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.

  • Letter
  • Published:

Accumulation of cytotoxic T cells in the aged CNS leads to axon degeneration and contributes to cognitive and motor decline

Abstract

Aging is a major risk factor for the development of nervous system functional decline, even in the absence of diseases or trauma. The axon–myelin units and synaptic terminals are some of the neural structures most vulnerable to aging-related deterioration1,2,3,4,5,6, but the underlying mechanisms are poorly understood. In the peripheral nervous system, macrophages—important representatives of the innate immune system—are prominent drivers of structural and functional decline of myelinated fibers and motor endplates during aging7. Similarly, in the aging central nervous system (CNS), microglial cells promote damage of myelinated axons and synapses8,9,10,11,12,13,14,15,16,17,18,19,20. Here we examine the role of cytotoxic CD8+ T lymphocytes, a type of adaptive immune cells previously identified as amplifiers of axonal perturbation in various models of genetically mediated CNS diseases21 but understudied in the aging CNS22,23,24,25. We show that accumulation of CD8+ T cells drives axon degeneration in the normal aging mouse CNS and contributes to age-related cognitive and motor decline. We characterize CD8+ T-cell population heterogeneity in the adult and aged mouse brain by single-cell transcriptomics and identify aging-related changes. Mechanistically, we provide evidence that CD8+ T cells drive axon degeneration in a T-cell receptor- and granzyme B-dependent manner. Cytotoxic neural damage is further aggravated by systemic inflammation in aged but not adult mice. We also find increased densities of T cells in white matter autopsy material from older humans. Our results suggest that targeting CD8+ CNS-associated T cells in older adults might mitigate aging-related decline of brain structure and function.

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: Neuroinflammation-related axon degeneration in the aged CNS.
Fig. 2: Cytotoxic CD8+ T cells drive axonal degeneration in the aged CNS.
Fig. 3: Systemic inflammation fosters CD8+ T cell-driven axonal degeneration in the aged CNS.
Fig. 4: CD8+ T cells accumulate in aged human white matter.

Similar content being viewed by others

Data availability

The sequencing data are available from the Gene Expression Omnibus under accession number GSE138891. Source data are provided with this paper. Other data that support the findings of this study are available from the corresponding author.

References

  1. Samuel, M. A., Zhang, Y., Meister, M. & Sanes, J. R. Age-related alterations in neurons of the mouse retina. J. Neurosci. 31, 16033–16044 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Burke, S. N. & Barnes, C. A. Neural plasticity in the ageing brain. Nat. Rev. Neurosci. 7, 30–40 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Dickstein, D. L. et al. Changes in the structural complexity of the aged brain. Aging Cell 6, 275–284 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Bowley, M. P., Cabral, H., Rosene, D. L. & Peters, A. Age changes in myelinated nerve fibers of the cingulate bundle and corpus callosum in the rhesus monkey. J. Comp. Neurol. 518, 3046–3064 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Salvadores, N., Sanhueza, M., Manque, P. & Court, F. A. Axonal degeneration during aging and its functional role in neurodegenerative disorders. Front. Neurosci. 11, 451 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Milde, S., Adalbert, R., Elaman, M. H. & Coleman, M. P. Axonal transport declines with age in two distinct phases separated by a period of relative stability. Neurobiol. Aging 36, 971–981 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Yuan, X. et al. Macrophage depletion ameliorates peripheral neuropathy in aging mice. J. Neurosci. 38, 4610–4620 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Streit, W. J., Walter, S. A. & Pennell, N. A. Reactive microgliosis. Prog. Neurobiol. 57, 563–581 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Au, N. P. B. & Ma, C. H. E. Recent advances in the study of bipolar/rod-shaped microglia and their roles in neurodegeneration. Front. Aging Neurosci. 9, 128 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Bisht, K. et al. Dark microglia: a new phenotype predominantly associated with pathological states. Glia 64, 826–839 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Grabert, K. et al. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat. Neurosci. 19, 504–516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Galatro, T. F. et al. Transcriptomic analysis of purified human cortical microglia reveals age-associated changes. Nat. Neurosci. 20, 1162–1171 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Deczkowska, A. et al. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell 173, 1073–1081 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Hickman, S., Izzy, S., Sen, P., Morsett, L. & El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 21, 1359–1369 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mrdjen, D. et al. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity 48, 380–395 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Hong, S. et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Stephan, A. H., Barres, B. A. & Stevens, B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu. Rev. Neurosci. 35, 369–389 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Shi, Q. et al. Complement C3-deficient mice fail to display age-related hippocampal decline. J. Neurosci. 35, 13029–13042 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cantuti-Castelvetri, L. et al. Defective cholesterol clearance limits remyelination in the aged central nervous system. Science 359, 684–688 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Safaiyan, S. et al. Age-related myelin degradation burdens the clearance function of microglia during aging. Nat. Neurosci. 19, 995–998 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Groh, J. & Martini, R. Neuroinflammation as modifier of genetically caused neurological disorders of the central nervous system: understanding pathogenesis and chances for treatment. Glia 65, 1407–1422 (2017).

