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:

Selenium–GPX4 axis protects follicular helper T cells from ferroptosis

An Author Correction to this article was published on 04 October 2021

This article has been updated

Abstract

Follicular helper T (TFH) cells are a specialized subset of CD4+ T cells that essentially support germinal center responses where high-affinity and long-lived humoral immunity is generated. The regulation of TFH cell survival remains unclear. Here we report that TFH cells show intensified lipid peroxidation and altered mitochondrial morphology, resembling the features of ferroptosis, a form of programmed cell death that is driven by iron-dependent accumulation of lipid peroxidation. Glutathione peroxidase 4 (GPX4) is the major lipid peroxidation scavenger and is necessary for TFH cell survival. The deletion of GPX4 in T cells selectively abrogated TFH cells and germinal center responses in immunized mice. Selenium supplementation enhanced GPX4 expression in T cells, increased TFH cell numbers and promoted antibody responses in immunized mice and young adults after influenza vaccination. Our findings reveal the central role of the selenium–GPX4–ferroptosis axis in regulating TFH homeostasis, which can be targeted to enhance TFH cell function in infection and following vaccination.

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: Mouse TFH cells show features of ferroptosis.
Fig. 2: Human TFH cells are vulnerable to ferroptosis.
Fig. 3: GPX4 in T cells supports TFH cell homeostasis and humoral immune responses.
Fig. 4: TFH–BGC interactions exaggerate lipid peroxidation and promote TFH ferroptosis.
Fig. 5: Tuning ferroptosis regulates TFH cell function.
Fig. 6: Selenium supplementation mitigates ferroptosis in TFH cells and boosts humoral immunity.
Fig. 7: Dietary selenium supplementation enhances TFH response and humoral immunity following influenza vaccination in young adults.

Similar content being viewed by others

Data availability

All data are provided in the article and its supplementary files or from the corresponding author upon reasonable request. Source data are provided with this paper.

Change history

References

  1. Victora, G. D. & Nussenzweig, M. C. Germinal centers. Annu Rev. Immunol. 30, 429–457 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Vinuesa, C. G., Linterman, M. A., Yu, D. & MacLennan, I. C. Follicular helper T cells. Ann. Rev. Immunol. https://doi.org/10.1146/annurev-immunol-041015-055605 (2016).

  3. Crotty, S. T. Follicular helper cell biology: a decade of discovery and diseases. Immunity 50, 1132–1148 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Deng, J., Wei, Y., Fonseca, V. R., Graca, L. & Yu, D. T follicular helper cells and T follicular regulatory cells in rheumatic diseases. Nat. Rev. Rheumatol. 15, 475–490 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. He, J. et al. Low-dose interleukin-2 treatment selectively modulates CD4(+) T cell subsets in patients with systemic lupus erythematosus. Nat. Med. 22, 991–993 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Edner, N. M. et al. Follicular helper T cell profiles predict response to costimulation blockade in type 1 diabetes. Nat. Immunol. 21, 1244–1255 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bentebibel, S. E. et al. Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Sci. Transl. Med. 5, 176ra132 (2013).

    Article  Google Scholar 

  8. Koutsakos, M. et al. Circulating TFH cells, serological memory, and tissue compartmentalization shape human influenza-specific B cell immunity. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aan8405 (2018).

  9. Jun, D. et al. The metabolic hormone leptin promotes the function of TFH cells and supports vaccine responses. Nat. Commun. https://doi.org/10.1038/s41467-021-23220-x (2021).

  10. Linterman, M. A. & Hill, D. L. Can follicular helper T cells be targeted to improve vaccine efficacy? F1000Res https://doi.org/10.12688/f1000research.7388.1 (2016).

  11. Peperzak, V., Vikstrom, I. B. & Tarlinton, D. M. Through a glass less darkly: apoptosis and the germinal center response to antigen. Immunol. Rev. 247, 93–106 (2012).

    Article  PubMed  Google Scholar 

  12. Hildeman, D. A. et al. Reactive oxygen species regulate activation-induced T cell apoptosis. Immunity 10, 735–744 (1999).

