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:

Cell-autonomous effects of APOE4 in restricting microglial response in brain homeostasis and Alzheimer’s disease

Abstract

Microglial involvement in Alzheimer’s disease (AD) pathology has emerged as a risk-determining pathogenic event. While apolipoprotein E (APOE) is known to modify AD risk, it remains unclear how microglial apoE impacts brain cognition and AD pathology. Here, using conditional mouse models expressing apoE isoforms in microglia and central nervous system-associated macrophages (CAMs), we demonstrate a cell-autonomous effect of apoE3-mediated microglial activation and function, which are negated by apoE4. Expression of apoE3 in microglia/CAMs improves cognitive function, increases microglia surrounding amyloid plaque and reduces amyloid pathology and associated toxicity, whereas apoE4 expression either compromises or has no effects on these outcomes by impairing lipid metabolism. Single-cell transcriptomic profiling reveals increased antigen presentation and interferon pathways upon apoE3 expression. In contrast, apoE4 expression downregulates complement and lysosomal pathways, and promotes stress-related responses. Moreover, in the presence of mouse endogenous apoE, microglial apoE4 exacerbates amyloid pathology. Finally, we observed a reduction in Lgals3-positive responsive microglia surrounding amyloid plaque and an increased accumulation of lipid droplets in APOE4 human brains and induced pluripotent stem cell-derived microglia. Our findings establish critical isoform-dependent effects of microglia/CAM-expressed apoE in brain function and the development of amyloid pathology, providing new insight into how apoE4 vastly increases AD risk.

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: ApoE3 expression in microglia/CAMs protects against the development of amyloid pathology and associated neurotoxicity, whereas apoE4 expression is associated with lipid accumulation.
Fig. 2: Transcriptomic analysis reveals apoE4-associated microglial dysfunction in activation and inflammatory responses to amyloid pathology.
Fig. 3: ApoE4 microglia display compromised microglial dynamics and responses to injury with reduced ability to support synaptic function compared to apoE3 microglia.
Fig. 4: Expression of apoE isoforms in microglia/CAMs differentially impacts the development of amyloid pathology and gliosis in the presence of mouse endogenous apoE.
Fig. 5: APOE4 reduces microglial responses to AD pathology in human brains and human microglia.

Similar content being viewed by others

Data availability

All datasets generated and/or analyzed in the current study are attached. The scRNA-seq data that support the findings of this study are deposited in the Gene Expression Omnibus repository under accession number https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE241553. Source data are provided with this paper.

References

  1. Ross, C. A. & Poirier, M. A. Protein aggregation and neurodegenerative disease. Nat. Med. 10, S10–S17 (2004).

    Article  PubMed  Google Scholar 

  2. Jucker, M. & Walker, L. C. Pathogenic protein seeding in Alzheimer disease and other neurodegenerative disorders. Ann. Neurol. 70, 532–540 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Guo, T. et al. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol. Neurodegener. 15, 40 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Leng, F. & Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat. Rev. Neurol. 17, 157–172 (2021).

    Article  PubMed  Google Scholar 

  5. Chen, Y. & Colonna, M. Microglia in Alzheimer’s disease at single-cell level. Are there common patterns in humans and mice? J. Exp. Med. https://doi.org/10.1084/jem.20202717 (2021).

  6. Magno, L., Bunney, T. D., Mead, E., Svensson, F. & Bictash, M. N. TREM2/PLCγ2 signalling in immune cells: function, structural insight, and potential therapeutic modulation. Mol. Neurodegener. 16, 22 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wang, C. et al. Selective removal of astrocytic APOE4 strongly protects against tau-mediated neurodegeneration and decreases synaptic phagocytosis by microglia. Neuron 109, 1657–1674 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kang, S. S. et al. Microglial translational profiling reveals a convergent APOE pathway from aging, amyloid, and tau. J. Exp. Med. 215, 2235–2245 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sala Frigerio, C. et al. The major risk factors for Alzheimer’s disease: age, sex, and genes modulate the microglia response to abeta plaques. Cell Rep. 27, 1293–1306 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Liu, C. C., Liu, C. C., Kanekiyo, T., Xu, H. & Bu, G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat. Rev. Neurol. 9, 106–118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yamazaki, Y., Zhao, N., Caulfield, T. R., Liu, C. C. & Bu, G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat. Rev. Neurol. 15, 501–518 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Raulin, A. C. et al. ApoE in Alzheimer’s disease: pathophysiology and therapeutic strategies. Mol. Neurodegener. 17, 72 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fernandez-Calle, R. et al. APOE in the bullseye of neurodegenerative diseases: impact of the APOE genotype in Alzheimer’s disease pathology and brain diseases. Mol. Neurodegener. 17, 62 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Spittau, B. Aging microglia-phenotypes, functions and implications for age-related neurodegenerative diseases. Front. Aging Neurosci. 9, 194 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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 