    Article  PubMed  Google Scholar 

  22. Stichel, C. C. & Luebbert, H. Inflammatory processes in the aging mouse brain: participation of dendritic cells and T-cells. Neurobiol. Aging 28, 1507–1521 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Gemechu, J. M. & Bentivoglio, M. T cell recruitment in the brain during normal aging. Front. Cell. Neurosci. 6, 38 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ritzel, R. M. et al. Age-associated resident memory CD8 T cells in the central nervous system are primed to potentiate inflammation after ischemic brain injury. J. Immunol. 196, 3318–3330 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Dulken, B. W. et al. Single-cell analysis reveals T cell infiltration in old neurogenic niches. Nature 571, 205–210 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Spear, P. D. Neural bases of visual deficits during aging. Vision Res. 33, 2589–2609 (1993).

    Article  CAS  PubMed  Google Scholar 

  27. Bridge, K. E. et al. Late onset distal axonal swelling in YFP-H transgenic mice. Neurobiol. Aging 30, 309–321 (2009).

    Article  PubMed  Google Scholar 

  28. Stilling, R. M. et al. De-regulation of gene expression and alternative splicing affects distinct cellular pathways in the aging hippocampus. Front. Cell. Neurosci. 8, 373 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Baruch, K. et al. Aging-induced type I interferon response at the choroid plexus negatively affects brain function. Science 346, 89–93 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Norden, D. M. & Godbout, J. P. Review: microglia of the aged brain: primed to be activated and resistant to regulation. Neuropathol. Appl. Neurobiol. 39, 19–34 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Korin, B. et al. High-dimensional, single-cell characterization of the brain’s immune compartment. Nat. Neurosci. 20, 1300–1309 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Wakim, L. M. et al. The molecular signature of tissue resident memory CD8 T cells isolated from the brain. J. Immunol. 189, 3462–3471 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Smolders, J. et al. Tissue-resident memory T cells populate the human brain. Nat. Commun. 9, 4593 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Schetters, S. T. T., Gomez-Nicola, D., Garcia-Vallejo, J. J. & Van Kooyk, Y. Neuroinflammation: microglia and T cells get ready to tango. Front. Immunol. 8, 1905 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Mohebiany, A. N. et al. Microglial A20 protects the brain from CD8 T-cell-mediated immunopathology. Cell Rep. 30, 1585–1597 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Kurd, N. S. et al. Early precursors and molecular determinants of tissue-resident memory CD8+ T lymphocytes revealed by single-cell RNA sequencing. Sci. Immunol. 5, eaaz6894 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Urban, S. L. et al. Peripherally induced brain tissue-resident memory CD8+ T cells mediate protection against CNS infection. Nat. Immunol. 21, 938–949 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. McGinnis, G. J. & Raber, J. CNS side effects of immune checkpoint inhibitors: preclinical models, genetics and multimodality therapy. Immunotherapy 9, 929–941 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kroner, A., Ip, C. W., Thalhammer, J., Nave, K.-A. & Martini, R. Ectopic T-cell specificity and absence of perforin and granzyme B alleviate neural damage in oligodendrocyte mutant mice. Am. J. Pathol. 176, 549–555 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Leder, C. et al. Clonal expansions of pathogenic CD8+ effector cells in the CNS of myelin mutant mice. Mol. Cell. Neurosci. 36, 416–424 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Filiano, A. J., Gadani, S. P. & Kipnis, J. How and why do T cells and their derived cytokines affect the injured and healthy brain? Nat. Rev. Neurosci. 18, 375–384 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Norris, G. T. & Kipnis, J. Immune cells and CNS physiology: microglia and beyond. J. Exp. Med. 216, 60–70 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Deczkowska, A., Baruch, K. & Schwartz, M. Type I/II interferon balance in the regulation of brain physiology and pathology. Trends Immunol. 37, 181–192 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Zarif, H. et al. CD8+ T cells are essential for the effects of enriched environment on hippocampus-dependent behavior, hippocampal neurogenesis and synaptic plasticity. Brain Behav. Immun. 69, 235–254 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Smith, C. J. et al. Probiotics normalize the gut-brain-microbiota axis in immunodeficient mice. Am. J. Physiol. Gastrointest. Liver Physiol. 307, G793–G802 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Faraco, G. et al. Dietary salt promotes neurovascular and cognitive dysfunction through a gut-initiated TH17 response. Nat. Neurosci. 21, 240–249 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Feng, G. et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron 28, 41–51 (2000).

