Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer's disease

Alex E Roher, Chera L Esh, Tyler A Kokjohn, Eduardo M Castaño, Gregory D Van Vickle, Walter M Kalback, R Lyle Patton, Dean C Luehrs, Ian D Daugs, Yu-Min Kuo, Mark R Emmerling, Holly Soares, Joseph F Quinn, Jeffrey Kaye, Donald J Connor, Nina B Silverberg, Charles H Adler, James D Seward, Thomas G Beach, Marwan N Sabbagh, Alex E Roher, Chera L Esh, Tyler A Kokjohn, Eduardo M Castaño, Gregory D Van Vickle, Walter M Kalback, R Lyle Patton, Dean C Luehrs, Ian D Daugs, Yu-Min Kuo, Mark R Emmerling, Holly Soares, Joseph F Quinn, Jeffrey Kaye, Donald J Connor, Nina B Silverberg, Charles H Adler, James D Seward, Thomas G Beach, Marwan N Sabbagh

Abstract

Background: We evaluated the amounts of amyloid beta (Abeta)) peptides in the central nervous system (CNS) and in reservoirs outside the CNS and their potential impact on Abeta plasma levels and Alzheimer's disease (AD) pathology.

Methods: Amyloid beta levels were measured in (1) the plasma of AD and nondemented (ND) controls in a longitudinal study, (2) the plasma of a cohort of AD patients receiving a cholinesterase inhibitor, and (3) the skeletal muscle, liver, aorta, platelets, leptomeningeal arteries, and in gray and white matter of AD and ND control subjects.

Results: Plasma Abeta levels fluctuated over time and among individuals, suggesting continuous contributions from brain and peripheral tissues and associations with reactive circulating proteins. Arteries with atherosclerosis had larger amounts of Abeta40 than disease-free vessels. Inactivated platelets contained more Abeta peptides than activated ones. Substantially more Abeta was present in liver samples from ND patients. Overall, AD brain and skeletal muscle contained increased levels of Abeta.

Conclusions: Efforts to use plasma levels of Abeta peptides as AD biomarkers or disease-staging scales have failed. Peripheral tissues might contribute to both the circulating amyloid pool and AD pathology within the brain and its vasculature. The wide spread of plasma Abeta values is also due in part to the ability of Abeta to bind to a variety of plasma and membrane proteins. Sources outside the CNS must be accounted for because pharmacologic interventions to reduce cerebral amyloid are assessed by monitoring Abeta plasma levels. Furthermore, the long-range impact of Abeta immunotherapy on peripheral Abeta sources should also be considered.

Conflict of interest statement

Conflicts of Interest: The authors do not have any conflicts of interests with the expection of Dr. Marwan N. Sabbagh who receives support from Pfizer, Novartis, Elan, Wyeth, Medivation and Lilly. He is on the speaker’s bureau for Eisai, Pfizer, Novartis and Forest and a consultant for Lilly.

Figures

Figure 1
Figure 1
Longitudinal comparison of AD (black) and ND control (white) plasma Aβ levels as measured by ELISA. Measurements were taken at baseline (T=0) and at 3, 6 and 12 months. The horizontal bars represent the mean values. A) Aβ40 pg/ml B) Aβ42 pg/ml.
Figure 2
Figure 2
Plasma Aβ levels measured by ELISA in the therapeutic study with donepezil. Measurements were taken at baseline and 12 weeks later (follow-up). A) Aβ40 pg/ml in donepezil-initiation group B) Aβ42 pg/ml in donepezil-initiation group C) Aβ40 pg/ml in stable-donepezil group D) Aβ42 pg/ml in stable-donepezil group.