  17. Shi, Y. & Holtzman, D. M. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat. Rev. Immunol. 18, 759–772 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shi, Y. et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549, 523–527 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. 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 

  21. Ulrich, J. D. et al. ApoE facilitates the microglial response to amyloid plaque pathology. J. Exp. Med. 215, 1047–1058 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. Miyazaki, S., Miyazaki, T., Tashiro, F., Yamato, E. & Miyazaki, J. Development of a single-cassette system for spatiotemporal gene regulation in mice. Biochem. Biophys. Res Commun. 338, 1083–1088 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Liu, C. C. et al. ApoE4 accelerates early seeding of amyloid pathology. Neuron 96, 1024–1032 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Goldmann, T. et al. Origin, fate and dynamics of macrophages at central nervous system interfaces. Nat. Immunol. 17, 797–805 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Masuda, T. et al. Novel Hexb-based tools for studying microglia in the CNS. Nat. Immunol. 21, 802–815 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  29. Ajami, B., Bennett, J. L., Krieger, C., Tetzlaff, W. & Rossi, F. M. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat. Neurosci. 10, 1538–1543 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Lawson, L. J., Perry, V. H. & Gordon, S. Turnover of resident microglia in the normal adult mouse brain. Neuroscience 48, 405–415 (1992).

    Article  CAS  PubMed  Google Scholar 

  31. van Furth, R. & Cohn, Z. A. The origin and kinetics of mononuclear phagocytes. J. Exp. Med. 128, 415–435 (1968).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Fogg, D. K. et al. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science 311, 83–87 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Jankowsky, J. L. et al. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum. Mol. Genet. 13, 159–170 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Holtzman, D. M. et al. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. Proc. Natl Acad. Sci. USA 97, 2892–2897 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Pankiewicz, J. E. et al. Blocking the apoE/Aβ interaction ameliorates Aβ-related pathology in APOE ε2 and ε4 targeted replacement Alzheimer model mice. Acta Neuropathol. Commun. 2, 75 (2014).

    PubMed  PubMed Central  Google Scholar 

  36. Sharoar, M. G., Palko, S., Ge, Y., Saido, T. C. & Yan, R. Accumulation of saposin in dystrophic neurites is linked to impaired lysosomal functions in Alzheimer’s disease brains. Mol. Neurodegener. 16, 45 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Boucher, D. M., Vijithakumar, V. & Ouimet, M. Lipid droplets as regulators of metabolism and immunity. Immunometabolism 3, e210021 (2021).

    Article  Google Scholar 

  38. Marschallinger, J. et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat. Neurosci. 23, 194–208 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sienski, G. et al. APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aaz4564 (2021).

  40. Keane, L. et al. mTOR-dependent translation amplifies microglia priming in aging mice. J. Clin. Invest. https://doi.org/10.1172/JCI155208 (2021).

