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:

Aging-associated HELIOS deficiency in naive CD4+ T cells alters chromatin remodeling and promotes effector cell responses

Abstract

Immune aging combines cellular defects in adaptive immunity with the activation of pathways causing a low-inflammatory state. Here we examined the influence of age on the kinetic changes in the epigenomic and transcriptional landscape induced by T cell receptor (TCR) stimulation in naive CD4+ T cells. Despite attenuated TCR signaling in older adults, TCR activation accelerated remodeling of the epigenome and induced transcription factor networks favoring effector cell differentiation. We identified increased phosphorylation of STAT5, at least in part due to aberrant IL-2 receptor and lower HELIOS expression, as upstream regulators. Human HELIOS-deficient, naive CD4+ T cells, when transferred into human-synovium-mouse chimeras, infiltrated tissues more efficiently. Inhibition of IL-2 or STAT5 activity in T cell responses of older adults restored the epigenetic response pattern to the one seen in young adults. In summary, reduced HELIOS expression in non-regulatory naive CD4+ T cells in older adults directs T cell fate decisions toward inflammatory effector cells that infiltrate tissue.

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: Chromatin accessibility changes induced by TCR stimulation of different signaling strengths.
Fig. 2: Dissociation of TCR-induced signaling and chromatin accessibility changes in naive CD4+ T cells from older adults.
Fig. 3: Accelerated chromatin changes in T cells from older adults are driven by the STAT5 pathway.
Fig. 4: Age-associated expansion of naive CD4+ T cell subsets with increased activation of the STAT5 pathway.
Fig. 5: CD25 upregulation is induced by a loss of HELIOS with age.
Fig. 6: Forced HELIOS degradation in human naive CD4+ T cells induces tissue inflammation in vivo.
Fig. 7: Inhibition of IL-2-STAT5 signaling rewires activated naive CD4+ T cells from older individuals to a younger epigenetic response pattern.

Similar content being viewed by others

Data availability

Raw sequencing data have been deposited in SRA with the BioProject accession no. PRJNA757466. Raw experimental data will be provided by the corresponding author upon request. Source data are provided with this paper.

References

  1. O’Driscoll, M. et al. Age-specific mortality and immunity patterns of SARS-CoV-2. Nature 590, 140–145 (2021).

    Article  Google Scholar 

  2. Rydyznski Moderbacher, C. et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell 183, 996–1012 (2020).

    Article  CAS  Google Scholar 

  3. Gustafson, C. E., Kim, C., Weyand, C. M. & Goronzy, J. J. Influence of immune aging on vaccine responses. J. Allergy Clin. Immunol. 145, 1309–1321 (2020).

    Article  CAS  Google Scholar 

  4. Weng, N. P. Aging of the immune system: how much can the adaptive immune system adapt? Immunity 24, 495–499 (2006).

    Article  CAS  Google Scholar 

  5. Furman, D. et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 25, 1822–1832 (2019).

    Article  CAS  Google Scholar 

  6. Goronzy, J. J. & Weyand, C. M. Mechanisms underlying T cell ageing. Nat. Rev. Immunol. 19, 573–583 (2019).

    Article  CAS  Google Scholar 

  7. Mittelbrunn, M. & Kroemer, G. Hallmarks of T cell aging. Nat. Immunol. 22, 687–698 (2021).

    Article  CAS  Google Scholar 

  8. Whiting, C. C. et al. Large-scale and comprehensive immune profiling and functional analysis of normal human aging. PLoS ONE 10, e0133627 (2015).

    Article  Google Scholar 

  9. Qi, Q. et al. Diversity and clonal selection in the human T-cell repertoire. Proc. Natl Acad. Sci. USA 111, 13139–13144 (2014).

    Article  CAS  Google Scholar 

  10. Fülöp, T. et al. Age-related impairment of p56lck and ZAP-70 activities in human T lymphocytes activated through the TcR/CD3 complex. Exp. Gerontol. 34, 197–216 (1999).

