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:

Quantitative phosphoproteomics uncovers dysregulated kinase networks in Alzheimer’s disease

Abstract

Alzheimer’s disease (AD) is a form of dementia characterized by amyloid-β plaques and tau neurofibrillary tangles that progressively disrupt neural circuits in the brain. The signaling networks underlying AD pathological changes are poorly characterized at the phosphoproteome level. Using mass spectrometry, we analyzed the proteome and tyrosine, serine and threonine phosphoproteomes of temporal cortex tissue from patients with AD and aged-matched controls. We identified cocorrelated peptide clusters that were linked to varying levels of phospho-tau, oligodendrocyte, astrocyte, microglia and neuron pathologies. We found that neuronal synaptic protein abundances were strongly anti-correlated with markers of microglial reactivity. We also observed that phosphorylation sites on kinases targeting tau and other new signaling factors were correlated with these peptide modules. Finally, we used data-driven statistical modeling to identify individual peptides and peptide clusters that were predictive of AD histopathologies. Together, these results build a map of pathology-associated phosphorylation signaling events occurring in AD.

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: Combined phosphoproteomics and proteomics analysis captures the molecular signaling profile of AD.
Fig. 2: Clustering analysis identifies pathology-associated peptide modules.
Fig. 3: Correlation analysis finds pathology-associated change in the full peptidome.
Fig. 4: Kinome map of the pathology-associated phosphoproteome.
Fig. 5: PLSR model ties clusters and peptides to AD histopathologies.

Similar content being viewed by others

Data availability

Data supporting findings from this study are available from the lead contact, F.M.W., upon request. The MS raw and searched proteomics data files have been deposited to the ProteomeXchange Consortium via the PRIDE114 partner repository with the dataset accession no. PXD020087 and 10.6019/PXD020087. Patient clinical histopathology data and MS-run information are available in Supplementary Data 1. Processed peptide data are available in Supplementary Data 2. All calculated peptide feature correlations and group fold-changes are available in Supplementary Data 4 and 5. Cell type-specific sequencing data were downloaded from BrainRNASeq (https://web.stanford.edu/group/barres_lab/brainseq2/TableS4-HumanMouseMasterFPKMList.xlsx). Kinase–substrate and homology mapping data were downloaded from PhosphoSite Plus (https://www.phosphosite.org/downloads/Kinase_Substrate_Dataset.gz, https://phosphosite.org/downloads/Phosphorylation_site_dataset.gz). BRSK1 and BRSK2 substrates were derived from MaxQuant processed ‘_phosphoSites_filtered_imputed.txt’ file, shared directly by Tamir et al.112. For GO analysis, ‘gene2go’ (ftp://ftp.ncbi.nlm.nih.gov/gene/DATA/gene2go.gz) and ‘go-basic.obo’ (http://geneontology.org/ontology/go-basic.obo) were downloaded using goatools. Mouse phosphoproteome data for analysis was sourced from Morshed et al.22: dataset EV1. Mouse source MS data are available on PRIDE via accession no. PXD018757.

Code availability

The proteomics data integration software is available on GitHub (https://github.com/white-lab/pyproteome). This repository also includes detailed software for concatenating peptide fractions on a Gibson FC 204 Fraction Collection (https://github.com/white-lab/fc-cycle) and an updated tool for validating PSMs (https://github.com/white-lab/CAMV). Sources for other code used in the present study are indicated in Supplementary Information.

References

  1. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement. 16, 391–460 (2020).

  2. Musiek, E. S. & Holtzman, D. M. Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat. Neurosci. 18, 800–806 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. De Strooper, B. & Karran, E. The cellular phase of Alzheimer’s disease. Cell 164, 603–615 (2016).

    Article  PubMed  CAS  Google Scholar 

  4. Long, J. M. & Holtzman, D. M. Alzheimer disease: an update on pathobiology and treatment strategies. Cell 179, 312–339 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ittner, L. M. & Götz, J. Amyloid-β and tau—a toxic pas de deux in Alzheimer’s disease. Nat. Rev. Neurosci. 12, 65–72 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Cavallini, A. et al. An unbiased approach to identifying tau kinases that phosphorylate tau at sites associated with Alzheimer disease. J. Biol. Chem. 288, 23331–23347 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jicha, G. A. et al. cAMP-dependent protein kinase phosphorylations on Tau in Alzheimer’s disease. J. Neurosci. 19, 7486–7494 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Alfonso, S. I. et al. Gain-of-function mutations in protein kinase Cα (PKCα) may promote synaptic defects in Alzheimer’s disease. Sci. Signal. 9, ra47 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Callender, J. A. et al. Protein kinase Cα gain-of-function variant in Alzheimer’s disease displays enhanced catalysis by a mechanism that evades down-regulation. Proc. Natl Acad. Sci. USA 115, E5497–E5505 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Reichenbach, N. et al. Inhibition of Stat3‐mediated astrogliosis ameliorates pathology in an Alzheimer’s disease model. EMBO Mol. Med. 11, e9665 (2019).

