Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Identification of a protective microglial state mediated by miR-155 and interferon-γ signaling in a mouse model of Alzheimer’s disease

Abstract

Microglia play a critical role in brain homeostasis and disease progression. In neurodegenerative conditions, microglia acquire the neurodegenerative phenotype (MGnD), whose function is poorly understood. MicroRNA-155 (miR-155), enriched in immune cells, critically regulates MGnD. However, its role in Alzheimer’s disease (AD) pathogenesis remains unclear. Here, we report that microglial deletion of miR-155 induces a pre-MGnD activation state via interferon-γ (IFN-γ) signaling, and blocking IFN-γ signaling attenuates MGnD induction and microglial phagocytosis. Single-cell RNA-sequencing analysis of microglia from an AD mouse model identifies Stat1 and Clec2d as pre-MGnD markers. This phenotypic transition enhances amyloid plaque compaction, reduces dystrophic neurites, attenuates plaque-associated synaptic degradation and improves cognition. Our study demonstrates a miR-155-mediated regulatory mechanism of MGnD and the beneficial role of IFN-γ-responsive pre-MGnD in restricting neurodegenerative pathology and preserving cognitive function in an AD mouse model, highlighting miR-155 and IFN-γ as potential therapeutic targets for AD.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Targeting microglial miR-155 enhances MGnD signature in APP/PS1 mice.
Fig. 2: miR-155 ablation primes microglial transition to the pre-MGnD cluster through IFN-γ signaling.
Fig. 3: Targeting interferon-γ signaling abolishes the beneficial effect of miR-155 deletion in response to phagocytic stress.
Fig. 4: miR-155-deficient microglia restrict amyloid-β pathology.
Fig. 5: Deletion of microglial miR-155 enhanced synaptic functions in the brain milieu of APP/PS1 mice at 4 and 8 months of age.
Fig. 6: Microglia-specific miR-155 ablation rescues cognitive function in APP/PS1 mice.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available in this paper and Supplementary Information. The Smart-seq2 RNA-seq and scRNA-seq data that support the findings of this study have been deposited into Gene Expression Omnibus under SuperSeries GSE205569.

Code availability

Single-cell and bulk RNA-seq data were analyzed using customized code available at https://github.com/The-Butovsky-Lab/Yin-Herron-et-al.-miR-155-deletion-study/.

References

  1. Efthymiou, A. G. & Goate, A. M. Late-onset Alzheimer’s disease genetics implicates microglial pathways in disease risk. Mol. Neurodegener. 12, 43 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Lambert, J. C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 45, 1452–1458 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kunkle, B. W. et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat. Genet. 51, 414–430 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Colonna, M. & Butovsky, O. Microglia function in the central nervous system during health and neurodegeneration. Annu. Rev. Immunol. 35, 441–468 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Heneka, M. T. et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 14, 388–405 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Butovsky, O. et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat. Neurosci. 17, 131–143 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. 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 

  9. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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 

  11. Butovsky, O. & Weiner, H. L. Microglial signatures and their role in health and disease. Nat. Rev. Neurosci. 19, 622–635 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Yuan, P. et al. TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 90, 724–739 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cheng, Q. et al. TREM2-activating antibodies abrogate the negative pleiotropic effects of the Alzheimer’s disease variant Trem2R47H on murine myeloid cell function. J. Biol. Chem. 293, 12620–12633 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cignarella, F. et al. TREM2 activation on microglia promotes myelin debris clearance and remyelination in a model of multiple sclerosis. Acta Neuropathol. 140, 513–534 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ellwanger, D. C. et al. Prior activation state shapes the microglia response to antihuman TREM2 in a mouse model of Alzheimer’s disease. Proc. Natl Acad. Sci. USA 118, e2017742118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Fassler, M., Rappaport, M. S., Cuño, C. B. & George, J. Engagement of TREM2 by a novel monoclonal antibody induces activation of microglia and improves cognitive function in Alzheimer’s disease models. J. Neuroinflammation 18, 19 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Schlepckow, K. et al. Enhancing protective microglial activities with a dual function TREM 2 antibody to the stalk region. EMBO Mol. Med. 12, e11227 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. Clayton, K. et al. Plaque-associated microglia hyper-secrete extracellular vesicles and accelerate tau propagation in a humanized APP mouse model. Mol. Neurodegener. 16, 18 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Stevens, B. et al. The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Faraoni, I., Antonetti, F. R., Cardone, J. & Bonmassar, E. miR-155 gene: a typical multifunctional microRNA. Biochim. Biophys. Acta 1792, 497–505 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. O’Connell, R. M., Taganov, K. D., Boldin, M. P., Cheng, G. & Baltimore, D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc. Natl Acad. Sci. USA 104, 1604–1609 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Butovsky, O. et al. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann. Neurol. 77, 75–99 (2015).

