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.

  • Review Article
  • Published:

The multiplex model of the genetics of Alzheimer’s disease

Abstract

Genes play a strong role in Alzheimer’s disease (AD), with late-onset AD showing heritability of 58–79% and early-onset AD showing over 90%. Genetic association provides a robust platform to build our understanding of the etiology of this complex disease. Over 50 loci are now implicated for AD, suggesting that AD is a disease of multiple components, as supported by pathway analyses (immunity, endocytosis, cholesterol transport, ubiquitination, amyloid-β and tau processing). Over 50% of late-onset AD heritability has been captured, allowing researchers to calculate the accumulation of AD genetic risk through polygenic risk scores. A polygenic risk score predicts disease with up to 90% accuracy and is an exciting tool in our research armory that could allow selection of those with high polygenic risk scores for clinical trials and precision medicine. It could also allow cellular modelling of the combined risk. Here we propose the multiplex model as a new perspective from which to understand AD. The multiplex model reflects the combination of some, or all, of these model components (genetic and environmental), in a tissue-specific manner, to trigger or sustain a disease cascade, which ultimately results in the cell and synaptic loss observed in AD.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Schematic of Mendelian disease-causing genes and loci reaching GWS for single-variant (not gene-wide) association with sporadic AD.
Fig. 2: Multiplex model of Alzheimer’s disease.
Fig. 3: Polygenic risk score (PRS) in Alzheimer’s disease.
Fig. 4: Schematic demonstrating the complexity and methods of discovering Alzheimer’s disease mechanisms.

Similar content being viewed by others

References

  1. Goate, A. et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 349, 704–706 (1991).

    Article  CAS  PubMed  Google Scholar 

  2. Sherrington, R. et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 375, 754–760 (1995).

    Article  CAS  PubMed  Google Scholar 

  3. Rogaev, E. I. et al. Familial Alzheimer’s disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer’s disease type 3 gene. Nature 376, 775–778 (1995).

    Article  CAS  PubMed  Google Scholar 

  4. Hardy, J. & Selkoe, D. J. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Ricciarelli, R. & Fedele, E. The amyloid cascade hypothesis in Alzheimer’s disease: it’s time to change our mind. Curr. Neuropharmacol. 15, 926–935 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Doody, R. S., Farlow, M. & Aisen, P. S., Alzheimer’s Disease Cooperative Study Data Analysis and Publication Committee. Phase 3 trials of solanezumab and bapineuzumab for Alzheimer’s disease. N. Engl. J. Med. 370, 1460 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Honig, L. S. et al. Trial of solanezumab for mild dementia due to Alzheimer’s disease. N. Engl. J. Med. 378, 321–330 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Galimberti, D. & Scarpini, E. Disease-modifying treatments for Alzheimer’s disease. Ther. Adv. Neurol. Disord. 4, 203–216 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yiannopoulou, K. G. & Papageorgiou, S. G. Current and future treatments for Alzheimer’s disease. Ther. Adv. Neurol. Disord. 6, 19–33 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jagust, W. Imaging the evolution and pathophysiology of Alzheimer disease. Nat. Rev. Neurosci. 19, 687–700 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rajan, K. B., Wilson, R. S., Weuve, J., Barnes, L. L. & Evans, D. A. Cognitive impairment 18 years before clinical diagnosis of Alzheimer disease dementia. Neurology 85, 898–904 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rosenblum, W. I. Why Alzheimer trials fail: removing soluble oligomeric beta amyloid is essential, inconsistent, and difficult. Neurobiol. Aging 35, 969–974 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Gatz, M. et al. Role of genes and environments for explaining Alzheimer disease. Arch. Gen. Psychiatry 63, 168–174 (2006).

    Article  PubMed  Google Scholar 

  14. Wingo, T. S., Lah, J. J., Levey, A. I. & Cutler, D. J. Autosomal recessive causes likely in early-onset Alzheimer disease. Arch. Neurol. 69, 59–64 (2012).