  11. Haim, L. Ben et al. The JAK/STAT3 pathway is a common inducer of astrocyte reactivity in Alzheimer’s and Huntington’s diseases. J. Neurosci. 35, 2817–2829 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Styr, B. & Slutsky, I. Imbalance between firing homeostasis and synaptic plasticity drives early-phase Alzheimer’s disease. Nat. Neurosci. 21, 463–473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Seo, J. et al. Inhibition of p25/Cdk5 attenuates tauopathy in mouse and iPSC models of frontotemporal dementia. J. Neurosci. 37, 9917–9924 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Seo, J. et al. Activity-dependent p25 generation regulates synaptic plasticity and aβ-induced cognitive impairment. Cell 157, 486–498 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rigbolt, K. T. G. & Blagoev, B. Quantitative phosphoproteomics to characterize signaling networks. Semin. Cell Dev. Biol. https://doi.org/10.1016/j.semcdb.2012.05.006 (2012).

  16. Riley, N. M. & Coon, J. J. Phosphoproteomics in the age of rapid and deep proteome profiling. Anal. Chem. https://doi.org/10.1021/acs.analchem.5b04123 (2016).

  17. Lemeer, S. & Heck, A. J. The phosphoproteomics data explosion. Curr. Opin. Chem. Biol. https://doi.org/10.1016/j.cbpa.2009.06.022 (2009).

  18. Ficarro, S. B. et al. Phosphoproteome analysis by mass spectrometry and its application to Saccharomyces cerevisiae. Nat. Biotechnol. 20, 301–305 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Zhang, Y. et al. Time-resolved mass spectrometry of tyrosine phosphorylation sites in the epidermal growth factor receptor signaling network reveals dynamic modules. Mol. Cell Proteomics 4, 1240–1250 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. White, F. M. & Wolf-Yadlin, A. Methods for the analysis of protein phosphorylation-mediated cellular signaling networks. Annu. Rev. Anal. Chem. https://doi.org/10.1146/annurev-anchem-071015-041542 (2016).

  21. Bai, B. et al. Deep multilayer brain proteomics identifies molecular networks in Alzheimer’s disease progression. Neuron 105, 975–991.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Morshed, N. et al. Phosphoproteomics identifies microglial Siglec‐F inflammatory response during neurodegeneration. Mol. Syst. Biol. 16, e9819 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tagawa, K. et al. Comprehensive phosphoproteome analysis unravels the core signaling network that initiates the earliest synapse pathology in preclinical Alzheimer’s disease brain. Hum. Mol. Genet. 24, 540–558 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Dammer, E. B. et al. Quantitative phosphoproteomics of Alzheimer’s disease reveals cross-talk between kinases and small heat shock proteins. Proteomics 15, 508–519 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Marttinen, M. et al. A multiomic approach to characterize the temporal sequence in Alzheimer’s disease-related pathology. Neurobiol. Dis. 124, 454–468 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. 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 

  27. Ping, L. et al. Global quantitative analysis of the human brain proteome in Alzheimer’s and Parkinson’s disease. Sci. Data 5, 180036 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wingo, A. P. et al. Large-scale proteomic analysis of human brain identifies proteins associated with cognitive trajectory in advanced age. Nat. Commun. 10, 1619 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Di Domenico, F. et al. Quantitative proteomics analysis of phosphorylated proteins in the hippocampus of Alzheimer’s disease subjects. J. Proteomics 74, 1091–1103 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Triplett, J. C., Swomley, A. M., Cai, J., Klein, J. B. & Butterfield, D. A. Quantitative phosphoproteomic analyses of the inferior parietal lobule from three different pathological stages of Alzheimer’s disease. J. Alzheimers Dis. 49, 45–62 (2016).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mattsson, N. et al. Association of plasma neurofilament light with neurodegeneration in patients with Alzheimer disease. JAMA Neurol. https://doi.org/10.1001/jamaneurol.2016.6117 (2017).

  34. 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. https://doi.org/10.1001/jamaneurol.2019.0765 (2019).

  35. Preische, O. et al. Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer’s disease. Nat. Med. https://doi.org/10.1038/s41591-018-0304-3 (2019).

  36. Davidsson, P., Jahn, R., Bergquist, J., Ekman, R. & Blennow, K. Synaptotagmin, a synaptic vesicle protein, is present in human cerebrospinal fluid—a new biochemical marker for synaptic pathology in Alzheimer disease? Mol. Chem. Neuropathol. https://doi.org/10.1007/BF02815094 (1996).

  37. Galasko, D. et al. Synaptic biomarkers in CSF aid in diagnosis, correlate with cognition and predict progression in MCI and Alzheimer’s disease. Alzheimer’s Dement. Transl. Res. Clin. Interv. https://doi.org/10.1016/j.trci.2019.11.002 (2019).

