ArticlePDF Available

Impaired Adult Neurogenesis in the Dentate Gyrus of a Triple Transgenic Mouse Model of Alzheimer's Disease

PLOS
PLOS ONE
Authors:

Abstract and Figures

It has become generally accepted that new neurones are added and integrated mainly in two areas of the mammalian CNS, the subventricular zone and the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus, which is of central importance in learning and memory. The newly generated cells display neuronal morphology, are able to generate action potentials and receive functional synaptic inputs, i.e. their properties are similar to those found in mature neurones. Alzheimer's disease (AD) is the primary and widespread cause of dementia and is an age-related, progressive and irreversible neurodegenerative disease that deteriorates cognitive functions. Here, we have used male and female triple transgenic mice (3xTg-AD) harbouring three mutant genes (beta-amyloid precursor protein, presenilin-1 and tau) and their respective non-transgenic (non-Tg) controls at 2, 3, 4, 6, 9 and 12 months of age to establish the link between AD and neurogenesis. Using immunohistochemistry we determined the area density of proliferating cells within the SGZ of the DG, measured by the presence of phosphorylated Histone H3 (HH3), and their possible co-localisation with GFAP to exclude a glial phenotype. Less than 1% of the HH3 labeled cells co-localised with GFAP. Both non-Tg and 3xTg-AD showed an age-dependent decrease in neurogenesis. However, male 3xTg-AD mice demonstrated a further reduction in the production of new neurones from 9 months of age (73% decrease) and a complete depletion at 12 months, when compared to controls. In addition, female 3xTg-AD mice showed an earlier but equivalent decrease in neurogenesis at 4 months (reduction of 63%) with an almost inexistent rate at 12 months (88% decrease) compared to controls. This reduction in neurogenesis was directly associated with the presence of beta-amyloid plaques and an increase in the number of beta-amyloid containing neurones in the hippocampus; which in the case of 3xgTg females was directly correlated. These results suggest that 3xTg-AD mice have an impaired ability to generate new neurones in the DG of the hippocampus, the severity of which increases with age and might be directly associated with the known cognitive impairment observed from 6 months of age onwards . The earlier reduction of neurogenesis in females, from 4 months, is in agreement with the higher prevalence of AD in women than in men. Thus it is conceivable to speculate that a recovery in neurogenesis rates in AD could help to rescue cognitive impairment.
Content may be subject to copyright.
Impaired Adult Neurogenesis in the Dentate Gyrus of a
Triple Transgenic Mouse Model of Alzheimer’s Disease
Jose
´J. Rodrı
´guez
1
*, Victoria C. Jones
1
, Masashi Tabuchi
1
, Stuart M. Allan
1
, Elysse M. Knight
1
, Frank M.
LaFerla
2
, Salvatore Oddo
2
, Alexei Verkhratsky
1,3
1Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom, 2Department of Neurobiology and Behaviour, University of California Irvine, Irvine,
California, United States of America, 3Institute of Experimental Medicine, ASCR, Prague, Czech Republic
Abstract
It has become generally accepted that new neurones are added and integrated mainly in two areas of the mammalian CNS, the
subventricular zone and the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus, which is of central
importance in learning and memory. The newly generated cells display neuronal morphology, are able to generate action
potentials and receive functional synaptic inputs, i.e. their properties are similar to those found in mature neurones. Alzheimer’s
disease (AD) is the primary and widespread cause of dementia and is an age-related, progressive and irreversible
neurodegenerative disease that deteriorates cognitive functions. Here, we have used male and female triple transgenic mice
(3xTg-AD) harbouring three mutant genes (b-amyloid precursor protein, presenilin-1 and tau) and their respective non-
transgenic (non-Tg) controls at 2, 3, 4, 6, 9 and 12 months of age to establish the link between AD and neurogenesis. Using
immunohistochemistry we determined the area density of proliferating cells within the SGZ of the DG, measured by the
presence of phosphorylated Histone H3 (HH3), and their possible co-localisation with GFAP to exclude a glial phenotype. Less
than 1% of the HH3 labeled cells co-localised with GFAP. Both non-Tg and 3xTg-AD showed an age-dependent decrease in
neurogenesis. However, male 3xTg-AD mice demonstrated a further reduction in the production of new neurones from
9 months of age (73% decrease) and a complete depletion at 12 months, when compared to controls. In addition, female 3xTg-
AD mice showed an earlier but equivalent decrease in neurogenesis at 4 months (reduction of 63%) with an almost inexistent
rate at 12 months (88% decrease) compared to controls. This reduction in neurogenesis was directly associated with the
presence of b-amyloid plaques and an increase in the number of b-amyloid containing neurones in the hippocampus; which in
the case of 3xgTg females was directly correlated. These results suggest that 3xTg-AD mice have an impaired ability to generate
new neurones in the DG of the hippocampus, the severity of which increases with age and might be directly associated with the
known cognitive impairment observed from 6 months of age onwards . The earlier reduction of neurogenesis in females, from
4 months, is in agreement with the higher prevalence of AD in women than in men. Thus it is conceivable to speculate that a
recovery in neurogenesis rates in AD could help to rescue cognitive impairment.
Citation: Rodrı
´guez JJ, Jones VC, Tabuchi M, Allan SM, Knight EM, et al. (2008) Impaired Adult Neurogenesis in the Dentate Gyrus of a Triple Transgenic Mouse
Model of Alzheimer’s Disease. PLoS ONE 3(8): e2935. doi:10.1371/journal.pone.0002935
Editor: Katrina Gwinn, Baylor College of Medicine, United States of America
Received March 24, 2008; Accepted July 21, 2008; Published August 13, 2008
Copyright: ß2008 Rodrı
´guez Arellano et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: Alzheimer’s Research Trust Programme Grant (ART/PG2004A/1) to JJR and AV.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: Jose.Rodriguez-arellano@manchester.ac.uk
Introduction
The classical view that all neurones are generated (via neurogen-
esis) during prenatal development and early postnatal life has been
challenged by the seminal study of Altman and Das (1965) [1] and
now it is generally accepted that neurogenesis does also occur in
adulthood mainly in two areas of the mammalian CNS [1–5]. These
areas, which are involved in both plasticity and stability of the brain,
are the anterior part of the subventricular zone (SVZ) along the
lateral ventricles, which is also an important a site of gliogenesis [6,7]
and the subgranular zone (SGZ) of the dentate gyrus (DG) of the
hippocampus [4,5]. In both areas neurogenesis progress as a complex
multi-step process which starts with the proliferation of precursors
residing in the SVZ or in the SGZ. For the hippocampus, it has been
estimated that several thousand new cells are generated daily [5].
However, within several days after their birth at least fifty percent of
the newborn cells die [5]. The cells surviving this initial period of cell
death differentiate mainly into granule neurones and endure for
several months. These newly generated neurones receive synaptic
inputs, extend axons along the mossy fibres tract and exhibit
electrophysiological properties similar to those of mature dentate
granule cells [8,9]. In addition these new cells express a full
complement of membrane receptors [10]. From a functional point
of view, hippocampal neurogenesis plays an important role in
memory processes. Decline in neurogenesis within SGZ has been
involved in cognitive impairments linked with ageing and neurode-
generative disorders, and was suggested to play a role in Alzheimer
disease (AD) [5,11].
AD is a progressive neurodegenerative disease which is the
primary cause of dementia in the elderly and is characterized by
damage of the brain regions associated with learning and memory,
such as the hippocampus [12,13]. Decline in neurogenic capacity
could participate in AD-associated cognitive impairments and
contribute to early AD symptoms such as the inability to acquire
and store new information [14–16]. Incidentally, the use of
endogenous neuronal precursors to replace lost and/or damaged
cells has been proposed as a potential therapeutic approach to
treat AD [17,18]. Experimental studies of neurogenesis in various
PLoS ONE | www.plosone.org 1 August 2008 | Volume 3 | Issue 8 | e2935
AD animal models, however, resulted in contradictory findings
[19–26]. For example, animals carrying presenilin or some APP
mutant genes demonstrated an impaired neurogenesis in the DG
[20–22,24,27,28]. Conversely, recent studies, performed on the
APP
Sw, Ind
(Swiss/Indian mutation) PDGF-APP mutant and on
FAD post-mortem human material [18,29], reported an increase
of neurogenesis. It has to be noted, though, that the study on
human FAD cases analysed only immature newly generated
neurones without providing definitive probes of further develop-
ment and/or progress into mature cells.