    Article  PubMed  Google Scholar 

  15. Saunders, A. M. et al. Apolipoprotein E epsilon 4 allele distributions in late-onset Alzheimer’s disease and in other amyloid-forming diseases. Lancet 342, 710–711 (1993).

    Article  CAS  PubMed  Google Scholar 

  16. Strittmatter, W. J. et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc. Natl Acad. Sci. USA 90, 1977–1981 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Corder, E. H. et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat. Genet. 7, 180–184 (1994).

    Article  CAS  PubMed  Google Scholar 

  18. Liao, F., Yoon, H. & Kim, J. Apolipoprotein E metabolism and functions in brain and its role in Alzheimer’s disease. Curr. Opin. Lipidol. 28, 60–67 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Deane, R. et al. ApoE isoform-specific disruption of amyloid beta peptide clearance from mouse brain. J. Clin. Invest. 118, 4002–4013 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Verghese, P. B., Castellano, J. M. & Holtzman, D. M. Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurol. 10, 241–252 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Harold, D. et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet. 41, 1088–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lambert, J. C. et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat. Genet. 41, 1094–1099 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Seshadri, S. et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. J. Am. Med. Assoc. 303, 1832–1840 (2010).

    Article  CAS  Google Scholar 

  24. Naj, A. C. et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat. Genet. 43, 436–441 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hollingworth, P. et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat. Genet. 43, 429–435 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lambert, J. C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 45, 1452–1458 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Desikan, R. S. et al. Genetic assessment of age-associated Alzheimer disease risk: development and validation of a polygenic hazard score. PLoS Med. 14, e1002258 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Johnson, E. C. B. et al. Deep proteomic network analysis of Alzheimer’s disease brain reveals alterations in RNA binding proteins and RNA splicing associated with disease. Mol. Neurodegener. 13, 52 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Anttila, V. et al. Analysis of shared heritability in common disorders of the brain. Science 360, eaap8757 (2018).

    Article  PubMed  CAS  Google Scholar 

  30. Ruiz, A. et al. Follow-up of loci from the International Genomics of Alzheimer’s Disease Project identifies TRIP4 as a novel susceptibility gene. Transl. Psychiatry 4, e358 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jansen, I. E. et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 51, 404–413 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Jun, G. R. et al. Transethnic genome-wide scan identifies novel Alzheimer’s disease loci. Alzheimers Dement. 13, 727–738 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. de Rojas, I., et al. Common variants in Alzheimer’s disease: novel association of six genetic variants with AD and risk stratification by polygenic risk scores. Preprint at medRxiv https://doi.org/10.1101/19012021 (2019).

  35. Escott-Price, V. et al. Gene-wide analysis detects two new susceptibility genes for Alzheimer’s disease. PLoS One 9, e94661 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Baker, E. et al. Gene-based analysis in HRC imputed genome wide association data identifies three novel genes for Alzheimer’s disease. PLoS One 14, e0218111 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Marioni, R. E. et al. GWAS on family history of Alzheimer’s disease. Transl. Psychiatry 8, 99 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Liu, J. Z., Erlich, Y. & Pickrell, J. K. Case-control association mapping by proxy using family history of disease. Nat. Genet. 49, 325–331 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. So, H. C., Gui, A. H., Cherny, S. S. & Sham, P. C. Evaluating the heritability explained by known susceptibility variants: a survey of ten complex diseases. Genet. Epidemiol. 35, 310–317 (2011).

    Article  PubMed  Google Scholar 

  40. Ridge, P. G., Mukherjee, S., Crane, P. K. & Kauwe, J. S. Alzheimer’s Disease Genetics Consortium. Alzheimer’s disease: analyzing the missing heritability. PLoS One 8, e79771 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Meng, W. et al. A genome-wide association study finds genetic associations with broadly-defined headache in UK Biobank (N=223,773). EBioMedicine 28, 180–186 (2018).

