Skip to main content

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

  • Article
  • Published:

Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease

Abstract

The gene-regulatory landscape of the brain is highly dynamic in health and disease, coordinating a menagerie of biological processes across distinct cell types. Here, we present a multi-omic single-nucleus study of 191,890 nuclei in late-stage Alzheimer’s disease (AD), accessible through our web portal, profiling chromatin accessibility and gene expression in the same biological samples and uncovering vast cellular heterogeneity. We identified cell-type-specific, disease-associated candidate cis-regulatory elements and their candidate target genes, including an oligodendrocyte-associated regulatory module containing links to APOE and CLU. We describe cis-regulatory relationships in specific cell types at a subset of AD risk loci defined by genome-wide association studies, demonstrating the utility of this multi-omic single-nucleus approach. Trajectory analysis of glial populations identified disease-relevant transcription factors, such as SREBF1, and their regulatory targets. Finally, we introduce single-nucleus consensus weighted gene coexpression analysis, a coexpression network analysis strategy robust to sparse single-cell data, and perform a systems-level analysis of the AD transcriptome.

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: Single-nucleus ATAC-seq and single-nucleus RNA-seq for the study of cellular diversity in the diseased brain.
Fig. 2: Epigenetically and transcriptionally distinct cell subpopulations in human AD prefrontal cortex.
Fig. 3: Linking cis-regulatory elements to downstream target genes in specific cell types.
Fig. 4: Cell-subpopulation-specific transcription factor regulation in late-stage AD.
Fig. 5: Multi-omic oligodendrocyte trajectory analysis.
Fig. 6: Multi-omic microglia and astrocyte trajectory analyses.
Fig. 7: Cell-type-specific regulatory landscapes of GWAS loci in the AD brain.
Fig. 8: Robust gene expression modules revealed using integrated bulk and single-cell coexpression network analysis.

Similar content being viewed by others

Data availability

All the multi-omics raw and processed data are available at https://www.synapse.org/#!Synapse:syn22079621/. Raw sequencing data have been deposited into the National Center for Biotechnology Information Gene Expression Omnibus under accession number GSE174367. Additionally, these data can be accessed through our web app at http://swaruplab.bio.uci.edu/singlenucleiAD. Source data are provided with this paper.

Code availability

The custom code used for this paper is available on GitHub74 (https://doi.org/10.5281/zenodo.4681643) at https://github.com/swaruplab/Single-nuclei-epigenomic-and-transcriptomic-landscape-in-Alzheimer-disease/.

References

  1. Lake, B. B. et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science 352, 1586–1590 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tasic, B. et al. Adult mouse cortical cell taxonomy revealed by single cell transcriptomics. Nat. Neurosci. 19, 335–346 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hodge, R. D. et al. Conserved cell types with divergent features in human versus mouse cortex. Nature 573, 61–68 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. 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).

    Article  CAS  PubMed  Google Scholar 

  8. Leng, K. et al. Molecular characterization of selectively vulnerable neurons in Alzheimer’s disease. Nat. Neurosci. 24, 276–287 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Del-Aguila, J. L. et al. A single-nuclei RNA-sequencing study of Mendelian and sporadic AD in the human brain. Alzheimers. Res. Ther. https://doi.org/10.1186/s13195-019-0524-x (2019).

  10. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Carrasquillo, M. M. et al. Genetic variation in PCDH11X is associated with susceptibility to late-onset Alzheimer’s disease. Nat. Genet. 41, 192–198 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Harold, D. et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet. 41, 1088–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hollingworth, P. et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat. Genet. 43, 429–436 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hibar, D. P. et al. Novel genetic loci associated with hippocampal volume. Nat. Commun. 8, 13624 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nott, A. et al. Brain cell-type-specific enhancer–promoter interactome maps and disease-risk association. Science 366, 1134–1139 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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).

    Article  CAS  PubMed  Google Scholar 

  20. Corces, M. R. et al. Single-cell epigenomic analyses implicate candidate causal variants at inherited risk loci for Alzheimer’s and Parkinson’s diseases. Nat. Genet. 52, 1158–1168 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Welch, J. D. et al. Single-cell multi-omic integration compares and contrasts features of brain cell identity. Cell 177, 1873–1887 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: Uniform Manifold Approximation and Projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  24. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. ChromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Masuda, T. et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature 566, 388–392 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Habib, N. et al. Disease-associated astrocytes in Alzheimer’s disease and aging. Nat. Neurosci. https://doi.org/10.1038/s41593-020-0624-8 (2020).