    Article  Google Scholar 

  11. Li, G. et al. Decline in miR-181a expression with age impairs T cell receptor sensitivity by increasing DUSP6 activity. Nat. Med. 18, 1518–1524 (2012).

    Article  CAS  Google Scholar 

  12. Pereira, B. I. et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nat. Immunol. 21, 684–694 (2020).

    Article  CAS  Google Scholar 

  13. Kim, C. et al. Activation of miR-21-regulated pathways in immune aging selects against signatures characteristic of memory T cells. Cell Rep. 25, 2148–2162 (2018).

    Article  CAS  Google Scholar 

  14. Goronzy, J. J. & Weyand, C. M. Successful and maladaptive T cell aging. Immunity 46, 364–378 (2017).

    Article  CAS  Google Scholar 

  15. Elyahu, Y. et al. Aging promotes reorganization of the CD4 T cell landscape toward extreme regulatory and effector phenotypes. Sci. Adv. 5, eaaw8330 (2019).

    Article  CAS  Google Scholar 

  16. Almanzar, N. et al. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature 583, 590–595 (2020).

    Article  CAS  Google Scholar 

  17. Mogilenko, D. A. et al. Comprehensive profiling of an aging immune system reveals clonal GZMK+CD8+ T cells as conserved hallmark of inflammaging. Immunity 54, 99–115 (2021).

    Article  CAS  Google Scholar 

  18. den Braber, I. et al. Maintenance of peripheral naive T cells is sustained by thymus output in mice but not humans. Immunity 36, 288–297 (2012).

    Article  Google Scholar 

  19. Zhang, H., Weyand, C. M. & Goronzy, J. J. Hallmarks of the aging T-cell system. FEBS J. 288, 7123–7142 (2021).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Kurachi, M. et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat. Immunol. 15, 373–383 (2014).

    Article  CAS  Google Scholar 

  22. Davenport, M. P., Smith, N. L. & Rudd, B. D. Building a T cell compartment: how immune cell development shapes function. Nat. Rev. Immunol. 20, 499–506 (2020).

    Article  CAS  Google Scholar 

  23. Roychoudhuri, R. et al. BACH2 regulates CD8+ T cell differentiation by controlling access of AP-1 factors to enhancers. Nat. Immunol. 17, 851–860 (2016).

    Article  CAS  Google Scholar 

  24. 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  Google Scholar 

  25. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789 (2019).

    Article  CAS  Google Scholar 

  26. Pulko, V. et al. Human memory T cells with a naive phenotype accumulate with aging and respond to persistent viruses. Nat. Immunol. 17, 966–975 (2016).

    Article  CAS  Google Scholar 

  27. Simeonov, D. R. et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 549, 111–115 (2017).

    Article  CAS  Google Scholar 

  28. Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).

    Article  CAS  Google Scholar 

  29. Rosenbloom, K. R. et al. ENCODE data in the UCSC genome browser: year 5 update. Nucleic Acids Res. 41, D56–D63 (2013).

    Article  CAS  Google Scholar 

  30. Kim, H. J. et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science 350, 334–339 (2015).

    Article  CAS  Google Scholar 

  31. Thornton, A. M. et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J. Immunol. 184, 3433–3441 (2010).

    Article  CAS  Google Scholar 

  32. Yang, Z. et al. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci. Transl. Med. 8, 331ra338 (2016).

    Article  Google Scholar 

  33. Wang, E. S. et al. Acute pharmacological degradation of Helios destabilizes regulatory T cells. Nat. Chem. Biol. 17, 711–717 (2021).

    Article  Google Scholar 

  34. Liao, W., Lin, J.-X., Wang, L., Li, P. & Leonard, W. J. Modulation of cytokine receptors by IL-2 broadly regulates differentiation into helper T cell lineages. Nat. Immunol. 12, 551–559 (2011).

    Article  CAS  Google Scholar 

  35. Jin, J. et al. Activation of mTORC1 at late endosomes misdirects T cell fate decision in older individuals. Sci. Immunol. 6, 791–791 (2021).

