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:

Functional T cells are capable of supernumerary cell division and longevity

Abstract

Differentiated somatic mammalian cells putatively exhibit species-specific division limits that impede cancer but may constrain lifespans1,2,3. To provide immunity, transiently stimulated CD8+ T cells undergo unusually rapid bursts of numerous cell divisions, and then form quiescent long-lived memory cells that remain poised to reproliferate following subsequent immunological challenges. Here we addressed whether T cells are intrinsically constrained by chronological or cell-division limits. We activated mouse T cells in vivo using acute heterologous prime–boost–boost vaccinations4, transferred expanded cells to new mice, and then repeated this process iteratively. Over 10 years (greatly exceeding the mouse lifespan)5 and 51 successive immunizations, T cells remained competent to respond to vaccination. Cells required sufficient rest between stimulation events. Despite demonstrating the potential to expand the starting population at least 1040-fold, cells did not show loss of proliferation control and results were not due to contamination with young cells. Persistent stimulation by chronic infections or cancer can cause T cell proliferative senescence, functional exhaustion and death6. We found that although iterative acute stimulations also induced sustained expression and epigenetic remodelling of common exhaustion markers (including PD1, which is also known as PDCD1, and TOX) in the cells, they could still proliferate, execute antimicrobial functions and form quiescent memory cells. These observations provide a model to better understand memory cell differentiation, exhaustion, cancer and ageing, and show that functionally competent T cells can retain the potential for extraordinary population expansion and longevity well beyond their organismal lifespan.

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: CD8+ T cells can undergo seemingly unlimited bursts of proliferation if rested between stimulations.
Fig. 2: Iteratively boosted T cells maintain telomere length, cell cycle control and durability.
Fig. 3: Iterative boosting induces progressive changes in gene and protein expression and acquisition of exhaustion markers.
Fig. 4: Transcriptional, epigenetic and functional profiling distinguishes ISTCs from exhausted T cells.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. Sequencing data are available through GEO (accession number GSE213230). Source data are provided with this paper.

References

  1. Stanley, J. F., Pye, D. & MacGregor, A. Comparison of doubling numbers attained by cultured animal cells with life span of species. Nature 255, 158–159 (1975).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Röhme, D. Evidence for a relationship between longevity of mammalian species and life spans of normal fibroblasts in vitro and erythrocytes in vivo. Proc. Natl Acad. Sci. USA 78, 5009–5013 (1981).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  3. Bodnar, A. G. et al. Extension of life-span by introduction of telomerase into normal human cells. Science 279, 349–352 (1998).

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Masopust, D., Ha, S.-J., Vezys, V. & Ahmed, R. Stimulation history dictates memory CD8 T cell phenotype: implications for prime-boost vaccination. J. Immunol. 177, 831–839 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Kunstyr, I. & Leuenberger, H. G. Gerontological data of C57BL/6J mice. I. Sex differences in survival curves. J. Gerontol. 30, 157–162 (1975).

    Article  CAS  PubMed  Google Scholar 

  6. 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).

    Article  CAS  PubMed  Google Scholar 

  7. Carrel, A. On the permanent life of tissues outside of the organism. J. Exp. Med. 15, 516–528 (1912).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ebeling, A. H. A ten year old strain of fibroblasts. J. Exp. Med. 35, 755–759 (1922).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Witkowski, J. A. Dr. Carrel’s immortal cells. Med. Hist. 24, 129–142 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hayflick, L. & Moorhead, P. S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621 (1961).

    Article  CAS  PubMed  Google Scholar 

  11. Allsopp, R. C. et al. Telomere length predicts replicative capacity of human fibroblasts. Proc. Natl Acad. Sci. USA 89, 10114–10118 (1992).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Goldstein, S. Aging in vitro growth of cultured cells from the Galapagos tortoise. Exp. Cell Res. 83, 297–302 (1974).

    Article  Google Scholar 

  13. de Magalhães, J. P. & Toussaint, O. Telomeres and telomerase: a modern fountain of youth? Rejuv. Res. 7, 126–133 (2004).

    Article  Google Scholar 

  14. Cagan, A. et al. Somatic mutation rates scale with lifespan across mammals. Nature 604, 517–524 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Boer, R. J. D., Homann, D. & Perelson, A. S. Different dynamics of CD4+ and CD8+ T cell responses during and after acute lymphocytic choriomeningitis virus infection. J. Immunol. 171, 3928–3935 (2003).