  30. Pliner, H. A. et al. Cicero predicts cis-regulatory DNA interactions from single-cell chromatin accessibility data. Mol. Cell 71, 858–871 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Satoh, J. I., Kawana, N. & Yamamoto, Y. Pathway analysis of ChIP–seq-based NRF1 target genes suggests a logical hypothesis of their involvement in the pathogenesis of neurodegenerative diseases. Gene Regul. Syst. Bio. 2013, 139–152 (2013).

    Google Scholar 

  32. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Qiu, X. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Mitra, R. & Maclean, A. L. RVAgene: generative modeling of gene expression time-series data. Preprint at bioRxiv https://doi.org/10.1101/2020.11.10.375436 (2020).

  36. 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 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Shimano, H. & Sato, R. SREBP-regulated lipid metabolism: convergent physiology-divergent pathophysiology. Nat. Rev. Endocrinol. 13, 710–730 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Mohamed, A., Viveiros, A., Williams, K. & De Chaves, E. P. Aβ inhibits SREBP-2 activation through Akt inhibition. J. Lipid Res. 59, 1–13 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. 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 

  41. Pugacheva, E. M. et al. CTCF mediates chromatin looping via N-terminal domain-dependent cohesin retention. Proc. Natl Acad. Sci. USA 117, 2020–2031 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim, S., Yu, N. K. & Kaang, B. K. CTCF as a multifunctional protein in genome regulation and gene expression. Exp. Mol. Med. 47, e166 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ferrari, R. et al. Frontotemporal dementia and its subtypes: a genome-wide association study. Lancet Neurol. 13, 686–699 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Chen, J. A. et al. Joint genome-wide association study of progressive supranuclear palsy identifies novel susceptibility loci and genetic correlation to neurodegenerative diseases. Mol. Neurodegener. 13, 41 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Pardiñas, A. F. et al. Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection. Nat. Genet. 50, 381–389 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Andlauer, T. F. M. et al. Novel multiple sclerosis susceptibility loci implicated in epigenetic regulation. Sci. Adv. 2, 1–12 (2016).

    Article  CAS  Google Scholar 

  48. Liu, J. Z. et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 47, 979–986 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wood, A. R. et al. Defining the role of common variation in the genomic and biological architecture of adult human height. Nat. Genet. 46, 1173–1186 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Willer, C. J. et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1285 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cusanovich, D. A., Hill, A. J., Disteche, C. M., Trapnell, C. & Shendure, J. A single-cell atlas of in vivo mammalian chromatin accessibility. Cell 174, 1309–1324 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ulirsch, J. C. et al. Interrogation of human hematopoiesis at single-cell and single-variant resolution. Nat. Genet. 51, 683–693 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Zhang, B. & Horvath, S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 4, 1128 (2005).

    Article  Google Scholar 

  55. Zhang, B. et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 153, 707–720 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Rexach, J. E. et al. Tau pathology drives dementia risk-associated gene networks toward chronic inflammatory states and immunosuppression. Cell Rep. 33, 108398 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Morabito, S., Miyoshi, E., Michael, N. & Swarup, V. Integrative genomics approach identifies conserved transcriptomic networks in Alzheimer’s disease. Hum. Mol. Genet. 29, 2899–2919 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Swarup, V. et al. Identification of conserved proteomic networks in neurodegenerative dementia. Cell Rep. 31, 107807 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Swarup, V. et al. Identification of evolutionarily conserved gene networks mediating neurodegenerative dementia. Nat. Med. 25, 152–164 (2019).

    Article  CAS  PubMed  Google Scholar 

  61. Allen, M. et al. Conserved brain myelination networks are altered in Alzheimer’s and other neurodegenerative diseases. Alzheimer’s Dement. 14, 352–366 (2018).

    Article  Google Scholar 

  62. Wu, Y. E., Pan, L., Zuo, Y., Li, X. & Hong, W. Detecting activated cell populations using single-cell RNA-seq. Neuron 96, 313–329 (2017).

    Article  CAS  PubMed  Google Scholar 

  63. Sun, Y., Ip, P. & Chakrabartty, A. Simple elimination of background fluorescence in formalin-fixed human brain tissue for immunofluorescence microscopy. J. Vis. Exp. 127, 56188 (2017).