In the present study we sought to determine the rate and possible
changes in hippocampal neurogenesis in the recently developed
triple-transgenic AD (3xTg-AD) mouse model, that harbours the
mutant genes for amyloid precursor protein (APP
Swe
), for presenilin
1PS1
M146V
and for tau
P301L
[30,31]. These animals are recognised
as relevant AD model since they show temporal- and region-specific
Aband tau pathology, which closely resembles that seen in the
human AD brain [30,31]. As well as progressively developing
plaques and tangles the 3xTg-AD mice also show clear functional
and cognitive impairments including LTP, spatial memory and long
term memory deficits; which are manifest in an age-related manner
importantly preceding the appearance of histological markers
[30,31]. Cognitive deficits in the 3xTg-AD model correlate with
the accumulation of intraneuronal Ab[30–34]. Subsequently, we
decided to investigate a gender difference in the number of
proliferating cells, because it is well establish that AD affects women
more than men [35,36]. Finally we also aimed to correlate the rate of
neurogenesis with the presence of intracellular b-amyloid.
Materials and Methods
Mice
All animal procedures were performed according to the Animal
Scientific Procedures Act of 1986 under the license from the
United Kingdom Home Office.
The generation of the 3xTg-AD mice was done as previously
described [30,31,37]. Briefly, human amyloid precursor protein
with the Swedish mutation (APP
Swe
) and human tau with the
P301L (tau
P301L
) mutation were microinjected into single-cell
embryos from homozygous presenilin 1PS1
M146V
knockin mice.
The background of the PS1 knockin mouse is a hybrid 129/
C57BL6. The Non-Tg mice used were from the same strain and
genetic background as the PS1 knockin mice, but they harbor the
endogenous wild-type mouse PS1. All 3xTg-AD and Non-Tg mice
were obtained from crossing homozygous breeders. Male and
female mice were independently group housed and kept on daily
12 h light-dark cycles dark schedule. All mice were given ad
libitum access to food and water.
Fixation and tissue processing
Male and female 3xTg-AD and their respective non-transgenic
(non-Tg) controls were anaesthetized with an intraperitoneal
injection of sodium pentobarbital at different time points (2, 3, 4,
6, 9 and 12 months of age; n = 3–7). The brains were fixed by
perfusion through the aortic arch with 25 ml of 3.8% acrolein
(TAAB, UK) in a solution of 2% paraformaldehyde and 0.1 M
phosphate buffer (PB) pH 7.4, followed by 75 ml of 2%
paraformaldehyde. Brains were removed from the cranium and
cut into 4–5 mm coronal slabs of tissue containing the entire
rostrocaudal extent of the hippocampus. This tissue was then post-
fixed for 30 minutes in 2% paraformaldehyde and sectioned at
40–50 mm on a vibrating microtome (VT1000, Leica, Milton
Keynes, UK). To remove excess reactive aldehyde groups, sections
were treated with 1% sodium borohydride in 0.1 M PB for
30 minutes. The tissue sections were then freeze-thawed to
optimize the penetration of immunoreagents. For this procedure,
sections were incubated in cryoprotectant solution containing 25%
sucrose and 3.5% glycerol in 0.05 M PB at pH 7.4 and
subsequently rapidly immersed in chlorodifluoromethane followed
by liquid nitrogen and then thawed at room temperature in PB.
Sections were then rinsed in 0.1 M PB followed by 0.1 M Tris-
buffered saline (TBS), pH 7.6.
Antibodies
A polyclonal affinity-purified rabbit antiserum raised against
phosphorylated Histone 3 (Upstate, USA; #06-570) and a
monoclonal mouse antiserum generated against GFAP from pig
spinal cord (Sigma-Aldrich Company Ltd., UK; #G3893) were used
for the determination of proliferating cells and glia. Specificity of
these antisera was confirmed by immunoblot and western blot [38].
For identification of intracellular beta amyloid (Ab)depositsweused
a monoclonal mouse antiserum that reacts with abnormally
processed isoforms, as well as precursor forms of Ab,recognizing
an epitope within amino acids 3–8 (EFRHDS; anti-Ab 6E10 [SIG-
39320]. Signet Laboratories, Dedham, MA).The immunolabelling
pattern we obtained with this antibody is equivalent to that obtained
previously in different brain regions [30,31].
To assess for non-specific background labelling or cross reactivity
between antibodies derived from different host species, a series of
control experiments were performed. Omission of primary and/or
secondary antibodies from the incubation solutions resulted in a total
absence of target labelling. These primary antibodies are therefore
regarded as specific to their designated targets.
Immunohistochemistry
To optimize detection of all HH3 and GFAP cells and
containing profiles we used the highly sensitive avidin–biotin
peroxidase complex (ABC) method [39]; and to minimize
methodological variability, sections through the dorsal hippocam-
pus containing both hemispheres of all animals were processed at
the same time using precisely the same experimental conditions.
For this procedure, the vibratome sections were first incubated for
30 minutes in 0.5% bovine serum albumin in TBS to minimize
non-specific labelling. The tissue sections were then incubated for
48 hours at 4uC in 0.1% bovine serum albumin in TBS
containing: (1) rabbit polyclonal antiserum for HH3 (1:1,000)
and (2) mouse monoclonal antiserum for GFAP (1:60,000).
Subsequently, the HH3 and GFAP antibodies were detected in
a sequential manner on the same sections. For HH3 labelling,
sections were washed and placed in (1) 1:200 dilutions of
biotinylated donkey anti-rabbit IgG (Jackson Immunoresearch,
Stratech Scientific Ltd., Soham, UK) and (2) 1:200 dilutions of
biotin-avidin complex from the Elite kit (Vector Laboratories Ltd.,
Peterborough, UK). All antisera dilutions were prepared in TBS,
and the incubations were carried out at room temperature. The
peroxidase reaction product was visualized by incubation in a
solution containing 0.022% of 3,39diaminobenzidine (DAB,
Aldrich, Gillingham, UK) and 0.003% H
2
O
2
in TBS for
6 minutes. For GFAP labelling, sections were then rinsed again
in TBS and incubated (1) 1:200 dilution of biotynilated horse anti-
mouse IgG (1:200; Vector Laboratories Ltd., Peterborough, UK)
and (2) placed in a 1:200 dilution of biotin-avidin complex from
the Elite kit (Vector Laboratories Ltd., Peterborough, UK). The
GFAP peroxidase product was then visualized in a solution
prepared from the Novared or SGZ kits (Vector Laboratories Ltd.,
Peterborough, UK) for 3–4 minutes. This allowed us to see the
GFAP labelling in red and/or blue respectively; allowing us to
differentiate it from the HH3 labelled cells (brown).
Neurogenesis Reduction in AD
PLoS ONE | www.plosone.org 2 August 2008 | Volume 3 | Issue 8 | e2935
The same immunoperoxidase approach, but for single labelling,
was used for the detection of intracellular Ab. Briefly, adjacent
sections were incubated for 48 hours at 4uCin0.1%bovineserum
albumin in TBS containing mouse monoclonal antiserum for Ab
(1:2000). Subsequently, sections were then washed and placed in (1)
1:200 horse anti-mouse IgG (Vector Laboratories Ltd., Peterbor-
ough, UK) and (2) 1:200 dilution of biotin-avidin complex from the
Elite kit (Vector Laboratories Ltd., Peterborough, UK). The
peroxidase reaction product was visualized by incubation in a
solution containing 0.022% of 3,39diaminobenzidine (DAB,
Aldrich, Gillingham, UK) and 0.003% H
2
O
2
in TBS for 6 minutes.