    PubMed  PubMed Central  Google Scholar 

  42. Kunkle, B. et al. Meta-analysis of genetic association with diagnosed Alzheimer’s disease identifies novel risk loci and implicates Abeta, Tau, immunity and lipid processing. Nat. Genet. 51, 414–430 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Guerreiro, R. et al. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med. 368, 117–127 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Bis, J.C. et al. Whole exome sequencing study identifies novel rare and common Alzheimer’s-associated variants involved in immune response and transcriptional regulation. Mol. Psychiatry https://doi.org/10.1038/s41380-018-0112-7 (2018).

  45. Jonsson, T. et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 368, 107–116 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Jonsson, T. et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature 488, 96–99 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Kunkle, B. W. et al. Early-onset Alzheimer disease and candidate risk genes involved in endolysosomal transport. JAMA Neurol. 74, 1113–1122 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Bellenguez, C. et al. Contribution to Alzheimer’s disease risk of rare variants in TREM2, SORL1, and ABCA7 in 1779 cases and 1273 controls. Neurobiol. Aging 59, 220.e1–220.e9 (2017).

    Article  CAS  Google Scholar 

  49. Louwersheimer, E. et al. Influence of genetic variants in SORL1 gene on the manifestation of Alzheimer’s disease. Neurobiol. Aging 36, 1605.e13–1605.e20 (2015).

    Article  CAS  Google Scholar 

  50. Steinberg, S. et al. Loss-of-function variants in ABCA7 confer risk of Alzheimer’s disease. Nat. Genet. 47, 445–447 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Cruchaga, C. et al. Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer’s disease. Nature 505, 550–554 (2014).

    Article  CAS  PubMed  Google Scholar 

  52. Logue, M. W. et al. Two rare AKAP9 variants are associated with Alzheimer’s disease in African Americans. Alzheimers Dement. 10, 609–618.e11 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Wetzel-Smith, M. K. et al. A rare mutation in UNC5C predisposes to late-onset Alzheimer’s disease and increases neuronal cell death. Nat. Med. 20, 1452–1457 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sims, R. et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat. Genet. 49, 1373–1384 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Magno, L. et al. Alzheimer’s disease phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph. Alzheimers Res. Ther. 11, 16 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Gusareva, E. S. et al. Male-specific epistasis between WWC1 and TLN2 genes is associated with Alzheimer’s disease. Neurobiol. Aging 72, 188.e3–188.e12 (2018).

    Article  CAS  Google Scholar 

  57. Ridge, P. G. et al. Linkage, whole genome sequence, and biological data implicate variants in RAB10 in Alzheimer’s disease resilience. Genome Med. 9, 100 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Ropacki, S. A. & Jeste, D. V. Epidemiology of and risk factors for psychosis of Alzheimer’s disease: a review of 55 studies published from 1990 to 2003. Am. J. Psychiatry 162, 2022–2030 (2005).

    Article  PubMed  Google Scholar 

  59. Shin, I. S., Carter, M., Masterman, D., Fairbanks, L. & Cummings, J. L. Neuropsychiatric symptoms and quality of life in Alzheimer disease. Am. J. Geriatr. Psychiatry 13, 469–474 (2005).

    Article  PubMed  Google Scholar 

  60. Wilkosz, P. A. et al. Trajectories of cognitive decline in Alzheimer’s disease. Int. Psychogeriatr. 22, 281–290 (2010).

    Article  PubMed  Google Scholar 

  61. Lopez, O. L. et al. Long-term effects of the concomitant use of memantine with cholinesterase inhibition in Alzheimer disease. J. Neurol. Neurosurg. Psychiatry 80, 600–607 (2009).

