Abstract
Human microglia play a pivotal role in neurological diseases, but we still have an incomplete understanding of microglial heterogeneity, which limits the development of targeted therapies directly modulating their state or function. Here, we use single-cell RNA sequencing to profile 215,680 live human microglia from 74 donors across diverse neurological diseases and CNS regions. We observe a central divide between oxidative and heterocyclic metabolism and identify microglial subsets associated with antigen presentation, motility and proliferation. Specific subsets are enriched in susceptibility genes for neurodegenerative diseases or the disease-associated microglial signature. We validate subtypes in situ with an RNAscope–immunofluorescence pipeline and high-dimensional MERFISH. We also leverage our dataset as a classification resource, finding that induced pluripotent stem cell model systems capture substantial in vivo heterogeneity. Finally, we identify and validate compounds that recapitulate certain subtypes in vitro, including camptothecin, which downregulates the signature of disease-enriched subtypes and upregulates a signature previously associated with Alzheimer’s disease.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
27,99 € / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
209,00 € per year
only 17,42 € per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout








Similar content being viewed by others
Data availability
Raw scRNA-seq data (fastq files) generated from CD45+ cells isolated from autopsy samples were deposited to the Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE204702. Bulk RNA-seq data from compound-treated HMC3 cells were deposited to the GEO under accession number GSE202556. Bulk proteomic data from compound-treated HMC3 cells were deposited to ProteomeXChange (http://www.proteomexchange.org/) under accession number PXD033844. Data repurposed for label transfer was retrieved from the GEO under accession numbers GSE133432, GSE178317 and GSE103224.
Code availability
Code used to perform preprocessing, clustering, cluster validation and label transfer of scRNA-seq data in the current study is available publicly at https://github.com/jtuddenham/single-cell-microglia-v2/. The CellProfiler pipeline used to analyze joint immunofluorescence–RNAscope data is available as Supplementary Information (Supplementary Table 5), and in the aforementioned GitHub repository. Code for visualization, analysis of bulk RNA-seq/proteomic data and downstream analysis of CellProfiler outputs is available from the corresponding author upon request.
References
Ginhoux, F., Lim, S., Hoeffel, G., Low, D. & Huber, T. Origin and differentiation of microglia. Front. Cell. Neurosci. 7, 45 (2013).
Li, Q. & Barres, B. A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 18, 225–242 (2018).
Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).
Olah, M. et al. Single cell RNA sequencing of human microglia uncovers a subset associated with Alzheimer’s disease. Nat. Commun. 11, 6129 (2020).
Butovsky, O. & Weiner, H. L. Microglial signatures and their role in health and disease. Nat. Rev. Neurosci. 19, 622–635 (2018).
Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290 (2017).
Ayata, P. et al. Epigenetic regulation of brain region-specific microglia clearance activity. Nat. Neurosci. 21, 1049–1060 (2018).
Gerrits, E. et al. Distinct amyloid-β and tau-associated microglia profiles in Alzheimer’s disease. Acta Neuropathol. 141, 681–696 (2021).
Masuda, T., Sankowski, R., Staszewski, O. & Prinz, M. Microglia heterogeneity in the single-cell era. Cell Rep. 30, 1271–1281 (2020).
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).
Colonna, M. & Brioschi, S. Neuroinflammation and neurodegeneration in human brain at single-cell resolution. Nat. Rev. Immunol. 20, 81–82 (2020).
Dumas, A. A., Borst, K. & Prinz, M. Current tools to interrogate microglial biology. Neuron 109, 2805–2819 (2021).
Masuda, T. et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature 566, 388–392 (2019).
Kracht, L. et al. Human fetal microglia acquire homeostatic immune-sensing properties early in development. Science https://doi.org/10.1126/science.aba5906 (2020).
Young, A. M. H. et al. A map of transcriptional heterogeneity and regulatory variation in human microglia. Nat. Genet. 53, 861–868 (2021).
Thrupp, N. et al. Single-nucleus RNA-seq is not suitable for detection of microglial activation genes in humans. Cell Rep. 32, 108189 (2020).
Bakken, T. E. et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types. PLoS ONE 13, e0209648 (2018).
Marsh, S. E. et al. Dissection of artifactual and confounding glial signatures by single-cell sequencing of mouse and human brain. Nat. Neurosci. 25, 306–316 (2022).
Mattei, D. et al. Enzymatic dissociation induces transcriptional and proteotype bias in brain cell populations. Int. J. Mol. Sci. 21, 7944 (2020).
Subramanian, A. et al. A next generation connectivity map: L1000 platform and the first 1,000,000 profiles. Cell 171, 1437–1452 (2017).
Lamb, J. et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006).
Olah, M. et al. A transcriptomic atlas of aged human microglia. Nat. Commun. 9, 539 (2018).
Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).
Gerrits, E., Heng, Y., Boddeke, E. W. G. M. & Eggen, B. J. L. Transcriptional profiling of microglia; current state of the art and future perspectives. Glia 68, 740–755 (2020).
