- Article
- Published:
A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk
- Andrew C. Yang ORCID:orcid.org/0000-0002-6756-47461,2,3,
- Ryan T. Vest3 na1,
- Fabian Kern ORCID:orcid.org/0000-0002-8223-37503,4 na1,
- Davis P. Lee3,
- Maayan Agam3,
- Christina A. Maat3,
- Patricia M. Losada3,
- Michelle B. Chen1,
- Nicholas Schaum3,
- Nathalie Khoury3,
- Angus Toland5,
- Kruti Calcuttawala3,
- Heather Shin3,
- Róbert Pálovics3,
- Andrew Shin3,
- Elizabeth Y. Wang6,
- Jian Luo ORCID:orcid.org/0000-0002-2064-84677,
- David Gate ORCID:orcid.org/0000-0003-0481-96573,
- Walter J. Schulz-Schaeffer ORCID:orcid.org/0000-0001-5886-23228,
- Pauline Chu4,
- Julie A. Siegenthaler9,
- M. Windy McNerney10,
- Andreas Keller ORCID:orcid.org/0000-0002-5361-08953,4 &
- …
- Tony Wyss-Coray ORCID:orcid.org/0000-0001-5893-08312,3,11,12
Naturevolume 603, pages885–892 (2022)Cite this article
120kAccesses
382Citations
294Altmetric
Abstract
The human brain vasculature is of great medical importance: its dysfunction causes disability and death1, and the specialized structure it forms—the blood–brain barrier—impedes the treatment of nearly all brain disorders2,3. Yet so far, we have no molecular map of the human brain vasculature. Here we develop vessel isolation and nuclei extraction for sequencing (VINE-seq) to profile the major vascular and perivascular cell types of the human brain through 143,793 single-nucleus transcriptomes from 25 hippocampus and cortex samples of 9 individuals with Alzheimer’s disease and 8 individuals with no cognitive impairment. We identify brain-region- and species-enriched genes and pathways. We reveal molecular principles of human arteriovenous organization, recapitulating a gradual endothelial and punctuated mural cell continuum. We discover two subtypes of human pericytes, marked by solute transport and extracellular matrix (ECM) organization; and define perivascular versus meningeal fibroblast specialization. In Alzheimer’s disease, we observe selective vulnerability of ECM-maintaining pericytes and gene expression patterns that implicate dysregulated blood flow. With an expanded survey of brain cell types, we find that 30 of the top 45 genes that have been linked to Alzheimer’s disease risk by genome-wide association studies (GWASs) are expressed in the human brain vasculature, and we confirm this by immunostaining. Vascular GWAS genes map to endothelial protein transport, adaptive immune and ECM pathways. Many are microglia-specific in mice, suggesting a partial evolutionary transfer of Alzheimer’s disease risk. Our work uncovers the molecular basis of the human brain vasculature, which will inform our understanding of overall brain health, disease and therapy.
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
9,800 Yen / 30 days
cancel any time
Subscription info for Japanese customers
We have a dedicated website for our Japanese customers. Please go tonatureasia.com to subscribe to this journal.
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
Raw sequencing data have been deposited in the NCBI Gene Expression Omnibus (GEO) under accession codeGSE163577. Data are also available to explore via an interactive web browser:https://twc-stanford.shinyapps.io/human_bbb.
References
Feigin, V. L. et al. Global and regional burden of stroke during 1990–2010: findings from the Global Burden of Disease Study 2010.Lancet383, 245–255 (2014).
Chow, B. W. & Gu, C. The molecular constituents of the blood–brain barrier.Trends Neurosci.38, 598–608 (2015).
Profaci, C. P., Munji, R. N., Pulido, R. S. & Daneman, R. The blood–brain barrier in health and disease: important unanswered questions.J. Exp. Med.217, e20190062 (2020).
Obermeier, B., Daneman, R. & Ransohoff, R. M. Development, maintenance and disruption of the blood–brain barrier.Nat. Med.19, 1584–1596 (2013).
Sweeney, M. D., Zhao, Z., Montagne, A., Nelson, A. R. & Zlokovic, B. V. Blood–brain barrier: from physiology to disease and back.Physiol. Rev.99, 21–78 (2019).