  38. Wingo, A. P. et al. Shared proteomic effects of cerebral atherosclerosis and Alzheimer’s disease on the human brain. Nat. Neurosci. https://doi.org/10.1038/s41593-020-0635-5 (2020).

  39. Jack, C. R. et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 12, 207–216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Janelidze, S. et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nat. Commun. 11 (2020).

  41. Dujardin, S. et al. Tau molecular diversity contributes to clinical heterogeneity in Alzheimer’s disease. Nat. Med. https://doi.org/10.1038/s41591-020-0938-9 (2020).

  42. Wesseling, H. et al. Tau PTM profiles identify patient heterogeneity and stages of Alzheimer’s disease. Cell https://doi.org/10.1016/j.cell.2020.10.029 (2020).

  43. Harauz, G. & Boggs, J. M. Myelin management by the 18.5-kDa and 21.5-kDa classic myelin basic protein isoforms. J. Neurochem. 125, 334–361 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lin, H., Lin, T. Y. & Juang, J. L. Abl deregulates Cdk5 kinase activity and subcellular localization in Drosophila neurodegeneration. Cell Death Differ. 14, 607–615 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Zukerberg, L. R. et al. Cables links Cdk5 and c-Abl and facilitates Cdk5 tyrosine phosphorylation, kinase upregulation, and neurite outgrowth Cdks are involved in events unrelated to cell division. A function of Cdk5 has been demonstrated only in the development of the nervous system. Neuron 26, 633–646 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Muñoz, I. M. et al. Phosphoproteomic screening identifies physiological substrates of the CDKL5 kinase. EMBO J. 37, e99559 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Mikolcevic, P. et al. Cyclin-dependent kinase 16/PCTAIRE kinase 1 is sctivated by cyclin Y and is essential for spermatogenesis. Mol. Cell. Biol. 32, 868–879 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chaput, D., Kirouac, L., Stevens, S. M., Padmanabhan, J. & Padmanabhan, J. Potential role of PCTAIRE-2, PCTAIRE-3 and P-Histone H4 in amyloid precursor protein-dependent Alzheimer pathology. Oncotarget 7, 8481–8497 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Patrick, G. N. et al. Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodegeneration. Nature 402, 615–622 (1999).

    Article  CAS  PubMed  Google Scholar 

  50. Herskovits, A. Z. & Davies, P. The regulation of tau phosphorylation by PCTAIRE 3: Implications for the pathogenesis of Alzheimer’s disease. Neurobiol. Dis. 23, 398–408 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Hensley, K. et al. p38 kinase is activated in the Alzheimer’s disease brain. J. Neurochem. 72, 2053–2058 (1999).

    Article  CAS  PubMed  Google Scholar 

  52. Ghosh, A. & Giese, K. P. Calcium/calmodulin-dependent kinase II and Alzheimer’s disease. Mol. Brain 8, 78 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Yarza, R., Vela, S., Solas, M. & Ramirez, M. J. c-Jun N-terminal kinase (JNK) signaling as a therapeutic target for Alzheimer’s disease. Front. Pharmacol. 6, 321 (2015).

    PubMed  Google Scholar 

  54. Lisnock, J. et al. Activation of JNK3 alpha 1 requires both MKK4 and MKK7: kinetic characterization of in vitro phosphorylated JNK3 alpha 1. Biochemistry 39, 3141–3148 (2000).

    Article  CAS  PubMed  Google Scholar 

  55. Brichkina, A. et al. P38MAPK builds a hyaluronan cancer niche to drive lung tumorigenesis. Genes Dev. 30, 2623–2636 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Seidler, J., Adal, M., Kübler, D., Bossemeyer, D. & Lehmann, W. D. Analysis of autophosphorylation sites in the recombinant catalytic subunit alpha of cAMP-dependent kinase by nano-UPLC-ESI-MS/MS. Anal. Bioanal. Chem. 395, 1713–1720 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Geng, J. et al. Kinases Mst1 and Mst2 positively regulate phagocytic induction of reactive oxygen species and bactericidal activity. Nat. Immunol. 16, 1142–1152 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Robison, A. J., Winder, D. G., Colbran, R. J. & Bartlett, R. K. Oxidation of calmodulin alters activation and regulation of CaMKII. Biochem. Biophys. Res. Commun. https://doi.org/10.1016/j.bbrc.2007.02.087 (2007).

  59. Giese, K. P., Fedorov, N. B., Filipkowski, R. K. & Silva, A. J. Autophosphorylation at Thr286 of the α calcium-calmodulin kinase II in LTP and learning. Science 279, 870–873 (1998).

