Alzheimer’s Disease: Genes
Sapeck Agrawal
Gaithersburg, MD, USA
DOI
//dx.doi.org/10.13070/mm.en.7.2226
Date
last modified : 2024-09-11; original version : 2017-04-07
Cite as
MATER METHODS 2017;7:2226
Abstract

A comprehensive review of susceptibility genes for Alzheimer's disease and a summary of antibodies against the Alzheimer's disease-related proteins cited among the over 60,000 formal publications Labome has surveyed for its Validated Antibody Database.

Introduction

Alzheimer’s disease (AD) is a progressive, neurodegenerative disorder known to occur in the elderly, with an onset age of around 60 years. AD represents the most common form of elderly dementia, characterized by memory loss (92% of all patients), confusion (71%), short attention span (63%), a declining sense of direction (53%), and personality changes (31%) [1]. Autopsy studies on brains of affected patients reveal the neuropathological hallmarks of AD, which include senile plaques of amyloid β (Aβ) protein and intraneuronal tangles of hyperphosphorylated tau protein, as well as gross cortical atrophy and ventricular dilation [2]. Approximately 1 in 8 people over the age of 65 years suffer from this debilitating disease, 65% of which are women [1]. Alzheimer’s disease poses a tremendous public health challenge to the world, as the average human life expectancy continues to improve. Todate, a total of ~ 46.8 million cases of dementia are recorded worldwide, with an associated health-care cost in the billions. By the year 2030, the number of cases will almost double [3].

Extensive research on the genetics of AD using traditional and modern genetic tools has, to a large extent, helped elucidate the etiology of AD, although many questions remain. This article presents a review of the genetics of AD attributed to the hallmark neuropathology of AD, including accumulation of Aβ and tau protein.

Strategies for Understanding the Genetics of Alzheimer’s Disease

There are two main strategies used to identify the genetic risk factors of AD. The first strategy employs a phenotype to genotype approach. Researchers examine polymorphic genomic markers such as short tandem repeats found commonly in families with a high burden of AD (two or more cases of AD among first–degree relatives) across multiple generations to identify broad genomic regions co-transmitted with the disease. This method, used to determine linkage or tendency of genes to be inherited together because of their spatial proximity, is known as linkage analysis. The technique is typically followed by positional cloning of candidate genes to test their contribution to AD. Linkage analyses were instrumental in the identification of genes associated with familial or early-onset AD, which is described in more detail below [4].

The second strategy for identifying the genetic risks of AD employs a genotype to phenotype approach. Researchers compare allele frequencies of available polymorphic markers such as single nucleotide polymorphisms (SNPs) between healthy (control) and sick (case) people to identify genes associated with AD. These studies are known as genome-wide association studies and represent modern genetic tools for studying complex or sporadic forms of AD [5] or other parameters such as the concentration of soluble TREM2 in cerebrospinal fluid [6].

Table 1 lists each method in detail, including its advantages and disadvantages and its contribution to the genetics of AD.

Alzheimer’s Disease: Genes  figure 1
Figure 1. Categories of Alzheimer’s Disease.
Categories of Alzheimer’s Disease

AD is commonly categorized based on the age of onset of symptoms. Early-onset AD (EOAD) typically begins before 65 years of age, while late-onset AD (LOAD) most commonly starts after 60 (see Figure 1). EOAD, also known as familial AD, is caused by inheritance of autosomal dominant mutations in three genes: APP, PSEN1, and PNSEN2, and accounts only for ~ 5% of all AD cases. A rare mutation of ADAM17 was also found to cause late-onset familial Alzheimer disease with an autosomal-dominant pattern [7]. The majority of AD cases fall under the LOAD category, also known as sporadic AD, which represents a much more complex disorder, and is associated with a multitude of genetic risk factors. While EOAD is 100% inheritable, LOAD is only 60-80% inheritable; the remaining cases of LOAD can be attributed to environmental influences (i.e., diet, brain injury, lifestyle, certain medications) [1].

Early-onset AD

Linkage analyses of the affected families led to the discovery of APP, PSEN1, and PSEN2 (Table 2), with approximately 250 mutations identified in these genes that are responsible for disrupting their normal biological function, leading to deposition of Aβ plaques in the brain. APOE E4 may explain around 10.1% of the variance of EOAD and rare coding variants in APOB may also contribute to EOAD [8].

Assay Method Advantages Disadvantages Genes
Linkage Analysis Phenotype to genotype approach:
Studies examine polymorphic genomic markers such as short tandem repeats found commonly in families with a high AD burden to identify broad genomic regions co-transmitted with the disease. This is followed by positional cloning of candidate genes to identify genes contributing to AD.
Help identify genomic regions associated with disease
Can simultaneously study multiple genetic markers
Need to identify families with a high burden of the disease over multiple generations.
Not suited for complex conditions affected by multiple genes, such as LOAD.
APP, PS1, PS2, APOE [9-13]
Genome-wide Association Studies Genotype to phenotype approach:
Compare allele frequencies of available polymorphic markers such as SNPs between healthy (control) and sick (case) people to identify genes associated with a trait of interest.
Used to study less common diseases without relying on studying many families and generations with the disease
A high-throughput technique that can study millions of single nucleotide polymorphisms simultaneously using SNP arrays
Can study entire genome at the same time by whole genome sequencing or whole exome sequencing
Relies on statistical association, thus, cannot determine causality
Studies examine genes with a hypothetical function related to disease or proximity to linkage signals.
Tables 4 and 5
Table 1. Methods for identifying genes related to AD.
Amyloid precursor protein (APP)

The APP gene is on chromosome 21. Researchers have identified over 50 AD-associated APP mutations with new missense/deletion APP mutations being reported [14, 15]. The gene encodes the APP protein that undergoes proteolytic cleavage by three distinct proteases called α-, β- and γ-secretase. Proteolysis of APP by β and γ-secretase produces two amyloid-β species called sAPPβ and β-CTF. Aβ, the protein responsible for the senile amyloid plaques seen in AD-affected brains, is produced from further processing of βCTF. While most AD-causing APP mutations are dominant, some recessive mutations have also been described [1]. Down syndrome patients older than 65 years, with the trisomy of chromosome 21 (hence the overexpression of APP gene), has a prevalence of nearly 80% for Alzheimer disease [16] ; for those older than 55 years, in another study, the prevalence is 32.7% [17]. A short, 17-amino acid peptide in sAPP binds to the sushi 1 domain of GABABR subunit 1a and is a physiological ligand for metabotropic GABA receptors [18]. APP gene, more specifically, the beta amyloid peptide derived from it, is the most common therapeutic target for Alzheimer's disease [19].

