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.

  • Letter
  • Published:

Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology

This article has been updated

Abstract

Senescent cells contribute to pathology and dysfunction in animal models1. Their sparse distribution and heterogenous phenotype have presented challenges to their detection in human tissues. We developed a senescence eigengene approach to identify these rare cells within large, diverse populations of postmortem human brain cells. Eigengenes are useful when no single gene reliably captures a phenotype, like senescence. They also help to reduce noise, which is important in large transcriptomic datasets where subtle signals from low-expressing genes can be lost. Each of our eigengenes detected 2% senescent cells from a population of 140,000 single nuclei derived from 76 postmortem human brains with various levels of Alzheimer’s disease (AD) pathology. More than 97% of the senescent cells were excitatory neurons and overlapped with neurons containing neurofibrillary tangle (NFT) tau pathology. Cyclin-dependent kinase inhibitor 2D (CDKN2D/p19) was predicted as the most significant contributor to the primary senescence eigengene. RNAscope and immunofluorescence confirmed its elevated expression in AD brain tissue. The p19-expressing neuron population had 1.8-fold larger nuclei and significantly more cells with lipofuscin than p19-negative neurons. These hallmark senescence phenotypes were further elevated in the presence of NFTs. Collectively, CDKN2D/p19-expressing neurons with NFTs represent a unique cellular population in human AD with a senescence-like phenotype. The eigengenes developed may be useful in future senescence profiling studies as they identified senescent cells accurately in snRNA-Seq datasets and predicted biomarkers for histological investigation.

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: The prominent senescent cell type in the dorsolateral prefrontal cortex were excitatory neurons.
Fig. 2: NFT eigengene expression correlated significantly with senescence expression.
Fig. 3: Senescent excitatory neurons contain NFTs, and NFT-bearing neurons are senescent.
Fig. 4: Upregulated CDKN2D and p19 deposition co-occur with tau neuropathology and morphological characteristics of cellular senescence in human AD.

Similar content being viewed by others

Data availability

The snRNA-Seq data analyzed in this study are available from https://www.synapse.org/ with synapse IDs: syn18485175 and syn21126462 for cohorts 1 and 2, respectively. Accessing these data requires submitting a Data Use Certificate through the AMP–AD website. Clinical data were available in the corresponding publications. The scRNA-Seq data from the embryonic cortex and the scRNA-Seq data from the entorhinal cortex are also available from the Gene Expression Omnibus54 with accession numbers GSE103723 and GSE138852.

Code availability

Our R scripts, which are available as Supplementary material, can be used to fully reproduce our results. Our code is also publicly available at https://bitbucket.org/habilzare/alzheimer/src/master/code/senescence/Shiva/.

Change history

  • 16 December 2021

    In the version of this article initially appearing online, the Supplementary Code file was missing, and has been included as of 16 December 2021.

References

  1. González-Gualda, E., Baker, A. G., Fruk, L. & Muñoz-Espín, D. A guide to assessing cellular senescence in vitro and in vivo. FEBS J. 288, 56–80 (2020).

    Article  PubMed  Google Scholar 

  2. Acosta, J. C. et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 15, 978–990 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Musi, N. et al. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 17, e12840 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Chow, H. M. et al. Age-related hyperinsulinemia leads to insulin resistance in neurons and cell-cycle-induced senescence. Nat. Neurosci. 22, 1806–1819 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Ogrodnik, M. et al. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab. 29, 1061–1077 e1068 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Riessland, M. et al. Loss of SATB1 induces p21-dependent cellular senescence in post-mitotic dopaminergic neurons. Cell Stem Cell 25, 514–530 e518 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chinta, S. J. et al. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell Rep. 22, 930–940 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bussian, T. J. et al. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562, 578–582 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhang, P. et al. Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat. Neurosci. 22, 719–728 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Justice, J. N. et al. Cellular senescence biomarker p16INK4a+ cell burden in thigh adipose is associated with poor physical function in older women. J. Gerontol. A Biol. Sci. Med. Sci. 73, 939–945 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Hickson, L. J. et al. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 47, 446–456 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Gillispie, G. J. Evidence of the cellular senescence stress response in mitotically active brain cells – implications for cancer and neurodegeneration.Life (Basel) 11, 153 (2021).

    CAS  Google Scholar 

  13. Sah, E. The cellular senescence stress response in post-mitotic brain cells – cell survival at the expense of tissue degeneration. Life (Basel) 11, 229 (2021).

    CAS  Google Scholar 

  14. Arendt, T., Rodel, L., Gartner, U. & Holzer, M. Expression of the cyclin-dependent kinase inhibitor p16 in Alzheimer’s disease. Neuroreport 7, 3047–3049 (1996).