Haage, V. et al. Comprehensive gene expression meta-analysis identifies signature genes that distinguish microglia from peripheral monocytes/macrophages in health and glioma. Acta Neuropathol. Commun. 7, 20 (2019).
Jurga, A. M., Paleczna, M. & Kuter, K. Z. Overview of general and discriminating markers of differential microglia phenotypes. Front. Cell. Neurosci. 14, 198 (2020).
Masuda, T. et al. Novel Hexb-based tools for studying microglia in the CNS. Nat. Immunol. 21, 802–815 (2020).
Van Hove, H. et al. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat. Neurosci. 22, 1021–1035 (2019).
Kierdorf, K., Masuda, T., Jordão, M. J. C. & Prinz, M. Macrophages at CNS interfaces: ontogeny and function in health and disease. Nat. Rev. Neurosci. 20, 547–562 (2019).
Lee, J. et al. QUAKING regulates microexon alternative splicing of the Rho GTPase pathway and controls microglia homeostasis. Cell Rep. 33, 108560 (2020).
Ren, J. et al. Qki is an essential regulator of microglial phagocytosis in demyelination. J. Exp. Med. 218, e20190348 (2021).
Nguyen, A. T. et al. APOE and TREM2 regulate amyloid-responsive microglia in Alzheimer’s disease. Acta Neuropathol. 140, 477–493 (2020).
Zhou, Y. et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and -independent cellular responses in Alzheimer’s disease. Nat. Med. 26, 131–142 (2020).
Tasic, B. et al. Adult mouse cortical cell taxonomy revealed by single cell transcriptomics. Nat. Neurosci. 19, 335–346 (2016).
Tasic, B. et al. Shared and distinct transcriptomic cell types across neocortical areas. Nature 563, 72–78 (2018).
Ashburner, M. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).
The Gene Ontology Consortium. The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 47, D330–D338 (2019).
Alexa, A. & Rahnenfuhrer, J. topGO: enrichment analysis for Gene Ontology. Bioconductor version: release 3.13. https://doi.org/10.18129/B9.bioc.topGO (2021).
Sayols, S. rrvgo: a Bioconductor package to reduce and visualize Gene Ontology terms. https://doi.org/10.18129/B9.bioc.rrvgo (2020).
Jassal, B. et al. The reactome pathway knowledgebase. Nucleic Acids Res. 48, D498–D503 (2020).
Wu, G. & Haw, R. Functional interaction network construction and analysis for disease discovery. Methods Mol. Biol. 1558, 235–253 (2017).
Yu, G., Wang, L. -G., Han, Y. & He, Q. -Y. clusterProfiler: an R Package for comparing biological themes among gene clusters. OMICS. 16, 284–287 (2012).
Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).
Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).
Qiu, X. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).
Mathys, H. et al. Temporal tracking of microglia activation in neurodegeneration at single-cell resolution. Cell Rep. 21, 366–380 (2017).
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).
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).
Krasemann, S. et al. The TREM2–APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47, 566–581 (2017).
Marschallinger, J. et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat. Neurosci. 23, 194–208 (2020).
International Multiple Sclerosis Genetics Consortium. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science 365, eaav7188 (2019).
Buniello, A. et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 47, D1005–D1012 (2019).
Sekar, A. et al. Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183 (2016).
David, A. B., Julie, A. S., Zoe, A. & Robert, S. W. Overview and findings from the Religious Orders Study. Curr. Alzheimer Res. 9, 628–645 (2012).
David, A. B. et al. Overview and findings from the Rush Memory and Aging Project. Curr. Alzheimer Res. 9, 646–663 (2012).
Patrick, E. et al. A cortical immune network map identifies distinct microglial transcriptional programs associated with β-amyloid and Tau pathologies. Transl. Psychiatry 11, 50 (2021).
Wang, F. et al. RNAscope. J. Mol. Diagn. 14, 22–29 (2012).
Carpenter, A. E. et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 7, R100 (2006).
McQuin, C. et al. CellProfiler 3.0: next-generation image processing for biology. PLoS Biol. 16, e2005970 (2018).
Kamentsky, L. et al. Improved structure, function and compatibility for CellProfiler: modular high-throughput image analysis software. Bioinformatics 27, 1179–1180 (2011).
Stirling, D. R. et al. CellProfiler 4: improvements in speed, utility and usability. BMC Bioinf. 22, 433 (2021).
Stence, N., Waite, M. & Dailey, M. E. Dynamics of microglial activation: a confocal time-lapse analysis in hippocampal slices. Glia 33, 256–266 (2001).
Green, G. S. et al. Cellular communities reveal trajectories of brain ageing and Alzheimer’s disease. Nature 633, 634–645 (2024).
Yuan, J. et al. Single-cell transcriptome analysis of lineage diversity in high-grade glioma. Genome Med. 10, 57 (2018).
Hambardzumyan, D., Gutmann, D. H. & Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 19, 20–27 (2016).
Hasselmann, J. et al. Development of a chimeric model to study and manipulate human microglia in vivo. Neuron 103, 1016–1033 (2019).