    Article  Google Scholar 

  36. Pekalski, M. L. et al. Postthymic expansion in human CD4 naive T cells defined by expression of functional high-affinity IL-2 receptors. J. Immunol. 190, 2554–2566 (2013).

    Article  CAS  Google Scholar 

  37. Kalia, V. et al. Prolonged interleukin-2Rα expression on virus-specific CD8+ T cells favors terminal-effector differentiation in vivo. Immunity 32, 91–103 (2010).

    Article  CAS  Google Scholar 

  38. Lindahl, L. M. et al. STAT5 induces miR-21 expression in cutaneous T cell lymphoma. Oncotarget 7, 45730–45744 (2016).

    Article  Google Scholar 

  39. Johnston, R. J., Choi, Y. S., Diamond, J. A., Yang, J. A. & Crotty, S. STAT5 is a potent negative regulator of TFH cell differentiation. J. Exp. Med. 209, 243–250 (2012).

    Article  CAS  Google Scholar 

  40. Canale, F. P. et al. CD39 expression defines cell exhaustion in tumor-infiltrating CD8+ T cells. Cancer Res. 78, 115–128 (2018).

    Article  CAS  Google Scholar 

  41. Cao, W. et al. Ecto-NTPDase CD39 is a negative checkpoint that inhibits follicular helper cell generation. J. Clin. Invest. 130, 3422–3436 (2020).

    Article  CAS  Google Scholar 

  42. Fang, F. et al. Expression of CD39 on activated T cells impairs their survival in older individuals. Cell Rep. 14, 1218–1231 (2016).

    Article  CAS  Google Scholar 

  43. Hetemäki, I. et al. Loss-of-function mutation in IKZF2 leads to immunodeficiency with dysregulated germinal center reactions and reduction of MAIT cells. Sci. Immunol. 6, 3454 (2021).

    Article  Google Scholar 

  44. Serre, K. et al. Helios is associated with CD4 T cells differentiating to T helper 2 and follicular helper T cells in vivo independently of Foxp3 expression. PLoS ONE 6, e20731 (2011).

    Article  CAS  Google Scholar 

  45. Shahin, T. et al. Germline biallelic mutation affecting the transcription factor Helios causes pleiotropic defects of immunity. Sci. Immunol. 6, 3981 (2021).

    Article  Google Scholar 

  46. Baine, I., Basu, S., Ames, R., Sellers, R. S. & Macian, F. Helios induces epigenetic silencing of IL2 gene expression in regulatory T cells. J. Immunol. 190, 1008 (2013).

    Article  CAS  Google Scholar 

  47. Sheng, W. et al. STAT5 programs a distinct subset of GM-CSF-producing T helper cells that is essential for autoimmune neuroinflammation. Cell Res. 24, 1387–1402 (2014).

    Article  CAS  Google Scholar 

  48. Fu, Y. et al. STAT5 promotes accessibility and is required for BATF-mediated plasticity at the Il9 locus. Nat. Commun. 11, 1–16 (2020).

    Article  Google Scholar 

  49. Hu, B. et al. Transcription factor networks in aged naive CD4 T cells bias lineage differentiation. Aging Cell 18, e12957 (2019).

    Article  Google Scholar 

  50. 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, 21.29.21–21.29.29 (2015).

    Article  Google Scholar 

  51. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    Article  CAS  Google Scholar 

  52. Hansen, K. D., Irizarry, R. A. & Wu, Z. Removing technical variability in RNA-seq data using conditional quantile normalization. Biostatistics 13, 204–216 (2012).

    Article  Google Scholar 

  53. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  Google Scholar 

  54. Yu, G., Wang, L.-G. & He, Q.-Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  Google Scholar 

  55. Granja, J. M. et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat. Genet. 53, 403–411 (2021).

    Article  CAS  Google Scholar 

  56. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  Google Scholar 

  57. 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).

    Article  CAS  Google Scholar 

  58. Abbas, A. R. et al. Immune response in silico (IRIS): immune-specific genes identified from a compendium of microarray expression data. Genes Immun. 6, 319–331 (2005).

