Abstract
Aging is a major risk factor for various neurological disorders, including Alzheimer’s disease, and is associated with the accumulation of senescent cells, which can themselves propagate the senescence process through paracrine signaling. Migrasomes are organelles that form during cellular migration, detach from parent cells and mediate intercellular communication. Here we demonstrate that border-associated macrophages (BAMs) acquire senescence-associated properties during early brain aging, possibly due to prolonged exposure to amyloid beta. Senescent-like BAMs show elevated production of migrasomes, which convey senescence-associated signals including the apoptosis inhibitor of macrophage to neighboring cells. We show that microglia are prominent recipients of senescent-like BAM-derived migrasomes, and that through activation of CD16 in recipient cells, the apoptosis inhibitor of macrophage inhibits apoptosis and promotes senescence induction. Blocking migrasome induction in senescent-like BAMs through treatment with Tspan4-targeting siRNA-encapsulated liposomes ameliorates cognitive deficits in aged mice. Our findings suggest that migrasomes are potent vehicles of senescence-regulatory signals and represent a promising target for senomorphic therapy.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
All the databases/datasets used in the study are appropriately accessible via the following accession codes. Raw scRNA-seq data have been deposited in the GEO (accession no. GSE257560). Raw RNA-seq data have been deposited in the Sequence Read Archive (accession no. PRJNA1072693). Public databases GSE208292, GSE175546 and GSE147495 were also used for the data analysis in this study. Source data are provided with this paper. All other data needed to evaluate the conclusions in the paper are present in the paper or are available from the corresponding authors upon request.
Change history
30 September 2025
A Correction to this paper has been published: https://doi.org/10.1038/s43587-025-00994-z
References
Manabe, T. & Heneka, M. T. Cerebral dysfunctions caused by sepsis during ageing. Nat. Rev. Immunol. 22, 444–458 (2022).
Pluvinage, J. V. & Wyss-Coray, T. Systemic factors as mediators of brain homeostasis, ageing and neurodegeneration. Nat. Rev. Neurosci. 21, 93–102 (2020).
Schonhoff, A. M. et al. Border-associated macrophages mediate the neuroinflammatory response in an alpha-synuclein model of Parkinson disease. Nat. Commun. 14, 3754 (2023).
Kim, J. S. et al. A binary cre transgenic approach dissects microglia and CNS border-associated macrophages. Immunity 54, 176–190 (2021).
Foret, M. K. et al. Early oxidative stress and DNA damage in Abeta-burdened hippocampal neurons in an Alzheimer’s-like transgenic rat model. Commun. Biol. 7, 861 (2024).
Hu, Y. et al. Aβ promotes CD38 expression in senescent microglia in Alzheimer’s disease. Biol. Res. 55, 10 (2022).
An, Y., Li, Y., Hou, Y., Huang, S. & Pei, G. Alzheimer’s amyloid-beta accelerates cell senescence and suppresses the SIRT1/NRF2 pathway in human microglial cells. Oxid. Med. Cell Longev. 2022, 3086010 (2022).
Miranda, S. et al. The role of oxidative stress in the toxicity induced by amyloid beta-peptide in Alzheimer’s disease. Prog. Neurobiol. 62, 633–648 (2000).
Giraldo, E., Lloret, A., Fuchsberger, T. & Vina, J. Abeta and tau toxicities in Alzheimer’s are linked via oxidative stress-induced p38 activation: protective role of vitamin E. Redox Biol. 2, 873–877 (2014).
Huang, W., Hickson, L. J., Eirin, A., Kirkland, J. L. & Lerman, L. O. Cellular senescence: the good, the bad and the unknown. Nat. Rev. Nephrol. 18, 611–627 (2022).
Urbanelli, L., Buratta, S., Sagini, K., Tancini, B. & Emiliani, C. Extracellular vesicles as new players in cellular senescence. Int. J. Mol. Sci. 17, 1408 (2016).
Ou, B. et al. Senescent neutrophils-derived exosomal piRNA-17560 promotes chemoresistance and EMT of breast cancer via FTO-mediated m6A demethylation. Cell Death Dis. 13, 905 (2022).
Ma, L. et al. Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration. Cell Res. 25, 24–38 (2015).
Zhang, C. et al. Monocytes deposit migrasomes to promote embryonic angiogenesis. Nat. Cell Biol. 24, 1726–1738 (2022).
Tan, X., He, S., Wang, F., Li, L. & Wang, W. Migrasome, a novel organelle, differs from exosomes. Biochem. Biophys. Rep. 35, 101500 (2023).
Hu, M. et al. Macrophage lineage cells-derived migrasomes activate complement-dependent blood–brain barrier damage in cerebral amyloid angiopathy mouse model. Nat. Commun. 14, 3945 (2023).
Saul, D. et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat. Commun. 13, 4827 (2022).
Ogrodnik, M. et al. Guidelines for minimal information on cellular senescence experimentation in vivo. Cell 187, 4150–4175 (2024).
