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:

The kinase complex mTORC2 promotes the longevity of virus-specific memory CD4+ T cells by preventing ferroptosis

Abstract

Antigen-specific memory CD4+ T cells can persist and confer rapid and efficient protection from microbial reinfection. However, the mechanisms underlying the long-term maintenance of the memory CD4+ T cell pool remain largely unknown. Here, using a mouse model of acute infection with lymphocytic choriomeningitis virus (LCMV), we found that the serine/threonine kinase complex mammalian target of rapamycin complex 2 (mTORC2) is critical for the long-term persistence of virus-specific memory CD4+ T cells. The perturbation of mTORC2 signaling at memory phase led to an enormous loss of virus-specific memory CD4+ T cells by a unique form of regulated cell death (RCD), ferroptosis. Mechanistically, mTORC2 inactivation resulted in the impaired phosphorylation of downstream AKT and GSK3β kinases, which induced aberrant mitochondrial reactive oxygen species (ROS) accumulation and ensuing ferroptosis-causative lipid peroxidation in virus-specific memory CD4+ T cells; furthermore, the disruption of this signaling cascade also inhibited glutathione peroxidase 4 (GPX4), a major scavenger of lipid peroxidation. Thus, the mTORC2–AKT–GSK3β axis functions as a key signaling hub to promote the longevity of virus-specific memory CD4+ T cells by preventing ferroptosis.

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: mTORC2 signaling is required for the formation and maintenance of the memory CD4+ T cell pool.
Fig. 2: mTORC2 deficiency induces non-apoptotic cell death in virus-specific memory CD4+ T cells.
Fig. 3: mTORC2 deficiency induces ferroptosis in memory CD4+ T cells.
Fig. 4: Overexpression of GPX4 restricts ferroptosis in mTORC2-deficient memory CD4+ T cells.
Fig. 5: mTORC2 deficiency induces aberrant mitochondrial ROS production in memory CD4+ T cells.
Fig. 6: mTORC2–p-AKTSer 473–p-GSK3βSer 9 signaling regulates HK2–VDAC interaction to orchestrate mitochondrial ROS generation.
Fig. 7: The mTORC2 kinase supports GPX4 peroxidase activity via p-AKTSer 473–p-GSK3βSer 9-mediated NRF2 nuclear translocation.
Fig. 8: Constitutively inactive GSK3β-S9D restores memory CD4+ T cells from mTORC2 deficiency-induced ferroptosis.

Similar content being viewed by others

Data availability

The microarray raw data are deposited and available from the GEO database under the accession number GSE151800. All other data supporting the findings are available in this paper or from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Crotty, S. Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 29, 621–663 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Zhou, L., Chong, M. M. W. & Littman, D. R. Plasticity of CD4+ T cell lineage differentiation. Immunity 30, 646–655 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Fahey, L. M. et al. Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells. J. Exp. Med. 208, 987–999 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ahmed, R. & Gray, D. Immunological memory and protective immunity: understanding their relation. Science 272, 54–60 (1996).

    Article  CAS  PubMed  Google Scholar 

  5. Kaech, S. M., Wherry, E. J. & Ahmed, R. Effector and memory T-cell differentiation: implications for vaccine development. Nat. Rev. Immunol. 2, 251–262 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Huang, H., Long, L., Zhou, P., Chapman, N. M. & Chi, H. mTOR signaling at the crossroads of environmental signals and T-cell fate decisions. Immunol. Rev. 295, 15–38 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Waickman, A. T. & Powell, J. D. mTOR, metabolism, and the regulation of T-cell differentiation and function. Immunol. Rev. 249, 43–58 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chi, H. Regulation and function of mTOR signalling in T cell fate decisions. Nat. Rev. Immunol. 12, 325–338 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wullschleger, S., Loewith, R. J. & Hall, M. N. TOR signaling in growth and metabolism. Cell 124, 471–484 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Liu, G. Y. & Sabatini, D. M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol. 21, 183–203 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rao, R. R., Li, Q., Odunsi, K. & Shrikant, P. A. The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and eomesodermin. Immunity 32, 67–78 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Pollizzi, K. N. et al. mTORC1 and mTORC2 selectively regulate CD8+ T cell differentiation. J. Clin. Invest. 125, 2090–2108 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Michelini, R. H., 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  CAS  PubMed Central  Google Scholar 

