Skip to main content
Top
Published in: Alzheimer's Research & Therapy 1/2023

Open Access 01-12-2023 | Alzheimer's Disease | Research

APOE ε4 gene dose effect on imaging and blood biomarkers of neuroinflammation and beta-amyloid in cognitively unimpaired elderly

Authors: Anniina Snellman, Laura L. Ekblad, Jouni Tuisku, Mikko Koivumäki, Nicholas J. Ashton, Juan Lantero-Rodriguez, Thomas K. Karikari, Semi Helin, Marco Bucci, Eliisa Löyttyniemi, Riitta Parkkola, Mira Karrasch, Michael Schöll, Henrik Zetterberg, Kaj Blennow, Juha O. Rinne

Published in: Alzheimer's Research & Therapy | Issue 1/2023

Login to get access

Abstract

Background

Neuroinflammation, characterized by increased reactivity of microglia and astrocytes in the brain, is known to be present at various stages of the Alzheimer’s disease (AD) continuum. However, its presence and relationship with amyloid pathology in cognitively normal at-risk individuals is less clear. Here, we used positron emission tomography (PET) and blood biomarker measurements to examine differences in neuroinflammation and beta-amyloid (Aβ) and their association in cognitively unimpaired homozygotes, heterozygotes, or non-carriers of the APOE ε4 allele, the strongest genetic risk for sporadic AD.

Methods

Sixty 60–75-year-old APOE ε4 homozygotes (n = 19), heterozygotes (n = 21), and non-carriers (n = 20) were recruited in collaboration with the local Auria biobank. The participants underwent 11C-PK11195 PET (targeting 18-kDa translocator protein, TSPO), 11C-PiB PET (targeting Aβ), brain MRI, and neuropsychological testing including a preclinical cognitive composite (APCC). 11C-PK11195 distribution volume ratios and 11C-PiB standardized uptake value ratios (SUVRs) were calculated for regions typical for early Aβ accumulation in AD. Blood samples were drawn for measuring plasma glial fibrillary acidic protein (GFAP) and plasma Aβ1-42/1.40.

Results

In our cognitively unimpaired sample, cortical 11C-PiB-binding increased according to APOE ε4 gene dose (median composite SUVR 1.47 (range 1.38–1.66) in non-carriers, 1.55 (1.43–2.02) in heterozygotes, and 2.13 (1.61–2.83) in homozygotes, P = 0.002). In contrast, cortical composite 11C-PK11195-binding did not differ between the APOE ε4 gene doses (P = 0.27) or between Aβ-positive and Aβ-negative individuals (P = 0.81) and associated with higher Aβ burden only in APOE ε4 homozygotes (Rho = 0.47, P = 0.043). Plasma GFAP concentration correlated with cortical 11C-PiB (Rho = 0.35, P = 0.040), but not 11C-PK11195-binding (Rho = 0.13, P = 0.47) in Aβ-positive individuals. In the total cognitively unimpaired population, both higher composite 11C-PK11195-binding and plasma GFAP were associated with lower hippocampal volume, whereas elevated 11C-PiB-binding was associated with lower APCC scores.