References

    1. Hansson O, Zetterberg H, Vanmechelen E, Vanderstichele H, Andreasson U, Londos E, et al. Evaluation of plasma Abeta(40) and Abeta(42) as predictors of conversion to Alzheimer’s disease in patients with mild cognitive impairment. Neurobiol Aging. 2008
    1. Kuo YM, Kokjohn TA, Watson MD, Woods AS, Cotter RJ, Sue LI, et al. Elevated abeta42 in skeletal muscle of Alzheimer disease patients suggests peripheral alterations of AbetaPP metabolism. Am J Pathol. 2000;156:797–805.
    1. Li QX, Whyte S, Tanner JE, Evin G, Beyreuther K, Masters CL. Secretion of Alzheimer’s disease Abeta amyloid peptide by activated human platelets. Lab Invest. 1998;78:461–469.
    1. Van Nostrand WE, Melchor JP. Disruption of pathologic amyloid beta-protein fibril assembly on the surface of cultured human cerebrovascular smooth muscle cells. Amyloid. 2001;8(Suppl 1):20–27.
    1. Selkoe DJ, Podlisny MB, Joachim CL, Vickers EA, Lee G, Fritz LC, et al. Beta-amyloid precursor protein of Alzheimer disease occurs as 110- to 135-kilodalton membrane-associated proteins in neural and nonneural tissues. Proc Natl Acad Sci U S A. 1988;85:7341–7345.
    1. Catteruccia N, Willingale-Theune J, Bunke D, Prior R, Masters CL, Crisanti A, et al. Ultrastructural localization of the putative precursors of the A4 amyloid protein associated with Alzheimer’s disease. Am J Pathol. 1990;137:19–26.
    1. Sandbrink R, Masters CL, Beyreuther K. Beta A4-amyloid protein precursor mRNA isoforms without exon 15 are ubiquitously expressed in rat tissues including brain, but not in neurons. J Biol Chem. 1994;269:1510–1517.
    1. Biere AL, Ostaszewski B, Stimson ER, Hyman BT, Maggio JE, Selkoe DJ. Amyloid beta-peptide is transported on lipoproteins and albumin in human plasma. J Biol Chem. 1996;271:32916–32922.
    1. Kuo YM, Emmerling MR, Lampert HC, Hempelman SR, Kokjohn TA, Woods AS, et al. High levels of circulating Abeta42 are sequestered by plasma proteins in Alzheimer’s disease. Biochem Biophys Res Commun. 1999;257:787–791.
    1. Kuo YM, Kokjohn TA, Kalback W, Luehrs D, Galasko DR, Chevallier N, et al. Amyloid-beta peptides interact with plasma proteins and erythrocytes: implications for their quantitation in plasma. Biochem Biophys Res Commun. 2000;268:750–756.
    1. Beach TG, Sue LI, Walker DG, Roher AE, Lue L, Vedders L, et al. The Sun Health Research Institute Brain Donation Program: Description and Experience, 1987–2007. Cell Tissue Bank. 2008;9:229–245.
    1. De JC, Cras P, Vanderstichele H, Cruts M, Vanderhoeven I, Smouts I, et al. Evidence that Abeta42 plasma levels in presenilin-1 mutation carriers do not allow for prediction of their clinical phenotype. Neurobiol Dis. 1999;6:280–287.
    1. Roher AE, Weiss N, Kokjohn TA, Kuo YM, Kalback W, Anthony J, et al. Increased A beta peptides and reduced cholesterol and myelin proteins characterize white matter degeneration in Alzheimer’s disease. Biochemistry. 2002;41:11080–11090.
    1. Li QX, Evin G, Small DH, Multhaup G, Beyreuther K, Masters CL. Proteolytic processing of Alzheimer’s disease beta A4 amyloid precursor protein in human platelets. J Biol Chem. 1995;270:14140–14147.
    1. Basun H, Nilsberth C, Eckman C, Lannfelt L, Younkin S. Plasma levels of Abeta42 and Abeta40 in Alzheimer patients during treatment with the acetylcholinesterase inhibitor tacrine. Dement Geriatr Cogn Disord. 2002;14:156–160.
    1. Lopez OL, Kuller LH, Mehta PD, Becker JT, Gach HM, Sweet RA, et al. Plasma amyloid levels and the risk of AD in normal subjects in the Cardiovascular Health Study. Neurology. 2008;70:1664–1671.
    1. Watson DE, Roher AE, Kim KS, Spiegel K, Emmerling MR. Complement interactions with amyloid-β1-42: a nidus for inflammation in AD brains. Amyloid: International Journal of Experimental and Clinical Investigation. 1997;4:147–156.