    Article  CAS  PubMed  Google Scholar 

  48. Mombaerts, P. et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    Article  CAS  PubMed  Google Scholar 

  49. Fung-Leung, W. P. et al. CD8 is needed for development of cytotoxic T cells but not helper T cells. Cell 65, 443–449 (1991).

    Article  CAS  PubMed  Google Scholar 

  50. Heusel, J. W., Wesselschmidt, R. L., Shresta, S., Russell, J. H. & Ley, T. J. Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell 76, 977–987 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Hogquist, K. A. et al. T cell receptor antagonist peptides induce positive selection. Cell 76, 17–27 (1994).

    Article  CAS  PubMed  Google Scholar 

  52. Oghumu, S. et al. Distinct populations of innate CD8+ T cells revealed in a CXCR3 reporter mouse. J. Immunol. 190, 2229–2240 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Jung, S. et al. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell. Biol. 20, 4106–4114 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Groh, J. et al. Pathogenic inflammation in the CNS of mice carrying human PLP1 mutations. Hum. Mol. Genet. 25, 4686–4702 (2016).

    CAS  PubMed  Google Scholar 

  55. Ferreira, T. A. et al. Neuronal morphometry directly from bitmap images. Nat. Methods 11, 982–984 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Williams, R. W., Strom, R. C., Rice, D. S. & Goldowitz, D. Genetic and environmental control of variation in retinal ganglion cell number in mice. J. Neurosci. 16, 7193–7205 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Supek, F., Bošnjak, M., Škunca, N. & Šmuc, T. REVIGO summarizes and visualizes long lists of gene ontology terms. PLoS ONE 6, e21800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Groh, J., Stadler, D., Buttmann, M. & Martini, R. Non-invasive assessment of retinal alterations in mouse models of infantile and juvenile neuronal ceroid lipofuscinosis by spectral domain optical coherence tomography. Acta Neuropathol. Commun. 2, 54 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Lueptow, L. M. Novel object recognition test for the investigation of learning and memory in mice. J. Vis. Exp. 126, 55718 (2017).

    Google Scholar 

  63. Groh, J. et al. Immune cells perturb axons and impair neuronal survival in a mouse model of infantile neuronal ceroid lipofuscinosis. Brain 136, 1083–1101 (2013).

    Article  PubMed  Google Scholar 

  64. Khan, A. B. et al. Redirection to the bone marrow improves T cell persistence and antitumor functions. J. Clin. Invest. 128, 2010–2024 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Smith, C. & Millar, T. Brain donation procedures in the Sudden Death Brain Bank in Edinburgh. Handb. Clin. Neurol. 150, 17–27 (2018).