    Article  CAS  PubMed  Google Scholar 

  62. Hollingworth, P. et al. Genome-wide association study of Alzheimer’s disease with psychotic symptoms. Mol. Psychiatry 17, 1316–1327 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Del-Aguila, J. L. et al. Assessment of the genetic architecture of Alzheimer’s disease risk in rate of memory decline. J. Alzheimers Dis. 62, 745–756 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Criswell, L. A. et al. Analysis of families in the multiple autoimmune disease genetics consortium (MADGC) collection: the PTPN22 620W allele associates with multiple autoimmune phenotypes. Am. J. Hum. Genet. 76, 561–571 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Eaton, W. W., Rose, N. R., Kalaydjian, A., Pedersen, M. G. & Mortensen, P. B. Epidemiology of autoimmune diseases in Denmark. J. Autoimmun. 29, 1–9 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Solovieff, N., Cotsapas, C., Lee, P. H., Purcell, S. M. & Smoller, J. W. Pleiotropy in complex traits: challenges and strategies. Nat. Rev. Genet. 14, 483–495 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sivakumaran, S. et al. Abundant pleiotropy in human complex diseases and traits. Am. J. Hum. Genet. 89, 607–618 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Moskvina, V. et al. Analysis of genome-wide association studies of Alzheimer disease and of Parkinson disease to determine if these 2 diseases share a common genetic risk. JAMA Neurol. 70, 1268–1276 (2013).

    PubMed  PubMed Central  Google Scholar 

  69. Guerreiro, R. et al. Genome-wide analysis of genetic correlation in dementia with Lewy bodies, Parkinson’s and Alzheimer’s diseases. Neurobiol. Aging 38, 214.e7–214.e10 (2016).

    Article  CAS  Google Scholar 

  70. Geiger, J. T. et al. Next-generation sequencing reveals substantial genetic contribution to dementia with Lewy bodies. Neurobiol. Dis. 94, 55–62 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Liu, G. et al. Cardiovascular disease contributes to Alzheimer’s disease: evidence from large-scale genome-wide association studies. Neurobiol. Aging 35, 786–792 (2014).

    Article  PubMed  Google Scholar 

  72. Holmans, P. et al. Gene ontology analysis of GWA study data sets provides insights into the biology of bipolar disorder. Am. J. Hum. Genet. 85, 13–24 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lee, P. H., O’Dushlaine, C., Thomas, B. & Purcell, S. M. INRICH: interval-based enrichment analysis for genome-wide association studies. Bioinformatics 28, 1797–1799 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. de Leeuw, C. A., Mooij, J. M., Heskes, T. & Posthuma, D. MAGMA: generalized gene-set analysis of GWAS data. PLOS Comput. Biol. 11, e1004219 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. International Genomics of Alzheimer’s Disease Consortium (IGAP). Convergent genetic and expression data implicate immunity in Alzheimer’s disease. Alzheimers Dement. 11, 658–671 (2015).

  76. Yu, C. H., Pal, L. R. & Moult, J. Consensus genome-wide expression quantitative trait loci and their relationship with human complex trait disease. OMICS 20, 400–414 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Gusev, A. et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48, 245–252 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Gamazon, E. R. et al. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet. 47, 1091–1098 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Raj, T. et al. Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer’s disease susceptibility. Nat. Genet. 50, 1584–1592 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Ramasamy, A. et al. Genetic variability in the regulation of gene expression in ten regions of the human brain. Nat. Neurosci. 17, 1418–1428 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Akbarian, S. et al. The PsychENCODE project. Nat. Neurosci. 18, 1707–1712 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Fromer, M. et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat. Neurosci. 19, 1442–1453 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Raj, T. et al. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science 344, 519–523 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Skene, N. G. & Grant, S. G. Identification of vulnerable cell types in major brain disorders using single cell transcriptomes and expression weighted cell type enrichment. Front. Neurosci. 10, 16 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Skene, N. G. et al. Genetic identification of brain cell types underlying schizophrenia. Nat. Genet. 50, 825–833 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290.e17 (2017).