    Article  CAS  PubMed  Google Scholar 

  15. Katsouri, L. et al. Ablation of reactive astrocytes exacerbates disease pathology in a model of Alzheimer’s disease. Glia 68, 1017–1030 (2020).

    Article  PubMed  Google Scholar 

  16. Hansen, D. V., Hanson, J. E. & Sheng, M. Microglia in Alzheimer’s disease. J. Cell Biol. 217, 459–472 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Oldham, M. C., Horvath, S. & Geschwind, D. H. Conservation and evolution of gene coexpression networks in human and chimpanzee brains. Proc. Natl Acad. Sci. USA 103, 17973–17978 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Foroushani, A. et al. Large-scale gene network analysis reveals the significance of extracellular matrix pathway and homeobox genes in acute myeloid leukemia: an introduction to the Pigengene package and its applications. BMC Med. Genomics 10, 16 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. Fan, X. et al. Spatial transcriptomic survey of human embryonic cerebral cortex by single-cell RNA-seq analysis. Cell Res. 28, 730–745 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bryant, A. G. et al. Cerebrovascular senescence is associated with tau pathology in Alzheimer’s disease. Front. Neurol. 11, 575953 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Bhat, R. et al. Astrocyte senescence as a component of Alzheimer’s disease. PLoS ONE 7, e45069 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Tchkonia, T. & Kirkland, J. L. Aging, cell senescence, and chronic disease: emerging therapeutic strategies. JAMA 320, 1319–1320 (2018).

    Article  PubMed  Google Scholar 

  25. Hooper, A. T. et al. Angiomodulin is a specific marker of vasculature and regulates vascular endothelial growth factor-A-dependent neoangiogenesis. Circ. Res. 105, 201–208 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Boraas, L. C. & Ahsan, T. Lack of vimentin impairs endothelial differentiation of embryonic stem cells. Sci. Rep. 6, 30814 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. Avelar, R. A. et al. A multidimensional systems biology analysis of cellular senescence in aging and disease. Genome Biol. 21, 91 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gene Ontology, C. The Gene Ontology resource: enriching a GOld mine. Nucleic Acids Res. 49, D325–D334 (2021).

    Article  Google Scholar 

  31. Kanehisa, M. & Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Orr, M. E., Sullivan, A. C. & Frost, B. A brief overview of tauopathy: causes, consequences, and therapeutic strategies. Trends Pharmacol. Sci. 38, 637–648 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fu, H. et al. A tau homeostasis signature is linked with the cellular and regional vulnerability of excitatory neurons to tau pathology. Nat. Neurosci. 22, 47–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Dunckley, T. et al. Gene expression correlates of neurofibrillary tangles in Alzheimer’s disease. Neurobiol. Aging 27, 1359–1371 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Otero-Garcia, T. et al. Single-soma transcriptomics of tangle-bearing neurons in Alzheimer’s disease reveals the signatures of tau-associated synaptic dysfunction. Preprint at bioRxiv https://doi.org/10.1101/2020.05.11.088591 (2020).

  36. Furcila, D., Dominguez-Alvaro, M., DeFelipe, J. & Alonso-Nanclares, L. Subregional density of neurons, neurofibrillary tangles and amyloid plaques in the hippocampus of patients with Alzheimer’s disease. Front. Neuroanat. 13, 99 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Neurohr, G. E. et al. Excessive cell growth causes cytoplasm dilution and contributes to senescence. Cell 176, 1083–1097.e1018 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Georgakopoulou, E. A. et al. Specific lipofuscin staining as a novel biomarker to detect replicative and stress-induced senescence. A method applicable in cryo-preserved and archival tissues. Aging (Albany NY) 5, 37–50 (2013).

    Article  CAS  Google Scholar 

  39. Frost, B., Bardai, F. H. & Feany, M. B. Lamin dysfunction mediates neurodegeneration in tauopathies. Curr. Biol. 26, 129–136 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Barascu, A. et al. Oxidative stress induces an ATM-independent senescence pathway through p38 MAPK-mediated lamin B1 accumulation. EMBO J. 31, 1080–1094 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hodes, R. J. & Buckholtz, N. Accelerating medicines partnership: Alzheimer’s disease (AMP–AD) knowledge portal aids Alzheimer’s drug discovery through open data sharing. Expert Opin. Ther. Targets 20, 389–391 (2016).

    Article  PubMed  Google Scholar 

  42. Bennett, D. A. et al. Religious orders study and rush memory and aging project. J. Alzheimers Dis. 64, S161–S189 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  43. R Core Team. R.: A language and environment for statistical computing https://www.R-project.org/ (2013).

