Prediction of Alzheimer's disease using multi-variants from a Chinese genome-wide association study

Longfei Jia, Fangyu Li, Cuibai Wei, Min Zhu, Qiumin Qu, Wei Qin, Yi Tang, Luxi Shen, Yanjiang Wang, Lu Shen, Honglei Li, Dantao Peng, Lan Tan, Benyan Luo, Qihao Guo, Muni Tang, Yifeng Du, Jiewen Zhang, Junjian Zhang, Jihui Lyu, Ying Li, Aihong Zhou, Fen Wang, Changbiao Chu, Haiqing Song, Liyong Wu, Xiumei Zuo, Yue Han, Junhua Liang, Qi Wang, Hongmei Jin, Wei Wang, Yang Lü, Fang Li, Yuying Zhou, Wei Zhang, Zhengluan Liao, Qiongqiong Qiu, Yan Li, Chaojun Kong, Yan Li, Haishan Jiao, Jie Lu, Jianping Jia, Longfei Jia, Fangyu Li, Cuibai Wei, Min Zhu, Qiumin Qu, Wei Qin, Yi Tang, Luxi Shen, Yanjiang Wang, Lu Shen, Honglei Li, Dantao Peng, Lan Tan, Benyan Luo, Qihao Guo, Muni Tang, Yifeng Du, Jiewen Zhang, Junjian Zhang, Jihui Lyu, Ying Li, Aihong Zhou, Fen Wang, Changbiao Chu, Haiqing Song, Liyong Wu, Xiumei Zuo, Yue Han, Junhua Liang, Qi Wang, Hongmei Jin, Wei Wang, Yang Lü, Fang Li, Yuying Zhou, Wei Zhang, Zhengluan Liao, Qiongqiong Qiu, Yan Li, Chaojun Kong, Yan Li, Haishan Jiao, Jie Lu, Jianping Jia

Abstract

Previous genome-wide association studies have identified dozens of susceptibility loci for sporadic Alzheimer's disease, but few of these loci have been validated in longitudinal cohorts. Establishing predictive models of Alzheimer's disease based on these novel variants is clinically important for verifying whether they have pathological functions and provide a useful tool for screening of disease risk. In the current study, we performed a two-stage genome-wide association study of 3913 patients with Alzheimer's disease and 7593 controls and identified four novel variants (rs3777215, rs6859823, rs234434, and rs2255835; Pcombined = 3.07 × 10-19, 2.49 × 10-23, 1.35 × 10-67, and 4.81 × 10-9, respectively) as well as nine variants in the apolipoprotein E region with genome-wide significance (P < 5.0 × 10-8). Literature mining suggested that these novel single nucleotide polymorphisms are related to amyloid precursor protein transport and metabolism, antioxidation, and neurogenesis. Based on their possible roles in the development of Alzheimer's disease, we used different combinations of these variants and the apolipoprotein E status and successively built 11 predictive models. The predictive models include relatively few single nucleotide polymorphisms useful for clinical practice, in which the maximum number was 13 and the minimum was only four. These predictive models were all significant and their peak of area under the curve reached 0.73 both in the first and second stages. Finally, these models were validated using a separate longitudinal cohort of 5474 individuals. The results showed that individuals carrying risk variants included in the models had a shorter latency and higher incidence of Alzheimer's disease, suggesting that our models can predict Alzheimer's disease onset in a population with genetic susceptibility. The effectiveness of the models for predicting Alzheimer's disease onset confirmed the contributions of these identified variants to disease pathogenesis. In conclusion, this is the first study to validate genome-wide association study-based predictive models for evaluating the risk of Alzheimer's disease onset in a large Chinese population. The clinical application of these models will be beneficial for individuals harbouring these risk variants, and particularly for young individuals seeking genetic consultation.

Keywords: Alzheimer’s disease; Chinese; genome-wide association study; longitudinal cohort; predictive model.

© The Author(s) (2020). Published by Oxford University Press on behalf of the Guarantors of Brain.

