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.

  • Resource
  • Published:

Human early-onset dementia caused by DAP12 deficiency reveals a unique signature of dysregulated microglia

A Publisher Correction to this article was published on 20 February 2023

This article has been updated

Abstract

The TREM2–DAP12 receptor complex sustains microglia functions. Heterozygous hypofunctional TREM2 variants impair microglia, accelerating late-onset Alzheimer’s disease. Homozygous inactivating variants of TREM2 or TYROBP-encoding DAP12 cause Nasu–Hakola disease (NHD), an early-onset dementia characterized by cerebral atrophy, myelin loss and gliosis. Mechanisms underpinning NHD are unknown. Here, single-nucleus RNA-sequencing analysis of brain specimens from DAP12-deficient NHD individuals revealed a unique microglia signature indicating heightened RUNX1, STAT3 and transforming growth factor-β signaling pathways that mediate repair responses to injuries. This profile correlated with a wound healing signature in astrocytes and impaired myelination in oligodendrocytes, while pericyte profiles indicated vascular abnormalities. Conversely, single-nuclei signatures in mice lacking DAP12 signaling reflected very mild microglial defects that did not recapitulate NHD. We envision that DAP12 signaling in microglia attenuates wound healing pathways that, if left unchecked, interfere with microglial physiological functions, causing pathology in human. The identification of a dysregulated NHD microglia signature sparks potential therapeutic strategies aimed at resetting microglia signaling pathways.

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: NHD microglia show a unique signature.
Fig. 2: The NHD microglia signature in part overlaps with the IL-10-primed macrophage signature.
Fig. 3: The NHD pericyte signature indicates vascular abnormalities.
Fig. 4: NHD astrocyte signatures reflect a wound healing response.
Fig. 5: The microglia profile of aged KΔ75 mice is distinct from that of NHD.
Fig. 6: KΔ75 mice exhibit mild oligodendrocyte and astrocyte defects.

Similar content being viewed by others

Data availability

snRNA-seq differential expression gene lists with statistics are available in Supplementary Tables 3 and 6. All requests for human snRNA-seq data are promptly reviewed by the Brain Research Institute at Niigata University and Mihara Memorial Hospital (Isesaki, Japan) to verify if the request is subject to any intellectual property or confidentiality obligations. Human-related data not included in the paper were generated as part of clinical trials and may be subject to patient confidentiality. Any data and materials that can be shared will be released via a material transfer agreement. Raw and processed mouse and human snRNA-seq data have been deposited to the Gene Expression Omnibus (GEO) database under accession number GSE190015. Processed data, UMAP coordinates and annotations for the human NHD snRNA-seq have also been made freely available to download and visualize at the University of California Santa Cruz Cell Browser (https://nhd-brain.cells.ucsc.edu). scRNA-seq and microarray data on macrophage polarization were downloaded from GEO under accession numbers GSE199378 and GSE61298, respectively. Please contact the corresponding authors for additional information. Source data for Figs. 2, 4, 5 and 6 are provided with the paper.

Code availability

All software used in this study is published and cited either in the main text or Methods. No custom code was used for data processing or analysis. All the code used in the analysis for NHD snRNA-seq data can be found at https://github.com/yingyue-zhou/NHD_snRNA.

Change history

References

  1. Ulland, T. K. & Colonna, M. TREM2—a key player in microglial biology and Alzheimer disease. Nat. Rev. Neurol. 14, 667–675 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Li, R.-Y. et al. TREM2 in the pathogenesis of AD: a lipid metabolism regulator and potential metabolic therapeutic target. Mol. Neurodegener. 17, 40 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 

  4. Song, W. M. et al. Humanized TREM2 mice reveal microglia-intrinsic and -extrinsic effects of R47H polymorphism. J. Exp. Med. 215, 745–760 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Chen, Y. & Colonna, M. Microglia in Alzheimer’s disease at single-cell level. Are there common patterns in humans and mice? J. Exp. Med. 218, e20202717 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Paloneva, J. et al. Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am. J. Hum. Genet. 71, 656–662 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chouery, E. et al. Mutations in TREM2 lead to pure early-onset dementia without bone cysts. Hum. Mutat. 29, E194–E204 (2008).

    Article  PubMed  Google Scholar 

  9. Paloneva, J. et al. CNS manifestations of Nasu–Hakola disease: a frontal dementia with bone cysts. Neurology 56, 1552–1558 (2001).