    Article  CAS  PubMed  Google Scholar 

  13. Seman, M. et al. NAD-induced T cell death: ADP-ribosylation of cell surface proteins by ART2 activates the cytolytic P2X7 purinoceptor. Immunity 19, 571–582 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Sena, L. A. et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 38, 225–236 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mak, T. W. et al. Glutathione primes T cell metabolism for inflammation. Immunity 46, 1089–1090 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. 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 

  17. 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 

  18. Li, J. et al. Ferroptosis: past, present and future. Cell Death Dis. 11, 88 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Seiler, A. et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab. 8, 237–248 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Ingold, I. et al. Selenium utilization by GPX4 Is required to prevent Hydroperoxide-Induced ferroptosis. Cell 172, 409–422 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Zhang, N., Hartig, H., Dzhagalov, I., Draper, D. & He, Y. W. The role of apoptosis in the development and function of T lymphocytes. Cell Res. 15, 749–769 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. 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 

  23. 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 

  24. Ayala, A., Munoz, M. F. & Arguelles, S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid. Med. Cell Longev. 2014, 360438 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Yu, D. et al. Roquin represses autoimmunity by limiting inducible T-cell co-stimulator messenger RNA. Nature 450, 299–303 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Liu, D. et al. T-B-cell entanglement and ICOSL-driven feed-forward regulation of germinal centre reaction. Nature 517, 214–218 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Sakaguchi, S. et al. Regulatory T cells and human disease. Annu. Rev. Immunol. 38, 541–566 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Leong, Y. A. et al. CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat. Immunol. 17, 1187–1196 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Gerriets, V. A. et al. Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J. Clin. Invest. 125, 194–207 (2015).

    Article  PubMed  Google Scholar 

  30. Magtanong, L. et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem. Biol. 26, 420–432 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Devadas, S., Zaritskaya, L., Rhee, S. G., Oberley, L. & Williams, M. S. Discrete generation of superoxide and hydrogen peroxide by T cell receptor stimulation: selective regulation of mitogen-activated protein kinase activation and fas ligand expression. J. Exp. Med. 195, 59–70 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. DiToro, D. et al. Differential IL-2 expression defines developmental fates of follicular versus nonfollicular helper T cells. Science https://doi.org/10.1126/science.aao2933 (2018).

  33. Victora, G. D. et al. Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell 143, 592–605 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Baumjohann, D. et al. Persistent antigen and germinal center B cells sustain T follicular helper cell responses and phenotype. Immunity 38, 596–605 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Reinhardt, R. L., Liang, H. E. & Locksley, R. M. Cytokine-secreting follicular T cells shape the antibody repertoire. Nat. Immunol. 10, 385–393 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cannons, J. L. et al. Optimal germinal center responses require a multistage T cell:B cell adhesion process involving integrins, SLAM-associated protein, and CD84. Immunity 32, 253–265 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li, W. et al. Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment after heart transplantation. J. Clin. Invest. 129, 2293–2304 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Alim, I. et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell 177, 1262–1279 (2019).

    Article  CAS  PubMed  Google Scholar 

  39. He, J. et al. Circulating precursor CCR7loPD-1hi CXCR5+ CD4+ T cells indicate TFH cell activity and promote antibody responses upon antigen reexposure. Immunity 39, 770–781 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Hagan, T. et al. Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans. Cell 178, 1313–1328 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Goodwin, K., Viboud, C. & Simonsen, L. Antibody response to influenza vaccination in the elderly: a quantitative review. Vaccine 24, 1159–1169 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Nagata, S. & Tanaka, M. Programmed cell death and the immune system. Nat. Rev. Immunol. 17, 333–340 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Proietti, M. et al. ATP-gated ionotropic P2X7 receptor controls follicular T helper cell numbers in Peyer’s patches to promote host-microbiota mutualism. Immunity 41, 789–801 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Faliti, C. E. et al. P2X7 receptor restrains pathogenic TFH cell generation in systemic lupus erythematosus. J. Exp. Med. 216, 317–336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kunzli, M. et al. Long-lived T follicular helper cells retain plasticity and help sustain humoral immunity. Sci. Immunol. https://doi.org/10.1126/sciimmunol.aay5552 (2020).

  46. Muri, J., Thut, H., Bornkamm, G. W. & Kopf, M. B1 and marginal zone B cells but not follicular B2 cells require Gpx4 to prevent lipid peroxidation and ferroptosis. Cell Rep. 29, 2731–2744 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. Kusam, S., Toney, L. M., Sato, H. & Dent, A. L. Inhibition of TH2 differentiation and GATA-3 expression by BCL-6. J. Immunol. 170, 2435–2441 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Yu, D. et al. The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity 31, 457–468 (2009).