    Article  CAS  PubMed  Google Scholar 

  87. Hammond, T. R. et al. Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity 50, 253–271.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  88. Li, Q. et al. Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing. Neuron 101, 207–223.e10 (2019).

    Article  CAS  PubMed  Google Scholar 

  89. Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Farlik, M. et al. Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics. Cell Rep. 10, 1386–1397 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Peterson, V. M. et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol. 35, 936–939 (2017).

    Article  CAS  PubMed  Google Scholar 

  93. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Olah, M. et al. A single cell-based atlas of human microglial states reveals associations with neurological disorders and histopathological features of the aging brain. Preprint at bioRxiv https://doi.org/10.1101/343780 (2018).

  95. Mostafavi, S. et al. A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer’s disease. Nat. Neurosci. 21, 811–819 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhang, B. et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 153, 707–720 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Salih, D.A. et al. Genetic variability in response to amyloid beta deposition influences Alzheimer’s risk. Brain Commun. 1, fcz022 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  98. Gjoneska, E. et al. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature 518, 365–369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Lu, Q. et al. Systematic tissue-specific functional annotation of the human genome highlights immune-related DNA elements for late-onset Alzheimer’s disease. PLoS Genet. 13, e1006933 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Novikova, G. et al. Integration of Alzheimer’s disease genetics and myeloid cell genomics identifies novel causal variants, regulatory elements, genes and pathways. Preprint at bioRxiv https://doi.org/10.1101/694281 (2019).

  101. Amlie-Wolf, A. et al. Inferring the molecular mechanisms of noncoding Alzheimer’s disease-associated genetic variants. J. Alzheimers Dis. 72, 301–318 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Finucane, H. K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228–1235 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Huang, K. L. et al. A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer’s disease. Nat. Neurosci. 20, 1052–1061 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Tansey, K. E., Cameron, D. & Hill, M. J. Genetic risk for Alzheimer’s disease is concentrated in specific macrophage and microglial transcriptional networks. Genome Med. 10, 14 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Gosselin, D. et al. An environment-dependent transcriptional network specifies human microglia identity. Science 356, eaal3222 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Matarin, M. et al. A genome-wide gene-expression analysis and database in transgenic mice during development of amyloid or tau pathology. Cell Rep. 10, 633–644 (2015).

    Article  CAS  PubMed  Google Scholar 

  107. Liu, X., Li, Y. I. & Pritchard, J. K. Trans effects on gene expression can drive omnigenic inheritance. Cell 177, 1022–1034.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Alasoo, K. et al. Shared genetic effects on chromatin and gene expression indicate a role for enhancer priming in immune response. Nat. Genet. 50, 424–431 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Lodato, M. A. et al. Somatic mutation in single human neurons tracks developmental and transcriptional history. Science 350, 94–98 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. McConnell, M. J. et al. Intersection of diverse neuronal genomes and neuropsychiatric disease: the brain somatic mosaicism network. Science 356, eaal1641 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Arendt, T., Brückner, M. K., Mosch, B. & Lösche, A. Selective cell death of hyperploid neurons in Alzheimer’s disease. Am. J. Pathol. 177, 15–20 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Bushman, D. M. et al. Genomic mosaicism with increased amyloid precursor protein (APP) gene copy number in single neurons from sporadic Alzheimer’s disease brains. eLife 4, e05116 (2015).

    Article  PubMed Central  Google Scholar 

  113. Lee, M. H. et al. Somatic APP gene recombination in Alzheimer’s disease and normal neurons. Nature 563, 639–645 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Lee, S. H. et al. Estimation and partitioning of polygenic variation captured by common SNPs for Alzheimer’s disease, multiple sclerosis and endometriosis. Hum. Mol. Genet. 22, 832–841 (2013).

    Article  CAS  PubMed  Google Scholar 

  115. Escott-Price, V. et al. Common polygenic variation enhances risk prediction for Alzheimer’s disease. Brain 138, 3673–3684 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Escott-Price, V. & Jones, L. Genomic profiling and diagnostic biomarkers in Alzheimer’s disease. Lancet Neurol. 16, 582–583 (2017).