    Google Scholar 

  64. Stuart, T., Srivastava, A., Lareau, C. & Satija, R. Multimodal single-cell chromatin analysis with Signac. Preprint at bioRxiv https://doi.org/10.1101/2020.11.09.373613 (2020).

  65. Granja, J. M. et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat. Genet. 53, 403–411 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  67. Khan, A. et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46, D260–D266 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Gaujoux, R. & Seoighe, C. A flexible R package for nonnegative matrix factorization. BMC Bioinformatics 11, 367 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Kuleshov, M. V. et al. Enrichr: a comprehensive gene-set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Lage, K. et al. A human phenome–interactome network of protein complexes implicated in genetic disorders. Nat. Biotechnol. 25, 309–316 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Stark, C. BioGRID: a general repository for interaction datasets. Nucleic Acids Res. 34, D535–D539 (2006).

    Article  CAS  PubMed  Google Scholar 

  73. Szklarczyk, D. et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Morabito, S. & Swarup Lab. swaruplabUCI/single-nuclei-epigenomic-and-transcriptomic-landscape-in-Alzheimer-disease: publication. Zenodo https://doi.org/10.5281/zenodo.4681643 (2021).

Download references

Acknowledgements

Funding for this work was provided by UCI and UCI MIND start-up funds, a National Institute on Aging grant 1RF1AG071683, Adelson Medical Research Foundation funds and an American Federation of Aging Research young investigator award to V.S. The funders had no role in the study design, data collection and analysis, decision to publish or in the preparation of the manuscript. We thank the New York Genome Center for sequencing our bulk and single-nucleus RNA-seq libraries and UCI’s Genomic High Throughput Facility for providing their facilities and sequencing our single-nucleus ATAC-seq libraries. In addition, we thank J. Atwood and UCI’s Institute for Immunology Flow Cytometry Core for assisting us in FACS sorting. We also thank K. Green and F. LaFerla for generously providing their imaging facilities and M. Arreola, Y.-Q. Xue and I. Cobos for their technical advice. This work utilized the infrastructure for high-performance and high-throughput computing, research data storage and analysis, and scientific software tool integration built, operated and updated by the Research Cyberinfrastructure Center (RCIC) at the University of California, Irvine (UCI). For ROSMAP, study data were provided by the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago. Data collection was supported through funding by National Institute on Aging grants P30AG10161, R01AG15819, R01AG17917, R01AG30146, R01AG36836, U01AG32984 and U01AG46152, the Illinois Department of Public Health and the Translational Genomics Research Institute. The results presented here are in whole or in part based on data obtained from the AMP-AD Knowledge Portal (https://doi.org/10.7303/syn2580853).

Author information

Authors and Affiliations

Authors

Contributions

S.M., E.M., N.M. and V.S. planned and directed all experiments and analyses and wrote the manuscript with assistance from all authors. N.M., E.M. and V.S. performed all dissections and sequencing experiments, and S.M., E.M. and V.S. performed all bioinformatic analyses. A.C.M., E.H., J.S., K.L., M.P.-R., E.M., N.M. and V.S. reviewed clinical information. E.M. performed RNAscope validation, and N.M., S.S. and A.C.M. performed immunofluorescence/immunohistochemistry.

Corresponding author

Correspondence to Vivek Swarup.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Goncalo Castelo-Branco, Alexandra Grubman and Yin Shen for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Batch correction of snATAC-seq, snRNA-seq, and merged datasets.

a, snRNA-seq UMAPs before (left) and after iNMF batch correction (right), colored by sequencing batch. b, snATAC-seq UMAPs before (left) and after MNN batch correction (right), colored by sequencing batch. c, Dot plot of iNMF metagene expression in each snRNA-seq cluster. d, snRNA-seq UMAPs colored by metagene expression of selected iNMF metagenes. e, Dot plots showing the iNMF loading for the top 30 genes for the same metagenes in d.

Extended Data Fig. 2 Cell-type immunostaining and in situ hybridization.

a–d, Representative immunofluorescence images from postmortem human brain tissue from control and late-stage AD cases for Iba-1 (a), GFAP (b) MAP2 (c), and 6E10 (d). e, Quantification of 6E10-positive amyloid plaques. n = 3 cognitively healthy controls, 3 late-stage AD. Data is presented as the average of three different sections per sample. Linear mixed-effects model **** p < 0.0001. Box boundaries and line correspond to the interquartile range (IQR) and median respectively. Whiskers extend to the lowest or highest data points that are no further than 1.5 times the IQR from the box boundaries. f, Representative immunofluorescence images from postmortem human brain tissue from control and late-stage AD cases for OLIG2 with PDGFRA co-labeling. g, h, Representative RNAscope images from postmortem human brain tissue from control and late-stage AD cases for CNP (g) and PLP1 (h) with DAPI counterstain.

