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:

CSF proteome profiling across the Alzheimer’s disease spectrum reflects the multifactorial nature of the disease and identifies specific biomarker panels

Abstract

Development of disease-modifying therapies against Alzheimer’s disease (AD) requires biomarkers reflecting the diverse pathological pathways specific for AD. We measured 665 proteins in 797 cerebrospinal fluid (CSF) samples from patients with mild cognitive impairment with abnormal amyloid (MCI(Aβ+): n = 50), AD-dementia (n = 230), non-AD dementias (n = 322) and cognitively unimpaired controls (n = 195) using proximity ligation-based immunoassays. Here we identified >100 CSF proteins dysregulated in MCI(Aβ+) or AD compared to controls or non-AD dementias. Proteins dysregulated in MCI(Aβ+) were primarily related to protein catabolism, energy metabolism and oxidative stress, whereas those specifically dysregulated in AD dementia were related to cell remodeling, vascular function and immune system. Classification modeling unveiled biomarker panels discriminating clinical groups with high accuracies (area under the curve (AUC): 0.85–0.99), which were translated into custom multiplex assays and validated in external and independent cohorts (AUC: 0.8–0.99). Overall, this study provides novel pathophysiological leads delineating the multifactorial nature of AD and potential biomarker tools for diagnostic settings or clinical trials.

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: Study overview and differential abundance of CSF proteins across groups.
Fig. 2: Differentially regulated CSF proteins change along AD spectrum and reflect different biological processes.
Fig. 3: CSF biomarker panels for early and specific diagnosis of AD.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available at https://www.synapse.org/PRIDE_AD. MS CSF results from previous studies have been provided by co-authors (J.J.L., N.T.S., E.B.D. and A.I.L.)13,25, and all data are available at https://www.synapse.org/consensus and https://www.synapse.org/#!Synapse:syn20821165/wiki/596086.

Code availability

The code that supports the findings of this study is available from the authors on reasonable request. All models were built using publicly available packages and functions in R.

References

  1. Jellinger, K. A. Neuropathology of dementia disorders.J. Alzheimers Dis. Parkinsonism 4, 135 (2014).

    Article  Google Scholar 

  2. Beach, T. G., Monsell, S. E., Phillips, L. E. & Kukull, W. Accuracy of the clinical diagnosis of Alzheimer disease at National Institute on Aging Alzheimer Disease Centers, 2005–2010. J. Neuropathol. Exp. Neurol. 71, 266–273 (2012).

    Article  PubMed  Google Scholar 

  3. Gauthier, S. et al. Why has therapy development for dementia failed in the last two decades? Alzheimers Dement. 12, 60–64 (2016).

    Article  PubMed  Google Scholar 

  4. Jansen, I. E. et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 12, 43 (2019).

    Google Scholar 

  5. van der Kant, R., Goldstein, L. S. B. & Ossenkoppele, R. Amyloid-β-independent regulators of tau pathology in Alzheimer disease. Nat. Rev. Neurosci. 21, 21–35 (2020).

    Article  PubMed  Google Scholar 

  6. Park, S. A., Han, S. M. & Kim, C. E. New fluid biomarkers tracking non-amyloid-β and non-tau pathology in Alzheimer’s disease. Exp. Mol. Med. 52, 556–568 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Schoonenboom, N. S. M. et al. Cerebrospinal fluid markers for differential dementia diagnosis in a large memory clinic cohort. Neurology 78, 47–54 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Jack, C. R. et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 14, 535–562 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Robinson, J. L. et al. Neurodegenerative disease concomitant proteinopathies are prevalent, age-related and APOE4-associated. Brain 141, 2181–2193 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ewers, M. et al. CSF biomarkers for the differential diagnosis of Alzheimer’s disease: A large-scale international multicenter study. Alzheimers Dement. 11, 1306–1315 (2015).

    Article  PubMed  Google Scholar 

  11. van Bokhoven, P. et al. The Alzheimer’s disease drug development landscape. Alzheimers Res. Ther. 13, 186 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bader, J. M. et al. Proteome profiling in cerebrospinal fluid reveals novel biomarkers of Alzheimer’s disease. Mol. Syst. Biol. 16, e9356 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Higginbotham, L. et al. Integrated proteomics reveals brain-based cerebrospinal fluid biomarkers in asymptomatic and symptomatic Alzheimer’s disease. Sci. Adv. 6, eeaz9360 (2020).

