Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Aug 1.
Published in final edited form as: Neurosci Lett. 2008 May 27;440(2):190–192. doi: 10.1016/j.neulet.2008.05.082

No association of SORL1 SNPs with Alzheimer’s disease

Ryan L Minster a, Steven T DeKosky b, M Ilyas Kamboh a,*
PMCID: PMC2519047  NIHMSID: NIHMS58022  PMID: 18562096

Abstract

SORL1 is an element of the amyloid precursor protein processing pathway and is therefore a good candidate for affecting Alzheimer’s disease (AD) risk. Indeed, there have been reports of associations between variation in SORL1 and AD risk. We examined six statistically significant single-nucleotide polymorphisms from the initial observation in a large Caucasian American case–controls cohort (1000 late-onset AD [LOAD] cases and 1000 older controls). Analysis of allele, genotype and haplotype frequencies revealed no association with LOAD risk in our cohort.

Keywords: Alzheimer’s disease, SORL1, genetics


It is estimated that up to 79% of the risk for late-onset Alzheimer’s disease (LOAD) is attributable to genetics [4]. Thus far, only variation in APOE has been definitively associated with LOAD [2], but only 20%–29% of risk is attributable to this variation [20, 22]. Association between variation in an excellent biological candidate gene, sortilin-related receptor 1 (SORL1), and the risk of LOAD has been reported [18]. We genotyped and analyzed six of the single nucleotide polymorphisms (SNPs), listed in Table 1, that were reported to be associated with LOAD in a multiple case–control cohort or family-based samples [18] to verify this report.

Table 1.

Examined SORL1 markers, their genotype and allele frequencies, and statistical significance.

RefSNP Variation Gene Element AD Status n Genotype Frequencies p Allele Frequencies p
TT TC CC T C


rs661057 c.285+5629T>C intron 1 cases 1000 0.336 0.490 0.174 0.980 0.581 0.419 0.852
controls 1001 0.339 0.491 0.171 0.584 0.416
CC CT TT C T


rs668387 c.939+163C>T intron 6 cases 1005 0.353 0.473 0.174 0.177 0.590 0.410 0.526
controls 1004 0.323 0.514 0.163 0.580 0.420
GG GA AA G A


rs689021 c.939+3362G>A intron 6 cases 1000 0.345 0.174 0.178 0.370 0.584 0.417 0.786
controls 1003 0.326 0.163 0.167 0.579 0.421
GG GA AA G A


rs641120 c.940-2747G>A intron 6 cases 1004 0.348 0.480 0.172 0.462 0.588 0.412 0.518
controls 999 0.324 0.507 0.169 0.578 0.422
TT TG GG T G


rs2070045 c.3561T>G exon 26 cases 994 0.607 0.344 0.049 0.711 0.779 0.221 0.823
controls 1001 0.608 0.335 0.057 0.776 0.224
TT TA AA T A


rs3824968 c.4752T>A exon 34 cases 1001 0.498 0.480 0.084 0.674 0.707 0.293 0.420
controls 1007 0.485 0.507 0.094 0.695 0.305

Case subjects were Caucasian Americans with LOAD (n = 1009; mean age-at-onset [AAO] 72.8 ± 6.2 [SD] years; 67.7% female; 7.3% autopsy-confirmed) recruited by the University of Pittsburgh Alzheimer’s Disease Research Center. All cases were evaluated clinically and met criteria for probable or possible AD [11] or by autopsy and met neuropathological criteria for definite AD [13, 14]. Controls were Caucasian Americans of age 60 or above with no psychiatric or neurological disorders (n = 1009; mean age-at-baseline 74.1 ± 6.2 [SD] years; 59.9% female; 1.3% autopsy-confirmed). All experiments on human subjects were conducted in accordance with the Declaration of Helsinki, and all procedures were carried out with the adequate understanding and written consent of the subjects. The genetic study was approved by the University of Pittsburgh Institutional Review Board.

Genotypes for the six SORL1 SNPs were ascertained from genomic DNA using TaqMan SNP genotyping assays (Applied Biosystems, Foster City, California). Case and control samples were present on each 384-well plate used in genotyping. To estimate genotyping error rates, 10% of the samples were selected at random and repeated. Genotypes for APOE were determined either as previously described [5] or using TaqMan SNP genotyping assays.