Iadecola, C. The pathobiology of vascular dementia.Neuron80, 844–866 (2013).
Pardridge, W. M. Drug transport across the blood–brain barrier.J. Cereb. Blood Flow Metab.32, 1959–1972 (2012).
Yang, A. C. et al. Physiological blood–brain transport is impaired with age by a shift in transcytosis.Nature583, 425–430 (2020).
Daneman, R., Zhou, L., Kebede, A. A. & Barres, B. A. Pericytes are required for blood–brain barrier integrity during embryogenesis.Nature468, 562–566 (2010).
Armulik, A. et al. Pericytes regulate the blood–brain barrier.Nature468, 557–561 (2010).
Janzer, R. C. & Raff, M. C. Astrocytes induce blood–brain barrier properties in endothelial cells.Nature325, 253–257 (1987).
Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature.Nature554, 475–480 (2018).
Sabbagh, M. F. et al. Transcriptional and epigenomic landscapes of CNS and non-CNS vascular endothelial cells.Elife7, e36187 (2018).
Kalucka, J. et al. Single-cell transcriptome atlas of murine endothelial cells.Cell180, 764–779 (2020).
Chen, M. B. et al. Brain endothelial cells are exquisite sensors of age-related circulatory cues.Cell Rep.30, 4418–4432 (2020).
Mathys, H. et al. Single-cell transcriptomic analysis of Alzheimer’s disease.Nature570, 332–337 (2019).
Grubman, A. et al. A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation.Nat. Neurosci.22, 2087–2097 (2019).
Jäkel, S. et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis.Nature566, 543–547 (2019).
Velmeshev, D. et al. Single-cell genomics identifies cell type–specific molecular changes in autism.Science364, 685–689 (2019).
Keller, D., Erö, C. & Markram, H. Cell densities in the mouse brain: a systematic review.Front. Neuroanat.12, 83 (2018).
Niedowicz, D. M. et al. Obesity and diabetes cause cognitive dysfunction in the absence of accelerated β-amyloid deposition in a novel murine model of mixed or vascular dementia.Acta Neuropathol. Commun.2, 64 (2014).
Montagne, A. et al. Blood–brain barrier breakdown in the aging human hippocampus.Neuron85, 296–302 (2015).
Geirsdottir, L. et al. Cross-species single-cell analysis reveals divergence of the primate microglia program.Cell179, 1609–1622 (2019).
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).
Uhlén, M. et al. Tissue-based map of the human proteome.Science347, 1260419 (2015).
De Meyer, S. F., Stoll, G., Wagner, D. D. & Kleinschnitz, C. Von Willebrand factor: an emerging target in stroke therapy.Stroke43, 599–606 (2012).
Mao, M., Alavi, M. V., Labelle-Dumais, C. & Gould, D. B. Type IV collagens and basement membrane diseases: cell biology and pathogenic mechanisms.Curr. Top. Membr.76, 61–116 (2015).
DeSisto, J. et al. Single-cell transcriptomic analyses of the developing meninges reveal meningeal fibroblast diversity and function.Dev. Cell54, 43–59 (2020).
Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels.Nature523, 337–341 (2015).
Aspelund, A. et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules.J. Exp. Med.212, 991–999. (2015).
Dorrier, C. E. et al. CNS fibroblasts form a fibrotic scar in response to immune cell infiltration.Nat. Neurosci.24, 234–244 (2021).
Iliff, J. J. et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β.Sci. Transl. Med.4, 147ra111 (2012).
Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease.Cell169, 1276–1290 (2017).
Brown, W. R. A review of string vessels or collapsed, empty basement membrane tubes.J. Alzheimer’s Dis.21, 725–739 (2010).
Roher, A. E. et al. Cerebral blood flow in Alzheimer’s disease.Vasc. Health Risk Manag.8, 599 (2012).
Montagne, A. et al.APOE4 leads to blood–brain barrier dysfunction predicting cognitive decline.Nature581, 71–76 (2020).
Rockenstein, E., Mallory, M., Mante, M., Sisk, A. & Masliaha, E. Early formation of mature amyloid-β protein deposits in a mutant APP transgenic model depends on levels of Aβ1–42.J. Neurosci. Res.66, 573–582 (2001).
