ArticlePDF AvailableLiterature Review

Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective

Authors:
  • Xuanwu Hospital, Capital Medical University

Abstract

Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
REVIEWhttps://doi.org/10.1007/s11427-022-2357-x
...........................................................................................................
Critical thinking of Alzheimer’s transgenic mouse model: current
research and future perspective
Xinyue Li1†, Meina Quan1,2†, Yiping Wei1, Wei Wang1,2, Lingzhi Xu1,
Qi Wang1,2 & Jianping Jia1,2,3,4,5,6*
1Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University,
Beijing 100053, China;
2National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing 100053, China;
3Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing 100053, China;
4Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing 100053, China;
5Center of Alzheimer’s Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical
University, Beijing 100053, China;
6Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China
Received December 22, 2022; accepted April 23, 2023; published online July 17, 2023
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer’s Disease (AD). AD
models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each
transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic
mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model
built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the
preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multi-
target drug development in AD.
Alzheimer’s disease, transgenic mouse model, amyloid-β oligomers, multi-target therapy
Citation: Li, X., Quan, M., Wei, Y., Wang, W., Xu, L., Wang, Q., and Jia, J. (2023). Critical thinking of Alzheimer’s transgenic mouse model: current research
and future perspective. Sci China Life Sci 66, https://doi.org/10.1007/s11427-022-2357-x
Introduction
Current animal models for Alzheimer’s disease (AD) include
intermediate state animal vectors such as C.elegans, fruit fly
and zebrafish, high level models such as rodents and non-
human primates (Drummond and Wisniewski, 2017). There
are several methods for AD modeling, such as spontaneous
aging, physical or chemical lesion, and gene modification,
which include transgenosis and gene editing (Drummond
and Wisniewski, 2017;Götz et al., 2018;Zhang et al., 2020).
AD transgenic (Tg) Drosophila models can show patholo-
gical changes such as synaptic degeneration, axonal changes,
and apoptosis, and can shorten experimental cycle (Guna-
wardena and Goldstein, 2001). However, the lower order
animal models have limitations such as great distance from
human physiology, lack of genetic homology with humans,
lack of amyloid-β (Aβ), complexity in nervous system, and
behaviors seen in human (Alexander et al., 2014;Fernandez-
Funez et al., 2015). Non-human primates have the limitations
of long lifespan, high price, immature cognitive behavioral
paradigm, rare tauopathy and ethical concerns for research
studies (Drummond and Wisniewski, 2017;Heuer et al.,
© Science China Press 2023 life.scichina.com link.springer.com
SCIENCE CHINA
Life Sciences
†Contributed equally to this work
*Corresponding author (email: jiajp@vip.126.com)
2012;Perez et al., 2013). The most frequently used animal
models are rodents, especially transgenic models. Though
transgenic rat models of AD have also been developed, they
are not as well characterized as mouse models, and amyloid
plaque expression is at lower levels than in transgenic mice
(Cohen et al., 2013;Leon et al., 2010;Pang et al., 2022).
Mice have the advantages of small size, short reproductive
cycle, mature gene modification technology, high homology
with human genes and high-level neural activities, thus are
suitable for understanding AD pathogenesis and develop-
ment of candidate AD therapies (Hafezparast et al., 2002).
There are three methods to construct transgenic mouse
models of AD: random transgenic, CRISPR/Cas9 and em-
bryonic stem cell targeting. Each of the three technologies
has its advantages and disadvantages. Random transgenic
technology is to randomly integrate foreign genes into mouse
chromosomes to obtain randomly inserted transgenic mice
(Jaenisch and Mintz, 1974;Oakley et al., 2006). Generally,
the founder (F0) mice obtained need to be bred in-
dependently, and backcrossed with wild type (WT) mice for
more than two generations to detect the expression of foreign
genes, in order to screen out mouse strains that meet the
experimental requirements. The advantage of this method is
that mice with multiple expression levels of target genes can
be obtained. The disadvantage is that the reproduction work
is cumbersome and not sure to obtain an ideal model of
foreign gene expression, and there is a risk that the genetic
information of the mouse itself might be destroyed. CRISPR/
Cas9 introduces the target DNA fragment through homo-
logous recombination with Cas9 protein and guide RNA as
the core, then gets the exogenous gene knock-in (KI) or gene
editing mice (Gaj et al., 2013;Li et al., 2013;Shen et al.,
2013). Embryonic stem cell targeting technology carries out
DNA homologous recombination in mouse embryonic stem
cells to obtain gene KI or gene editing mice (Thomas and
Capecchi, 1987). Both CRISPR/Cas9 and embryonic stem
cell targeting can obtain mouse humanization, gene knock-
out (KO) and point mutation models. CRISPR/Cas9 has a
short cycle and low cost, but there is a risk of missing target
and it is not suitable for complex genetic transformation.
Embryonic stem cell targeting technology has a long cycle
and high cost, but there is no risk of missing target and it is
suitable for complex gene transformation.
The first AD transgenic mouse model was generated in the
1990s, based on overexpression of human amyloid precursor
protein (APP) with familial AD (FAD) mutation (Games et
al., 1995). Thereafter, more and more transgenic models
were generated, including single transgenic and multiple
transgenic models (Borchelt et al., 1997;Mucke et al., 2000;
Scearce-Levie et al., 2020;Sturchler-Pierrat et al., 1997).
Transgenic mouse models have some common features as
well as heterogeneities. The most commonly used mouse
models carry two or more mutations, and have prominent
pathological characteristics and early onset age (Mullane and
Williams, 2019;Rao and Yamada, 2021). Different mutation
sites have different pathogenic functions for AD (Qiu et al.,
2020;Shen et al., 2019). Neuropsychological and imaging
studies also found that APP mutation, PSEN1 mutation and
APOE affect cognitive phenotypes and imaging biomarkers
in a different way (Almkvist et al., 2019;Quan et al., 2020;
Wong et al., 2020). As a result, the mechanisms based on
each model may only be a subtype of AD pathogenesis, not
as a whole.
According to AlzForum (https://www.alzforum.org/re-
search-models/alzheimers-disease), there are 197 AD mouse
models. This review intends to review the current research
findings and progress on transgenic mouse models, including
three major pathogenic genes (APP,PSEN1,PSEN2), AD
risk genes and some AD-related genes such as MAPT gene.
The usage of these mouse models in drug development stu-
dies is also reviewed. This review aims to deepen our un-
derstanding of AD pathogenesis and shed light on the future
applications of transgenic mice in drug development.
Comparisons between single-transgenic mouse
models
The pathological, electrophysiological, and cognitive fea-
tures were compared between single mutation transgenic
mouse models reported in AlzForum and the literature, and
the timing of each feature was also summarized. Most AD
single transgenic mouse models are constructed with APP,
PSEN1 and MAPT gene mutations. Two PSEN2 models were
found in AlzForum, but they lack pathological evidence
(Hwang et al., 2002;Richards et al., 2003).
PSEN1 single-transgenic mouse models
Seven mouse models carrying a single mutation of PSEN1
with more than one pathological change or cognitive im-
pairments are summarized in Table 1 (Barrow et al., 2000;
Begley et al., 1999;Duff et al., 1996;Guo et al., 1999;
Holcomb et al., 1998;Lalonde and Strazielle, 2005;Nakano
et al., 1999;Schneider et al., 2001;Tanemura et al., 2006;
Wang et al., 2004;Wang et al., 2016;Wang et al., 2012;Wen
et al., 2004;Wen et al., 2002;Zhang et al., 2014;Zufferey et
al., 2013). We can see that PS1(I213T) KI, PS1(M146L),
PS1(M146V), PS1(P117L), and PS1V97L Tg mice showed
elevated Aβ42 level, and plaques were not seen in any
PSENs single transgenic models (Barrow et al., 2000;Begley
et al., 1999;Nakano et al., 1999;Wen et al., 2004;Zhang et
al., 2014). Only PS1(P117L) and PS1V97L Tg mice showed
accumulation of oligomer (AβO) (Wang et al., 2016;
Zhang et al., 2014;Zufferey et al., 2013). Increased tau
phosphorylation was detected in PS1(P117L), PS1(I213T)
2Li, X., et al. Sci China Life Sci
Table 1 Characteristics of PSEN1 single mutation mouse modelsa)
Mutation
site Model name Tau Neuronal damage Gliosis Synaptic changes Changes in LTP/
LTD Cognitive dysfunc-
tion References
PSEN1
A246E PS1(A246E) Absent up to 24 months. Absent up to
24 months. ———
A weak stimulation
at Schaeffer’s
collaterals to CA1
neuron synapses
elicited LTP.
Absent at 1 and
9 months. (Schneider et al.,
2001)
PSEN1
I213T PS1(I213T)
KI Elevated Aβ42(43) at
4–5 months.
Tau phosphorylation
in hippocampal neu-
rons at 7 months and
in CA3 neurons at
15 months
β-sheet structure of
tau fibrils and
tangles at 16 months.
————Spatial learning
deficits at
11 months.
(Lalonde and Stra-
zielle, 2005;Naka-
no et al., 1999;
Tanemura et al.,
2006)
PSEN1
M146L
(A>C) PS1(M146L) Elevated Aβ42(43). Absent. Absent.
Larger medium and
late after hyperpo-
larizations in CA3
pyramidal cells at
1–2 months.
Absent in Y maze
at 3–3.5 months.
(Barrow et al.,
2000;Begley et al.,
1999;Duff et al.,
1996;Holcomb et
al., 1998)
PSEN1
M146V
PS1(M146V) Selective increase
in Aβ42(43). Absent. Absent.
Larger medium and
late after hyperpo-
larizations in CA3
pyramidal cells at
1–2 months.
(Barrow et al.,
2000;Begley et al.,
1999;Duff et al.,
1996)
PS1(M146V)
KI Absent. Absent. Reduced adult
neurogenesis in DG
at 3 months. Absent. Absent. Absent at
3 months.
Impaired contex-
tual fear learning at
3 months.
(Guo et al., 1999;
Wang et al., 2004)
PSEN1
P117L PS1(P117L)
(line 13)
Increased Aβ42 at 2 months;
increased AβOs in CA1
and amygdala at 10
months.
Tau phosphorylation
at 10 months
Impaired neurogen-
esis at 1 month;
impaired neural
progenitor cells
survival.
————
(Wen et al., 2004;
Wen et al., 2002;
Zufferey et al.,
2013)
PSEN1
V97L PS1V97L Tg
Increased Aβ42 and Aβ42/
Aβ40 ratio at 9 months;
intraneuronal AβO
accumulation in cortex at
6 months and in
hippocampus at 9 months;
absent of plaques up to
24 months.
Tau phosphorylation
at 9 months; tangles
formed at 12 months.
Neuronal damage
occasionally at
10-14 months;
neuronal degenera-
tion at 24 months.
Microglia activa-
tion in cortex and
astrocyte activation
at 6 months;
microglia activa-
tion in hippocam-
pus at 9 months.
Synaptophysin re-
duction in
hippocampus at
9 months;
decreased synaptic
density at
12 months.
Decreased LTP
in hippocampus
at 6 months.
Spatial learning
and memory defi-
cits at 9 months.
(Wang et al., 2016;
Wang et al., 2012;
Zhang et al., 2014)
a) The table is ordered by mutation site alphabetically. “—” means no data.
3
Li, X., et al. Sci China Life Sci
KI and PS1V97L Tg mice, but neurofibrillary tangles (NFTs)
could be observed only in PS1V97L Tg mice (Tanemura et
al., 2006;Wang et al., 2012;Zufferey et al., 2013). PS1
(M146V) KI and PS1(P117L) mice showed impaired neu-
rogenesis at young age (Guo et al., 1999;Wen et al., 2004).
PS1(M146V) KI, PS1V97L Tg and PS1(I213T) KI mice
showed cognitive impairment at 3, 9 and 11 months of age,
respectively (Lalonde and Strazielle, 2005;Wang et al.,
2004;Zhang et al., 2014). None of the PSENs transgenic
models showed gliosis, synaptic changes, or neuronal de-
generation except PS1V97L Tg mice (Wang et al., 2012).
None of the PSENs transgenic models showed neuronal
death. Major PSEN1 models with more than three patholo-
gical or cognitive changes are described below.
PS1(I213T) KI mice
PS1(I213T) KI mice were constructed using embryonic stem
cell gene targeting technology and introducing I213T mis-
sense mutation into exon 7 of mouse PSEN1 gene (Nakano et
al., 1999). The mice showed elevated Aβ42 and Aβ42/40
ratio in brain at 4–5 months (Nakano et al., 1999), by im-
pairing catalytic activity of γ-secretase-mediated γ-site
cleavage (Shimojo et al., 2008). Tau phosphorylation were
detected in hippocampal neurons at 7 and 15 months, and β-
sheet structure of tau fibrils and tangles appeared at
16 months (Tanemura et al., 2006). Spatial learning deficits
were detected by Morris water maze (MWM) at 11 months
(Lalonde and Strazielle, 2005).
PS1(P117L) mice
PS1(P117L) mice were constructed using random transgenic
technology and overexpressing the P117L mutation of hu-
man PSEN1 driven by neuron specific enolase promoter
(Wen et al., 2002). The mice showed impaired neurogenesis
as early as 1 month (Wen et al., 2004), plaque and tau
phosphorylation in CA1 region at 10 months (Zufferey et al.,
2013). This mouse model was crossed with APP KI mice to
study the synergistic effect of the pathogenic mutations in
APP and PS1 genes in AD, such as AppNL-FPsen1P117L dou-
ble-mutant mice (Sato et al., 2021).
PS1V97L Tg mice
PS1V97LTg mice were constructed using random transgenic
technology and overexpressing human PSEN1 gene with
V97L mutation (from Chinese FAD) driven by the platelet-
derived growth factor (PDGF)-β promoter (Wang et al.,
2012). The earliest pathologies in PS1V97L Tg mice were
accumulation, glial cell activation and decreased hippo-
campal long-term potentiation (LTP) at the age of 6 months
(Zhang et al., 2014). Intraneuronal AβOs appeared first in the
cortex at 6 months, and progressed to hippocampus at
9 months (Zhang et al., 2014). Microglia activation appeared
in the cortex at 6 months and hippocampus at 9 months
(Zhang et al., 2014). Astrocyte activation in the cortex and
hippocampus started at 6 months, and accumulated over time
(Zhang et al., 2014). Spatial learning and memory decline
started at 9 months (Wang et al., 2012). At the same time, tau
phosphorylations at Ser199, Ser202, Thr231, Ser396, and
Ser404 were significantly increased in CA3/CA4 regions of
hippocampus (Wang et al., 2012). Age-related synaptophy-
sin decreased in hippocampus at 9 months, especially in the
CA3 region (Zhang et al., 2014). Tangle-like structures and
atrophic degenerated neurons appeared in the frontal cortex
at 10 months (Wang et al., 2012). Synaptic density decreased
significantly at 12 months. Neuronal damage could be seen
occasionally at 10 and 14 months, accompanied by en-
largement of lateral ventricle. Neuronal degeneration oc-
cured at 24 months, but there were no neuronal loss or
plaques even at 24 months (Zhang et al., 2014). PS1V97L Tg
mice also exhibited enhanced neuroinflammation and oxi-
dative stress (Hou et al., 2018;Wang et al., 2016), abnorm-
alities of mitochondrial membrane potential via the Sirtuin 3
(SIRT3) pathway (Li et al., 2018), and decreased Nrf2/ARE
signaling pathway activity (Tian et al., 2019). Using this
model, sulforaphane and honokiol were found to alleviate
cognitive impairment caused by AβOs (Hou et al., 2018;Li
et al., 2018;Tian et al., 2019). Figure 1 summarized the
timeline and major pathological changes of PS1V97L Tg
mice.
APP single-transgenic mouse models
APP encodes amyloid precursor protein, a transmembrane
protein which is cleaved to form amyloidogenic peptides.
73 mutations in APP have been reported, and are associated
with familial forms of early onset AD as well as with cerebral
amyloid angiopathy (CAA). Thirteen mouse models carrying
single mutation of APP with more than one pathological
change or cognitive impairment are shown in Table 2
(Boncristiano et al., 2005;Brown et al., 2005;Calhoun et al.,
1998;Chapman et al., 1999;Dodart et al., 1999;Domnitz et
al., 2005;Frautschy et al., 1998;Games et al., 1995;Herzig
et al., 2004;Hsiao et al., 1996;Hwang et al., 2004;Irizarry et
al., 1997;Jacobsen et al., 2006;Kawasumi et al., 2004;Kelly
et al., 2003;Kim et al., 2012;Kulnane and Lamb, 2001;Lanz
et al., 2003;Larson et al., 1999;Lehman et al., 2003;Lord et
al., 2006;Lord et al., 2011;Marazuela et al., 2022;Masliah
et al., 2001;Moechars et al., 1999;Philipson et al., 2009;
Puig et al., 2004;Reichwald et al., 2009;Richards et al.,
2003;Richardson et al., 2003;Roder et al., 2003;Rönnbäck
et al., 2012;Snellman et al., 2013;Stalder et al., 1999;
Sturchler-Pierrat et al., 1997;Tomiyama et al., 2010;To-
miyama et al., 2008;Van Dam et al., 2003;Van Dorpe et al.,
4Li, X., et al. Sci China Life Sci
2000). Plaques appeared in the human APP Swedish muta-
tion (KM670/671NL), Indiana mutation (V717F), and Lon-
don mutation (V717I) mice, but not in APPSw-NSE mice
(Games et al., 1995;Hsiao et al., 1996;Lehman et al., 2003;
Lord et al., 2011;Moechars et al., 1999;Richards et al.,
2003;Richardson et al., 2003;Sturchler-Pierrat et al., 1997).
NFTs were absent in all APP single mutant mice. Only APP
E693Δ-Tg and APP23 mice showed significant hippocampal
neuron death (Calhoun et al., 1998;Tomiyama et al., 2010).
APP23 and PDAPP mice showed the earliest cognitive im-
pairment, at 3 months of age (Dodart et al., 1999;Van Dam et
al., 2003), while other models including APP E693Δ-Tg,
APPSw-NSE, Tg2576, TAS10 and APP (V717I) mice
showed spatial memory deficits at 6–12 months (Hsiao et al.,
1996;Hwang et al., 2004;Moechars et al., 1999;Richardson
et al., 2003;Tomiyama et al., 2010). Different from other
APP models, APP(V642) KI mouse model was generated
using targeted knock-in of the V642I (corresponds to human
APP V717I) mutation into the exon 17 of mouse APP gene,
which showed cognitive impairment at 26–27 months (Ka-
wasumi et al., 2004). Major APP models with more than
three pathological or cognitive changes are described below.
APPE693Δ-Tg mice
This model expresses low levels of human APP (isoform
695) carrying the E693Δ(Osaka) mutation under the control
of the mouse prion promoter (Tomiyama et al., 2010). The
expression levels of human APP in the mice were similar to
those of endogenous mouse APP (Tomiyama et al., 2010).
Figure 1 The timeline and major pathological changes of the PS1V97L Tg mice. PS1V97L mutation was found in 2003, and PS1V97L Tg mouse model
was established in 2012–2014. Series of neuropathological and behavioral changes as well as underlying mechanisms were discovered subsequently,
including increased intraneuronal AβOs, microglial activation, impairment of LTP, increased phosphorylation of tau, increased levels of various inflammatory
factors, cognitive impairment in the MWM, NFT-like structures, and reduced synaptic density. No significant amyloid plaque or neuronal loss was found.
5
Li, X., et al. Sci China Life Sci
Table 2 Characteristics of APP single mutation mouse modelsa)
Mutation site Model name Tau Neuronal damage Gliosis Synaptic
changes Changes in
LTP/LTD Cognitive
dysfunction References
APP E693del
(Osaka) APP E693Δ-Tg
(Osaka)
AβO accumulation within
neurons at 8 months; no
plaques.
No tangles;
abnormal tau
phosphorylation
at 8 months.
Neuronal loss in
hippocampal CA3
region at
24 months.
Microgliosis at 12 months;
astrocytosis at 18 months.
Decrease in
synaptic den-
sity in hippo-
campal CA3
region at
8 months.
Reduced short
term plasticity
and LTP at
8 months.
Memory
impairment in
MWM
at 8 months.
(Tomiyama et al.,
2010;Tomiyama et
al., 2008)
APP E693G
(Arctic) TgAPParc
Diffused extracellular
deposition in subiculum
at 7–9 months; in inter-
connected regions at
12–15 months.
(Rönnbäck et al.,
2012)
APP E693Q
(Dutch) APPDutch
No plaques; increased
Aβ40/Aβ42 ratio; de-
position in blood vessel at
22–24 months.
Absent.
Microgliosis adjacent to
amyloid-laden vessels and
astrocyte activation in CAA
affected neocortical regions
earlier than 29 months.
(Herzig et al.,
2004)
APP
KM670/
671NL
(Swedish)
APP23
plaques at 6 months,
faster in female; de-
position in neocortex at
12 months;
age-related increase in
vascular deposits by
12 months; plaques occu-
py more than 25% of
neocortex and hippocam-
pus by 24 months.
No tangles;
hyperphosphorylated
tau in dystrophic
neurites around
plaques at 6 months.
14%–28%
neuronal loss in
hippocampal CA1
region at
14–18 months.
Microglia activation at
4–9 months; increased C1q
at 9 months; increased C3 at
18 months; upregulation of
neuro-inflammation mar-
kers, activated astrocytes
and macrophage at
20 months.
Absent at
24 months.
Absent at 3, 6,
9, 12, 18, or
24 months.
Spatial memory
deficit in MWM
at 3 months.
(Boncristiano et al.,
2005;Calhoun et
al., 1998;Kelly et
al., 2003;Marazue-
la et al., 2022;
Reichwald et al.,
2009;Roder et al.,
2003;Snellman et
al., 2013;Stalder et
al., 1999;Sturchler-
Pierrat et al., 1997;
Van Dam et al.,
2003)
APPSwe
plaques in neocortex
and hippocampus at
17–18 months; Aβ40 5–
10 times higher than
Aβ42.
(Richards et al.,
2003)
APP(Swedish)
(R1.40)
deposition in frontal
cortex at 14–16 months;
spread to whole cortex at
18–20 months; occasional
deposit in corpus callo-
sum and hippocampus.
No tangles;
phosphorylated
tau at
14–16 months.
Plaque-related reactive
astrocyte and microglia
at 14–16 months. Absent. (Kulnane and
Lamb, 2001;Leh-
man et al., 2003)
APPSw-NSE Increased Aβ42 in cortex
and hippocampus at
12 months; no plaques.
Increased tau
phosphorylation at
12 months.
Increased caspase-3
staining neuron and
TdT-mediated
dUTP Nick-End
Labeling (TU-
NEL)-stained nu-
clei in cortex and
hippocampus at
12 months.
Longer escape
latency in MWM
at 12 months.
(Hwang et al.,
2004;Kim et al.,
2012)
(To be continued on the next page)
6Li, X., et al. Sci China Life Sci
(Continued)
Mutation site Model name Tau Neuronal damage Gliosis Synaptic
changes Changes in
LTP/LTD Cognitive
dysfunction References
TAS10
(thy1-APPswe)
Aβ40 and Aβ42 accumu-
lation and fibrillar amy-
loid plaques in cortex and
hippocampus
at 12 months; increase
progressively up to
24 months.
Absent. Astrocytes and microglia
proliferation in DG at
6 months.
Dystrophic
synapses at
6 months;
decreased
synapse num-
ber at
24 months.
Spatial memory
deficits in MWM
at 6 months;
defects of
spontaneous
alternating
behavior in Y
maze at
12 months.
(Brown et al.,
2005;Richardson
et al., 2003)
tg-APPSwe
Intraneuronal aggre-
gation at 6 months; CAA
and congophilic parench-
ymal plaques in cortex,
hippocampus and
thalamus at 9 months;
extracellular amyloid
deposition and plaques
formation at 12 months.
Absent. Absent.
Microgliosis and astroglio-
sis in hippocampus, thala-
mus, and around plaques in
cortex at 12 months.
(Lord et al., 2006;
Lord et al., 2011;
Philipson et al.,
2009)
Tg2576
plaques and vascular
deposition at
11–13 months; extensive
CAA at 22 months.
Hyperphosphorylated
tau in neurites around
plaques at 16 months.
Absent or very
limited.
Increase in microglial
density and size at
10–16 months.
Dendritic spine
loss by
4.5 months in
hippocampal
CA1 region.
Decline in
LTP in DG at
5 months.
Impaired spatial
learning, working
memory, and
contextual fear
conditioning at
9 months.
(Chapman et al.,
1999;Domnitz et
al., 2005;Frautschy
et al., 1998;Hsiao
et al., 1996;Irizarry
et al., 1997;Jacob-
sen et al., 2006;
Lanz et al., 2003;
Puig et al., 2004)
APP V717F
(Indiana) PDAPP (line109)
No plaques at
4–6 months; deposi-
tion at 6 months; detect-
able CAA at 24 months.
No tangles;
phosphorylated tau in
dystrophic neurites
after 14 months.
Absent. Microglia and astrocytes
activation at 6–9 months.
Decreased sy-
naptic density
in DG at
8 months.
Alterations in
theta burst
stimulation
induced LTP
at 4–5 months.
Robust deficits
in RAM at
3 months.
(Dodart et al.,
1999;Domnitz et
al., 2005;Games et
al., 1995;Larson et
al., 1999;Masliah
et al., 2001)
APP V717I
(London)
APP (V717I)
Plaques in cortex and
subiculum at 10 months;
diffuse amyloid deposi-
tion and dense neuritis
plaques at 13–18 months;
amyloid deposition in
cerebral vessels and de-
creased Aβ42/40 ratio at
15 months.
No tangles;
hyperphosphorylated
tau at 16 months. Absent.
Microglia activation and
other neuro-inflammatory
markers increase at
10 months.
LTP deficit in
hippocampal
CA1 region
at 6 months.
Spatial and
non-spatial
orientation,
memory
impairment in
MWM and
associative
learning deficits
at 6 months.
(Moechars et al.,
1999;Van Dorpe et
al., 2000)
APP (V642I)KI Increased Aβ42(43)/40
ratio at 29 months. Absent. Absent. Long-term
memory deficit
at 27–29 months.
(Kawasumi et al.,
2004)
a) The table is ordered by mutation site alphabetically. “—” means no data.
7
Li, X., et al. Sci China Life Sci
Accumulating AβO within neurons of the hippocampus and
cortex were found at 8 months of age, but no amyloid pla-
ques even at 24 months of age (Tomiyama et al., 2010). The
mice exhibited hyperphosphorylated tau, decreased synaptic
density in the hippocampal CA3 region and memory im-
pairment at 8 months of age, microgliosis in the hippo-
campus and cortex at 12 months, astrocytosis at around
18 months, and neuronal loss in the CA3 region at 24 months
(Tomiyama et al., 2010). This mouse model was crossed with
other transgenic mice to study the combined role of two
genes in AD, such as tau264 mice (Umeda et al., 2014).
APP23 mice
This transgenic mouse model expresses human APP (iso-
form 751) containing the KM670/671NL (Swedish) muta-
tion under the murine Thy1 promoter, and has a 7-fold
overexpression of mutant human APP (Sturchler-Pierrat et
al., 1997). The mice first developed cognitive impairment at
3 months of age (Van Dam et al., 2003). deposits were
first observed at 6 months of age, visualized in neocortex by
pittsburgh compound B (PiB) positron emission tomography
(PET) at 12 months (Snellman et al., 2013). The mice also
showed age-associated increase in components of the com-
plement system, namely C1q and C3, at 9 months (Reich-
wald et al., 2009). Dense-core amyloid plaques were formed
at 6 months which increased with age and faster in female
mice, and occupied more than 25% of neocortex and hip-
pocampus at 24 months of age (Calhoun et al., 1998;
Sturchler-Pierrat et al., 1997). Plaques were surrounded by
activated microglia, astrocytes, and dystrophic neurites with
hyperphosphorylated tau, but NFTs were not found (Stalder
et al., 1999;Sturchler-Pierrat et al., 1997). The number of
hippocampal neurons decreased by 14%–28% at
14–18 months (Calhoun et al., 1998). This mouse model was
used to study the treatment of AD with anti-Aβ antibodies
and other drugs that block the formation of multimers
(Leinenga et al., 2021;Wilhelmus et al., 2022).
TAS10 (Thy1-APPswe) mice
This transgenic mouse overexpresses human APP (isoform
695) with the KM670/671NL (Swedish) mutation driven by
the murine Thy-1 promoter (Richardson et al., 2003). These
mice accumulated Aβ40 and Aβ42 in the cortex and hippo-
campus, and developed amyloid plaques by 12 months, with
higher overall levels in the cortex by 18 months (Richardson
et al., 2003). Cognitive impairment appeared in MWM at
6 months, and in Y maze at 12 months. The mice also ex-
hibited dystrophic neurites, lipid deposits, astrogliosis and
microgliosis in the dentate gyrus (DG) at 6 months. The mice
had more synapses than non-Tg mice before 12 and
18 months of age, but had fewer synapses by 24 months of
age (Richardson et al., 2003).
Tg2576 mice
This transgenic mouse model overexpresses human APP
(isoform 695) with the KM670/671NL (Swedish) mutation
driven by the hamster prion protein promoter (Hsiao et al.,
1996). The mice first showed synaptic morphological and
functional changes such as dendritic spine loss in CA1 region
and severely impaired LTP in CA1 and DG of the hippo-
campus at 4–5 months (Chapman et al., 1999;Lanz et al.,
2003), and impaired spatial learning, working memory and
contextual fear conditioning after 9 months of age (Hsiao et
al., 1996;Irizarry et al., 1997). Hemizygous mice developed
many plaques along with some vascular amyloid by
11–13 months (Irizarry et al., 1997), and significantly in-
creased microglial density and size in plaque-forming areas
of hippocampus, frontal, entorhinal and occipital cortex at 10
and 16 months (Frautschy et al., 1998). There was hyper-
phosphorylated tau in neurites around plaques at 16 months,
but no tangles or neuron loss (Irizarry et al., 1997;Puig et al.,
2004). These mice developed extensive CAA at 22 months
(Domnitz et al., 2005). This mouse model was widely used in
researches of pathogenesis and therapeutics of AD, and was
often used in constructing new AD models, such as 5×FAD
(Oakley et al., 2006). Aducanumab, bexarotene, memantine,
and high-fat diet treatment were studied using this mouse
model and showed beneficial effects on cognitive behaviors
(Cramer et al., 2012;Elhaik Goldman et al., 2018;Nagakura
et al., 2013;Sevigny et al., 2016). Oat extract avenan-
thramide-C prevented the impairment of hippocampal sy-
naptic plasticity in this model (Lee et al., 2021b).
PDAPP (line109) mice
This model has a PDGF-driven human APP minigene with
the V717F (Indiana) mutation, and overexpresses human
APP more than 10-fold higher than endogenous murine APP
(Games et al., 1995). The mice showed cognitive deficits in a
bar press learning task and radial arm maze (RAM) at
3 months, in spatial working memory in MWM at 4 months,
in recognition memory in novel object recognition (NOR)
task at 6 months, and worsened with age (Dodart et al., 1999;
Hartman et al., 2005). The mice showed changes in synaptic
transmission and plasticity including enhanced paired-pulse
facilitation, distorted responses to high frequency stimula-
tion bursts, and more rapidly decayed LTP at 4–5 months,
and decreased synaptic and dendrite density at 8 months
(Larson et al., 1999). deposition occurred in the hippo-
campus, corpus callosum and cerebral cortex at 6–9 months,
and plaques deformed extensively with age. Astrocytes and
activated microglia were associated with plaques. Tau
phosphorylation was observed in dystrophic neurites at
8Li, X., et al. Sci China Life Sci
16 months of age (Dodart et al., 1999;Games et al., 1995).
CAA was detectable at 24 months (Domnitz et al., 2005).
This mouse model was often used in the researches of pa-
thogenesis and therapeutics of AD. In this model, 2B3 (an
antibody that recognizes the β-secretase cleavage site on
APP but not Aβ) was reported to selectively affect APP
metabolism by reducing beta-site APP-cleaving enzyme 1
(BACE1) activity and reverse an age-dependent associative
recognition memory deficit (Evans et al., 2019), and sema-
gacestat (a γ-secretase inhibitor) showed sustained and dose-
dependent inhibition of production in plasma and cere-
brospinal fluid (CSF) (Karran and Hardy, 2014).
APP(V717I) mice
This model overexpresses human APP (isoform 695) with
the V717I (London) mutation driven by the Thy1 promoter
(Moechars et al., 1999). The earliest pathological manifes-
tations included reduced LTP, impairments in spatial and
non-spatial orientation, and deficits in memory in MWM and
associative learning at 6 months of age. Plaques began to
develop in the cortex and subiculum at 10 months. Diffused
amyloid deposits and compact neuritic plaques were ob-
served at 13–18 months, especially in the hippocampus and
cortex, with occasional deposits in the thalamus and fimbria,
external capsule, pontine nuclei, and white matter (Moechars
et al., 1999). Prominent amyloid deposits were observed in
brain vessels and microbleeds at 15 and 25–30 months of
age, respectively (Van Dorpe et al., 2000). The mice ex-
hibited microglia activation and increased neuro-in-
flammatory markers at 10 months, tau hyperphosphorylation
in dystrophic neurites at 16 months, but was absent of tan-
gles. This mouse model was used to generate mouse model to
study the combined role of two genes in AD, including APP
(V717I)×PS1(A246E) mice (Dewachter et al., 2000).
MAPT single-transgenic mouse models
MAPT encodes the microtubule associated protein tau, a
protein central to AD neuropathology. Human tau protein has
six isoforms, that differ in the number of amino-terminal
repeats (0N, 1N or 2N) and microtubule-binding repeats (3R
or 4R) (Götz et al., 2018). The transformation from 3R to 4R
is the basis for the development of NFTs (Hara et al., 2013).