Extended Data Fig. 3 Comparison of gene expression and gene activity.

a, Scatter plot comparing average gene activity from snATAC-seq and average gene expression from snRNA-seq by each major cell-type, with accompanying Pearson correlation statistics and linear regression lines. b, Donut chart showing the percent of genes with high chromatin accessibility and low gene expression in grey for each major cell-type. High chromatin accessibility was defined as genes in the top 20% of gene activity, while low gene expression was defined as genes in the bottom 20% of gene expression. Percent of all other genes are colored by the cell-type.

Extended Data Fig. 4 NEAT1 validation and neuronal TFs.

a, b, Representative RNAscope images from postmortem human brain tissue for NEAT1 and AQP4 staining (a) and NEAT1 and MOG staining (b) with DAPI nuclear counterstain. c, Boxplots showing quantification of NEAT1 puncta per AQP4 + astrocyte as in a. n = 4 cognitively healthy controls, 5 late-stage AD. d, Boxplots showing quantification of NEAT1 puncta per MOG + oligodendrocyte as in b. n = 4 cognitively healthy controls, 4 late-stage AD. Data is represented as the mean of four equally sized regions per sample. Linear mixed-effects model e, Tn5 bias subtracted TF footprinting for JUN by snATAC-seq neuron cluster (top) and by AD diagnosis (bottom), with TF binding motif logo above and Tn5 bias insertions below. f, Left: Co-embedding UMAP colored by JUN motif variability (top) and JUN target gene score (bottom). Right: Violin plots of JUN motif variability (top) and JUN target gene score (bottom) in excitatory neuron clusters, split by diagnosis. Wilcoxon test (ns: p > 0.05, *: p < = 0.05, **: p < = 0.01, ***: p < = 0.001, ****: p < = 0.0001). g, Tn5 bias subtracted TF footprinting for EGR1 by snATAC-seq neuron cluster (top) and by AD diagnosis (bottom), as in e. h, Left: Co-embedding UMAP colored by EGR1 motif variability (top) and EGR1 target gene score (bottom). Right: Violin plots of EGR1 motif variability (top) and EGR1 target gene score (bottom) in excitatory neuron clusters, split by diagnosis, as in f. i, Violin plots of SREBF1 motif variability in oligodendrocyte snATAC-seq clusters, as in f. j, Violin plots of SREBF1 gene expression in oligodendrocyte snRNA-seq clusters, as in i. For boxplots, box boundaries and line correspond to the interquartile range (IQR) and median respectively. Whiskers extend to the lowest or highest data points that are no further than 1.5 times the IQR from the box boundaries.

Source data

Extended Data Fig. 5 Schematics of analyses.

a, Schematic diagram linking cCREs to target genes and downstream analysis. First, we identify co-accessible chromatin peaks in each cell-type for control and late-stage AD. Second, we identify pairs of co-accessible peaks where one peak overlaps a gene promoter and correlate the expression of that gene with the chromatin accessibility of the other peak. Third, NMF is used to group gl-cCREs into functional modules. b, Schematic of construction of TF regulatory networks for each cell-type. c, Schematic representation of scWGCNA analysis, including iNMF integration with the Mathys et al. 2019 dataset, metacell aggregation, construction of co-expression networks, and downstream analysis of gene modules.

Extended Data Fig. 6 Pseudotime trajectory analysis to identify dysregulated TFs and gene expression in glia.

a, Line plot showing the RVAgene training loss at each epoch for oligodendrocyte (ODC), microglia (MG), and astrocyte (ASC) RVAE models. b, c, d, Heatmaps showing TF motif variability smoothed using loess regression and scaled to minimum and maximum values for TFs up- and down-regulated in AD as well as cell-type marker TFs along the oligodendrocyte trajectory (b), microglia trajectory (c), and astrocyte trajectory (d). TFs are ordered by trajectory rank (point in trajectory where of 75% maximum value is reached). e, f, g, Dot plot showing the enrichR combined score for the top enriched GO terms in oligodendrocyte (e), microglia (f), and astrocyte (g) t-DEGs.