  44. R Core Team. R: a language and environment for statistical computing. (R Foundation for Statistical Computing, 2017).

  45. Zainulabadeen, A., Yao, P. & Zare, H. Underexpression of specific interferon genes is associated with poor prognosis of melanoma. PLoS ONE 12, e0170025 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Agrahari, R. et al. Applications of Bayesian network models in predicting types of hematological malignancies. Sci. Rep. 8, 6951 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Samimi, H. DNA methylation analysis improves the prognostication of acute myeloid leukemia. eJHaem 2, 211–218 (2021).

    Article  PubMed  Google Scholar 

  48. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Altman, N. S. An introduction to kernel and nearest-neighbor nonparametric regression. Am. Stat. 46, 175–185 (1992).

    Google Scholar 

  51. Blondel, V. D., Guillaume, J.-L., Lambiotte, R. & Lefebvre, E. Fast unfolding of communities in large networks. J. Stat. Mech. 2008, P10008 (2008).

    Article  Google Scholar 

  52. McInnes, L., Healy, J. & Melville, J. Umap: Uniform manifold approximation and projection for dimension reduction. Preprint at arXiv (2018).

  53. Merritt, C. R. et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol. 38, 586–599 (2020).

    Article  CAS  PubMed  Google Scholar 

  54. Edgar, R., Domrachev, M. & Lash, A. E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work is supported by NIH/NIA (R01AG068293, R01AG057896, U01AG046170, RF1AG057440, R01AG057907, K99AG061259; P30AG062421; RF1AG051485, R21AG059176, and RF1AG059082 and T32AG021890), Cure Alzheimer’s Fund, New Vision Research Charleston Conference for Alzheimer’s Disease, and Veterans Affairs (IK2BX003804). We obtained ROSMAP data from the AD Knowledge Portal (https://adknowledgeportal.synapse.org). Study data were provided by the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago. Data generation was supported by National Institute on Aging (NIA) and grants RF1AG57473, P30AG10161, R01AG15819, R01AG17917, U01G46152, U01AG61356 and RF1AG059082. Additional phenotypic ROSMAP data can be requested at https://www.radc.rush.edu. We acknowledge the Texas Advanced Computing Center (TACC) at the University of Texas at Austin for providing high-performance computing resources: http://www.tacc.utexas.edu. We acknowledge the Biggs Institute Brain Bank and Massachusetts ADRC for providing postmortem human tissue for analyses. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

All authors edited and approved the final manuscript. In addition, S.K.D. developed code, designed and performed in silico experiments, performed all bioinformatic and statistical analyses, prepared figures and drafted the manuscript; J.W. provided tissue from the Biggs Brain Bank, performed p19 and AT8 IHC validation and wrote IHC methods; E.S. performed immunofluorescence validation (p19, AT8 and lamin B1), performed confocal microscopy and wrote the corresponding methods; E.B. performed blinded analyses of IHC and immunofluorescence and wrote the corresponding methods; F.A. performed immunofluorescence (p19, AT8 and Map2) validation and wrote corresponding methods; B.F. provided oversight to F.A. and provided Lamin b1 expertise; B.W. performed RNAScope; R.E.B. requested and acquired brain tissue from MGH ADRC, performed confocal microscopy, provided oversight to B.W., generated RNAscope figures and wrote RNAscope methods, RNAscope data interpretation; T.C.O. developed methods to analyze lamin b1 expression, performed blinded lamin b1 analyses and wrote lamin B1 analyses methods; Y.Z. provided access and expertise on cohort 2 snRNA-Seq data; P.S.A. provided access and expertise on cohort 2 snRNA-Seq data; M.C. provided access and expertise on cohort 2 snRNA-Seq data; P.H.S. provided bioinformatics consultation and validation; P.X. performed cell clustering analyses and wrote corresponding methods; M.W. performed cell clustering analyses and wrote the corresponding methods; B.Z. provided oversight to P.X. and provided bioinformatic consultation; H.Z. provided oversight to S.K.D. (bioinformatic experimental design and development), acquired access to all datasets, designed and confirmed bioinformatic data analyses and interpreted results; M.E.O. conceived the project, acquired funding, provided supervision (E.S., E.B. and T.C.O.), analyzed and interpreted histology data, interpreted bioinformatic data, and drafted manuscript and submitted the manuscript.

Corresponding authors

Correspondence to Habil Zare or Miranda E. Orr.

Ethics declarations

Competing interests

Patent applicant: Wake Forest University Health Sciences. Name of inventor: Miranda Orr. Application number: 63/199,927 & 63/261,630. Status of application: Pending. The specific aspect of manuscript covered in patent application: data from this manuscript was used to file a patent, ‘Biosignature and therapeutic approach for neuronal senescence’.

