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.

Autophagy is essential for effector CD8+ T cell survival and memory formation

Abstract

The importance of autophagy in the generation of memory CD8+ T cells in vivo is not well defined. We report here that autophagy was dynamically regulated in virus-specific CD8+ T cells during acute infection of mice with lymphocytic choriomeningitis virus. In contrast to the current paradigm, autophagy decreased in activated proliferating effector CD8+ T cells and was then upregulated when the cells stopped dividing just before the contraction phase. Consistent with those findings, deletion of the gene encoding either of the autophagy-related molecules Atg5 or Atg7 had little to no effect on the proliferation and function of effector cells, but these autophagy-deficient effector cells had survival defects that resulted in compromised formation of memory T cells. Our studies define when autophagy is needed during effector and memory differentiation and warrant reexamination of the relationship between T cell activation and autophagy.

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

Figure 1: Analysis of autophagy in virus-specific CD8+ T cells during a viral infection.
Figure 2: Autophagic flux in virus-specific CD8+ T cells is inversely correlated with cell proliferation status.
Figure 3: Autophagic flux is inhibited during the T cell clonal expansion phase via impairment of autophagosome maturation into autophagolysosomes.
Figure 4: Atg7 deficiency results in survival defects in effector CD8+ T cells during the effector-to-memory transition.
Figure 5: Survival defects of Atg5-deficient T cells during the contraction phase following infection with LCMV Armstrong strain.
Figure 6: Antigen-specific CD8+ T cells lacking Atg7 exhibit cell-intrinsic defects in the development into long-term memory cells in chimeras.
Figure 7: Metabolomic and transcriptomic analysis of Atg7-deficient CD8+ T cells.
Figure 8: Autophagy in CD8+ T cells is essential for regulating chronic infection with LCMV.

Accession codes

Primary accessions

Gene Expression Omnibus

References

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

    CAS  PubMed  Google Scholar 

  2. Kaech, S.M. & Wherry, E.J. Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection. Immunity 27, 393–405 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kaech, S.M. et al. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat. Immunol. 4, 1191–1198 (2003).

    CAS  PubMed  Google Scholar 

  4. Sarkar, S. et al. Functional and genomic profiling of effector CD8 T cell subsets with distinct memory fates. J. Exp. Med. 205, 625–640 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Joshi, N.S. et al. Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Levine, B., Mizushima, N. & Virgin, H.W. Autophagy in immunity and inflammation. Nature 469, 323–335 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Mizushima, N., Yoshimori, T. & Levine, B. Methods in mammalian autophagy research. Cell 140, 313–326 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Walsh, C.M. & Edinger, A.L. The complex interplay between autophagy, apoptosis, and necrotic signals promotes T-cell homeostasis. Immunol. Rev. 236, 95–109 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Pua, H.H., Dzhagalov, I., Chuck, M., Mizushima, N. & He, Y.W. A critical role for the autophagy gene Atg5 in T cell survival and proliferation. J. Exp. Med. 204, 25–31 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Stephenson, L.M. et al. Identification of Atg5-dependent transcriptional changes and increases in mitochondrial mass in Atg5-deficient T lymphocytes. Autophagy 5, 625–635 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, R.C. & Levine, B. Autophagy in cellular growth control. FEBS Lett. 584, 1417–1426 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Hubbard, V.M. et al. Macroautophagy regulates energy metabolism during effector T cell activation. J. Immunol. 185, 7349–7357 (2010).

    CAS  PubMed  Google Scholar 

  13. Li, C. et al. Autophagy is induced in CD4+ T cells and important for the growth factor-withdrawal cell death. J. Immunol. 177, 5163–5168 (2006).

    CAS  PubMed  Google Scholar 

  14. Rubinsztein, D.C., Codogno, P. & Levine, B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat. Rev. Drug Discov. 11, 709–730 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Pua, H.H., Guo, J., Komatsu, M. & He, Y.W. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J. Immunol. 182, 4046–4055 (2009).

    CAS  PubMed  Google Scholar 

  16. Pankiv, S. et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 282, 24131–24145 (2007).

    CAS  PubMed  Google Scholar 

  17. Shvets, E., Fass, E. & Elazar, Z. Utilizing flow cytometry to monitor autophagy in living mammalian cells. Autophagy 4, 621–628 (2008).