Figures

Figure 1
Figure 1
Study flow chart. AD = Alzheimer’s disease.
Figure 2
Figure 2
Regional association plots. (AD) Association results are shown for the analysed SNPs with recombination rates in the four loci associated with genome-wide significance at chromosome 5 (A and B), 14 (C), and 21 (D). The −log10 (P-values) (y-axis) of SNPs within the ±500 kb region centred on each marker SNP are presented according to the chromosomal positions of the SNPs (x-axis; NCBI Build 37). Purple diamonds represent the most significantly associated SNP (marker SNP) in the combined analysis. SNPs are coloured according to their linkage disequilibrium with the marker SNP. Linkage disequilibrium values were based on the 1000 Genome Project Asian data. Blue lines represent the estimated recombination rates based on the 1000 Genome Project samples. Arrows depict genes in the regions of interest annotated from the UCSC Genome Browser.
Figure 3
Figure 3
Differential expression of the annotated genes in Gene Expression Omnibus datasets. (AC) shows the differential expression of RHOBTB3 (A), GLRX (B), CHODL (C) in frontal cortex, hippocampus, and temporal cortex. The bold red line indicates the median of each group, and the black dotted lines show the quartiles. AD = Alzheimer’s disease; CN = cognitively normal; FC = fold change; GSE = Gene Expression Omnibus Series; ns = no significance; *P <0.05; **P <0.01; ***P <0.001; ****P <0.0001.
Figure 4
Figure 4
ROC curves for 11 predictive models with different predictors in the three cohorts and survival curves in a longitudinal cohort. The factors included in the 11 models are as follows. (A) A1: APOE ε4 status, rs3777215, rs6859823, rs234434 and rs2255835; (B) A2: APOE ε4 status, rs3777215, rs6859823, rs234434, rs2255835, rs11668861, rs71352238 and rs4420638; (C) A3: APOE ε4 status, rs3777215, rs6859823, rs234434, rs2255835, rs11668861, rs6859, rs3852860, rs71352238, rs157580, rs2075650, rs157582, rs439401 and rs4420638; (D) A4: rs3777215, rs6859823, rs234434, rs2255835, rs11668861, rs6859, rs3852860, rs71352238, rs157580, rs2075650, rs157582, rs439401 and rs4420638; (E) B1: rs3777215, rs6859823, rs234434 and rs2255835; (F) B2: rs3777215, rs6859823, rs234434, rs2255835, rs11668861, rs71352238 and rs4420638; (G) B3: rs3777215, rs6859823, rs234434, rs71352238 and rs4420638; (H) B4: rs3777215, rs234434, rs71352238 and rs4420638; (I) B5: rs6859823, rs234434, rs71352238 and rs4420638; (J) B6: rs3777215, rs6859823, rs234434 and rs71352238; (K) B7: rs3777215, rs6859823, rs234434 and rs4420638. (L) Survival curves of the longitudinal cohort, *P <0.001. AUC1 indicates AUC of the first stage; AUC2 indicates AUC of the second stage; AUC3 indicates AUC of the longitudinal cohort; ROC = receiver operating characteristic curve.