    Article  CAS  Google Scholar 

  59. Kolmykov, S. et al. GTRD: an integrated view of transcription regulation. Nucleic Acids Res. 49, D104–D111 (2021).

    Article  CAS  Google Scholar 

  60. Marzec, M. et al. Differential effects of interleukin-2 and interleukin-15 versus interleukin-21 on CD4+ cutaneous T-cell lymphoma cells. Cancer Res. 68, 1083–1091 (2008).

    Article  CAS  Google Scholar 

  61. Wen, Z. et al. N-myristoyltransferase deficiency impairs activation of kinase AMPK and promotes synovial tissue inflammation. Nat. Immunol. 20, 313–325 (2019).

    Article  CAS  Google Scholar 

  62. Wu, B. et al. Mitochondrial aspartate regulates TNF biogenesis and autoimmune tissue inflammation. Nat. Immunol. 22, 1551–1562 (2021).

    Article  CAS  Google Scholar 

  63. Moskowitz, D. M. et al. Epigenomics of human CD8 T cell differentiation and aging. Sci. Immunol. 2, eaag0192 (2017).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institutes of Health (R01 AR042527, R01 HL117913, R01 AI108906, R01 HL142068 and P01 HL129941 to C.M.W. and R01 AI108891, R01 AG045779, U19 AI057266 and R01 AI129191 to J.J.G.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. We thank E. Fischer from Harvard Medical School for providing the ALV2 compound, P. Li from the National Institutes of Health for providing processed ChIP-seq data files of human CD4+ T cells, C. Gustafson and F. Müller for suggestions on single-cell data analysis, X. Wang and F. Cao for suggestions on ATAC-seq time course analysis and the Stanford Genome Sequencing Service Center and Novogen for providing sequencing services.

Author information

Authors and Affiliations

Authors

Contributions

H.Z., R.R.J., W.J.G., C.M.W. and J.J.G. designed the research and interpreted data. H.Z., W.C., T.Z., J.J., S.O., J.M. and Z.H. performed experimental work. R.R.J. and I.N.G. analyzed high-throughput data. H.Z., R.R.J. and J.J.G. wrote the manuscript.

Corresponding author

Correspondence to Jörg J. Goronzy.

Ethics declarations

Competing interests

W.J.G. has affiliations with 10x Genomics (consultant). The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Graham Pawelec, Benedict Seddon and Harinder Singh for their contribution to the peer review of this work. Primary Handling Editor: Laurie A. Dempsey, in collaboration with the Nature Immunology team. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Related to Fig. 1: Titration of TCR stimulation.

a, Naive CD4+ T cells were stimulated with polystyrene beads coated with indicated amounts of anti-CD3 Ab together with a constant amount of 1 µg mL-1 antiCD28. Bead-cell conjugates were gated and the percent of bound to total cells were determined. b, Histograms of phosphorylated ERK gated on bead-bound cells at 0 and 5 min stimulated with indicated amounts of anti-CD3. c, Mean fluorescence intensity (MFI) of phosphorylated ZAP70, SLP76 and ERK was measured gated on bead-bound cells. d, Histogram of CD69 gated on bead-bound cells at 24 h of stimulation (left panel). Frequency of CD69+ cells as percentage of total bead-bound cells from 0–48 h with indicated anti-CD3 concentrations (right panel). e, Cell Trace Violet dye dilution of naive CD4+ T cells stimulated with 1 µg mL-1 anti-CD3. Experiments shown are representative of three experiments.

Source data

Extended Data Fig. 2 Related to Fig. 1a, b: Longitudinal epigenomic and transcriptomic changes induced by TCR stimulation.