    Article  Google Scholar 

  14. Assarsson, E. et al. Homogenous 96-Plex PEA immunoassay exhibiting high sensitivity, specificity, and excellent scalability. PLoS ONE 9, e95192 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Enroth, S. et al. High throughput proteomics identifies a high-accuracy 11 plasma protein biomarker signature for ovarian cancer. Commun. Biol. 2, 221 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Fang, M. et al. The ER UDPase ENTPD5 promotes protein N-glycosylation, the Warburg effect, and proliferation in the PTEN pathway. Cell 143, 711–724 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Clements, C. M., Mcnally, R. S., Conti, B. J., Mak, T. W. & Ting, J. P. protein, stabilizes the antioxidant transcriptional master regulator Nrf2. Proc. Natl Acad. Sci. USA 103, 15091–15096 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Maes, M. B. et al. Kinetic investigation of human dipeptidyl peptidase II (DPPII)-mediated hydrolysis of dipeptide derivatives and its identification as quiescent cell proline dipeptidase (QPP)/dipeptidyl peptidase 7 (DPP7). Biochem. J 386, 315–324 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Soisson, S. M. et al. Structural definition and substrate specificity of the S28 protease family: The crystal structure of human prolylcarboxypeptidase. BMC Struct. Biol. 10, 1–8 (2010).

    Article  Google Scholar 

  20. Patiño-Lopez, G. et al. Human class-I restricted T cell associated molecule is highly expressed in the cerebellum and is a marker for activated NKT and CD8+ T lymphocytes. J. Neuroimmunol. 171, 145–155 (2006).

    Article  PubMed  Google Scholar 

  21. Derkinderen, P. et al. Tyrosine 394 is phosphorylated in Alzheimer’s paired helical filament tau and in fetal tau with c-Abl as the candidate tyrosine kinase. J. Neurosci. 25, 6584–6593 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Beroun, A. et al. MMPs in learning and memory and neuropsychiatric disorders. Cell. Mol. Life Sci. 76, 3207–3228 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Erickson-Viitanen, S., Ruggieri, S., Natalini, P. & Horecker, B. L. Thymosin beta 10, a new analog of thymosin beta 4 in mammalian tissues. Arch. Biochem. Biophys. 225, 407–413 (1983).

    Article  CAS  PubMed  Google Scholar 

  24. Baietti, M. F. et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 14, 677–685 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Johnson, E. C. B. et al. Large-scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation. Nat. Med. 26, 769–780 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Isgrò, M. A., Bottoni, P. & Scatena, R. Neuron-Specific enolase as a biomarker: Biochemical and clinical aspects. Adv. Exp. Med. Biol. 867, 125–143 (2015).

    Article  PubMed  Google Scholar 

  27. Pollio, G. et al. Increased expression of the oligopeptidase THOP1 is a neuroprotective response to Aβ toxicity. Neurobiol. Dis. https://doi.org/10.1016/j.nbd.2008.04.004 (2008).

    Article  PubMed  Google Scholar 

  28. Dayoub, H. et al. Dimethylarginine dimethylaminohydrolase regulates nitric oxide synthesis: genetic and physiological evidence. Circulation 108, 3042–3047 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Kannan, L. et al. R-spondin3 prevents mesenteric ischemia/reperfusion-induced tissue damage by tightening endothelium and preventing vascular leakage. Proc. Natl Acad. Sci. USA 110, 14348–14353 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wesenhagen, K. E. J., Teunissen, C. E., Visser, P. J. & Tijms, B. M. Cerebrospinal fluid proteomics and biological heterogeneity in Alzheimer’s disease: A literature review. Crit. Rev. Clin. Lab. Sci. 57, 86–98 (2019).