  14. Ye, L. et al. mTOR promotes antiviral humoral immunity by differentially regulating CD4 helper T cell and B cell responses. J. Virol. 91, e01653-16 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Hao, Y. et al. The kinase complex mTOR complex 2 promotes the follicular migration and functional maturation of differentiated follicular helper CD4+ T cells during viral infection. Front. Immunol. 23, 1127 (2018).

    Article  CAS  Google Scholar 

  16. Zeng, H. et al. mTORC1 and mTORC2 kinase signaling and glucose metabolism drive follicular helper T cell differentiation. Immunity 45, 540–554 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yang, J. et al. Critical roles of mTOR complex 1 and 2 for T follicular helper cell differentiation and germinal center responses. eLife 30, e17936 (2016).

    Article  Google Scholar 

  18. Opferman, J. T. & Korsmeyer, S. J. Apoptosis in the development and maintenance of the immune system. Nat. Immunol. 4, 410–415 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Galluzzi, L. et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25, 486–541 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Michael, P. & Murphy, M. P. How mitochondria produce reactive oxygen species. Biochem. J. 417, 1–13 (2009).

    Article  CAS  Google Scholar 

  23. Paola et al. Mitochondrial metabolism of reactive oxygen species. Mitochondrion 13, 71–82 (2013).

    Article  CAS  Google Scholar 

  24. Chen, X., Kang, R., Kroemer, G. & Tang, D. Broadening horizons: the role of ferroptosis in cancer. Nat. Rev. Clin. Oncol. 18, 280–296 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Latunde-Dada & Gladys, O. Ferroptosis: role of lipid peroxidation, iron and ferritinophagy. Biochim. Biophys. Acta Gen. Subj. 1861, 1893–1900 (2017).

  26. Tobias, M. et al. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic. Biol. Med. 133, 144–152 (2018).

    Google Scholar 

  27. Ursini, F. & Maiorino, M. Lipid peroxidation and ferroptosis: the role of GSH and GPx4. Free Radic. Biol. Med. 152, 175–185 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Stockwell, B. R., Jiang, X. & Gu, W. Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol. 30, 478–490 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Laplante, M. & Sabatini, D. M. mTOR signaling at a glance. J. Cell Sci. 122, 3589–3594 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dixon, S. J., Patel, D. N., Welsch, M., Skouta, R. & Stockwell, B. R. Pharmacological inhibition of cystine–glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife 3, e02523 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Matsushita, M. et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J. Exp. Med. 212, 555–568 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Imai, H., Matsuoka, M., Kumagai, T., Sakamoto, T. & Koumura, T. Lipid peroxidation-dependent cell death regulated by GPx4 and ferroptosis. Curr. Top. Microbiol. Immunol. 403, 143–170 (2017).

