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:

Identification of senescent, TREM2-expressing microglia in aging and Alzheimer’s disease model mouse brain

Abstract

Alzheimer’s disease (AD) and dementia in general are age-related diseases with multiple contributing factors, including brain inflammation. Microglia, and specifically those expressing the AD risk gene TREM2, are considered important players in AD, but their exact contribution to pathology remains unclear. In this study, using high-throughput mass cytometry in the 5×FAD mouse model of amyloidosis, we identified senescent microglia that express high levels of TREM2 but also exhibit a distinct signature from TREM2-dependent disease-associated microglia (DAM). This senescent microglial protein signature was found in various mouse models that show cognitive decline, including aging, amyloidosis and tauopathy. TREM2-null mice had fewer microglia with a senescent signature. Treating 5×FAD mice with the senolytic BCL2 family inhibitor ABT-737 reduced senescent microglia, but not the DAM population, and this was accompanied by improved cognition and reduced brain inflammation. Our results suggest a dual and opposite involvement of TREM2 in microglial states, which must be considered when contemplating TREM2 as a therapeutic target in AD.

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: Senescent microglia with a distinct signature accumulate in aged mice and in 5×FAD mice.
Fig. 2: High levels of TREM2 are associated with expression of senescence markers in microglia.
Fig. 3: Senescent microglia are found in postmortem brain of patients with AD and in 5×FAD mice.
Fig. 4: Transcriptional signature of senescent microglia.
Fig. 5: Senolytic therapy using ABT-737 reduced the accumulation of senescent microglia, cognitive decline and neuroinflammation in 5×FAD mice.

Similar content being viewed by others

Data availability

Data to reproduce the figures are available as of the date of publication at https://github.com/noarachmian/Trem2_senescent_microglia (ref. 77).

Any additional information required to reanalyze the data reported in this paper will be available from the lead contact upon reasonable request.

Code availability

The code is available as of the date of publication at https://github.com/noarachmian/Trem2_senescent_microglia (ref. 77).

Any additional information required to reanalyze the data reported in this paper will be available from the lead contact upon reasonable request.

References

  1. De Strooper, B. & Karran, E. The cellular phase of Alzheimer’s disease. Cell 164, 603–615 (2016).

    Article  PubMed  Google Scholar 

  2. Wyss-Coray, T. & Rogers, J. Inflammation in Alzheimer disease—a brief review of the basic science and clinical literature. Cold Spring Harb. Perspect. Med. 2, a006346 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Pascoal, T. A. et al. Microglial activation and tau propagate jointly across Braak stages. Nat. Med. 27, 1592–1599 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Moore, Z., Taylor, J. M. & Crack, P. J. The involvement of microglia in Alzheimer’s disease: a new dog in the fight. Br. J. Pharmacol. 176, 3533–3543 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Guerreiro, R. et al. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med. 368, 117–127 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Jonsson, T. et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 368, 107–116 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Matcovitch-Natan, O. et al. Microglia development follows a stepwise program to regulate brain homeostasis. Science 353, aad8670 (2016).

    Article  PubMed  Google Scholar 

  8. Castellani, G. & Schwartz, M. Immunological features of non-neuronal brain cells: implications for Alzheimer’s disease immunotherapy. Trends Immunol. 41, 794–804 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Karanfilian, L., Tosto, M. G. & Malki, K. The role of TREM2 in Alzheimer’s disease; evidence from transgenic mouse models. Neurobiol. Aging 86, 39–53 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Ulrich, J. D., Ulland, T. K., Colonna, M. & Holtzman, D. M. Elucidating the role of TREM2 in Alzheimer’s disease. Neuron 94, 237–248 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Rea, I. M. et al. Age and age-related diseases: role of inflammation triggers and cytokines. Front. Immunol. 9, 586 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Jaul, E. & Barron, J. Age-related diseases and clinical and public health implications for the 85 years old and over population. Front. Public Health 5, 335 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Di Micco, R., Krizhanovsky, V., Baker, D. & d’Adda di Fagagna, F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat. Rev. Mol. Cell Biol. 22, 75–95 (2021).

