Tài liệu Báo cáo khoa học: Animal models of amyloid-b-related pathologies in Alzheimer’s disease - Pdf 10

REVIEW ARTICLE
Animal models of amyloid-b-related pathologies in
Alzheimer’s disease
Ola Philipson
1
, Anna Lord
2
, Astrid Gumucio
1
, Paul O’Callaghan
1
, Lars Lannfelt
1
and Lars
N.G. Nilsson
1
1 Department of Public Health and Caring Sciences ⁄ Molecular Geriatrics, Uppsala University, Sweden
2 BioArctic Neuroscience AB, Stockholm, Sweden
Introduction
Alzheimer’s disease (AD) accounts for  60–70% of
all dementia cases. Prevalence increases with age from
 1% in the 60 to 64-year age group, to 24–33% in
those aged > 85 years. There is an insidious onset
with an initial loss of short-term memory, followed by
progressive impairment of multiple cognitive functions
that affect the activities of daily living. The AD diag-
nosis is based on a patient’s medical history, neurolog-
ical assessment and neuropsychiatric testing of
cognitive functions. Neuroimaging techniques and
biomarkers in cerebrospinal fluid (CSF) are invaluable
in differential diagnosis.

doi:10.1111/j.1742-4658.2010.07564.x
In the early 1990s, breakthrough discoveries on the genetics of Alzheimer’s
disease led to the identification of missense mutations in the amyloid-b
precursor protein gene. Research findings quickly followed, giving insights
into molecular pathogenesis and possibilities for the development of new
types of animal models. The complete toolbox of transgenic techniques,
including pronuclear oocyte injection and homologous recombination, has
been applied in the Alzheimer’s disease field, to produce overexpressors,
knockouts, knockins and regulatable transgenics. Transgenic models have
dramatically advanced our understanding of pathogenic mechanisms and
allowed therapeutic approaches to be tested. Following a brief introduction
to Alzheimer’s disease, various nontransgenic and transgenic animal models
are described in terms of their values and limitations with respect to patho-
genic, therapeutic and functional understandings of the human disease.
Abbreviations
AD, Alzheimer’s disease; ApoE, apolipoprotein E; APP, amyloid-b precursor protein; Ab, Amyloid-b; BACE-1, b-site APP cleaving enzyme-1;
CAA, cerebral amyloid angiopathy; CCR2, chemokine (C-C motif) receptor 2; CSF, cerebrospinal fluid; MWM, Morris water maze; NFTs,
neurofibrillary tangles; PDGF, platelet-derived growth factor; PS, presenilin; SMC, smooth muscle cells; wt, wild-type.
FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS 1389
lack b-sheet structure and are therefore by definition
not amyloid. Cerebral amyloid angiopathy (CAA)
results in the degeneration of vessel walls and hemor-
rhages. CAA is found in  80% of AD brains, but is
not a diagnostic criterion. NFTs are intracellular fila-
mentous lesions with amyloid properties. They contain
hyperphosphorylated and aggregated forms of tau, a
microtubule-associated protein that normally serves to
assemble and stabilize microtubules.
Genetics and risk factors implicated in
Alzheimer’s disease pathogenesis

AD neuropathology, and most experience dementia by
60–70 years of age [9].
The major genetic risk factor for developing late-
onset AD is the apolipoprotein E (ApoE) e4 allele
[10,11]. One ApoE e4 allele increases the risk of AD by
two- to threefold, and two e4 alleles confer a 12-fold
increase in risk. In the brain, ApoE is primarily synthe-
sized by astrocytes and serves to regulate the transport
of cholesterol-containing lipoprotein particles. ApoE
binds to Ab and becomes a component of amyloid in
AD senile plaques. The pathogenic mechanism of
ApoE likely relates to altered deposition and ⁄ or clear-
ance of Ab in the brain, although the details are still
not fully understood [12]. A large number of other dis-
ease-related loci and candidate genes have been pro-
posed, but not generally verified, indicating that these
genes have a modest impact on the pathogenesis. The
major risk factors for AD are age and a family history
of the disease. Low education or cognitive reserve
capacity, female gender, head trauma, hypertension,
cardiovascular disease and a high-cholesterol diet are
proposed risk factors for AD [13] (Fig. 2).
Nontransgenic animal models
Based on the cholinergic hypothesis, scopolamine-
induced amnesia, excitotoxic lesions of the basal
forebrain and aged primates have been used to assess
cognitive deficits. Current symptomatic drugs for AD
were successfully evaluated in these models, but their
etiological relevance is low [14]. Nontransgenic rodents
Fig. 1. Disease-causing APP mutations used in transgenic models. The Swedish mutation (1) favors b-secretase (b) cleavage, while the

tion, spatial disorientation and disturbed diurnal
rhythm. Cognitive dysfunction in old dogs is associated
with diffuse Ab deposits [20], neuritic dystrophy and gli-
osis, but few amyloid plaques and no NFTs. Ventricular
dilation, cortical and hippocampal atrophy, CAA with
degeneration of smooth muscle cells (SMC) and hemor-
rhages can all be found in aged canine brain. Interest in
nonhuman primate models has grown following the fail-
ure to predict meningoencephalitis as a side-effect of the
AN1792 vaccination trial from transgenic studies. The
efficacy and safety of an Ab vaccine has been tested in
the Carribean vervet monkey [21]. Alternatives are aged
lemurs [22], cotton-top tamarins [23], rhesus monkeys
[24] or squirrel monkeys [25]. An aged chimpanzee with
complete AD neuropathology, including neuritic
plaques and paired helical filament-containing NFTs,
was recently reported [26].
Fig. 2. AD pathogenesis according to the
amyloid cascade hypothesis. This theory
suggests that altered metabolism of Ab,in
particular aggregation-prone Ab species like
Ab42, initiates AD pathogenesis. Oligomeric
assemblies of Ab trigger aggregation of tau
and the formation of NFTs, but also inflam-
mation and oxidative stress, by rather
unclear mechanisms. These downstream
processes give rise to progressive neurode-
generation, which ultimately results in
dementia. The main pathogenic pathway of
AD is illustrated with red arrows, whereas

aggregates or amyloid deposists were found in the pan-
creas [28], intestine [29] or skeletal muscles [30]. The
level of plasma Ab in C99-based models was similar to
Tg2576, an APP transgenic model with high peripheral
promoter activity.
In an alternative strategy, a yeast artificial chromo-
some, harboring the whole APPwt gene, was used to
maintain transcriptional regulation, alternative splicing
and normal APP processing. In these Py8.9 mice,
proper APP protein synthesis and alternative splicing
was demonstrated, but the brain was devoid of neuro-
pathology and the levels of Ab were low [31]. How-
ever, when wild-type human APP was expressed at
very high levels, under the Thy1 promoter, sparse
parenchymal and vascular amyloid deposits were
found in aged mice [32]. Thus a pathogenic APP muta-
tion is not a prerequisite for amyloid deposition.
Instead it seems to depend upon producing sufficient
Ab levels in the brain to ensure fibrillization. To
explore the pathogenic impact of individual Ab spe-
cies, a fusion protein, BRI–wt-Ab42, was designed
from which Ab was released by furin-like enzymes on
the cell surface. BRI is a transmembrane protein that
is involved in amyloid deposition in British familial
dementia. The fusion design permitted the synthesis of
high Ab levels in the brain in a manner similar to APP
transgenic mice, but in the absence of APP overexpres-
sion. Transgenic mice expressing BRI–wt-Ab42 devel-
oped extensive vascular and parenchymal amyloid
pathology, accompanied by dystrophic neurites and