    CAS  PubMed  Google Scholar 

  33. Pfeifer, H. et al. Identification of a specific sperm nuclei selenoenzyme necessary for protamine thiol cross-linking during sperm maturation. FASEB J. 15, 1236–1238 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Arai, M. et al. Mitochondrial phospholipid hydroperoxide glutathione peroxidase plays a major role in preventing oxidative injury to cells. J. Biol. Chem. 274, 4924–4933 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Gao, M. et al. Role of mitochondria in ferroptosis. Mol. Cell 73, 354–363 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Liu, X. et al. Acetate production from glucose and coupling to mitochondrial metabolism in mammals. Cell 175, 502–513 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Camara, A. K. S., Zhou, Y. F., Wen, P. C., Emad, T. & Wai-Meng, K. Mitochondrial VDAC1: a key gatekeeper as potential therapeutic target. Front. Physiol. 8, 460 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Pastorino, J. G. Activation of glycogen synthase kinase 3β disrupts the binding of hexokinase II to mitochondria by phosphorylating voltage-dependent anion channel and potentiates chemotherapy-induced cytotoxicity. Cancer Res. 65, 10545–10554 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Bantug, G. R. et al. Mitochondria-endoplasmic reticulum contact sites function as immunometabolic hubs that orchestrate the rapid recall response of memory CD8+ T cells. Immunity 48, 542–555 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hayes, J. D., & Dinkova-Kostova, A. T. et al. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 39, 199–218 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Bijur, G. N. & Jope, R. S. Rapid accumulation of Akt in mitochondria following phosphatidylinositol 3-kinase activation. J. Neurochem. 87, 1427–1435 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hayes, J. D., Chowdhry, S., Dinkova-Kostova, A. T. & Sutherland, C. et al. Dual regulation of transcription factor Nrf2 by Keap1 and by the combined actions of β-TrCP and GSK-3. Biochem. Soc. Trans. 43, 611–620 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Jam, A., Jaiswal, K. & Anil, K. GSK-3β acts upstream of Fyn kinase in regulation of nuclear export and degradation of NF-E2 related factor 2. J. Biol. Chem. 282, 16502–16512 (2007).

    Article  CAS  Google Scholar 

  44. Paramasivan, P., Kankia, I. H., Langdon, S. P. & Deeni, Y. Y. Emerging role of nuclear factor erythroid 2-related factor 2 in the mechanism of action and resistance to anticancer therapies. Cancer Drug Resist. 2, 490–515 (2019).

    Google Scholar 

  45. Weldy, C. S. et al. Glutathione (GSH) and the GSH synthesis gene Gclm modulate vascular reactivity in mice. Free Radic. Biol. Med. 53, 1264–1278 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Conrad, M. & Sato, H. The oxidative stress-inducible cystine/glutamate antiporter, system xc: cystine supplier and beyond. Amino Acids 42, 231–246 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. 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 

  48. 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 

  49. Intlekofer, A. M. et al. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat. Immunol. 6, 1236–1244 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Belabid, J. & Cheddadi, A. Structural and functional characterization of Nrf2 degradation by glycogen synthase kinase 3/β-TrCP. Free Radic. Biol. Med. 88, 147–157 (2015).

    Article  CAS  Google Scholar 

  51. Liu, Q. et al. Discovery of 1-(4-(4-propionylpiperazin-1-yl)-3-(trifluoromethyl)phenyl)-9-(quinolin-3-yl)benzo[h][1,6]naphthyridin-2(1H)-one as a highly potent, selective mammalian target of rapamycin (mTOR) inhibitor for the treatment of cancer. J. Med. Chem. 53, 7146–7155 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Badgley, M. A. et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ingold, I. et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell 172, 409–422 (2017).

    Article  PubMed  CAS  Google Scholar 

  56. Liu, H., Mi, S., Li, Z., Hua, F. & Hu, Z.-W. Interleukin 17A inhibits autophagy through activation of PIK3CA to interrupt the GSK3B-mediated degradation of BCL2 in lung epithelial cells. Autophagy 9, 730–742 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Morita, S., Kojima, T. & Kitamura, T. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Ther. 7, 1063–1066 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. Yao, C. et al. BACH2 enforces the transcriptional and epigenetic programs of stem-like CD8 T cells. Nat. Immunol. 22, 370–380 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank R. Ahmed (Emory University) for SMARTA transgenic mice and retroviral vectors. We thank CapitalBio Corporation (Beijing, China) for performing microarray experiments and data analysis, and we thank the core facility center of Third Military Medical University for cell sorting and TEM. This study was supported by grants from the National Key Research Development Plan (SQ2021YFC2300059 to L.Y.), the National Natural Science Fund for Distinguished Young Scholars (31825011 to L.Y.), the National Natural Science Foundation of China (32030041 to L.Y, 31800748 to Y.W., 31800742 to Q.T. and 31900642 to Y.H.), the China Postdoctoral Science Foundation (2019M663389 to Y.W.) and Chongqing Special Postdoctoral Science Foundation (XmT2018021 to Q.T.).