Claes, C. et al. Plaque-associated human microglia accumulate lipid droplets in a chimeric model of Alzheimer’s disease. Mol. Neurodegener. 16, 50 (2021).
Dräger, N. M. et al. A CRISPRi/a platform in human iPSC-derived microglia uncovers regulators of disease states. Nat. Neurosci. 5, 1149–1162 (2022).
Chen, T. & Guestrin, C. XGBoost: a scalable tree boosting system. in Proceedings of the 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining 785–794 https://doi.org/10.1145/2939672.2939785 (Association for Computing Machinery, 2016).
Szulzewsky, F. et al. Glioma-associated microglia/macrophages display an expression profile different from M1 and M2 polarization and highly express Gpnmb and Spp1. PLoS ONE 10, e0116644 (2015).
Ryan, K. J. et al. A human microglia-like cellular model for assessing the effects of neurodegenerative disease gene variants. Sci. Transl. Med. 9, eaai7635 (2017).
Dello Russo, C. et al. The human microglial HMC3 cell line: where do we stand? A systematic literature review. J. Neuroinflammation 15, 259 (2018).
Boutej, H. et al. Diverging mRNA and protein networks in activated microglia reveal SRSF3 suppresses translation of highly upregulated innate immune transcripts. Cell Rep. 21, 3220–3233 (2017).
Miedema, A. et al. Brain macrophages acquire distinct transcriptomes in multiple sclerosis lesions and normal appearing white matter. Acta Neuropathol. Commun. 10, 8 (2022).
Absinta, M. et al. A lymphocyte–microglia–astrocyte axis in chronic active multiple sclerosis. Nature 597, 709–714 (2021).
Patel, T. et al. Transcriptional landscape of human microglia implicates age, sex, and APOE ‐related immunometabolic pathway perturbations. Aging Cell 21, e13606 (2022).
Alsema, A. M. et al. Profiling microglia from Alzheimer’s disease donors and non-demented elderly in acute human postmortem cortical tissue. Front. Mol. Neurosci. 13, 134 (2020).
Li, Y. et al. Decoding the temporal and regional specification of microglia in the developing human brain. Cell Stem Cell 29, 620–634 (2022).
Kumar, P. et al. Single-cell transcriptomics and surface epitope detection in human brain epileptic lesions identifies pro-inflammatory signaling. Nat. Neurosci. 25, 956–966 (2022).
Amin, A. G. et al. Targeting the mTOR pathway using novel ATP‑competitive inhibitors, Torin1, Torin2 and XL388, in the treatment of glioblastoma. Int. J. Oncol. 59, 83 (2021).
Johnson, S. C. et al. mTOR inhibition alleviates mitochondrial disease in a mouse model of Leigh syndrome. Science 342, 1524–1528 (2013).
He, D. et al. Camptothecin regulates microglia polarization and exerts neuroprotective effects via activating AKT/Nrf2/HO-1 and inhibiting NF-κB pathways in vivo and in vitro. Front. Immunol. 12, 619761 (2021).
Stark, A. et al. Narciclasine exerts anti-inflammatory actions by blocking leukocyte-endothelial cell interactions and down-regulation of the endothelial TNF receptor 1. FASEB J. 33, 8771–8781 (2019).
Vonsattel, J. P. G. et al. Twenty-first century brain banking: practical prerequisites and lessons from the past: the experience of New York Brain Bank, Taub Institute, Columbia University. Cell Tissue Bank. 9, 247–258 (2008).
R Core Team. R: a language and environment for statistical computing. R Foundation for Statistical Computing (2021).
RStudio Team. RStudio: integrated development for R. RStudio, PBC (2020).
Osorio, D. & Cai, J. J. Systematic determination of the mitochondrial proportion in human and mice tissues for single-cell RNA-sequencing data quality control. Bioinformatics 37, 963–967 (2021).
Klein, H. -U. demuxmix: demultiplexing oligonucleotide-barcoded single-cell RNA sequencing data with regression mixture models. Bioinformatics 39, btad481 (2023).
McGinnis, C. S. et al. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices. Nat. Methods 16, 619–626 (2019).
Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).
Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).
Haghverdi, L., Lun, A. T. L., Morgan, M. D. & Marioni, J. C. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat. Biotechnol. 36, 421–427 (2018).
Allaire, J. & Chollet, F. keras: R interface to ‘Keras’. https://CRAN.R-project.org/package=keras (2021).
Taiyun, W. & Viliam, S. R package ‘corrplot’: visualization of a correlation matrix (version 0.90) (2021).
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).
Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 9440–9445 (2003).
Zheng, S. et al. Molecular transitions in early progenitors during human cord blood hematopoiesis. Mol. Sys. Biol. 14, e8041 (2018).
Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Methodol. 57, 289–300 (1995).
MacArthur, J. et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 45, D896–D901 (2017).
Li, M. J. et al. GWASdb v2: an update database for human genetic variants identified by genome-wide association studies. Nucleic Acids Res. 44, D869–D876 (2016).