Allele and genotype frequencies were calculated by the direct allele-counting method. Goodness of fit to Hardy–Weinberg equilibrium was tested using the χ2 test. Differences between genotype and allele frequencies in cases and controls were tested with the χ2 test. Differences between cases and controls stratified by APOE*4 carrier status were also tested with the χ2 test. Haplotype frequencies were estimated in cases and controls, and the global difference in frequencies was tested. Linkage disequilibrium (LD) between the SNPs was estimated by calculating D′ and r2 between each pair. Haplotype frequencies were then again compared after eliminating highly-correlated SNPs (those with r2 > 0.8). These statistics were calculated using R 2.2.0 with the genetics and haplo.stats packages attached [16, 21, 24]. Power to detect associations was determined with PS 2.1.30 [3].

The allele and genotype frequencies for the SORL1 SNPs are shown in Table 1. Neither cases nor controls had genotype frequencies which differed significantly from those expected under Hardy–Weinberg equilibrium. The differences between cases and controls do not rise to statistical significance (range: p = 0.177–0.980).

Among, non–APOE*4 carriers, the differences were not statistically significant (range: p = 0.357–0.922). Among APOE*4 carriers, rs661057 was associated with AD risk (genotype frequency differences, p = 0.022; alleles, p = 0.008). None of the other SNPs were observed to be associated (range: p = 0.109–0.765).

Haplotype frequencies calculated for all six SNPs did not differ with statistical significance (p = 0.627) (data not shown). LD between the SNPs is shown in Table 2. Because the three SNPs in intron 6 (rs668387, rs689021 and rs641120) were highly correlated (r2 = 0.988–0.996), haplotype frequencies were recalculated between just four of the SNPs (rs661057, rs668387, rs2070045 and rs3824968). The frequency differences were not significant (p = 0.858).

Table 2.

Linkage disequilibrium between examined SORL1 SNPs

D′
rs661057 0.6363 0.6373 0.6437 0.0989 0.0437
0.4014 rs668387 0.9956 0.9900 0.1414 0.0680
r2 0.4044 0.9780 rs689021 0.9876 0.1470 0.0642
0.4141 0.9720 0.9704 rs641120 0.1487 0.0640
0.0039 0.0081 0.0086 0.0088 rs2070045 0.8643
0.0011 0.0014 0.0013 0.0013 0.5019 rs3824968

Given the minor allele frequencies at α = 0.05, we had 80% power to detect a risk odds-ratio of 1.23 with statistical significance for rs2070045 which had the lowest minor allele frequencies and 1.20 for rs641120 which had the highest.

Sortilin-related receptor 1 (also known as sorLA and LR11) is an excellent biological candidate gene due to its role in amyloid precursor protein (APP) processing and evidence that variation in SORL1 levels are associated with both APP [1, 15] and LOAD [19]. Association between variation in SORL1 and LOAD has been reported in Northern Europeans, Caucasian Americans, Caribbean Hispanics and Israeli Arabs [18]; Caribbean Hispanics and Caucasian Americans [6]; Han Chinese [23]; and Caucasian Americans with Down syndrome [7]. However, the same large study that reported association in multiple groups [18] found no association in Caucasian American or African American families. Two groups examined SORL1 SNPs in publicly available data from the same genomewide association studies of AD [17] and found marginal associations with some variants in specific regions of SORL1. Meng et al. [12] observed associated SNPs in the interval from exon 7 to exon 18;Webster et al. [25], from intron 25 to intron 39 (SORL1 comprises 48 exons). However, neither replicated any of Rogaeva et al.’s [18] significant SNPs in particular. Another high-density genomewide association study of AD failed to find any association between disease risk and any of 41 SNPs included from the SORL1 region [8].

Liu et al. [10] also failed to find evidence of association between SORL1 and LOAD in a large Dutch pedigree, although they did observe evidence of linkage nearby at 11q25 that they believe to be associated not with SORL1 but with OPCML and HNT. Li et al. [9] examined several SNPs and haplotypes in a population of size and demographics similar to ours, and observed only marginal association with AD risk for a single SNP (rs2070045) which they dismissed because it is not statistically significant after correcting for multiple testing.

We did not find evidence of association in our Caucasian American cohort. It is possible that the effect of SORL1 variation on AD risk is specific to particular ethnic groups or that the effect is not large enough to be detected reliably by a cohort of our size. Further examinations into this gene and the region surrounding it are necessary to determine the role of SORL1 if any in modulating LOAD risk.

