IMR Press / JIN / Volume 21 / Issue 4 / DOI: 10.31083/j.jin2104105
Open Access Original Research
TREM2 coding variants in Slovak Alzheimer's disease patients
Show Less
1 Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, 81108 Bratislava, Slovakia
2 Medirex Group Academy, Ltd, 82016 Bratislava, Slovakia
3 Psychiatry Outpatient Clinic, University Hospital and Policlinic the Brothers of Saint John of God, 81106 Bratislava, Slovakia
4 Care Centre Centrum MEMORY, 85103 Bratislava, Slovakia
5 1st Department of Neurology, Faculty of Medicine, Comenius University in Bratislava and University Hospital, 81367 Bratislava, Slovakia
6 Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava and University Hospital, 81369 Bratislava, Slovakia
*Correspondence: vladimira.durmanova@fmed.uniba.sk (Vladimira Durmanova)
Academic Editor: Qiu-Lan Ma
J. Integr. Neurosci. 2022, 21(4), 105; https://doi.org/10.31083/j.jin2104105
Submitted: 13 December 2021 | Revised: 14 January 2022 | Accepted: 26 January 2022 | Published: 1 June 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Triggering receptor expressed on myeloid cells 2 (TREM2) is an important modulator of innate immune responses. In the human brain, TREM2 is primarily expressed on microglia and is involved in cell survival, phagocytosis, and regulation of inflammation. TREM2 dysfunction has been linked to the pathogenesis of various neurodegenerative diseases including Alzheimer’s disease (AD). Rare coding variants of the TREM2 gene have been reported to modulate AD risk in several populations, however, data on their association with susceptibility to AD in the Slovak population have been missing. Methods: We have analyzed 10 non-synonymous coding variants located in TREM2 exon 2 by direct sequencing in 270 late-onset Alzheimer’s disease (LOAD) patients and 331 controls. Results: Four out of 10 TREM2 mutant variants have been identified in the analyzed groups, namely rs75932628 C > T (R47H), rs142232675 C > T (D87N), rs143332484 C > T (R62H), and rs2234253 G > T (T96K). R47H was found only in the AD group, while T96K was present only in the controls. Although no significant association between TREM2 coding variants and LOAD susceptibility has been detected, the observed odds ratio (OR) of 3.69 for R47H carriers suggests an increased risk of LOAD for this variant in the Slovak population. Moreover, we also found a higher OR for the rs143332484-T allele in APOEε4 non-carriers (1.99) when compared to APOEε4 carriers (0.62). Conclusions: Our results suggest an impact of specific TREM2 rare coding variants on AD risk in the Slovak population.

Keywords
Alzheimer's disease
TREM2 variants
apolipoprotein E
case-control study
sequencing
1. Introduction

Triggering Receptor Expressed on Myeloid cells 2 (TREM2) is a pattern recognition receptor present on dendritic cells, monocytes, and tissue-specific macrophages [1, 2, 3]. This 230 amino acid long transmembrane glycoprotein consists of an extracellular immunoglobulin-like V type domain, a transmembrane domain, and a cytoplasmic tail [4]. Because of the short cytoplasmic tail, TREM2 acts through the intracellular adaptor molecule DNAX-activation protein 12 (DAP12), also known as TYRO protein tyrosine kinase binding protein (TYROBP) [5]. TREM2 binds to anionic lipids, high- and low-density lipoproteins, and to several apolipoproteins such as APOA1, APOA2, APOB, APOE, and APOJ (clusterin) [6, 7, 8, 9]. The molecule is implicated in a wide array of functions including cell maturation, survival, proliferation, activation, phagocytosis, and the regulation of inflammation [10]. Anti-inflammatory properties of TREM2 after TLR stimulation have been confirmed in several in vitro and in vivo studies [11, 12, 13].

In the human brain, TREM2 is primarily expressed on microglia and is involved in the phagocytosis of apoptotic neurons, and modulation of inflammation [10, 14, 15, 16]. As revealed in microglia, the reduction of TREM2 signaling increases TNF and NO synthase-2 (NOS2) transcription, while overexpression of TREM2 decreases transcription of TNF, IL1β, and NOS2 [14].

Enhanced expression of TREM2 was found in various neurodegenerative disorders such as Parkinson’s disease [17], amyotrophic lateral sclerosis [18], stroke [19, 20], traumatic brain injury [21], and Alzheimer’s disease [22, 23, 24, 25]. In Alzheimer’s disease (AD) subjects, increased TREM2 expression has been associated with the recruitment of microglia to amyloid plaques [24, 26]. In various in vitro and in vivo models, TREM2 has been associated with Aβ40 and Aβ42 uptake by microglia suggesting its effect on Aβ clearance in AD models [27, 28, 29, 30, 31]. TREM2 can also increase microglial cell number, proliferation, and survival resulting in clustering of microglia to amyloid plaques [27, 32, 33].

Several TREM2 genetic variants have been identified that increase the risk of late-onset Alzheimer’s disease (LOAD). The most-well studied variant is rs75932628 C > T, a single nucleotide polymorphism (SNP) in exon 2 encoding an arginine to histidine missense substitution at amino acid 47 (R47H). Two independent studies in 2013 found for the first time that heterozygous form of the TREM2 R47H is associated with LOAD risk [34, 35]. This rare TREM2 variant can increase AD risk by 2–4 fold which is comparable to the increased risk associated with one copy of the known APOEε4 risk allele [36, 37]. The association of the TREM2 R47H variant with AD has been observed in European [38, 39, 40, 41, 42, 43, 44, 45, 46], and Afro-American populations [47, 48]. However, the R47H variant does not seem to be associated with LOAD risk in the Asian population [49, 50, 51, 52, 53, 54]. Other TREM2 variants suggested as risk factors for developing LOAD include D87N, R62H, T96K, Y38C, L211P, and H157Y [34, 38, 47, 55, 56, 57, 58, 59, 60]. Most variants are associated with decreased cell-surface expression of TREM2 (R47H, Y38C, R136Q) or by impaired in vitro interactions with ligands like APOE, LDL, and clusterin (R47H, R62H and D87N) [27, 61, 62]. R47H carriers in AD patients also demonstrated upregulation of IFN type I response and pro-inflammatory cytokines [63]. Moreover, the R47H variant of sTREM2 is less able to bind and disaggregate oligomeric Aβ, which leads to Aβ protofibril formation and neurotoxicity [64].

The aim of our study was to perform an association analysis between TREM2 coding variants and risk of LOAD in the Slovak population. We have analyzed 10 non-synonymous rare variants in exon 2 of the TREM2 by means of direct sequencing. To our knowledge, no such analysis has been performed in the Slovak population until now.

2. Materials and methods
2.1 Study groups

The case-control study involved 270 late-onset AD patients (99 men and 171 women, mean age: 78.56 ± 6.18 years). The diagnosis of probably AD was established by physicians according to the National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and Related Disorders Association (NINCDS-ADRDA) diagnostic criteria [65]. The clinical examination included personal medical and family history assessment, neurologic examination, neuropsychological assessment, and neuroimaging. The clinicians estimated age at onset (AAO) using standardized methodology; it is the age, at which the patient started to show significant symptoms of memory loss and cognitive impairment with a progressive clinical course. Unrelated cases with early-onset AD (age at onset <65 years) and having other neurologic or psychiatric diseases were excluded from the study. All AD patients have been recruited in the period from 2016 until 2020 via several psychiatric clinics throughout Slovakia. The mean age at disease onset was 75.50 ± 6.38 years. The reference cohort in our case-control study comprised 331 unrelated volunteers (133 males and 198 females with mean age of 76.05 ± 7.64 years). Montreal Cognitive Assessment (MoCA) was selected as the screening test for cognitive impairment in this study [66]. The cut-off score of 26 from 30 has been considered for normal cognition. All control subjects were without any personal or family history of AD and they were recruited from a matched group of a larger population sample. All patients and controls were Caucasians of Slovak descent. APOEε4 allele as a known genetic risk factor for AD was evaluated in both study groups and implemented as a stratification factor in further analyses. Detailed parameters of the study groups are summarized in Table 1. This case-control study was approved by the Independent Ethical Committee of the Bratislava Municipality under the No. 05440/2021/HF. All the investigations were carried out in accordance with the International Ethical Guidelines and the Declaration of Helsinki. From all patients or their representatives, as well as from control subjects, informed written consent for enrolling in the study and for personal data management was obtained.

Table 1.Characteristics of the study groups.
Parameter LOAD patients (n = 270) Controls (n = 331) p value
Age at examination, y; mean × SD (age range) 78.56 × 6.18 (65‒95) 76.05 × 7.64 (65‒95) <0.0001
Age at onset, y; mean × SD (age range) 75.50 × 6.38 (65‒95) - -
Sex, n; female/male, (% female) 171/99 (63.33%) 198/133 (59.82%) 0.38
MoCA score; mean × SD 14.53 × 5.78 27.53 × 1.53 <0.0001
APOEε4 status, n; positive/negative (% positive) 134/136 (49.63%) 63/268 (19.03%) <0.0001
Differences in age and MoCA score between the two groups were examined by the Mann-Whitney unparametric test. Differences in sex and APOEε4 status were assessed using the Pearson χ2 test. p < 0.05 is considered statistically significant. CI, confidence interval; LOAD, late-onset Alzheimer’s disease; MoCA, Montreal Cognitive Assessment; n, number; OR, odds ratio; SD, standard deviation; y, years.
2.2 Sequence analysis

Genomic DNA was isolated from ethylenediamine tetraacetic acid (EDTA)-treated whole blood samples (2 mL) by a modified salting-out procedure [67]. TREM2 exon 2 region from 6472 to 7004 bp was amplified using forward primer 5-TCCTTCAGGGCAGGATTTTT-3 and reverse primer 5-AGTGGGTGGTTCTGCACAC-3. A 25 μL PCR reaction mixture contained 50 ng of template DNA, 0.2 mM of each dNTP (Thermo Fisher Scientific Inc., Lot. Nr. 00654321, MA, USA), 1 U of Taq DNA polymerase (Thermo Fisher Scientific Inc., Lot Nr. 1817560, MA, USA), 1.5 mmol MgCl2 (Thermo Fisher Scientific Inc., Lot Nr. 1817560, MA, USA), and 10 pmol of each specific primer. The PCR conditions consisted of initial denaturation at 94 °C for 3 min, followed 30 cycles of denaturation at 94 °C for 1 min, annealing at 56 °C for 30 s, and elongation at 72 °C for 1 min. The final elongation at 72 °C for 10 min completed the reaction. The PCR products were run in 1.0% agarose gel for 20 min and then visualized under UV light. A fragment size of 533 bp was confirmed using the 100 bp DNA ladder (Solis BioDyne Inc., Lot. Nr. 07110000, Estonia, Europe). To perform sequence analysis, the PCR products were purified using an EXO SAP-IT kit according to the manufacturer’s recommendations (USB Inc., Lot. Nr. 00123457, WI, USA) and then sequenced (approximately 100 ng) using BigDye Terminator v3.1 Cycle Sequencing Kit (Thermo Fisher Scientific Inc., Lot Nr. 00567001, MA, USA). Direct sequencing was performed individually on both strands using forward and reverse PCR primers. The sequencing reaction was run on a 3130 ABI PRISM Genetic analyzer (Applied Biosystems Inc., MA, USA) and the sequence data were analyzed by SeqScape software (version 3.0, Applied Biosystems Inc., MA, USA). The reference sequence was reported on the website http://www.ebi.ac.uk/imgt/hla/align.html.

Genotyping of the APOEε4 risk allele was performed by the determination of rs429358 (C > T) and rs7412 (T > C) polymorphisms in the fourth exon using direct sequencing as described previously [68].

2.3 Statistical analysis

Allele and genotype frequencies were determined by direct counting. Genotypes were tested for their fit to Hardy-Weinberg equilibrium using the chi-squared goodness-of-fit test. Statistical differences in allele and genotype frequencies between AD patients and the control group were evaluated by the Pearson chi-squared test using the InStat statistical software (version 3.10, GraphPad Software Inc., CA, USA). The p values, odds ratios (OR), and 95% confidence intervals (95% CI) were calculated in the codominant inheritance model. The multivariate logistic regression analysis adjusted for sex, age (age at onset in patients and age at the examination in controls), and APOEε4 carrier status as possible modifying factors was performed by the SNPstats web software available at http://bioinfo.iconcologia.net/SNPstats [69]. Regression analysis and synergy factor (SF) measurement were also performed to assess the significance and size of interaction between TREM2 variants and the APOEε4 allele, as previously described [70]. The p value of <0.05 was considered as statistically significant.

3. Results
3.1 Characteristics of the study groups

The demographic and clinical characteristics of the study groups are shown in Table 1. The study included 270 AD patients and 331 unrelated controls. There was no statistically significant difference between the AD group and controls in relation to gender (p = 0.38), with females having a higher prevalence in both AD patients (63.33%) and controls (59.82%). The mean age at examination was significantly higher in the AD group than in controls (78.56 versus 76.05 years; p < 0.0001). Concerning the MoCA assessment, there was a significantly lower MoCA score in AD patients compared to controls (14.53 versus 27.53; p < 0.0001). A significantly higher prevalence of the APOE ε4 risk allele was found in the AD group compared to controls (49.63% vs 19.03%, p < 0.0001).

3.2 Association of TREM2 variants with LOAD risk

Ten non-synonymous rare variants in TREM2 exon 2 have been analyzed in 270 AD patients and 331 controls: rs75932628 C > T (R47H), rs147564421 G > A (R98W), rs142232675 C > T (D87N), rs201258663 G > A (T66M), rs797044603 A > G (Y38C), rs104894002 G > A (Q33X), rs143332484 C > T (R62H), rs2234252 G > A (A28V), rs2234253 G > T (T96K), and rs374851046 C > G (R52H). Allele and genotype frequencies of the TREM2 variants observed in the analyzed groups are shown in Table 2. Four out of 10 TREM2 coding variants have been identified in the analyzed groups, namely R47H, D87N, R62H and T96K. Genotype frequencies of TREM2 variants fit the Hardy-Weinberg equilibrium in AD patients (p = 0.98 for R47H; p = 0.93 for D87N, p = 0.80 for R62H, p = NA for T96K) as well as in controls (p = NA for R47H, p = 0.87 for D87N, p = 0.82 for R62H, p = 0.98 for T96K).

Table 2.Association between TREM2 coding variants and LOAD risk.
TREM-2 LOAD (n = 270) Controls (n = 331) a vs A-crude analysis Aa vs AA-crude analysis Aa vs AA-adjusted analysis* Interaction p values
a frequency a frequency p OR (95% CI) p OR (95% CI) p OR (95% CI) TREM-2× APOE ε4
6671 C > T (R47H) 1 (0.19%) 0 (0.00%) 0.27 3.68 (0.15‒90.69) 0.21 3.69 (0.15‒91.02) 0.38 - NA
6823 G > A (R98W) 0 (0.00%) 0 (0.00%) NA NA NA NA NA NA NA
6790 C > T (D87N) 3 (0.56%) 6 (0.91%) 0.48 0.61 (0.15‒2.46) 0.48 0.61 (0.15-2.46) 0.62 0.69 (0.16‒3.03) 0.75/0.72
6728 G > A (T66M) 0 (0.00%) 0 (0.00%) NA NA NA NA NA NA NA
6645 A > G (Y38C) 0 (0.00%) 0 (0.00%) NA NA NA NA NA NA NA
6628 G > A (Q33X) 0 (0.00%) 0 (0.00%) NA NA NA NA NA NA NA
6716 C > T (R62H) 8 (1.48%) 7 (1.06%) 0.51 1.41 (0.51‒3.91) 0.51 1.41 (0.51‒3.95) 0.73 1.21 (0.41‒3.59) 0.27/0.32
6614 G > A (A28V) 0 (0.00%) 0 (0.00%) NA NA NA NA NA NA NA
6818 G > T (T96K) 0 (0.00%) 1 (0.15%) 0.37 0.41 (0.02‒10.04) 0.27 0.41 (0.02‒10.04) 0.31 - NA
6686 C > G (R52H) 0 (0.00%) 0 (0.00%) NA NA NA NA NA NA NA
*Logistic regression analysis adjusted for age (age at onset in patients and age at examination in controls), sex and APOE ε4 status. Crude/adjusted analysis (for age and sex) of interaction between TREM-2 mutant allele and APOE ε4 allele. p < 0.05 is considered statistically significant. -: OR values couldn’t be calculated by software program. a, mutant allele; A, wild-type allele; CI, confidence interval; LOAD, late-onset Alzheimer’s disease; n, number; NA, not available; OR, odds ratio.

The carriage of the rs75932628-T allele (R47H) was identified in one of the AD cases (0.19%), while this allele was missing in the control group. Univariate chi-square analysis revealed that the carrier of the minor T allele had a 3.69-fold increased risk to develop AD compared to non-T carriers. Regarding other TREM2 variants, the D87N variant was identified in 3 AD cases (0.56%) and 6 controls (0.91%), R62H was identified in 8 AD cases (1.48%) and 7 controls (1.06%) and T96K variant was identified in one control subject (0.15%). No statistically significant differences in either TREM2 mutant allele or genotype frequencies were found between the AD group and controls (p > 0.05, Table 2). Multivariate analysis of the association between the TREM2 variants and LOAD risk adjusted for age, sex and APOEε4 status as potential confounding variables revealed no changes in comparison with the univariate analysis (p > 0.05, Table 2). Logistic regression analysis also revealed no statistically significant p value for the interaction of TREM2 variants with APOE ε4 allele dosage (p > 0.05, Table 2).

3.3 Analysis of TREM2 variants in relation to APOE ε4 carrier status

Association between the TREM2 variants and LOAD risk in subjects stratified by APOE ε4 carrier status was also performed. Analyses in APOE ε4-positive and APOE ε4-negative groups revealed no statistically significant differences in the distribution of TREM2 variants between AD patients and the control group (p > 0.05 for R62H variant, Table 3). However, we found a somewhat higher odds ratio for rs143332484-T (R62H) in APOE ε4 non-carriers (1.99) than in APOE ε4 carriers (0.62).

Table 3.Association between TREM2 rs143332484 C > T (R62H) variant and LOAD risk in subjects stratified by APOE ε4 status.
LOAD Controls T vs C-crude analysis CT vs CC-crude analysis CT vs CC-adjusted analysis*
T frequency T frequency p OR (95% CI) p OR (95% CI) p OR (95% CI)
APOEε4 positive individuals 4 (1.49%) 3 (2.38%) 0.53 0.62 (0.14‒2.82) 0.54 0.62 (0.13‒2.84) 0.58 0.64 (0.14‒3.02)
APOEε4 negative individuals 4 (1.47%) 4 (0.75%) 0.33 1.99 (0.49‒8.00) 0.34 2.00 (0.49‒8.12) 0.34 1.99 (0.48‒8.21)
*Logistic regression analysis adjusted for age (age at onset in patients and age at examination in controls) and sex. p < 0.05 is considered statistically significant. CI, confidence interval; LOAD, late-onset Alzheimer’s disease; OR, odds ratio.

To further determine the genetic interaction between the APOE ε4 allele and the TREM2 variants, we assessed the combined gene effects of TREM2 rs143332484-T (R62H) and the APOE ε4 alleles on LOAD risk (Table 4). When compared to subjects without both alleles, carriers of rs143332484-T, but no APOEε4 were at two-fold increased odds of developing LOAD (p = 0.32), while the OR in subjects with at least one copy of APOEε4, but no TREM2 rs143332484-T (R62H) was 4.33 (p < 0.0001). In subjects with both alleles, the OR was reduced to 2.67 (p = 0.19). This observed combined effect size of the two alleles was markedly lower than the predicted joint OR assuming independent effects of both rs143332484-T and APOEε4 (OR = 8.66), however, the difference was not significant (p = 0.27). The calculated SF value of 0.31 indicates antagonism between TREM2 rs143332484-T (R62H) and APOEε4 alleles in LOAD risk (Table 4).

Table 4.Combined effects of TREM2 rs143332484-T (R62H) and APOEε4 allele carriage in LOAD risk.
TREM2 rs143332484-T APOE ε4 LOAD Controls p OR (95% CI) SF (p value)
132 (48.89%) 264 (79.76%) - reference 0.31 (0.27)
+ 4 (1.48%) 4 (1.21%) 0.32 2.00 (0.49‒8.13) -
+ 130 (48.15%) 60 (18.13%) <0.0001 4.33 (2.99‒6.28) -
+ + 4 (1.48%) 3 (0.91%) 0.19 2.67 (0.59‒12.09) -
Note: − = no copies of the allele; + = one or two copies of the allele. p, OR and 95% CI values were obtained by χ2 analysis. SF was calculated as follows: 2.67/(2.00 × 4.33). p < 0.05 is considered statistically significant. CI, confidence interval; LOAD, late-onset Alzheimer’s disease; OR, odds ratio; SF, synergy factor.
4. Discussion

TREM2 is a pattern recognition receptor expressed on myeloid cells involved in the modulation of the innate immune response [3]. In the human brain, TREM2 is primarily expressed on microglia, and is involved in cell survival, chemotaxis, phagocytosis, and regulation of inflammation [10, 14, 15, 16]. It was found that TREM2 can protect against AD by binding with Aβ40 and Aβ42 monomers [27, 28, 29, 30, 31, 33]. Moreover, the soluble form of TREM2 is involved in the inhibition of Aβ aggregation thus it prevents the formation of pathological amyloid plaques [64].

Rare coding variants in TREM2 were identified as risk factors for Alzheimer’s disease in several populations. In 2013 two independent studies described for the first time the association of R47H with LOAD risk [34, 35]. Since then the R47H variant has been consistently reported to increase the risk for AD across ethnicities as stated in meta-analyses [37, 46, 54, 59, 60] and large-scale GWAS analyses [71, 72].

In this study, we examined the contribution of TREM2 rare variants on risk for LOAD in the Slovak population. We have analyzed 10 non-synonymous coding variants located in TREM2 exon 2 encoding the ectodomain. Four out of 10 TREM2 mutant variants have been identified in analyzed groups, namely R47H, D87N, R62H and T96K. No statistically significant differences in the distribution of the TREM2 R47H variant were found between AD patients and the control group. This finding is due to the low frequency of the risk allele in the study population and agrees with other reports [39, 55]. In our study, the R47H variant was identified in one AD case (0.19%), but it was absent in the control group. The OR for R47H was 3.69 what is comparable with previous studies on this variant in Caucasian populations with pooled OR of 3.93 [37].

The distribution of R47H variant in AD patients seems to differ across ethnicities. The rs75932628-T was found in 0.6 to 1.4% of AD cases in the Spanish population [38, 40], 1.6 to 2.1% in the French population [73, 74], 0.74% of AD cases in the Belgian population [55], 0.4% in the Polish population [58], 1.4 to 2.5% in UK [34, 41, 42], 1.8% in the Icelandic population [43], 2.3% in the Iranian population [75], 1.8% to 6% in North Americans [56, 76, 77, 78], 0.2% in Afro-Americans [47], 1.7% in the Colombian population [48], 0.98% in the Argentinian population [59], and in 0.05% in the Japanese population [51].

Regarding other TREM2 variants in the Slovak population, D87N and R62H were found in both AD patients and controls, while T96K was identified in one control subject. No statistically significant differences in the distribution of TREM2 mutant variants were found between AD patients and the control group, as reported in other studies [34, 47, 55].

We are aware of several limitations of the current study. First, the relatively small sample sizes may reduce the power of the study to detect associations between TREM2 gene variants and the risk of late-onset Alzheimer’s disease. A larger number of LOAD patients would be desirable for replication, especially with regard to the low prevalence of most TREM variants. We assume our present-day results as useful for sample size planning in future investigations on this topic. Secondly, the estimation of age at onset may be biased by different factors such as the significance of clinical symptoms as experienced by patients.

A significant association of R47H with the risk of AD in the Caucasian population (OR = 3.93, 95% CI: 3.15–4.90, p < 0.001) was confirmed in the latest meta-analysis examining 22,175 AD cases and 33,049 controls [37]. Furthermore, meta-analysis of R62H and D87N in Caucasians identified no significant association of these TREM2 variants with AD susceptibility (R62H: OR = 1.17, 95% CI: 0.90–1.52, p = 0.231; D87N: OR = 1.62, 95% CI: 0.94–2.82, p = 0.084). However, a three-stage case-control study of 48,343 AD cases and 36,790 controls revealed significant association between the R62H variant and AD risk in Caucasians (p = 1.55 × 10-14, OR = 1.67) [79]. A most recent meta-analysis in Caucasians also indicated that H157Y carriers were more predisposed to AD (OR = 4.22, 95% CI: 1.93–9.21, p < 0.001) as compared to controls [37]. In contrast to the Caucasian population, the R47H variant has a lower frequency in the East Asian population (0.03% vs 0.08%) and thus no association of the R47H variant with AD risk has been identified [37, 52]. Similarly, no association of H157Y or L211P with AD in the East Asian population was found [52]. Concerning other TREM2 variants, S31F, R47C, G55R, L205P, and G219C were identified as novel variants in the AD patient group [56, 75, 77]. Moreover, TREM2 rs187370608 A > G was significantly associated with AD susceptibility as revealed in GWAS using combined AD-by-proxy approach and case-control study (p = 1.45 × 10-16, p = 1.26 × 10-25) [80, 81].

The diminished effect of TREM2 rare coding variants on protein function has been confirmed by biochemical analyses. The most studied R47H variant showed decreased cell-surface expression and impaired ligand-binding [9, 77]. R47H variant also affects TREM2 maturation [82]. Finally, the R47H variant of soluble TREM2 is less capable of binding and disaggregating oligomeric Aβ, thus promoting Aβ-induced neurotoxicity [64]. As a consequence of loss of TREM2 function, R47H carriers showed reduced myeloid cell responses to amyloid deposition and more frequent α-synucleinopathy [45, 83]. Some studies found that R47H carriers had a trend toward a decrease in the age of AD onset [41, 45], however, others revealed no significant association [40, 43].

Other TREM2 rare variants associated with decreased cell-surface receptor expression include R136Q, R136W Y38C, T66M, S31F, R47C, and E151K [77]. R62H and D87N showed impaired interactions with ligands, however, the T96K variant increased TREM2 affinity to their ligands [61, 84]. Y38C variant exhibited impaired TREM2 maturation and folding leading to changes in microglia morphology, loss of synaptic proteins, and reduced hippocampal synaptic plasticity in mouse models [85].

As APOE ε4 allele belongs to known risk factors for susceptibility to AD [36, 86, 87], we also examined the combined effect of APOEε4 and TREM2 variants on LOAD risk. Analyses in APOEε4-positive and APOEε4-negative groups revealed no statistically significant differences in the distribution of TREM2 variants between AD patients and the control group. However, we found a somewhat higher OR for rs143332484-T (R62H) in APOE ε4 non-carriers (1.99) than in APOE ε4 carriers (0.62). A recent meta-analysis in non-APOEε4 carriers based on whole-exome sequencing data identified 4 candidate variants with strong statistical power on AD risk. They include APOE/rs7412 coding for the APOEε2 allele, (OR = 0.40; p = 5.46 × 10-24), TOMM40/rs157581 (OR = 1.49; p = 4.04 × 10-7) and TREM2/rs75932628 (R47H) (OR = 4.00; p = 1.15 × 10-7). The fourth significant variant, NSF/rs199533 (K702K), was found on chromosome 17 (OR = 0.78; p = 2.88 × 10-7). These candidate gene variants showed either protective or negative effects on AD risk [88].

In our study we also calculated the synergy factor value that has predicted an antagonism between TREM2 rs143332484-T (R62H) and APOE ε4 alleles in LOAD risk. It is not clear whether the observed antagonistic interaction between TREM2 rs143332484-T (R62H) and APOE ε4 alleles correlates with the biological events in AD subjects. APOE ε4 has been associated with impaired binding and clearance of Aβ leading to deposition of amyloid plaques in the brain [89, 90]. Moreover, APOE ε4 has been involved in impaired synaptic integrity and plasticity [91]. Carriers of at least one APOEε4 allele exhibited an earlier disease onset, faster disease progression, and increased brain atrophy [36, 92, 93].

The APOE- TREM2 relationship has been studied by several authors [6, 7, 8, 94]. It was found that TREM2 is binding to APOE to enhance the phagocytosis of apoptotic neurons [6]. Regarding the TREM2 R62H variant its decreased ligand affinity has been observed [9]. It can be hypothesized that impaired ligand affinity of R62H affects phagocytosis of APOE-bound apoptotic cells by microglia contributing to AD pathology.

5. Conclusions

In our case-control study, we assessed the contribution of TREM2 rare variants on risk for LOAD in the Slovak population. We have analyzed 10 non-synonymous coding variants located in TREM2 exon 2 encoding for the extracellular domain. Four out of 10 TREM2 mutant variants have been identified in both analyzed groups, namely R47H, D87N, R62H, and T96K. R47H substitution was found only in the AD group, while T96K was present only in the controls. The OR of 3.69 in TREM2 R47H carriers suggests an increased risk of this variant for LOAD also in the Slovak population.

Abbreviations

AD, Alzheimer’s disease; APO, apolipoprotein; Aβ, amyloid β; DAP12, DNAX-activation protein 12; EDTA, ethylenediamine tetraacetic acid; GWAS, genome wide association studies; LDL, low density lipoproteins; LOAD, late-onset Alzheimer’s disease; LPS, lipopolysaccharide; MoCA, Montreal Cognitive Assessment; NA, not available; OR, odds ratio; SNP, single nucleotide polymorphism; sTREM2, soluble TREM2; TLR, toll-like receptor; TNF, tumor necrosis factor; TREM2, triggering receptor expressed on myeloid cells 2; TYROBP, TYRO protein tyrosine kinase binding protein.

Author contributions

VD and IS—study design and manuscript writing. ZP, BV and IK—sample collection. GM and RP— analysis by direct sequencing. JJ—data analysis and interpretation. VD and AO—PCR assays. SS—data collection.

Ethics approval and consent to participate

The study was approved by the Independent Ethical Committee of the Bratislava Municipality under the No. 05440/2021/HF. Informed written consent was obtained from all participants.

Acknowledgment

The authors thank all individuals for participating in this study. We gratefully acknowledge B. Misovic Faragova and Z. Nurnberger for their technical assistance.

Funding

This research was financially supported by the Scientific Grant Agency of Ministry of Education, Science, Research and Sport of Slovak Republic and Slovak Academy of Sciences (VEGA grant No. 1/0738/20).

Conflict of interest

The authors declare no conflict of interest.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References
[1]
Bouchon A, Hernández-Munain C, Cella M, Colonna M. A Dap12-Mediated Pathway Regulates Expression of Cc Chemokine Receptor 7 and Maturation of Human Dendritic Cells. Journal of Experimental Medicine. 2001; 194: 1111–1122.
[2]
Cella M, Buonsanti C, Strader C, Kondo T, Salmaggi A, Colonna M. Impaired differentiation of osteoclasts in TREM-2-deficient individuals. Journal of Experimental Medicine. 2003; 198: 645–651.
[3]
Colonna M. TREMs in the immune system and beyond. Nature Reviews Immunology. 2003; 3: 445–453.
[4]
Allcock RJN, Barrow AD, Forbes S, Beck S, Trowsdale J. The human TREM gene cluster at 6p21.1 encodes both activating and inhibitory single IgV domain receptors and includes NKp44. European Journal of Immunology. 2003; 33: 567–577.
[5]
Daws MR, Lanier LL, Seaman WE, Ryan JC. Cloning and characterization of a novel mouse myeloid DAP12-associated receptor family. European Journal of Immunology. 2001; 31: 783–791.
[6]
Atagi Y, Liu C, Painter MM, Chen X, Verbeeck C, Zheng H, et al. Apolipoprotein E is a Ligand for Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) Journal of Biological Chemistry. 2015; 290: 26043–26050.
[7]
Bailey CC, DeVaux LB, Farzan M. The Triggering Receptor Expressed on Myeloid Cells 2 Binds Apolipoprotein E. Journal of Biological Chemistry. 2016; 290: 26033–26042.
[8]
Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M. TREM2 Binds to Apolipoproteins, Including APOE and CLUAPOJ, and thereby Facilitates Uptake of Amyloid-Beta by Microglia. Neuron. 2016; 91: 328–340.
[9]
Kober DL, Brett TJ. TREM2-Ligand Interactions in Health and Disease. Journal of Molecular Biology. 2017; 429: 1607–1629.
[10]
Colonna M, Wang Y. TREM2 variants: new keys to decipher Alzheimer disease pathogenesis. Nature Reviews. Neuroscience. 2016; 17: 201–207.
[11]
Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. Journal of Immunology. 2006; 177: 2051–2055.
[12]
Gao X, Dong Y, Liu Z, Niu B. Silencing of triggering receptor expressed on myeloid cells-2 enhances the inflammatory responses of alveolar macrophages to lipopolysaccharide. Molecular Medicine Reports. 2014; 7: 921–926.
[13]
Zhong L, Chen X, Zhang Z, Wang Z, Shi X, Xu K, et al. DAP12 Stabilizes the C-terminal Fragment of the Triggering Receptor Expressed on Myeloid Cells-2 (TREM2) and Protects against LPS-induced Pro-inflammatory Response. Journal of Biological Chemistry. 2015; 290: 15866–15877.
[14]
Takahashi K, Rochford CDP, Neumann H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. Journal of Experimental Medicine. 2005; 201: 647–657.
[15]
Hsieh CL, Koike M, Spusta SC, Niemi EC, Yenari M, Nakamura MC, et al. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. Journal of Neurochemistry. 2009; 109: 1144–1156.
[16]
Liu W, Taso O, Wang R, Bayram S, Graham AC, Garcia-Reitboeck P, et al. TREM2 promotes anti-inflammatory responses in microglia and is suppressed under pro-inflammatory conditions. Human Molecular Genetics. 2020; 29: 3224–3248.
[17]
Liu G, Liu Y, Jiang Q, Jiang Y, Feng R, Zhang L, et al. Convergent Genetic and Expression Datasets Highlight TREM2 in Parkinson’s Disease Susceptibility. Molecular Neurobiology. 2018; 53: 4931–4938.
[18]
Cady J, Koval ED, Benitez BA, Zaidman C, Jockel-Balsarotti J, Allred P, et al. TREM2Variant p.R47H as a Risk Factor for Sporadic Amyotrophic Lateral Sclerosis. JAMA Neurology. 2014; 71: 449.
[19]
Kawabori M, Kacimi R, Kauppinen T, Calosing C, Kim JY, Hsieh CL, et al. Triggering receptor expressed on myeloid cells 2 (TREM2) deficiency attenuates phagocytic activities of microglia and exacerbates ischemic damage in experimental stroke. Journal of Neuroscience. 2015; 35: 3384–3396.
[20]
Backes FN, de Souza A, Bianchin MM. Biomarkers in the prognostic evaluation of ischemic stroke: is there benefit in the measurements of TREM-1 and TREM-2 in the acute phase? Clinical Biochemistry. 2021; 98: 10–16.
[21]
Saber M, Kokiko-Cochran O, Puntambekar SS, Lathia JD, Lamb BT. Triggering Receptor Expressed on Myeloid Cells 2 Deficiency Alters Acute Macrophage Distribution and Improves Recovery after Traumatic Brain Injury. Journal of Neurotrauma. 2018; 34: 423–435.
[22]
Celarain N, Sánchez-Ruiz de Gordoa J, Zelaya MV, Roldán M, Larumbe R, Pulido L, et al. TREM2 upregulation correlates with 5-hydroxymethycytosine enrichment in Alzheimer’s disease hippocampus. Clinical Epigenetics. 2016; 8: 37.
[23]
Ma L, Allen M, Sakae N, Ertekin-Taner N, Graff-Radford NR, Dickson DW, et al. Expression and processing analyses of wild type and p.R47H TREM2 variant in Alzheimer’s disease brains. Molecular Neurodegeneration. 2017; 11: 72.
[24]
Perez SE, Nadeem M, He B, Miguel JC, Malek-Ahmadi MH, Chen K, et al. Neocortical and hippocampal TREM2 protein levels during the progression of Alzheimer’s disease. Neurobiology of Aging. 2017; 54: 133–143.
[25]
Liu D, Cao B, Zhao Y, Huang H, McIntyre RS, Rosenblat JD, et al. Soluble TREM2 changes during the clinical course of Alzheimer’s disease: a meta-analysis. Neuroscience Letters. 2019; 686: 10–16.
[26]
Yeh FL, Hansen DV, Sheng M. TREM2, Microglia, and Neurodegenerative Diseases. Trends in Molecular Medicine. 2018; 23: 512–533.
[27]
Wang Y, Cella M, Mallinson K, Ulrich JD, Young KL, Robinette ML, et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell. 2015; 160: 1061–1071.
[28]
Yuan P, Condello C, Keene CD, Wang Y, Bird TD, Paul SM, et al. TREM2 Haplodeficiency in Mice and Humans Impairs the Microglia Barrier Function Leading to Decreased Amyloid Compaction and Severe Axonal Dystrophy. Neuron. 2017; 90: 724–739.
[29]
Zhao Y, Wu X, Li X, Jiang L, Gui X, Liu Y, et al. TREM2 is a Receptor for β-Amyloid that Mediates Microglial Function. Neuron. 2018; 97: 1023–1031.
[30]
Zhong L, Wang Z, Wang D, Wang Z, Martens YA, Wu L, et al. Amyloid-beta modulates microglial responses by binding to the triggering receptor expressed on myeloid cells 2 (TREM2) Molecular Neurodegeneration. 2018; 13: 15.
[31]
Kober DL, Stuchell-Brereton MD, Kluender CE, Dean HB, Strickland MR, Steinberg DF, et al. Functional insights from biophysical study of TREM2 interactions with apoE and Aβ1-42. To be published in Alzheimer’s & Dementia. 2020. [Preprint].
[32]
Zheng H, Jia L, Liu C, Rong Z, Zhong L, Yang L, et al. TREM2 Promotes Microglial Survival by Activating Wnt/β-Catenin Pathway. Journal of Neuroscience. 2017; 37: 1772–1784.
[33]
Wang Y, Ulland TK, Ulrich JD, Song W, Tzaferis JA, Hole JT, et al. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. Journal of Experimental Medicine. 2016; 213: 667–675.
[34]
Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, et al. TREM2 variants in Alzheimer’s disease. New England Journal of Medicine. 2013; 368: 117–127.
[35]
Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. New England Journal of Medicine. 2013; 368: 107–116.
[36]
Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, 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.
[37]
Li R, Wang X, He P. The most prevalent rare coding variants of TREM2 conferring risk of Alzheimer’s disease: A systematic review and meta-analysis. Experimental and Therapeutic Medicine. 2021; 21: 347.
[38]
Benitez BA, Cooper B, Pastor P, Jin S, Lorenzo E, Cervantes S, et al. TREM2 is associated with the risk of Alzheimer’s disease in Spanish population. Neurobiology of Aging. 2013; 34: e15–e17.
[39]
Hooli BV, Parrado AR, Mullin K, Yip W, Liu T, Roehr JT, et al. The rare TREM2 R47H variant exerts only a modest effect on Alzheimer disease risk. Neurology. 2014; 83: 1353–1358.
[40]
Ruiz A, Dols-Icardo O, Bullido MJ, Pastor P, Rodríguez-Rodríguez E, López de Munain A, et al. Assessing the role of the TREM2 p.R47H variant as a risk factor for Alzheimer’s disease and frontotemporal dementia. Neurobiology of Aging. 2014; 35: e1–e4.
[41]
Slattery CF, Beck JA, Harper L, Adamson G, Abdi Z, Uphill J, et al. R47H TREM2 variant increases risk of typical early-onset Alzheimer’s disease but not of prion or frontotemporal dementia. Alzheimer’s & Dementia. 2014; 10: 602–608.
[42]
Finelli D, Rollinson S, Harris J, Jones M, Richardson A, Gerhard A, et al. TREM2 analysis and increased risk of Alzheimer’s disease. Neurobiology of Aging. 2015; 36: e9–e13.
[43]
Rosenthal SL, Bamne MN, Wang X, Berman S, Snitz BE, Klunk WE, et al. More evidence for association of a rare TREM2 mutation (R47H) with Alzheimer’s disease risk. Neurobiology of Aging. 2016; 36: e21–e26.
[44]
Lill CM, Rengmark A, Pihlstrøm L, Fogh I, Shatunov A, Sleiman PM, et al. The role of TREM2 R47H as a risk factor for Alzheimer’s disease, frontotemporal lobar degeneration, amyotrophic lateral sclerosis, and Parkinson’s disease. Alzheimer’s & Dementia. 2016; 11: 1407–1416.
[45]
Korvatska O, Leverenz JB, Jayadev S, McMillan P, Kurtz I, Guo X, et al. R47H Variant of TREM2 Associated with Alzheimer Disease in a Large Late-Onset Family: Clinical, Genetic, and Neuropathological Study. JAMA Neurology. 2015; 72: 920–927.
[46]
Lu Y, Liu W, Wang X. TREM2 variants and risk of Alzheimer’s disease: a meta-analysis. Neurological Sciences. 2015; 36: 1881–1888.
[47]
Jin SC, Carrasquillo MM, Benitez BA, Skorupa T, Carrell D, Patel D, et al. TREM2 is associated with increased risk for Alzheimer’s disease in African Americans. Molecular Neurodegeneration. 2015; 10: 19.
[48]
Arboleda-Bustos CE, Ortega-Rojas J, Mahecha MF, Arboleda G, Vásquez R, Pardo R, et al. The p.R47H Variant of TREM2 Gene is Associated with Late-onset Alzheimer Disease in Colombian Population. Alzheimer Disease and Associated Disorders. 2019; 32: 305–308.
[49]
Yu J, Jiang T, Wang Y, Wang H, Zhang W, Hu N, et al. Triggering receptor expressed on myeloid cells 2 variant is rare in late-onset Alzheimer’s disease in Han Chinese individuals. Neurobiology of Aging. 2014; 35: e1–e3.
[50]
Ma J, Zhou Y, Xu J, Liu X, Wang Y, Deng Y, et al. Association study of TREM2 polymorphism rs75932628 with late-onset Alzheimer’s disease in Chinese Han population. Neurological Research. 2015; 36: 894–896.
[51]
Miyashita A, Wen Y, Kitamura N, Matsubara E, Kawarabayashi T, Shoji M, et al. Lack of genetic association between TREM2 and late-onset Alzheimer’s disease in a Japanese population. Journal of Alzheimer’s Disease. 2015; 41: 1031–1038.
[52]
Huang M, Wang D, Xu Z, Xu Y, Xu X, Ma Y, et al. Lack of Genetic Association between TREM2 and Alzheimer’s Disease in East Asian Population. American Journal of Alzheimer’s Disease & other Dementiasr. 2015; 30: 541–546.
[53]
Wang P, Guo Q, Zhou Y, Chen K, Xu Y, Ding D, et al. Lack of association between triggering receptor expressed on myeloid cells 2 polymorphism rs75932628 and late-onset Alzheimer’s disease in a Chinese Han population. Psychiatric Genetics. 2018; 28: 16–18.
[54]
Zhang B, Li R, Zhang Y, Gao X. Differential role of triggering receptors expressed on myeloid cells 2 R47H in 3 neurodegenerative diseases based on a systematic review and meta-analysis. Medicine. 2020; 99: e18921.
[55]
Cuyvers E, Bettens K, Philtjens S, Van Langenhove T, Gijselinck I, van der Zee J, et al. Investigating the role of rare heterozygous TREM2 variants in Alzheimer’s disease and frontotemporal dementia. Neurobiology of Aging. 2014; 35: e11–e19.
[56]
Ghani M, Sato C, Kakhki EG, Gibbs JR, Traynor B, St George-Hyslop P, et al. Mutation analysis of the MS4a and TREM gene clusters in a case-control Alzheimer’s disease data set. Neurobiology of Aging. 2017; 42: e7–e13.
[57]
Jiang T, Hou J, Gao Q, Yu J, Zhou J, Zhao H, et al. TREM2 p.H157Y Variant and the Risk of Alzheimer’s Disease: a Meta-Analysis Involving 14,510 Subjects. Current Neurovascular Research. 2016; 13: 318–320.
[58]
Peplonska B, Berdynski M, Mandecka M, Barczak A, Kuzma-Kozakiewicz M, Barcikowska M, et al. TREM2 variants in neurodegenerative disorders in the Polish population. Homozygosity and compound heterozygosity in FTD patients. Amyotrophic Lateral Sclerosis & Frontotemporal Degeneration. 2019; 19: 407–412.
[59]
Dalmasso MC, Brusco LI, Olivar N, Muchnik C, Hanses C, Milz E, et al. Transethnic meta-analysis of rare coding variants in PLCG2, ABI3, and TREM2 supports their general contribution to Alzheimer’s disease. Translational Psychiatry. 2019; 9: 55.
[60]
Zhou S, Tan C, Hou X, Cao X, Tan L, Yu J. TREM2 Variants and Neurodegenerative Diseases: a Systematic Review and Meta-Analysis. Journal of Alzheimer’s Disease. 2019; 68: 1171–1184.
[61]
Kober DL, Alexander-Brett JM, Karch CM, Cruchaga C, Colonna M, Holtzman MJ, et al. Neurodegenerative disease mutations in TREM2 reveal a functional surface and distinct loss-of-function mechanisms. ELife. 2016; 5: e20391.
[62]
Song WM, Joshita S, Zhou Y, Ulland TK, Gilfillan S, Colonna M. Humanized TREM2 mice reveal microglia-intrinsic and -extrinsic effects of R47H polymorphism. Journal of Experimental Medicine. 2018; 215: 745–760.
[63]
Korvatska O, Kiianitsa K, Ratushny A, Matsushita M, Beeman N, Chien WM, et al. Triggering Receptor Expressed on Myeloid Cell 2 R47H Exacerbates Immune Response in Alzheimer’s Disease Brain. Frontiers in Immunology. 2020; 11: 559342.
[64]
Vilalta A, Zhou Y, Sevalle J, Griffin JK, Satoh K, Allendorf DH, et al. Wild-type s TREM2 blocks aβ aggregation and neurotoxicity, but the Alzheimer’s R47H mutant increases aβ aggregation. Journal of Biological Chemistry. 2021; 296: 100631.
[65]
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: 939–939.
[66]
Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. Journal of the American Geriatrics Society. 2005; 53: 695–699.
[67]
Miller SA, Dykes DD, Polesky HF. A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Research. 1988; 16: 1215.
[68]
Durmanova V, Parnicka Z, Javor J, Minarik G, Vrazda L, Vaseckova B, et al. A Novel Association of Polymorphism in the ITGA4 Gene Encoding the VLA-4 α4 Subunit with Increased Risk of Alzheimer’s Disease. Mediators of Inflammation. 2018; 2018: 7623823.
[69]
Solé X, Guinó E, Valls J, Iniesta R, Moreno V. SNPStats: a web tool for the analysis of association studies. Bioinformatics. 2006; 22: 1928–1929.
[70]
Cortina-Borja M, Smith AD, Combarros O, Lehmann DJ. The synergy factor: a statistic to measure interactions in complex diseases. BMC Research Notes. 2009; 2: 105.
[71]
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 Aβ, tau, immunity and lipid processing. Nature Genetics. 2019; 51: 414–430.
[72]
de Rojas I, Moreno-Grau S, Tesi N, Grenier-Boley B, Andrade V, Jansen IE, et al. Common variants in Alzheimer’s disease and risk stratification by polygenic risk scores. Nature Communications. 2021; 12: 3417.
[73]
Pottier C, Wallon D, Rousseau S, Rovelet-Lecrux A, Richard A, Rollin-Sillaire A, et al. TREM2 R47H variant as a risk factor for early-onset Alzheimer’s disease. Journal of Alzheimer’s Disease. 2013; 35: 45–49.
[74]
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. Neurobiology of Aging. 2018; 59: e1–e9.
[75]
Mehrjoo Z, Najmabadi A, Abedini SS, Mohseni M, Kamali K, Najmabadi H, et al. Association Study of the TREM2 Gene and Identification of a Novel Variant in Exon 2 in Iranian Patients with Late-Onset Alzheimer’s Disease. Medical Principles and Practice. 2016; 24: 351–354.
[76]
Roussos P, Katsel P, Fam P, Tan W, Purohit DP, Haroutunian V. The triggering receptor expressed on myeloid cells 2 (TREM2) is associated with enhanced inflammation, neuropathological lesions and increased risk for Alzheimer’s dementia. Alzheimer’s & Dementia. 2016; 11: 1163–1170.
[77]
Sirkis DW, Bonham LW, Aparicio RE, Geier EG, Ramos EM, Wang Q, et al. Rare TREM2 variants associated with Alzheimer’s disease display reduced cell surface expression. Acta Neuropathologica Communications. 2017; 4: 98.
[78]
Ayer AH, Wojta K, Ramos EM, Dokuru D, Chen JA, Karydas AM, et al. Frequency of the TREM2 R47H Variant in Various Neurodegenerative Disorders. Alzheimer Disease & Associated Disorders. 2019; 33: 327–330.
[79]
Sims R, van der Lee SJ, Naj AC, 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. Nature Genetics. 2017; 49: 1373–1384.
[80]
Jansen IE, Savage JE, Watanabe K, Bryois J, Williams DM, Steinberg S, et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nature Genetics. 2019; 51: 404–413.
[81]
Wightman DP, Jansen IE, Savage JE, Shadrin AA, Bahrami S, Holland D, et al. A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer’s disease. Nature Genetics. 2021; 53: 1276–1282.
[82]
Kleinberger G, Yamanishi Y, Suárez-Calvet M, Czirr E, Lohmann E, Cuyvers E, et al. TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Science Translational Medicine. 2014; 6: 243ra86.
[83]
Cheng-Hathaway PJ, Reed-Geaghan EG, Jay TR, Casali BT, Bemiller SM, Puntambekar SS, et al. The TREM2 R47H variant confers loss-of-function-like phenotypes in Alzheimer’s disease. Molecular Neurodegeneration. 2018; 13: 29.
[84]
Song W, Hooli B, Mullin K, Jin SC, Cella M, Ulland TK, et al. Alzheimer’s disease-associated TREM2 variants exhibit either decreased or increased ligand-dependent activation. Alzheimer’s & Dementia. 2017; 13: 381–387.
[85]
Jadhav VS, Lin PBC, Pennington T, Di Prisco GV, Jannu AJ, Xu G, et al. TREM2 Y38C mutation and loss of TREM2 impairs neuronal synapses in adult mice. Molecular Neurodegeneration. 2020; 15: 62.
[86]
Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, 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. Journal of the American Medical Association. 1997; 278: 1349–1356.
[87]
Crean S, Ward A, Mercaldi CJ, Collins JM, Cook MN, Baker NL, et al. Apolipoprotein E ε4 prevalence in Alzheimer’s disease patients varies across global populations: a systematic literature review and meta-analysis. Dementia and Geriatric Cognitive Disorders. 2011; 31: 20–30.
[88]
Fan K, Feingold E, Rosenthal SL, Demirci FY, Ganguli M, Lopez OL, et al. Whole-Exome Sequencing Analysis of Alzheimer’s Disease in Non-APOE*4 Carriers. Journal of Alzheimer’s Disease. 2020; 76: 1553–1565.
[89]
Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, et al. Human apoE isoforms differentially regulate brain amyloid-β peptide clearance. Science Translational Medicine. 2011; 3: 89ra57.
[90]
Mahoney-Sanchez L, Belaidi AA, Bush AI, Ayton S. The Complex Role of Apolipoprotein E in Alzheimer’s Disease: an Overview and Update. Journal of Molecular Neuroscience. 2016; 60: 325–335.
[91]
Love S, Siew LK, Dawbarn D, Wilcock GK, Ben-Shlomo Y, Allen SJ. Premorbid effects of APOE on synaptic proteins in human temporal neocortex. Neurobiology of Aging. 2006; 27: 797–803.
[92]
Cosentino S, Scarmeas N, Helzner E, Glymour MM, Brandt J, Albert M, et al. APOE epsilon 4 allele predicts faster cognitive decline in mild Alzheimer disease. Neurology. 2008; 70: 1842–1849.
[93]
Agosta F, Vossel KA, Miller BL, Migliaccio R, Bonasera SJ, Filippi M, et al. Apolipoprotein E epsilon4 is associated with disease-specific effects on brain atrophy in Alzheimer’s disease and frontotemporal dementia. Proceedings of the National Academy of Sciences of the United States of America. 2009; 106: 2018–2022.
[94]
Wolfe CM, Fitz NF, Nam KN, Lefterov I, Koldamova R. The Role of APOE and TREM2 in Alzheimer’s Disease-Current Understanding and Perspectives. International Journal of Molecular Sciences. 2018; 20: 81.
Share
Back to top