Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile

Charisse N Winston, Edward J Goetzl, Johnny C Akers, Bob S Carter, Edward M Rockenstein, Douglas Galasko, Eliezer Masliah, Robert A Rissman, Charisse N Winston, Edward J Goetzl, Johnny C Akers, Bob S Carter, Edward M Rockenstein, Douglas Galasko, Eliezer Masliah, Robert A Rissman

Abstract

Introduction: Levels of Alzheimer's disease (AD)-related proteins in plasma neuronal derived exosomes (NDEs) were quantified to identify biomarkers for prediction and staging of mild cognitive impairment (MCI) and AD.

Methods: Plasma exosomes were extracted, precipitated, and enriched for neuronal source by anti-L1CAM antibody absorption. NDEs were characterized by size (Nanosight) and shape (TEM) and extracted NDE protein biomarkers were quantified by ELISAs. Plasma NDE cargo was injected into normal mice, and results were characterized by immunohistochemistry to determine pathogenic potential.

Results: Plasma NDE levels of P-T181-tau, P-S396-tau, and Aβ1-42 were significantly higher, whereas those of neurogranin (NRGN) and the repressor element 1-silencing transcription factor (REST) were significantly lower in AD and MCI converting to AD (ADC) patients compared to cognitively normal controls (CNC) subjects and stable MCI patients. Mice injected with plasma NDEs from ADC patients displayed increased P-tau (PHF-1 antibody)-positive cells in the CA1 region of the hippocampus compared to plasma NDEs from CNC and stable MCI patients.

Conclusions: Abnormal plasma NDE levels of P-tau, Aβ1-42, NRGN, and REST accurately predict conversion of MCI to AD dementia. Plasma NDEs from demented patients seeded tau aggregation and induced AD-like neuropathology in normal mouse CNS.

Keywords: Alzheimer's disease; Beta amyloid; Biomarker; Exosomes; Mild cognitive impairment; Neuron; Phospho-tau.

Figures

Fig. 1
Fig. 1
Plasma NDEs derived from stable MCI and ADC patients are similar in size and shape to previously reported exosome preparations. (A) Representative TEM image of plasma NDEs derived from a stable MCI patient (scale bars 350 nm; 100 nm). NTA detects high concentrations of plasma NDEs (B) with similar size distributions (C) that are not significantly different between the two patient populations. (C) Representative NTA plot of averaged size/concentration for plasma NDEs derived from a MCI and ADC patient (n = 4/group). Abbreviations: NDE, neuronal derived exosome; MCI, mild cognitive impairment; ADC, MCI converting to AD; TEM, transmission electron microscopy; NTA, nanoparticle tracking analysis.
Fig. 2
Fig. 2
Human plasma NDE levels of P-T181-tau, P-S396-tau, Aβ1–42, NRGN, and REST delineate stages of AD. (A) Plasma NDE levels of CD81 were not statistically different among the four clinical cohorts as measured by ELISA. Significantly different levels of (B) Aβ1–42, (C) P-T181-tau, (D) P-S396-tau, (E) NRGN, and (F) REST were detected by ELISAs in the plasmas of CNC (n = 10), stable MCI (n = 20), ADC (n = 20), and AD (n = 10) patients. Plasma NDE levels of P-T181-tau, P-S396-tau, and Aβ1–42 were significantly higher, whereas those of NRGN and REST were significantly lower for ADC and AD patients than CNC subjects and stable MCI patients. The horizontal line in each cluster depicts the mean for that set. * = P < .05, ** = P < .01, *** = P < .0001 versus CNC; ## = P < .01, ### = P < .0001 versus MCI. Abbreviations: NDE, neuronal derived exosome; CNC, cognitively normal controls; MCI, mild cognitive impairment; ADC, MCI converting to AD.
Fig. 3
Fig. 3
ROC curve analyses assess diagnostic accuracy for plasma NDEs levels of P-T181-tau, P-S396-tau, Aβ1–42, NRGN, and REST. ROC curves based on the plasma NDE levels of P-T181-tau, P-S396-tau, Aβ1–42, NRGN, and REST for (A) CNC subjects versus AD patients, (B) CNC subjects versus stable MCI patients, (C) stable MCI versus ADC patients, and (D) stable MCI versus AD patients. Abbreviations: ROC, receiver operating curve; NDE, neuronal derived exosome; CNC, cognitively normal control; MCI, mild cognitive impairment; ADC, MCI converting to AD.
Fig. 4
Fig. 4
Plasma NDEs from stable MCI and AD patients seed tau aggregation in the brains of normal mice. Representative photomicrographs of hippocampal sections obtained from wild-type, C57/Bl6 mice that were injected with plasma NDEs from control, stable MCI, and ADC patients. At 1-month post-injection, mice injected with plasma NDEs from stable MCI patients display modest enhancement of PHF-1 immunoreactivity in the CA1 region of the hippocampus, whereas mice injected with plasma NDEs from ADC patients display a significant increase in PHF-1 immunoreactivity in the CA1. Extensive cellular staining as well as staining within the dendritic processes can be observed in these mice. Mice-injected NDEs from control plasma displayed no PHF-1 immunoreactivity (n = 6/group; scale bars 250 μm; 25 μm). Abbreviations: NDE, neuronal derived exosome; MCI, mild cognitive impairment; ADC, MCI converting to AD.