Acknowledgements

This study was supported by National Institute on Aging grants AG13672 and AG05133. We thank Jessica Figgins, Oussama Khalifa and YueeWang for their technical contributions.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Andersen OM, Reiche J, Schmidt V, Gotthardt M, Spoelgen R, Behlke J, von Arnim CAF, Breiderhoff T, Jansen P, Wu X, Bales KR, Cappai R, Masters CL, Gliemann J, Mufson EJ, Hyman BT, Paul SM, Nykjær A, Willnow TE. Neuronal sorting protein-related receptor sorLA/LR11 regulates processing of the amyloid precursor protein. Proc. Natl Acad. Sci. U.S.A. 2005;102:13461–13466. doi: 10.1073/pnas.0503689102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Roses AD, Haines JL, Pericak-Vance MA. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–923. doi: 10.1126/science.8346443. [DOI] [PubMed] [Google Scholar]
  • 3.Dupont WD, Plummer WD., Jr Power and sample size calculations for studies involving linear regression. Control. Clin. Trials. 1998;19:589–601. doi: 10.1016/s0197-2456(98)00037-3. [DOI] [PubMed] [Google Scholar]
  • 4.Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, Fiske A, Pedersen NL. Role of genes and environments for explaining Alzheimer disease. Arch. Gen. Psychiatry. 2006;63:168–174. doi: 10.1001/archpsyc.63.2.168. [DOI] [PubMed] [Google Scholar]
  • 5.Kamboh MI, Aston CE, Hamman RF. The relationship of APOE polymorphism and cholesterol levels in normoglycemic and diabetic subjects in a biethnic population from the San Luis Valley, Colorado. Atherosclerosis. 1995;112:145–159. doi: 10.1016/0021-9150(94)05409-c. [DOI] [PubMed] [Google Scholar]
  • 6.Lee JH, Cheng R, Schupf N, Manly J, Lantigua R, Stern Y, Rogaeva E, Wakutani Y, Farrer L, St George-Hyslop P, Mayeux R. The association between genetic variants in SORL1 and Alzheimer disease in an urban, multiethnic, community-based cohort. Arch. Neurol. 2007;64:501–506. doi: 10.1001/archneur.64.4.501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lee JH, Chulikavit M, Pang D, Zigman WB, Silverman W, Schupf N. Association between genetics variants in sortilin-related receptor 1 (SORL1) and Alzheimer’s disease in adults with Down syndrome. Neurosci. Lett. 2007;425:105–109. doi: 10.1016/j.neulet.2007.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Li H, Wetten S, Li L, St Jean PL, Upmanyu R, Surh L, Hosford D, Barnes MR, Briley JD, Borrie M, Coletta N, Delisle R, Dhalla D, Ehm MG, Feldman HH, Fornazzari L, Gauthier S, Goodgame N, Guzman D, Hammond S, Hollingworth P, Hsiung G-Y, Johnson J, Kelly DD, Keren R, Kertesz A, King KS, Lovestone S, Loy-English I, Matthews PM, Owen MJ, Plumpton M, Pryse-Phillips W, Prinjha RK, Richardson JC, Saunders A, Slater AJ, St George-Hyslop PH, Stinnett SW, Swartz JE, Taylor RL, Wherrett J, Williams J, Yarnall DP, Gibson RA, Irizarry MC, Middleton LT, Roses AD. Candidate single-nucleotide polymorphisms from a genomewide association study on Alzheimer disease. Arch. Neurol. 2008;65:45–53. doi: 10.1001/archneurol.2007.3. [DOI] [PubMed] [Google Scholar]
  • 9.Li Y, Rowland C, Catanese J, Morris J, Lovestone S, O’Donovan MC, Goate A, Owen M, Williams J, Grupe A. SORL1 variants and risk of late-onset Alzheimer’s disease. Neurobiol. Dis. 2008;209:293–296. doi: 10.1016/j.nbd.2007.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Liu F, Arias-Vásquez A, Sleegers K, Aulchenko YS, Kayser M, Sanchez-Juan P, Feng B-J, Bertoli-Avella AM, van Swieten J, Axenovich TI, Heutink P, van Broeckhoven C, Oostra BA, van Duijn CM. A genomewide screen for late-onset Alzheimer disease in a genetically isolated Dutch population. Am. J. Hum. Genet. 2007;81:17–31. doi: 10.1086/518720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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;34:939–944. doi: 10.1212/wnl.34.7.939. [DOI] [PubMed] [Google Scholar]