    Article  PubMed  Google Scholar 

  31. Boström, G. et al. Different inflammatory signatures in Alzheimer’s disease and frontotemporal dementia cerebrospinal fluid. J. Alzheimers Dis. 81, 629–640 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Konijnenberg, E. et al. APOE ϵ4 genotype-dependent cerebrospinal fluid proteomic signatures in Alzheimer’s disease. Alzheimers Res. Ther. 12, 65 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Whelan, C. D. et al. Multiplex proteomics identifies novel CSF and plasma biomarkers of early Alzheimer’s disease. Acta Neuropathol. Commun. 7, 169 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Duits, F. H. et al. Matrix metalloproteinases in Alzheimer’s disease and concurrent cerebral microbleeds. J. Alzheimers Dis. 48, 711–720 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Dayon, L. et al. Alzheimer disease pathology and the cerebrospinal fluid proteome. Alzheimers Res. Ther. 10, 66 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Suhre, K., McCarthy, M. I. & Schwenk, J. M. Genetics meets proteomics: perspectives for large population-based studies. Nat. Rev. Genet. 22, 19–37 (2021).

    Article  CAS  PubMed  Google Scholar 

  37. Dammer, E. B. et al. Multi-platform proteomic analysis of Alzheimer’s disease cerebrospinal fluid and plasma reveals network biomarkers associated with proteostasis and the matrisome. Preprint at bioRxiv https://doi.org/10.1101/2022.06.20.494087 (2022).

  38. Wildsmith, K. R. et al. Identification of longitudinally dynamic biomarkers in Alzheimer’s disease cerebrospinal fluid by targeted proteomics. Mol. Neurodegener. 9, 22 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Begcevic, I. et al. Brain-related proteins as potential CSF biomarkers of Alzheimer’s disease: A targeted mass spectrometry approach. J. Proteomics 182, 12–20 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Bai, B. et al. Deep multilayer brain proteomics identifies molecular networks in alzheimer’s disease progression. Neuron 105, 975–991 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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  PubMed  Google Scholar 

  42. Gao, S., Casey, A. E., Sargeant, T. J. & Mäkinen, V.-P. Genetic variation within endolysosomal system is associated with late-onset Alzheimer’s disease. Brain 141, 2711–2720 (2018).

    Article  PubMed  Google Scholar 

  43. Butterfield, D. A. & Halliwell, B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat. Rev. Neurosci. 20, 148–160 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Butkiewicz, M. 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  PubMed  PubMed Central  Google Scholar 

  45. Lepelletier, F. X., Mann, D. M. A., Robinson, A. C., Pinteaux, E. & Boutin, H. Early changes in extracellular matrix in Alzheimer’s disease. Neuropathol. Appl. Neurobiol. 43, 167–182 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Wang, H. et al. Regulation of beta-amyloid production in neurons by astrocyte-derived cholesterol. Proc. Natl Acad. Sci. USA 118, e2102191118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Philibert, K. D., Marr, R. A., Norstrom, E. M. & Glucksman, M. J. Identification and characterization of Aβ peptide interactors in Alzheimer’s disease by structural approaches. Front. Aging Neurosci. 6, 1–9 (2014).

    Article  Google Scholar 

  48. Lonskaya, I., Hebron, M. L., Desforges, N. M., Franjie, A. & Moussa, C. E.-H. Tyrosine kinase inhibition increases functional parkin‐Beclin‐1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol. Med. 5, 1247–1262 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Siddiqui, T. J. et al. An LRRTM4-HSPG complex mediates excitatory synapse development on dentate gyrus granule cells. Neuron 79, 680–695 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Gong, C.-X., Liu, F. & Iqbal, K. Perspective multifactorial hypothesis and multi-targets for Alzheimer’s disease. J. Alzheimers Dis. 64, 107–117 (2018).

    Article  Google Scholar 

  51. Johnstone, D., Milward, E. A., Berretta, R. & Moscato, P. Multivariate protein signatures of pre-clinical Alzheimer’s disease in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) Plasma Proteome Dataset. PLoS ONE 7, e34341 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kim, Y. et al. Targeted proteomics identifies liquid-biopsy signatures for extracapsular prostate cancer. Nat. Commun. 7, 11906 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Teunissen, C. E., Otto, M., Engelborghs, S., Herukka, S.-K. & Lehmann, S. White paper by the Society for CSF Analysis and Clinical Neurochemistry: Overcoming barriers in biomarker development and clinical translation. Alzheimers Res. Ther. 12, 23 (2018).

