Use of genetic variation as biomarkers for Alzheimer's disease

Christiane Reitz, Richard Mayeux, Christiane Reitz, Richard Mayeux

Abstract

Late-onset Alzheimer's disease (LOAD) is the most common cause of late-onset dementia in western societies. Despite remarkable achievements in human genetics throughout the years, in particular technological advances in gene mapping and in statistical methods that relate genetic variants to disease, to date only a small proportion of the genetic contribution to LOAD can be explained leaving several remaining genetic risk factors to be identified. A possible explanation for the difficulty in gene identification is that LOAD is a multifactorial complex disorder with both genetic and environmental components. Multiple genes with small effects each ("quantitative trait loci"[QTLs]) are likely to contribute to the quantitative traits associated with the disease, such as memory performance, amyloid/tau pathology, or hippocampal atrophy. The motivation for identifying the genetics of LOAD is clear. Not only could it shed light on disease pathogenesis, but it may also provide potential targets for effective treatment, screening, and prevention. Here, we review the usefulness of genetic variation as diagnostic tools and biomarkers in LOAD and discuss the potentials and difficulties researchers face in designing appropriate studies for gene discovery.

Conflict of interest statement

Conflicts of Interest

The authors declare no conflicts of interest.

Figures

Figure 1
Figure 1
Pooled odds ratios (95% CI) of the 40 studies included in the meta-analysis by Farrer et al. relating APOE genotype with LOAD (ε4 allele versus ε3 allele). †No data provided; ‡Hardy-Weinberg-Equilibrium (HWE) deviation in controls (P ≤ 0.05). (In color in Annals online.)
Figure 2
Figure 2
Protein sorting as a key mechanism in Alzheimer’s cell biology. (A) Aβ (red bar) is liberated from its parent protein, APP (multicolor bar), in two enzymatic steps. In the first β-cleavage step, BACE (blue bar) splits full-length APP into an sAPPβ fragment (brown bar) and a C-terminal fragment (CTFβ, red/green bar). Then, in the γ-cleavage step, the γ-secretase (star) splits CTFβ into Aβ (red bar) and amyloid intracellular domain (AICD, green bar). β-cleavage is the committed step in APP processing and may be upregulated in LOAD. (B) Both APP (red bar) and BACE (blue bar) are type-I transmembrane proteins that are sorted through multiple membranous compartments of the cell. The sorting triangle that interconnects the trans-Golgi network (TGN), cell surface, and the endosome is critically important for APP and BACE sorting. As indicated, clathrin is the coat complex that regulates transport from the cell surface and the TGN to the endosome, whereas the retromer is the coat complex that regulates transport from the endosome back to the TGN. APP and BACE interact within the membranes of the endosomal system, initiating the amyloidogenic pathway. (Illustration adapted from Small and Gandy.75) (In color in Annals online.)
Figure 3
Figure 3
Role of SORL1 in transmembrane sorting of APP. The green arrows track re-entry of APP from the cell surface when SORL1 is present. The red arrows show that, when SORL1 is absent, more APP moves into domains, such as the late endosome/lysosome, where the black arrows show how it is subsequently cut by beta-secretase (BACE1) and gamma-secretase (PS1 γ-sec), generating the neurotoxic amyloid beta-peptide (Aβ). (Illustration adapted from Rogaeva et al.77) (In color in Annals online.)