Additional information

Peer review information Nature Aging thanks Markus Riessland and the other, anonymous, reviewer(s) 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 figures and tables

Extended Data Fig. 1 Prominent senescent cell types in prefrontal cortex of the embryonic control.

Cell types and counts represented in the senescent cell population discovered in (A) CSP, (B) SIP and (C) SRP. The cutoff and statistical test definitions are the same as in Fig. 1. Cell populations: astrocytes [Ast], blood cells [Blood], Cajal-Retzius cells [Cajal], endothelial cells [Endo], excitatory neurons [Ext], immune cells [Immune], inhibitory neuron [Inh], microglia [Micro], neural stem cells [NSC], and oligodendrocyte precursor cells [Oligo] were classified in the original publication.

Extended Data Fig. 2 Prominent senescent cell types in the dorsal lateral prefrontal cortex in Cohort 2.

Cell types and counts represented in the senescent cell population discovered in (A) CSP, (B) SIP and (C) SRP with n = 57,857. The cutoff, statistical test and abbreviations definitions are the same as in Fig. 1.

Extended Data Fig. 3 Overlap between senescent and NFT neurons.

Each vertical bar represents the number of neurons in Cohort 1 that express the eigengenes marked by green circles below the bar. Each row at the bottom corresponds to an eigengene, and the number of neurons expressing that eigengene is shown in the right end on each row. The probability distributions of multi-set intersections have been calculated and the significance was tested using a hypergeometric test. The scale bar at top right shows the level of significance for each intersection. The largest p-value is −232 in log10 scale, which corresponds to the intersection between SRP and CSP expressing cells.

Extended Data Fig. 4 Prominent senescent cell types using CellAge, GO and KEGG gene lists in Cohort 1.

Cell types and counts represented in the senescent cell population discovered in (A) CellAge, (B) GO and (C) KEGG. The cutoff, statistical test and abbreviations definitions are the same as in Fig. 1.

Extended Data Fig. 5 Overlap between senescent cell populations.

Each vertical bar represents the number of senescent cells in Cohort 1 that express the senescence eigengenes, marked by green circles below the bar. Each row at the bottom corresponds to a senescence eigengene, and the number of senescent cells expressing that eigengene is shown at the end of each row. The probability distributions of multi-set intersections have been calculated and the significance was tested using a hypergeometric test. The scale bar at top right shows the level of significance for each intersection. The largest p-value is-260 in log10 scale corresponding to the intersection of SRP and CSP.

Extended Data Fig. 6 Excitatory neurons are the prominent senescent cell types based on CDKN2D in (A) Cohort 1 and (B) Cohort 2.

Cell types and counts represented in the senescent cell population using only CDKN2D. The cutoff, statistical tests and abbreviations definitions are the same as in Fig. 1.

Extended Data Fig. 7 RNAscope reveals higher CDKN2D expression in postmortem brains from cases with AD than age-matched control brains.

A. CDKN2D negative and positive control probe signal. B. CDKN2D RNAscope on three separate AD cases (n = 3) compared to a representative age-matched non-demented control (n = 3) (refer to Supplementary Table 5 for case characteristics. Scale bar 50 μm.

Extended Data Fig. 8 CDKN2D RNAscope co-localized with neuronal marker, HuD.

Postmortem AD tissue was processed for RNAscope with CDKN2D (green) and co-labeled for total nuclei (DAPI, gray) and neurons (HuD, cyan)/ Merged image display strong overlap between CDKN2D and neurons, but not other cell types (that is, blue and green co-localization with infrequent green co-localization in nuclei without HuD staining). Scale bar 10 μm. Representative images from postmortem human brains (n = 3 control and n = 3 AD cases).

Supplementary information

Supplementary Information Map of Extended Data, Supplementary Figs. 1–14 and Supplementary Tables 4–5.

Reporting Summary

Supplementary Table 1

This file contains Supplemental Tables 1-3 combined into a single excel workbook file with multiple Tabs. Supplementary Table 1 contains the gene lists used to create each of the eigengene; Supplementary Table 2 (multiple tabs) contains eigengene expression data from every cell analyzed across datasets. Supplementary Table 3 (multiple tabs) contains data describing the overlap between cells expressing the senescence eigengenes and those expressing the NFT eigengenes for both cohorts (1 and 2) using both NFT eigengenes (Dunckley and Garcia).

Supplementary Code

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dehkordi, S.K., Walker, J., Sah, E. et al. Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology. Nat Aging 1, 1107–1116 (2021). https://doi.org/10.1038/s43587-021-00142-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00142-3

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