751
and APP
770
, with strong and
selective neuronal expression was enabled by the plate-
let derived growth factor (PDGF)b promoter. Impor-
tantly, young PDAPP mice produced high Ab42 levels
in the brain, particularly in the hippocampus. The ani-
mals preferentially accumulated Ab42 peptides and
developed senile plaques, but also a substantial num-
ber of diffuse Ab deposits at 9–10 months of age [37].
Plaque formation began in the cingulate cortex and
was accompanied by phospho-tau immunoreactive
dystrophic neurites, synaptic loss and gliosis in the
adjacent tissue, but not by overt neuronal loss [38,39].
Ultrastructural analyses revealed neurons in close
proximity to senile plaques and amyloid fibrils. The
latter had a diameter of 9–11 nm and were surrounded
by neuronal membranes and vesicles [40]. Young
PDAPP mice showed deficits in spatial learning and
memory, which worsened with increasing age and Ab
burden, although their performance in a novel
object-recognition task was unimpaired [41]. By
contrast, others found age-dependent deficits in object
recognition and place learning impairments that were
independent of age [42]. These discrepancies could be
because of differences in experimental procedures or
unintentional genetic drift of mouse colonies. PDAPP
mice are typically bred on a mixed genetic background
(Swiss Webster, DBA ⁄ 2 and C57Bl ⁄ 6). Hippocampal

extracts. Ab42 could still have initiated deposition of
Ab in vessels, because some focal lesions were only
Ab42-immunoreactive [45]. By contrast, brains from
patients with the London mutation contained mainly
Ab-immunoreactive plaques and cytoskeletal pathol-
ogy, but only modest or little CAA [46]. Thus, the
CAA phenotype in the transgenic mice might not have
been caused by the London mutation. Instead it may
be the result of strong APP expression, advanced age,
strain background (FVB ⁄ N) and ⁄ or differences in APP
processing between species.
Models with the Swedish APP mutation
The Swedish mutation (KM670 ⁄ 671NL) is located just
outside the N-terminus of the Ab domain in APP. It
was identified in 1992 [47] and shown to increase Ab
levels by six- to eightfold [4]. These discoveries created
intense interest in APP processing and paved the way
for the development of more sophisticated ELISAs to
selectively measure Ab40 and Ab42 [48]. Later, the
Swedish mutation became essential in the identification
and characterization of BACE-1 [49]. The clinical and
neuropathological features associated with the Swedish
mutation are those of typical AD [47,50]. Tg2576
mice, the most frequently used APP transgenic model,
harbor the Swedish mutation and display both AD-like
Ab neuropathology and cognitive deficits [51]. The
Swedish mutation redirects APP processing to secre-
tory vesicles en route to the cell surface in cell culture
[52], whereas APPwt is largely processed in recycling
endosomes [53]. This difference may be largely irrele-

around the murine Ab domain was replaced with the
human Ab sequence and the Swedish mutation. The
MoPrP.Xho vector was much smaller than the cos-
SHaPrP, and selectively directed twofold overexpres-
sion of APP to the brain [58,59]. In an even more
refined strategy, the murine Ab sequence was human-
ized and the Swedish mutation introduced with gene
targeting. In this knockin model, APP
NLh ⁄ NLh
, only
five single amino acids were altered in the entire mur-
ine genome. Consequently, APP protein synthesis
remained unchanged in terms of its spatial and tempo-
ral expression pattern and mRNA localization. The
Swedish mutation led to markedly enhanced b-secre-
tase activity and a ninefold increase in Ab level,
compared with normal aged human brain [60]. By
22 months of age, APP
NLh ⁄ NLh
mice had not devel-
oped Ab neuropathology [61], but young mice were
elegantly used to estimate the turnover of Ab, APP
and APP fragments in vivo [62]. In another genomic
approach, a 650 kb yeast artificial chromosome vector
harboring the whole human APP gene locus with the
O. Philipson et al. Animal models of Alzheimer’s disease
FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS 1393
Swedish mutation was used. In the homozygous mice
(R.1.40), in which Ab42 levels were 15 to 20-fold
higher than in mice expressing wild-type human APP,

mutations
Patients never inherit multiple pathogenic mutations in
APP, presenilin, tau or a-synuclein genes, nor do they
overexpress chimeric APP mRNA under a heterolo-
gous promoter. Thus none of the transgenic models
fully mimic the genetics of familial AD. By combining
genetic lesions one can accelerate Ab aggregation and
lower the cost of research. One can also confer certain
characteristics to Ab and dissect molecular interac-
tions. The Swedish mutation has often been used
together with other mutations in transgenic models
because it is located outside the Ab domain and serves
to enhance Ab levels.
No Ab pathology was evident at 24 months of age
in J.1.96 homozygous transgenic mice when a genomic
vector with both the Swedish and London mutations
was introduced, despite life-long exposure to a four- to
sixfold increased levels of Ab42, compared to human
APPwt [64]. In contrast, APP22 mice presented with
diffuse Ab deposits and few amyloid plaques when the
mutations were combined in a cDNA-strategy to
create twofold overexpression under the human Thy1
promoter [65].
The Tg-CRND8 model was designed by inserting
human APP
695
with Swedish and Indiana mutations in
the cosSHaPrP vector [55]. It resulted in an aggressive
neuropathology with onset of amyloid deposition and
place learning impairment as early as 3 months of age.

from  8 weeks of age, a consequence of 10 to 30-fold
increased APP expression and two pathogenic APP
mutations. APP transgene expression was suppressed
> 95% when 4-week-old mice were given doxycycline
for 2 weeks. Relative to doxycycline-free mice, Ab in
PBS- and SDS-soluble pools were efficiently cleared,
although Ab42 partially remained in the formic acid
soluble pool. By contrast, brains of animals reared on
doxycycline from birth to 6 weeks of age contained
essentially no human Ab, suggesting a very early onset
of Ab aggregation and a tightly controlled transgene
expression with little leakage. Interestingly, suppression
of the transgene in 6-month-old mice arrested amyloid
deposition, but did not promote clearance. Moreover,
astrogliosis and ubiquitin-positive dystrophic neurites
in the vicinity of senile plaques were unchanged. Thus,
the endogenous clearance systems were unable to
Animal models of Alzheimer’s disease O. Philipson et al.
1394 FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS
eliminate existing Ab aggregates and secondary pathol-
ogy, at least in this aggressive transgenic model [72].
Models with the Flemish, Arctic, Dutch or Iowa
APP mutation inside the Ab domain
Mutations at positions 21–23 in the Ab domain of
APP, near the hydrophobic cluster, are a heterogeneous
group of genetic lesions. They affect Ab aggregation
and degradation, but also APP processing. The Dutch
(E693Q) [73] and Iowa (D694N) [74] mutations are
associated with CAA and diffuse Ab deposits, resulting
in hemorrhagic strokes and ⁄ or infarcts and dementia.