However, there is only 4R isoform in mice, which cannot
naturally develop tangles with age, so MAPT transgenic mice
were established as tau pathological models for AD research
(Watamura et al., 2022a). MAPT mutations are commonly
found in frontotemporal dementia, and the A152T, K280Δ
and R406W mutations have been reported in sporadic AD
(Coppola et al., 2012). MAPT single mutation mouse models
are lack of pathology. Eleven mouse models carrying a
single mutation of MAPT with more than one pathological
change or cognitive impairments are shown in Table 3
(Bellucci et al., 2004;Boekhoorn et al., 2006;Cook et al.,
2014;Decker et al., 2016;Decker et al., 2015;Dennissen et
al., 2016;Eckermann et al., 2007;Gelman et al., 2018;
Hampton et al., 2010;Helboe et al., 2017;Hoover et al.,
2010;Kopeikina et al., 2013;Lasagna-Reeves et al., 2016;
Lewis et al., 2000;Maeda et al., 2016;Maurin et al., 2014;
Mocanu et al., 2008;Ramsden et al., 2005;SantaCruz et al.,
2005;Scattoni et al., 2010;Spires et al., 2006;Sydow et al.,
2016;Sydow et al., 2011;Takeuchi et al., 2011;Tanemura et
al., 2001;Tanemura et al., 2002;Tatebayashi et al., 2002;
Terwel et al., 2005;Van der Jeugd et al., 2012;Xu et al.,
2014;Yoshiyama et al., 2007;Yue et al., 2011). All MAPT
single mutant mice showed tau hyperphosphorylation, and
tangles were seen in most mice except hTau-AT or Tau
R406W Tg mice (Decker et al., 2016;Eckermann et al.,
2007;Lewis et al., 2000;Maeda et al., 2016;Mocanu et al.,
2008;Ramsden et al., 2005;Sydow et al., 2016;Tanemura et
al., 2001;Tatebayashi et al., 2002;Terwel et al., 2005;Xu et
al., 2014;Yoshiyama et al., 2007). Significantly neuronal
loss, gliosis, synaptic changes and cognitive dysfunction
were observed in hTau-AT, TauRDΔK280, JNPL3(P301L),
rTg4510, hTau.P301S and Tau P301S (PS19) mice (Bellucci
et al., 2004;Cook et al., 2014;Hampton et al., 2010;Helboe
et al., 2017;Kopeikina et al., 2013;Lewis et al., 2000;Maeda
et al., 2016;Mocanu et al., 2008;Ramsden et al., 2005;
SantaCruz et al., 2005;Scattoni et al., 2010;Spires et al.,
2006;Sydow et al., 2016;Sydow et al., 2011;Xu et al., 2014;
Yue et al., 2011). The MAPT mouse models commonly used
in AD studies are described below.
JNPL3(P301L) mice
JNPL3 mice were constructed using random transgenic
technology and overexpressing human tau (4R0N) with the
P301L mutation driven by the mouse prion promoter
(MoPrP) (Lewis et al., 2000). Homozygous JNPL3 mice
expressed human tau at about twice the endogenous level and
were most strongly expressed in the cerebellum and hippo-
campus (Lewis et al., 2000). NFTs appeared at 4.5 months in
homozygotes and 6.5 months in heterozygotes. NFTs were
morphologically heterogeneous, including flame- or glo-
bose-shaped NFT, Pick bodies and smaller, more irregular,
dense cytoplasmic inclusions (Lewis et al., 2000). Neuronal
loss, especially in the spinal cord, and astrogliosis near NFTs
were observed at 10 months (Lewis et al., 2000). 90% JNPL3
mice exhibited motor and behavioral problems by 10 months
(Lewis et al., 2000). This model is often used to evaluate the
effect of different types of tau antibodies on tau pathology of
AD, such as single domain antibodies (Congdon et al., 2022),
phosphorylated tau-specific intrabodies (Goodwin et al.,
2021), vectorized single chain variable fragment (Vitale et
al., 2020), and the interaction between and tau by
9
Li, X., et al. Sci China Life Sci
Table 3 Characteristics of MAPT single mutation mouse modelsa)
Mutation site Model name Tau Neuronal damage Gliosis Synaptic changes Changes in LTP/
LTD Cognitive dysfunc-
tion References
MAPT A152T
hTau-AT
(hTau40-AT)
Hyperphosphorylated tau
and tangles in
hippocampus, cortex,
cerebellum and spinal
cord at 3 months.
Neuronal loss in
hippocampus and
cortex at
12 months.
Astrocytosis and
microgliosis at
10 months.
Decreased synaptophysin
in hippocampus and cor-
tex at 12 months; in-
creased spines in CA3 at
12 months; decreased in
CA1 and CA3 at
16 months.
Absent at
20 months.
Learning and
memory deficits in
MWM at 16 months.
(Decker et al.,
2016;Sydow et al.,
2016)
hTau-A152T
No tangles; abnormal
accumulations and
phosphorylation of
soluble tau in neurons
at 8 months.
Neuron loss in
hippocampus at
20 months.
Astrocytosis at
6 months. No changes at
20 months.
Impaired nest
building at
10–14 months;
learning and
memory deficits
at 17 months.
(Maeda et al.,
2016)
MAPT
K280del
TauΔK280
(“Proaggregation
mutant”) Absent.
Widespread pre-tangle tau
pathology and
hyper-phosphorylation;
tangles after 24 months.
Absent.
Synaptic loss in
hippocampus at
13 months; pre- and
post-synaptic protein
reduction; decreased
number of dendrite
and spine.
LTP reduction
from DG to CA3
in hippocampus
at 12 months.
Cognitive deficit
at 16 months.
(Dennissen et al.,
2016;Eckermann
et al., 2007;Van
der Jeugd et al.,
2012)
TauRDΔK280
(“Proaggregation
mutant”) Absent.
Tangles at 2–3 months;
from entorhinal to
neocortex by
15 months.
Neuronal loss in
hippocampus at
5 months.
Astrocytosis in
hippocampus,
entorhinal and
piriform cortex
at 21 months.
Structural changes of
synapse in mossy fiber
and CA3; synaptic loss
in hippocampus after
9.5 months.
LTP and
long-term
depression
(LTD) deficits
after 10 months.
Learning and
memory deficits in
MWM and passive
avoidance test after
10 months.
(Decker et al.,
2015;Mocanu et
al., 2008;Sydow et
al., 2011)
MAPT P301L
JNPL3(P301L) Absent. NFTs at 4.5 months. Motor neuron loss,
especially in
spinal cord.
Astrogliosis in
brainstem,
diencephalon, and
basal telencephalon
by 10 months.
Motor impairment
and behavioral
problems at
10 months.
(Lewis et al., 2000)
rTg(tauP301L)
4510 Absent.
Pre-tangles at 2.5 months;
argyrophilic tangle-like
inclusions in cortex at
4 months and in
hippocampus at 5.5 months.
Decreased
hippocampal CA1
neurons by
5.5 months; cortical
cell loss at
8.5 months;
forebrain
atrophy at
10 months.
Microgliosis and
astrocytosis at
2.5 months.
Decreased pre- and
postsynaptic proteins
at 6–12 months;
dendritic spine loss
at 8–9 months.
Impaired LTP
at 4.5 months.
Spatial navigation
deficits in MWM
at 1.3 months;
spatial memory
deficits at
2.5–4 months;
fear conditioning
deficits at
2–6 months.
(Cook et al., 2014;
Gelman et al.,
2018;Helboe et al.,
2017;Hoover et al.,
2010;Kopeikina et
al., 2013;Ramsden
et al., 2005;Santa-
Cruz et al., 2005;
Spires et al., 2006;
Yue et al., 2011)
Tau P301L Absent.
Tau hyperphosphorylation
and conformational changes
at 7 months; tangle-like
pathology by 8 months;
age-dependent increase
in total tau.
Astrogliosis by
7 months.
LTP deficit in
hippocampal
CA1 region at
6 months.
Deficits in passive
avoidance task at
5 months; in NOR
task at 9 months;
motor impairment
at 7 months.
(Boekhoorn et al.,
2006;Maurin et al.,
2014;Terwel et al.,
2005)
(To be continued on the next page)
10 Li, X., et al. Sci China Life Sci
(Continued)
Mutation site Model name Tau Neuronal damage Gliosis Synaptic changes Changes in LTP/
LTD Cognitive dysfunc-
tion References
MAPT P301S
hTau.P301S Absent. NFTs at 4 months. Neuronal loss at
3 months.
Microglial
activation in brain
stem and spinal
cord at 5 months;
astrocytosis at
6 months.
Decreased spine density
and length in
hippocampus at
2.5 months.
Memory deficit in
MWM at
2.5 months.
(Bellucci et al.,
2004;Hampton et
al., 2010;Scattoni
et al., 2010;Xu et
al., 2014)
Tau P301S
(Line PS19) Absent.
Tangles in neocortex,
amygdala, hippocampus,
brain stem and spinal cord
at 6 months.
Neuron loss in
hippocampus at
8 months, and in
entorhinal cortex,
piriform cortex
and amygdala at
12 months.
Microgliosis at
3 months, espe-
cially in white
matter and spinal
cord;
increased
microgliosis by
6 months in
hippocampus,
amygdala,
entorhinal cortex
and spinal cord.
Decreased synaptophysin
in hippocampal CA3
region at 3–6 months.
Reduced LTP
in CA1 region
of hippocampus
at 6 months.
Impaired spatial
learning and memory
in MWM at
6 months; impaired
memory
in contextual fear
conditioning test at
7.5 months.
(Lasagna-Reeves et
al., 2016;Takeuchi
et al., 2011;
Yoshiyama et al.,
2007)
MAPT R406W Tau R406W
transgenic Absent.
Tau inclusions (mainly
filaments) in neurons of
forebrain, hippocampus
and amygdala at
18 months.
Impairmed contextual
and cued fear
conditioning at
16–23 months.
(Tatebayashi et al.,
2002)
MAPT V337M
(Seattle) Tau V337M Absent.
Phosphorylated and
ubiquitinated tau
aggregates in
hippocampus at
11 months; tangles
formation in
hippocampus between
4–11 months.
Irregularly shaped
neurons and
non-apoptotic
atrophic
degeneration in
hippocampus at
10 months;
decreased
hippocampal neural
activity at
15 months.
No differences in
MWM at
11 months.
(Tanemura et al.,
2001;Tanemura et
al., 2002)
a) The table is ordered by mutation site alphabetically. “—” means no data.
11
Li, X., et al. Sci China Life Sci
hybridizing with Tg2576 or 5×FAD mice models (Kang et
al., 2021;Kwan et al., 2022).
rTg(tauP301L)4510 mice
rTg(tauP301L)4510 mice express human tau (4R0N) with
the P301L mutation driven by a tetracycline operator up-
stream of a cytomegalovirus minimal promoter and a tetra-
cycline-controlled transactivator driven by forebrain-specific
Ca2+ calmodulin kinase II promoter system, which restricts
transgene expression to the forebrain and can be inhibited by
doxycycline (Ramsden et al., 2005). The expression of mu-
tant tau protein in rTg(tauP301L)4510 mice was about 13
times higher than that of endogenous tau protein, with the
highest in the hippocampus and cortex (Ramsden et al.,
2005). The model mice showed a series of behavioral
changes, such as spatial navigation deficits at 1.3 months,
impaired spatial reference memory at 2.5 months (Ramsden
et al., 2005), deficits in fear conditioning between 2 months
and 6 months (Cook et al., 2014), anxiety at 6 months and
motor dysfunction at 10 months (Cook et al., 2014;Ramsden
et al., 2005). The mice exhibited gliosis, pathological tau
conformation and phosphorylation in the hippocampus and
neocortex at 2.5 months, developed NTFs in the cortex at
4 months and hippocampus at 5.5 months (Helboe et al.,
2017;Ramsden et al., 2005). Neuronal loss first occurred in
DG region at 2.5 months, followed by CA1 and CA2/3 at
5.5 months, and later in cortex at 8.5 months (Spires et al.,
2006). Impaired LTP in the CA1 hippocampal region were
observed at 4.5 months (Hoover et al., 2010). Decreased pre-
and post-synaptic proteins began between 6 and 12 months,
decreased dendritic spine density occurred at 8.5 months
(Kopeikina et al., 2013). Brain weight decreased at 7 months
and forebrain atrophy was observed at 10 months (SantaCruz
et al., 2005). Doxycycline can suppress tau P301L expres-
sion at different time points, and delay tau pathology,
maintain the number of neurons and brain weight, and im-
prove spatial memory (Helboe et al., 2017;SantaCruz et al.,
2005). This model has also been used to evaluate the treat-
ment effects of trazodone, epicatechin, LDN/OSU-0215111
(a novel excitatory amino acid transporter 2 translational
activator) on tau pathology and cognitive deficits (de Oli-
veira et al., 2022;Foster et al., 2019;Hole et al., 2021).
Tau P301S (Line PS19) mice
Tau P301S (Line PS19) mice were constructed using random
transgenic technology and overexpressing human tau
(4R1N) with the P301S mutation driven by the MoPrP
(Yoshiyama et al., 2007). The expression of mutant tau
protein in Tau P301S (Line PS19) mice was about 5 times
higher than that of endogenous tau protein (Yoshiyama et al.,
2007). This model showed tau hyperphosphorylation, im-
paired axonal transport, microgliosis, decreased synapto-
physin, and motor deficits as early as 3 months (Yoshiyama
et al., 2007). Increased inflammatory factors in CA3 region
and synaptic dysfunction in CA1 region of the hippocampus
occurred at 4 and 6 months, respectively (Yoshiyama et al.,
2007). NFTs were observed in the neocortex, amygdala,
hippocampus, brain stem and spinal cord at 6 months with
progressive accumulation (Yoshiyama et al., 2007). Astro-
gliosis was seen in white and gray matter of hippocampus,
amygdala, entorhinal cortex, and spinal cord at 6 months
(Yoshiyama et al., 2007). At 6 months of age, the mice
showed a variety of cognitive impairments, including
working memory, spatial learning and memory, contextual
memory, sociability, and object recognition (Takeuchi et al.,
2011). Neuron loss in the hippocampus at 8 months, and in
the entorhinal cortex, piriform cortex and amygdala at
12 months (Yoshiyama et al., 2007). Remarkable atrophy of
hippocampus and entorhinal cortex plus ventricular dilata-
tion were detected at 9 months (Yoshiyama et al., 2007). 80%
PS19 mice died by 12 months (Yoshiyama et al., 2007). This
model has been used to evaluate the therapeutic effect of
metformin, tryptophan-tyrosine-related β-lactopeptides,
ibrutinib on tau pathology and tau-related cognitive impair-
ment (Ano et al., 2023;Lee et al., 2021a;Zhao et al., 2023).
Studies have also combined this model with AD risk factors,
such as Aβ42, APOE and TREM2, to study their effects on
tau pathology (Gratuze et al., 2020;Williams et al., 2022;
Zampar and Wirths, 2021).
Other AD-related single gene mouse models
Some single gene mouse models carrying AD-related genes
with AD pathological change and cognitive impairment are
summarized in Table 4 (Heckmann et al., 2020;Pensalfini et
al., 2020;Plucińska et al., 2014;Zhong et al., 2022). These
genetically modified models in the background of murine
APP can cause AD-related pathological changes and cog-
nitive impairment, thus are important to elucidate novel AD
pathogenesis and potential therapeutic targets such as au-
tophagy, neuroinflammation, endosomal dysfunction, cal-
cium dysregulation, and palmitoylation.
ATG16L encodes the part of a large protein complex that is
necessary for autophagy, and its expression level is down-
regulated in AD patients (Heckmann et al., 2020). Mice
lacking the WD domain of ATG16L showed age-associated
development of AD pathologies, such as deposits, tau
hyperphosphorylation, neurodegeneration, microgliosis,
neuroinflammation, and impaired synaptic plasticity.
MCC095, a brain-penetrant inflammasome inhibitor, ame-
liorated AD pathology and cognitive impairment in ATG16L
mice (Heckmann et al., 2020). BACE1 encodes β-site APP
cleaving enzyme 1, which directly relates to plaque
formation and APP metabolism. The PLB4 (hBACE1) mice
12 Li, X., et al. Sci China Life Sci
Table 4 Characteristics of other AD-related gene mouse modelsa)
Gene Model name Tau Neuronal damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive
dysfunction References
ATG16L Atg16LΔWD
Elevated extracellar
and intraneural Aβ,
but no plaque at
2 years.
Pervasive
hyperphosphorylation
of microtubule-stabilizing
protein tau in
hippocampus with
pronounced accumulation
in CA3 field at 2 years.
Apoptotic death,
apoptosis and
suggestive of active
neurodegeneration,
reduction in total
neurons within
hippocampus at
2 years.
exacerbated
activation of
microglia in
hippocampi at
2 years.
substantial
reduction in LTP
at CA3-CA1
synapses at
2 years.
Impaired
performances in
sucrose preference
test, Y-maze, and
NOR at 2 years.
(Heckmann et al.,
2020)
BACE1 PLB4
(hBACE1)
Elevated
extracellular and
intraneural at
12 months; plaques
virtually absent,
minimal small
sparse plaques;
extracellular
staining includes
multimers.
Preliminary analysis did
not find abnormal
phosphorylation or
conformational changes
in tau. Increase trends
of total tau levels at
6 months.
Increased
astrogliosis in DG,
CA1 region of
hippocampus, and
piriform cortex at
12 months. Gliosis
is suspected to
begin earlier than
12 months.
Impaired spatial
representation in a
habituation task at
3 months; impaired
learning and
memory in Y-maze,
MWM, and a test
of social transmis-
sion of food pre-
ference at
6 months.
(Plucińska et al.,
2014)
GluN3A GluN3A KO
Significant
depositions of
endogenous
overlaid with
neurons in
hippocampus at
10–15 months.
Increased total tau
expression in cortex at
6–7 months; tau
hyperphosphorylation in
both hippocampus and
cortex at 10–15 months.
Neurodegeneration
of olfactory sensory
neurons at 2–3 months;
neuron loss in olfactory
bulb and hippocampal
CA3 region at
10–12 months;
increased apoptotic
marker cleaved by
caspase-3 in cortex
at 10–12 months.
Reactive astrocytes
in hippocampus
at 10–15 months.
Decreased level of
synapsin in
hippocampus at
7 months; no
significant
difference in ex-
pression of
synaptophysin and
PSD-95.
Enhanced LTP in
hippocampus at
2–4 months;
suppressed LTP
in hippocampus
at 6–7 months.
Impaired
recognition mem-
ory and slower
learning at
5–6 months;
impaired spatial
working memory
at 7 months; poor
social novelty
performance at
5–12 months.
(Zhong et al., 2022)
Rab5a PA-Rab5
Levels of total tau and
tau hyperphosphorylation
are increased in cortices
at 15 months.
Loss of basal forebrain
cholinergic neurons
started at 7 months;
abnormal swelling and
thickening of proximal
dendrites and dys-
morphic cell bodies.
Loss of spines in
CA3 and DG
regions of
hippocampus at
8.5 months, but not
in CA1 region.
Defect in LTD and
slight impairment
in LTP in hippo-
campal slices at
Schaffer collateral-
CA1
synapses from
6 months.
Reduced retention
in NOR test in
6-month-old
mice.
(Pensalfini et al.,
2020)
ZDHHC21 ZDHHC21
T209S KI
Increased
intraneural Aβ42
in hippocampus
at 9 months
Phosphorylated tau
aggregates in
hippocampus at
9 months.
Neuronal loss in
hippocampus at
9 months.
Decreased synapse
density in hippo-
campus at
6 months; reduced
number of dendritic
branches and
spines in
hippocampus at
9 months.
Reduced LTP in
hippocampus at
3 months
Impaired spatial
learning and
memory ability in
MWM at
9 months.
(Li et al., 2023)
a) The table is ordered by mutation site alphabetically. “—” means no data.
13
Li, X., et al. Sci China Life Sci
showed accumulation of Aβ56 and hexamers without
plaques, gliosis, neuroinflammation and cognitive impair-
ment relevant to AD, which suggests that mouse APP can be
cleaved by human BACE1 (Plucińska et al., 2014). GluN3A
encodes a subunit of the NMDA receptors that belong to the
superfamily of glutamate-regulated ion channels, and func-
tions in physiological and pathological processes in the
central nervous system (Zhong et al., 2022). GluN3A KO
mice developed olfactory dysfunction followed by cognitive
deficits prior to and tau pathology, and also showed
neuronal hyperactivity, calcium dysregulation, neuroin-
flammation, impaired synaptic plasticity and neuronal loss
(Zhong et al., 2022). Rab5 is a small GTPase that regulates
endosome trafficking, sorting, and fusion and involves
clearance. Rab5-mediated endosomal dysfunction occurs
very early in AD. Overexpressing human Rab5 in mice
neuron led to tau hyperphosphorylation, basal forebrain
cholinergic neurodegeneration, impaired LTP and LDP,
hippocampal spine abnormalities, endosomal dysfunction
and memory deficits (Pensalfini et al., 2020). ZDHHC21
encodes zinc finger DHHC-type palmitoyltransferase 21,
and enables palmitoyltransferase activity (Pedram et al.,
2012). The T209S mutation of ZDHHC21 was found in a
Han Chinese family with AD. Homozygous mice carrying
this mutation showed elevated palmitoylation of APP and
Fyn, resulting in AD-related pathologies including ac-
cumulation, tau hyperphosphorylation, deficits in LTP, de-
creased dendritic spines and branches, and cognitive
impairment (Li et al., 2023).
Heterogeneity in single transgenic mouse models
Taken together, most PSENs transgenic models are lack of
gliosis or synaptic changes, except PS1V97L Tg mice. Most
APP transgenic models are lack of tau tangles and neuronal
damages. None of the PSENs or MAPT transgenic models
show plaques. The possible reasons for lacking pla-
ques might be due to the differences between mouse and
human APP. Studies have shown that when mice over-
expressing or knocking in human PSEN1 mutation are bred
with human APP mutant mice, plaque deposition begins at
an earlier age and increases with aging as compared to the
parental APP lines (Borchelt et al., 1997;Dewachter et al.,
2000;Elder et al., 2010;Holcomb et al., 1998;Lamb et al.,
1999;McGowan et al., 1999). These data indicate that a
human APP might be necessary for the formation of
plaques.
Figure 2 shows the ethnic origin of AD pathogenic gene
mutation sites used in all AD mouse models, including APP,
PSENs and ZDHHC21. Three mutations have been reported
only in Asian, namely APP E693Δ(Osaka) mutation from
two Japanese AD families (Tomiyama and Shimada, 2020),
PSEN1 V97L mutation (Jia et al., 2005) and ZDHHC21
T209S mutation (Li et al., 2023) from two Chinese AD fa-
milies. These three mutations have been used to construct
mouse models to explore genetic mechanism and drug
treatment (Tomiyama et al., 2010;Wang et al., 2016), and
provide new ideas and therapeutic targets for AD drug de-
velopment. Two mutations have been reported only in
American or European populations, including APP KM670/
671NL (Swedish) mutation from two Sweden families
(Mullan et al., 1992) and APP I716V (Florida) mutation from
one American family. APP KM670/671NL (Swedish) mu-
tation is the most commonly used mutation to establish
mouse models, which can promote plaque formation and
cognitive impairment, and BACE1 was confirmed as a key
driver of AD and responsible for plaque formation based on
these mutant mouse models (Armbrust et al., 2022). How-
ever, clinical trials of BACE1 inhibitors have all failed, and it
is necessary to rethink the importance of N-terminal trun-
cated species and alternative β-secretases as therapeutic
targets (Armbrust et al., 2022). APP KM670/671NL
(Swedish) and I716V (Florida) were both used in the con-
struction of 5×FAD mice, the most commonly used mouse
models for AD. However, it is unknown whether the po-
tential mechanisms revealed by these mutant mouse models
are unique to each racial/ethnic group or apply to other ra-
cial/ethnic groups as well, which needs further research and
verification.
Comparisons between multi-transgenic mouse
models
Multi-transgenic mouse models with more than one patho-
logical and cognitive changes are summarized in Table 5
(Adalbert et al., 2009;Arancio et al., 2004;Arendash et al.,
2001;Baglietto-Vargas et al., 2021;Bellucci et al., 2007;
Billings et al., 2005;Borchelt et al., 1997;Borchelt et al.,
1996;Bouter et al., 2013;Brautigam et al., 2012;Breyhan et
al., 2009;Buskila et al., 2013;Cacciottolo et al., 2021;
Caruso et al., 2013;Casas et al., 2004;Cheng-Hathaway et
al., 2018;Cheng et al., 2004;Cheng et al., 2007;Chishti et
al., 2001;Codita et al., 2010;Colton et al., 2008;Crouzin et
al., 2013;Davis et al., 2004;Davis et al., 2006;Dewachter et
al., 2000;Domnitz et al., 2005;Dudal et al., 2004;Flanigan
et al., 2014;Garcia-Alloza et al., 2006;Gengler et al., 2010;
Giannoni et al., 2016;Gordon et al., 2002;Gratuze et al.,
2020;Grueninger et al., 2010;Hashimoto et al., 2019;Havas
et al., 2011;Holcomb et al., 1998;Hong et al., 2016;Howlett
et al., 2008;Howlett et al., 2004;Hsia et al., 1999;Hu et al.,
2018;Hyman and Tanzi, 2019;Jackson et al., 2013;Jackson
et al., 2016;Jankowsky et al., 2004;Jankowsky et al., 2005;
Jankowsky et al., 2001;Jawhar et al., 2012;Jay et al., 2017;
Kamphuis et al., 2012;Kang et al., 2021;Kim and Jeong,
2015;Kimura et al., 2012;Kimura and Ohno, 2009;Kno-
14 Li, X., et al. Sci China Life Sci
Table 5 Characteristics of AD mouse model with multiple gene mutationsa)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APOE,
APP,
PSEN1
APP KM670/671NL
(Swedish), I716V
(Florida), V717I
(London); PSEN1
M146L (A>C),
L286V
E2FAD Plaques at 4 months and
increase with age.
Microgliosis and
astrocytosis in
subiculum and
cortex at
6 months.
Decreased synap-
tic proteins of
NMDA receptor
subunits at
2–6 months.
Better than E4FAD
mice, and compar-
able to E3FAD mice
in Y maze and
MWM.
(Youmans et al.,
2012)
E3FAD
Plaques at 4 months and
increase with age; in-
creased deposits
primarily in cerebral
blood vessels at
6 months.
Microgliosis and
astrocytosis in
subiculum and
cortex at
6 months.
Decreased synap-
tic proteins of
NMDA receptor
subunits at
2–6 months.
Better than E4FAD
mice, and compar-
able to E2FAD mice
in Y maze and
MWM.
(Cacciottolo et
al., 2021;You-
mans et al., 2012)
E4FAD
Plaques at 4 months and
increase with age; in-
creased deposits
primarily in cerebral
blood vessels at
6 months.
Microgliosis and
astrocytosis in
subiculum and
cortex at
6 months.
Decreased protein
levels of PSD95
and NMDA re-
ceptor subunits by
4 months.
Learning deficits in
MWM by
2 months; progres-
sive decrease in
learning and mem-
ory tasks.
(Cacciottolo et
al., 2021;You-
mans et al., 2012)
APP
Aβ4-42 sequence
fused to murine thyr-
otropin-releasing hor-
mone (TRH) signal
peptide
Tg4-42
No plaques; Aβ4-42
expression in hippo-
campal CA1 region and
detectable around
2 months.
Absent.
Age- and dose-de-
pendent hippocam-
pal neuronal loss
in CA1 region;
hemizygotes had
38% neuronal loss
at 8 months, and
49% loss at
12 months.
Reactive micro-
glia and astro-
cytes in
hippocampus at
2 months.
Altered synapto-
physin staining in
hippocampal CA3
region at
8 months.
Age-dependent spa-
tial learning deficit
in MWM at
8 months; impaired
contextual fear con-
ditioning at
12 months.
(Bouter et al.,
2013)
APP KM670/671NL
(Swedish), S679C TgDimer Intracellular depos-
its in hippocampus and
cortex at 12 months.
Absent through
24 months. Absent through
24 months. Absent through
24 months.
Impaired LTP in
Schaffer collat-
eral-CA1 synaptic
system at
6–8 months.
Learning deficits in
MWM at 7 months. (Müller-Schiff-
mann et al., 2016)
APP KM670/671NL
(Swedish), E693G
(Arctic) ArcAβ
Intracellular punctate
deposits in cortex
and hippocampus;
abundant plaques by
9–15 months.
Absent.
Impaired LTP in
hippocampus
slices at 3.5–7.5
months.
Cognitive impair-
ment in MWM and
Y maze at
6 months.
(Knobloch et al.,
2007a;Knobloch
et al., 2007b)
APP KM670/671NL
(Swedish), E693G
(Arctic) Tg-ArcSwe
Extracellular amyloid
plaque deposition starts
at around 5–6 months in
cortex, hippocampus,
and thalamus; CAA in
cerebral cortical vessels
at 12 months.
Absent. Absent.
Microgliosis and
astrogliosis most
prominent in hip-
pocampus, also
around deposits in
cortex and thala-
mus.
Spatial learning im-
pairment in MWM
at 4–8 months and
in passive avoid-
ance test at
16 months.
(Codita et al.,
2010;Lillehaug et
al., 2014;Lord et
al., 2009;Lord et
al., 2006;Skaar-
aas et al., 2021)
APP KM670/671NL
(Swedish), E693Q
(Dutch), D694N
(Iowa)
Tg-SwDI
(APP-Swedish,
Dutch, Iowa)
deposits in subicu-
lum, hippocampus and
cortex at 3 months;
more numerous at
6 months; throughout
most forebrain by
12 months; microvascu-
lar amyloid deposits at
24 months.
Absent.
Pronounced in-
creased astrocytes
and activated mi-
croglia at
6–24 months
especially in tha-
lamus and subi-
culum.
Impaired learning
and memory in
Barnes maze task at
3, 9, and 12 months.
(Davis et al.,
2004;Davis et al.,
2006;Miao et al.,
2005b;Xu et al.,
2007)
(To be continued on the next page)
15
Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP
APP KM670/671NL
(Swedish), T714I
(Austrian)
A7 APP
transgenic
Amyloid deposition in
cortex at 9–12 months;
extensive deposition at
21 months.
(Yamada et al.,
2009)
APP KM670/671NL
(Swedish), V717F
(Indiana)
J20 (PDGF-
APPSw, Ind)
Diffuse plaques in
DG and neocortex at
5–7 months; widespread
plaques by
8–10 months.
Absent.
Decreased neuronal
numbers in hippo-
campal CA1 region
at 3, 6 and
9 months.
Significant in-
crease in the
number of astro-
cytes and micro-
glia in
hippocampus at
4–6 months
Loss of hippocam-
pal synaptophysin,
synaptotagmin,
PSD-95 and homer
immunoreactivity
by 3 months; im-
paired basal sy-
naptic transmission
at 3–6 months.
Impaired LTP at
Schaffer collat-
eral-CA1 synapse
at 3–6 months.
Spatial reference
memory deficits as
measured by RAM
and MWM at
4 months.
(Cheng et al.,
2007;Hong et al.,
2016;Mucke et
al., 2000;Wright
et al., 2013)
PDGF-APPSw,
Ind (line J9)
20% had plaques at
5–7 months, 50% at
8–10 months, and 100%
by 21–25 months.
Deficits in synap-
tic transmission at
2–4 months. (Hsia et al., 1999;
Mucke et al.,
2000)
TetO-APPS-
weInd (line
102, 107, 885)
Progressive amyloid
plaques at 2 months;
extensive amyloid
pathology in cortex and
hippocampus by
9 months.
(Jankowsky et al.,
2005)
TgCRND8
Amyloid deposition at
3 months; plaques by
5 months; first in sub-
iculum, amygdala and
frontal cortex; severe
vascular deposition
at 11 months.
Absent of NFTs;
hyperphosphory-
lated, nitrosy-
lated and
aggregated tau at
7–12 months
especially in
neocortex, DG,
and hippocampal
CA1/CA3 areas.
Neuritic pathology
by 5 months; fewer
hippocampal neu-
rons at 6 months.
Microglia activa-
tion and robust
astrogliosis in
cortex and hippo-
campus by
3 months; numer-
ous by 5 months.
Reduced synapto-
physin immunor-
eactivity at
6 months; re-
duced excitatory
synaptic trans-
mission at
6–12 months.
Reduced LTP in
hippocampal
slices at 6–12
months.
Impaired acquisi-
tion and reversal
learning in MWM
at 3 months.
(Adalbert et al.,
2009;Bellucci et
al., 2007;Brauti-
gam et al., 2012;
Chishti et al.,
2001;Domnitz et
al., 2005;Dudal
et al., 2004;Ki-
mura et al., 2012)
APP KM670/671NL
(Swedish), V717F
(Indiana), E693G
(Arctic)
Arc48 (APPSw/
Ind/Arc)
Parenchymal neuritic
plaques by 2 months;
prominent hippocampal
plaques by 3–4 months;
abundant plaques by
10–12 months.
Absent. Dystrophic neurites
by 10–12 months.
Reactive astrocy-
tosis at
3–4 months in
DG.
Impaired ability to
use extramaze cues
to navigate to the
hidden platform in
MWM at 3–4
months
(Cheng et al.,
2004;Cheng et
al., 2007)
APP KM670/671NL
(Swedish), V717I
(London)
APPSweLon Increased Aβ42 at
3–4 months; no amyloid
deposits by 24 months. (Lamb et al.,
1999;Lamb et al.,
1997)
mThy1-
hAPP751
(TASD41)
Increased Aβ40, Aβ42
and AβOs at 6 months;
plaques at 3–6 months
in frontal cortex.
Absent. Dystrophic neurites
at 12 months.
Inflammation re-
lated to activated
microglia and re-
active astrocytes
by 6 months.
Synaptic loss at
12 months. Learning and mem-
ory deficits in
MWM at 6 months.
(Havas et al.,
2011;Rocken-
stein et al., 2001;
Rockenstein et
al., 2005)
rTg9191
Plaques in cerebral cor-
tex at 8 months; in hip-
pocampus at 10.5–
12.5 months.
Hyperpho-
sphorylated tau
develops with
age.
Reduced forebrain
weight and smaller
DG at 2–6 months.
Astrocytosis and
microgliosis near
plaques at
24 months.
Absent. (Liu et al., 2015)
(To be continued on the next page)
16 Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP
Glutamate at position
three of amino
acid sequence was
mutated into gluta-
mine
TBA42
accumulates in hip-
pocampal neurons at
3 months and cerebellar
nuclei by 6 months;
(pE3-42) accumulates in
spinal cord and pyrami-
dal neurons of motor
cortex at 12 months.
Absent.
35% neuronal loss
in hippocampal
CA1 region at
12 months.
Marked gliosis in
hippocampus at
12 months.
Working and spatial
reference memory
deficits at
12 months.
(Wittnam et al.,
2012)
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H)
hAβ-KI
Decreased soluble
and increased insoluble
Aβ; no plaques up to
22 months.
Absent.
No cell loss in pyr-
amidal layer of hip-
pocampal CA1
region, but signifi-
cant hippocampal
volume differences at
22 months
Astrocytosis in
hippocampus at
22 months.
Reduction of sy-
naptophysin and
PSD95 expres-
sion in hippo-
campus at
18 months.
Impaired LTP at
18 months.
Cognitive impair-
ment in contextual
fear conditioning at
10 months, and
NOR at 14 months.
(Baglietto-Vargas
et al., 2021)
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H); APP
KM670/671NL
(Swedish)
APPSwe (line
C3-3)
deposition at
18–20 months; plaques
formation at
24–26 months.
—— ———Absent at
12-14 months. (Savonenko et al.,
2003)
APPSwe (line
E1-2)
increase slightly at
6–14 months, and sig-
nificantly at
24–26 months.
—— ———Cognitive deficits at
12-13 months. (Savonenko et al.,
2003)
TgAPPSwe-KI
Absent of mouse de-
rived overexpres-
sion; increased human
derived deposition.
(Reaume et al.,
1996)
Humanize mouse
region: G676R (G5R),
F681Y (F10Y),
R684H (R13H); APP
KM670/671NL
(Swedish), E693G
(Arctic), T714I (Aus-
trian)
AppSAA KI
Increased Aβ42 to 40
ratio in brain, CSF and
plasma at 2 months;
plaques in homozygotes
at 4 months, most pro-
nounced in cortex and
hippocampus.