  41. Baik, S. H. et al. A breakdown in metabolic reprogramming causes microglia dysfunction in Alzheimer’s disease. Cell Metab. 30, 493–507 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Damani, M. R. et al. Age-related alterations in the dynamic behavior of microglia. Aging Cell 10, 263–276 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Davalos, D. et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 8, 752–758 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Nimmerjahn, A., Kirchhoff, F. & Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 308, 1314–1318 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Madry, C. & Attwell, D. Receptors, ion channels, and signaling mechanisms underlying microglial dynamics. J. Biol. Chem. 290, 12443–12450 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Schafer, D. P., Lehrman, E. K. & Stevens, B. The ‘quad-partite’ synapse: microglia–synapse interactions in the developing and mature CNS. Glia 61, 24–36 (2013).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  48. Olah, M. et al. Single-cell RNA sequencing of human microglia uncovers a subset associated with Alzheimer’s disease. Nat. Commun. 11, 6129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Neftel, C. et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell 178, 835–849 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lau, S. F., Cao, H., Fu, A. K. Y. & Ip, N. Y. Single-nucleus transcriptome analysis reveals dysregulation of angiogenic endothelial cells and neuroprotective glia in Alzheimer’s disease. Proc. Natl Acad. Sci. USA 117, 25800–25809 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Boza-Serrano, A. et al. Galectin-3, a novel endogenous TREM2 ligand, detrimentally regulates inflammatory response in Alzheimer’s disease. Acta Neuropathol. 138, 251–273 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Pantazis, C. B. et al. A reference human induced pluripotent stem cell line for large-scale collaborative studies. Cell Stem Cell 29, 1685–1702 (2022).

    Article  CAS  PubMed  Google Scholar 

  53. Querfurth, H. & Lee, H. K. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol. Neurodegener. 16, 44 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ding, X. et al. Loss of microglial SIRPα promotes synaptic pruning in preclinical models of neurodegeneration. Nat. Commun. 12, 2030 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hong, S., Dissing-Olesen, L. & Stevens, B. New insights on the role of microglia in synaptic pruning in health and disease. Curr. Opin. Neurobiol. 36, 128–134 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Nguyen, A. T. et al. APOE and TREM2 regulate amyloid-responsive microglia in Alzheimer’s disease. Acta Neuropathol. 140, 477–493 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Shi, Y. et al. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J. Exp. Med. 216, 2546–2561 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yin, Z. et al. APOE4 impairs the microglial response in Alzheimer’s disease by inducing TGFβ-mediated checkpoints. Nat. Immunol. https://doi.org/10.1038/s41590-023-01627-6 (2023).

  59. Condello, C., Yuan, P., Schain, A. & Grutzendler, J. Microglia constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques. Nat. Commun. 6, 6176 (2015).

    Article  CAS  PubMed  Google Scholar 

  60. Wang, Y. et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell 160, 1061–1071 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Parhizkar, S. et al. Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE. Nat. Neurosci. 22, 191–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Fitz, N. F. et al. Trem2 deficiency differentially affects phenotype and transcriptome of human APOE3 and APOE4 mice. Mol. Neurodegener. 15, 41 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lanfranco, M. F., Sepulveda, J., Kopetsky, G. & Rebeck, G. W. Expression and secretion of apoE isoforms in astrocytes and microglia during inflammation. Glia 69, 1478–1493 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Huynh, T. V. et al. Lack of hepatic apoE does not influence early Aβ deposition: observations from a new APOE knock-in model. Mol. Neurodegener. 14, 37 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Martens, Y. A. et al. ApoE cascade hypothesis in the pathogenesis of Alzheimer’s disease and related dementias. Neuron 110, 1304–1317 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Lian, H. et al. Astrocyte-microglia cross talk through complement activation modulates amyloid pathology in mouse models of Alzheimer’s disease. J. Neurosci. 36, 577–589 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Castellano, J. M. et al. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci. Transl. Med. 3, 89ra57 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Liu, C. C. et al. Neuronal heparan sulfates promote amyloid pathology by modulating brain amyloid-beta clearance and aggregation in Alzheimer’s disease. Sci. Transl. Med. 8, 332ra344 (2016).

    Article  Google Scholar 

  70. Chakrabarty, P. et al. IFN-gamma promotes complement expression and attenuates amyloid plaque deposition in amyloid beta precursor protein transgenic mice. J. Immunol. 184, 5333–5343 (2010).

