ArticlePDF Available

A Recessive Mutation in the APP Gene with Dominant-Negative Effect on Amyloidogenesis

Authors:

Abstract and Figures

β-Amyloid precursor protein (APP) mutations cause familial Alzheimer's disease with nearly complete penetrance. We found an APP mutation [alanine-673→valine-673 (A673V)] that causes disease only in the homozygous state, whereas heterozygous carriers were unaffected, consistent with a recessive Mendelian trait of inheritance. The A673V mutation affected APP processing, resulting in enhanced β-amyloid (Aβ) production and formation of amyloid fibrils in vitro. Co-incubation of mutated and wild-type peptides conferred instability on Aβ aggregates and inhibited amyloidogenesis and neurotoxicity. The highly amyloidogenic effect of the A673V mutation in the homozygous state and its anti-amyloidogenic effect in the heterozygous state account for the autosomal recessive pattern of inheritance and have implications for genetic screening and the potential treatment of Alzheimer's disease.
Content may be subject to copyright.
A Recessive Mutation in the APP Gene with Dominant-Negative
Effect on Amyloidogenesis
Giuseppe Di Fede1, Marcella Catania1, Michela Morbin1, Giacomina Rossi1, Silvia Suardi1,
Giulia Mazzoleni1, Marco Merlin1, Anna Rita Giovagnoli1, Sara Prioni1, Alessandra
Erbetta2, Chiara Falcone3, Marco Gobbi4, Laura Colombo4, Antonio Bastone4, Marten
Beeg4, Claudia Manzoni4, Bruna Francescucci5, Alberto Spagnoli5, Laura Cantù6, Elena Del
Favero6, Efrat Levy7, Mario Salmona4, and Fabrizio Tagliavini1,*
1Division of Neurology and Neuropathology, “Carlo Besta” National Neurological Institute, 20133
Milan, Italy.
2Division of Neuroradiology, “Carlo Besta” National Neurological Institute, 20133 Milan, Italy.
3Division of Neuroepidemiology, “Carlo Besta” National Neurological Institute, 20133 Milan, Italy.
4Department of Molecular Biochemistry and Pharmachology, Istituto di Ricerche Farmacologiche
“Mario Negri,” 20156 Milan, Italy.
5Division of Cognitive Disorders, Centro Sant’Ambrogio Fatebenefratelli, Cernusco sul Naviglio,
20063 Milan, Italy.
6Department of Medical Chemistry, Biochemistry, and Biotechnology, University of Milan, Segrate,
20090 Milan, Italy.
7Departments of Pharmacology and Psychiatry, New York University School of Medicine, and
Nathan S. Kline Institute, Orangeburg, NY 10962, USA.
Abstract
β-Amyloid precursor protein (APP) mutations cause familial Alzheimer’s disease with nearly
complete penetrance. We found an APP mutation [alanine-673valine-673 (A673V)] that causes
disease only in the homozygous state, whereas heterozygous carriers were unaffected, consistent
with a recessive Mendelian trait of inheritance. The A673V mutation affected APP processing,
resulting in enhanced β-amyloid (Aβ) production and formation of amyloid fibrils in vitro. Co-
incubation of mutated and wild-type peptides conferred instability on Aβ aggregates and inhibited
amyloidogenesis and neurotoxicity. The highly amyloidogenic effect of the A673V mutation in the
homozygous state and its anti-amyloidogenic effect in the heterozygous state account for the
autosomal recessive pattern of inheritance and have implications for genetic screening and the
potential treatment of Alzheimer’s disease.
Acentral pathological feature of Alzheimer’s disease (AD) is the accumulation of β-Aβ in the
form of oligomers and amyloid fibrils in the brain (1). Aβ is generated by sequential cleavage
of the APP by β- and γ-secretases and exists as short and long isoforms, Aβ1-40 and Aβ1-42
(2). Aβ1-42 is especially prone to misfolding and builds up aggregates that are thought to be
the primary neurotoxic species involved in AD pathogenesis (2,3). AD is usually sporadic, but
*To whom correspondence should be addressed. E-mail: ftagliavini@istituto-besta.it.
Publisher's Disclaimer: This manuscript has been accepted for publication in Science. This version has not undergone final editing.
Please refer to the complete version of record at http://www.sciencemag.org/. The manuscript may not be reproduced or used in any
manner that does not fall within the fair use provisions of the Copyright Act without the prior, written permission of AAAS.
NIH Public Access
Author Manuscript
Science. Author manuscript; available in PMC 2010 March 13.
Published in final edited form as:
Science. 2009 March 13; 323(5920): 1473–1477. doi:10.1126/science.1168979.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
a small fraction of cases is familial (4). The familial forms show an autosomal dominant pattern
of inheritance with virtually complete penetrance and are linked to mutations in the APP,
presenilin 1, and presenilin 2 genes (5). The APP mutations close to the sites of β- or γ-secretase
cleavage flanking the Aβ sequence overproduce total Aβ or only Aβ1-42, respectively, whereas
those that alter amino acids within Aβ result in greater propensity to aggregation in vitro (6,
7).