    Article  PubMed  Google Scholar 

  66. Groh, J., Berve, K. & Martini, R. Fingolimod and teriflunomide attenuate neurodegeneration in mouse models of neuronal ceroid lipofuscinosis. Mol. Ther. 25, 1889–1899 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Faul, F., Erdfelder, E., Lang, A.-G. & Buchner, A. G*Power 3: a flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behav. Res. Methods 39, 175–191 (2007).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Hörner and S. Popp for initial advice regarding the NOR analysis and D. Klein for fruitful discussions and suggestions. We thank H. Blazyca, S. Loserth and B. Meyer for technical assistance and J. Schreiber, A. Weidner and T. Bimmerlein for their attentive care of mice. We thank the Edinburgh Brain and Tissue Bank for providing the postmortem brain tissue. This work was supported by the German Research Foundation (grant no. MA1053/6-2 to R.M. and grant no. GR5240/1-1 to J.G.), the Roman, Marga und Mareille Sobek Foundation (to R.M.), the Charitable Hertie Foundation (grant no. P1150084 to J.G.) and the Interdisciplinary Centre for Clinical Research of the University of Würzburg (grant no. A-302 to R.M. and grant no. Z-6 to P.A.).

Author information

Authors and Affiliations

Authors

Contributions

J.G. and R.M. planned and oversaw all aspects of the study. J.G. performed and analyzed most of the experiments. K.K. and W.K. performed the flow cytometry and analyzed the scRNA-seq experiments. P.A. and A.E.S. performed the cell sorting and scRNA-seq. X.Y., L.L. and R.M. designed and performed the animal experiments. J.G. and R.M. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Janos Groh.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Aging 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 Aging results in myelin alterations and T cell-associated axon degeneration in the CNS.

a, Representative electron micrographs of optic nerve cross-sections from adult (12-month-old) and aged (24-month-old) mice. Redundant myelin and myelin fragments (open arrowheads) as well as axonal spheroids typically showing swelling and accumulation of organelles (asterisks) or neurofilaments (hashtag) are detectable in aged but not adult nerves. Additionally, periaxonal vacuoles (circle) as well as degenerating fibres (arrowhead) are occasionally present in aged nerves. Scale bars, 2 μm. b, Z-stack projections of YFPtg axons in optic nerves and c, YFPtg axon terminals in the superior colliculi of adult and aged Thy1-YFP-H transgenic mice. Irregular and discontinuous axons with spheroids (arrows) and reduced density of axon terminals are observed in aged mice. Scale bar, 20 μm. d, Automated optokinetic response analysis of visual acuity (c/d, cycles per degree) shows no significant difference between adult and aged mice. Only at a more advanced age (28-month-old), there is a significant decline of visual acuity. (n = 5 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (2, 11) = 12.95, P = 0.001). e, Immunofluorescent detection and quantification of CD4+ T lymphocytes (arrow) in optic nerves from mice at distinct ages and aged Rag1−/− mice. Densities of CD4+ T cells are significantly increased in aged compared with adult mice (n = 5 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (5, 24) = 16.58, P < 0.001). Scale bar, 20 µm. f, Immunofluorescent detection and Z-stack 3D reconstruction of CD8+ T lymphocytes and SMI32+ axonal spheroids in an aged optic nerve by confocal microscopy (see also Supplementary Video 1). Higher magnification of individual optical slices and quantifications show proximity (< 10 µm) of CD8+ T cells to SMI32+ axonal spheroids and vice versa (n = 150 CD8+ cells and 211 SMI32+ profiles from 5 aged mice). Scale bars, 10 µm. g, Electron micrograph of a longitudinal optic nerve section and quantification shows that most damaged axons (axonal spheroids or degenerating axons; n = 102 from 5 aged mice) are ensheathed by myelin. Scale bar, 1 µm.

Source data

Extended Data Fig. 2 Aging results in retinal thinning with modest neuron loss in the CNS.