    Article  CAS  PubMed  Google Scholar 

  71. Liu, C. C. et al. APOE3-Jacksonville (V236E) variant reduces self-aggregation and risk of dementia. Sci. Transl. Med. 13, eabc9375 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu, C. C. et al. Deficiency in LRP6-mediated Wnt signaling contributes to synaptic abnormalities and amyloid pathology in Alzheimer’s disease. Neuron 84, 63–77 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Liu, C. C. et al. Peripheral apoE4 enhances Alzheimer’s pathology and impairs cognition by compromising cerebrovascular function. Nat. Neurosci. 25, 1020–1033 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Drew, P. J. et al. Chronic optical access through a polished and reinforced thinned skull. Nat. Methods 7, 981–984 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kuchibhotla, K. V., Lattarulo, C. R., Hyman, B. T. & Bacskai, B. J. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science 323, 1211–1215 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Schafer, D. P. et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Subramanian, A. et al. Gene-set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Zhao, J. et al. APOE ε4/ε4 diminishes neurotrophic function of human iPSC-derived astrocytes. Hum. Mol. Genet. 26, 2690–2700 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Abud, E. M. et al. iPSC-derived human microglia-like cells to study neurological diseases. Neuron 94, 278–293 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zhao, J. et al. Apolipoprotein E regulates lipid metabolism and alpha-synuclein pathology in human iPSC-derived cerebral organoids. Acta Neuropathol. 142, 807–825 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  82. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at https://arxiv.org/abs/1802.03426 (2018).

  83. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by NIH grants U19AG069701 (to G.B., C.-C.L. and L.-J.W.), R01AG062110 (to C.-C.L. and J.D.F) and R37AG027924 (to G.B.), a grant from the Brightfocus Foundation (to C.-C.L.) and a grant from the Cure Alzheimer’s Fund (to G.B.). We are grateful to D. Dickson and M. Murry at Mayo Clinic Brain Bank for providing human AD brain slides. We thank M. C. Casey, V. Phillips and A. Librero at Mayo Clinic Histology Core for the immunohistochemical analyses.

Author information

Authors and Affiliations

Authors

Contributions

C.-C.L. and G.B. developed the research concept, designed the experiments, and wrote the paper. C.-C.L. and N.W. executed the experiments and analyzed the data. L.-J.W. provided knowledge support for the study. N.W., L.J., S.J., C.L.R., J.Z., S.V.D. and A.A. performed immunostaining, western blot analysis and quantification on mouse brain tissues. N.W. and F.Q. performed western blot analysis. Y.C. and B.Y.S.K. performed two-photon imaging and provided knowledge support for two-photon experiments. F.S. performed quantification for immunostaining. Y.A.M., Z.L. and C-C.L. performed scRNA-seq. Y.R., M.W., T.C.I. and B.Z. performed bioinformatics analysis and provided knowledge support for scRNA-seq analyses. A.-C.R. performed iPSC characterization. Y.I. and Y.Z. performed human iPSC-derived microglia-related experiments. W.Q. performed electrophysiological analyses. A.K. and J.D.F. performed behavioral analysis and provided technical support. C.L., Y.C., J.K. and C.-C.L. contributed to animal maintenance, genotyping and tissue isolation. All authors read and approved the final version of the paper.

Corresponding authors

Correspondence to Chia-Chen Liu or Guojun Bu.

Ethics declarations

Competing interests

During the initial phase of these studies, G.B. consulted for SciNeuro, AbbVie, E-Scape, Vida Ventures and Eisai, and was on the scientific advisory board for Kisbee Therapeutics. G.B. was an employee of SciNeuro during the revision phase and is currently employed at Hong Kong University of Science and Technology. C.-C.L. was employed with Biogen during the revision phase and is currently an employee of Biogen. Y.M. is currently an employee of SciNeuro. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: S. Houston, in collaboration with the rest of the Nature Immunology team.

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 Induction of apoE isoforms in microglia/CAMs in iE3/CreER/+ and iE4/CreER/+ mice.