We have identified an APP mutation [Ala673Val673 (A673V)] that causes disease only in
the homozygous state. The mutation consists of a C-to-T transition that results in an alanine-
to-valine substitution at position 673 (APP770 numbering) corresponding to position 2 of Aβ
(Fig. 1A and fig. S1) (8). The genetic defect was found in a patient with early-onset dementia
and in his younger sister, who now shows multiple-domain mild cognitive impairment (MCI)
(9). Six relatives aged between 21 and 88 years, from both parental lineages, who carry the
A673V mutation in the heterozygous state were not affected, as deduced by formal
neuropsychological assessment [supporting online material (SOM) text, fig. S2, and table S1],
consistent with a recessive Mendelian trait of inheritance. The A673V mutation was not found
in 200 healthy individuals and 100 sporadic AD patients. Both mutated and wild-type APP
mRNA were expressed in heterozygous carriers (8).
In the patient, the disease presented with behavioral changes and cognitive deficits at the age
of 36 years and evolved toward severe dementia with spastic tetraparesis, leading to complete
loss of autonomy in about 8 years (SOM text). Serial magnetic resonance imaging showed
progressive cortico-subcortical atrophy (fig. S3). Cerebrospinal fluid analysis evidenced
decreased Aβ1-42 and increased total and 181T-phosphorylated tau compared with that of
nondemented controls and similarly to AD subjects (table S2 and fig. S4) (8). In the plasma of
the patient and his A673V homozygous sister, Aβ1-40 and Aβ1-42 were higher than those in
nondemented controls, whereas the six A673V heterozygous carriers had intermediate amounts
(table S2 and fig. S4).
In conditioned media of fibroblasts prepared from skin biopsies (8), Aβ1-40 and Aβ1-42 were
2.1- and 1.7-fold higher in the patient than in four age-matched controls with no change in
Aβ1-42:Aβ1-40 ratio (table S2 and fig. S4), suggesting that the A673V variant alters APP
processing, which promotes an increase in Aβ formation. To confirm this, we transiently
transfected Chinese hamster ovary (CHO) and COS-7 cells with either mutant or wild-type
APP cDNA and measured Aβ in conditioned media by enzyme-linked immunosorbent assay
(ELISA) (8). Cells expressing A673V APP had significantly higher amounts of both Aβ1-40
and Aβ1-42 than did cells transfected with wild-type APP, with no change in Aβ1-42:Aβ1-40
ratio (table S2). CHO and COS-7 cells with the A673V mutation also had increased secretion
of amino-terminally truncated Aβ species, including Aβ11-40, Aβ11-42, and AβN3pE-42
(table S2). These differences were paralleled by differences in the production of soluble forms
of APP (sAPPβ and sAPPα) and of APP carboxy-terminal fragments (C99 and C83) that
derived from the amyloidogenic β-secretase or nonamyloidogenic α-secretase processing (8).
Fibroblasts of the patient showed increased secretion of sAPPβ and 2.5-fold increase in
sAPPβ:sAPPα ratio (mean of three determinations: 0.5) compared with those of four age-
matched controls [0.2 ± 0.01 (SD)] as deduced by ELISA. Similarly, the sAPPβ:sAPPα ratio
was significantly higher in media from CHO cells expressing the A673V mutation (0.4 ± 0.1)
than in media from control cells (0.1 ± 0.03, P =0.03) (Fig. 1B). Immunoblot analysis of cell
lysates with an antibody to the carboxy-terminal region of APP (8) showed a 1.9 ± 0.2 increase
in C99:C83 ratio in patient’s fibroblasts (mean of three determinations: 0.67) compared with
that in control fibroblasts (0.35 ± 0.02) and 2.5 ± 0.2 increase in mutated CHO cells (0.52 ±
0.10) compared with that of control cells (0.21 ± 0.05, P = 0.0001) (Fig. 1, C and D).
Di Fede et al. Page 2
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
We then investigated the effects of the A673V mutation on the aggregation and amyloidogenic
properties of Aβ by using synthetic peptides homologous to residues 1 to 40 with and without
the A-to-V substitution at position 2 (Aβ1-40mut and Aβ1-40wt) (8). Laser light scattering
measurements over short periods (first 24 hours after sample preparation) showed that the
aggregation kinetics was faster for Aβ1-40mut than for Aβ1-40wt and that the time constants
of the exponential increase were 1.3 hours and 5.8 hours, respectively (Fig. 2A). Furthermore,
although the initial size distribution of particles generated by the two peptides was similar,
after 24 hours Aβ1-40mut assemblies were much larger than Aβ1-40wt aggregates (Fig. 2B).
Polarized-light and electron microscopy (EM) showed that Aβ1-40mut aggregates with the
tinctorial properties of amyloid (i.e., birefringence after Congo red staining) ultrastructurally
formed by straight, unbranched, 8-nm-diameter fibrils were already apparent after 4 hours.
Amyloid progressively increased up to 5 days, when the samples contained only fibrils
organized in dense meshwork (Fig. 3, B, E, H, and K). Aβ1-40wt followed a qualitatively
similar assembly path but with much slower kinetics. The 8-nm-diameter amyloid fibrils were
first observed after 72 hours, mingled with oligomers and protofibrils (Fig. 3, A and G), and
the size and density of congophilic aggregates reached a plateau only after 20 days (Fig. 3, D
and J). Similar differences were observed between wild-type and mutated peptides homologous
to residues 1 to 42 of Aβ (Fig. 3, M and N), although the aggregation kinetics was faster
compared with that of Aβ1-40.