    Article  PubMed  Google Scholar 

  27. Koval, E. D. et al. Method for widespread microRNA-155 inhibition prolongs survival in ALS-model mice. Hum. Mol. Genet 22, 4127–4135 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Readhead, B. et al. miR-155 regulation of behavior, neuropathology, and cortical transcriptomics in Alzheimer’s disease. Acta Neuropathol. 140, 295–315 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Aloi, M. S. et al. The pro-inflammatory microRNA miR-155 influences fibrillar β-amyloid1–42 catabolism by microglia. Glia 69, 1736–1748 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hu, R. et al. miR-155 promotes T follicular helper cell accumulation during chronic, low-grade inflammation. Immunity 41, 605–619 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. 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 

  32. Radde, R. et al. Aβ42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 7, 940–946 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. Roy, E. R. et al. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. J. Clin. Invest. 130, 1912–1930 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  36. Lai, J. J., Cruz, F. M. & Rock, K. L. Immune sensing of cell death through recognition of histone sequences by C-type lectin-receptor-2d causes inflammation and tissue injury. Immunity 52, 123–135 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Gracias, D. T. et al. The microRNA miR-155 controls CD8+ T cell responses by regulating interferon signaling. Nat. Immunol. 14, 593–602 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Li, H., Gade, P., Xiao, W. & Kalvakolanu, D. V. The interferon signaling network and transcription factor C/EBP-β. Cell. Mol. Immunol. 4, 407 (2007).

    CAS  PubMed  Google Scholar 

  39. He, M., Xu, Z., Ding, T., Kuang, D.-M. & Zheng, L. MicroRNA-155 regulates inflammatory cytokine production in tumor-associated macrophages via targeting C/EBPβ. Cell. Mol. Immunol. 6, 343–352 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Prencipe, G. et al. Neutralization of IFN-γ reverts clinical and laboratory features in a mouse model of macrophage activation syndrome. J. Allergy Clin. Immunol. 141, 1439–1449 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. 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 

  42. Serneels, L. et al. γ-Secretase heterogeneity in the Aph1 subunit: relevance for Alzheimer’s disease. Science 324, 639–642 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hammond, T. R. et al. Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity 50, 253–271 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Gerrits, E. et al. Distinct amyloid-β and tau-associated microglia profiles in Alzheimer’s disease. Acta Neuropathol. 141, 681–696 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mathys, H. et al. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 570, 332–337 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sierksma, A. et al. Deregulation of neuronal miRNAs induced by amyloid-β or TAU pathology. Mol. Neurodegener. 13, 54 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Culpan, D., Kehoe, P. G. & Love, S. Tumour necrosis factor-α (TNF-α) and miRNA expression in frontal and temporal neocortex in Alzheimer’s disease and the effect of TNF-α on miRNA expression in vitro. Int. J. Mol. Epidemiol. Genet. 2, 156–162 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Patrick, E. et al. Dissecting the role of non-coding RNAs in the accumulation of amyloid and tau neuropathologies in Alzheimer’s disease. Mol. Neurodegener. 12, 51 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Lau, P. et al. Alteration of the microRNA network during the progression of Alzheimer’s disease. EMBO Mol. Med. 5, 1613–1634 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Landgraf, P. et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401–1414 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Korotkov, A. et al. Increased expression of miR142 and miR-155 in glial and immune cells after traumatic brain injury may contribute to neuroinflammation via astrocyte activation. Brain Pathol. 30, 897–912 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Lee, S. H. et al. Identifying the initiating events of anti-Listeria responses using mice with conditional loss of IFN-γ receptor subunit 1 (IFNGR1). J. Immunol. 191, 4223–4234 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Garcia-Alonso, L., Holland, C. H., Ibrahim, M. M., Turei, D. & Saez-Rodriguez, J. Benchmark and integration of resources for the estimation of human transcription factor activities. Genome Res. 29, 1363–1375 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Germain, P. L., Lun, A., Garcia Meixide, C., Macnair, W. & Robinson, M. D. Doublet identification in single-cell sequencing data using scDblFinder. F1000Res 10, 979 (2021).