References

    1. Ferri CP, Prince M, Brayne C, et al. Global prevalence of dementia: a Delphi consensus study. Lancet. 2005;366:2112–2117.
    1. Fratiglioni L, De Ronchi D, Aguero-Torres H. Worldwide prevalence and incidence of dementia. Drugs Aging. 1999;15:365–375.
    1. Brookmeyer R, Gray S, Kawas C. Projections of Alzheimer’s disease in the United States and the public health impact of delaying disease onset. Am J Public Health. 1998;88:1337–1342.
    1. McKhann G, Drachman D, Folstein M, et al. 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.
    1. Lopez OL, Swihart AA, Becker JT, et al. Reliability of NINCDS-ADRDA clinical criteria for the diagnosis of Alzheimer’s disease. Neurology. 1990;40:1517–1522.
    1. Kukull WA, Larson EB, Reifler BV, et al. Interrater reliability of Alzheimer’s disease diagnosis. Neurology. 1990;40:257–260.
    1. Schofield PW, Tang M, Marder K, et al. Consistency of clinical diagnosis in a community-based longitudinal study of dementia and Alzheimer’s disease. Neurology. 1995;45:2159–2164.
    1. Morris JC, McKeel DW, Jr, Fulling K, et al. Validation of clinical diagnostic criteria for Alzheimer’s disease. Ann Neurol. 1988;24:17–22.
    1. Burns A, Luthert P, Levy R, et al. Accuracy of clinical diagnosis of Alzheimer’s disease. BMJ (Clinical Research Ed) 1990;301:1026.
    1. Meyer JM, Breitner JC. Multiple threshold model for the onset of Alzheimer’s disease in the NAS-NRC twin panel. Am J Med Genet. 1998;81:92–97.
    1. Ertekin-Taner N, Allen M, Fadale D, et al. Genetic variants in a haplotype block spanning IDE are significantly associated with plasma Abeta42 levels and risk for Alzheimer disease. Hum Mutat. 2004;23:334–342.
    1. Ertekin-Taner N, Ronald J, Feuk L, et al. Elevated amyloid beta protein (Abeta42) and late onset Alzheimer’s disease are associated with single nucleotide polymorphisms in the urokinase-type plasminogen activator gene. Hum Mol Genet. 2005;14:447–460.
    1. Farris W, Mansourian S, Leissring MA, et al. Partial loss-of-function mutations in insulin-degrading enzyme that induce diabetes also impair degradation of amyloid beta-protein. Am J Pathol. 2004;164:1425–1434.
    1. Breitner JC, Wyse BW, Anthony JC, et al. APOE-epsilon4 count predicts age when prevalence of AD increases, then declines: the Cache County Study. Neurology. 1999;53:321–331.
    1. Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–923.
    1. Gomez-Isla T, West HL, Rebeck GW, et al. Clinical and pathological correlates of apolipoprotein E epsilon 4 in Alzheimer’s disease. Ann Neurol. 1996;39:62–70.
    1. Holmes C, Levy R, McLoughlin DM, et al. Apolipoprotein E: non-cognitive symptoms and cognitive decline in late onset Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1996;61:580–583.
    1. Hyman BT, Gomez-Isla T, Rebeck GW, et al. Epidemiological, clinical, and neuropathological study of apolipoprotein E genotype in Alzheimer’s disease. Ann N Y Acad Sci. 1996;802:1–5.
    1. Kurz A, Altland K, Lautenschlager N, et al. Apolipoprotein E type 4 allele and Alzheimer’s disease: effect on age at onset and relative risk in different age groups. J Neurol. 1996;243:452–456.
    1. Murman DL, Foster NL, Kilgore SP, et al. Apolipoprotein E and Alzheimer’s disease: strength of association is related to age at onset. Dementia. 1996;7:251–255.
    1. Poirier J, Davignon J, Bouthillier D, et al. Apolipoprotein E polymorphism and Alzheimer’s disease. Lancet. 1993;342:697–699.
    1. Roses AD. Alzheimer’s disease: the genetics of risk. Hosp Pract (Minneap) 1997;32:51–55. 58–63, 67–69.
    1. Tang MX, Maestre G, Tsai WY, et al. Relative risk of Alzheimer disease and age-at-onset distributions, based on APOE genotypes among elderly African Americans, Caucasians, and Hispanics in New York City. Am J Hum Genet. 1996;58:574–584.
    1. Slooter AJ, Cruts M, Kalmijn S, et al. Risk estimates of dementia by apolipoprotein E genotypes from a population-based incidence study: the Rotterdam Study. Arch Neurol. 1998;55:964–968.