Gene Chromosome Biological Function Method Used to Identify Seminal Studies
APP 21 Encodes the APP protein needed for neuronal development, synaptic formation and repair, β-amyloid production, somatic gene recombination (gencDNAs) [20] Linkage analysis [13, 21-23]
PSEN1 14 Encodes the PSEN1 protein, a major component of γ-secretase complex that processes the APP protein Linkage analysis [12, 24-26]
PSEN2 1 Encodes the PSEN2 protein, a major component of γ-secretase complex that processes the APP protein Linkage analysis [10, 11]
Table 2. Genes implicated in early-onset AD (Adapted from [1] ).
Presenilins

The presenilin-1 (PSEN1) gene is located on Chromosome 14. The encoded protein is a component of the γ-secretase complex that is needed in APP processing. Over 200 AD-associated PSEN1 mutations have been identified so far. PSEN1 are responsible for 50% of all EOAD, with new ones being identified. For example, Jia L et al examined 404 pedigrees and identified 10 new missense PSEN1 mutations [27]. The PSEN2 gene is located on Chromosome 1. New mutations are identified constantly. Van Giau V et al locate a new mutation at Trp 165 of PSEN1 associated with EOAD [28]. Itzcovich T et al identified a novel T119I dominant mutation in PSEN1 in an Argentine family with early and late-onset Alzheimer’s disease [29]. Kim YE et al reported both pathogenic variants of PSEN1 gene and variants of unknown significance in early onset Korean Alzheimer's patients [30]. PSEN2 mutations are much rarer; a total of only 13 pathogenic mutations have been identified in 29 families. Similar to PSEN1, PSEN2 is also a component of the γ-secretase complex and is needed for APP processing [1].

Allele Amino acids Frequency Role in LOAD
112 158 Popu* AD
E2 Cys Cys 8% 3.9% May be protective
E3 Cys Arg 78% 59.4% Not associated
E4 Arg Arg 14% 36.7% Associated
Table 3. Common APOE alleles and contribution to LOAD. Popu*: The frequencies reported represent those for the Caucasian population, which are different from frequencies reported in other ethnicities. Reference Naj et al, 2016 [4] for details.
Late-onset AD (LOAD)
Apolipoprotein E

LOAD represents a complex form of Alzheimer’s disease. However, one gene, Apolipoprotein E (APOE) has the strongest causative effect on LOAD. APOE was first identified in 1991 using linkage analysis on chromosome 19 [31]. The encoded protein is produced by astrocytes in the nervous system and is responsible for transporting cholesterol to neurons, as well as neuronal growth, repair response to tissue injury, nerve regeneration, immunoregulation, and activation of lipolytic enzymes [32]. APOE is a polymorphic gene, which is expressed in three different isoforms (Table 3). These include E2, E3, and E4. Isoform E4 represents a genetic risk factor for LOAD that can increase a person’s risk for LOAD by 3 to 8 times. Isoform E4 is associated with approximately 36.7% of LOAD cases [33] and likely accelerates amyloid deposition, possibly beginning as early as the 30-40 years of age [34]. ApoE E4/4 homozygotes also tend to have olfactory dysfunction [35]. Interestingly, isoform E2 may decrease a person’s risk of developing LOAD [33], while E3 appears to have a neutral biological effect. A mutation at ApoE3 (R136S) potentially delays or prevents the onset of Alzheimer’s disease in an autosomal dominant PSEN1 E280A mutation carrier [36]. A gain-of-function mutation of RELN, which, like ApoE, binds to VLDLr and APOEr2 receptors, delays the onset of dementia [37].