Suryadevara, V. et al. SenNet recommendations for detecting senescent cells in different tissues. Nat. Rev. Mol. Cell Biol. 25, 1001–1023 (2024).
Zhao, X. et al. Identification of markers for migrasome detection. Cell Discov. 5, 27 (2019).
Chen, L., Ma, L. & Yu, L. WGA is a probe for migrasomes. Cell Discov. 5, 13 (2019).
Jiao, H. et al. Mitocytosis, a migrasome-mediated mitochondrial quality-control process. Cell 184, 2896–2910 (2021).
Papadopoulos, Z., Herz, J. & Kipnis, J. Meningeal lymphatics: from anatomy to central nervous system immune surveillance. J Immunol. 204, 286–293 (2020).
Drieu, A. et al. Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid. Nature 611, 585–593 (2022).
Gerganova, G., Riddell, A. & Miller, A. A. CNS border-associated macrophages in the homeostatic and ischaemic brain. Pharmacol. Ther. 240, 108220 (2022).
Chakraborty, S., ThimmaReddygari, J. & Selvaraj, D. G-lymphatic, vascular and immune pathways for Aβ clearance cascade and therapeutic targets for Alzheimer’s disease. Comb. Chem. High Throughput Screen. 24, 1083–1092 (2021).
Chen, H. et al. Perivascular space in Parkinson’s disease: association with CSF amyloid/tau and cognitive decline. Parkinsonism Relat. Disord. 95, 70–76 (2022).
Gouveia-Freitas, K. & Bastos-Leite, A. J. Perivascular spaces and brain waste clearance systems: relevance for neurodegenerative and cerebrovascular pathology. Neuroradiology 63, 1581–1597 (2021).
Kolbe, S. C. et al. Lesion volume in relapsing multiple sclerosis is associated with perivascular space enlargement at the level of the basal ganglia. AJNR Am. J. Neuroradiol. 43, 238–244 (2022).
Giorgetti, S., Greco, C., Tortora, P. & Aprile, F. A. Targeting amyloid aggregation: an overview of strategies and mechanisms. Int. J. Mol. Sci. 19, 2677 (2018).
Gu, L. & Guo, Z. Alzheimer’s Aβ42 and Aβ40 peptides form interlaced amyloid fibrils. J. Neurochem. 126, 305–311 (2013).
Cao, L. et al. Abeta-induced senescent retinal pigment epithelial cells create a proinflammatory microenvironment in AMD. Invest. Ophthalmol. Vis. Sci. 54, 3738–3750 (2013).
Lewis, S. Mechanics of amyloid clearance. Nat. Rev. Neurosci. 24, 1 (2023).
Shuken, S. R. et al. Limited proteolysis-mass spectrometry reveals aging-associated changes in cerebrospinal fluid protein abundances and structures. Nat. Aging 2, 379–388 (2022).
Castelblanco, E. et al. Circulating CD5L is associated with cardiovascular events and all-cause mortality in individuals with chronic kidney disease. Aging 13, 22690–22709 (2021).
Fang, Y. et al. Association between inflammatory biomarkers and cognitive aging. PLoS ONE 17, e0274350 (2022).
Yi, M. et al. Comparative proteomic analysis identifies biomarkers for renal aging. Aging 12, 21890–21903 (2020).
Sanjurjo, L. et al. The human CD5L/AIM-CD36 axis: a novel autophagy inducer in macrophages that modulates inflammatory responses. Autophagy 11, 487–502 (2015).
Li, J., Lin, W. & Zhuang, L. CD5L-induced activation of autophagy is associated with hepatoprotection in ischemic reperfusion injury via the CD36/ATG7 axis. Exp. Ther. Med. 19, 2588–2596 (2020).
Huang, Y. et al. Migrasome formation is mediated by assembly of micron-scale tetraspanin macrodomains. Nat. Cell Biol. 21, 991–1002 (2019).
Dharan, R. et al. Tetraspanin 4 stabilizes membrane swellings and facilitates their maturation into migrasomes. Nat. Commun. 14, 1037 (2023).
Walton, C. C., Begelman, D., Nguyen, W. & Andersen, J. K. Senescence as an amyloid cascade: the amyloid senescence hypothesis. Front. Cell Neurosci. 14, 129 (2020).
Wardlaw, J. M. et al. Perivascular spaces in the brain: anatomy, physiology and pathology. Nat. Rev. Neurol. 16, 137–153 (2020).
Bordon, Y. Macrophages bordering the brain parenchyma regulate the flow of cerebrospinal fluid. Nat. Rev. Immunol. 23, 3 (2023).
Jiang, D. et al. Migrasomes provide regional cues for organ morphogenesis during zebrafish gastrulation. Nat. Cell Biol. 21, 966–977 (2019).
