Skip to main content

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

  • Resource
  • Published:

Large-scale plasma proteomic analysis identifies proteins and pathways associated with dementia risk

This article has been updated

Abstract

The plasma proteomic changes that precede the onset of dementia could yield insights into disease biology and highlight new biomarkers and avenues for intervention. We quantified 4,877 plasma proteins in nondemented older adults in the Atherosclerosis Risk in Communities cohort and performed a proteome-wide association study of dementia risk over five years (n = 4,110; 428 incident cases). Thirty-eight proteins were associated with incident dementia after Bonferroni correction. Of these, 16 were also associated with late-life dementia risk when measured in plasma collected nearly 20 years earlier, during mid-life. Two-sample Mendelian randomization causally implicated two dementia-associated proteins (SVEP1 and angiostatin) in Alzheimer’s disease. SVEP1, an immunologically relevant cellular adhesion protein, was found to be part of larger dementia-associated protein networks, and circulating levels were associated with atrophy in brain regions vulnerable to Alzheimer’s pathology. Pathway analyses for the broader set of dementia-associated proteins implicated immune, lipid, metabolic signaling and hemostasis pathways in dementia pathogenesis.

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: Schematic overview of the study design.
Fig. 2: Proteome-wide associations with incident dementia, mid-life replication and external replication.
Fig. 3: Association of dementia-associated proteins with neuroimaging markers and gene coexpression analyses.
Fig. 4: Biological pathways and upstream regulators implicated in dementia risk.
Fig. 5: Protein networks identified among dementia-associated proteins.

Similar content being viewed by others

Data availability

All data generated in this study are either included in this article (and its Supplementary Information), available upon reasonable request or are available in an online public database. Consistent with a prespecified policy for access of ARIC and AGES data, requests may be submitted to the ARIC and AGES steering committees for review. Requests for clinical or proteomic data from individual investigators will be reviewed to ensure that data can be shared without compromising patient confidentiality or breaching intellectual property restrictions. Reasonable requests will be considered and promptly processed. Participant-level demographic, clinical and proteomic data may be partially restricted based on previously obtained participant consent. Data sharing restrictions may also be applied to ensure consistency with confidentiality or privacy laws and considerations. For information on how to access available data and study protocols, see www2.cscc.unc.edu/aric/. Data from the AGES-Reykjavik study used in this study are available through collaboration (AGES_data_request@hjarta.is) under a data usage agreement with the Icelandic Heart Association. Tissue-specific gene expression data are available at https://www.gtexportal.org/home/. Gene coexpression analyses were conducted using data available at http://www.explainbio.com. Brain gene expression data were derived from the Brain eQTL Almanac (http://www.braineac.org/) and the Functional Mapping and Annotation of Genome-Wide Association Studies105 platform (https://fuma.ctglab.nl/). eQTL gene enrichment was performed using data from the Molecular Signatures database (http://www.broadinstitute.org/msigdb) and the NHGRI-EBI Catalog of Published GWAS (https://www.ebi.ac.uk/gwas/). Functional enrichment of protein networks was conducted using the g:Profiler web tool (https://biit.cs.ut.ee/gprofiler/gost).

Code availability

All software used in this study are publicly available: R v.3.6.2 (https://www.r-project.org/); Stata, v.14 (https://www.stata.com/stata14/); IPA (https://digitalinsights.qiagen.com/products-overview/discovery-insights-portfolio/analysis-and-visualization/qiagen-ipa/); ExplainBio (http://www.explainbio.com/); and GraphPad Prism v.8.4.3. (https://www.graphpad.com/scientific-software/prism/). The code used in this study can be made available from the corresponding author upon reasonable request.

Change history

  • 04 July 2022

    In the version of this article initially published, the Supplementary Tables 1–31 file was missing and has now been restored to the online version of the article.

References

  1. Nakamura, A. et al. High-performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature 554, 249–254 (2018).

    Article  CAS  Google Scholar 

  2. Mattsson, N., Cullen, N. C., Andreasson, U., Zetterberg, H. & Blennow, K. Association between longitudinal plasma neurofilament light and neurodegeneration in patients with Alzheimer disease. JAMA Neurol. 76, 791–799 (2019).

    Article  Google Scholar 

  3. Palmqvist, S. et al. Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in Alzheimer’s disease. EMBO Mol. Med. https://doi.org/10.15252/emmm.201911170 (2019).

  4. Kim, K. et al. Clinically accurate diagnosis of Alzheimer’s disease via multiplexed sensing of core biomarkers in human plasma. Nat. Commun. 11, 119 (2020).