Method Sample Size Strongest AD-correlative Genes
Cases Controls
GWAS111,326 677,663 111,326 clinically diagnosed/‘proxy’ AD cases. Identified 75 risk loci: 33 loci already known to be associated with the risk of developing Alzheimer's Disease including BIN, CR1, CLU, MS4A, PICALM etc. and 42 new loci associated with GRN, TSPAN14 etc. [38].
2,247 1,669 2247 subjects from 605 multiplex AD families. Identified 13 new risk gene variants: FNBP1L, SEL1L, LINC00298, PRKCH, C15ORF41, C2CD3, KIF2A, APC, LHX9, NALCN, CTNNA2, SYTL3, and CLSTN2 [39].
21,982 41,944 Discovered five new risk genes: IQCK, ACE, ADAM10, ADAMTS1, and WWOX; Confirmed 20 known genes: CR1, BIN1, INPP5D, HLA-DRB1, TREM2, CD2AP, NYAP1, EPHA1, PTK2B, CLU, ECHDC3, SPI1, MS4A2, PICALM, SORL1, FERMT2, SLC24A4, ABCA7, CASS4 [40].
4120 3289 AD cases were grouped into 6 endophenotypes. APOE, BIN1, MAPT, MS4A2, PICALM; ANKRD31 (meta-analysis with nine additional databases); CD33, HBEGF(?) (meta-analysis with IGAP stage I summary statistics; NDUFAF6 (meta-analysis with IGAP I and II summary statistics); SCIMP ( meta-analysis of GR@ACE AD+++ endophenotype data with IGAP I and II) [41]
71,880 383,378 Cases include AD-by-proxy. ABCA7, ABI3, AC074212.3, ADAM10, ADAMTS4, ALPK2, APH1B, APOE, BIN1, CASS4, CD2AP, CD33, CLNK, CLU/PTK2B, CNTNAP2, CR1, ECHDC3, EPHA1, HESX1, HLA-DRB1, INPPD5, KAT8, MS4A6A, PICALM, SCIMP, SLC24A4, SORL1, TREM2, ZCWPW1 [42]
21392 38164 Two-stage data; CR1, BIN1, PTK2B, CLU, MS4A4A, PICALM, ABCA7, TPBG; SNP-level: PFDN1/HBEGF, USP6NL/ECHDC3, BZRAP1-AS1, and NFIC [43]
2032 5328 CLU (clusterin/ApoJ), CR1 [44]
3941 7848 CLU, PICALM [45]
3006 14642 BIN1, XOC3L2/BLOC1S3/MARK4, CLU, PICALM [46]
8309 7366 MS4A4A, CD2AP, CD33, EPHA1, CR1, CLU, BIN1, PICALM [47]
6688 13685 ABCA7, MS4A6A/MS4A4E, EPHA1, CD33, CD2AP [48]
17008 37154 CR1, BIN1, CD2AP, EPHA1, CLU, MS4A4A, PICALM, ABCA7, HLA-region, PTK2B, SORL1, SLC24A4-RIN3, INPP5D, MEF2C, NME8, ZCWPW1, CELF1, FERMT2, CASS4 [49]
2025 5328 FRMD4A [50]
1968 3928 ABCA7, intergenic locus on 5q35.2 [51]
3001 2299 FBXL7 [52]
85133 a protective variant in PLCG2, risk variants in ABI3 and TREM2 [53]
Whole Exome Sequencing or Whole Genome Sequencing 3550 8888 TREM2 [54]
LOAD patients from 5 families 410 pedigrees from NIMH Alzheimer's Disease Genetics Initiative a rare mutation in GGA3 [55]
2 LOAD 4 ADAM17 [7], an autosomal-dominant mutation in the European population, validated in additional 5450 cases/control samples
8050 98194 UNC5C [56], with an autosomal dominant variant identified first among a large pedigree with eight LOAD cases, fifteen unaffected family members, and four individuals of unknown LOAD status
132 53 PILRA [57]
12 sporadic EOAD trois PSEN1, VPS35, MARK4 [58]
1092 1107 TREM2 [59]
506 86 ABCA7, BIN1, CD2AP, EPHA1 [60]
365 80 SORL1 [61]
14 None PLD3 [62]
1459 2263 AKAP9 [63]
927 LOAD; 852 EOAD 1273 exome-wide significant association between EOAD risk and rare variants in SORL1, TREM2 and ABCA7; No exome-wide significant signal was obtained in the LOAD sample [64].
Candidate Gene Analyses 436 None ADAM10 [65]
311 360 NICASTRIN [66]
311 360 ABCA1 [67]
Table 4. Genetic risk factors for LOAD identified in genome-wide association studies and other association studies (Table adapted from [5] ).
Other genetic risk factors

Until 2009, APOE was the only genetic risk factor described for LOAD. However, only about 30% of all LOAD cases can be attributed to APOE [32]. Genetic risk factors that can account for the remaining cases of LOAD include polymorphisms or mutations of multiple genes involved in several biological pathways. Lately, emerging techniques such as Genome-wide Association Studies (GWAS), whole exome/genome sequencing, and candidate gene analysis studies are spearheading the identification of the genetic risk factors contributing to onset and/or progression of LOAD (Table 4).

Gene Biological Function Cause
CLU Encodes a chaperone protein needed for apoptosis and clearance of cellular debris, as well as lipid transport and inflammation SNP
CR1 Encodes the main receptor of complement C3b protein that binds Aβ, mediates innate immunity SNP
BIN1 Clathrin-mediated endocytosis, inflammation, calcium homeostasis, and apoptosis; may mediate AD risk through tau pathology. SNP
PICALM Phosphatidylinositol-binding clathrin assembly protein (PICALM) is involved in synaptic neurotransmitter release and intracellular trafficking SNP
CD33 Immunological protein expressed on myeloid cells; mediates cell-cell interactions SNP
EPHA1 Encodes the EPH protein needed for developmental events in the nervous system SNP
TREM2 Encodes a protein involved in immune responses, particularly inflammatory. Protein impacts the function of microglia involved in AB pathway and the switching of microglia types [68]. Loss of function (missense) mutation
ABCA7 Highly expressed in hippocampal neurons; involved in ABC transport protein; function unclear SNP / haplodeficiency [69]
SORL1 Protein involved in endocytosis and sorting SNP/ nonsense and missense mutations; somatic mutations [70]
ADAM10 Gene encodes a major α-secretase in the brain. The encoded protein contains both adhesion and protease domain that cleaves between the Aβ domain of APP to prevent the formation of β-amyloid Mutations
ADAM17 Gene encodes an α-secretase in the brain. The encoded protein contains both adhesion and protease domain that cleaves between the Aβ domain of APP to prevent the formation of β-amyloid Loss of function (missense) mutation
Table 5. Genes identified in multiple studies with strong AD association [1, 32].