The microvascular pathology was accompanied by
astro- and microgliosis as well as increased levels of
proinflammatory cytokines.
The Flemish (A692G) [78] mutation can start either
with presenile dementia or CAA. In contrast to the
other intra-Ab mutations, it makes APP an inferior
substrate for a-secretase and increases Ab levels
[79,80]. APP cleavage by b-secretase was favored in
transgenic mice with the Flemish mutation (APP ⁄ Fl),
with modestly increased Ab1-40 levels. There was
spongiosis and gliosis in APP ⁄ Fl mice, but no Ab or
tau pathology. Male APP ⁄ Fl mice, which were bred on
the FVB background, were aggressive and suffered
premature death and seizures. APP expression was
likely insufficient to generate neuropathology and it is
unclear if the findings in APP ⁄ Fl were specifically
caused by the Flemish mutation. An APP ⁄ Du model,
developed in parallel, displayed similar phenotypes
[81].
The Arctic APP mutation (E693G) [79] is associated
with clinical features of early-onset AD commencing at
52–62 years. There are NFTs, severe CAA in the
absence of hemorrhage and an abundance of paren-
chymal Ab deposits lacking amyloid cores in postmor-
tem brain [82]. The Arctic mutation promotes Ab
protofibril and fibril formation, but also favors intra-
cellular b-secretase processing of APP [79,83,84].
Tg-ArcSwe models with both the Swedish and Arctic
mutation were developed by two independent groups.
In young mice, the Arctic mutation increased intraneu-

mice, amyloid deposition was markedly accelerated in
O. Philipson et al. Animal models of Alzheimer’s disease
FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS 1395
bigenic PS-1 · APP transgenic mice [91,92]. Borchelt
et al. generated PS-1 transgenic models expressing
mutant protein (M146L, A246E or PS1DE9), which
were cross-bred with APP transgenic mice with the
Swedish mutation, line C3-3 [58,91]. Other researchers
cross-bred Tg2576 with PS-1 transgenic mice, in which
PS-1 cDNA (M146L or M146V) had been linked to
the PDGFb2 promoter [90], resulting in the PSAPP
model [92]. PS2APP mice, generated by crossing PS2
(N141I) and APP-Swe, also displayed an aggressive
Ab pathology with age-dependent spatial learning and
memory deficits [93].
Autosomal dominant mutations in tau causing
frontotemporal lobe dementia were quickly utilized for
transgenic experiments. The JNPL3 model, expressing
4R0N tau isoform with the P301L mutation, was the
first model with Gallyas-positive NFTs. When double-
transgenic mice were created by cross-breeding with
Tg2576 a more complete AD neuropathology was
generated. Moreover tau pathology, but not Ab patho-
logy, was enhanced in these mice, suggesting that effects
on tau are downstream of Ab in AD pathogenesis [94]
(Fig. 2). However triple-transgenic mice, which were
generated by crossing mice producing the wild-type tau
isoform (3R0N) with mice carrying the Swedish and
London APP mutations and a PS-1 mutation (M146L),
only resulted in somadendritic accumulation of tau and

in 12 to 15-month-old mice, whereas paired helical
filament-1 immunoreactivity and Gallyas staining,
which indicate NFT formation, were not seen until
18 months of age [98]. In more recent studies of this
model, amyloid deposition commenced at  15 months
in the hippocampus and was widespread > 18 months
[99]. Triple-transgenic mice have been elegantly used to
study interactions between Ab and tau pathologies and
their impact on phenotypes of synaptic and cognitive
dysfunction [100,101].
Advanced animal models have recently been gener-
ated in which neuronal degeneration is clearly evident.
In the 5xFAD transgenic model, Thy1 promoter-driven
transgenes of APP (with the Swedish, Florida and
London AD mutations) and PS-1 (with the AD muta-
tions M146L and L286V) were coinjected into pronu-
clei of C57BL ⁄ 6xSJL mice. The model was made in an
effort to alter the Ab42 ⁄ Ab40 ratio in favor of Ab42
synthesis ([102] and references therein). Indeed the
strategy resulted in a high level of Ab42 and an
Ab42 ⁄ Ab40 synthesis ratio of 25 : 1 in young mice, in
comparison with 0.1–0.2 : 1 in Tg2576 mice with only
the Swedish APP mutation. Amyloid deposits formed
within 2 months, and the mice also developed intran-
euronal Ab aggregates. The intraneuronal deposits
were in a b-pleated sheet conformation, and located to
large pyramidal neurons of cerebral cortex layer V.
Interestingly, in 9-month-old 5xFAD-mice there was a
selective loss of these neurons and a decrease of several
synaptic markers. Importantly, these phenotypes and

CSF
⁄ [Ab]
plasma
should be the same in all trans-
genic models. A much higher plasma Ab level in
Tg2576 mice than in APP23, but a comparable
central nervous system Ab level, is inconsistent with
the idea of a dynamic equilibrium. The higher plasma
Ab levels in Tg2576 is most likely explained by the
stronger peripheral activity of the hamster PrP
promoter, compared with the neuron-specific Thy1
promoter in APP23. This emphasises the influence of
promoter selection on differential expression patterns
of APP and steady-state Ab levels in the central
nervous system and in peripheral tissue; consequently,
interpretations from transgenic models regarding Ab
dynamics should be made with caution.
In the 1980s it was debated whether Ab amyloid
deposits, and in particular CAA at the cerebral vessel
wall, had a central nervous system or a peripheral
source. Models driven by the Thy1 promoter, like
APPDutch transgenic mice, with almost exclusive
neuronal central nervous system expression of APP
develop almost only CAA, but by introducing a
presenilin transgene and raising the ratio of
Ab42 ⁄ Ab40 synthesis instead mainly parenchymal
senile plaques develop [32]. By contrast, models with
peripheral APP synthesis and high plasma Ab levels
present with amyloid deposits in peripheral organs
and neither CAA nor Ab plaques are found in the

1–40
. A density 1 gÆL
)1
for brain tissue has been assumed when the ratio of brain Ab ⁄ plasma Ab
has been calculated. Thus, for example, 20 pmolÆg
)1
 20 nM. Studies on the initial description and on metabolic levels of brain, CSF and plasma Ab that were used as sources of informa-
tion are cited. APP, amyloid-b precursor protein; CAA, cerebral amyloid angiopathy; NFT, neurofibrillary tangles; PDGF, platelet-derived growth factor.
Model Transgene Promoter Neuropathology
Age-of
-onset
(months)
Brain
Ab
(pmolÆg
)1
)
Plasma
Ab
Ratio
brain Ab ⁄
plasma Ab
CSF
Ab
Ratio
brain Ab ⁄
CSF Ab Contact
PDAPP [36,37,105] APP minigene V717F
(Indiana)
PDGF Diffuse and neuritic