    Article  CAS  PubMed  Google Scholar 

  60. Boggon, T. J. & Eck, M. J. Structure and regulation of Src family kinases. Oncogene 23, 7918–7927 (2004).

    Article  CAS  PubMed  Google Scholar 

  61. Pan, Q. et al. Cdk5 targets active Src for ubiquitin-dependent degradation by phosphorylating Src(S75). Cell Mol. Life Sci. 68, 3425–3436 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Weir, M. E. et al. Novel autophosphorylation sites of Src family kinases regulate kinase activity and SH2 domain-binding capacity. FEBS Lett. 590, 1042–1052 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Manser, C., Vagnoni, A., Guillot, F., Davies, J. & Miller, C. C. J. Cdk5/p35 phosphorylates lemur tyrosine kinase-2 to regulate protein phosphatase-1C phosphorylation and activity. J. Neurochem. 121, 343–348 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Brunton, V. G. et al. Identification of Src-specific phosphorylation site on focal adhesion kinase: dissection of the role of Src SH2 and catalytic functions and their consequences for tumor cell behavior. Cancer Res. 65, 1335–1342 (2005).

    Article  CAS  PubMed  Google Scholar 

  65. Fu, H. L. et al. Glycosylation at Asn211 regulates the activation state of the discoidin domain receptor 1 (DDR1). J. Biol. Chem. 289, 9275–9287 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Hebron, M. et al. Discoidin domain receptor inhibition reduces neuropathology and attenuates inflammation in neurodegeneration models. J. Neuroimmunol. 311, 1–9 (2017).

    Article  CAS  PubMed  Google Scholar 

  67. Fowler, A. J. et al. Multikinase Abl/DDR/Src inhibition produces optimal effects for tyrosine kinase inhibition in neurodegeneration. Drugs R. D. 19, 149–166 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Fowler, A. J. et al. Discoidin domain receptor 1 is a therapeutic target for neurodegenerative diseases. Hum. Mol. Genet. https://doi.org/10.1093/hmg/ddaa177 (2020).

  69. Reddy, R. J. et al. Early signaling dynamics of the epidermal growth factor receptor. Proc. Natl Acad. Sci. USA 113, 3114–3119 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hornbeck, P. V. et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 43, D512–D520 (2015).

    Article  CAS  PubMed  Google Scholar 

  71. Okuno, S., Kitani, T. & Fujisawa, H. Regulation of Ca2+/calmodulin-dependent protein kinase kinase alpha by cAMP-dependent protein kinase: I. Biochemical analysis. J. Biochem. 130, 503–513 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Bürklen, T. S. et al. The creatine kinase/creatine connection to Alzheimer’s disease: CK-inactivation, APP–CK complexes and focal creatine deposits. J. Biomed. Biotechnol. 2006, 35936 (2006).

  73. Aksenov, M., Aksenova, M., Butterfield, D. A. & Markesbery, W. R. Oxidative modification of creatine kinase BB in Alzheimer’s disease brain. J. Neurochem. 74, 2520–2527 (2000).

    Article  CAS  PubMed  Google Scholar 

  74. Gonzales, E. B., Smith, R. N. & Agharkar, A. S. A review of creatine supplementation in age-related diseases: more than a supplement for athletes. F1000Research https://doi.org/10.12688/f1000research.5218.1 (2014).

  75. Matthews, R. T. et al. Neuroprotective effects of creatine and cyclocreatine in animal models of Huntington’s disease. J. Neurosci. 18, 156–163 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Ulland, T. K. et al. TREM2 maintains microglial metabolic fitness in Alzheimer’s disease. Cell 170, 649–663 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Carbajosa, G. et al. Loss of Trem2 in microglia leads to widespread disruption of cell coexpression networks in mouse brain. Neurobiol. Aging 69, 151–166 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kirouac, L., Rajic, A. J., Cribbs, D. H. & Padmanabhan, J. Activation of Ras-ERK signaling and GSK-3 by amyloid precursor protein and amyloid beta facilitates neurodegeneration in Alzheimer’s disease. eNeuro https://doi.org/10.1523/ENEURO.0149-16.2017 (2017).

  79. Arstikaitis, P. et al. Paralemmin-1, a modulator of filopodia induction is required for spine maturation. Mol. Biol. Cell 19, 2026–2038 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zorzetto, M. et al. Complement C4A and C4B gene copy number study in Alzheimer’s disease patients. Curr. Alzheimer Res. https://doi.org/10.2174/1567205013666161013091934 (2016).

  81. Sellgren, C. M. et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat. Neurosci. https://doi.org/10.1038/s41593-018-0334-7 (2019).

  82. Hong, S. et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 8373, 712–716 (2016).

    Google Scholar 

  83. Beckmann, N. D. et al. Multiscale causal networks identify VGF as a key regulator of Alzheimer’s disease. Nat. Commun. 11, 3942 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53 (2016).

    Article  CAS  PubMed  Google Scholar 

  85. 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  PubMed  PubMed Central  Google Scholar 

  86. Cullen, N. C. et al. Individualized prognosis of cognitive decline and dementia in mild cognitive impairment based on plasma biomarker combinations. Nat. Aging https://doi.org/10.1038/s43587-020-00003-5 (2020).