Extended Data Fig. 7 Metacell aggregation and SREBP.

a, Heatmap showing the enrichment of cell-type marker genes in standard WGCNA modules constructed from our snRNA-seq data. b, Schematic showing generation of 30,218 metacells from the integrated transcriptomic dataset of 132,106 nuclei from our snRNA-seq and Mathys et al. c, d, e, Heatmap showing enrichment of oligodendrocyte (c), microglia (d), and astrocyte (e) scWGCNA modules constructed with 12 metacells, 25 metacells, 100 metacells, and 200 metacells in the scWGCNA modules constructed with 50 metacells, as shown in Fig. 7 and Supplementary Fisg. 15 and 16. f, SREBP protein-protein interaction (PPI) network. Green circle denotes proteins involved in ribosome processing and transcription pathway, cyan circle for mTOR pathway, and red circle for lipid processing pathway. g, Left: Representative immunohistochemistry images from postmortem human brain tissue for SREBP with nuclear counterstain. Right: Quantification of SREBP staining. n = 4 pathological controls, 3 late-stage AD. Data is represented as the mean of four equally sized regions per sample. Scale bar represents 100 μm. Linear mixed-effects model ** p < 0.01. Box boundaries and line correspond to the interquartile range (IQR) and median respectively. Whiskers extend to the lowest or highest data points that are no further than 1.5 times the IQR from the box boundaries.

Source data

Extended Data Fig. 8 iNMF integration of snRNA-seq with Mathys et al. snRNA-seq.

a, Schematic representation of iNMF integration of snRNA-seq with Mathys et al. snRNA-seq. UMAP plots are colored by cell-type assignments. b, Dot plot of iNMF metagene expression in each cell-type, split by dataset of origin. c, UMAP plots of the integrated dataset colored by selected iNMF metagenes. d, Dot plots showing the iNMF loading for the top 30 genes for the same metagenes in c.

Extended Data Fig. 9 scWGCNA in microglia and astrocytes.

a, Signed correlation of astrocyte modules to AD diagnosis. b, c, d, Co-expression plots for modules AM1 (b), AM2 (c), and AM5 (d). e, GO term enrichment of astrocyte modules. f, Heatmaps showing row-normalized Seurat module scores of astrocyte modules in snRNA-seq (left) and snATAC-seq (right) astrocyte clusters. g, Signed correlation of microglia co-expression modules with AD diagnosis. h, i, j, Co-expression plots for modules MM1 (h), MM2 (i), and MM4 (j). k, GO term enrichment of microglia modules. l, Heatmaps showing row-normalized Seurat module scores of microglia modules in snRNA-seq (left) and snATAC-seq (right) microglia clusters.

Extended Data Fig. 10 scWGCNA in neurons.

a, Signed correlation of excitatory neuron modules to AD diagnosis. b, c, d, e, Co-expression plots for modules EM1 (b), EM2 (c), EM5 (d), and EM7 (e). f, GO term enrichment of excitatory neuron modules. g, Heatmaps showing row-normalized Seurat module scores of excitatory neuron modules in snRNA-seq (left) and snATAC-seq (right) excitatory neuron clusters. h, Signed correlation of inhibitory neuron modules to AD diagnosis. i, j, k, l, m, n, Co-expression plots for modules IM1 (i), IM2 (j), IM3 (k), IM4 (l), IM5 (m), and IM6 (n). o, GO term enrichment of inhibitory neuron modules. p, Heatmaps showing row-normalized Seurat module scores of inhibitory neuron modules in snRNA-seq (left) and snATAC-seq (right) inhibitory neuron clusters.

Supplementary information

Supplementary Information

Supplementary results, discussion, methods and Figs. 1–12

Reporting Summary

Peer Review Information

Supplementary Tables 1–9

Supplementary Data 1

Supplementary Data 1

Supplementary Data 2

Supplementary Data 2

Supplementary Data 3

Supplementary Data 3

Supplementary Data 4

Supplementary Data 4

Supplementary Data 5

Supplementary Data 5

Supplementary Data 6

Supplementary Data 6

Supplementary Data 7

Supplementary Data 7

Source data

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morabito, S., Miyoshi, E., Michael, N. et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease. Nat Genet 53, 1143–1155 (2021). https://doi.org/10.1038/s41588-021-00894-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00894-z

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