    1. Pillot T, Goethals M, Najib J, Labeur C, Lins L, Chambaz J, et al. Beta-amyloid peptide interacts specifically with the carboxy-terminal domain of human apolipoprotein E: relevance to Alzheimer’s disease. J Neurochem. 1999;72:230–237.
    1. Munson GW, Roher AE, Kuo YM, Gilligan SM, Reardon CA, Getz GS, et al. SDS-stable complex formation between native apolipoprotein E3 and beta-amyloid peptides. Biochemistry. 2000;39:16119–16124.
    1. Kivipelto M, Helkala EL, Hanninen T, Laakso MP, Hallikainen M, Alhainen K, et al. Midlife vascular risk factors and late-life mild cognitive impairment: A population-based study. Neurology. 2001;56:1683–1689.
    1. Notkola IL, Sulkava R, Pekkanen J, Erkinjuntti T, Ehnholm C, Kivinen P, et al. Serum total cholesterol, apolipoprotein E epsilon 4 allele, and Alzheimer’s disease. Neuroepidemiology. 1998;17:14–20.
    1. Refolo LM, Malester B, LaFrancois J, Bryant-Thomas T, Wang R, Tint GS, et al. Hypercholesterolemia accelerates the Alzheimer’s amyloid pathology in a transgenic mouse model. Neurobiol Dis. 2000;7:321–331.
    1. Mori T, Paris D, Town T, Rojiani AM, Sparks DL, Delledonne A, et al. Cholesterol accumulates in senile plaques of Alzheimer disease patients and in transgenic APP(SW) mice. J Neuropathol Exp Neurol. 2001;60:778–785.
    1. Wolozin B, Bednar MM. Interventions for heart disease and their effects on Alzheimer’s disease. Neurol Res. 2006;28:630–636.
    1. Roher AE, Esh C, Kokjohn TA, Kalback W, Luehrs DC, Seward JD, et al. Circle of Willis atherosclerosis is a risk factor for sporadic Alzheimer’s disease. Arterioscler Thromb Vasc Biol. 2003;23:2055–2062.
    1. Beach TG, Wilson JR, Sue LI, Newell A, Poston M, Cisneros R, et al. Circle of Willis atherosclerosis: association with Alzheimer’s disease, neuritic plaques and neurofibrillary tangles. Acta Neuropathol. 2007;113:13–21.
    1. Kalback W, Esh C, Castano EM, Rahman A, Kokjohn T, Luehrs DC, et al. Atherosclerosis, vascular amyloidosis and brain hypoperfusion in the pathogenesis of sporadic Alzheimer’s disease. Neurol Res. 2004;26:525–539.
    1. Honig LS, Kukull W, Mayeux R. Atherosclerosis and AD: analysis of data from the US National Alzheimer’s Coordinating Center. Neurology. 2005;64:494–500.
    1. Ferreiro JA, Ansbacher LE, Vinters HV. Stroke related to cerebral amyloid angiopathy: the significance of systemic vascular disease. J Neurol. 1989;236:267–272.
    1. Grammas P, Ovase R. Inflammatory factors are elevated in brain microvessels in Alzheimer’s disease. Neurobiol Aging. 2001;22:837–842.
    1. Ridker PM, Libby P. Risk Factors for Atherothrombic Disease. In: Zipes DP, Libby P, Bonow RO, Braunwald E, editors. Braunwald’s Heart Disease. Philadelphia: Elsevier Saunders; 2005. pp. 939–958.
    1. Licastro F, Pedrini S, Caputo L, Annoni G, Davis LJ, Ferri C, et al. Increased plasma levels of interleukin-1, interleukin-6 and alpha-1-antichymotrypsin in patients with Alzheimer’s disease: peripheral inflammation or signals from the brain? J Neuroimmunol. 2000;103:97–102.
    1. Zuliani G, Ranzini M, Guerra G, Rossi L, Munari MR, Zurlo A, et al. Plasma cytokines profile in older subjects with late onset Alzheimer’s disease or vascular dementia. J Psychiatr Res. 2007;41:686–693.
    1. Thal DR, Ghebremedhin E, Orantes M, Wiestler OD. Vascular pathology in Alzheimer disease: correlation of cerebral amyloid angiopathy and arteriosclerosis/lipohyalinosis with cognitive decline. J Neuropathol Exp Neurol. 2003;62:1287–1301.
    1. Attems J, Lintner F, Jellinger KA. Amyloid beta peptide 1-42 highly correlates with capillary cerebral amyloid angiopathy and Alzheimer disease pathology. Acta Neuropathol. 2004;107:283–291.
    1. Sjogren M, Blennow K. The link between cholesterol and Alzheimer’s disease. World J Biol Psychiatry. 2005;6:85–97.