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
Mostafavi, S. et al. A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer’s disease. Nat. Neurosci. 21, 811–819 (2018).
Welch, B. L. The generalisation of student’s problems when several different population variances are involved. Biometrika 34, 28–35 (1947).
Holm, S. A simple sequentially rejective multiple test procedure. Scand. J. Stat. 6, 65–70 (1979).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell https://doi.org/10.1016/j.cell.2021.04.048 (2021).
Griffiths, J. A., Richard, A. C., Bach, K., Lun, A. T. L. & Marioni, J. C. Detection and removal of barcode swapping in single-cell RNA-seq data. Nat. Commun. 9, 2667 (2018).
Patterson-Cross, R. B., Levine, A. J. & Menon, V. Selecting single cell clustering parameter values using subsampling-based robustness metrics. BMC Bioinformatics 22, 39 (2021).
Kuhn, M. et al. caret: classification and regression training. R Core Team. https://CRAN.R-project.org/package=caret (2021).
Fleming, S. J. et al. Unsupervised removal of systematic background noise from droplet-based single-cell experiments using CellBender. Nat. Methods 20, 1323–1335 (2023).
Kang, H. M. et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat. Biotechnol. 36, 89–94 (2018).
Cain, A. et al. Multi-cellular communities are perturbed in the aging human brain and Alzheimer's disease. Nat. Neurosci. 26, 1267–1280 (2023).
Stephens, M. et al. ashr: methods for adaptive shrinkage, using empirical Bayes. R package ashr version 2.2-47 (2022).
Meier, F. et al. diaPASEF: parallel accumulation–serial fragmentation combined with data-independent acquisition. Nat. Methods 17, 1229–1236 (2020).
Kulak, N. A., Pichler, G., Paron, I., Nagaraj, N. & Mann, M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat. Methods 11, 319–324 (2014).
Demichev, V., Messner, C. B., Vernardis, S. I., Lilley, K. S. & Ralser, M. DIA-NN: neural networks and interference correction enable deep proteome coverage in high throughput. Nat. Methods 17, 41–44 (2020).
Yu, G. et al. ggtree: an R package for visualization of tree and annotation data. Bioconductor version: release 3.14. https://doi.org/10.18129/B9.bioc.ggtree (2022).
Wickham, H. Ggplot2: elegant graphics for data analysis. (Springer-Verlag, 2016).
Slowikowski, K. ggrepel: automatically position non-overlapping text labels with ‘ggplot2’. R package version 0.9.1. https://CRAN.R-project.org/package=ggrepel (2021).
Tang, Y., Horikoshi, M. & Li, W. ggfortify: unified interface to visualize statistical results of popular R packages. R. J. 8, 474–485 (2016).
Pedersen, T. L. Patchwork: the composer of plots. https://CRAN.R-project.org/package=patchwork (2020).
Wilke, C. O. cowplot: streamlined plot theme and plot annotations for ‘ggplot2’. https://wilkelab.org/cowpl (2020).
Xiao, N. ggsci: scientific journal and Sci-Fi themed color palettes for ‘ggplot2’. R package version 2.9 https://cran.r-project.org/package=ggsci (2018).
Kolde, R. pheatmap: pretty heatmaps. R package version 1(2):726 https://cran.r-project.org/package=pheatmap (2019).
Blighe, K. et al. EnhancedVolcano: Publication-ready volcano plots with enhanced colouring and labeling. Bioconductor version: release (3.14) https://doi.org/10.18129/B9.bioc.EnhancedVolcano (2022).
Acknowledgements
We thank the individuals and their families who donated the brain samples used in this project. The work was supported by the Chan-Zuckerberg Initiative’s Neurodegeneration Challenge Network grant CS-02018-191971. Some of the work also emerged from support from National Institutes of Health (NIH)/National Institute on Aging (NIA) grants R01 AG070438, U01 AG061356, RF1 AG057473 and R01AG048015. Research reported in this publication was supported by the National Institute of General Medical Sciences of the NIH under award number T32GM007367 and by the National Cancer Institute of the NIH under award number F30CA261090. The Parkinsonism brain bank at Columbia University is supported by the Parkinson’s Foundation. R.A.H. was supported by grant funding from the Huntington Disease Society of America and Hereditary Disease Foundation and was a Columbia University Irving Medical Center ADRC Research Education Component trainee (P30AG066462). We are grateful to the Banner Sun Health Research Institute Brain and Body Donation Program of Sun City, Arizona for the provision of human brain tissue. The Brain and Body Donation Program has been supported by the National Institute of Neurological Disorders and Stroke (U24NS072026 National Brain and Tissue Resource for Parkinson’s Disease and Related Disorders), the NIA (P30AG19610 and P30AG072980 Arizona Alzheimer’s Disease Core Center), the Arizona Department of Health Services (contract 211002, Arizona Alzheimer’s Research Center), the Arizona Biomedical Research Commission (contracts 4001, 0011, 05-901 and 1001 to the Arizona Parkinson’s Disease Consortium) and the Michael J. Fox Foundation for Parkinson’s Research. The Rocky Mountain Multiple Sclerosis Tissue Bank is supported by the National Multiple Sclerosis Society. The Genomics Shared Resource is supported by NIH/NCI P30CA013696. ROSMAP is supported by P30AG10161, P30AG72975, R01AG15819, R01AG17917, U01AG46152 and U01AG61356. ROSMAP resources can be requested at https://www.radc.rush.edu/. Research reported in this publication was partially performed in the Columbia Center for Translational Immunology Flow Cytometry Core, supported in part by the Office of the Director, NIH under award S10OD020056. All figures were created using BioRender.com and Figs. 2–8 and Supplementary Fig. 1 were generated in Inkscape (https://inkscape.org).