Conclusions

Only Aβ burden measured by PET, but not markers of neuroinflammation, differed among cognitively unimpaired elderly with different APOE ε4 gene dose. However, APOE ε4 gene dose seemed to modulate the association between neuroinflammation and Aβ.
Appendix
Available only for authorised users
Literature
1.
go back to reference Gustavsson A, Norton N, Fast T, Frolich L, Georges J, Holzapfel D, et al. Global estimates on the number of persons across the Alzheimer’s disease continuum. Alzheimers Dement. 2022;19(2):658-70. https://doi.org/10.1002/alz.12694. Epub 2022 Jun 2. Gustavsson A, Norton N, Fast T, Frolich L, Georges J, Holzapfel D, et al. Global estimates on the number of persons across the Alzheimer’s disease continuum. Alzheimers Dement. 2022;19(2):658-70. https://​doi.​org/​10.​1002/​alz.​12694. Epub 2022 Jun 2.
2.
go back to reference Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405.PubMedPubMedCentralCrossRef Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405.PubMedPubMedCentralCrossRef
3.
go back to reference Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17(3):157–72.PubMedCrossRef Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17(3):157–72.PubMedCrossRef
4.
go back to reference Fan Z, Brooks DJ, Okello A, Edison P. An early and late peak in microglial activation in Alzheimer’s disease trajectory. Brain. 2017;140(3):792–803.PubMedPubMedCentral Fan Z, Brooks DJ, Okello A, Edison P. An early and late peak in microglial activation in Alzheimer’s disease trajectory. Brain. 2017;140(3):792–803.PubMedPubMedCentral
5.
go back to reference Albrecht DS, Sagare A, Pachicano M, Sweeney MD, Toga A, Zlokovic B, et al. Early neuroinflammation is associated with lower amyloid and tau levels in cognitively normal older adults. Brain Behav Immun. 2021;94:299–307.PubMedPubMedCentralCrossRef Albrecht DS, Sagare A, Pachicano M, Sweeney MD, Toga A, Zlokovic B, et al. Early neuroinflammation is associated with lower amyloid and tau levels in cognitively normal older adults. Brain Behav Immun. 2021;94:299–307.PubMedPubMedCentralCrossRef
7.
go back to reference Hamelin L, Lagarde J, Dorothee G, Leroy C, Labit M, Comley RA, et al. Early and protective microglial activation in Alzheimer’s disease: a prospective study using 18F-DPA-714 PET imaging. Brain. 2016;139(Pt 4):1252–64.PubMedCrossRef Hamelin L, Lagarde J, Dorothee G, Leroy C, Labit M, Comley RA, et al. Early and protective microglial activation in Alzheimer’s disease: a prospective study using 18F-DPA-714 PET imaging. Brain. 2016;139(Pt 4):1252–64.PubMedCrossRef
8.
go back to reference Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261(5123):921–3.PubMedCrossRef Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261(5123):921–3.PubMedCrossRef
9.
go back to reference Reiman EM, Chen K, Liu X, Bandy D, Yu M, Lee W, et al. Fibrillar amyloid-beta burden in cognitively normal people at 3 levels of genetic risk for Alzheimer’s disease. Proc Natl Acad Sci U S A. 2009;106(16):6820–5.PubMedPubMedCentralCrossRef Reiman EM, Chen K, Liu X, Bandy D, Yu M, Lee W, et al. Fibrillar amyloid-beta burden in cognitively normal people at 3 levels of genetic risk for Alzheimer’s disease. Proc Natl Acad Sci U S A. 2009;106(16):6820–5.PubMedPubMedCentralCrossRef
10.
go back to reference Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, et al. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci Transl Med. 2011;3(89):89ra57.PubMedPubMedCentralCrossRef Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, et al. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci Transl Med. 2011;3(89):89ra57.PubMedPubMedCentralCrossRef
11.