Nott, A. et al. Brain cell type-specific enhancer–promoter interactome maps and disease-risk association.Science366, 1134–1139 (2019).
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).
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).
Jansen, I. E. et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk.Nat. Genet.51, 404–413 (2019).
Skene, N. G. & Grant, S. G. N. Identification of vulnerable cell types in major brain disorders using single cell transcriptomes and expression weighted cell type enrichment.Front. Neurosci.10, 16 (2016).
Karch, C. M. & Goate, A. M. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis.Biol. Psychiatry77, 43–51 (2015).
Zhao, Z. et al. Central role for PICALM in amyloid-β blood–brain barrier transcytosis and clearance.Nat. Neurosci.18, 978–987 (2015).
Cirrito, J. R. et al. Synaptic activity regulates interstitial fluid amyloid-β levels in vivo.Neuron48, 913–922 (2005).
Safaiyan, S. et al. Age-related myelin degradation burdens the clearance function of microglia during aging.Nat. Neurosci.19, 995–998 (2016).
Spangenberg, E. et al. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model.Nat. Commun.10, 3758 (2019).
Gate, D. et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease.Nature577, 399–404 (2020).
Farh, K. K. H. et al. Genetic and epigenetic fine mapping of causal autoimmune disease variants.Nature518, 337–343 (2015).
Villar, D. et al. Enhancer evolution across 20 mammalian species.Cell160, 554–566 (2015).
Wightman, D. P. et al. Largest GWAS (N = 1,126,563) of Alzheimer’s disease implicates microglia and immune cells. Preprint athttps://doi.org/10.1101/2020.11.20.20235275 (2020).
Lee, Y. K., Uchida, H., Smith, H., Ito, A., & Sanchez, T. The isolation and molecular characterization of cerebral microvessels.Nat. Protoc.14, 3059–3081 (2019).
Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues.Nat. Methods14, 959–962 (2017).
Yang, A. C. et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19.Nature595, 565–571 (2021).
McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint athttps://arxiv.org/abs/1802.03426 (2018).
Satija, R., Farrell, J. A., Gennert, D., Schier, A. F., & Regev, A. Spatial reconstruction of single-cell gene expression data.Nat. Biotechnol.33, 495–502 (2015).
McGinnis, C. S., Murrow, L. M., & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors.Cell Syst.8, 329–337 (2019).
Zeisel, A. et al. Molecular architecture of the mouse nervous system.Cell174, 999–1014 (2018).
Zeisel, A. et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq.Science347, 1138–1142 (2015).
Yang, A. C. et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19.Nature595, 565–571 (2021).
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).
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).
Lake, B. B. et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain.Nat. Biotechnol.36, 70–80 (2018).
Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool.BMC Bioinformatics14, 128 (2013).
Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets.Nat. Commun.10, 1523 (2019).
Conway, J. R., Lex, A. & Gehlenborg, N. UpSetR: an R package for the visualization of intersecting sets and their properties.Bioinformatics33, 2938–2940 (2017).
Jin, S. et al. Inference and analysis of cell–cell communication using CellChat.Nat. Commun.12, 1088 (2021).
The Tabula Muris Consortium. Single-cell transcriptomics of 20 mouse organs creates aTabula Muris.Nature562, 367–372 (2018).
Zuchero, Y. J. Y. et al. Discovery of novel blood–brain barrier targets to enhance brain uptake of therapeutic antibodies.Neuron89, 70–82 (2016).
Yousef, H., et al. Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1.Nat. Med.25, 988–1000 (2019).
Swiech, L. et al. In vivo interrogation of gene function in the mammalian brain using CRISPR–Cas9.Nat. Biotechnol.33, 102–106 (2015).
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).
Yang, A. C. et al. Multiple click-selective tRNA Synthetases expand mammalian cell-specific proteomics.J. Am. Chem. Soc.140, 7046–7051 (2018).
Thul, P. J. et al. A subcellular map of the human proteome.Science356, eaal3321 (2017).