    Article  PubMed  Google Scholar 

  15. 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 

  16. Kipnis, J., Cohen, H., Cardon, M., Ziv, Y. & Schwartz, M. T cell deficiency leads to cognitive dysfunction: implications for therapeutic vaccination for schizophrenia and other psychiatric conditions. Proc. Natl Acad. Sci. USA 101, 8180–8185 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ziv, Y. et al. Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood. Nat. Neurosci. 9, 268–275 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Baruch, K. et al. Breaking immune tolerance by targeting Foxp3+ regulatory T cells mitigates Alzheimer’s disease pathology. Nat. Commun. 6, 7967 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Giunta, B. et al. Inflammaging as a prodrome to Alzheimer’s disease. J. Neuroinflammation 5, 51 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Oakley, H. et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J. Neurosci. 26, 10129–10140 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rosenmann, H. et al. A novel transgenic mouse expressing double mutant tau driven by its natural promoter exhibits tauopathy characteristics. Exp. Neurol. 212, 71–84 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Palframan, R. T. et al. Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J. Exp. Med. 194, 1361–1374 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yang, J., Zhang, L., Yu, C., Yang, X.-F. & Wang, H. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark. Res. 2, 1 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Kamphuis, W., Kooijman, L., Schetters, S., Orre, M. & Hol, E. M. Transcriptional profiling of CD11c-positive microglia accumulating around amyloid plaques in a mouse model for Alzheimer’s disease. Biochim. Biophys. Acta 1862, 1847–1860 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Carroll, J. A., Striebel, J. F., Baune, C., Chesebro, B. & Race, B. CD11c is not required by microglia to convey neuroprotection after prion infection. PLoS ONE 18, e0293301 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hickman, S. E. et al. The microglial sensome revealed by direct RNA sequencing. Nat. Neurosci. 16, 1896–1905 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Haynes, S. E. et al. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 9, 1512–1519 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Mildner, A., Huang, H., Radke, J., Stenzel, W. & Priller, J. P2Y12 receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia 65, 375–387 (2017).

    Article  PubMed  Google Scholar 

  29. Merino, J. J., Muñetón-Gómez, V., Alvárez, M.-I. & Toledano-Díaz, A. Effects of CX3CR1 and Fractalkine chemokines in amyloid beta clearance and p-tau accumulation in Alzheimerʼs disease (AD) rodent models: is Fractalkine a systemic biomarker for AD? Curr. Alzheimer Res. 13, 403–412 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Lowe, S. W. & Sherr, C. J. Tumor suppression by Ink4aArf: progress and puzzles. Curr. Opin. Genet. Dev. 13, 77–83 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Burton, D. G. & Krizhanovsky, V. Physiological and pathological consequences of cellular senescence. Cell. Mol. Life Sci. 71, 4373–4386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Palmero, I., Pantoja, C. & Serrano, M. p19ARF links the tumour suppressor p53 to Ras. Nature 395, 125–126 (1998).

    Article  CAS  PubMed  Google Scholar 

  34. Freund, A., Patil, C. K. & Campisi, J. p38MAPK is a novel DNA damage response‐independent regulator of the senescence‐associated secretory phenotype. EMBO J. 30, 1536–1548 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Di Micco, R. et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006).

    Article  PubMed  Google Scholar 

  36. Beauséjour, C. M. et al. Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J. 22, 4212–4222 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Vaziri, H. et al. ATM‐dependent telomere loss in aging human diploid fibroblasts and DNA damage lead to the post‐translational activation of p53 protein involving poly (ADP‐ribose) polymerase. EMBO J. 16, 6018–6033 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mercurio, D. et al. Protein expression of the microglial marker Tmem119 decreases in association with morphological changes and location in a mouse model of traumatic brain injury. Front. Cell. Neurosci. 16, 820127 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gisslén, M. et al. CSF concentrations of soluble TREM2 as a marker of microglial activation in HIV-1 infection. Neurol. Neuroimmunol. Neuroinflamm. 6, e512 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Cantoni, C. et al. TREM2 regulates microglial cell activation in response to demyelination in vivo. Acta Neuropathol. 129, 429–447 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Jin, C. et al. A unique type of highly-activated microglia evoking brain inflammation via Mif/Cd74 signaling axis in aged mice. Aging Dis. 12, 2125–2139 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Krasemann, S. et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47, 566–581 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gasque, P., Singhrao, S. K., Neal, J. W., Götze, O. & Morgan, B. P. Expression of the receptor for complement C5a (CD88) is up-regulated on reactive astrocytes, microglia, and endothelial cells in the inflamed human central nervous system. Am. J. Pathol. 150, 31–41 (1997).