go back to reference Mecca AP, Barcelos NM, Wang S, Bruck A, Nabulsi N, Planeta-Wilson B, et al. Cortical beta-amyloid burden, gray matter, and memory in adults at varying APOE epsilon4 risk for Alzheimer’s disease. Neurobiol Aging. 2018;61:207–14.PubMedCrossRef Mecca AP, Barcelos NM, Wang S, Bruck A, Nabulsi N, Planeta-Wilson B, et al. Cortical beta-amyloid burden, gray matter, and memory in adults at varying APOE epsilon4 risk for Alzheimer’s disease. Neurobiol Aging. 2018;61:207–14.PubMedCrossRef
12.
go back to reference Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, et al. Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med. 2004;10(7):719–26.PubMedCrossRef Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, et al. Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med. 2004;10(7):719–26.PubMedCrossRef
13.
go back to reference Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, et al. ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc Natl Acad Sci U S A. 2013;110(19):E1807–16.PubMedPubMedCentralCrossRef Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, et al. ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc Natl Acad Sci U S A. 2013;110(19):E1807–16.PubMedPubMedCentralCrossRef
14.
go back to reference Egensperger R, Kosel S, von Eitzen U, Graeber MB. Microglial activation in Alzheimer disease: association with APOE genotype. Brain Pathol. 1998;8(3):439–47.PubMedCrossRef Egensperger R, Kosel S, von Eitzen U, Graeber MB. Microglial activation in Alzheimer disease: association with APOE genotype. Brain Pathol. 1998;8(3):439–47.PubMedCrossRef
15.
go back to reference Rodriguez GA, Tai LM, LaDu MJ, Rebeck GW. Human APOE4 increases microglia reactivity at Abeta plaques in a mouse model of Abeta deposition. J Neuroinflammation. 2014;11:111.PubMedPubMedCentralCrossRef Rodriguez GA, Tai LM, LaDu MJ, Rebeck GW. Human APOE4 increases microglia reactivity at Abeta plaques in a mouse model of Abeta deposition. J Neuroinflammation. 2014;11:111.PubMedPubMedCentralCrossRef
16.
go back to reference Cosenza-Nashat M, Zhao ML, Suh HS, Morgan J, Natividad R, Morgello S, et al. Expression of the translocator protein of 18 kDa by microglia, macrophages and astrocytes based on immunohistochemical localization in abnormal human brain. Neuropathol Appl Neurobiol. 2009;35(3):306–28.PubMedCrossRef Cosenza-Nashat M, Zhao ML, Suh HS, Morgan J, Natividad R, Morgello S, et al. Expression of the translocator protein of 18 kDa by microglia, macrophages and astrocytes based on immunohistochemical localization in abnormal human brain. Neuropathol Appl Neurobiol. 2009;35(3):306–28.PubMedCrossRef
17.
go back to reference Nutma E, Fancy N, Weinert M, Marzin MC, Tsartsalis S, Muirhead RCJ, et al. Translocator protein is a marker of activated microglia in rodent models but not human neurodegenerative diseases bioRxiv preprint. 2022. Nutma E, Fancy N, Weinert M, Marzin MC, Tsartsalis S, Muirhead RCJ, et al. Translocator protein is a marker of activated microglia in rodent models but not human neurodegenerative diseases bioRxiv preprint. 2022.
18.
go back to reference Tournier BB, Tsartsalis S, Ceyzeriat K, Fraser BH, Gregoire MC, Kovari E, et al. Astrocytic TSPO upregulation appears before microglial TSPO in Alzheimer’s disease. J Alzheimers Dis. 2020;77(3):1043–56.PubMedPubMedCentralCrossRef Tournier BB, Tsartsalis S, Ceyzeriat K, Fraser BH, Gregoire MC, Kovari E, et al. Astrocytic TSPO upregulation appears before microglial TSPO in Alzheimer’s disease. J Alzheimers Dis. 2020;77(3):1043–56.PubMedPubMedCentralCrossRef
19.
go back to reference Venneti S, Lopresti BJ, Wang G, Hamilton RL, Mathis CA, Klunk WE, et al. PK11195 labels activated microglia in Alzheimer’s disease and in vivo in a mouse model using PET. Neurobiol Aging. 2009;30(8):1217–26.PubMedCrossRef Venneti S, Lopresti BJ, Wang G, Hamilton RL, Mathis CA, Klunk WE, et al. PK11195 labels activated microglia in Alzheimer’s disease and in vivo in a mouse model using PET. Neurobiol Aging. 2009;30(8):1217–26.PubMedCrossRef
20.