Butovsky, O. et al. Identification of a unique TGF-β–dependent molecular and functional signature in microglia.Nat. Neurosci.17, 131–143 (2014).
Szabo, P. A. et al. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease.Nat. Commun.10, 4706 (2019).
Iadecola, C., Anrather, J. & Kamel, H. Effects of COVID-19 on the nervous system.Cell183, 16–27 (2020).
Månberg, A. et al. Altered perivascular fibroblast activity precedes ALS disease onset.Nat. Med.27, 640–646 (2021).
Parker, K. R. et al. Single-cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies.Cell183, 126–142 (2020).
Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature.Nature554, 475–480 (2018).
Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse.Neuron89, 37–53 (2016).
Acknowledgements
We thank T. Iram, E. Tapp, N. Lu, M. Haney, O. Hahn, M. J. Estrada, S. M. Shi and other members of the Wyss-Coray laboratory for feedback and support; H. Mathys, D. A. Bennett and participants in the CSHL BBB 2021 meeting for advice; and H. Zhang and K. Dickey for laboratory management. This work was funded by the NOMIS Foundation (T.W.-C.), the National Institute on Aging (T32-AG0047126 to A.C.Y. and 1RF1AG059694 to T.W.-C), Nan Fung Life Sciences (T.W.-C.), the Bertarelli Brain Rejuvenation Sequencing Cluster (an initiative of the Stanford Wu Tsai Neurosciences Institute) and the Stanford Alzheimer’s Disease Research Center (P30 AG066515). This work was supported by a grant from the Simons Foundation Award (811253TWC). A.C.Y. was supported by a Siebel Scholarship. F.K. and A.K. are part of the CORSAAR study supported by the State of Saarland, the Saarland University and the Rolf M. Schwiete Stiftung. This study was supported by the AHA–Allen Initiative in Brain Health and Cognitive Impairment (19PABHI34580007). The statements in this work are solely the responsibility of the authors and do not necessarily represent the views of the American Heart Association (AHA) or the Paul G. Allen Frontiers Group. Graphics were created with BioRender.com.
Author information
These authors contributed equally: Ryan T. Vest, Fabian Kern
Authors and Affiliations
Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
Andrew C. Yang & Michelle B. Chen
Bakar Aging Research Institute, University of California San Francisco, San Francisco, CA, USA
Andrew C. Yang & Tony Wyss-Coray
Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
Andrew C. Yang, Ryan T. Vest, Fabian Kern, Davis P. Lee, Maayan Agam, Christina A. Maat, Patricia M. Losada, Nicholas Schaum, Nathalie Khoury, Kruti Calcuttawala, Heather Shin, Róbert Pálovics, Andrew Shin, David Gate, Andreas Keller & Tony Wyss-Coray
Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
Fabian Kern, Pauline Chu & Andreas Keller
Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
Angus Toland
Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
Elizabeth Y. Wang
Veterans Administration Palo Alto Healthcare System, Palo Alto, CA, USA
Jian Luo
Institute for Neuropathology, Saarland University Hospital and Medical Faculty of Saarland University, Homburg, Germany
Walter J. Schulz-Schaeffer
Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
Julie A. Siegenthaler
Department of Psychiatry, Stanford University School of Medicine, Stanford, CA, USA
M. Windy McNerney
Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
Tony Wyss-Coray
Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
Tony Wyss-Coray
- Andrew C. Yang
You can also search for this author inPubMed Google Scholar
- Ryan T. Vest
You can also search for this author inPubMed Google Scholar
- Fabian Kern
You can also search for this author inPubMed Google Scholar
- Davis P. Lee
You can also search for this author inPubMed Google Scholar
- Maayan Agam
You can also search for this author inPubMed Google Scholar