    PubMed  PubMed Central  Google Scholar 

  44. Olmos-Alonso, A. et al. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain 139, 891–907 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Kopatz, J. et al. Siglec‐h on activated microglia for recognition and engulfment of glioma cells. Glia 61, 1122–1133 (2013).

    Article  PubMed  Google Scholar 

  46. Jakovljevic, M. et al. Induction of NTPDase1/CD39 by reactive microglia and macrophages is associated with the functional state during EAE. Front. Neurosci. 13, 410 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Dvir-Szternfeld, R. et al. Alzheimer’s disease modification mediated by bone marrow-derived macrophages via a TREM2-independent pathway in mouse model of amyloidosis. Nat. Aging 2, 60–73 (2022).

    Article  CAS  PubMed  Google Scholar 

  48. Wang, Y. et al. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J. Exp. Med. 213, 667–675 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bancaro, N. et al. Apolipoprotein E induces pathogenic senescent-like myeloid cells in prostate cancer. Cancer Cell 41, 602–619.e11 (2023).

    Article  CAS  PubMed  Google Scholar 

  50. Zhou, Y. et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nat. Med. 26, 131–142 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Safaiyan, S. et al. White matter aging drives microglial diversity. Neuron 109, 1100–1117 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. Ovadya, Y. & Krizhanovsky, V. Strategies targeting cellular senescence. J. Clin. Invest. 128, 1247–1254 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Yosef, R. et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat. Commun. 7, 11190 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Leger, M. et al. Object recognition test in mice. Nat. Protoc. 8, 2531–2537 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Streit, W. J., Khoshbouei, H. & Bechmann, I. Dystrophic microglia in late‐onset Alzheimer’s disease. Glia 68, 845–854 (2020).

    Article  PubMed  Google Scholar 

  57. Streit, W. J., Braak, H., Xue, Q.-S. & Bechmann, I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 118, 475–485 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bussian, T. J. et al. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562, 578–582 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Talma, N., Gerrits, E., Wang, B., Eggen, B. J. & Demaria, M. Identification of distinct and age‐dependent p16High microglia subtypes. Aging Cell 20, e13450 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Schlepckow, K. et al. Enhancing protective microglial activities with a dual function TREM2 antibody to the stalk region. EMBO Mol. Med. 12, e11227 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Jain, N., Lewis, C. A., Ulrich, J. D. & Holtzman, D. M. Chronic TREM2 activation exacerbates Aβ-associated tau seeding and spreading. J. Exp. Med. 220, e20220654 (2023).

    Article  CAS  PubMed  Google Scholar 

  62. Allen, W. E., Blosser, T. R., Sullivan, Z. A., Dulac, C. & Zhuang, X. Molecular and spatial signatures of mouse brain aging at single-cell resolution. Cell 186, 194–208.e18 (2023).

    Article  CAS  PubMed  Google Scholar 

  63. Zhang, P. et al. Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat. Neurosci. 22, 719–728 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ben-Yehuda, H. et al. Key role of the CCR2-CCL2 axis in disease modification in a mouse model of tauopathy. Mol. Neurodegener. 16, 39 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhao, N. & Bu, G. A TREM2 antibody energizes microglia. Nat. Neurosci. 26, 366–369 (2023).

  66. Rachmian, N. & Krizhanovsky, V. Senescent cells in the brain and where to find them. FEBS J. 290, 1256–1266 (2022).

  67. Xing, Z. et al. Influenza vaccine combined with moderate-dose PD1 blockade reduces amyloid-β accumulation and improves cognition in APP/PS1 mice. Brain Behav. Immun. 91, 128–141 (2021).

    Article  CAS  PubMed  Google Scholar 

  68. Zou, Y. et al. Programmed cell death protein 1 blockade reduces glycogen synthase kinase 3β activity and tau hyperphosphorylation in Alzheimer’s disease mouse models. Front. Cell Dev. Biol. 9, 769229 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Rosenzweig, N. et al. PD-1/PD-L1 checkpoint blockade harnesses monocyte-derived macrophages to combat cognitive impairment in a tauopathy mouse model. Nat. Commun. 10, 465 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Baruch, K. et al. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer’s disease. Nat. Med. 22, 135–137 (2016).

    Article  CAS  PubMed  Google Scholar 

  71. Majewska, J. et al. p16-dependent upregulation of PD-L1 impairs immunosurveillance of senescent cells. Preprint at bioRxiv https://doi.org/10.1101/2023.01.30.524522 (2023).

