Compilation of AD-associated and non-AD

AD-associated (positives) and non-AD (positives) genes are needed to build a machine learning model. First of all, we performed intensive hand-curation to identify confident AD-associated genes (positives) from various disease genes resources, including AlzGene1, AlzBase2, OMIM3, DisGenet4, DistiLD5, and UniProt6, Open

Targets7, GWAS Catalog8, differentially expressed genes (DEGs) in ROSMAP9, and published literature. The curated genes from each resource as well as the corresponding criteria were provided in Table 1 in this file. As the AD-associated genes as well as their reliability vary across these resources, we used a vote strategy and selected only the genes that are present in at least two of the above-described resources to ensure higher reliability. In this way, we collected 147 AD-associated genes (Table 2). Of them, most (n=103, 70%) were associated with AD based on

GWAS.

Next, we selected a set of non-AD genes, (i.e. negatives), which refer to the genes that have no or minimal association with AD. The main idea of our method for non-AD selection was to remove any genes that exhibit potential associations with AD.

Let Gp be the set of the 147 collected positives. We then selected a negative gene set

GN containing genes that are potentially associated with AD in the following procedure.

We first performed a (GO) enrichment analysis for the positives using

PANTHER10 and obtained 737 biological process terms (FDR<0.05). It is reasonable to assume that the genes annotated to these GO terms exhibit a potential association with AD compared with a random baseline because they were annotated to the same

GO terms (functions) as the positive genes. In addition, we also collected genes that showed any potential association with AD from the above-described resources. These genes were then combined with the genes annotated to the GO terms, forming a set of genes of potential association with AD, denoted by GS. Next, the negative gene set

was then calculated as GN = GA - GS, where GA represents all the genes in the network.

By removing the 19471 genes identified in GS from GA (21122 genes), 1651 genes that were not associated with AD were identified and used as non-AD (negatives) genes.

Table 1. Resources for compiling AD-associated genes. No. of Resources Genes Filtering Criteria genes PSEN2, HFE, NOS3, PLAU, CALHM1, A2M, PSEN1, OMIM3 All genes associated with AD 12 ADAM10, MPO, ABCA7, APOE, APP APOC1, BCL3, MS4A4A, FRMD4A, GAB2, MTHFD1L, CR1, EPHA1, CLU, PICALM, Genome-wide significant association DistiLD5 19 SORL1, CD33, ABCA7, with AD (p-value < 5.0×10-8) CEACAM16, GLIS3, CD2AP, SRRM4, BCHE, ZNF292 CLU, TNK1, APOE, SOAT1, PVRL2, TRAK2, CHRNB2, TFAM, CTSD, IL33, THRA, IL1RN, IL10, PICALM, STH, IL6, CD33, TFCP2, VLDLR, SORCS1, IL1B, GAB2, ABCA1, CTNNA3, APOC2, IL1A, BIN1, DAPK1, PON1, CST3, CCR2, The top ranked AD genes provided AlzGene11 GAPDHS, IL8, GRN, ECE1, 68 on the AlzGene website. OLR1, TF, LPL, LRP1, HHEX, GAPDH, LDLR, OTC, ARID5B, TNF, TOMM40, EXOC3L2, PGBD1, PRNP, BCAM, CALHM1, UBQLN1, PCK1, NEDD9, SORL1, ADAM10, MME, ENTPD7, FAS, CH25H, APOC1, ACE, CYP19A1, NCAPD2, MTHFR, APOC4, CHAT, CR1 APP, PSEN1, PSEN2, MAPT, APOE, TREM2, PLD3, SORL1, NOTCH3, CLU, PICALM, CR1, ABCA7, BIN1, CD33, CD2AP, MS4A6A, MS4A4E, NME8, Top ranked genes supported by AlzBase2 32 EPHA1, PTK2B, SLC24A4, genetic evidences CELF1, FERMT2, ZCWPW1, INPP5D, HLA-DRB1, HLA- DRB5, MEF2C, TP53INP1, CASS4, IGHV1-67 PSEN1, APP, PSEN2, APOE, Genes associated with AD and score DisGenet12 SORL1, MAPT, BDNF, IL1B, 12 > 0.3 BACE1, ACE, GSK3B, PLAU ADAM10, APOE, APP, MT-ND1, UniProt 6 MT-ND2, PSEN1, PSEN2, All genes associated with AD 8 SORL1 SLC30A6(ZNT6)13, 14, SLC30A4(ZNT4)13, 14, MAOB15, 16, CHRNA717, 18, IGF119, 20, VEGFA21, 22, ARC23, 24, PPARG25, 26, BCL227, 28, 29, IGF2R30, CASP331, 32, NECTIN2 (PVRL2)33, 34, 35, IGF230, 36, 37, VSNL138, 39, ACHE40, 41, 42, CYP46A143, 44, 45, IDE46, 47, 1. For GWAS studies: p- NPY48, 49, 50, IGF1R20, 51, value<5.0×10-8 Published PM20D152, APBB253, 54, ESR155, 2. For differential 56 literature 56, 57, PAXIP158, 59, CRH60, 61, studies on AD, the gene which is SOD262, 63, 64, SLC2A465, 66, differentially expressed in at least HMOX167, 68, DPYSL269, 70, two studies were included. INSR71, 72, DHCR2473, 74, 75, RELN76, 77, 78, 79, 80, BAX81, 82, 83, REST84, MEOX285, 86, EIF2AK387, 88, MAN2A189, 90, (ABI3, PLCG2)91, 92, DSG226, 93, MS4A294, 95,PPP1R3796, RELB33, 97, TREML298, (HESX1, ADAMTS4, CLNK, HS3ST1, CNTNAP2, ADAM10, APH1B, KAT8, ABI3, ALPK2, ECHDC3, SCIMP, SUZ12P1)99 p-value<5.0×10-8; from both GWAS- GWAS reported and GWAS-mapped genes 265 catalog https://www.ebi.ac.uk/gwas as provided in the GWAS catalog database. Open Genes with association score with AD 175 Targets7 https://www.opentargets.org/ > 0.5 ROSMAP See ref9 All DEGs with FDR<0.05 2614

