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:

Central nervous system-associated macrophages modulate the immune response following stroke in aged mice

Abstract

Age is a major nonmodifiable risk factor for ischemic stroke. Central nervous system-associated macrophages (CAMs) are resident immune cells located along the brain vasculature at the interface between the blood circulation and the parenchyma. By using a clinically relevant thromboembolic stroke model in young and aged male mice and corresponding human tissue samples, we show that during aging, CAMs acquire a central role in orchestrating immune cell trafficking after stroke through the specific modulation of adhesion molecules by endothelial cells. The absence of CAMs provokes increased leukocyte infiltration (neutrophils and CD4+ and CD8+ T lymphocytes) and neurological dysfunction after stroke exclusively in aged mice. Major histocompatibility complex class II, overexpressed by CAMs during aging, plays a significant role in the modulation of immune responses to stroke. We demonstrate that during aging, CAMs become central coordinators of the neuroimmune response that ensure a long-term fine-tuning of the immune responses triggered by stroke.

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: Age-associated immune responses and BBB leakage in the naive mouse brain occur without changes in the number or morphology of CAMs.
Fig. 2: CAMs change their transcriptomic phenotype during aging and overexpress genes implicated in the regulation of innate and adaptive immune responses, including antigen presentation and leukocyte cell–cell adhesion.
Fig. 3: Upregulation of HLA and CD74 expression by CAMs in the cortex of older humans.
Fig. 4: CAM depletion increases functional deficit after stroke in aged mice but not in young mice.
Fig. 5: CAM depletion increases the expression of the P-sel adhesion molecule and leukocyte trafficking in cerebral vessels after stroke in aged mice but not in young mice.
Fig. 6: CAM depletion increases neutrophil and CD4+ and CD8+ T cell infiltration after stroke in aged mice.
Fig. 7: Blockade of MHC class II mimics some of the features of global CAM depletion by CLO treatment in aged mice.

Similar content being viewed by others

Data availability

Mouse macrophage RNA-seq data are available at the Gene Expression Omnibus under accession number GSE267623. All data necessary for the conclusions of the study are available in the main text, figures and Extended Data figures. Source data are provided with this paper.

References

  1. Iadecola, C., Buckwalter, M. S. & Anrather, J. Immune responses to stroke: mechanisms, modulation, and therapeutic potential. J. Clin. Invest. 130, 2777–2788 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Drieu, A. et al. Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid. Nature 611, 585–593 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Amann, L., Masuda, T. & Prinz, M. Mechanisms of myeloid cell entry to the healthy and diseased central nervous system. Nat. Immunol. 24, 393–407 (2023).

    CAS  PubMed  Google Scholar 

  4. Kida, S., Steart, P. V., Zhang, E. T. & Weller, R. O. Perivascular cells act as scavengers in the cerebral perivascular spaces and remain distinct from pericytes, microglia and macrophages. Acta Neuropathol. 85, 646–652 (1993).

    Article  CAS  PubMed  Google Scholar 

  5. Mendes-Jorge, L. et al. Scavenger function of resident autofluorescent perivascular macrophages and their contribution to the maintenance of the blood–retinal barrier. Invest. Ophthalmol. Vis. Sci. 50, 5997–6005 (2009).

    Article  PubMed  Google Scholar 

  6. Fabriek, B. O. et al. CD163-positive perivascular macrophages in the human CNS express molecules for antigen recognition and presentation. Glia 51, 297–305 (2005).

    Article  PubMed  Google Scholar 

  7. Faraco, G. et al. Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension. J. Clin. Invest. 126, 4674–4689 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Park, L. et al. Brain perivascular macrophages initiate the neurovascular dysfunction of Alzheimer Aβ peptides. Circ. Res. 121, 258–269 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pedragosa, J. et al. CNS-border associated macrophages respond to acute ischemic stroke attracting granulocytes and promoting vascular leakage. Acta Neuropathol. Commun. 6, 76 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Drieu, A. et al. Alcohol exposure-induced neurovascular inflammatory priming impacts ischemic stroke and is linked with brain perivascular macrophages. JCI Insight 5, e129226 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Popa-Wagner, A. et al. Ageing as a risk factor for cerebral ischemia: underlying mechanisms and therapy in animal models and in the clinic. Mech. Ageing Dev. 190, 111312 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Virani, S. S. et al. Heart disease and stroke statistics-2020 update: a report from the American Heart Association. Circulation 141, e139–e596 (2020).