Author information

Authors and Affiliations

Authors

Contributions

Y.W., Q.T., Y.H., W.Y., J.L., C.C., X.C., Y. Lin, Q.H., L.X., J.H., S.L., Z.W., Y. Luo and Q.W. performed the experiments. Z.L., L.H. and J.T. provided reagents, materials and support. L.Y., Y.W. and Q.T. designed the study, analyzed the data and wrote the paper with X.Z. and Y.W. L.Y., J.X. and Z.Y. supervised the study.

Corresponding authors

Correspondence to Zhinan Yin, Jianqing Xu or Lilin Ye.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks the anonymous reviewers 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 IL-7 signaling sustains mTORC2 activity in memory CD4+ T cells.

a, Naïve SMARTA cells (CD45.1+) were adoptively transferred into C57BL/6J recipient mice (CD45.2+) and followed by infection with LCMV Arm+ the next day. SMARTA cells were analyzed via flow cytometry at D0 (Naïve, green), D5 (early Effector, blue), and D60 (Memory, red) post-infection. CD127 levels on transferred SMARTA cells in the spleens of recipients, from two independent experiments (n=4 for Naive and D5, n=5 for D90). b, p-AKTSer473 levels on memory transferred SMARTA cells as in a after stimulation with (red) or without (blue) recombinant human IL-7 ex vivo, from two independent experiments (n=4). c Experimental setup of IL-7 signaling blockade with αIL-7-mAb at the maintenance stage of CD4+ T cell memory. d, p-STAT5Tyr694 and p-AKTSer473 levels on memory SMARTA cells in the spleens of recipients as in c after administration with αIL-7-mAb at day 60 post-infection. The presented data are representative of two independent experiments (n=4 for αIL-7-mAb, n=5 for Control). e, f, Validation of the knockout of Rictor. Immunoblot analysis (e) of lysates of sorted memory CD4+ T cells from either CD4Cre-Rictorfl/fl (Rictor−/−) or Rictorfl/fl mice. The presented data are representative of four independent experiments. (f) p-AKTSer473 and p-S6Ser235/236 levels on memory CD4+ T cells in the spleens of recipients as in c. The presented data are representative of four independent experiments (n=4). Grey area in histogram indicates staining with rabbit isotype IgG. Data are presented as mean values ± SD and in a-b, d, f. Box plots show min to max, each data point represents one result from an independent experiment, and horizontal bars denote the median in e. Data are analyzed by unpaired two-tailed t-test in a-b, d-f.

Source data

Extended Data Fig. 2 The mTORC2-deficiency impairs memory CD4+ T cell pool but enhances memory CD8+ T cell pool in acute viral infection.

a, b, Analysis of TH1 (SLAMhighCXCR5 in a, CXCR5 in b) and TFH (SLAMlowCXCR5+ in a, CXCR5+ in b) cells among viral-specific memory CD4+ T (positive for LCMV GP66-77 tetramer) cells in the spleens of either Rictorfl/fl (blue) or CD4Cre-Rictorfl/fl (Rictor−/−, red) mice at day 8 (a) and day 100 (b) post-infection of LCMV Arm+. The presented data are representative of three independent experiments (n=6 for Rictorfl/fl and n=4 for Rictor−/− in a, n=6 for both groups in b). c, Viral-specific memory CD8+ T (positive for LCMV GP33-41 tetramer) cells in the spleens of either Rictorfl/fl (blue) or CD4Cre-Rictorfl/fl (Rictor−/−, red) mice. The presented data are representative of three independent experiments (n=4). d, Ratios of total CD8+ T cells and virus-specific CD8+ T cells (positive for LCMV GP33-41 tetramer) between ERT2Cre-Rictorfl/fl origin (CD45.2+) and wild-type origin (CD45.1+) in the spleens of bone marrow chimeras (BMCs) after administration of Tamoxifen (red) or vehicle (blue) at the memory maintenance stage as described in Fig. 2b, from three independent experiments (n=4). Data are presented as mean values ± SD and analyzed by unpaired two-tailed t-test in a-d.