- Christina A. Maat
You can also search for this author inPubMed Google Scholar
- Patricia M. Losada
You can also search for this author inPubMed Google Scholar
- Michelle B. Chen
You can also search for this author inPubMed Google Scholar
- Nicholas Schaum
You can also search for this author inPubMed Google Scholar
- Nathalie Khoury
You can also search for this author inPubMed Google Scholar
- Angus Toland
You can also search for this author inPubMed Google Scholar
- Kruti Calcuttawala
You can also search for this author inPubMed Google Scholar
- Heather Shin
You can also search for this author inPubMed Google Scholar
- Róbert Pálovics
You can also search for this author inPubMed Google Scholar
- Andrew Shin
You can also search for this author inPubMed Google Scholar
- Elizabeth Y. Wang
You can also search for this author inPubMed Google Scholar
- Jian Luo
You can also search for this author inPubMed Google Scholar
- David Gate
You can also search for this author inPubMed Google Scholar
- Walter J. Schulz-Schaeffer
You can also search for this author inPubMed Google Scholar
- Pauline Chu
You can also search for this author inPubMed Google Scholar
- Julie A. Siegenthaler
You can also search for this author inPubMed Google Scholar
- M. Windy McNerney
You can also search for this author inPubMed Google Scholar
- Andreas Keller
You can also search for this author inPubMed Google Scholar
- Tony Wyss-Coray
You can also search for this author inPubMed Google Scholar
Contributions
A.C.Y. and T.W.-C. conceptualized the study. A.C.Y. devised the isolation method. M.W.M. and W.J.S.-S. provided and A.C.Y. organized tissue samples. D.P.L. and A.C.Y. performed tissue dissociations. N.S., R.T.V., D.G., K.C., H.S. and A.C.Y. prepared libraries for sequencing. R.T.V., F.K., A.K., C.A.M., M.B.C., R.P., A.S., N.K., J.A.S. and A.C.Y. performed computational analysis. D.P.L., C.A.M., M.A., D.G., E.Y.W., J.L. A.T., P.C. and A.C.Y. performed immunohistochemical stains. P.M.L. developed the searchable web interface (Shiny app). C.A.M. and A.C.Y. drew diagrams. A.C.Y. wrote the manuscript with input from all authors. A.C.Y. and T.W.-C. supervised the study.
Corresponding authors
Correspondence toAndrew C. Yang orTony Wyss-Coray.
Ethics declarations
Competing interests
T.W.-C. is a co-founder and scientific advisor of Alkahest. A.C.Y., R.T.V. and T.W.-C. are co-founders and scientific advisors of Qinotto.
Peer review
Peer review information
Nature thanks Trygve Bakken, Neelroop Parikshak 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 figures and tables
Extended Data Fig. 1 Enhanced capture and characterization of human brain vascular nuclei.
a, Detailed schematic of the VINE-seq method to capture human brain vascular and immune cell types for single-nucleus sequencing.b, Total number of nuclei, median number of unique molecular identifiers (UMI), and median number of genes for each human sample sequenced from hippocampus and superior frontal cortex.c, Quantification of the median number of genes detected per nuclei across groups (n = 17 hippocampus andn = 8 cortex;n = 8 NCI andn = 9 AD, two-sidedt-test; mean +/− s.e.m.).d,e, Quantification of the number (d) and proportion (e) of cerebrovascular cell types captured via the VINE-seq method introduced here compared to recent snRNA-seq studies16,17.f, Summary quantification of the proportion of captured cell types, by individuals with NCI and individuals with AD.g, Quantification of the proportion of captured cell types across individuals.h, Summary (left) and quantification (right) of the proportion of captured cell types by brain region (n = 17 hippocampus andn = 8 cortex;n = 8 NCI andn = 9 AD, two-sidedt-test; mean +/− s.e.m.).
Extended Data Fig. 2 Diversity and heterogeneity of human brain vascular cell types.