go back to reference Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer FE, et al. In-vivo measurement of activated microglia in dementia. Lancet. 2001;358(9280):461–7.PubMedCrossRef Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer FE, et al. In-vivo measurement of activated microglia in dementia. Lancet. 2001;358(9280):461–7.PubMedCrossRef
21.
go back to reference Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer FE, et al. Microglia, amyloid, and cognition in Alzheimer’s disease: An [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol Dis. 2008;32(3):412–9.PubMedCrossRef Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer FE, et al. Microglia, amyloid, and cognition in Alzheimer’s disease: An [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol Dis. 2008;32(3):412–9.PubMedCrossRef
22.
go back to reference Fan Z, Okello AA, Brooks DJ, Edison P. Longitudinal influence of microglial activation and amyloid on neuronal function in Alzheimer’s disease. Brain. 2015;138(Pt 12):3685–98.PubMedCrossRef Fan Z, Okello AA, Brooks DJ, Edison P. Longitudinal influence of microglial activation and amyloid on neuronal function in Alzheimer’s disease. Brain. 2015;138(Pt 12):3685–98.PubMedCrossRef
23.
go back to reference Femminella GD, Ninan S, Atkinson R, Fan Z, Brooks DJ, Edison P. Does microglial activation influence hippocampal volume and neuronal function in Alzheimer’s disease and Parkinson’s disease dementia? J Alzheimers Dis. 2016;51(4):1275–89.PubMedCrossRef Femminella GD, Ninan S, Atkinson R, Fan Z, Brooks DJ, Edison P. Does microglial activation influence hippocampal volume and neuronal function in Alzheimer’s disease and Parkinson’s disease dementia? J Alzheimers Dis. 2016;51(4):1275–89.PubMedCrossRef
24.
go back to reference Okello A, Edison P, Archer HA, Turkheimer FE, Kennedy J, Bullock R, et al. Microglial activation and amyloid deposition in mild cognitive impairment: a PET study. Neurology. 2009;72(1):56–62.PubMedPubMedCentralCrossRef Okello A, Edison P, Archer HA, Turkheimer FE, Kennedy J, Bullock R, et al. Microglial activation and amyloid deposition in mild cognitive impairment: a PET study. Neurology. 2009;72(1):56–62.PubMedPubMedCentralCrossRef
25.
go back to reference Parbo P, Ismail R, Hansen KV, Amidi A, Marup FH, Gottrup H, et al. Brain inflammation accompanies amyloid in the majority of mild cognitive impairment cases due to Alzheimer’s disease. Brain. 2017;140(7):2002–11.PubMedCrossRef Parbo P, Ismail R, Hansen KV, Amidi A, Marup FH, Gottrup H, et al. Brain inflammation accompanies amyloid in the majority of mild cognitive impairment cases due to Alzheimer’s disease. Brain. 2017;140(7):2002–11.PubMedCrossRef
26.
go back to reference Zou J, Tao S, Johnson A, Tomljanovic Z, Polly K, Klein J, et al. Microglial activation, but not tau pathology, is independently associated with amyloid positivity and memory impairment. Neurobiol Aging. 2020;85:11–21.PubMedCrossRef Zou J, Tao S, Johnson A, Tomljanovic Z, Polly K, Klein J, et al. Microglial activation, but not tau pathology, is independently associated with amyloid positivity and memory impairment. Neurobiol Aging. 2020;85:11–21.PubMedCrossRef
27.
go back to reference Wiley CA, Lopresti BJ, Venneti S, Price J, Klunk WE, DeKosky ST, et al. Carbon 11-labeled Pittsburgh Compound B and carbon 11-labeled (R)-PK11195 positron emission tomographic imaging in Alzheimer disease. Arch Neurol. 2009;66(1):60–7.PubMedPubMedCentralCrossRef Wiley CA, Lopresti BJ, Venneti S, Price J, Klunk WE, DeKosky ST, et al. Carbon 11-labeled Pittsburgh Compound B and carbon 11-labeled (R)-PK11195 positron emission tomographic imaging in Alzheimer disease. Arch Neurol. 2009;66(1):60–7.PubMedPubMedCentralCrossRef
28.