    1. Ashford JW, Mortimer JA. Non-familial Alzheimer’s disease is mainly due to genetic factors. J Alzheimers Dis. 2002;4:169–177.
    1. Strittmatter WJ, Weisgraber KH, Huang DY, et al. Binding of human apolipoprotein E to synthetic amyloid beta peptide: isoform-specific effects and implications for late-onset Alzheimer disease. Proc Natl Acad Sci USA. 1993;90:8098–8102.
    1. Schmechel DE, Saunders AM, Strittmatter WJ, et al. Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc Natl Acad Sci USA. 1993;90:9649–9653.
    1. Rebeck GW, Reiter JS, Strickland DK, Hyman BT. Apolipoprotein E in sporadic Alzheimer’s disease: allelic variation and receptor interactions. Neuron. 1993;11:575–580.
    1. Bales KR, Verina T, Cummins DJ, et al. Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 1999;96:15233–15238.
    1. Bales KR, Verina T, Dodel RC, et al. Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nat Genet. 1997;17:263–264.
    1. Kindy MS, Rader DJ. Reduction in amyloid A amyloid formation in apolipoprotein-E-deficient mice. Am J Pathol. 1998;152:1387–1395.
    1. Ma J, Yee A, Brewer HB, Jr, et al. Amyloid-associated proteins alpha 1-antichymotrypsin and apolipoprotein E promote assembly of Alzheimer beta-protein into filaments. Nature. 1994;372:92–94.
    1. Sanan DA, Weisgraber KH, Russell SJ, et al. Apolipoprotein E associates with beta amyloid peptide of Alzheimer’s disease to form novel monofibrils. Isoform apoE4 associates more efficiently than apoE3. J Clin Investig. 1994;94:860–869.
    1. Holtzman DM, Bales KR, Tenkova T, et al. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2000;97:2892–2897.
    1. Holtzman DM, Bales KR, Wu S, et al. Expression of human apolipoprotein E reduces amyloid-beta deposition in a mouse model of Alzheimer’s disease. J Clin Investig. 1999;103:R15–R21.
    1. Sparks DL, Scheff SW, Hunsaker JC, 3rd, et al. Induction of Alzheimer-like beta-amyloid immunoreactivity in the brains of rabbits with dietary cholesterol. Exp Neurol. 1994;126:88–94.
    1. Refolo LM, Malester B, LaFrancois J, et al. Hypercholesterolemia accelerates the Alzheimer’s amyloid pathology in a transgenic mouse model. Neurobiol Disease. 2000;7:321–331.
    1. Bodovitz S, Klein WL. Cholesterol modulates alpha-secretase cleavage of amyloid precursor protein. J Biol Chem. 1996;271:4436–4440.
    1. Howland DS, Trusko SP, Savage MJ, et al. Modulation of secreted beta-amyloid precursor protein and amyloid beta-peptide in brain by cholesterol. J Biol Chem. 1998;273:16576–16582.
    1. Farrer LA, Cupples LA, Haines JL, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278:1349–1356.
    1. Gozal D, Capdevila OS, Kheirandish-Gozal L, Crabtree VM. APOE epsilon 4 allele, cognitive dysfunction, and obstructive sleep apnea in children. Neurology. 2007;69:243–249.
    1. Caselli RJ, Graff-Radford NR, Reiman EM, et al. Preclinical memory decline in cognitively normal apolipoprotein E-epsilon4 homozygotes. Neurology. 1999;53:201–207.
    1. Flory JD, Manuck SB, Ferrell RE, et al. Memory performance and the apolipoprotein E polymorphism in a community sample of middle-aged adults. Am J Med Genet. 2000;96:707–711.
    1. Liu F, Pardo LM, Schuur M, et al. The apolipoprotein E gene and its age-specific effects on cognitive function. Neurobiol Aging. 2008 Epub ahead of print.
    1. Schultz MR, Lyons MJ, Franz CE, et al. Apolipoprotein E genotype and memory in the sixth decade of life. Neurology. 2008;70(19 Pt 2):1771–1777.
    1. Wehling E, Lundervold AJ, Standnes B, et al. APOE status and its association to learning and memory performance in middle aged and older Norwegians seeking assessment for memory deficits. Behav Brain Funct. 2007;3:57.
    1. Mak YT, Chiu H, Woo J, et al. Apolipoprotein E genotype and Alzheimer’s disease in Hong Kong elderly Chinese. Neurology. 1996;46:146–149.
    1. Hallman DM, Boerwinkle E, Saha N, et al. The apolipoprotein E polymorphism: a comparison of allele frequencies and effects in nine populations. Am J Hum Genet. 1991;49:338–349.