The finding that the A673V mutation strongly boosts Aβ production and fibrillogenesis raises
the question of why heterozygous carriers do not develop disease, so we analyzed the effects
of the interaction between Aβ1-40mut and Aβ1-40wt. Laser light scattering showed that the
time constant of aggregate formation of equimolar mixtures of wild-type and mutated peptides
was higher (8.3 hours) than the time to aggregate for either Aβ1-40mut (1.3 hours) or
Aβ1-40wt alone (5.8 hours) (Fig. 2A) and that the size distribution of particles was lowest both
at time 0 and after 24 hours (Fig. 2B). Furthermore, the aggregates formed by peptide mixtures
were far more unstable than those generated by either Aβ1-40wt or Aβ1-40mut after dilution
with buffer, with a characteristic dissolution time of 8 min. At the same time, no dissolution
kinetics was observed for samples of Aβ1-40wt and Aβ1-40mut alone (Fig. 2C). This was
confirmed by urea denaturation studies of peptide aggregates (8). Size exclusion
chromatography showed that the elution profiles of Aβ1-40wt and Aβ1-40mut were marked by
a single peak corresponding to the dimer, whereas the mixture gave a smaller peak area
corresponding to the dimer and a second small peak corresponding to the monomer (Fig. 4A).
Polarized light and EM showed that the peptide mixture built up much fewer congophilic
aggregates than not only Aβ1-40mut but also Aβ1-40wt (Fig. 3, C, F, I, and L). Similar results
were observed with Aβ1-42 peptides (Fig. 3, M to O). Amyloid formation was also inhibited
when Aβ1-40wt was incubated with a hexapeptide homologous to residues 1 to 6 containing
the A-to-V substitution in position 2 (Aβ1-6mut) at 1:4 molar ratio (fig. S5).
We analyzed the binding of Aβ peptides with and without the A673V mutation to Aβ1-40wt
by using surface plasmon resonance (8). In addition to Aβ1-40wt and Aβ1-40mut, we used the
hexa-peptides Aβ1-6wt and Aβ1-6mut to evaluate the independent contribution of the amino-
terminal sequence containing the mutation. No difference in binding to immobilized
Aβ1-40wt fibrils was observed between Aβ1-40wt and Aβ1-40mut, consistent with the finding
that Aβ aggregation is primarily driven by hydrophobic stretches in the central and carboxy-
terminal parts of the peptide (Fig. 4B) (10). However, the amino-terminal fragment
Aβ1-6mut showed greater ability to bind to wild-type Aβ1-40 than did Aβ1-6wt (Fig. 4C),
indicating that the A-to-V substitution at position 2 favors the interaction between mutant and
wild-type Aβ.
Lastly, we treated human neuroblastoma SH-SY5Y cells with Aβ1-42wt, Aβ1-42mut, or
mixtures thereof at 5 μM for 24 hours and assessed cell viability by 3-(4,5-dimethylthiazol-2-
Di Fede et al. Page 3
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
yl)-2,5-diphenyl tetrasodium bromide (8): Aβ1-42mut was more toxic than Aβ1-42wt, and the
mixture was significantly less toxic than either peptide alone (Fig. 4D).
We have identified a mutation in the APP gene showing a recessive Mendelian trait of
inheritance. Recently, a homozygous APP mutation (A693Δ) was detected in three AD patients
from two Japanese pedigrees (11). Because one out of four heterozygous individuals had MCI,
in the absence of experimental studies mimicking the situation in heterozygotes, it is hard to
establish whether A693Δ is a recessive mutation or a dominant APP variant with incomplete
penetrance.
The A673V APP mutation has two pathogenic effects: it (i) shifts APP processing toward the
amyloidogenic pathway and (ii) enhances the aggregation and fibrillogenic properties of Aβ.
However, the interaction between mutant and wild-type Aβ, favored by the A-to-V substitution
at position 2, interferes with nucleation or nucleation-dependent polymerization, or both,
hindering amyloidogenesis and neurotoxicity and thus protecting the heterozygous carriers.
Until recently, the importance of the aminoterminal sequence of Aβ in misfolding and disease
was underestimated because this region is highly disordered in the fibrillar form of the peptide
(12). However, the amino-terminal domain of Aβ is selectively perturbed in amyloidogenesis,
and, most importantly, changes in its primary sequence trigger peptide assembly and fibril
formation (13,14). The importance of this domain is further supported by the finding that
antibodies against it are optimal for plaque clearance in animal models (15). A previous study
reported a distinct heterozygous APP mutation at codon 673 [Ala673Tyr673 (A673T)] in a
participant without clinical signs of dementia (16). Histological analysis did not detect amyloid
deposits in the brain. However, when the A673T mutation was introduced in a synthetic
Aβ1-40 peptide, it increased the propensity to aggregate, with a much shorter lag phase than
that of the wild-type peptide (17). These observations, together with our results, suggest that
mutations at position 2 of Aβ confer amyloidogenic properties that lead to AD only in the
homozygous state. The finding that the interaction between A673V-mutated and wild-type
Aβ hinders amyloidogenesis, and especially the anti-amyloidogenic properties of the mutated
six-residue peptide, may offer grounds for the development of therapeutic strategies based on
modified Aβ peptides or peptido-mimetic compounds (18,19) for both sporadic and familial
AD.