  87. Hong, S., Dissing-Olesen, L. & Stevens, B. New insights on the role of microglia in synaptic pruning in health and disease. Curr. Opin. Neurobiol. 36, 128–134 (2016).

    Article  CAS  PubMed  Google Scholar 

  88. Yilmaz, M. et al. Overexpression of schizophrenia susceptibility factor human complement C4A promotes excessive synaptic loss and behavioral changes in mice. Nat. Neurosci. https://doi.org/10.1038/s41593-020-00763-8 (2020).

  89. Sekar, A. et al. Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ferreira, S. et al. Oligodendrogenesis increases in hippocampal grey and white matter prior to locomotor or memory impairment in an adult mouse model of tauopathy. Eur. J. Neurosci. https://doi.org/10.1111/ejn.14726 (2020).

  91. Birdsill, A. C., Walker, D. G., Lue, L. F., Sue, L. I. & Beach, T. G. Postmortem interval effect on RNA and gene expression in human brain tissue. Cell Tissue Bank. 12, 311–318 (2011).

    Article  CAS  PubMed  Google Scholar 

  92. Walker, D. G. et al. Characterization of RNA isolated from eighteen different human tissues: results from a rapid human autopsy program. Cell Tissue Bank. 17, 361–375 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Emdal, K. B. et al. Characterization of in vivo resistance to osimertinib and JNJ-61186372, an EGFR/Met bi-specific antibody, reveals unique and consensus mechanisms of resistance. Mol. Cancer Ther. https://doi.org/10.1158/1535-7163.MCT-17-0413 (2017).

  94. Huang, P. H. et al. Phosphotyrosine signaling analysis of site-specific mutations on EGFRvIII identifies determinants governing glioblastoma cell growth. Mol. Biosyst. 6, 1227–1237 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Miraldi, E. R. et al. Molecular network analysis of phosphotyrosine and lipid metabolism in hepatic PTP1b deletion mice. Integr. Biol. (United Kingdom) 5, 940–963 (2013).

    Article  CAS  Google Scholar 

  96. Zecha, J. et al. TMT labeling for the masses: a robust and cost-efficient, in-solution labeling approach. Mol. Cell. Proteomics 18, 1468–1478 (2019).

  97. Wolf-Yadlin, A. et al. Effects of HER2 overexpression on cell signaling networks governing proliferation and migration. Mol. Syst. Biol. 2, 54 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Johnson, H. & White, F. M. Toward quantitative phosphotyrosine profiling in vivo. Semin. Cell Dev. Biol. 23, 854–862 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Johnson, H. et al. Molecular characterization of EGFR and EGFRvIII signaling networks in human glioblastoma tumor xenografts. Mol. Cell. Proteomics 11, 1724–1740 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Gajadhar, A. S. et al. Phosphotyrosine signaling analysis in human tumors is confounded by systemic ischemia-driven artifacts and intra-specimen heterogeneity. Cancer Res. 75, 1495–1503 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Taus, T. et al. Universal and confident phosphorylation site localization using phosphoRS. J. Proteome Res. https://doi.org/10.1021/pr200611n (2011).

  102. Maes, E. et al. CONSTANd: a normalization method for isobaric labeled spectra by constrained optimization. Mol. Cell. Proteomics 15, 2779–2790 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Pedregosa, F. et al. Scikit-learn: machine learning in python. J. Mach. Learn. Res. 12, 2825–2830 (2012).

    Google Scholar 

  104. Virtanen, P. et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat. Methods https://doi.org/10.1038/s41592-019-0686-2 (2020).

  105. Van Der Walt, S., Colbert, S. C. & Varoquaux, G. The NumPy array: a structure for efficient numerical computation. Comput. Sci. Eng. 13, 22–30 (2011).

    Article  Google Scholar 

  106. McKinney, W. Data structures for statistical computing in Python. In Proceedings of the 9th Python in Science Conference (eds van der Walt, S. & Millman, K. J.) 51–56 (SciPy, 2010).

  107. Waskom, M. mwaskom/seaborn: v0.9.0. Zenodo https://doi.org/10.5281/zenodo.1313201 (2018).

  108. Hunter, J. D. Matplotlib: a 2D graphics environment. Comput. Sci. Eng. 9, 99–104 (2007).

    Article  Google Scholar 

  109. Gaudet, S. et al. A compendium of signals and responses triggered by prodeath and prosurvival cytokines. Mol. Cell. Proteomics 4, 1569–1590 (2005).

    Article  CAS  PubMed  Google Scholar 

  110. Metz, K. S. et al. Coral: clear and customizable visualization of human kinome data. Cell Syst. 7, 347–350 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Krug, K. et al. A curated resource for phosphosite-specific signature analysis. Mol. Cell. Proteomics 18, 576–593 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  112. Tamir, T. Y. et al. Gain-of-function genetic screen of the kinome reveals BRSK2 as an inhibitor of the NRF2 transcription factor. J. Cell Sci. https://doi.org/10.1242/jcs.241356 (2020).