    1. Deane R, Zlokovic BV. Role of the blood-brain barrier in the pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2007;4:191–197.
    1. Ito S, Ohtsuki S, Kamiie J, Nezu Y, Terasaki T. Cerebral clearance of human amyloid-beta peptide (1-40) across the blood-brain barrier is reduced by self-aggregation and formation of low-density lipoprotein receptor-related protein-1 ligand complexes. J Neurochem. 2007;103:2482–2490.
    1. Wilhelmus MM, Otte-Holler I, van Triel JJ, Veerhuis R, Maat-Schieman ML, Bu G, et al. Lipoprotein receptor-related protein-1 mediates amyloid-beta-mediated cell death of cerebrovascular cells. Am J Pathol. 2007;171:1989–1999.
    1. Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al. Clearance of Alzheimer’s amyloid-ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest. 2000;106:1489–1499.
    1. Martel CL, Mackic JB, McComb JG, Ghiso J, Zlokovic BV. Blood -brain barrier uptake of the 40 and 42 amino acid sequences of circulating Alzheimer’s amyloid beta in guinea pigs. Neurosci Lett. 1996;206:157–160.
    1. Weller RO, Cohen NR, Nicoll JA. Cerebrovascular disease and the pathophysiology of Alzheimer’s disease. Implications for therapy. Panminerva Med. 2004;46:239–251.
    1. Preston SD, Steart PV, Wilkinson A, Nicoll JA, Weller RO. Capillary and arterial cerebral amyloid angiopathy in Alzheimer’s disease: defining the perivascular route for the elimination of amyloid beta from the human brain. Neuropathol Appl Neurobiol. 2003;29:106–117.
    1. Roher AE, Kuo YM, Esh C, Knebel C, Weiss N, Kalback W, et al. Cortical and leptomeningeal cerebrovascular amyloid and white matter pathology in Alzheimer’s disease. Mol Med. 2003;9:112–122.
    1. Chen M, Inestrosa NC, Ross GS, Fernandez HL. Platelets are the primary source of amyloid beta-peptide in human blood. Biochem Biophys Res Commun. 1995;213:96–103.
    1. Casoli T, Di Stefano G, Giorgetti B, Grossi Y, Balietti M, Fattoretti P, et al. Release of beta-amyloid from high-density platelets: implications for Alzheimer’s disease pathology. Ann N Y Acad Sci. 2007;1096:170–178.
    1. Shen MY, Hsiao G, Fong TH, Chen HM, Chou DS, Lin CH, et al. Amyloid beta peptide-activated signal pathways in human platelets. Eur J Pharmacol. 2008;588:259–266.
    1. Di LM, Pastorino L, Bianchetti A, Perez J, Vignolo LA, Lenzi GL, et al. Differential level of platelet amyloid beta precursor protein isoforms: an early marker for Alzheimer disease. Arch Neurol. 1998;55:1195–1200.
    1. Padovani A, Borroni B, Colciaghi F, Pettenati C, Cottini E, Agosti C, et al. Abnormalities in the pattern of platelet amyloid precursor protein forms in patients with mild cognitive impairment and Alzheimer disease. Arch Neurol. 2002;59:71–75.
    1. Liu HC, Wang HC, Ko SY, Wang PN, Chi CW, Hong CJ, et al. Correlation between platelet amyloid precursor protein isoform ratio and cognition in Alzheimer’s disease. J Alzheimers Dis. 2007;11:77–84.
    1. Van Nostrand WE, Schmaier AH, Farrow JS, Cines DB, Cunningham DD. Protease nexin-2/amyloid beta-protein precursor in blood is a platelet-specific protein. Biochem Biophys Res Commun. 1991;175:15–21.
    1. Xu F, Davis J, Miao J, Previti ML, Romanov G, Ziegler K, et al. Protease nexin-2/amyloid beta-protein precursor limits cerebral thrombosis. Proc Natl Acad Sci U S A. 2005;102:18135–18140.
    1. De Meyer GR, De Cleen DM, Cooper S, Knaapen MW, Jans DM, Martinet W, et al. Platelet phagocytosis and processing of beta-amyloid precursor protein as a mechanism of macrophage activation in atherosclerosis. Circ Res. 2002;90:1197–1204.
    1. Jans DM, Martinet W, Van De Parre TJ, Herman AG, Bult H, Kockx MM, et al. Processing of amyloid precursor protein as a biochemical link between atherosclerosis and Alzheimer’s disease. Cardiovasc Hematol Disord Drug Targets. 2006;6:21–34.