Gasek, N. S., Kuchel, G. A., Kirkland, J. L. & Xu, M. Strategies for targeting senescent cells in human disease. Nat. Aging 1, 870–879 (2021).
Gurkar, A. U. et al. Spatial mapping of cellular senescence: emerging challenges and opportunities. Nat. Aging 3, 776–790 (2023).
Sanchez-Moral, L. et al. Multifaceted roles of CD5L in infectious and sterile inflammation. Int. J. Mol. Sci. 22, 4076 (2021).
Salminen, A., Ojala, J. & Kaarniranta, K. Apoptosis and aging: increased resistance to apoptosis enhances the aging process. Cell. Mol. Life Sci. 68, 1021–1031 (2011).
Baar, M. P. et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169, 132–147 (2017).
Ni, J., Xie, Z., Quan, Z., Meng, J. & Qing, H. How brain ‘cleaners’ fail: mechanisms and therapeutic value of microglial phagocytosis in Alzheimer’s disease. Glia 72, 227–244 (2024).
Lau, V., Ramer, L. & Tremblay, M. E. An aging, pathology burden, and glial senescence build-up hypothesis for late onset Alzheimer’s disease. Nat. Commun. 14, 1670 (2023).
Greenwood, E. K. & Brown, D. R. Senescent microglia: the key to the ageing brain? Int. J. Mol. Sci. 22, 4402 (2021).
Romashkan, S., Chang, H. & Hadley, E. C. National Institute on Aging Workshop: repurposing drugs or dietary supplements for their senolytic or senomorphic effects: considerations for clinical trials. J. Gerontol. A Biol. Sci. Med. Sci. 76, 1144–1152 (2021).
Saif, M. et al. Non-invasive monitoring of chronic liver disease via near-infrared and shortwave-infrared imaging of endogenous lipofuscin. Nat. Biomed. Eng. 4, 801–813 (2020).
Balogh, E. et al. Pseudouridylation defect due to DKC1 and NOP10 mutations causes nephrotic syndrome with cataracts, hearing impairment, and enterocolitis. Proc. Natl Acad. Sci. USA 117, 15137–15147 (2020).
Luis, T. C. et al. Perivascular niche cells sense thrombocytopenia and activate hematopoietic stem cells in an IL-1 dependent manner. Nat. Commun. 14, 6062 (2023).
Acknowledgements
This work was supported by grants from the National Natural Science Foundation of China (82271348 to W.C.), the Guangdong Basic and Applied Basic Research Foundation (2024B1515020021 to W.C.), the National Natural Science Foundation of China (82471338 to W.C.), the National Natural Science Foundation of China (82471335 to Z.L)., China Postdoctoral Science Foundation (2023M744023 to M.H.), the National Natural Science Foundation of China (82171307 to Z.L.), the Guangzhou Key Research Program on Brain Science (202206060001), the National Natural Science Foundation of China (81971110 to Z.L.), the Guangzhou Science and Technology Program Key Project (202007030010), Guangzhou Municipal School (Hospital) Joint Funding (Dengfeng Hospital) Municipal Key Laboratory Construction Project (202102010009), The Science and Technology Planning Project of Guangdong Province (2023B1212060018) and the Science and Technology program of Guangzhou (202201020588).
Author information
Authors and Affiliations
Contributions
M.H. designed and performed the experiments, collected and analyzed the data, and drafted the paper. X.K. and Z. Liu contributed to the experimental design and revised the paper. S.W. collected human samples. S.L. collected the clinical data and revised the paper. C.L. and D.L. performed the animal experiments and collected the data. Q.Q., H.Y. and L.Y. collected the clinical data and revised the paper. Y.L. contributed to the experimental design and the paper. Q.L., Z. Lu and W.C. designed and supervised the study and critically revised the paper. The authors read and approved the final paper.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Aging thanks the 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 BAMs are the leading cells during brain senescence.