go back to reference Schuitemaker A, Kropholler MA, Boellaard R, van der Flier WM, Kloet RW, van der Doef TF, et al. Microglial activation in Alzheimer’s disease: an (R)-[(1)(1)C]PK11195 positron emission tomography study. Neurobiol Aging. 2013;34(1):128–36.PubMedCrossRef Schuitemaker A, Kropholler MA, Boellaard R, van der Flier WM, Kloet RW, van der Doef TF, et al. Microglial activation in Alzheimer’s disease: an (R)-[(1)(1)C]PK11195 positron emission tomography study. Neurobiol Aging. 2013;34(1):128–36.PubMedCrossRef
29.
go back to reference Knezevic D, Verhoeff NPL, Hafizi S, Strafella AP, Graff-Guerrero A, Rajji T, et al. Imaging microglial activation and amyloid burden in amnestic mild cognitive impairment. J Cereb Blood Flow Metab. 2018;38(11):1885–95.PubMedCrossRef Knezevic D, Verhoeff NPL, Hafizi S, Strafella AP, Graff-Guerrero A, Rajji T, et al. Imaging microglial activation and amyloid burden in amnestic mild cognitive impairment. J Cereb Blood Flow Metab. 2018;38(11):1885–95.PubMedCrossRef
30.
go back to reference Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simren J, Montoliu-Gaya L, et al. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer’s disease. J Intern Med. 2021;290(3):583-601. https://doi.org/10.1111/joim.13332. Epub 2021 Jun 26. Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simren J, Montoliu-Gaya L, et al. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer’s disease. J Intern Med. 2021;290(3):583-601. https://​doi.​org/​10.​1111/​joim.​13332. Epub 2021 Jun 26.
31.
go back to reference Zetterberg H, Schott JM. Blood biomarkers for Alzheimer’s disease and related disorders. Acta Neurol Scand. 2022;146(1):51–5.PubMedCrossRef Zetterberg H, Schott JM. Blood biomarkers for Alzheimer’s disease and related disorders. Acta Neurol Scand. 2022;146(1):51–5.PubMedCrossRef
33.
go back to reference Cicognola C, Janelidze S, Hertze J, Zetterberg H, Blennow K, Mattsson-Carlgren N, et al. Plasma glial fibrillary acidic protein detects Alzheimer pathology and predicts future conversion to Alzheimer dementia in patients with mild cognitive impairment. Alzheimers Res Ther. 2021;13(1):68.PubMedPubMedCentralCrossRef Cicognola C, Janelidze S, Hertze J, Zetterberg H, Blennow K, Mattsson-Carlgren N, et al. Plasma glial fibrillary acidic protein detects Alzheimer pathology and predicts future conversion to Alzheimer dementia in patients with mild cognitive impairment. Alzheimers Res Ther. 2021;13(1):68.PubMedPubMedCentralCrossRef
34.
go back to reference Benedet AL, Mila-Aloma M, Vrillon A, Ashton NJ, Pascoal TA, Lussier F, et al. Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer disease continuum. JAMA Neurol. 2021;78(12):1471-83. Benedet AL, Mila-Aloma M, Vrillon A, Ashton NJ, Pascoal TA, Lussier F, et al. Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer disease continuum. JAMA Neurol. 2021;78(12):1471-83.
35.
go back to reference Jack CR Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535–62.PubMedPubMedCentralCrossRef Jack CR Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535–62.PubMedPubMedCentralCrossRef
36.
go back to reference Snellman A, Ekblad LL, Koivumaki M, Lindgren N, Tuisku J, Perala M, et al. ASIC-E4: interplay of beta-amyloid, synaptic density and neuroinflammation in cognitively normal volunteers with three levels of genetic risk for late-onset Alzheimer’s disease - study protocol and baseline characteristics. Front Neurol. 2022;13:826423.PubMedPubMedCentralCrossRef Snellman A, Ekblad LL, Koivumaki M, Lindgren N, Tuisku J, Perala M, et al. ASIC-E4: interplay of beta-amyloid, synaptic density and neuroinflammation in cognitively normal volunteers with three levels of genetic risk for late-onset Alzheimer’s disease - study protocol and baseline characteristics. Front Neurol. 2022;13:826423.PubMedPubMedCentralCrossRef
37.