    Article  PubMed  Google Scholar 

  58. Zappia, L. & Oshlack, A. Clustering trees: a visualization for evaluating clusterings at multiple resolutions. Gigascience 7, giy083 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  59. 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 

  60. Subramanian, A. Obesity-instructed TREM2high macrophages identified by comparative analysis of diabetic mouse and human kidney at single cell resolution. Preprint at bioRxiv https://doi.org/10.1101/2021.05.30.446342 (2021).

  61. Gabriely, G. et al. Myeloid cell subsets that express latency-associated peptide promote cancer growth by modulating T cells. iScience 24, 103347 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mantri, M. et al. Spatiotemporal single-cell RNA sequencing of developing chicken hearts identifies interplay between cellular differentiation and morphogenesis. Nat. Commun. 12, 1771 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. 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 

  65. Griffiths-Jones, S., Grocock, R. J., van Dongen, S., Bateman, A. & Enright, A. J. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 34, D140–D144 (2006).

    Article  CAS  PubMed  Google Scholar 

  66. Wolf, A., Bauer, B., Abner, E. L., Ashkenazy-Frolinger, T. & Hartz, A. M. A comprehensive behavioral test battery to assess learning and memory in 129S6/Tg2576 mice. PLoS ONE 11, e0147733 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Lalonde, R. The neurobiological basis of spontaneous alternation. Neurosci. Biobehav. Rev. 26, 91–104 (2002).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. Krishnan (Ann Romney Center for Neurologic Diseases Flow Cytometry Core facility) and B. Tilton (Boston University Flow Cytometry Core facility) for FACS sorting; Y. Alekseyev, Boston University Single Cell Sequencing Core facility and Boston University Microarray and Sequencing Resource Core Facility for single-cell sequencing; the Single Cell Core at Harvard Medical School for performing the single-cell RNA-seq sample preparation; L. Ding and NeuroTechnology Studio at Brigham and Women’s Hospital for consultation on data acquisition and data analysis; G. Garden for sharing miR-155-flox mice; S. Garamszegi and Genomic Platform of Broad Institute of MIT and Harvard for Smart-seq2 RNA-seq; D. Holtzman for sharing the anti-Apoe and anti-Aβ antibodies. This work was supported by grants from the following organizations to the indicated authors. O.B. and T.I.: NIH-NIA (R01AG054672); O.B.: NIH-NIA (R01AG051812, R01AG075509, R01AG080992, R21AG076982 and R41AG073059), NIH-NINDS (R01NS088137, R21NS104609, R21NS101673), NIH-NEI (R01EY027921), NIH-NIGMS (R01GM132668), Cure Alzheimer’s Fund (ApoE Consortium) and BrightFocus Foundation (2020A016806); T.I. and S.I.: Cure Alzheimer’s Fund, T.I.: NIH-NIA (RF1AG054199, R01AG066429, R01AG072719 and R01AG067763); S.I.: NIH-NIA (R01AG079859), S.H.: NIH-NIA (F31AG071106) and NIH-NIGMS (T32GM008541). M.A.M: NIH-NEI (K12 EY016335, K08 EY030160) and Research to Prevent Blindness Career Development Award.