Among the genetic risk factors described in the table above, some have been identified repeatedly in multiple genetic studies. Thus, a more prominent association with LOAD is presumed. These genes include CLU, CR1, BIN1, PICALM, CD33, EPHA1, TREM2, ABCA7, SORL1, and ADAM10 [5]. The contribution of common and rare variants of SORL1 in Alzheimer's disease is summarized in a recent review [71]. The encoded proteins of these genes are involved in a multitude of biological pathways such as immune response, protein trafficking, endocytosis and sorting, lipid metabolism, APP processing, tau pathology, and gene regulation. Mutations in the TREM2 gene, in particular, are associated with 3 to 5 times higher risk of developing AD [54]. Variants in TREM2 and also in another gene, NOTCH3, may result in not only Alzheimer's disease but also other dementias (Nasu-Hakola disease and cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy) [72]. Increased soluble TREM2 in CSF correlates with reduced cognitive and clinical decline in Alzheimer’s disease [73], which has been explored to identify variants in the MS4A gene region as modulators of Alzheimer disease risk [6]. TREM2 and APOE forms an axis that might convert microglia from a homeostatic phenotype to a neurodegenerative one after phagocytosis of apoptotic neurons [74, 75]. TREM2, present in tumor-infiltrating macrophages, is also explored in immuno-oncotherapy [76]. INPP5D transmits the signalling of CD22, a regulator of microglial phagocytosis [77].

Table 5 includes details on each genetic risk factor. It is important to note that some genetic risk factors may weigh heavier in some populations than in others. For example, a rare variant in TM2D3 is disproportionately-enriched in Icelanders (~0.5% compared to <0.05% in other European populations) and is associated with a 7.5 times increased risk of LOAD, as shown in an exome-wide association analysis-based study [78], while a rare variant of ADAM17 causing late-onset familial AD is identified only in Europeans.

Pathway Genes involved
Immune response / inflammation CLU, ABCA7 [69], HLA-DRB5, INPP5D, MEF2C, CR1
APP processing SORL1, CASS4, VPS35, ADAM10, ADAM17
Tau pathology CASS4, FERMT2, BIN1, MARK4
Cell migration PTK2B
Lipid transport and endocytosis SORL1, PICALM, BIN1
Synaptic function MEF2C, PTK2B
Cytoskeletal function and axonal transport CELF1, NME8, CASS4
Regulation of gene expression, post-translational modification of protein, microglial and myeloid cell function INPPD5
Cholesterol metabolism APOE, CLU
Transcytosis PICALM, BIN1
Table 6. Biological pathways involved in AD pathogenesis [33, 79].
GeneGene descriptionTop three suppliers
ABCA1 ATP binding cassette subfamily A member 1 Abcam ab18180 (27), Santa Cruz Biotechnology sc-58219 (5), Novus Biologicals NB100-2068 (4)
ADAM10 ADAM metallopeptidase domain 10 Abcam ab124695 (8), Santa Cruz Biotechnology sc-48400 (2), Invitrogen MA5-32616 (1)
APC APC regulator of WNT signaling pathway Abcam ab16794 (21), Santa Cruz Biotechnology sc-53165 (1)
APOB apolipoprotein B Santa Cruz Biotechnology sc-13538 (2)
APOE apolipoprotein E Abcam ab1906 (14), Novus Biologicals NB110-60531 (7), Santa Cruz Biotechnology sc-13521 (4)
APP amyloid beta precursor protein BioLegend 803001 (151), Invitrogen 13-0200 (47), Abcam ab32136 (44)
BACE1 beta-secretase 1 Cell Signaling Technology 5606 (45), Abcam ab108394 (13), Invitrogen MA1-177 (2)
BACE2 beta-site APP-cleaving enzyme 2 Santa Cruz Biotechnology sc-271212 (1)
BIN1 bridging integrator 1 Santa Cruz Biotechnology sc-13575 (2), Abcam ab185950 (2)
CD2AP CD2 associated protein Santa Cruz Biotechnology sc-25272 (5)
CD33 CD33 BD Biosciences 561157 (18), BioLegend 303419 (12), Beckman Coulter A54824 (7)
CELF1 CUGBP Elav-like family member 1 Santa Cruz Biotechnology sc-20003 (15), Invitrogen MA1-16675 (2), MBL International rn034pw (2)
CLU clusterin Santa Cruz Biotechnology sc-5289 (7), Sino Biological 11297-R210 (3), Abcam ab92548 (2)
CNTNAP2 contactin associated protein like 2 Neuromab 75-075 (3)
CR1 complement C3b/C4b receptor 1 (Knops blood group) BD Biosciences 555451 (5), Dako M0846 (4), Santa Cruz Biotechnology sc-166329 (3)
CTNNA2 catenin alpha 2 Invitrogen 13-9700 (20), Cell Signaling Technology 2163 (2), BD Biosciences 562505 (2)
FERMT2 fermitin family member 2 OriGene TA500505 (1)
GABBR1 gamma-aminobutyric acid type B receptor subunit 1 Abcam ab55051 (7)
GGA3 Golgi-associated, gamma adaptin ear containing, ARF binding protein 3 BD Biosciences 610502 (14), Cell Signaling Technology 8027 (1)
HBEGF heparin binding EGF like growth factor MBL International M220-3 (4)
INPP5D inositol polyphosphate-5-phosphatase D Santa Cruz Biotechnology sc-8425 (4), Cell Signaling Technology 2727 (3), Miltenyi Biotec 130-109-155 (1)
KAT8 lysine acetyltransferase 8 Santa Cruz Biotechnology sc-271691 (5), Abcam ab200660 (2), Invitrogen MA5-15345 (1)
MAPT microtubule associated protein tau Invitrogen MN1020 (751), Abcam ab80579 (32), Cell Signaling Technology 9632 (17)
MEF2C myocyte enhancer factor 2C Cell Signaling Technology 5030 (14), Invitrogen MA5-17119 (1), Santa Cruz Biotechnology sc-365862 (1)
MS4A4A membrane spanning 4-domains A4A BioLegend 372502 (1)
NCSTN nicastrin Cell Signaling Technology 9447 (5)
NFIC nuclear factor I C Santa Cruz Biotechnology sc-74445 X (1)
PLCG2 phospholipase C gamma 2 Santa Cruz Biotechnology sc-5283 (5)
PRKCH protein kinase C eta Abcam ab179524 (1)
PSEN1 presenilin 1 Cell Signaling Technology 5643 (19), Abcam ab76083 (4), Invitrogen MA1-752 (3)
PTK2B protein tyrosine kinase 2 beta Cell Signaling Technology 3480 (10), Abcam ab32571 (3), Santa Cruz Biotechnology sc-81512 (1)
PYCARD PYD and CARD domain containing Santa Cruz Biotechnology sc-514414 (12), MBL International D086-3 (5), LifeSpan Biosciences LS-C175123 (4)
RELN reelin Abcam ab78540 (8), Santa Cruz Biotechnology sc-25346 (3)
SEL1L SEL1L adaptor subunit of ERAD E3 ubiquitin ligase Santa Cruz Biotechnology sc-377350 (2)
SORL1 sortilin related receptor 1 BD Biosciences 612633 (3), Abcam ab190684 (2), Santa Cruz Biotechnology sc-136073 (1)
TPBG trophoblast glycoprotein Invitrogen MA5-32120 (1), Abcam ab134162 (1)
TREM2 triggering receptor expressed on myeloid cells 2 Santa Cruz Biotechnology sc-373828 (3), R&D Systems MAB1828 (2), Abcam ab209814 (1)
VPS35 VPS35, retromer complex component Abcam ab57632 (8), Santa Cruz Biotechnology sc-374372 (5), Abnova H00055737-M02 (2)
Table 7. A list of Alzheimer's disease-related genes, and top suppliers of antibodies against them cited in the over 60,000 publications Labome has surveyed for Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.