a, b, Upon TAM administration, GFP fluorescence which represents apoE distribution was exclusively detected in the Iba1+ microglia (a, Scale bar, 40 µm) and TMEM119+ microglia (b, Scale bar, 20 µm) in the iE3/CreER/+ and iE4/CreER/+ mice but not in control mice. The percentage of GFP+ cells in Iba1+microglia or TMEM119+ microglia in experimental mice (iE3/CreER/+: Ctrl: n = 5, TAM-induced: n = 5; iE4/CreER/+: Ctrl: n = 5, TAM-induced: n = 5) was quantified. **, P < 0.0001. Two-tailed t-test. c, Brain tissues from TAM-induced mice were co-immunostained for GFP which represents apoE distribution (green), and astrocyte-specific (anti-GFAP; pink) or neuron-specific (anti-NeuN; red) markers. Experiments were repeated two times with similar results. Scale bar, 20 µm. d, The GFP fluorescence was colocalized with apoE signal (Red) detected by apoE antibody. Scale bar, 20 μm. e, The percentage of apoE+ cells in GFP+ microglia in iE3/CreER/+ (Ctrl: n = 5, TAM-induced: n = 8) and iE4/CreER/+ (Ctrl: n = 5, TAM-induced: n = 8) mice was quantified. **, P < 0.0001. f, ApoE levels in the cortex of the iE3/CreER/+ and iE4/CreER/+ mice at 6 months of age upon vehicle or TAM administration (iE3/CreER/+: Ctrl: n = 7, TAM-induced: n = 5; iE4/CreER/+: Ctrl: n = 7, TAM-induced: n = 5) were analyzed by ELISA. **, P < 0.0001. g, ApoE levels in the cortex of the iE3/CreER/+ (Ctrl: n = 8, TAM-induced: n = 8) and iE4/CreER/+ (Ctrl: n = 9, TAM-induced: n = 7) mice were examined by Western blotting and quantified. **, P < 0.0001. h, ApoE levels in the plasma of the iE3/CreER/+ (Ctrl: n = 5, TAM-induced: n = 5) and iE4/CreER/+ (Ctrl: n = 5, TAM-induced: n = 3) mice upon vehicle or TAM administration were analyzed by ELISA. Plasma from APOE3-TR and APOE4-TR mice (n = 4/group) was included as positive controls. **, P < 0.0001, one-way ANOVA. a, b, g-h, Data represent mean ± s.e.m. N.S., not significant. e-g, two-way ANOVA with a Tukey’s post-hoc test.

Source data

Extended Data Fig. 2 Differential effects of microglia/CAM-derived apoE isoforms on amyloid pathology.

a, Brain sections from 9-month-old APP/iE3/CreER/+ mice (Ctrl, n = 16; TAM-induced, n = 14) or APP/iE4/CreER/+ mice (Ctrl, n = 19; TAM-induced, n = 21) were immunostained with a pan-Aβ antibody. The Aβ plaque burden in the hippocampus was quantified (*, P = 0.048). Scale bar, 200 µm. b, c, Insoluble Aβ levels in the cortical tissues of APP/iE3/CreER/+ mice (Ctrl, n = 17; TAM, n = 15) and APP/iE4/CreER/+ mice (Ctrl, n = 16; TAM, n = 16) were examined by specific Aβ ELISA. (Aβ40:*, P = 0.045; Aβ42:*, P = 0.043) d, TBS- and TBSX-soluble Aβ levels in the cortex of the APP/iE3/CreER/+ mice (Ctrl, n = 17; TAM-induced, n = 16) or APP/iE4/CreER/+ mice (Ctrl, n = 16; TAM-induced, n = 16) were examined. *, P = 0.010. e, TBS soluble, detergent-soluble (TBSX fraction) and insoluble (GDN fraction) apoE levels in the cortex of the APP/iE3/CreER/+ (Ctrl, n = 17; TAM-induced, n = 16) or APP/iE4/CreER/+ (Ctrl, n = 16; TAM-induced, n = 16) mice were examined. **, P < 0.0001. f, Thioflavin S-positive plaques in the cortex of APP/iE3/CreER/+ mice (Ctrl, n = 10; TAM, n = 10) and APP/iE4/CreER/+ mice (Ctrl, n = 13; TAM, n = 17) were shown and quantified. Scale bar, 50 µm. In f-h, Images from APP/PS1 mice bred to apoE3-targeted replacement (TR) mice (APP/ApoE3) or apoE4-TR mice (APP/ApoE4) were included for a visualization of plaque morphology. g, Representative images of Aβ staining in the cortex of 9-month-old APP/iE3/CreER/+ or APP/iE4/CreER/+ mice (mouse Apoe-/- background) are shown. Staining was performed on more than 10 animals/group with similar results. Scale bar, 200 µm. h, Representative images of plaque compactness examined by Thio S staining in APP/iE3/CreER/+ mice (Ctrl, n = 11; TAM, n = 10) and APP/iE4/CreER/+ mice (Ctrl, n = 12; TAM, n = 16) with vehicle treatment (Ctrl) or TAM administration. Scale bars, 10 μm. The shape compactness of amyloid plaque in the brain of experimental mice was quantified. *, P = 0.048. a-f, h. Data represent mean ± s.e.m. N.S., not significant. Two-tailed, t-test.