The present data highlight the importance of screening demented and nondemented human
populations for mutations of the Aβ encoding region of APP. Genetic variants that could be
regarded as normal polymorphisms may turn out to be pathogenic in homozygous individuals.
The identification of such mutations would help to prevent the occurrence of the disease in
their carriers.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
This work was supported by grants from the Italian Ministry of Health (533F/Q/1 to F.T. and M.S., and 71.6/2006
and RFPS 2007/02 to F.T.), CARIPLO Foundation (Guard) to F.T. and M.S., ERA-Net Neuron (nEUROsyn) to F.T.,
Negri-Weizmann Foundation to M.S., the National Institute of Neurological Disorders and Stroke (NS42029) to E.L.,
and the American Heart Association (0040102N) to E.L. A patent application related to this work has been filed by
Fondazione IRCCS Istituto Nazionale Neurologico “Carlo Besta.”
References and Notes
1. Hardy J, Selkoe DJ. Science 2002;297:353. [PubMed: 12130773]
Di Fede et al. Page 4
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
2. Selkoe DJ. Physiol. Rev 2001;81:741. [PubMed: 11274343]
3. Walsh DM, et al. Nature 2002;416:535. [PubMed: 11932745]
4. Bertram L, Tanzi RE. J. Clin. Investig 2005;115:1449. [PubMed: 15931380]
5. Rocchi A, Pellegrini S, Siciliano G, Murri L. Brain Res. Bull 2003;61:1. [PubMed: 12788204]
6. George-Hyslop PH. Biol. Psychiatry 2000;47:183. [PubMed: 10682216]
7. Levy E, et al. Science 1990;248:1124. [PubMed: 2111584]
8. Materials and methods are available as supporting material on Science Online.
9. Petersen RC, et al. Arch. Neurol 2001;58:1985. [PubMed: 11735772]
10. Cecchini M, Curcio R, Pappalardo M, Melki R, Caflisch A. J. Mol. Biol 2006;357:1306. [PubMed:
16483608]
11. Tomiyama T, et al. Ann. Neurol 2008;63:377. [PubMed: 18300294]
12. Williams AD, et al. J. Mol. Biol 2004;335:833. [PubMed: 14687578]
13. Lim KH. ChemBioChem 2006;7:1662. [PubMed: 17013984]
14. Hori Y, et al. J. Biol. Chem 2007;282:4916. [PubMed: 17170111]
15. Bard F, et al. Proc. Natl. Acad. Sci. U.S.A 2003;100:2023. [PubMed: 12566568]
16. Peacock ML, Warren JT, Roses AD, Fink JK. Neurology 1993;43:1254. [PubMed: 8170579]
17. Meinhardt J, et al. Protein Sci 2007;16:1214. [PubMed: 17525469]
18. Soto C, et al. Nat. Med 1998;4:822. [PubMed: 9662374]
19. Adessi C, et al. J. Biol. Chem 2003;278:13905. [PubMed: 12578830]
Di Fede et al. Page 5
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
Analysis of APP gene and APP processing. (A) APP gene analysis by restriction fragment
length polymorphism of 169—base pair (bp) polymerase chain reaction (PCR) products
amplified from homozygous (III-16), heterozygous (II-11), and control subjects. In the absence
of the A673V mutation, the enzyme HpyCH4V generates two fragments of 91 and 78 bp. The
mutation abolishes the restriction site, and the PCR product remains uncut. (B)
sAPPβ:sAPPα ratio in conditioned media from CHO cells transfected with wild-type or
A673V-mutated APP and fibroblasts of the proband and four controls. Error bars represent
means ± SD. (C) APP carboxy-terminal fragments C99 and C83 (arrowheads) in CHO cells
transfected with wild-type (lane 2) or A673V-mutated (lane 3) APP and fibroblasts from a
control (lane 4) and the proband (lane 5), as shown by immunoblot analysis. Lane 1 corresponds
to from nontransfected CHO cells. (D) Densitometric analysis of immunoblots, showing a
significant increase in the C99:C83 ratio (P = 0.0001) in cells carrying the A673V mutation.
Error bars represent means ± SD.
Di Fede et al. Page 6
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Short-time kinetics of Aβ assembly and disassembly, determined by laser light scattering.
(A) Course of the light intensity scattered by solutions of Aβ1-40wt (blue), Aβ1-40mut (red),
and their equimolar mixture (green). The corresponding exponential fits are indicated by full
lines. (B) Particle size distribution of Aβ1-40wt (blue), Aβ1-40mut (red), and the peptide mixture
(green) immediately after sample preparation (time 0) and after 24 hours. (C) Short-time
dissolution kinetics of 48-hour-aged peptide aggregates after fivefold dilution with buffer. a.u.,
arbitrary units.
Di Fede et al. Page 7
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 3.
Aggregation properties of mutated and wild-type Aβ peptides. (A to F) Electron micrographs
of aggregates generated by Aβ1-40wt, Aβ1-40mut, and equimolar mixtures after 72 hours [(A)
to (C), negative staining] and 20 days incubation [(D) to (F), positive staining]. (G to L)
Polarized light microscopy of Aβ aggregates stained with Congo red after 72 hours [(G) to (I)]
and 20 days [(J) to (L)]. (M to O) Electron micrographs of negatively stained aggregates
generated by Aβ1-42wt (M), Aβ1-42mut (N), and equimolar mixtures (O) after 5 days
incubation. The peptide mixture contains mainly amorphous material (O), whereas wild-type
and mutated Aβ1-42 are assembled in fibrillary structures. Scale bars indicate 250 nm [(A) to
(F)], 50 μm [(G) to (L)], and 125 nm [(M) to (O)].