    Google Scholar 

  54. Teunissen, C. E. et al. Novel diagnostic cerebrospinal fluid protein biomarkers for pathologic subtypes of frontotemporal dementia identified by proteomics. Alzheimers Dement. 9, 86–94 (2016).

    Google Scholar 

  55. Teunissen, C. E. et al. Blood-based biomarkers for Alzheimer’s disease: Towards clinical implementation. Lancet Neurol. 21, 66–77 (2021).

    Article  PubMed  Google Scholar 

  56. van der Flier, W. M. & Scheltens, P. Amsterdam Dementia Cohort: Performing research to optimize care. J. Alzheimers Dis. 62, 1091–1111 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Lleó, A. et al. A 2-step cerebrospinal algorithm for the selection of frontotemporal lobar degeneration subtypes. JAMA Neurol. 75, 738–745 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Del Campo, M. et al. Recommendations to standardize preanalytical confounding factors in Alzheimer’s and Parkinson’s disease cerebrospinal fluid biomarkers: An update. Biomark Med. 6, 419–430 (2012).

    Article  PubMed  Google Scholar 

  59. Albert, M. S. et al. The diagnosis of mild cognitive impairment 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, 270–279 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Dubois, B. et al. Research criteria for the diagnosis of Alzheimer’s disease: Revising the NINCDS-ADRDA criteria. Lancet Neurol. 6, 734–746 (2007).

    Article  PubMed  Google Scholar 

  61. McKeith, I. G. et al. Diagnosis and management of dementia with Lewy bodies: Third report of the DLB Consortium. Neurology 65, 1863–1872 (2005).

    Article  CAS  PubMed  Google Scholar 

  62. McKeith, I. G. et al. Diagnosis and management of dementia with Lewy bodies: Fourth consensus report of the DLB Consortium. Neurology 89, 88–100 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Rascovsky, K. et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain 134, 2456–2477 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Armstrong, M. J. et al. Criteria for the diagnosis of corticobasal degeneration. Neurology 80, 496–503 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Litvan, I. et al. Clinical research criteria for the diagnosis of progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome): report of the NINDS-SPSP international workshop. Neurology 47, 1–9 (1996).

    Article  CAS  PubMed  Google Scholar 

  66. Shaw, L. M. et al. Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann. Neurol. 65, 403–413 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Duits, F. H. et al. The cerebrospinal fluid ‘alzheimer profile’: Easily said, but what does it mean? Alzheimers Dement. 10, 713–723 (2014).

    Article  PubMed  Google Scholar 

  68. Alcolea, D. et al. The Sant Pau Initiative on Neurodegeneration (SPIN) cohort: A data set for biomarker discovery and validation in neurodegenerative disorders. Alzheimers Dement. 5, 597–609 (2019).

    Article  Google Scholar 

  69. Tijms, B. M. et al. Unbiased approach to counteract upward drift in cerebrospinal fluid amyloid-β 1–42 analysis results. Clin. Chem. 64, 576–585 (2018).

    Article  CAS  PubMed  Google Scholar 

  70. Jessen, F. et al. A conceptual framework for research on subjective cognitive decline in preclinical Alzheimer’s disease. Alzheimers Dement. 10, 844–852 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Leeuw, F. A. et al. Blood‐based metabolic signatures in Alzheimer’s disease. Alzheimers Dement. 8, 196–207 (2017).

    Google Scholar 

  72. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: A practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).