No NFTs at
8 months; in-
creased CSF
total tau in
homozygotes at
8 months.
AT8-positive dys-
trophic neurites in
homozygous mice
at 8 months.
Plaque-associated
microgliosis at
4 months. (Xia et al., 2021)
Humanize mouse
region: G676R (G5R),
F681Y (F10Y),
R684H (R13H); APP
KM670/671NL
(Swedish), E693G
(Arctic), I716F (Iber-
ian)
APPNL-G-F KI Plaques at 2 months
with near saturation by
7 months.
No tangles; ele-
vated levels of
phosphorylated
tau in dys-
trophic neur-
ites.
Dystrophic neurites
Microglia and ac-
tivated astrocytes
accumulate at
2 months, concur-
rent with plaque
formation.
Changes in synap-
tic microstructure
and reduction of
synaptophysin and
PSD95 immunor-
eactivities in cor-
tices and
hippocampus at
6 months.
Memory impair-
ment in Y maze test
at 6 months, and in
NOR at 9 months.
(Liu et al., 2021b;
Mehla et al.,
2019;Nilsson et
al., 2014;Saito et
al., 2014)
Humanize mouse
region: G676R (G5R),
F681Y (F10Y),
R684H (R13H); APP
KM670/671NL
(Swedish), I716F
(Iberian)
APPNL-F KI
Increased Aβ42/Aβ40
ratio and plaques in
cortex and hippocampus
at 6 months.
Elevated phos-
phorylated tau
in dystrophic
neurites at
6 months.
Dystrophic neurites
at 6 months.
Microglia and ac-
tivated astrocytes
accumulate at
6 months, concur-
rent with plaque
formation.
Reduced synapto-
physin and
PSD95 immunor-
eactivities at
9–12 months.
Memory impair-
ment in Y maze at
18 months; absent
in MWM at
18 months.
(Saito et al.,
2014)
(To be continued on the next page)
17
Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H); APP E693G
(Arctic), I716F (Iber-
ian)
AppG-F KI
deposition in the
cortex at 4–6 months, in
the hippocampus at
8–12 months.
Reactive astro-
cyte and activated
microglia around
plaques in the
cortex at
22 months.
Decreased synap-
tophysin and
postsynaptic den-
sity protein 95
immunoreactivity
in hippocampus at
22 months.
(Watamura et al.,
2022b)
APP,
BACE1,
PSEN1
APP KM670/671NL
(Swedish), I716V
(Florida), V717I
(London); PSEN1
M146L (A>C),
L286V
BACE1 cKO
(Hu, Yan)×5×-
FAD
Plaques accumulate up
to 4 months and even-
tually disappear.
Dystrophic neurites
accumulate up to
4 months, to a lesser
degree than in con-
trol 5×FAD.
Reactive astro-
cytes and micro-
glia accumulate
up to 4 months.
Deficit in LTP at
Schaffer collat-
eral-CA1 sy-
napses by
10–12 months.
Normal contextual
and cued fear con-
ditioning at
8–10 months.
(Hu et al., 2018)
APP,
MAPT
APP KM670/671NL
(Swedish); MAPT
P301L
Tg2576/Tau
(P301L)
(APPSwe-Tau) Plaques at 6–7 months.
Tangles in
spinal cord and
pons at
3 months; in-
creased in lim-
bic areas by
6 months.
Reactive astro-
cytes and micro-
glia at 3 months
in hippocampus;
increased astrocy-
tosis in limbic
areas; microglia
around plaques at
9 and 12 months.
(Lewis et al.,
2001)
Human Aβ4-42 se-
quence; MAPT P301S PS19/Tg4-
42hom Amyloid deposition at
3 months.
Phosphorylated
tau accumula-
tions at
3 months; abun-
dant tau pathol-
ogy with NFTs
in CA1 pyrami-
dal layer at
9 months.
Neuron loss in dis-
tal and proximal
CA1 at 3 months. ———
Impaired motor and
spatial learning at
5 months; impaired
recognition memory
at 9 months.
(Zampar and
Wirths, 2021)
Humanize mouse
region: G676R (G5R),
F681Y (F10Y), R684H
(R13H); APP KM670/
671NL (Swedish),
E693G (Arctic), I716F
(Iberian)
AppNL-G-F/
MAPT KI Plaques at 2 months. Absent. Absent. Astrogliosis and
microgliosis by
4 months. Working memory
deficits in Y-maze
at 12 months
(Hashimoto et al.,
2019;Saito et al.,
2019)
APP,
MAPT,
PSEN1
APP KM670/671NL
(Swedish); MAPT
P301L; PSEN1
M146V
3×Tg
Intraneuronal
pathology in hippocam-
pus, cortex and amyg-
dala at 4 months;
extracellular depos-
its in frontal cortex by
6 months.
Extensive tau
immunoreactiv-
ity in hippo-
campal CA1
pyramidal neu-
rons by
12 months, later
in cortex.
Increased density
of astrocytes and
microglia at
7 months.
Impaired basal
synaptic trans-
mission at
6 months
Decreased LTP at
6 months
Long-term retention
deficit at 4 months;
spatial learning and
memory deficit in
Barns maze at
6.5 months.
(Billings et al.,
2005;Caruso et
al., 2013;Oddo et
al., 2003;Stover
et al., 2015)
APP KM670/671NL
(Swedish), I716V
(Florida), V717I (Lon-
don); MAPT P301L;
PSEN1 M146L (A>C),
L286V
6×Tg
Aβ42 accumulation at
2 months; plaques in
cortex and hippocampus
at 4 months.
Hyperpho-
sphorylated tau
at 4 months;
develops with
age.
Neuronal loss in
cortex at 4 months
and in hippocampus
at 6 months.
Microglial activa-
tion in cortex and
hippocampus at
2 months.
Synaptic loss in
cortex from
4 months, and in
hippocampus
from 6 months.
Decreased cognitive
and memory capa-
city at 2 months.
(Kang et al.,
2021)
(To be continued on the next page)
18 Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP,
MAPT,
PSEN1
APP KM670/671NL
(Swedish), V717I
(London); MAPT
P301L, R406W;
PSEN1 A246E
PLB1-triple
(hAPP/hTau/
hPS1)
Sparse plaques by
21 months; intracellular
accumulation and
formed oligomers.
Tau hyperpho-
sphorylation in
hippocampus
and cortex from
6 months.
Increased inflam-
mation at
12 months in cor-
tex and hippo-
campus.
Impaired synaptic
transmission at
12 months.
Reduced LTP and
paired-pulse fa-
cilitation at 6 and
12 months.
Reduced social re-
cognition at 5 and
12 months; object
recognition at
8 months spatial
learning at
12 months.
(Platt et al., 2011)
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H); APP
KM670/671NL
(Swedish); PSEN1
deltaE9
APP/PS1/
rTg21221
Cortical plaques ob-
served between
8–10 months.
Aggregates of
misfolded and
phosphorylated
tau between
8–10 months.
Neuronal loss near
plaques. Astrocytosis near
plaques.
Decreased sy-
napse density
near plaques. (Jackson et al.,
2016)
APP,
MAPT,
PSEN2
APP KM670/671NL
(Swedish); MAPT
P301L; PSEN2 N141I TauPS2APP
Rare amyloid plaques at
4 months; increase in
subiculum and hippo-
campal CA1 region by
8 months.
Abnormally
phosphorylated
tau at 4 months,
in subiculum,
amygdala, and
hippocampal
CA1 region;
tangle-like de-
posits in hippo-
campus by
16 months.
Absent at
16 months. ———
Impaired spatial
learning in MWM
at 4 months, but not
progressive be-
tween 4 and
12 months, and in-
dependent of
pathology.
(Grueninger et al.,
2010)
APP,
NOS2
APP KM670/671NL
(Swedish), E693Q
(Dutch), D694N
(Iowa)
APPSwDI×-
NOS2 KO
Plaques especially in
thalamus and subicu-
lum.
Aggregated,
hyperpho-
sphorylated tau
tangles.
Neuronal loss in
hippocampus and
subiculum. ———
Learning and mem-
ory deficits mea-
sured by RAM and
Barnes maze at
12–13 months.
(Colton et al.,
2008;Wilcock et
al., 2008)
APP,
Pdgfrβ
APP KM670/671NL
(Swedish); Pdgfrβ
gene deleted
APPsw/0;
Pdgfrβ+/-
Elevated plaque load in
cortex and hippocampus
at 9 months.
Tau hyperpho-
sphorylation in
cortical and
hippocampal
neurons at
9 months.
Reduced neurite
density and neuro-
nal loss in cortex
and hippocampus at
9 months.
———
Impaired nest
building, burrow-
ing, and NOR at
9 months.
(Sagare et al.,
2013)
APP,
PSEN1
APP KM670/671NL
(Swedish), I716V
(Florida), V717I
(London); PSEN1
M146L (A>C),
L286V
5×FAD
(B6SJL)
Extracellular amyloid
deposition in cortex at
2 months; throughout
hippocampus and cortex
by 6 months.
Absent.
Neuron loss in cor-
tical layer 5 and
subiculum at
6 months.
Gliosis at
2 months.
Decreased presy-
naptic marker sy-
naptophysin by
4 months; de-
creased presynap-
tic marker syntaxin
and postsynaptic
marker PSD-95 by
9 months.
Deteriorated LTP
in hippocampal
CA1 at
4–6 months; defi-
cits in basal sy-
naptic transmis-
sion by 6 months.
Impaired spatial
working memory in
Y-maze and MWM
by 4–6 months;
impaired remote
memory
stabilization by
4–5 months;
impaired learning
at 9 months;
impaired social
recognition at
9–12 months.
(Crouzin et al.,
2013;Flanigan et
al., 2014;Kimura
and Ohno, 2009;
Oakley et al.,
2006;Xiao et al.,
2015)
(To be continued on the next page)
19
Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP,
PSEN1
APP KM670/671NL
(Swedish), I716V
(Florida), V717I
(London); PSEN1
M146L (A>C),
L286V
5×FAD
(C57BL6)
Plaques in hippocam-
pus, cortex, and thala-
mus at 2 months, and in
spinal cord by
3 months; progressive
CAA on leptomeningeal
and penetrating vessels
started at 3 months.
40% neuron loss in
cortical layer V at
12 months.
Plaques asso-
ciated microglio-
sis and
astrogliosis; vas-
cular damage as-
sociated micro-
gliosis.
Reduced spine
density in pyra-
midal neurons in
somatosensory
and prefrontal
cortices at
6 months.
Layer V neurons
from 2–3 months
old mice have lost
the ability to un-
dergo LTP and
have a small yet
significant in-
crease in LTD
following spike-
timing-dependent
plasticity induc-
tion.
Impaired spatial
working memory at
3–6 months and
worsen with age.
(Buskila et al.,
2013;Giannoni et
al., 2016;Jawhar
et al., 2012;Ri-
chard et al., 2015)
AD-BXD Strain-dependent amy-
loid plaques by
6 months. Strain-dependent
gliosis by
6 months.
Impaired in contex-
tual fear condition-
ing at 14 months.
Age of onset and
severity of impair-
ment are strain-de-
pendent.
(Hyman and Tan-
zi, 2019)
APP KM670/671NL
(Swedish); PSEN1
M146L (A>C) PS/APP accumulate in cortex
and hippocampus at
6 months.
No tangles; hy-
perphosphory-
lated tau
detected at
6 months, near
amyloid depos-
its in the cortex
and hippocam-
pus.
Neuronal loss in
hippocampal CA1
region at
22 months.
Numbers of as-
trocytes increase
in the cortex;
numbers of rest-
ing microglia in-
crease in the
vicinity of depos-
its at 6 months.
Reduced sponta-
neous alternation
performance in Y
maze at 3 months;
progressive cogni-
tive impairment as-
sessed by RAM.
(Arendash et al.,
2001;Gordon et
al., 2002;Hol-
comb et al., 1998;
Kurt et al., 2003;
Sadowski et al.,
2004)
APP KM670/671NL
(Swedish); PSEN1
M146V
ARTE10
Plaques by 3 months in
homozygotes and
5 months in hemizy-
gotes; start in anterior
neocortex and subicu-
lum.
No tangles; hy-
perphosphory-
lated tau in
dystrophic
neurites at
8 months.
Reduced dendritic
length, surface area,
and branches in
hippocampal neu-
rons at
10–14 months.
Reactive astro-
cytes and acti-
vated microglia
around plaques at
5 months in
homozygotes, la-
ter in hemizy-
gotes
Decreased ex-
pression of sy-
naptophysin
mRNA by
3–4 months
Cognitive deficits in
an object-recogni-
tion task and MWM
at 12 months.
(Willuweit et al.,
2009)
TASTPM
(TAS10×TPM)
deposit starts at
3 months; plaques evi-
dent by 6 months in
cortex and hippocam-
pus.
Absent.
Minimal neuronal
loss up to
10 months; some
signs of loss near
plaques in hippo-
campus.
Greater numbers
of reactive astro-
cytes and micro-
glia by 6 months
in hippocampus
and cortex.
Age-dependent im-
pairment in object
recognition memory
at 6 months.
(Howlett et al.,
2008;Howlett et
al., 2004)
APP KM670/671NL
(Swedish); PSEN1
L166P APPPS1
Amyloid plaque
deposition in neocortex
at 1.5 months;
hippocampus at
3–4 months; striatum,
thalamus and
brainstem at
4–5 months.
Phosphorylated
tau-positive
neuritic pro-
cesses around
plaques.
Neuron loss in DG
and other subre-
gions with high
neuronal density at
17 months.
Activated micro-
glia and increased
astrogliosis at
1.5 months.
Dendritic spine
loss at 2.5 month.
Hippocampal
CA1 LTP im-
paired at 8 and
15 months
Spatial learning and
memory deficits in
MWM at 7 months;
impaired reversal
learning in four-arm
spatial maze at
8 months.
(Gengler et al.,
2010;Radde et
al., 2006;Rupp et
al., 2011)
(To be continued on the next page)
20 Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP,
PSEN1
APP KM670/671NL
(Swedish); PSEN1
R278I
APP23×PS1-
R278I
Elevated Aβ43 by
3 months; amyloid de-
position by 6 months in
cortex and hippocam-
pus; high density of
cored plaques.
Absent.
Astrocytosis near
plaques in hippo-
campus and cor-
tex by 9 months.
Short-term memory
deficits in Y maze
by 3–4 months.
(Saito et al.,
2011)
APP KM670/671NL
(Swedish), V717I
(London); PSEN1
M233T, L235P
APP751SL/PS1
KI
deposition at
2.5 months; compact
deposits in
hippocampus, cortex
and thalamus at
6 months.
Neuronal loss de-
tected in female
mice at 6 months; in
hippocampus CA1/
2 at 10 months.
Astrogliosis in
proximity of Aβ-
positive neurons
at 2.5 months.
Reduced pre- and
post-synaptic
markers are at
6 months.
Reduced LTP and
disrupted paired
pulse facilitation
at 6 months.
Impaired working
memory by Y maze
and T-maze at 6 and
12 months.
(Breyhan et al.,
2009;Casas et al.,
2004)
APP V717I (London);
PSEN1 A246E
APP
(V717I)×PS1
(A246E)
Soluble AβOs at
2 months; plaques in
cortex, hippocampus
and subiculum at
4–6 months; CAA
pathology at 8 months.
Hyperpho-
sphorylated
murine tau; no
tangles.
Dystrophic neurites.
Microglial activa-
tion and other
markers of brain
inflammation in-
crease at
4.5 months.
Significant deficit
in LTP in hippo-
campal CA1 re-
gion at 6 months.
Spatial and
non-spatial
orientation and
memory deficits
by MWM at
5 months;
impaired
associative
learning.
(Dewachter et al.,
2000)
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H); APP
KM670/671NL
(Swedish), I716F
(Iberian); PSEN1
P117L
AppNL-F
Psen1P117L
double-mutant
Increased Aβ40, Aβ42
and Aβ43 in cortices at
3 months; dense cored
plaques at 12 months.
Significantly mi-
crogliosis and as-
trocytosis in
hippocampus at
12 months.
(Sato et al., 2021)
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H); APP
KM670/671NL
(Swedish); PSEN1
A246E
APPSwe/
PSEN1(A246E)
Amyloid plaques in
hippocampus and subi-
culum by 9 months, la-
ter extending to the
cortex.
Absent. Absent. Gliosis in cortex
and hippocampus
at 12 months.
Age-associated
cognitive impair-
ment in MWM at
11–12 months.
(Borchelt et al.,
1997;Borchelt et
al., 1996;Puoli-
väli et al., 2002;
Wang et al., 2003)
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H); APP
KM670/671NL
(Swedish); PSEN1
deltaE9
APPSwe/
PSEN1dE9
(C3-3×S-9)
Elevated Aβ42 and pla-
ques in hippocampus
and cortex at 6 months. Absent. Age-related cogni-
tive deficits at
18 months.
(Savonenko et al.,
2005)
APPswe/
PSEN1dE9
(C57BL6)
Plaques in cortex at
4 months and in
hippocampus at
6 months; amyloid
deposits on the
cerebral arteriole
wall at
7 months.
Absent of neuron
loss up to
12 months.
Plaque-associated
astrogliosis and
microgliosis by 4
and 8 months, re-
spectively.
Synapse loss in
the hippocampus
by 4 months. Deficits in MWM at
6–10 months and
worsen with age.
(Jackson et al.,
2013;Jankowsky
et al., 2001;Kim
and Jeong, 2015;
Malm et al.,
2007;Minkevi-
ciene et al., 2008)
(To be continued on the next page)
21
Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP,
PSEN1
Humanize mouse
region: G676R
(G5R), F681Y
(F10Y), R684H
(R13H); APP
KM670/671NL
(Swedish); PSEN1
deltaE9
APPswe/
PSEN1dE9
(line 85)
Occasional deposits
by 6 months; abundant
plaques in hippocampus
and cortex by 9 months;
progressive increase up
to 12 months.
Absent. Neuronal loss ob-
served adjacent to
plaques.
Minimal astrocy-
tosis at 3 months;
significant astro-
cytosis by
6 months; exten-
sive GFAP+
staining at
15 months.
Loss of
synaptophysin,
synaptotagmin,
PSD-95, and
homer immunor-
eactivity in
hippocampus by
4 months.
Reduced transient
LTP by 3 months;
not age related
from 3 to
12 months.
Deficits in spatial
memory in MWM
at 12 months.
(Garcia-Alloza et
al., 2006;Hong et
al., 2016;Jan-
kowsky et al.,
2004;Jankowsky
et al., 2001;
Kamphuis et al.,
2012;Lalonde et
al., 2005;Vo-
lianskis et al.,
2010)
CAST.APP/PS1
Plaques in cortex and
hippocampus by
8 months; more severe
in females.
Absent. Possible neuron loss
in hippocampal
CA1 area.
Plaque-associated
gliosis.
Impaired short-term
memory in Y-maze
in males, not avail-
able for females.
(Onos et al.,
2019)
PWK.APP/PS1 Plaques in the cortex
and hippocampal CA1
region by 8 months. Absent. Absent. Plaque-associated
microgliosis by
8 months. Absent assessed by
Y maze at
7–8 months.
(Onos et al.,
2019)
WSB.APP/PS1 Plaques in the cortex
and hippocampal CA1
region by 8 months. Absent. Fewer neurons in
cortex and CA1 of
female mice.
Plaque-associated
microgliosis by
8 months.
Deficits in short-
term memory by
8 months in female
mice.
(Onos et al.,
2019)
APP,
PSEN1,
TREM2
APP KM670/671NL
(Swedish), I716V
(Florida), V717I
(London); PSEN1
M146L (A>C),
L286V
TREM2-
BAC×5×FAD Plaques at 7 months. Microgliosis at
7 months. Absent (Lee et al., 2018)
Trem2 KO
(Colonna)×5×-
FAD Plaques at 4 months. Loss of cortical
layer V neurons at
8 months.
Microgliosis at
4 months. (Wang et al.,
2015)
TREM2, huma-
nized (common
variant)×5×-
FAD
Plaques at 8.5 months. Microgliosis at
8.5 months. (Song et al.,
2018)
APP KM670/671NL
(Swedish), I716V
(Florida), V717I
(London); PSEN1
M146L (A>C),
L286V; TREM2
R47H
TREM2, huma-
nized
(R47H)×5×FA-
D
Plaques at 8.5 months
Lower density of
activated micro-
glia surrounding
plaques at
8.5 months.
(Song et al.,
2018)
APP KM670/671NL
(Swedish); PSEN1
L166P
Trem2 KO
(KOM-
P)×APPPS1
Plaques observed by
2 months. Gliosis observed
by 2 months. (Jay et al., 2017)
APP KM670/671NL
(Swedish); PSEN1
L166P; TREM2 R47H
Trem2 R47H
KI (Lamb/
Landreth)×
APPPS1-21
Plaques observed at
4 months.
No tangles;
hyperpho-
sphorylated
tau detected at
4 months;
Dystrophic neurites
surrounding plaques
at 4 months (Cheng-Hathaway
et al., 2018)
(To be continued on the next page)
22 Li, X., et al. Sci China Life Sci
(Continued)
Gene Mutation site Model name Tau Neuronal
damage Gliosis Synaptic changes Changes in
LTP/LTD Cognitive dysfunc-
tion References
APP,
PSEN2
APP KM670/671NL
(Swedish); PSEN2
N141I
PS2APP
deposition at
6 months; heavy plaque
load in hippocampus,
frontal cortex, and sub-
iculum at 10 months.
Absent. Gliosis at
6 months.
A strong increase
in LTP in hippo-
campal slices at
10 months.
Cognitive impair-
ment in MWM at 8
and 12 months.
(Ozmen et al.,
2009;Poirier et
al., 2010;Wei-
densteiner et al.,
2009)
PS2APP (PS2
(N141I)×APPs-
we)
Rare amyloid deposits
at 5 months; consistent
deposits in subiculum
and frontolateral cor-
tices by 9 months; pla-
ques spread throughout
neocortex, hippocam-
pus, amygdala, thalamic
and pontine nuclei at
17 months.
Absent. Activated micro-
glia and astro-
cytes at 9 months. Absent at 3 and
10 months.
Impaired acquisi-
tion of spatial
learning in MWM
from 8 months.
(Richards et al.,
2003)
APP,
RAGE
APP KM670/671NL
(Swedish), V717F
(Indiana)
Tg-mAPP/
RAGE
Increased activa-
tion of microglia
and astrocytes at
14–18 months.
Decreased LTP at
8–10 months. Impaired spatial
learning and mem-
ory at 3–4 months.
(Arancio et al.,
2004)
MAPT,
TREM2
MAPT P301S
PS19 with hu-
manized
TREM2 (com-
mon variant)
Tangles at
9 months.
Atrophy of hippo-
campus and entorh-
inal/piriform cortex
and pronounced
ventricular expan-
sion at 9 months;
thinner DG granule
cell layer and piri-
form cortex pyra-
midal cell layer
compared to those
carrying R47H var-
iant of TREM2.
Elevated expres-
sion of markers of
astroglial and mi-
croglial reactivity,
compared with
those carrying the
R47H variant of
TREM2 at
9 months.
Fewer synapses
and more dys-
trophic synapses
compared with
those carrying the
R47H variant of
TREM2 at
9 months.
(Gratuze et al.,
2020)
MAPT P301S;
TREM2 R47H
PS19 with hu-
manized
TREM2
(R47H)
Tangles at
9 months (Gratuze et al.,
2020)
(To be continued on the next page)
a) The table is ordered by gene and mutation site alphabetically. “—” means no data.
23
Li, X., et al. Sci China Life Sci
bloch et al., 2007a;Knobloch et al., 2007b;Kurt et al., 2003;
Lalonde et al., 2005;Lamb et al., 1999;Lamb et al., 1997;
Lee et al., 2018;Lewis et al., 2001;Lillehaug et al., 2014;
Liu et al., 2015;Liu et al., 2021b;Lord et al., 2009;Lord et
al., 2006;Malm et al., 2007;Mehla et al., 2019;Miao et al.,
2005b;Minkeviciene et al., 2008;Mucke et al., 2000;Mül-
ler-Schiffmann et al., 2016;Nilsson et al., 2014;Oakley et
al., 2006;Oddo et al., 2003;Onos et al., 2019;Ozmen et al.,
2009;Platt et al., 2011;Poirier et al., 2010;Puoliväli et al.,
2002;Radde et al., 2006;Reaume et al., 1996;Richard et al.,
2015;Richards et al., 2003;Rockenstein et al., 2001;
Rockenstein et al., 2005;Rupp et al., 2011;Sadowski et al.,
2004;Sagare et al., 2013;Saito et al., 2014;Saito et al., 2019;
Saito et al., 2011;Sato et al., 2021;Savonenko et al., 2005;
Savonenko et al., 2003;Skaaraas et al., 2021;Song et al.,
2018;Stover et al., 2015;Volianskis et al., 2010;Wang et al.,
2003;Wang et al., 2015;Watamura et al., 2022b;Weiden-
steiner et al., 2009;Wilcock et al., 2008;Willuweit et al.,
2009;Wittnam et al., 2012;Wright et al., 2013;Xia et al.,
2021;Xiao et al., 2015;Xu et al., 2007;Yamada et al., 2009;
Youmans et al., 2012;Zampar and Wirths, 2021). These
models show different timelines and severity of AD-related
phenotypes. The mouse models carrying multiple mutants of
APP showed deposits and cognitive deficits, but absent
of NFTs and neuronal death. Mouse models carrying a
combination of APP and other gene mutations (such as
PSEN1,PSEN2,MAPT) exhibited earlier and more ag-
gressive pathological and behavioral changes than those only
carrying APP mutants. In addition, some multi-transgenic
mouse models also introduced risk genes such as APOE,
TREM2,ABCA7 and SORL1. To study the role of the three
human isoforms of APOE on AD phenotypes, APOE4,
APOE3, and APOE2 targeted replacement mice were crossed
with 5×FAD mice, respectively. Among them, E4FAD
mouse model carrying the APOE ε4 subtype had more pla-
ques and worse cognitive impairment than E3FAD and
E2FAD mice carrying APOE ε3 and APOE ε2 subtypes,
respectively (Youmans et al., 2012). TREM2 R47H was re-
ported as a significant risk modifier for late-onset AD, and
was constructed into the 5×FAD, APP/PS1-21 and PS19
mouse models. These mice carrying the TREM2 R47H mu-
tation had less pathology than those carrying wild type or
common variant of human TREM2 (Cheng-Hathaway et al.,
2018;Gratuze et al., 2020;Song et al., 2018). Other AD risk
genes mouse models such as Trem2 R47H KI×APOE4,
Abca7*A1527G/APOE4/Trem2*R47H, Sorl1*A528T/
APOE4/Trem2*R47H were not listed in the table due to lack
of pathological and cognitive evidence.
Figure 2 The ethnic origin of AD pathogenic gene mutation sites used in all single-transgenic AD mouse models. A, Number of single mutation sites found
in FAD in each continent. B, Overlaps of single mutation sites used in single-transgenic AD mouse models among continents.
24 Li, X., et al. Sci China Life Sci
Tg-SwDI mice
Tg-SwDI mice were constructed by random transgenic
technology and overexpressing the human APP gene (iso-
form 770) containing the K670N/M671L (Swedish), E693Q
(Dutch), and D694N (Iowa) mutations under the control of
the mouse Thy1 promoter (Davis et al., 2004). At 3 months
of age, insoluble deposits were found in the subcortex,
hippocampus and cortex of Tg-SwDI mice. By 6 months,
plaques-like deposits became more numerous and appeared
in the olfactory bulb and thalamus, and fibrillary accu-
mulation increased in the cerebral microvasculature. At
12 months, deposits were detected throughout the fore-
brain (Davis et al., 2004). The mice developed gliosis with
age (6–24 months) especially in the thalamus and sub-
iculum, and to a lesser extent in the cortex. Learning and
memory deficits were observed in the Barnes maze task at
3 months. This model serves as a cerebral microvascular
amyloid model for AD drug research, such as cGMP-grade
AV-1959D vaccine (Petrushina et al., 2020), taxifolin (Saito
et al., 2017), and minocycline (Fan et al., 2007).
J20 mice
J20 mice were constructed by random transgenic technology
and overexpressing the human APP gene containing the
K670N/M671L (Swedish) and V717F (Indiana) mutations
driven by the PDGF promoter (Mucke et al., 2000). The
expression of human APP in the brain of J20 mice was
moderate, but the accumulation of was the highest among
all PDGF-APP mice. Increased deposits in the hippo-
campus began at 1 month, and plaque formation started at
5–7 months (Mucke et al., 2000). hippocampal neuronal loss,
synapse loss and cognitive impairment were observed at
3 months (Cheng et al., 2007;Hong et al., 2016;Wright et
al., 2013). J20 mice also showed deficits in basal synaptic
transmission and LTP at the Schaffer collateral-CA1 synapse
at 3–6 months (Saganich et al., 2006), and developed mi-
crogliosis and astrogliosis in the hippocampus at 6–9 months
(Wright et al., 2013). In J20 mice, tumor necrosis factor
receptor 2 stimulation, adult neurogenesis modulation and
mTOR attenuation were found to be potential treatments for
AD (Ortí-Casañ et al., 2022;Van Skike et al., 2021;Zhang et
al., 2021).
TgCRND8 mice
TgCRND8 mice were constructed by random transgenic
technology and overexpressing the human APP gene (iso-
form 695) containing the K670N/M671L (Swedish) and
V717F (Indiana) mutations under the control of the hamster
prion gene promoter (Chishti et al., 2001). This model is an
early-onset AD model with deposition but no tangles.
The mice showed plaques, plaque-associated microglia
and astrocyte activation, and memory impairment at
3 months (Chishti et al., 2001;Dudal et al., 2004). Neuronal
loss, reduced LTP and decreased synaptophysin around the
plaques were seen in the hippocampus at 6 months (Adalbert
et al., 2009;Brautigam et al., 2012;Kimura et al., 2012). Tau
hyperphosphorylation and aggregation occurred at
7–12 months, particularly in neocortex and hippocampus
(Bellucci et al., 2007). Severe vascular deposition was
observed at 11 months (Domnitz et al., 2005). Nano-hono-
kiol, magnolol, and isorhynchophylline could ameliorate
neuropathology and cognitive deficits in TgCRND8 mice (Li
et al., 2019;Qu et al., 2022;Xian et al., 2020).
APPNL-G-F KI mice
APPNL-G-F KI mice were constructed by knocking in tech-
nology and humanizing mouse sequence and introducing
K670N/M671L (Swedish), E693G (Arctic), V716F (Beyr-
euther/Iberian) mutations into exon 16 and 17 of mouse APP
gene (Nilsson et al., 2014). In homozygous mice, elevated
levels of Aβ42 in the cortex started at 2 months, and plaques
appeared in the cortex, hippocampus and other subcortical
regions at 4 months (Saito et al., 2014). Gliosis, degenerative
neurons, decreased dendritic spines, reduced synaptophysin
and PSD95 associated with plaques, changes in the sy-
naptic microstructure, and impaired spatial learning and
memory in the MWM and Y maze were observed at
6 months (Liu et al., 2021b;Mehla et al., 2019;Nilsson et al.,
2014;Saito et al., 2014). Memory impairment in NOR trials
was observed at 9 months (Mehla et al., 2019;Saito et al.,
2014). Tau pathologies were not found in APPNL-G-F KI mice.
The mice also showed neuroinflammation and dysregulation
of β-catenin and NF-κB signaling pathways at 6 months (Liu
et al., 2021b). Using this model, studies demonstrated that
Cathepsin E could modulate microglial activation and neu-
rodegeneration, and inhibit pathology and neuroin-
flammation (Xie et al., 2022); two non-steroidal anti-
inflammatory drugs (dexketoprofen and etodolac) and two
anti-hypertensives (penbutolol and bendroflumethiazide)
could reduce plaque burden and reverse cognitive im-
pairment (Pauls et al., 2021).
3×Tg-AD mice
3×Tg-AD mice were constructed by microinjection of two
human transgenes (APP KM670/671NL mutation and MAPT
P30IL mutation, under the control of mouse Thy1.2 pro-
moter) into single-cell embryos from homozygous PS1
(M146V) KI mice (Oddo et al., 2003). The mice showed
intraneuronal deposits in the hippocampus, cortex and
amygdala, retention/retrieval deficit at 4 months (Billings et
al., 2005). The mice exhibited extracellular deposits in
25
Li, X., et al. Sci China Life Sci
the frontal cortex, decreased LTP, impaired basal synaptic
transmission, and learning and memory deficits in the Barnes
maze at 6 months (Billings et al., 2005;Oddo et al., 2003;
Stover et al., 2015). Gliosis appeared at 7 months of age and
increased with age (Caruso et al., 2013). Tau phosphoryla-
tion and NFTs appeared in the hippocampal CA1 neurons at
12 months, and progressed to the cortex (Oddo et al., 2003).
The mice had sexual dimorphism, as reflected by increased
cognitive deficits in females and increased levels of novelty-
induced behavioral inhibition in males (Blázquez et al.,
2014). Plasma BDNF levels in mice could predict synaptic
plasticity and memory deficits (Cocco et al., 2020). This
mouse model is usually used to study AD treatment. For
example, overexpression of phospholipid transfer protein
decreased senile plaques and NFTs, thereby improving
cognitive impairment in the mice (Wang et al., 2021c). Pa-
clitaxel delivered via the intranasal route could increase
axonal transport rates, reduce phospho-tau-containing neu-
rons, and improve performance in behavioral testing in
3×Tg-AD mice (Cross et al., 2021). Shortening theryanodine
receptor 2 open time with R-carvedilol rescued these AD-
related deficits in 3×Tg mice (Liu et al., 2021c).
5×FAD mice
5×FAD mice were constructed by co-injecting two vectors
encoding APP with KM670/671NL (Sweden), I716V
(Florida), and V717I (London) mutations and PSEN1 with
M146L and L286V mutations, each driven by the mouse
Thy1 promoter (Oakley et al., 2006). The 5×FAD model was
initially constructed with three lines, including Tg6799,
Tg7092 and Tg7031. Among them Tg6799 is the fast line for
behavioral and amyloid pathological changes, Tg7031 is the
slow line, and Tg7092 is in the middle. Tg6799 is the most
widely used line, so in this review we mainly described the
line Tg6799 (Oakley et al., 2006). The 5×FAD (B6SJL) mice
are the original 5×FAD line, on a hybrid B6SJL background.