    Article  CAS  Google Scholar 

  5. Soares, H. D. et al. Plasma biomarkers associated with the apolipoprotein E genotype and Alzheimer disease. Arch. Neurol. 69, 1310 (2012).

    Article  Google Scholar 

  6. Doecke, J. D. et al. Blood-based protein biomarkers for diagnosis of Alzheimer disease. Arch. Neurol. 69, 1318 (2012).

    Article  Google Scholar 

  7. Hye, A. et al. Proteome-based plasma biomarkers for Alzheimer’s disease. Brain 129, 3042–3050 (2006).

    Article  CAS  Google Scholar 

  8. Cutler, P. et al. Proteomic identification and early validation of complement 1 inhibitor and pigment epithelium-derived factor: two novel biomarkers of Alzheimer’s disease in human plasma. Proteomics. Clin. Appl. 2, 467–477 (2008).

    Article  CAS  Google Scholar 

  9. Choi, J., Malakowsky, C. A., Talent, J. M., Conrad, C. C. & Gracy, R. W. Identification of oxidized plasma proteins in Alzheimer’s disease. Biochem. Biophys. Res. Commun. 293, 1566–1570 (2002).

    Article  CAS  Google Scholar 

  10. Henkel, A. W. et al. Multidimensional plasma protein separation technique for identification of potential Alzheimer’s disease plasma biomarkers: a pilot study. J. Neural Transm. 119, 779–788 (2012).

    Article  CAS  Google Scholar 

  11. Ray, S. et al. Classification and prediction of clinical Alzheimer’s diagnosis based on plasma signaling proteins. Nat. Med. 13, 1359–1362 (2007).

    Article  CAS  Google Scholar 

  12. Ijsselstijn, L. et al. Serum levels of pregnancy zone protein are elevated in presymptomatic Alzheimer’s disease. J. Proteome Res. 10, 4902–4910 (2011).

    Article  CAS  Google Scholar 

  13. O’Bryant, S. E. et al. A serum protein-based algorithm for the detection of Alzheimer disease. Arch. Neurol. 67, 1077–1081 (2010).

    Article  Google Scholar 

  14. Sattlecker, M. et al. Alzheimer’s disease biomarker discovery using SOMAscan multiplexed protein technology. Alzheimers Dement. 10, 724–734 (2014).

    Article  Google Scholar 

  15. Kiddle, S. J. et al. Candidate blood proteome markers of Alzheimer’s disease onset and progression: a systematic review and replication study. J. Alzheimers Dis. 38, 515–531 (2014).

    Article  CAS  Google Scholar 

  16. Sattlecker, M. et al. Longitudinal protein changes in blood plasma associated with the rate of cognitive decline in Alzheimer’s disease. J. Alzheimers Dis. 49, 1105–1114 (2016).

    Article  CAS  Google Scholar 

  17. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  Google Scholar 

  18. Sharma, K. et al. Cell type- and brain region-resolved mouse brain proteome. Nat. Neurosci. 18, 1819–1831 (2015).

    Article  CAS  Google Scholar 

  19. Seyfried, N. T. et al. A multi-network approach identifies protein-specific co-expression in asymptomatic and symptomatic Alzheimer’s disease. Cell Syst. 4, 60–72 (2017).

    Article  CAS  Google Scholar 

  20. Ganz, P. et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. JAMA 315, 2532–2541 (2016).

    Article  CAS  Google Scholar 

  21. Gold, L. et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE 5, e15004 (2010).

    Article  CAS  Google Scholar 

  22. 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  Google Scholar 

  23. Price, A. L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    Article  CAS  Google Scholar 

  24. Shadlen, M. F. et al. Education, cognitive test scores, and black-white differences in dementia risk. J. Am. Geriatr. Soc. 54, 898–905 (2006).

    Article  Google Scholar 

  25. Lehallier, B. et al. Undulating changes in human plasma proteome profiles across the lifespan. Nat. Med. 25, 1843–1850 (2019).

    Article  CAS  Google Scholar 

  26. Harris, T. B. et al. Age, gene/environment susceptibility-Reykjavik study: multidisciplinary applied phenomics. Am. J. Epidemiol. 165, 1076–1087 (2007).

    Article  Google Scholar 

  27. Emilsson, V. et al. Co-regulatory networks of human serum proteins link genetics to disease. Science 361, 769–773 (2018).