Author information
Authors and Affiliations
Contributions
Conceptualization, J.F.T. and P.L.D.; Methodology, J.F.T., M.T., Y.Z., P.A.S., V.M. and P.L.D.; Software, J.F.T. and M.T.; Validation, J.F.T., M.T., V.H. and P.L.D.; Formal analysis, J.F.T., V.M., H.K., C.W. and P.A.S.; Investigation, J.F.T., M.T., V.H., A.K., M.O. and P.L.D.; Resources, B.H., T.R., A.J.L., G.G., G.E.S., N.H., M.F., S.H., T.G.B., J.C., R.K.S., A.F.T., R.A.H., R.N.A., N.S., J.S. and D.A.B.; Data curation, J.F.T., M.O. and V.M.; Writing–original draft, J.F.T. and P.L.D.; Writing–review and editing, all authors; Visualization, J.F.T. and V.H.; Supervision, P.L.D.; Project administration, P.L.D.; Funding acquisition, P.L.D.
Corresponding author
Ethics declarations
Competing interests
R.N.A. is funded by the NIH, DoD, the Parkinson’s Foundation and the Michael. J. Fox Foundation. R.N.A. received consultation fees from Avrobio, Caraway, GSK, Merck, Ono Therapeutics and Genzyme/Sanofi. P.L.D. has served as a consultant for Biogen, Merck-Serono and PureTech. All other authors declare no competing interests.
Peer review
Peer review information
Nature Neuroscience thanks Özgün Gökçe 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 Proportions of overarching cell types in our dataset.
(A) Different cell types are discriminable in UMAP space or by marker genes. Unsupervised Jaccard-Louvain clustering on a kNN neighbor graph delineates distinct cell types, including adaptive immune cells, monocytes, glial/neuronal cells, and erythrocytes. UMAP plots are binned in hexagons: each single hexagon represents a merged representation of all cells falling within the region. The central UMAP plot is colored by the majority cell type. Different cell types are easily distinguishable in 2-D UMAP plots. The other schex-UMAP plots show gene expression values of selected characteristic marker genes projected onto cells. The color gradient bar represents log-normalized gene expression values. Yellow represents the maximal expressed value, while purple represents the lowest expression values. Markers of distinct immune subpopulations are detected in our data: CD8 T-cells (CD8A), NK cells (GZMB), B cells (MS4A1). Similarly, different non-neuronal cells can be detected in our analysis: astrocytes (GFAP), neurons (SNAP25), and oligodendrocytes (OLIG2). Monocytes (LYZ) localize close to our microglial cells and were used for comparative expression of marker genes in Fig. 2b. Red blood cells (HBB) were also easily discriminable. (B) Microglia are the predominant cell type recovered across regions and diseases. Bar plots showing the relative representation of different cell types across different metadata parameters, with each bar summing to 100%. Overall, 95.7% of cells are microglial, 2.2% are adaptive immune, 1.5% are glial/neuronal, 0.4% are monocytic, and 0.3% are erythrocytes. The upper bar plot shows proportion of each overarching cell group across regions, while the lower plot shows the same across diseases. Mono monocytes, RBC red blood cells, LOAD late-onset Alzheimer’s disease, EOAD early onset Alzheimer’s disease, MCI mild cognitive impairment, CNTRL control, DLBD-PD diffuse Lewy body disease-Parkinson’s disease, PSP progressive supranuclear palsy, TLE temporal lobe epilepsy, MS multiple sclerosis, ALS amyotrophic lateral sclerosis, FTD frontotemporal dementia, HD Huntington’s disease, DNET dysembryoplastic neuroepithelial tumor, BA Brodmann area, AWS anterior watershed, OC occipital cortex, TNC temporal neocortex, H hippocampus, TH thalamus, SC spinal cord, SN substantia nigra, FN facial nucleus.
Extended Data Fig. 2 Quality control metrics across our data after downsampling to account for 10x chemistry differences.