    Article  CAS  PubMed  Google Scholar 

  10. Sasaki, A. et al. Variable expression of microglial DAP12 and TREM2 genes in Nasu–Hakola disease. Neurogenetics 16, 265–276 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Kaifu, T. et al. Osteopetrosis and thalamic hypomyelinosis with synaptic degeneration in DAP12-deficient mice. J. Clin. Invest. 111, 323–332 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nataf, S. et al. Brain and bone damage in KARAP/DAP12 loss-of-function mice correlate with alterations in microglia and osteoclast lineages. Am. J. Pathol. 166, 275–286 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Poliani, P. L. et al. TREM2 sustains microglial expansion during aging and response to demyelination. J. Clin. Invest. 125, 2161–2170 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Roumier, A. et al. Impaired synaptic function in the microglial KARAP/DAP12-deficient mouse. J. Neurosci. 24, 11421–11428 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tomasello, E. et al. Combined natural killer cell and dendritic cell functional deficiency in KARAP/DAP12 loss-of-function mutant mice. Immunity 13, 355–364 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Kondo, T. et al. Heterogeneity of presenile dementia with bone cysts (Nasu–Hakola disease): three genetic forms. Neurology 59, 1105–1107 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Zusso, M. et al. Regulation of postnatal forebrain amoeboid microglial cell proliferation and development by the transcription factor Runx1. J. Neurosci. 32, 11285–11298 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Griffiths, E. K. & Penninger, J. M. ADAP-ting TCR signaling to integrins. Sci. STKE 2002, re3 (2002).

    Article  PubMed  Google Scholar 

  19. Zheng, B. et al. VEGFR2 promotes metastasis and PD-L2 expression of human osteosarcoma cells by activating the STAT3 and RhoA–ROCK–LIMK2 pathways. Front. Oncol. 10, 543562 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Simoncic, P. D., Lee-Loy, A., Barber, D. L., Tremblay, M. L. & McGlade, C. J. The T cell protein tyrosine phosphatase is a negative regulator of janus family kinases 1 and 3. Curr. Biol. 12, 446–453 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Matak, P. et al. Activated macrophages induce hepcidin expression in HuH7 hepatoma cells. Haematologica 94, 773–780 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Katoh, M. & Katoh, M. STAT3-induced WNT5A signaling loop in embryonic stem cells, adult normal tissues, chronic persistent inflammation, rheumatoid arthritis and cancer (review). Int. J. Mol. Med. 19, 273–278 (2007).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Srinivasan, K. et al. Alzheimer’s patient microglia exhibit enhanced aging and unique transcriptional activation. Cell Rep. 31, 107843 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. McKay, E. C. et al. Peri-infarct upregulation of the oxytocin receptor in vascular dementia. J. Neuropathol. Exp. Neurol. 78, 436–452 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Absinta, M. et al. A lymphocyte–microglia–astrocyte axis in chronic active multiple sclerosis. Nature 597, 709–714 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Martinez, F. O., Sica, A., Mantovani, A. & Locati, M. Macrophage activation and polarization. Front. Biosci. 13, 453–461 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. Ohradanova-Repic, A. et al. Extracellular purine metabolism is the switchboard of immunosuppressive macrophages and a novel target to treat diseases with macrophage imbalances. Front. Immunol. 9, 852 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Noda, M. et al. FGF-2 released from degenerating neurons exerts microglial-induced neuroprotection via FGFR3–ERK signaling pathway. J. Neuroinflammation 11, 76 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Garcia, F. J. et al. Single-cell dissection of the human brain vasculature. Nature 603, 893–899 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kelley, C., D’Amore, P., Hechtman, H. B. & Shepro, D. Vasoactive hormones and cAMP affect pericyte contraction and stress fibres in vitro. J. Muscle Res. Cell Motil. 9, 184–194 (1988).

    Article  CAS  PubMed  Google Scholar 

  32. Gerthoffer, W. T., Solway, J. & Camoretti-Mercado, B. Emerging targets for novel therapy of asthma. Curr. Opin. Pharmacol. 13, 324–330 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Parthasarathy, S. & Fehr, A. R. PARP14: a key ADP-ribosylating protein in host–virus interactions? PLoS Pathog. 18, e1010535 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Eddleston, M. et al. Astrocytes are the primary source of tissue factor in the murine central nervous system. A role for astrocytes in cerebral hemostasis. J. Clin. Invest. 92, 349–358 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wiese, S., Karus, M. & Faissner, A. Astrocytes as a source for extracellular matrix molecules and cytokines. Front. Pharmacol. 3, 120 (2012).