Naive CD4+ T cells from eight individuals were stimulated with 0.01 (low stimulation strength, square) or 1 µg mL-1 (medium strength, triangle) anti-CD3-coated beads and subjected to ATAC-seq and RNA-seq at indicated time points. a, Bar graphs show the number of peaks more or less accessible in activated cells compared to unstimulated cells at each time point. b, Differential peaks comparing unstimulated cells to either low (left) or high (right) TCR stimulation are plotted as average log2 fold-change (logFC) versus log2 mean reads per peak. c, Bar graphs show the number of significantly up- or downregulated gene transcripts for each time point. d, Up- or downregulated gene transcripts in cells activated as indicated compared to unstimulated cells are shown as volcano plots of logFC versus significance levels. To illustrate similarities between stimulation conditions, few transcripts are labeled. Comparisons in b, d were done by two-sided robust empirical Bayes moderated statistical test; a Benjamini–Hochberg adjusted P value of 0.001 was used as cutoff.

Extended Data Fig. 3 Related to Fig. 2: Higher intensity TCR stimulation amplifies age-associated chromatin accessibility differences seen at baseline or with low intensity stimulation.

Peak sets from ATAC-seq analysis of naive CD4+ T cells from young (left) or old (right) adults were compared for each of the three conditions, unstimulated and after TCR stimulation with beads coated with low and medium concentrations of anti-CD3. Comparisons were done by robust empirical Bayes moderated statistical test; a Benjamini–Hochberg adjusted P value of 0.05 was used as cutoff. Relationships between the three sets of differential peaks are shown as Venn diagrams.

Extended Data Fig. 4 Related to Fig. 2c, d: Activation-induced temporal patterns of epigenetic changes in naive CD4+ T cells from young and older adults.

a, Genes assigned to differentially accessible regulatory sites were examined for significant enrichment in biological pathways. Selected GO terms associated with the different temporal patterns in young (upper panel) and older adults (lower panel) are shown. Comparisons were done by two-sided robust empirical Bayes moderated statistical test; a Benjamini–Hochberg adjusted P value of 0.05 was used as cutoff. b, Gene expression of indicated TFs shown as mean + s.e.m. n = 8 biologically independent samples.

Extended Data Fig. 5 Related to Fig. 3c: Relationship between differential accessibility of regulatory regions and transcriptome.

a, Differential peaks between young and older individuals at indicated TCR stimulation conditions and time points are plotted as logFC versus log mean reads per peak. Comparisons were done by robust empirical Bayes moderated statistical test; a Benjamini–Hochberg adjusted P value of 0.05 was used as cutoff. b, Aggregate genome accessibility tracks (top panel) of representative sites of cluster 6–10 shown in the heat plot in Fig. 3c. Cyan-shaded areas indicate peaks that are more open in young compared to older adults, magenta-shaded areas are more open in old. Transcript data from corresponding genes as determined by RNA-seq (lower panel) and shown as mean + s.e.m. n = 8 biologically independent samples. c, LogFC difference of transcripts differentially expressed between young and older adults from 48-h RNA-seq data is plotted against logFC difference of differential peaks from ATAC-seq data annotated to the same gene at the indicated time point. Data are fitted with two-sided simple linear regression. P < 0.0001.

Extended Data Fig. 6 Related to Fig. 3h: Time course of TF expression after STAT5 inhibition.

Naive CD4+ T cells from older adults were activated for 0–24 h with polystyrene beads coated with 1 µg mL-1 anti-CD3 and anti-CD28 in the presence of DMSO solvent control or a STAT5 inhibitor. Representative western blots (top panel). Summary data from 3 experiments showing band intensities of IRF4 and BATF relative to β-actin (bottom panel) and expressed as mean + s.e.m. n = 3 biologically independent samples. Data were analyzed with two-sided paired t-test. *P < 0.05 (p = 0.02), **P < 0.01 (p = 0.0094).