The 5×FAD (B6SJL) mice showed intraneuronal Aβ42 ac-
cumulation at 1.5 months, extracellular amyloid deposition
and gliosis at 2 months, that increased with age, and plaques
throughout the hippocampus and cortex at 6 months (Oakley
et al., 2006). Deficits in basal synaptic transmission and LTP
of the hippocampus were observed between 4 to 6 months
(Crouzin et al., 2013;Kimura and Ohno, 2009). Synaptic
degeneration including presynaptic marker synaptophysin,
syntaxin and postsynaptic marker PSD-95 decline was ob-
served at 4 and 9 months, respectively, and neuronal loss in
cortical layer 5 and subiculum was observed at about
6 months, which progressed to other regions (Oakley et al.,
2006). Impaired spatial working memory in the MWM and
Y-maze test and impaired remote memory stabilization in a
contextual-fear-conditioning test were observed by 4 to
6 months, while impaired learning and social recognition
were observed at 9–12 months (Flanigan et al., 2014;Kimura
and Ohno, 2009;Oakley et al., 2006;Xiao et al., 2015). The
5×FAD (B6SJL)mice also showed other behavioral changes,
such as olfactory, motor, and hearing impairment at 6, 9, and
14–16 months, respectively (Flanigan et al., 2014;O’Leary
et al., 2018;O’Leary et al., 2017;Xiao et al., 2015). The
5×FAD (C57BL6) is obtained by 5×FAD (B6SJL) back-
crossing C57BL6 mice for several generations. The earliest
pathological features of 5×FAD (C57BL6) mice are in-
traneuronal accumulation and synaptic deficits in cortical
layer V neurons at 1.5–2 months (Buskila et al., 2013;Ri-
chard et al., 2015). Extracellular plaques were observed in
hippocampus, cortex, and thalamus of 2-month animals
(Richard et al., 2015). Progressive CAA on leptomeningeal
and penetrating vessels started at 3 months (Giannoni et al.,
2016). Impairment of spatial working memory in
3–6 months old 5×FAD (C57BL6) mice is later than the
5×FAD (B6SJL) mice (Jawhar et al., 2012). 40% loss of
layer V pyramidal neurons was detected in 12 months old
5×FAD (C57BL6) mice, but the total number of neurons in
hippocampus and cortex was not different from that in
wildtype mice (Jawhar et al., 2012). The 5×FAD (C57BL6)
mice also showed other behavioral changes, such as reduced
anxiety, abnormal reflexes, sensorimotor deficits, at 3, 5, and
9 months, respectively (Jawhar et al., 2012;Richard et al.,
2015). 5×FAD mouse model is widely used in AD drug
development (Keren-Shaul et al., 2017;Wang et al., 2019;
Xiao et al., 2015). For example, GV-971, approved by the
Chinese Food and Drug Administration for the treatment of
AD, was reported to harness neuroinflammation and reverse
cognition impairment in this model (Wang et al., 2019).
Salidroside protected neurite morphology, mitigated pa-
thology, and improved cognitive function in this model (Yao
et al., 2022). Exenatide reduced Aβ1-42 deposition in the
hippocampal CA1 region, alleviated synaptic degradation in
the hippocampus, and improved the learning ability and
spatial memory ability in this model (An et al., 2019). Many
drugs targeting AβO had shown therapeutic efficacy in this
model, which were described in detail in the next section.
APPPS1 mice
APPPS1 (APPPS1-21) mouse model was constructed by co-
injecting two vectors that encode mutant APP KM670/
671NL(Sweden) and PSEN1 L166P, respectively. de-
position began in the cortex at 6 weeks of age, and activated
microglia around deposits as well as increased astro-
gliosis, deposition in the hippocampus at 3–4 months of
age (Radde et al., 2006). Synaptic loss began at 4 weeks after
plaque formation and continued for several months (Bittner
et al., 2012). LTP was impaired in hippocampal CA1 region
at 8 and 15 months of age (Gengler et al., 2010). Modest
neuron loss was found in the granule cell layer of the DG and
26 Li, X., et al. Sci China Life Sci
other subregions with high neuronal density in 17-month-old
mice (Rupp et al., 2011). Spatial learning and memory were
impaired at 7 months (Serneels et al., 2009), and reversal
learning was impaired at 8 months of age (Radde et al.,
2006). This mouse model is commonly used in AD treatment
studies. Specifically, 7-day treatment of preweaned male
mice with antibiotic (ABX) is associated with reductions of
amyloidosis, plaque-localized microglia morphologies,
and Aβ-associated degenerative changes at 9 weeks of age.
Furthermore, fecal microbiota transplantation into ABX-
treated male mice completely restored the above pathologi-
cal changes (Dodiya et al., 2020;Dodiya et al., 2022).
toxicity inhibitor SEN1500 showed a positive effect on
cognition in APPPS1 mice (Lo et al., 2013). Another study
showed that prolonged treatment with the pan-histone dea-
cetylase inhibitor sodium butyrate improved associative
memory in APPPS1 mice (Govindarajan et al., 2011). A
recent study used a genetic modification strategy by injecting
virus into the median raphe nucleus of APPPS1 mice, and
found that ETV4 activation of VGLUT3 transcription could
upscale the efficacy of spatial memory retrieval, which
provided a promising therapeutic target for AD (He et al.,
2022).
APPswe/PSEN1dE9 mice
APPswe/PSEN1dE9 mice were constructed by co-injecting
two vectors that encode mutant APP and PSEN1, respec-
tively. The APP sequence contains three humanized sites
and KM670/671NL (Sweden) mutation, and the PSEN1
sequence encodes human presenilin-1 lacking exon 9
(Jankowsky et al., 2001). The mice first showed plaques,
gliosis and synapse loss at 4 months (Jackson et al., 2013;
Jankowsky et al., 2001;Malm et al., 2007;Minkeviciene et
al., 2008). Plaques emerged in cortex at 4 months, pro-
gressed to the hippocampus at 6 months, and increased in
size and number with age (Jackson et al., 2013;Minkevi-
ciene et al., 2008). There was amyloid deposition on the
cerebral arteriole wall at 7 months (Kim and Jeong, 2015).
Cognitive impairment measured by MWM was observed
between 6 and 10 months and worsened with age. Phos-
phorylated tau was observed in hippocampal CA3 neurons
at 16 months (Malm et al., 2007;Minkeviciene et al., 2008).
This mouse model is also commonly used in AD drug stu-
dies. For example, GV-971 was reported to harness neu-
roinflammation and reverse the cognitive impairment in this
model (Wang et al., 2019). Chronic oral masitinib treatment
promoted a recovery of synaptic markers, and restored
normal spatial learning performance in this model (Li et al.,
2020).
The phenotypic characteristics and applications of com-
monly used AD transgenic mouse models are described be-
low (Table 6).
AβO pathology related transgenic mouse models
and clinical translation
The earliest mouse model to report accumulation of AβO in
the brain is the Tg2576 mice, which carry 2 mutations (hu-
man APP695 containing K670N, M671L) (Chang et al.,
2003). It started to show Aβ42 oligomers in the cortex at
11 months (Takahashi et al., 2004), and gradually spread to
other regions especially hippocampus and striatum at
13 months, and reached the highest level in hippocampus
and cortex at 17 months (Chang et al., 2003). The number
and size of Aβ42 oligomer deposits increased after
20 months (Takahashi et al., 2004). Subsequently, Tg-SwDI
mice with four mutations (human APP770 containing
K670N/M671L, E693Q, D694N) (Miao et al., 2005a),
APP23 with two mutations (human APP751 containing
K670N, M671L) (Lefterov et al., 2009), J20 (Murata et al.,
2010) and McGill-Thyl-APP Tg mice (Ferretti et al., 2012;
Ferretti et al., 2011) with three mutations (human APP751
containing K670N, M671L, V717F) were reported to show
age-dependent AβO accumulation in brain neurons prior to
plaque appearance. While APP E693Δ-Tg (human APP695
containing E693Δ) (Tomiyama et al., 2010;Umeda et al.,
2011), APP Dutch (human APP751 containing E693Q)
(Gandy et al., 2010;Price et al., 2014), OSK-KI (APP
E693Δ) (Umeda et al., 2017), and PS1V97L Tg mice (Zhang
et al., 2014) that carry single mutations were reported to
show age-related AβO accumulation in cortical and hippo-
campal neurons, no extracellular deposition of any forms
was found.
The model carrying multiple gene mutations of APP,
PSEN1,PSEN2,MAPT or BACE1 also showed pathological
oligomers accumulation. In 3×Tg mice brain, AβOs in-
creased at 6 months, decreased briefly at 9–12 months, and
increased again with age at 12–20 months (Oddo et al.,
2006). AβOs appeared in the brains of 5×FAD (Tg6799)
mice at 6 months of age, and BACE1 gene deletion resulted
in the reduction of AβOs in the mice brains (Ohno et al.,
2006). Age-dependent accumulation of AβO started at
7 months in the brain of APP/PS1 mice (Bruggink et al.,
2013). PS2APP mice lacking Trem2 showed increased AβOs
in hippocampus, in addition to more diffused plaques in mice
brain (Meilandt et al., 2020).
Drug treatment is needed to prevent, delay the onset, slow
the progression, and improve the symptoms of AD (Cum-
mings et al., 2022). The large body of evidence that AβOs are
both necessary and sufficient to trigger AD-associated
memory malfunction and neurodegeneration, coupled with
the extensive literature of AβO-triggered cellular and beha-
vioral effects, sets the stage for new AD therapeutic ap-
proaches targeting AβO (Bayer, 2022;Selkoe and Hardy,
2016;Sun et al., 2021). Several drugs targeting AβO have
been tested in AβO-induced transgenic mouse models, with
27
Li, X., et al. Sci China Life Sci
Table 6 The main characters and applications of each line
Model name Main characters Applications
PS1(I213T) KI Elevated Aβ42 and Aβ42/40 ratio, phosphorylated tau started at young age,
tangles and spatial learning deficits appeared in middle age. No data
PS1(P117L) Impaired neurogenesis as early as 1 month; plaque and tau phosphorylation
were seen at young age. Mixed mouse models with
other genes
PS1V97L Tg AβO accumulation, glial cell activation, decreased LTP, phosphorylated tau, NFTs,
spatial learning and memory impairment occurred in young age; neuronal damage
was occasionally seen in middle age; neurodegeneration occurred in old age. Sulforaphane, honokiol
APPE693Δ-Tg AβO accumulation, hyperphosphorylated tau, decreased synaptic density started at
young age; microgliosis was observed in middle age; astrocytosis and neuron loss
occurred in old age.
Mixed mouse models with
other genes
APP23 Spatial memory impairment as early as 3 months; plaque, CAA, glial cell
activation and hyperphosphorylated tau appeared in young adult mice;
neuron loss occurred in old age.
Anti-Aβ antibodies and other drugs that
block the formation of multimers
TAS10 (Thy1-APPswe) Spatial learning and memory decline, and dystrophic neurites, lipid deposits, gliosis
started at young age; plaques and working memory deficits appeared in middle age;
synapses loss occurred in old age. No data
Tg2576 Reduced LTP, spatial and non-spatial orientation and memory deficits appeared
in young age; increased plaques, hyperphosphorylated tau and extensive CAA
were seen in old age.
Aducanumab, bexarotene, memantine,
oat extract avenanthramide-C
PDAPP (line109) Learning, working memory, recognition memory and synaptic dysfunction appeared
at <6 months, followed by plaques deformed extensively with age and glial cell
activation; phosphorylated tau and CAA were detectable in old age. 2B3, semagacestat
APP(V717I) Reduced LTP and impairment of learning and memory started at young age;
increased accumulation (including plaques and CAA) and inflammation occurred
in middle age. Mixed mouse models with other genes
JNPL3(P301L) NFTs appeared in young age, neuronal loss and astrogliosis occurred in middle age;
motor and behavioral problems by 10 months. Anti-tau antibodies
rTg(tauP301L)4510 Gliosis, tau pathology, neuron loss, synaptic dysfunction, brain weight decreased
and cognitive impairment appeared in young adults; forebrain atrophy was
seen in middle age.
Doxycycline, trazodone, epicatechin,
LDN/OSU-0215111
Tau P301S (Line PS19)
Tau hyperphosphorylation, impaired axonal transport, microgliosis, decreased
synaptophysin and motor deficits as early as 3 months; followed by increased
inflammatory factors and synaptic dysfunction in CA1, progressive NFTs and
gliosis in multiple brain regions and spinal cord; working memory, impaired
spatial learning and memory, contextual memory, sociability, and object
recognition; hippocampal atrophy and cortical atrophy occurred in middle age;
80% percent of mice died by 12 months.
Metformin, tryptophan-tyrosine-related
β-lactopeptides, ibrutinib, combined
with AD risk factors
Tg-SwDI Insoluble accumulation and cognitive deficits began at 3 months;
plaques-like deposits, CAA and gliosis increased with age at 6 months. cGMP-grade AV-1959D vaccine,
taxifolin, minocycline
J20 Increased deposits in the hippocampus began at 1 months; synaptic
dysfunction and gliosis occurred in young age.
Tumor necrosis factor receptor
2 stimulation, adult neurogenesis
modulation, mTOR attenuation
TgCRND8 Plaques, glial cell activation and cognitive impairment began at 3 months; followed
by neuronal loss, reduced LTP and decreased synaptophysin around the plaques in
young age; hyperphosphorylated tau aggregation and CAA occurred in middle age.
Nano-Honokiol, magnolol,
isorhynchophylline
APPNL-G-F KI Elevated level of Aβ42 in the cortex started at 2 months; gliosis, neurodegeneration,
synaptic dysfunction and cognitive impairment appeared in young age. Cathepsin E, dexketoprofen , etodolac,
penbutolol , bendroflumethiazide
3×Tg-AD deposits, synaptic dysfunction, gliosis, decreased LTP and cognitive impairment
occurred in young age; hyperphosphorylated tau and NFTs were seen in middle age. Paclitaxel, R-carvedilol,
butylphthalide
5×FAD (B6SJL)
Intraneuronal Aβ42 accumulation at 1.5 months; followed by extracellular amyloid
deposition and gliosis increased with age; plaques, neuron loss, synaptic dysfunction,
spatial working memory, remote memory deficits at 6 months; impaired learning,
social recognition, motor and hearing were observed in middle age. GV-971, salidroside, exenatide, 9D5,
coassembly composed of
cyclodextrin and calixarene
5×FAD (C57BL6)
Intraneuronal Aβ42 accumulation and synaptic deficits in cortical layer V neurons at
1.5-2 months; followed by extracellular amyloid deposition, CAA and gliosis
increased with age; spatial working memory deficits in young age; layer V neurons
loss in middle age.
APPPS1 Plaques, activated microglia, astrogliosis, dendritic spine loss appeared at
<3 months; impaired LTP and cognitive deficits, modest neuron loss was
found in old age.
ABX, fecal microbiota transplantation,
SEN1500, sodium butyrate, tenuifolin,
ginsenoside, cryptotanshinone, carnosic
acid, butylphthalide, lycium barbarum
extract, genetic modification strategy by
virus injection
APPswe/PSEN1dE9 Plaques, gliosis and synapse loss began at 4 months; followed by CAA and cognitive
impairment; phosphorylated tau was observed in middle age. GV-971, masitinib
28 Li, X., et al. Sci China Life Sci
strategies including directly clearing AβOs, blocking AβO
surface receptors, interfering with AβO-induced signaling
pathways, or decreasing downstream effectors such as tau
(Overk and Masliah, 2014). Selective AβO antibodies pre-
vent AD-like pathology and rescue memory performance
(Klyubin et al., 2005;Lambert et al., 2007;Sebollela et al.,
2017). Several drugs have shown therapeutic effects, such as
aducanumab that binds to both soluble AβOs and insoluble
fibrils in Tg2576 mice (Gamage and Kumar, 2017). The
murine version of humanized antibody BAN2401 (mAb158)
exhibited a strong binding preference for soluble AβOs and
protofibrils over monomers in tg-ArcSwe mice (Tucker
et al., 2015). Gantenerumab and crenezumab were also found
binding to AβOs in the PS2APP mouse model (Barrow et al.,
2017;Meilandt et al., 2019). Passive immunization of
5×FAD mice with the monoclonal antibody 9D5, which re-
acted with low molecular weight AβOs, reduced plaques and
rescued behavioral deficits (Wirths et al., 2010). The
monoclonal antibody 07/01 prevented in vitro toxicity of
AβOs and normalized in vivo behavioral deficits in AD
mouse model (Frost et al., 2015). A coassembly composed of
cyclodextrin (CD) and calixarene (CA) was able to eliminate
amyloid plaques, and degrade Aβ1-42 monomers and oli-
gomers in 5×FAD mice (Wang et al., 2021a).
In addition to the above antibody drugs, a variety of drugs
especially traditional Chinese medicine monomers were ef-
fective in AβO based mouse models (Chen et al., 2020), and
had multi-target anti-dementia components (Jia et al., 2020c;
Li et al., 2021a). More recently, sulforaphane was found to
improve spatial cognition in PS1V97L Tg mice and improve
memory impairment and depressive-like behavior in rats
with intracerebroventricular injection AβOs, with potential
mechanism of anti-AβOs, anti-tau, anti-oxidative stress and
anti-neuroinflammation, and protecting serotonergic system
(Hou et al., 2018;Wang et al., 2020). Honokiol was found
effective in PS1V97L Tg mice, with a potential mechanism
of up-regulating peroxisome proliferator-activated receptor γ
and peroxisome proliferator-activated receptor-γ coactivator-
through SIRT3 pathway, thus improving mitochondrial
function and apoptosis, and promoting microglial phagocytic
function (Li et al., 2018;Li et al., 2021b). Tenuifolin (Zhang
et al., 2008), ginsenoside (Li et al., 2016), cryptotanshinone
(Mei et al., 2009) and carnosic acid (Yi-Bin et al., 2022) were
found effective in APP/PS1 mice, with potential mechanisms
of anti-neuroinflammation and anti-oxidative stress through
Nrf2/NF-κB and CEBPβ-NFκB signaling pathways, redu-
cing tau phosphorylation, synaptoprotection and mitopro-
tection via PI3K/Akt/GSK3β pathway and regulating
NMDAR2B downstream cascades (Chen and Jia, 2020;
Chen et al., 2022;Jiao and Jia, 2022;Liu et al., 2023;Lyu
and Jia, 2022). In addition, butylphthalide and lycium bar-
barum extract were found effective in 3×Tg and/or APP/PS1
mice, with potential mechanisms of reducing tau phosphor-
ylation, reducing AβO-induced inflammatory reactions, sy-
napse stabilization, anti-oxidation and anti-apoptosis through
CaMKα pathway (Li et al., 2022;Liu et al., 2021a;Peng et
al., 2010;Sun et al., 2022;Zhang et al., 2016).
Among the drugs that showed therapeutic efficacy in AβO
based mouse models, several drugs have shown efficacy in
clinical trials or have biomarker data, including two anti-
bodies that engage AβOs, aducanumab and lecanemab (Se-
vigny et al., 2016;Swanson et al., 2021;van Dyck et al.,
2023). Butylphthalide, in combination with donepezil, was
found effective in delaying cognitive decline in patients with
mild to moderate AD (Wang et al., 2021b). Sulforaphane has
entered clinical trials for AD population (NCT04213391).
Advantages and limitations of transgenic mouse
models
Transgenic mouse models are useful tools for studying early
AD pathology as well as the spatial and temporal features of
each pathology, which would be helpful for developing early
diagnostic markers for AD even before symptoms appear
(behavioral changes in mice), namely Aβ, tau, neuronal da-
mage, microglia activation, and synaptic changes as shown
in the tables. The mouse models are also useful in in-
vestigating the upstream and downstream mechanisms and
molecular pathways of each mechanism, which would be
helpful for developing target treatments. Having that said,
the heterogeneity and variation of each transgenic mouse
model in terms of phenotypes make it inconclusive about AD
pathogenesis, which requires further verification in higher
order animals (namely non-human primates) and/or clinical
studies.
Single transgenic mouse models
Single-transgenic mouse models have the following ad-
vantages and limitations. First, the behavioral changes ap-
pear relatively late, not as early as multi-transgenic models
such as 5×FAD model, thus the duration for drug experi-
ments might be longer, which is not efficient for drug
screening. However, it is more consistent with real world
situation and thus is more suitable for translational research
of promising drugs with known targets of action that match
the pathogenic features of the mouse models. There is evi-
dence showing that the chance of translation of findings in
transgenic mouse models to humans decreases as the focus
shifts from the targets of interest to behavioral or other
biomarker end points (Scearce-Levie et al., 2020). Second,
many single-transgenic mouse models only mimic a few
pathogenic characters of AD, thus cannot reflect the features
of the whole AD population. In this regard, models with
more pathogenic features such as PS1V97L Tg mice might
29
Li, X., et al. Sci China Life Sci
be a good choice. Third, many single-transgenic mouse
models are developed locally, and not widely used or fully
studied for their pathogenic features or mechanisms, and not
verified by other labs. In the future, more efforts are needed
for cross-validation.
Multi-transgenic mouse models
Multi-transgenic mouse models have the following ad-
vantages and limitations. It was found that models expressing
multiple FAD gene mutations could lead to earlier and more
severe pathology (Drummond and Wisniewski, 2017). In-
creased number of APP mutations or superimposed with
PSEN1 or PSEN2 mutations can lead to earlier AD-related
pathology (Irizarry et al., 1997;Jankowsky et al., 2004;
Mehla et al., 2019;Nilsson et al., 2014;Oakley et al., 2006;
Richards et al., 2003;Sato et al., 2021), and shorten the
experimental period. The 3×Tg mice constructed in combi-
nation with APP,PSEN1, and MAPT gene mutations can
simulate plaque and NFTs, the two typical AD pathological
manifestations (Oddo et al., 2003), but it is debatable whether
this combination can represent AD (King, 2018). In addition,
crossbreeding 5×FAD mice with risk gene mice such as
BACE,APOE, and TREM2, can illustrate the interaction of
these risk genes with pathogenic gene mutations during the
onset and development of AD (Hu et al., 2018;Song et al.,
2018;Youmans et al., 2012). Although studies in these mouse
models have made important contributions in explaining the
pathologic changes of AD, drug efficacy provided by these
models has seldom been translated into clinical practice
(Josephine Boder and Banerjee, 2021;Zhang et al., 2020).
First, the genetic background of such models is rarely seen in
human genetic characteristics (Martini et al., 2018). FAD
patients usually carry only one pathogenic variant, and more
than 80% of FAD patients are not found to carry the reported
pathogenic variant (Jia et al., 2020b). Whether mechanisms
and potentially effective drugs obtained from multi-trans-
genic animals can be completely translated into AD patients
with a single mutation or without these mutations still re-
quires more validation and interpretation (King, 2018;
Mckean et al., 2021). Second, the additive effect of multiple
gene mutations in the multi-transgenic model has shortened
the process of AD, that are not in line with the chronic pro-
gression feature especially in preclinical stage of AD before
symptoms onset. As a result, it is difficult to study the
treatment strategy in terms of timing and duration. Third, the
phenotype changes are likely to be faster and more severe
than in single-transgenic mouse models, thus it is difficult for
prevention study as well as risk factor managements. Pre-
vention trials take the longest duration, even longer than
disease-modifying therapy (Cummings et al., 2022). Results
of positive effects in young models cannot be verified in
elderly AD patients (King, 2018).
Tg mice vs. KI mice
Tg is to integrate foreign genes into mouse chromosomes at
random to obtain randomly inserted transgenic mice (Jae-
nisch and Mintz, 1974;Oakley et al., 2006). Because the
target gene is randomly integrated into the genome, the ex-
pression of the target gene is affected by the integrated lo-
cation, and the expression may be high or low, or even not
expressed (Garrick et al., 1998). This feature has two sides.
The advantage is that researchers can select the mice with the
best expression level from mice with various expression le-
vels for scientific research. However, different expression
strains need to be established in the early stage, so the time
and labor costs are high. At the same time, due to random
integration, other gene sequences of mice may be destroyed
to obtain phenotypes unrelated to the target gene. Further-
more, Tg mice have suffered from artificial phenotypes
caused by overproduction or mislocalization, as well as
calpastatin deficiency-induced early lethality, calpain acti-
vation, neuronal cell death without tau pathology, en-
doplasmic reticulum stresses, and inflammasome
involvement. Such artifacts generate more uncertainties in
the interpretation of experimental results (Sasaguri et al.,
2022). KI is a gene-targeting technology that inserts the
target gene segment into a pre-selected site at a fixed point,
usually the Rosa26 locus (Friedrich and Soriano, 1991;
Soriano, 1999). Inserting the target gene at this site does not
affect the expression of endogenous genes in mice. The
mouse model constructed by this method has only one strain,
and the expression amount is controllable, but the over-
expression model cannot be obtained at one time.
Intraneuronal Aβ/APP vs. extracellular plaque
deposition
Zou et al. (2015) reported that the accumulation APP but not
in neurons was observed in 4–5 months old APP23 mice,
and the intraneuronal APP content was inversely correlated
with the absolute spine density and the relative number of
mushroom spines. This elucidates that APP accumulation
can lead to the decline of dendritic spines and cognitive
impairment, which is different from the mechanism of AD
caused by accumulation, suggesting that APP accumu-
lation in neurons should be paid attention to in the over-
expressed APP mouse models. Extracellular plaque
deposition has been considered as the major neuropatholo-
gical marker of AD, those mouse models showing only
plaque deposition provide strong evidence for understanding
plaque formation and the effect of plaque on other AD fea-
tures (Sanchez-Varo et al., 2022). Furthermore, no matter the
pathological features in these AD mouse models were in-
traneuronal pathology or extracellular plaque deposition,
they were accompanied by other pathological changes or
30 Li, X., et al. Sci China Life Sci
cognitive impairment, but the time of occurrence was dif-
ferent. From the perspective of drug research on AD, multi-
target therapy and asymptomatic phase intervention are the
current hotspot and key (Cheong et al., 2022;Crous-Bou et
al., 2017). Intraneuronal accumulation is an early event
of plaque formation. Intervention at the stage of accu-
mulation may provide more evidence for AD prevention and
treatment. Some mouse models showing intraneuronal
pathology without plaque formation also have other AD-
related pathological/behavioral changes, such as PS1V97L
Tg (Zhang et al., 2014) and APP E693Δ-Tg (Tomiyama et
al., 2010). In addition, much evidence indicates that simply
removing amyloid plaques may not significantly affect AD
progression, many drugs that are based on targets aim to
reduce production or accumulation (Karran and De
Strooper, 2022). As a result, these models can be used as
preclinical AD models for ultra-early intervention studies
(Sasaguri et al., 2017) and to study the effects of accu-
mulation on neurons, glial cells, synaptic morphology and
function, mitochondria, oxidative stress and neuroin-
flammation (Sehar et al., 2022), that are conducive to the
discovery of other pathways and therapeutic targets affecting
the pathogenesis of AD.
Inconsistency between transgenic mouse models and
clinical trials in drug development
In animal models, which are mechanistically simpler by
design, only a small subset of the molecules that may con-
tribute to disease are likely to be impacted, whereas other
aspects of human disease are not accurately reflected. There
is seldom a good model which reproduces all features of AD.
It is still unknown to what extent cellular phenotypes induced
in these models mimic human situation. Sixty-five million
years of evolutionary divergence cannot be ignored
(Sierksma et al., 2020). Due to overexpression artifacts,
unwanted genetic alterations, lengthy experiments and/or
ethical approval and considerations, drugs that show good
efficacy and safety in transgenic mouse models are often
failed to enter clinical trials (Sierksma et al., 2020). Targeted
drugs, either enhancers or suppressors, can exert their effects
on the expression of genes that are remote from their location
(Nott et al., 2019). Many enhancers affected by causal var-
iants are highly specific to brain region, cell type or state
(Huang et al., 2017;Nott et al., 2019). Many enhancer re-
gions rather than promoter regions, often myeloid and mi-
croglial, are significantly enriched for AD-associated
variants (Gjoneska et al., 2015;Nott et al., 2019). Further-
more, the genomic variants captured from different AD pa-
tients are different, but the pathological phenotypes of AD
patients are similar (Sierksma et al., 2020). In contrast, in
transgenic mouse models, different genomic variants lead to
various pathological phenotypes. Such inconsistency makes
it difficult to specify how AD-associated variants may exert
their effects on pathological phenotypes. As a result, there is
a huge gap between pathological phenotypes and genetic
phenotypes, which further contributes to the inconsistency
between drug development in transgenic models and clinical
trials.
In addition to the pathological phenotypes that were
present in AD, in clinical field, symptoms usually start to
show up along with the occurrence of tau pathology. In-
creasing evidence has demonstrated a strong correlation
between tau pathology and neurodegeneration and memory
impairment, indicating an essential role of pathological tau in
driving AD symptoms (Long and Holtzman, 2019;Veitch et
al., 2019). Mouse models that show tau pathology might be
indicative of symptom onset as well as disease progression,
which might be translated into clinical trials as outcome
measures of disease progression. The limitations, however,
are that tau pathology usually appears after amyloid pa-
thology, thus transgenic mouse models showing only tau
pathology were probably not suitable for early detection of
disease in preclinical or prodromal phase. In addition to
pathology and tau pathology, neurodegeneration (e.g., brain
atrophy) is another important pathologic feature for AD
patients, however, it is not developed in the amyloid mouse
models with known pathogenic gene mutation (regardless of
single or multi-transgenic), but developed in most MAPT
single-transgenic mouse models. A possible reason is that
neurodegeneration is a downstream effect, and more related
to tau pathology and disease progression, while amyloid
pathology usually occurs earlier and is probably an upstream
mechanism that acts together with many other pathogenic
pathways to finally lead to neurodegeneration. As a result,
these mouse models are limited in testing downstream ef-
fects.
Although this review mainly focuses on AD transgenic
mouse models, which are the most widely used ones at
present, there are other AD models that have some ad-
vantages over mouse models in some way. Drosophila and
C.elegans are also often used animal models for AD
(Prüßing et al., 2013;van Ham et al., 2009). Although they
are somewhat deficient in cognitive function assessment
compared to rodents, both are model animals with simple
genetic manipulation, which can easily construct transgenic
models of single or double copies of Aβ42 expressed by
whole neurons or motor neurons in the central nervous sys-
tem, thus are suitable as preliminary screening models for
new drugs of AD treatment. At present, the lag of experi-
mental model animal research and the failure to fully cover
and replicate various pathologies and phenotypes of AD are
the bottlenecks restricting the research on AD mechanism
and drug development. Recently, Pang et al. constructed a
transgenic rat model that carries three human-derived APP
gene mutations AppNL-G-F, and the study showed that the rat
31
Li, X., et al. Sci China Life Sci
model presented more similar pathology and disease pro-
gression to human AD patients than other existing AD ani-
mal models (Pang et al., 2022), which suggests that rat model
might be of great significance in studying the pathological
development and mechanism of AD in the future, and may
also be more suitable for the development and verification of
some potential anti-AD drugs. Non-human primates and
human beings are similar in brain anatomy, neuropathy
characteristics and biological behavior patterns, especially
rhesus monkeys. In the brains of rhesus monkeys, observed
deposition containing senile plaques and neurofibrillary
tangled phenomenon, whether spontaneous or induced
models, can better replicate AD-related pathological and
physiological characteristics, but expensive costs and scarce
resources limit the large number of applications of non-hu-
man primates (Capitanio and Emborg, 2008).
APP-based transgenic models were successful in predict-
ing molecular pathways that could impact amyloid accu-
mulation in the clinic, but had less value in predicting the
specific properties of anti-Aβ therapies that were most likely
to reduce plaque load in patients, and had not yet translated
into robust effects on cognition in well-powered clinical
trials (Scearce-Levie et al., 2020). Such discrepancy may be
due to: the specific APP-based model, end points used in
preclinical studies, the limitations inherent in clinical trials,
and uncertainty about the specific form of that drives
pathology (Karran and Hardy, 2014). In addition, pa-
thology may not be the direct cause of neurodegeneration
and cognitive deficits. It is very likely that the very few
antibody-based clinical trials reported to show some effects
in the early stage of AD take effects by reducing pa-
thology-induced downstream detrimental events such as tau
pathology and neuroinflammation, that serve as the real
driver of neurodegenerative and cognitive impairment. As a
result, APP-based transgenic models might be useful in
testing anti-Aβ therapies at the early stage of disease, while
multi-target therapeutic strategies might be more suitable for
later stage treatment. The probability of translation of pre-
clinical findings to the clinic depends on both the type of
model and the end points selected. The chance of translating
to human decreases as the focus shifts from end points of the
actual target to end points such as cognitive behaviors or
brain atrophy (Scearce-Levie et al., 2020). Scientists have
proposed a framework that is centered on the use of animal
models to explore disease-relevant biology proximal to the
target of interest, and to identify candidate biomarkers with
potential clinical translatability (Scearce-Levie et al., 2020).
Bridging the gap would be critical and helpful for stratifi-
cation of patients for clinical trials, and personalized medi-
cine based on genetic profile.
Furthermore, in clinical practice, the phenomenon of fa-
milial co-segregation is very strong, however, in animal
models, we sometimes fail to get positive results to verify the
genetic phenomenon. Possible reasons are: (i) there might be
other synergistic gene(s) in the family, which was not con-
structed into the mouse model; (ii) there might be a lack of
corresponding intermediate link or pathway for the patho-
genic gene mutation to play the same role in the mouse
model, which could lead to the limited pathogenic effects of
the gene; (iii) the pathogenesis of AD is complex, which is
affected by various factors including genetic and environ-
mental factors, as well as lifestyles, and these factors are
difficult if not impossible to mimic or model in mice, which
may affect the results.
Conclusion and future perspectives
Transgenic mouse models have development prospects,
which can explain the different mechanisms of different
mutations and can be used for a wider range of studies. First,
they can be used for studying gene specific mechanism,
targeted drug development and gene therapy. Besides the
known pathogenic genes, more and more gene mutation sites
of the three known pathogenic genes, as well as unknown
mutations, have been found in FAD, and subsequent devel-
opment of corresponding mouse models is on the way.
Second, they can be used to study drugs with multi-target
potential efficacy. There have been studies of multi-target
drugs in the early stage of transgenic mouse models, namely
various Chinese medicine monomers, and some have entered
clinical trials. Such preclinical findings of multi-target
therapy need to be further verified or reproduced in-
dependently in other models and labs. Third, they can be
used to combine with risk factor models so as to study the
interaction of genes and risk factors, which provides a basis
for developing prevention strategies. In large-scale popula-
tion studies, a variety of risk factors for AD have been found,
namely diabetes, cerebrovascular diseases, depression, an-
xiety, malaise and fatigue, high cholesterol level and ab-
normal weight loss (Gauthier et al., 2012;Jia et al., 2020a;
Nedelec et al., 2022;Yu et al., 2020). More and more evi-
dence suggests that future AD treatment should develop to-
wards multi targets and combination therapy (Delrieu et al.,
2022), and pay more attention to the diversity and hetero-
geneity of the population (Peroutka, 2022), as well as gene-
environment interaction (Migliore and Coppedè, 2022), so as
to better reflect the real world situation.
Among the various AD transgenic models, special atten-
tion should be paid to AβO related models due to their pa-
thogenic features and promising translational advantages.
Future studies in investigating the trajectories of AβO related
pathologic changes, extended upstream and downstream
mechanisms and pathways are needed to optimize treatment
and prevention strategy. In addition, AD-related biomarkers
and behavioral phenotypes shared by models and humans
32 Li, X., et al. Sci China Life Sci
should be developed for better translation from models to
clinical trials.
Compliance and ethics The author(s) declare that they have no conflict
of interest.