    1. Wisniewski HM, Frackowiak J, Mazur-Kolecka B. In vitro production of beta-amyloid in smooth muscle cells isolated from amyloid angiopathy-affected vessels. Neurosci Lett. 1995;183:120–123.
    1. Shi C, Guo K, Yew DT, Yao Z, Forster EL, Wang H, et al. Effects of ageing and Alzheimer’s disease on mitochondrial function of human platelets. Exp Gerontol. 2008;43:589–594.
    1. Franchini M. Hemostasis and aging. Crit Rev Oncol Hematol. 2006;60:144–151.
    1. Wilkerson WR, Sane DC. Aging and thrombosis. Semin Thromb Hemost. 2002;28:555–568.
    1. Borroni B, Akkawi N, Martini G, Colciaghi F, Prometti P, Rozzini L, et al. Microvascular damage and platelet abnormalities in early Alzheimer’s disease. J Neurol Sci. 2002;203–204:189–193.
    1. Schubert W, Prior R, Weidemann A, Dircksen H, Multhaup G, Masters CL, et al. Localization of Alzheimer beta A4 amyloid precursor protein at central and peripheral synaptic sites. Brain Res. 1991;563:184–194.
    1. Askanas V, Engel WK. Molecular pathology and pathogenesis of inclusion-body myositis. Microsc Res Tech. 2005;67:114–120.
    1. Finch CE. A perspective on sporadic inclusion-body myositis: the role of aging and inflammatory processes. Neurology. 2006;66:S1–S6.
    1. Querfurth HW, Suhara T, Rosen KM, McPhie DL, Fujio Y, Tejada G, et al. Beta-amyloid peptide expression is sufficient for myotube death: implications for human inclusion body myopathy. Mol Cell Neurosci. 2001;17:793–810.
    1. Turner AJ, Nalivaeva NN. New insights into the roles of metalloproteinases in neurodegeneration and neuroprotection. Int Rev Neurobiol. 2007;82:113–135.
    1. Ghiso J, Shayo M, Calero M, Ng D, Tomidokoro Y, Gandy S, et al. Systemic catabolism of Alzheimer’s Abeta40 and Abeta42. J Biol Chem. 2004;279:45897–45908.
    1. Ghiso J, Calero M, Matsubara E, Governale S, Chuba J, Beavis R, et al. Alzheimer’s soluble amyloid beta is a normal component of human urine. FEBS Lett. 1997;408:105–108.
    1. Tamaki C, Ohtsuki S, Terasaki T. Insulin facilitates the hepatic clearance of plasma amyloid beta-peptide (1 40) by intracellular translocation of low-density lipoprotein receptor-related protein 1 (LRP-1) to the plasma membrane in hepatocytes. Mol Pharmacol. 2007;72:850–855.
    1. Bateman RJ, Wen G, Morris JC, Holtzman DM. Fluctuations of CSF amyloid-beta levels: implications for a diagnostic and therapeutic biomarker. Neurology. 2007;68:666–669.
    1. Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia-Alloza M, de Calignon A, Rozkalne A, et al. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer’s disease. Nature. 2008;451:720–724.
    1. Hawkes CA, McLaurin J. Immunotherapy as treatment for Alzheimer’s disease. Expert Rev Neurother. 2007;7:1535–1548.
    1. Paris D, Townsend K, Quadros A, Humphrey J, Sun J, Brem S, et al. Inhibition of angiogenesis by Abeta peptides. Angiogenesis. 2004;7:75–85.
    1. Plant LD, Boyle JP, Smith IF, Peers C, Pearson HA. The production of amyloid beta peptide is a critical requirement for the viability of central neurons. J Neurosci. 2003;23:5531–5535.
    1. Pearson HA, Peers C. Physiological roles for amyloid beta peptides. J Physiol. 2006;575:5–10.
    1. Heininger K. A unifying hypothesis of Alzheimer’s disease. IV. Causation and sequence of events. Rev Neurosci. 2000;(11 Spec No):213–328.
    1. Kuo YM, Emmerling MR, Vigo-Pelfrey C, Kasunic TC, Kirkpatrick JB, Murdoch GH, et al. Water-soluble Abeta (N-40, N-42) oligomers in normal and Alzheimer disease brains. J Biol Chem. 1996;271:4077–4081.
    1. Roher AE, Chaney MO, Kuo YM, Webster SD, Stine WB, Haverkamp LJ, et al. Morphology and toxicity of Abeta-(1-42) dimer derived from neuritic and vascular amyloid deposits of Alzheimer’s disease. J Biol Chem. 1996;271:20631–20635.

Source: PubMed

3
Subscribe