  113. Klopfenstein, D. V. et al. GOATOOLS: a python library for gene ontology analyses. Sci. Rep. https://doi.org/10.1038/s41598-018-28948-z (2018).

  114. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. https://doi.org/10.1093/nar/gky1106 (2019).

Download references

Acknowledgements

We thank members of the laboratories of F.M.W., L.-H. Tsai and D.A.L. for numerous discussions and feedback. N.M. was partially supported by the NIH Biotechnology Training grant (no. T32GM008334). M.J.L. was partially supported through the US Army Research Office Cooperative Agreement (no. W911NF-19-2-0026) for the Institute for Collaborative Biotechnologies and the National Science Foundation Graduate Research Fellowship Program (award no. 1745302). We thank the BSHRI Brain and Body Donation Program of Sun City, AZ for the provision of human brain tissue. The Brain and Body Donation Program has been supported by the National Institute of Neurological Disorders and Stroke (U24 NS072026 National Brain and Tissue Resource for Parkinson’s Disease and Related Disorders), the National Institute on Aging (P30 AG19610 Arizona Alzheimer’s Disease Core Center), the Arizona Department of Health Services (contract no. 211002, Arizona Alzheimer’s Research Center), the Arizona Biomedical Research Commission (contract nos. 4001, 0011, 05-901 and 1001 to the Arizona Parkinson’s Disease Consortium) and the Michael J. Fox Foundation for Parkinson’s Research.

Author information

Authors and Affiliations

Authors

Contributions

N.M. and F.M.W. designed the study. D.M. collected human brain tissue samples. N.M. and F.H.R. collected proteomics datasets. N.M. wrote proteomics data integration and enrichment software. N.M. and M.J.L. performed statistical analyses with methodology and analysis oversight from D.A.L. N.M., M.J.L. and F.M.W. wrote the manuscript.

Corresponding author

Correspondence to Forest M. White.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks the anonymous reviewers 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 Proteome and phosphoproteome dataset statistics and quantification reproducibility.

a–c, Venn diagram showing peptide overlap between (a) pTyr sites, (b) pSer/pThr sites, and (c) proteins identified in each 10-plex analysis. d, Cumulative number of peptides identified compared to the number of patients with peptide quantification. e-f, Reproducibility analysis of peptide quantification after data integration and normalization. Shown are the log2 quantification ratios between two separate brain samples from patients (e) 06-49 and (f) 06-03. Peptides are separated on the x-axis by log10 total TMT intensity from all PSMs in both 10-plex analyses. The standard deviation of abundance ratios and Pearson’s correlation coefficient between all peptide replicates are listed in the panels. g,h, Reproducibility of peptide quantification between technical replicates of peptide aliquots for each TMT10-plex analysis. Shown are (g) the standard deviation of abundance ratios and (h) the Pearson’s correlation coefficient calculated over all peptides. Box plot indicates quartiles, and whiskers indicate the last datum within 1.5 interquartile range. Individual data points are shown for each technical replicate.

Extended Data Fig. 2 Accessory clustering and dataset comparison analyses.

a-b, Properties of clusters generated by clustering with increasing number of clusters. Shown are the (a) mean Pearson correlation coefficient for cluster peptides with their respective cluster centroid, averaged over all clusters, and (b) minimum absolute Pearson correlation coefficient for cluster peptides with their respective cluster centroid, summed and normalized over all clusters. Dotted line indicates the final cluster count used for downstream analysis. c, Co-correlation heatmap between all cluster centroids, group by cluster. Selected named clusters are shown on the y-axis. d, Phosphopeptide composition of all peptides associated with cluster centroids. Peptides are labeled as ‘pY’ if they contain at least one pTyr site, ‘pST’ if they contain at least one pSer or pThr site, or ‘no phospho’ if they are not phosphorylated. Statistics calculations are same as Fig. 2g. e, Binomial enrichment for proteome module marker proteins identified in Johnson et al. (2020). Heatmap shows log-odds enrichment (LOE) values for the overlap of marker transcripts with pTyr, pSer/pThr, and proteome datasets that are correlated with cluster centroids. f, Binomial enrichment for single-nuclei populations markers identified in Mathys et al. (2019). Legend is same as (e). g, Venn diagram overlap between proteins correlated with Oligo in our dataset, M2 Oligo proteome module from (e) and Oli0 marker transcripts from (f). Numbers indicate the total number of proteins in each overlapping set and selected protein names are shown. h, Transcript log2 Fold Change values Oli0 population from Mathys et al. (2019) for markers of the Oligo cluster.