    CAS  PubMed  Google Scholar 

  18. Gump, J.M. et al. Autophagy variation within a cell population determines cell fate through selective degradation of Fap-1. Nat. Cell Biol. 16, 47–54 (2014).

    CAS  PubMed  Google Scholar 

  19. Kimura, S., Noda, T. & Yoshimori, T. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 3, 452–460 (2007).

    CAS  PubMed  Google Scholar 

  20. Kuma, A. et al. The role of autophagy during the early neonatal starvation period. Nature 432, 1032–1036 (2004).

    CAS  PubMed  Google Scholar 

  21. Hara, T. et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441, 885–889 (2006).

    CAS  PubMed  Google Scholar 

  22. Jacob, J. & Baltimore, D. Modelling T-cell memory by genetic marking of memory T cells in vivo. Nature 399, 593–597 (1999).

    CAS  PubMed  Google Scholar 

  23. Masopust, D., Murali-Krishna, K. & Ahmed, R. Quantitating the magnitude of the lymphocytic choriomeningitis virus-specific CD8 T-cell response: it is even bigger than we thought. J. Virol. 81, 2002–2011 (2007).

    CAS  PubMed  Google Scholar 

  24. Waggoner, S.N., Cornberg, M., Selin, L.K. & Welsh, R.M. Natural killer cells act as rheostats modulating antiviral T cells. Nature 481, 394–398 (2012).

    CAS  Google Scholar 

  25. Lang, P.A. et al. Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity. Proc. Natl. Acad. Sci. USA 109, 1210–1215 (2012).

    CAS  PubMed  Google Scholar 

  26. 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).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  28. O'Sullivan, D. et al. Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 41, 75–88 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Wellen, K.E. et al. The hexosamine biosynthetic pathway couples growth factor-induced glutamine uptake to glucose metabolism. Genes Dev. 24, 2784–2799 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Wherry, E.J., Blattman, J.N., Murali-Krishna, K., van der Most, R. & Ahmed, R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77, 4911–4927 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Zajac, A.J. et al. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205–2213 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Jia, W., Pua, H.H., Li, Q.J. & He, Y.W. Autophagy regulates endoplasmic reticulum homeostasis and calcium mobilization in T lymphocytes. J. Immunol. 186, 1564–1574 (2011).

    CAS  PubMed  Google Scholar 

  34. Kaech, S.M., Hemby, S., Kersh, E. & Ahmed, R. Molecular and functional profiling of memory CD8 T cell differentiation. Cell 111, 837–851 (2002).

    CAS  PubMed  Google Scholar 

  35. Rathmell, J.C., Farkash, E.A., Gao, W. & Thompson, C.B. IL-7 enhances the survival and maintains the size of naive T cells. J. Immunol. 167, 6869–6876 (2001).

    CAS  PubMed  Google Scholar 

  36. Ma, A., Koka, R. & Burkett, P. Diverse functions of IL-2, IL-15, and IL-7 in lymphoid homeostasis. Annu. Rev. Immunol. 24, 657–679 (2006).

    CAS  PubMed  Google Scholar 

  37. Jones, R.G. & Thompson, C.B. Revving the engine: signal transduction fuels T cell activation. Immunity 27, 173–178 (2007).

    CAS  PubMed  Google Scholar 

  38. Grayson, J.M., Laniewski, N.G., Lanier, J.G. & Ahmed, R. Mitochondrial potential and reactive oxygen intermediates in antigen-specific CD8+ T cells during viral infection. J. Immunol. 170, 4745–4751 (2003).

    CAS  PubMed  Google Scholar 

  39. Paul, S., Kashyap, A.K., Jia, W., He, Y.W. & Schaefer, B.C. Selective autophagy of the adaptor protein Bcl10 modulates T cell receptor activation of NF-κB. Immunity 36, 947–958 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Komatsu, M. et al. The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Nat. Cell Biol. 12, 213–223 (2010).