    Article  PubMed  Google Scholar 

  117. Escott-Price, V., Shoai, M., Pither, R., Williams, J. & Hardy, J. Polygenic score prediction captures nearly all common genetic risk for Alzheimer’s disease. Neurobiol. Aging 49, 214.e7–214.e11 (2017).

    Article  Google Scholar 

  118. Ridge, P. G. et al. Assessment of the genetic variance of late-onset Alzheimer’s disease. Neurobiol. Aging 41, 200.e13–200.e20 (2016).

    Article  CAS  Google Scholar 

  119. Lewis, C. M. & Vassos, E. Prospects for using risk scores in polygenic medicine. Genome Med. 9, 96 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Escott-Price, V., Myers, A. J., Huentelman, M. & Hardy, J. Polygenic risk score analysis of pathologically confirmed Alzheimer disease. Ann. Neurol. 82, 311–314 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  121. Ahmad, S. et al. Disentangling the biological pathways involved in early features of Alzheimer’s disease in the Rotterdam Study. Alzheimers Dement. 14, 848–857 (2018).

    Article  PubMed  Google Scholar 

  122. Biffi, A. et al. Genetic variation and neuroimaging measures in Alzheimer disease. Arch. Neurol. 67, 677–685 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Braskie, M. N., Ringman, J. M. & Thompson, P. M. Neuroimaging measures as endophenotypes in Alzheimer’s disease. Int. J. Alzheimers Dis. 2011, 490140 (2011).

    PubMed  PubMed Central  Google Scholar 

  124. Saykin, A. J. et al. Alzheimer’s Disease Neuroimaging Initiative biomarkers as quantitative phenotypes: genetics core aims, progress, and plans. Alzheimers Dement. 6, 265–273 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Mormino, E. C. et al. Polygenic risk of Alzheimer disease is associated with early- and late-life processes. Neurology 87, 481–488 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Bralten, J. et al. CR1 genotype is associated with entorhinal cortex volume in young healthy adults. Neurobiol. Aging 32, 2106.e7–2106.e11 (2011).

    Article  CAS  Google Scholar 

  127. Lancaster, T. M. et al. Alzheimer’s disease risk variant in CLU is associated with neural inefficiency in healthy individuals. Alzheimers Dement. 11, 1144–1152 (2015).

    Article  PubMed  Google Scholar 

  128. Foley, S. F. et al. Multimodal brain imaging reveals structural differences in Alzheimer’s disease polygenic risk carriers: a study in healthy young adults. Biol. Psychiatry 81, 154–161 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Axelrud, L. K. et al. Polygenic risk score for Alzheimer’s disease: implications for memory performance and hippocampal volumes in early life. Am. J. Psychiatry 175, 555–563 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Chandler, H. L. et al. Polygenic impact of common genetic risk loci for Alzheimer’s disease on cerebral blood flow in young individuals. Sci. Rep. 9, 467 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Lupton, M. K. et al. The effect of increased genetic risk for Alzheimer’s disease on hippocampal and amygdala volume. Neurobiol. Aging 40, 68–77 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Chauhan, G. et al. Association of Alzheimer’s disease GWAS loci with MRI markers of brain aging. Neurobiol. Aging 36, 1765.e7–1765.e16 (2015).

    Article  CAS  Google Scholar 

  133. Hibar, D. P. et al. Novel genetic loci associated with hippocampal volume. Nat. Commun. 8, 13624 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Satizabal, C.L. et al. Genetic architecture of subcortical brain structures in 38,851 individuals. Nat. Genet. 51, 1624–1636 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Thompson, P. M. et al. ENIGMA and the individual: predicting factors that affect the brain in 35 countries worldwide. Neuroimage 145, 389–408 (2017). Pt B.