Author information

Authors and Affiliations

Authors

Contributions

O.B., T.I., S.I., Z.Y. and S.H. conceived the study. Z.Y. and S.H. did experimental design, performed experiments, analyzed data and wrote the manuscript. O.B., T.I. and S.I. provided funding, supervision of the study, data interpretation and manuscript editing. J.-C.D. assisted and supervised microglia isolation and behavioral experiments. S.H. performed behavioral tests. S.S., Y.C. and S.W.H. performed experiments. D.M., C.G. and K.K. performed analysis for Smart-seq2 RNA-seq, scRNA-seq and proteomic analysis. A.S. performed statistical analysis of scRNA-seq for comparison of proportions. C.M., W.B., M.A.M., K.M.P., J.-L.B., H.S. and A.D. developed and procured mouse models. M.J. performed mass spectrometry for proteomics. G.M. and A.K.A. performed the luciferase reporter assay.

Corresponding authors

Correspondence to Tsuneya Ikezu or Oleg Butovsky.

Ethics declarations

Competing interests

O.B. is an inventor of a patent for the use of miR-155 inhibitors for the treatment of neurodegenerative diseases. O.B. collaborated with GSK and Regulus Therapeutics, received research funding from Sanofi and GSK, has received honoraria for lectures from and has provided consultancy services to Camp4 and Ono Pharma USA. T.I. consults Takeda. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Evgenia Salta 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 miR-155 is specifically detected in Iba1+ cells in the brain of APP/PS1 mice.

a, Gating strategy for sorting Ly6CCD11b+Fcrls+ microglia. b, Confocal microscopy images of Iba1 or CD206 immunoreactivity and detection of miR-155 gene expression using miRNAscope in 8-month-old APP/PS1 mice, scale bar: 50 µm. c, Quantification of fluorescence intensity of miR-155 in Iba1+ cells, Iba1 cells, and CD206+ cells per ROI using one-way ANOVA with Tukey’s post hoc analysis, P < 0.0001 (n = 8 ROIs for Iba1+ group, n = 12 ROIs for Iba1 group, n = 5 ROIs for CD206+ group). Data were presented as mean values ± s.e.m.

Extended Data Fig. 2 Targeting microglial miR-155 causes subtle changes in APP/PS1 mice at 8 months old.

a, qPCR of miR-155 expression in microglia from 8-month-old mice showing the efficiency of tamoxifen treatment, P = 0.0003 using one-way ANOVA with Tukey’s post hoc test (n = 5 mice for WT and APP/PS1:miR-155 cKO group, n = 3 mice for miR-155 cKO group, n = 4 mice for APP/PS1 group). b, Heatmap of DEGs identified in microglia of APP/PS1:miR-155 cKO group at 8 months of age, compared to APP/PS1 group using DESeq2 analysis (Two-sided LRT, FDR-corrected P < 0.05, n = 5-6 per sex/group). c, PCA analysis showing no separation among all the groups at 8 months old. d, Venn diagram showing the difference of DEGs between 4- and 8-month-old groups. Data were presented as mean values ± s.e.m. See also Supplementary Table 1.

Extended Data Fig. 3 Targeting microglial miR-155 suppresses homeostatic signature in WT mice.

a, Experimental design. b, Number of DEGs comparing miR-155 cKO vs. WT at 4- and 8 months old (n = 5-6 mice per sex/group). Heat map of DEGs identified in miR-155 cKO microglia, compared to WT microglia for male (c) and female (d) groups using DESeq2 analysis, two-sided Wald test, FDR-corrected P < 0.05 (n = 5-6 mice per sex/group). e, Top-affected pathways based on the comparison of female miR-155 cKO microglia vs. WT microglia at 8 months of age using IPA (DEGs were identified with two-sided Wald test, FDR-corrected P < 0.05). See also Supplementary Table 1.

Extended Data Fig. 4 Male APP/PS1 mice show more robust changes after targeting microglial miR-155.