Source data

Extended Data Fig. 3 Expression of apoE4 compromises microglial barrier and phagocytic functions.

a, b, Brain sections from APP/iE3/CreER/+ (Ctrl, n = 18; TAM-induced, n = 16) or APP/iE4/CreER/+ mice (Ctrl, n = 19; TAM-induced, n = 21) at 9 months of age were immunostained with Iba1 antibody. The immunoreactivity of Iba1-positive microglia in cortex and hippocampus was quantified. Scale bar, 100 µm. c, d, TAM-induced APP/iE3/CreER/+ mice (n = 3) and APP/iE4/CreER/+ male mice (n = 4) at 9 months of age were subjected to 2-photon imaging. c, Representative images of GFP-positive microglia (green) and methoxy-X04-stained amyloid plaques (blue) monitored by two-photon in vivo imaging. Plaque associated microglia (number of microglia per 100 mm2; 1-3 fields/mouse) were quantified. Scale bar, 20 µm. *, P = 0.018. d, Representative images of microglial phagocytosis in APP/iE3/CreER/+ (n = 3) and APP/iE4/CreER/+ mice (n = 4) examined by two-photon imaging. The colocalization (pink) of microglia and Aβ are pseudo colored to illustrate the phagocytosed Aβ in microglia which was quantified. Scale bar, 50 µm. *, P = 0.032. a-d. Data represent mean ± s.e.m. N.S., not significant. Two-tailed, t-test.

Source data

Extended Data Fig. 4 Differential effects of microglia/CAM-derived apoE isoforms on astrogliosis.

a, b, Brain sections from the APP/iE3/CreER/+ mice (Ctrl, n = 16; TAM-induced, n = 16) or APP/iE4/CreER/+ mice (Ctrl, n = 19; TAM-induced, n = 22) at 9 months of age were immunostained with GFAP antibody for astrogliosis. The immunoreactivities of GFAP-positive astrocyte in cortex and hippocampus were quantified. Scale bar, 100 µm. iE3:*, P = 0.028; iE4:*, P = 0.016. c, The correlations between the levels of GFAP-positive astrocytes and amyloid plaque in the APP/iE3/CreER/+ mice and APP/iE4/CreER/+ mice. Correlation coefficient (r) and P-value were acquired by Pearson’s correlation analysis. d, The levels of APP, PSD-95, (iE3: Ctrl, n = 9; TAM, n = 8; iE4: Ctrl, n = 9; TAM, n = 8), GFAP (iE3: Ctrl, n = 9; TAM, n = 8; iE4: Ctrl, n = 9; TAM, n = 7), synaptophysin (Syp) and aquaporin 4 (Aqp4) (iE3: Ctrl, n = 7; TAM, n = 7; iE4: Ctrl, n = 7; TAM, n = 7) in the cortex of the APP/iE3/CreER/+ mice or APP/iE4/CreER/+ mice were examined by Western blotting and quantified. *, P = 0.015 (GFAP); *, P = 0.013 (Aqp4). a, b, d. Data represent mean ± s.e.m. N.S., not significant. Two-tailed, t-test.

Source data

Extended Data Fig. 5 Single cell transcriptomic analysis of mice with apoE3 or apoE4 expression in microglia/CAMs.