Extended Data Fig. 3 Peptide cluster principle component and stratification analyses.

a, Principal component analysis (PCA) of peptides quantified in all samples. Patients are colored by (a) AD or ND status, (b) Tau, (c) Oligo, (d) Astro, (e) Micro, (f) pNeuro, and (g) Neuro cluster centroid values. h–j, PCA of samples colored by patient stratifications using (h) Tau and Oligo, (i) Astro and Micro, and (j) pNeuro and Neuro cluster centroids. k,l, Patients numbers for (k) Astro;Micro or (l) pNeuro;Neuro patient stratifications. m,n, Overlap between Tau;Oligo and (m) Astro;Micro or (n) pNeuro;Neuro patient stratifications.

Extended Data Fig. 4 Peptide correlation analyses.

a,b, Distribution of Spearman’s correlation coefficient for peptides identified in ≥15 patients with (a) scrambled cluster centroids or (b) true cluster centroids. c,d, Distribution of Spearman’s correlation p-values for peptides identified in ≥15 patients with (c) scrambled cluster centroids or (d) true cluster centroids. (e–g) Venn diagram showing the number of correlated peptides with (e) Tau and Oligo, (f) Astro and Micro, and (g) pNeuro and Neuro cluster centroids. Plots include the total number of associated peptides for each cluster, as well as the number of overlapping peptides between similar clusters. h, Heatmap showing phosphopeptide fold changes for peptides that are significantly correlated with Tau cluster centroid. Samples are ordered by centroid values. Left color bars indicate proteins that were predicted to be cell type-specific. Green = Astrocyte, Purple = Oligodendrocyte, Yellow = Neuron, Blue = Microglia. Missing values are indicated as white boxes. i, Heatmap showing fold changes cell type-specific proteins that are significantly correlated with Oligo cluster centroid. Samples are ordered by centroid value. Left color bar legend is same as (h).

Extended Data Fig. 5 Analyses of peptide partial cluster correlation.

a-d, Fold changes for (a) MAPT pS579 (pS262), (b) MAPT pT534 (pS217), (c) MBP protein, and (d) PLP1 protein. Protein levels were estimated from the median abundance of all unmodified peptides that map uniquely to that protein. Values are normalized to the median of Tau Oligo patients. Box plot indicates quartiles, and whiskers indicate the last datum within 1.5 interquartile range. Individual data points are shown for each sample. Stars indicate two-sided t-test values for each group compared with Tau Oligo and Tau Oligo+ patients (n = 8-14 patients in each group). ns, not significant (p > 0.05); * p < 5e-2, **p < 1e-2; ***p < 1e-3; ****p < 1e-4. e, All peptides mapping to the peripheral nervous system (PNS) isoform of MAPT. Colored bars indicate directional correlation for non-phosphorylated peptides. Red = correlated, blue = anti-correlated, grey = uncorrelated, light-grey = only phosphopeptides were seen in that region. Colored circles indicate phosphorylation sites that were quantified. Red circle = correlated, blue circle = anti-correlated, black circle = uncorrelated. Green dividers indicate the borders between the N-terminal projection, proline-rich domain, microtubule-binding repeats, and C-terminal flanking region. f, All peptides mapping to the full-length isoform of MBP. Legend is same as (e). g–r, Fold changes across Astro+ and Micro+ groups for (g) GFAP protein, (h) VIM protein, (i) GNA13 protein, (j) AIF1 protein, (k) ITGB2 protein, (l) SPP1 pS270, (m) C1QA protein, (n) C1QB protein, (o) C1QC protein, (p) C3 protein, (q) C4A protein, and (r) C4B protein. Legend is same as (a-d). Stars indicate two-sided t-test values for each group compared with Astro Micro and Astro+ Micro patients (n = 12-14 patients in each group). ns, not significant (p > 0.05); * p < 5e-2, **p < 1e-2; ***p < 1e-3; ****p < 1e-4.

Extended Data Fig. 6 Tau kinase and CDK phosphosite abundances.

a-d, Fold changes for (a) TNIK pS680, (b) BRSK1 pS443, (c) BRSK2 pS423, and (d) TTBK1 pS483 across Tau and Oligo groups. Values are normalized to the median of Tau Oligo patients. Box plot indicates quartiles, and whiskers indicate the last datum within 1.5 interquartile range. Individual data points are shown for each sample. Stars indicate two-sided t-test values for each group compared with Tau Oligo and Tau Oligo+ patients (n = 8-14 patients in each group). ns, not significant (p > 0.05); * p < 5e-2, **p < 1e-2; ***p < 1e-3; ****p < 1e-4. e–h, Fold changes for all non-phospho peptides and significantly changing phosphosites on (e) TNIK, (f) BRSK1, (g) BRSK2, (h) TTBK1. Each dot indicates one unique peptide. Phosphosite ranges are estimated from peptides with missed cleavages. Box plot indicates quartiles, and whiskers indicate the last datum within 1.5 interquartile range. Phosphosites labeled with ‘*’ are on peptides that map ambiguously to more than one protein. i–m, Fold changes for (i) CDK5 pY15, (j) CDKL5 pY171, (k) CDK16 pS153, (l) CDK17 pS180, and (m) CDK18 pS132 across Tau and Oligo groups. Legend is same as (a-d). n–r, Fold changes for all non-phospho peptides and significantly changing phosphosites on (n) CDK5, (o) CDKL5, (p) CDK16, (q) CDK17, and (r) CDK18. Legend is same as (e-h).