References

    1. Akterin S, Cowburn RF, Miranda-Vizuete A, Jimenez A, Bogdanovic N, Winblad B, et al.Involvement of glutaredoxin-1 and thioredoxin-1 in beta-amyloid toxicity and Alzheimer's disease. Cell Death Differ 2006; 13: 1454–65.
    1. Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ.. Second-generation PLINK: rising to the challenge of larger and richer datasets. GigaSci 2015; 4: 7.
    1. Chaudhury S, Brookes KJ, Patel T, Fallows A, Guetta-Baranes T, Turton JC, et al.Alzheimer's disease polygenic risk score as a predictor of conversion from mild-cognitive impairment. Transl Psychiatry 2019; 9: 154.
    1. Chelala C, Khan A, Lemoine NR.. SNPnexus: a web database for functional annotation of newly discovered and public domain single nucleotide polymorphisms. Bioinformatics 2009; 25: 655–61.
    1. Chen J, Zheng H, Bei JX, Sun L, Jia WH, Li T, et al.Genetic structure of the Han Chinese population revealed by genome-wide SNP variation. Am J Hum Genet 2009; 85: 775–85.
    1. Choi SW, O'Reilly PF.. PRSice-2: polygenic Risk Score software for biobank-scale data. Gigascience 2019; 8:1–6. doi: 10.1093/gigascience/giz082.
    1. Chouraki V, Reitz C, Maury F, Bis JC, Bellenguez C, Yu L, et al.Evaluation of a genetic risk score to improve risk prediction for Alzheimer's disease. J Alzheimers Dis 2016; 53: 921–32.
    1. Dayem Ullah AZ, Lemoine NR, Chelala C.. SNPnexus: a web server for functional annotation of novel and publicly known genetic variants (2012 update). Nucleic Acids Res 2012; 40(Web Server issue): W65–70.
    1. Dayem Ullah AZ, Lemoine NR, Chelala C.. A practical guide for the functional annotation of genetic variations using SNPnexus. Brief Bioinform 2013; 14: 437–47.
    1. Dayem Ullah AZ, Oscanoa J, Wang J, Nagano A, Lemoine NR, Chelala C.. SNPnexus: assessing the functional relevance of genetic variation to facilitate the promise of precision medicine. Nucleic Acids Res 2018; 46: W109–W13.
    1. Delaneau O, Marchini J, Zagury JF.. A linear complexity phasing method for thousands of genomes. Nat Methods 2011; 9: 179–81.
    1. Desikan RS, Schork AJ, Wang Y, Thompson WK, Dehghan A, Ridker PM, et al.Polygenic overlap between C-reactive protein, plasma lipids, and Alzheimer disease. Circulation 2015; 131: 2061–9.
    1. Ellingson SR, Fardo DW.. Automated quality control for genome wide association studies. F1000Res 2016; 5: 1889.
    1. Escott-Price V, Myers A, Huentelman M, Shoai M, Hardy J.. Polygenic risk score analysis of Alzheimer's disease in cases without APOE4 or APOE2 alleles. J Prev Alzheimers Dis 2019; 6: 16–9.
    1. Escott-Price V, Myers AJ, Huentelman M, Hardy J.. Polygenic risk score analysis of pathologically confirmed Alzheimer disease. Ann Neurol 2017. a; 82: 311–4.
    1. 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 2017. b; 49: 214 e7–e11.
    1. Escott-Price V, Sims R, Bannister C, Harold D, Vronskaya M, Majounie E, et al.Common polygenic variation enhances risk prediction for Alzheimer's disease. Brain 2015; 138: 3673–84.
    1. Espinosa EJ, Calero M, Sridevi K, Pfeffer SR.. RhoBTB3: a Rho GTPase-family ATPase required for endosome to Golgi transport. Cell 2009; 137: 938–48.
    1. Gatz M, Pedersen NL, Berg S, Johansson B, Johansson K, Mortimer JA, et al.Heritability for Alzheimer's disease: the study of dementia in Swedish twins. J Gerontol A Biol Sci Med Sci 1997; 52: M117–M25.
    1. Gauderman WJ, Morrison JM, Morrison WGJ. QUANTO 1.1: A computer program for power and sample size calculations for genetic-epidemiology studies [Internet]. 2006. Available from:
    1. Genomes Project CAbecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, et integrated map of genetic variation from 1,092 human genomes. Nature 2012; 491: 56–65.
    1. Giambartolomei C, Vukcevic D, Schadt EE, Franke L, Hingorani AD, Wallace C, et al.Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet 2014; 10: e1004383.
    1. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, et al.TREM2 variants in Alzheimer's disease. N Engl J Med 2013; 368: 117–27.
    1. Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, et al.Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease. Nat Genet 2009; 41: 1088–93.
    1. Higgins JP, Thompson SG.. Quantifying heterogeneity in a meta-analysis. Stat Med 2002; 21: 1539–58.
