Skip to main content
Top
Published in: BMC Neurology 1/2022

Open Access 01-12-2022 | Alzheimer's Disease | Research

Amyloid-β and APOE genotype predict memory decline in cognitively unimpaired older individuals independently of Alzheimer’s disease polygenic risk score

Authors: Jori Tomassen, Anouk den Braber, Sven J. van der Lee, Lianne M. Reus, Elles Konijnenberg, Stephen F. Carter, Maqsood Yaqub, Bart N.M. van Berckel, Lyduine E. Collij, Dorret I. Boomsma, Eco J.C. de Geus, Philip Scheltens, Karl Herholz, Betty M. Tijms, Pieter Jelle Visser

Published in: BMC Neurology | Issue 1/2022

Login to get access

Abstract

Background:

What combination of risk factors for Alzheimer’s disease (AD) are most predictive of cognitive decline in cognitively unimpaired individuals remains largely unclear. We studied associations between APOE genotype, AD-Polygenic Risk Scores (AD-PRS), amyloid-β pathology and decline in cognitive functioning over time in a large sample of cognitively unimpaired older individuals.

Methods:

We included 276 cognitively unimpaired older individuals (75 ± 10 years, 63% female) from the EMIF-AD PreclinAD cohort. An AD-PRS was calculated including 83 genome-wide significant variants. The APOE gene was not included in the PRS and was analyzed separately. Baseline amyloid-β status was assessed by visual read of [18F]flutemetamol-PET standardized uptake value images. At baseline and follow-up (2.0 ± 0.4 years), the cognitive domains of memory, attention, executive function, and language were measured. We used generalized estimating equations corrected for age, sex and center to examine associations between APOE genotype and AD-PRS with amyloid-β status. Linear mixed models corrected for age, sex, center and education were used to examine associations between APOE genotype, AD-PRS and amyloid-β status, and their interaction on changes in cognitive functioning over time.

Results:

Fifty-two participants (19%) had abnormal amyloid-β, and 84 participants (31%) carried at least one APOE ε4 allele. APOE genotype and AD-PRS were both associated with abnormal amyloid-β status. Increasingly more risk-full APOE genotype, a high AD-PRS and an abnormal amyloid-β status were associated with steeper decline in memory functioning in separate models (all p ≤ 0.02). A model including 4-way interaction term (APOE×AD-PRS×amyloid-β×time) was not significant. When modelled together, both APOE genotype and AD-PRS predicted steeper decline in memory functioning (APOE β(SE)=-0.05(0.02); AD-PRS β(SE)=-0.04(0.01)). Additionally, when modelled together, both amyloid-β status and AD-PRS predicted a steeper decline in memory functioning (amyloid-β β(SE)=-0.07(0.04); AD-PRS β(SE)=-0.04(0.01)). Modelling both APOE genotype and amyloid-β status, we observed an interaction, in which APOE genotype was related to steeper decline in memory and language functioning in amyloid-β abnormal individuals only (β(SE)=-0.13(0.06); β(SE)=-0.22(0.07), respectively).

Conclusion:

Our results suggest that APOE genotype is related to steeper decline in memory and language functioning in individuals with abnormal amyloid-β only. Furthermore, independent of amyloid-β status other genetic risk variants contribute to memory decline in initially cognitively unimpaired older individuals.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jack CRJ, et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. The lancet neurology. 2013;12.2: 207-216.CrossRef Jack CRJ, et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. The lancet neurology. 2013;12.2: 207-216.CrossRef
2.
go back to reference Gordon BA, et al. Spatial patterns of neuroimaging biomarker change in individuals from families with autosomal dominant Alzheimer’s disease: a longitudinal study. Lancet Neurol. 2018;17:241–50.CrossRef Gordon BA, et al. Spatial patterns of neuroimaging biomarker change in individuals from families with autosomal dominant Alzheimer’s disease: a longitudinal study. Lancet Neurol. 2018;17:241–50.CrossRef
4.
go back to reference Tomlinson BE, Blessed G, Roth M. Observations on the brains of non-demented old people. J Neurol Sci. 1968;7:331–56.CrossRef Tomlinson BE, Blessed G, Roth M. Observations on the brains of non-demented old people. J Neurol Sci. 1968;7:331–56.CrossRef
7.
go back to reference Sperling RA, et al. Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & dementia. 2011;7.3: 280-292. Sperling RA, et al. Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & dementia. 2011;7.3: 280-292.
9.
go back to reference Dubois B, et al. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimer’s Dement. 2016;12:292–323.CrossRef Dubois B, et al. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimer’s Dement. 2016;12:292–323.CrossRef
10.
go back to reference Corder EH, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.CrossRef Corder EH, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.CrossRef
11.
go back to reference Caselli RJ, et al. Cognitive domain decline in healthy apolipoprotein E ε4 homozygotes before the diagnosis of mild cognitive impairment. Arch Neurol. 2007;64:1306–11.CrossRef Caselli RJ, et al. Cognitive domain decline in healthy apolipoprotein E ε4 homozygotes before the diagnosis of mild cognitive impairment. Arch Neurol. 2007;64:1306–11.CrossRef
13.
go back to reference Bellenguez C, et al. New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nature genetics. 2022:1–25. Bellenguez C, et al. New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nature genetics. 2022:1–25.
14.
15.
go back to reference Ebenau JL, et al. Risk of dementia in APOE ε4 carriers is mitigated by a polygenic risk score. Alzheimer’s & Dementia: Diagnosis Assessment & Disease Monitoring. 2021;13:e12229. Ebenau JL, et al. Risk of dementia in APOE ε4 carriers is mitigated by a polygenic risk score. Alzheimer’s & Dementia: Diagnosis Assessment & Disease Monitoring. 2021;13:e12229.
16.
go back to reference Darst BF, et al. Pathway-specific polygenic risk scores as predictors of amyloid-β deposition and cognitive function in a sample at increased risk for Alzheimer’s disease. J Alzheimers Dis. 2017;55:473–84.CrossRef Darst BF, et al. Pathway-specific polygenic risk scores as predictors of amyloid-β deposition and cognitive function in a sample at increased risk for Alzheimer’s disease. J Alzheimers Dis. 2017;55:473–84.CrossRef
18.
go back to reference Porter T, et al. Utility of an Alzheimer’s disease risk-weighted polygenic risk score for predicting rates of cognitive decline in preclinical Alzheimer’s disease: a prospective longitudinal study. J Alzheimers Dis. 2018;66:1193–211.CrossRef Porter T, et al. Utility of an Alzheimer’s disease risk-weighted polygenic risk score for predicting rates of cognitive decline in preclinical Alzheimer’s disease: a prospective longitudinal study. J Alzheimers Dis. 2018;66:1193–211.CrossRef
19.
go back to reference Carrasquillo MM, et al. Late-onset Alzheimer’s risk variants in memory decline, incident mild cognitive impairment, and Alzheimer’s disease. Neurobiol Aging. 2015;36:60–7.CrossRef Carrasquillo MM, et al. Late-onset Alzheimer’s risk variants in memory decline, incident mild cognitive impairment, and Alzheimer’s disease. Neurobiol Aging. 2015;36:60–7.CrossRef
20.
go back to reference Skoog I, et al. A non-APOE polygenic risk score for Alzheimer’s disease is associated with CSF neurofilament light in a representative sample of cognitively unimpaired 70-year-olds. The Journals of Gerontology: Series A (2021). Skoog I, et al. A non-APOE polygenic risk score for Alzheimer’s disease is associated with CSF neurofilament light in a representative sample of cognitively unimpaired 70-year-olds. The Journals of Gerontology: Series A (2021).
21.
go back to reference Marden JR, et al. Using an Alzheimer’s Disease polygenic risk score to predict memory decline in black and white Americans over 14 years of follow-up Running head: AD polygenic risk score predicting memory decline. Alzheimer Dis Assoc Disord. 2016;30:195.CrossRef Marden JR, et al. Using an Alzheimer’s Disease polygenic risk score to predict memory decline in black and white Americans over 14 years of follow-up Running head: AD polygenic risk score predicting memory decline. Alzheimer Dis Assoc Disord. 2016;30:195.CrossRef
22.
go back to reference Verhaaren BF, et al. Alzheimer’s disease genes and cognition in the nondemented general population. Biol Psychiatry. 2013;73:429–34.CrossRef Verhaaren BF, et al. Alzheimer’s disease genes and cognition in the nondemented general population. Biol Psychiatry. 2013;73:429–34.CrossRef
23.
go back to reference Andrews SJ, Das D, Cherbuin N, Anstey KJ, Easteal S. Association of genetic risk factors with cognitive decline: the PATH through life project. Neurobiol Aging. 2016;41:150–8.CrossRef Andrews SJ, Das D, Cherbuin N, Anstey KJ, Easteal S. Association of genetic risk factors with cognitive decline: the PATH through life project. Neurobiol Aging. 2016;41:150–8.CrossRef
24.
go back to reference Riaz M, et al. Effect of APOE and a polygenic risk score on incident dementia and cognitive decline in a healthy older population. Aging Cell. 2021;20:e13384.CrossRef Riaz M, et al. Effect of APOE and a polygenic risk score on incident dementia and cognitive decline in a healthy older population. Aging Cell. 2021;20:e13384.CrossRef
26.
go back to reference Tan CH, et al. Polygenic hazard score: an enrichment marker for Alzheimer’s associated amyloid and tau deposition. Acta Neuropathol. 2018;135:85–93.CrossRef Tan CH, et al. Polygenic hazard score: an enrichment marker for Alzheimer’s associated amyloid and tau deposition. Acta Neuropathol. 2018;135:85–93.CrossRef
27.
go back to reference Cruchaga C, et al. Polygenic risk score of sporadic late-onset Alzheimer’s disease reveals a shared architecture with the familial and early-onset forms. Alzheimer’s Dement. 2018;14:205–14.CrossRef Cruchaga C, et al. Polygenic risk score of sporadic late-onset Alzheimer’s disease reveals a shared architecture with the familial and early-onset forms. Alzheimer’s Dement. 2018;14:205–14.CrossRef
31.
go back to reference de Jager CA, Budge MM, Clarke R. Utility of TICS‐M for the assessment of cognitive function in older adults. International journal of geriatric psychiatry. 2003;18.4:318-324.CrossRef de Jager CA, Budge MM, Clarke R. Utility of TICS‐M for the assessment of cognitive function in older adults. International journal of geriatric psychiatry. 2003;18.4:318-324.CrossRef
32.
go back to reference Morris, JC, et al. The consortium to establish a registry for Alzheimer’s disease (CERAD): I. Clinical and neuropsychological assessment of Alzheimer’s disease. Neurology. 1989 Morris, JC, et al. The consortium to establish a registry for Alzheimer’s disease (CERAD): I. Clinical and neuropsychological assessment of Alzheimer’s disease. Neurology. 1989
33.
go back to reference Morris, JC. Current vision and scoring rules the clinical dementia rating (CDR). Neurology. 1993;43:2412-2414. Morris, JC. Current vision and scoring rules the clinical dementia rating (CDR). Neurology. 1993;43:2412-2414.
34.
go back to reference Yesavage, JA, et al. Development and validation of a geriatric depression screening scale: a preliminary report. Journal of psychiatric research. 1982;17.1:37-49.CrossRef Yesavage, JA, et al. Development and validation of a geriatric depression screening scale: a preliminary report. Journal of psychiatric research. 1982;17.1:37-49.CrossRef
35.
go back to reference Saan R, Deelman B De 15-woordentest A en B (een voorlopige handleiding). Groningen: Afdeling Neuropsychologie, AZG (1986). Saan R, Deelman B De 15-woordentest A en B (een voorlopige handleiding). Groningen: Afdeling Neuropsychologie, AZG (1986).
36.
go back to reference Rey A L’examen clinique en psychologie [Clinical psychological examination] Presses Universitaires de France. Paris, France (1964). Rey A L’examen clinique en psychologie [Clinical psychological examination] Presses Universitaires de France. Paris, France (1964).
37.
go back to reference Meyers JE, Bayless JD, Meyers KR. Rey complex figure: memory error patterns and functional abilities. Applied Neuropsychology. 1996;3.2:89-92.CrossRef Meyers JE, Bayless JD, Meyers KR. Rey complex figure: memory error patterns and functional abilities. Applied Neuropsychology. 1996;3.2:89-92.CrossRef
40.
go back to reference Robbins TW, et al. Cambridge Neuropsychological Test Automated Battery (CANTAB): a factor analytic study of a large sample of normal elderly volunteers. Dement Geriatr Cogn Disord. 1994;5:266–81.CrossRef Robbins TW, et al. Cambridge Neuropsychological Test Automated Battery (CANTAB): a factor analytic study of a large sample of normal elderly volunteers. Dement Geriatr Cogn Disord. 1994;5:266–81.CrossRef
42.
go back to reference Rentz, DM, et al. Face-name associative memory performance is related to amyloid burden in normal elderly. Neuropsychologia. 2011;49.9:2776-2783. Rentz, DM, et al. Face-name associative memory performance is related to amyloid burden in normal elderly. Neuropsychologia. 2011;49.9:2776-2783.
43.
go back to reference Reitan, RM. Validity of the Trail Making Test as an indicator of organic brain damage. Perceptual and motor skills. 1958;8.3:271-276. Reitan, RM. Validity of the Trail Making Test as an indicator of organic brain damage. Perceptual and motor skills. 1958;8.3:271-276.
44.
go back to reference Wechsler, D. Wechsler adult intelligence scale-revised (WAIS-R). Psychological Corporation. 1981. Wechsler, D. Wechsler adult intelligence scale-revised (WAIS-R). Psychological Corporation. 1981.
46.
go back to reference Lindeboom J, Schmand B, Tulner L, Walstra G, Jonker C. J Neurol Neurosurg Psychiatry. 2002;73:126–33.CrossRef Lindeboom J, Schmand B, Tulner L, Walstra G, Jonker C. J Neurol Neurosurg Psychiatry. 2002;73:126–33.CrossRef
47.
52.
go back to reference Tesi N, et al. Immune response and endocytosis pathways are associated with the resilience against Alzheimer’s disease. Translational psychiatry. 2020;10:1–12.CrossRef Tesi N, et al. Immune response and endocytosis pathways are associated with the resilience against Alzheimer’s disease. Translational psychiatry. 2020;10:1–12.CrossRef
53.
go back to reference Narasimhan V, et al. BCFtools/RoH: a hidden Markov model approach for detecting autozygosity from next-generation sequencing data. Bioinformatics. 2016;32:1749–51.CrossRef Narasimhan V, et al. BCFtools/RoH: a hidden Markov model approach for detecting autozygosity from next-generation sequencing data. Bioinformatics. 2016;32:1749–51.CrossRef
56.
go back to reference McCarthy S, et al Haplotype Reference, C.(2016). A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet48, 1279–1283. McCarthy S, et al Haplotype Reference, C.(2016). A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet48, 1279–1283.
57.
go back to reference Dudbridge F. Power and predictive accuracy of polygenic risk scores. PLoS Genet. 2013;9:e1003348.CrossRef Dudbridge F. Power and predictive accuracy of polygenic risk scores. PLoS Genet. 2013;9:e1003348.CrossRef
59.
go back to reference Sibomana M, et al. in IEEE Symposium Conference Record Nuclear Science 2004. 2647–2651 (IEEE). Sibomana M, et al. in IEEE Symposium Conference Record Nuclear Science 2004. 2647–2651 (IEEE).
61.
go back to reference GEHealthcare. EPAR product information - summary of product characteristics. (2014). GEHealthcare. EPAR product information - summary of product characteristics. (2014).
62.
go back to reference Van Buuren S, Groothuis-Oudshoorn K. mice: Multivariate imputation by chained equations in R. J Stat Softw. 2011;45:1–67.CrossRef Van Buuren S, Groothuis-Oudshoorn K. mice: Multivariate imputation by chained equations in R. J Stat Softw. 2011;45:1–67.CrossRef
63.
go back to reference Benjamini Y, Hochberg Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J Roy Stat Soc: Ser B (Methodol). 1995;57:289–300. Benjamini Y, Hochberg Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J Roy Stat Soc: Ser B (Methodol). 1995;57:289–300.
64.
go back to reference Sleegers K, et al. A 22-single nucleotide polymorphism Alzheimer’s disease risk score correlates with family history, onset age, and cerebrospinal fluid Aβ42. Alzheimer’s Dement. 2015;11:1452–60.CrossRef Sleegers K, et al. A 22-single nucleotide polymorphism Alzheimer’s disease risk score correlates with family history, onset age, and cerebrospinal fluid Aβ42. Alzheimer’s Dement. 2015;11:1452–60.CrossRef
65.
go back to reference Lopresti BJ, et al. Influence of apolipoprotein-E genotype on brain amyloid load and longitudinal trajectories. Neurobiol Aging. 2020;94:111–20.CrossRef Lopresti BJ, et al. Influence of apolipoprotein-E genotype on brain amyloid load and longitudinal trajectories. Neurobiol Aging. 2020;94:111–20.CrossRef
66.
go back to reference Bellenguez C, et al New insights on the genetic etiology of Alzheimer’s and related dementia. MedRxiv (2020). Bellenguez C, et al New insights on the genetic etiology of Alzheimer’s and related dementia. MedRxiv (2020).
67.
go back to reference Konijnenberg E, et al. Association of amyloid pathology with memory performance and cognitive complaints in cognitively normal older adults: a monozygotic twin study. Neurobiol Aging. 2019;77:58–65.CrossRef Konijnenberg E, et al. Association of amyloid pathology with memory performance and cognitive complaints in cognitively normal older adults: a monozygotic twin study. Neurobiol Aging. 2019;77:58–65.CrossRef
69.
go back to reference Doraiswamy PM, et al. Amyloid-β assessed by florbetapir F 18 PET and 18-month cognitive decline: a multicenter study. Neurology. 2012;79:1636–44.CrossRef Doraiswamy PM, et al. Amyloid-β assessed by florbetapir F 18 PET and 18-month cognitive decline: a multicenter study. Neurology. 2012;79:1636–44.CrossRef
70.
go back to reference Landau SM, et al. Amyloid deposition, hypometabolism, and longitudinal cognitive decline. Ann Neurol. 2012;72:578–86.CrossRef Landau SM, et al. Amyloid deposition, hypometabolism, and longitudinal cognitive decline. Ann Neurol. 2012;72:578–86.CrossRef
71.
go back to reference De Jager PL, et al. A genome-wide scan for common variants affecting the rate of age-related cognitive decline. Neurobiol Aging 33, 1017. e1011–7. e1015 (2012).CrossRef De Jager PL, et al. A genome-wide scan for common variants affecting the rate of age-related cognitive decline. Neurobiol Aging 33, 1017. e1011–7. e1015 (2012).CrossRef
72.
go back to reference Mormino EC, et al. Amyloid and APOE ε4 interact to influence short-term decline in preclinical Alzheimer disease. Neurology. 2014;82:1760–7.CrossRef Mormino EC, et al. Amyloid and APOE ε4 interact to influence short-term decline in preclinical Alzheimer disease. Neurology. 2014;82:1760–7.CrossRef
74.
go back to reference Kunkle BW, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat Genet. 2019;51:414–30.CrossRef Kunkle BW, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat Genet. 2019;51:414–30.CrossRef
76.
go back to reference Tomassen J, et al. Abnormal cerebrospinal fluid levels of amyloid and tau are associated with cognitive decline over time in cognitively normal older adults: A monozygotic twin study. Alzheimer’s & Dementia: Translational Research & Clinical Interventions. 2022;8:e12346. doi:https://doi.org/10.1002/trc2.12346.CrossRef Tomassen J, et al. Abnormal cerebrospinal fluid levels of amyloid and tau are associated with cognitive decline over time in cognitively normal older adults: A monozygotic twin study. Alzheimer’s & Dementia: Translational Research & Clinical Interventions. 2022;8:e12346. doi:https://​doi.​org/​10.​1002/​trc2.​12346.CrossRef
78.
go back to reference Villeneuve S, et al. Existing Pittsburgh Compound-B positron emission tomography thresholds are too high: statistical and pathological evaluation. Brain. 2015;138:2020–33.CrossRef Villeneuve S, et al. Existing Pittsburgh Compound-B positron emission tomography thresholds are too high: statistical and pathological evaluation. Brain. 2015;138:2020–33.CrossRef
79.
go back to reference Liu C-C, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Reviews Neurol. 2013;9:106–18.CrossRef Liu C-C, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Reviews Neurol. 2013;9:106–18.CrossRef
80.
go back to reference Duncan L, et al. Analysis of polygenic risk score usage and performance in diverse human populations. Nat Commun. 2019;10:1–9.CrossRef Duncan L, et al. Analysis of polygenic risk score usage and performance in diverse human populations. Nat Commun. 2019;10:1–9.CrossRef
81.
go back to reference Vermunt L, et al. Duration of preclinical, prodromal, and dementia stages of Alzheimer’s disease in relation to age, sex, and APOE genotype. Alzheimer’s Dement. 2019;15:888–98.CrossRef Vermunt L, et al. Duration of preclinical, prodromal, and dementia stages of Alzheimer’s disease in relation to age, sex, and APOE genotype. Alzheimer’s Dement. 2019;15:888–98.CrossRef
Metadata
Title
Amyloid-β and APOE genotype predict memory decline in cognitively unimpaired older individuals independently of Alzheimer’s disease polygenic risk score
Authors
Jori Tomassen
Anouk den Braber
Sven J. van der Lee
Lianne M. Reus
Elles Konijnenberg
Stephen F. Carter
Maqsood Yaqub
Bart N.M. van Berckel
Lyduine E. Collij
Dorret I. Boomsma
Eco J.C. de Geus
Philip Scheltens
Karl Herholz
Betty M. Tijms
Pieter Jelle Visser
Publication date
01-12-2022
Publisher
BioMed Central
Published in
BMC Neurology / Issue 1/2022
Electronic ISSN: 1471-2377
DOI
https://doi.org/10.1186/s12883-022-02925-6

Other articles of this Issue 1/2022

BMC Neurology 1/2022 Go to the issue