(a-c) Publicly available single-cell RNA sequencing (scRNAseq) datasets (GSE208292 and GSE175546), including transcriptomics of brain cells from 3-, 6- and 12-month-old female wild-type (WT) C57/Bl6J mice, were analyzed. Our scRNAseq data (GSE257560), comprising transcriptomic profiles of brain cells from 4- and 6-month-old male WT C57/Bl6J mice, was integrated with the public dataset GSE147495, which includes transcriptomics of brain cells from 10-month-old male WT C57/Bl6J mice. (a) Cellular senescence of brain cells from 3-, 6- and 12-month-old female mice (GSE208292 and GSE175546) (left) and 4-, 6- and 10-month-old male mice (GSE257560 and GSE147495) (right) was assessed using the SenMayo gene set, with cell types ranked by gene expression levels. **P < 0.01, ***P < 0.001, compared to 3-month-old female mice; #P < 0.05, ##P < 0.01, ###P < 0.001, compared to 4-month-old male mice; by one-way ANOVA. (b) tSNE plots showing the expression of SenMayo genes (calculated with the Function of Addmodulescore in R) among brain cells of 3-, 6- and 12-month-old female mice (GSE208292 and GSE175546) (upper) and 4-, 6- and 10-month-old male mice (GSE257560 and GSE147495) (lower). (c) Dot plots showing the expression of SASPs in BAM from 3-, 6- and 12-month-old female mice (GSE208292 and GSE175546) (upper) and 4-, 6- and 10-month-old male mice (GSE257560 and GSE147495) (lower). (d) Naïve female and male wild type (WT) C57/BL6J mice were sacrificed at indicated age. Left: Coronal brain sections from female and male mice at 3, 6, 12 and 18 months were immunostained for the BAM marker CD206 and SASPs (TNFα or IFNγ). Experiments were conducted four times, and representative confocal microscopic images are shown. Single channel images were shown in Supplementary Fig. 1a. Right: Expression of TNFα or IFNγ in BAMs (CD206+) from 3, 6, 12 and 18-month-old female and male mice was analyzed via flow cytometry. Data are presented as mean values +/- SEM. N = 5 per group. *P < 0.05, **P < 0.01, ***P < 0.001, compared to 3-month-old female mice; ##P < 0.01, ###P < 0.001, compared to 3-month-old male mice; by one-way ANOVA. (e) Heat maps showing the expression of markers of cellular senescence in BAM from 3-, 6-, and 12-month-old female mice (GSE208292 and GSE175546) (upper) and 4-, 6- and 10-month-old male mice (GSE257560 and GSE147495) (lower). (f) The number of BAMs (CD206+) from 3, 6, 12 and 18-month-old female and male mice was quantified with flow cytometry. Data are presented as mean values +/- SEM. N = 3 per group. *P < 0.05, **P < 0.01, compared to 3-month-old female mice; #P < 0.05, ##P < 0.01, compared to 3-month-old male mice; by one-way ANOVA. (g-i) Coronal brain sections from female (g) and male (h) mice at 3, 6, 12 and 18 months were subjected to fluorescence in situ hybridization (FISH) for Cdkn2a, followed by co-staining with markers for neurons (NeuN+), astrocytes (GFAP+), oligodendrocytes (CC1+) and microglia (IBA1+). Single channel images were shown in Supplementary Fig. 1d. Senescence was not evident in these cells until 12 months of age. Experiments were conducted four times, and representative confocal microscopic images are displayed. (i) Coronal brain sections from female and male mice at 3, 6, 12 and 18 months of age were subjected to fluorescence in situ hybridization (FISH) for Cdkn2a. The percentages of different cell types (NeuN+ neurons, GFAP+ astrocytes, CC1+ oligodendrocytes and IBA1+ microglia) with Cdkn2a expression at different ages were calculated. Data are presented as mean values +/- SEM. N = 6 per group. ***P < 0.001, compared to 3-month-old female mice; ###P < 0.001, compared to 3-month-old male mice; by two-way ANOVA.
Extended Data Fig. 2 The in situ expression of cellular senescence markers is elevated in 12-month-old neurons, astrocytes, oligodendrocytes and microglia.
(a-d) Coronal brain sections from male mice at 3, 6, 12 and 18 months were subjected to analysis of LMNB1, HMGB1, p-γH2AX, Cenpb, p-STAT3, PLIN2, lipofuscin and SA-β-Gal, followed by co-staining with the marker for neurons (TUJ1, a), astrocytes (GFAP, b), oligodendrocytes (CC1, c) and microglia (IBA1, d). Representative images were displayed. Single channel images were shown in Supplementary Fig. 2. The locations of neurons and astrocytes were demarcated by red dashed lines in images showing intracellular lipofuscin and SA-β-Gal expression. Data are presented as mean values +/- SEM. N = 6 per group. *P < 0.05, **P < 0.01, ***P < 0.001; by one-way ANOVA.
Extended Data Fig. 3 Relevant correlation analysis of the community cohort, and analysis of Aβ40 accumulation in BAMs of mice.
(a-c) A total of 80 individuals (age = 23–82y) from community population were recruited for the study. (a) Inclusion and exclusion process of the community cohort. (b-c) In the first batch of cohort recruitment, blood samples from 52 individuals (age = 55–82 years) were collected. (b) The association between the percentage of TSPAN4-expressing monocytes or TSPAN4 MFI in monocytes and age, along with the listed parameters in the recruited cohort, was assessed using Spearman correlation analysis. *P < 0.05, **P < 0.01. (c) The association between the percentage of AIM-expressing monocytes or AIM MFI in monocytes and age, along with the listed parameters in the recruited cohort, was assessed using Spearman correlation analysis. *P < 0.05. (d) Coronal brain sections from naïve wild-type (WT) or Tg-SwDI/B+/+ mice of the indicated ages were subjected to Thioflavin-S staining to visualize amyloid tangle accumulation. Experiments were performed four times, and representative confocal microscopic images are shown. (e) The retention of Aβ42, APOE, α-SYN, and MBP in leptomeningeal BAM was assessed using immunostaining. Single channel images were shown in Supplementary Fig. 3c. The percentage of BAMs in the leptomeninges engulfing these substances was calculated, with statistical data presented in Fig. 3a. Experiments were conducted on brain slices from four independent mice, and representative images depicting the accumulation of these substances in leptomeningeal BAMs are displayed. (f) Fluorescent Aβ40-TAMRA (5 μl, 5 mg/ml) was injected into healthy young WT male mice (3 months old) via the cisterna magna (i.c.m.). Mice were sacrificed one day post-injection. Aβ40 engulfment by monocyte-derived macrophages (MoM, F4/80+), microglia (TMEM119+) and BAM (CD206+) was quantified using flow cytometry. Data are presented as mean values +/- SEM. N = 3 in each group. *P < 0.05, **P < 0.01, by one-way ANOVA.