References

    1. Hebert L.E., Weuve J., Scherr P.A., Evans D.A. Alzheimer disease in the United States (2010-2050) estimated using the 2010 census. Neurology. 2013;80:1778–1783.
    1. Ward A., Arrighi H.M., Michels S., Cedarbaum J.M. Mild cognitive impairment: disparity of incidence and prevalence estimates. Alzheimers Dement. 2012;8:14–21.
    1. Grundman M., Petersen R.C., Ferris S.H., Thomas R.G., Aisen P.S., Bennett D.A. Mild cognitive impairment can be distinguished from Alzheimer disease and normal aging for clinical trials. Arch Neurol. 2004;61:59–66.
    1. van Rossum I.A., Vos S., Handels R., Visser P.J. Biomarkers as predictors for conversion from mild cognitive impairment to Alzheimer-type dementia: implications for trial design. J Alzheimers Dis. 2010;20:881–891.
    1. Parnetti L., Farotti L., Eusebi P., Chiasserini D., De Carlo C., Giannandrea D. Differential role of CSF alpha-synuclein species, tau, and Aβ42 in Parkinson's Disease. Front Aging Neurosci. 2014;6:53.
    1. Lanari A., Parnetti L. Cerebrospinal fluid biomarkers and prediction of conversion in patients with mild cognitive impairment: 4-year follow-up in a routine clinical setting. ScientificWorldJournal. 2009;9:961–966.
    1. Blennow K., Hampel H. CSF markers for incipient Alzheimer's disease. Lancet Neurol. 2003;2:605–613.
    1. Agarwal R., Tripathi C.B. Diagnostic Utility of CSF Tau and Abeta(42) in Dementia: A Meta-Analysis. Int J Alzheimers Dis. 2011;2011:503293.
    1. Andreasson U., Lautner R., Schott J.M., Mattsson N., Hansson O., Herukka S.K. CSF biomarkers for Alzheimer's pathology and the effect size of APOE varepsilon4. Mol Psychiatry. 2014;19:148–149.
    1. Zetterberg H., Lautner R., Skillback T., Rosen C., Shahim P., Mattsson N. CSF in Alzheimer's disease. Adv Clin Chem. 2014;65:143–172.
    1. Mattsson N., Insel P.S., Landau S., Jagust W., Donohue M., Shaw L.M. Diagnostic accuracy of CSF Ab42 and florbetapir PET for Alzheimer's disease. Ann Clin Transl Neurol. 2014;1:534–543.
    1. Rosen C., Hansson O., Blennow K., Zetterberg H. Fluid biomarkers in Alzheimer's disease - current concepts. Mol Neurodegener. 2013;8:20.
    1. Rosen C., Rosen H., Andreasson U., Bremell D., Bremler R., Hagberg L. Cerebrospinal fluid biomarkers in cardiac arrest survivors. Resuscitation. 2014;85:227–232.
    1. Rosen C., Zetterberg H. Cerebrospinal fluid biomarkers for pathological processes in Alzheimer's disease. Curr Opin Psychiatry. 2013;26:276–282.
    1. Okamura N., Harada R., Furumoto S., Arai H., Yanai K., Kudo Y. Tau PET imaging in Alzheimer's disease. Curr Neurol Neurosci Rep. 2014;14:500.
    1. Fiandaca M.S., Kapogiannis D., Mapstone M., Boxer A., Eitan E., Schwartz J.B. Identification of preclinical Alzheimer's disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement. 2015;11:600–607.e1.