Acknowledgements This work was supported by the National Natural
Science Foundation of China (U20A20354, 81530036), Beijing Brain In-
itiative from Beijing Municipal Science & Technology Commission
(Z201100005520016, Z201100005520017), the National Major R&D Pro-
jects of China-Scientific Technological Innovation 2030 (2021ZD0201802),
the National Key Scientific Instrument and Equipment Development Project
(31627803), Youth Program of National Natural Science Foundation of
China (81801048, 82101503) and Youth Elite Scientists Sponsorship Pro-
gram by CAST (YESS20200155). We would like to thank Shuya Nie, Fangyu
Li, Bingqiu Li and Wenying Liu for literature search for the clinical trans-
lation related sections.
References
Adalbert, R., Nogradi, A., Babetto, E., Janeckova, L., Walker, S.A.,
Kerschensteiner, M., Misgeld, T., and Coleman, M.P. (2009). Severely
dystrophic axons at amyloid plaques remain continuous and connected
to viable cell bodies. Brain 132, 402–416.
Alexander, A.G., Marfil, V., and Li, C. (2014). Use of Caenorhabditis
elegans as a model to study Alzheimer’s disease and other
neurodegenerative diseases. Front Genet 5, 279.
Almkvist, O., Rodriguez-Vieitez, E., Thordardottir, S., Nordberg, A.,
Viitanen, M., Lannfelt, L., and Graff, C. (2019). Longitudinal cognitive
decline in autosomal-dominant Alzheimer’s disease varies with
mutations in APP and PSEN1 genes. Neurobiol Aging 82, 40–47.
An, J., Zhou, Y., Zhang, M., Xie, Y., Ke, S., Liu, L., Pan, X., and Chen, Z.
(2019). Exenatide alleviates mitochondrial dysfunction and cognitive
impairment in the 5×FAD mouse model of Alzheimer’s disease. Behav
Brain Res 370, 111932.
Ano, Y., Takaichi, Y., Ohya, R., Uchida, K., Nakayama, H., and Takashima,
A. (2023). Tryptophan-tyrosine dipeptide improves tau-related
symptoms in tauopathy mice. Nutr Neurosci 26, 766–777.
Arancio, O., Zhang, H.P., Chen, X., Lin, C., Trinchese, F., Puzzo, D., Liu,
S., Hegde, A., Yan, S.F., Stern, A., et al. (2004). RAGE potentiates Aβ-
induced perturbation of neuronal function in transgenic mice. EMBO J
23, 4096–4105.
Arendash, G.W., King, D.L., Gordon, M.N., Morgan, D., Hatcher, J.M.,
Hope, C.E., and Diamond, D.M. (2001). Progressive, age-related
behavioral impairments in transgenic mice carrying both mutant
amyloid precursor protein and presenilin-1 transgenes. Brain Res 891,
42–53.
Armbrust, F., Bickenbach, K., Marengo, L., Pietrzik, C., and Becker-Pauly,
C. (2022). The Swedish dilemma—the almost exclusive use of
APPswe-based mouse models impedes adequate evaluation of
alternative β-secretases. Biochim Biophys Acta 1869, 119164.
Baglietto-Vargas, D., Forner, S., Cai, L., Martini, A.C., Trujillo-Estrada, L.,
Swarup, V., Nguyen, M.M.T., Do Huynh, K., Javonillo, D.I., Tran, K.
M., et al. (2021). Generation of a humanized expressing mouse
demonstrating aspects of Alzheimer’s disease-like pathology. Nat
Commun 12, 2421.
Barrow, P., Villabruna, L., Hoberman, A., Bohrmann, B., Richter, W.F., and
Schubert, C. (2017). Reproductive and developmental toxicology
studies with gantenerumab in PS2APP transgenic mice. Reprod
Toxicol 73, 362–371.
Barrow, P.A., Empson, R.M., Gladwell, S.J., Anderson, C.M., Killick, R.,
Yu, X., Jefferys, J.G.R., and Duff, K. (2000). Functional phenotype in
transgenic mice expressing mutant human presenilin-1. Neurobiol Dis
7, 119–126.
Bayer, T.A. (2022). Pyroglutamate cascade as drug target in
Alzheimer’s disease. Mol Psychiatry 27, 1880–1885.
Begley, J.G., Duan, W., Chan, S., Duff, K., and Mattson, M.P. (1999).
Altered calcium homeostasis and mitochondrial dysfunction in cortical
synaptic compartments of presenilin-1 mutant mice. J Neurochem 72,
1030–1039.
Bellucci, A., Rosi, M.C., Grossi, C., Fiorentini, A., Luccarini, I., and
Casamenti, F. (2007). Abnormal processing of tau in the brain of aged
TgCRND8 mice. Neurobiol Dis 27, 328–338.
Bellucci, A., Westwood, A.J., Ingram, E., Casamenti, F., Goedert, M., and
Spillantini, M.G. (2004). Induction of inflammatory mediators and
microglial activation in mice transgenic for mutant human P301S tau
protein. Am J Pathol 165, 1643–1652.
Billings, L.M., Oddo, S., Green, K.N., McGaugh, J.L., and LaFerla, F.M.
(2005). Intraneuronal causes the onset of early Alzheimer’s disease-
related cognitive deficits in transgenic mice. Neuron 45, 675–688.
Bittner, T., Burgold, S., Dorostkar, M.M., Fuhrmann, M., Wegenast-Braun,
B.M., Schmidt, B., Kretzschmar, H., and Herms, J. (2012). Amyloid
plaque formation precedes dendritic spine loss. Acta Neuropathol 124,
797–807.
Blázquez, G., Cañete, T., Tobeña, A., Giménez-Llort, L., and Fernández-
Teruel, A. (2014). Cognitive and emotional profiles of aged
Alzheimer’s disease (3×TgAD) mice: Effects of environmental
enrichment and sexual dimorphism. Behav Brain Res 268, 185–201.
Boekhoorn, K., Terwel, D., Biemans, B., Borghgraef, P., Wiegert, O.,
Ramakers, G.J.A., de Vos, K., Krugers, H., Tomiyama, T., Mori, H., et
al. (2006). Improved long-term potentiation and memory in young tau-
P301L transgenic mice before onset of hyperphosphorylation and
tauopathy. J Neurosci 26, 3514–3523.
Boncristiano, S., Calhoun, M.E., Howard, V., Bondolfi, L., Kaeser, S.A.,
Wiederhold, K.H., Staufenbiel, M., and Jucker, M. (2005). Neocortical
synaptic bouton number is maintained despite robust amyloid
deposition in APP23 transgenic mice. Neurobiol Aging 26, 607–613.
Borchelt, D.R., Ratovitski, T., van Lare, J., Lee, M.K., Gonzales, V.,
Jenkins, N.A., Copeland, N.G., Price, D.L., and Sisodia, S.S. (1997).
Accelerated amyloid deposition in the brains of transgenic mice
coexpressing mutant presenilin 1 and amyloid precursor proteins.
Neuron 19, 939–945.
Borchelt, D.R., Thinakaran, G., Eckman, C.B., Lee, M.K., Davenport, F.,
Ratovitsky, T., Prada, C.M., Kim, G., Seekins, S., Yager, D., et al.
(1996). Familial Alzheimer’s disease-linked presenilin 1 variants
elevate Aβ1–42/1–40 ratio in vitro and in vivo.Neuron 17, 1005–1013.
Bouter, Y., Dietrich, K., Wittnam, J.L., Rezaei-Ghaleh, N., Pillot, T., Papot-
Couturier, S., Lefebvre, T., Sprenger, F., Wirths, O., Zweckstetter, M., et
al. (2013). N-truncated amyloid β (Aβ) 4-42 forms stable aggregates
and induces acute and long-lasting behavioral deficits. Acta
Neuropathol 126, 189–205.
Brautigam, H., Steele, J.W., Westaway, D., Fraser, P.E., George-Hyslop, P.
H.S., Gandy, S., Hof, P.R., and Dickstein, D.L. (2012). The isotropic
fractionator provides evidence for differential loss of hippocampal
neurons in two mouse models of Alzheimer’s disease. Mol
Neurodegener 7, 58.
Breyhan, H., Wirths, O., Duan, K., Marcello, A., Rettig, J., and Bayer, T.A.
(2009). APP/PS1KI bigenic mice develop early synaptic deficits and
hippocampus atrophy. Acta Neuropathol 117, 677–685.
Brown, J.T., Richardson, J.C., Collingridge, G.L., Randall, A.D., and
Davies, C.H. (2005). Synaptic transmission and synchronous activity is
disrupted in hippocampal slices taken from aged TAS10 mice.
Hippocampus 15, 110–117.
Bruggink, K.A., Jongbloed, W., Biemans, E.A.L.M., Veerhuis, R.,
Claassen, J.A.H.R., Kuiperij, H.B., and Verbeek, M.M. (2013).
Amyloid-β oligomer detection by ELISA in cerebrospinal fluid and
brain tissue. Anal Biochem 433, 112–120.
Buskila, Y., Crowe, S.E., and Ellis-Davies, G.C.R. (2013). Synaptic deficits
in layer 5 neurons precede overt structural decay in 5xFAD mice.
Neuroscience 254, 152–159.
Cacciottolo, M., Morgan, T.E., and Finch, C.E. (2021). Age, sex, and
33
Li, X., et al. Sci China Life Sci
cerebral microbleeds in EFAD Alzheimer disease mice. Neurobiol
Aging 103, 42–51.
Calhoun, M.E., Wiederhold, K.H., Abramowski, D., Phinney, A.L., Probst,
A., Sturchler-Pierrat, C., Staufenbiel, M., Sommer, B., and Jucker, M.
(1998). Neuron loss in APP transgenic mice. Nature 395, 755–756.
Capitanio, J.P., and Emborg, M.E. (2008). Contributions of non-human
primates to neuroscience research. Lancet 371, 1126–1135.
Caruso, D., Barron, A.M., Brown, M.A., Abbiati, F., Carrero, P., Pike, C.J.,
Garcia-Segura, L.M., and Melcangi, R.C. (2013). Age-related changes
in neuroactive steroid levels in 3xTg-AD mice. Neurobiol Aging 34,
1080–1089.
Casas, C., Sergeant, N., Itier, J.M., Blanchard, V., Wirths, O., van der Kolk,
N., Vingtdeux, V., van de Steeg, E., Ret, G., Canton, T., et al. (2004).
Massive CA1/2 neuronal loss with intraneuronal and N-terminal
truncated Aβ42 accumulation in a novel Alzheimer transgenic model.
Am J Pathol 165, 1289–1300.
Chang, L., Bakhos, L., Wang, Z., Venton, D.L., and Klein, W.L. (2003).
Femtomole immunodetection of synthetic and endogenous amyloid-β
oligomers and its application to Alzheimer’s Disease drug candidate
screening. J Mol Neurosci 20, 305–314.
Chapman, P.F., White, G.L., Jones, M.W., Cooper-Blacketer, D., Marshall,
V.J., Irizarry, M., Younkin, L., Good, M.A., Bliss, T.V.P., Hyman, B.T.,
et al. (1999). Impaired synaptic plasticity and learning in aged amyloid
precursor protein transgenic mice. Nat Neurosci 2, 271–276.
Chen, S., and Jia, J. (2020). Tenuifolin attenuates amyloid-β42-induced
neuroinflammation in microglia through the NF-κB signaling pathway.
J Alzheimer Dis 76, 195–205.
Chen, S.Y., Gao, Y., Sun, J.Y., Meng, X.L., Yang, D., Fan, L.H., Xiang, L.,
and Wang, P. (2020). Traditional Chinese medicine: role in reducing β-
amyloid, apoptosis, autophagy, neuroinflammation, oxidative stress,
and mitochondrial dysfunction of Alzheimer’s disease. Front Pharmacol
11, 497.
Chen, Y., Wang, Y., Qin, Q., Zhang, Y., Xie, L., Xiao, J., Cao, Y., Su, Z.,
and Chen, Y. (2022). Carnosic acid ameliorated Aβ-mediated (amyloid-
β peptide) toxicity, cholinergic dysfunction and mitochondrial defect in
Caenorhabditis elegans of Alzheimer’s Model. Food Funct 13, 4624–
4640.
Cheng-Hathaway, P.J., Reed-Geaghan, E.G., Jay, T.R., Casali, B.T.,
Bemiller, S.M., Puntambekar, S.S., von Saucken, V.E., Williams, R.
Y., Karlo, J.C., Moutinho, M., et al. (2018). The Trem2 R47H variant
confers loss-of-function-like phenotypes in Alzheimer’s disease. Mol
Neurodegener 13, 29.
Cheng, I.H., Palop, J.J., Esposito, L.A., Bien-Ly, N., Yan, F., and Mucke, L.
(2004). Aggressive amyloidosis in mice expressing human amyloid
peptides with the Arctic mutation. Nat Med 10, 1190–1192.
Cheng, I.H., Scearce-Levie, K., Legleiter, J., Palop, J.J., Gerstein, H., Bien-
Ly, N., Puolivaöli, J., Lesné, S., Ashe, K.H., Muchowski, P.J., et al.
(2007). Accelerating amyloid-β fibrillization reduces oligomer levels
and functional deficits in Alzheimer disease mouse models. J Biol
Chem 282, 23818–23828.
Cheong, S.L., Tiew, J.K., Fong, Y.H., Leong, H.W., Chan, Y.M., Chan, Z.
L., and Kong, E.W.J. (2022). Current pharmacotherapy and multi-target
approaches for Alzheimer’s disease. Pharmaceuticals 15, 1560.
Chishti, M.A., Yang, D.S., Janus, C., Phinney, A.L., Horne, P., Pearson, J.,
Strome, R., Zuker, N., Loukides, J., French, J., et al. (2001). Early-onset
amyloid deposition and cognitive deficits in transgenic mice expressing
a double mutant form of amyloid precursor protein 695. J Biol Chem
276, 21562–21570.
Cocco, S., Rinaudo, M., Fusco, S., Longo, V., Gironi, K., Renna, P., Aceto,
G., Mastrodonato, A., Li Puma, D.D., Podda, M.V., et al. (2020).
Plasma BDNF levels following transcranial direct current stimulation
allow prediction of synaptic plasticity and memory deficits in 3×Tg-AD
mice. Front Cell Dev Biol 8, 541.
Codita, A., Gumucio, A., Lannfelt, L., Gellerfors, P., Winblad, B.,
Mohammed, A.H., and Nilsson, L.N.G. (2010). Impaired behavior of
female tg-ArcSwe APP mice in the IntelliCage: A longitudinal study.
Behav Brain Res 215, 83–94.
Cohen, R.M., Rezai-Zadeh, K., Weitz, T.M., Rentsendorj, A., Gate, D.,
Spivak, I., Bholat, Y., Vasilevko, V., Glabe, C.G., Breunig, J.J., et al.
(2013). A transgenic Alzheimer rat with plaques, tau pathology,
behavioral impairment, oligomeric Aβ, and frank neuronal loss. J
Neurosci 33, 6245–6256.
Colton, C.A., Wilcock, D.M., Wink, D.A., Davis, J., Van Nostrand, W.E.,
and Vitek, M.P. (2008). The effects of NOS2 gene deletion on mice
expressing mutated human AbetaPP. J Alzheimers Dis 15, 571–587.
Congdon, E.E., Pan, R., Jiang, Y., Sandusky-Beltran, L.A., Dodge, A., Lin,
Y., Liu, M., Kuo, M.H., Kong, X.P., and Sigurdsson, E.M. (2022).
Single domain antibodies targeting pathological tau protein: Influence
of four IgG subclasses on efficacy and toxicity. Ebiomedicine 84,
104249.
Cook, C., Dunmore, J.H., Murray, M.E., Scheffel, K., Shukoor, N., Tong,
J., Castanedes-Casey, M., Phillips, V., Rousseau, L., Penuliar, M.S., et
al. (2014). Severe amygdala dysfunction in a MAPT transgenic mouse
model of frontotemporal dementia. Neurobiol Aging 35, 1769–1777.
Coppola, G., Chinnathambi, S., Lee, J.J.Y., Dombroski, B.A., Baker, M.C.,
Soto-Ortolaza, A.I., Lee, S.E., Klein, E., Huang, A.Y., Sears, R., et al.
(2012). Evidence for a role of the rare p.A152T variant in MAPT in
increasing the risk for FTD-spectrum and Alzheimer’s diseases. Hum
Mol Genet 21, 3500–3512.
Cramer, P.E., Cirrito, J.R., Wesson, D.W., Lee, C.Y.D., Karlo, J.C., Zinn, A.
E., Casali, B.T., Restivo, J.L., Goebel, W.D., James, M.J., et al. (2012).
ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits
in AD mouse models. Science 335, 1503–1506.
Cross, D.J., Huber, B.R., Silverman, M.A., Cline, M.M., Gill, T.B., Cross,
C.G., Cook, D.G., and Minoshima, S. (2021). Intranasal paclitaxel alters
Alzheimer’s disease phenotypic features in 3xTg-AD mice. J Alzheimer
Dis 83, 379–394.
Crous-Bou, M., Minguillón, C., Gramunt, N., and Molinuevo, J.L. (2017).
Alzheimer’s disease prevention: from risk factors to early intervention.
Alzheimers Res Ther 9, 71.
Crouzin, N., Baranger, K., Cavalier, M., Marchalant, Y., Cohen-Solal, C.,
Roman, F.S., Khrestchatisky, M., Rivera, S., Feron, F., and Vignes, M.
(2013). Area-specific alterations of synaptic plasticity in the 5XFAD
mouse model of Alzheimer’s disease: dissociation between somato-
sensory cortex and hippocampus. PLoS ONE 8, e74667.
Cummings, J., Lee, G., Nahed, P., Kambar, M.E.Z.N., Zhong, K., Fonseca,
J., and Taghva, K. (2022). Alzheimer’s disease drug development
pipeline: 2022. Alzheimers Dement 8, e12295.
Davis, J., Xu, F., Deane, R., Romanov, G., Previti, M.L., Zeigler, K.,
Zlokovic, B.V., and Van Nostrand, W.E. (2004). Early-onset and robust
cerebral microvascular accumulation of amyloid β-protein in transgenic
mice expressing low levels of a vasculotropic Dutch/Iowa mutant form
of amyloid β-protein precursor. J Biol Chem 279, 20296–20306.
Davis, J., Xu, F., Miao, J., Previti, M.L., Romanov, G., Ziegler, K., and Van
Nostrand, W.E. (2006). Deficient cerebral clearance of vasculotropic
mutant Dutch/Iowa double in human AßPP transgenic mice.
Neurobiol Aging 27, 946–954.
de Oliveira, P., Cella, C., Locker, N., Ravindran, K.K.G., Mendis, A.,
Wafford, K., Gilmour, G., Dijk, D.J., and Winsky-Sommerer, R. (2022).
Improved sleep, memory, and cellular pathological features of
tauopathy, including the NLRP3 inflammasome, after chronic
administration of trazodone in rTg4510 mice. J Neurosci 42, 3494–
3509.
Decker, J.M., Krüger, L., Sydow, A., Dennissen, F.J., Siskova, Z.,
Mandelkow, E., and Mandelkow, E. (2016). The Tau/A152T
mutation, a risk factor for frontotemporal-spectrum disorders, leads to
NR2B receptor-mediated excitotoxicity. EMBO Rep 17, 552–569.
Decker, J.M., Krüger, L., Sydow, A., Zhao, S., Frotscher, M., Mandelkow,
E., and Mandelkow, E.M. (2015). Pro-aggregant Tau impairs mossy
fiber plasticity due to structural changes and Ca++ dysregulation. Acta
Neuropathol Commun 3, 23.
Delrieu, J., Bateman, R.J., Touchon, J., Sabbagh, M., and Cummings, J.
(2022). The future of AD clinical trials with the advent of anti-amyloid
therapies: an CTAD Task force report. J Prev Alzheimers Dis 9, 393–
34 Li, X., et al. Sci China Life Sci
399.
Dennissen, F.J.A., Anglada-Huguet, M., Sydow, A., Mandelkow, E., and
Mandelkow, E.M. (2016). Adenosine A1receptor antagonist rolofylline
alleviates axonopathy caused by human Tau ΔK280. Proc Natl Acad Sci
USA 113, 11597–11602.
Dewachter, I., Van Dorpe, J., Smeijers, L., Gilis, M., Kuipéri, C., Laenen,
I., Caluwaerts, N., Moechars, D., Checler, F., Vanderstichele, H., et al.
(2000). Aging increased amyloid peptide and caused amyloid plaques in
brain of old APP/V717I transgenic mice by a different mechanism than
mutant presenilin1. J Neurosci 20, 6452–6458.
Dodart, J.C., Meziane, H., Mathis, C., Bales, K.R., Paul, S.M., and
Ungerer, A. (1999). Behavioral disturbances in transgenic mice
overexpressing the V717F Β-amyloid precursor protein. Behav
Neurosci 113, 982–990.
Dodiya, H.B., Frith, M., Sidebottom, A., Cao, Y., Koval, J., Chang, E., and
Sisodia, S.S. (2020). Synergistic depletion of gut microbial consortia,
but not individual antibiotics, reduces amyloidosis in APPPS1-21
Alzheimer’s transgenic mice. Sci Rep 10, 8183.
Dodiya, H.B., Lutz, H.L., Weigle, I.Q., Patel, P., Michalkiewicz, J., Roman-
Santiago, C.J., Zhang, C.M., Liang, Y., Srinath, A., Zhang, X., et al.
(2022). Gut microbiota-driven brain amyloidosis in mice requires
microglia. J Exp Med 219.
Domnitz, S.B., Robbins, E.M., Hoang, A.W., Garcia-Alloza, M., Hyman,
B.T., Rebeck, G.W., Greenberg, S.M., Bacskai, B.J., and Frosch, M.P.
(2005). Progression of cerebral amyloid angiopathy in transgenic mouse
models of Alzheimer disease. J Neuropathol Exp Neurol 64, 588–594.
Drummond, E., and Wisniewski, T. (2017). Alzheimer’s disease:
experimental models and reality. Acta Neuropathol 133, 155–175.
Dudal, S., Krzywkowski, P., Paquette, J., Morissette, C., Lacombe, D.,
Tremblay, P., and Gervais, F. (2004). Inflammation occurs early during
the deposition process in TgCRND8 mice. Neurobiol Aging 25,
861–871.
Duff, K., Eckman, C., Zehr, C., Yu, X., Prada, C.M., Perez-Tur, J., Hutton,
M., Buee, L., Harigaya, Y., Yager, D., et al. (1996). Increased amyloid-
β42(43) in brains of mice expressing mutant presenilin 1. Nature 383,
710–713.
Eckermann, K., Mocanu, M.M., Khlistunova, I., Biernat, J., Nissen, A.,
Hofmann, A., Schönig, K., Bujard, H., Haemisch, A., Mandelkow, E., et
al. (2007). The β-propensity of Tau determines aggregation and synaptic
loss in inducible mouse models of tauopathy. J Biol Chem 282, 31755–
31765.
Elder, G.A., Gama Sosa, M.A., De Gasperi, R., Dickstein, D.L., and Hof, P.
R. (2010). Presenilin transgenic mice as models of Alzheimer’s disease.
Brain Struct Funct 214, 127–143.
Elhaik Goldman, S., Goez, D., Last, D., Naor, S., Liraz Zaltsman, S.,
Sharvit-Ginon, I., Atrakchi-Baranes, D., Shemesh, C., Twitto-
Greenberg, R., Tsach, S., et al. (2018). High-fat diet protects the
blood-brain barrier in an Alzheimer’s disease mouse model. Aging Cell
17, e12818.
Evans, C.E., Thomas, R.S., Freeman, T.J., Hvoslef-Eide, M., Good, M.A.,
and Kidd, E.J. (2019). Selective reduction of APP-BACE1 activity
improves memory via NMDA-NR2B receptor-mediated mechanisms in
aged PDAPP mice. Neurobiol Aging 75, 136–149.
Fan, R., Xu, F., Previti, M.L., Davis, J., Grande, A.M., Robinson, J.K., and
Van Nostrand, W.E. (2007). Minocycline reduces microglial activation
and improves behavioral deficits in a transgenic model of cerebral
microvascular amyloid. J Neurosci 27, 3057–3063.
Fernandez-Funez, P., de Mena, L., and Rincon-Limas, D.E. (2015).
Modeling the complex pathology of Alzheimer’s disease in
Drosophila.Exp Neurol 274, 58–71.
Ferretti, M.T., Bruno, M.A., Ducatenzeiler, A., Klein, W.L., and Cuello, A.
C. (2012). Intracellular Aβ-oligomers and early inflammation in a
model of Alzheimer’s disease. Neurobiol Aging 33, 1329–1342.
Ferretti, M.T., Partridge, V., C. Leon, W., Canneva, F., Allard, S., N.
Arvanitis, D., Vercauteren, F., Houle, D., Ducatenzeiler, A., L. Klein,
W., et al. (2011). Transgenic mice as a model of pre-clinical Alzheimers
disease. Curr Alzheimer Res 8, 4–23.
Flanigan, T.J., Xue, Y., Kishan Rao, S., Dhanushkodi, A., and McDonald,
M.P. (2014). Abnormal vibrissa-related behavior and loss of barrel field
inhibitory neurons in 5xFAD transgenics. Genes Brain Behav 13, 488–
500.
Foster, J.B., Lashley, R., Zhao, F., Wang, X., Kung, N., Askwith, C.C., Lin,
L., Shultis, M.W., Hodgetts, K.J., and Lin, C.L.G. (2019). Enhancement
of tripartite synapses as a potential therapeutic strategy for Alzheimer’s
disease: a preclinical study in rTg4510 mice. Alzheimers Res Ther 11,
75.
Frautschy, S.A., Yang, F., Irrizarry, M., Hyman, B., Saido, T.C., Hsiao, K.,
and Cole, G.M. (1998). Microglial response to amyloid plaques in
APPsw transgenic mice. Am J Pathol 152, 307–317.
Friedrich, G., and Soriano, P. (1991). Promoter traps in embryonic stem
cells: a genetic screen to identify and mutate developmental genes in
mice. Genes Dev 5, 1513–1523.
Frost, J.L., Liu, B., Rahfeld, J.U., Kleinschmidt, M., O’Nuallain, B., Le, K.
X., Lues, I., Caldarone, B.J., Schilling, S., Demuth, H.U., et al. (2015).
An anti-pyroglutamate-3 vaccine reduces plaques and improves
cognition in APPswe/PS1ΔE9 mice. Neurobiol Aging 36, 3187–3199.
Gaj, T., Gersbach, C.A., and Barbas Iii, C.F. (2013). ZFN, TALEN, and
CRISPR/Cas-based methods for genome engineering. Trends
Biotechnol 31, 397–405.
Gamage, K.K., and Kumar, S. (2017). Aducanumab therapy ameliorates
calcium overload in a mouse model of Alzheimer’s disease. J Neurosci
37, 4430–4432.
Games, D., Adams, D., Alessandrini, R., Barbour, R., Borthelette, P.,
Blackwell, C., Carr, T., Clemens, J., Donaldson, T., Gillespie, F., et al.
(1995). Alzheimer-type neuropathology in transgenic mice
overexpressing V717F β-amyloid precursor protein. Nature 373, 523–
527.
Gandy, S., Simon, A.J., Steele, J.W., Lublin, A.L., Lah, J.J., Walker, L.C.,
Levey, A.I., Krafft, G.A., Levy, E., Checler, F., et al. (2010). Days to
criterion as an indicator of toxicity associated with human Alzheimer
amyloid-β oligomers. Ann Neurol 68, 220–230.
Garcia-Alloza, M., Robbins, E.M., Zhang-Nunes, S.X., Purcell, S.M.,
Betensky, R.A., Raju, S., Prada, C., Greenberg, S.M., Bacskai, B.J., and
Frosch, M.P. (2006). Characterization of amyloid deposition in the
APPswe/PS1dE9 mouse model of Alzheimer disease. Neurobiol Dis 24,
516–524.
Garrick, D., Fiering, S., Martin, D.I.K., and Whitelaw, E. (1998). Repeat-
induced gene silencing in mammals. Nat Genet 18, 56–59.
Gauthier, S., Wu, L., Rosa-Neto, P., and Jia, J. (2012). Prevention strategies
for Alzheimer’s disease. Transl Neurodegener 1, 13.
Gelman, S., Palma, J., Tombaugh, G., and Ghavami, A. (2018). Differences
in synaptic dysfunction between rTg4510 and APP/PS1 mouse models
of Alzheimer’s disease. J Alzheimer Dis 61, 195–208.
Gengler, S., Hamilton, A., and Holscher, C. (2010). Synaptic plasticity in
the hippocampus of a APP/PS1 mouse model of Alzheimer’s disease is
impaired in old but not young mice. PLoS ONE 5, e9764.
Giannoni, P., Arango-Lievano, M., Neves, I.D., Rousset, M.C., Baranger,
K., Rivera, S., Jeanneteau, F., Claeysen, S., and Marchi, N. (2016).
Cerebrovascular pathology during the progression of experimental
Alzheimer’s disease. Neurobiol Dis 88, 107–117.
Gjoneska, E., Pfenning, A.R., Mathys, H., Quon, G., Kundaje, A., Tsai, L.
H., and Kellis, M. (2015). Conserved epigenomic signals in mice and
humans reveal immune basis of Alzheimer’s disease. Nature 518, 365–
369.
Goodwin, M.S., Sinyavskaya, O., Burg, F., O’Neal, V., Ceballos-Diaz, C.,
Cruz, P.E., Lewis, J., Giasson, B.I., Davies, P., Golde, T.E., et al. (2021).
Anti-tau scFvs targeted to the cytoplasm or secretory pathway variably
modify pathology and neurodegenerative phenotypes. Mol Ther 29,
859–872.
Gordon, M.N., Holcomb, L.A., Jantzen, P.T., DiCarlo, G., Wilcock, D.,
Boyett, K.W., Connor, K., Melachrino, J., O’Callaghan, J.P., and
Morgan, D. (2002). Time course of the development of Alzheimer-like
pathology in the doubly transgenic PS1+APP mouse. Exp Neurol 173,
183–195.
35
Li, X., et al. Sci China Life Sci
Götz, J., Bodea, L.G., and Goedert, M. (2018). Rodent models for
Alzheimer disease. Nat Rev Neurosci 19, 583–598.
Govindarajan, N., Agis-Balboa, R.C., Walter, J., Sananbenesi, F., and
Fischer, A. (2011). Sodium butyrate improves memory function in an
Alzheimer’s disease mouse model when administered at an advanced
stage of disease progression. J Alzheimer Dis 26, 187–197.
Gratuze, M., Leyns, C.E.G., Sauerbeck, A.D., St-Pierre, M.K., Xiong, M.,
Kim, N., Serrano, J.R., Tremblay, M.È., Kummer, T.T., Colonna, M., et
al. (2020). Impact of TREM2R47H variant on tau pathology-induced
gliosis and neurodegeneration. J Clin Invest 130, 4954–4968.
Grueninger, F., Bohrmann, B., Czech, C., Ballard, T.M., Frey, J.R.,
Weidensteiner, C., von Kienlin, M., and Ozmen, L. (2010).
Phosphorylation of Tau at S422 is enhanced by in TauPS2APP
triple transgenic mice. Neurobiol Dis 37, 294–306.
Gunawardena, S., and Goldstein, L.S.B. (2001). Disruption of axonal
transport and neuronal viability by amyloid precursor protein mutations
in Drosophila.Neuron 32, 389–401.
Guo, Q., Fu, W., Sopher, B.L., Miller, M.W., Ware, C.B., Martin, G.M., and
Mattson, M.P. (1999). Increased vulnerability of hippocampal neurons
to excitotoxic necrosis in presenilin-1 mutant knock-in mice. Nat Med
5, 101–106.
Hafezparast, M., Ahmad-Annuar, A., Wood, N.W., Tabrizi, S.J., and Fisher,
E.M. (2002). Mouse models for neurological disease. Lancet Neurol 1,
215–224.
Hampton, D.W., Webber, D.J., Bilican, B., Goedert, M., Spillantini, M.G.,
and Chandran, S. (2010). Cell-mediated neuroprotection in a mouse
model of human tauopathy. J Neurosci 30, 9973–9983.
Hara, M., Hirokawa, K., Kamei, S., and Uchihara, T. (2013). Isoform
transition from four-repeat to three-repeat tau underlies dendrosomatic
and regional progression of neurofibrillary pathology. Acta Neuropathol
125, 565–579.
Hartman, R.E., Izumi, Y., Bales, K.R., Paul, S.M., Wozniak, D.F., and
Holtzman, D.M. (2005). Treatment with an amyloid-β antibody
ameliorates plaque load, learning deficits, and hippocampal long-term
potentiation in a mouse model of Alzheimer’s disease. J Neurosci 25,
6213–6220.
Hashimoto, S., Matsuba, Y., Kamano, N., Mihira, N., Sahara, N., Takano,
J., Muramatsu, S., Saido, T.C., and Saito, T. (2019). Tau binding protein
CAPON induces tau aggregation and neurodegeneration. Nat Commun
10, 2394.
Havas, D., Hutter-Paier, B., Ubhi, K., Rockenstein, E., Crailsheim, K.,
Masliah, E., and Windisch, M. (2011). A longitudinal study of
behavioral deficits in an AβPP transgenic mouse model of
Alzheimer’s disease. J Alzheimer Dis 25, 231–243.
He, A., Zhang, C., Ke, X., Yi, Y., Yu, Q., Zhang, T., Yu, H., Du, H., Li, H.,
Tian, Q., et al. (2022). VGLUT3 neurons in median raphe control the
efficacy of spatial memory retrieval via ETV4 regulation of VGLUT3
transcription. Sci China Life Sci 65, 1590–1607.
Heckmann, B.L., Teubner, B.J.W., Boada-Romero, E., Tummers, B., Guy,
C., Fitzgerald, P., Mayer, U., Carding, S., Zakharenko, S.S., Wileman,
T., et al. (2020). Noncanonical function of an autophagy protein
prevents spontaneous Alzheimer’s disease. Sci Adv 6, eabb9036.
Helboe, L., Egebjerg, J., Barkholt, P., and Volbracht, C. (2017). Early
depletion of CA1 neurons and late neurodegeneration in a mouse
tauopathy model. Brain Res 1665, 22–35.
Herzig, M.C., Winkler, D.T., Burgermeister, P., Pfeifer, M., Kohler, E.,
Schmidt, S.D., Danner, S., Abramowski, D., Stürchler-Pierrat, C.,
Bürki, K., et al. (2004). is targeted to the vasculature in a mouse
model of hereditary cerebral hemorrhage with amyloidosis. Nat
Neurosci 7, 954–960.
Heuer, E., F. Rosen, R., Cintron, A., and C. Walker, L. (2012). Nonhuman
primate models of Alzheimer-like cerebral proteopathy. Curr Pharm
Des 18, 1159–1169.
Holcomb, L., Gordon, M.N., McGowan, E., Yu, X., Benkovic, S., Jantzen,
P., Wright, K., Saad, I., Mueller, R., Morgan, D., 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.
Hole, K.L., Staniaszek, L.E., Menon Balan, G., Mason, J.M., Brown, J.T.,
and Williams, R.J. (2021). Oral (−)-epicatechin inhibits progressive tau
pathology in rTg4510 mice independent of direct actions at GSK3β.