(A-F). Violin plots showing the distribution of our cellular data with overlaid boxplots. The center of boxplots is the median, and the hinges of the box span the 25% to 75% percentiles. Whiskers represent 1.5 IQR from the nearest hinge. Outliers are not shown in this visualization, nor are minima or maxima. Further information about metadata traits and number of cells included in each violin plot may be found in Supplementary Table 1 under ‘QC_’ tabs. The distributions of unique molecular identifiers (UMIs) and genes detected on a per-cell level after downsampling are similar across donors (A), clusters (B), genders (C), 10x chemistry versions (D), regions, (E), and diagnoses (F). Notably, after downsampling, differences between 10x chemistry versions in these metrics are largely eliminated. (G) Validation of population stability by resampling and reclustering demonstrates that overlap of gene expression is largely observed for clusters with similarly related families, such as 2 and 4, or for intermediate subsets such as 5 and 3. To evaluate clustering stability, we randomly sampled ¾ of the cells from our dataset and ran our clustering pipeline with identical parameters. We recorded the frequency of ‘misclassification’, where cells were re-clustered into clusters different from the one that contained most cells with the same original classification. This process was repeated between pairs of cells, and repeated 50 times for each comparison. Cells were considered to be classified into the ‘correct’ class if they were assigned correctly in ¾ of classification runs. Otherwise, they were considered ‘misclassified’ into a different cluster. Classification frequency is visualized in a heatmap here. LOAD late-onset Alzheimer’s disease, EOAD early onset Alzheimer’s disease, MCI mild cognitive impairment, CNTRL control, DLBD-PD diffuse Lewy body disease-Parkinson’s disease, PSP progressive supranuclear palsy, TLE temporal lobe epilepsy, MS multiple sclerosis, ALS amyotrophic lateral sclerosis, FTD frontotemporal dementia, HD Huntington’s disease, DNET dysembryoplastic neuroepithelial tumor, BA Brodmann area, AWS anterior watershed, OC occipital cortex, TNC temporal neocortex, H hippocampus, TH thalamus, SC spinal cord, SN substantia nigra, FN facial nucleus.
Extended Data Fig. 3 Microglial proportions across individual donors and donor-region pairings.
(A) Proportions of microglial subtypes across single donors. Proportions of microglial subtypes are plotted by donor, with selected metadata annotated in a header bar above. Each bar represents a single donor and sums to 100%. Samples are clustered hierarchically based on proportions of each subtype. Donors have variability in the exact proportions of different subtypes but exhibit consistent amounts of the most common subtypes in our dataset, clusters 1 through 6. (B) Proportions of microglial subtypes across region-donor pairings. Samples are aggregated to donor-region pairings (for example, AD1-BA9) to give a proportion of different clusters for each region for each individual. Boxplots are computed for specific region-disease pairings showing the median (center), 25% (left hinge), and 75% (right hinge), for the proportion of cells across all samples for which that combination of disease and region was sampled. Whiskers represent 1.5 IQR from the nearest hinge, and outliers are not shown, nor are minima or maxima. Proportions are shown on the x-axis, and the scale varies depending on the cluster in question. [Number of independent samples per category: TNC_TLE (6), TNC_PSP (1), TH_MS (2), SN_PSP (1), SN_LOAD (3), SN_DLBD-PD (5), SN_CNTRL (1), SC_ALS/FTD (2), SC_ALS (9), OC_TLE (1), OC_Stroke_lesion (1), Lesion_MS (1), H_TLE (2), H_PSP (1), H_LOAD (14), H_HD (1), H_FTD (1), H_EOAD (2), H_CNTRL (1), FN_ALS (4), DNET_DNET (1), BA9_Stroke_lesion (1), BA9_PSP (1), BA9_MS (2), BA9_MCI (4), BA9_LOAD (35), BA9_HD (1), BA9_FTD (1), BA9_EOAD (2), BA9_DLBD-PD (5), BA9_CNTRL (1), BA9_ALS/FTD (2), BA9_ALS (8), BA4_CNTRL (1), BA4_ALS/FTD (2), BA4_ALS (9), BA20_LOAD (9), BA20_HD (1), BA20_EOAD (2), AWS_MS (2), AWS_MCI (3), AWS_LOAD (13)].
Extended Data Fig. 4 Further exploration of microglial phenotypes with pseudotime analysis and GO annotation validates our trajectory map and reveals subsets associated with motility, lipid trafficking, and proliferation.
(A) Cluster 5, an intermediate cluster, shows association with motility. On the left, the size of the circle represents the percentage of cells in a cluster that express the gene, with no circle plotted if less than 10% of cells in a cluster express the gene. The color of the circle represents the z-scored expression of the gene. Cluster 5 expresses a transcriptional signature partially overlapping with the core homeostatic or transitional clusters, 2 and 3, but expresses unique sets of genes associated with motility. GO annotation was performed with topGO and summarized with rrvgo. Parent terms are shown in white, overlaid over child terms. Terms associated with motility are enriched in cluster 5. (B) Cluster 12 is associated with oxidative phosphorylation and proliferation. (C) Cluster 11 interfaces with lipids and beta-amyloid. (D) GO annotation of clusters 8/10 parallels results of Reactome pathway analysis, highlighting common immunological activation but divergence in other aspects of phenotype. (E) Trajectories of state shift in pseudotime analysis parallel those seen in other analyses. Monocle3 was used to build a pseudotime trajectory across our dataset, setting the root point at the boundary of clusters 2 and 3. Shifts in pseudotime from this root point reinforces the directionality laid out in the constellation diagram, suggesting that a broad intermediate gradient between a series of terminal points exists, with pseudotime scores in 6-7, 4, and 10 showing most divergence from the root point. GO gene ontology.