    CAS  PubMed  Google Scholar 

  41. Araki, K. et al. mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108–112 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim, J., Kundu, M., Viollet, B. & Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13, 132–141 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Guo, J.Y. et al. Autophagy suppresses progression of K-ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev. 27, 1447–1461 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Singh, R. et al. Autophagy regulates lipid metabolism. Nature 458, 1131–1135 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Jordan, M.S. Genetic reconstitution of bone marrow for the study of signal transduction ex vivo. Methods Mol. Biol. 332, 331–342 (2006).

    CAS  PubMed  Google Scholar 

  46. Murali-Krishna, K. et al. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity 8, 177–187 (1998).

    CAS  PubMed  Google Scholar 

  47. Yu, T., Park, Y., Johnson, J.M. & Jones, D.P. apLCMS–adaptive processing of high-resolution LC/MS data. Bioinformatics 25, 1930–1936 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Li, S. et al. Predicting network activity from high throughput metabolomics. PLoS Comput. Biol. 9, e1003123 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Rao (La Jolla Institute for Allergy and Immunology) for the MSCV-IRES-Thy-1.1 retroviral vector; J. Jacob (Emory University) for Gzmb-Cre transgenic mice; V. Tran for the LC-MS experiments; R. Karaffa and S. Durham for sorting cells by flow cytometry at the Emory Flow Cytometry Core Facility; and A. Rae for assistance in the use of ImageStream. Supported by the US National Institutes of Health (R01 AI030048 to R.A. and R01 AI084887 to H.W.V.), the Mérieux Foundation (R.A.) and the Crohn's and Colitis Foundation (H.W.V.).

Author information

Authors and Affiliations

Authors

Contributions

X.X., K.A., S.L., M.W.B., J.M., D.R.G., D.P.J., H.W.V. and R.A. designed the research; X.X., K.A., S.L., J.-H.H., L.Y., W.G.T. and B.T.K. performed experiments. X.X., K.A., S.L., M.W.B., E.L.P., D.R.G., D.P.J., H.W.V. and R.A. analyzed data; and X.X., K.A., S.L., E.L.P., H.W.V. and R.A. wrote the manuscript.

Corresponding authors

Correspondence to Koichi Araki, Herbert W Virgin or Rafi Ahmed.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Dynamic regulation of autophagy in antigen-specific CD8+ T cells.

(a) A cartoon illustration of autophagy pathway highlighting molecules that are used to assess autophagy activity in this study. LC3b and p62 are both targeted to autophagosomes, which subsequently fuse with lysosomes for degradation. (b and c) mRNA levels of LC3b (b) and p62 (c) in P14 at different time points post LCMV Armstrong infection. (d) Protein expression levels of LC3b and p62 at early stages of P14 cell activation. GP33 peptide (200 µg) was intraveneously injected into P14 transgenic mice. The corresponding mRNA levels are show in (e). Errors bars in (b), (c) and (e) represent SEM. (b) - (e) are representative of at least two independent experiments.

Supplementary Figure 2 Autophagy activity during the early expansion phase of CD8+ T cells.

(a) Experimental set-up. Retrovirus transduced P14 cells were adoptively transferred into B6 mice, followed by LCMV Armstrong infection. (b-e) Flow cytometry plots of adoptively transferred P14 cells in spleens. P14 cells transduced with MIT retrovirus harboring GFP-LC3b (either wild type or G120A mutant) are positive for the congenic marker Thy1.1. Day 2 (b) and day 3 (d) p.i. splenocytes were used for the analysis. The percentage of GFP-negative cells out of the transduced P14 (Thy1.1+) cells from each group is summarized in (c) and (e). Errors bars in (c) and (e) represent SEM. (b) and (d) are representative of two independent experiments, n3 in each group.

Supplementary Figure 3 Autophagy activity measured in endogenous CD8+ T cells using retrovirus carrying the reporter gene encoding GFP-LC3b.

(a) Experimental set-up. Hematopoietic stem cells transduced with MIT retrovirus harboring GFP-LC3b (either wild type or G120A mutant) were introduced into irradiated host mice, which were left for 8-10 weeks for reconstitution of the hematopoietic system. (b) Flow plots showing changes in GFP intensity over the course of LCMV Armstrong infection in MIT-GFP-LC3b and MIT-G120A groups. Summary plots were shown on the right. (b) is representative of two independent experiments, n2 in each group.