    Article  CAS  Google Scholar 

  28. Sun, B. B. et al. Genomic atlas of the human plasma proteome. Nature 558, 73–79 (2018).

    Article  CAS  Google Scholar 

  29. Cissé, M. & Checler, F. Eph receptors: new players in Alzheimer’s disease pathogenesis. Neurobiol. Dis. 73, 137–149 (2015).

    Article  Google Scholar 

  30. Shi, L. et al. A decade of blood biomarkers for Alzheimer’s disease research: an evolving field, improving study designs, and the challenge of replication. J. Alzheimers Dis. 62, 1181–1198 (2018).

    Article  Google Scholar 

  31. Palmqvist, S. et al. Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 324, 772–781 (2020).

    Article  CAS  Google Scholar 

  32. Ghosh, S. et al. Sustained interleukin-1 overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J. Neurosci. 33, 5053–5064 (2013).

    Article  CAS  Google Scholar 

  33. Paolicelli, R. C. et al. TDP-43 depletion in microglia promotes amyloid clearance but also induces synapse loss. Neuron 95, 297–308 (2017).

    Article  CAS  Google Scholar 

  34. Varma, S. Network model of normal gene expression predicts gene perturbation fold changes. In Proc. RECOMB/ISCB Conference on Regulatory and Systems Genomics with DREAM Challenges 78–79 (2015).

  35. Savli, H., Szendröi, A., Romics, I. & Nagy, B. Gene network and canonical pathway analysis in prostate cancer: a microarray study. Exp. Mol. Med. 40, 176–185 (2008).

    Article  CAS  Google Scholar 

  36. Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290 (2017).

    Article  CAS  Google Scholar 

  37. 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  Google Scholar 

  38. 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  Google Scholar 

  39. Jones, L. et al. Convergent genetic and expression data implicate immunity in Alzheimer’s disease. Alzheimers Dement. 11, 658–671 (2015).

    Article  Google Scholar 

  40. Felsky, D. et al. Neuropathological correlates and genetic architecture of microglial activation in elderly human brain. Nat. Commun. https://doi.org/10.1038/s41467-018-08279-3 (2019).

  41. Walker, K. A. et al. The association of mid-to late-life systemic inflammation with white matter structure in older adults: the Atherosclerosis Risk in Communities study. Neurobiol. Aging 68, 26–33 (2018).

    Article  CAS  Google Scholar 

  42. Steeland, S. et al. Counteracting the effects of TNF receptor‐1 has therapeutic potential in Alzheimer’s disease. EMBO Mol. Med. https://doi.org/10.15252/emmm.201708300 (2018).

  43. Krasemann, S. et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47, 566–581 (2017).

    Article  CAS  Google Scholar 

  44. Neumann, H. & Daly, M. J. Variant TREM2 as risk factor for Alzheimer’s disease. N. Engl. J. Med. 368, 182–184 (2013).

    Article  CAS  Google Scholar 

  45. Stegle, O., Parts, L., Piipari, M., Winn, J. & Durbin, R. Using probabilistic estimation of expression residuals (PEER) to obtain increased power and interpretability of gene expression analyses. Nat. Protoc. 7, 500–507 (2012).

    Article  CAS  Google Scholar 

  46. Parts, L., Stegle, O., Winn, J. & Durbin, R. Joint genetic analysis of gene expression data with inferred cellular phenotypes. PLoS Genet. 7, e1001276 (2011).

    Article  CAS  Google Scholar 

  47. Trimbuch, T. & Rosenmund, C. Should I stop or should I go? The role of complexin in neurotransmitter release. Nat. Rev. Neurosci. 17, 118–125 (2016).

    Article  CAS  Google Scholar 

  48. An, S. J., Grabner, C. P. & Zenisek, D. Real-time visualization of complexin during single exocytic events. Nat. Neurosci. 13, 577–583 (2010).

    Article  CAS  Google Scholar 

  49. Tannenberg, R. K., Scott, H. L., Tannenberg, A. E. G. & Dodd, P. R. Selective loss of synaptic proteins in Alzheimer’s disease: evidence for an increased severity with APOE ε4. Neurochem. Int. 49, 631–639 (2006).

    Article  CAS  Google Scholar 

  50. Nakada, T. A., Russell, J. A., Boyd, J. H., Thair, S. A. & Walley, K. R. Identification of a nonsynonymous polymorphism in the SVEP1 gene associated with altered clinical outcomes in septic shock. Crit. Care Med. 43, 101–108 (2015).