Venn diagram (a) and violin plot (b) showing the difference of up-regulated DEGs or down-regulated DEGs by comparing APP/PS1:miR-155 cKO vs. APP/PS1 (P < 0.05) between male and female samples at 4 months of age. c, Sex-specific top-15 upregulated and downregulated DEGs with the Log2 fold changes (FC) between APP/PS1 vs. WT group and APP/PS1:miR-155 cKO vs. WT group. DEGs were identified using DESeq2 analysis with FDR-corrected P < 0.05. d, Volcano plots using common DEGs of sex-specific APP/PS1:miR-155 cKO microglia vs. APP/PS1 microglia and non-plaque-associated microglia or plaque-associated microglia, compared to WT microglia (Two-sided LRT, P < 0.05). See also Supplementary Table 1.

Extended Data Fig. 5 miR-155 ablation induces interferon signaling in microglia.

a, GSEA analysis of interferon alpha response and interferon gamma response made using bulk RNA sequencing data from isolated microglia. b, DEG Heatmaps of genes involved in gene ontology terms for phagocytosis, antigen presentation and cellular response to IFN-γ derived from bulk RNAseq data (Two-sided LRT, FDR-corrected P < 0.05, n = 4-6 mice per sex/group). c, Transcription factor functional enrichment analysis in microglia comparing APP/PS1:miR-155 cKO vs. APP/PS1 mice at 4 months old (R_Dorothea, FDR-corrected P < 0.05). d, Workflow of generating miR-155 targetome. e, Volcano plots showing common genes (highlighted) between miR-155 targetome and DEGs from four different comparisons: male and female miR-155 cKO vs. WT at the age of 8 months, APP/PS1:miR-155 cKO vs. APP/PS1 at the age of 4 and 8 months (Two-sided Wald test, P < 0.05, n = 5-13 mice per group). f, Table of common miR-155 targeted genes among four different comparisons in e. See also Supplementary Table 1.

Extended Data Fig. 6 miR-155 ablation sensitizes microglial transition to the pre-MGnD cluster through interferon-γ signaling.

a, Gating strategy for sorting CD11b+CD45+ myeloid cells. b, Unbiased UMAP plots analysis of female APP/PS1 and APP/PS1:miR-155 cKO microglia from single cell sequencing. Initial clustering generated 17 sub-clusters (n = 2 mice per group, 15,429 cells). c, Cell cycle phase analysis for each cluster (clusters 15 and 16 removed). d, Percentage of cell cycle phase within each sub-cluster. e, Volcano plots of DEGs in the proliferating microglia clusters 8 and 9 comparing APP/PS1:miR-155 cKO microglia and APP/PS1 microglia (Two-sided Wald test, P < 0.05). f, Effect-size of differences in the proportion of microglia clusters between genotypes in female APP/PS1:miR-155 cKO mice determined by Poisson regression. The color bar indicates the P value, while the size of the points indicates the effect size. g, UMAP plots analysis of three major microglia clusters identified in male APP/PS1 and APP/PS1:miR-155 cKO microglia from scRNA sequencing (n = 3 mice per group, 43,667 cells). h, Bar charts showing relative percentage of each major microglia cluster in APP/PS1 and APP/PS1:miR-155 cKO mice. i, Violin plots showing the mean z-score of pre-MGnD markers in APP/PS1 and APP/PS1:miR-155 cKO male mice. P = 3.54507e-91 by Kruska-Wallis test, FDR corrected using Benjamini Hochberg. j, Top-affected pathways from IPA analysis of all three clusters based on the comparison of APP/PS1:miR-155 cKO microglia vs. APP/PS1 microglia at 4 months of age (DEGs were identified with two-sided Wilcox FDR-corrected P < 0.05). k, UMAP plots analysis of microglia clusters identified in male APP/PS1 and APP/PS1:miR-155 cKO microglia including subclusters of pre-MGnD. l, Dot plot showing top markers for four microglia clusters (M0, early IFN-responsive pre-MGnD, late IFN-responsive pre-MGnD and MGnD). m, Trajectory analysis showing the transition from M0 to MGnD via IFN-responsive pre-MGnD clusters. n, Dynamic plots showing the expression of selected genes in all four major microglia clusters. o, Effect-size of differences in the proportion of microglia clusters between genotypes in male APP/PS1:miR-155 cKO mice determined by Poisson regression. The color bar indicates the P value, while the size of the points indicates the effect size. Standard error bars were shown. See also Supplementary Table 2.