  36. Matthies, A. M., Low, Q. E. H., Lingen, M. W. & DiPietro, L. A. Neuropilin-1 participates in wound angiogenesis. Am. J. Pathol. 160, 289–296 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hara, T. et al. Interactions between cancer cells and immune cells drive transitions to mesenchymal-like states in glioblastoma. Cancer Cell 39, 779–792 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Moulson, A. J., Squair, J. W., Franklin, R. J. M., Tetzlaff, W. & Assinck, P. Diversity of reactive astrogliosis in CNS pathology: heterogeneity or plasticity? Front. Cell. Neurosci. 15, 703810 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Satoh, J.-I. et al. Immunohistochemical characterization of microglia in Nasu–Hakola disease brains. Neuropathology 31, 363–375 (2011).

    Article  PubMed  Google Scholar 

  40. Erwig, M. S. et al. Anillin facilitates septin assembly to prevent pathological outfoldings of central nervous system myelin. eLife 8, e43888 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Jäkel, S. et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566, 543–547 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Ximerakis, M. et al. Single-cell transcriptomic profiling of the aging mouse brain. Nat. Neurosci. 22, 1696–1708 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. 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 

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

    Article  CAS  PubMed  Google Scholar 

  45. 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 

  46. Scherneck, S. et al. in Sensory and Metabolic Control of Energy Balance (eds Meyerhof, W., Beisiegel, U. & Joost, H. -G.) 57–68 (Springer, 2010).

  47. Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yaffe, Y. et al. The myelin proteolipid plasmolipin forms oligomers and induces liquid-ordered membranes in the Golgi complex. J. Cell Sci. 128, 2293–2302 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Yu, T. & Lieberman, A. P. Npc1 acting in neurons and glia is essential for the formation and maintenance of CNS myelin. PLoS Genet. 9, e1003462 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kenigsbuch, M. et al. A shared disease-associated oligodendrocyte signature among multiple CNS pathologies. Nat. Neurosci. 25, 876–886 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Peng, Q., Long, C. L., Malhotra, S. & Humphrey, M. B. A physical interaction between the adaptor proteins DOK3 and DAP12 is required to inhibit lipopolysaccharide signaling in macrophages.Sci. Signal. 6, ra72 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Turnbull, I. R. & Colonna, M. Activating and inhibitory functions of DAP12. Nat. Rev. Immunol. 7, 155–161 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Wang, S. et al. TREM2 drives microglia response to amyloid-β via SYK-dependent and -independent pathways. Cell 185, 4153–4169 (2022).

    Article  CAS  PubMed  Google Scholar 

  54. Lapenna, A., De Palma, M. & Lewis, C. E. Perivascular macrophages in health and disease. Nat. Rev. Immunol. 18, 689–702 (2018).

    Article  CAS  PubMed  Google Scholar 

  55. Haure-Mirande, J.-V. et al. Deficiency of TYROBP, an adapter protein for TREM2 and CR3 receptors, is neuroprotective in a mouse model of early Alzheimer’s pathology. Acta Neuropathol. 134, 769–788 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Fonseka, C. Y. et al. Mixed-effects association of single cells identifies an expanded effector CD4+ T cell subset in rheumatoid arthritis. Sci. Transl. Med. 10, eaaq0305 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  57. 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  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Andreatta, M. & Carmona, S. J. UCell: robust and scalable single-cell gene signature scoring. Comput. Struct. Biotechnol. J. 19, 3796–3798 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).

    Article  CAS  PubMed  Google Scholar 

  63. Zenkova, D. et al. phantasus: visual and interactive gene expression analysis. Bioconductor https://doi.org/10.18129/B9.bioc.phantasus (2022).

  64. Takeuchi, R. et al. Heterogeneity of cerebral TDP-43 pathology in sporadic amyotrophic lateral sclerosis: evidence for clinico-pathologic subtypes. Acta Neuropathologica Commun. 4, 61 (2016).