Table 2. The list of 147 AD-associated genes. AD-associated genes APOE, SORL1, GAB2, CR1, PICALM, CLU, CD33, ABCA7, ADAM10, CD2AP, BIN1, APOC1, TOMM40, INPP5D, PSEN2, EPHA1, APP, MTHFD1L, CNTNAP2, HLA-DRB1, CASS4, BCAM, ABCA1, PTK2B, MS4A6A, FRMD4A, BCL3, SLC24A4, GLIS3, FERMT2, PSEN1, TREM2, ZCWPW1, EXOC3L2, MS4A4A, ACE, APOC4, BZW2, SUCLG2, APOB, SCIMP, SCARB1, RELB, CRY2, PVRL2, CLASRP, ADAMTS4, MMP3, UBE2L3, PPP1R37, ECHDC3, TCF7L2, IL6R, MS4A2, LIPG, MAN2A1, MAPT, ALDH1A2, ABI3, LILRA5, CELF1, PLCG2, HMGCR, OARD1, APH1B, APOC2, OR4S1, STAT4, MS4A4E, PVR, MT-ND2, HS3ST1, CCR2, VASP, CYP8B1, BLOC1S3, PPP1R13L, NFIC, NKPD1, INSR, CNTNAP5, BCAS3, BCHE, BCL2, NME8, CLPTM1, CLNK, UBQLN1, CLMN, IL1B, TRAPPC6A, VSNL1, SORCS1, PPARG, IGSF23, CRH, PSMA1, CHRNB2, FBXL7, CHRNA7, SPON1, MYO16, CHRNA2, VLDLR, KIR3DL2, KIT, HLA-DRB5, BACE1, HLA- DRA, DSG2, CALHM1, RBFOX1, HFE, PILRA, LRP4, HARBI1, TFCP2, CBLC, DPP10, SYNJ1, CDC25B, ACP2, ACHE, PACSIN3, MADD, ZNF652, GSK3B, PFDN1, RIN3, MARK4, GRIN2A, PDGFRB, MAPK8IP1, GRIN3B, CCRL2, ECE1, SCN1A, HBEGF, CACNA1G, CEACAM16, MMP13, ESR1, ALDH5A1, PLAU, SCN8A, CACNA2D1, MMP12

References

1. Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet 39, 17–23 (2007).