Extended Data Fig. 4 Aβ40 induces cellular senescence of mouse bone marrow derived macrophages (BMDM).
BMDM were cultured from cells isolated from the femurs and tibiae of healthy wild-type (WT) C57/BL6 donors (2–3 months old). Macrophage precursors were cultivated and differentiated into macrophages over 6 days in macrophage colony-stimulating factor (MCSF)-conditioned medium. On day 7, BMDM cultures were subjected to in vitro experiments and treated with Aβ40 (20 μg/ml) for the indicated time period. PBS treatment was used as control. (a) Experimental schematics. (b) The cell cycle of BMDMs was assessed using Ki67 and DAPI staining with flow cytometry. The percentage of cells in the G0 phase (Ki67−DAPI−), G1 phase (Ki67+DAPI−) and S-G2-M phase (Ki67+DAPI+) was calculated. Data are presented as mean values +/- SEM. Experiments were conducted five times. *P < 0.05, **P < 0.01, ***P < 0.001 compared with control group; by two-way ANOVA. (c) BMDMs treated with Aβ40 were stained with phalloidin and DAPI to outline the cell body and nucleus. The volumes of the cell and nucleus were calculated. Data are presented as mean values +/- SEM. Experiments were conducted five times. *P < 0.05, ***P < 0.001 compared with control group; by two-way ANOVA. (d) BMDM cultures were subjected to SA-β-Gal labeling. Data are presented as mean values +/- SEM. Experiments were repeated three times. ***P < 0.001, compared with control group; by two-way ANOVA. (e) Telomere length was assessed using a telomere probe (C-rich) (TELC) and flow cytometry. Data are presented as mean values +/- SEM. Experiments were repeated four times. ***P < 0.001, by two-way ANOVA, compared with control group. (f) QPCR analysis of telomere length in BMDMs. Data are presented as mean values +/- SEM. Experiments were repeated five times. *P < 0.05, **P < 0.01, ***P < 0.001, by two-way ANOVA, compared with control group. (g) BMDMs were cultured and treated with Aβ40 (20 μg/ml) for 1 day. The BMDM cultures were subjected to western blot analysis to assess the expression of the indicated senescence markers. (h) BMDM cultures treated with Aβ40 (20 μg/ml) for indicated time period were subjected to QPCR. Experiments were repeated four times. *P < 0.05, **P < 0.01, ***P < 0.001, compared with the 0h group; by one-way ANOVA. (i) BMDM cultures treated with Aβ40 (20 μg/ml) for indicated time period were subjected to staining for Cdkn2a, P21, LMNB1, HMGB1, p-γH2AX, Cenpb, p-STAT3, PLIN2, lipofuscin, followed by co-staining with phalloidin. Experiments were repeated three times. Representative images were displayed. Single channel images were shown in Supplementary Fig. 4a. (j) BMDM cultures treated with Aβ40 for the indicated time periods were co-cultured with organotypic brain slices (OSC) for 1 day. The cell cycle of OSC cells was assessed using Ki67 and DAPI staining. The percentage of cells in the G0 phase (Ki67−DAPI−), G1 phase (Ki67+DAPI−) and S-G2-M phase (Ki67+DAPI+) was calculated. Data are presented as mean values +/- SEM. Experiments were conducted four times. *P < 0.05, **P < 0.01; compared 0h group; by one-way ANOVA.
Extended Data Fig. 5 Aβ40 induces cellular senescence of RAW264.7 and human macrophages.