go back to reference Karjalainen T, Tuisku J, Santavirta S, Kantonen T, Bucci M, Tuominen L, et al. Magia: robust automated image processing and kinetic modeling toolbox for PET neuroinformatics. Front Neuroinform. 2020;14:3.PubMedPubMedCentralCrossRef Karjalainen T, Tuisku J, Santavirta S, Kantonen T, Bucci M, Tuominen L, et al. Magia: robust automated image processing and kinetic modeling toolbox for PET neuroinformatics. Front Neuroinform. 2020;14:3.PubMedPubMedCentralCrossRef
38.
go back to reference Turkheimer FE, Edison P, Pavese N, Roncaroli F, Anderson AN, Hammers A, et al. Reference and target region modeling of [11C]-(R)-PK11195 brain studies. J Nucl Med. 2007;48(1):158–67.PubMed Turkheimer FE, Edison P, Pavese N, Roncaroli F, Anderson AN, Hammers A, et al. Reference and target region modeling of [11C]-(R)-PK11195 brain studies. J Nucl Med. 2007;48(1):158–67.PubMed
39.
go back to reference Yaqub M, van Berckel BN, Schuitemaker A, Hinz R, Turkheimer FE, Tomasi G, et al. Optimization of supervised cluster analysis for extracting reference tissue input curves in (R)-[(11)C]PK11195 brain PET studies. J Cereb Blood Flow Metab. 2012;32(8):1600–8.PubMedPubMedCentralCrossRef Yaqub M, van Berckel BN, Schuitemaker A, Hinz R, Turkheimer FE, Tomasi G, et al. Optimization of supervised cluster analysis for extracting reference tissue input curves in (R)-[(11)C]PK11195 brain PET studies. J Cereb Blood Flow Metab. 2012;32(8):1600–8.PubMedPubMedCentralCrossRef
40.
go back to reference Gunn RN, Lammertsma AA, Hume SP, Cunningham VJ. Parametric imaging of ligand-receptor binding in PET using a simplified reference region model. Neuroimage. 1997;6(4):279–87.PubMedCrossRef Gunn RN, Lammertsma AA, Hume SP, Cunningham VJ. Parametric imaging of ligand-receptor binding in PET using a simplified reference region model. Neuroimage. 1997;6(4):279–87.PubMedCrossRef
41.
go back to reference Gonzalez-Escamilla G, Lange C, Teipel S, Buchert R, Grothe MJ, Alzheimer’s Disease Neuroimaging I. PETPVE12: an SPM toolbox for Partial Volume Effects correction in brain PET - application to amyloid imaging with AV45-PET. Neuroimage. 2017;147:669–77.PubMedCrossRef Gonzalez-Escamilla G, Lange C, Teipel S, Buchert R, Grothe MJ, Alzheimer’s Disease Neuroimaging I. PETPVE12: an SPM toolbox for Partial Volume Effects correction in brain PET - application to amyloid imaging with AV45-PET. Neuroimage. 2017;147:669–77.PubMedCrossRef
42.
go back to reference Scholl M, Lockhart SN, Schonhaut DR, O’Neil JP, Janabi M, Ossenkoppele R, et al. PET imaging of Tau deposition in the aging human brain. Neuron. 2016;89(5):971–82.PubMedPubMedCentralCrossRef Scholl M, Lockhart SN, Schonhaut DR, O’Neil JP, Janabi M, Ossenkoppele R, et al. PET imaging of Tau deposition in the aging human brain. Neuron. 2016;89(5):971–82.PubMedPubMedCentralCrossRef
43.
go back to reference Jack CR Jr, Lowe VJ, Senjem ML, Weigand SD, Kemp BJ, Shiung MM, et al. 11C PiB and structural MRI provide complementary information in imaging of Alzheimer’s disease and amnestic mild cognitive impairment. Brain. 2008;131(Pt 3):665–80.PubMedCrossRef Jack CR Jr, Lowe VJ, Senjem ML, Weigand SD, Kemp BJ, Shiung MM, et al. 11C PiB and structural MRI provide complementary information in imaging of Alzheimer’s disease and amnestic mild cognitive impairment. Brain. 2008;131(Pt 3):665–80.PubMedCrossRef
44.
go back to reference Rowe CC, Ellis KA, Rimajova M, Bourgeat P, Pike KE, Jones G, et al. Amyloid imaging results from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging. Neurobiol Aging. 2010;31(8):1275–83.PubMedCrossRef Rowe CC, Ellis KA, Rimajova M, Bourgeat P, Pike KE, Jones G, et al. Amyloid imaging results from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging. Neurobiol Aging. 2010;31(8):1275–83.PubMedCrossRef
45.