  • 12.Meng Y, Lee JH, Cheng R, St George-Hyslop P, Mayeux R, Farrer LA. Association between SORL1 and Alzheimer’s disease in a genome-wide study. Neuroreport. 2007;18:1761–1764. doi: 10.1097/WNR.0b013e3282f13e7a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, Brownlee LM, Vogel FS, Hughes JP, van Belle G, Berg L. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD), Part II, Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology. 1991;41:479–486. doi: 10.1212/wnl.41.4.479. [DOI] [PubMed] [Google Scholar]
  • 14.National Institute on Aging. Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease, Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease. Neurobiol. Aging. 1997;18:S1–S2. [PubMed] [Google Scholar]
  • 15.Offe K, Dodson SE, Shoemaker JT, Fritz JJ, Gearing M, Levey AI, Lah JJ. The lipoprotein receptor LR11 regulates amyloid β production and amyloid precursor protein traffic in endosomal compartments. J. Neurosci. 2006;26:1596–1603. doi: 10.1523/JNEUROSCI.4946-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.R Development Core Team. R: A language and environment for statistical computing, version 2.2.0. Vienna, Austria: R Foundation for Statistical Computing; 2005. www.r-project.org. [Google Scholar]
  • 17.Reiman EM, Webster JA, Myers AJ, Hardy J, Dunckley T, Zismann VL, Joshipura KD, Pearson JV, Hu-Lance D, Huentelman MJ, Craig DW, Coon KD, Liang WS, Herbert RH, Beach T, Rohrer KC, Zhao AS, Leung D, Bryden L, Marlowe L, Kaleem M, Mastroeni D, Grover A, Heward CB, Ravid R, Rogers J, Hutton ML, Melquist S, Petersen RC, Alexander GE, Caselli RJ, Kukull W, Papassotiropoulos A, Stephan DA. GAB2 alleles modify Alzheimer’s risk in APOE ε4 carriers. Neuron. 2007;54:713–720. doi: 10.1016/j.neuron.2007.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T, Zou F, Katayama T, Baldwin CT, Cheng R, Hasegawa H, Chen F, Shibata N, Lunetta KL, Pardossi-Piquard R, Bohm C, Wakutani Y, Cupples LA, Cuenco KT, Green RC, Pinessi L, Rainero I, Sorbi S, Bruni A, Duara R, Friedland RP, Inzelberg R, Hampe W, Bujo H, Song Y-Q, Andersen OM, Willnow TE, Graff-Radford N, Petersen RC, Dickson D, Der SD, Fraser PE, Schmitt-Ulms G, Younkin S, Mayeux R, Farrer LA, St George-Hyslop P. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat. Genet. 2007;39:168–177. doi: 10.1038/ng1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Scherzer CR, Offe K, Gearing M, Rees HD, Fang G, Heilman CJ, Schaller C, Bujo H, Levey AI, Lah JJ. Loss of apolipoprotein E receptor LR11 in Alzheimer disease. Arch. Neurol. 2004;61:1200–1205. doi: 10.1001/archneur.61.8.1200. [DOI] [PubMed] [Google Scholar]
  • 20.Seshadri S, Drachman DA, Lippa CF. Apolipoprotein E ε4 allele and the lifetime risk of Alzheimer’s disease. What physicians know, and what they should know. Arch. Neurol. 1995;52:1074–1079. doi: 10.1001/archneur.1995.00540350068018. [DOI] [PubMed] [Google Scholar]
  • 21.Sinnwell JP, Schaid DJ. haplo.stats: Statistical Analysis of Haplotypes with Traits and Covariates when Linkage Phase is Ambiguous. R package version 1.2.2. 2005 [Google Scholar]
  • 22.Slooter AJC, Cruts M, Kalmijn S, Hofman A, Breteler MMB, Van Broeckhoven C, van Duijn CM. Risk estimates of dementia by apolipoprotein E genotypes from a population-based study: the Rotterdam study. Arch. Neurol. 1998;55:964–968. doi: 10.1001/archneur.55.7.964. [DOI] [PubMed] [Google Scholar]
  • 23.Tan EK, Lee J, Chen CP, Teo YY, Zhao Y, Lee WL. SORL1 haplotypes modulate risk of Alzheimer’s disease in Chinese. Neurobiol. Aging. doi: 10.1016/j.neurobiolaging.2007.10.013. in press. [DOI] [PubMed] [Google Scholar]
  • 24.Warnes G, Leisch F. genetics: Population Genetics. R package version 1.2.0. 2005 [Google Scholar]
  • 25.Webster JA, Myers AJ, Pearson JV, Craig DW, Hu-Lince D, Coon KD, Zismann VL, Beach T, Leung D, Bryden L, Halperin RF, Marlowe L, Kaleem M, Huentelman MJ, Joshipura K, Walker D, Heward CB, Ravid R, Rogers J, Papassotiropoulos A, Hardy J, Reiman EM, Stephan DA. Sorl1 as an Alzheimer’s disease predisposition gene? Neurodegener. Dis. 2008;5:60–64. doi: 10.1159/000110789. [DOI] [PubMed] [Google Scholar]

RESOURCES