(a-b) RAW264.7 cells were subjected to in vitro experiments and treated with Aβ40 (20 μg/ml) for the indicated time period. (a) Telomere length was assessed using a telomere probe (C-rich) (TELC) and flow cytometry. Data are presented as mean values +/- SEM. Experiments were repeated four times. **P < 0.001, ***P < 0.001, by one-way ANOVA, compared with the 0h group. (b) RAW264.7 treated with Aβ40 (20 μg/ml) for indicated time period were subjected to immunostaining for LMNB1, HMGB1, PLIN2. Experiments were conducted three times. Representative images were displayed. Single channel images were shown in Supplementary Fig. 4b. (c-f) Human macrophages were derived from blood monocytes donated by healthy young adults (age = 20–30 years) using human serum and MCSF. (c) Human macrophages were treated with PBS for indicated time points then subjected to analysis of LMNB1, HMGB1, p-γH2AX, pSTAT3, and SA-β-Gal, followed by co-staining with phalloidin. Single channel images were shown in Supplementary Fig. 4c. Experiments were conducted four times. (d) The cell cycle of the human macrophage cultures was analyzed using Ki67 and DAPI staining via flow cytometry. (e) Telomere length was assessed using a telomere probe (C-rich) (TELC) and flow cytometry. (f) The cultures were labeled with WGA-AF488, and migrasomes produced by macrophages were quantified.
Extended Data Fig. 6 Migrasomes are accessible to brain cells and could induce senescence in organotypic brain slice cultures (OSCs) by AIM.
(a-e) Brain migrasomes from 3- and 18-month-old healthy WT male mice were isolated through serial centrifugation. BAM-derived migrasomes (CD206+) were further enriched via magnetic isolation (a) The membrane structures of the iosolated migrasomes were observed by TEM after negative staining. Red arrowheads were used to indicate magnetic beads associated with migrasomes. (b) Diameter distribution of isolated migrasomes. Data represent three biologically independent experiments. (c) Expression of migrasome markers in migrasomes was assessed via western blot. Data represent three biologically independent experiments. (d) BAM-derived migrasomes from 3- and 18-month-old healthy WT male mice were labeled with DIL. The DIL+ migrasomes were administered to 3-month-old healthy male recipients (0.25 mg/kg, i.c.m). Seven days post-migrasome transfer, coronal brain sections of the recipients were examined using microscopy. The penetration of migrasomes from the perivascular space (PVS) into the brain parenchyma was recorded. Experiments were conducted four times. (e) BAM-derived migrasomes from 3- and 18-month-old healthy WT male mice were treated to OSCs (50 μg/ml, 24 h). The membrane structures of the iosolated migrasomes were observed by TEM after negative staining. The cell cycle of brain cells in OSCs was assessed using Ki67 and DAPI staining with flow cytometry. The percentages of cells in the G0 phase (Ki67−DAPI−), G1 phase (Ki67+DAPI−), and S-G2-M phase (Ki67+DAPI+) were calculated. Data are presented as mean values +/- SEM. Experiments were repeated five times. **P < 0.01, compared to magnetic beads group; by one-way ANOVA. (f-g) OSCs were treated with AIM (100 ng/ml, 24 h) or equal volume of PBS. (f) OSCs were subjected to western blot analysis to assess the listed senescence markers. Data are presented as mean values +/- SEM. Experiments were repeated four times. *P < 0.05, **P < 0.01, by Student’s t-test. (g) OSCs were subjected to SA-β-Gal labeling. Data are presented as mean values +/- SEM. Experiments were repeated six times. ***P < 0.001, by Student’s t-test.
Extended Data Fig. 7 Aβ40-induced migrasomes promote senescence of microglia.
(a-g) Bone marrow-derived macrophages (BMDMs) were cultured from 2-month-old male mice and stimulated with Aβ40 (20 μg/ml, 24 h). Migrasomes derived from PBS- or Aβ40-treated BMDMs were collected. The PBS-M and Aβ40-M were administered to primary microglia cultures (50 μg/ml) for the indicated time periods. (a) The cell cycle of microglia cultures was assessed using Ki67 and DAPI staining with flow cytometry. The percentages of microglia in the G0 phase (Ki67−DAPI−), G1 phase (Ki67+DAPI−) and S-G2-M phase (Ki67+DAPI+) were calculated. Data are presented as mean values +/- SEM. Experiments were conducted four times. **P < 0.01, ***P < 0.001, compared to PBS group; by one-way ANOVA. (b) The microglia cultures were subjected to Sudan Black staining to reveal the accumulation of lipofuscin. Experiments were repeated three times. (c) The microglia cultures were subjected to SA-β-Gal labeling. Experiments were repeated three times. (d) Telomere length was assessed using a C-rich telomere probe (TELC) and flow cytometry. Data are presented as mean values +/- SEM. Experiments were repeated three times. **P < 0.01, ***P < 0.001, compared with the PBS-M group; by two-way ANOVA. (e) The microglia cultures were subjected to western blot analysis for the indicated senescence markers. Data are presented as mean values +/- SEM. Experiments were repeated three times. *P < 0.05, compared to PBS group; by one-way ANOVA. (f-g) The microglia cultures were subjected to flow cytometric analysis for the senescence marker P21 (f) and the SASPs of TNFα and IFNγ (g). Data are presented as mean values +/- SEM. Experiments were conducted four times. *P < 0.05, **P < 0.01, ***P < 0.001; by one-way ANOVA. (h-m) BMDMs were cultured and treated with Aβ40 (20 μg/ml, 24 h). The migrasomes (50 μg/ml), exosomes (50 μg/ml), and soluble substances (50%) were collected and used to treat the microglial cultures for 24 h. (h) Representative transmission electron microscopic (TEM) images of negatively stained exosomes are shown. (i) Diameter distribution of PBS-induced and Aβ40-induced exosomes. Data represent three biologically independent experiments. (j) Expression of exosome markers in PBS-induced and Aβ40-induced exosomes was assessed via western blot. Data represent three biologically independent experiments. (k) The cell cycle of microglia cultures treated with migrasomes, exosomes, or solutes was assessed using Ki67 and DAPI staining with flow cytometry. The percentages of microglia in the G0 phase (Ki67−DAPI−), G1 phase (Ki67+DAPI−) and S-G2-M phase (Ki67+DAPI+) were calculated. Data are presented as mean values +/- SEM. Experiments were conducted four times. **P < 0.01, ***P < 0.001, compared to PBS group; by one-way ANOVA. (l) The microglia cultures treated with migrasomes, exosomes, or solutes were subjected to SA-β-Gal labeling. Experiments were repeated three times. (m) The microglia cultures treated with migrasomes, exosomes, or solutes were subjected to western blot analysis to assess the listed senescence markers. Experiments were repeated three times.