    Google Scholar 

  73. Friedman, J., Hastie, T. & Tibshirani, R. Regularization paths for generalized linear models via coordinate descent. J. Stat. Softw. 33, 1–22 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research is part of the neurodegeneration research program of Amsterdam Neuroscience. This study was supported by Alzheimer Nederland (WE.03-2018-05, M.d.C. and C.E.T.) and Selfridges Group Foundation (NR170065, M.d.C. and C.E.T.). M.d.C. and G.H. are supported by the attraction talent fellowship of Comunidad de Madrid (2018-T2/BMD-11885) and San Pablo CEU University. D.A. acknowledges support from Institute of Health Carlos III (PI18/00435, INT19/00016) and the Department of Health Generalitat de Catalunya PERIS program (SLT006/17/125). Collection of patients samples and data from Penn University (A.C.-P. and D.J.I.) was supported by different funding sources, including National Institute on Aging NINDS R01-NS109260-01A1, P01-AG066597, P30-AG072979 (formerly P30-AG10124) and U19-AG062418-03 (formerly NINDSP50-NS053488-09). Alzheimer Center Amsterdam is supported by Stichting Alzheimer Nederland and Stichting VUmc fonds. The chair (W.M.v.d.F.) is supported by the Pasman stichting. The clinical database structure was developed with funding from Stichting Dioraphte. Research programs of W.M.v.d.F. have been funded by ZonMW, NWO, EU-FP7, EU-JPND, Alzheimer Nederland, Hersenstichting CardioVascular Onderzoek Nederland, Health~Holland, Topsector Life Sciences & Health, stichting Dioraphte, Gieskes-Strijbisfonds, stichting Equilibrio, Edwin Bouw fonds, Pasman stichting, stichting Alzheimer & Neuropsychiatrie Foundation, Philips, Biogen MA, Novartis-NL, Life-MI, AVID, Roche BV, Fujifilm and Combinostics. W.M.v.d.F. holds the Pasman chair. W.M.v.d.F. is a recipient of ABOARD, which is a public-private partnership receiving funding from ZonMW (73305095007) and Health~Holland, Topsector Life Sciences & Health (PPP-allowance; #LSHM20106). All funding is paid to her institution. Research of C.E.T. is supported by the European Commission (Marie Curie International Training Network, grant agreement No. 860197 (MIRIADE)), Innovative Medicines Initiatives 3TR (Horizon 2020, grant 831434), EPND (IMI 2 Joint Undertaking (JU) under grant agreement 101034344) and JPND (bPRIDE), National MS Society (Progressive MS alliance) and Health Holland, the Dutch Research Council (ZonMW), Alzheimer Drug Discovery Foundation, The Selfridges Group Foundation, Alzheimer Netherlands and Alzheimer Association. C.E.T. is a recipient of ABOARD, which is a public-private partnership receiving funding from ZonMW (3305095007) and Health~Holland, Topsector Life Sciences & Health (PPP allowance; LSHM20106). ABOARD also receives funding from Edwin Bouw Fonds and Gieskes-Strijbisfonds. 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

M.d.C. and C.T conceived the study. M.d.C., C.F.W.P., L.V. and M.v.N. performed the statistical analysis. L.V., A.C.-P., D.J.I., W.T.H., L.H.M., J.v.S., D.A., A.L., A.W.L, Y.A.L.P., P.J.V., B.T.M. and W.M.v.d.F. recruited participants and collected clinical data and samples. M.d.C. and Y.S.H. arranged and prepared samples for proteomics analysis. J.J.L., N.T.S., E.B.D. and A.I.L. provided the proteomic data (both Olink and MS) from previous published studies13,25. M.d.C. and C.E.T. drafted the manuscript. All authors contributed to revision and editing of the manuscript.

Corresponding author

Correspondence to Marta del Campo.

Ethics declarations

Competing interests

M.d.C. has been an invited speaker at Eisai. L.V. received a grant for the CORAL consortium by Olink. B.M.T. and P.J.V. are inventors on a patent (WO2020197399A1; owned by Stichting VUmc). D.J.I. is a Scientific Advisory Board Member for Denali Therapeutics. D.A. participated in advisory boards from Fujirebio-Europe and Roche Diagnostics and received speaker honoraria from Fujirebio-Europe, Roche Diagnostics, Nutricia, Krka Farmacéutica S.L., Zambon S.A.U. and Esteve Pharmaceuticals S.A. D.A. declares a filed patent application (WO2019175379 A1 Markers of synaptopathy in neurodegenerative disease). W.M.v.d.F. has performed contract research for Biogen MA and Boehringer Ingelheim. W.M.v.d.F. has been an invited speaker at Boehringer Ingelheim, Biogen MA, Danone, Eisai, WebMD Neurology (Medscape) and Springer Healthcare. W.M.v.d.F. is consultant to Oxford Health Policy Forum CIC, Roche and Biogen MA. W.M.v.d.F. participated in advisory boards of Biogen MA and Roche. All funding is paid to her institution. W.M.v.d.F. is member of the steering committee of PAVE and Think Brain Health. W.M.v.d.F. was associate editor of Alzheimer’s Research & Therapy in 2020/2021. W.M.v.d.F. is associate editor at Brain. C.E.T. has a collaboration contract with ADx Neurosciences, Quanterix and Eli Lilly and performed contract research or received grants from AC-Immune, Axon Neurosciences, Bioconnect, Bioorchestra, Brainstorm Therapeutics, Celgene, EIP Pharma, Eisai, Grifols, Novo Nordisk, PeopleBio, Roche, Toyama and Vivoryon. She serves on editorial boards of Medidact Neurologie/Springer, Alzheimer’s Research & Therapy and Neurology: Neuroimmunology & Neuroinflammation and is editor of a Neuromethods book (Springer). She had speaker contracts for Roche, Grifols and Novo Nordisk. The other authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Niklas Mattsson-Carlgren and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Protein replicates measured through different PEA panels showed comparable differences.