    1. Kapogiannis D., Boxer A., Schwartz J.B., Abner E.L., Biragyn A., Masharani U. Dysfunctionally phosphorylated type 1 insulin receptor substrate in neural-derived blood exosomes of preclinical Alzheimer's disease. FASEB J. 2015;29:589–596.
    1. Goetzl E.J., Boxer A., Schwartz J.B., Abner E.L., Petersen R.C., Miller B.L. Low neural exosomal levels of cellular survival factors in Alzheimer's disease. Ann Clin Transl Neurol. 2015;2:769–773.
    1. Goetzl E.J., Boxer A., Schwartz J.B., Abner E.L., Petersen R.C., Miller B.L. Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease. Neurology. 2015;85:40–47.
    1. Simons M., Raposo G. Exosomes–vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21:575–581.
    1. Théry C., Amigorena S., Raposo G., Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;Chapter 3:Unit 3.22.
    1. Rajendran L., Honsho M., Zahn T.R., Keller P., Geiger K.D., Verkade P. Alzheimer's disease beta-amyloid peptides are released in association with exosomes. Proc Natl Acad Sci U S A. 2006;103:11172–11177.
    1. Budnik V., Ruiz-Cañada C., Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17:160–172.
    1. Vingtdeux V., Hamdane M., Loyens A., Gele P., Drobeck H., Begard S. Alkalizing drugs induce accumulation of amyloid precursor protein by-products in luminal vesicles of multivesicular bodies. J Biol Chem. 2007;282:18197–18205.
    1. Davidsson P., Blennow K. Neurochemical dissection of synaptic pathology in Alzheimer's disease. Int Psychogeriatr. 1998;10:11–23.
    1. Kester M.I., Teunissen C.E., Crimmins D.L., Herries E.M., Ladenson J.H., Scheltens P. Neurogranin as a Cerebrospinal Fluid Biomarker for Synaptic Loss in Symptomatic Alzheimer Disease. JAMA Neurol. 2015;72:1275–1280.
    1. Portelius E., Zetterberg H., Skillbäck T., Törnqvist U., Andreasson U., Trojanowski J.Q. Cerebrospinal fluid neurogranin: relation to cognition and neurodegeneration in Alzheimer's disease. Brain. 2015;138:3373–3385.
    1. Ingelsson M., Fukumoto H., Newell K.L., Growdon J.H., Hedley-Whyte E.T., Frosch M.P. Early Abeta accumulation and progressive synaptic loss, gliosis, and tangle formation in AD brain. Neurology. 2004;62:925–931.
    1. Terry R.D., Masliah E., Salmon D.P., Butters N., DeTeresa R., Hill R. Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–580.
    1. Chang J.W., Schumacher E., Coulter P.M., Vinters H.V., Watson J.B. Dendritic translocation of RC3/neurogranin mRNA in normal aging, Alzheimer disease and fronto-temporal dementia. J Neuropathol Exp Neurol. 1997;56:1105–1118.
    1. Kvartsberg H., Duits F.H., Ingelsson M., Andreasen N., Öhrfelt A., Andersson K. Cerebrospinal fluid levels of the synaptic protein neurogranin correlates with cognitive decline in prodromal Alzheimer's disease. Alzheimers Dement. 2015;11:1180–1190.
    1. Chong J.A., Tapia-Ramírez J., Kim S., Toledo-Aral J.J., Zheng Y., Boutros M.C. REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons. Cell. 1995;80:949–957.