Extended Data Fig. 7 Validation of Pre-MGnD markers as Stat1 and Clec2d.

a, Confocal microscopy images of Stat1+ and Iba1+ cells in the brain of APP/PS1 and APP/PS1:miR-155 cKO mice at 4 months of age. Scale bar: 50 µm. b, Quantification of Stat1positive are in Iba1+ cells per ROI, P = 0.0001 by two-tailed Student’s unpaired t-test (n = 12 ROIs from 3 mice for APP/PS1 group, n = 18 ROIs from 6 mice for APP/PS1:miR-155 cKO group). c, Comparison of transcript per million (TPM) level of Clec2d between WT and APP/PS1 mice, P = 0.0458 by two-tailed Student’s unpaired t-test (n = 11 APP/PS1, n = 9 APP/PS1:miR-155 cKO). d, FACS plots showing four clusters in WT, 2- and 8-month-old APP/PS1 mice, which are Clec7aClec2d (M0), Clec7aClec2d+ (early pre-MGnD), Clec7a+Clec2d+ (late pre-MGnD), and Clec7a+Clec2d (MGnD). e, Quantification of percentage of four clusters mentioned in d from WT, 2- and 8-month-old APP/PS1 mice, P < 0.0001 for M0 and late pre-MGnD, P = 0.0011 (WT vs. 8-month-old APP/PS1) and P = 0.0084 (2-month-old APP/PS1 vs. 8-month-old APP/PS1) for early pre-MGnD, P = 0.0017 (WT vs. 8-month-old APP/PS1) and P = 0.0014 (2-month-old APP/PS1 vs. 8-month-old APP/PS1) for MGnD as determined by one-way ANOVA (n = 3 WT, n = 4 APP/PS1). f, qPCR Quantification of gene expression of P2ry12, Clec2d, and Clec7a in M0, early pre-MGnD, late pre-MGnD, and MGnD clusters, P < 0.0001 for P2ry12, P = 0.0046 (early pre-MGnD vs. MGnD) and P = 0.0103 (late pre-MGnD vs. MGnD) for Clec2d, P = 0.0136 (M0 vs. early pre-MGnD), P = 0.0060 (M0 vs. late pre-MGnD), and P = 0.0023 (M0 vs. MGnD) for Clec7a as determined by one-way ANOVA (n = 5 mice/group). Data were presented as mean values ± s.e.m.

Extended Data Fig. 8 miR-155 targets Stat1 and regulates Interferon signaling pathway.

a, Illustration of 3’UTR of Stat1 Gaussia luciferase assay co-transfected with miR-155 mimic or miR-155 antagomir into N9 microglial cells; Created with Biorender.com. b, Control plasmid activity in miR-155 mimic and miR-155 antagomir transfected cells. Fold changes were calculated as compared to control mimic or antagomir using two-tailed Student’s unpaired t-test (n = 3). c, Fold changes of luciferase activity after co-transfection of mutant STAT1 3’UTR and control mimic or miR-155 mimic to check the specific binding of miR-155 to Stat1, P = 0.0234 by two-way ANOVA with Holm-Sidak post hoc analysis (n = 3). d, Fold changes of luciferase activity after non-mutated Stat1 3’UTR co-transfected with the non-relevant miRNA mimic (miR-18), P = 0.0072 by one-way ANOVA with post hoc Tukey’s test (n = 3). Data are representative of 2 independent experiments and show biologically independent replicates (b-d). e, Venn diagram depicting overlap of Ifngr1 cKO downregulated genes and APP/PS1:miR-155 cKO upregulated genes. f, Heat map showing expression of genes which were downregulated by Ifngr1 cKO phagocytic microglia and upregulated in APP/PS1:miR-155 cKO microglia. Data were presented as mean values ± s.e.m. See also Supplementary Tables 1, 3.