The major biological pathways associated with AD pathogenesis include protein sorting, an immune/inflammatory response such as inflammasome and ASC specks [80], cholesterol and lipid metabolism, and APP processing. Table 6 lists the key biological pathways associated with LOAD that have been discovered with GWAS.

Microtubule-associated protein Tau

The Tau protein, encoded by the MAPT gene, is associated with microtubules in the neurons and functions to stabilize them. However, hyperphosphorylated variants of tau proteins, especially those phosphorylated at Ser262 [81], fail to interact with microtubules and clump together to form intraneuronal tangles. The result is a collapsed neuronal cytoskeletal system, rendering the neurons unable to function properly. Increased tau proteins in cerebrospinal fluid (CSF) correlate with risk of AD onset. GWAS reveal the role of APOE and TREM2 in the variability of tau levels in the CSF. Other genes implicated in tau pathology include BIN1, PICALM, and FERMT2 [32].

Genes Not Related to Alzheimer's Disease

Common and rare TBK1 variants are not significantly represented in early-onset Alzheimer disease in a European cohort of 1253 patients; they are known to cause frontotemporal dementia and amyotrophic lateral sclerosis [82].

Conclusions

Alzheimer’s disease is a major public health concern as people’s life expectancy improves worldwide. While mutations on key genes can explain familial AD or early-onset AD, i.e., APP, PSEN1, and PSEN2, identified using linkage analyses, much of sporadic AD or late-onset AD stems from a combination of several etiological risk factors. These include both environmental (i.e., diet, brain injury, lifestyle, certain medications) and genetic factors. genome-wide association studies have been instrumental in identifying several genetic risk factors that can predispose people to the disease. These risk factors include polymorphisms or mutations in several genes that play important roles in a multitude of biological pathways protein sorting, immune/inflammatory response, cholesterol, and lipid metabolism, and APP processing. These genes are CLU, CR1, BIN1, PICALM, CD33, EPHA1, TREM2, ABCA7, SORL1, and ADAM10. Scientists continue to make progress in gaining a deeper understanding on the neuropathology of the disease by relying on traditional research models such as transgenic and non-transgenic animals and modern research models including induced pluripotent stem cells, cultured brain tissue, and molecular simulation models. Research on the genes involved in Alzheimer's disease is essential to develop effective preventive and/or therapeutical intervention approaches. Table 7 lists the antibodies against the genes discussed in this article, based on Labome's Validated Antibody Database.