    Article  CAS  Google Scholar 

  51. Siddiqui, S., Gurung, R. L., Liu, S., Seet, E. C. P. & Lim, S. C. Genetic polymorphisms and cytokine profile of different ethnicities in septic shock patients, and their association with mortality. Indian J. Crit. Care Med. 23, 135–138 (2019).

    Article  CAS  Google Scholar 

  52. Sato-Nishiuchi, R. et al. Polydom/SVEP1 is a ligand for integrin α9β1. J. Biol. Chem. 287, 25615–25630 (2012).

    Article  CAS  Google Scholar 

  53. Schwanzer-Pfeiffer, D., Roßmanith, E., Schildberger, A. & Falkenhagen, D. Characterization of SVEP1, KIAA, and SRPX2 in an in vitro cell culture model of endotoxemia. Cell. Immunol. 263, 65–70 (2010).

    Article  CAS  Google Scholar 

  54. Richter, V. et al. Circulating vascular cell adhesion molecules VCAM-1, ICAM-1, and E-selectin in dependence on aging. Gerontology 49, 293–300 (2003).

    Article  CAS  Google Scholar 

  55. 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).

    Article  CAS  Google Scholar 

  56. Janelidze, S. et al. CSF biomarkers of neuroinflammation and cerebrovascular dysfunction in early Alzheimer disease. Neurology 91, e867–e877 (2018).

    Article  CAS  Google Scholar 

  57. K Ryu, J., P Little, J., Klegeris, A., Jantaratnotai, N. & G McLarnon, J. Actions of the anti-angiogenic compound angiostatin in an animal model of Alzheimer’s disease. Curr. Alzheimers Res. 10, 252–260 (2013).

    Article  Google Scholar 

  58. Jahanshad, N. et al. Genome-wide scan of healthy human connectome discovers SPON1 gene variant influencing dementia severity. Proc. Natl Acad. Sci. USA 110, 4768–4773 (2013).

    Article  CAS  Google Scholar 

  59. Bossù, P. et al. Interleukin-18 produced by peripheral blood cells is increased in Alzheimer’s disease and correlates with cognitive impairment. Brain. Behav. Immun. 22, 487–492 (2008).

    Article  Google Scholar 

  60. Li, X., Long, J., He, T., Belshaw, R. & Scott, J. Integrated genomic approaches identify major pathways and upstream regulators in late-onset Alzheimer’s disease. Sci. Rep. 5, 12393 (2015).

    Article  Google Scholar 

  61. Painter, M. M. et al. TREM2 in CNS homeostasis and neurodegenerative disease. Mol. Neurodegener. https://doi.org/10.1186/s13024-015-0040-9 (2015).

  62. The ARIC Investigators. The Atherosclerosis Risk in Communities (ARIC) study: design and objectives. Am. J. Epidemiol. 129, 687–702 (1989).

    Article  Google Scholar 

  63. Tin, A. et al. Reproducibility and variability of protein analytes measured using a multiplexed modified aptamer assay. J. Appl. Lab. Med. 4, 30–39 (2019).

    Article  Google Scholar 

  64. Kim, C. H. et al. Stability and reproducibility of proteomic profiles measured with an aptamer-based platform. Sci. Rep. 8, 8382 (2018).

    Article  Google Scholar 

  65. Candia, J. et al. Assessment of variability in the SOMAscan assay. Sci. Rep. 7, 14248 (2017).

    Article  Google Scholar 

  66. Williams, S. A. et al. Plasma protein patterns as comprehensive indicators of health. Nat. Med. 25, 1851–1857 (2019).

    Article  CAS  Google Scholar 

  67. Muntner, P., He, J., Astor, B. C., Folsom, A. R. & Coresh, J. Traditional and nontraditional risk factors predict coronary heart disease in chronic kidney disease: results from the Atherosclerosis Risk in Communities study. J. Am. Soc. Nephrol. 16, 529–538 (2005).

    Article  Google Scholar 

  68. Knopman, D. S. et al. Mild cognitive impairment and dementia prevalence: the atherosclerosis risk in communities neurocognitive study. Alzheimers Dement. Diagnosis Assess. Dis. Monit. 2, 1–11 (2016).

    Google Scholar 

  69. Hayden, K. M. et al. Cognitive decline in the elderly: an analysis of population heterogeneity. Age Ageing 40, 684–689 (2011).

    Article  Google Scholar 

  70. Wilson, R. S., Li, Y., Bienias, L. & Bennett, D. A. Cognitive decline in old age: separating retest effects from the effects of growing older. Psychol. Aging 21, 774–789 (2006).