CAA NFTs, atrophy
20 90 p
M 220 1.6 nM 12
O. Philipson et al. Animal models of Alzheimer’s disease
FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS 1397
formed very rapidly, within 1–2 days, reaching a size
that was surprisingly stable. Within 1 week, early
changes were accompanied by the recruitment of
reactive microglia, and shortly thereafter by neuritic
dystrophy [113]. However, in a subsequent study,
plaque growth occurred over a period of weeks when a
thinned-skull cranial window was used instead [114].
There is also a local neurotoxic effect on nerve endings
near amyloid plaques in APP transgenic models and in
AD postmortem brain [115], whereby dendritic spines
decrease in density but do not change in structure
[116]. Loss of CA1 pyramidal neurons in the hippo-
campus was reported in aged APP23 mice with a high
plaque load [117], although subtle cell loss is difficult
to distinguish from physical displacement. Today,
almost every research article in the AD field contains
an introductory statement in which the neurotoxicity
of Ab is described as a well-established fact, yet analy-
ses of mouse brain in which large amounts of Ab have
accumulated provide no or very sparse support for this
hypothesis. Neurodegenerative mechanisms of proteop-
athies are still largely unknown. Perhaps the neurotox-
icity is sparse because APP trafficking and subcellular
Ab accumulation in AD brain is poorly mimicked in
most models, as chimeric APP mRNAs are overexpres-

expressed under the glial fibrillary acidic protein
promoter in PDAPP mice lacking murine ApoE.Ab
burden was then accelerated by the risk allele ApoE e4
and decelerated by the protective allele ApoE e2, rela-
tive to the ApoE e3 allele [120]. These findings fit well
with observations in postmortem AD brain [121].
However, although murine ApoE facilitates Ab deposi-
tion in a gene-dose-dependent manner, human ApoE
decelerates Ab deposition compared with murine
ApoE [122]. This may be because a human transgene
was introduced into a complex feedback network
involving murine lipoprotein receptors. Alternatively,
ApoE may affect both Ab clearance and deposition. It
illustrates the complexity of detailed mechanistic stud-
ies. Deletion of apolipoprotein J, which also binds to
Ab, decelerated amyloid formation [123], whereas abla-
tion of both ApoE and apolipoprotein J strongly
increased Ab deposition [124]. Possibly the lipoprotein
metabolism in the brain is altered when two abun-
dantly expressed apolipoproteins, E and J, are both
absent. Lipidation of ApoE-containing lipoparticles via
the ATP-binding cassette family of active transporters
regulates Ab deposition. PDAPP mice overexpressing
murine ATP-binding cassette family of active transport-
ers 1 are phenotypically similar to those devoid of
murine ApoE. In contrast, Ab and amyloid deposition
is accelerated in APP transgenic mice devoid of ATP-
binding cassette family of active transporters 1 ([12] and
references therein).
Neuroinflammation has been suggested to influence

play a role in Ab biology,
and possibly in AD pathogenesis. Both ions bind APP
and Ab with high affinity, and stimulate Ab aggrega-
tion and oxidizing effects of Ab in vitro. By deleting
the gene encoding a zinc transporter, ZnT3, the endog-
enous pool of synaptic Zn
2+
was depleted. Senile
plaque deposition was then markedly decelerated in
Tg2576 mice in a dose-dependent manner, whereas
soluble Ab was modestly increased. Synaptic zinc and
ZnT3 changed in response to ovariectomy and estro-
gen replacement possibly explaining why increased Ab
burden is often observed in female APP transgenic
mice [57,135,136].
As described above, knowledge on pathogenic
mechanisms has often been gained by removing or
expressing normal or mutant genes and examining
APP ⁄ Ab-related phenotypes. A major caveat when
interpreting the results from a cross-breeding experi-
ment is the influence of the genetic background. This
problem can be circumvented by expressing a trans-
gene at a specific location in the brain with a lentiviral
vector [137]. The relevance of cross-breeding experi-
ments is strengthened if expression and deletion of the
gene(s) results in the opposite outcome, or if the results
are substantiated by in vitro studies or analyses of
clinical samples. The ability to track pathogenic events
in living animals with intravital multiphoton micros-
copy, small animal PET cameras and microdialysis

placebo-treated groups can then lead to a systematic
error that is mistakenly interpreted as evidence of ther-
apeutic efficacy. Many APP transgenic models also
suffer spontaneous death, which drives selection in a
cohort of animals by unknown mechanisms. Conse-
quently, a drug under investigation might modulate
spontaneous death and not AD-pathogenesis. Ran-
domizing experimental groups, matching them for gen-
der, and having sufficient power are therefore
extremely important in minimizing the influence of
confounding factors arising from inherent problems
associated with breeding APP transgenic mice and
maintaining stable phenotypes. We would argue that a
single report that has not been replicated, preferably
by other researchers, provides weak evidence of thera-
peutic efficacy.
Knowledge of the pathway one intends to target
and the pharmacological mechanism of the drug are
equally important. In vitro and in vivo pharmacology
of the drug should preferably be carried out before
efficacy is tested in APP transgenic mice. Unfortu-
nately, many drug candidates or dietary supplements
are simply directly tested for in vivo efficacy in the
absence of prior pharmacological and pharmacoki-
netic experiments, and without mechanistic knowl-
edge. One also needs to carefully consider which
transgenes to express ⁄ suppress and what pathogenic
AD mutation to include in the animal model. For
example, a putative BACE-1 inhibitor can be evalu-
ated in a transgenic model with the Swedish mutation

nately, in a clinical trial AD patients (and likely some
healthy subjects) will have substantial amounts of Ab
and tau pathology at the commencement of treatment.
Protein aggregation has often only been prevented in
APP transgenic mice, but it is far more difficult to
clear existing Ab deposits. This has also been seen in,
for example, superoxide dismutase 1 transgenic mice
models of amyotrophic lateral sclerosis [141]. There-
fore, to avoid overinterpreting therapeutic studies in
transgenic mice, it is important to record the age and
evaluate the stage of neuropathology when the animals
were first given the drug. Here we present a few exam-
ples of Ab-based drug candidates that are in preclinical
or clinical development.
Ab immunotherapy is perhaps the most promising
disease-modifying treatment strategy for AD, and it
also illustrates the potential clinical value of transgenic
mice. Immunization would probably never have been
pursued if APP transgenic mice had not been avail-
able. Human clinical trials of active immunization with
fibrillar Ab
1–42
(AN1792) were rapidly initiated when
biochemical and functional efficacy had been proven in
APP transgenic mice [142–144]. Studies were halted in
phase II, because meningoencephalitis developed in a
subgroup of patients [145]. Encouragingly, there was
evidence of Ab plaque clearance in postmortem brain
of vaccinated patients, resembling that of transgenic
mice [146]. Passive immunization, i.e. direct adminis-