2. Bai Z, et al. AlzBase: an integrative database for gene dysregulation in Alzheimer’s disease. Mol Neurobiol 53, 310–319 (2016).

3. Hamosh A, Scott AF, Amberger JS, Bocchini CA, McKusick VA. Online Mendelian Inheritance in Man (OMIM), a knowledgebase of human genes and genetic disorders. Nucleic Acids Res 33, D514–D517 (2005).

4. Pinero J, et al. DisGeNET: a discovery platform for the dynamical exploration of human diseases and their genes. Database (Oxford) 2015, bav028 (2015).

5. Palleja A, Horn H, Eliasson S, Jensen LJ. DistiLD Database: diseases and traits in linkage disequilibrium blocks. Nucleic Acids Res 40, D1036–D1040 (2012).

6. Wu CH, et al. The Universal Resource (UniProt): an expanding universe of protein information. Nucleic Acids Res 34, D187–D191 (2006).

7. Koscielny G, et al. Open Targets: a platform for therapeutic target identification and validation. Nucleic Acids Research 45, D985-D994 (2016).

8. MacArthur J, et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Research 45, D896-D901 (2016).

9. Canchi S, et al. Integrating Gene and Protein Expression Reveals Perturbed Functional Networks in Alzheimer’s Disease. Cell Reports 28, 1103-1116.e1104 (2019).

10. Mi H, Muruganujan A, Ebert D, Huang X, Thomas PD. PANTHER version 14: more genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res 47, D419–D426 (2018).

11. Alzforum. https://www.alzforum.org.

12. Piñero J, et al. DisGeNET: a comprehensive platform integrating information on human disease-associated genes and variants. Nucleic Acids Research 45, D833-D839 (2016).

13. Lyubartseva G, Smith JL, Markesbery WR, Lovell MA. Alterations of zinc transporter ZnT-1, ZnT-4 and ZnT-6 in preclinical Alzheimer's disease . Brain pathology (Zurich, Switzerland) 20, 343-350 (2010).

14. Smith JL, Xiong S, Markesbery WR, Lovell MA. Altered expression of zinc transporters-4 and -6 in mild cognitive impairment, early and late Alzheimer’s disease brain. Neuroscience 140, 879-888 (2006).

15. Schedin-Weiss S, et al. Monoamine oxidase B is elevated in Alzheimer disease neurons, is associated with γ-secretase and regulates neuronal β-peptide levels. Alzheimer's Research & Therapy 9, 57 (2017).

16. Riederer P, Danielczyk W, Grünblatt E. Monoamine Oxidase-B Inhibition in Alzheimer’s Disease. NeuroToxicology 25, 271-277 (2004).

17. Clarelli F, et al. CHRNA7 Gene and Response to Inhibitors in an Italian Cohort of Alzheimer's Disease Patients. J Alzheimers Dis 52, 1203-1208 (2016).

18. Carson R, et al. Alpha7 nicotinic acetylcholine receptor gene and reduced risk of Alzheimer's disease. J Med Genet 45, 244-248 (2008).

19. Westwood AJ, et al. Insulin-like growth factor-1 and risk of Alzheimer dementia and brain atrophy. Neurology 82, 1613-1619 (2014).

20. Ostrowski PP, Barszczyk A, Forstenpointner J, Zheng W, Feng Z-P. Meta-Analysis of Serum Insulin-Like Growth Factor 1 in Alzheimer’s Disease. PLOS ONE 11, e0155733 (2016).

21. Chiappelli M, et al. VEGF gene and phenotype relation with Alzheimer's disease and mild cognitive impairment. Rejuvenation research 9, 485-493 (2006).