a, Beta coefficients (log2 fold change) of the protein replicates from the comparisons between MCI(Aβ+) or AD and controls (yellow and red respectively) and AD and non-AD dementias (blue). Stars represent the corresponding significant q-value (* q < 0.05, ** q < 0.01, ***q < 0.001) in the discrimination between groups. b, Scatter plots show the correlation of the NPX values (protein abundance) between the protein replicates that showed significant differences across the groups of interest in the complete cohort (n = 797). Regression line and 95% confident intervals are depicted. Insert indicate the spearman correlation coefficient. CON, cognitively unimpaired controls; MCI(Aβ+), mild cognitive impairment with Aβ pathological changes; AD, Alzheimer’s disease; non-AD dem: non-AD dementias.

Extended Data Fig. 2 Sensitivity analysis AD versus control.

a, Volcano plot represent the proteins differentially regulated in the validation cohort (AD = 18, CON = 44)23,24 after two-sided nested F-test analysis with no adjustments for multiple comparisons. The beta coefficients (log2 fold change) are plotted versus p values (-log10-transformed). Scatter plots show the correlation between the p-values or beta coefficients obtained in the discovery and validation cohorts. Insert indicate the spearman correlation coefficient using all proteins. Proteins showing concordance between discovery and validation (that is, consistency regarding the proteins that are dysregulated and nondysregulated or show the same direction of changes between the groups of interest in both cohorts) are colored in green and the corresponding concordance % is indicated. b, Venn diagram depicts the number and % of overlapping proteins that are significantly dysregulated (p < 0.05) between AD and controls in the discovery and validation cohorts after applying two-sided nested F-test analysis with no adjustments for multiple comparisons. The name of the overlapping proteins is specified in extended data Table 1. AD, Alzheimer’s disease; CON, cognitively unimpaired controls.

Extended Data Fig. 3 Biological processes enriched with randomly selected markers.

Bar graphs depicting the biological pathways enriched when a random selection of markers was performed. The number of proteins randomly selected is the same as those in the unique protein subsets identified along AD stages (Fig. 2b). Functional enrichment was performed using Metascape selecting GO Biological Processes as ontology source. Terms with a p-value < 0.01, a minimum count of 3, and an enrichment factor > 1.5 were collected and grouped into clusters based on their membership similarities. p-values were calculated based on the accumulative hypergeometric distribution. Kappa scores are used as the similarity metric when performing hierarchical clustering on the enriched terms, and subtrees with a similarity of > 0.3 are considered a cluster. The most statistically significant term within a cluster is chosen to represent the cluster. The corresponding GO number and biological process is defined in the right side. Stronger colors represent higher significant enrichment. Vertical line represent the significant threshold (unadjusted p < 0.01).

Extended Data Fig. 4 Mean intensity values of the protein markers included in the CSF diagnostic protein panels for early and specific diagnosis of AD.

Violins represent the abundance (log2 NPX) of the CSF proteins that combined can accurately discriminate MCI(Aβ+) (n = 50) or AD (n = 230, orange) from controls (n = 195) and AD from other types of dementias (n = 322, blue). Horizontal black and dash lines indicate median and interquartile range of the protein abundance. aITGB2 and ABL1 are also markers within the AD versus non-AD dementia panel.*q < 0.05, **q < 0.01, ***q < 0.001. n.s: non-significant. MCI(Aβ+), mild cognitive impairment with Aβ pathological changes; AD, Alzheimer’s disease; non-ADdem: non-AD dementias; CON, cognitively unimpaired controls.