References
  1. Giri M, Zhang M, Lu Y. Genes associated with Alzheimer's disease: an overview and current status. Clin Interv Aging. 2016;11:665-81 pubmed publisher
  2. Karch C, Cruchaga C, Goate A. Alzheimer's disease genetics: from the bench to the clinic. Neuron. 2014;83:11-26 pubmed publisher
  3. Cuyvers E, Sleegers K. Genetic variations underlying Alzheimer's disease: evidence from genome-wide association studies and beyond. Lancet Neurol. 2016;15:857-868 pubmed publisher
  4. Naj A, Schellenberg G. Genomic variants, genes, and pathways of Alzheimer's disease: An overview. Am J Med Genet B Neuropsychiatr Genet. 2017;174:5-26 pubmed publisher
  5. Tosto G, Reitz C. Genomics of Alzheimer's disease: Value of high-throughput genomic technologies to dissect its etiology. Mol Cell Probes. 2016;30:397-403 pubmed publisher
  6. Deming Y, Filipello F, Cignarella F, Cantoni C, Hsu S, Mikesell R, et al. The MS4A gene cluster is a key modulator of soluble TREM2 and Alzheimer's disease risk. Sci Transl Med. 2019;11: pubmed publisher
  7. Hartl D, May P, Gu W, Mayhaus M, Pichler S, Spaniol C, et al. A rare loss-of-function variant of ADAM17 is associated with late-onset familial Alzheimer disease. Mol Psychiatry. 2018;: pubmed publisher
  8. Wingo T, Cutler D, Wingo A, Le N, Rabinovici G, Miller B, et al. Association of Early-Onset Alzheimer Disease With Elevated Low-density Lipoprotein Cholesterol Levels and Rare Genetic Coding Variants of APOB. JAMA Neurol. 2019;: pubmed publisher
  9. Goate A. Segregation of a missense mutation in the amyloid beta-protein precursor gene with familial Alzheimer's disease. J Alzheimers Dis. 2006;9:341-7 pubmed
  10. Levy Lahad E, Wasco W, Poorkaj P, Romano D, Oshima J, Pettingell W, et al. Candidate gene for the chromosome 1 familial Alzheimer's disease locus. Science. 1995;269:973-7 pubmed
  11. Rogaev E, Sherrington R, Rogaeva E, Levesque G, Ikeda M, Liang Y, 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. 1995;376:775-8 pubmed
  12. Sherrington R, Rogaev E, Liang Y, Rogaeva E, Levesque G, Ikeda M, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature. 1995;375:754-60 pubmed
  13. Suzuki N, Cheung T, Cai X, Odaka A, Otvos L, Eckman C, et al. An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants. Science. 1994;264:1336-40 pubmed
  14. Boyer M, Cayrefourcq L, Garima F, Foulongne V, Dereure O, Alix Panabières C. Circulating Tumor Cell Detection and Polyomavirus Status in Merkel Cell Carcinoma. Sci Rep. 2020;10:1612 pubmed publisher
  15. Pagnon de la Vega M, Giedraitis V, Michno W, Kilander L, Guner G, Zielinski M, et al. The Uppsala APP deletion causes early onset autosomal dominant Alzheimer's disease by altering APP processing and increasing amyloid β fibril formation. Sci Transl Med. 2021;13: pubmed publisher
  16. McCarron M, McCallion P, Reilly E, Dunne P, Carroll R, Mulryan N. A prospective 20-year longitudinal follow-up of dementia in persons with Down syndrome. J Intellect Disabil Res. 2017;61:843-852 pubmed publisher
  17. RUBENSTEIN E, Hartley S, Bishop L. Epidemiology of Dementia and Alzheimer Disease in Individuals With Down Syndrome. JAMA Neurol. 2019;: pubmed publisher
  18. Rice H, de Malmazet D, Schreurs A, Frere S, Van Molle I, Volkov A, et al. Secreted amyloid-β precursor protein functions as a GABABR1a ligand to modulate synaptic transmission. Science. 2019;363: pubmed publisher
  19. Hettmann T, Gillies S, Kleinschmidt M, Piechotta A, Makioka K, Lemere C, et al. Development of the clinical candidate PBD-C06, a humanized pGlu3-Aβ-specific antibody against Alzheimer's disease with reduced complement activation. Sci Rep. 2020;10:3294 pubmed publisher
  20. Lee M, Siddoway B, Kaeser G, Segota I, Rivera R, Romanow W, et al. Somatic APP gene recombination in Alzheimer's disease and normal neurons. Nature. 2018;563:639-645 pubmed publisher
  21. Goldgaber D, Lerman M, McBride O, Saffiotti U, Gajdusek D. Characterization and chromosomal localization of a cDNA encoding brain amyloid of Alzheimer's disease. Science. 1987;235:877-80 pubmed
  22. Tanzi R, Bird E, Latt S, Neve R. The amyloid beta protein gene is not duplicated in brains from patients with Alzheimer's disease. Science. 1987;238:666-9 pubmed
  23. Hilbich C, Kisters Woike B, Reed J, Masters C, Beyreuther K. Human and rodent sequence analogs of Alzheimer's amyloid beta A4 share similar properties and can be solubilized in buffers of pH 7.4. Eur J Biochem. 1991;201:61-9 pubmed
  24. Selkoe D. Presenilin, Notch, and the genesis and treatment of Alzheimer's disease. Proc Natl Acad Sci U S A. 2001;98:11039-41 pubmed
  25. Lemere C, Blusztajn J, Yamaguchi H, Wisniewski T, Saido T, Selkoe D. Sequence of deposition of heterogeneous amyloid beta-peptides and APO E in Down syndrome: implications for initial events in amyloid plaque formation. Neurobiol Dis. 1996;3:16-32 pubmed
  26. Mann D, Iwatsubo T, Cairns N, Lantos P, Nochlin D, Sumi S, et al. Amyloid beta protein (Abeta) deposition in chromosome 14-linked Alzheimer's disease: predominance of Abeta42(43). Ann Neurol. 1996;40:149-56 pubmed
  27. Jia L, Fu Y, Shen L, Zhang H, Zhu M, Qiu Q, et al. PSEN1, PSEN2, and APP mutations in 404 Chinese pedigrees with familial Alzheimer's disease. Alzheimers Dement. 2020;16:178-191 pubmed publisher
  28. Van Giau V, Pyun J, Suh J, Bagyinszky E, An S, Kim S. A pathogenic PSEN1 Trp165Cys mutation associated with early-onset Alzheimer's disease. BMC Neurol. 2019;19:188 pubmed publisher