Di Fede et al. Page 8
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 4.
Physicochemical and biological properties of mutated and wild-type Aβ peptides. (A) Size
exclusion chromatograms of Aβ1-40wt, Aβ1-40mut, and equimolar peptide mixture aggregates
after treatment with 1 M urea for 24 hours. Monomeric species (arrow) are only seen in the
peptide mixture. (B and C) Binding of wild-type and mutated Aβ1-40 (B) or Aβ1-6 (C) to
amyloid fibrils of Aβ1-40wt determined by surface plasmon resonance. Solutions of Aβ1-40
(1 μM) or Aβ1-6 (500 μM) were injected onto Aβ1-40wt fibrils immobilized on the sensor chip
for the time indicated by the bars. (D) Viability of human neuroblastoma cells after 24 hours
exposure to 5 μM Aβ1-42wt, Aβ1-42mut, and the equimolar mixture. Error bars represent SD
of the mean of eight replicates. *P = 0.026, **P < 0.001.
Di Fede et al. Page 9
Science. Author manuscript; available in PMC 2010 March 13.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... state and is protective in heterozygous state [50]. It induces a reduction of fibrillogenic properties of wild-type Aβ in the presence of Aβ A2V [50]. ...
... state and is protective in heterozygous state [50]. It induces a reduction of fibrillogenic properties of wild-type Aβ in the presence of Aβ A2V [50]. Thus potential therapies were developped using small synthetic peptides limited to the first amino acids of Aβ A2V [49]. ...
Article
Full-text available
The amyloid cascade hypothesis assumes that the development of Alzheimer’s disease (AD) is driven by a self-perpetuating cycle, in which β-amyloid (Aβ) accumulation leads to Tau pathology and neuronal damages. A particular mutation (A673T) of the amyloid precursor protein (APP) was identified among Icelandic population. It provides a protective effect against Alzheimer- and age-related cognitive decline. This APP mutation leads to the reduced production of Aβ with A2T (position in peptide sequence) change (Aβice). In addition, Aβice has the capacity to form protective heterodimers in association with wild-type Aβ. Despite the emerging interest in Aβice during the last decade, the impact of Aβice on events associated with the amyloid cascade has never been reported. First, the effects of Aβice were evaluated in vitro by electrophysiology on hippocampal slices and by studying synapse morphology in cortical neurons. We showed that Aβice protects against endogenous Aβ-mediated synaptotoxicity. Second, as several studies have outlined that a single intracerebral administration of Aβ can worsen Aβ deposition and cognitive functions several months after the inoculation, we evaluated in vivo the long-term effects of a single inoculation of Aβice or Aβ-wild-type (Aβwt) in the hippocampus of transgenic mice (APPswe/PS1dE9) over-expressing Aβ1-42 peptide. Interestingly, we found that the single intra-hippocampal inoculation of Aβice to mice rescued synaptic density and spatial memory losses four months post-inoculation, compared with Aβwt inoculation. Although Aβ load was not modulated by Aβice infusion, the amount of Tau-positive neuritic plaques was significantly reduced. Finally, a lower phagocytosis by microglia of post-synaptic compounds was detected in Aβice-inoculated animals, which can partly explain the increased density of synapses in the Aβice animals. Thus, a single event as Aβice inoculation can improve the fate of AD-associated pathology and phenotype in mice several months after the event. These results open unexpected fields to develop innovative therapeutic strategies against AD.
... 74 Whereas the APP mutation (alanine-673valine-673 [A673V]) affects APP processing only in the homozygous state, resulting in enhanced Aβ production and amyloid fibril formation in vitro, heterozygous carriers are not affected. 75 In addition, the APP intracellular domain (AICD) downregulates Wiskott-Aldrich syndrome protein (WASP)-family verprolin homologous protein 1, limiting Aβ production, which is another potential mechanism to regulate Aβ production. 76 The mutation of the APP gene is the most important risk factor of Aβ. ...
Article
Full-text available
Amyloid‐β (Aβ) is a pivotal biomarker in Alzheimer's disease (AD), attracting considerable attention from numerous researchers. There is uncertainty regarding whether clearing Aβ is beneficial or harmful to cognitive function. This question has been a central topic of research, especially given the lack of success in developing Aβ‐targeted drugs for AD. However, with the Food and Drug Administration's approval of Lecanemab as the first anti‐Aβ medication in July 2023, there is a significant shift in perspective on the potential of Aβ as a therapeutic target for AD. In light of this advancement, this review aims to illustrate and consolidate the molecular structural attributes and pathological ramifications of Aβ. Furthermore, it elucidates the determinants influencing its expression levels while delineating the gamut of extant Aβ‐targeted pharmacotherapies that have been subjected to clinical or preclinical evaluation. Subsequently, a comprehensive analysis is presented, dissecting the research landscape of Aβ across the domains above, culminating in the presentation of informed perspectives. Concluding reflections contemplate the supplementary advantages conferred by nanoparticle constructs, conceptualized within the framework of multivalent theory, within the milieu of AD diagnosis and therapeutic intervention, supplementing conventional modalities.