Front Neurosci 15, 697319.
Hong, S., Beja-Glasser, V.F., Nfonoyim, B.M., Frouin, A., Li, S.,
Ramakrishnan, S., Merry, K.M., Shi, Q., Rosenthal, A., Barres, B.A.,
et al. (2016). Complement and microglia mediate early synapse loss in
Alzheimer mouse models. Science 352, 712–716.
Hoover, B.R., Reed, M.N., Su, J., Penrod, R.D., Kotilinek, L.A., Grant, M.
K., Pitstick, R., Carlson, G.A., Lanier, L.M., Yuan, L.L., et al. (2010).
Tau mislocalization to dendritic spines mediates synaptic dysfunction
independently of neurodegeneration. Neuron 68, 1067–1081.
Hou, T.T., Yang, H.Y., Wang, W., Wu, Q.Q., Tian, Y.R., and Jia, J.P. (2018).
Sulforaphane inhibits the generation of amyloid-β oligomer and
promotes spatial learning and memory in Alzheimer’s disease
(PS1V97L) transgenic mice. J Alzheimer Dis 62, 1803–1813.
Howlett, D.R., Bowler, K., Soden, P.E., Riddell, D., Davis, J.B., Richard-
son, J.C., Burbidge, S.A., Gonzalez, M.I., Irving, E.A., Lawman, A., et
al. (2008). Abeta deposition and related pathology in an APP x PS1
transgenic mouse model of Alzheimer’s disease. Histol Histopathol 23,
67–76.
Howlett, D.R., Richardson, J.C., Austin, A., Parsons, A.A., Bate, S.T.,
Davies, D.C., and Gonzalez, M.I. (2004). Cognitive correlates of
deposition in male and female mice bearing amyloid precursor protein
and presenilin-1 mutant transgenes. Brain Res 1017, 130–136.
Hsia, A.Y., Masliah, E., McConlogue, L., Yu, G.Q., Tatsuno, G., Hu, K.,
Kholodenko, D., Malenka, R.C., Nicoll, R.A., and Mucke, L. (1999).
Plaque-independent disruption of neural circuits in Alzheimer’s disease
mouse models. Proc Natl Acad Sci USA 96, 3228–3233.
Hsiao, K., Chapman, P., Nilsen, S., Eckman, C., Harigaya, Y., Younkin, S.,
Yang, F., and Cole, G. (1996). Correlative memory deficits,
elevation, and amyloid plaques in transgenic mice. Science 274, 99–
103.
Hu, X., Das, B., Hou, H., He, W., and Yan, R. (2018). BACE1 deletion in
the adult mouse reverses preformed amyloid deposition and improves
cognitive functions. J Exp Med 215, 927–940.
Huang, K., Marcora, E., Pimenova, A.A., Di Narzo, A.F., Kapoor, M., Jin,
S.C., Harari, O., Bertelsen, S., Fairfax, B.P., Czajkowski, J., et al.
(2017). A common haplotype lowers PU.1 expression in myeloid cells
and delays onset of Alzheimer’s disease. Nat Neurosci 20, 1052–1061.
Hwang, D.Y., Chae, K.R., Kang, T.S., Hwang, J.H., Lim, C.H., Kang, H.K.,
Goo, J.S., Lee, M.R., Lim, H.J., Min, S.H., et al. (2002). Alterations in
behavior, amyloid p-42, caspase-3, and Cox-2 in mutant PS2 transgenic
mouse model of Alzheimer’s disease. FASEB J 16, 805–813.
Hwang, D.Y., Cho, J.S., Lee, S.H., Chae, K.R., Lim, H.J., Min, S.H., Seo,
S.J., Song, Y.S., Song, C.W., Paik, S.G., et al. (2004). Aberrant
expressions of pathogenic phenotype in Alzheimer’s diseased
transgenic mice carrying NSE-controlled APPsw. Exp Neurol 186,
20–32.
Hyman, B., and Tanzi, R.E. (2019). Effects of species-specific genetics on
Alzheimer’s mouse models. Neuron 101, 351–352.
Irizarry, M.C., Mcnamara, M., Fedorchak, K., Hsiao, K., and Hyman, B.T.
(1997). APPSW transgenic mice develop age-related deposits and
neuropil abnormalities, but no neuronal loss in CA1. J Neuropathol Exp
Neurol 56, 965–973.
Jackson, H.M., Soto, I., Graham, L.C., Carter, G.W., and Howell, G.R.
(2013). Clustering of transcriptional profiles identifies changes to
insulin signaling as an early event in a mouse model of Alzheimer’s
disease. BMC Genomics 14, 831.
Jackson, R.J., Rudinskiy, N., Herrmann, A.G., Croft, S., Kim, J.M., Pet-
rova, V., Ramos-Rodriguez, J.J., Pitstick, R., Wegmann, S., Garcia-
Alloza, M., Carlson, G.A., Hyman, B.T., and Spires-Jones, T.L. (2016).
Human tau increases amyloid beta plaque size but not amyloid beta-
mediated synapse loss in a novel mouse model of Alzheimer’s disease.
Eur J Neurosci 44, 3056–3066.
Jacobsen, J.S., Wu, C.C., Redwine, J.M., Comery, T.A., Arias, R., Bowlby,
36 Li, X., et al. Sci China Life Sci
M., Martone, R., Morrison, J.H., Pangalos, M.N., Reinhart, P.H., et al.
(2006). Early-onset behavioral and synaptic deficits in a mouse model
of Alzheimer’s disease. Proc Natl Acad Sci USA 103, 5161–5166.
Jaenisch, R., and Mintz, B. (1974). Simian virus 40 DNA sequences in
DNA of healthy adult mice derived from preimplantation blastocysts
injected with viral DNA. Proc Natl Acad Sci USA 71, 1250–1254.
Jankowsky, J.L., Fadale, D.J., Anderson, J., Xu, G.M., Gonzales, V.,
Jenkins, N.A., Copeland, N.G., Lee, M.K., Younkin, L.H., Wagner, S.
L., et al. (2004). Mutant presenilins specifically elevate the levels of the
42 residue β-amyloid peptide in vivo: evidence for augmentation of a
42-specific γ secretase. Hum Mol Genet 13, 159–170.
Jankowsky, J.L., Slunt, H.H., Gonzales, V., Savonenko, A.V., Wen, J.C.,
Jenkins, N.A., Copeland, N.G., Younkin, L.H., Lester, H.A., Younkin,
S.G., et al. (2005). Persistent amyloidosis following suppression of
Abeta production in a transgenic model of Alzheimer disease. PLoS
Med 2, e355.
Jankowsky, J.L., Slunt, H.H., Ratovitski, T., Jenkins, N.A., Copeland, N.G.,
and Borchelt, D.R. (2001). Co-expression of multiple transgenes in
mouse CNS: a comparison of strategies. Biomol Eng 17, 157–165.
Jawhar, S., Trawicka, A., Jenneckens, C., Bayer, T.A., and Wirths, O.
(2012). Motor deficits, neuron loss, and reduced anxiety coinciding with
axonal degeneration and intraneuronal aggregation in the 5XFAD
mouse model of Alzheimer’s disease. Neurobiol Aging 33, 196.e29–
196.e40.
Jay, T.R., Hirsch, A.M., Broihier, M.L., Miller, C.M., Neilson, L.E.,
Ransohoff, R.M., Lamb, B.T., and Landreth, G.E. (2017). Disease
progression-dependent effects of TREM2 deficiency in a mouse model
of Alzheimer’s disease. J Neurosci 37, 637–647.
Jia, J., Xu, E., Shao, Y., Jia, J., Sun, Y., and Li, D. (2005). One novel
presenilin-1 gene mutation in a Chinese pedigree of familial
Alzheimer’s disease. J Alzheimer Dis 7, 119–124. ; discussion 173-180.
Jia, L., Du, Y., Chu, L., Zhang, Z., Li, F., Lyu, D., Li, Y., Li, Y., Zhu, M.,
Jiao, H., et al. (2020a). Prevalence, risk factors, and management of
dementia and mild cognitive impairment in adults aged 60 years or
older in China: a cross-sectional study. Lancet Public Health 5, e661–
e671.
Jia, L., Fu, Y., Shen, L., Zhang, H., Zhu, M., Qiu, Q., Wang, Q., Yan, X.,
Kong, C., Hao, J., et al. (2020b). PSEN1,PSEN2, and APP mutations in
404 Chinese pedigrees with familial Alzheimer’s disease. Alzheimers
Dement 16, 178–191.
Jia, L., Quan, M., Fu, Y., Zhao, T., Li, Y., Wei, C., Tang, Y., Qin, Q., Wang,
F., Qiao, Y., et al. (2020c). Dementia in China: epidemiology, clinical
management, and research advances. Lancet Neurol 19, 81–92.
Jiao, H., and Jia, J. (2022). Ginsenoside compound K acts via LRP1 to
alleviate Amyloid β42-induced neuroinflammation in microglia by
suppressing NF-κB. Biochem Biophys Res Commun 590, 14–19.
Josephine Boder, E., and Banerjee, I.A. (2021). Alzheimer’s disease:
current perspectives and advances in physiological modeling.
Bioengineering 8, 211.
Kamphuis, W., Mamber, C., Moeton, M., Kooijman, L., Sluijs, J.A., Jansen,
A.H., Verveer, M., de Groot, L.R., Smith, V.D., Rangarajan, S., et al.
(2012). GFAP isoforms in adult mouse brain with a focus on neurogenic
astrocytes and reactive astrogliosis in mouse models of Alzheimer
disease. PLoS ONE 7, e42823.
Kang, S., Kim, J., and Chang, K.A. (2021). Spatial memory deficiency
early in 6xTg Alzheimer’s disease mouse model. Sci Rep 11, 1334.
Karran, E., and De Strooper, B. (2022). The amyloid hypothesis in
Alzheimer disease: new insights from new therapeutics. Nat Rev Drug
Discov 21, 306–318.
Karran, E., and Hardy, J. (2014). A critique of the drug discovery and phase
3 clinical programs targeting the amyloid hypothesis for Alzheimer
disease. Ann Neurol 76, 185–205.
Kawasumi, M., Chiba, T., Yamada, M., Miyamae-Kaneko, M., Matsuoka,
M., Nakahara, J., Tomita, T., Iwatsubo, T., Kato, S., Aiso, S., et al.
(2004). Targeted introduction of V642I mutation in amyloid precursor
protein gene causes functional abnormality resembling early stage of
Alzheimer’s disease in aged mice. Eur J Neurosci 19, 2826–2838.
Kelly, P.H., Bondolfi, L., Hunziker, D., Schlecht, H.P., Carver, K., Maguire,
E., Abramowski, D., Wiederhold, K.H., Sturchler-Pierrat, C., Jucker,
M., et al. (2003). Progressive age-related impairment of cognitive
behavior in APP23 transgenic mice. Neurobiol Aging 24, 365–378.
Keren-Shaul, H., Spinrad, A., Weiner, A., Matcovitch-Natan, O., Dvir-
Szternfeld, R., Ulland, T.K., David, E., Baruch, K., Lara-Astaiso, D.,
Toth, B., et al. (2017). A unique microglia type associated with
restricting development of Alzheimer’s disease. Cell 169, 1276–1290.
e17.
Kim, J., and Jeong, Y. (2015). In vivo image of cerebral amyloid angiopathy
in an Alzheimer’s disease mouse model. J Stroke 17, 87–88.
Kim, J.H., Nam, Y.P., Jeon, S.M., Han, H.S., and Suk, K. (2012). Amyloid
neurotoxicity is attenuated by metallothionein: dual mechanisms at
work. J Neurochem 121, 751–762.
Kimura, R., MacTavish, D., Yang, J., Westaway, D., and Jhamandas, J.H.
(2012). Beta amyloid-induced depression of hippocampal long-term
potentiation is mediated through the amylin receptor. J Neurosci 32,
17401–17406.
Kimura, R., and Ohno, M. (2009). Impairments in remote memory
stabilization precede hippocampal synaptic and cognitive failures in
5XFAD Alzheimer mouse model. Neurobiol Dis 33, 229–235.
King, A. (2018). The search for better animal models of Alzheimer’s
disease. Nature 559, S13–S15.
Klyubin, I., Walsh, D.M., Lemere, C.A., Cullen, W.K., Shankar, G.M.,
Betts, V., Spooner, E.T., Jiang, L., Anwyl, R., Selkoe, D.J., et al. (2005).
Amyloid β protein immunotherapy neutralizes oligomers that
disrupt synaptic plasticity in vivo.Nat Med 11, 556–561.
Knobloch, M., Farinelli, M., Konietzko, U., Nitsch, R.M., and Mansuy, I.
M. (2007a). oligomer-mediated long-term potentiation impairment
involves protein phosphatase 1-dependent mechanisms. J Neurosci 27,
7648–7653.
Knobloch, M., Konietzko, U., Krebs, D.C., and Nitsch, R.M. (2007b).
Intracellular and cognitive deficits precede β-amyloid deposition in
transgenic arcAβ mice. Neurobiol Aging 28, 1297–1306.
Kopeikina, K.J., Polydoro, M., Tai, H.C., Yaeger, E., Carlson, G.A.,
Pitstick, R., Hyman, B.T., and Spires-Jones, T.L. (2013). Synaptic
alterations in the rTg4510 mouse model of tauopathy. J Comp Neurol
521, 1334–1353.
Kulnane, L.S., and Lamb, B.T. (2001). Neuropathological characterization
of mutant amyloid precursor protein yeast artificial chromosome
transgenic mice. Neurobiol Dis 8, 982–992.
Kurt, M.A., Davies, D.C., Kidd, M., Duff, K., and Howlett, D.R. (2003).
Hyperphosphorylated tau and paired helical filament-like structures in
the brains of mice carrying mutant amyloid precursor protein and
mutant presenilin-1 transgenes. Neurobiol Dis 14, 89–97.
Kwan, P., Ho, A., and Baum, L. (2022). Effects of deferasirox in
Alzheimer’s disease and tauopathy animal models. Biomolecules 12,
365.
Lalonde, R., Kim, H.D., Maxwell, J.A., and Fukuchi, K. (2005).
Exploratory activity and spatial learning in 12-month-old
APP695SWE/co+PS1/ΔE9 mice with amyloid plaques. Neurosci Lett
390, 87–92.
Lalonde, R., and Strazielle, C. (2005). PS1 knockin mice with the Japanese
I213T mutation: effects on exploratory activity, motor coordination, and
spatial learning. Behav Brain Res 162, 182–190.
Lamb, B.A., Bardel, K.A., Kulnane, L.S., Anderson, J.J., Holtz, G.,
Wagner, S.L., Sisodia, S.S., and Hoeger, E.J. (1999). Amyloid
production and deposition in mutant amyloid precursor protein and
presenilin-1 yeast artificial chromosome transgenic mice. Nat Neurosci
2, 695–697.
Lamb, B.T., Call, L.M., Slunt, H.H., Bardel, K.A., Lawler, A.M., Eckman,
C.B., Younkin, S.G., Holtz, G., Wagner, S.L., Price, D.L., et al. (1997).
Altered metabolism of familial Alzheimer’s disease-linked amyloid
precursor protein variants in yeast artificial chromosome transgenic
mice. Hum Mol Genet 6, 1535–1541.
Lambert, M.P., Velasco, P.T., Chang, L., Viola, K.L., Fernandez, S., Lacor,
P.N., Khuon, D., Gong, Y., Bigio, E.H., Shaw, P., et al. (2007).
37
Li, X., et al. Sci China Life Sci
Monoclonal antibodies that target pathological assemblies of Aβ. J
Neurochem 100, 23–35.
Lanz, T.A., Carter, D.B., and Merchant, K.M. (2003). Dendritic spine loss
in the hippocampus of young PDAPP and Tg2576 mice and its
prevention by the ApoE2 genotype. Neurobiol Dis 13, 246–253.
Larson, J., Lynch, G., Games, D., and Seubert, P. (1999). Alterations in
synaptic transmission and long-term potentiation in hippocampal slices
from young and aged PDAPP mice. Brain Res 840, 23–35.
Lasagna-Reeves, C.A., de Haro, M., Hao, S., Park, J., Rousseaux, M.W.C.,
Al-Ramahi, I., Jafar-Nejad, P., Vilanova-Velez, L., See, L., De Maio, A.,
et al. (2016). Reduction of Nuak1 decreases tau and reverses
phenotypes in a tauopathy mouse model. Neuron 92, 407–418.
Lee, C.Y.D., Daggett, A., Gu, X., Jiang, L.L., Langfelder, P., Li, X., Wang,
N., Zhao, Y., Park, C.S., Cooper, Y., et al. (2018). Elevated TREM2
gene dosage reprograms microglia responsivity and ameliorates
pathological phenotypes in Alzheimer’s disease models. Neuron 97,
1032–1048.e5.
Lee, H.J., Jeon, S.G., Kim, J., Kang, R.J., Kim, S.M., Han, K.M., Park, H.,
Kim, K.T., Sung, Y.M., Nam, H.Y., et al. (2021a). Ibrutinib modulates
Aβ/tau pathology, neuroinflammation, and cognitive function in mouse
models of Alzheimer’s disease. Aging Cell 20, e13332.
Lee, H.S., Kim, M.W., Jin, K.S., Shin, H.C., Kim, W.K., Lee, S.C., Kim, S.
J., Lee, E.W., and Ku, B. (2021b). Molecular analysis of the interaction
between human PTPN21 and the oncoprotein E7 from human
papillomavirus genotype 18. MolCells 44, 26–37.
Lefterov, I., Fitz, N.F., Cronican, A., Lefterov, P., Staufenbiel, M., and
Koldamova, R. (2009). Memory deficits in APP23/Abca1+/− mice
correlate with the level of Aβoligomers. ASN Neuro 1, AN20090015.
Lehman, E.J.H., Kulnane, L.S., and Lamb, B.T. (2003). Alterations in β-
amyloid production and deposition in brain regions of two transgenic
models. Neurobiol Aging 24, 645–653.
Leinenga, G., Koh, W.K., and Götz, J. (2021). A comparative study of the
effects of Aducanumab and scanning ultrasound on amyloid plaques
and behavior in the APP23 mouse model of Alzheimer disease.
Alzheimers Res Ther 13, 76.
Leon, W.C., Canneva, F., Partridge, V., Allard, S., Ferretti, M.T., DeWilde,
A., Vercauteren, F., Atifeh, R., Ducatenzeiler, A., Klein, W., et al.
(2010). A novel transgenic rat model with a full Alzheimer’s-like
amyloid pathology displays pre-plaque intracellular amyloid-β-
associated cognitive impairment. J Alzheimer Dis 20, 113–126.
Lewis, J., Dickson, D.W., Lin, W.L., Chisholm, L., Corral, A., Jones, G.,
Yen, S.H., Sahara, N., Skipper, L., Yager, D., et al. (2001). Enhanced
neurofibrillary degeneration in transgenic mice expressing mutant tau
and APP. Science 293, 1487–1491.
Lewis, J., McGowan, E., Rockwood, J., Melrose, H., Nacharaju, P., Van
Slegtenhorst, M., Gwinn-Hardy, K., Murphy, M.P., Baker, M., Yu, X., et
al. (2000). Neurofibrillary tangles, amyotrophy and progressive motor
disturbance in mice expressing mutant (P301L) tau protein. Nat Genet
25, 402–405.
Li, B., Xu, L., Li, F., Li, Y., Zhao, Y., Zhang, H., Quan, M., and Jia, J.
(2022). CaMKIIα signaling is required for the neuroprotective effects of
Dl-3-n-butylphthalide in Alzheimer’s disease. Mol Neurobiol 59, 3370–
3381.
Li, D., Qiu, Z., Shao, Y., Chen, Y., Guan, Y., Liu, M., Li, Y., Gao, N., Wang,
L., Lu, X., et al. (2013). Heritable gene targeting in the mouse and rat
using a CRISPR-Cas system. Nat Biotechnol 31, 681–683.
Li, F., Wu, X., Li, J., and Niu, Q. (2016). Ginsenoside Rg1 ameliorates
hippocampal long-term potentiation and memory in an Alzheimer’s
disease model. Mol Med Rep 13, 4904–4910.
Li, H., Jia, J., Wang, W., Hou, T., Tian,, Y., Wu, Q., Xu, L., Wei, Y., and
Wang, X. (2018). Honokiol alleviates cognitive deficits of Alzheimer’s
disease (PS1V97L) transgenic mice by activating mitochondrial SIRT3.
J Alzheimer Dis 64, 291–302.
Li, H.Q., Ip, S.P., Yuan, Q.J., Zheng, G.Q., Tsim, K.K.W., Dong, T.T.X.,
Lin, G., Han, Y., Liu, Y., Xian, Y.F., et al. (2019). Isorhynchophylline
ameliorates cognitive impairment via modulating amyloid pathology,
tau hyperphosphorylation and neuroinflammation: Studies in a
transgenic mouse model of Alzheimer’s disease. Brain Behav Immun
82, 264–278.
Li, S., Wu, Z., and Le, W. (2021a). Traditional Chinese medicine for
dementia. Alzheimers Dement 17, 1066–1071.
Li, T., Martin, E., Abada, Y., Boucher, C., Cès, A., Youssef, I., Fenaux, G.,
Forand, Y., Legrand, A., Nachiket, N., et al. (2020). Effects of chronic
masitinib treatment in APPswe/PSEN1dE9 transgenic mice modeling
Alzheimer’s disease. J Alzheimer Dis 76, 1339–1345.
Li, W., Pang, Y., Wang, Y., Mei, F., Guo, M., Wei, Y., Li, X., Qin, W.,
Wang, W., Jia, L., et al. (2023). Aberrant palmitoylation caused by a
ZDHHC21 mutation contributes to pathophysiology of Alzheimer's
disease. BMC Med 21, 223.
Li, W., Wang, S., Zhang, H., Li, B., Xu, L., Li, Y., Kong, C., Jiao, H.,
Wang, Y., Pang, Y., et al. (2021b). Honokiol restores microglial pha-
gocytosis by reversing metabolic reprogramming. J Alzheimers Dis 82,
1475–1485.
Lillehaug, S., Syverstad, G.H., Nilsson, L.N.G., Bjaalie, J.G., Leergaard, T.
B., and Torp, R. (2014). Brainwide distribution and variance of
amyloid-beta deposits in tg-ArcSwe mice. Neurobiol Aging 35, 556–
564.
Liu, J., Baum, L., Yu, S., Lin, Y., Xiong, G., Chang, R.C.C., So, K.F., and
Chiu, K. (2021a). Preservation of retinal function through synaptic
stabilization in Alzheimer’s disease model mouse retina by lycium
barbarum extracts. Front Aging Neurosci 13, 788798.
Liu, P., Paulson, J.B., Forster, C.L., Shapiro, S.L., Ashe, K.H., and Zahs, K.
R. (2015). Characterization of a novel mouse model of Alzheimer’s
disease—amyloid pathology and unique β-amyloid oligomer profile.
PLoS ONE 10, e0126317.
Liu, W., Li, Y., Li, Y., Xu, L., and Jia, J. (2023). Carnosic acid attenuates
AβOs-induced apoptosis and synaptic impairment via regulating
NMDAR2B and its downstream cascades in SH-SY5Y cells. Mol
Neurobiol 60, 133–144.
Liu, X., Zhou, Q., Zhang, J.H., Wang, K.Y., Saito, T., Saido, T.C., Wang,
X., Gao, X., and Azuma, K. (2021b). Microglia-based sex-biased
neuropathology in early-stage Alzheimer’s disease model mice and the
potential pharmacologic efficacy of dioscin. Cells 10, 3261.
Liu, Y., Yao, J., Song, Z., Guo, W., Sun, B., Wei, J., Estillore, J.P., Back, T.
G., and Chen, S.R.W. (2021c). Limiting RyR2 open time prevents
Alzheimer’s disease-related deficits in the 3xTG-AD mouse model. J
Neurosci Res 99, 2906–2921.
Lo, A.C., Tesseur, I., Scopes, D.I.C., Nerou, E., Callaerts-Vegh, Z.,
Vermaercke, B., Treherne, J.M., De Strooper, B., and D’Hooge, R.
(2013). Dose-dependent improvements in learning and memory deficits
in APPPS1-21 transgenic mice treated with the orally active toxicity
inhibitor SEN1500. Neuropharmacology 75, 458–466.
Long, J.M., and Holtzman, D.M. (2019). Alzheimer disease: an update on
pathobiology and treatment strategies. Cell 179, 312–339.
Lord, A., Englund, H., Söderberg, L., Tucker, S., Clausen, F., Hillered, L.,
Gordon, M., Morgan, D., Lannfelt, L., Pettersson, F.E., et al. (2009).
Amyloid-β protofibril levels correlate with spatial learning in Arctic
Alzheimer’s disease transgenic mice. FEBS J 276, 995–1006.
Lord, A., Kalimo, H., Eckman, C., Zhang, X.Q., Lannfelt, L., and Nilsson,
L.N.G. (2006). The Arctic Alzheimer mutation facilitates early
intraneuronal aggregation and senile plaque formation in
transgenic mice. Neurobiol Aging 27, 67–77.
Lord, A., Philipson, O., Klingstedt, T., Westermark, G., Hammarström, P.,
Nilsson, K.P.R., and Nilsson, L.N.G. (2011). Observations in APP
bitransgenic mice suggest that diffuse and compact plaques form via
independent processes in Alzheimer’s disease. Am J Pathol 178, 2286–
2298.
Lyu, D., and Jia, J. (2022). Cryptotanshinone attenuates amyloid-β42-
induced tau phosphorylation by regulating PI3K/Akt/GSK3β pathway
in HT22 cells. Mol Neurobiol 59, 4488–4500.
Maeda, S., Djukic, B., Taneja, P., Yu, G., Lo, I., Davis, A., Craft, R., Guo,
W., Wang, X., Kim, D., et al. (2016). Expression of A152T human tau
causes age-dependent neuronal dysfunction and loss in transgenic mice.
EMBO Rep 17, 530–551.
38 Li, X., et al. Sci China Life Sci
Malm, T.M., Iivonen, H., Goldsteins, G., Keksa-Goldsteine, V., Ahtoniemi,
T., Kanninen, K., Salminen, A., Auriola, S., Van Groen, T., Tanila, H., et
al. (2007). Pyrrolidine dithiocarbamate activates Akt and improves
spatial learning in APP/PS1 mice without affecting β-amyloid burden. J
Neurosci 27, 3712–3721.
Marazuela, P., Paez-Montserrat, B., Bonaterra-Pastra, A., Solé, M., and
Hernández-Guillamon, M. (2022). Impact of cerebral amyloid
angiopathy in two transgenic mouse models of cerebral β-
amyloidosis: a neuropathological study. Int J Mol Sci 23, 4972.
Martini, A.C., Forner, S., Trujillo-Estrada, L., Baglietto-Vargas, D., and
LaFerla, F.M. (2018). Past to future: what animal models have taught us
about Alzheimer’s disease. J Alzheimers Dis 64, S365–S378.
Masliah, E., Sisk, A., Mallory, M., and Games, D. (2001). Neurofibrillary
pathology in transgenic mice overexpressing V717F β-amyloid
precursor protein. J Neuropathol Exp Neurol 60, 357–368.
Maurin, H., Chong, S.A., Kraev, I., Davies, H., Kremer, A., Seymour, C.
M., Lechat, B., Jaworski, T., Borghgraef, P., Devijver, H., et al. (2014).
Early structural and functional defects in synapses and myelinated ax-
ons in stratum lacunosum moleculare in two preclinical models for
tauopathy. PLoS ONE 9, e87605.
McGowan, E., Sanders, S., Iwatsubo, T., Takeuchi, A., Saido, T., Zehr, C.,
Yu, X., Uljon, S., Wang, R., Mann, D., et al. (1999). Amyloid phenotype
characterization of transgenic mice overexpressing both mutant amyloid
precursor protein and mutant presenilin 1 transgenes. Neurobiol Dis 6,
231–244.
Mckean, N.E., Handley, R.R., and Snell, R.G. (2021). A review of the
current mammalian models of Alzheimer’s disease and challenges that
need to be overcome. Int J Mol Sci 22, 13168.
Mehla, J., Lacoursiere, S.G., Lapointe, V., McNaughton, B.L., Sutherland,
R.J., McDonald, R.J., and Mohajerani, M.H. (2019). Age-dependent
behavioral and biochemical characterization of single APP knock-in
mouse (APPNL-G-F/NL-G-F) model of Alzheimer’s disease. Neurobiol
Aging 75, 25–37.
Mei, Z., Zhang, F., Tao, L., Zheng, W., Cao, Y., Wang, Z., Tang, S., Le, K.,
Chen, S., Pi, R., et al. (2009). Cryptotanshinone, a compound from
Salvia miltiorrhiza modulates amyloid precursor protein metabolism
and attenuates β-amyloid deposition through upregulating α-secretase in
vivo and in vitro.Neurosci Lett 452, 90–95.
Meilandt, W.J., Maloney, J.A., Imperio, J., Lalehzadeh, G., Earr, T.,
Crowell, S., Bainbridge, T.W., Lu, Y., Ernst, J.A., Fuji, R.N., et al.
(2019). Characterization of the selective in vitro and in vivo binding
properties of crenezumab to oligomeric Aβ. Alzheimers Res Ther 11,
97.
Meilandt, W.J., Ngu, H., Gogineni, A., Lalehzadeh, G., Lee, S.H.,
Srinivasan, K., Imperio, J., Wu, T., Weber, M., Kruse, A.J., et al.
(2020). Trem2 deletion reduces late-stage amyloid plaque
accumulation, elevates the Aβ42:Aβ40 ratio, and exacerbates axonal
dystrophy and dendritic spine loss in the PS2APP Alzheimer’s mouse
model. J Neurosci 40, 1956–1974.
Miao, J., Vitek, M.P., Xu, F., Previti, M.L., Davis, J., and Van Nostrand, W.
E. (2005a). Reducing cerebral microvascular amyloid-β protein
deposition diminishes regional neuroinflammation in vasculotropic
mutant amyloid precursor protein transgenic mice. J Neurosci 25, 6271–
6277.
Miao, J., Xu, F., Davis, J., Otte-Höller, I., Verbeek, M.M., and Van
Nostrand, W.E. (2005b). Cerebral microvascular amyloid β protein
deposition induces vascular degeneration and neuroinflammation in
transgenic mice expressing human vasculotropic mutant amyloid β
precursor protein. Am J Pathol 167, 505–515.
Migliore, L., and Coppedè, F. (2022). Gene-environment interactions in
Alzheimer disease: the emerging role of epigenetics. Nat Rev Neurol
18, 643–660.
Minkeviciene, R., Ihalainen, J., Malm, T., Matilainen, O., Keksa-
Goldsteine, V., Goldsteins, G., Iivonen, H., Leguit, N., Glennon, J.,
Koistinaho, J., et al. (2008). Age-related decrease in stimulated
glutamate release and vesicular glutamate transporters in APP/PS1
transgenic and wild-type mice. J Neurochem 105, 584–594.
Mocanu, M.M., Nissen, A., Eckermann, K., Khlistunova, I., Biernat, J.,
Drexler, D., Petrova, O., Schönig, K., Bujard, H., Mandelkow, E., et al.
(2008). The potential for β-structure in the repeat domain of tau protein
determines aggregation, synaptic decay, neuronal loss, and coassembly
with endogenous tau in inducible mouse models of tauopathy. J
Neurosci 28, 737–748.
Moechars, D., Dewachter, I., Lorent, K., Reversé, D., Baekelandt, V.,
Naidu, A., Tesseur, I., Spittaels, K., Haute, C.V.D., Checler, F., et al.
(1999). Early phenotypic changes in transgenic mice that overexpress
different mutants of amyloid precursor protein in brain. J Biol Chem
274, 6483–6492.
Mucke, L., Masliah, E., Yu, G.Q., Mallory, M., Rockenstein, E.M.,
Tatsuno, G., Hu, K., Kholodenko, D., Johnson-Wood, K., and
McConlogue, L. (2000). High-level neuronal expression of 1–42 in
wild-type human amyloid protein precursor transgenic mice:
synaptotoxicity without plaque formation. J Neurosci 20, 4050–4058.
Mullan, M., Crawford, F., Axelman, K., Houlden, H., Lilius, L., Winblad,
B., and Lannfelt, L. (1992). A pathogenic mutation for probable
Alzheimer’s disease in the APP gene at the N-terminus of β-amyloid.
Nat Genet 1, 345–347.
Mullane, K., and Williams, M. (2019). Preclinical models of Alzheimer’s
disease: relevance and translational validity. Curr Protoc Pharmacol 84,
e57.
Müller-Schiffmann, A., Herring, A., Abdel-Hafiz, L., Chepkova, A.N.,
Schäble, S., Wedel, D., Horn, A.H.C., Sticht, H., de Souza Silva, M.A.,
Gottmann, K., et al. (2016). Amyloid-β dimers in the absence of plaque
pathology impair learning and synaptic plasticity. Brain 139, 509–525.
Murata, N., Murakami, K., Ozawa, Y., Kinoshita, N., Irie, K., Shirasawa,
T., and Shimizu, T. (2010). Silymarin attenuated the amyloid β plaque
burden and improved behavioral abnormalities in an Alzheimer’s
disease mouse model. Biosci Biotechnol Biochem 74, 2299–2306.
Nagakura, A., Shitaka, Y., Yarimizu, J., and Matsuoka, N. (2013).
Characterization of cognitive deficits in a transgenic mouse model of
Alzheimer’s disease and effects of donepezil and memantine. Eur J
Pharmacol 703, 53–61.
Nakano, Y., Kondoh, G., Kudo, T., Imaizumi, K., Kato, M., Miyazaki, J.,
Tohyama, M., Takeda, J., and Takeda, M. (1999). Accumulation of
murine amyloidbeta42 in a gene-dosage-dependent manner in PS1
‘knock-in’ mice. Eur J Neurosci 11, 2577–2581.
Nedelec, T., Couvy-Duchesne, B., Monnet, F., Daly, T., Ansart, M.,
Gantzer, L., Lekens, B., Epelbaum, S., Dufouil, C., and Durrleman, S.
(2022). Identifying health conditions associated with Alzheimer’s
disease up to 15 years before diagnosis: an agnostic study of French
and British health records. Lancet Digital Health 4, e169–e178.
Nilsson, P., Saito, T., and Saido, T.C. (2014). New mouse model of
Alzheimer’s. ACS Chem Neurosci 5, 499–502.
Nott, A., Holtman, I.R., Coufal, N.G., Schlachetzki, J.C.M., Yu, M., Hu, R.,
Han, C.Z., Pena, M., Xiao, J., Wu, Y., et al. (2019). Brain cell type-
specific enhancer-promoter interactome maps and disease-risk
association. Science 366, 1134–1139.
O’Leary, T.P., Robertson, A., Chipman, P.H., Rafuse, V.F., and Brown, R.E.
(2018). Motor function deficits in the 12 month-old female 5xFAD
mouse model of Alzheimer’s disease. Behav Brain Res 337, 256–263.
O’Leary, T.P., Shin, S., Fertan, E., Dingle, R.N., Almuklass, A., Gunn, R.