  29. Itzcovich T, Chrem Mendez P, Vazquez S, Barbieri Kennedy M, Niikado M, Martinetto H, et al. A novel mutation in PSEN1 (p.T119I) in an Argentine family with early- and late-onset Alzheimer's disease. Neurobiol Aging. 2019;: pubmed publisher
  30. Kim Y, Cho H, Kim H, Na D, Seo S, Ki C. PSEN1 variants in Korean patients with clinically suspicious early-onset familial Alzheimer's disease. Sci Rep. 2020;10:3480 pubmed publisher
  31. Pericak Vance M, Bebout J, Gaskell P, Yamaoka L, Hung W, Alberts M, et al. Linkage studies in familial Alzheimer disease: evidence for chromosome 19 linkage. Am J Hum Genet. 1991;48:1034-50 pubmed
  32. Van Cauwenberghe C, Van Broeckhoven C, Sleegers K. The genetic landscape of Alzheimer disease: clinical implications and perspectives. Genet Med. 2016;18:421-30 pubmed publisher
  33. Martins R, Turner B, Carroll R, Sweeney D, Kim K, Wisniewski H, et al. High levels of amyloid-beta protein from S182 (Glu246) familial Alzheimer's cells. Neuroreport. 1995;7:217-20 pubmed
  34. Bussy A, Snider B, Coble D, Xiong C, Fagan A, Cruchaga C, et al. Effect of apolipoprotein E4 on clinical, neuroimaging, and biomarker measures in noncarrier participants in the Dominantly Inherited Alzheimer Network. Neurobiol Aging. 2019;75:42-50 pubmed publisher
  35. Oleson S, Murphy C. Olfactory Dysfunction in ApoE ?4/4 Homozygotes with Alzheimer's Disease. J Alzheimers Dis. 2015;46:791-803 pubmed publisher
  36. Arboleda Velasquez J, Lopera F, O Hare M, Delgado Tirado S, Marino C, Chmielewska N, et al. Resistance to autosomal dominant Alzheimer's disease in an APOE3 Christchurch homozygote: a case report. Nat Med. 2019;: pubmed publisher
  37. Resilience to autosomal dominant Alzheimer’s disease in a Reelin-COLBOS heterozygous man. Available from: www.nature.com/articles/s41591-023-02318-3
  38. Bellenguez C, Küçükali F, Jansen I, Kleineidam L, Moreno Grau S, Amin N, et al. New insights into the genetic etiology of Alzheimer's disease and related dementias. Nat Genet. 2022;54:412-436 pubmed publisher
  39. Prokopenko D, Morgan S, Mullin K, Hofmann O, Chapman B, Kirchner R, et al. Whole-genome sequencing reveals new Alzheimer's disease-associated rare variants in loci related to synaptic function and neuronal development. medRxiv. 2020;: pubmed publisher
  40. Kunkle B, Grenier Boley B, Sims R, Bis J, Damotte V, Naj A, et al. Genetic meta-analysis of diagnosed Alzheimer's disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat Genet. 2019;51:414-430 pubmed publisher
  41. Moreno Grau S, de Rojas I, Hernandez I, Quintela I, Montrreal L, Alegret M, et al. Genome-wide association analysis of dementia and its clinical endophenotypes reveal novel loci associated with Alzheimer's disease and three causality networks: The GR@ACE project. Alzheimers Dement. 2019;15:1333-1347 pubmed publisher
  42. Jansen I, Savage J, Watanabe K, Bryois J, Williams D, Steinberg S, et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer's disease risk. Nat Genet. 2019;51:404-413 pubmed publisher
  43. Jun G, Chung J, Mez J, Barber R, Beecham G, Bennett D, et al. Transethnic genome-wide scan identifies novel Alzheimer's disease loci. Alzheimers Dement. 2017;13:727-738 pubmed publisher
  44. Lambert J, 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 pubmed publisher
  45. Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere M, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease. Nat Genet. 2009;41:1088-93 pubmed publisher
  46. Seshadri S, Fitzpatrick A, Ikram M, Destefano A, Gudnason V, Boada M, et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA. 2010;303:1832-40 pubmed publisher
  47. Naj A, Jun G, Beecham G, Wang L, Vardarajan B, 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 pubmed publisher
  48. Hollingworth P, Harold D, Sims R, Gerrish A, Lambert J, Carrasquillo M, et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat Genet. 2011;43:429-35 pubmed publisher
  49. Lambert J, Ibrahim Verbaas C, Harold D, Naj A, Sims R, Bellenguez C, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat Genet. 2013;45:1452-8 pubmed publisher
  50. Lambert J, Grenier Boley B, Harold D, Zelenika D, Chouraki V, Kamatani Y, et al. Genome-wide haplotype association study identifies the FRMD4A gene as a risk locus for Alzheimer's disease. Mol Psychiatry. 2013;18:461-70 pubmed publisher
  51. Reitz C, Jun G, Naj A, Rajbhandary R, Vardarajan B, Wang L, et al. Variants in the ATP-binding cassette transporter (ABCA7), apolipoprotein E ?4,and the risk of late-onset Alzheimer disease in African Americans. JAMA. 2013;309:1483-92 pubmed publisher
  52. Tosto G, Fu H, Vardarajan B, Lee J, Cheng R, Reyes Dumeyer D, et al. F-box/LRR-repeat protein 7 is genetically associated with Alzheimer's disease. Ann Clin Transl Neurol. 2015;2:810-20 pubmed publisher
  53. Sims R, van der Lee S, Naj A, Bellenguez C, Badarinarayan N, Jakobsdottir J, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer's disease. Nat Genet. 2017;49:1373-1384 pubmed publisher
  54. Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson P, Snaedal J, et al. Variant of TREM2 associated with the risk of Alzheimer's disease. N Engl J Med. 2013;368:107-16 pubmed publisher
  55. S. LOMOIO, R. WILLEN, K. Z. HO, W. KIM, E. K. ROBINSON, R. E. TANZI, G. TESCO. GGA3 gene knock out and a novel AD-linked GGA3 gene mutation result in BACE1 accumulation in axonal swellings. Program No. 626.02. 2018 Neuroscience Meeting Planner. San Diego, CA: Society for Neuroscience, 2018. Online.