    Article  Google Scholar 

Download references

Acknowledgements

We thank E. Vivier (Aix Marseille Université) and W. M. Yokoyama (Washington University in St. Louis) for providing the KΔ75 mice, R. Cho (Cell Signaling Technology) for kindly providing STAT3 and pSTAT3 antibodies, L. L. Lanier (University of California at San Francisco) for providing DAP12 antibody, V. Peng and M. Terekhova for advice on bioinformatic analyses, S. Brioschi for great scientific discussions and M. Tano for technical help. M. Tada was supported by the Japan Society for the Promotion of Science (JSPS, KAKENHI, 22H02995). A.K. was supported by the Strategic Research Program for Brain Sciences from the Japan Agency for Medical Research and Development (AMED, JP21wm0425019) and the Collaborative Research Project of the Brain Research Institute, Niigata University, Japan. M. Takao was supported by AMED (JP21wm0425019). M.C. was supported by the NIH (RF1 AG051485, R21 AG077244, P01 AG078106 and RF1 AG059082) Cure Alzheimer’s Fund, Fred and Ginger Haberle Charitable Fund at East Texas Communities Foundation and Centene.

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. and M.C. designed the study and interpreted the results. Y.Z. performed computational analyses. M. Tada performed IHC and IF staining in human sections. Y.Z. and Z.C. performed validation experiments. Y.Z. and A.S. prepared single-nuclei suspension samples. K.R.M. helped with single-cell technology. S.G. bred all mice. M. Tada, M. Takao and A.K. provided human postmortem brain samples and clinical advice. P.S.A. and M.N.A. provided guidance for computational analyses. Y.Z. and M.C. wrote the manuscript with feedback from all authors.

Corresponding author

Correspondence to Marco Colonna.

Ethics declarations

Competing interests

M.C. is a member of the Vigil Neuro scientific advisory board (SAB), is a consultant for Cell Signaling Technology, has received research grants from Vigil Neuro during the conduct of the study and has a patent to TREM2 pending. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Mariko Bennett and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with 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 Cluster characterization of NHD and control samples.

a, UMAP plots of NHD and control samples before (left) and after (right) batch correction by Harmony. n = 66,324 total nuclei; n = 11 healthy individuals and n = 3 individuals with NHD. b, Bar graphs of total nuclei number, median of number of genes and median of number of UMIs of each sample sequenced. c, Heatmap showing cell type markers. d, Nuclei frequency of all cell types in each sample.

Extended Data Fig. 2 Characterization of NHD microglia subclusters.

a, Pathways enriched in genes upregulated in NHD microglia compared to controls, using GSEA analysis (left, q value by FDR adjustment) or Metascape analysis (right, P value by hypergeometric distribution). b, PCA plot of NHD and control microglia demarcated by sex. c, Average expression of top DEGs [log2(Fold Change) > 0.5, FDR-adjusted P < 0.05, two-part, generalized linear model] upregulated in NHD microglia plotted against age at death. Trend line calculated for control samples only.

Extended Data Fig. 3 NHD microglia signature overlaps with IL-10-induced macrophage signature.

a, UMAP plot of human monocyte-derived macrophages stimulated with cocktails of LPS + IFNγ, IL-4 + IL-13 or IL-10 from GSE199378, designated by treatment. b, Venn diagram showing little overlap among signatures of polarized macrophages. c-e, Violin plot showing gene set scores (by UCell) of the designated macrophage polarization signatures (top 100 genes upregulated under each condition) in the macrophage polarization dataset; c, LPS + IFNγ polarization, n = 8,232 nuclei; d, IL-4 + IL-13 polarization, n = 6,741 nuclei; e, IL-10 polarization, n = 6,890 nuclei. Box center lines, bounds of box, and whiskers indicate median, first and third quartiles, and minima and maxima within 1.5X IQR, respectively. P value by two-sided Wilcoxon Rank Sum test. f-h, GSEA plots showing enrichment of NHD microglia signature in signatures of polarized macrophages stimulated by LPS and IFNγ (f), IL-4 (g) and IL-10 (h) from GSE6129828. P values by permutation. NES, normalized enrichment score. i, Histogram of phospho-STAT3 (pSTAT3) staining of bone marrow derived macrophages (BMDMs) from WT and DAP12 KΔ75 mice, stimulated with 0 ng/ml or 20 ng/ml IL-10 for 15 min after starvation overnight. Data are representative of two independent experiments. j, Quantification of pSTAT3 mean fluorescence intensity (MFI) in i. P value by two-way ANOVA. Data are presented as mean ± s.e.m. n = 3 independent cell culture wells per genotype per experiment; n = two independent experiments. k, Histogram of total STAT3 staining in BMDM from WT and DAP12 KΔ75 mice, treated as in i. Data are representative of two independent experiments. l, Quantification of STAT3 mean fluorescence intensity in k. P value by two-way ANOVA. Data are presented as mean ± s.e.m. n = 3 independent cell culture wells per genotype per experiment; n = two independent experiments. m, Gating strategy for BMDMs from WT and KΔ75 mice. Numbers indicate the percentage of cells within the gate.