Source data

Extended Data Fig. 7 Related to Fig. 4: Single cell epigenetic and transcriptional analysis of naive CD4+ T cell responses.

a, CD31 and PTK7 surface expression on naive CD4+ T cells. b, Nuclei pooled from unstimulated and activated naive CD4+ T cells were subjected to scATAC- and scRNA-seq. Imputed CD69 gene expression is projected on the UMAP of scATAC-seq. Clusters of resting and activated cells were distinguished based on containing CD69-expressing cells (left panel). UMAP maps of scATAC-seq and scRNA-seq data are shown for activated cells (right panels). c, B3GAT1 and KLRG1 gene expression were projected onto scMultiome UMAPs (top panel). Genome tracks of bulk ATAC-seq and gene expression in bulk RNA-seq expressed as mean + s.e.m. (bottom). n = 8 biologically independent samples. d, IL4 and IL17A gene expression were projected onto scMultiome UMAPs (top panel). Genome tracks of bulk ATAC-seq at 24 h of TCR stimulation (bottom).

Extended Data Fig. 8 Related to Fig. 5b: TFs binding to IL2RA enhancers.

a, Gating strategy for naive CD4+ T cells from PBMC of young and older individuals. b, ATAC-seq genome tracks of unstimulated naive CD4+ T cells from young and older adults were aligned with ENCODE ChIP-seq genome tracks of indicated TFs at IL2RA enhancer locus (see Fig. 5c). The location of enhancer A and enhancer B is indicated by red lines. c, IKZF2 and IL2RA transcripts from sorted CD25lo and CD25- naive CD4+ T cells from 7 donors. Data were fitted with two-sided simple linear regression. P = 0.0076.

Source data

Extended Data Fig. 9 Related to Fig. 6: Naive CD4+ T cell tissue infiltration after HELIOS degradation.

Naive CD4+ T cells were treated with DMSO or 1 µM ALV2 for 5 days. a, Histograms of CD25 gated on cells conjugated to beads after 4 h stimulation with 1 µg mL-1 anti-CD3 (left). Summary of CD25 MFI from four experiments (right). n = 4 biologically independent samples. Data were analyzed with two-sided paired t-test. *P < 0.05 (P = 0.01). b, IL-21 and IFNγ were stained after five days of activation with 5 µg mL-1 plate-coated anti-CD3 and 1 µg mL-1 anti-CD28. Data shown are representative of 4 (IFNγ) and 5 (IL21) experiments. c, Human CD4+ T cells recovered from mouse spleen after 7-day engraftment. n = 7 biologically independent samples. Data were analyzed with two-sided paired t-test.

Source data

Extended Data Fig. 10 Related to Fig. 7: Genome accessibility changes induced by IL-2 receptor blocking and effector TF expression induced by STAT5 inhibition.

a, LogFC of differential peaks between IL-2 receptor (IL-2R) blocking and PBS treatment are plotted against average peak size. Peaks that are also differentially accessible between young and older individuals are indicated in dark red. Comparisons were done by two-sided robust empirical Bayes moderated statistical test; a Benjamini–Hochberg adjusted P value of 0.05 was used as cutoff. b, Genome tracks of representative genes. Magenta-shaded areas indicate peaks that are more open in older adults and with PBS treatment; cyan indicates peaks that are more open in young adults or with IL-2R blocking treatment of older adults. c, IRF4 protein expression were measured by Immuno blot in 48-h activated naive CD4+ T cells from four young adults with and without STAT5 inhibition. n = 4 biologically independent samples. d, BLIMP1 and TCF1 expression was measured by flow cytometry in 5-day activated naive CD4+ T cells from four young adults. The MFI ratio of BLIMP1 to TCF1 was plotted. n = 4 biologically independent samples. Data in c,d were analyzed with two-sided paired t-test.

Source data

Supplementary information

Source data

Source Data Fig. 2

Unprocessed blots and statistical source data.

Source Data Fig. 3

Unprocessed blots and statistical dource data.

Source Data Fig. 5

Unprocessed blots and statistical source data.

Source Data Fig. 6

Unprocessed blots and statistical source data.

Source Data Fig. 7

Unprocessed blots and statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical 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

Zhang, H., Jadhav, R.R., Cao, W. et al. Aging-associated HELIOS deficiency in naive CD4+ T cells alters chromatin remodeling and promotes effector cell responses. Nat Immunol 24, 96–109 (2023). https://doi.org/10.1038/s41590-022-01369-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41590-022-01369-x

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