go back to reference Koikkalainen J, Rhodius-Meester H, Tolonen A, Barkhof F, Tijms B, Lemstra AW, et al. Differential diagnosis of neurodegenerative diseases using structural MRI data. Neuroimage Clin. 2016;11:435–49.PubMedPubMedCentralCrossRef Koikkalainen J, Rhodius-Meester H, Tolonen A, Barkhof F, Tijms B, Lemstra AW, et al. Differential diagnosis of neurodegenerative diseases using structural MRI data. Neuroimage Clin. 2016;11:435–49.PubMedPubMedCentralCrossRef
46.
go back to reference Lotjonen J, Wolz R, Koikkalainen J, Julkunen V, Thurfjell L, Lundqvist R, et al. Fast and robust extraction of hippocampus from MR images for diagnostics of Alzheimer’s disease. Neuroimage. 2011;56(1):185–96.PubMedCrossRef Lotjonen J, Wolz R, Koikkalainen J, Julkunen V, Thurfjell L, Lundqvist R, et al. Fast and robust extraction of hippocampus from MR images for diagnostics of Alzheimer’s disease. Neuroimage. 2011;56(1):185–96.PubMedCrossRef
47.
go back to reference Keshavan A, Pannee J, Karikari TK, Rodriguez JL, Ashton NJ, Nicholas JM, et al. Population-based blood screening for preclinical Alzheimer’s disease in a British birth cohort at age 70. Brain. 2021;144(2):434–49.PubMedPubMedCentral Keshavan A, Pannee J, Karikari TK, Rodriguez JL, Ashton NJ, Nicholas JM, et al. Population-based blood screening for preclinical Alzheimer’s disease in a British birth cohort at age 70. Brain. 2021;144(2):434–49.PubMedPubMedCentral
48.
go back to reference Meyer PF, Ashton NJ, Karikari TK, Strikwerda-Brown C, Kobe T, Gonneaud J, et al. Plasma p-tau231, p-tau181, PET biomarkers, and cognitive change in older adults. Ann Neurol. 2022;91(4):548–60.PubMedCrossRef Meyer PF, Ashton NJ, Karikari TK, Strikwerda-Brown C, Kobe T, Gonneaud J, et al. Plasma p-tau231, p-tau181, PET biomarkers, and cognitive change in older adults. Ann Neurol. 2022;91(4):548–60.PubMedCrossRef
49.
go back to reference Passamonti L, Rodriguez PV, Hong YT, Allinson KSJ, Bevan-Jones WR, Williamson D, et al. [(11)C]PK11195 binding in Alzheimer disease and progressive supranuclear palsy. Neurology. 2018;90(22):e1989–96.PubMedPubMedCentralCrossRef Passamonti L, Rodriguez PV, Hong YT, Allinson KSJ, Bevan-Jones WR, Williamson D, et al. [(11)C]PK11195 binding in Alzheimer disease and progressive supranuclear palsy. Neurology. 2018;90(22):e1989–96.PubMedPubMedCentralCrossRef
50.
go back to reference Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity. 2017;47(3):566-81e9.PubMedPubMedCentralCrossRef Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity. 2017;47(3):566-81e9.PubMedPubMedCentralCrossRef
51.
go back to reference Ulrich JD, Ulland TK, Mahan TE, Nystrom S, Nilsson KP, Song WM, et al. ApoE facilitates the microglial response to amyloid plaque pathology. J Exp Med. 2018;215(4):1047–58.PubMedPubMedCentralCrossRef Ulrich JD, Ulland TK, Mahan TE, Nystrom S, Nilsson KP, Song WM, et al. ApoE facilitates the microglial response to amyloid plaque pathology. J Exp Med. 2018;215(4):1047–58.PubMedPubMedCentralCrossRef
52.
go back to reference Nguyen AT, Wang K, Hu G, Wang X, Miao Z, Azevedo JA, et al. APOE and TREM2 regulate amyloid-responsive microglia in Alzheimer’s disease. Acta Neuropathol. 2020;140(4):477–93.PubMedPubMedCentralCrossRef Nguyen AT, Wang K, Hu G, Wang X, Miao Z, Azevedo JA, et al. APOE and TREM2 regulate amyloid-responsive microglia in Alzheimer’s disease. Acta Neuropathol. 2020;140(4):477–93.PubMedPubMedCentralCrossRef
53.
go back to reference Ghisays V, Goradia DD, Protas H, Bauer RJ 3rd, Devadas V, Tariot PN, et al. Brain imaging measurements of fibrillar amyloid-beta burden, paired helical filament tau burden, and atrophy in cognitively unimpaired persons with two, one, and no copies of the APOE epsilon4 allele. Alzheimers Dement. 2020;16(4):598–609.PubMedPubMedCentralCrossRef Ghisays V, Goradia DD, Protas H, Bauer RJ 3rd, Devadas V, Tariot PN, et al. Brain imaging measurements of fibrillar amyloid-beta burden, paired helical filament tau burden, and atrophy in cognitively unimpaired persons with two, one, and no copies of the APOE epsilon4 allele. Alzheimers Dement. 2020;16(4):598–609.PubMedPubMedCentralCrossRef
54.
go back to reference Ekblad LL, Johansson J, Helin S, Viitanen M, Laine H, Puukka P, et al. Midlife insulin resistance, APOE genotype, and late-life brain amyloid accumulation. Neurology. 2018;90(13):e1150–7.PubMedPubMedCentralCrossRef Ekblad LL, Johansson J, Helin S, Viitanen M, Laine H, Puukka P, et al. Midlife insulin resistance, APOE genotype, and late-life brain amyloid accumulation. Neurology. 2018;90(13):e1150–7.PubMedPubMedCentralCrossRef
55.
go back to reference Jansen WJ, Janssen O, Tijms BM, Vos SJB, Ossenkoppele R, Visser PJ, et al. Prevalence estimates of amyloid abnormality across the Alzheimer disease clinical spectrum. JAMA Neurol. 2022;79(3):228–43.PubMedCrossRef Jansen WJ, Janssen O, Tijms BM, Vos SJB, Ossenkoppele R, Visser PJ, et al. Prevalence estimates of amyloid abnormality across the Alzheimer disease clinical spectrum. JAMA Neurol. 2022;79(3):228–43.PubMedCrossRef
56.
go back to reference Toppala S, Ekblad LL, Tuisku J, Helin S, Johansson JJ, Laine H, et al. Association of early beta-amyloid accumulation and neuroinflammation measured with [(11)C]PBR28 in elderly individuals without dementia. Neurology. 2021;96(12):e1608–19.PubMedPubMedCentralCrossRef Toppala S, Ekblad LL, Tuisku J, Helin S, Johansson JJ, Laine H, et al. Association of early beta-amyloid accumulation and neuroinflammation measured with [(11)C]PBR28 in elderly individuals without dementia. Neurology. 2021;96(12):e1608–19.PubMedPubMedCentralCrossRef
57.
go back to reference Dani M, Wood M, Mizoguchi R, Fan Z, Walker Z, Morgan R, et al. Microglial activation correlates in vivo with both tau and amyloid in Alzheimer’s disease. Brain. 2018;141(9):2740–54.PubMed Dani M, Wood M, Mizoguchi R, Fan Z, Walker Z, Morgan R, et al. Microglial activation correlates in vivo with both tau and amyloid in Alzheimer’s disease. Brain. 2018;141(9):2740–54.PubMed
58.
go back to reference Kreisl WC, Lyoo CH, McGwier M, Snow J, Jenko KJ, Kimura N, et al. In vivo radioligand binding to translocator protein correlates with severity of Alzheimer’s disease. Brain. 2013;136(Pt 7):2228–38.PubMedPubMedCentralCrossRef Kreisl WC, Lyoo CH, McGwier M, Snow J, Jenko KJ, Kimura N, et al. In vivo radioligand binding to translocator protein correlates with severity of Alzheimer’s disease. Brain. 2013;136(Pt 7):2228–38.PubMedPubMedCentralCrossRef
59.
go back to reference Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, et al. Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27(9):1592–9.PubMedCrossRef Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, et al. Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27(9):1592–9.PubMedCrossRef
60.
go back to reference Chatterjee P, Pedrini S, Stoops E, Goozee K, Villemagne VL, Asih PR, et al. Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer’s disease. Transl Psychiatry. 2021;11(1):27.PubMedPubMedCentralCrossRef Chatterjee P, Pedrini S, Stoops E, Goozee K, Villemagne VL, Asih PR, et al. Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer’s disease. Transl Psychiatry. 2021;11(1):27.PubMedPubMedCentralCrossRef
Metadata
Title
APOE ε4 gene dose effect on imaging and blood biomarkers of neuroinflammation and beta-amyloid in cognitively unimpaired elderly
Authors
Anniina Snellman
Laura L. Ekblad
Jouni Tuisku
Mikko Koivumäki
Nicholas J. Ashton
Juan Lantero-Rodriguez
Thomas K. Karikari
Semi Helin
Marco Bucci
Eliisa Löyttyniemi
Riitta Parkkola
Mira Karrasch
Michael Schöll
Henrik Zetterberg
Kaj Blennow
Juha O. Rinne
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Alzheimer's Research & Therapy / Issue 1/2023
Electronic ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-023-01209-6

Other articles of this Issue 1/2023

Alzheimer's Research & Therapy 1/2023 Go to the issue