K., Yu, Z., Wang, J., and Brown, R.E. (2017). Reduced acoustic startle
response and peripheral hearing loss in the 5xFAD mouse model of
Alzheimer’s disease. Genes Brain Behav 16, 554–563.
Oakley, H., Cole, S.L., Logan, S., Maus, E., Shao, P., Craft, J., Guillozet-
Bongaarts, A., Ohno, M., Disterhoft, J., Van Eldik, L., et al. (2006).
Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron
loss in transgenic mice with five familial Alzheimer’s disease
mutations: potential factors in amyloid plaque formation. J Neurosci
26, 10129–10140.
Oddo, S., Caccamo, A., Shepherd, J.D., Murphy, M.P., Golde, T.E., Kayed,
R., Metherate, R., Mattson, M.P., Akbari, Y., and LaFerla, F.M. (2003).
Triple-transgenic model of Alzheimer’s disease with plaques and
tangles. Neuron 39, 409–421.
39
Li, X., et al. Sci China Life Sci
Oddo, S., Caccamo, A., Tran, L., Lambert, M.P., Glabe, C.G., Klein, W.L.,
and LaFerla, F.M. (2006). Temporal profile of amyloid-β (Aβ)
oligomerization in an in vivo model of Alzheimer disease. J Biol
Chem 281, 1599–1604.
Ohno, M., Chang, L., Tseng, W., Oakley, H., Citron, M., Klein, W.L.,
Vassar, R., and Disterhoft, J.F. (2006). Temporal memory deficits in
Alzheimer’s mouse models: rescue by genetic deletion of BACE1. Eur J
Neurosci 23, 251–260.
Onos, K.D., Uyar, A., Keezer, K.J., Jackson, H.M., Preuss, C., Acklin, C.J.,
O’Rourke, R., Buchanan, R., Cossette, T.L., Sukoff Rizzo, S.J., et al.
(2019). Enhancing face validity of mouse models of Alzheimer’s dis-
ease with natural genetic variation. PLoS Genet 15, e1008155.
Ortí-Casañ, N., Zuhorn, I.S., Naudé, P.J.W., De Deyn, P.P., van Schaik, P.E.
M., Wajant, H., and Eisel, U.L.M. (2022). A TNF receptor 2 agonist
ameliorates neuropathology and improves cognition in an Alzheimer’s
disease mouse model. Proc Natl Acad Sci USA 119, e2201137119.
Overk, C.R., and Masliah, E. (2014). Toward a unified therapeutics
approach targeting putative amyloid-β oligomer receptors. Proc Natl
Acad Sci USA 111, 13680–13681.
Ozmen, L., Albientz, A., Czech, C., and Jacobsen, H. (2009). Expression of
transgenic APP mRNA is the key determinant for beta-amyloid
deposition in PS2APP transgenic mice. Neurodegener Dis 6, 29–36.
Pang, K., Jiang, R., Zhang, W., Yang, Z., Li, L.L., Shimozawa, M.,
Tambaro, S., Mayer, J., Zhang, B., Li, M., et al. (2022). An App knock-
in rat model for Alzheimer’s disease exhibiting and tau pathologies,
neuronal death and cognitive impairments. Cell Res 32, 157–175.
Pauls, E., Bayod, S., Mateo, L., Alcalde, V., Juan-Blanco, T., Sánchez-Soto,
M., Saido, T.C., Saito, T., Berrenguer-Llergo, A., Attolini, C.S.O., et al.
(2021). Identification and drug-induced reversion of molecular
signatures of Alzheimer’s disease onset and progression in AppNL-G-F,
AppNL-F, and 3xTg-AD mouse models. Genome Med 13, 168.
Pedram, A., Razandi, M., Deschenes, R.J., and Levin, E.R. (2012). DHHC-
7 and -21 are palmitoylacyltransferases for sex steroid receptors. Mol
Biol Cell 23, 188–199.
Peng, Y., Sun, J., Hon, S., Nylander, A.N., Xia, W., Feng, Y., Wang, X., and
Lemere, C.A. (2010). L-3-n-butylphthalide improves cognitive
impairment and reduces amyloid-beta in a transgenic model of
Alzheimer’s disease. J Neurosci 30, 8180–8189.
Pensalfini, A., Kim, S., Subbanna, S., Bleiwas, C., Goulbourne, C.N.,
Stavrides, P.H., Jiang, Y., Lee, J.H., Darji, S., Pawlik, M., et al. (2020).
Endosomal dysfunction induced by directly overactivating Rab5
recapitulates prodromal and neurodegenerative features of
Alzheimer’s disease. Cell Rep 33, 108420.
Perez, S.E., Raghanti, M.A., Hof, P.R., Kramer, L., Ikonomovic, M.D.,
Lacor, P.N., Erwin, J.M., Sherwood, C.C., and Mufson, E.J. (2013).
Alzheimer’s disease pathology in the neocortex and hippocampus of the
western lowland gorilla (Gorilla gorilla gorilla). J Comp Neurol 521,
4318–4338.
Peroutka, S.J. (2022). Defining demographic cohorts in clinical trial
populations using large electronic health records databases. Contemp
Clin Trials 121, 106890.
Petrushina, I., Hovakimyan, A., Harahap-Carrillo, I.S., Davtyan, H.,
Antonyan, T., Chailyan, G., Kazarian, K., Antonenko, M., Jullienne,
A., Hamer, M.M., et al. (2020). Characterization and preclinical
evaluation of the cGMP grade DNA based vaccine, AV-1959D to enter
the first-in-human clinical trials. Neurobiol Dis 139, 104823.
Philipson, O., Hammarström, P., Nilsson, K.P.R., Portelius, E., Olofsson,
T., Ingelsson, M., Hyman, B.T., Blennow, K., Lannfelt, L., Kalimo, H.,
et al. (2009). A highly insoluble state of similar to that of
Alzheimer’s disease brain is found in Arctic APP transgenic mice.
Neurobiol Aging 30, 1393–1405.
Platt, B., Drever, B., Koss, D., Stoppelkamp, S., Jyoti, A., Plano, A., Utan,
A., Merrick, G., Ryan, D., Melis, V., et al. (2011). Abnormal cognition,
sleep, EEG and brain metabolism in a novel knock-in Alzheimer mouse,
PLB1. PLoS ONE 6, e27068.
Plucińska, K., Crouch, B., Koss, D., Robinson, L., Siebrecht, M., Riedel,
G., and Platt, B. (2014). Knock-in of human BACE1 cleaves murine
APP and reiterates Alzheimer-like phenotypes. J Neurosci 34, 10710–
10728.
Poirier, R., Veltman, I., Pflimlin, M.C., Knoflach, F., and Metzger, F.
(2010). Enhanced dentate gyrus synaptic plasticity but reduced
neurogenesis in a mouse model of amyloidosis. Neurobiol Dis 40,
386–393.
Price, K.A., Varghese, M., Sowa, A., Yuk, F., Brautigam, H., Ehrlich, M.E.,
and Dickstein, D.L. (2014). Altered synaptic structure in the
hippocampus in a mouse model of Alzheimer’s disease with soluble
amyloid-β oligomers and no plaque pathology. Mol Neurodegener 9,
41.
Prüßing, K., Voigt, A., and Schulz, J.B. (2013). Drosophila melanogaster as
a model organism for Alzheimer’s disease. Mol Neurodegener 8, 35.
Puig, B., Gómez-Isla, T., Ribé, E., Cuadrado, M., Torrejón-Escribano, B.,
Dalfó, E., and Ferrer, I. (2004). Expression of stress-activated kinases c-
Jun N-terminal kinase (SAPK/JNK-P) and p38 kinase (p38-P), and tau
hyperphosphorylation in neurites surrounding βA plaques in APP
Tg2576 mice. Neuropathol Appl Neurobiol 30, 491–502.
Puoliväli, J., Wang, J., Heikkinen, T., Heikkilä, M., Tapiola, T., van Groen,
T., and Tanila, H. (2002). Hippocampal Aβ42 levels correlate with
spatial memory deficit in APP and PS1 double transgenic mice.
Neurobiol Dis 9, 339–347.
Qiu, Q., Jia, L., Wang, Q., Zhao, L., Jin, H., Li, T., Quan, M., Xu, L., Li, B.,
Li, Y., et al. (2020). Identification of a novel PSEN1 Gly111Val
missense mutation in a Chinese pedigree with early-onset Alzheimer’s
disease. Neurobiol Aging 85, 155.e1–155.e4.
Qu, C., Li, Q.P., Su, Z.R., Ip, S.P., Yuan, Q.J., Xie, Y.L., Xu, Q.Q., Yang,
W., Huang, Y.F., Xian, Y.F., et al. (2022). Nano-Honokiol ameliorates
the cognitive deficits in TgCRND8 mice of Alzheimer’s disease via
inhibiting neuropathology and modulating gut microbiota. J Adv Res
35, 231–243.
Quan, M., Zhao, T., Tang, Y., Luo, P., Wang, W., Qin, Q., Li, T., Wang, Q.,
Fang, J., and Jia, J. (2020). Effects of gene mutation and disease
progression on representative neural circuits in familial Alzheimer’s
disease. Alzheimers Res Ther 12, 14.
Radde, R., Bolmont, T., Kaeser, S.A., Coomaraswamy, J., Lindau, D.,
Stoltze, L., Calhoun, M.E., Jäggi, F., Wolburg, H., Gengler, S., et al.
(2006). Aβ42-driven cerebral amyloidosis in transgenic mice reveals
early and robust pathology. EMBO Rep 7, 940–946.
Ramsden, M., Kotilinek, L., Forster, C., Paulson, J., McGowan, E.,
SantaCruz, K., Guimaraes, A., Yue, M., Lewis, J., Carlson, G., et al.
(2005). Age-dependent neurofibrillary tangle formation, neuron loss,
and memory impairment in a mouse model of human tauopathy
(P301L). J Neurosci 25, 10637–10647.
Rao, C.V., and Yamada, H.Y. (2021). How would preclinical Alzheimer’s
disease (AD pathology) occur? An insight from a genomic instability
mouse model. Neural Regen Res 16, 2012–2014.
Reaume, A.G., Howland, D.S., Trusko, S.P., Savage, M.J., Lang, D.M.,
Greenberg, B.D., Siman, R., and Scott, R.W. (1996). Enhanced
amyloidogenic processing of the β-amyloid precursor protein in gene-
targeted mice bearing the Swedish familial Alzheimers disease
mutations and a “humanized” sequence. J Biol Chem 271, 23380–
23388.
Reichwald, J., Danner, S., Wiederhold, K.H., and Staufenbiel, M. (2009).
Expression of complement system components during aging and
amyloid deposition in APP transgenic mice. J Neuroinflammation 6, 35.
Richard, B.C., Kurdakova, A., Baches, S., Bayer, T.A., Weggen, S., and
Wirths, O. (2015). Gene dosage dependent aggravation of the
neurological phenotype in the 5XFAD mouse model of Alzheimer’s
disease. J Alzheimer Dis 45, 1223–1236.
Richards, J.G., Higgins, G.A., Ouagazzal, A.M., Ozmen, L., Kew, J.N.C.,
Bohrmann, B., Malherbe, P., Brockhaus, M., Loetscher, H., Czech, C.,
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.
Richardson, J.C., Kendal, C.E., Anderson, R., Priest, F., Gower, E., Soden,
P., Gray, R., Topps, S., Howlett, D.R., Lavender, D., et al. (2003).
40 Li, X., et al. Sci China Life Sci
Ultrastructural and behavioural changes precede amyloid deposition in
a transgenic model of Alzheimer’s disease. Neuroscience 122, 213–228.
Rockenstein, E., Mallory, M., Mante, M., Sisk, A., and Masliaha, E. (2001).
Early formation of mature amyloid-beta protein deposits in a mutant
APP transgenic model depends on levels of Aβ1-42. J Neurosci Res 66,
573–582.
Rockenstein, E., Mante, M., Alford, M., Adame, A., Crews, L., Hashimoto,
M., Esposito, L., Mucke, L., and Masliah, E. (2005). High β-secretase
activity elicits neurodegeneration in transgenic mice despite reductions
in amyloid-β levels. J Biol Chem 280, 32957–32967.
Roder, S., Danober, L., Pozza, M.F., Lingenhoehl, K., Wiederhold, K.H.,
and Olpe, H.R. (2003). Electrophysiological studies on the
hippocampus and prefrontal cortex assessing the effects of
amyloidosis in amyloid precursor protein 23 transgenic mice.
Neuroscience 120, 705–720.
Rönnbäck, A., Sagelius, H., Bergstedt, K.D., Näslund, J., Westermark, G.
T., Winblad, B., and Graff, C. (2012). Amyloid neuropathology in the
single Arctic APP transgenic model affects interconnected brain
regions. Neurobiol Aging 33, 831.e11–831.e19.
Rupp, N.J., Wegenast-Braun, B.M., Radde, R., Calhoun, M.E., and Jucker,
M. (2011). Early onset amyloid lesions lead to severe neuritic
abnormalities and local, but not global neuron loss in APPPS1
transgenic mice. Neurobiol Aging 32, 2324.e1–2324.e6.
Sadowski, M., Pankiewicz, J., Scholtzova, H., Ji, Y., Quartermain, D.,
Jensen, C.H., Duff, K., Nixon, R.A., Gruen, R.J., and Wisniewski, T.
(2004). Amyloid-β deposition is associated with decreased hippocampal
glucose metabolism and spatial memory impairment in APP/PS1 mice.
J Neuropathol Exp Neurol 63, 418–428.
Saganich, M.J., Schroeder, B.E., Galvan, V., Bredesen, D.E., Koo, E.H.,
and Heinemann, S.F. (2006). Deficits in synaptic transmission and
learning in amyloid precursor protein (APP) transgenic mice require C-
terminal cleavage of APP. J Neurosci 26, 13428–13436.
Sagare, A.P., Bell, R.D., Zhao, Z., Ma, Q., Winkler, E.A., Ramanathan, A.,
and Zlokovic, B.V. (2013). Pericyte loss influences Alzheimer-like
neurodegeneration in mice. Nat Commun 4, 2932.
Saito, S., Yamamoto, Y., Maki, T., Hattori, Y., Ito, H., Mizuno, K., Harada-
Shiba, M., Kalaria, R.N., Fukushima, M., Takahashi, R., et al. (2017).
Taxifolin inhibits amyloid-β oligomer formation and fully restores
vascular integrity and memory in cerebral amyloid angiopathy. Acta
Neuropathol Commun 5, 26.
Saito, T., Matsuba, Y., Mihira, N., Takano, J., Nilsson, P., Itohara, S., Iwata,
N., and Saido, T.C. (2014). Single App knock-in mouse models of
Alzheimer’s disease. Nat Neurosci 17, 661–663.
Saito, T., Mihira, N., Matsuba, Y., Sasaguri, H., Hashimoto, S.,
Narasimhan, S., Zhang, B., Murayama, S., Higuchi, M., Lee, V.M.Y.,
et al. (2019). Humanization of the entire murine Mapt gene provides a
murine model of pathological human tau propagation. J Biol Chem 294,
12754–12765.
Saito, T., Suemoto, T., Brouwers, N., Sleegers, K., Funamoto, S., Mihira,
N., Matsuba, Y., Yamada, K., Nilsson, P., Takano, J., et al. (2011).
Potent amyloidogenicity and pathogenicity of Aβ43. Nat Neurosci 14,
1023–1032.
Sanchez-Varo, R., Mejias-Ortega, M., Fernandez-Valenzuela, J.J., Nuñez-
Diaz, C., Caceres-Palomo, L., Vegas-Gomez, L., Sanchez-Mejias, E.,
Trujillo-Estrada, L., Garcia-Leon, J.A., Moreno-Gonzalez, I., et al.
(2022). Transgenic mouse models of Alzheimer’s disease: an integrative
analysis. Int J Mol Sci 23, 5404.
SantaCruz, K., Lewis, J., Spires, T., Paulson, J., Kotilinek, L., Ingelsson,
M., Guimaraes, A., DeTure, M., Ramsden, M., McGowan, E., et al.
(2005). Tau suppression in a neurodegenerative mouse model improves
memory function. Science 309, 476–481.
Sasaguri, H., Hashimoto, S., Watamura, N., Sato, K., Takamura, R., Nagata,
K., Tsubuki, S., Ohshima, T., Yoshiki, A., Sato, K., et al. (2022). Recent
advances in the modeling of Alzheimer’s disease. Front Neurosci 16,
807473.
Sasaguri, H., Nilsson, P., Hashimoto, S., Nagata, K., Saito, T., De Strooper,
B., Hardy, J., Vassar, R., Winblad, B., and Saido, T.C. (2017). APP mouse
models for Alzheimer’s disease preclinical studies. EMBO J 36, 2473–
2487.
Sato, K., Watamura, N., Fujioka, R., Mihira, N., Sekiguchi, M., Nagata, K.,
Ohshima, T., Saito, T., Saido, T.C., and Sasaguri, H. (2021). A third-
generation mouse model of Alzheimer’s disease shows early and
increased cored plaque pathology composed of wild-type human
amyloid β peptide. J Biol Chem 297, 101004.
Savonenko, A., Xu, G.M., Melnikova, T., Morton, J.L., Gonzales, V.,
Wong, M.P.F., Price, D.L., Tang, F., Markowska, A.L., and Borchelt, D.
R. (2005). Episodic-like memory deficits in the APPswe/PS1dE9 mouse
model of Alzheimer’s disease: Relationships to β-amyloid deposition
and neurotransmitter abnormalities. Neurobiol Dis 18, 602–617.
Savonenko, A.V., Xu, G.M., Price, D.L., Borchelt, D.R., and Markowska,
A.L. (2003). Normal cognitive behavior in two distinct congenic lines
of transgenic mice hyperexpressing mutant APPSWE.Neurobiol Dis 12,
194–211.
Scattoni, M.L., Gasparini, L., Alleva, E., Goedert, M., Calamandrei, G.,
and Spillantini, M.G. (2010). Early behavioural markers of disease in
P301S tau transgenic mice. Behav Brain Res 208, 250–257.
Scearce-Levie, K., Sanchez, P.E., and Lewcock, J.W. (2020). Leveraging
preclinical models for the development of Alzheimer disease
therapeutics. Nat Rev Drug Discov 19, 447–462.
Schneider, I., Reversé, D., Dewachter, I., Ris, L., Caluwaerts, N., Kuipéri,
C., Gilis, M., Geerts, H., Kretzschmar, H., Godaux, E., et al. (2001).
Mutant presenilins disturb neuronal calcium homeostasis in the brain of
transgenic mice, decreasing the threshold for excitotoxicity and
facilitating long-term potentiation. J Biol Chem 276, 11539–11544.
Sebollela, A., Cline, E.N., Popova, I., Luo, K., Sun, X., Ahn, J., Barcelos,
M.A., Bezerra, V.N., Lyra e Silva, N.M., Patel, J., et al. (2017). A
human scFv antibody that targets and neutralizes high molecular weight
pathogenic amyloid-β oligomers. J Neurochem 142, 934–947.
Sehar, U., Rawat, P., Reddy, A.P., Kopel, J., and Reddy, P.H. (2022).
Amyloid beta in aging and Alzheimer’s disease. Int J Mol Sci 23,
12924.
Selkoe, D.J., and Hardy, J. (2016). The amyloid hypothesis of Alzheimer’s
disease at 25 years. EMBO Mol Med 8, 595–608.
Serneels, L., Van Biervliet, J., Craessaerts, K., Dejaegere, T., Horré, K., Van
Houtvin, T., Esselmann, H., Paul, S., Schäfer, M.K., Berezovska, O., et
al. (2009). γ-Secretase heterogeneity in the Aph1 subunit: relevance for
Alzheimer’s disease. Science 324, 639–642.
Sevigny, J., Chiao, P., Bussière, T., Weinreb, P.H., Williams, L., Maier, M.,
Dunstan, R., Salloway, S., Chen, T., Ling, Y., et al. (2016). The antibody
aducanumab reduces plaques in Alzheimer’s disease. Nature 537,
50–56.
Shen, B., Zhang, J., Wu, H., Wang, J., Ma, K., Li, Z., Zhang, X., Zhang, P.,
and Huang, X. (2013). Generation of gene-modified mice via Cas9/
RNA-mediated gene targeting. Cell Res 23, 720–723.
Shen, L., Qin, W., Wu, L., Zhou, A., Tang, Y., Wang, Q., Jia, L., and Jia, J.
(2019). Two novel presenilin-1 mutations (I249L and P433S) in early
onset Chinese Alzheimer’s pedigrees and their functional
characterization. Biochem Biophys Res Commun 516, 264–269.
Shimojo, M., Sahara, N., Mizoroki, T., Funamoto, S., Morishima-
Kawashima, M., Kudo, T., Takeda, M., Ihara, Y., Ichinose, H., and
Takashima, A. (2008). Enzymatic characteristics of I213T mutant
presenilin-1/γ-secretase in cell models and knock-in mouse brains. J
Biol Chem 283, 16488–16496.
Sierksma, A., Escott-Price, V., and De Strooper, B. (2020). Translating
genetic risk of Alzheimer’s disease into mechanistic insight and drug
targets. Science 370, 61–66.
Skaaraas, G.H.E.S., Melbye, C., Puchades, M.A., Leung, D.S.Y., Jacobsen,
Ø., Rao, S.B., Ottersen, O.P., Leergaard, T.B., and Torp, R. (2021).
Cerebral amyloid angiopathy in a mouse model of Alzheimer’s disease
associates with upregulated angiopoietin and downregulated hypoxia-
inducible factor. J Alzheimer Dis 83, 1651–1663.
Snellman, A., López-Picón, F.R., Rokka, J., Salmona, M., Forloni, G.,
Scheinin, M., Solin, O., Rinne, J.O., and Haaparanta-Solin, M. (2013).
Longitudinal amyloid imaging in mouse brain with 11C-PIB:
41
Li, X., et al. Sci China Life Sci
comparison of APP23, Tg2576, and APPswe-PS1dE9 mouse models of
Alzheimer disease. J Nucl Med 54, 1434–1441.
Song, W.M., Joshita, S., Zhou, Y., Ulland, T.K., Gilfillan, S., and Colonna,
M. (2018). Humanized TREM2 mice reveal microglia-intrinsic and
-extrinsic effects of R47H polymorphism. J Exp Med 215, 745–760.
Soriano, P. (1999). Generalized lacZ expression with the ROSA26 Cre
reporter strain. Nat Genet 21, 70–71.
Spires, T.L., Orne, J.D., SantaCruz, K., Pitstick, R., Carlson, G.A., Ashe, K.
H., and Hyman, B.T. (2006). Region-specific dissociation of neuronal
loss and neurofibrillary pathology in a mouse model of tauopathy. Am J
Pathol 168, 1598–1607.
Stalder, M., Phinney, A., Probst, A., Sommer, B., Staufenbiel, M., and
Jucker, M. (1999). Association of microglia with amyloid plaques in
brains of APP23 transgenic mice. Am J Pathol 154, 1673–1684.
Stover, K.R., Campbell, M.A., Van Winssen, C.M., and Brown, R.E.
(2015). Early detection of cognitive deficits in the 3xTg-AD mouse
model of Alzheimer’s disease. Behav Brain Res 289, 29–38.
Sturchler-Pierrat, C., Abramowski, D., Duke, M., Wiederhold, K.H., Mistl,
C., Rothacher, S., Ledermann, B., Bürki, K., Frey, P., Paganetti, P.A., et
al. (1997). Two amyloid precursor protein transgenic mouse models
with Alzheimer disease-like pathology. Proc Natl Acad Sci USA 94,
13287–13292.
Sun, B., Chen, Y., Fan, D., Zhu, C., Zeng, F., and Wang, Y. (2021). Critical
thinking on amyloid-beta-targeted therapy: challenges and perspectives.
Sci China Life Sci 64, 926–937.
Sun, Z.Q., Liu, J.F., Luo, W., Wong, C.H., So, K.F., Hu, Y., and Chiu, K.
(2022). Lycium barbarum extract promotes M2 polarization and reduces
oligomeric amyloid-β-induced inflammatory reactions in microglial
cells. Neural Regen Res 17, 203–209.
Swanson, C.J., Zhang, Y., Dhadda, S., Wang, J., Kaplow, J., Lai, R.Y.K.,
Lannfelt, L., Bradley, H., Rabe, M., Koyama, A., et al. (2021). A
randomized, double-blind, phase 2b proof-of-concept clinical trial in
early Alzheimer’s disease with lecanemab, an anti-Aβ protofibril
antibody. Alzheimers Res Ther 13, 80.
Sydow, A., Hochgräfe, K., Könen, S., Cadinu, D., Matenia, D., Petrova, O.,
Joseph, M., Dennissen, F.J., and Mandelkow, E.M. (2016). Age-
dependent neuroinflammation and cognitive decline in a novel
Ala152Thr-Tau transgenic mouse model of PSP and AD. Acta
Neuropathol Commun 4, 17.
Sydow, A., Van der Jeugd, A., Zheng, F., Ahmed, T., Balschun, D., Petrova,
O., Drexler, D., Zhou, L., Rune, G., Mandelkow, E., et al. (2011). Tau-
induced defects in synaptic plasticity, learning, and memory are
reversible in transgenic mice after switching off the toxic Tau mutant.
J Neurosci 31, 2511–2525.
Takahashi, R.H., Almeida, C.G., Kearney, P.F., Yu, F., Lin, M.T., Milner, T.
A., and Gouras, G.K. (2004). Oligomerization of Alzheimer’s β-
amyloid within processes and synapses of cultured neurons and brain. J
Neurosci 24, 3592–3599.
Takeuchi, H., Iba, M., Inoue, H., Higuchi, M., Takao, K., Tsukita, K.,
Karatsu, Y., Iwamoto, Y., Miyakawa, T., Suhara, T., et al. (2011). P301S
mutant human tau transgenic mice manifest early symptoms of human
tauopathies with dementia and altered sensorimotor gating. PLoS ONE
6, e21050.
Tanemura, K., Akagi, T., Murayama, M., Kikuchi, N., Murayama, O.,
Hashikawa, T., Yoshiike, Y., Park, J.M., Matsuda, K., Nakao, S., et al.
(2001). Formation of filamentous tau aggregations in transgenic mice
expressing V337M human tau. Neurobiol Dis 8, 1036–1045.
Tanemura, K., Chui, D.H., Fukuda, T., Murayama, M., Park, J.M., Akagi,
T., Tatebayashi, Y., Miyasaka, T., Kimura, T., Hashikawa, T., et al.
(2006). Formation of tau inclusions in knock-in mice with familial
Alzheimer disease (FAD) mutation of presenilin 1 (PS1). J Biol Chem
281, 5037–5041.
Tanemura, K., Murayama, M., Akagi, T., Hashikawa, T., Tominaga, T.,
Ichikawa, M., Yamaguchi, H., and Takashima, A. (2002).
Neurodegeneration with tau accumulation in a transgenic mouse
expressing V337M human tau. J Neurosci 22, 133–141.
Tatebayashi, Y., Miyasaka, T., Chui, D.H., Akagi, T., Mishima, K., Iwasaki,
K., Fujiwara, M., Tanemura, K., Murayama, M., Ishiguro, K., et al.
(2002). Tau filament formation and associative memory deficit in aged
mice expressing mutant (R406W) human tau. Proc Natl Acad Sci USA
99, 13896–13901.
Terwel, D., Lasrado, R., Snauwaert, J., Vandeweert, E., Van Haesendonck,
C., Borghgraef, P., and Van Leuven, F. (2005). Changed conformation
of mutant Tau-P301L underlies the moribund tauopathy, absent in
progressive, nonlethal axonopathy of Tau-4R/2N transgenic mice. J
Biol Chem 280, 3963–3973.
Thomas, K.R., and Capecchi, M.R. (1987). Site-directed mutagenesis by
gene targeting in mouse embryo-derived stem cells. Cell 51, 503–512.
Tian, Y., Wang, W., Xu, L., Li, H., Wei, Y., Wu, Q., and Jia, J. (2019).
Activation of Nrf2/ARE pathway alleviates the cognitive deficits in
PS1V97L-Tg mouse model of Alzheimer’s disease through modulation
of oxidative stress. J Neurosci Res 97, 492–505.
Tomiyama, T., Matsuyama, S., Iso, H., Umeda, T., Takuma, H., Ohnishi,
K., Ishibashi, K., Teraoka, R., Sakama, N., Yamashita, T., et al. (2010).
A mouse model of amyloid β oligomers: their contribution to synaptic
alteration, abnormal tau phosphorylation, glial activation, and neuronal
loss in vivo.J Neurosci 30, 4845–4856.
Tomiyama, T., Nagata, T., Shimada, H., Teraoka, R., Fukushima, A.,
Kanemitsu, H., Takuma, H., Kuwano, R., Imagawa, M., Ataka, S., et al.
(2008). A new amyloid β variant favoring oligomerization in
Alzheimer’s-type dementia. Ann Neurol 63, 377–387.
Tomiyama, T., and Shimada, H. (2020). APP Osaka mutation in familial
Alzheimer’s disease—its discovery, phenotypes, and mechanism of
recessive inheritance. Int J Mol Sci 21, 1413.
Tucker, S., Möller, C., Tegerstedt, K., Lord, A., Laudon, H., Sjödahl, J.,
Söderberg, L., Spens, E., Sahlin, C., Waara, E.R., et al. (2015). The
murine version of BAN2401 (mAb158) selectively reduces amyloid-β
protofibrils in brain and cerebrospinal fluid of tg-ArcSwe mice. J
Alzheimer Dis 43, 575–588.
Umeda, T., Kimura, T., Yoshida, K., Takao, K., Fujita, Y., Matsuyama, S.,
Sakai, A., Yamashita, M., Yamashita, Y., Ohnishi, K., et al. (2017).
Mutation-induced loss of APP function causes GABAergic depletion in
recessive familial Alzheimer’s disease: analysis of Osaka mutation-
knockin mice. Acta Neuropathol Commun 5, 59.
Umeda, T., Maekawa, S., Kimura, T., Takashima, A., Tomiyama, T., and
Mori, H. (2014). Neurofibrillary tangle formation by introducing wild-
type human tau into APP transgenic mice. Acta Neuropathol 127, 685–
698.
Umeda, T., Tomiyama, T., Sakama, N., Tanaka, S., Lambert, M.P., Klein,
W.L., and Mori, H. (2011). Intraneuronal amyloid β oligomers cause
cell death via endoplasmic reticulum stress, endosomal/lysosomal
leakage, and mitochondrial dysfunction in vivo.J Neurosci Res 89,
1031–1042.
Van Dam, D., D’Hooge, R., Staufenbiel, M., Van Ginneken, C., Van Meir,
F., and De Deyn, P.P. (2003). Age-dependent cognitive decline in the
APP23 model precedes amyloid deposition. Eur J Neurosci 17, 388–
396.
Van der Jeugd, A., Hochgräfe, K., Ahmed, T., Decker, J.M., Sydow, A.,
Hofmann, A., Wu, D., Messing, L., Balschun, D., D’Hooge, R., et al.
(2012). Cognitive defects are reversible in inducible mice expressing
pro-aggregant full-length human Tau. Acta Neuropathol 123, 787–805.
Van Dorpe, J., Smeijers, L., Dewachter, I., Nuyens, D., Spittaels, K., Van
den Haute, C., Mercken, M., Moechars, D., Laenen, I., Kuiperi, C., et
al. (2000). Prominent cerebral amyloid angiopathy in transgenic mice
overexpressing the london mutant of human APP in neurons. Am J
Pathol 157, 1283–1298.
van Dyck, C.H., Swanson, C.J., Aisen, P., Bateman, R.J., Chen, C., Gee,
M., Kanekiyo, M., Li, D., Reyderman, L., Cohen, S., et al. (2023).
Lecanemab in early Alzheimer’s disease. N Engl J Med 388, 9–21.
van Ham, T.J., Breitling, R., Swertz, M.A., and Nollen, E.A.A. (2009).
Neurodegenerative diseases: Lessons from genome-wide screens in
small model organisms. EMBO Mol Med 1, 360–370.
Van Skike, C.E., Hussong, S.A., Hernandez, S.F., Banh, A.Q., DeRosa, N.,
and Galvan, V. (2021). mTOR attenuation with rapamycin reverses
42 Li, X., et al. Sci China Life Sci
neurovascular uncoupling and memory deficits in mice modeling
Alzheimer’s disease. J Neurosci 41, 4305–4320.
Veitch, D.P., Weiner, M.W., Aisen, P.S., Beckett, L.A., Cairns, N.J., Green,
R.C., Harvey, D., Jack Jr., C.R., Jagust, W., Morris, J.C., et al. (2019).
Understanding disease progression and improving Alzheimer’s disease
clinical trials: recent highlights from the Alzheimer’s Disease
Neuroimaging Initiative. Alzheimers Dement 15, 106–152.
Vitale, F., Ortolan, J., Volpe, B.T., Marambaud, P., Giliberto, L., and
d’Abramo, C. (2020). Intramuscular injection of vectorized-scFvMC1
reduces pathological tau in two different tau transgenic models. Acta
Neuropathol Commun 8, 126.
Volianskis, A., Køstner, R., Mølgaard, M., Hass, S., and Jensen, M.S.
(2010). Episodic memory deficits are not related to altered
glutamatergic synaptic transmission and plasticity in the CA1
hippocampus of the APPswe/PS1ΔE9-deleted transgenic mice model
of β-amyloidosis. Neurobiol Aging 31, 1173–1187.
Wang, H., Xu, X.X., Pan, Y.C., Yan, Y.X., Hu, X.Y., Chen, R.W., Ravoo, B.
J., Guo, D.S., and Zhang, T. (2021a). Recognition and removal of
amyloid-β by a heteromultivalent macrocyclic coassembly: a potential
strategy for the treatment of Alzheimer’s disease. Adv Mater 33,
2006483.
Wang, J., Guo, X., Lu, W., Liu, J., Zhang, H., Quan, Q., Su, H., Ma, L.,
Gao, F., and Qu, Q. (2021b). Donepezil combined with DL-3-n-
butylphthalide delays cognitive decline in patients with mild to
moderate Alzheimer’s disease: a multicenter, prospective cohort
study. J Alzheimer Dis 80, 673–681.
Wang, J., Tanila, H., Puoliväli, J., Kadish, I., and Groen, T. (2003). Gender
differences in the amount and deposition of amyloidβ in APPswe and
PS1 double transgenic mice. Neurobiol Dis 14, 318–327.
Wang, R., Dineley, K.T., Sweatt, J.D., and Zheng, H. (2004). Presenilin 1
familial Alzheimer’s disease mutation leads to defective associative
learning and impaired adult neurogenesis. Neuroscience 126, 305–312.
Wang, W., Lu, L., Wu, Q., and Jia, J. (2016). Brain amyloid-β plays an
initiating role in the pathophysiological process of the PS1V97L-Tg
mouse model of Alzheimer’s disease. J Alzheimer Dis 52, 1089–1099.
Wang, W., Wei, C., Quan, M., Li, T., and Jia, J. (2020). Sulforaphane
reverses the amyloid-β oligomers induced depressive-like behavior. J
Alzheimer Dis 78, 127–137.