Source data

Extended Data Fig. 3 Torin-1 induced mTORC2-deficiency impairs the virus-specific memory CD4+ T cell pool in vivo.

a, Experimental setup of in vivo administration of Torin-1, Rapamycin or vehicle on adoptively transferred memory SMARTA cells (CD45.1+) in recipients (CD45.2+). b, p-AKTSer473 levels on transferred SMARTA cells in spleens of recipients as in a. The presented data are representative of three independent experiments. c, p-S6Ser235/236 levels on memory SMARTA cells in the presence or absence of αCD3 plus αCD28 ex vivo in spleens of recipients. The presented data are representative of three independent experiments (n=4). d, Frequency change of memory SMARTA cells in PBMCs after administration of either Torin-1, Rapamycin, or vehicle in recipients as in a. The presented data are representative of three independent experiments (n=5). Grey area in histogram indicates staining with rabbit isotype IgG in b-c. Data are presented as mean values ± SD and analyzed by unpaired two-tailed t-test in c. Data are analyzed by paired two-tailed t-test in d.

Source data

Extended Data Fig. 4 No aberrant metabolic profiles in mTORC2-deficient memory CD4+ T cells.

a, b, Profile of (a) extracellular acidification rate (ECAR) and (b) oxygen consumption rate (OCR) of sorted memory CD4+ T cells (CD25GITRCD44highCD4+) from either CD4Cre-Rictorfl/fl (Rictor−/−, red) or Rictorfl/fl (blue) mice at day 60 post infection of LCMV Arm+ (n=3 biological replicates, pooled from at least 20 mice in CD4Cre-Rictorfl/fl group and 10 mice in Rictorfl/fl group). The presented data are representative of two independent experiments. Data are presented as mean values ± SD and analyzed by unpaired two-tailed t-test in a-b.

Source data

Extended Data Fig. 5 mTORC2-deficiency induces ferroptosis in both memory TH1 and TFH CD4+ T cells.

a, Bodipy C11 intensity on either wild-type SMARTA (blue) or Rictor−/−-SMARTA (red) TH1 and TFH cells in spleens of recipients at day 60 post infection of LCMV Arm+. The presented data are representative of three independent experiments (n=5 for WT, n=6 for Rictor−/−). b, Bodipy C11 intensity on bulk memory TH1 (CD25GITRCD44highCXCR5CD4+) and TFH (CD25GITRCD44highCXCR5+CD4+) cells in spleens of either Rictorfl/fl (blue) or ERT2Cre-Rictorfl/fl (Rictor−/−, red) mice after administration of Tamoxifen. The data presented are representative of three independent experiments (n=4). Data are presented as mean values ± SD and analyzed by unpaired two-tailed t-test in a-b.

Source data

Extended Data Fig. 6 IL-7 signaling is indispensable to protect memory CD4+ T cells from ferroptosis.

a, Experimental setup of IL-7 signaling blockade with αIL-7-mAb at the maintenance stage of CD4+ T cell memory. Naïve SMARTA cells (CD45.1+) were adoptively transferred into C57BL/6J recipient mice (CD45.2+) and followed by infection with LCMV Arm+ the next day. Recipients were injected intraperitoneally with αIL-7-mAb or PBS (Vehicle) at day 60 post infection every three days for a total four doses before sacrifice. b, Representative contour plots of memory SMARTA cell frequency and statistical analysis of the decreased proportion of memory SMARTA cells in the PMBCs before and after αIL-7-mAb administration and the endpoint cell number of memory SMARTA cells in the spleens of recipients at day 60 post LCMV Arm+ infection in the context of IL-7 signaling blockade as described in a. c, d, Dead cells (c, 7-AAD+) and apoptotic cell death (d, Annexin V+ 7-AAD) in memory SMARTA cells in spleens of recipients as described in a. e, Bodipy C11 intensity on SMARTA cells in spleens of recipients as described in a. The presented data are representative of two independent experiments (b-e, n=4 for αIL-7-mAb, n=5 for Control). Data are presented as mean values ± SD and analyzed by unpaired two-tailed t-test in b-e.