    Article  PubMed  Google Scholar 

  16. Morgan, D. A., Ruscetti, F. W. & Gallo, R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science 193, 1007–1008 (1976).

    Article  ADS  CAS  PubMed  Google Scholar 

  17. Gillis, S. & Smith, K. A. Long term culture of tumour-specific cytotoxic T cells. Nature 268, 154–156 (1977).

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Effros, R. B., Dagarag, M., Spaulding, C. & Man, J. The role of CD8+ T‐cell replicative senescence in human aging. Immunol. Rev. 205, 147–157 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Rubin, H. The disparity between human cell senescence in vitro and lifelong replication in vivo. Nat. Biotechnol. 20, 675–681 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Akbar, A. N. & Henson, S. M. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat. Rev. Immunol. 11, 289–295 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Rosenberg, S. A. et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. N. Engl. J. Med. 319, 1676–1680 (1988).

    Article  CAS  PubMed  Google Scholar 

  22. Rosenberg, S. A. et al. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J. Natl Cancer Inst. 86, 1159–1166 (1994).

    Article  CAS  PubMed  Google Scholar 

  23. Dudley, M. E. & Rosenberg, S. A. Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nat. Rev. Cancer 3, 666–675 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang, Y., Joe, G., Hexner, E., Zhu, J. & Emerson, S. G. Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nat. Med. 11, 1299–1305 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Gattinoni, L. et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 15, 808–813 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gattinoni, L. et al. A human memory T cell subset with stem cell–like properties. Nat. Med. 17, 1290–1297 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Graef, P. et al. Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8+ central memory T cells. Immunity 41, 116–126 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Wirth, T. C. et al. Repetitive antigen stimulation induces stepwise transcriptome diversification but preserves a core signature of memory CD8+ T cell differentiation. Immunity 33, 128–140 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Rai, D., Martin, M. D. & Badovinac, V. P. The longevity of memory CD8 T cell responses after repetitive antigen stimulations. J. Immunol. 192, 5652–5659 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Fraser, K. A., Schenkel, J. M., Jameson, S. C., Vezys, V. & Masopust, D. Preexisting high frequencies of memory CD8+ T cells favor rapid memory differentiation and preservation of proliferative potential upon boosting. Immunity 39, 171–183 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Thompson, E. A., Beura, L. K., Nelson, C. E., Anderson, K. G. & Vezys, V. Shortened intervals during heterologous boosting preserve memory CD8 T cell function but compromise longevity. J. Immunol. 196, 3054–3063 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Rubin, H. Telomerase and cellular lifespan: ending the debate? Nat. Biotechnol. 16, 396–397 (1998).

    Article  CAS  PubMed  Google Scholar 

  33. Mondello, C. et al. Telomere length in fibroblasts and blood cells from healthy centenarians. Exp. Cell Res. 248, 234–242 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Weng, N. P., Levine, B. L., June, C. H. & Hodes, R. J. Regulated expression of telomerase activity in human T lymphocyte development and activation. J. Exp. Med. 183, 2471–2479 (1996).

    Article  CAS  PubMed  Google Scholar 

  35. Weng, N., Hathcock, K. S. & Hodes, R. J. Regulation of telomere length and telomerase in T and B cells: a mechanism for maintaining replicative potential. Immunity 9, 151–157 (1998).

    Article  CAS  PubMed  Google Scholar 

  36. Hathcock, K. S., Kaech, S. M., Ahmed, R. & Hodes, R. J. Induction of telomerase activity and maintenance of telomere length in virus-specific effector and memory CD8+ T cells. J. Immunol. 170, 147–152 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Callicott, R. J. & Womack, J. E. Real-time PCR assay for measurement of mouse telomeres. Comp. Med. 56, 17–22 (2006).