a, Representative H&E images used by two neuropathologists to evaluate NCI cortical and hippocampal tissue for vascular pathology. No significant vascular pathology was observed. Scale bars, 200 μm.b, Discovery of the top cell-type-specific marker genes across the major classes of cells captured. The colour bar indicates gene expression from low (blue) to high (yellow).c, Validation of cell-type annotations and confirmation of minimal doublet contamination using established cell-type markers.d, UMAP projection of captured myeloid cells, forming two distinct clusters corresponding to parenchymal microglia and brain barrier macrophages. Example marker genes listed.e, Immunohistochemical validation of microglial and perivascular macrophage markers. Scale bars, 50 μm. Image credit: Human Protein Atlas25,74 (http://www.proteinatlas.org).f, Global view of DEGs comparing human brain macrophages and microglia (left, MAST, Benjamini–Hochberg correction; FDR < 0.01 and logFC>0.5 [log2FC>0.72] to be coloured significant). Pathways enriched in microglia versus macrophages (right), recapitulating interesting biology such as the unique TGF-β molecular signature in microglia75.g, Expression of top gene markers for various T cell subtypes (top), and quantification of their expression as a module (bottom)76. Brain T cells exhibit highest expression of markers corresponding to CD8 cytotoxic and CD4 Naive/Central memory (NV/CM) T cells.h, UMAP projection of captured astrocytes, forming two distinct clusters, and split by brain region. Example marker genes listed.i–j, Quantification of astrocyte cluster 0 (b) and 1 (c) frequency in the cortex and hippocampus (n = 8 cortex andn = 17 hippocampus, Mann-Whitneyt-test; mean +/− s.e.m.).k, Immunohistochemical validation of the brain region-specific astrocyte markerTENM4. Scale bars, 50 μm.
Extended Data Fig. 3 Species-specific gene expression across brain cell types and their pharmacological relevance.
a–f, Identification of species-specific genes. Both mouse and human transcriptomes were generated and analysed similarly via single-nucleus RNA-sequencing. Mice were 19 months of age to match the average age of our human cohort. Species-specific/enriched are coloured.g, Immunohistochemical confirmation of genes predicted to be enriched or specific to human cerebrovascular cells compared to mouse (isolated mouse nuclei and per Vanlandewijck, et al., 2018)12, in terms of overall expression or zonation. In parenthesis is the cell type predicted to be uniquely or exhibiting enriched expressed in human over mouse. Scale bars, 50 μm. Image credit: Human Protein Atlas25,74 (http://www.proteinatlas.org).h–i, Mouse and human BEC expression of genes mediating protein transcytosis (h) and small molecule influx and efflux (i).
Extended Data Fig. 4 Human brain vascular expression of genes relevant to disease.
a, Brain vascular expression of genes relevant to SARS-CoV-2 brain entry, as summarized in Iadecola, et al. 202077.b, Expression of the mouse perivascular fibroblast-like geneSpp1 is instead specifically expressed in human myeloid cells and oligodendrocytes (SPP1, top)78.c, No expression of the immuno-oncology targetCD19 and its chaperoneCD81 across human adult brain pericytes and SMCs79. Note: cells with any finite expression are ordered to the front to ensure all expression is visible, but this carries the potential to visually overestimate average expression.
Extended Data Fig. 5 Brain endothelial and mural cell zonation and subpopulations.
a, UMAP projection of captured BECs, organizing by arteriovenous zonation. Bottom, tip cell markers expressed in the tip-like/ proteostatic EC cluster.b, Validation of BEC zonation clusters using established zonation markers12. Violin plots are centred around the median, with their shape representing cell distribution.c,d, As ina,b but for pericytes and SMCs. Note that the anatomical locations of pericyte 0 and 1 have not yet been determined.e, Immunohistochemical validation ofACTA2 (α-SMA) expression in human SMCs and less so in capillary pericytes. A denotes arterial and C denotes capillary. Arrowheads specify capillary pericytes expressingACTA2. Scale bars, 50 μm.f,g, As ina,b but for perivascular fibroblast-like cells, as recently discovered in mice12.
Extended Data Fig. 6 Brain endothelial zonation and mural cell subtype markers.
a, Immunohistochemical validation of zonation and cell subtype markers in BECs. Scale bars, 50 μm. Image credit: Human Protein Atlas25,74 (http://www.proteinatlas.org).b, Comparison of the zonal specificity of genes in arterial, capillary, and venous cells. Axis plot a specificity score, as defined in the Methods. For example, specificity score for capillaries = avg(logFC(cap/ven), logFC(cap/art)).c, Immunohistochemical validation of capillary expression in human brains of the mouse venous-specific marker VWF and CA4, with similar patterns observed across multiple primary antibody clones. Scale bars, 100 μm. Image credit: Human Protein Atlas25,74 (http://www.proteinatlas.org).d, Immunohistochemical validation of zonation and cell subtype markers in brain SMCs and pericytes. Scale bars, 50 μm. Image credit: Human Protein Atlas25,74 (http://www.proteinatlas.org).