Brain cortical tissues from the APP/iE3/CreER/+ mice (Ctrl, n = 6; TAM-induced, n = 6) or APP/iE4/CreER/+ mice (Ctrl, n = 6; TAM-induced, n = 6) at 9 months of age were subjected to single cell RNA-sequencing (scRNA-seq). a, Uniform manifold approximation and projection (UMAP) plot showing the clusters of single cell events captured in scRNA-seq analysis. MG, microglia; EC, endothelial cell; SMC, smooth muscle cell; PC, pericyte; MФ, microphage; CP, choroid plexus. b, Split dot plot depicting marker genes for each cell population in the APP/iE3/CreER/+ mice and APP/iE4/CreER/+ mice. The expression level (color intensity) and the percentage of cells in a cluster expressing a given gene (size of the dot) are reflected in circles. c, Feature plots of canonical markers defining major cell types identified from scRNA-seq analysis. d, The relative proportion of each cell cluster identified in the APP/iE3/CreER/+ mice and APP/iE4/CreER/+ mice with or without TAM administration. e, Proportions of cell types identified from the APP/iE3/CreER/+ mice and APP/iE4/CreER/+ mice with or without TAM administration. MG, microglia; AS, astrocyte; OL, oligodendrocyte; VC, vascular cell; MФ, microphage; CP, choroid plexus.

Source data

Extended Data Fig. 6 Single cell transcriptomic profiling reveals microglial apoE isoform-dependent pathways responding to amyloid pathology.

a, Feature plots showing the expression of selected markers in various microglial subtypes. Legend shows a color gradient of normalized expression. b, UMAP revealed an increased ARM4 (Cluster 9) cell population in mice with apoE3 expression in microglia. c, Percentage of cells from the APP/iE3/CreER/+ and APP/iE4/CreER/+ groups (Ctrl, n = 6; TAM-induced, n = 6) for each microglial cluster identified. Data represent mean ± s.e.m. **, P < 0.0001; N.S., not significant, two-tailed, t-test. The fold changes of cell numbers for each microglial cluster in TAM-induced mice compared with control mice (set as 1) are shown. d, Gene ontology (GO) enrichment analysis for genes in the Cluster 7 (ARM3) of the APP/iE3/CreER/+ mice. e, Gene ontology (GO) enrichment analysis for genes in the Cluster 7 (ARM3) of the APP/iE4/CreER/+ mice.

Source data

Extended Data Fig. 7 eIF2 pathway, oxidative stress and components of complement system are differentially modulated by apoE3 or apoE4 expression in microglia/CAMs.

a, Feature plots of Cluster 7 (ARM3) and Cluster 9 (ARM4) microglial populations. b, c, Heatmap and Violin plots for selected genes in cluster 7 (ARM3) identified from scRNA-seq of the APP/iE4/CreER/+ mice. d, e, Heatmap and Violin plots for selected genes in cluster 9 (ARM4) identified from scRNA-seq of the APP/iE4/CreER/+ mice. f, Venn diagrams indicating up-regulated and down-regulated DEGs in microglia cluster 7 (ARM3) between the APP/iE3/CreER/+ mice and APP/iE4/CreER/+ mice. g, h, Heatmap and Violin plots for selected genes in cluster 9 (ARM4) identified from scRNA-seq of the APP/iE3/CreER/+ mice and APP/iE4/CreER/+ mice. i, Gene ontology (GO) enrichment analysis for genes in the Cluster 9 (ARM4) of the APP/iE3/CreER/+ mice or APP/iE4/CreER/+ mice.

Extended Data Fig. 8 ApoE isoforms differentially impact transcriptional signatures for ARM1 and ARM2 in amyloid pathogenesis.

a, b, Volcano plots illustrating up- or down-regulated DEGs in activated microglia Cluster 3 (ARM1) or Cluster 4 (ARM2) from the APP/iE3/CreER/+ mice (a) or APP/iE4/CreER/+ mice (b) identified from scRNA-seq analysis. Genes significant at the P value ≤ 0.05 and fold change ≥ 1.2 are denoted red in color for apoE3 mice, and blue in color for apoE4 mice. c, Violin plots showing the mean and variance differences for selected DEGs from ARM1 or ARM2 identified from scRNA-seq analysis in the experimental mice. d, Venn diagrams indicating up-regulated and down-regulated DEGs in microglia cluster 3 (ARM1) between the APP/iE3/CreER/+ mice and APP/iE4/CreER/+ mice. e, Pseudotime analyses of scRNA-seq data showing the transition and trajectory of microglia from homeostatic status to activated signature.