a, Optical coherence tomography (OCT) analysis of the innermost retinal composite layer (NFL/GCL/IPL, nerve fibre layer, ganglion cell layer, inner plexiform layer) in peripapillary circle scans at distinct ages demonstrates retinal thinning in aged (24-month-old) compared with adult (12-month-old) mice (n = 5 mice per group, repeated measures one-way ANOVA with Bonferroni’s multiple comparisons test, F (2.039, 8.154) = 31.52, P < 0.001, each circle represents the mean value of one mouse), v- blood vessel. b, Retinal area is not changed during aging (n = 5 mice per group, one-way ANOVA, F (4, 20) = 1.757, P = 0.18). c, Histological and d, immunofluorescent visualization of neurons in the ganglion cell layer (GCL) of retinal flat mount preparations. Scale bars, 50 μm. e, f, Quantification of respective cell densities in adult and aged mice in the middle region of the retina. Densities of Nissl-stained GCL neurons are not significantly reduced (n = 5 mice per group, one-way ANOVA, F (4, 20) = 2.217, P = 0.10), whereas RBPMS+ retinal ganglion cells (of which 91% were also Brn3a+) but not ChAT+ displaced amacrine cells are significantly decreased in aged compared with adult mice (n = 4 mice per group, two-sided Student’s t-test, RBPMS+: t = 4.339 df = 6, ChAT+: t = 1.008, df = 6). g, Brain weights of aged mice are mildly reduced compared with adult mice (n = 5 mice per group, two-sided Student’s t-test, t = 2.466, df = 8).

Source data

Extended Data Fig. 3 Aging reduces dendritic arborization of alpha-RGCs.

a, Representative example of immunohistochemical identification of YFPtg SMI32+RBPMS+ alpha-RGCs (arrow) in retinal flat mount preparations from Thy1-YFP-H transgenic mice for subsequent neurite tracing and Sholl analysis. Scale bar, 50 μm b, Tracing and Sholl analysis of the complete dendritic arbor (purple) in z-stack projections of YFPtg alpha-RGCs (Extended Data Fig. 1b) demonstrates a significant reduction in total dendrite length and ramification index of aged (24-month-old) compared with adult (12-month-old) Thy1-YFP-H transgenic mice (n = 9 RGCs from 3 mice per group, two-sided Student’s t-test, dendrite length: t = 9.867 df = 4, ramification index: t = 4.501, df=4. Each circle represents one RGC and each diamond represents the mean value of one mouse). Scale bar, 50 μm. c, Representative plots of Sholl analysis for traced dendritic arbors of alpha-RGCs from adult and aged mice.

Source data

Extended Data Fig. 4 Aging-related accumulation of CD8+ T cells in the CNS is specific to the white matter.

a, Immunofluorescence-based quantification of CD8+ T cells and SMI32+ axonal spheroids in distinct CNS white matter regions of adult (12-month-old) and aged (24-month-old) mice (each circle represents the mean value of one mouse). Densities of CD8+ T cells and SMI32+ axonal spheroids are significantly increased in optic nerve (also shown in Fig. 1e,f, n = 5 mice per group), corpus callosum (n = 4 mice per group), and cerebellar white matter (n = 4 mice per group) of aged mice (two-sided Student’s t-test, CD8+: optic nerve: t = 12.95, df = 8, corpus callosum: t = 4.849, df = 6, cerebellar white matter: t = 4.025, df = 6, SMI32+: optic nerve: t = 6.247, df = 8, corpus callosum: t = 9.003, df = 6, cerebellar white matter: t = 5.846, df = 6). b, Quantification of CD8+ T cells in distinct CNS gray matter regions of adult and aged mice. Densities of CD8+ T cells are not significantly increased in retina (n = 5 mice per group), frontal cortex (n = 4 mice per group), and cerebellar cortex (n = 4 mice per group) of aged mice (two-sided Student’s t-test, retina: t = 1, df = 8, frontal cortex: t = 0.6547, df = 6, cerebellar cortex: t = 1.163, df = 6). c, Immunofluorescent detection of MHC-I on Iba1+ microglia (arrowheads) or d, on APC+ oligodendrocytes (arrows) in optic nerves from adult and aged mice. Scale bars, 20 µm. e, Quantification reveals increased percentages of MHC-I+ oligodendrocytes and MHC-I+ microglia in aged nerves (n = 5 mice per group, two-sided Student’s t-test, oligodendrocytes: t = 5.425, df = 8, microglia: t = 8.794, df = 8).