Source data

Extended Data Fig. 7 The mTORC2 signaling generally inhibits ferroptosis in memory CD4+ T cells.

LM-GP61 or IAV-GP61 is the recombinant Listeria monocytogenes or Influenza A Virus expressing LCMV glycoprotein-specific I-Ab restricted CD4 T cell epitope GP66-77. a, Experimental setup of inducible knockout of Rictor in BMCs generated with BM cells from C57BL/6 (WT, CD45.1+) and ERT2Cre-Rictorfl/fl (Rictor−/−, CD45.2+) mice at day 60 post infection of either LM-GP61 or IAV-GP61. b-e, Bodipy C11 intensity on memory CD4+ T cells in spleens of BMCs after administration of Tamoxifen at day 60 post infection with LM-GP61 (b) or IAV-GP61 (d) as described in a. Ratio between Rictor−/− (CD45.2+) and WT (CD45.1+) viral specific memory CD4+ T cells (GP66-77+) in PBMCs from BMCs before and after administration of Tamoxifen at day 60 post infection with LM-GP61(c) or IAV-GP61 (e) as described in a. The presented data are representative of two independent experiments (n=4 in b-e). f, Experimental setup and gating strategy for flow cytometry. Human naïve (Tnaive), central memory (TCM), and effector memory (TEM) CD4+ T cells isolated from PBMCs of healthy human donors were treated with Torin-1, Rapamycin, or vehicle, then analyzed via flow cytometry. g, h, Bodipy C11 intensity (g) and percentage of dead cells (Live/Dead+) in enumerated CD4+ T cell subsets in PBMCs after treatment of Torin-1, Rapamycin, or vehicle as described in f. The presented data are representative of two independent experiments (n=5 in g-h). Data are analyzed by paired two-tailed t-test in b-e. Data are presented as mean values ± SD and analyzed by unpaired two-tailed t-test in g-h.

Source data

Extended Data Fig. 8 GSEA reveals hyperactive mitochondrial ROS generation in mTORC2-deficienct memory CD4+ T cells.

a, b, c, GSEA plots for GO term about mitochondrial organization and respiratory chain complex assembly (a), cellular regulation of reactive oxygen species metabolic process (b), and cellular oxidative stress (c) in sorted memory TH1 cells (CD25GITRCD44highCXCR5CD4+) and memory TFH cells (CD25GITRCD44highCXCR5+CD4+) from either CD4Cre-Rictorfl/fl (Rictor−/−) or Rictorfl/fl mice at day 60 post-infection of LCMV Arm+. NES, normalized enrichment score; FDR, false discovery rates.

Extended Data Fig. 9 Blockade of IL-7 signaling results in mitochondrial dysfunction in memory CD4+ T cells.

a, Experimental setup is described as in Extended Data Fig. 6. TMRE, MitoTracker, and MitoSOX intensity on memory SMARTA cells in spleens of recipients at day 60 post LCMV Arm+ infection in the context of IL-7 signaling blockade (αIL-7-mAb) or not (Control). The presented data are representative of two independent experiments (n=4 for αIL-7-mAb, or 5 for Control). Data are presented as mean values ± SD and analyzed by unpaired two-tailed t-test in a.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed immunoblots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed immunoblots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed immunoblots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed immunoblots.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed immunoblots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Tian, Q., Hao, Y. et al. The kinase complex mTORC2 promotes the longevity of virus-specific memory CD4+ T cells by preventing ferroptosis. Nat Immunol 23, 303–317 (2022). https://doi.org/10.1038/s41590-021-01090-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01090-1

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