8+ TCM cells, as defined in Fig. 4d. Listed are the top transcription factor or hybrid motifs and corresponding logos, along with statistical significance and frequency of occurrence at the target ChrAcc sites.

Extended Data Fig. 5 Tcf1 critically regulates chromatin interactions in resting CD8+ TCM cells.

a. Scatter-plots showing reproducibility of two biological replicates of Hi-C libraries from resting wild-type or Tcf7ΔGzmb CD8+ TCM cells. The x- and y-axis values for each data point (marked with a dot) represent the interaction scores of an anchor in replicate 1 (Rep1) and replicate 2 (Rep2), respectively, and the R values denote Pearson correlation coefficient. b. Functional annotation of Clusters 3 + 4a genes associated with Tcf1-dependent ChrInt using DAVID, with selected gene ontology (GO) terms shown in dot plots and statistical significance denoted with a color scale. c. Proposed model for Tcf1-mediated pre-programming of responsiveness of CD8+ TCM cells to recall stimulation. Left, CD8+ TCM cells at resting state express Tcf1 and basal expression of secondary effector CD8+ T cell-associated genes, including transcription regulators such as Id3 and glycolytic enzymes. In this context, the presence of Tcf1 mediates chromatin interactions that engage the effector gene promoters with distal regulatory regions (top row) or span the effector gene loci (bottom row). The Tcf1-dependent chromatin interactions may thus bring the effector gene promotors and the poised enhancer elements (blue flags) into spatial proximity, which constitutes a ‘preprogrammed’ state predisposed for transcriptional activation. Right, recall-stimulated CD8+ TCM cells downregulate Tcf1, but TCR-mobilized transcription factors, such as those in the AP-1, NFAT and NF-κB families, increase chromatin accessibility of the poised regulatory elements to become active enhancers (red flags), and hence induce the poised effector genes with higher efficacy. The induced Id3 can further boost activation of glycolytic genes in recall-stimulated CD8+ TCM cells to ensure bioenergetic supplies.

Extended Data Fig. 6 Integrative analysis of Tcf1 occupancy and chromatin accessibility at recall-induced genes in CD8+ TCM cells.

Sequencing tracks at select glycolytic genes (a) and Irf8 (b) as displayed on the UCSC genome browser, with gene structure and transcription orientation marked on the top. The tracks included Tcf1 ChIP-seq in WT CD8+ TCM cells and Tcf1-deficient naïve CD8+ T cells, ATAC-seq in resting WT and Tcf7ΔGzmb, and GP33-stimulated WT and Tcf7ΔGzmb CD8+ TCM cells, where black bars on the top denoted high-confidence Tcf1-binding sites and those at the bottom denoted WT-prepotent (that is, Tcf1-dependent, recall-‘opened’) ChrAcc sites with corresponding regions highlighted with yellow vertical bars.

Extended Data Fig. 7 Effect of ectopic expression of select factors on recall responses by CD8+ TCM cells.

a–b. Analysis of cell surface markers (a) and Id3 expression (b) in CD8+ TCM cells at resting state or after 24-h GP33 stimulation. Note that recall-induced CD25 and CD69 proteins (right panel in a) better demarcated CD8+ TCM cell activation than recall-repressed CD62L and IL-7Rα proteins (left panels in a). Sell and Il7r genes were greatly diminished in WT CD8+ TCM cells after 24-hr GP33 stimulation, as detected among the 2,998 genes in Fig. 3b, but their encoded proteins, CD62L and IL-7Rα, remained highly expressed at this time point (left panels), likely due to longer protein half-lives. Data are representative from 3 independent experiments with similar results. c. Experimental design for ‘rescue’ studies by gene delivery into BM progenitors. mCherry- or hNGFR-expressing retroviruses were used for gene delivery into CD45.2+ BM progenitors, which were transplanted into irradiatied CD45.1+ wild-type hosts to generate BM chimeras. mCherry+ (hNGFR+) naïve CD8+ T cells were then sorted from the BM chimeras to generate TCM cells, followed by in vivo and ex vivo analyses. d. Analysis of cell surface markers in mCherry+ (hNGFR+) CD45.2+CD8+ T cells in the spleens of the BM chimeras reconstituted with BM progenitors infected with mCherry (hNGFR)-expressing retroviruses, where the frequency of CD62L+CD44med-lo naïve phenotype cells is shown in representative contour plots from 2 independent experiments. e. Detection of glycolytic gene expression in mCherry+hNGFR+CD8+ TCM cells with RT-PCR at resting state, where the transcripts of each gene were first normalized to Hprt in each cell type, and further normalized to resting WT CD8+ TCM cells to calculate the relative expression of the gene in all cell types. Data are mean ± s.d. (n = 4) from 2 independent experiments. f. Detection of CD62L+CD8+ TCM cell formation in recipient spleens at ≥30 days after CD45.2+ mCherry+ (hNGFR+) naïve P14 CD8+ T cells were sort-purified from the BM chimeras, transferred into CD45.1+ wild-type recipients followed by LCMV infection the next day. The frequency of CD62L+ CD8+ TCM cells is shown in representative contour plots (left) from 2 independent experiments, and cumulative data on CD8+ TCM cell frequency (right) are means ± s.d. g. Experimental design for ‘rescue’ studies by gene delivery into CD8+ T cells primed in vivo. WT or Tcf7ΔGzmb P14 transgenic mice were i.v. infected with LCMV-Arm to prime P14 CD8+ T cells, which were then infected with mCherry-expressing retroviruses. The retrovirus-transduced cells were transferred into primary recipients to generated CD8+ TCM cells, and the mCherry+ CD8+ TCM cells were then sort-purified for in vivo analyses in secondary Tcra–/– recipients. h. Enumerating CD45.2+mCherry+CD8+ TCM cells in the recipient spleens at ≥day 25 after EV- or Id3-mCherry retrovirus-transduced WT or Tcf7ΔGzmb P14 CD8+ T cells were transferred into CD45.1+ wild-type recipients, followed by LCMV-Arm infection the next day. Data are means ± s.d. from 2 independent experiments. Statistical significance for multiple group comparisons in e, f, and h was first determined with one-way ANOVA for multi-group comparison, and Tukey’s test was used as post hoc correction for indicated pairwise comparisons. ns, not statistically significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shan, Q., Hu, S.S., Zhu, S. et al. Tcf1 preprograms the mobilization of glycolysis in central memory CD8+ T cells during recall responses. Nat Immunol 23, 386–398 (2022). https://doi.org/10.1038/s41590-022-01131-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01131-3

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