Supplementary Figure 4 Characterization of Atg7-deficient H-2Db–gp33–specific CD8+ T cells.

(a) DbGP33-specific CD8 T cells were purified from day 8 mice infected with LCMV Armstrong (2x105 pfu). Representative plots of target cells before and after cell sorting, showing a typical > 95% purity. (b) Flow cytometry plot of total activated CD8 T cells (CD44hiCD62Llo) before and after cell sorting. Spleens were collected from mice infected with LCMV Armstrong (2x105 pfu) at day 8 p.i.. (c) Phenotypic properties of Atg7-deficient DbGP33-specific T cells at day 8 p.i (black outlined histograms). Solid gray histograms represent wild-type DbGP33-specific T cells. (d) Numbers of tetramer-positive cells in spleens 5 days p.i. (e) Flow plots showing percent of cells showing Brdu+ tetramer+ CD8+ T cells 5 hr after Brdu i.p. injection at day 5 p.i.; data summarized in (f). (g) Flow plots showing percent of cells are Annexin V+ tetramer+ CD8+ T cells at day 5 p.i; data summarized in (h). Errors bars in (d), (f) and (h) represent SEM. (c), (e) and (g) are representative of at least two independent experiments, n3 in each group.

Supplementary Figure 5 Bone marrow chimera reconstitution and T cell response following infection with LCMV Armstrong strain.

(a) Experimental set-up for generating Atg7fl/fl plus C57BL/6 (CD45.2/CD45.1) control and Atg7fl/fl Gzmb-Cre plus C57BL/6 (CD45.2/CD45.1) experimental mixed bone marrow chimera mice. CD8 T cell response was evaluated following the LCMV Armstrong infection. (b) Peripheral blood mononuclear cells were used to assess the level of reconstitution in the bone marrow chimeras prior to infection. Gated on total CD8 T cells. Plots to the right of the arrows indicate the level of reconstitution 30 days p.i in CD44lo populations. Gated on total CD8 T cells. Number on each quadrant represents the percentage in each. (c) Summary plot of percentage of DbGP33- and DbNP396-specific T cells from day 8 to day 30 in the peripheral blood of the chimeric mice. The number of tetramer-positive cells on day 8 p.i. is normalized to 100%. The dashed line represents tetramer-positive cell of C57BL/6 origin (CD45.1+) and the solid black and red lines of Atg7fl/fl and Atg7fl/fl Gzmb-Cre origin (CD45.2+), respectively. Each line represents data from one experimental mouse. Data are representative of two independent experiments.

Supplementary Figure 6 Metabolic activity network of Atg7-deficient H-2Db–gp33+ CD8+ T cells.

Prediction directly from m/z feature tables (Supplementary Table 1) by method previously described1. Metabolites are colored according to log2 fold change.

1. Li, S. et al. Predicting network activity from high throughput metabolomics. PLoS Comput Biol 9, e1003123 (2013).

Supplementary Figure 7 Virus-specific CD8+ T cell response during chronic infection with LCMV in the absence of Atg7.

Atg7fl/fl Gzmb-Cre and control mice were infected with LCMV Clone-13. (a) and (c) Flow cytometric analysis of LCMV-specific T cells post LCMV Clone 13 infection in PBMC, spleen, liver and lung on days 8 and 15 p.i., respectively. The numbers on FACS plots indicate the percentage of DbGP276-specific T cells on the gated CD8 T cells in each sample examined. (b) and (d) The total numbers of DbGP276-specific T cells in each tissue are plotted at days 8 and 15 p.i., respectively. n=3-5 in each group. Data are representative of two independent experiments. Error bars indicate SEM. *, p0.05. ***, p0.0005

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 1226 kb)

Supplementary Table 1

List of metabolites that were significantly different between Atg7f/f and Atg7f/f Gzmb-Cre (XLS 27 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, X., Araki, K., Li, S. et al. Autophagy is essential for effector CD8+ T cell survival and memory formation. Nat Immunol 15, 1152–1161 (2014). https://doi.org/10.1038/ni.3025

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3025

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