and providing suggestive biomarkers to be used in
clinical trials. Examples of pioneering studies are the
demonstration of impaired remyelination of sciatic
nerves and reduced cleavage of the neuregulin-1 pre-
cursor in BACE-1 knockout mice [154], and lethal phe-
notypes observed in PS-1 knockout mice. The latter
experiments led to the realization that the c-secretase
complex also regulates the Notch cell signaling
pathway [155].
Functional studies with AD models
We regard behavioral studies with APP transgenic
mice as being relevant to prove that a certain Ab
species or a pathological lesion has a functional effect
on neurotransmission. Because our knowledge of the
mechanisms and neuropathology of AD are both
incomplete, it is not possible to predict if a new
drug will impact on AD symptomatology based on
functional studies with APP transgenic mice.
Effects of the promoter, transgene overexpression
and strain background all need to be considered when
interpreting functional studies. These parameters can
have a major impact on behavior and also generate
variability. It is also difficult to distinguish between
deficits caused by Ab or APP overexpression. A trans-
genic model should ideally first be investigated in a
comprehensive battery of cognitive and sensorimotor
tests [156], which is labor intensive but can be reward-
ing if combined with statistical analyses [157]. The
MWM task is one of the most widely used cognitive
tests. It depends on hippocampal formation, which is

drawbacks and limit variability associated with
conventional behavioral tasks. These systems should
facilitate reproducible functional studies with animal
models of AD.
Concluding remarks and future
perspectives
Transgenic techniques have revolutionized our ability
to develop animal models of AD, and also contributed
significantly to the understanding of molecular patho-
genesis. Today a wide range of animal models are
available for mechanistic, therapeutic and functional
studies. They offer an appealing means to rapidly
move from simplistic in vitro experiments to clinical
trials. It is important to understand the strengths and
limitations of the models (Table 2). We foresee that
technical advances in RNA interference and gene tar-
geting with, for example, zinc-finger nucleases will be
increasingly utilized in the future. This could lead to
new animal models of AD where proteins are not
overexpressed and also to more sophisticated studies
of pathogenic mechanisms in APP transgenic mice.
Moreover, chimeric proteins will frequently be
designed to target transgene expression to certain sub-
cellular locations in postmitotic neurons and to then
express only a defined Ab peptide. The first transgenic
nonhuman primate model of AD will be developed, as
a result of recent successes in modeling Huntington’s
disease [163]. This will enable limited therapeutic stud-
ies where effects of a new drug on higher cognitive
functions can be better evaluated with high-resolution

C3-3 [58] 18 + + nr nr + ) nr
R.1.40 [64] 14 +++ +++ + nr )) nr
APP23 [65] 6 +++ + + nr +++ ) nr
Tg-CRND8 [69] 3 +++ + + nr + ) nr
APPDutch [32] 22–25 ) + +++ + +++ ) nr
SweDI [75] 3 ) +++ +++ nr +++ ) nr
Tg-ArcSwe [86] 6 +++ + + +++ +++ ) nr
APParc [89] > 12 + + + ) +++ ) nr
PSAPP [92] 6 +++ + + nr + ) nr
3xTg-AD [98] 6 +++ +++ + +++ +++ ) nr
5xFAD [102] 2 +++ + nr +++ +++ + nr
APP ⁄ PS1 KI [104] 2–3 +++ + nr +++ +++ + +++
TBA2 [118] 2 nr +++ ) +++ +++ +++ +++
O. Philipson et al. Animal models of Alzheimer’s disease
FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS 1401
careful in vitro experiments and advanced early clinical
studies will provide important contributions to the
development of the first approved disease-modifying
drug for AD.
Acknowledgements
Mattias Staufenbiel and Thomas Bayer are greatly
acknowledged for providing information on the
APP23 and TBA ⁄ 2 models, and the Swedish Research
Council (2009-4567, LL; 2009-4389, LN) provided
financial support.
References
1 Glenner GG & Wong CW (1984) Alzheimer’s disease:
initial report of the purification and characterization
of a novel cerebrovascular amyloid protein. Biochem
Biophys Res Commun 120, 885–890.

Brice A et al. (2006) APP locus duplication causes
autosomal dominant early-onset Alzheimer disease with
cerebral amyloid angiopathy. Nat Genet 38, 24–26.
9 Olson MI & Shaw CM (1969) Presenile dementia and
Alzheimer’s disease in mongolism. Brain 92 , 147–156.
10 Corder EH, Saunders AM, Strittmatter WJ, Schmechel
DE, Gaskell PC, Small GW, Roses AD, Haines JL &
Pericak-Vance MA (1993) Gene dose of apolipoprotein
E type 4 allele and the risk of Alzheimer’s disease in
late onset families. Science 261, 921–923.
11 Strittmatter WJ, Saunders AM, Schmechel D, Pericak-
Vance M, Enghild J, Salvesen GS & Roses AD (1993)
Apolipoprotein E: high-avidity binding to beta-amy-
loid and increased frequency of type 4 allele in
late-onset familial Alzheimer disease. Proc Natl Acad
Sci USA 90, 1977–1981.
12 Kim J, Basak JM & Holtzman DM (2009) The role of
apolipoprotein E in Alzheimer’s disease. Neuron 63,
287–303.
13 Blennow K, de Leon MJ & Zetterberg H (2006)
Alzheimer’s disease. Lancet 368, 387–403.
14 Bartus RT (2000) On neurodegenerative diseases,
models, and treatment strategies: lessons learned and
lessons forgotten a generation following the cholinergic
hypothesis. Exp Neurol 163, 495–529.
15 Geula C, Wu CK, Saroff D, Lorenzo A, Yuan M &
Yankner BA (1998) Aging renders the brain vulnerable
to amyloid beta-protein neurotoxicity. Nat Med 4,
827–831.
16 Hauss-Wegrzyniak B, Dobrzanski P, Stoehr JD &

1402 FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS
23 Lemere CA, Oh J, Stanish HA, Peng Y, Pepivani I,
Fagan AM, Yamaguchi H, Westmoreland SV &
Mansfield KG (2008) Cerebral amyloid-beta protein
accumulation with aging in cotton-top tamarins:
a model of early Alzheimer’s disease? Rejuvenation Res
11, 321–332.
24 Gandy S, DeMattos RB, Lemere CA, Heppner FL,
Leverone J, Aguzzi A, Ershler WB, Dai J, Fraser P
et al. (2004) Alzheimer’s Abeta vaccination of rhesus
monkeys (Macaca mulatta). Mech Ageing Dev 125,
149–151.
25 Elfenbein HA, Rosen RF, Stephens SL, Switzer RC,
Smith Y, Pare J, Mehta PD, Warzok R & Walker LC
(2007) Cerebral beta-amyloid angiopathy in aged
squirrel monkeys. Histol Histopathol 22, 155–167.
26 Rosen RF, Farberg AS, Gearing M, Dooyema J, Long
PM, Anderson DC, Davis-Turak J, Coppola G,
Geschwind DH et al. (2008) Tauopathy with paired
helical filaments in an aged chimpanzee. J Comp
Neurol 509, 259–270.
27 Wirak D, Bayney R, Ramabhadran TV, Fracasso RP,
Hart JT, Hauer PE, Hsiau P, Pekar SK, Scangos GA
et al. (1992) Age-associated inclusions in normal and
transgenic mouse brain. Science 255, 1445.
28 Kawarabayashi T, Shoji M, Sato M, Sasaki A, Ho L,
Eckman CB, Prada CM, Younkin SG, Kobayashi T
et al. (1996) Accumulation of beta-amyloid fibrils in
pancreas of transgenic mice. Neurobiol Aging 17,
215–222.