Extended Data Fig. 9 The effects of apoE expression in microglia/CAMs on anxiety-related behaviors and synaptic transmission.

a, The general locomotor activity, and anxiety-like behaviors in the iE3/CreER/+ mice (Ctrl, n = 19; TAM-induced, n = 18) or iE4/CreER/+ mice (Ctrl, n = 19; TAM-induced, n = 20) at 6 months of age were assessed by open field tests. The total distance traveled, average speed, time spent mobile, and the center vs. total distance travelled are shown. b, The anxiety-like behaviors in the iE3/CreER/+ (Ctrl, n = 19; TAM-induced, n = 18) or iE4/CreER/+ mice (Ctrl, n = 19; TAM-induced, n = 20) were assessed by elevated plus maze test. Total distance traveled, average speed, total time spent mobile, and ratio of open vs. closed arms are shown. In a, b, Data represent mean ± s.e.m. N.S., not significant. Two-tailed, t-test. c, d, Input-output curve in Schaeffer collaterals-CA1 network showing fEPSPs in CA1 stratum radiatum in response to increased stimulus in the iE3/CreER/+ mice (c; Ctrl, n = 12; TAM, n = 10 brain slices from 5-6 mice/group) or iE4/CreER/+ mice (d; Ctrl, n = 16; TAM, n = 12 brain slices brain slices from 5-6 mice/group). The linear regression of fEPSP slope is shown. e, f, Paired-pulse facilitation (PPF) achieved with increased inter-pulse intervals in the iE3/CreER/+ mice (e; Ctrl, n = 10; TAM, n = 8 brain slices from 5-6 mice/group) or iE4/CreER/+ mice (f; Ctrl, n = 11; TAM, n = 7 brain slices from 5-6 mice/group) are shown. *, P = 0.044; 0.011; 0.015; 0.004; 0.0027. Wilcoxon Rank-sum tests.

Source data

Extended Data Fig. 10 Analyses on behaviors, synaptic plasticity, and microglial responses to focal injury in mice expressing apoE3 or apoE4 in microglia in the absence of Cx3cr1.

a, b, The associative memory of iE3/CreER/ER mice (Ctrl, n = 13; TAM, n = 14) or iE4/CreER/ER mice (Ctrl, n = 15; TAM, n = 14) at 6 months of age was assessed by contextual and cued fear conditioning tests. **, P = 0.007. c, The general locomotor activity, and anxiety-related behaviors in the iE3/CreER/ER mice (Ctrl, n = 13; TAM, n = 14) or iE4/CreER/ER mice (Ctrl, n = 15; TAM, n = 14) at 6 months of age were examined by open field tests. The total distance traveled, average speed, and center: distance are shown. d, The anxiety-like behaviors in the iE3/CreER/ER mice (Ctrl, n = 13; TAM, n = 14) or iE4/CreER/ER mice (Ctrl, n = 15; TAM, n = 13) were examined by elevated plus maze test. Total distance traveled, average speed, and open: closed arm are shown. Data represent mean ± SEM. N.S., not significant, Student’s t-test. e, f, The time courses of normalized initial slope measurements of fEPSP in hippocampal CA1 neurons in brain slices from the iE3/CreER/ER mice (Ctrl, n = 12; TAM, n = 13 brain slices from 5-6 mice/group) or iE4/CreER/ER mice (Ctrl, n = 7; TAM, n = 9 brain slices from 4-5 mice/group). Averages of the last 5 min of fEPSP recording were quantified. **, P = 0.001. g, Microglial processes responded and extended toward the ablation (15–20 µm in diameter). h, The kinetics of microglial responses over 30 min; microglial process movement towards a laser-induced focal lesion (circle) in the iE3/CreER/ER mice or iE4/CreER/ER mice (7–9 microglia from 4 mice/group). a-f. Data represent mean ± s.e.m. N.S., not significant. Two-tailed, t-test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2.

Reporting Summary

Supplementary Data 1

Statistical source data for Supplementary Fig. 1.

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 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. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots or gels.

Source Data Extended Data Fig. 4d_1

Unprocessed western blots or gels.

Source Data Extended Data Fig. 4d_2

Unprocessed western blots or gels.

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

Liu, CC., Wang, N., Chen, Y. et al. Cell-autonomous effects of APOE4 in restricting microglial response in brain homeostasis and Alzheimer’s disease. Nat Immunol 24, 1854–1866 (2023). https://doi.org/10.1038/s41590-023-01640-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01640-9

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