    Article  Google Scholar 

  71. McKhann, G. M. et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 7, 263–269 (2011).

    Article  Google Scholar 

  72. American Psychiatric Association. DSM-5: Diagnostic and Statistical Manual of Mental Disorders (American Psychiatric Association, 2013).

  73. Carpenter, C. R., Despain, B., Keeling, T. N., Shah, M. & Rothenberger, M. The six-item screener and AD8 for the detection of cognitive impairment in geriatric emergency department patients. Ann. Emerg. Med. 57, 653–661 (2011).

    Article  Google Scholar 

  74. Galvin, J. E., Roe, C. M., Xiong, C. & Morris, J. C. Validity and reliability of the AD8 informant interview in dementia. Neurology 67, 1942–1948 (2006).

    Article  Google Scholar 

  75. Walker, K. A. et al. Association of midlife to late-life blood pressure patterns with incident dementia. JAMA 322, 535–545 (2019).

    Article  Google Scholar 

  76. Gottesman, R. F. et al. Associations between midlife vascular risk factors and 25-year incident dementia in the Atherosclerosis Risk in Communities (ARIC) cohort. JAMA Neurol. 388, 797–805 (2017).

    Google Scholar 

  77. Diagnostic and Statistical Manual of Mental Disorders: DSM-IV-TR (American Psychiatric Association, 2000).

  78. Knopman, D. S. et al. Vascular imaging abnormalities and cognition: Mediation by cortical volume in nondemented individuals: atherosclerosis risk in communities-neurocognitive study. Stroke 46, 433–440 (2015).

    Article  Google Scholar 

  79. Fischl, B. et al. Whole brain segmentation: automated labeling of neuroanatomical structures in the human brain. Neuron 33, 341–355 (2002).

    Article  CAS  Google Scholar 

  80. Dickerson, B. C. et al. Alzheimer-signature MRI biomarker predicts AD dementia in cognitively normal adults. Neurology 76, 1395–1402 (2011).

    Article  CAS  Google Scholar 

  81. Raz, L. et al. Thrombogenic microvesicles and white matter hyperintensities in postmenopausal women. Neurology 80, 911–918 (2013).

    Article  CAS  Google Scholar 

  82. Gottesman, R. F. et al. The ARIC-PET amyloid imaging study: brain amyloid differences by age, race, sex, and APOE. Neurology 87, 473–480 (2016).

    Article  Google Scholar 

  83. Smith, G. D. & Ebrahim, S. ‘Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease? Int. J. Epidemiol. 32, 1–22 (2003).

    Article  Google Scholar 

  84. Burgess, S., Butterworth, A. & Thompson, S. G. Mendelian randomization analysis with multiple genetic variants using summarized data. Genet. Epidemiol. 37, 658–665 (2013).

    Article  Google Scholar 

  85. Zheng, J. et al. Recent developments in Mendelian randomization studies. Curr. Epidemiol. Reports 4, 330–345 (2017).

    Article  Google Scholar 

  86. Machiela, M. J. & Chanock, S. J. LDlink: a web-based application for exploring population-specific haplotype structure and linking correlated alleles of possible functional variants. Bioinformatics 31, 3555–3557 (2015).

    Article  CAS  Google Scholar 

  87. Bowden, J., Davey Smith, G. & Burgess, S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int. J. Epidemiol. 44, 512–525 (2015).

    Article  Google Scholar 

  88. Bowden, J., Davey Smith, G., Haycock, P. C. & Burgess, S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet. Epidemiol. 40, 304–314 (2016).

    Article  Google Scholar 

  89. Burgess, S., Foley, C. N., Allara, E., Staley, J. R. & Howson, J. M. M. A robust and efficient method for Mendelian randomization with hundreds of genetic variants. Nat. Commun. 11, 376 (2020).

    Article  CAS  Google Scholar 

  90. Staiger, D. & Stock, J. H. Instrumental Variables Regression with Weak Instruments (National Bureau of Economic Research Inc., 1994).

  91. Burgess, S. & Thompson, S. G. Interpreting findings from Mendelian randomization using the MR-Egger method. Eur. J. Epidemiol. 32, 377–389 (2017).

    Article  Google Scholar 

  92. Verbanck, M., Chen, C. Y., Neale, B. & Do, R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat. Genet. 50, 693–698 (2018).

    Article  CAS  Google Scholar 

  93. Cochran, W. The combination of estimates from different experiments. Biometrics 10, 101–129 (1954).

    Article  Google Scholar 

  94. Hemani, G. et al. The MR-Base platform supports systematic causal inference across the human phenome. eLife https://doi.org/10.7554/eLife.34408 (2018).