  56. Wetzel Smith M, Hunkapiller J, Bhangale T, Srinivasan K, Maloney J, Atwal J, et al. A rare mutation in UNC5C predisposes to late-onset Alzheimer's disease and increases neuronal cell death. Nat Med. 2014;20:1452-7 pubmed publisher
  57. Patel T, Brookes K, Turton J, Chaudhury S, Guetta Baranes T, Guerreiro R, et al. Whole-exome sequencing of the BDR cohort: Evidence to support the role of the PILRA gene in Alzheimer's disease. Neuropathol Appl Neurobiol. 2017;: pubmed publisher
  58. Rovelet Lecrux A, Charbonnier C, Wallon D, Nicolas G, Seaman M, Pottier C, et al. De novo deleterious genetic variations target a biological network centered on A? peptide in early-onset Alzheimer disease. Mol Psychiatry. 2015;20:1046-56 pubmed publisher
  59. 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 pubmed publisher
  60. Vardarajan B, Ghani M, Kahn A, Sheikh S, Sato C, Barral S, et al. Rare coding mutations identified by sequencing of Alzheimer disease genome-wide association studies loci. Ann Neurol. 2015;78:487-98 pubmed publisher
  61. Vardarajan B, Zhang Y, Lee J, Cheng R, Bohm C, Ghani M, et al. Coding mutations in SORL1 and Alzheimer disease. Ann Neurol. 2015;77:215-27 pubmed publisher
  62. Cruchaga C, Karch C, Jin S, Benítez B, Cai Y, Guerreiro R, et al. Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer's disease. Nature. 2014;505:550-554 pubmed publisher
  63. Logue M, Schu M, Vardarajan B, Farrell J, Bennett D, Buxbaum J, et al. Two rare AKAP9 variants are associated with Alzheimer's disease in African Americans. Alzheimers Dement. 2014;10:609-618.e11 pubmed publisher
  64. Bellenguez C, Charbonnier C, Grenier Boley B, Quenez O, Le Guennec K, Nicolas G, et al. Contribution to Alzheimer's disease risk of rare variants in TREM2, SORL1, and ABCA7 in 1779 cases and 1273 controls. Neurobiol Aging. 2017;59:220.e1-220.e9 pubmed publisher
  65. Kim M, Suh J, Romano D, Truong M, Mullin K, Hooli B, et al. Potential late-onset Alzheimer's disease-associated mutations in the ADAM10 gene attenuate {alpha}-secretase activity. Hum Mol Genet. 2009;18:3987-96 pubmed publisher
  66. Lupton M, Proitsi P, Danillidou M, Tsolaki M, Hamilton G, Wroe R, et al. Deep sequencing of the Nicastrin gene in pooled DNA, the identification of genetic variants that affect risk of Alzheimer's disease. PLoS ONE. 2011;6:e17298 pubmed publisher
  67. Lupton M, Proitsi P, Lin K, Hamilton G, Daniilidou M, Tsolaki M, et al. The role of ABCA1 gene sequence variants on risk of Alzheimer's disease. J Alzheimers Dis. 2014;38:897-906 pubmed publisher
  68. Keren Shaul H, Spinrad A, Weiner A, Matcovitch Natan O, Dvir Szternfeld R, Ulland T, et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer's Disease. Cell. 2017;169:1276-1290.e17 pubmed publisher
  69. Aikawa T, Ren Y, Yamazaki Y, Tachibana M, Johnson M, Anderson C, et al. ABCA7 haplodeficiency disturbs microglial immune responses in the mouse brain. Proc Natl Acad Sci U S A. 2019;: pubmed publisher
  70. Nicolas G, Acuna Hidalgo R, Keogh M, Quenez O, Steehouwer M, Lelieveld S, et al. Somatic variants in autosomal dominant genes are a rare cause of sporadic Alzheimer's disease. Alzheimers Dement. 2018;14:1632-1639 pubmed publisher
  71. Campion D, Charbonnier C, Nicolas G. SORL1 genetic variants and Alzheimer disease risk: a literature review and meta-analysis of sequencing data. Acta Neuropathol. 2019;: pubmed publisher
  72. Patel D, Mez J, Vardarajan B, Staley L, Chung J, Zhang X, et al. Association of Rare Coding Mutations With Alzheimer Disease and Other Dementias Among Adults of European Ancestry. JAMA Netw Open. 2019;2:e191350 pubmed publisher
  73. Ewers M, Franzmeier N, Suarez Calvet M, Morenas Rodríguez E, Caballero M, Kleinberger G, et al. Increased soluble TREM2 in cerebrospinal fluid is associated with reduced cognitive and clinical decline in Alzheimer's disease. Sci Transl Med. 2019;11: pubmed publisher
  74. Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, et al. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity. 2017;47:566-581.e9 pubmed publisher
  75. Maniatis S, Aijö T, Vicković S, Braine C, Kang K, Mollbrink A, et al. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science. 2019;364:89-93 pubmed publisher
  76. Molgora M, Esaulova E, Vermi W, Hou J, Chen Y, Luo J, et al. TREM2 Modulation Remodels the Tumor Myeloid Landscape Enhancing Anti-PD-1 Immunotherapy. Cell. 2020;182:886-900.e17 pubmed publisher
  77. Pluvinage J, Haney M, Smith B, Sun J, Iram T, Bonanno L, et al. CD22 blockade restores homeostatic microglial phagocytosis in ageing brains. Nature. 2019;568:187-192 pubmed publisher
  78. Jakobsdottir J, van der Lee S, Bis J, Chouraki V, Li Kroeger D, Yamamoto S, et al. Rare Functional Variant in TM2D3 is Associated with Late-Onset Alzheimer's Disease. PLoS Genet. 2016;12:e1006327 pubmed publisher
  79. Mozell M, Hornung D, Leopold D, Youngentob S. Initial mechanisms basic to olfactory perception. Am J Otolaryngol. 1983;4:238-45 pubmed
  80. Venegas C, Kumar S, Franklin B, Dierkes T, Brinkschulte R, Tejera D, et al. Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer's disease. Nature. 2017;552:355-361 pubmed publisher
  81. Kellogg E, Hejab N, Poepsel S, Downing K, DiMaio F, Nogales E. Near-atomic model of microtubule-tau interactions. Science. 2018;360:1242-1246 pubmed publisher
  82. Verheijen J, van der Zee J, Gijselinck I, Van den Bossche T, Dillen L, Heeman B, et al. Common and rare TBK1 variants in early-onset Alzheimer disease in a European cohort. Neurobiol Aging. 2018;62:245.e1-245.e7 pubmed publisher
ISSN : 2329-5139