hereditary Alzheimer disease with a guanine to
thymine missense change at position 1924 of the APP
gene. Brain Res 571, 133–139.
36 Games D, Adams D, Alessandrini R, Barbour R,
Berthelette P, Blackwell C, Carr T, Clemens J,
Donaldson T et al. (1995) Alzheimer-type neuropatho-
logy in transgenic mice overexpressing V717F
beta-amyloid precursor protein. Nature 373, 523–527.
37 Johnson-Wood K, Lee M, Motter R, Hu K, Gordon
G, Barbour R, Khan K, Gordon M, Tan H et al.
(1997) Amyloid precursor protein processing and A
beta42 deposition in a transgenic mouse model of
Alzheimer disease. Proc Natl Acad Sci USA 94, 1550–
1555.
38 Irizarry MC, Soriano F, McNamara M, Page KJ,
Schenk D, Games D & Hyman BT (1997) Abeta depo-
sition is associated with neuropil changes, but not with
overt neuronal loss in the human amyloid precursor
protein V717F (PDAPP) transgenic mouse. J Neurosci
17, 7053–7059.
39 Masliah E, Sisk A, Mallory M & Games D (2001)
Neurofibrillary pathology in transgenic mice
overexpressing V717F beta-amyloid precursor protein.
J Neuropathol Exp Neurol 60, 357–368.
40 Masliah E, Sisk A, Mallory M, Mucke L, Schenk D &
Games D (1996) Comparison of neurodegenerative
pathology in transgenic mice overexpressing V717F
beta-amyloid precursor protein and Alzheimer’s
disease. J Neurosci 16, 5795–5811.
41 Chen G, Chen KS, Knox J, Inglis J, Bernard A,

Kaneko K, Shimizu T, Kojima T, Nakano I et al.
(1993) Clinical comparison of Alzheimer’s disease in
pedigrees with the codon 717 Val fi Ile mutation in
the amyloid precursor protein gene. Neurobiol Aging
14, 407–419.
47 Mullan M, Crawford F, Axelman K, Houlden H,
Lilius L, Winblad B & Lannfelt L (1992) A pathogenic
mutation for probable Alzheimer’s disease in the APP
gene at the N-terminus of beta-amyloid. Nat Genet 1,
345–347.
48 Cai XD, Golde TE & Younkin SG (1993) Release of
excess amyloid beta protein from a mutant amyloid
beta protein precursor. Science 259, 514–516.
49 Vassar R, Bennett BD, Babu-Khan S, Kahn S,
Mendiaz EA, Denis P, Teplow DB, Ross S, Amarante P
et al. (1999) Beta-secretase cleavage of Alzheimer’s
amyloid precursor protein by the transmembrane
aspartic protease BACE. Science 286, 735–741.
50 Mann DM, Iwatsubo T, Ihara Y, Cairns NJ, Lantos
PL, Bogdanovic N, Lannfelt L, Winblad B, Maat-
Schieman ML et al. (1996) Predominant deposition of
amyloid-beta 42(43) in plaques in cases of Alzheimer’s
disease and hereditary cerebral hemorrhage associated
with mutations in the amyloid precursor protein gene.
Am J Pathol 148, 1257–1266.
51 Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya
Y, Younkin S, Yang F & Cole G (1996) Correlative
memory deficits, Abeta elevation, and amyloid plaques
in transgenic mice. Science 274, 99–102.
52 Haass C, Lemere CA, Capell A, Citron M, Seubert P,

linked presenilin 1 variants elevate Abeta1-42 ⁄ 1-40
ratio in vitro and in vivo. Neuron 17, 1005–1013.
59 Borchelt DR, Davis J, Fischer M, Lee MK, Slunt HH,
Ratovitsky T, Regard J, Copeland NG, Jenkins NA
et al. (1996) A vector for expressing foreign genes in
the brains and hearts of transgenic mice. Genet Anal
13, 159–163.
60 Reaume AG, Howland DS, Trusko SP, Savage MJ,
Lang DM, Greenberg BD, Siman R & Scott RW
(1996) Enhanced amyloidogenic processing of the
beta-amyloid precursor protein in gene-targeted mice
bearing the Swedish familial Alzheimer’s disease
mutations and a ‘humanized’ Abeta sequence. J Biol
Chem 271, 23380–23388.
61 Flood DG, Reaume AG, Dorfman KS, Lin YG, Lang
DM, Trusko SP, Savage MJ, Annaert WG, De
Strooper B et al. (2002) FAD mutant PS-1 gene-
targeted mice: increased A beta 42 and A beta
deposition without APP overproduction. Neurobiol
Aging 23, 335–348.
62 Savage MJ, Trusko SP, Howland DS, Pinsker LR,
Mistretta S, Reaume AG, Greenberg BD, Siman R &
Scott RW (1998) Turnover of amyloid beta-protein in
mouse brain and acute reduction of its level by
phorbol ester. J Neurosci 18, 1743–1752.
63 Lehman EJ, Kulnane LS & Lamb BT (2003)
Alterations in beta-amyloid production and deposition
in brain regions of two transgenic models. Neurobiol
Aging 24, 645–653.
64 Lamb BT, Bardel KA, Kulnane LS, Anderson JJ,

deficits in transgenic mice expressing a double mutant
form of amyloid precursor protein 695. J Biol Chem
276, 21562–21570.
70 Mucke L, Masliah E, Yu GQ, Mallory M,
Rockenstein EM, Tatsuno G, Hu K, Kholodenko D,
Johnson-Wood K et al. (2000) High-level neuronal
expression of abeta 1-42 in wild-type human amyloid
protein precursor transgenic mice: synaptotoxicity
without plaque formation. J Neurosci 20, 4050–4058.
71 Kim J, Onstead L, Randle S, Price R, Smithson L,
Zwizinski C, Dickson DW, Golde T & McGowan E
(2007) Abeta40 inhibits amyloid deposition in vivo.
J Neurosci 27, 627–633.
72 Jankowsky JL, Slunt HH, Gonzales V, Savonenko
AV, Wen JC, Jenkins NA, Copeland NG, Younkin
LH, Lester HA et al. (2005) Persistent amyloidosis
following suppression of Abeta production in a
transgenic model of Alzheimer disease. PLoS Med 2,
e355.
73 Levy E, Carman MD, Fernandez-Madrid IJ, Power
MD, Lieberburg I, van Duinen SG, Bots GT,
Luyendijk W & Frangione B (1990) Mutation of the
Alzheimer’s disease amyloid gene in hereditary cerebral
hemorrhage, Dutch type. Science 248, 1124–1126.
74 Grabowski TJ, Cho HS, Vonsattel JP, Rebeck GW
& Greenberg SM (2001) Novel amyloid precursor
protein mutation in an Iowa family with dementia
and severe cerebral amyloid angiopathy. Ann Neurol
49, 697–705.
75 Davis J, Xu F, Deane R, Romanov G, Previti ML,