    Article  PubMed  Google Scholar 

  13. Mozaffarian, D. et al. Heart disease and stroke statistics-2016 update: a report from the American Heart Association. Circulation 133, e38–e360 (2016).

    PubMed  Google Scholar 

  14. Franceschi, C., Garagnani, P., Parini, P., Giuliani, C. & Santoro, A. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 14, 576–590 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Jordão, M. J. C. et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 363, eaat7554 (2019).

    Article  PubMed  Google Scholar 

  16. Prinz, M., Masuda, T., Wheeler, M. A. & Quintana, F. J. Microglia and central nervous system-associated macrophages-from origin to disease modulation. Annu. Rev. Immunol. 39, 251–277 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Masuda, T. et al. Specification of CNS macrophage subsets occurs postnatally in defined niches. Nature 604, 740–748 (2022).

    Article  CAS  PubMed  Google Scholar 

  18. Orset, C. et al. Mouse model of in situ thromboembolic stroke and reperfusion. Stroke 38, 2771–2778 (2007).

    Article  PubMed  Google Scholar 

  19. Gauberti, M., Fournier, A. P., Docagne, F., Vivien, D. & Martinez de Lizarrondo, S. Molecular magnetic resonance imaging of endothelial activation in the central nervous system. Theranostics 8, 1195–1212 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Martinez de Lizarrondo, S. et al. Tracking the immune response by MRI using biodegradable and ultrasensitive microprobes. Sci. Adv. 8, eabm3596 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fournier, A. P. et al. Prediction of disease activity in models of multiple sclerosis by molecular magnetic resonance imaging of P-selectin. Proc. Natl Acad. Sci. USA 114, 6116–6121 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mato, M. et al. Involvement of specific macrophage-lineage cells surrounding arterioles in barrier and scavenger function in brain cortex. Proc. Natl Acad. Sci. USA 93, 3269–3274 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Kierdorf, K., Masuda, T., Jordão, M. J. C. & Prinz, M. Macrophages at CNS interfaces: ontogeny and function in health and disease. Nat. Rev. Neurosci. 20, 547–562 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Se, H. et al. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 9, 1512–1519 (2006).

    Article  Google Scholar 

  26. 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 

  27. 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 

  28. Knox, E. G., Aburto, M. R., Clarke, G., Cryan, J. F. & O’Driscoll, C. M. The blood–brain barrier in aging and neurodegeneration. Mol. Psychiatry 27, 2659–2673 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yousef, H. et al. Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nat. Med. 25, 988–1000 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Paolicelli, R. C. et al. Microglia states and nomenclature: a field at its crossroads. Neuron 110, 3458–3483 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schonhoff, A. M. et al. Border-associated macrophages mediate the neuroinflammatory response in an α-synuclein model of Parkinson disease. Nat. Commun. 14, 3754 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Garcia-Bonilla, L. et al. Analysis of brain and blood single-cell transcriptomics in acute and subacute phases after experimental stroke. Nat. Immunol. 25, 357–370 (2024).