Extended Data Fig. 5 Correlations between each biomarker panel composite score and AD CSF biomarkers and cognitive function.

Correlation matrix heatmaps represent the Spearman’s correlation coefficient in-between the biomarker panel composite score (PC) of each of the identified CSF panels, the classical AD CSF biomarkers (Aβ42, tTau, pTau181 and tTau/Aβ42 ratio) and MMSE score in the corresponding groups for which the specific panel was built. Positive correlations are depicted in red; negative correlations are depicted in blue. Correlation coefficients are indicated. AD, Alzheimer’s disease; non-AD dem: non-AD dementias; CON, cognitively unimpaired controls.

Extended Data Fig. 6 Additional sensitivity analysis for the CSF biomarker panels discriminating AD from controls or AD from non-AD dementias.

a, Receiver operating characteristic (ROC) curves showed that the performance of the CSF AD-diagnostic to discriminate AD from controls was similar between APOE4 + (AD = 133; CON = 44) and APOE4- (AD = 88; CON = 125) patients. b, ROC curve depicting the performance of the CSF AD-differential diagnostic discriminating AD from non-AD dementias in the subset of patients in which non-AD dementias was confirmed (AD = 230; cNon-AD dem = 101). Black line is the mean area under the curve (AUC) over all resamplings (1000 repeats of 5-fold cross-validation, gray lines). Inserts outline corresponding AUC and 95% CI. AD, Alzheimer’s disease; CON, cognitively unimpaired controls; cNon-Ad dem: confirmed non-AD dementias.

Extended Data Fig. 7 Performance of CSF biomarker panels discriminating other groups of interest.

Receiver operating characteristic (ROC) curves depicting the performance of the different panels (rows, Fig. 3, Table 2) discriminating other groups of interest (columns). Black line is the mean area under the curve (AUC) over all resamplings (1000 repeats of 5-fold cross-validation, gray lines). Inserts outline corresponding AUC and 95% CI. MCI(Aβ+), mild cognitive impairment with Aβ pathological changes; AD, Alzheimer’s disease; non-AD dem: non-AD dementias; CON, cognitively unimpaired controls.

Extended Data Fig. 8 CSF biomarker panel for MCI(Aβ+) was not fully replicated in the validation cohort using custom PEA assays.

a, Receiver operating characteristic (ROC) curve depicting the performance of CSF biomarker panel discriminating MCI(Aβ+) from controls (10 CSF proteins: ENTPD5, CRTAM, PARK7, DPP7, CLEC5A, CYR61, KYNU, PCSK9, DDAH1 and PRCP) in the discovery (a, CON = 195, MCI(Aβ+) = 50). Black line is the mean area under the curve (AUC) over all resamplings (1000 repeats of 5-fold cross-validation, gray lines). Inserts outline corresponding AUC and 95% CI. b) ROC curve depicting the performance of CSF MCI(Aβ+)-diagnostic panel in the validation cohort (CON = 110 and MCI(Aβ+) = 101) using the custom assays. CRTAM assay could not be developed due to technical difficulties and thus was not included in the analysis of the validation cohort. c) Scatter plots show the correlation between the beta-coefficients obtained in the discovery phase to those obtained with the custom assays in validation step 2. Insert indicate the spearman correlation coefficient.*ABL1 and ITGB2 are proteins that are present in both AD CSF panels. CLEC5A is present in both the AD and the MCI(Aβ+) diagnostic panel. MCI(Aβ+), mild cognitive impairment with Aβ pathological changes; AD, Alzheimer’s disease; non-AD dem: non-AD dementias; CON, cognitively unimpaired controls.

Supplementary information

Source data

Source Data Table 1

Table outlining the statistics of the CSF proteins differentially regulated across the different diagnostic groups

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

del Campo, M., Peeters, C.F.W., Johnson, E.C.B. et al. CSF proteome profiling across the Alzheimer’s disease spectrum reflects the multifactorial nature of the disease and identifies specific biomarker panels. Nat Aging 2, 1040–1053 (2022). https://doi.org/10.1038/s43587-022-00300-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-022-00300-1

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