Constitutive and regulated alpha-secretase cleavage of
Alzheimer’s amyloid precursor protein by a disintegrin
metalloprotease. Proc Natl Acad Sci USA 96, 3922–
3927.
81 Kumar-Singh S, Dewachter I, Moechars D, Lubke U,
De Jonghe C, Ceuterick C, Checler F, Naidu A,
Cordell B et al. (2000) Behavioral disturbances without
amyloid deposits in mice overexpressing human
amyloid precursor protein with Flemish (A692G) or
Dutch (E693Q) mutation. Neurobiol Dis 7, 9–22.
82 Basun H, Bogdanovic N, Ingelsson M, Almkvist O,
Naslund J, Axelman K, Bird TD, Nochlin D,
Schellenberg GD et al. (2008) Clinical and
neuropathological features of the Arctic APP gene
mutation causing early-onset Alzheimer disease.
Arch Neurol 65, 499–505.
83 Stenh C, Nilsberth C, Hammarback J, Engvall B,
Naslund J & Lannfelt L (2002) The Arctic mutation
interferes with processing of the amyloid precursor
protein. Neuroreport 13, 1857–1860.
84 Sahlin C, Lord A, Magnusson K, Englund H, Almeida
CG, Greengard P, Nyberg F, Gouras GK, Lannfelt L
et al. (2007) The Arctic Alzheimer mutation favors
intracellular amyloid-beta production by making amy-
loid precursor protein less available to alpha-secretase.
J Neurochem 101, 854–862.
85 Knobloch M, Konietzko U, Krebs DC & Nitsch RM
(2007) Intracellular Abeta and cognitive deficits
precede beta-amyloid deposition in transgenic
arcAbeta mice. Neurobiol Aging 28, 1297–1306.

19, 939–945.
92 Holcomb L, Gordon MN, McGowan E, Yu X,
Benkovic S, Jantzen P, Wright K, Saad I, Mueller R
et al. (1998) Accelerated Alzheimer-type phenotype in
transgenic mice carrying both mutant amyloid
precursor protein and presenilin 1 transgenes.
Nat Med 4, 97–100.
93 Richards JG, Higgins GA, Ouagazzal AM, Ozmen L,
Kew JN, Bohrmann B, Malherbe P, Brockhaus M,
Loetscher H et al. (2003) PS2APP transgenic mice,
coexpressing hPS2mut and hAPPswe, show age-related
cognitive deficits associated with discrete brain amyloid
deposition and inflammation. J Neurosci 23, 8989–9003.
94 Lewis J, Dickson DW, Lin WL, Chisholm L, Corral
A, Jones G, Yen SH, Sahara N, Skipper L et al.
(2001) Enhanced neurofibrillary degeneration in trans-
genic mice expressing mutant tau and APP. Science
293, 1487–1491.
95 Boutajangout A, Authelet M, Blanchard V, Touchet
N, Tremp G, Pradier L & Brion JP (2004) Characteri-
sation of cytoskeletal abnormalities in mice transgenic
for wild-type human tau and familial Alzheimer’s
disease mutants of APP and presenilin-1. Neurobiol
Dis 15, 47–60.
96 Masliah E, Rockenstein E, Veinbergs I, Sagara Y,
Mallory M, Hashimoto M & Mucke L (2001) beta-
Amyloid peptides enhance alpha-synuclein accumula-
tion and neuronal deficits in a transgenic mouse model
linking Alzheimer’s disease and Parkinson’s disease.
Proc Natl Acad Sci USA 98, 12245–12250.

tial factors in amyloid plaque formation. J Neurosci
26, 10129–10140.
103 Ohno M, Cole SL, Yasvoina M, Zhao J, Citron M,
Berry R, Disterhoft JF & Vassar R (2007) BACE1
gene deletion prevents neuron loss and memory deficits
in 5XFAD APP ⁄ PS1 transgenic mice. Neurobiol Dis
26, 134–145.
104 Casas C, Sergeant N, Itier JM, Blanchard V, Wirths
O, van der Kolk N, Vingtdeux V, van de Steeg E, Ret
G et al. (2004) Massive CA1 ⁄ 2 neuronal loss with
intraneuronal and N-terminal truncated Abeta42 accu-
mulation in a novel Alzheimer transgenic model. Am J
Pathol 165, 1289–1300.
105 DeMattos RB, Bales KR, Parsadanian M, O’Dell MA,
Foss EM, Paul SM & Holtzman DM (2002) Plaque-
associated disruption of CSF and plasma amyloid-beta
(Abeta) equilibrium in a mouse model of Alzheimer’s
disease. J Neurochem 81, 229–236.
106 Christie R, Yamada M, Moskowitz M & Hyman B
(2001) Structural and functional disruption of vascular
smooth muscle cells in a transgenic mouse model of
amyloid angiopathy. Am J Pathol 158, 1065–1071.
107 Maeda A, Yamada M, Itoh Y, Otomo E, Hayakawa
M & Miyatake T (1993) Computer-assisted three-
dimensional image analysis of cerebral amyloid
angiopathy. Stroke 24, 1857–1864.
108 Lazarov O, Lee M, Peterson DA & Sisodia SS
(2002) Evidence that synaptically released
Animal models of Alzheimer’s disease O. Philipson et al.
1406 FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS

Gonzales E, Holtzman DM & Lee JM (2009) Charac-
terizing the appearance and growth of amyloid plaques
in APP ⁄ PS1 mice. J Neurosci 29, 10706–10714.
115 Knowles RB, Wyart C, Buldyrev SV, Cruz L, Urbanc
B, Hasselmo ME, Stanley HE & Hyman BT (1999)
Plaque-induced neurite abnormalities: implications for
disruption of neural networks in Alzheimer’s disease.
Proc Natl Acad Sci USA 96, 5274–5279.
116 Spires TL, Meyer-Luehmann M, Stern EA, McLean
PJ, Skoch J, Nguyen PT, Bacskai BJ & Hyman BT
(2005) Dendritic spine abnormalities in amyloid
precursor protein transgenic mice demonstrated by
gene transfer and intravital multiphoton microscopy.
J Neurosci 25, 7278–7287.
117 Calhoun ME, Wiederhold KH, Abramowski D,
Phinney AL, Probst A, Sturchler-Pierrat C,
Staufenbiel M, Sommer B & Jucker M (1998) Neuron
loss in APP transgenic mice. Nature 395, 755–756.
118 Wirths O, Breyhan H, Cynis H, Schilling S, Demuth
HU & Bayer TA (2009) Intraneuronal pyroglutamate-
Abeta 3-42 triggers neurodegeneration and lethal
neurological deficits in a transgenic mouse model.
Acta Neuropathol 118, 487–496.
119 Bales KR, Verina T, Dodel RC, Du Y, Altstiel L,
Bender M, Hyslop P, Johnstone EM, Little SP et al.
(1997) Lack of apolipoprotein E dramatically reduces
amyloid beta-peptide deposition. Nat Genet 17, 263–
264.
120 Fagan AM, Watson M, Parsadanian M, Bales KR,
Paul SM & Holtzman DM (2002) Human and murine