Extended Data Fig. 7 Specialization and functions of human brain fibroblasts.
a, Expression of example markers demarcating perivascular from meningeal fibroblasts.b, UMAP of 428 meningeal fibroblast nuclei, subclustering into anatomically segregated dural and arachnoid space fibroblasts.c, Expression of the genes constituting the major fibrotic scar component collagen I in pericytes and fibroblasts. Collagen I is composed of two components, COL1A1 and COL1A2. Column annotations: T-PC = solute transport pericyte and M-PC = Extracellular matrix regulating pericyte, P. FB = Perivascular fibroblast, and M. FB = Meningeal fibroblast.d,e, Protein immunostaining validation of polarized expression of human brain meningeal and perivascular fibroblast pumps: the common marker CYP1B1 (d, serves as a control) and the meningeal fibroblast-specific influx pump SLC47A1 (e). Scale bars, 50 μm.f, Overlap between the top 100 perivascular fibroblast-like cell markers and those identified in mice. A more lenient set of 500 (instead of 100) mouse markers80 were used for comparison to ensure claims of species-specificity were robust. Note: the species-conservation of a cell-type marker depends on species-specific changes in the given cell type and changes amongst the remaining background cell types.
Extended Data Fig. 8 Vascular cell-type-specific perturbations in patients with AD and ApoE4 carriers.
a, Immunohistochemistry with anti-β-amyloid antibody (D54D2, white), Thioflavin S (green), and Hoechst (blue) in the hippocampus of individuals with NCI and individuals with AD. Scale bars, 40 μm.b, Quantification of β-amyloid immunostaining ina for overall β-amyloid (n = 4 NCI and AD, two-sidedt-test; mean +/− s.e.m.).c, As inb but for cored and neuritic β-amyloid plaques (n = 3 NCI and AD, two-sidedt-test; mean +/− s.e.m.).d, UMAP of 143,793 nuclei captured from 17 human hippocampus and superior frontal cortex samples, coloured by AD diagnosis.e, Quantification controls for Fig.5b. Quantification of Collagen IV+ vasculature (left) and number of total (regardless of Collagen IV+ overlap) Hoechst+ nuclei (n = 5 NCI and AD, nested two-sidedt-test; mean +/− s.e.m.).f, Matrix layout for intersections of AD DEGs shared across and specific to each cell type. Circles in the matrix indicate sets that are part of the intersection, showing that most DEGs are cell-type-specific.g, Example DEGs in AD: arterial (Art), capillary (Cap), venous (Vein), pericyte (Peri), perivascular fibro blast-like cell (P. fibro), and SMC. Blue arrow indicates upregulated and grey arrow downregulated genes.h, Summary of the number of AD DEGs by pericyte class: T-, M-, and all pericytes combined to evaluate DEGs that could arise due to a disproportionate loss of M-pericytes in AD.i, DEG counts for each cell type in ApoE4 carriers (n = 5 ApoE3/3,n = 11 ApoE3/4 or ApoE4/4): arterial (Art), capillary (Cap), venous (Vein), pericyte (Peri), perivascular fibro blast-like cell (P. fibro), and SMC. The intensity of the blue colour and the size of the squares are proportional to entry values.j, Matrix layout for intersections of ApoE4 DEGs shared across and specific to each cell type. Circles in the matrix indicate sets that are part of the intersection, showing that most DEGs are cell-type-specific.k, Immunohistochemical validation of the predicted upregulated anti-inflammatory DEGSLC39A10 in venous BECs of ApoE4 carriers. Scale bars, 50 μm (n = 4 ApoE3/3 and ApoE4 carriers, nested two-sidedt-test; mean +/− s.e.m.).l, Among patients with both hippocampus and superior frontal cortex profiled (n = 4 NCI andn = 4 AD), quantification of the relative abundance of major vascular cell types (NCI hippocampus set as reference, unpaired two-sidedt-test; mean +/− s.e.m.). *BECP = 0.0260, **BEC P = 0.0023, *Pericyte P (left) = 0.0357, *Pericyte P (mid) = 0.0237, **Pericyte P = 0.0077, **SMC P = 0.0075, *Fibroblast P = 0.0109, *Astrocyte P = 0.0357.m, As in(l), but comparison of the number of DEGs between brain regions for each cerebrovascular cell type. Analysis done separately for NCI and AD samples (n = 7 cell types, unpaired two-sidedt-test; mean +/− s.e.m.).