HH3 Area density and Abcell number
To determine the area density (Sv, number/mm
2
) of HH3-
immunoreactive neurons, the labelled cells were counted on both
hemispheres in six non-consecutive coronal Vibratome sections,
separate by at least 80 mm, taken through representative sections
of both the dorsal (3) and ventral (3) DG of the hippocampus at
Figure 1. Photomicrographs showing phosphorilated Histone H3 (HH3, a proliferating mitotic marker) within the dentate gyrus of
Non Tg mice. A–B: Single labelling of HH3 positive cells (arrows) in the dentate gyrus of 2 (A) and 12 months (B) Non Tg mice. C–D: Dual labeling
of HH3 positive cells (arrows) and glial cells (GFAP, blue) in the dentate gyrus of 2 (C) and 12 months (D) Non Tg mice. E–F: Bar graphs showing the
area density of HH3 positive cells within the dorsal dentate gyrus (all layers included) of Non-Tg males (E) and females (F) mice. GCL: Granular Cell
Layer, ML: Molecular Layer. ** =p,0.01 compared to 2 months; *** =p,0.001 compared to 2 months;
NN
=p,0.01 compared to 3 months;
## =p,0.01 compared to 2 and 3 months; ### =p,0.001 compared to 3 months; e=p,0.05 compared to 4 months; ee =p,0.01 compared
to 4 months; =p,0.05 compared to 4 and 6 months; +=p,0.05 compared to 2, 3 and 6 months.
doi:10.1371/journal.pone.0002935.g001
Neurogenesis Reduction in AD
PLoS ONE | www.plosone.org 3 August 2008 | Volume 3 | Issue 8 | e2935
levels 1.22 mm/2.46 mm and 2.54 mm/3.80 mm posterior
anterior to bregma, respectively, according to the mouse brain
atlas of Paxinos and Watson (1986) [40]. The number of HH3
positive cells and the area measurements of the complete dentate
gyrus and its different layers (granule cell layer –GCL-, molecular
layer –ML- and hilus) were determined blindly.
The number of Abcontaining neurones was examined in the CA1
region of the hippocampus CA1, since this field shows the earliest
and strongest accumulation of Abintracellular deposits. This
quantification was carried out on six non-consecutive hippocampal
sections of the same animals used for the proliferation analysis.
Statistical analysis
An analysis of variance (ANOVA) was used to examine differences
in the mean area density of labelled HH3 cells between the 3xTg-AD
and non-Tg animals and sexes, followed by unpaired t-test
comparisons at the different time points. Spearman correlation
was used to correlate the mean area density of HH3 positive cells
with the mean number of Abcontaining neurons (implemented
through GraphPad Prism 4.0, GraphPad Software, Inc.).
Results
In the dorsal hippocampus and more specifically within the
dentate gyrus of both non-Tg and 3xTg-AD mice a fair number of
newly generated cells could be visualized, as indicated by HH3
immunoreactivity (HH3-IR; Figs.1A, 2A). These newly formed
cells showed the distinctive characteristics of proliferating cells;
they were mainly localized in the inferior part of the granule cell
layer (GCL) and demonstrated typical morphology such as
irregular shape and small size; sometimes they appeared close
together and/or formed clusters (Fig. 1A–B).
Effects of ageing on neurogenesis in non-Tg animals
Quantitative analysis of the rate of cell proliferation showed a
reduction in the number of HH3-IR cells with age. This reduction
was apparent within the dentate gyrus of non-Tg males and
females mice (F
5,20
= 5.643, p = 0.0021 and F
5,16
= 44.8,
p,0.0001, respectively; Fig 1E–F) and in the GCL where we
observed the highest number of proliferating cells (F
5,20
= 5.362,
p = 0.0028 and F
5,16
= 17.54, p,0.0001, respectively; Fig 3A–B).
Decrease in proliferation rate with age was quite significant: at
6 months of age in both sexes the proliferation rate was reduced
by more than 60% when compared to the 2 months old animals
(males 2.6460.23 vs. 6.6161.74; females 1.4860.39 vs.
7.4660.45, Fig. 1E–H). Decrease in the number of HH3 cells in
dentate gyrus and the subgranular zone of the GCL appears
earlier in females (3–4 month) than in males as confirmed by the t-
test analysis (Figs. 1E–F; Fig. 3).
Figure 2. Brightfield micrographs showing HH3 labelled cells within the dentate gyrus of 3xTg-AD e. A–B: Dual labeling of HH3 positive
cells (arrows) and glial cells (GFAP, red) in the dentate gyrus of 2 (A) and 12 months (B) 3xTg-AD mice. C–D: Bar graphs showing the area density of
HH3 positive cells within the dorsal dentate gyrus (all layers included) of 3xTg-AD males (C) and females (D) mice. GCL: Granular Cell Layer, ML:
Molecular Layer. ** =p,0.01 compared to 3 months; *** =p,0.001 compared to 3 months;
N
=p,0.05 compared to 2 months
NN
=p,0.001
compared to 2 months; ## =p,0.01 compared to 2 and 3 months; e=p,0.05 compared to 4 months; =p,0.05 compared to 2 and 4 months.
doi:10.1371/journal.pone.0002935.g002
Neurogenesis Reduction in AD
PLoS ONE | www.plosone.org 4 August 2008 | Volume 3 | Issue 8 | e2935
Dual labelling showed that the majority of HH3-IR cells did not
possess the astroglia marker GFAP; in fact, less then 1% of HH3-
IR cells co-expressed GFAP in the dentate gyrus, including the
GCL (data not shown).
Impairment of neurogenesis in 3xTg-mice
Similarly to the controls, the rate of neurogenesis in 3xTg-AD
mice was decreased with age (Fig. 2, 3) in both males and females
throughout the dentate gyrus (F
5,20
=5.437, p=0.0026 and
F
5,18
=12.39, p,0.0001, respectively) and in the GCL
(F
5,20
= 8.524, p = 0.0002 and F
5,18
=11.81, p,0.0001, respectively;
Figs 2C–D). Decrease in proliferation rate in 3xTg-AD animals was
about 60–90% more pronounced compared to control animals: at
6 months compared to the 2 months 3xTg-AD mice (males
2.2360.5 vs. 5.7161.47; females 0.47+0.23 vs 5.0961) Fig. 2 C–
D). Furthermore, at 12 months of age males and females showed
very little capacity of forming new cells within the GCL (Fig. 2 C–D;
Fig. 3); whilst in both males and females non-Tg animals we could
still observe approximately a 20–35% of the number of HH3-IR
observed at young ages (2–3 months ; Fig 3).
Neurogenesis depression in 3xTg-mice is gender
dependent
When we analysed the number of HH3-IR cells within the GCL
of the 3xTg-AD mice and compared it with the cell proliferation
rate of the non-Tg it became evident that at young ages (2 and
3 months) the levels were very similar whilst at older ages,
especially 9 and 12 months the neurogenesis levels have decreased
over 70% in both groups (Figs. 1 E–F, 2 C–D, 3). The
quantification and consequent statistical analysis showed that the
age associated reduction in HH3-IR cells in 3xTg-AD males
compared to non-Tg animals start to be significant at 9 months
(73%; showing a trend to significant difference) and is completely
disappeared at 12 months of age (Fig. 3A; age effect, F
5,1
= 53.57,
p,0.0001; group effect F
5,1
= 1.73, p = 0.1744; age x group effect
F
5,1
= 3.75, p = 0.5353; p,0.05 at 12 months). In contrast in
females 3xTg-AD, compared to non-Tg animals, HH3-IR cells
were already significantly reduced at 4 months of age (63%) being
maximal (88% reduction) at 12 months (Fig. 3B; age effect,
F
5,1
= 68.08, p,0.0001; group effect F
5,1
= 10.31, p,0.0001; age
x group effect F
5,1
= 2.31, p = 0.4961; p,0.05 at 4 and 6 months;
p,0.001 at 9 and 12 months). This impairment in neurogenesis
rate within the 3xTg-AD mice is mainly due to changes at dorsal
more than ventral hippocampal levels (Fig. 3), which is consistent
with their specific preferential roles in learning and memory and
affective behaviour respectively [41].