  95. Burgess, S., Scott, R. A., Timpson, N. J., Davey Smith, G. & Thompson, S. G. Using published data in Mendelian randomization: a blueprint for efficient identification of causal risk factors. Eur. J. Epidemiol. 30, 543–552 (2015).

    Article  Google Scholar 

  96. Bowden, J. et al. Improving the visualization, interpretation and analysis of two-sample summary data Mendelian randomization via the Radial plot and Radial regression. Int. J. Epidemiol. 47, 1264–1278 (2018).

    Article  Google Scholar 

  97. Watanabe, K., Taskesen, E., Van Bochoven, A. & Posthuma, D. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun. https://doi.org/10.1038/s41467-017-01261-5 (2017).

  98. Kircher, M. et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 46, 310–315 (2014).

    Article  CAS  Google Scholar 

  99. Ramasamy, A. et al. Genetic variability in the regulation of gene expression in ten regions of the human brain. Nat. Neurosci. 17, 1418–1428 (2014).

    Article  CAS  Google Scholar 

  100. GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).

  101. Lonsdale, J. et al. The genotype-tissue expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

    Article  CAS  Google Scholar 

  102. Varma, V. R. et al. α-2 macroglobulin in Alzheimer’s disease: a marker of neuronal injury through the RCAN1 pathway. Mol. Psychiatry 22, 13–23 (2017).

    Article  CAS  Google Scholar 

  103. Hage, C. et al. Inflammatory biomarkers predict heart failure severity and prognosis in patients with heart failure with preserved ejection fraction: a holistic proteomic approach. Circ. Cardiovasc. Genet. https://doi.org/10.1161/CIRCGENETICS.116.001633 (2017).

  104. Helleman, J., Smid, M., Jansen, M. P. H. M., van der Burg, M. E. L. & Berns, E. M. J. J. Pathway analysis of gene lists associated with platinum-based chemotherapy resistance in ovarian cancer: the big picture. Gynecol. Oncol. 117, 170–176 (2010).

    Article  CAS  Google Scholar 

  105. Krämer, A., Green, J., Pollard, J. & Tugendreich, S. Causal analysis approaches in ingenuity pathway analysis. Bioinformatics 30, 523–530 (2014).

    Article  Google Scholar 

  106. Raudvere, U. et al. G:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  Google Scholar 

  107. Gene, T. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25 (2000).

    Article  Google Scholar 

  108. The Gene Ontology, C. et al. The gene ontology resource: 20 years and still GOing strong. Nucleic Acids Res. https://doi.org/10.17863/CAM.36439 (2019).

  109. Kanehisa, M., Sato, Y., Kawashima, M., Furumichi, M. & Tanabe, M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 44, D457–D462 (2016).

    Article  CAS  Google Scholar 

  110. Bohler, A. et al. Reactome from a WikiPathways perspective. PLoS Comput. Biol. https://doi.org/10.1371/journal.pcbi.1004941 (2016).

  111. Stegle, O., Parts, L., Durbin, R. & Winn, J. A Bayesian framework to account for complex non-genetic factors in gene expression levels greatly increases power in eQTL studies. PLoS Comput. Biol. 6, 1–11 (2010).

    Article  Google Scholar 

  112. Seddighi, S. et al. SPARCL1 accelerates symptom onset in Alzheimer’s disease and influences brain structure and function during aging. J. Alzheimers Dis. 61, 401–414 (2018).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the staff and participants of the ARIC study for their important contributions. We also thank B. Chen for her valuable assistance with aspects of the manuscript. The ARIC study has been funded in whole or in part with Federal funds from the National Heart, Lung and Blood Institute (NHLBI), NIH, Department of Health and Human Services (contract nos. HHSN268201700001I, HHSN268201700002I, HHSN268201700003I, HHSN268201700004I, HHSN268201700005I, R01HL087641 and R01HL086694); National Human Genome Research Institute contract U01HG004402; and NIH contract HHSN268200625226C. Neurocognitive data are collected by U01 2U01HL096812, 2U01HL096814, 2U01HL096899, 2U01HL096902, 2U01HL096917 from the NIH (NHLBI, National Institute of Neurological Disorders and Stroke (NINDS), NIA and National Institute on Deafness and Other Communication Disorders (NIDCD)) and with previous brain MRI examinations funded by R01-HL70825 from the NHLBI. The ARIC-PET study is funded by the NIA (R01AG040282). Infrastructure was partly supported by grant no. UL1RR025005, a component of the NIH and NIH Roadmap for Medical Research. This study was also supported by contracts K23 AG064122 (to K.A.W.), K24 AG052573 (to R.F.G.) and U01-AG052409 (to M.F.) from NIA; and R01-HL134320 (to C.M.B.) from NHLBI. Avid Radiopharmaceuticals provided the florbetapir isotope for the study but had no role in the study design or interpretation of results. The AGES-Reykjavik study was supported by the Icelandic Heart Association, the NIA (N01-AG-12100 and HHSN271201200022C), the Intramural Program at the NIA, the Althingi (the Icelandic Parliament), the Icelandic Centre for Research grant 141101-051 and the Novartis Institute for Biomedical Research. SomaLogic provided assays as an in-kind contribution in a data flex change collaboration agreement. This research was supported in part by the Intramural Research Program of the NIH, NIA. Funders had no control over the publication.