Extended Data Fig. 9 Decreased 6E10+ amyloid plaque burden and enhanced phagocytosis function in APP/PS1:miR-155 cKO mice.

a, Representative images of 6E10 staining in APP/PS1 and APP/PS1:miR-155 cKO brains at the age of 4 months. Scale bar: 500 µm. b, 6E10 plaque counts normalized to area and percent area 6E10 positivity in 4-month-old mice. P values were determined using two-tailed Student’s unpaired t-test, P = 0.0084 and P = 0.0104, respectively (n = 10 APP/PS1, n = 9 APP/PS1:miR-155 cKO). c, 6E10 plaque counts normalized to area and percent area 6E10 positivity in 8-month-old mice (n = 14 APP/PS1, n = 16 APP/PS1:miR-155 cKO). P values were determined using two-tailed Student’s unpaired t-test. d, Representative images of Thioflavin-S in the cortex and hippocampus of APP/PS1 and APP/PS1:miR-155 cKO mice at 4 months old. Scale bar: 500 µm. P values were determined using two-tailed Student’s unpaired t-test. e, Thioflavin-S plaque counts normalized to area and percent area Thioflavin-S positivity in 4-month-old mice. P values were determined using two-tailed Student’s unpaired t-test (n = 10 APP/PS1, n = 9 APP/PS1:miR-155 cKO, P = 0.09). f, Thioflavin-S plaque counts normalized to area and percent area Thioflavin-S positivity in 8-month-old mice (n = 14 APP/PS1, n = 16 APP/PS1:miR-155 cKO). P values were determined using two-tailed Student’s unpaired t-test. g, Representative images of Thioflavin-S in the cortex and hippocampus of APP/PS1 and APP/PS1:miR-155 cKO mice at 2.5 months of age. Scale bar: 500 µm. h, Quantification of Thioflavin-S+ plaque area in the cortical region at 2.5 months of age using two-tailed Student’s unpaired t-test (n = 6 APP/PS1, n = 5 APP/PS1:miR-155 cKO). i, Representative images of HJ3.4B staining in APP/PS1 and APP/PS1:miR-155 cKO brains. Scale bar: 500 µm. j, Quantification of HJ3.4B+ plaque area normalized to cortical area at 2.5 months of age using two-tailed Student’s unpaired t-test (n = 6 APP/PS1, n = 5 APP/PS1:miR-155 cKO). k, Apoptotic neuron injection scheme. l, Mean fluorescence intensity (MFI) of phagocytic microglia sorted from WT and miR-155 cKO mice following apoptotic neuron injection, P = 0.0042 using two-tailed Student’s unpaired t-test (n = 7 WT, n = 14 miR-155 cKO). Data were combined from two independent experiments and normalized to control group (l). Data were presented as mean values ± s.e.m.

Extended Data Fig. 10 Proteomics analysis of whole-brain tissue showed enhanced synaptic functions in APP/PS1:miR-155 cKO mice at 4 and 8 months of age.

a, Heat maps of significantly changed proteins between male APP/PS1:miR-155 cKO and APP/PS1 whole brain tissue at 4 and 8 months of age (one-way ANOVA with Student’s t-test between APP/PS1 and APP/PS1:miR-155 cKO mice, P < 0.05, n = 3-4 male mice per group). See also Supplementary Table 4.

Supplementary information

Reporting Summary

Supplementary Table 1

RNA-seq data for Fig. 1 and related Extended Data figures.

Supplementary Table 2

scRNA-seq data for Fig. 2 and related Extended Data figures.

Supplementary Table 3

RNA-seq data for Fig. 3.

Supplementary Table 4

Proteomics data for Fig. 5.

Supplementary Table 5

Reagents and resources.

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

Yin, Z., Herron, S., Silveira, S. et al. Identification of a protective microglial state mediated by miR-155 and interferon-γ signaling in a mouse model of Alzheimer’s disease. Nat Neurosci 26, 1196–1207 (2023). https://doi.org/10.1038/s41593-023-01355-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-023-01355-y

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