Relationship between intraneuronal b-amyloid
accumulation and neurogenesis
The 3xTg-AD mice had also an age-dependent increase in the
number of hippocampal neurones accumulating b-amyloid
(Fig. 4A–D). Within the hippocampus neurones containing b-
amyloid could be found as early as 2 months of age, and this
number was higher in females compared to males (Fig. 4C–D).
Quantitative analysis showed that in both males and females
3xTg-Ad mice there was an age-dependent increase of b-amyloid
containing neurones that was maximal and very significant at
9 months of age when compared to young animals (Fig. 4 C–D;
F
4,10
= 4.231, p = 0.0293 and F
4,10
= 4.948, p = 0.0184, respec-
tively). The fully formed b-amyloid plaques within the neuropil,
however, were observed much later, at 9 and 12 months (Fig. 4B).
Increase in the number of b-amyloid containing hippocampal
neurones seem to match with the reduction of GCL proliferating
cells in both males and females (Fig. 4E–F). However, only females
showed a significant indirect correlation between the number of
HH3-IR cells with the number of b-amyloid positive cells
Figure 3. Bar graphs showing the mean area density HH3 labelled cells within the GCL of the dentate gyrus of both 3xTg-AD and
control nonTg-AD mice. A–B: Males (A) and females (B) dorsal GCL. C–D: Males (C) and females (D) dorsal GCL. Asterisks indicate a significant
difference in the means.
doi:10.1371/journal.pone.0002935.g003
Neurogenesis Reduction in AD
PLoS ONE | www.plosone.org 5 August 2008 | Volume 3 | Issue 8 | e2935
(R
2
= 0.7018; Fig 4F). This finding showing a higher prevalence of
b-amyloid positive cells in 3xTg-AD females is also in agreement
with the recently reported 20% increase in plaque load observed in
APP23 transgenic mice females when compared to males [42].
Furthermore,The presence of high number of b-amyloid contain-
ing neurones and occurrence of plaques at 12 months were
occasionally concomitant with the first appearance of phosphori-
lated Tau (Fig. 4G).
Figure 4. Photomicrographs showing the presence of b-amyloid within the pyramidal neurones of CA1 as well as the presence of a
plaque in 12 months 3xTg-AD mice (B) compared to a nonTg control animal (A). C–D: Bar graphs showing the number of cells containing
b-amyloidin the hippocampal CA1 of males (C) and females (D) 3xTg-AD mice. E–F: Linear correlations between the mean number of cells containing
b-amyloid in the hippocampal CA1 and the mean area density of HH3 positive cells in the GCL of the dentate gyrus of males (E) and females (F) 3xTg-
AD mice. In Gwe can see the accumulation phosphorilated Tau within the CA1 of a 3xTg-AD mice. Or: CA1 Stratum Oriens, Py: CA1 stratum
Pyramidale, Rad: CA1 Stratum Radiatum, LMol: CA1 Stratum Lacunosum Moleculare. *=p,0.05 compared to 2 months; ** =p,0.01 compared to
2 months; =p,0.05 compared to 3 months.
doi:10.1371/journal.pone.0002935.g004
Neurogenesis Reduction in AD
PLoS ONE | www.plosone.org 6 August 2008 | Volume 3 | Issue 8 | e2935
Discussion
In the present study we have used single and dual labelling
immunohistochemistry to demonstrate changes in cell prolifera-
tion and neurogenesis within the hippocampal dentate gyrus of an
AD animal model. The experiments were performed on a recently
developed 3xTg-AD mouse model, which is recognised as an
extremely relevant, since these transgenic animals show temporal-
and region-specific Aband tau pathology, which closely resembles
that seen in the human AD brain [30,31]. These pathological
hallmarks are concomitant with clear functional and cognitive
impairments including LTP, spatial memory and long term
memory deficits, which are manifest in an age-related manner
[30–34]. We found that only a very small proportion of HH3-IR
proliferating cells in either 3xTg-AD or Non-Tg mice express
GFAP (,1%), suggesting that those HH3-IR cells are likely of a
neuronal lineage and thus are an indicator of neurogenesis.
Our main findings are that 3xTg-AD mice have a decreased GCL
neurogenesis, which is also is gender dependent. As happens in
normal rodents (including our control non-Tg animals) the decrease
in the dentate gyrus GCL neurogenesis develops with age [5,14]
However, we found that this effect is much exacerbated in 3xTg-AD,
being at least 60% stronger than in normal animals. These results are
in agreement with findings in other transgenic models of AD in
which transgenic mice having mutant forms of APP or presenilin-1
demonstrated impaired neurogenesis [20–22,24,27,28]. Since none
of models used previously fully reproduces the features of familial
and/or sporadic AD, our findings made in the 3xTg-AD mice
become of major relevance and importance.
We also demonstrated that the impairment of neurogenesis in the
dentate gyrus of the hippocampus is closely associated with AD
pathogenesis. Indeed hippocampus is affected early in AD; impaired
memory related to hippocampal damage may be associated with
deregulations of neurogenesis [5,11,29,43]. Two previous studies,
however, are in contradiction with our findings; one performed on
the APP
Sw, Ind
(Swiss/Indian mutation) mutant and one on FAD
post-mortem human material, reporting an increase of neurogenesis
[18,29]. These discrepancies could be explained by methodological
and preservation differences, including the post-mortem fixation
delay which could contribute to antigenmasking and in consequence
a misevaluation of the proliferation rates [44]. In addition, in these
studies the Doublecortin (Dcx) labelling, which marks both young
and immature neurons was used. This could affect the data
interpretation because more than 50% of the newborn cells die
[5]. Another discrepancy may reside in the fact that even if they use
5-Bromo-29-Deoxyuridine (BrdU) as an accurate index to label
proliferating cells, very few of them survives to 4 weeks [28].
Furthermore, these methods suffer from uncertainty since they may
not detect a distinct proliferative state but instead mark repaired
DNA in post-mitotic neurons and/or an abortive cell cycle [45,46].
In this study we also investigated the gender difference of the rate
of neurogenesis in 3xTg-AD mice. We found that neurogenesis in
female 3xTg-AD mice is affected earlier; already at 4 months of age
we found significant depression of neurogenesis in female AD
animals. This difference is in line with the recently reported sexual
dimorphism observed in cognitive performance such as the Morris
water-maze in which female 3xTg-AD mice also perform worse than
males [37]. In addition, it correlates to the well known fact, that AD
affects women earlier and with more severity than men (according to
some findings the incidence of the disease is double in females
[35,36]. Importantly such a gender-based predisposition toward
females is specific of AD and not found in other dementias [36].
Several lines of evidence suggest that this prevalence is directly
related with the circulating levels of estrogens [35,36,47]. As a result
females have higher levels of cell proliferation, but not cell survival
when compared with males cell proliferation rate in turn depends on
the endocrine status [48]. Only proestrus females, with high levels of
estradiol, show higher levels of cell proliferation; which seems to be
mediated through its effect on estrogen receptors [34,48,49]. Be it all
as it may, all these results are in agreement with our findings of a
greater degree of Abpathology in female versus male 3xTg-AD
animals from 4 months of age; this is also in agreement with recent
results observed in anothere AD transgenic mouse model (APP23) as
well as in line with clinical evidence of higher prevalence of AD in
females [35,36,42,50,51]. However, future studies are needed to
confirm this estrogen active role on cell proliferation and
neurogenesis whilst lately it has been shown controversial evidence
in some mouse strains, such as C57BL/6, in which female
hippocampal cell proliferation is not influenced by estrous cycle or
ovariectomy [52].