    CAS  PubMed  Google Scholar 

  38. Bally, A. P. R., Austin, J. W. & Boss, J. M. Genetic and epigenetic regulation of PD-1 expression. J. Immunol. 196, 2431–2437 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Duraiswamy, J. et al. Phenotype, function, and gene expression profiles of programmed death-1hi CD8 T cells in healthy human adults. J. Immunol. 186, 4200–4212 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Sekine, T. et al. TOX is expressed by exhausted and polyfunctional human effector memory CD8+ T cells. Sci. Immunol. 5, eaba7918 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Lee, K. et al. Characterization of age‐associated exhausted CD8+ T cells defined by increased expression of Tim‐3 and PD‐1. Aging Cell 15, 291–300 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bengsch, B. et al. Epigenomic-guided mass cytometry profiling reveals disease-specific features of exhausted CD8 T cells. Immunity 48, 1029–1045 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Altman, J. D. & Davis, M. M. MHC‐peptide tetramers to visualize antigen‐specific T cells. Curr. Protoc. Immunol. 115, 17.3.1–17.3.44 (2016).

    Article  PubMed  Google Scholar 

  44. Obar, J. J., Khanna, K. M. & Lefrançois, L. Endogenous naive CD8+ T cell precursor frequency regulates primary and memory responses to infection. Immunity 28, 859–869 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC‐seq: a method for assaying chromatin accessibility genome‐wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  PubMed  Google Scholar 

  46. Künzli, M. et al. Long-lived T follicular helper cells retain plasticity and help sustain humoral immunity. Sci. Immunol. 5, eaay5552 (2020).

    Article  PubMed  Google Scholar 

  47. Youngblood, B. et al. Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells. Immunity 35, 400–412 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Steinert, E. M. et al. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell 161, 737–749 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc. 9, 209–222 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the laboratories of D.M. and V.V. for helpful discussions. We thank the University of Minnesota Flow Cytometry resource for cell sorting (J. Motl, T. Martin and R. Arora). This study was supported by National Institutes of Health grants R01 AI084913, R01 AI146032 (D.M.) and T32HL007741 (A.G.S.), and Swiss National Science Foundation grant P2BSP3_200187 (M.K.).

Author information

Authors and Affiliations

Authors

Contributions

A.G.S., M.K., C.F.Q., M.C.S., L.S., J.L. and H.E.G. carried out the experiments, analysed data and prepared visualizations. B.Y. analysed data and prepared visualizations. D.Z. created and shared reagents. A.G.S., M.K., V.V. and D.M. designed the experiments and wrote the manuscript.

Corresponding author

Correspondence to David Masopust.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Susan Kaech and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Iteratively stimulated T cells populate non-lymphoid tissues.

a) CD45.1+ 48° ISTC memory CD8 T cells were transferred to naïve mice, followed by three heterologous prime-boost-boost immunizations. Non-lymphoid tissues were analyzed 48 days after the 51° boost. Flow cytometry plots are gated on live CD8a+ lymphocytes that were not stained by intravascular in vivo antibody labeling. b) CD69 expression on endogenous 3° cells (CD45.1-) and 48° memory CD8 T cells (CD45.1+).Plots concatenated from four mice and experiment is representative of three similar experiments with similar results.

Extended Data Fig. 2 Gene set enrichment analysis of iteratively stimulated CD8 T cells.

Gene set enrichment analysis was performed on differentially expressed genes between 45° ISTCs and primary memory cells using the category “Biological process” of the Gene Ontology database. The top 20 upregulated (activated) and top 20 downregulated (suppressed) pathways in ISTCs are shown. Statistical significance was determined using Over-Representation analysis.

Extended Data Fig. 3 A single naïve CD8 T cell shows the potential to produce >1040 memory cell progeny.

a) Schematic showing estimate of cell expansion potential after 51 HPBB immunizations. HPBB: three successive Heterologous Prime, Boost, Boost immunizations. b) Schematic comparing the volume of earth with the theoretical volume of memory T cells implied by the calculated proliferation potential of a naïve CD8 T cell.

Supplementary information

Supplementary Information 1

This file contains Supplementary Figs. 1–4 showing the gating schemes used in Figs. 14 and Supplementary Table 1, which shows the number of animals at each time point in Fig. 1a,b.

Reporting Summary

Supplementary Information 2

This file lists the differentially expressed genes between 45° cells and 3° cells, 1° or naive CD8+ T cells. Empirical Bayes moderation was used to test for significance.

Supplementary Information 3

Extended statistical results showing the full details of statistics carried out for Fig. 4g.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Soerens, A.G., Künzli, M., Quarnstrom, C.F. et al. Functional T cells are capable of supernumerary cell division and longevity. Nature 614, 762–766 (2023). https://doi.org/10.1038/s41586-022-05626-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05626-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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