  33. Frosch, M., Amann, L. & Prinz, M. CNS-associated macrophages shape the inflammatory response in a mouse model of Parkinson’s disease. Nat. Commun. 14, 3753 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Drieu, A. et al. Parenchymal border macrophages regulate tau pathology and tau-mediated neurodegeneration. Life Sci. Alliance 6, e202302087 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Schmidt, A. et al. Targeting different monocyte/macrophage subsets has no impact on outcome in experimental stroke. Stroke 48, 1061–1069 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Drieu, A., Levard, D., Vivien, D. & Rubio, M. Anti-inflammatory treatments for stroke: from bench to bedside. Ther. Adv. Neurol. Disord. 11, 1756286418789854 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Vogelgesang, A. et al. Siponimod (BAF312) treatment reduces brain infiltration but not lesion volume in middle-aged mice in experimental stroke. Stroke 50, 1224–1231 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Llovera, G. et al. The choroid plexus is a key cerebral invasion route for T cells after stroke. Acta Neuropathol. 134, 851–868 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Benakis, C. et al. T cells modulate the microglial response to brain ischemia. eLife 11, e82031 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Roth, S., Yang, J., Cramer, J. V., Malik, R. & Liesz, A. Detection of cytokine-induced sickness behavior after ischemic stroke by an optimized behavioral assessment battery. Brain Behav. Immun. 91, 668–672 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Iadecola, C. et al. Cell autonomous role of border associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. Preprint at Research Square https://doi.org/10.21203/rs.3.rs-3222611/v1 (2023).

  42. Gervois, P. & Lambrichts, I. The emerging role of triggering receptor expressed on myeloid cells 2 as a target for immunomodulation in ischemic stroke. Front. Immunol. 10, 1668 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Atagi, Y. et al. Apolipoprotein E is a ligand for triggering receptor expressed on myeloid cells 2 (TREM2). J. Biol. Chem. 290, 26043–26050 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Krumbholz, M. et al. Chemokines in multiple sclerosis: CXCL12 and CXCL13 up-regulation is differentially linked to CNS immune cell recruitment. Brain 129, 200–211 (2006).

    Article  PubMed  Google Scholar 

  45. Polfliet, M. M. J. et al. The role of perivascular and meningeal macrophages in experimental allergic encephalomyelitis. J. Neuroimmunol. 122, 1–8 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Alves de Lima, K., Rustenhoven, J. & Kipnis, J. Meningeal immunity and its function in maintenance of the central nervous system in health and disease. Annu. Rev. Immunol. 38, 597–620 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Lehenkari, P. P. et al. Further insight into mechanism of action of clodronate: inhibition of mitochondrial ADP/ATP translocase by a nonhydrolyzable, adenine-containing metabolite. Mol. Pharmacol. 61, 1255–1262 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Quenault, A. et al. Molecular magnetic resonance imaging discloses endothelial activation after transient ischaemic attack. Brain 140, 146–157 (2017).

    Article  PubMed  Google Scholar 

  49. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bindea, G. et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25, 1091–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This project has received funding from the European Union’s Horizon 2020 research and innovation programme under Marie Skłodowska-Curie grant agreement number 813294 (ENTRAIN). This work was also supported by grants from the Ministère de l’Enseignement Supérieur et de la Recherche and INSERM (French National Institute for Health and Medical Research; HCERES U1237-2017/2022), Throne (ANR-22-CE14-002), Eranet-Neuron MeniPSYs (ANR-22-NEU2-0005) and the Fondation pour la Recherche Médicale (ARF202005011926; E.L.). The work of A.M. is supported by the UK Dementia Research Institute (DRI), which receives its funding from UK DRI, funded by the UK Medical Research Council, Alzheimer’s Society and Alzheimer’s Research UK. A.M. also holds a UKRI Medical Research Council fellowship (Career Development Award MR/V032488/1) and a Foundation for Research on Alzheimer’s disease award. A.P.F. is funded by the WINNINGNormandy Program supported by the Normandy Region and the European Union’s Horizon 2020 research and innovation programme under Marie Skłodowska-Curie grant agreement number 101034329. Some illustrations were created using BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

D.L. designed and performed the experiments, analyzed and interpreted the data, created the figures and wrote the manuscript. C. Seillier performed the flow cytometry experiments. M.B.-S. performed the quantification of human sample staining and analyzed and interpreted the data. A.P.F. performed the molecular MRI experiments. E.L. and C.D. performed immunohistological experiments and data analyses. G.R. performed the cell sorting experiments. A.M., K.M., C. Smith and C.M. performed the human sample staining experiments and participated in methods writing. A.M., M.P. and D.V. provided intellectual contributions. L.A. and M.P. performed the mouse RNA-seq data analyses and participated in methods writing. D.V. provided resources and intellectual contributions. M.R. designed and performed the experiments, analyzed and interpreted the data and wrote the manuscript.