    1. do Couto FS, de Mendonca A, Garcia C, et al. Age of onset in patients with Alzheimer’s disease with different apoE genotypes. J Neurol Neurosurg Psychiatry. 1998;64:817.
    1. Dal Forno G, Rasmusson DX, Brandt J, et al. Apolipoprotein E genotype and rate of decline in probable Alzheimer’s disease. Arch Neurol. 1996;53:345–350.
    1. Welsh-Bohmer KA, Ostbye T, Sanders L, et al. Neuropsychological performance in advanced age: influences of demographic factors and apolipoprotein E: findings from the Cache County Memory Study. Clin Neuropsychol. 2008:1–23.
    1. Salo A, Ylikoski R, Verkkoniemi A, et al. Does apolipoprotein E influence learning and memory in the nondemented oldest old? Int Psychogeriatr. 2001;13:451–459.
    1. Murphy GM, Jr, Taylor J, Kraemer HC, et al. No association between apolipoprotein E epsilon 4 allele and rate of decline in Alzheimer’s disease. Am J Psychiatry. 1997;154:603–608.
    1. Cosentino S, Scarmeas N, Helzner E, et al. APOE epsilon 4 allele predicts faster cognitive decline in mild Alzheimer disease. Neurology. 2008;70(19 Pt 2):1842–1849.
    1. Hirono N, Hashimoto M, Yasuda M, et al. Accelerated memory decline in Alzheimer’s disease with apolipoprotein epsilon4 allele. J Neuropsychiatry Clin Neurosci. 2003;15:354–358.
    1. Mayeux R, Small SA, Tang M, et al. Memory performance in healthy elderly without Alzheimer’s disease: effects of time and apolipoprotein-E. Neurobiol Aging. 2001;22:683–689.
    1. Wilson RS, Bienias JL, Berry-Kravis E, et al. The apolipoprotein E epsilon 2 allele and decline in episodic memory. J Neurol Neurosurg Psychiatry. 2002;73:672–677.
    1. Lehmann DJ, Refsum H, Nurk E, et al. Apolipoprotein E epsilon4 and impaired episodic memory in community-dwelling elderly people: a marked sex difference. The Hordaland Health Study. J Neurol Neurosurg Psychiatry. 2006;77:902–908.
    1. Bondi MW, Salmon DP, Monsch AU, et al. Episodic memory changes are associated with the APOE-epsilon 4 allele in nondemented older adults. Neurology. 1995;45:2203–2206.
    1. Dik MG, Jonker C, Comijs HC, et al. Memory complaints and APOE-epsilon4 accelerate cognitive decline in cognitively normal elderly. Neurology. 2001;57:2217–2222.
    1. Hsiung GY, Sadovnick AD, Feldman H. Apolipoprotein E epsilon4 genotype as a risk factor for cognitive decline and dementia: data from the Canadian Study of Health and Aging. CMAJ. 2004;171:863–867.
    1. Petersen RC, Smith GE, Ivnik RJ, et al. Apolipoprotein E status as a predictor of the development of Alzheimer’s disease in memory-impaired individuals. JAMA. 1995;273:1274–1278.
    1. Reynolds CA, Prince JA, Feuk L, et al. Longitudinal memory performance during normal aging: twin association models of APOE and other Alzheimer candidate genes. Behav Genet. 2006;36:185–194.
    1. Saunders AM, Hulette O, Welsh-Bohmer KA, et al. Specificity, sensitivity, and predictive value of apolipoprotein-E genotyping for sporadic Alzheimer’s disease. Lancet. 1996;348:90–93.
    1. Mayeux R, Saunders AM, Shea S, et al. Utility of the apolipoprotein E genotype in the diagnosis of Alzheimer’s disease. Alzheimer’s disease centers consortium on apolipoprotein E and Alzheimer’s disease. N Engl J Med. 1998;338:506–511.
    1. Roses AD. Apolipoprotein E genotyping in the differential diagnosis, not prediction, of Alzheimer’s disease. Ann Neurol. 1995;38:6–14.
    1. Slooter AJ, Breteler MB, Ott A, et al. APOE genotyping in differential diagnosis of Alzheimer’s disease. Lancet. 1996;348:334.
    1. Kakulas BA, Wilton SD, Fabian VA, Jones TM. Apolipoprotein-E genotyping in diagnosis of Alzheimer’s disease. Lancet. 1996;348:483.
    1. Edbauer D, Winkler E, Regula JT, et al. Reconstitution of gamma-secretase activity. Nat Cell Biol. 2003;5:486–488.
    1. Cobbold C, Monaco AP, Sivaprasadarao A, Ponnambalam S. Aberrant trafficking of transmembrane proteins in human disease. Trends Cell Biol. 2003;13:639–647.