Extended Data Fig. 4 The NHD endothelial cell signature shows altered function.

a, Volcano plot depicting genes differentially expressed (log2(Fold Change) > 0.25, FDR-adjusted P < 0.05, two-part, generalized linear model) in NHD versus control endothelial cells. b, Box plots showing average individual expression levels for selected DEGs in endothelial cells. Box center lines, bounds of box, and whiskers indicate median, first and third quartiles, and minima and maxima within 1.5X IQR, respectively. P values by two-sided Wilcoxon Rank Sum test. n = 11 healthy individuals and n = 3 individuals with NHD. c, Heatmap showing average gene expression of top DEGs in endothelial cells per sample.

Extended Data Fig. 5 Comparison of NHD astrocyte and oligodendrocyte signatures with those in other neurodegenerative diseases.

a, b, Venn diagram revealing overlapping genes commonly upregulated in NHD astrocytes and two human AD datasets (a), and in NHD astrocytes and AIMS (astrocytes inflamed in MS) (b). c, Dot plot showing marker genes for each oligodendrocyte subcluster. d, Volcano plot depicting genes differentially expressed in NHD and control oligodendrocytes (log2(fold change) > 0.25, FDR-adjusted P < 0.05, two-part, generalized linear model). e, Box plots showing average of individual levels of expression for selected genes upregulated in NHD oligodendrocytes. Box center lines, bounds of box, and whiskers indicate median, first and third quartiles, and minima and maxima within 1.5X IQR, respectively. P values by two-sided Wilcoxon rank-sum test. n = 11 healthy individuals and n = 3 individuals with NHD. f, Heatmap showing average gene expression of top DEGs (log2(fold change) > 0.25, FDR-adjusted P < 0.05, two-part, generalized linear model) in oligodendrocytes from each sample. g, Pathways enriched in genes upregulated in NHD oligodendrocytes. P values calculated based on the cumulative hypergeometric distribution. h, i, Venn diagram revealing genes differentially expressed in NHD oligodendrocytes as well as in two human MS datasets. h, upregulated genes. i, downregulated genes.

Extended Data Fig. 6 NHD neurons present signatures of diminished activity.

a, UMAP plot of neuron sub-clustering. n = 36,336 total neuronal nuclei; n = 11 healthy individuals and n = 3 individuals with NHD. Ex, excitatory neurons; In, inhibitory neurons. b, Nuclei frequency of neuronal subclusters in each sample. c, Number of upregulated (up) and downregulated (down) genes differentially expressed in (log2(fold change) > 0.5, FDR-adjusted P < 0.05, two-part, generalized linear model) NHD and controls in each neuronal subcluster. d,e, Pathways enriched in genes downregulated in NHD in excitatory neurons (d) or inhibitory neurons (e). P values calculated based on the cumulative hypergeometric distribution. f, Violin plots showing percentage of reads that map to ribosomal genes split by disease condition. Differences between NHD and controls in all subclusters are significant by two-sided Wilcoxon Rank Sum test. Each dot represents the mean ± s.d. A full list of P values and number of nuclei is given in Supplementary Table 2. g, Heatmap showing average expression of genes in synaptic vesicle endocytosis pathway per condition per cluster.

Extended Data Fig. 7 Cluster characterization of 2-year-old KΔ75 and WT cortical samples.

a, Bar graphs of total nuclei number, median of number of genes and median of number of UMIs of each sample sequenced. b, UMAP plot of 2-year-old KΔ75 and WT cortical samples grouped by clusters, which were manually assigned to each cell type. n = 60,851 total nuclei; n = 4 animals per genotype. c, UMAP plots showing expression of cluster markers. d, Nuclei distribution of all samples in each cluster. e, UMAP plot of all nuclei from KΔ75 and WT samples. n = 26,492 WT nuclei and n = 34,359 KΔ75 nuclei; n = 4 animals per genotype.