Extended Data Fig. 7 Accessory kinome analyses.

a, Maximum phosphosite fold changes and median protein abundances for all kinases shown in Fig. 4d. Changes are calculated for each Tau;Oligo group compared to Tau;Oligo. Stars indicate two-sided t-test values for each group compared with Tau Oligo patients (n = 8-14 patients in each group). *p < 5e-2; **p < 1e-3; ***p < 1e-4. b–m, Fold changes for all non-phospho peptides and significantly changing phosphosites on (b) MAPK8 (c) MAPK10, (d) MAPK11, (e) MAPK14, (f) PRKACA, (g) PRKCA, (h) all CaMKII subunits, (i) SRC, (j) LMTK2, (k) PTK2, (l) DDR1, and (m) EGFR. Legend is same as Extended Data Figure 6e.

Extended Data Fig. 8 Kinase-substrate and AD mouse model analyses.

a–d, Volcano plots showing Spearman’s correlation coefficient and p-values between PKA Cα substrates and (a) Tau, (b) Astro, (d) Oligo, and (d) pNeuro cluster centroids. Labels are colored using the same scheme as Extended Data Figure 4h. e, Number of phosphosites from AD mouse models identified in three mouse models of neurodegeneration that could be translated to human and were significantly correlated with each cluster centroid and shared directional changes. f, Phosphoproteins that were identified to have at least one shared phosphosite with three mouse models of neurodegeneration. Proteins are shown as a Venn diagram between downregulated phosphosites and pNeuro or Neuro cluster centroids.

Extended Data Fig. 9 PLSR accessory analyses

a–c, Latent variable plots for multivariate PLSR analysis for X scores. Points are colored by patient (a) NIA-R score, (b) CERAD-NP score, and (c) TangleT score. d, K-fold validation of the multivariate PLSR model for all included histopathology variables. R2 and Q2 values are shown for training and test data predictions respectively. e, Scatter plot showing PLSR patient scores generated from cluster centroids compared with subsets of the matrix of peptides quantified in all patients. f, Fold changes for all non-phospho peptides and significantly changing phosphosites on CKB. Legend is same as Extended Data Fig. 6e. g–j, Fold changes for (g) SYN1, (h) SYT1, (i) CAMKK1 pS52, and (j) CASKIN pS935 across pNeuro and Neuro groups. Box plot indicates quartiles, and whiskers indicate the last datum within 1.5 interquartile range. Individual data points are shown for each sample. Stars indicate two-sided t-test values for each group compared with pNeuro Neuro patients (n = 5-16 patients in each group). ns, not significant (p > 0.05); * p < 5e-2, **p < 1e-2; ***p < 1e-3; ****p < 1e-4. k–m, Fold changes for (k) EMILIN3, (l) C4A, and (m) VGF across Tau and Oligo groups. Legend is same as (g-j). Stars indicate two-sided t-test values for each group compared with Tau Oligo and Tau Oligo+ patients (n = 8-14 patients in each group). ns, not significant (p > 0.05); * p < 5e-2, **p < 1e-2; ***p < 1e-3; ****p < 1e-4.

Extended Data Fig. 10 Additional ROC and PMI analyses.

a,b, ROC curves predicting AD status from (a) protein and (b) phosphosites highlighted in Fig. 5f–h.

Supplementary information

Supplementary Information

Supplementary Note 1, Table 1 and Fig. 1.

Reporting Summary

Supplementary Data 1

Demographic information and clinical histopathology measurements for individual patients.

Supplementary Data 2

Full peptidome dataset with individual quantification values.

Supplementary Data 3

Peptide cluster composition: cluster identities and centroid values for peptides quantified in all samples.

Supplementary Data 4

Proteome and phosphoproteome cluster correlations: the table includes the Spearman’s correlation and associated P values between each peptide and each cluster centroid.

Supplementary Data 5

Proteome and phosphoproteome cluster fold-changes: the table includes fold-changes and P values for each peptide and cluster group against their respective negative cases.

Supplementary Data 6

AD mouse phosphoproteome overlap: AD mouse model phosphosites from Morshed et al.22 that had homologous phosphosites detected in the human AD phosphoproteome.

Supplementary Data 7

Peptide scores and VIP values for peptide-centric PLSR models: the table includes tabs for PLSR models built from pTyr, pSer/pThr, protein and all data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morshed, N., Lee, M.J., Rodriguez, F.H. et al. Quantitative phosphoproteomics uncovers dysregulated kinase networks in Alzheimer’s disease. Nat Aging 1, 550–565 (2021). https://doi.org/10.1038/s43587-021-00071-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00071-1

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