    1. Harter C, Reinhard C. The secretory pathway from history to the state of the art. Sub-Cell Biochem. 2000;34:1–38.
    1. Le Borgne R, Hoflack B. Protein transport from the secretory to the endocytic pathway in mammalian cells. Biochim Biophys Acta. 1998;1404:195–209.
    1. Chen WJ, Goldstein JL, Brown MS. NPXY, a sequence often found in cytoplasmic tails, is required for coated pit-mediated internalization of the low density lipoprotein receptor. J Biol Chem. 1990;265:3116–3123.
    1. He X, Li F, Chang WP, Tang J. GGA proteins mediate the recycling pathway of memapsin 2 (BACE) J Biol Chem. 2005;280:11696–11703.
    1. Small SA, Gandy S. Sorting through the cell biology of Alzheimer’s disease: intracellular pathways to pathogenesis. Neuron. 2006;52:15–31.
    1. Traub LM. Common principles in clathrin-mediated sorting at the Golgi and the plasma membrane. Biochim Biophys Acta. 2005;1744:415–437.
    1. Rogaeva E, Meng Y, Lee JH, et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet. 2007;39:168–177.
    1. Bettens K, Brouwers N, Engelborghs S, et al. SORL1 is genetically associated with increased risk for late-onset Alzheimer disease in the Belgian population. Hum Mutat. 2008;29:769–770.
    1. Lee JH, Cheng R, Honig LS, et al. Association between genetic variants in SORL1 and autopsy-confirmed Alzheimer disease. Neurology. 2008;70:887–889.
    1. Lee JH, Cheng R, Schupf N, et al. The association between genetic variants in SORL1 and Alzheimer disease in an urban, multiethnic, community-based cohort. Arch Neurol. 2007;64:501–506.
    1. Meng Y, Lee JH, Cheng R, et al. Association between SORL1 and Alzheimer’s disease in a genome-wide study. Neuroreport. 2007;18:1761–1764.
    1. Seshadri S, DeStefano AL, Au R, et al. Genetic correlates of brain aging on MRI and cognitive test measures: a genome-wide association and linkage analysis in the Framingham Study. BMC Med Genet. 2007;8(Suppl 1):S15.
    1. Tan EK, Lee J, Chen CP, et al. SORL1 haplotypes modulate risk of Alzheimer’s disease in Chinese. Neurobiol Aging. 2009;30:1048–1051.
    1. Li H, Wetten S, Li L, et al. Candidate single-nucleotide polymorphisms from a genomewide association study of Alzheimer disease. Arch Neurol. 2008;65:45–53.
    1. Li Y, Rowland C, Catanese J, et al. SORL1 variants and risk of late-onset Alzheimer’s disease. Neurobiol Dis. 2008;29:293–296.
    1. Webster JA, Myers AJ, Pearson JV, et al. Sorl1 as an Alzheimer’s disease predisposition gene? Neurodegener Dis. 2008;5:60–64.
    1. Shibata N, Ohnuma T, Baba H, et al. Genetic association between SORL1 polymorphisms and Alzheimer’s disease in a Japanese population. Dement Geriatr Cogn Disord. 2008;26:161–164.
    1. Lee JH, Shibata N, Cheng R, Mayeux R. Possible association between SORL1 and Alzheimer disease? Reanalyzing the data of Shibata et al. Dement Geriatr Cogn Disord. 2008;26:482.
    1. Bertram L, McQueen MB, Mullin K, et al. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39:17–23.
    1. Grupe A, Abraham R, Li Y, et al. Evidence for novel susceptibility genes for late-onset Alzheimer’s disease from a genome-wide association study of putative functional variants. Hum Mol Genet. 2007;16:865–873.
    1. Holmans P, Hamshere M, Hollingworth P, et al. Genome screen for loci influencing age at onset and rate of decline in late onset Alzheimer’s disease. Am J Med Genet B Neuropsychiatr Genet. 2005;135:24–32.
    1. Liu F, Arias-Vasquez A, Sleegers K, et al. A genomewide screen for late-onset Alzheimer disease in a genetically isolated dutch population. Am J Hum Genet. 2007;81:17–31.
    1. Myers A, Wavrant De-Vrieze F, Holmans P, et al. Full genome screen for Alzheimer disease: stage II analysis. Am J Med Genet. 2002;114:235–244.
    1. De Ferrari GV, Papassotiropoulos A, Biechele T, et al. Common genetic variation within the low-density lipoprotein receptor-related protein 6 and late-onset Alzheimer’s disease. Proc Natl Acad Sci USA. 2007;104:9434–9439.