    1. Higgins JPT, Thompson SG, Deeks JJ, Altman DG.. Measuring inconsistency in meta-analyses. Bmj 2003; 327: 557–60.
    1. Hollingworth P, Harold D, Sims R, Gerrish A, Lambert J-C, Carrasquillo MM, et al.Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat Genet 2011; 43: 429–35.
    1. Howie BN, Donnelly P, Marchini J.. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet 2009; 5: e1000529.
    1. Jia J, Wang F, Wei C, Zhou A, Jia X, Li F, et al.The prevalence of dementia in urban and rural areas of China. Alzheimers Dement 2014; 10: 1–9.
    1. Jun G, Ibrahim-Verbaas CA, Vronskaya M, Lambert JC, Chung J, Naj AC, et al.A novel Alzheimer disease locus located near the gene encoding tau protein. Mol Psychiatry 2016; 21: 108–17.
    1. Jun G, Vardarajan BN, Buros J, Yu CE, Hawk MV, Dombroski BA, et al.Comprehensive search for Alzheimer disease susceptibility loci in the APOE region. Arch Neurol 2012; 69: 1270–9.
    1. Kommaddi RP, Tomar DS, Karunakaran S, Bapat D, Nanguneri S, Ray A, et al.Glutaredoxin1 diminishes amyloid beta-mediated oxidation of F-actin and reverses cognitive deficits in an Alzheimer's disease mouse model. Antioxid Redox Signal 2019; 31: 1321–38.
    1. Kunkle BW, Grenier-Boley B, Sims R, Bis JC, Damotte V, Naj AC, et al.Genetic meta-analysis of diagnosed Alzheimer's disease identifies new risk loci and implicates Abeta, tau, immunity and lipid processing. Nat Genet 2019; 51: 414–30.
    1. Lacour A, Espinosa A, Louwersheimer E, Heilmann S, Hernandez I, Wolfsgruber S, et al.Genome-wide significant risk factors for Alzheimer's disease: role in progression to dementia due to Alzheimer's disease among subjects with mild cognitive impairment. Mol Psychiatry 2017; 22: 153–60.
    1. Lambert J-C, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, et al.Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease. Nat Genet 2009; 41: 1094–9.
    1. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, et al.Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat Genet 2013. a; 45: 1452–8.
    1. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, et al.Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat Genet 2013. b; 45: 1452–8.
    1. Leonenko G, Sims R, Shoai M, Frizzati A, Bossu P, Spalletta G, et al.Polygenic risk and hazard scores for Alzheimer's disease prediction. Ann Clin Transl Neurol 2019; 6: 456–65.
    1. Lloyd-Jones LR, Holloway A, McRae A, Yang J, Small K, Zhao J, et al.The genetic architecture of gene expression in peripheral blood. Am J Hum Genet 2017; 100: 228–37.
    1. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM.. Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's Disease. Neurology 1984; 34: 939–44.
    1. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr., Kawas CH, et al.The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 2011; 7: 263–9.
    1. McNaughton D, Knight W, Guerreiro R, Ryan N, Lowe J, Poulter M, et al.Duplication of amyloid precursor protein (APP), but not prion protein (PRNP) gene is a significant cause of early onset dementia in a large UK series. Neurobiol Aging 2012; 33: 426 e13.
    1. Miller JA, Woltjer RL, Goodenbour JM, Horvath S, Geschwind DH.. Genes and pathways underlying regional and cell type changes in Alzheimer's disease. Genome Med 2013; 5: 48.
    1. Miron J, Picard C, Nilsson N, Frappier J, Dea D, Theroux L, et al.CDK5RAP2 gene and tau pathophysiology in late-onset sporadic Alzheimer's disease. Alzheimers Dement 2018; 14: 787–96.
    1. Miyashita A, Koike A, Jun G, Wang L-S, Takahashi S, Matsubara E, et al.SORL1 is genetically associated with late-onset Alzheimer's disease in Japanese, Koreans and Caucasians. PLoS One 2013; 8: e58618.
    1. Naj AC, Jun G, Beecham GW, Wang LS, Vardarajan BN, Buros J, et al.Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat Genet 2011; 43: 436–41.
    1. Nasiri H, Forouzandeh M, Rasaee MJ, Rahbarizadeh F.. Modified salting-out method: high-yield, high-quality genomic DNA extraction from whole blood using laundry detergent. J Clin Lab Anal 2005; 19: 229–32.
    1. Patterson N, Price AL, Reich D.. Population structure and eigenanalysis. PLoS Genet 2006; 2: e190.
    1. Pruim RJ, Welch RP, Sanna S, Teslovich TM, Chines PS, Gliedt TP, et al.LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 2010; 26: 2336–7.
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, et al.PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 2007; 81: 559–75.
