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:

Clonally expanded memory CD8+ T cells accumulate in atherosclerotic plaques and are pro-atherogenic in aged mice

Abstract

Aging is a strong risk factor for atherosclerosis and induces accumulation of memory CD8+ T cells in mice and humans. Biological changes that occur with aging lead to enhanced atherosclerosis, yet the role of aging on CD8+ T cells during atherogenesis is unclear. In this study, using female mice, we found that depletion of CD8+ T cells attenuated atherogenesis in aged, but not young, animals. Furthermore, adoptive transfer of splenic CD8+ T cells from aged wild-type, but not young wild-type, donor mice significantly enhanced atherosclerosis in recipient mice lacking CD8+ T cells. We also characterized T cells in healthy and atherosclerotic young and aged mice by single-cell RNA sequencing. We found specific subsets of age-associated CD8+ T cells, including a Granzyme K+ effector memory subset, that accumulated and was clonally expanded within atherosclerotic plaques. These had transcriptomic signatures of T cell activation, migration, cytotoxicity and exhaustion. Overall, our study identified memory CD8+ T cells as therapeutic targets for atherosclerosis in aging.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: CD8+ T cell depletion reduces atherosclerotic plaque size in aged WT mice but not young WT mice.
Fig. 2: Aged CD8+ T cells contain more exhausted memory cells and are more pro-atherogenic compared to young CD8+ T cells.
Fig. 3: Aging leads to expansion of memory CD8 cells that infiltrate aortic atherosclerotic plaque.
Fig. 4: CD8 T cell populations in atherosclerotic plaques are more differentiated, exhausted and cytotoxic with aging.
Fig. 5: Granzyme K+ CD8+ EM and CD8+ CM cells from aged mice are clonally expanded compared to young mice.
Fig. 6: Chemokine receptors are upregulated in CD8+ T cell subsets in aged mice compared to young mice.

Similar content being viewed by others

Data availability

Raw and processed mouse single-cell RNA sequencing and TCR sequencing data are deposited in the Gene Expression Omnibus (GSE210719). Genome builds used can be found at https://support.10xgenomics.com/single-cell-gene-expression/software/release-notes/build. Human atherosclerotic plaque data were re-analyzed from publicly available data archived on Zenodo: 3361716. The source data figures in this manuscript are published along with the manuscript. Raw data are available from the corresponding author upon reasonable request.

Code availability

No original code was written for the experiments in this study. The packages for the code used are detailed in the Methods subsection titled ‘scRNA-seq’.

References

  1. Benjamin, E. J. et al. Heart disease and stroke statistics—2018 update: a report from the American Heart Association. Circulation 137, e67–e492 (2018).

    PubMed  Google Scholar 

  2. Libby, P. et al. Atherosclerosis. Nat. Rev. Dis. Primers 5, 56 (2019).

    PubMed  Google Scholar 

  3. Tyrrell, D. J. et al. Age-associated mitochondrial dysfunction accelerates atherogenesis. Circ. Res. 126, 298–314 (2020).

    CAS  PubMed  Google Scholar 

  4. Hansson, G. K., Holm, J. & Jonasson, L. Detection of activated T lymphocytes in the human atherosclerotic plaque. Am. J. Pathol. 135, 169–175 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Schäfer, S. & Zernecke, A. CD8 T cells in atherosclerosis. Cells 10, 37 (2020).

    PubMed  PubMed Central  Google Scholar 

  6. Ait-Oufella, H. et al. Natural regulatory T cells control the development of atherosclerosis in mice. Nat. Med. 12, 178–180 (2006).

    CAS  PubMed  Google Scholar 

  7. Benagiano, M. et al. T helper type 1 lymphocytes drive inflammation in human atherosclerotic lesions. Proc. Natl Acad. Sci. USA 100, 6658–6663 (2003).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cardilo-Reis, L. et al. Interleukin-13 protects from atherosclerosis and modulates plaque composition by skewing the macrophage phenotype. EMBO Mol. Med. 4, 1072–1086 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Elhage, R. et al. Deleting TCRαβ+ or CD4+ T lymphocytes leads to opposite effects on site-specific atherosclerosis in female apolipoprotein E-deficient mice. Am. J. Pathol. 165, 2013–2018 (2004).

    PubMed  PubMed Central  Google Scholar 

  10. Cochain, C. et al. CD8+ T cells regulate monopoiesis and circulating Ly6Chigh monocyte levels in atherosclerosis in mice. Circ. Res. 117, 244–253 (2015).

    CAS  PubMed  Google Scholar 

  11. Kyaw, T. et al. Cytotoxic and proinflammatory CD8+ T lymphocytes promote development of vulnerable atherosclerotic plaques in apoE-deficient mice. Circulation 127, 1028–1039 (2013).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Palmer, D. B. The effect of age on thymic function. Front. Immunol. 4, 316 (2013).

    PubMed  PubMed Central  Google Scholar 

  14. Saule, P. et al. Accumulation of memory T cells from childhood to old age: central and effector memory cells in CD4+ versus effector memory and terminally differentiated memory cells in CD8+ compartment. Mech. Ageing Dev. 127, 274–281 (2006).

    CAS  PubMed  Google Scholar 

  15. Bjorklund, M. M. et al. Induction of atherosclerosis in mice and hamsters without germline genetic engineering. Circ. Res. 114, 1684–1689 (2014).

    CAS  PubMed  Google Scholar 

  16. 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).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  18. Zheng, C. et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356 (2017).

    CAS  PubMed  Google Scholar 

  19. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    CAS  PubMed  Google Scholar 

  20. Jonsson, A. H. et al. Granzyme K. Sci. Transl. Med. 14, eabo0686 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at https://doi.org/10.48550/arXiv.1802.03426 (2018).

  22. Saul, D. et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat. Commun. 13, 4827 (2022).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fernandez, D. M. et al. Single-cell immune landscape of human atherosclerotic plaques. Nat. Med. 25, 1576–1588 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Lantz, O. & Bendelac, A. An invariant T cell receptor alpha chain is used by a unique subset of major histocompatibility complex class I-specific CD4+ and CD4-8 T cells in mice and humans. J. Exp. Med. 180, 1097–1106 (1994).

    CAS  PubMed  Google Scholar 

  25. Carey, A. J. et al. Public clonotypes and convergent recombination characterize the naïve CD8. Front. Immunol. 8, 1859 (2017).

    PubMed  PubMed Central  Google Scholar 

  26. Bedel, R. et al. Effective functional maturation of invariant natural killer T cells is constrained by negative selection and T-cell antigen receptor affinity. Proc. Natl Acad. Sci. USA 111, E119–E128 (2014).

    CAS  PubMed  Google Scholar 

  27. Kimmel, J. C. et al. Murine single-cell RNA-seq reveals cell-identity- and tissue-specific trajectories of aging. Genome Res. 29, 2088–2103 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  29. Winkels, H. et al. Atlas of the immune cell repertoire in mouse atherosclerosis defined by single-cell RNA-sequencing and mass cytometry. Circ. Res. 122, 1675–1688 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Cochain, C. et al. Single-cell RNA-seq reveals the transcriptional landscape and heterogeneity of aortic macrophages in murine atherosclerosis. Circ. Res. 122, 1661–1674 (2018).

    CAS  PubMed  Google Scholar 

  31. Cole, J. E. et al. Immune cell census in murine atherosclerosis: cytometry by time of flight illuminates vascular myeloid cell diversity. Cardiovasc. Res. 114, 1360–1371 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Getz, G. S. & Reardon, C. A. Animal models of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 32, 1104–1115 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Uyar, B. et al. Single-cell analyses of aging, inflammation and senescence. Ageing Res. Rev. 64, 101156 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Barbe-Tuana, F., Funchal, G., Schmitz, C. R. R., Maurmann, R. M. & Bauer, M. E. The interplay between immunosenescence and age-related diseases. Semin. Immunopathol. 42, 545–557 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kirkland, J. L. & Tchkonia, T. Cellular senescence: a translational perspective. EBioMedicine 21, 21–28 (2017).

    PubMed  PubMed Central  Google Scholar 

  36. Koltsova, E. K. et al. Dynamic T cell–APC interactions sustain chronic inflammation in atherosclerosis. J. Clin. Invest. 122, 3114–3126 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Shaw, M. K. et al. T-cells specific for a self-peptide of ApoB-100 exacerbate aortic atheroma in murine atherosclerosis. Front Immunol 8, 95 (2017).

    PubMed  PubMed Central  Google Scholar 

  38. Stemme, S. et al. T lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proc. Natl Acad. Sci. USA 92, 3893–3897 (1995).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Chou, M. Y. et al. Oxidation-specific epitopes are dominant targets of innate natural antibodies in mice and humans. J. Clin. Invest. 119, 1335–1349 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Ribot, J. C., Lopes, N. & Silva-Santos, B. γδ T cells in tissue physiology and surveillance. Nat. Rev. Immunol. 21, 221–232 (2021).

    CAS  PubMed  Google Scholar 

  41. Cheng, H.-Y., Wu, R. & Hedrick, C. C. Gammadelta (γδ) T lymphocytes do not impact the development of early atherosclerosis. Atherosclerosis 234, 265–269 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Vu, D. M. et al. γδT cells are prevalent in the proximal aorta and drive nascent atherosclerotic lesion progression and neutrophilia in hypercholesterolemic mice. PLoS ONE 9, e109416 (2014).

    ADS  PubMed  PubMed Central  Google Scholar 

  43. Clément, M. et al. Deletion of IRF8 (interferon regulatory factor 8)-dependent dendritic cells abrogates proatherogenic adaptive immunity. Circ. Res. 122, 813–820 (2018).

    PubMed  Google Scholar 

  44. Tyrrell, D. J., Blin, M. G., Song, J., Wood, S. C. & Goldstein, D. R. Aging impairs mitochondrial function and mitophagy and elevates interleukin 6 within the cerebral vasculature. J. Am. Heart Assoc. 9, e017820 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Du, W., Shen, H., Galan, A. & Goldstein, D. R. An age-specific CD8+ T cell pathway that impairs the effectiveness of strategies to prolong allograft survival. J. Immunol. 187, 3631–3640 (2011).

    CAS  PubMed  Google Scholar 

  46. Mulholland, M. et al. IL-2Rβγ signalling in lymphocytes promotes systemic inflammation and reduces plasma cholesterol in atherosclerotic mice. Atherosclerosis 326, 1–10 (2021).

    CAS  PubMed  Google Scholar 

  47. Paigen, B., Morrow, A., Holmes, P. A., Mitchell, D. & Williams, R. A. Quantitative assessment of atherosclerotic lesions in mice. Atherosclerosis 68, 231–240 (1987).

    CAS  PubMed  Google Scholar 

  48. Daugherty, A. et al. Recommendation on design, execution, and reporting of animal atherosclerosis studies: a scientific statement from the American Heart Association. Arterioscler. Thromb. Vasc. Biol. 37, e131–e157 (2017).

    CAS  PubMed  Google Scholar 

  49. Fernandez-Hernando, C. et al. Loss of Akt1 leads to severe atherosclerosis and occlusive coronary artery disease. Cell Metab. 6, 446–457 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Germain, P. L., Lun, A., Garcia Meixide, C., Macnair, W. & Robinson, M. D. Doublet identification in single-cell sequencing data using scDblFinder. F1000Res. 10, 979 (2021).

    PubMed  Google Scholar 

  52. Shao, X. et al. scCATCH: automatic annotation on cell types of clusters from single-cell RNA sequencing data. iScience 23, 100882 (2020).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. Le, T. et al. BBrowser: making single-cell data easily accessible. Preprint at bioRxiv https://doi.org/10.1101/2020.12.11.414136 (2020).

  54. Vuong, H., Truong, T., Phan, T. & Pham, S. Venice: a new algorithm for finding marker genes in single-cell transcriptomic data. Preprint at bioRxiv https://doi.org/10.1101/2020.11.16.384479 (2020).

  55. Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res. 9, 47 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by National Institutes of Health (NIH) awards AG068309 (D.J.T.), HL155169, AG028082, AI138347 (D.R.G.), HL158003 (J.C.) and AHA898210 (J.S.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We acknowledge support from the Bioinformatics Core of the University of Michigan Medical School’s Biomedical Research Core Facilities, especially D. King. We acknowledge W. Rosebury-Smith in the Unit for Laboratory Animal Management, In-Vivo Animal Core, for expertise and assistance with histology. Research reported in this publication was supported by the National Cancer Institute under award P30CA046592 by use of the following Cancer Center Shared Resource: Single Cell and Spatial Analysis Shared Resource.

Author information

Authors and Affiliations

Authors

Contributions

D.J.T. and D.R.G. conceived the project and performed experiments. D.J.T. and D.R.G. secured funding. D.J.T. and D.R.G. analyzed the data. D.J.T. and D.R.G. wrote the manuscript. All authors edited the manuscript.

Corresponding author

Correspondence to Daniel J. Tyrrell.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Klaus Ley, Tin Kyaw and Holger Winkels for their contributions to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Aging leads to expansion of memory CD8+ T cells in circulation.

(a) Schematic of experimental procedure to isolate blood and perform flow cytometry at 3 timepoints during atherogenesis in young wild-type, aged wild-type, and young Ldlr-/- mice. (b) Blood-cell flow cytometry gating strategy to identify CD8+ T cells. Live CD45+ lymphocytes were gated on CD3e+ cells, CD8+ cells, and subdivided into naive (CD62L+CD44), central memory (CD62L+CD44+), and effector memory (CD44+CD62L) then PD1+ and either granzyme K+ or Tox+ cells. (c-l) Flow cytometry quantification of T cell populations as a frequency of CD3+ T cells for young C57BL/6, aged C57BL/6, and young Ldlr-/-, mice at baseline, midway through the western diet feeding period (5-weeks), and just prior to sacrifice (10-weeks). N = 6 biological replicates per group over 1 independent experiment. Measurements were taken from distinct samples. Data in this figure represents mean ± SEM. 2-Way ANOVA with Tukey’s post-hoc test. PCSK9 = proprotein convertase subtilisin/kexin type 9 serine protease, AAV = adeno-associated virus, CM = central memory, EM = effector memory.

Source data

Extended Data Fig. 2 Aging leads to expansion of memory CD8+ T cells in circulation.

Representative flow cytometry plots of T cells for young WT, aged WT, and young Ldlr-/- mice after PCSK9-AAV and 10-weeks of western diet feeding. Representative flow cytometry plots demonstrating increased frequency of central memory CD8+ T cells in aged C57BL/6 WT mice and young Ldlr-/- mice compared to young C57BL/6 WT mice, increased frequency of effector memory CD8+ T cells in aged C57BL/6 WT mice versus young C57BL/6 WT and Ldlr-/- mice, and greater frequency of naive CD8+ T cells in young C57BL/6 WT mice and young Ldlr-/- mice compared with aged C57BL/6 WT mice. Aged WT mice also demonstrate greater frequency of PD1+ EM CD8+ T cells and granzyme K+ PD1+ EM CD8+ T cells compared to young WT and Ldlr-/- mice. PCSK9 = proprotein convertase subtilisin/kexin type 9 serine protease, AAV = adeno-associated virus, CM = central memory, EM = effector memory.

Extended Data Fig. 3 Similar fasting total cholesterol level in young and aged C57BL/6 WT mice treated with anti-IgG or anti-CD8 treatment over 10-weeks of WD feeding.

Fasting total cholesterol was quantified from plasma via colorimetric assay. 2-way ANOVA with Tukey’s post-hoc test. N = 11 young WT anti-CD8a mice, N = 10 young WT anti-IgG mice, N = 16 aged WT anti-CD8 mice, N = 17 aged WT anti-IgG mice, N = 6 young Ldlr-/- anti-IgG mice, and N = 7 young Ldlr-/- anti-CD8 mice over 3 biological replicates. Data in this figure represents mean ± SEM. Measurements were taken from distinct samples. Ldlr = low-density lipoprotein receptor. WD = western diet, WT = wild-type.

Source data

Extended Data Fig. 4 CD8+ T cell depletion reduces brachiocephalic artery atherosclerotic plaque size in Aged WT mice but not young WT mice.

Murine PCSK9-AAV-induced hypercholesterolemia model of atherosclerosis with anti-CD8 or anti-IgG antibody injection every 2 weeks during the WD feeding period. Representative histology and atherosclerotic plaque size quantification of the brachiocephalic artery of young PCSK9-AAV (a), aged PCSK9-AAV (b), and Ldlr-/- mice (c) treated with either anti-CD8 antibody treatment or anti-IgG treatment. Scale bar = 100 µm. In A-C, data are pooled from 3 independent experiments and all data are shown. N = 10 for young anti-IgG, N = 11 for young anti-CD8, N = 17 for aged anti-IgG, N = 16 for aged anti-CD8, N = 7 for young Ldlr-/- anti-IgG, and N = 6 for young Ldlr-/- anti-CD8 over 3 independent experiments. Measurements were taken from distinct samples. Data in this figure represents mean ± SEM. 2-Way ANOVA with Tukey’s post-hoc test. BCA = brachiocephalic artery, CM = central memory, EM = effector memory, PCSK9 = proprotein convertase subtilisin/kexin type 9 serine protease.

Source data

Extended Data Fig. 5 Anti-CD8 treatment significantly reduces the number of CD8+ T cells in atherosclerotic aortas.

Aortas from aged mice transfected with PCSK9-AAV and subjected to 10-weeks of western diet feeding and either treated with anti-CD8 or anti-IgG isotype control antibody during western diet feeding were digested and analyzed by flow cytometry. The flow cytometry gating strategy is shown demonstrating CD4+ and CD8+ T cells in anti-CD8 and anti-IgG treated atherosclerotic mice. Quantification demonstrates a significant reduction of CD8+ T cells in anti-CD8 treated mice compared to isotype control. N = 5 anti-IgG treated mice and N = 5 anti-CD8 treated mice over 1 independent experiment. Data in this figure represents mean ± SEM. Each point is a biological replicate and measurements were taken from distinct samples. Two-tailed Mann-Whitney U test.

Source data

Extended Data Fig. 6 CD8+ T cell enrichment from young and aged C57BL/6 WT donor spleens.

(a) Flow cytometry gating strategy for young and aged WT mice before and after CD8+ T cell enrichment prior to CD8+ T cell adoptive transfer into Cd8-/- mice showing gating and frequency of previous gate. (b) Representative flow cytometry plots demonstrating CD4+ and CD8+ T cells, memory and naive CD8+ T cells, PD1+ effector memory CD8+ T cells, and PD1+ central memory CD8+ T cells showing gating and frequency of parent gate.

Extended Data Fig. 7 Similar fasting total cholesterol level in young Cd8-/- mice adoptively transferred with 10 million CD8+ T cells or vehicle from either young C57BL/6 WT or aged C57BL/6 WT mice over 10-weeks of WD feeding.

All mice were adoptively transferred and allowed to rest for 4-weeks before PCSK9-AAV injection and 10-week WD feeding. Fasting total cholesterol was quantified from plasma via colorimetric assay. 2-way ANOVA with Tukey’s post-hoc test. N = 13 young Cd8-/- mice + young CD8+ T cells, N = 8 young Cd8-/- mice + aged CD8+ T cells, and N = 7 young Cd8-/- mice + no CD8+ T cells over 2 independent experiments. Data in this figure represents mean ± SEM. Measurements were taken from distinct samples. WD = western diet, WT = wild-type.

Source data

Extended Data Fig. 8 Human atherosclerotic plaque CD8 T cells express GZMK and associate with symptomatic atherosclerotic disease.

(a) UMAP plot of T cells from human atherosclerotic plaques from public study deposited to Zenodo: 3361716. (b) GZMK expression overlayed on UMAP plot of T cells in human atherosclerotic plaques with dashed line indicating cells with the highest GZMK expression. (c) top 15 differentially expressed genes in the CD8+ T cell cluster. (d) Violin plot of GZMK expression on 3 T cell clusters, stratified by symptomatic group. For D, two-tailed Venice non-parametric benchmarking method within BBrowser3 was used to compare groups (see Methods) and data represents mean with lines above and below representing 1st and 3rd quartiles. Raw data for this figure is from a publicly available dataset archived on Zenodo: 3361716. UMAP = uniform manifold approximation projection.

Extended Data Fig. 9 Atherosclerotic plaque T cells are more clonally expanded than splenic T cells and aging enhances clonal expansion.

(a) Abundance of unique clonotypes. (b) Clonotype diversity estimation by Chao1 index. (c) Length of CDR3 sequences by group. (d) Size of clonotypes by group. (e) Clonotype tracking of the top 5 clonotypes from each sample across all other samples. (f) Overlap of CDR3 sequences by sample in circos plot. Top 100 expanded CDR3 sequences, colored by group, of 15 amino acids (g), 10 amino acids (h), and 5 amino acids (i). Composition of amino acid sequences of different lengths including 15 amino acids (j), 10 amino acids (k), and 5 amino acids (l). Depiction of which samples had the greatest clonal expansion shared across the number of cells (x-axis) and number of samples (y-axis) and stratified by group (m), age (n), and tissue-type (o). Data includes young spleens (N = 3 biological replicates and 18,412 total cells), aged spleen (N = 3 biological replicates and 11,151 total cells), young aorta (N = 4 biological replicates and 1,999 total cells), and aged aorta (N = 4 biological replicates and 5,698 total cells) over 1 independent experiment.

Extended Data Fig. 10 Aged atherosclerotic mice have more CD3+, CD8+, and F4/80+ cells in the aorta compared to young mice.

Aortas from 3-mo old and 18-mo old mice transfected with PCSK9-AAV and subjected to 10-weeks of western diet feeding were digested and analyzed by flow cytometry. (a) The flow cytometry gating strategy is shown CD3+, CD8+, and CD11b+F4/80+ cells from the aortas of atherosclerotic mice. Quantification demonstrates a significant increase of CD3+ and CD8+ T cells as well as CD11b+F4/80+ macrophages in aged atherosclerotic mice compared with young atherosclerotic mice normalized to total aorta weight (b-d) or by mg of aorta weight (e-g). In B-G, each point is a biological replicate and measurements were taken from distinct samples. N = 6 aged and N = 7 young mice at each timepoint for a total of N = 18 aged mice and N = 21 young mice over 1 independent experiment. Data in this figure represents mean ± SEM. 2-Way ANOVA with Šídák’s post-hoc test. PCSK9 = proprotein convertase subtilisin/kexin type 9 serine protease, AAV = adeno-associated virus, CM = central memory, EM = effector memory, WD = western diet.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–12.

Reporting Summary

Supplementary Tables 1-9

Supplementary Tables 1–9.

Supplementary Data 1

Source data for Supplementary Fig. 1b–h.

Supplementary Data 2

Source data for Supplementary Fig. 2a–c.

Supplementary Data 3

Source data for Supplementary Fig. 3.

Supplementary Data 4

Source data for Supplementary Fig. 4.

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 Extended Data Fig. 1

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. 7

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

Tyrrell, D.J., Wragg, K.M., Chen, J. et al. Clonally expanded memory CD8+ T cells accumulate in atherosclerotic plaques and are pro-atherogenic in aged mice. Nat Aging 3, 1576–1590 (2023). https://doi.org/10.1038/s43587-023-00515-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-023-00515-w

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