    1. Coulson J.M. Transcriptional regulation: cancer, neurons and the REST. Curr Biol. 2005;15:R665–R668.
    1. Lu T., Aron L., Zullo J., Pan Y., Kim H., Chen Y. REST and stress resistance in ageing and Alzheimer's disease. Nature. 2014;507:448–454.
    1. Burghardt R.C., Droleskey R. Transmission electron microscopy. Curr Protoc Microbiol. 2006;Chapter 2:Unit 2B.1.
    1. Mitsuhashi M., Taub D.D., Kapogiannis D., Eitan E., Zukley L., Mattson M.P. Aging enhances release of exosomal cytokine mRNAs by Abeta1-42-stimulated macrophages. FASEB J. 2013;27:5141–5150.
    1. Campbell S.N., Zhang C., Monte L., Roe A.D., Rice K.C., Tache Y. Increased tau phosphorylation and aggregation in the hippocampus of mice overexpressing corticotropin-releasing factor. J Alzheimers Dis. 2015;43:967–976.
    1. Shu S.Y., Ju G., Fan L.Z. The glucose oxidase-DAB-nickel method in peroxidase histochemistry of the nervous system. Neurosci Lett. 1988;85:169–171.
    1. van der Pol E., Coumans F.A., Grootemaat A.E., Gardiner C., Sargent I.L., Harrison P. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J Thromb Haemost. 2014;12:1182–1192.
    1. Laske C., Sohrabi H.R., Frost S.M., López-de-Ipiña K., Garrard P., Buscema M. Innovative diagnostic tools for early detection of Alzheimer's disease. Alzheimers Dement. 2015;11:561–578.
    1. Ray S., Britschgi M., Herbert C., Takeda-Uchimura Y., Boxer A., Blennow K. Classification and prediction of clinical Alzheimer's diagnosis based on plasma signaling proteins. Nat Med. 2007;13:1359–1362.
    1. Soares H.D., Chen Y., Sabbagh M., Roher A., Rohrer A., Schrijvers E. Identifying early markers of Alzheimer's disease using quantitative multiplex proteomic immunoassay panels. Ann N Y Acad Sci. 2009;1180:56–67.
    1. Sparks D.L., Kryscio R.J., Sabbagh M.N., Ziolkowski C., Lin Y., Sparks L.M. Tau is reduced in AD plasma and validation of employed ELISA methods. Am J Neurodegener Dis. 2012;1:99–106.
    1. Alvarez-Erviti L., Seow Y., Schapira A.H., Gardiner C., Sargent I.L., Wood M.J. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol Dis. 2011;42:360–367.
    1. Shi M., Liu C., Cook T.J., Bullock K.M., Zhao Y., Ginghina C. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson's disease. Acta Neuropathol. 2014;128:639–650.
    1. Fevrier B., Vilette D., Archer F., Loew D., Faigle W., Vidal M. Cells release prions in association with exosomes. Proc Natl Acad Sci U S A. 2004;101:9683–9688.
    1. Grad L.I., Pokrishevsky E., Silverman J.M., Cashman N.R. Exosome-dependent and independent mechanisms are involved in prion-like transmission of propagated Cu/Zn superoxide dismutase misfolding. Prion. 2014;8:331–335.
    1. Stern R.A., Tripodis Y., Baugh C.M., Fritts N.G., Martin B.M., Chaisson C. Preliminary Study of Plasma Exosomal Tau as a Potential Biomarker for Chronic Traumatic Encephalopathy. J Alzheimers Dis. 2016;10:1099–1109.

Source: PubMed

3
購読する