Source data

Extended Data Fig. 5 Aging-related CD8+ T cell-driven axon degeneration responds to systemic challenge.

a, Electron microscopy-based quantification of profiles showing axonal spheroid formation or undergoing degeneration in optic nerves (each circle represents the mean value of one mouse). Scale bar, 2 μm. Absence of adaptive immune cells attenuates axon damage in aged (24-month-old) mice (n = 4 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (2, 9) = 10.7, P = 0.004). b, Axonal damage is reestablished to Wt levels in aged Rag1−/− BMC Wt mice but not in Rag1−/− BMC Cd8−/−, BMC Gzmb−/−, or BMC OT-I mice (n = 4 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (3, 12) = 13.2, P < 0.001). c, LPS injection aggravates axonal damage in aged Wt but not aged Cd8−/− or adult (12-month-old) Wt mice (n = 4 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (3, 12) = 49.17, P < 0.001). d, OCT analysis of the innermost retinal composite layer (NFL/GCL/IPL) in peripapillary circle scans. Aging-related retinal thinning is attenuated in Rag1−/− mice (n = 5 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (2, 12) = 46.27, P < 0.001). e, LPS injection enhances retinal thinning in aged Wt but not aged Cd8−/− or adult Wt mice (n = 5 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (3, 16) = 95.37, P < 0.001). f, Quantification of RBPMS+ RGCs in retinae from adult, aged, and aged Rag1−/− mice. As opposed to normal aged mice, there is no significant loss of RGCs detectable in mice lacking lymphocytes (n = 4 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (2, 9) = 9.356, P = 0.006). Scale bar, 20 μm. g, Quantification of RBPMS+ RGCs in aged Rag1−/− BMC Wt and Rag1−/− BMC Cd8−/−, BMC Gzmb−/−, or BMC OT-I mice mice (n = 4 mice per group, one-way ANOVA with Bonferroni’s multiple comparisons test, F (3, 12) = 6.973, P = 0.006). h, Electron microscopy-based estimation of total axonal numbers in optic nerves of adult and aged Cd8−/− mice (n = 5 mice per group, two-sided Student’s t-test, t = 4.107, df = 10). Scale bar, 2 μm. i, Novel object recognition test shows no aging-related impairment of memory in Cd8−/− mice (n = 5 mice per group, two-sided Student’s t-test, t = 1.036, df = 8).

Source data

Extended Data Fig. 6 scRNA-seq reveals heterogeneity and activation of CD8+ T lymphocytes in the aged CNS.

a, UMAP visualization of CD8+ T cells freshly sorted from adult (12-month-old, n = 5) and aged (24-month-old, n = 4) mouse brains and analyzed by scRNA-seq. Combined (top, 8,809 cells) and separate visualization of cells from adult (middle, 4,338 cells) and aged (bottom, 4,471 cells) brains are displayed. b, Heatmap of top 10 cluster-specific genes. The color scale is based on a z-score distribution from -2 (purple) to 2 (yellow). c, Heatmaps of differentially expressed genes comparing cells isolated from adult and aged brains across all clusters (left) or within cluster 2 (right) as identified in panel a. d, Contribution of the samples to each cluster is displayed in percent and e, absolute numbers extrapolated to total cells per brain. f, Heatmap of differentially expressed genes between cluster 1 and cluster 3 as annotated in panel a. g, Violin plots of Lag3 (top) and Gzmb (bottom) gene expression in clusters 1 to 5. IST, interferon-stimulated T cells; TEFF, effector T cells; TCM, central memory T cells.

Source data

Extended Data Fig. 7 scRNA-seq reveals heterogeneity of CD8+ CNS-associated T cells.

a, UMAP visualizations of CD8+ T cells from brains of adult and aged mice showing the expression of selected control genes (Cd3e, Cd8a) and cluster-enriched marker genes (as annotated in Extended Data Fig. 6a). Transcript levels are color-coded: gray, not expressed; red, expressed. Complete lists of cluster-specific marker genes can be found in Supplementary Table 1. b, UMAP visualizations of CD8+ T cells showing the expression of selected marker genes that indicate effector/memory function and are likely involved in tissue recruitment, residency, or egress.