Wang, W.Z., Li, M.W., Chen, Y., Liu, L.Y., Xu, Y., Xia, Z.H., Yu, Y., Wang,
X.D., Chen, W., Zhang, F., et al. (2021c). 3xTg-AD mice over-
expressing phospholipid transfer protein improves cognition through
decreasing amyloid-beta production and tau hyperphosphorylation. J
Alzheimers Dis 82, 1635–1649.
Wang, X., Sun, G., Feng, T., Zhang, J., Huang, X., Wang, T., Xie, Z., Chu,
X., Yang, J., Wang, H., et al. (2019). Sodium oligomannate
therapeutically remodels gut microbiota and suppresses gut bacterial
amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease
progression. Cell Res 29, 787–803.
Wang, Y., Cella, M., Mallinson, K., Ulrich, J.D., Young, K.L., Robinette,
M.L., Gilfillan, S., Krishnan, G.M., Sudhakar, S., Zinselmeyer, B.H., et
al. (2015). TREM2 lipid sensing sustains the microglial response in an
Alzheimer’s disease model. Cell 160, 1061–1071.
Wang, Y., Cheng, Z., Qin, W., and Jia, J. (2012). Val97Leu mutant
presenilin-1 induces tau hyperphosphorylation and spatial memory
deficit in mice and the underlying mechanisms. J Neurochem 121, 135–
145.
Watamura, N., Sato, K., and Saido, T.C. (2022a). Mouse models of
Alzheimer’s disease for preclinical research. Neurochem Int 158,
105361.
Watamura, N., Sato, K., Shiihashi, G., Iwasaki, A., Kamano, N., Takahashi,
M., Sekiguchi, M., Mihira, N., Fujioka, R., Nagata, K., et al. (2022b).
An isogenic panel of App knock-in mouse models: Profiling β-secretase
inhibition and endosomal abnormalities. Sci Adv 8, eabm6155.
Weidensteiner, C., Metzger, F., Bohrmann, A.B.B., Kuennecke, B., and von
Kienlin, M. (2009). Cortical hypoperfusion in the B6.PS2APP mouse
model for Alzheimer’s disease: Comprehensive phenotyping of vascular
and tissular parameters by MRI. Magn Reson Med 62, 35–45.
Wen, P.H., Hof, P.R., Chen, X., Gluck, K., Austin, G., Younkin, S.G.,
Younkin, L.H., DeGasperi, R., Gama Sosa, M.A., Robakis, N.K., et al.
(2004). The presenilin-1 familial Alzheimer disease mutant P117L
impairs neurogenesis in the hippocampus of adult mice. Exp Neurol
188, 224–237.
Wen, P.H., Shao, X., Shao, Z., Hof, P.R., Wisniewski, T., Kelley, K.,
Friedrich Jr., V.L., Ho, L., Pasinetti, G.M., Shioi, J., et al. (2002).
Overexpression of wild type but not an FAD mutant presenilin-1
promotes neurogenesis in the hippocampus of adult mice. Neurobiol
Dis 10, 8–19.
Wilcock, D.M., Lewis, M.R., Van Nostrand, W.E., Davis, J., Previti, M.L.,
Gharkholonarehe, N., Vitek, M.P., and Colton, C.A. (2008). Progression
of amyloid pathology to Alzheimer’s disease pathology in an amyloid
precursor protein transgenic mouse model by removal of nitric oxide
synthase 2. J Neurosci 28, 1537–1545.
Wilhelmus, M.M.M., Chouchane, O., Loos, M., Jongenelen, C.A.M.,
Brevé, J.J.P., Jonker, A., Bol, J.G.J.M., Smit, A.B., and Drukarch, B.
(2022). Absence of tissue transglutaminase reduces amyloid-beta
pathology in APP23 mice. Neuropathol Appl Neurobio 48, e12796.
Williams, T., Ruiz, A.J., Ruiz, A.M., Vo, Q., Tsering, W., Xu, G.,
McFarland, K., Giasson, B.I., Sullivan, P., Borchelt, D.R., et al. (2022).
Impact of APOE genotype on prion-type propagation of tauopathy. Acta
Neuropathol Commun 10, 57.
Willuweit, A., Velden, J., Godemann, R., Manook, A., Jetzek, F., Tintrup,
H., Kauselmann, G., Zevnik, B., Henriksen, G., Drzezga, A., et al.
(2009). Early-onset and robust amyloid pathology in a new
homozygous mouse model of Alzheimer’s disease. PLoS ONE 4,
e7931.
Wirths, O., Erck, C., Martens, H., Harmeier, A., Geumann, C., Jawhar, S.,
Kumar, S., Multhaup, G., Walter, J., Ingelsson, M., et al. (2010).
Identification of low molecular weight pyroglutamate oligomers in
Alzheimer disease. J Biol Chem 285, 41517–41524.
Wittnam, J.L., Portelius, E., Zetterberg, H., Gustavsson, M.K., Schilling, S.,
Koch, B., Demuth, H.U., Blennow, K., Wirths, O., and Bayer, T.A.
(2012). Pyroglutamate amyloid β (Aβ) aggravates behavioral deficits in
transgenic amyloid mouse model for Alzheimer disease. J Biol Chem
287, 8154–8162.
Wong, T.H., Seelaar, H., Melhem, S., Rozemuller, A.J.M., and van Swieten,
J.C. (2020). Genetic screening in early-onset Alzheimer’s disease
identified three novel presenilin mutations. Neurobiol Aging 86, 201.
e9–201.e14.
Wright, A.L., Zinn, R., Hohensinn, B., Konen, L.M., Beynon, S.B., Tan, R.
P., Clark, I.A., Abdipranoto, A., and Vissel, B. (2013).
Neuroinflammation and neuronal loss precede plaque deposition
in the hAPP-J20 mouse model of Alzheimer’s disease. PLoS ONE 8,
e59586.
Xia, D., Lianoglou, S., Sandmann, T., Calvert, M., Suh, J., Thomsen, E.,
Dugas, J., Pizzo, M., Devos, S., Earr, T., et al. (2021). Fibrillar
causes profound microglial metabolic perturbations in a novel APP
knock-in mouse model. BioRxiv, 2021.01.19.426731.
Xian, Y.F., Qu, C., Liu, Y., Ip, S.P., Yuan, Q.J., Yang, W., and Lin, Z.X.
(2020). Magnolol ameliorates behavioral impairments and
neuropathology in a transgenic mouse model of Alzheimer’s disease.
Oxid Med Cell Longev 2020, 1–17.
Xiao, N.A., Zhang, J., Zhou, M., Wei, Z., Wu, X.L., Dai, X.M., Zhu, Y.G.,
and Chen, X.C. (2015). Reduction of glucose metabolism in olfactory
bulb is an earlier Alzheimer’s disease-related biomarker in 5XFAD
mice. Chin Med J 128, 2220–2227.
Xie, Z., Meng, J., Kong, W., Wu, Z., Lan, F., Narengaowa, F., Hayashi, Y.,
Yang, Q., Bai, Z., Nakanishi, H., et al. (2022). Microglial cathepsin E
plays a role in neuroinflammation and amyloid β production in
Alzheimer’s disease. Aging Cell 21, e13565.
Xu, F., Grande, A.M., Robinson, J.K., Previti, M.L., Vasek, M., Davis, J.,
and Van Nostrand, W.E. (2007). Early-onset subicular microvascular
amyloid and neuroinflammation correlate with behavioral deficits in
vasculotropic mutant amyloid β-protein precursor transgenic mice.
Neuroscience 146, 98–107.
43
Li, X., et al. Sci China Life Sci
Xu, H., Rösler, T.W., Carlsson, T., de Andrade, A., Bruch, J., Höllerhage,
M., Oertel, W.H., and Höglinger, G.U. (2014). Memory deficits
correlate with tau and spine pathology in P301S MAPT transgenic
mice. Neuropathol Appl Neurobiol 40, 833–843.
Yamada, K., Yabuki, C., Seubert, P., Schenk, D., Hori, Y., Ohtsuki, S.,
Terasaki, T., Hashimoto, T., and Iwatsubo, T. (2009).
immunotherapy: intracerebral sequestration of by an anti-Aβ
monoclonal antibody 266 with high affinity to soluble Aβ. J Neurosci
29, 11393–11398.
Yao, Y., Ren, Z., Yang, R., Mei, Y., Dai, Y., Cheng, Q., Xu, C., Xu, X.,
Wang, S., Kim, K.M., et al. (2022). Salidroside reduces neuropathology
in Alzheimer’s disease models by targeting NRF2/SIRT3 pathway. Cell
Biosci 12, 180.
Yi-Bin, W., Xiang, L., Bing, Y., Qi, Z., Fei-Tong, J., Minghong, W.,
Xiangxiang, Z., Le, K., Yan, L., Ping, S., et al. (2022). Inhibition of the
CEBPβ-NFκB interaction by nanocarrier-packaged Carnosic acid
ameliorates glia-mediated neuroinflammation and improves cognitive
function in an Alzheimer’s disease model. Cell Death Dis 13, 318.
Yoshiyama, Y., Higuchi, M., Zhang, B., Huang, S.M., Iwata, N., Saido, T.
C., Maeda, J., Suhara, T., Trojanowski, J.Q., and Lee, V.M.Y. (2007).
Synapse loss and microglial activation precede tangles in a P301S
tauopathy mouse model. Neuron 53, 337–351.
Youmans, K.L., Tai, L.M., Nwabuisi-Heath, E., Jungbauer, L., Kanekiyo,
T., Gan, M., Kim, J., Eimer, W.A., Estus, S., Rebeck, G.W., et al.
(2012). APOE4-specific changes in accumulation in a new
transgenic mouse model of Alzheimer disease. J Biol Chem 287,
41774–41786.
Yu, J.T., Xu, W., Tan, C.C., Andrieu, S., Suckling, J., Evangelou, E., Pan,
A., Zhang, C., Jia, J., Feng, L., et al. (2020). Evidence-based prevention
of Alzheimer’s disease: systematic review and meta-analysis of 243
observational prospective studies and 153 randomised controlled trials.
J Neurol Neurosurg Psychiatry 91, 1201–1209.
Yue, M., Hanna, A., Wilson, J., Roder, H., and Janus, C. (2011). Sex
difference in pathology and memory decline in rTg4510 mouse model
of tauopathy. Neurobiol Aging 32, 590–603.
Zampar, S., and Wirths, O. (2021). Characterization of a mouse model of
Alzheimer’s disease expressing Aβ4-42 and human mutant tau. Int J
Mol Sci 22, 5191.
Zhang, H., Han, T., Zhang, L., Yu, C.H., Wan, D.G., Rahman, K., Qin, L.P.,
and Peng, C. (2008). Effects of tenuifolin extracted from radix
polygalae on learning and memory: a behavioral and biochemical
study on aged and amnesic mice. Phytomedicine 15, 587–594.
Zhang, L., Chen, C., Mak, M.S., Lu, J., Wu, Z., Chen, Q., Han, Y., Li, Y.,
and Pi, R. (2020). Advance of sporadic Alzheimer’s disease animal
models. Med Res Rev 40, 431–458.
Zhang, X., Wei, X., Mei, Y., Wang, D., Wang, J., Zhang, Y., Li, X., Gu, Y.,
Peng, G., and Sun, B. (2021). Modulating adult neurogenesis affects
synaptic plasticity and cognitive functions in mouse models of
Alzheimer’s disease. Stem Cell Rep 16, 3005–3019.
Zhang, Y., Huang, L.J., Shi, S., Xu, S.F., Wang, X.L., and Peng, Y. (2016).
L-3-n-butylphthalide rescues hippocampal synaptic failure and
attenuates neuropathology in aged APP/PS1 mouse model of
Alzheimer’s disease. CNS Neurosci Ther 22, 979–987.
Zhang, Y., Lu, L., Jia, J., Jia, L., Geula, C., Pei, J., Xu, Z., Qin, W., Liu, R.,
Li, D., et al. (2014). A lifespan observation of a novel mouse model: in
vivo evidence supports oligomer hypothesis. PLoS ONE 9, e85885.
Zhao, S., Fan, Z., Zhang, X., Li, Z., Shen, T., Li, K., Yan, Y., Yuan, Y., Pu,
J., Tian, J., et al. (2023). Metformin attenuates tau pathology in tau-
seeded PS19 mice. Neurotherapeutics 20, 452–463.
Zhong, W., Wu, A., Berglund, K., Gu, X., Jiang, M.Q., Talati, J., Zhao, J.,
Wei, L., and Yu, S.P. (2022). Pathogenesis of sporadic Alzheimer’s
disease by deficiency of NMDA receptor subunit GluN3A. Alzheimers
Dement 18, 222–239.
Zou, C., Montagna, E., Shi, Y., Peters, F., Blazquez-Llorca, L., Shi, S.,
Filser, S., Dorostkar, M.M., and Herms, J. (2015). Intraneuronal APP
and extracellular independently cause dendritic spine pathology in
transgenic mouse models of Alzheimer’s disease. Acta Neuropathol
129, 909–920.
Zufferey, V., Vallet, P.G., Moeri, M., Moulin-Sallanon, M., Piotton, F.,
Marin, P., and Savioz, A. (2013). Maladaptive exploratory behavior and
neuropathology of the PS-1 P117L Alzheimer transgenic mice. Brain
Res Bull 94, 17–22.
44 Li, X., et al. Sci China Life Sci
... These experimental facts suggest that DTD1 may be involved in physiological or pathological brain aging. Other pathological processes, such as schizophrenia, depression, and bipolar disorder, are associated with D-Ser (Bendikov et al., 2007;El-Tallawy et al., 2017;Fuchs et al., 2008;Guercio and Panizzutti, 2018;Hashimoto et al., 2007;Hashimoto et al., 2016;Li et al., 2023;Pålsson et al., 2015;Yamamori et al., 2014). Therefore, DTD1 may be potentially affected by the above physiological or pathological processes. ...
Article
D-serine and D-aspartate are involved in N-methyl-D-aspartate receptor (NMDAR)-related physiological and pathological processes. D-aminoacyl-tRNA deacylase 1 (DTD1) may biochemically contribute to D-serine or D-aspartate production. However, it is unclear thus far whether DTD1 regulates D-serine or D-aspartate content in neurobiological processes. In the present research, we found that DTD1 was essential to maintain the D-serine or D-aspartate homeostasis, which was consistent with the phenomenon that DTD1-deficiency resulted in changes in the quantity changes of functional NMDAR subunits in postsynaptic compartments. Moreover, DTD1 played a considerable role in regulating dendritic morphology and synaptic structure. As a consequence, DTD1 affected neurobiological events, including the synaptic strength of the CA3-to-CA1 circuit, dendritic spine density of hippocampal pyramidal neurons, and behavioral performance of mice in the Morris water maze. These findings highlight the important role of DTD1 in synaptic transmission, neuronal morphology, and spatial learning and memory and suggest an undisclosed mechanism of DTD1 that participates the regulation of D-serine or D-aspartate homeostasis in hippocampal neurons.
... On the contrary, what is not controversial is the observation that there is no widespread frank neuronal death, and no neurofibrillary tangles (NTFs) are present in the vast majority of transgenic murine models of FAD. This is observed in mice overexpressing human mutant APP (note that there is no evidence of APP overexpression in AD patients, except in very rare forms of early onset FAD [36]) or mutated hAPP/hPSEN1 that, on the other hand, produce high levels of Aβ [37,38]. NTFs are observed in transgenic mice when, in addition to mutated hAPP or hAPP/hPSEN1 genes, they carry also the P301L human gene for protein tau (hMAPT), a mutation not associated with FAD [39,40]. ...
Article
Full-text available
Over the past 30 years, the majority of (pre)clinical efforts to find an effective therapy for Alzheimer’s disease (AD) focused on clearing the β-amyloid peptide (Aβ) from the brain since, according to the amyloid cascade hypothesis, the peptide was (and it is still considered by many) the pathogenic determinant of this neurodegenerative disorder. However, as reviewed in this article, results from the numerous clinical trials that have tested anti-Aβ therapies to date indicate that this peptide plays a minor role in the pathogenesis of AD. Indeed, even Aducanumab and Lecanemab, the two antibodies recently approved by the FDA for AD therapy, as well as Donanemab showed limited efficacy on cognitive parameters in phase III clinical trials, despite their capability of markedly lowering Aβ brain load. Furthermore, preclinical evidence demonstrates that Aβ possesses several physiological functions, including memory formation, suggesting that AD may in part be due to a loss of function of this peptide. Finally, it is generally accepted that AD could be the result of many molecular dysfunctions, and therefore, if we keep chasing only Aβ, it means that we cannot see the forest for the trees.
Article
Alzheimer’s disease (AD) is a leading cause of dementia and a growing public health concern worldwide. Despite decades of research, effective disease-modifying treatments remain elusive, partly due to limitations in current experimental models. The purpose of this review is to critically assess and compare existing murine and alternative models of AD to identify key strengths, limitations, and future directions for model development that can enhance translational relevance and therapeutic discovery. Traditional transgenic mouse models have advanced the understanding of amyloid-beta and tau pathologies, but often fail to capture the complexity of sporadic, late-onset AD. In response, alternative models—including zebrafish, Drosophila melanogaster, Caenorhabditis elegans, non-human primates, and human brain organoids—are gaining traction due to their complementary insights and diverse experimental advantages. This review also discusses innovations in genetic engineering, neuroimaging, computational modelling, and drug repurposing that are reshaping the landscape of AD research. By integrating these diverse approaches, the review advocates for a multi-model, multidisciplinary strategy to improve the predictive power, accelerate clinical translation, and inform personalised therapeutic interventions. Ethical considerations and equitable access to diagnostics and emerging treatments are also emphasised. Ultimately, this work aims to support the development of more accurate, effective, and human-relevant models to combat AD.
Article
Full-text available
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline, memory loss, and behavioural changes. While genetic predispositions and pathological processes have been the traditional focus, this review highlights the fundamental role of environmental factors, particularly nutrition, within the exposome framework in modulating the risk and progression of AD. The exposome, which includes the totality of environmental exposures in an individual’s lifetime, provides a comprehensive approach to understanding the complex aetiology of AD. In this review, we explore the impact of dietary factors and cyclic nucleotide pathways (cAMP/cGMP) on AD, emphasizing the potential of dietary interventions as therapeutic strategies. We investigate key aspects of how nutrition affects the accumulation of β-amyloid, the aggregation of tau proteins, and neuroinflammation. We also examine the impact of specific nutrients on cognitive performance and the risk of AD. Additionally, we discuss the potential of nutraceuticals with anti-phosphodiesterase activity and the role of various animal models of AD (such as 5xFAD, 3xTg-AD, Tg2576, and APP/PS1 mice) in demonstrating the effects of dietary interventions on disease onset and progression.
Article
Full-text available
Alzheimer’s disease (AD) is a common neurodegenerative disorder characterized by the accumulation of amyloid-beta (Aβ), hyperphosphorylation of tau, and neuroinflammation in the brain. The blood–brain barrier (BBB) limits solutes from circulating blood from entering the brain, which is essential for neuronal functioning. Focusing on BBB function is important for the early detection of AD and in-depth study of AD pathogenic mechanisms. However, the mechanism of BBB alteration in AD is still unclear, which hinders further research on therapeutics that target the BBB to delay the progression of AD. The exact timing of the vascular abnormalities in AD and the complex cause-and-effect relationships remain uncertain. Thus, it is necessary to summarize and emphasize this process. First, in this review, the current evidence for BBB dysfunction in AD is summarized. Then, the interrelationships and pathogenic mechanisms between BBB dysfunction and the risk factors for AD, such as Aβ, tau, neuroinflammation, apolipoprotein E (ApoE) genotype and aging, were analyzed. Finally, we discuss the current status and future directions of therapeutic AD strategies targeting the BBB. We hope that these summaries or reviews will allow readers to better understand the relationship between the BBB and AD.
Article
Full-text available
Background The identification of pathogenic mutations in Alzheimer’s disease (AD) causal genes led to a better understanding of the pathobiology of AD. Familial Alzheimer’s disease (FAD) is known to be associated with mutations in the APP, PSEN1, and PSEN2 genes involved in Aβ production; however, these genetic defects occur in only about 10–20% of FAD cases, and more genes and new mechanism causing FAD remain largely obscure. Methods We performed exome sequencing on family members with a FAD pedigree and identified gene variant ZDHHC21 p.T209S. A ZDHHC21T209S/T209S knock-in mouse model was then generated using CRISPR/Cas9. The Morris water navigation task was then used to examine spatial learning and memory. The involvement of aberrant palmitoylation of FYN tyrosine kinase and APP in AD pathology was evaluated using biochemical methods and immunostaining. Aβ and tau pathophysiology was evaluated using ELISA, biochemical methods, and immunostaining. Field recordings of synaptic long-term potentiation were obtained to examine synaptic plasticity. The density of synapses and dendritic branches was quantified using electron microscopy and Golgi staining. Results We identified a variant (c.999A > T, p.T209S) of ZDHHC21 gene in a Han Chinese family. The proband presented marked cognitive impairment at 55 years of age (Mini-Mental State Examination score = 5, Clinical Dementia Rating = 3). Considerable Aβ retention was observed in the bilateral frontal, parietal, and lateral temporal cortices. The novel heterozygous missense mutation (p.T209S) was detected in all family members with AD and was not present in those unaffected, indicating cosegregation. ZDHHC21T209S/T209S mice exhibited cognitive impairment and synaptic dysfunction, suggesting the strong pathogenicity of the mutation. The ZDHHC21 p.T209S mutation significantly enhanced FYN palmitoylation, causing overactivation of NMDAR2B, inducing increased neuronal sensitivity to excitotoxicity leading to further synaptic dysfunction and neuronal loss. The palmitoylation of APP was also increased in ZDHHC21T209S/T209S mice, possibly contributing to Aβ production. Palmitoyltransferase inhibitors reversed synaptic function impairment. Conclusions ZDHHC21 p.T209S is a novel, candidate causal gene mutation in a Chinese FAD pedigree. Our discoveries strongly suggest that aberrant protein palmitoylation mediated by ZDHHC21 mutations is a new pathogenic mechanism of AD, warranting further investigations for the development of therapeutic interventions.
Article
Full-text available
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by decreased synaptic transmission and cerebral atrophy with appearance of amyloid plaques and neurofibrillary tangles. Cognitive, functional, and behavioral alterations are commonly associated with the disease. Different pathophysiological pathways of AD have been proposed, some of which interact and influence one another. Current treatment for AD mainly involves the use of therapeutic agents to alleviate the symptoms in AD patients. The conventional single-target treatment approaches do not often cause the desired effect in the disease due to its multifactorial origin. Thus, multi-target strategies have since been undertaken, which aim to simultaneously target multiple targets involved in the development of AD. In this review, we provide an overview of the pathogenesis of AD and the current drug therapies for the disease. Additionally, rationales of the multi-target approaches and examples of multi-target drugs with pharmacological actions against AD are also discussed.
Article
Full-text available
Accumulation of neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau is a histopathological hallmark of Alzheimer’s disease (AD) and related tauopathies. Growing evidence demonstrated that tau pathology in AD spreads in a prion-like manner. Previous studies showed that metformin might have a positive effect on cognition. However, the underlying mechanisms are still unknown. Therefore, the present study aimed to investigate the effects of metformin on tau propagation. Brain extracts containing tau aggregates were unilaterally injected into the hippocampus and the overlying cerebral cortex of PS19 mice. Metformin was administrated through drinking water for four months, and we observed tau spreading in the brain of tau-seeded PS19 mice. Metformin inhibited the spreading of tau pathology in the ipsilateral hemisphere, attenuated tau pathology in the contralateral hemisphere, and reduced the hyperphosphorylation of tau at Ser202/Thr205, Thr231, and Ser422 sites in the soluble fraction and Ser202/Thr205, Ser262, Thr396, Thr231, and Ser422 sites in the insoluble fraction of tau-seeded PS19 mice brains. Metformin did not affect tau kinases or phosphatase 2A protein levels but reduced mTORC1 protein levels. Additionally, metformin reduced learning and memory deficits of the tau-seeded PS19 mice. These findings indicate that metformin reduced tau hyperphosphorylation, attenuated tau pathology in tau-seeded PS19 mice, and improved learning and memory deficits. These findings highlight the potential mechanisms underlying the beneficial effects of metformin on cognition, implying that metformin could be a promising drug for the prevention and early treatment of AD. Supplementary Information The online version contains supplementary material available at 10.1007/s13311-022-01316-6.
Article
Full-text available
Background Neurite dystrophy is a pathologic hallmark of Alzheimer’s disease (AD). However, drug discovery targeting neurite protection in AD remains largely unexplored. Methods Aβ-induced neurite and mitochondrial damage assays were used to evaluate Aβ toxicity and the neuroprotective efficacy of a natural compound salidroside (SAL). The 5×FAD transgenic mouse model of AD was used to study the neuroprotective function of SAL. To verify the direct target of SAL, we used surface plasmon resonance and cellular thermal shift assays to analyze the drug-protein interaction. Results SAL ameliorates Aβ-mediated neurite damage in cell culture. We further reveal that SAL represses mitochondrial damage in neurites by promoting mitophagy and maintaining mitochondrial homeostasis, dependent on an NAD-dependent deacetylase SIRT3. In AD mice, SAL protects neurite morphology, mitigates Aβ pathology, and improves cognitive function, which are all SIRT3-dependent. Notably, SAL directly binds to transcription factor NRF2, inhibits its degradation by blocking its interaction with KEAP1 ubiquitin ligase, and then advances NRF2-mediated SIRT3 transcription. Conclusions Overall, we demonstrate that SAL, a potential anti-aging drug candidate, attenuates AD pathology by targeting NRF2/SIRT3 pathway for mitochondrial and neurite protection. Drug discovery strategies focusing on SAL may thus provide promising therapeutics for AD.
Article
Full-text available
Alzheimer’s disease (AD), is a progressive neurodegenerative disease that affects behavior, thinking, learning, and memory in elderly individuals. AD occurs in two forms, early onset familial and late-onset sporadic; genetic mutations in PS1, PS2, and APP genes cause early onset familial AD, and a combination of lifestyle, environment and genetic factors causes the late-onset sporadic form of the disease. However, accelerated disease progression is noticed in patients with familial AD. Disease-causing pathological changes are synaptic damage, and mitochondrial structural and functional changes, in addition to increased production and accumulation of phosphorylated tau (p-tau), and amyloid beta (Aβ) in the affected brain regions in AD patients. Aβ is a peptide derived from amyloid precursor protein (APP) by proteolytic cleavage of beta and gamma secretases. APP is a glycoprotein that plays a significant role in maintaining neuronal homeostasis like signaling, neuronal development, and intracellular transport. Aβ is reported to have both protective and toxic effects in neurons. The purpose of our article is to summarize recent developments of Aβ and its association with synapses, mitochondria, microglia, astrocytes, and its interaction with p-tau. Our article also covers the therapeutic strategies that reduce Aβ toxicities in disease progression and discusses the reasons for the failures of Aβ therapeutics.
Article
Full-text available
Neuronal death and synaptic loss are principal pathological features of Alzheimer's disease (AD). Amyloid beta oligomers (AβOs) constitute the main neurotoxin underscoring AD pathology. AβOs interact with N-methyl-D-aspartate receptors (NMDARs), resulting in neurotoxic events, including activation of apoptosis and synaptic impairment. Carnosic acid (CA), extracted from Salvia rosmarinus, has been verified its neuroprotective effects in AD. However, the precise mechanisms by which CA induces synaptic protection remain unclear. In this study, we established an in vitro AD model using SH-SY5Y human neuroblastoma cells. We observed that CA improved neuronal survival by suppressing apoptosis. Moreover, CA restored synaptic impairments by increasing expression levels of brain-derived neurotrophic factor (BDNF), postsynaptic density protein-95 (PSD-95), and synaptophysin (Syn). Furthermore, we found these protective effects were dependent on inhibiting the phosphorylation of NMDAR subtype 2B (NMDAR2B), which further suppressed calcium overload and promoted activation of the extracellular signal-regulated kinase (ERK)-cAMP response element-binding protein (CREB) pathway. Administration of N-methyl-D-aspartic acid (NMDA), an agonist of NMDARs, abolished these effects of CA. Our findings demonstrate that CA exerts neuroprotective effects in an in vitro model of AD by regulating NMDAR2B and its downstream cascades, highlighting the therapeutic potential of CA as a NMDARs-targeted candidate in the treatment of AD.
Article
Full-text available
Background Eleven tau immunoglobulin G (IgG) antibodies have entered clinical trials to treat tauopathies, including Alzheimer's disease, but it is unclear which IgG subclass/subtype has the ideal efficacy and safety profile. Only two subtypes, with or without effector function, have been examined in the clinic and not for the same tau antibody. The few preclinical studies on this topic have only compared two subtypes of one antibody each and have yielded conflicting results. Methods We selected two single domain antibodies (sdAbs) derived from a llama immunized with tau proteins and utilized them to generate an array of Fc-(sdAb)2 subclasses containing identical tau binding domains but differing Fc region. Unmodified sdAbs and their IgG subclasses were tested for efficacy in primary cultures and in vivo microdialysis using JNPL3 tauopathy mice. Findings Unmodified sdAbs were non-toxic, blocked tau toxicity and promoted tau clearance. However, the efficacy/safety profile of their Fc-(sdAb)2 subclasses varied greatly within and between sdAbs. For one of them, all its subtypes were non-toxic, only those with effector function cleared tau, and were more effective in vivo than unmodified sdAb. For the other sdAb, all its subtypes were toxic in tauopathy cultures but not in wild-type cells, suggesting that bivalent binding of its tau epitope stabilizes a toxic conformation of tau, with major implications for tau pathogenesis. Likewise, its subclasses were less effective than the unmodified sdAb in clearing tau in vivo. Interpretation These findings indicate that tau antibodies with effector function are safe and better at clearing pathological tau than effectorless antibodies, Furthermore, tau antibodies can provide a valuable insight into tau pathogenesis, and some may aggravate it. Funding Funding for these studies was provided by the National Institute of Health (R01 AG032611, R01 NS077239, RF1 NS120488, R21 AG 069475, R21 AG 058282, T32AG052909), and the NYU Alzheimer's Disease Center Pilot Grant Program (via P30 AG008051).
Article
Full-text available
Tumor necrosis factor-α (TNF-α) is a pleiotropic, proinflammatory cytokine related to different neurodegenerative diseases, including Alzheimer’s disease (AD). Although the linkage between increased TNF-α levels and AD is widely recognized, TNF-α–neutralizing therapies have failed to treat AD. Previous research has associated this with the antithetic functions of the two TNF receptors, TNF receptor 1, associated with inflammation and apoptosis, and TNF receptor 2 (TNFR2), associated with neuroprotection. In our study, we investigated the effects of specifically stimulating TNFR2 with a TNFR2 agonist (NewStar2) in a transgenic Aβ-overexpressing mouse model of AD by administering NewStar2 in two different ways: centrally, via implantation of osmotic pumps, or systemically by intraperitoneal injections. We found that both centrally and systemically administered NewStar2 resulted in a drastic reduction in amyloid β deposition and β-secretase 1 expression levels. Moreover, activation of TNFR2 increased microglial and astrocytic activation and promoted the uptake and degradation of Aβ. Finally, cognitive functions were also improved after NewStar2 treatment. Our results demonstrate that activation of TNFR2 mitigates Aβ-induced cognitive deficits and neuropathology in an AD mouse model and indicates that TNFR2 stimulation might be a potential treatment for AD.
Article
Background: The accumulation of soluble and insoluble aggregated amyloid-beta (Aβ) may initiate or potentiate pathologic processes in Alzheimer's disease. Lecanemab, a humanized IgG1 monoclonal antibody that binds with high affinity to Aβ soluble protofibrils, is being tested in persons with early Alzheimer's disease. Methods: We conducted an 18-month, multicenter, double-blind, phase 3 trial involving persons 50 to 90 years of age with early Alzheimer's disease (mild cognitive impairment or mild dementia due to Alzheimer's disease) with evidence of amyloid on positron-emission tomography (PET) or by cerebrospinal fluid testing. Participants were randomly assigned in a 1:1 ratio to receive intravenous lecanemab (10 mg per kilogram of body weight every 2 weeks) or placebo. The primary end point was the change from baseline at 18 months in the score on the Clinical Dementia Rating-Sum of Boxes (CDR-SB; range, 0 to 18, with higher scores indicating greater impairment). Key secondary end points were the change in amyloid burden on PET, the score on the 14-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog14; range, 0 to 90; higher scores indicate greater impairment), the Alzheimer's Disease Composite Score (ADCOMS; range, 0 to 1.97; higher scores indicate greater impairment), and the score on the Alzheimer's Disease Cooperative Study-Activities of Daily Living Scale for Mild Cognitive Impairment (ADCS-MCI-ADL; range, 0 to 53; lower scores indicate greater impairment). Results: A total of 1795 participants were enrolled, with 898 assigned to receive lecanemab and 897 to receive placebo. The mean CDR-SB score at baseline was approximately 3.2 in both groups. The adjusted least-squares mean change from baseline at 18 months was 1.21 with lecanemab and 1.66 with placebo (difference, -0.45; 95% confidence interval [CI], -0.67 to -0.23; P<0.001). In a substudy involving 698 participants, there were greater reductions in brain amyloid burden with lecanemab than with placebo (difference, -59.1 centiloids; 95% CI, -62.6 to -55.6). Other mean differences between the two groups in the change from baseline favoring lecanemab were as follows: for the ADAS-cog14 score, -1.44 (95% CI, -2.27 to -0.61; P<0.001); for the ADCOMS, -0.050 (95% CI, -0.074 to -0.027; P<0.001); and for the ADCS-MCI-ADL score, 2.0 (95% CI, 1.2 to 2.8; P<0.001). Lecanemab resulted in infusion-related reactions in 26.4% of the participants and amyloid-related imaging abnormalities with edema or effusions in 12.6%. Conclusions: Lecanemab reduced markers of amyloid in early Alzheimer's disease and resulted in moderately less decline on measures of cognition and function than placebo at 18 months but was associated with adverse events. Longer trials are warranted to determine the efficacy and safety of lecanemab in early Alzheimer's disease. (Funded by Eisai and Biogen; Clarity AD ClinicalTrials.gov number, NCT03887455.).
Article
With the exception of a few monogenic forms, Alzheimer disease (AD) has a complex aetiology that is likely to involve multiple susceptibility genes and environmental factors. The role of environmental factors is difficult to determine and, until a few years ago, the molecular mechanisms underlying gene-environment (G × E) interactions in AD were largely unknown. Here, we review evidence that has emerged over the past two decades to explain how environmental factors, such as diet, lifestyle, alcohol, smoking and pollutants, might interact with the human genome. In particular, we discuss how various environmental AD risk factors can induce epigenetic modifications of key AD-related genes and pathways and consider how epigenetic mechanisms could contribute to the effects of oxidative stress on AD onset. Studies on early-life exposures are helping to uncover critical time windows of sensitivity to epigenetic influences from environmental factors, thereby laying the foundations for future primary preventative approaches. We conclude that epigenetic modifications need to be considered when assessing G × E interactions in AD.