126 Nilsson LN, Arendash GW, Leighty RE, Costa DA,
Low MA, Garcia MF, Cracciolo JR, Rojiani A, Wu X
et al. (2004) Cognitive impairment in PDAPP mice
depends on ApoE and ACT-catalyzed amyloid
formation. Neurobiol Aging 25, 1153–1167.
127 Nilsson LN, Bales KR, DiCarlo G, Gordon MN,
Morgan D, Paul SM & Potter H (2001) Alpha-1-antic-
hymotrypsin promotes beta-sheet amyloid plaque
deposition in a transgenic mouse model of Alzheimer’s
disease. J Neurosci 21, 1444–1451.
128 Mucke L, Yu GQ, McConlogue L, Rockenstein EM,
Abraham CR & Masliah E (2000) Astroglial expres-
sion of human alpha(1)-antichymotrypsin enhances
alzheimer-like pathology in amyloid protein precursor
transgenic mice. Am J Pathol 157, 2003–2010.
129 Wyss-Coray T, Lin C, Yan F, Yu GQ, Rohde M,
McConlogue L, Masliah E & Mucke L (2001) TGF-
beta1 promotes microglial amyloid-beta clearance and
reduces plaque burden in transgenic mice. Nat Med 7,
612–618.
O. Philipson et al. Animal models of Alzheimer’s disease
FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS 1407
130 Wyss-Coray T, Masliah E, Mallory M, McConlogue
L, Johnson-Wood K, Lin C & Mucke L (1997)
Amyloidogenic role of cytokine TGF-beta1 in
transgenic mice and in Alzheimer’s disease. Nature
389, 603–606.
131 Simard AR, Soulet D, Gowing G, Julien JP & Rivest
S (2006) Bone marrow-derived microglia play a critical
role in restricting senile plaque formation in Alzhei-

1996.
138 Sebastiani G, Krzywkowski P, Dudal S, Yu M,
Paquette J, Malo D, Gervais F & Tremblay P (2006)
Mapping genetic modulators of amyloid plaque
deposition in Tg-CRND8 transgenic mice. Hum Mol
Genet 15, 2313–2323.
139 Page RM, Baumann K, Tomioka M, Perez-Revuelta
BI, Fukumori A, Jacobsen H, Flohr A, Luebbers T,
Ozmen L et al. (2008) Generation of Abeta38 and
Abeta42 is independently and differentially affected by
familial Alzheimer disease-associated presenilin
mutations and gamma-secretase modulation. J Biol
Chem 283, 677–683.
140 Philipson O, Hammarstrom P, Nilsson KP, Portelius
E, Olofsson T, Ingelsson M, Hyman BT, Blennow K,
Lannfelt L et al. (2009) A highly insoluble state of
Abeta similar to that of Alzheimer’s disease brain is
found in Arctic APP transgenic mice. Neurobiol Aging
30, 1393–1405.
141 Schnabel J (2008) Neuroscience: standard model.
Nature 454, 682–685.
142 Janus C, Pearson J, McLaurin J, Mathews PM,
Jiang Y, Schmidt SD, Chishti MA, Horne P,
Heslin D et al. (2000) A beta peptide immunization
reduces behavioural impairment and plaques in
a model of Alzheimer’s disease. Nature 408, 979–
982.
143 Morgan D, Diamond DM, Gottschall PE, Ugen KE,
Dickey C, Hardy J, Duff K, Jantzen P, DiCarlo G
et al. (2000) A beta peptide vaccination prevents

of Alzheimer’s disease. Proc Natl Acad Sci USA 98,
8850–8855.
150 Lord A, Gumucio A, Englund H, Sehlin D, Sundquist
VS, Soderberg L, Moller C, Gellerfors P, Lannfelt L
et al. (2009) An amyloid-beta protofibril-selective anti-
body prevents amyloid formation in a mouse model of
Alzheimer’s disease. Neurobiol Dis 36, 425–434.
151 Siemers ER, Dean RA, Friedrich S, Ferguson-Sells L,
Gonzales C, Farlow MR & May PC (2007) Safety,
tolerability, and effects on plasma and cerebrospinal
fluid amyloid-beta after inhibition of gamma-secretase.
Clin Neuropharmacol 30, 317–325.
Animal models of Alzheimer’s disease O. Philipson et al.
1408 FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS
152 Henley DB, May PC, Dean RA & Siemers ER (2009)
Development of semagacestat (LY450139), a func-
tional gamma-secretase inhibitor, for the treatment of
Alzheimer’s disease. Expert Opin Pharmacother 10,
1657–1664.
153 May PC, Yang ZX, Li WY, Hyslop PA, Siemers E &
Boggs LN (2004) Multi-compartmental pharmacody-
namic assessment of the functional gamma-secretase
inhibitor LY450139 in PDAPP transgenic mice and
non-transgenic mice. Neurobiol Aging 25, S65–S65.
154 Willem M, Garratt AN, Novak B, Citron M,
Kaufmann S, Rittger A, DeStrooper B, Saftig P,
Birchmeier C et al. (2006) Control of peripheral nerve
myelination by the beta-secretase BACE1. Science 314,
664–666.
155 Shen J, Bronson RT, Chen DF, Xia W, Selkoe DJ &

of a murine model of Alzheimer’s disease in a seminat-
uralistic environment using RFID tracking. Behav Res
Methods 41, 850–856.
163 Yang SH, Cheng PH, Banta H, Piotrowska-Nitsche K,
Yang JJ, Cheng EC, Snyder B, Larkin K, Liu J
et al. (2008) Towards a transgenic model of
Huntington’s disease in a non-human primate. Nature
453, 921–924.
164 Tremblay C, Pilote M, Phivilay A, Emond V, Bennett
DA & Calon F (2007) Biochemical characterization of
Abeta and tau pathologies in mild cognitive impair-
ment and Alzheimer’s disease. J Alzheimers Dis 12,
377–390.
165 Giedraitis V, Sundelof J, Irizarry MC, Garevik N,
Hyman BT, Wahlund LO, Ingelsson M & Lannfelt L
(2007) The normal equilibrium between CSF and
plasma amyloid beta levels is disrupted in Alzheimer’s
disease. Neurosci Lett 427, 127–131.
O. Philipson et al. Animal models of Alzheimer’s disease
FEBS Journal 277 (2010) 1389–1409 ª 2010 The Authors Journal compilation ª 2010 FEBS 1409


Nhờ tải bản gốc

Tài liệu, ebook tham khảo khác

Music ♫

Copyright: Tài liệu đại học © DMCA.com Protection Status