    Article  CAS  PubMed  Google Scholar 

  49. Mahnke, K., Qian, Y., Knop, J. & Enk, A. H. Induction of CD4+/CD25+ regulatory T cells by targeting of antigens to immature dendritic cells. Blood 101, 4862–4869 (2003).

    Article  CAS  PubMed  Google Scholar 

  50. Bonifaz, L. C. et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J. Exp. Med. 199, 815–824 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Imaging and Cytometry Facility and the Biomolecular Research Facility at the John Curtin School of Medical Research and the Biomolecular Research Facility at Translational Research Institute for technical support; the Centre of Advanced Microscopy at the Australian National University for the scientific and technical assistance for TEM; R. Steptoe and Y. Zhan for the hybridoma cell line; D. Gray for Foxp3-GFP reporter mice; S. Ming Man for Casp1/11−/− mice; J. Adams for Bcl2-transgenic mice; H. Wu and Q. Li for tissue imaging and E. Bartlett for reading the manuscript. This study was supported by Australian National Health and Medical Research Council project GNT1085509; the Bellberry-Viertel Senior Medical Research fellowship; National Natural Science Foundation of China grants 81630024, 81920108011 and 82071792; Health Commission of Hubei Province Scientific Research Project WJ2019H136 and WJ2019H137; Natural Science Foundation of Shandong Province ZR2020ZD41 and ZR2019BB080; Taishan Scholars Program of Shandong Province; and a China Scholarship Council scholarship 201706160045.

Author information

Authors and Affiliations

Authors

Contributions

Y. Yao. planned and performed most experiments with major support from Z.A.C. and H.Z.. Y.B.W. performed plasma selenium levels analysis. C.L.C., M.Z., Z.Q.L., M.Z.Z., C.Q., Y.M.W., Y.K.D., N. Wang, C.S., C.L.G., H.B.S., H.L.Y. and W.T.Z. participated in the human trial. J.V. helped with imaging. N. Wu. and A.H. helped with flow cytometry. H.S.O., H.W., F.V.M., Z.H.Y., Y. Yang and P.C.Z. participated in animal experiments; Q.X.Z., J.D. and N.S. helped with human cell sorting and cell culture. N.Q.W. participated in animal experiments and helped with data analysis. W.H.Z., J.B.H., Y.X., J.F., D.M.L., M.X. and L.Y. participated in discussion and human study. D.Y. and Z.L. designed the study and supervised the project.

Corresponding authors

Correspondence to Zheng Liu or Di Yu.

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.

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

Extended data

Extended Data Fig. 1 The cell death of TFH cells is not regulated by apoptosis or pyroptosis.

a, b, As indicated by the schematic of bone marrow (BM) reconstitution to generate chimeric mice, indicated mixtures of BM cells were transferred into lethally irradiated Rag1–/– recipients. After 6 weeks, chimeric mice were subcutaneously immunized with NP-OVA in Alum. b, At day 14 post immunisation, frequencies of TFH and BGC cells derived from WT or transgenic were analysed by flow cytometry (n = 5 for control and n = 6 for Bcl-2-Tg and Casp1/11-KO). c, C57/BL6/J mice were subcutaneously immunized with NP-OVA in Alum. Cell size of TFH and non-TFH cells at day 7 post immunisation was analysed by flow cytometry (n = 6). d, Congenic marked C57/BL6/J mice were adoptively transferred of OT-II cells and then intraperitoneally immunised with OVA in Alum. At day 7 post immunisation, cell size of TFH and non-TFH OT-II cells was analysed by flow cytometry (n = 6). e, Mice were intraperitoneally immunised with SRBC, TFH and non-TFH cells were sorted at day 8 post immunisation. Lipid ROS production in TFH and non-TFH cells was analysed by flow cytometry using C11-BODIPY (n = 5). f, Flow cytometric analysis of lipid ROS production in purified tonsillar TFH and non-TFH cells (n = 4). Each dot represents one individual. g, Purified naïve CD4+ T, non-TFH, and TFH cells from human tonsils were stimulated with αCD3/CD28 and treated with different doses of RSL-3 or H2O2 for 12 h. Frequencies of Annexin V7-AAD viable cells were assessed by flow cytometry (n = 3). Data are representative of three independent experiments. For b, g, data are expressed as mean ± SEM. Data are analysed by two-sided Student’s t-test (b), paired-sample t-test (c-f), or one-way ANOVA (g). For g, bonferroni correction was used to adjust the significance level by using a value of 0.017 for each comparison. Data are representative of three (ce) experiments with at least three mice per group.