Extended Data Fig. 5 Additional representative images from our joint RNAscope/IF and CellProfiler measures highlight morphological differences between expression-defined subtypes.
Representative images are shown for both panel 1 (A) and panel 2 (B) across different diseases. (C) Compactness is highest in the medium classes of CD74, GPX1, and SPP1-defined expression groups. Compactness (a measure of ramification, where high values indicate high ramification) is shown across CD74-, GPX1-, and SPP1-expressing IBA1+ microglial cells quantified using CellProfiler. For this and following panels, significance was calculated with two-sided, two-sample Welch’s t-tests. Multiple testing correction was performed with Holm-Bonferroni correction. For boxplots in these visualizations, the center is the median, and the hinges of the box span the 25% to 75% percentiles. Whiskers represent 1.5 IQR from the nearest hinge. Outliers are shown as circles, but minima and maxima are not explicitly depicted. Significance thresholds for p-values: >0.05 = ns, <0.05 = *, <0.01 = **, <0.005 = ***. (D) Compactness is higher in the CXCR4+ class. (E) Eccentricity is highest in the low classes for CD74 and GPX1. Eccentricity (a measure of shape, where 0 is a circle and 1 is a line), is shown across CD74- and GPX1- expressing Iba1+ microglia. (F) CD74 distance is highest in the CD74 medium group, but also in the CXCR4+ group. CD74 distance (calculated as the median of all puncta for a given cell from the cellular centroid) is shown across CD74-, and CXCR4-expressing Iba1+ microglia. Number of cells per expression class are as follows. CD74: low (3756), medium (3333), high (329), GPX1: low (1404), medium (1653), high (329), SPP1: low (3216), medium (388), high (125), CXCR4: positive (322), negative (7096). 16 tissue sections were stained with panel 1 (CD74/CXCR4) and eight were stained with panel 2 (GPX1/SPP1).
Extended Data Fig. 6 In situ merFISH validation of microglia subtypes.
(A) Projection of microglial cells into the established scRNAseq model. UMAP space showing predicted cluster subtypes within a projected UMAP space (established model shown in greyed-out background). Seven out of twelve microglial subtypes were identified across AD (blue) and non-AD (yellow) cortex tissue, with different observed proportions. Clusters 8/10 show depletion in AD cortex ( < 1%) compared to non-AD cortex (35.7%). (B) Expression signatures of predicted clusters in situ. Microglia predicted to belong to clusters 8/10 show a greater average expression and percent expression of CXCR4, SRGN, and CD74. Showing clusters with at least 5 predicted microglia.
Extended Data Fig. 7 Performance metrics across models trained for different datasets.
Each row contains a different performance metric, while each column represents a single dataset. Training and validation sets were identical, but mNN correction incorporates the query dataset, slightly modifying input data. Accuracy metrics are derived from analysis of the holdout validation set, consisting of approximately 50% of the original dataset not used for training either SVM or XGB models (104902 cells). The first row presents histograms of XGBoost classification confidence for cells in the validation set, highlighting cells below 70% confidence in yellow and below 50% in red (the latter cells are dropped). Most cells in the validation set are classified with high confidence. Row 2 contains a UMAP visualization of classification confidence, revealing higher confidence for cells at the UMAP periphery and lower confidence for intermediate cells. Row 3 shows confusion matrices for the validation set. Row 4 presents sensitivity and specificity per class, which are comparable across different datasets. Row 5 shows boxplots for XGB classification confidence across the 4 classes. Boxplots represent the median (center), 25% (lower hinge), and 75% (upper hinge) percentiles. Whiskers extend to 1.5 times the IQR from the nearest hinge, with more extreme values represented as circles. Minima and maxima are not explicitly depicted. Classification confidence varies substantially depending on the data, with the ROSMAP data being the only dataset where classification confidence for families 167 and 24 is generally comparable to that for 3 and 5. Row 6 contains histograms of XGBoost classification confidence for the query cells. Notably, the glioblastoma and xenograft data have similar classification confidence to the validation set, but the ROSMAP data, and to a lesser extent, the Dräger data, diverge noticeably. Finally, row 7 shows marker gene expression across assigned labels in the query datasets. The size of the circle represents the percentage of cells in each cluster expressing the gene (no circle plotted if less than 10% of cells in a cluster express the gene). The color of the circle represents z-scored expression of the gene. Despite systematic differences, label transfer aligns expression profiles effectively.
Extended Data Fig. 8 Screening of in silico predictions identifies successful hits and compounds that fail to drive predicted signatures.