... Naturally occurring mutations offer evidence that N-terminal structure may influence phenotype. For instance, while the protective A2T mutation (Jonsson et al., 2012) decreases the hydrophobicity of the N-terminus, the A2V mutation (Di Fede et al., 2009) increases the amyloidogenesis and neurotoxicity of the mutated Aβ peptide. The increased hydrophobic character of the N-terminus increases the likelihood of β-sheet formation in the region, as evidenced computationally by reduced disorder and increased β-hairpin population in A2V-Aβ(1-28) (Nguyen et al., 2014). ...
... The β-secretase binding site in APP was located between APP Val669 and Ala673 with cleavage site between Met671 and Asp672 (Bagyinszky et al. 2019). Adjacent to p.M671L, 4 mutations were found in this area Met 670/671 Asn,Leu, the Val669Leu, the autosomal recessive Ala673Val and the protective Ala673Thr (Bagyinszky et al. 2019;Di Fede et al. 2009;Jonsson et al. 2012). The Met 670/671 Asn,Leu, the Val669Leu, the autosomal recessive Ala673Val were pathogenic to Alzheimer's disease by increasing the affinity of enzyme to APP and enhancing the amyloid aggregation (Bagyinszky et al. 2019). ...
Article
Full-text available
Familial Alzheimer’s disease (AD) is a rare disease caused by autosomal-dominant mutations. APP (encoding amyloid precursor protein), PSEN1 (encoding presenilin 1), and PSEN2 (encoding presenilin 2) are the most common genes cause dominant inherited AD. This study aimed to demonstrate a Chinese early-onset AD pedigree presenting as progressive memory impairment, apraxia, visual-spatial disorders, psychobehavioral disorders, and personality changes with a novel APP gene mutation. The family contains four patients, three carries and three normal family members. The proband underwent brain magnetic resonance imaging (MRI), ¹⁸F-fludeoxyglucose positron emission tomography (¹⁸F-FDG-PET), cerebrospinal fluid amyloid detection, ¹⁸F-florbetapir (AV-45) Positron Emission Computed Tomography (PET) imaging, whole-exome sequencing and Sanger sequencing. Brain MRI images showed brain atrophy, especially in the entorhinal cortex, temporal hippocampus, and lateral ventricle dilation. The FDG-PET showed hypometabolism in the frontotemporal, parietal, and hippocampal regions. ¹⁸F-florbetapir (AV-45) PET imaging showed cerebral cortex Aβ protein deposition. The cerebrospinal fluid amyloid protein test showed Aβ42/Aβ40 ratio decreases, pathological phosphor-tau level increases. Whole-exome sequencing detected a new missense mutation of codon 671 (M671L), which was a heterozygous A to T point mutation at position 2011 (c.2011A > T) in exon 16 of the amyloid precursor protein, resulting in the replacement of methionine to Leucine. The co-separation analysis was validated in this family. The mutation was found in 3 patients, 3 clinical normal members in the family, but not in the other 3 unaffected family members, 100 unrelated normal subjects, or 100 sporadic patients with AD. This mutation was probably pathogenic and novel in a Chinese Han family with early-onset AD.
Article
Alzheimer’s disease (AD) is one of the most unanswered diseases in the world as its complicated pathological mechanism makes it a formidable challenge in modern healthcare with limited intervention options. Due to its chronic nature in neurodegeneration, leading to cognitive decline and brain damage, mitigating this disease is now of major concern globally. The revelation of certain key hallmarks of AD pathology such as cholinergic dysfunction and amyloid plaque toxicity has thrown some insight into identifying therapeutic targets. Only symptomatic relief has been achieved by a single-target therapeutic approach, thus, the development of multi-impediment drugs is urgently needed. The adverse effects of current AD medication and repeated failure of futuristic drugs led us to hunt for a natural compound with beneficial properties that can target multi-facets of AD pathology, and cure this devastating brain disorder. The hypothesis of targeting different pathways contributing to progressive neurodegeneration in AD pathophysiology can be considered a new-age intervention. Coptisine, a bioactive alkaloid with benzyl tetrahydroisoquinoline in structure possesses enormous pharmacological benefits including neuroprotective abilities. Together with the potential of coptisine to inhibit the major targets of AD pathogenesis namely acetylcholinesterase (AChE), beta-secretase (BACE1), and gamma-secretase, this alkaloid can emerge as a new-age multi-target therapeutic for AD. However, robust research on coptisine’s suitability against AD pathogenesis is pivotal considering its therapeutic promises and an unambiguous understanding of the coptisine’s future can be predicted by evaluating its efficacy and safety for ameliorating AD.