Extended Data Fig. 8 KΔ75 microglia sub-clustering in detail.

a, UMAP plot showing expression of Tyrobp in myeloid clusters. b, Venn diagram highlighting similarities between IFN-R and the interferon responsive signature from ref. 45. Overlapping genes are shown in the box. c, Scatter plot depicting differential cell type abundance calculated by MASC. Data are represented as the MASC OR of a nucleus being in that cluster for WT versus KΔ75 (with 95% CI), against the -log(P value) of the association. Center of bar corresponds to OR. Red labeled cluster was significant with FDR-adjusted P value < 0.05 using Benjamini-Hochberg correction. A full list of P values and number of nuclei is provided in Supplementary Table 5. d, Donut plots showing nuclei contribution from each sample to each myeloid subcluster. Numbers represent the percentage of nuclei from each sample within the designated subcluster. e, Volcano plot showing DEGs (log2(fold change) > 0.5, adjusted P < 0.05, two-sided Wilcoxon Rank Sum test, Bonferroni correction) within the HM cluster from KΔ75 and WT mice. f, Heatmap showing average gene expression of top DEGs from e in HM cluster. g, Pathways enriched in genes downregulated in KΔ75 HM vs. WT HM. q values calculated based on Benjamini-Hochberg.

Extended Data Fig. 9 KΔ75 oligodendrocyte and astrocyte sub-clustering in detail.

a, UMAP plots showing cluster marker expression in each oligodendrocyte sub-cluster. b, Violin plots showing expression of reactive oligodendrocyte genes in each sub-cluster. c, Violin plots showing expression of reactive oligodendrocyte genes in the ROL clusters from KΔ75 and WT mice. d, Violin plots showing the DOL signature score (by UCell) in each oligodendrocyte sub-cluster. DOL signature was extracted from ref. 50. Box center lines, bounds of box, and whiskers indicate median, first and third quartiles, and minima and maxima within 1.5X IQR, respectively. P values by two-sided Wilcoxon Rank Sum test. n = 8,398 MFOL, 1,643 OPC, 754 MOL, 74 COP and 51 ROL nuclei. e, Violin plots showing DOL signature score (by UCell) in ROL from KΔ75 and WT mice. f, Representative IF images of MBP (red) in corpus callosum of 2-year-old WT and KΔ75 mice. Scale bar, 30 μm. g, Quantification of MBP intensity in f. P value by two-tailed unpaired t test. Bars at mean. n = 4 WT and n = 3 KΔ75 mice. h, UMAP plot showing expression of Vim and Gfap in astrocyte subcultures. i, Violin plots showing expression of reactive astrocyte genes in each sub-cluster. j, Pathways enriched in astrocyte cluster 8 marker genes. q-values calculated based on Benjamini-Hochberg. k, UMAP plot showing expression of Crym in astrocyte subclusters.

Extended Data Fig. 10 Transcriptional profiles of KΔ75 and WT neurons are similar.

a, UMAP plot of neuron sub-clustering. n = 40,657 total neuronal nuclei; n = 4 animals per genotype. b, UMAP plot showing expression of Slc17a7 and Gad1. c, UMAP plot of neuron subclustering grouped by excitatory neurons (Ex_neuron) and inhibitory neurons (In_neuron). d, Number of upregulated (up) and downregulated (down) DEGs (log2(fold change) > 0.5, adjusted P < 0.05, non-parametric two-sided Wilcoxon rank sum test, Bonferroni correction) in excitatory and inhibitory neurons in KΔ75 versus WT mice. e, Model of NHD pathogenesis. A genetic defect in DAP12 or TREM2 leads to imbalance between TREM2/DAP12 signaling pathway and pathways driven by STAT3, RUNX1 and TGFβ in microglia. This imbalance results in demyelination through unknown mechanisms, which may include dysregulated phagocytosis and secretion of cytokines. Accumulation of myelin and cellular debris leads to progressive and extensive tissue damage that includes astrocytosis, oligodendrocyte malfunction and dysfunctional vasculature, which further activates microglia. In addition, perivascular macrophages may directly alter vascular function, leading to its dysregulation. Created with BioRender.com.

Supplementary information

Reporting Summary

Supplementary Table 1

Supplementary Tables 1–6.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source 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

Zhou, Y., Tada, M., Cai, Z. et al. Human early-onset dementia caused by DAP12 deficiency reveals a unique signature of dysregulated microglia. Nat Immunol 24, 545–557 (2023). https://doi.org/10.1038/s41590-022-01403-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01403-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