Generation of new neurones is an important feature of the adult
brain; in hippocampus the newborn neuronal cells, governed by
multiple factors, undergo complex stages of morphological and
functional maturation and integrate into existing neural circuitry
[53]. The hippocampal neurogenesis can be directly involved in
variety of cognitive processes; decreased neurogenesis negatively
affects certain learning and memory processes such as spatial
memory [51,53]. In contrast, increased load on the cognitive
processes (e.g. enriched environment) and physical exercises
positively affect neurogenesis [54]. On the other hand the role of
impaired neurogenesis in cognitive deficits in neurodegenerative
diseases is much less characterized. However, our observations
showing an impairment in neurogenesis correlate with the recent
evidence of age dependent impairment in spatial and long-term
memory tasks observed in the 3xTg-AD animals [32–34]; suggesting
a critical implication of neurogenesis in cognitive deficits
Thus, we consider that our data are specifically important as we
directly addressed the contradicting issue of the neurogenesis status
in the AD. By using longitudinal study on newly developed
transgenic model of the disease we demonstrated clear inhibition
of neurogenesis in diseased animals; this inhibition is specifically
pronounced in females, which correlates with clinical observations.
Therefore we conclude that inhibited neurogenesis can play an
active role in development of cognitive deficits and progression of
the AD.
Acknowledgments
The authors would like to thank Mr. Harun Noristani and Markel
Olabarria for their help editing this manuscript.
Author Contributions
Conceived and designed the experiments: JJRA. Performed the experi-
ments: JJRA MT EMK. Analyzed the data: JJRA VCJ EMK. Contributed
reagents/materials/analysis tools: SMA FML SO AV. Wrote the paper:
JJRA AV.
References
1. Altman J, Das GD (1965) Autoradiographic and histological evidence of
postnatal hippocampal neurogenesis in rats. J Comp Neurol 124: 319–335.
2. Ramo´ n y Cajal S (1913) Estudios sobre la degeneracio´n y regeneracio´ n del
sistema nervioso. Madrid: Imprenta de Hijos de Nicola´s Moya.
3. Gross CG (2000) Neurogenesis in the adult brain: death of a dogma. Nat Rev
Neurosci 1: 67–73.
4. Taupin P, Gage FH (2002) Adult neurogenesis and neural stem cells of the
central nervous system in mammals. J Neurosci Res 69: 745–749.
Neurogenesis Reduction in AD
PLoS ONE | www.plosone.org 7 August 2008 | Volume 3 | Issue 8 | e2935
5. Abrous DN, Koehl M, Le Moal M (2005) Adult neurogenesis: from precursors to
network and physiology. Physiol Rev 85: 523–569.
6. Paterson JA, Privat A, Ling EA, Leblond CP (1973) Investigation of glial cells in
semithin sections. 3. Transformation of subependymal cells into glial cells, as
shown by radioautography after 3 H-thymidine injection into the lateral
ventricle of the brain of young rats. J Comp Neurol. 149: 83–102.
7. Levison SW, Goldman JE (1993) Both oligodendrocytes and astrocytes develop
from progenitors in the subventricular zone of postnatal rat forebrain. Neuron.
10: 201–12.
8. Kempermann G, Gage FH (1999) New nerve cells for the adult brain. Sci Am
280: 48–53.
9. van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, et al. (2002)
Functional neurogenesis in the adult hippocampus. Nature 415: 1030–1034.
10. Mayo W, Lemaire V, Malaterre J, Rodriguez JJ, Cayre M, et al. (2005)
Pregnenolone sulfate enhances neurogenesis and PSA-NCAM in young and
aged hippocampus. Neurobiol Aging 26: 103–114.
11. Tatebayashi Y, Lee MH, Li L, Iqbal K, Grundke-Iqbal I (2003) The dentate
gyrus neurogenesis: a therapeutic target for Alzheimer’s disease. Acta
Neuropathol 105: 225–232.
12. Price DL, Struble RG, Whiteouse PJ, Kitt CA, Cork LC, et al. (1986)
Alzheimer’s disease: a multisystem disorder. Res Publ Assoc Res Nerv Dis 64:
209–214.
13. Braak R, Braak H (1991) Neuropathological stageing of Alzheimer-related
changes. Acta Neuropathol 82: 239–259.
14. Kuhn HG, Winkler J, Kempermann G, Thal LJ, Gage FH (1997) Epidermal
growth factor and fibroblast growth factor-2 have different effects on neural
progenitors in the adult rat brain. J Neurosci 17: 5820–5829.
15. Kempermann G, Kuhn HG, Gage FH (1998) Experience-induced neurogenesis
in the senescent dentate gyrus. J Neurosci 18: 3206–3212.
16. Verret L, Jankowsky JL, Xu GM, Borchelt DR, Rampon C (2007) Alzheimer’s-
type amyloidosis in transgenic mice impairs survival of newborn neurons derived
from adult hippocampal neurogenesis. J Neurosci 27: 6771–6780.
17. Gage FH, Kempermann G, Palmer TD, Peterson DA, Ray J (1998) Multipotent
progenitor cells in the adult dentate gyrus. J Neurobiol 36: 249–266.
18. Jin K, Peel AL, Mao XO, Xie L, Cottrell BA, et al. (2004) Increased
hippocampal neurogenesis in Alzheimer’s disease. Proc Natl Acad Sci U S A
101: 343–347.
19. Feng R, Rampon C, Tang YP, Shrom D, Jin J, et al. (2001) Deficient
neurogenesis in forebrain-specific presenilin-1 knockout mice is associated with
reduced clearance of hippocampal memory traces. Neuron 32: 911–926.
20. Haughey NJ, Nath A, Chan SL, Borchard AC, Rao MS, et al. (2002) Disrupti on
of neurogenesis by amyloid beta-peptide, and perturbed neural progenitor cell
homeostasis, in models of Alzheimer’s disease. J Neurochem 83: 1509–1524.
21. Haughey NJ, Liu D, Nath A, Borchard AC, Mattson MP (2002) Disruption of
neurogenesis in the subventricular zone of adult mice, and in human cortical
neuronal precursor cells in culture, by amyloid beta-peptide: implications for the
pathogenesis of Alzheimer’s disease. Neuromolecular Med 1: 125–135.
22. Wen PH, Shao X, Shao Z, Hof PR, Wisniewski T, 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.
23. Caille´ I, Allinquant B, Dupont E, Bouillot C, Langer A, et al. (2004) Soluble
form of amyloid precursor protein regulates proliferation of progenitors in the
adult subventricular zone. Development 131: 2173–2181.
24. Dong H, Goico B, Martin M, Csernansky CA, Bertchume A, et al. (2004)
Modulation of hippocampal cell proliferation, memory, and amyloid plaque
deposition in APPsw (Tg2576) mutant mice by isolation stress. Neuroscience
127: 601–609.
25. Wang R, Dineley KT, Sweatt JD, Zheng H (2004) Presenilin 1 familial
Alzheimer’s disease mutation leads to defective associative learning and impaired
adult neurogenesis. Neuroscience 126: 305–312.
26. Wolf SA, Kronenberg G, Lehmann K, Blankenship A, Overall R, et al. (2006)
Cognitive and physical activity differently modulate disease progression in the
amyloid precursor protein (APP)-23 model of Alzheimer’s disease. Biol
Psychiatry 60: 1314–1323.
27. Chevallier NL, Soriano S, Kang DE, Masliah E, Hu G, et al. (2005) Perturbed
neurogenesis in the adult hippocampus associated with presenilin-1 A246E
mutation. Am J Pathol 167: 151–159.
28. Donovan MH, Yazdani U, Norris RD, Games D, German DC, et al. (2006)
Decreased adult hippocampal neurogenesis in the PDAPP mouse model of
Alzheimer’s disease. J Comp Neurol 495: 70–83.