Extended Data Fig. 8 AIM in migrasomes induces microglial senescence.
(a) Primary microglia were cultured and treated with AIM (100 ng/ml, 24h) or an equal volume of PBS. The microglia cultures were then subjected to Sudan Black staining to reveal the accumulation of lipofuscin. Experiments were repeated three times. (b-h) Cd5l−/− () RAW264.7 macrophages were generated and stimulated with Aβ40 (20 μg/ml, 24 h). Migrasomes derived from Aβ40-treated AIM KO macrophages (KO Aβ40-M) were collected and used to treat microglia cultures (50 μg/ml, 24 h). (b) The cell cycle of microglia cultures was assessed using Ki67 and DAPI staining with flow cytometry. The percentages of microglia in the G0 phase (Ki67−DAPI−), G1 phase (Ki67+DAPI−) and S-G2-M phase (Ki67+DAPI+) were calculated. Data are presented as mean values +/- SEM. Experiments were conducted four times. **P < 0.01, ***P < 0.001, compared to WT PBS-M group; by one-way ANOVA. (c) The microglia cultures were subjected to Sudan Black staining to reveal the accumulation of lipofuscin. Experiments were repeated three times. (d) The microglia cultures were subjected to SA-β-Gal labeling. Experiments were repeated three times. (e) Telomere length was assessed using a C-rich telomere probe (TELC) and flow cytometry. Data are presented as mean values +/- SEM. Experiments were repeated three times. *P < 0.05, by one-way ANOVA, compared with the 0h group. (f) The microglia cultures were subjected to western blot analysis for the indicated senescence markers. Experiments were repeated four times. (g-h) The microglia cultures were subjected to flow cytometric analysis for the senescence marker P21 (g) and the SASPs of TNFα and IFNγ (h). Data are presented as mean values +/- SEM. Experiments were conducted four times. ***P < 0.001, compared to WT PBS-M group; by one-way ANOVA. (i) Our scRNAseq data (GSE257560), comprising transcriptomic profiles of brain cells from 4- and 6-month-old male WT C57/Bl6J mice, was integrated with the public dataset GSE147495, which includes transcriptomics of brain cells from 10-month-old male WT C57/Bl6J mice. Expression of Fcgr3 in microglia from 10m-old mice and 4m-old mice was analyzed. ***P < 0.001, by Student’s t-test, compared with the 4-month-old mice. (j-k) Mouse bone marrow-derived macrophages (BMDMs) were cultured and treated with Aβ40 (20 μg/ml, 24 h). The migrasome products were collected and administered to brain organotypic slice cultures (OSCs) or primary microglia cultures (50 μg/ml). (j) The OSCs were treated with Aβ40-M or PBS-M for 24 h and then subjected to western blot analysis to assess the expression of CD16. Experiments were repeated three times. (k) Primary microglia cultures were treated with Aβ40-M or PBS-M for the indicated time period. The cultures were then subjected to western blot analysis to assess the expression of CD16. Data are presented as mean values +/- SEM. Experiments were repeated three times. *P < 0.05, compared to PBS group; by one-way ANOVA.
Extended Data Fig. 9 Senescence-associated migrasomes propagate senescence signals by activating CD16 on recipient cells.