Source data

Extended Data Fig. 2 Characterisation of GPX4-deficient T cells in mice.

a, GPX4 protein expression in splenic CD4+ cells from Cd4-CreNegGpx4flox/flox (WT) and Cd4-CreGpx4flox/flox (T-KO) mice were analysed by flow cytometry (n = 5). b, c, Naïve CD4+ T cells were purified from WT and T-KO mice and cultured in 2-Mercaptoethanol-deprived culture medium. The frequencies of Annexin V7-AAD viable cells (b) and lipid ROS production (c) were assessed by flow cytometry at indicated time points (n = 3). Data are representative of three independent experiments. d, Naïve CD4+ T cells from WT or T-KO mice were cultured for 2 h and imaged by transmission electron microscopy. Blue arrow indicates mitochondria from WT mice, and the red arrow indicates shrunken and damaged mitochondria from T-KO mice. Scale bars, 1 μm. The area of each mitochondrion was calculated using ImageJ. Each dot represents one mitochondrion. Box plots shows medians and the extent from the 25th to 75th percentiles, with error bars covering the data range. e-g, Flow cytometric analysis of T cell subsets of WT and T-KO mice at 8-weeks old. Total numbers of thymocytes (e) and splenic cells (f) were quantified and CD44 and CD62L expression on splenic CD4+ T cells (g) were analysed (n = 5). hl, WT and T-KO mice were subcutaneously immunised with NP-OVA in Alum (n = 5 for WT and n = 3 for T-KO). h, At day 7 post immunisation, frequencies of CD44+, CD69+, and Ki-67+ cells in CD4+ T cells of draining popliteal lymph nodes were analysed by flow cytometry. i, TFR cells were analysed at day 7 and 14 after immunisation. Gata3 (j) and Rorγt (k) expression in CD4+Foxp3 cells were analysed by flow cytometry. l, TREG cells were analysed at day 7 and 14 after immunisation. Numbers indicate the frequency of cells in the gated region. For a, el, each dot represents one mouse. Data are presented as mean ± SEM and analysed by two-sided Student’s t-test (a, el). For a, el, data are representative of two independent experiments with at least three mice per group.

Source data

Extended Data Fig. 3 Vitamin E rescues TFH cell from ferroptosis induced by GPX4 deletion.

ad, Cd28–/– mice were adoptively transferred of WT or T-KO OT-II cells and then intraperitoneally immunised with OVA in Alum. At day 10 post immunisation, recipient CD8+ T (a) and Foxp3+ TREG (b) cells and OT-II TREG (c) and TFR (d) cells in spleen were analysed by flow cytometry (n = 5). e, f, Cd28–/– mice were adoptively transferred OT-II cells from T-KO mice, subcutaneously immunised with NP-OVA in Alum and treated every 2 days with control or Vitamin E (100 mg/kg) from day 1–9 post immunisation. f, At day 10, frequencies and numbers of CXCR5+Bcl-6+ TFH in popliteal lymph nodes were analysed by flow cytometry (n = 5). Numbers indicate the frequency of cells in the gated region. Each dot represents one mouse. Data are presented as mean ± SEM and analysed by two-sided Student’s t-test. Data are representative of two experiments with at least five mice per group.

Source data

Extended Data Fig. 4 High cytosolic ROS expression in TFH cells.

a, Flow cytometric analysis of cytosolic ROS production in purified CD44+CD25CXCR5highPD-1high TFH and CD44+CD25CXCR5PD-1 non-TFH cells from spleen of SRBC-immunised mice at day 8 post immunisation using CM-H2DCFDA (n = 3). Each dot represents one mouse. b, Comparison of cytosolic ROS levels between circulating CD25–/lowCD127–/+CD45RACXCR5+ TFH and CD25–/lowCD127–/+CD45RACXCR5 non-TFH effector cells in human peripheral blood (n = 5). Each dot represents a single blood donor. c, Comparison of cytosolic ROS levels between CD45RACXCR5high TFH and CD45RACXCR5 non-TFH effector cells in human tonsils (n = 10). Each dot represents a single tonsil donor. Data are analysed by paired-sample t-test.