29. Jin K, Galvan V, Xie L, Mao XO, Gorostiza OF, et al. (2004) Enhanced
neurogenesis in Alzheimer’s disease transgenic (PDGF-APPSw,Ind) mice. Proc
Natl Acad Sci U S A 101: 13363–13367.
30. Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, et al. (2003) Triple-
transgenic model of Alzheimer’s disease with plaques and tangles: intracellular
Abeta and synaptic dysfunction. Neuron 39: 409–421.
31. Oddo S, Caccamo A, Kitazawa M, Tseng BP, LaFerla FM (2003) Amyloid
deposition precedes tangle formation in a triple transgenic model of Alzheimer’s
disease. Neurobiol Aging 24: 1063–1070.
32. Frazer ME, Hughes JE, Mastrangelo MA, Tibbens JL, Federoff HJ, et al. (2008)
Reduced pathology and improved behavioral performance in Alzheimer’s
disease mice vaccinated with HSV amplicons expressing amyloid-beta and
interleukin-4. Mol Ther. 16: 845–853.
33. McKee AC, Carreras I, Hossain L, Ryu H, Klein WL, et al. (2008) Ibuprofen
reduces Abeta, hyperphosphorylated tau and memory deficits in Alzheimer
mice. Brain Res. 1207: 225–236.
34. Carroll JC, Rosario ER, Chang L, Stanczyk FZ, Oddo S, et al. (2007)
Progesterone and estrogen regulate Alzheimer-like neuropathology in female
3xTg-AD mice. J Neurosci. 27: 13357–13365.
35. Baum LW (2005) Sex, hormones, and Alzheimer’s disease. J Gerontol A Biol Sci
Med Sci 60: 736–743.
36. Webber KM, Casadesus G, Perry G, Atwood CS, Bowen R, et al. (2005) Gender
differences in Alzheimer disease: the role of luteinizing hormone in disease
pathogenesis. Alzheimer Dis Assoc Disord 19: 95–99.
37. Clinton LK, Billings LM, Green KN, Caccamo A, Ngo J, et al. (2007) Age-
dependent sexual dimorphism in cognition and stress response in the 3xTg-AD
mice. Neurobiol Dis. 28: 76–82.
38. Eng LF, Ghirnikar RS, Lee YL (2000) Glial fibrillary acidic protein: GFAP-
thirty-one years (1969–2000). Neurochem Res 25: 1439–1451.
39. Hsu S, Raine L, Franger H (1981) Use of avidin-biotin-peroxidase complex
(ABC) in immunoperoxidase techniques: a comparison between ABC and
unlabeled antibody (PAP) procedures. J Histochem Cytochem 29: 577–580.
40. Paxinos G, Watson C (1986) The Rat Brain in Stereotaxic Coordinates. New
York: Academic Press.
41. Bannerman DM, Rawlins JN, McHugh SB, Deacon RM, Yee BK, et al. (2004)
Regional dissociations within the hippocampus–memory and anxiety. Neurosci
Biobehav Rev. 28: 273–283.
42. Sykova´ E, Vorı
´sek I, Antonova T, Mazel T, Meyer-Luehmann M, et al. (2005) ,
Jucker M (2005) Changes in extracellular space size and geometry in APP23
transgenic mice: a model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 102:
479–484.
43. Shors TJ, Miesegaes G, Beylin A, Zhao M, Rydel T, et al. (2001) Neurogenesis
in the adult is involved in the formation of trace memories. Nature 410:
372–376.
44. Boekhoorn K, Joels M, Lucassen PJ (2006) Increased proliferation reflects glial
and vascular-associated changes, but not neurogenesis in the presenile Alzheimer
hippocampus. Neurobiol Dis. 24: 1–14.
45. Cooper-Kuhn CM, Kuhn HG (2002) Is it all DNA repair? Methodological
considerations for detecting neurogenesis in the adult brain. Brain Res Dev
Brain Res 134: 13–21.
46. Rakic P (2002) Adult neurogenesis in mammals: an identity crisis. J Neurosci 22:
614–618.
47. Manly JJ, Merchant CA, Jacobs DM, Small SA, Bell K, et al. (2000) Endogenous
estrogen levels and Alzheimer’s disease among postmenopausal women.
Neurology 54: 833–837.
48. Galea LA, Spritzer MD, Barker JM, Pawluski JL (2006) Gonadal hormone
modulation of hippocampal neurogenesis in the adult. Hippocampus 16:
225–232.
49. Tanapat P, Hastings NB, Reeves AJ, Gould E (1999) Estrogen stimulates a
transient increase in the number of new neurons in the dentate gyrus of the adult
female rat. J Neurosci 19: 5792–5801.
50. Jacobsen JS, Wu CC, Redwine JM, Comery TA, Arias R, et al. (2006) Early-
onset behavioral and synaptic deficits in a mouse model of Alzheimer’s disease.
Proc Natl Acad Sci U S A 103: 5161–5166.
51. Zhao C, Deng W, Gage FH (2008) Mechanisms and functional implications of
adult neurogenesis Cell 132: 645–660.
52. Lagace DC, Fischer SJ, Eisch AJ (2007) Gender and endogenous levels of
estradiol do not influence adult hippocampal neurogenesis in mice. Hippocam-
pus 17: 175–180.
53. Ge S, Goh EL, Sailor KA, Kitabatake Y, Ming GL, et al. (2006) GABA regulates
synaptic integration of newly generated neurons in the adult brain. Nature 439:
589–593.
54. Olson AK, Eadie BD, Ernst C, Christie BR (2006) Environmental enrichment
and voluntary exercise massively increase neurogenesis in the adult hippocam-
pus via dissociable pathways. Hippocampus 16: 250–260.
Neurogenesis Reduction in AD
PLoS ONE | www.plosone.org 8 August 2008 | Volume 3 | Issue 8 | e2935
... Neurogenesis ↓ brain tissue postmortem (hippocampus) [23] ↓Proliferation and differentiation of neural progenitor cells in APP/PS1 and 3xTg-AD mouse models [24,25] Calmodulin ↓ brain tissue postmortem [26] Primary cilia ↓ in dentate gyrus cells of 3xTg-AD mice [27] ↑ in length in hippocampus of APP/PS1 mice [28] Acetylcholine ↓ [29] This Table summarizes the findings regarding the most accepted and widely described biomarkers, such as amyloid-β and tau proteins in blood and cerebrospinal fluid (CSF), postmortem human brains, and PET images. In addition, in biological processes altered in Alzheimer's disease, molecules such as calmodulin and organelles as primary cilia are shown as sources of potential biomarkers in different human fluids and tissues, as well as in transgenic animal models of AD. ↑: increased; ↓: decreased; *: in blood exosomes. ...
... In AD transgenic animal models, particularly in 3xTg mice, neurogenesis in the hippocampus is markedly altered. The reduction in neurogenesis in the SVZ and the dentate gyrus is directly associated with the presence of amyloid beta, which is exacerbated in aged mice (Table 2) [24,27]. In addition, impaired neurogenesis precedes plaque and tangle formation, suggesting that deficits in adult neurogenesis may mediate premature cognitive decline in AD [71]. ...
... The 3xTg-AD mouse model that carries mutations in APP, Tau, and PSEN1 exhibits impaired neurogenesis in the hippocampus SGZ, and dentate granular cells display a reduction in cilia length. Notably, this impairment in neuronal proliferation is exacerbated in females and aging mice (Table 2) [27]. In the APP/PS1 model, there is an abnormal elongation of 5-HT6-positive cilia on hippocampal neurons, along with an increase in the length of the axonal initial segment. ...