Author information

Authors and Affiliations

Authors

Contributions

K.A.W., S.L.Z., R.F.G., T.H.M., E.B., C.M.B. and J. Coresh conceptualized and designed the study. M.G., N.D., R.C.H., C.M.B., V.E., L.J.L., L.L.J., V.G. and J. Coresh contributed to data acquisition. K.A.W., J. Chen, J.Z., M.F., Y.Y., L.Z., A.W. and E.F.G. analyzed data. K.A.W., M.F., Y.Y., M.G., A.T., P.G., N.C., R.F.G. and J. Coresh contributed to interpretation of data. K.A.W., M.F., Y.Y., M.G., A.T., R.C.H., A.W., K.J.S., P.G., L.J.L., R.F.G., C.M.B. and J. Coresh drafted and provided substantial revision of the manuscript.

Corresponding authors

Correspondence to Keenan A. Walker or Josef Coresh.

Ethics declarations

Competing interests

R.C.H. has received grants and consulting fees from Denka Seiken outside the scope of the current research study. A.W. received fees from the Analysis Group as a consultant outside the scope of the current research study. P.G. is a member of the SomaLogic Medical Advisory board, for which he receives no remuneration of any kind. L.L.J. is an employee and stockholder of Novartis. R.F.G. received fees from the American Academy of Neurology for her role as an Associate Editor for the journal Neurology. Proteomic assays in ARIC were conducted free of charge as part of a data exchange agreement with SomaLogic. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks Sebastian Palmqvist 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

Extended Data Fig. 1 P-values for dementia-associated proteins in mid-life replication and AGES-Reykjavik external replication analyses.

a, P-values (two-sided) for the mid-life replication analysis (index visit 1993-1995, ages 49-73) of the 38 dementia-associated proteins identified in older adults (y-axis) plotted against P-values for each dementia-associated protein derived from the primary analysis (index visit 2011-2013) on the x-axis. The horizontal dotted red line represents the Bonferroni-corrected threshold for statistical significance in the mid-life replication analysis (0.05/38; P < 0.0013). The vertical dotted black line represents the Bonferroni-corrected threshold for statistical significance in the primary analysis (0.05/4,877; P < 1.03×10-5). b, P-values (two-sided) for the AGES-Reykjavik (index visit 2002-2006) replication of proteins that were significantly associated with dementia risk in both the primary and the mid-life replication analysis (y-axis) plotted against P-values for each dementia-associated protein derived from the primary analysis (index visit 2011-2013) on the x-axis. The horizontal dotted red line represents the Bonferroni-corrected threshold for statistical significance in the AGES-Reykjavik replication analysis (0.05/13; P < 0.0038). The vertical dotted black line represents the Bonferroni-corrected threshold for statistical significance in the primary analysis (0.05/4,877; P < 1.03×10-5).

Extended Data Fig. 2 Prediction of incident dementia using proteins, demographic and clinical variables, and their combination measured at late-life baseline (2011-2013).

Elastic net machine learning with Cox proportional hazards regression was used to select the best combination of proteins from the top 50 proteins in each model. This table shows results from 10-fold cross validated analyses. Two-sided P-values were calculated for the C statistic comparisons. No corrections for multiple comparisons were performed. a Protein combination defined using elastic net machine learning algorithm. b Includes age, sex, race-center, education, and APOEε4. c Includes body mass index, diabetes, hypertension, smoking status, and eGFR-creatinine. Abbreviations: C stat. Δ, change in C statistic with the addition of elastic net proteins.