Article
Full-text available
The “amyloid cascade” hypothesis of Alzheimer’s disease (AD) pathogenesis invokes the accumulation in the brain of plaques (containing the amyloid-β protein precursor [AβPP] cleavage product amyloid-β [Aβ]) and tangles (containing hyperphosphorylated tau) as drivers of pathogenesis. However, the poor track record of clinical trials based on this hypothesis suggests that the accumulation of these peptides is not the only cause of AD. Here, an alternative hypothesis is proposed in which the AβPP cleavage product C99, not Aβ, is the main culprit, via its role as a regulator of cholesterol metabolism. C99, which is a cholesterol sensor, promotes the formation of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a cholesterol-rich lipid raft-like subdomain of the ER that communicates, both physically and biochemically, with mitochondria. We propose that in early-onset AD (EOAD), MAM-localized C99 is elevated above normal levels, resulting in increased transport of cholesterol from the plasma membrane to membranes of intracellular organelles, such as ER/endosomes, thereby upregulating MAM function and driving pathology. By the same token, late-onset AD (LOAD) is triggered by any genetic variant that increases the accumulation of intracellular cholesterol that, in turn, boosts the levels of C99 and again upregulates MAM function. Thus, the functional cause of AD is upregulated MAM function that, in turn, causes the hallmark disease phenotypes, including the plaques and tangles. Accordingly, the MAM hypothesis invokes two key interrelated elements, C99 and cholesterol, that converge at the MAM to drive AD pathogenesis. From this perspective, AD is, at bottom, a lipid disorder.
Article
Full-text available
Fibril formation resulting from protein aggregation is a hallmark of a large group of neurodegenerative human diseases, including Alzheimer's disease, type 2 diabetes, amyotrophic lateral sclerosis, and Parkinson's disease, among many others. Key factors governing protein fibril formation have been identified over the past decades to elucidate various facets of misfolding and aggregation. However, surprisingly little is known about how and why fibril structure is achieved, and it remains a fundamental problem in molecular biology. In this review, we discuss the relationship between fibril formation kinetics and various characteristics, including sequence, mutations, monomer secondary structure, mechanical stability of the fibril state, aromaticity, hydrophobicity, charge, and population of fibril-prone conformations in the monomeric state.
Article
Full-text available
An amyloid protein that precipitates in the cerebral vessel walls of Dutch patients with hereditary cerebral hemorrhage with amyloidosis is similar to the amyloid protein in vessel walls and senile plaques in brains of patients with Alzheimer's disease, Down syndrome, and sporadic cerebral amyloid angiopathy. Cloning and sequencing of the two exons that encode the amyloid protein from two patients with this amyloidosis revealed a cytosine-to-guanine transversion, a mutation that caused a single amino acid substitution (glutamine instead of glutamic acid) at position 22 of the amyloid protein. The mutation may account for the deposition of this amyloid protein in the cerebral vessel walls of these patients, leading to cerebral hemorrhages and premature death.
Article
Full-text available
The field of aging and dementia is focusing on the characterization of the earliest stages of cognitive impairment. Recent research has identified a transitional state between the cognitive changes of normal aging and Alzheimer's disease (AD), known as mild cognitive impairment (MCI). Mild cognitive impairment refers to the clinical condition between normal aging and AD in which persons experience memory loss to a greater extent than one would expect for age, yet they do not meet currently accepted criteria for clinically probable AD. When these persons are observed longitudinally, they progress to clinically probable AD at a considerably accelerated rate compared with healthy age-matched individuals. Consequently, this condition has been recognized as suitable for possible therapeutic intervention, and several multicenter international treatment trials are under way. Because this is a topic of intense interest, a group of experts on aging and MCI from around the world in the fields of neurology, psychiatry, geriatrics, neuropsychology, neuroimaging, neuropathology, clinical trials, and ethics was convened to summarize the current state of the field of MCI. Participants reviewed the world scientific literature on aging and MCI and summarized the various topics with respect to available evidence on MCI. Diagnostic criteria and clinical outcomes of these subjects are available in the literature. Mild cognitive impairment is believed to be a high-risk condition for the development of clinically probable AD. Heterogeneity in the use of the term was recognized, and subclassifications were suggested. While no treatments are recommended for MCI currently, clinical trials regarding potential therapies are under way. Recommendations concerning ethical issues in the diagnosis and the management of subjects with MCI were made.
Article
Rapid progress in deciphering the biological mechanism of Alzheimer's disease (AD) has arisen from the application of molecular and cell biology to this complex disorder of the limbic and association cortices. In turn, new insights into fundamental aspects of protein biology have resulted from research on the disease. This beneficial interplay between basic and applied cell biology is well illustrated by advances in understanding the genotype-to-phenotype relationships of familial Alzheimer's disease. All four genes definitively linked to inherited forms of the disease to date have been shown to increase the production and/or deposition of amyloid β-protein in the brain. In particular, evidence that the presenilin proteins, mutations in which cause the most aggressive form of inherited AD, lead to altered intramembranous cleavage of the β-amyloid precursor protein by the protease called γ-secretase has spurred progress toward novel therapeutics. The finding that presenilin itself may be the long-sought γ-secretase, coupled with the recent identification of β-secretase, has provided discrete biochemical targets for drug screening and development. Alternate and novel strategies for inhibiting the early mechanism of the disease are also emerging. The progress reviewed here, coupled with better ability to diagnose the disease early, bode well for the successful development of therapeutic and preventative drugs for this major public health problem.