(a-b) Primary microglia cultures were treated with Aβ40-M or PBS-M for 24 h, with the addition of IgG or anti-CD16 neutralizing antibodies (BD Pharmingen 553142, clone: 2.4G2, 0.5 μg/ml) and/or anti-CD36 neutralizing antibodies (Abcam ab23680, clone: JC63.1, 0.1 μg/ml). The cultures were subjected to western blot analysis to assess the listed senescence markers (a) or Sudan Black staining to reveal the accumulation of lipofuscin (b). Data are presented as mean values +/- SEM. Experiments were repeated three times. Experiments were repeated three times. *P < 0.05, **P < 0.01, ***P < 0.001, compared to PBS IgG group; by one-way ANOVA. (c) Cd5l−/− (AIM KO) RAW264.7 macrophages were generated and stimulated with Aβ40 (20 μg/ml, 24 h). Migrasomes derived from Aβ40-treated AIM KO macrophages were collected and administered to microglia cultures (50 μg/ml, 24 h). WT Aβ40-M were also administered to the cultures along with IgG or anti-CD16 neutralizing antibodies (BD Pharmingen 553142, clone: 2.4G2, 0.5 μg/ml). The cultures were then subjected to western blot analysis to assess the listed senescence markers. Data are presented as mean values +/- SEM. Experiments were repeated three times. *P < 0.05, **P < 0.01, ***P < 0.001, by one-way ANOVA.
Extended Data Fig. 10 Suppressing migrasome production by BAM attenuates the pro-inflammatory properties and enhances the scavenging activity of microglia.
A total of 2 nmol of Tspan4 siRNA was encapsulated in 25 μg of liposomes, dissolved in 5 μl of PBS, and injected into 18-month-old WT female and male mice (i.c.m.) to suppress migrasome production by BAM. Mice injected with equal amount of empty liposomes served as controls. The mice were sacrificed 7 days post-injection. (a) Schematic of the experimental design. (b) Brain cells were analyzed by flow cytometry for TSPAN4 expression in BAM (CD206+), microglia (TMEM119+), astrocyte (GFAP+), oligodendrocyte (O4+) and neuron (TUJ1+). Data are presented as mean values +/- SEM. N = 8 in each group. ***P < 0.001; by Student’s t-test. (c) Coronal brain sections were subjected to immunostaining to assess TSPAN4 expression in BAM (CD206+), microglia (IBA1+), astrocyte (GFAP+), oligodendrocyte (CC1+) and neuron (TUJ1+). Representative confocal microscopy images are shown. Single channel images were shown in Supplementary Fig. 8c. Data are representative of three biologically independent experiments. (d) Brain vesicles were isolated by serial centrifugation and analyzed by flow cytometry. Purified migrasomes derived from BMDM in vitro were used as FSC/SSC scale standards. Migrasomes were identified as TSPAN4+ particles. The percentage of total migrasomes (TSPAN4+) derived from BAM (CD206+), microglia (TMEM119+), astrocyte (GFAP+), oligodendrocyte (O4+) and neuron (TUJ1+) were recorded. Data are presented as mean values +/- SEM. N = 6 in each group. ***P < 0.001, by Student’s t-test. (e) Coronal brain sections from female and male recipients were subjected to fluorescence in situ hybridization (FISH) for Cdkn2a, followed by co-staining with the microglia marker IBA1. Single channel images were shown in Supplementary Fig. 8d. The percentage of microglia expressing Cdkn2a was calculated. Data are presented as mean values +/- SEM. N = 4 in each group. ***P < 0.001, in female mice; ###P < 0.001, in male mice; by Student’s t-test. (f-i) Coronal brain sections from female and male recipients were subjected to immunostaining for IBA1/TNFα (f), IBA1/IFNγ (g), IBA1/Aβ (clone: 6E10) (h) and IBA1/MBP (i). Single channel images were shown in Supplementary Fig. 8e–h. Data are presented as mean values +/- SEM. N = 6 in each group. ***P < 0.001, in female mice; ###P < 0.001, in male mice; by Student’s t-test.
Supplementary information
Supplementary Information
Supplementary Methods, Tables 1–4 and Figs. 1–9.
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. 4
Unprocessed western blots.
Source Data Fig. 5
Statistical source data.
Source Data Fig. 5
Unprocessed western blots.
Source Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 1
Statistical source data.
Source Data Extended Data Fig. 2
Statistical source data.
Source Data Extended Data Fig. 3
Statistical source data.
Source Data Extended Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 5
Statistical source data.
Source Data Extended Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 6
Unprocessed western blots.
Source Data Extended Data Fig. 7
Statistical source data.
Source Data Extended Data Fig. 7
Unprocessed western blots.
Source Data Extended Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 8
Unprocessed western blots.
Source Data Extended Data Fig. 9
Statistical source data.
Source Data Extended Data Fig. 9
Unprocessed western blots.
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.
About this article
Cite this article
Hu, M., Kang, X., Liu, Z. et al. Senescent-like border-associated macrophages regulate cognitive aging via migrasome-mediated induction of paracrine senescence in microglia. Nat Aging (2025). https://doi.org/10.1038/s43587-025-00956-5
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s43587-025-00956-5