    1. Reiman EM, Webster JA, Myers AJ, et al. GAB2 alleles modify Alzheimer’s risk in APOE epsilon4 carriers. Neuron. 2007;54:713–720.
    1. Dreses-Werringloer U, Lambert JC, Vingtdeux V, et al. A polymorphism in CALHM1 influences Ca2+ homeostasis, Abeta levels, and Alzheimer’s disease risk. Cell. 2008;133:1149–1161.
    1. Blacker D, Wilcox MA, Laird NM, et al. Alpha-2 macroglobulin is genetically associated with Alzheimer disease. Nat Genet. 1998;19:357–360.
    1. Giedraitis V, Hedlund M, Skoglund L, et al. New Alzheimer’s disease locus on chromosome 8. J Med Genet. 2006;43:931–935.
    1. Li Y, Nowotny P, Holmans P, et al. Association of late-onset Alzheimer’s disease with genetic variation in multiple members of the GAPD gene family. Proc Natl Acad Sci USA. 2004;101:15688–15693.
    1. Ozturk A, Desai PP, Minster RL, et al. Three SNPs in the GSTO1, GSTO2 and PRSS11 genes on chromosome 10 are not associated with age at onset of Alzheimer’s disease. Neurobiol Aging. 2005;26:1161–1165.
    1. Bertram L, Hiltunen M, Parkinson M, et al. Family-based association between Alzheimer’s disease and variants in UBQLN1. N Engl J Med. 2005;352:884–894.
    1. Blacker D, Bertram L, Saunders AJ, et al. Results of a high-resolution genome screen of 437 Alzheimer’s disease families. Hum Mol Genet. 2003;12:23–32.
    1. Farrer LA, Bowirrat A, Friedland RP, et al. Identification of multiple loci for Alzheimer disease in a consanguineous Israeli-Arab community. Hum Mol Genet. 2003;12:415–422.
    1. Hahs DW, McCauley JL, Crunk AE, et al. A genome-wide linkage analysis of dementia in the Amish. Am J Med Genet B Neuropsychiatr Genet. 2006;141:160–166.
    1. Lee JH, Cheng R, Santana V, et al. Expanded genomewide scan implicates a novel locus at 3q28 among Caribbean hispanics with familial Alzheimer disease. Arch Neurol. 2006;63:1591–1598.
    1. Pericak-Vance MA, Grubber J, Bailey LR, et al. Identification of novel genes in late-onset Alzheimer’s disease. Exp Gerontol. 2000;35:1343–1352.
    1. Rademakers R, Cruts M, Sleegers K, et al. Linkage and association studies identify a novel locus for Alzheimer disease at 7q36 in a Dutch population-based sample. Am J Hum Genet. 2005;77:643–652.
    1. Scott WK, Hauser ER, Schmechel DE, et al. Ordered-subsets linkage analysis detects novel Alzheimer disease loci on chromosomes 2q34 and 15q22. Am J Hum Genet. 2003;73:1041–1051.
    1. Li M, Atmaca-Sonmez P, Othman M, et al. CFH haplotypes without the Y402H coding variant show strong association with susceptibility to age-related macular degeneration. Nat Genet. 2006;38:1049–1054.
    1. Sweet RA, V, Nimgaonkar L, Devlin B, Jeste DV. Psychotic symptoms in Alzheimer disease: evidence for a distinct phenotype. Mol Psychiatry. 2003;8:383–392.
    1. Wijsman EM, Daw EW, Yu CE, et al. Evidence for a novel late-onset Alzheimer disease locus on chromosome 19p13.2. Am J Hum Genet. 2004;75:398–409.
    1. Daw EW, Heath SC, Wijsman EM. Multipoint oligogenic analysis of age-at-onset data with applications to Alzheimer disease pedigrees. Am J Hum Genet. 1999;64:839–851.
    1. Daw EW, Payami H, Nemens EJ, et al. The number of trait loci in late-onset Alzheimer disease. Am J Hum Genet. 2000;66:196–204.
    1. Gottesman II, Gould TD. The endophenotype concept in psychiatry: etymology and strategic intentions. Am J Psychiatry. 2003;160:636–645.
    1. Blangero J, Williams JT, Almasy L. Novel family-based approaches to genetic risk in thrombosis. J Thromb Haemost. 2003;1:1391–1397.
    1. Ebrahim S, Davey Smith G. Mendelian randomization: can genetic epidemiology help redress the failures of observational epidemiology? Hum Genet. 2008;123:15–33.
    1. Lehtovirta M, Helisalmi S, Mannermaa A, et al. Apolipoprotein E polymorphism and Alzheimer’s disease in eastern Finland. Neurosci Lett. 1995;185:13–15.
    1. Geschwind DH, Miller BL, DeCarli C, Carmelli D. Heritability of lobar brain volumes in twins supports genetic models of cerebral laterality and handedness. Proc Natl Acad Sci USA. 2002;99:3176–3181.

Source: PubMed

3
Subscribe