Extended Data Fig. 3 Gene expression in whole blood, brain, heart, kidney, spleen, and adipose tissue of genes coding for dementia-associated proteins.

Using gene expression data available from postmortem samples in the GTEx database, this heatmap shows the expression of genes coding for dementia-associated proteins (in transcripts per million) in whole blood, select brain regions, and other selected tissue. Hierarchical cluster analysis was used to group dementia-associated proteins based on gene expression across multiple tissues.

Extended Data Fig. 4 Functional profiling of protein Network 1 identified among the set of dementia-associated proteins.

Protein networks were assembled based on evidence of known gene/molecule interactions in the Ingenuity Knowledge Base. The g:Profiler toolkit106 was used to analyze the proteins in each dementia-associated protein network for functional enrichment. We defined the biological pathways/processes associated with each protein set using the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and WikiPathways databases. Images were generated using the g:Profiler web tool: https://biit.cs.ut.ee/gprofiler/. Abbreviations: GO:BP, gene ontology biological process; GO:MF, gene ontology molecular function.

Extended Data Fig. 5 Functional profiling of protein Network 2 and Network 3 identified among the set of dementia-associated proteins.

Protein networks were assembled based on evidence of known gene/molecule interactions in the Ingenuity Knowledge Base. The g:Profiler toolkit106 was used to analyze the proteins in each dementia-associated protein network for functional enrichment. We defined the biological pathways/processes associated with each protein set using the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and WikiPathways databases. Images were generated using the g:Profiler web tool: https://biit.cs.ut.ee/gprofiler/. Abbreviations: GO:BP, gene ontology biological process; GO:MF, gene ontology molecular function.

Extended Data Fig. 6 Functional profiling of protein Network 4, Network 5, and Network 6 identified among the set of dementia-associated proteins.

Protein networks were assembled based on evidence of known gene/molecule interactions in the Ingenuity Knowledge Base. The g:Profiler toolkit106 was used to analyze the proteins in each dementia-associated protein network for functional enrichment. We defined the biological pathways/processes associated with each protein set using the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and WikiPathways databases. Images were generated using the g:Profiler web tool: https://biit.cs.ut.ee/gprofiler/. Abbreviations: GO:BP, gene ontology biological process; GO:MF, gene ontology molecular function.

Extended Data Fig. 7 Functional profiling of protein Network 7, Network 8, and Network 10 identified among the set of dementia-associated proteins.

Protein networks were assembled based on evidence of known gene/molecule interactions in the Ingenuity Knowledge Base. The g:Profiler toolkit106 was used to analyze the proteins in each dementia-associated protein network for functional enrichment. We defined the biological pathways/processes associated with each protein set using the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and WikiPathways databases. Images were generated using the g:Profiler web tool: https://biit.cs.ut.ee/gprofiler/. No enriched biological pathways/processes were found for Network 9. Abbreviations: GO:BP, gene ontology biological process; GO:MF, gene ontology molecular function.

Extended Data Fig. 8 Functional profiling of protein Network 11 and Network 12 identified among the set of dementia-associated proteins.

Protein networks were assembled based on evidence of known gene/molecule interactions in the Ingenuity Knowledge Base. The g:Profiler toolkit106 was used to analyze the proteins in each dementia-associated protein network for functional enrichment. We defined the biological pathways/processes associated with each protein set using the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and WikiPathways databases. Images were generated using the g:Profiler web tool: https://biit.cs.ut.ee/gprofiler/. Abbreviations: GO:BP, gene ontology biological process; GO:MF, gene ontology molecular function.

Extended Data Fig. 9 PEER factors associated with incident dementia after visit 5.

a, Volcano plot showing the hazard ratio (x-axis) and two-sided P-value (y-axis) for the association of 110 PEER factors with incident dementia. PEER factors above the horizontal dotted line were significantly associated with incident dementia after Bonferroni correction (0.05/110; P < 0.00045). P-values for dementia-associated PEER factors were 1.52E-06, 2.24E-05, and 2.00E-04 for PEER factors 9, 88, and 3, respectively. b, Spearman correlations between dementia-associated protein level and each PEER factors associated with dementia risk.

Supplementary information

Supplementary Information

Supplementary Methods and Figs. 1 and 2.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–31

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Walker, K.A., Chen, J., Zhang, J. et al. Large-scale plasma proteomic analysis identifies proteins and pathways associated with dementia risk. Nat Aging 1, 473–489 (2021). https://doi.org/10.1038/s43587-021-00064-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00064-0

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research