(A) Schematic overview of workflow for compound treatment. To explore the correct dosage for downstream studies, we conducted dose titration to examine viability of cells after treatment with varying dosages of our drugs. After choosing optimal concentrations, we conducted initial screening with qPCR to select candidates for final validation, then conducted final validation with bulk RNA-seq and proteomics. (B)-(D) qPCR results for different cluster families. Results not shown in Fig. 8b-d are shown here. Some compounds had effects on specific marker genes, but these did not pass our criteria for further study. Bars represent mean fold change expression, and error bars represent SD. All replicates are biological. Number of replicates per experiment as follows - Dorsomorphin: 6hrs: CXCR4 - n = 6, SRGN – n = 7; 24hrs: both n = 6, BX-795: 6hrs: CXCR4 - n = 5, SRGN – n = 8; 24hrs: CXCR4 - n = 3, SRGN – n = 5, BMS-2455421: 6hrs: both - n = 4; 24hrs: CXCR4 - n = 3, SRGN – n = 4, BRD: 6hrs: both - n = 7; 24hrs: TYROPB - n = 6, GPX1 – n = 7, Budesonide: 6hrs: n = 3; 24hrs: n = 3, Naltrexone: 6hrs: n = 3; 24hrs: n = 3, Cytochalasin b: 6hrs: SRGAP2 - n = 6, MEF2A – n = 5; 24hrs: both n = 6.
Extended Data Fig. 9 Different compounds modulate different aspects of the cluster 1/6 signature at the transcriptomic level.
(A) Camptothecin downregulates the cluster 1/6 signature. Bulk RNA-seq was generated from HMC3 cells treated with our candidate drugs for 24 h. Data was analyzed with DESeq2, which fits a negative binomial model to the data then uses Wald significance tests with Benjamini-Hochberg correction, and fold change shrinkage was performed with ashr. To examine the genes associated with cluster families, we took the top 20 non-overlapping genes for each individual cluster in our overarching groupings that were present in the differentially expressed gene list for each compound, irrespective of directionality and plotted them in volcano plots. FDR threshold was set to 0.01 and fold change threshold was set at 1.5. (B) Narciclasine does not upregulate the cluster 1/6 signature. (C) Narciclasine upregulates GO processes also found in cluster 1/6. GO annotation was computed on differentially expressed genes that passed an FDR threshold of 0.01 and a fold change threshold of 1.5. Terms were grouped based on similar etiology and parent terms were overlaid. Notably, Narciclasine drives metabolic shifts such as in nitrogen-containing metabolism, heterocyclic metabolism, and nucleic acid metabolism, that are strongly enriched in clusters 1/6 (Fig. 3a). (D) Narciclasine and Torin-2 drive distinct modules of cluster 1/6 marker genes. Cluster 1/6 genes were selected and shown in a row-scaled, zero-centered heatmap. Columns are individual replicates, and rows are genes. These two compounds appear to drive separate modules of genes associated with cluster 1/6. Camptothecin downregulates almost all 1/6 associated genes.
Extended Data Fig. 10 Representative flow gating images.
Cells that were stained with anti-CD11b and anti-CD45 antibodies and 7AAD were sorted by flow cytometry. Flow gates demonstrate selection of live singlets that are CD45-positive.
Supplementary information
Supplementary Information
Supplementary Fig. 1
Supplementary Table 1
Overview of demographics, hashing strategy and QC.
Supplementary Table 2
Pairwise marker genes across clusters. Differential expression calculations are down with MAST, which fits a two-part generalized regression model using logistic and Gaussian components, and Bonferroni multiple-testing correction.
Supplementary Table 3
ROSMAP trait association results. Analysis was performed in DESeq2, which fits a negative binomial model with Wald tests and Benjamini–Hochberg correction.
Supplementary Table 4
Overview of samples used for in situ confirmation.
Supplementary Table 5
Training results of machine learning models. P values for evaluation calculated with one-sided exact binomial test without multiple-testing correction.
Supplementary Table 6
Results of in silico CMAP analysis by cluster. CMAP uses a two-sided, nonparametric similarity measure based on the weighted Kolmogorov–Smirnov enrichment statistic that includes multiple-testing correction to calculate significance. Tau scores show the percentage of reference perturbations that show stronger connectivity to the query.
Supplementary Table 7
In vitro validation results at the transcriptomic and proteomic levels. qPCR results were analyzed with two-way analysis of variance without multiple-testing correction. Bulk RNA-seq data were analyzed with DESeq2, fitting a negative binomial model with Wald tests and Benjamini–Hochberg correction. Bulk proteomic data were analyzed with pairwise differential testing between DMSO control and each treated condition used Welch’s t-test with Benjamini–Hochberg correction.
Supplementary Code 1
CellProfiler pipeline.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Tuddenham, J.F., Taga, M., Haage, V. et al. A cross-disease resource of living human microglia identifies disease-enriched subsets and tool compounds recapitulating microglial states. Nat Neurosci 27, 2521–2537 (2024). https://doi.org/10.1038/s41593-024-01764-7
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41593-024-01764-7
This article is cited by
-
A versatile mouse model to advance human microglia transplantation research in neurodegenerative diseases
Molecular Neurodegeneration (2025)
-
A dynamic and multimodal framework to define microglial states
Nature Neuroscience (2025)