Article
Full-text available
Alzheimer's disease (AD), as the main cause of dementia, affects millions of people around the world, whose diagnosis is based mainly on clinical criteria. Unfortunately, the diagnosis is obtained very late, when the neurodegenerative damage is significant for most patients. Therefore, the exhaustive study of biomarkers is indispensable for diagnostic, prognostic, and even follow-up support. AD is a multifactorial disease, and knowing its underlying pathological mechanisms is crucial to propose new and valuable biomarkers. In this review, we summarize some of the main biomarkers described in AD, which have been evaluated mainly by imaging studies in cerebrospinal fluid and blood samples. Furthermore, we describe and propose neuronal precursors derived from the olfactory neuroepithelium as a potential resource to evaluate some of the widely known biomarkers of AD and to gear toward searching for new biomarkers. These neuronal lineage cells, which can be obtained directly from patients through a non-invasive and outpatient procedure, display several characteristics that validate them as a surrogate model to study the central nervous system, allowing the analysis of AD pathophysiological processes. Moreover, the ease of obtaining and harvesting endows them as an accessible and powerful resource to evaluate biomarkers in clinical practice.
... 130,131 Studies using animal models of AD have shown that region-specific impairment of neurogenesis may develop before the gross degeneration of neurons 132 and the formation of senile plaques or neurofibrillary tangles. 133 The presence of Aβ plaques and the increase in the proportion of Aβ-containing hippocampal neurons have also both been linked to a decrease in neurogenesis in animal models of AD. 134 Antidepressants, such as MAOIs, SSRIs, and norepinephrine-selective reuptake inhibitors have been found to promote neurogenesis in the hippocampus, suggesting that they may provide therapeutic effect in neurodegenerative diseases such as AD. [135][136][137] In mice treated with the SSRI fluoxetine for 28 days, an increase of 60% in the number of dividing neural progenitor cells was observed in the hippocampus; 70% of these neural progenitor cells displayed a neuronal marker while those remaining expressed a glial differentiation marker. ...
Article
Alzheimer disease (AD) is a devastating neuro- degenerative disorder that affects millions of individuals worldwide, with no effective cure. The main symptoms include learning and memory loss, and the inability to carry out the simplest tasks, sig- nificantly affecting patients’ quality of life. Over the past few years, tremendous progress has been made in research demonstrating a link between AD and major depressive disorder (MDD). Evidence suggests that MDD is commonly associated with AD and that it can serve as a precipitating factor for this disease. Antidepressants such as selective serotonin reuptake inhibitors, which are the first line of treatment for MDD, have shown great promise in the treatment of depression in AD, although their effectiveness remains controversial. The goal of this review is to summarize current knowledge regarding the association between AD, MDD, and antidepressant treatment. It first provides an overview of the interaction between AD and MDD at the level of genes, brain regions, neurotransmitter systems, and neuroinflammatory markers. The review then presents current evidence regarding the effective- ness of various antidepressants for AD-related pathophysiology and then finally discusses current limitations, challenges, and future directions.
... The accumulation of senescent neural precursor cells decreased adult neurogenesis in the hippocampus with age in mice [14]. Alzheimer's disease is an age-related neurodegenerative disease, including defective neurogenesis [55][56][57][58]. Defective neurogenesis causes a reduction in new-born neurons, potentially leading to the impairment of cognitive function [59]. ...
Article
Full-text available
As a lifelong source of neurons, neural stem cells (NSCs) serve multiple crucial functions in the brain. The senescence of NSCs may be associated with the onset and progression of Alzheimer’s disease (AD). Our study reveals a noteworthy finding, indicating that the AD-associated pathogenic protein amyloid-β (Aβ) substantially enhances senescence-related characteristics of human NSCs. These characteristics encompass the enhanced expression of p16 and p21, the upregulation of genes associated with the senescence-associated secretory phenotype (SASP), increased SA-β-gal activity, and the activation of the DNA damage response. Further studies revealed that Aβ treatment significantly downregulates the SIRT1 protein which plays a crucial role in regulating the aging process and decreases downstream PGC-1α and FOXO3. Subsequently, we found that SIRT1 overexpression significantly alleviates a range of Aβ-induced senescent markers in human NSCs. Taken together, our results uncover that Aβ accelerates cellular senescence in human NSCs, making SIRT1 a highly promising therapeutic target for senescent NSCs which may contribute to age-related neurodegenerative diseases, including AD.
... 58,59 In both Alzheimer's disease patients and animal models, dysregulation of NPC proliferation and self-renewal leads to abnormal neurogenesis. 54,[60][61][62] According to Elena et al 57 neurogenesis is still observed in the brains of healthy individuals up to the age of 90. However, in patients with AD, the generation of new neurons is significantly reduced. ...
Article
Full-text available
Alzheimer’s disease is a prevalent form of dementia among elderly individuals and is characterized by irreversible neurodegeneration. Despite extensive research, the exact causes of this complex disease remain unclear. Currently available drugs for Alzheimer’s disease treatment are limited in their effectiveness, often targeting a single aspect of the disease and causing significant adverse effects. Moreover, these medications are expensive, placing a heavy burden on patients’ families and society as a whole. Natural compounds and extracts offer several advantages, including the ability to target multiple pathways and exhibit high efficiency with minimal toxicity. These attributes make them promising candidates for the prevention and treatment of Alzheimer’s disease. In this paper, we provide a summary of the common natural products used in Chinese medicine for different pathogeneses of AD. Our aim is to offer new insights and ideas for the further development of natural products in Chinese medicine and the treatment of AD.
... The Aβ accumulates in the cortex and progresses to the hippocampus with age while tau pathology follows an inverse trend. Additionally, neurogenesis and cognitive dysfunction defects appear within 4 months [101] and gliosis after seven months with no neuronal loss [72,92]. Further investigations showed that neutrophils infiltrate the brain in 4-6-month-old 3xTg-AD mice, contributing to early cognitive decline. ...
Preprint
Full-text available
A form of dementia distinct from healthy cognitive aging, Alzheimer's disease (AD) is a complex multi-stage disease that currently afflicts over 50 million people worldwide. Unfortunately, previous therapeutic strategies developed from murine models emulating different aspects of AD pathogenesis have failed. Consequently, researchers are now developing models that express several aspects of pathogenesis that better reflect the clinical situation in humans. As such, this review seeks to provide insight regarding current applications of mammalian models in AD research by addressing recent developments and characterizations of prominent transgenic models and their contributions to pathogenesis as well as discuss the advantages, limitations, and application of emerging models (hAβ-KI) that better capture genetic heterogeneity and mixed pathologies observed in the clinical situation.
Article
Alzheimer’s disease (AD) is an approaching, progressive public health crisis which presently lacks an effective treatment. Various non-invasive novel therapies like repetitive transcranial magnetic stimulation have shown potential to improve cognitive performance in AD patients. In the present study, the effect of extremely low intensity magnetic field (MF) stimulation on neurogenesis and cortical electrical activity was explored. Adult Wistar rats were divided into Sham, AD and AD + MF groups. Streptozotocin (STZ) was injected intracerebroventricularly, at a dose of 3 mg/kg body weight for developing AD model. The AD rats were then exposed to MF (17.96 µT) from 8th day of STZ treatment until 15th day, followed by cognitive assessments and electrocortical recording. In brain tissue samples, cresyl violet staining and BrdU immunohistochemistry were done. MF exposure, improved passive avoidance and recognition memory, attenuated neuronal degeneration and enhanced cell proliferation (BrdU positive cells) in comparison to AD rats. It also significantly restores delta wave power from frontal lobe. Our results suggest that early-stage MF exposure could be an asset for AD research and open new avenues in slowing down the progression of Alzheimer’s disease.