22. Del Bo R, Ghezzi S, Scarpini E, Bresolin N, Comi GP. VEGF genetic variability is associated with increased risk of developing Alzheimer's disease. Journal of the neurological sciences 283, 66-68 (2009).

23. Bi R, et al. The Arc Gene Confers Genetic Susceptibility to Alzheimer's Disease in Han Chinese. Mol Neurobiol 55, 1217-1226 (2018).

24. Ridge PG, 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).

25. Jiang Q, Heneka M, Landreth GE. The role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in Alzheimer's disease: therapeutic implications. CNS drugs 22, 1-14 (2008).

26. Barrera J, Subramanian S, Chiba-Falek O. Probing the role of PPARγ in the regulation of late-onset Alzheimer’s disease-associated genes. PLOS ONE 13, e0196943 (2018).

27. O'Barr S, Schultz J, Rogers J. Expression of the protooncogene bcl-2 in Alzheimer's disease brain. Neurobiol Aging 17, 131-136 (1996).

28. Paradis E, Douillard H, Koutroumanis M, Goodyer C, LeBlanc A. Amyloid β Peptide of Alzheimer’s Disease Downregulates Bcl-2 and Upregulates Bax Expression in Human Neurons. The Journal of Neuroscience 16, 7533 (1996).

29. Sajan FD, et al. Apoptotic gene expression in Alzheimer's disease hippocampal tissue. American journal of Alzheimer's disease and other dementias 22, 319-328 (2007).

30. Pascual-Lucas M, et al. Insulin-like growth factor 2 reverses memory and synaptic deficits in APP transgenic mice. EMBO molecular medicine 6, 1246-1262 (2014).

31. Su JH, Zhao M, Anderson AJ, Srinivasan A, Cotman CW. Activated caspase-3 expression in Alzheimer's and aged control brain: correlation with Alzheimer pathology. Brain research 898, 350-357 (2001).

32. D'Amelio M, et al. Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer's disease. Nat Neurosci 14, 69-76 (2011).

33. Xiao E, et al. Late-Onset Alzheimer’s Disease Polygenic Risk Profile Score Predicts Hippocampal Function. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging 2, 673-679 (2017).

34. Yashin AI, et al. Hidden heterogeneity in Alzheimer's disease: Insights from genetic association studies and other analyses. Exp Gerontol 107, 148-160 (2018).

35. Takei N, et al. Genetic association study on in and around the APOE in late-onset Alzheimer disease in Japanese. Genomics 93, 441-448 (2009).

36. Williams DM, Karlsson IK, Pedersen NL, Hägg S. Circulating insulin-like growth factors and Alzheimer disease: A mendelian randomization study. Neurology 90, e291-e297 (2018).

37. Mellott TJ, Pender SM, Burke RM, Langley EA, Blusztajn JK. IGF2 ameliorates amyloidosis, increases cholinergic marker expression and raises BMP9 and neurotrophin levels in the hippocampus of the APPswePS1dE9 Alzheimer's disease model mice. PLoS One 9, e94287 (2014).

38. Lin C-W, Chang L-C, Tseng GC, Kirkwood CM, Sibille EL, Sweet RA. VSNL1 Co- Expression Networks in Aging Include Calcium Signaling, Synaptic Plasticity, and Alzheimer's Disease Pathways. Frontiers in psychiatry 6, 30-30 (2015).

39. Gómez Ravetti M, Rosso OA, Berretta R, Moscato P. Uncovering Molecular Biomarkers That Correlate Cognitive Decline with the Changes of Hippocampus' Gene Expression Profiles in Alzheimer's Disease. PLOS ONE 5, e10153 (2010).

40. McGleenon BM, Dynan KB, Passmore AP. inhibitors in Alzheimer's disease. British journal of clinical pharmacology 48, 471-480 (1999).

41. Lanctôt KL, Herrmann N, LouLou MM. Correlates of response to acetylcholinesterase inhibitor therapy in Alzheimer's disease. Journal of psychiatry & neuroscience : JPN 28, 13-26 (2003).

42. Lane RM, Kivipelto M, Greig NH. Acetylcholinesterase and its inhibition in Alzheimer disease. Clinical neuropharmacology 27, 141-149 (2004).

43. Kolsch H, et al. CYP46A1 variants influence Alzheimer's disease risk and brain cholesterol metabolism. European psychiatry : the journal of the Association of European Psychiatrists 24, 183-190 (2009).

44. Djelti F, et al. CYP46A1 inhibition, brain cholesterol accumulation and neurodegeneration pave the way for Alzheimer's disease. Brain 138, 2383-2398 (2015).

45. Li L, et al. CYP46A1 and the APOEepsilon4 Allele Polymorphisms Correlate with the Risk of Alzheimer's Disease. Mol Neurobiol 55, 8179-8187 (2018).

46. Carrasquillo MM, et al. Concordant Association of Insulin Degrading Gene (IDE) Variants with IDE mRNA, Aß, and Alzheimer's Disease. PLOS ONE 5, e8764 (2010).

47. Vepsäläinen S, et al. Insulin-degrading enzyme is genetically associated with Alzheimer's disease in the Finnish population. Journal of medical genetics 44, 606- 608 (2007).

48. Spencer B, et al. Systemic Central Nervous System (CNS)-targeted Delivery of Neuropeptide Y (NPY) Reduces Neurodegeneration and Increases Neural Precursor Cell Proliferation in a Mouse Model of Alzheimer Disease. The Journal of biological chemistry 291, 1905-1920 (2016).

49. Minthon L, Edvinsson L, Gustafson L. Correlation between clinical characteristics and cerebrospinal fluid neuropeptide Y levels in dementia of the Alzheimer type and frontotemporal dementia. Alzheimer disease and associated disorders 10, 197-203 (1996).

50. Koide S, Onishi H, Hashimoto H, Kai T, Yamagami S. Plasma neuropeptide Y is reduced in patients with Alzheimer's disease. Neurosci Lett 198, 149-151 (1995).

51. George C, Gontier G, Lacube P, Francois JC, Holzenberger M, Aid S. The Alzheimer's disease transcriptome mimics the neuroprotective signature of IGF-1 receptor- deficient neurons. Brain 140, 2012-2027 (2017).

52. Sanchez-Mut JV, et al. PM20D1 is a quantitative trait associated with Alzheimer’s disease. Nature Medicine 24, 598-603 (2018).

53. Golanska E, et al. Analysis of APBB2 gene polymorphisms in sporadic Alzheimer's disease. Neurosci Lett 447, 164-166 (2008).

54. Li Y, et al. Genetic association of the APP binding protein 2 gene (APBB2) with late onset Alzheimer disease. Human mutation 25, 270-277 (2005).

55. Luckhaus C, Sand PG. Estrogen Receptor 1 gene (ESR1) variants in Alzheimer's disease. Results of a meta-analysis. Aging clinical and experimental research 19, 165- 168 (2007).

56. Elcoroaristizabal Martin X, et al. Progression from amnesic mild cognitive impairment to Alzheimer's disease: ESR1 and ESR2 polymorphisms and APOE gene. Dementia and geriatric cognitive disorders 32, 332-341 (2011).

57. Goumidi L, et al. Study of estrogen receptor-alpha and receptor-beta gene polymorphisms on Alzheimer's disease. J Alzheimers Dis 26, 431-439 (2011).

58. Rademakers R, et al. Linkage and Association Studies Identify a Novel Locus for Alzheimer Disease at 7q36 in a Dutch Population-Based Sample. The American Journal of Human Genetics 77, 643-652 (2005).

59. Chen JA, et al. A multiancestral genome-wide exome array study of Alzheimer disease, frontotemporal dementia, and progressive supranuclear palsy. JAMA neurology 72, 414-422 (2015).

60. Pedersen WA, McCullers D, Culmsee C, Haughey NJ, Herman JP, Mattson MP. Corticotropin-releasing hormone protects neurons against insults relevant to the pathogenesis of Alzheimer's disease. Neurobiology of disease 8, 492-503 (2001).

61. Futch HS, Croft CL, Truong VQ, Krause EG, Golde TE. Targeting psychologic stress signaling pathways in Alzheimer’s disease. Molecular Neurodegeneration 12, 49 (2017).

62. Massaad CA, Washington TM, Pautler RG, Klann E. Overexpression of SOD-2 reduces hippocampal superoxide and prevents memory deficits in a mouse model of Alzheimer's disease. Proceedings of the National Academy of Sciences 106, 13576 (2009).

63. Flynn JM, Melov S. SOD2 in mitochondrial dysfunction and neurodegeneration. Free radical biology & medicine 62, 4-12 (2013).

64. Wiener HW, Perry RT, Chen Z, Harrell LE, Go RC. A polymorphism in SOD2 is associated with development of Alzheimer's disease. Genes, brain, and behavior 6, 770-775 (2007).

65. Wang X, et al. Genetic determinants of disease progression in Alzheimer's disease. Journal of Alzheimer's disease : JAD 43, 649-655 (2015).

66. Katsel P, et al. Parahippocampal gyrus expression of endothelial and insulin receptor signaling pathway genes is modulated by Alzheimer’s disease and normalized by treatment with anti-diabetic agents. PLOS ONE 13, e0206547 (2018).

67. Sung HY, Choi BO, Jeong JH, Kong KA, Hwang J, Ahn JH. -Mediated Hypomethylation of Oxygenase 1 Correlates with Cognitive Impairment in Alzheimer's Disease. PLoS One 11, e0153156 (2016).

68. Schipper HM, Song W. A heme oxygenase-1 transducer model of degenerative and developmental brain disorders. Int J Mol Sci 16, 5400-5419 (2015).

69. Silva PN, et al. CNP and DPYSL2 mRNA expression and promoter methylation levels in brain of Alzheimer's disease patients. J Alzheimers Dis 33, 349-355 (2013).

70. Lubec G, Nonaka M, Krapfenbauer K, Gratzer M, Cairns N, Fountoulakis M. Expression of the dihydropyrimidinase related protein 2 (DRP-2) in Down syndrome and Alzheimer's disease brain is downregulated at the mRNA and dysregulated at the protein level. Journal of neural transmission Supplementum 57, 161-177 (1999).

71. da Costa IB, et al. Change in INSR, APBA2 and IDE Gene Expressions in of Alzheimer's Disease Patients. Curr Alzheimer Res 14, 760-765 (2017).

72. Ling J, Yang S, Huang Y, Wei D, Cheng W. Identifying key genes, pathways and screening therapeutic agents for manganese-induced Alzheimer disease using bioinformatics analysis. 97, e10775 (2018).

73. Wang Y, et al. The selective Alzheimer's disease indicator-1 gene (Seladin-1/DHCR24) is a liver X receptor target gene. Molecular pharmacology 74, 1716-1721 (2008).

74. Feher A, Juhasz A, Pakaski M, Kalman J, Janka Z. Gender dependent effect of DHCR24 polymorphism on the risk for Alzheimer's disease. Neurosci Lett 526, 20-23 (2012).

75. Crameri A, et al. The role of seladin-1/DHCR24 in cholesterol biosynthesis, APP processing and Abeta generation in vivo. The EMBO journal 25, 432-443 (2006).

76. Seripa D, et al. The RELN locus in Alzheimer's disease. J Alzheimers Dis 14, 335-344 (2008).

77. Herring A, et al. Reelin depletion is an early phenomenon of Alzheimer's pathology. J Alzheimers Dis 30, 963-979 (2012).

78. Bufill E, et al. Reelin signaling pathway genotypes and Alzheimer disease in a Spanish population. Alzheimer disease and associated disorders 29, 169-172 (2015).

79. Cuchillo-Ibañez I, Mata-Balaguer T, Balmaceda V, Arranz JJ, Nimpf J, Sáez-Valero J. The β-amyloid peptide compromises Reelin signaling in Alzheimer’s disease. Scientific Reports 6, 31646 (2016).