    1. Qi T, Wu Y, Zeng J, Zhang F, Xue A, Jiang L, et al.Identifying gene targets for brain-related traits using transcriptomic and methylomic data from blood. Nat Commun 2018; 9: 2282.
    1. Ramasamy A, Trabzuni D, Gibbs JR, Dillman A, Hernandez DG, Arepalli S, et al.Resolving the polymorphism-in-probe problem is critical for correct interpretation of expression QTL studies. Nucleic Acids Res 2013; 41: e88.
    1. Ramasamy A, Trabzuni D, Guelfi S, Varghese V, Smith C, Walker R, et al.Genetic variability in the regulation of gene expression in ten regions of the human brain. Nat Neurosci 2014; 17: 1418–28.
    1. Reitz C, Jun G, Naj A, Rajbhandary R, Vardarajan BN, Wang L-S, et al.Variants in the ATP-binding cassette transporter (ABCA7), apolipoprotein E ϵ4, and the risk of late-onset Alzheimer disease in African Americans. Jama 2013; 309: 1483–92.
    1. Sanchez-Mut JV, Heyn H, Silva BA, Dixsaut L, Garcia-Esparcia P, Vidal E, et al.PM20D1 is a quantitative trait locus associated with Alzheimer's disease. Nat Med 2018; 24: 598–603.
    1. Sanger F, Nicklen S, Coulson AR.. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci USA 1977; 74: 5463–7.
    1. Selkoe DJ, Hardy J.. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med 2016; 8: 595–608.
    1. Seshadri S, Fitzpatrick AL, Ikram MA, DeStefano AL, Gudnason V, Boada M, et al.Genome-wide analysis of genetic loci associated with Alzheimer disease. Jama 2010; 303: 1832–40.
    1. Sleigh JN, Barreiro-Iglesias A, Oliver PL, Biba A, Becker T, Davies KE, et al.Chondrolectin affects cell survival and neuronal outgrowth in in vitro and in vivo models of spinal muscular atrophy. Hum Mol Genet 2014; 23: 855–69.
    1. Stocker H, Mollers T, Perna L, Brenner H.. The genetic risk of Alzheimer's disease beyond APOE epsilon4: systematic review of Alzheimer's genetic risk scores. Transl Psychiatry 2018; 8: 166.
    1. Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al.STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 2015; 43(Database issue): D447–52.
    1. Tan L, Yu JT, Zhang W, Wu ZC, Zhang Q, Liu QY, et al.Association of GWAS-linked loci with late-onset Alzheimer's disease in a northern Han Chinese population. Alzheimers Dement 2013; 9: 546–53.
    1. Tao QQ, Liu ZJ, Sun YM, Li HL, Yang P, Liu DS, et al.Decreased gene expression of CD2AP in Chinese patients with sporadic Alzheimer's disease. Neurobiol Aging 2017; 56: 212 e5–e10.
    1. Trabzuni D, Ryten M, Walker R, Smith C, Imran S, Ramasamy A, et al.Quality control parameters on a large dataset of regionally dissected human control brains for whole genome expression studies. J Neurochem 2011; 119: 275–82.
    1. Wang H-Z, Bi R, Hu Q-X, Xiang Q, Zhang C, Zhang D-F, et al.Validating GWAS-identified risk loci for Alzheimer's disease in Han Chinese populations. Mol Neurobiol 2016; 53: 379–90.
    1. Wang Y, Lu D, Chung YJ, Xu S.. Genetic structure, divergence and admixture of Han Chinese, Japanese and Korean populations. Hereditas 2018; 155: 19.
    1. Westra HJ, Peters MJ, Esko T, Yaghootkar H, Schurmann C, Kettunen J, et al.Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet 2013; 45: 1238–43.
    1. Willer CJ, Li Y, Abecasis GR.. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 2010; 26: 2190–1.
    1. Wiseman FK, Al-Janabi T, Hardy J, Karmiloff-Smith A, Nizetic D, Tybulewicz VL, et al.A genetic cause of Alzheimer disease: mechanistic insights from Down syndrome. Nat Rev Neurosci 2015; 16: 564–74.
    1. Zelensky AN, Gready JE.. The C-type lectin-like domain superfamily. FEBS J 2005; 272: 6179–217.
    1. Zheng J, Erzurumluoglu AM, Elsworth BL, Kemp JP, Howe L, Haycock PC, et al.LD Hub: a centralized database and web interface to perform LD score regression that maximizes the potential of summary level GWAS data for SNP heritability and genetic correlation analysis. Bioinformatics 2017; 33: 272–9.
    1. Zhou X, Chen Y, Mok KY, Zhao Q, Chen K, Chen Y, et al.Identification of genetic risk factors in the Chinese population implicates a role of immune system in Alzheimer's disease pathogenesis. Proc Natl Acad Sci USA 2018; 115: 1697–706.
    1. Zhu Z, Zhang F, Hu H, Bakshi A, Robinson MR, Powell JE, et al.Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat Genet 2016; 48: 481–7.

Source: PubMed

3
Subscribe