Article
Full-text available
Early diagnosis of Alzheimer's is still difficult to do, thus it is important to carry out further research to find biomarkers that can be used to detect early Alzheimer's disease. Proliferating Cell Nuclear Antigen (PCNA) is known as a proliferation marker, which has the potential to detect neurogenesis in the brain. This study aimed to identify PCNA gene expression in the brain as a marker of Alzheimer’s disease in Macaca fascicularis. Macaca fascicularis was used in this study because of their similarities with humans in terms of their behavioral complexity, as well as high cognitive abilities and the formation of a pathological characteristic of Alzheimer's disease in the brain. This study used the brains of 7 monkeys in the hippocampus and cortical regions. Monkeys previously have been divided into old and adult age groups. The detection of PCNA gene expression was done using RT-qPCR method. The results showed the gene expression tended to be higher in the adult group and the hippocampus region, although based on statistical analysis showed no significant differences
Article
Full-text available
A form of dementia distinct from healthy cognitive aging, Alzheimer's disease (AD) is a complex multi-stage disease that currently afflicts over 50 million people worldwide. Unfortunately, previous therapeutic strategies developed from murine models emulating different aspects of AD pathogenesis were limited. Consequently, researchers are now developing models that express several aspects of pathogenesis that better reflect the clinical situation in humans. As such, this review seeks to provide insight regarding current applications of mammalian models in AD research by addressing recent developments and characterizations of prominent transgenic models and their contributions to pathogenesis as well as discuss the advantages, limitations, and application of emerging models that better capture genetic heterogeneity and mixed pathologies observed in the clinical situation.
Article
Full-text available
Astroglia are a broad class of neural parenchymal cells primarily dedicated to homoeostasis and defence of the central nervous system (CNS). Astroglia contribute to the pathophysiology of all neurological and neuropsychiatric disorders in ways that can be either beneficial or detrimental to disorder outcome. Pathophysiological changes in astroglia can be primary or secondary and can result in gain or loss of functions. Astroglia respond to external, non-cell autonomous signals associated with any form of CNS pathology by undergoing complex and variable changes in their structure, molecular expression, and function. In addition, internally driven, cell autonomous changes of astroglial innate properties can lead to CNS pathologies. Astroglial pathophysiology is complex, with different pathophysiological cell states and cell phenotypes that are context-specific and vary with disorder, disorder-stage, comorbidities, age, and sex. Here, we classify astroglial pathophysiology into (i) reactive astrogliosis, (ii) astroglial atrophy with loss of function, (iii) astroglial degeneration and death, and (iv) astrocytopathies characterised by aberrant forms that drive disease. We review astroglial pathophysiology across the spectrum of human CNS diseases and disorders, including neurotrauma, stroke, neuroinfection, autoimmune attack and epilepsy, as well as neurodevelopmental, neurodegenerative, metabolic and neuropsychiatric disorders. Characterising cellular and molecular mechanisms of astroglial pathophysiology represents a new frontier to identify novel therapeutic strategies.
Article
Full-text available
The neuropathological correlates of Alzheimer's disease (AD) include amyloid-β (Aβ) plaques and neurofibrillary tangles. To study the interaction between Aβ and tau and their effect on synaptic function, we derived a triple-transgenic model (3×Tg-AD) harboring PS1M146V, APPSwe, and tauP301L transgenes. Rather than crossing independent lines, we microinjected two transgenes into single-cell embryos from homozygous PS1M146V knockin mice, generating mice with the same genetic background. 3×Tg-AD mice progressively develop plaques and tangles. Synaptic dysfunction, including LTP deficits, manifests in an age-related manner, but before plaque and tangle pathology. Deficits in long-term synaptic plasticity correlate with the accumulation of intraneuronal Aβ. These studies suggest a novel pathogenic role for intraneuronal Aβ with regards to synaptic plasticity. The recapitulation of salient features of AD in these mice clarifies the relationships between Aβ, synaptic dysfunction, and tangles and provides a valuable model for evaluating potential AD therapeutics as the impact on both lesions can be assessed.
Article
Eighty-three brains obtained at autopsy from nondemented and demented individuals were examined for extracellular amyloid deposits and intraneuronal neurofibrillary changes. The distribution pattern and packing density of amyloid deposits turned out to be of limited significance for differentiation of neuropathological stages. Neurofibrillary changes occurred in the form of neuritic plaques, neurofibrillary tangles and neuropil threads. The distribution of neuritic plaques varied widely not only within architectonic units but also from one individual to another. Neurofibrillary tangles and neuropil threads, in contrast, exhibited a characteristic distribution pattern permitting the differentiation of six stages. The first two stages were characterized by an either mild or severe alteration of the transentorhinal layer Pre-alpha (transentorhinal stages I-II). The two forms of limbic stages (stages III-IV) were marked by a conspicuous affection of layer Pre-alpha in both transentorhinal region and proper entorhinal cortex. In addition, there was mild involvement of the first Ammon's horn sector. The hallmark of the two isocortical stages (stages V-VI) was the destruction of virtually all isocortical association areas. The investigation showed that recognition of the six stages required qualitative evaluation of only a few key preparations.
Article
Amyloid-β (Aβ) containing plaques and tau-laden neurofibrillary tangles are the defining neuropathological features of Alzheimer’s disease (AD). To better mimic this neuropathology, we generated a novel triple transgenic model of AD (3xTg-AD) harboring three mutant genes: β-amyloid precursor protein (βAPPSwe), presenilin-1 (PS1M146V), and tauP301L. The 3xTg-AD mice progressively develop Aβ and tau pathology, with a temporal- and regional-specific profile that closely mimics their development in the human AD brain. We find that Aβ deposits initiate in the cortex and progress to the hippocampus with aging, whereas tau pathology is first apparent in the hippocampus and then progresses to the cortex. Despite equivalent overexpression of the human βAPP and human tau transgenes, Aβ deposition develops prior to the tangle pathology, consistent with the amyloid cascade hypothesis. As these 3xTg-AD mice phenocopy critical aspects of AD neuropathology, this model will be useful in pre-clinical intervention trials, particularly because the efficacy of anti-AD compounds in mitigating the neurodegenerative effects mediated by both signature lesions can be evaluated.
Article
Since the early 1960s, in vivo observations have shown the generation of new neurons from dividing precursor cells. Nevertheless, these experiments suffered from skepticism, suggesting that the prevailing labeling method, which incorporates tagged thymidine analogs, such as [3H]-thymidine or bromodeoxyuridine (BrdU), may not be detecting a proliferative event, but could rather mark DNA repair in postmitotic neurons. Even today many scientists outside the field are still skeptical, because the question of specificity for thymidine labeling has not been sufficiently answered. This current paper aims at evaluating the arguments that are used by proponents and skeptics of this method by (i) presenting histological evidence of specificity of BrdU labeling for neural stem cell/progenitor activity in the adult brain; (ii) validating and comparing BrdU labeling with other histological methods; and (iii) combining BrdU and labeling methods for apoptosis to argue against DNA repair being a major contribution of BrdU-positive cells.
Article
The adult mammalian brain contains populations of stem cells that can proliferate and then differentiate into neurons or glia. The highest concentration of such neural progenitor cells (NPC) is located in the subventricular zone (SVZ) and these cells can produce new olfactory bulb and cerebral cortical neurons. NPC may provide a cellular reservoir for replacement of cells lost during normal cell turnover and after brain injury. However, neurogenesis does not compensate for neuronal loss in age-related neurodegenerative disorders such as Alzheimer’s disease (AD), suggesting the possibility that impaired neurogenesis contributes to the pathogenesis of such disorders. We now report that amyloid β-peptide (Aβ), a self-aggregating neurotoxic protein thought to cause AD, can impair neurogenesis in the SVZ/cerebral cortex of adult mice and in human cortical NPC in culture. The proliferation and migration of NPC in the SVZ of amyloid precursor protein (APP) mutant mice, and in mice receiving an intraventricular infusion of Aβ, were greatly decreased compared to control mice. Studies of NPC neurosphere cultures derived from human embryonic cerebral cortex showed that Aβ can suppress NPC proliferation and differentiation, and can induce apoptosis. The adverse effects of Aβ on neurogenesis were associated with a disruption of calcium regulation in the NPC. Our data show that Aβ can impair cortical neurogenesis, and suggest that this adverse effect of Aβ contributes to the depletion of neurons and the resulting olfactory and cognitive deficits in AD.