80. Kocherhans S, et al. Reduced Reelin Expression Accelerates Amyloid-β Plaque Formation and Tau Pathology in Transgenic Alzheimer's Disease Mice. The Journal of Neuroscience 30, 9228 (2010).

81. Su JH, Deng G, Cotman CW. Bax protein expression is increased in Alzheimer's brain: correlations with DNA damage, Bcl-2 expression, and brain pathology. J Neuropathol Exp Neurol 56, 86-93 (1997).

82. Kudo W, Lee HP, Smith MA, Zhu X, Matsuyama S, Lee Hg. Inhibition of Bax protects neuronal cells from oligomeric Aβ neurotoxicity. Cell Death &Amp; Disease 3, e309 (2012).

83. Paradis E, Douillard H, Koutroumanis M, Goodyer C, LeBlanc A. Amyloid beta peptide of Alzheimer's disease downregulates Bcl-2 and upregulates bax expression in human neurons. J Neurosci 16, 7533-7539 (1996).

84. Meyer K, et al. REST and Neural Gene Network Dysregulation in iPSC Models of Alzheimer’s Disease. Cell Reports 26, 1112-1127.e1119 (2019).

85. Wang Y, et al. Generation of induced pluripotent stem cell line (ZZUi0013-A) from a 65-year-old patient with a novel MEOX2 gene mutation in Alzheimer's disease. Stem Cell Research 34, 101366 (2019).

86. Soto I, et al. Meox2 haploinsufficiency increases neuronal cell loss in a mouse model of Alzheimer's disease. Neurobiology of Aging 42, 50-60 (2016).

87. Wong TH, et al. EIF2AK3 variants in Dutch patients with Alzheimer's disease. Neurobiology of Aging 73, 229.e211-229.e218 (2019).

88. Liu Q-Y, et al. An exploratory study on STX6, MOBP, MAPT, and EIF2AK3 and late- onset Alzheimer's disease. Neurobiology of Aging 34, 1519.e1513-1519.e1517 (2013).

89. DeMichele-Sweet MAA, et al. Genetic risk for schizophrenia and psychosis in Alzheimer disease. Molecular psychiatry 23, 963-972 (2018).

90. Sherva R, et al. Genome-wide association study of the rate of cognitive decline in Alzheimer's disease. Alzheimer's & dementia : the journal of the Alzheimer's Association 10, 45-52 (2014).

91. Sims R, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial- mediated innate immunity in Alzheimer’s disease. Nature Genetics 49, 1373 (2017).

92. Conway OJ, et al. ABI3 and PLCG2 missense variants as risk factors for neurodegenerative diseases in Caucasians and African Americans. Molecular Neurodegeneration 13, 53 (2018).

93. Kim H, et al. Modelling APOE ɛ3/4 allele-associated sporadic Alzheimer’s disease in an induced neuron. Brain 140, 2193-2209 (2017).

94. Carter C. Alzheimer's Disease: APP, , APOE, CLU, CR1, PICALM, ABCA7, BIN1, CD2AP, CD33, EPHA1, and MS4A2, and Their Relationships with Herpes Simplex, C. Pneumoniae, Other Suspect Pathogens, and the Immune System. International journal of Alzheimer's disease 2011, 501862-501862 (2011).

95. Raj T, et al. Alzheimer disease susceptibility loci: evidence for a protein network under natural selection. American journal of human genetics 90, 720-726 (2012).

96. Han Z, Huang H, Gao Y, Huang Q. Functional annotation of Alzheimer's disease associated loci revealed by GWASs. PloS one 12, e0179677-e0179677 (2017).

97. Nho K, et al. Association analysis of rare variants near the APOE region with CSF and neuroimaging biomarkers of Alzheimer’s disease. BMC Medical Genomics 10, 29 (2017).

98. Benitez BA, et al. Missense variant in TREML2 protects against Alzheimer's disease. Neurobiology of aging 35, 1510.e1519-1510.e1526 (2014).

99. Jansen IE, et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nature Genetics 51, 404-413 (2019).