    Article  PubMed  Google Scholar 

  136. Lancaster, T. M., Hill, M. J., Sims, R. & Williams, J. Microglia - mediated immunity partly contributes to the genetic association between Alzheimer’s disease and hippocampal volume. Brain Behav. Immun. 79, 267–273 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Esquerda-Canals, G., Montoliu-Gaya, L., Güell-Bosch, J. & Villegas, S. Mouse models of Alzheimer’s disease. J. Alzheimers Dis. 57, 1171–1183 (2017).

    Article  CAS  PubMed  Google Scholar 

  138. Bouleau, S. & Tricoire, H. Drosophila models of Alzheimer’s disease: advances, limits, and perspectives. J. Alzheimers Dis. 45, 1015–1038 (2015).

    Article  CAS  PubMed  Google Scholar 

  139. Yagi, T. et al. [Modeling familial Alzheimer’s disease with induced pluripotent stem cells]. Rinsho Shinkeigaku 52, 1134–1136 (2012).

    Article  PubMed  Google Scholar 

  140. Israel, M. A. et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 482, 216–220 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Garcia-Reitboeck, P. et al. Human induced pluripotent stem cell-derived microglia-like cells harboring TREM2 missense mutations show specific deficits in phagocytosis. Cell Rep. 24, 2300–2311 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Claes, C. et al. Human stem cell-derived monocytes and microglia-like cells reveal impaired amyloid plaque clearance upon heterozygous or homozygous loss of TREM2. Alzheimers Dement. 15, 453–464 (2019).

    Article  PubMed  Google Scholar 

  143. Zhao, J. et al. APOE ε4/ε4 diminishes neurotrophic function of human iPSC-derived astrocytes. Hum. Mol. Genet. 26, 2690–2700 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Lin, Y. T. et al. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 98, 1141–1154.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhao, Z. et al. Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance. Nat. Neurosci. 18, 978–987 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Robbins, J. P. et al. Clusterin is required for β-amyloid toxicity in human iPSC-derived neurons. Front. Neurosci. 12, 504 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Huang, Y. A., Zhou, B., Wernig, M. & Südhof, T. C. ApoE2, ApoE3, and ApoE4 differentially stimulate APP transcription and Aβ secretion. Cell 168, 427–441.e21 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Arber, C., Lovejoy, C. & Wray, S. Stem cell models of Alzheimer’s disease: progress and challenges. Alzheimers Res. Ther. 9, 42 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Rius-Pérez, S., Tormos, A. M., Pérez, S. & Taléns-Visconti, R. Vascular pathology: cause or effect in Alzheimer disease? Neurologia 33, 112–120 (2018).

    Article  PubMed  Google Scholar 

  150. Nelson, M. R. et al. The support of human genetic evidence for approved drug indications. Nat. Genet. 47, 856–860 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Cardiff University was supported by the UK Dementia Research Institute at Cardiff (MC_PC_17112), Medical Research Council MR/K013041/1), the Alzheimer’s Society, Alzheimer’s Research UK (ARUK-NC2018-WAL), Dementia Platform UK (HQR00720), the European Joint Programme for Neurodegenerative Disease (MR/N029402/1), the Welsh Assembly Government, Centre for Ageing & Dementia Research (SGR544:CADR) and a donation from the Moondance Charitable Foundation. We thank T. D. Cushion, G. Leonenko, J. Harwood and B. Lan-Leung for their support preparing this manuscript. Finally, we thank all patients and affected families for their continued generosity and willingness to participate in medical research; without their involvement, there would be no advancement in our knowledge of disease genetics.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Julie Williams.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Mina Ryten and Bryan Traynor 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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sims, R., Hill, M. & Williams, J. The multiplex model of the genetics of Alzheimer’s disease. Nat Neurosci 23, 311–322 (2020). https://doi.org/10.1038/s41593-020-0599-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-020-0599-5

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing