Skip to main content
Erschienen in: BMC Neurology 1/2019

Open Access 01.12.2019 | Case report

A pathogenic PSEN1 Trp165Cys mutation associated with early-onset Alzheimer’s disease

verfasst von: Vo Van Giau, Jung-Min Pyun, Jeewon Suh, Eva Bagyinszky, Seong Soo A. An, Sang Yun Kim

Erschienen in: BMC Neurology | Ausgabe 1/2019

Abstract

Background

Presenilin-1 (PSEN1) is one of the causative genes for early onset Alzheimer’s disease (EOAD). Recently, emerging studies reported several novel PSEN1 mutations among Asian. We describe a male with EOAD had a pathogenic PSEN1 mutation.

Case presentation

A 53-year-old male presented with memory decline, followed by difficulty in finding ways. Patient had positive family history, since his mother and one of his brother was also affected with dementia. Brain magnetic resonance imaging (MRI) scan showed mild degree of atrophy of bilateral hippocampus and parietal lobe. 18F-Florbetaben-PET (FBB-PET) revealed increased amyloid deposition in bilateral frontal, parietal, temporal lobe and precuneus. Whole exome analysis revealed a heterozygous, probably pathogenic PSEN1 (c.695G > T, p.W165C) mutation. Interestingly, Trp165Cys mutation is located in trans membrane (TM)-III region, which is conserved between PSEN1/PSEN2. In vitro studies revealed that PSEN1 Trp165Cys could result in disturbances in amyloid metabolism. This prediction was confirmed by structure predictions and previous in vitro studies that the p.Trp165Cys could result in decreased Aβ42/Aβ40 ratios.

Conclusion

We report a case of EOAD having a pathogenic PSEN1 (Trp165Cys) confirmed with in silico and in vitro predictions.
Hinweise
Vo Van Giau and Jung-Min Pyun contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AD
Alzheimer’s disease
APP
Amyloid precursor protein
β-amyloid
CDR
clinical dementia rating scale
EOAD
Early-onset Alzheimer’s disease
FDG-PET
Fludeoxyglucose- positron emission tomography
KRGDB
Korean Reference Genome Database
MMSE
Minimental state examination
MRI
Magnetic resonance imaging
PolyPhen2
Polymorphism phenotyping v2
PS1
Presenilin-1
PSEN1
Presenilin 1
PSEN2
Presenilin 2
SIFT
Sorting intolerant from tolerant
TM-III
Transmembrane segment III
WES
Whole exome sequencing

Background

Alzheimer’s disease (AD) (MIM #104300) is a neurodegenerative disease among elderlies, which is resulted by abnormal protein assembly inside the brain. Extracellular and intracellular amyloid beta (Aβ) and Tau protein, respectively, were associated as the main hallmarks of the disease. Early onset AD (EOAD) and late onset AD (LOAD) are the two main forms of the disease. Three genes were verified as causative factor for EOAD: amyloid precursor protein (APP) (MIM #104760) [1], presenilin 1 (PSEN1) (MIM #104311) [2], and presenilin 2 (PSEN2) (MIM #600759) [3]. Approximately 300 mutations of PSEN1, PSEN2, and APP in 635 affected individuals or families have been reported in the Dementia Mutation Database [4] (https://​www.​alzforum.​org/​mutations). Majority of mutations were observed in PSEN1 [57] (n = 219, 76.6%) with over 230 mutations reported as pathogenic in the Alzforum database (https://​www.​alzforum.​org/​mutations/​psen-1), as compared to APP (n = 51, 17.8%), and PSEN2 (n = 16, 5.6%) [811].
PSEN1 protein contains nine transmembrane (TM) domains, connected with hydrophilic loop regions. As the member of γ- secretase complex, PSEN1 could function as a catalytic subunit of aspartyl protease, involved in the cleavage of C99 residue in APP protein into β-amyloid (Aβ) peptide. PSEN1 mutations may impair the γ-secretase processing, resulting in altered of Aβ production. Gain-of-function mutations could increase the amyoid processing and the ratio Aβ42/Aβ40 [12, 13]. Loss- of- function mutations may reduce protective mechanisms, such as α-secretase cleavage [14]. In addition, accumulation of amyloid peptides may also associated with the reduced Aβ42 clearance [15] and neuronal loss [1517]. In majority of patients, disease occurred at 40–50 years of age [1821]. Several cases of young onset AD have been reported, where patients were less than 30–40 years of age [6, 7, 2231]. Guerrio et al. (2010) designed an algorithm for variants in EOAD causative genes [32] on prediction of the pathogenic nature of novel mutations. Investigating patients carrying novel as well as previously known mutations along with the associated phenotypes will aid in classification of these variants and may eventually support genetic counseling [33]. In this study, we reported a pathogenic PSEN1 W165C mutation as determined by genetic testing in a Korean patient with EOAD.

Case presentation

A 53-year-old man with 13 years of education presented with progressive memory decline. At aged 50, he complained forgetfulness of meetings or details of story, and repeating the same questions. He had difficulty in orienting to date and in finding way to a new place. His past medical history revealed myocardial infarction with proper medical treatment. His Korean version of Mini-Mental Status Examination (K-MMSE) score was 21/30 and clinical dementia rating scale (CDR) score was 0.5 at three years after symptom onset. And follow-up K-MMSE score at six years after symptom onset was 19/30 and CDR score was 1. His brain MRI at three years after symptom onset revealed mild atrophy of bilateral hippocampus and parietal lobe (Fig. 1a). FBB-PET at five years after symptom onset showed increased amyloid deposition in bilateral parietal, frontal, temporal lobe and precuneus (Fig. 1b). The patient had an APOE ε2 /ε3 polymorphism.
A 53-year-old man (III-1, Fig. 2) visited the Seoul National University Bundang Hospital with gradually impaired cognitive function over the previous years. The proband’s family history had a strong family history of dementia, and presented several family members affected by EOAD (Fig. 2). His mother (II-2) suffered from AD with onset in her fifties and deceased. The patient was one of the 4 siblings, comprising 2 brothers and 2 sisters. His first younger brother (III-2) was also diagnosed AD deceased in his forties, and had 2 children. His second younger brother (III-5) and his two younger sisters (III-3, III-4) displayed normal cognitive function. The health condition of the rest of his family members remained unknown, since all living family members and relatives declined to provide any additional information regarding their health.

Genetic analysis of PSEN1 and structural prediction of mutant PSEN 1 protein

Methods

An in depth genetic screen was performed using a specifically expanded panel of 50 causative and risk factor genes for various neurodegenerative disorders [34]. Whole exome sequencing (WES) was performed in Novogene. Standard Sanger sequencing was carried out by BioNeer Inc. (Dajeon, Republic of Korea) [34]. Big Dye Terminator Cyclic sequencing was performed using the ABI 3730XL DNA Analyzer (Bioneer Inc., Dajeon, Republic of Korea). Sequencing data was analyzed using NCBI Blast (http://​blast.​ncbi.​nlm.​nih.​gov/​Blast.​cgi) and the chromatograms were screened using the DNA BASER (http://​www.​dnabaser.​com) tool. Possible novel mutations were checked in the Korean Reference Genome Database (KRGDB; http://​nih.​go.​kr/​menu.​es?​mid=​a50303020300), which was obtained by whole genome sequencing of 622 healthy Korean individuals. The mutations were also screened against Broad Institute’s Genome Association Database (genome AD, http://​gnomad.​broadinstitute.​org) and 1000 Genomes (http://​www.​1000genomes.​org/​) databases.
The possible pathogenic nature of missense variants was predicted using simple online tools, such as PolyPhen-2 (http://​genetics.​bwh.​harvard.​edu/​pph2), Sorting Intolerant from Tolerant (SIFT; http://​sift.​jcvi.​org/​), and PROVEAN (http://​provean.​jcvi.​org) algorithms. ExPasy analysis was also performed (https://​www.​expasy.​org/​) using different parameters, such as Kyte and Doolittle hydrophobicity index, bulkiness, and polarity. Mutant and normal prion protein structures were compared by 3D modelling. Protein structures were built using the Raptor X web server (http://​raptorx.​uchicago.​edu/​), while Discovery Studio 3.5 Visualizer (Accelrys, San Diego, USA) was used to display the 3D images [35].

Results

A heterozygous G > T substitution (chr14; g.73653575: G > T) was discovered and confirmed to occur in the PSEN1 coding region using both WES and standard sequencing. This mutation caused the change from tryptophan to cysteine (c.495G > C; p.Trp165Cys) substitution, located at exon 6 of PSEN1 gene, and in transmembrane (TM) helix domain-III of the PSEN1 protein (Fig. 3).
The mutation is associated with EOAD patient with memory decline, followed by difficulty in finding ways and had a strong family history of AD; however, the specific mechanism is not functionally uncovered. The mutation was found in subject a Korean patient with EOAD and not observed in KRGDB, ExAC, and 1000genome control data sets. All in silico pathogenicity programs predicted the mutation to be deleterious. Figure 4 predicted that abnormal conformation inside the helix results in the mutated form due to an abnormal intra-or intermolecular disulfide bridge associated with the potential re-activities with metals or other compounds with thiol groups. Additionally, cysteine is not common recorded in the helix, further suggesting that this mutation might lead to abnormal conformation within the TM region. The intramolecular interactions may also change with the mutation: Trp165 has strong interaction with Ser169 (two hydrogen bonds), and forms another hydrogen bond with Val161. Cys165 changes the hydrogen bonds significantly. The interaction with Ser169 and Val 169 remained, but with Ser169, only one hydrogen bond was visible. Two new interactions could be seen with Ile162 and Ile168 (Fig. 5) In addition, the mutation is localized to trans membrane-III region conserved between PSEN1/PSEN2 and expected to affect Ab42 levels. This hypothesis was previously demonstrated that PSEN1 W165C led to increase Aβ42 and decreased Aβ40, resulting in elevated Aβ42/Aβ40 ratio in gaining loss of function in presenilin [3639].

Discussions

Since four different populations have been previously described the PSEN1 Trp165 from four familial AD cases (Table 1), the 165 codon seems to be a very vulnerable site. Initially, PSEN1 Trp165Cys mutation was found in a French family, with a codon combination of TGG > TGC. Mutation was associated with strongly was positive family history, since several affected family members were identified in three generations. Age of onset ranged between 37 and 47 years in the relatives with disease. No details were available on clinical symptoms of affected patients [36]. Second case of Trp165Cys was discovered with alternative codon exchange of TGG > TGT in an Indian family. Affected patients developed disease in their 40s, and disease phenotypes were rapid progressive disease progression and cerebral/cerebellar atrophies [37]. Our case was associated with probable EOAD case in a male patient, and his family members presented AD in their 40s. It may be difficult to find out, whether there could be a common founder between the Indian and Korean families. Since the Korea and India may be geographically isolated from each other, we suggest that PSEN1 Trp165Cys occur independently in these two families.
Table 1
Clinical findings in the published at codon 165 of PSEN1
 
Campion et al., 1999 [36] Wallon et al., 2012 [38]
Higuchi et al., 2000 [39]
Syama et al., 2018 [37]
This study
Country
France
Japan
India
Korea
Mutation
Trp165Cys
Trp165Gly
Trp165Cys
Trp165Cys
Codon change
TGG to TGC
TGG to GGG
TGG to TGT
TGG to TGT
Familiar
Yes
3 generation
Yes
Yes
Yes
Two generations
Age at onset (year)
37–47
34–38
45
50
ApoE genotype
ε3/ε3
NA
NA
ε2/ε3
Clinical signs and symptoms
EOAD
EOAD
NA
Memory decline, followed by difficulty in finding ways and had a strong family history of dementia
Brain Imaging (MRI, CT)
NA
NA
MRI: indicated diffuse cerebral and cerebellar atrophy in one case.
18F-Florbetaben-PET (FBB-PET): increased amyloid deposition in bilateral frontal, parietal, temporal lobe and precuneus
Functional data
↑Aβ42 and ↓Aβ40 in vitro; elevated Aβ42/Aβ40 ratio
No functional data
↑Aβ42 and ↓Aβ40 production in vitro; elevated Aβ42/Aβ40 ratio
↑Aβ42 and ↓Aβ40 production in vitro; elevated Aβ42/Aβ40 ratio.
PSEN1 Trp165Cys is located in the TM-III region of PSEN1 protein. An exchange from native amino acid to Cys may increase the risk of an abnormal intramolecular disulfide bond formation with another Cys. These new S-S bonds may create novel inter- or intramolecular structures, involving in pathogenic mechanisms. The potential mechanism was displayed and revealed the Fig. 5. In addition, at the residue, another mutation to glycine (Gly, G) was previously documented in a Japanese family with young onset AD [39], suggesting that this residue may be critical for PSEN1 function. Interestingly, the mutation is located to TM-III region conserved between PSEN1/PSEN2 and expected to affect Ab42 levels. This prediction was previously demonstrated that PSEN1 Trp165Cys resulted in increased Aβ42 and decreased Aβ40, respectively. It could lead to elevated Aβ42/Aβ40 ratio in gaining loss of function in presenilin [3639], verifying as a pathogenic mutation, involved in EOAD. This mutation was associated with rapid progression of disease, since the duration from the first clinical symptoms to the death ranged 4–10 years. Earlier onset of disease (37–47 years) was observed in a French family, but there was no information on the clinical symptoms in this family (Table 1).
Several pathogenic PSEN genes are found to cluster within the predicted α-helical TMs. Among other TMs, TM3 has been identified as one of the critical site in PSEN1, where several familiar AD (FAD) mutations were found. Recently, more than twenty mutations associated with FAD have been reported in TM-III of PSEN1 [36, 4050] (Fig. 6), and several of them were associated with EOAD (Table 2). Among these mutations, the PSEN1 Trp165Cys mutation is of particular interest because it may be associated with disease early onset. Moreover, the Trp165Cys mutation could cause increase in the Aβ42/Aβ total ratio [3639], similarly to other FAD-associated PSEN1 H163R [51], H163Y [52], L166P [53], I167del [54], S170F [31] S170P [40], L174del [44], L173 W [36] and L174 M [45] mutations.
Table 2
Comparison of PSEN1 Trp165Cys with all mutations, located in membrane associated TM-III domain
Mutation
Clinical data
Age of Onset (Year)
Family History
Functional Studies
Reference
H163P
FLAIR, showing bilateral hippocampal, temporal lobe atrophy, and paraventricular hyperintensity.
PET, revealing moderate-to-severe hypometabolism in the diffuse cortical area.
Immunohistochemical stain revealed expression of amyloid beta in the senile plaque
34
No
↑Aβ42/Aβ40 ratio; ↑Aβ42; Aβ40.
[18] Kim et al., 2012
H163R
Data are limited, but neuropathology consistent with AD has been observed in at least one case.
42–47
Yes
↓Aβ42/Aβ total ratio in COS-1 cells; ↓Aβ42 and Aβ40 production in vitro. Involving γ-secretase- neurexin processing.
[51] Martin et al., 1995;
H163Y
Typical AD neuropathology (one case); decreased glucose metabolism in presymptomatic mutation carriers, especially in the thalamus. Widespread brain amyloid (PiB-PET) and shrunken hippocampi.
47
Yes
↓CSF Aβ42 and Aβ38 levels. ↑Aβ42/Aβ total ratio when expressed in COS-1 cells, and ↑Aβ42 production
[52] Clark et al., 1995
A164V
MRI revealed atrophy in brain involved anterior temporal lobe, and the hippocampus.
45–50
Yes
Possibly damaging via in silico.
[19] Roeber et al., 2015
W165C (G > C)
Not available
37–47
Yes
↑Aβ42 and ↓Aβ40; elevated Aβ42/Aβ40 ratio
[36] Campion et al., 1999
W165C (G > T)
EOAD, a severe form of atrophies and rapid deterioration in cerebral and cerebellar
45
Yes
↓Aβ42 and ↓Aβ40; elevated Aβ42/Aβ40 ratio
[37] Syama et al., 2018
W165G
Not available
34–38
Yes
Not available
[39] Higuchi et al., 2000
L166H
MRI: hippocampal atrophy and cortical atrophy. SPECT: bilateral hypometabolism in the parietal and frontal lobes.
30
Yes
Not available
[26] Pantieri et al., 2005
L166P
numerous Aβ-positive neuritic and cotton-wool plaques; abundant Aβ-positive amyloid cores in the cerebellar cortex.
  
↑Aβ42/Aβ ratio; ↓Aβ40, Aβ42, and AICD.↓cleavage of Notch and N-cadherin.
[53] Moehlmann et al., 2002
L166R
MRI revealed cortical atrophy;
PET revealed parietal hypoperfusion.
32–34
Yes
Not available
[27] Ezquerra et al., 2000
L166 V
SPECT indicated temporoparietal hypoperfusion. Advanced plaques and tangles
42–50
Yes
Possibly damaging by in silico
[20] Sassi et al., 2014
L166del
MRI strongly revealed symmetrical cerebral atrophy,
40–46
Yes
Predicted possibly damaging in silico
[21] Knight et al., 2007
I167del
Symptom was progressive memory loss, behavior variants and spastic paraparesis.
38–46
 
Pathogenic by in silico
[54] Jiao et al., 2014
I168del
Not available
 
Yes
Pathogenic by in silico
[9] Janssen et al., 2003
I168T
Neuropathology consistent with AD.
86–94
Yes
Pathogenic by in silico
[20] Sassi et al., 2014
S169del (ΔS169, Ser169del, ΔS170)
MRI revealed cerebral atrophy involvement of the ventricles and widening of the sulci.
40
Yes
Not available
[28] Guo et al., 2010
S169 L
Aβ deposition in the cerebellum and white matter
33–37
Yes
Not available
[29] Taddei et al., 1998
S169P
Numerous plaques and neurofibrillary tangles could be seen in brain of the case
35
Yes
Not available
[30] Ezquerra et al., 1999
S170F
Much Lewy bodies in the substantia nigra and had severe cerebellar pathology
Also, abundant amyloid deposition and loss of Purkinje cells reported.
27
Yes
↑Aβ42 and Aβ40, altering the ratio.
[31] Snider et al., 2005
S170P
MRI shown hypointensity, globus pallidus, and substantia nigra, as well as frontotemporal cortical atrophy.
SPECT revealed severe nigrostriatal dopaminergic deficit bilaterally, and 18F-FDG PET hypometabolism in striatal and posterior cingulate.
25
No
Pathogenic by in silico
[40] Carecchio et al., 2017
L171P
Not available
36–40
Yes
Not available
[41] Ramirez-Dueñas et al., 1998
L173F (G > T)
Not available
50.5
Yes
↑Aβ42; ↑Aβ42:Aβ40 ratio
[42] Jin et al., 2012
L173F (G > C)
MRI revealed atrophy of the medial temporal lobe. SPECT indicated hypoperfusion of the posterior cingulate gyri and other cortical areas.
40–42
Yes
↑Aβ42 than cells
↑Aβ42:Aβ40 ratio
[43] Kasuga et al., 2009
L173 W
Not available
24–29
Yes
Not available
[36] Campion et al., 1999
L174del
MRI shown slight temporal lobe atrophy.
53
Yes
↑Aβ40, and ↓Aβ42 and Aβ42/Aβ40
[44] Tiedt et al., 2013
L174 M
Neuropathology consistent with AD and CAA associated
58
No
↓Aβ40 and ↑ Aβ42/Aβ40 ratio
[45] Tedde et al., 2003
L174R
EOAD
46–56
Yes
Not available
[46] Klünemann et al., 2004
F175S
Not available
NA
Yes
Not available
[47] Colacicco et al., 2002
F176 L
Much abundant amyloid plaques and neurofibrillary tangles in the cortex.
51
Yes
Not available
[48] Müller et al., 2013
F177 L
Not available
60
Not available
Not available
[49] Rogaeva et al., 2001
F177S
Not available
60
Not available
Not available
[49] Rogaeva et al., 2001
S178P
Not available
60
Not available
Not available
[49] Rogaeva et al., 2001
In our case, the PSEN1 p.Trp165Cys variant has been identified in a patient with early onset of age (50s years at diagnosis), suggesting that disease phenotype may be the result of amino acid substitution in this conservative residue. Furthermore, the amino acid position 165, located in the TM-III of PSEN1 indicated a significant phylogenetic conservation among vertebrates, and in homologous proteins such as PSEN2, suggesting that the position is of functional significance. Importantly, the patient’s mother and his brother were also affected by AD that is likely to involve autosomal dominant AD.
Guerrio et al. (2010) designed an algorithm on mutations on PSENs. which may be helpful in prediction on their pathogenic nature [32]. PSEN1 Trp165Cys may be a definitely pathogenic mutation. The Korean case of PSEN1 Trp165Cys may be associated with positive family history of disease, since the mother and one of the brother of patient was affected with AD. This is the third case of Trp165Cys, described all around the world, and EOAD was observed in all cases of AD. All of these findings suggested that Trp165 may be an important in PSEN1, since it bound two pathogenic mutations, Trp165Cys and Trp165Gly [3638]. Functional studies, performed by Sun et al. (2016) revealed that mutation may impair the gamma secretase activity, resulting in elevated amyloid beta 42 production [55]. Our findings confirmed the significance of PSEN1 Trp165Cys in EOAD.

Conclusions

We confirm that PSEN1 p.Trp165Cys may be commonly associated with EOAD. Our findings were consistent with the previously reported cases of this mutation, and supported the hypothesis that PSs contribute the identification of at risk relatives who may be potential candidates for clinical trials.

Acknowledgements

Not applicable.
This study was conducted with approval from the Institutional Review Board of Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital (B-1302/192–006).
Written informed consent was obtained from the patient for publication of this Case report and any accompanying images. A copy of the written consent is available for review by the Editor of this journal.

Competing interests

The authors declare that they have no competing of interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F, Fidani L, Giuffra L, Haynes A, Irving N, James L, et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature. 1991;349(6311):704–6.PubMedCrossRef Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F, Fidani L, Giuffra L, Haynes A, Irving N, James L, et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature. 1991;349(6311):704–6.PubMedCrossRef
2.
Zurück zum Zitat Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, Chi H, Lin C, Li G, Holman K, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature. 1995;375(6534):754–60.PubMedCrossRef Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, Chi H, Lin C, Li G, Holman K, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature. 1995;375(6534):754–60.PubMedCrossRef
3.
Zurück zum Zitat Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, Yu CE, Jondro PD, Schmidt SD, Wang K, et al. Candidate gene for the chromosome 1 familial Alzheimer's disease locus. Science. 1995;269(5226):973–7.PubMedCrossRef Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, Yu CE, Jondro PD, Schmidt SD, Wang K, et al. Candidate gene for the chromosome 1 familial Alzheimer's disease locus. Science. 1995;269(5226):973–7.PubMedCrossRef
4.
5.
Zurück zum Zitat Loy CT, Schofield PR, Turner AM, Kwok JB. Genetics of dementia. Lancet. 2014;383(9919):828–40.PubMedCrossRef Loy CT, Schofield PR, Turner AM, Kwok JB. Genetics of dementia. Lancet. 2014;383(9919):828–40.PubMedCrossRef
6.
Zurück zum Zitat Giau VV, Bagyinszky E, Yang YS, Youn YC, An SSA, Kim SY. Genetic analyses of early-onset Alzheimer’s disease using next generation sequencing. Sci Rep. 2019;9(1):8368.PubMedPubMedCentralCrossRef Giau VV, Bagyinszky E, Yang YS, Youn YC, An SSA, Kim SY. Genetic analyses of early-onset Alzheimer’s disease using next generation sequencing. Sci Rep. 2019;9(1):8368.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Giau VV, Senanarong V, Bagyinszky E, An SSA, Kim S. Analysis of 50 neurodegenerative genes in clinically diagnosed early-onset Alzheimer's disease. Int J Mol Sci. 2019;20(6).PubMedCentralCrossRef Giau VV, Senanarong V, Bagyinszky E, An SSA, Kim S. Analysis of 50 neurodegenerative genes in clinically diagnosed early-onset Alzheimer's disease. Int J Mol Sci. 2019;20(6).PubMedCentralCrossRef
8.
Zurück zum Zitat Raux G, Guyant-Marechal L, Martin C, Bou J, Penet C, Brice A, Hannequin D, Frebourg T, Campion D. Molecular diagnosis of autosomal dominant early onset Alzheimer's disease: an update. J Med Genet. 2005;42(10):793–5.PubMedPubMedCentralCrossRef Raux G, Guyant-Marechal L, Martin C, Bou J, Penet C, Brice A, Hannequin D, Frebourg T, Campion D. Molecular diagnosis of autosomal dominant early onset Alzheimer's disease: an update. J Med Genet. 2005;42(10):793–5.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Janssen JC, Beck JA, Campbell TA, Dickinson A, Fox NC, Harvey RJ, Houlden H, Rossor MN, Collinge J. Early onset familial Alzheimer's disease: mutation frequency in 31 families. Neurology. 2003;60(2):235–9.PubMedCrossRef Janssen JC, Beck JA, Campbell TA, Dickinson A, Fox NC, Harvey RJ, Houlden H, Rossor MN, Collinge J. Early onset familial Alzheimer's disease: mutation frequency in 31 families. Neurology. 2003;60(2):235–9.PubMedCrossRef
10.
Zurück zum Zitat Giau VV, Bagyinszky E, An SSA, Kim S. Clinical genetic strategies for early onset neurodegenerative diseases. Molecular & Cellular Toxicology. 2018;14(2):123–42.CrossRef Giau VV, Bagyinszky E, An SSA, Kim S. Clinical genetic strategies for early onset neurodegenerative diseases. Molecular & Cellular Toxicology. 2018;14(2):123–42.CrossRef
11.
Zurück zum Zitat Giau VV, Pyun JM, Bagyinszky E, An SSA, Kim S. A pathogenic PSEN2 p.His169Asn mutation associated with early-onset Alzheimer's disease. Clin Interv Aging. 2018;13:1321–9.PubMedPubMedCentralCrossRef Giau VV, Pyun JM, Bagyinszky E, An SSA, Kim S. A pathogenic PSEN2 p.His169Asn mutation associated with early-onset Alzheimer's disease. Clin Interv Aging. 2018;13:1321–9.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Tolia A, De Strooper B. Structure and function of gamma-secretase. Semin Cell Dev Biol. 2009;20(2):211–8.PubMedCrossRef Tolia A, De Strooper B. Structure and function of gamma-secretase. Semin Cell Dev Biol. 2009;20(2):211–8.PubMedCrossRef
13.
Zurück zum Zitat De Strooper B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-secretase complex. Neuron. 2003;38(1):9–12.PubMedCrossRef De Strooper B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-secretase complex. Neuron. 2003;38(1):9–12.PubMedCrossRef
14.
Zurück zum Zitat Giau VV, Lee H, Shim KH, Bagyinszky E, An SSA. Genome-editing applications of CRISPR-Cas9 to promote in vitro studies of Alzheimer's disease. Clin Interv Aging. 2018;13:221–33.PubMedPubMedCentralCrossRef Giau VV, Lee H, Shim KH, Bagyinszky E, An SSA. Genome-editing applications of CRISPR-Cas9 to promote in vitro studies of Alzheimer's disease. Clin Interv Aging. 2018;13:221–33.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Fernandez MA, Klutkowski JA, Freret T, Wolfe MS. Alzheimer presenilin-1 mutations dramatically reduce trimming of long amyloid beta-peptides (Abeta) by gamma-secretase to increase 42-to-40-residue Abeta. J Biol Chem. 2014;289(45):31043–52.PubMedPubMedCentralCrossRef Fernandez MA, Klutkowski JA, Freret T, Wolfe MS. Alzheimer presenilin-1 mutations dramatically reduce trimming of long amyloid beta-peptides (Abeta) by gamma-secretase to increase 42-to-40-residue Abeta. J Biol Chem. 2014;289(45):31043–52.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Xia D, Watanabe H, Wu B, Lee SH, Li Y, Tsvetkov E, Bolshakov VY, Shen J, Kelleher RJ 3rd. Presenilin-1 knockin mice reveal loss-of-function mechanism for familial Alzheimer's disease. Neuron. 2015;85(5):967–81.PubMedPubMedCentralCrossRef Xia D, Watanabe H, Wu B, Lee SH, Li Y, Tsvetkov E, Bolshakov VY, Shen J, Kelleher RJ 3rd. Presenilin-1 knockin mice reveal loss-of-function mechanism for familial Alzheimer's disease. Neuron. 2015;85(5):967–81.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8(2):101–12.PubMedCrossRef Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8(2):101–12.PubMedCrossRef
18.
Zurück zum Zitat Kim J, Bagyinszky E, Chang YH, Choe G, Choi BO, An SS, Kim S. A novel PSEN1 H163P mutation in a patient with early-onset Alzheimer's disease: clinical, neuroimaging, and neuropathological findings. Neurosci Lett. 2012;530(2):109–14.PubMedCrossRef Kim J, Bagyinszky E, Chang YH, Choe G, Choi BO, An SS, Kim S. A novel PSEN1 H163P mutation in a patient with early-onset Alzheimer's disease: clinical, neuroimaging, and neuropathological findings. Neurosci Lett. 2012;530(2):109–14.PubMedCrossRef
19.
Zurück zum Zitat Roeber S, Muller-Sarnowski F, Kress J, Edbauer D, Kuhlmann T, Tuttelmann F, Schindler C, Winter P, Arzberger T, Muller U, et al. Three novel presenilin 1 mutations marking the wide spectrum of age at onset and clinical patterns in familial Alzheimer's disease. J Neural Transm (Vienna). 2015;122(12):1715–9.CrossRef Roeber S, Muller-Sarnowski F, Kress J, Edbauer D, Kuhlmann T, Tuttelmann F, Schindler C, Winter P, Arzberger T, Muller U, et al. Three novel presenilin 1 mutations marking the wide spectrum of age at onset and clinical patterns in familial Alzheimer's disease. J Neural Transm (Vienna). 2015;122(12):1715–9.CrossRef
20.
Zurück zum Zitat Sassi C, Guerreiro R, Gibbs R, Ding J, Lupton MK, Troakes C, Lunnon K, Al-Sarraj S, Brown KS, Medway C, et al. Exome sequencing identifies 2 novel presenilin 1 mutations (p.L166V and p.S230R) in British early-onset Alzheimer's disease. Neurobiol Aging. 2014;35(10):2422.e2413–26.CrossRef Sassi C, Guerreiro R, Gibbs R, Ding J, Lupton MK, Troakes C, Lunnon K, Al-Sarraj S, Brown KS, Medway C, et al. Exome sequencing identifies 2 novel presenilin 1 mutations (p.L166V and p.S230R) in British early-onset Alzheimer's disease. Neurobiol Aging. 2014;35(10):2422.e2413–26.CrossRef
21.
Zurück zum Zitat Knight WD, Kennedy J, Mead S, Rossor MN, Beck J, Collinge J, Mummery C. A novel presenilin 1 deletion (p.L166del) associated with early onset familial Alzheimer's disease. Eur J Neurol. 2007;14(7):829–31.PubMedCrossRef Knight WD, Kennedy J, Mead S, Rossor MN, Beck J, Collinge J, Mummery C. A novel presenilin 1 deletion (p.L166del) associated with early onset familial Alzheimer's disease. Eur J Neurol. 2007;14(7):829–31.PubMedCrossRef
22.
Zurück zum Zitat Larner AJ, Doran M. Clinical phenotypic heterogeneity of Alzheimer's disease associated with mutations of the presenilin-1 gene. J Neurol. 2006;253(2):139–58.PubMedCrossRef Larner AJ, Doran M. Clinical phenotypic heterogeneity of Alzheimer's disease associated with mutations of the presenilin-1 gene. J Neurol. 2006;253(2):139–58.PubMedCrossRef
23.
Zurück zum Zitat Giau VV, Wang MJ, Bagyinszky E, Youn YC, An SSA, Kim S. Novel PSEN1 p.Gly417Ala mutation in a Korean patient with early-onset Alzheimer's disease with parkinsonism. Neurobiol Aging. 2018;72:188.e113–7.CrossRef Giau VV, Wang MJ, Bagyinszky E, Youn YC, An SSA, Kim S. Novel PSEN1 p.Gly417Ala mutation in a Korean patient with early-onset Alzheimer's disease with parkinsonism. Neurobiol Aging. 2018;72:188.e113–7.CrossRef
24.
Zurück zum Zitat Bagyinszky E, Lee HM, Van Giau V, Koh SB, Jeong JH, An SSA, Kim S. PSEN1 p.Thr116Ile variant in two Korean families with young onset Alzheimer's disease. Int J Mol Sci. 2018;19(9):2604.PubMedCentralCrossRef Bagyinszky E, Lee HM, Van Giau V, Koh SB, Jeong JH, An SSA, Kim S. PSEN1 p.Thr116Ile variant in two Korean families with young onset Alzheimer's disease. Int J Mol Sci. 2018;19(9):2604.PubMedCentralCrossRef
25.
Zurück zum Zitat Park J, An SSA, Giau VV, Shim K, Youn YC, Bagyinszky E, Kim S. Identification of a novel PSEN1 mutation (Leu232Pro) in a Korean patient with early-onset Alzheimer's disease and a family history of dementia. Neurobiol Aging. 2017;56:212.e211–7.CrossRef Park J, An SSA, Giau VV, Shim K, Youn YC, Bagyinszky E, Kim S. Identification of a novel PSEN1 mutation (Leu232Pro) in a Korean patient with early-onset Alzheimer's disease and a family history of dementia. Neurobiol Aging. 2017;56:212.e211–7.CrossRef
26.
Zurück zum Zitat Pantieri R, Pardini M, Cecconi M, Dagna-Bricarelli F, Vitali A, Piccini A, Russo R, Borghi R, Tabaton M. A novel presenilin 1 L166H mutation in a pseudo-sporadic case of early-onset Alzheimer's disease. Neurol Sci. 2005;26(5):349–50.PubMedCrossRef Pantieri R, Pardini M, Cecconi M, Dagna-Bricarelli F, Vitali A, Piccini A, Russo R, Borghi R, Tabaton M. A novel presenilin 1 L166H mutation in a pseudo-sporadic case of early-onset Alzheimer's disease. Neurol Sci. 2005;26(5):349–50.PubMedCrossRef
27.
Zurück zum Zitat Ezquerra M, Carnero C, Blesa R, Oliva R. A novel presenilin 1 mutation (Leu166Arg) associated with early-onset Alzheimer disease. Arch Neurol. 2000;57(4):485–8.PubMedCrossRef Ezquerra M, Carnero C, Blesa R, Oliva R. A novel presenilin 1 mutation (Leu166Arg) associated with early-onset Alzheimer disease. Arch Neurol. 2000;57(4):485–8.PubMedCrossRef
28.
Zurück zum Zitat Guo J, Wei J, Liao S, Wang L, Jiang H, Tang B. A novel presenilin 1 mutation (Ser169del) in a Chinese family with early-onset Alzheimer's disease. Neurosci Lett. 2010;468(1):34–7.PubMedCrossRef Guo J, Wei J, Liao S, Wang L, Jiang H, Tang B. A novel presenilin 1 mutation (Ser169del) in a Chinese family with early-onset Alzheimer's disease. Neurosci Lett. 2010;468(1):34–7.PubMedCrossRef
29.
Zurück zum Zitat Taddei K, Kwok JB, Kril JJ, Halliday GM, Creasey H, Hallupp M, Fisher C, Brooks WS, Chung C, Andrews C, et al. Two novel presenilin-1 mutations (Ser169Leu and Pro436Gln) associated with very early onset Alzheimer's disease. Neuroreport. 1998;9(14):3335–9.PubMedCrossRef Taddei K, Kwok JB, Kril JJ, Halliday GM, Creasey H, Hallupp M, Fisher C, Brooks WS, Chung C, Andrews C, et al. Two novel presenilin-1 mutations (Ser169Leu and Pro436Gln) associated with very early onset Alzheimer's disease. Neuroreport. 1998;9(14):3335–9.PubMedCrossRef
30.
Zurück zum Zitat Ezquerra M, Carnero C, Blesa R, Gelpi JL, Ballesta F, Oliva R. A presenilin 1 mutation (Ser169Pro) associated with early-onset AD and myoclonic seizures. Neurology. 1999;52(3):566–70.PubMedCrossRef Ezquerra M, Carnero C, Blesa R, Gelpi JL, Ballesta F, Oliva R. A presenilin 1 mutation (Ser169Pro) associated with early-onset AD and myoclonic seizures. Neurology. 1999;52(3):566–70.PubMedCrossRef
31.
Zurück zum Zitat Snider BJ, Norton J, Coats MA, Chakraverty S, Hou CE, Jervis R, Lendon CL, Goate AM, McKeel DW Jr, Morris JC. Novel presenilin 1 mutation (S170F) causing Alzheimer disease with Lewy bodies in the third decade of life. Arch Neurol. 2005;62(12):1821–30.PubMedCrossRef Snider BJ, Norton J, Coats MA, Chakraverty S, Hou CE, Jervis R, Lendon CL, Goate AM, McKeel DW Jr, Morris JC. Novel presenilin 1 mutation (S170F) causing Alzheimer disease with Lewy bodies in the third decade of life. Arch Neurol. 2005;62(12):1821–30.PubMedCrossRef
32.
Zurück zum Zitat Guerreiro RJ, Baquero M, Blesa R, Boada M, Bras JM, Bullido MJ, Calado A, Crook R, Ferreira C, Frank A, et al. Genetic screening of Alzheimer's disease genes in Iberian and African samples yields novel mutations in presenilins and APP. Neurobiol Aging. 2010;31(5):725–31.PubMedCrossRef Guerreiro RJ, Baquero M, Blesa R, Boada M, Bras JM, Bullido MJ, Calado A, Crook R, Ferreira C, Frank A, et al. Genetic screening of Alzheimer's disease genes in Iberian and African samples yields novel mutations in presenilins and APP. Neurobiol Aging. 2010;31(5):725–31.PubMedCrossRef
33.
Zurück zum Zitat Moulder KL, Snider BJ, Mills SL, Buckles VD, Santacruz AM, Bateman RJ, Morris JC. Dominantly inherited Alzheimer network: facilitating research and clinical trials. Alzheimers Res Ther. 2013;5(5):48.PubMedPubMedCentralCrossRef Moulder KL, Snider BJ, Mills SL, Buckles VD, Santacruz AM, Bateman RJ, Morris JC. Dominantly inherited Alzheimer network: facilitating research and clinical trials. Alzheimers Res Ther. 2013;5(5):48.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Van Giau V, An SSA, Bagyinszky E, Kim S. Gene panels and primers for next generation sequencing studies on neurodegenerative disorders. Molecular & Cellular Toxicology. 2015;11(2):89–143.CrossRef Van Giau V, An SSA, Bagyinszky E, Kim S. Gene panels and primers for next generation sequencing studies on neurodegenerative disorders. Molecular & Cellular Toxicology. 2015;11(2):89–143.CrossRef
35.
Zurück zum Zitat Källberg M, Wang H, Wang S, Peng J, Wang Z, Lu H, Xu J. Template-based protein structure modeling using the RaptorX web server. Nat Protoc. 2012;7(8):1511–22.PubMedPubMedCentralCrossRef Källberg M, Wang H, Wang S, Peng J, Wang Z, Lu H, Xu J. Template-based protein structure modeling using the RaptorX web server. Nat Protoc. 2012;7(8):1511–22.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Campion D, Dumanchin C, Hannequin D, Dubois B, Belliard S, Puel M, Thomas-Anterion C, Michon A, Martin C, Charbonnier F, et al. Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet. 1999;65(3):664–70.PubMedPubMedCentralCrossRef Campion D, Dumanchin C, Hannequin D, Dubois B, Belliard S, Puel M, Thomas-Anterion C, Michon A, Martin C, Charbonnier F, et al. Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet. 1999;65(3):664–70.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Syama A, Sen S, Kota LN, Viswanath B, Purushottam M, Varghese M, Jain S, Panicker MM, Mukherjee O. Mutation burden profile in familial Alzheimer's disease cases from India. Neurobiol Aging. 2018;64:158.e157–13.CrossRef Syama A, Sen S, Kota LN, Viswanath B, Purushottam M, Varghese M, Jain S, Panicker MM, Mukherjee O. Mutation burden profile in familial Alzheimer's disease cases from India. Neurobiol Aging. 2018;64:158.e157–13.CrossRef
38.
Zurück zum Zitat Wallon D, Rousseau S, Rovelet-Lecrux A, Quillard-Muraine M, Guyant-Marechal L, Martinaud O, Pariente J, Puel M, Rollin-Sillaire A, Pasquier F, et al. The French series of autosomal dominant early onset Alzheimer's disease cases: mutation spectrum and cerebrospinal fluid biomarkers. J Alzheimers Dis. 2012;30(4):847–56.PubMedCrossRef Wallon D, Rousseau S, Rovelet-Lecrux A, Quillard-Muraine M, Guyant-Marechal L, Martinaud O, Pariente J, Puel M, Rollin-Sillaire A, Pasquier F, et al. The French series of autosomal dominant early onset Alzheimer's disease cases: mutation spectrum and cerebrospinal fluid biomarkers. J Alzheimers Dis. 2012;30(4):847–56.PubMedCrossRef
39.
Zurück zum Zitat Higuchi S, Yoshino A, Matsui T. A novel PS1 mutation (W165G) in a Japanese family with early-onset Alzheimer’s disease. Alzheimers Rep. 2000;3:227–31. Higuchi S, Yoshino A, Matsui T. A novel PS1 mutation (W165G) in a Japanese family with early-onset Alzheimer’s disease. Alzheimers Rep. 2000;3:227–31.
40.
Zurück zum Zitat Carecchio M, Picillo M, Valletta L, Elia AE, Haack TB, Cozzolino A, Vitale A, Garavaglia B, Iuso A, Bagella CF, et al. Rare causes of early-onset dystonia-parkinsonism with cognitive impairment: a de novo PSEN-1 mutation. neurogenetics. 2017;18(3):175–8.PubMedCrossRef Carecchio M, Picillo M, Valletta L, Elia AE, Haack TB, Cozzolino A, Vitale A, Garavaglia B, Iuso A, Bagella CF, et al. Rare causes of early-onset dystonia-parkinsonism with cognitive impairment: a de novo PSEN-1 mutation. neurogenetics. 2017;18(3):175–8.PubMedCrossRef
41.
Zurück zum Zitat Ramirez-Duenas MG, Rogaeva EA, Leal CA, Lin C, Ramirez-Casillas GA, Hernandez-Romo JA, St George-Hyslop PH, Cantu JM. A novel Leu171Pro mutation in presenilin-1 gene in a Mexican family with early onset Alzheimer disease. Ann Genet. 1998;41(3):149–53.PubMed Ramirez-Duenas MG, Rogaeva EA, Leal CA, Lin C, Ramirez-Casillas GA, Hernandez-Romo JA, St George-Hyslop PH, Cantu JM. A novel Leu171Pro mutation in presenilin-1 gene in a Mexican family with early onset Alzheimer disease. Ann Genet. 1998;41(3):149–53.PubMed
42.
Zurück zum Zitat Jin SC, Pastor P, Cooper B, Cervantes S, Benitez BA, Razquin C, Goate A, Cruchaga C. Pooled-DNA sequencing identifies novel causative variants in PSEN1, GRN and MAPT in a clinical early-onset and familial Alzheimer's disease Ibero-American cohort. Alzheimers Res Ther. 2012;4(4):34.PubMedPubMedCentralCrossRef Jin SC, Pastor P, Cooper B, Cervantes S, Benitez BA, Razquin C, Goate A, Cruchaga C. Pooled-DNA sequencing identifies novel causative variants in PSEN1, GRN and MAPT in a clinical early-onset and familial Alzheimer's disease Ibero-American cohort. Alzheimers Res Ther. 2012;4(4):34.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Kasuga K, Ohno T, Ishihara T, Miyashita A, Kuwano R, Onodera O, Nishizawa M, Ikeuchi T. Depression and psychiatric symptoms preceding onset of dementia in a family with early-onset Alzheimer disease with a novel PSEN1 mutation. J Neurol. 2009;256(8):1351–3.PubMedCrossRef Kasuga K, Ohno T, Ishihara T, Miyashita A, Kuwano R, Onodera O, Nishizawa M, Ikeuchi T. Depression and psychiatric symptoms preceding onset of dementia in a family with early-onset Alzheimer disease with a novel PSEN1 mutation. J Neurol. 2009;256(8):1351–3.PubMedCrossRef
44.
Zurück zum Zitat Tiedt HO, Lueschow A, Winter P, Muller U. Previously not recognized deletion in presenilin-1 (p.Leu174del.) in a patient with early-onset familial Alzheimer's disease. Neurosci Lett. 2013;544:115–8.PubMedCrossRef Tiedt HO, Lueschow A, Winter P, Muller U. Previously not recognized deletion in presenilin-1 (p.Leu174del.) in a patient with early-onset familial Alzheimer's disease. Neurosci Lett. 2013;544:115–8.PubMedCrossRef
45.
Zurück zum Zitat Tedde A, Nacmias B, Ciantelli M, Forleo P, Cellini E, Bagnoli S, Piccini C, Caffarra P, Ghidoni E, Paganini M, et al. Identification of new Presenilin gene mutations in early-onset familial Alzheimer disease. Arch Neurol. 2003;60(11):1541–4.PubMedCrossRef Tedde A, Nacmias B, Ciantelli M, Forleo P, Cellini E, Bagnoli S, Piccini C, Caffarra P, Ghidoni E, Paganini M, et al. Identification of new Presenilin gene mutations in early-onset familial Alzheimer disease. Arch Neurol. 2003;60(11):1541–4.PubMedCrossRef
46.
Zurück zum Zitat Klunemann HH, Rogaeva E, Neumann M, Kretzschmar HA, Kandel M, Toulina A, Sato C, Salehi-Rad S, Pfister K, Klein HE, et al. Novel PS1 mutation in a Bavarian kindred with familial Alzheimer disease. Alzheimer Dis Assoc Disord. 2004;18(4):256–8.PubMed Klunemann HH, Rogaeva E, Neumann M, Kretzschmar HA, Kandel M, Toulina A, Sato C, Salehi-Rad S, Pfister K, Klein HE, et al. Novel PS1 mutation in a Bavarian kindred with familial Alzheimer disease. Alzheimer Dis Assoc Disord. 2004;18(4):256–8.PubMed
47.
Zurück zum Zitat Colacicco AM, Panza F, Basile AM, Solfrizzi V, Capurso C, D’Introno A, Torres F, Capurso S, Cozza S, Flora R, et al. F175S change and a novel polymorphism in Presenilin-1 gene in late-onset familial Alzheimer’s disease. Eur Neurol. 2002;47(4):209–13.PubMedCrossRef Colacicco AM, Panza F, Basile AM, Solfrizzi V, Capurso C, D’Introno A, Torres F, Capurso S, Cozza S, Flora R, et al. F175S change and a novel polymorphism in Presenilin-1 gene in late-onset familial Alzheimer’s disease. Eur Neurol. 2002;47(4):209–13.PubMedCrossRef
48.
Zurück zum Zitat Muller U, Winter P, Graeber MB. A presenilin 1 mutation in the first case of Alzheimer's disease. Lancet Neurol. 2013;12(2):129–30.PubMedCrossRef Muller U, Winter P, Graeber MB. A presenilin 1 mutation in the first case of Alzheimer's disease. Lancet Neurol. 2013;12(2):129–30.PubMedCrossRef
49.
Zurück zum Zitat Rogaeva EA, Fafel KC, Song YQ, Medeiros H, Sato C, Liang Y, Richard E, Rogaev EI, Frommelt P, Sadovnick AD, et al. Screening for PS1 mutations in a referral-based series of AD cases: 21 novel mutations. Neurology. 2001;57(4):621–5.PubMedCrossRef Rogaeva EA, Fafel KC, Song YQ, Medeiros H, Sato C, Liang Y, Richard E, Rogaev EI, Frommelt P, Sadovnick AD, et al. Screening for PS1 mutations in a referral-based series of AD cases: 21 novel mutations. Neurology. 2001;57(4):621–5.PubMedCrossRef
50.
Zurück zum Zitat Sassi C, Guerreiro R, Gibbs R, Ding J, Lupton MK, Troakes C, Al-Sarraj S, Niblock M, Gallo JM, Adnan J, et al. Investigating the role of rare coding variability in Mendelian dementia genes (APP, PSEN1, PSEN2, GRN, MAPT, and PRNP) in late-onset Alzheimer's disease. Neurobiol Aging. 2014;35(12):2881.e2881–6.CrossRef Sassi C, Guerreiro R, Gibbs R, Ding J, Lupton MK, Troakes C, Al-Sarraj S, Niblock M, Gallo JM, Adnan J, et al. Investigating the role of rare coding variability in Mendelian dementia genes (APP, PSEN1, PSEN2, GRN, MAPT, and PRNP) in late-onset Alzheimer's disease. Neurobiol Aging. 2014;35(12):2881.e2881–6.CrossRef
51.
Zurück zum Zitat Martin C, Charbonnier F, Flaman J-M, Frebourg T, Moreau V, Campion D, Calenda A, Bellis M, Heilig R, Mallet J, et al. Mutations of the presenilin I gene in families with early-onset Alzheimer's disease. Hum Mol Genet. 1995;4(12):2373–7.PubMedCrossRef Martin C, Charbonnier F, Flaman J-M, Frebourg T, Moreau V, Campion D, Calenda A, Bellis M, Heilig R, Mallet J, et al. Mutations of the presenilin I gene in families with early-onset Alzheimer's disease. Hum Mol Genet. 1995;4(12):2373–7.PubMedCrossRef
52.
Zurück zum Zitat Clark RF, Hutton M, Fuldner M, Froelich S, Karran E, Talbot C, Crook R, Lendon C, Prihar G, He C, et al. The structure of the presenilin 1 (S182) gene and identification of six novel mutations in early onset AD families. Nat Genet. 1995;11(2):219–22.CrossRef Clark RF, Hutton M, Fuldner M, Froelich S, Karran E, Talbot C, Crook R, Lendon C, Prihar G, He C, et al. The structure of the presenilin 1 (S182) gene and identification of six novel mutations in early onset AD families. Nat Genet. 1995;11(2):219–22.CrossRef
53.
Zurück zum Zitat Moehlmann T, Winkler E, Xia X, Edbauer D, Murrell J, Capell A, Kaether C, Zheng H, Ghetti B, Haass C, et al. Presenilin-1 mutations of leucine 166 equally affect the generation of the notch and APP intracellular domains independent of their effect on Abeta 42 production. Proc Natl Acad Sci U S A. 2002;99(12):8025–30.PubMedPubMedCentralCrossRef Moehlmann T, Winkler E, Xia X, Edbauer D, Murrell J, Capell A, Kaether C, Zheng H, Ghetti B, Haass C, et al. Presenilin-1 mutations of leucine 166 equally affect the generation of the notch and APP intracellular domains independent of their effect on Abeta 42 production. Proc Natl Acad Sci U S A. 2002;99(12):8025–30.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Jiao B, Tang B, Liu X, Xu J, Wang Y, Zhou L, Zhang F, Yan X, Zhou Y, Shen L. Mutational analysis in early-onset familial Alzheimer's disease in mainland China. Neurobiol Aging. 2014;35(8):1957.e1951–6.CrossRef Jiao B, Tang B, Liu X, Xu J, Wang Y, Zhou L, Zhang F, Yan X, Zhou Y, Shen L. Mutational analysis in early-onset familial Alzheimer's disease in mainland China. Neurobiol Aging. 2014;35(8):1957.e1951–6.CrossRef
55.
Zurück zum Zitat Sun L, Zhou R, Yang G, Shi Y. Analysis of 138 pathogenic mutations in presenilin-1 on the in vitro production of Abeta42 and Abeta40 peptides by gamma-secretase. Proc Natl Acad Sci U S A. 2017;114(4):E476–e485.PubMedCrossRef Sun L, Zhou R, Yang G, Shi Y. Analysis of 138 pathogenic mutations in presenilin-1 on the in vitro production of Abeta42 and Abeta40 peptides by gamma-secretase. Proc Natl Acad Sci U S A. 2017;114(4):E476–e485.PubMedCrossRef
Metadaten
Titel
A pathogenic PSEN1 Trp165Cys mutation associated with early-onset Alzheimer’s disease
verfasst von
Vo Van Giau
Jung-Min Pyun
Jeewon Suh
Eva Bagyinszky
Seong Soo A. An
Sang Yun Kim
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Neurology / Ausgabe 1/2019
Elektronische ISSN: 1471-2377
DOI
https://doi.org/10.1186/s12883-019-1419-y

Weitere Artikel der Ausgabe 1/2019

BMC Neurology 1/2019 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.