  72. Wang, T.-W. et al. Blocking PD-L1–PD-1 improves senescence surveillance and ageing phenotypes. Nature 611, 358–364 (2022).

    Article  CAS  PubMed  Google Scholar 

  73. Ulland, T. K. et al. TREM2 maintains microglial metabolic fitness in Alzheimer’s disease. Cell 170, 649–663 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Han, G., Spitzer, M. H., Bendall, S. C., Fantl, W. J. & Nolan, G. P. Metal-isotope-tagged monoclonal antibodies for high-dimensional mass cytometry. Nat. Protoc. 13, 2121–2148 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Bevins, R. A. & Besheer, J. Object recognition in rats and mice: a one-trial non-matching-to-sample learning task to study ‘recognition memory’. Nat. Protoc. 1, 1306–1311 (2006).

    Article  PubMed  Google Scholar 

  76. Björklund, Å. Single RNA-seq data analysis with R: quality control. Github https://nbisweden.github.io/excelerate-scRNAseq/session-qc/Quality_control.html (2019).

  77. Rachmian, N. & Deitch, D. Trem2_senescent_microglia. Github https://github.com/NoaRachmian/Trem2_senescent_microglia (2024).

Download references

Acknowledgements

We sincerely thank A. Tsitsou-Kampeli, H. Ben-Yehuda, S. Suzzi, G. Castellani, M. A. Abellanas, L. Roitman, H. Gal, N. Papismadov, Y. Addadi and I. Sher for technical assistance. We also thank all members of the Krizhanovsky laboratory and the Schwartz laboratory for helpful discussions. This study was supported by a grant from the Advanced European Research Council (no. 741744); Israel Science Foundation (ISF) research grant 991/16; ISF–Legacy Heritage Biomedical Science Partnership research grant 1354/15; grants from the Thompson Foundation and the Adelis Foundation (given to M.S.); and by grants from the European Research Council H2020 program (no. 856487), the Weizmann Centers for Research on Positive Neuroscience and Research on Neurodegeneration, the ISF (no. 1626/20), the Deutsche Forschungsgemeinschaft (CRC 1506), the Israel Ministry of Health, the Belle S. and Irving E. Meller Center for the Biology of Aging and the Sagol Institute for Longevity Research (given to V.K.). V.K. is an incumbent of the Georg F. Duckwitz Professorial Chair and the Shimon and Golde Picker–Weizmann Award.

Author information

Authors and Affiliations

Authors

Contributions

N.R., M.S. and V.K. conceptualized the project. N.R., M.S. and V.K. planned the experiments. N.R. designed and performed the CyTOF and flow cytometry experiments and the mouse experiments. S.M. performed and analyzed the NOR task. U.C. and N.R. analyzed flow cytometry. U.C. and N.R. injected mice. H.A. analyzed the snRNA-seq data. N.R. and D.D. analyzed the CyTOF results. H.A., D.D. and N.R. performed data analysis. D.E. and N.R. performed and analyzed RT–PCR. N.R., T.C. and T.M.S. established the CyTOF panel. J.M.P.R. established the flow cytometry panel. S.M. and U.C. contributed to the experiment’s performance and experiment design. L.C. organized the mice colonies and performed genotyping. N.R., M.S. and V.K. wrote the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Valery Krizhanovsky or Michal Schwartz.

Ethics declarations

Competing interests

V.K. is a co-inventor of patents on senolytics and senolytic approaches and is a consultant for Sentaur Bio. None of these interests influenced the data presented in this manuscript. M.S. is a scientific co-founder of ImmunoBrain Checkpoint, which develops anti-PD-L1 to treat Alzheimerʼs disease. The other authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Rudolph Tanzi, V. Wee Yong and the other, anonymous, reviewer(s) for their contribution 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 Association between ApoE and senescence markers.

(a) Pearson’s correlation between the expression of ApoE with TREM2 and with senescence markers, p16, p19 and p21. Each color represents a different experiment from Fig. 1; the median expression was Z-scored within each experimental group to account for batch identity.

Extended Data Fig. 2 Differential protein expression between disease-associated microglia (DAM) and senescent microglia.

Differentially expressed proteins between by senescent microglia compared to disease-associated microglia. Each dot represents one protein. The horizontal line marks the significance threshold p < 0.0016 after Bonferroni correction). The vertical dashed lines represent two fold differences in expression.

Extended Data Fig. 3 Proportion of CD45+ cells following senolytic treatment with ABT-737.

(a-g) 10-11 month-old female mice, vehicle control (WT, vehicle) (n = 4), compared to 10-11-month-old 5xFAD female mice, vehicle control (5xFAD, vehicle) (n = 5), and 10-11-month-old 5xFAD female mice, treated with ABT-737 (5xFAD, ABT) (n = 4). Quantitative analysis showing percentage of (a) resting microglia; (b) activated microglia; (c) disease associated microglia; (d) senescent microglia; (e) border-associated macrophages; (f) monocytes; and (g) CD11b- cells among the CNS CD45+ cells. Data are presented as mean values +/- SEM; *P < 0.05, **P < 0.01, ***P < 0.001.

Extended Data Fig. 4 Flow cytometry of splenocytes two weeks following ABT-737 treatment.

12.5-13-month-old male mice, vehicle control (WT, vehicle) (n = 7), compared to 12.5-13-month-old 5xFAD male mice, vehicle control (5xFAD, vehicle; n = 7), and 12.5-13-month-old 5xFAD male mice, treated with ABT-737 (5xFAD, ABT; n = 7). (a) Gating strategy. (b-k) Quantitative analysis by flow cytometry; pink and circles represent the WT group, blue and squares represent the 5xFAD vehicle group, and green and triangles represent 5xFAD ABT group (b) Quantitative analysis of B-cell percentage from total CD45+ cells. (c) Quantitative analysis of myeloid cells percentage from total CD45+ cells. (d) Quantitative analysis of monocyte percentage from total CD45+ cells. (e) Quantitative analysis of neutrophil percentage from total CD45+ cells. (f) Quantitative analysis of T-cell percentage from total CD45+ cells. (g) Quantitative analysis of CD4 + T-cell percentage from total CD45+ cells. (h) Quantitative analysis of naïve T-cell percentage from total CD45+ cells. (i) Quantitative analysis of T-effector cell percentage from total CD45+ cells. (j) Quantitative analysis of T-regulatory cell percentage from total CD45+ cells. (k) Quantitative analysis of CD8 + T-cell percentage from total CD45+ cells. Data are presented as mean values +/- SEM; *P < 0.05, **P < 0.01, ***P < 0.001.

Extended Data Fig. 5 Open Field test.

Two cohorts of mice were combined for the analysis. First cohort of mice: 10-11 month-old female mice, vehicle control (WT, vehicle) (n = 5), compared to 10-11-month-old 5xFAD female mice, vehicle control (5xFAD, vehicle; n = 4), and 10-11-month-old 5xFAD female mice, treated with ABT-737 (5xFAD, ABT; n = 5). Second cohort of mice: 10-11-month-old male mice, vehicle control (WT, vehicle; n = 5), compared to 10-11-month-old 5xFAD male and female, vehicle control (5xFAD, vehicle) (n = 5), and 10-11 month-old 5xFAD male and female mice, treated with ABT-737 (5xFAD, ABT) (n = 7). One-way ANOVA was used for the analyses. Locomotor activity and anxiety were assessed in an open field test and (a) distance moved; (b) mean velocity; (c) cumulative duration in the center zone; (d) frequency entering the center zone; and (e) latency until center entry were recorded, P(WT vs. AD vehicle) = 0.0100, P(WT vs. AD ABT) = 0.8283, P(AD vehicle vs. AD ABT) = 0.0125. Data are presented as mean values +/- SEM; *P < 0.05, **P < 0.01, ***P < 0.001.

Extended Data Fig. 6 Mass cytometry gating strategy.

In the analysis workflow, i) we initially selected CD11b as a stable signal over time and applied gating. ii) Subsequently, gating was performed based on event length and Gaussian residual parameters. iii) To eliminate bead-related interference, the 140Ce bead channel was utilized for gating. iv) Next, live single cells were identified using cisplatin in the 195Pt channel, and iridium DNA labeling in the 193Ir channel, and v) the single-cell population was further refined using two iridium channels. vi) Finally, gating was applied to isolate CD45-positive cells.

Extended Data Table 1 CyTOF panel
Extended Data Table 2 CyTOF small panel

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Table 1: differentially expressed genes of each cluster.

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

Rachmian, N., Medina, S., Cherqui, U. et al. Identification of senescent, TREM2-expressing microglia in aging and Alzheimer’s disease model mouse brain. Nat Neurosci (2024). https://doi.org/10.1038/s41593-024-01620-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41593-024-01620-8

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