Extended Data Fig. 8 Localization of CD8+ CNS-associated T cell subtypes.

a, Immunofluorescent localization of CD8+ T cells (arrows) and laminin+ blood vessels in longitudinal optic nerve and coronal brain sections of adult (6-month-old) CXCR3-GFP reporter mice. CD8+ T cells associated with the CNS parenchyma, meninges, choroid plexus and cerebrospinal fluid express CXCR3, while intravascular and circulating T cells are negative for CXCR3. Scale bar, 10 μm. b, Immunohistochemical characterization of CD8+ T cells (arrows) in distinct CNS-compartments of aged (24-month-old) Wt and adult CX3CR1-GFP or CXCR3-GFP reporter mice. Parenchymal (Paren) and meningeal (Men) CD8+ T cells (clusters 1 to 5) show heterogeneous expression of Lag3, CD103 and PD-1. A20 and high Stat1 expression is detected in meningeal and choroid plexus (CP)-resident CD8+ T cells (IST). CD8+CD62L-CX3CR1+ TEFF and CD8+CD62L+CXCR3- TCM2 are detected in blood and CD8+CD62L+CXCR3+ TCM1 are detected in meninges and cerebrospinal fluid (CSF; inset). Scale bar, 10 μm. Micrographs represent three independent experiments with similar results.

Extended Data Fig. 9 Transcriptional changes in CD8+ CNS-associated T cells during aging.

a, Violin plots showing the expression of selected genes with significant differences between CD8+ T cells from brains of adult (12-months-old) and aged (24-month-old) mice when comparing cells from all clusters (as annotated in Extended Data Fig. 6a). Upper panels show genes enriched in CD8+ T cells from adult mice, lower panels show genes enriched in CD8+ T cells from aged mice. b, Heatmaps showing differentially expressed genes between adult and aged mice in each cluster. c, Violin plots showing the expression of selected genes with significant differences between CD8+ T cells from brains of adult and aged mice within cluster 2. Upper panels show genes enriched in CD8+ T cells from adult mice, lower panels show genes enriched in CD8+ T cells from aged mice. d, Violin plots showing the expression of selected genes with significant differences between CD8+ T cells from cluster 1 and cluster 3. Upper panels show genes enriched in cluster 1, lower panels show genes enriched in cluster 3. e, Overlap of significantly decreased (top) or enriched (bottom) transcripts when comparing cells from aged with adult mice over all clusters (upper-left circle), aged with adult mice within cluster 2 (upper-right circle) and cells from cluster 1 against cluster 3 (lower circle). There are global and cluster-specific transcriptional changes in CD8+ T cells during brain aging. Complete lists of genes with significant differences in expression for all comparisons can be found in Supplementary Table 1.

Extended Data Fig. 10 Validation of CD8+ T cell subtypes during CNS aging.

a, Representative plots of general flow cytometric gating of single, viable CD45highCD8+ T cells freshly isolated from brains. b, Gated CD8+ T cells from adult (12-month-old) mice or c, from aged (24-month-old) mice are analyzed for expression of CD62L, CX3CR1, IL7R, PD-1 and CD103. Percentages and identity of the respective cells are indicated in the quadrants or below the gate. CD8+ T cells comprise small populations of CD62L+CX3CR1- cells (TCM1 and 2) and CD62L-CX3CR1+ cells (TEFF). TCM1 and 2 show high IL7R and mostly low PD-1 expression. Among the remaining CD62L-CX3CR1- cells, some are IL7R-CD103- (cluster 3) and IL7R+CD103+ (cluster 2), but the majority are IL7R+CD103- cells which can be further subdivided by PD-1 expression (clusters 1, 5, and IST being PD-1+). There is an increase in PD-1 expression on CD8+ CNS-associated T cells during aging (n =3 mice per group).

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Table 2.

Reporting Summary

Supplementary Table 1

Cluster-specific marker genes, differentially expressed genes and GO lists for the scRNA-seq data.

Supplementary Video 1

Z-stack 3D reconstruction of CD8+ T lymphocytes and SMI32+ axonal spheroids in an aged optic nerve by confocal microscopy.

Source data

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Groh, J., Knöpper, K., Arampatzi, P. et al. Accumulation of cytotoxic T cells in the aged CNS leads to axon degeneration and contributes to cognitive and motor decline. Nat Aging 1, 357–367 (2021). https://doi.org/10.1038/s43587-021-00049-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00049-z

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