Corresponding authors

Correspondence to Denis Vivien or Marina Rubio.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Ádám Dénes, Robert Harris 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 Immunofluorescent characterization of CAMs in young and aged mice cortex.

a) Representative immunofluorescence images of CD206+ CAMs (red), P2Y12R+ microglia (green) and collagen IV in the basal lamina of blood vessels (gray) in young male mice brain cortex. Scale bar: 100 µm. b-d) Representative immunofluorescence images of CD206+ CAMs (red), P2Y12R+ microglia (green), GFAP+ astrocytes (cyan), AQP4+ astrocytic endfeet (cyan) in young mice brain cortex. Scale bar: 100 µm. e) Representative immunofluorescence images of CD206+ CAMs (red), Iba1+ microglia (green) and collagen IV in the basal lamina of blood vessels (gray) in young and aged male mice brain cortex. Scale bar: 100 µm. f) Representative immunofluorescence images of CD206+ and Lyve1+ CAMs (red) and podocalyxin-labelled blood vessels (gray), in young or aged male mice brain cortex. Scale bar: 100 µm. g) Representative immunofluorescence images of Iba1+ microglia (green) and CD68+ macrophage/phagocytic microglia (red) in young or aged male mice brain cortex. Scale bar: 100 µm. h) Immunohistological quantification of Iba1+ microglia and Iba1+/CD68+ phagocytic microglia or macrophages in young or aged male mice brain cortex. n=6 mice/groupe. Two-sided Mann-Whitney U-test. Data are presented as mean values +/− SEM.

Source data

Extended Data Fig. 2 Flow cytometry dot-plots and gating strategies.

a) Representative flow cytometry dot-plots and gating strategy used for quantification of CAMs, microglia or macrophages and neutrophils from young and aged male mice brain. b) Representative flow cytometry dot-plots and gating strategy used for quantification of CD4 and CD8 T cells from young and aged mice brain. c) Representative flow cytometry dot-plots and gating strategy used for quantification of CD19+ B220+ B cells from young and aged mice brain. d) Representative flow cytometry dot-plots and gating strategy used for isolation of CD11b+ CD45+ CD206+ CAMs by fluorescence activated cell sorting (FACS) from young and aged mice brain to analyze the RNA expression profile.

Extended Data Fig. 3 Immunohistochemical analysis of microglia in young and aged human brain cortex.

a) Table representing the information about the gender, the age and the Broadmann area of interest of the subjects within the two groups "Young controls" and "Aged controls". b) Representative IHC staining of human brain cortex within grey matter area. Hematoxylin (Blue), Iba1 (DAB), HLA (green). Scale bar: 50 µm. c) Representative IHC staining of human brain cortex within grey matter area. Hematoxylin (Blue), Iba1 (DAB), CD74 (green) Scale bar: 50 µm. d) Quantification of Iba1+, Iba1+ HLA+ and Iba1+ CD74+ microglia in the human grey matter. n= 5 young controls/ 5 aged controls. Two-sided Mann-Whitney Utest. Data are presented as mean values +/− SEM.