Source data

Extended Data Fig. 5 Comparing the levels of ferroptosis-related factors in TFH and non-TFH cells.

a, Gene expression related to cytosolic and lipid ROS and ferroptosis in TFH and non-TFH cells in human and mice. Data were obtained by analysing published RNA-seq data. b, Glutathione (GSH) levels in purified tonsillar CD4+CD19CD25CD45RACXCR5high TFH and CD4+CD19CD25CD45RACXCR5 non-TFH cells were detected by ELISA (n = 3). c, Total Fe levels in purified tonsillar TFH and non-TFH cells were analysed by ICP-AES (n = 4). d, Fe2+ levels in tonsillar TFH and non-TFH cells were analysed by flow cytometry using FerroOrange (n = 5). e, Total SFAs and the ratio of total PUFAs/total MUFA in purified tonsillar TFH and non-TFH cells were analysed by GC-MS (n = 4). For b-e, each dot represents a single tonsil donor. Data are analysed by paired-sample t-test.

Source data

Extended Data Fig. 6 Intensified TCR stimulation induces high ROS production in TFH cells.

a, Purified naïve CD4+ T cells were stimulated with αCD3/CD28 in 2-Mercaptoethanol-deprived culture medium, and cytosolic ROS production was assessed by flow cytometry at 24 and 72 h after incubation (n = 3 for each dose). Data are represented as fold change by normalizing to no stimulation and representative of three independent experiments. b, c, Purified CD4+CD19CD25CD45RACXCR5high TFH cells from human tonsils were cultured with indicated cytokines or stimulators in the absence (b) or presence (c) of 3 µM RSL-3 in 2-Mercaptoethanol-deprived culture medium for 12 h. Lipid ROS levels and frequencies of 7AAD viable cells were analysed by flow cytometry (n = 3). Data are representative of three independent experiments. d, Mice were subcutaneously immunised with NP-OVA in Alum. Flow cytometric analysis of cytosolic ROS production in CD44+CD25CD19CXCR5highPD-1high TFH and FAS+GL-7+B220+ BGC cells at day 10 post immunisation (n = 6). Each dot represents one mouse. Data are representative of two independent experiments with at least five mice per group. Data are presented as mean ± SEM (ac) and analysed by two-sided Student’s t-test (ac) or paired-sample t-test (d).

Source data

Extended Data Fig. 7 Ferrostatin-1 rescues TFH cells in mice immunised with NP-OVA.

ad, Cd4-CreGpx4wt/wt (WT) and Cd4-CreGpx4wt/flox (T-HET) mice were subcutaneously immunised with NP-OVA in Alum. At day 14 post immunisation, the frequencies of CD3+ and CD4+ T cells (a), CD44, CD69, and Ki-67 expression by CD4+ T cells (b), frequencies of Foxp3+ TREG and Bcl-6+Foxp3+ TFR cells (c), and IFNγ, IL-4, and IL-17A production by CD44+CD25CXCR5PD-1 non-TFH cells (d) were analysed by flow cytometry (n = 5). eg, As the schematic of experiment design (e), mice were adoptively transferred OT-II cells, intraperitoneally immunised with NP-OVA in Alum and treated daily with DMSO (control) or Ferrostatin-1 (10 mg/kg) from day 7–9 post immunisation. At day 10, frequencies of donor OT-II cells in total CD4+ T cells (f) and frequencies and numbers of TFH and non-TFH OT-II cells (g) in spleen were analysed by flow cytometry (n = 5). hl, C57/BL6/J mice were intraperitoneally immunised with NP-OVA in Alum and treated every 2 days with DMSO (control) or Ferrostatin-1 (2 mg/kg) s.c. from day 5–13 post immunization. At day 14, CD44+Foxp3CXCR5 non-TFH cells (i), Ki-67 expression by non-TFH cells (j), IFNγ, IL-4, and IL-17A production by non-TFH cells (k), Foxp3+ TREG cells, and Bcl-6+Foxp3+ TFR cells (l) in draining popliteal lymph nodes were analysed by flow cytometry (n = 6). Each dot represents one mouse. Data are presented as mean ± SEM and analysed by two-sided Student’s t-test. Data are representative of two experiments with at least four mice per group.

Source data

Extended Data Fig. 8 Selenium supplementation promotes TFH cell responses in mice.

ag, C57/BL6/J mice were fed with water containing methionine (Met, 1 mg/L) or selenomethionine (Se-Met, 1 mg/L) one month before immunisation with NP-OVA in Alum. b, Selenoprotein P (SelP) and glutathione peroxidase 3 (GPX3) levels in serum were measured by ELISA at indicated time points (n = 6). At day 7 and 14 post immunisation, Ki-67 expression in TFH cells (c), Annexin V and cytosolic ROS staining of non-TFH (d), IFNγ, IL-4, and IL-17A production by non-TFH cells (e), Foxp3+ TREG cells (f), and CD44+Foxp3+Bcl-6+ TFR cells (g) in popliteal lymph nodes were analysed by flow cytometry (n = 6). h-l, C57/BL6/J mice were fed with adequate selenium (0.15 mg Se/kg) or high selenium (1 mg Se/kg) diets one month before the immunisation with NP-OVA in Alum. i, Selenoprotein P (SelP) levels in serum were measured by ELISA at indicated time points (n = 5). At day 14 post immunisation, TFH (j) and BGC cells (k) in popliteal lymph nodes were analysed by flow cytometry (n = 5). l, Serum NP23- and NP2-binding IgG1 were measured by ELISA (n = 5). Numbers indicate frequency of cells in gated region. Each dot represents one mouse. Data are presented as mean ± SEM and analysed by two-sided Student’s t-test. Data are representative of three experiments with at least five mice per group.

Source data

Extended Data Fig. 9 The gating strategy to analyse peripheral CD4+ T cell subsets in human blood.

Within viable CD3+CD8CD4+ T cells, TREG cells were defined as CD25highCD127low cells, circulating TFH cells as CD25–/lowCD45RACXCR5+ cells, activated TFH cells as ICOShighPD-1high or PD-1high CCR7low TFH cells, activated non-TFH cells as CXCR5ICOShighPD-1high cells, TH1 cells as CD25–/lowCD45RACXCR3+CCR6CCR7lowCCR4 cells, TH2 cells as CD25–/lowCD45RACXCR3CCR6CCR7lowCCR4+ cells, and TH17 cells as CD25–/lowCD45RACXCR3CCR6+CCR7CCR4+ cells.

Extended Data Fig. 10 Changes in circulating T cell subsets after selenium supplementation.

a, Young healthy subjects were enrolled and randomised into selenium supplementation (200 μg/day) and control groups. Thirty days after the start of supplementation, all subjects received seasonal influenza vaccination, and peripheral blood was collected at baseline (day −30), day 0 (before vaccination), day 7 (day 7 post vaccination), and day 30 (day 30 post vaccination). b, Frequency of circulating CXCR5+CCR7lowPD-1high TFH cells in total CD4+ T cells were analysed by flow cytometry (n = 29 for control and n = 28 for selenium). Changes of TFH cells at the indicated time points were normalised to the baseline and represented as fold changes. ce, Frequencies and fold changes normalised to the baseline of indicated populations were analysed by flow cytometry (n = 29 for control and n = 28 for selenium). f, g, IL-21 production by circulating CD25–/lowCD45RACXCR5+ TFH cells (f) and IFNγ, IL-4, and IL-17A production by CD25–/lowCD45RACXCR5 non-TFH cells (g) were analysed by flow cytometry (n = 23 for control and n = 24 for selenium). Numbers indicate the frequency of cells in the gated region. Each dot represents one individual. Data are presented as median and interquartile range and analysed by Mann-Whitney U test.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 7

Unprocessed immunoblots.

Source Data Extended Data Fig. 1

Statistical Source Data.

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. 8

Statistical Source Data.

Source Data Extended Data Fig. 10

Statistical Source Data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yao, Y., Chen, Z., Zhang, H. et al. Selenium–GPX4 axis protects follicular helper T cells from ferroptosis. Nat Immunol 22, 1127–1139 (2021). https://doi.org/10.1038/s41590-021-00996-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00996-0

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