Extended Data Fig. 9 Re-evaluation and characterization of top AD GWAS genes expressed in the human brain vasculature.
a, Heterogeneous expression of AD GWAS genes across T- and M-pericyte subtypes.b, RNA-seq data of the predicted T cell-specific AD GWAS genesEPHA1 andABCA7 in an independent dataset81, corroborating minimal expression across resident/ parenchymal brain cells.c, Immunohistochemical confirmation of vascular localization of proteins encoded by 12 top AD GWAS genes froma. Scale bars, 25 μm. Arrowheads in APOE point to signal around larger-diameter vessels, consistent with predicted SMC expression. Image credit: Human Protein Atlas25,74 (http://www.proteinatlas.org).d, Heat map comparing expression patterns of top AD GWAS genes in the hippocampus and superior frontal cortex: e.g., several microglia-expressed GWAS genes likeAPOE,MS4A4A, andTREM2 are more highly expressed in hippocampal compared to cortical microglia/ macrophages.e, GWAS genes found to be expressed specifically in microglia among cells captured using the conventional nuclei isolation process (from Grubman et al. 2019)17 are also expressed in vascular cells (asterisks).f, Summary of AD GWAS genes enriched in microglia and vascular cells mediating common pathways in protein clearance and inflammation. Mouse and human superscripts denote whether expression has been confirmed in that species for a given gene. Proposed model is described in Discussion.
Extended Data Fig. 10 Brain vascular and perivascular expression of AD and AD-related GWAS genes.
a, Expression of AD and AD-related GWAS risk genes (from Grubman et al. 2019)17 across human vascular cells.b, Enriched biological pathways amongst AD and AD-related trait GWAS genes expressed in each cell type.c, For each cell type, the top 10 most specifically expressed AD and AD-related trait GWAS genes.
Supplementary information
Supplementary Table 1
Patient samples.
Supplementary Table 2
Cell type markers.
Supplementary Table 3
Brain region enriched genes.
Supplementary Table 4
Mouse versus human gene expression.
Supplementary Table 5
Vascular cell subtype markers.
Supplementary Table 6
Vascular Alzheimer’s disease differentially expressed genes.
Supplementary Table 7
Vascular ApoE4 carrier disease differentially expressed genes.
Supplementary Table 8
Expression of top Alzheimer’s disease and related GWAS genes.
Rights and permissions
About this article
Cite this article
Yang, A.C., Vest, R.T., Kern, F.et al. A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk.Nature603, 885–892 (2022). https://doi.org/10.1038/s41586-021-04369-3
Received:
Accepted:
Published:
Issue Date:
This article is cited by
Edaravone dexborneol alleviates pericyte-mediated fibrosis depositing extracellular matrix through TGF-β1/IL-11 in cerebral small vessel disease
- Qingrui Duan
- Zhiyang Liu
- Lijuan Wang
Journal of Translational Medicine (2025)
Cell-specific transcriptional signatures of vascular cells in Alzheimer’s disease: perspectives, pathways, and therapeutic directions
- Soumilee Chaudhuri
- Minyoung Cho
- Kwangsik Nho
Molecular Neurodegeneration (2025)
How the gut microbiota impacts neurodegenerative diseases by modulating CNS immune cells
- Philipp Schaible
- Julia Henschel
- Daniel Erny
Journal of Neuroinflammation (2025)
Peripheral and central neuroimmune mechanisms in Alzheimer’s disease pathogenesis
- Shuo Zhang
- Yue Gao
- Xin Wang
Molecular Neurodegeneration (2025)
Molecular and cellular characteristics of cerebrovascular cell types and their contribution to neurodegenerative diseases
- Francisco J. Garcia
- Myriam Heiman
Molecular Neurodegeneration (2025)