Source data

Extended Data Fig. 4 CAMs depletion by intracerebroventricular injection of clodronate-liposomes.

a) Schematic representation of the experimental protocol for CAMs depletion and quantification. b) Representative flow cytometry dot-plots and gating strategy used for quantification of CAMs, microglia, activated microglia or macrophages and neutrophils from young and aged male mice brain treated with vehicle or clodronate-liposomes. c) Flow cytometry quantification of CAMs, microglia, activated microglia or macrophages and neutrophils from young and aged mice brain treated with vehicle or clodronate-liposomes. n=4 mice/group. Two-sided Mann-Whitney U test. d) Representative immunofluorescence images of CD206+ CAMs (red), P2Y12R+ or Iba1+ microglia (green) and collagen IV in the basal lamina of blood vessels (gray), in vehicle or clodronate-liposomes treated mice cortex. Scale bar: 100 µm. e) Immunohistological quantification of CD206+ CAMs and Iba1+ microglia in vehicle or clodronate-liposomes treated mice cortex. n=5, Two-sided Mann-Whitney U-test. Data are presented as mean values +/− SEM (c, e).

Source data

Extended Data Fig. 5 Immunohistochemical analysis of P-selectin expression and fibrinogen extravasation.

a) Representative immunofluorescence images of Psel+ blood vessels in mice brain cortex at 5 days after stroke in vehicle or clodronate-treated young male mice. Scale bar: 50 µm. b) Representative immunofluorescence images of Psel+ blood vessels in mice brain cortex at 5 days after stroke in vehicle or clodronate-treated aged mice. Scale bar: 50 µm. c) Immunohistological quantification of the number of Psel+ blood vessels in mice brain cortex at 5 days after stroke. n=5 mice/group. d) Immunohistological quantification of the Psel+ area in mice brain cortex at 5 days after stroke. n=5 mice/group. e) Immunofluorescence images of Psel+ (magenta) blood vessels (gray) and Iba1+ microglia (green) in mice brain cortex at 5 days after stroke in vehicle or clodronate-treated aged mice. Scale bar: 50 µm. f) Representative immunofluorescence images of fibrinogen extravasation in mice brain cortex 1 day after stroke in young and aged mice treated with PBS (Vehicle) or clodronate (CLO). Scale bar 100µm. g) Quantification of the fibrinogen extravasation area. n=6 vehicle, young / n=6 CLO, young / n=5 vehicle, aged / n=5 CLO, aged. Multiple Mann-Whitney U-test with two-stage step up method for false discovery rate (c, d, g). Data are presented as mean values +/− SEM (c, d, g).

Source data

Extended Data Fig. 6 CAMs and microglia characterization after stroke in young and aged mice.

a) Representative flow cytometry dot-plots and gating strategy used for quantification of CAMs, microglia, macrophages and neutrophils 2 days after stroke in vehicle or clodronate-treated young and aged male mice brain. b-c) Flow cytometry quantification of CAMs, microglia, macrophages and neutrophils 2 days after stroke in vehicle or clodronate-treated young and aged mice brain. n=7 mice/group. d) Representative immunofluorescence images of CD206+ CAMs and Iba1+ microglia in perilesional area at 5 days after stroke in vehicle or CLO treated young and aged mice. Scale bar: 100 µm. e) Immunohistological quantification of CD206+ CAMs 1 day (left) and 5 days (right) after stroke. n=5 mice/group. f) Representative immunofluorescence images of Iba1+/CD68+ phagocytic microglia in perilesional area 1 day after stroke in vehicle or clodronate-treated young or aged mice. Scale bar: 100 µm. g) Immunohistological quantification of Iba1+ microglia 1 day and 5 days after stroke. n=5 mice/group. h) Immunohistological quantification of Iba1+/CD68+ phagocytic microglia 1 day and 5 days after stroke. n=5 mice/group. Multiple Mann-Whitney U-test with two-stage step up method for false discovery rate (b, c, e, g, h). Data are presented as mean values +/− SEM (b, c, e, g, h).

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Levard, D., Seillier, C., Bellemain-Sagnard, M. et al. Central nervous system-associated macrophages modulate the immune response following stroke in aged mice. Nat Neurosci 27, 1721–1733 (2024). https://doi.org/10.1038/s41593-024-01695-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41593-024-01695-3

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