Article
Application of genetic paradigms to Alzheimer's disease (AD) has led to confirmation that genetic factors play a role in this disease. Additionally, researchers now understand that AD is genetically heterogeneous and that some genetic isoforms appear to have similar or related biochemical consequences. Genetic epidemiologic studies indicate that first-degree relatives of AD probands have an age-dependent risk for AD approximately equal to 38% by age 90 years (range 10% to 50%). This incidence strongly suggests that transmission may be more complicated than a simple autosomal dominant trait. Nevertheless, a small proportion of AD cases with unequivocal autosomal dominant transmission have been identified. Studies of these autosomal dominant familial AD (FAD) pedigrees have thus far identified four distinct FAD genes. The beta-amyloid precursor protein (beta APP) gene (on chromosome 21), the presenilin 1 (PS1) gene (on chromosome 14), and the presenilin 2 (PS2) gene (on chromosome 1) gene are all associated with early-onset AD. Missense mutations in these genes cause abnormal beta APP processing with resultant overproduction of A beta 42 peptides. In addition, the epsilon 4 allele of apolipoprotein E (APOE) is associated with a increased risk for late-onset AD. Although attempts to develop symptomatic treatments based on neurotransmitter replacement continue, some laboratories are attempting to design treatments that will modulate production or disposition of A beta peptides.
Article
Inhibition of cerebral amyloid -protein deposition seems to be an important target for Alzheimer's disease therapy. Amyloidogenesis could be inhibited by short synthetic peptides designed as -sheet breakers. Here we demonstrate a 5-residue peptide that inhibits amyloid protein fibrillogenesis, disassembles preformed fibrils in vitro and prevents neuronal death induced by fibrils in cell culture. In addition, the -sheet breaker peptide significantly reduces amyloid protein deposition in vivo and completely blocks the formation of amyloid fibrils in a rat brain model of amyloidosis. These findings may provide the basis for a new therapeutic approach to prevent amyloidosis in Alzheimer's disease.
Article
We found a novel polymorphism in the amyloid precursor protein (APP) gene in a patient with ischemic cerebrovascular disease who had no evidence of Alzheimer's disease (AD). This polymorphism deletes a Fok I restriction enzyme site and causes the substitution of threonine for alanine at codon 673. This is adjacent to the site at which APP is thought to undergo cleavage in AD. Analysis of this polymorphism may provide insight into the basis of APP processing.
Article
Inhibition of cerebral amyloid beta-protein deposition seems to be an important target for Alzheimer's disease therapy. Amyloidogenesis could be inhibited by short synthetic peptides designed as beta-sheet breakers. Here we demonstrate a 5-residue peptide that inhibits amyloid beta-protein fibrillogenesis, disassembles preformed fibrils in vitro and prevents neuronal death induced by fibrils in cell culture. In addition, the beta-sheet breaker peptide significantly reduces amyloid beta-protein deposition in vivo and completely blocks the formation of amyloid fibrils in a rat brain model of amyloidosis. These findings may provide the basis for a new therapeutic approach to prevent amyloidosis in Alzheimer's disease.
Article
Rapid progress in deciphering the biological mechanism of Alzheimer's disease (AD) has arisen from the application of molecular and cell biology to this complex disorder of the limbic and association cortices. In turn, new insights into fundamental aspects of protein biology have resulted from research on the disease. This beneficial interplay between basic and applied cell biology is well illustrated by advances in understanding the genotype-to-phenotype relationships of familial Alzheimer's disease. All four genes definitively linked to inherited forms of the disease to date have been shown to increase the production and/or deposition of amyloid beta-protein in the brain. In particular, evidence that the presenilin proteins, mutations in which cause the most aggressive form of inherited AD, lead to altered intramembranous cleavage of the beta-amyloid precursor protein by the protease called gamma-secretase has spurred progress toward novel therapeutics. The finding that presenilin itself may be the long-sought gamma-secretase, coupled with the recent identification of beta-secretase, has provided discrete biochemical targets for drug screening and development. Alternate and novel strategies for inhibiting the early mechanism of the disease are also emerging. The progress reviewed here, coupled with better ability to diagnose the disease early, bode well for the successful development of therapeutic and preventative drugs for this major public health problem.
Article
Although extensive data support a central pathogenic role for amyloid beta protein (Abeta) in Alzheimer's disease, the amyloid hypothesis remains controversial, in part because a specific neurotoxic species of Abeta and the nature of its effects on synaptic function have not been defined in vivo. Here we report that natural oligomers of human Abeta are formed soon after generation of the peptide within specific intracellular vesicles and are subsequently secreted from the cell. Cerebral microinjection of cell medium containing these oligomers and abundant Abeta monomers but no amyloid fibrils markedly inhibited hippocampal long-term potentiation (LTP) in rats in vivo. Immunodepletion from the medium of all Abeta species completely abrogated this effect. Pretreatment of the medium with insulin-degrading enzyme, which degrades Abeta monomers but not oligomers, did not prevent the inhibition of LTP. Therefore, Abeta oligomers, in the absence of monomers and amyloid fibrils, disrupted synaptic plasticity in vivo at concentrations found in human brain and cerebrospinal fluid. Finally, treatment of cells with gamma-secretase inhibitors prevented oligomer formation at doses that allowed appreciable monomer production, and such medium no longer disrupted LTP, indicating that synaptotoxic Abeta oligomers can be targeted therapeutically.
Article
It has been more than 10 years since it was first proposed that the neurodegeneration in Alzheimer's disease (AD) may be caused by deposition of amyloid ??-peptide (A??) in plaques in brain tissue. According to the amyloid hypothesis, accumulation of A?? in the brain is the primary influence driving AD pathogenesis. The rest of the disease process, including formation of neurofibrillary tangles containing tau protein, is proposed to result from an imbalance between A?? production and A?? clearance.